WO2025123137A1 - Heterocyclic compounds and methods of preparation thereof - Google Patents
Heterocyclic compounds and methods of preparation thereof Download PDFInfo
- Publication number
- WO2025123137A1 WO2025123137A1 PCT/CA2024/051651 CA2024051651W WO2025123137A1 WO 2025123137 A1 WO2025123137 A1 WO 2025123137A1 CA 2024051651 W CA2024051651 W CA 2024051651W WO 2025123137 A1 WO2025123137 A1 WO 2025123137A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- alkyl
- compound
- group
- cycloalkyl
- aryl
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Pending
Links
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P25/00—Drugs for disorders of the nervous system
- A61P25/24—Antidepressants
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/40—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
- A61K31/403—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
- A61K31/404—Indoles, e.g. pindolol
- A61K31/4045—Indole-alkylamines; Amides thereof, e.g. serotonin, melatonin
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K45/00—Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
- A61K45/06—Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P25/00—Drugs for disorders of the nervous system
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D209/00—Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
- C07D209/02—Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
- C07D209/04—Indoles; Hydrogenated indoles
- C07D209/10—Indoles; Hydrogenated indoles with substituted hydrocarbon radicals attached to carbon atoms of the hetero ring
- C07D209/14—Radicals substituted by nitrogen atoms, not forming part of a nitro radical
- C07D209/16—Tryptamines
Definitions
- the subject matter disclosed generally relates to heterocyclic compounds and methods of preparing the same.
- the subject matter disclosed also relates to the use of heterocyclic compounds as selective agents of the serotonin receptors.
- Psilocybin is a naturally occurring psychedelic compound produced by more than 200 species of mushrooms collectively known as “psilocybin mushrooms”. psilocybin mushrooms
- psilocybin As a prodrug, psilocybin is quickly metabolized by the body to generate the bioactive compound psilocin, which has mind-altering effects not unlike those produced by other psychedelics such as lysergic acid diethylamide (LSD), mescaline, and N,N-dimethyltryptamine (DMT).
- LSD lysergic acid diethylamide
- DMT N,N-dimethyltryptamine
- CNS diseases include both difficult-to-treat mental health disorders (Daniel J, Haberman M. Clinical potential of psilocybin as a treatment for mental health conditions. Ment. Health Clin 2017, 7(1), 24-8), such as treatment resistant depression or drug resistant depression, and neurological disorders such as cluster headaches.
- psilocybin While psilocybin has recognized therapeutic potential for treating certain CNS diseases and disorders, it is also recognized as a 5-HT2B receptor agonist and is therefore cardiotoxic. As such, there is an unmet need for safer drugs and analogs of psilocybin and psilocin that maintain 5-HT2A receptor agonist activity but that lack cardiotoxic 5-HT2B agonist activity; furthermore, and at least in some instances, there is an unmet need for safer drugs that maintain 5-HT 2 A receptor agonist activity but that lack cardiotoxic 5-HT2B agonist activity.
- R 1 is OH, and when R 1 is OH, R 2 is different than OH and (i) selected from the group consisting of H, halogen, lower alkyl, CHF 2 , CF 3 , NO, OCH 3 , OCHF 2 , OCF 3 , SCHF 2 , SCH 3 , SCF 3 , and cyano; or (ii) together with a form a ring of 3 or 4 atoms, one atom of which is selected from the group consisting of C, N, O, and S, while the remainder are carbon, which chain contains 0, 1 , or 2 double bonds, and to which chain are attached substituents independently selected from the group consisting of H, halogen, OH, Ci-Ce alkoxy, Ci-Ce alkyl, C3-C6 cycloalkyl, CHF2, CF3, OCHF2, OCF3, SCH3, SCHF2, SCF3, cyano, and oxo;
- R 2 is OH, and when R 2 is OH, R 1 is different than OH and (i) selected from the group consisting of H, halogen, lower alkyl, CHF2, CF3, OCH3, OCHF2, OCF3, SCHF2, SCH3, SCF3, and cyano; or (ii) together with b or c form a ring of 6 to 8 atoms, one atom of which is selected from the group consisting of C, N, O, and S, while the remainder are carbon, which chain contains 1 , or 2 double bonds, and to which chain are attached substituents independently selected from the group consisting of H, halogen, OH, Ci-Ce alkoxy, Ci-Ce alkyl, C3-C6 cycloalkyl, CHF2, CF3, OCHF2, OCF3, SCH3, SCHF2, SCF3, cyano, and oxo;
- R 4 is (i) is selected from the group consisting of H, Ci-Ce alkyl, Ci-Ce substituted alkyl, C2-C6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, (C 3 -C 6 cycloalkyl)(Ci-C 6 alkyl), C 3 -C 6 heterocyclyl, (C 3 - Ce heterocyclyl)(Ci-C6 alkyl), aryl(Ci-Ce alkyl), arylsulfonyl, heteroarylsulfonyl, aryl(Ci-C6 alkyl)sulfonyl, (Ci-C6)alkylsufonyl and heteroaryl(Ci-Ce alkyl)sulfonyl; or (ii) together with R 3 form a chain of 2 to 4 carbon atoms to which are attached substituents independently selected from the group consisting of H, Ci-C
- R 5 is H, D, Ci-Ce alkyl, Ci-Ce substituted alkyl;
- R 6 (i) is selected from the group consisting of H, Ci-Ce alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3- Ce cycloalkyl, (C3-C6 cycloalkyl)(Ci-Ce alkyl), aryl(Ci-Ce alkyl), acetyl, and heteroaryl(Ci-Ce alkyl); or (ii) together with R 7 and the N atom to which they are attached form a 4-7 membered heterocyclyl ring; or (iii) together with e and the N atom to which R 6 is attached form an azetidine or pyrrolidine ring, such ring carrying substituents independently selected from the group consisting of H, aryl, heteroaryl, Ci-C 6 alkyl, and C 3 -C 6 cycloalkyl; or (iv) together with b and the N atom to which R 6 is attached form an azetidine
- R 7 (i) is selected from the group consisting of H, Ci-Ce alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3- Ce cycloalkyl, (C3-C6 cycloalkyl)(Ci-Ce alkyl), aryl(Ci-Ce alkyl), acetyl, and heteroaryl(Ci-Ce alkyl); or (ii) together with R 6 and the N atom to which they are attached form a 4-7 membered heterocyclyl ring; a: (i) is selected from a group consisting of H, halogen, CH3, CHF2, CF3, OCH3, OCHF2, OCF3, SCH3, SCHF2, SCF3, NC and cyano; or (ii) together with a form a ring of 3 or 4 atoms, one atom of which is selected from the group consisting of C, N, O, and S, while the remainder are carbon, which chain contains 0, 1
- the a may be halogen.
- the a may be F.
- the a may be CF3.
- the a may be H.
- the R 1 may be OH and R 2 may be H.
- the R 1 may be H and R 2 may be OH.
- the R 4 may be H.
- the R 5 may be H.
- each of b, c, d, and e may be H.
- the compound of formula I may be a compound of formula II:
- the compound of formula I may be a compound of formula III:
- each of R 6 and R 7 may be CH3.
- the compound of formula I may be a compound of structural formula VI, or a pharmaceutically acceptable salt thereof, and stereoisomers thereof: wherein:
- R 1 is OH, and when R 1 is OH, R 2 is different than OH and selected from the group consisting of H, halogen, lower alkyl, CHF2, CF3, NO, OCH3, OCHF2, OCF3, SCHF2, SCH3, SCF3, and cyano;
- R 2 is OH, and when R 2 is OH, R 1 is different than OH and selected from the group consisting of H, halogen, lower alkyl, CHF2, CF3, OCH3, OCHF2, OCF3, SCHF2, SCH3, SCF3, and cyano;
- R 4 is selected from the group consisting of H, Ci-Ce alkyl, Ci-Ce substituted alkyl, C 2 - Ce alkenyl, C 2 -Ce alkynyl, C3-C6 cycloalkyl, (C3-C6 cycloalkyl)(Ci-Ce alkyl), C3-C6 heterocyclyl, (C 3 -C 6 heterocyclyl)(Ci-C 6 alkyl), aryl(Ci-C 6 alkyl), arylsulfonyl, heteroarylsulfonyl, aryl(Ci-C 6 alkyl)sulfonyl, (Ci-Ce)alkylsufonyl and heteroaryl(Ci-Ce alkyl)sulfonyl;
- R 5 is H, D, Ci-C 6 alkyl, Ci-C 6 substituted alkyl;
- R 6 is selected from the group consisting of H, Ci-Ce alkyl, C 2 -Ce alkenyl, C 2 -Ce alkynyl, C3-C6 cycloalkyl, (C3-C6 cycloalkyl)(Ci-Ce alkyl), aryl(Ci-Ce alkyl), acetyl, and heteroaryl(Ci-Ce alkyl);
- R 7 is selected from the group consisting of H, Ci-Ce alkyl, C 2 -Ce alkenyl, C 2 -Ce alkynyl, C 3 -C 6 cycloalkyl, (C 3 -C 6 cycloalkyl)(Ci-C 6 alkyl), aryl(Ci-C 6 alkyl), acetyl, and heteroaryl(Ci-C 6 alkyl); a: is selected from a group consisting of H, halogen, CH 3 , CHF 2 , CF 3 , OCH 3 , OCHF 2 , OCF3, SCH3, SCHF 2 , SCF3, NC and cyano; and b, c, d, and e, are each independently H; or three of b, c, d, and e are H and the remaining substituent is a lower alkyl group.
- the compound of formula I may be a compound of formula IV:
- the compound of formula IV may be a compound of formula IV-S:
- the compound of formula IV may be a compound of formula IV-/?:
- the compound of formula IV may be a racemate of a compound of formula IV-S and a compound of formula IV-/?;
- the compound of formula I may be a compound of formula V :
- the compound of formula V may be a compound of formula V-S:
- the compound of formula V may be a compound of formula V-R:
- the compound of formula V may be a racemate of a compound of formula V-S and a compound of formula ⁇ /-R: [0026] According to another embodiment, there is provided a pharmaceutical composition comprising the compound of the present invention, and a pharmaceutically acceptable carrier.
- a compound of the present invention for use in the treatment of a disorder in a patient in need thereof.
- a method of treating a disorder comprising administering to a patient an effective amount of the compound of the present invention, or the pharmaceutical composition of the present invention.
- the disorder is one or more of major depressive disorder, drug resistant depression, and psychotic depression, addiction including alcoholism, tobacco addiction, cocaine addiction, and opioid addiction, pain indications including neuropathic pain, pain from chemotherapy associated neuropathy, phantom limb pain and fibromyalgia, inflammation (including chronic and acute), eating disorders including anorexia, autism, cluster headaches, migraines, dementia including Alzheimer’s dementia, Parkinson’s disease dementia, and Lewy body dementia, mild cognitive impairment, post-traumatic stress disorder, emotional distress associated with cancer, Fragile-X syndrome, autism spectrum disorder, bipolar disease, obsessive compulsive disease, and Rett syndrome.
- major depressive disorder including alcoholism, tobacco addiction, cocaine addiction, and opioid addiction
- pain indications including neuropathic pain, pain from chemotherapy associated neuropathy, phantom limb pain and fibromyalgia, inflammation (including chronic and acute), eating disorders including anorexia, autism, cluster headaches, migraines, dementia including Alzheimer’s dementia, Parkinson’s disease dementia,
- a combination drug therapy comprising: a compound according to the present invention, or the pharmaceutical composition of the present invention; and a A/-methyl-D-aspartate (NMDA) receptor antagonist.
- NMDA A/-methyl-D-aspartate
- NMDA receptor antagonist is at least one of ketamine, nitrous oxide, memantine, amantadine, noribogaine, dextromethorphan, dextrorphan, and dextromethadone, or a pharmaceutically acceptable salt, stereoisomer, or solvate thereof.
- the combination drug therapy of the present invention for use in the treatment of a disorder in a patient in need thereof.
- a method of treating a disorder comprising administering to a patient an effective amount of the combination drug therapy of the present invention.
- substituted refers to a compound in which one or more hydrogen atoms have been replaced by other atoms or groups.
- alkenyl' refers to a substituted or unsubstituted, linear or branched, univalent hydrocarbon chain having at least two carbon atoms and at least one carbon-carbon (CC) double bond.
- alkenyl groups include allyl, vinyl, 2-propenyl, crotyl, 2-isopentenyl, 1 ,3-butadien-2-yl, 2,4-pentadien-1-yl, 1 ,4-pentadien- 3-yl, and the like.
- alkoxy used alone or as part of a larger moiety, refers to the groups -O-alkyl and -O-cycloalkyl.
- substituted alkoxy used alone or as part of a larger moiety, refers to the groups - ⁇ -(substituted alkyl) and -O-(substituted cycloalkyl).
- alkyl used alone or as part of a larger moiety, means a substituted or unsubstituted, linear or branched, univalent hydrocarbon chain that is completely saturated. Unless otherwise specified, an alkyl group contains 1 to 7 carbon atoms (“C1-C7 alkyl”).
- alkyl groups contain 1 to 6 carbon atoms (“Ci-Ce alkyl”); in some embodiments, alkyl groups contain 1 to 5 carbon atoms (“C1-C5 alkyl”); in some embodiments, alkyl groups contain 1 to 4 carbon atoms (“C1-C4 alkyl”, alternatively “lower alkyl”); and in some embodiments, alkyl groups contain 3 to 7 carbon atoms (“C3-C7 alkyl”).
- Non-limiting examples of saturated alkyl groups include methyl, ethyl, n-propyl, i- propyl, n-butyl, t-butyl, i-butyl, s-butyl, homologs and isomers of, for example, n-pentyl, n-hexyl, n-heptyl, n-octyl, and the like.
- Examples of lower alkyl groups include methyl, ethyl, n-propyl, i- propyl, n-butyl, s-butyl, i-butyl, and t-butyl.
- a substituted alkyl group is one having at least one or more substituents, and no more substituents than the number of hydrogen atoms in the unsubstituted group.
- the substituents are fluorine atoms.
- substituted alkyl groups include 2-hydroxyethyl, 2- methoxyethyl, CHF 2 , CF 3 , CH2CF3, CF 2 CF 3 ,4-fluorobutyl, and the like.
- alkynyl refers to a substituted or unsubstituted, linear or branched, univalent hydrocarbon chain having at least two carbon atoms and at least one carbon-carbon triple bond.
- alkynyl groups include ethynyl, 1- and 3-propynyl, 3-butyn-1-yl, and the like.
- aryl refers to a univalent monocyclic or bicyclic carbocyclic aromatic ring system. Unless otherwise specified, aryl groups contain 6 or 10 ring members. Nonlimiting examples of aryl include phenyl, naphthyl, and the like. The term “aryl” also refers to aryl groups that may be unsubstituted or substituted.
- aryl groups can be unsubstituted or can be substituted with one, two, three or more groups selected independently from the group consisting of halogen, OH, Ci-C 6 alkoxy, substituted Ci-C 6 alkoxy, Ci-C 6 alkylthio, substituted Ci-C 6 alkylthio, Ci- Ce alkyl, substituted Ci-Ce alkyl, C3-C6 cycloalkyl, substituted C3-C6 cycloalkyl, C(O)OH, C(O)(Ci-Ce alkyl), C(N-OH)(CI-C 6 alkyl), C(O)(Ci-C 6 alkoxy), C(O)NH 2 , C(O)NH(CI-C 6 alkyl), C(O)N(C C 4 alkyl)(Ci-C 4 alkyl), C(O)-heterocyclyl, NHC(O)(CI-C 6 alkyl), N(CH 3 )C(CH 3
- the term “chemical entity” refers to a compound having the indicated structure, whether in its “free” form (e g., “free compound” or “free base” or “free acid” form, as applicable), or in a salt form, particularly a pharmaceutically acceptable salt form, and furthermore whether in solid state form or otherwise.
- a solid state form is an amorphous (/.e., non-crystalline) form; in some embodiments, a solid state form is a crystalline form (e.g., a polymorph, pseudohydrate, hydrate, or solvate).
- the term encompasses the compound whether provided in solid form or otherwise. Unless otherwise specified, all statements made herein regarding “compounds” apply to the associated chemical entities, as defined.
- compositions, use, or method denotes that additional elements, method steps or both additional elements and method steps may be present, but that these additions do not materially affect the manner in which the recited composition, method, or use functions.
- cycloalkyl used alone or as part of a larger moiety, for example “(cycloalkyl)alkyl”, refers to: (i) a substituted or unsubstituted, univalent monocyclic hydrocarbon radical that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic; or (ii) bicyclo[m.n.o]alkyl wherein each of “m”, “n”, and “0” is independently an integer ranging from zero to 5, and the sum “m”+’n"+”o” ranges from 2 to 6.
- cycloalkyl groups contain 3 to 8 ring carbon atoms (“C3-C8 cycloalkyl”).
- Nonlimiting examples of cycloalkyl include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, 1 -cyclohexenyl, 3-cyclohexenyl, cycloheptyl, and the like, as well as bicyclo[2.2.1]heptyl (also called norbornyl) and bicyclo[1.1.1]pentyl.
- a substituted cycloalkyl group is one having at least one or more substituents. In some embodiments, the substituents are fluorine atoms.
- Non-limiting examples of substituted cycloalkyl groups include 2- methylcyclopropyl, 4-hydroxycyclohexyl, 2-methoxycyclopentyl, 4,4- difluorocyclohexyl, and the like.
- halogen or “halo”, used alone or as part of a larger moiety, refers to fluoro, chloro, bromo, or iodo.
- heteroalkyl refers to a substituted or unsubstituted, saturated or unsaturated alkyl group, as defined herein, in which one or more of the constituent carbon atoms have been replaced by nitrogen, oxygen, or sulfur.
- heteroaryl used alone or as part of a larger moiety, e.g., “(heteroaryl)alkyl”, refers to a univalent monocyclic or bicyclic group having 5 to 10 ring atoms, preferably 5, 6, 9, or 10 ring atoms, having 6 or 10 n electrons shared in a cyclic array, and having, in addition to ring carbon atoms, from one to four ring heteroatoms.
- heteroaryl groups include thienyl, furanyl, pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, oxadiazolyl, thiazolyl, isothiazolyl, thiadiazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, indolyl, indolizinyl, benzofuranyl, benzothiophenyl, benzimidazolyl, benzoxazolyl, benzothiazolyl, benzotriazolyl, quinolyl, isoquinolyl, purinyl, naphthyridinyl, ptendmyl, and the like.
- Heteroaryl groups may be unsubstituted or may be substituted with one, two, three or more groups selected independently from halogen, OH, Ci-Ce alkoxy, substituted Ci-Ce alkoxy, Ci-Ce alkylthio, substituted Ci-Ce alkylthio, Ci-Ce alkyl, substituted Ci-Ce alkyl, C3-C6 cycloalkyl, substituted C3-C6 cycloalkyl, C(O)OH, C(O)(Ci-C 6 alkoxy), C(O)NH 2 , C(O)NH(CI-C 6 alkyl), C(O)N(CI-C 4 alkyl)(Ci-C 4 alkyl), C(O)- heterocyclyl, NHC(O)(Ci-Ce alkyl), N(CH3)C(O)(CI- Ce alkyl), and cyano.
- heterocyclyl refers to a univalent stable 4- to 7- membered monocyclic or 7- to 10-membered bicyclic heterocyclic moiety that is either saturated or partially unsaturated, and has, in addition to ring carbon atoms, one to four heteroatoms.
- heterocyclyl groups include tetrahydrofuranyl, pyrrolidinyl, tetrahydropyranyl, piperidinyl, morpholinyl, and the like.
- Heterocyclyl groups can be unsubstituted or can be substituted.
- heterocyclyl groups can be unsubstituted or can be substituted with one, two, three or more groups selected independently from the group consisting of halogen, OH, O(Ci-Ce alkyl), O(substituted Ci-Ce alkyl), Ci-Ce alkyl, substituted Ci-Ce alkyl, and C3-C6 cycloalkyl.
- the term “inactive” when used the context of “EC50 (nM)” and “ Eff%” as such terms would be understood by a person skilled in the art or equivalent skilled person, and when used in reference to the activity at the 5-HT2B receptor, means a concentration of greater than 10,000 nM (when used in the context of “EC50 (nM)”) or an efficacy of 30% or lower (when used in the context of “ Eff%”).
- isotopologue refers to a species that differs from a specific compound only in the isotopic composition thereof.
- all hydrogen atoms in a compound are independently of natural isotopic composition or of any isotopic composition enriched or depleted in one or both of the heavy isotopes, 2 H (D, deuterium) and 3 H (T, tritium), ranging from a depletion to zero% to an enrichment to 100%.
- Deuterium may be incorporated into the compounds described herein in various ways, using deuterated versions of reagents and building blocks under the same or similar conditions as those employed for their counterparts with natural hydrogen isotope composition.
- LiAl D 4 can be used in the same manner to reduce urethane functions, such as Boc- orCbz-derivatized amines, to N-CD 3 .
- the building blocks methyl-d 3 iodide, ethyl-cfe iodide, allyl-cfe bromide, formaldehyde- d2 aqueous solution, paraformaldehyde- ⁇ , and dimethylamine-cfe (free base and hydrochloride) are commercially available, as are the reducing agents commonly employed in reductive aminations/alkylations, NaBD4 and NaBDsCN.
- Deuterium gas is available for the catalytic deuteration and deuterolysis of CC multiple bonds and C-heteroatom bonds, respectively. Indole-d? is commercially available.
- Electron-rich aromatics of which indoles are an example, can be ring- deuterated with D2O in the presence of the catalyst, B(CeFs)3, specifically in those positions that are more susceptible to electrophilic attack than an unactivated aromatic ring (Li, W.; Wang, M.-M.; Hu, Y.; Werner, T. Org. Lett. 2017, 19, 5768).
- Aromatics and heteroaromatics may also be deuterated by reaction with an excess of D 2 O in the presence of a heterogeneous transition metal catalyst (Sawama, Y.; Park, K.; Yamada, T.; Sajiki, H. Chem. Pharm. Bull. 2018, 66, 21-28).
- Deuteration of specific positions in the indole ring is achievable by halogen-metal exchange reactions on compounds that bear a halogen atom (typically Br or I) at the position to be deuterated, followed by quenching of the indolylmetal intermediate with a deuterating agent such as D2O or CH3OD; or by free-radical deuterodehalogenation of the same precursors with BusSnD and a radical starter such as azobis(isobutyronitrile) or di benzoyl peroxide; or by reaction of the same precursors with a deuteride source such as BusSnD or formic acid-cfe and a transition metal catalyst.
- the indole ring can as needed be reduced to indoline using a deuterium containing reducing agent such as EfaSiD.
- neurological disorder is intended to mean disorders that affect the brain as well as the nerves found throughout the human body and the spinal cord that may cause significant changes in thinking, emotion, and/or behavior, and distress and/or problems functioning in social, work, or family activities.
- neurodegenerative disorder is intended to mean disorders and diseases that affect the brain as well as the nerves found throughout the human body and the spinal cord that may cause significant changes in thinking, emotion, and/or behavior, and distress and/or problems functioning in social, work, or family activities.
- psychiatric disorder is intended to mean disorders that cause significant changes in thinking, emotion, and/or behavior, and distress and/or problems functioning in social, work, or family activities.
- time sufficient for/to or is intended to mean any period of time suitable to effect treatment with the method of the present invention.
- salts refers to those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response, and the like, and are commensurate with a reasonable benefit/risk ratio.
- Pharmaceutically acceptable salts of the compounds provided in this disclosure include salts derived from suitable inorganic and organic acids and bases.
- Non-limiting examples of pharmaceutically acceptable salts include salts of compounds comprising an amino group that are formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid, and perchloric acid or with organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid, or malonic acid.
- inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid, and perchloric acid
- organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid, or malonic acid.
- salts include adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydriodide, 2-hydroxyethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, digluconate,
- compositions include those that are derived from appropriate bases such as alkali metal, alkaline earth metal, ammonium, and N + (CI-4 alkyl)4 salts.
- Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like.
- Further non-limiting examples of pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, lower alkyl sulfonate, and aryl sulfonate.
- the term “subject” includes a mammal (e.g., a human, and in some embodiments including prenatal human forms).
- a subject suffers from a relevant disease, disorder, or condition.
- a subject is susceptible to a disease, disorder, or condition.
- a subject displays one or more symptoms or characteristics of a disease, disorder, or condition.
- a subject does not display any symptom or characteristic of a disease, disorder, or condition.
- a subject is a mammal with one or more features characteristic of susceptibility to or risk of a disease, disorder, or condition.
- a subject is a patient.
- a subject is an individual to whom diagnosis and/or therapy is and/or has been administered.
- a subject is a fetus, an infant, a child, a teenager, an adult, or a senior citizen (i.e. , the subject is of advanced age, such as older than 50).
- a child refers to a human that is between two and 18 years of age.
- an adult refers to a human that is eighteen years of age or older.
- the phrase “such as” is intended to be open-ended.
- the phrase “A can be a halogen, such as chlorine or bromine” means that “A” can be, but is not limited to, chlorine or bromine.
- structures depicted herein include all isomeric (e.g., enantiomeric, diastereomeric, and geometric (or conformational)) forms of the structure (e.g., the R and S configurations for each asymmetric center, Z and E double bond isomers, and Z and E conformational isomers).
- isomeric e.g., enantiomeric, diastereomeric, and geometric (or conformational) forms of the structure
- the compounds disclosed, taught, or otherwise suggested in this disclosure contemplate all single stereochemical isomers as well as enantiomeric, diastereomeric, and geometric (or conformational) mixtures thereof.
- the compounds disclosed, taught, or suggested in this disclosure contemplate all tautomeric forms thereof.
- structures depicted herein include compounds that differ only in the presence of one or more isotopically enriched atoms. Such compounds may be useful, for example, as analytical tools, as probes in biological assays, or as therapeutic agents. Additionally, incorporation of heavier isotopes such as deuterium ( 2 H) may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increase in vivo half-life, or reduced dosage requirements.
- isotopes such as deuterium ( 2 H) may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increase in vivo half-life, or reduced dosage requirements.
- structures depicted herein are also meant to include all stereoisomeric (e g., enantiomeric, diastereomeric, and geometric (or conformational)) forms of the structure; for example, the R and S configurations for each asymmetric center, Z and E double bond isomers, and Z and E conformational isomers. Therefore, the present compounds contemplate all single stereochemical isomers as well as enantiomeric, diastereomeric, and geometric (or conformational) mixtures thereof. Unless otherwise stated, the present compounds contemplate all tautomeric forms thereof. Additionally, unless otherwise stated, structures depicted herein are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms.
- Such compounds may be useful, for example, as analytical tools, as probes in biological assays, or as therapeutic agents. Additionally, incorporation of heavier isotopes such as deuterium ( 2 H) may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increase in vivo half-life, or reduced dosage requirements.
- isotopes such as deuterium ( 2 H)
- 2 H deuterium
- Fig. 1 illustrates EC50 graphs for compounds of formula IV in optically pure form (racemate), and PURM100 and PURM200 which are enantiomeric forms of Compound IV, against 5- HTIA, 5-HT2A, 5-HT2B, 5-HT2C, and m5-HT2A. ECso values are summarised in Table 1.
- Fig. 2 illustrates Concentration Response Curves for compounds of formula IV in optically pure form (racemate), and PURM100 and PURM200 which are enantiomeric forms of Compound IV, against 5-HT2A in agonist mode.
- Fig. 3 illustrates the plasma concentration following intravenous (IV) or oral (PO) of the racemic mixture of Compound PURM100 and Compound PURM200.
- Fig. 4 illustrates the plasma concentration following intravenous (IV), intraperinoneal (IPA or IPB) or oral (PO) of Compound PURM100.
- Fig. 5 illustrates the plasma concentration following intravenous (IV), intraperinoneal (IPA or IPB) or oral (PO) of Compound PURM200.
- Fig. 6 illustrates the head twitch response (HTR) in mice administered with 0.1 mg/kg to 10 mg/kg of the racemic mixture of Compound IV. HTR for the Compound IV is minimal in comparison to DOI as the control. Positive control is a racemic mixture of 2,5-Dimethoxy-4- iodoamphetamine (DOI), which is a a psychedelic drug and a substituted amphetamine.
- DOI 2,5-Dimethoxy-4- iodoamphetamine
- Fig. 7 illustrates that in mice administered with 0.1 mg/kg to 3 mg/kg of the racemic mixture of Compound IV, there are no changes to locomotor activity.
- Fig. 8 illustrates the results of Sholl Analysis which reveals that neurons treated with compounds PURM100 and PURM200 (which individually represent one of the enantiomeric forms of Compound IV) exhibit an increase in the number of process crossings in comparison to vehicle.
- Fig. 9 illustrates the results of Sholl Analysis which reveals that neurons treated with compounds PURM100 or PURM200 in the presence of IPTG, exhibit an increase in the number of process crossings in comparison to vehicle.
- Fig. 10 illustrates that neuronal stem cell cultures treated with compounds PURM100 and PURM200 (at 10 pM, which individually represent one of the enantiomeric forms of Compound IV) for 48h exhibit an increase in the number of branching of dendrites when compared to control treatments.
- FIG. 11 illustrates that neuronal stem cell cultures treated with compounds PURM100 and PURM200 (at 10 pM, which individually represent one of the enantiomeric forms of Compound IV) for48h, in the presence of IPTG to silence the HT 2A receptors.
- Statistical analysis indicates a significant effect of drug treatment on dendritogenesis produced only by BDNF treatment, implicating the 5-HT 2 A receptors in the effect observed with these compounds.
- Fig. 12 illustrates the results shown in Figs. 10 and 11 that were normalized with respect to their corresponding vehicle (control) as fold over basal in order to compare them.
- Statistical analysis indicates a significant effect of both variables — drug treatment and expression of 5-HT 2 A receptors — and the interaction between them. This means that the effect of drugs on dendritogenesis requires the expression of 5-HT 2 A receptors.
- Fig. 13 shows images of cells treated with water (control) and with Compound IV, where the treatment with compound IV for 48h causes an increase in the number of branching of dendrites when compared to control treatments.
- Fig. 14 illustrates the expression of c-fos in neurons treated with the pure enantiomeric forms of Compound IV, namely PURM100 and PURM200, with or without IPTG.
