WO2022187973A1 - Dosing regimens of pharmaceutical and nutraceutical mushroom and cannabis compositions and their use to treat cns disorders and improve mental health - Google Patents

Dosing regimens of pharmaceutical and nutraceutical mushroom and cannabis compositions and their use to treat cns disorders and improve mental health Download PDF

Info

Publication number
WO2022187973A1
WO2022187973A1 PCT/CA2022/050374 CA2022050374W WO2022187973A1 WO 2022187973 A1 WO2022187973 A1 WO 2022187973A1 CA 2022050374 W CA2022050374 W CA 2022050374W WO 2022187973 A1 WO2022187973 A1 WO 2022187973A1
Authority
WO
WIPO (PCT)
Prior art keywords
composition
days
cannabis
unit dosage
dosage form
Prior art date
Application number
PCT/CA2022/050374
Other languages
French (fr)
Inventor
Radka Milanova
James Smeeding
Calum HUGHES
Original Assignee
Allied Corp.
Priority date (The priority date 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 date listed.)
Filing date
Publication date
Application filed by Allied Corp. filed Critical Allied Corp.
Publication of WO2022187973A1 publication Critical patent/WO2022187973A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/045Hydroxy compounds, e.g. alcohols; Salts thereof, e.g. alcoholates
    • A61K31/05Phenols
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/34Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide
    • A61K31/343Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide condensed with a carbocyclic ring, e.g. coumaran, bufuralol, befunolol, clobenfurol, amiodarone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/35Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom
    • A61K31/352Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom condensed with carbocyclic rings, e.g. methantheline 
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic 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
    • A61K31/403Heterocyclic 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
    • A61K31/404Indoles, e.g. pindolol
    • A61K31/4045Indole-alkylamines; Amides thereof, e.g. serotonin, melatonin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/455Nicotinic acids, e.g. niacin; Derivatives thereof, e.g. esters, amides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/675Phosphorus compounds having nitrogen as a ring hetero atom, e.g. pyridoxal phosphate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7135Compounds containing heavy metals
    • A61K31/714Cobalamins, e.g. cyanocobalamin, i.e. vitamin B12
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/06Fungi, e.g. yeasts
    • A61K36/062Ascomycota
    • A61K36/066Clavicipitaceae
    • A61K36/068Cordyceps
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/06Fungi, e.g. yeasts
    • A61K36/07Basidiomycota, e.g. Cryptococcus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/18Magnoliophyta (angiosperms)
    • A61K36/185Magnoliopsida (dicotyledons)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca

Definitions

  • Described herein are pharmaceutical and nutraceutical compositions from mushrooms and Cannabis which are useful to treat CNS and mental health disorders and to improve mental health, for example when administered in the disclosed distinct daily dosing regimens.
  • BACKGROUND OF THE INVENTION [03]
  • Mental health conditions are the leading cause of disability worldwide. They lead to over a million lives lost to suicide, cost the global economy $1 trillion annually in lost productivity, and affect 1 in 5 adults. Their incidence has been increasing over past decades, and accelerating over the last few years. Despite the recognition that such conditions, and CNS disorders more broadly, are a great and growing problem, only half of those afflicted receive treatment, largely due to a dearth of accessible and effective treatment options.
  • formulations are useful for modulating neurotransmission, endocannabinoid system activity, and neurosteroid biosynthesis, for treating mental health conditions and other CNS disorders, and for improving mental health, in particular when used in a variety of distinct and uniquely designed daily and combination dosing regimens, as taught in different embodiments herein.
  • the medical benefits of Cannabis have been recognized for at least 4,750 years of recorded history. Across these millennia however, the hundreds of chemical compounds in the plant have resisted study in a systematic and comprehensive way.
  • the present invention discloses formulations from mushrooms and Cannabis , including with synergistic and entourage enhanced combinations of tryptamines, cannabinoids, and other active agents, and which are useful in methods for treating CNS and mental health disorders and for improving mental health, for example when administered in the daily dosing regimens. Such compositions and methods have various improvements and advantages over current treatments, as will be readily appreciated from the disclosure herein. [11] In some aspects are disclosed methods of treating a CNS disorder patient in need thereof, comprising administering to the patient a mushroom composition and a second composition according to a defined dosing regimen.
  • a defined dosing regimen is a daily dosing regimen consisting of a first number of days of administration of the mushroom composition, followed by a second number of days of administration of the second composition.
  • the first number of days is between 2 and 7 days.
  • the first number of days is between 2 and 5 days.
  • the first number of days is either 3 or 4 days.
  • the first number of days is 4 days.
  • the second number of days is between 2 and 7 days.
  • the second number of days is between 2 and 5 days.
  • the second number of days is either 3 or 4 days.
  • the second number of days is 3 days.
  • a patient is administered a psychedelic mushroom composition.
  • the psychedelic mushroom composition comprises a psychedelic mushroom extract or a compound from a psychedelic mushroom.
  • the psychedelic mushroom extract is a Psilocybe extract.
  • the Psilocybe extract is an extract of Psilocybe cubensis .
  • the psychedelic mushroom extract is an extract of a species within a fungal genera selected from the group consisting of Conocybe, Copelandia, Galerina, Gymnopilus, Inocybe, Mycena, Panaeolina, Panaeolus, Pholiotina , and Pluteus .
  • the compound from a psychedelic mushroom is any of psilocybin, psilocin, norbaeocystin, baeocystin, aeruginascin, and norpsilocin.
  • the compound from a psychedelic mushroom is produced by chemical synthesis or biosynthesis.
  • the functional mushroom comprises a Hericium extract or a compound from Hericium .
  • the compound from Hericium is any of any of Hericenone A, Hericenone B, Hericenone C, Hericenone D, Hericenone E, Hericenone F, Hericenone G, Hericenone H, Hericenone I, Hericenone J, Hericenone K, 3HF, DLPE, Isohericerinol A, Hericerin, NDPIH, and Corallocin A.
  • the functional mushroom comprises a Cordyceps extract or a compound from Cordyceps .
  • the compound from Cordyceps is any of cordycepin, cordycepic acid, N-acetylgalactosamine, adenosine, ergosterol, an ergosteryl ester, a bioxanthracene, hypoxanthine, a macrolide, a cicadapeptin, myriocin, superoxide dismutase, naphthoquinone, cordyheptapeptide, dipicolinic acid, a fibrinolytic enzyme, and cordymin.
  • a patient is administered a cannabis composition.
  • the cannabis composition is an intoxicating cannabis composition.
  • the intoxicating cannabis composition comprises THC.
  • the THC is ⁇ 9 -THC. In embodiments, the THC is ⁇ 8 -THC .
  • the cannabis composition is a non-intoxicating cannabis composition. In embodiments, the non-intoxicating cannabis composition comprises CBD, and is free of THC. [14] In embodiments, a patient is administered a washout composition.
  • the washout composition comprises an active compound such as amino acids, antioxidants, anti-inflammatory agents, analgesics, antineuropathic and antinociceptive agents, antimigraine agents, anxiolytics, antidepressants, antipsychotics, anti-PTSD agents, immunostimulants, anti-cancer agents, antiemetics, orexigenics, antiulcer agents, antihistamines, antihypertensives, anticonvulsants, antiepileptics, bronchodilators, neuroprotectants, entactogens and empathogens, entheogens, psychedelics, nootropics, monoamine oxidase inhibitors, sedatives, stimulants, supplements, and vitamins.
  • an active compound such as amino acids, antioxidants, anti-inflammatory agents, analgesics, antineuropathic and antinociceptive agents, antimigraine agents, anxiolytics, antidepressants, antipsychotics, anti-PTSD agents, immunostimulants, anti-
  • a patient abstains from any administration of a composition of the invention.
  • the days of abstinence are after the first number of days of administration of the mushroom composition, and before the second number of days of administration of the second composition.
  • the days of abstinence are after the second number of days of administration of the second composition, and before a next administration of the mushroom composition.
  • the days of abstinence are within the first number of days of administration of the mushroom composition.
  • the days of abstinence are within the second number of days of administration of the second composition.
  • the number of days of abstinence is between 2 and 7 days. In embodiments, the number of days of abstinence is between 2 and 5 days.
  • the number of days of abstinence is either 3 or 4 days.
  • a defined dosing regimen is repeated between 2 and 10 times to provide a therapeutic regime.
  • the defined dosing regimen is repeated six times, seven times, or eight times.
  • the daily dosing regimen is repeated between 2 and 10 times to provide a therapeutic regime.
  • the daily dosing regimen is repeated six times, seven times, or eight times.
  • the daily dosing regimen lasts for a period of six weeks, seven weeks, or eight weeks.
  • the regime lasts from between 15 and 90 days.
  • the regime lasts from between 30 and 70 days.
  • the regime lasts from between 50 and 65 days.
  • the regime lasts for 60 days.
  • a therapeutic regime comprises at least two different defined dosing regimens, or three or more different daily dosing regimens of the invention.
  • mushroom compositions for use in the methods of the invention, comprising synergistically and therapeutically effective amounts of: a Hericium extract or a compound from Hericium, and a Cordyceps extract or a compound from Cordyceps together with at least one carrier, diluent, or excipient, and wherein the at least one carrier, diluent, or excipient is not from a fungal source or found in fungi.
  • a mushroom composition further comprises a Psilocybe extract or a compound from Psilocybe.
  • a mushroom composition further comprises one or more of niacin, valerian root extract, vitamin B12, and a CB 1 receptor modulator. In embodiments, a mushroom composition further comprises two or more of niacin, valerian root extract, vitamin B12, and a CB 1 receptor modulator. In embodiments, a mushroom composition further comprises three or more of niacin, valerian root extract, vitamin B12, and a CB 1 receptor modulator. In embodiments, a mushroom composition further comprises niacin, valerian root extract, vitamin B12, and a CB 1 receptor modulator. In embodiments, the CB 1 receptor modulator is THC. In embodiments, the CB 1 receptor modulator is CBD.
  • a mushroom composition further comprises one or more of rice hull concentrate powder, rice bran extract powder, and rice bran 100 mesh powder. In embodiments, a mushroom composition further comprises a therapeutically effective amount of an additional active compound.
  • cannabis compositions for use in the methods of the invention, comprising synergistically and therapeutically effective amounts of: cannabidiol (CBD); an entourage-enhancing cannabinoid fraction; and an entourage-enhancing isoprenoid fraction; together with at least one carrier, diluent, or excipient, and wherein the at least one carrier, diluent, or excipient is not from a plant source or found in plants.
  • CBD cannabidiol
  • entourage-enhancing cannabinoid fraction an entourage-enhancing isoprenoid fraction
  • a cannabis composition further comprises THC.
  • the THC is ⁇ 9 -THC.
  • the THC is ⁇ 8 -THC.
  • the CBD is natural CBD.
  • the CBD is synthetic CBD.
  • the ratio of THC:CBD is from about 1:20 to about 20:1.
  • the ratio of THCCBD is between 1:20 to 1:10, between 1:10 and 1:5, between 1:5 and 1:2, between 1:2 and about 1:1, between about 1:1 and 2:1, between 2:1 and 5:1, between 5:1 and 10:1, and between 10:1 and 20:1.
  • the ratio of THCCBD is about 1:4.
  • the entourage-enhancing cannabinoid fraction comprises two or more cannabinoids.
  • the cannabinoids in the entourage-enhancing cannabinoid fraction are natural.
  • at least one of the natural cannabinoids is derived from a botanical source.
  • the botanical source is a Cannabis plant.
  • at least one of the cannabinoids in the entourage-enhancing cannabinoid fraction is synthetic.
  • all of the cannabinoids in the entourage-enhancing cannabinoid fraction are synthetic.
  • at least one of the cannabinoids in the entourage-enhancing cannabinoid fraction is selected for its ability to provide therapeutic effects or synergistic effects.
  • At least one of the cannabinoids in the entourage-enhancing cannabinoid fraction is selected for its ability to modulate the endocannabinoid system, modulate neurosteroid biosynthesis, or modulate a neurotransmitter system.
  • the entourage-enhancing cannabinoid fraction comprises at least two or more of: cannabichromene (CBC), cannabielsoin (CBE), cannabigerol (CBG), cannabicyclol (CBL), cannabinol (CBN), cannabidinodiol (CBND), cannabicitran (CBT), cannabitriol (CBT), cannabivarin (CBV), cannabigerol monomethyl ether (CBGM), cannabidivarin (CBDV), tetrahydrocannabivarin (THCV), cannabidiphorol (CBDP), tetrahydrocannabiphorol (THCP), and iso-tetrahydrocannabinol (iso-THC).
  • CBC cannabichromene
  • CBE cannabielsoin
  • CBG cannabigerol
  • CBL cannabinol
  • CBN cannabinol
  • the entourage-enhancing cannabinoid fraction comprises two or more cannabinoids independently selected from any of CBC-type cannabinoids, CBD-type cannabinoids, CBE-type cannabinoids, CBG-type cannabinoids, CBL-type cannabinoids, CBN-type cannabinoids, CBND-type cannabinoids, CBT-type cannabinoids, ⁇ 8 -THC -type cannabinoids, ⁇ 9 -THC-type cannabinoids, and miscellaneous-type cannabinoids, as specifically disclosed below.
  • the entourage-enhancing isoprenoid fraction comprises two or more isoprenoids.
  • all of the isoprenoids in the entourage-enhancing isoprenoid fraction are natural.
  • at least one of the natural isoprenoids is derived from a botanical source.
  • the botanical source is a Cannabis plant.
  • at least one of the isoprenoids in the entourage-enhancing isoprenoid fraction is synthetic.
  • all of the isoprenoids in the entourage-enhancing isoprenoid fraction are synthetic.
  • at least one of the isoprenoids in the entourage-enhancing isoprenoid fraction is selected for its ability to provide therapeutic effects or synergistic effects.
  • At least one of the isoprenoids in the entourage-enhancing isoprenoid fraction is selected for its ability to modulate the endocannabinoid system, modulate neurosteroid biosynthesis, or modulate a neurotransmitter system.
  • two or more isoprenoids are selected from the group consisting of: alpha-bisabolol, beta-caryophyllene, camphene, carene, caryophyllene oxide, alpha-humulene, fenchol, guaiene, guaiol, limonene, linalool, myrcene, nerolidol, ocimene, alpha-phellandrene, alpha-pinene, beta-pinene, alpha-terpinene, gamma-terpinene, terpineol, and terpinolene.
  • the entourage-enhancing isoprenoid fraction is myrcene dominant.
  • the myrcene dominant entourage-enhancing isoprenoid fraction contains at least 20% myrcene by weight.
  • the entourage enhancing isoprenoid fraction includes two or more isoprenoids independently selected from any of monoterpenes, sesquiterpenes, diterpenes, triterpenes, and miscellaneous terpenes, as disclosed below. [21]
  • the entourage-enhancing cannabinoid fraction or the entourage-enhancing isoprenoid fraction provides a therapeutic effect, decreases an unwanted effect, increases stability or shelf-life, or alters pharmacokinetics or pharmacodynamics.
  • the therapeutic effect is an antioxidant, anti-inflammatory, analgesic, antineuropathic, antinociceptive, antimigraine, anxiolytic, antidepressant, antipsychotic, anti-PTSD, immunostimulant, anti-cancer, antiemetic, orexigenic, antiulcer, antihistamine, antihypertensive, anticonvulsant, antiepileptic, bronchodilator, neuroprotective, entactogenic, empathogenic, entheogenic, psychedelic, sedative, or stimulant effect.
  • the entourage-enhancing cannabinoid fraction or the entourage-enhancing isoprenoid fraction provides a synergistic effect.
  • the synergistic effect is a greater than additive increase in potency, bioactivity, bioaccessibility, bioavailability, or therapeutic effect.
  • the entourage-enhancing cannabinoid fraction or the entourage-enhancing isoprenoid fraction modulates the endocannabinoid system, modulates neurosteroid biosynthesis, or modulates a neurotransmitter system.
  • the entourage-enhancing cannabinoid fraction comprises from 0.1% to about 25% of the composition by weight.
  • the entourage-enhancing cannabinoid fraction comprises about 10% of the composition by weight.
  • the entourage-enhancing isoprenoid fraction comprises from 0.1% to about 10% of the composition by weight.
  • the entourage-enhancing isoprenoid fraction comprises about 5% of the composition by weight.
  • the cannabis composition further comprises one or more flavonoids, as specifically disclosed below.
  • a cannabis composition of is formulated for any one of: (1) oral administration, and comprising a hydrophobic solvent or an alcohol-based solvent, and optionally a co-solvent; (2) use with a transdermal drug delivery system, wherein the composition is combined with a permeation enhancing agent, and optionally with one or more stabilizers, solubilizers, or adhesives; or (3) administration as a nanostructured formulation selected from the group consisting of: a nanoemulsion, a nanocapsule, a nanoparticle conjugate, or a nano-encapsulated oral or nasal spray.
  • a composition is prepared for administration as a formulation selected from the group consisting of: a soft or hard gelatin capsule, an oral tablet, a timed release capsule or tablet, a controlled release capsule or tablet, an extended release capsule or tablet, gastrorententive tablet, a sublingual or buccal tablet, an oral spray, a nasal spray, a suppository, a suspension, a solution suitable for subcutaneous, intravenous, intraperitoneal, or intramuscular administration, a liquid suitable for use in a liquid vaporization appliance, a topical preparation, or a transdermal patch.
  • a cannabis composition further comprises a therapeutically effective amount of an additional active compound.
  • the additional active compound acts to increase therapeutic efficacy, provide additional therapeutic effects, decrease unwanted effects, increase stability or shelf-life, improve bioavailability, induce synergy, or alter pharmacokinetics or pharmacodynamics.
  • the additional active compound is selected from the group consisting of: amino acids, antioxidants, anti-inflammatory agents, analgesics, antineuropathic and antinociceptive agents, antimigraine agents, anxiolytics, antidepressants, antipsychotics, anti-PTSD agents, immunostimulants, anti-cancer agents, antiemetics, orexigenics, antiulcer agents, antihistamines, antihypertensives, anticonvulsants, antiepileptics, bronchodilators, neuroprotectants, entactogens and empathogens, entheogens, psychedelics, nootropics, monoamine oxidase inhibitors, sedatives, stimulants, supplements, and vitamins.
  • unit dosage forms useful to treat a mammal with a CNS disorder, and which comprise a therapeutically effective amount of: a functional mushroom composition; a psychedelic mushroom composition; a non-intoxicating cannabis composition; and an intoxicating cannabis composition, as well as a washout composition.
  • the unit dosage form is suitable for oral, mucosal, rectal, injectable, intranasal, inhaled, or transdermal administration.
  • the unit dosage form is an oral solid form or an oral liquid form.
  • the unit dosage form comprises ground or pulverized Psilocybe fungal matter in a total amount of between 1 mg and 1 g.
  • the unit dosage form comprises a Psilocybe extract in a total amount of between 0.5 mg and 500 mg. In embodiments, the unit dosage form comprises psilocybin in a total amount of between 0.01 mg and 10 mg. In embodiments, the unit dosage form comprises psilocybin in a total amount of between 0.5 mg and 3 mg. In embodiments, the unit dosage form comprises psilocybin in a total amount of 2.5 mg. In embodiments, the unit dosage form comprises psilocybin in a total amount of 1.25 mg. In embodiments, the unit dosage form comprises THC in a total amount of between 0.1 mg and 100 mg. In embodiments, the unit dosage form comprises THC in a total amount of between 2 mg and 50 mg.
  • the unit dosage form comprises THC in a total amount of 10 mg. In embodiments, the unit dosage form comprises THC in a total amount of 5 mg.
  • the defined dosing regimen is a daily dosing regimen consisting of a first number of days of administration of one unit dosage form, followed by a second number of days of administration of a second unit dosage form. In embodiments, the first number of days is between 2 and 7 days. In embodiments, the first number of days is between 2 and 5 days.
  • the first number of days is either 3 or 4 days. In embodiments, the first number of days is 4 days.
  • the second number of days is between 2 and 7 days. In embodiments, the second number of days is between 2 and 5 days. In embodiments, the second number of days is either 3 or 4 days. In embodiments, the second number of days is 3 days.
  • the defined dosing regimen is repeated between 2 and 10 times to provide a therapeutic regime. In embodiments, the defined dosing regimen is repeated six times, seven times, or eight times. In embodiments, the daily dosing regimen is repeated between 2 and 10 times to provide a therapeutic regime. In embodiments, the daily dosing regimen is repeated six times, seven times, or eight times.
  • the daily dosing regimen lasts for a period of six weeks, seven weeks, or eight weeks. In embodiments, the regime lasts from between 15 and 90 days. In embodiments, the regime lasts from between 30 and 70 days. In embodiments, the regime lasts from between 50 and 65 days. In embodiments, the regime lasts for 60 days. [27] In some aspects are disclosed daily dosing kits according to the methods of the invention, comprising the unit dosage forms to be administered according to said methods, and in such amount as the number of total days of their administration. In embodiments, a daily dosing kit is provided as a blister pack, a blister tray, a blister card, a strip pack, a push-through pack, a compliance pack, or a calendar pack.
  • the neurotransmission is catecholaminergic, gabaminergic, glutamatergic, or serotonergic neurotransmission.
  • the serotonergic neurotransmission is a modulation of 5-HT 2A receptors, without a significant modulation of 5-HT 2B receptors.
  • the serotonergic neurotransmission is a modulation of 5-HT 1A and 5-HT 2A receptors.
  • endocannabinoid system activity in a mammal in need of such modulation comprising administering to the mammal a first unit dosage form together with a second unit dosage form, according to a defined dosing regimen.
  • methods for modulating biosynthesis of a neuroactive steroid in a mammal in need of such modulation comprising administering to the mammal a first unit dosage form together with a second unit dosage form, according to a defined dosing regimen.
  • the medical condition is a disorder linked to dysregulation or inadequate functioning of neurotransmission.
  • the disorder linked to dysregulation or inadequate functioning of neurotransmission is that of catecholaminergic, gabaminergic, glutamatergic, or serotonergic neurotransmission.
  • the disorder linked to dysregulation or inadequate functioning of neurotransmission is that of serotonergic neurotransmission.
  • the medical condition is a disorder linked to dysregulation or inadequate functioning of the endocannabinoid system.
  • the medical condition is a disorder linked to dysregulation or inadequate functioning neurosteroid biosynthesis.
  • the medical condition treated by the invention is a mental health condition.
  • the mental health condition is selected from the group consisting of: post-traumatic stress disorder (PTSD), adjustment disorder, affective disorder, depression, dysthymia, anxiety, phobia disorders, binge disorders, body dysmorphic disorder, alcohol or drug abuse or dependence disorders, disruptive behavior disorders, eating disorders, impulse control disorders, obsessive compulsive disorder (OCD), attention deficit hyperactivity disorder (ADHD), personality disorders, attachment disorders, and dissociative disorders.
  • the mental health condition is a disorder related to rigid modes of thinking.
  • the disorder related to rigid modes of thinking is an anxiety disorder, a depressive disorder, addiction, an eating disorder, OCD, or PTSD.
  • a mental health condition in a human, the method comprising identifying a human in need of said reducing, and administering to the human a first unit dosage form of any of claims 134-149 together with a second unit dosage form of any of claims 134-149, according to a defined dosing regimen.
  • the mental health condition is PTSD.
  • the symptoms of PTSD that are reduced include one or more of flashbacks, nightmares, distressing and intense memories, distress or physical reactions after being exposed to triggers, blaming self or others for the trauma, decreased interest in things that were once enjoyable, negative feelings about self and the world, inability to remember the trauma clearly, difficulty feeling positive, feelings of isolation, negative affect, difficulty feeling positive, avoidance, aggression or irritability, hypervigilance and hyper-awareness, difficulty concentrating, difficulty sleeping, heightened startle response, engaging in self-destructive, or risky behavior, difficulty sleeping or staying asleep, or suicidal ideation.
  • a human in need of said improving comprising identifying a human in need of said improving, and administering to the human a first unit dosage form together with a second unit dosage form, according to a defined dosing regimen, and wherein the improvement in psychological functioning is a reduction of neuroticism or psychological defensiveness, an increase in creativity or openness to experience, an increase in decision-making ability, an increase in feelings of wellness or satisfaction, or an increase in ability to fall or stay asleep.
  • a CNS disorder in a human patient comprising administering to the patient a first unit dosage form together with a second unit dosage form, according to a defined dosing regimen, and in combination with one or more psychotherapy sessions, or in conjunction with participation in a therapeutically beneficial self-care activity which is chosen from the group consisting of: breathing exercises, meditation and concentration practices, focusing on an object or mantra, listening to music, physical exercise, stretching or bodywork, journaling, grounding techniques, positive self-talk, or engaging with a pet or animal.
  • a therapeutically beneficial self-care activity which is chosen from the group consisting of: breathing exercises, meditation and concentration practices, focusing on an object or mantra, listening to music, physical exercise, stretching or bodywork, journaling, grounding techniques, positive self-talk, or engaging with a pet or animal.
  • FIG.1 illustrates a number of exemplary daily dosing kits containing a 21-day supply of the mushroom and cannabis compositions of the invention, according to embodiments.
  • FIG.2 illustrates a number of exemplary daily dosing kits containing a 21-day supply of the mushroom and cannabis compositions of the invention, according to embodiments, and additionally includes abstinent days, wherein no dosing is completed.
  • FIG.3 illustrates a number of exemplary daily dosing kits containing a 21-day supply of certain mushroom and cannabis compositions of the invention according to certain embodiments, wherein a “M+” mushroom composition of EXAMPLE 2 or EXAMPLE 3 is administered for four consecutive days, followed by any combination of administration of any of: (1) a mushroom composition that does not contain psilocybin (“M–”); (2) a cannabis composition containing THC (“C+”); a cannabis composition that does not contain THC (“C–”); and abstinence or “washout” (“A”), for the next three consecutive days.
  • DETAILED DESCRIPTION [39] Among the various aspects of the present invention are formulations of compounds from mushrooms and Cannabis , including with synergistic and entourage enhanced combinations of tryptamines, cannabinoids, and other active agents, and which are useful in methods for treating CNS disorders and mental health conditions and for improving mental health, for example when administered in daily dosing regimens and therapeutic regimes.
  • the term “including” as used herein means, and is used interchangeably with, the phrase “including but not limited to.”
  • the term “or” is used herein to mean, and is used interchangeably with, the term “and/or,” unless context clearly indicates otherwise.
  • all numbers expressing quantities of ingredients, properties such as concentration, reaction conditions, and so forth, used to describe and claim certain embodiments of the invention are to be understood as being modified in some instances by the term “about.” Accordingly, in some embodiments, the numerical parameters set forth in the description and claims are approximations that can vary depending upon the desired properties sought to be obtained by a particular embodiment. In embodiments, “about” refers to plus or minus five percent ( ⁇ 5%) of the recited unit of measure.
  • Botanical Drug Substance may refer to the term as defined through FDA implementing rules and regulations, and as described in the Guidance for Industry Botanical Drug Products, December 2016 (Docket No.
  • Botanical Drug Guidance Terms including “Botanical Drug Product” and “Botanical Raw Materials” shall be known similarly. [50] For avoidance of doubt, even though “botanical” is commonly used to mean “relating to plants,” for purposes herein “botanical” as in a “botanical” drug substance or drug product will be understood to include substances and products (e.g., extracts) from fungal material as well, such as psilocybin-containing or other mushrooms, for example as defined by FDA.
  • a substance or product is from fungal material, it may be from any part of the fruiting body (commonly referred to as a “mushroom”), regardless of whether fruiting occurs above or below ground (as with “truffles”), or from mycelia or other material.
  • Mushroom will be used throughout this description to mean and to be the equivalent of terms like “fungi” or “fungal,” to the extent those terms would be broader.
  • a “mushroom composition” thus should not be interpreted as being narrower than a “fungi composition” or a “fungal composition.”
  • a “mushroom composition” may be either an “M+” composition or an “M-” composition.
  • An “M+” composition which also may be referred to as a “psychedelic mushroom composition” will contain at least one compound known to produce or capable of producing “psychedelic effects” when ingested in humans, or that demonstrates such capabilities (e.g., by showing a head-twitch response or “HTR” in rodents).
  • a “psychedelic mushroom composition” will contain at least one compound known to produce or capable of producing “psychedelic effects” when ingested in humans, or that demonstrates such capabilities (e.g., by showing a head-twitch response or “HTR” in rodents).
  • an “M+” composition may be a “psychedelic mushroom composition” even if it does not or cannot produce psychedelic effects when ingested or administered at the dosages of or according to the methods of the invention, and indeed, the lack of psychedelic effects in embodiments will be preferred, and it is therefore referred to as such because it comprises certain psychedelic compounds, not because those compounds produce psychedelic effects.
  • a mushroom composition comprising psilocybin, psilocin, and other psychedelic tryptamines (referred to together as “compounds from a psychedelic mushroom”) will be an “M+” or “psychedelic” mushroom composition, as will a composition comprising an extract from a Psilocybe or other psilocybin-containing mushroom.
  • a “M-” composition by contrast, will be a mushroom composition that does not contain any compound known to produce or capable of producing psychedelic effects when ingested in humans, or that demonstrates such capabilities.
  • An “M-” composition may also be referred to as a “functional mushroom composition.”
  • a functional mushroom composition may comprise extracts from lion’s mane and/or cordyceps, but not Psilocybe mushrooms.
  • a functional mushroom composition comprises merely trace amounts of a psilocybin or a psychedelic mushroom (e.g., less than 1% w/w, less than 0.5% w/w, less than 0.1% w/w, less than 0.05% w/w, less than 0.01% w/w, or less than 0.005% w/w of the mushroom composition), which may occur, for example, because of shared manufacturing equipment, it will nonetheless be considered an “M-” or “functional” mushroom composition, and may further be considered a mushroom composition “substantially without psychoactive compounds” or “substantially free of’ psychoactive compounds, or “substantially” without or free of one or more specific psychedelic compounds, such as psilocybin or psilocin.
  • a “cannabis composition” may be either a “C+” composition or a “C-” composition.
  • An “C+” composition which also may be referred to as an “intoxicating cannabis composition” or a “cannabis composition with THC” will contain at least one cannabinoid known to produce or capable of producing “intoxicating effects” when ingested in humans, or that demonstrates such capabilities.
  • cannabis compositions not comprising a cannabinoid known to produce or capable of producing “intoxicating effects” when ingested in humans, or that demonstrates such capabilities will be referred to as a “C-” composition or a “non-intoxicating cannabis composition” or a “cannabis composition without THC.”
  • the “intoxicating” compound will be a tetrahydrocannabinol (e.g., ⁇ 9 -THC. ⁇ 8 -THC , or another intoxicating THC compound).
  • C+ compositions are referred to as “intoxicating” instead of “psychoactive” because some compounds such as CBD that can be considered psychoactive, nevertheless do not produce the “high” associated with Cannabis.
  • a “C+” composition may be an “intoxicating cannabis composition” even if it does not or cannot produce intoxicating effects or produce a “high” when ingested or administered at the dosages of or according to the methods of the invention, or does not for a specific individual.
  • the inclusion of THC may be to balance the entourage effects of other cannabinoids or other compounds in the cannabis composition.
  • the inclusion of THC may be for purposes of causing at least some degree of intoxication or high, such as the inclusion of about 5 mg of THC or more, or an equivalent amount of another intoxicating cannabinoid, based on relative potency.
  • an intoxicating amount may be less than or more than 5 mg, but the amount that may cause intoxication will be readily appreciated and readily determinable both by those of ordinary skill as well as by subjects seeking to either obtain or avoid such effects in the self-administration of the compositions of the invention, or by the practice of the methods of the invention, such as when intoxication is desired, or when it is sought to be avoided, for example when it might interfere with one or more daily activities during the course of a daily dosing regimen.
  • a subject will administer a daily dosing regimen specifically selected or timed so that days of administration of intoxicating cannabis compositions, where such compositions produce intoxicating effects, will be on specific days, such as on weekend days.
  • an intoxicating cannabis compositions comprises merely trace amounts of THC or an intoxicating cannabinoid (e.g., less than 1% w/w, less than 0.5% w/w, less than 0.1% w/w, less than 0.05% w/w, less than 0.01% w/w, or less than 0.005% w/w of the cannabis composition), which may occur, for example, because of shared manufacturing equipment, it will nonetheless be considered an “C-” or “non-intoxicating” cannabis composition, and may further be considered a cannabis composition “substantially without THC” or “substantially free of’ THC.
  • Botanical drug substances and products may be available by prescription or over-the-counter (“OTC”), or as nutritional or dietary supplements, or under any other regulatory regime; they also may be unregulated (e.g., “natural products”).
  • OTC over-the-counter
  • Cannabis ” or “Cannabis plant” may refer to wild-type C. sativa , C. indica , and C. ruderalis , as well as genetic crosses, self-crosses, hybrids, variants, and chemovars thereof.
  • “Cannabis-derived drug substance” may refer to a botanical drug substance which is derived from cannabis plants (including plant parts, plant part biomass, and plant exudates), as non-limiting, purely illustrative examples, primary extracts prepared by processes including maceration, percolation, extraction with solvents such as C1–C5 alcohols (e.g., ethanol), hydrocarbons (e.g., propane, butane), and subcritical or supercritical carbon dioxide.
  • “Cannabis-derived drug product” may refer to a primary cannabis extract that is further purified, for example by distillation or chromatography. It will be known to those of skill that when certain solvents are used to prepare primary extracts, the resultant extract may contain non-specific lipid-soluble material.
  • CB 1 receptor modulator may refer to a compound which is a ligand of and thus binds to and modulates the cannabinoid type 1 (CB 1 ) receptor.
  • CB 1 receptor modulators include positive and negative allosteric modulators which bind to the receptor and change the conformation of its binding site, without also acting as an orthosteric agonist or antagonist. Allosteric CB 1 receptor modulators may treat CNS disorders without some adverse effects caused by orthosteric binding.
  • Negative allosteric modulators of CB 1 include cannabidiol (CBD) and N -palmitoylethanolamine (PEA). Additional allosteric modulators of CB 1 , as well as orthosteric modulators, will be known to those of skill ( see, e.g., Laprairie et al., 2015).
  • Extract may refer to a botanical extract (e.g., a psilocybin-containing mushroom extract, a lion’s mane extract, a cordyceps extract, or a valerian root extract) prepared from a botanical (including fungal) source that does not undergo isolation, purification, or other processing to obtain specific active compounds separate from other constituents (i.e., is obtained as a whole-plant medicine).
  • a botanical extract e.g., a psilocybin-containing mushroom extract, a lion’s mane extract, a cordyceps extract, or a valerian root extract
  • a fungal extract is prepared by taking lyophilized, dehydrated, or desiccated mushrooms or other mycelial or fungal matter (first drying such matter, if obtained fresh), and pulverizing or grinding it into a fine powder, and optionally passing it through a sieve or fine mesh followed by repeated grinding of remaining material, to ensure a preparation having uniformity of particle size.
  • “Purified extract” may refer to a botanical extract that has undergone further processing after preparation, such as soaking or heating the preparation in water and/or alcohol (depending on whether the compounds sought are water soluble), agitating, cooling the resulting liquid, straining, filtering, and removing unwanted products (repeating if necessary), and then evaporating sufficient liquid solvent to obtain a desired concentration (or entirely, to obtain a crystalline precipitate), or using a spray dryer to create a purified dried powder.
  • Extracts in some embodiments may be “standardized,” which refers to extracts that include one or more desired compounds (e.g., psilocybin) in a specific concentration. Methods to produce standardized extracts, such as by quantifying the potency of an extract and then adding excipient to dilute the extract to a standardized concentration, or further concentrating an extract to increase its potency, will be well known to those of skill.
  • Natural may refer to a substance which was isolated, extracted, or otherwise obtained from a natural source, such as a plant or fungus.
  • a natural compound such as a natural cannabinoid, a natural isoprenoid (or natural terpene), or a natural flavonoid, would therefore be within the scope of the invention if it was isolated or extracted from a Cannabis plant, or from a Psilocybe spp. or other mushroom (i.e., as a Botanical drug substance or drug product).
  • the compositions shall be derived from botanical sources including Cannabis plants and mushrooms, as extracts or by other means, and shall comprise one or more Botanical drug substances or Cannabis -derived drug substances.
  • compositions shall comprise one or more Botanical drug products or Cannabis -derived drug products. In other embodiments, such compositions shall comprise one or more Botanical drug products or Cannabis -derived drug products which are substantially free from impurities.
  • Methods of extracting cannabinoids, isoprenoids, and flavonoids from Cannabis and other plants, and creating plant and fungal extracts, and methods of obtaining purified products containing desired compounds free from undesired plant or fungal matter, chemicals, and other impurities are known in the art and described in, e.g., U.S. Pat. Nos.
  • Synthetic may refer to a substance which is manufactured artificially in a laboratory, by means of chemical synthesis (e.g., by a series of chemical processes or reactions using chemical substrates, reagents, and optionally one or more catalysts) or biosynthesis (e.g., from a bioengineered organism, and thus including those compounds also referred to as “biosynthetic” or as involving “synthetic biology” or “synbio”). (“Natural” will be understood to therefore exclude such compounds.) For example, yeast, E.
  • coli, or other host cells can be bioengineered to produce synthetic psilocybin or other compounds by inserting genes that produce appropriate enzymes and/or by altering the natural metabolic pathway to achieve the production of the desired compounds from one or more precursors.
  • Compounds can then be obtained and purified. Through such means, compounds may be obtained that are the same as natural compounds from plants or fungi, or that are derivatives or analogs thereof; examples are provided below.
  • “Nutraceutical” may refer to a preparation that could be marketed as a dietary supplement (sometimes called a nutritional supplement), e.g., in the U.S. under the appropriate regulations of the Federal Food, Drug, and Cosmetic Act (FDCA) (22 U.S.C. ⁇ 301 et seq.) and Dietary Supplement Health and Education Act (DSHEA) of 1994.
  • a dietary supplement is a product taken by mouth that contains a dietary ingredient intended to supplement the diet.
  • the dietary ingredients in these products may include: vitamins, minerals, herbs or other botanicals, amino acids, and substances such as enzymes, organ tissues, glandulars, and metabolites.
  • Dietary supplements can also be extracts or concentrates, and may be found in many forms such as tablets, capsules, softgels, gelcaps, liquids or powders.
  • “nutraceuticals” could be marketed as dietary supplements, products need not in fact meet any specific regulatory standards (such as under DSHEA or other FDA regulations), or be considered under any specific regulatory standards to be considered nutraceuticals for purposes of the definition herein.
  • nutraceuticals are also products that are sold as “natural products,” or otherwise outside of any specific regulatory regime.
  • cosmeticals which, although without legal definition under the FDCA, are understood to refer to cosmetic products that have medicinal or drug-like benefits.
  • compositions of the invention may be used interchangeably herein with regard to compositions of the invention, it will be appreciated that depending on the regulatory regime in which such compositions are available to an individual, a disclosed embodiment may or may not need to be prescribed or “recommended” by a doctor or medical professional, may or may not be available for purchase at a “dispensary” or other state or local jurisdiction licensed facility, and may or may not be able to be self-administered, including with or without supervision.
  • Embodiments may include (depending on the regulatory regime) a dosing regimen of a psilocybin-comprising mushroom composition, which is prescribed by a doctor or received under the advice or care of a medical or healthcare professional, followed by a cannabis composition which may be obtained at a dispensary or otherwise over the counter, or without medical prescription.
  • a dosing regimen of a psilocybin-comprising mushroom composition which is prescribed by a doctor or received under the advice or care of a medical or healthcare professional, followed by a cannabis composition which may be obtained at a dispensary or otherwise over the counter, or without medical prescription.
  • Psychedelic effects refer to subjective alterations in perception, cognition, emotion, or consciousness that can affect and usually interfere with the ability to perform everyday tasks and activities.
  • psychedelic effects can be made by a subject following consumption of a composition of the invention, such determination also can be made by use of or by reference to psychometric rating scales such as those known in the art, e.g., the Hallucinogen Rating Scale (HRS), Mystical Experience Questionnaire (MEQ), and Addiction Research Center Inventory (ARCI) ( see Bouso et al., 2016).
  • HRS Hallucinogen Rating Scale
  • MEQ Mystical Experience Questionnaire
  • ARCI Addiction Research Center Inventory
  • Psychoactive may refer to a substance that is capable of changing nervous system function or causing short- or long-term alterations in perception, mood, consciousness, cognition, or behavior.
  • prodrug may refer to a precursor of a biologically active pharmaceutical agent. Prodrugs undergo a chemical or a metabolic conversion to become a biologically active pharmaceutical agent. A prodrug can be converted ex vivo to the biologically active pharmaceutical agent by chemical transformative processes.
  • a prodrug is converted to the biologically active pharmaceutical agent by the action of a metabolic process, an enzymatic process or a degradative process that removes the prodrug moiety, such as a glycoside, to form the biologically active pharmaceutical agent.
  • the compounds of the invention include the prodrugs thereof.
  • psilocybin is a prodrug of psilocin, but other compounds, e.g., those which are also prodrugs of psilocybin, such as psilacetin (or such other compounds used in the compositions of the invention), will be understood to be prodrugs within the meaning herein, along with prodrugs of other compounds, including cannabinoids (e.g., with THC-O-acetate, a prodrug of THC), and known prodrugs of CBD.
  • cannabinoids e.g., with THC-O-acetate, a prodrug of THC
  • isolated may refer to material that is substantially or essentially free from components that normally accompany the material in its raw, untreated, or native state or when the material is produced.
  • a “substantially pure” or “isolated” preparation of a compound may accordingly be defined as a preparation having a chromatographic purity (of the desired compound) of greater than 90%, more preferably greater than 95%, more preferably greater than 96%, more preferably greater than 97%, more preferably greater than 98%, more preferably greater than 99% and most preferably greater than 99.5%, as determined by area normalization of an HPLC profile or other similar detection method.
  • the substantially pure compound used in the invention is itself substantially free of any other active compounds, which may cause effect in a subject when administered, but which are not intended to be administered to the subject.
  • substantially free can be taken to mean that no compounds other than the target compound are detectable by HPLC or other similar detection method.
  • the treatment options provided by the compositions and methods of the invention will be better than one or more current treatment options for mental health conditions available to patients. Such current treatment options consist generally of psychotherapy, pharmacotherapy, and direct brain intervention. All suffer serious drawbacks.
  • the compositions and methods of the invention will be better than psychotherapy.
  • Psychotherapy takes various forms including cognitive behavioral therapy (CBT), exposure therapy, eye movement desensitization and reprocessing (EMDR), and numerous forms of talk therapy (often, paired together with a pharmacological agent).
  • CBT cognitive behavioral therapy
  • EMDR eye movement desensitization and reprocessing
  • talk therapy often fails to provide complete or even partial symptom reduction.
  • compositions and methods of the invention will be no worse than, at least as good as, and preferably better than, psychotherapy.
  • compositions and methods of the invention will be no worse than, at least as good as, and preferably better than, CBT, exposure therapy, and/or EMDR. “No worse than,” “at least as good as,” and “better than” may be determined, for example, by use of a non-inferiority trial, an equivalence trial, or a superiority trial, as known in the art and described in Examples herein.
  • compositions and methods of the invention will be provided in conjunction with psychotherapy, and will be no worse than, at least as good as, and preferably better than, psychotherapy alone; or no worse than, at least as good as, and preferably better than, psychotherapy in conjunction with known pharmacotherapies, such as SSRIs or other antidepressants, or other pharmacological treatments.
  • known pharmacotherapies such as SSRIs or other antidepressants, or other pharmacological treatments.
  • the treatment options provided by the compositions and methods of the invention will be better than one or more pharmacological treatment options for mental health conditions available to patients.
  • the compositions and methods of the invention will be no worse than, at least as good as, and preferably better than, one or more “first-line” pharmacological treatment options, including for mental health conditions such as depression, panic and anxiety disorders, obsessive compulsive disorder, phobias, bulimia, adjustment disorder, and PTSD.
  • compositions and methods of the invention will be no worse than, at least as good as, and preferably better than, use of selective serotonin reuptake inhibitors (SSRIs), which block the reabsorption (i.e., reuptake) of serotonin into neurons, thereby increasing levels of serotonin in the brain.
  • SSRIs selective serotonin reuptake inhibitors
  • the drugs are generally slow to achieve meaningful benefit, requiring weeks or months to produce therapeutic effects, while also significantly increasing the risk of serious adverse events.
  • patients once prescribed, patients generally continue to take them for their entire lives.
  • compositions and methods of the invention will be no worse than, at least as good as, and preferably better than, SSRIs.
  • monoamine releasers Another broad class of agents for treatment of mental health conditions is monoamine releasers, which induce the release of monoamine neurotransmitters (e.g., dopamine, serotonin, or epinephrine) from neurons.
  • monoamine releasers rapidly modulate the brain systems that are more gradually affected by SSRIs. However, their stimulant and euphoric effects create high abuse liability.
  • compositions and methods of the invention will be no worse than, at least as good as, and preferably better than, psychostimulants and other monoamine releasers.
  • Various FDA-approved drugs are also prescribed to treat symptoms of mental health conditions “off label” (i.e., for unapproved indications). Prazosin (Minipress ® ), for instance, is an alpha-blocker and antihypertensive often used off label for PTSD-related nightmares and sleep disruption.
  • compositions and methods of the invention will be no worse than, at least as good as, and preferably better than, current off label treatments for mental health conditions.
  • a last resort for patients with serious or refractory (i.e., treatment resistant) conditions is direct brain interventions. For instance, one option when other treatments are unsuccessful is electroconvulsive therapy (ECT).
  • ECT is a procedure, done under general anesthesia, in which electric currents are passed through the brain, intentionally triggering a brief seizure.
  • ECT electroconvulsive therapy
  • Various surgical procedures also have been developed. These primarily involve targeted ablative procedures, i.e., causing the irreversible destruction of specific brain tissue.
  • compositions and methods of the invention will be no worse than, at least as good as, and preferably better than, direct brain interventions like ECT, surgical procedures such as targeted ablative procedures, and deep brain stimulation.
  • compositions and methods of the invention will be no worse than, at least as good as, and preferably better than, psilocybin-assisted and other psychedelic-assisted psychotherapy.
  • Psilocybin and its History [81] Psilocybin is a naturally occurring compound produced by several hundred species of fungus, most from the genus Psilocybe , but also from a number of other genera. Psilocybin acts as a prodrug in animals, and is rapidly converted to psilocin.
  • psilocybin causes mind-altering “psychedelic” effects such as euphoria, visual and mental hallucinations, changes in consciousness, a distorted sense of time, and experiences labeled as numinous, mystical, or “spiritual,” even by those who do not profess to be religious.
  • the term “psychedelic” was coined in 1953 by the psychiatrist Humphrey Osmond, during written correspondence with author Aldous Huxley.
  • Psychedelic derives from two Ancient Greek words, psyche meaning “mind” or “soul,” and delos meaning “reveal,” or “manifest.” At psychedelic dosages, psilocybin also can cause possible adverse reactions such as nausea, panic attacks, and negative or disturbing experiences known as “bad trips.” [83] Evidence is cited to suggest that psychoactive mushrooms containing psilocybin have been used by humans in religious ceremonies for thousands of years.
  • psilocybin must be taken in an appropriate psychotherapeutic setting. Further, because a psychedelic experience can last 6-8 hours, psychedelic-assisted therapy requires significant time from trained personnel. [90] At these higher dosages, besides significant psychedelic effects, potential adverse effects also increase in risk or severity. For example, psilocybin not only activates the 5-HT 2A receptors thought to be responsible for its beneficial therapeutic effects, but also activates 5-HT 2B receptors.
  • compositions and methods of the invention will be no worse than, at least as good as, and preferably better than, current treatments by having reduced cardiovascular risk or reduced cardiac adverse effects, by having reduced effects including long-term effects at 5-HT 2B receptors.
  • compositions and methods of the invention will be no worse than, at least as good as, and preferably better than, current treatments by having increased synergistic and entourage effects.
  • Cannabis and Mental Health Conditions [92] Cannabis is a genus of flowering plant in the family Cannabaceae that is commonly recognized as containing the three species C. sativa , C. indica , and C. ruderalis . Cannabis is believed to be one of the first plants to be cultivated, and has a long history of human use for medicinal purposes.
  • Cannabis use became increasingly vilified, culminating in its illegalization and criminalization in most countries. Although extensive use continued, including for medical purposes (occasionally gaining wide notice, for instance during the AIDS crisis), legalization attempts throughout the end of the 20 th century repeatedly stalled.
  • Cannabis remains illegal in most countries, and is illegal at the federal level in the United States, on Schedule I of the CSA, and determined to have “no currently accepted medical use.”
  • U.S. states have now legalized Cannabis for medical and/or recreational (“adult use”) purposes, research into the therapeutic benefits of Cannabis (particularly as randomized controlled trials [RCTs], the gold standard for clinical research) remains underfunded and arduous to pursue.
  • RCTs randomized controlled trials
  • the primary U.S. federal agency that funds Cannabis research is the National Institute on Drug Abuse (NIDA), resulting in research with a focus on dependence and adverse effects, rather than on potential therapies.
  • NIDA National Institute on Drug Abuse
  • Cannabis use prevalent among those with mental disorders? Or are mental health conditions prevalent among Cannabis users? While the scientific literature is clear that use is highly correlated with major depressive, psychotic, anxiety, post-traumatic stress, bipolar, and other disorders, the bias toward viewing Cannabis use through the lens of dependency, addiction, and illegality has led many to conclude that its use causes or contributes to these disorders, rather than is a means to treat them. [95] Anecdotally, Cannabis users commonly report reliance on Cannabis to treat symptoms of mental health conditions.
  • Cannabis users broadly report that Cannabis helps with anxiety, a review of scientific literature reveals a relationship that is uncertain and complex.
  • some compounds in Cannabis e.g., THC
  • CBD include anxiolytic properties (i.e., reducing symptoms).
  • THC is shown to mitigate stress at relatively lower doses (7.5 mg), but exacerbate stress at relatively higher doses (12.5 mg).
  • dose dependent effects tend to decrease with frequency of use, with infrequent Cannabis users less tolerant of larger doses of THC.
  • Novel compositions that use different Cannabis constituents (or are based on such constituents, as derivatives, analogs, prodrugs, or synthetic versions thereof), and that are able to account for each of these factors, may improve treatment of mental disorders.
  • the bioactive compounds most readily associated with Cannabis are cannabinoids, generally held to be responsible for the primary mental and physical effects of Cannabis when consumed.
  • Another class of compounds is terpenes, which are responsible for the unique fragrance of different Cannabis strains, and also cause numerous mental and physical effects.
  • flavonoids provide color and aroma as well as mental and physical effects.
  • Cannabinoids are a diverse class of small molecules that are grouped together because of their ability to act on cannabinoid receptors found in the brain and throughout the central and peripheral nervous systems of humans and other mammals.
  • CBD 1 cannabinoid receptor 1
  • CBD 2 cannabinoid receptor 2
  • CB 1 receptors are primarily found in the central nervous system (i.e., the brain and spinal cord), as well as in the lungs, liver and kidneys. In fact, CB 1 receptors are the most abundant G protein-coupled receptor in the brain, and are widely expressed on almost all neuronal types in the brain, including gabaminergic, glutamatergic, serotonergic, noradrenergic, and dopaminergic terminals. CB 1 receptor-mediated signaling plays a critical role in neural circuitry that mediates mood, motivation, and emotional behaviors. CB 2 receptors are primarily found in the immune system and hematopoietic stem and progenitor cells, and may be found in neurons.
  • endocannabinoids produced endogenously in humans and other mammals may be termed “endocannabinoids.” These endocannabinoids are a class of lipid mediators, including amides, esters, and ethers of long-chain polyunsaturated fatty acids. Endocannabinoids are abundantly present in the cerebral cortex, basal ganglia, and limbic structures and exert their effects mainly through CB receptors. The first endocannabinoid was isolated in 1992, and was characterized to be N-arachidonoyl ethanolamide (AEA). AEA was subsequently named anandamide, from the Indian Sanskrit word “ananda,” meaning inner bliss.
  • AEA N-arachidonoyl ethanolamide
  • Other endocannabinoids include N-arachidonoylglycine (NAGly), N-arachidonoyldopamine (NADA), 2-arachidonoylglyceryl ether (2-AGE), and 9-octadecenoamide (oleamide).
  • endocannabinoids may produce effects upon a variety of other targets; for example, oleamide has shown activity at CB 1 receptors as well as 5-HT 1A , 5-HT 2A/2C , 5-HT 7 , and GABA A receptors (Fowler, 2004).
  • endocannabinoids and the endogenous cannabinoid receptors they act on form the “endocannabinoid system” (ECS).
  • ECS endocannabinoid system
  • the ECS is shown or suggested to be involved in a variety of physiological processes including appetite, digestion, pain sensation, mood, memory, reproduction, stress response, immune function, thermoregulation, energy balance, and sleep.
  • Cannabinoids also can be isolated from plants, including at least 100 from the cannabis plant, among other plants including echinacea, kava, tea, and flax. Cannabinoids from plants may be termed “phytocannabinoids.” These non-endogenous cannabinoids also act on cannabinoid receptors in the body, and have many structural and functional similarities with endocannabinoids. Among the naturally-occurring phytocannabinoids from Cannabis , tetrahydrocannabinol, cannabidiol, and cannabichromene are three major constituents.
  • THC Tetrahydrocannabinol
  • ⁇ 9 -THC THC isomer-trans- ⁇ 9 - tetrahydrocannabinol
  • numerous other double bond- and stereo-isomers exist.
  • One such isomer is ⁇ 8 -THC , an analog with anxiolytic, antiemetic, analgesic, appetite-stimulating, and neuroprotective qualities.
  • ⁇ 8 -THC is shown to be twice as effective an anti-emetic than ⁇ 9 -THC , but have up to 10 times less psychoactivity.
  • ⁇ 8 -THC binds to CB 1 receptors located in the CNS, with potential affinity for CB 2 receptors, but due to its altered molecular structure, has a different affinity for those receptors than ⁇ 9 -THC .
  • Trace amounts of other THC isomers can be isolated from Cannabis .
  • the seven-carbon THC homologue ⁇ 9 -trans-tetrahydrocannabiphorol (THCP) has been reported to be much more potent than five-carbon ⁇ 9 -THC , but provide similar pharmacological actions.
  • CBD cannabidiol
  • CBD is an inverse agonist of CB 2 receptors, and an indirect antagonist of CB 1 and CB 2 cannabinoid receptor agonists (i.e., an antagonist to other ligands at CB 1 and CB 2 receptors). Interaction with CB 2 receptors appears to be primarily responsible for the anti-inflammatory and other therapeutic effects of Cannabis seen in animal models. [109] While THC directly activates CB 1 and CB 2 receptors, CBD does not bind to either, but instead impacts them indirectly. The pharmacology of CBD is enigmatic, involving interactions with multiple neurochemical systems, including serotonergic and adenosinergic systems.
  • CBD Suggested mechanisms of action supporting the pharmacological effects of CBD include activation of TRPV1 channels, inhibition of uptake and metabolism of the endocannabinoid anandamide (in part through FAAH inhibition), inhibition of adenosine uptake, antagonism of GPR55 and agonism of PPAR ⁇ receptors, and increase of intracellular calcium.
  • CBD is also shown to act as an agonist of serotonergic 5-HT 1A receptors, possibly contributing to the ability of CBD to improve mood and reduce anxiety and depression.
  • Cannabichromene (CBC) is a third major naturally-occurring phytocannabinoid, which potentiates THC effects in vivo and has anti-nociceptive and anti-inflammatory effects.
  • CBC has been shown to have potential to stimulate the growth of brain cells and the ability to normalize gastrointestinal hypermotility, and is viewed as a potential anti-cancer agent.
  • cannabinoids are almost exclusively found in their “acidic” form (e.g., THCA, CBDA, CBCA), rather than their “neutral” form (e.g., THC, CBD, CBC) (and further, may have isomers, e.g., THCA-A and THCA-B).
  • Cannabinoids in acidic form have an extra carboxyl (–COOH) group attached, which acts as an acid by donating its proton (H + ) and becoming ionized (to –COO - ).
  • decarboxylation the deprotonated carboxyl group is lost as carbon dioxide (CO 2 ), leaving the neutral form.
  • the acidic cannabinoid forms have pharmacological properties, but generally must be converted into their neutral forms through decarboxylation before being active and bioavailable in humans (for instance, it is THC, and not THCA, that is responsible for the intoxicating effects of Cannabis ). How quickly decarboxylation occurs is a function of pressure, heat, and time. At lower temperatures, such as when a plant is dried and cured after harvesting, decarboxylation occurs slowly and only partially.
  • Cannabigerolic acid CBGA
  • CBDA cannabigerol
  • CBCA cannabigerol
  • Cannabinol (CBN) although not directly synthesized in Cannabis , is found in trace amounts and is weakly psychoactive.
  • CBN is a breakdown product of THC, when THC is oxidized to CBN via prolonged exposure to heat, oxygen and light.
  • Cannabis specimens or flower products tend to have increasing levels of CBN as they age.
  • CBN is observed to produce greater sedation when combined with THC, and may have anticonvulsant, anti-inflammatory, antibiotic, and analgesic properties.
  • CBN has affinity to CB 2 receptors, and acts as a partial agonist of CB 1 receptors (with lower affinity than THC).
  • phytocannabinoids from Cannabis have a pentyl side-chain, but traces of methyl and ethyl and minor amounts of propyl homologues also occur.
  • CBDV cannabidivarin
  • THCV trans-tetrahydrocannabivarin
  • cannabinoids include those as set forth and described by Radwan et al. in Cannabinoids, Phenolics, Terpenes and Alkaloids of Cannabis , Molecules, 26(9), 2774 (2021), which is incorporated by reference as if fully set forth herein.
  • cannabinoids include compounds with a characteristic C21 terpenophenolic backbone that are part of one of 11 cannabinoid sub-classes, namely: cannabichromene (CBC)-type, cannabidiol (CBD) type, cannabielsoin (CBE) type, cannabigerol (CBG) type, cannabicyclol (CBL) type, cannabinol (CBN) type, cannabinodiol (CBND) type, cannabitriol (CBT) type, ( ⁇ )- ⁇ 8 - trans -tetrahydrocannabinol ( ⁇ 8 -THC) type, ( ⁇ )- ⁇ 9 -trans-tetrahydrocannabinol ( ⁇ 9 -THC ) type, and miscellaneous-type cannabinoids.
  • CBC cannabichromene
  • CBD cannabidiol
  • CBE cannabielsoin
  • CBG cannabig
  • a cannabis composition of the invention comprises one or more ⁇ 9 -THC -type cannabinoids which include ⁇ 9 -THC -C 5 , ⁇ 9 -THC AA-C 5 , ⁇ 9 -THC AB-C 5 , ⁇ 9 -THC -C 4 , ⁇ 9 -THC AA-C 4 , ⁇ 9 -THC V, ⁇ 9 -THC VAA, ⁇ 9 -THC O, ⁇ 9 -THC OAA, ⁇ 9 -THC –aldehyde, ⁇ -fenchyl ( ⁇ )- ⁇ 9-trans-tetrahydrocannabinolate, ⁇ -fenchyl ( ⁇ )- ⁇
  • a cannabis composition of the invention comprises one or more ⁇ 9 -THC-type cannabinoids which include ⁇ 8 -THC , ⁇ 8 -THC A, 10 ⁇ -OH- ⁇ 8 -THC , 10 ⁇ -OH- ⁇ 8 -THC , and 10a- ⁇ -hydroxy-10-oxo- ⁇ 8-THC. c.
  • a cannabis composition of the invention comprises one or more CBG-type cannabinoids which include ( E )CBG, ( E )CBGA, ( E )CBGG, ( E )CBGAM, ( E )CBGV, ( E )CBGVA, (Z)CBGA, 5-acetyl-4-hydroxy-cannabigerol, ( ⁇ )-6,7- trans - epoxycannabigerolic acid, ( ⁇ )-6,7- cis -epoxycannabigerolic acid, ( ⁇ )-6,7- cis - epoxycannabigerol, ( ⁇ )-6,7- trans -epxoycannabigerol, camagerol, and sesquicannabigerol.
  • CBG-type Cannabinoids which include ( E )CBG, ( E )CBGA, ( E )CBGG, ( E )CBGAM, ( E )CBGV, ( E )CBGVA, (Z)CBGA, 5-
  • a cannabis composition of the invention comprises one or more CBD, CBND, CBE, and CBL-type cannabinoids which include CBD-C 5 , CBDA-C 5 , CBDM–C 5 , CBD-C 4 , CBDV, CBDVA, CBD-C 1 , CBDH, CBDP, CBDD, CBND-C 3 , CBND-C 5 , CBE-C 5 , CBEAA-C 5 , CBEAB-C 5 , CBE-C 3 , CBEAB-C 3 , CBL, CBLA, and CBLV.
  • CBD-C 5 CBDA-C 5 , CBDM–C 5 , CBD-C 4 , CBDV, CBDVA, CBD-C 1 , CBDH, CBDP, CBDD, CBND-C 3 , CBND-C 5 , CBE-C 5 , CBEAA-C 5 , CBEAB-C 5 , CBE-C 3 , CBEAB-C 3 , CBL, CBLA, and
  • a cannabis composition of the invention comprises one or more CBC and CBN-type cannabinoids which include CBC, CBCA, ⁇ CBCV, +CBCV, CBCVA, 4-acetoxy-CBC, ( ⁇ )-3”-hydroxy- ⁇ 4”-cannabichromene, (–)-7-hydroxy- cannabichromane, CBC-C 3 , CBN-C 5 , CBNA-C 5 , CBN-C 4 , CBN-C 3 , CBN-C 2 , CBN-C 1 , CBNM–C 5 , 8-OH-CBN, 8-OH-CBNA, 1’ S -OH-CBN, and 4-terpenyl-cannabinolate.
  • a cannabis composition of the invention comprises one or more CBT-type cannabinoids which include ( ⁇ )- trans -CBT-C 5 , (+)- trans -CBT-C 5 , ( ⁇ )- cis -CBT-C 5 , ( ⁇ )- trans -CBT-C 3 , CBT-C 3 -homologue, ( ⁇ )- trans -CBT-OEt-C 5 , (–)- trans -CBT-OEt-C 3 , 8,9-Di-OH-CBT-C 5 , and CBDA-C 5 , and 9-OH-CBT-C 5 ester.
  • CBT-type cannabinoids which include ( ⁇ )- trans -CBT-C 5 , (+)- trans -CBT-C 5 , ( ⁇ )- cis -CBT-C 5 , ( ⁇ )- trans -CBT-C 3 , CBT-C 3 -homologue, ( ⁇ )- trans -CBT-OEt-
  • a cannabis composition of the invention comprises one or more miscellaneous-type cannabinoids which include DCBF-C 5 , CBF-C 5 , OH-iso-HHCV-C 3 OTHC, cannabicitran, cis - ⁇ 9 -THC, CBCON-C 5 , CBR, CBTT, CBCN-C 5 , CBCN-C 3 , cis -iso- ⁇ 7 -THCV, trans -iso- ⁇ 7 -THCV, trans -iso- ⁇ 7 -THC, CBCNB, CBCNC, CBCND, (–)-(7 R )-cannabicoumarononic acid, 4-acetoxy-2-geranyl-5-hydroxy-3- n - pentylphenol, 2-geranyl-5-hydroxy-3- n -pentyl-1,4-benzoquinone, 5-acetoxy-6-geranyl-3-
  • Each cannabinoid also will be understood to include its isomers, such as structural isomers and stereoisomers (including enantiomers), the -A and -B isomers for each cannabinoid, double bond isomers, and other such isomers known to those of skill.
  • THC will, in embodiments, be understood to include THC–A and THC–B.
  • cannabinoids included among cannabinoids are the cannabinoid carboxylic acids and their carboxylate salts (see, e.g., U.S. Pat. No.9,376,367).
  • reference to a cannabinoid includes the various alkyl chain lengths associated therewith, as illustrated by C n , wherein “ n ” refers to the number of carbon atoms in each alkyl chain.
  • THC also includes THC-C1, THC-C2, THC-C3, THC-C4, THC-C5, THC-C6, and THC-C7.
  • reference to a given cannabinoid will include all possible isomers, such as but not limited to its -A and -B isomers, and all possible combinations of alkyl chain lengths, including chains comprised of 1, 2, 3, 4, 5, 6, or 7 carbon atoms.
  • THC will include THC-C1 A, THC-C1 B, THC-C2 A, THC-C2 B, THC-C3 A, THC-C3 B, THC-C4 A, THC-C4 B, THC-C5 A, THC-C5 B, THC-C6 A, THC-C6 B, and THC-C7 A, and THC-C7 B.
  • THC is merely used to illustrate such logic, and should not be construed as limiting.
  • cannabinoids may further comprise one or more chemical moieties including, but not limited to, methyl, alkyl, alkenyl, methoxy, alkoxy, acetyl, carboxyl, carbonyl, oxo, ester, hydroxyl, aryl, heteroaryl, cycloalkyl, cycloalkenyl, cycloalkylalkenyl, cycloalkenylalkyl, cycloalkenylalkenyl, heterocyclylalkenyl, heteroarylalkenyl, arylalkenyl, heterocyclyl, aralkyl, cycloalkylalkyl, heterocyclylalkyl, heteroarylalkyl, and the like, and still be within the spirit of the invention as disclosed herein.
  • Non-Cannabinoid Compounds [125] In addition to cannabinoids, many hundreds of non-cannabinoid compounds are found in Cannabis , including non-cannabinoid phenols (including spiro-indans, dihydrostilbenes, dihydrophenathrenes, and simple phenols, according for purposes of organization and explication only, and using the classification system discussed above, but not to be limited thereby), as well as flavonoids and terpenes (as discussed in separate sections below), and alkaloids, such as the spermidine alkaloids cannabisativine and anhydrocannabisativine. a.
  • non-cannabinoid phenols including spiro-indans, dihydrostilbenes, dihydrophenathrenes, and simple phenols, according for purposes of organization and explication only, and using the classification system discussed above, but not to be limited thereby
  • flavonoids and terpenes as discussed in separate sections below
  • alkaloids such as
  • a cannabis composition of the invention comprises one or more Spiro-Indans which include cannabispiran, cannabispirenone, cannabispirenone isomer, cannabispiradienone, ⁇ -cannabispirol, ⁇ -cannabispirol acetate, 7-methoxyindan-1-spiro- cyclohexane, 7-hydroxy-5-methoxyindan-1-spiro-cyclohexane, 5,7-dihydroxyindan-1-spiro- cyclohexane, isocannabispiran, 7-O-methyl-cannabispirone, isocannabispiradienone, ⁇ -cannabispiranol, cannabispirketal, ⁇ -cannabispiranol-4′- O- ⁇ -glucopyranose, and prenylspirodienone.
  • Spiro-Indans which include cannabispiran, cannabispirenone, cannabispirenone isomer, cannabispiradienone, ⁇ -cannabispirol
  • a cannabis composition of the invention comprises one or more dihydrostilbenes which include 3-[2-(4-hydroxyphenyl)-ethyl]-5- methoxyphenol, 3-[2-(3-hydroxy-4-methoxyphenyl)-ethyl]-5-methoxyphenol, 3-[2-(3- isoprenyl-4-hydroxy-5- methoxy-phenyl)-ethyl]-5-methoxyphenol, canniprene, cannabistilbene I, cannabistilbene II, 3,4′,5-trihydroxy-dihydrostilbene, ⁇ , ⁇ ′-dihydro- 3′,4,5′-trihydroxy-4′-methoxy- 3-isopentenylstilbene, ⁇ , ⁇ ′-dihydro-3,4′,5-trihydroxy-4-methoxy-2,6-diisopentenylstilbene, ⁇ , ⁇ ′-dihydro- 3′,4,5′-trihydroxy-
  • a cannabis composition of the invention comprises one or more dihydrophenanthrenes which include cannabidihydrophenanthrene (cannithrene 1), cannithrene 2, 4,5-dihydroxy-2,3,7-trimethoxy-9,10-dihydrophenanthrene, 4-hydroxy- 2,3,6,7-tetramethoxy-9,10-dihydrophenanthrene, 4,7-dimethoxy-1,2,5- trihydroxy- phenanthrene, denbinobin, and dihydrophenanthrenedione.
  • cannabidihydrophenanthrene cannithrene 1
  • cannithrene 2 4,5-dihydroxy-2,3,7-trimethoxy-9,10-dihydrophenanthrene
  • 4-hydroxy- 2,3,6,7-tetramethoxy-9,10-dihydrophenanthrene 4,7-dimethoxy-1,2,5- trihydroxy- phenanthrene, denbinobin, and dihydrophenanthren
  • a cannabis composition of the invention comprises one or more cannabis simple phenols which include eugenol, methyleugenol, iso-eugenol, trans-anethol, cis-anethol, vanillin, and phloroglucinol ⁇ -D-glucoside.
  • Terpenes are a large and diverse class of organic hydrocarbon compounds, produced by a broad variety of plants, including Cannabis . Terpenes are ubiquitous throughout nature, with some estimates concluding that as many as 60 percent of all naturally-produced chemical compounds are members of the class (totaling over 20,000 unique compounds).
  • Terpene is derived from “turpentine,” the pungent resin extracted from the terebinth tree Pistacia terebinthus .
  • Terpenes may be referred to as “terpenoids” when they have experienced oxidation and have an additional oxygen-containing functional group (for instance, after cannabis is cured and dried), or when they are otherwise modified by addition or removal of a functional group (e.g., a methyl group (–CH 3 )).
  • a functional group e.g., a methyl group (–CH 3 )
  • the presence and particular combination of terpenes gives different plants (and different cannabis strains) their distinctive smells and tastes.
  • terpenes are the largest group of phytochemicals, with at least 120 identified molecules. Terpenes generally make up between 10-20 percent of the total oil content produced by Cannabis resin glands.
  • Terpenes also constitute the majority of chemicals in the heated or vaporized smoke of Cannabis flowers, often consisting of greater than 50%, with cannabinoids normally accounting for 10-20%.
  • terpenes are constituted by one or more repeating units of a five-carbon building block known as an isoprene unit (i.e., 2-methyl-1,3-butadiene , having the molecular formula C 5 H 8 ).
  • Isoprene units may be linked together end-to-end to form linear chains, or may be arranged so as to form rings (thus having the molecular formula (C 5 H 8 ) n , where n is the number of linked isoprene units).
  • Terpenes are classified in families according to the number of isoprene units from which they are constituted: as hemiterpenes (one unit), monoterpenes (two), sesquiterpenes (three), diterpenes (four), sesterterpenes (five), triterpenes (six), sesquarterpenes (seven), tetraterpenes (eight), and polyterpenes (nine or more).
  • Monoterpenes generally dominate the terpene profile of Cannabis .
  • Monoterpenes include myrcene, d-limonene, ⁇ - and ⁇ -pinene, terpinolene and linalool.
  • Sesquiterpenes and ⁇ -caryophyllene and ⁇ -humulene in particular also occur to a large extent in cannabis, as do triterpenes (as ⁇ -amyrin, friedelin and epifriedelanol, cycloartenol, and dammaradienol).
  • triterpenes as ⁇ -amyrin, friedelin and epifriedelanol, cycloartol, and dammaradienol.
  • cannabinoids are more popularly understood to be responsible for the mental and physical effects of Cannabis , terpenes have demonstrated a wide array of such effects as well ( see generally, e.g ., Russo, 2011).
  • the terpene ⁇ -myrcene has been shown to have a sedative effect, and is believed to be responsible for a heavy “body high.” Indeed, preparations of ⁇ -myrcene from hops have been employed as a sleep aid. Furthermore, the effects of ⁇ -myrcene have been demonstrated to be blocked by the drug naloxone, which is also used to block the effects of opioids, especially in overdose. ⁇ -myrcene has also demonstrated the ability to reduce inflammation and block hepatic carcinogenesis, and act as an analgesic and muscle relaxant.
  • ⁇ -myrcene the terpenes linalool, nerolidol, and pulegone have also shown sedative effects. Others, such as limonene and terpinolene, have by contrast shown stimulating effects. And others show yet different effects.
  • ⁇ -humulene acts as an appetite suppressant. Limonene exhibits anti-cancer, anxiolytic, and immunostimulating properties, and nerolidol also has anti-cancer properties.
  • ⁇ -caryophyllene possesses anti-inflammatory and gastric cytoprotector effects.
  • pentacyclic triterpenes such as ⁇ -amyrin and cycloartol, show anti-bacterial, anti-fungal, anti-inflammatory, and anti-cancer properties.
  • Terpenes may cause effects when consumed because of the modulation of neurotransmitter systems in the brain, as terpenes easily cross the blood-brain barrier (BBB).
  • BBB blood-brain barrier
  • Linalool for example, is shown to modulate the glutamatergic and gabaminergic neurotransmitter systems, which may explain its analgesic, anti-anxiety, anti-inflammatory, and anticonvulsant properties.
  • ⁇ -Pinene is an acetylcholinesteral inhibitor, and may thereby aid memory. And phytol, a diterpene, increases gabaminergic expression. Other terpenes have been shown to affect serotonergic and dopaminergic neurotransmitter systems. [135] Furthermore, some terpenes interact directly with the ECS. For example, ⁇ -caryophyllene selectively binds to CB 2 receptors as a functional CB 2 agonist, supporting ⁇ -caryophyllene as having an anxiolytic and antidepressant effect. Terpenes also have been shown to alter the permeability of cell membranes and thereby modulate the effects of THC and other cannabinoids.
  • terpenes are lipophilic, they interact with lipid membranes, ion channels, a variety of different receptors (including both G protein-coupled odorant and neurotransmitter receptors), and enzymes. Through these and other mechanisms, the terpenes in cannabis do not only cause effects individually, and in combination with other terpenes, but may also modulate the effects of the different cannabinoids that are present.
  • terpenes according to the categorization of Radwan 2021 include monoterpenes (C 10 skeleton), sesquiterpenes (C 15 skeleton), diterpenes (C 20 skeleton), triterpenes (C 30 skeleton), and miscellaneous terpenes.
  • monoterpenes C 10 skeleton
  • sesquiterpenes C 15 skeleton
  • diterpenes C 20 skeleton
  • triterpenes C 30 skeleton
  • miscellaneous terpenes Non-limiting examples of terpenes, all understood to be useful in the practice of the invention, are known by reference to the disclosure of Radwan 2021 and the below. a.
  • a cannabis composition of the invention comprises one or more monoterpenes which include myrcene, cis- ⁇ -ocimene, trans- ⁇ -ocimene, p-cymene, ⁇ -terpinene, ⁇ -phellandrene, ⁇ -terpinene, ⁇ -terpinolene, ⁇ -phellandrene, 3-phenyl-2-methyl-prop-1-ene, ⁇ -pinene, ⁇ -pinene, camphene, ⁇ 3 -carene, ⁇ 4 -carene, sabinene, ⁇ -thujene, linalool, citral B, nerol, geraniol, ipsienol, citronellol, 2-methyl-2-heptene-6-on, geranyl acetone, m-mentha-1,8-(9)-dien-5-ol, carvacrol, carvon
  • a cannabis composition of the invention comprises one or more sesquiterpenes which include ⁇ -caryophyllene, ⁇ -caryophyllene, caryophyllene oxide, curcumene, ⁇ -trans-bergamotene, ⁇ -selinene, ⁇ -farnesene, longifolene, humulene epoxide I, humulene epoxide II, caryophyllene alcohol (caryophyllenol), ⁇ -bisabolene, allo-aromadendrene, calamenene, ⁇ -copaene, nerolidol, ⁇ -gurjunene, iso-caryophyllene, ⁇ -selinene, selina-3,7(11)-diene, selina-4(14),7(11)-diene, ⁇ -bisabolol, ⁇ -cedrene, ⁇
  • a cannabis composition of the invention comprises one or more diterpenes, triterpenes, and miscellaneous terpenes which include phytol and neophytadiene (diterpenes); friedeline and epifriedelanol (triterpenes); and vomifoliol, dihydrovomifoliol, ⁇ -ionone, and dihydroactinidiolide (miscellaneous terpenes).
  • J. Flavonoids A third class of phytochemicals produced by Cannabis are flavonoids, which account for roughly 10 percent of the bioactive compounds in the plant.
  • Flavonoids are a broad class of thousands of different water-soluble polyphenolic molecules (i.e., consisting of a phenyl group ( ⁇ C 6 H 5 ) bonded to a hydroxy group ( ⁇ OH)), of which about 20 are widely found in cannabis. Flavonoids get their name from the Latin word “flavus” for the color yellow. In fact, a primary function of flavonoids is to provide color pigmentation to plants, particularly flowers. In Cannabis , for instance, deep purple strains owe their coloration to the flavonoids known as anthocyanins and anthoxanthins.
  • flavonoids also have been shown to provide health benefits through modulation of cell signaling pathways and through various anti-inflammatory, antioxidant, anti-fungal, anti-cancer, and other effects.
  • the Cannabis flavonoid apigenin has potent anti-anxiety, anti-inflammatory, and anti-cancer properties; butin has been shown to reduce oxidative stress-related cell dysfunction.
  • Other bioactive flavonoids found in cannabis include cannaflavins, kaempferol, orientin, luteolin, quercetin, silymarin, and vitexin.
  • flavonoids all of which will be understood to be useful in the practice of the invention, are known by reference to the disclosure of Radwan 2021, and include orientin, vitexin, isovitexin, apigenin, luteolin, kaempferol, and quercetin flavonoids that may be methylated, glycosylated, prenylated, or geranylated.
  • flavonoids include the orientin, vitexin, isovitexin, apigenin, luteolin, kaempferol, and quercetin flavonoids that may be methylated, glycosylated, prenylated, or geranylated; disclosed by Radwan et al., 2021.
  • a cannabis composition of the invention comprises one or more flavonoids which include orientin, orientin-O-glucoside, orientin-7-O-glucoside, orientin-7-O-rhamnoglucoside, vitexin, vitexin-O-glucoside, vitexin-7-O-glucoside, vitexin-7-O-rhamnoglucoside, cytisoside, cytisoside-glucoside, isovitexin, isovitexin-O- glucoside, isovitexin-7-O-glucoarbinoside, isovitexin-7-O-rhamnoglucoside, apigenin-7-O- glucoside, apigenin-7-O-glucuronoid, apigenin-7-O'P-coumaroylglucoside, 6-prenylapigenin, apigenin-6,8-di-glucopyranoside, luteolin-C–glucuronide, luteolin-C–glucur
  • THCV also influences the effects of THC, although in a dose-dependent fashion, diminishing the ability of THC to activate CB 1 receptors at lower doses (i.e., counteracting the effects of THC), but activating CB 1 receptors itself at higher doses (i.e., reinforcing the effects of THC).
  • compositions of the invention comprise psilocybin and/or other tryptamines, and optionally L-theanine, niacin, and extracts from cordyceps and lion’s mane.
  • these compositions further comprise valerian extract.
  • these compositions further comprise vitamin B12.
  • they further comprise a CB 1 receptor modulator, such as CBD or PEA.
  • they may further comprise one or more other active compounds.
  • these are formulated with at least one carrier, excipient, or diluent.
  • compositions of the invention comprise an entourage-enhancing cannabinoid fraction, which may include a cannabinoid such as cannabidiol (CBD) and in “C+” embodiments, a tetrahydrocannabinol (e.g., ⁇ 9 -THC, ⁇ 8 -THC, or another intoxicating THC compound); an entourage-enhancing isoprenoid fraction; and optionally, one or more flavonoids. In embodiments, these are formulated with at least one carrier, excipient, or diluent.
  • the methods of the invention include “daily dosing regimens” comprising administration of the mushroom compositions and cannabis compositions.
  • a daily dosing regimen comprises administering both of a mushroom composition and a cannabis composition in a defined daily dosing pattern according to the distinct methods and protocols that are taught herein and claimed.
  • the methods of the invention include “therapeutic regimes” comprising administration of mushroom compositions and cannabis compositions across two or more daily dosing regimens, wherein each such daily dosing regimen may be the same (as in a repeated daily dosing regimen) or different (as in a combination daily dosing regimen).
  • compositions of the invention shall be used interchangeably when referring to the compositions of the invention, as in the “nutraceutical and/or pharmaceutical compositions of the invention,” and both shall refer to a composition that is contemplated or shown to possess therapeutic effects when administered for its intended purpose to a mammal, such as a human. Even where not so defined, the mushroom compositions and cannabis compositions of the invention shall be known to be both “pharmaceutical compositions” and “nutraceutical compositions.” “Compositions of the invention” will be used as shorthand, and include these meanings.
  • compositions or “cannabis compositions” (or the term “mushroom and cannabis compositions,” which will encompass both) it will be appreciated that the terms include such compositions as both “nutraceutical” and “pharmaceutical” compositions, even if not so stated specifically. It should be further understood that the compositions described herein shall be useful regardless of the regulatory regime under which they are ultimately sold (e.g., as prescription pharmaceutical drug products, non-prescription over-the-counter (OTC) drug products, or dietary supplements), and even if not sold under a specific regulatory regime at all (e.g., as natural products).
  • one or more supplements may be paired with, or incorporated into, the compositions of the invention.
  • supplements are known to those of skill as having a wide range of health effects, and may, in embodiments, provide additional synergistic or unexpected results when paired with certain other compounds in a composition.
  • Some embodiments may include a standardized Ginkgo biloba extract, which has demonstrated in a study on the effects of three doses (120 mg, 240 mg, 360 mg), that were assessed on performance of two related serial subtraction tasks with different levels of mental effort, an improvement in cognitive function compared with placebo (Scott, 2019).
  • “paired with” means used in a daily dosing kit as one of the “A” days.
  • the supplements paired with or incorporated into the compositions of the invention include, but are not limited to, calcium, echinacea, fish oil, ginseng, glucosamine, chondroitin sulfate, garlic, St.
  • DMAE or DMEA dimethylethanolamine
  • vitamins may be paired with and/or incorporated into the compositions of the invention.
  • vitamins offer a wide range of health benefits.
  • vitamins incorporated into, and/or paired with the compositions of the invention include, but are not limited to vitamin A (retinol, retinoic acid), vitamin B1 (thiamin), vitamin B2 (riboflavin ) , vitamin B3 (niacin), vitamin B6 (pyridoxine), vitamin B12 (cobalamin), vitamin C (ascorbic acid, L-ascorbic acid), vitamin D (calciferol), vitamin E (tocopherols and/or tocotrienols), and vitamin K (phytonadione or phylloquinone).
  • nootropics may be paired with and/or incorporated into the compositions of the invention.
  • “Nootropics,” sometimes called “smart drugs,” may refer to the class of OTC or prescription substances that are believed to boost brain performance, and sometimes also called “cognition enhancers” or “memory enhancers.”
  • Prescription nootropics include, e.g., modafinil (Provigil), certain mixed amphetamine salts (Adderall, Mydayis), methylphenidate (Ritalin), and memantine (Axura).
  • OTC nootropics include, e.g., caffeine, L-theanine, omega-3 fatty acids, racetams (e.g., piracetam, pramiracetam, phenylpiracetam, aniracetam), Ginkgo biloba , Panax ginseng , Rhodiola rosea (roseroot), and creatine. It will be readily appreciated that compounds hence may fall into more than one category herein (e.g., a “nootropic” may also be a supplement or amino acid) [157]
  • “cordyceps” may refer to any species from the genus of ascomycete fungi known as Cordyceps, whether from the subgenera Cordyceps subgen.
  • Cordyceps or Cordyceps subgen.
  • Cordylia e.g., Cordyceps sinesis or Cordyceps militaris, especially those species relatively high in the compound cordycepin (3'-deoxyadenosine), which has demonstrated antidepressant effects in animal models of depression.
  • a “compound from Cordyceps” will be understood to include, besides cordycepin, any one or more of cordycepic acid, N-acetylgalactosamine, adenosine, ergosterol, ergosteryl esters, bioxanthracenes, hypoxanthine, acid deoxyribonuclease, polysaccharide, exopolysaccharide, chitinase, macrolides, cicadapeptins, myriocin, superoxide dismutase, protease, naphthoquinone, cordy heptapeptide, dipicolinic acid, fibrinolytic enzyme, lectin, and cordymin (Tuli, 2014).
  • the invention will include fungi from the genus Hericium.
  • Hericium Species of Hericium are white and fleshy, and generally grown on dead or dying wood.
  • the fruiting bodies of Hericium generally contain a mass of icicle-like spines suspended from a branched supporting framework or an unbranched cushion of tissue, giving the species its colloquial names including “Lion’s mane,” “monkey head mushroom,” “bearded tooth mushroom,” “satyr’s beard,” “bearded hedgehog mushroom,” “pom pom mushroom,” and/or “bearded tooth fungus” (Miller, 1933).
  • “Lion’s mane” may refer to Hericium erinaceus.
  • Hericium species within the spirit and scope of the invention include H. abietis, H. alpestre, H. americanum, H. bharengense, H. botryoides, H. cirrhatum, H. clathroides , H. coralloides , H. novae-zealandiae, H. rajchenbergii, and H. yumthangense .
  • Hericenones may be extracted from the fruiting bodies of Hericium spp. (i.e., Lion’s mane), and include, as non-limiting examples: Hericenone A (5-[(2E)-3,7-dimethyl-5-oxoocta-2,6-dienyl]-4-hydroxy-6-methoxy-3H-2- benzofuran-l-one), Hericenone B (6-[(2Z)-3,7-dimethyl-5-oxoocta-2,6-dienyl]-7-hydroxy- 5-methoxy-2-(2-phenylethyl)-3H-isoindol-l-one), Hericenone C ([4-[(2E)-3,7-dimethyl-5- oxoocta-2,6-dienyl]-2-formyl-3-hydroxy-5-methoxyphenyl]methylhexadecanoate), Hericenone DD (
  • L-theanine refers to L- ⁇ -glutamylethylamide ( N 5 -ethyl-L-glutamine) in enantiomeric form, but also includes unless specified or implied by context the D-enantiomer, as well as racemic DL- mixtures, and non-racemic mixtures of any proportions.
  • Psilocin refers to 4-hydroxy-N,N-dimethyltryptamine in pure or substantially pure form, in purified or isolated form produced by biosynthesis or by chemical synthesis, or as extracts from Psilocybe and other psilocin-containing mushrooms, such as those in the genera Conocybe , Copelandia , Galerina , Gymnopilus , Inocybe , Mycena , Panaeolina , Panaeolus , Pholiotina , and Pluteus .
  • Psilocybe mushrooms may be referred to herein as shorthand, one will readily appreciate than any of the above psilocybin-producing genera, as well as other psilocybe-producing genera known in the art, will be within the meaning of the term or be equivalents.
  • psilocin are also its precursors and prodrugs, and the enantiomers, metabolites, polymorphs, salts, derivatives, analogs, and variants thereof.
  • Psilocybin refers to 4-phosphoryloxy-N,N-dimethyltryptamine in pure or substantially pure form, in purified or isolated form produced by biosynthesis or by chemical synthesis, or as extracts from Psilocybe and other psilocybin-containing mushrooms, such as those in the genera Conocybe , Copelandia , Galerina , Gymnopilus , Inocybe , Mycena , Panaeolina , Panaeolus , Pholiotina , and Pluteus .
  • Chemically synthesized psilocybin is available as a crystalline solid from, e.g., Cayman Chemical Co., Ann Arbor, MI, with purity ⁇ 98%, as Item No.14041, CAS No.520-52-5, or can be synthesized following any of several known methods including as discussed, e.g., in Nichols and Frescas (1999).
  • Biosynthesized psilocybin can be produced in genetically-modified E. coli , S. cerevisiae , and other host cells, and can be commercially sourced ( see, e.g., Fricke, 2017).
  • psilocybin Within the scope of psilocybin are also its precursors and prodrugs, and further including the enantiomers, metabolites, polymorphs (any of several), salts, derivatives, analogs, and variants thereof.
  • extraction may utilize any such extraction means described herein or otherwise known in the art.
  • a polar solvent may be used for extraction of polar molecules. While a variety of polar solvents may be utilized, water, methanol, and/or ethanol, and including combinations thereof, may be used in embodiments herein.
  • an isolated, purified, extracted, biosynthesized, or synthesized compound such as a mushroom compound, such as any one or more compounds from Psilocybe , cordyceps, or lion’s mane, ground or pulverized dried mushroom is used directly.
  • the “mushroom composition” of the invention may comprise dried Psilocybe , cordyceps, and/or lion’s mane mushroom (in some embodiments, together with a carrier, diluent, or excipient, for example to make it easier to work with and capsulize), in some embodiments, may consist essentially of dried Psilocybe , cordyceps, and/or lion’s mane mushroom, and in some embodiments, may consist of dried Psilocybe , cordyceps, and/or lion’s mane mushroom, or consist of dried mushrooms and other active ingredients.
  • extraction systems span from the relatively simple (e.g., those that include a solvent, a funnel, a cheesecloth, etc.), to those large and complex enough to fill a warehouse. Regardless of scale, many generally follow similar principles.
  • Psilocybe mushrooms are harvested and collected, dried, and pulverized.
  • the pulverized mushrooms are then combined with a solvent capable of extracting the compounds of interest (e.g., methanol, ethanol, water, or a mixture thereof) to form a slurry, and the slurry is then agitated to facilitate extraction for a specified duration of time that is generally up to, or more than, 24 hours in length.
  • a solvent capable of extracting the compounds of interest e.g., methanol, ethanol, water, or a mixture thereof
  • the extraction process is like that above, in that fungal matter may or may not be first dried, then ground and pulverized, prior to introducing the ground fungal matter to a solvent, or solvent to the fungal matter, capable of extracting the desired compounds.
  • a solvent or solvent to the fungal matter, capable of extracting the desired compounds.
  • this may be completed to extract the desired hericenones from lion’s mane, cordycepin from Cordyceps, or any other desired compound from any other such mushroom species.
  • any solvent known in the art methanol, ethanol, water, a combination thereof, etc.
  • a mushroom composition of the invention is an entourage enhanced extract.
  • an entourage enhanced extract comprises predetermined amounts of two or more of psilocybin, psilocin, norbaeocystin, baeocystin, aeruginascin, norpsilocin, and other psychoactive or “psychedelic tryptamines” from Psilocybe spp.
  • an entourage enhanced extract is produced through the extraction of a single full spectrum or broad spectrum mushroom extract comprising two or more active compounds of interest.
  • an entourage enhanced extract is produced through the extraction of and/or by the combination of multiple compounds from more than one mushroom species, wherein the mushrooms are mixed (e.g., as fungal matter, such as pulverized fungal matter) before extraction.
  • an entourage enhanced extract is produced through the extraction of multiple compounds (e.g., a broad spectrum or full spectrum mushroom extract) and/or by the combination of two separate extracts, wherein such extracts are individually produced, and combined after production, and comprise multiple compounds of interest.
  • an entourage enhanced extract is a standardized extract, which is standardized for one or more tryptamines such as psilocybin.
  • an entourage enhanced extract may be produced by combining more than one mushroom species from genera including Psilocybe (or another psilocybin-containing, or psychoactive tryptamine-containing genus), Cordyceps , and Hericium ; drying, grinding, and pulverizing the mushrooms (and including more broadly, for avoidance of doubt, all such fungal matter), introducing a solvent sufficient to extract the desired compounds (e.g., methanol, ethanol, water, or a mixture thereof), continuously agitating the mixture for between about 1 to about 24 hours, or for more than 24 hours, and then allowing the excess solvent to evaporate under appropriate conditions.
  • a solvent sufficient to extract the desired compounds
  • the mushroom composition may contain any desired composition, such that a first mushroom species may comprise between about 0% to about 99% of the mushroom composition, inclusive; a second mushroom species may comprise between about 0% to about 99% of the mushroom composition, inclusive; and a third mushroom species may comprise between about 0% to about 99% of the mushroom composition, inclusive.
  • a mushroom composition may include compounds from a fourth, a fifth, a sixth, a seventh, an eighth, a ninth, a tenth, or more than a 10th other mushroom species.
  • An entourage enhanced extract also may be produced by combining more than one mushroom extract from at least one of the Psilocybe (or other psilocybin-containing), Cordyceps , and Hericium genera.
