WO2021245675A1 - Cannabidiolic acid (cbda) derivatives and uses thereof - Google Patents

Cannabidiolic acid (cbda) derivatives and uses thereof Download PDF

Info

Publication number
WO2021245675A1
WO2021245675A1 PCT/IL2021/050666 IL2021050666W WO2021245675A1 WO 2021245675 A1 WO2021245675 A1 WO 2021245675A1 IL 2021050666 W IL2021050666 W IL 2021050666W WO 2021245675 A1 WO2021245675 A1 WO 2021245675A1
Authority
WO
WIPO (PCT)
Prior art keywords
formula
represented
linear
pharmaceutical composition
substituted
Prior art date
Application number
PCT/IL2021/050666
Other languages
French (fr)
Inventor
Reshef SWISA
Anat IOSUB-AMIR
Ron SHAHARABANI
Raphael Mechoulam
Natalya M. Kogan
Original Assignee
Epm (Ip), Inc.
Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd.
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 Epm (Ip), Inc., Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd. filed Critical Epm (Ip), Inc.
Publication of WO2021245675A1 publication Critical patent/WO2021245675A1/en

Links

Classifications

    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/08Drugs for disorders of the alimentary tract or the digestive system for nausea, cinetosis or vertigo; Antiemetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/08Antiepileptics; Anticonvulsants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/22Anxiolytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/24Antidepressants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/06Antiglaucoma agents or miotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C69/00Esters of carboxylic acids; Esters of carbonic or haloformic acids
    • C07C69/76Esters of carboxylic acids having a carboxyl group bound to a carbon atom of a six-membered aromatic ring
    • C07C69/84Esters of carboxylic acids having a carboxyl group bound to a carbon atom of a six-membered aromatic ring of monocyclic hydroxy carboxylic acids, the hydroxy groups and the carboxyl groups of which are bound to carbon atoms of a six-membered aromatic ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C69/00Esters of carboxylic acids; Esters of carbonic or haloformic acids
    • C07C69/76Esters of carboxylic acids having a carboxyl group bound to a carbon atom of a six-membered aromatic ring
    • C07C69/84Esters of carboxylic acids having a carboxyl group bound to a carbon atom of a six-membered aromatic ring of monocyclic hydroxy carboxylic acids, the hydroxy groups and the carboxyl groups of which are bound to carbon atoms of a six-membered aromatic ring
    • C07C69/86Esters of carboxylic acids having a carboxyl group bound to a carbon atom of a six-membered aromatic ring of monocyclic hydroxy carboxylic acids, the hydroxy groups and the carboxyl groups of which are bound to carbon atoms of a six-membered aromatic ring with esterified hydroxyl groups