- C-fos is a functional marker of neuronal activity and initiation of protein transcription cascades.
- Fig. 15 illustrates the increase in expression of erg-1 , suggesting that PURM100 and PURM200 may activate neurons in absence of 5-HT 2 A signaling.
- Fig. 16 illustrates the increase in expression of erg-2, suggesting that PURM100 and PURM200 may activate neurons in absence of 5-HT 2 A signaling.
- R 1 is OH, and when R 1 is OH, R 2 is different than OH and (i) selected from the group consisting of H, halogen, lower alkyl, CHF 2 , CF 3 , NO, OCH 3 , OCHF 2 , OCF 3 , SCHF 2 , SCH 3 , SCF 3 , and cyano; or (ii) together with a form a ring of 3 or 4 atoms, one atom of which is selected from the group consisting of C, N, O, and S, while the remainder are carbon, which chain contains 0, 1 , or 2 double bonds, and to which chain are attached substituents independently selected from the group consisting of H, halogen, OH, Ci-Ce alkoxy, Ci-Ce alkyl, C 3 -Ce cycloalkyl, CHF 2 , CF 3 , OCHF 2 , OCF 3 , SCH 3 , SCHF 2 , SCF 3 , cyano, and ox
- R 2 is OH, and when R 2 is OH, R 1 is different than OH and (i) selected from the group consisting of H, halogen, lower alkyl, CHF 2 , CF 3 , OCH 3 , OCHF 2 , OCF 3 , SCHF 2 , SCH 3 , SCF 3 , and cyano; or (ii) together with b or c form a ring of 6 to 8 atoms, one atom of which is selected from the group consisting of C, N, O, and S, while the remainder are carbon, which chain contains 1 , or 2 double bonds, and to which chain are attached substituents independently selected from the group consisting of H, halogen, OH, Ci-Ce alkoxy, Ci-Ce alkyl, C 3 -Ce cycloalkyl, CHF 2 , CF 3 , OCHF 2 , OCF 3 , SCH 3 , SCHF 2 , SCF 3 , cyano, and ox
- R 4 is (i) is selected from the group consisting of H, Ci-Ce alkyl, Ci-Ce substituted alkyl, C2-C6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, (C 3 -C 6 cycloalkyl)(Ci-C 6 alkyl), C 3 -C 6 heterocyclyl, (C 3 - Ce heterocyclyl)(Ci-C6 alkyl), aryl(Ci-C6 alkyl), arylsulfonyl, heteroarylsulfonyl, , aryl(Ci-C6 alkyl)sulfonyl, (Ci-C 6 )alkylsufonyl and heteroaryl(Ci-C 6 alkyl)sulfonyl; or (ii) together with R 3 form a chain of 2 to 4 carbon atoms to which are attached substituents independently selected from the group consisting of H
- R 5 is H, D, Ci-Ce alkyl, Ci-Ce substituted alkyl;
- R 6 (i) is selected from the group consisting of H, Ci-C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 - Ce cycloalkyl, (C3-C6 cycloalkyl)(Ci-Ce alkyl), aryl(Ci-Ce alkyl), acetyl, and heteroaryl(Ci-Ce alkyl); or (ii) together with R 7 and the N atom to which they are attached form a 4-7 membered heterocyclyl ring; or (iii) together with e and the N atom to which R 6 is attached form an azetidine or pyrrolidine ring, such ring carrying substituents independently selected from the group consisting of H, aryl, heteroaryl, Ci-Ce alkyl, and C3-C6 cycloalkyl; or (iv) together with b and the N atom to which R 6 is attached form an an
- R 7 (i) is selected from the group consisting of H, Ci-Ce alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3- Ce cycloalkyl, (C3-C6 cycloalkyl)(Ci-Ce alkyl), aryl(Ci-Ce alkyl), acetyl, and heteroaryl(Ci-Ce alkyl); or (ii) together with R 6 and the N atom to which they are attached form a 4-7 membered heterocyclyl ring; a: (i) is selected from a group consisting of H, halogen, CH3, CHF2, CF3, OCH3, OCHF2, OCF3, SCH3, SCHF2, SCF3, NC and cyano; or (ii) together with a form a ring of 3 or 4 atoms, one atom of which is selected from the group consisting of C, N, O, and S, while the remainder are carbon, which chain contains 0, 1
- R 1 is OH and R 2 is H or the R 1 is H and R 2 is OH.
- the compound is a compound of formula IV:
- the compound is a compound of formula IV-S:
- the compound is a compound of formula IV-R:
- the compound is a racemate of a compound of formula IV-S and a compound of formula IV-R:
- the compound is a compound of formula V:
- the compound of formula V may be a compound of formula V-S:
- the compound of formula V may be a compound of formula V-R:
- the compound of formula V may be a racemate of a compound of formula V-S and a compound of formula V-R:
- R 2 is different than OH and (i) selected from the group consisting of H, halogen, lower alkyl, CHF2, CF3, OCH3, OCHF2, OCF3, SCHF2, SCH3, SCF3, and cyano; or (ii) together with a form a ring of 3 or4 atoms, one atom of which is selected from the group consisting of C, N, O, and S, while the remainder are carbon, which chain contains 0, 1 , or 2 double bonds, and to which chain are attached substituents independently selected from the group consisting of H, halogen, OH, Ci-Ce alkoxy, Ci-Ce alkyl, C3-C6 cycloalkyl, CHF2, CF3, OCHF2, OCF3, SCH3, SCHF2, SCF3, cyano, and oxo;
- R 4 is (i) is selected from the group consisting of H, Ci-Ce alkyl, Ci-Ce substituted alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, (C3-C6 cycloalkyl)(Ci-Ce alkyl), C3-C6 heterocyclyl, (C3- Ce heterocyclyl)(Ci-C6 alkyl), aryl(Ci-Ce alkyl), arylsulfonyl, heteroarylsulfonyl, aryl(Ci-Ce alkyl)sulfonyl, (Ci-Ce)alkylsufonyl and heteroaryl(Ci-Ce alkyl)sulfonyl; or (ii) together with R 3 form a chain of 2 to 4 carbon atoms to which are attached substituents independently selected from the group consisting of H, Ci-Ce alkyl,
- R 5 is H, D, Ci-Ce alkyl, Ci-Ce substituted alkyl
- R 7 (i) is selected from the group consisting of H, Ci-Ce alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3- Ce cycloalkyl, (C3-C6 cycloalkyl)(Ci-Ce alkyl), aryl(Ci-Ce alkyl), acetyl, and heteroaryl(Ci-Ce alkyl); or (ii) together with R 6 and the N atom to which they are attached form a 4-7 membered heterocyclyl ring; a: (i) is selected from a group consisting of H, halogen, CH3, CHF2, CF3, OCH3, OCHF2, OCF3, SCH3, SCHF2, SCF3, and cyano; or (ii) together with a form a ring of 3 or 4 atoms, one atom of which is selected from the group consisting of C, N, O, and S, while the remainder are carbon, which chain contains 0, 1
- the compound is a compound of formula IV:
- the compound is a compound of formula IV-S:
- the compound is a compound of formula IV-R:
- the compound is a racemate of a compound of formula IV-S and a compound of formula IV-/?:
- R 1 is different than OH and (i) selected from the group consisting of H, halogen, lower alkyl, CHF2, CF3, OCH3, OCHF2, OCF3, SCHF2, SCH3, SCF3, and cyano; or (ii) together with b or c form a ring of 6 to 8 atoms, one atom of which is selected from the group consisting of C, N, O, and S, while the remainderare carbon, which chain contains 1 , or2 double bonds, and to which chain are attached substituents independently selected from the group consisting of H, halogen, OH, Ci-C 6 alkoxy, Ci-C 6 alkyl, C 3 -C 6 cycloalkyl, CHF 2 , CF 3 , OCHF 2 , OCF 3 , SCH 3 , SCHF2, SCF3, cyano, and
- R 4 is (i) is selected from the group consisting of H, Ci-Ce alkyl, Ci-Ce substituted alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, (C3-C6 cycloalkyl)(Ci-C6 alkyl), C3-C6 heterocyclyl, (C3- C 6 heterocyclyl)(Ci-C 6 alkyl), aryl(Ci-C 6 alkyl), arylsulfonyl, heteroarylsulfonyl, aryl(Ci-C 6 alkyl)sulfonyl, (Ci-C6)alkylsufonyl and heteroaryl(Ci-Ce alkyl)sulfonyl; or (ii) together with R 3 form a chain of 2 to 4 carbon atoms to which are attached substituents independently selected from the group consisting of H, Ci-Ce alkyl,
- R 5 is H, D, Ci-Ce alkyl, Ci-Ce substituted alkyl;
- R 6 (i) is selected from the group consisting of H, Ci-Ce alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3- Ce cycloalkyl, (C3-C6 cycloalkyl)(Ci-Ce alkyl), aryl(Ci-Ce alkyl), acetyl, and heteroaryl(Ci-Ce alkyl); or (ii) together with R 7 and the N atom to which they are attached form a 4-7 membered heterocyclyl ring; or (iii) together with e and the N atom to which R 6 is attached form an azetidine or pyrrolidine ring, such ring carrying substituents independently selected from the group consisting of H, aryl, heteroaryl, Ci-Ce alkyl, and C3-C6 cycloalkyl; or (iv) together with b and the N atom to which R 6 is attached form an azetidine or
- R 7 (i) is selected from the group consisting of H, Ci-Ce alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3- Ce cycloalkyl, (C3-C6 cycloalkyl)(Ci-Ce alkyl), aryl(Ci-Ce alkyl), acetyl, and heteroaryl(Ci-Ce alkyl); or (ii) together with R 6 and the N atom to which they are attached form a 4-7 membered heterocyclyl ring; a: (i) is selected from a group consisting of H, halogen, CH3, CHF2, CF3, OCH3, OCHF2, OCF3, SCH3, SCHF2, SCF3, and cyano; or (ii) together with a form a ring of 3 or 4 atoms, one atom of which is selected from the group consisting of C, N, O, and S, while the remainder are carbon, which chain contains 0, 1
- the compound is a compound of formula V:
- the compound of formula V may be a compound of formula V-S:
- the compound of formula V may be a compound of formula V-R:
- the compound of formula V may be a racemate of a compound of formula V-S and a compound of formula V-R:
- R 1 is OH, and when R 1 is OH, R 2 is different than OH and selected from the group consisting of H, halogen, lower alkyl, CHF 2 , CF 3 , NO, OCH 3 , OCHF 2 , OCF 3 , SCHF 2 , SCH 3 , SCF 3 , and cyano;
- R 2 is OH, and when R 2 is OH, R 1 is different than OH and selected from the group consisting of H, halogen, lower alkyl, CHF2, CF3, OCH3, OCHF2, OCF3, SCHF2, SCH3, SCF3, and cyano;
- R 4 is selected from the group consisting of H, Ci-Ce alkyl, Ci-Ce substituted alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, (C3-C6 cycloalkyl)(Ci-C6 alkyl), C3-C6 heterocyclyl, (C3-C6 heterocyclyl)(Ci-C6 alkyl), aryl(Ci-C6 alkyl), arylsulfonyl, heteroarylsulfonyl, aryl(Ci-C6 alkyl)sulfonyl, (Ci-C6)alkylsufonyl and heteroaryl(Ci-C6 alkyl)sulfonyl;
- R 5 is H, D, Ci-Ce alkyl, Ci-Ce substituted alkyl;
- R 6 is selected from the group consisting of H, Ci-Ce alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, (C3-C6 cycloalkyl)(Ci-Ce alkyl), aryl(Ci-Ce alkyl), acetyl, and heteroaryl(Ci-Ce alkyl);
- R 7 is selected from the group consisting of H, Ci-Ce alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, (C3-C6 cycloalkyl)(Ci-Ce alkyl), aryl(Ci-Ce alkyl), acetyl, and heteroaryl(Ci-Ce alkyl);
- a: is selected from a group consisting of H, halogen, CH3, CHF2, CF3, OCH3, OCHF2, OCF3, SCH3, SCHF2, SCF3, NC and cyano; and
- b, c, d, and e, are each independently H; or three of b, c, d, and e are H and the remaining substituent is a lower alkyl group.
- Indoline compounds described herein are believed to be useful in the treatment of drug resistant depression based on several clinical trials that have been reported using psilocybin itself.
- Another potential use of the compounds of the present invention is in the treatment of seizure disorders, including but not limited to infantile seizure disorders such as but not limited to Dravet syndrome (Sourbon, J. et al. "Serotonergic Modulation as Effective Treatment for Dravet Syndrome in a Zebrafish Mutant Model”, ACS Chem. Neurosci. 2016, 7, 588-593).
- infantile seizure disorders such as but not limited to Dravet syndrome (Sourbon, J. et al. "Serotonergic Modulation as Effective Treatment for Dravet Syndrome in a Zebrafish Mutant Model”, ACS Chem. Neurosci. 2016, 7, 588-593).
- indoline compounds described herein are believed to be safer than psilocybin, given their lack of at least some of the undesirable characteristics of 5-HT2B-agonist related activities.
- Other potential uses of compounds of the present invention is one or more of major depressive disorder, drug resistant depression, and psychotic depression, addiction including alcoholism, tobacco addiction, cocaine addiction, and opioid addiction, pain indications including neuropathic pain, pain from chemotherapy associated neuropathy, phantom limb pain and fibromyalgia, inflammation (including chronic and acute), eating disorders including anorexia, autism, cluster headaches, migraines, dementia including Alzheimer’s dementia, Parkinson’s disease dementia, and Lewy body dementia, mild cognitive impairment, post-traumatic stress disorder, emotional distress associated with cancer, Fragile-X syndrome, autism spectrum disorder, bipolar disease, obsessive compulsive disease, and Rett syndrome.
- major depressive disorder including alcoholism, tobacco addiction, cocaine addiction, and opioid addiction
- pain indications including neuropathic pain, pain from chemotherapy associated neuropathy, phantom limb pain and fibromyalgia, inflammation (including chronic and acute), eating disorders including anorexia, autism, cluster headaches, migraines, dementia including Alzheimer’s dementia
- NMDA A/-methyl-D-aspartate
- the NMDA receptor antagonist is at least one of ketamine, nitrous oxide, memantine, amantadine, noribogaine, dextromethorphan, dextrorphan, and dextromethadone, or a pharmaceutically acceptable salt, stereoisomer, or solvate thereof.
- the NMDA receptor antagonist is dextromethorphan.
- the combination drug therapy may be useful for the treatment of one or more of major depressive disorder, drug resistant depression, and psychotic depression, addiction including alcoholism, tobacco addiction, cocaine addiction, and opioid addiction, pain indications including neuropathic pain, pain from chemotherapy associated neuropathy, phantom limb pain and fibromyalgia, inflammation (including chronic and acute), eating disorders including anorexia, autism, cluster headaches, migraines, dementia including Alzheimer’s dementia, Parkinson’s disease dementia, and Lewy body dementia, mild cognitive impairment, post-traumatic stress disorder, emotional distress associated with cancer, Fragile-X syndrome, autism spectrum disorder, bipolar disease, obsessive compulsive disease, and Rett syndrome.
- the combination drug therapy may be useful for the treatment of one or more of major depressive disorder, drug resistant depression, and psychotic depression.
- an indoline compound described herein is administered to a subject in need thereof. Whether such treatment is indicated depends on the subject case, and is further subject to medical assessment (diagnosis) that takes into consideration signs, symptoms, and/or malfunctions that are present, the risks of developing particular signs, symptoms and/or malfunctions, and other factors.
- an indoline compound described herein may be administered by any suitable route known in the art.
- Such routes include, but are not limited to, oral, buccal, inhalation, topical, sublingual, rectal, vaginal, intracisternal or intrathecal through lumbar puncture, transurethral, nasal, percutaneous, transdermal, and parenteral administration (including intravenous, intramuscular, subcutaneous, intracoronary, intradermal, intramammary, intraperitoneal, intraarticular, intrathecal, retrobulbar, intrapulmonary injection and/or surgical implantation at a particular site).
- Parenteral administration may be accomplished using a needle and syringe or using a high-pressure technique.
- compositions include those wherein an indoline compound described herein is present in a sufficient amount to be administered in an effective amount to achieve its intended purpose.
- the exact formulation, route of administration, and dosage is determined by a qualified medical practitioner in view of the diagnosed condition or disease. Dosage amount and interval can be adjusted individually to provide levels of an indoline compound described herein that is sufficient to maintain the desired therapeutic effects. It is possible that the indoline compound described herein may only require infrequent administration (e.g. monthly, as opposed to daily) to achieve the desired therapeutic effect.
- a therapeutically effective amount of an indoline compound described herein adapted for use in therapy varies with the nature of the condition being treated, the length of time that activity is desired, and the age and the condition of the patient, and ultimately is determined by the attendant physician. Dosage amounts and intervals can be adjusted individually to provide plasma levels of the indoline compound that are sufficient to maintain the desired therapeutic effects.
- the desired dose conveniently may be administered in a single dose, or as multiple doses administered at appropriate intervals, for example as one, two, three, four, or more subdoses per day. Multiple doses often may be desired or required.
- an indoline compound described herein may be administered at a frequency of: four doses delivered as one dose per day at four-day intervals (q4d x 4); four doses delivered as one dose per day at three-day intervals (q3d x 4); one dose delivered per day at five-day intervals (qd x 5); one dose per week for three weeks (qwk3); five daily doses, with two days’ rest, and another five daily doses (5/2/5); or, any dose regimen determined to be appropriate for the circumstance.
- the indoline compounds described herein may be administered in admixture with a pharmaceutical carrier selected with regard to the intended route of administration and standard pharmaceutical practice.
- Pharmaceutical compositions for use in accordance with the indoline compounds described herein are formulated in a conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries that facilitate processing of the compounds described herein.
- Water is a preferred carrier when an indoline compounds described herein is administered intravenously.
- Saline solutions and aqueous dextrose and glycerol solutions may also be used as liquid carriers, particularly for injectable solutions.
- Suitable pharmaceutical carriers also include excipients such as starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene glycol, water, ethanol, and the like.
- the present compositions if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents.
- compositions may be manufactured, for example, by conventional mixing, dissolving, granulating, dragee-making, emulsifying, encapsulating, entrapping, or lyophilizing processes. Proper formulation is dependent upon the route of administration chosen.
- a therapeutically effective amount of an indoline compound described herein is administered orally, the composition typically is in the form of a tablet, capsule, powder, solution, or elixir.
- the composition additionally can contain a solid carrier, such as a gelatin or an adjuvant.
- the tablet, capsule, and powder contain about 0.01% to about 95%, and preferably from about 1 % to about 50%, of an indoline compound described herein.
- a liquid carrier such as water, petroleum, or oils of animal or plant origin
- the liquid form of the composition can further contain physiological saline solution, dextrose or other saccharide solutions, or glycols.
- the composition When administered in liquid form, the composition contains about 0.1 % to about 90%, and preferably about 1% to about 50%, by weight, of a compound described herein.
- an indoline compound described herein described herein When a therapeutically effective amount of an indoline compound described herein described herein is administered by intravenous, cutaneous, or subcutaneous injection, the composition is in the form of a pyrogen-free, parenterally acceptable aqueous solution.
- a preferred composition for intravenous, cutaneous, or subcutaneous injection typically contains an isotonic vehicle.
- An indoline compound described herein described herein can be infused with other fluids over a 10-30 minute span or over several hours.
- indoline compounds described herein may be readily combined with pharmaceutically acceptable carriers well-known in the art. Such carriers enable the active agents to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a patient to be treated.
- Pharmaceutical preparations for oral use can be obtained by adding an indoline compound described herein to a solid excipient, with or without grinding the resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores. Suitable excipients include, for example, fillers and cellulose preparations. If desired, disintegrating agents can be added.
- An indoline compound described herein may be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion.
- Formulations for injection can be presented in unit dosage form, e.g., in ampules or in multidose containers, with an added preservative.
- the compositions can take such forms as suspensions, solutions, or emulsions in oily or aqueous vehicles, and can contain formulatory agents such as suspending, stabilizing, and/or dispersing agents.
- compositions for parenteral administration include aqueous solutions of the active agent in water-soluble form. Additionally, suspensions of an indoline compounds described herein can be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils or synthetic fatty acid esters. Aqueous injection suspensions can contain substances which increase the viscosity of the suspension.
- the suspension also can contain suitable stabilizers or agents that increase the solubility of the compounds and allow for the preparation of highly concentrated solutions.
- a present composition can be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
- an indoline compound described herein also may be formulated in rectal compositions, such as suppositories or retention enemas, e.g., containing conventional suppository bases.
- an indoline compound described herein also can be formulated as a depot preparation.
- Such long-acting formulations can be administered by implantation (for example, subcutaneously or intramuscularly) or by intramuscular injection.
- an indoline compound described herein may be formulated with suitable polymeric or hydrophobic materials (for example, as an emulsion in an acceptable oil) or ion exchange resins.
- An indoline compound described herein may be administered orally, buccally, or sublingually in the form of tablets containing excipients, such as starch or lactose, or in capsules or ovules, either alone or in admixture with excipients, or in the form of elixirs or suspensions containing flavoring or coloring agents.
- excipients such as starch or lactose
- capsules or ovules either alone or in admixture with excipients, or in the form of elixirs or suspensions containing flavoring or coloring agents.
- Such liquid preparations can be prepared with pharmaceutically acceptable additives, such as suspending agents.
- the indoline compounds described herein also may be injected parenterally, for example, intravenously, intramuscularly, subcutaneously, or intracoronarily.
- indoline compounds described herein may be best used in the form of a sterile aqueous solution which can contain other substances, for example, salts or monosaccharides, such as mannitol or glucose, to make the solution isotonic with blood.
- indoline compounds described herein are psilocybin analogs.
- Gq dissociation by bioluminescence resonance energy transfer (BRET) or Gq-dependent calcium flux is performed for selected compounds.
- BRET bioluminescence resonance energy transfer
- 5-HT 2 receptor-mediated Gq activation via Gq/y1 dissociation as measured by BRET (McCorvy JD, Wacker D, Wang S, Agegnehu B, Liu J, Lansu K, Tribo AR, Olsen RHJ, Che T, Jin J, Roth BL. Structural determinants of 5-HT2B receptor activation and biased agonism. Nat Struct Mol Biol.
- HEK293T cells are sub-cultured in Dulbecco’s Modified Eagle Medium (DMEM) supplemented with 10% dialyzed fetal bovine serum (FBS) and are co-transfected in a 1 :1 :1 :1 ratio with RLuc3-fused human Gaq (Gaq- RLucS), a GFP 2 -fused to the C-terminus of human Gy1 (Gy1-GFP 2 ), human Gp1 , and 5- HT2 receptor using TransiT-2020.
- DMEM Modified Eagle Medium
- FBS dialyzed fetal bovine serum
- transfected cells are plated in poly-L-lysine coated 96-well white clear bottom cell culture plates in DMEM containing 1 % dialyzed FBS at a density of 25,000-40,000 cells in 200 pL per well and incubated overnight. The next day, medium is decanted, and cells are washed with 60 pL of drug buffer (1x HBSS, 20 mM HEPES, pH 7.4), then 60 pL of drug buffer is added per well. Cells are pre-incubated in a humidified atmosphere at 37°C before receiving drug stimulation.
- drug buffer (1x HBSS, 20 mM HEPES, pH 7.4
- Drug stimulation utilized 30 pL addition of drug (3X) diluted in McCorvy buffer (1x HBSS, 20 mM HEPES, pH 7.4, supplemented with 0.3% BSA fatty acid free, 0.03% ascorbic acid), and plates are incubated for 1 hour at 37°C. Substrate addition occurred 15 minutes before reading and utilized 10 pL of the RLuc substrate coelenterazine 400a for Gq dissociation BRET 2 (Prolume/Nanolight, 5 pM final concentration). Plates are read for luminescence at 400 nm and fluorescent GFP 2 emission at 510 nm at 1 second perwell using a Mithras LB940 (multimode microplate reader (e.g. one provided by Berthold)).
- the BRET ratios of fluorescence/luminescence are calculated per well and are plotted as a function of drug concentration using Graphpad Prism 8 (Graphpad Software Inc., San Diego, CA). Data are normalized to % 5-HT stimulation and analyzed using nonlinear regression “log(agonist) vs. response” to yield E ma x and EC50 parameter estimates.
- receptor expression is induced with tetracycline (2 pg/mL), and cells are seeded into 384-well poly-L-lysine- coated black plates at a density of 7,500 cells/well in DMEM containing 1% dialyzed FBS.
- the cells are incubated for 1 hour at 37 J C with Fluo-4 Direct dye (Invitrogen, 20 pL/well) reconstituted in drug buffer (20 mM HEPES- buffered HBSS, pH 7.4) containing 2.5 mM probenecid.
- Drug dilutions are prepared at 5X final concentration in McCorvy buffer (20 mM HEPES-buffered HBSS, 0.1% BSA, 0.01 % ascorbic acid, pH 7.4). After dye load, cells are allowed to equilibrate to room temperature for 15 minutes, and then placed in a FLIPR® TETRA fluorescence imaging plate reader (Molecular Devices). The FLIPR® TETRA is programmed to read baseline fluorescence for 10 s (1 read/s), and afterward 5 pL of drug per well is added, and fluorescence is read for a total of 5-10 min (1 read/s).
- Fluorescence in each well is normalized to the average of the first 10 reads for baseline fluorescence, and then either maximum-fold peak increase over baseline or area under the curve (AUC) is calculated. Either peak or AUC is plotted as a function of drug concentration, and data are normalized to percent 5-HT stimulation. Data are plotted, and non-linear regression is performed using “log(agonist) vs. response” in Graphpad Prism 8 to yield E ma x and EC50 parameter estimates.
- the synthesis of the compound of Formula IV comprises several steps, namely: (1) construction of the heterocyclic core; (2) installation or modification of the 3-substituent; and (3) functional group transformations.
- Non-limiting examples of procedures for preparing the compound of Formula V described herein is provided below.
- the synthesis of the compound of Formula V comprises several steps, namely: (1) construction of the heterocyclic core; (2) installation or modification of the 3-substituent; and (3) functional group transformations.
- HEK293 cells expressing 5-HT receptors are trypsinised, counted and seeded in black, clear bottomed 96 well plates at a density of 25,000 cells per well and incubated overnight in media containing 1 % dialysed serum. Next day, media is removed from cell plates and replaced with 30 pl assay buffer (20 mM HEPES: HBSS, pH 7.4). Calcium 5 (Molecular Devices, R8186) dye solution (10 pl) is added to the wells and incubated at 37°C for 60 minutes. Dye solution is made up in 20 mM HEPES: HBSS, pH 7.4 + 2.5 mM probenecid.
- Compound dilutions are performed in 100% DMSO then transferred to intermediate dilutions for a very limited amount of time ( ⁇ 10 minutes) just before adding to the cell plate.
- Testing of 3 compounds (racemate, compound PURM100 and compound PURM200) at 7 concentrations in triplicate against a selected panel of 5- HT Receptors assays (5-HTIA, 5-HT2A, 5-HT2B, 5-HT2C and m5-HT2A).
- Compounds are tested as agonists at 100 pM, 10 pM, 1 pM, 0.1 pM, 0.01 pM, 1 nM and 0.1 nM.
- test compound or reference agonist (10 pl) is added to the wells and the fluorescence monitored for 5 minutes at ex/emm: 488nm/510- 570nm.
- the ECso values for the test compounds and reference compound were determined using GraphPad Prism software.
- the EC50 values for the reference compound are compared to historical assay and or literature data to ensure that it is within acceptable ranges in order to validate each assay.
- the % efficacy of test compounds measured against Emax of 5-HT is also determined.
- High potency the compounds PURM100 and -R have high potency against the 5-HT2A receptor (below 60nM). High potency can lead to therapeutic effects using a smaller dose. Lower doses can lead to fewer adverse side effects and greater patient outcomes. High potency leads to improved target receptor engagement in complex biological systems (e.g., competition against endogenous molecules).
- the compounds have high selectivity for 5-HT2A in comparison to other 5-HT receptors. For example, an unexpected and approximately 13x greater potency at 5-HT2A than 5-HT2C, minimizing off-target 5-HT effects.
- Low risk for cardio toxicity mitral and aortic valves are densely populated with 5-HT2B receptors; activation increases risk of valvular heart disease. The compounds PURM100 and - R lack of 5-HT2B activation.
- HEK cells expressing LgBiT tagged 5-HT2A and smBiT Beta-Arrestin are washed with PBS, to remove serum containing media, harvested in 1% dialysed serum containing media, counted and seeded in white 96 well plates at a density of 25,000 cells per well.
- the following day 25pl of NanoGio live cell substrate (Promega N2012) is added to each well and cells incubated for 25 minutes.
- Compounds are prepared as described for the 5-HTR calcium assay procedure detailed in Examiner 4 above and 20pl of 5X compound solution added to the cells. Cells are incubated for 90 minutes at 37°C and plates read for luminescence using the HidexTM plate reader.
- the EC50 of the test compounds and reference compound were determined using Graph Pad Prism software.
- the EC50 values for the reference compound are compared to historical assay and or literature data to ensure that it is within our acceptable ranges in order to validate each assay.
- the % efficacy of test compounds was measured against Emax of 5-HT. The results in were generated by the excel formulae (compound RFU - average min RFU)/(average max RFU- average min RFU))*100.
- the EC50 values were generated using the equation log(agonist) v response (four parameters) by Prism software (GraphPad Inc). The the raw data was normalized to the top and bottom of the 5-HT EC50 values using Prism software.
- the EC50 values were generated using the equation log(agonist) v response (three parameters) by Prism software (GraphPad Inc).
- Biased Gq signaling can activate phospholipase C (PLC), which hydrolyzes Phosphatidylinositol 4,5-bisphosphate (PIP2) into two second messengers, Inositol Trisphosphate (IP3) and Diacyclglycerol (DAG). These have several functions in cellular processes such as influencing cell proliferation and neurotransmitter release.
- PLC phospholipase C
- IP3 Inositol Trisphosphate
- DAG Diacyclglycerol
- the compound IV enantiomers are each more biased to the calcium/Gq signaling pathway than the racemic form. Reduced bias towards P-Arrestin2 suggests potentially less desensitization/downregulation of 5- HT 2 A receptors with the repeated administration of Compounds PURM100 and -R.