  • extracts to be combined may be sourced in the same, or a different manner, including through use of the same or a different solvent, and under the same, or different conditions, depending on the specific extract profile desired.
  • extracts may be combined in any ratio, such that a first mushroom species may comprise between about 0% to about 99% of the resultant extract, inclusive; a second mushroom species may comprise between about 0% to about 99% of the resultant extract, inclusive; a third mushroom species may comprise between about 0% to about 99% of the resultant extract, inclusive, and the like for up to or greater than 10 mushroom species.
  • a first mushroom species may comprise between about 0% to about 99% of the resultant extract, inclusive
  • a second mushroom species may comprise between about 0% to about 99% of the resultant extract, inclusive
  • a third mushroom species may comprise between about 0% to about 99% of the resultant extract, inclusive, and the like for up to or greater than 10 mushroom species.
  • an extract of 100 mg P. cubensis may contain approximately 0.63 mg of psilocybin.
  • an extract of 275 mg P. azurenscens may contain approximately 4.895 mg.
  • a mushroom extract including an entourage enhanced mushroom extract, does not contain any of psilocybin, psilocin, and/or any other psychedelic tryptamines (i.e., those that would cause psychedelic effects in a human when administered at a psychedelic dose, or cause an equivalent effect in a non-human animal, such as a head-twitch response, as will be readily understood by those of ordinary skill).
  • “Valerian” may refer to extracts of Valeriana officinalis , from the honeysuckle family Caprifoliaceae .
  • Valerian therefore may contain one or more of the known compounds found therein, and preferably found in Valerian root, such as the alkaloids actinidine, chatinine, shyanthine, valerianine, and valerene; isovaleramide; gamma-aminobutyric acid (GABA); valeric acid or isovaleric acid; iridoids, including the valepotriates valtrate and isovaltrate; the sesquiterpenes valerenic acid, hydroxyvalerenic acid, and acetoxyvalerenic acid; and the flavanones hesperidin, 6-methylapigenin, and linarin.
  • the alkaloids actinidine, chatinine, shyanthine, valerianine, and valerene isovaleramide
  • GABA gamma-aminobutyric acid
  • valeric acid or isovaleric acid iridoids, including the valepotriates valtrate and isovaltrate
  • a composition is a mixture of natural compounds.
  • the composition is a mixture of synthetic or biosynthetic compounds which may reproduce or replicate the effects of a whole plant full spectrum, or broad spectrum extract.
  • the composition is a mixture of natural, synthetic, and/or biosynthetic compounds. Accordingly, it should be understood that the compounds used in the compositions of the invention, where such compounds are as defined above, all may be obtained as natural compounds (e.g., from plants or fungi), or by chemical synthesis or from a bioengineered organism, or as a mixture or combination thereof.
  • “Entourage-enhanced,” in reference to compositions containing such above-defined compounds, refers to a composition so described that has beneficial effects in a human or other mammal, when taken for its intended use, that are greater than the beneficial effects that would be obtained or expected from each of the same compounds when taken alone or in isolated and purified form.
  • entourage-enhanced compounds may increase an existing therapeutic effect, provide an additional therapeutic effect, increase a desired property such as stability or shelf-life, decrease an unwanted effect or property, alter a property in a desirable way (such as pharmacokinetics or pharmacodynamics), or modulate a desired system or pathway (e.g., a neurotransmitter system such as the serotonergic system).
  • the cannabis compositions contain at least one cannabinoid.
  • the at least one cannabinoid is any of CBC-type cannabinoids, CBD-type cannabinoids, CBE-type cannabinoids, CBG-type cannabinoids, CBL-type cannabinoids, CBN-type cannabinoids, CBND-type cannabinoids, CBT-type cannabinoids, ⁇ 8 -THC-type cannabinoids, ⁇ 9 -THC-type cannabinoids, and miscellaneous-type cannabinoids.
  • a cannabinoid is a natural cannabinoid (e.g., a phytocannabinoid).
  • a cannabinoid is a synthetic cannabinoid.
  • the cannabis composition comprises more than one cannabinoid.
  • the cannabinoids may be natural cannabinoids, may be synthetic cannabinoids, or may be a combination of natural and synthetic (including biosynthetic) cannabinoids.
  • synthetic cannabinoids may include any of AKB48, AZ-11713908, AZD-1940, CP55940, CP-47497, CP-47497 C8, JWH-015, WIN55212-2, HU-210, HU-239, JWH-018, AM–2201, JWH-122, JWH-073, JWH-081, JWH-200, JWH-210, UR-144, and XLR11.
  • synthetic cannabinoids may include pharmaceutical compounds such as nabilone (Cesamet ® ), which is structurally distinct from ⁇ 9 -THC, but mimics its structure and pharmacological activity through weak partial agonist activity at CB 1 and CB 2 receptors, although twice as active. Numerous other synthetic variants and analogs of natural cannabinoids exist, and some are shown to have altered or enhanced activity.
  • nabilone Cesamet ®
  • the cannabis composition comprises at least one isoprenoid.
  • Isoprenoids may refer to any of the class of organic hydrocarbon isoprene polymers constituted by one or more repeating units of the five-carbon building block known as the isoprene unit (i.e., 2-methyl-1,3-butadiene, C 5 H 8 ), including those compounds known as “terpenoids” and “terpenes.” “Isoprenoids” thus includes such terpenes as those structurally found in linear chains or in rings, and those having any number of isoprene units, i.e., whether as hemiterpenes (one unit), monoterpenes (two), sesquiterpenes (three), diterpenes (four), sesterterpenes (five), triterpenes (six), sesquarterpenes (seven), tetraterpenes (eight), or polyterpenes (nine or more).
  • isoprene unit i.e., 2-methyl-1,3-butadiene, C 5 H 8
  • Isoprenoids may be derived from a botanical source, whether from Cannabis or another plant, or may be synthetic.
  • One of skill, in practicing the invention, will understand where to obtain isoprenoids, and how to balance various factors (e.g., purity, cost, ease of sourcing, marketing and advertising goals and constraints) when determining whether to use natural and/or synthetic versions.
  • the isoprenoids are terpenes selected from the group consisting of alpha-bisabolol, beta-caryophyllene, camphene, carene, caryophyllene oxide, alpha-humulene, fenchol, guaiene, guaiol, limonene, linalool, myrcene, nerolidol, ocimene, alpha-phellandrene, alpha-pinene, beta-pinene, alpha-terpinene, gamma-terpinene, terpineol, and terpinolene.
  • At least one of the terpenes in an entourage-enhancing isoprenoid fraction is selected for its ability to provide therapeutic effects or synergistic effects, or is selected for its ability to modulate the endocannabinoid system, modulate neurosteroid biosynthesis, or modulate a neurotransmitter system
  • terpenes have other effects and uses as well, and may contribute to a composition in multiple distinct ways. For instance, limonene shows stimulating effects, and also exhibits anti-cancer, anxiolytic, and immunostimulating properties.
  • an isoprenoid or terpene may be selected for its aromatic and flavor masking properties which make it suitable or preferable for certain formulations, as will be taught herein or generally known in the art.
  • a Cannabis composition comprises a flavonoid.
  • preferred flavonoids are those from Cannabis , including anthocyanins, anthoxanthins, apigenin, butin, cannaflavins, kaempferol, luteolin, orientin, quercetin, silymarin, and vitexin.
  • flavonoids are derived from or found in other plant sources.
  • a composition comprising cannabinoids, isoprenoids, and/or flavonoids is a mixture of natural compounds.
  • the composition is a mixture of synthetic compounds.
  • the composition is a mixture of natural and synthetic compounds.
  • cannabinoids, isoprenoids, and/or flavonoids used in compositions herein all may be obtained as natural compounds (from Cannabis or other plants), as synthetic or biosynthetic compounds (by chemical synthesis or from bioengineered organisms), or as mixtures or combinations thereof.
  • Entourage-enhancing when used to describe the “entourage-enhancing cannabinoid fractions” and “entourage-enhancing isoprenoid fractions,” or any other “entourage-enhancing” fraction of a cannabis composition of the invention (as well as, for example, “entourage-enhancing tryptamine fractions” or other “entourage-enhancing” fractions of a mushroom composition), means that the composition comprising the fraction(s) so described has beneficial effects in a human or other mammal, when taken for its intended use, that are greater than: (1) the beneficial effects that would be obtained or expected from a composition identical in every way, but without comprising those same fraction(s), added to (2) the beneficial effects that would be obtained or expected from those fraction(s) alone.
  • entourage-enhancing effects may include increasing an existing therapeutic effect, providing an additional therapeutic effect, increasing a desired property such as stability or shelf-life, decreasing an unwanted effect or property, altering a property in a desirable way (such as pharmacokinetics or pharmacodynamics), or modulating a desired system or pathway (e.g., the endocannabinoid system, a neurotransmitter system, or the biosynthesis of a neuroactive steroid).
  • “Therapeutic effects” that are contemplated as being increased in embodiments of the invention include, but are not limited to, antioxidant, anti-inflammatory, analgesic, antineuropathic, antinociceptive, antimigraine, anxiolytic, antidepressant, antipsychotic, anti-PTSD, immunostimulant, anti-cancer, antiemetic, orexigenic, antiulcer, antihistamine, antihypertensive, anticonvulsant, antiepileptic, bronchodilator, neuroprotective, entactogenic, empathogenic, entheogenic, euphoric, psychedelic, sedative, and stimulant effects.
  • “Synergistic effects” will be understood as being “entourage-enhanced” effects, and thus the entourage-enhanced effects of different embodiments of the invention should be understood to include increases in potency, bioactivity, bioaccessibility, bioavailability, or therapeutic effect, that are greater than the additive contributions of the components acting alone, and/or are greater than the contribution of the isolated compounds on their own, if incorporated not as part of a whole plant (including whole fungal) extract.
  • Such additional active compounds may include any one or more of amino acids, antioxidants, anti-inflammatory agents, analgesics, antineuropathic and antinociceptive agents, antimigraine agents, anxiolytics, antidepressants, antipsychotics, anti-PTSD agents, immunostimulants, anti-cancer agents, antiemetics, orexigenics, antiulcer agents, antihistamines, antihypertensives, anticonvulsants, antiepileptics, bronchodilators, neuroprotectants, entactogens and empathogens, entheogens, psychedelics, nootropics, monoamine oxidase inhibitors, sedatives, stimulants, supplements, and vitamins.
  • an extract containing at least one cannabinoid may be obtained, wherein the at least one cannabinoid is one of, or more than one of a cannabinoid selected from the group consisting of CBC-type cannabinoids, CBD-type cannabinoids, CBE-type cannabinoids, CBG-type cannabinoids, CBL-type cannabinoids, CBN-type cannabinoids, CBND-type cannabinoids, CBT-type cannabinoids, ⁇ 8 -THC-type cannabinoids, ⁇ 9 -THC-type cannabinoids, and miscellaneous-type cannabinoids; wherein, in some embodiments, the at least cannabinoid is one of, or more than one of THC, CBD, CBC, CBND, CBDL, CBE, CBL, CBT
  • the extract may be produced by any means known to those of skill.
  • An exemplary extraction system that should not be construed as limiting is as follows: Obtaining dried cannabis flower, grinding and optionally pulverizing the flower, and placing it into an extraction system generally comprising a loop wherein chilled ethanol (kept at between -30C ⁇ and -40 ⁇ C) is circulated. The resultant extract is then evaporated, so as to remove the latent solvent, and then distilled—which occurs at a higher temperature and pressure than the evaporation stage—so as to remove impurities, and further concentrate the extract.
  • decarboxylation occurs, which removes the carboxylic acid group from the THC molecule, rendering it “active” (i.e., in a state where it will cause mind-altering effects characterized by THC ingestion or inhalation) (Harli, 2021).
  • an extraction method that does not heat the compounds to such a temperature/pressure wherein decarboxylation occurs (roughly 110 C ⁇ at standard pressure), or may remove the THC from the extract via, among other means, column chromatography.
  • compositions intended for ingestion that comprise THCA will not be decarboxylated prior to or during administration
  • compounds intended for ingestion that comprise THCA will be decarboxylated prior to or during administration, and therefore are capable of providing intoxicating effects as the neutral form of THC, will be considered “C+” compositions within the meaning of the invention.
  • Compositions comprising other acidic cannabinoids will be considered C+ or C– similarly.
  • Each chiral center may be defined, in terms of absolute stereochemistry, as (R)- or (S)-.
  • the present invention is meant to include all such possible isomers, as well as mixtures thereof, including racemic and optically pure forms.
  • Optically active (R)- and (S)-, (-)- and (+)-, or (D)- and (L)-isomers may be prepared or resolved using conventional techniques.
  • the compounds described herein contain olefinic double bonds or other centers of geometric asymmetry, and unless specified otherwise, it is intended that the compounds include both E and Z geometric isomers. Likewise, tautomeric forms are also intended to be included. [193] All compounds disclosed will be understood to include the pharmaceutically acceptable salts of such compounds.
  • “Pharmaceutically acceptable salts” refers to salts prepared from pharmaceutically acceptable non-toxic acids or bases, using conventional chemical methods. Generally, such salts are prepared by reacting the free acid or base forms of a compound with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two. “Pharmaceutically acceptable salt” when generally referring to a salt of a disclosed compound will be understood to mean a salt which is pharmaceutically acceptable and possesses the desired pharmacological activity of the parent compound ( see, e.g., Berge et al., 1977). Production of the salts of natural and synthetic cannabinoid carboxylic acids is disclosed, e.g., in U.S. Pat.
  • compositions and formulations include “mushroom compositions” and “cannabis compositions,” such as M+, M–, C+, and C– compositions.
  • the mushroom compositions and cannabis compositions are administered, e.g., to a patient in need thereof.
  • the mushroom compositions and cannabis compositions comprise a pharmaceutically acceptable carrier, diluent, or excipient.
  • a composition will comprise one or more plant or fungi extracts or compounds alone without carriers, diluents, or excipients
  • some embodiments do not have a single carrier, diluent, or excipient alone, but include multiple carriers, diluents, and/or excipients.
  • the carrier, diluent, and/or excipient (including all such carriers, diluents, and/or excipients used in a composition) will not be from a fungal source or found in fungi.
  • such carriers, diluents, and/or excipients not from a fungal source or found in fungi will further not be found in any natural source, such as any botanical source, and will not be able to be obtained as a natural product or as products in or of nature.
  • such carriers, diluents, and/or excipients will further have markedly different characteristics than any found in nature.
  • such carriers, diluents, and/or excipients will contribute to the composition as a whole such characteristics or qualities that the composition as a whole is transformed and has markedly different characteristics than any such combination of natural products found in nature.
  • compositions can be prepared by standard pharmaceutical formulation techniques such as described below or disclosed in, e.g., Remington: The Science and Practice of Pharmacy (2005) 21th ed., Mack Publ. Co., Easton, Pa.; The Merck Index (1996) 12th ed., Merck Publ. Group, Whitehouse, N.J.; Pharm. Principles of Solid Dosage Forms (1993), Technomic Publ. Co., Inc., Lancaster, Pa.; and Ansel and Stoklosa, Pharm. Calculations (2001) 11th ed., Lippincott Williams & Wilkins, Baltimore, Md.; and Poznansky et al. Drug Delivery Systems (1980), R.L.
  • “Pharmaceutically acceptable” in connection with one or more ingredients means the ingredient(s) are generally safe and, within the scope of sound medical judgment, suitable for use in contact with cells of humans and other animals without undue toxicity, irritation, allergic response, or complication, and commensurate with a reasonable risk/benefit ratio.
  • pure or substantially pure individual compounds described herein are administered as part of the mushroom compositions and cannabis compositions disclosed herein.
  • the terms “pure” or “substantially pure,” as used herein, refer to material that is substantially or essentially free from components that normally accompany the material when the material is synthesized, manufactured, or otherwise produced.
  • a “pure” or “substantially pure” preparation of a compound is accordingly defined as a preparation having a chromatographic purity (of the desired compound) of greater than 90%, more preferably greater than 95%, more preferably greater than 96%, more preferably greater than 97%, more preferably greater than 98%, more preferably greater than 99%, more preferably greater than 99.5%, and most preferably greater than 99.9%, as determined by area normalization of an HPLC profile or other similar detection method.
  • the pure or substantially pure compound used in the invention is substantially free of any other active compounds which are not intended to be administered to a subject.
  • substantially free refers to the fact that no active compound(s), other than the active compound intended to be administered to a subject, are detectable by HPLC or another similar detection method, or are below a desired threshold of detection such as defined above. a.
  • the mushroom compositions and cannabis compositions disclosed herein can be formulated into any suitable dosage form, including aqueous oral dispersions, aqueous oral suspensions, solid dosage forms including oral solid dosage forms, aerosols, controlled release formulations, fast melt formulations, effervescent formulations, self-emulsifying dispersions, solid solutions, liposomal dispersions, lyophilized formulations, tablets, capsules, pills, powders, delayed-release formulations, immediate-release formulations, modified release formulations, extended-release formulations, pulsatile release formulations, multi particulate formulations, and mixed immediate release and controlled release formulations.
  • nutraceutical and/or pharmaceutical compositions including “mushroom compositions” and “cannabis compositions,” may be referred to collectively as “formulations,” for instance when prepared with at least one carrier, diluent, or excipient, or when prepared according to any of the non-limiting exemplary formulation examples below.
  • formulations are in unit dosage form.
  • Unit dosage form may refer to a physically discrete unit suited as a unitary dosage for a subject to be treated, comprising a predetermined quantity of a composition.
  • Unit dosage forms may be used for ease of administration and uniformity of dosage.
  • Unit dosage forms can contain a single or individual dose or unit, a sub-dose, or an appropriate fraction thereof of a composition administered.
  • Unit dosage forms may include capsules, troches, cachets, lozenges, tablets, ampules and vials, which may include a composition in a freeze-dried or lyophilized state; a sterile liquid carrier, for example, can be added prior to administration or delivery.
  • Unit dosage forms may also include ampules and vials comprising a composition in aqueous or liquid form.
  • Unit dosage forms may be for transdermal administration, such as “patches” that can be adhered to the epidermis of a subject, comprising a predetermined amount of a composition.
  • the formulations are in a pharmaceutically acceptable oral dosage form, including oral solid dosage forms and oral liquid dosage forms.
  • the formulations are a pharmaceutically acceptable oral solid dosage form.
  • Oral solid dosage forms may include but are not limited to, lozenges, troches, tablets, capsules, caplets, powders, pellets, multiparticulates, beads, spheres, and/or any combinations thereof.
  • Oral solid dosage forms may be formulated as immediate release, controlled release, sustained release, extended release, or modified release formulations.
  • the solid dosage provided herein may be in the form of a tablet (including a suspension tablet, a fast-melt tablet, a bite-disintegration tablet, a rapid-disintegration tablet, an effervescent tablet, or a caplet), a pill, a powder (including a sterile packaged powder, a dispensable powder, or an effervescent powder), a capsule (including both soft or hard capsules, e.g., capsules made from animal-derived gelatin or plant-derived HPMC, or “sprinkle capsules”), solid dispersion, solid solution, bioerodible dosage form, controlled release formulations, pulsatile release dosage forms, multiparticulate dosage forms, pellets, granules, or an aerosol.
  • a tablet including a suspension tablet, a fast-melt tablet, a bite-disintegration tablet, a rapid-disintegration tablet, an effervescent tablet, or a caplet
  • a pill including a sterile packaged powder
  • the formulation is in the form of a powder. In still other embodiments, the formulation is in the form of a tablet, including a fast-melt tablet. In embodiments, formulations may be administered as a single capsule or in multiple capsule dosage form. In embodiments, a formulation is administered in two, three, four, or more capsules or tablets.
  • solid dosage forms may contain pharmaceutically acceptable excipients such as fillers, diluents, lubricants, surfactants, glidants, binders, dispersing agents, suspending agents, disintegrants, viscosity-increasing agents, film-forming agents, granulation aids, flavoring agents, sweetener, coating agents, solubilizing agents, and combinations thereof.
  • solid dosage forms also can comprise one or more pharmaceutically acceptable additives, as well as supplementary agent(s).
  • supplementary agents include preservatives, antioxidants, antimicrobial agents including biocides and biostats such as antibacterial, antiviral and antifungal agents.
  • tablets provided herein are prepared by methods well known in the art. Various methods for the preparation of the immediate release, modified release, controlled release, and extended-release dosage forms (e.g., as matrix tablets having one or more modified, controlled, or extended-release layers) and the vehicles therein are well known in the art.
  • a tablet may be made by compression or molding.
  • compressed tablets may be prepared by compressing, in a suitable machine, an active ingredient in a free-flowing form such as a powder or granules, optionally mixed with a binder, lubricant, inert diluent, preservative, surface-active or dispersing agent.
  • molded tablets may be produced by molding, in a suitable apparatus, a mixture of powdered compound moistened with an inert liquid diluent.
  • the tablets may optionally be coated or scored and may be formulated so as to provide a slow or controlled release of the active ingredient therein.
  • Generally recognized compendia of methods include Remington 2020 and Sheth et al.1980.
  • solid dosage forms are prepared by mixing the active agents of the invention with one or more pharmaceutical excipients to form a “bulk blend” composition.
  • the bulk blend composition is homogeneous, i.e., the active agents are dispersed evenly throughout so that the bulk blend may be readily subdivided into equally effective unit dosage forms, such as tablets, pills, and capsules.
  • the individual unit dosages may also comprise film coatings, which disintegrate upon oral ingestion or upon contact with diluents.
  • these formulations are manufactured by conventional pharmaceutical techniques.
  • Conventional pharmaceutical techniques for preparation of solid dosage forms include, but are not limited to, the following methods, which may be used alone or in combination: (1) dry mixing, (2) direct compression, (3) milling, (4) dry or non-aqueous granulation, (5) wet granulation, or (6) fusion (see, e.g., Lachman et al.1986).
  • Other methods include spray drying, pan coating, melt granulation, granulation, fluidized bed spray drying or coating (e.g., Wurster coating), tangential coating, top spraying, tableting, and extruding.
  • the formulations have a specific release profile.
  • oral solid dosage forms may be prepared as immediate release or as modified release formulations, e.g., controlled release, extended release, sustained release, or delayed release.
  • immediate release formulations a therapeutically effective amount of one or more active compounds is administered to facilitate rapid release.
  • Immediate release formulations may be prepared by combining a superdisintegrant such as croscarmellose sodium and different grades of microcrystalline cellulose in various ratios.
  • the plasma half-life compared to the plasma half-life of an immediate release formulation is greater by at least 5%, at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 50%, at least 75%, at least 100%, or values in between.
  • the formulations are designed to result in a comparable area under the curve, or AUC 0-24 , and a similar safety and efficacy profile, but having a delayed time to maximum concentration (t max ) of at least 5%, at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 50%, at least 75%, at least 100%, or values in between, as would be appreciated by one of skill.
  • t max delayed time to maximum concentration
  • a formulation is designed to be a product with a specific time course based on an optimum “therapeutic window,” such as less than about 30 minutes, about 30 minutes, about 45 minutes, about 60 minutes, about 90 minutes, about 2 hours, about 3 hours, about 4 hours, about 5 hours, about 6 hours, about 7 hours, about 8 hours, and more than 8 hours, and lengths of time in between.
  • oral solid dosage forms are formulated as a delayed release dosage form by utilizing an enteric coating to affect release in the small intestine of the gastrointestinal tract.
  • An enteric-coated oral dosage form may be a compressed or molded or extruded tablet/mold (coated or uncoated) containing granules, powder, pellets, beads or particles of the active ingredient and/or other composition components, which are themselves coated or uncoated.
  • the enteric-coated oral dosage form may also be a capsule (coated or uncoated) containing pellets, beads or granules of the solid carrier or the composition, which are themselves coated or uncoated.
  • enteric coatings may be used to prepare other controlled release dosage forms, including but not limited to extended release and pulsatile release dosage forms. Pulsatile release dosage forms may be formulated using techniques known in the art, such as those described in U.S. Pat.
  • the controlled release dosage form is a pulsatile release solid oral dosage form comprising at least two groups of particles, each containing active agents of the invention.
  • the first group of particles upon ingestion by a subject, provides a substantially immediate dose of the active agents of the invention, and may either be uncoated, or comprise a coating and/or sealant.
  • a single unit dosage form can provide both a first and a second dosage amount in the single form (i.e., a first dosage amount in an immediate release form, and a second dosage amount in a delayed release form).
  • gastroretentive sustained release tablets are formulated by using a combination of hydrophilic polymer (e.g., hydroxypropyl methylcellulose), together with at least one swelling agent (e.g., crospovidone, sodium starch glycolate, and croscarmellose sodium), and an effervescent substance (e.g., sodium bicarbonate).
  • hydrophilic polymer e.g., hydroxypropyl methylcellulose
  • swelling agent e.g., crospovidone, sodium starch glycolate, and croscarmellose sodium
  • effervescent substance e.g., sodium bicarbonate
  • coatings for providing a controlled, delayed, or extended release may be applied to the compositions of the invention or to a core containing the compositions, and may comprise a pharmaceutically acceptable ingredient in an amount sufficient to provide a delayed release from, for example, about 1 hour to about 7 hours following ingestion before release of the active agents.
  • suitable coatings include one or more differentially degradable coatings including pH-sensitive coatings (enteric coatings), or non-enteric coatings having variable thickness to provide differential release of the active agents. Many other types of modified release systems will be known to those of skill in the art.
  • Non-limiting examples of additional delivery systems include both polymer- and nonpolymer-based systems, silastic systems, peptide-based systems, wax coatings, bioerodible dosage forms, and compressed tablets using conventional binders (see, e.g, Liberman et al. 1990; Singh et al. 2002; U.S. Pat. Nos. 4,327,725; 4,624,848; 4,968,509; 5,461,140; 5,456,923; 5,516,527; 5,622,721; 5,686,105; 5,700,410; 5,977,175; 6,465,014; and 6,932,983).
  • the formulation is a pharmaceutically acceptable oral liquid dosage form.
  • oral liquid dosage forms include tinctures, drops, emulsions, syrups, elixirs, suspensions, and solutions, and the like.
  • oral liquid dosage forms are formulated with any pharmaceutically acceptable excipient known to those of skill for preparation of liquid dosage forms, and with solvents, diluents, carriers, excipients, and the like, chosen as appropriate to the solubility and other properties of the active agents and other ingredients.
  • solvents include, e.g., water, glycerin, simple syrup, alcohol, medium chain triglycerides (MCT), and combinations thereof.
  • oral liquid dosage forms may be monophasic or biphasic, the former being a substantially homogenous solution dissolved in water or non-aqueous solvent, while the latter refers to oral liquid dosage forms in which the active ingredients do not fully dissolve in common solvents.
  • the solid particles within a oral liquid dosage form may form a precipitate at the bottom of the container, requiring shaking to redisperse them.
  • monophasic liquid forms include syrups, linctuses, spirits/essences, elixirs, and fluid extracts.
  • Non-limiting examples of biphasic liquid forms include oral suspensions, oral emulsions, and mixtures.
  • Liquid dosage forms for oral administration may be prepared as liquid suspensions or solutions using a sterile liquid, such as but not limited to, an oil, water, an alcohol, combinations of pharmaceutically suitable surfactants, suspending agents, and emulsifying agents.
  • liquid formulations also may be prepared as single dose or multi-dose beverages.
  • suspensions may include oils. Such oils include but are not limited to peanut oil, sesame oil, cottonseed oil, corn oil, and olive oil. Suitable oils also include carrier oils such as MCT and long chain triglyceride (LCT) oils.
  • suspension preparation may also contain esters of fatty acids such as ethyl oleate, isopropyl myristate, fatty acid glycerides, and acetylated fatty acid glycerides.
  • suspension formulations may include alcohols, such as ethanol, isopropyl alcohol, hexadecyl alcohol; glycerol, and propylene glycol.
  • ethers such as polyethylene glycol; petroleum hydrocarbons, such as mineral oil and petrolatum; and water may also be used in suspension formulations.
  • a suspension is an aqueous liquid or a non-aqueous liquid, an oil-in-water liquid emulsion, or a water-in-oil emulsion.
  • Dosage forms for oral administration may be aqueous suspensions such as aqueous oral dispersions, emulsions, solutions, and syrups ( see, e.g., Singh et al.2002).
  • the liquid dosage forms may comprise additives, such as one or more (a) disintegrating agents, (b) dispersing agents, (c) wetting agents, (d) preservatives, (e) viscosity enhancing agents, (f) sweetening agents, and/or (g) flavoring agents.
  • the liquid formulations of the invention may also comprise inert diluents commonly used in the art such as water or other solvents, solubilizing agents, emulsifiers, flavoring agents, and/or sweeteners.
  • co-solvents and adjuvants also may be added to a formulation.
  • effervescent powders containing the compositions are prepared.
  • effervescent salts are used to disperse medicines in water for oral administration.
  • effervescent salts also may be packaged as single dose or multi-dose drink mixes, alone or in combination with other ingredients, such as vitamins or electrolytes.
  • effervescent salts are granules or coarse powders containing a medicinal agent in a dry mixture, usually composed of sodium bicarbonate and sodium carbonate, citric acid, and/or tartaric acid.
  • a medicinal agent in a dry mixture
  • the acids and the base react to liberate carbon dioxide gas, thereby causing “effervescence.”
  • any acid-base combination that results in the liberation of carbon dioxide may be used, as long as the ingredients are suitable for pharmaceutical use, and result in a pH of about 6.0 or higher.
  • the formulations are delivered transdermally.
  • transdermal delivery involves contacting a formulation with a subject’s skin under conditions effective for the active agent(s) in a composition to penetrate the skin and cause an effect.
  • transdermal formulations include ointments, creams, suspensions, lotions, pastes, gels, sprays, foams, oils, and the like, and combinations thereof.
  • An exemplary transdermal delivery form is a transdermal “patch” which contains a formulation.
  • Transdermal patches may be used to provide continuous or discontinuous infusion of the compounds described herein in controlled amounts. Such patches may be constructed for continuous, gradual, pulsatile, or on demand delivery of active agents.
  • a patch will be a medicated adhesive patch, i.e., a patch impregnated with a composition of the invention for application onto the skin.
  • a patch may be a single-layer or multi-layer drug-in-adhesive patch, wherein the one or more adhesive layers also contain the active agents.
  • a patch also may be a “matrix” (or “monolithic”) patch, wherein the adhesive layer surrounds and overlays the drug layer (wherein a solution or suspension of the active agents is in a semisolid matrix).
  • a “reservoir” patch may also be used, comprising a drug layer, typically as a solution or suspension of the active agents in a liquid compartment (i.e., the reservoir), separate from an adhesive layer.
  • a patch also may be part of a delivery system, for instance used with an electronic device communicatively coupled to the mobile device of a user, and coupled with a mobile application (e.g., to control the delivery rate from the reservoir, and optionally to provide information about delivery back to the application or user).
  • a mobile application e.g., to control the delivery rate from the reservoir, and optionally to provide information about delivery back to the application or user.
  • Various transdermal patch technologies may be accordingly utilized.
  • formulations are for subcutaneous, intravenous, intra-arterial, intraperitoneal, intraosseous, intramuscular, intrathecal, or intracerebroventricular injection.
  • injection formulations are prepared by dissolving, suspending, or emulsifying active agent(s) in an aqueous or nonaqueous solvent, examples of which include oils, such as vegetable oil, synthetic aliphatic acid glycerides, and esters of higher aliphatic acids or propylene glycol; and may also contain additives such as solubilizers, stabilizers, and suspending, preserving, wetting, emulsifying, dispensing, and isotonic agents.
  • oils such as vegetable oil, synthetic aliphatic acid glycerides, and esters of higher aliphatic acids or propylene glycol
  • additives such as solubilizers, stabilizers, and suspending, preserving, wetting, emulsifying, dispensing, and isotonic agents.
  • nanostructured formulations such as a nanoemulsion, a nanocapsule, a nanoparticle conjugate, or a nano-encapsulated oral or nasal spray.
  • nano as used in the terms describing various embodiments of a nanostructured formulation denotes a size range in the nanometer (“nm”) scale. Accordingly, sizes of such nanoparticle delivery vehicles include those in the range of about 1 to about 100 nm, about 100 to about 200 nm, about 200 to about 400 nm, about 400 to about 600 nm, about 600 to about 800 nm, and about 800 to about 1000 nm, as well as “microparticles” in the range of about 1000 to about 2000 nm (1-2 micrometer (“ ⁇ m”) scale).
  • lipid-based nanoparticles such as liposomes, solid lipid nanoparticles (SLN), or nanostructured lipid carriers (NLC) are used.
  • the active ingredients of a formulation are mixed with an excipient, diluted by an excipient, or enclosed within, encapsulated by, or attached to a carrier in the manufacture of the formulation.
  • the excipient serves as a diluent, it can be a solid, semi-solid, or liquid material, which acts as a vehicle, carrier, or medium for the active ingredient.
  • the formulations can be in the form of, e.g., tablets, pills, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, aerosols (as a solid or in a liquid medium), ointments, soft or hard gelatin capsules, suppositories, sterile injectable solutions, and sterile packaged powders.
  • compositions of the invention are combined with other compounds, including active compounds (i.e., those providing or capable of providing a biological effect), such as vitamins, antioxidants, amino acids (including natural L-amino acids, as well as D- and DL- amino acids, and their analogs and derivatives), probiotics, natural herbs or plant extracts, and/or food or dietary supplements, to form patches, masks, wraps, creams, gels, oral films, or the like, for various purposes such as for energy, reduced fatigue, or improved mental and physical performance (e.g., with guarana, amino acids, and vitamins), for improved performance and reduced gastrointestinal discomfort during exercise (e.g., with L-leucine, L-isoleucine, L-valine, Vitamin
  • EXAMPLE 1 Formulation of “M+” capsules Gelatin capsules, containing approximately 1 g of active ingredients, are made as follows: The psilocybe, L-theanine, niacin, and cordyceps and lion’s mane extracts are obtained (or prepared into extracts, from obtained raw materials), blended, passed through a mesh sieve (e.g., between No.10 and 20 U.S., or to obtain another suitable particle size), further ground or milled if necessary, and filled into hard or soft gelatin capsules in 1 g quantities (optionally, together with valerian root extract and/or PEA if desired; if not included, the total 1 g quantity will be reduced accordingly).
  • a mesh sieve e.g., between No.10 and 20 U.S., or to obtain another suitable particle size
  • a suitable amount of cellulose, starch, or magnesium stearate can be added.
  • Purified extracts may be used, with quantities adjusted accordingly, based on concentration.
  • standardized extracts are used, or the amount of an active agent of interest (e.g., psilocybin or another bioactive compound) is quantified.
  • an active agent of interest e.g., psilocybin or another bioactive compound
  • the amount of extract may be increased or decreased to provide a known or predetermined amount of an active agent of interest. For example, in one exemplary embodiment of this Example, 1 mg of psilocybin is desired per capsule.
  • a Psilocybe extract comprises 1% w/w of psilocybin (or a standardized extract, containing 1% w/w of psilocybin is created or obtained).100 mg of Psilocybe extract is therefore used, resulting in a capsule with 1 mg (i.e., 1% w/w of 100 mg) psilocybin. Isolated compounds also may be used, and quantities adjusted accordingly, using known methods.
  • the formulation is made as above, but without Psilocybe extract, and not containing psilocybin, psilocin, or any other psychedelic tryptamines.
  • EXAMPLE 2 Alternative formulation of “M+” capsules
  • the formulation of EXAMPLE 2 is created in the same way as that of EXAMPLE 1 , except that valerian root extract and PEA are replaced with rice hull concentrate powder (nu-flow), rice bran extract powder (nu-rice), and rice bran 100 mesh powder ribran 100.
  • the formulation is made as above, but without psilocybe extract, and not containing psilocybin, psilocin, or any other psychedelic tryptamines.
  • EXAMPLE 3 Alternative formulation of “M+” capsules The formulation of EXAMPLE 3 is created in the same way as that of EXAMPLE 2.
  • the formulation is made as above, but without Psilocybe extract, and not containing psilocybin, psilocin, or any other psychedelic tryptamines.
  • EXAMPLE 4 Formulation of “M+” tablets Tablets, each containing 790 mg of active ingredients are made as follows: The components are blended and compressed to form tablets, each weighing 1 g.
  • the formulation is made as above, but without Psilocybe extract, and not containing psilocybin, psilocin, or any other psychedelic tryptamines.
  • EXAMPLE 5 Formulation of “M+” capsules with an additional active agent
  • Capsules comprising an M+ composition and an additional active agent are made as follows: The active ingredients are blended, passed through a mesh sieve, and filled into hard or soft gelatin capsules.
  • the antidepressant or anxiolytic may be any pharmaceutical agent known to act as such by one of skill (e.g., SSRIs, serotonin-norepinephrine reuptake inhibitors (SNRIs), tricyclic antidepressants (TCAs), atypical antidepressants, benzodiazepines, buspirone, etc.).
  • Antidepressants and anxiolytics may be chosen for their ability to act as 5-HT1A agonists.
  • CBD also directly activates 5-HT1A receptors, further conferring an anti-anxiety effect. If useful for ease of blending or filling, cellulose, a starch, or magnesium stearate can be added.
  • the formulation is made as above, but without Psilocybe extract, and not containing psilocybin, psilocin, or any other psychedelic tryptamines.
  • Capsules each containing a composition of present invention with additional active agents, are made as follows: The active ingredients are blended, passed through a mesh sieve, and filled into hard of soft gelatin capsules, for example in 675 mg quantities.
  • the analgesic or anti-inflammatory may be any agent known to act as such (including both OTC and prescription medications) by one of skill (e.g., aspirin, acetaminophen, ibuprofen, naproxen, prescription NSAIDs, etc.). If useful for ease of blending or filling, cellulose, a starch, or magnesium stearate can be added.
  • the formulation is made as above, but without Psilocybe extract, and not containing psilocybin, psilocin, or any other psychedelic tryptamines.
  • EXAMPLE 7 Formulation of “M+” suspension or solution, such as a tincture Liquid suspensions or solutions, with below amounts per 1.0 ml dose, are made as follows: The active agents are measured out, blended, passed through a mesh sieve, and then mixed with a previously made solution of microcrystalline cellulose and sodium carboxymethyl cellulose in excipient (e.g., ethanol, for a tincture, or purified water). The sodium benzoate, flavor, and color are diluted with excipient and added with stirring.
  • excipient e.g., ethanol, for a tincture, or purified water
  • Sweetener e.g., sucrose or sucralose
  • Additional compounds may be included, e.g., vitamins, other active ingredients such as antioxidants or antiinflammatories herbal extracts and essential oils.
  • Sufficient further excipient is then added to produce the required volume.
  • Suspensions and solutions may be prepared in volumes of 5 ml, 10ml, 25 mL, 30 mL, 50 mL, 100 mL, or such other total volumes as practical for research use or for sale as a pharmaceutical or OTC preparation, a nutraceutical preparation or dietary supplement, or a natural product.
  • liquid formulations may be used to prepare dropper bottles (e.g., 30 mL/1 oz.
  • Liquid formulations of this Example also can be used to prepare filled softgel capsules, ampoules, or other single unit dosage forms, using methods herein disclosed or known to those of skill in the art.
  • the formulation is made as above, but without Psilocybe extract, and not containing psilocybin, psilocin, or any other psychedelic tryptamines. [226] It will be appreciated that the above formulation examples are illustrative only.
  • 100 mg of “Psilocybe extract” from Psilocybe cubensis fungal material may be replaced with 10 mg of “Psilocybe purified extract” from such material at a 10x concentration, or an equivalent amount (e.g., vis-à-vis psilocybin) of purified extract at a 2x, 3x, 4x, 5x, 6x, 7x, 8x, 9x, or greater than 10x concentration, or may be replaced with an equivalent milligram amount of isolated and purified natural psilocybin, synthetic psilocybin, or biosynthetic psilocybin, calculated based on ordinary skill, all of which will be appreciated to provide like effects (if speaking of therapeutic effect; or, if speaking of psychedelic “effects,” a like absence of such effects, if so desired).
  • Synergistic Cannabis Compositions EXAMPLE 8: Formulation of “C+” oil Liquid suspensions, each containing the below amounts per 1.0 ml dose are made as follows: Cannabinoids can be commercially sourced or obtained from, e.g., Cayman Chemical Co., Ann Arbor, MI. The CBD, THC, cannabinoids, and isoprenoids are measured out, blended, passed through a No.10 mesh U.S.
  • a carrier oil as an excipient, such as MCT oil, coconut oil, or hemp seed oil
  • a polyol e.g., vegetable glycerin
  • a lecithin e.g., soy or sunflower lecithin
  • “Weight percent,” “wt %,” or “% w/w” will be understood (here, and below, and similarly for example with regard to the mushroom compositions, unless otherwise defined) to mean as a percentage of the total cannabinoid+isoprenoid content, and further including the flavonoid content (where applicable).
  • the entourage-enhancing cannabinoid fraction contains CBG, CBN, and optionally CBDV
  • the entourage-enhancing isoprenoid fraction contains the terpenes beta-myrcene, terpinolene, limonene, ocimene, beta-pinene, beta-caryophyllene, alpha-pinene, and gamma-terpinene.
  • the entourage-enhancing cannabinoid fraction is beta-myrcene dominant. Additional compounds may be included if desired, e.g., flavonoids, vitamins, active ingredients such as antioxidants or anti-inflammatories, herbal extracts, and essential oils.
  • Suspensions may be prepared in volumes of 5 mL, 10mL, 25 mL, 30mL, 50 mL, 100 mL, or such other total volumes as practical for research use or for sale as a pharmaceutical or OTC preparation, or a nutraceutical preparation or dietary supplement.
  • suspensions may be used to prepare dropper bottles (e.g., 30 mL/1 oz. bottles) or fine mist spray (i.e., oral spray) bottles (e.g., 10 mL bottles).
  • Liquid suspensions of this Example also can be used to prepare softgel capsules, ampoules, or other single unit dosage forms, through methods herein disclosed or known to those of skill in the art.
  • the formulation is made as above, but without THC.
  • EXAMPLE 9 Formulation of “C+” tincture Liquid suspensions, each containing the below amounts per 1.0 ml dose are made as follows:
  • the CBD, cannabinoids, isoprenoids, flavonoids, sweetener (e.g., sucrose or sucralose), and xanthan gum are measured out, blended, passed through a No.10 mesh U.S. sieve, and then mixed with a previously made solution of microcrystalline cellulose and sodium carboxymethyl cellulose in excipient (e.g., ethanol or purified water).
  • excipient e.g., ethanol or purified water.
  • the sodium benzoate, flavor, and color are diluted with excipient and added with stirring.
  • the entourage-enhancing cannabinoid fraction contains cannabinoids selected from the group consisting of CBC-type cannabinoids, CBD-type cannabinoids, CBE-type cannabinoids, CBG-type cannabinoids, CBL-type cannabinoids, CBN-type cannabinoids, CBND-type cannabinoids, CBT-type cannabinoids, ⁇ 8 -THC-type cannabinoids, ⁇ 9 -THC-type cannabinoids, and miscellaneous-type cannabinoids; including but not limited to CBC, CBE, CBG, CBL, CBN, CBND, CBT, CBV, CBGM, CBDV, THCV, CBDP, THCP, and iso-THC; and the entourage-enhancing isoprenoid fraction contains terpenes selected from the group consisting of monoterpenes, diterpenes, triterpenes, sesquiterpenes
  • the cannabinoid fraction includes CBG, CBN, and optionally CBDV
  • the isoprenoid fraction includes limonene and myrcene, wherein limonene comprises 6 mg, and myrcene comprises 4 mg.
  • Additional compounds may be included if desired, e.g., flavonoids, vitamins, active ingredients such as antioxidants or anti-inflammatories, herbal extracts, and essential oils. Sufficient further excipient is then added to produce the required volume.
  • Suspensions may be prepared in volumes of 5 ml, 10ml, 25 mL, 30 mL, 50 mL, 100 mL, or such other total volumes as practical for research use or for sale as a pharmaceutical or OTC preparation, or a nutraceutical preparation or dietary supplement.
  • the formulation is made as above, but without THC.
  • EXAMPLE 10 Formulation of “C+” capsules Gelatin capsules, each containing the below active ingredients, are made as follows: The CBD, THC, cannabinoids, isoprenoids, cellulose, starch, and magnesium stearate are blended, passed through a No. 20 mesh U.S. sieve, and filled into hard of soft gelatin capsules in 250 mg quantities.
  • the isoprenoid fraction is ⁇ -myrcene dominant, with at least 20% w/w of the isoprenoid fraction consisting of ⁇ -myrcene.
  • Beta-myrcene is selected because when combined with ⁇ 9 -THC it will intensify the sedative effects thereof, increase the anxiolytic effects of the formulation, and generally improve the tolerability of THC.
  • the formulation is made as above, but without THC.
  • EXAMPLE 11 Formulation of “C+” tablets Tablets, each containing the below active ingredients, are made as follows: The components are blended and compressed to form tablets, each weighing 250 mg.
  • ⁇ 8 -THC is selected for its stability and longer shelf life compared to ⁇ 9 -THC, which is more easily oxidized to cannabinol (CBN).
  • Delta-8-THC is therapeutically effective to reduce anxiety symptoms and its anxiolytic effects are enhanced by addition of the flavonoid apigenin.
  • the formulation is made as above, but without ⁇ 8 -THC.
  • EXAMPLE 12 Formulation of “C+” sublingual or buccal tablets
  • Sublingual or buccal tablets containing the below active ingredients, are made as follows: The glycerol, water, sodium citrate, polyvinyl alcohol, and polyvinylpyrrolidone are admixed together by continuous stirring and maintaining the temperature at about 90° C. When the polymers have gone into solution, the solution is cooled to about 50-55° C and the medicament is slowly admixed. The homogenous mixture is poured into forms made of an inert material to produce a drug-containing diffusion matrix having a thickness of about 2-4 mm. This diffusion matrix is then cut to form individual tablets having the appropriate size. Butin is added to promote the reduction of oxidative stress.
  • EXAMPLE 13 Formulation of “C+” capsules with an additional active agent Capsules, each containing a composition with an additional active agent are made as follows: The active ingredients, cellulose, starch, and magnesium stearate are blended, passed through a No.20 mesh U.S. sieve, and filled into hard of soft gelatin capsules in 300 mg quantities.
  • the antidepressant or anxiolytic would be any pharmaceutical agent known to act as such by one of skill (e.g., SSRIs, serotonin-norepinephrine reuptake inhibitors (SNRIs), tricyclic antidepressants (TCAs), atypical antidepressants, benzodiazepines, buspirone, etc.).
  • SSRIs serotonin-norepinephrine reuptake inhibitors
  • TCAs tricyclic antidepressants
  • benzodiazepines benzodiazepines
  • buspirone etc.
  • EXAMPLE 14 Formulation of “C+” capsules with additional active agents
  • Capsules, each containing a composition with additional active agents are made as follows: The active ingredients, cellulose, starch, and magnesium stearate are blended, passed through a No.20 mesh U.S.
  • analgesic or anti-inflammatory would be any agent known to act as such (including both OTC and prescription medications) by one of skill (e.g., aspirin, acetaminophen, ibuprofen, naproxen, prescription NSAIDs, etc.). Cannaflavins are further included to enhance the analgesic or anti-inflammatory effects.
  • the formulation is made as above, but without THC.
  • EXAMPLE 15 Formulation of “C+” liquid for vaporization A liquid formulation is prepared containing the following ingredients:
  • composition is made as above, but without THCA.
  • EXAMPLE 16 Formulation of “C–” topical form A topical formulation may be prepared as follows: The white soft paraffin is heated until molten. The active ingredients are added and stirring is continued until dispersed. The liquid paraffin and emulsifying wax are incorporated and stirred until dissolved. The mixture is then cooled until solid.
  • the formulation is made as above, but with the addition of 6.25 mg of THC.
  • EXAMPLE 17 Formulation of “C+” transdermal delivery form
  • a formulation for a transdermal delivery device may be prepared as follows: The stabilizer, solubilizer, and permeation enhancing agent are heated and stirred until combined. The active ingredients are added after partially cooled (at a temperature suitable to prevent premature decarboxylation) but before setting and stirring is continued until dispersed. The mixture is then cooled until in its desired final form (e.g., for use in a reservoir delivery system) or admixed with an adhesive and then cooled (e.g., for use in a drug-in-adhesive patch).
  • the device may be configured to sufficiently heat the CBDA and THCA so as to decarboxylate them before absorption.
  • CBDA and THCA will have therapeutic effects in their acidic form (CBDA for instance has been shown to have anti-inflammatory effects and to reduce nausea and vomiting and enhance 5-HT 1A receptor activation in animal models; THCA also has shown anti-nausea and anti-emetic properties).
  • CBDA for instance has been shown to have anti-inflammatory effects and to reduce nausea and vomiting and enhance 5-HT 1A receptor activation in animal models; THCA also has shown anti-nausea and anti-emetic properties.
  • the formulation is made as above, but without THCA.
  • EXAMPLE 18 Formulation of “C+” injectable form
  • a formulation for injection may be prepared as follows: Active ingredients are dissolved in dimethyl sulphoxide (DMSO) in proportions of 1 g to .5 mL. Solution is brought to 37° C and vortexed for 3-5 minutes. Tetraethyleneglycol (TEG) in amount of 5 mL is added, and solution is returned to 37° C and vortexed again for 3-5 mins. Solution is mixed 1:1 with saline containing 1% cremaphor to prevents precipitation.
  • DMSO dimethyl sulphoxide
  • TAG Tetraethyleneglycol
  • Final solution will be at 10mg/mL active ingredients in 49.5% TEG, 49.5% saline, .5% DMSO, and .5% cremaphor.
  • Injection may be by any suitable means, e.g., bolus injection, IV infusion, or subcutaneous infusion, for example using a drug delivery device comprising a reservoir and a pump mechanism, configured for subcutaneous administration, and which may optionally contain a user interface or be coupled to a device with a user interface such as a smartphone.
  • a drug delivery device comprising a reservoir and a pump mechanism, configured for subcutaneous administration, and which may optionally contain a user interface or be coupled to a device with a user interface such as a smartphone.
  • the formulation is made as above, but without THC.
  • a nasal spray formulation for intranasal delivery may be prepared as follows: Solution at 10mg/mL active ingredients in 49.5% MCT, 49.5% saline, .5% DMSO, and .5% cremaphor is prepared, as above (but with MCT in place of TEG), for use in nasal spray device.
  • a nasal formulation is prepared as a dry powder for inhalation, e.g., by combining the active agents with lactose and mixing for use with a dry powder inhaling appliance, or as in U.S. Pub. No. US2015/0367091A1 and references cited therein.
  • any one or more cannabinoids such as CBD and THC, as well as any entourage-enhancing cannabinoid fraction, including combinations thereof, may be replaced with a cannabis extract comprising such cannabinoids.
  • compositions of the invention will be combined with other compounds such as vitamins, antioxidants, amino acids, probiotics, natural herbs or plant extracts, and other food or dietary supplements, for various purposes such as for energy, reduced fatigue, or improved mental and physical performance (e.g., guarana, amino acids, and vitamins such as B12), for improved performance and reduced gastrointestinal discomfort during exercise (e.g., L-leucine, L-isoleucine, L-valine, Vitamin B1, and Vitamin B5), for osteoporosis or function of bones and teeth (e.g., vitamins C, D3, and K2), for sexual dysfunction, improved sexual vigor, or fertility (e.g., vitamins, amino acids, and plant extracts such as maca root, ginkgo biloba, tributes terrestris, minus pinaster, muir puma, damiana, or catuaba), for improved circulation or to sooth heavy legs (e.g., vitas vinifera extract and biof
  • a cannabis composition will contain: 1.176 grams of cannabis distillate, 0.09 grams of cherry terps, 0.22 grams of mint, and 28.514 mL of MCT oil.
  • any other active compounds e.g., the antidepressant or anxiolytic in Examples 3 and 11, or analgesic, anti-inflammatory, or antioxidants in Examples 4 and 12
  • any of the compounds could be provided in another amount, adapted to achieve the same or like effect when administered to a human.
  • substitution of the compound by its prodrug, free base, salt, or hydrochloride salt shall be understood to provide merely an alternative embodiment still within the scope of the invention.
  • compositions within the scope of the invention should be understood to be open-ended and may include additional active or inactive compounds and ingredients.
  • the type of formulation employed for the administration of the compounds employed in the methods of the present invention generally may be dictated by the compound(s) employed, the type of pharmacokinetic profile desired from the route of administration and the compound(s), and the state of the patient.
  • N. Dosage [230] Administration of a mushroom composition or a cannabis composition in a “therapeutically effective amount,” or an “effective amount” to a subject means administration of an amount of the mushroom composition or the cannabis composition sufficient to achieve the desired effect. In embodiments, a “therapeutically effective amount,” or“ effective amount” thus would be the amount sufficient to cause a modulation of a neurotransmitter system.
  • an “effective amount” means an amount effective in treating the stated disorder or symptoms in a subject
  • therapeutic effect would be understood to mean the responses(s) in a mammal after treatment that are judged to be desirable and beneficial.
  • Mushroom compositions disclosed herein may comprise therapeutic amounts of any of psilocybin and other compounds from Psilocybe or other psilocybin-containing mushrooms, such as other psychedelic or psychoactive tryptamines (“minor tryptamines”), L-theanine, niacin, and compounds from lion’s mane mushrooms and cordyceps mushrooms, and optionally valerian (or other plant extracts or compounds) and/or vitamin B12 (or other vitamins), optionally a CB 1 receptor modulator (such as CBD or PEA), and in some embodiments, other active ingredients which may include minerals, amino acids, proteins, and dietary or nutritional supplements.
  • other active ingredients which may include minerals, amino acids, proteins, and dietary or nutritional supplements.
  • a mushroom composition where it contains psilocybin and other compounds from Psilocybe or other psilocybin-containing mushrooms, and/or compounds from lion’s mane mushrooms and cordyceps mushrooms, will comprise ground or pulverized fungal matter (i.e., fungal matter from Psilocybe or other psilocybin-containing mushrooms, lion’s mane mushrooms, and/or cordyceps mushrooms), one or more fungal extracts from any of the foregoing, and/or one or more isolated or purified fungal compounds therefrom.
  • ground or pulverized fungal matter i.e., fungal matter from Psilocybe or other psilocybin-containing mushrooms, lion’s mane mushrooms, and/or cordyceps mushrooms
  • a mushroom composition comprises ground or pulverized fungal matter, which may be fungal matter from one or more fungal species, and may further comprise one or more carriers, diluents, or excipients.
  • a mushroom composition comprises ground or pulverized fungal matter, including where a composition preferably comprises ground or pulverized dried fungal matter, such as fungal matter from a Psilocybe or other psilocybin-producing species, from lion’s mane, or cordyceps, it may be present in an amount so that a single dose is (whether or not such dose is present in a unit dosage form), e.g., 10 mg or less, at least 20 mg, at least 30 mg, at least 40 mg, at least 50 mg, at least 100 mg, at least 125 mg, at least 150 mg, at least 175 mg, at least 200 mg, at least 225 mg, at least 250 mg, at least 275 mg, at least 300 mg, at least 350
  • a mushroom composition comprises an extract from a psilocybin-containing mushroom, truffle, mycelial body, or other fungal material, such as from a Psilocybe or other psilocybin-producing species
  • it may be present in an amount so that a single dose is (whether or not such dose is present in a unit dosage form), e.g., 10 mg or less, at least 20 mg, at least 30 mg, at least 40 mg, at least 50 mg, at least 100 mg, at least 125 mg, at least 150 mg, at least 175 mg, at least 200 mg, at least 225 mg, at least 250 mg, at least 275 mg, at least 300 mg, at least 350 mg, at least 400 mg, at least 450 mg, or at least 500 mg.
  • a mushroom composition comprises purified, synthetic, or biosynthetic psilocybin
  • it may be present in an amount so that a single dose is (whether or not such dose is present in a unit dosage form), e.g., .01 mg or less, at least .05 mg, at least .1 mg, at least .15 mg, at least .2 mg, at least .3 mg, at least .4 mg, at least .5 mg, at least 1 mg, at least 1.5 mg, at least 2 mg, at least 2.5 mg, at least 3 mg, at least 3.5 mg, at least 4 mg, at least 5 mg, at least 6 mg, at least 7 mg, at least 8 mg, at least 9 mg, or at least 10 mg.
  • the compositions and formulations will be without psychedelic effect when taken by a human.
  • dosages shall be below the threshold at which human subjects report the presence of such psychedelic effects (i.e., “microdoses”). Because such dosages may differ from subject to subject, an individual subject will be able to take a smaller dosage, if desired, and unit dosage forms at various dosage levels can be offered to provide optimal results across the whole of the dose-response curve, for differently responding individuals in a population.
  • dose ranges under about 2.0-3.0 mg isolated or pure psilocybin are considered sub-psychedelic dosages, and this amount can be converted based on the psilocybin concentration in a whole fungal preparation to determine reasonable and appropriate starting dosages, which then can be adjusted upwards or downwards based on subjective effects, using known methods.
  • Therapeutic benefits may be found optimal in a range of about 0.5 to about 2.0 mg psilocybin, and some preferred embodiments contain purified, synthetic, or biosynthetic psilocybin in such amounts, or in approximately equivalent amounts. For example, a P.
  • cubensis strain with an approximate w/w% of psilocybin of 0.63% may be used in a composition of the invention as an equivalent extract by weight of between about 75 to about 325 mg. Amounts of between 100 to 275 mg may be preferred. Likewise, a 10x purified extract from the same strain may be used in an amount of between about 7.5 to about 32.5 mg. It will be appreciated that similar calculations may be made for other extracts, such as cordyceps, lion’s mane, and valerian root, based on the same principle, when using purified extracts or isolated compounds.
  • a mushroom composition comprises lion’s mane extract
  • it may be present in an amount so that a single dose is (whether or not such dose is present in a unit dosage form), e.g., 10 mg or less, at least 20 mg, at least 30 mg, at least 40 mg, at least 50 mg, at least 100 mg, at least 150 mg, at least 200 mg, at least 250 mg, at least 300 mg, at least 350 mg, at least 400 mg, at least 450 mg, at least 500 mg, at least 550 mg, at least 600 mg, at least 650 mg, at least 700 mg, at least 750 mg, at least 800 mg, at least 850 mg, at least 900 mg, at least 950 mg, or at least 1000 mg.
  • a mushroom composition comprises a purified, synthetic, or biosynthetic compound from lion’s mane, such as a hericenone extracted from or extractable from Hericium erinaceum , and including any of Hericenone A, Hericenone B, Hericenone C, Hericenone D, Hericenone E, Hericenone F, Hericenone G, Hericenone H, Hericenone I, Hericenone J, Hericenone K, 3HF, DLPE, Isohericerinol A, Hericerin, NDPIH, and Corallocin A
  • it may be present in an amount so that a single dose is (whether or not such dose is present in a unit dosage form), e.g., .01 mg or less, at least .05 mg, at least .1 mg, at least .15 mg, at least .2 mg, at least .3 mg, at least .4 mg, at least .5 mg, at least 1 mg
  • a mushroom composition comprises cordyceps extract
  • it may be present in an amount so that a single dose is (whether or not such dose is present in a unit dosage form), e.g., 10 mg or less, at least 20 mg, at least 30 mg, at least 40 mg, at least 50 mg, at least 100 mg, at least 150 mg, at least 200 mg, at least 250 mg, at least 300 mg, at least 350 mg, at least 400 mg, at least 450 mg, at least 500 mg, at least 550 mg, at least 600 mg, at least 650 mg, at least 700 mg, at least 750 mg, at least 800 mg, at least 850 mg, at least 900 mg, at least 950 mg, or at least 1000 mg.
  • a mushroom composition comprises a purified, synthetic, or biosynthetic compound from cordyceps, such as cordycepin, cordycepic acid, N-acetylgalactosamine, adenosine, ergosterol, ergosteryl esters, bioxanthracenes, hypoxanthine, acid deoxyribonuclease, polysaccharide, exopolysaccharide, chitinase, macrolides, cicadapeptins, myriocin, superoxide dismutase, protease, naphthoquinone, cordyheptapeptide, dipicolinic acid, fibrinolytical enzyme, lectin, and cordymin, it may be present in an amount so that a single dose is (whether or not such dose is present in a unit dosage form), e.g., .01 mg or less, at least .05 mg, at least .1 mg
  • cordyceps such
  • a mushroom composition comprises L-theanine
  • it may be present in an amount so that a single dose is (whether or not such dose is present in a unit dosage form), e.g., 10 mg or less, at least 20 mg, at least 30 mg, at least 40 mg, at least 50 mg, at least 100 mg, at least 150 mg, at least 200 mg, at least 250 mg, at least 300 mg, at least 350 mg, at least 400 mg, at least 450 mg, at least 500 mg, at least 550 mg, at least 600 mg, at least 650 mg, at least 700 mg, at least 750 mg, at least 800 mg, at least 850 mg, at least 900 mg, at least 950 mg, or at least 1000 mg.
  • a mushroom composition comprises another amino acid or amino acid analog, at dose amounts known in the art.
  • a mushroom composition comprises niacin
  • it may be present in an amount so that a single dose is (whether or not such dose is present in a unit dosage form), e.g., 10 mg or less, at least 20 mg, at least 30 mg, at least 40 mg, at least 50 mg, at least 100 mg, at least 150 mg, at least 200 mg, at least 250 mg, at least 300 mg, at least 350 mg, at least 400 mg, at least 450 mg, at least 500 mg, at least 550 mg, at least 600 mg, at least 650 mg, at least 700 mg, at least 750 mg, at least 800 mg, at least 850 mg, at least 900 mg, at least 950 mg, or at least 1000 mg.
  • a mushroom composition comprises another vitamin, at dose amounts known in the art.
  • a mushroom composition comprises valerian root extract, it may be present in an amount so that a single dose is (whether or not such dose is present in a unit dosage form), e.g., 10 mg or less, at least 20 mg, at least 30 mg, at least 40 mg, at least 50 mg, at least 100 mg, at least 150 mg, at least 200 mg, at least 250 mg, at least 300 mg, at least 350 mg, at least 400 mg, at least 450 mg, at least 500 mg, at least 550 mg, at least 600 mg, at least 650 mg, at least 700 mg, at least 750 mg, at least 800 mg, at least 850 mg, at least 900 mg, at least 950 mg, or at least 1000 mg.
  • a mushroom composition comprises another plant extract or compound, at dose amounts known in the art.
  • a mushroom composition comprises Vitamin B12
  • it may be present in an amount so that a single dose is (whether or not such dose is present in a unit dosage form), e.g., 1 mcg or less, at least 2 mcg, at least 3 mcg, at least 4 mcg, at least 5 mcg, at least 10 mcg, at least 15 mcg, at least 20 mcg, at least 25 mcg, at least 30 mcg, at least 35 mcg, at least 40 mcg, at least 45 mcg, at least 50 mcg, at least 55 mcg, at least 60 mcg, at least 65 mcg, at least 70 mcg, at least 75 mcg, at least 80 mcg, at least 85 mcg, at least 90 mcg, at least 95
  • a mushroom composition comprises another vitamin, at dose amounts known in the art.
  • a mushroom composition comprises a CB 1 receptor modulator such as CBD or PEA
  • it may be present in an amount so that a single dose is (whether or not such dose is present in a unit dosage form), e.g., 1 mg or less, at least 2 mg, at least 3 mg, at least 4 mg, at least 5 mg, at least 10 mg, at least 15 mg, at least 20 mg, at least 25 mg, at least 30 mg, at least 35 mg, at least 40 mg, at least 45 mg, at least 50 mg, at least 55 mg, at least 60 mg, at least 65 mg, at least 70 mg, at least 75 mg, at least 80 mg, at least 85 mg, at least 90 mg, at least 95 mg, or at least 100 mg.
  • a mushroom composition comprises another CB 1 receptor modulator, at dose amounts known in the art.
  • Cannabis compositions disclosed herein may comprise therapeutic amounts of any of THC, CBD, and other psychoactive or non-psychoactive cannabinoids (besides THC and CBD, referred to “minor cannabinoids”), isoprenoids or terpenes, and in some embodiments, flavonoids and/or other active ingredients, such as vitamins, minerals, natural and unnatural (analog) amino acids (e.g., L-theanine), proteins, and dietary or nutritional supplements.
  • a cannabis composition comprises a cannabis extract, and may further comprise one or more carriers, diluents, or excipients.
  • a cannabis extract may be, for example, a cannabis full spectrum extract, a cannabis broad spectrum extract, a cannabis oil, a cannabis distillate, or a cannabis isolate, as such terms will be readily understood by those of skill.
  • a cannabis extract may be any of kief, hashish, bubble hash, solvent reduced oils, sludges, e-juice, and tinctures.
  • a cannabis composition comprises CBD
  • it may be present in an amount so that a single dose is (whether or not such dose is present in a unit dosage form), e.g., 5 mg or less, at least 5 mg, at least 10 mg, at least 15 mg, at least 20 mg, at least 25 mg, at least 30 mg, at least 35 mg, at least 40 mg, at least 45 mg, at least 50 mg, at least 55 mg, at least 60 mg, at least 65 mg, at least 70 mg, at least 75 mg, at least 80 mg, at least 85 mg, at least 90 mg, at least 95 mg, or at least 100 mg.
  • a single dose e.g., 5 mg or less, at least 5 mg, at least 10 mg, at least 15 mg, at least 20 mg, at least 25 mg, at least 30 mg, at least 35 mg, at least 40 mg, at least 45 mg, at least 50 mg, at least 55 mg, at least 60 mg, at least 65 mg, at least 70 mg, at least 75 mg, at least 80 mg, at least 85 mg, at least 90
  • CBD may be present in an amount of 5 mg/mL or less, at least 5 mg/mL, at least 10 mg/mL, at least 15 mg/mL, at least 20 mg/mL, at least 25 mg/mL, at least 30 mg/mL, at least 35 mg/mL, at least 40 mg/mL, at least 45 mg/mL, at least 50 mg/mL, at least 55 mg/mL, at least 60 mg/mL, at least 65 mg/mL, at least 70 mg/mL, at least 75 mg/mL, at least 80 mg/mL, at least 85 mg/mL, at least 90 mg/mL, at least 95 mg/mL, or at least 100 mg/mL.
  • a cannabis composition comprises THC
  • it may be present in an amount so that a single dose is (whether or not present in a unit dosage form), e.g., 1 mg or less, at least 2 mg, at least 3 mg, at least 4 mg, at least 5 mg, at least 10 mg, at least 15 mg, at least 20 mg, at least 25 mg, at least 30 mg, at least 35 mg, at least 40 mg, at least 45 mg, or at least 50 mg.
  • THC may be present in an amount of 1 mg/mL or less, at least 2 mg/mL, at least 3 mg/mL, at least 4 mg/mL, at least 5 mg/mL, at least 10 mg/mL, at least 15 mg/mL, at least 20 mg/mL, at least 25 mg/mL, at least 30 mg/mL, at least 35 mg/mL, at least 40 mg/mL, at least 45 mg/mL, or at least 50 mg/mL.
  • the ratio of THC:CBD is between 1:20 to 1:10, between 1:10 and 1:5, between 1:5 and 1:2, between 1:2 and about 1:1, but preferably about 1:4; in other embodiments it is between about 1:1 and 2:1, between 2:1 and 5:1, between 5:1 and 10:1, and between 10:1 and 20:1.
  • the entourage-enhancing cannabinoid fraction is contemplated as consisting of a lower amount (by % w/w) than the amounts of CBD and THC (or their synthetic counterparts, analogs, metabolites, prodrugs, salts, and the like).
  • the entourage-enhancing cannabinoid fraction comprises from about 0.1% to about 25% of the total amount of cannabinoids and isoprenoids (and flavonoids, when present) by weight, and preferably from about 1-15% w/w, about 2.5-12.5% w/w, or about 5-10% w/w.
  • the individual cannabinoids of the entourage-enhancing cannabinoid fraction shall be in relatively equal proportions, but it is contemplated that such cannabinoids may be in unequal proportions, and particularly contemplated that certain cannabinoids may be dominant with others only present in trace amounts, especially where a cannabinoid fraction contains three or more cannabinoids.
  • Terpenes are generally considered to be pharmacologically relevant in whole plant material when present in concentrations of at least 0.05% (Hazekamp and Fischedick 2010.
  • isoprenoid fractions are preferably present in an amount of about 1% w/w of the total amount of cannabinoids and isoprenoids (and flavonoids, when present).
  • the isoprenoid may be ⁇ 0.1% w/w, 0.1-0.5 % w/w, 0.5-1 % w/w, 1-1.5% w/w, 1.5-2.5% w/w, 2.5-5% w/w, 5-10% w/w, or greater than 10% w/w.
  • the individual isoprenoids of the entourage-enhancing isoprenoid fraction shall be in relatively equal proportions, but it is contemplated that such isoprenoids may be in unequal proportions, and particularly contemplated that certain isoprenoids may be dominant with others only present in trace amounts, especially where an isoprenoid fraction contains three or more isoprenoids.
  • the entourage-enhancing isoprenoid fraction contains from about 19.9-29.9% myrcene by weight, and about 4.2-6.3% beta-caryophyllene, 2.8-4.2% and gamma-terpinene, 12.2-18.3% limonene, 13.0-19.5% terpinolene, 8.6-12.9% ocimene, 4.5-6.8% beta-pinene, 2.7-4.0% alpha-pinene, and about 12.0-18.0% other terpenes, where no such other terpenes are in an amount of 2% w/w or greater.
  • the composition comprises an amount of myrcene by w/w of at least 1.5x, at least 2x, at least 3x, at least 4x, at least 5x, or more than 5x the amount of the terpene that is in the next greatest amount.
  • the dominant terpene may be by w/w at least 1.5x, at least 2x, at least 3x, at least 4x, at least 5x, or more than 5x the amount of the terpene that is in the next greatest amount.
  • Such embodiments may be referred to as “1.5x dominant” of the terpene, or “2x dominant,” “3x dominant,” “4x dominant,” and the like.
  • Dose amount, frequency or duration may be increased or reduced, as indicated by the clinical outcome desired, status of the infection, reactivation, pathology or symptom, or any adverse side effects of the treatment or therapy.
  • the skilled artisan will appreciate the factors that may influence the dosage, frequency, and timing required to provide an amount sufficient or effective for providing a prophylactic or therapeutic effect or benefit.
  • the dose actually administered will be determined by a physician, in light of the relevant circumstances, including the disorder to be treated, the chosen route of administration, the actual composition administered, the age, weight, and response of the individual patient, and the severity of the patient’s symptoms, and therefore any dosage ranges disclosed herein are not intended to limit the scope of the invention.
  • compositions of the invention may be administered and dosed in accordance with good medical practice, taking into account the method and scheduling of administration, prior and concomitant medications and medical supplements, the clinical condition of the individual patient and the severity of the underlying disease, the patient’s age, sex, body weight, and other such factors relevant to medical practitioners, and knowledge of the particular compound(s) used.
  • Starting and maintenance dosage levels thus may differ from patient to patient, for individual patients across time, and for different pharmaceutical compositions, but shall be able to be determined with ordinary skill.
  • appropriate dosages to achieve a therapeutic effect can be determined by an individual by reference to available public information and knowledge, and by reference to subjective considerations regarding desired outcomes and effects (including, in some embodiments, a desired absence of psychedelic effects).
  • a patient will be prescribed, administered, or will obtain without a prescription, a therapeutically effective dose of the compositions of the invention on a regular or “chronic” basis, wherein the patient is administered the compositions of the invention daily, several times per day (at least one, at least two, at least three, at least four, or greater than four times per day); on a set, repeating schedule, wherein the patient is administered a therapeutically effective dose of the compositions of the invention every other day, every three days, every four days, every five days, every six days, every seven days, or more than every seven days; or, in some embodiments, a varying schedule comprised of a plurality of days “on” (wherein the therapeutically effective dose of the pharmaceutical composition of the invention is administered), and a plurality of days “off” (wherein no administration occurs), such as one day on two days off, two days on three days off, three days on four days off, or other such schedules as would be apparent to those of skill.
  • a varying schedule comprised of a plurality of
  • the dosage administered to, or taken by a patient in need thereof is a “macrodose” (whether or not so named), wherein the dose is a sufficient amount to cause at least one perceptible subjective effect within a patient, and preferably the full range of subjective effects desired through administration of the drug, or a desired subset thereof, as chosen through ordinary skill.
  • perceptible subjective effects are those characteristic of, or substantially characteristic of, or sharing at least one or more subjective qualities of, a psychedelic.
  • the dosage administered to, or taken by a patient in need thereof is a “microdose” (whether or not so named), wherein the dose is not perceptible by the patient to which it is administered.
  • a microdose may be 0.001 to 0.25 of a macro-dose, such as but not limited to 0.003, 0.005, 0.007, 0.009, 0.01, 0.03, 0.05, 0.07, 0.09, 0.1, 0.2, and 0.25 of a macrodose (wherein the list is inclusive, and modified by the term “about”).
  • a microdose is between about 0.001 mg and about 50 mg, including 0.001 mg, 0.005 mg, 0.01 mg, 0.05 mg, 0.1 mg, 0.5 mg, 1 mg, 1.25 mg, 1.5 mg, 1.75 mg, 2 mg, 2.25 mg, 2.5 mg, 2.75 mg, 3 mg, 3.5 mg, 4 mg, 4.5 mg, 5 mg, 5.5 mg, 6 mg, 6.5 mg, 7 mg, 7.5 mg, 8 mg, 8.5 mg, 9 mg, 9.5 mg, 10 mg, 11 mg, 12 mg, 13 mg, 14 mg, 15 mg, and values in between, wherein each value is modified by the term “about.”
  • Determination of appropriate dosing shall include not only the determination of single dosage amounts, but also determination of the number and timing of doses, e.g., administration of a particular dosage amount once per day, twice per day, or more than twice per day, and the time(s) of day preferable for administration, and any daily dosing regimen, including different and distinct alternating daily dosing regimens,
  • dosing regimens will be “daily dosing” regimens, but modification of any dosing regimen, including any daily dosing regimen herein, may be made within ordinary skill, for example that every “daily dose” be taken at 12-hour intervals (e.g., at “morning” and at “night”), rather than at 24-hour intervals, and thus, in one exemplary such modification, that 14-day therapeutic regimes will be administered during a single week. Other such modifications and alterations will be readily appreciated by those of skill. [265] In embodiments, especially where a formulation is prepared in single unit dosage form, suggested dosage amounts may be known by reference to the format of the preparation itself.
  • the method of treating a mental health condition or improving mental health comprises administering a composition according to a distinct and predetermined dosing regimen, provided for its novel and unique therapeutic effects.
  • a distinct and predetermined dosing regimen provided for its novel and unique therapeutic effects.
  • a composition will be taken on a daily basis, and a defined dosing regimen may also be referred to as a “daily dosing regimen”; conversely, as noted above, daily dosing regimens may be modified, and can also be readily embodied as “defined dosing regimens.”
  • the defined dosing regimen comprises alternating phases of administration (“dosing,” which depending on embodiments will include what is commonly referred to equivalently, for example with psilocybin-comprising products, as “microdosing”) and abstaining (“washout”).
  • the defined dosing regimen comprises alternating phases of dosing a mushroom composition and washout.
  • the defined dosing regimen comprises alternating phases of dosing an M+ mushroom composition and washout.
  • the defined dosing regimen comprises alternating phases of dosing an M– mushroom composition and washout.
  • the defined dosing regimen comprises alternating phases of dosing a cannabis composition and washout.
  • the defined dosing regimen comprises alternating phases of dosing an C+ mushroom composition and washout.
  • the defined dosing regimen comprises alternating phases of dosing an C– mushroom composition and washout.
  • the defined dosing regimen comprises alternating phases of dosing a first composition, and dosing a second composition.
  • the defined dosing regimen comprises alternating phases of dosing a first mushroom composition and dosing a second mushroom composition.
  • the defined dosing regimen comprises alternating phases of dosing a first M+ mushroom composition and dosing a second M+ mushroom composition.
  • the defined dosing regimen comprises alternating phases of dosing a first M+ mushroom composition and dosing a second M– mushroom composition.
  • the defined dosing regimen comprises alternating phases of dosing a first M– mushroom composition and dosing a second M– mushroom composition.
  • the defined dosing regimen comprises alternating phases of dosing a mushroom composition and dosing a cannabis composition.
  • the defined dosing regimen comprises alternating phases of dosing an M+ mushroom composition and dosing a cannabis composition.
  • the defined dosing regimen comprises alternating phases of dosing an M+ mushroom composition and dosing a C+ cannabis composition.
  • the defined dosing regimen comprises alternating phases of dosing an M+ mushroom composition and dosing a C– cannabis composition.
  • the defined dosing regimen comprises alternating phases of dosing an M– mushroom composition and dosing a cannabis composition.
  • the defined dosing regimen comprises alternating phases of dosing an M– mushroom composition and dosing a C+ cannabis composition. [285] In embodiments, the defined dosing regimen comprises alternating phases of dosing an M– mushroom composition and dosing a C– cannabis composition. [286] In embodiments, the defined dosing regimen comprises alternating phases of dosing a first cannabis composition and dosing a second cannabis composition. [287] In embodiments, the defined dosing regimen comprises alternating phases of dosing a first C+ cannabis composition and dosing a second C+ cannabis composition.
  • the defined dosing regimen comprises alternating phases of dosing a first C+ cannabis composition and dosing a second C– cannabis composition.
  • the defined dosing regimen comprises alternating phases of dosing a first C– cannabis composition and dosing a second C– cannabis composition.
  • the defined dosing regimen comprises three or more alternating phases of dosing and/or washout, including three alternating phases, four alternating phases, five alternating phases, six alternating phases, seven alternating phases, eight alternating phases, nine alternating phases, ten alternating phases, and more than ten alternating phases, including 11 or more, 12 or more, or greater than 12 alternating phases.
  • any or all of such alternating phases may be dosing phases or washout phases, including three or more phase of dosing, including three dosing phases, four dosing phases, five dosing phases, six dosing phases, seven dosing phases, eight dosing phases, nine dosing phases, ten dosing phases, and more than ten dosing phases, including 11 or more, 12 or more, or greater than 12 dosing phases.
  • Such dosing regimens may, in some embodiments, include one or more washout phases, including three washout phases, four washout phases, five washout phases, six washout phases, or greater than six washout phases.
  • any one or more of the defined dosing regimens above is a “daily dosing regimen,” wherein the alternating phases of dosing are on consecutive days.
  • any one or more of the defined dosing regimens above is a “twice-a-day dosing regimen,” wherein the alternating phases of dosing are at morning and at night, or at two other times per day, including predetermined times chosen by a patient or a practitioner, or chosen based on a software analysis of one or more objective indicia.
  • any one or more of the defined dosing regimens above is a “three-times-per-day dosing regimen,” wherein the alternating phases of dosing are at breakfast, lunch, and dinner, or at three other times per day, including predetermined times chosen by a patient or a practitioner, or chosen based on a software analysis of one or more objective indicia.
  • Defined dosing regimens also may be four, five, or more than five times per day.
  • Defined dosing regimens may also be on a greater than single day schedule, such as every other day, every third day, every fourth day, every fifth day, every sixth day, every week, or every number of days greater than one week.
  • Defined dosing regimens also may be on schedules not defined by an integer number of days, such as every 1.5 days, and the use of an electronic tool, mobile app, or other software app, may optionally assist with compliance.
  • the number of administrations of each phase will be independently selected, any may include from one administration to seven administrations inclusive, such as one administration, two administrations, three administrations, four administrations, five administrations, six administrations, and one week, as well as a number of administrations greater than seven administrations, such as eight administrations, nniinnee administrations, 10 administrations, 11 administrations, 12 administrations, and greater than 12 administrations.
  • administration will be understood to include an “administration” period or day of abstinence or “washout,” wherein such period or day of abstinence or washout may also include the administration of a composition that is not a mushroom or cannabis composition of the invention, but which may for example comprise additional active agents, such as vitamins, nootropics, or supplements
  • the defined dosing regimen comprises alternating phases of between 2-7 periods of administration and 2-7 periods of abstaining (washout). In embodiments, the defined dosing regimen comprises alternating phases of between 2-6 periods of administration and 2-6 periods of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 2-5 periods of administration and 2-5 periods of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 2-4 periods of administration and 2-4 periods of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 2-3 periods of administration and 2-3 periods of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 3-4 periods of administration and 3-4 periods of abstaining.
  • the defined dosing regimen comprises alternating phases of between 3-5 periods of administration and 3-5 periods of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 3-6 periods of administration and 3-6 periods of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 3-7 periods of administration and 3-7 periods of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 4-7 periods of administration and 4-7 periods of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 4-6 periods of administration and 4-6 periods of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 4-5 periods of administration and 4-5 periods of abstaining.
  • the defined dosing regimen comprises alternating phases of between 6-7 periods of administration and 6-7 periods of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 5-7 periods of administration and 5-7 periods of abstaining. In embodiments, the defined dosing regimen comprises alterating phases of between 1-7 periods of administration and 1-7 periods of abstaining.
  • the defined dosing regimen comprises alternating phases of between 2-7 periods of a first composition and 2-7 periods of a second composition. In embodiments, the defined dosing regimen comprises alternating phases of between 2-6 periods of a first composition and 2-6 periods of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 2-5 periods of a first composition and 2-5 periods of a second composition. In embodiments, the defined dosing regimen comprises alternating phases of between 2-4 periods of a first composition and 2-4 periods of a second composition. In embodiments, the defined dosing regimen comprises alternating phases of between 2-3 periods of a first composition and 2-3 periods of a second composition.
  • the defined dosing regimen comprises alternating phases of between 3-4 periods of a first composition and 3-4 periods of a second composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-5 periods of a first composition and 3-5 periods of a second composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-6 periods of a first composition and 3-6 periods of a second composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-7 periods of a first composition and 3-7 periods of a second composition. In embodiments, the defined dosing regimen comprises alternating phases of between 4-7 periods of a first composition and 4-7 periods of a second composition.
  • the defined dosing regimen comprises alternating phases of between 4-6 periods of a first composition and 4-6 periods of a second composition. In embodiments, the defined dosing regimen comprises alternating phases of between 4-5 periods of a first composition and 4-5 periods of a second composition. In embodiments, the defined dosing regimen comprises alternating phases of between 6-7 periods of a first composition and 6-7 periods of a second composition. In embodiments, the defined dosing regimen comprises alternating phases of between 5-7 periods of a first composition and 5-7 periods of a second composition. In embodiments, the defined dosing regimen comprises alternating phases of between 1-7 periods of a first composition and 1-7 periods of a second composition.
  • the defined dosing regimen comprises alternating phases of between 2-7 periods of a mushroom composition and 2-7 periods of a cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 2-6 periods of a mushroom composition and 2-6 periods of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 2-5 periods of a mushroom composition and 2-5 periods of a cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 2-4 periods of a mushroom composition and 2-4 periods of a cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 2-3 periods of a mushroom composition and 2-3 periods of a cannabis composition.
  • the defined dosing regimen comprises alternating phases of between 3-4 periods of a mushroom composition and 3-4 periods of a cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-5 periods of a mushroom composition and 3-5 periods of a cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-6 periods of a mushroom composition and 3-6 periods of a cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-7 periods of a mushroom composition and 3-7 periods of a cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 4-7 periods of a mushroom composition and 4-7 periods of a cannabis composition.
  • the defined dosing regimen comprises alternating phases of between 4-6 periods of a mushroom composition and 4-6 periods of a cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 4-5 periods of a mushroom composition and 4-5 periods of a cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 6-7 periods of a mushroom composition and 6-7 periods of a cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 5-7 periods of a mushroom composition and 5-7 periods of a cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 1-7 periods of a mushroom composition and 1-7 periods of a cannabis composition.
  • the defined dosing regimen comprises alternating phases of between 2-7 periods of a first mushroom composition and 2-7 periods of a second mushroom composition. In embodiments, the defined dosing regimen comprises alternating phases of between 2-6 periods of a first mushroom composition and 2-6 periods of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 2-5 periods of a first mushroom composition and 2-5 periods of a second mushroom composition. In embodiments, the defined dosing regimen comprises alternating phases of between 2-4 periods of a first mushroom composition and 2-4 periods of a second mushroom composition. In embodiments, the defined dosing regimen comprises alternating phases of between 2-3 periods of a first mushroom composition and 2-3 periods of a second mushroom composition.
  • the defined dosing regimen comprises alternating phases of between 3-4 periods of a first mushroom composition and 3-4 periods of a second mushroom composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-5 periods of a first mushroom composition and 3-5 periods of a second mushroom composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-6 periods of a first mushroom composition and 3-6 periods of a second mushroom composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-7 periods of a first mushroom composition and 3-7 periods of a second mushroom composition. In embodiments, the defined dosing regimen comprises alternating phases of between 4-7 periods of a first mushroom composition and 4-7 periods of a second mushroom composition.