Definitions

  • the present invention relates to cannabidiolic acid (CBDA) derivatives, including acids and esters, and uses thereof in the treatment of various diseases, disorders, conditions and symptoms.
  • CBDA cannabidiolic acid
  • Cannabidiol is the major non-psycho tropic phytocannabinoid compound in the plant Cannabis sativa, making up to 40% of the cannabinoids in Cannabis extracts (Grlic, Bull. Narc., 1976, 14:37-46). CBD is considered a lead compound for treating and preventing inflammatory and oxidative damage, see for example WO 1999/053917.
  • Cannabidiolic acid is also a major constituent of the Cannabis sativa plant and was first isolated in 1955 (Krejci and Santavy, 1955, Acta Univ Palacki Olomuc 6:59- 66). Its structure was elucidated in 1965 by analysis of the physical properties of its methyl ester, cannabidiolic acid methyl ester (CBDA-ME) (Mechoulam and Gaoni, Tetrahedron, 1965, 21:1223-1229). Its synthesis from cannabidiol was subsequently reported (Mechoulam and Ben-Zvi, J. Chem. Soc. Commun., 1969, 7:343-344).
  • the cannabinoid acids are precursors of the natural cannabinoids (Potter et al., J. Forensic Sci., 2008, 53:90-94), potentially lowering the amount of drug required to induce effects.
  • the decarboxylation of CBDA into CBD is enhanced by heat, indicating the relative instability of CBDA, thus lowering its potential to serve as a medication (Mechoulam, Academic Press, New York, 1973, 1-99; Citti et al., J. Pharm. Biomed. Anal., 2018, 16:532-540).
  • CBDA-ME is a relatively unknown cannabinoid and remains understudied and its effects are only just starting to become clear.
  • CBDA-ME is a stable derivative of CBDA and can be pharmacological active in vivo.
  • Pertwee et al. (Brit. J. Pharmacology, 2018, 175:100-112) reported that the methyl ester of CBDA, designated HU-580 (also denoted herein EPM-301), displays a greater potency than CBDA in suppressing signs of both acute and anticipatory nausea, and of stress-induced anxiety in rats, and that it produces these effects in a 5-HT IA receptor- dependent manner.
  • Another recent study (Hen-Shoval et al., Behav. Brain Res., 2018, 351:1-3) provides support for a potent anti-depressant effect after oral ingestion of a low dose (1 mg/kg) of HU-580 in two rat models.
  • WO 2018/235079 discloses compositions comprising CBDA esters and uses thereof in the treatment of a condition, disease or symptom associated with 5-HT IA receptors.
  • the present invention provides novel compounds comprising cannabidiolic acid (CBDA) derivatives, including acid and esters, and uses thereof.
  • CBDA cannabidiolic acid
  • the present invention further provides pharmaceutical compositions comprising CBDA derivatives, alone or in combination with one or more additional cannabinoid compounds, for use in treating various diseases, disorders and symptoms, including but not limited to pain, impaired neurological function, inflammation including respiratory inflammation, inflammatory bowel disease and autoimmune diseases, nausea, vomiting, convulsions, low appetite and glaucoma.
  • compositions comprising cannabidiolic acid
  • CBDA cannabidiol
  • CBDA ester derivatives exhibit prolonged and significant therapeutic effects compared to cannabidiol (CBD).
  • CBDA cannabidiol
  • the pharmaceutical compositions disclosed herein are non-psycho active.
  • the present invention provides pharmaceutical compositions comprising CBDA derivatives having superior pharmacokinetics compared to the commercially available cannabidiol (CBD).
  • the present invention provides a compound represented by the structure of Formula (la),
  • Ri is selected from the group consisting of a hydrogen, a linear or branched unsubstituted or substituted C 1 -C 15 alkyl, a linear or branched unsubstituted or substituted C 2 -C 15 alkenyl, and a linear or branched unsubstituted or substituted C2-C15 alkynyl;
  • R 2 is selected from the group consisting of a linear or branched unsubstituted or substituted C 1 -C 15 alkyl, a linear or branched unsubstituted or substituted C 2 - Ci 5 alkenyl, and a linear or branched unsubstituted or substituted C 2 -C 15 alkynyl; each of R 3 and R 4 is independently selected from the group consisting of hydrogen and -C(0)-R 5 , wherein R 5 is a C 1 -C 15 alkyl; and stereoisomers, and salts thereof.
  • the compound is represented by the structure of
  • Ri is hydrogen.
  • Ri is a linear unsubstituted C 1 -C 15 alkyl.
  • Ri is a linear substituted C 1 -C 15 alkyl.
  • Ri is a branched unsubstituted C 1 -C 15 alkyl.
  • Ri is a branched substituted C 1 -C 15 alkyl.
  • Ri is methyl.
  • Ri is a linear unsubstituted C 2 -C 15 alkenyl.
  • Ri is a linear substituted C 2 -C 15 alkenyl. According to some embodiments, Ri is a branched unsubstituted C 2 -C 15 alkenyl. According to some embodiments, Ri is a branched substituted C 2 -C 15 alkenyl. According to some embodiments, Ri is a linear unsubstituted C 2 -C 15 alkynyl. According to some embodiments, Ri is a linear substituted C 2 -C 15 alkynyl. According to some embodiments, Ri is a branched unsubstituted C 2 -C 15 alkynyl. According to some embodiments, Ri is a branched substituted C 2 -C 15 alkynyl. According to some embodiments, Ri is a branched substituted C 2 -C 15 alkynyl.
  • R 2 is a linear unsubstituted C 1 -C 15 alkyl. According to some embodiments, R 2 is a linear substituted C 1 -C 15 alkyl. According to some embodiments, R 2 is a branched unsubstituted C 1 -C 15 alkyl. According to some embodiments, R 2 is a branched substituted C 1 -C 15 alkyl. According to some embodiments, R 2 is a linear unsubstituted C 5 alkyl. According to some embodiments, R 2 is a linear unsubstituted C 2 -C 15 alkenyl. According to some embodiments, R 2 is a linear substituted C 2 -C 15 alkenyl.
  • R 2 is a branched unsubstituted C 2 -C 15 alkenyl. According to some embodiments, R 2 is a branched substituted C 2 -C 15 alkenyl. According to some embodiments, R 2 is a linear unsubstituted C 2 -C 15 alkynyl. According to some embodiments, R 2 is a linear substituted C 2 -C 15 alkynyl. According to some embodiments, R 2 is a branched unsubstituted C 2 -C 15 alkynyl. According to some embodiments, R 2 is a branched substituted C 2 -C 15 alkynyl.
  • R3 is hydrogen. According to some embodiments, R3 is -C(0)-CH3. According to some embodiments, R3 is -C(0)-C2H5. According to some embodiments, R3 is -C(0)-C 3 H 7 . According to some embodiments, R3 is -C(0)-C 4 H 9 . According to some embodiments, R3 is -C(0)-C 5 Hn.
  • R 4 is hydrogen. According to some embodiments, R 4 is -C(0)-CH 3 . According to some embodiments, R 4 is -C(0)-C 2 H 5 . According to some embodiments, R 4 is -C(0)-C 3 H 7 . According to some embodiments, R 4 is -C(0)-C 4 H 9 . According to some embodiments, R 4 is -C(0)-C 5 Hn.
  • R3 and R 4 are each hydrogen. According to some embodiments, one of R3 and R 4 is hydrogen and the other is -C(0)-CH 3 . According to some embodiments, R3 and R 4 are each -C(0)-CH 3 .
  • R 5 is -CH 3 . According to some embodiments, R 5 is -C 2 H 5 . According to some embodiments, R 5 is -C 3 H 7 . According to some embodiments, R 5 is -C 4 H 9 . According to some embodiments, R 5 is -C 5 H 11 . According to some embodiments, R 5 is a Ci- 15 alkyl. According to some embodiments, R 5 is a C 1-5 alkyl. According to some embodiments, R 5 is C 5-10 alkyl. According to some embodiments, R 5 is a Cio- 15 alkyl. According to some embodiments, the compound is represented by the structure of Formula (Ila),
  • the compound is represented by the structure of Formula (lib),
  • the compound is represented by the structure of Formula (lie),
  • the compound is represented by the structure of Formula (lid),
  • Formula (lid) and salts thereof According to some embodiments, the compound is represented by the structure of Formula (He), and salts thereof.
  • the compound is represented by the structure of Formula (Ilia),
  • the compound is represented by the structure of Formula (Mb) (designated herein EPM308),
  • the compound is represented by the structure of Formula (IIIc),
  • the compound is represented by the structure of Formula (Hid),
  • the compound is represented by the structure of Formula (Hie),
  • the compound is represented by the structure of Formula (IVa), Formula (IVa) and stereoisomers, and salts thereof.
  • the compound is represented by the structure of Formula (IVb) (designated herein EPM 313), and salts thereof.
  • the compound is represented by the structure of Formula (IVc),
  • the compound is represented by the structure of Formula (IVd), and salts thereof.
  • the compound is represented by the structure of Formula (IVe), and salts thereof.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective amount of a compound represented by the structure of Formula (la) as defined herein, and a pharmaceutically acceptable carrier or excipient.
  • the pharmaceutically acceptable carrier or excipient comprises at least one of a binder, a filler, a diluent, a surfactant or emulsifier, a glidant or lubricant, a buffering or pH adjusting agent, a tonicity enhancing agent, a wetting agent, a preservative, an antioxidant, a flavoring agent, a colorant, and a mixture or combinations thereof.
  • the pharmaceutical composition is in a form selected from the group consisting of tablet, pill, capsule, pellets, granules, powder, a wafer, coated or uncoated beads, lozenge, sachet, cachet, elixir, a depot system, a patch, suspension, dispersion, emulsion, solution, syrup, aerosol, oil, ointment, suppository, a gel, and a cream.
  • the pharmaceutical composition is formulated into a dosage form suitable for, oral, intranasal, intravenous, intraarterial, topical, intramuscular, transdermal, intraperitoneal, intrathecal, vaginal, rectal or subcutaneous administration.
  • the pharmaceutical composition is for use in the treatment of a disease, disorder or symptom amenable to treatment with cannabidiol (CBD).
  • CBD cannabidiol
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective amount of a compound represented by the structure of Formula (la), for use in the treatment of a disease, disorder or symptom amenable to treatment with cannabidiol (CBD),
  • Ri is selected from the group consisting of a hydrogen, a linear or branched unsubstituted or substituted C 1 -C 15 alkyl, a linear or branched unsubstituted or substituted C 2 -C 15 alkenyl, and a linear or branched unsubstituted or substituted C2-C15 alkynyl;
  • R 2 is selected from the group consisting of a linear or branched unsubstituted or substituted C 1 -C 15 alkyl, a linear or branched unsubstituted or substituted C 2 -C 15 alkenyl, and a linear or branched unsubstituted or substituted C 2 -C 15 alkynyl; each of R 3 and R 4 is independently selected from the group consisting of hydrogen and -C(0)-R 5 , wherein R 5 is a C 1 -C 15 alkyl; and stereoisomers, and salts thereof.
  • Ri is hydrogen. According to some embodiments, Ri is a linear unsubstituted C 1 -C 15 alkyl. According to some embodiments, Ri is methyl. According to some embodiments, R 2 is a linear unsubstituted C 1 -C 15 alkyl. According to some embodiments, R 2 is a linear unsubstituted C 5 alkyl.
  • R 3 and R 4 are each Hydrogen. According to some embodiments, one of R 3 and R 4 is Hydrogen and the other is -C(0)-CH 3 . According to some embodiments, R 3 and R 4 are each -C(0)-CH 3 . According to some embodiments, R 5 is -CH 3 .
  • the disease, disorder or symptom amenable to treatment with CBD is selected from the group consisting of pain, autoimmune disease, cancer, bacterial infection, impaired neurological function, inflammation, nausea, vomiting, convulsions, low appetite and glaucoma.
  • the pharmaceutical composition is for use in the treatment of pain.
  • the pharmaceutical composition is for use in the treatment of autoimmune disease. According to some embodiments, the pharmaceutical composition is for use in the treatment of cancer.
  • the pharmaceutical composition is for use in the treatment of bacterial infection.
  • the pharmaceutical composition is for use in the treatment of impaired neurological function.
  • the impaired neurological function is selected from the group consisting of stroke, trauma, Parkinson’s Disease, vascular dementia, senile dementia, Alzheimer’s disease, mild cognitive impairment, Huntington’s Disease, Amyotrophic lateral sclerosis (ALS), epilepsy, multiple sclerosis, and psychiatric disorders.
  • stroke trauma, Parkinson’s Disease, vascular dementia, senile dementia, Alzheimer’s disease, mild cognitive impairment, Huntington’s Disease, Amyotrophic lateral sclerosis (ALS), epilepsy, multiple sclerosis, and psychiatric disorders.
  • the impaired neurological function is epilepsy.
  • the impaired neurological function is a psychiatric disorder.
  • the psychiatric disorder is selected from the group consisting of depression, anxiety, acute or chronic stress, schizophrenia, panic attacks, ADHD, bipolar disorder, obsessive compulsive disorder and personality disorders. Each possibility represents a separate embodiment of the present invention.
  • the pharmaceutical composition is for use in the treatment of an inflammation.
  • the pharmaceutical composition is for use in treating joint inflammatory diseases and joint degeneration.
  • the pharmaceutical composition is for use in treating respiratory inflammation.
  • the pharmaceutical composition is for use in treating inflammatory bowel disease.
  • the pharmaceutical composition is for use in the treatment of nausea, vomiting or low appetite.
  • the pharmaceutical composition is for use in the treatment of convulsions.
  • the pharmaceutical composition is for use in the treatment of glaucoma.
  • the pharmaceutical composition is for use in the treatment of gastrointestinal diseases or disorders.
  • the disease, disorder or symptom amenable to treatment with CBD is selected from the group consisting of Non-Alcoholic Fatty Liver Disease (NAFLD), chronic kidney disease (CKD), obesity, hyperglycemia, diabetes, metabolic syndrome and/or obesity related diseases.
  • NAFLD Non-Alcoholic Fatty Liver Disease
  • CKD chronic kidney disease
  • obesity hyperglycemia
  • diabetes metabolic syndrome
  • obesity related diseases include obesity, hyperglycemia, diabetes, metabolic syndrome and/or obesity related diseases.
  • the pharmaceutical composition is for use in the treatment of obesity. According to some embodiments, the pharmaceutical composition is for use in the treatment of muscular dystrophy.
  • the compound is represented by the structure of Formula (Ila), Formula (Ila) and stereoisomers, and salts thereof.
  • the compound is represented by the structure of Formula (Ilia), Formula (Ilia) and stereoisomers, and salts thereof.
  • the compound is represented by the structure of Formula (IVa),
  • the pharmaceutical composition further comprising at least one additional active pharmaceutical ingredient.
  • the additional active pharmaceutical ingredient comprises at least one additional cannabinoid compound.
  • the additional cannabinoid compound is selected from the group consisting of cannabidiol (CBD), cannabigerol (CBG), D 8 - tetrahydrocannabinol (A S -THC), A 9 -tetrahydrocannabinol (A 9 -THC), cannabinol (CBN), A 9 (ll)-tetrahydrocannabinol (exo-THC), cannabichromene (CBC), tetrahydrocannabinol-C3 (THC-C3), tetrahydrocannabinol-C4 (THC-C4), tetrahydrocannabinol-C7 (THC-C7), esters thereof and combinations thereof.
  • CBD cannabidiol
  • CBG cannabigerol
  • the additional cannabinoid compound comprises a cannabigerolic acid (CBGA) ester represented by Formula (V), a cannabinolic acid (CBNA) ester represented by Formula (VI), a cannabidiolic acid (CBDA) ester represented by Formula (VII), a tetrahydrocannabinolic acid (THCA) ester represented by Formula (VIII) or any combination thereof,
  • CBDGA cannabigerolic acid
  • CBNA cannabinolic acid
  • VII cannabidiolic acid
  • THCA tetrahydrocannabinolic acid
  • each of R 6 , R 7 , Rs, R 9 , Rio, R 11 , R 12 and R 13 is independently selected from the group consisting of a linear or branched, unsubstituted or substituted C 1 -C 15 alkyl, a linear or branched, unsubstituted or substituted C 2 -C 15 alkenyl, and a linear or branched, unsubstituted or substituted C 2 -C 15 alkynyl; one of the dashed lines in Formula (VIII) represents a double bond and the other dashed line represents a single bond; and stereoisomers, and salts thereof.
  • Each one of the cannabigerolic acid (CBGA) ester represented by Formula (IV), the cannabinolic acid (CBNA) ester represented by Formula (V), the cannabidiolic acid (CBDA) ester represented by Formula (VI), and the tetrahydrocannabinolic acid (THCA) ester represented by Formula (VII) represents a separate embodiment of the present invention.
  • the at least one additional cannabinoid compound is cannabidiolic acid methyl ester (CBDA-ME).
  • the at least one additional cannabinoid compound is present in one or more extracts of a cannabis plant.
  • the cannabis plant extract is obtained from a strain selected from the group consisting of Cannabis sativa, Cannabis indica, Cannabis ruderalis, a hybrid strain, and combinations thereof.
  • the cannabis plant extract is obtained from a strain selected from the group consisting of a high-CBD strain, a high- THC strain, and a combination thereof.
  • the cannabis plant extract comprises at least one cannabinoid selected from the group consisting of cannabidiol (CBD), tetrahydrocannabinol (THC), cannabinol (CBN), cannabigerol (CBG), cannabichromene (CBC), acids thereof and combinations thereof.
  • CBD cannabidiol
  • THC tetrahydrocannabinol
  • CBN cannabinol
  • CBG cannabigerol
  • CBC cannabichromene
  • the cannabis plant extract comprises from about 1 to about 25% (w/w) of CBD. According to some embodiments, the cannabis plant extract comprises about 1% (w/w) of CBD. According to some embodiments, the cannabis plant extract comprises about 10% (w/w) of CBD. According to some embodiments, the cannabis plant extract comprises about 25% (w/w) of CBD.
  • the cannabis plant extract comprises from about 1 to about 25% (w/w) of THC. According to some embodiments, the cannabis plant extract comprises about 1% (w/w) of THC. According to some embodiments, the cannabis plant extract comprises about 10% (w/w) of THC. According to some embodiments, the cannabis plant extract comprises about 25% (w/w) of THC.
  • the present invention provides a method of treating a disease, disorder or symptom amenable to treatment with cannabidiol (CBD), the method comprising the step of administering to a subject in need thereof a pharmaceutical composition comprising a therapeutically effective amount of a compound represented by the structure of Formula (la) and a pharmaceutically acceptable carrier or excipient,
  • CBD cannabidiol
  • Ri is selected from the group consisting of a hydrogen, a linear or branched unsubstituted or substituted C 1 -C 15 alkyl, a linear or branched unsubstituted or substituted C 2 -C 15 alkenyl, and a linear or branched unsubstituted or substituted C 2 -C 15 alkynyl;
  • R 2 is selected from the group consisting of a linear or branched unsubstituted or substituted C 1 -C 15 alkyl, a linear or branched unsubstituted or substituted C 2 -C 15 alkenyl, and a linear or branched unsubstituted or substituted C 2 -C 15 alkynyl; each of R 3 and R 4 is independently selected from the group consisting of hydrogen and -C(0)-R 5 , wherein R 5 is a C 1 -C 15 alkyl; and stereoisomers, and salts thereof.
  • Ri is a hydrogen. According to some embodiments, wherein Ri is a linear unsubstituted C 1 -C 15 alkyl. According to some embodiments, Ri is Ci alkyl. According to some embodiments, Ri is methyl. According to some embodiments, R 2 is a linear unsubstituted C 1 -C 15 alkyl. According to some embodiments, wherein R 2 is a linear unsubstituted C 5 alkyl.
  • the compound is represented by the structure of Formula (Ila),
  • the compound is represented by the structure of Formula (Ilia),
  • the compound is represented by the structure of Formula (IVa),
  • the pharmaceutical composition is administered in combination with at least one additional active pharmaceutical ingredient, wherein the at least one additional active pharmaceutical ingredient comprises at least one additional cannabinoid compound.
  • the pharmaceutical composition comprises at least one additional active pharmaceutical ingredient, wherein the at least one additional active pharmaceutical ingredient comprises at least one additional cannabinoid compound.
  • compound represented by any one of the structure of Formulae (Ia)-(I-e), (Ila)-(II-e) or (Illa)-(III-e) is administered in combination with at least one additional active pharmaceutical ingredient, wherein the at least one additional active pharmaceutical ingredient comprises at least one additional cannabinoid compound.
  • the additional cannabinoid compound is selected from the group consisting of cannabidiol (CBD), cannabigerol (CBG), D 8 - tetrahydrocannabinol (A S -THC), A 9 -tetrahydrocannabinol (A 9 -THC), cannabinol (CBN), A 9 (ll)- tetrahydrocannabinol (exo-THC), cannabichromene (CBC), tetrahydrocannabinol-C3 (THC-C3), tetrahydrocannabinol-C4 (THC-C4), tetrahydrocannabinol-C7 (THC-C7), esters thereof and combination thereof.
  • CBD cannabidiol
  • CBG cannabigerol
  • a S -THC cannabinol
  • a 9 -THC cannabinol
  • CBN cannabinol
  • the additional cannabinoid compound comprises a cannabigerolic acid (CBGA) ester represented by Formula (V), a cannabinolic acid (CBNA) ester represented by Formula (VI), a cannabidiolic acid (CBDA) ester represented by Formula (VII), a tetrahydrocannabinolic acid (THCA) ester represented by Formula (VIII) or any combination thereof,
  • CBDGA cannabigerolic acid
  • CBNA cannabinolic acid
  • VII cannabidiolic acid
  • THCA tetrahydrocannabinolic acid
  • each of R 6 , R 7 , Rs, R 9 , Rio, R 11 , R 12 and R 13 is independently selected from the group consisting of a linear or branched, unsubstituted or substituted C 1 -C 15 alkyl, a linear or branched, unsubstituted or substituted C 2 -C 15 alkenyl, and a linear or branched, unsubstituted or substituted C 2 -C 15 alkynyl; one of the dashed lines in Formula (VIII) represents a double bond and the other dashed line represents a single bond; and stereoisomers, and salts thereof.
  • Figure 1 represents the 'H NMR spectra of EPM308.
  • the present invention provides novel cannabidiolic acid (CBDA) derivatives including acids and esters, and uses thereof.
  • CBDA cannabidiolic acid
  • the present invention further provides pharmaceutical compositions comprising CBDA derivatives, alone or in combination with one or more additional cannabinoid compounds, for use in treating various diseases, disorders and symptoms, including but not limited to pain, impaired neurological function, inflammation including respiratory inflammation, inflammatory bowel disease and autoimmune diseases, nausea, vomiting, convulsions, low appetite and glaucoma.
  • the present invention provides a compound represented by the structure of Formula (la),
  • Ri is selected from the group consisting of a hydrogen, a linear or branched unsubstituted or substituted C 1 -C 15 alkyl, a linear or branched unsubstituted or substituted C 2 -C 15 alkenyl, and a linear or branched unsubstituted or substituted C2-C15 alkynyl;
  • R 2 is selected from the group consisting of a linear or branched unsubstituted or substituted C 1 -C 15 alkyl, a linear or branched unsubstituted or substituted C 2 - Ci 5 alkenyl, and a linear or branched unsubstituted or substituted C 2 -C 15 alkynyl; each of R 3 and R 4 is independently selected from the group consisting of hydrogen and -C(0)-Rs, wherein R 5 is a C 1 -C 15 alkyl; and stereoisomers, and salts thereof.
  • the compound is represented by the structure of Formula (lb),
  • the compound is represented by the structure of Formula (Ic), Formula (Ic) wherein Ri, R 2 , R 3 and R 4 are defined as for Formula (la); and salts thereof.
  • the compound is represented by the structure of Formula (Id),
  • the compound is represented by the structure of Formula (lib),
  • the compound is represented by the structure of Formula (lid),
  • the compound is represented by the structure of Formula (He),
  • the compound is represented by the structure of Formula (Ilia),
  • the compound is represented by the structure of Formula (Mb) (designated herein EPM308), Formula (Mb) and salts thereof.
  • the compound is represented by the structure of Formula (IIIc), Formula (IIIc) and salts thereof.
  • the compound is represented by the structure of Formula (Hid),
  • the compound is represented by the structure of Formula (Me),
  • the compound is represented by the structure of Formula (IVa),
  • the compound is represented by the structure of Formula (IVb) (designated herein EPM 313),
  • the compound is represented by the structure of Formula (IVc),
  • the compound is represented by the structure of Formula (IVd),
  • the compound is represented by the structure of Formula (IVe),
  • Ri is hydrogen. According to some embodiments, Ri is a linear unsubstituted C 1 -C 15 alkyl. According to some embodiments, Ri is a linear unsubstituted C 2 alkyl. According to some embodiments, Ri is a linear unsubstituted C 3 alkyl. According to some embodiments, Ri is a linear unsubstituted Ci- C 4 alkyl. According to some embodiments, Ri is a linear unsubstituted C 5 alkyl. According to some embodiments, Ri is a linear substituted C 1 -C 15 alkyl. According to some embodiments, Ri is a branched unsubstituted C 1 -C 15 alkyl.
  • Ri is a branched substituted C 1 -C 15 alkyl. According to some embodiments, Ri is methyl. According to some embodiments, Ri is a linear unsubstituted C 2 -C 15 alkenyl. According to some embodiments, Ri is a linear substituted C 2 -C 15 alkenyl. According to some embodiments, Ri is a branched unsubstituted C 2 -C 15 alkenyl. According to some embodiments, Ri is a branched substituted C 2 -C 15 alkenyl. According to some embodiments, Ri is a linear unsubstituted C 2 -C 15 alkynyl.
  • Ri is a linear substituted C 2 -C 15 alkynyl. According to some embodiments, Ri is a branched unsubstituted C 2 -C 15 alkynyl. According to some embodiments, Ri is a branched substituted C 2 -C 15 alkynyl.