- Hepatocyte stability is a measure of how quickly a drug candidate is metabolized in the liver.
- a hepatocyte stability assay is an in vitro test that uses liver cells to determine how a compound is cleared. Hepatocyte stability assays can be used for a variety of purposes, including: Drug efflux, Uptake studies, Metabolite identification, and CYP induction/inhibition studies.
- Thaw the cells by placing the vial in a 37°C water bath and gently shaking the vials for 2 minutes. After thawing was completed, spray vial with 70% ethanol, transfer the vial to a biosafety cabinet.
- T ransfer well contents in 25 pL aliquots at time points of 0.5, 15, 30, 60, 90 and 120 minutes. The aliquots were then mixed with 6 volumes (150 pL) of acetonitrile containing with internal standard, IS (100 nM alprazolam, 200 nM caffeine and 100 nM tolbutamide) to terminate the reaction. Vortex for 5 minutes. Samples were centrifuges for 45 minutes at 3,220 g. Aliquot of 100 pL of the supernatant was diluted by 100 pL ultra-pure water, and the mixture was used for LC/MS/MS analysis. All incubations were performed in duplicate.
- IS internal standard
- V incubation volume (0.2 mL);
- N number of hepatocytes per well (0.1 x 10 6 cells).
- the pharmacokinetics parameters were measured following intravenous (IV), Oral (PO), and intraperitoneal administration (IP) of either of the racemic mixture, Compound PURM100 or Compound PURM200 administration to CD-1 male mice.
- IV administration the compounds were administered at 2 mg/kg
- PO administration the compounds were administered at 15 mg/kg
- IP administration the compounds were administered at 5 mg/kg, in saline solution.
- Dosing solutions for IV was 0.4 mg/mL
- PO was 3 mg/mL
- IP was 1 mg/mL.
- Brain samples were homogenized at a ratio of 1 :3 with PBS (W/V, 1 :3). The final brain concentration in the tissue was corrected by multiplying 4.
- the desired serial concentrations of working solutions were achieved by diluting stock solution of analyte with 50% acetonitrile in water solution.
- 5 pL of working solutions (1 , 2, 4, 10, 20, 100, 200, 1000, 2000 ng/mL) were added to 10 pL of blank plasma to achieve calibration standards of 0.5 ⁇ 1000 ng/mL (0.5, 1 , 2, 5, 10, 50, 100, 500, 1000 ng/mL) in a total volume of 15 pL.
- 4 quality control samples at 1 ng/mL, 2 ng/mL, 50 ng/mL and 800 ng/mL for plasma were prepared independently of those used for the calibration curves. These QC samples were prepared on the day of analysis in the same way as calibration standards.
- the desired serial concentrations of working solutions were achieved by diluting stock solution of analyte with 50% acetonitrile in water solution.15 pL of working solutions (1 , 2, 4, 10, 20, 100, 200, 1000, 2000 ng/mL) were added to 30 pL of blank brain homogenate to achieve calibration standards of 0.5 ⁇ 1000 ng/mL (0.5, 1 , 2, 5, 10, 50, 100, 500, 1000 ng/mL) in a total volume of 45 pL. 4 quality control samples at 1 ng/mL, 2 ng/mL, 50 ng/mL and 800 ng/mL for brain homogenate were prepared independently of those used for the calibration curves. These QC samples were prepared on the day of analysis in the same way as calibration standards.
- human ether-a-go-go-related gene is a potassium channel that plays a key role in the heart's electrical activity.
- In vitro hERG assays are a key part of assessing a drug’s liability for TdP (torsade de pointes) ventricular arrhythmias.
- TdP torsade de pointes
- the potential inhibitory effect of the racemic mixture of the compound IV on hERG channel was evaluated by using manual patch-clamp system according to the protocol.
- HEK 293 cell line stably expressing hERG channel (Cat. K1236) was purchased from InvitrogenTM. The cells are cultured in medium containing of 85% DMEM, 10% dialyzed FBS, 0.1 mM non-essential amino acid (NEAA), 25 mM HEPES, 100 U/mL Penicillin-Streptomycin, 5 pg/mL Blasticidin and 400 pg/mL Geneticin, cells grow in 25 cm2 cell culture bottles with 5% CO 2 and 37°C. Cells are split using TrypLETM Express about three times a week, and maintained between ⁇ 40% to ⁇ 80% confluence.
- the solution is filtered by filter system and stored at 4°C prior to use.
- Test compounds were initially prepared in DMSO with final concentration of 10 mM as stock solution.
- Dofetilide was initially prepared in DMSO with final concentration of 75 mM as stock solution.
- the working solutions are finally prepared by dilution of above described intermediate solutions in 1000 folds using extracellular solution, so that the final concentration of working solution was 150, 50, 16.67, 5.56 and 1 .85 nM.
- the final DMSO concentration in working solutions was maintained in range of 0.1% (v/v).
- hERG current in presence of 5 doses were tested for IC50 determination, consisting of 150, 50, 16.67, 5.56 and 1.85 pM.
- Peak current inhibition 1 - : - : - : - : - X 100
- the IC50 of the positive control Dofetilide is in line with the literature reports and exhibits good consistency between different batches of experiment, suggesting that current study running under appropriate condition and results were reliable.
- the IC50 ⁇ 0.37 pM is reported if the percentage hERG inhibition is more than 50% in presence of this compound at 0.37 pM dose concentration.
- the IC50 > 30 pM was reported if the fitting results of IC50 was greater than 30 pM or if the maximum inhibition at 30 pM was lower than 50%.
- a generally acceptable ranking system used to identify the potency of a test compound inhibiting hERG channel is listed as follows: a) Low: IC50 > 10 pM; b) Moderate: 1 pM ⁇ IC50 ⁇ 10 pM, or c) High: IC50 ⁇ 1 pM.
- test compound and control compounds were prepared in DMSO at the concentrations of 10 mM.
- the plate was placed into the well plate autosampler. The samples were evaluated by LC-MS/MS analysis.
- DF means the dilution factor. Any value of the compounds that was not within the specified limits was rejected and the experiment was repeated.
- Table 19 The solubility data of test compound and control compounds in 0.9% saline [00232] The upper limit for this assay was set at 300 pM. Any value close to or above 300 pM indicates that the compound may have a solubility at or above 300 pM.
- test Compounds PURM100 and PURM200 and control compound were prepared in DMSO at the concentration of 1 mM.
- a basic solution was prepared by dissolving 14.2 g/L Na2HPO4 and 8.77 g/L NaCI in deionized water and the solution could be stored at 4°C for up to 7 days.
- An acidic solution was prepared by dissolving 12.0 g/L NaH2PO4 and 8.77 g/L NaCI in deionized water and the solution could be stored at 4°C for up to 7 days.
- the basic solution was titrated with the acidic solution to pH 7.4 and store at 4°C for up to 7 days. pH was checked on the day of experiment and was adjusted if outside specification of 7.4 ⁇ 0.1 .
- the remaining spiked plasma solution sample in the plastic plate or separate plastic tube is incubated for 6 hours at 37°C with 5% CO2 in the CO2 incubator.
- PBS dialysis buffer
- [00241] Add 50 pL of plasma solution to the buffer samples, and an equal volume of PBS to the collected plasma solution samples. Shake the plate at 1000 rpm for 2 minutes and add 400 pL of acetonitrile containing an appropriate internal standard (IS) to precipitate protein and release compound. Vortex at 1000 rpm for 10 minutes. Centrifuge for 30 minutes at 3,220 g. Then transfer 100 pL of the supernatant to new 96-well plates for analysis. Add 100 pL of distilled water to each sample and mix for analysis by LC-MS/MS.
- IS internal standard
- a basic solution was prepared by dissolving 14.2 g/L Na 2 HPO 4 and 8.77 g/L NaCI in deionized water and the solution could be stored at 4°C for up to 7 days.
- An acidic solution was prepared by dissolving 12.0 g/L Na 2 HPO 4 and 8.77 g/L NaCI in deionized water and the solution could be stored at 4°C for up to 7 days.
- the basic solution was titrated with the acidic solution to pH 7.4 and store at 4°C for up to 7 days. pH was checked on the day of experiment and was adjusted if outside specification of 7.4 ⁇ 0.1 .
- Frozen mouse brain tissues were thawed immediately at room temperature. Then, it will be weighted and homogenized with buffer by brain weight (g) to buffer volume (mL) ratio 1 :4. The mouse brain tissues were from male CD1 mice.
- test compound and control compound were prepared in DMSO at the concentration of 200 pM.
- Preparation of operation plate [00248] Soak the dialysis membranes in ultrapure water for 60 minutes to separate strips, then in 20% ethanol for 20 minutes, finally in dialysis buffer for 20 minutes. Load the prepared membranes into the dialysis device and install the device again following manufacturers guidelines. Turn on air bath and allow to pre-heat to 37°C. If the dialysis membranes are not used immediately, they can be store at 4°C for no longer than 4 weeks.
- the remaining spiked brain homogenate sample in the plastic plate or separate plastic tube is incubated for 6 hours at 37°C with 5% CO2 in the CO2 incubator.
- 5-HT2 A R knockout mice were performed on adult (10-20 weeks old) 129S6/SvEv male mice.
- 5-HT 2 A-KO and controls in these experiments denote Htr2a-i- and Htr2a+/+ individuals, respectively, born to heterozygote (Htr2a+i-) breeders.
- Surgical implantation of the magnet was adapted from a previously reported protocol (Hanks, J. B. & Gonzalez-Maeso). Mice were anesthetized using a ketamine and xylazine (120 mg/kg and 12 mg/kg, respectively). A circular incision was performed to remove part of the scalp.
- Membranous tissue in and around the area was repeatedly cleaned with H2O2 and dried until the bone surface was exposed.
- Dental cement was applied in the exposed area to attach a small neodymium magnet (6.0, 6.0, 1 .5 mm, 490 mg) with the south pole facing the bone surface.
- the implant remained exposed with the dental cement bordering the skin.
- the mice were allowed to recover for 1 or 2 weeks. Animals were used repeatedly with a washout period of at least 1 week between tests.
- Isoflurane anesthesia was rapidly induced at an initial dose of 2% (vol/vol). When the effects of the anesthesia were apparent the animals were quickly administered DOI (1 mg/kg, i.p.) and placed on a heating pad on the isoflurane chamber. The dose of isoflurane was maintained at 1.5% for the following 30 min. Animals were regularly monitored.
- HTR were identified when: i) amplitude of the voltage signal exceeded the background noise level, and ii) corresponded in time with a peak maximum in the region of 70-110 Hz in the spectrogram heat-map often accompanied by a harmonic between 40-60 Hz of comparable or greater magnitude.
- HTR events relied on a script based on detection of individual events between 70-110 Hz. Recorded data was band-passed through a digital Butterworth filter between 70-110 Hz and transformed to absolute values. After double local maxima processing each original wavelet is transformed into a unipolar peak that is identified as HTR event when (i) peak prominence exceeds 0.075V, (ii) is separated from any other event by at least 200 ms, and (iii) has a width inferior to 90 ms at half the maximum value of prominence.
- Analytical reference ( ⁇ )-2,5-dimethoxy-4-iodoamphetamine, 2,5-dimethoxy-4- bromophenethylamine were purchased from Cerilliant Corporation (Round Rock, TX). Acetonitrile, ammonium acetate, ethyl acetate, formic acid, hexane, methanol, sodium hydroxide and water were purchased from Fisher Scientific (Hanover Park, IL, USA). All reagents were ACS grade or higher.
- Adherent neural stem cell lines previously generated from murine fetuses, were used as an in vitro experimental model. These cells are proliferative and susceptible to differentiation into neurons and glia by growing under appropriate cell culture conditions.
- the original NSC line was genetically engineered to harbor a shRNA sequence against the gene encoding the 5- HT 2 A receptor within its genome. The expression of this shRNA is regulated by a promoter modulated by the presence of IPTG in the cell culture medium. Experiments were conducted under both conditions, i.e., with cells treated or not treated with IPTG throughout the process.
- Figs. 8 and 9 show representative graphs showing Sholl analysis from one of the three independent experiments with cells grown in the absence (Fig. 8) or presence (Fig. 9) of IPTG in the culture medium.
- Sholl Analysis revealed that neurons treated with these compounds exhibit an increase in the number of process crossings in comparison to vehicle treated neurons.
- PSI represents psilocin.
- Statistical analysis of the results from Fig. 10 with One-way ANOVA resulted in a significant outcome, indicating an effect on neuronal dendritic arborization dependent on drug treatment. Bonferroni post hoc analysis to compare this data among the different treatment groups revealed a significant difference between all the drugs and their corresponding vehicle. No differences were found between Compound PURM100 and Compound PURM200 compared to the psilocin effect.
- FIG. 11 shows results from the same experiment carried out in conditions including IPTG in the cell culture medium, where the expression of 5-HT2A receptors is silenced.
- Statistical analysis indicates a significant effect of drug treatment on dendritogenesis produced only by BDNF treatment, implicating the 5-HT 2 A receptors in the effect observed with Compound PURM100 and Compound PURM200.
- Fig. 12 shows data from the results shown in Figs. 10 and 11 that were normalized with respect to their corresponding vehicle (control) as fold over basal in order to compare them.
- a two-way ANOVA was performed considering drug treatments and the presence of IPTG in the culture medium as variables. The statistical analysis indicates a significant effect of both variables — drug treatment and expression of 5-HT2A receptors — and the interaction between them. This means that the effect of drugs on dendritogenesis requires the expression of 5-HT2A receptors.
- Fig. 13 shows images of cells treated with water (control) and with a racemic mixture of Compound IV, where the treatment with compound IV for 48h causes an increase in the number of branching of dendrites when compared to control treatments.
- Compound PURM100 and Compound PURM200 treatment of differentiated neural progenitors into neurons and glia induces a significant increase in dendritogenesis in neurons, to a similar extent as psilocin treatment carried out in parallel.
- Equivalent assays conducted in cells grown in the presence of IPTG to silence the expression of 5-HT2A receptors resulted in no effect of these drugs on dendritogenesis, in contrast to BDNF, which was included in the experiments as a positive control.
- treatment with Compound PURM100 and Compound PURM200 for 48 hours promotes neuronal plasticity, assessed as the increment of dendritic arborization, and this effect is dependent on the expression of 5-HT2A receptors.
- Cell lines are the same as those used in neuronal plasticity experiments of Example 11 , and treatments were performed in parallel with the same drugs and at the same concentrations. In this case, after 5 days of differentiation, cells were treated with the different drugs and vehicle for 45 minutes at 37°C and 5% CO2. After incubation, cells were scraped and collected in 1 .5 mL centrifuge tubes.
- RNA extraction was accomplished by centrifuging the samples at 800g for 5 minutes at 4°C. The supernatant was discarded, and cells were resuspended in 400 pL of RNA-Solv reagent (Omega Bio-Tek®). Samples were incubated for 5 minutes at room temperature, and 80 pL of chloroform was added. Samples were mixed by inversion, left to rest for 5 minutes at room temperature, and then centrifuged at 12,000g for 15 minutes at 4°C. The aqueous phases of the supernatants were collected and transferred to new centrifuge tubes, where the same volume of isopropanol was added. Once again, samples were mixed by inversion and allowed to rest for 10 minutes at room temperature.
- RNA concentration was measured using a Nanodrop.
- cDNA synthesis was performed using TakaraO’s PrimeScriptTM RT Master Mix. For each sample, 500 ng of RNA, enough water to reach a final volume of 8 pL, and 2 pL of Takara’s PrimeScriptTM RT Master Mix were added to a 1.5 mL centrifuge tube. Samples were incubated at 37°C for 15 minutes, and the reaction was stopped using a 5-second cycle at 85°C. cDNA was measured using a Nanodrop, and samples were then diluted with water to a final concentration of 100 ng/pL.
- qPCRs were performed in BIO-RAD®’s clear 96-well Multiplate® PCR PlatesTM.
- the expression of c-fos, egr-1 , and egr-2 was tested for each biological replicate.
- the expression of each gene was measured using the median cq value of 4 technical replicates.
- Gene expression in each biological sample was normalized to the housekeeper gene GAPDH.
- Each qPCR plate was loaded with 16 wells worth of cDNA for each treatment for a total of 96 wells. Each condition was tested for our three genes of interest and the housekeeper (4 wells for each gene).
Landscapes
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- Life Sciences & Earth Sciences (AREA)
- Medicinal Chemistry (AREA)
- Pharmacology & Pharmacy (AREA)
- Animal Behavior & Ethology (AREA)
- General Health & Medical Sciences (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Engineering & Computer Science (AREA)
- Epidemiology (AREA)
- Biomedical Technology (AREA)
- Neurology (AREA)
- Neurosurgery (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Pain & Pain Management (AREA)
- Psychiatry (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
The present document describes compounds of structural formula (I), or a pharmaceutically acceptable salt thereof, and stereoisomers thereof, pharmaceutical compositions comprising the compounds of formula (I), as well as methods and/or uses of the compounds of formula (I) for treating disorders, particularly disorders depressive disorders.
Description
HETEROCYCLIC COMPOUNDS AND METHODS OF PREPARATION THEREOF
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] This application claims priority of United States Patent Application No. 63/610,614 filed on December 15, 2023, the specification of which is hereby incorporated by reference in its entirety.
BACKGROUND
(a) Field
[0002] The subject matter disclosed generally relates to heterocyclic compounds and methods of preparing the same. The subject matter disclosed also relates to the use of heterocyclic compounds as selective agents of the serotonin receptors.
(b) Related Prior Art
[0003] Psilocybin is a naturally occurring psychedelic compound produced by more than 200 species of mushrooms collectively known as “psilocybin mushrooms”.
psilocybin
[0004] As a prodrug, psilocybin is quickly metabolized by the body to generate the bioactive compound psilocin, which has mind-altering effects not unlike those produced by other psychedelics such as lysergic acid diethylamide (LSD), mescaline, and N,N-dimethyltryptamine (DMT). These effects include, among other things, euphoria, visual and mental hallucinations, changes in perception, a distorted sense of time, and spiritual experiences, and can also include possible adverse reactions such as nausea and panic attacks.
[0005] As agonists of the 5-HT2A and 5-HT2C receptors, psilocybin and psilocin have been recognized for their therapeutic potential. Since 5-HT2A receptor activation appears to increase locomotor activity, whereas 5-HT2C receptor activation appears to decrease locomotor activity, compounds possessing varying degrees of 5-HT2A and 5-HT2C activity will show varying levels of psychedelic activity. While psilocybin, along with other psychedelic drugs, were explored more than 60 years ago by Hofmann and co-workers at Sandoz (see for example, Hofmann, A., Troxler, F. US3075992; US3078214), clinical investigations into these drugs waned substantially by the early
1970s - particularly after these drugs were placed on Schedule 1 of the Controlled Substance Act in the United States of America. Notwithstanding their listing as controlled substances in certain jurisdictions however, research into psilocybin and other psychedelic drugs never fully stopped, and recent clinical investigations have led to a revived interest in the potential application of psychedelic drugs (including psilocybin) in evolving medical areas, such as the treatment of central nervous system (CNS) diseases. CNS diseases include both difficult-to-treat mental health disorders (Daniel J, Haberman M. Clinical potential of psilocybin as a treatment for mental health conditions. Ment. Health Clin 2017, 7(1), 24-8), such as treatment resistant depression or drug resistant depression, and neurological disorders such as cluster headaches.
[0006] While psilocybin has recognized therapeutic potential for treating certain CNS diseases and disorders, it is also recognized as a 5-HT2B receptor agonist and is therefore cardiotoxic. As such, there is an unmet need for safer drugs and analogs of psilocybin and psilocin that maintain 5-HT2A receptor agonist activity but that lack cardiotoxic 5-HT2B agonist activity; furthermore, and at least in some instances, there is an unmet need for safer drugs that maintain 5-HT2A receptor agonist activity but that lack cardiotoxic 5-HT2B agonist activity.
SUMMARY
[0007] According to an embodiment, there is provided a compound of structural formula I, or a pharmaceutically acceptable salt thereof, and stereoisomers thereof:
I wherein:
R1 is OH, and when R1 is OH, R2 is different than OH and (i) selected from the group consisting of H, halogen, lower alkyl, CHF2, CF3, NO, OCH3, OCHF2, OCF3, SCHF2, SCH3, SCF3, and cyano; or (ii) together with a form a ring of 3 or 4 atoms, one atom of which is selected from the group consisting of C, N, O, and S, while the remainder are carbon, which chain contains 0, 1 , or 2 double bonds, and to which chain are attached substituents independently selected
from the group consisting of H, halogen, OH, Ci-Ce alkoxy, Ci-Ce alkyl, C3-C6 cycloalkyl, CHF2, CF3, OCHF2, OCF3, SCH3, SCHF2, SCF3, cyano, and oxo;
R2 is OH, and when R2 is OH, R1 is different than OH and (i) selected from the group consisting of H, halogen, lower alkyl, CHF2, CF3, OCH3, OCHF2, OCF3, SCHF2, SCH3, SCF3, and cyano; or (ii) together with b or c form a ring of 6 to 8 atoms, one atom of which is selected from the group consisting of C, N, O, and S, while the remainder are carbon, which chain contains 1 , or 2 double bonds, and to which chain are attached substituents independently selected from the group consisting of H, halogen, OH, Ci-Ce alkoxy, Ci-Ce alkyl, C3-C6 cycloalkyl, CHF2, CF3, OCHF2, OCF3, SCH3, SCHF2, SCF3, cyano, and oxo;
R3 is (i) is selected from the group consisting of Ci-Ce alkyl, Ci-Ce substituted alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, (C3-C6 cycloalkyl)(Ci-C6 alkyl), C3-C6 heterocyclyl, (C3- Ce heterocyclyl)(Ci-C6 alkyl), aryl, aryl(Ci-C6 alkyl), heteroaryl, heteroaryl(Ci-Ce alkyl), CN, C(O)NH2, C(O)NH(CI-C6 alkyl), C(O)N(CI-C3 alkyl)(Ci-C6 alkyl), C(=NOH)(CI-C6 alkyl), and C(=NOH)(CI-C6 substituted alkyl), C(O)NH(aryl), C(O)N(alkyl)(aryl), NC, S(O)2NH2, S(O)2NH(Ci-Ce alkyl) and S(O)2NH(aryl); or (ii) together with R4 form a chain of 2 to 4 carbon atoms to which are attached substituents independently selected from the group consisting of H, Ci-Ce alkyl, aryl, and heteroaryl; or (iii) togetherwith a form a ring of 3 or4 atoms, one atom of which is selected from the group consisting of C, N, O, and S, while the remainder are carbon, which chain contains 0, 1 , or 2 double bonds, and to which chain are attached substituents independently selected from the group consisting of H, halogen, OH, Ci-C6 alkoxy, Ci-Ce alkyl, C3-C6 cycloalkyl, CHF2, CF3, OCHF2, OCF3, SCH3, SCF3, cyano, and oxo; or (iv) is selected from the group consisting of H, Ci-Ce alkyl, Ci-Ce substituted alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, (C3-C6 cycloalkyl)(Ci-C6 alkyl), C3-C6 heterocyclyl, (C3-C6 heterocyclyl)(Ci-C6 alkyl), aryl, aryl(Ci-C6 alkyl), heteroaryl, heteroaryl(Ci-Ce alkyl), CN, C(O)NH2, C(O)NH(CI-C6 alkyl), C(O)N(CI-C3 alkyl)(Ci-C6 alkyl), C(=NOH)(CI-C6 alkyl), and C(=NOH)(CI-C6 substituted alkyl), if b is halogen, CH3, CHF2, CF3, OCH3, OCHF2, OCF3, SCH3, SCHF2, SCF3, or cyano, optionally, R3 together with a form any one of CH2CH2, CH2CH2CH2, CH2CH2CH2CH2, CH=CHCH=CH, OCH2CH2, CH2OCH2, CH2CH2O, OCH=CH, CH=CHO, OCH2O, SCH2CH2, CH2SCH2, CH2CH2S, SCH=CH, CH=CHS, NHCH2CH2, CH2NHCH2, CH2CH2NH, NHCH=CH, CH=CHNH, ON=CH, CH=NO, OCH=N, N=CHO, SN=CH, CH=NS, SCH=N, N=CHS, NHN=CH, CH=NNH, NHCH=N, N=CHNH, NHN=N, N=NNH, OCH2CH2CH2, CH2OCH2CH2, CH2CH2OCH2, CH2CH2CH2O, SCH2CH2CH2, CH2SCH2CH2, CH2CH2SCH2, CH2CH2CH2S NHCH2CH2CH2, CH2NHCH2CH2, CH2CH2NCH2, CH2CH2CH2NH, N=CHCH=CH,
CH=NCH=CH, CH=CHN=CH, CH=CHCH=N; and optionally, wherein one hydrogen atom or two hydrogen atoms, if present on a moiety, are replaced with substituents selected independently from the group consisting of halogen, OH, Ci-Ce alkoxy, Ci-Ce alkyl, C3-C6 cycloalkyl, CHF2, CF3, OCHF2, OCF3, SCH3, SCFs.and cyano, or wherein two hydrogens, if attached to the same carbon atom, are replaced with an oxo group;
R4 is (i) is selected from the group consisting of H, Ci-Ce alkyl, Ci-Ce substituted alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, (C3-C6 cycloalkyl)(Ci-C6 alkyl), C3-C6 heterocyclyl, (C3- Ce heterocyclyl)(Ci-C6 alkyl), aryl(Ci-Ce alkyl), arylsulfonyl, heteroarylsulfonyl, aryl(Ci-C6 alkyl)sulfonyl, (Ci-C6)alkylsufonyl and heteroaryl(Ci-Ce alkyl)sulfonyl; or (ii) together with R3 form a chain of 2 to 4 carbon atoms to which are attached substituents independently selected from the group consisting of H, Ci-Ce alkyl, aryl, heteroaryl, and any combination thereof;
R5 is H, D, Ci-Ce alkyl, Ci-Ce substituted alkyl;
R6: (i) is selected from the group consisting of H, Ci-Ce alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3- Ce cycloalkyl, (C3-C6 cycloalkyl)(Ci-Ce alkyl), aryl(Ci-Ce alkyl), acetyl, and heteroaryl(Ci-Ce alkyl); or (ii) together with R7 and the N atom to which they are attached form a 4-7 membered heterocyclyl ring; or (iii) together with e and the N atom to which R6 is attached form an azetidine or pyrrolidine ring, such ring carrying substituents independently selected from the group consisting of H, aryl, heteroaryl, Ci-C6 alkyl, and C3-C6 cycloalkyl; or (iv) together with b and the N atom to which R6 is attached form an azetidine or pyrrolidine ring, such ring carrying substituents independently selected from the group consisting of H, aryl, heteroaryl, halogen, Ci-Ce alkyl, and C3-C6 cycloalkyl;
R7: (i) is selected from the group consisting of H, Ci-Ce alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3- Ce cycloalkyl, (C3-C6 cycloalkyl)(Ci-Ce alkyl), aryl(Ci-Ce alkyl), acetyl, and heteroaryl(Ci-Ce alkyl); or (ii) together with R6 and the N atom to which they are attached form a 4-7 membered heterocyclyl ring; a: (i) is selected from a group consisting of H, halogen, CH3, CHF2, CF3, OCH3, OCHF2, OCF3, SCH3, SCHF2, SCF3, NC and cyano; or (ii) together with a form a ring of 3 or 4 atoms, one atom of which is selected from the group consisting of C, N, O, and S, while the remainder are carbon, which chain contains 0, 1 , or 2 double bonds, and to which chain are attached substituents independently selected from the group consisting of H, halogen, OH, Ci-Ce alkoxy, Ci-C6 alkyl, C3-C6 cycloalkyl, CHF2, CF3, OCHF2, OCF3, SCH3, SCHF2, SCF3, cyano, and oxo; or (iii) together with R3 form a ring of 3 or 4 atoms, one atom of which is selected from the group consisting of C, N, O, and S, while the remainder are carbon, which chain contains 0, 1 ,
or 2 double bonds, and to which chain are attached substituents independently selected from the group consisting of H, halogen, OH, Ci-Ce alkoxy, Ci-Ce alkyl, C3-C6 cycloalkyl, CHF2, CF3, OCHF2, OCF3, SCH3, SCHF2, SCF3, cyano, and oxo; and b, c, d, and e, are each independently H; or three of b, c, d, and e are H and the remaining substituent is a lower alkyl group; or b and e are each H, and c and d together are -CH2- or - CH2CH2-, thereby giving rise to a cyclopropane or cyclobutane ring; or b, c, and d are each H, and e, R6, and the N atom to which R6 is attached form together an azetidine or pyrrolidine ring, such ring carrying substituents independently selected from the group consisting of H, aryl, heteroaryl, Ci-Ce alkyl, and C3-C6 cycloalkyl; or c, d, and e are each H, and b, R6, and the N atom to which R6 is attached form together an azetidine or pyrrolidine ring, such ring carrying substituents independently selected from the group consisting of H, aryl, heteroaryl, halogen, Ci-Ce alkyl, and C3-C6 cycloalkyl.
[0008] The a may be halogen. The a may be F. The a may be CF3. The a may be H.
[0009] The R1 may be OH and R2 may be H.
[0010] The R1 may be H and R2 may be OH.
[0011] The R4 may be H.
[0012] The R5 may be H.
[0013] In the compound of the present invention each of b, c, d, and e may be H.
II wherein each of R2, R3, R4, R5, R6, R7, a, b, c, d, and e are as defined above.
[0015] The compound of formula I may be a compound of formula III:
Ill wherein each of R1, R3, R4, R5, R6, R7, a, b, c, d, and e are as defined in claim 1.
[0016] In the compound of the present invention, each of R6 and R7 may be CH3.
[0017] The compound of formula I may be a compound of structural formula VI, or a pharmaceutically acceptable salt thereof, and stereoisomers thereof:
wherein:
R1 is OH, and when R1 is OH, R2 is different than OH and selected from the group consisting of H, halogen, lower alkyl, CHF2, CF3, NO, OCH3, OCHF2, OCF3, SCHF2, SCH3, SCF3, and cyano;
R2 is OH, and when R2 is OH, R1 is different than OH and selected from the group consisting of H, halogen, lower alkyl, CHF2, CF3, OCH3, OCHF2, OCF3, SCHF2, SCH3, SCF3, and cyano;
R3 is selected from the group consisting of Ci-Ce alkyl, Ci-Ce substituted alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, (C3-C6 cycloalkyl)(Ci-C6 alkyl), C3-C6 heterocyclyl, (C3- Ce heterocyclyl)(Ci-C6 alkyl), aryl, aryl(Ci-C6 alkyl), heteroaryl, heteroaryl(Ci-C6 alkyl), ON, C(O)NH2, C(O)NH(CI-C6 alkyl), C(O)N(CI-C3 alkyl)(Ci-C6 alkyl), C(=NOH)(CI-C6 alkyl), and
C(=NOH)(CI-C6 substituted alkyl), C(O)NH(aryl), C(O)N(alkyl)(aryl), NC, S(O)2NH2, S(O)2NH(CI-C6 alkyl) and S(O)2NH(aryl);
R4 is selected from the group consisting of H, Ci-Ce alkyl, Ci-Ce substituted alkyl, C2- Ce alkenyl, C2-Ce alkynyl, C3-C6 cycloalkyl, (C3-C6 cycloalkyl)(Ci-Ce alkyl), C3-C6 heterocyclyl, (C3-C6 heterocyclyl)(Ci-C6 alkyl), aryl(Ci-C6 alkyl), arylsulfonyl, heteroarylsulfonyl, aryl(Ci-C6 alkyl)sulfonyl, (Ci-Ce)alkylsufonyl and heteroaryl(Ci-Ce alkyl)sulfonyl;
R5 is H, D, Ci-C6 alkyl, Ci-C6 substituted alkyl;
R6: is selected from the group consisting of H, Ci-Ce alkyl, C2-Ce alkenyl, C2-Ce alkynyl, C3-C6 cycloalkyl, (C3-C6 cycloalkyl)(Ci-Ce alkyl), aryl(Ci-Ce alkyl), acetyl, and heteroaryl(Ci-Ce alkyl);
R7: is selected from the group consisting of H, Ci-Ce alkyl, C2-Ce alkenyl, C2-Ce alkynyl, C3-C6 cycloalkyl, (C3-C6 cycloalkyl)(Ci-C6 alkyl), aryl(Ci-C6 alkyl), acetyl, and heteroaryl(Ci-C6 alkyl); a: is selected from a group consisting of H, halogen, CH3, CHF2, CF3, OCH3, OCHF2, OCF3, SCH3, SCHF2, SCF3, NC and cyano; and b, c, d, and e, are each independently H; or three of b, c, d, and e are H and the remaining substituent is a lower alkyl group.
[0019] The compound of formula IV may be a compound of formula IV-S:
IV-S.
IV-R
[0021] The compound of formula IV may be a racemate of a compound of formula IV-S and a compound of formula IV-/?;
[0022] The compound of formula I may be a compound of formula V :
[0025] The compound of formula V may be a racemate of a compound of formula V-S and a compound of formula \/-R:
[0026] According to another embodiment, there is provided a pharmaceutical composition comprising the compound of the present invention, and a pharmaceutically acceptable carrier.
[0027] According to another embodiment, there is provided a compound of the present invention, or the pharmaceutical composition of the present invention, for use in the treatment of a disorder in a patient in need thereof.
[0028] According to another embodiment, there is provided a method of treating a disorder comprising administering to a patient an effective amount of the compound of the present invention, or the pharmaceutical composition of the present invention.
[0029] According to another embodiment, there is provided a use of the compound of of the present invention, or the pharmaceutical composition of the present invention, for the treatment of a disorder in a patient in need thereof.
[0030] The compound for use of the present invention, the method of the present invention, or the use of the present invention, wherein the disorder is one or more of major depressive disorder, drug resistant depression, and psychotic depression, addiction including alcoholism, tobacco addiction, cocaine addiction, and opioid addiction, pain indications including neuropathic pain, pain from chemotherapy associated neuropathy, phantom limb pain and fibromyalgia, inflammation (including chronic and acute), eating disorders including anorexia, autism, cluster headaches, migraines, dementia including Alzheimer’s dementia, Parkinson’s disease dementia, and Lewy body dementia, mild cognitive impairment, post-traumatic stress disorder, emotional distress associated with cancer, Fragile-X syndrome, autism spectrum disorder, bipolar disease, obsessive compulsive disease, and Rett syndrome.
[0031] The compound for use of the present invention, the method of the present invention, or the use of the present invention, wherein the disorder is one or more of major depressive disorder, drug resistant depression, and psychotic depression.
[0032] According to another embodiment, there is provided a combination drug therapy, comprising: a compound according to the present invention, or the pharmaceutical composition of the present invention; and a A/-methyl-D-aspartate (NMDA) receptor antagonist.
[0033] The combination drug therapy of the present invention, wherein the NMDA receptor antagonist is at least one of ketamine, nitrous oxide, memantine, amantadine, noribogaine,
dextromethorphan, dextrorphan, and dextromethadone, or a pharmaceutically acceptable salt, stereoisomer, or solvate thereof.
[0034] The combination drug therapy of the present invention, wherein the NMDA receptor antagonist is dextromethorphan.
[0035] The combination drug therapy of the present invention, for use in the treatment of a disorder in a patient in need thereof.
[0036] According to another embodiment, there is provided a method of treating a disorder comprising administering to a patient an effective amount of the combination drug therapy of the present invention.
[0037] According to another embodiment, there is provided a use of the combination drug therapy of the present invention, for the treatment of a disorder in a patient in need thereof.
[0038] The combination drug therapy for use of the present invention, the method of the present invention, or the use of the present invention, wherein the disorder is one or more of major depressive disorder, drug resistant depression, and psychotic depression, addiction including alcoholism, tobacco addiction, cocaine addiction, and opioid addiction, pain indications including neuropathic pain, pain from chemotherapy associated neuropathy, phantom limb pain and fibromyalgia, inflammation (including chronic and acute), eating disorders including anorexia, autism, cluster headaches, migraines, dementia including Alzheimer’s dementia, Parkinson’s disease dementia, and Lewy body dementia, mild cognitive impairment, post-traumatic stress disorder, emotional distress associated with cancer, Fragile-X syndrome, autism spectrum disorder, bipolar disease, obsessive compulsive disease, and Rett syndrome.
[0039] The combination drug therapy for use of the present invention, the method of the present invention, or the use of the present invention, wherein the disorder is one or more of major depressive disorder, drug resistant depression, and psychotic depression.
[0040] The following terms are defined below.
[0041] The use of the word “a” or “an” when used herein in conjunction with the term “comprising” may mean “one,” but it is also consistent with the meaning of “one or more,” “at least one” and “one or more than one.” Any element expressed in the singular form also encompasses its plural form. Any element expressed in the plural form also encompasses its singular form. The term “plurality” as used herein means more than one, for example, two or more, three or more, four or more, and the like.
[0042] As used herein and unless otherwise specified, the term “about”, when used to describe a recited value, means within 10% of the recited value.
[0043] As used herein and unless otherwise specified, the term “substituted” refers to a compound in which one or more hydrogen atoms have been replaced by other atoms or groups.
[0044] As used herein and unless otherwise specified, the term “alkenyl'” refers to a substituted or unsubstituted, linear or branched, univalent hydrocarbon chain having at least two carbon atoms and at least one carbon-carbon (CC) double bond. Examples of alkenyl groups include allyl, vinyl, 2-propenyl, crotyl, 2-isopentenyl, 1 ,3-butadien-2-yl, 2,4-pentadien-1-yl, 1 ,4-pentadien- 3-yl, and the like.
[0045] As used herein and unless otherwise specified, the term “alkoxy”, used alone or as part of a larger moiety, refers to the groups -O-alkyl and -O-cycloalkyl. As used herein and unless otherwise specified, the term “substituted alkoxy”, used alone or as part of a larger moiety, refers to the groups - □-(substituted alkyl) and -O-(substituted cycloalkyl).
[0046] As used herein and unless otherwise specified, the term “alkyl”, used alone or as part of a larger moiety, means a substituted or unsubstituted, linear or branched, univalent hydrocarbon chain that is completely saturated. Unless otherwise specified, an alkyl group contains 1 to 7 carbon atoms (“C1-C7 alkyl”). For example, in some embodiments, alkyl groups contain 1 to 6 carbon atoms (“Ci-Ce alkyl”); in some embodiments, alkyl groups contain 1 to 5 carbon atoms (“C1-C5 alkyl”); in some embodiments, alkyl groups contain 1 to 4 carbon atoms (“C1-C4 alkyl”, alternatively “lower alkyl”); and in some embodiments, alkyl groups contain 3 to 7 carbon atoms (“C3-C7 alkyl”). Non-limiting examples of saturated alkyl groups include methyl, ethyl, n-propyl, i- propyl, n-butyl, t-butyl, i-butyl, s-butyl, homologs and isomers of, for example, n-pentyl, n-hexyl, n-heptyl, n-octyl, and the like. Examples of lower alkyl groups include methyl, ethyl, n-propyl, i- propyl, n-butyl, s-butyl, i-butyl, and t-butyl. A substituted alkyl group is one having at least one or more substituents, and no more substituents than the number of hydrogen atoms in the unsubstituted group. In some embodiments, the substituents are fluorine atoms. Non-limiting examples of substituted alkyl groups include 2-hydroxyethyl, 2- methoxyethyl, CHF2, CF3, CH2CF3, CF2CF3,4-fluorobutyl, and the like.
[0047] As used herein and unless otherwise specified, the term “alkynyl” refers to a substituted or unsubstituted, linear or branched, univalent hydrocarbon chain having at least two carbon atoms and at least one carbon-carbon triple bond. Non-limiting examples of alkynyl groups include ethynyl, 1- and 3-propynyl, 3-butyn-1-yl, and the like.
[0048] As used herein and unless otherwise specified, the term “aryl”, used alone or as part of a larger moiety (for example, “(aryl)alkyl”) refers to a univalent monocyclic or bicyclic carbocyclic
aromatic ring system. Unless otherwise specified, aryl groups contain 6 or 10 ring members. Nonlimiting examples of aryl include phenyl, naphthyl, and the like. The term “aryl” also refers to aryl groups that may be unsubstituted or substituted. For example, aryl groups can be unsubstituted or can be substituted with one, two, three or more groups selected independently from the group consisting of halogen, OH, Ci-C6 alkoxy, substituted Ci-C6 alkoxy, Ci-C6 alkylthio, substituted Ci-C6 alkylthio, Ci- Ce alkyl, substituted Ci-Ce alkyl, C3-C6 cycloalkyl, substituted C3-C6 cycloalkyl, C(O)OH, C(O)(Ci-Ce alkyl), C(N-OH)(CI-C6 alkyl), C(O)(Ci-C6 alkoxy), C(O)NH2, C(O)NH(CI-C6 alkyl), C(O)N(C C4 alkyl)(Ci-C4 alkyl), C(O)-heterocyclyl, NHC(O)(CI-C6 alkyl), N(CH3)C(O)(CI-C6 alkyl), and cyano.
[0049] As used herein and unless otherwise specified or clear from context, the term “chemical entity” refers to a compound having the indicated structure, whether in its “free” form (e g., “free compound” or “free base” or “free acid” form, as applicable), or in a salt form, particularly a pharmaceutically acceptable salt form, and furthermore whether in solid state form or otherwise. In some embodiments, a solid state form is an amorphous (/.e., non-crystalline) form; in some embodiments, a solid state form is a crystalline form (e.g., a polymorph, pseudohydrate, hydrate, or solvate). Similarly, the term encompasses the compound whether provided in solid form or otherwise. Unless otherwise specified, all statements made herein regarding “compounds” apply to the associated chemical entities, as defined.
[0050] As used herein and unless otherwise specified, the terms “comprising”, “having”, “including”, “containing”, and grammatical variations thereof, are inclusive or open-ended and do not exclude additional, un-recited elements and/or method steps. For example, “A includes 1 ,2, and 3” means that A includes but is not limited to 1 , 2, and 3.
[0051] As used herein and unless otherwise specified, the term “consisting essentially of’ when used herein in connection with a composition, use, or method, denotes that additional elements, method steps or both additional elements and method steps may be present, but that these additions do not materially affect the manner in which the recited composition, method, or use functions.
[0052] As used herein and unless otherwise specified, the term “consisting of’ when used herein in connection with a composition, use, or method, excludes the presence of additional elements and/or method steps.
[0053] As used herein and unless otherwise specified, the term “cycloalkyl”, used alone or as part of a larger moiety, for example “(cycloalkyl)alkyl”, refers to: (i) a substituted or unsubstituted, univalent monocyclic hydrocarbon radical that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic; or (ii) bicyclo[m.n.o]alkyl wherein each of “m”, “n”, and “0" is independently an integer ranging from zero to 5, and the sum “m”+’n"+”o" ranges from 2 to 6. In
some embodiments, cycloalkyl groups contain 3 to 8 ring carbon atoms (“C3-C8 cycloalkyl”). Nonlimiting examples of cycloalkyl include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, 1 -cyclohexenyl, 3-cyclohexenyl, cycloheptyl, and the like, as well as bicyclo[2.2.1]heptyl (also called norbornyl) and bicyclo[1.1.1]pentyl. A substituted cycloalkyl group is one having at least one or more substituents. In some embodiments, the substituents are fluorine atoms. Non-limiting examples of substituted cycloalkyl groups include 2- methylcyclopropyl, 4-hydroxycyclohexyl, 2-methoxycyclopentyl, 4,4- difluorocyclohexyl, and the like.
[0054] As used herein and unless otherwise specified, the term “halogen” or “halo”, used alone or as part of a larger moiety, refers to fluoro, chloro, bromo, or iodo.
[0055] As used herein and unless otherwise specified, the term “heteroalkyl” refers to a substituted or unsubstituted, saturated or unsaturated alkyl group, as defined herein, in which one or more of the constituent carbon atoms have been replaced by nitrogen, oxygen, or sulfur.
[0056] As used herein and unless otherwise specified, the term “heteroaryl”, used alone or as part of a larger moiety, e.g., “(heteroaryl)alkyl”, refers to a univalent monocyclic or bicyclic group having 5 to 10 ring atoms, preferably 5, 6, 9, or 10 ring atoms, having 6 or 10 n electrons shared in a cyclic array, and having, in addition to ring carbon atoms, from one to four ring heteroatoms. Examples of heteroaryl groups include thienyl, furanyl, pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, oxadiazolyl, thiazolyl, isothiazolyl, thiadiazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, indolyl, indolizinyl, benzofuranyl, benzothiophenyl, benzimidazolyl, benzoxazolyl, benzothiazolyl, benzotriazolyl, quinolyl, isoquinolyl, purinyl, naphthyridinyl, ptendmyl, and the like. Heteroaryl groups may be unsubstituted or may be substituted with one, two, three or more groups selected independently from halogen, OH, Ci-Ce alkoxy, substituted Ci-Ce alkoxy, Ci-Ce alkylthio, substituted Ci-Ce alkylthio, Ci-Ce alkyl, substituted Ci-Ce alkyl, C3-C6 cycloalkyl, substituted C3-C6 cycloalkyl, C(O)OH, C(O)(Ci-C6 alkoxy), C(O)NH2, C(O)NH(CI-C6 alkyl), C(O)N(CI-C4 alkyl)(Ci-C4 alkyl), C(O)- heterocyclyl, NHC(O)(Ci-Ce alkyl), N(CH3)C(O)(CI- Ce alkyl), and cyano.
[0057] As used herein and unless otherwise specified, the term “heterocyclyl”, used alone or as part of a larger moiety (for example, “(heterocyclyl)alkyl”) refers to a univalent stable 4- to 7- membered monocyclic or 7- to 10-membered bicyclic heterocyclic moiety that is either saturated or partially unsaturated, and has, in addition to ring carbon atoms, one to four heteroatoms. Non-limiting examples of heterocyclyl groups include tetrahydrofuranyl, pyrrolidinyl, tetrahydropyranyl, piperidinyl, morpholinyl, and the like. Heterocyclyl groups can be unsubstituted or can be substituted. For example, heterocyclyl groups can be unsubstituted or can be substituted with one, two, three or more groups
selected independently from the group consisting of halogen, OH, O(Ci-Ce alkyl), O(substituted Ci-Ce alkyl), Ci-Ce alkyl, substituted Ci-Ce alkyl, and C3-C6 cycloalkyl.
[0058] As used herein and unless otherwise specified, the term “inactive” (and all related terms thereto including “inactivity”), when used the context of “EC50 (nM)” and “ Eff%” as such terms would be understood by a person skilled in the art or equivalent skilled person, and when used in reference to the activity at the 5-HT2B receptor, means a concentration of greater than 10,000 nM (when used in the context of “EC50 (nM)”) or an efficacy of 30% or lower (when used in the context of “ Eff%”).
[0059] As used herein and unless otherwise specified, the term “isotopologue” refers to a species that differs from a specific compound only in the isotopic composition thereof. For example, all hydrogen atoms in a compound are independently of natural isotopic composition or of any isotopic composition enriched or depleted in one or both of the heavy isotopes, 2H (D, deuterium) and 3H (T, tritium), ranging from a depletion to zero% to an enrichment to 100%. Deuterium may be incorporated into the compounds described herein in various ways, using deuterated versions of reagents and building blocks under the same or similar conditions as those employed for their counterparts with natural hydrogen isotope composition. The reduction of 3-acyl groups on the indole nucleus and of carboxamides with commercially available LiAl D4 or BD3-THF complex has already been mentioned. LiAl D4 can be used in the same manner to reduce urethane functions, such as Boc- orCbz-derivatized amines, to N-CD3. The building blocks methyl-d3 iodide, ethyl-cfe iodide, allyl-cfe bromide, formaldehyde- d2 aqueous solution, paraformaldehyde-^, and dimethylamine-cfe (free base and hydrochloride) are commercially available, as are the reducing agents commonly employed in reductive aminations/alkylations, NaBD4 and NaBDsCN. Deuterium gas is available for the catalytic deuteration and deuterolysis of CC multiple bonds and C-heteroatom bonds, respectively. Indole-d? is commercially available. Electron-rich aromatics, of which indoles are an example, can be ring- deuterated with D2O in the presence of the catalyst, B(CeFs)3, specifically in those positions that are more susceptible to electrophilic attack than an unactivated aromatic ring (Li, W.; Wang, M.-M.; Hu, Y.; Werner, T. Org. Lett. 2017, 19, 5768). Aromatics and heteroaromatics may also be deuterated by reaction with an excess of D2O in the presence of a heterogeneous transition metal catalyst (Sawama, Y.; Park, K.; Yamada, T.; Sajiki, H. Chem. Pharm. Bull. 2018, 66, 21-28). Deuteration of specific positions in the indole ring is achievable by halogen-metal exchange reactions on compounds that bear a halogen atom (typically Br or I) at the position to be deuterated, followed by quenching of the indolylmetal intermediate with a deuterating agent such as D2O or CH3OD; or by free-radical deuterodehalogenation of the same precursors with BusSnD and a radical starter such as azobis(isobutyronitrile) or di benzoyl peroxide; or by reaction of the same precursors with a deuteride
source such as BusSnD or formic acid-cfe and a transition metal catalyst. The indole ring can as needed be reduced to indoline using a deuterium containing reducing agent such as EfaSiD.
[0060] The term “neurological disorder” is intended to mean disorders that affect the brain as well as the nerves found throughout the human body and the spinal cord that may cause significant changes in thinking, emotion, and/or behavior, and distress and/or problems functioning in social, work, or family activities.
[0061] The term “neurodegenerative disorder” is intended to mean disorders and diseases that affect the brain as well as the nerves found throughout the human body and the spinal cord that may cause significant changes in thinking, emotion, and/or behavior, and distress and/or problems functioning in social, work, or family activities.
[0062] The term “psychiatric disorder” is intended to mean disorders that cause significant changes in thinking, emotion, and/or behavior, and distress and/or problems functioning in social, work, or family activities.
[0063] The term “time sufficient for/to” or is intended to mean any period of time suitable to effect treatment with the method of the present invention.
[0064] The term “a therapeutically effective amount” is intended to mean an amount that is effective in therapy, or an amount sufficient to provide a therapeutic effect. An amount that is effective in therapy is an amount which produces a biological activity and will relieve to some extent one or more symptoms of the disorder being treated, or result in inhibition of the progress or at least partial reversal of the condition. It will depend, among other things, on the individual. The typical daily dose of the active substance(s) may vary and will depend on various factors such as the individual requirements of the patients (e.g., the age, weight, etc. of the subject to be treated), the mode of administration, the disease or condition and its severity.
[0065] As used herein and unless otherwise specified, the term “pharmaceutically acceptable salt” refers to those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response, and the like, and are commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts of the compounds provided in this disclosure include salts derived from suitable inorganic and organic acids and bases. Non-limiting examples of pharmaceutically acceptable salts include salts of compounds comprising an amino group that are formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid, and perchloric acid or with organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid, or malonic acid. Other non-limiting examples of pharmaceutically acceptable salts include adipate, alginate,
ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydriodide, 2-hydroxyethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, pivalate, propionate, stearate, thiocyanate, p- toluenesulfonate, undecanoate, valerate salts, and the like. Other pharmaceutically acceptable salts include those that are derived from appropriate bases such as alkali metal, alkaline earth metal, ammonium, and N+(CI-4 alkyl)4 salts. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like. Further non-limiting examples of pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, lower alkyl sulfonate, and aryl sulfonate.
[0066] As used herein and unless otherwise specified, the term “subject" includes a mammal (e.g., a human, and in some embodiments including prenatal human forms). In some embodiments, a subject suffers from a relevant disease, disorder, or condition. In some embodiments, a subject is susceptible to a disease, disorder, or condition. In some embodiments, a subject displays one or more symptoms or characteristics of a disease, disorder, or condition. In some embodiments, a subject does not display any symptom or characteristic of a disease, disorder, or condition. In some embodiments, a subject is a mammal with one or more features characteristic of susceptibility to or risk of a disease, disorder, or condition. In some embodiments, a subject is a patient. In some embodiments, a subject is an individual to whom diagnosis and/or therapy is and/or has been administered. In some embodiments, a subject is a fetus, an infant, a child, a teenager, an adult, or a senior citizen (i.e. , the subject is of advanced age, such as older than 50). In some embodiments, a child refers to a human that is between two and 18 years of age. In some embodiments, an adult refers to a human that is eighteen years of age or older.
[0067] As used herein and unless otherwise specified, the phrase “such as” is intended to be open-ended. For example, the phrase “A can be a halogen, such as chlorine or bromine” means that “A” can be, but is not limited to, chlorine or bromine.
[0068] Reference to specific moieties, functional groups, or substituents contemplates (where applicable) tautomers thereof.
[0069] Unless otherwise stated, structures depicted herein include all isomeric (e.g., enantiomeric, diastereomeric, and geometric (or conformational)) forms of the structure (e.g., the R and S configurations for each asymmetric center, Z and E double bond isomers, and Z and E
conformational isomers). Unless otherwise stated, the compounds disclosed, taught, or otherwise suggested in this disclosure contemplate all single stereochemical isomers as well as enantiomeric, diastereomeric, and geometric (or conformational) mixtures thereof. Unless otherwise stated, the compounds disclosed, taught, or suggested in this disclosure contemplate all tautomeric forms thereof. Additionally, unless otherwise stated, structures depicted herein include compounds that differ only in the presence of one or more isotopically enriched atoms. Such compounds may be useful, for example, as analytical tools, as probes in biological assays, or as therapeutic agents. Additionally, incorporation of heavier isotopes such as deuterium (2H) may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increase in vivo half-life, or reduced dosage requirements.
[0070] Chemical entities described herein are further illustrated by the classes, subclasses, and species disclosed herein. For purposes of this disclosure, the chemical elements are identified in accordance with the Periodic Table of the Elements, CAS version. Handbook of Chemistry and Physics, 75th Ed., inside cover, and specific functional groups are generally defined as described therein. Additionally, general principles of organic chemistry, as well as specific functional moieties and reactivity, are described in Thomas Sorrell, Organic Chemistry, University Science Books, Sausalito, 1999; Smith and March, March's Advanced Organic Chemistry, 5th Edition, John Wiley & Sons, Inc., New York, 2001 ; Larock, Comprehensive Organic Transformations, VCH Publishers, Inc., New York, 1989; and Carruthers, Some Modem Methods of Organic Synthesis, 3rd Edition, Cambridge University Press, Cambridge, 1987. In this disclosure, any atom not specifically designated as a particular isotope is meant to represent any stable isotope of that atom.
[0071] Unless otherwise stated, structures depicted herein are also meant to include all stereoisomeric (e g., enantiomeric, diastereomeric, and geometric (or conformational)) forms of the structure; for example, the R and S configurations for each asymmetric center, Z and E double bond isomers, and Z and E conformational isomers. Therefore, the present compounds contemplate all single stereochemical isomers as well as enantiomeric, diastereomeric, and geometric (or conformational) mixtures thereof. Unless otherwise stated, the present compounds contemplate all tautomeric forms thereof. Additionally, unless otherwise stated, structures depicted herein are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms. Such compounds may be useful, for example, as analytical tools, as probes in biological assays, or as therapeutic agents. Additionally, incorporation of heavier isotopes such as deuterium (2H) may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increase in vivo half-life, or reduced dosage requirements.
[0072] Features and advantages of the subject matter hereof will become more apparent in light of the following detailed description of selected embodiments, as illustrated in the accompanying
figures. As will be realized, the subject matter disclosed and claimed is capable of modifications in various respects, all without departing from the scope of the claims. Accordingly, the drawings and the description are to be regarded as illustrative in nature, and not as restrictive and the full scope of the subject matter is set forth in the claims.
BRIEF DESCRIPTION OF THE DRAWINGS
[0073] Further features and advantages of the present disclosure will become apparent from the following detailed description, taken in combination with the appended drawings, in which:
[0074] Fig. 1 illustrates EC50 graphs for compounds of formula IV in optically pure form (racemate), and PURM100 and PURM200 which are enantiomeric forms of Compound IV, against 5- HTIA, 5-HT2A, 5-HT2B, 5-HT2C, and m5-HT2A. ECso values are summarised in Table 1.
[0075] Fig. 2 illustrates Concentration Response Curves for compounds of formula IV in optically pure form (racemate), and PURM100 and PURM200 which are enantiomeric forms of Compound IV, against 5-HT2A in agonist mode.
[0076] Fig. 3 illustrates the plasma concentration following intravenous (IV) or oral (PO) of the racemic mixture of Compound PURM100 and Compound PURM200.
[0077] Fig. 4 illustrates the plasma concentration following intravenous (IV), intraperinoneal (IPA or IPB) or oral (PO) of Compound PURM100.
[0078] Fig. 5 illustrates the plasma concentration following intravenous (IV), intraperinoneal (IPA or IPB) or oral (PO) of Compound PURM200.
[0079] Fig. 6 illustrates the head twitch response (HTR) in mice administered with 0.1 mg/kg to 10 mg/kg of the racemic mixture of Compound IV. HTR for the Compound IV is minimal in comparison to DOI as the control. Positive control is a racemic mixture of 2,5-Dimethoxy-4- iodoamphetamine (DOI), which is a a psychedelic drug and a substituted amphetamine.
[0080] Fig. 7 illustrates that in mice administered with 0.1 mg/kg to 3 mg/kg of the racemic mixture of Compound IV, there are no changes to locomotor activity.
[0081] Fig. 8 illustrates the results of Sholl Analysis which reveals that neurons treated with compounds PURM100 and PURM200 (which individually represent one of the enantiomeric forms of Compound IV) exhibit an increase in the number of process crossings in comparison to vehicle.
[0082] Fig. 9 illustrates the results of Sholl Analysis which reveals that neurons treated with compounds PURM100 or PURM200 in the presence of IPTG, exhibit an increase in the number of process crossings in comparison to vehicle.
[0083] Fig. 10 illustrates that neuronal stem cell cultures treated with compounds PURM100 and PURM200 (at 10 pM, which individually represent one of the enantiomeric forms of Compound IV) for 48h exhibit an increase in the number of branching of dendrites when compared to control treatments.
[0084] Fig. 11 illustrates that neuronal stem cell cultures treated with compounds PURM100 and PURM200 (at 10 pM, which individually represent one of the enantiomeric forms of Compound IV) for48h, in the presence of IPTG to silence the HT2A receptors. Statistical analysis indicates a significant effect of drug treatment on dendritogenesis produced only by BDNF treatment, implicating the 5-HT2A receptors in the effect observed with these compounds.
[0085] Fig. 12 illustrates the results shown in Figs. 10 and 11 that were normalized with respect to their corresponding vehicle (control) as fold over basal in order to compare them. Statistical analysis indicates a significant effect of both variables — drug treatment and expression of 5-HT2A receptors — and the interaction between them. This means that the effect of drugs on dendritogenesis requires the expression of 5-HT2A receptors.
[0086] Fig. 13 shows images of cells treated with water (control) and with Compound IV, where the treatment with compound IV for 48h causes an increase in the number of branching of dendrites when compared to control treatments.
[0087] Fig. 14 illustrates the expression of c-fos in neurons treated with the pure enantiomeric forms of Compound IV, namely PURM100 and PURM200, with or without IPTG. C-fos is a functional marker of neuronal activity and initiation of protein transcription cascades.
[0088] Fig. 15 illustrates the increase in expression of erg-1 , suggesting that PURM100 and PURM200 may activate neurons in absence of 5-HT2A signaling.
[0089] Fig. 16 illustrates the increase in expression of erg-2, suggesting that PURM100 and PURM200 may activate neurons in absence of 5-HT2A signaling.
[0090] It will be noted that throughout the appended drawings, like features are identified by like reference numerals.
DETAILED DESCRIPTION
[0091] In embodiments there are disclosed compounds of structural formula I, or a pharmaceutically acceptable salt thereof, and stereoisomers thereof:
R1 is OH, and when R1 is OH, R2 is different than OH and (i) selected from the group consisting of H, halogen, lower alkyl, CHF2, CF3, NO, OCH3, OCHF2, OCF3, SCHF2, SCH3, SCF3, and cyano; or (ii) together with a form a ring of 3 or 4 atoms, one atom of which is selected from the group consisting of C, N, O, and S, while the remainder are carbon, which chain contains 0, 1 , or 2 double bonds, and to which chain are attached substituents independently selected from the group consisting of H, halogen, OH, Ci-Ce alkoxy, Ci-Ce alkyl, C3-Ce cycloalkyl, CHF2, CF3, OCHF2, OCF3, SCH3, SCHF2, SCF3, cyano, and oxo;
R2 is OH, and when R2 is OH, R1 is different than OH and (i) selected from the group consisting of H, halogen, lower alkyl, CHF2, CF3, OCH3, OCHF2, OCF3, SCHF2, SCH3, SCF3, and cyano; or (ii) together with b or c form a ring of 6 to 8 atoms, one atom of which is selected from the group consisting of C, N, O, and S, while the remainder are carbon, which chain contains 1 , or 2 double bonds, and to which chain are attached substituents independently selected from the group consisting of H, halogen, OH, Ci-Ce alkoxy, Ci-Ce alkyl, C3-Ce cycloalkyl, CHF2, CF3, OCHF2, OCF3, SCH3, SCHF2, SCF3, cyano, and oxo;
R3 is (i) is selected from the group consisting of Ci-Ce alkyl, Ci-Ce substituted alkyl, C2-Ce alkenyl, C2-Ce alkynyl, C3-Ce cycloalkyl, (C3-Ce cycloalkyl)(Ci-C6 alkyl), C3-Ce heterocyclyl, (C3- C6 heterocyclyl)(Ci-C6 alkyl), aryl, aryl(Ci-C6 alkyl), heteroaryl, heteroaryl(Ci-C6 alkyl), CN, C(O)NH2, C(O)NH(CI-C6 alkyl), C(O)N(CI-C3 alkyl)(Ci-C6 alkyl), C(=NOH)(CI-C6 alkyl), and C(=NOH)(CI-C6 substituted alkyl), C(O)NH(aryl), C(O)N(alkyl)(aryl), NC, S(O)2NH2, S(O)2NH(Ci-Ce alkyl) and S(O)2NH(aryl); or (ii) together with R4 form a chain of 2 to 4 carbon atoms to which are attached substituents independently selected from the group consisting of H, Ci-Ce alkyl, aryl, and heteroaryl; or (iii) togetherwith a form a ring of 3 or4 atoms, one atom of which is selected from the group consisting of C, N, O, and S, while the remainder are
carbon, which chain contains 0, 1 , or 2 double bonds, and to which chain are attached substituents independently selected from the group consisting of H, halogen, OH, Ci-Ce alkoxy, Ci-C6 alkyl, C3-C6 cycloalkyl, CHF2, CF3, OCHF2, OCF3, SCH3, SCF3, cyano, and oxo; or (iv) is selected from the group consisting of H, Ci-Ce alkyl, Ci-Ce substituted alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, (C3-C6 cycloalkyl)(Ci-C6 alkyl), C3-C6 heterocyclyl, (C3-C6 heterocyclyl)(Ci-C6 alkyl), aryl, aryl(Ci-C6 alkyl), heteroaryl, heteroaryl(Ci-Ce alkyl), CN, C(O)NH2, C(O)NH(CI-C6 alkyl), C(O)N(CI-C3 alkyl)(Ci-C6 alkyl), C(=NOH)(CI-C6 alkyl), and C(=NOH)(CI-C6 substituted alkyl), if b is halogen, CH3, CHF2, CF3, OCH3, OCHF2, OCF3, SCH3, SCHF2, SCF3, or cyano, optionally, R3 together with a form any one of CH2CH2, CH2CH2CH2, CH2CH2CH2CH2, CH=CHCH=CH, OCH2CH2, CH2OCH2, CH2CH2O, OCH=CH, CH=CHO, OCH2O, SCH2CH2, CH2SCH2, CH2CH2S, SCH=CH, CH=CHS, NHCH2CH2, CH2NHCH2, CH2CH2NH, NHCH=CH, CH=CHNH, ON=CH, CH=NO, OCH=N, N=CHO, SN=CH, CH=NS, SCH=N, N=CHS, NHN=CH, CH=NNH, NHCH=N, N=CHNH, NHN=N, N=NNH, OCH2CH2CH2, CH2OCH2CH2, CH2CH2OCH2, CH2CH2CH2O, SCH2CH2CH2, CH2SCH2CH2, CH2CH2SCH2, CH2CH2CH2S NHCH2CH2CH2, CH2NHCH2CH2, CH2CH2NCH2, CH2CH2CH2NH, N=CHCH=CH, CH=NCH=CH, CH=CHN=CH, CH=CHCH=N; and optionally, wherein one hydrogen atom or two hydrogen atoms, if present on a moiety, are replaced with substituents selected independently from the group consisting of halogen, OH, Ci-Ce alkoxy, Ci-Ce alkyl, C3-C6 cycloalkyl, CHF2, CF3, OCHF2, OCF3, SCH3, SCFs.and cyano, or wherein two hydrogens, if attached to the same carbon atom, are replaced with an oxo group;
R4 is (i) is selected from the group consisting of H, Ci-Ce alkyl, Ci-Ce substituted alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, (C3-C6 cycloalkyl)(Ci-C6 alkyl), C3-C6 heterocyclyl, (C3- Ce heterocyclyl)(Ci-C6 alkyl), aryl(Ci-C6 alkyl), arylsulfonyl, heteroarylsulfonyl, , aryl(Ci-C6 alkyl)sulfonyl, (Ci-C6)alkylsufonyl and heteroaryl(Ci-C6 alkyl)sulfonyl; or (ii) together with R3 form a chain of 2 to 4 carbon atoms to which are attached substituents independently selected from the group consisting of H, Ci-C6 alkyl, aryl, heteroaryl, and any combination thereof;
R5 is H, D, Ci-Ce alkyl, Ci-Ce substituted alkyl;
R6: (i) is selected from the group consisting of H, Ci-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3- Ce cycloalkyl, (C3-C6 cycloalkyl)(Ci-Ce alkyl), aryl(Ci-Ce alkyl), acetyl, and heteroaryl(Ci-Ce alkyl); or (ii) together with R7 and the N atom to which they are attached form a 4-7 membered heterocyclyl ring; or (iii) together with e and the N atom to which R6 is attached form an azetidine or pyrrolidine ring, such ring carrying substituents independently selected from the group consisting of H, aryl, heteroaryl, Ci-Ce alkyl, and C3-C6 cycloalkyl; or (iv) together with b
and the N atom to which R6 is attached form an azetidine or pyrrolidine ring, such ring carrying substituents independently selected from the group consisting of H, aryl, heteroaryl, halogen, Ci-Ce alkyl, and C3-C6 cycloalkyl;
R7: (i) is selected from the group consisting of H, Ci-Ce alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3- Ce cycloalkyl, (C3-C6 cycloalkyl)(Ci-Ce alkyl), aryl(Ci-Ce alkyl), acetyl, and heteroaryl(Ci-Ce alkyl); or (ii) together with R6 and the N atom to which they are attached form a 4-7 membered heterocyclyl ring; a: (i) is selected from a group consisting of H, halogen, CH3, CHF2, CF3, OCH3, OCHF2, OCF3, SCH3, SCHF2, SCF3, NC and cyano; or (ii) together with a form a ring of 3 or 4 atoms, one atom of which is selected from the group consisting of C, N, O, and S, while the remainder are carbon, which chain contains 0, 1 , or 2 double bonds, and to which chain are attached substituents independently selected from the group consisting of H, halogen, OH, Ci-Ce alkoxy, Ci-Ce alkyl, C3-C6 cycloalkyl, CHF2, CF3, OCHF2, OCF3, SCH3, SCHF2, SCF3, cyano, and oxo; or (iii) together with R3 form a ring of 3 or 4 atoms, one atom of which is selected from the group consisting of C, N, O, and S, while the remainder are carbon, which chain contains 0, 1 , or 2 double bonds, and to which chain are attached substituents independently selected from the group consisting of H, halogen, OH, Ci-Ce alkoxy, Ci-Ce alkyl, C3-C6 cycloalkyl, CHF2, CF3, OCHF2, OCF3, SCH3, SCHF2, SCF3, cyano, and oxo; and b, c, d, and e, are each independently H; or three of b, c, d, and e are H and the remaining substituent is a lower alkyl group; or b and e are each H, and c and d together are -CH2- or - CH2CH2-, thereby giving rise to a cyclopropane or cyclobutane ring; or b, c, and d are each H, and e, R6, and the N atom to which R6 is attached form together an azetidine or pyrrolidine ring, such ring carrying substituents independently selected from the group consisting of H, aryl, heteroaryl, Ci-Ce alkyl, and C3-C6 cycloalkyl; or c, d, and e are each H, and b, R6, and the N atom to which R6 is attached form together an azetidine or pyrrolidine ring, such ring carrying substituents independently selected from the group consisting of H, aryl, heteroaryl, halogen, Ci-C6 alkyl, and C3-C6 cycloalkyl.
[0092] According to embodiments of compounds of structural formula I, or a pharmaceutically acceptable salt thereof, and stereoisomers thereof, the R1 is OH and R2 is H or the R1 is H and R2 is OH.
[0093] According to an embodiment of compounds of structural formula I, or a pharmaceutically acceptable salt thereof, and stereoisomers thereof, the compound is a compound of formula IV:
[0094] According to an embodiment of the compound of structural formula I, or a pharmaceutically acceptable salt thereof, and stereoisomers thereof, the compound is a compound of formula IV-S:
[0095] According to an embodiment of the compound of structural formula I, or a pharmaceutically acceptable salt thereof, and stereoisomers thereof, the compound is a compound of formula IV-R:
IV-/?.
[0096] According to an embodiment of the compound of structural formula I, or a pharmaceutically acceptable salt thereof, and stereoisomers thereof, the compound is a racemate of a compound of formula IV-S and a compound of formula IV-R:
[0097] According to an embodiment of compounds of structural formula I, or a pharmaceutically acceptable salt thereof, and stereoisomers thereof, the compound is a compound of formula V:
[00100] The compound of formula V may be a racemate of a compound of formula V-S and a compound of formula V-R:
[00101] According to other embodiments there are disclosed compounds of structural formula II, or a pharmaceutically acceptable salt thereof, and stereoisomers thereof:
wherein:
R2 is different than OH and (i) selected from the group consisting of H, halogen, lower alkyl, CHF2, CF3, OCH3, OCHF2, OCF3, SCHF2, SCH3, SCF3, and cyano; or (ii) together with a form a ring of 3 or4 atoms, one atom of which is selected from the group consisting of C, N, O, and S, while the remainder are carbon, which chain contains 0, 1 , or 2 double bonds, and to which
chain are attached substituents independently selected from the group consisting of H, halogen, OH, Ci-Ce alkoxy, Ci-Ce alkyl, C3-C6 cycloalkyl, CHF2, CF3, OCHF2, OCF3, SCH3, SCHF2, SCF3, cyano, and oxo;
R3 is (i) is selected from the group consisting of Ci-Ce alkyl, Ci-Ce substituted alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, (C3-C6 cycloalkyl)(Ci-C6 alkyl), C3-C6 heterocyclyl, (C3- Ce heterocyclyl)(Ci-C6 alkyl), aryl, aryl(Ci-C6 alkyl), heteroaryl, heteroaryl(Ci-Ce alkyl), CN, C(O)NH2, C(O)NH(CI-C6 alkyl), C(O)N(CI-C3 alkyl)(Ci-C6 alkyl), C(=NOH)(CI-C6 alkyl), and C(=NOH)(Ci-Ce substituted alkyl); or (ii) together with R4 form a chain of 2 to 4 carbon atoms to which are attached substituents independently selected from the group consisting of H, Ci- Ce alkyl, aryl, and heteroaryl; or (iii) together with a form a ring of 3 or 4 atoms, one atom of which is selected from the group consisting of C, N, O, and S, while the remainder are carbon, which chain contains 0, 1 , or 2 double bonds, and to which chain are attached substituents independently selected from the group consisting of H, halogen, OH, Ci-Ce alkoxy, Ci-Ce alkyl, C3-C6 cycloalkyl, CHF2, CF3, OCHF2, OCF3, SCH3, SCF3, cyano, and oxo; or (iv) is selected from the group consisting of H, Ci-Ce alkyl, Ci-Ce substituted alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, (C3-C6 cycloalkyl)(Ci-C6 alkyl), C3-C6 heterocyclyl, (C3-C6 heterocyclyl)(Ci-C6 alkyl), aryl, aryl(Ci-Ce alkyl), heteroaryl, heteroaryl(Ci-Ce alkyl), CN, C(O)NH2, C(O)NH(Ci-Ce alkyl), C(O)N(CI-C3 alkyl)(Ci-C6 alkyl), C(=NOH)(CI-C6 alkyl), and C(=NOH)(CI-C6 substituted alkyl), if b is halogen, CH3, CHF2, CF3, OCH3, OCHF2, OCF3, SCH3, SCHF2, SCF3, or cyano, optionally, R3 together with a form any one of CH2CH2, CH2CH2CH2, CH2CH2CH2CH2, CH=CHCH=CH, OCH2CH2, CH2OCH2, CH2CH2O, OCH=CH, CH=CHO, OCH2O, SCH2CH2, CH2SCH2, CH2CH2S, SCH=CH, CH=CHS, NHCH2CH2, CH2NHCH2, CH2CH2NH, NHCH=CH, CH=CHNH, ON=CH, CH=NO, OCH=N, N=CHO, SN=CH, CH=NS, SCH=N, N=CHS, NHN=CH, CH=NNH, NHCH=N, N=CHNH, NHN=N, N=NNH, OCH2CH2CH2, CH2OCH2CH2, CH2CH2OCH2, CH2CH2CH2O, SCH2CH2CH2, CH2SCH2CH2, CH2CH2SCH2, CH2CH2CH2S NHCH2CH2CH2, CH2NHCH2CH2, CH2CH2NCH2, CH2CH2CH2NH, N=CHCH=CH, CH=NCH=CH, CH=CHN=CH, CH=CHCH=N; and optionally, wherein one hydrogen atom or two hydrogen atoms, if present on a moiety, are replaced with substituents selected independently from the group consisting of halogen, OH, Ci-Ce alkoxy, Ci-Ce alkyl, C3-C6 cycloalkyl, CHF2, CF3, OCHF2, OCF3, SCH3, SCFs.and cyano, or wherein two hydrogens, if attached to the same carbon atom, are replaced with an oxo group;
R4 is (i) is selected from the group consisting of H, Ci-Ce alkyl, Ci-Ce substituted alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, (C3-C6 cycloalkyl)(Ci-Ce alkyl), C3-C6 heterocyclyl, (C3- Ce heterocyclyl)(Ci-C6 alkyl), aryl(Ci-Ce alkyl), arylsulfonyl, heteroarylsulfonyl, aryl(Ci-Ce
alkyl)sulfonyl, (Ci-Ce)alkylsufonyl and heteroaryl(Ci-Ce alkyl)sulfonyl; or (ii) together with R3 form a chain of 2 to 4 carbon atoms to which are attached substituents independently selected from the group consisting of H, Ci-Ce alkyl, aryl, heteroaryl, and any combination thereof;
R5 is H, D, Ci-Ce alkyl, Ci-Ce substituted alkyl
R6: (i) is selected from the group consisting of H, Ci-Ce alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3- Ce cycloalkyl, (C3-C6 cycloalkyl)(Ci-Ce alkyl), aryl(Ci-Ce alkyl), acetyl, and heteroaryl(Ci-Ce alkyl); or (ii) together with R7 and the N atom to which they are attached form a 4-7 membered heterocyclyl ring; or (iii) together with e and the N atom to which R6 is attached form an azetidine or pyrrolidine ring, such ring carrying substituents independently selected from the group consisting of H, aryl, heteroaryl, Ci-Ce alkyl, and C3-C6 cycloalkyl; or (iv) together with b and the N atom to which R6 is attached form an azetidine or pyrrolidine ring, such ring carrying substituents independently selected from the group consisting of H, aryl, heteroaryl, halogen, Ci-Ce alkyl, and C3-C6 cycloalkyl;
R7: (i) is selected from the group consisting of H, Ci-Ce alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3- Ce cycloalkyl, (C3-C6 cycloalkyl)(Ci-Ce alkyl), aryl(Ci-Ce alkyl), acetyl, and heteroaryl(Ci-Ce alkyl); or (ii) together with R6 and the N atom to which they are attached form a 4-7 membered heterocyclyl ring; a: (i) is selected from a group consisting of H, halogen, CH3, CHF2, CF3, OCH3, OCHF2, OCF3, SCH3, SCHF2, SCF3, and cyano; or (ii) together with a form a ring of 3 or 4 atoms, one atom of which is selected from the group consisting of C, N, O, and S, while the remainder are carbon, which chain contains 0, 1 , or 2 double bonds, and to which chain are attached substituents independently selected from the group consisting of H, halogen, OH, Ci-Ce alkoxy, Ci-Ce alkyl, C3-C6 cycloalkyl, CHF2, CF3, OCHF2, OCF3, SCH3, SCHF2, SCF3, cyano, and oxo; or (iii) together with R3 form a ring of 3 or 4 atoms, one atom of which is selected from the group consisting of C, N, O, and S, while the remainder are carbon, which chain contains 0, 1 , or 2 double bonds, and to which chain are attached substituents independently selected from the group consisting of H, halogen, OH, Ci-C6 alkoxy, Ci-C6 alkyl, C3-C6 cycloalkyl, CHF2, CF3, OCHF2, OCF3, SCH3, SCHF2, SCF3, cyano, and oxo; and b, c, d, and e, are each independently H; or three of b, c, d, and e are H and the remaining substituent is a lower alkyl group; or b and e are each H, and c and d together are -CH2- or - CH2CH2-, thereby giving rise to a cyclopropane or cyclobutane ring; or b, c, and d are each H, and e, R6, and the N atom to which R6 is attached form together an azetidine or pyrrolidine ring, such ring carrying substituents independently selected from the group consisting of H, aryl,
heteroaryl, Ci-Ce alkyl, and C3-C6 cycloalkyl; or c, d, and e are each H, and b, R6, and the N atom to which R6 is attached form together an azetidine or pyrrolidine ring, such ring carrying substituents independently selected from the group consisting of H, aryl, heteroaryl, halogen, Ci-Ce alkyl, and C3-C6 cycloalkyl.
[00102] According to an embodiment of compounds of structural formula II, or a pharmaceutically acceptable salt thereof, and stereoisomers thereof, the compound is a compound of formula IV:
[00103] According to an embodiment of the compound of structural formula II, or a pharmaceutically acceptable salt thereof, and stereoisomers thereof, the compound is a compound of formula IV-S:
IV-S.
[00104] According to an embodiment of the compound of structural formula II, or a pharmaceutically acceptable salt thereof, and stereoisomers thereof, the compound is a compound of formula IV-R:
IV-/?.
[00105] According to an embodiment of the compound of structural formula II, or a pharmaceutically acceptable salt thereof, and stereoisomers thereof, the compound is a racemate of a compound of formula IV-S and a compound of formula IV-/?:
[00106] According to other embodiments there are disclosed compounds of structural formula III, or a pharmaceutically acceptable salt thereof, and stereoisomers thereof:
wherein:
R1 is different than OH and (i) selected from the group consisting of H, halogen, lower alkyl, CHF2, CF3, OCH3, OCHF2, OCF3, SCHF2, SCH3, SCF3, and cyano; or (ii) together with b or c form a ring of 6 to 8 atoms, one atom of which is selected from the group consisting of C, N, O, and S, while the remainderare carbon, which chain contains 1 , or2 double bonds, and to which chain are attached substituents independently selected from the group consisting of H, halogen, OH, Ci-C6 alkoxy, Ci-C6 alkyl, C3-C6 cycloalkyl, CHF2, CF3, OCHF2, OCF3, SCH3, SCHF2, SCF3, cyano, and oxo;
R3 is (i) is selected from the group consisting of Ci-Ce alkyl, Ci-Ce substituted alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, (C3-C6 cycloalkyl)(Ci-C6 alkyl), C3-C6 heterocyclyl, (C3- Ce heterocyclyl)(Ci-C6 alkyl), aryl, aryl(Ci-C6 alkyl), heteroaryl, heteroaryl(Ci-Ce alkyl), CN, C(O)NH2, C(O)NH(CI-C6 alkyl), C(O)N(CI-C3 alkyl)(Ci-C6 alkyl), C(=NOH)(CI-C6 alkyl), and C(=NOH)(Ci-Ce substituted alkyl); or (ii) together with R4 form a chain of 2 to 4 carbon atoms to which are attached substituents independently selected from the group consisting of H, Ci- Ce alkyl, aryl, and heteroaryl; or (iii) together with a form a ring of 3 or 4 atoms, one atom of which is selected from the group consisting of C, N, O, and S, while the remainder are carbon, which chain contains 0, 1 , or 2 double bonds, and to which chain are attached substituents independently selected from the group consisting of H, halogen, OH, Ci-Ce alkoxy, Ci-Ce alkyl, C3-C6 cycloalkyl, CHF2, CF3, OCHF2, OCF3, SCH3, SCF3, cyano, and oxo; or (iv) is selected from the group consisting of H, Ci-Ce alkyl, Ci-Ce substituted alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, (C3-C6 cycloalkyl)(Ci-Ce alkyl), C3-C6 heterocyclyl, (C3-C6 heterocyclyl)(Ci-C6 alkyl), aryl, aryl(Ci-Ce alkyl), heteroaryl, heteroaryl(Ci-Ce alkyl), CN, C(O)NH2, C(O)NH(Ci-Ce alkyl), C(O)N(CI-C3 alkyl)(Ci-C6 alkyl), C(=NOH)(CI-C6 alkyl), and C(=NOH)(CI-C6 substituted alkyl), if b is halogen, CH3, CHF2, CF3, OCH3, OCHF2, OCF3, SCH3, SCHF2, SCF3, or cyano, optionally, R3 together with a form any one of CH2CH2, CH2CH2CH2, CH2CH2CH2CH2, CH=CHCH=CH, OCH2CH2, CH2OCH2, CH2CH2O, OCH=CH, CH=CHO, OCH2O, SCH2CH2, CH2SCH2, CH2CH2S, SCH=CH, CH=CHS, NHCH2CH2, CH2NHCH2, CH2CH2NH, NHCH=CH, CH=CHNH, ON=CH, CH=NO, OCH=N, N=CHO, SN=CH, CH=NS, SCH=N, N=CHS, NHN=CH, CH=NNH, NHCH=N, N=CHNH, NHN=N, N=NNH, OCH2CH2CH2, CH2OCH2CH2, CH2CH2OCH2, CH2CH2CH2O, SCH2CH2CH2, CH2SCH2CH2, CH2CH2SCH2, CH2CH2CH2S NHCH2CH2CH2, CH2NHCH2CH2, CH2CH2NCH2, CH2CH2CH2NH, N=CHCH=CH, CH=NCH=CH, CH=CHN=CH, CH=CHCH=N; and optionally, wherein one hydrogen atom or
two hydrogen atoms, if present on a moiety, are replaced with substituents selected independently from the group consisting of halogen, OH, Ci-Ce alkoxy, Ci-Ce alkyl, C3-C6 cycloalkyl, CHF2, CF3, OCHF2, OCF3, SCH3, SCFs.and cyano, or wherein two hydrogens, if attached to the same carbon atom, are replaced with an oxo group;
R4 is (i) is selected from the group consisting of H, Ci-Ce alkyl, Ci-Ce substituted alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, (C3-C6 cycloalkyl)(Ci-C6 alkyl), C3-C6 heterocyclyl, (C3- C6 heterocyclyl)(Ci-C6 alkyl), aryl(Ci-C6 alkyl), arylsulfonyl, heteroarylsulfonyl, aryl(Ci-C6 alkyl)sulfonyl, (Ci-C6)alkylsufonyl and heteroaryl(Ci-Ce alkyl)sulfonyl; or (ii) together with R3 form a chain of 2 to 4 carbon atoms to which are attached substituents independently selected from the group consisting of H, Ci-Ce alkyl, aryl, heteroaryl, and any combination thereof;
R5 is H, D, Ci-Ce alkyl, Ci-Ce substituted alkyl;
R6: (i) is selected from the group consisting of H, Ci-Ce alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3- Ce cycloalkyl, (C3-C6 cycloalkyl)(Ci-Ce alkyl), aryl(Ci-Ce alkyl), acetyl, and heteroaryl(Ci-Ce alkyl); or (ii) together with R7 and the N atom to which they are attached form a 4-7 membered heterocyclyl ring; or (iii) together with e and the N atom to which R6 is attached form an azetidine or pyrrolidine ring, such ring carrying substituents independently selected from the group consisting of H, aryl, heteroaryl, Ci-Ce alkyl, and C3-C6 cycloalkyl; or (iv) together with b and the N atom to which R6 is attached form an azetidine or pyrrolidine ring, such ring carrying substituents independently selected from the group consisting of H, aryl, heteroaryl, halogen, Ci-C6 alkyl, and C3-C6 cycloalkyl;
R7: (i) is selected from the group consisting of H, Ci-Ce alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3- Ce cycloalkyl, (C3-C6 cycloalkyl)(Ci-Ce alkyl), aryl(Ci-Ce alkyl), acetyl, and heteroaryl(Ci-Ce alkyl); or (ii) together with R6 and the N atom to which they are attached form a 4-7 membered heterocyclyl ring; a: (i) is selected from a group consisting of H, halogen, CH3, CHF2, CF3, OCH3, OCHF2, OCF3, SCH3, SCHF2, SCF3, and cyano; or (ii) together with a form a ring of 3 or 4 atoms, one atom of which is selected from the group consisting of C, N, O, and S, while the remainder are carbon, which chain contains 0, 1 , or 2 double bonds, and to which chain are attached substituents independently selected from the group consisting of H, halogen, OH, Ci-C6 alkoxy, Ci-Ce alkyl, C3-C6 cycloalkyl, CHF2, CF3, OCHF2, OCF3, SCH3, SCHF2, SCF3, cyano, and oxo; or (iii) together with R3 form a ring of 3 or 4 atoms, one atom of which is selected from the group consisting of C, N, O, and S, while the remainder are carbon, which chain contains 0, 1 , or 2 double bonds, and to which chain are attached substituents independently selected from
the group consisting of H, halogen, OH, Ci-Ce alkoxy, Ci-Ce alkyl, C3-C6 cycloalkyl, CHF2, CF3, OCHF2, OCF3, SCH3, SCHF2, SCF3, cyano, and oxo; and b, c, d, and e, are each independently H; or three of b, c, d, and e are H and the remaining substituent is a lower alkyl group; or b and e are each H, and c and d together are -CH2- or - CH2CH2-, thereby giving rise to a cyclopropane or cyclobutane ring; or b, c, and d are each H, and e, R6, and the N atom to which R6 is attached form together an azetidine or pyrrolidine ring, such ring carrying substituents independently selected from the group consisting of H, aryl, heteroaryl, Ci-Ce alkyl, and C3-C6 cycloalkyl; or c, d, and e are each H, and b, R6, and the N atom to which R6 is attached form together an azetidine or pyrrolidine ring, such ring carrying substituents independently selected from the group consisting of H, aryl, heteroaryl, halogen, Ci-Ce alkyl, and C3-C6 cycloalkyl.
[00107] According to an embodiment of compounds of structural formula III, or a pharmaceutically acceptable salt thereof, and stereoisomers thereof, the compound is a compound of formula V:
[00110] The compound of formula V may be a racemate of a compound of formula V-S and a compound of formula V-R:
[00111] According to other embodiments there are disclosed compounds of structural formula VI, or a pharmaceutically acceptable salt thereof, and stereoisomers thereof:
wherein:
R1 is OH, and when R1 is OH, R2 is different than OH and selected from the group consisting of H, halogen, lower alkyl, CHF2, CF3, NO, OCH3, OCHF2, OCF3, SCHF2, SCH3, SCF3, and cyano;
R2 is OH, and when R2 is OH, R1 is different than OH and selected from the group consisting of H, halogen, lower alkyl, CHF2, CF3, OCH3, OCHF2, OCF3, SCHF2, SCH3, SCF3, and cyano;
R3 is selected from the group consisting of Ci-Ce alkyl, Ci-Ce substituted alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, (C3-C6 cycloalkyl)(Ci-C6 alkyl), C3-C6 heterocyclyl, (C3-C6 heterocyclyl)(Ci-C6 alkyl), aryl, aryl(Ci-C6 alkyl), heteroaryl, heteroaryl(Ci-C6 alkyl), ON, C(O)NH2, C(O)NH(CI-C6 alkyl), C(O)N(CI-C3 alkyl)(Ci-C6 alkyl), C(=NOH)(CI-C6 alkyl), and C(=NOH)(CI-C6 substituted alkyl), C(O)NH(aryl), C(O)N(alkyl)(aryl), NO, S(O)2NH2, S(O)2NH(CI-C6 alkyl) and S(O)2NH(aryl);
R4 is selected from the group consisting of H, Ci-Ce alkyl, Ci-Ce substituted alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, (C3-C6 cycloalkyl)(Ci-C6 alkyl), C3-C6 heterocyclyl, (C3-C6 heterocyclyl)(Ci-C6 alkyl), aryl(Ci-C6 alkyl), arylsulfonyl, heteroarylsulfonyl, aryl(Ci-C6 alkyl)sulfonyl, (Ci-C6)alkylsufonyl and heteroaryl(Ci-C6 alkyl)sulfonyl;
R5 is H, D, Ci-Ce alkyl, Ci-Ce substituted alkyl;
R6: is selected from the group consisting of H, Ci-Ce alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, (C3-C6 cycloalkyl)(Ci-Ce alkyl), aryl(Ci-Ce alkyl), acetyl, and heteroaryl(Ci-Ce alkyl);
R7: is selected from the group consisting of H, Ci-Ce alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, (C3-C6 cycloalkyl)(Ci-Ce alkyl), aryl(Ci-Ce alkyl), acetyl, and heteroaryl(Ci-Ce alkyl); a: is selected from a group consisting of H, halogen, CH3, CHF2, CF3, OCH3, OCHF2, OCF3, SCH3, SCHF2, SCF3, NC and cyano; and b, c, d, and e, are each independently H; or three of b, c, d, and e are H and the remaining substituent is a lower alkyl group.
[00112] According to other embodiments there are disclosed compounds of structural formula I, II, III, or VI or a pharmaceutically acceptable salt thereof, and stereoisomers thereof wherein a is halogen, preferably F. According to another embodiment, in the compounds of formula I, II or III, a is CF3. According to another embodiment, in the compounds of formula I, II, III, or VI a is H.
[00113] According to other embodiments there are disclosed compounds of structural formula I, II III, or VI or a pharmaceutically acceptable salt thereof, and stereoisomers thereof wherein R4 is H.
[00114] According to other embodiments there are disclosed compounds of structural formula I, II III, or VI or a pharmaceutically acceptable salt thereof, and stereoisomers thereof R5 is H.
[00115] According to other embodiments there are disclosed compounds of structural formula I, II III, or VI or a pharmaceutically acceptable salt thereof, and stereoisomers thereof wherein each of b, c, d, and e is H.
[00116] According to other embodiments there are disclosed compounds of structural formula I, II III, or VI or a pharmaceutically acceptable salt thereof, and stereoisomers thereof wherein each of R6 and R7 are CH3.
Methods of use
[00117] Indoline compounds described herein are believed to be useful in the treatment of drug resistant depression based on several clinical trials that have been reported using psilocybin itself.
[00118] A US STAR*D study has reported that more than half of all patients recruited through primary care and psychiatric clinics fail to achieve remission after first-line antidepressant treatment, and one-third were unable to experience remission after four courses of acute treatment (Rush AJ, Trivedi MH, Wisniewski SR, Nierenberg AA, Stewart JW, Warden D, et al. Acute and longer-term outcomes in depressed outpatients requiring one or several treatment steps: a STAR*D report. Am. J. Psychiatry 2006; 163:1905-17).
[00119] In addition to the potential use of the compounds of the present invention in the treatment of depression, of major depressive disorder, drug resistant depression, and psychotic depression. Other studies by third party groups of human volunteers have revealed that psilocybin can be used to treat tobacco and alcohol addiction. Moreover, in a controlled clinical environment, psilocybin was safely administered to subjects with OCD, and this drug treatment was found to lead to acute reductions in core OCD symptoms in several subjects (Moreno, F. A., Wiegand, C. B., Taitano, E. K., and Delgado, P.L. "Safety, tolerability, and efficacy of psilocybin in 9 patients with obsessive- compulsive disorder” J. Clin. Psychiatry 2006, 67,1735-1740).
[00120] Another potential use of the compounds of the present invention is in the treatment of seizure disorders, including but not limited to infantile seizure disorders such as but not limited to Dravet syndrome (Sourbon, J. et al. "Serotonergic Modulation as Effective Treatment for Dravet Syndrome in a Zebrafish Mutant Model”, ACS Chem. Neurosci. 2016, 7, 588-593).
[00121] The indoline compounds described herein are believed to be safer than psilocybin, given their lack of at least some of the undesirable characteristics of 5-HT2B-agonist related activities.
[00122] Other potential uses of compounds of the present invention is one or more of major depressive disorder, drug resistant depression, and psychotic depression, addiction including alcoholism, tobacco addiction, cocaine addiction, and opioid addiction, pain indications including
neuropathic pain, pain from chemotherapy associated neuropathy, phantom limb pain and fibromyalgia, inflammation (including chronic and acute), eating disorders including anorexia, autism, cluster headaches, migraines, dementia including Alzheimer’s dementia, Parkinson’s disease dementia, and Lewy body dementia, mild cognitive impairment, post-traumatic stress disorder, emotional distress associated with cancer, Fragile-X syndrome, autism spectrum disorder, bipolar disease, obsessive compulsive disease, and Rett syndrome.
[00123] Other potential uses of compounds of the present invention is as combination drug therapy, comprising: a compound according to the present invention of formulae I, II, III or IV; and a A/-methyl-D-aspartate (NMDA) receptor antagonist.
[00124] The NMDA receptor antagonist is at least one of ketamine, nitrous oxide, memantine, amantadine, noribogaine, dextromethorphan, dextrorphan, and dextromethadone, or a pharmaceutically acceptable salt, stereoisomer, or solvate thereof. Preferably, the NMDA receptor antagonist is dextromethorphan.
[00125] The combination drug therapy may be useful for the treatment of one or more of major depressive disorder, drug resistant depression, and psychotic depression, addiction including alcoholism, tobacco addiction, cocaine addiction, and opioid addiction, pain indications including neuropathic pain, pain from chemotherapy associated neuropathy, phantom limb pain and fibromyalgia, inflammation (including chronic and acute), eating disorders including anorexia, autism, cluster headaches, migraines, dementia including Alzheimer’s dementia, Parkinson’s disease dementia, and Lewy body dementia, mild cognitive impairment, post-traumatic stress disorder, emotional distress associated with cancer, Fragile-X syndrome, autism spectrum disorder, bipolar disease, obsessive compulsive disease, and Rett syndrome.
[00126] The combination drug therapy may be useful for the treatment of one or more of major depressive disorder, drug resistant depression, and psychotic depression.
Methods of Administration
[00127] As contemplated herein, a therapeutically effective amount of an indoline compound described herein is administered to a subject in need thereof. Whether such treatment is indicated depends on the subject case, and is further subject to medical assessment (diagnosis) that takes into consideration signs, symptoms, and/or malfunctions that are present, the risks of developing particular signs, symptoms and/or malfunctions, and other factors.
[00128] As contemplated herein, an indoline compound described herein may be administered by any suitable route known in the art. Such routes include, but are not limited to, oral, buccal, inhalation, topical, sublingual, rectal, vaginal, intracisternal or intrathecal through lumbar puncture, transurethral, nasal, percutaneous, transdermal, and parenteral administration (including intravenous, intramuscular, subcutaneous, intracoronary, intradermal, intramammary, intraperitoneal, intraarticular, intrathecal, retrobulbar, intrapulmonary injection and/or surgical implantation at a particular site). Parenteral administration may be accomplished using a needle and syringe or using a high-pressure technique.
[00129] Pharmaceutical compositions include those wherein an indoline compound described herein is present in a sufficient amount to be administered in an effective amount to achieve its intended purpose. The exact formulation, route of administration, and dosage is determined by a qualified medical practitioner in view of the diagnosed condition or disease. Dosage amount and interval can be adjusted individually to provide levels of an indoline compound described herein that is sufficient to maintain the desired therapeutic effects. It is possible that the indoline compound described herein may only require infrequent administration (e.g. monthly, as opposed to daily) to achieve the desired therapeutic effect.
[00130] As contemplated herein, a therapeutically effective amount of an indoline compound described herein adapted for use in therapy varies with the nature of the condition being treated, the length of time that activity is desired, and the age and the condition of the patient, and ultimately is determined by the attendant physician. Dosage amounts and intervals can be adjusted individually to provide plasma levels of the indoline compound that are sufficient to maintain the desired therapeutic effects. The desired dose conveniently may be administered in a single dose, or as multiple doses administered at appropriate intervals, for example as one, two, three, four, or more subdoses per day. Multiple doses often may be desired or required. For example, an indoline compound described herein may be administered at a frequency of: four doses delivered as one dose per day at four-day intervals (q4d x 4); four doses delivered as one dose per day at three-day intervals (q3d x 4); one dose delivered per day at five-day intervals (qd x 5); one dose per week for three weeks (qwk3); five daily doses, with two days’ rest, and another five daily doses (5/2/5); or, any dose regimen determined to be appropriate for the circumstance.
[00131] As contemplated herein, the indoline compounds described herein may be administered in admixture with a pharmaceutical carrier selected with regard to the intended route of administration and standard pharmaceutical practice. Pharmaceutical compositions for use in accordance with the indoline compounds described herein are formulated in a conventional manner
using one or more physiologically acceptable carriers comprising excipients and auxiliaries that facilitate processing of the compounds described herein.
[00132] Water is a preferred carrier when an indoline compounds described herein is administered intravenously. Saline solutions and aqueous dextrose and glycerol solutions may also be used as liquid carriers, particularly for injectable solutions. Suitable pharmaceutical carriers also include excipients such as starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene glycol, water, ethanol, and the like. The present compositions, if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents.
[00133] These pharmaceutical compositions may be manufactured, for example, by conventional mixing, dissolving, granulating, dragee-making, emulsifying, encapsulating, entrapping, or lyophilizing processes. Proper formulation is dependent upon the route of administration chosen. When a therapeutically effective amount of an indoline compound described herein is administered orally, the composition typically is in the form of a tablet, capsule, powder, solution, or elixir. When administered in tablet form, the composition additionally can contain a solid carrier, such as a gelatin or an adjuvant. The tablet, capsule, and powder contain about 0.01% to about 95%, and preferably from about 1 % to about 50%, of an indoline compound described herein. When administered in liquid form, a liquid carrier, such as water, petroleum, or oils of animal or plant origin, can be added. The liquid form of the composition can further contain physiological saline solution, dextrose or other saccharide solutions, or glycols. When administered in liquid form, the composition contains about 0.1 % to about 90%, and preferably about 1% to about 50%, by weight, of a compound described herein.
[00134] When a therapeutically effective amount of an indoline compound described herein described herein is administered by intravenous, cutaneous, or subcutaneous injection, the composition is in the form of a pyrogen-free, parenterally acceptable aqueous solution. The preparation of such parenterally acceptable solutions, having due regard to pH, isotonicity, stability, and the like, is within the skill in the art. A preferred composition for intravenous, cutaneous, or subcutaneous injection typically contains an isotonic vehicle. An indoline compound described herein described herein can be infused with other fluids over a 10-30 minute span or over several hours.
[00135] The indoline compounds described herein may be readily combined with pharmaceutically acceptable carriers well-known in the art. Such carriers enable the active agents to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a patient to be treated.
[00136] Pharmaceutical preparations for oral use can be obtained by adding an indoline compound described herein to a solid excipient, with or without grinding the resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores. Suitable excipients include, for example, fillers and cellulose preparations. If desired, disintegrating agents can be added.
[00137] An indoline compound described herein may be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion. Formulations for injection can be presented in unit dosage form, e.g., in ampules or in multidose containers, with an added preservative. The compositions can take such forms as suspensions, solutions, or emulsions in oily or aqueous vehicles, and can contain formulatory agents such as suspending, stabilizing, and/or dispersing agents.
[00138] Pharmaceutical compositions for parenteral administration include aqueous solutions of the active agent in water-soluble form. Additionally, suspensions of an indoline compounds described herein can be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils or synthetic fatty acid esters. Aqueous injection suspensions can contain substances which increase the viscosity of the suspension.
[00139] In some embodiments, the suspension also can contain suitable stabilizers or agents that increase the solubility of the compounds and allow for the preparation of highly concentrated solutions. Alternatively, a present composition can be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
[00140] An indoline compound described herein also may be formulated in rectal compositions, such as suppositories or retention enemas, e.g., containing conventional suppository bases. In addition to the formulations described previously, an indoline compound described herein also can be formulated as a depot preparation. Such long-acting formulations can be administered by implantation (for example, subcutaneously or intramuscularly) or by intramuscular injection. Thus, for example, an indoline compound described herein may be formulated with suitable polymeric or hydrophobic materials (for example, as an emulsion in an acceptable oil) or ion exchange resins.
[00141] An indoline compound described herein may be administered orally, buccally, or sublingually in the form of tablets containing excipients, such as starch or lactose, or in capsules or ovules, either alone or in admixture with excipients, or in the form of elixirs or suspensions containing flavoring or coloring agents. Such liquid preparations can be prepared with pharmaceutically acceptable additives, such as suspending agents. The indoline compounds described herein also may be injected parenterally, for example, intravenously, intramuscularly, subcutaneously, or
intracoronarily. For parenteral administration, the indoline compounds described herein may be best used in the form of a sterile aqueous solution which can contain other substances, for example, salts or monosaccharides, such as mannitol or glucose, to make the solution isotonic with blood. At least in some embodiments, indoline compounds described herein are psilocybin analogs.
[00142] The present invention will be more readily understood by referring to the following examples which are given to illustrate the invention rather than to limit its scope.
EXAMPLE 1
SEROTONIN RECEPTOR 5-HT2 FUNCTIONAL ASSAYS
[00143] Non-limiting examples of methods of measuring serotonin receptor functional activation are described as follows.
[00144] To measure serotonin receptor functional activation, either Gq dissociation by bioluminescence resonance energy transfer (BRET) or Gq-dependent calcium flux is performed for selected compounds. To measure 5-HT2 receptor-mediated Gq activation via Gq/y1 dissociation as measured by BRET (McCorvy JD, Wacker D, Wang S, Agegnehu B, Liu J, Lansu K, Tribo AR, Olsen RHJ, Che T, Jin J, Roth BL. Structural determinants of 5-HT2B receptor activation and biased agonism. Nat Struct Mol Biol. 2013; 25(9):787-96), HEK293T cells are sub-cultured in Dulbecco’s Modified Eagle Medium (DMEM) supplemented with 10% dialyzed fetal bovine serum (FBS) and are co-transfected in a 1 :1 :1 :1 ratio with RLuc3-fused human Gaq (Gaq- RLucS), a GFP2-fused to the C-terminus of human Gy1 (Gy1-GFP2), human Gp1 , and 5- HT2 receptor using TransiT-2020. After at least 18-24 hours, transfected cells are plated in poly-L-lysine coated 96-well white clear bottom cell culture plates in DMEM containing 1 % dialyzed FBS at a density of 25,000-40,000 cells in 200 pL per well and incubated overnight. The next day, medium is decanted, and cells are washed with 60 pL of drug buffer (1x HBSS, 20 mM HEPES, pH 7.4), then 60 pL of drug buffer is added per well. Cells are pre-incubated in a humidified atmosphere at 37°C before receiving drug stimulation. Drug stimulation utilized 30 pL addition of drug (3X) diluted in McCorvy buffer (1x HBSS, 20 mM HEPES, pH 7.4, supplemented with 0.3% BSA fatty acid free, 0.03% ascorbic acid), and plates are incubated for 1 hour at 37°C. Substrate addition occurred 15 minutes before reading and utilized 10 pL of the RLuc substrate coelenterazine 400a for Gq dissociation BRET2 (Prolume/Nanolight, 5 pM final concentration). Plates are read for luminescence at 400 nm and fluorescent GFP2 emission at 510 nm at 1 second perwell using a Mithras LB940 (multimode microplate reader (e.g. one provided by Berthold)). The BRET ratios of fluorescence/luminescence are calculated per well and are plotted as a function of drug concentration using Graphpad Prism 8 (Graphpad Software Inc., San Diego, CA). Data are normalized to % 5-HT
stimulation and analyzed using nonlinear regression “log(agonist) vs. response” to yield Emax and EC50 parameter estimates.
[00145] Calcium flux is measured using stable-expressing 5-HT2 Flp-ln 293 T-Rex Tetracycline inducible system by methods known in the art (e.g. Investigation of the Structure-Activity Relationships of Psilocybin Analogues, ACS Pharmacol. Trans Sei. 2020, Publication Date: December 14, 2020, https://doi.org/10.1021/acsptsci.0c00176). Cell lines are maintained in DMEM containing 10% FBS, 10 pg/mL Blasticidin, and 100 pg/mL hygromycin B. At least 20-24 hours before the assay, receptor expression is induced with tetracycline (2 pg/mL), and cells are seeded into 384-well poly-L-lysine- coated black plates at a density of 7,500 cells/well in DMEM containing 1% dialyzed FBS. On the day of the assay, the cells are incubated for 1 hour at 37JC with Fluo-4 Direct dye (Invitrogen, 20 pL/well) reconstituted in drug buffer (20 mM HEPES- buffered HBSS, pH 7.4) containing 2.5 mM probenecid. Drug dilutions are prepared at 5X final concentration in McCorvy buffer (20 mM HEPES-buffered HBSS, 0.1% BSA, 0.01 % ascorbic acid, pH 7.4). After dye load, cells are allowed to equilibrate to room temperature for 15 minutes, and then placed in a FLIPR®TETRA fluorescence imaging plate reader (Molecular Devices). The FLIPR®TETRA is programmed to read baseline fluorescence for 10 s (1 read/s), and afterward 5 pL of drug per well is added, and fluorescence is read for a total of 5-10 min (1 read/s). Fluorescence in each well is normalized to the average of the first 10 reads for baseline fluorescence, and then either maximum-fold peak increase over baseline or area under the curve (AUC) is calculated. Either peak or AUC is plotted as a function of drug concentration, and data are normalized to percent 5-HT stimulation. Data are plotted, and non-linear regression is performed using “log(agonist) vs. response” in Graphpad Prism 8 to yield Emax and EC50 parameter estimates.
[00146] The functional activity of various compounds disclosed herein at each of the 5-HT2A, 5- HT2B, and 5-HT2c receptors is measured against and relative to the functional activity of 4- hydroxytryptamine at those receptors.
EXAMPLE 2
METHOD OF CHEMICAL SYNTHESIS OF COMPOUND OF FORMULA IV
[00147] Non-limiting examples of procedures for preparing the compound of Formula IV described herein is provided below.
[00148] The indoline core of the compounds of Formula I is numbered as follows:
[00149] Analogous numbering applies for the compounds of Formula II, III and IV as the numbers are attached to the indole ring regardless of its substituents.
[00150] The synthesis of the compound of Formula IV comprises several steps, namely: (1) construction of the heterocyclic core; (2) installation or modification of the 3-substituent; and (3) functional group transformations.
EXAMPLE 3
METHOD OF CHEMICAL SYNTHESIS OF COMPOUND OF FORMULA V
[00151] Non-limiting examples of procedures for preparing the compound of Formula V described herein is provided below.
[00152] The synthesis of the compound of Formula V comprises several steps, namely: (1) construction of the heterocyclic core; (2) installation or modification of the 3-substituent; and (3) functional group transformations.
Pharmacology
EXAMPLE 4
SEROTONIN RECEPTOR 5-HT FUNCTIONAL ASSAY #1
[00153] Non-limiting examples of methods of measuring serotonin receptor functional activation are described as follows.
[00154] HEK293 cells expressing 5-HT receptors are trypsinised, counted and seeded in black, clear bottomed 96 well plates at a density of 25,000 cells per well and incubated overnight in media containing 1 % dialysed serum. Next day, media is removed from cell plates and replaced with 30 pl assay buffer (20 mM HEPES: HBSS, pH 7.4). Calcium 5 (Molecular Devices, R8186) dye solution (10 pl) is added to the wells and incubated at 37°C for 60 minutes. Dye solution is made up in 20 mM HEPES: HBSS, pH 7.4 + 2.5 mM probenecid. Compound dilutions (including serial dilutions) are performed in 100% DMSO then transferred to intermediate dilutions for a very limited amount of time (<10 minutes) just before adding to the cell plate. Testing of 3 compounds (racemate, compound PURM100 and compound PURM200) at 7 concentrations in triplicate against a selected panel of 5- HT Receptors assays (5-HTIA, 5-HT2A, 5-HT2B, 5-HT2C and m5-HT2A). Compounds are tested as agonists at 100 pM, 10 pM, 1 pM, 0.1 pM, 0.01 pM, 1 nM and 0.1 nM.
[00155] For agonist testing: the plates are placed in the FLIPR Penta High-Throughput Cellular Screening System, after incubation with dye, and fluorescence monitored every 1 second to monitor baseline. After 20 seconds test compound or reference agonist (10 pl) is added to the wells and the fluorescence monitored for 5 minutes at ex/emm: 488nm/510- 570nm. The ECso values for the test
compounds and reference compound were determined using GraphPad Prism software. The EC50 values for the reference compound are compared to historical assay and or literature data to ensure that it is within acceptable ranges in order to validate each assay. The % efficacy of test compounds measured against Emax of 5-HT is also determined.
[00156] The functional activity of the racemate of compound IV, compound PURM100 and compound PURM200 disclosed herein at each of the 5-HTIA, 5-HT2A, 5-HT2B, and 5-HT2C receptors was measured. A comparison of the functional activities is provided in Table 1 and in Fig. 1 , as follows.
Table 1 - 5-HT receptor binding profile
[00157] High potency: the compounds PURM100 and -R have high potency against the 5-HT2A receptor (below 60nM). High potency can lead to therapeutic effects using a smaller dose. Lower doses can lead to fewer adverse side effects and greater patient outcomes. High potency leads to improved target receptor engagement in complex biological systems (e.g., competition against endogenous molecules). The compounds have high selectivity for 5-HT2A in comparison to other 5-HT receptors. For example, an unexpected and approximately 13x greater potency at 5-HT2A than 5-HT2C, minimizing off-target 5-HT effects. Low risk for cardio toxicity: mitral and aortic valves are densely populated with 5-HT2B receptors; activation increases risk of valvular heart disease. The compounds PURM100 and - R lack of 5-HT2B activation.
EXAMPLE 5
SEROTONIN RECEPTOR 5-HT FUNCTIONAL ASSAY #2
[00158] HEK cells expressing LgBiT tagged 5-HT2A and smBiT Beta-Arrestin are washed with PBS, to remove serum containing media, harvested in 1% dialysed serum containing media, counted and seeded in white 96 well plates at a density of 25,000 cells per well. The following day 25pl of NanoGio live cell substrate (Promega N2012) is added to each well and cells incubated for 25 minutes. Compounds are prepared as described for the 5-HTR calcium assay procedure detailed in Examiner
4 above and 20pl of 5X compound solution added to the cells. Cells are incubated for 90 minutes at 37°C and plates read for luminescence using the Hidex™ plate reader.
[00159] The EC50 of the test compounds and reference compound were determined using Graph Pad Prism software. The EC50 values for the reference compound are compared to historical assay and or literature data to ensure that it is within our acceptable ranges in order to validate each assay. The % efficacy of test compounds was measured against Emax of 5-HT. The results in were generated by the excel formulae (compound RFU - average min RFU)/(average max RFU- average min RFU))*100. The EC50 values were generated using the equation log(agonist) v response (four parameters) by Prism software (GraphPad Inc). The the raw data was normalized to the top and bottom of the 5-HT EC50 values using Prism software. The EC50 values were generated using the equation log(agonist) v response (three parameters) by Prism software (GraphPad Inc).
Table 4 - activation of Ca2+/Gq pathway
[00160] Now referring to Fig. 2 and the tables above. Biased Gq signaling can activate phospholipase C (PLC), which hydrolyzes Phosphatidylinositol 4,5-bisphosphate (PIP2) into two second messengers, Inositol Trisphosphate (IP3) and Diacyclglycerol (DAG). These have several functions in cellular processes such as influencing cell proliferation and neurotransmitter release. The compound IV enantiomers are each more biased to the calcium/Gq signaling pathway than the racemic form. Reduced bias towards P-Arrestin2 suggests potentially less desensitization/downregulation of 5- HT2A receptors with the repeated administration of Compounds PURM100 and -R. Potency against the Gq pathway: Compound IB-/?, > Compound PURM100 > racemic mixture. The results of the assay measuring activation of Ca2+/Gq pathway indicate that the compounds IV all have high potency at 5- HT2A (below 60nM).
EXAMPLE 6
LIVER HEPATOCYTE STABILITY
[00161] Hepatocyte stability is a measure of how quickly a drug candidate is metabolized in the liver. A hepatocyte stability assay is an in vitro test that uses liver cells to determine how a compound is cleared. Hepatocyte stability assays can be used for a variety of purposes, including: Drug efflux, Uptake studies, Metabolite identification, and CYP induction/inhibition studies.
[00162] The hepatocyte stability assay protocol was carried out according to the following protocol:
Preparation of Working Solutions
[00163] 1) Prepare 10 mM stock solutions of test compounds and positive control compound in appropriate solvent (DMSO).
[00164] 2) In separate conical tubes, dilute the 10 mM test compound and positive control to
100 pM by combining 198 pL of 50% acetonitrile 150% water and 2 pL of 10 mM stock.
Preparation of Hepatocytes
[00165] 1) Incubation medium (William’s E Medium supplemented with GlutaMAX) and hepatocyte thawing medium were placed in a 37°C water bath, and allow warming for at least 15 minutes prior to use. Boiled hepatocytes was prepared in 100°C water bath at least 15 minutes.
[00166] 2) T ransfer a vial of cryopreserved hepatocytes from storage, ensuring that vials remain at cryogenic temperatures until thawing process ensues. Thaw the cells by placing the vial in a 37°C water bath and gently shaking the vials for 2 minutes. After thawing was completed, spray vial with 70% ethanol, transfer the vial to a biosafety cabinet.
[00167] 3) Use wide-bore pipette tip to transfer hepatocytes into 50 mL conical tube containing thawing medium. Place the 50 mL conical tube into a centrifuge and spin at 100 g for 10 minutes. Upon completion of spin, aspirate thawing medium and resuspend hepatocytes in enough incubation medium to yield ~1 .5 x 106 cells/mL.
[00168] 4) Using a AO/PI Staining, count cells and determine the viable cell density. Dilute cells with incubation medium to a working cell density of 0.5 x 106 viable cells/mL.
[00169] Procedure for Stability Determination
[00170] 1) Pipette 198 pL of hepatocytes and boiled hepatocytes into each wells of a 96-well non-coated plate. Place the plate in the incubator to allow the hepatocytes to warm for 10 minutes.
[00171] 2) Pipette 2 pL of the 100 pM test compound or positive control into respective wells of the 96-well non-coated plate to start the reaction. Return the plate to the incubator for the designed time points.
[00172] 3) T ransfer well contents in 25 pL aliquots at time points of 0.5, 15, 30, 60, 90 and 120 minutes. The aliquots were then mixed with 6 volumes (150 pL) of acetonitrile containing with internal standard, IS (100 nM alprazolam, 200 nM caffeine and 100 nM tolbutamide) to terminate the reaction. Vortex for 5 minutes. Samples were centrifuges for 45 minutes at 3,220 g. Aliquot of 100 pL of the supernatant was diluted by 100 pL ultra-pure water, and the mixture was used for LC/MS/MS analysis. All incubations were performed in duplicate.
[00173] Data Analysis
[00174] All calculations were carried out using Microsoft Excel. Peak areas were determined from extracted ion chromatograms. Determine the in vitro half-life (ti/2) of parent compound by regression analysis of the percent parent disappearance vs. time curve.
[00175] The in vitro half-life (in vitro ti/2) was determined from the slope value: in vitro ti/2 = -0.693 / k
[00176] Conversion of the in vitro ti/2 (in min) into the in vitro intrinsic clearance in vitro CLint, in L/min/1 x106 cells) was done using the following equation (mean of duplicate determinations): in vitro CLint = -kV/N
V = incubation volume (0.2 mL);
N = number of hepatocytes per well (0.1 x 106 cells).
[00177] The calculations of Scale-up CLint (mL/min/kg), Predicted Hepatic CLH (mL/min/kg) and Hepatic Extraction Ratio (ER) were done using the following equation:
Scale-up CLint = (0.693/TI/2) x (1 /(hepatocytes concentration (0.5 x 106 cells/mL))) x Scaling Factors
Predicted Hepatic CLH = (QH x Scale-up CLint x fub) I (QH + Scale-up CLint x fub),
ER = Predicted Hepatic CLH/QH
[00178] Where QH is the hepatic blood flow (mL/min/kg), fub is the fraction of unbound drug in plasma which is assumed to be 1. Table 5 below provides values for the scaling factors and other values used for these calculations.
aScaling Factor = (Hepatocellularity) x (Liver weight)
Table 5 - Scaling Factors for Intrinsic Clearance Prediction in Human, Monkey, Dog, Rat and Mouse Hepatocytes
Remaining % Processing Rules
If T-test with p<0.05 is obtained, report the calculated Clint value.
If T-test with p<0.05 is not obtained, then report 0 for Clint value >80% at 120 min when all the other data points fall in the range of 80%~ 120% (one data point within the range of 70%~130% is accepted), otherwise _ the experiment should be repeated. _
Always remove from the calculation all points with < 10% left of 0.5 min sample, but leave at least 2 points
<80% at 120 min |f -|-_fest wjth p<0.05 is obtained, report the calculated CLint value.
If T-test with p<0.05 is not obtained, the experiment must be repeated.
Table 8 - Hepatic clearance
[00179] The results show that compared to the verapamil control, the compound IV of the present invention exhibits slow hepatic clearance.
EXAMPLE 7
PLASMA AND BRAIN PHARMACOKINETICS IN MICE
[00180] The pharmacokinetics parameters were measured following intravenous (IV), Oral (PO), and intraperitoneal administration (IP) of either of the racemic mixture, Compound PURM100 or Compound PURM200 administration to CD-1 male mice. For IV administration, the compounds were administered at 2 mg/kg, for PO administration, the compounds were administered at 15 mg/kg, for IP administration, the compounds were administered at 5 mg/kg, in saline solution. Dosing solutions for IV was 0.4 mg/mL, PO was 3 mg/mL, and IP was 1 mg/mL. Sampling of blood was performed at 0.083,
0.25, 0.5, 1 , 2, 4 and 8 hours per dose (IV), or 0.25, 0.5, 1 , 2, 4 and 8 hours per dose (PO), or 0.25, 0.5, 1 , 2, 4 and 8 hours per dose (IP). Brain samples were collected 0.25, 1 , 4 and 8 hours per dose.
Tissue preparation
[00181] Brain samples were homogenized at a ratio of 1 :3 with PBS (W/V, 1 :3). The final brain concentration in the tissue was corrected by multiplying 4.
Sample preparation for plasma
[00182] The desired serial concentrations of working solutions were achieved by diluting stock solution of analyte with 50% acetonitrile in water solution. 5 pL of working solutions (1 , 2, 4, 10, 20, 100, 200, 1000, 2000 ng/mL) were added to 10 pL of blank plasma to achieve calibration standards of 0.5~1000 ng/mL (0.5, 1 , 2, 5, 10, 50, 100, 500, 1000 ng/mL) in a total volume of 15 pL. 4 quality control samples at 1 ng/mL, 2 ng/mL, 50 ng/mL and 800 ng/mL for plasma were prepared independently of those used for the calibration curves. These QC samples were prepared on the day of analysis in the same way as calibration standards.
[00183] 15 pL standards, 15 pL QC samples and 15 pL unknown samples (10 pL plasma with
5 pL blank solution) were added to 200 pL of acetonitrile containing IS mixture for precipitating protein respectively. Then the samples were vortexed for 30 s. After centrifugation at 4 “Celsius, 4700 rpm for 15 min, the supernatant was diluted with ultrapure water at a ratio of 1 :2 (V/V), then 10 pL of diluted supernatant was injected into the LC/MS/MS system for quantitative analysis.
Sample preparation forbrain
[00184] The desired serial concentrations of working solutions were achieved by diluting stock solution of analyte with 50% acetonitrile in water solution.15 pL of working solutions (1 , 2, 4, 10, 20, 100, 200, 1000, 2000 ng/mL) were added to 30 pL of blank brain homogenate to achieve calibration standards of 0.5~ 1000 ng/mL (0.5, 1 , 2, 5, 10, 50, 100, 500, 1000 ng/mL) in a total volume of 45 pL. 4 quality control samples at 1 ng/mL, 2 ng/mL, 50 ng/mL and 800 ng/mL for brain homogenate were prepared independently of those used for the calibration curves. These QC samples were prepared on the day of analysis in the same way as calibration standards.
[00185] 45 pL standards, 45 pL QC samples and 45 pL unknown samples (30 pL brain homogenate with 15 pL blank solution) were added to 200 pL of acetonitrile containing IS mixture for precipitating protein respectively. Then the samples were vortexed for 30 s. After centrifugation at 4 “Celsius, 4700 rpm for 15 min, the supernatant was diluted with ultrapure water at a ratio of 1 :2 (V/V), then 1 pL of diluted supernatant was injected into the LC/MS/MS system for quantitative analysis. Limit
of quantitation (LOQ) is 0.5ng/mL for plasma, 1 ng/mL for brain in this study; Upper limit of quantitation (UOQ) is 1000 ng/mL for plasma and brain in this study.
Table 11 - Brain Plasma Ratio for racemic mixture
Table 13 - Brain concentration and Kp following IP dose of 10 mpk for Compound PURM100
Table 15 - Brain concentration and Kp following IP dose of 10 mpk for Compound -PURM200 [00186] Brain penetrant: from the results above it can be seen, based on peak concentration, that both Compound PURM100 (KpCmax = 4.46) and Compound PURM200 (KpCmax = 7 49) effectively enter the brain shortly after administration (Compound PURM100 and -R, Tmax = 15 min).
[00187] Orally bioavailable: also, both Compound PURM100 (F = 70%) and Compound PURM200 (F = 80%) effectively enter the bloodstream when orally administered.
[00188] Drug exposure: the Compound IV racemic mixture is quickly removed from the body (IV Cl_obs = 110 mL/min/Kg) and plasma concentration is reduced by 50% 52 min after administration. Compound PURM100 shows similar clearance to the racemic mixture (IV Cl_obs = 71 mL/min/Kg) and plasma concentration is reduced by 50% 30 min after administration. Compound PURM200 clearance is the slowest (IV Cl_obs = 56 mL/min/Kg) and plasma concentration is reduced by 50% 58 min after administration. In vitro hepatic vs in vivo clearance suggest mechanisms beyond liver contribute to metabolism of these compounds.
EXAMPLE 8
HUMAN ETHER-A-GO-GO-RELATED GENE (HERG) ASSAY ON RACEMIC MIXTURE
[00189] human ether-a-go-go-related gene (hERG) is a potassium channel that plays a key role in the heart's electrical activity. In vitro hERG assays are a key part of assessing a drug’s liability for TdP (torsade de pointes) ventricular arrhythmias. The potential inhibitory effect of the racemic mixture
of the compound IV on hERG channel was evaluated by using manual patch-clamp system according to the protocol.
Cell lines and cell culture
[00190] HEK 293 cell line stably expressing hERG channel (Cat. K1236) was purchased from Invitrogen™. The cells are cultured in medium containing of 85% DMEM, 10% dialyzed FBS, 0.1 mM non-essential amino acid (NEAA), 25 mM HEPES, 100 U/mL Penicillin-Streptomycin, 5 pg/mL Blasticidin and 400 pg/mL Geneticin, cells grow in 25 cm2 cell culture bottles with 5% CO2 and 37°C. Cells are split using TrypLE™ Express about three times a week, and maintained between ~40% to ~80% confluence. Before the assay, cells were induced with doxycycline at 1 pg/mL for 48 hours. On the experiment day, the induced cells are resuspended and plated onto the coverslips about 5 x 105 cells /per 3.5 cm cell culture dish prior to use, cultured in medium without Blasticidin and Geneticin. Note: The passage of cell line used in safety evaluation assay is less than 60.
Solution preparations
. Vendor&Catalog MW/ stock _ . .. . ...
Chemical .. . .. Concentration mM
No. concentration ' '
NaCI Sigma S6191 58,44 132
KCI Sigma P5405 74,55 4
CaCI2 Sigma 21115 1 M 3
MgCh Sigma 63069 1 M 0,5
D-(+) Glucose sigma G7528 180,16 11 ,1
HEPES Gibco 15630080 1 M 10 pH: adjusted to 7.35 with NaOH.
Osmolarity range: 285~295.
The solution is filtered by filter system and stored at 4°C prior to use.
Table 16 - Extracellular solution
. Vendor&Catalog MW/ stock _ . .. . ...
Chemical .. . .. Concentration(mM)
No. concentration ' '
EGTA Sigma E3889 0.1 M 10
HEPES Solarbio H8090 238,3 10
KCI Sigma P5405 74,55 10
Osmolarity range: 285~295
Use stock solutions OOmM EGTA adjusted to pH 8.2 with KOH
Note: The solution is filtered by filter system and stored at 4°C prior to use
Table 17 - Intercellular solution
Working solution preparation for test compound
[00191] 1) Test compounds were initially prepared in DMSO with final concentration of 10 mM as stock solution.
[00192] 2)Then stock solution of each compound was serially diluted in ratio of 1 :3 with DMSO to prepare additional 3 intermediate solutions, including 3.33, 1.11 and 0.37 mM, respectively.
[00193] 3) Before the experiment, the working solutions are finally prepared by dilution of above-described intermediate solutions in 1000 folds using extracellular solution, so that the final concentration of working solution was 30, 10, 3.33, 1.11 and 0.37 pM, while 30 pM working solution was prepared by 333.333-folds dilution of 10 mM DMSO stock. The final DMSO concentration in working solutions was maintained in range of 0.1 -0.3% (v/v).
[00194] 4) hERG current in presence of 5 doses were tested for IC50 determination, consisting of 30, 10, 3.33, 1.11 and 0.37 pM.
[00195] 5) Dofetilide was initially prepared in DMSO with final concentration of 75 mM as stock solution.
[00196] 6) Then stock solution of dofetilide was serially diluted with DMSO to prepare additional
5 intermediate solutions, including 150, 50, 16.67, 5.56 and 1.85 pM, respectively.
[00197] 7) Before the experiment, the working solutions are finally prepared by dilution of above described intermediate solutions in 1000 folds using extracellular solution, so that the final
concentration of working solution was 150, 50, 16.67, 5.56 and 1 .85 nM. The final DMSO concentration in working solutions was maintained in range of 0.1% (v/v).
[00198] 8) hERG current in presence of 5 doses were tested for IC50 determination, consisting of 150, 50, 16.67, 5.56 and 1.85 pM.
Experimental procedure
[00199] 1 ) Remove the coverslip from the cell culture dish and place it on the microscope stage in bath chamber, fill the intracellular onto the glass electrode tip; then use thermal controller (Warner Instruments™, TC-324B) to allow the bath temperature of the bath chamber to reach 34 °C.
[00200] 2) Locate a desirable cell using the *10 objective. Locate the tip of the electrode under the microscope using the *10 objective by focusing above the plane of the cell. Once the tip is in focus, advance the electrode downwards towards the cell using the coarse controls of the manipulator, while simultaneously moving the objective to keep the tip in focus.
[00201] 3) When directly over the cell, switch to the 40* objective and use the fine controls of the manipulator to approach the surface of the cell in small steps.
[00202] 4) Apply gentle suction through the side-port of the electrode holder to form a giga-ohm seal.
[00203] 5) Use the Cfast to remove the capacity current that is in coincidence with the voltage step. Obtain the whole cell configuration by applying repetitive, brief, strong suction until the membrane patch has ruptured.
[00204] 6) Set membrane potential to -60 mV at this point to ensure that hERG channels are not open. The spikes of capacity current should then be cancelled using the Csiow on the amplifier.
[00205] 7) Set holding potential to -90 mV for 500 ms; record current at 20 kHz and filter at 10 kHz. Leaking current was tested at -80 mV for 500 ms.
[00206] 8) The hERG current was elicited by depolarizing at +30 mV for 4.8 seconds and then the voltage was taken back to -50 mV for 5.2 seconds to remove the inactivation and observe the deactivating tail current. The maximum amount of tail current size was used to determine hERG current amplitude.
[00207] 9) Record current for 120 seconds to assess the current stability. Only stable cells with recording parameters above threshold were applied for the drug administrations.
[00208] 10) Firstly vehicle control was applied to the cells to establish the baseline. Once the hERG current was found to be stabilized for 5 minutes, compound was applied. hERG current in the
presence of test compound were recorded for approximately 5 minutes to reach steady state and then 5 sweeps were captured. For dose response testing, 5 doses of compound was applied to the cells accumulatively from low to high concentrations. The positive control article, dofetilide at concentration of 450 nM was also applied to each cell post hERG current measurement at highest concentration of test compound as the internal low control for normalization of percentage inhibition. In order to ensure the good performance of cultured cells and operations, the positive control, Dofetilide, with 5 dosing concentrations was also used test the same batch of cells that were used for compounds.
Data analysis
[00209] Data acceptance criteria
[00210] The following criteria were used to determine data acceptability.
[00211] 1) Initial seal resistance > 1 GQ;
[00212] 2) Leak currents < 50% of the control peak tail currents at any time;
[00213] 3) Normal test pulse current waveform (e.g., hERG peak tail) current amplitude greater than prepulse current amplitude and the peak tail amplitude >250pA;
[00214] 4) Membrane resistance Rm > 500 MQ;
[00215] 5) Access resistance (Ra) < 15 MQ;
[00216] 6) Apparent run-down of peak current < 2.5% per min.
[00217] Data that met the above criteria for hERG current quality were further analyzed as the following steps.
[00218] 1) Percent current inhibition was calculated using the following equation. PatchMaster software was used to extract the peak current from the original data.
/ Peak tail currentcompoimd - Peak tail currcnt
\ Peak tail curr entBlank vehicle - Peak tail currcnt;)0S[t[ve contro
[00219] 2) The dose response curve of test compounds was plotted with %inhibition against the concentration of test compounds using Graphpad Prism 8.0, and fit the data to a sigmoid doseresponse curve with a variable slope.
[00220] Based on the above, the results are shown in the following table:
Table 18 - hERG assay results
[00221] The IC50 of the positive control Dofetilide is in line with the literature reports and exhibits good consistency between different batches of experiment, suggesting that current study running under appropriate condition and results were reliable. The IC50 <0.37 pM is reported if the percentage hERG inhibition is more than 50% in presence of this compound at 0.37 pM dose concentration. The IC50 > 30 pM was reported if the fitting results of IC50 was greater than 30 pM or if the maximum inhibition at 30 pM was lower than 50%. A generally acceptable ranking system used to identify the potency of a test compound inhibiting hERG channel is listed as follows: a) Low: IC50 > 10 pM; b) Moderate: 1 pM < IC50 < 10 pM, or c) High: IC50 < 1 pM. These results indicate that the compound IV of the present invention pose a low hERG cardiovascular safety risk.
EXAMPLE 9
SOLUBILITY ASSAY
[00222] The solubility of the Compound PURM100 of the present invention was evaluated in 0.9% saline.
Study Procedure
Preparation of stock solutions
[00223] The stock solutions of test compound and control compounds (progesterone and diclofenac) were prepared in DMSO at the concentrations of 10 mM.
Procedure for solubility determination
[00224] 15 pL of stock solution (10 mM) of each sample was placed in order into a 96-well rack.
485 pL of buffer was added into each vial of the cap-less Solubility Sample plate. The assay was performed in duplicate. Add one stir stick to each vial and seal using a molded PTFE/Silicone plug. Then the solubility sample plate was transferred to the Eppendorf® Thermomixer® Comfort plate shaker and shaken at 25°C at 1100 rpm for 2 hours. After completion of the 2 hours, plugs were removed and the stir sticks were removed using a big magnet, the samples from the Solubility Sample plate were transferred into the filter plate. Using the Vacuum Manifold, all the samples were filtered. Aliquot 5 pL filtrate and 5 pL DMSO followed by addition of 490 pL of a mixture of H2O and methyl alcohol containing internal standard (1 :1) as 100-fold diluted samples. Then 20 pL of 100-fold diluted samples are further diluted by addition of 180 pL of a mixture of H2O and methyl alcohol containing internal standard (1 :1) as 1000-fold diluted samples. A certain proportion of a mixture of H2O and methyl alcohol containing internal standard (1 :1) was used to dilute the diluent according to the peak
shape. The dilution factor was changed according to the solubility values and the LC-MS signal response.
[00225] Preparation of 300 pM standards (STD)
[00226] From the 10 mM DMSO STD plate, 6 pL was transferred into the remaining empty plate, and then 194 pL of DMSO was added to that plate to have a STD concentration of 300 pM. Aliquot 5 pL 300 pM STD and 5 pL buffer followed by addition of 490 pL of a mixture of H2O and acetonitrile containing internal standard (1 :1) as 100 fold diluted STD concentration of 3 pM. Then 20 pL of 100-fold diluted STD samples are further diluted by addition of 180 pL of a mixture of H2O and methyl alcohol containing internal standard (1 :1) as 1000-fold diluted STD to have a final STD concentration of 0.3 pM. A certain proportion of a mixture of H2O and methyl alcohol containing internal standard (1 :1) was used to dilute the diluent according to the peak shape. The concentrations of the standard samples were changed according to the LC-MS signal response
Procedure for sample analysis
[00227] The plate was placed into the well plate autosampler. The samples were evaluated by LC-MS/MS analysis.
Data analysis
[00228] All calculations were carried out using Microsoft® Excel®.
[00229] The filtrate was analyzed and quantified against a standard of known concentration using LC coupled with mass spectral peak identification and quantitation. Solubility values of the test compound and control compound were calculated as follows:
[00230] DF means the dilution factor. Any value of the compounds that was not within the specified limits was rejected and the experiment was repeated.
Table 19 - The solubility data of test compound and control compounds in 0.9% saline
[00232] The upper limit for this assay was set at 300 pM. Any value close to or above 300 pM indicates that the compound may have a solubility at or above 300 pM.
EXAMPLE 10
PROTEIN BINDING MEASUREMENT OF COMPOUNDS IN HUMAN AND MOUSE PLASMA BY USING EQUILIBRIUM DIALYSIS METHOD
Study Procedure
Preparation of compound working solutions
[00233] The working solution of test Compounds PURM100 and PURM200 and control compound were prepared in DMSO at the concentration of 1 mM.
Preparation of buffer solution pH 7.4
[00234] A basic solution was prepared by dissolving 14.2 g/L Na2HPO4 and 8.77 g/L NaCI in deionized water and the solution could be stored at 4°C for up to 7 days. An acidic solution was prepared by dissolving 12.0 g/L NaH2PO4 and 8.77 g/L NaCI in deionized water and the solution could be stored at 4°C for up to 7 days. The basic solution was titrated with the acidic solution to pH 7.4 and store at 4°C for up to 7 days. pH was checked on the day of experiment and was adjusted if outside specification of 7.4 ± 0.1 .
Preparation of plasma
[00235] Set the temperature of water bath to 37°C. Thaw the frozen Plasma (stored at -80°C) immediately in a 37°C water bath. Plasma samples were obtained from mixed gender human plasma samples as well as mix gender CD-1 mice.
Preparation of operation plate
[00236] Soak the dialysis membranes in ultrapure water for 60 minutes to separate strips, then in 20% ethanol for 20 minutes, finally in dialysis buffer for 20 minutes. Load the prepared membranes into the dialysis device and install the device again following manufacturers guidelines. Turn on air bath and allow to pre-heat to 37°C. If the dialysis membranes are not used immediately, they can be store at 4°C for no longer than 4 weeks.
Preparation of control sample at 0 hour
[00237] Add 597 pL of blank plasma solution into each vial of a new plastic plate or separate plastic tube by addition of 3 pL of the working solution of test compound, vortex at 1000 rpm for 2 minutes. The final percent volume of organic solvent is 0.5% and the final concentration for test compound is 5 pM. Immediately transfer 50 pL of the spiked plasma solution suspension to a 96-well
plate to act as T=0 control sample. The samples are treated the same as the samples after incubation. Place all remaining spiked plasma solution in the incubator for the duration of the study.
Stability determination of test compound in plasma solution
[00238] At the same time, the remaining spiked plasma solution sample in the plastic plate or separate plastic tube is incubated for 6 hours at 37°C with 5% CO2 in the CO2 incubator.
[00239] At T=6 hours, transfer 50 pL of the original spiked plasma solution suspension to the 96-well plate for analysis."
Procedure for equilibrium dialysis
[00240] Assemble the dialysis set up following the manufacturer’s instructions. Load cells with 120 pL of plasma sample and dialyzed against equal volume of dialysis buffer (PBS). The assay is performed in duplicate. Cover the unit with gas permeable lid and incubate for 6 hours at 37°C at 100 rpm with 5% CO2 on an orbital shaker in the CO2 incubator. At the end of incubation, remove lid and pipette 50 pL of post-dialysis samples from both buffer and plasma solution chambers into separated 96-well plate for analysis, respectively.
Procedure for sample preparation
[00241] Add 50 pL of plasma solution to the buffer samples, and an equal volume of PBS to the collected plasma solution samples. Shake the plate at 1000 rpm for 2 minutes and add 400 pL of acetonitrile containing an appropriate internal standard (IS) to precipitate protein and release compound. Vortex at 1000 rpm for 10 minutes. Centrifuge for 30 minutes at 3,220 g. Then transfer 100 pL of the supernatant to new 96-well plates for analysis. Add 100 pL of distilled water to each sample and mix for analysis by LC-MS/MS.
Data Analysis
[00242] All calculations are carried out using Microsoft Excel. Determine the concentrations of test compound and control compound in the buffer and plasma solution chambers. Calculate the percentages of test compound(s) and control compound bound as follows:
% Unbound — (Area ratio buffer chamber / Area ratio plasma solution chamber) x 100
% Bound = 100 - % Unbound
% Recovery — (Area ratio buffer chamber + Area ratio plasma solution chamber) I (Area ratio Total sample) x 100
% Remaining = Area ratio 6hr / Area ratio ohr x 100
Ketoconazole in human and mouse plasma
[00244] The results show that the free fraction of drug is very good, and thus both compounds will be available to interact with the target receptors.
EXAMPLE 11
PROTEIN BINDING MEASUREMENT OF COMPOUNDS IN MOUSE BRAIN HOMOGENATE USING EQUILIBRIUM DIALYSIS METHOD
Study procedure
Preparation of buffer solution pH 7.4
[00245] A basic solution was prepared by dissolving 14.2 g/L Na2HPO4 and 8.77 g/L NaCI in deionized water and the solution could be stored at 4°C for up to 7 days. An acidic solution was prepared by dissolving 12.0 g/L Na2HPO4 and 8.77 g/L NaCI in deionized water and the solution could be stored at 4°C for up to 7 days. The basic solution was titrated with the acidic solution to pH 7.4 and store at 4°C for up to 7 days. pH was checked on the day of experiment and was adjusted if outside specification of 7.4 ± 0.1 .
Preparation of blank brain homogenate
[00246] Frozen mouse brain tissues were thawed immediately at room temperature. Then, it will be weighted and homogenized with buffer by brain weight (g) to buffer volume (mL) ratio 1 :4. The mouse brain tissues were from male CD1 mice.
Preparation of working solutions
[00247] The working solution of test compound and control compound was prepared in DMSO at the concentration of 200 pM.
Preparation of operation plate
[00248] Soak the dialysis membranes in ultrapure water for 60 minutes to separate strips, then in 20% ethanol for 20 minutes, finally in dialysis buffer for 20 minutes. Load the prepared membranes into the dialysis device and install the device again following manufacturers guidelines. Turn on air bath and allow to pre-heat to 37°C. If the dialysis membranes are not used immediately, they can be store at 4°C for no longer than 4 weeks.
Preparation of control sample at 0 hour
[00249] Add 597 pL of blank brain homogenate into each vial of a new plastic plate or separate plastic tube by addition of 3 pL of the working solution of each cassette, vortex at 1000 rpm for 5 minutes. The final percent volume of organic solvent is 0.5% and the final concentration for test compound is 1 pM. Immediately transfer 50 pL of the spiked brain homogenate suspension to a 96- well plate to act as T=0 control sample. The samples are treated the same as the samples after incubation. Place all remaining spiked brain homogenate in the incubator for the duration of the study.
Stability determination of test compound in brain homogenate
[00250] At the same time, the remaining spiked brain homogenate sample in the plastic plate or separate plastic tube is incubated for 6 hours at 37°C with 5% CO2 in the CO2 incubator. At T=6 hours, transfer 50 pL of the original spiked brain homogenate to the 96-well plate for analysis.
Procedure for eguilibrium dialysis
[00251] Assemble the dialysis set up following the manufacturer’s instructions. Load cells with 120 pL of brain homogenate sample and dialyzed against equal volume of dialysis buffer (PBS). The assay is performed in duplicate. Seal the dialysis plate and place the plate in an incubator at 37°C with 5% CO2 at approximately 100 rpm for 6 hours. At the end of dialysis, remove seal and pipette 50 pL each of post-dialysis samples from both buffer and brain homogenate chambers into separate tubes in plate.
Procedure for sample preparation
[00252] Add 50 pL of mouse brain homogenate to the buffer samples, and an equal volume of PBS to the collected brain homogenate samples. Shake the plate at 1000 rpm for 2 minutes and add 400 pL of acetonitrile containing an appropriate internal standard (IS) to precipitate protein and release compound. Vortex at 1000 rpm for 10 minutes. Centrifuge for 30 minutes at 3,220 g. Transfer 250 pL of the supernatant to new 96-well plates and centrifuge again (3,220 g, 30 minutes). Then transfer 100 pL of the supernatant to new 96-well plates for analysis. Add 100 pL of distilled water to each sample and mix for analysis by LC-MS/MS. All compounds are tested in duplicate at 1 pM in mouse brain homogenate.
Data analysis
[00253] All calculations are carried out using Microsoft Excel.
[00254] Calculate the percentage of unbound, percentage of bound and recovery of test compound as follows:
% Unbound homogenate- (Area ratio buffer chamber I Area ratio homogenate chamber) x 100
% Unbound Brain=1/5/((1 /(% Unbound homogenate/100)-1 )+1 /5) X 1 00
% Bound Brain= 100 - % Unbound Brain
% Recovery — (Area ratio buffer chamber + Area ratio homogenate chamber) I (Area ratio Total sample) x 100
% Remaining = Area ratio 6hr / Area ratio ohr x 100
Table 21 - Protein binding results of test compounds PURM100 and PURM200 and control compound Telmisartan in mouse brain homogenate
[00256] The results show that the free fraction of drug is very good, and thus both compounds will be available to interact with the target receptors.
EXAMPLE 12
NON-HALLUCINOGENIC NEUROPLASTOGEN ASSAY
[00257] This experiment was carried out according to the method presented in de la Fuente Revenga, M. et al. Nature (2019) 9:14247
Animals
[00258] Experiments were performed on adult (10-20 weeks old) C57BL/6 male mice (Taconic), unless otherwise stated. Animals were housed in cages with up to 5 littermates at 12 h light/dark cycle at 23°C with food and water ad libitum, except during behavioral testing.
[00259] Experiments involving the use of 5-HT2AR knockout (5-HT2A-KO) mice were performed on adult (10-20 weeks old) 129S6/SvEv male mice. 5-HT2A-KO and controls in these experiments denote Htr2a-i- and Htr2a+/+ individuals, respectively, born to heterozygote (Htr2a+i-) breeders.
[00260] Surgical implantation of the magnet was adapted from a previously reported protocol (Hanks, J. B. & Gonzalez-Maeso). Mice were anesthetized using a ketamine and xylazine (120 mg/kg and 12 mg/kg, respectively). A circular incision was performed to remove part of the scalp. Membranous tissue in and around the area was repeatedly cleaned with H2O2 and dried until the bone surface was exposed. Dental cement was applied in the exposed area to attach a small neodymium magnet (6.0, 6.0, 1 .5 mm, 490 mg) with the south pole facing the bone surface. The implant remained exposed with the dental cement bordering the skin. The mice were allowed to recover for 1 or 2 weeks. Animals were used repeatedly with a washout period of at least 1 week between tests.
[00261] Isoflurane anesthesia was rapidly induced at an initial dose of 2% (vol/vol). When the effects of the anesthesia were apparent the animals were quickly administered DOI (1 mg/kg, i.p.) and placed on a heating pad on the isoflurane chamber. The dose of isoflurane was maintained at 1.5% for the following 30 min. Animals were regularly monitored.
Drugs
[00262] (±)-2,5-Dimethoxy-4-iodoamphetamine hydrochloride (DOI), compound PURM100 and compound PURM200. All drugs were dissolved in saline (0.9% NaCI) to the appropriate volume (0.005-0.01 ml/g body weight) and concentration for administration (i.p.). Vehicle-treated condition denotes injection of saline (i.p.) to the equivalent volume of the drug administered.
Materials
[00263] Data acquisition was performed in non-overlapping ~500-turn enameled wire (30 AWG) coils supported in closed plastic containers (inner dimensions, 11 cm diameter x 14 cm tall) with both terminals of each coil connected to a phono preamplifier (Pyle PP444). The amplified signal output was recorded at a 1000 Hz sampling rate using a myDAQ (National Instruments) data acquisition system controlled through MATLAB (Mathworks, R2018a version, along with the Nl myDAQ support package). The analog input range for each channel was ±10 V with an ADC (analog-digital conversion) of 16 bits. The amplified signal amplitude rarely exceeded a ±2.5 V range which eliminated the risk of signal clipping. In total, we were able to perform the simultaneous recording of 4 coils per session in two data acquisition systems (2 coils per myDAQ unit), but these number can be scaled up by using additional data acquisition systems, or systems with more analog input channels. Visual identification of HTR was performed as previously described5 on clips recorded at 60 FPS and 1080p resolution. For visual identification of HTR as signals on the magnetometer recorded data, the raw data was processed on the MATLAB suite. The recording was band-passed through a digital Butterworth filter between 40-200 Hz as previously described in Halberstadt, A. L. & Geyer, M. A., and the filtered data plotted in parallel with a spectrogram heat-map. HTR were identified when: i) amplitude of the voltage
signal exceeded the background noise level, and ii) corresponded in time with a peak maximum in the region of 70-110 Hz in the spectrogram heat-map often accompanied by a harmonic between 40-60 Hz of comparable or greater magnitude.
[00264] Automated detection of HTR events relied on a script based on detection of individual events between 70-110 Hz. Recorded data was band-passed through a digital Butterworth filter between 70-110 Hz and transformed to absolute values. After double local maxima processing each original wavelet is transformed into a unipolar peak that is identified as HTR event when (i) peak prominence exceeds 0.075V, (ii) is separated from any other event by at least 200 ms, and (iii) has a width inferior to 90 ms at half the maximum value of prominence.
[00265] Experiments that included the recording of the piezo sensor output were carried out with a paper tube (46 cm height) surrounding the coil and open top so that the animal could jump vertically without escaping the container. Simultaneous recording of the coil and piezo sensor signal was done by connecting the amplified output of the coil to one analog input channel of the data acquisition system (as described above), and the output of the piezoelectric sensor (ceramic diaphragm, 27 mm brass disc) secured under the plastic container with tape directly into the other available analog input channel. The recording data from the piezo sensor baseline was corrected and the signal transformed to absolute values before performing a maxima analysis in the MATLAB environment. Employing the peak-picking function timestamps for maxima exceeding an absolute voltage of 0.3 V were annotated. The value for the 0.3 V threshold was determined experimentally as described above, and corresponded to ~8 standard deviations of the piezo sensor signal from mice injected with 1 mg/kg of DOI.
[00266] The matching analysis of HTR detection and annotated piezo sensor maxima timestamps was automated. Two events were matched if the HTR detection system timestamp fitted within the range defined by the piezo sensor maxima timestamp ±0.1 s.
[00267] Analytical reference (±)-2,5-dimethoxy-4-iodoamphetamine, 2,5-dimethoxy-4- bromophenethylamine were purchased from Cerilliant Corporation (Round Rock, TX). Acetonitrile, ammonium acetate, ethyl acetate, formic acid, hexane, methanol, sodium hydroxide and water were purchased from Fisher Scientific (Hanover Park, IL, USA). All reagents were ACS grade or higher.
Statistical analysis
[00268] Statistical analyses were performed with GraphPad™ Prism™ software version 8. Animals were randomly allocated into the different experimental groups. Experiments combining DOI, PURM100 and PURM200 were performed crossing groups with 1 week between tests. Statistical significance of experiments involving different treatment groups were assessed by one-way followed
by Bonferroni’s post hoc test, and in experiments involving two or more variables and treatment groups by two-way ANOVA followed by Bonferroni’s post hoc test. Statistical significance of experiments involving two treatment groups was assessed by Student’s t-test. The level of significance was chosen at P<0.05. All data are presented as mean±standard error of the mean (S.E.M). Correlation between visual identification vs. automated detection of HTR, and visual analysis of signal vs. automated detection of HTR were assessed by linear regression. Correlation between levels of DOI in whole blood, forebrain and HTR was assessed by Pearson’s correlation coefficient.
[00269] Now referring to Figs. 6 and 7. The results show that the racemic mixture, when administered in mice at a dose of 0.1 mg/kg to 10 mg/kg, causes minimal head twitch responses (a proxy for hallucinogenic responses) (Fig. 6), and causes no changes to locomotor activity in mice administered 0.1 mg/kg to 3 mg/kg (Fig. 7).