  • the defined dosing regimen comprises alternating phases of between 4-6 periods of a first mushroom composition and 4-6 periods of a second mushroom composition. In embodiments, the defined dosing regimen comprises alternating phases of between 4-5 periods of a first mushroom composition and 4-5 periods of a second mushroom composition. In embodiments, the defined dosing regimen comprises alternating phases of between 6-7 periods of a first mushroom composition and 6-7 periods of a second mushroom composition. In embodiments, the defined dosing regimen comprises alternating phases of between 5-7 periods of a first mushroom composition and 5-7 periods of a second mushroom composition. In embodiments, the defined dosing regimen comprises alternating phases of between 1-7 periods of a first mushroom composition and 1-7 periods of a second mushroom composition.
  • the defined dosing regimen comprises alternating phases of between 2-7 periods of a first cannabis composition and 2-7 periods of a second cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 2-6 periods of a first cannabis composition and 2-6 periods of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 2-5 periods of a first cannabis composition and 2-5 periods of a second cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 2-4 periods of a first cannabis composition and 2-4 periods of a second cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 2-3 periods of a first cannabis composition and 2-3 periods of a second cannabis composition.
  • the defined dosing regimen comprises alternating phases of between 3-4 periods of a first cannabis composition and 3-4 periods of a second cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-5 periods of a first cannabis composition and 3-5 periods of a second cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-6 periods of a first cannabis composition and 3-6 periods of a second cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-7 periods of a first cannabis composition and 3-7 periods of a second cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 4-7 periods of a first cannabis composition and 4-7 periods of a second cannabis composition.
  • the defined dosing regimen comprises alternating phases of between 4-6 periods of a first cannabis composition and 4-6 periods of a second cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 4-5 periods of a first cannabis composition and 4-5 periods of a second cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 6-7 periods of a first cannabis composition and 6-7 periods of a second cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 5-7 periods of a first cannabis composition and 5-7 periods of a second cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 1-7 periods of a first cannabis composition and 1-7 periods of a second cannabis composition.
  • the number of periods of a phase may include greater than seven periods, including eight periods, nine periods, 10 periods, 11 periods, 12 periods, 13 periods, 14 periods, 15 periods, 20 periods, 30 periods, 60 periods, 90 periods, or greater than 90 periods, and any number of periods in between.
  • the “period” as above will be by definition a day, and the number of days of each phase will be independently selected, any may include from one day to seven days inclusive, such as one day, two days, three days, four days, five days, six days, and one week, as well as a number of days greater than seven days, such as eight days, nine days, 10 days, 11 days, 12 days, and greater than 12 days.
  • the defined dosing regimen comprises alternating phases of between 2-7 days of administration and 2-7 days of abstaining (washout). In embodiments, the defined dosing regimen comprises alternating phases of between 2-6 days of administration and 2-6 days of abstaining.
  • the defined dosing regimen comprises alternating phases of between 2-5 days of administration and 2-5 days of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 2-4 days of administration and 2-4 days of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 2-3 days of administration and 2-3 days of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 3-4 days of administration and 3-4 days of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 3-5 days of administration and 3-5 days of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 3-6 days of administration and 3-6 days of abstaining.
  • the defined dosing regimen comprises alternating phases of between 3-7 days of administration and 3-7 days of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 4-7 days of administration and 4-7 days of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 4-6 days of administration and 4-6 days of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 4-5 days of administration and 4-5 days of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 6-7 days of administration and 6-7 days of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 5-7 days of administration and 5-7 days of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 1-7 days of administration and 1-7 days of abstaining.
  • the defined dosing regimen comprises alternating phases of between
  • the defined dosing regimen comprises alternating phases of between 2-6 days of a first composition and 2-6 days of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 2-5 days of a first composition and 2-5 days of a second composition. In embodiments, the defined dosing regimen comprises alternating phases of between 2-4 days of a first composition and 2-4 days of a second composition. In embodiments, the defined dosing regimen comprises alternating phases of between 2-3 days of a first composition and 2-3 days of a second composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-4 days of a first composition and
  • the defined dosing regimen comprises alternating phases of between 3-5 days of a first composition and 3-5 days of a second composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-6 days of a first composition and 3-6 days of a second composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-7 days of a first composition and 3-7 days of a second composition. In embodiments, the defined dosing regimen comprises alternating phases of between 4-7 days of a first composition and 4-7 days of a second composition. In embodiments, the defined dosing regimen comprises alternating phases of between 4-6 days of a first composition and 4-6 days of a second composition.
  • the defined dosing regimen comprises alternating phases of between 4-5 days of a first composition and 4-5 days of a second composition. In embodiments, the defined dosing regimen comprises alternating phases of between 6-7 days of a first composition and 6-7 days of a second composition. In embodiments, the defined dosing regimen comprises alternating phases of between 5-7 days of a first composition and
  • the defined dosing regimen comprises alternating phases of between 1-7 days of a first composition and 1-7 days of a second composition.
  • the defined dosing regimen comprises alternating phases of between 2-7 days of a mushroom composition and 2-7 days of a cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 2-6 days of a mushroom composition and 2-6 days of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 2-5 days of a mushroom composition and
  • the defined dosing regimen comprises alternating phases of between 2-4 days of a mushroom composition and 2-4 days of a cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 2-3 days of a mushroom composition and 2-3 days of a cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-4 days of a mushroom composition and 3-4 days of a cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-5 days of a mushroom composition and 3-5 days of a cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-6 days of a mushroom composition and 3-6 days of a cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-7 days of a mushroom composition and
  • the defined dosing regimen comprises alternating phases of between 4-7 days of a mushroom composition and 4-7 days of a cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 4-6 days of a mushroom composition and 4-6 days of a cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 4-5 days of a mushroom composition and 4-5 days of a cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 6-7 days of a mushroom composition and 6-7 days of a cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 5-7 days of a mushroom composition and 5-7 days of a cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 1-7 days of a mushroom composition and
  • the defined dosing regimen comprises alternating phases of between
  • the defined dosing regimen comprises alternating phases of between 2-6 days of a first mushroom composition and 2-6 days of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 2-5 days of a first mushroom composition and 2-5 days of a second mushroom composition. In embodiments, the defined dosing regimen comprises alternating phases of between 2-4 days of a first mushroom composition and 2-4 days of a second mushroom composition. In embodiments, the defined dosing regimen comprises alternating phases of between 2-3 days of a first mushroom composition and 2-3 days of a second mushroom composition.
  • the defined dosing regimen comprises alternating phases of between 3-4 days of a first mushroom composition and 3-4 days of a second mushroom composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-5 days of a first mushroom composition and 3-5 days of a second mushroom composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-6 days of a first mushroom composition and 3-6 days of a second mushroom composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-7 days of a first mushroom composition and 3-7 days of a second mushroom composition. In embodiments, the defined dosing regimen comprises alternating phases of between 4-7 days of a first mushroom composition and 4-7 days of a second mushroom composition.
  • the defined dosing regimen comprises alternating phases of between 4-6 days of a first mushroom composition and 4-6 days of a second mushroom composition. In embodiments, the defined dosing regimen comprises alternating phases of between 4-5 days of a first mushroom composition and 4-5 days of a second mushroom composition. In embodiments, the defined dosing regimen comprises alternating phases of between 6-7 days of a first mushroom composition and 6-7 days of a second mushroom composition. In embodiments, the defined dosing regimen comprises alternating phases of between 5-7 days of a first mushroom composition and 5-7 days of a second mushroom composition. In embodiments, the defined dosing regimen comprises alternating phases of between 1-7 days of a first mushroom composition and 1-7 days of a second mushroom composition.
  • the defined dosing regimen comprises alternating phases of between 2-7 days of a first cannabis composition and 2-7 days of a second cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 2-6 days of a first cannabis composition and 2-6 days of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 2-5 days of a first cannabis composition and 2-5 days of a second cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 2-4 days of a first cannabis composition and 2-4 days of a second cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 2-3 days of a first cannabis composition and 2-3 days of a second cannabis composition.
  • the defined dosing regimen comprises alternating phases of between 3-4 days of a first cannabis composition and 3-4 days of a second cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-5 days of a first cannabis composition and 3-5 days of a second cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-6 days of a first cannabis composition and 3-6 days of a second cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-7 days of a first cannabis composition and 3-7 days of a second cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 4-7 days of a first cannabis composition and 4-7 days of a second cannabis composition.
  • the defined dosing regimen comprises alternating phases of between 4-6 days of a first cannabis composition and 4-6 days of a second cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 4-5 days of a first cannabis composition and 4-5 days of a second cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 6-7 days of a first cannabis composition and 6-7 days of a second cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 5-7 days of a first cannabis composition and 5-7 days of a second cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 1-7 days of a first cannabis composition and 1-7 days of a second cannabis composition.
  • the number of days of a phase may include greater than seven days, including eight days, nine days, 10 days, 11 days, 12 days, 13 days, two weeks, three weeks, one month, 60 days, 90 days, or greater than 90 days, and any number of days in between.
  • the defined dosing regimen is a “weekly” dosing regimen.
  • one weekly regimen comprises alternating phases of four days and three days (understood to be equivalent to alternating phases of three days and four days, wherein the phase that is considered as “first” or “second” is simply reversed, and immaterial to the ultimate practice of the embodiment).
  • an alternating dosing regimen may comprise phases of two days and five days. In other embodiments, an alternating dosing regimen may comprise phases of one day and six days, or any other combination of phases that add up to seven days, such as two days and three days and two days, or one day and four days and two days, or one day and five days and one day, and the like (including variations thereof, wherein the “order” of the phases is immaterial, as above).
  • “weekly” regimens are easier to remember and thus in some embodiments may be preferable for compliance
  • “every other day” regimens also aid in compliance, and regimens can be adjusted according to the needs of a patient, and may include other such schedules such as two on and two off, three on and three off, four on and four off, etc., or various other such schedules (two on, three off, two on, two off; three on, four off, three on, two off; two on, two off, two on, five off; etc.).
  • a longer “washout” between doses is adopted, such as one dose every third day, or one every fourth day (i.e., two or three days of abstaining in between).
  • calendar alerts and reminders can be used, as well as web-based or computer-based programs, applications, services, schedules, and the like.
  • a phase of days “abstaining” or “washout” will mean that no composition of the invention is administered.
  • a phase will comprise administration of a mushroom composition of the invention wherein psilocybin, psilocin, or their salts, prodrugs, metabolites, polymorphs, analogs, or derivatives, are not present, nor are present any other psychedelic or psychoactive tryptamines, such as minor tryptamines from a Psilocybe or other fungi, including even at dosages insufficient to have psychedelic effects (microdoses).
  • M- mushroom compositions will in some embodiments include a mushroom composition identical in all other respects but absent psilocybin or a psilocybin-containing ingredient such as a Psilocybe extract (e.g., any of the above mushroom compositions of the Examples, without psilocybin or a Psilocybe extract, but with the additional extracts and ingredients, and otherwise modified as will be readily appreciated by those of ordinary skill for general consistency of the formulation).
  • a Psilocybe extract e.g., any of the above mushroom compositions of the Examples, without psilocybin or a Psilocybe extract, but with the additional extracts and ingredients, and otherwise modified as will be readily appreciated by those of ordinary skill for general consistency of the formulation.
  • a phase will comprise administration of a cannabis composition of the invention (such as those disclosed in the Examples) that does not contain THC or an intoxicating THC derivative or analog (i.e., a THC derivative or analog capable of causing a subject to get “high” when ingested or otherwise administered at dosages used in the art) (herein, “ C– ” cannabis compositions or “ C– ” compositions).
  • a cannabis composition of the invention such as those disclosed in the Examples
  • an intoxicating THC derivative or analog i.e., a THC derivative or analog capable of causing a subject to get “high” when ingested or otherwise administered at dosages used in the art
  • the mushroom composition may or may not contain psilocybin (i.e., M+ and M– , respectively) and the cannabis composition may or may not contain THC (i.e., C+ and C– , respectively).
  • the defined dosing regimen is a dosing regimen comprising between about 1 and about 5 administrations of a mushroom composition that does contain psilocybin (including a composition of an effective amount of psilocybin alone, all other ingredients, whether active or active agents, optional), followed by 1-5 administrations of any of, or a combination thereof, a cannabis composition containing THC, a cannabis composition that does not contain THC, a mushroom composition that does not contain psilocybin, and abstaining from administration.
  • a total therapy regime is completed for between about 15 and about 45 days, inclusive. In some preferred embodiments, the therapy regime is completed for 30 days. In embodiments, the therapy regime may be repeated a second time, for a total of about 30-90 days, or for 60 days. The therapy regime may be repeated more than two times, including three times, four times, five times, six times, seven times, eight times, nine times, 10 times, and more than 10 times.
  • the defined dosing regimen is a daily dosing regimen comprising between about 1 and about 5 days of a mushroom composition that does contain psilocybin (including a composition of an effective amount of psilocybin alone, all other ingredients, whether active or active agents, optional), followed by 1-5 days of any of, or a combination thereof, a cannabis composition containing THC, a cannabis composition that does not contain THC, a mushroom composition that does not contain psilocybin, and abstaining from dosing.
  • a total therapy regime is completed for between about 30 and about 90 days, inclusive. In some preferred embodiments, the therapy regime is completed for 60 days.
  • the therapy regime may be repeated a second time, for a total of about 60-180 days, or for 120 days.
  • the therapy regime may be repeated more than two times, including three times, four times, five times, six times, seven times, eight times, nine times, 10 times, and more than 10 times.
  • the defined dosing regimen is a dosing regimen comprising an alternating schedule of administration, wherein the mushroom composition containing psilocybin is administered for a set number of administrations, followed by administration of any of a cannabis composition containing THC, a cannabis composition that does not contain THC, a mushroom composition that does not contain psilocybin, and abstaining from administration, for a set number of total administrations.
  • the set number of administrations is between about 1 to about 7 administrations.
  • a therapy regime comprises the set number of administrations.
  • the set number of administrations comprises more than one defined dosing regimen, and is part of a therapy regime, which comprises two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, 10 or more, 11 or more, 12 or more, or more than 12 defined dosing regimens, which may be all the same or may be independently selected.
  • the therapy regime comprising the defined dosing regimen is for at least 21 administrations, at least 22 administrations, at least 23 administrations, at least 24 administrations, at least 25 administrations, at least 26 administrations, at least 27 administrations, at least 28 administrations, at least 29 administrations, at least 30 administrations, at least 31 administrations, at least 32 administrations, at least 33 administrations, at least 34 administrations, at least 35 administrations, at least 36 administrations, at least 37 administrations, at least 38 administrations, at least 39 administrations, at least 40 administrations, at least 41 administrations, at least 42 administrations, at least 43 administrations, at least 44 administrations, at least 45 administrations, at least 46 administrations, at least 47 administrations, at least 48 administrations, at least 49 administrations, at least 50 administrations, at least 51 administrations, at least 52 administrations, at least 53 administrations, at least 54 administrations, at least 55 administrations, at least 56 administrations, at least 57 administrations, at least 58 administrations, at least 59
  • an “administration” may include an “administration” of “abstinence” or “washout.” “Administration” may in such embodiments be used equivalently to “period,” as used above.
  • the defined dosing regimen is a daily dosing regimen comprising an alternating schedule of daily administration, wherein the mushroom composition containing psilocybin is administered for a set number of days, followed by administration of any of a cannabis composition containing THC, a cannabis composition that does not contain THC, a mushroom composition that does not contain psilocybin, and abstaining from dosing, for a set number of total days.
  • the set number of days is between about 1 to about 7 days.
  • a therapy regime comprises the set number of days.
  • the set number of days comprises more than one daily dosing regimen, and is part of a therapy regime, which comprises two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, 10 or more, 11 or more, 12 or more, or more than 12 daily dosing regimens, which may be all the same or may be independently selected.
  • the therapy regime comprising the more than one daily dosing regimens is for at least 21 days, at least 22 days, at least 23 days, at least 24 days, at least 25 days, at least 26 days, at least 27 days, at least 28 days, at least 29 days, at least 30 days, at least 31 days, at least 32 days, at least 33 days, at least 34 days, at least 35 days, at least 36 days, at least 37 days, at least 38 days, at least 39 days, at least 40 days, at least 41 days, at least 42 days, at least 43 days, at least 44 days, at least 45 days, at least 46 days, at least 47 days, at least 48 days, at least 49 days, at least 50 days, at least 51 days, at least 52 days, at least 53 days, at least 54 days, at least 55 days, at least 56 days, at least 57 days, at least 58 days, at least 59 days, at least 60 days, at least 61 days, at least 62 days, at least 63 days, at least 64 days, at least 65 days, at least 60 days, at
  • a mushroom composition containing psilocybin is administered for 4 consecutive days.
  • the 4 consecutive days of administration of the mushroom composition containing psilocybin is followed and/or preceded by any of, or a combination thereof, administration of a cannabis composition containing THC, a cannabis composition that does not contain THC, a mushroom composition that does not contain psilocybin; or abstinence from dosing for 3 consecutive days.
  • the therapeutic regime may continue for between about 45 days to about 90 days inclusive, including about 60 days.
  • a mushroom composition of the invention containing psilocybin may be administered consecutively for for one, two, three, four, five, six, or seven days; may be administered for one day followed by one, two, three, four, five, six, or seven days of a M–, C+, C– composition or of abstaining; may be administered for two days followed by one, two, three, four, five, six, or seven days a M–, C+, C– composition or of abstaining; may be administered for three days followed by one, two, three, four, five, six, or seven days a M–, C+, C– composition or of abstaining; may be administered for four days followed by one, two, three, four, five, six, or seven days a M–, C+, C– composition or of abstaining; may be administered for five days followed by one, two, three, four, five, six, or seven days a M–, C+, C– composition or of abstaining; may be administered for five days followed by one
  • a mushroom composition for instance as in EXAMPLES 1-7 (either as M+ or as M– compositions), can be alternated with doses containing a cannabis composition, for instance as in EXAMPLES 8-19 (either as C+ or as C– compositions), or alternated with days abstaining (washout), including where days abstaining are of a composition that is not an M or a C composition, but comprises other active ingredients.
  • a psychedelic mushroom composition e.g., a composition comprising psilocybin and/or psilocin
  • a cannabis composition e.g., a composition comprising a cannabinoid and/or a terpene
  • such compositions are not administered sequentially because one or more washout days are in between a day administering a composition comprising psilocybin and/or psilocin, and a day administering a composition comprising a cannabinoid and/or a terpene.
  • compositions are not administered sequentially because one or more days of administering an M– or functional mushroom composition (i.e., not comprising psilocybin and/or psilocin) are in between a day administering a composition comprising psilocybin and/or psilocin, and a day administering a composition comprising a cannabinoid and/or a terpene.
  • compositions are not administered sequentially because one or more administrations of an M– or functional mushroom composition (i.e., not comprising psilocybin and/or psilocin) are in between administrations of a composition comprising psilocybin and/or psilocin, and administrations of a composition comprising a cannabinoid and/or a terpene.
  • an M– or functional mushroom composition i.e., not comprising psilocybin and/or psilocin
  • administrations of a composition comprising a cannabinoid and/or a terpene are in between administrations of a composition comprising psilocybin and/or psilocin, and administrations of a composition comprising a cannabinoid and/or a terpene.
  • kits containing the mushroom compositions and/or cannabis compositions of the invention, suggested administration guidelines or prescribing information (if necessary, if by prescription) for the composition, and a suitable container.
  • Individual unit dosage forms can be included in multi-dose kits or containers.
  • Mushroom and cannabis compositions also can be packaged in single or multiple unit dosage forms for ease of administration and uniformity of dosage.
  • kits may be utilized to great advantage to assist with compliance, e.g., by using marked blister packs.
  • kits may be provided to patients or individuals with or without a prescription, including sold OTC, online, at “dispensaries,” and in one example, offered for “take home” purchase by a state legal psilocybin service center. “Pharmaceutical kits” is equivalent to and includes “defined dosing” and “daily dosing” kits.
  • a daily dosing kit or “daily dosing kit” (the terms having equivalent meaning, except for the suggestion, in some embodiments, or when needed for technical effect, that a daily dosing kit be used for “daily” administration, which a “defined” dosing kit may be used for administration on any schedule, including but not limited to daily) capsules, tablets, caplets, or other unit dosage forms are packaged in blister packs.
  • “Blister pack” may refer to any of several types of pre-formed containers, especially with plastic packaging, that contain separate receptacles (e.g., cavities or pockets) for single unit doses, where the receptacles are individually sealed and opened individually.
  • Blister packs thus include such pharmaceutical blister packs known to those of ordinary skill, including Aclar® Rx160, Rx20e, SupRx, and UltRx 2000, 3000, 4000, and 6000 (Honeywell).
  • multi-dose containers and also often referred to as blister packs, are blister trays, blister cards, strip packs, push-through packs, and the like.
  • dosing kits can be used to keep track of defined dosing regimens.
  • daily dosing kits can be used to keep track of daily dosing regimens.
  • a “compliance pack” or a “calendar pack” is created by printing the dose numbers or the days of the week above each dose.
  • the printing may comprise “day” and “night,” or “breakfast,” “lunch,” and “dinner,” or any like appropriate predetermined dosing times.
  • a number of compliance or calendar packs, containers, and kits can be utilized, including labeled blister packs, packages having indicators for each day, and dial dispenser containers.
  • information pertaining to dosing and proper administration is printed on a multi-dose kit directly (e.g., on a blister pack or other interior packaging holding the compositions of the invention).
  • kits can further contain package inserts and other printed instructions (e.g., on exterior packaging) for administering the compositions of the invention and for their appropriate therapeutic use.
  • the defined and daily dosing kits containing the compositions of the invention are designed to facilitate adherence to the defined and daily dosing regimens and therapeutic regimes described herein.
  • the kits are designed for daily oral administration over 7-day, 14-day, 21-day, 28-day, 30-day, 45-day, 60-day, 75-day, or 90 day cycle, wherein the mushroom composition and cannabis composition are packaged such that their consumption in the prescribed order is facilitated.
  • Daily dosing kits can comprise the same period daily dosing regimen repeated a set number of times (e.g., a 7-day daily dosing regimen, repeated two times, three times, four times, five times, six times, seven times, eight times, nine times, 10 times, 11 times, 12 times, or greater than 12 times; or a shorter or longer daily dosing regimen, with less or more days).
  • defined and daily dosing kits will comprise different dosing regimens repeated a number of times, in a distinct pattern, where together the pattern of dosing regimens comprises a therapeutic regime.
  • a kit of FIG.1 can be viewed as showing a single 7-day daily dosing regimen ( MMMMCCC ) three times (although it will be readily appreciated that in some embodiments, each “M” and “C” may be selected independently, and need not all be the same M and C composition.
  • the kit of FIG.2 can be viewed as showing different 7-day daily dosing regimens ( MMMMACA , MMMMCAC , MAMAACA ), which may be followed in order, comprising a 21-day therapeutic regime. Distinct or daily dosing regimens, and distinct or daily dosing kits, can be further combined to create longer therapeutic regimes, e.g., the daily dosing kit of FIG.1 and/or FIG.
  • FIG.1 illustrates an exemplary daily dosing kit ( 1901 ) containing a 21-day supply of the compositions of the invention, according to one embodiment.
  • the daily dosing kit ( 1901 ) contains a total of 21 doses of the mushroom composition ( 1903 ) of EXAMPLE 2 or EXAMPLE 3 , and the cannabis composition ( 1904 ) of EXAMPLE 9 , as well as an indicator that alerts the patient to when each dose should be administered ( 1902 ), i.e., on what day of a seven day cycle (according to this embodiment).
  • this is an exemplary embodiment, and not to be construed as limiting the invention.
  • the daily dosing kit containing doses of the compositions of the invention may contain any conceivable dosing schedule (i.e., regimen) involving any such mushroom and/or cannabis composition, including those that do or do not contain psilocybin and THC, respectively.
  • the daily dosing kit may additionally contain periodic “abstinent” or “washout” days, wherein a patient does not take any of the compositions of the invention, as is further illustrated in FIG.2.
  • FIG.2 illustrates an exemplary daily dosing kit ( 2001 ) according to an embodiment of the invention comprising the administration on given days ( 2002 ) a mushroom composition, which may or may not contain psilocybin ( 2003 ), i.e., where 2003 may be either “M+” or “M–”; and the administration of a cannabis composition, which may or may not contain THC ( 2005 ), i.e., where 2005 may be either “C+” or “C–”.
  • FIG.2 additionally includes a number of “abstinent” days (e.g., 2004 ), wherein the individual is not administered (either by the individual or a third party) any of the mushroom or cannabis compositions disclosed herein.
  • an abstinent (“A”) day when part of a daily dosing kit, may be a “blank” space or container, e.g., an empty blister where a blister pack is used as the daily dosing kit.
  • an “A” day may be a “blank” pill or composition, e.g., a placebo or “sugar pill,” as may be contained within kits comprising birth control pills.
  • an “A” day may be a pill that is not any mushroom composition or cannabis composition of the invention, but comprises one or more bioactive compounds, such as vitamins, nootropics, dietary supplements, and the like.
  • the M and C compositions therefore are not administered sequentially. In embodiments, as shown in FIG.2 , the M+ and C compositions therefore are not administered sequentially. In embodiments, as shown in FIG.2 , the M+ and C+ compositions therefore are not administered sequentially. In embodiments, as shown in FIG. 2 , the M+ and C– compositions therefore are not administered sequentially. In embodiments, as shown in FIG.2 , M+ and C compositions are administered with at least or more of a M– day or an A day in between.
  • a therapeutic regime including any one or more distinct and/or daily dosing regimens which comprise it, may be tailored to an individual patient, for example based on any number of patient-selected, prescriber-selected, or other patient-related information, such as based on one or more “objective health measurements” (including “objective indicia of improvement”), the specific indication being treated, and the like.
  • a “distinct dosing kit” may contain a dosing schedule requiring the administration of a mushroom composition for one administration, or two, three, four, five, six, or seven consecutive administrations; with one day, or two, three, four, five, six, or seven consecutive administrations of a cannabis composition; or one day, or two, three, four, five, six, or seven consecutive dosing periods of abstinence separating the mushroom administrations and/or cannabis administrations; or a combination thereof (wherein the mushroom composition may or may not contain psilocybin, and the cannabis composition may or may not contain THC).
  • the daily or distinct dosing kit may facilitate the administration of a mushroom composition containing psilocybin; followed by any of administration of a mushroom composition containing psilocybin, administration of a mushroom composition that does not contain psilocybin, administration of a cannabis composition containing THC, administration of a cannabis composition that does not contain THC, or abstinence, wherein no administration occurs.
  • the pattern may continue until the duration of the treatment program ends, or may fluctuate according to the specific patient’s needs, or the indication being treated.
  • a “daily dosing kit” may contain a dosing schedule requiring the administration of a mushroom composition for one day, or two, three, four, five, six, or seven consecutive days; with one day, or two, three, four, five, six, or seven consecutive days of administration of a cannabis composition; or one day, or two, three, four, five, six, or seven consecutive days of abstinence separating the mushroom dosing days and/or cannabis dosing days; or a combination thereof (wherein the mushroom composition may or may not contain psilocybin, and the cannabis composition may or may not contain THC). [341] In embodiments, as illustrated in FIG.
  • the non-psilocybin (M–) mushroom administration days are not scheduled abstinent, mushroom, or cannabis composition dosing days, but may fluctuate depending on patient needs.
  • the daily dosing kit ( 2101 ) includes four consecutive days ( 2103 ) of administration of the mushroom composition of EXAMPLE 2 or EXAMPLE 3 , illustrated as “ M+ ,” which indicates it contains psilocybin or another psychedelic tryptamine ( 2103 ); followed by three consecutive days of any of the administration of a cannabis composition containing THC or another intoxicating cannabinoid (including but not limited to that of EXAMPLE 9 ), illustrated as “ C+ ;” administration of a cannabis composition that does not contain THC or another intoxicating cannabinoid, illustrated as “ C– ;” administration of a mushroom composition that does not contain psilocybin or another psychedelic tryptamine, illustrated as “ M– ;” or abstinence (washout
  • the patient may be in communication with a practitioner who may evaluate the patient based on monitoring of the “objective health measurements” disclosed infra , or via information obtained from the patient regarding their subjective feelings via questionnaires, patient interviews, journaling, or any other such measures known to those of skill. Based on said information gathered, the clinician would then instruct the patient to either abstain from dosing, or be administered (either by the patient or a third party) the appropriate substance. Likewise, the patient may decide, based on how the patient personally feels, whether to abstain from dosing, or take one of the cannabis compositions, or a mushroom composition that does not contain psilocybin.
  • daily dosing kits may comprise all options (e.g., a patient may select between M+, M–, C+, C–, and A, day by day).
  • a daily dosing kit may be mailed to a patient or readied for pickup by a patient depending on prior communication to the provider of the kit of the patient needs, goals, results, data, biometrics, digital biomarkers, and the like, in advance of the start of the daily dosing regimen in the kit.
  • a clinician may instruct the patient to be administered (either by the patient or a third party, which may or may not be the clinician) more than one of the compositions on a given day.
  • the patient may be administered (by the patient or a third party) at least one of a mushroom composition that does not contain psilocybin ( M– ), a cannabis composition that contains THC ( C+ ), and a cannabis composition that does not contain THC ( C– ).
  • the patient may be administered (by the patient or a third party), any two of a mushroom composition that does not contain psilocybin ( M– ), a cannabis composition that contains THC ( C+ ), and a cannabis composition that does not contain THC ( C– ); or all three of a mushroom composition that does not contain psilocybin ( M– ), a cannabis composition that contains THC ( C+ ), and a cannabis composition that does not contain THC ( C– ).
  • a clinician may instruct a patient to be administered (either by the patient or a third party, which may or may not be the clinician) more than one of the compositions on a given day, including on days wherein a composition containing psilocybin is being administered.
  • a patient may be administered (either by the patient or a third party) a mushroom composition containing psilocybin ( M+ ), a mushroom composition not containing psilocybin ( M– ), a cannabis composition containing THC ( C+ ), and a cannabis composition not containing THC ( C– ).
  • administration may be in any order preferred by the patient and/or clinician.
  • a patient also may abstain from dosing, such that no compounds are being administered.
  • a patient will have the option of using online software such as a website, or downloadable software such as a mobile application, to assist with compliance or to collect or provide data relating to treatment.
  • Such software can be used to, e.g., keep track of last dose taken and total doses taken, provide reminders and alerts for upcoming doses, provide feedback to discourage taking doses outside of set schedules, and allow for recording of specific subjective effects, to collect or analyze one or more biometrics or digital biomarkers, or to provide means for subject directed activities such as unstructured journaling.
  • Such data collection can assist with individual patient compliance, can be used to improve or tailor individual patient care plans, and can be anonymized, aggregated, and analyzed (including by AI or natural language processing means) to allow research into the effects of various methods of treatment.
  • Q. Methods for Modulating Neurotransmission and Other Biological Systems [347] Methods are provided for using therapeutically effective amounts of the mushroom compositions of the invention in a mammal, and preferably a human. Such methods include those for modulating neurotransmission, such as serotonergic neurotransmission, using the described and claimed mushroom compositions. Methods are provided for using therapeutically effective amounts of the cannabis compositions of the invention in a mammal, and preferably a human.
  • Such methods include those for modulating endocannabinoid system activity, for modulating neurosteroid biosynthesis, and for modulating neurotransmission, using the described and claimed cannabis compositions.
  • methods are provided for using therapeutically effective amounts of the mushroom compositions and the cannabis compositions in a mammal, and preferably a human, in defined dosing regimens, daily dosing regimens, and/or therapeutic regimes, as taught herein.
  • the mushroom compositions of the invention are useful in methods for modulating neurotransmission, and such modulation will result in treatment of a mental health condition or improvement of psychological functioning.
  • compositions of the invention that modulate serotonergic neurotransmission are effective for treatment of CNS disorders such as PTSD, and other disorders correlated with dysfunction of serotonergic neurotransmission, such as feeding disturbances (annorexia nervosa, bulimia, binge eating), depression and suicide, impulsivity and violence, anxiety and harm avoidance, obsessive-compulsive features, addiction, and disturbances in neuroendocrine and vascular tissues, as well as related to other neurochemical systems linked to serotonin (Brewerton, 1995).
  • feeding disturbances annorexia nervosa, bulimia, binge eating
  • depression and suicide depression and suicide
  • impulsivity and violence anxiety and harm avoidance
  • obsessive-compulsive features addiction, and disturbances in neuroendocrine and vascular tissues
  • the invention is not limited to disorders characterized by serotonin dysregulation.
  • CNS disorders with other causes can be effectively treated by the compositions of the invention, as demonstrated through the teachings herein.
  • mushroom compositions of the invention are used to modulate serotonergic neurotransmission, and will be effective to treat disorders and conditions associated with such serotonergic modulation, or that can be treated thereby.
  • the methods of the invention that comprise the administration of a mushroom composition will therefore find use in such modulation.
  • the cannabis compositions of the invention are useful in methods for modulating endocannabinoid system activity and for modulating neurotransmission, and such modulation will result in treatment of a mental health condition or improvement of psychological functioning.
  • the cannabis compositions of the invention modulate this threat response by attenuating amygdala reactivity, facilitating fear extinction, enhancing consolidation of extinction learning, and reducing the reinstatement of fear responding.
  • the cannabis compositions of the invention will reduce perceived threat responding, improve general stress reactivity, and assist in the relearning of negatively charged emotional memories to reduce their aversive intensity.
  • modulations of neurotransmission and other biological systems caused by the compositions and methods of the invention both the emotional and cognitive aspects of PTSD will be effectively addressed, providing clinically significant benefits in the treatment of PTSD.
  • Certain cannabis compositions of the present invention can augment and increase such benefits through, e.g., synergistic effects resulting in greater activity at CB 1 receptors, greater bioactivity of CBD, or additional therapeutic effects, and such benefits will be shown to be increased by the combination of the cannabis compositions with the mushroom compositions of the invention using the dosing methods taught herein.
  • ECS-related gene polymorphisms i.e., ECS genotypes
  • CNR1 cannabinoid receptor gene
  • a subject with a genetic marker associated with higher anandamide, or a genetic marker associated with a reduction in the expression of CNR1 will be treated with a method specifically suited to such genetic marker, such as an increased dose of a cannabis composition, or an increased number of days of a phase of a daily dosing regimen, where such phase is for a cannabis composition.
  • a method specifically suited to such genetic marker such as an increased dose of a cannabis composition, or an increased number of days of a phase of a daily dosing regimen, where such phase is for a cannabis composition.
  • the compositions and methods of the invention will modulate neurosteroid biosynthesis, for example by assisting in the reinstatement of normal allopregnanolone levels in allopregnanolone deficient patients.
  • compositions and methods are used to increase or rebalance levels of allopregnanolone
  • improvement is found in any of one or more neuropsychopathologies, such as PTSD, epilepsy, major depression, perceived social isolation, postpartum depression, premenstrual syndrome, and anorexia nervosa or obesity complicated by anxiety and depression (Pinna, 2018).
  • neuropsychopathologies such as PTSD, epilepsy, major depression, perceived social isolation, postpartum depression, premenstrual syndrome, and anorexia nervosa or obesity complicated by anxiety and depression (Pinna, 2018).
  • allopregnanolone deficient patients will have improved symptoms of these and related disorders.
  • patients deficient in the equipotent isomer of allopregnanolone, pregnanolone will be treated.
  • compositions and methods of the invention therefore are used to modulate and restore levels of allopregnanolone and pregnanolone and have therapeutic benefits in patients with mental health conditions.
  • cannabis compositions of the invention are used to modulate endocannabinoid system (ECS) activity, and will be effective to treat disorders and conditions associated with such ECS modulation, or that can be treated thereby.
  • ECS endocannabinoid system
  • cannabis compositions of the invention are used to modulate neurosteroid biosynthesis, and will be effective to treat disorders and conditions associated with such neurosteroid biosynthesis modulation, or that can be treated thereby.
  • cannabis compositions of the invention are used to modulate serotonergic neurotransmission, and will be effective to treat disorders and conditions associated with such serotonergic modulation, or that can be treated thereby.
  • the methods of the invention that comprise the administration of a cannabis composition will therefore find use in such modulation.
  • methods of treatment comprising administration of the mushroom and cannabis compositions of the invention are provided, such as administration by a distinct or daily dosing regimen.
  • the mushroom and cannabis compositions are used to treat CNS disorders including mental health conditions (such as psychiatric or neuropsychiatric disorders), neurodevelopmental disorders, and neurodegenerative conditions and disorders.
  • “Treating” or “treatment” of a disorder includes (i) inhibiting the disorder, i.e., arresting or reducing the development or progression of the disorder or its clinical symptoms; or (ii) relieving the disorder, i.e., causing regression of the disorder or its clinical symptoms. Inhibiting the disorder, for example, would include prophylaxis.
  • a therapeutic amount necessary to effect treatment for purposes of this invention will, for example, be an amount that provides for objective indicia of improvement in patients having clinically-diagnosable symptoms.
  • Treatment covers any treatment of a disorder in a mammal, and particularly in a human, and includes: (a) preventing a disorder from occurring in a subject who may be predisposed to the disorder but has not yet been diagnosed with it: (b) inhibiting a disorder, i.e., arresting its development; (c) relieving a disorder, i.e., causing regression thereof; (d) protection from or relief of a symptom or pathology caused by or related to a disorder; (e) reduction, decrease, inhibition, amelioration, or prevention of onset, severity, duration, progression, frequency or probability of one or more symptoms or pathologies associated with a disorder; and (f) prevention or inhibition of a worsening or progression of symptoms or pathologies associated with a disorder.
  • subject refers to any mammal, including murines, simians, humans, mammalian farm animals, mammalian sport animals, and mammalian pets.
  • subject is human.
  • subject includes a subject or patient who has a mental health condition, or a condition related to a mental health condition for which similar treatment may be efficacious.
  • Such terms shall also refer to patients in need of treatment for such a disorder, persons predisposed to such a disorder, and subjects who have been diagnosed with such a disorder. Moreover, these terms shall likewise refer to persons who have received treatment or therapy for a mental health condition, are currently receiving therapy or treatment for a mental health condition, or who may receive therapy or treatment for such a disorder in the future. In embodiments, the disclosed methods also can be used to improve mental health and improve psychological functioning in non-disease states, i.e., in an individual without a diagnosed mental disorder, or specific symptoms thereof. [358] In embodiments, a therapeutically effective amount of a mushroom or cannabis composition is administered to a subject.
  • Effective refers to an amount of a composition (or an active agent thereof, depending on context) that is sufficient to provide a desired therapeutic effect.
  • An effective amount will vary depending upon the subject and the disease condition being treated or health benefit sought, the weight and age of the subject, the severity of the disease condition or degree of health benefit sought, the manner of administration, and the like, all of which will be readily determined by one of skill.
  • “Therapeutic effect” or “therapeutic efficacy” means the responses(s) in a mammal, and preferably a human, after treatment that is judged to be desirable and beneficial. Hence, depending on the disorder to be treated, or improvement in physiological or psychological functioning sought, and depending on the particular constituent(s) in the compositions of the invention under consideration, those responses shall differ, but would be readily understood by those of skill.
  • the mammal is a human.
  • Measures of therapeutic effect include outcome measures (primary or secondary), endpoints, effect measures, and measures of effect within clinical or medical practice or research which can be used to assess an effect (positive and/or negative) of an intervention or treatment, whether patient-reported (e.g., questionnaires); based on other patient data (e.g., patient monitoring); gathered through laboratory tests such as from blood or urine; through medical examination by a doctor or medical professional, or by digital or electronic means, including online tools, smartphones, wireless devices, biosensors, health apps, and the like.
  • compositions of the invention may be administered to the patient (alone, with another patient as in couples therapy, or as part of group therapy) in the presence of one or more therapists in a therapeutic setting to facilitate psychotherapy.
  • psychedelic-assisted psychotherapy or drug-assisted therapy see, e.g., Sessa, 2019, and references therein.
  • the compositions of the invention will be administered not as part of a psychotherapeutic regimen, or as part of drug-assisted therapy, but will be self-administered by a patient, and such self-administration may or may not be under the supervision of a medical professional.
  • a patient may be administered the compositions of the invention and be monitored, a patient may be administered the compositions of the invention and receive psychological support, and a patient may be administered the compositions of the invention and receive psychotherapy, and such may take place for example with a psychiatrist, medical doctor, clinical psychologist, or other trained clinician, as well as with a “guide” or non-clinical practitioner.
  • “Therapist” herein therefore may refer to any person who treats a patient using the compositions and methods of the invention, whether that person is a care provider, a psychiatrist, clinical psychologist, clinical therapist, psychotherapist, or other trained counselor, facilitator, or guide, and who may or may not be a trained counselor.
  • the patient is supervised or monitored by a clinician, guide, therapist, or care provider.
  • supervision or monitoring may be completed in person or remotely, for example, over the telephone (e.g., telemedicine), through video conferencing (e.g., Zoom, Teams, Skype, FaceTime), with the use of virtual reality (VR), including both synchronous and asynchronous means, or otherwise through software or an app.
  • a subject may or may not be supervised, and may or may not be instructed on how to self-administer the composition.
  • instruction it may be in the form of physical instructions that accompany a composition of the invention, through software or an app, or through publicly available information.
  • a subject completes one or more remote consultations with a care provider, e.g., by phone, by video conference, or in virtual reality (VR), prior to administration of the compositions.
  • remote consultations are used by the care provider to determine whether administration of the compositions is appropriate for the subject.
  • the one or more remote consultations may be used by the care provider to determine and/or confirm diagnosis of a condition, such as but not limited to a CNS disorder, which can be treated by administration of a compound provided herein.
  • the one or more consultations is conducted in person.
  • a subject completes one or more in person preparatory sessions with a care provider, or one or more remote preparatory sessions, e.g., by phone, by video conference, or in virtual reality, prior to administration of the compositions of the invention.
  • a subject completes one or more in person integration sessions with a care provider, or one or more remote integration sessions, e.g., by phone, by video conference, or in virtual reality, after administration of the compositions of the invention.
  • the compositions of the invention are used to treat CNS disorders, including mental conditions and mental health disorders, psychiatric and neuropsychiatric disorders, and neurodegenerative conditions.
  • such disorders may be characterized as those codable as “mental, behavioral, or neurodevelopmental disorders” per the ICD-11 and/or its predecessor ICD-10.
  • ICD-11 such disorders are characterized by clinically significant disturbance in an individual's cognition, emotional regulation, or behavior that reflects a dysfunction in the psychological, biological, or developmental processes that underlie mental and behavioral functioning. These disturbances are usually associated with distress or impairment in personal, family, social, educational, occupational, or other important areas of functioning (World Health Organization, 2019).
  • CNS disorders further include conditions involving the brain and spinal cord caused by any of genetic factors, trauma, infections, degenerations, structural defects and/or damages, tumors, blood flow disruption, autoimmune disorders, or a combination thereof.
  • CNS disorders refer to any disease of the nervous system, including diseases that involve the central nervous system (brain, brainstem, spinal cord, and cerebellum), the peripheral nervous system (including cranial nerves), and the autonomic nervous system (parts of which are located in both central and peripheral nervous system).
  • Neurodegenerative disorders refer to a type of neurological disease marked by the loss of nerve cells, including, but not limited to, Alzheimer's disease, Parkinson's disease, frontotemporal dementia, multiple sclerosis, amyotrophic lateral sclerosis, tauopathies (including frontotemporal dementia), multiple system atrophy, and Huntington's disease.
  • neurological diseases include, but are not limited to, headache, stupor and coma, dementia, seizure, sleep disorders, trauma, infections, neoplasms, neuro-ophthalmopathy, movement disorders, demyelinating diseases, spinal cord disorders, and disorders of peripheral nerves, muscle and neuromuscular junctions.
  • Addiction and mental illness include, but are not limited to, bipolar disorder, depression and schizophrenia, are also included in the definition of CNS diseases.
  • neurological diseases include acquired epileptiform aphasia; aaccuuttee disseminated encephalomyelitis; adrenoleukodystrophy; agenesis of the corpus callosum; agnosia; Aicardi syndrome; Alexander disease; Alpers' disease; alternating hemiplegia; Alzheimer's disease; amyotrophic lateral sclerosis; anencephaly; Angelman syndrome; angiomatosis; anoxia; aphasia; apraxia; arachnoid cysts; arachnoiditis; Arnold-Chiari malformation; arteriovenous malformation; Asperger syndrome; ataxia telangiectasia; attention deficit hyperactivity disorder; autism; autonomic dysfunction; back pain; Batten disease; Behcet's disease; Bell's palsy; benign essential blepharospasm; benign focal amyotrophy; benign intracranial hypertension; Binswanger's disease; ble
  • MMEennttaall hheeaalltthh ccoonnddiittiioonnss may refer to disease conditions that generally involve negative changes in emotion, mood, thinking, or behavior, and include, but are not limited to, those disclosed herein. Broadly, mental health conditions include those characterized by the DSM-5, Merck Manual, ICD-11, or other such diagnostic resources known to those of skill.
  • a variety of methods for screening or assessing a subject for a mental health condition exist.
  • a diagnosis of a mental health condition is facilitated with use of the Diagnostic and Statistical Manual of Mental Disorders, such as the DSM-5.
  • diagnosis of a mental health condition is facilitated with use of self-reported or observer-report surveys or questionnaires.
  • Non-limiting examples of such questionnaires include the Patient Health Questionnaire 9 (PHQ-9), the Generalized Anxiety Disorder 7 (GAD-7), PTSD Checklist for DSM-5 (PCL-5), the Alcohol Use Disorders Identification Test (AUDIT), Binge Eating Scale (BES), Obsessive-Compulsive Inventory (OCI), the Personality Disorders Questionnaire (PDQ-IV), Dissociative Experiences Scale (DES), Drug Use Questionnaire (D AST-20), the Mood Disorder Questionnaire (MDQ), and other similar questionnaires.
  • alternative questionnaires such as the Clinical Global Impression-Improvement scale (CGI-I), may be used to assess improvement of a subject’s mental health state, such as by comparing baseline responses to responses after a treatment intervention.
  • any of the diagnostic manuals and assessments described, and other similar tools may be used to confirm a reduction in symptoms, a reduction in symptom severity, or elimination of symptoms and/or a previous diagnosis.
  • the compositions of the invention are useful to treat a patient diagnosed with or diagnosable with at least one mental health condition, or that has any one or more symptoms thereof.
  • a patient diagnosed with a mental health condition is prescribed a therapeutically effective amount of the compositions of the invention, e.g., a prescription is provided for one or more compounds.
  • the compositions are prescribed to a patient diagnosed with a mental health condition as a therapeutic regime, a defined dosing regimen, or a daily dosing regimen, any of which comprising effective amounts of the compositions described herein, e.g., a prescription is provided for one or more dosing method(s).
  • a patient diagnosed with a mental health condition obtains the compositions of the invention without a prescription.
  • a patient diagnosed with a mental health condition obtains the compositions of the invention, and instructions for their use, without a prescription.
  • a patient diagnosed with a mental health condition is administered an effective amount of the compositions of the invention by a clinician.
  • a patient diagnosed with a mental health condition self-administers the compositions of the invention.
  • a patient diagnosed with a mental health condition is supervised by a health professional while self-administering the compositions of the invention while, in other embodiments, the patient is not supervised by a health professional while self-administering the compositions of the invention.
  • “Supervision” and “supervised” will include remote supervision, supervision by electronic means, supervision by patient adherence to a protocol where evidence of such is shared with a health professional, supervision by an individual dispensing one or more compositions by confirming that a patient is allowed to purchase such composition(s), in person supervision during administration itself, confirmation by a patient that said patient is in compliance, and other forms of “supervision” as will be readily appreciated in the art.
  • a patient diagnosed with a mental health condition is administered, either by the patient or a third-party, an effective amount of the compositions of the invention as part of a psychotherapy regimen.
  • a patient diagnosed with a mental health condition is administered, either by the patient or a third-party, an effective amount of the compositions of the invention not part of a psychotherapy regimen, without any accompanying psychotherapy or other therapy, without any supervision or monitoring, or without any intervention by a health care professional.
  • compositions of the invention including those comprising psilocybin or other psychedelic tryptamines, or Psilocybe extracts, may be obtainable without a prescription, but merely for example by providing proof of the patient’s age, or by taking and passing a “licensing test” (as has been suggested by MAPS), or another method to ensure safe and responsible use.
  • the “patient diagnosed with at least one mental health condition” or “patient diagnosed with a mental health condition” is a patient having an undiagnosed mental health condition, or a patient having symptoms of one or more mental health conditions, but not meeting all criteria for its diagnosis, or a patient having no mental health conditions and no symptoms thereof.
  • a patient will take the compositions of the invention for the improvement of mental health and mental functioning, for increased feelings of wellbeing, or for other purposes sometimes referred to broadly as the “betterment of the well.”
  • the mushroom or cannabis compositions disclosed herein are efficacious in reducing at least one symptom of a mental health condition within a patient diagnosed with the same.
  • symptoms for each mental health condition will be different, however, through medical monitoring (such as monitoring of objective measurements, as described herein), patient reporting (such as, but not limited to through journaling), completion of questionnaires, etc., one will be able to objectively determine if a symptom has reduced in its frequency and/or magnitude.
  • compositions of the invention are shown to be useful in methods for treating a variety of mental health conditions linked to dysregulation or inadequate functioning of the ECS or of neurotransmission in mammals.
  • depression including in forms such as drug resistant and treatment resistant depression (TRD) as well as major depressive disorder (MDD), dysthymia, dysthymia, anxiety and phobia disorders (e.g., generalized anxiety or GAD, social anxiety, panic, post-traumatic stress and adjustment disorders), feeding and eating disorders (including binge eating, bulimia, and anorexia nervosa), other binge behaviors, body dysmorphic syndromes, alcoholism, tobacco abuse, drug abuse or dependence disorders including substance use disorders (SUDs) such as opioid use disorders, alcohol use disorder (AUD), cannabis use disorder (CUD), amphetamine and methamphetamine use disorders, caffeine and other stimulant use disorders, prescription drug abuse disorders, disruptive behavior disorders, impulse control disorders, gaming disorders, gambling disorders, pornography and s
  • mental health conditions of the invention are specifically the “trauma- and stressor-related disorders,” which include acute stress disorder, adjustment disorders, and PTSD (Merck Manual), as well as reactive attachment disorder, disinhibited social engagement disorder, and others (DSM-5).
  • the mental health condition is specifically anxiety, depression, or PTSD.
  • the neurophysiology underlying mental health conditions may be distinct, an aspect in common of many is the presence of a deleterious, repetitive, and often “rigid” thought process that negatively impacts an individual’s ability to function.
  • symptoms involve re-experiencing trauma and the feelings associated with it; for depression it can take the form of a recurrent internal editor that attaches negative connotations to normal life events; and for addiction it is the preoccupation with acquiring and using the substance of choice.
  • the method of treating a mental health condition involves the treatment of a disorder related to rigid modes of thinking.
  • the disorder related to rigid modes of thinking can be anxiety, depression, addiction, an eating disorder, obsessive compulsive disorder, or PTSD.
  • the compositions of the invention are used to reduce the symptoms of a mental health condition.
  • the symptoms of the mental health condition to be treated shall be able to be determined by one of skill, by reference to the general understanding of the art regarding that disorder.
  • Symptoms of PTSD include transient waking dissociative states in which events are relived as if happening (“flashbacks”), nightmares, distressing and intense memories, other intrusive negative memories, distress or physical reactions after being exposed to triggers, blaming self or others for the trauma, decreased interest in things that were once enjoyable and other feelings of emotional numbness, negative feelings about self and the world, inability to remember the trauma clearly, difficulty feeling positive, feelings of isolation, negative affect, difficulty feeling positive, other negative alterations in cognition and mood, avoidance, aggression or irritability, hypervigilance and hyper-awareness, difficulty concentrating, difficulty sleeping, heightened startle response, engaging in self-destructive, or risky behavior, difficulty sleeping or staying asleep, and suicidal
  • compositions used are the psilocybin compositions of the invention
  • such reductions occur without psychedelic effects, and therefore allow for treatment at home or otherwise outside of a clinic and without the need for concomitant psychotherapy.
  • Reductions can also be measured using questionnaires or assessments known in the art, such as for PTSD, CAPS.
  • assessments known in the art, such as for PTSD, CAPS.
  • a patient may complete a pre-treatment assessment to determine a baseline measure, and then repeat the process at set points during treatment, at the conclusion of treatment, and/or at one or more predetermined periods of time following treatment, to objectively measure the effectiveness of treatment, or one or more aspects of treatment and its efficacy, wherein a decrease or increase in an overall score (e.g., as measured by a test’s “scale”) may determine whether the severity and/or frequency of symptoms has increased, stayed the same, or decreased.
  • a reduction in score generally indicates symptom improvement, but in some scales, such as GAF, an increase in score indicates improvement.
  • Neurodegenerative conditions encompass a wide range of disorders that result from progressive damage to cells and nervous system connections essential for at least one of mobility, coordination, strength, sensation, and cognition; and encompass those disclosed herein.
  • Symptoms of neurodegenerative diseases include any of reductions in mobility and balance, abnormal movements, difficulty swallowing, abnormal bladder and bowel movement function, wide variations in blood pressure, difficulty sleeping, difficulty breathing, abnormal heart function, reduced memory and cognitive abilities, alterations in mood, and changes to speech (UT Soiled Medical Center, 2022).
  • the compositions of the invention are useful in treating a patient diagnosed with or diagnosable with at least one neurodegenerative condition, or that has any one or more symptoms thereof.
  • a patient diagnosed with at least one neurodegenerative condition is prescribed a therapeutically effective amount of the compositions of the invention.
  • a patient diagnosed with at least one neurodegenerative condition obtains the compositions of the invention without a prescription.
  • a patient diagnosed with at least one neurodegenerative condition is administered an effective amount of the compositions of the invention by a clinician.
  • a patient diagnosed with at least one neurodegenerative condition self-administers the compositions of the invention.
  • a patient diagnosed with at least one neurodegenerative condition is supervised by a health professional while self-administering the compositions of the invention while, in other embodiments, the patient is not supervised by a health professional while self-administering the compositions of the invention.
  • a patient diagnosed with at least one neurodegenerative condition is administered, either by the patient or a third-party, an effective amount of the compositions of the invention as part of a psychotherapy regimen.
  • a patient diagnosed with at least one neurodegenerative condition is administered, either by the patient or a third-party, an effective amount of the compositions of the invention not part of a psychotherapy regimen.
  • the compositions of the invention are efficacious in reducing at least one symptom of a neurodegenerative condition within a patient diagnosed with the same.
  • compositions of the invention are useful to treat the conditions and disorders discussed herein may be shown in part by the following Examples.
  • EXAMPLE 20 Determination of Therapeutic Efficacy to Treat PTSD and MDD [384] A study is designed to determine the efficacy of the compositions of the invention in the treatment of PTSD and/or major depressive disorder (MDD). The study will include 10 subject (preferably 5 male and 5 female) whose diagnoses for at least one of PTSD and/or MDD will be confirmed prior to the study beginning.
  • MDD major depressive disorder
  • the objectives of the study will be to demonstrate the initial subject acceptance of a therapeutic regime of the invention, to measure pre- and post- changes from baseline to final dosage, and to measure brain activity and gain subject reported response during therapy.
  • Inclusion criteria for the subjects include ensuring the subject is at least 21 years old, is diagnosed with at least one of PTSD or MDD, has not experienced satisfactory treatment for such condition(s), and will abstain from using contraindicated drugs during treatment.
  • the protocol itself the subject will undergo a dosing regimen comprising 4 continuous days of dosing of a M+ mushroom composition of EXAMPLE 2 followed by 3 continuous days of dosing of a C+ cannabis composition of EXAMPLE 9.
  • the subject will undergo a dosing regimen comprising 4 continuous days of dosing of a M+ mushroom composition of EXAMPLE 2 followed by 3 continuous days of dosing of a C– cannabis composition of EXAMPLE 9.
  • the subject will undergo a dosing regimen comprising 4 continuous days of dosing of a M– mushroom composition of EXAMPLE 2 followed by 3 continuous days of dosing of a C+ cannabis composition of EXAMPLE 9.
  • the subject will undergo a dosing regimen comprising 4 continuous days of dosing of a M– mushroom composition of EXAMPLE 2 followed by 3 continuous days of dosing of a C– cannabis composition of EXAMPLE 9.
  • the subject will undergo a dosing regimen comprising 4 continuous days of dosing of a M+ mushroom composition of EXAMPLE 2 followed by 3 continuous days of abstaining (washout).
  • the subject will undergo a dosing regimen comprising 4 continuous days of dosing of a M– mushroom composition of EXAMPLE 2 followed by 3 continuous days of abstaining (washout).
  • the alternative protocols also may be used as additional treatment arms, to compare the compositions and dosing regimens of the invention against each other.
  • the first four days of dosing a M+ composition are done in a controlled setting with initial education prior to the first dose, to ensure a lack of psychedelic effects by the subject, and sufficient comfort by the subject with microdosing psychedelics.
  • subjects may be assessed by a physician to gauge the intensity of any observed side effects. Patient reported outcomes are then performed prior to each dosing day. At the end of the first four days, the patient is evaluated regarding the need for continued controlled dosing. If it is determined that self-dosing is safe and may be effective, an ongoing symptom and outcome record and separate liability form must be completed.
  • the mushroom composition may be dispensed at a time.
  • a medication compliance form may be filled out and collected prior to dispensing another dosing regimen’s worth (e.g., four days’ supply) of the mushroom composition.
  • the treatment period is 8 weeks, or about 60 days, after which the evaluation and testing will be completed. Results will indicate that at least one symptom of PTSD and/or MDD are lessened, reduced in frequency, or otherwise attenuated by the compositions of the invention.
  • EXAMPLE 19 Determination of Therapeutic Efficacy for Other CNS Disorders
  • a study is designed to determine the efficacy of the compositions of the invention in the treatment of other CNS disorders besides PTSD and MDD, including any one or more of adjustment disorder, affective disorder, depression, atypical depression, postpartum depression, catatonic depression, a depressive disorder due to a medical condition, premenstrual dysphoric disorder, seasonal affective disorder, dysthymia, anxiety, phobia disorders, binge disorders, body dysmorphic disorder, alcohol or drug abuse or dependence disorders, substance-related disorders, substance use disorders, alcohol use disorder, substance-induced mood disorder, a mood disorder related to another health condition, disruptive behavior disorders, eating disorders, impulse control disorders, obsessive compulsive disorder (OCD), attention deficit hyperactivity disorder (ADHD), personality disorders, attachment disorders, and dissociative
  • results will indicate that at least one symptom of the CNS disorder are lessened, reduced in frequency, or otherwise attenuated by the compositions of the invention. Results will be compiled for reporting to Health Canada to build a dossier for Phase 0 and Phase I trials, and IND guidance.
  • EXAMPLE 20 Use as a Medication
  • the compounds described herein are prescribed in a therapeutically effective amount to a patient suffering from at least one CNS disorder, including mental health conditions such as post-traumatic stress disorder (PTSD), adjustment disorder, affective disorder, depression, atypical depression, postpartum depression, catatonic depression, a depressive disorder due to a medical condition, premenstrual dysphoric disorder, seasonal affective disorder, dysthymia, anxiety, phobia disorders, binge disorders, body dysmorphic disorder, alcohol or drug abuse or dependence disorders, substance-related disorders, substance use disorders, alcohol use disorder, substance-induced mood disorder, a mood disorder related to another health condition, disruptive behavior disorders, eating disorders, impulse control disorders, obsessive compulsive disorder (OCD), attention deficit hyperactivity disorder (ADHD), personality disorders, attachment disorders, and dissociative disorders; and neurodegenerative conditions, such as but not limited to Alzheimer’s disease, ataxia, Huntington’s disease, Parkinson’s disease, motor neuron disease, multiple system atrophy,
  • a patient is evaluated to determine whether the compositions are appropriate for the patient.
  • An evaluation may include completing questionnaires, obtaining objective health measurements from the patient, such as one or more of weight, body temperature, heart rate, respiratory rate, blood oxygenation, BP and its variables (e.g., SBP, SBP, MAP, and PP); CNIBP; ECG measurements (e.g., RR interval or its variability), QT interval or its variability, HRV (including measured by devices other than an ECG); hemodynamic response; levels of glucose, cortisol, serotonin, dopamine, or cholesterol; EEG measures; BDNF; genetic markers including relating to CYP enzymes or drug metabolism; genetic variation in mGluR5 or FKBP5; other biomarkers; and digital biomarkers, including digital biomarkers associated with any one or more of the foregoing.
  • the patient may be prescribed the compositions of the invention, and the type or amount or other characteristics of the compositions of the invention may be modified thereby, for example a prescribed dose will be appropriate for the age and weight of the patient, and may additionally take into consideration other factors known to those of skill, such as comorbidities, current medications (if any), and metabolic or genetic variations among patient populations.
  • one or more diagnostic or clinical tools is used, such as the DSM–5, one or more self-reported or observer-report surveys or questionnaires, and any one or more of the Patient Health Questionnaire 9 (PHQ-9), the Hamilton Depression Rating Scale (HAM–D); the Generalized Anxiety Disorder 7 (GAD-7), PTSD Checklist for DSM–5 (PCL-5), The Alcohol Use Disorders Identification Test (AUDIT), Binge Eating Scale (BES), Obsessive-Compulsive Inventory (OCI), the Personality Disorders Questionnaire (PDQ-IV), Dissociative Experiences Scale (DES), Drug Use Questionnaire (DAST-20), the Mood Disorder Questionnaire (MDQ), and one or more other similar questionnaires, which may be chosen based on the condition or disorder to be treated, as known in the art.
  • PHQ-9 Patient Health Questionnaire 9
  • HAM–D Hamilton Depression Rating Scale
  • GCD-7 Generalized Anxiety Disorder 7
  • EXAMPLE 21 Dosing Regimens for the Compositions of the Invention [397] Exemplary and non-limiting dosing regimens for the compositions of the invention, with M+, M–, C+, C–, and A as herein defined are listed below.
  • a “therapeutic regime” will comprise one or more “defined dosing regimens” and/or “daily dosing regimens” as listed in the Examples below, and that such therapeutic regimes will therefore comprise the treatment administrations or days from 1-11 as shown (or any smaller number, e.g., from 1-7, 1-8, 1-9, or 1-10), repeating any number of times of at least one, and including in some embodiments one or more defined dosing regimens, two or more defined dosing regimens, three or more defined dosing regimens, four or more defined dosing regimens, five or more defined dosing regimens, six or more defined dosing regimens, seven or more defined dosing regimens, eight or more defined dosing regimens, nine or more defined dosing regimens, 10 or more defined dosing regimens, 11 or more defined dosing regimens, 12 or more defined dosing regimens, or greater than 12 defined dosing regimens, which together will comprise a single “therapeutic regime” of the invention.
  • a therapeutic regime will comprise, together, one or more daily dosing regimens, two or more daily dosing regimens, three or more daily dosing regimens, four or more daily dosing regimens, five or more daily dosing regimens, six or more daily dosing regimens, seven or more daily dosing regimens, eight or more daily dosing regimens, nine or more daily dosing regimens, 10 or more daily dosing regimens, 11 or more daily dosing regimens, 12 or more daily dosing regimens, or greater than 12 daily dosing regimens.
  • an Example provides for less than 11 total administrations or days (as indicated by blank treatment administrations/days in the below) the therapeutic regime will repeat following the last administration period (“period” including days, and also including other periods of shorter or longer duration) or day shown.
  • a defined dosing regimen of Examples 22-55, each lasting seven administrations (or “periods”) will repeat every seven administrations, with each seven administrations repeating as below, or of Examples 56-71, each lasting eight administrations, will repeat every eight administrations, with each eight administrations repeating as below.
  • each will be understood to last seven days, and each will repeat weekly, with each week repeating as below; or of Examples 56-71, each lasting eight days, and repeating every eight days, with each eight days repeating as below.
  • a therapeutic regime may be a “combination therapeutic regime,” wherein it comprises multiple defined dosing regimens and/or daily dosing regimens from different examples. For example, in some embodiments it can combine multiple defined dosing regimens from different examples (a “combination defined dosing regimen”), rather than a single defined dosing regimen, repeated a specific number of times (a “repeated defined dosing regimen”).
  • Such combination therapeutic regimes may comprise two or more different defined dosing regimens, including three or more different defined dosing regimens, four or more different defined dosing regimens, five or more different defined dosing regimens, six or more different defined dosing regimens, seven or more different defined dosing regimens, eight or more different defined dosing regimens, nine or more different defined dosing regimens, 10 or more different defined dosing regimens, 11 or more different defined dosing regimens, 12 or more different defined dosing regimens, or greater than 12 different defined dosing regimens, each following the last, end to end, comprising a combination therapeutic regimen.
  • a therapeutic regime can combine daily dosing regimens from different examples (a “combination daily dosing regimen”), rather than a single daily dosing regimen, repeated a specific number of times (a “repeated daily dosing regimen”).
  • Such combination therapeutic regimes may comprise two or more different daily dosing regimens, including three or more different daily dosing regimens, four or more different daily dosing regimens, five or more different daily dosing regimens, six or more different daily dosing regimens, seven or more different daily dosing regimens, eight or more different daily dosing regimens, nine or more different daily dosing regimens, 10 or more different daily dosing regimens, 11 or more different daily dosing regimens, 12 or more different daily dosing regimens, or greater than 12 different daily dosing regimens, each following the last, end to end, comprising a combination therapeutic regimen.
  • a therapeutic regime can combine both defined dosing regimens and daily dosing regimens from different examples (a “combination dosing regimen”), rather than a single distinct or daily dosing regimen, repeated a specific number of times (a “repeated dosing regimen”).
  • Such combination therapeutic regimes may comprise two or more different distinct and daily dosing regimens, including three or more different distinct and daily dosing regimens, four or more different distinct and daily dosing regimens, five or more different distinct and daily dosing regimens, six or more different distinct and daily dosing regimens, seven or more different distinct and daily dosing regimens, eight or more different distinct and daily dosing regimens, nine or more different distinct and daily dosing regimens, 10 or more different distinct and daily dosing regimens, 11 or more different distinct and daily dosing regimens, 12 or more different distinct and daily dosing regimens, or greater than 12 different distinct and daily dosing regimens, each following the last, end to end, comprising a combination therapeutic regimen.
  • a combination therapeutic regime will include an “all plus” distinct and/or daily dosing regimen (e.g., M+ and C+), followed by an “all minus” distinct and/or daily dosing regimen (e.g., M– and C—), such as Example 34 followed by Example 37, the two in alternating fashion.
  • an “all plus” distinct and/or daily dosing regimen e.g., M+ and C+
  • an “all minus” distinct and/or daily dosing regimen e.g., M– and C—
  • EXAMPLES 22-205 include, in certain exemplary embodiments, the administration of a psychedelic mushroom composition (e.g., comprising psilocybin and/or a Psilocybe extract) ( M+ ), followed or preceded by the administration of at least one of a functional mushroom composition (e.g., comprising no psilocybin and no Psilocybe extract) ( M– ), an intoxicating cannabis composition (e.g., comprising THC) ( C+ ), or a non-intoxicating cannabis composition (e.g., comprising no THC) ( C– ); alternatively, as illustrated in certain of EXAMPLES 22-205 , administration of the M+ mushroom composition may be preceded and/or followed by abstinence or washout ( A ).
  • a psychedelic mushroom composition e.g., comprising psilocybin and/or a Psilocybe extract
  • M– a functional mushroom composition
  • the M+ mushroom composition is a composition of EXAMPLE 2 or 3.
  • the (C+) cannabis composition is a composition of EXAMPLE 9.
  • the M– mushroom composition is a composition of EXAMPLE 2 or 3 , but without a Psilocybe extract.
  • the (C–) cannabis composition is a composition of EXAMPLE 9 , but without THC.
  • the dosing regimen illustrated in each of EXAMPLES 22-205 will be repeated and continue for a specified or predetermined duration of time, such duration of time being the therapeutic regime.
  • the specified duration of time is between about 15 to about 90 days.
  • the specified duration of time is between about 30 to about 75 days.
  • the specified duration of time is between about 50-65 days.
  • the specified duration of time is 60 days.
  • the specified duration of time is 45 days.
  • the specified duration of time is 30 days.
  • the specified duration of time is a period of weeks, such as two weeks, three weeks, four weeks, five weeks, six weeks, seven weeks, eight weeks, nine weeks, 10 weeks, 11 weeks, 12 weeks, or more than 12 weeks.
  • the specified duration of time is a specific number of repetitions of the defined dosing regimen(s), the daily dosing regimen(s), or the combination of dosing regimens.
  • the therapeutic regime lasts for a specified duration of between about 15 to about 90 days, inclusive; however, the pattern illustrated in EXAMPLES 22-205 does not continue for the entirety of the specified duration.
  • the first 11 treatment administrations or days are illustrated in one of EXAMPLES 22-205 , but after the initial 11 administrations or days, the treatment schedule is fluid, and is instead determined by one, or a combination of, objective health measurements, as disclosed herein; or subjective patient data, obtained through any of a questionnaire, interview, and journaling.
  • the subsequent portion of the therapeutic regime may include set administration or dosing days (e.g., administration of a M+ mushroom composition, that may or may not be the formulation of EXAMPLE 2 or 3 , for four consecutive administrations or days), but may then be “modular,” such that other administrations or days may comprise any of abstinence (A), or administration of at least one of a M+ mushroom composition, a C+ cannabis composition, or a C– cannabis composition.
  • the patient, a clinician, or both may determine which compositions(s) is(are) administered, or if abstinence will occur, and if the A day will include another composition comprising additional active agents, such as vitamins, nootropics, and/or supplements.
  • the M+ dosing days also may be modular, such that, preceding or following an M+ dosing day, a patient may be administered, by a patient or a third party, a M– mushroom composition, a C+ or C– cannabis composition, or may have a washout.
  • EXAMPLES 22-205 are merely illustrative examples, and should not be construed as limiting.
  • a patient may be prescribed a treatment regimen of a specified duration of between about 15 to about 90 days, inclusive, wherein on any given administration period or day, a patient may be administered one of, or a combination thereof, a M+ or M– mushroom composition, a C+ or C– cannabis composition, or may have a washout, which includes administration of a composition of additional active agents.
  • a patient will not be prescribed a therapeutic regime with a predetermined length or number of dosing regimens, such that therapeutic regime may be as short or as long in duration as necessary, and may be determined by the patient, a clinician prescribing the treatment, or a combination.
  • the formulations detailed in EXAMPLES 22-205 are packaged as distinct dosing kits or daily dosing kits.
  • the distinct dosing kits may be kits that contain 7, 11, 14, 21, 28, 29, 30, 31, 45, 50, 60, 75, or 90 administrations-worth of the mushroom and/or cannabis compositions described herein, optionally including “A” administration compositions of additional active agents.
  • the daily dosing kits may be kits that contain 7, 11, 14, 21, 28, 29, 30, 31, 45, 50, 60, 75, or 90 days-worth of the mushroom and/or cannabis compositions described herein, optionally including “A” day compositions of additional active agents.
  • the number of administrations or days in a kit will be any number from 2 to 90 or greater than 90.
  • the daily dosing kits will include a M+ mushroom composition of either EXAMPLE 2 or 3 according to a set schedule, and will include one, two, three, or all of a C+ cannabis composition, a C– cannabis composition, an M– mushroom composition, and an A composition of additional active agents, for each administration or day not scheduled to be an administration or day wherein an M+ mushroom composition is to be administered (such as that illustrated in FIG.3 ).
  • the patient may be administered or may self-administer one, two, three, or all of a C+, C–, M–, or A composition as described herein.
  • an M+ composition comprises a Psilocybe spp. extract, or a like “psychedelic mushroom extract” from another genera, such as Conocybe, Copelandia, Galerina, Gymnopilus, Inocybe, Mycena, Panaeolina, Panaeolus, Pholiotina, or Pluteus .
  • 1 kg psilocybin fungal material is first obtained as fruiting bodies, truffles, and/or mycelium, and then sufficiently dried in an air drier so as to remove moisture, but not damage the fungal material.
  • the sufficiently dried fungal material is then placed in an industrial processor having a sufficient number of blades to facilitate the fine chopping thereof (such as a BSC coarse crusher).
  • the finely-chopped material is then placed in an industrial pulverizer sufficient to reduce the fungal material into a sufficiently fine powder (such as a BSP pulverizer). Once the fungal material is reduced to a fine powder, it is transferred to a vat.
  • An ethanol solvent (e.g., of at least 99%) is then introduced in a volume sufficient to fully immerse the dried material, transforming the mixture into a homogeneous slurry.
  • the entire mixture is then agitated within the vat, ensuring constant exchange between the fungal material and the ethanol solvent. After roughly 24 hours, the agitation ceases, and the slurry is transferred to a filtration device of sufficient porosity to separate the fungal material from the alcohol solution.
  • the alcohol solution is then evaporated under ambient air conditions, via fans blowing ambient air over the solution to facilitate evaporation, or evaporated under heat and vacuum pressure.
  • the solution may be spray dried using methods and techniques generally known in the art.
  • Non-inferiority trials are performed to demonstrate that the compositions and methods herein perform no worse than at least one comparator composition or method, by more than a pre-specified amount. Such trials show that the compositions and methods of the invention are at least as useful as for treatment of a specified indication and have approximately the same efficacy as the comparator(s). In embodiments, the compositions and methods of the invention are shown to offer other benefits such as a better safety profile.
  • EXAMPLE 207 Exemplary Equivalence Trial of Formulations and Methods [414] Equivalence trials are performed to demonstrate that the compositions and methods herein perform at least as good as at least one comparator composition or method, within a pre-specified range. Such trials show that the compositions and methods of the invention are at least as useful for treatment of a specified indication and are at least as effective as the comparator(s). In embodiments, the compositions and methods of the invention are shown to offer other benefits such as a better safety profile.
  • EXAMPLE 208 Exemplary Superiority Trial of Formulations and Methods [415] Superiority trials are performed to demonstrate that the compositions and methods herein perform better than at least one comparator composition or method, by more than a pre-specified amount.
  • compositions and methods of the invention are more useful for treatment of a specified indication and have better efficacy than the comparator(s).
  • the compositions and methods of the invention are shown to offer other benefits such as a better safety profile.
  • EXAMPLE 209 Exemplary Study to Illustrate Synergistic Properties [416]
  • a study is designed to determine the presence and magnitude of synergistic properties within the entourage enhanced compositions of the invention.
  • the study includes a population of 90 individuals (45 men and 45 women) diagnosed with at least one CNS disorder, as disclosed above, who are each administered the compositions as part of a therapeutic treatment regimen that lasts a total of 60 days, comprising the administration of one composition for four consecutive days, followed by the administration of a separate composition for three consecutive days.
  • each participant Prior to commencing the dosing regimen, each participant’s symptoms, and the severity thereof, are determined to obtain a baseline. Methods of measuring such symptoms are as discussed herein or known in the art, and may include participation in a clinical outcome assessment specific to that disorder. [417] Of the study population, the participants are split into nine groups of 10 each (5 men, 5 women), with each group being assigned a separate dosing regimen: 1. Group 1 : An entourage enhanced mushroom composition of the invention, paired with an entourage enhanced cannabis composition of the invention. 2. Group 2 : An entourage enhanced mushroom composition of the invention, paired with a cannabis composition that is not entourage enhanced 3. Group 3 : A mushroom composition that is not entourage enhanced, with an entourage enhanced cannabis composition of the invention. 4.
  • Group 4 A mushroom composition that is not entourage enhanced, with a cannabis composition that is not entourage enhanced. 5.
  • Group 5 A placebo, with an entourage enhanced cannabis composition of the invention. 6.
  • Group 6 An entourage enhanced mushroom composition of the invention, with a placebo. 7.
  • Group 7 A placebo, with a cannabis composition that is not entourage enhanced. 8.
  • Group 8 A mushroom composition that is not entourage enhanced, with a placebo.
  • Group 9 A placebo, with a placebo. [418] At the end of the 60-day period, the same method used to determine the symptoms and their severity is again completed by each patient.
  • compositions and methods for enhancing working and long-term memory support using new combination of ginkgo biloba and panax ginseng and associated components 34. Sherwood, A. M., Halberstadt, A. L., Klein, A. K., McCorvy, J. D., Kaylo, K. W., Kargbo, R. B., & Meisenheimer, P. (2020). Synthesis and biological evaluation of tryptamines found in hallucinogenic mushrooms: Norbaeocystin, Baeocystin, norpsilocin, and aeruginascin. Journal of Natural Products, 83(2), 461–467. 35.