  • R 2 is a linear unsubstituted C 1 -C 15 alkyl. According to some embodiments, R 2 is a linear substituted C 1 -C 15 alkyl. According to some embodiments, R 2 is a branched unsubstituted C 1 -C 15 alkyl. According to some embodiments, R 2 is a branched substituted C 1 -C 15 alkyl. According to some embodiments, R 2 is a linear unsubstituted C 2 alkyl. According to some embodiments, R 2 is a linear unsubstituted C 3 alkyl. According to some embodiments, R 2 is a linear unsubstituted C 4 alkyl.
  • R 2 is a linear unsubstituted C 5 alkyl (n-pentyl). According to some embodiments, R2 is a linear unsubstituted Ce alkyl. According to some embodiments, R 2 is a linear unsubstituted C 2 -C 15 alkenyl. According to some embodiments, R 2 is a linear substituted C 2 -C 15 alkenyl. According to some embodiments, R 2 is a branched unsubstituted C 2 -C 15 alkenyl. According to some embodiments, R 2 is a branched substituted C 2 -C 15 alkenyl. According to some embodiments, R 2 is a linear unsubstituted C 2 -C 15 alkynyl. According to some embodiments, R 2 is a linear substituted C 2 -C 15 alkynyl. According to some embodiments, R 2 is a linear substituted C 2 -C 15 alkynyl. According to some embodiments, R 2 is a linear
  • R 2 is a branched unsubstituted C 2 -C 15 alkynyl. According to some embodiments, R 2 is a branched substituted C 2 -C 15 alkynyl.
  • R 3 is hydrogen. According to some embodiments, R 3 is -C(0)-CH 3 . According to some embodiments, R 3 is -C(0)-C 2 H 5 . According to some embodiments, R 3 is -C(0)-C 3 H 7 . According to some embodiments, R 3 is -C(0)-C 4 H 9 . According to some embodiments, R 3 is -C(0)-C 5 Hn.
  • R 4 is hydrogen. According to some embodiments, R 4 is -C(0)-CH 3 . According to some embodiments, R 4 is -C(0)-C 2 H 5 . According to some embodiments, R 4 is -C(0)-C 3 H 7 . According to some embodiments, R 4 is -C(0)-C 4 H 9 . According to some embodiments, R 4 is -C(0)-C 5 Hn.
  • R 3 and R 4 are each Hydrogen. According to some embodiments, one of R 3 and R 4 is Hydrogen and the other is -C(0)-CH 3 . According to some embodiments, R 3 and R 4 are each -C(0)CH 3 . According to some embodiments, R 5 is -CH 3 . According to some embodiments, R 5 is -C 2 H 5 . According to some embodiments, R 5 is -C 3 H 7 . According to some embodiments, R 5 is -C 4 H 9 . According to some embodiments, R 5 is -C 5 H 11 . According to some embodiments, R 5 is a Ci- 15 alkyl. According to some embodiments, R 5 is a C 1-5 alkyl. According to some embodiments, R 5 is C 5-10 alkyl. According to some embodiments, R 5 is a Cio- 15 alkyl.
  • the compounds provided herein may contain one or more chiral centers. Such chiral centers may each be of either of the ( R ) or (S) configuration. In case a compound of the invention contains more than one chiral center, each one of those chiral centers may be of the (R) or ( S ) configuration, independently. Thus, the compounds provided herein may be enantiomerically pure, or be stereoisomeric or diastereomeric mixtures.
  • alkyl refers to a saturated aliphatic hydrocarbon, including straight- chain also referred to as linear-chain, and branched-chain.
  • the alkyl group has 1-15 carbons designated here as Ci-Ci5-alkyl.
  • the alkyl group has 2-6 carbons designated here as C2-C6-alkyl.
  • the alkyl group has 2-4 carbons designated here as C2-C4-alkyl.
  • the alkyl group may be unsubstituted or substituted by one or more groups selected from the group consisting of hydroxyl, halogen, amino, thiol, phosphate, and combinations thereof.
  • unsubstituted or substituted alkyl refers to an alkyl group which is either unsubstituted (i.e. it is a hydrocarbon) or to an alkyl substituted with at least one of hydroxyl, halogen, amino, thiol, phosphate, and combinations thereof.
  • halo and halogen refer to the fluoro, chloro, bromo or iodo atoms. There can be one or more halogens, which are the same or different.
  • alkenyl refers to an aliphatic hydrocarbon group containing at least one carbon-carbon double bond including straight-chain, linear-chain, branched-chain and cyclic alkenyl groups.
  • the alkenyl group has 2-15 carbon atoms (a C2-15 alkenyl).
  • the alkenyl group has 2-4 carbon atoms in the chain (a C2-4 alkenyl).
  • alkenyl groups include, but are not limited to, ethenyl, propenyl, n-butenyl, i-butenyl, 3-methylbut-2-enyl, n-pentenyl, heptenyl, octenyl, cyclohexyl-butenyl and decenyl.
  • An alkylalkenyl is an alkyl group as defined herein bonded to an alkenyl group as defined herein.
  • the alkenyl group can be unsubstituted or substituted through available carbon atoms with one or more groups defined herein for alkyl.
  • unsubstituted or substituted alkenyl refers to an alkenyl group which is either unsubstituted (i.e. it is a hydrocarbon) or to an alkenyl substituted with at least one of hydroxyl, halogen, amino, thiol, phosphate, and combinations thereof.
  • alkynyl refers to an aliphatic hydrocarbon group containing at least one carbon-carbon triple bond including straight-chain or linear-chain and branched-chain.
  • the alkynyl group has 2-15 carbon atoms in the chain (a C2-15 alkynyl).
  • the alkynyl group has 2-4 carbon atoms in the chain (a C2-4 alkynyl).
  • alkynyl groups include, but are not limited to, ethynyl, propynyl, n- butynyl, 2-butynyl, 3-methylbutynyl, n-pentynyl, heptynyl, octynyl and decynyl.
  • An alkylalkynyl is an alkyl group as defined herein bonded to an alkynyl group as defined herein.
  • the alkynyl group can be unsubstituted or substituted through available carbon atoms with one or more groups defined herein for alkyl.
  • unsubstituted or substituted alkynyl refers to an alkynyl group which is either unsubstituted (i.e. it is a hydrocarbon) or to an alkynyl substituted with at least one of hydroxyl, halogen, amino, thiol, phosphate, and combinations thereof.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective amount of a compound represented by the structure of Formula (la) and a pharmaceutically acceptable carrier or excipient,
  • Ri is selected from the group consisting of a hydrogen, a linear or branched unsubstituted or substituted C 1 -C 15 alkyl, a linear or branched unsubstituted or substituted C 2 -C 15 alkenyl, and a linear or branched unsubstituted or substituted C2-C15 alkynyl;
  • R2 is selected from the group consisting of a linear or branched unsubstituted or substituted C 1 -C 15 alkyl, a linear or branched unsubstituted or substituted C 2 - Ci 5 alkenyl, and a linear or branched unsubstituted or substituted C 2 -C 15 alkynyl; each of R 3 and R 4 is independently selected from the group consisting of hydrogen and -C(0)-R 5 , wherein R 5 is a C 1 -C 15 alkyl; and stereoisomers, and salts thereof.
  • cannabinoid refers to any compound that interacts with cannabinoid receptors including endocannabinoids (produced naturally in the body by humans and animals), phytocannabinoids (found in cannabis and some other plants), and synthetic cannabinoids (manufactured artificially).
  • cannabinoid acid refers to the acid form of the above-mentioned cannabinoids.
  • CBD cannabidiol
  • CBD derivatives CBD may be obtained from industrial hemp extract with a trace amount of THC or from cannabis extract using high CBD cannabis cultivars.
  • cannabidiol may be obtained from plant extract, or may be prepared synthetically (manufactured artificially), the structure of CBD is presented below:
  • CBDA cannabidiolic acid
  • CBDA-ME cannabidiolic acid methyl ester
  • CBDA canbidiolic acid derivative
  • CBDA ester derivative refers to various molecules, which are the alkyl, alkenyl or alkynyl form of CBDA derivative as described herein.
  • Ri is selected from the group consisting of a linear or branched unsubstituted or substituted C 1 -C 15 alkyl, a linear or branched unsubstituted or substituted C 2 - Ci 5 alkenyl, and a linear or branched unsubstituted or substituted C 2 -C 15 alkynyl; each of R 3 and R 4 is independently selected from the group consisting of hydrogen and -C(0)-R 5 , wherein R 5 is -a Ci- 15 alkyl; and stereoisomers, and salts thereof.
  • CBDA cannabidiolic acid
  • derivatives of CBDA may include other substituents, in particular alkyl, alkenyl or alkynyl groups. Therefore, the term cannabidiolic acid derivative includes corresponding structures, in which position 4 is substituted by a group, which is either an n-CsHn or a different chemical group, in particular alkyl, alkenyl or alkynyl groups.
  • cannabidiolic acid and/or cannabidiolic acid ester should be interpreted broadly referring to all possible stereoconfigurations and salts of the relevant formula.
  • the present invention thus provides pharmaceutical compositions comprising the compounds disclosed herein and a pharmaceutically acceptable carrier and/or excipient.
  • the compounds of the present invention can be safely administered orally or non-orally. Routes of administration include, but are not limited to, oral, topical, mucosal, nasal, parenteral, gastrointestinal, intraspinal, intraperitoneal, intramuscular, intravenous, intrauterine, intraocular, intradermal, intracranial, intratracheal, intravaginal, intracerebroventricular, intracerebral, subcutaneous, ophthalmic, transdermal, rectal, buccal, epidural and sublingual. Each possibility represents a separate embodiment.
  • the pharmaceutical compositions can be formulated as tablets (including e.g. film-coated tablets), powders, granules, capsules (including soft capsules), orally disintegrating tablets, and sustained-release preparations as is well known in the art. Each possibility represents a separate embodiment.
  • the pharmaceutical composition is formulated into a dosage form suitable for, oral, intranasal, intravenous, intraarterial, topical, intramuscular, transdermal, intraperitoneal, intrathecal, vaginal, rectal or subcutaneous administration.
  • the pharmaceutical composition is formulated as tablet, pill, capsule (e.g. soft or hard gelatin capsule), pellets, granules, powder, a wafer, coated or uncoated beads, lozenge, sachet, cachet, elixir, a depot system, a patch, suspension, dispersion, emulsion, solution, syrup, aerosol, oil, ointment, suppository, a gel, and a cream.
  • a pharmaceutical composition is formulated as tablet, pill, capsule (e.g. soft or hard gelatin capsule), pellets, granules, powder, a wafer, coated or uncoated beads, lozenge, sachet, cachet, elixir, a depot system, a patch, suspension, dispersion, emulsion, solution, syrup, aerosol, oil, ointment, suppository, a gel, and a cream.
  • the active pharmaceutical ingredient is mixed with a pharmaceutical carrier or excipient to form a solid pre formulation composition containing a substantially homogeneous distribution of the compound disclosed herein in the pharmaceutical carrier or excipient.
  • solid dosage forms can be prepared by wet granulation, dry granulation, direct compression and the like as is known in the art.
  • the liquid forms in which the compositions disclosed herein may be incorporated, for administration via a route selected from oral, topical or by injection include aqueous solutions, suitably flavored syrups, aqueous or oil suspensions, and flavored emulsions with edible oils such as cottonseed oil, sesame oil, coconut oil, or peanut oil, as well as elixirs and similar pharmaceutical vehicles.
  • aqueous solutions suitably flavored syrups, aqueous or oil suspensions, and flavored emulsions with edible oils such as cottonseed oil, sesame oil, coconut oil, or peanut oil, as well as elixirs and similar pharmaceutical vehicles.
  • aqueous solutions suitably flavored syrups, aqueous or oil suspensions, and flavored emulsions with edible oils such as cottonseed oil, sesame oil, coconut oil, or peanut oil, as well as
  • the composition may be formulated as single phase aqueous, emulsion or multiple emulsions.
  • the composition is formulated as emulsion.
  • These emulsions may be oil-in-water (o/w) emulsions, water-in-oil (w/o) emulsions, or multiple emulsions such as oil-in-water-in-oil (o/w/o) or water-in-oil-in-water (w/o/w).
  • the oil phase can comprise silicone oils, non-silicone organic oils, or mixtures thereof.
  • the compositions can comprise two immiscible phases that are reconstituted prior to use. Each possibility represents a separate embodiment.
  • compositions for inhalation include solutions and suspensions in pharmaceutically acceptable aqueous or organic solvents, or mixtures thereof, and powders.
  • the liquid or solid compositions may contain suitable pharmaceutically acceptable carriers or excipients as described herein.
  • the compositions may be administered by the oral or nasal respiratory route.
  • Compositions may also be nebulized by use of inert gases. Nebulized solutions may be breathed directly from the nebulizing device or the nebulizing device may be attached to a face mask tent, or intermittent positive pressure breathing machine. Solution, suspension, or powder compositions may be administered, inter alia, orally or nasally, from devices that deliver the formulation in an appropriate manner.
  • the composition is prepared for topical administration, e.g. as an oil, ointment, gel or cream.
  • Adjuvants for topical administration may include, for example, sodium carboxymethylcellulose, polyacrylates, polyoxyethylene-polyoxypropylene-block polymers, polyethylene glycol and wood wax alcohols.
  • gel refers to a substantially dilute cross-linked system, which exhibits little or no flow when in the steady-state having a solid jelly-like matrix.
  • gel may comprise hydrogel, organogel, thermosensitive gel, non-thermosensitive gel, and aerogel. Each possibility represents a separate embodiment.
  • Hydrogels are highly hydrated, macromolecular networks, dispersed in water or other biological fluids. Hydrogels that exhibit the specific property of increased viscosity with increased temperatures are known as thermosensitive (or thermosetting) hydrogels. Such hydrogels have been shown to have easier application combined with longer survival periods at the site of application as compared to non-thermosensitive hydrogels, and are therefore advantageous as sustained-release drug delivery systems. Gel may be characterized by a critical gelation temperature, wherein gelation is effected at the critical gelation temperature or at temperatures above the critical gelation temperature. As used herein, “critical gelation temperature” refers to the lowest temperature at which some gelation of a material is observed (e.g., by increase in shear storage modulus).
  • the pharmaceutical composition can be formulated for local administration into the vagina.
  • the compositions may also be embedded in an article or administered using an applicator.
  • Suitable forms for vaginal administration include, but are not limited to, bandage, suppository, pessary, douche, ointment, tablet, tampon, foam, cream, gel, paste, microcapsules, vaginal sponge, vaginal ring, and syringe-like applicator. Each possibility represents a separate embodiment.
  • An optional carrier for vaginal administration comprises a lipid carrier, e.g. fatty acids, glycerin and pharmaceutically acceptable oils.
  • compositions disclosed herein may exhibit release mode which may be immediate release, controlled release or a mixture thereof. Each possibility represents a separate embodiment.
  • “Immediate release” (IR) compositions in the context of the present invention refers to compositions in which the active ingredient is released without delay following administration.
  • “Controlled release” (CR) compositions in the context of the present invention refers to compositions in which the active ingredient is released gradually over a period of time following administration.
  • Suitable pharmaceutically acceptable carriers or excipients include, but are not limited to, a binder, a filler, a diluent, a surfactant or emulsifier, a glidant or lubricant, buffering or pH adjusting agent, a tonicity enhancing agent, a wetting agent, a preservative, an antioxidant, a flavoring agent, a colorant, and a mixture or combination thereof.
  • a binder a filler, a diluent, a surfactant or emulsifier, a glidant or lubricant, buffering or pH adjusting agent, a tonicity enhancing agent, a wetting agent, a preservative, an antioxidant, a flavoring agent, a colorant, and a mixture or combination thereof.
  • Suitable binders include, but are not limited to, polyvinylpyrrolidone, copovidone, hydroxypropyl methylcellulose, starch, and gelatin. Each possibility represents a separate embodiment.
  • Suitable fillers include, but are not limited to, sugars such as lactose, sucrose, mannitol or sorbitol and derivatives therefore (e.g. amino sugars), ethylcellulose, microcrystalline cellulose, and silicified microcrystalline cellulose. Each possibility represents a separate embodiment.
  • Suitable lubricants include, but are not limited to, sodium stearyl fumarate, stearic acid, polyethylene glycol or stearates, such as magnesium stearate. Each possibility represents a separate embodiment.
  • Suitable diluents include, but are not limited to, dicalcium phosphate dihydrate, sugars, lactose, calcium phosphate, cellulose, kaolin, mannitol, sodium chloride, and dry starch. Each possibility represents a separate embodiment.
  • Suitable surfactants or emulsifiers include, but are not limited to, polyvinyl alcohol (PVA), polysorbate, polyethylene glycols, polyoxyethylene- polyoxypropylene block copolymers known as “poloxamer”, polyglycerin fatty acid esters such as decaglyceryl monolaurate and decaglyceryl monomyristate, sorbitan fatty acid ester such as sorbitan monostearate, polyoxyethylene sorbitan fatty acid ester such as polyoxyethylene sorbitan monooleate (Tween), polyethylene glycol fatty acid ester such as polyoxyethylene monostearate, polyoxyethylene alkyl ether such as polyoxyethylene lauryl ether, polyoxyethylene castor oil and hardened castor oil such as polyoxyethylene hardened castor oil.
  • PVA polyvinyl alcohol
  • polysorbate polyethylene glycols
  • Suitable glidants or lubricants include, but are not limited to, colloidal silicon dioxide, magnesium stearate, talc, and mineral oil. Each possibility represents a separate embodiment.
  • Suitable buffering or pH adjusting agent include, but are not limited to, acidic buffering agents such as short chain fatty acids, citric acid, acetic acid, hydrochloric acid, sulfuric acid and fumaric acid; and basic buffering agents such as tris, sodium carbonate, sodium bicarbonate, sodium hydroxide, potassium hydroxide and magnesium hydroxide. Each possibility represents a separate embodiment.
  • Suitable tonicity enhancing agents include, but are not limited to, ionic and non ionic agents such as, alkali metal or alkaline earth metal halides, urea, glycerol, sorbitol, mannitol, propylene glycol, and dextrose. Each possibility represents a separate embodiment.
  • Suitable wetting agents include, but are not limited to, glycerin, cetyl alcohol, and glycerol monostearate. Each possibility represents a separate embodiment.
  • Suitable preservatives include, but are not limited to, benzalkonium chloride, benzoxonium chloride, thiomersal, phenylmercuric nitrate, phenylmercuric acetate, phenylmercuric borate, methylparaben, propylparaben, chlorobutanol, benzyl alcohol, phenyl alcohol, chlorohexidine, and polyhexamethylene biguanide. Each possibility represents a separate embodiment.
  • Suitable antioxidants include, but are not limited to, sorbic acid, ascorbic acid, ascorbate, glycine, a-tocopherol, butylated hydroxyanisole (BHA), and butylated hydroxytoluene (BHT). Each possibility represents a separate embodiment.
  • Suitable flavoring agents include, but are not limited to, sweeteners such as sucralose and synthetic flavor oils and flavoring aromatics, natural oils, extracts from plants, leaves, flowers, and fruits, and combinations thereof.
  • Exemplary flavoring agents include cinnamon oils, oil of wintergreen, peppermint oils, clover oil, hay oil, anise oil, eucalyptus, vanilla, citrus oil such as lemon oil, orange oil, grape and grapefruit oil, and fruit essences including apple, peach, pear, strawberry, raspberry, cherry, plum, pineapple, and apricot. Each possibility represents a separate embodiment.
  • Suitable colorants include, but are not limited to, alumina (dried aluminum hydroxide), annatto extract, calcium carbonate, canthaxanthin, caramel, b-carotene, cochineal extract, carmine, potassium sodium copper chlorophyllin (chlorophyllin-copper complex), dihydroxy acetone, bismuth oxychloride, synthetic iron oxide, ferric ammonium ferrocyanide, ferric ferrocyanide, chromium hydroxide green, chromium oxide greens, guanine, mica-based pearlescent pigments, pyrophyllite, mica, dentifrices, talc, titanium dioxide, aluminum powder, bronze powder, copper powder, and zinc oxide.
  • alumina dried aluminum hydroxide
  • annatto extract calcium carbonate
  • canthaxanthin caramel
  • b-carotene cochineal extract
  • carmine potassium sodium copper chlorophyllin (chlorophyllin-copper complex)
  • dihydroxy acetone
  • solvate refers to a physical association of a compound disclosed herein with one or more solvent molecules. This physical association involves varying degrees of ionic and covalent bonding, including hydrogen bonding. In certain instances, the solvate will be capable of isolation. “Solvate” encompasses both solution-phase and isolatable solvates. Non-limiting examples of suitable solvates include ethanolates, methanolates and the like. A “hydrate” is a solvate in which the solvent molecule is water.
  • the present disclosure also includes any polymorphs thereof.
  • polymorph refers to a particular crystalline or amorphous state of a substance, which can be characterized by particular physical properties such as X-ray diffraction, electron diffraction, IR spectra, Raman spectra, melting point, and the like.
  • cannabinoids any of the cannabinoids disclosed herein, specifically, the abnormal cannabidiolic acids and their esters, can be prepared synthetically or semi-synthetically .
  • Natural cannabinoids may be isolated or extracted from a natural source or prepared by synthetic or semi- synthetic means.
  • cannabinoids can be isolated by extraction from cannabis plants. Plants in the cannabis genus include, but are not limited to, Cannabis sativa, Cannabis ruderalis, and Cannabis indica. Each possibility represents a separate embodiment. These plants are the natural sources of cannabinoids.
  • the pharmaceutical composition further comprising at least one additional active pharmaceutical ingredient.
  • the additional active pharmaceutical ingredient comprises at least one additional cannabinoid compound.
  • the additional cannabinoid compound is selected from the group consisting of cannabidiol (CBD), cannabigerol (CBG), D 8 - tetrahydrocannabinol (A S -THC), A 9 -tetrahydrocannabinol (A 9 -THC), cannabinol (CBN), A 9 (ll)-tetrahydrocannabinol (exo-THC), cannabichromene (CBC), tetrahydrocannabinol-C3 (THC-C3), tetrahydrocannabinol-C4 (THC-C4), tetrahydrocannabinol-C7 (THC-C7), esters thereof and combinations thereof.
  • CBD cannabidiol
  • CBG cannabigerol
  • a S -THC D 8 - tetrahydrocannabinol
  • a 9 -THC cannabinol
  • CBN can
  • the additional cannabinoid compound comprises a cannabigerolic acid (CBGA) ester represented by Formula (V), a cannabinolic acid (CBNA) ester represented by Formula (VI), a cannabidiolic acid (CBDA) ester represented by Formula (VII), a tetrahydrocannabinolic acid (THCA) ester represented by Formula (VIII) or any combination thereof,
  • each of R 6 , R 7 , Rs, R 9 , Rio, R 11 , R 12 and R 13 is independently selected from the group consisting of a linear or branched, unsubstituted or substituted C 1 -C 15 alkyl, a linear or branched, unsubstituted or substituted C 2 -C 15 alkenyl, and a linear or branched, unsubstituted or substituted C 2 -C 15 alkynyl; one of the dashed lines in Formula (VIII) represents a double bond and the other dashed line represents a single bond; and stereoisomers, and salts thereof.
  • the additional cannabinoid compound is cannabidiolic acid methyl ester.
  • the additional cannabinoid compound is present in one or more extracts of a cannabis plant.
  • the cannabis plant extract is obtained from a strain selected from the group consisting of Cannabis sativa, Cannabis indica, Cannabis ruderalis, a hybrid strain, and combinations thereof.
  • the cannabis plant extract is obtained from a strain selected from the group consisting of a high-CBD strain, a high-THC strain, and a combination thereof.
  • the cannabis plant extract comprises at least one cannabinoid selected from the group consisting of cannabidiol (CBD), tetrahydrocannabinol (THC), cannabinol (CBN), cannabigerol (CBG), cannabichromene (CBC), acids thereof and combinations thereof.
  • CBD cannabidiol
  • THC tetrahydrocannabinol
  • CBN cannabinol
  • CBG cannabigerol
  • CBC cannabichromene
  • the cannabis plant extract comprises from about 1 to about 25% (w/w) of CBD. According to some embodiments, the cannabis plant extract comprises about 1% (w/w) of CBD. According to some embodiments, the cannabis plant extract comprises about 10% (w/w) of CBD. According to some embodiments, the cannabis plant extract comprises about 25% (w/w) of CBD.
  • the cannabis plant extract comprises from about 1 to about 25% (w/w) of THC. According to some embodiments, the cannabis plant extract comprises about 1% (w/w) of THC. According to some embodiments, the cannabis plant extract comprises about 10% (w/w) of THC. According to some embodiments, the cannabis plant extract comprises about 25% (w/w) of THC.
  • extract refers a product prepared by extraction by physical means (e.g. by comminuting, pressing, heating, pulsed electric field assisted treatments, shear treatments and pressure wave treatments), by chemical means (e.g. by treatment with an acid, a base, a solvent) and/or by biochemical means (e.g. by treatment with hydrolytic enzymes, microorganisms).
  • the term refers to a liquid substance obtained through extraction from a given substance, or to a concentrate or essence which is free of, or substantially free of solvent.