EXAMPLE 13
COMPOUNDS PURM100 AND PURM200 INDUCE POSITIVE STRUCTURAL CHANGES TO CULTURED NEURONS
Methods
[00270] Adherent neural stem cell lines (NSCs), previously generated from murine fetuses, were used as an in vitro experimental model. These cells are proliferative and susceptible to differentiation into neurons and glia by growing under appropriate cell culture conditions. The original NSC line was genetically engineered to harbor a shRNA sequence against the gene encoding the 5- HT2A receptor within its genome. The expression of this shRNA is regulated by a promoter modulated by the presence of IPTG in the cell culture medium. Experiments were conducted under both conditions, i.e., with cells treated or not treated with IPTG throughout the process.
[00271] After 5 days of differentiation when neurons acquired their phenotype, cells were submitted to drug treatments as follows:
- Vehicle: H20 or DMSO
- BDNF at [50 ng/ml]
- Psilocin at 10 pM
- Compound PURM100 at 10 pM
- Compound PURM200 at 10 pM
[00272] BDNF, Compound PURM100, and Compound PURM200 were dissolved in H2O, while
Psilocin was dissolved in DMSO.
[00273] Phase-contrast microscopy images were captured using a 40x objective after 48 hours of treatment. Neuronal dendritic arborization was evaluated through image Sholl analysis. The Sholl analysis tool, included in Imaged software, was employed to estimate dendritic complexity. This analysis entails establishing concentric circles from the center of the neuronal body and quantifying the number of intersections these circles make with the various dendritic processes emerging from the neuron soma. The resulting data can be graphed to depict the number of intersections in relation to the distance from the center of the neuron. The area under the curve (AUC) obtained from this graph was considered to compare and average the results from different experiments. Experiments were independently performed three times, with analysis of 20 individual neurons per experimental condition.
Results
[00274] Figs. 8 and 9 show representative graphs showing Sholl analysis from one of the three independent experiments with cells grown in the absence (Fig. 8) or presence (Fig. 9) of IPTG in the culture medium. Sholl Analysis revealed that neurons treated with these compounds exhibit an increase in the number of process crossings in comparison to vehicle treated neurons. PSI represents psilocin.
[00275] Figs. 10 and 11 are bar graph which represent the mean ± SEM of the AUC from three independent experiments (n = 3) corresponding to the condition without IPTG (Fig. 10) or with IPTG (Fig. 11). Statistical analysis of the results from Fig. 10 with One-way ANOVA resulted in a significant outcome, indicating an effect on neuronal dendritic arborization dependent on drug treatment. Bonferroni post hoc analysis to compare this data among the different treatment groups revealed a significant difference between all the drugs and their corresponding vehicle. No differences were found between Compound PURM100 and Compound PURM200 compared to the psilocin effect.
[00276] Fig. 11 shows results from the same experiment carried out in conditions including IPTG in the cell culture medium, where the expression of 5-HT2A receptors is silenced. Statistical analysis indicates a significant effect of drug treatment on dendritogenesis produced only by BDNF treatment, implicating the 5-HT2A receptors in the effect observed with Compound PURM100 and Compound PURM200.
[00277] Fig. 12 shows data from the results shown in Figs. 10 and 11 that were normalized with respect to their corresponding vehicle (control) as fold over basal in order to compare them. A two-way ANOVA was performed considering drug treatments and the presence of IPTG in the culture medium as variables. The statistical analysis indicates a significant effect of both variables — drug treatment
and expression of 5-HT2A receptors — and the interaction between them. This means that the effect of drugs on dendritogenesis requires the expression of 5-HT2A receptors.
[00278] Fig. 13 shows images of cells treated with water (control) and with a racemic mixture of Compound IV, where the treatment with compound IV for 48h causes an increase in the number of branching of dendrites when compared to control treatments.
CONCLUSION
[00279] Compound PURM100 and Compound PURM200 treatment of differentiated neural progenitors into neurons and glia induces a significant increase in dendritogenesis in neurons, to a similar extent as psilocin treatment carried out in parallel. Equivalent assays conducted in cells grown in the presence of IPTG to silence the expression of 5-HT2A receptors resulted in no effect of these drugs on dendritogenesis, in contrast to BDNF, which was included in the experiments as a positive control. In conclusion, treatment with Compound PURM100 and Compound PURM200 for 48 hours promotes neuronal plasticity, assessed as the increment of dendritic arborization, and this effect is dependent on the expression of 5-HT2A receptors.
EXAMPLE 14
COMPOUNDS PURM100 AND PURM200 INCREASES IMMEDIATE EARLY GENE EXPRESSION
Methods
[00280] Cell lines are the same as those used in neuronal plasticity experiments of Example 11 , and treatments were performed in parallel with the same drugs and at the same concentrations. In this case, after 5 days of differentiation, cells were treated with the different drugs and vehicle for 45 minutes at 37°C and 5% CO2. After incubation, cells were scraped and collected in 1 .5 mL centrifuge tubes.
[00281] RNA extraction was accomplished by centrifuging the samples at 800g for 5 minutes at 4°C. The supernatant was discarded, and cells were resuspended in 400 pL of RNA-Solv reagent (Omega Bio-Tek®). Samples were incubated for 5 minutes at room temperature, and 80 pL of chloroform was added. Samples were mixed by inversion, left to rest for 5 minutes at room temperature, and then centrifuged at 12,000g for 15 minutes at 4°C. The aqueous phases of the supernatants were collected and transferred to new centrifuge tubes, where the same volume of isopropanol was added. Once again, samples were mixed by inversion and allowed to rest for 10 minutes at room temperature. Samples were centrifuged once more at 12 000g for 10 minutes at 4°C, and this time the supernatants were discarded. Pellets were resuspended in 1000 pL of chilled 100% ethanol, and samples were centrifuged at 7600g for 10 minutes at room temperature. Supernatants
were discarded, pellets were set to dry for 10 minutes and then resuspended in 10 pL of DEPC water (diethyl pyrocarbonate in water 1 :1000). After 20 minutes at room temperature, RNA concentration was measured using a Nanodrop.
[00282] cDNA synthesis was performed using TakaraO’s PrimeScript™ RT Master Mix. For each sample, 500 ng of RNA, enough water to reach a final volume of 8 pL, and 2 pL of Takara’s PrimeScript™ RT Master Mix were added to a 1.5 mL centrifuge tube. Samples were incubated at 37°C for 15 minutes, and the reaction was stopped using a 5-second cycle at 85°C. cDNA was measured using a Nanodrop, and samples were then diluted with water to a final concentration of 100 ng/pL.
[00283] qPCRs were performed in BIO-RAD®’s clear 96-well Multiplate® PCR Plates™. The expression of c-fos, egr-1 , and egr-2 was tested for each biological replicate. The expression of each gene was measured using the median cq value of 4 technical replicates. Gene expression in each biological sample was normalized to the housekeeper gene GAPDH. Each qPCR plate was loaded with 16 wells worth of cDNA for each treatment for a total of 96 wells. Each condition was tested for our three genes of interest and the housekeeper (4 wells for each gene). 4 pL of cDNA and 6 pL of a master mix containing iTaq Universal SYBR® Green Supermix (5 pL per well), forward primer (0.5 pL per well), and reverse primer (0.5 pL per well) were added to each well.
[00284] Now referring to Figs. 14 - 16. The results show an increase in c-fos expression in neurons treated with Compound PURM100 and Compound PURM200 with or without IPTG (Fig. 14). C-fos is a functional marker of neuronal activity and initiation of protein transcription cascades. Cells treated with Compound PURM200 and IPTG show increase in expression of erg-1 and erg-2, suggesting that Compound PURM200 may activate neurons in absence of 5-HT2A signaling (Fig. 15 and Fig. 16).
[00285] While preferred embodiments have been described above and illustrated in the accompanying drawings, it will be evident to those skilled in the art that modifications may be made without departing from this disclosure. Such modifications are considered as possible variants comprised in the scope of the disclosure.
References
1 : Roche et al. A Virtual Screening Method for Prediction of the hERG Potassium Channel Liability of Compound Libraries. (2002) ChemBioChem. 3, 455-459.
2: Glenn E. Kirsch et al. Variability in the measurement of hERG potassium channel inhibition: effects of temperature and stimulus patter. (2004) Journal of Pharmacological and Toxicological Methods 50, 93-101.
3: Roger Marrannes et al. Computer programs to facilitate the estimation of time-dependent drug effects on ion channels. (2004) Computer Methods and Programs in Biomedicine 74, 167-181.
4: Hanks, J. B. & Gonzalez-Maeso, J. Animal models of serotonergic psychedelics. ACS Chem. Neurosci. 4, 33-42 (2013).
5: Halberstadt, A. L. & Geyer, M. A. Characterization of the head-twitch response induced by hallucinogens in mice. Psychopharmacology (Berl). 227, 727-739 (2013).
Claims
1. A compound of structural formula I, or a pharmaceutically acceptable salt thereof, and stereoisomers thereof:
wherein:
R1 is OH, and when R1 is OH, R2 is different than OH and (i) selected from the group consisting of H, halogen, lower alkyl, CHF2, CF3, NO, OCH3, OCHF2, OCF3, SCHF2, SCH3, SCF3, and cyano; or (ii) together with a form a ring of 3 or 4 atoms, one atom of which is selected from the group consisting of C, N, O, and S, while the remainder are carbon, which chain contains 0, 1 , or 2 double bonds, and to which chain are attached substituents independently selected from the group consisting of H, halogen, OH, Ci-Ce alkoxy, Ci-Ce alkyl, C3-C6 cycloalkyl, CHF2, CF3, OCHF2, OCF3, SCH3, SCHF2, SCF3, cyano, and oxo;
R2 is OH, and when R2 is OH, R1 is different than OH and (i) selected from the group consisting of H, halogen, lower alkyl, CHF2, CF3, OCH3, OCHF2, OCF3, SCHF2, SCH3, SCF3, and cyano; or (ii) togetherwith b or c form a ring of 6 to 8 atoms, one atom of which is selected from the group consisting of C, N, O, and S, while the remainder are carbon, which chain contains 1 , or 2 double bonds, and to which chain are attached substituents independently selected from the group consisting of H, halogen, OH, Ci-Ce alkoxy, Ci-Ce alkyl, C3-C6 cycloalkyl, CHF2, CF3, OCHF2, OCF3, SCH3, SCHF2, SCF3, cyano, and oxo;
R3 is (i) is selected from the group consisting of Ci-Ce alkyl, Ci-Ce substituted alkyl, C2- Ce alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, (C3-C6 cycloalkyl)(Ci-C6 alkyl), C3-C6 heterocyclyl, (C3-C6 heterocyclyl)(Ci-C6 alkyl), aryl, aryl(Ci-C6 alkyl), heteroaryl, heteroaryl(Ci-C6 alkyl), CN, C(O)NH2, C(O)NH(CI-C6 alkyl), C(O)N(CI-C3 alkyl)(Ci-C6 alkyl), C(=NOH)(CI-C6 alkyl), and C(=NOH)(CI-C6 substituted alkyl), C(O)NH(aryl), C(O)N(alkyl)(aryl), NC, S(O)2NH2, S(O)2NH(Ci-Ce alkyl) and S(O)2NH(aryl); or (ii) together with R4 form a chain of 2 to 4 carbon atoms to which are attached substituents independently selected from the group consisting of
H, Ci-Ce alkyl, aryl, and heteroaryl; or (iii) togetherwith a form a ring of 3 or4 atoms, one atom of which is selected from the group consisting of C, N, O, and S, while the remainder are carbon, which chain contains 0, 1 , or 2 double bonds, and to which chain are attached substituents independently selected from the group consisting of H, halogen, OH, Ci-Ce alkoxy, Ci-C6 alkyl, C3-C6 cycloalkyl, CHF2, CF3, OCHF2, OCF3, SCH3, SCF3, cyano, and oxo; or (iv) is selected from the group consisting of H, Ci-Ce alkyl, Ci-Ce substituted alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, (C3-C6 cycloalkyl)(Ci-C6 alkyl), C3-C6 heterocyclyl, (C3-C6 heterocyclyl)(Ci-C6 alkyl), aryl, aryl(Ci-C6 alkyl), heteroaryl, heteroaryl(Ci-Ce alkyl), CN, C(O)NH2, C(O)NH(CI-C6 alkyl), C(O)N(CI-C3 alkyl)(Ci-C6 alkyl), C(=NOH)(CI-C6 alkyl), and C(=NOH)(CI-C6 substituted alkyl), if b is halogen, CH3, CHF2, CF3, OCH3, OCHF2, OCF3, SCH3, SCHF2, SCF3, or cyano, optionally, R3 together with a form any one of CH2CH2, CH2CH2CH2, CH2CH2CH2CH2, CH=CHCH=CH, OCH2CH2, CH2OCH2, CH2CH2O, OCH=CH, CH=CHO, OCH2O, SCH2CH2, CH2SCH2, CH2CH2S, SCH=CH, CH=CHS, NHCH2CH2, CH2NHCH2, CH2CH2NH, NHCH=CH, CH=CHNH, ON=CH, CH=NO, OCH=N, N=CHO, SN=CH, CH=NS, SCH=N, N=CHS, NHN=CH, CH=NNH, NHCH=N, N=CHNH, NHN=N, N=NNH, OCH2CH2CH2, CH2OCH2CH2, CH2CH2OCH2, CH2CH2CH2O, SCH2CH2CH2, CH2SCH2CH2, CH2CH2SCH2, CH2CH2CH2S NHCH2CH2CH2, CH2NHCH2CH2, CH2CH2NCH2, CH2CH2CH2NH, N=CHCH=CH, CH=NCH=CH, CH=CHN=CH, CH=CHCH=N; and optionally, wherein one hydrogen atom or two hydrogen atoms, if present on a moiety, are replaced with substituents selected independently from the group consisting of halogen, OH, Ci-Ce alkoxy, Ci-Ce alkyl, C3-Ce cycloalkyl, CHF2, CF3, OCHF2, OCF3, SCH3, SCF3,and cyano, or wherein two hydrogens, if attached to the same carbon atom, are replaced with an oxo group;
R4 is (i) is selected from the group consisting of H, Ci-Ce alkyl, Ci-Ce substituted alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-Ce cycloalkyl, (C3-Ce cycloalkyl)(Ci-C6 alkyl), C3-Ce heterocyclyl, (C3-Ce heterocyclyl)(Ci-C6 alkyl), aryl(Ci-Ce alkyl), arylsulfonyl, heteroarylsulfonyl, aryl(Ci-Ce alkyl)sulfonyl, (Ci-C6)alkylsufonyl and heteroaryl(Ci-C6 alkyl)sulfonyl; or (ii) togetherwith R3 form a chain of 2 to 4 carbon atoms to which are attached substituents independently selected from the group consisting of H, Ci-Ce alkyl, aryl, heteroaryl, and any combination thereof;
R5 is H, D, Ci-Ce alkyl, Ci-Ce substituted alkyl;
R6: (i) is selected from the group consisting of H, Ci-Ce alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-Ce cycloalkyl, (C3-Ce cycloalkyl)(Ci-Ce alkyl), aryl(Ci-Ce alkyl), acetyl, and heteroaryl(Ci-Ce alkyl); or (ii) togetherwith R7 and the N atom to which they are attached form a 4-7 membered heterocyclyl ring; or (iii) togetherwith e and the N atom to which R6 is attached
form an azetidine or pyrrolidine ring, such ring carrying substituents independently selected from the group consisting of H, aryl, heteroaryl, Ci-Ce alkyl, and C3-C6 cycloalkyl; or (iv) together with b and the N atom to which R6 is attached form an azetidine or pyrrolidine ring, such ring carrying substituents independently selected from the group consisting of H, aryl, heteroaryl, halogen, Ci-C6 alkyl, and C3-C6 cycloalkyl;
R7: (i) is selected from the group consisting of H, Ci-Ce alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, (C3-C6 cycloalkyl)(Ci-C6 alkyl), aryl(Ci-C6 alkyl), acetyl, and heteroaryl(Ci-Ce alkyl); or (ii) together with R6 and the N atom to which they are attached form a 4-7 membered heterocyclyl ring; a: (i) is selected from a group consisting of H, halogen, CH3, CHF2, CF3, OCH3, OCHF2, OCF3, SCH3, SCHF2, SCF3, NC and cyano; or (ii) together with a form a ring of 3 or 4 atoms, one atom of which is selected from the group consisting of C, N, O, and S, while the remainder are carbon, which chain contains 0, 1 , or 2 double bonds, and to which chain are attached substituents independently selected from the group consisting of H, halogen, OH, Ci-C6 alkoxy, Ci-Ce alkyl, C3-C6 cycloalkyl, CHF2, CF3, OCHF2, OCF3, SCH3, SCHF2, SCF3, cyano, and oxo; or (iii) together with R3 form a ring of 3 or 4 atoms, one atom of which is selected from the group consisting of C, N, O, and S, while the remainder are carbon, which chain contains 0, 1 , or 2 double bonds, and to which chain are attached substituents independently selected from the group consisting of H, halogen, OH, Ci-Ce alkoxy, Ci-Ce alkyl, C3-Ce cycloalkyl, CHF2, CF3, OCHF2, OCF3, SCH3, SCHF2, SCF3, cyano, and oxo; and b, c, d, and e, are each independently H; or three of b, c, d, and e are H and the remaining substituent is a lower alkyl group; or b and e are each H, and c and d together are - CH2- or -CH2CH2-, thereby giving rise to a cyclopropane or cyclobutane ring; or b, c, and d are each H, and e, R6, and the N atom to which R6 is attached form together an azetidine or pyrrolidine ring, such ring carrying substituents independently selected from the group consisting of H, aryl, heteroaryl, Ci-Ce alkyl, and C3-Ce cycloalkyl; or c, d, and e are each H, and b, R6, and the N atom to which R6 is attached form together an azetidine or pyrrolidine ring, such ring carrying substituents independently selected from the group consisting of H, aryl, heteroaryl, halogen, Ci-Ce alkyl, and C3-Ce cycloalkyl.
2. The compound of claim 1 , wherein a is halogen.
3. The compound of claim 2, wherein a is F.
4. The compound of claim 1 , wherein a is CF3.
5. The compound of claim 1 , wherein a is H.
6. The compound of claim 1 or 5, wherein the R1 is OH and R2 is H.
7. The compound of claim 1 or 5, wherein the R1 is H and R2 is OH.
8. The compound of claim any one of claims 1 and 5-7, wherein R4 is H.
9. The compound of claim any one of claims 1 and 5-8, wherein R5 is H.
10. The compound of claim any one of claims 1 and 5-9, wherein, wherein each of b, c, d, and e is H.
13. The compound of claim 1 , wherein said compound is a compound of structural formula VI, or a pharmaceutically acceptable salt thereof, and stereoisomers thereof:
wherein:
R1 is OH, and when R1 is OH, R2 is different than OH and selected from the group consisting of H, halogen, lower alkyl, CHF2, CF3, NO, OCH3, OCHF2, OCF3, SCHF2, SCH3, SCF3, and cyano;
R2 is OH, and when R2 is OH, R1 is different than OH and selected from the group consisting of H, halogen, lower alkyl, CHF2, CF3, OCH3, OCHF2, OCF3, SCHF2, SCH3, SCF3, and cyano;
R3 is selected from the group consisting of Ci-Ce alkyl, Ci-Ce substituted alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, (C3-C6 cycloalkyl)(Ci-C6 alkyl), C3-C6 heterocyclyl, (C3- Ce heterocyclyl)(Ci-C6 alkyl), aryl, aryl(Ci-C6 alkyl), heteroaryl, heteroaryl(Ci-C6 alkyl), ON, C(O)NH2, C(O)NH(CI-C6 alkyl), C(O)N(CI-C3 alkyl)(Ci-C6 alkyl), C(=NOH)(CI-C6 alkyl), and C(=NOH)(CI-C6 substituted alkyl), C(O)NH(aryl), C(O)N(alkyl)(aryl), NO, S(O)2NH2, S(O)2NH(CI-C6 alkyl) and S(O)2NH(aryl);
R4 is selected from the group consisting of H, Ci-Ce alkyl, Ci-Ce substituted alkyl, C2- Ce alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, (C3-C6 cycloalkyl)(Ci-C6 alkyl), C3-C6 heterocyclyl, (C3-C6 heterocyclyl)(Ci-C6 alkyl), aryl(Ci-Ce alkyl), arylsulfonyl, heteroarylsulfonyl, aryl(Ci-Ce alkyl)sulfonyl, (Ci-C6)alkylsufonyl and heteroaryl(Ci-C6 alkyl)sulfonyl;
R5 is H, D, Ci-Ce alkyl, Ci-Ce substituted alkyl;
R6: is selected from the group consisting of H, Ci-Ce alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, (C3-C6 cycloalkyl)(Ci-Ce alkyl), aryl(Ci-Ce alkyl), acetyl, and heteroaryl(Ci-Ce alkyl);
R7: is selected from the group consisting of H, Ci-Ce alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, (C3-C6 cycloalkyl)(Ci-Ce alkyl), aryl(Ci-Ce alkyl), acetyl, and heteroaryl(Ci-Ce alkyl); a: is selected from a group consisting of H, halogen, CH3, CHF2, CF3, OCH3, OCHF2, OCF3, SCH3, SCHF2, SCF3, NC and cyano; and b, c, d, and e, are each independently H; or three of b, c, d, and e are H and the remaining substituent is a lower alkyl group.
14. The compound of any one of claims 1 to 13, wherein each of R6 and R7 are CH3.
16. The compound of claim 15, wherein said compound of formula IV is a compound of formula IV-
17. The compound of claim 15, wherein said compound of formula IV is a compound of formula IV-
R
IV-/?.
20. A pharmaceutical composition comprising the compound of any one of claims 1 to 19, and a pharmaceutically acceptable carrier.
21. The compound of any one of claims 1 to 19, or the pharmaceutical composition of claim 20, for use in the treatment of a disorder in a patient in need thereof.
22. A method of treating a disorder comprising administering to a patient an effective amount of the compound of any one of claims 1 to 19, or the pharmaceutical composition of claim 20.
23. Use of the compound of any one of claims 1 to 19, or the pharmaceutical composition of claim 20, for the treatment of a disorder in a patient in need thereof.
24. The compound for use of claim 21 , the method of claim 22, or the use of claim 23, wherein the disorder is one or more of major depressive disorder, drug resistant depression, and psychotic depression, addiction including alcoholism, tobacco addiction, cocaine addiction, and opioid addiction, pain indications including neuropathic pain, pain from chemotherapy associated neuropathy, phantom limb pain and fibromyalgia, inflammation (including chronic and acute), eating disorders including anorexia, autism, cluster headaches, migraines, dementia including Alzheimer’s dementia, Parkinson’s disease dementia, and Lewy body dementia, mild cognitive impairment, post-traumatic stress disorder, emotional distress associated with cancer, Fragile-X syndrome, autism spectrum disorder, bipolar disease, obsessive compulsive disease, and Rett syndrome.
25. The compound for use of claim 21 , the method of claim 22, or the use of claim 23, wherein the disorder is one or more of major depressive disorder, drug resistant depression, and psychotic depression.
26. A combination drug therapy, comprising: a compound according to any one of claims 1 to 19, or the pharmaceutical composition of claim 20; and a A/-methyl-D-aspartate (NMDA) receptor antagonist.
27. The combination drug therapy of claim 26, wherein the NMDA receptor antagonist is at least one of ketamine, nitrous oxide, memantine, amantadine, noribogaine, dextromethorphan, dextrorphan, and dextromethadone, or a pharmaceutically acceptable salt, stereoisomer, or solvate thereof.
28. The combination drug therapy of claim 27, wherein the NMDA receptor antagonist is dextromethorphan.
29. The combination drug therapy of any one of claims 26 to 28, for use in the treatment of a disorder in a patient in need thereof.
30. A method of treating a disorder comprising administering to a patient an effective amount of the combination drug therapy of any one of claims 26 to 28.
31. Use of the combination drug therapy of any one of claims 26 to 28, for the treatment of a disorder in a patient in need thereof.
32. The combination drug therapy for use of claim 29, the method of claim 30, or the use of claim 31 , wherein the disorder is one or more of major depressive disorder, drug resistant depression, and psychotic depression, addiction including alcoholism, tobacco addiction, cocaine addiction, and opioid addiction, pain indications including neuropathic pain, pain from chemotherapy associated neuropathy, phantom limb pain and fibromyalgia, inflammation (including chronic and acute), eating disorders including anorexia, autism, cluster headaches, migraines, dementia including Alzheimer’s dementia, Parkinson’s disease dementia, and Lewy body dementia, mild cognitive impairment, post-traumatic stress disorder, emotional distress associated with cancer, Fragile-X syndrome, autism spectrum disorder, bipolar disease, obsessive compulsive disease, and Rett syndrome.
33. The combination drug therapy for use of claim 29, the method of claim 30, or the use of claim 31 , wherein the disorder is one or more of major depressive disorder, drug resistant depression, and psychotic depression.
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| US202363610614P | 2023-12-15 | 2023-12-15 | |
| US63/610,614 | 2023-12-15 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2025123137A1 true WO2025123137A1 (en) | 2025-06-19 |
Family
ID=96056228
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/CA2024/051651 Pending WO2025123137A1 (en) | 2023-12-15 | 2024-12-12 | Heterocyclic compounds and methods of preparation thereof |
Country Status (1)
| Country | Link |
|---|---|
| WO (1) | WO2025123137A1 (en) |
Citations (3)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2023036473A1 (en) * | 2021-09-08 | 2023-03-16 | Cybin Irl Limited | Combination drug therapies |
| WO2023201421A1 (en) * | 2022-04-19 | 2023-10-26 | Mindset Pharma Inc. | Indoline derivatives as serotonergic agents useful for the treatment of disorders related thereto |
| WO2024044847A1 (en) * | 2022-08-29 | 2024-03-07 | Mindset Pharma Inc. | Indoline derivatives as serotonergic agents useful for the treatment of disorders related thereto |
-
2024
- 2024-12-12 WO PCT/CA2024/051651 patent/WO2025123137A1/en active Pending
Patent Citations (3)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2023036473A1 (en) * | 2021-09-08 | 2023-03-16 | Cybin Irl Limited | Combination drug therapies |
| WO2023201421A1 (en) * | 2022-04-19 | 2023-10-26 | Mindset Pharma Inc. | Indoline derivatives as serotonergic agents useful for the treatment of disorders related thereto |
| WO2024044847A1 (en) * | 2022-08-29 | 2024-03-07 | Mindset Pharma Inc. | Indoline derivatives as serotonergic agents useful for the treatment of disorders related thereto |
Non-Patent Citations (1)
| Title |
|---|
| SCHYMAN PATRIC, USHARANI DANDAMUDI, WANG YONG, SHAIK SASON: "Brain Chemistry: How Does P450 Catalyze the O-Demethylation Reaction of 5-Methoxytryptamine to Yield Serotonin?", JOURNAL OF PHYSICAL CHEMISTRY PART B, AMERICAN CHEMICAL SOCIETY, US, vol. 114, no. 20, 27 May 2010 (2010-05-27), US , pages 7078 - 7089, XP093328005, ISSN: 1520-6106, DOI: 10.1021/jp1008994 * |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| EP2424366B1 (en) | Pyrido [4, 3-b]indoles and methods of use | |
| EP2274307B1 (en) | Bridged heterocyclic compounds and methods of use | |
| Schweitzer et al. | Suppression of arrhythmia by enhancing mitochondrial Ca2+ uptake in catecholaminergic ventricular tachycardia models | |
| Ahnaou et al. | Blocking melanin-concentrating hormone MCH1 receptor affects rat sleep–wake architecture | |
| BRPI0718247B1 (en) | COMPOUND USED AS AN INHIBITOR OF THE VESICULAR MONOAMINE CONVEYOR 2, PHARMACEUTICAL COMPOSITION INCLUDING SUCH COMPOUND AND USE OF SUCH PHARMACEUTICAL COMPOSITION IN THE TREATMENT OF HYPERKINETIC DISORDERS | |
| Beitzel et al. | β2-Adrenoceptor agonist fenoterol enhances functional repair of regenerating rat skeletal muscle after injury | |
| JP2022521789A (en) | Azepino-indole and other heterocyclic compounds for the treatment of brain disorders | |
| WO2011038164A1 (en) | Bridged heterocyclic compounds and methods of use | |
| CA3007673A1 (en) | Metabolism resistant fenfluramine analogs and methods of using the same | |
| EP3760631B1 (en) | Crystal salt of pyrazolo[1,5-a]pyrimidine compound | |
| WO2023212811A1 (en) | Azepinoindoles and methods of preparation thereof | |
| US10538482B2 (en) | Adamantane and memantine derivatives as peripheral NMDA receptor antagonists | |
| WO2025123137A1 (en) | Heterocyclic compounds and methods of preparation thereof | |
| EP3829579B1 (en) | 1,3,8-triazaspiro compounds and their use as medicaments for the treatment of reperfusion injury | |
| KR20240133739A (en) | Phenethylamine and its preparation method | |
| KR20220158290A (en) | Compositions for the treatment of neurodegenerative and mitochondrial diseases and methods of using the same | |
| AU2018243482A1 (en) | Substituted (4'-hydroxyphenyl)cycloalkane and (4'-hydroxyphenyl)cycloalkene compounds and uses thereof as selective agonists of the estrogen receptor beta isoform for enhanced memory consolidation | |
| CN105636943B (en) | Chromene Derivatives as Inhibitors of TCR-Nck Interaction | |
| Cortés et al. | A new isoxazolic compound acts as α7 nicotinic receptor agonist in human umbilical vein endothelial cells | |
| Luis et al. | Synthesis and preliminary ex vivo evaluation of the spasmolytic activity of 1, 3-thiazolium-and 1, 3, 4-thiadiazolium-5-methylthio-and 5-tioacetate derivatives | |
| US20200316087A1 (en) | Novel combination therapy for anxiety disorders, epilepsy, and pain | |
| EP4375286A1 (en) | Oxaspiro derivative, and preparation method therefor and use thereof | |
| CN118891248A (en) | Phenethylamine and preparation method thereof | |
| TW202442225A (en) | An indole compound and its application | |
| EP3328826B1 (en) | Novel adamantane and memantine derivatives as peripheral nmda receptor antagonists |
Legal Events
| Date | Code | Title | Description |
|---|---|---|---|
| 121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 24901861 Country of ref document: EP Kind code of ref document: A1 |