Abstract

The present invention discloses compositions from mushrooms and cannabis, including with synergistic and entourage enhanced combinations of psilocybin and other psychoactive tryptamines, THC, CBD, and other cannabinoids, and other active agents, all of which useful in methods to treat CNS and mental health disorders and to improve mental health, for example when administered in distinct or daily dosing regimens as taught herein.

Description

DOSING REGIMENS OF PHARMACEUTICAL AND NUTRACEUTICAL MUSHROOM AND CANNABIS COMPOSITIONS AND THEIR USE TO TREAT CNS DISORDERS AND IMPROVE MENTAL HEALTH INVENTORS: Radka Milanova, James Smeeding, Calum Hughes CROSS-REFERENCE [01] This application claims priority under PCT Article 8(1) and PCT Rule 4.10 to U.S. Provisional Application No.63/160,235, filed March 12, 2021, which is incorporated by reference for all purposes as if fully set forth herein. TECHNICAL FIELD [02] Described herein are pharmaceutical and nutraceutical compositions from mushrooms and Cannabis which are useful to treat CNS and mental health disorders and to improve mental health, for example when administered in the disclosed distinct daily dosing regimens. BACKGROUND OF THE INVENTION [03] Mental health conditions are the leading cause of disability worldwide. They lead to over a million lives lost to suicide, cost the global economy $1 trillion annually in lost productivity, and affect 1 in 5 adults. Their incidence has been increasing over past decades, and accelerating over the last few years. Despite the recognition that such conditions, and CNS disorders more broadly, are a great and growing problem, only half of those afflicted receive treatment, largely due to a dearth of accessible and effective treatment options. [04] There is accordingly a clear need for novel treatment options to treat patients who cannot access or do not respond to current interventions, and which also lack the numerous drawbacks of those interventions, such as detrimental or dangerous side effects. Such options will be highly sought after, both to relieve the suffering of individual patients, as well as to reduce the societal burdens created by untreated and undertreated mental health conditions. [05] Providing these advancements and others, Applicant herein discloses formulations from fungi, Cannabis , and other natural, biosynthetic, and synthetic sources, comprising active compounds that together demonstrate a variety of synergistic and enhanced entourage effects, as demonstrated through the disclosure below. These formulations are useful for modulating neurotransmission, endocannabinoid system activity, and neurosteroid biosynthesis, for treating mental health conditions and other CNS disorders, and for improving mental health, in particular when used in a variety of distinct and uniquely designed daily and combination dosing regimens, as taught in different embodiments herein. [06] The medical benefits of Cannabis have been recognized for at least 4,750 years of recorded history. Across these millennia however, the hundreds of chemical compounds in the plant have resisted study in a systematic and comprehensive way. As this ancient therapeutic is revealed again, like a phoenix rising from the ashes of decades of vilification and illegality, its full potential will only be realized by combining the teachings of the past with the insights of current science, just as is occurring with psilocybin and Psilocybe mushrooms and the “psychedelic renaissance.” [07] Marrying their medical wisdom with their discoveries and innovations, the inventors herein bring forward and accelerate the natural evolution of these plant and fungi based medicines and modalities, providing the unique and novel contributions detailed below. INCORPORATION BY REFERENCE [08] Each patent, publication, and non-patent literature cited in the application or the section entitled References is hereby incorporated by reference in its entirety as if each was incorporated by reference individually. Unless specifically stated otherwise, reference is not to be construed as an admission that a document referred to or any underlying information in the document is prior art in any jurisdiction, or part of common general knowledge in the art. BRIEF SUMMARY OF THE INVENTION [09] The following presents a simplified summary of some embodiments of the invention in order to provide a basic understanding of the invention. This summary is not an extensive overview of the invention. It is not intended to identify key or critical elements of the invention or to delineate the scope of the invention. Its sole purpose is to present some embodiments of the invention in a simplified form as a prelude to the more detailed description that is presented later. [10] The present invention discloses formulations from mushrooms and Cannabis , including with synergistic and entourage enhanced combinations of tryptamines, cannabinoids, and other active agents, and which are useful in methods for treating CNS and mental health disorders and for improving mental health, for example when administered in the daily dosing regimens. Such compositions and methods have various improvements and advantages over current treatments, as will be readily appreciated from the disclosure herein. [11] In some aspects are disclosed methods of treating a CNS disorder patient in need thereof, comprising administering to the patient a mushroom composition and a second composition according to a defined dosing regimen. In embodiments, a defined dosing regimen is a daily dosing regimen consisting of a first number of days of administration of the mushroom composition, followed by a second number of days of administration of the second composition. In embodiments, the first number of days is between 2 and 7 days. In embodiments, the first number of days is between 2 and 5 days. In embodiments, the first number of days is either 3 or 4 days. In embodiments, the first number of days is 4 days. In embodiments, the second number of days is between 2 and 7 days. In embodiments, the second number of days is between 2 and 5 days. In embodiments, the second number of days is either 3 or 4 days. In embodiments, the second number of days is 3 days. [12] In embodiments, a patient is administered a psychedelic mushroom composition. In embodiments, the psychedelic mushroom composition comprises a psychedelic mushroom extract or a compound from a psychedelic mushroom. In embodiments, the psychedelic mushroom extract is a Psilocybe extract. In embodiments, the Psilocybe extract is an extract of Psilocybe cubensis . In embodiments, the psychedelic mushroom extract is an extract of a species within a fungal genera selected from the group consisting of Conocybe, Copelandia, Galerina, Gymnopilus, Inocybe, Mycena, Panaeolina, Panaeolus, Pholiotina , and Pluteus . In embodiments, the compound from a psychedelic mushroom is any of psilocybin, psilocin, norbaeocystin, baeocystin, aeruginascin, and norpsilocin. In embodiments, the compound from a psychedelic mushroom is produced by chemical synthesis or biosynthesis. In embodiments, the functional mushroom comprises a Hericium extract or a compound from Hericium . In embodiments, the compound from Hericium is any of any of Hericenone A, Hericenone B, Hericenone C, Hericenone D, Hericenone E, Hericenone F, Hericenone G, Hericenone H, Hericenone I, Hericenone J, Hericenone K, 3HF, DLPE, Isohericerinol A, Hericerin, NDPIH, and Corallocin A. In embodiments, the functional mushroom comprises a Cordyceps extract or a compound from Cordyceps . In embodiments, the compound from Cordyceps is any of cordycepin, cordycepic acid, N-acetylgalactosamine, adenosine, ergosterol, an ergosteryl ester, a bioxanthracene, hypoxanthine, a macrolide, a cicadapeptin, myriocin, superoxide dismutase, naphthoquinone, cordyheptapeptide, dipicolinic acid, a fibrinolytic enzyme, and cordymin. [13] In embodiments, a patient is administered a cannabis composition. In embodiments, the cannabis composition is an intoxicating cannabis composition. In embodiments, the intoxicating cannabis composition comprises THC. In embodiments, the THC is Δ9 -THC. In embodiments, the THC is Δ8-THC . In embodiments, the cannabis composition is a non-intoxicating cannabis composition. In embodiments, the non-intoxicating cannabis composition comprises CBD, and is free of THC. [14] In embodiments, a patient is administered a washout composition. In embodiments, the washout composition comprises an active compound such as amino acids, antioxidants, anti-inflammatory agents, analgesics, antineuropathic and antinociceptive agents, antimigraine agents, anxiolytics, antidepressants, antipsychotics, anti-PTSD agents, immunostimulants, anti-cancer agents, antiemetics, orexigenics, antiulcer agents, antihistamines, antihypertensives, anticonvulsants, antiepileptics, bronchodilators, neuroprotectants, entactogens and empathogens, entheogens, psychedelics, nootropics, monoamine oxidase inhibitors, sedatives, stimulants, supplements, and vitamins.
[15] In embodiments, a patient abstains from any administration of a composition of the invention. In embodiments, the days of abstinence are after the first number of days of administration of the mushroom composition, and before the second number of days of administration of the second composition. In embodiments, the days of abstinence are after the second number of days of administration of the second composition, and before a next administration of the mushroom composition. In embodiments, the days of abstinence are within the first number of days of administration of the mushroom composition. In embodiments, the days of abstinence are within the second number of days of administration of the second composition. In embodiments, the number of days of abstinence is between 2 and 7 days. In embodiments, the number of days of abstinence is between 2 and 5 days. In embodiments, the number of days of abstinence is either 3 or 4 days. In embodiments, a defined dosing regimen is repeated between 2 and 10 times to provide a therapeutic regime. In embodiments, the defined dosing regimen is repeated six times, seven times, or eight times. In embodiments, the daily dosing regimen is repeated between 2 and 10 times to provide a therapeutic regime. In embodiments, the daily dosing regimen is repeated six times, seven times, or eight times. In embodiments, the daily dosing regimen lasts for a period of six weeks, seven weeks, or eight weeks. In embodiments, the regime lasts from between 15 and 90 days. In embodiments, the regime lasts from between 30 and 70 days. In embodiments, the regime lasts from between 50 and 65 days. In embodiments, the regime lasts for 60 days. In embodiments, a therapeutic regime comprises at least two different defined dosing regimens, or three or more different daily dosing regimens of the invention.
[16] In some aspects are disclosed mushroom compositions for use in the methods of the invention, comprising synergistically and therapeutically effective amounts of: a Hericium extract or a compound from Hericium, and a Cordyceps extract or a compound from Cordyceps together with at least one carrier, diluent, or excipient, and wherein the at least one carrier, diluent, or excipient is not from a fungal source or found in fungi. [17] In embodiments, a mushroom composition further comprises a Psilocybe extract or a compound from Psilocybe. In embodiments, a mushroom composition further comprises one or more of niacin, valerian root extract, vitamin B12, and a CB1 receptor modulator. In embodiments, a mushroom composition further comprises two or more of niacin, valerian root extract, vitamin B12, and a CB1 receptor modulator. In embodiments, a mushroom composition further comprises three or more of niacin, valerian root extract, vitamin B12, and a CB1 receptor modulator. In embodiments, a mushroom composition further comprises niacin, valerian root extract, vitamin B12, and a CB1 receptor modulator. In embodiments, the CB1 receptor modulator is THC. In embodiments, the CB1 receptor modulator is CBD. In embodiments, the CB1 receptor modulator is PEA. In embodiments, a mushroom composition further comprises one or more of rice hull concentrate powder, rice bran extract powder, and rice bran 100 mesh powder. In embodiments, a mushroom composition further comprises a therapeutically effective amount of an additional active compound.
[18] In some aspects are disclosed cannabis compositions for use in the methods of the invention, comprising synergistically and therapeutically effective amounts of: cannabidiol (CBD); an entourage-enhancing cannabinoid fraction; and an entourage-enhancing isoprenoid fraction; together with at least one carrier, diluent, or excipient, and wherein the at least one carrier, diluent, or excipient is not from a plant source or found in plants.
[19] In embodiments, a cannabis composition further comprises THC. In embodiments, the THC is Δ9-THC. In embodiments, the THC is Δ8-THC. In embodiments, the CBD is natural CBD. In embodiments, the CBD is synthetic CBD. In embodiments, the ratio of THC:CBD is from about 1:20 to about 20:1. In embodiments, the ratio of THCCBD is between 1:20 to 1:10, between 1:10 and 1:5, between 1:5 and 1:2, between 1:2 and about 1:1, between about 1:1 and 2:1, between 2:1 and 5:1, between 5:1 and 10:1, and between 10:1 and 20:1. In embodiments, the ratio of THCCBD is about 1:4. In embodiments, the entourage-enhancing cannabinoid fraction comprises two or more cannabinoids. In embodiments, the cannabinoids in the entourage-enhancing cannabinoid fraction are natural. In embodiments, at least one of the natural cannabinoids is derived from a botanical source. In embodiments, the botanical source is a Cannabis plant. In embodiments, at least one of the cannabinoids in the entourage-enhancing cannabinoid fraction is synthetic. In embodiments, all of the cannabinoids in the entourage-enhancing cannabinoid fraction are synthetic. In embodiments, at least one of the cannabinoids in the entourage-enhancing cannabinoid fraction is selected for its ability to provide therapeutic effects or synergistic effects. In embodiments, at least one of the cannabinoids in the entourage-enhancing cannabinoid fraction is selected for its ability to modulate the endocannabinoid system, modulate neurosteroid biosynthesis, or modulate a neurotransmitter system. In embodiments, the entourage-enhancing cannabinoid fraction comprises at least two or more of: cannabichromene (CBC), cannabielsoin (CBE), cannabigerol (CBG), cannabicyclol (CBL), cannabinol (CBN), cannabidinodiol (CBND), cannabicitran (CBT), cannabitriol (CBT), cannabivarin (CBV), cannabigerol monomethyl ether (CBGM), cannabidivarin (CBDV), tetrahydrocannabivarin (THCV), cannabidiphorol (CBDP), tetrahydrocannabiphorol (THCP), and iso-tetrahydrocannabinol (iso-THC). In embodiments, the entourage-enhancing cannabinoid fraction comprises two or more cannabinoids independently selected from any of CBC-type cannabinoids, CBD-type cannabinoids, CBE-type cannabinoids, CBG-type cannabinoids, CBL-type cannabinoids, CBN-type cannabinoids, CBND-type cannabinoids, CBT-type cannabinoids, Δ8-THC -type cannabinoids, Δ9 -THC-type cannabinoids, and miscellaneous-type cannabinoids, as specifically disclosed below. [20] In embodiments, the entourage-enhancing isoprenoid fraction comprises two or more isoprenoids. In embodiments, all of the isoprenoids in the entourage-enhancing isoprenoid fraction are natural. In embodiments, at least one of the natural isoprenoids is derived from a botanical source. In embodiments, the botanical source is a Cannabis plant. In embodiments, at least one of the isoprenoids in the entourage-enhancing isoprenoid fraction is synthetic. In embodiments, all of the isoprenoids in the entourage-enhancing isoprenoid fraction are synthetic. In embodiments, at least one of the isoprenoids in the entourage-enhancing isoprenoid fraction is selected for its ability to provide therapeutic effects or synergistic effects. In embodiments, at least one of the isoprenoids in the entourage-enhancing isoprenoid fraction is selected for its ability to modulate the endocannabinoid system, modulate neurosteroid biosynthesis, or modulate a neurotransmitter system. In embodiments, two or more isoprenoids are selected from the group consisting of: alpha-bisabolol, beta-caryophyllene, camphene, carene, caryophyllene oxide, alpha-humulene, fenchol, guaiene, guaiol, limonene, linalool, myrcene, nerolidol, ocimene, alpha-phellandrene, alpha-pinene, beta-pinene, alpha-terpinene, gamma-terpinene, terpineol, and terpinolene. In embodiments, the entourage-enhancing isoprenoid fraction is myrcene dominant. In embodiments, the myrcene dominant entourage-enhancing isoprenoid fraction contains at least 20% myrcene by weight. In embodiments, the entourage enhancing isoprenoid fraction includes two or more isoprenoids independently selected from any of monoterpenes, sesquiterpenes, diterpenes, triterpenes, and miscellaneous terpenes, as disclosed below. [21] In embodiments, the entourage-enhancing cannabinoid fraction or the entourage-enhancing isoprenoid fraction provides a therapeutic effect, decreases an unwanted effect, increases stability or shelf-life, or alters pharmacokinetics or pharmacodynamics. In embodiments, the therapeutic effect is an antioxidant, anti-inflammatory, analgesic, antineuropathic, antinociceptive, antimigraine, anxiolytic, antidepressant, antipsychotic, anti-PTSD, immunostimulant, anti-cancer, antiemetic, orexigenic, antiulcer, antihistamine, antihypertensive, anticonvulsant, antiepileptic, bronchodilator, neuroprotective, entactogenic, empathogenic, entheogenic, psychedelic, sedative, or stimulant effect. In embodiments, the entourage-enhancing cannabinoid fraction or the entourage-enhancing isoprenoid fraction provides a synergistic effect. In embodiments, the synergistic effect is a greater than additive increase in potency, bioactivity, bioaccessibility, bioavailability, or therapeutic effect. In embodiments, the entourage-enhancing cannabinoid fraction or the entourage-enhancing isoprenoid fraction modulates the endocannabinoid system, modulates neurosteroid biosynthesis, or modulates a neurotransmitter system. In embodiments, the entourage-enhancing cannabinoid fraction comprises from 0.1% to about 25% of the composition by weight. In embodiments, the entourage-enhancing cannabinoid fraction comprises about 10% of the composition by weight. In embodiments, the entourage-enhancing isoprenoid fraction comprises from 0.1% to about 10% of the composition by weight. In embodiments, the entourage-enhancing isoprenoid fraction comprises about 5% of the composition by weight. In embodiments, the cannabis composition further comprises one or more flavonoids, as specifically disclosed below. [22] In embodiments, a cannabis composition of is formulated for any one of: (1) oral administration, and comprising a hydrophobic solvent or an alcohol-based solvent, and optionally a co-solvent; (2) use with a transdermal drug delivery system, wherein the composition is combined with a permeation enhancing agent, and optionally with one or more stabilizers, solubilizers, or adhesives; or (3) administration as a nanostructured formulation selected from the group consisting of: a nanoemulsion, a nanocapsule, a nanoparticle conjugate, or a nano-encapsulated oral or nasal spray. [23] In embodiments, a composition is prepared for administration as a formulation selected from the group consisting of: a soft or hard gelatin capsule, an oral tablet, a timed release capsule or tablet, a controlled release capsule or tablet, an extended release capsule or tablet, gastrorententive tablet, a sublingual or buccal tablet, an oral spray, a nasal spray, a suppository, a suspension, a solution suitable for subcutaneous, intravenous, intraperitoneal, or intramuscular administration, a liquid suitable for use in a liquid vaporization appliance, a topical preparation, or a transdermal patch. [24] In embodiments, a cannabis composition further comprises a therapeutically effective amount of an additional active compound. In embodiments, the additional active compound acts to increase therapeutic efficacy, provide additional therapeutic effects, decrease unwanted effects, increase stability or shelf-life, improve bioavailability, induce synergy, or alter pharmacokinetics or pharmacodynamics. In embodiments, the additional active compound is selected from the group consisting of: amino acids, antioxidants, anti-inflammatory agents, analgesics, antineuropathic and antinociceptive agents, antimigraine agents, anxiolytics, antidepressants, antipsychotics, anti-PTSD agents, immunostimulants, anti-cancer agents, antiemetics, orexigenics, antiulcer agents, antihistamines, antihypertensives, anticonvulsants, antiepileptics, bronchodilators, neuroprotectants, entactogens and empathogens, entheogens, psychedelics, nootropics, monoamine oxidase inhibitors, sedatives, stimulants, supplements, and vitamins. [25] In some aspects are disclosed unit dosage forms, useful to treat a mammal with a CNS disorder, and which comprise a therapeutically effective amount of: a functional mushroom composition; a psychedelic mushroom composition; a non-intoxicating cannabis composition; and an intoxicating cannabis composition, as well as a washout composition. In embodiments, the unit dosage form is suitable for oral, mucosal, rectal, injectable, intranasal, inhaled, or transdermal administration. In embodiments, the unit dosage form is an oral solid form or an oral liquid form. In embodiments, the unit dosage form comprises ground or pulverized Psilocybe fungal matter in a total amount of between 1 mg and 1 g. In embodiments, the unit dosage form comprises a Psilocybe extract in a total amount of between 0.5 mg and 500 mg. In embodiments, the unit dosage form comprises psilocybin in a total amount of between 0.01 mg and 10 mg. In embodiments, the unit dosage form comprises psilocybin in a total amount of between 0.5 mg and 3 mg. In embodiments, the unit dosage form comprises psilocybin in a total amount of 2.5 mg. In embodiments, the unit dosage form comprises psilocybin in a total amount of 1.25 mg. In embodiments, the unit dosage form comprises THC in a total amount of between 0.1 mg and 100 mg. In embodiments, the unit dosage form comprises THC in a total amount of between 2 mg and 50 mg. In embodiments, the unit dosage form comprises THC in a total amount of 10 mg. In embodiments, the unit dosage form comprises THC in a total amount of 5 mg. [26] In some aspects are disclosed methods of treating a mental health condition or improving mental health, in a patient in need thereof, comprising administering to the patient a unit dosage forms of the invention, according to a defined dosing regimen. In embodiments, the defined dosing regimen is a daily dosing regimen consisting of a first number of days of administration of one unit dosage form, followed by a second number of days of administration of a second unit dosage form. In embodiments, the first number of days is between 2 and 7 days. In embodiments, the first number of days is between 2 and 5 days. In embodiments, the first number of days is either 3 or 4 days. In embodiments, the first number of days is 4 days. In embodiments, the second number of days is between 2 and 7 days. In embodiments, the second number of days is between 2 and 5 days. In embodiments, the second number of days is either 3 or 4 days. In embodiments, the second number of days is 3 days. In embodiments, the defined dosing regimen is repeated between 2 and 10 times to provide a therapeutic regime. In embodiments, the defined dosing regimen is repeated six times, seven times, or eight times. In embodiments, the daily dosing regimen is repeated between 2 and 10 times to provide a therapeutic regime. In embodiments, the daily dosing regimen is repeated six times, seven times, or eight times. In embodiments, the daily dosing regimen lasts for a period of six weeks, seven weeks, or eight weeks. In embodiments, the regime lasts from between 15 and 90 days. In embodiments, the regime lasts from between 30 and 70 days. In embodiments, the regime lasts from between 50 and 65 days. In embodiments, the regime lasts for 60 days. [27] In some aspects are disclosed daily dosing kits according to the methods of the invention, comprising the unit dosage forms to be administered according to said methods, and in such amount as the number of total days of their administration. In embodiments, a daily dosing kit is provided as a blister pack, a blister tray, a blister card, a strip pack, a push-through pack, a compliance pack, or a calendar pack. [28] In some aspects are disclosed methods for modulating neurotransmission in a mammal in need thereof, comprising administering thereto a first unit dosage form together with a second unit dosage form, according to a defined dosing regimen. In embodiments, the neurotransmission is catecholaminergic, gabaminergic, glutamatergic, or serotonergic neurotransmission. In embodiments, the serotonergic neurotransmission is a modulation of 5-HT 2A receptors, without a significant modulation of 5-HT 2B receptors. In embodiments, the serotonergic neurotransmission is a modulation of 5-HT 1A and 5-HT 2A receptors. [29] In some aspects are disclosed methods for modulating endocannabinoid system activity in a mammal in need of such modulation, the method comprising administering to the mammal a first unit dosage form together with a second unit dosage form, according to a defined dosing regimen. In some aspects are disclosed methods for modulating biosynthesis of a neuroactive steroid in a mammal in need of such modulation, the method comprising administering to the mammal a first unit dosage form together with a second unit dosage form, according to a defined dosing regimen. In some aspects are disclosed methods for treating a medical condition in a mammal in need of such treatment, including in a human, the method comprising administering to the mammal a first unit dosage form together with a second unit dosage form, according to a defined dosing regimen. In embodiments, the medical condition is a disorder linked to dysregulation or inadequate functioning of neurotransmission. In embodiments, the disorder linked to dysregulation or inadequate functioning of neurotransmission is that of catecholaminergic, gabaminergic, glutamatergic, or serotonergic neurotransmission. In embodiments, the disorder linked to dysregulation or inadequate functioning of neurotransmission is that of serotonergic neurotransmission. In embodiments, the medical condition is a disorder linked to dysregulation or inadequate functioning of the endocannabinoid system. In embodiments, the medical condition is a disorder linked to dysregulation or inadequate functioning neurosteroid biosynthesis.
[30] In embodiments, the medical condition treated by the invention is a mental health condition. In embodiments, the mental health condition is selected from the group consisting of: post-traumatic stress disorder (PTSD), adjustment disorder, affective disorder, depression, dysthymia, anxiety, phobia disorders, binge disorders, body dysmorphic disorder, alcohol or drug abuse or dependence disorders, disruptive behavior disorders, eating disorders, impulse control disorders, obsessive compulsive disorder (OCD), attention deficit hyperactivity disorder (ADHD), personality disorders, attachment disorders, and dissociative disorders. In embodiments, the mental health condition is a disorder related to rigid modes of thinking. In embodiments, the disorder related to rigid modes of thinking is an anxiety disorder, a depressive disorder, addiction, an eating disorder, OCD, or PTSD.
[31] In some aspects are disclosed methods of reducing the symptoms of a mental health condition in a human, the method comprising identifying a human in need of said reducing, and administering to the human a first unit dosage form of any of claims 134-149 together with a second unit dosage form of any of claims 134-149, according to a defined dosing regimen. In embodiments, the mental health condition is PTSD. In embodiments, the symptoms of PTSD that are reduced include one or more of flashbacks, nightmares, distressing and intense memories, distress or physical reactions after being exposed to triggers, blaming self or others for the trauma, decreased interest in things that were once enjoyable, negative feelings about self and the world, inability to remember the trauma clearly, difficulty feeling positive, feelings of isolation, negative affect, difficulty feeling positive, avoidance, aggression or irritability, hypervigilance and hyper-awareness, difficulty concentrating, difficulty sleeping, heightened startle response, engaging in self-destructive, or risky behavior, difficulty sleeping or staying asleep, or suicidal ideation. [32] In some aspects are disclosed methods of improving psychological functioning in a human, the method comprising identifying a human in need of said improving, and administering to the human a first unit dosage form together with a second unit dosage form, according to a defined dosing regimen, and wherein the improvement in psychological functioning is a reduction of neuroticism or psychological defensiveness, an increase in creativity or openness to experience, an increase in decision-making ability, an increase in feelings of wellness or satisfaction, or an increase in ability to fall or stay asleep. In some aspects are disclosed methods of treating a CNS disorder in a human patient, the method comprising administering to the patient a first unit dosage form together with a second unit dosage form, according to a defined dosing regimen, and in combination with one or more psychotherapy sessions, or in conjunction with participation in a therapeutically beneficial self-care activity which is chosen from the group consisting of: breathing exercises, meditation and concentration practices, focusing on an object or mantra, listening to music, physical exercise, stretching or bodywork, journaling, grounding techniques, positive self-talk, or engaging with a pet or animal. [33] The foregoing has outlined rather broadly the more pertinent and important features of the present disclosure so that the detailed description of the invention that follows may be better understood and so that the present contribution to the art can be more fully appreciated. Additional features of the invention will be described hereinafter which form the subject of the claims of the invention. It should be appreciated by those skilled in the art that the conception and the disclosed specific methods and structures may be readily utilized as a basis for modifying or designing other structures for carrying out the same purposes of the present disclosure. It should be also realized that such equivalent structures do not depart from the spirit and scope of the invention as set forth in the appended claims. Hence, this summary has been made with the understanding that it is to be considered as a brief and general synopsis of only some of the objects and embodiments disclosed herein, is provided solely for the benefit and convenience of the reader, and is not intended to limit in any manner the scope, or range of equivalents, to which the claims are lawfully entitled. BRIEF SUMMARY OF THE DRAWINGS [34] To further clarify various aspects of the invention, a more particular description of the invention will be rendered by reference to certain exemplary embodiments thereof which are illustrated in the included figures. It should be understood and appreciated that the figures depict only illustrated embodiments of the invention and are therefore not to be considered limiting of its scope. They are simply provided as exemplary illustrations of certain concepts of some embodiments of the invention. [35] Certain aspects of the invention are therefore further described and explained with additional specificity and detail, but still by way of example only, with reference to the accompanying figures in which: [36] FIG.1 illustrates a number of exemplary daily dosing kits containing a 21-day supply of the mushroom and cannabis compositions of the invention, according to embodiments. [37] FIG.2 illustrates a number of exemplary daily dosing kits containing a 21-day supply of the mushroom and cannabis compositions of the invention, according to embodiments, and additionally includes abstinent days, wherein no dosing is completed. [38] FIG.3 illustrates a number of exemplary daily dosing kits containing a 21-day supply of certain mushroom and cannabis compositions of the invention according to certain embodiments, wherein a “M+” mushroom composition of EXAMPLE 2 or EXAMPLE 3 is administered for four consecutive days, followed by any combination of administration of any of: (1) a mushroom composition that does not contain psilocybin (“M–”); (2) a cannabis composition containing THC (“C+”); a cannabis composition that does not contain THC (“C–”); and abstinence or “washout” (“A”), for the next three consecutive days. DETAILED DESCRIPTION [39] Among the various aspects of the present invention are formulations of compounds from mushrooms and Cannabis , including with synergistic and entourage enhanced combinations of tryptamines, cannabinoids, and other active agents, and which are useful in methods for treating CNS disorders and mental health conditions and for improving mental health, for example when administered in daily dosing regimens and therapeutic regimes. [40] While the present invention is described in terms of particular embodiments and applications, it is not intended that these descriptions in any way limit its scope to any such embodiments and applications, and it will be understood that many modifications, substitutions, changes, and variations in the described embodiments, applications, and details of the invention illustrated herein can be made by those skilled in the art without departing from the spirit of the invention, or the scope of the invention as described in the appended claims. The headings within this document are being utilized only to expedite its review by a reader. They should not be construed as limiting the present invention in any manner. The full scope of the invention shall be properly determined by reference to the appended claims. A. Definitions and Terms [41] The terminology used herein is for describing embodiments and is not intended to be limiting. As used herein, the singular forms “a,” “an,” “and,” “the,” and “said” mean that there are one or more of the elements, and hence include plural referents, unless the content and context clearly dictate otherwise. Thus, for example, a reference to “an extract” or “a compound” may include a single such extract or compound, or a combination of two or more. [42] The terms “comprising,” “including,” “such as,” and “having” are intended to be inclusive and not exclusive (i.e., there may be other elements in addition to the recited elements). Thus, the term “including” as used herein means, and is used interchangeably with, the phrase “including but not limited to.” The term “or” is used herein to mean, and is used interchangeably with, the term “and/or,” unless context clearly indicates otherwise. [43] Unless otherwise indicated, all numbers expressing quantities of ingredients, properties such as concentration, reaction conditions, and so forth, used to describe and claim certain embodiments of the invention are to be understood as being modified in some instances by the term “about.” Accordingly, in some embodiments, the numerical parameters set forth in the description and claims are approximations that can vary depending upon the desired properties sought to be obtained by a particular embodiment. In embodiments, “about” refers to plus or minus five percent (±5%) of the recited unit of measure. [44] The term “substantially,” when applied to modify a parameter or characteristic herein, will be read in the context of the invention and in light of the knowledge in the art to provide certainty, e.g., by using a standard that is recognized in the art for measuring the meaning of substantially as a term of degree, or by ascertaining the scope as would one of skill. [45] In embodiments, the numerical parameters should be construed in light of the number of reported significant digits and by applying ordinary rounding techniques. Notwithstanding that the numerical ranges and parameters setting forth the broad scope of some embodiments of the invention are approximations, the numerical values set forth in the specific examples are reported as precisely as practicable. The numerical values presented in some embodiments of the invention may contain certain errors necessarily resulting from the standard deviation found in their respective testing measurements. [46] Unless defined otherwise, all technical and scientific terms herein have the meaning as commonly understood by a person having ordinary skill in the art to which this invention belongs, who as a shorthand may be referred to simply as “one of skill.” [47] Further definitions that may assist the reader in understanding the disclosed embodiments are as follows; however, it will be appreciated that they will not be used to limit the scope of the invention, which shall be properly interpreted and understood by reference to the full specification (as well as any plain meaning known to one of skill in the relevant art) in view of the language used in the appended claims. That is, the terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting. [48] Generally, the nomenclature used and procedures performed herein are those known in field(s) relating to that of one or more aspects of the invention, and are those that will be well-known and commonly employed in such field(s). Standard techniques and procedures will be those generally performed according to conventional methods in the art. [49] Terms having a specific meaning within the regulatory law of a jurisdiction in which this patent was filed or may be in force generally should be given such meaning unless context dictates otherwise. For example, “Botanical Drug Substance” may refer to the term as defined through FDA implementing rules and regulations, and as described in the Guidance for Industry Botanical Drug Products, December 2016 (Docket No. FDA-2000-D-0103), U.S. Department of Health and Human Services, Food and Drug Administration Center for Drug Evaluation and Research (“2016 FDA Botanical Drug Guidance”). Terms including “Botanical Drug Product” and “Botanical Raw Materials” shall be known similarly. [50] For avoidance of doubt, even though “botanical” is commonly used to mean “relating to plants,” for purposes herein “botanical” as in a “botanical” drug substance or drug product will be understood to include substances and products (e.g., extracts) from fungal material as well, such as psilocybin-containing or other mushrooms, for example as defined by FDA. Where a substance or product is from fungal material, it may be from any part of the fruiting body (commonly referred to as a “mushroom”), regardless of whether fruiting occurs above or below ground (as with “truffles”), or from mycelia or other material. “Mushroom” will be used throughout this description to mean and to be the equivalent of terms like “fungi” or “fungal,” to the extent those terms would be broader. A “mushroom composition” thus should not be interpreted as being narrower than a “fungi composition” or a “fungal composition.” [51] A “mushroom composition” may be either an “M+” composition or an “M-” composition. An “M+” composition, which also may be referred to as a “psychedelic mushroom composition” will contain at least one compound known to produce or capable of producing “psychedelic effects” when ingested in humans, or that demonstrates such capabilities (e.g., by showing a head-twitch response or “HTR” in rodents). For clarity and avoidance of doubt, it will be understood that an “M+” composition may be a “psychedelic mushroom composition” even if it does not or cannot produce psychedelic effects when ingested or administered at the dosages of or according to the methods of the invention, and indeed, the lack of psychedelic effects in embodiments will be preferred, and it is therefore referred to as such because it comprises certain psychedelic compounds, not because those compounds produce psychedelic effects.
[52] For example, a mushroom composition comprising psilocybin, psilocin, and other psychedelic tryptamines (referred to together as “compounds from a psychedelic mushroom”) will be an “M+” or “psychedelic” mushroom composition, as will a composition comprising an extract from a Psilocybe or other psilocybin-containing mushroom. A “M-” composition, by contrast, will be a mushroom composition that does not contain any compound known to produce or capable of producing psychedelic effects when ingested in humans, or that demonstrates such capabilities. An “M-” composition may also be referred to as a “functional mushroom composition.” A functional mushroom composition, for example, may comprise extracts from lion’s mane and/or cordyceps, but not Psilocybe mushrooms. Where a functional mushroom composition comprises merely trace amounts of a psilocybin or a psychedelic mushroom (e.g., less than 1% w/w, less than 0.5% w/w, less than 0.1% w/w, less than 0.05% w/w, less than 0.01% w/w, or less than 0.005% w/w of the mushroom composition), which may occur, for example, because of shared manufacturing equipment, it will nonetheless be considered an “M-” or “functional” mushroom composition, and may further be considered a mushroom composition “substantially without psychoactive compounds” or “substantially free of’ psychoactive compounds, or “substantially” without or free of one or more specific psychedelic compounds, such as psilocybin or psilocin.
[53] A “cannabis composition” may be either a “C+” composition or a “C-” composition. An “C+” composition, which also may be referred to as an “intoxicating cannabis composition” or a “cannabis composition with THC” will contain at least one cannabinoid known to produce or capable of producing “intoxicating effects” when ingested in humans, or that demonstrates such capabilities. By contrast, cannabis compositions not comprising a cannabinoid known to produce or capable of producing “intoxicating effects” when ingested in humans, or that demonstrates such capabilities, will be referred to as a “C-” composition or a “non-intoxicating cannabis composition” or a “cannabis composition without THC.” In some preferred embodiments, the “intoxicating” compound will be a tetrahydrocannabinol (e.g., Δ9-THC. Δ8-THC , or another intoxicating THC compound). For avoidance of confusion, C+ compositions are referred to as “intoxicating” instead of “psychoactive” because some compounds such as CBD that can be considered psychoactive, nevertheless do not produce the “high” associated with Cannabis. In embodiments, it will be understood that a “C+” composition may be an “intoxicating cannabis composition” even if it does not or cannot produce intoxicating effects or produce a “high” when ingested or administered at the dosages of or according to the methods of the invention, or does not for a specific individual. In embodiments, the inclusion of THC may be to balance the entourage effects of other cannabinoids or other compounds in the cannabis composition. In other embodiments, the inclusion of THC may be for purposes of causing at least some degree of intoxication or high, such as the inclusion of about 5 mg of THC or more, or an equivalent amount of another intoxicating cannabinoid, based on relative potency. Depending on embodiment and individual, an intoxicating amount may be less than or more than 5 mg, but the amount that may cause intoxication will be readily appreciated and readily determinable both by those of ordinary skill as well as by subjects seeking to either obtain or avoid such effects in the self-administration of the compositions of the invention, or by the practice of the methods of the invention, such as when intoxication is desired, or when it is sought to be avoided, for example when it might interfere with one or more daily activities during the course of a daily dosing regimen. In embodiments, a subject will administer a daily dosing regimen specifically selected or timed so that days of administration of intoxicating cannabis compositions, where such compositions produce intoxicating effects, will be on specific days, such as on weekend days. Where an intoxicating cannabis compositions comprises merely trace amounts of THC or an intoxicating cannabinoid (e.g., less than 1% w/w, less than 0.5% w/w, less than 0.1% w/w, less than 0.05% w/w, less than 0.01% w/w, or less than 0.005% w/w of the cannabis composition), which may occur, for example, because of shared manufacturing equipment, it will nonetheless be considered an “C-” or “non-intoxicating” cannabis composition, and may further be considered a cannabis composition “substantially without THC” or “substantially free of’ THC.
[54] Botanical drug substances and products may be available by prescription or over-the-counter (“OTC”), or as nutritional or dietary supplements, or under any other regulatory regime; they also may be unregulated (e.g., “natural products”). [55] “ Cannabis ” or “Cannabis plant” may refer to wild-type C. sativa , C. indica , and C. ruderalis , as well as genetic crosses, self-crosses, hybrids, variants, and chemovars thereof. [56] “Cannabis-derived drug substance” may refer to a botanical drug substance which is derived from cannabis plants (including plant parts, plant part biomass, and plant exudates), as non-limiting, purely illustrative examples, primary extracts prepared by processes including maceration, percolation, extraction with solvents such as C1–C5 alcohols (e.g., ethanol), hydrocarbons (e.g., propane, butane), and subcritical or supercritical carbon dioxide. [57] “Cannabis-derived drug product” may refer to a primary cannabis extract that is further purified, for example by distillation or chromatography. It will be known to those of skill that when certain solvents are used to prepare primary extracts, the resultant extract may contain non-specific lipid-soluble material. Those of skill will know that such impurities can be removed by a variety of processes including winterization (e.g., chilling to −20° C followed by filtration to remove waxy ballast), further extraction or filtration, and distillation. [58] “CB 1 receptor modulator” may refer to a compound which is a ligand of and thus binds to and modulates the cannabinoid type 1 (CB 1 ) receptor. CB 1 receptor modulators include positive and negative allosteric modulators which bind to the receptor and change the conformation of its binding site, without also acting as an orthosteric agonist or antagonist. Allosteric CB 1 receptor modulators may treat CNS disorders without some adverse effects caused by orthosteric binding. Negative allosteric modulators of CB 1 include cannabidiol (CBD) and N -palmitoylethanolamine (PEA). Additional allosteric modulators of CB 1 , as well as orthosteric modulators, will be known to those of skill ( see, e.g., Laprairie et al., 2015). [59] “Extract” may refer to a botanical extract (e.g., a psilocybin-containing mushroom extract, a lion’s mane extract, a cordyceps extract, or a valerian root extract) prepared from a botanical (including fungal) source that does not undergo isolation, purification, or other processing to obtain specific active compounds separate from other constituents (i.e., is obtained as a whole-plant medicine). As a non-limiting, illustrative example, a fungal extract is prepared by taking lyophilized, dehydrated, or desiccated mushrooms or other mycelial or fungal matter (first drying such matter, if obtained fresh), and pulverizing or grinding it into a fine powder, and optionally passing it through a sieve or fine mesh followed by repeated grinding of remaining material, to ensure a preparation having uniformity of particle size. [60] “Purified extract” may refer to a botanical extract that has undergone further processing after preparation, such as soaking or heating the preparation in water and/or alcohol (depending on whether the compounds sought are water soluble), agitating, cooling the resulting liquid, straining, filtering, and removing unwanted products (repeating if necessary), and then evaporating sufficient liquid solvent to obtain a desired concentration (or entirely, to obtain a crystalline precipitate), or using a spray dryer to create a purified dried powder. A starch or other carrier subsequently can be added to maintain the purified dried powder as a free-flowing powder that is easy to work with during formulation, for instance if the dried powder will be added to capsules; however, when referring to the weight of a “purified extract,” any such carrier, diluent, or excipient is excluded from the total amount. [61] Extracts in some embodiments may be “standardized,” which refers to extracts that include one or more desired compounds (e.g., psilocybin) in a specific concentration. Methods to produce standardized extracts, such as by quantifying the potency of an extract and then adding excipient to dilute the extract to a standardized concentration, or further concentrating an extract to increase its potency, will be well known to those of skill. [62] “Natural” may refer to a substance which was isolated, extracted, or otherwise obtained from a natural source, such as a plant or fungus. A natural compound, such as a natural cannabinoid, a natural isoprenoid (or natural terpene), or a natural flavonoid, would therefore be within the scope of the invention if it was isolated or extracted from a Cannabis plant, or from a Psilocybe spp. or other mushroom (i.e., as a Botanical drug substance or drug product). Thus, in various embodiments of the invention, the compositions shall be derived from botanical sources including Cannabis plants and mushrooms, as extracts or by other means, and shall comprise one or more Botanical drug substances or Cannabis -derived drug substances. In embodiments, such compositions shall comprise one or more Botanical drug products or Cannabis -derived drug products. In other embodiments, such compositions shall comprise one or more Botanical drug products or Cannabis -derived drug products which are substantially free from impurities. [63] Methods of extracting cannabinoids, isoprenoids, and flavonoids from Cannabis and other plants, and creating plant and fungal extracts, and methods of obtaining purified products containing desired compounds free from undesired plant or fungal matter, chemicals, and other impurities, are known in the art and described in, e.g., U.S. Pat. Nos. 6,403,126, 8,846,409, 8,895,078, 10,059,684, 10,239,808, 10,246,431, 10,300,494, 10,307,447, 10,406,453, 10,413,845, and 10,414,709; and U.S. App. Nos.2003/0017216A1 and 2016/0038437A1; including references cited, or are otherwise within ordinary skill. [64] “Synthetic” may refer to a substance which is manufactured artificially in a laboratory, by means of chemical synthesis (e.g., by a series of chemical processes or reactions using chemical substrates, reagents, and optionally one or more catalysts) or biosynthesis (e.g., from a bioengineered organism, and thus including those compounds also referred to as “biosynthetic” or as involving “synthetic biology” or “synbio”). (“Natural” will be understood to therefore exclude such compounds.) For example, yeast, E. coli, or other host cells can be bioengineered to produce synthetic psilocybin or other compounds by inserting genes that produce appropriate enzymes and/or by altering the natural metabolic pathway to achieve the production of the desired compounds from one or more precursors. (See PCT Pub. No. W02019/180309A1.) Compounds can then be obtained and purified. Through such means, compounds may be obtained that are the same as natural compounds from plants or fungi, or that are derivatives or analogs thereof; examples are provided below.
[65] “Nutraceutical” may refer to a preparation that could be marketed as a dietary supplement (sometimes called a nutritional supplement), e.g., in the U.S. under the appropriate regulations of the Federal Food, Drug, and Cosmetic Act (FDCA) (22 U.S.C. §§ 301 et seq.) and Dietary Supplement Health and Education Act (DSHEA) of 1994. A dietary supplement is a product taken by mouth that contains a dietary ingredient intended to supplement the diet. The dietary ingredients in these products may include: vitamins, minerals, herbs or other botanicals, amino acids, and substances such as enzymes, organ tissues, glandulars, and metabolites. Dietary supplements can also be extracts or concentrates, and may be found in many forms such as tablets, capsules, softgels, gelcaps, liquids or powders. Although “nutraceuticals” could be marketed as dietary supplements, products need not in fact meet any specific regulatory standards (such as under DSHEA or other FDA regulations), or be considered under any specific regulatory standards to be considered nutraceuticals for purposes of the definition herein. Thus, it will be appreciated that within the definition of nutraceuticals are also products that are sold as “natural products,” or otherwise outside of any specific regulatory regime. Also within the meaning of nutraceuticals are “cosmeceuticals” which, although without legal definition under the FDCA, are understood to refer to cosmetic products that have medicinal or drug-like benefits.
[66] Although “pharmaceutical” and “nutraceutical” may be used interchangeably herein with regard to compositions of the invention, it will be appreciated that depending on the regulatory regime in which such compositions are available to an individual, a disclosed embodiment may or may not need to be prescribed or “recommended” by a doctor or medical professional, may or may not be available for purchase at a “dispensary” or other state or local jurisdiction licensed facility, and may or may not be able to be self-administered, including with or without supervision. Embodiments, for example, may include (depending on the regulatory regime) a dosing regimen of a psilocybin-comprising mushroom composition, which is prescribed by a doctor or received under the advice or care of a medical or healthcare professional, followed by a cannabis composition which may be obtained at a dispensary or otherwise over the counter, or without medical prescription. [67] “Psychedelic effects” refer to subjective alterations in perception, cognition, emotion, or consciousness that can affect and usually interfere with the ability to perform everyday tasks and activities. Although a determination of psychedelic effects can be made by a subject following consumption of a composition of the invention, such determination also can be made by use of or by reference to psychometric rating scales such as those known in the art, e.g., the Hallucinogen Rating Scale (HRS), Mystical Experience Questionnaire (MEQ), and Addiction Research Center Inventory (ARCI) ( see Bouso et al., 2016). [68] “Psychoactive” may refer to a substance that is capable of changing nervous system function or causing short- or long-term alterations in perception, mood, consciousness, cognition, or behavior. Unlike psychedelic effects, “psychoactive effects” may be present without noticeable subjective (inward) or objective (outward) alterations or changes, or feelings of “intoxication.” Accordingly, to distinguish between cannabinoids which can produce a subjective feeling of “being high,” such as ∆ 9 -THC and certain of its analogs like ∆ 8 -THC, the term “intoxicating” generally will be used instead of “psychoactive.” [69] The term “prodrug” may refer to a precursor of a biologically active pharmaceutical agent. Prodrugs undergo a chemical or a metabolic conversion to become a biologically active pharmaceutical agent. A prodrug can be converted ex vivo to the biologically active pharmaceutical agent by chemical transformative processes. In vivo, a prodrug is converted to the biologically active pharmaceutical agent by the action of a metabolic process, an enzymatic process or a degradative process that removes the prodrug moiety, such as a glycoside, to form the biologically active pharmaceutical agent. The compounds of the invention include the prodrugs thereof. For example, psilocybin is a prodrug of psilocin, but other compounds, e.g., those which are also prodrugs of psilocybin, such as psilacetin (or such other compounds used in the compositions of the invention), will be understood to be prodrugs within the meaning herein, along with prodrugs of other compounds, including cannabinoids (e.g., with THC-O-acetate, a prodrug of THC), and known prodrugs of CBD. [70] The terms “isolated,” “purified,” “substantially pure,” or “biologically pure” may refer to material that is substantially or essentially free from components that normally accompany the material in its raw, untreated, or native state or when the material is produced. A “substantially pure” or “isolated” preparation of a compound may accordingly be defined as a preparation having a chromatographic purity (of the desired compound) of greater than 90%, more preferably greater than 95%, more preferably greater than 96%, more preferably greater than 97%, more preferably greater than 98%, more preferably greater than 99% and most preferably greater than 99.5%, as determined by area normalization of an HPLC profile or other similar detection method. Preferably the substantially pure compound used in the invention is itself substantially free of any other active compounds, which may cause effect in a subject when administered, but which are not intended to be administered to the subject. In this context “substantially free” can be taken to mean that no compounds other than the target compound are detectable by HPLC or other similar detection method. [71] This disclosure is not limited to particular embodiments described as such may vary. The terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting. Although any materials and methods similar or equivalent to those described herein can be used in the practice or testing of the present invention, certain preferred materials and methods will be described. B. Current Treatment Options for Mental Health Conditions [72] In embodiments herein, the treatment options provided by the compositions and methods of the invention will be better than one or more current treatment options for mental health conditions available to patients. Such current treatment options consist generally of psychotherapy, pharmacotherapy, and direct brain intervention. All suffer serious drawbacks. [73] In embodiments herein, the compositions and methods of the invention will be better than psychotherapy. Psychotherapy takes various forms including cognitive behavioral therapy (CBT), exposure therapy, eye movement desensitization and reprocessing (EMDR), and numerous forms of talk therapy (often, paired together with a pharmacological agent). Psychotherapy frequently fails to provide complete or even partial symptom reduction. For example, CBT is fully effective for only a third of patients with mood disorders, with another third receiving partial benefit, and a last third receiving no benefit at all. Even for those who receive benefit, psychotherapy must be continued for years or decades. In long-term studies of patients using psychotherapy to deal with symptoms of post-traumatic stress disorder (PTSD), only 10-15 percent of patients were considered to have fully recovered; the remainder either dropped out or required further treatment. Because of the extended length of time typically required to see a clinically significant benefit, and because it is usually received in a one-on-one setting with a clinical professional, psychotherapy for many patients is difficult or prohibitively expensive to access or obtain consistently. In embodiments herein, the compositions and methods of the invention will be no worse than, at least as good as, and preferably better than, psychotherapy. In embodiments herein, the compositions and methods of the invention will be no worse than, at least as good as, and preferably better than, CBT, exposure therapy, and/or EMDR. “No worse than,” “at least as good as,” and “better than” may be determined, for example, by use of a non-inferiority trial, an equivalence trial, or a superiority trial, as known in the art and described in Examples herein. [74] In embodiments herein, the compositions and methods of the invention will be provided in conjunction with psychotherapy, and will be no worse than, at least as good as, and preferably better than, psychotherapy alone; or no worse than, at least as good as, and preferably better than, psychotherapy in conjunction with known pharmacotherapies, such as SSRIs or other antidepressants, or other pharmacological treatments. [75] With pharmacological treatments of the current state of the art, although some patients benefit, many patients find only partial relief, and many no relief at all. And for many, while relief may come, it comes accompanied by unacceptable medication-related side effects. In embodiments herein, the treatment options provided by the compositions and methods of the invention will be better than one or more pharmacological treatment options for mental health conditions available to patients. In embodiments herein, the compositions and methods of the invention will be no worse than, at least as good as, and preferably better than, one or more “first-line” pharmacological treatment options, including for mental health conditions such as depression, panic and anxiety disorders, obsessive compulsive disorder, phobias, bulimia, adjustment disorder, and PTSD. In embodiments herein, the compositions and methods of the invention will be no worse than, at least as good as, and preferably better than, use of selective serotonin reuptake inhibitors (SSRIs), which block the reabsorption (i.e., reuptake) of serotonin into neurons, thereby increasing levels of serotonin in the brain. Even for those patients who respond to SSRIs, the drugs are generally slow to achieve meaningful benefit, requiring weeks or months to produce therapeutic effects, while also significantly increasing the risk of serious adverse events. Moreover, once prescribed, patients generally continue to take them for their entire lives. A recent review has concluded that for most patients the “potential small beneficial effects [of SSRIs] seem to be outweighed by harmful effects” (Jackobsen et al., 2017). In embodiments herein, the compositions and methods of the invention will be no worse than, at least as good as, and preferably better than, SSRIs. [76] Another broad class of agents for treatment of mental health conditions is monoamine releasers, which induce the release of monoamine neurotransmitters (e.g., dopamine, serotonin, or epinephrine) from neurons. Monoamine releasers rapidly modulate the brain systems that are more gradually affected by SSRIs. However, their stimulant and euphoric effects create high abuse liability. Although psychostimulants in this class were widely employed in the mid-20th century to treat affective disorders, OCD, and schizophrenia, use of such agents (e.g., amphetamines) is now primarily limited to treating attention deficit hyperactivity disorder (ADHD), and even this use is increasingly restricted. In embodiments herein, the compositions and methods of the invention will be no worse than, at least as good as, and preferably better than, psychostimulants and other monoamine releasers. [77] Various FDA-approved drugs are also prescribed to treat symptoms of mental health conditions “off label” (i.e., for unapproved indications). Prazosin (Minipress ® ), for instance, is an alpha-blocker and antihypertensive often used off label for PTSD-related nightmares and sleep disruption. However, evidence supporting off label treatments is sparse, and they are not widely considered to be effective for most patients. In embodiments herein, the compositions and methods of the invention will be no worse than, at least as good as, and preferably better than, current off label treatments for mental health conditions. [78] A last resort for patients with serious or refractory (i.e., treatment resistant) conditions is direct brain interventions. For instance, one option when other treatments are unsuccessful is electroconvulsive therapy (ECT). ECT is a procedure, done under general anesthesia, in which electric currents are passed through the brain, intentionally triggering a brief seizure. Various surgical procedures also have been developed. These primarily involve targeted ablative procedures, i.e., causing the irreversible destruction of specific brain tissue. Others involve implantation of devices directly into the brain, an example being “deep brain stimulation,” where electrodes are inserted into a specific brain region to generate electrical pulses. But like psychotherapeutic and pharmacotherapeutic interventions, neurosurgical interventions are only successful in a minority of patients; more significantly, they are also associated with serious adverse events including changes in personality, development of epilepsy, and the risks associated with surgery itself, such as hemorrhage, infection, and death. In embodiments herein, the compositions and methods of the invention will be no worse than, at least as good as, and preferably better than, direct brain interventions like ECT, surgical procedures such as targeted ablative procedures, and deep brain stimulation. [79] In human clinical studies currently underway, evidence has started to accumulate demonstrating that certain tryptamine agents that engage serotonin receptors directly show promise of rapid and long-lasting therapeutic effects in treating mental health conditions following only limited doses. For instance, studies using the Schedule I controlled drug psilocybin (the active ingredient in “magic” mushrooms) have shown promise in rapidly and effectively treating mental health conditions such as AUD, TRD, and PTSD, when taken in combination with psychotherapy (“psychedelic-assisted psychotherapy”). [80] However, such treatment can produce significant side effects. Psilocybin, at psychedelic dosages, causes hallucinations and distortions of perception, cognition, emotion, and consciousness. These effects, and their often unpredictable consequences, renders the drug unsuitable for many patients, and difficult to use without supervision. Psilocybin-assisted therapy, moreover, generally requires two trained therapists or facilitators to be present during a drug administration session for at least 6-8 hours. Although efforts are underway to shorten the duration of “psychedelic” or “hallucinogenic” effects, thus reducing or eliminating the need for lengthy supervision, as well as to improve the efficacy of known drugs like psilocybin and reduce its deleterious side effects, whether derivatives or analogs will have such therapeutic benefits remains to be seen. And if use is ultimately restricted to psychedelic-assisted psychotherapy, adoption may be limited. In embodiments herein, the compositions and methods of the invention will be no worse than, at least as good as, and preferably better than, psilocybin-assisted and other psychedelic-assisted psychotherapy. C. Psilocybin and its History [81] Psilocybin is a naturally occurring compound produced by several hundred species of fungus, most from the genus Psilocybe , but also from a number of other genera. Psilocybin acts as a prodrug in animals, and is rapidly converted to psilocin. Taken at certain dosages, psilocybin causes mind-altering “psychedelic” effects such as euphoria, visual and mental hallucinations, changes in consciousness, a distorted sense of time, and experiences labeled as numinous, mystical, or “spiritual,” even by those who do not profess to be religious. [82] The term “psychedelic” was coined in 1953 by the psychiatrist Humphrey Osmond, during written correspondence with author Aldous Huxley. Psychedelic derives from two Ancient Greek words, psyche meaning “mind” or “soul,” and delos meaning “reveal,” or “manifest.” At psychedelic dosages, psilocybin also can cause possible adverse reactions such as nausea, panic attacks, and negative or disturbing experiences known as “bad trips.” [83] Evidence is cited to suggest that psychoactive mushrooms containing psilocybin have been used by humans in religious ceremonies for thousands of years. Murals dated 9000 to 7000 BCE found in the Sahara desert in southeast Algeria depict what have been asserted to be mushrooms used in such ceremonies, and archaeological artifacts from Central and South America, including the “mushroom stones” of Guatemala have been interpreted by some scholars as evidence for ritual usage of psilocybin-containing mushrooms in the Mayan and Aztec cultures of Mesoamerica. In his book “Food of the Gods,” Terrence McKenna in fact proposed that the transformation from humans’ early ancestors Homo erectus to the species Homo sapiens was a direct result of the addition of Psilocybe cubensis mushrooms in the diet, occurring about 100,000 BCE. According to this “Stoned Ape” theory, access to and ingestion of psilocybin-containing mushrooms advantaged humans’ omnivorous hunter-gatherer ancestors, and was the “evolutionary catalyst” from which language, projective imagination, art, religion, philosophy, science, and human culture sprang. [84] In modern times, psilocybin-containing mushrooms came to the attention of many in the West when American banker and amateur ethnomycologist R. Gordon Wasson and his physician wife Valentina P. Wasson studied the ritual use of psychoactive mushrooms by the indigenous population in the Mazatec village Huautla de Jiménez, Mexico. In 1957, after their visit, Wasson described the visions that he experienced during these rituals in “Seeking the Magic Mushroom,” an article published in the popular American weekly Life magazine. [85] Samples of the psychoactive Psilocybe mexicana mushrooms they ingested were later sent to Albert Hofmann, a chemist at the Swiss pharmaceutical company Sandoz. Hofmann, who had in 1938 synthesized lysergic acid diethylamide (LSD), led a research group that isolated the psychoactive compounds and identified them as psilocybin and psilocin. Hofmann was aided in the discovery process by his willingness to ingest mushroom extracts to help verify the presence of active compounds. For his research, Hoffman and Sandoz were ultimately granted U.S. Pat. Nos.3,183,172 and 3,192,111 in 1957 and 1958, for methods of extracting psilocybin and psilocin from fungal material, and for using them as tranquilizers. D. Clinical Research with Psilocybin [86] In the early 1960s, psilocybin became available to academics for research. At Harvard University, Timothy Leary and his associates Ralph Metzner and Richard Alpert (later Ram Dass) performed research using synthesized psilocybin obtained from Hofmann’s Sandoz lab. Some studies, such as the Concord Prison Experiment, suggested promising results using psilocybin in clinical psychiatry. However, due to concerns about the increase in unauthorized use of psychedelic drugs by the general public, psilocybin became federally illegal in 1970, added to Schedule I of the Controlled Substances Act (CSA)—the most restrictive class of drugs, for substances that “have no currently accepted medical use,” “a lack of accepted safety for use under medical supervision, and a high potential for abuse.” These restrictions on its use made funding for academic research on psilocybin difficult to obtain, and scientists working with it terminated their research or became increasingly marginalized. [87] After several decades in the shadows, psychedelic drugs and their effects have again become the subject of scientific study, starting in the 1990s and increasing to the present. The return of interest in psychedelics, and the advances in neuropharmacology, neuropsychology, and brain imaging techniques that have opened new research paths, have refocused attention on psilocybin, as part of what has been called “the psychedelic renaissance” ( see generally , Carhart-Harris and Goodwin, 2017, and references cited therein). [88] At the time of this filing, well over a dozen research studies with psilocybin have been completed, and dozens more are currently in progress or recruiting. Such studies have examined or are examining the effects of psilocybin in the treatment of obsessive-compulsive disorder (OCD), major depressive disorder (MDD), depression and anxiety in life-threatening cancer, alcohol and nicotine dependence, and other diseases and mental health conditions. E. Drawbacks of Current Research with Psilocybin [89] Although many clinical studies have completed or are underway, they generally investigate psilocybin at “psychedelic” dosages, meaning subjects have significant mind-altering or psychedelic effects. Indeed, such psychedelic effects are often not only intended but also considered to be an integral part of the treatment, and an inseparable aspect of therapeutic efficacy. This is typically understood to mean that psilocybin must be taken in an appropriate psychotherapeutic setting. Further, because a psychedelic experience can last 6-8 hours, psychedelic-assisted therapy requires significant time from trained personnel. [90] At these higher dosages, besides significant psychedelic effects, potential adverse effects also increase in risk or severity. For example, psilocybin not only activates the 5-HT 2A receptors thought to be responsible for its beneficial therapeutic effects, but also activates 5-HT 2B receptors. Serotonergic agonists at 5-HT 2B receptors have been shown (especially at higher dosages, or with chronic longer-term use) to have harmful cardiotoxic effects, causing the formation of “valvular strands,” which can lead to Valvular Heart Disease (VHD) in a proportion of cases. Indeed, the 5-HT 2B receptor agonist fenfluramine was withdrawn from the market in the 1990s because of the incidence of VHD in patients (Hutcheson et al., 2011). In embodiments herein, the compositions and methods of the invention will be no worse than, at least as good as, and preferably better than, current treatments by having reduced cardiovascular risk or reduced cardiac adverse effects, by having reduced effects including long-term effects at 5-HT 2B receptors. [91] Clinical studies of psilocybin also generally investigate it in isolated form, as a purified fungal extract or a purified compound made by chemical synthesis. It is known, however, that Psilocybe and other psilocybin-containing mushrooms also contain dozens of related compounds and natural analogs that may have therapeutic benefits, or may modulate the therapeutic benefits of psilocybin. For example, aeruginascin, a structural analog of psilocybin (having an additional methyl group on the ethanolamine moiety) is believed to temper the negative effects of psilocybin, and lead to euphoric experiences (Sherwood et al., 2020). In embodiments, by formulating mushroom compositions comprising psilocybin together with additional “minor tryptamines,” an “entourage effect” is provided, including in some embodiments an “enhanced entourage effect.” In embodiments, the compositions and methods of the invention will be no worse than, at least as good as, and preferably better than, current treatments by having increased synergistic and entourage effects. F. Cannabis and Mental Health Conditions [92] Cannabis is a genus of flowering plant in the family Cannabaceae that is commonly recognized as containing the three species C. sativa , C. indica , and C. ruderalis . Cannabis is believed to be one of the first plants to be cultivated, and has a long history of human use for medicinal purposes. Indeed, in what is considered to be the first pharmacopoeia entry, in 2737 BC, the Chinese Emperor Shen-Nung described cannabis as useful to treat over 100 ailments including malaria, dysentery, constipation, gout, and rheumatic pain. Use of Cannabis for medicinal purposes continued with the Egyptians, Greeks, and Romans. [93] Beginning in the 19 th century, however, Cannabis use became increasingly vilified, culminating in its illegalization and criminalization in most countries. Although extensive use continued, including for medical purposes (occasionally gaining wide notice, for instance during the AIDS crisis), legalization attempts throughout the end of the 20 th century repeatedly stalled. To this day, Cannabis remains illegal in most countries, and is illegal at the federal level in the United States, on Schedule I of the CSA, and determined to have “no currently accepted medical use.” Hence, although a majority of U.S. states have now legalized Cannabis for medical and/or recreational (“adult use”) purposes, research into the therapeutic benefits of Cannabis (particularly as randomized controlled trials [RCTs], the gold standard for clinical research) remains underfunded and arduous to pursue. [94] In addition, the primary U.S. federal agency that funds Cannabis research is the National Institute on Drug Abuse (NIDA), resulting in research with a focus on dependence and adverse effects, rather than on potential therapies. Besides driving research into the negative consequences of Cannabis use, framing it as a dependency issue has also affected the way correlations between use and mental health conditions are understood. Logically, the cross-sectional relationship between Cannabis use and mental health conditions can be viewed in two ways: That is to say, is Cannabis use prevalent among those with mental disorders? Or are mental health conditions prevalent among Cannabis users? While the scientific literature is clear that use is highly correlated with major depressive, psychotic, anxiety, post-traumatic stress, bipolar, and other disorders, the bias toward viewing Cannabis use through the lens of dependency, addiction, and illegality has led many to conclude that its use causes or contributes to these disorders, rather than is a means to treat them. [95] Anecdotally, Cannabis users commonly report reliance on Cannabis to treat symptoms of mental health conditions. However, there is limited RCT data demonstrating the efficacy of such use, and data that exist are often equivocal, contradictory, or confounded by associations with other chronic conditions or substance abuse disorders. For example, substantial numbers of individuals suffering from PTSD report using Cannabis to cope with their symptoms, including anxiety, stress, and insomnia. A retrospective study of medical Cannabis users suggests that self-reported PTSD symptoms are lower when smoking Cannabis (Greer et al., 2014). Other studies have questioned these findings. In a longitudinal study of 2,276 veterans, Cannabis use was associated with greater PTSD symptom severity, increased violent behavior, and more alcohol and drug use (Wilkinson et al., 2014). The study concluded that Cannabis may actually worsen symptoms or nullify the benefits of specialized intensive treatment, and that cessation or prevention of use may be a goal of PTSD treatment. [96] Similarly, while Cannabis users broadly report that Cannabis helps with anxiety, a review of scientific literature reveals a relationship that is uncertain and complex. When analyzed in more detail, some compounds in Cannabis (e.g., THC) are shown to be potentially anxiogenic (i.e., exacerbating symptoms), while others (e.g., CBD) have shown anxiolytic properties (i.e., reducing symptoms). Complicating things further, while most medical research has focused on synthetic versions of single cannabinoids (and in particular THC and CBD), whole-plant Cannabis and full- or broad-spectrum extracts and preparations have hundreds of constituents that may work together in synergistic combinations, making the contribution of any individual compound(s) challenging if not impossible to tease out. [97] Indeed, other studies on anxiety using Cannabis have suggested that feelings of stress versus relaxation are impacted by the specific balance of cannabinoids (e.g., THC:CBD). Therefore, ingesting a formulation with differing amounts of particular compounds (e.g., an extract containing 20:1 THC:CBD versus one with 1:20 THC:CBD), may be expected to have clinically meaningful differential effects. However, studies into the effects of different cannabinoids at different proportions have not been performed in any systematic way. Evidence also indicates that patient responses are dose dependent, and can have biphasic (or even multiphasic) effect profiles, a result of the pharmacodynamic characteristics of different cannabinoid compounds. For instance, THC is shown to mitigate stress at relatively lower doses (7.5 mg), but exacerbate stress at relatively higher doses (12.5 mg). In addition, dose dependent effects tend to decrease with frequency of use, with infrequent Cannabis users less tolerant of larger doses of THC. Because of such effects, methods of administration that allow a person to choose and closely titrate the dosage (e.g., with accurately measured edibles or oral preparations, or by vaping low-THC plant material) reduce the risk of over-ingestion. However, while knowing total cannabinoid content per dose allows patients and consumers to only take the amount intended, the ultimate therapeutic effects of a formulation will still depend on the overall combination of chemical constituents, and their proportions. [98] In sum, the relationship between Cannabis and mental health conditions is highly nuanced and multifaceted. It is clear that many factors, such as the method, frequency, and duration of administration, the dosage and potency of individual constituents, and the specific combinations and proportions of those constituents together, all play a role in the ultimate therapeutic effect. Novel compositions that use different Cannabis constituents (or are based on such constituents, as derivatives, analogs, prodrugs, or synthetic versions thereof), and that are able to account for each of these factors, may improve treatment of mental disorders. [99] The bioactive compounds most readily associated with Cannabis are cannabinoids, generally held to be responsible for the primary mental and physical effects of Cannabis when consumed. Another class of compounds is terpenes, which are responsible for the unique fragrance of different Cannabis strains, and also cause numerous mental and physical effects. Last, the class of compounds known as flavonoids provide color and aroma as well as mental and physical effects. The interplay of different effects provided by the particular mix of cannabinoids, terpenes, and flavonoids in Cannabis flower (or in an extract or formulation prepared from or based on Cannabis ) will be shown to provide novel therapeutic treatments. G. Cannabinoids [100] Cannabinoids are a diverse class of small molecules that are grouped together because of their ability to act on cannabinoid receptors found in the brain and throughout the central and peripheral nervous systems of humans and other mammals. [101] There are two major types of cannabinoid receptors, known as cannabinoid receptor 1 (CB 1 ) and cannabinoid receptor 2 (CB 2 ). CB 1 receptors are primarily found in the central nervous system (i.e., the brain and spinal cord), as well as in the lungs, liver and kidneys. In fact, CB 1 receptors are the most abundant G protein-coupled receptor in the brain, and are widely expressed on almost all neuronal types in the brain, including gabaminergic, glutamatergic, serotonergic, noradrenergic, and dopaminergic terminals. CB 1 receptor-mediated signaling plays a critical role in neural circuitry that mediates mood, motivation, and emotional behaviors. CB 2 receptors are primarily found in the immune system and hematopoietic stem and progenitor cells, and may be found in neurons. [102] Cannabinoids produced endogenously in humans and other mammals may be termed “endocannabinoids.” These endocannabinoids are a class of lipid mediators, including amides, esters, and ethers of long-chain polyunsaturated fatty acids. Endocannabinoids are abundantly present in the cerebral cortex, basal ganglia, and limbic structures and exert their effects mainly through CB receptors. The first endocannabinoid was isolated in 1992, and was characterized to be N-arachidonoyl ethanolamide (AEA). AEA was subsequently named anandamide, from the Indian Sanskrit word “ananda,” meaning inner bliss. A second endocannabinoid, 2-arachidonoylglycerol (2-AG), was identified in 1994.2-AG is present at higher levels in mammalian CNS than AEA.2-AG and AEA act as full and partial agonists at the CB 1 receptor, respectively. Other endocannabinoids include N-arachidonoylglycine (NAGly), N-arachidonoyldopamine (NADA), 2-arachidonoylglyceryl ether (2-AGE), and 9-octadecenoamide (oleamide). Besides CB receptors, endocannabinoids may produce effects upon a variety of other targets; for example, oleamide has shown activity at CB 1 receptors as well as 5-HT 1A , 5-HT 2A/2C , 5-HT 7 , and GABA A receptors (Fowler, 2004). [103] Taken together, endocannabinoids and the endogenous cannabinoid receptors they act on (along with proteins involved in their metabolism, regulation, and degradation) form the “endocannabinoid system” (ECS). The ECS is shown or suggested to be involved in a variety of physiological processes including appetite, digestion, pain sensation, mood, memory, reproduction, stress response, immune function, thermoregulation, energy balance, and sleep. [104] Cannabinoids also can be isolated from plants, including at least 100 from the cannabis plant, among other plants including echinacea, kava, tea, and flax. Cannabinoids from plants may be termed “phytocannabinoids.” These non-endogenous cannabinoids also act on cannabinoid receptors in the body, and have many structural and functional similarities with endocannabinoids. Among the naturally-occurring phytocannabinoids from Cannabis , tetrahydrocannabinol, cannabidiol, and cannabichromene are three major constituents. [105] Tetrahydrocannabinol (THC) is a widely-known cannabinoid derived from Cannabis , particularly because of its intoxicating effects, resulting in the “high” associated with whole-plant Cannabis use. The psychoactive effects of THC are thought primarily due to interaction with CB 1 receptors. Besides the principle THC isomer (−)-trans-Δ 9 - tetrahydrocannabinol (Δ9-THC ), numerous other double bond- and stereo-isomers exist. [106] One such isomer is Δ8-THC , an analog with anxiolytic, antiemetic, analgesic, appetite-stimulating, and neuroprotective qualities. Δ8-THC is shown to be twice as effective an anti-emetic than Δ9-THC , but have up to 10 times less psychoactivity. Δ8-THC binds to CB 1 receptors located in the CNS, with potential affinity for CB 2 receptors, but due to its altered molecular structure, has a different affinity for those receptors than Δ9-THC . [107] Trace amounts of other THC isomers can be isolated from Cannabis . For example, the seven-carbon THC homologue Δ9 -trans-tetrahydrocannabiphorol (THCP) has been reported to be much more potent than five-carbon Δ9-THC , but provide similar pharmacological actions. The butyl and heptyl homologues of THC (Δ 9 -THCB and Δ 9 -THCP, respectively) have also been shown to have high CB 1 affinity and overall high cannabimimetic activity. [108] Cannabidiol (CBD) is another widely-known cannabinoid derived from Cannabis . Unlike THC, it does not produce intoxicating effects in humans, potentially antagonizing them instead. CBD is reported to demonstrate numerous pharmacological properties, including analgesic, antioxidant, anti-inflammatory, antiemetic, anticonvulsant, antipsychotic, anxiolytic, antidepressant, anticompulsive, antitumoral, neuroprotective, and immunomodulatory effects. CBD is an inverse agonist of CB 2 receptors, and an indirect antagonist of CB 1 and CB 2 cannabinoid receptor agonists (i.e., an antagonist to other ligands at CB 1 and CB 2 receptors). Interaction with CB 2 receptors appears to be primarily responsible for the anti-inflammatory and other therapeutic effects of Cannabis seen in animal models. [109] While THC directly activates CB 1 and CB 2 receptors, CBD does not bind to either, but instead impacts them indirectly. The pharmacology of CBD is enigmatic, involving interactions with multiple neurochemical systems, including serotonergic and adenosinergic systems. Suggested mechanisms of action supporting the pharmacological effects of CBD include activation of TRPV1 channels, inhibition of uptake and metabolism of the endocannabinoid anandamide (in part through FAAH inhibition), inhibition of adenosine uptake, antagonism of GPR55 and agonism of PPARγ receptors, and increase of intracellular calcium. CBD is also shown to act as an agonist of serotonergic 5-HT 1A receptors, possibly contributing to the ability of CBD to improve mood and reduce anxiety and depression. [110] Cannabichromene (CBC) is a third major naturally-occurring phytocannabinoid, which potentiates THC effects in vivo and has anti-nociceptive and anti-inflammatory effects. CBC has been shown to have potential to stimulate the growth of brain cells and the ability to normalize gastrointestinal hypermotility, and is viewed as a potential anti-cancer agent. [111] Importantly, in fresh Cannabis plant material cannabinoids are almost exclusively found in their “acidic” form (e.g., THCA, CBDA, CBCA), rather than their “neutral” form (e.g., THC, CBD, CBC) (and further, may have isomers, e.g., THCA-A and THCA-B). Cannabinoids in acidic form have an extra carboxyl (–COOH) group attached, which acts as an acid by donating its proton (H + ) and becoming ionized (to –COO - ). Then, in a process called decarboxylation, the deprotonated carboxyl group is lost as carbon dioxide (CO2), leaving the neutral form. The acidic cannabinoid forms have pharmacological properties, but generally must be converted into their neutral forms through decarboxylation before being active and bioavailable in humans (for instance, it is THC, and not THCA, that is responsible for the intoxicating effects of Cannabis ). How quickly decarboxylation occurs is a function of pressure, heat, and time. At lower temperatures, such as when a plant is dried and cured after harvesting, decarboxylation occurs slowly and only partially. At higher temperatures, such as when cannabis is combusted by smoking, the cannabinoids are almost instantaneously decarboxylated, and are thus immediately available for absorption through inhalation. [112] Cannabigerolic acid (CBGA), the original biogenetic precursor phytocannabinoid, metabolically yields THCA, CBDA, and CBCA via their respective synthases, by enzymatic processes in the plant. The neutral form of CBGA, cannabigerol (CBG), is shown to have anti-inflammatory, antibiotic and antifungal properties, and to inhibit cancer cell growth. [113] Cannabinol (CBN), although not directly synthesized in Cannabis , is found in trace amounts and is weakly psychoactive. CBN is a breakdown product of THC, when THC is oxidized to CBN via prolonged exposure to heat, oxygen and light. Thus, Cannabis specimens or flower products tend to have increasing levels of CBN as they age. CBN is observed to produce greater sedation when combined with THC, and may have anticonvulsant, anti-inflammatory, antibiotic, and analgesic properties. CBN has affinity to CB 2 receptors, and acts as a partial agonist of CB 1 receptors (with lower affinity than THC). [114] Generally, phytocannabinoids from Cannabis have a pentyl side-chain, but traces of methyl and ethyl and minor amounts of propyl homologues also occur. The propyl homologues of CBD and THC, named cannabidivarin (CBDV) and trans-tetrahydrocannabivarin (THCV) respectively, are present in cannabis plants at various ratios, depending on the specific strain. Research shows that THCV and CBDV may have therapeutic potential for curbing anxiety, reducing nausea, treating neuronal hyperexcitability, stimulating bone growth, and improving insulin sensitivity. THCV also influences the effects of THC. At lower doses, THCV appears to diminish the ability of THC to activate CB 1 receptors, similar to CBD (i.e., counteracting the effects of THC). However, at higher doses, THCV also activates CB 1 receptors, like THC (i.e., reinforcing effects of THC). [115] In embodiments of the invention, cannabinoids include those as set forth and described by Radwan et al. in Cannabinoids, Phenolics, Terpenes and Alkaloids of Cannabis , Molecules, 26(9), 2774 (2021), which is incorporated by reference as if fully set forth herein. In general, but without being bound by theory, cannabinoids according to such categorization include compounds with a characteristic C21 terpenophenolic backbone that are part of one of 11 cannabinoid sub-classes, namely: cannabichromene (CBC)-type, cannabidiol (CBD) type, cannabielsoin (CBE) type, cannabigerol (CBG) type, cannabicyclol (CBL) type, cannabinol (CBN) type, cannabinodiol (CBND) type, cannabitriol (CBT) type, (−)- Δ8 - trans -tetrahydrocannabinol (Δ8 -THC) type, (−)-Δ9 -trans-tetrahydrocannabinol (Δ9-THC ) type, and miscellaneous-type cannabinoids. Non-limiting examples of such cannabinoids, all of which will be understood to be useful in the practice of the invention, are known by reference to the disclosure of Radwan 2021 and the below. a. ∆ 9 -THC-type Cannabinoids [116] In embodiments, a cannabis composition of the invention comprises one or moreΔ9-THC -type cannabinoids which include Δ9-THC -C 5 , Δ9-THC AA-C 5 , Δ9-THC AB-C 5 , Δ9-THC -C 4 , Δ9-THC AA-C 4 , Δ9-THC V, Δ9-THC VAA, Δ9-THC O, Δ9-THC OAA, Δ9-THC –aldehyde, β-fenchyl (−)-Δ9-trans-tetrahydrocannabinolate, α-fenchyl (−)-Δ9-trans-tetrahydrocannabinolate, epi-bornyl (−)-Δ9-trans-tetrahydrocannabinolate, bornyl (−)-Δ9-trans-tetrahydrocannabinolate, α-terpenyl (−)-Δ9-trans- tetrahydrocannabinolate, 4-terpenyl (−)-Δ9-trans-tetrahydrocannabinolate, α-cadinyl (−)-Δ9-trans-tetrahydrocannabinolate, γ-eudesmyl (−)-Δ9-trans- tetrahydrocannabinolate, 8α-hydroxy-(−)-Δ9-trans-tetrahydrocannabinol, 8β-hydroxy-(−)-Δ9- trans-tetrahydro cannabinol, 11-acetoxy-(−)-Δ9-trans-tetrahydrocannabinolic acid A, 8-oxo-(−)-Δ9-trans-tetrahydrocannabinol, cannabisol, (−)-Δ9-trans-tetrahydro-cannabi- phorol, and (−)-Δ9-trans-tetrahydrocannabihexol. b. Δ8 -THC-type Cannabinoids [117] In embodiments, a cannabis composition of the invention comprises one or more Δ9 -THC-type cannabinoids which include Δ8-THC , Δ8-THC A, 10α-OH-Δ8-THC , 10β-OH- Δ8-THC , and 10a-α-hydroxy-10-oxo-Δ8-THC. c. CBG-type Cannabinoids [118] In embodiments, a cannabis composition of the invention comprises one or more CBG-type cannabinoids which include ( E )CBG, ( E )CBGA, ( E )CBGG, ( E )CBGAM, ( E )CBGV, ( E )CBGVA, (Z)CBGA, 5-acetyl-4-hydroxy-cannabigerol, (±)-6,7- trans - epoxycannabigerolic acid, (±)-6,7- cis -epoxycannabigerolic acid, (±)-6,7- cis - epoxycannabigerol, (±)-6,7- trans -epxoycannabigerol, camagerol, and sesquicannabigerol. d. CBD, CBND, CBE, and CBL-type Cannabinoids [119] In embodiments, a cannabis composition of the invention comprises one or more CBD, CBND, CBE, and CBL-type cannabinoids which include CBD-C 5 , CBDA-C 5 , CBDM–C 5 , CBD-C 4 , CBDV, CBDVA, CBD-C 1 , CBDH, CBDP, CBDD, CBND-C 3 , CBND-C 5 , CBE-C 5 , CBEAA-C 5 , CBEAB-C 5 , CBE-C 3 , CBEAB-C 3 , CBL, CBLA, and CBLV. e. CBC and CBN-type Cannabinoids [120] In embodiments, a cannabis composition of the invention comprises one or more CBC and CBN-type cannabinoids which include CBC, CBCA, ±CBCV, +CBCV, CBCVA, 4-acetoxy-CBC, (±)-3”-hydroxy-Δ4”-cannabichromene, (–)-7-hydroxy- cannabichromane, CBC-C 3 , CBN-C 5 , CBNA-C 5 , CBN-C 4 , CBN-C 3 , CBN-C 2 , CBN-C 1 , CBNM–C 5 , 8-OH-CBN, 8-OH-CBNA, 1’ S -OH-CBN, and 4-terpenyl-cannabinolate. f. CBT-type Cannabinoids [121] In embodiments, a cannabis composition of the invention comprises one or more CBT-type cannabinoids which include (−)- trans -CBT-C 5 , (+)- trans -CBT-C 5 , (±)- cis -CBT-C 5 , (±)- trans -CBT-C 3 , CBT-C 3 -homologue, (−)- trans -CBT-OEt-C 5 , (–)- trans -CBT-OEt-C 3 , 8,9-Di-OH-CBT-C 5 , and CBDA-C 5 , and 9-OH-CBT-C 5 ester. g. Miscellaneous-type Cannabinoids [122] In embodiments, a cannabis composition of the invention comprises one or more miscellaneous-type cannabinoids which include DCBF-C 5 , CBF-C 5 , OH-iso-HHCV-C 3 OTHC, cannabicitran, cis -Δ 9 -THC, CBCON-C 5 , CBR, CBTT, CBCN-C 5 , CBCN-C 3 , cis -iso-Δ 7 -THCV, trans -iso-Δ 7 -THCV, trans -iso-Δ 7 -THC, CBCNB, CBCNC, CBCND, (–)-(7 R )-cannabicoumarononic acid, 4-acetoxy-2-geranyl-5-hydroxy-3- n - pentylphenol, 2-geranyl-5-hydroxy-3- n -pentyl-1,4-benzoquinone, 5-acetoxy-6-geranyl-3-n- pentyl-1,4-benzoquinone, CBM, CBX, 10α-hydroxy-Δ 9,11 -hexahydrocannabinol, 9β,10β- epoxyhexahydrocannabinol, 9α-hydroxyhexahydrocannabinol, 7-oxo-9α-hydroxyhexa- hydrocannabinol, 10α-hydroxyhexahydrocannabinol, 10aR-hydroxyhexahydrocannabinol, and 9α-hydroxy-10-oxo-Δ 6a,10a -THC. [123] Each cannabinoid also will be understood to include its isomers, such as structural isomers and stereoisomers (including enantiomers), the -A and -B isomers for each cannabinoid, double bond isomers, and other such isomers known to those of skill. Thus, “THC” will, in embodiments, be understood to include THC–A and THC–B. In embodiments, included among cannabinoids are the cannabinoid carboxylic acids and their carboxylate salts (see, e.g., U.S. Pat. No.9,376,367). Additionally, reference to a cannabinoid includes the various alkyl chain lengths associated therewith, as illustrated by C n , wherein “ n ” refers to the number of carbon atoms in each alkyl chain. Thus, “THC” also includes THC-C1, THC-C2, THC-C3, THC-C4, THC-C5, THC-C6, and THC-C7. Further, reference to a given cannabinoid will include all possible isomers, such as but not limited to its -A and -B isomers, and all possible combinations of alkyl chain lengths, including chains comprised of 1, 2, 3, 4, 5, 6, or 7 carbon atoms. So, herein, reference to “THC” will include THC-C1 A, THC-C1 B, THC-C2 A, THC-C2 B, THC-C3 A, THC-C3 B, THC-C4 A, THC-C4 B, THC-C5 A, THC-C5 B, THC-C6 A, THC-C6 B, and THC-C7 A, and THC-C7 B. As would be apparent to one of skill, such logic applies to all such cannabinoids to which the invention applies; THC is merely used to illustrate such logic, and should not be construed as limiting. [124] In embodiments, cannabinoids may further comprise one or more chemical moieties including, but not limited to, methyl, alkyl, alkenyl, methoxy, alkoxy, acetyl, carboxyl, carbonyl, oxo, ester, hydroxyl, aryl, heteroaryl, cycloalkyl, cycloalkenyl, cycloalkylalkenyl, cycloalkenylalkyl, cycloalkenylalkenyl, heterocyclylalkenyl, heteroarylalkenyl, arylalkenyl, heterocyclyl, aralkyl, cycloalkylalkyl, heterocyclylalkyl, heteroarylalkyl, and the like, and still be within the spirit of the invention as disclosed herein. H. Non-Cannabinoid Compounds [125] In addition to cannabinoids, many hundreds of non-cannabinoid compounds are found in Cannabis , including non-cannabinoid phenols (including spiro-indans, dihydrostilbenes, dihydrophenathrenes, and simple phenols, according for purposes of organization and explication only, and using the classification system discussed above, but not to be limited thereby), as well as flavonoids and terpenes (as discussed in separate sections below), and alkaloids, such as the spermidine alkaloids cannabisativine and anhydrocannabisativine. a. Cannabis Spiryo-Indans [126] In embodiments, a cannabis composition of the invention comprises one or more Spiro-Indans which include cannabispiran, cannabispirenone, cannabispirenone isomer, cannabispiradienone, β-cannabispirol, β-cannabispirol acetate, 7-methoxyindan-1-spiro- cyclohexane, 7-hydroxy-5-methoxyindan-1-spiro-cyclohexane, 5,7-dihydroxyindan-1-spiro- cyclohexane, isocannabispiran, 7-O-methyl-cannabispirone, isocannabispiradienone, α-cannabispiranol, cannabispirketal, α-cannabispiranol-4′- O-β-glucopyranose, and prenylspirodienone. b. Cannabis Dihydrostilbenes [127] In embodiments, a cannabis composition of the invention comprises one or more dihydrostilbenes which include 3-[2-(4-hydroxyphenyl)-ethyl]-5- methoxyphenol, 3-[2-(3-hydroxy-4-methoxyphenyl)-ethyl]-5-methoxyphenol, 3-[2-(3- isoprenyl-4-hydroxy-5- methoxy-phenyl)-ethyl]-5-methoxyphenol, canniprene, cannabistilbene I, cannabistilbene II, 3,4′,5-trihydroxy-dihydrostilbene, α,α′-dihydro- 3′,4,5′-trihydroxy-4′-methoxy- 3-isopentenylstilbene, α,α′-dihydro-3,4′,5-trihydroxy-4-methoxy-2,6-diisopentenylstilbene, α,α′-dihydro- 3′,4,5′-trihydroxy- 4′-methoxy-2′,3-diisopentenylstilbene, α,α′-dihydro-3,4′,5- trihydroxy-4,5′-diisopentenylstilbene, and combretastatin B-2. c. Cannabis Dihydrophenanthrenes [128] In embodiments, a cannabis composition of the invention comprises one or more dihydrophenanthrenes which include cannabidihydrophenanthrene (cannithrene 1), cannithrene 2, 4,5-dihydroxy-2,3,7-trimethoxy-9,10-dihydrophenanthrene, 4-hydroxy- 2,3,6,7-tetramethoxy-9,10-dihydrophenanthrene, 4,7-dimethoxy-1,2,5- trihydroxy- phenanthrene, denbinobin, and dihydrophenanthrenedione. d. Cannabis Simple Phenols [129] In embodiments, a cannabis composition of the invention comprises one or more cannabis simple phenols which include eugenol, methyleugenol, iso-eugenol, trans-anethol, cis-anethol, vanillin, and phloroglucinol β-D-glucoside. I. Terpenes [130] Terpenes are a large and diverse class of organic hydrocarbon compounds, produced by a broad variety of plants, including Cannabis . Terpenes are ubiquitous throughout nature, with some estimates concluding that as many as 60 percent of all naturally-produced chemical compounds are members of the class (totaling over 20,000 unique compounds). “Terpene” is derived from “turpentine,” the pungent resin extracted from the terebinth tree Pistacia terebinthus . Terpenes may be referred to as “terpenoids” when they have experienced oxidation and have an additional oxygen-containing functional group (for instance, after cannabis is cured and dried), or when they are otherwise modified by addition or removal of a functional group (e.g., a methyl group (–CH 3 )). The presence and particular combination of terpenes gives different plants (and different cannabis strains) their distinctive smells and tastes. In Cannabis , terpenes are the largest group of phytochemicals, with at least 120 identified molecules. Terpenes generally make up between 10-20 percent of the total oil content produced by Cannabis resin glands. Terpenes also constitute the majority of chemicals in the heated or vaporized smoke of Cannabis flowers, often consisting of greater than 50%, with cannabinoids normally accounting for 10-20%. [131] Chemically, terpenes are constituted by one or more repeating units of a five-carbon building block known as an isoprene unit (i.e., 2-methyl-1,3-butadiene , having the molecular formula C 5 H 8 ). Being isoprene polymers, terpenes as a group are also often referred to as “isoprenoids.” Isoprene units may be linked together end-to-end to form linear chains, or may be arranged so as to form rings (thus having the molecular formula (C 5 H 8 ) n , where n is the number of linked isoprene units). Terpenes are classified in families according to the number of isoprene units from which they are constituted: as hemiterpenes (one unit), monoterpenes (two), sesquiterpenes (three), diterpenes (four), sesterterpenes (five), triterpenes (six), sesquarterpenes (seven), tetraterpenes (eight), and polyterpenes (nine or more). [132] Monoterpenes generally dominate the terpene profile of Cannabis . Monoterpenes include myrcene, d-limonene, α- and β-pinene, terpinolene and linalool. Sesquiterpenes (and β-caryophyllene and α-humulene in particular) also occur to a large extent in cannabis, as do triterpenes (as β-amyrin, friedelin and epifriedelanol, cycloartenol, and dammaradienol). [133] Although cannabinoids are more popularly understood to be responsible for the mental and physical effects of Cannabis , terpenes have demonstrated a wide array of such effects as well ( see generally, e.g ., Russo, 2011). For example, the terpene β-myrcene has been shown to have a sedative effect, and is believed to be responsible for a heavy “body high.” Indeed, preparations of β-myrcene from hops have been employed as a sleep aid. Furthermore, the effects of β-myrcene have been demonstrated to be blocked by the drug naloxone, which is also used to block the effects of opioids, especially in overdose. β-myrcene has also demonstrated the ability to reduce inflammation and block hepatic carcinogenesis, and act as an analgesic and muscle relaxant. Beyond β-myrcene, the terpenes linalool, nerolidol, and pulegone have also shown sedative effects. Others, such as limonene and terpinolene, have by contrast shown stimulating effects. And others show yet different effects. For example α-humulene acts as an appetite suppressant. Limonene exhibits anti-cancer, anxiolytic, and immunostimulating properties, and nerolidol also has anti-cancer properties. β-caryophyllene possesses anti-inflammatory and gastric cytoprotector effects. And pentacyclic triterpenes, such as β-amyrin and cycloartenol, show anti-bacterial, anti-fungal, anti-inflammatory, and anti-cancer properties. As research progresses, many other terpene properties and potential uses continue to be discovered. [134] Terpenes may cause effects when consumed because of the modulation of neurotransmitter systems in the brain, as terpenes easily cross the blood-brain barrier (BBB). Linalool, for example, is shown to modulate the glutamatergic and gabaminergic neurotransmitter systems, which may explain its analgesic, anti-anxiety, anti-inflammatory, and anticonvulsant properties. α-Pinene is an acetylcholinesteral inhibitor, and may thereby aid memory. And phytol, a diterpene, increases gabaminergic expression. Other terpenes have been shown to affect serotonergic and dopaminergic neurotransmitter systems. [135] Furthermore, some terpenes interact directly with the ECS. For example, β-caryophyllene selectively binds to CB 2 receptors as a functional CB 2 agonist, supporting β-caryophyllene as having an anxiolytic and antidepressant effect. Terpenes also have been shown to alter the permeability of cell membranes and thereby modulate the effects of THC and other cannabinoids. Because terpenes are lipophilic, they interact with lipid membranes, ion channels, a variety of different receptors (including both G protein-coupled odorant and neurotransmitter receptors), and enzymes. Through these and other mechanisms, the terpenes in cannabis do not only cause effects individually, and in combination with other terpenes, but may also modulate the effects of the different cannabinoids that are present. [136] In general, but without being bound by theory, terpenes according to the categorization of Radwan 2021 include monoterpenes (C 10 skeleton), sesquiterpenes (C 15 skeleton), diterpenes (C 20 skeleton), triterpenes (C 30 skeleton), and miscellaneous terpenes. Non-limiting examples of terpenes, all understood to be useful in the practice of the invention, are known by reference to the disclosure of Radwan 2021 and the below. a. Monoterpenes [137] In embodiments, a cannabis composition of the invention comprises one or more monoterpenes which include myrcene, cis-β-ocimene, trans-β-ocimene, p-cymene, α-terpinene, β-phellandrene, γ-terpinene, α-terpinolene, α-phellandrene, 3-phenyl-2-methyl-prop-1-ene, α-pinene, β-pinene, camphene, Δ 3 -carene, Δ 4 -carene, sabinene, α-thujene, linalool, citral B, nerol, geraniol, ipsienol, citronellol, 2-methyl-2-heptene-6-on, geranyl acetone, m-mentha-1,8-(9)-dien-5-ol, carvacrol, carvone, α-terpineol, terpinene-4-ol, pulegone, dihydrocarvone, β-terpineol, dihydrocarveyl acetate, p-cymene-8-ol, β-cyclocitral, safranal, cis-linalool oxide, perillene, sabinol, thujyl alcohol, linalool oxide, cis-carveol, cis-sabinene hydrate, sabinene hydrate, ,8-cineol, 1,4-cineol, piperitone oxide, piperitenone oxide, fenchyl alcohol, fenchone, borneol, bornyl acetate, camphor, camphene hydrate, α-pinene oxide, pinocarveol, and pinocarvone. b. Sesquiterpenes [138] In embodiments, a cannabis composition of the invention comprises one or more sesquiterpenes which include α-caryophyllene, β-caryophyllene, caryophyllene oxide, curcumene, α-trans-bergamotene, α-selinene, β-farnesene, longifolene, humulene epoxide I, humulene epoxide II, caryophyllene alcohol (caryophyllenol), β-bisabolene, allo-aromadendrene, calamenene, α-copaene, nerolidol, α-gurjunene, iso-caryophyllene, β-selinene, selina-3,7(11)-diene, selina-4(14),7(11)-diene, α-bisabolol, α-cedrene, α-cubebene, δ-cadinene, epi-β-santalene, farnesol, γ-cadinene, γ-elemene, γ-eudesmol, guaiol, ledol, trans-trans-α-farnesene, (Z)-β-farnesene, farnesyl acetone, α-cadinene, α-cis-bergamotene, α-eudesmol, α-guaiene, α-longipinene, α-ylangene, β-elemene, β-eudesmol, epi-α-bisabolol, γ-cis-bisabolene, γ-curcumene, γ-muurolene, γ-trans-bisabolene, viridiflorene, germacrene-B, and clovandiol. c. Diterpenes, Triterpenes, and Miscellaneous Terpenes [139] In embodiments, a cannabis composition of the invention comprises one or more diterpenes, triterpenes, and miscellaneous terpenes which include phytol and neophytadiene (diterpenes); friedeline and epifriedelanol (triterpenes); and vomifoliol, dihydrovomifoliol, β-ionone, and dihydroactinidiolide (miscellaneous terpenes). J. Flavonoids [140] A third class of phytochemicals produced by Cannabis are flavonoids, which account for roughly 10 percent of the bioactive compounds in the plant. Flavonoids are a broad class of thousands of different water-soluble polyphenolic molecules (i.e., consisting of a phenyl group (−C 6 H 5 ) bonded to a hydroxy group (−OH)), of which about 20 are widely found in cannabis. Flavonoids get their name from the Latin word “flavus” for the color yellow. In fact, a primary function of flavonoids is to provide color pigmentation to plants, particularly flowers. In Cannabis , for instance, deep purple strains owe their coloration to the flavonoids known as anthocyanins and anthoxanthins. Besides providing color, flavonoids also have been shown to provide health benefits through modulation of cell signaling pathways and through various anti-inflammatory, antioxidant, anti-fungal, anti-cancer, and other effects. For example, the Cannabis flavonoid apigenin has potent anti-anxiety, anti-inflammatory, and anti-cancer properties; butin has been shown to reduce oxidative stress-related cell dysfunction. Other bioactive flavonoids found in cannabis include cannaflavins, kaempferol, orientin, luteolin, quercetin, silymarin, and vitexin. [141] In general, but without being bound by theory, non-limiting examples of flavonoids, all of which will be understood to be useful in the practice of the invention, are known by reference to the disclosure of Radwan 2021, and include orientin, vitexin, isovitexin, apigenin, luteolin, kaempferol, and quercetin flavonoids that may be methylated, glycosylated, prenylated, or geranylated. In embodiments, flavonoids include the orientin, vitexin, isovitexin, apigenin, luteolin, kaempferol, and quercetin flavonoids that may be methylated, glycosylated, prenylated, or geranylated; disclosed by Radwan et al., 2021. [142] In embodiments, a cannabis composition of the invention comprises one or more flavonoids which include orientin, orientin-O-glucoside, orientin-7-O-glucoside, orientin-7-O-rhamnoglucoside, vitexin, vitexin-O-glucoside, vitexin-7-O-glucoside, vitexin-7-O-rhamnoglucoside, cytisoside, cytisoside-glucoside, isovitexin, isovitexin-O- glucoside, isovitexin-7-O-glucoarbinoside, isovitexin-7-O-rhamnoglucoside, apigenin-7-O- glucoside, apigenin-7-O-glucuronoid, apigenin-7-O'P-coumaroylglucoside, 6-prenylapigenin, apigenin-6,8-di-glucopyranoside, luteolin-C–glucuronide, luteolin-7-O-glucuronide, canniflavin A, canniflavin B, canniflavin C, chrysoeriol, kaempferol-3-O-diglucoside, quercetin-3-O-glucoside, quercetin-3-O-diglucoside, kaempferol-3-O-sophoroside, quercetin-3-O-sophoroside, rutin, quercetin, naringenin, and naringin. K. The “Entourage Effect” [143] As discussed above, Cannabis contains hundreds of different bioactive compounds. This polypharmacy of phytochemicals, together with their countless possible targets in the body, and combined with various endocannabinoids, neurotransmitters, and other molecules with which they interact, proves an endless potential for pharmacological permutations. [144] However, the dominant model for pharmaceutical development has long been to focus on single agent therapies. Indeed, the first Cannabis -derived drug to be FDA-approved (to treat AIDS-related loss of appetite, and chemotherapy-related nausea and vomiting) was dronabinol (Marinol ® )—pure, synthetic THC. When dronabinol became available in the mid-1980s, scientists and physicians thought it would have the same effect as the whole-plant Cannabis that was then widely-used illegally to treat the same symptoms. But it became clear that most patients preferred using the whole plant to dronabinol, and many discontinued dronabinol because of serious adverse events, including severe paranoia and acute psychosis. The question therefore was raised of what other compounds in cannabis are responsible for creating and modulating its therapeutic effects. [145] In 1998, a pair of pharmacologically inactive monoacylglycerols were found to modulate activity of the endocannabinoid 2-AG through inhibition of its metabolism. This observation led to the coining of the term “entourage effect” to describe how putatively “inactive” metabolites could markedly increase the activity of the primary endogenous cannabinoids (Ben-Shabat et al., 1998). In years since, “entourage effect” has been used more broadly to explain how whole-plant extracts and multiple-compound compositions are often more efficacious, or have different effects, than their isolated components. And in succeeding years, reports have started to accumulate that various cannabis phytochemicals working together can potentially achieve effects beyond the mere additive effects of compounds alone. [146] For instance, studies have confirmed the benefits of whole-plant and broad-spectrum formulations over pure THC preparations, and demonstrated the role of CBD in particular to mitigate the negative effects of THC, although other cannabinoids including CBC and Δ 8 -THC also appear to potentiate some THC effects. THCV also influences the effects of THC, although in a dose-dependent fashion, diminishing the ability of THC to activate CB 1 receptors at lower doses (i.e., counteracting the effects of THC), but activating CB 1 receptors itself at higher doses (i.e., reinforcing the effects of THC). The superiority of whole-plant extracts to pure THC in studies of human cancer cell lines have been attributed to the presence of trace amounts of CBG and THCA. In studies of analgesia, pure CBD produces a biphasic dose-response curve such that smaller doses reduce pain responses until a peak is reached, after which further dose increases are ineffective. However, the application of a broad-spectrum cannabis extract with equivalent CBD doses eliminates the biphasic response in favor of a linear dose-response curve such that an extract is analgesic at any dose with no observed ceiling effect. Studies also show that CBD-predominant whole-plant extracts perform better than pure CBD for treatment of patients with epilepsy, based on improvement in seizure frequency and reduction of adverse events. The synergistic contribution of other entourage compounds with anticonvulsant properties, such as THC, THCA, THCV, CBDV, linalool, and β-caryophyllene, are posited to potentially underlie the effect. [147] However, systematic research into different combinations of Cannabis compounds has been lacking, and much of the evidence purported to support the entourage effect is anecdotal, or based solely on in vitro or animal studies, or on comparisons between single synthetic cannabinoid molecules and whole-plant preparations (versus defined multi- cannabinoid, cannabinoid+terpenoid, or cannabinoid+terpenoid+flavonoid formulations). Because of these limitations, to this day scientists still question whether the contributions of different cannabis compounds impact the overall pharmacological effect of a cannabis formulation to any meaningful extent. As one recent review observed, “[c]ompelling clinical data is lacking to support the notion of an entourage effect as a reliable phenomenon that is predictive of beneficial outcomes” (Cogan, 2020). Cogan further explains “the polypharmacy of cannabis does not at present predict therapeutic outcomes [and] does not reliably yield superior effects from [whole-plant and full- and broad-spectrum] products relative to isolated compounds,” and in fact, “the polypharmacy of cannabis can in many instances be better framed as a detriment to its proposed therapeutic utility.” ( Id .) With psychedelics, research on entourage effects and multiple-compound compositions has lagged even further behind. L. Compounds and Compositions [148] In some aspects (“mushroom compositions”), the compositions of the invention comprise psilocybin and/or other tryptamines, and optionally L-theanine, niacin, and extracts from cordyceps and lion’s mane. In embodiments, these compositions further comprise valerian extract. In embodiments, these compositions further comprise vitamin B12. In embodiments, they further comprise a CB 1 receptor modulator, such as CBD or PEA. In yet other embodiments, they may further comprise one or more other active compounds. In embodiments, these are formulated with at least one carrier, excipient, or diluent. [149] In other aspects (“cannabis compositions”), the compositions of the invention comprise an entourage-enhancing cannabinoid fraction, which may include a cannabinoid such as cannabidiol (CBD) and in “C+” embodiments, a tetrahydrocannabinol (e.g., Δ9 -THC, ∆ 8 -THC, or another intoxicating THC compound); an entourage-enhancing isoprenoid fraction; and optionally, one or more flavonoids. In embodiments, these are formulated with at least one carrier, excipient, or diluent. [150] In certain aspects, the methods of the invention include “daily dosing regimens” comprising administration of the mushroom compositions and cannabis compositions. In embodiments, a daily dosing regimen comprises administering both of a mushroom composition and a cannabis composition in a defined daily dosing pattern according to the distinct methods and protocols that are taught herein and claimed. [151] In other aspects, the methods of the invention include “therapeutic regimes” comprising administration of mushroom compositions and cannabis compositions across two or more daily dosing regimens, wherein each such daily dosing regimen may be the same (as in a repeated daily dosing regimen) or different (as in a combination daily dosing regimen). [152] As used herein, unless otherwise defined or otherwise dictated by context, “nutraceutical” and “pharmaceutical” shall be used interchangeably when referring to the compositions of the invention, as in the “nutraceutical and/or pharmaceutical compositions of the invention,” and both shall refer to a composition that is contemplated or shown to possess therapeutic effects when administered for its intended purpose to a mammal, such as a human. Even where not so defined, the mushroom compositions and cannabis compositions of the invention shall be known to be both “pharmaceutical compositions” and “nutraceutical compositions.” “Compositions of the invention” will be used as shorthand, and include these meanings. Said otherwise, when referring simply to “mushroom compositions” or “cannabis compositions” (or the term “mushroom and cannabis compositions,” which will encompass both) it will be appreciated that the terms include such compositions as both “nutraceutical” and “pharmaceutical” compositions, even if not so stated specifically. It should be further understood that the compositions described herein shall be useful regardless of the regulatory regime under which they are ultimately sold (e.g., as prescription pharmaceutical drug products, non-prescription over-the-counter (OTC) drug products, or dietary supplements), and even if not sold under a specific regulatory regime at all (e.g., as natural products). [153] In embodiments, one or more supplements may be paired with, or incorporated into, the compositions of the invention. Broadly, supplements are known to those of skill as having a wide range of health effects, and may, in embodiments, provide additional synergistic or unexpected results when paired with certain other compounds in a composition. Some embodiments, for example, may include a standardized Ginkgo biloba extract, which has demonstrated in a study on the effects of three doses (120 mg, 240 mg, 360 mg), that were assessed on performance of two related serial subtraction tasks with different levels of mental effort, an improvement in cognitive function compared with placebo (Scott, 2019). In embodiments, “paired with” means used in a daily dosing kit as one of the “A” days. [154] In embodiments, the supplements paired with or incorporated into the compositions of the invention include, but are not limited to, calcium, echinacea, fish oil, ginseng, glucosamine, chondroitin sulfate, garlic, St. John’s wort, saw palmetto, ginkgo, green tea extract, fiber, selenium, chromium, creatine, iodine, iron, magnesium, potassium, Acacia rigidula, cesium chloride, folic acid, betaine anhydrous, choline bitartrate, agmatine sulfate, linoleic acid, coleus extract, caffeine, zinc, maca root, quercetin, dimethylethanolamine (DMAE or DMEA), acetyl-L-carnitine, epigallocatechin, epicatechin gallate, epicatechin, kaempferol, myricetin, guarana, and any other such supplements known to those of skill. [155] In embodiments, one or more vitamins may be paired with and/or incorporated into the compositions of the invention. Like supplements, vitamins offer a wide range of health benefits. In such embodiments, vitamins incorporated into, and/or paired with the compositions of the invention include, but are not limited to vitamin A (retinol, retinoic acid), vitamin B1 (thiamin), vitamin B2 (riboflavin ) , vitamin B3 (niacin), vitamin B6 (pyridoxine), vitamin B12 (cobalamin), vitamin C (ascorbic acid, L-ascorbic acid), vitamin D (calciferol), vitamin E (tocopherols and/or tocotrienols), and vitamin K (phytonadione or phylloquinone). [156] In embodiments, one or more nootropics may be paired with and/or incorporated into the compositions of the invention. “Nootropics,” sometimes called “smart drugs,” may refer to the class of OTC or prescription substances that are believed to boost brain performance, and sometimes also called “cognition enhancers” or “memory enhancers.” Prescription nootropics include, e.g., modafinil (Provigil), certain mixed amphetamine salts (Adderall, Mydayis), methylphenidate (Ritalin), and memantine (Axura). OTC nootropics include, e.g., caffeine, L-theanine, omega-3 fatty acids, racetams (e.g., piracetam, pramiracetam, phenylpiracetam, aniracetam), Ginkgo biloba , Panax ginseng , Rhodiola rosea (roseroot), and creatine. It will be readily appreciated that compounds hence may fall into more than one category herein (e.g., a “nootropic” may also be a supplement or amino acid) [157] Herein, “cordyceps” may refer to any species from the genus of ascomycete fungi known as Cordyceps, whether from the subgenera Cordyceps subgen. Cordyceps, or Cordyceps subgen. Cordylia, e.g., Cordyceps sinesis or Cordyceps militaris, especially those species relatively high in the compound cordycepin (3'-deoxyadenosine), which has demonstrated antidepressant effects in animal models of depression. A “compound from Cordyceps” will be understood to include, besides cordycepin, any one or more of cordycepic acid, N-acetylgalactosamine, adenosine, ergosterol, ergosteryl esters, bioxanthracenes, hypoxanthine, acid deoxyribonuclease, polysaccharide, exopolysaccharide, chitinase, macrolides, cicadapeptins, myriocin, superoxide dismutase, protease, naphthoquinone, cordy heptapeptide, dipicolinic acid, fibrinolytic enzyme, lectin, and cordymin (Tuli, 2014).
[158] In embodiments, the invention will include fungi from the genus Hericium. Species of Hericium are white and fleshy, and generally grown on dead or dying wood. The fruiting bodies of Hericium generally contain a mass of icicle-like spines suspended from a branched supporting framework or an unbranched cushion of tissue, giving the species its colloquial names including “Lion’s mane,” “monkey head mushroom,” “bearded tooth mushroom,” “satyr’s beard,” “bearded hedgehog mushroom,” “pom pom mushroom,” and/or “bearded tooth fungus” (Miller, 1933). As used herein, “Lion’s mane” may refer to Hericium erinaceus. Other Hericium species within the spirit and scope of the invention include H. abietis, H. alpestre, H. americanum, H. bharengense, H. botryoides, H. cirrhatum, H. clathroides , H. coralloides , H. novae-zealandiae, H. rajchenbergii, and H. yumthangense .