  • extract may be a single extract obtained from a particular extraction step or series of extraction steps. Extract also may be a combination of extracts obtained from separate extraction steps or separate feedstocks.
  • extract Any methods of extraction with suitable solvent are encompassed. Exemplary extraction methods can be found for example in US patent 6,403,126, the contents of which are incorporated by reference herein.
  • the extract may be obtained from any part of the plant e.g. from leaves, flowers, stems, roots, fruits and seeds.
  • the extract may be aqueous or oily.
  • the cannabis plant extract is formed through contact with a suitable solvent or a combination of solvents.
  • the solvent is selected from the group consisting of a polar solvent, a hydrocarbon solvent, carbon dioxide, and a combination thereof.
  • extract further refers to a liquid or semi-solid or resinous substance obtained through extraction from plants defined in the present application, i.e. extracts obtained from cannabis plant e.g. Cannabis sativa, Cannabis indica, and Cannabis ruderalis.
  • the term refers to a mixture of liquid or semi-solid, resinous substances obtained through extraction from two or more different plants.
  • the term refers also to a compound purified from the extract.
  • extract has the meaning of a mixture or combination of two or more extracts.
  • cannabisbis extract refers to one or more plant extracts from the cannabis plant.
  • a cannabis extract contains, in addition to one or more cannabinoids, one or more non-cannabinoid components which are co-extracted with the cannabinoids from the plant material. Their respective ranges in weight will vary according to the starting plant material and the extraction methodology used.
  • Cannabinoid-containing plant extracts may be obtained by various means of extraction of cannabis plant material. Such means include but are not limited to supercritical or subcritical extraction with CO 2 , extraction with hot or cold gas and extraction with solvents.
  • the term refers to a mixture of liquid or semi-solid, resinous substances obtained through extraction from two or more different cannabis species.
  • the term refers also to a compound purified from the extract.
  • cannabis plant is a CBD-rich strain of cannabis plant or THC-rich strain of cannabis plant. Each possibility represents a separate embodiment.
  • hybrid strain refers to different strains of Cannabis which include differing amounts and/or ratios of the various cannabinoid compounds.
  • Cannabis sativa typically has a relatively high THC/CBD ratio.
  • Cannabis indica has a relative low THC/CBD ratio compared to Cannabis sativa (although the absolute amount of THC can be higher in Cannabis indica than in Cannabis sativa).
  • high-CBD strain and “CBD-rich strain” refer to a strain of cannabis plant which comprises CBD and optionally one or more additional cannabinoids, such as, for example, but not limited to: THC, CBN, and the like. According to some embodiments, CBD is the main component in the high-CBD strain.
  • the terms “high-THC strain” and “THC-rich strain” are directed to a strain of cannabis plant which comprises THC and optionally one or more additional cannabinoids, such as, for example, but not limited to: CBD, CBN, and the like.
  • THC is the main component in the high-THC strain.
  • the present invention provides a pharmaceutical composition comprising a therapeutically effective amount of a compound represented by the structure of Formula (la) and a pharmaceutically acceptable carrier or excipient, for use in the treatment of a disease, disorder or symptom amenable to treatment with CBD,
  • Ri is selected from the group consisting of a hydrogen, a linear or branched unsubstituted or substituted C 1 -C 15 alkyl, a linear or branched unsubstituted or substituted C 2 -C 15 alkenyl, and a linear or branched unsubstituted or substituted C2-C15 alkynyl;
  • R2 is selected from the group consisting of a linear or branched unsubstituted or substituted C 1 -C 15 alkyl, a linear or branched unsubstituted or substituted C 2 - Ci 5 alkenyl, and a linear or branched unsubstituted or substituted C 2 -C 15 alkynyl; each of R 3 and R 4 is independently selected from the group consisting of hydrogen and -C(0)-R 5 , wherein R 5 is a C 1 -C 15 alkyl; and stereoisomers, and salts thereof.
  • the disease, disorder or symptom amenable to treatment with cannabidiol is selected from the group consisting of pain, autoimmune disease, cancer, bacterial infection, impaired neurological function, inflammation, nausea, vomiting, convulsions, low appetite and glaucoma.
  • the composition disclosed herein is for use in the treatment of pain. According to some embodiments, the composition disclosed herein is for use in the attenuation of pain. According to some embodiments, the composition disclosed herein is for use in the alleviation of pain.
  • the composition disclosed herein is for use in the treatment of autoimmune disease. According to some embodiments, the composition disclosed herein is for use in the treatment of cancer. According to some embodiments, the composition disclosed herein is for use as an anti-tumor agent.
  • the composition disclosed herein is for use in the treatment of bacterial infection. According to some embodiments, the composition disclosed herein is for use as an anti-bacterial agent.
  • composition disclosed herein is for use in the treatment of impaired neurological function.
  • the composition disclosed herein is for use in the treatment of impaired neurological function selected from the group consisting of stroke, trauma, Parkinson’s disease, vascular dementia, senile dementia, Alzheimer’s disease, mild cognitive impairment, Huntington’s Disease, Amyotrophic lateral sclerosis (ALS), epilepsy, multiple sclerosis, and psychiatric disorders.
  • impaired neurological function selected from the group consisting of stroke, trauma, Parkinson’s disease, vascular dementia, senile dementia, Alzheimer’s disease, mild cognitive impairment, Huntington’s Disease, Amyotrophic lateral sclerosis (ALS), epilepsy, multiple sclerosis, and psychiatric disorders.
  • impaired neurological function selected from the group consisting of stroke, trauma, Parkinson’s disease, vascular dementia, senile dementia, Alzheimer’s disease, mild cognitive impairment, Huntington’s Disease, Amyotrophic lateral sclerosis (ALS), epilepsy, multiple sclerosis, and psychiatric disorders.
  • the impaired neurological function is epilepsy.
  • the composition disclosed herein is for use in the treatment of psychiatric disorders.
  • the psychiatric disorder is selected from the group consisting of depression, anxiety, acute or chronic stress, schizophrenia, panic attacks, ADHD, bipolar disorder, obsessive compulsive disorder or personality disorders.
  • the composition disclosed herein is for use in the treatment of depression. According to some embodiments, the composition disclosed herein is for use in the treatment of anxiety. According to some embodiments, the composition disclosed herein is for use in the treatment of acute or chronic stress. According to some embodiments, the composition disclosed herein is for use in the treatment of schizophrenia. According to some embodiments, the composition disclosed herein is for use in the treatment of panic attacks. According to some embodiments, the composition disclosed herein is for use in the treatment of ADHD. According to some embodiments, the composition disclosed herein is for use in the treatment of bipolar disorder. According to some embodiments, the composition disclosed herein is for use in the treatment of obsessive-compulsive disorder.
  • composition disclosed herein is for use in the treatment of inflammation.
  • the composition disclosed herein is for use in treating joint inflammatory diseases and joint degeneration.
  • the pharmaceutical composition is for use in treating respiratory inflammation.
  • the composition disclosed herein is for use in treating inflammatory bowel disease.
  • the inflammation is respiratory inflammation.
  • the respiratory inflammation is acute respiratory inflammation.
  • the respiratory inflammation can be associated with a disease or disorder including, but not limited to, asthma, chronic obstructive airway disorder, chronic obstructive pulmonary disease (COPD), pneumonia, respiratory syncytial viral infection, bronchitis, bronchiolitis, idiopathic pulmonary fibrosis, cystic fibrosis, acute respiratory distress syndrome (ARDS), bronchopulmonary dysplasia, occupational respiratory disease, particulate exposure, pleurisy, emphysema, and pulmonary edema.
  • COPD chronic obstructive pulmonary disease
  • COPD chronic obstructive pulmonary disease
  • pneumonia respiratory syncytial viral infection
  • bronchitis bronchiolitis
  • idiopathic pulmonary fibrosis cystic fibrosis
  • ARDS acute respiratory distress syndrome
  • bronchopulmonary dysplasia occupational respiratory disease, particulate exposure, pleurisy, emphysema, and pulmonary edema.
  • the respiratory inflammation is chronic respiratory inflammation.
  • the respiratory inflammation is pneumonitis.
  • the respiratory inflammation is asthma.
  • the respiratory inflammation is COPD.
  • the respiratory inflammation is caused by a pathogen.
  • the respiratory inflammation is caused by a pathogen selected from the group consisting of bacteria, viruses, parasites and fungi.
  • the respiratory inflammation is caused by a virus.
  • the respiratory inflammation is caused by Severe acute respiratory syndrome (SARS) vims.
  • the respiratory inflammation is caused by SARS-CoV-2 (Covid-19).
  • the composition disclosed herein is for use as a neuroprotective agent. According to some embodiments, the composition disclosed herein is for use in treating multiple sclerosis.
  • composition disclosed herein is for use in treating cancer.
  • the composition disclosed herein is for use in the treatment of a symptom selected from the group consisting of nausea, vomiting and low appetite. According to some embodiments, the composition disclosed herein is for use in the treatment of nausea. According to some embodiments, the composition disclosed herein is for use in the treatment of vomiting. According to some embodiments, the composition disclosed herein is for use in the treatment of low appetite.
  • composition disclosed herein is for use in the treatment of convulsions.
  • composition disclosed herein is for use in the treatment of glaucoma.
  • composition disclosed herein is for use in the treatment of gastrointestinal diseases or disorders.
  • composition disclosed herein is for use in the treatment of Non-Alcoholic Fatty Liver Disease (NAFLD), chronic kidney disease (CKD), obesity, hyperglycemia, diabetes, metabolic syndrome and/or obesity related diseases.
  • NAFLD Non-Alcoholic Fatty Liver Disease
  • CKD chronic kidney disease
  • obesity hyperglycemia
  • diabetes metabolic syndrome and/or obesity related diseases.
  • composition disclosed herein is for use in the treatment of obesity.
  • composition disclosed herein is for use in the treatment of muscular dystrophy.
  • the present invention provides a method of treating a disease, disorder or symptom amenable to treatment with cannabidiol (CBD), the method comprising administering to a subject in need thereof a pharmaceutical composition comprising a therapeutically effective amount of a compound represented by the structure of Formula (la) and a pharmaceutically acceptable carrier or excipient,
  • CBD cannabidiol
  • Ri is selected from the group consisting of a hydrogen, a linear or branched unsubstituted or substituted C 1 -C 15 alkyl, a linear or branched unsubstituted or substituted C 2 -C 15 alkenyl, and a linear or branched unsubstituted or substituted C2-C15 alkynyl;
  • R 2 is selected from the group consisting of a linear or branched unsubstituted or substituted C 1 -C 15 alkyl, a linear or branched unsubstituted or substituted C 2 - Ci 5 alkenyl, and a linear or branched unsubstituted or substituted C 2 -C 15 alkynyl; each of R 3 and R 4 is independently selected from the group consisting of hydrogen and -C(0)-R 5 , wherein R 5 is a C 1 -C 15 alkyl; and stereoisomers, and salts thereof.
  • the method of treating, a disease, disorder or symptom is selected from the group consisting of impaired neurological function, inflammation, nausea, vomiting, convulsions, low appetite, and glaucoma, the method comprising administering to a subject in need thereof the pharmaceutical composition disclosed herein.
  • the impaired neurological function is selected from the group consisting of stroke, trauma, Parkinson’ s disease, vascular dementia, senile dementia, Alzheimer’s disease, mild cognitive impairment, Huntington’s Disease, Amyotrophic lateral sclerosis (ALS), epilepsy, multiple sclerosis, and psychiatric disorders.
  • a method of treating, attenuating or alleviating pain comprising administering to a subject in need thereof the pharmaceutical composition disclosed herein.
  • a method of treating, attenuating or alleviating pain comprising administering to a subject in need thereof a pharmaceutical composition comprising a therapeutically effective amount of a compound represented by the structure of Formula (la) as described herein, and a pharmaceutically acceptable carrier or excipient.
  • the pharmaceutical composition is administered in combination with at least one additional active pharmaceutical ingredient as described herein.
  • Suitable additional active pharmaceutical ingredients include agents for pain relief such as non-steroidal anti-inflammatory agents (NSAIDs).
  • NSAIDs non-steroidal anti-inflammatory agents
  • Suitable NS A TPs include, but are not limited to, acetyl salicylic acid, indometacin, sulindac, phenylbutazone, diclofenac, fentiazac, ketorolac, piroxicam, tenoxicam, mecoxicam, meloxicam, cinnoxicam, ibufenac, ibuprofen, naproxen, ketoprofen, nabumetone, niflumic acid, nimesulide, and pharmaceutically acceptable salts thereof.
  • acetyl salicylic acid indometacin
  • sulindac phenylbutazone
  • diclofenac fentiazac
  • ketorolac piroxicam
  • tenoxicam mecoxicam
  • meloxicam
  • Cox-2 inhibitors suitable for use in the combinations of the present invention include, but are not limited to, (4-(4-cyclohexyl-2-methyloxazol-5-yl)-2-fluorobenzenesulfonamide), 5-chloro-3-(4-(methylsulfonyl)phenyl)-2-(methyl-5-pyridinyl)pyridine, 2-(3,5- difluorophenyl)-3-4 (methylsulfonyl)phenyl)-2-cyclopenten-l-one, 4-[5-(4- methylphenyl)-3-(trifluoromethyl)-lH-pyrazol-l-yl]-benzenesulfonamide, rofecoxib, (4- (4-methylsulfonyl)phenyl]-3-phenyl-2(5H)-furanone), 4- (5-methyl-3-phenylisoxazol-4
  • a method of treating psychiatric disorders comprising administering to a subject in need thereof the pharmaceutical composition disclosed herein.
  • a method of treating psychiatric disorders comprising administering to a subject in need thereof a pharmaceutical composition comprising a therapeutically effective amount of a compound represented by the structure of Formula (la) as described herein, and a pharmaceutically acceptable carrier or excipient.
  • the pharmaceutical composition is administered in combination with at least one additional active pharmaceutical ingredient as described herein.
  • a method of treating impaired neurological function comprising administering to a subject in need thereof the pharmaceutical composition disclosed herein.
  • a method of treating epilepsy comprising administering to a subject in need thereof the pharmaceutical composition disclosed herein.
  • a method of treating impaired neurological function comprising administering to a subject in need thereof a pharmaceutical composition comprising a therapeutically effective amount of a compound represented by the structure of Formula (la) as described herein, and a pharmaceutically acceptable carrier or excipient.
  • a method of treating epilepsy comprising administering to a subject in need thereof a pharmaceutical composition comprising a therapeutically effective amount of a compound ester represented by the structure of Formula (la) as described herein, and a pharmaceutically acceptable carrier or excipient.
  • the pharmaceutical composition is administered in combination with at least one additional active pharmaceutical ingredient as described herein.
  • a method of treating inflammation comprising administering to a subject in need thereof the pharmaceutical composition disclosed herein.
  • a method of treating inflammation comprising administering to a subject in need thereof a pharmaceutical composition comprising a therapeutically effective amount of a compound represented by the structure of Formula (la) as described herein, and a pharmaceutically acceptable carrier or excipient.
  • a method of treating joint inflammatory diseases and joint degeneration comprising administering to a subject in need thereof a pharmaceutical composition comprising a therapeutically effective amount of a compound represented by the structure of Formula (la) as described herein, and a pharmaceutically acceptable carrier or excipient.
  • a method of treating respiratory inflammation comprising administering to a subject in need thereof a pharmaceutical composition comprising a therapeutically effective amount of a compound represented by the structure of Formula (la) as described herein, and a pharmaceutically acceptable carrier or excipient.
  • a method of treating inflammatory bowel disease comprising administering to a subject in need thereof a pharmaceutical composition comprising a therapeutically effective amount of a compound represented by the structure of Formula (la) as described herein, and a pharmaceutically acceptable carrier or excipient.
  • the pharmaceutical composition is administered in combination with at least one additional active pharmaceutical ingredient as described herein.
  • a method of treating a symptom selected from the group consisting of nausea, vomiting and low appetite comprising administering to a subject in need thereof the pharmaceutical composition disclosed herein.
  • a symptom selected from the group consisting of nausea, vomiting and low appetite comprising administering to a subject in need thereof the pharmaceutical composition disclosed herein.
  • a method of treating a symptom selected from the group consisting of nausea, vomiting and low appetite comprising administering to a subject in need thereof a pharmaceutical composition comprising a therapeutically effective amount of a compound represented by the structure of Formula (la) as described herein, and a pharmaceutically acceptable carrier or excipient.
  • a pharmaceutical composition comprising a therapeutically effective amount of a compound represented by the structure of Formula (la) as described herein, and a pharmaceutically acceptable carrier or excipient.
  • the pharmaceutical composition is administered in combination with at least one additional active pharmaceutical ingredient as described herein.
  • the symptom is nausea.
  • the symptom is vomiting.
  • the symptom is low appetite.
  • a method of treating convulsions comprising administering to a subject in need thereof the pharmaceutical composition disclosed herein.
  • a method of treating convulsions comprising administering to a subject in need thereof a pharmaceutical composition comprising a therapeutically effective amount of a compound represented by the structure of Formula (la) as described herein, and a pharmaceutically acceptable carrier or excipient.
  • the pharmaceutical composition is administered in combination with at least one additional active pharmaceutical ingredient as described herein.
  • a method of treating glaucoma comprising administering to a subject in need thereof the pharmaceutical composition disclosed herein.
  • a method of treating glaucoma comprising administering to a subject in need thereof a pharmaceutical composition comprising a therapeutically effective amount of a compound represented by the structure of Formula (la) as described herein, and a pharmaceutically acceptable carrier or excipient.
  • the pharmaceutical composition is administered in combination with at least one additional active pharmaceutical ingredient as described herein.
  • composition comprising the compound of the present invention may be administered as the single therapeutic agent for the treatment of diseases or disorders described herein, combination therapy including co-administration of one or more additional active pharmaceutical ingredients is within the scope of the present invention.
  • Co-administration of the compound of the present invention with one or more therapeutic agents may take place sequentially in any order, simultaneously or a combination thereof.
  • administration of the compound of the present invention can take place prior to, after or at the same time as the administration of the additional therapeutic agent(s).
  • a total treatment period can be decided for the compound of the present invention.
  • the additional agent(s) can be administered prior to the onset of treatment with the compound of the present invention or following treatment with the compound of the present invention.
  • the additional agent(s) can be administered during the period of administering the compound of the present invention but does not need to occur over the entire treatment period.
  • the treatment regimen includes pre-treatment with one agent, followed by the addition of the other agent or agents.
  • Alternating sequences of administration are also contemplated. Alternating administration includes administration of the compound of the present invention, followed by the additional agent, followed by the compound of the present invention, etc.
  • the aforementioned sequences can also be administrated in several cycles wherein each cycle may be similar or different with each possibility representing a separate embodiment.
  • the therapeutic efficacy of the combination of the compound of the present invention and the additional agent(s) is at least additive.
  • the therapeutic efficacy is synergistic, namely the overall dose of each of the components may be lower, thus resulting in significantly lower side effects experienced by the subject, while a sufficient desirable therapeutic effect is nonetheless achieved.
  • the compound of the present invention and the additional therapeutic agent(s) may he provided in a single dosage form such as a fixed-dose combination or in separate compositions intended for simultaneous administration.
  • An optional dosage will be within the range of about 0.01-1000 mg/kg of body weight, about 0.1 mg/kg to 100 mg/kg, about 1 mg/kg to lOOmg/kg, about 10 mg/kg to 75 mg/kg, about 0.1 to 1 mg/kg etc., including each value within the specified range.
  • the composition comprises the compound of the present invention at a concentration of between 10 to 200 mg/ml.
  • the administration schedule will depend on several factors such as the severity and progression of the disease, disorder or symptom.
  • the compositions of the invention can be taken once-daily, twice-daily, thrice daily, once-weekly or once- monthly.
  • the administration can be continuous, i.e., every day, or intermittently.
  • intermittent administration can be administration one to six days per week or it may mean administration in cycles (e.g. daily administration for two to eight consecutive weeks, then a rest period with no administration for up to one week) or it may mean administration on alternate days.
  • intermittent administration can be administration one to six days per week or it may mean administration in cycles (e.g. daily administration for two to eight consecutive weeks, then a rest period with no administration for up to one week) or it may mean administration on alternate days.
  • the effectiveness of said compositions could enable a shortened period of treatment with superior results.
  • a “therapeutically effective amount” as used herein refers to an amount of an agent which is effective, upon single or multiple dose administration to the subject in providing a therapeutic benefit to the subject.
  • the compounds of the present invention are used for the preparation of a medicament for treating any one of the diseases, disorders, conditions or symptoms disclosed herein.
  • CBDA-ME (1.2gr) was dissolved in 30 ml dry pyridine, a catalytic amount of DMAP and 4.5ml of acetic anhydride was added. After lh stirring at room temperature under nitrogen full conversion is observed, the reaction was stopped by adding ice-cold HC1 till pH 1, the aqueous layer was washed with diethyl ether 5 times, then the combined ether phase washed with sodium bicarbonate and brine and dryer over MgS04. Fast purification on silica gel provided around 95% yield of the product.
  • mice Two groups of mice are treated, each group with a different compound at different doses ninety (90) minutes before the hot plate test.
  • the percent change in response time (latency) is calculated by comparing the mean of the control values with the mean of the test values and statistical significance determined by a paired t test.
  • EPM313 and EPM308 are tested in an acute experimental epilepsy model in pentylenetetrazole (PTZ) injected rats as described in Obay et. al. (Peptides, 2007, 28, 1214-1219). Briefly, adult male Wistar albino rats are divided into a control group, and seven experimental groups with seven rats in each group. In order to generate epileptic seizures, PTZ (50 mg/kg) is injected intraperitoneally. The experimental groups receive intraperitoneal injections, each group treated with a different compound at different doses 30 min before PTZ injection. After PTZ injection, the rats are kept in cages and their behaviors are observed for 30 min. The latencies are separated into three components: first myoclonic jerk (FMJ), generalized clonic seizures (GCS) and tonic generalized extension (TGE).
  • FMJ myoclonic jerk
  • GCS generalized clonic seizures
  • TGE tonic generalized extension

Abstract

The present invention provides novel compounds comprising cannabidiolic acid (CBDA) derivatives, including acids and esters, and uses thereof in the treatment of various diseases, disorders, conditions and symptoms.

Description

CANNABIDIOLIC ACID (CBDA) DERIVATIVES AND USES THEREOF
FIELD OF THE INVENTION
The present invention relates to cannabidiolic acid (CBDA) derivatives, including acids and esters, and uses thereof in the treatment of various diseases, disorders, conditions and symptoms.
BACKGROUND OF THE INVENTION
Cannabidiol (CBD) is the major non-psycho tropic phytocannabinoid compound in the plant Cannabis sativa, making up to 40% of the cannabinoids in Cannabis extracts (Grlic, Bull. Narc., 1976, 14:37-46). CBD is considered a lead compound for treating and preventing inflammatory and oxidative damage, see for example WO 1999/053917.
Figure imgf000002_0001
Cannabidiolic acid (CBDA) is also a major constituent of the Cannabis sativa plant and was first isolated in 1955 (Krejci and Santavy, 1955, Acta Univ Palacki Olomuc 6:59- 66). Its structure was elucidated in 1965 by analysis of the physical properties of its methyl ester, cannabidiolic acid methyl ester (CBDA-ME) (Mechoulam and Gaoni, Tetrahedron, 1965, 21:1223-1229). Its synthesis from cannabidiol was subsequently reported (Mechoulam and Ben-Zvi, J. Chem. Soc. Commun., 1969, 7:343-344).
The cannabinoid acids are precursors of the natural cannabinoids (Potter et al., J. Forensic Sci., 2008, 53:90-94), potentially lowering the amount of drug required to induce effects. The decarboxylation of CBDA into CBD is enhanced by heat, indicating the relative instability of CBDA, thus lowering its potential to serve as a medication (Mechoulam, Academic Press, New York, 1973, 1-99; Citti et al., J. Pharm. Biomed. Anal., 2018, 16:532-540). Thus, CBDA-ME is a relatively unknown cannabinoid and remains understudied and its effects are only just starting to become clear.
Figure imgf000003_0001
CBDA-ME is a stable derivative of CBDA and can be pharmacological active in vivo. Pertwee et al. (Brit. J. Pharmacology, 2018, 175:100-112) reported that the methyl ester of CBDA, designated HU-580 (also denoted herein EPM-301), displays a greater potency than CBDA in suppressing signs of both acute and anticipatory nausea, and of stress-induced anxiety in rats, and that it produces these effects in a 5-HTIA receptor- dependent manner. Another recent study (Hen-Shoval et al., Behav. Brain Res., 2018, 351:1-3) provides support for a potent anti-depressant effect after oral ingestion of a low dose (1 mg/kg) of HU-580 in two rat models.
WO 2018/235079 discloses compositions comprising CBDA esters and uses thereof in the treatment of a condition, disease or symptom associated with 5-HTIA receptors.
There is still an unmet medical need for effective therapies for use of non psychoactive cannabinoids, particularly derivatives of CBD with improved properties and decreased potential for detrimental activity. These derivatives will be advantageous in the treatment of various diseases, conditions, disorders and symptoms, such as pain, impaired neurological functions including depression and anxiety, and inflammation.
SUMMARY OF THE INVENTION
The present invention provides novel compounds comprising cannabidiolic acid (CBDA) derivatives, including acid and esters, and uses thereof. The present invention further provides pharmaceutical compositions comprising CBDA derivatives, alone or in combination with one or more additional cannabinoid compounds, for use in treating various diseases, disorders and symptoms, including but not limited to pain, impaired neurological function, inflammation including respiratory inflammation, inflammatory bowel disease and autoimmune diseases, nausea, vomiting, convulsions, low appetite and glaucoma.
It is now disclosed, unexpectedly, that compositions comprising cannabidiolic acid
(CBDA) or CBDA ester derivatives exhibit prolonged and significant therapeutic effects compared to cannabidiol (CBD). Advantageously, the pharmaceutical compositions disclosed herein are non-psycho active. According to some embodiments the present invention provides pharmaceutical compositions comprising CBDA derivatives having superior pharmacokinetics compared to the commercially available cannabidiol (CBD). According to one aspect, the present invention provides a compound represented by the structure of Formula (la),
Figure imgf000004_0001
Formula (la) wherein
Ri is selected from the group consisting of a hydrogen, a linear or branched unsubstituted or substituted C1-C15 alkyl, a linear or branched unsubstituted or substituted C2-C15 alkenyl, and a linear or branched unsubstituted or substituted C2-C15 alkynyl;
R2 is selected from the group consisting of a linear or branched unsubstituted or substituted C1-C15 alkyl, a linear or branched unsubstituted or substituted C2- Ci5 alkenyl, and a linear or branched unsubstituted or substituted C2-C15 alkynyl; each of R3 and R4 is independently selected from the group consisting of hydrogen and -C(0)-R5, wherein R5 is a C1-C15 alkyl; and stereoisomers, and salts thereof. According to some embodiments, the compound is represented by the structure of
Formula (lb),
Figure imgf000004_0002
Formula (lb) wherein Ri, R2, R3 and R4 are as defined for Formula (la); and salts thereof. According to some embodiments, the compound is represented by the structure of Formula (Ic),
Figure imgf000005_0001
Formula (Ic) wherein Ri, R2, R3 and R4 are defined as for Formula (la); and salts thereof. According to some embodiments, the compound is represented by the structure of
Formula (Id),
Figure imgf000005_0002
Formula (Id) wherein Ri, R2, R3 and R4 are defined as for Formula (la); and salts thereof. According to some embodiments, the compound is represented by the structure of
Formula (Ie),
Figure imgf000005_0003
Formula (Ie) wherein Ri, R2, R3 and R4 are defined as for Formula (la); and salts thereof. According to some embodiments, Ri is hydrogen. According to some embodiments, Ri is a linear unsubstituted C1-C15 alkyl. According to some embodiments, Ri is a linear substituted C1-C15 alkyl. According to some embodiments, Ri is a branched unsubstituted C1-C15 alkyl. According to some embodiments, Ri is a branched substituted C1-C15 alkyl. According to some embodiments, Ri is methyl. According to some embodiments, Ri is a linear unsubstituted C2-C15 alkenyl. According to some embodiments, Ri is a linear substituted C2-C15 alkenyl. According to some embodiments, Ri is a branched unsubstituted C2-C15 alkenyl. According to some embodiments, Ri is a branched substituted C2-C15 alkenyl. According to some embodiments, Ri is a linear unsubstituted C2-C15 alkynyl. According to some embodiments, Ri is a linear substituted C2-C15 alkynyl. According to some embodiments, Ri is a branched unsubstituted C2-C15 alkynyl. According to some embodiments, Ri is a branched substituted C2-C15 alkynyl.
According to some embodiments, R2 is a linear unsubstituted C1-C15 alkyl. According to some embodiments, R2 is a linear substituted C1-C15 alkyl. According to some embodiments, R2 is a branched unsubstituted C1-C15 alkyl. According to some embodiments, R2 is a branched substituted C1-C15 alkyl. According to some embodiments, R2 is a linear unsubstituted C5 alkyl. According to some embodiments, R2 is a linear unsubstituted C2-C15 alkenyl. According to some embodiments, R2 is a linear substituted C2-C15 alkenyl. According to some embodiments, R2 is a branched unsubstituted C2-C15 alkenyl. According to some embodiments, R2 is a branched substituted C2-C15 alkenyl. According to some embodiments, R2 is a linear unsubstituted C2-C15 alkynyl. According to some embodiments, R2 is a linear substituted C2-C15 alkynyl. According to some embodiments, R2 is a branched unsubstituted C2-C15 alkynyl. According to some embodiments, R2 is a branched substituted C2-C15 alkynyl.
According to some embodiments, R3 is hydrogen. According to some embodiments, R3 is -C(0)-CH3. According to some embodiments, R3 is -C(0)-C2H5. According to some embodiments, R3 is -C(0)-C3H7. According to some embodiments, R3 is -C(0)-C4H9. According to some embodiments, R3 is -C(0)-C5Hn.
According to some embodiments, R4 is hydrogen. According to some embodiments, R4 is -C(0)-CH3. According to some embodiments, R4 is -C(0)-C2H5. According to some embodiments, R4 is -C(0)-C3H7. According to some embodiments, R4 is -C(0)-C4H9. According to some embodiments, R4 is -C(0)-C5Hn.
According to some embodiments, R3 and R4 are each hydrogen. According to some embodiments, one of R3 and R4 is hydrogen and the other is -C(0)-CH3. According to some embodiments, R3 and R4 are each -C(0)-CH3.
According to some embodiments, R5 is -CH3. According to some embodiments, R5 is -C2H5. According to some embodiments, R5 is -C3H7. According to some embodiments, R5 is -C4H9. According to some embodiments, R5 is -C5H11. According to some embodiments, R5 is a Ci-15 alkyl. According to some embodiments, R5 is a C1-5 alkyl. According to some embodiments, R5 is C5-10 alkyl. According to some embodiments, R5 is a Cio-15 alkyl. According to some embodiments, the compound is represented by the structure of Formula (Ila),
Figure imgf000007_0001
Formula (Ila) and stereoisomers, and salts thereof.
According to some embodiments, the compound is represented by the structure of Formula (lib),
Figure imgf000007_0002
Formula (lib) and salts thereof.
According to some embodiments, the compound is represented by the structure of Formula (lie),
Figure imgf000007_0003
Formula (lie) and salts thereof.
According to some embodiments, the compound is represented by the structure of Formula (lid),
Figure imgf000007_0004
Formula (lid) and salts thereof. According to some embodiments, the compound is represented by the structure of Formula (He),
Figure imgf000008_0001
and salts thereof.
According to some embodiments, the compound is represented by the structure of Formula (Ilia),
Figure imgf000008_0002
Formula (Ilia) and stereoisomers, and salts thereof.
According to some embodiments, the compound is represented by the structure of Formula (Mb) (designated herein EPM308),
Figure imgf000008_0003
Formula (Mb) and salts thereof.
According to some embodiments, the compound is represented by the structure of Formula (IIIc),
Figure imgf000009_0001
Formula (IIIc) and salts thereof.
According to some embodiments, the compound is represented by the structure of Formula (Hid),
Figure imgf000009_0002
Formula (Hid) and salts thereof.
According to some embodiments, the compound is represented by the structure of Formula (Hie),
Figure imgf000009_0003
Formula (Me) and salts thereof.
According to some embodiments, the compound is represented by the structure of Formula (IVa), Formula (IVa) and stereoisomers, and salts thereof.
According to some embodiments, the compound is represented by the structure of Formula (IVb) (designated herein EPM 313),
Figure imgf000010_0001
and salts thereof.
According to some embodiments, the compound is represented by the structure of Formula (IVc),
Figure imgf000010_0002
Formula (IVc) and salts thereof.
According to some embodiments, the compound is represented by the structure of Formula (IVd),
Figure imgf000010_0003
and salts thereof.
According to some embodiments, the compound is represented by the structure of Formula (IVe),
Figure imgf000011_0001
and salts thereof.
According to another aspect, the present invention provides a pharmaceutical composition comprising a therapeutically effective amount of a compound represented by the structure of Formula (la) as defined herein, and a pharmaceutically acceptable carrier or excipient.
According to some embodiments, the pharmaceutically acceptable carrier or excipient comprises at least one of a binder, a filler, a diluent, a surfactant or emulsifier, a glidant or lubricant, a buffering or pH adjusting agent, a tonicity enhancing agent, a wetting agent, a preservative, an antioxidant, a flavoring agent, a colorant, and a mixture or combinations thereof.
According to some embodiments, the pharmaceutical composition is in a form selected from the group consisting of tablet, pill, capsule, pellets, granules, powder, a wafer, coated or uncoated beads, lozenge, sachet, cachet, elixir, a depot system, a patch, suspension, dispersion, emulsion, solution, syrup, aerosol, oil, ointment, suppository, a gel, and a cream.
According to some embodiments, the pharmaceutical composition is formulated into a dosage form suitable for, oral, intranasal, intravenous, intraarterial, topical, intramuscular, transdermal, intraperitoneal, intrathecal, vaginal, rectal or subcutaneous administration.
According to some embodiments, the pharmaceutical composition is for use in the treatment of a disease, disorder or symptom amenable to treatment with cannabidiol (CBD).
According to another aspect, the present invention provides a pharmaceutical composition comprising a therapeutically effective amount of a compound represented by the structure of Formula (la), for use in the treatment of a disease, disorder or symptom amenable to treatment with cannabidiol (CBD),
Figure imgf000012_0001
Formula (la) wherein
Ri is selected from the group consisting of a hydrogen, a linear or branched unsubstituted or substituted C1-C15 alkyl, a linear or branched unsubstituted or substituted C2-C15 alkenyl, and a linear or branched unsubstituted or substituted C2-C15 alkynyl;
R2 is selected from the group consisting of a linear or branched unsubstituted or substituted C1-C15 alkyl, a linear or branched unsubstituted or substituted C2-C15 alkenyl, and a linear or branched unsubstituted or substituted C2-C15 alkynyl; each of R3 and R4 is independently selected from the group consisting of hydrogen and -C(0)-R5, wherein R5 is a C1-C15 alkyl; and stereoisomers, and salts thereof.
According to some embodiments, Ri is hydrogen. According to some embodiments, Ri is a linear unsubstituted C1-C15 alkyl. According to some embodiments, Ri is methyl. According to some embodiments, R2 is a linear unsubstituted C1-C15 alkyl. According to some embodiments, R2 is a linear unsubstituted C5 alkyl.
According to some embodiments, R3 and R4 are each Hydrogen. According to some embodiments, one of R3 and R4 is Hydrogen and the other is -C(0)-CH3. According to some embodiments, R3 and R4 are each -C(0)-CH3. According to some embodiments, R5 is -CH3.
According to some embodiments, the disease, disorder or symptom amenable to treatment with CBD is selected from the group consisting of pain, autoimmune disease, cancer, bacterial infection, impaired neurological function, inflammation, nausea, vomiting, convulsions, low appetite and glaucoma.
According to some embodiments, the pharmaceutical composition is for use in the treatment of pain.
According to some embodiments, the pharmaceutical composition is for use in the treatment of autoimmune disease. According to some embodiments, the pharmaceutical composition is for use in the treatment of cancer.
According to some embodiments, the pharmaceutical composition is for use in the treatment of bacterial infection.
According to some embodiments, the pharmaceutical composition is for use in the treatment of impaired neurological function.
According to some embodiments, the impaired neurological function is selected from the group consisting of stroke, trauma, Parkinson’s Disease, vascular dementia, senile dementia, Alzheimer’s disease, mild cognitive impairment, Huntington’s Disease, Amyotrophic lateral sclerosis (ALS), epilepsy, multiple sclerosis, and psychiatric disorders. Each possibility represents a separate embodiment of the present invention.
According to some embodiments, the impaired neurological function is epilepsy.
According to some embodiments, the impaired neurological function is a psychiatric disorder.
According to some embodiments, the psychiatric disorder is selected from the group consisting of depression, anxiety, acute or chronic stress, schizophrenia, panic attacks, ADHD, bipolar disorder, obsessive compulsive disorder and personality disorders. Each possibility represents a separate embodiment of the present invention.
According to some embodiments, the pharmaceutical composition is for use in the treatment of an inflammation. According to some embodiments, the pharmaceutical composition is for use in treating joint inflammatory diseases and joint degeneration. According to additional embodiments, the pharmaceutical composition is for use in treating respiratory inflammation. According to additional embodiments, the pharmaceutical composition is for use in treating inflammatory bowel disease.
According to some embodiments, the pharmaceutical composition is for use in the treatment of nausea, vomiting or low appetite.
According to some embodiments, the pharmaceutical composition is for use in the treatment of convulsions.
According to some embodiments, the pharmaceutical composition is for use in the treatment of glaucoma.
According to some embodiments, the pharmaceutical composition is for use in the treatment of gastrointestinal diseases or disorders.
According to some embodiments, the disease, disorder or symptom amenable to treatment with CBD is selected from the group consisting of Non-Alcoholic Fatty Liver Disease (NAFLD), chronic kidney disease (CKD), obesity, hyperglycemia, diabetes, metabolic syndrome and/or obesity related diseases.
According to some embodiments, the pharmaceutical composition is for use in the treatment of obesity. According to some embodiments, the pharmaceutical composition is for use in the treatment of muscular dystrophy.
According to some embodiments, the compound is represented by the structure of Formula (Ila),
Figure imgf000014_0001
Formula (Ila) and stereoisomers, and salts thereof.
According to some embodiments, the compound is represented by the structure of Formula (Ilia),
Figure imgf000014_0002
Formula (Ilia) and stereoisomers, and salts thereof.
According to some embodiments, the compound is represented by the structure of Formula (IVa),
Figure imgf000014_0003
Formula (IVa) and stereoisomers, and salts thereof. According to some embodiments, the pharmaceutical composition further comprising at least one additional active pharmaceutical ingredient.
According to some embodiments, the additional active pharmaceutical ingredient comprises at least one additional cannabinoid compound. According to some embodiments, the additional cannabinoid compound is selected from the group consisting of cannabidiol (CBD), cannabigerol (CBG), D8- tetrahydrocannabinol (AS-THC), A9-tetrahydrocannabinol (A9-THC), cannabinol (CBN), A9(ll)-tetrahydrocannabinol (exo-THC), cannabichromene (CBC), tetrahydrocannabinol-C3 (THC-C3), tetrahydrocannabinol-C4 (THC-C4), tetrahydrocannabinol-C7 (THC-C7), esters thereof and combinations thereof.
According to some embodiments, the additional cannabinoid compound comprises a cannabigerolic acid (CBGA) ester represented by Formula (V), a cannabinolic acid (CBNA) ester represented by Formula (VI), a cannabidiolic acid (CBDA) ester represented by Formula (VII), a tetrahydrocannabinolic acid (THCA) ester represented by Formula (VIII) or any combination thereof,
Figure imgf000015_0001
Formula (VIII) wherein each of R6, R7, Rs, R9, Rio, R11, R12 and R13 is independently selected from the group consisting of a linear or branched, unsubstituted or substituted C1-C15 alkyl, a linear or branched, unsubstituted or substituted C2-C15 alkenyl, and a linear or branched, unsubstituted or substituted C2-C15 alkynyl; one of the dashed lines in Formula (VIII) represents a double bond and the other dashed line represents a single bond; and stereoisomers, and salts thereof. Each one of the cannabigerolic acid (CBGA) ester represented by Formula (IV), the cannabinolic acid (CBNA) ester represented by Formula (V), the cannabidiolic acid (CBDA) ester represented by Formula (VI), and the tetrahydrocannabinolic acid (THCA) ester represented by Formula (VII) represents a separate embodiment of the present invention.
According to some embodiments, the at least one additional cannabinoid compound is cannabidiolic acid methyl ester (CBDA-ME).
According to some embodiments, the at least one additional cannabinoid compound is present in one or more extracts of a cannabis plant. According to some embodiments, the cannabis plant extract is obtained from a strain selected from the group consisting of Cannabis sativa, Cannabis indica, Cannabis ruderalis, a hybrid strain, and combinations thereof. According to some embodiments, the cannabis plant extract is obtained from a strain selected from the group consisting of a high-CBD strain, a high- THC strain, and a combination thereof.
According to some embodiments, the cannabis plant extract comprises at least one cannabinoid selected from the group consisting of cannabidiol (CBD), tetrahydrocannabinol (THC), cannabinol (CBN), cannabigerol (CBG), cannabichromene (CBC), acids thereof and combinations thereof.
According to some embodiments, the cannabis plant extract comprises from about 1 to about 25% (w/w) of CBD. According to some embodiments, the cannabis plant extract comprises about 1% (w/w) of CBD. According to some embodiments, the cannabis plant extract comprises about 10% (w/w) of CBD. According to some embodiments, the cannabis plant extract comprises about 25% (w/w) of CBD.
According to some embodiments, the cannabis plant extract comprises from about 1 to about 25% (w/w) of THC. According to some embodiments, the cannabis plant extract comprises about 1% (w/w) of THC. According to some embodiments, the cannabis plant extract comprises about 10% (w/w) of THC. According to some embodiments, the cannabis plant extract comprises about 25% (w/w) of THC.
According to another aspect, the present invention provides a method of treating a disease, disorder or symptom amenable to treatment with cannabidiol (CBD), the method comprising the step of administering to a subject in need thereof a pharmaceutical composition comprising a therapeutically effective amount of a compound represented by the structure of Formula (la) and a pharmaceutically acceptable carrier or excipient,
Figure imgf000017_0001
Formula (la) wherein
Ri is selected from the group consisting of a hydrogen, a linear or branched unsubstituted or substituted C1-C15 alkyl, a linear or branched unsubstituted or substituted C2-C15 alkenyl, and a linear or branched unsubstituted or substituted C2-C15 alkynyl;
R2 is selected from the group consisting of a linear or branched unsubstituted or substituted C1-C15 alkyl, a linear or branched unsubstituted or substituted C2-C15 alkenyl, and a linear or branched unsubstituted or substituted C2-C15 alkynyl; each of R3 and R4 is independently selected from the group consisting of hydrogen and -C(0)-R5, wherein R5 is a C1-C15 alkyl; and stereoisomers, and salts thereof.
According to some embodiments, wherein Ri is a hydrogen. According to some embodiments, wherein Ri is a linear unsubstituted C1-C15 alkyl. According to some embodiments, Ri is Ci alkyl. According to some embodiments, Ri is methyl. According to some embodiments, R2 is a linear unsubstituted C1-C15 alkyl. According to some embodiments, wherein R2 is a linear unsubstituted C5 alkyl.
According to some embodiments, the compound is represented by the structure of Formula (Ila),
Figure imgf000018_0001
Formula (Ila) and stereoisomers, and salts thereof.
According to some embodiments, the compound is represented by the structure of Formula (Ilia),
Figure imgf000018_0002
Formula (Ilia) and stereoisomers, and salts thereof.
According to some embodiments, the compound is represented by the structure of Formula (IVa),
Figure imgf000018_0003
Formula (IVa) and stereoisomers thereof.
According to some embodiments, the pharmaceutical composition is administered in combination with at least one additional active pharmaceutical ingredient, wherein the at least one additional active pharmaceutical ingredient comprises at least one additional cannabinoid compound. According to some embodiments, the pharmaceutical composition comprises at least one additional active pharmaceutical ingredient, wherein the at least one additional active pharmaceutical ingredient comprises at least one additional cannabinoid compound. According to some embodiments, compound represented by any one of the structure of Formulae (Ia)-(I-e), (Ila)-(II-e) or (Illa)-(III-e) is administered in combination with at least one additional active pharmaceutical ingredient, wherein the at least one additional active pharmaceutical ingredient comprises at least one additional cannabinoid compound.
According to some embodiments, the additional cannabinoid compound is selected from the group consisting of cannabidiol (CBD), cannabigerol (CBG), D8- tetrahydrocannabinol (AS-THC), A9-tetrahydrocannabinol (A9-THC), cannabinol (CBN), A9(ll)- tetrahydrocannabinol (exo-THC), cannabichromene (CBC), tetrahydrocannabinol-C3 (THC-C3), tetrahydrocannabinol-C4 (THC-C4), tetrahydrocannabinol-C7 (THC-C7), esters thereof and combination thereof. According to some embodiments, the additional cannabinoid compound comprises a cannabigerolic acid (CBGA) ester represented by Formula (V), a cannabinolic acid (CBNA) ester represented by Formula (VI), a cannabidiolic acid (CBDA) ester represented by Formula (VII), a tetrahydrocannabinolic acid (THCA) ester represented by Formula (VIII) or any combination thereof,
Figure imgf000019_0001
Formula (VIII) wherein each of R6, R7, Rs, R9, Rio, R11, R12 and R13 is independently selected from the group consisting of a linear or branched, unsubstituted or substituted C1-C15 alkyl, a linear or branched, unsubstituted or substituted C2-C15 alkenyl, and a linear or branched, unsubstituted or substituted C2-C15 alkynyl; one of the dashed lines in Formula (VIII) represents a double bond and the other dashed line represents a single bond; and stereoisomers, and salts thereof.
It is to be understood that any combination of each of the aspects and the embodiments disclosed herein is explicitly encompassed within the disclosure of the present invention.
Further embodiments and the full scope of applicability of the present invention will become apparent from the detailed description given hereinafter. However, it should be understood that the detailed description and specific examples, while indicating preferred embodiments of the invention, are given by way of illustration only, since various changes and modifications within the spirit and scope of the invention will become apparent to those skilled in the art from this detailed description.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 represents the 'H NMR spectra of EPM308.
DETAILED DESCRIPTION OF THE INVENTION
The present invention provides novel cannabidiolic acid (CBDA) derivatives including acids and esters, and uses thereof. The present invention further provides pharmaceutical compositions comprising CBDA derivatives, alone or in combination with one or more additional cannabinoid compounds, for use in treating various diseases, disorders and symptoms, including but not limited to pain, impaired neurological function, inflammation including respiratory inflammation, inflammatory bowel disease and autoimmune diseases, nausea, vomiting, convulsions, low appetite and glaucoma.
According to one aspect, the present invention provides a compound represented by the structure of Formula (la),
Figure imgf000020_0001
Formula (la) wherein Ri is selected from the group consisting of a hydrogen, a linear or branched unsubstituted or substituted C1-C15 alkyl, a linear or branched unsubstituted or substituted C2-C15 alkenyl, and a linear or branched unsubstituted or substituted C2-C15 alkynyl; R2 is selected from the group consisting of a linear or branched unsubstituted or substituted C1-C15 alkyl, a linear or branched unsubstituted or substituted C2- Ci5 alkenyl, and a linear or branched unsubstituted or substituted C2-C15 alkynyl; each of R3 and R4 is independently selected from the group consisting of hydrogen and -C(0)-Rs, wherein R5 is a C1-C15 alkyl; and stereoisomers, and salts thereof.
According to some embodiments, the compound is represented by the structure of Formula (lb),
Figure imgf000021_0001
Formula (lb) wherein Ri, R2, R3 and R4 are defined as for Formula (la); and salts thereof. According to some embodiments, the compound is represented by the structure of Formula (Ic),
Figure imgf000021_0002
Formula (Ic) wherein Ri, R2, R3 and R4 are defined as for Formula (la); and salts thereof. According to some embodiments, the compound is represented by the structure of Formula (Id),
Figure imgf000022_0001
Formula (Id) wherein Ri, R2, R3 and R4 are defined as for Formula (la); and salts thereof. According to some embodiments, the compound is represented by the structure of Formula (Ie),
Figure imgf000022_0002
Formula (Ie) wherein Ri, R2, R3 and R4 are defined as for Formula (la); and salts thereof. According to some embodiments, the compound is represented by the structure of Formula (Ila),
Figure imgf000022_0003
Formula (Ila) and stereoisomers, and salts thereof.
According to some embodiments, the compound is represented by the structure of Formula (lib),
Figure imgf000022_0004
Formula (lib) and salts thereof. According to some embodiments, the compound is represented by the structure of Formula (lie),
Figure imgf000023_0001
Formula (lie) and salts thereof.
According to some embodiments, the compound is represented by the structure of Formula (lid),
Figure imgf000023_0002
Formula (lid) and salts thereof.
According to some embodiments, the compound is represented by the structure of Formula (He),
Figure imgf000023_0003
Formula (He) and salts thereof.
According to some embodiments, the compound is represented by the structure of Formula (Ilia),
Figure imgf000023_0004
Formula (Ilia) and stereoisomers, and salts thereof.
According to some embodiments, the compound is represented by the structure of Formula (Mb) (designated herein EPM308),
Figure imgf000024_0001
Formula (Mb) and salts thereof.
According to some embodiments, the compound is represented by the structure of Formula (IIIc),
Figure imgf000024_0002
Formula (IIIc) and salts thereof.
According to some embodiments, the compound is represented by the structure of Formula (Hid),
Figure imgf000024_0003
Formula (Hid) and salts thereof.
According to some embodiments, the compound is represented by the structure of Formula (Me),
Figure imgf000025_0001
Formula (Me) and salts thereof.
According to some embodiments, the compound is represented by the structure of Formula (IVa),
Figure imgf000025_0002
Formula (IVa) and stereoisomers thereof.
According to some embodiments, the compound is represented by the structure of Formula (IVb) (designated herein EPM 313),
Figure imgf000025_0003
Formula (IVb).
According to some embodiments, the compound is represented by the structure of Formula (IVc),
Figure imgf000026_0001
Formula (IVc).
According to some embodiments, the compound is represented by the structure of Formula (IVd),
Figure imgf000026_0002
Formula (IVd).
According to some embodiments, the compound is represented by the structure of Formula (IVe),
Figure imgf000026_0003
Formula (IVe).
According to some embodiments, Ri is hydrogen. According to some embodiments, Ri is a linear unsubstituted C1-C15 alkyl. According to some embodiments, Ri is a linear unsubstituted C2 alkyl. According to some embodiments, Ri is a linear unsubstituted C3 alkyl. According to some embodiments, Ri is a linear unsubstituted Ci- C4 alkyl. According to some embodiments, Ri is a linear unsubstituted C5 alkyl. According to some embodiments, Ri is a linear substituted C1-C15 alkyl. According to some embodiments, Ri is a branched unsubstituted C1-C15 alkyl. According to some embodiments, Ri is a branched substituted C1-C15 alkyl. According to some embodiments, Ri is methyl. According to some embodiments, Ri is a linear unsubstituted C2-C15 alkenyl. According to some embodiments, Ri is a linear substituted C2-C15 alkenyl. According to some embodiments, Ri is a branched unsubstituted C2-C15 alkenyl. According to some embodiments, Ri is a branched substituted C2-C15 alkenyl. According to some embodiments, Ri is a linear unsubstituted C2-C15 alkynyl. According to some embodiments, Ri is a linear substituted C2-C15 alkynyl. According to some embodiments, Ri is a branched unsubstituted C2-C15 alkynyl. According to some embodiments, Ri is a branched substituted C2-C15 alkynyl.
According to some embodiments, R2 is a linear unsubstituted C1-C15 alkyl. According to some embodiments, R2 is a linear substituted C1-C15 alkyl. According to some embodiments, R2 is a branched unsubstituted C1-C15 alkyl. According to some embodiments, R2 is a branched substituted C1-C15 alkyl. According to some embodiments, R2 is a linear unsubstituted C2 alkyl. According to some embodiments, R2 is a linear unsubstituted C3 alkyl. According to some embodiments, R2 is a linear unsubstituted C4 alkyl. According to some embodiments, R2 is a linear unsubstituted C5 alkyl (n-pentyl). According to some embodiments, R2 is a linear unsubstituted Ce alkyl. According to some embodiments, R2 is a linear unsubstituted C2-C15 alkenyl. According to some embodiments, R2 is a linear substituted C2-C15 alkenyl. According to some embodiments, R2 is a branched unsubstituted C2-C15 alkenyl. According to some embodiments, R2 is a branched substituted C2-C15 alkenyl. According to some embodiments, R2 is a linear unsubstituted C2-C15 alkynyl. According to some embodiments, R2 is a linear substituted C2-C15 alkynyl. According to some embodiments,
R2 is a branched unsubstituted C2-C15 alkynyl. According to some embodiments, R2 is a branched substituted C2-C15 alkynyl.
According to some embodiments, R3 is hydrogen. According to some embodiments, R3 is -C(0)-CH3. According to some embodiments, R3 is -C(0)-C2H5. According to some embodiments, R3 is -C(0)-C3H7. According to some embodiments, R3 is -C(0)-C4H9. According to some embodiments, R3 is -C(0)-C5Hn.
According to some embodiments, R4 is hydrogen. According to some embodiments, R4 is -C(0)-CH3. According to some embodiments, R4 is -C(0)-C2H5. According to some embodiments, R4 is -C(0)-C3H7. According to some embodiments, R4 is -C(0)-C4H9. According to some embodiments, R4 is -C(0)-C5Hn.
According to some embodiments, R3 and R4 are each Hydrogen. According to some embodiments, one of R3 and R4 is Hydrogen and the other is -C(0)-CH3. According to some embodiments, R3 and R4 are each -C(0)CH3. According to some embodiments, R5 is -CH3. According to some embodiments, R5 is -C2H5. According to some embodiments, R5 is -C3H7. According to some embodiments, R5 is -C4H9. According to some embodiments, R5 is -C5H11. According to some embodiments, R5 is a Ci-15 alkyl. According to some embodiments, R5 is a C1-5 alkyl. According to some embodiments, R5 is C5-10 alkyl. According to some embodiments, R5 is a Cio-15 alkyl.
It is to be understood that the compounds provided herein may contain one or more chiral centers. Such chiral centers may each be of either of the ( R ) or (S) configuration. In case a compound of the invention contains more than one chiral center, each one of those chiral centers may be of the (R) or ( S ) configuration, independently. Thus, the compounds provided herein may be enantiomerically pure, or be stereoisomeric or diastereomeric mixtures.
An “alkyl” group refers to a saturated aliphatic hydrocarbon, including straight- chain also referred to as linear-chain, and branched-chain. In one embodiment, the alkyl group has 1-15 carbons designated here as Ci-Ci5-alkyl. In another embodiment, the alkyl group has 2-6 carbons designated here as C2-C6-alkyl. In another embodiment, the alkyl group has 2-4 carbons designated here as C2-C4-alkyl. Each possibility represents a separate embodiment of the invention. The alkyl group may be unsubstituted or substituted by one or more groups selected from the group consisting of hydroxyl, halogen, amino, thiol, phosphate, and combinations thereof. Thus, the phrase "unsubstituted or substituted alkyl" refers to an alkyl group which is either unsubstituted (i.e. it is a hydrocarbon) or to an alkyl substituted with at least one of hydroxyl, halogen, amino, thiol, phosphate, and combinations thereof.
The terms "halo" and "halogen" refer to the fluoro, chloro, bromo or iodo atoms. There can be one or more halogens, which are the same or different.
An "alkenyl" group refers to an aliphatic hydrocarbon group containing at least one carbon-carbon double bond including straight-chain, linear-chain, branched-chain and cyclic alkenyl groups. In one embodiment, the alkenyl group has 2-15 carbon atoms (a C2-15 alkenyl). In another embodiment, the alkenyl group has 2-4 carbon atoms in the chain (a C2-4 alkenyl). Exemplary alkenyl groups include, but are not limited to, ethenyl, propenyl, n-butenyl, i-butenyl, 3-methylbut-2-enyl, n-pentenyl, heptenyl, octenyl, cyclohexyl-butenyl and decenyl. An alkylalkenyl is an alkyl group as defined herein bonded to an alkenyl group as defined herein. The alkenyl group can be unsubstituted or substituted through available carbon atoms with one or more groups defined herein for alkyl. Thus, the phrase "unsubstituted or substituted alkenyl" refers to an alkenyl group which is either unsubstituted (i.e. it is a hydrocarbon) or to an alkenyl substituted with at least one of hydroxyl, halogen, amino, thiol, phosphate, and combinations thereof.
An "alkynyl" group refers to an aliphatic hydrocarbon group containing at least one carbon-carbon triple bond including straight-chain or linear-chain and branched-chain. In one embodiment, the alkynyl group has 2-15 carbon atoms in the chain (a C2-15 alkynyl). In another embodiment, the alkynyl group has 2-4 carbon atoms in the chain (a C2-4 alkynyl). Exemplary alkynyl groups include, but are not limited to, ethynyl, propynyl, n- butynyl, 2-butynyl, 3-methylbutynyl, n-pentynyl, heptynyl, octynyl and decynyl. An alkylalkynyl is an alkyl group as defined herein bonded to an alkynyl group as defined herein. The alkynyl group can be unsubstituted or substituted through available carbon atoms with one or more groups defined herein for alkyl. Thus, the phrase "unsubstituted or substituted alkynyl" refers to an alkynyl group which is either unsubstituted (i.e. it is a hydrocarbon) or to an alkynyl substituted with at least one of hydroxyl, halogen, amino, thiol, phosphate, and combinations thereof.
According to another aspect, the present invention provides a pharmaceutical composition comprising a therapeutically effective amount of a compound represented by the structure of Formula (la) and a pharmaceutically acceptable carrier or excipient,
Figure imgf000029_0001
Formula (la) wherein
Ri is selected from the group consisting of a hydrogen, a linear or branched unsubstituted or substituted C1-C15 alkyl, a linear or branched unsubstituted or substituted C2-C15 alkenyl, and a linear or branched unsubstituted or substituted C2-C15 alkynyl;
R2 is selected from the group consisting of a linear or branched unsubstituted or substituted C1-C15 alkyl, a linear or branched unsubstituted or substituted C2- Ci5 alkenyl, and a linear or branched unsubstituted or substituted C2-C15 alkynyl; each of R3 and R4 is independently selected from the group consisting of hydrogen and -C(0)-R5, wherein R5 is a C1-C15 alkyl; and stereoisomers, and salts thereof.
The word "cannabinoid" as used herein refers to any compound that interacts with cannabinoid receptors including endocannabinoids (produced naturally in the body by humans and animals), phytocannabinoids (found in cannabis and some other plants), and synthetic cannabinoids (manufactured artificially). The term "cannabinoid acid" refers to the acid form of the above-mentioned cannabinoids.
The word "cannabidiol" refers to cannabidiol (CBD) and CBD derivatives. CBD may be obtained from industrial hemp extract with a trace amount of THC or from cannabis extract using high CBD cannabis cultivars. According to some embodiments, cannabidiol may be obtained from plant extract, or may be prepared synthetically (manufactured artificially), the structure of CBD is presented below:
Figure imgf000030_0001
The abbreviation "CBDA" is used herein to refer to the common cannabidiolic acid, which is the acid form of CBD. The term "cannabidiolic acid ester" or "cannabidiolic ester" refers to various molecules, which are the alkyl, alkenyl or alkynyl form of CBDA. The abbreviation "CBDA-ME" is used herein to refer to cannabidiolic acid methyl ester, which is the methyl ester form of CBDA. The structures of CBDA and CBDA-ME are presented below:
Figure imgf000030_0002
Figure imgf000031_0001
The term "cannabidiolic acid derivative" refers to derivatives of CBDA, which are substituted at position 3 by a carboxylic acid group, at position 9 by a hydroxyl group, independently at positions 2 and 6 by a different chemical group. The term "cannabidiolic acid ester derivative" refers to various molecules, which are the alkyl, alkenyl or alkynyl form of CBDA derivative as described herein. The chemical structures of CBDA and CBDA ester derivatives are presented below:
Figure imgf000031_0002
wherein
Ri is selected from the group consisting of a linear or branched unsubstituted or substituted C1-C15 alkyl, a linear or branched unsubstituted or substituted C2- Ci5 alkenyl, and a linear or branched unsubstituted or substituted C2-C15 alkynyl; each of R3 and R4 is independently selected from the group consisting of hydrogen and -C(0)-R5, wherein R5 is -a Ci-15 alkyl; and stereoisomers, and salts thereof.
It is to be understood that although the common cannabidiolic acid (CBDA) isomers include an n-CsHn chain at position 4, derivatives of CBDA may include other substituents, in particular alkyl, alkenyl or alkynyl groups. Therefore, the term cannabidiolic acid derivative includes corresponding structures, in which position 4 is substituted by a group, which is either an n-CsHn or a different chemical group, in particular alkyl, alkenyl or alkynyl groups. The term "cannabidiolic acid" and/or cannabidiolic acid ester" should be interpreted broadly referring to all possible stereoconfigurations and salts of the relevant formula.
The present invention thus provides pharmaceutical compositions comprising the compounds disclosed herein and a pharmaceutically acceptable carrier and/or excipient. The compounds of the present invention can be safely administered orally or non-orally. Routes of administration include, but are not limited to, oral, topical, mucosal, nasal, parenteral, gastrointestinal, intraspinal, intraperitoneal, intramuscular, intravenous, intrauterine, intraocular, intradermal, intracranial, intratracheal, intravaginal, intracerebroventricular, intracerebral, subcutaneous, ophthalmic, transdermal, rectal, buccal, epidural and sublingual. Each possibility represents a separate embodiment. The pharmaceutical compositions can be formulated as tablets (including e.g. film-coated tablets), powders, granules, capsules (including soft capsules), orally disintegrating tablets, and sustained-release preparations as is well known in the art. Each possibility represents a separate embodiment.
According to some embodiments, the pharmaceutical composition is formulated into a dosage form suitable for, oral, intranasal, intravenous, intraarterial, topical, intramuscular, transdermal, intraperitoneal, intrathecal, vaginal, rectal or subcutaneous administration.
According to some embodiments, the pharmaceutical composition is formulated as tablet, pill, capsule (e.g. soft or hard gelatin capsule), pellets, granules, powder, a wafer, coated or uncoated beads, lozenge, sachet, cachet, elixir, a depot system, a patch, suspension, dispersion, emulsion, solution, syrup, aerosol, oil, ointment, suppository, a gel, and a cream. Each possibility represents a separate embodiment.
For preparing solid compositions such as tablets, the active pharmaceutical ingredient is mixed with a pharmaceutical carrier or excipient to form a solid pre formulation composition containing a substantially homogeneous distribution of the compound disclosed herein in the pharmaceutical carrier or excipient.
Any method can be used to prepare the pharmaceutical compositions. For example, solid dosage forms can be prepared by wet granulation, dry granulation, direct compression and the like as is known in the art. The liquid forms in which the compositions disclosed herein may be incorporated, for administration via a route selected from oral, topical or by injection, include aqueous solutions, suitably flavored syrups, aqueous or oil suspensions, and flavored emulsions with edible oils such as cottonseed oil, sesame oil, coconut oil, or peanut oil, as well as elixirs and similar pharmaceutical vehicles. Each possibility represents a separate embodiment.
According to some embodiments, the composition may be formulated as single phase aqueous, emulsion or multiple emulsions. According to some embodiments, the composition is formulated as emulsion. These emulsions may be oil-in-water (o/w) emulsions, water-in-oil (w/o) emulsions, or multiple emulsions such as oil-in-water-in-oil (o/w/o) or water-in-oil-in-water (w/o/w). It is understood that the oil phase can comprise silicone oils, non-silicone organic oils, or mixtures thereof. The compositions can comprise two immiscible phases that are reconstituted prior to use. Each possibility represents a separate embodiment.
Compositions for inhalation include solutions and suspensions in pharmaceutically acceptable aqueous or organic solvents, or mixtures thereof, and powders. The liquid or solid compositions may contain suitable pharmaceutically acceptable carriers or excipients as described herein. The compositions may be administered by the oral or nasal respiratory route. Compositions may also be nebulized by use of inert gases. Nebulized solutions may be breathed directly from the nebulizing device or the nebulizing device may be attached to a face mask tent, or intermittent positive pressure breathing machine. Solution, suspension, or powder compositions may be administered, inter alia, orally or nasally, from devices that deliver the formulation in an appropriate manner.
According to some embodiments, the composition is prepared for topical administration, e.g. as an oil, ointment, gel or cream. Adjuvants for topical administration may include, for example, sodium carboxymethylcellulose, polyacrylates, polyoxyethylene-polyoxypropylene-block polymers, polyethylene glycol and wood wax alcohols. The term “gel” as used herein, refers to a substantially dilute cross-linked system, which exhibits little or no flow when in the steady-state having a solid jelly-like matrix. As contemplated herein, gel may comprise hydrogel, organogel, thermosensitive gel, non-thermosensitive gel, and aerogel. Each possibility represents a separate embodiment.
Hydrogels are highly hydrated, macromolecular networks, dispersed in water or other biological fluids. Hydrogels that exhibit the specific property of increased viscosity with increased temperatures are known as thermosensitive (or thermosetting) hydrogels. Such hydrogels have been shown to have easier application combined with longer survival periods at the site of application as compared to non-thermosensitive hydrogels, and are therefore advantageous as sustained-release drug delivery systems. Gel may be characterized by a critical gelation temperature, wherein gelation is effected at the critical gelation temperature or at temperatures above the critical gelation temperature. As used herein, “critical gelation temperature” refers to the lowest temperature at which some gelation of a material is observed (e.g., by increase in shear storage modulus).
According to some embodiments, the pharmaceutical composition can be formulated for local administration into the vagina. In accordance with these embodiments, the compositions may also be embedded in an article or administered using an applicator. Suitable forms for vaginal administration include, but are not limited to, bandage, suppository, pessary, douche, ointment, tablet, tampon, foam, cream, gel, paste, microcapsules, vaginal sponge, vaginal ring, and syringe-like applicator. Each possibility represents a separate embodiment. An optional carrier for vaginal administration comprises a lipid carrier, e.g. fatty acids, glycerin and pharmaceutically acceptable oils.
The pharmaceutical compositions disclosed herein may exhibit release mode which may be immediate release, controlled release or a mixture thereof. Each possibility represents a separate embodiment. “Immediate release” (IR) compositions in the context of the present invention refers to compositions in which the active ingredient is released without delay following administration. “Controlled release” (CR) compositions in the context of the present invention refers to compositions in which the active ingredient is released gradually over a period of time following administration.
Suitable pharmaceutically acceptable carriers or excipients include, but are not limited to, a binder, a filler, a diluent, a surfactant or emulsifier, a glidant or lubricant, buffering or pH adjusting agent, a tonicity enhancing agent, a wetting agent, a preservative, an antioxidant, a flavoring agent, a colorant, and a mixture or combination thereof. Each possibility represents a separate embodiment.
Suitable binders include, but are not limited to, polyvinylpyrrolidone, copovidone, hydroxypropyl methylcellulose, starch, and gelatin. Each possibility represents a separate embodiment.
Suitable fillers include, but are not limited to, sugars such as lactose, sucrose, mannitol or sorbitol and derivatives therefore (e.g. amino sugars), ethylcellulose, microcrystalline cellulose, and silicified microcrystalline cellulose. Each possibility represents a separate embodiment. Suitable lubricants include, but are not limited to, sodium stearyl fumarate, stearic acid, polyethylene glycol or stearates, such as magnesium stearate. Each possibility represents a separate embodiment.
Suitable diluents include, but are not limited to, dicalcium phosphate dihydrate, sugars, lactose, calcium phosphate, cellulose, kaolin, mannitol, sodium chloride, and dry starch. Each possibility represents a separate embodiment.
Suitable surfactants or emulsifiers include, but are not limited to, polyvinyl alcohol (PVA), polysorbate, polyethylene glycols, polyoxyethylene- polyoxypropylene block copolymers known as “poloxamer”, polyglycerin fatty acid esters such as decaglyceryl monolaurate and decaglyceryl monomyristate, sorbitan fatty acid ester such as sorbitan monostearate, polyoxyethylene sorbitan fatty acid ester such as polyoxyethylene sorbitan monooleate (Tween), polyethylene glycol fatty acid ester such as polyoxyethylene monostearate, polyoxyethylene alkyl ether such as polyoxyethylene lauryl ether, polyoxyethylene castor oil and hardened castor oil such as polyoxyethylene hardened castor oil. Each possibility represents a separate embodiment.
Suitable glidants or lubricants include, but are not limited to, colloidal silicon dioxide, magnesium stearate, talc, and mineral oil. Each possibility represents a separate embodiment.
Suitable buffering or pH adjusting agent include, but are not limited to, acidic buffering agents such as short chain fatty acids, citric acid, acetic acid, hydrochloric acid, sulfuric acid and fumaric acid; and basic buffering agents such as tris, sodium carbonate, sodium bicarbonate, sodium hydroxide, potassium hydroxide and magnesium hydroxide. Each possibility represents a separate embodiment.
Suitable tonicity enhancing agents include, but are not limited to, ionic and non ionic agents such as, alkali metal or alkaline earth metal halides, urea, glycerol, sorbitol, mannitol, propylene glycol, and dextrose. Each possibility represents a separate embodiment.
Suitable wetting agents include, but are not limited to, glycerin, cetyl alcohol, and glycerol monostearate. Each possibility represents a separate embodiment.
Suitable preservatives include, but are not limited to, benzalkonium chloride, benzoxonium chloride, thiomersal, phenylmercuric nitrate, phenylmercuric acetate, phenylmercuric borate, methylparaben, propylparaben, chlorobutanol, benzyl alcohol, phenyl alcohol, chlorohexidine, and polyhexamethylene biguanide. Each possibility represents a separate embodiment. Suitable antioxidants include, but are not limited to, sorbic acid, ascorbic acid, ascorbate, glycine, a-tocopherol, butylated hydroxyanisole (BHA), and butylated hydroxytoluene (BHT). Each possibility represents a separate embodiment.
Suitable flavoring agents include, but are not limited to, sweeteners such as sucralose and synthetic flavor oils and flavoring aromatics, natural oils, extracts from plants, leaves, flowers, and fruits, and combinations thereof. Exemplary flavoring agents include cinnamon oils, oil of wintergreen, peppermint oils, clover oil, hay oil, anise oil, eucalyptus, vanilla, citrus oil such as lemon oil, orange oil, grape and grapefruit oil, and fruit essences including apple, peach, pear, strawberry, raspberry, cherry, plum, pineapple, and apricot. Each possibility represents a separate embodiment.
Suitable colorants include, but are not limited to, alumina (dried aluminum hydroxide), annatto extract, calcium carbonate, canthaxanthin, caramel, b-carotene, cochineal extract, carmine, potassium sodium copper chlorophyllin (chlorophyllin-copper complex), dihydroxy acetone, bismuth oxychloride, synthetic iron oxide, ferric ammonium ferrocyanide, ferric ferrocyanide, chromium hydroxide green, chromium oxide greens, guanine, mica-based pearlescent pigments, pyrophyllite, mica, dentifrices, talc, titanium dioxide, aluminum powder, bronze powder, copper powder, and zinc oxide. Each possibility represents a separate embodiment.
It is contemplated that the compounds include any solvate thereof. The term “solvate” as used herein refers to a physical association of a compound disclosed herein with one or more solvent molecules. This physical association involves varying degrees of ionic and covalent bonding, including hydrogen bonding. In certain instances, the solvate will be capable of isolation. “Solvate” encompasses both solution-phase and isolatable solvates. Non-limiting examples of suitable solvates include ethanolates, methanolates and the like. A “hydrate” is a solvate in which the solvent molecule is water.
In embodiments in which the compound is incorporated into a pharmaceutical composition in solid state form, the present disclosure also includes any polymorphs thereof. The term “polymorph” refers to a particular crystalline or amorphous state of a substance, which can be characterized by particular physical properties such as X-ray diffraction, electron diffraction, IR spectra, Raman spectra, melting point, and the like.
Any of the cannabinoids disclosed herein, specifically, the abnormal cannabidiolic acids and their esters, can be prepared synthetically or semi-synthetically .
Natural cannabinoids may be isolated or extracted from a natural source or prepared by synthetic or semi- synthetic means. For example, cannabinoids can be isolated by extraction from cannabis plants. Plants in the cannabis genus include, but are not limited to, Cannabis sativa, Cannabis ruderalis, and Cannabis indica. Each possibility represents a separate embodiment. These plants are the natural sources of cannabinoids.
According to some embodiments, the pharmaceutical composition further comprising at least one additional active pharmaceutical ingredient.
According to some embodiments, the additional active pharmaceutical ingredient comprises at least one additional cannabinoid compound.
According to some embodiments, the additional cannabinoid compound is selected from the group consisting of cannabidiol (CBD), cannabigerol (CBG), D8- tetrahydrocannabinol (AS-THC), A9-tetrahydrocannabinol (A9-THC), cannabinol (CBN), A9(ll)-tetrahydrocannabinol (exo-THC), cannabichromene (CBC), tetrahydrocannabinol-C3 (THC-C3), tetrahydrocannabinol-C4 (THC-C4), tetrahydrocannabinol-C7 (THC-C7), esters thereof and combinations thereof. Each possibility represents a separate embodiment of the present invention. According to some embodiments, the additional cannabinoid compound comprises a cannabigerolic acid (CBGA) ester represented by Formula (V), a cannabinolic acid (CBNA) ester represented by Formula (VI), a cannabidiolic acid (CBDA) ester represented by Formula (VII), a tetrahydrocannabinolic acid (THCA) ester represented by Formula (VIII) or any combination thereof,
Figure imgf000038_0001
Formula (VIII) wherein each of R6, R7, Rs, R9, Rio, R11, R12 and R13 is independently selected from the group consisting of a linear or branched, unsubstituted or substituted C1-C15 alkyl, a linear or branched, unsubstituted or substituted C2-C15 alkenyl, and a linear or branched, unsubstituted or substituted C2-C15 alkynyl; one of the dashed lines in Formula (VIII) represents a double bond and the other dashed line represents a single bond; and stereoisomers, and salts thereof. According to some embodiments, the additional cannabinoid compound is cannabidiolic acid methyl ester.
According to some embodiments, the additional cannabinoid compound is present in one or more extracts of a cannabis plant. According to some embodiments, the cannabis plant extract is obtained from a strain selected from the group consisting of Cannabis sativa, Cannabis indica, Cannabis ruderalis, a hybrid strain, and combinations thereof. According to some embodiments, the cannabis plant extract is obtained from a strain selected from the group consisting of a high-CBD strain, a high-THC strain, and a combination thereof. According to some embodiments, the cannabis plant extract comprises at least one cannabinoid selected from the group consisting of cannabidiol (CBD), tetrahydrocannabinol (THC), cannabinol (CBN), cannabigerol (CBG), cannabichromene (CBC), acids thereof and combinations thereof.
According to some embodiments, the cannabis plant extract comprises from about 1 to about 25% (w/w) of CBD. According to some embodiments, the cannabis plant extract comprises about 1% (w/w) of CBD. According to some embodiments, the cannabis plant extract comprises about 10% (w/w) of CBD. According to some embodiments, the cannabis plant extract comprises about 25% (w/w) of CBD.
According to some embodiments, the cannabis plant extract comprises from about 1 to about 25% (w/w) of THC. According to some embodiments, the cannabis plant extract comprises about 1% (w/w) of THC. According to some embodiments, the cannabis plant extract comprises about 10% (w/w) of THC. According to some embodiments, the cannabis plant extract comprises about 25% (w/w) of THC.
The term “extract” as used herein refers a product prepared by extraction by physical means (e.g. by comminuting, pressing, heating, pulsed electric field assisted treatments, shear treatments and pressure wave treatments), by chemical means (e.g. by treatment with an acid, a base, a solvent) and/or by biochemical means (e.g. by treatment with hydrolytic enzymes, microorganisms). The term refers to a liquid substance obtained through extraction from a given substance, or to a concentrate or essence which is free of, or substantially free of solvent. The term extract may be a single extract obtained from a particular extraction step or series of extraction steps. Extract also may be a combination of extracts obtained from separate extraction steps or separate feedstocks. Such combined extracts are thus also encompassed by the term “extract”. Any methods of extraction with suitable solvent are encompassed. Exemplary extraction methods can be found for example in US patent 6,403,126, the contents of which are incorporated by reference herein. The extract may be obtained from any part of the plant e.g. from leaves, flowers, stems, roots, fruits and seeds. The extract may be aqueous or oily.
According to some embodiments, the cannabis plant extract is formed through contact with a suitable solvent or a combination of solvents. According to some embodiments, the solvent is selected from the group consisting of a polar solvent, a hydrocarbon solvent, carbon dioxide, and a combination thereof.
The term “extract” further refers to a liquid or semi-solid or resinous substance obtained through extraction from plants defined in the present application, i.e. extracts obtained from cannabis plant e.g. Cannabis sativa, Cannabis indica, and Cannabis ruderalis. In some embodiments, the term refers to a mixture of liquid or semi-solid, resinous substances obtained through extraction from two or more different plants. In some embodiments, the term refers also to a compound purified from the extract. According to some embodiments, the term “extract” has the meaning of a mixture or combination of two or more extracts.
The term "cannabis extract" as used herein refers to one or more plant extracts from the cannabis plant. A cannabis extract contains, in addition to one or more cannabinoids, one or more non-cannabinoid components which are co-extracted with the cannabinoids from the plant material. Their respective ranges in weight will vary according to the starting plant material and the extraction methodology used. Cannabinoid-containing plant extracts may be obtained by various means of extraction of cannabis plant material. Such means include but are not limited to supercritical or subcritical extraction with CO2, extraction with hot or cold gas and extraction with solvents. In some embodiments, the term refers to a mixture of liquid or semi-solid, resinous substances obtained through extraction from two or more different cannabis species. In some embodiments, the term refers also to a compound purified from the extract. The term "cannabis plant" as used herein, refers to plants of the genus Cannabis, including but not limited to Cannabis sativa, Cannabis indica, and Cannabis ruderalis. According to some embodiments, cannabis plant is a CBD-rich strain of cannabis plant or THC-rich strain of cannabis plant. Each possibility represents a separate embodiment.
The term “hybrid strain” refers to different strains of Cannabis which include differing amounts and/or ratios of the various cannabinoid compounds. For example, Cannabis sativa typically has a relatively high THC/CBD ratio. Conversely, Cannabis indica has a relative low THC/CBD ratio compared to Cannabis sativa (although the absolute amount of THC can be higher in Cannabis indica than in Cannabis sativa).
As used herein the terms “high-CBD strain” and “CBD-rich strain” refer to a strain of cannabis plant which comprises CBD and optionally one or more additional cannabinoids, such as, for example, but not limited to: THC, CBN, and the like. According to some embodiments, CBD is the main component in the high-CBD strain.
As used herein the terms “high-THC strain” and “THC-rich strain” are directed to a strain of cannabis plant which comprises THC and optionally one or more additional cannabinoids, such as, for example, but not limited to: CBD, CBN, and the like. According to some embodiments, THC is the main component in the high-THC strain. According to another aspect, the present invention provides a pharmaceutical composition comprising a therapeutically effective amount of a compound represented by the structure of Formula (la) and a pharmaceutically acceptable carrier or excipient, for use in the treatment of a disease, disorder or symptom amenable to treatment with CBD,
Figure imgf000041_0001
Formula (la) wherein
Ri is selected from the group consisting of a hydrogen, a linear or branched unsubstituted or substituted C1-C15 alkyl, a linear or branched unsubstituted or substituted C2-C15 alkenyl, and a linear or branched unsubstituted or substituted C2-C15 alkynyl;
R2 is selected from the group consisting of a linear or branched unsubstituted or substituted C1-C15 alkyl, a linear or branched unsubstituted or substituted C2- Ci5 alkenyl, and a linear or branched unsubstituted or substituted C2-C15 alkynyl; each of R3 and R4 is independently selected from the group consisting of hydrogen and -C(0)-R5, wherein R5 is a C1-C15 alkyl; and stereoisomers, and salts thereof.
According to some embodiments, the disease, disorder or symptom amenable to treatment with cannabidiol (CBD) is selected from the group consisting of pain, autoimmune disease, cancer, bacterial infection, impaired neurological function, inflammation, nausea, vomiting, convulsions, low appetite and glaucoma.
According to some embodiments, the composition disclosed herein is for use in the treatment of pain. According to some embodiments, the composition disclosed herein is for use in the attenuation of pain. According to some embodiments, the composition disclosed herein is for use in the alleviation of pain.
According to some embodiments, the composition disclosed herein is for use in the treatment of autoimmune disease. According to some embodiments, the composition disclosed herein is for use in the treatment of cancer. According to some embodiments, the composition disclosed herein is for use as an anti-tumor agent.
According to some embodiments, the composition disclosed herein is for use in the treatment of bacterial infection. According to some embodiments, the composition disclosed herein is for use as an anti-bacterial agent.
According to some embodiments, the composition disclosed herein is for use in the treatment of impaired neurological function.
According to some embodiments, the composition disclosed herein is for use in the treatment of impaired neurological function selected from the group consisting of stroke, trauma, Parkinson’s disease, vascular dementia, senile dementia, Alzheimer’s disease, mild cognitive impairment, Huntington’s Disease, Amyotrophic lateral sclerosis (ALS), epilepsy, multiple sclerosis, and psychiatric disorders. Each possibility represents a separate embodiment of the present invention.
According to some embodiments, the impaired neurological function is epilepsy.
According to some embodiments, the composition disclosed herein is for use in the treatment of psychiatric disorders. According to some embodiments, the psychiatric disorder is selected from the group consisting of depression, anxiety, acute or chronic stress, schizophrenia, panic attacks, ADHD, bipolar disorder, obsessive compulsive disorder or personality disorders.
According to some embodiments, the composition disclosed herein is for use in the treatment of depression. According to some embodiments, the composition disclosed herein is for use in the treatment of anxiety. According to some embodiments, the composition disclosed herein is for use in the treatment of acute or chronic stress. According to some embodiments, the composition disclosed herein is for use in the treatment of schizophrenia. According to some embodiments, the composition disclosed herein is for use in the treatment of panic attacks. According to some embodiments, the composition disclosed herein is for use in the treatment of ADHD. According to some embodiments, the composition disclosed herein is for use in the treatment of bipolar disorder. According to some embodiments, the composition disclosed herein is for use in the treatment of obsessive-compulsive disorder.
According to some embodiments, the composition disclosed herein is for use in the treatment of inflammation.
According to some embodiments, the composition disclosed herein is for use in treating joint inflammatory diseases and joint degeneration. According to additional embodiments, the pharmaceutical composition is for use in treating respiratory inflammation. According to additional embodiments, the composition disclosed herein is for use in treating inflammatory bowel disease.
According to some embodiments, the inflammation is respiratory inflammation.
According to some embodiments, the respiratory inflammation is acute respiratory inflammation.
The respiratory inflammation can be associated with a disease or disorder including, but not limited to, asthma, chronic obstructive airway disorder, chronic obstructive pulmonary disease (COPD), pneumonia, respiratory syncytial viral infection, bronchitis, bronchiolitis, idiopathic pulmonary fibrosis, cystic fibrosis, acute respiratory distress syndrome (ARDS), bronchopulmonary dysplasia, occupational respiratory disease, particulate exposure, pleurisy, emphysema, and pulmonary edema.
According to some embodiments, the respiratory inflammation is chronic respiratory inflammation. According to certain embodiments, the respiratory inflammation is pneumonitis. According to certain embodiments, the respiratory inflammation is asthma. According to certain embodiments, the respiratory inflammation is COPD.
According to some embodiments, the respiratory inflammation is caused by a pathogen. According to certain embodiments, the respiratory inflammation is caused by a pathogen selected from the group consisting of bacteria, viruses, parasites and fungi.
According to some embodiments, the respiratory inflammation is caused by a virus. According to additional embodiments, the respiratory inflammation is caused by Severe acute respiratory syndrome (SARS) vims. According to additional embodiments, the respiratory inflammation is caused by SARS-CoV-2 (Covid-19).
According to some embodiments, the composition disclosed herein is for use as a neuroprotective agent. According to some embodiments, the composition disclosed herein is for use in treating multiple sclerosis.
According to some embodiments, the composition disclosed herein is for use in treating cancer.
According to some embodiments, the composition disclosed herein is for use in the treatment of a symptom selected from the group consisting of nausea, vomiting and low appetite. According to some embodiments, the composition disclosed herein is for use in the treatment of nausea. According to some embodiments, the composition disclosed herein is for use in the treatment of vomiting. According to some embodiments, the composition disclosed herein is for use in the treatment of low appetite.
According to some embodiments, the composition disclosed herein is for use in the treatment of convulsions.
According to some embodiments, the composition disclosed herein is for use in the treatment of glaucoma.
According to some embodiments, the composition disclosed herein is for use in the treatment of gastrointestinal diseases or disorders.
According to some embodiments, the composition disclosed herein is for use in the treatment of Non-Alcoholic Fatty Liver Disease (NAFLD), chronic kidney disease (CKD), obesity, hyperglycemia, diabetes, metabolic syndrome and/or obesity related diseases.
According to some embodiments, the composition disclosed herein is for use in the treatment of obesity.
According to some embodiments, the composition disclosed herein is for use in the treatment of muscular dystrophy.
According to another aspect, the present invention provides a method of treating a disease, disorder or symptom amenable to treatment with cannabidiol (CBD), the method comprising administering to a subject in need thereof a pharmaceutical composition comprising a therapeutically effective amount of a compound represented by the structure of Formula (la) and a pharmaceutically acceptable carrier or excipient,
Figure imgf000044_0001
Formula (la) wherein
Ri is selected from the group consisting of a hydrogen, a linear or branched unsubstituted or substituted C1-C15 alkyl, a linear or branched unsubstituted or substituted C2-C15 alkenyl, and a linear or branched unsubstituted or substituted C2-C15 alkynyl;
R2 is selected from the group consisting of a linear or branched unsubstituted or substituted C1-C15 alkyl, a linear or branched unsubstituted or substituted C2- Ci5 alkenyl, and a linear or branched unsubstituted or substituted C2-C15 alkynyl; each of R3 and R4 is independently selected from the group consisting of hydrogen and -C(0)-R5, wherein R5 is a C1-C15 alkyl; and stereoisomers, and salts thereof.
According to some embodiments, the method of treating, a disease, disorder or symptom is selected from the group consisting of impaired neurological function, inflammation, nausea, vomiting, convulsions, low appetite, and glaucoma, the method comprising administering to a subject in need thereof the pharmaceutical composition disclosed herein.
According to some embodiments, the impaired neurological function is selected from the group consisting of stroke, trauma, Parkinson’ s disease, vascular dementia, senile dementia, Alzheimer’s disease, mild cognitive impairment, Huntington’s Disease, Amyotrophic lateral sclerosis (ALS), epilepsy, multiple sclerosis, and psychiatric disorders.
According to some embodiments, there is provided a method of treating, attenuating or alleviating pain, the method comprising administering to a subject in need thereof the pharmaceutical composition disclosed herein.
According to some embodiments, there is provided a method of treating, attenuating or alleviating pain, the method comprising administering to a subject in need thereof a pharmaceutical composition comprising a therapeutically effective amount of a compound represented by the structure of Formula (la) as described herein, and a pharmaceutically acceptable carrier or excipient.
According to some embodiments, the pharmaceutical composition is administered in combination with at least one additional active pharmaceutical ingredient as described herein.
Other suitable additional active pharmaceutical ingredients include agents for pain relief such as non-steroidal anti-inflammatory agents (NSAIDs). Suitable NS A TPs include, but are not limited to, acetyl salicylic acid, indometacin, sulindac, phenylbutazone, diclofenac, fentiazac, ketorolac, piroxicam, tenoxicam, mecoxicam, meloxicam, cinnoxicam, ibufenac, ibuprofen, naproxen, ketoprofen, nabumetone, niflumic acid, nimesulide, and pharmaceutically acceptable salts thereof. Each possibility represents a separate embodiment of the invention. A group of NSAIDs within the scope of the present invention are Cox-2 inhibitors. Cox-2 inhibitors suitable for use in the combinations of the present invention include, but are not limited to, (4-(4-cyclohexyl-2-methyloxazol-5-yl)-2-fluorobenzenesulfonamide), 5-chloro-3-(4-(methylsulfonyl)phenyl)-2-(methyl-5-pyridinyl)pyridine, 2-(3,5- difluorophenyl)-3-4 (methylsulfonyl)phenyl)-2-cyclopenten-l-one, 4-[5-(4- methylphenyl)-3-(trifluoromethyl)-lH-pyrazol-l-yl]-benzenesulfonamide, rofecoxib, (4- (4-methylsulfonyl)phenyl]-3-phenyl-2(5H)-furanone), 4- (5-methyl-3-phenylisoxazol-4- yl)benzenesulfonamide, N - [ [4-(5-methyl-3 -phenylisoxazol-4yl]phenyl] sulfonyl] propanamide, 4- [5-(4-chlorophenyl)-3 -(trifluoromethyl)- 1 H-pyrazole- 1 - yl]benzenesulfonamide, N- (2, 3-dihydro-l, l-dioxido-6-phenoxy-l, 2-benzisothiazol-5- yl) methanesulfonamide, 6- [ [5-(4-chlorobenzoyl)- 1 ,4-dimethyl- 1 H-pyrrol-2-yl] methyl] - 3(2H)-pyridazinone, N-(4-nitro-2-phenoxyphenyl) methanesulfonamide, 3-(3,4- difluorophenoxy)-5,5-dimethyl-4-[4-(methylsulfonyl) phenyl]-2 (SH)-furanone, N-[6- [(2, 4-difluorophenyl) thio]-2, 3 -dihydro- l-oxo-lH-inden-5-yl] methanesulfonamide, 3- (4-chlorophenyl)-4-[-4-(methylsulfonyl) phenyl]-2 (3H)-oxazolone, 4-[3-(4- fluorophenyl)-2,3-dihydro-2-oxo-4-oxazolyl] benzenesulfonamide, 3- [4-
(methylsulfonyl) phenyll-2-phonyl-2-cyclopenten- 1 -one, 4-(2-methyl-4-phenyl-5 - oxazolyl)benzenesulfonamide, 3-(4-fluorophenyl)-4-[4- (methylsulfonyl) phenyl]-2 (3H)-oxazolone, 5- (4-fluorophenyl)-l- [4- (methylsulfonyl) phenyl]-3-
(trifluoromethyl)-lH-pyrazole, 4- [5-phenyl)-3- (trifluoromethyl)-lH-pyrazol-l- yl)benzenesulfonamide, 4- [ 1 -phenyl-3 -(trifluoromethyl)-lH-pyrazol-5-yl] benzenesulfonamide, 4-[5-(4-fluorophenyl)-3- (trifluoromethyl) -lH-pyrazol- 1 -yl] benzenesulfonamide, N-[2-(cyclohexyloxy)-4-nitrophenyl] methanesulfonamide, N- [6- (2, 4-difluorophenoxy)-2, 3 -dihydro- l-oxo-lH-inden-5-yl] methanesulfonamide, 3-(4- chlorophenoxy-4-[(methylsulfonyl)amino]benzenesulfonamide, 3-(4-chlorophenoxy-4- [(methylsulfonyl)amino]benzenesulfonamide, 3-[(I-methyl-IH-imidzaol-2-yl) thio]-4 [(methylsulfonyl) amino] benzenesulfonamide, 5,5-dimethyl-4-[4-(methylsulfonyl) phenyl]-3-phenoxy-2(5H)-furanone, N-[6-[(4-ethyl-2-thiazolyl) thio]-l, 3-dihydro-l-oxo- 5 -isobenzofuranyl] methanesulfonamide, 3 - [(2,4-dichlorophenyl)thio] -4-
[(methylsulfonyl)amino]benzenesulfonamide, l-fluoro-4-[2-[4-(methylsulfonyl) phenyl] cyclopenten- 1 -yl] benzene, 4-[5-(4-chlorophenyl)-3-(difluoromethyl)- lH-pyrazol- 1 - yl]benzenesulfonamide, 3-[l-[4-(methylsulfonyl)phenyl]-4- (trifluoromethyl) -1H- imidazol-2-yl] pyridine, 4- [2- (3-pyridinyll)-4- (trifluoromethyl)-lH-imidazol-l-yl] benzenesulfonamide, 4-[5-(hydroxymethyl)-3-phenylisoxazol-4-yl]benzenesulfonamide, 4-[3-(4-chlorophenyl)-2,3-dihydro-2-oxo-4-oxazolyl] benzenesulfonamide, 4-[5- (difluoromethyl)-3-phenylisoxazol-4-yl] benzenesulfonamide, [1, l':2', l"-terphenyl]-4- sulfonamide, 4-(methylsulfonyl)-l,l', 2], l"-terpheynyl, 4-(2-phenyl-3- pyridinyl)benzenesulfonamide, N-[3-(fonnylamino)-4-oxo-6-phenoxy-4H-l- benzopyran-7-yl] methanesulfonamide, darbufelone, celecoxib, rofecoxib, parecoxib, and valdecoxib. Each possibility represents a separate embodiment of the invention. Exemplary Cox-2 inhibitors include, but are not limited to, celecoxib, rofecoxib, parecoxib and valdecoxib. Each possibility represents a separate embodiment of the invention.
According to some embodiments, there is provided a method of treating psychiatric disorders, the method comprising administering to a subject in need thereof the pharmaceutical composition disclosed herein.
According to some embodiments, there is provided a method of treating psychiatric disorders, the method comprising administering to a subject in need thereof a pharmaceutical composition comprising a therapeutically effective amount of a compound represented by the structure of Formula (la) as described herein, and a pharmaceutically acceptable carrier or excipient.
According to some embodiments, the pharmaceutical composition is administered in combination with at least one additional active pharmaceutical ingredient as described herein.
According to some embodiments, there is provided a method of treating impaired neurological function, the method comprising administering to a subject in need thereof the pharmaceutical composition disclosed herein.
According to some embodiments, there is provided a method of treating epilepsy comprising administering to a subject in need thereof the pharmaceutical composition disclosed herein.
According to some embodiments, there is provided a method of treating impaired neurological function, the method comprising administering to a subject in need thereof a pharmaceutical composition comprising a therapeutically effective amount of a compound represented by the structure of Formula (la) as described herein, and a pharmaceutically acceptable carrier or excipient.
According to some embodiments, there is provided a method of treating epilepsy, the method comprising administering to a subject in need thereof a pharmaceutical composition comprising a therapeutically effective amount of a compound ester represented by the structure of Formula (la) as described herein, and a pharmaceutically acceptable carrier or excipient.
According to some embodiments, the pharmaceutical composition is administered in combination with at least one additional active pharmaceutical ingredient as described herein.
According to some embodiments, there is provided a method of treating inflammation, the method comprising administering to a subject in need thereof the pharmaceutical composition disclosed herein.
According to some embodiments, there is provided a method of treating inflammation, the method comprising administering to a subject in need thereof a pharmaceutical composition comprising a therapeutically effective amount of a compound represented by the structure of Formula (la) as described herein, and a pharmaceutically acceptable carrier or excipient.
According to some embodiments, there is provided a method of treating joint inflammatory diseases and joint degeneration, the method comprising administering to a subject in need thereof a pharmaceutical composition comprising a therapeutically effective amount of a compound represented by the structure of Formula (la) as described herein, and a pharmaceutically acceptable carrier or excipient.
According to some embodiments, there is provided a method of treating respiratory inflammation, the method comprising administering to a subject in need thereof a pharmaceutical composition comprising a therapeutically effective amount of a compound represented by the structure of Formula (la) as described herein, and a pharmaceutically acceptable carrier or excipient.
According to some embodiments, there is provided a method of treating inflammatory bowel disease, the method comprising administering to a subject in need thereof a pharmaceutical composition comprising a therapeutically effective amount of a compound represented by the structure of Formula (la) as described herein, and a pharmaceutically acceptable carrier or excipient.
According to some embodiments, the pharmaceutical composition is administered in combination with at least one additional active pharmaceutical ingredient as described herein.
According to some embodiments, there is provided a method of treating a symptom selected from the group consisting of nausea, vomiting and low appetite, the method comprising administering to a subject in need thereof the pharmaceutical composition disclosed herein. Each possibility represents a separate embodiment of the present invention.
According to some embodiments, there is provided a method of treating a symptom selected from the group consisting of nausea, vomiting and low appetite, the method comprising administering to a subject in need thereof a pharmaceutical composition comprising a therapeutically effective amount of a compound represented by the structure of Formula (la) as described herein, and a pharmaceutically acceptable carrier or excipient. Each possibility represents a separate embodiment of the present invention.
According to some embodiments, the pharmaceutical composition is administered in combination with at least one additional active pharmaceutical ingredient as described herein. According to some embodiments, the symptom is nausea. According to some embodiments, the symptom is vomiting. According to some embodiments, the symptom is low appetite.
According to some embodiments, there is provided a method of treating convulsions, the method comprising administering to a subject in need thereof the pharmaceutical composition disclosed herein.
According to some embodiments, there is provided a method of treating convulsions, the method comprising administering to a subject in need thereof a pharmaceutical composition comprising a therapeutically effective amount of a compound represented by the structure of Formula (la) as described herein, and a pharmaceutically acceptable carrier or excipient.
According to some embodiments, the pharmaceutical composition is administered in combination with at least one additional active pharmaceutical ingredient as described herein.
According to some embodiments, there is provided a method of treating glaucoma, the method comprising administering to a subject in need thereof the pharmaceutical composition disclosed herein.
According to some embodiments, there is provided a method of treating glaucoma, the method comprising administering to a subject in need thereof a pharmaceutical composition comprising a therapeutically effective amount of a compound represented by the structure of Formula (la) as described herein, and a pharmaceutically acceptable carrier or excipient. According to some embodiments, the pharmaceutical composition is administered in combination with at least one additional active pharmaceutical ingredient as described herein.
Although the pharmaceutical composition comprising the compound of the present invention may be administered as the single therapeutic agent for the treatment of diseases or disorders described herein, combination therapy including co-administration of one or more additional active pharmaceutical ingredients is within the scope of the present invention. Co-administration of the compound of the present invention with one or more therapeutic agents may take place sequentially in any order, simultaneously or a combination thereof. For example, administration of the compound of the present invention can take place prior to, after or at the same time as the administration of the additional therapeutic agent(s). For example, a total treatment period can be decided for the compound of the present invention. The additional agent(s) can be administered prior to the onset of treatment with the compound of the present invention or following treatment with the compound of the present invention. In addition, the additional agent(s) can be administered during the period of administering the compound of the present invention but does not need to occur over the entire treatment period. In another embodiment, the treatment regimen includes pre-treatment with one agent, followed by the addition of the other agent or agents. Alternating sequences of administration are also contemplated. Alternating administration includes administration of the compound of the present invention, followed by the additional agent, followed by the compound of the present invention, etc. The aforementioned sequences can also be administrated in several cycles wherein each cycle may be similar or different with each possibility representing a separate embodiment. The therapeutic efficacy of the combination of the compound of the present invention and the additional agent(s) is at least additive. In some embodiments, the therapeutic efficacy is synergistic, namely the overall dose of each of the components may be lower, thus resulting in significantly lower side effects experienced by the subject, while a sufficient desirable therapeutic effect is nonetheless achieved. When combination therapy is involved, the compound of the present invention and the additional therapeutic agent(s) may he provided in a single dosage form such as a fixed-dose combination or in separate compositions intended for simultaneous administration.
The precise dose to be employed in the pharmaceutical composition disclosed herein will depend on the route of administration, and the seriousness of the disease, and should be decided according to the judgment of the practitioner and each patient's circumstances. An optional dosage will be within the range of about 0.01-1000 mg/kg of body weight, about 0.1 mg/kg to 100 mg/kg, about 1 mg/kg to lOOmg/kg, about 10 mg/kg to 75 mg/kg, about 0.1 to 1 mg/kg etc., including each value within the specified range.
According to some embodiments, the composition comprises the compound of the present invention at a concentration of between 10 to 200 mg/ml.
The administration schedule will depend on several factors such as the severity and progression of the disease, disorder or symptom. For example, the compositions of the invention can be taken once-daily, twice-daily, thrice daily, once-weekly or once- monthly. In addition, the administration can be continuous, i.e., every day, or intermittently. The terms “intermittent” or “intermittently” as used herein means stopping and starting at either regular or irregular intervals. For example, intermittent administration can be administration one to six days per week or it may mean administration in cycles (e.g. daily administration for two to eight consecutive weeks, then a rest period with no administration for up to one week) or it may mean administration on alternate days. The effectiveness of said compositions could enable a shortened period of treatment with superior results.
A "therapeutically effective amount" as used herein refers to an amount of an agent which is effective, upon single or multiple dose administration to the subject in providing a therapeutic benefit to the subject. In additional embodiments, the compounds of the present invention are used for the preparation of a medicament for treating any one of the diseases, disorders, conditions or symptoms disclosed herein.
The following examples describe specific aspects of the invention to illustrate the invention and provide a description of the present methods for those skilled in the art. It should be noted that the term “and” or the term “or” is generally employed in its sense including “and/or” unless the context clearly dictates otherwise. As used herein, the term “about” is meant to encompass variations of ±10%.
EXAMPLES
Example 1. Synthesis of Cannabidiolic Acid (CBDA)
The preparation process described in the PCT application WO 2018/235079 was applied. A mixture of cannabidiol (CBD, 314 mg, 1 mmol) and 2 molar solution of magnesium methyl carbonate (MMC/2M, 1.5 ml, 3 mmol) in dimethylformamide (DMF) was heated at 130°C for 3 hours. Then the reaction was cooled to 0°C, acidified with 10% hydrochloric acid and extracted with ether. The organic layer was washed with saline, dried over the drying agent magnesium sulfate (MgSCC) and then evaporated. The crude compound was then cleaned by column chromatography (20% ether-petroleum ether).
Example 2. Synthesis of Cannabidiolic Acid Methyl Ester (CBDA-ME)
The preparation process described in the PCT application WO 2018/235079 was applied. To a solution of cannabidiolic Acid (CBDA) (175 mg, 0.488 mmol) in 2.5 ml dichloromethane (CH2CI2), was added 0.02 ml of methanol (CH3OH, 0.488 mmol) and 7.2 mg of 4-Pyrrolidinopyridine (0.048 mmol). The reaction was stirred for 5 minutes at room temperature followed by the addition of the coupling agent, N,N' dicyclohexylcarbodiimide (DCC) (121 mg, 0.585 mmol) and stirred overnight. Then the solvent was evaporated and the crude mixture acidified with 5% hydrochloric acid and extracted with dichloromethane (CH2CI2). The organic layer was washed with saturated aqueous sodium bicarbonate (NaHCCh), dried over the drying agent magnesium sulfate (MgSCC) and then evaporated. The crude compound was then cleaned by column chromatography (2% ether-petroleum ether).
'H-NMR spectra were obtained using a Bruker AMX 300 MHz apparatus using the deuterated DMSO. Thin-layer chromatography (TLC) was run on silica gel 60F254 plates (Merck). Column chromatography was performed on silica gel 60 A (Merck). Compounds were located using a UV lamp at 254 nm.
Ή NMR (300 MHz, ((CD3)2SO)) d 6.18 (1H, s, Ar), 5.07 (1H, s), 4.44 (1H, s), 4.41 (1H, s), 3.82 (3H, s), 3.35 (1H, m), 2.66 (1H, m), 2.49 (2H, t), 2.09 (1H, b), 1.95 (3H, s), 1.71-1.05 (12, ms), 0.86 (3H, t).
Example 3. Synthesis of EPM 313
CBDA-ME (1.2gr) was dissolved in 30 ml dry pyridine, a catalytic amount of DMAP and 4.5ml of acetic anhydride was added. After lh stirring at room temperature under nitrogen full conversion is observed, the reaction was stopped by adding ice-cold HC1 till pH 1, the aqueous layer was washed with diethyl ether 5 times, then the combined ether phase washed with sodium bicarbonate and brine and dryer over MgS04. Fast purification on silica gel provided around 95% yield of the product. Then, 1.6 gr of the diacetate was dissolved in 80 ml dry dichloromethane, the 2.2 ml of t-butyl hydroperoxide is added and 220 mg of selenium dioxide. The reaction was mixed under nitrogen overnight, to yield 60% of 9-hydroxy derivative and around 10% of 9-oxo derivative, which can be readily separated by silica gel.
Example 4. Synthesis of EPM308
Compound EPM313 (400mg) was dissolved in 20ml dry THF + 40ml dry ethanol. 64.32mg of NaBH4 was added (leq for each acetate) under N2 flow, followed by the addition of 188.68mg of anhydrous CaCh (leq for each acetate). The reaction was stirred under the N2 atmosphere, at 70*C for 5.5h, to get almost full deprotection of the diacetate (yield 93%). Workup: THF and ethanol were evaporated, the reaction crude dissolved in acidic water + ethyl acetate, the water phase washed twice with ethyl acetate, the combined organic phase washed to neutrality, dried over MgSCC, filtered and evaporated. The 'H NMR spectrum of EPM 308 is shown in Figure 1.
Example 5. In-vivo model of Pain
The therapeutic effect of compounds EPM313 and EPM308 on pain are examined in the experimental model of hot-plate test, measuring the analgesic activity of compounds based on the reaction time of mice to lick their forepaws and/or jump after being placed on an aluminum hot plate heated to, and maintained at about 55° C as described in the US Patent 5,338,753, with minor modifications. An aluminum surface is maintained at about 55° C by circulating water through the passages in the metal. A clear plastic cylinder is placed on the surface to prevent escape. The end point is reached when the mouse either performed a hind paw lick or jumped off the surface; in no case are the animals kept more than 30 seconds on the plate. Control values are measured 3 hours before the test values. Two groups of mice are treated, each group with a different compound at different doses ninety (90) minutes before the hot plate test. The percent change in response time (latency) is calculated by comparing the mean of the control values with the mean of the test values and statistical significance determined by a paired t test.
Example 6. In-vivo model of Inflammatory
The therapeutic effect of compounds EPM313 and EPM308 on inflammatory is examined in the experimental model of inflammation (paw edema) in arachidonic acid or platelet activating factor (PAF) injected mice, as described in the US Patent 5,338,753. Briefly, PAF (1.0 mg) or arachidonic acid (1.0 mg) dissolved in 50 pL of 5% ethanol in saline, is injected subcutaneously into the plantar surface of the right hind paw of ether- anesthetized CD-I female mice (20-25 g). The volume of the right foot is measured to the level of the lateral malleous by water displacement before treatment, fifteen minutes after PAF injection, or thirty minutes after arachidonic acid injection. Two groups of mice are then treated, each group with a different compound injection at different doses. The change in paw volume is calculated for each mouse and the significance for each group is determined by a paired t test.
Example 7. In-vivo . Pentylenetetrazole-induced (PTZ) model of generalized seizures
The antiepileptic effects of compounds EPM313 and EPM308 are tested in an acute experimental epilepsy model in pentylenetetrazole (PTZ) injected rats as described in Obay et. al. (Peptides, 2007, 28, 1214-1219). Briefly, adult male Wistar albino rats are divided into a control group, and seven experimental groups with seven rats in each group. In order to generate epileptic seizures, PTZ (50 mg/kg) is injected intraperitoneally. The experimental groups receive intraperitoneal injections, each group treated with a different compound at different doses 30 min before PTZ injection. After PTZ injection, the rats are kept in cages and their behaviors are observed for 30 min. The latencies are separated into three components: first myoclonic jerk (FMJ), generalized clonic seizures (GCS) and tonic generalized extension (TGE).
While the present invention has been particularly described, persons skilled in the art will appreciate that many variations and modifications can be made. Therefore, the invention is not to be construed as restricted to the particularly described embodiments, and the scope and concept of the invention will be more readily understood by reference to the claims, which follow.

Claims

1. A compound represented by the structure of Formula (la),
Figure imgf000055_0001
Formula (la) wherein
Ri is selected from the group consisting of a hydrogen, a linear or branched unsubstituted or substituted C1-C15 alkyl, a linear or branched unsubstituted or substituted C2-C15 alkenyl, and a linear or branched unsubstituted or substituted C2-C15 alkynyl; R2 is selected from the group consisting of a linear or branched unsubstituted or substituted C1-C15 alkyl, a linear or branched unsubstituted or substituted C2- Ci5 alkenyl, and a linear or branched unsubstituted or substituted C2-C15 alkynyl; each of R3 and R4 is independently selected from the group consisting of hydrogen and -C(0)-R5 wherein R5 is a linear or branched unsubstituted or substituted Ci-15 alkyl; and stereoisomers, and salts thereof.
2. The compound according to claim 1 , which is represented by the structure of Formula
(lb),
Figure imgf000055_0002
Formula (lb) wherein Ri, R2, R3 and R4 are defined as for Formula (la); and salts thereof.
3. The compound according to claim 1 , which is represented by the structure of Formula
(Ic),
Figure imgf000056_0001
Formula (Ic) wherein Ri, R2, R3 and R4 are defined as for Formula (la); and salts thereof.
4. The compound according to claim 1, which represented by the structure of Formula (Id),
Figure imgf000056_0002
Formula (Id) wherein Ri, R2, R3 and R4 are defined as for Formula (la); and salts thereof.
5. The compound according to claim 1, which represented by the structure of Formula (Ie),
Figure imgf000056_0003
Formula (Ie) wherein Ri, R2, R3 and R4 are defined as for Formula (la); and salts thereof.
6. The compound according to any one of claims 1-5, wherein Ri is a linear unsubstituted C1-C15 alkyl.
7. The compound according to claim 6, wherein Ri is methyl.
8. The compound according to any one of claims 1-5, wherein Ri is hydrogen.
9. The compound according to any one of claims 1-8, wherein R2 is a linear unsubstituted C1-C15 alkyl.
10. The compound according to claim 9, wherein R2 is a linear unsubstituted C5 alkyl.
11. The compound according to any one of claims 1-10, wherein R3 and R4 are each hydrogen.
12. The compound according to any one of claims 1-10, wherein R3 and R4 are each -C(0)-CH3.
13. The compound according to any one of claims 1-10, wherein one of R3 and R4 is hydrogen and the other is -C(0)-CH3.
14. The compound according to claim 1, which is represented by the structure of Formula
(Ilia),
Figure imgf000057_0001
Formula (Ilia) and stereoisomers, and salts thereof.
15. The compound according to claim 1, which is represented by the structure of Formula
(mb)
Figure imgf000057_0002
Formula (Mb) and salts thereof.
16. The compound according to claim 1, which is represented by the structure of Formula
(Hie),
Figure imgf000057_0003
Formula (IIIc) and salts thereof.
17. The compound according to claim 1, which is represented by the structure of Formula (Hid),
Figure imgf000058_0001
and salts thereof.
18. The compound according to claim 1, which is represented by the structure of Formula
(me),
Figure imgf000058_0002
and salts thereof.
19. The compound according to claim 1, which is represented by the structure of Formula (IVa),
Figure imgf000058_0003
Formula (IVa) and stereoisomers thereof.
20. The compound according to claim 1, which is represented by the structure of Formula (IVb),
Figure imgf000058_0004
Formula (IVb).
21. The compound according to claim 1, which is represented by the structure of Formula (IVc),
Figure imgf000059_0001
Formula (IVc).
22. The compound according to claim 1, which is represented by the structure of Formula
(IVd),
Figure imgf000059_0002
Formula (IVd).
23. The compound according to claim 1, which is represented by the structure of Formula (IVe),
Figure imgf000059_0003
Formula (IVe).
24. The compound according to claim 1, which is represented by the structure of Formula
(Ha),
Figure imgf000059_0004
Formula (Ila) and stereoisomers, and salts thereof.
25. The compound according to claim 1, which is represented by the structure of Formula (lib),
Figure imgf000060_0001
and salts thereof.
26. The compound according to claim 1, which is represented by the structure of Formula
(lie),
Figure imgf000060_0002
and salts thereof.
27. The compound according to claim 1, which is represented by the structure of Formula (lid),
Figure imgf000060_0003
and salts thereof.
28. The compound according to claim 1, which is represented by the structure of Formula (He),
Figure imgf000060_0004
and salts thereof.
29. A pharmaceutical composition comprising a therapeutically effective amount of a compound represented by the structure of Formula (la) according to any one of claims 1 to 28, and a pharmaceutically acceptable carrier or excipient.
30. The pharmaceutical composition according to claim 29, wherein the pharmaceutically acceptable carrier or excipient comprises at least one of a binder, a filler, a diluent, a surfactant or emulsifier, a glidant or lubricant, a buffering or pH adjusting agent, a tonicity enhancing agent, a wetting agent, a preservative, an antioxidant, a flavoring agent, a colorant, and a mixture or combinations thereof.
31. The pharmaceutical composition according to claim 29 or 30, in a form selected from the group consisting of tablet, pill, capsule, pellets, granules, powder, a wafer, coated or uncoated beads, lozenge, sachet, cachet, elixir, a depot system, an a patch, suspension, dispersion, emulsion, solution, syrup, aerosol, oil, ointment, suppository, a gel, and a cream.
32. The pharmaceutical composition according to claim 29, which is formulated into a dosage form suitable for, oral, intranasal, intravenous, intraarterial, topical, intramuscular, transdermal, intraperitoneal, vaginal, rectal or subcutaneous administration.
33. The pharmaceutical composition according to any one of claim 29-33, further comprising at least one additional active ingredient.
34. The pharmaceutical composition according to claim 33, wherein the additional active ingredient comprises at least one additional cannabinoid compound.
35. The pharmaceutical composition according to claim 34, wherein the additional cannabinoid compound comprises a cannabigerolic acid (CBGA) ester represented by Formula (V), a cannabinolic acid (CBNA) ester represented by Formula (VI), a cannabidiolic acid (CBDA) ester represented by Formula (VII), a tetrahydrocannabinolic acid (THCA) ester represented by Formula (VIII) or any combination thereof,
Figure imgf000062_0001
Formula (VIII) wherein each of R6, R7, Rs, R9, Rio, R11, R12 and R13 is independently selected from the group consisting of a linear or branched, unsubstituted or substituted C1-C15 alkyl, a linear or branched, unsubstituted or substituted C2-C15 alkenyl, and a linear or branched, unsubstituted or substituted C2-C15 alkynyl; one of the dashed lines in Formula (VIII) represents a double bond and the other dashed line represents a single bond; and stereoisomers, and salts thereof.
36. The pharmaceutical composition according to any one of claims 34 or 35, wherein the additional cannabinoid compound is present in one or more extracts of a cannabis plant.
37. The pharmaceutical composition according to claim 36, wherein the cannabis plant extract is obtained from a strain selected from the group consisting of Cannabis sativa, Cannabis indica, Cannabis ruderalis, a hybrid strain, and combinations thereof.
38. The pharmaceutical composition according to claim 36 or 37, wherein the cannabis plant extract is obtained from a strain selected from the group consisting of a high- CBD strain, a high-THC strain, and a combination thereof.
39. The pharmaceutical composition according to claim 36, wherein the cannabis plant extract comprises at least one cannabinoid selected from the group consisting of cannabidiol (CBD), tetrahydrocannabinol (THC), cannabinol (CBN), cannabigerol (CBG), cannabichromene (CBC), acids thereof and combinations thereof.
40. The pharmaceutical composition according to claim 36, wherein the cannabis plant extract comprises from about 1 to about 25% (w/w) of CBD.
41. The pharmaceutical composition according to claim 36, wherein the cannabis plant extract comprises from about 1 to about 25% (w/w) of THC.
42. The pharmaceutical composition according to any one of claims 29-41, for use in the treatment of a disease, disorder or symptom amenable to treatment with cannabidiol
(CBD).
43. The pharmaceutical composition for use according to claim 42, wherein the disease, disorder or symptom is selected from the group consisting of pain, autoimmune disease, cancer, bacterial infection, impaired neurological function, inflammation, nausea, vomiting, convulsions, low appetite and glaucoma.
44. The pharmaceutical composition for use according to claim 43, wherein the disease is an autoimmune disease.
45. The pharmaceutical composition for use according to claim 43, wherein the disease is cancer.
46. The pharmaceutical composition for use according to claim 43, wherein the disease is treating bacterial infection.
47. The pharmaceutical composition for use according to claim 43, wherein the impaired neurological function is a psychiatric disorder selected from the group consisting of depression and anxiety.
48. The pharmaceutical composition for use according to claim 43, wherein the inflammation is a respiratory inflammation or inflammatory bowel disease.
49. The pharmaceutical composition for use according to claim 42, wherein the disease, disorder or symptom amenable to treatment with CBD is selected from the group consisting of Non-Alcoholic Fatty Liver Disease (NAFLD), chronic kidney disease (CKD), obesity, hyperglycemia, diabetes, metabolic syndrome and/or obesity related diseases.
50. The pharmaceutical composition for use according to claim 49, wherein the disease is obesity.
51. A method of treating a disease, disorder or symptom amenable to treatment with cannabidiol (CBD), the method comprising the step of administering to a subject in need thereof a pharmaceutical composition comprising a therapeutically effective amount of a compound represented by the structure of Formula (la) and a pharmaceutically acceptable carrier or excipient,
Figure imgf000064_0001
Formula (la) wherein
Ri is selected from the group consisting of a hydrogen, a linear or branched unsubstituted or substituted C1-C15 alkyl, a linear or branched unsubstituted or substituted C2-C15 alkenyl, and a linear or branched unsubstituted or substituted
C2-C15 alkynyl;
R2 is selected from the group consisting of a linear or branched unsubstituted or substituted C1-C15 alkyl, a linear or branched unsubstituted or substituted C2- Ci5 alkenyl, and a linear or branched unsubstituted or substituted C2-C15 alkynyl; each of R3 and R4 is independently selected from the group consisting of hydrogen and -C(0)-R5, wherein R5 is a C1-C15 alkyl; and stereoisomers, and salts thereof.
PCT/IL2021/050666 2020-06-03 2021-06-03 Cannabidiolic acid (cbda) derivatives and uses thereof WO2021245675A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202063033849P 2020-06-03 2020-06-03
US63/033,849 2020-06-03

Publications (1)

Publication Number Publication Date
WO2021245675A1 true WO2021245675A1 (en) 2021-12-09

Family

ID=78830170

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IL2021/050666 WO2021245675A1 (en) 2020-06-03 2021-06-03 Cannabidiolic acid (cbda) derivatives and uses thereof

Country Status (1)

Country Link
WO (1) WO2021245675A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023213715A1 (en) * 2022-05-04 2023-11-09 Société des Produits Nestlé S.A. Ampk activator (cbda) and sglt2 inhibitor for metabolic health

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030158191A1 (en) * 1999-03-22 2003-08-21 Travis Craig R. Cannabinol derivatives
WO2003080043A1 (en) * 2002-03-18 2003-10-02 Immugen Pharmaceuticals, Inc. Topical formulations of resorcinols and cannibinoids and methods of use
WO2016022936A1 (en) * 2014-08-07 2016-02-11 Murty Pharmaceuticals, Inc. An improved oral gastrointestinal dosage form delivery system of cannabinoids and/or standardized marijuana extracts
WO2020024009A1 (en) * 2018-07-31 2020-02-06 AusCann Group Holdings Ltd "solid self-emulsifying pharmaceutical compositions"
WO2020041321A1 (en) * 2018-08-20 2020-02-27 Berkowitz Barry A Process for the production of cannabinoids
WO2020092987A1 (en) * 2018-11-01 2020-05-07 Molecular Infusions, Llc Polymer-based oral cannabinoid and/or terpene formulations

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030158191A1 (en) * 1999-03-22 2003-08-21 Travis Craig R. Cannabinol derivatives
WO2003080043A1 (en) * 2002-03-18 2003-10-02 Immugen Pharmaceuticals, Inc. Topical formulations of resorcinols and cannibinoids and methods of use
WO2016022936A1 (en) * 2014-08-07 2016-02-11 Murty Pharmaceuticals, Inc. An improved oral gastrointestinal dosage form delivery system of cannabinoids and/or standardized marijuana extracts
WO2020024009A1 (en) * 2018-07-31 2020-02-06 AusCann Group Holdings Ltd "solid self-emulsifying pharmaceutical compositions"
WO2020041321A1 (en) * 2018-08-20 2020-02-27 Berkowitz Barry A Process for the production of cannabinoids
WO2020092987A1 (en) * 2018-11-01 2020-05-07 Molecular Infusions, Llc Polymer-based oral cannabinoid and/or terpene formulations

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
BORNHEIM, L.M. ; EVERHART, E. ; LI, J. ; CORREIA, M.: "Characterization of cannabidiol-mediated cytochrome P450 inactivation", BIOCHEMICAL PHARMACOLOGY, ELSEVIER, US, vol. 45, no. 6, 24 March 1993 (1993-03-24), US , pages 1323 - 1331, XP025541942, ISSN: 0006-2952, DOI: 10.1016/0006-2952(93)90286-6 *
DHIR, A: "Cannabidiol in Refractory Epilepsy", STUDIES IN NATURAL PRODUCTS CHEMISTRY, vol. 58, 14 August 2018 (2018-08-14), pages 419 - 438, XP009532738, DOI: 10.1016/B978-0-444-64056-7.00014-3 *
GREEN K, ET AL.: "INTRAOCULAR PRESSURE FOLLOWING SYSTEMIC ADMINISTRATION OF CANNABINOIDS", CURRENT EYE RESEARCH, INFORMA HEALTHCARE USA, US, vol. 02, no. 04, 1 January 1982 (1982-01-01), US , pages 247 - 253, XP001076642, ISSN: 0271-3683 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023213715A1 (en) * 2022-05-04 2023-11-09 Société des Produits Nestlé S.A. Ampk activator (cbda) and sglt2 inhibitor for metabolic health

Similar Documents

Publication Publication Date Title
US20230192588A1 (en) Cannabidiolic acid (cbda) derivatives and uses thereof
US7586008B2 (en) Propofol analogs, process for their preparation, and methods of use
KR20210151816A (en) Cannabinoid acid ester compositions and uses thereof
BRPI0608819B1 (en) crystalline forms of an imidazole derivative
UA119147C2 (en) Sulfonamides as modulators of sodium channels
WO2021245676A1 (en) Cannabidiolic acid (cbda) derivatives and uses thereof
JP2002322096A (en) Combination of medicine for therapy of attack and traumatic brain impediment
EP4161901A1 (en) Cannabigerolic acid (cbga) derivatives and uses thereof
JP2022130689A (en) Solid forms of sgc stimulator
JP2008542359A (en) Benzofuran derivatives with therapeutic activity
JP2022062115A (en) Phosphorus prodrugs of sgc stimulators
CN112839942A (en) 1,3, 4-oxadiazole derivative compounds as histone deacetylase 6 inhibitors and pharmaceutical compositions comprising the same
US9770432B2 (en) Deuterated benzopyran compounds and the use in the treatment of pain and inflammation
WO2021034403A1 (en) Cannabinoid acid ester compositions and uses thereof
WO2021245675A1 (en) Cannabidiolic acid (cbda) derivatives and uses thereof
WO2013147161A1 (en) Nipecotic acid derivative and use thereof for medical purposes
JPS62108814A (en) Inhibitor against formation of peroxide lipide
MX2013013355A (en) Polymorph form of 4-{[4-({[4-(2,2,2-trifluoroethoxy)-1,2-benzisox azol-3-yl]oxy}methyl)piperidin-1-yl]methyl}-tetrahydro-2h-pyran- 4-carboxylic acid.
TW202132249A (en) Cannabidiol-type cannabinoid compound
TWI823903B (en) CRYSTALLINE FORMS OF AN sGC STIMULATOR
WO2021245669A1 (en) Abnormal cannabidiol (abn-cbd) acid derivatives and uses thereof
JP2009515943A (en) Amino acid derivatives
WO2006056874A1 (en) Salt form
CN108658964B (en) Polymorphic substance
WO2006027681A1 (en) Combination of a 5-ht(1) receptor agonist and an alpha-2-delta ligand for the treatment of migraine

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: 21818839

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 21818839

Country of ref document: EP

Kind code of ref document: A1