[159] Various compounds referred to as “hericenones” may be extracted from the fruiting bodies of Hericium spp. (i.e., Lion’s mane), and include, as non-limiting examples: Hericenone A (5-[(2E)-3,7-dimethyl-5-oxoocta-2,6-dienyl]-4-hydroxy-6-methoxy-3H-2- benzofuran-l-one), Hericenone B (6-[(2Z)-3,7-dimethyl-5-oxoocta-2,6-dienyl]-7-hydroxy- 5-methoxy-2-(2-phenylethyl)-3H-isoindol-l-one), Hericenone C ([4-[(2E)-3,7-dimethyl-5- oxoocta-2,6-dienyl]-2-formyl-3-hydroxy-5-methoxyphenyl]methylhexadecanoate), Hericenone DD ([4-[(2E)-3,7-dimethyl-5-oxoocta-2,6-dienyl]-2-formyl-3-hydroxy-5- methoxyphenyl]methyloctadecanoate), Hericenone E ([4-[(2E)-3,7-dimethyl-5-oxoocta- 2,6-dienyl]-2-formyl-3-hydroxy-5-methoxyphenyl]methyl(9E,12E)-octadeca-9,12-di enoate), Hericenone F ([8-formyl-5-methoxy-2-methyl-2-(4-methyl-2-oxopent-3-enyl)-3,4- dihydrochromen-7-yl] methylhexadecanoate), Hericenone G ([8-formyl-5-methoxy-2- methyl-2-(4-methyl-2-oxopent-3-enyl)-3,4-dihydrochromen-7-yl]methyloctadecanoate), Hericenone HH ([8-formyl-5-methoxy-2-methyl-2-(4-methyl-2-oxopent-3-enyl)-3,4- dihydrochromen-7-yl] methyl (9E,12E)-octadeca-9,12-di enoate), Hericenone I (5-Methoxy-2-methyl-2-(4-methyl-2-oxopent-3-en-1-yl)-3,4-dihydro-2H-furo[3,4-h]chromen -7(9H)-one), Hericenone J (6-[(2E)-3,7-dimethylocta-2,6-dienyl]-7-hydroxy-5-methoxy- 3H-2-benzofuran-1-one), and Hericenone K (2-(4-hydroxy-4-methyl-2-oxopentyl)- 5-methoxy-2-methyl-4,7-dihydro-3H-furo[3,4-h]chromen-9-one), 3HF (3-Hydroxy- hericenone F, i.e., [(2S,3S)-8-formyl-3-hydroxy-5-methoxy-2-methyl-2-(4-methyl- 2-oxopent-3-enyl)-3,4-dihydrochromen-7-yl]methylhexadecanoate), DLPE (Dilinoleoyl- phosphatidylethanolamine), Isohericerinol A, Hericerin 6-[(2Z)-3,7-dimethylocta- 2,6-dienyl]-7-hydroxy-5-methoxy-2-(2-phenylethyl)-3H-isoindol-1-one, NDPIH (N-de(phenylethyl)isohericerin, i.e., 5-(3,7-dimethylocta-2,6-dienyl)-4-hydroxy-6-methoxy- 2,3-dihydroisoindol-1-one), and Corallocin A (2E,6E)-8-(4-hydroxy-6-methoxy- 1-oxo-3H-2-benzofuran-5-yl)- 2,6-dimethylocta-2,6-dienoic acid. [160] “L-theanine” refers to L-γ-glutamylethylamide ( N 5 -ethyl-L-glutamine) in enantiomeric form, but also includes unless specified or implied by context the D-enantiomer, as well as racemic DL- mixtures, and non-racemic mixtures of any proportions. [161] “Psilocin” refers to 4-hydroxy-N,N-dimethyltryptamine in pure or substantially pure form, in purified or isolated form produced by biosynthesis or by chemical synthesis, or as extracts from Psilocybe and other psilocin-containing mushrooms, such as those in the genera Conocybe , Copelandia , Galerina , Gymnopilus , Inocybe , Mycena , Panaeolina , Panaeolus , Pholiotina , and Pluteus . Although “ Psilocybe ” mushrooms may be referred to herein as shorthand, one will readily appreciate than any of the above psilocybin-producing genera, as well as other psilocybe-producing genera known in the art, will be within the meaning of the term or be equivalents. Within the scope of psilocin are also its precursors and prodrugs, and the enantiomers, metabolites, polymorphs, salts, derivatives, analogs, and variants thereof. [162] “Psilocybin” refers to 4-phosphoryloxy-N,N-dimethyltryptamine in pure or substantially pure form, in purified or isolated form produced by biosynthesis or by chemical synthesis, or as extracts from Psilocybe and other psilocybin-containing mushrooms, such as those in the genera Conocybe , Copelandia , Galerina , Gymnopilus , Inocybe , Mycena , Panaeolina , Panaeolus , Pholiotina , and Pluteus . Chemically synthesized psilocybin is available as a crystalline solid from, e.g., Cayman Chemical Co., Ann Arbor, MI, with purity ≥ 98%, as Item No.14041, CAS No.520-52-5, or can be synthesized following any of several known methods including as discussed, e.g., in Nichols and Frescas (1999). Biosynthesized psilocybin can be produced in genetically-modified E. coli , S. cerevisiae , and other host cells, and can be commercially sourced ( see, e.g., Fricke, 2017). Within the scope of psilocybin are also its precursors and prodrugs, and further including the enantiomers, metabolites, polymorphs (any of several), salts, derivatives, analogs, and variants thereof. [163] In embodiments wherein psilocybin, psilocin, and/or other psychedelic or psychoactive tryptamines are extracted, extraction may utilize any such extraction means described herein or otherwise known in the art. For extraction of polar molecules, a polar solvent may be used. While a variety of polar solvents may be utilized, water, methanol, and/or ethanol, and including combinations thereof, may be used in embodiments herein. In embodiments, instead of an isolated, purified, extracted, biosynthesized, or synthesized compound, such as a mushroom compound, such as any one or more compounds from Psilocybe , cordyceps, or lion’s mane, ground or pulverized dried mushroom is used directly. That is, the “mushroom composition” of the invention, in some embodiments, may comprise dried Psilocybe , cordyceps, and/or lion’s mane mushroom (in some embodiments, together with a carrier, diluent, or excipient, for example to make it easier to work with and capsulize), in some embodiments, may consist essentially of dried Psilocybe , cordyceps, and/or lion’s mane mushroom, and in some embodiments, may consist of dried Psilocybe , cordyceps, and/or lion’s mane mushroom, or consist of dried mushrooms and other active ingredients. [164] Broadly, extraction systems span from the relatively simple (e.g., those that include a solvent, a funnel, a cheesecloth, etc.), to those large and complex enough to fill a warehouse. Regardless of scale, many generally follow similar principles. In one exemplary embodiment, first, Psilocybe mushrooms are harvested and collected, dried, and pulverized. The pulverized mushrooms are then combined with a solvent capable of extracting the compounds of interest (e.g., methanol, ethanol, water, or a mixture thereof) to form a slurry, and the slurry is then agitated to facilitate extraction for a specified duration of time that is generally up to, or more than, 24 hours in length. The solvent is then filtered and collected, before evaporation of the excess solvent is completed to yield a more concentrated extract. Note, as would be apparent to one of skill, one may additionally decide to re-saturate the mushroom plant material after the initial filtration to complete a secondary, tertiary, etc., extraction. Likewise, one may choose an extraction method different from the exemplary one outlined above. All such cases would be within the spirit of the invention disclosed within—the only requirement being that, depending on the embodiment, an extract comprising the compounds of interest be obtained. [165] In embodiments, desired compounds other than those that are psychedelic or psychoactive also may be obtained through extraction. In such embodiments, the extraction process is like that above, in that fungal matter may or may not be first dried, then ground and pulverized, prior to introducing the ground fungal matter to a solvent, or solvent to the fungal matter, capable of extracting the desired compounds. As would be apparent to one of skill, this may be completed to extract the desired hericenones from lion’s mane, cordycepin from Cordyceps, or any other desired compound from any other such mushroom species. As it relates to the solvent itself, any solvent known in the art (methanol, ethanol, water, a combination thereof, etc.) may effectively extract the desired compounds. As with the psilocybin extraction, the pulverized mushroom and solvent mixture is continuously or substantially continuously agitated for a specified duration of time (e.g., between about 1 and about 24 hours), and excess solvent may be removed through distillation or evaporation. [166] In embodiments, a mushroom composition of the invention is an entourage enhanced extract. In embodiments, an entourage enhanced extract comprises predetermined amounts of two or more of psilocybin, psilocin, norbaeocystin, baeocystin, aeruginascin, norpsilocin, and other psychoactive or “psychedelic tryptamines” from Psilocybe spp. and other psilocybin-containing mushrooms (i.e., “minor tryptamines”). Such compounds may also be referred to herein as “compounds from Psilocybe ,” although as noted, they need not be obtained from Psilocybe , and are found in and may come from other sources. The same will be understood to be true with “compounds from” Hericium and Cordyceps . [167] In embodiments, an entourage enhanced extract is produced through the extraction of a single full spectrum or broad spectrum mushroom extract comprising two or more active compounds of interest. In embodiments, an entourage enhanced extract is produced through the extraction of and/or by the combination of multiple compounds from more than one mushroom species, wherein the mushrooms are mixed (e.g., as fungal matter, such as pulverized fungal matter) before extraction. In embodiments, an entourage enhanced extract is produced through the extraction of multiple compounds (e.g., a broad spectrum or full spectrum mushroom extract) and/or by the combination of two separate extracts, wherein such extracts are individually produced, and combined after production, and comprise multiple compounds of interest. In embodiments, an entourage enhanced extract is a standardized extract, which is standardized for one or more tryptamines such as psilocybin. [168] In an exemplary embodiment, an entourage enhanced extract may be produced by combining more than one mushroom species from genera including Psilocybe (or another psilocybin-containing, or psychoactive tryptamine-containing genus), Cordyceps , and Hericium ; drying, grinding, and pulverizing the mushrooms (and including more broadly, for avoidance of doubt, all such fungal matter), introducing a solvent sufficient to extract the desired compounds (e.g., methanol, ethanol, water, or a mixture thereof), continuously agitating the mixture for between about 1 to about 24 hours, or for more than 24 hours, and then allowing the excess solvent to evaporate under appropriate conditions. In such an embodiment, the mushroom composition may contain any desired composition, such that a first mushroom species may comprise between about 0% to about 99% of the mushroom composition, inclusive; a second mushroom species may comprise between about 0% to about 99% of the mushroom composition, inclusive; and a third mushroom species may comprise between about 0% to about 99% of the mushroom composition, inclusive. In embodiments, a mushroom composition may include compounds from a fourth, a fifth, a sixth, a seventh, an eighth, a ninth, a tenth, or more than a 10th other mushroom species. [169] An entourage enhanced extract also may be produced by combining more than one mushroom extract from at least one of the Psilocybe (or other psilocybin-containing), Cordyceps , and Hericium genera. As apparent to one of skill, extracts to be combined may be sourced in the same, or a different manner, including through use of the same or a different solvent, and under the same, or different conditions, depending on the specific extract profile desired. Once obtained, extracts may be combined in any ratio, such that a first mushroom species may comprise between about 0% to about 99% of the resultant extract, inclusive; a second mushroom species may comprise between about 0% to about 99% of the resultant extract, inclusive; a third mushroom species may comprise between about 0% to about 99% of the resultant extract, inclusive, and the like for up to or greater than 10 mushroom species. [170] One of skill will understand how to calculate how much active psilocybin and/or psilocin is in unprocessed (fresh or dried) fungal matter, for example by reference to concentration guidelines available online (allowing a rough estimate), or more precisely by chemical quantification or analysis, as known to those of skill (e.g., a reagent test). It will additionally be within ordinary skill to calculate how much active psilocybin is in a particular fungal extract, or in a purified extract by using the specific extract ratio, as would be known when producing or sourcing a fungal extract for use in the invention. For instance, the concentration of active compounds in various Psilocybe species is presented in Table 1 below, showing the w/w% of psilocybin and psilocin in dried mushrooms (containing negligible water weight, or sometimes referred to as “cracker dry”): P. P.
Figure imgf000051_0001
P. P. P. P. P. P. P. P. P. P.
Figure imgf000052_0001
[171] Thus, an extract of 100 mg P. cubensis may contain approximately 0.63 mg of psilocybin. An extract of 275 mg P. azurenscens may contain approximately 4.895 mg. [172] In embodiments, a mushroom extract, including an entourage enhanced mushroom extract, does not contain any of psilocybin, psilocin, and/or any other psychedelic tryptamines (i.e., those that would cause psychedelic effects in a human when administered at a psychedelic dose, or cause an equivalent effect in a non-human animal, such as a head-twitch response, as will be readily understood by those of ordinary skill). [173] “Valerian” may refer to extracts of Valeriana officinalis , from the honeysuckle family Caprifoliaceae . Valerian therefore may contain one or more of the known compounds found therein, and preferably found in Valerian root, such as the alkaloids actinidine, chatinine, shyanthine, valerianine, and valerene; isovaleramide; gamma-aminobutyric acid (GABA); valeric acid or isovaleric acid; iridoids, including the valepotriates valtrate and isovaltrate; the sesquiterpenes valerenic acid, hydroxyvalerenic acid, and acetoxyvalerenic acid; and the flavanones hesperidin, 6-methylapigenin, and linarin. In embodiments, only one or more purified biologically active compounds, such as valerenic acid, is included. However, with valerian extract or valerian root extract, as with certain fungal extracts of the invention, a “whole plant” extract (or in some embodiments, a “full spectrum” or “broad spectrum” extract) is preferred over one or more isolated compound(s), to provide synergistic and other “entourage” effects, and the accompanying full range of benefits of whole plant medicines. [174] In certain embodiments of the above-defined compounds, a composition is a mixture of natural compounds. In other embodiments, the composition is a mixture of synthetic or biosynthetic compounds which may reproduce or replicate the effects of a whole plant full spectrum, or broad spectrum extract. In yet other embodiments, the composition is a mixture of natural, synthetic, and/or biosynthetic compounds. Accordingly, it should be understood that the compounds used in the compositions of the invention, where such compounds are as defined above, all may be obtained as natural compounds (e.g., from plants or fungi), or by chemical synthesis or from a bioengineered organism, or as a mixture or combination thereof. [175] “Entourage-enhanced,” in reference to compositions containing such above-defined compounds, refers to a composition so described that has beneficial effects in a human or other mammal, when taken for its intended use, that are greater than the beneficial effects that would be obtained or expected from each of the same compounds when taken alone or in isolated and purified form. Put differently, there are beneficial (and in embodiments, synergistic) effects resulting from the combination that result from the combination in itself (i.e., the fact that each of the various compounds are taken together, in a specific way) and/or from the incorporation and use of whole plant extracts, full- or broad-spectrum extracts, or purified extracts (e.g., concentrated extracts) instead of isolated compounds. Recall, as a model, the original combination that led to coining the term “entourage effect,” above (where two otherwise “inactive” compounds markedly increased the activity of another). [176] It therefore will be readily appreciated that entourage-enhanced compounds may increase an existing therapeutic effect, provide an additional therapeutic effect, increase a desired property such as stability or shelf-life, decrease an unwanted effect or property, alter a property in a desirable way (such as pharmacokinetics or pharmacodynamics), or modulate a desired system or pathway (e.g., a neurotransmitter system such as the serotonergic system). [177] In embodiments, the cannabis compositions contain at least one cannabinoid. In embodiments, the at least one cannabinoid is any of CBC-type cannabinoids, CBD-type cannabinoids, CBE-type cannabinoids, CBG-type cannabinoids, CBL-type cannabinoids, CBN-type cannabinoids, CBND-type cannabinoids, CBT-type cannabinoids, ∆ 8 -THC-type cannabinoids, ∆ 9 -THC-type cannabinoids, and miscellaneous-type cannabinoids. [178] In embodiments, a cannabinoid is a natural cannabinoid (e.g., a phytocannabinoid). In embodiments, a cannabinoid is a synthetic cannabinoid. In embodiments, the cannabis composition comprises more than one cannabinoid. In such embodiments, the cannabinoids may be natural cannabinoids, may be synthetic cannabinoids, or may be a combination of natural and synthetic (including biosynthetic) cannabinoids. [179] In embodiments comprising synthetic cannabinoids, synthetic cannabinoids may include any of AKB48, AZ-11713908, AZD-1940, CP55940, CP-47497, CP-47497 C8, JWH-015, WIN55212-2, HU-210, HU-239, JWH-018, AM–2201, JWH-122, JWH-073, JWH-081, JWH-200, JWH-210, UR-144, and XLR11. In embodiments, synthetic cannabinoids may include pharmaceutical compounds such as nabilone (Cesamet ® ), which is structurally distinct from Δ9 -THC, but mimics its structure and pharmacological activity through weak partial agonist activity at CB 1 and CB 2 receptors, although twice as active. Numerous other synthetic variants and analogs of natural cannabinoids exist, and some are shown to have altered or enhanced activity. Breuer et al. (2016) report, for example, synthesis of fluorinated CBD derivatives, showing greater potency than CBD in behavioral assays in mice predictive of anxiolytic, antidepressant, antipsychotic, and anticompulsive activity. [180] In embodiments, the cannabis composition comprises at least one isoprenoid. “Isoprenoids” may refer to any of the class of organic hydrocarbon isoprene polymers constituted by one or more repeating units of the five-carbon building block known as the isoprene unit (i.e., 2-methyl-1,3-butadiene, C 5 H 8 ), including those compounds known as “terpenoids” and “terpenes.” “Isoprenoids” thus includes such terpenes as those structurally found in linear chains or in rings, and those having any number of isoprene units, i.e., whether as hemiterpenes (one unit), monoterpenes (two), sesquiterpenes (three), diterpenes (four), sesterterpenes (five), triterpenes (six), sesquarterpenes (seven), tetraterpenes (eight), or polyterpenes (nine or more). Isoprenoids may be derived from a botanical source, whether from Cannabis or another plant, or may be synthetic. One of skill, in practicing the invention, will understand where to obtain isoprenoids, and how to balance various factors (e.g., purity, cost, ease of sourcing, marketing and advertising goals and constraints) when determining whether to use natural and/or synthetic versions. In some preferred embodiments, the isoprenoids are terpenes selected from the group consisting of alpha-bisabolol, beta-caryophyllene, camphene, carene, caryophyllene oxide, alpha-humulene, fenchol, guaiene, guaiol, limonene, linalool, myrcene, nerolidol, ocimene, alpha-phellandrene, alpha-pinene, beta-pinene, alpha-terpinene, gamma-terpinene, terpineol, and terpinolene. [181] While, in embodiments, at least one of the terpenes in an entourage-enhancing isoprenoid fraction is selected for its ability to provide therapeutic effects or synergistic effects, or is selected for its ability to modulate the endocannabinoid system, modulate neurosteroid biosynthesis, or modulate a neurotransmitter system, it will be appreciated that terpenes have other effects and uses as well, and may contribute to a composition in multiple distinct ways. For instance, limonene shows stimulating effects, and also exhibits anti-cancer, anxiolytic, and immunostimulating properties. In embodiments, an isoprenoid or terpene may be selected for its aromatic and flavor masking properties which make it suitable or preferable for certain formulations, as will be taught herein or generally known in the art. [182] In embodiments, a Cannabis composition comprises a flavonoid. In embodiments, preferred flavonoids are those from Cannabis , including anthocyanins, anthoxanthins, apigenin, butin, cannaflavins, kaempferol, luteolin, orientin, quercetin, silymarin, and vitexin. In other embodiments, flavonoids are derived from or found in other plant sources. [183] In certain embodiments, a composition comprising cannabinoids, isoprenoids, and/or flavonoids is a mixture of natural compounds. In other embodiments, the composition is a mixture of synthetic compounds. In other embodiments, the composition is a mixture of natural and synthetic compounds. Thus, it will be appreciated that cannabinoids, isoprenoids, and/or flavonoids used in compositions herein all may be obtained as natural compounds (from Cannabis or other plants), as synthetic or biosynthetic compounds (by chemical synthesis or from bioengineered organisms), or as mixtures or combinations thereof. [184] “Entourage-enhancing,” when used to describe the “entourage-enhancing cannabinoid fractions” and “entourage-enhancing isoprenoid fractions,” or any other “entourage-enhancing” fraction of a cannabis composition of the invention (as well as, for example, “entourage-enhancing tryptamine fractions” or other “entourage-enhancing” fractions of a mushroom composition), means that the composition comprising the fraction(s) so described has beneficial effects in a human or other mammal, when taken for its intended use, that are greater than: (1) the beneficial effects that would be obtained or expected from a composition identical in every way, but without comprising those same fraction(s), added to (2) the beneficial effects that would be obtained or expected from those fraction(s) alone. Put differently, there are beneficial effects that result from the combination of the entourage-enhancing fraction(s), that are the result of the combination in itself (i.e., the fact that each of the various compounds are taken together, in a specific way). Recall again, as a model, the original combination that led to coining the term “entourage effect,” above (where two otherwise “inactive” compounds markedly increased the activity of a cannabinoid). [185] It therefore will be readily appreciated that such entourage-enhancing effects may include increasing an existing therapeutic effect, providing an additional therapeutic effect, increasing a desired property such as stability or shelf-life, decreasing an unwanted effect or property, altering a property in a desirable way (such as pharmacokinetics or pharmacodynamics), or modulating a desired system or pathway (e.g., the endocannabinoid system, a neurotransmitter system, or the biosynthesis of a neuroactive steroid). [186] “Therapeutic effects” that are contemplated as being increased in embodiments of the invention include, but are not limited to, antioxidant, anti-inflammatory, analgesic, antineuropathic, antinociceptive, antimigraine, anxiolytic, antidepressant, antipsychotic, anti-PTSD, immunostimulant, anti-cancer, antiemetic, orexigenic, antiulcer, antihistamine, antihypertensive, anticonvulsant, antiepileptic, bronchodilator, neuroprotective, entactogenic, empathogenic, entheogenic, euphoric, psychedelic, sedative, and stimulant effects. [187] “Synergistic effects” will be understood as being “entourage-enhanced” effects, and thus the entourage-enhanced effects of different embodiments of the invention should be understood to include increases in potency, bioactivity, bioaccessibility, bioavailability, or therapeutic effect, that are greater than the additive contributions of the components acting alone, and/or are greater than the contribution of the isolated compounds on their own, if incorporated not as part of a whole plant (including whole fungal) extract. [188] [348] Numerous methods known to those of skill in the art exist to determine whether there is synergy as to a particular effect, i.e., whether, when two or more components are mixed together, the effect is greater than the sum of the effects of the individual components applied alone, thereby producing “1+1 > 2.” Suitable methods include isobologram (or contour) analysis (Huang et al.2019), or the equation of Loewe additivity (Loewe & Muischnek, 1926, Arch. Exp. Pathol Pharmacol.114: 313-326). A synergistic effect also may be calculated using methods such as the Sigmoid-Emax equation (Holford & Scheiner, 1981, Clin. Pharmacokinet.6: 429-453) and the median-effect equation (Chou & Talalay, 1984, Adv. Enzyme Regul.22:27-55). The corresponding graphs associated with the equations referred to above are the concentration-effect curve and combination index curve, respectively. Each equation referred to above may be applied to experimental data to generate a corresponding graph to aid in assessing the effects of the drug combination. [189] The goal of increasing an existing therapeutic effect, providing an additional therapeutic effect, increasing a desired property such as stability or shelf-life, decreasing an unwanted effect or property, altering a property in a desirable way (e.g., pharmacokinetics or pharmacodynamics), modulating a desired system or pathway (e.g., a neurotransmitter system), or otherwise inducing synergy, also can be achieved by inclusion of an additional active compound. Such additional active compounds may include any one or more of amino acids, antioxidants, anti-inflammatory agents, analgesics, antineuropathic and antinociceptive agents, antimigraine agents, anxiolytics, antidepressants, antipsychotics, anti-PTSD agents, immunostimulants, anti-cancer agents, antiemetics, orexigenics, antiulcer agents, antihistamines, antihypertensives, anticonvulsants, antiepileptics, bronchodilators, neuroprotectants, entactogens and empathogens, entheogens, psychedelics, nootropics, monoamine oxidase inhibitors, sedatives, stimulants, supplements, and vitamins. These ingredients may be in ion, freebase, or salt form, and may be isomers or prodrugs. [190] In embodiments, an extract containing at least one cannabinoid may be obtained, wherein the at least one cannabinoid is one of, or more than one of a cannabinoid selected from the group consisting of CBC-type cannabinoids, CBD-type cannabinoids, CBE-type cannabinoids, CBG-type cannabinoids, CBL-type cannabinoids, CBN-type cannabinoids, CBND-type cannabinoids, CBT-type cannabinoids, ∆ 8 -THC-type cannabinoids, ∆ 9 -THC-type cannabinoids, and miscellaneous-type cannabinoids; wherein, in some embodiments, the at least cannabinoid is one of, or more than one of THC, CBD, CBC, CBND, CBDL, CBE, CBL, CBT, CBT, CBV, CBGM, CBDP, THCP, and iso-THC; as well as their acidic forms, their propyl, methyl, and ethyl homologues, and the acid forms of those homologues forms (i.e., their acidic propyl, acidic methyl, and acidic ethyl homologue forms). In such embodiments, the extract may be produced by any means known to those of skill. An exemplary extraction system that should not be construed as limiting is as follows: Obtaining dried cannabis flower, grinding and optionally pulverizing the flower, and placing it into an extraction system generally comprising a loop wherein chilled ethanol (kept at between -30C˚ and -40 ˚C) is circulated. The resultant extract is then evaporated, so as to remove the latent solvent, and then distilled—which occurs at a higher temperature and pressure than the evaporation stage—so as to remove impurities, and further concentrate the extract. In such an embodiment, decarboxylation occurs, which removes the carboxylic acid group from the THC molecule, rendering it “active” (i.e., in a state where it will cause mind-altering effects characterized by THC ingestion or inhalation) (Harli, 2021). [191] In an embodiment wherein mind-altering or intoxicating effects are not desired, one may choose an extraction method that does not heat the compounds to such a temperature/pressure wherein decarboxylation occurs (roughly 110 C˚ at standard pressure), or may remove the THC from the extract via, among other means, column chromatography. Accordingly, compounds intended for ingestion that comprise THCA, wherein such THCA will not be decarboxylated prior to or during administration, will not be considered “C+” compositions within the meaning of the invention. By contrast, compounds intended for ingestion that comprise THCA, wherein such THCA will be decarboxylated prior to or during administration, and therefore are capable of providing intoxicating effects as the neutral form of THC, will be considered “C+” compositions within the meaning of the invention. Compositions comprising other acidic cannabinoids will be considered C+ or C– similarly. [192] All of the compounds described herein may contain one or more asymmetric centers and give rise to enantiomers, diastereomers, and other stereoisomeric forms. Each chiral center may be defined, in terms of absolute stereochemistry, as (R)- or (S)-. The present invention is meant to include all such possible isomers, as well as mixtures thereof, including racemic and optically pure forms. Optically active (R)- and (S)-, (-)- and (+)-, or (D)- and (L)-isomers may be prepared or resolved using conventional techniques. When the compounds described herein contain olefinic double bonds or other centers of geometric asymmetry, and unless specified otherwise, it is intended that the compounds include both E and Z geometric isomers. Likewise, tautomeric forms are also intended to be included. [193] All compounds disclosed will be understood to include the pharmaceutically acceptable salts of such compounds. “Pharmaceutically acceptable salts” refers to salts prepared from pharmaceutically acceptable non-toxic acids or bases, using conventional chemical methods. Generally, such salts are prepared by reacting the free acid or base forms of a compound with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two. “Pharmaceutically acceptable salt” when generally referring to a salt of a disclosed compound will be understood to mean a salt which is pharmaceutically acceptable and possesses the desired pharmacological activity of the parent compound ( see, e.g., Berge et al., 1977). Production of the salts of natural and synthetic cannabinoid carboxylic acids is disclosed, e.g., in U.S. Pat. No.9,376,367. [194] The compounds used in the practice of the invention now having been generally described will be more readily understood by further reference to the following description and examples, which are included for the purposes of illustration of certain aspects of the embodiments of the present invention. The following is not intended to limit the invention, as one of skill would recognize from the teachings and examples herein that other techniques and methods can satisfy the claims and be employed without departing from the scope of the invention. Indeed, while this invention has been particularly shown and described with reference to certain exemplary embodiments, it will be understood by those skilled in the art that various changes in form and details may be made without departing from the scope or spirit of the invention encompassed by the appended claims. M. Nutraceutical and Pharmaceutical Compositions and Formulations [195] Provided are “nutraceutical compositions,” “pharmaceutical compositions” and “formulations,” and methods of their administration. Herein, “compositions” and “formulations” may be used interchangeably, unless context dictates otherwise, or one such term is needed to give technical effect. Compositions and formulations include “mushroom compositions” and “cannabis compositions,” such as M+, M–, C+, and C– compositions. [196] In embodiments, the mushroom compositions and cannabis compositions are administered, e.g., to a patient in need thereof. In embodiments, the mushroom compositions and cannabis compositions comprise a pharmaceutically acceptable carrier, diluent, or excipient. It will be appreciated, however, that some embodiments do not have any carrier, diluent, or excipient (e.g., a composition will comprise one or more plant or fungi extracts or compounds alone without carriers, diluents, or excipients), and some embodiments do not have a single carrier, diluent, or excipient alone, but include multiple carriers, diluents, and/or excipients. In some preferred embodiments, the carrier, diluent, and/or excipient (including all such carriers, diluents, and/or excipients used in a composition) will not be from a fungal source or found in fungi. In embodiments, such carriers, diluents, and/or excipients not from a fungal source or found in fungi will further not be found in any natural source, such as any botanical source, and will not be able to be obtained as a natural product or as products in or of nature. In embodiments, such carriers, diluents, and/or excipients will further have markedly different characteristics than any found in nature. In embodiments, such carriers, diluents, and/or excipients will contribute to the composition as a whole such characteristics or qualities that the composition as a whole is transformed and has markedly different characteristics than any such combination of natural products found in nature. [197] Compositions can be prepared by standard pharmaceutical formulation techniques such as described below or disclosed in, e.g., Remington: The Science and Practice of Pharmacy (2005) 21th ed., Mack Publ. Co., Easton, Pa.; The Merck Index (1996) 12th ed., Merck Publ. Group, Whitehouse, N.J.; Pharm. Principles of Solid Dosage Forms (1993), Technomic Publ. Co., Inc., Lancaster, Pa.; and Ansel and Stoklosa, Pharm. Calculations (2001) 11th ed., Lippincott Williams & Wilkins, Baltimore, Md.; and Poznansky et al. Drug Delivery Systems (1980), R.L. Juliano, ed., Oxford, N.Y., pp.253-315). [198] “Pharmaceutically acceptable” in connection with one or more ingredients means the ingredient(s) are generally safe and, within the scope of sound medical judgment, suitable for use in contact with cells of humans and other animals without undue toxicity, irritation, allergic response, or complication, and commensurate with a reasonable risk/benefit ratio. [199] In embodiments, pure or substantially pure individual compounds described herein are administered as part of the mushroom compositions and cannabis compositions disclosed herein. The terms “pure” or “substantially pure,” as used herein, refer to material that is substantially or essentially free from components that normally accompany the material when the material is synthesized, manufactured, or otherwise produced. A “pure” or “substantially pure” preparation of a compound is accordingly defined as a preparation having a chromatographic purity (of the desired compound) of greater than 90%, more preferably greater than 95%, more preferably greater than 96%, more preferably greater than 97%, more preferably greater than 98%, more preferably greater than 99%, more preferably greater than 99.5%, and most preferably greater than 99.9%, as determined by area normalization of an HPLC profile or other similar detection method. Preferably the pure or substantially pure compound used in the invention is substantially free of any other active compounds which are not intended to be administered to a subject. In this context, “substantially free” refers to the fact that no active compound(s), other than the active compound intended to be administered to a subject, are detectable by HPLC or another similar detection method, or are below a desired threshold of detection such as defined above. a. Compositions, Dosage Forms, and Methods for Preparing the Same [200] In embodiments, the mushroom compositions and cannabis compositions disclosed herein can be formulated into any suitable dosage form, including aqueous oral dispersions, aqueous oral suspensions, solid dosage forms including oral solid dosage forms, aerosols, controlled release formulations, fast melt formulations, effervescent formulations, self-emulsifying dispersions, solid solutions, liposomal dispersions, lyophilized formulations, tablets, capsules, pills, powders, delayed-release formulations, immediate-release formulations, modified release formulations, extended-release formulations, pulsatile release formulations, multi particulate formulations, and mixed immediate release and controlled release formulations. Generally speaking, one will desire to administer an amount of the active agents of a composition that is effective to achieve a plasma level commensurate with the concentrations found to be effective in vivo for a period of time effective to elicit the desired therapeutic effect(s). As noted above, as shorthand and for ease and simplicity of reference, nutraceutical and/or pharmaceutical compositions, including “mushroom compositions” and “cannabis compositions,” may be referred to collectively as “formulations,” for instance when prepared with at least one carrier, diluent, or excipient, or when prepared according to any of the non-limiting exemplary formulation examples below. [201] In embodiments, formulations are in unit dosage form. “Unit dosage form” may refer to a physically discrete unit suited as a unitary dosage for a subject to be treated, comprising a predetermined quantity of a composition. Unit dosage forms may be used for ease of administration and uniformity of dosage. Unit dosage forms can contain a single or individual dose or unit, a sub-dose, or an appropriate fraction thereof of a composition administered. [202] Unit dosage forms may include capsules, troches, cachets, lozenges, tablets, ampules and vials, which may include a composition in a freeze-dried or lyophilized state; a sterile liquid carrier, for example, can be added prior to administration or delivery. Unit dosage forms may also include ampules and vials comprising a composition in aqueous or liquid form. Unit dosage forms may be for transdermal administration, such as “patches” that can be adhered to the epidermis of a subject, comprising a predetermined amount of a composition. [203] In embodiments, the formulations are in a pharmaceutically acceptable oral dosage form, including oral solid dosage forms and oral liquid dosage forms. In embodiments, the formulations are a pharmaceutically acceptable oral solid dosage form. Oral solid dosage forms may include but are not limited to, lozenges, troches, tablets, capsules, caplets, powders, pellets, multiparticulates, beads, spheres, and/or any combinations thereof. Oral solid dosage forms may be formulated as immediate release, controlled release, sustained release, extended release, or modified release formulations. [204] In embodiments, the solid dosage provided herein, such as oral solid dosage forms, may be in the form of a tablet (including a suspension tablet, a fast-melt tablet, a bite-disintegration tablet, a rapid-disintegration tablet, an effervescent tablet, or a caplet), a pill, a powder (including a sterile packaged powder, a dispensable powder, or an effervescent powder), a capsule (including both soft or hard capsules, e.g., capsules made from animal-derived gelatin or plant-derived HPMC, or “sprinkle capsules”), solid dispersion, solid solution, bioerodible dosage form, controlled release formulations, pulsatile release dosage forms, multiparticulate dosage forms, pellets, granules, or an aerosol. In other embodiments, the formulation is in the form of a powder. In still other embodiments, the formulation is in the form of a tablet, including a fast-melt tablet. In embodiments, formulations may be administered as a single capsule or in multiple capsule dosage form. In embodiments, a formulation is administered in two, three, four, or more capsules or tablets. [205] In embodiments, solid dosage forms may contain pharmaceutically acceptable excipients such as fillers, diluents, lubricants, surfactants, glidants, binders, dispersing agents, suspending agents, disintegrants, viscosity-increasing agents, film-forming agents, granulation aids, flavoring agents, sweetener, coating agents, solubilizing agents, and combinations thereof. In embodiments, solid dosage forms also can comprise one or more pharmaceutically acceptable additives, as well as supplementary agent(s). In embodiments, supplementary agents include preservatives, antioxidants, antimicrobial agents including biocides and biostats such as antibacterial, antiviral and antifungal agents. [206] In embodiments, tablets provided herein are prepared by methods well known in the art. Various methods for the preparation of the immediate release, modified release, controlled release, and extended-release dosage forms (e.g., as matrix tablets having one or more modified, controlled, or extended-release layers) and the vehicles therein are well known in the art. In embodiments, a tablet may be made by compression or molding. In embodiments, compressed tablets may be prepared by compressing, in a suitable machine, an active ingredient in a free-flowing form such as a powder or granules, optionally mixed with a binder, lubricant, inert diluent, preservative, surface-active or dispersing agent. In embodiments, molded tablets may be produced by molding, in a suitable apparatus, a mixture of powdered compound moistened with an inert liquid diluent. In embodiments, the tablets may optionally be coated or scored and may be formulated so as to provide a slow or controlled release of the active ingredient therein. Generally recognized compendia of methods include Remington 2020 and Sheth et al.1980. [207] In embodiments, solid dosage forms are prepared by mixing the active agents of the invention with one or more pharmaceutical excipients to form a “bulk blend” composition. In embodiments, the bulk blend composition is homogeneous, i.e., the active agents are dispersed evenly throughout so that the bulk blend may be readily subdivided into equally effective unit dosage forms, such as tablets, pills, and capsules. In embodiments, the individual unit dosages may also comprise film coatings, which disintegrate upon oral ingestion or upon contact with diluents. In embodiments, these formulations are manufactured by conventional pharmaceutical techniques. [208] Conventional pharmaceutical techniques for preparation of solid dosage forms include, but are not limited to, the following methods, which may be used alone or in combination: (1) dry mixing, (2) direct compression, (3) milling, (4) dry or non-aqueous granulation, (5) wet granulation, or (6) fusion (see, e.g., Lachman et al.1986). Other methods include spray drying, pan coating, melt granulation, granulation, fluidized bed spray drying or coating (e.g., Wurster coating), tangential coating, top spraying, tableting, and extruding. [209] In embodiments, the formulations have a specific release profile. For example, oral solid dosage forms may be prepared as immediate release or as modified release formulations, e.g., controlled release, extended release, sustained release, or delayed release. [210] In embodiments of immediate release formulations, a therapeutically effective amount of one or more active compounds is administered to facilitate rapid release. Immediate release formulations may be prepared by combining a superdisintegrant such as croscarmellose sodium and different grades of microcrystalline cellulose in various ratios. [211] In embodiments of modified release formulations, the plasma half-life compared to the plasma half-life of an immediate release formulation is greater by at least 5%, at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 50%, at least 75%, at least 100%, or values in between. In embodiments of modified release formulations, the formulations are designed to result in a comparable area under the curve, or AUC 0-24 , and a similar safety and efficacy profile, but having a delayed time to maximum concentration (t max ) of at least 5%, at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 50%, at least 75%, at least 100%, or values in between, as would be appreciated by one of skill. In embodiments, a formulation is designed to be a product with a specific time course based on an optimum “therapeutic window,” such as less than about 30 minutes, about 30 minutes, about 45 minutes, about 60 minutes, about 90 minutes, about 2 hours, about 3 hours, about 4 hours, about 5 hours, about 6 hours, about 7 hours, about 8 hours, and more than 8 hours, and lengths of time in between. [212] In embodiments, oral solid dosage forms are formulated as a delayed release dosage form by utilizing an enteric coating to affect release in the small intestine of the gastrointestinal tract. An enteric-coated oral dosage form may be a compressed or molded or extruded tablet/mold (coated or uncoated) containing granules, powder, pellets, beads or particles of the active ingredient and/or other composition components, which are themselves coated or uncoated. The enteric-coated oral dosage form may also be a capsule (coated or uncoated) containing pellets, beads or granules of the solid carrier or the composition, which are themselves coated or uncoated. In embodiments, enteric coatings may be used to prepare other controlled release dosage forms, including but not limited to extended release and pulsatile release dosage forms. Pulsatile release dosage forms may be formulated using techniques known in the art, such as those described in U.S. Pat. Nos.5,011,692, 5,017,381, 5,229,135, and 5,840,329. Other suitable dosage forms are described in U.S. Pat. Nos. 4,871,549, 5,260,068, 5,260,069, 5,508,040, 5,567,441 and 5,837,284.
[213] In embodiments, the controlled release dosage form is a pulsatile release solid oral dosage form comprising at least two groups of particles, each containing active agents of the invention. In embodiments, the first group of particles, upon ingestion by a subject, provides a substantially immediate dose of the active agents of the invention, and may either be uncoated, or comprise a coating and/or sealant. In embodiments, using such means, a single unit dosage form can provide both a first and a second dosage amount in the single form (i.e., a first dosage amount in an immediate release form, and a second dosage amount in a delayed release form). In embodiments, gastroretentive sustained release tablets are formulated by using a combination of hydrophilic polymer (e.g., hydroxypropyl methylcellulose), together with at least one swelling agent (e.g., crospovidone, sodium starch glycolate, and croscarmellose sodium), and an effervescent substance (e.g., sodium bicarbonate). Using known methods, gastroretentive tablets can be formulated to prolong the gastric emptying time and extend the mean residence time (MRT) in the stomach for optimal drug release and absorption (see, e.g., Arza 2009). In embodiments, coatings for providing a controlled, delayed, or extended release may be applied to the compositions of the invention or to a core containing the compositions, and may comprise a pharmaceutically acceptable ingredient in an amount sufficient to provide a delayed release from, for example, about 1 hour to about 7 hours following ingestion before release of the active agents. In embodiments, suitable coatings include one or more differentially degradable coatings including pH-sensitive coatings (enteric coatings), or non-enteric coatings having variable thickness to provide differential release of the active agents. Many other types of modified release systems will be known to those of skill in the art. Non-limiting examples of additional delivery systems include both polymer- and nonpolymer-based systems, silastic systems, peptide-based systems, wax coatings, bioerodible dosage forms, and compressed tablets using conventional binders (see, e.g, Liberman et al. 1990; Singh et al. 2002; U.S. Pat. Nos. 4,327,725; 4,624,848; 4,968,509; 5,461,140; 5,456,923; 5,516,527; 5,622,721; 5,686,105; 5,700,410; 5,977,175; 6,465,014; and 6,932,983).
[214] In embodiments, the formulation is a pharmaceutically acceptable oral liquid dosage form. Non-limiting examples of oral liquid dosage forms include tinctures, drops, emulsions, syrups, elixirs, suspensions, and solutions, and the like. In embodiments, oral liquid dosage forms are formulated with any pharmaceutically acceptable excipient known to those of skill for preparation of liquid dosage forms, and with solvents, diluents, carriers, excipients, and the like, chosen as appropriate to the solubility and other properties of the active agents and other ingredients. Non-limiting examples of solvents include, e.g., water, glycerin, simple syrup, alcohol, medium chain triglycerides (MCT), and combinations thereof. [215] In embodiments, oral liquid dosage forms may be monophasic or biphasic, the former being a substantially homogenous solution dissolved in water or non-aqueous solvent, while the latter refers to oral liquid dosage forms in which the active ingredients do not fully dissolve in common solvents. In embodiments, over time, the solid particles within a oral liquid dosage form may form a precipitate at the bottom of the container, requiring shaking to redisperse them. Non-limiting examples of monophasic liquid forms include syrups, linctuses, spirits/essences, elixirs, and fluid extracts. Non-limiting examples of biphasic liquid forms include oral suspensions, oral emulsions, and mixtures. [216] Liquid dosage forms for oral administration may be prepared as liquid suspensions or solutions using a sterile liquid, such as but not limited to, an oil, water, an alcohol, combinations of pharmaceutically suitable surfactants, suspending agents, and emulsifying agents. In embodiments, liquid formulations also may be prepared as single dose or multi-dose beverages. In embodiments, suspensions may include oils. Such oils include but are not limited to peanut oil, sesame oil, cottonseed oil, corn oil, and olive oil. Suitable oils also include carrier oils such as MCT and long chain triglyceride (LCT) oils. In embodiments, as suspension preparation may also contain esters of fatty acids such as ethyl oleate, isopropyl myristate, fatty acid glycerides, and acetylated fatty acid glycerides. In embodiments, suspension formulations may include alcohols, such as ethanol, isopropyl alcohol, hexadecyl alcohol; glycerol, and propylene glycol. In embodiments, ethers, such as polyethylene glycol; petroleum hydrocarbons, such as mineral oil and petrolatum; and water may also be used in suspension formulations. In embodiments, a suspension is an aqueous liquid or a non-aqueous liquid, an oil-in-water liquid emulsion, or a water-in-oil emulsion. [217] Dosage forms for oral administration may be aqueous suspensions such as aqueous oral dispersions, emulsions, solutions, and syrups ( see, e.g., Singh et al.2002). In addition to the active agents, the liquid dosage forms may comprise additives, such as one or more (a) disintegrating agents, (b) dispersing agents, (c) wetting agents, (d) preservatives, (e) viscosity enhancing agents, (f) sweetening agents, and/or (g) flavoring agents. In addition to the additives listed above, the liquid formulations of the invention, in some embodiments, may also comprise inert diluents commonly used in the art such as water or other solvents, solubilizing agents, emulsifiers, flavoring agents, and/or sweeteners. In embodiments, co-solvents and adjuvants also may be added to a formulation. [218] In embodiments, effervescent powders containing the compositions are prepared. In embodiments, effervescent salts are used to disperse medicines in water for oral administration. In embodiments, effervescent salts also may be packaged as single dose or multi-dose drink mixes, alone or in combination with other ingredients, such as vitamins or electrolytes. In embodiments, effervescent salts are granules or coarse powders containing a medicinal agent in a dry mixture, usually composed of sodium bicarbonate and sodium carbonate, citric acid, and/or tartaric acid. In embodiments, when salts of the invention are added to water, the acids and the base react to liberate carbon dioxide gas, thereby causing “effervescence.” In embodiments, any acid-base combination that results in the liberation of carbon dioxide may be used, as long as the ingredients are suitable for pharmaceutical use, and result in a pH of about 6.0 or higher. [219] In embodiments, the formulations are delivered transdermally. Generally speaking, transdermal delivery involves contacting a formulation with a subject’s skin under conditions effective for the active agent(s) in a composition to penetrate the skin and cause an effect. Non-limiting examples of transdermal formulations include ointments, creams, suspensions, lotions, pastes, gels, sprays, foams, oils, and the like, and combinations thereof. [220] An exemplary transdermal delivery form is a transdermal “patch” which contains a formulation. Transdermal patches may be used to provide continuous or discontinuous infusion of the compounds described herein in controlled amounts. Such patches may be constructed for continuous, gradual, pulsatile, or on demand delivery of active agents. In embodiments, a patch will be a medicated adhesive patch, i.e., a patch impregnated with a composition of the invention for application onto the skin. In embodiments, a patch may be a single-layer or multi-layer drug-in-adhesive patch, wherein the one or more adhesive layers also contain the active agents. In embodiments, a patch also may be a “matrix” (or “monolithic”) patch, wherein the adhesive layer surrounds and overlays the drug layer (wherein a solution or suspension of the active agents is in a semisolid matrix). In embodiments, a “reservoir” patch may also be used, comprising a drug layer, typically as a solution or suspension of the active agents in a liquid compartment (i.e., the reservoir), separate from an adhesive layer. In embodiments, a patch also may be part of a delivery system, for instance used with an electronic device communicatively coupled to the mobile device of a user, and coupled with a mobile application (e.g., to control the delivery rate from the reservoir, and optionally to provide information about delivery back to the application or user). Various transdermal patch technologies may be accordingly utilized. [221] In embodiments, formulations are for subcutaneous, intravenous, intra-arterial, intraperitoneal, intraosseous, intramuscular, intrathecal, or intracerebroventricular injection. In embodiments, injection formulations are prepared by dissolving, suspending, or emulsifying active agent(s) in an aqueous or nonaqueous solvent, examples of which include oils, such as vegetable oil, synthetic aliphatic acid glycerides, and esters of higher aliphatic acids or propylene glycol; and may also contain additives such as solubilizers, stabilizers, and suspending, preserving, wetting, emulsifying, dispensing, and isotonic agents. [222] In embodiments are disclosed pharmaceutically acceptable nanostructured formulations, such as a nanoemulsion, a nanocapsule, a nanoparticle conjugate, or a nano-encapsulated oral or nasal spray. In embodiments, preparations of the compositions of the invention as certain nanostructured formulations may be prepared by reference to the general knowledge of the art ( see, e.g., Jaiswal 2015). The prefix “nano” as used in the terms describing various embodiments of a nanostructured formulation denotes a size range in the nanometer (“nm”) scale. Accordingly, sizes of such nanoparticle delivery vehicles include those in the range of about 1 to about 100 nm, about 100 to about 200 nm, about 200 to about 400 nm, about 400 to about 600 nm, about 600 to about 800 nm, and about 800 to about 1000 nm, as well as “microparticles” in the range of about 1000 to about 2000 nm (1-2 micrometer (“μm”) scale). Particles of certain sizes may be particularly advantageous depending on the method of administration, as will be immediately appreciated by one of skill (e.g., for oral liquid emulsion versus for transdermal or topical application). In embodiments, lipid-based nanoparticles (LBNPs) such as liposomes, solid lipid nanoparticles (SLN), or nanostructured lipid carriers (NLC) are used. [223] In embodiments, the active ingredients of a formulation are mixed with an excipient, diluted by an excipient, or enclosed within, encapsulated by, or attached to a carrier in the manufacture of the formulation. In embodiments, the excipient serves as a diluent, it can be a solid, semi-solid, or liquid material, which acts as a vehicle, carrier, or medium for the active ingredient. In embodiments, the formulations can be in the form of, e.g., tablets, pills, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, aerosols (as a solid or in a liquid medium), ointments, soft or hard gelatin capsules, suppositories, sterile injectable solutions, and sterile packaged powders. In embodiments, it may be necessary to mill the active agent to provide the appropriate particle size prior to combining with the other ingredients. In embodiments, if an active agent is substantially insoluble, it ordinarily is milled to a particle size of less than about 200 mesh. Different embodiments include immediate, delayed, extended, and controlled release forms. Many other variations are possible and known to those skilled in the art. [224] In embodiments, compositions of the invention are combined with other compounds, including active compounds (i.e., those providing or capable of providing a biological effect), such as vitamins, antioxidants, amino acids (including natural L-amino acids, as well as D- and DL- amino acids, and their analogs and derivatives), probiotics, natural herbs or plant extracts, and/or food or dietary supplements, to form patches, masks, wraps, creams, gels, oral films, or the like, for various purposes such as for energy, reduced fatigue, or improved mental and physical performance (e.g., with guarana, amino acids, and vitamins), for improved performance and reduced gastrointestinal discomfort during exercise (e.g., with L-leucine, L-isoleucine, L-valine, Vitamin B1, and Vitamin B5), for osteoporosis or function of bones and teeth (e.g., with vitamins C, D3, and K2), for sexual dysfunction, improved sexual vigor, or fertility (e.g., with vitamins, amino acids, and plant extracts such as maca root, ginkgo biloba, tributes terrestris, minus pinaster, muir puma, damiana, or catuaba), for improved circulation or to sooth heavy legs (e.g., with vitas vinifera extract and bioflavonoids), for relaxation and sleep (e.g., with melatonin, passionflower, Californian poppy, and other herbal ingredients), or to relieve muscle and joint pain or trauma, (e.g., with hemp seed oil, arnica, and harpagophytum). [225] By way of non-limiting and merely suggestive examples, the following formulations may be used in the methods of the present invention. b. Synergistic Mushroom Compositions EXAMPLE 1: Formulation of “M+” capsules Gelatin capsules, containing approximately 1 g of active ingredients, are made as follows:
Figure imgf000068_0001
The psilocybe, L-theanine, niacin, and cordyceps and lion’s mane extracts are obtained (or prepared into extracts, from obtained raw materials), blended, passed through a mesh sieve (e.g., between No.10 and 20 U.S., or to obtain another suitable particle size), further ground or milled if necessary, and filled into hard or soft gelatin capsules in 1 g quantities (optionally, together with valerian root extract and/or PEA if desired; if not included, the total 1 g quantity will be reduced accordingly). If useful for ease of blending or filling, a suitable amount of cellulose, starch, or magnesium stearate can be added. Purified extracts may be used, with quantities adjusted accordingly, based on concentration. In some preferred embodiments, standardized extracts are used, or the amount of an active agent of interest (e.g., psilocybin or another bioactive compound) is quantified. One will readily appreciate that the amount of extract may be increased or decreased to provide a known or predetermined amount of an active agent of interest. For example, in one exemplary embodiment of this Example, 1 mg of psilocybin is desired per capsule. It is determined that a Psilocybe extract comprises 1% w/w of psilocybin (or a standardized extract, containing 1% w/w of psilocybin is created or obtained).100 mg of Psilocybe extract is therefore used, resulting in a capsule with 1 mg (i.e., 1% w/w of 100 mg) psilocybin. Isolated compounds also may be used, and quantities adjusted accordingly, using known methods. In an alternative “ M– ” embodiment, the formulation is made as above, but without Psilocybe extract, and not containing psilocybin, psilocin, or any other psychedelic tryptamines. EXAMPLE 2: Alternative formulation of “M+” capsules
Figure imgf000069_0001
The formulation of EXAMPLE 2 is created in the same way as that of EXAMPLE 1 , except that valerian root extract and PEA are replaced with rice hull concentrate powder (nu-flow), rice bran extract powder (nu-rice), and rice bran 100 mesh powder ribran 100. In an alternative “ M– ” embodiment, the formulation is made as above, but without psilocybe extract, and not containing psilocybin, psilocin, or any other psychedelic tryptamines. EXAMPLE 3: Alternative formulation of “M+” capsules
Figure imgf000070_0001
The formulation of EXAMPLE 3 is created in the same way as that of EXAMPLE 2. In an alternative “ M– ” embodiment, the formulation is made as above, but without Psilocybe extract, and not containing psilocybin, psilocin, or any other psychedelic tryptamines. EXAMPLE 4: Formulation of “M+” tablets Tablets, each containing 790 mg of active ingredients are made as follows:
Figure imgf000070_0002
The components are blended and compressed to form tablets, each weighing 1 g. In an alternative “ M– ” embodiment, the formulation is made as above, but without Psilocybe extract, and not containing psilocybin, psilocin, or any other psychedelic tryptamines. EXAMPLE 5: Formulation of “M+” capsules with an additional active agent Capsules comprising an M+ composition and an additional active agent are made as follows:
Figure imgf000071_0001
The active ingredients are blended, passed through a mesh sieve, and filled into hard or soft gelatin capsules. The antidepressant or anxiolytic may be any pharmaceutical agent known to act as such by one of skill (e.g., SSRIs, serotonin-norepinephrine reuptake inhibitors (SNRIs), tricyclic antidepressants (TCAs), atypical antidepressants, benzodiazepines, buspirone, etc.). Antidepressants and anxiolytics may be chosen for their ability to act as 5-HT1A agonists. At higher concentrations, CBD also directly activates 5-HT1A receptors, further conferring an anti-anxiety effect. If useful for ease of blending or filling, cellulose, a starch, or magnesium stearate can be added. In an alternative “ M– ” embodiment, the formulation is made as above, but without Psilocybe extract, and not containing psilocybin, psilocin, or any other psychedelic tryptamines. EXAMPLE 6: Formulation of “M+” capsules with additional active agents Capsules, each containing a composition of present invention with additional active agents, are made as follows:
Figure imgf000071_0002
Figure imgf000072_0002
The active ingredients are blended, passed through a mesh sieve, and filled into hard of soft gelatin capsules, for example in 675 mg quantities. The analgesic or anti-inflammatory may be any agent known to act as such (including both OTC and prescription medications) by one of skill (e.g., aspirin, acetaminophen, ibuprofen, naproxen, prescription NSAIDs, etc.). If useful for ease of blending or filling, cellulose, a starch, or magnesium stearate can be added. In an alternative “ M– ” embodiment, the formulation is made as above, but without Psilocybe extract, and not containing psilocybin, psilocin, or any other psychedelic tryptamines. EXAMPLE 7: Formulation of “M+” suspension or solution, such as a tincture Liquid suspensions or solutions, with below amounts per 1.0 ml dose, are made as follows:
Figure imgf000072_0001
The active agents are measured out, blended, passed through a mesh sieve, and then mixed with a previously made solution of microcrystalline cellulose and sodium carboxymethyl cellulose in excipient (e.g., ethanol, for a tincture, or purified water). The sodium benzoate, flavor, and color are diluted with excipient and added with stirring. Sweetener (e.g., sucrose or sucralose) may be added if desired. Additional compounds may be included, e.g., vitamins, other active ingredients such as antioxidants or antiinflammatories herbal extracts and essential oils. Sufficient further excipient is then added to produce the required volume. Suspensions and solutions may be prepared in volumes of 5 ml, 10ml, 25 mL, 30 mL, 50 mL, 100 mL, or such other total volumes as practical for research use or for sale as a pharmaceutical or OTC preparation, a nutraceutical preparation or dietary supplement, or a natural product. Depending on unit dosage volume and total volume, liquid formulations may be used to prepare dropper bottles (e.g., 30 mL/1 oz. bottles) or fine mist spray (i.e., oral spray) bottles (e.g., 10 mL bottles). Liquid formulations of this Example also can be used to prepare filled softgel capsules, ampoules, or other single unit dosage forms, using methods herein disclosed or known to those of skill in the art. In an alternative “ M– ” embodiment, the formulation is made as above, but without Psilocybe extract, and not containing psilocybin, psilocin, or any other psychedelic tryptamines. [226] It will be appreciated that the above formulation examples are illustrative only. Hence, it will be understood that reference to particular compound(s) is likewise illustrative, and the compound(s) in any Example may be substituted by other compound(s) of the invention. For any compound referred to as an “extract,” a “purified extract” or an isolated compound may be used in its place. For example, 100 mg of “Psilocybe extract” from Psilocybe cubensis fungal material may be replaced with 10 mg of “Psilocybe purified extract” from such material at a 10x concentration, or an equivalent amount (e.g., vis-à-vis psilocybin) of purified extract at a 2x, 3x, 4x, 5x, 6x, 7x, 8x, 9x, or greater than 10x concentration, or may be replaced with an equivalent milligram amount of isolated and purified natural psilocybin, synthetic psilocybin, or biosynthetic psilocybin, calculated based on ordinary skill, all of which will be appreciated to provide like effects (if speaking of therapeutic effect; or, if speaking of psychedelic “effects,” a like absence of such effects, if so desired). c. Synergistic Cannabis Compositions EXAMPLE 8: Formulation of “C+” oil Liquid suspensions, each containing the below amounts per 1.0 ml dose are made as follows:
Figure imgf000073_0001
Cannabinoids can be commercially sourced or obtained from, e.g., Cayman Chemical Co., Ann Arbor, MI. The CBD, THC, cannabinoids, and isoprenoids are measured out, blended, passed through a No.10 mesh U.S. sieve, and then mixed with a carrier oil as an excipient, such as MCT oil, coconut oil, or hemp seed oil, and optionally a polyol (e.g., vegetable glycerin) and/or a lecithin (e.g., soy or sunflower lecithin). “Weight percent,” “wt %,” or “% w/w” will be understood (here, and below, and similarly for example with regard to the mushroom compositions, unless otherwise defined) to mean as a percentage of the total cannabinoid+isoprenoid content, and further including the flavonoid content (where applicable). In an embodiment, the entourage-enhancing cannabinoid fraction contains CBG, CBN, and optionally CBDV, and the entourage-enhancing isoprenoid fraction contains the terpenes beta-myrcene, terpinolene, limonene, ocimene, beta-pinene, beta-caryophyllene, alpha-pinene, and gamma-terpinene. In a preferred embodiment, the entourage-enhancing cannabinoid fraction is beta-myrcene dominant. Additional compounds may be included if desired, e.g., flavonoids, vitamins, active ingredients such as antioxidants or anti-inflammatories, herbal extracts, and essential oils. Sufficient further excipient is then added to produce the required volume. Suspensions may be prepared in volumes of 5 mL, 10mL, 25 mL, 30mL, 50 mL, 100 mL, or such other total volumes as practical for research use or for sale as a pharmaceutical or OTC preparation, or a nutraceutical preparation or dietary supplement. Depending on unit dosage volume and total volume, suspensions may be used to prepare dropper bottles (e.g., 30 mL/1 oz. bottles) or fine mist spray (i.e., oral spray) bottles (e.g., 10 mL bottles). Liquid suspensions of this Example also can be used to prepare softgel capsules, ampoules, or other single unit dosage forms, through methods herein disclosed or known to those of skill in the art. In an alternative “ C– ” embodiment, the formulation is made as above, but without THC. EXAMPLE 9: Formulation of “C+” tincture Liquid suspensions, each containing the below amounts per 1.0 ml dose are made as follows:
Figure imgf000074_0001
Figure imgf000075_0001
The CBD, cannabinoids, isoprenoids, flavonoids, sweetener (e.g., sucrose or sucralose), and xanthan gum are measured out, blended, passed through a No.10 mesh U.S. sieve, and then mixed with a previously made solution of microcrystalline cellulose and sodium carboxymethyl cellulose in excipient (e.g., ethanol or purified water). The sodium benzoate, flavor, and color are diluted with excipient and added with stirring. In an embodiment, the entourage-enhancing cannabinoid fraction contains cannabinoids selected from the group consisting of CBC-type cannabinoids, CBD-type cannabinoids, CBE-type cannabinoids, CBG-type cannabinoids, CBL-type cannabinoids, CBN-type cannabinoids, CBND-type cannabinoids, CBT-type cannabinoids, ∆ 8 -THC-type cannabinoids, ∆ 9 -THC-type cannabinoids, and miscellaneous-type cannabinoids; including but not limited to CBC, CBE, CBG, CBL, CBN, CBND, CBT, CBV, CBGM, CBDV, THCV, CBDP, THCP, and iso-THC; and the entourage-enhancing isoprenoid fraction contains terpenes selected from the group consisting of monoterpenes, diterpenes, triterpenes, sesquiterpenes, miscellaneous terpenes; including but not limited to alpha-bisabolol, beta-caryophyllene, camphene, carene, caryophyllene oxide, alpha-humulene, fenchol, guaiene, guaiol, limonene, linalool, myrcene, nerolidol, ocimene, alpha-phellandrene, alpha-pinene, beta-pinene, alpha-terpinene, gamma-terpinene, terpineol, and terpinolene. In preferred embodiments, the cannabinoid fraction includes CBG, CBN, and optionally CBDV, while the isoprenoid fraction includes limonene and myrcene, wherein limonene comprises 6 mg, and myrcene comprises 4 mg. Additional compounds may be included if desired, e.g., flavonoids, vitamins, active ingredients such as antioxidants or anti-inflammatories, herbal extracts, and essential oils. Sufficient further excipient is then added to produce the required volume. Suspensions may be prepared in volumes of 5 ml, 10ml, 25 mL, 30 mL, 50 mL, 100 mL, or such other total volumes as practical for research use or for sale as a pharmaceutical or OTC preparation, or a nutraceutical preparation or dietary supplement. In an alternative “ C– ” embodiment, the formulation is made as above, but without THC. EXAMPLE 10: Formulation of “C+” capsules Gelatin capsules, each containing the below active ingredients, are made as follows:
Figure imgf000076_0001
The CBD, THC, cannabinoids, isoprenoids, cellulose, starch, and magnesium stearate are blended, passed through a No. 20 mesh U.S. sieve, and filled into hard of soft gelatin capsules in 250 mg quantities. In this Example, the isoprenoid fraction is β-myrcene dominant, with at least 20% w/w of the isoprenoid fraction consisting of β-myrcene. Beta-myrcene is selected because when combined with Δ 9 -THC it will intensify the sedative effects thereof, increase the anxiolytic effects of the formulation, and generally improve the tolerability of THC. In an alternative “ C– ” embodiment, the formulation is made as above, but without THC. EXAMPLE 11: Formulation of “C+” tablets Tablets, each containing the below active ingredients, are made as follows:
Figure imgf000076_0002
The components are blended and compressed to form tablets, each weighing 250 mg. In this Example, Δ 8 -THC is selected for its stability and longer shelf life compared to Δ 9 -THC, which is more easily oxidized to cannabinol (CBN). Delta-8-THC is therapeutically effective to reduce anxiety symptoms and its anxiolytic effects are enhanced by addition of the flavonoid apigenin. In an alternative “ C– ” embodiment, the formulation is made as above, but without Δ 8 -THC. EXAMPLE 12: Formulation of “C+” sublingual or buccal tablets Sublingual or buccal tablets, containing the below active ingredients, are made as follows:
Figure imgf000077_0001
The glycerol, water, sodium citrate, polyvinyl alcohol, and polyvinylpyrrolidone are admixed together by continuous stirring and maintaining the temperature at about 90° C. When the polymers have gone into solution, the solution is cooled to about 50-55° C and the medicament is slowly admixed. The homogenous mixture is poured into forms made of an inert material to produce a drug-containing diffusion matrix having a thickness of about 2-4 mm. This diffusion matrix is then cut to form individual tablets having the appropriate size. Butin is added to promote the reduction of oxidative stress. In an alternative “ C– ” embodiment, the formulation is made as above, but without THC. EXAMPLE 13: Formulation of “C+” capsules with an additional active agent Capsules, each containing a composition with an additional active agent are made as follows:
Figure imgf000077_0002
Figure imgf000078_0003
The active ingredients, cellulose, starch, and magnesium stearate are blended, passed through a No.20 mesh U.S. sieve, and filled into hard of soft gelatin capsules in 300 mg quantities. The antidepressant or anxiolytic would be any pharmaceutical agent known to act as such by one of skill (e.g., SSRIs, serotonin-norepinephrine reuptake inhibitors (SNRIs), tricyclic antidepressants (TCAs), atypical antidepressants, benzodiazepines, buspirone, etc.). In an alternative “ C– ” embodiment, the formulation is made as above, but without THC. EXAMPLE 14: Formulation of “C+” capsules with additional active agents Capsules, each containing a composition with additional active agents are made as follows:
Figure imgf000078_0001
The active ingredients, cellulose, starch, and magnesium stearate are blended, passed through a No.20 mesh U.S. sieve, and filled into hard of soft gelatin capsules in 350 mg quantities. The analgesic or anti-inflammatory would be any agent known to act as such (including both OTC and prescription medications) by one of skill (e.g., aspirin, acetaminophen, ibuprofen, naproxen, prescription NSAIDs, etc.). Cannaflavins are further included to enhance the analgesic or anti-inflammatory effects. In an alternative “ C– ” embodiment, the formulation is made as above, but without THC. EXAMPLE 15: Formulation of “C+” liquid for vaporization A liquid formulation is prepared containing the following ingredients:
Figure imgf000078_0002
Figure imgf000079_0001
The ingredients are mixed and prepared for use with a liquid vaporization appliance. In an alternative “ C– ” embodiment, the formulation is made as above, but without THCA. EXAMPLE 16: Formulation of “C–” topical form A topical formulation may be prepared as follows:
Figure imgf000079_0002
The white soft paraffin is heated until molten. The active ingredients are added and stirring is continued until dispersed. The liquid paraffin and emulsifying wax are incorporated and stirred until dissolved. The mixture is then cooled until solid. In an alternative “ C+ ” embodiment, the formulation is made as above, but with the addition of 6.25 mg of THC. EXAMPLE 17: Formulation of “C+” transdermal delivery form A formulation for a transdermal delivery device may be prepared as follows:
Figure imgf000079_0003
Figure imgf000080_0002
The stabilizer, solubilizer, and permeation enhancing agent are heated and stirred until combined. The active ingredients are added after partially cooled (at a temperature suitable to prevent premature decarboxylation) but before setting and stirring is continued until dispersed. The mixture is then cooled until in its desired final form (e.g., for use in a reservoir delivery system) or admixed with an adhesive and then cooled (e.g., for use in a drug-in-adhesive patch). If used with an electric transdermal delivery device with reservoir, the device may be configured to sufficiently heat the CBDA and THCA so as to decarboxylate them before absorption. If used in a matrix or other passive delivery device, the CBDA and THCA will have therapeutic effects in their acidic form (CBDA for instance has been shown to have anti-inflammatory effects and to reduce nausea and vomiting and enhance 5-HT 1A receptor activation in animal models; THCA also has shown anti-nausea and anti-emetic properties). In an alternative “ C– ” embodiment, the formulation is made as above, but without THCA. EXAMPLE 18: Formulation of “C+” injectable form A formulation for injection (e.g., for subcutaneous, intramuscular, intraperitoneal, or intravenous delivery) may be prepared as follows:
Figure imgf000080_0001
Active ingredients are dissolved in dimethyl sulphoxide (DMSO) in proportions of 1 g to .5 mL. Solution is brought to 37° C and vortexed for 3-5 minutes. Tetraethyleneglycol (TEG) in amount of 5 mL is added, and solution is returned to 37° C and vortexed again for 3-5 mins. Solution is mixed 1:1 with saline containing 1% cremaphor to prevents precipitation. Final solution will be at 10mg/mL active ingredients in 49.5% TEG, 49.5% saline, .5% DMSO, and .5% cremaphor. Injection may be by any suitable means, e.g., bolus injection, IV infusion, or subcutaneous infusion, for example using a drug delivery device comprising a reservoir and a pump mechanism, configured for subcutaneous administration, and which may optionally contain a user interface or be coupled to a device with a user interface such as a smartphone. In an alternative “ C– ” embodiment, the formulation is made as above, but without THC. EXAMPLE 19: Formulation of “C+” intranasal delivery form A nasal spray formulation for intranasal delivery may be prepared as follows:
Figure imgf000081_0001
Solution at 10mg/mL active ingredients in 49.5% MCT, 49.5% saline, .5% DMSO, and .5% cremaphor is prepared, as above (but with MCT in place of TEG), for use in nasal spray device. In other embodiments, a nasal formulation is prepared as a dry powder for inhalation, e.g., by combining the active agents with lactose and mixing for use with a dry powder inhaling appliance, or as in U.S. Pub. No. US2015/0367091A1 and references cited therein. In an alternative “ C– ” embodiment, the formulation is made as above, but without THC. [227] In any of the above cannabis composition examples, any one or more cannabinoids such as CBD and THC, as well as any entourage-enhancing cannabinoid fraction, including combinations thereof, may be replaced with a cannabis extract comprising such cannabinoids. [228] In some other described embodiments, the compositions of the invention will be combined with other compounds such as vitamins, antioxidants, amino acids, probiotics, natural herbs or plant extracts, and other food or dietary supplements, for various purposes such as for energy, reduced fatigue, or improved mental and physical performance (e.g., guarana, amino acids, and vitamins such as B12), for improved performance and reduced gastrointestinal discomfort during exercise (e.g., L-leucine, L-isoleucine, L-valine, Vitamin B1, and Vitamin B5), for osteoporosis or function of bones and teeth (e.g., vitamins C, D3, and K2), for sexual dysfunction, improved sexual vigor, or fertility (e.g., vitamins, amino acids, and plant extracts such as maca root, ginkgo biloba, tributes terrestris, minus pinaster, muir puma, damiana, or catuaba), for improved circulation or to sooth heavy legs (e.g., vitas vinifera extract and bioflavonoids), for relaxation and sleep (e.g., melatonin, passionflower, Californian poppy, and other herbal ingredients), or to relieve muscle and joint pain or trauma, (e.g., hemp seed oil, arnica, and harpagophytum). In one preferred embodiment, a cannabis composition will contain: 1.176 grams of cannabis distillate, 0.09 grams of cherry terps, 0.22 grams of mint, and 28.514 mL of MCT oil. [229] It should be readily appreciated that the above formulation examples are illustrative only. Thus, it should be understood that reference to particular compound(s) is likewise illustrative, and the compound(s) in any Example may be substituted by other compound(s) of the invention. Similarly, any other active compounds (e.g., the antidepressant or anxiolytic in Examples 3 and 11, or analgesic, anti-inflammatory, or antioxidants in Examples 4 and 12) may be substituted by a different active compound, as may be any one or more inactive compounds. Similarly, any of the compounds could be provided in another amount, adapted to achieve the same or like effect when administered to a human. Moreover, for any of the other active compounds of the invention, substitution of the compound by its prodrug, free base, salt, or hydrochloride salt shall be understood to provide merely an alternative embodiment still within the scope of the invention. Further, compositions within the scope of the invention should be understood to be open-ended and may include additional active or inactive compounds and ingredients. The type of formulation employed for the administration of the compounds employed in the methods of the present invention generally may be dictated by the compound(s) employed, the type of pharmacokinetic profile desired from the route of administration and the compound(s), and the state of the patient. It will be readily appreciated that any of the above embodiments and classes of embodiments can be combined to form additional embodiments and formulations. N. Dosage [230] Administration of a mushroom composition or a cannabis composition in a “therapeutically effective amount,” or an “effective amount” to a subject means administration of an amount of the mushroom composition or the cannabis composition sufficient to achieve the desired effect. In embodiments, a “therapeutically effective amount,” or“ effective amount” thus would be the amount sufficient to cause a modulation of a neurotransmitter system. When an “effective amount” means an amount effective in treating the stated disorder or symptoms in a subject, “therapeutic effect” would be understood to mean the responses(s) in a mammal after treatment that are judged to be desirable and beneficial. Hence, depending on the mental health condition to be treated, or improvement in psychological functioning sought, and depending on the particular constituent(s) in the compositions of the invention under consideration, those responses shall differ, but would be readily understood by those of ordinary skill. a. Mushroom Compositions—Dosage [231] Mushroom compositions disclosed herein may comprise therapeutic amounts of any of psilocybin and other compounds from Psilocybe or other psilocybin-containing mushrooms, such as other psychedelic or psychoactive tryptamines (“minor tryptamines”), L-theanine, niacin, and compounds from lion’s mane mushrooms and cordyceps mushrooms, and optionally valerian (or other plant extracts or compounds) and/or vitamin B12 (or other vitamins), optionally a CB 1 receptor modulator (such as CBD or PEA), and in some embodiments, other active ingredients which may include minerals, amino acids, proteins, and dietary or nutritional supplements. [232] In embodiments, a mushroom composition, where it contains psilocybin and other compounds from Psilocybe or other psilocybin-containing mushrooms, and/or compounds from lion’s mane mushrooms and cordyceps mushrooms, will comprise ground or pulverized fungal matter (i.e., fungal matter from Psilocybe or other psilocybin-containing mushrooms, lion’s mane mushrooms, and/or cordyceps mushrooms), one or more fungal extracts from any of the foregoing, and/or one or more isolated or purified fungal compounds therefrom. [233] In embodiments, a mushroom composition comprises ground or pulverized fungal matter, which may be fungal matter from one or more fungal species, and may further comprise one or more carriers, diluents, or excipients. [234] In embodiments, where a mushroom composition comprises ground or pulverized fungal matter, including where a composition preferably comprises ground or pulverized dried fungal matter, such as fungal matter from a Psilocybe or other psilocybin-producing species, from lion’s mane, or cordyceps, it may be present in an amount so that a single dose is (whether or not such dose is present in a unit dosage form), e.g., 10 mg or less, at least 20 mg, at least 30 mg, at least 40 mg, at least 50 mg, at least 100 mg, at least 125 mg, at least 150 mg, at least 175 mg, at least 200 mg, at least 225 mg, at least 250 mg, at least 275 mg, at least 300 mg, at least 350 mg, at least 400 mg, at least 450 mg, at least 500 mg, at least 600 mg, at least 700 mg, at least 800 mg, at least 900 mg, or at least 1 gram. [235] In embodiments, where a mushroom composition comprises an extract from a psilocybin-containing mushroom, truffle, mycelial body, or other fungal material, such as from a Psilocybe or other psilocybin-producing species, it may be present in an amount so that a single dose is (whether or not such dose is present in a unit dosage form), e.g., 10 mg or less, at least 20 mg, at least 30 mg, at least 40 mg, at least 50 mg, at least 100 mg, at least 125 mg, at least 150 mg, at least 175 mg, at least 200 mg, at least 225 mg, at least 250 mg, at least 275 mg, at least 300 mg, at least 350 mg, at least 400 mg, at least 450 mg, or at least 500 mg. [236] In embodiments, where a mushroom composition comprises purified, synthetic, or biosynthetic psilocybin, it may be present in an amount so that a single dose is (whether or not such dose is present in a unit dosage form), e.g., .01 mg or less, at least .05 mg, at least .1 mg, at least .15 mg, at least .2 mg, at least .3 mg, at least .4 mg, at least .5 mg, at least 1 mg, at least 1.5 mg, at least 2 mg, at least 2.5 mg, at least 3 mg, at least 3.5 mg, at least 4 mg, at least 5 mg, at least 6 mg, at least 7 mg, at least 8 mg, at least 9 mg, or at least 10 mg. [237] In one aspect of the invention, the compositions and formulations will be without psychedelic effect when taken by a human. In accordance with this aspect of the invention, dosages shall be below the threshold at which human subjects report the presence of such psychedelic effects (i.e., “microdoses”). Because such dosages may differ from subject to subject, an individual subject will be able to take a smaller dosage, if desired, and unit dosage forms at various dosage levels can be offered to provide optimal results across the whole of the dose-response curve, for differently responding individuals in a population. [238] In general, dose ranges under about 2.0-3.0 mg isolated or pure psilocybin are considered sub-psychedelic dosages, and this amount can be converted based on the psilocybin concentration in a whole fungal preparation to determine reasonable and appropriate starting dosages, which then can be adjusted upwards or downwards based on subjective effects, using known methods. Therapeutic benefits may be found optimal in a range of about 0.5 to about 2.0 mg psilocybin, and some preferred embodiments contain purified, synthetic, or biosynthetic psilocybin in such amounts, or in approximately equivalent amounts. For example, a P. cubensis strain with an approximate w/w% of psilocybin of 0.63% ( see Table 1 above) may be used in a composition of the invention as an equivalent extract by weight of between about 75 to about 325 mg. Amounts of between 100 to 275 mg may be preferred. Likewise, a 10x purified extract from the same strain may be used in an amount of between about 7.5 to about 32.5 mg. It will be appreciated that similar calculations may be made for other extracts, such as cordyceps, lion’s mane, and valerian root, based on the same principle, when using purified extracts or isolated compounds. [239] In embodiments, where a mushroom composition comprises lion’s mane extract, it may be present in an amount so that a single dose is (whether or not such dose is present in a unit dosage form), e.g., 10 mg or less, at least 20 mg, at least 30 mg, at least 40 mg, at least 50 mg, at least 100 mg, at least 150 mg, at least 200 mg, at least 250 mg, at least 300 mg, at least 350 mg, at least 400 mg, at least 450 mg, at least 500 mg, at least 550 mg, at least 600 mg, at least 650 mg, at least 700 mg, at least 750 mg, at least 800 mg, at least 850 mg, at least 900 mg, at least 950 mg, or at least 1000 mg. [240] In embodiments, where a mushroom composition comprises a purified, synthetic, or biosynthetic compound from lion’s mane, such as a hericenone extracted from or extractable from Hericium erinaceum , and including any of Hericenone A, Hericenone B, Hericenone C, Hericenone D, Hericenone E, Hericenone F, Hericenone G, Hericenone H, Hericenone I, Hericenone J, Hericenone K, 3HF, DLPE, Isohericerinol A, Hericerin, NDPIH, and Corallocin A, it may be present in an amount so that a single dose is (whether or not such dose is present in a unit dosage form), e.g., .01 mg or less, at least .05 mg, at least .1 mg, at least .15 mg, at least .2 mg, at least .3 mg, at least .4 mg, at least .5 mg, at least 1 mg, at least 1.5 mg, at least 2 mg, at least 2.5 mg, at least 3 mg, at least 3.5 mg, at least 4 mg, at least 5 mg, at least 6 mg, at least 7 mg, at least 8 mg, at least 9 mg, or at least 10 mg. [241] In embodiments, where a mushroom composition comprises cordyceps extract, it may be present in an amount so that a single dose is (whether or not such dose is present in a unit dosage form), e.g., 10 mg or less, at least 20 mg, at least 30 mg, at least 40 mg, at least 50 mg, at least 100 mg, at least 150 mg, at least 200 mg, at least 250 mg, at least 300 mg, at least 350 mg, at least 400 mg, at least 450 mg, at least 500 mg, at least 550 mg, at least 600 mg, at least 650 mg, at least 700 mg, at least 750 mg, at least 800 mg, at least 850 mg, at least 900 mg, at least 950 mg, or at least 1000 mg. [242] In embodiments, where a mushroom composition comprises a purified, synthetic, or biosynthetic compound from cordyceps, such as cordycepin, cordycepic acid, N-acetylgalactosamine, adenosine, ergosterol, ergosteryl esters, bioxanthracenes, hypoxanthine, acid deoxyribonuclease, polysaccharide, exopolysaccharide, chitinase, macrolides, cicadapeptins, myriocin, superoxide dismutase, protease, naphthoquinone, cordyheptapeptide, dipicolinic acid, fibrinolytical enzyme, lectin, and cordymin, it may be present in an amount so that a single dose is (whether or not such dose is present in a unit dosage form), e.g., .01 mg or less, at least .05 mg, at least .1 mg, at least .15 mg, at least .2 mg, at least .3 mg, at least .4 mg, at least .5 mg, at least 1 mg, at least 1.5 mg, at least 2 mg, at least 2.5 mg, at least 3 mg, at least 3.5 mg, at least 4 mg, at least 5 mg, at least 6 mg, at least 7 mg, at least 8 mg, at least 9 mg, or at least 10 mg. [243] In embodiments, where a mushroom composition comprises L-theanine, it may be present in an amount so that a single dose is (whether or not such dose is present in a unit dosage form), e.g., 10 mg or less, at least 20 mg, at least 30 mg, at least 40 mg, at least 50 mg, at least 100 mg, at least 150 mg, at least 200 mg, at least 250 mg, at least 300 mg, at least 350 mg, at least 400 mg, at least 450 mg, at least 500 mg, at least 550 mg, at least 600 mg, at least 650 mg, at least 700 mg, at least 750 mg, at least 800 mg, at least 850 mg, at least 900 mg, at least 950 mg, or at least 1000 mg. In embodiments, a mushroom composition comprises another amino acid or amino acid analog, at dose amounts known in the art. [244] In embodiments, where a mushroom composition comprises niacin, it may be present in an amount so that a single dose is (whether or not such dose is present in a unit dosage form), e.g., 10 mg or less, at least 20 mg, at least 30 mg, at least 40 mg, at least 50 mg, at least 100 mg, at least 150 mg, at least 200 mg, at least 250 mg, at least 300 mg, at least 350 mg, at least 400 mg, at least 450 mg, at least 500 mg, at least 550 mg, at least 600 mg, at least 650 mg, at least 700 mg, at least 750 mg, at least 800 mg, at least 850 mg, at least 900 mg, at least 950 mg, or at least 1000 mg. In embodiments, a mushroom composition comprises another vitamin, at dose amounts known in the art. [245] In embodiments, where a mushroom composition comprises valerian root extract, it may be present in an amount so that a single dose is (whether or not such dose is present in a unit dosage form), e.g., 10 mg or less, at least 20 mg, at least 30 mg, at least 40 mg, at least 50 mg, at least 100 mg, at least 150 mg, at least 200 mg, at least 250 mg, at least 300 mg, at least 350 mg, at least 400 mg, at least 450 mg, at least 500 mg, at least 550 mg, at least 600 mg, at least 650 mg, at least 700 mg, at least 750 mg, at least 800 mg, at least 850 mg, at least 900 mg, at least 950 mg, or at least 1000 mg. In embodiments, a mushroom composition comprises another plant extract or compound, at dose amounts known in the art. [246] In embodiments, where a mushroom composition comprises Vitamin B12, it may be present in an amount so that a single dose is (whether or not such dose is present in a unit dosage form), e.g., 1 mcg or less, at least 2 mcg, at least 3 mcg, at least 4 mcg, at least 5 mcg, at least 10 mcg, at least 15 mcg, at least 20 mcg, at least 25 mcg, at least 30 mcg, at least 35 mcg, at least 40 mcg, at least 45 mcg, at least 50 mcg, at least 55 mcg, at least 60 mcg, at least 65 mcg, at least 70 mcg, at least 75 mcg, at least 80 mcg, at least 85 mcg, at least 90 mcg, at least 95 mcg, at least 100 mcg, at least 250 mcg, at least 500 mcg, at least 750 mcg, at least 1000 mcg, at least 2500 mcg, or at least 5000 mcg. In embodiments, a mushroom composition comprises another vitamin, at dose amounts known in the art. [247] In embodiments, where a mushroom composition comprises a CB 1 receptor modulator such as CBD or PEA, it may be present in an amount so that a single dose is (whether or not such dose is present in a unit dosage form), e.g., 1 mg or less, at least 2 mg, at least 3 mg, at least 4 mg, at least 5 mg, at least 10 mg, at least 15 mg, at least 20 mg, at least 25 mg, at least 30 mg, at least 35 mg, at least 40 mg, at least 45 mg, at least 50 mg, at least 55 mg, at least 60 mg, at least 65 mg, at least 70 mg, at least 75 mg, at least 80 mg, at least 85 mg, at least 90 mg, at least 95 mg, or at least 100 mg. In embodiments, a mushroom composition comprises another CB 1 receptor modulator, at dose amounts known in the art. b. Cannabis Compositions—Dosage [248] Cannabis compositions disclosed herein may comprise therapeutic amounts of any of THC, CBD, and other psychoactive or non-psychoactive cannabinoids (besides THC and CBD, referred to “minor cannabinoids”), isoprenoids or terpenes, and in some embodiments, flavonoids and/or other active ingredients, such as vitamins, minerals, natural and unnatural (analog) amino acids (e.g., L-theanine), proteins, and dietary or nutritional supplements. [249] In embodiments, a cannabis composition comprises a cannabis extract, and may further comprise one or more carriers, diluents, or excipients. A cannabis extract may be, for example, a cannabis full spectrum extract, a cannabis broad spectrum extract, a cannabis oil, a cannabis distillate, or a cannabis isolate, as such terms will be readily understood by those of skill. In other embodiments, a cannabis extract may be any of kief, hashish, bubble hash, solvent reduced oils, sludges, e-juice, and tinctures. [250] In embodiments, where a cannabis composition comprises CBD, it may be present in an amount so that a single dose is (whether or not such dose is present in a unit dosage form), e.g., 5 mg or less, at least 5 mg, at least 10 mg, at least 15 mg, at least 20 mg, at least 25 mg, at least 30 mg, at least 35 mg, at least 40 mg, at least 45 mg, at least 50 mg, at least 55 mg, at least 60 mg, at least 65 mg, at least 70 mg, at least 75 mg, at least 80 mg, at least 85 mg, at least 90 mg, at least 95 mg, or at least 100 mg. [251] Thus, for example, where the CBD is dissolved or suspended in a liquid formulation having a single dosage unit of 1 mL, that CBD may be present in an amount of 5 mg/mL or less, at least 5 mg/mL, at least 10 mg/mL, at least 15 mg/mL, at least 20 mg/mL, at least 25 mg/mL, at least 30 mg/mL, at least 35 mg/mL, at least 40 mg/mL, at least 45 mg/mL, at least 50 mg/mL, at least 55 mg/mL, at least 60 mg/mL, at least 65 mg/mL, at least 70 mg/mL, at least 75 mg/mL, at least 80 mg/mL, at least 85 mg/mL, at least 90 mg/mL, at least 95 mg/mL, or at least 100 mg/mL. However, it should be readily appreciated that both higher doses and higher concentrations for active ingredients are within the scope of the present invention. [252] In embodiments, where a cannabis composition comprises THC, it may be present in an amount so that a single dose is (whether or not present in a unit dosage form), e.g., 1 mg or less, at least 2 mg, at least 3 mg, at least 4 mg, at least 5 mg, at least 10 mg, at least 15 mg, at least 20 mg, at least 25 mg, at least 30 mg, at least 35 mg, at least 40 mg, at least 45 mg, or at least 50 mg. Thus, for example, where the THC is dissolved or suspended in a liquid formulation having a single dosage unit of 1 mL, that THC may be present in an amount of 1 mg/mL or less, at least 2 mg/mL, at least 3 mg/mL, at least 4 mg/mL, at least 5 mg/mL, at least 10 mg/mL, at least 15 mg/mL, at least 20 mg/mL, at least 25 mg/mL, at least 30 mg/mL, at least 35 mg/mL, at least 40 mg/mL, at least 45 mg/mL, or at least 50 mg/mL. [253] Although it will be understood that doses of THC and CBD are selected independently depending on the desired effects of a composition, in some embodiments with THC, the ratio of THC:CBD is between 1:20 to 1:10, between 1:10 and 1:5, between 1:5 and 1:2, between 1:2 and about 1:1, but preferably about 1:4; in other embodiments it is between about 1:1 and 2:1, between 2:1 and 5:1, between 5:1 and 10:1, and between 10:1 and 20:1. [254] Generally, in preferred embodiments the entourage-enhancing cannabinoid fraction is contemplated as consisting of a lower amount (by % w/w) than the amounts of CBD and THC (or their synthetic counterparts, analogs, metabolites, prodrugs, salts, and the like). In these embodiments, the entourage-enhancing cannabinoid fraction comprises from about 0.1% to about 25% of the total amount of cannabinoids and isoprenoids (and flavonoids, when present) by weight, and preferably from about 1-15% w/w, about 2.5-12.5% w/w, or about 5-10% w/w. In preferred embodiments, the individual cannabinoids of the entourage-enhancing cannabinoid fraction shall be in relatively equal proportions, but it is contemplated that such cannabinoids may be in unequal proportions, and particularly contemplated that certain cannabinoids may be dominant with others only present in trace amounts, especially where a cannabinoid fraction contains three or more cannabinoids. [255] Terpenes are generally considered to be pharmacologically relevant in whole plant material when present in concentrations of at least 0.05% (Hazekamp and Fischedick 2010. “Metabolic fingerprinting of Cannabis sativa L., cannabinoids and terpenoids for chemotaxonomic and drug standardization purposes” Phytochemistry 2058-73; Russo 2011). For purposes of the compositions and formulations disclosed herein, isoprenoid fractions are preferably present in an amount of about 1% w/w of the total amount of cannabinoids and isoprenoids (and flavonoids, when present). However, in other embodiments, the isoprenoid may be < 0.1% w/w, 0.1-0.5 % w/w, 0.5-1 % w/w, 1-1.5% w/w, 1.5-2.5% w/w, 2.5-5% w/w, 5-10% w/w, or greater than 10% w/w. [256] In embodiments, the individual isoprenoids of the entourage-enhancing isoprenoid fraction shall be in relatively equal proportions, but it is contemplated that such isoprenoids may be in unequal proportions, and particularly contemplated that certain isoprenoids may be dominant with others only present in trace amounts, especially where an isoprenoid fraction contains three or more isoprenoids. For example, in certain preferred “myrcene-dominant” embodiments, the entourage-enhancing isoprenoid fraction contains from about 19.9-29.9% myrcene by weight, and about 4.2-6.3% beta-caryophyllene, 2.8-4.2% and gamma-terpinene, 12.2-18.3% limonene, 13.0-19.5% terpinolene, 8.6-12.9% ocimene, 4.5-6.8% beta-pinene, 2.7-4.0% alpha-pinene, and about 12.0-18.0% other terpenes, where no such other terpenes are in an amount of 2% w/w or greater. In other myrcene-dominant embodiments, the composition comprises an amount of myrcene by w/w of at least 1.5x, at least 2x, at least 3x, at least 4x, at least 5x, or more than 5x the amount of the terpene that is in the next greatest amount. In general, for any single terpene-dominant embodiment, regardless of the terpene that is dominant, the dominant terpene may be by w/w at least 1.5x, at least 2x, at least 3x, at least 4x, at least 5x, or more than 5x the amount of the terpene that is in the next greatest amount. Such embodiments may be referred to as “1.5x dominant” of the terpene, or “2x dominant,” “3x dominant,” “4x dominant,” and the like. c. General Dosing Guidelines [257] It should be readily appreciated that dosages may vary depending upon whether the treatment is therapeutic or prophylactic, the onset, progression, severity, frequency, duration, probability of, or susceptibility of the symptom to which treatment is directed, clinical endpoint desired, previous, simultaneous or subsequent treatments, general health, age, gender, and race of the subject, bioavailability, potential adverse systemic, regional or local side effects, the presence of other disorders or diseases in the subject, and other factors that will be appreciated by the skilled artisan (e.g., medical or familial history). Dose amount, frequency or duration may be increased or reduced, as indicated by the clinical outcome desired, status of the infection, reactivation, pathology or symptom, or any adverse side effects of the treatment or therapy. The skilled artisan will appreciate the factors that may influence the dosage, frequency, and timing required to provide an amount sufficient or effective for providing a prophylactic or therapeutic effect or benefit. [258] Indeed, it will be understood that, in some embodiments, the dose actually administered will be determined by a physician, in light of the relevant circumstances, including the disorder to be treated, the chosen route of administration, the actual composition administered, the age, weight, and response of the individual patient, and the severity of the patient’s symptoms, and therefore any dosage ranges disclosed herein are not intended to limit the scope of the invention. In some instances, dosage levels below the lower limit of a disclosed range may be more than adequate, while in other cases doses above a range may be employed without causing any harmful side effects, provided for instance that such larger doses also may be divided into several smaller doses for administration. [259] In these embodiments, the compositions of the invention may be administered and dosed in accordance with good medical practice, taking into account the method and scheduling of administration, prior and concomitant medications and medical supplements, the clinical condition of the individual patient and the severity of the underlying disease, the patient’s age, sex, body weight, and other such factors relevant to medical practitioners, and knowledge of the particular compound(s) used. Starting and maintenance dosage levels thus may differ from patient to patient, for individual patients across time, and for different pharmaceutical compositions, but shall be able to be determined with ordinary skill. [260] It should be appreciated that in other embodiments, e.g., when the compositions of the invention are for “nutraceutical” use or purpose, and sold over-the-counter or otherwise without a prescription (e.g., as dietary supplements or as natural products), and therefore taken without the intervention or guidance of a medical professional, appropriate dosages to achieve a therapeutic effect, including the upper and lower bounds of any dose ranges, can be determined by an individual by reference to available public information and knowledge, and by reference to subjective considerations regarding desired outcomes and effects (including, in some embodiments, a desired absence of psychedelic effects). [261] In embodiments, a patient will be prescribed, administered, or will obtain without a prescription, a therapeutically effective dose of the compositions of the invention on a regular or “chronic” basis, wherein the patient is administered the compositions of the invention daily, several times per day (at least one, at least two, at least three, at least four, or greater than four times per day); on a set, repeating schedule, wherein the patient is administered a therapeutically effective dose of the compositions of the invention every other day, every three days, every four days, every five days, every six days, every seven days, or more than every seven days; or, in some embodiments, a varying schedule comprised of a plurality of days “on” (wherein the therapeutically effective dose of the pharmaceutical composition of the invention is administered), and a plurality of days “off” (wherein no administration occurs), such as one day on two days off, two days on three days off, three days on four days off, or other such schedules as would be apparent to those of skill. [262] In embodiments, the dosage administered to, or taken by a patient in need thereof, is a “macrodose” (whether or not so named), wherein the dose is a sufficient amount to cause at least one perceptible subjective effect within a patient, and preferably the full range of subjective effects desired through administration of the drug, or a desired subset thereof, as chosen through ordinary skill. In embodiments, such perceptible subjective effects are those characteristic of, or substantially characteristic of, or sharing at least one or more subjective qualities of, a psychedelic. [263] In embodiments, the dosage administered to, or taken by a patient in need thereof, is a “microdose” (whether or not so named), wherein the dose is not perceptible by the patient to which it is administered. In embodiments, a microdose may be 0.001 to 0.25 of a macro-dose, such as but not limited to 0.003, 0.005, 0.007, 0.009, 0.01, 0.03, 0.05, 0.07, 0.09, 0.1, 0.2, and 0.25 of a macrodose (wherein the list is inclusive, and modified by the term “about”). In embodiments, a microdose is between about 0.001 mg and about 50 mg, including 0.001 mg, 0.005 mg, 0.01 mg, 0.05 mg, 0.1 mg, 0.5 mg, 1 mg, 1.25 mg, 1.5 mg, 1.75 mg, 2 mg, 2.25 mg, 2.5 mg, 2.75 mg, 3 mg, 3.5 mg, 4 mg, 4.5 mg, 5 mg, 5.5 mg, 6 mg, 6.5 mg, 7 mg, 7.5 mg, 8 mg, 8.5 mg, 9 mg, 9.5 mg, 10 mg, 11 mg, 12 mg, 13 mg, 14 mg, 15 mg, and values in between, wherein each value is modified by the term “about.” [264] Determination of appropriate dosing shall include not only the determination of single dosage amounts, but also determination of the number and timing of doses, e.g., administration of a particular dosage amount once per day, twice per day, or more than twice per day, and the time(s) of day preferable for administration, and any daily dosing regimen, including different and distinct alternating daily dosing regimens, as are described herein. In preferred embodiments, dosing regimens will be “daily dosing” regimens, but modification of any dosing regimen, including any daily dosing regimen herein, may be made within ordinary skill, for example that every “daily dose” be taken at 12-hour intervals (e.g., at “morning” and at “night”), rather than at 24-hour intervals, and thus, in one exemplary such modification, that 14-day therapeutic regimes will be administered during a single week. Other such modifications and alterations will be readily appreciated by those of skill. [265] In embodiments, especially where a formulation is prepared in single unit dosage form, suggested dosage amounts may be known by reference to the format of the preparation itself. In other embodiments, where a formulation is prepared in multiple dosage form, for instance liquid suspensions of oils and tinctures and topical preparations, suggested dosage amounts may be known by reference to the means of administration (e.g., measured dropper bottles, “push button” spray bottles), or by reference to the packaging and labeling, package insert(s), marketing materials, or information and knowledge available to the public. O. Dosing Regimens [266] In embodiments, the method of treating a mental health condition or improving mental health comprises administering a composition according to a distinct and predetermined dosing regimen, provided for its novel and unique therapeutic effects. Such “defined dosing regimens” are among the advantages and improvements over the prior art. In embodiments, a composition will be taken on a daily basis, and a defined dosing regimen may also be referred to as a “daily dosing regimen”; conversely, as noted above, daily dosing regimens may be modified, and can also be readily embodied as “defined dosing regimens.” [267] In embodiments, the defined dosing regimen comprises alternating phases of administration (“dosing,” which depending on embodiments will include what is commonly referred to equivalently, for example with psilocybin-comprising products, as “microdosing”) and abstaining (“washout”). [268] In embodiments, the defined dosing regimen comprises alternating phases of dosing a mushroom composition and washout. [269] In embodiments, the defined dosing regimen comprises alternating phases of dosing an M+ mushroom composition and washout. [270] In embodiments, the defined dosing regimen comprises alternating phases of dosing an M– mushroom composition and washout. [271] In embodiments, the defined dosing regimen comprises alternating phases of dosing a cannabis composition and washout. [272] In embodiments, the defined dosing regimen comprises alternating phases of dosing an C+ mushroom composition and washout. [273] In embodiments, the defined dosing regimen comprises alternating phases of dosing an C– mushroom composition and washout. [274] In embodiments, the defined dosing regimen comprises alternating phases of dosing a first composition, and dosing a second composition. [275] In embodiments, the defined dosing regimen comprises alternating phases of dosing a first mushroom composition and dosing a second mushroom composition. [276] In embodiments, the defined dosing regimen comprises alternating phases of dosing a first M+ mushroom composition and dosing a second M+ mushroom composition. [277] In embodiments, the defined dosing regimen comprises alternating phases of dosing a first M+ mushroom composition and dosing a second M– mushroom composition. [278] In embodiments, the defined dosing regimen comprises alternating phases of dosing a first M– mushroom composition and dosing a second M– mushroom composition. [279] In embodiments, the defined dosing regimen comprises alternating phases of dosing a mushroom composition and dosing a cannabis composition. [280] In embodiments, the defined dosing regimen comprises alternating phases of dosing an M+ mushroom composition and dosing a cannabis composition. [281] In embodiments, the defined dosing regimen comprises alternating phases of dosing an M+ mushroom composition and dosing a C+ cannabis composition. [282] In embodiments, the defined dosing regimen comprises alternating phases of dosing an M+ mushroom composition and dosing a C– cannabis composition. [283] In embodiments, the defined dosing regimen comprises alternating phases of dosing an M– mushroom composition and dosing a cannabis composition. [284] In embodiments, the defined dosing regimen comprises alternating phases of dosing an M– mushroom composition and dosing a C+ cannabis composition. [285] In embodiments, the defined dosing regimen comprises alternating phases of dosing an M– mushroom composition and dosing a C– cannabis composition. [286] In embodiments, the defined dosing regimen comprises alternating phases of dosing a first cannabis composition and dosing a second cannabis composition. [287] In embodiments, the defined dosing regimen comprises alternating phases of dosing a first C+ cannabis composition and dosing a second C+ cannabis composition. [288] In embodiments, the defined dosing regimen comprises alternating phases of dosing a first C+ cannabis composition and dosing a second C– cannabis composition. [289] In embodiments, the defined dosing regimen comprises alternating phases of dosing a first C– cannabis composition and dosing a second C– cannabis composition. [290] In embodiments, the defined dosing regimen comprises three or more alternating phases of dosing and/or washout, including three alternating phases, four alternating phases, five alternating phases, six alternating phases, seven alternating phases, eight alternating phases, nine alternating phases, ten alternating phases, and more than ten alternating phases, including 11 or more, 12 or more, or greater than 12 alternating phases. [291] Any or all of such alternating phases may be dosing phases or washout phases, including three or more phase of dosing, including three dosing phases, four dosing phases, five dosing phases, six dosing phases, seven dosing phases, eight dosing phases, nine dosing phases, ten dosing phases, and more than ten dosing phases, including 11 or more, 12 or more, or greater than 12 dosing phases. Such dosing regimens may, in some embodiments, include one or more washout phases, including three washout phases, four washout phases, five washout phases, six washout phases, or greater than six washout phases. [292] In embodiments of the one or more of the defined dosing regimens above, the alternating phases of dosing are on consecutive periods, which may or may not be days, but may be periods shorter than days (including fractions of days) or may be longer than days. [293] In embodiments, any one or more of the defined dosing regimens above is a “daily dosing regimen,” wherein the alternating phases of dosing are on consecutive days. [294] In embodiments, any one or more of the defined dosing regimens above is a “twice-a-day dosing regimen,” wherein the alternating phases of dosing are at morning and at night, or at two other times per day, including predetermined times chosen by a patient or a practitioner, or chosen based on a software analysis of one or more objective indicia. [295] In embodiments, any one or more of the defined dosing regimens above is a “three-times-per-day dosing regimen,” wherein the alternating phases of dosing are at breakfast, lunch, and dinner, or at three other times per day, including predetermined times chosen by a patient or a practitioner, or chosen based on a software analysis of one or more objective indicia. Defined dosing regimens also may be four, five, or more than five times per day. Defined dosing regimens may also be on a greater than single day schedule, such as every other day, every third day, every fourth day, every fifth day, every sixth day, every week, or every number of days greater than one week. Defined dosing regimens also may be on schedules not defined by an integer number of days, such as every 1.5 days, and the use of an electronic tool, mobile app, or other software app, may optionally assist with compliance. [296] In embodiments with a defined dosing regimen, the number of administrations of each phase will be independently selected, any may include from one administration to seven administrations inclusive, such as one administration, two administrations, three administrations, four administrations, five administrations, six administrations, and one week, as well as a number of administrations greater than seven administrations, such as eight administrations, nniinnee administrations, 10 administrations, 11 administrations, 12 administrations, and greater than 12 administrations. Here, as elsewhere the term “administration” will be understood to include an “administration” period or day of abstinence or “washout,” wherein such period or day of abstinence or washout may also include the administration of a composition that is not a mushroom or cannabis composition of the invention, but which may for example comprise additional active agents, such as vitamins, nootropics, or supplements
[297] In embodiments, the defined dosing regimen comprises alternating phases of between 2-7 periods of administration and 2-7 periods of abstaining (washout). In embodiments, the defined dosing regimen comprises alternating phases of between 2-6 periods of administration and 2-6 periods of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 2-5 periods of administration and 2-5 periods of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 2-4 periods of administration and 2-4 periods of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 2-3 periods of administration and 2-3 periods of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 3-4 periods of administration and 3-4 periods of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 3-5 periods of administration and 3-5 periods of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 3-6 periods of administration and 3-6 periods of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 3-7 periods of administration and 3-7 periods of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 4-7 periods of administration and 4-7 periods of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 4-6 periods of administration and 4-6 periods of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 4-5 periods of administration and 4-5 periods of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 6-7 periods of administration and 6-7 periods of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 5-7 periods of administration and 5-7 periods of abstaining. In embodiments, the defined dosing regimen comprises alterating phases of between 1-7 periods of administration and 1-7 periods of abstaining.
[298] In embodiments, the defined dosing regimen comprises alternating phases of between 2-7 periods of a first composition and 2-7 periods of a second composition. In embodiments, the defined dosing regimen comprises alternating phases of between 2-6 periods of a first composition and 2-6 periods of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 2-5 periods of a first composition and 2-5 periods of a second composition. In embodiments, the defined dosing regimen comprises alternating phases of between 2-4 periods of a first composition and 2-4 periods of a second composition. In embodiments, the defined dosing regimen comprises alternating phases of between 2-3 periods of a first composition and 2-3 periods of a second composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-4 periods of a first composition and 3-4 periods of a second composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-5 periods of a first composition and 3-5 periods of a second composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-6 periods of a first composition and 3-6 periods of a second composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-7 periods of a first composition and 3-7 periods of a second composition. In embodiments, the defined dosing regimen comprises alternating phases of between 4-7 periods of a first composition and 4-7 periods of a second composition. In embodiments, the defined dosing regimen comprises alternating phases of between 4-6 periods of a first composition and 4-6 periods of a second composition. In embodiments, the defined dosing regimen comprises alternating phases of between 4-5 periods of a first composition and 4-5 periods of a second composition. In embodiments, the defined dosing regimen comprises alternating phases of between 6-7 periods of a first composition and 6-7 periods of a second composition. In embodiments, the defined dosing regimen comprises alternating phases of between 5-7 periods of a first composition and 5-7 periods of a second composition. In embodiments, the defined dosing regimen comprises alternating phases of between 1-7 periods of a first composition and 1-7 periods of a second composition.
[299] In embodiments, the defined dosing regimen comprises alternating phases of between 2-7 periods of a mushroom composition and 2-7 periods of a cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 2-6 periods of a mushroom composition and 2-6 periods of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 2-5 periods of a mushroom composition and 2-5 periods of a cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 2-4 periods of a mushroom composition and 2-4 periods of a cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 2-3 periods of a mushroom composition and 2-3 periods of a cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-4 periods of a mushroom composition and 3-4 periods of a cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-5 periods of a mushroom composition and 3-5 periods of a cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-6 periods of a mushroom composition and 3-6 periods of a cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-7 periods of a mushroom composition and 3-7 periods of a cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 4-7 periods of a mushroom composition and 4-7 periods of a cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 4-6 periods of a mushroom composition and 4-6 periods of a cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 4-5 periods of a mushroom composition and 4-5 periods of a cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 6-7 periods of a mushroom composition and 6-7 periods of a cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 5-7 periods of a mushroom composition and 5-7 periods of a cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 1-7 periods of a mushroom composition and 1-7 periods of a cannabis composition.
[300] In embodiments, the defined dosing regimen comprises alternating phases of between 2-7 periods of a first mushroom composition and 2-7 periods of a second mushroom composition. In embodiments, the defined dosing regimen comprises alternating phases of between 2-6 periods of a first mushroom composition and 2-6 periods of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 2-5 periods of a first mushroom composition and 2-5 periods of a second mushroom composition. In embodiments, the defined dosing regimen comprises alternating phases of between 2-4 periods of a first mushroom composition and 2-4 periods of a second mushroom composition. In embodiments, the defined dosing regimen comprises alternating phases of between 2-3 periods of a first mushroom composition and 2-3 periods of a second mushroom composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-4 periods of a first mushroom composition and 3-4 periods of a second mushroom composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-5 periods of a first mushroom composition and 3-5 periods of a second mushroom composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-6 periods of a first mushroom composition and 3-6 periods of a second mushroom composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-7 periods of a first mushroom composition and 3-7 periods of a second mushroom composition. In embodiments, the defined dosing regimen comprises alternating phases of between 4-7 periods of a first mushroom composition and 4-7 periods of a second mushroom composition. In embodiments, the defined dosing regimen comprises alternating phases of between 4-6 periods of a first mushroom composition and 4-6 periods of a second mushroom composition. In embodiments, the defined dosing regimen comprises alternating phases of between 4-5 periods of a first mushroom composition and 4-5 periods of a second mushroom composition. In embodiments, the defined dosing regimen comprises alternating phases of between 6-7 periods of a first mushroom composition and 6-7 periods of a second mushroom composition. In embodiments, the defined dosing regimen comprises alternating phases of between 5-7 periods of a first mushroom composition and 5-7 periods of a second mushroom composition. In embodiments, the defined dosing regimen comprises alternating phases of between 1-7 periods of a first mushroom composition and 1-7 periods of a second mushroom composition. [301] In embodiments, the defined dosing regimen comprises alternating phases of between 2-7 periods of a first cannabis composition and 2-7 periods of a second cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 2-6 periods of a first cannabis composition and 2-6 periods of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 2-5 periods of a first cannabis composition and 2-5 periods of a second cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 2-4 periods of a first cannabis composition and 2-4 periods of a second cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 2-3 periods of a first cannabis composition and 2-3 periods of a second cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-4 periods of a first cannabis composition and 3-4 periods of a second cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-5 periods of a first cannabis composition and 3-5 periods of a second cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-6 periods of a first cannabis composition and 3-6 periods of a second cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-7 periods of a first cannabis composition and 3-7 periods of a second cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 4-7 periods of a first cannabis composition and 4-7 periods of a second cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 4-6 periods of a first cannabis composition and 4-6 periods of a second cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 4-5 periods of a first cannabis composition and 4-5 periods of a second cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 6-7 periods of a first cannabis composition and 6-7 periods of a second cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 5-7 periods of a first cannabis composition and 5-7 periods of a second cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 1-7 periods of a first cannabis composition and 1-7 periods of a second cannabis composition. [302] In embodiments, including any of the embodiments specifically disclosed herein, the number of periods of a phase may include greater than seven periods, including eight periods, nine periods, 10 periods, 11 periods, 12 periods, 13 periods, 14 periods, 15 periods, 20 periods, 30 periods, 60 periods, 90 periods, or greater than 90 periods, and any number of periods in between. [303] In embodiments with a daily dosing regimen, the “period” as above will be by definition a day, and the number of days of each phase will be independently selected, any may include from one day to seven days inclusive, such as one day, two days, three days, four days, five days, six days, and one week, as well as a number of days greater than seven days, such as eight days, nine days, 10 days, 11 days, 12 days, and greater than 12 days. [304] In embodiments, the defined dosing regimen comprises alternating phases of between 2-7 days of administration and 2-7 days of abstaining (washout). In embodiments, the defined dosing regimen comprises alternating phases of between 2-6 days of administration and 2-6 days of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 2-5 days of administration and 2-5 days of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 2-4 days of administration and 2-4 days of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 2-3 days of administration and 2-3 days of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 3-4 days of administration and 3-4 days of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 3-5 days of administration and 3-5 days of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 3-6 days of administration and 3-6 days of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 3-7 days of administration and 3-7 days of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 4-7 days of administration and 4-7 days of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 4-6 days of administration and 4-6 days of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 4-5 days of administration and 4-5 days of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 6-7 days of administration and 6-7 days of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 5-7 days of administration and 5-7 days of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 1-7 days of administration and 1-7 days of abstaining.
[305] In embodiments, the defined dosing regimen comprises alternating phases of between
2-7 days of a first composition and 2-7 days of a second composition. In embodiments, the defined dosing regimen comprises alternating phases of between 2-6 days of a first composition and 2-6 days of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 2-5 days of a first composition and 2-5 days of a second composition. In embodiments, the defined dosing regimen comprises alternating phases of between 2-4 days of a first composition and 2-4 days of a second composition. In embodiments, the defined dosing regimen comprises alternating phases of between 2-3 days of a first composition and 2-3 days of a second composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-4 days of a first composition and
3-4 days of a second composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-5 days of a first composition and 3-5 days of a second composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-6 days of a first composition and 3-6 days of a second composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-7 days of a first composition and 3-7 days of a second composition. In embodiments, the defined dosing regimen comprises alternating phases of between 4-7 days of a first composition and 4-7 days of a second composition. In embodiments, the defined dosing regimen comprises alternating phases of between 4-6 days of a first composition and 4-6 days of a second composition. In embodiments, the defined dosing regimen comprises alternating phases of between 4-5 days of a first composition and 4-5 days of a second composition. In embodiments, the defined dosing regimen comprises alternating phases of between 6-7 days of a first composition and 6-7 days of a second composition. In embodiments, the defined dosing regimen comprises alternating phases of between 5-7 days of a first composition and
5-7 days of a second composition. In embodiments, the defined dosing regimen comprises alternating phases of between 1-7 days of a first composition and 1-7 days of a second composition.
[306] In embodiments, the defined dosing regimen comprises alternating phases of between 2-7 days of a mushroom composition and 2-7 days of a cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 2-6 days of a mushroom composition and 2-6 days of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 2-5 days of a mushroom composition and
2-5 days of a cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 2-4 days of a mushroom composition and 2-4 days of a cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 2-3 days of a mushroom composition and 2-3 days of a cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-4 days of a mushroom composition and 3-4 days of a cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-5 days of a mushroom composition and 3-5 days of a cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-6 days of a mushroom composition and 3-6 days of a cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-7 days of a mushroom composition and
3-7 days of a cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 4-7 days of a mushroom composition and 4-7 days of a cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 4-6 days of a mushroom composition and 4-6 days of a cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 4-5 days of a mushroom composition and 4-5 days of a cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 6-7 days of a mushroom composition and 6-7 days of a cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 5-7 days of a mushroom composition and 5-7 days of a cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 1-7 days of a mushroom composition and
1-7 days of a cannabis composition.
[307] In embodiments, the defined dosing regimen comprises alternating phases of between
2-7 days of a first mushroom composition and 2-7 days of a second mushroom composition. In embodiments, the defined dosing regimen comprises alternating phases of between 2-6 days of a first mushroom composition and 2-6 days of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 2-5 days of a first mushroom composition and 2-5 days of a second mushroom composition. In embodiments, the defined dosing regimen comprises alternating phases of between 2-4 days of a first mushroom composition and 2-4 days of a second mushroom composition. In embodiments, the defined dosing regimen comprises alternating phases of between 2-3 days of a first mushroom composition and 2-3 days of a second mushroom composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-4 days of a first mushroom composition and 3-4 days of a second mushroom composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-5 days of a first mushroom composition and 3-5 days of a second mushroom composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-6 days of a first mushroom composition and 3-6 days of a second mushroom composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-7 days of a first mushroom composition and 3-7 days of a second mushroom composition. In embodiments, the defined dosing regimen comprises alternating phases of between 4-7 days of a first mushroom composition and 4-7 days of a second mushroom composition. In embodiments, the defined dosing regimen comprises alternating phases of between 4-6 days of a first mushroom composition and 4-6 days of a second mushroom composition. In embodiments, the defined dosing regimen comprises alternating phases of between 4-5 days of a first mushroom composition and 4-5 days of a second mushroom composition. In embodiments, the defined dosing regimen comprises alternating phases of between 6-7 days of a first mushroom composition and 6-7 days of a second mushroom composition. In embodiments, the defined dosing regimen comprises alternating phases of between 5-7 days of a first mushroom composition and 5-7 days of a second mushroom composition. In embodiments, the defined dosing regimen comprises alternating phases of between 1-7 days of a first mushroom composition and 1-7 days of a second mushroom composition. [308] In embodiments, the defined dosing regimen comprises alternating phases of between 2-7 days of a first cannabis composition and 2-7 days of a second cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 2-6 days of a first cannabis composition and 2-6 days of abstaining. In embodiments, the defined dosing regimen comprises alternating phases of between 2-5 days of a first cannabis composition and 2-5 days of a second cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 2-4 days of a first cannabis composition and 2-4 days of a second cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 2-3 days of a first cannabis composition and 2-3 days of a second cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-4 days of a first cannabis composition and 3-4 days of a second cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-5 days of a first cannabis composition and 3-5 days of a second cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-6 days of a first cannabis composition and 3-6 days of a second cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 3-7 days of a first cannabis composition and 3-7 days of a second cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 4-7 days of a first cannabis composition and 4-7 days of a second cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 4-6 days of a first cannabis composition and 4-6 days of a second cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 4-5 days of a first cannabis composition and 4-5 days of a second cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 6-7 days of a first cannabis composition and 6-7 days of a second cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 5-7 days of a first cannabis composition and 5-7 days of a second cannabis composition. In embodiments, the defined dosing regimen comprises alternating phases of between 1-7 days of a first cannabis composition and 1-7 days of a second cannabis composition.
[309] In embodiments, including any of the embodiments specifically disclosed herein, the number of days of a phase may include greater than seven days, including eight days, nine days, 10 days, 11 days, 12 days, 13 days, two weeks, three weeks, one month, 60 days, 90 days, or greater than 90 days, and any number of days in between. [310] In certain preferred embodiments, the defined dosing regimen is a “weekly” dosing regimen. For example, one weekly regimen comprises alternating phases of four days and three days (understood to be equivalent to alternating phases of three days and four days, wherein the phase that is considered as “first” or “second” is simply reversed, and immaterial to the ultimate practice of the embodiment). In other embodiments, an alternating dosing regimen may comprise phases of two days and five days. In other embodiments, an alternating dosing regimen may comprise phases of one day and six days, or any other combination of phases that add up to seven days, such as two days and three days and two days, or one day and four days and two days, or one day and five days and one day, and the like (including variations thereof, wherein the “order” of the phases is immaterial, as above).
[311] Although such “weekly” regimens are easier to remember and thus in some embodiments may be preferable for compliance, “every other day” regimens also aid in compliance, and regimens can be adjusted according to the needs of a patient, and may include other such schedules such as two on and two off, three on and three off, four on and four off, etc., or various other such schedules (two on, three off, two on, two off; three on, four off, three on, two off; two on, two off, two on, five off; etc.). In embodiments, a longer “washout” between doses is adopted, such as one dose every third day, or one every fourth day (i.e., two or three days of abstaining in between). Longer alternating schedules (one week on, one week off; one week on, two weeks off, etc.) are also contemplated. To aid in compliance, calendar alerts and reminders can be used, as well as web-based or computer-based programs, applications, services, schedules, and the like.
[312] In embodiments, a phase of days “abstaining” or “washout” will mean that no composition of the invention is administered. In other embodiments, a phase will comprise administration of a mushroom composition of the invention wherein psilocybin, psilocin, or their salts, prodrugs, metabolites, polymorphs, analogs, or derivatives, are not present, nor are present any other psychedelic or psychoactive tryptamines, such as minor tryptamines from a Psilocybe or other fungi, including even at dosages insufficient to have psychedelic effects (microdoses). Such mushroom compositions (herein, “M-” mushroom compositions or “M-” compositions) will in some embodiments include a mushroom composition identical in all other respects but absent psilocybin or a psilocybin-containing ingredient such as a Psilocybe extract (e.g., any of the above mushroom compositions of the Examples, without psilocybin or a Psilocybe extract, but with the additional extracts and ingredients, and otherwise modified as will be readily appreciated by those of ordinary skill for general consistency of the formulation). In other embodiments, a phase will comprise administration of a cannabis composition of the invention (such as those disclosed in the Examples) that does not contain THC or an intoxicating THC derivative or analog (i.e., a THC derivative or analog capable of causing a subject to get “high” when ingested or otherwise administered at dosages used in the art) (herein, “ C– ” cannabis compositions or “ C– ” compositions). [313] In embodiments, doses of a mushroom composition of the invention, for instance as in EXAMPLES 1-7 , will be alternated with doses of a cannabis composition of the invention, for instance as in EXAMPLES 8-19. In such embodiments, the mushroom composition may or may not contain psilocybin (i.e., M+ and M– , respectively) and the cannabis composition may or may not contain THC (i.e., C+ and C– , respectively). [314] In embodiments, the defined dosing regimen is a dosing regimen comprising between about 1 and about 5 administrations of a mushroom composition that does contain psilocybin (including a composition of an effective amount of psilocybin alone, all other ingredients, whether active or active agents, optional), followed by 1-5 administrations of any of, or a combination thereof, a cannabis composition containing THC, a cannabis composition that does not contain THC, a mushroom composition that does not contain psilocybin, and abstaining from administration. In embodiments, a total therapy regime is completed for between about 15 and about 45 days, inclusive. In some preferred embodiments, the therapy regime is completed for 30 days. In embodiments, the therapy regime may be repeated a second time, for a total of about 30-90 days, or for 60 days. The therapy regime may be repeated more than two times, including three times, four times, five times, six times, seven times, eight times, nine times, 10 times, and more than 10 times. [315] In embodiments, the defined dosing regimen is a daily dosing regimen comprising between about 1 and about 5 days of a mushroom composition that does contain psilocybin (including a composition of an effective amount of psilocybin alone, all other ingredients, whether active or active agents, optional), followed by 1-5 days of any of, or a combination thereof, a cannabis composition containing THC, a cannabis composition that does not contain THC, a mushroom composition that does not contain psilocybin, and abstaining from dosing. In embodiments, a total therapy regime is completed for between about 30 and about 90 days, inclusive. In some preferred embodiments, the therapy regime is completed for 60 days. In embodiments, the therapy regime may be repeated a second time, for a total of about 60-180 days, or for 120 days. The therapy regime may be repeated more than two times, including three times, four times, five times, six times, seven times, eight times, nine times, 10 times, and more than 10 times. [316] In embodiments, the defined dosing regimen is a dosing regimen comprising an alternating schedule of administration, wherein the mushroom composition containing psilocybin is administered for a set number of administrations, followed by administration of any of a cannabis composition containing THC, a cannabis composition that does not contain THC, a mushroom composition that does not contain psilocybin, and abstaining from administration, for a set number of total administrations.
[317] In embodiments, the set number of administrations is between about 1 to about 7 administrations. In embodiments, a therapy regime comprises the set number of administrations. In embodiments, the set number of administrations comprises more than one defined dosing regimen, and is part of a therapy regime, which comprises two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, 10 or more, 11 or more, 12 or more, or more than 12 defined dosing regimens, which may be all the same or may be independently selected.
[318] In embodiments, the therapy regime comprising the defined dosing regimen is for at least 21 administrations, at least 22 administrations, at least 23 administrations, at least 24 administrations, at least 25 administrations, at least 26 administrations, at least 27 administrations, at least 28 administrations, at least 29 administrations, at least 30 administrations, at least 31 administrations, at least 32 administrations, at least 33 administrations, at least 34 administrations, at least 35 administrations, at least 36 administrations, at least 37 administrations, at least 38 administrations, at least 39 administrations, at least 40 administrations, at least 41 administrations, at least 42 administrations, at least 43 administrations, at least 44 administrations, at least 45 administrations, at least 46 administrations, at least 47 administrations, at least 48 administrations, at least 49 administrations, at least 50 administrations, at least 51 administrations, at least 52 administrations, at least 53 administrations, at least 54 administrations, at least 55 administrations, at least 56 administrations, at least 57 administrations, at least 58 administrations, at least 59 administrations, at least 60 administrations, at least 61 administrations, at least 62 administrations, at least 63 administrations, at least 64 administrations, at least 65 administrations, at least 66 administrations, at least 67 administrations, at least 68 administrations, at least 69 administrations, at least 70 administrations, at least 71 administrations, at least 72 administrations, at least 73 administrations, at least 74 administrations, at least 75 administrations, at least 76 administrations, at least 77 administrations, at least 78 administrations, at least 79 administrations, at least 80 administrations, at least 90 administrations, or more than 90 administrations. As above, in some embodiments an “administration” may include an “administration” of “abstinence” or “washout.” “Administration” may in such embodiments be used equivalently to “period,” as used above. [319] In embodiments, the defined dosing regimen is a daily dosing regimen comprising an alternating schedule of daily administration, wherein the mushroom composition containing psilocybin is administered for a set number of days, followed by administration of any of a cannabis composition containing THC, a cannabis composition that does not contain THC, a mushroom composition that does not contain psilocybin, and abstaining from dosing, for a set number of total days. [320] In embodiments, the set number of days is between about 1 to about 7 days. In embodiments, a therapy regime comprises the set number of days. In embodiments, the set number of days comprises more than one daily dosing regimen, and is part of a therapy regime, which comprises two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, 10 or more, 11 or more, 12 or more, or more than 12 daily dosing regimens, which may be all the same or may be independently selected. [321] In embodiments, the therapy regime comprising the more than one daily dosing regimens is for at least 21 days, at least 22 days, at least 23 days, at least 24 days, at least 25 days, at least 26 days, at least 27 days, at least 28 days, at least 29 days, at least 30 days, at least 31 days, at least 32 days, at least 33 days, at least 34 days, at least 35 days, at least 36 days, at least 37 days, at least 38 days, at least 39 days, at least 40 days, at least 41 days, at least 42 days, at least 43 days, at least 44 days, at least 45 days, at least 46 days, at least 47 days, at least 48 days, at least 49 days, at least 50 days, at least 51 days, at least 52 days, at least 53 days, at least 54 days, at least 55 days, at least 56 days, at least 57 days, at least 58 days, at least 59 days, at least 60 days, at least 61 days, at least 62 days, at least 63 days, at least 64 days, at least 65 days, at least 66 days, at least 67 days, at least 68 days, at least 69 days, at least 70 days, at least 71 days, at least 72 days, at least 73 days, at least 74 days, at least 75 days, at least 76 days, at least 77 days, at least 78 days, at least 79 days, at least 80 days, at least 90 days, or more than 90 days. [322] In some preferred embodiments, a mushroom composition containing psilocybin is administered for 4 consecutive days. In such embodiments, the 4 consecutive days of administration of the mushroom composition containing psilocybin is followed and/or preceded by any of, or a combination thereof, administration of a cannabis composition containing THC, a cannabis composition that does not contain THC, a mushroom composition that does not contain psilocybin; or abstinence from dosing for 3 consecutive days. In such embodiments, the therapeutic regime may continue for between about 45 days to about 90 days inclusive, including about 60 days. [323] In embodiments, a mushroom composition of the invention containing psilocybin (i.e., a M+ composition) may be administered consecutively for for one, two, three, four, five, six, or seven days; may be administered for one day followed by one, two, three, four, five, six, or seven days of a M–, C+, C– composition or of abstaining; may be administered for two days followed by one, two, three, four, five, six, or seven days a M–, C+, C– composition or of abstaining; may be administered for three days followed by one, two, three, four, five, six, or seven days a M–, C+, C– composition or of abstaining; may be administered for four days followed by one, two, three, four, five, six, or seven days a M–, C+, C– composition or of abstaining; may be administered for five days followed by one, two, three, four, five, six, or seven days a M–, C+, C– composition or of abstaining; may be administered for six days followed by one, two, three, four, five, six, or seven days a M–, C+, C– composition or of abstaining; may be administered for seven days, followed by one, two, three, four, five, six, or seven days a M–, C+, C– composition or of abstaining; or may be administered in any other such combination of days “on” (i.e., days wherein a mushroom composition of the invention containing psilocybin is administered to the patient), combined with any number of days “a M–, C+, C– composition or of abstaining” (i.e., wherein either no dosing occurs, or dosing of any of a mushroom composition of the invention without psilocybin, a cannabis composition of the invention with THC, or a cannabis composition of the invention without THC occurs). Exemplary dosing regimens are in EXAMPLES 22-205. [324] In other embodiments, a mushroom composition, for instance as in EXAMPLES 1-7 (either as M+ or as M– compositions), can be alternated with doses containing a cannabis composition, for instance as in EXAMPLES 8-19 (either as C+ or as C– compositions), or alternated with days abstaining (washout), including where days abstaining are of a composition that is not an M or a C composition, but comprises other active ingredients. [325] In embodiments, including some preferred embodiments, a psychedelic mushroom composition (e.g., a composition comprising psilocybin and/or psilocin) is not administered sequentially with a cannabis composition (e.g., a composition comprising a cannabinoid and/or a terpene). In some exemplary embodiments, such compositions are not administered sequentially because one or more washout days are in between a day administering a composition comprising psilocybin and/or psilocin, and a day administering a composition comprising a cannabinoid and/or a terpene. In embodiments, such compositions are not administered sequentially because one or more days of administering an M– or functional mushroom composition (i.e., not comprising psilocybin and/or psilocin) are in between a day administering a composition comprising psilocybin and/or psilocin, and a day administering a composition comprising a cannabinoid and/or a terpene. In embodiments, such compositions are not administered sequentially because one or more administrations of an M– or functional mushroom composition (i.e., not comprising psilocybin and/or psilocin) are in between administrations of a composition comprising psilocybin and/or psilocin, and administrations of a composition comprising a cannabinoid and/or a terpene. [326] The teaching of dosage ranges at which therapeutic benefits are achieved without certain drawbacks (e.g., side effects, or undesired psychedelic or intoxicating effects), as well as dosage regimens at which these benefits are maintained while also minimizing risk, are among the advantages and improvements of the compositions and methods of the invention. P. Defined Dosing Kits and Daily Dosing Kits [327] Another aspect of this disclosure provides “pharmaceutical kits” containing the mushroom compositions and/or cannabis compositions of the invention, suggested administration guidelines or prescribing information (if necessary, if by prescription) for the composition, and a suitable container. Individual unit dosage forms can be included in multi-dose kits or containers. Mushroom and cannabis compositions also can be packaged in single or multiple unit dosage forms for ease of administration and uniformity of dosage. In embodiments with an alternating dosing schedule, kits may be utilized to great advantage to assist with compliance, e.g., by using marked blister packs. It will be readily appreciated that use of the term “pharmaceutical” in “pharmaceutical kit” does not imply or necessitate that the kit be offered solely by prescription, or that the compounds or compositions within must be “pharmaceuticals” or for administration by prescription or under the supervision of a doctor or health professional, and “pharmaceutical kits” may be provided to patients or individuals with or without a prescription, including sold OTC, online, at “dispensaries,” and in one example, offered for “take home” purchase by a state legal psilocybin service center. “Pharmaceutical kits” is equivalent to and includes “defined dosing” and “daily dosing” kits. [328] In an exemplary “defined dosing kit” or “daily dosing kit” (the terms having equivalent meaning, except for the suggestion, in some embodiments, or when needed for technical effect, that a daily dosing kit be used for “daily” administration, which a “defined” dosing kit may be used for administration on any schedule, including but not limited to daily) capsules, tablets, caplets, or other unit dosage forms are packaged in blister packs. “Blister pack” may refer to any of several types of pre-formed containers, especially with plastic packaging, that contain separate receptacles (e.g., cavities or pockets) for single unit doses, where the receptacles are individually sealed and opened individually. Blister packs thus include such pharmaceutical blister packs known to those of ordinary skill, including Aclar® Rx160, Rx20e, SupRx, and UltRx 2000, 3000, 4000, and 6000 (Honeywell). Within the definition of multi-dose containers, and also often referred to as blister packs, are blister trays, blister cards, strip packs, push-through packs, and the like. [329] Defined dosing kits can be used to keep track of defined dosing regimens. As an example, daily dosing kits can be used to keep track of daily dosing regimens. In embodiments, a “compliance pack” or a “calendar pack” is created by printing the dose numbers or the days of the week above each dose. Where doses are administered more than once per day, such as twice per day, the printing may comprise “day” and “night,” or “breakfast,” “lunch,” and “dinner,” or any like appropriate predetermined dosing times. A number of compliance or calendar packs, containers, and kits can be utilized, including labeled blister packs, packages having indicators for each day, and dial dispenser containers. [330] In embodiments, information pertaining to dosing and proper administration (if needed) is printed on a multi-dose kit directly (e.g., on a blister pack or other interior packaging holding the compositions of the invention). In other embodiments, kits can further contain package inserts and other printed instructions (e.g., on exterior packaging) for administering the compositions of the invention and for their appropriate therapeutic use. [331] In embodiments, the defined and daily dosing kits containing the compositions of the invention are designed to facilitate adherence to the defined and daily dosing regimens and therapeutic regimes described herein. In one such exemplary, non-limiting embodiment, the kits are designed for daily oral administration over 7-day, 14-day, 21-day, 28-day, 30-day, 45-day, 60-day, 75-day, or 90 day cycle, wherein the mushroom composition and cannabis composition are packaged such that their consumption in the prescribed order is facilitated. [332] Daily dosing kits can comprise the same period daily dosing regimen repeated a set number of times (e.g., a 7-day daily dosing regimen, repeated two times, three times, four times, five times, six times, seven times, eight times, nine times, 10 times, 11 times, 12 times, or greater than 12 times; or a shorter or longer daily dosing regimen, with less or more days). [333] In other embodiments, defined and daily dosing kits will comprise different dosing regimens repeated a number of times, in a distinct pattern, where together the pattern of dosing regimens comprises a therapeutic regime. For example, a kit of FIG.1 can be viewed as showing a single 7-day daily dosing regimen ( MMMMCCC ) three times (although it will be readily appreciated that in some embodiments, each “M” and “C” may be selected independently, and need not all be the same M and C composition. To compare, the kit of FIG.2 can be viewed as showing different 7-day daily dosing regimens ( MMMMACA , MMMMCAC , MAMAACA ), which may be followed in order, comprising a 21-day therapeutic regime. Distinct or daily dosing regimens, and distinct or daily dosing kits, can be further combined to create longer therapeutic regimes, e.g., the daily dosing kit of FIG.1 and/or FIG. 2 can be combined to create six week, nine week, 12 week, and greater therapeutic regimes, and like logic will be understood as being able to be used to create any number of regimes, including by splitting a daily regimen into a multi-dose per day regimen. [334] To illustrate several exemplary kits, reference can be made to FIGS. 1-3. For example, FIG.1 illustrates an exemplary daily dosing kit ( 1901 ) containing a 21-day supply of the compositions of the invention, according to one embodiment. As is evident from the figure, the daily dosing kit ( 1901 ) contains a total of 21 doses of the mushroom composition ( 1903 ) of EXAMPLE 2 or EXAMPLE 3 , and the cannabis composition ( 1904 ) of EXAMPLE 9 , as well as an indicator that alerts the patient to when each dose should be administered ( 1902 ), i.e., on what day of a seven day cycle (according to this embodiment). [335] As apparent to one of skill, this is an exemplary embodiment, and not to be construed as limiting the invention. In other embodiments, the daily dosing kit containing doses of the compositions of the invention may contain any conceivable dosing schedule (i.e., regimen) involving any such mushroom and/or cannabis composition, including those that do or do not contain psilocybin and THC, respectively. Moreover, the daily dosing kit may additionally contain periodic “abstinent” or “washout” days, wherein a patient does not take any of the compositions of the invention, as is further illustrated in FIG.2. [336] As mentioned, FIG.2 illustrates an exemplary daily dosing kit ( 2001 ) according to an embodiment of the invention comprising the administration on given days ( 2002 ) a mushroom composition, which may or may not contain psilocybin ( 2003 ), i.e., where 2003 may be either “M+” or “M–”; and the administration of a cannabis composition, which may or may not contain THC ( 2005 ), i.e., where 2005 may be either “C+” or “C–”. FIG.2 additionally includes a number of “abstinent” days (e.g., 2004 ), wherein the individual is not administered (either by the individual or a third party) any of the mushroom or cannabis compositions disclosed herein. In embodiments an abstinent (“A”) day, when part of a daily dosing kit, may be a “blank” space or container, e.g., an empty blister where a blister pack is used as the daily dosing kit. In other embodiments, an “A” day may be a “blank” pill or composition, e.g., a placebo or “sugar pill,” as may be contained within kits comprising birth control pills. In yet other embodiments, including in some preferred embodiments, an “A” day may be a pill that is not any mushroom composition or cannabis composition of the invention, but comprises one or more bioactive compounds, such as vitamins, nootropics, dietary supplements, and the like. [337] In embodiments, as shown in FIG.2 , the M and C compositions therefore are not administered sequentially. In embodiments, as shown in FIG.2 , the M+ and C compositions therefore are not administered sequentially. In embodiments, as shown in FIG.2 , the M+ and C+ compositions therefore are not administered sequentially. In embodiments, as shown in FIG. 2 , the M+ and C– compositions therefore are not administered sequentially. In embodiments, as shown in FIG.2 , M+ and C compositions are administered with at least or more of a M– day or an A day in between. [338] It will be readily appreciated that the placement of abstinent days in FIG.2 (as well as the placement of the administration of either of a mushroom composition or a cannabis composition) should not be construed as limiting the spirit or scope of the invention. As will be apparent to one of skill, a therapeutic regime, including any one or more distinct and/or daily dosing regimens which comprise it, may be tailored to an individual patient, for example based on any number of patient-selected, prescriber-selected, or other patient-related information, such as based on one or more “objective health measurements” (including “objective indicia of improvement”), the specific indication being treated, and the like. [339] Thus, a “distinct dosing kit” may contain a dosing schedule requiring the administration of a mushroom composition for one administration, or two, three, four, five, six, or seven consecutive administrations; with one day, or two, three, four, five, six, or seven consecutive administrations of a cannabis composition; or one day, or two, three, four, five, six, or seven consecutive dosing periods of abstinence separating the mushroom administrations and/or cannabis administrations; or a combination thereof (wherein the mushroom composition may or may not contain psilocybin, and the cannabis composition may or may not contain THC). Meaning, in some embodiments, the daily or distinct dosing kit may facilitate the administration of a mushroom composition containing psilocybin; followed by any of administration of a mushroom composition containing psilocybin, administration of a mushroom composition that does not contain psilocybin, administration of a cannabis composition containing THC, administration of a cannabis composition that does not contain THC, or abstinence, wherein no administration occurs. In such embodiments, the pattern may continue until the duration of the treatment program ends, or may fluctuate according to the specific patient’s needs, or the indication being treated. [340] Further, a “daily dosing kit” may contain a dosing schedule requiring the administration of a mushroom composition for one day, or two, three, four, five, six, or seven consecutive days; with one day, or two, three, four, five, six, or seven consecutive days of administration of a cannabis composition; or one day, or two, three, four, five, six, or seven consecutive days of abstinence separating the mushroom dosing days and/or cannabis dosing days; or a combination thereof (wherein the mushroom composition may or may not contain psilocybin, and the cannabis composition may or may not contain THC). [341] In embodiments, as illustrated in FIG. 3 , the non-psilocybin (M–) mushroom administration days are not scheduled abstinent, mushroom, or cannabis composition dosing days, but may fluctuate depending on patient needs. As illustrated in FIG.3 , the daily dosing kit ( 2101 ) includes four consecutive days ( 2103 ) of administration of the mushroom composition of EXAMPLE 2 or EXAMPLE 3 , illustrated as “ M+ ,” which indicates it contains psilocybin or another psychedelic tryptamine ( 2103 ); followed by three consecutive days of any of the administration of a cannabis composition containing THC or another intoxicating cannabinoid (including but not limited to that of EXAMPLE 9 ), illustrated as “ C+ ;” administration of a cannabis composition that does not contain THC or another intoxicating cannabinoid, illustrated as “ C– ;” administration of a mushroom composition that does not contain psilocybin or another psychedelic tryptamine, illustrated as “ M– ;” or abstinence (washout), illustrated by the letter “ A ” (i.e., the options shown in 2104 ). [342] In such an embodiment, the patient may be in communication with a practitioner who may evaluate the patient based on monitoring of the “objective health measurements” disclosed infra , or via information obtained from the patient regarding their subjective feelings via questionnaires, patient interviews, journaling, or any other such measures known to those of skill. Based on said information gathered, the clinician would then instruct the patient to either abstain from dosing, or be administered (either by the patient or a third party) the appropriate substance. Likewise, the patient may decide, based on how the patient personally feels, whether to abstain from dosing, or take one of the cannabis compositions, or a mushroom composition that does not contain psilocybin. [343] In embodiments, daily dosing kits may comprise all options (e.g., a patient may select between M+, M–, C+, C–, and A, day by day). In embodiments, a daily dosing kit may be mailed to a patient or readied for pickup by a patient depending on prior communication to the provider of the kit of the patient needs, goals, results, data, biometrics, digital biomarkers, and the like, in advance of the start of the daily dosing regimen in the kit. [344] In embodiments, a clinician may instruct the patient to be administered (either by the patient or a third party, which may or may not be the clinician) more than one of the compositions on a given day. In such embodiments—on days wherein the patient is not being administered a mushroom composition containing psilocybin ( M+ )—the patient may be administered (by the patient or a third party) at least one of a mushroom composition that does not contain psilocybin ( M– ), a cannabis composition that contains THC ( C+ ), and a cannabis composition that does not contain THC ( C– ). Thus, in some embodiments, on days wherein the patient is not being administered a mushroom composition containing psilocybin ( M+ ), the patient may be administered (by the patient or a third party), any two of a mushroom composition that does not contain psilocybin ( M– ), a cannabis composition that contains THC ( C+ ), and a cannabis composition that does not contain THC ( C– ); or all three of a mushroom composition that does not contain psilocybin ( M– ), a cannabis composition that contains THC ( C+ ), and a cannabis composition that does not contain THC ( C– ). [345] In embodiments, a clinician may instruct a patient to be administered (either by the patient or a third party, which may or may not be the clinician) more than one of the compositions on a given day, including on days wherein a composition containing psilocybin is being administered. In such embodiments, on any given day, a patient may be administered (either by the patient or a third party) a mushroom composition containing psilocybin ( M+ ), a mushroom composition not containing psilocybin ( M– ), a cannabis composition containing THC ( C+ ), and a cannabis composition not containing THC ( C– ). In such embodiments, administration may be in any order preferred by the patient and/or clinician. On any such day, a patient also may abstain from dosing, such that no compounds are being administered. [346] In embodiments, a patient will have the option of using online software such as a website, or downloadable software such as a mobile application, to assist with compliance or to collect or provide data relating to treatment. Such software can be used to, e.g., keep track of last dose taken and total doses taken, provide reminders and alerts for upcoming doses, provide feedback to discourage taking doses outside of set schedules, and allow for recording of specific subjective effects, to collect or analyze one or more biometrics or digital biomarkers, or to provide means for subject directed activities such as unstructured journaling. Such data collection can assist with individual patient compliance, can be used to improve or tailor individual patient care plans, and can be anonymized, aggregated, and analyzed (including by AI or natural language processing means) to allow research into the effects of various methods of treatment. Q. Methods for Modulating Neurotransmission and Other Biological Systems [347] Methods are provided for using therapeutically effective amounts of the mushroom compositions of the invention in a mammal, and preferably a human. Such methods include those for modulating neurotransmission, such as serotonergic neurotransmission, using the described and claimed mushroom compositions. Methods are provided for using therapeutically effective amounts of the cannabis compositions of the invention in a mammal, and preferably a human. Such methods include those for modulating endocannabinoid system activity, for modulating neurosteroid biosynthesis, and for modulating neurotransmission, using the described and claimed cannabis compositions. In some preferred embodiments, methods are provided for using therapeutically effective amounts of the mushroom compositions and the cannabis compositions in a mammal, and preferably a human, in defined dosing regimens, daily dosing regimens, and/or therapeutic regimes, as taught herein. [348] In embodiments, the mushroom compositions of the invention are useful in methods for modulating neurotransmission, and such modulation will result in treatment of a mental health condition or improvement of psychological functioning. Compositions of the invention that modulate serotonergic neurotransmission, as disclosed in embodiments herein, are effective for treatment of CNS disorders such as PTSD, and other disorders correlated with dysfunction of serotonergic neurotransmission, such as feeding disturbances (annorexia nervosa, bulimia, binge eating), depression and suicide, impulsivity and violence, anxiety and harm avoidance, obsessive-compulsive features, addiction, and disturbances in neuroendocrine and vascular tissues, as well as related to other neurochemical systems linked to serotonin (Brewerton, 1995). Further, the invention is not limited to disorders characterized by serotonin dysregulation. CNS disorders with other causes can be effectively treated by the compositions of the invention, as demonstrated through the teachings herein. [349] In embodiments, mushroom compositions of the invention are used to modulate serotonergic neurotransmission, and will be effective to treat disorders and conditions associated with such serotonergic modulation, or that can be treated thereby. In embodiments, the methods of the invention that comprise the administration of a mushroom composition will therefore find use in such modulation. [350] In embodiments, the cannabis compositions of the invention are useful in methods for modulating endocannabinoid system activity and for modulating neurotransmission, and such modulation will result in treatment of a mental health condition or improvement of psychological functioning. In one non-limiting example of patients with PTSD, various brain systems respond to threat-related and emotional stimuli in a dysfunctional way (i.e., the amygdala is hyperresponsive, whereas the ventral/medial prefrontal cortex and hippocampus are hypoactive, resulting in inadequate top–down control of amygdalar activation relative to healthy controls). This dysfunction is correlated with PTSD symptoms such as anxiety and emotional dysregulation. In embodiments, the cannabis compositions of the invention modulate this threat response by attenuating amygdala reactivity, facilitating fear extinction, enhancing consolidation of extinction learning, and reducing the reinstatement of fear responding. Through resultant modulations of neurotransmission and the ECS, the cannabis compositions of the invention will reduce perceived threat responding, improve general stress reactivity, and assist in the relearning of negatively charged emotional memories to reduce their aversive intensity. Through modulations of neurotransmission and other biological systems caused by the compositions and methods of the invention, both the emotional and cognitive aspects of PTSD will be effectively addressed, providing clinically significant benefits in the treatment of PTSD. Certain cannabis compositions of the present invention can augment and increase such benefits through, e.g., synergistic effects resulting in greater activity at CB 1 receptors, greater bioactivity of CBD, or additional therapeutic effects, and such benefits will be shown to be increased by the combination of the cannabis compositions with the mushroom compositions of the invention using the dosing methods taught herein. [351] Connections between the ECS and PTSD are supported by studies showing how ECS-related gene polymorphisms (i.e., ECS genotypes) affect the risk of PTSD and the severity of specific symptoms. For example, subjects who carry alleles associated with higher anandamide levels show lower levels of PTSD symptoms, lower stress reactivity, and more rapid extinction of fear memories. Similar correlations have been made with carriers of different alleles for the cannabinoid receptor gene (CNR1) associated with differential CB 1 gene expression. In embodiments herein, a subject with a genetic marker associated with higher anandamide, or a genetic marker associated with a reduction in the expression of CNR1 will be treated with a method specifically suited to such genetic marker, such as an increased dose of a cannabis composition, or an increased number of days of a phase of a daily dosing regimen, where such phase is for a cannabis composition. [352] In embodiments, the compositions and methods of the invention will modulate neurosteroid biosynthesis, for example by assisting in the reinstatement of normal allopregnanolone levels in allopregnanolone deficient patients. In embodiments where the compositions and methods are used to increase or rebalance levels of allopregnanolone, improvement is found in any of one or more neuropsychopathologies, such as PTSD, epilepsy, major depression, perceived social isolation, postpartum depression, premenstrual syndrome, and anorexia nervosa or obesity complicated by anxiety and depression (Pinna, 2018). Accordingly, in such embodiments allopregnanolone deficient patients will have improved symptoms of these and related disorders. In other embodiments, patients deficient in the equipotent isomer of allopregnanolone, pregnanolone, will be treated. In some aspects disclosed herein, compositions and methods of the invention therefore are used to modulate and restore levels of allopregnanolone and pregnanolone and have therapeutic benefits in patients with mental health conditions. [353] In embodiments, cannabis compositions of the invention are used to modulate endocannabinoid system (ECS) activity, and will be effective to treat disorders and conditions associated with such ECS modulation, or that can be treated thereby. In embodiments, cannabis compositions of the invention are used to modulate neurosteroid biosynthesis, and will be effective to treat disorders and conditions associated with such neurosteroid biosynthesis modulation, or that can be treated thereby. In embodiments, cannabis compositions of the invention are used to modulate serotonergic neurotransmission, and will be effective to treat disorders and conditions associated with such serotonergic modulation, or that can be treated thereby. In embodiments, the methods of the invention that comprise the administration of a cannabis composition will therefore find use in such modulation. R. Methods of Treating CNS Disorders [354] In some aspects, methods of treatment comprising administration of the mushroom and cannabis compositions of the invention are provided, such as administration by a distinct or daily dosing regimen. In embodiments, provided are methods for administration of the mushroom and cannabis compositions to treat a disease or condition in a subject or patient. In embodiments, the mushroom and cannabis compositions are used to treat CNS disorders including mental health conditions (such as psychiatric or neuropsychiatric disorders), neurodevelopmental disorders, and neurodegenerative conditions and disorders. [355] “Treating” or “treatment” of a disorder includes (i) inhibiting the disorder, i.e., arresting or reducing the development or progression of the disorder or its clinical symptoms; or (ii) relieving the disorder, i.e., causing regression of the disorder or its clinical symptoms. Inhibiting the disorder, for example, would include prophylaxis. Hence, one will understand that a therapeutic amount necessary to effect treatment for purposes of this invention will, for example, be an amount that provides for objective indicia of improvement in patients having clinically-diagnosable symptoms. The effect may be prophylactic in terms of completely or partially preventing a disorder or symptom thereof and/or may be therapeutic in terms of a partial or complete cure for a disorder and/or adverse effect attributable to the disorder. [356] “Treatment” covers any treatment of a disorder in a mammal, and particularly in a human, and includes: (a) preventing a disorder from occurring in a subject who may be predisposed to the disorder but has not yet been diagnosed with it: (b) inhibiting a disorder, i.e., arresting its development; (c) relieving a disorder, i.e., causing regression thereof; (d) protection from or relief of a symptom or pathology caused by or related to a disorder; (e) reduction, decrease, inhibition, amelioration, or prevention of onset, severity, duration, progression, frequency or probability of one or more symptoms or pathologies associated with a disorder; and (f) prevention or inhibition of a worsening or progression of symptoms or pathologies associated with a disorder. Other such measurements, benefits, and surrogate or clinical endpoints, alone or in combination, would be understood to one of ordinary skill. [357] The terms “subject,” “patient,” and “individual” are used interchangeably, and refer to any mammal, including murines, simians, humans, mammalian farm animals, mammalian sport animals, and mammalian pets. Preferably, the subject herein is human. As further used herein, the terms “subject,” “patient,” and “individual” includes a subject or patient who has a mental health condition, or a condition related to a mental health condition for which similar treatment may be efficacious. Such terms shall also refer to patients in need of treatment for such a disorder, persons predisposed to such a disorder, and subjects who have been diagnosed with such a disorder. Moreover, these terms shall likewise refer to persons who have received treatment or therapy for a mental health condition, are currently receiving therapy or treatment for a mental health condition, or who may receive therapy or treatment for such a disorder in the future. In embodiments, the disclosed methods also can be used to improve mental health and improve psychological functioning in non-disease states, i.e., in an individual without a diagnosed mental disorder, or specific symptoms thereof. [358] In embodiments, a therapeutically effective amount of a mushroom or cannabis composition is administered to a subject. “Effective,” “an effective amount,” “effective dose,” “sufficient amount,” “sufficient dose,” “therapeutically effective,” “a therapeutically effective amount,” “a therapeutically effective dose,” “therapeutically sufficient amount,” “therapeutically sufficient dose,” or “a pharmacologically effective amount” refer to an amount of a composition (or an active agent thereof, depending on context) that is sufficient to provide a desired therapeutic effect. An effective amount will vary depending upon the subject and the disease condition being treated or health benefit sought, the weight and age of the subject, the severity of the disease condition or degree of health benefit sought, the manner of administration, and the like, all of which will be readily determined by one of skill. [359] “Therapeutic effect” or “therapeutic efficacy” means the responses(s) in a mammal, and preferably a human, after treatment that is judged to be desirable and beneficial. Hence, depending on the disorder to be treated, or improvement in physiological or psychological functioning sought, and depending on the particular constituent(s) in the compositions of the invention under consideration, those responses shall differ, but would be readily understood by those of skill. In preferred embodiments, the mammal is a human. [360] Measures of therapeutic effect include outcome measures (primary or secondary), endpoints, effect measures, and measures of effect within clinical or medical practice or research which can be used to assess an effect (positive and/or negative) of an intervention or treatment, whether patient-reported (e.g., questionnaires); based on other patient data (e.g., patient monitoring); gathered through laboratory tests such as from blood or urine; through medical examination by a doctor or medical professional, or by digital or electronic means, including online tools, smartphones, wireless devices, biosensors, health apps, and the like. [361] In embodiments, the compositions of the invention may be administered to the patient (alone, with another patient as in couples therapy, or as part of group therapy) in the presence of one or more therapists in a therapeutic setting to facilitate psychotherapy. Numerous such treatment paradigms are described in the art, and referred to as psychedelic-assisted psychotherapy or drug-assisted therapy ( see, e.g., Sessa, 2019, and references therein). In other embodiments, including some preferred embodiments, the compositions of the invention will be administered not as part of a psychotherapeutic regimen, or as part of drug-assisted therapy, but will be self-administered by a patient, and such self-administration may or may not be under the supervision of a medical professional. [362] In various embodiments, a patient may be administered the compositions of the invention and be monitored, a patient may be administered the compositions of the invention and receive psychological support, and a patient may be administered the compositions of the invention and receive psychotherapy, and such may take place for example with a psychiatrist, medical doctor, clinical psychologist, or other trained clinician, as well as with a “guide” or non-clinical practitioner. “Therapist” herein therefore may refer to any person who treats a patient using the compositions and methods of the invention, whether that person is a care provider, a psychiatrist, clinical psychologist, clinical therapist, psychotherapist, or other trained counselor, facilitator, or guide, and who may or may not be a trained counselor. [363] In embodiments, after having received a dose of a composition of the invention, the patient is supervised or monitored by a clinician, guide, therapist, or care provider. In embodiments, such supervision or monitoring may be completed in person or remotely, for example, over the telephone (e.g., telemedicine), through video conferencing (e.g., Zoom, Teams, Skype, FaceTime), with the use of virtual reality (VR), including both synchronous and asynchronous means, or otherwise through software or an app. In embodiments, a subject may or may not be supervised, and may or may not be instructed on how to self-administer the composition. In embodiments wherein instruction is provided, it may be in the form of physical instructions that accompany a composition of the invention, through software or an app, or through publicly available information. [364] In embodiments, a subject completes one or more remote consultations with a care provider, e.g., by phone, by video conference, or in virtual reality (VR), prior to administration of the compositions. In embodiments, remote consultations are used by the care provider to determine whether administration of the compositions is appropriate for the subject. In embodiments, the one or more remote consultations may be used by the care provider to determine and/or confirm diagnosis of a condition, such as but not limited to a CNS disorder, which can be treated by administration of a compound provided herein. In embodiments, the one or more consultations is conducted in person. [365] In embodiments, a subject completes one or more in person preparatory sessions with a care provider, or one or more remote preparatory sessions, e.g., by phone, by video conference, or in virtual reality, prior to administration of the compositions of the invention. [366] In embodiments, a subject completes one or more in person integration sessions with a care provider, or one or more remote integration sessions, e.g., by phone, by video conference, or in virtual reality, after administration of the compositions of the invention. [367] In embodiments, the compositions of the invention are used to treat CNS disorders, including mental conditions and mental health disorders, psychiatric and neuropsychiatric disorders, and neurodegenerative conditions. In embodiments, such disorders may be characterized as those codable as “mental, behavioral, or neurodevelopmental disorders” per the ICD-11 and/or its predecessor ICD-10. Per the ICD-11, such disorders are characterized by clinically significant disturbance in an individual's cognition, emotional regulation, or behavior that reflects a dysfunction in the psychological, biological, or developmental processes that underlie mental and behavioral functioning. These disturbances are usually associated with distress or impairment in personal, family, social, educational, occupational, or other important areas of functioning (World Health Organization, 2019). In embodiments, CNS disorders further include conditions involving the brain and spinal cord caused by any of genetic factors, trauma, infections, degenerations, structural defects and/or damages, tumors, blood flow disruption, autoimmune disorders, or a combination thereof.
[368] Non-limiting examples of disorders and conditions that may be treated by the compositions and methods disclosed herein include CNS disorders, which refer to any disease of the nervous system, including diseases that involve the central nervous system (brain, brainstem, spinal cord, and cerebellum), the peripheral nervous system (including cranial nerves), and the autonomic nervous system (parts of which are located in both central and peripheral nervous system). Neurodegenerative disorders refer to a type of neurological disease marked by the loss of nerve cells, including, but not limited to, Alzheimer's disease, Parkinson's disease, frontotemporal dementia, multiple sclerosis, amyotrophic lateral sclerosis, tauopathies (including frontotemporal dementia), multiple system atrophy, and Huntington's disease. Examples of neurological diseases include, but are not limited to, headache, stupor and coma, dementia, seizure, sleep disorders, trauma, infections, neoplasms, neuro-ophthalmopathy, movement disorders, demyelinating diseases, spinal cord disorders, and disorders of peripheral nerves, muscle and neuromuscular junctions. Addiction and mental illness, include, but are not limited to, bipolar disorder, depression and schizophrenia, are also included in the definition of CNS diseases. Further examples of neurological diseases include acquired epileptiform aphasia; aaccuuttee disseminated encephalomyelitis; adrenoleukodystrophy; agenesis of the corpus callosum; agnosia; Aicardi syndrome; Alexander disease; Alpers' disease; alternating hemiplegia; Alzheimer's disease; amyotrophic lateral sclerosis; anencephaly; Angelman syndrome; angiomatosis; anoxia; aphasia; apraxia; arachnoid cysts; arachnoiditis; Arnold-Chiari malformation; arteriovenous malformation; Asperger syndrome; ataxia telangiectasia; attention deficit hyperactivity disorder; autism; autonomic dysfunction; back pain; Batten disease; Behcet's disease; Bell's palsy; benign essential blepharospasm; benign focal amyotrophy; benign intracranial hypertension; Binswanger's disease; blepharospasm; Bloch Sulzberger syndrome; brachial plexus injury; brain abscess; brain injury; brain tumors (including glioblastoma multiforme); spinal cord tumor; Brown-Sequard syndrome; Canavan disease; carpal tunnel syndrome (CTS); causalgia; central pain syndrome; central pontine myelinolysis; cephalic disorder; cerebral aneurysm; cerebral arteriosclerosis; cerebral atrophy; cerebral gigantism; cerebral palsy; Charcot-Marie-Tooth disease; chemotherapy-induced neuropathy and neuropathic pain; Chiari malformation; chorea; chronic inflammatory demyelinating polyneuropathy (CIDP); chronic pain; chronic regional pain syndrome; Coffin Lowry syndrome; coma, including persistent vegetative state; congenital facial diplegia; corticobasal degeneration; cranial arteritis; craniosynostosis; Creutzfeldt-Jakob disease; cumulative trauma disorders; Cushing's syndrome; cytomegalic inclusion body disease (CIBD); cytomegalovirus infection; dancing eyes-dancing feet syndrome; Dandy-Walker syndrome; Dawson disease; De Morsier's syndrome; Dejerine-Klumpke palsy; dementia; dermatomyositis; diabetic neuropathy; diffuse sclerosis; dysautonomia; dysgraphia; dyslexia; dystonias; early infantile epileptic encephalopathy; empty sella syndrome; encephalitis; encephaloceles; encephalotrigeminal angiomatosis; epilepsy; Erb's palsy; essential tremor; Fabry's disease; Fahr's syndrome; fainting; familial spastic paralysis; febrile seizures; Fisher syndrome; Friedreich's ataxia; frontotemporal dementia and other “tauopathies”; Gaucher's disease; Gerstmann's syndrome; giant cell arteritis; giant cell inclusion disease; globoid cell leukodystrophy; Guillain-Barre syndrome; HTLV-1 associated myelopathy; Hallervorden-Spatz disease; head injury; headache; hemifacial spasm; hereditary spastic paraplegia; heredopathia atactica polyneuritiformis; herpes zoster oticus; herpes zoster; Hirayama syndrome; HIV-associated dementia and neuropathy (see also neurological manifestations of AIDS); holoprosencephaly; Huntington's disease and other polyglutamine repeat diseases; hydranencephaly; hydrocephalus; hypercortisolism; hypoxia; immune-mediated encephalomyelitis; inclusion body myositis; incontinentia pigmenti; infantile phytanic acid storage disease; Infantile Refsum disease; infantile spasms; inflammatory myopathy; intracranial cyst; intracranial hypertension; Joubert syndrome; Keams-Sayre syndrome; Kennedy disease; Kinsboume syndrome; Klippel Feil syndrome; Krabbe disease; Kugelberg-Welander disease; kuru; Lafora disease; Lambert-Eaton myasthenic syndrome; Landau-Kleffner syndrome; lateral medullary (Wallenberg) syndrome; learning disabilities; Leigh's disease; Lennox-Gastaut syndrome; Lesch-Nyhan syndrome; leukodystrophy; Lewy body dementia; lissencephaly; locked-in syndrome; Lou Gehrig's disease (aka motor neuron disease or amyotrophic lateral sclerosis); lumbar disc disease; lyme disease-neurological sequelae; Machado-Joseph disease; macrencephaly; megalencephaly; Melkersson-Rosenthal syndrome; Menieres disease; meningitis; Menkes disease; metachromatic leukodystrophy; microcephaly; migraine; Miller Fisher syndrome; mini-strokes; mitochondrial myopathies; Mobius syndrome; monomelic amyotrophy; mmoottoorr nneeuurroonnee disease; moyamoya disease; mucopolysaccharidoses; multi -infarct dementia; multifocal motor neuropathy; multiple sclerosis and other demyelinating disorders; multiple system atrophy with postural hypotension; muscular dystrophy; myasthenia gravis; myelinoclastic diffuse sclerosis; myoclonic encephalopathy of infants; myoclonus; myopathy; myotonia congenital; narcolepsy; neurofibromatosis; neuroleptic malignant syndrome; neurological manifestations of AIDS; neurological sequelae of lupus; neuromyotonia; neuronal ceroid lipofuscinosis; neuronal migration disorders; Niemann-Pick disease; O'Sullivan-McLeod syndrome; occipital neuralgia; occult spinal dysraphism sequence; Ohtahara syndrome; olivopontocerebellar atrophy; opsoclonus myoclonus; optic neuritis; orthostatic hypotension; overuse syndrome; paresthesia; Parkinson's disease; paramyotonia congenita; paraneoplastic diseases; paroxysmal attacks; Parry Romberg syndrome; Pelizaeus-Merzbacher disease; periodic paralyses; peripheral neuropathy; painful neuropathy and neuropathic pain; persistent vegetative state; pervasive developmental disorders; photic sneeze reflex; phytanic acid storage disease; Pick's disease; pinched nerve; pituitary tumors; polymyositis; porencephaly; Post-Polio syndrome; postherpetic neuralgia (PHN); postinfectious encephalomyelitis; postural hypotension; Prader-Willi syndrome; primary lateral sclerosis; prion diseases; progressive; hemifacial atrophy; progressive multifocal leukoencephalopathy; progressive sclerosing poliodystrophy; progressive supranuclear palsy; pseudotumor cerebri; Ramsay-Hunt syndrome (Type I and Type II); Rasmussen's Encephalitis; reflex sympathetic dystrophy syndrome; Refsum disease; repetitive motion disorders; repetitive ssttrreessss injuries; restless legs syndrome; retrovirus-associated myelopathy; Rett syndrome; Reye's syndrome; Saint Vitus Dance; Sandhoff disease; Schilder's disease; schizencephaly; septo-optic dysplasia; shaken baby syndrome; shingles; Shy-Drager syndrome; Sjogren's syndrome; sleep apnea; Soto's syndrome; spasticity; spina bifida; spinal cord injury; spinal cord tumors; spinal muscular atrophy; stiff-person syndrome; stroke; Sturge-Weber syndrome; subacute sclerosing panencephalitis; subarachnoid hemorrhage; subcortical arteriosclerotic encephalopathy; sydenham chorea; syncope; syringomyelia; tardive dyskinesia; Tay-Sachs disease; temporal arteritis; tethered spinal cord syndrome; Thomsen disease; thoracic outlet syndrome; tic douloureux; Todd's paralysis; Tourette syndrome; transient ischemic attack; transmissible spongiform encephalopathies; transverse myelitis; traumatic brain injury; tremor; trigeminal neuralgia; tropical spastic paraparesis; tuberous sclerosis; vascular dementia (multi-infarct dementia); vasculitis including temporal arteritis; Von Hippel-Lindau Disease (VHL); Wallenberg's syndrome; Werdnig-Hoffman disease; West syndrome; whiplash; Williams syndrome; Wilson's disease; and Zellweger syndrome; and mental health conditions which include, but are not limited to, anxiety disorders (e.g., acute stress disorder, agoraphobia, generalized anxiety disorder, obsessive-compulsive disorder, panic disorder, posttraumatic stress disorder, separation anxiety disorder, social phobia, and specific phobia), childhood disorders, (e.g., attention-deficit/hyperactivity disorder, conduct disorder, and oppositional defiant disorder), eating disorders (e.g., anorexia nervosa and bulimia nervosa), mood disorders (e.g., depression, bipolar disorder I and II, cyclothymic disorder, dysthymic disorder, and major depressive disorder), personality disorders (e.g., antisocial personality disorder, avoidant personality disorder, borderline personality disorder, dependent personality disorder, histrionic personality disorder, narcissistic personality disorder, obsessive-compulsive personality disorder, paranoid personality disorder, schizoid personality disorder, and schizotypal personality disorder), psychotic disorders (e.g., brief psychotic disorder, delusional disorder, schizoaffective disorder, schizophreniform disorder, schizophrenia, and shared psychotic disorder), substance-related disorders (e.g., alcohol dependence or abuse, amphetamine dependence or abuse, cannabis dependence or abuse, cocaine dependence or abuse, hallucinogen dependence or abuse, inhalant dependence or abuse, nicotine dependence or abuse, opioid dependence or abuse, phencyclidine dependence or abuse, and sedative dependence or abuse), adjustment disorders, autism, Asperger's disorder, delirium, dementia, multi -infarct dementia, learning and memory disorders (e.g., amnesia and age-related memory loss), tic disorder and Tourette's disorder. a. Mental Health Conditions
[369] “ “MMeennttaall hheeaalltthh ccoonnddiittiioonnss”” may refer to disease conditions that generally involve negative changes in emotion, mood, thinking, or behavior, and include, but are not limited to, those disclosed herein. Broadly, mental health conditions include those characterized by the DSM-5, Merck Manual, ICD-11, or other such diagnostic resources known to those of skill.
[370] A variety of methods for screening or assessing a subject for a mental health condition exist. In embodiments, a diagnosis of a mental health condition is facilitated with use of the Diagnostic and Statistical Manual of Mental Disorders, such as the DSM-5. In embodiments, diagnosis of a mental health condition is facilitated with use of self-reported or observer-report surveys or questionnaires. Non-limiting examples of such questionnaires include the Patient Health Questionnaire 9 (PHQ-9), the Generalized Anxiety Disorder 7 (GAD-7), PTSD Checklist for DSM-5 (PCL-5), the Alcohol Use Disorders Identification Test (AUDIT), Binge Eating Scale (BES), Obsessive-Compulsive Inventory (OCI), the Personality Disorders Questionnaire (PDQ-IV), Dissociative Experiences Scale (DES), Drug Use Questionnaire (D AST-20), the Mood Disorder Questionnaire (MDQ), and other similar questionnaires. In embodiments, alternative questionnaires, such as the Clinical Global Impression-Improvement scale (CGI-I), may be used to assess improvement of a subject’s mental health state, such as by comparing baseline responses to responses after a treatment intervention. In embodiments, any of the diagnostic manuals and assessments described, and other similar tools, may be used to confirm a reduction in symptoms, a reduction in symptom severity, or elimination of symptoms and/or a previous diagnosis.
[371] In embodiments, the compositions of the invention are useful to treat a patient diagnosed with or diagnosable with at least one mental health condition, or that has any one or more symptoms thereof. In embodiments, a patient diagnosed with a mental health condition is prescribed a therapeutically effective amount of the compositions of the invention, e.g., a prescription is provided for one or more compounds. In embodiments, the compositions are prescribed to a patient diagnosed with a mental health condition as a therapeutic regime, a defined dosing regimen, or a daily dosing regimen, any of which comprising effective amounts of the compositions described herein, e.g., a prescription is provided for one or more dosing method(s). In embodiments, a patient diagnosed with a mental health condition obtains the compositions of the invention without a prescription. In embodiments, a patient diagnosed with a mental health condition obtains the compositions of the invention, and instructions for their use, without a prescription. In embodiments, a patient diagnosed with a mental health condition is administered an effective amount of the compositions of the invention by a clinician. In embodiments, a patient diagnosed with a mental health condition self-administers the compositions of the invention. In embodiments, a patient diagnosed with a mental health condition is supervised by a health professional while self-administering the compositions of the invention while, in other embodiments, the patient is not supervised by a health professional while self-administering the compositions of the invention. “Supervision” and “supervised” will include remote supervision, supervision by electronic means, supervision by patient adherence to a protocol where evidence of such is shared with a health professional, supervision by an individual dispensing one or more compositions by confirming that a patient is allowed to purchase such composition(s), in person supervision during administration itself, confirmation by a patient that said patient is in compliance, and other forms of “supervision” as will be readily appreciated in the art. In embodiments, a patient diagnosed with a mental health condition is administered, either by the patient or a third-party, an effective amount of the compositions of the invention as part of a psychotherapy regimen. In embodiments, a patient diagnosed with a mental health condition is administered, either by the patient or a third-party, an effective amount of the compositions of the invention not part of a psychotherapy regimen, without any accompanying psychotherapy or other therapy, without any supervision or monitoring, or without any intervention by a health care professional. For example, in some embodiments, and depending on the regulatory regime of a jurisdiction and such time as this application may be allowed an in force, the compositions of the invention, including those comprising psilocybin or other psychedelic tryptamines, or Psilocybe extracts, may be obtainable without a prescription, but merely for example by providing proof of the patient’s age, or by taking and passing a “licensing test” (as has been suggested by MAPS), or another method to ensure safe and responsible use. In all of the aforementioned embodiments of this paragraph, an additional and otherwise equivalent embodiment exists and will be readily understood in which the “patient diagnosed with at least one mental health condition” or “patient diagnosed with a mental health condition” is a patient having an undiagnosed mental health condition, or a patient having symptoms of one or more mental health conditions, but not meeting all criteria for its diagnosis, or a patient having no mental health conditions and no symptoms thereof. In embodiments, a patient will take the compositions of the invention for the improvement of mental health and mental functioning, for increased feelings of wellbeing, or for other purposes sometimes referred to broadly as the “betterment of the well.”
[372] In embodiments, the mushroom or cannabis compositions disclosed herein are efficacious in reducing at least one symptom of a mental health condition within a patient diagnosed with the same. As would be apparent to one of skill, symptoms for each mental health condition will be different, however, through medical monitoring (such as monitoring of objective measurements, as described herein), patient reporting (such as, but not limited to through journaling), completion of questionnaires, etc., one will be able to objectively determine if a symptom has reduced in its frequency and/or magnitude.
[373] Further, in some embodiments, the compositions of the invention are shown to be useful in methods for treating a variety of mental health conditions linked to dysregulation or inadequate functioning of the ECS or of neurotransmission in mammals. Included among such disorders are depression including in forms such as drug resistant and treatment resistant depression (TRD) as well as major depressive disorder (MDD), dysthymia, dysthymia, anxiety and phobia disorders (e.g., generalized anxiety or GAD, social anxiety, panic, post-traumatic stress and adjustment disorders), feeding and eating disorders (including binge eating, bulimia, and anorexia nervosa), other binge behaviors, body dysmorphic syndromes, alcoholism, tobacco abuse, drug abuse or dependence disorders including substance use disorders (SUDs) such as opioid use disorders, alcohol use disorder (AUD), cannabis use disorder (CUD), amphetamine and methamphetamine use disorders, caffeine and other stimulant use disorders, prescription drug abuse disorders, disruptive behavior disorders, impulse control disorders, gaming disorders, gambling disorders, pornography and sex addiction, memory loss, dementia of aging, ADHD, personality disorders (e.g., antisocial, avoidant, borderline, histrionic, narcissistic, obsessive compulsive, paranoid, schizoid and schizotypal personality disorders), attachment disorders, autism, and dissociative disorders.
[374] In embodiments, mental health conditions of the invention are specifically the “trauma- and stressor-related disorders,” which include acute stress disorder, adjustment disorders, and PTSD (Merck Manual), as well as reactive attachment disorder, disinhibited social engagement disorder, and others (DSM-5). In other embodiments, the mental health condition is specifically anxiety, depression, or PTSD.
[375] While the neurophysiology underlying mental health conditions may be distinct, an aspect in common of many is the presence of a deleterious, repetitive, and often “rigid” thought process that negatively impacts an individual’s ability to function. For someone with PTSD, for instance, symptoms involve re-experiencing trauma and the feelings associated with it; for depression it can take the form of a recurrent internal editor that attaches negative connotations to normal life events; and for addiction it is the preoccupation with acquiring and using the substance of choice. Thus, in many embodiments, the method of treating a mental health condition involves the treatment of a disorder related to rigid modes of thinking. In different embodiments, the disorder related to rigid modes of thinking can be anxiety, depression, addiction, an eating disorder, obsessive compulsive disorder, or PTSD.
[376] In embodiments, the compositions of the invention are used to reduce the symptoms of a mental health condition. The symptoms of the mental health condition to be treated shall be able to be determined by one of skill, by reference to the general understanding of the art regarding that disorder. Symptoms of PTSD, for example, include transient waking dissociative states in which events are relived as if happening (“flashbacks”), nightmares, distressing and intense memories, other intrusive negative memories, distress or physical reactions after being exposed to triggers, blaming self or others for the trauma, decreased interest in things that were once enjoyable and other feelings of emotional numbness, negative feelings about self and the world, inability to remember the trauma clearly, difficulty feeling positive, feelings of isolation, negative affect, difficulty feeling positive, other negative alterations in cognition and mood, avoidance, aggression or irritability, hypervigilance and hyper-awareness, difficulty concentrating, difficulty sleeping, heightened startle response, engaging in self-destructive, or risky behavior, difficulty sleeping or staying asleep, and suicidal ideation. Accordingly, methods of the invention that reduce the symptoms of PTSD would be understood to reduce any such symptoms. Preferably, where the compositions used are the psilocybin compositions of the invention, such reductions occur without psychedelic effects, and therefore allow for treatment at home or otherwise outside of a clinic and without the need for concomitant psychotherapy. Reductions can also be measured using questionnaires or assessments known in the art, such as for PTSD, CAPS. [377] As will be apparent to one of skill, a plurality of clinical outcome assessments (COAs) are known in the art for disorders characterized by rigid modes of thought. Such are used to measure the efficacy of treatment, and determine the level of symptom reduction in a patient diagnosed with such diseases, non-limiting examples of which include the Addiction Severity Index (ASI), which assesses 30-day and lifetime alcohol and drug use, as well as medical, psychiatric, legal, and family issues; Beck Depression Inventory (BDI), which is a 21 -item measure used to assess depression, involving self-reporting or verbal administration by an administrator, with a possible range of 0-63; the Clinician- Administered PTSD Scale (CAPS), which is a 30-item structured interview administered by a trained individual that corresponds to the DSM-IV criteria for PTSD, and discusses one of the past week, the past month, or the worst month since trauma, with a scale of 0-136; the Davidson Trauma Scale (DTS), which is a 17-item self-report measure that assesses the 17 DSM-IV symptoms of PTSD and rates them based on frequency and severity, having a scale of 0-136; the Global Appraisal of Individual Needs (GAIN), which includes an assessment of background information, substance use, physical health, risk behaviors, and mental health; the Global Assessment of Functioning (GAF), which is a clinician administered scale used to assess the social, occupational, and psychological functioning of adults, with a score range of 0-42; the Hospital Anxiety and Depression Scale (HADS), which is a 14-item self-report measure designed to assess anxiety and depression in non-psychiatric populations, and has a range of 0-42; the Hamilton Anxiety Scale (HAM- A or HAS), which is aa 14-item clinician-administered measure used to assess the severity of anxiety symptoms having a scale of 0-56; the Hamilton Depression Scale (HAM-D) which, similar to HAM-A/HAS, is a clinician-administered scale used to measure the severity of depressive symptoms, having a scale of 0-54; the Impact of Event Scale (IES), which is a 15-item self-reported measure used to assess the frequency with which experiences of “intrusions,” “avoidance,” and emotional numbing related to stressful events occurring within the last week, and has a scale of 0-75; the Maudsley Addiction Profde (MAP), which measures substance use, health risk behavior, physical and psychological health, and social functioning; the Montgomery-Asberg Depression Rating Scale (MADRS), which is a 10-item clincian rated measure that assesses the severity of depression, having a scale of 0-60; the Mississippi Scale for Combat-related PTSD, which is a 35-item self-report questionnaire used to assess DSM-III combat-related PTSD and related features, having a scale of 35-175; the Modified PTSD Symptom Scale, which is a 17-item self-report measure assessing the 17 DSM-III-R symptoms of PTSD, with a scale of 0-68 for intensity and 0-51 for frequency; the Posttraumatic Diagnostic Scale (PTDS or PDS) which is a 49-item self-report measure for severity of PTSD symptoms related to a single identified traumatic event, assessing all DSM-IV criteria in the past month (although the timeframe may be adjusted), having a scale of 0-51; the PTSD Checklist (PCL), which is a 17-item self-report measure of the 17 DSM-IV symptoms of PTSD, having a scale of 17-85; the PTSD Symptom Scale Interview (PSS-I), which is a 17-item semistructured interview that assesses the presence/severity of DSM-IV PTSD symptoms related to single identified traumatic event in individuals with a known trauma history, with a scale of 0-51; the PTSD Symptom Scale Self-report Version (PSS-SR), which is a 17-item self-report scale used to diagnose PTSD according to DSM-III-R criteria, having a scale of 0-51; the Structured interview for PTSD (SI-PTSD or SIP), which assesses the 17 PTSD symptoms as well as survival and behavioral guilt, having a scale of 0-68; the Structured Clinical Interview PTSD Module (SCID), which is a semi-structured interview used to assess the prevalence, absence, and subthreshold presence of PTSD used across trauma populations; the Short PTSD Rating Interview (SPRINT), which is an 8-item self-report measure that assesses the core symptoms of PTSD (intrusion, avoidance, numbing, and arousal) with a scale of 0-32; and the Treatment Outcome Post-Traumatic Stress Disorder Scale (TOP-8), which is an 8-item measure based on all three clusters of posttraumatic stress disorder, having a scale of 0-32 (Forman-Hoffman, Middleton, and Feltner, 2018; Deady, 2009).
[378] In embodiments where an outcome assessment scales such as discussed above is used, a patient may complete a pre-treatment assessment to determine a baseline measure, and then repeat the process at set points during treatment, at the conclusion of treatment, and/or at one or more predetermined periods of time following treatment, to objectively measure the effectiveness of treatment, or one or more aspects of treatment and its efficacy, wherein a decrease or increase in an overall score (e.g., as measured by a test’s “scale”) may determine whether the severity and/or frequency of symptoms has increased, stayed the same, or decreased. As apparent to those of skill, a reduction in score generally indicates symptom improvement, but in some scales, such as GAF, an increase in score indicates improvement. b. Neurodegenerative Conditions [379] Neurodegenerative conditions encompass a wide range of disorders that result from progressive damage to cells and nervous system connections essential for at least one of mobility, coordination, strength, sensation, and cognition; and encompass those disclosed herein. Symptoms of neurodegenerative diseases include any of reductions in mobility and balance, abnormal movements, difficulty swallowing, abnormal bladder and bowel movement function, wide variations in blood pressure, difficulty sleeping, difficulty breathing, abnormal heart function, reduced memory and cognitive abilities, alterations in mood, and changes to speech (UT Southwestern Medical Center, 2022). [380] In embodiments, the compositions of the invention are useful in treating a patient diagnosed with or diagnosable with at least one neurodegenerative condition, or that has any one or more symptoms thereof. In embodiments, a patient diagnosed with at least one neurodegenerative condition is prescribed a therapeutically effective amount of the compositions of the invention. In embodiments, a patient diagnosed with at least one neurodegenerative condition obtains the compositions of the invention without a prescription. In embodiments, a patient diagnosed with at least one neurodegenerative condition is administered an effective amount of the compositions of the invention by a clinician. In embodiments, a patient diagnosed with at least one neurodegenerative condition self-administers the compositions of the invention. In embodiments, a patient diagnosed with at least one neurodegenerative condition is supervised by a health professional while self-administering the compositions of the invention while, in other embodiments, the patient is not supervised by a health professional while self-administering the compositions of the invention. In embodiments, a patient diagnosed with at least one neurodegenerative condition is administered, either by the patient or a third-party, an effective amount of the compositions of the invention as part of a psychotherapy regimen. In embodiments, a patient diagnosed with at least one neurodegenerative condition is administered, either by the patient or a third-party, an effective amount of the compositions of the invention not part of a psychotherapy regimen. In embodiments, the compositions of the invention are efficacious in reducing at least one symptom of a neurodegenerative condition within a patient diagnosed with the same. As would be apparent to one of skill, symptoms for each neurodegenerative condition will be different, however, through medical monitoring (such as monitoring of objective measurements, as described herein), patient reporting (such as, but not limited to through journaling), completion of questionnaires, etc., one will be able to objectively determine if a symptom has reduced in its frequency and/or magnitude. c. General Treatment [381] In addition to treating CNS disorders as discussed herein, the disclosed methods also can be used to improve mental health and improve psychological functioning in non-disease states, such as by reducing neuroticism and psychological defensiveness, increasing creativity and openness to experience, aiding decision-making ability, increasing subjective feelings of wellbeing, wellness, and satisfaction, and increasing the ability to fall or stay asleep. [382] Although in some embodiments, psychotherapy is neither necessitated nor desired, any of the disclosed methods can be used in combination with one or more psychotherapy sessions. Furthermore, a patient can participate in numerous therapeutically beneficial activities, where such participation follows or is in conjunction with the administration of the composition, including but not limited to breathing exercises, meditation and concentration practices, focusing on an object or mantra, listening to music, physical exercise, stretching or bodywork, journaling, grounding techniques, positive self-talk, or engaging with a pet or animal, and it should be understood that such participation can occur with or without the participation or guidance of a therapist or other professional. S. Prophetic Treatment Examples [383] That the compositions of the invention are useful to treat the conditions and disorders discussed herein may be shown in part by the following Examples. While every attempt is made to use the present and future tenses, for avoidance of doubt, these Examples (and any examples discussed elsewhere, even if not so labeled, and unless explicitly stated otherwise) are prophetic examples. It will be readily understood and appreciated that these Examples are merely exemplary and are intended to be illustrative and not to be limiting. EXAMPLE 20: Determination of Therapeutic Efficacy to Treat PTSD and MDD [384] A study is designed to determine the efficacy of the compositions of the invention in the treatment of PTSD and/or major depressive disorder (MDD). The study will include 10 subject (preferably 5 male and 5 female) whose diagnoses for at least one of PTSD and/or MDD will be confirmed prior to the study beginning. [385] The objectives of the study will be to demonstrate the initial subject acceptance of a therapeutic regime of the invention, to measure pre- and post- changes from baseline to final dosage, and to measure brain activity and gain subject reported response during therapy. [386] Inclusion criteria for the subjects include ensuring the subject is at least 21 years old, is diagnosed with at least one of PTSD or MDD, has not experienced satisfactory treatment for such condition(s), and will abstain from using contraindicated drugs during treatment. [387] Regarding the protocol itself, the subject will undergo a dosing regimen comprising 4 continuous days of dosing of a M+ mushroom composition of EXAMPLE 2 followed by 3 continuous days of dosing of a C+ cannabis composition of EXAMPLE 9. In an alternative protocol, the subject will undergo a dosing regimen comprising 4 continuous days of dosing of a M+ mushroom composition of EXAMPLE 2 followed by 3 continuous days of dosing of a C– cannabis composition of EXAMPLE 9. In an alternative protocol, the subject will undergo a dosing regimen comprising 4 continuous days of dosing of a M– mushroom composition of EXAMPLE 2 followed by 3 continuous days of dosing of a C+ cannabis composition of EXAMPLE 9. In an alternative protocol, the subject will undergo a dosing regimen comprising 4 continuous days of dosing of a M– mushroom composition of EXAMPLE 2 followed by 3 continuous days of dosing of a C– cannabis composition of EXAMPLE 9. In an alternative protocol, the subject will undergo a dosing regimen comprising 4 continuous days of dosing of a M+ mushroom composition of EXAMPLE 2 followed by 3 continuous days of abstaining (washout). In an alternative protocol, the subject will undergo a dosing regimen comprising 4 continuous days of dosing of a M– mushroom composition of EXAMPLE 2 followed by 3 continuous days of abstaining (washout). It will be readily appreciated that the alternative protocols also may be used as additional treatment arms, to compare the compositions and dosing regimens of the invention against each other. [388] In one embodiment, the first four days of dosing a M+ composition are done in a controlled setting with initial education prior to the first dose, to ensure a lack of psychedelic effects by the subject, and sufficient comfort by the subject with microdosing psychedelics. [389] After the first dose, subjects may be assessed by a physician to gauge the intensity of any observed side effects. Patient reported outcomes are then performed prior to each dosing day. At the end of the first four days, the patient is evaluated regarding the need for continued controlled dosing. If it is determined that self-dosing is safe and may be effective, an ongoing symptom and outcome record and separate liability form must be completed. [390] In embodiments, and depending on the regulatory regime, if self-dosing is conducted by a patient, only four doses of the mushroom composition may be dispensed at a time. A medication compliance form may be filled out and collected prior to dispensing another dosing regimen’s worth (e.g., four days’ supply) of the mushroom composition. [391] In total, the treatment period is 8 weeks, or about 60 days, after which the evaluation and testing will be completed. Results will indicate that at least one symptom of PTSD and/or MDD are lessened, reduced in frequency, or otherwise attenuated by the compositions of the invention. Results will be compiled for reporting to Health Canada to build a dossier for Phase 0 and Phase I trials, and IND (investigational new drug) guidance. EXAMPLE 19: Determination of Therapeutic Efficacy for Other CNS Disorders [392] A study is designed to determine the efficacy of the compositions of the invention in the treatment of other CNS disorders besides PTSD and MDD, including any one or more of adjustment disorder, affective disorder, depression, atypical depression, postpartum depression, catatonic depression, a depressive disorder due to a medical condition, premenstrual dysphoric disorder, seasonal affective disorder, dysthymia, anxiety, phobia disorders, binge disorders, body dysmorphic disorder, alcohol or drug abuse or dependence disorders, substance-related disorders, substance use disorders, alcohol use disorder, substance-induced mood disorder, a mood disorder related to another health condition, disruptive behavior disorders, eating disorders, impulse control disorders, obsessive compulsive disorder (OCD), attention deficit hyperactivity disorder (ADHD), personality disorders, attachment disorders, and dissociative disorders; and neurodegenerative conditions, such as but not limited to Alzheimer’s disease, ataxia, Huntington’s disease, Parkinson’s disease, motor neuron disease, multiple system atrophy, progressive supranuclear palsy, migraines, cluster headaches, short-lasting unilateral neuralgiform headaches, fibromyalgia, traumatic brain injury, and mild-traumatic brain injury (mTBI) . [393] Following the outline of the Example above with regard to PTSD and MDD, the subject receives treatment in total for a period of 8 weeks, or about 60 days, after which evaluation and testing will be completed. Results will indicate that at least one symptom of the CNS disorder are lessened, reduced in frequency, or otherwise attenuated by the compositions of the invention. Results will be compiled for reporting to Health Canada to build a dossier for Phase 0 and Phase I trials, and IND guidance. EXAMPLE 20: Use as a Medication [394] The compounds described herein are prescribed in a therapeutically effective amount to a patient suffering from at least one CNS disorder, including mental health conditions such as post-traumatic stress disorder (PTSD), adjustment disorder, affective disorder, depression, atypical depression, postpartum depression, catatonic depression, a depressive disorder due to a medical condition, premenstrual dysphoric disorder, seasonal affective disorder, dysthymia, anxiety, phobia disorders, binge disorders, body dysmorphic disorder, alcohol or drug abuse or dependence disorders, substance-related disorders, substance use disorders, alcohol use disorder, substance-induced mood disorder, a mood disorder related to another health condition, disruptive behavior disorders, eating disorders, impulse control disorders, obsessive compulsive disorder (OCD), attention deficit hyperactivity disorder (ADHD), personality disorders, attachment disorders, and dissociative disorders; and neurodegenerative conditions, such as but not limited to Alzheimer’s disease, ataxia, Huntington’s disease, Parkinson’s disease, motor neuron disease, multiple system atrophy, progressive supranuclear palsy, migraines, cluster headaches, short-lasting unilateral neuralgiform headaches, fibromyalgia, traumatic brain injury, and mild-traumatic brain injury (mTBI) . [395] In embodiments, prior to being prescribed the compositions of the invention, a patient is evaluated to determine whether the compositions are appropriate for the patient. An evaluation may include completing questionnaires, obtaining objective health measurements from the patient, such as one or more of weight, body temperature, heart rate, respiratory rate, blood oxygenation, BP and its variables (e.g., SBP, SBP, MAP, and PP); CNIBP; ECG measurements (e.g., RR interval or its variability), QT interval or its variability, HRV (including measured by devices other than an ECG); hemodynamic response; levels of glucose, cortisol, serotonin, dopamine, or cholesterol; EEG measures; BDNF; genetic markers including relating to CYP enzymes or drug metabolism; genetic variation in mGluR5 or FKBP5; other biomarkers; and digital biomarkers, including digital biomarkers associated with any one or more of the foregoing. If one or more objective measurements obtained from the patient are within established safety standards known to those of skill, the patient may be prescribed the compositions of the invention, and the type or amount or other characteristics of the compositions of the invention may be modified thereby, for example a prescribed dose will be appropriate for the age and weight of the patient, and may additionally take into consideration other factors known to those of skill, such as comorbidities, current medications (if any), and metabolic or genetic variations among patient populations. [396] To confirm a reduction in symptoms, a reduction in symptom severity, or elimination of symptoms and/or a previous diagnosis, one or more diagnostic or clinical tools is used, such as the DSM–5, one or more self-reported or observer-report surveys or questionnaires, and any one or more of the Patient Health Questionnaire 9 (PHQ-9), the Hamilton Depression Rating Scale (HAM–D); the Generalized Anxiety Disorder 7 (GAD-7), PTSD Checklist for DSM–5 (PCL-5), The Alcohol Use Disorders Identification Test (AUDIT), Binge Eating Scale (BES), Obsessive-Compulsive Inventory (OCI), the Personality Disorders Questionnaire (PDQ-IV), Dissociative Experiences Scale (DES), Drug Use Questionnaire (DAST-20), the Mood Disorder Questionnaire (MDQ), and one or more other similar questionnaires, which may be chosen based on the condition or disorder to be treated, as known in the art. By comparing baseline responses to responses after treatment, the methods and compositions of the invention are shown to be effective to treat the conditions herein. EXAMPLE 21: Dosing Regimens for the Compositions of the Invention [397] Exemplary and non-limiting dosing regimens for the compositions of the invention, with M+, M–, C+, C–, and A as herein defined are listed below. It will be understood that a “therapeutic regime” will comprise one or more “defined dosing regimens” and/or “daily dosing regimens” as listed in the Examples below, and that such therapeutic regimes will therefore comprise the treatment administrations or days from 1-11 as shown (or any smaller number, e.g., from 1-7, 1-8, 1-9, or 1-10), repeating any number of times of at least one, and including in some embodiments one or more defined dosing regimens, two or more defined dosing regimens, three or more defined dosing regimens, four or more defined dosing regimens, five or more defined dosing regimens, six or more defined dosing regimens, seven or more defined dosing regimens, eight or more defined dosing regimens, nine or more defined dosing regimens, 10 or more defined dosing regimens, 11 or more defined dosing regimens, 12 or more defined dosing regimens, or greater than 12 defined dosing regimens, which together will comprise a single “therapeutic regime” of the invention. In other embodiments, a therapeutic regime will comprise, together, one or more daily dosing regimens, two or more daily dosing regimens, three or more daily dosing regimens, four or more daily dosing regimens, five or more daily dosing regimens, six or more daily dosing regimens, seven or more daily dosing regimens, eight or more daily dosing regimens, nine or more daily dosing regimens, 10 or more daily dosing regimens, 11 or more daily dosing regimens, 12 or more daily dosing regimens, or greater than 12 daily dosing regimens. [398] Where an Example provides for less than 11 total administrations or days (as indicated by blank treatment administrations/days in the below) the therapeutic regime will repeat following the last administration period (“period” including days, and also including other periods of shorter or longer duration) or day shown. For instance, a defined dosing regimen of Examples 22-55, each lasting seven administrations (or “periods”), will repeat every seven administrations, with each seven administrations repeating as below, or of Examples 56-71, each lasting eight administrations, will repeat every eight administrations, with each eight administrations repeating as below. In embodiments with a daily dosing regimen of Examples 22-55, each will be understood to last seven days, and each will repeat weekly, with each week repeating as below; or of Examples 56-71, each lasting eight days, and repeating every eight days, with each eight days repeating as below.
[399] In embodiments, a therapeutic regime may be a “combination therapeutic regime,” wherein it comprises multiple defined dosing regimens and/or daily dosing regimens from different examples. For example, in some embodiments it can combine multiple defined dosing regimens from different examples (a “combination defined dosing regimen”), rather than a single defined dosing regimen, repeated a specific number of times (a “repeated defined dosing regimen”). Such combination therapeutic regimes may comprise two or more different defined dosing regimens, including three or more different defined dosing regimens, four or more different defined dosing regimens, five or more different defined dosing regimens, six or more different defined dosing regimens, seven or more different defined dosing regimens, eight or more different defined dosing regimens, nine or more different defined dosing regimens, 10 or more different defined dosing regimens, 11 or more different defined dosing regimens, 12 or more different defined dosing regimens, or greater than 12 different defined dosing regimens, each following the last, end to end, comprising a combination therapeutic regimen.
[400] In other embodiments, a therapeutic regime can combine daily dosing regimens from different examples (a “combination daily dosing regimen”), rather than a single daily dosing regimen, repeated a specific number of times (a “repeated daily dosing regimen”). Such combination therapeutic regimes may comprise two or more different daily dosing regimens, including three or more different daily dosing regimens, four or more different daily dosing regimens, five or more different daily dosing regimens, six or more different daily dosing regimens, seven or more different daily dosing regimens, eight or more different daily dosing regimens, nine or more different daily dosing regimens, 10 or more different daily dosing regimens, 11 or more different daily dosing regimens, 12 or more different daily dosing regimens, or greater than 12 different daily dosing regimens, each following the last, end to end, comprising a combination therapeutic regimen. [401] In yet other embodiments, a therapeutic regime can combine both defined dosing regimens and daily dosing regimens from different examples (a “combination dosing regimen”), rather than a single distinct or daily dosing regimen, repeated a specific number of times (a “repeated dosing regimen”). Such combination therapeutic regimes may comprise two or more different distinct and daily dosing regimens, including three or more different distinct and daily dosing regimens, four or more different distinct and daily dosing regimens, five or more different distinct and daily dosing regimens, six or more different distinct and daily dosing regimens, seven or more different distinct and daily dosing regimens, eight or more different distinct and daily dosing regimens, nine or more different distinct and daily dosing regimens, 10 or more different distinct and daily dosing regimens, 11 or more different distinct and daily dosing regimens, 12 or more different distinct and daily dosing regimens, or greater than 12 different distinct and daily dosing regimens, each following the last, end to end, comprising a combination therapeutic regimen. [402] In embodiments, a combination therapeutic regime will include an “all plus” distinct and/or daily dosing regimen (e.g., M+ and C+), followed by an “all minus” distinct and/or daily dosing regimen (e.g., M– and C–), such as Example 34 followed by Example 37, the two in alternating fashion. For avoidance of doubt, it should be understood that any single treatment administration and/or day may be independently selected; that is, as an example, where a defined dosing regimen or a daily dosing regimen comprises more than one “M+”, each M+ may be an independently selected M+ mushroom composition.
Figure imgf000137_0001
Figure imgf000138_0001
Figure imgf000139_0001
Figure imgf000140_0001
Figure imgf000141_0001
Figure imgf000142_0001
Figure imgf000143_0001
[403] EXAMPLES 22-205 include, in certain exemplary embodiments, the administration of a psychedelic mushroom composition (e.g., comprising psilocybin and/or a Psilocybe extract) ( M+ ), followed or preceded by the administration of at least one of a functional mushroom composition (e.g., comprising no psilocybin and no Psilocybe extract) ( M– ), an intoxicating cannabis composition (e.g., comprising THC) ( C+ ), or a non-intoxicating cannabis composition (e.g., comprising no THC) ( C– ); alternatively, as illustrated in certain of EXAMPLES 22-205 , administration of the M+ mushroom composition may be preceded and/or followed by abstinence or washout ( A ). [404] In embodiments of EXAMPLES 22-205 , the M+ mushroom composition is a composition of EXAMPLE 2 or 3. In embodiments of EXAMPLES 22-205 , which may or may not be the same embodiments, the (C+) cannabis composition is a composition of EXAMPLE 9. In embodiments of EXAMPLES 22-205 , the M– mushroom composition is a composition of EXAMPLE 2 or 3 , but without a Psilocybe extract. In embodiments of EXAMPLES 22-205 , which may or may not be the same embodiments, the (C–) cannabis composition is a composition of EXAMPLE 9 , but without THC. [405] In embodiments, regardless of whether the M+ mushroom composition is the formulation of EXAMPLE 2 or 3 , and/or the C+ cannabis composition is the composition of EXAMPLE 9 , the dosing regimen illustrated in each of EXAMPLES 22-205 will be repeated and continue for a specified or predetermined duration of time, such duration of time being the therapeutic regime. In embodiments, the specified duration of time is between about 15 to about 90 days. In embodiments, the specified duration of time is between about 30 to about 75 days. In embodiments, the specified duration of time is between about 50-65 days. In embodiments, the specified duration of time is 60 days. In embodiments, the specified duration of time is 45 days. In embodiments, the specified duration of time is 30 days. In embodiments, the specified duration of time is a period of weeks, such as two weeks, three weeks, four weeks, five weeks, six weeks, seven weeks, eight weeks, nine weeks, 10 weeks, 11 weeks, 12 weeks, or more than 12 weeks. In embodiments, the specified duration of time is a specific number of repetitions of the defined dosing regimen(s), the daily dosing regimen(s), or the combination of dosing regimens. [406] In embodiments, the therapeutic regime lasts for a specified duration of between about 15 to about 90 days, inclusive; however, the pattern illustrated in EXAMPLES 22-205 does not continue for the entirety of the specified duration. Instead, in some embodiments, the first 11 treatment administrations or days are illustrated in one of EXAMPLES 22-205 , but after the initial 11 administrations or days, the treatment schedule is fluid, and is instead determined by one, or a combination of, objective health measurements, as disclosed herein; or subjective patient data, obtained through any of a questionnaire, interview, and journaling. [407] In such an embodiment, the subsequent portion of the therapeutic regime may include set administration or dosing days (e.g., administration of a M+ mushroom composition, that may or may not be the formulation of EXAMPLE 2 or 3 , for four consecutive administrations or days), but may then be “modular,” such that other administrations or days may comprise any of abstinence (A), or administration of at least one of a M+ mushroom composition, a C+ cannabis composition, or a C– cannabis composition. In such embodiments, the patient, a clinician, or both, may determine which compositions(s) is(are) administered, or if abstinence will occur, and if the A day will include another composition comprising additional active agents, such as vitamins, nootropics, and/or supplements. [408] Likewise, in some embodiments, after the initial 11 administrations or days illustrated in EXAMPLES 22-205 , the M+ dosing days also may be modular, such that, preceding or following an M+ dosing day, a patient may be administered, by a patient or a third party, a M– mushroom composition, a C+ or C– cannabis composition, or may have a washout. [409] It will be readily appreciated that EXAMPLES 22-205 are merely illustrative examples, and should not be construed as limiting. As a result, in some embodiments, a patient may be prescribed a treatment regimen of a specified duration of between about 15 to about 90 days, inclusive, wherein on any given administration period or day, a patient may be administered one of, or a combination thereof, a M+ or M– mushroom composition, a C+ or C– cannabis composition, or may have a washout, which includes administration of a composition of additional active agents. Furthermore, in some embodiments, a patient will not be prescribed a therapeutic regime with a predetermined length or number of dosing regimens, such that therapeutic regime may be as short or as long in duration as necessary, and may be determined by the patient, a clinician prescribing the treatment, or a combination. [410] In embodiments, regardless of whether the M+ mushroom composition is a formulation of EXAMPLE 2 or 3 , and/or the C+ cannabis composition is a composition of EXAMPLE 9 , the formulations detailed in EXAMPLES 22-205 are packaged as distinct dosing kits or daily dosing kits. In embodiments, the distinct dosing kits may be kits that contain 7, 11, 14, 21, 28, 29, 30, 31, 45, 50, 60, 75, or 90 administrations-worth of the mushroom and/or cannabis compositions described herein, optionally including “A” administration compositions of additional active agents. In embodiments, the daily dosing kits may be kits that contain 7, 11, 14, 21, 28, 29, 30, 31, 45, 50, 60, 75, or 90 days-worth of the mushroom and/or cannabis compositions described herein, optionally including “A” day compositions of additional active agents. In other embodiments, the number of administrations or days in a kit will be any number from 2 to 90 or greater than 90. [411] In some preferred embodiments, the daily dosing kits will include a M+ mushroom composition of either EXAMPLE 2 or 3 according to a set schedule, and will include one, two, three, or all of a C+ cannabis composition, a C– cannabis composition, an M– mushroom composition, and an A composition of additional active agents, for each administration or day not scheduled to be an administration or day wherein an M+ mushroom composition is to be administered (such as that illustrated in FIG.3 ). In such an embodiment, on administrations or days wherein the patient is not being administered an M+ composition, the patient may be administered or may self-administer one, two, three, or all of a C+, C–, M–, or A composition as described herein. In such embodiments, as disclosed supra , such decisions may be made by a patient, a clinician or health professional, or them together. EXAMPLE 206: Exemplary Ethanol Psilocybin Extraction System [412] In embodiments, an M+ composition comprises a Psilocybe spp. extract, or a like “psychedelic mushroom extract” from another genera, such as Conocybe, Copelandia, Galerina, Gymnopilus, Inocybe, Mycena, Panaeolina, Panaeolus, Pholiotina, or Pluteus . In this exemplary extraction system, which will produce such an extract, 1 kg psilocybin fungal material is first obtained as fruiting bodies, truffles, and/or mycelium, and then sufficiently dried in an air drier so as to remove moisture, but not damage the fungal material. The sufficiently dried fungal material is then placed in an industrial processor having a sufficient number of blades to facilitate the fine chopping thereof (such as a BSC coarse crusher). The finely-chopped material is then placed in an industrial pulverizer sufficient to reduce the fungal material into a sufficiently fine powder (such as a BSP pulverizer). Once the fungal material is reduced to a fine powder, it is transferred to a vat. An ethanol solvent (e.g., of at least 99%) is then introduced in a volume sufficient to fully immerse the dried material, transforming the mixture into a homogeneous slurry. To further facilitate extraction, the entire mixture is then agitated within the vat, ensuring constant exchange between the fungal material and the ethanol solvent. After roughly 24 hours, the agitation ceases, and the slurry is transferred to a filtration device of sufficient porosity to separate the fungal material from the alcohol solution. The alcohol solution is then evaporated under ambient air conditions, via fans blowing ambient air over the solution to facilitate evaporation, or evaporated under heat and vacuum pressure. In alternative embodiments, the solution may be spray dried using methods and techniques generally known in the art. After sufficient evaporation, the extraction may be standardized, or may be tested to ensure it is at an appropriate concentration to be utilized in the M+ mushroom compositions of the invention. EXAMPLE 206: Exemplary Non-Inferiority Trial of Formulations and Methods [413] Non-inferiority trials are performed to demonstrate that the compositions and methods herein perform no worse than at least one comparator composition or method, by more than a pre-specified amount. Such trials show that the compositions and methods of the invention are at least as useful as for treatment of a specified indication and have approximately the same efficacy as the comparator(s). In embodiments, the compositions and methods of the invention are shown to offer other benefits such as a better safety profile. EXAMPLE 207: Exemplary Equivalence Trial of Formulations and Methods [414] Equivalence trials are performed to demonstrate that the compositions and methods herein perform at least as good as at least one comparator composition or method, within a pre-specified range. Such trials show that the compositions and methods of the invention are at least as useful for treatment of a specified indication and are at least as effective as the comparator(s). In embodiments, the compositions and methods of the invention are shown to offer other benefits such as a better safety profile. EXAMPLE 208: Exemplary Superiority Trial of Formulations and Methods [415] Superiority trials are performed to demonstrate that the compositions and methods herein perform better than at least one comparator composition or method, by more than a pre-specified amount. Such trials show that the compositions and methods of the invention are more useful for treatment of a specified indication and have better efficacy than the comparator(s). In embodiments, the compositions and methods of the invention are shown to offer other benefits such as a better safety profile. EXAMPLE 209: Exemplary Study to Illustrate Synergistic Properties [416] A study is designed to determine the presence and magnitude of synergistic properties within the entourage enhanced compositions of the invention. The study includes a population of 90 individuals (45 men and 45 women) diagnosed with at least one CNS disorder, as disclosed above, who are each administered the compositions as part of a therapeutic treatment regimen that lasts a total of 60 days, comprising the administration of one composition for four consecutive days, followed by the administration of a separate composition for three consecutive days. Prior to commencing the dosing regimen, each participant’s symptoms, and the severity thereof, are determined to obtain a baseline. Methods of measuring such symptoms are as discussed herein or known in the art, and may include participation in a clinical outcome assessment specific to that disorder. [417] Of the study population, the participants are split into nine groups of 10 each (5 men, 5 women), with each group being assigned a separate dosing regimen: 1. Group 1 : An entourage enhanced mushroom composition of the invention, paired with an entourage enhanced cannabis composition of the invention. 2. Group 2 : An entourage enhanced mushroom composition of the invention, paired with a cannabis composition that is not entourage enhanced 3. Group 3 : A mushroom composition that is not entourage enhanced, with an entourage enhanced cannabis composition of the invention. 4. Group 4 : A mushroom composition that is not entourage enhanced, with a cannabis composition that is not entourage enhanced. 5. Group 5 : A placebo, with an entourage enhanced cannabis composition of the invention. 6. Group 6 : An entourage enhanced mushroom composition of the invention, with a placebo. 7. Group 7 : A placebo, with a cannabis composition that is not entourage enhanced. 8. Group 8 : A mushroom composition that is not entourage enhanced, with a placebo. 9. Group 9 : A placebo, with a placebo. [418] At the end of the 60-day period, the same method used to determine the symptoms and their severity is again completed by each patient. It will be discovered that those patients taking at least one entourage enhanced composition of the invention have improvements to at least one symptom of their diagnosed condition that are more than one would expect from administration of the individual compounds themselves. Moreover, those in Group 1 , who are administered the entourage enhanced mushroom composition and entourage enhanced cannabis composition of the invention, will have improvements to at least one symptom of their diagnosed condition that are more significant than one would expect or find from administration of the individual entourage enhanced compositions by themselves. This will be further supported by analysis of the groups comparatively, as Group 1 will see the greatest improvements, followed by Groups 2 and 3 , and Groups 5 and 6. [419] The foregoing description, for purposes of explanation, uses specific nomenclature to provide a thorough understanding of the invention. However, it will be apparent to one skilled in the art that specific details are not required in order to practice the invention. Thus, the foregoing description of specific embodiments of the invention is presented for purposes of illustration and description. It is not intended to be exhaustive or to limit the invention to the precise forms disclosed; of course, many modifications and variations are possible in view of the above teachings. The embodiments were chosen and described in order to best explain certain key principles of the invention and its practical applications, through the elucidation of specific examples, and to thereby enable others skilled in the art to best make and utilize the invention and various embodiments with various modifications as are suited to the particular use contemplated, even when such uses are beyond the specific examples disclosed. Accordingly, the scope of the invention shall be defined solely by the following claims and their equivalents. REFERENCES 1. APA 2013. Diagnostic and statistical manual of mental disorders: DSM–5. Washington D.C.: American Psychiatric Association. 2. Ben-Shabat et al., An entourage effect: inactive endogenous fatty acid glycerol esters enhance 2-arachidonoyl-glycerol cannabinoid activity, Eur. J. Pharmacol.353(1):23-31 (1998). 3. Berge et al. (1977) “Pharmaceutical Salts,” J. Pharm. Sci.66:1-19. 4. Bouso et al. Measuring the subjective: revisiting the psychometric properties of three rating scales that assess the acute effects of hallucinogens. Hum Psychopharmacol.2016 Sep.31(5):356-72). 5. Breuer et al., Fluorinated Cannabidiol Derivatives: Enhancement of Activity in Mice Models, PLOS ONE, 2016. 6. Brewerton T. D. (1995). Toward a unified theory of serotonin dysregulation in eating and related disorders. Psychoneuroendocrinology, 20 (6), 561–590. 7. Carhart-Harris, R. L.; Goodwin, G. M. (2017) The Therapeutic Potential of Psychedelic Drugs: Past, Present, and Future, Neuropsychopharmacol., 42, 2105-13. 8. Castillo-Carniglia, Alvaro et al., Psychiatric comorbidities in alcohol use disorder, The Lancet Psychiatry, Vol.6, Issue 12, 1068-1080. 9. Center for Food Safety and Applied Nutrition. (2019, December 16). New dietary ingredients (NDI) notification process. U.S. FDA. Retrieved March 10, 2022, from www.fda.gov/food/dietary-supplements/new-dietary-ingredients-ndi-notification-process 10. Cogan, The ‘entourage effect’ or ‘hodge-podge hashish’: the questionable rebranding, marketing, and expectations of cannabis polypharmacy, Expert Rev. Clin. Pharmacol., 1-11 (2020). 11. Deady, M. (2009). A Review of Screening, Assessment & Outcome Measures for Drug & Alcohol Settings . Network of Alohol. & Other Drug Agencies (NADA). Retrieved March 2, 2022. 12. Forman-Hoffman V, Middleton JC, Feltner C, et al. Psychological and Pharmacological Treatments for Adults With PTSD: A Systematic Review Update [Internet]. Rockville (MD): Agency for Healthcare Research and Quality (US); 2018 May. (Comparative Effectiveness Review, No.207.) Appendix B, Outcome Measures and Instruments. 13. Fowler, Oleamide: a member of the endocannabinoid family?, Br. J. Pharmacol., 141(2):195-96 (2004)). 14. Fricke J. Enzymatic Synthesis of Psilocybin. Angew Chem Int Ed Engl.2017 Sep 25;56(40):12352-55. 15. Green (1996) The Trauma History Questionnaire (THQ). 16. Green B. (2014). Prazosin in the treatment of PTSD. J psych practice, 20 (4), 253–259. 17. Greer et al., PTSD symptom reports of patients evaluated for the New Mexico Medical Cannabis Program, J. Psychoactive Drugs, 46:73-77 (2014)). 18. Harli, M. (2022, January 14). Cold ethanol extraction of cannabis: How it works. Prospiant. Retrieved March 10, 2022, from https://www.prospiant.com/cold-ethanol-extraction/ 19. Huang et al. (2019) Isobologram Analysis: A Comprehensive Review of Methodology and Current Research, Front. Pharmacol.10:1222. doi:10.3389/fphar.2019.01222. 20. Hutcheson et al. (2011). Serotonin receptors and heart valve disease—it was meant 2B. Pharmacol. Therapeutics, 132(2), 146-57. 21. Jackobsen et al., SSRIs versus placebo in patients with major depressive disorder. A systematic review, BMC Psychiatry (2017)17:58. 22. Kawagishi, H., Ando, M., Mizuno, T. (1990). Hericenone A and B as cytotoxic principles from the mushroom. Tetrahedron Letters, 31(3), 373–376. 23. Kobayashi, S., Tamanoi, H., Hasegawa, Y., Segawa, Y., & Masuyama, A. (2014). Divergent synthesis of bioactive resorcinols isolated from the fruiting bodies of Hericium erinaceum: total syntheses of hericenones A, B, and I, hericenols B-D, and erinacerins A and B. The Journal of organic chemistry, 79(11), 5227–5238. 24. Laprairie et al. (2015) Cannabidiol is a negative allosteric modulator of the cannabinoid CB1 receptor, British J. Pharmacol., 172(20):4790-4805. 25. Miller, L. W. (1933). The Genera of Hydnaceae. Mycologia, 25 (4), 286–302. 26. Morean, M. E., de Wit, H., King, A. C., Sofuoglu, M., Rueger, S. Y., & O'Malley, S. S. (2013). The drug effects questionnaire: psychometric support across three drug types. Psychopharmacology, 227 (1), 177–192. 27. Nichols, David E. and Frescas, Stewart Synthesis No.6, 935-938 (1999). 28. Pinna, Biomarkers for PTSD at the Interface of the Endocannabinoid and Neurosteroid Axis, Front. Neurosci., 12:482 (2018)). 29. Radwan, M. M., Chandra, S., Gul, S., & ElSohly, M. A. (2021). Cannabinoids, Phenolics, Terpenes and Alkaloids of Cannabis. Molecules , 26 (9), 2774. 30. Richard Finkel; Michelle Alexia Clark; Pamela C. Champe; Luigi X. Cubeddu (16 July 2008). Lippincott's Illustrated Reviews: Pharmacology. Lippincott Williams & Wilkins. p.141. ISBN 978-0-7817-7155-9. Retrieved 12 May 2012. 31. Russo, Taming THC: potential cannabis synergy and phytocannabinoid-terpenoid entourage effects, British J. of Pharm., 163:1344-64 (2011). 32. Ryu, S. H., Hong, S. M., Khan, Z., Lee, S. K., Vishwanath, M., Turk, A., Yeon, S. W., Jo, Y. H., Lee, D. H., Lee, J. K., Hwang, B. Y., Jung, J. K., Kim, S. Y., & Lee, M. K. (2021). Neurotrophic isoindolinones from the fruiting bodies of Hericium erinaceus. Bioorganic & medicinal chemistry letters, 31, 127714. 33. Scott, E. (February 12, 2019). U.S. Pat. No.10,201,577. “Compositions and methods for enhancing working and long-term memory support using new combination of ginkgo biloba and panax ginseng and associated components” 34. Sherwood, A. M., Halberstadt, A. L., Klein, A. K., McCorvy, J. D., Kaylo, K. W., Kargbo, R. B., &amp; Meisenheimer, P. (2020). Synthesis and biological evaluation of tryptamines found in hallucinogenic mushrooms: Norbaeocystin, Baeocystin, norpsilocin, and aeruginascin. Journal of Natural Products, 83(2), 461–467. 35. Sornchaithawatwong, C., Kunthakudee, N., Sunsandee, N., &amp; Ramakul, P. (2020). Selective extraction of Cordycepin from cordyceps militaris – optimisation, kinetics and Equilibrium Studies. Indian Chemical Engineer, 1–13. 36. Spitzer R.L., Kroenke K., Williams J.B., Löwe B. (2006) A brief measure for assessing generalized anxiety disorder: the GAD-7. Arch intern med.22;166(10):1092-7. 37. Studerus, E., Gamma, A., & Vollenweider, F. X. (2010). Psychometric evaluation of the altered states of consciousness rating scale (OAV). PloS one, 5(8), e12412. https://doi.org/10.1371/journal.pone.0012412 38. Tuli, H. S., Sandhu, S. S., & Sharma, A. K. (2014). Pharmacological and therapeutic potential of Cordyceps with special reference to Cordycepin.3 Biotech, 4(1), 1–12. 39. Wilkinson et al., Impact of cannabis use on the development of psychotic disorders, Curr. Addict. Rep.1:115-28 (2014). 40. World Health Organization. (2019). ICD-11: International classification of diseases (11th revision). Retrieved from https://icd.who.int/

Claims

CLAIMS What is claimed is: 1. A method of treating a CNS disorder patient in need thereof, comprising administering to the patient a mushroom composition and a second composition according to a defined dosing regimen.
2. The method of claim 1, wherein the defined dosing regimen is a daily dosing regimen consisting of a first number of days of administration of the mushroom composition, followed by a second number of days of administration of the second composition.
3. The method of claim 2, wherein the first number of days is between 2 and 7 days.
4. The method of claim 3, wherein the first number of days is between 2 and 5 days.
5. The method of claim 4, wherein the first number of days is either 3 or 4 days.
6. The method of claim 5, wherein the first number of days is 4 days.
7. The method of claim 2, wherein the second number of days is between 2 and 7 days.
8. The method of claim 7, wherein the second number of days is between 2 and 5 days.
9. The method of claim 8, wherein the second number of days is either 3 or 4 days.
10. The method of claim 9, wherein the second number of days is 3 days.
11. The method of any of claims 1-10, wherein the mushroom composition is a psychedelic mushroom composition.
12. The method of claim 6, wherein the mushroom composition is a psychedelic mushroom composition.
13. The method of claim 12, wherein the psychedelic mushroom composition comprises a psychedelic mushroom extract or a compound from a psychedelic mushroom.
14. The method of claim 13, wherein the psychedelic mushroom extract is a Psilocybe extract.
15. The method of claim 14, wherein the Psilocybe extract is an extract of Psilocybe cubensis .
16. The method of claim 13, wherein the psychedelic mushroom extract is an extract of a species within a fungal genera selected from the group consisting of Conocybe, Copelandia, Galerina, Gymnopilus, Inocybe, Mycena, Panaeolina, Panaeolus, Pholiotina , and Pluteus .
17. The method of claim 13, wherein the compound from a psychedelic mushroom is any of psilocybin, psilocin, norbaeocystin, baeocystin, aeruginascin, and norpsilocin.
18. The method of claim 17, wherein the compound from a psychedelic mushroom is produced by chemical synthesis or biosynthesis.
19. The method of any of claims 1-10, wherein the mushroom composition is a functional mushroom composition.
20. The method of claim 6, wherein the mushroom composition is a functional mushroom composition.
21. The method of any of claims 1-10, wherein the second composition is a functional mushroom composition.
22. The method of claim 10, wherein the second composition is a functional mushroom composition.
23. The method of either of claim 20 or 22, wherein the functional mushroom comprises a Hericium extract or a compound from Hericium .
24. The method of claim 23, wherein the compound from Hericium is any of any of Hericenone A, Hericenone B, Hericenone C, Hericenone D, Hericenone E, Hericenone F, Hericenone G, Hericenone H, Hericenone I, Hericenone J, Hericenone K, 3HF, DLPE, Isohericerinol A, Hericerin, NDPIH, and Corallocin A.
25. The method of either of claim 20 or 22, wherein the functional mushroom comprises a Cordyceps extract or a compound from Cordyceps .
26. The method of claim 25, wherein the compound from Cordyceps is any of cordycepin, cordycepic acid, N-acetylgalactosamine, adenosine, ergosterol, an ergosteryl ester, a bioxanthracene, hypoxanthine, a macrolide, a cicadapeptin, myriocin, superoxide dismutase, naphthoquinone, cordyheptapeptide, dipicolinic acid, a fibrinolytic enzyme, and cordymin.
27. The method of any of claims 1-10, wherein the second composition is a cannabis composition.
28. The method of claim 10, wherein the second composition is a cannabis composition.
29. The method of claim 28, wherein the cannabis composition is an intoxicating cannabis composition.
30. The method of claim 29, wherein the intoxicating cannabis composition comprises THC.
31. The method of claim 30, wherein THC is ∆ 9 -THC.
32. The method of claim 30, wherein THC is ∆ 8 -THC.
33. The method of claim 28, wherein the cannabis composition is a non-intoxicating cannabis composition.
34. The method of claim 33, wherein the non-intoxicating cannabis composition comprises CBD, and is free of THC.
35. The method of any of claims 1-10, wherein the second composition is a washout composition.
36. The method of claim 10, wherein the second composition is a washout composition.
37. The method of claim 36, wherein the washout composition comprises an active compound selected from the group consisting of: amino acids, antioxidants, anti-inflammatory agents, analgesics, antineuropathic and antinociceptive agents, antimigraine agents, anxiolytics, antidepressants, antipsychotics, anti-PTSD agents, immunostimulants, anti-cancer agents, antiemetics, orexigenics, antiulcer agents, antihistamines, antihypertensives, anticonvulsants, antiepileptics, bronchodilators, neuroprotectants, entactogens and empathogens, entheogens, psychedelics, nootropics, monoamine oxidase inhibitors, sedatives, stimulants, supplements, and vitamins.
38. The method of any of claims 2-10, wherein the second number of days are an equivalent number of days of abstinence as the administration of a second composition of the invention, and are instead of administration of the second composition.
39. The method of any of claims 1-10, further comprising a number of days of abstinence from administration of any composition of the invention.
40. The method of claim 39, wherein the days of abstinence are after the first number of days of administration of the mushroom composition, and before the second number of days of administration of the second composition.
41. The method of claim 39, wherein the days of abstinence are after the second number of days of administration of the second composition, and before a next administration of the mushroom composition.
42. The method of claim 39, wherein the days of abstinence are within the first number of days of administration of the mushroom composition.
43. The method of claim 39, wherein the days of abstinence are within the second number of days of administration of the second composition.
44. The method of claim 39, wherein the number of days of abstinence is between 2 and 7 days.
45. The method of claim 39, wherein the number of days of abstinence is between 2 and 5 days.
46. The method of claim 39, wherein the number of days of abstinence is either 3 or 4 days
47. The method of any of claims 1-10, wherein the defined dosing regimen is repeated between 2 and 10 times to provide a therapeutic regime.
48. The method of claim 47, wherein the defined dosing regimen is repeated six times, seven times, or eight times.
49. The method of any of claims 2-10, wherein the daily dosing regimen is repeated between 2 and 10 times to provide a therapeutic regime.
50. The method of claim 49, wherein the daily dosing regimen is repeated six times, seven times, or eight times.
51. The method of claim 49, wherein the daily dosing regimen lasts for a period of six weeks, seven weeks, or eight weeks.
52. The method of claim 49, wherein the regime lasts from between 15 and 90 days.
53. The method of claim 52, wherein the regime lasts from between 30 and 70 days.
54. The method of claim 53, wherein the regime lasts from between 50 and 65 days.
55. The method of claim 54, wherein the regime lasts for 60 days.
56. A therapeutic regime comprising at least two different defined dosing regimens of claim 1.
57. The therapeutic regime of claim 56, comprising three or more different defined dosing regimens.
58. A therapeutic regime comprising at least two different daily dosing regimens of claim 2.
59. The therapeutic regime of claim 58, comprising three or more different daily dosing regimens.
60. A mushroom composition for use in the method of any of claims 1-10, comprising synergistically and therapeutically effective amounts of: a) a Hericium extract or a compound from Hericium ; and b) a Cordyceps extract or a compound from Cordyceps ; together with at least one carrier, diluent, or excipient, and wherein the at least one carrier, diluent, or excipient is not from a fungal source or found in fungi.
61. The mushroom composition of claim 60, further comprising a Psilocybe extract or a compound from Psilocybe .
62. The mushroom composition of claim 61, further comprising one or more of niacin, valerian root extract, vitamin B12, and a CB 1 receptor modulator.
63. The mushroom composition of claim 61, further comprising two or more of niacin, valerian root extract, vitamin B12, and a CB 1 receptor modulator.
64. The mushroom composition of claim 61, further comprising three or more of niacin, valerian root extract, vitamin B12, and a CB 1 receptor modulator.
65. The mushroom composition of claim 61, further comprising niacin, valerian root extract, vitamin B12, and a CB 1 receptor modulator.
66. The mushroom extract of claim 65, wherein the CB 1 receptor modulator is THC.
67. The mushroom extract of claim 65, wherein the CB 1 receptor modulator is CBD.
68. The mushroom extract of claim 65, wherein the CB 1 receptor modulator is PEA.
69. The mushroom composition of claim 61, further comprising one or more of rice hull concentrate powder, rice bran extract powder, and rice bran 100 mesh powder.
70. The mushroom composition of any of claims 60-69, further comprising a therapeutically effective amount of an additional active compound.
71. A cannabis composition for use in the method of any of claims 1-10, comprising synergistically and therapeutically effective amounts of: c) cannabidiol (CBD); d) an entourage-enhancing cannabinoid fraction; and e) an entourage-enhancing isoprenoid fraction; together with at least one carrier, diluent, or excipient, and wherein the at least one carrier, diluent, or excipient is not from a plant source or found in plants.
72. The cannabis composition of claim 71, further comprising THC.
73. The cannabis composition of claim 72, wherein the THC is ∆ 9 -THC.
74. The cannabis composition of claim 72, wherein the THC is ∆ 8 -THC.
75. The cannabis composition of claim 72, wherein the CBD is natural CBD.
76. The cannabis composition of claim 72, wherein the CBD is synthetic CBD.
77. The cannabis composition of claim 72, wherein the ratio of THC:CBD is from about 1:20 to about 20:1.
78. The cannabis composition of claim 72, wherein the ratio of THC:CBD is between 1:20 to 1:10, between 1:10 and 1:5, between 1:5 and 1:2, between 1:2 and about 1:1, between about 1:1 and 2:1, between 2:1 and 5:1, between 5:1 and 10:1, and between 10:1 and 20:1.
79. The cannabis composition of claim 78, wherein the ratio of THC:CBD is about 1:4.
80. The cannabis composition of any of claims 72, wherein the entourage-enhancing cannabinoid fraction comprises two or more cannabinoids.
81. The cannabis composition of claim 80, wherein all of the cannabinoids in the entourage-enhancing cannabinoid fraction are natural.
82. The cannabis composition of claim 81, wherein at least one of the natural cannabinoids is derived from a botanical source.
83. The cannabis composition of claim 82, wherein the botanical source is a Cannabis plant.
84. The cannabis composition of claim 80, wherein at least one of the cannabinoids in the entourage-enhancing cannabinoid fraction is synthetic.
85. The cannabis composition of claim 84, wherein all of the cannabinoids in the entourage-enhancing cannabinoid fraction are synthetic.
86. The cannabis composition of claim 80, wherein at least one of the cannabinoids in the entourage-enhancing cannabinoid fraction is selected for its ability to provide therapeutic effects or synergistic effects.
87. The cannabis composition of claim 80, wherein at least one of the cannabinoids in the entourage-enhancing cannabinoid fraction is selected for its ability to modulate the endocannabinoid system, modulate neurosteroid biosynthesis, or modulate a neurotransmitter system.
88. The cannabis composition of claim 80, wherein the entourage-enhancing cannabinoid fraction comprises at least two or more of: cannabichromene (CBC), cannabielsoin (CBE), cannabigerol (CBG), cannabicyclol (CBL), cannabinol (CBN), cannabidinodiol (CBND), cannabicitran (CBT), cannabitriol (CBT), cannabivarin (CBV), cannabigerol monomethyl ether (CBGM), cannabidivarin (CBDV), tetrahydrocannabivarin (THCV), cannabidiphorol (CBDP), tetrahydrocannabiphorol (THCP), and iso-tetrahydrocannabinol (iso-THC).
89. The cannabis composition of claim 80, wherein the entourage-enhancing cannabinoid fraction comprises two or more cannabinoids independently selected from any of CBC-type cannabinoids, CBD-type cannabinoids, CBE-type cannabinoids, CBG-type cannabinoids, CBL-type cannabinoids, CBN-type cannabinoids, CBND-type cannabinoids, CBT-type cannabinoids, ∆ 8 -THC-type cannabinoids, ∆ 9 -THC-type cannabinoids, and miscellaneous-type cannabinoids.
90. The cannabis composition of claim 89, wherein at least one cannabinoid is a CBC-type cannabinoid from the group including CBC, CBCA, ±CBCV, +CBCV, CBCVA, 4-acetoxy-CBC, and CBC-C 3.
91. The cannabis composition of claim 89, wherein at least one cannabinoid is a CBD-type cannabinoid from the group including CBD-C 5 , CBDA-C 5 , CBDM–C 5 , CBD-C 4 , CBDV, CBDVA, CBD-C 1 , CBDH, CBDP, and CBDD.
92. The cannabis composition of claim 89, wherein at least one cannabinoid is a CBE-type cannabinoid from the group including CBE-C 5 , CBEAA-C 5 , CBEAB-C 5 , CBE-C 3 , and CBEAB-C 3.
93. The cannabis composition of claim 89, wherein at least one cannabinoid is a CBG-type cannabinoid from the group including ( E )CBG, ( E )CBGA, ( E )CBGG, ( E )CBGAM, ( E )CBGV, ( E )CBGVA, (Z)CBGA, 5-acetyl-4-hydroxy-cannabigerol, (±)-6,7- trans -epoxycannabigerolic acid, (±)-6,7- cis -epoxycannabigerolic acid, (±)-6,7- cis -epoxycannabigerol, (±)-6,7- trans -epxoycannabigerol, camagerol, and sesquicannabigerol.
94. The cannabis composition of claim 89, wherein at least one cannabinoid is a CBL-type cannabinoid from the group including CBL, CBLA, and CBLV.
95. The cannabis composition of claim 89, wherein at least one cannabinoid is a CBN-type cannabinoid from the group including CBN-C 5 , CBNA-C 5 , CBN-C 4 , CBN-C 3 , CBN-C 2 , CBN-C 1 , CBNM–C 5 , 8-OH-CBN, 8-OH-CBNA, and 1’ S -OH-CBN.
96. The cannabis composition of claim 89, wherein at least one cannabinoid is a CBND-type cannabinoid from the group including CBND-C 3 and CBND-C 5.
97. The cannabis composition of claim 89, wherein at least one cannabinoid is a CBT-type cannabinoid from the group including (−)- trans -CBT-C 5 , (+)- trans -CBT-C 5 , (±)- cis -CBT-C 5 , (±)- trans -CBT-C 3 , CBT-C 3 -homologue, (−)- trans -CBT-OEt-C 5 , (–)- trans -CBT-OEt-C 3 , 8,9-Di-OH-CBT-C 5 , and CBDA-C 5 , and 9-OH-CBT-C 5 ester.
98. The cannabis composition of claim 89, wherein at least one cannabinoid is a ∆ 8 -THC-type cannabinoid from the group including ∆ 8 -THC, ∆ 8 -THCA, 10α-OH-∆ 8 -THC, 10β-OH-∆ 8 -THC, and 10a-α-hydroxy-10-oxo-∆ 8 -THC.
99. The cannabis composition of claim 89, wherein at least one cannabinoid is a ∆ 9 -THC-type cannabinoid from the group including ∆ 9 -THC-C 5 , ∆ 9 -THCAA-C 5 , ∆ 9 -THCAB-C 5 , ∆ 9 -THC-C 4 , ∆ 9 -THCAA-C 4 , ∆ 9 -THCV, ∆ 9 -THCVAA, ∆ 9 -THCO, ∆ 9 -THCOAA, ∆ 9 -THC–aldehyde, β-fenchyl (−)-Δ9-trans-tetrahydrocannabinolate, α-fenchyl (−)-Δ9-trans-tetrahydrocannabinolate, epi-bornyl (−)-Δ9-trans-tetrahydrocannabinolate, bornyl (−)-Δ9-trans-tetrahydrocannabinolate, α-terpenyl (−)-Δ9-trans-tetrahydrocannabinolate, 4-terpenyl (−)-Δ9-trans-tetrahydrocannabinolate, α-cadinyl (−)-Δ9-trans-tetrahydrocannabinolate, γ-eudesmyl (−)-Δ9-trans-tetrahydrocannabinolate, 8α-hydroxy-(−)-Δ9-trans-tetrahydrocannabinol, 8β-hydroxy-(−)-Δ9-trans-tetrahydro cannabinol 11-acetoxy-(−)-Δ9-trans-tetrahydrocannabinolic acid A, 8-oxo-(−)-Δ9-trans-tetrahydrocannabinol, cannabisol, (−)-Δ9-trans-tetrahydro-cannabi- phorol, and (−)-Δ9-trans-tetrahydrocannabihexol.
100. The cannabis composition of claim 89, wherein at least one cannabinoid is a miscellaneous-type cannabinoid from the group including DCBF-C 5 , CBF-C 5 , OH-iso-HHCV-C 3 OTHC, cannabicitran, cis -Δ 9 -THC, CBCON-C 5 , CBR, CBTT, CBCN-C 5 , CBCN-C 3 , cis -iso-Δ 7 -THCV, trans -iso-Δ 7 -THCV, trans -iso-Δ 7 -THC, CBCNB, CBCNC, CBCND, (–)-(7 R )-cannabicoumarononic acid, 4-acetoxy-2-geranyl-5-hydroxy-3- n -pentylphenol, 2-geranyl-5-hydroxy-3- n -pentyl-1,4-benzoquinone, 5-acetoxy-6-geranyl-3-n-pentyl-1,4-benzoquinone, CBM, CBX, 10α-hydroxy-Δ 9,11 -hexahydrocannabinol, 9β,10β-epoxyhexahydrocannabinol, 9α-hydroxyhexahydrocannabinol, 7-oxo-9α-hydroxyhexa-hydrocannabinol, 10α-hydroxyhexahydrocannabinol, 10aR-hydroxyhexahydrocannabinol, and 9α-hydroxy-10-oxo-Δ 6a,10a -THC.
101. The cannabis composition of any of claims 72, wherein the entourage-enhancing isoprenoid fraction comprises two or more isoprenoids.
102. The cannabis composition of claim 101, wherein all of the isoprenoids in the entourage-enhancing isoprenoid fraction are natural.
103. The cannabis composition of claim 102, wherein at least one of the natural isoprenoids is derived from a botanical source.
104. The cannabis composition of claim 103, wherein the botanical source is a Cannabis plant.
105. The cannabis composition of claim 101, wherein at least one of the isoprenoids in the entourage-enhancing isoprenoid fraction is synthetic.
106. The cannabis composition of claim 105, wherein all of the isoprenoids in the entourage-enhancing isoprenoid fraction are synthetic.
107. The cannabis composition of claim 101, wherein at least one of the isoprenoids in the entourage-enhancing isoprenoid fraction is selected for its ability to provide therapeutic effects or synergistic effects.
108. The cannabis composition of claim 101, wherein at least one of the isoprenoids in the entourage-enhancing isoprenoid fraction is selected for its ability to modulate the endocannabinoid system, modulate neurosteroid biosynthesis, or modulate a neurotransmitter system.
109. The cannabis composition of claim 101, wherein the two or more isoprenoids are selected from the group consisting of: alpha-bisabolol, beta-caryophyllene, camphene, carene caryophyllene oxide alpha-humulene fenchol guaiene guaiol limonene linalool, myrcene, nerolidol, ocimene, alpha-phellandrene, alpha-pinene, beta-pinene, alpha-terpinene, gamma-terpinene, terpineol, and terpinolene.
110. The cannabis composition of any of claims 71-109, wherein the entourage-enhancing isoprenoid fraction is myrcene dominant.
111. The cannabis composition of claim 110, wherein the myrcene dominant entourage-enhancing isoprenoid fraction contains at least 20% myrcene by weight.
112. The cannabis composition of claim 101, wherein the entourage enhancing isoprenoid fraction includes two or more isoprenoids independently selected from any of monoterpenes, sesquiterpenes, diterpenes, triterpenes, and miscellaneous terpenes.
113. The cannabis composition of claim 112, wherein at least one isoprenoid is from the group of monoterpenes which include myrcene, cis-β-ocimene, trans-β-ocimene, p-cymene, α-terpinene, β-phellandrene, γ-terpinene, α-terpinolene, α-phellandrene, 3-phenyl-2-methyl-prop-1-ene, α-pinene, β-pinene, camphene, Δ 3 -carene, Δ 4 -carene, sabinene, α-thujene, linalool, citral B, nerol, geraniol, ipsienol, citronellol, 2-methyl-2-heptene-6-on, geranyl acetone, m-mentha-1,8-(9)-dien-5-ol, carvacrol, carvone, α-terpineol, terpinene-4-ol, pulegone, dihydrocarvone, β-terpineol, dihydrocarveyl acetate, p-cymene-8-ol, β-cyclocitral, safranal, cis-linalool oxide, perillene, sabinol, thujyl alcohol, linalool oxide, cis-carveol, cis-sabinene hydrate, sabinene hydrate, ,8-cineol, 1,4-cineol, piperitone oxide, piperitenone oxide, fenchyl alcohol, fenchone, borneol, bornyl acetate, camphor, camphene hydrate, α-pinene oxide, pinocarveol, and pinocarvone.
114. The cannabis composition of claim 112, wherein at least one isoprenoid is from the group of sesquiterpenes which include α-caryophyllene, β-caryophyllene, caryophyllene oxide, curcumene, α-trans-bergamotene, α-selinene, β-farnesene, longifolene, humulene epoxide I, humulene epoxide II, caryophyllene alcohol (caryophyllenol), β-bisabolene, allo-aromadendrene, calamenene, α-copaene, nerolidol, α-gurjunene, iso-caryophyllene, β-selinene, selina-3,7(11)-diene, selina-4(14),7(11)-diene, α-bisabolol, α-cedrene, α-cubebene, δ-cadinene, epi-β-santalene, farnesol, γ-cadinene, γ-elemene, γ-eudesmol, guaiol, ledol, trans-trans-α-farnesene, (Z)-β-farnesene, farnesyl acetone, α-cadinene, α-cis-bergamotene, α-eudesmol, α-guaiene, α-longipinene, α-ylangene, β-elemene, β-eudesmol, epi-α-bisabolol, γ-cis-bisabolene, γ-curcumene, γ-muurolene, γ-trans-bisabolene, viridiflorene, germacrene-B, and clovandiol.
115. The cannabis composition of claim 112, wherein at least one isoprenoid is from the group of diterpenes which include phytol and neophytadiene.
116. The cannabis composition of claim 112, wherein at least one isoprenoid is from the group of triterpenes which include friedeline and epifriedelanol.
117. The cannabis composition of claim 112, wherein at least one isoprenoid is from the group of miscellaneous terpenes which include vomifoliol, dihydrovomifoliol, β-ionone, and dihydroactinidiolide.
118. The cannabis composition of any of claims 71-117, wherein the entourage-enhancing cannabinoid fraction or the entourage-enhancing isoprenoid fraction provides a therapeutic effect, decreases an unwanted effect, increases stability or shelf-life, or alters pharmacokinetics or pharmacodynamics.
119. The cannabis composition of claim 118, wherein the therapeutic effect is an antioxidant, anti-inflammatory, analgesic, antineuropathic, antinociceptive, antimigraine, anxiolytic, antidepressant, antipsychotic, anti-PTSD, immunostimulant, anti-cancer, antiemetic, orexigenic, antiulcer, antihistamine, antihypertensive, anticonvulsant, antiepileptic, bronchodilator, neuroprotective, entactogenic, empathogenic, entheogenic, psychedelic, sedative, or stimulant effect.
120. The cannabis composition of any of claims 71-119, wherein the entourage-enhancing cannabinoid fraction or the entourage-enhancing isoprenoid fraction provides a synergistic effect.
121. The cannabis composition of claim 120, wherein the synergistic effect is a greater than additive increase in potency, bioactivity, bioaccessibility, bioavailability, or therapeutic effect.
122. The cannabis composition of any of claims 71-121 wherein the entourage-enhancing cannabinoid fraction or the entourage-enhancing isoprenoid fraction modulates the endocannabinoid system, modulates neurosteroid biosynthesis, or modulates a neurotransmitter system.
123. The cannabis composition of any of claims 71-122 wherein the entourage-enhancing cannabinoid fraction comprises from 0.1% to about 25% of the composition by weight.
124. The cannabis composition of claim 123 wherein the entourage-enhancing cannabinoid fraction comprises about 10% of the composition by weight.
125. The cannabis composition of any of claims 71-124 wherein the entourage-enhancing isoprenoid fraction comprises from 0.1% to about 10% of the composition by weight.
126. The cannabis composition of claim 125 wherein the entourage-enhancing isoprenoid fraction comprises about 5% of the composition by weight.
127. The cannabis composition of claim 72, further comprising one or more flavonoids.
128. The cannabis composition of claim 127, wherein the one or more flavonoids is any of orientin, orientin-O-glucoside, orientin-7-O-glucoside, orientin-7-O-rhamnoglucoside, vitexin, vitexin-O-glucoside, vitexin-7-O-glucoside, vitexin-7-O-rhamnoglucoside, cytisoside, cytisoside-glucoside, isovitexin, isovitexin-O-glucoside, isovitexin-7-O-glucoarbinoside, isovitexin-7-O-rhamnoglucoside, apigenin-7-O-glucoside, apigenin-7-O-glucuronoid, apigenin-7-O'P-coumaroylglucoside, 6-prenylapigenin, apigenin-6,8-di-glucopyranoside, luteolin-C–glucuronide, luteolin-7-O-glucuronide, canniflavin A, canniflavin B, canniflavin C, chrysoeriol, kaempferol-3-O-diglucoside, quercetin-3-O-glucoside, quercetin-3-O-diglucoside, kaempferol-3-O-sophoroside, quercetin-3-O-sophoroside, rutin, quercetin, naringenin, and naringin.
129. The cannabis composition of claim 72, formulated for any one of: (1) oral administration, and comprising a hydrophobic solvent or an alcohol-based solvent, and optionally a co-solvent; (2) use with a transdermal drug delivery system, wherein the composition is combined with a permeation enhancing agent, and optionally with one or more stabilizers, solubilizers, or adhesives; or (3) administration as a nanostructured formulation selected from the group consisting of: a nanoemulsion, a nanocapsule, a nanoparticle conjugate, or a nano-encapsulated oral or nasal spray.
130. The cannabis composition of any of claims 71-129, further comprising a therapeutically effective amount of an additional active compound.
131. The mushroom composition of claim 61 or the cannabis composition of claim 72, wherein said composition is prepared for administration as a formulation selected from the group consisting of: a soft or hard gelatin capsule, an oral tablet, a timed release capsule or tablet, a controlled release capsule or tablet, an extended release capsule or tablet, gastrorententive tablet, a sublingual or buccal tablet, an oral spray, a nasal spray, a suppository, a suspension, a solution suitable for subcutaneous, intravenous, intraperitoneal, or intramuscular administration, a liquid suitable for use in a liquid vaporization appliance, a topical preparation, or a transdermal patch.
132. The mushroom composition of claim 70 or the cannabis composition of claim 130, wherein the additional active compound acts to increase therapeutic efficacy, provide additional therapeutic effects, decrease unwanted effects, increase stability or shelf-life, improve bioavailability, induce synergy, or alter pharmacokinetics or pharmacodynamics.
133. The mushroom composition of claim 70 or the cannabis composition of claim 130, wherein the additional active compound is selected from the group consisting of: amino acids, antioxidants, anti-inflammatory agents, analgesics, antineuropathic and antinociceptive agents, antimigraine agents, anxiolytics, antidepressants, antipsychotics, anti-PTSD agents, immunostimulants, anti-cancer agents, antiemetics, orexigenics, antiulcer agents, antihistamines, antihypertensives, anticonvulsants, antiepileptics, bronchodilators, neuroprotectants, entactogens and empathogens, entheogens, psychedelics, nootropics, monoamine oxidase inhibitors, sedatives, stimulants, supplements, and vitamins.
134. A unit dosage form, useful to treat a mammal with a CNS disorder, which comprises a therapeutically effective amount of the functional mushroom composition of claim 60.
135. A unit dosage form, useful to treat a mammal with a CNS disorder, which comprises a therapeutically effective amount of the psychedelic mushroom composition of claim 61.
136. A unit dosage form, useful to treat a mammal with a CNS disorder, which comprises a therapeutically effective amount of the non-intoxicating cannabis composition of claim 71.
137. A unit dosage form, useful to treat a mammal with a CNS disorder, which comprises a therapeutically effective amount of the intoxicating cannabis composition of claim 72.
138. The unit dosage form any of claims 134-137, wherein the unit dosage form is suitable for oral, mucosal, rectal, injectable, intranasal, inhaled, or transdermal administration.
139. The unit dosage form claim 138, as an oral solid form or an oral liquid form.
140. The unit dosage form of claim 135, comprising ground or pulverized Psilocybe fungal matter in a total amount of between 1 mg and 1 g.
141. The unit dosage form of claim 135, comprising a Psilocybe extract in a total amount of between 0.5 mg and 500 mg.
142. The unit dosage form of claim 135, comprising psilocybin in a total amount of between 0.01 mg and 10 mg.
143. The unit dosage form of claim 142, comprising psilocybin in a total amount of between 0.5 mg and 3 mg.
144. The unit dosage form of claim 143, comprising psilocybin in a total amount of 2.5 mg.
145. The unit dosage form of claim 143, comprising psilocybin in a total amount of 1.25 mg.
146. The unit dosage form of claim 137, comprising THC in a total amount of between 0.1 mg and 100 mg.
147. The unit dosage form of claim 137, comprising THC in a total amount of between 2 mg and 50 mg.
148. The unit dosage form of claim 147, comprising THC in a total amount of 10 mg.
149. The unit dosage form of claim 148, comprising THC in a total amount of 5 mg.
150. A method of treating a mental health condition or improving mental health, in a patient in need thereof, comprising administering to the patient a unit dosage form of claim 135, followed by a unit dosage form of claim 137, according to a defined dosing regimen.
151. The method of claim 150, wherein the defined dosing regimen is a daily dosing regimen consisting of a first number of days of administration of the unit dosage form of claim 135, followed by a second number of days of administration of the unit dosage form of claim 137.
152. A method of treating a mental health condition or improving mental health, in a patient in need thereof, comprising administering to the patient a unit dosage form of claim 134, followed by a unit dosage form of claim 137, according to a defined dosing regimen.
153. The method of claim 152, wherein the defined dosing regimen is a daily dosing regimen consisting of a first number of days of administration of the unit dosage form of claim 134, followed by a second number of days of administration of the unit dosage form of claim 137.
154. A method of treating a mental health condition or improving mental health, in a patient in need thereof, comprising administering to the patient a unit dosage form of claim 134, followed by a unit dosage form of claim 136, according to a defined dosing regimen.
155. The method of claim 154, wherein the defined dosing regimen is a daily dosing regimen consisting of a first number of days of administration of the unit dosage form of claim 134, followed by a second number of days of administration of the unit dosage form of claim 136.
156. A method of treating a mental health condition or improving mental health, in a patient in need thereof, comprising administering to the patient a unit dosage form of claim 135, followed by a unit dosage form of claim 136, according to a defined dosing regimen.
157. The method of claim 156, wherein the defined dosing regimen is a daily dosing regimen consisting of a first number of days of administration of the unit dosage form of claim 135, followed by a second number of days of administration of the unit dosage form of claim 136.
158. A method of treating a mental health condition or improving mental health, in a patient in need thereof, comprising administering to the patient a unit dosage form of claim 135, followed by a unit dosage form of claim 134, according to a defined dosing regimen.
159. The method of claim 158, wherein the defined dosing regimen is a daily dosing regimen consisting of a first number of days of administration of the unit dosage form of claim 135, followed by a second number of days of administration of the unit dosage form of claim 134.
160. A method of treating a mental health condition or improving mental health, in a patient in need thereof, comprising administering to the patient a unit dosage form of claim 134, followed by a unit dosage form of a washout composition, according to a defined dosing regimen.
161. The method of claim 160, wherein the defined dosing regimen is a daily dosing regimen consisting of a first number of days of administration of the unit dosage form of claim 134, followed by a second number of days of administration of the washout composition.
162. The method of either of claims 160 or 161, wherein the washout composition comprises an active compound selected from the group consisting of: amino acids, antioxidants, anti-inflammatory agents, analgesics, antineuropathic and antinociceptive agents, antimigraine agents, anxiolytics, antidepressants, antipsychotics, anti-PTSD agents, immunostimulants, anti-cancer agents, antiemetics, orexigenics, antiulcer agents, antihistamines, antihypertensives, anticonvulsants, antiepileptics, bronchodilators, neuroprotectants, entactogens and empathogens, entheogens, psychedelics, nootropics, monoamine oxidase inhibitors, sedatives, stimulants, supplements, and vitamins.
163. The method of any of claims 151, 153, 155, 157, 159, or 161, wherein the first number of days is between 2 and 7 days.
164. The method of claim 163, wherein the first number of days is between 2 and 5 days.
165. The method of claim 164, wherein the first number of days is either 3 or 4 days.
166. The method of claim 165, wherein the first number of days is 4 days.
167. The method of any of claims 151, 153, 155, 157, 159, or 161, wherein the second number of days is between 2 and 7 days.
168. The method of claim 167, wherein the second number of days is between 2 and 5 days.
169. The method of claim 168, wherein the second number of days is either 3 or 4 days.
170. The method of claim 169, wherein the second number of days is 3 days.
171. The method of any of claims 150-170, wherein the defined dosing regimen is repeated between 2 and 10 times to provide a therapeutic regime.
172. The method of claim 171, wherein the defined dosing regimen is repeated six times, seven times, or eight times.
173. The method of any of claims 151, 153, 155, 157, 159, or 161, wherein the daily dosing regimen is repeated between 2 and 10 times to provide a therapeutic regime.
174. The method of claim 173, wherein the daily dosing regimen is repeated six times, seven times, or eight times.
175. The method of claim 173, wherein the daily dosing regimen lasts for a period of six weeks, seven weeks, or eight weeks.
176. The method of either of claims 171 or 173, wherein the regime lasts from between 15 and 90 days.
177. The method of either of claims 171 or 173, wherein the regime lasts from between 30 and 70 days.
178. The method of either of claims 171 or 173, wherein the regime lasts from between 50 and 65 days.
179. The method of either of claims 171 or 173, wherein the regime lasts for 60 days.
180. A daily dosing kit according to the method of any of claims 151, 153, 155, 157, 159, or 161, comprising the unit dosage forms to be administered according to said method, and in such amount as the number of total days of their administration.
181. The daily dosing kit of claim 180, comprising a first amount of a unit dosage form of claim 135, and a second amount of a unit dosage form of claim 137.
182. The daily dosing kit of claim 180, comprising a first amount of a unit dosage form of claim 134, and a second amount of a unit dosage form of claim 137.
183. The daily dosing kit of claim 180, comprising a first amount of a unit dosage form of claim 135, and a second amount of a unit dosage form of claim 136.
184. The daily dosing kit of claim 180, comprising a first amount of a unit dosage form of claim 134, and a second amount of a unit dosage form of claim 136.
185. The daily dosing kit of claim 180, comprising a first amount of a unit dosage form of claim 135, and a second amount of a unit dosage form of claim 134.
186. The daily dosing kit of claim 180, comprising a first amount of a unit dosage form of claim 135, and a second amount of a unit dosage form of a washout composition.
187. The daily dosing kit of any of claims 180-186, provided as a blister pack, a blister tray, a blister card, a strip pack, a push-through pack, a compliance pack, or a calendar pack.
188. A method for modulating neurotransmission in a mammal in need of such modulation, comprising administering to the mammal a first unit dosage form of any of claims 134-149 together with a second unit dosage form of any of claims 134-149, according to a defined dosing regimen.
189. The method of claim 188. wherein said neurotransmission is catecholaminergic, gabaminergic, glutamatergic, or serotonergic neurotransmission.
190. The method of claim 189, wherein said serotonergic neurotransmission is a modulation of 5-HT2A receptors, without a significant modulation of 5-HT2B receptors.
191. The method of claim 189, wherein said serotonergic neurotransmission is a modulation of 5-HT1A and 5-HT2A receptors.
192. A method for modulating endocannabinoid system activity in a mammal in need of such modulation, the method comprising administering to the mammal a first unit dosage form of any of claims 134-149 together with a second unit dosage form of any of claims 134-149, according to a defined dosing regimen.
193. A method for modulating biosynthesis of a neuroactive steroid in a mammal in need of such modulation, the method comprising administering to the mammal a first unit dosage form of any of claims 134-149 together with a second unit dosage form of any of claims 134-149, according to a defined dosing regimen.
194. A method of treating a medical condition in a mammal in need of such treatment, the method comprising administering to the mammal a first unit dosage form of any of claims 134-149 together with a second unit dosage form of any of claims 134-149, according to a defined dosing regimen.
195. The method of claim 194, wherein the mammal is a human.
196. The method of claim 194, wherein the medical condition is a disorder linked to dysregulation or inadequate functioning of neurotransmission.
197. The method of claim 196, wherein the disorder linked to dysregulation or inadequate functioning of neurotransmission is that of catecholaminergic, gabaminergic, glutamatergic, or serotonergic neurotransmission.
198. The method of claim 196, wherein the disorder linked to dysregulation or inadequate functioning of neurotransmission is that of serotonergic neurotransmission.
199. The method of claim 194, wherein the medical condition is a disorder linked to dysregulation or inadequate functioning of the endocannabinoid system.
200. The method of claim 194, wherein the medical condition is a disorder linked to dysregulation or inadequate functioning neurosteroid biosynthesis.
201. The method of claim 194, wherein the medical condition is a mental health condition.
202. The method of claim 201, wherein the mental health condition is selected from the group consisting of: post-traumatic stress disorder (PTSD), adjustment disorder, affective disorder, depression, dysthymia, anxiety, phobia disorders, binge disorders, body dysmorphic disorder, alcohol or drug abuse or dependence disorders, disruptive behavior disorders, eating disorders, impulse control disorders, obsessive compulsive disorder (OCD), attention deficit hyperactivity disorder (ADHD), personality disorders, attachment disorders, and dissociative disorders.
203. The method of claim 201, wherein the mental health condition is a disorder related to rigid modes of thinking.
204. The method of claim 201, wherein the disorder related to rigid modes of thinking is an anxiety disorder, a depressive disorder, addiction, an eating disorder, OCD, or PTSD.
205. A method of reducing the symptoms of a mental health condition in a human, the method comprising identifying a human in need of said reducing, and administering to the human a first unit dosage form of any of claims 134-149 together with a second unit dosage form of any of claims 134-149, according to a defined dosing regimen.
206. The method of claim 205, wherein the mental health condition is PTSD.
207. The method of claim 206, wherein the symptoms of PTSD that are reduced include one or more of flashbacks, nightmares, distressing and intense memories, distress or physical reactions after being exposed to triggers, blaming self or others for the trauma, decreased interest in things that were once enjoyable, negative feelings about self and the world, inability to remember the trauma clearly, difficulty feeling positive, feelings of isolation, negative affect, difficulty feeling positive, avoidance, aggression or irritability, hypervigilance and hyper-awareness, difficulty concentrating, difficulty sleeping, heightened startle response, engaging in self-destructive, or risky behavior, difficulty sleeping or staying asleep, or suicidal ideation.
208. A method of improving psychological functioning in a human, the method comprising identifying a human in need of said improving, and administering to the human a first unit dosage form of any of claims 134-149 together with a second unit dosage form of any of claims 134-149, according to a defined dosing regimen, and wherein the improvement in psychological functioning is a reduction of neuroticism or psychological defensiveness, an increase in creativity or openness to experience, an increase in decision-making ability, an increase in feelings of wellness or satisfaction, or an increase in ability to fall or stay asleep.
209. A method of treating a CNS disorder in a human patient, the method comprising administering to the patient a first unit dosage form of any of claims 134-149 together with a second unit dosage form of any of claims 134-149, according to a defined dosing regimen, and in combination with one or more psychotherapy sessions, or in conjunction with participation in a therapeutically beneficial self-care activity wherein the therapeutically beneficial self-care activity is chosen from the group consisting of: breathing exercises, meditation and concentration practices, focusing on an object or mantra, listening to music, physical exercise, stretching or bodywork, journaling, grounding techniques, positive self-talk, or engaging with a pet or animal.
PCT/CA2022/050374 2021-03-12 2022-03-14 Dosing regimens of pharmaceutical and nutraceutical mushroom and cannabis compositions and their use to treat cns disorders and improve mental health WO2022187973A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202163160235P 2021-03-12 2021-03-12
US63/160,235 2021-03-12

Publications (1)

Publication Number Publication Date
WO2022187973A1 true WO2022187973A1 (en) 2022-09-15

Family

ID=83226150

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CA2022/050374 WO2022187973A1 (en) 2021-03-12 2022-03-14 Dosing regimens of pharmaceutical and nutraceutical mushroom and cannabis compositions and their use to treat cns disorders and improve mental health

Country Status (1)

Country Link
WO (1) WO2022187973A1 (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20220370483A1 (en) * 2021-05-07 2022-11-24 Turtle Bear Holdings, Llc Fungal compound compositions and methods for modulating inflammation
WO2023082003A1 (en) * 2021-11-10 2023-05-19 London Pharmaceuticals And Research Corporation New cannabinoid-gabapentinoid conjugates and uses thereof
CN116549478A (en) * 2023-05-06 2023-08-08 云南中医药大学 Pharmaceutical composition and preparation method and application thereof
US11850254B2 (en) 2019-11-19 2023-12-26 Turtle Bear Holdings, Llc Tryptamine compositions for enhancing neurite outgrowth
US11896587B2 (en) 2016-07-23 2024-02-13 Turtle Bear Holdings, Llp Tryptamine compositions for enhancing neurite outgrowth

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018135943A1 (en) * 2017-01-18 2018-07-26 Procare Beheer B.V. Psilocybin and/or psilocin in combination with cannabinoids and/or terpenes
WO2019099745A1 (en) * 2017-11-16 2019-05-23 CaaMTech, LLC Compositions comprising a psilocybin derivative and a cannabinoid
WO2020188577A1 (en) * 2019-03-21 2020-09-24 Alvit Lcs Pharma Ltd. Fixed dose combination of cannabinoids and medical mushrooms for prevention and treatment of cancer, inflammatory or immune-mediated inflammatory diseases
WO2021022378A1 (en) * 2019-08-08 2021-02-11 Neptune Wellness Solutions Inc. Oral formulations of cannabis extracts and methods of making same
US20210069170A1 (en) * 2016-07-23 2021-03-11 Paul Edward Stamets Tryptamine compositions for enhancing neurite outgrowth

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20210069170A1 (en) * 2016-07-23 2021-03-11 Paul Edward Stamets Tryptamine compositions for enhancing neurite outgrowth
WO2018135943A1 (en) * 2017-01-18 2018-07-26 Procare Beheer B.V. Psilocybin and/or psilocin in combination with cannabinoids and/or terpenes
WO2019099745A1 (en) * 2017-11-16 2019-05-23 CaaMTech, LLC Compositions comprising a psilocybin derivative and a cannabinoid
WO2020188577A1 (en) * 2019-03-21 2020-09-24 Alvit Lcs Pharma Ltd. Fixed dose combination of cannabinoids and medical mushrooms for prevention and treatment of cancer, inflammatory or immune-mediated inflammatory diseases
WO2021022378A1 (en) * 2019-08-08 2021-02-11 Neptune Wellness Solutions Inc. Oral formulations of cannabis extracts and methods of making same

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11896587B2 (en) 2016-07-23 2024-02-13 Turtle Bear Holdings, Llp Tryptamine compositions for enhancing neurite outgrowth
US11850254B2 (en) 2019-11-19 2023-12-26 Turtle Bear Holdings, Llc Tryptamine compositions for enhancing neurite outgrowth
US11911401B2 (en) 2019-11-19 2024-02-27 Turtle Bear Holdings, Llc Tryptamine compositions for enhancing neurite outgrowth
US20220370483A1 (en) * 2021-05-07 2022-11-24 Turtle Bear Holdings, Llc Fungal compound compositions and methods for modulating inflammation
WO2023082003A1 (en) * 2021-11-10 2023-05-19 London Pharmaceuticals And Research Corporation New cannabinoid-gabapentinoid conjugates and uses thereof
CN116549478A (en) * 2023-05-06 2023-08-08 云南中医药大学 Pharmaceutical composition and preparation method and application thereof
CN116549478B (en) * 2023-05-06 2024-03-19 云南中医药大学 Pharmaceutical composition and preparation method and application thereof

Similar Documents

Publication Publication Date Title
WO2022187973A1 (en) Dosing regimens of pharmaceutical and nutraceutical mushroom and cannabis compositions and their use to treat cns disorders and improve mental health
Mazzanti et al. Curcumin and resveratrol in the management of cognitive disorders: what is the clinical evidence?
Pritam et al. Antioxidants in Alzheimer’s disease: Current therapeutic significance and future prospects
AU2018100924A4 (en) Composition and method for treating pain
Rigacci et al. Nutraceuticals and amyloid neurodegenerative diseases: a focus on natural phenols
Aboukhatwa et al. Antidepressants are a rational complementary therapy for the treatment of Alzheimer's disease
Do et al. Mangosteen pericarp and its bioactive xanthones: Potential therapeutic value in Alzheimer’s disease, Parkinson’s disease, and depression with pharmacokinetic and safety profiles
WO2020044116A1 (en) Therapeutic combinations of cannabinoids with curcumin
Zanforlin et al. The medicinal chemistry of natural and semisynthetic compounds against Parkinson’s and Huntington’s diseases
US20240024340A1 (en) Compositions and Methods For Treating Migraine
Singh et al. Antidepressant activity of methanolic extract of Foeniculum vulgare (fennel) fruits in experimental animal models
Arnold et al. The safety and efficacy of low oral doses of cannabidiol: An evaluation of the evidence
WO2020044119A2 (en) Oral formulations of lavender and cannabinoids
US20210205236A1 (en) Therapeutic combinations of boswellia extract and cannabinoids
EP2694051B1 (en) Use of l-carnitine and salts thereof for reducing or preventing fatigue and improving cognitive function
Andrade et al. Therapeutic potential of natural compounds in neurodegenerative diseases: Insights from clinical trials
US20210338629A1 (en) Oral formulations of phenylalanine and cannabinoids
Olatunji et al. Sceletium tortuosum: A review on its phytochemistry, pharmacokinetics, biological and clinical activities
US10201577B2 (en) Compositions and methods for enhancing working and long-term memory support using new combination of ginkgo biloba and panax ginseng and associated components
EGHTESADI et al. The effects of saffron on psychiatric disorders: A review.
Muss et al. Neuroprotective impact of a vitamin trace element composition-a randomized, double blind, placebo controlled clinical trial with healthy volunteers
WO2020115751A1 (en) Cannabis-based compositions for the treatment of alzheimer&#39;s disease and dementia
Ayati et al. Advances in treatment of mild cognitive impairment (MCI) and dementia: A review of promising non-pharmaceutical modalities
WO2024054688A2 (en) Therapeutic combinations for movement disorders
AU2021390590B2 (en) Antiinflammatory compositions comprising cannabidiol, delta-9- tetrahydrocannabinol and linalool

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: 22766054

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE