US20220168428A1 - Cannabinoid glycoside prodrugs and methods of synthesis - Google Patents

Cannabinoid glycoside prodrugs and methods of synthesis Download PDF

Info

Publication number
US20220168428A1
US20220168428A1 US17/527,685 US202117527685A US2022168428A1 US 20220168428 A1 US20220168428 A1 US 20220168428A1 US 202117527685 A US202117527685 A US 202117527685A US 2022168428 A1 US2022168428 A1 US 2022168428A1
Authority
US
United States
Prior art keywords
cannabinoid
glucopyranosyl
ugt76g1
glucose
glycosides
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
US17/527,685
Inventor
Brandon J. Zipp
Janee M. Hardman
Robert T. Brooke
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Graphium Biosciences Inc
Original Assignee
Graphium Biosciences Inc
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 Graphium Biosciences Inc filed Critical Graphium Biosciences Inc
Priority to US17/527,685 priority Critical patent/US20220168428A1/en
Publication of US20220168428A1 publication Critical patent/US20220168428A1/en
Priority to US18/182,618 priority patent/US20230346952A1/en
Assigned to GRAPHIUM BIOSCIENCES, INC. reassignment GRAPHIUM BIOSCIENCES, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: VITALITY BIOPHARMA, INC.
Assigned to VITALITY BIOPHARMA, INC. reassignment VITALITY BIOPHARMA, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BROOKE, Robert T., HARDMAN, Janee' M., ZIPP, Brandon J.
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/549Sugars, nucleosides, nucleotides or nucleic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/35Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom
    • A61K31/352Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom condensed with carbocyclic rings, e.g. methantheline 
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7012Compounds having a free or esterified carboxyl group attached, directly or through a carbon chain, to a carbon atom of the saccharide radical, e.g. glucuronic acid, neuraminic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7016Disaccharides, e.g. lactose, lactulose
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7028Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages
    • A61K31/7034Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin
    • 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
    • A61P1/10Laxatives
    • 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
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H15/00Compounds containing hydrocarbon or substituted hydrocarbon radicals directly attached to hetero atoms of saccharide radicals
    • C07H15/02Acyclic radicals, not substituted by cyclic structures
    • C07H15/04Acyclic radicals, not substituted by cyclic structures attached to an oxygen atom of the saccharide radical
    • C07H15/10Acyclic radicals, not substituted by cyclic structures attached to an oxygen atom of the saccharide radical containing unsaturated carbon-to-carbon bonds
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H15/00Compounds containing hydrocarbon or substituted hydrocarbon radicals directly attached to hetero atoms of saccharide radicals
    • C07H15/20Carbocyclic rings
    • C07H15/203Monocyclic carbocyclic rings other than cyclohexane rings; Bicyclic carbocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0014Skin, i.e. galenical aspects of topical compositions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration

Definitions

  • the present invention pertains to the field of drug development and in particular to novel cannabinoid glycoside prodrugs and methods for their production by enzyme-mediated carbohydrate transfer.
  • CBD cannabidiol
  • Cannabinoids are extremely hydrophobic in nature, complicating their use in drug formulations.
  • Non-covalent methods have been found to improve the solubility of cannabinoids by utilizing carrier carbohydrates such as cyclized maltodextrins (Jarho 1998).
  • Covalent chemical manipulations have produced novel CBD prodrugs with improved solubility (WO2009018389, WO 2012011112).
  • Even fluorine substituted CBD compounds have been created through synthetic chemical manipulations in an effort to functionalize CBD (WO2014108899).
  • the aforementioned strategies were somewhat successful in improving the solubility of CBD, but they create unnatural compositions which alter the composition and will release the unnatural prodrug moieties upon hydrolysis.
  • glycosides are capable of acting as prodrugs and also to have direct therapeutic effects.
  • Glycoside prodrugs may enable improved drug bioavailability or improved drug pharmacokinetics including more site-specific or tissue-specific drug delivery, more consistent levels of drug in the plasma, and sustained or delayed release of the drug.
  • Site-specific delivery of steroid glycosides to the colon has previously been demonstrated (Friend 1985, Friend 1984), and could enable treatment of local disorders such as inflammatory bowel disease.
  • Glycosylation of steroids enabled survival of stable bioactive molecules in the acidic stomach environment and delivery into the large intestine, where the aglycones were liberated by glycosidases produced by colonic bacteria, and then absorbed into the systemic circulation.
  • glycosidesidases are also present universally in different tissues (Conchie 1959), so delivery of glycosides by methods that bypass the digestive tract and colon, such as intravenous delivery, will enable targeted delivery to other cells and tissues that have increased expression of glycosidases.
  • delivery of alpha-glycosidase and beta-glycosidase enzymes differ throughout the intestinal tract and other tissues, and different forms of glycosides may therefore provide unique pharmacokinetic profiles, including formulations that target delivery of specific diseased areas, or targeted release at locations that can promote or restrict systemic absorption of the cannabinoids and other compounds described herein.
  • glycosides including members of classes of compounds such as hormones, antibiotics, sweeteners, alkaloids, and flavonoids. While it is generally accepted that glycosides will be more water-soluble than the aglycones, literature reviews have analyzed structure-activity relationships and determined that it is nearly impossible to define a general pattern for the biological activities of glycosides across different classes of compounds (Kren 2008).
  • CBD is glucuronidated in humans by the liver glucosyltransferases, but to date only minor activity has been demonstrated with UGT1A9 and UGT2B7 in in vitro assays (U.S. Pat. No. 8,410,064).
  • cannabinol (CBN) is efficiently glucuronidated by the Human UGT1A10 (U.S. Pat. No. 8,410,064).
  • CBN cannabinol
  • the glucuronidation of CBD is one mechanism to increase CBD solubility and facilitate removal and excretion through the kidneys.
  • cannabinol was found to be glycosylated when incubated with in vitro cell culture of Pinellia ternata (Tanaka 1993). Similarly, cannabidiol was shown to be glycosylated when incubated with tissue cultures from Pinellia ternata and Datura inoxia , yielding CBD-6′-O- ⁇ -D-glucopyranoside and CBD-(2′,6′)-O- ⁇ -D-diglucopyranoside (Tanaka 1996).
  • Cannabinoids contain a hydroxylated hydrophobic backbone, similar to the steviol backbone of steviol glycosides found in the Stevia rebaudiana plant.
  • UGT76G1 is a glucosyltransferase from Stevia that is capable of transferring a secondary glucose to the 3C-hydroxyl of the primary glycosylation on both C13-OH and C19-COOH position of the steviol glycoside, and thus its substrates include steviolmonoside, stevioside, rubusoside, RebA, RebD, RebG, RebE, etc. (Richman et al. 2005, Stevia First Corp unpublished work).
  • the substrate recognition site of UGT76G1 is capable of binding and glycosylating multiple steviol glycosides, but it was previously not known to have glycosylation activity towards any other glycosides, and there previously was no established activity of UGT76G1 towards any aglycone compounds at all.
  • UGT76G1 is capable of glycosylating steviol glycosides on the primary sugar located on both C13 hydroxyl group and the C19 carboxyl group it demonstrates bi-functional glycosylation.
  • Cyclodextrin glucanotransferase (CGTase, Toruzyme 3.0L, Novozymes Inc.) is a member of the amylase family of enzymes and is best known for its ability to cyclize maltodextrin chains. A lesser known activity of CGTase is disproportionation of linear maltodextrin chains and transfer to an acceptor sugar molecule (Li 2012).
  • cannabinoid glycosides available as cannabinoid prodrugs.
  • there is a known method for the efficient regioselective production of cannabinoid glycosides which is necessary in order to produce large, purified quantities of individual glycosides and to assess their pharmaceutical properties, including evaluation of in vivo drug pharmacokinetics and pharmacodynamics.
  • screening of glucosyltransferase enzymes from various organisms has been conducted to identify candidates for the glycosylation of cannabinoids, and to identify cannabinoid glycosides as potential prodrugs of cannabinoids, and as novel cannabinoid compositions with novel properties and functions.
  • the present invention relates to novel cannabinoid glycoside prodrugs and methods for their production by enzyme-mediated carbohydrate transfer.
  • An object of the present invention is to provide a cannabinoid glycoside prodrug.
  • a cannabinoid glycoside prodrug compound having formula (I):
  • R is H, ⁇ -D-glucopyranosyl, or 3-O- ⁇ -D-glucopyranosyl- ⁇ -D-glucopyranosyl;
  • R′ is H or ⁇ -D-glucopyranosyl, or 3-O- ⁇ -D-glucopyranosyl- ⁇ -D-glucopyranosyl;
  • A is an aglycone moiety formed through reaction of a hydroxyl group on a cannabinoid compound, an endocannabinoid compound, or a vanilloid compound, or a pharmaceutically compatible salt thereof.
  • a method for the site-specific delivery of a cannabinoid drug to a subject comprising the step of administering a cannabinoid glycoside prodrug in accordance with the present invention to a subject in need thereof.
  • a method of producing a cannabinoid glycoside comprising incubating a cannabinoid aglycone with one or more sugar donors in the presence of one or more glycosyltransferases.
  • FIG. 1A illustrates aglycones employed in the glycosylation methods of the present invention.
  • FIG. 1B illustrates the possible points of glycosylation on the aglycones.
  • FIG. 2 illustrates possible products of the glycosylation of cannabidiol (CBD).
  • FIG. 3 illustrates possible products of the glycosylation of cannabidivarin (CBDV).
  • FIG. 4 illustrates possible rotational products of the glycosylation of cannabidiol (CBD).
  • FIG. 5 illustrates possible rotational products of the glycosylation of cannabidivarin (CBDV).
  • FIG. 6 illustrates the proposed super positioning of the substrate cannabidiol (CBD) in the catalytic site of UGT76G1.
  • CBD cannabidiol
  • FIG. 7 illustrates possible products of the glycosylation of tetrahydrocannabinol (A9-THC).
  • FIG. 8 illustrates possible products of the glycosylation of cannabinol (CBN).
  • FIG. 9 illustrates possible products of the glycosylation of arachidonoyl ethanolamide (AEA).
  • FIG. 10 illustrates possible products of the glycosylation of 2-arachidonoyl ethanolamide (2-AG).
  • FIG. 11 illustrates possible products of the glycosylation of 1-arachidonoyl ethanolamide (1-AG).
  • FIG. 12 illustrates possible products of the glycosylation of N-docosahexaenoylethanolamine (DHEA).
  • DHEA N-docosahexaenoylethanolamine
  • FIG. 13 illustrates possible products of the glycosylation of capsaicin.
  • FIG. 14 illustrates possible products of the glycosylation of vanillin.
  • FIGS. 15A and 15B illustrate possible products of the glycosylation of curcumin.
  • FIG. 16 is an HPLC linetrace of the reaction products of the glycosylation of CBD.
  • FIG. 17 is an HPLC linetrace of the reaction products of the glycosylation of CBDV.
  • FIG. 18 is an HPLC linetrace of the reaction products of the glycosylation of A9-THC.
  • FIG. 19 is an HPLC linetrace of the reaction products of the glycosylation of CBN.
  • FIG. 20 is an HPLC linetrace of the reaction products of the glycosylation of 1-AG and 2-AG.
  • FIG. 21 is an HPLC linetrace of the reaction products of the glycosylation of synaptamide (DHEA).
  • FIG. 22 is an HPLC linetrace of the reaction products of the glycosylation of AEA.
  • FIG. 23 is an HPLC linetrace of the reaction products of the glycosylation of vanillin.
  • FIG. 24 is an HPLC linetrace of the reaction products of the glycosylation of capsaicin.
  • FIG. 25 is an HPLC linetrace of the reaction products of the glycosylation of CBDg1 (VB104) with the glycosyltransferase UGT76G1.
  • FIG. 26 is an HPLC linetrace of the reaction products of the glycosylation of CBDg1 (VB104) with the glycosyltransferase Os03g0702000
  • FIG. 27 is a 1 NMR spectrum of an isolated product, VB104, of the glycosylation of CBD.
  • FIG. 28 is a 1 NMR spectrum of an isolated product, VB110 of the glycosylation of CBD.
  • FIG. 29 is a plot of C18 retention times vs c Log P values for selected cannabinoids and cannabinoid glycosides.
  • FIG. 30A is a graphical presentation of the results of the analysis of the small intestine extracts of a bioavailability assay.
  • FIG. 30B is a graphical presentation of the results of the analysis of the large intestine extracts of a bioavailability assay
  • CBD Cannabidiol
  • CBDV Cannabidivarin
  • CBDA Cannabidiolic acid
  • glucosecopyranoside is used for naming molecules and is shorthand for a 3-D-glucose attached through the hydroxyl at the 1-position (the anomeric carbon) of the glucose to the aglycone.
  • glycoside compound is used in the present application to refer to the non-glycosidic portion of a glycoside compound.
  • prodrug refers to a compound that, upon administration, must undergo a chemical conversion by metabolic processes before becoming an active pharmacological agent.
  • cannabinoid glycoside prodrug refers generally to the glycosides of cannabinoid compounds, endocannabinoid compounds and vanilloid compounds.
  • the cannabinoid glycoside prodrug undergoes hydrolysis of the glycosidic bond, typically by action of a glycosidase, to release the active cannabinoid, endocannabinoid or vanilloid compounds to a desired site in the body of the subject.
  • the cannabinoid glycoside prodrug of the present invention may also be referred to using the term “cannaboside”.
  • cannabinoid is used in the present application to refer generally to compounds found in cannabis and which act on cannabinoid receptors.
  • Cannabinoid compounds include, but are not limited to, cannabidiol (CBD), cannabidivarin (CBDV), cannabigerol (CBG), tetrahydrocannabinol ( ⁇ 9-THC or THC), cannabinol (CBN), cannabidiolic acid (CBDA), and tetrahydrocannabivarin (THCV).
  • CBD cannabidiol
  • CBDV cannabidivarin
  • CBG cannabigerol
  • THC cannabinol
  • CBDA cannabidiolic acid
  • THCV tetrahydrocannabivarin
  • Particularly preferred cannabinoids compounds are CBD, CBDV, THC and CBN.
  • endocannabinoid is used in the present application to refer to compounds including arachidonoyl ethanolamide (anandamide, AEA), 2-arachidonoyl ethanolamide (2-AG), 1-arachidonoyl ethanolamide (1-AG), and docosahexaenoyl ethanolamide (DHEA, synaptamide), oleoyl ethanolamide (OEA), eicsapentaenoyl ethanolamide, prostaglandin ethanolamide, docosahexaenoyl ethanolamide, linolenoyl ethanolamide, 5(Z),8(Z),11(Z)-eicosatrienoic acid ethanolamide (mead acid ethanolamide), heptadecanoul ethanolamide, stearoyl ethanolamide, docosaenoyl ethanolamide, nervonoyl ethanolamide, tricosanoyl ethanolamide, lignoceroyl ethanolamide, my
  • vanilloid is used in the present application to refer to compounds comprising a vanillyl group and which act on vanilloid receptors like TRPV1.
  • vanilloid compounds include, but are not limited to, vanillin, capsaicin and curcumin.
  • the term “about” refers to a +/ ⁇ 10% variation from the nominal value. It is to be understood that such a variation is always included in a given value provided herein, whether or not it is specifically referred to.
  • subject or “patient” as used herein refers to an animal in need of treatment.
  • the animal is a human.
  • cannabinoids, endocannabinoids and vanilloids are employed as substrates for glucosyltransferases to which one or more sugar molecules are attached to create novel cannabinoid glycoside prodrugs.
  • the resulting cannabinoid glycoside prodrugs demonstrate site-specific or tissue-specific delivery, improved aqueous solubility for improved pharmacological delivery, and/or sustained or delayed release of the cannabinoid, endocannabinoid and vanilloid drug molecules.
  • the cannabinoid glycoside prodrugs are converted upon hydrolysis of the glycosidic bond to provide the active cannabinoid, endocannabinoid and vanilloid drug. Accordingly, the present invention has demonstrated that glycosides with a hydrophobic aglycone moiety undergo glucose hydrolysis in the gastrointestinal tract or in tissues having increased expression of glycosidases, yielding the hydrophobic cannabinoid compound in the targeted tissue or organ.
  • glycosides are commonly acid-hydrolyzed in the stomach or cleaved by glycosidase enzymes in the intestinal tract, including by alpha-glycosidases and beta-glycosidases, which are expressed by intestinal microflora across different regions of the intestine. Accordingly, glycosides are hydrolyzed upon ingestion to release the desired compound into the intestines or target tissues.
  • glycosylation of cannabinoid drugs provides cannabinoid glycoside prodrugs capable of persisting in the acidic stomach environment upon oral administration, thereby allowing delivery of the prodrug into the large intestine, where the cannabinoid aglycones can be liberated by glycosidases produced by colonic bacteria.
  • glycosylation of cannabinoid drugs provides cannabinoid glycoside prodrugs suitable for targeted delivery to tissues having increased expression of glycosidases.
  • the cannabinoid glycoside prodrug formulation Upon parenteral administration of the cannabinoid glycoside prodrug formulation to the subject, the cannabinoid aglycones are liberated by the glycosidases in the target tissues.
  • the cannabinoid glycoside prodrug are also useful as pharmaceutical agents without glucose cleavage, where they exhibit novel pharmacodynamic properties compared to the parent compound alone.
  • the increased aqueous solubility of the cannabinoid glycoside prodrugs of the present invention also enables new formulations for delivery in transdermal or aqueous formulations that would not have been achievable if formulating hydrophobic cannabinoid, endocannabinoid and vanilloid molecules.
  • cannabinoid glycoside prodrug compounds having formula (I):
  • R is H, ⁇ -D-glucopyranosyl, or 3-O- ⁇ -D-glucopyranosyl- ⁇ -D-glucopyranosyl
  • R′ is H or ⁇ -D-glucopyranosyl, or 3-O- ⁇ -D-glucopyranosyl- ⁇ -D-glucopyranosyl
  • A is an aglycone moiety formed through reaction of a hydroxyl group on a cannabinoid compound, an endocannabinoid compound, or a vanilloid compound.
  • A is A′, A′′ or A′′′;
  • A′ is:
  • G is H, ⁇ -D-glucopyranosyl, 3-O- ⁇ -D-glucopyranosyl- ⁇ -D-glucopyranosyl, or ⁇ -D-glucopyranosyl-(1 ⁇ 3)- ⁇ -D-glucopyranosyl-(1 ⁇ 3)-D-glucopyranosyl; or a pharmaceutically compatible salt thereof.
  • the cannabinoid glycoside prodrug is a glycoside of a cannabinoid, wherein the prodrug has the formula (I′):
  • R is H, ⁇ -D-glucopyranosyl, or 3-O- ⁇ -D-glucopyranosyl- ⁇ -D-glucopyranosyl
  • R′ is H, ⁇ -D-glucopyranosyl, or 3-O- ⁇ -D-glucopyranosyl- ⁇ -D-glucopyranosyl
  • A′ is:
  • G is ⁇ -D-glucopyranosyl, 3-O- ⁇ -D-glucopyranosyl- ⁇ -D-glucopyranosyl, or ⁇ -D-glucopyranosyl-(1-3)- ⁇ -D-glucopyranosyl-(1-3)-D-glucopyranosyl.
  • Compounds of Formula (I′) include the compounds listed in Tables 1 to 4.
  • CBD-glycosides falling within the scope of Formula (I′), produced by the glycosylation of CBD (VB101) in accordance with the present invention, include:
  • CBDV cannabidivarin-glycosides falling within the scope of Formula (I′), produced by the glycosylation of CBDV (VB201) in accordance with the present invention, include:
  • Exemplary tetrahydrocannabinol ( ⁇ 9-THC)-glycosides falling within the scope of Formula (I′), produced by the glycosylation of ⁇ 9-THC (VB301) in accordance with the present invention, include:
  • Exemplary cannabinol (CBN)-glycosides falling within the scope of Formula (I′), produced by the glycosylation of CBN (VB401) in accordance with the present invention include:
  • the cannabinoid glycoside prodrug is a glycoside of an endocannabinoid, the prodrug having the formula (I′′):
  • R is H, ⁇ -D-glucopyranosyl, or 3-O- ⁇ -D-glucopyranosyl- ⁇ -D-glucopyranosyl;
  • R′ is H, ⁇ -D-glucopyranosyl, or 3-O- ⁇ -D-glucopyranosyl- ⁇ -D-glucopyranosyl;
  • A′′ is:
  • Compounds of Formula (I′′) include the compounds listed in Tables 5 to 8.
  • AEA arachidonoyl ethanolamide
  • Exemplary 2-arachidonoyl ethanolamide (2-AG)-glycosides falling within the scope of Formula (I′′), produced by the glycosylation of 2-AG (VB601) in accordance with the present invention, include:
  • Exemplary 1-arachidonoyl ethanolamide (1-AG)-glycosides falling within the scope of Formula (I′′), produced by the glycosylation of 1-AG (VB701) in accordance with the present invention, include:
  • DHEA-glycosides falling within the scope of Formula (I′′), produced by the glycosylation of DHEA (VB801) in accordance with the present invention include:
  • the cannabinoid glycoside prodrug is a glycoside of a vanilloid, the prodrug having the formula (I′′′):
  • R is H, ⁇ -D-glucopyranosyl, or 3-O- ⁇ -D-glucopyranosyl- ⁇ -D-glucopyranosyl;
  • R′ is H or ⁇ -D-glucopyranosyl, or 3-O- ⁇ -D-glucopyranosyl- ⁇ -D-glucopyranosyl; and, wherein A′′′ is:
  • Compounds of Formula (I′′′) include the compounds listed in Tables 9 to 11.
  • Exemplary capsaicin-glycosides falling within the scope of Formula (I′′′), produced by the glycosylation of capsaicin (VB901) in accordance with the present invention, include:
  • Exemplary vanillin-glycosides falling within the scope of Formula (I′′′), produced by the glycosylation of vanillin (VB1001) in accordance with the present invention include:
  • curcumin-glycosides falling within the scope of Formula (I′′′), produced by the glycosylation of curcumin (VB1101) in accordance with the present invention include:
  • a method for the site-specific delivery of a cannabinoid drug to a subject comprising the step of administering to a subject in need thereof one or more cannabinoid glycoside prodrugs in accordance with the present invention.
  • the site of delivery is the large intestine.
  • the site of delivery is the rectum.
  • the site of delivery is the liver.
  • the site of delivery is the skin.
  • a method for facilitating the transport of a cannabinoid drug to the brain through intranasal, stereotactic, or intrathecal delivery, or delivery across the blood brain barrier of a subject comprising administering a cannabinoid glycoside prodrug in accordance with the present invention to a subject in need thereof.
  • the cannabinoid glycoside prodrugs are useful in the treatment of conditions that benefit from or can be ameliorated with the administration of a cannabinoid drug.
  • Conditions that can be treated or ameliorated through the administration of cannabinoid glycoside prodrugs of the present invention include but are not limited to, inflammatory bowel disease including induction of remission from Crohn's disease, and colitis and induction of remission from ulcerative colitis.
  • cannabinoid glycoside prodrugs of the present invention are decreased inflammation of the intestines and rectum, decreased pain in the intestines, rectum, as well as decrease in neuropathic pain and abdominal pain, and inhibition of proliferation or cytotoxicity against colorectal cancer.
  • Additional treatment indications, effects, or applications for cannabinoids or cannabinoid glycosides may include but are not limited to anorexia, nausea, emesis, pain, wasting syndrome, HIV-wasting, chemotherapy induced nausea and vomiting, epilepsy, schizophrenia, irritable bowel syndrome, cramping, spasticity, seizure disorders, alcohol use disorders, substance abuse disorders, addiction, cancer, amyotrophic lateral sclerosis, glioblastoma multiforme, glioma, increased intraocular pressure, glaucoma, cannabis use disorders, Tourette's syndrome, dystonia, multiple sclerosis, white matter disorders, demyelinating disorders, chronic traumatic encephalopathy, leukoencephalopathies, Guillain-Barre syndrome, inflammatory bowel disorders, gastrointestinal disorders, bacterial infections, MRSA, sepsis, septic shock, viral infections, arthritis, dermatitis, Rheumatoid arthritis, systemic lupus erythematosus, anti-
  • the cannabinoid glycoside prodrug is administered in a pharmaceutical composition further comprising a pharmaceutically acceptable carrier, diluent, excipient, or adjuvant.
  • the pharmaceutical compositions comprise one or more cannabinoid glycoside prodrugs and one or more pharmaceutically acceptable carriers, diluents, excipients and/or adjuvants.
  • the pharmaceutical compositions can be formulated for administration by a variety of routes including but not limited to oral, topical, rectal, parenteral, and intranasal administration.
  • compositions may comprise from about 1% to about 95% of a cannabinoid glycoside prodrug of the invention.
  • Compositions formulated for administration in a single dose form may comprise, for example, about 20% to about 90% of the cannabinoid glycoside prodrug of the invention, whereas compositions that are not in a single dose form may comprise, for example, from about 5% to about 20% of the cannabinoid glycoside prodrug of the invention.
  • unit dose forms include tablets, ampoules, dragees, suppositories, and capsules.
  • compositions are formulated for oral administration.
  • Pharmaceutical compositions for oral administration can be formulated, for example, as tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsion hard or soft capsules, or syrups or elixirs.
  • Such compositions can be prepared according to standard methods known in the art for the manufacture of pharmaceutical compositions and may contain one or more agents selected from the group of sweetening agents, flavouring agents, colouring agents and preserving agents in order to provide pharmaceutically elegant and palatable preparations.
  • Tablets contain the active ingredient in admixture with suitable non-toxic pharmaceutically acceptable excipients including, for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, such as corn starch, or alginic acid; binding agents, such as starch, gelatine or acacia, and lubricating agents, such as magnesium stearate, stearic acid or talc.
  • the tablets can be uncoated, or they may be coated by known techniques in order to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period.
  • a time delay material such as glyceryl monosterate or glyceryl distearate may be employed to further facilitate delivery of the drug compound to the desired location in the digestive tract.
  • compositions for oral use can also be presented as hard gelatine capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatine capsules wherein the active ingredient is mixed with water or an oil medium such as peanut oil, liquid paraffin or olive oil.
  • an inert solid diluent for example, calcium carbonate, calcium phosphate or kaolin
  • an oil medium such as peanut oil, liquid paraffin or olive oil.
  • compositions formulated as aqueous suspensions contain the active compound(s) in admixture with one or more suitable excipients, for example, with suspending agents, such as sodium carboxymethylcellulose, methyl cellulose, hydropropylmethylcellulose, sodium alginate, polyvinylpyrrolidone, hydroxypropyl- ⁇ -cyclodextrin, gum tragacanth and gum acacia; dispersing or wetting agents such as a naturally-occurring phosphatide, for example, lecithin, or condensation products of an alkylene oxide with fatty acids, for example, polyoxyethyene stearate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example, hepta-decaethyleneoxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol for example, polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids
  • the aqueous suspensions may also contain one or more preservatives, for example ethyl, or n-propyl p-hydroxy-benzoate, one or more colouring agents, one or more flavouring agents or one or more sweetening agents, such as sucrose, stevia , or saccharin.
  • preservatives for example ethyl, or n-propyl p-hydroxy-benzoate
  • colouring agents for example ethyl, or n-propyl p-hydroxy-benzoate
  • flavouring agents such as sucrose, stevia , or saccharin.
  • sweetening agents such as sucrose, stevia , or saccharin.
  • compositions can be formulated as oily suspensions by suspending the active compound(s) in a vegetable oil, for example, arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin.
  • the oily suspensions may contain a thickening agent, for example, beeswax, hard paraffin or cetyl alcohol.
  • Sweetening agents such as those set forth above, and/or flavouring agents may be added to provide palatable oral preparations.
  • These compositions can be preserved by the addition of an anti-oxidant such as ascorbic acid.
  • the pharmaceutical compositions can be formulated as a dispersible powder or granules, which can subsequently be used to prepare an aqueous suspension by the addition of water.
  • Such dispersible powders or granules provide the active ingredient in admixture with one or more dispersing or wetting agents, suspending agents and/or preservatives. Suitable dispersing or wetting agents and suspending agents are exemplified by those already mentioned above. Additional excipients, for example, sweetening, flavouring and colouring agents, can also be included in these compositions.
  • compositions of the invention can also be formulated as oil-in-water emulsions.
  • the oil phase can be a vegetable oil, for example, olive oil or arachis oil, or a mineral oil, for example, liquid paraffin, or it may be a mixture of these oils.
  • Suitable emulsifying agents for inclusion in these compositions include naturally-occurring gums, for example, gum acacia or gum tragacanth; naturally-occurring phosphatides, for example, soy bean, lecithin; or esters or partial esters derived from fatty acids and hexitol, anhydrides, for example, sorbitan monoleate, and condensation products of the said partial esters with ethylene oxide, for example, polyoxyethylene sorbitan monoleate.
  • the emulsions can also optionally contain sweetening and flavouring agents.
  • compositions can be formulated as a syrup or elixir by combining the active ingredient(s) with one or more sweetening agents, for example glycerol, propylene glycol, sorbitol or sucrose.
  • sweetening agents for example glycerol, propylene glycol, sorbitol or sucrose.
  • Such formulations can also optionally contain one or more demulcents, preservatives, flavouring agents and/or colouring agents.
  • the glycoside prodrugs may be combined with other ingredients or substances that have glycosidase activity, or that may in other ways alter drug metabolism and pharmacokinetic profile of various compounds in vivo, including ones in purified form as well as such compounds found within food, beverages, and other products.
  • the cannabinoid glycoside prodrug is administered in combination with, or formulated together with, substances that have direct glycosidase activity, or that contribute to modifications to the gut microflora that will alter the glycosidase activity in one or more regions of the intestines. Examples of such compositions include, but are not limited to, yogurt, prebiotics, probiotics, or fecal transplants.
  • compositions are formulated for parenteral administration.
  • parenteral as used herein includes subcutaneous injections, intravenous, intramuscular, intrathecal, intrasternal injection or infusion techniques.
  • Parenteral pharmaceutical compositions can be formulated as a sterile injectable aqueous or oleaginous suspension according to methods known in the art and using one or more suitable dispersing or wetting agents and/or suspending agents, such as those mentioned above.
  • the sterile injectable preparation can be a sterile injectable solution or suspension in a non-toxic parentally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol.
  • Acceptable vehicles and solvents that can be employed include, but are not limited to, water, Ringer's solution, lactated Ringer's solution and isotonic sodium chloride solution.
  • sterile, fixed oils which are conventionally employed as a solvent or suspending medium
  • a variety of bland fixed oils including, for example, synthetic mono- or diglycerides.
  • Fatty acids such as oleic acid can also be used in the preparation of injectables.
  • cannabinoids Due to the highly lipophilic nature of cannabinoids, these molecules are typically poorly absorbed through membranes such as the skin of mammals, including humans, and the success of transdermally administering therapeutically effective quantities of cannabinoid to a subject in need thereof within a reasonable time frame and over a suitable surface area has been substantially limited. It is therefore proposed that the cannabinoid glycoside prodrugs of the present invention, through conjugation of the hydrophobic cannabinoid aglycone to the hydrophilic glycosidic moieties, provide a molecule having an amphiphilic character favourable for passive diffusion which should be more readily absorbed through the skin.
  • the pharmaceutical compositions are formulated for topical administration.
  • topical formulations may be presented as, for example, aerosol sprays, powders, sticks, granules, creams, liquid creams, pastes, gels, lotions, ointments, on sponges or cotton applicators, or as a solution or a suspension in an aqueous liquid, a non-aqueous liquid, an oil-in-water emulsion, or a water-in-oil liquid emulsion.
  • Topical pharmaceutical compositions can be formulated with thickening (gelling) agents.
  • the thickening agent used herein may include anionic polymers such as polyacrylic acid (CARBOPOL® by Noveon, Inc., Cleveland, Ohio), carboxypolymethylene, carboxymethylcellulose and the like, including derivatives of Carbopol® polymers, such as Carbopol® Ultrez 10, Carbopol® 940, Carbopol® 941, Carbopol® 954, Carbopol® 980, Carbopol® 981, Carbopol® ETD 2001, Carbopol® EZ-2 and Carbopol® EZ-3, and other polymers such as Pemulen® polymeric emulsifiers, and Noveon® polycarbophils.
  • Thickening agents or gelling agents are present in an amount sufficient to provide the desired rheological properties of the composition.
  • Topical pharmaceutical compositions can be formulated with a penetration enhancer.
  • penetration enhancing agents include C 8 -C 22 fatty acids such as isostearic acid, octanoic acid, and oleic acid; C 8 -C 22 fatty alcohols such as oleyl alcohol and lauryl alcohol; lower alkyl esters of C 8 -C 22 fatty acids such as ethyl oleate, isopropyl myristate, butyl stearate, and methyl laurate; di(lower)alkyl esters of C 6 -C 22 diacids such as diisopropyl adipate; monoglycerides of C 8 -C 22 fatty acids such as glyceryl monolaurate; tetrahydrofurfuryl alcohol polyethylene glycol ether; polyethylene glycol, propylene glycol; 2-(2-ethoxyethoxyl)ethanol; diethylene glycol monomethyl ether; alkylaryl ethers
  • the topical pharmaceutical compositions can further comprise wetting agents (surfactants), lubricants, emollients, antimicrobial preservatives, and emulsifying agents as are known in the art of pharmaceutical formations.
  • wetting agents surfactants
  • lubricants emollients
  • antimicrobial preservatives emulsifying agents
  • Transdermal delivery of the cannabinoid glycoside prodrug can be further facilitated through the use of a microneedle array drug delivery system.
  • compositions and methods of preparing pharmaceutical compositions are known in the art and are described, for example, in “ Remington: The Science and Practice of Pharmacy ” (formerly “ Remingtons Pharmaceutical Sciences ”); Gennaro, A., Lippincott, Williams & Wilkins, Philadelphia, Pa. (2000).
  • compositions of the present invention described above include one or more cannabinoid glycoside prodrugs of the invention in an amount effective to achieve the intended purpose.
  • therapeutically effective dose refers to the amount of the cannabinoid glycoside prodrug that improves the status of the subject to be treated, for example, by ameliorating the symptoms of the disease or disorder to be treated, preventing the disease or disorder, or altering the pathology of the disease. Determination of a therapeutically effective dose of a compound is well within the capability of those skilled in the art.
  • cannabinoid glycosides can be combined to enable simultaneous delivery of multiple cannabinoids in a site-specific manner, including THC and CBD, whose effects in some ways may be synergistic (Russo 2006).
  • the pharmaceutical composition comprises one or more CBD-glycosides and one or more THC-glycosides formulated together in a single dosage form.
  • the exact dosage to be administered to a subject can be determined by the practitioner, in light of factors related to the subject requiring treatment. Dosage and administration are adjusted to provide desired levels of the cannabinoid glycoside prodrug and/or the cannabinoid drug compound obtained upon hydrolysis of the prodrug. Factors which may be taken into account when determining an appropriate dosage include the severity of the disease state, general health of the subject, age, weight, and gender of the subject, diet, time and frequency of administration, drug combination(s), reaction sensitivities, and tolerance/response to therapy. Dosing regimens can be designed by the practitioner depending on the above factors as well as factors such as the half-life and clearance rate of the particular formulation.
  • a method of producing a cannabinoid glycoside comprising incubating a cannabinoid aglycone with one or more sugar donors in the presence of one or more glycosyltransferases.
  • the one or more glycosyltransferases is a UGT76G1 or UGT76G1-like glucosyltransferase. In one embodiment, the one or more glycosyltransferases comprise a UGT76G1 or UGT76G1-like glucosyltransferase and a Os03g0702000 or Os03g0702000-like glucosyltransferase.
  • the one or more sugar donors are selected from the group consisting of UDP-glucose, UDP-glucuronic acid, UDP-mannose, UDP-fructose, UDP-xylose, UDP-rhamnose, UDP-fluoro-deoxyglucose, and combinations thereof.
  • the sugar donor is UDP-glucose.
  • the cannabinoid aglycone is a cannabinoid, an endocannabinoid, or a vanilloid.
  • the cannabinoid glycoside prodrug produced by the methods of the present invention is a compound of the Formula (I).
  • the method of producing a cannabinoid glycoside comprises incubating a cannabinoid aglycone with UDP-glucose, in the presence of a UGT76G1 or UGT76G1-like glucosyltransferase under conditions that allow for glycosylation.
  • the method of producing a cannabinoid glycoside comprises incubating a cannabinoid aglycone with one or more sugar donors in the presence of a first glycosyltransferase and a second glycosyltransferase under conditions which allow for glycosylation.
  • sugar donor is UDP-glucose
  • the first glycosyltransferase is a UGT76G1 or UGT76G1-like glucosyltransferase
  • the second glycosyltransferase is a Os03g0702000 or Os03g0702000-like glucosyltransferase.
  • the method of producing a cannabinoid glycoside comprises incubating a cannabinoid aglycone with UDP-glucose in the presence of a UGT76G1 or UGT76G1-like glucosyltransferase and Os03g0702000 or Os03g0702000-like glucosyltransferase under conditions which allow for glycosylation.
  • the method of producing a cannabinoid glycoside comprises incubating a cannabinoid aglycone with maltodextrin, in the presence of a cyclodextrin glucanotransferase under conditions that allow for glycosylation.
  • the method of producing a cannabinoid glycoside comprises incubating a cannabinoid aglycone with UDP-glucose and maltodextrin in the presence of a UGT76G1 or UGT76G1-like glucosyltransferase and cyclodextrin glucanotransferase under conditions which allow for glycosylation.
  • the glycosyltransferase employed in the methods of producing the cannabinoid glycoside is UGT76G1 or UGT76G1-like glucosyltransferase.
  • the UGT76G1 or UGT76G1-like glucosyltransferase comprises the sequence as set forth in SEQ ID NO:1, 3, 5 or 7.
  • the glycosyltransferase employed in the methods of producing the cannabinoid glycoside is Os03g0702000 or Os03g0702000-like glucosyltransferase.
  • the Os03g0702000 or Os03g0702000-like glucosyltransferase comprises the sequence as set forth in SEQ ID NO:9.
  • the method of producing the cannabinoid glycoside further comprises incubating with sucrose synthase.
  • the sucrose synthase comprises the sequence as set forth in SEQ ID NO: 15, 17, 19, 21, 23 or 25.
  • the method for the production of a cannabinoid glycoside prodrug comprises expressing one or more of the glycosyltransferases in a cell or plant which produces the cannabinoid aglycone and isolating the cannabinoid glycoside prodrug.
  • glycosylated cannabinoids can act as efficient prodrugs for selective delivery of cannabinoids to desired tissues where the glucose molecules can be hydrolyzed to release the aglycone cannabinoids. Additionally the glycosylations promote stability of CBD and CBDV by protecting them from oxidation and ring-closure of the C6′-hydroxyl group, which prevents degradation into ⁇ 9-THC or ⁇ 9-THCV, respectively, and subsequently into cannabinol (CBN) or cannabinavarin (CBNV), respectively
  • the primary detoxification mechanism for cannabinoids in humans is CYP450 mediated hydroxylation of the C7 methyl group of CBD and CBDV, or the C11 methyl group of THC and CBN, glycosylation of the acceptor hydroxyl groups of the cannabinoid resorcinol ring may afford protection from C7/C11 hydroxylation and subsequent elimination from the body due to steric hindrance preventing the cannabinoid-glycoside from binding in the CYP450 active site.
  • the hydroxyl groups of CBD are thought to facilitate the binding to the detoxification cytochrome P450 CYP3A4 in the epithelium of the small intestine (Yamaori 2011). Reduced degradation or metabolism in the stomach and small intestine due to these effects could also lead to higher total bioavailability of any glycosylated product upon oral delivery.
  • glycoside prodrugs may enable stable drug formulations that are resistant to abuse, due to the potential for their primary biological effects to only occur after oral ingestion. As most abuse-deterrent compounds are simply mixing or formulation based deterrents, they can still be compromised by simple physical and chemical methods. As one example, the beta-glycosides described herein will only release the aglycone upon the action of beta-glycosidase enzymes.
  • Beta-glycosidases are known to be secreted by microbes that occupy the large intestines of mammals, therefore upon oral ingestion the glycoside prodrugs will remain glycosylated until they reach the large intestine.
  • a similar approach may be used for abuse-resistant, abuse-deterrent, and site-specific delivery of other compounds through glycosylation. It has been found that the UGT76G1 enzyme (SEQ ID NO.1) from Stevia rebaudiana transfers a glucose molecule from the sugar donor UDP-glucose (UDPG) to the hydroxyl groups of CBD to create novel CBD-O-glycosides (Table 1, FIGS. 2 & 4 ).
  • the UDPG is inverted by UGT76G1 to produce ⁇ -D-glucose residues covalently linked through the to the hydroxyl acceptor sites on CBD.
  • UGT76G1 open reading frame (ORF) codon optimization was performed (SEQ ID NOs. 4 and 6) for expression in Pichia pastoris . Similar to its activity towards steviol glycosides, UGT76G1 is highly productive and has an equilibrium constant (Keq) for CBD of ⁇ 24. Through experimentation and analysis it was determined that UGT76G1 has the unique ability to apply multiple glucose moieties to the CBD molecule.
  • CBD-glycoside product mobility groups also suggest that CBD can dock in the UGT76G1 active site both forwards and backwards creating a cis-like-conformation for the glycosylations relative to the cannabinoid backbone (mechanism depicted in FIG. 3 ), or possibly the rotational freedom about the bond at C1′ (C6 described by Mazur 2009) allows the hydroxyl group to rotate after glycosylation, placing the other hydroxyl group adjacent to the UDPG in the active site and creating a trans-like-conformation for the glycosylations on the cannabinoid backbone (mechanism depicted in FIG. 4 ).
  • Potential CBD molecular docking in the active site of UGT76G1 is depicted in FIG. 6 where CBD is superpositioned over the bi-functional substrate for UGT76G1, Rebaudioside E (RebE) ( FIG. 6 ).
  • CBDV was incubated with UGT76G1 and UDPG to test for glycosylation activity.
  • CBDV depletion was observed upon HPLC analysis, in addition to the appearance of four additional product peak mobility groups, which were dependent on addition of both UGT76G1 and UDPG.
  • the four new products formed displayed the same absorbance characteristics as CDBV and were determined to be the primary glycosides CBDV-2′-O-glucopyranosides, CBDV-6′-O-glucopyranosides, and the secondary glycosides CBDV-2′-O-(3-1)-diglucopyranoside, and CBDV-6′-O-(3-1)-diglucopyranoside (compounds VB202, VB206, VB204 and VB208, respectively, Table 2). With additional reaction time it was determined that higher order glycoside products were also formed. CBDV-glycoside production was similar to CBD-glycosides from UGT76G1 (Table 2), and proceeded to completion with a K eq ⁇ 24. Given the number of CBDV-glycoside products, UGT76G1 transfers multiple glucose molecules onto CBDV on both C2′ and C6′ hydroxyl groups, as well as onto the primary and secondary glycosylations.
  • the cannabinoid backbone is recognized in the active site of UGT76G1 with the ⁇ 9-THC C1 hydroxyl group situated towards the UDPG sugar donor (pyran numbering, FIG. 1B ).
  • UGT76G1 demonstrated glycosylation activity for all other phytocannabinoids analyzed, it was also tested for glycosylation activity against cannabinol (CBN). Effective glycosylation of CBN by UGT76G1 was observed, in a similar pattern to ⁇ 9-THC, as both share a single hydroxyl recipient group at the C1 position of the resorcinol ring. The activity seen with UGT76G1 is consistent with a broad recognition of cannabinoids by the enzyme active site.
  • CBN cannabinol
  • Cannabinoid substrates may be inserted into this UGT76G1 glycosylation reaction infrastructure to generate novel cannabinoid-glycosides, given they possess hydroxyl groups in similar positions on the cannabinoid backbone.
  • Ideal candidates are cannabigerol (CBG), cannabichromene (CBC), cannabidiol hydroxyquinone (CBDHQ), HU-331, other isomers of ⁇ 9-THC such as ⁇ 8-THC, etc., and synthetic analogues of ⁇ 9-THC such as HU-210.
  • the UGT enzyme Os03g0702000 (SEQ ID NO.9) from Oryza sativa is also capable of transferring an additional glucose moiety from UDP-glucose onto the C2-hydroxyl of the primary sugar (Tables 1-11, FIGS. 7-9 & 12-14 ).
  • This glycosylation activity is consistent with the activity of UGT Os03g0702000 towards steviol glycosides in establishing C2-hydroxyl secondary glycosylations (2 ⁇ 1 connectivity) on existing primary glucose residues.
  • This secondary glycosylation was observed with CBDV (Table 2, FIG. 3 ), and THC (Table 2, FIG.
  • cyclodextrin-glucanotransferase In addition to the UDPG-dependent glucosyltransferase activity, cyclodextrin-glucanotransferase (CGTase, Toruzyme 3.0L, trademark of Novozymes Inc.) is capable of transferring a short ⁇ -(1-4)-maltodextrin chain onto the hydroxyl groups of cannabinoids.
  • the CGTase is also capable of glycosylating primary and secondary glycosylations established by UGT76G1 and Os03g0702000, resulting in carbohydrate attachments that start with ⁇ -D-glucose molecules, but terminating in ⁇ -D-glucose molecules termed ⁇ -primed- ⁇ -glucosyl (Tables 1-11).
  • ⁇ -glycosylation by cyclodextrin glucanotransferase mediated maltodextrin transfer can occur on any of the hydroxyl groups of the primary or secondary sugars covalently linked to the cannabinoid.
  • cyclodextrin glucanotransferase mediated maltodextrin transfer can occur on any of the hydroxyl groups of the primary or secondary sugars covalently linked to the cannabinoid.
  • UGT76G1 and Os03g0702000 may be used to produce the same glycosylation of cannabinoids.
  • Suitable enzymes for establishing the primary glycosylation similar to UGT76G1 are additional members of the UGT76 clade such as UGT76G2 or UGT76H1.
  • BLAST results with the UGT76G1 protein sequence yield a maximum homology of 49% identity, as much as 66% positives (similar identity). Ideal candidates may have low overall peptide identity or similarity, but will likely have conserved amino acids at the opening adjacent to the UDPG catalytic site.
  • This sequence is exemplified by a leucine at position 379, and a broader peptide sequence of SDFGLDQ (AA's 375 to 381 of UGT76G1).
  • Suitable enzymes for producing the secondary glycosylation of Os03g0702000 are members of the UGT91 clade, including UGT91 D1 and UGT91 D2.
  • Glucuronic acid is the predominant nucleotide sugar utilized by phase-II detoxification UGTs in the liver, and cannabinoid-glucuronides are a common detoxification product.
  • nucleotide sugars which could be used to donate carbohydrate moieties to create novel glycosides with similar properties include UDP-glucuronic acid, UDP-mannose, UDP-fructose, UDP-xylose, UDP-rhamnose, UDP-fluorodeoxyglucose, etc.
  • nucleotide sugars can also be used in combination to create glycosides that contain multiple types of residues on the same aglycone backbone.
  • Alternative strategies to further improve the solubility and delivery of cannabinoids and other compounds described herein include their glycosylation and then functionalizing the sugar moieties with additional ligands or modifications. Examples of this include sulfation, myristoylation, phosphorylation, acetylation, etc.
  • endocannabinoids such as AEA, 2-AG, 1-AG, and synaptamide are glycosylated by UGT76G1, it is hypothesized that similar endocannabinoids will also be suitable substrates for glycosylation by UGT76G1.
  • endocannabinoid candidates that are likely to be glycosylated by UGT76G1 include oleoyl ethanolamide (OEA), eicsapentaenoyl ethanolamide, prostaglandin ethanolamide, docosahexaenoyl ethanolamide, linolenoyl ethanolamide, 5(Z),8(Z),11(Z)-eicosatrienoic acid ethanolamide (mead acid ethanolamide), heptadecanoul ethanolamide, stearoyl ethanolamide, docosaenoyl ethanolamide, nervonoyl ethanolamide, tricosanoyl ethanolamide, lignoceroyl ethanolamide, myristoyl ethanolamide, pentadecanoyl ethanolamide, palmitoleoyl ethanolamide, docosahexaenoic acid (DHA), and similar compounds.
  • OOA oleoyl ethanolamide
  • glycolipids may have a wide range of commercial uses, ranging from pharmaceutical use as a novel endocannabinoid drug with improved solubility and pharmacokinetic properties, to use as an antibacterial agent, to use as a detergent similar to other glycolipids, etc.
  • TRPV1 toll-like vanilloid receptor type 1
  • CBD cannabinoids and botanical extracts
  • CBN cannabigerol
  • CBG cannabigerol
  • TRPs transient receptor potential channels
  • TRPV1 Although stimulation of TRPV1 leads to vasodilation and inflammation, capsaicin and its analogues act to desensitize the receptors to stimulants, and provide potent anti-inflammatory effects (Bisogno 2001). Analogous effects may occur with TRPA1 in addition to other TRPs. For CBD, this may occur at concentrations that are lower than what is required for binding of cannabinoid receptors, and at concentrations that are within the range of those typically attained in human clinical testing and use.
  • CBD has been shown to inhibit fatty acid amide hydroxylase (FAAH), the enzyme responsible for facilitating the metabolism of the endocannabinoid anandamide (Watanabe, 1998; DE e Petrocellis 2010).
  • FAAH fatty acid amide hydroxylase
  • UGT76G1 would be capable of glycosylating many different ligands of the same TRPs, including TRPM8, TRPV2, TRPA1, and TRPV1.
  • Capsaicin is capable of contorting into a CBD-like structure (Bisogno 2001), therefore it was postulated that capsaicin was likely to be a suitable substrate for glycosylation by UGT76G1.
  • UGT76G1 is capable of glycosylating the vanilloid moiety of capsaicin in a structurally identical way to PaGT3 from Phytolacca americana (Noguchi 2009).
  • the glycosylated structure of capsaicin is the vanilloid head, it was further hypothesized that UGT76G1 would be capable of glycosylation of the minimal vanilloid, i.e., vanillin, as well as many analogues.
  • UGT76G1 created multiple glycoside products of vanillin ( FIG. 14 , Table 10). Seeking to test the ability of UGT76G1 to glycosylate vanilloids more broadly, curcumin, the well characterized vanilloid found in turmeric spice, isolated from the ginger Curcuma longa was applied as a substrate in the glycosylation reaction. Consistent with the glycosylation of vanillin, UGT76G1 effectively glycosylated curcumin, creating multiple glycoside product peaks, suggesting a bifunctional recognition and glycosylation by UGT76G1 similar to that seen with CBD and steviol glycosides ( FIGS. 15A & 15B , Table 11).
  • Cannabinoid glycosides may also have direct bioactive and therapeutic effects, beyond their utility a prodrug for their aglycone form.
  • Quercetin is an antioxidant flavonoid that is ubiquitous in vegetables and often present both in its aglyone and glycosylated forms. It has been demonstrated through in vitro studies that quercetin glucuronides act as a bioactive agent as well as a precursor molecule to aglycone quercetin (Terao 2011). In many cases, including with glycosides that exert antibacterial and antitumor effects, the glycosidic residues are crucial to activity (Kren & Rezanka 2008).
  • Glycosides have also been demonstrated to receive facilitated transport across the blood brain barrier (BBB) by the glucose transporter GLUT1.
  • BBB blood brain barrier
  • a prime example is the glycoside of ibuprofen achieving a significant increase of ibuprofen aglycone concentration in the brain (Chen 2009). Similar to these glycosides, glycosides of cannabinoids and other compounds described herein may benefit from enhanced facilitated transport across the BBB or other barriers.
  • Glucose transporters are a wide group of membrane proteins encoded by the human genome and that are found not only in the BBB but across many different cells and tissues, including brain, erythrocytes, fat, muscle, kidney, liver, intestine, and pancreas, so glycosylation will be tailored to provide site-specific delivery to any of these tissues. Accordingly, in one embodiment, there is provided a method for facilitating the transport of a cannabinoid drug across the blood brain barrier of a subject comprising administering to the subject a cannabinoid glycoside prodrug in accordance with the present invention.
  • cannabinoids and cannabidiol may be especially useful because of oligodendrocyte protective (oligoprotective) and general neuroprotective effects. It has been demonstrated that cannabinoid signaling is involved with both oligodendrocyte differentiation (Gomez 2010) and that cannabinoids promote oligodendrocyte progenitor survival (Molina-Holgado 2002). Drug formulations that include cannabidiol as a major ingredient have been approved to treat muscle spasticity and pain from multiple sclerosis, a neurodegenerative disorder that causes loss of myelin and oligodendrocyte progenitor cells.
  • cannabidiol has been demonstrated to mediate oligoprotective effects through attenuation of endoplasmic reticulum stress pathways (Mecha 2012). Cannabidiol has also been studied extensively for its antipsychotic effects, however the exact role in protection of oligodendroctyes and promotion of remyelination has not yet been described (Zuardi 2012). Despite the correlation between the clinical symptoms of psychosis with neuropathological analysis that indicates dysmyelination is involved, the role of dysmyelination as a driver or cause of schizophrenia and other psychoses remains controversial (Mighdoll 2015). Remyelination has also been described as potentially useful for treatment of Alzheimer's disease and other forms of dementia (Bartzokis 2004).
  • cannabinoids delivery of cannabinoids to the brain may be especially useful for its established neuroprotective and oligoprotective effects.
  • Cannabinoid glycoside drug formulations co-administered in combination with other agents that influence other aspects of repair or regeneration, such as oligodendrocyte progenitor differentiation or remyelination, may also prove to be beneficial. This includes compounds such as anti-LINGO-1 monoclonal antibodies, guanabenz, sephin1, benzatropine, clemastine, polyunsaturated fatty acids, etc.
  • UGT76G1, Os03g0702000 and cyclodextrin glucanotransferase were capable of primary, secondary and tertiary glycosylations of steviol glycosides and aglycone products of diverse chemical structure, including cannabinoids, endocannabinoids, vanillin, curcumin, and capsaicin.
  • UGT76G1 is an enzyme from the plant Stevia rebaudiana , it will be compatible with expression in the genus Cannabis .
  • the ideal strategy for expression of UGT76G1 within the Cannabis plant is to genetically engineer the UGT76G1 open reading frame under a promoter element that is specific for the same tissue that cannabinoids are produced in, namely the secretory trichomes of the plant.
  • Suitable promoter elements include the promoter for the cytosolic O-acetylserine(thiol)lyase (OASA1) enzyme from Arabidopsis thaliana (Gutierrez-Alcala 2005).
  • Candidates for transformation with UGT76G1 include Cannabis sativa, Cannabis indica , and Cannabis ruderalis .
  • a similar approach may be used with UGT76G1 and similar enzymes for in planta production of glycosylated secondary metabolites within many other different plant species, and may be especially useful when plant species already produce large quantities of the desired aglycone product or known enzyme substrate.
  • an antimicrobial agent comprising an effective amount of a cannabinoid glycoside prodrug in accordance with the present invention.
  • a detersive agent comprising an effective amount of a cannabinoid glycoside prodrug in accordance with the present invention.
  • the present invention provides for nucleic acids comprising nucleotide sequences encoding a glycosyltransferase.
  • the glycosyltransferases of the present invention are capable of primary, secondary, tertiary glycosylations or a combination thereof.
  • the glycosyltransferases are capable of primary, secondary and tertiary glycosylations.
  • the glycosyltransferases are capable of secondary and tertiary glycosylations.
  • the nucleic acids encode a glucosyltransferase, including but not limited to a UDP-glucosyltransferase.
  • the glucosyltransferases include but are not limited to a Stevia rebaudiana UDP-glucosyltransferase, such as UGT76G1 or UGT74G1 or an Oryza sativa glucosyltrasferase, such as Os03g0702000.
  • the invention provides for nucleic acids comprising nucleotide sequences encoding a cyclodextrin glucanotransferase. Also provided are nucleic acids comprising nucleotide sequences that encode a sucrose synthase.
  • Nucleic acids include, but are not limited to, genomic DNA, cDNA, RNA, fragments and modified versions, including but not limited to codon optimized versions thereof.
  • the nucleotide sequences may be codon optimized for expression in Pichia pastoris or E. coli .
  • the nucleic acids may include the coding sequence of the glycosyltransferase or sucrose synthase, in isolation, in combination with additional coding sequences (e.g., including but not limited to a purification tag).
  • the nucleic acid comprises a sequence encoding UGT76G1 or UGT76G1-like glucosyltransferase.
  • UGT76G1-like glucosyltransferase include for example, other members of the UGT76G1 clade such as UGT76G2 or UGT76H1.
  • the nucleic acid comprises a sequence encoding an UGT76G1 glucosyltransferase having the amino acid sequence as set forth in any one of SEQ ID NOs:1, 3, 5 and 7 and listed below or fragments and variants thereof.
  • the nucleic acid comprises a sequence encoding UGT76G1 having the amino acid sequence as set forth in AAR06912.1.
  • the nucleic acid molecule comprises a sequence encoding UGT76G1 glucosyltransferase and comprising the nucleotide sequence as set forth in any one of SEQ ID NOs: 2, 4, 6 and 8 and listed below, or fragments and variants thereof.
  • the nucleic acid molecule encodes an UGT76G1 glucosyltransferase and comprises the nucleotide sequence as set forth in GenBank Accession number AY345974.1 or a variant or fragment thereof.
  • the nucleic acid comprises a sequence encoding UGT76G2 glucosyltransferase. In specific embodiments, the nucleic acid comprises a sequence encoding UGT76G2 glucosyltransferase having the amino acid sequence as set forth in SEQ ID NO:27 and listed below or variants and fragments thereof.
  • the nucleic acid comprises a sequence encoding UGT76G2 glucosyltransferase and having the nucleic acid sequence as set forth in SEQ ID NO:28 and listed below or variants and fragments thereof.
  • the nucleic acid comprises a sequence encoding UGT76H1 glucosyltransferase. In specific embodiments, the nucleic acid comprises a sequence encoding UGT76H1 glucosyltransferase having the amino acid sequence as set forth in SEQ ID NO:29 and listed below or variants and fragments thereof.
  • the nucleic acid comprises a sequence encoding UGT76H1 glucosyltransferase and having the nucleic acid sequence as set forth in SEQ ID NO:30 and listed below or variants and fragments thereof.
  • the nucleic acid comprises a sequence encoding Oryza sativa Os03g0702000 or Os03g0702000-like glucosyltransferase.
  • Os03g0702000-like glucosyltransferase include for example, other members of the UGT91clade such as UGT91D1 or UGT91 D2.
  • the nucleic acid comprises a sequence encoding Os03g0702000 glucosyltransferase having the amino acid sequence as set forth in SEQ ID NO: 9 and listed below or a variant or fragment thereof.
  • the nucleic acid molecule encodes Os03g0702000 glucosyltransferase and comprises the sequence as set forth in GenBank Accession number XM_015773655 or a variant or fragment thereof.
  • the nucleic acid comprises a sequence encoding UGT91 D1 glucosyltransferase. In certain embodiments, the nucleic acid comprises a sequence encoding UGT91 D1 glucosyltransferase having the amino acid sequence as set forth in SEQ ID NO:31 and listed below or a variant or fragment thereof.
  • the nucleic acid molecule encodes UGT91D1 glucosyltransferase and comprises a nucleotide sequence as set forth in SEQ ID NO: 32 and as detailed below or a variant or fragment thereof.
  • the nucleic acid comprises a sequence encoding UGT91 D2 glucosyltransferase. In certain embodiments, the nucleic acid comprises a sequence encoding UGT91 D2 glucosyltransferase having the amino acid sequence as set forth in SEQ ID NO: 33 and listed below or a variant or fragment thereof.
  • the nucleic acid molecule encodes UGT91D2 glucosyltransferase and comprises a nucleotide sequence as set forth in SEQ ID NO: 34 and as detailed below or a variant or fragment thereof.
  • the nucleic acid comprises a sequence encoding Stevia rebaudiana UDP-glycosyltransferase 74G1. In certain embodiments, the nucleic acid comprises a sequence encoding Stevia rebaudiana UDP-glycosyltransferase 74G1 which comprises the amino acid sequence as set forth in SEQ ID NO: 13 and as listed below or a variant or fragment thereof.
  • the nucleic acid molecule encodes Stevia rebaudiana UDP-glycosyltransferase 74G1 and comprises a nucleotide sequence as set forth in SEQ ID NO: 14 and as listed below or a variant or fragment thereof.
  • the nucleic acid molecule encodes Stevia rebaudiana UDP-glycosyltransferase 74G1 and comprises the sequence as set forth in GenBank Accession number AY345982 or a variant or fragment thereof.
  • the invention provides for nucleic acids comprising nucleotide sequences encoding a cyclodextrin glucanotransferase (WO1996033267; U.S. Pat. No. 6,271,010).
  • nucleic acids comprising nucleotide sequences that encode a sucrose synthase. Accordingly, in certain embodiments, the nucleic acid comprises a sequence encoding sucrose synthase which comprises the amino acid sequence as set forth in SEQ ID NO: 15, 17, 19, 21, 23 or 25 and listed below or a variant or fragment thereof.
  • SEQ ID NO: 15 Stevia rebaudiana SUS1 isoform
  • MAERVLTRVHSLRERLDSTLATHRNEILLFLSRIESHGKGILKPHQVMTEFEAICKEDQSKLSDG AFYEVLKCTQEAIVQPPWVALAIRLRPGVWEYVRVNVNVLVVEELSVPEYLHFKEELVNGTSN
  • the nucleic acid molecule encodes sucrose synthase and comprises a nucleotide sequence as set forth in SEQ ID NO: 16, 18, 20, 22, 24 or 26 and listed below or a fragment or variant thereof.
  • SEQ ID NO: 16 (encodes SUS1 isoform) ATGGCGGAACGTGTACTCACTCGTGTTCACAGTCTTCGTGAGCGTCTCGATTCAACTCTCG CAACTCATCGTAATGAAATCCTCTTGTTTCTTTCAAGGATTGAAAGCCATGGAAAAGGAATA TTGAAGCCTCATCAAGTTATGACTGAATTTGAAGCTATCTGCAAAGAAGATCAGAGCAAAC TCTCTGATGGTGCTTTTTATGAAGTTCTTAAATGCACACAGGAAGCAATAGTGCAACCTCC ATGGGTTGCACTCGCGATCCGTCTTCGACCCGGTGTTTGGGAATATGTTAGAGTCAATGTT AATGTTTTGGTGGTTGAAGAATTAAGTGTTCCTGAATATCTTCACTTCAAAGAAGAATTGGT TAATGGAACATCGAATGGCAACTTCGTGTTGGAACTGGATTTTGAACCTTACCGCATCG TTTCCTCGACCAACTTTAACCAAGTCTATTGGTAATGGTGTTGAGTTTCTAAACAGACAT
  • nucleic acid comprising a sequence having at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identity to any one of the sequences set forth in SEQ ID NOs: 2, 4, 6, 8, 10, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32 and 34 and fragments thereof or the complement thereof.
  • nucleic acid encoding a polypeptide comprising a sequence at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% percent identity to any one of the sequences set forth in SEQ ID NOs: 1, 3, 5, 7, 9, 13, 15, 17, 19, 21, 23, 25, 27, 29, 30, 31 and 33 and fragments thereof.
  • SEQ ID NOs: 1, 3, 5, 7, 9, 13, 15, 17, 19, 21, 23, 25, 27, 29, 30, 31 and 33 and fragments thereof A worker skilled in the art would readily appreciate that overall sequence identity or similarity may be less than 50% but regions of the enzyme (such as the catalytic site or areas adjacent to the catalytic site) may have conserved amino acids. For example, there are conserved amino acids at the opening adjacent to the UDPG catalytic site.
  • the nucleic acid encodes an UDP-glucosyltransferase having the sequence SDFGLDQ at a position corresponding to amino acid residues 375 to 381 of the UGT76G1 set forth in SEQ ID NO:1.
  • fragments are at least 10, at least 20, at least 50 nucleotides in length.
  • the fragments may be used, for example, as primers or probes.
  • nucleic acids that hybridize to the nucleic acids of the present invention or the complement thereof.
  • a nucleic acid that hybridizes to any one of the sequences set forth in SEQ ID NOs: 2, 4, 6, 8, 10, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32 and 34 or the complement thereof under conditions of low, moderate or high stringency.
  • hybridization and the strength of hybridization i.e., the strength of the association between the nucleic acids is impacted by such factors as the degree of complementary between the nucleic acids, stringency of the conditions involved, the T m of the formed hybrid, and the G:C ratio within the nucleic acids.
  • high stringency conditions when used in reference to nucleic acid hybridization comprise conditions equivalent to binding or hybridization at 42° C. in a solution consisting of 5 ⁇ SSPE (43.8 g/I NaCl, 6.9 g/I NaH 2 PO 4 H 2 O and 1.85 g/I EDTA, pH adjusted to 7.4 with NaOH), 0.5% SDS, 5 ⁇ Denhardt's reagent and 100 ⁇ g/ml denatured salmon sperm DNA followed by washing in a solution comprising 0.1 ⁇ SSPE, 1.0% SDS at 42° C. when a probe of about 500 nucleotides in length is employed.
  • 5 ⁇ SSPE 43.8 g/I NaCl, 6.9 g/I NaH 2 PO 4 H 2 O and 1.85 g/I EDTA, pH adjusted to 7.4 with NaOH
  • SDS 5 ⁇ Denhardt's reagent
  • 100 ⁇ g/ml denatured salmon sperm DNA followed by washing in a solution comprising 0.1 ⁇ SSPE, 1.0%
  • a non-limiting example of “medium stringency conditions” when used in reference to nucleic acid hybridization comprise conditions equivalent to binding or hybridization at 42° C. in a solution consisting of 5 ⁇ SSPE (43.8 g/I NaCl, 6.9 g/I NaH 2 PO 4 H 2 O and 1.85 g/I EDTA, pH adjusted to 7.4 with NaOH), 0.5% SDS, 5 ⁇ Denhardt's reagent and 100 ⁇ g/ml denatured salmon sperm DNA followed by washing in a solution comprising 1.0 ⁇ SSPE, 1.0% SDS at 42° C. when a probe of about 500 nucleotides in length is employed.
  • Low stringency conditions when used in reference to nucleic acid hybridization comprise conditions equivalent to binding or hybridization at 42.degree. C. in a solution consisting of 5 ⁇ SSPE (43.8 g/I NaCl, 6.9 g/I NaH 2 PO 4 H 2 O and 1.85 g/I EDTA, pH adjusted to 7.4 with NaOH), 0.5% SDS, 5 ⁇ Denhardt's reagent and 100 ⁇ g/ml denatured salmon sperm DNA followed by washing in a solution comprising 5 ⁇ SSPE, 0.1% SDS at 42° C. when a probe of about 500 nucleotides in length is employed.
  • 5 ⁇ SSPE 43.8 g/I NaCl, 6.9 g/I NaH 2 PO 4 H 2 O and 1.85 g/I EDTA, pH adjusted to 7.4 with NaOH
  • SDS 5 ⁇ Denhardt's reagent
  • 100 ⁇ g/ml denatured salmon sperm DNA followed by washing in a solution comprising 5 ⁇ SSPE, 0.1%
  • the polynucleotides include the coding sequence polypeptide, in isolation, in combination with additional coding sequences (e.g., a purification tag, a localization signal, as a fusion-protein, as a pre-protein, or the like), in combination with non-coding sequences (e.g., introns or inteins, regulatory elements such as promoters (including inducible promoters, tissue-specific promoters (such as root-specific or leaf specific promoters), enhancers, terminators, and the like), and/or in a vector or host environment in which the polynucleotide encoding a transcription factor or transcription factor homologue polypeptide is an endogenous or exogenous gene.
  • additional coding sequences e.g., a purification tag, a localization signal, as a fusion-protein, as a pre-protein, or the like
  • non-coding sequences e.g., introns or inteins, regulatory elements such as promoters (including
  • coding sequences e.g., a purification tag, a localization signal, as a fusion-protein, as a pre-protein, or the like
  • non-coding sequences e.g. regulatory elements such as promoters (including inducible promoters, tissue-specific promoters (such as root-specific or leaf specific promoters), enhancers, terminators, and the like)
  • vectors for use in prokaryotic such as E. coli and eukaryotic cells, including but not limited to yeast and plant cells are known in the art.
  • the present invention provides for glycosyltransferases.
  • the glycosyltransferases of the present invention are capable of primary, secondary and/or tertiary glycosylations.
  • the glycosyltransferases are capable of primary, secondary and tertiary glycosylations.
  • the glycosyltransferases are capable of secondary and/or tertiary glycosylations.
  • the glycosyltransferases is a glucosyltransferase, including but not limited to a UDP-glycotransferase.
  • the glucosyltransferases include but are not limited to a Stevia rebaudiana UDP-glucosyltransferase, such as UGT76G1 or UGT74G1 or an Oryza sativa glucosyltrasferase, such as Os03g0702000.
  • the invention provides for a cyclodextrin glucanotransferase.
  • sucrose synthases are also provided.
  • an UGT76G1 or UGT76G1-like glucosyltransferase include for example, other members of the UGT76G1 clade such as UGT76G2 or UGT76H1. Accordingly, in certain embodiments, there is provided an UGT76G1 comprising the amino acid sequence as set forth in any one of SEQ ID NOs: 1, 3, 5 and 7 or fragments and variants thereof. In certain embodiments, there is provided an UGT76G1 encoded by the nucleic acid molecule comprising the sequence as set forth in any one of SEQ ID NOs: 2, 4, 6 and 8.
  • an UGT76G2 comprising the amino acid sequence as set forth in SEQ ID NO: 27 or fragments and variants thereof.
  • an UGT76G1 encoded by the nucleic acid molecule comprising the sequence as set forth in SEQ ID NO: 28.
  • an UGT76H1 comprising the amino acid sequence as set forth in SEQ ID NO: 29 or fragments and variants thereof.
  • an UGT76G1 encoded by the nucleic acid molecule comprising the sequence as set forth in SEQ ID NO: 30.
  • Os03g0702000 or Os03g0702000-like glucosyltransferase include for example, other members of the UGT91clade such as UGT91D1 or UGT91D2. Accordingly, in certain embodiments, there is provided an Os03g0702000 comprising an amino acid sequence as set forth in SEQ ID NO: 9 or fragments and variants thereof. In certain embodiments, there is provided an Os03g0702000 encoded by the nucleic acid molecule comprising the sequence as set forth in SEQ ID NO: 10.
  • an UGT91 D1 comprising the amino acid sequence as set forth in SEQ ID NO: 31 or fragments and variants thereof. In certain embodiments, there is provided an UGT91 D1 encoded by the nucleic acid molecule comprising the sequence as set forth in SEQ ID NO: 32.
  • an UGT91 D2 comprising the amino acid sequence as set forth in SEQ ID NO: 33 or fragments and variants thereof.
  • an UGT76G1 encoded by the nucleic acid molecule comprising the sequence as set forth in SEQ ID NO: 34.
  • the UGT74G1 comprises the amino acid sequence as set forth in SEQ ID NO: 13 or fragments and variants thereof.
  • the UGT74G1 is encoded by the nucleic acid molecule comprising the sequence as set forth in SEQ ID NO: 14.
  • the invention provides for a cyclodextrin glucanotransferase.
  • Cyclodextrin-glucanotransferase is commercially available (CGTase, Toruzyme 3.0L, trademark of Novozymes Inc.).
  • sucrose synthase comprises the amino acid sequence as set forth in SEQ ID NO: 15, 17, 19, 21, 23 or 25 or fragments and variants thereof.
  • the polypeptide comprises an amino acid sequence encoded by the nucleic acid molecule comprises comprising the sequence as set forth in SEQ ID NO: 16, 18, 20, 22, 24 or 26.
  • a polypeptide comprising a sequence at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% percent identity to any one of the sequences set forth in SEQ ID NOs: 1, 3, 5, 7, 9, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31 and 33 and fragments thereof.
  • the nucleic acid encodes an UDP-glucosyltransferase having the sequence SDFGLDQ at a position
  • fragments are at least 10, at least 20, at least 50 amino acids in length.
  • the polypeptide sequences contain heterologous sequences including but not limited to purification tags such as a HIS tag.
  • a polypeptide comprising a 6 ⁇ HIS tag at the N-terminus there is provided a polypeptide comprising a 6 ⁇ HIS tag at the C-terminus.
  • glycosyltransferases including glucosyltransferases and cyclodextrin glucanotransferases are known in the art. As such, a worker skilled in the art could readily determine if the glycosyltransferases are capable of primary, secondary and/or tertiary glycosylations (see, for example Dewitte et al., J Biotechnol. 2016 Sep. 10; 233:49-55. doi: 10.1016/j.jbiotec.2016.06.034; Grubb et al., Plant J . (2014) 79, 92-105; Richman et al., Plant J .
  • the present invention further provides cells and plants which express one or more of the polypeptides of the present invention.
  • the cells and plants may naturally express one or more of the polypeptides of the present invention or have been modified to express one or more the polypeptides of the present invention.
  • the cells may be prokaryotic or eukaryotic cells and include but are not limited to, E. coli , yeast such as Pichia pastoris, Stevia rebaudiana, Phytolacca Americana, Cannabis including but not limited to Cannabis sativa, Cannabis indica and Cannabis ruderalis.
  • a cell which expresses an UGT76G1 or UGT76G1-like glucosyltransferase (such as UGT76G2 and UGT76H1). Accordingly, in certain embodiments, there is provided a cell which expresses an UGT76G1 glucosyltransferase comprising a sequence encoding the amino acid sequence as set forth in any one of SEQ ID NOs: 1, 3, 5 and 7. In certain embodiments, there is provided a cell which expresses an UGT76G1-like glucosyltransferase comprising a sequence encoding the amino acid sequence as set forth in SEQ ID NO: 27 or 29.
  • the cell may further express further glucosyltransferases, such as Os03g0702000 or Os03g0702000-like glucosyltransferase (such as UGT91 D1 and UGT91 D2) and/or a sucrose synthase, such as the sucrose synthase comprising the sequence as set forth in SEQ ID NO: 15, 17, 19, 21, 23 or 25.
  • glucosyltransferases such as Os03g0702000 or Os03g0702000-like glucosyltransferase (such as UGT91 D1 and UGT91 D2) and/or a sucrose synthase, such as the sucrose synthase comprising the sequence as set forth in SEQ ID NO: 15, 17, 19, 21, 23 or 25.
  • a cell which expresses UGT76G1 glucosyltransferase comprising a sequence encoding the amino acid sequence as set forth in any one of SEQ ID NOs: 1, 3, 5 and 7 and Os03g0702000 glucosyltransferase comprising the sequence as set forth in SEQ ID NO:10.
  • the cell may further express a sucrose synthase comprising the sequence as set forth in SEQ ID NO: 15, 17, 19, 21, 23 or 25.
  • a cell which expresses an Os03g0702000 or Os03g0702000-like glucosyltransferase expresses an Os03g0702000 or Os03g0702000-like glucosyltransferase. Accordingly, in certain embodiments, there is provided a cell which expresses Os03g0702000 glucosyltransferase comprising a sequence encoding the amino acid sequence as set forth in SEQ ID NO: 10. The cell may further express a sucrose synthase, such as the sucrose synthase comprising the sequence as set forth in SEQ ID NO: 15, 17, 19, 21, 23 or 25.
  • Transgenic cells and plants can be produced by a variety of well established techniques. Following construction of a vector, most typically an expression cassette, including a polynucleotide of the invention, standard techniques can be used to introduce the polynucleotide into cell or a plant. Optionally, the plant cell, explant or tissue can be regenerated to produce a transgenic plant.
  • Cannabis plants genetically engineered to express one or more of the proteins of the invention are provided.
  • a tissue specific promoter such as a secretory trichomes specific promoter may be used such that the proteins of the invention are expressed in the same tissue that cannabinoids are produced in, namely the secretory trichomes of the plant.
  • Suitable promoter elements include the promoter for the cytosolic O-acetylserine(thiol)lyase (OASA1) enzyme from Arabidopsis thaliana (Gutierrez-Alcala 2005).
  • Transformation and regeneration of plant cells is now routine, and the selection of the most appropriate transformation technique will be determined by the practitioner.
  • Suitable methods can include, but are not limited to: electroporation of plant protoplasts; liposome-mediated transformation; polyethylene glycol (PEG) mediated transformation; transformation using viruses; micro-injection of plant cells; micro-projectile bombardment of plant cells; vacuum infiltration; and Agrobacterium tumeficiens mediated transformation.
  • Transformation means introducing a nucleotide sequence into a plant in a manner to cause stable or transient expression of the sequence.
  • plants may be selected using a dominant selectable marker incorporated into the transformation vector.
  • a dominant selectable marker incorporated into the transformation vector.
  • such a marker will confer antibiotic or herbicide resistance on the transformed plants, and selection of transformants can be accomplished by exposing the plants to appropriate concentrations of the antibiotic or herbicide.
  • the present invention further provides methods for the production of cannabinoid glycoside prodrugs and the cannabinoid glycosides prodrugs produced by the methods.
  • the methods may be in vitro or in vivo (in a cell system or in planta).
  • a method of producing cannabinoid glycoside prodrugs comprising incubating a cannabinoid aglycone with one or more sugar donors in the presence of one or more glycosyltransferases.
  • the aglycones include but are not limited to: cannabinoids, including but not limited to cannabidiol, cannabidivarin, cannabigerol, tetrahydrocannabinol, cannabinol and cannabidiolic acid, endocannabinoids including but not limited to arachidonoylethanolamide (anandamide, AEA), 2-arachidonoylethanolamide (2-AG), 1-arachidonoylethanolamide (1-AG), and docosahexaenoyl ethanolamide (DHEA, synaptamide); and vanilloids including but not limited to vanillin, curcumin, and capsaicin.
  • cannabinoids including but not limited to cannabidiol, cannabidivarin, cannabigerol, tetrahydrocannabinol, cannabinol and cannabidiolic acid
  • endocannabinoids including but not limited to arachidonoy
  • the one or more sugar donors will be dependent on the one or more glycosyltransferases used in the method and/or the desired end products.
  • the sugar donors include but are not limited to UDP-glucose, UDP-glucuronic acid, UDP-mannose, UDP-fructose, UDP-xylose, UDP-fluorodeoxyglucose, and UDP-rhamnose.
  • the sugar donor includes maltodextrin.
  • a method of producing a cannabinoid glycoside comprising incubating an aglycone with a sugar donor in the presence of a glycosyltransferase. Also provided are the cannabinoid glycosides produced by the above method. In specific embodiments, there is provided a method of producing a cannabinoid glycoside, said method comprising incubating an aglycone with UDP-glucose, in the presence of a UGT76G1 or UGT76G1-like glucosyltransferase under conditions that allow for glycosylation.
  • a method of producing a glycoside prodrug comprising incubating an aglycone with maltodextrin, in the presence of a cyclodextrin glucanotransferase under conditions that allow for glycosylation.
  • An exemplary method for producing cannabinoid-glycosides comprises incubating a cannabinoid, with UDP-glucose in the presence of a UGT76G1 or UGT76G1-like glucosyltransferase under conditions which allow for glycosylation. Also provided are cannabinoid-glycosides produced by the above method.
  • a further exemplary method for producing cannabinoid-glycosides comprises incubating a cannabinoid with maltodextrin in the presence of a cyclodextrin glucanotransferase under conditions which allow for glycosylation. Also provided are cannabinoid-glycosides produced by the above method.
  • a method of producing a cannabinoid glycoside comprising incubating an aglycone with one or more sugar donors in the presence of a first glycosyltransferase and a second glycosyltransferase under conditions which allow for glycosylation. Also provided are cannabinoid glycosides produced by the above method.
  • first glycosyltransferase and a second glycosyltransferase may be provided concurrently or added sequentially.
  • sugar donors may be provided concurrently or added sequentially.
  • the structure of the resulting cannabinoid glycoside may be dependent on the order the glycosyltransferases are provided.
  • the ratio of first to second glycosyltransferase may impact the resulting products.
  • the activity levels of the glycosyltransferases may dictate the ratios and the ratios could be readily determined by a worker skilled in the art.
  • the ratios first to second glycosyltransferase include but are not limited to 1:1, 1:2, 1:10, 1:50 and vice versa.
  • a method of producing a cannabinoid glycoside comprising incubating an aglycone with UDP-glucose in the presence of a UGT76G1 or UGT76G1-like glucosyltransferase and Os03g0702000 or Os03g0702000-like glucosyltransferase under conditions which allow for glycosylation.
  • a method of producing a cannabinoid glycoside comprising incubating an aglycone with UDP-glucose and maltodextrin in the presence of a UGT76G1 or UGT76G1-like glucosyltransferase and cyclodextrin glucanotransferase under conditions which allow for glycosylation.
  • cannabinoid glycosides produced by the above methods.
  • An exemplary method for producing cannabinoid-glycosides comprises incubating cannabinoid, including but not limited to cannabidiol, cannabidivarin, cannabigerol, tetrahydrocannabinol, cannabinol and cannabidiolic acid, with UDP-glucose in the presence of a UGT76G1 or UGT76G1-like glucosyltransferase and Os03g0702000 or Os03g0702000-like glucosyltransferase under conditions which allow for glycosylation. Also provided are cannabinoid-glycosides produced by the above method.
  • a further exemplary method for producing cannabinoid-glycosides comprises incubating cannabinoids with UDP-glucose and maltodextrin in the presence of a UGT76G1 or UGT76G1-like glucosyltransferase and and cyclodextrin glucanotransferase under conditions which allow for glycosylation. Also provided are cannabinoid-glycosides produced by the above method.
  • each of the above described glycosylation methods may be applied to a lower order cannabinoid glycoside to form a higher order cannabinoid glycoside.
  • a cannabinoid monoglycoside may be glycosylated using any of the glycosylation methods of the present invention to form a diglycoside, or a cannabinoid diglycoside may be glycosylated to form a triglycoside, etc.
  • Methods of purifying the cannabinoid glycosides include for example solid phase extraction, such as column purification.
  • the invention also provides cell culture and in planta methods for the production of cannabinoid glycosides.
  • the methods comprise expressing one or more of the glycosyltransferases in a cell or plant which produces the aglycone and isolating the cannabinoid glycosides.
  • one or more sucrose synthases are also expressed.
  • Appropriate vectors and genetic engineering methods are known in the art.
  • the invention also provides methods for the conversion of UDP to UDPG utilizing the sucrose synthases of the present invention. Accordingly, in certain embodiments of the methods of producing cannabinoid glycosides which utilize UDP-glucose as a sugar donor, the methods further comprise the use of sucrose synthase to recycle UDP. In certain embodiments, there is provided a method of producing a cannabinoid glycoside, said method comprising incubating aglycone with UDP-glucose, in the presence of a UGT76G1 glucosyltransferase and a sucrose synthase under conditions that allow for glycosylation.
  • Glycosylation reactions consisted of 50 mM KPO 4 pH 7.2, 3 mM MgCl 2 , 0.005% CBD, 2.5% UGT76G1 purified enzyme preparation, and 2.5 mM UDP-glucose. Buffers were degassed and tubes were purged with nitrogen, reactions were protected from light and incubated at 28° C. with 180 rpm agitation for 18 hours. Reactions were then extracted 3 ⁇ with an equal volume of ethyl acetate, evaporated to dryness, and dissolved in a half volume of HPLC grade methanol. 50 microliters was injected on a reverse phase C18 column and eluted with a gradient of acetonitrile starting at 10% and increasing to 99%.
  • UGT76G1 was produced through expression in Pichia pastoris and purified through standard molecular biology techniques.
  • the UGT76G1 enzyme was found to glycosylate CBD in a UDP-glucose dependent manner. This activity was also proportional to the amount of UDP-glucose present. Incubation temperature was 28° C., and an acceptable range would be 20° C. to 30° C. as high temperatures can cause significant degradation of CBD. Reactions were carried out in the dark to prevent photo-degradation of the substrates. Gentle agitation from 120 to 200 rpm were used to mix the reactions in an inert atmosphere.
  • Substrate CBD in the reactions was replaced with ⁇ 9THC and CBDV and performed in an identical fashion with similar results. Enzyme combinations needed to create various products are listed in Table 4 for CBD-glycosides, Table 5 for CBDV-glycosides, and Table 6 for ⁇ 9THC-glycosides.
  • Enzymatic reactions are performed as described in Example 1 but with the inclusion of recombinant Os03g0702000 enzyme at a 1:2 ratio relative to UGT76G1. Samples were extracted and analyzed as in Example 1. Recombinant Os03g0702000 enzyme was codon optimized and expressed in E. coli BL21-DE3 cells and purified by immobilized metal ion chromatography.
  • Recombinant cyclodextrin glucanotransferase (CGTase, Toruzyme 3.0L trade name, Novozymes Inc.) was added to reactions as indicated in Example 1 but without UDPG or UGT76G1. Maltodextrin was used at 0.05% final concentration, and Toruzyme 3.0L was used at 0.1%. Samples were extracted and analyzed as in example 1. Additionally, reactions from Example 1 were carried out to convert cannabinoids to cannabinoid-glycosides, and then CGTase and maltodextrin were added and given adequate time to incubate with the cannabinoid-glycosides.
  • the resulting products contain a ⁇ -glycosylation on the cannabinoid backbone, and ⁇ -glycosylations emanating from the primary sugar.
  • This additional treatment created a new category of compounds termed ⁇ -primed, ⁇ -glycosylated cannabinoids.
  • Glycoside products were generated through the aforementioned biocatalytic reactions and purified to homogeneity by C18 solid phase extraction.
  • 100 mg Hypersep C18 columns (Thermo) were hydrated in methanol, rinsed with 50% methanol in water, rinsed with water, glycosylation reaction passed through the column, washed with water, washed with 10%, 20%, and 30% methanol, and the glycoside products were eluted with 45 and 60% methanol in water. Eluates were dried and extracted with ethyl acetate, and dried to completion to yield >95% pure cannabinoid-glycosides for further analysis and testing.
  • the HPLC line traces of the reaction products of glycosylation reactions of the cannabinoid aglycones CBD, CBDV, ⁇ 9-THC, CBN, 1-AG and 2-AG, DHEA, AEA, capsaicin, and vanillin, are provided in FIGS. 16 to 24 , respectively. Enzymatic reactions were performed as described in Example 1. The solid lines indicate the elution profile of the starting aglycone and the dashed lines indicate the elution profile of the glycosylation reaction product mixture.
  • the CBD aglycone retention time is 13.65 minutes, and product peaks are observed at 8.87, 9.02, 9.97, 10.33, and 10.37 min.
  • the CBDV aglycone retention time is 12.75 minutes, and product peaks are observed at 8.53, 9.70, and 10.01 min.
  • the THC aglycone retention time is 14.45 minutes, and product peaks are observed at 9.46, 10.67, 10.97, 11.28, 11.67, and 12.49 min.
  • the CBN aglycone retention time is 14.32 minutes, and product peaks are observed at 10.87, 11.50, and 12.25 min.
  • the 1-AG aglycone retention time is 14.18 minutes and the 2-AG aglycone retention time is 14.32 minutes, and product peaks are observed at 11.40, 11.78, 11.83, 11.97, 12.53, 12.92, 13.07, and 13.35 min.
  • the DHEA aglycone retention time is 13.78 minutes, and product peaks are observed at 10.09 and 12.43 min.
  • the AEA aglycone retention time is 13.87 minutes, and product peaks are observed at 12.47 min.
  • the vanillin aglycone retention time is 1.95 minutes and product peaks are observed from 1.25 to 1.35 min.
  • the capsaicin aglycone retention time is 11.73 minutes, and product peaks are observed at 10.23 min.
  • the LC column used was a Silia Chrom XDB C18 Sum, 150A, 4.6 ⁇ 50 mm.
  • the method was 12 min 5 to 95 H 2 O:ACN gradient.
  • electrospray ionization (ESI) was performed in positive mode.
  • FIG. 27 depicts the 1 NMR spectra of isolated VB104 and FIG. 28 depicts the 1 H MR spectra of isolated VB110.
  • Each of these products was isolated from the reaction mixture produced by the glycosylation reaction of CBD.
  • the 1 H NMR spectra of 10 mg/ml solutions of each compound prepared in CD30D were obtained on a Bruker Avance II 400 MHz instrument using TopSpin acquisition and processing software.
  • C18 retention times were empirically determined on a linear ramp of increasing acetonitrile on a Phenomenex Kinetex 2.6u 100A C18 column, on a Dionex HPLC equipped with Diode Array Detector.
  • C Log P values in Table A were predicted by ChemDraw (CambridgeSoft). Reference cannabinoids were analyzed by HPLC and established log P values (http://pubchem.ncbi.nlm.nih.gov/) and used to create a calibration line as depicted in FIG. 29 . The predicted c Log P values correlated with the reference calibration line.
  • C18 reverse phase HPLC retention times were plotted against the c Log P values presented in Table A, as depicted in FIG. 29 .
  • VB110 was administered to three mice by oral gavage and the animals sacrificed at 30, 60, and 90 minutes. Eight week old male Swiss mice were fasted for 12 hours prior to administration of 120 mg/kg VB110 in 10% Ethanol USP, 10% Propylene Glycol USP, 0.05% Sodium Deoxycholate USP, 79.95% Saline USP. Following termination and tissue harvest, the intestinal contents were then extracted and analyzed by C18 reverse phase HPLC. As shown in FIG. 30A , the small intestinal contents showed intact VB110, but no decoupled CBD. As shown in FIG. 30B , the large intestinal contents contained both VB110 and CBD in the 60 and 90 minute time points. This decoupling of VB110 is consistent with the large intestinal decoupling seen for sennoside beta-glycosides, and is the result of secreted beta-glycosidases from the large intestinal microflora.
  • Example 10 Analysis of Large Intestine Contents Upon Administration of CBD and CBD Glycosides
  • an aqueous solution of a mixture of CBD-glycosides was administered to a mouse by oral gavage.
  • a solution of CBD in cremophor, ethanol, and saline was administered to a second mouse.
  • the animals were each sacrificed at 2 hours. Following termination and tissue harvest, the intestinal contents were then extracted and analyzed by C18 reverse phase HPLC.
  • the mice employed in this example were eight week old male Swiss mice fasted for 12 hours prior to administration of the solutions.
  • the resulting extracts were analyzed by LCMS performed using a Shimadzu LC-MS 2010 EV.
  • LC separation was carried out using a Silia Chrom XDB C18 5 um, 150A, 4.6 ⁇ 50 mm. The method was 12 min, 5 to 95 H 2 O:ACN gradient elution.
  • Low resolution MS was performed in negative mode via electrospray ionization (ESI). Acetic acid and formic acid were used as sample additives during analysis, and the injection volume was 20 ⁇ l.
  • the plasma and brains from the same animals were also extracted and analyzed by HPLC for the presence of CBD-glycosides and CBD.
  • CBD was only present in the control animal that received CBD aglycone (data not shown).
  • the contents of the small intestines from the same animals were also extracted and analyzed by HPLC for the presence of CBD-glycosides and CBD, but no CBD aglycone was present in the small intestines (data not shown, consistent with THC decoupling data shown in example 11).
  • the presence of the CBD aglycone in the large intestinal contents indicates the successful delivery of CBD-glycosides, and the subsequent hydrolysis of the glycosides by beta-glycosidase enzymes only present in the large intestine.
  • CBD detoxification metabolite CBD-2OH is also consistent with delivery of CBD and absorption into the intestinal epithelium where CBD begins to be metabolized.
  • This example illustrates the potential to administer CBD-glycosides, safely transit the CBD-glycosides through the small intestine without absorption, transit to the large intestine where the sugars can be decoupled to release CBD locally, avoiding systemic absorption and delivery of the CBD to other tissues where it can have unwanted effects.
  • mice In order to investigate the metabolism and decoupling of THC-glycosides in the large intestine, an aqueous solution of a mixture of THC-glycosides was administered to two mice by oral gavage. The first animal was sacrificed at 2 hours and the second animal was sacrificed at 4 hours. Following termination and tissue harvest, the intestinal contents were then extracted and analyzed by C18 reverse phase HPLC. The mice employed in this example were eight week old male Swiss mice fasted for 12 hours prior to administration of the solutions.
  • the plasma and brains from the same animals were also extracted and analyzed by HPLC for the presence of THC-glycosides and THC, but neither compound was seen in these tissues (data not shown).
  • the contents of the small intestines from the same animals were also extracted and analyzed by HPLC for the presence of THC-glycosides and THC, but no THC aglycone was observed (data not shown, consistent with CBD decoupling data shown in Example 10).
  • the presence of the THC aglycone in the large intestinal contents at 2 and 4 hours indicates the successful delivery of THC-glycosides, and their subsequent hydrolysis of the glycosides by beta-glycosidases in the large intestine.
  • THC-glycosides The presence of decoupled THC in the large intestine, but not in the small intestine, indicates that glycoside decoupling only occurs upon transit to the large intestine.
  • THC detoxification metabolites in the large intestine is further proof that the THC aglycone is present and being absorbed by the intestinal epithelium where it begins to be metabolized.
  • This example illustrates the potential to administer THC-glycosides orally, transit the THC-glycosides through the small intestine without absorption, transit to the large intestine where the sugars can be decoupled to release THC locally, avoiding systemic absorption and delivery of the THC to the central nervous system where it can have unwanted psychoactivity.
  • sucrose synthase isoforms were identified within the stevia transcriptome, all having similar homology to the 6 isoforms found in Arabidopsis thaliana and named in conjunction with their homologues. These transcripts were cloned as described in materials and methods with the corresponding sequence ID information listed herein.
  • Enzymatic activities were tested and assayed for their ability to enhance UGT reactions with decreased UDPG input.
  • the best isoform, SrSUS4 was capable of recycling UDP to UDPG with sucrose, in concert with the steviol 19-O-glucosyltransferase SrUGT74G1 mediated glycosylation of steviol bioside to stevioside.
  • Targeted mutagenesis was performed to mutate a serine residue at the N-terminus that is commonly phosphorylated in planta to prevent dimerization (Hardin 2004).
  • SrSUS1-S13D mutants were created by mutating serine at position 13 to an aspartic acid residue (S13D), thus forming a phospho-mimetic protein. Additionally, the creation of SrSus1-S13R,L14I was created to replace the serine with an arginine, a large charged residue, also to prevent dimerization and inactivation of the enzyme. Sucrose synthase mutants showed improved UDPG production activity compared to their native counterparts.
  • SrSUS5 SEQ ID NOs.
  • Relative activity for UDPG production with SUS isoforms is as follows:
  • cannabinoid glycosides via UGT enzyme requires the nucleotide sugar donor UDPG in stoichiometric amounts, it is advantageous to recycle or recapture the spent UDP following a glycosylation reaction.
  • SUS4 isoform from Stevia rebaudiana cannabinoid glycosides were successfully produced using only UMP as the input nucleotide.
  • a two step reaction took place, first to produce UDP from UMP, and second to produce UDPG from the UDP in tandem with the UGT reaction.
  • a 5L reaction containing 50 mM KPO4 pH7.2, 200 mM UMP disodium salt, 200 mM ATP disodium salt, 1M MgCl2, 10% UMPK recombinant enzyme in 50% glycerol was prepared. The reaction was incubated at 28 C with stirring for >24 hours.
  • the 5L reaction 1 was filtered at 0.45 microns to remove precipitate then applied to a 50 L reaction containing 50 mM KPO4 pH7.2, 50 mM MgCl2, 300 mM Sucrose, 200 mg of CBD in 200 ml DMSO, 5L UGT76G1 in 50% glycerol, 2.5L SrSUS4 in 50% glycerol.
  • the main 50 L reaction was then mixed and allowed to react.
  • An additional 200 mg of CBD in 200 ml DMSO was added after the reaction went to completion, and allowed to continue incubating at the same conditions.
  • the mixture was filtered by tangential flow filtration with a ultrafiltration membrane at 5 kDa to remove enzymes and particulate, and then concentrated using nanofiltration membrane at 500 Da.
  • the nanofiltration retentate containing the cannabosides was then applied to hydrated C18 flash columns, washed with 10-30% methanol, and eluted with 40-65% methanol.
  • the eluate was then concentrated by rotary evaporation to remove all solvent, shell-frozen in a vacuum beaker and lyophilized to dryness.
  • the powdered cannabosides produced were then collected and stored at ⁇ 20 C in sealed vials.
  • Sucrose should be sterile filtered to avoid carmelization or sugar breakdown, as autoclaving sucrose stock solutions greatly decreases reaction activity.
  • Vanillin glycoside compositions by R-group R-group location is as depicted in FIG. 1B VB# 1° 1-O Position 2° 2-O- 2° 3-O- 3° 3-O- Name 1° Ezyme 2° Enzyme VB1001
  • UGT76G1 VB1004 R1 ⁇ -D-glucose ⁇ -D-glucose Vanillin-1-O-(2-1)-diglcopyranoside

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Organic Chemistry (AREA)
  • Molecular Biology (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biochemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Biotechnology (AREA)
  • Genetics & Genomics (AREA)
  • Neurology (AREA)
  • Biomedical Technology (AREA)
  • Crystallography & Structural Chemistry (AREA)
  • Neurosurgery (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Saccharide Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)

Abstract

The present invention relates to cannabinoid glycoside prodrugs suitable for site- and tissue-specific delivery of cannabinoid molecules. The present invention also relates to methods of forming the cannabinoid glycoside prodrugs through glycosyltransferase mediated glycosylation of cannabinoid molecules.

Description

    FIELD OF THE INVENTION
  • The present invention pertains to the field of drug development and in particular to novel cannabinoid glycoside prodrugs and methods for their production by enzyme-mediated carbohydrate transfer.
  • BACKGROUND
  • Phytocannabinoids from Cannabis sativa have long been used for altering mental states, but recent findings have illuminated the potential of specific cannabinoid compounds for treatment and maintenance of various diseases and conditions. Of particular importance is the non-psychotropic molecule cannabidiol (CBD) which has potential therapeutic application as an anti-psychotic, a neuroprotectant, and has potential for treatment of numerous other maladies (Zuardi 2012, Iuvone 2009, for review Mechoulam 2002, respectively). One shortcoming of CBD is that it is easily oxidized to THC and CBN derivatives by light, heat, and acidic or basic conditions, and another detrimental attribute to CBD is that its extremely hydrophobic nature makes it difficult for formulation and delivery. Additionally, current pharmaceutical compositions of CBD and THC have unpleasant organoleptic properties, and their hydrophobic nature results in a lingering on the palate.
  • Cannabinoids are extremely hydrophobic in nature, complicating their use in drug formulations. Non-covalent methods have been found to improve the solubility of cannabinoids by utilizing carrier carbohydrates such as cyclized maltodextrins (Jarho 1998). Covalent chemical manipulations have produced novel CBD prodrugs with improved solubility (WO2009018389, WO 2012011112). Even fluorine substituted CBD compounds have been created through synthetic chemical manipulations in an effort to functionalize CBD (WO2014108899). The aforementioned strategies were somewhat successful in improving the solubility of CBD, but they create unnatural compositions which alter the composition and will release the unnatural prodrug moieties upon hydrolysis.
  • A growing body of evidence shows that glycosides are capable of acting as prodrugs and also to have direct therapeutic effects. Glycoside prodrugs may enable improved drug bioavailability or improved drug pharmacokinetics including more site-specific or tissue-specific drug delivery, more consistent levels of drug in the plasma, and sustained or delayed release of the drug. Site-specific delivery of steroid glycosides to the colon has previously been demonstrated (Friend 1985, Friend 1984), and could enable treatment of local disorders such as inflammatory bowel disease. Glycosylation of steroids enabled survival of stable bioactive molecules in the acidic stomach environment and delivery into the large intestine, where the aglycones were liberated by glycosidases produced by colonic bacteria, and then absorbed into the systemic circulation. Glycosidases are also present universally in different tissues (Conchie 1959), so delivery of glycosides by methods that bypass the digestive tract and colon, such as intravenous delivery, will enable targeted delivery to other cells and tissues that have increased expression of glycosidases. In addition, the distribution of alpha-glycosidase and beta-glycosidase enzymes differ throughout the intestinal tract and other tissues, and different forms of glycosides may therefore provide unique pharmacokinetic profiles, including formulations that target delivery of specific diseased areas, or targeted release at locations that can promote or restrict systemic absorption of the cannabinoids and other compounds described herein. Many biologically active compounds are glycosides, including members of classes of compounds such as hormones, antibiotics, sweeteners, alkaloids, and flavonoids. While it is generally accepted that glycosides will be more water-soluble than the aglycones, literature reviews have analyzed structure-activity relationships and determined that it is nearly impossible to define a general pattern for the biological activities of glycosides across different classes of compounds (Kren 2008).
  • As with synthetic chemistry, in vivo detoxification strategies serve as another model for improving the solubility of cannabinoids. CBD is glucuronidated in humans by the liver glucosyltransferases, but to date only minor activity has been demonstrated with UGT1A9 and UGT2B7 in in vitro assays (U.S. Pat. No. 8,410,064). In vitro assays showed that cannabinol (CBN) is efficiently glucuronidated by the Human UGT1A10 (U.S. Pat. No. 8,410,064). The glucuronidation of CBD is one mechanism to increase CBD solubility and facilitate removal and excretion through the kidneys. Searching for glucosyltransferase activity towards cannabinoids, cannabinol was found to be glycosylated when incubated with in vitro cell culture of Pinellia ternata (Tanaka 1993). Similarly, cannabidiol was shown to be glycosylated when incubated with tissue cultures from Pinellia ternata and Datura inoxia, yielding CBD-6′-O-β-D-glucopyranoside and CBD-(2′,6′)-O-β-D-diglucopyranoside (Tanaka 1996). These biotransformation studies demonstrate the potential for limited glycosylation of these two compounds to occur by unknown plant glucosyltransferases, and for them to be produced in minute quantities, but to date, no specific plant glucosyltransferase proteins capable of glycosylation of cannabinoids have been identified, no cannabinoid glycosides been produced in large, purified quantities, and the biological activity or pharmaceutical properties of cannabinoid glycosides have never been characterized.
  • Cannabinoids contain a hydroxylated hydrophobic backbone, similar to the steviol backbone of steviol glycosides found in the Stevia rebaudiana plant. UGT76G1 is a glucosyltransferase from Stevia that is capable of transferring a secondary glucose to the 3C-hydroxyl of the primary glycosylation on both C13-OH and C19-COOH position of the steviol glycoside, and thus its substrates include steviolmonoside, stevioside, rubusoside, RebA, RebD, RebG, RebE, etc. (Richman et al. 2005, Stevia First Corp unpublished work). The substrate recognition site of UGT76G1 is capable of binding and glycosylating multiple steviol glycosides, but it was previously not known to have glycosylation activity towards any other glycosides, and there previously was no established activity of UGT76G1 towards any aglycone compounds at all. As UGT76G1 is capable of glycosylating steviol glycosides on the primary sugar located on both C13 hydroxyl group and the C19 carboxyl group it demonstrates bi-functional glycosylation. Cyclodextrin glucanotransferase (CGTase, Toruzyme 3.0L, Novozymes Inc.) is a member of the amylase family of enzymes and is best known for its ability to cyclize maltodextrin chains. A lesser known activity of CGTase is disproportionation of linear maltodextrin chains and transfer to an acceptor sugar molecule (Li 2012).
  • There are no known cannabinoid glycosides available as cannabinoid prodrugs. Nor is there a known method for the efficient regioselective production of cannabinoid glycosides, which is necessary in order to produce large, purified quantities of individual glycosides and to assess their pharmaceutical properties, including evaluation of in vivo drug pharmacokinetics and pharmacodynamics. To solve the aforementioned problem, screening of glucosyltransferase enzymes from various organisms has been conducted to identify candidates for the glycosylation of cannabinoids, and to identify cannabinoid glycosides as potential prodrugs of cannabinoids, and as novel cannabinoid compositions with novel properties and functions.
  • This background information is provided to reveal information believed by the applicant to be of possible relevance to the present invention. No admission is necessarily intended, nor should be construed, that any of the preceding information constitutes prior art against the present invention.
  • SUMMARY OF THE INVENTION
  • The present invention relates to novel cannabinoid glycoside prodrugs and methods for their production by enzyme-mediated carbohydrate transfer.
  • An object of the present invention is to provide a cannabinoid glycoside prodrug. In accordance with an aspect of the present invention, there is provided a cannabinoid glycoside prodrug compound having formula (I):
  • Figure US20220168428A1-20220602-C00001
  • wherein R is H, β-D-glucopyranosyl, or 3-O-β-D-glucopyranosyl-β-D-glucopyranosyl; R′ is H or β-D-glucopyranosyl, or 3-O-β-D-glucopyranosyl-β-D-glucopyranosyl; and A is an aglycone moiety formed through reaction of a hydroxyl group on a cannabinoid compound, an endocannabinoid compound, or a vanilloid compound, or a pharmaceutically compatible salt thereof.
  • In accordance with another aspect of the present invention, there is provided a method for the site-specific delivery of a cannabinoid drug to a subject, comprising the step of administering a cannabinoid glycoside prodrug in accordance with the present invention to a subject in need thereof.
  • In accordance with another aspect of the present invention, there is provided a method of producing a cannabinoid glycoside, comprising incubating a cannabinoid aglycone with one or more sugar donors in the presence of one or more glycosyltransferases.
  • Further aspects of the technology described herein will be brought out in the following portions of the specification, wherein the detailed description is for the purpose of fully disclosing preferred embodiments of the technology without placing limitations thereon.
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1A illustrates aglycones employed in the glycosylation methods of the present invention. FIG. 1B illustrates the possible points of glycosylation on the aglycones.
  • FIG. 2 illustrates possible products of the glycosylation of cannabidiol (CBD).
  • FIG. 3 illustrates possible products of the glycosylation of cannabidivarin (CBDV).
  • FIG. 4 illustrates possible rotational products of the glycosylation of cannabidiol (CBD).
  • FIG. 5 illustrates possible rotational products of the glycosylation of cannabidivarin (CBDV).
  • FIG. 6 illustrates the proposed super positioning of the substrate cannabidiol (CBD) in the catalytic site of UGT76G1.
  • FIG. 7 illustrates possible products of the glycosylation of tetrahydrocannabinol (A9-THC).
  • FIG. 8 illustrates possible products of the glycosylation of cannabinol (CBN).
  • FIG. 9 illustrates possible products of the glycosylation of arachidonoyl ethanolamide (AEA).
  • FIG. 10 illustrates possible products of the glycosylation of 2-arachidonoyl ethanolamide (2-AG).
  • FIG. 11 illustrates possible products of the glycosylation of 1-arachidonoyl ethanolamide (1-AG).
  • FIG. 12 illustrates possible products of the glycosylation of N-docosahexaenoylethanolamine (DHEA).
  • FIG. 13 illustrates possible products of the glycosylation of capsaicin.
  • FIG. 14 illustrates possible products of the glycosylation of vanillin.
  • FIGS. 15A and 15B illustrate possible products of the glycosylation of curcumin.
  • FIG. 16 is an HPLC linetrace of the reaction products of the glycosylation of CBD.
  • FIG. 17 is an HPLC linetrace of the reaction products of the glycosylation of CBDV.
  • FIG. 18 is an HPLC linetrace of the reaction products of the glycosylation of A9-THC.
  • FIG. 19 is an HPLC linetrace of the reaction products of the glycosylation of CBN.
  • FIG. 20 is an HPLC linetrace of the reaction products of the glycosylation of 1-AG and 2-AG.
  • FIG. 21 is an HPLC linetrace of the reaction products of the glycosylation of synaptamide (DHEA).
  • FIG. 22 is an HPLC linetrace of the reaction products of the glycosylation of AEA.
  • FIG. 23 is an HPLC linetrace of the reaction products of the glycosylation of vanillin.
  • FIG. 24 is an HPLC linetrace of the reaction products of the glycosylation of capsaicin.
  • FIG. 25 is an HPLC linetrace of the reaction products of the glycosylation of CBDg1 (VB104) with the glycosyltransferase UGT76G1.
  • FIG. 26 is an HPLC linetrace of the reaction products of the glycosylation of CBDg1 (VB104) with the glycosyltransferase Os03g0702000
  • FIG. 27 is a 1NMR spectrum of an isolated product, VB104, of the glycosylation of CBD.
  • FIG. 28 is a 1NMR spectrum of an isolated product, VB110 of the glycosylation of CBD.
  • FIG. 29 is a plot of C18 retention times vs c Log P values for selected cannabinoids and cannabinoid glycosides.
  • FIG. 30A is a graphical presentation of the results of the analysis of the small intestine extracts of a bioavailability assay.
  • FIG. 30B is a graphical presentation of the results of the analysis of the large intestine extracts of a bioavailability assay
  • DETAILED DESCRIPTION OF THE INVENTION
  • The following abbreviations are used throughout:
  • CB Cannabinoid
  • CBD Cannabidiol.
  • CBDV Cannabidivarin
  • CBG Cannabigerol
  • Δ9-THC or THC Tetrahydrocannabinol
  • CBN Cannabinol
  • CBNV Cannabinavarin
  • CBDA Cannabidiolic acid
  • THCV Tetrahydrocannabivarin
  • UGT UDPG-dependent glucosyltransferase
  • UDPG Uridine diphosphoglucose
  • UDP Uridine diphosphate
  • AEA Arachidonoyl ethanolamide (aka, anandamide)
  • 2-AG 2-Arachidonoyl ethanolamide.
  • 1-AG 1-Arachidonoyl ethanolamide.,
  • DHEA N-Docosahexaenoylethanolamine (aka, synaptamide)
  • SUS Sucrose synthase.
  • The term “glucopyranoside” is used for naming molecules and is shorthand for a 3-D-glucose attached through the hydroxyl at the 1-position (the anomeric carbon) of the glucose to the aglycone.
  • The term “aglycone” is used in the present application to refer to the non-glycosidic portion of a glycoside compound.
  • The term “prodrug” refers to a compound that, upon administration, must undergo a chemical conversion by metabolic processes before becoming an active pharmacological agent.
  • The term “cannabinoid glycoside prodrug” refers generally to the glycosides of cannabinoid compounds, endocannabinoid compounds and vanilloid compounds. The cannabinoid glycoside prodrug undergoes hydrolysis of the glycosidic bond, typically by action of a glycosidase, to release the active cannabinoid, endocannabinoid or vanilloid compounds to a desired site in the body of the subject. The cannabinoid glycoside prodrug of the present invention may also be referred to using the term “cannaboside”.
  • The term “cannabinoid” is used in the present application to refer generally to compounds found in cannabis and which act on cannabinoid receptors. “Cannabinoid” compounds include, but are not limited to, cannabidiol (CBD), cannabidivarin (CBDV), cannabigerol (CBG), tetrahydrocannabinol (Δ9-THC or THC), cannabinol (CBN), cannabidiolic acid (CBDA), and tetrahydrocannabivarin (THCV). Particularly preferred cannabinoids compounds are CBD, CBDV, THC and CBN.
  • The term “endocannabinoid” is used in the present application to refer to compounds including arachidonoyl ethanolamide (anandamide, AEA), 2-arachidonoyl ethanolamide (2-AG), 1-arachidonoyl ethanolamide (1-AG), and docosahexaenoyl ethanolamide (DHEA, synaptamide), oleoyl ethanolamide (OEA), eicsapentaenoyl ethanolamide, prostaglandin ethanolamide, docosahexaenoyl ethanolamide, linolenoyl ethanolamide, 5(Z),8(Z),11(Z)-eicosatrienoic acid ethanolamide (mead acid ethanolamide), heptadecanoul ethanolamide, stearoyl ethanolamide, docosaenoyl ethanolamide, nervonoyl ethanolamide, tricosanoyl ethanolamide, lignoceroyl ethanolamide, myristoyl ethanolamide, pentadecanoyl ethanolamide, palmitoleoyl ethanolamide, docosahexaenoic acid (DHA). Particularly preferred endocannabinoids are AEA, 2-AG, 1-AG, and DHEA.
  • The term “vanilloid” is used in the present application to refer to compounds comprising a vanillyl group and which act on vanilloid receptors like TRPV1. “Vanilloid” compounds include, but are not limited to, vanillin, capsaicin and curcumin.
  • As used herein, the term “about” refers to a +/−10% variation from the nominal value. It is to be understood that such a variation is always included in a given value provided herein, whether or not it is specifically referred to.
  • The term “subject” or “patient” as used herein refers to an animal in need of treatment. In one embodiment, the animal is a human.
  • Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs.
  • In accordance with the present invention, cannabinoids, endocannabinoids and vanilloids are employed as substrates for glucosyltransferases to which one or more sugar molecules are attached to create novel cannabinoid glycoside prodrugs. The resulting cannabinoid glycoside prodrugs demonstrate site-specific or tissue-specific delivery, improved aqueous solubility for improved pharmacological delivery, and/or sustained or delayed release of the cannabinoid, endocannabinoid and vanilloid drug molecules.
  • Also in accordance with the present invention, the cannabinoid glycoside prodrugs are converted upon hydrolysis of the glycosidic bond to provide the active cannabinoid, endocannabinoid and vanilloid drug. Accordingly, the present invention has demonstrated that glycosides with a hydrophobic aglycone moiety undergo glucose hydrolysis in the gastrointestinal tract or in tissues having increased expression of glycosidases, yielding the hydrophobic cannabinoid compound in the targeted tissue or organ.
  • The glucose residues of glycosides are commonly acid-hydrolyzed in the stomach or cleaved by glycosidase enzymes in the intestinal tract, including by alpha-glycosidases and beta-glycosidases, which are expressed by intestinal microflora across different regions of the intestine. Accordingly, glycosides are hydrolyzed upon ingestion to release the desired compound into the intestines or target tissues.
  • In one embodiment, glycosylation of cannabinoid drugs provides cannabinoid glycoside prodrugs capable of persisting in the acidic stomach environment upon oral administration, thereby allowing delivery of the prodrug into the large intestine, where the cannabinoid aglycones can be liberated by glycosidases produced by colonic bacteria.
  • In one embodiment, glycosylation of cannabinoid drugs provides cannabinoid glycoside prodrugs suitable for targeted delivery to tissues having increased expression of glycosidases. Upon parenteral administration of the cannabinoid glycoside prodrug formulation to the subject, the cannabinoid aglycones are liberated by the glycosidases in the target tissues.
  • It is also within the scope of the present invention that the cannabinoid glycoside prodrug are also useful as pharmaceutical agents without glucose cleavage, where they exhibit novel pharmacodynamic properties compared to the parent compound alone. The increased aqueous solubility of the cannabinoid glycoside prodrugs of the present invention also enables new formulations for delivery in transdermal or aqueous formulations that would not have been achievable if formulating hydrophobic cannabinoid, endocannabinoid and vanilloid molecules.
  • In one embodiment of the present invention, there are provided cannabinoid glycoside prodrug compounds having formula (I):
  • Figure US20220168428A1-20220602-C00002
  • or a pharmaceutically compatible salt thereof, wherein R is H, β-D-glucopyranosyl, or 3-O-β-D-glucopyranosyl-β-D-glucopyranosyl; R′ is H or β-D-glucopyranosyl, or 3-O-β-D-glucopyranosyl-β-D-glucopyranosyl; and A is an aglycone moiety formed through reaction of a hydroxyl group on a cannabinoid compound, an endocannabinoid compound, or a vanilloid compound.
  • In accordance with one embodiment of the present invention, A is A′, A″ or A′″;
  • wherein A′ is:
  • Figure US20220168428A1-20220602-C00003
      • wherein A″ is:
  • Figure US20220168428A1-20220602-C00004
  • and wherein A′″ is:
  • Figure US20220168428A1-20220602-C00005
  • wherein G is H, β-D-glucopyranosyl, 3-O-β-D-glucopyranosyl-β-D-glucopyranosyl, or β-D-glucopyranosyl-(1→3)-β-D-glucopyranosyl-(1→3)-D-glucopyranosyl; or a pharmaceutically compatible salt thereof.
  • In accordance with one embodiment of the present invention, the cannabinoid glycoside prodrug is a glycoside of a cannabinoid, wherein the prodrug has the formula (I′):
  • Figure US20220168428A1-20220602-C00006
  • wherein R is H, β-D-glucopyranosyl, or 3-O-β-D-glucopyranosyl-β-D-glucopyranosyl; R′ is H, β-D-glucopyranosyl, or 3-O-β-D-glucopyranosyl-β-D-glucopyranosyl; and wherein A′ is:
  • Figure US20220168428A1-20220602-C00007
  • wherein G is β-D-glucopyranosyl, 3-O-β-D-glucopyranosyl-β-D-glucopyranosyl, or β-D-glucopyranosyl-(1-3)-β-D-glucopyranosyl-(1-3)-D-glucopyranosyl.
  • Compounds of Formula (I′) include the compounds listed in Tables 1 to 4.
  • Exemplary cannabidiol (CBD)-glycosides falling within the scope of Formula (I′), produced by the glycosylation of CBD (VB101) in accordance with the present invention, include:
  • Figure US20220168428A1-20220602-C00008
    Figure US20220168428A1-20220602-C00009
    Figure US20220168428A1-20220602-C00010
  • Exemplary cannabidivarin (CBDV)-glycosides falling within the scope of Formula (I′), produced by the glycosylation of CBDV (VB201) in accordance with the present invention, include:
  • Figure US20220168428A1-20220602-C00011
    Figure US20220168428A1-20220602-C00012
    Figure US20220168428A1-20220602-C00013
  • Exemplary tetrahydrocannabinol (Δ9-THC)-glycosides falling within the scope of Formula (I′), produced by the glycosylation of Δ9-THC (VB301) in accordance with the present invention, include:
  • Figure US20220168428A1-20220602-C00014
    Figure US20220168428A1-20220602-C00015
  • Exemplary cannabinol (CBN)-glycosides falling within the scope of Formula (I′), produced by the glycosylation of CBN (VB401) in accordance with the present invention, include:
  • Figure US20220168428A1-20220602-C00016
    Figure US20220168428A1-20220602-C00017
  • In accordance with one embodiment of the present invention, the cannabinoid glycoside prodrug is a glycoside of an endocannabinoid, the prodrug having the formula (I″):
  • Figure US20220168428A1-20220602-C00018
  • wherein
  • R is H, β-D-glucopyranosyl, or 3-O-β-D-glucopyranosyl-β-D-glucopyranosyl; R′ is H, β-D-glucopyranosyl, or 3-O-β-D-glucopyranosyl-β-D-glucopyranosyl; and
  • wherein A″ is:
  • Figure US20220168428A1-20220602-C00019
  • Compounds of Formula (I″) include the compounds listed in Tables 5 to 8.
  • Exemplary arachidonoyl ethanolamide (AEA)-glycosides falling within the scope of Formula (I″), produced by the glycosylation of AEA (VB501) in accordance with the present invention, include:
  • Figure US20220168428A1-20220602-C00020
  • Exemplary 2-arachidonoyl ethanolamide (2-AG)-glycosides falling within the scope of Formula (I″), produced by the glycosylation of 2-AG (VB601) in accordance with the present invention, include:
  • Figure US20220168428A1-20220602-C00021
    Figure US20220168428A1-20220602-C00022
  • Exemplary 1-arachidonoyl ethanolamide (1-AG)-glycosides falling within the scope of Formula (I″), produced by the glycosylation of 1-AG (VB701) in accordance with the present invention, include:
  • Figure US20220168428A1-20220602-C00023
    Figure US20220168428A1-20220602-C00024
  • Exemplary N-docosahexaenoylethanolamine (DHEA)-glycosides falling within the scope of Formula (I″), produced by the glycosylation of DHEA (VB801) in accordance with the present invention, include:
  • Figure US20220168428A1-20220602-C00025
  • In accordance with one embodiment of the present invention, the cannabinoid glycoside prodrug is a glycoside of a vanilloid, the prodrug having the formula (I′″):
  • Figure US20220168428A1-20220602-C00026
  • wherein
  • R is H, β-D-glucopyranosyl, or 3-O-β-D-glucopyranosyl-β-D-glucopyranosyl;
  • R′ is H or β-D-glucopyranosyl, or 3-O-β-D-glucopyranosyl-β-D-glucopyranosyl; and,
    wherein A′″ is:
  • Figure US20220168428A1-20220602-C00027
  • Compounds of Formula (I′″) include the compounds listed in Tables 9 to 11.
  • Exemplary capsaicin-glycosides falling within the scope of Formula (I′″), produced by the glycosylation of capsaicin (VB901) in accordance with the present invention, include:
  • Figure US20220168428A1-20220602-C00028
  • Exemplary vanillin-glycosides falling within the scope of Formula (I′″), produced by the glycosylation of vanillin (VB1001) in accordance with the present invention, include:
  • Figure US20220168428A1-20220602-C00029
    Figure US20220168428A1-20220602-C00030
  • Exemplary curcumin-glycosides falling within the scope of Formula (I′″), produced by the glycosylation of curcumin (VB1101) in accordance with the present invention, include:
  • Figure US20220168428A1-20220602-C00031
    Figure US20220168428A1-20220602-C00032
    Figure US20220168428A1-20220602-C00033
    Figure US20220168428A1-20220602-C00034
    Figure US20220168428A1-20220602-C00035
  • In one embodiment, there is provided a method for the site-specific delivery of a cannabinoid drug to a subject, comprising the step of administering to a subject in need thereof one or more cannabinoid glycoside prodrugs in accordance with the present invention. In one embodiment, the site of delivery is the large intestine. In one embodiment, the site of delivery is the rectum. In one embodiment, the site of delivery is the liver. In one embodiment, the site of delivery is the skin.
  • In one embodiment, there is provided a method for facilitating the transport of a cannabinoid drug to the brain through intranasal, stereotactic, or intrathecal delivery, or delivery across the blood brain barrier of a subject comprising administering a cannabinoid glycoside prodrug in accordance with the present invention to a subject in need thereof.
  • In accordance with the present invention, the cannabinoid glycoside prodrugs are useful in the treatment of conditions that benefit from or can be ameliorated with the administration of a cannabinoid drug. Conditions that can be treated or ameliorated through the administration of cannabinoid glycoside prodrugs of the present invention, include but are not limited to, inflammatory bowel disease including induction of remission from Crohn's disease, and colitis and induction of remission from ulcerative colitis. Among the benefits that can be achieved through the administration of cannabinoid glycoside prodrugs of the present invention are decreased inflammation of the intestines and rectum, decreased pain in the intestines, rectum, as well as decrease in neuropathic pain and abdominal pain, and inhibition of proliferation or cytotoxicity against colorectal cancer. Additional treatment indications, effects, or applications for cannabinoids or cannabinoid glycosides may include but are not limited to anorexia, nausea, emesis, pain, wasting syndrome, HIV-wasting, chemotherapy induced nausea and vomiting, epilepsy, schizophrenia, irritable bowel syndrome, cramping, spasticity, seizure disorders, alcohol use disorders, substance abuse disorders, addiction, cancer, amyotrophic lateral sclerosis, glioblastoma multiforme, glioma, increased intraocular pressure, glaucoma, cannabis use disorders, Tourette's syndrome, dystonia, multiple sclerosis, white matter disorders, demyelinating disorders, chronic traumatic encephalopathy, leukoencephalopathies, Guillain-Barre syndrome, inflammatory bowel disorders, gastrointestinal disorders, bacterial infections, MRSA, sepsis, septic shock, viral infections, arthritis, dermatitis, Rheumatoid arthritis, systemic lupus erythematosus, anti-inflammatory, anti-convulsant, anti-psychotic, anti-oxidant, neuroprotective, anti-cancer, immunomodulatory effects, neuropathic pain, neuropathic pain associated with post-herpetic neuralgia, diabetic neuropathy, shingles, burns, actinic keratosis, oral cavity sores and ulcers, post-episiotomy pain, psoriasis, pruritis, gout, chondrocalcinosis, joint pain, fibromyalgia, musculoskeletal pain, neuropathic-postoperative complications.
  • In one embodiment, the cannabinoid glycoside prodrug is administered in a pharmaceutical composition further comprising a pharmaceutically acceptable carrier, diluent, excipient, or adjuvant. In one embodiment, the pharmaceutical compositions comprise one or more cannabinoid glycoside prodrugs and one or more pharmaceutically acceptable carriers, diluents, excipients and/or adjuvants. For administration to a subject, the pharmaceutical compositions can be formulated for administration by a variety of routes including but not limited to oral, topical, rectal, parenteral, and intranasal administration.
  • The pharmaceutical compositions may comprise from about 1% to about 95% of a cannabinoid glycoside prodrug of the invention. Compositions formulated for administration in a single dose form may comprise, for example, about 20% to about 90% of the cannabinoid glycoside prodrug of the invention, whereas compositions that are not in a single dose form may comprise, for example, from about 5% to about 20% of the cannabinoid glycoside prodrug of the invention. Non-limiting examples of unit dose forms include tablets, ampoules, dragees, suppositories, and capsules.
  • In a preferred embodiment, the pharmaceutical compositions are formulated for oral administration. Pharmaceutical compositions for oral administration can be formulated, for example, as tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsion hard or soft capsules, or syrups or elixirs. Such compositions can be prepared according to standard methods known in the art for the manufacture of pharmaceutical compositions and may contain one or more agents selected from the group of sweetening agents, flavouring agents, colouring agents and preserving agents in order to provide pharmaceutically elegant and palatable preparations. Tablets contain the active ingredient in admixture with suitable non-toxic pharmaceutically acceptable excipients including, for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, such as corn starch, or alginic acid; binding agents, such as starch, gelatine or acacia, and lubricating agents, such as magnesium stearate, stearic acid or talc. The tablets can be uncoated, or they may be coated by known techniques in order to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a time delay material such as glyceryl monosterate or glyceryl distearate may be employed to further facilitate delivery of the drug compound to the desired location in the digestive tract.
  • Pharmaceutical compositions for oral use can also be presented as hard gelatine capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatine capsules wherein the active ingredient is mixed with water or an oil medium such as peanut oil, liquid paraffin or olive oil.
  • Pharmaceutical compositions formulated as aqueous suspensions contain the active compound(s) in admixture with one or more suitable excipients, for example, with suspending agents, such as sodium carboxymethylcellulose, methyl cellulose, hydropropylmethylcellulose, sodium alginate, polyvinylpyrrolidone, hydroxypropyl-β-cyclodextrin, gum tragacanth and gum acacia; dispersing or wetting agents such as a naturally-occurring phosphatide, for example, lecithin, or condensation products of an alkylene oxide with fatty acids, for example, polyoxyethyene stearate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example, hepta-decaethyleneoxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol for example, polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides, for example, polyethylene sorbitan monooleate. The aqueous suspensions may also contain one or more preservatives, for example ethyl, or n-propyl p-hydroxy-benzoate, one or more colouring agents, one or more flavouring agents or one or more sweetening agents, such as sucrose, stevia, or saccharin.
  • Pharmaceutical compositions can be formulated as oily suspensions by suspending the active compound(s) in a vegetable oil, for example, arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin. The oily suspensions may contain a thickening agent, for example, beeswax, hard paraffin or cetyl alcohol. Sweetening agents such as those set forth above, and/or flavouring agents may be added to provide palatable oral preparations. These compositions can be preserved by the addition of an anti-oxidant such as ascorbic acid.
  • The pharmaceutical compositions can be formulated as a dispersible powder or granules, which can subsequently be used to prepare an aqueous suspension by the addition of water. Such dispersible powders or granules provide the active ingredient in admixture with one or more dispersing or wetting agents, suspending agents and/or preservatives. Suitable dispersing or wetting agents and suspending agents are exemplified by those already mentioned above. Additional excipients, for example, sweetening, flavouring and colouring agents, can also be included in these compositions.
  • Pharmaceutical compositions of the invention can also be formulated as oil-in-water emulsions. The oil phase can be a vegetable oil, for example, olive oil or arachis oil, or a mineral oil, for example, liquid paraffin, or it may be a mixture of these oils. Suitable emulsifying agents for inclusion in these compositions include naturally-occurring gums, for example, gum acacia or gum tragacanth; naturally-occurring phosphatides, for example, soy bean, lecithin; or esters or partial esters derived from fatty acids and hexitol, anhydrides, for example, sorbitan monoleate, and condensation products of the said partial esters with ethylene oxide, for example, polyoxyethylene sorbitan monoleate. The emulsions can also optionally contain sweetening and flavouring agents.
  • Pharmaceutical compositions can be formulated as a syrup or elixir by combining the active ingredient(s) with one or more sweetening agents, for example glycerol, propylene glycol, sorbitol or sucrose. Such formulations can also optionally contain one or more demulcents, preservatives, flavouring agents and/or colouring agents.
  • If desired, other active ingredients may be included in the compositions. In one embodiment, the glycoside prodrugs may be combined with other ingredients or substances that have glycosidase activity, or that may in other ways alter drug metabolism and pharmacokinetic profile of various compounds in vivo, including ones in purified form as well as such compounds found within food, beverages, and other products. In one embodiment, the cannabinoid glycoside prodrug is administered in combination with, or formulated together with, substances that have direct glycosidase activity, or that contribute to modifications to the gut microflora that will alter the glycosidase activity in one or more regions of the intestines. Examples of such compositions include, but are not limited to, yogurt, prebiotics, probiotics, or fecal transplants.
  • In a further preferred embodiment, the pharmaceutical compositions are formulated for parenteral administration. The term “parenteral” as used herein includes subcutaneous injections, intravenous, intramuscular, intrathecal, intrasternal injection or infusion techniques.
  • Parenteral pharmaceutical compositions can be formulated as a sterile injectable aqueous or oleaginous suspension according to methods known in the art and using one or more suitable dispersing or wetting agents and/or suspending agents, such as those mentioned above. The sterile injectable preparation can be a sterile injectable solution or suspension in a non-toxic parentally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol. Acceptable vehicles and solvents that can be employed include, but are not limited to, water, Ringer's solution, lactated Ringer's solution and isotonic sodium chloride solution. Other examples include, sterile, fixed oils, which are conventionally employed as a solvent or suspending medium, and a variety of bland fixed oils including, for example, synthetic mono- or diglycerides. Fatty acids such as oleic acid can also be used in the preparation of injectables.
  • Due to the highly lipophilic nature of cannabinoids, these molecules are typically poorly absorbed through membranes such as the skin of mammals, including humans, and the success of transdermally administering therapeutically effective quantities of cannabinoid to a subject in need thereof within a reasonable time frame and over a suitable surface area has been substantially limited. It is therefore proposed that the cannabinoid glycoside prodrugs of the present invention, through conjugation of the hydrophobic cannabinoid aglycone to the hydrophilic glycosidic moieties, provide a molecule having an amphiphilic character favourable for passive diffusion which should be more readily absorbed through the skin.
  • Accordingly, in one embodiment, the pharmaceutical compositions are formulated for topical administration. Such topical formulations may be presented as, for example, aerosol sprays, powders, sticks, granules, creams, liquid creams, pastes, gels, lotions, ointments, on sponges or cotton applicators, or as a solution or a suspension in an aqueous liquid, a non-aqueous liquid, an oil-in-water emulsion, or a water-in-oil liquid emulsion.
  • Topical pharmaceutical compositions can be formulated with thickening (gelling) agents. The thickening agent used herein may include anionic polymers such as polyacrylic acid (CARBOPOL® by Noveon, Inc., Cleveland, Ohio), carboxypolymethylene, carboxymethylcellulose and the like, including derivatives of Carbopol® polymers, such as Carbopol® Ultrez 10, Carbopol® 940, Carbopol® 941, Carbopol® 954, Carbopol® 980, Carbopol® 981, Carbopol® ETD 2001, Carbopol® EZ-2 and Carbopol® EZ-3, and other polymers such as Pemulen® polymeric emulsifiers, and Noveon® polycarbophils. Thickening agents or gelling agents are present in an amount sufficient to provide the desired rheological properties of the composition.
  • Topical pharmaceutical compositions can be formulated with a penetration enhancer. Non-limiting examples of penetration enhancing agents include C8-C22 fatty acids such as isostearic acid, octanoic acid, and oleic acid; C8-C22 fatty alcohols such as oleyl alcohol and lauryl alcohol; lower alkyl esters of C8-C22 fatty acids such as ethyl oleate, isopropyl myristate, butyl stearate, and methyl laurate; di(lower)alkyl esters of C6-C22 diacids such as diisopropyl adipate; monoglycerides of C8-C22 fatty acids such as glyceryl monolaurate; tetrahydrofurfuryl alcohol polyethylene glycol ether; polyethylene glycol, propylene glycol; 2-(2-ethoxyethoxyl)ethanol; diethylene glycol monomethyl ether; alkylaryl ethers of polyethylene oxide; polyethylene oxide monomethyl ethers; polyethylene oxide dimethyl ethers; dimethyl sulfoxide; glycerol; ethyl acetate; acetoacetic ester; N-alkylpyrrolidone; and terpenes.
  • The topical pharmaceutical compositions can further comprise wetting agents (surfactants), lubricants, emollients, antimicrobial preservatives, and emulsifying agents as are known in the art of pharmaceutical formations.
  • Transdermal delivery of the cannabinoid glycoside prodrug can be further facilitated through the use of a microneedle array drug delivery system.
  • Other pharmaceutical compositions and methods of preparing pharmaceutical compositions are known in the art and are described, for example, in “Remington: The Science and Practice of Pharmacy” (formerly “Remingtons Pharmaceutical Sciences”); Gennaro, A., Lippincott, Williams & Wilkins, Philadelphia, Pa. (2000).
  • The pharmaceutical compositions of the present invention described above include one or more cannabinoid glycoside prodrugs of the invention in an amount effective to achieve the intended purpose. Thus the term “therapeutically effective dose” refers to the amount of the cannabinoid glycoside prodrug that improves the status of the subject to be treated, for example, by ameliorating the symptoms of the disease or disorder to be treated, preventing the disease or disorder, or altering the pathology of the disease. Determination of a therapeutically effective dose of a compound is well within the capability of those skilled in the art. In one embodiment, cannabinoid glycosides can be combined to enable simultaneous delivery of multiple cannabinoids in a site-specific manner, including THC and CBD, whose effects in some ways may be synergistic (Russo 2006). Accordingly, in one embodiment, the pharmaceutical composition comprises one or more CBD-glycosides and one or more THC-glycosides formulated together in a single dosage form.
  • The exact dosage to be administered to a subject can be determined by the practitioner, in light of factors related to the subject requiring treatment. Dosage and administration are adjusted to provide desired levels of the cannabinoid glycoside prodrug and/or the cannabinoid drug compound obtained upon hydrolysis of the prodrug. Factors which may be taken into account when determining an appropriate dosage include the severity of the disease state, general health of the subject, age, weight, and gender of the subject, diet, time and frequency of administration, drug combination(s), reaction sensitivities, and tolerance/response to therapy. Dosing regimens can be designed by the practitioner depending on the above factors as well as factors such as the half-life and clearance rate of the particular formulation.
  • In accordance with the present invention, there is provided a method of producing a cannabinoid glycoside, comprising incubating a cannabinoid aglycone with one or more sugar donors in the presence of one or more glycosyltransferases.
  • In one embodiment, the one or more glycosyltransferases is a UGT76G1 or UGT76G1-like glucosyltransferase. In one embodiment, the one or more glycosyltransferases comprise a UGT76G1 or UGT76G1-like glucosyltransferase and a Os03g0702000 or Os03g0702000-like glucosyltransferase.
  • In one embodiment, the one or more sugar donors are selected from the group consisting of UDP-glucose, UDP-glucuronic acid, UDP-mannose, UDP-fructose, UDP-xylose, UDP-rhamnose, UDP-fluoro-deoxyglucose, and combinations thereof. In a preferred embodiment, the sugar donor is UDP-glucose.
  • In accordance with the present invention, the cannabinoid aglycone is a cannabinoid, an endocannabinoid, or a vanilloid. In a preferred embodiment, the cannabinoid glycoside prodrug produced by the methods of the present invention is a compound of the Formula (I).
  • In one embodiment, the method of producing a cannabinoid glycoside comprises incubating a cannabinoid aglycone with UDP-glucose, in the presence of a UGT76G1 or UGT76G1-like glucosyltransferase under conditions that allow for glycosylation.
  • In one embodiment, the method of producing a cannabinoid glycoside comprises incubating a cannabinoid aglycone with one or more sugar donors in the presence of a first glycosyltransferase and a second glycosyltransferase under conditions which allow for glycosylation. In one embodiment, sugar donor is UDP-glucose, the first glycosyltransferase is a UGT76G1 or UGT76G1-like glucosyltransferase, and the second glycosyltransferase is a Os03g0702000 or Os03g0702000-like glucosyltransferase.
  • In one embodiment, the method of producing a cannabinoid glycoside comprises incubating a cannabinoid aglycone with UDP-glucose in the presence of a UGT76G1 or UGT76G1-like glucosyltransferase and Os03g0702000 or Os03g0702000-like glucosyltransferase under conditions which allow for glycosylation.
  • In one embodiment, the method of producing a cannabinoid glycoside comprises incubating a cannabinoid aglycone with maltodextrin, in the presence of a cyclodextrin glucanotransferase under conditions that allow for glycosylation.
  • In one embodiment, the method of producing a cannabinoid glycoside comprises incubating a cannabinoid aglycone with UDP-glucose and maltodextrin in the presence of a UGT76G1 or UGT76G1-like glucosyltransferase and cyclodextrin glucanotransferase under conditions which allow for glycosylation.
  • In a preferred embodiment, the glycosyltransferase employed in the methods of producing the cannabinoid glycoside is UGT76G1 or UGT76G1-like glucosyltransferase. In one embodiment, the UGT76G1 or UGT76G1-like glucosyltransferase comprises the sequence as set forth in SEQ ID NO:1, 3, 5 or 7.
  • In one embodiment, the glycosyltransferase employed in the methods of producing the cannabinoid glycoside is Os03g0702000 or Os03g0702000-like glucosyltransferase. In one embodiment, the Os03g0702000 or Os03g0702000-like glucosyltransferase comprises the sequence as set forth in SEQ ID NO:9.
  • In one embodiment, the method of producing the cannabinoid glycoside further comprises incubating with sucrose synthase. In one embodiment, the sucrose synthase comprises the sequence as set forth in SEQ ID NO: 15, 17, 19, 21, 23 or 25.
  • In one embodiment, the method for the production of a cannabinoid glycoside prodrug comprises expressing one or more of the glycosyltransferases in a cell or plant which produces the cannabinoid aglycone and isolating the cannabinoid glycoside prodrug.
  • Glycosylation of cannabinoids improves solubility in aqueous solutions, as demonstrated by accelerated elution from an C18 analytical HPLC column, indicating that the new cannabinoid-glycosides require far less organic solvent for elution from the hydrophobic chromatography column. This improved solubility was further demonstrated by testing the aqueous solubility of purified solid cannabosides, where solutions were successfully prepared up to 500 mg/ml (50% mass/volume) with a mixture of higher glycoside forms of cannabosides. Given the markedly improved solubility and novel secondary and tertiary glycosylation on cannabinoids, glycosylated cannabinoids can act as efficient prodrugs for selective delivery of cannabinoids to desired tissues where the glucose molecules can be hydrolyzed to release the aglycone cannabinoids. Additionally the glycosylations promote stability of CBD and CBDV by protecting them from oxidation and ring-closure of the C6′-hydroxyl group, which prevents degradation into Δ9-THC or Δ9-THCV, respectively, and subsequently into cannabinol (CBN) or cannabinavarin (CBNV), respectively
  • Increasing the diversity and complexity of sugar attachments to cannabinoids, and administration of a mixture of glycosides will provide altered prodrug delivery kinetics, thus providing an extended release formulation of the drug. The primary detoxification mechanism for cannabinoids in humans is CYP450 mediated hydroxylation of the C7 methyl group of CBD and CBDV, or the C11 methyl group of THC and CBN, glycosylation of the acceptor hydroxyl groups of the cannabinoid resorcinol ring may afford protection from C7/C11 hydroxylation and subsequent elimination from the body due to steric hindrance preventing the cannabinoid-glycoside from binding in the CYP450 active site. In fact, the hydroxyl groups of CBD are thought to facilitate the binding to the detoxification cytochrome P450 CYP3A4 in the epithelium of the small intestine (Yamaori 2011). Reduced degradation or metabolism in the stomach and small intestine due to these effects could also lead to higher total bioavailability of any glycosylated product upon oral delivery.
  • In some cases, removal of the sugar from glycosides in the body may be required in order for the compounds to exert their primary biological activity. Therefore, glycoside prodrugs may enable stable drug formulations that are resistant to abuse, due to the potential for their primary biological effects to only occur after oral ingestion. As most abuse-deterrent compounds are simply mixing or formulation based deterrents, they can still be compromised by simple physical and chemical methods. As one example, the beta-glycosides described herein will only release the aglycone upon the action of beta-glycosidase enzymes. Beta-glycosidases are known to be secreted by microbes that occupy the large intestines of mammals, therefore upon oral ingestion the glycoside prodrugs will remain glycosylated until they reach the large intestine. A similar approach may be used for abuse-resistant, abuse-deterrent, and site-specific delivery of other compounds through glycosylation. It has been found that the UGT76G1 enzyme (SEQ ID NO.1) from Stevia rebaudiana transfers a glucose molecule from the sugar donor UDP-glucose (UDPG) to the hydroxyl groups of CBD to create novel CBD-O-glycosides (Table 1, FIGS. 2 & 4). The UDPG is inverted by UGT76G1 to produce β-D-glucose residues covalently linked through the to the hydroxyl acceptor sites on CBD. To improve the catalytic efficiency UGT76G1 open reading frame (ORF) codon optimization was performed (SEQ ID NOs. 4 and 6) for expression in Pichia pastoris. Similar to its activity towards steviol glycosides, UGT76G1 is highly productive and has an equilibrium constant (Keq) for CBD of ˜24. Through experimentation and analysis it was determined that UGT76G1 has the unique ability to apply multiple glucose moieties to the CBD molecule. Upon prolonged incubation of CBD with UGT76G1 and UDPG, HPLC analysis of the reaction mixture yielded 8 glycoside product mobility groups, suggesting that UGT76G1 is able to glycosylate both the C2′ and C6′ hydroxyl groups on CBD, as well as glycosylating the primary glucose residues with a secondary and tertiary glucose moieties. The secondary and tertiary glycosylations by UGT76G1 occurs at the C3 hydroxyl group of the recipient sugar (3→1 connectivity), as would be suggested by its activity in Stevia, creating O-(3-1)-glycosides, and the subsequent products. The CBD-glycoside product mobility groups also suggest that CBD can dock in the UGT76G1 active site both forwards and backwards creating a cis-like-conformation for the glycosylations relative to the cannabinoid backbone (mechanism depicted in FIG. 3), or possibly the rotational freedom about the bond at C1′ (C6 described by Mazur 2009) allows the hydroxyl group to rotate after glycosylation, placing the other hydroxyl group adjacent to the UDPG in the active site and creating a trans-like-conformation for the glycosylations on the cannabinoid backbone (mechanism depicted in FIG. 4). Potential CBD molecular docking in the active site of UGT76G1 is depicted in FIG. 6 where CBD is superpositioned over the bi-functional substrate for UGT76G1, Rebaudioside E (RebE) (FIG. 6).
  • As CBD was successfully glycosylated by UGT76G1, CBDV was incubated with UGT76G1 and UDPG to test for glycosylation activity. CBDV depletion was observed upon HPLC analysis, in addition to the appearance of four additional product peak mobility groups, which were dependent on addition of both UGT76G1 and UDPG. The four new products formed displayed the same absorbance characteristics as CDBV and were determined to be the primary glycosides CBDV-2′-O-glucopyranosides, CBDV-6′-O-glucopyranosides, and the secondary glycosides CBDV-2′-O-(3-1)-diglucopyranoside, and CBDV-6′-O-(3-1)-diglucopyranoside (compounds VB202, VB206, VB204 and VB208, respectively, Table 2). With additional reaction time it was determined that higher order glycoside products were also formed. CBDV-glycoside production was similar to CBD-glycosides from UGT76G1 (Table 2), and proceeded to completion with a Keq˜24. Given the number of CBDV-glycoside products, UGT76G1 transfers multiple glucose molecules onto CBDV on both C2′ and C6′ hydroxyl groups, as well as onto the primary and secondary glycosylations.
  • When the cannabinoid Δ9-THC was incubated with UGT76G1 and UDPG, HPLC analysis of the reaction mixture showed three main product peak mobility groups. The three products were identified as Δ9-THC-1-O-glucopyranoside, Δ9-THC-1-O-(3-1)-diglucopyranoside, and Δ9-THC-1-O-(3-1,3-1)-thioglucopyranoside (formal pyran numbering, Table 3, FIG. 7). Given that the rigid structure of Δ9-THC does not have the same rotational freedom as CBD around the C1′ resorcinol ring attachment, the cannabinoid backbone is recognized in the active site of UGT76G1 with the Δ9-THC C1 hydroxyl group situated towards the UDPG sugar donor (pyran numbering, FIG. 1B).
  • As UGT76G1 demonstrated glycosylation activity for all other phytocannabinoids analyzed, it was also tested for glycosylation activity against cannabinol (CBN). Effective glycosylation of CBN by UGT76G1 was observed, in a similar pattern to Δ9-THC, as both share a single hydroxyl recipient group at the C1 position of the resorcinol ring. The activity seen with UGT76G1 is consistent with a broad recognition of cannabinoids by the enzyme active site.
  • Alternative cannabinoid substrates may be inserted into this UGT76G1 glycosylation reaction infrastructure to generate novel cannabinoid-glycosides, given they possess hydroxyl groups in similar positions on the cannabinoid backbone. Ideal candidates are cannabigerol (CBG), cannabichromene (CBC), cannabidiol hydroxyquinone (CBDHQ), HU-331, other isomers of Δ9-THC such as Δ8-THC, etc., and synthetic analogues of Δ9-THC such as HU-210.
  • Similar to the secondary 3→1 glycosylation activity of UGT76G1, it was determined that following a primary glycosylation by UGT76G1, the UGT enzyme Os03g0702000 (SEQ ID NO.9) from Oryza sativa is also capable of transferring an additional glucose moiety from UDP-glucose onto the C2-hydroxyl of the primary sugar (Tables 1-11, FIGS. 7-9 & 12-14). This glycosylation activity is consistent with the activity of UGT Os03g0702000 towards steviol glycosides in establishing C2-hydroxyl secondary glycosylations (2→1 connectivity) on existing primary glucose residues. This secondary glycosylation was observed with CBDV (Table 2, FIG. 3), and THC (Table 2, FIG. 7), generating novel CBDV and Δ9THC-1-O-(2-1)-diglucopyranoside species, respectively. Consistent with broad substrate recognition and reactivity, this activity of Os03g0702000 was further demonstrated for the remainder of the substrates identified in FIG. 1.
  • In addition to the UDPG-dependent glucosyltransferase activity, cyclodextrin-glucanotransferase (CGTase, Toruzyme 3.0L, trademark of Novozymes Inc.) is capable of transferring a short α-(1-4)-maltodextrin chain onto the hydroxyl groups of cannabinoids. The CGTase is also capable of glycosylating primary and secondary glycosylations established by UGT76G1 and Os03g0702000, resulting in carbohydrate attachments that start with β-D-glucose molecules, but terminating in α-D-glucose molecules termed β-primed-α-glucosyl (Tables 1-11). α-glycosylation by cyclodextrin glucanotransferase mediated maltodextrin transfer can occur on any of the hydroxyl groups of the primary or secondary sugars covalently linked to the cannabinoid. One skilled in the art will appreciate that this makes possible any number of conformations of α-glycosyl chains linked to the glycosides listed in Tables 1-11.
  • Alternative enzymes with homology to UGT76G1 and Os03g0702000 may be used to produce the same glycosylation of cannabinoids. Suitable enzymes for establishing the primary glycosylation similar to UGT76G1 are additional members of the UGT76 clade such as UGT76G2 or UGT76H1. BLAST results with the UGT76G1 protein sequence yield a maximum homology of 49% identity, as much as 66% positives (similar identity). Ideal candidates may have low overall peptide identity or similarity, but will likely have conserved amino acids at the opening adjacent to the UDPG catalytic site. This sequence is exemplified by a leucine at position 379, and a broader peptide sequence of SDFGLDQ (AA's 375 to 381 of UGT76G1). Suitable enzymes for producing the secondary glycosylation of Os03g0702000 are members of the UGT91 clade, including UGT91 D1 and UGT91 D2.
  • The glycosylation reactions performed herein included UDP-glucose as the nucleotide sugar donor, however there is some cross-reactivity amongst UGTs that allows for use of alternative nucleotide sugars such as UDP-glucuronic acid, etc. Glucuronic acid is the predominant nucleotide sugar utilized by phase-II detoxification UGTs in the liver, and cannabinoid-glucuronides are a common detoxification product. Additional nucleotide sugars which could be used to donate carbohydrate moieties to create novel glycosides with similar properties include UDP-glucuronic acid, UDP-mannose, UDP-fructose, UDP-xylose, UDP-rhamnose, UDP-fluorodeoxyglucose, etc. In addition, nucleotide sugars can also be used in combination to create glycosides that contain multiple types of residues on the same aglycone backbone. Alternative strategies to further improve the solubility and delivery of cannabinoids and other compounds described herein include their glycosylation and then functionalizing the sugar moieties with additional ligands or modifications. Examples of this include sulfation, myristoylation, phosphorylation, acetylation, etc.
  • The endocannabinoid system has recently been the subject of intense research efforts due to its demonstrated role in and impact on a broad range of clinical pathologies. As UGT76G1 has been determined to recognize a broad class of phytocannabinoids, it was hypothesized that the same enzyme would also recognize and glycosylate endocannabinoids, which are the endogenous signaling molecules recognized by the cannabinoid receptors in Humans. Upon testing a sample of four prototypic endocannabinoids including arachidonoylethanolamide (anandamide, AEA), 2-arachidonoylethanolamide (2-AG), 1-arachidonoylethanolamide (1-AG), and docosahexaenoyl ethanolamide (DHEA, synaptamide), it was found that UGT76G1 effectively glycosylated each endocannabinoid (Tables 5-8, FIGS. 9-12). Glycosylation of endocannabinoids enables the creation of endocannabinoid-glycosides and other fatty acid neurotransmitter-glycosides, representing a new method of targeted delivery of endocannabinoids.
  • As endocannabinoids such as AEA, 2-AG, 1-AG, and synaptamide are glycosylated by UGT76G1, it is hypothesized that similar endocannabinoids will also be suitable substrates for glycosylation by UGT76G1. Other endocannabinoid candidates that are likely to be glycosylated by UGT76G1 include oleoyl ethanolamide (OEA), eicsapentaenoyl ethanolamide, prostaglandin ethanolamide, docosahexaenoyl ethanolamide, linolenoyl ethanolamide, 5(Z),8(Z),11(Z)-eicosatrienoic acid ethanolamide (mead acid ethanolamide), heptadecanoul ethanolamide, stearoyl ethanolamide, docosaenoyl ethanolamide, nervonoyl ethanolamide, tricosanoyl ethanolamide, lignoceroyl ethanolamide, myristoyl ethanolamide, pentadecanoyl ethanolamide, palmitoleoyl ethanolamide, docosahexaenoic acid (DHA), and similar compounds. These glycolipids may have a wide range of commercial uses, ranging from pharmaceutical use as a novel endocannabinoid drug with improved solubility and pharmacokinetic properties, to use as an antibacterial agent, to use as a detergent similar to other glycolipids, etc.
  • It has been characterized that AEA and CBD are full agonists of the toll-like vanilloid receptor type 1 (TRPV1), which is the receptor for capsaicin. In addition, other cannabinoids and botanical extracts, including but not limited to CBD, CBN, cannabigerol (CBG), and various propyl homologues of CBD, THC, and CBG have been demonstrated to bind and have activity towards transient receptor potential channels (TRPs) (De Petrocellis 2011). This includes stimulating and desensitizing TRPV1, as well as TRPA1, TRPV2, and also antagonism of TRPM8. Although stimulation of TRPV1 leads to vasodilation and inflammation, capsaicin and its analogues act to desensitize the receptors to stimulants, and provide potent anti-inflammatory effects (Bisogno 2001). Analogous effects may occur with TRPA1 in addition to other TRPs. For CBD, this may occur at concentrations that are lower than what is required for binding of cannabinoid receptors, and at concentrations that are within the range of those typically attained in human clinical testing and use. In addition to acting as a direct agonist of the TRPV1 receptor, CBD has been shown to inhibit fatty acid amide hydroxylase (FAAH), the enzyme responsible for facilitating the metabolism of the endocannabinoid anandamide (Watanabe, 1998; DE e Petrocellis 2010). Given that these phytocannabinoids act as ligands of diverse TRPs, it was postulated that UGT76G1 would be capable of glycosylating many different ligands of the same TRPs, including TRPM8, TRPV2, TRPA1, and TRPV1. Capsaicin is capable of contorting into a CBD-like structure (Bisogno 2001), therefore it was postulated that capsaicin was likely to be a suitable substrate for glycosylation by UGT76G1. To this end, it was shown that UGT76G1 is capable of glycosylating the vanilloid moiety of capsaicin in a structurally identical way to PaGT3 from Phytolacca americana (Noguchi 2009). As the glycosylated structure of capsaicin is the vanilloid head, it was further hypothesized that UGT76G1 would be capable of glycosylation of the minimal vanilloid, i.e., vanillin, as well as many analogues. Consistent with this hypothesis, through HPLC analysis it was determined that UGT76G1 created multiple glycoside products of vanillin (FIG. 14, Table 10). Seeking to test the ability of UGT76G1 to glycosylate vanilloids more broadly, curcumin, the well characterized vanilloid found in turmeric spice, isolated from the ginger Curcuma longa was applied as a substrate in the glycosylation reaction. Consistent with the glycosylation of vanillin, UGT76G1 effectively glycosylated curcumin, creating multiple glycoside product peaks, suggesting a bifunctional recognition and glycosylation by UGT76G1 similar to that seen with CBD and steviol glycosides (FIGS. 15A & 15B, Table 11).
  • Cannabinoid glycosides may also have direct bioactive and therapeutic effects, beyond their utility a prodrug for their aglycone form. Quercetin is an antioxidant flavonoid that is ubiquitous in vegetables and often present both in its aglyone and glycosylated forms. It has been demonstrated through in vitro studies that quercetin glucuronides act as a bioactive agent as well as a precursor molecule to aglycone quercetin (Terao 2011). In many cases, including with glycosides that exert antibacterial and antitumor effects, the glycosidic residues are crucial to activity (Kren & Rezanka 2008).
  • Glycosides have also been demonstrated to receive facilitated transport across the blood brain barrier (BBB) by the glucose transporter GLUT1. A prime example is the glycoside of ibuprofen achieving a significant increase of ibuprofen aglycone concentration in the brain (Chen 2009). Similar to these glycosides, glycosides of cannabinoids and other compounds described herein may benefit from enhanced facilitated transport across the BBB or other barriers. Glucose transporters are a wide group of membrane proteins encoded by the human genome and that are found not only in the BBB but across many different cells and tissues, including brain, erythrocytes, fat, muscle, kidney, liver, intestine, and pancreas, so glycosylation will be tailored to provide site-specific delivery to any of these tissues. Accordingly, in one embodiment, there is provided a method for facilitating the transport of a cannabinoid drug across the blood brain barrier of a subject comprising administering to the subject a cannabinoid glycoside prodrug in accordance with the present invention.
  • Delivery of cannabinoids and cannabidiol to the brain may be especially useful because of oligodendrocyte protective (oligoprotective) and general neuroprotective effects. It has been demonstrated that cannabinoid signaling is involved with both oligodendrocyte differentiation (Gomez 2010) and that cannabinoids promote oligodendrocyte progenitor survival (Molina-Holgado 2002). Drug formulations that include cannabidiol as a major ingredient have been approved to treat muscle spasticity and pain from multiple sclerosis, a neurodegenerative disorder that causes loss of myelin and oligodendrocyte progenitor cells. The effects of cannabidiol have been demonstrated to mediate oligoprotective effects through attenuation of endoplasmic reticulum stress pathways (Mecha 2012). Cannabidiol has also been studied extensively for its antipsychotic effects, however the exact role in protection of oligodendroctyes and promotion of remyelination has not yet been described (Zuardi 2012). Despite the correlation between the clinical symptoms of psychosis with neuropathological analysis that indicates dysmyelination is involved, the role of dysmyelination as a driver or cause of schizophrenia and other psychoses remains controversial (Mighdoll 2015). Remyelination has also been described as potentially useful for treatment of Alzheimer's disease and other forms of dementia (Bartzokis 2004). Therefore, delivery of cannabinoids to the brain may be especially useful for its established neuroprotective and oligoprotective effects. Cannabinoid glycoside drug formulations co-administered in combination with other agents that influence other aspects of repair or regeneration, such as oligodendrocyte progenitor differentiation or remyelination, may also prove to be beneficial. This includes compounds such as anti-LINGO-1 monoclonal antibodies, guanabenz, sephin1, benzatropine, clemastine, polyunsaturated fatty acids, etc.
  • In the course of the present work, it was discovered that UGT76G1, Os03g0702000 and cyclodextrin glucanotransferase (CGTase) were capable of primary, secondary and tertiary glycosylations of steviol glycosides and aglycone products of diverse chemical structure, including cannabinoids, endocannabinoids, vanillin, curcumin, and capsaicin.
  • In the screening and analysis methods described by Dewitte 2016, a 50 mm HPLC separation column combined with a high solvent flow rate was used limiting the separation and overall detection of glycoside products. Thus, the interpretation of the glycosylation reaction products for many compounds is speculative, yet still reinforces the significance of the present finding that UGT76G1 has broad substrate specificity. Clearly, the work described herein demonstrate that UGT76G1 can glycosylate not only steviol glycosides, but other forms of glycosides, and novel aglycone compounds such as cannabidiol as well. Internal studies that used an improved separation methodology involving a 150 mm length C18 column coupled with a low solvent flowrate also enabled the clear detection of secondary and tertiary glycosides. These compounds were unable to be detected by the methods described in Dewitte 2016, and provide additional verification of the ability of UGT76G1 to not only glycosylate compounds with diverse chemical structures, but also to perform multiple higher order glycosylations on glycosides of these same compounds.
  • The reactions described herein take place in vitro using recombinant enzymes and all necessary cofactors, and the expression of UGT76G1 enzyme within the cells of a Cannabis plant is possible for the in vivo biotransformation of cannabinoids prior to extraction of cannabinoids from plant tissue. As UGT76G1 is an enzyme from the plant Stevia rebaudiana, it will be compatible with expression in the genus Cannabis. The ideal strategy for expression of UGT76G1 within the Cannabis plant is to genetically engineer the UGT76G1 open reading frame under a promoter element that is specific for the same tissue that cannabinoids are produced in, namely the secretory trichomes of the plant. Suitable promoter elements include the promoter for the cytosolic O-acetylserine(thiol)lyase (OASA1) enzyme from Arabidopsis thaliana (Gutierrez-Alcala 2005). Candidates for transformation with UGT76G1 include Cannabis sativa, Cannabis indica, and Cannabis ruderalis. A similar approach may be used with UGT76G1 and similar enzymes for in planta production of glycosylated secondary metabolites within many other different plant species, and may be especially useful when plant species already produce large quantities of the desired aglycone product or known enzyme substrate.
  • In the course of performing phytocannabinoid glycosylation reactions CBD and THC displayed noticeable antimicrobial activity, even preventing large-scale reaction mixtures from becoming contaminated after failure of the sterile filter apparatus. Prior pilot-scale glycosylation reaction utilizing steviol glycosides as substrates during enzymatic processing were quite susceptible to infection in the absence of strict sanitation techniques. CBD and THC pilot-scale reactions remained aseptic for over a week in the same reaction vessels with very limited ongoing maintenance or care. To this end, the use of the aglycone cannabinoids and their respective glycosides is proposed as efficient antimicrobial agents. Accordingly, in one embodiment, there is provided an antimicrobial agent comprising an effective amount of a cannabinoid glycoside prodrug in accordance with the present invention.
  • Similarly, upon the production of large quantities of cannabinoid-glycosides and formulation in aqueous solutions, it was observed that multiple cannabinoid-glycosides in water had foaming properties similar to detergents. This is consistent with other glycoside detergents like 8-octylglycoside, 8-octylthioglycoside, and similar, and establishes a potential use for cannabinoid-glycosides as a detergent. Accordingly, in one embodiment, there is provided a detersive agent comprising an effective amount of a cannabinoid glycoside prodrug in accordance with the present invention.
  • Nucleic Acids
  • The present invention provides for nucleic acids comprising nucleotide sequences encoding a glycosyltransferase. The glycosyltransferases of the present invention are capable of primary, secondary, tertiary glycosylations or a combination thereof. In certain embodiments, the glycosyltransferases are capable of primary, secondary and tertiary glycosylations. In other embodiments, the glycosyltransferases are capable of secondary and tertiary glycosylations. In certain embodiments, the nucleic acids encode a glucosyltransferase, including but not limited to a UDP-glucosyltransferase. The glucosyltransferases include but are not limited to a Stevia rebaudiana UDP-glucosyltransferase, such as UGT76G1 or UGT74G1 or an Oryza sativa glucosyltrasferase, such as Os03g0702000. In other embodiments, the invention provides for nucleic acids comprising nucleotide sequences encoding a cyclodextrin glucanotransferase. Also provided are nucleic acids comprising nucleotide sequences that encode a sucrose synthase.
  • Nucleic acids include, but are not limited to, genomic DNA, cDNA, RNA, fragments and modified versions, including but not limited to codon optimized versions thereof. For example, the nucleotide sequences may be codon optimized for expression in Pichia pastoris or E. coli. The nucleic acids may include the coding sequence of the glycosyltransferase or sucrose synthase, in isolation, in combination with additional coding sequences (e.g., including but not limited to a purification tag).
  • In certain embodiments, the nucleic acid comprises a sequence encoding UGT76G1 or UGT76G1-like glucosyltransferase. UGT76G1-like glucosyltransferase include for example, other members of the UGT76G1 clade such as UGT76G2 or UGT76H1. In certain embodiments, the nucleic acid comprises a sequence encoding an UGT76G1 glucosyltransferase having the amino acid sequence as set forth in any one of SEQ ID NOs:1, 3, 5 and 7 and listed below or fragments and variants thereof.
  • SEQ ID NO: 1
    (UGT76G1 (native protein sequence))
    MENKTETTVRRRRRIILFPVPFQGHINPILQLANVLYSKGFSITIFHTNF
    NKPKTSNYPHFTFRFILDNDPQDERISNLPTHGPLAGMRIPIINEHGADE
    LRRELELLMLASEEDEEVSCLITDALWYFAQSVADSLNLRRLVLMTSSLF
    NFHAHVSLPQFDELGYLDPDDKTRLEEQASGFPMLKVKDIKSAYSNWQIL
    KEILGKMIKQTKASSGVIWNSFKELEESELETVIREIPAPSFLIPLPKHL
    TASSSSLLDHDRTVFQWLDQQPPSSVLYVSFGSTSEVDEKDFLEIARGLV
    DSKQSFLWVVRPGFVKGSTWVEPLPDGFLGERGRIVKWVPQQEVLAHGAI
    GAFWTHSGWNSTLESVCEGVPMIFSDFGLDQPLNARYMSDVLKVGVYLEN
    GWERGEIANAIRRVMVDEEGEYIRQNARVLKQKADVSLMKGGSSYESLES
    LVSYISSL
    SEQ ID NO: 3
    (UGT76G1 with a 6x Histidine tag at the N-
    terminus)
    MHHHHHHGSGENKTETTVRRRRRIILFPVPFQGHINPILQLANVLYSKGF
    SITIFHTNFNKPKTSNYPHFTFRFILDNDPQDERISNLPTHGPLAGMRIP
    IINEHGADELRRELELLMLASEEDEEVSCLITDALWYFAQSVADSLNLRR
    LVLMTSSLFNFHAHVSLPQFDELGYLDPDDKTRLEEQASGFPMLKVKDIK
    SAYSNWQILKEILGKMIKQTKASSGVIWNSFKELEESELETVIREIPAPS
    FLIPLPKHLTASSSSLLDHDRTVFQWLDQQPPSSVLYVSFGSTSEVDEKD
    FLEIARGLVDSKQSFLWVVRPGFVKGSTWVEPLPDGFLGERGRIVKWVPQ
    QEVLAHGAIGAFWTHSGWNSTLESVCEGVPMIFSDFGLDQPLNARYMSDV
    LKVGVYLENGWERGEIANAIRRVMVDEEGEYIRQNARVLKQKADVSLMKG
    GSSYESLESLVSYISSL
    SEQ ID NO: 5
    (UGT76G1 with a 6x Histidine-Glutamine tag at the
    N-terminus)
    MHQHQHQSGSMENKTETTVRRRRRIILFPVPFQGHINPILQLANVLYSKG
    FSITIFHTNFNKPKTSNYPHFTFRFILDNDPQDERISNLPTHGPLAGMRI
    PIINEHGADELRRELELLMLASEEDEEVSCLITDALWYFAQSVADSLNLR
    RLVLMTSSLFNFHAHVSLPQFDELGYLDPDDKTRLEEQASGFPMLKVKDI
    KSAYSNWQILKEILGKMIKQTKASSGVIWNSFKELEESELETVIREIPAP
    SFLIPLPKHLTASSSSLLDHDRTVFQWLDQQPPSSVLYVSFGSTSEVDEK
    DFLEIARGLVDSKQSFLWVVRPGFVKGSTWVEPLPDGFLGERGRIVKWVP
    QQEVLAHGAIGAFWTHSGWNSTLESVCEGVPMIFSDFGLDQPLNARYMSD
    VLKVGVYLENGWERGEIANAIRRVMVDEEGEYIRQNARVLKQKADVSLMK
    GGSSYESLESLVSYISSL
    SEQ ID NO: 7
    MENKTETTVRRRRRIILFPVPFQGHINPILQLANVLYSKGFSITIFHTNF
    NKPKTSNYPHFTFRFILDNDPQDERISNLPTHGPLAGMRIPIINEHGADE
    LRRELELLMLASEEDEEVSCLITDALWYFAQSVADSLNLRRLVLMTSSLF
    NFHAHVSLPQFDELGYLDPDDKTRLEEQASGFPMLKVKDIKSAYSNWQIL
    KEILGKMIKQTRASSGVIWNSFKELEESELETVIREIPAPSFLIPLPKHL
    TASSSSLLDHDRTVFQWLDQQPPSSVLYVSFGSTSEVDEKDFLEIARGLV
    DSKQSFLWVVRPGFVKGSTWVEPLPDGFLGERGRIVKWVPQQEVLAHGAI
    GAFWTHSGWNSTLESVCEGVPMIFSDFGLDQPLNARYMSDVLKVGVYLEN
    GWERGEIANAIRRVMVDEEGEYIRQNARVLKQKADVSLMKGGSSYESLES
    LVSYISSLGSHHHHHH
  • In certain embodiments, the nucleic acid comprises a sequence encoding UGT76G1 having the amino acid sequence as set forth in AAR06912.1. In certain embodiments, the nucleic acid molecule comprises a sequence encoding UGT76G1 glucosyltransferase and comprising the nucleotide sequence as set forth in any one of SEQ ID NOs: 2, 4, 6 and 8 and listed below, or fragments and variants thereof.
  • SEQ ID NO: 2
    (UGT76G1 native nucleic acid sequence)
    ATGGAAAATAAAACGGAGACCACCGTTCGCCGGCGCCGGAGAATAATATTATTCCCGGTA
    CCATTTCAAGGCCACATTAACCCAATTCTTCAGCTAGCCAATGTGTTGTACTCTAAAGGATT
    CAGTATCACCATCTTTCACACCAACTTCAACAAACCCAAAACATCTAATTACCCTCACTTCA
    CTTTCAGATTCATCCTCGACAACGACCCACAAGACGAACGCATTTCCAATCTACCGACTCA
    TGGTCCGCTCGCTGGTATGCGGATTCCGATTATCAACGAACACGGAGCTGACGAATTACG
    ACGCGAACTGGAACTGTTGATGTTAGCTTCTGAAGAAGATGAAGAGGTATCGTGTTTAATC
    ACGGATGCTCTTTGGTACTTCGCGCAATCTGTTGCTGACAGTCTTAACCTCCGACGGCTTG
    TTTTGATGACAAGCAGCTTGTTTAATTTTCATGCACATGTTTCACTTCCTCAGTTTGATGAG
    CTTGGTTACCTCGATCCTGATGACAAAACCCGTTTGGAAGAACAAGCGAGTGGGTTTCCTA
    TGCTAAAAGTGAAAGACATCAAGTCTGCGTATTCGAACTGGCAAATACTCAAAGAGATATT
    AGGGAAGATGATAAAACAAACAAGAGCATCTTCAGGAGTCATCTGGAACTCATTTAAGGAA
    CTCGAAGAGTCTGAGCTCGAAACTGTTATCCGTGAGATCCCGGCTCCAAGTTTCTTGATAC
    CACTCCCCAAGCATTTGACAGCCTCTTCCAGCAGCTTACTAGACCACGATCGAACCGTTTT
    TCAATGGTTAGACCAACAACCGCCAAGTTCGGTACTGTATGTTAGTTTTGGTAGTACTAGT
    GAAGTGGATGAGAAAGATTTCTTGGAAATAGCTCGTGGGTTGGTTGATAGCAAGCAGTCG
    TTTTTATGGGTGGTTCGACCTGGGTTTGTCAAGGGTTCGACGTGGGTCGAACCGTTGCCA
    GATGGGTTCTTGGGTGAAAGAGGACGTATTGTGAAATGGGTTCCACAGCAAGAAGTGCTA
    GCTCATGGAGCAATAGGCGCATTCTGGACTCATAGCGGATGGAACTCTACGTTGGAAAGC
    GTTTGTGAAGGTGTTCCTATGATTTTCTCGGATTTTGGGCTCGATCAACCGTTGAATGCTA
    GATACATGAGTGATGTTTTGAAGGTAGGGGTGTATTTGGAAAATGGGTGGGAAAGAGGAG
    AGATAGCAAATGCAATAAGAAGAGTTATGGTGGATGAAGAAGGAGAATACATTAGACAGAA
    TGCAAGAGTTTTGAAACAAAAGGCAGATGTTTCTTTGATGAAGGGTGGTTCGTCTTACGAA
    TCATTAGAGTCTCTAGTTTCTTACATTTCATCGTTGTAA
    SEQ ID NO: 4
    (Sequence encoding SEQ ID NO: 3 codon optimized for expression
    in Pichia pastoris)
    ATGCACCACCATCACCACCATGGTTCTGGTGAAAACAAAACTGAAACTACTGTTAGAAGAA
    GAAGAAGAATCATTTTGTTTCCAGTACCATTTCAAGGCCATATCAATCCAATTCTTCAATTG
    GCCAATGTTTTGTACTCCAAAGGATTCTCCATCACCATTTTTCACACCAATTTCAACAAACC
    AAAGACTTCCAACTATCCTCACTTCACTTTCAGATTTATTTTGGATAATGATCCTCAAGATG
    AAAGAATTTCCAATCTTCCGACTCATGGTCCTTTGGCTGGTATGAGAATTCCAATCATCAAT
    GAACATGGTGCTGATGAATTAAGAAGAGAATTGGAACTTTTGATGTTGGCTTCTGAAGAAG
    ATGAAGAAGTTTCATGTTTAATCACTGATGCTTTATGGTATTTTGCTCAATCTGTTGCTGAT
    TCTTTGAATTTGCGACGGTTGGTTTTGATGACTTCTTCTTTGTTCAACTTTCATGCTCATGT
    TTCTTTACCTCAGTTTGATGAACTTGGATATTTGGATCCAGATGACAAAACTAGATTGGAAG
    AACAAGCTAGTGGGTTTCCTATGTTGAAAGTCAAAGATATCAAATCTGCTTACTCCAACTG
    GCAAATTCTCAAAGAAATTTTGGGAAAAATGATCAAACAAACAAAAGCTTCTTCTGGAGTCA
    TTTGGAACTCATTCAAAGAATTGGAAGAATCTGAATTGGAAACTGTTATTAGAGAAATTCCT
    GCTCCAAGTTTTTTGATTCCTTTGCCAAAACATTTGACTGCTTCTTCTTCTTCTTTATTGGAT
    CACGATAGAACTGTTTTTCAATGGTTAGATCAACAACCTCCATCTTCTGTTTTGTATGTTAG
    TTTTGGATCTACTTCTGAAGTTGATGAAAAAGATTTTTTGGAAATTGCTAGAGGTTTGGTTG
    ATTCCAAACAAAGTTTTTTATGGGTTGTTAGACCAGGATTTGTCAAAGGATCTACTTGGGTC
    GAACCTTTGCCAGATGGATTTTTGGGAGAAAGAGGAAGAATTGTCAAATGGGTTCCACAG
    CAAGAAGTTTTGGCTCATGGTGCTATTGGTGCTTTTTGGACTCATTCTGGATGGAACTCTA
    CTTTGGAATCTGTTTGTGAAGGTGTTCCAATGATTTTTTCTGATTTTGGTTTGGATCAACCA
    TTGAATGCTAGATACATGTCTGATGTTTTGAAAGTTGGTGTTTATTTGGAAAATGGGTGGG
    AAAGAGGTGAAATTGCCAATGCTATTAGAAGAGTCATGGTTGATGAAGAAGGAGAATACAT
    TAGACAAAATGCTAGAGTTTTGAAACAAAAAGCTGATGTTTCTTTGATGAAGGGTGGATCTT
    CTTATGAATCTTTGGAATCTTTGGTTTCTTACATTTCTTCTCTTTAA
    SEQ ID NO: 6
    (Sequence encoding SEQ ID NO: 5 codon optimized for expression
    in Pichia pastoris)
    ATGCATCAACATCAACACCAATCTGGATCTATGGAGAACAAGACCGAGACTACAGTTAGAA
    GAAGAAGAAGAATAATCCTGTTTCCAGTACCATTCCAAGGACACATCAACCCAATCTTGCA
    GTTAGCAAATGTACTTTATTCTAAAGGCTTTAGTATTACGATTTTTCACACTAATTTTAATAA
    GCCAAAAACATCCAATTACCCTCACTTCACATTCAGATTTATCTTGGATAACGATCCTCAAG
    ATGAACGTATCTCCAACCTGCCAACACATGGACCATTGGCCGGTATGCGTATTCCTATAAT
    CAACGAGCATGGTGCTGATGAGCTTAGACGTGAACTGGAACTGTTGATGCTGGCATCGGA
    GGAAGATGAAGAGGTTAGTTGCTTGATAACGGATGCCCTCTGGTATTTCGCACAATCAGTC
    GCTGACTCCTTGAACCTTAGGAGATTGGTATTGATGACTAGTTCGTTGTTCAACTTCCATG
    CCCATGTTTCTTTGCCTCAATTTGATGAGCTGGGTTATTTGGATCCTGACGATAAGACTCG
    TTTAGAAGAACAGGCGTCAGGCTTCCCCATGTTAAAGGTTAAAGATATTAAGTCCGCCTAT
    TCTAACTGGCAAATTCTCAAAGAGATTCTAGGGAAAATGATTAAACAAACCAAGGCCTCTTC
    AGGAGTAATCTGGAACAGTTTCAAAGAACTAGAAGAATCCGAGTTGGAAACTGTTATTCGT
    GAAATCCCTGCTCCATCTTTCCTTATCCCATTACCAAAGCACCTCACTGCCTCCTCTAGTTC
    TCTTCTGGACCATGATAGAACAGTCTTTCAGTGGCTCGATCAGCAACCTCCATCTTCTGTC
    TTGTACGTTAGTTTTGGTTCCACCTCGGAAGTAGATGAAAAAGACTTTCTGGAAATTGCTC
    GAGGACTAGTTGACTCCAAGCAATCCTTTCTGTGGGTTGTTAGACCTGGATTCGTAAAAGG
    ATCCACCTGGGTAGAACCCCTCCCAGATGGATTTTTGGGCGAAAGGGGAAGAATTGTTAA
    ATGGGTGCCTCAACAAGAAGTTTTAGCTCATGGGGCCATTGGAGCTTTTTGGACTCATAGT
    GGATGGAATTCTACCTTAGAATCTGTTTGTGAAGGAGTTCCAATGATTTTTTCTGATTTTGG
    ATTGGATCAGCCTCTTAATGCCAGATATATGTCCGATGTCCTCAAGGTCGGAGTGTACCTG
    GAAAATGGTTGGGAGAGAGGTGAGATTGCAAATGCTATACGTAGAGTCATGGTTGATGAA
    GAGGGCGAGTATATTAGACAAAACGCTAGAGTGCTAAAGCAGAAGGCCGATGTTTCCCTT
    ATGAAGGGGGGAAGTTCATATGAGAGTTTGGAATCCCTAGTGTCCTACATTTCTTCGCTAT
    AA
    SEQ ID NO: 8
    (Sequence encoding SEQ ID NO: 7 codon optimized for expression
    in Escherichia coli)
    ATGGAAAATAAAACCGAAACCACCGTCCGTCGCCGTCGTCGTATCATTCTGTTCCCGGTCC
    CGTTCCAAGGTCACATCAACCCGATTCTGCAGCTGGCCAACGTGCTGTATAGCAAAGGTTT
    CTCTATCACCATCTTCCATACGAACTTCAACAAACCGAAAACCTCTAACTACCCGCACTTTA
    CGTTCCGTTTTATTCTGGATAACGACCCGCAGGATGAACGCATCAGTAATCTGCCGACCCA
    TGGTCCGCTGGCGGGTATGCGTATTCCGATTATCAACGAACACGGCGCAGATGAACTGCG
    TCGCGAACTGGAACTGCTGATGCTGGCCTCTGAAGAAGATGAAGAAGTTAGTTGCCTGAT
    CACCGACGCACTGTGGTATTTTGCCCAGAGTGTTGCAGATTCCCTGAACCTGCGTCGCCT
    GGTCCTGATGACGAGCTCTCTGTTCAATTTTCATGCCCACGTTTCCCTGCCGCAGTTCGAT
    GAACTGGGTTATCTGGACCCGGATGACAAAACCCGCCTGGAAGAACAAGCTTCAGGCTTT
    CCGATGCTGAAAGTCAAAGATATTAAAAGTGCGTACTCCAACTGGCAGATTCTGAAAGAAA
    TCCTGGGTAAAATGATCAAACAAACCCGTGCAAGTTCCGGCGTCATCTGGAATTCCTTCAA
    AGAACTGGAAGAATCAGAACTGGAAACGGTGATTCGCGAAATCCCGGCTCCGTCTTTTCT
    GATTCCGCTGCCGAAACATCTGACCGCGTCATCGAGCTCTCTGCTGGATCACGACCGTAC
    GGTGTTTCAGTGGCTGGATCAGCAACCGCCGAGTTCCGTGCTGTACGTTAGCTTCGGTAG
    CACCTCTGAAGTGGATGAAAAAGACTTTCTGGAAATCGCTCGTGGCCTGGTTGATTCAAAA
    CAATCGTTCCTGTGGGTGGTTCGCCCGGGTTTTGTGAAAGGCAGCACGTGGGTTGAACC
    GCTGCCGGATGGCTTCCTGGGTGAACGTGGTCGCATTGTCAAATGGGTGCCGCAGCAAG
    AAGTGCTGGCACATGGTGCTATCGGCGCGTTTTGGACCCACTCAGGTTGGAACTCGACGC
    TGGAAAGCGTTTGTGAAGGTGTCCCGATGATTTTCTCGGATTTTGGCCTGGACCAGCCGC
    TGAATGCACGTTATATGAGCGATGTTCTGAAAGTCGGTGTGTACCTGGAAAACGGTTGGG
    AACGCGGCGAAATTGCGAATGCCATCCGTCGCGTTATGGTCGATGAAGAAGGCGAATATA
    TCCGTCAGAATGCTCGCGTCCTGAAACAAAAAGCGGACGTTAGTCTGATGAAAGGCGGTT
    CATCGTACGAATCCCTGGAATCACTGGTCTCCTACATTTCTTCTCTGGGCTCGCATCATCA
    TCATCATCATTAA
  • In certain embodiments, the nucleic acid molecule encodes an UGT76G1 glucosyltransferase and comprises the nucleotide sequence as set forth in GenBank Accession number AY345974.1 or a variant or fragment thereof.
  • In certain embodiments, the nucleic acid comprises a sequence encoding UGT76G2 glucosyltransferase. In specific embodiments, the nucleic acid comprises a sequence encoding UGT76G2 glucosyltransferase having the amino acid sequence as set forth in SEQ ID NO:27 and listed below or variants and fragments thereof.
  • SEQ ID NO: 27
    MENKTETTVRRRRRIILFPVPVQGHINPILQLANVLYSKGFSITIFHTNF
    NKPKTSNYPHFTFRFILDNDPQDVRISNLPTHGPLTVMRILIINEHGADE
    LQRELELLMLASEEDGEVSCLITDQIWYFTQSVADSLNLRRLVLMTSSLF
    NFHAHVSLPQFDELGYLDPDDKTRLEEQASGFPMLKVKDIKCGFSMWKQG
    KEIFENITKQTKASSGVIWNSFKELEESELETVIREIPAPSFLIPLPKHL
    TASSSSLLDHDRTVFPWLDQQPSRSVLYVSFGSATEVDAKDFLEIARGLV
    DSKQSFLWVVRPGFVKGSTWVEPLPDGFLGERGRIVKWVPQQEVLAHGAI
    GAFWTHSGWNSTLESVCEGVPMIFSAFAFDQPLNARYMSDVLKVGVYLEN
    GWERGEIANAIRRVMVDEEGGYIRQNASVLKQKADVSLMKGGSSYESLES
    LVAYISSL
  • In specific embodiments, the nucleic acid comprises a sequence encoding UGT76G2 glucosyltransferase and having the nucleic acid sequence as set forth in SEQ ID NO:28 and listed below or variants and fragments thereof.
  • SEQ ID NO: 28
    ATGGAAAATAAAACGGAGACCACCGTTCGCCGGCGCCGGAGAATAATATT
    ATTCCCGGTACCAGTTCAAGGCCACATTAACCCAATTCTTCAGCTAGCCA
    ATGTGTTGTACTCCAAAGGATTCAGTATCACCATCTTTCACACCAACTTC
    AACAAACCCAAAACATCTAATTACCCTCACTTCACTTTCAGATTCATCCT
    CGACAACGACCCACAAGACGTACGCATTTCCAATCTACCGACTCATGGTC
    CGCTCACTGTTATGCGGATTCTGATTATCAACGAACACGGAGCTGACGAA
    TTACAACGCGAACTGGAACTGTTGATGTTAGCTTCTGAAGAAGATGGAGA
    GGTATCGTGTTTAATCACCGATCAGATTTGGTACTTCACGCAATCTGTTG
    CTGACAGTCTTAACCTCCGACGGCTTGTTTTGATGACAAGCAGCTTGTTT
    AATTTTCATGCACATGTTTCACTTCCTCAGTTTGATGAGCTTGGTTACCT
    CGATCCTGATGACAAAACCCGTTTGGAAGAACAAGCGAGTGGGTTTCCTA
    TGCTGAAAGTGAAAGATATCAAGTGTGGTTTTTCGATGTGGAAACAAGGC
    AAAGAGATATTCGAGAACATTACGAAACAAACAAAAGCATCTTCAGGAGT
    CATCTGGAACTCATTTAAGGAACTCGAAGAGTCTGAGCTCGAAACTGTTA
    TCCGTGAGATCCCGGCTCCAAGTTTCTTGATACCACTCCCCAAGCATTTG
    ACAGCCTCTTCCAGCAGCTTACTAGACCACGATCGAACCGTTTTTCCATG
    GTTAGACCAACAACCGTCACGTTCGGTACTGTATGTTAGTTTTGGTAGTG
    CTACTGAAGTGGATGCGAAAGATTTCTTGGAAATAGCTCGTGGGTTGGTT
    GATAGCAAGCAGTCGTTTTTATGGGTGGTTCGACCTGGTTTTGTCAAGGG
    TTCGACGTGGGTCGAACCGTTGCCAGATGGGTTCTTGGGTGAAAGAGGAC
    GTATTGTGAAATGGGTTCCGCAGCAAGAAGTGCTAGCTCATGGAGCAATA
    GGCGCATTCTGGACTCATAGCGGATGGAACTCTACGTTGGAAAGCGTTTG
    TGAAGGTGTTCCTATGATTTTCTCGGCTTTTGCGTTCGATCAACCGTTGA
    ATGCTAGATACATGAGTGATGTTTTGAAGGTAGGGGTGTATTTGGAAAAT
    GGGTGGGAAAGAGGAGAGATAGCAAATGCAATAAGAAGAGTTATGGTGGA
    TGAAGAAGGAGGATACATTAGACAGAATGCAAGTGTTTTGAAACAAAAGG
    CAGATGTTTCTTTGATGAAGGGTGGTTCGTCTTACGAATCATTAGAGTCT
    CTAGTTGCTTACATTTCATCGTTGTAA
  • In certain embodiments, the nucleic acid comprises a sequence encoding UGT76H1 glucosyltransferase. In specific embodiments, the nucleic acid comprises a sequence encoding UGT76H1 glucosyltransferase having the amino acid sequence as set forth in SEQ ID NO:29 and listed below or variants and fragments thereof.
  • SEQ ID NO: 29
    MLQLATYLHSQGISITIAQYPNFNSPDSSNHPELTFLPLSSGNLSVADIS
    GGFFKFIQTLNHNCKPHFREYLVQNMSSDDKESIVIIRDNLMFFAGEIAG
    ELGLPSIILRGSNAVMLTASDIIPQLHQEGRFPPPDSLLQETIPELVPFR
    YKDLPFIGYPIHQTLEFSITMMTPKSPASAILINTLEFLEQSALTQIRDH
    YKVPVFTIGPLHKIVTTRSTSILEEDTSCINWLDKQSPKSVVYVSLGSLA
    KLDEKVASEMACGLAMSNHKFLWVVRPGMVHGFEWVEFLPDSLVGEMKAR
    GLIVKWAPQTTVLAHNAVGGFWSHCGWNSTIECLAEGVPMMCQPFFADQL
    LNARYVSDVWKTGFEIVIEKGEIACAIKRVLVDEEGEEMRQRAMEIKEKV
    KIAINDGGSSYDSFKDLVAFISSL
  • In specific embodiments, the nucleic acid comprises a sequence encoding UGT76H1 glucosyltransferase and having the nucleic acid sequence as set forth in SEQ ID NO:30 and listed below or variants and fragments thereof.
  • ATGCTTCAGCTTGCAACTTACCTCCATTCTCAAGGGATTTCAATAACCAT
    CGCTCAGTACCCCAACTTCAACTCGCCGGATTCTTCCAACCATCCAGAAC
    TAACCTTCCTCCCACTATCCTCCGGCAACTTATCCGTCGCCGACATCTCC
    GGCGGCTTTTTCAAGTTCATCCAAACTCTTAACCATAACTGCAAACCCCA
    TTTCCGGGAATACCTTGTTCAGAACATGAGTTCTGATGATAAGGAATCAA
    TCGTTATCATCCGTGATAATCTCATGTTTTTCGCCGGAGAAATCGCCGGC
    GAGCTGGGTCTGCCTTCGATCATTTTACGTGGCAGCAATGCTGTCATGTT
    GACTGCTAGCGACATCATCCCTCAACTTCATCAAGAAGGTCGTTTTCCGC
    CACCAGATTCTTTGTTGCAGGAAACAATTCCAGAACTGGTTCCATTCAGA
    TACAAAGATCTACCATTTATTGGCTATCCAATACATCAAACCCTTGAATT
    TAGTATCACCATGATGACCCCCAAATCACCTGCTTCCGCCATTCTTATCA
    ACACCCTCGAATTTCTTGAACAATCGGCATTAACCCAGATCCGTGATCAT
    TACAAAGTTCCAGTTTTTACAATCGGACCATTGCACAAAATAGTCACAAC
    TCGTTCCACTAGCATTCTTGAAGAAGATACAAGTTGCATCAATTGGTTAG
    ATAAACAATCACCCAAATCAGTGGTTTATGTGAGTTTAGGAAGCTTAGCA
    AAGTTGGATGAAAAGGTTGCATCTGAAATGGCATGTGGTTTAGCCATGAG
    TAACCATAAGTTCCTATGGGTGGTTCGACCCGGTATGGTTCATGGGTTTG
    AATGGGTCGAGTTTTTGCCGGATAGTTTGGTGGGTGAAATGAAGGCTAGA
    GGTTTGATTGTGAAATGGGCACCCCAGACGACGGTTTTGGCGCATAACGC
    GGTTGGTGGATTTTGGAGTCATTGCGGTTGGAACTCGACCATAGAATGCT
    TAGCTGAAGGGGTCCCGATGATGTGTCAACCGTTTTTTGCTGATCAGTTG
    TTGAATGCTAGGTATGTGAGTGATGTTTGGAAGACGGGTTTTGAGATTGT
    TATCGAGAAAGGTGAGATTGCGTGCGCGATTAAACGAGTTTTGGTGGATG
    AAGAAGGCGAAGAAATGAGGCAGAGAGCTATGGAGATTAAAGAAAAGGTT
    AAAATTGCAATCAACGATGGTGGTTCTTCTTATGACTCGTTCAAGGACTT
    GGTGGCGTTTATTTCATCACTCTAA
  • In certain embodiments, the nucleic acid comprises a sequence encoding Oryza sativa Os03g0702000 or Os03g0702000-like glucosyltransferase. Os03g0702000-like glucosyltransferase include for example, other members of the UGT91clade such as UGT91D1 or UGT91 D2. In certain embodiments, the nucleic acid comprises a sequence encoding Os03g0702000 glucosyltransferase having the amino acid sequence as set forth in SEQ ID NO: 9 and listed below or a variant or fragment thereof.
  • SEQ ID NO: 9
    MHQHQHQSGSMDSGYSSSYAAAAGMHVVICPWLAFGHLLPCLDLAQRLAS
    RGHRVSFVSTPRNISRLPPVRPALAPLVAFVALPLPRVEGLPDGAESTND
    VPHDRPDMVELHRRAFDGLAAPFSEFLGTACADWVIVDVFHHWAAAAALE
    HKVPCAMMLLGSAHMIASIADRRLERAETESPAAAGQGRPAAAPTFEVAR
    MKLIRTKGSSGMSLAERFSLTLSRSSLVVGRSCVEFEPETVPLLSTLRGK
    PITFLGLMPPLHEGRREDGEDATVRWLDAQPAKSVVYVALGSEVPLGVEK
    VHELALGLELAGTRFLWALRKPTGVSDADLLPAGFEERTRGRGVVATRWV
    PQMSILAHAAVGAFLTHCGWNSTIEGLMFGHPLIMLPIFGDQGPNARLIE
    AKNAGLQVARNDGDGSFDREGVAAAIRAVAVEEESSKVFQAKAKKLQEIV
    ADMACHERYIDGFIQQLRSYKD
  • In certain embodiments, the nucleic acid molecule encodes Os03g0702000 glucosyltransferase and comprises a nucleotide sequence as set forth in SEQ ID NO: 10 and as detailed below or a variant or fragment thereof.
  • SEQ ID NO: 10
    ATGCATCAGCACCAACATCAGAGCGGTTCTATGGACTCCGGCTACTCCTC
    CTCCTACGCCGCCGCCGCCGGGATGCACGTCGTGATCTGCCCGTGGCTCG
    CCTTCGGCCACCTGCTCCCGTGCCTCGACCTCGCCCAGCGCCTCGCGTCG
    CGGGGCCACCGCGTGTCGTTCGTCTCCACGCCGCGGAACATATCCCGCCT
    CCCGCCGGTGCGCCCCGCGCTCGCGCCGCTCGTCGCCTTCGTGGCGCTGC
    CGCTCCCGCGCGTCGAGGGGCTCCCCGACGGCGCCGAGTCCACCAACGAC
    GTCCCCCACGACAGGCCGGACATGGTCGAGCTCCACCGGAGGGCCTTCGA
    CGGGCTCGCCGCGCCCTTCTCGGAGTTCTTGGGCACCGCGTGCGCCGACT
    GGGTCATCGTCGACGTCTTCCACCACTGGGCCGCAGCCGCCGCTCTCGAG
    CACAAGGTGCCATGTGCAATGATGTTGTTGGGCTCTGCACATATGATCGC
    TTCCATAGCAGACAGACGGCTCGAGCGCGCGGAGACAGAGTCGCCTGCGG
    CTGCCGGGCAGGGACGCCCAGCGGCGGCGCCAACGTTCGAGGTGGCGAGG
    ATGAAGTTGATACGAACCAAAGGCTCATCGGGAATGTCCCTCGCCGAGCG
    CTTCTCCTTGACGCTCTCGAGGAGCAGCCTCGTCGTCGGGCGGAGCTGCG
    TGGAGTTCGAGCCGGAGACCGTCCCGCTCCTGTCGACGCTCCGCGGTAAG
    CCTATTACCTTCCTTGGCCTTATGCCGCCGTTGCATGAAGGCCGCCGCGA
    GGACGGCGAGGATGCCACCGTCCGCTGGCTCGACGCGCAGCCGGCCAAGT
    CCGTCGTGTACGTCGCGCTAGGCAGCGAGGTGCCACTGGGAGTGGAGAAG
    GTCCACGAGCTCGCGCTCGGGCTGGAGCTCGCCGGGACGCGCTTCCTCTG
    GGCTCTTAGGAAGCCCACTGGCGTCTCCGACGCCGACCTCCTCCCCGCCG
    GCTTCGAGGAGCGCACGCGCGGCCGCGGCGTCGTGGCGACGAGATGGGTT
    CCTCAGATGAGCATACTGGCGCACGCCGCCGTGGGCGCGTTCCTGACCCA
    CTGCGGCTGGAACTCGACCATCGAGGGGCTCATGTTCGGCCACCCGCTTA
    TCATGCTGCCGATCTTCGGCGACCAGGGACCGAACGCGCGGCTAATCGAG
    GCGAAGAACGCCGGATTGCAGGTGGCAAGAAACGACGGCGATGGATCGTT
    CGACCGAGAAGGCGTCGCGGCGGCGATTCGTGCAGTCGCGGTGGAGGAAG
    AAAGCAGCAAAGTGTTTCAAGCCAAAGCCAAGAAGCTGCAGGAGATCGTC
    GCGGACATGGCCTGCCATGAGAGGTACATCGACGGATTCATTCAGCAATT
    GAGATCTTACAAGGATTGA
  • In certain embodiments, the nucleic acid molecule encodes Os03g0702000 glucosyltransferase and comprises the sequence as set forth in GenBank Accession number XM_015773655 or a variant or fragment thereof.
  • In certain embodiments, the nucleic acid comprises a sequence encoding UGT91 D1 glucosyltransferase. In certain embodiments, the nucleic acid comprises a sequence encoding UGT91 D1 glucosyltransferase having the amino acid sequence as set forth in SEQ ID NO:31 and listed below or a variant or fragment thereof.
  • SEQ ID NO: 31
    MYNVTYHQNSKAMATSDSIVDDRKQLHVATFPWLAFGHILPFLQLSKLIA
    EKGHKVSFLSTTRNIQRLSSHISPLINVVQLTLPRVQELPEDAEATTDVH
    PEDIQYLKKAVDGLQPEVTRFLEQHSPDWIIYDFTHYWLPSIAASLGISR
    AYFCVITPWTIAYLAPSSDAMINDSDGRTTVEDLTTPPKWFPFPTKVCWR
    KHDLARMEPYEAPGISDGYRMGMVFKGSDCLLFKCYHEFGTQWLPLLETL
    HQVPVVPVGLLPPEIPGDEKDETWVSIKKWLDGKQKGSVVYVALGSEALV
    SQTEVVELALGLELSGLPFVWAYRKPKGPAKSDSVELPDGFVERTRDRGL
    VWTSWAPQLRILSHESVCGFLTHCGSGSIVEGLMFGHPLIMLPIFCDQPL
    NARLLEDKQVGIEIPRNEEDGCLTKESVARSLRSVVVENEGEIYKANARA
    LSKIYNDTKVEKEYVSQFVDYLEKNARAVAIDHES
  • In certain embodiments, the nucleic acid molecule encodes UGT91D1 glucosyltransferase and comprises a nucleotide sequence as set forth in SEQ ID NO: 32 and as detailed below or a variant or fragment thereof.
  • SEQ ID NO: 32
    ATGTACAACGTTACTTATCATCAAAATTCAAAAGCAATGGCTACCAGTGA
    CTCCATAGTTGACGACCGTAAGCAGCTTCATGTTGCGACGTTCCCATGGC
    TTGCTTTCGGTCACATCCTCCCTTTCCTTCAGCTTTCGAAATTGATAGCT
    GAAAAGGGTCACAAAGTCTCGTTTCTTTCTACCACCAGAAACATTCAACG
    TCTCTCTTCTCATATCTCGCCACTCATAAATGTTGTTCAACTCACACTTC
    CACGTGTCCAAGAGCTGCCGGAGGATGCAGAGGCGACCACTGACGTCCAC
    CCTGAAGATATTCAATATCTCAAGAAGGCTGTTGATGGTCTTCAACCGGA
    GGTCACCCGGTTTCTAGAACAACACTCTCCGGACTGGATTATTTATGATT
    TTACTCACTACTGGTTGCCATCCATCGCGGCTAGCCTCGGTATCTCACGA
    GCCTACTTCTGCGTCATCACTCCATGGACCATTGCTTATTTGGCACCCTC
    ATCTGACGCCATGATAAATGATTCAGATGGTCGAACCACGGTTGAGGATC
    TCACGACACCGCCCAAGTGGTTTCCCTTTCCGACCAAAGTATGCTGGCGG
    AAGCATGATCTTGCCCGAATGGAGCCTTACGAAGCTCCGGGGATATCTGA
    TGGATACCGTATGGGGATGGTTTTTAAGGGATCTGATTGTTTGCTTTTCA
    AATGTTACCATGAGTTTGGAACTCAATGGCTACCTCTTTTGGAGACACTA
    CACCAAGTACCGGTGGTTCCGGTGGGATTACTGCCGCCGGAAATACCCGG
    AGACGAGAAAGATGAAACATGGGTGTCAATCAAGAAATGGCTCGATGGTA
    AACAAAAAGGCAGTGTGGTGTACGTTGCATTAGGAAGCGAGGCTTTGGTG
    AGCCAAACCGAGGTTGTTGAGTTAGCATTGGGTCTCGAGCTTTCTGGGTT
    GCCATTTGTTTGGGCTTATAGAAAACCAAAAGGTCCCGCGAAGTCAGACT
    CGGTGGAGTTGCCAGACGGGTTCGTGGAACGAACTCGTGACCGTGGGTTG
    GTCTGGACGAGTTGGGCACCTCAGTTACGAATACTGAGCCACGAGTCAGT
    TTGTGGTTTCTTGACTCATTGTGGTTCTGGATCAATTGTGGAAGGGCTAA
    TGTTTGGTCACCCTCTAATCATGCTACCGATTTTTTGTGACCAACCTCTG
    AATGCTCGATTACTGGAGGACAAACAGGTGGGAATCGAGATACCAAGAAA
    TGAGGAAGATGGTTGCTTGACCAAGGAGTCGGTTGCTAGATCACTGAGGT
    CCGTTGTTGTGGAAAACGAAGGGGAGATCTACAAGGCGAACGCGAGGGCG
    CTGAGTAAAATCTATAACGACACTAAGGTGGAAAAAGAATATGTAAGCCA
    ATTCGTAGACTATTTGGAAAAGAATGCGCGTGCGGTTGCCATCGATCATG
    AGAGTTAA
  • In certain embodiments, the nucleic acid comprises a sequence encoding UGT91 D2 glucosyltransferase. In certain embodiments, the nucleic acid comprises a sequence encoding UGT91 D2 glucosyltransferase having the amino acid sequence as set forth in SEQ ID NO: 33 and listed below or a variant or fragment thereof.
  • SEQ ID NO: 33
    MATSDSIVDDRKQLHVATFPWLAFGHILPYLQLSKLIAEKGHKVSFLSTT
    RNIQRLSSHISPLINVVQLTLPRVQELPEDAEATTDVHPEDIPYLKKASD
    GLQPEVTRFLEQHSPDWIIYDYTHYWLPSIAASLGISRAHFSVTTPWAIA
    YMGPSADAMINGSDGRTTVEDLTTPPKWFPFPTKVCWRKHDLARLVPYKA
    PGISDGYRMGLVLKGSDCLLSKCYHEFGTQWLPLLETLHQVPVVPVGLLP
    PEIPGDEKDETWVSIKKWLDGKQKGSVVYVALGSEVLVSQTEVVELALGL
    ELSGLPFVWAYRKPKGPAKSDSVELPDGFVERTRDRGLVWTSWAPQLRIL
    SHESVCGFLTHCGSGSIVEGLMFGHPLIMLPIFGDQPLNARLLEDKQVGI
    EIPRNEEDGCLTKESVARSLRSVVVEKEGEIYKANARELSKIYNDTKVEK
    EYVSQFVDYLEKNARAVAIDHES
  • In certain embodiments, the nucleic acid molecule encodes UGT91D2 glucosyltransferase and comprises a nucleotide sequence as set forth in SEQ ID NO: 34 and as detailed below or a variant or fragment thereof.
  • SEQ ID NO: 34
    ATGGCCACATCTGACTCTATCGTTGATGACAGAAAACAATTGCATGTTGC
    TACTTTCCCATGGTTGGCCTTTGGACACATTCTGCCCTACTTGCAATTGT
    CAAAGCTGATTGCAGAAAAAGGTCATAAGGTGTCCTTTTTGTCTACCACA
    AGAAACATCCAGAGACTAAGTTCTCATATTTCTCCATTGATTAATGTGGT
    TCAGTTGACCTTGCCTAGAGTCCAAGAACTTCCCGAAGACGCAGAAGCTA
    CTACTGATGTTCACCCTGAAGATATCCCATATCTAAAGAAGGCATCTGAT
    GGACTTCAACCAGAAGTAACCAGGTTTTTGGAGCAGCACAGTCCTGACTG
    GATTATCTATGATTATACTCATTACTGGCTTCCATCCATCGCAGCTAGTC
    TAGGCATTTCCAGAGCTCATTTCTCTGTCACTACCCCATGGGCAATTGCA
    TATATGGGTCCTTCTGCTGATGCAATGATCAACGGTTCTGATGGTAGGAC
    CACTGTTGAAGATTTAACTACACCTCCAAAGTGGTTCCCATTTCCTACTA
    AAGTTTGTTGGCGAAAACACGATCTGGCACGTTTGGTCCCATATAAGGCT
    CCAGGTATCTCCGATGGATATCGAATGGGTCTGGTGCTAAAGGGTTCTGA
    TTGTCTGTTATCTAAGTGTTACCACGAATTTGGAACTCAATGGCTTCCTC
    TATTAGAGACTCTGCATCAAGTTCCAGTTGTTCCTGTCGGTCTGCTACCA
    CCTGAAATTCCCGGTGACGAAAAGGACGAAACTTGGGTTTCCATAAAAAA
    ATGGCTGGATGGTAAGCAGAAGGGTAGTGTTGTATATGTCGCTTTAGGCT
    CCGAGGTTTTGGTATCCCAGACTGAAGTTGTGGAACTTGCCTTAGGATTG
    GAGTTGTCCGGTTTGCCATTCGTCTGGGCATATAGAAAGCCAAAGGGACC
    AGCTAAGTCAGACTCAGTTGAATTGCCAGATGGTTTCGTAGAAAGGACAA
    GAGACAGAGGATTGGTTTGGACATCATGGGCCCCACAATTGAGAATTCTG
    AGTCATGAAAGTGTGTGTGGATTCTTGACTCACTGTGGCTCTGGCAGTAT
    TGTTGAAGGACTGATGTTTGGACACCCACTGATAATGTTGCCAATCTTCG
    GTGACCAACCTCTGAATGCAAGATTGCTGGAGGATAAACAAGTTGGTATC
    GAAATCCCAAGAAACGAGGAAGACGGCTGCCTGACTAAGGAATCAGTTGC
    ACGTAGTTTAAGATCTGTAGTTGTTGAAAAAGAAGGTGAAATATATAAGG
    CTAACGCTAGAGAACTTTCAAAGATATACAATGATACCAAGGTGGAGAAA
    GAATATGTTTCACAGTTTGTGGACTATTTGGAGAAAAACGCTAGAGCCGT
    TGCTATCGATCACGAATCATAG
  • In certain embodiments, the nucleic acid comprises a sequence encoding Stevia rebaudiana UDP-glycosyltransferase 74G1. In certain embodiments, the nucleic acid comprises a sequence encoding Stevia rebaudiana UDP-glycosyltransferase 74G1 which comprises the amino acid sequence as set forth in SEQ ID NO: 13 and as listed below or a variant or fragment thereof.
  • SEQ ID NO: 13
    MAEQQKIKKSPHVLLIPFPLQGHINPFIQFGKRLISKGVKTTLVTTIHTL
    NSTLNHSNTTTTSIEIQAISDGCDEGGFMSAGESYLETFKQVGSKSLADL
    IKKLQSEGTTIDAIIYDSMTEWVLDVAIEFGIDGGSFFTQACVVNSLYYH
    VHKGLISLPLGETVSVPGFPVLQRWETPLILQNHEQIQSPWSQMLFGQFA
    NIDQARWVFTNSFYKLEEEVIEWTRKIWNLKVIGPTLPSMYLDKRLDDDK
    DNGFNLYKANHHECMNWLDDKPKESVVYVAFGSLVKHGPEQVEEITRALI
    DSDVNFLWVIKHKEEGKLPENLSEVIKTGKGLIVAWCKQLDVLAHESVGC
    FVTHCGFNSTLEAISLGVPVVAMPQFSDQTTNAKLLDEILGVGVRVKADE
    NGIVRRGNLASCIKMIMEEERGVIIRKNAVKWKDLAKVAVHEGGSSDNDI
    VEFVSELIKA
  • In certain embodiments, the nucleic acid molecule encodes Stevia rebaudiana UDP-glycosyltransferase 74G1 and comprises a nucleotide sequence as set forth in SEQ ID NO: 14 and as listed below or a variant or fragment thereof.
  • SEQ ID NO: 14
    ATGGCGGAACAACAAAAGATCAAGAAATCACCACACGTTCTACTCATC
    CCATTCCCTTTACAAGGCCATATAAACCCTTTCATCCAGTTTGGCAAA
    CGATTAATCTCCAAAGGTGTCAAAACAACACTTGTTACCACCATCCAC
    ACCTTAAACTCAACCCTAAACCACAGTAACACCACCACCACCTCCATC
    GAAATCCAAGCAATTTCCGATGGTTGTGATGAAGGCGGTTTTATGAGT
    GCAGGAGAATCATATTTGGAAACATTCAAACAAGTTGGGTCTAAATCA
    CTAGCTGACTTAATCAAGAAGCTTCAAAGTGAAGGAACCACAATTGAT
    GCAATCATTTATGATTCTATGACTGAATGGGTTTTAGATGTTGCAATT
    GAGTTTGGAATCGATGGTGGTTCGTTTTTCACTCAAGCTTGTGTTGTA
    AACAGCTTATATTATCATGTTCATAAGGGTTTGATTTCTTTGCCATTG
    GGTGAAACTGTTTCGGTTCCTGGATTTCCAGTGCTTCAACGGTGGGAG
    ACACCGTTAATTTTGCAGAATCATGAGCAAATACAGAGCCCTTGGTCT
    CAGATGTTGTTTGGTCAGTTTGCTAATATTGATCAAGCACGTTGGGTC
    TTCACAAATAGTTTTTACAAGCTCGAGGAAGAGGTAATAGAGTGGACG
    AGAAAGATATGGAACTTGAAGGTAATCGGGCCAACACTTCCATCCATG
    TACCTTGACAAACGACTTGATGATGATAAAGATAACGGATTTAATCTC
    TACAAAGCAAACCATCATGAGTGCATGAACTGGTTAGACGATAAGCCA
    AAGGAATCAGTTGTTTACGTAGCATTTGGTAGCCTGGTGAAACATGGA
    CCCGAACAAGTGGAAGAAATCACACGGGCTTTAATAGATAGTGATGTC
    AACTTCTTGTGGGTTATCAAACATAAAGAAGAGGGAAAGCTCCCAGAA
    AATCTTTCGGAAGTAATAAAAACCGGAAAGGGTTTGATTGTAGCATGG
    TGCAAACAATTGGATGTGTTAGCACACGAATCAGTAGGATGCTTTGTT
    ACACATTGTGGGTTCAACTCAACTCTTGAAGCAATAAGTCTTGGAGTC
    CCCGTTGTTGCAATGCCTCAATTTTCGGATCAAACTACAAATGCCAAG
    CTTCTAGATGAAATTTTGGGTGTTGGAGTTAGAGTTAAGGCTGATGAG
    AATGGGATAGTGAGAAGAGGAAATCTTGCGTCATGTATTAAGATGATT
    ATGGAGGAGGAAAGAGGAGTAATAATCCGAAAGAATGCGGTAAAATGG
    AAGGATTTGGCTAAAGTAGCCGTTCATGAAGGTGGTAGCTCAGACAAT
    GATATTGTCGAATTTGTAAGTGAGCTAATTAAGGCTTAA
  • In certain embodiments, the nucleic acid molecule encodes Stevia rebaudiana UDP-glycosyltransferase 74G1 and comprises the sequence as set forth in GenBank Accession number AY345982 or a variant or fragment thereof.
  • In other embodiments, the invention provides for nucleic acids comprising nucleotide sequences encoding a cyclodextrin glucanotransferase (WO1996033267; U.S. Pat. No. 6,271,010).
  • Also provided are nucleic acids comprising nucleotide sequences that encode a sucrose synthase. Accordingly, in certain embodiments, the nucleic acid comprises a sequence encoding sucrose synthase which comprises the amino acid sequence as set forth in SEQ ID NO: 15, 17, 19, 21, 23 or 25 and listed below or a variant or fragment thereof.
  • SEQ ID NO: 15
    (Stevia rebaudiana SUS1 isoform)
    MAERVLTRVHSLRERLDSTLATHRNEILLFLSRIESHGKGILKPHQVMTEFEAICKEDQSKLSDG
    AFYEVLKCTQEAIVQPPWVALAIRLRPGVWEYVRVNVNVLVVEELSVPEYLHFKEELVNGTSN
    GNFVLELDFEPFTASFPRPTLTKSIGNGVEFLNRHLSAKMFHDKDSMHPLLDFLRTHHYKGKTM
    MLNDRIQNLNALQSVLRKASEYLSTLDAATPYSEFEHKFQEIGLERGWGDKAEVVMEMIHMLL
    DLLEAPDACTLEKFLGRIPMVFNVVILSPHGYFAQENVLGYPDTGGQVVYILDQVPALEREMLK
    RIKEQGLDIIPRILIVTRLLPDAVGTTCGQRLEKVFGAEHSHILRVPFRTEKGILRKWISRFEVWP
    YIETFTEDVAKEVTAELQAKPDLIIGNYSEGNLVASLLAHKLGVTQCTIAHALEKTKYPDSDIYWK
    NFEEKYHFSSQFTADLIAMNHTDFIITSTFQEIAGSKDTVGQYESHTAFTMPGLYRVVHGIDVFD
    PKFNIVSPGADMGIYYSYTEKEKRLTALHPEIDELLFSSVENEEHLCVLKDKSKPILFTMARLDNV
    KNLTGLVEWYAKNDRLRELVNLVVVGGDRRKESKDLEEQAQMQKMHELIETYKLNGQFRWIS
    SQMNRVRNGELYRVIADTRGAFIQPAFYEAFGLTVVEAMTCGLPTFATLHGGPAEIIVHGKSGF
    HIDPYHGDQVTELLVNFFEKTKQDPGHWEAISKGGLQRIQEKYTWQIYSDRLLTLAGVYGFWK
    HVSKLDRLEIRRYLEMFYALKYRKLAESVPLAVDE
    SEQ ID NO: 17
    (Stevia rebaudiana SUS2 isoform)
    MATSKLSRTHSMRERVEETLSAHRNEIVSLLSRYVAQGKAILQPHQILHELENIIGDVTSRQKLT
    DGPFGDALKTAQECIVLPPFVALAVRPRPGVWEYVRVDAYQLSVEQLTVSEYLTFKEELVGES
    NSSLMLELDFEPFNASFPRPTRSSSIGNGVQFLNRHLSSSMFRSKDCLEPLLDFLRTHRHNGH
    VMMLNDRITSMTRLQSSLVKAEEYLSKLPSDTDYSEFQYELQGMGFERGWGNNAERIIEMMHL
    LSDILQAPDPSILESFLARIPMVFNVVILSIHGYFGQANVLGLPDTGGQIVYILDQVRALENEMLLK
    LKHQGLDIKPRILIVTRLIPDAKGTSCNQRLERVSGTEHTHILRVPFRTEKGILRKWISRFDVWPF
    LEKFTQDAASEISAELHGTPDLIIGNYSDGNLVASLLSYKMGVTQCNIAHALEKTKYPDSDLYWK
    KFDEKYHFSCQFTADLLAMNNADFIITSTYQEIAGTKNTVGQYESHSSFTLPGLYRVVHGIDVFD
    PKFNIVSPGADMSIYFSYTEKEKRLTSLHTTIEKLLFDPTQTEDYIGNLSDKSKPIIFSMARLDHVK
    NITGLVEWYAKNEKLRGLANLVVVAGYNNVKRSSDREEIAEIEKMHQLIKKYKLDGQMRWISAQ
    TNRAQNGELYRYIADGRGIFVQPAIYEAFGLTVVEAMTCGLPTFATCHGGPGEIIENGVSGFHID
    PYHPDTASATMADFFQKCKEDPSYWFKISEAGLKRIYERYTWKIYSERLMTLAGVYSFWKYVS
    KLERRETRRYLEMFYILKFRDLVKSVPVATDDEA
    SEQ ID NO: 19
    (Stevia rebaudiana SUS3 isoform)
    MATPKLTRTPSMRERLEETLSAHRNDIVSLLSRYVDQGKAILQPHHLLDEIDNFIGDQNCRQKLA
    DSLFGEILKSAQEGIILPPYVTLAVRPRPGVWDFLRVNVDELSVEQLTVSEYLSFKEELVDGQSR
    NPFVLELDLEPFNATFPRMSRSSSIGNGVQFLNRHLSSIMFRNKDCMDPFLDFLRAHKHKGYA
    MMLNDRIQTMSRLESSLAKAEDHLSKLPPETPYSEFEYVLQGMGFERGWGDNCERVLGMMHL
    LSDILQAPDPSILEKFLGKMPMIFNVVVLSIHGYFGQANVLGLPDTGGQVVYILDQVRSLENEML
    LKLRHQGLDIKPKILIVTRLIPNAKGTSCNQRLEKVSGTEYTYILRVPFRTEKGILGKWLSRFDIW
    PYLEAFTTDAASEIAAELHGVPDLLIGNYSDGNLVASLLSNKLGVTQCNIAHALEKTKYPDSDLY
    WKKFEDKYHFSCQFTADLLAMNNADFIITSTYQEIAGTKNTVGQYENHSSFTLPGLYRVVHGID
    VFDPKFNIVSPGADMAIYFSYADKERRLTSLHPTIEKLLFDTEQNDVHIGNINDPSKPMIFTMARL
    DHVKNITGFVECYAKNNKLREHANLVVIAGYNDAKKSSDREEIAEIEKMHNLIKQYKLDGQMRW
    ISAQTNRARNGEFYRYIADGRGVFVQPAFYEAFGLTVVEAMTCGLPTFATCHGGPAEIIEDGVS
    GFHIDPYHPDKMSTTLADFFQKCKEEPSYWGKISDGGLKRISERYTWKIYSERLMTLAGVYSF
    WKYVSKLERRETRRYLEMFYILKFRQLVKSVPLAVDEEP
    SEQ ID NO: 21
    (Stevia rebaudiana SUS4 isoform)
    MASASSSIMKRSESIVDTMPEALKQSRYHMKKCFLKYVEKGIRMMKRHHLIQEMETAIEDKDEK
    AQLLDGLLGYILCTTQEAAVVPPCVAFAIRPNPGFWEFVKVNSNDLSVDGITATDYLKFKEMIVD
    ETWAKDENALEIDFGSMDFNLPNMSLSCSIGNGVNFTSKFITCKLYAQSSCQQLLVDYLLSLNH
    QGENLMINDALNSVSKLRAALIVAHASLSSLPNDTPYQSFELRFKEWGFEKGWGDNAERARET
    IRFLLEVLQAPDPINLEALFSRIPNIFNVVLFSIHGYFGQSNVLGLPDTGGQVVYVLDQVVAMEEE
    LLMRIKQQGLNFKPQILVVTRLLPDAKGTKCNQVLEPVLNTKHSHILRVPFRTDKGVLRKWVSR
    FDIYPYLENFTQDASAKIIEMMEGKPDLIIGNYTDGNLVASLMANKLGTTLGTIAHALEKTKYEDS
    DMNWKQFDPKYHFSCQFTADMIAMNSADFIITSTFQEIAGSKDRPGQYESHEAFTLPGLYRVV
    SGINVFDPKFNIASPGADQTVYFPYTETKKRFTAFQPAIEELLFSKVENEEHIGYLEDKTKPIIFS
    MARLDTVKNITGLTEWFGENKRLRSLVNLVIVAGFFDPSKSKDREEMAEIKKMHLLIEKYQLKG
    QIRWIAAQTDKNRNSELYRFIADSKGAFVQPALYEAFGLTVIEAMNCGLPTFATNQGGPAEIIVD
    GVSGFQIDPNFGDQSSNKIADFFQKCKEDPGYWNNISEGGLKRIYECYTWKIYANKVLNMGNI
    YSFWKRLNKEQKEAKQRYIELFYNLHYKNLVRTVPIASDEAQPAPVSRAKLATQPTRRTQSRL
    QRLFGA
    SEQ ID NO: 23
    (Stevia rebaudiana SUS5 isoform)
    MAASSSPIMKRSESVLDTMPEALRQSRYHMKKCFLKYVGKGKRMVKLHHLMQEMETVIEDKD
    EKAQLLEGLLGYILCTTQEAAVVPPYVAFAIRPNPGFWEFVKVNSNDLSVKGITSTDYLKFKEMI
    VDETWANDENALEIDFGAMDFNLPTMSLSSSIGNGVNFTSKFIISKLYAHSGSQLQSLVDYLLSL
    NHQGEKLMINDKLNTVSKLQAALIVAHSFLSSLPNDTPYQSFELRFKEWGFEKGWGDYAERVQ
    ETIRFLLEVLQAPDPVNLEAFFSRVPNIFNIVLFSIHGYFGQSNVLGLPDTGGQVVYVLDQVVAM
    EEELLLRIKQQGLSFKPHILVVTRLLPDAKGTECSQVLEPVLNTKHSHILRVPFRTEKGVLRKWV
    SRFDIYPYLEKFTQDASAKITEMMEGKPDLIIGNYTDGNLVASLMANKLGSTLGTIAHALEKTKYE
    DSDMKWKHLDTKYHFSCQFTADMIAMNSADFIITSTFQEIAGSKDRPGQYESHEAFTLPGLYRV
    VSGINVFDPKFNIASPGADQTVYFPYTETPKRFTTFQPAIQELLFSKVENDEHIGYLEDKNKPIIF
    SMARLDMVKNITGLTEWFGENKRLRSLVNLVIVAGFFDPSKSKDREEMEEIKKMHLLIEKYELK
    GQIRWIVAQTDKNRNSELYRCIADSKGAFVQPALYEAFGLTVIEAMNCGLPTFATNQGGPAEIIV
    DGVSGFQIDPNYGDESSNKIADFFQKCKQDPGYWNRISDGGLMRIYECYTWKIYANKVLNMG
    NIYTFWKQLNKEQKDAKQRYIELFYNQHYKNLVRTVPIVSDEDDQVTRAKPATQPSTRRTQSA
    LQRLLGA
    SEQ ID NO: 25
    (Stevia rebaudiana SUS6 isoform)
    MDFGIAETLAEALKQNRYHARRCFERFTSRGKRMVKPQELLHMIEKTIDDKLERTKVLEGSMG
    QILSSTQEAIVIPPYVILGLRANPGQWAYVKINADDVTVESLTPSQYLKFKESIYDQEWAKDENA
    LELDFGAFDFDTPRLILPSSIGNGLGYISKFMTSRIGGDLENAKPLLDHLLALKYHGEKLMINETID
    TVSKLQKALIVADVYLSAHPKDEQYQTLEPKLKEWGFEKGWGDTAERVRETMKMLSEILQAPD
    PINMQSFFSRLPVVFNIVIFSIHGYFGQSDVLGLPDTGGQVVYILDQVKALEEEILLRIKMQGLNA
    KPRILVVSRLIPDAQGTKCNEEMEPILNTMHSHILRVPFRTSKGVVPQWVSRFDIYPYLERFSQD
    AASKILEVMECKPDLILGNYTDGNIVASLIAKKFGVTQGTIAHALEKTKYEDSDVNWKNFEKKYH
    FSCQFTADLISMNAADFIITSTYQEIVGSKQRPGQYETHGAFSMPGLCRVVSGINVFDPKFNIAS
    PGAEQSVYFPYTEKEKRLTDFHPAIKELLFNEQDNDEHMGYLADVTKPIIFSMARLDTVKNITGL
    TEWFGKNKRLRSLVNLVVVAGFFDPSKSKDREEMEEIKKMHELIEKYKLKGQMRWIAAQNDRT
    RNGELYRCISDTKGAFVQPALYEAFGLTVIEAMNCGLPTFATNQGGPAEIIVDGVSGFHIDPVN
    GDESSNKIADFFTKCKVDGEYWDRVSQAGLORIYECYTWKMYANKALNMGSMYGFWRQLNK
    ETKQAKQRYIDILYNLQFKNLAKTIEIPDFVTPKLQEPVKTEPTKPLQEARPREPVQKLVPEETRL
    PKLELTKLGQPNLMSNARKPLIVLVSVLIVAYASKNLYRRYFK
  • In certain embodiments, the nucleic acid molecule encodes sucrose synthase and comprises a nucleotide sequence as set forth in SEQ ID NO: 16, 18, 20, 22, 24 or 26 and listed below or a fragment or variant thereof.
  • SEQ ID NO: 16
    (encodes SUS1 isoform)
    ATGGCGGAACGTGTACTCACTCGTGTTCACAGTCTTCGTGAGCGTCTCGATTCAACTCTCG
    CAACTCATCGTAATGAAATCCTCTTGTTTCTTTCAAGGATTGAAAGCCATGGAAAAGGAATA
    TTGAAGCCTCATCAAGTTATGACTGAATTTGAAGCTATCTGCAAAGAAGATCAGAGCAAAC
    TCTCTGATGGTGCTTTTTATGAAGTTCTTAAATGCACACAGGAAGCAATAGTGCAACCTCC
    ATGGGTTGCACTCGCGATCCGTCTTCGACCCGGTGTTTGGGAATATGTTAGAGTCAATGTT
    AATGTTTTGGTGGTTGAAGAATTAAGTGTTCCTGAATATCTTCACTTCAAAGAAGAATTGGT
    TAATGGAACATCGAATGGCAACTTCGTGTTGGAACTGGATTTTGAACCTTTTACCGCATCG
    TTTCCTCGACCAACTTTAACCAAGTCTATTGGTAATGGTGTTGAGTTTCTAAACAGACATTT
    ATCTGCTAAAATGTTTCATGATAAGGATAGCATGCACCCTCTTCTTGATTTCCTACGGACTC
    ACCACTATAAGGGAAAGACAATGATGTTGAATGATAGAATCCAAAACCTCAATGCTCTACAA
    TCGGTGTTGCGAAAGGCGTCAGAGTACTTATCAACACTCGACGCAGCAACACCGTACTCT
    GAGTTTGAACATAAGTTTCAAGAAATCGGGTTGGAGAGAGGTTGGGGTGATAAAGCGGAG
    GTCGTAATGGAGATGATCCACATGCTTCTAGACCTTCTAGAAGCACCCGACGCATGCACA
    CTCGAGAAGTTTCTCGGAAGAATCCCAATGGTTTTCAATGTTGTCATTCTTTCGCCTCACG
    GCTACTTCGCCCAAGAAAATGTGTTGGGATATCCCGACACTGGCGGTCAGGTTGTTTACAT
    CTTGGATCAAGTTCCCGCTCTGGAACGCGAGATGCTCAAAAGGATTAAGGAGCAAGGACT
    CGATATCATTCCTCGTATATTGATTGTTACGAGGCTTCTTCCCGACGCGGTTGGGACCACA
    TGCGGGCAACGTTTAGAGAAAGTGTTTGGAGCCGAACACTCGCATATTCTTCGGGTCCCG
    TTTAGAACCGAAAAGGGTATTCTTCGTAAATGGATCTCTCGTTTTGAGGTGTGGCCTTACA
    TCGAGACTTTCACCGAGGATGTTGCTAAAGAAGTTACAGCAGAGTTGCAAGCAAAACCAGA
    TTTGATCATTGGAAACTATAGTGAAGGAAATTTGGTTGCATCTTTGCTAGCTCACAAGTTGG
    GTGTCACTCAGTGTACCATTGCTCATGCTTTGGAGAAAACTAAATACCCGGATTCTGATAT
    CTACTGGAAGAACTTTGAGGAGAAATATCATTTCTCTTCGCAGTTTACCGCTGATCTTATCG
    CTATGAACCATACCGACTTCATCATCACCAGTACTTTCCAAGAAATTGCTGGAAGTAAGGA
    CACGGTTGGACAGTACGAGAGTCATACCGCGTTCACAATGCCGGGATTGTATCGGGTGGT
    TCACGGGATCGATGTTTTTGACCCCAAATTCAATATTGTTTCACCCGGGGCCGATATGGGA
    ATTTACTACTCGTATACCGAGAAAGAAAAGAGGCTCACTGCGCTTCACCCTGAAATCGATG
    AACTTCTCTTTAGTTCCGTCGAAAACGAAGAACACTTATGTGTGTTGAAGGATAAGAGTAAA
    CCAATCTTGTTCACAATGGCGCGATTGGATAATGTGAAGAATTTAACCGGACTGGTTGAAT
    GGTACGCTAAAAACGACCGCCTTCGTGAGCTCGTGAACCTCGTGGTCGTCGGTGGTGAC
    CGAAGGAAAGAGTCGAAAGATCTTGAAGAACAAGCTCAGATGCAGAAGATGCATGAACTT
    ATCGAAACCTACAAACTCAACGGTCAGTTCAGGTGGATATCCTCACAAATGAACCGCGTGA
    GGAACGGTGAGTTGTATCGCGTTATTGCTGACACACGAGGTGCGTTTATCCAGCCTGCGT
    TTTACGAGGCGTTTGGGTTGACGGTTGTGGAGGCCATGACTTGTGGCCTGCCGACATTCG
    CGACACTTCATGGTGGGCCCGCTGAGATTATTGTTCACGGGAAATCCGGGTTCCATATTG
    ACCCGTATCACGGTGACCAGGTCACCGAGTTGCTGGTCAATTTCTTTGAGAAAACTAAACA
    AGACCCGGGTCATTGGGAGGCCATTTCCAAGGGTGGTCTGCAACGTATTCAGGAGAAATA
    CACGTGGCAGATTTATTCAGATAGGTTGTTGACGCTTGCCGGAGTTTATGGATTCTGGAAG
    CATGTGTCGAAGCTTGACAGGCTCGAGATCCGTCGTTATCTTGAAATGTTTTACGCGCTCA
    AGTATCGCAAACTGGCTGAATCTGTTCCATTGGCTGTTGATGAGTGA
    SEQ ID NO: 18
    (encodes SUS2 isoform)
    ATGGCGACAAGTAAGTTGAGCAGAACGCATAGTATGCGTGAGCGTGTTGAAGAAACTCTT
    TCCGCTCATCGCAACGAAATCGTTTCTCTTCTTTCTAGGTATGTGGCTCAGGGGAAGGCGA
    TATTGCAGCCGCATCAGATACTCCATGAACTTGAGAATATCATCGGTGATGTTACTTCGCG
    CCAAAAGCTTACAGATGGTCCGTTTGGAGATGCGTTGAAGACAGCACAGGAATGTATAGTT
    CTACCTCCATTTGTAGCTTTAGCAGTTCGTCCAAGACCTGGTGTTTGGGAATACGTGCGCG
    TGGATGCATATCAACTAAGTGTGGAACAACTAACTGTTTCAGAGTATCTTACCTTCAAAGAA
    GAACTTGTTGGAGAGTCTAATAGTTCTTTAATGCTCGAGTTGGATTTTGAGCCATTTAATGC
    TTCGTTTCCTAGACCAACCCGTTCTTCATCCATTGGCAATGGAGTTCAGTTCCTGAATCGC
    CACCTGTCGTCAAGCATGTTTCGCAGCAAAGATTGTTTAGAACCGCTTCTGGATTTCCTAC
    GCACACACAGACATAATGGACATGTAATGATGTTAAATGACCGCATAACAAGCATGACTAG
    ACTTCAATCTTCTTTGGTCAAAGCAGAGGAATATCTTTCTAAACTACCATCTGATACAGACT
    ACTCTGAGTTTCAATATGAATTGCAAGGAATGGGTTTTGAAAGAGGATGGGGAAACAATGC
    TGAAAGAATCATTGAGATGATGCATCTTCTCTCAGACATTCTACAAGCTCCAGATCCTTCCA
    TTTTGGAATCTTTTCTTGCTAGAATACCTATGGTGTTTAATGTTGTTATATTATCAATACATG
    GCTACTTTGGGCAAGCAAATGTTTTGGGTTTGCCAGATACTGGTGGCCAGATTGTATATAT
    ATTGGATCAAGTCCGTGCATTGGAAAATGAGATGCTTCTTAAATTAAAGCACCAAGGACTG
    GATATCAAACCTAGGATTCTGATTGTGACTCGGTTAATACCTGATGCAAAAGGTACTTCAT
    GTAACCAACGACTGGAAAGAGTCAGTGGAACTGAACACACACATATACTTCGTGTTCCTTT
    TAGAACCGAGAAAGGAATTCTTCGTAAATGGATCTCAAGGTTTGATGTATGGCCTTTTTTG
    GAGAAATTTACACAGGATGCAGCAAGTGAAATTTCTGCTGAGTTGCATGGTACTCCAGATC
    TTATAATTGGAAATTATAGTGATGGCAATCTTGTTGCCTCTTTATTATCTTACAAAATGGGA
    GTAACCCAGTGTAACATTGCTCATGCTTTAGAGAAAACAAAGTATCCAGATTCTGATTTATA
    TTGGAAGAAATTTGATGAGAAATATCACTTTTCTTGTCAATTTACTGCTGATCTTTTAGCCAT
    GAACAATGCAGATTTTATCATCACCAGCACATACCAAGAAATCGCGGGAACGAAAAATACT
    GTCGGACAATACGAGAGTCATTCGTCTTTCACTCTCCCGGGGCTCTACAGGGTTGTTCAT
    GGTATTGACGTTTTTGACCCTAAGTTCAACATTGTGTCTCCAGGGGCAGATATGTCTATAT
    ACTTCTCATACACCGAGAAGGAAAAAAGACTTACATCTCTTCATACTACAATTGAGAAGTTA
    TTGTTTGACCCTACACAAACTGAAGATTACATTGGAAATCTGAGTGATAAATCAAAACCGAT
    AATTTTTTCAATGGCAAGACTTGATCATGTGAAGAACATTACGGGTCTGGTTGAGTGGTAC
    GCTAAGAATGAGAAGCTTAGAGGACTAGCAAACCTTGTTGTGGTTGCTGGTTATAATAATG
    TGAAGAGGTCTAGTGACAGAGAAGAAATTGCAGAAATTGAAAAAATGCATCAACTTATTAA
    GAAATACAAATTAGATGGTCAGATGAGATGGATTTCAGCACAAACAAACCGCGCACAAAAT
    GGTGAACTTTATCGCTATATTGCTGATGGAAGGGGAATCTTTGTACAGCCCGCTATTTATG
    AAGCTTTTGGGCTGACAGTGGTGGAGGCCATGACTTGTGGGCTTCCAACATTTGCAACTT
    GCCATGGTGGGCCAGGAGAGATAATTGAAAATGGTGTTTCGGGCTTCCATATCGACCCGT
    ATCATCCGGATACTGCATCAGCCACAATGGCTGATTTTTTTCAGAAATGCAAGGAGGACCC
    GAGTTATTGGTTCAAGATATCTGAAGCAGGGCTTAAAAGAATATATGAAAGGTACACATGG
    AAAATTTACTCTGAACGGTTGATGACATTAGCTGGAGTTTATAGCTTCTGGAAGTATGTCTC
    GAAACTTGAGAGACGTGAAACAAGACGATATCTTGAGATGTTTTATATTCTTAAGTTCCGTG
    ATCTGGTAAAATCTGTTCCAGTGGCTACTGATGATGAGGCTTAG
    SEQ ID NO: 20
    (encodes SUS3 isoform)
    ATGGCGACACCTAAGCTTACGCGAACACCAAGCATGCGAGAGCGTCTTGAAGAAACTTTA
    TCAGCTCATCGCAACGATATCGTCTCTCTTCTTTCCAGGTATGTAGATCAAGGTAAGGCCA
    TATTGCAGCCCCACCACCTACTTGACGAAATCGATAACTTCATCGGAGATCAAAATTGCCG
    CCAAAAGCTTGCTGATAGTCTATTCGGTGAAATCCTCAAGTCCGCACAGGAAGGTATAATT
    CTTCCTCCATATGTAACGCTTGCTGTTCGTCCAAGACCTGGTGTTTGGGACTTTTTGCGTG
    TGAATGTCGATGAATTGAGTGTCGAGCAACTTACTGTTTCTGAGTATTTAAGCTTCAAGGA
    GGAGCTTGTAGATGGCCAGAGTAGGAACCCGTTTGTGTTGGAACTGGATCTGGAACCGTT
    TAATGCAACATTTCCCCGGATGTCACGATCTTCATCCATCGGCAATGGAGTTCAGTTTCTC
    AACCGTCATCTCTCGTCAATTATGTTTCGCAACAAAGATTGTATGGATCCGTTTCTTGATTT
    CCTTCGTGCTCATAAACATAAAGGATACGCGATGATGTTGAATGATCGGATACAAACAATG
    TCTAGACTTGAATCTTCTTTAGCAAAAGCGGAGGATCATCTCTCTAAACTACCACCCGAAA
    CACCGTACTCCGAATTCGAATACGTATTGCAAGGAATGGGGTTTGAAAGAGGTTGGGGGG
    ATAATTGTGAAAGAGTTCTTGGTATGATGCATCTTCTTTCTGACATTCTTCAAGCTCCAGAT
    CCTTCGATTCTTGAAAAGTTTCTTGGAAAGATGCCGATGATCTTCAATGTTGTTGTGTTATC
    GATTCATGGTTACTTTGGTCAGGCTAATGTTTTGGGTTTGCCGGATACCGGTGGTCAGGTT
    GTATATATATTGGATCAAGTACGTTCTTTGGAGAATGAAATGTTACTTAAATTAAGGCATCA
    AGGACTTGATATCAAACCCAAGATTCTAATTGTAACTCGATTGATACCAAATGCCAAAGGTA
    CTTCATGCAACCAACGATTGGAGAAAGTAAGTGGAACCGAATACACGTATATATTACGTGT
    CCCTTTTAGGACAGAGAAAGGGATTCTTGGTAAATGGTTATCAAGGTTTGATATATGGCCT
    TATTTGGAGGCGTTTACAACGGATGCAGCAAGTGAAATTGCTGCTGAGTTACACGGTGTTC
    CGGATCTTTTAATAGGAAACTACAGTGATGGGAATCTCGTTGCCTCCTTGCTATCTAACAA
    ATTGGGCGTAACCCAGTGCAACATTGCACACGCGTTAGAGAAAACAAAGTATCCAGATTCC
    GACTTATATTGGAAGAAATTTGAGGACAAATATCACTTTTCATGTCAATTTACCGCCGACCT
    TCTAGCAATGAACAATGCAGATTTTATCATCACTAGCACATACCAAGAGATTGCAGGAACG
    AAAAACACCGTTGGACAATACGAGAATCATTCATCGTTCACTCTTCCGGGTCTATACAGGG
    TTGTTCACGGTATCGATGTCTTTGACCCGAAGTTCAACATCGTGTCACCAGGGGCAGATAT
    GGCAATTTACTTCTCATATGCCGATAAAGAGAGACGACTTACATCTCTACATCCCACAATTG
    AGAAGCTATTGTTCGACACTGAGCAGAACGATGTACACATTGGAAATATAAATGACCCGTC
    TAAACCCATGATTTTCACAATGGCGAGGCTTGATCATGTGAAGAATATAACTGGATTCGTC
    GAGTGTTATGCTAAAAATAATAAGTTGAGGGAACACGCAAATCTTGTGGTTATTGCTGGTT
    ATAATGACGCGAAGAAATCAAGTGATCGAGAAGAAATTGCGGAAATTGAAAAGATGCATAA
    TCTTATCAAGCAATACAAACTTGATGGTCAGATGAGATGGATATCAGCCCAAACAAACCGG
    GCCCGAAATGGGGAATTTTATCGGTATATCGCTGATGGTAGGGGCGTTTTCGTCCAGCCC
    GCTTTCTATGAAGCATTTGGGCTTACGGTTGTGGAGGCGATGACATGTGGGCTCCCAACA
    TTTGCCACGTGTCATGGTGGGCCTGCTGAGATCATTGAGGATGGTGTGTCGGGGTTCCAT
    ATTGATCCATATCATCCTGATAAGATGTCGACTACGTTAGCTGATTTTTTTCAAAAGTGCAA
    AGAGGAACCTAGTTACTGGGGTAAAATATCCGATGGCGGGCTGAAAAGAATAAGTGAAAG
    GTACACATGGAAGATATATTCGGAACGGTTGATGACGTTGGCGGGCGTATATAGCTTTTG
    GAAATATGTGTCAAAACTCGAGAGGCGTGAAACCCGTCGATACCTTGAGATGTTCTACATT
    TTAAAGTTTCGTCAACTGGTGAAGTCGGTTCCGCTAGCTGTTGATGAGGAGCCGTAA
    SEQ ID NO: 22
    (encodes SUS4 isoform)
    ATGGCATCTGCTTCAAGTTCTATCATGAAACGGTCTGAATCAATAGTTGACACCATGCCAG
    AAGCCTTAAAGCAGAGCCGCTATCATATGAAAAAATGTTTTCTAAAATATGTAGAAAAAGGA
    ATTCGCATGATGAAAAGACATCATTTGATACAAGAAATGGAGACCGCAATTGAAGACAAGG
    ATGAAAAGGCTCAGCTTCTAGATGGCTTACTTGGCTACATCTTGTGCACAACTCAGGAAGC
    AGCCGTTGTTCCTCCTTGTGTTGCATTTGCTATAAGACCGAATCCTGGATTCTGGGAGTTT
    GTTAAAGTCAACTCTAATGATCTATCGGTTGATGGGATAACTGCCACAGATTACTTGAAGTT
    CAAGGAAATGATCGTAGATGAGACATGGGCTAAAGATGAAAATGCATTGGAGATTGACTTT
    GGATCGATGGACTTTAACCTACCAAACATGAGTTTATCTTGTTCGATTGGAAATGGTGTTAA
    CTTCACATCAAAATTCATTACTTGTAAACTTTACGCACAATCTAGTTGCCAACAACTGCTTG
    TTGATTACTTGCTCTCATTGAATCATCAAGGAGAAAATCTTATGATCAATGATGCATTAAAC
    TCAGTCTCAAAACTTCGAGCGGCTTTAATTGTAGCTCATGCGTCGCTATCTTCGTTGCCCA
    ACGATACTCCATATCAAAGCTTCGAGCTTAGATTCAAAGAATGGGGATTTGAGAAGGGATG
    GGGAGATAACGCGGAACGCGCGAGGGAAACAATTCGGTTTCTTTTGGAGGTTCTTCAAGC
    ACCCGATCCGATAAACCTCGAGGCTTTATTCAGCAGGATTCCAAACATATTCAACGTTGTTT
    TATTCTCGATTCATGGGTATTTTGGTCAATCCAATGTTCTTGGATTGCCCGATACTGGTGG
    CCAAGTGGTTTATGTTTTGGATCAAGTGGTAGCTATGGAAGAAGAACTACTCATGAGGATC
    AAACAACAAGGACTCAACTTCAAGCCTCAAATTCTTGTGGTGACCCGACTTCTTCCTGATG
    CTAAAGGGACCAAGTGTAATCAGGTGTTGGAACCAGTTCTGAACACGAAACATTCGCATAT
    TCTTAGGGTTCCATTCAGGACTGATAAAGGTGTTCTTCGTAAATGGGTATCTCGATTTGATA
    TCTATCCATATCTCGAAAACTTCACTCAGGATGCAAGTGCGAAAATCATTGAAATGATGGAA
    GGGAAACCGGATCTTATCATCGGAAACTATACCGATGGAAACCTTGTTGCATCACTCATGG
    CTAACAAACTCGGAACGACATTGGGAACAATTGCACATGCTTTGGAGAAAACCAAATACGA
    AGATTCAGACATGAATTGGAAGCAATTCGACCCAAAATATCACTTCTCCTGCCAATTTACAG
    CCGATATGATTGCAATGAACTCAGCTGATTTCATCATCACAAGTACTTTCCAAGAAATCGCT
    GGAAGTAAAGATAGACCCGGACAATATGAAAGCCATGAAGCATTTACACTTCCAGGATTAT
    ACAGAGTTGTTTCAGGCATCAACGTGTTCGATCCCAAATTCAATATCGCGTCTCCAGGAGC
    CGATCAAACCGTTTATTTCCCGTACACCGAAACAAAGAAACGATTCACTGCATTTCAACCC
    GCCATAGAGGAATTACTCTTCAGTAAAGTTGAAAACGAAGAACACATTGGATACTTAGAAG
    ACAAAACCAAACCGATCATATTCTCAATGGCGCGTCTCGACACAGTTAAGAACATAACAGG
    ACTAACCGAATGGTTTGGAGAGAACAAACGGCTCCGAAGCTTGGTTAATCTTGTAATCGTG
    GCGGGTTTCTTTGACCCGTCAAAGTCAAAAGACAGAGAAGAAATGGCGGAAATAAAGAAA
    ATGCATTTATTGATTGAAAAATATCAGCTTAAAGGTCAAATAAGATGGATTGCTGCACAAAC
    TGATAAGAACCGAAACAGTGAGCTTTACCGGTTTATTGCTGACTCAAAAGGCGCGTTTGTG
    CAGCCCGCTTTGTATGAGGCGTTTGGGCTCACGGTTATTGAGGCGATGAACTGTGGTTTA
    CCGACTTTTGCAACTAATCAAGGTGGTCCAGCTGAGATTATCGTTGATGGTGTTTCTGGGT
    TCCAGATTGATCCTAATTTTGGTGATCAGTCTAGTAATAAGATTGCTGATTTCTTCCAGAAG
    TGTAAGGAAGATCCTGGTTATTGGAATAATATTTCAGAAGGCGGTTTGAAGCGTATATACG
    AATGTTATACTTGGAAGATTTATGCGAATAAAGTGTTGAATATGGGGAACATATACTCGTTT
    TGGAAGCGGTTAAACAAGGAACAAAAAGAAGCAAAACAAAGATACATTGAACTATTCTACA
    ATCTACACTACAAGAACTTGGTTAGGACTGTACCAATTGCTAGTGATGAAGCTCAACCTGC
    ACCAGTGTCAAGGGCAAAACTTGCAACACAACCCACAAGACGTACGCAATCCAGGTTGCA
    AAGGCTGTTTGGAGCTTAA
    SEQ ID NO: 24
    (encodes SUS5 isoform)
    ATGGCAGCTTCTTCAAGTCCCATTATGAAACGGTCTGAGTCAGTACTCGACACCATGCCAG
    AAGCTTTGAGGCAAAGTCGGTATCATATGAAAAAATGCTTTCTAAAATATGTAGGGAAAGG
    AAAGCGGATGGTGAAACTCCACCATTTGATGCAAGAAATGGAGACCGTCATTGAGGACAA
    GGACGAAAAGGCTCAGCTCTTGGAAGGCTTACTTGGTTACATCTTGTGCACCACTCAGGA
    AGCAGCAGTTGTTCCTCCTTATGTCGCCTTTGCAATAAGGCCAAACCCTGGATTTTGGGAG
    TTTGTTAAAGTCAACTCTAATGATCTCTCGGTTAAAGGGATCACTTCCACCGATTACTTGAA
    GTTCAAGGAAATGATCGTTGACGAAACATGGGCTAATGATGAAAATGCATTGGAGATCGAC
    TTTGGAGCAATGGACTTTAACTTGCCAACAATGAGCTTATCTTCTTCAATTGGAAATGGAGT
    TAACTTCACATCAAAGTTTATTATTTCTAAACTTTATGCTCATTCTGGCAGCCAATTACAATC
    TCTAGTTGATTACTTACTTTCATTAAATCATCAAGGAGAAAAACTTATGATAAATGACAAACT
    AAACACAGTTTCAAAACTTCAAGCCGCTCTAATAGTAGCTCATTCTTTCCTTTCTTCATTGC
    CCAACGACACACCGTATCAAAGCTTTGAACTTAGATTTAAAGAGTGGGGTTTTGAAAAAGG
    ATGGGGAGATTATGCAGAAAGGGTGCAAGAAACAATTCGGTTTTTGTTGGAGGTTCTTCAA
    GCACCCGACCCCGTAAACCTAGAGGCCTTTTTTAGCAGGGTTCCAAACATATTCAATATTG
    TTTTATTCTCGATTCATGGGTATTTTGGTCAATCCAATGTTCTTGGCTTGCCCGATACCGGA
    GGTCAGGTAGTTTATGTTTTGGATCAAGTTGTGGCAATGGAAGAAGAATTGCTACTTAGGA
    TTAAGCAACAAGGACTCAGCTTCAAGCCTCATATTCTTGTGGTGACTCGACTTCTTCCCGA
    TGCCAAAGGGACCGAGTGTAGCCAAGTTTTGGAACCAGTTCTCAACACGAAACACTCACA
    CATTCTTAGAGTCCCATTTAGGACAGAAAAAGGTGTTCTTCGTAAATGGGTGTCTCGATTT
    GATATCTATCCATACCTCGAAAAGTTTACTCAGGATGCAAGTGCAAAAATAACTGAAATGAT
    GGAAGGAAAACCTGATCTTATCATTGGAAACTACACTGACGGAAACTTGGTTGCATCTCTC
    ATGGCTAACAAACTCGGAAGCACATTGGGAACGATTGCACACGCGTTAGAGAAGACTAAA
    TACGAAGATTCAGACATGAAATGGAAACATTTGGACACAAAATATCACTTTTCTTGTCAATT
    TACAGCTGATATGATAGCAATGAATTCAGCAGATTTCATCATCACTAGTACTTTCCAAGAAA
    TTGCTGGAAGTAAAGATAGACCCGGTCAGTATGAAAGCCATGAAGCATTTACACTCCCGG
    GTTTATATAGAGTTGTTTCGGGCATCAACGTGTTTGATCCCAAATTCAACATTGCATCTCCG
    GGAGCTGATCAAACCGTTTATTTCCCTTACACGGAAACACCAAAACGATTCACTACTTTTCA
    ACCCGCTATACAAGAATTACTCTTTAGTAAAGTTGAAAACGACGAACACATTGGATATTTAG
    AAGATAAGAATAAACCAATCATCTTCTCAATGGCAAGACTCGACATGGTTAAGAACATAACG
    GGGCTAACCGAATGGTTTGGGGAAAACAAGCGGTTAAGAAGTTTGGTTAATCTTGTAATTG
    TGGCGGGGTTTTTTGATCCGTCAAAATCAAAAGATAGAGAAGAAATGGAAGAAATAAAGAA
    AATGCATTTGTTGATTGAGAAATATGAACTTAAAGGTCAAATAAGATGGATAGTAGCACAAA
    CTGATAAAAACAGAAATAGTGAACTTTATCGTTGTATCGCTGACTCAAAGGGGGCGTTTGT
    GCAACCGGCTTTATATGAAGCGTTTGGGTTAACCGTTATTGAGGCTATGAATTGTGGGTTA
    CCAACTTTTGCAACTAACCAAGGTGGTCCGGCTGAGATTATTGTTGATGGTGTTTCTGGGT
    TCCAAATCGATCCTAATTATGGCGACGAGTCTAGCAACAAGATCGCTGATTTTTTTCAAAAA
    TGCAAACAGGATCCAGGATACTGGAATAGGATTTCAGACGGTGGTTTGATGCGTATATACG
    AATGCTACACATGGAAGATTTATGCAAATAAAGTGTTGAATATGGGGAACATTTACACATTT
    TGGAAGCAGTTAAACAAGGAACAGAAAGATGCGAAACAAAGATACATTGAGCTATTCTACA
    ATCAACATTACAAGAATTTGGTTAGGACTGTGCCGATTGTAAGTGATGAAGATGACCAAGT
    TACAAGGGCAAAACCGGCAACACAACCTTCAACAAGGCGCACACAATCTGCCTTGCAAAG
    GCTGCTTGGAGCTTAA
    SEQ ID NO: 26
    (encodes SUS6 isoform)
    ATGGATTTCGGTATAGCAGAGACTTTGGCCGAGGCATTGAAGCAAAACCGGTACCATGCA
    AGGAGATGCTTTGAGCGTTTTACATCACGTGGAAAAAGGATGGTGAAGCCTCAAGAGTTAT
    TACACATGATTGAAAAAACCATTGACGACAAGCTTGAAAGAACGAAGGTCTTGGAGGGCTC
    AATGGGACAAATCTTGAGTTCCACACAGGAGGCAATCGTTATTCCACCATATGTTATTTTAG
    GATTGAGAGCGAATCCAGGACAATGGGCATACGTTAAGATCAATGCTGATGACGTCACTG
    TTGAGTCACTCACACCTTCACAATATCTAAAGTTCAAAGAATCCATCTACGATCAAGAATGG
    GCAAAGGACGAAAATGCCCTTGAACTAGATTTCGGAGCGTTCGACTTTGATACGCCTCGAT
    TAATCCTCCCGTCATCTATCGGCAACGGACTCGGTTACATTTCAAAGTTCATGACTTCAAG
    AATTGGTGGTGATCTAGAAAACGCGAAGCCGTTGCTTGACCACTTGCTTGCTCTAAAATAT
    CATGGAGAGAAGCTTATGATCAATGAGACAATAGATACAGTTTCAAAGCTCCAGAAAGCAT
    TAATTGTTGCTGATGTCTACTTATCTGCACACCCGAAAGACGAACAATATCAAACCTTAGAG
    CCCAAGCTTAAAGAATGGGGATTTGAGAAAGGATGGGGAGATACTGCTGAAAGAGTTAGA
    GAGACAATGAAAATGCTTTCGGAGATTCTTCAAGCACCCGACCCGATTAACATGCAATCGT
    TCTTTAGCAGGCTTCCGGTGGTCTTCAATATTGTCATATTTTCTATTCATGGGTATTTTGGT
    CAATCAGATGTTCTTGGATTACCTGATACCGGAGGGCAGGTTGTTTACATTCTTGATCAAG
    TTAAAGCATTAGAGGAAGAGATATTGCTAAGAATAAAAATGCAAGGATTGAATGCAAAGCC
    TCGGATTCTTGTGGTGAGTCGACTCATTCCCGACGCACAAGGAACAAAGTGTAACGAGGA
    AATGGAACCGATCTTGAACACAATGCATTCACACATCCTTCGGGTTCCTTTCAGAACCTCA
    AAAGGCGTTGTTCCTCAATGGGTATCGCGGTTTGACATCTACCCGTATCTTGAAAGATTCT
    CACAGGACGCTGCCTCTAAAATACTTGAAGTAATGGAATGTAAACCAGATCTCATACTTGG
    AAACTACACAGATGGAAACATTGTTGCATCACTTATAGCCAAAAAGTTTGGAGTAACACAG
    GGGACGATTGCACACGCGTTAGAGAAGACAAAGTACGAAGATTCGGATGTTAACTGGAAA
    AACTTTGAAAAAAAGTATCATTTCTCATGTCAATTTACCGCGGATTTGATCTCAATGAACGC
    TGCAGATTTCATAATCACAAGCACTTATCAAGAAATTGTGGGAAGCAAACAAAGACCCGGA
    CAGTATGAGACCCACGGGGCGTTTAGTATGCCCGGACTTTGTAGAGTCGTGTCGGGCATC
    AACGTGTTTGATCCTAAGTTCAACATTGCTTCACCCGGTGCGGAACAATCGGTTTATTTTC
    CGTACACCGAGAAGGAGAAACGGTTAACGGATTTTCATCCCGCAATTAAAGAACTACTTTT
    CAACGAACAAGACAATGACGAGCATATGGGATACCTCGCGGATGTAACCAAACCGATAATA
    TTCTCAATGGCGAGGCTCGATACGGTGAAGAACATAACAGGGTTAACCGAGTGGTTCGGT
    AAGAACAAACGACTTAGAAGTCTTGTAAACTTGGTTGTTGTCGCGGGGTTCTTCGATCCAT
    CAAAATCTAAAGACCGTGAAGAGATGGAGGAAATCAAGAAAATGCATGAACTAATAGAGAA
    ATACAAACTCAAGGGTCAGATGAGATGGATCGCGGCTCAAAACGATAGGACCCGCAATGG
    TGAATTGTATCGGTGTATTTCCGATACGAAGGGAGCGTTTGTGCAGCCCGCGTTGTATGA
    GGCTTTTGGGCTCACGGTTATCGAGGCAATGAACTGCGGTCTCCCGACTTTTGCAACCAA
    TCAAGGCGGGCCCGCGGAGATCATAGTTGACGGAGTTTCGGGATTTCATATTGATCCCGT
    TAACGGAGATGAATCAAGCAACAAGATTGCTGATTTCTTCACGAAATGCAAAGTCGATGGC
    GAGTATTGGGACCGCGTGTCGCAAGCGGGACTTCAACGTATTTACGAGTGCTACACATGG
    AAGATGTATGCTAACAAAGCATTGAACATGGGTTCGATGTATGGTTTTTGGAGGCAATTAA
    ACAAAGAAACTAAGCAAGCGAAGCAACGATACATCGATATCTTGTATAACTTACAATTCAAG
    AATTTGGCAAAAACCATTGAAATCCCTGATTTTGTGACTCCTAAACTTCAAGAACCGGTCAA
    AACCGAACCAACAAAACCATTACAAGAAGCAAGACCTCGAGAACCGGTGCAAAAACTGGTA
    CCGGAAGAAACCCGACTGCCAAAACTAGAGTTGACCAAGCTTGGTCAACCGAATTTGATG
    AGCAATGCAAGAAAACCATTGATTGTTCTTGTTTCTGTGTTGATAGTTGCATATGCATCCAA
    GAACTTGTATAGGAGGTATTTCAAATAG
  • In other embodiments, there is provided a nucleic acid comprising a sequence having at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identity to any one of the sequences set forth in SEQ ID NOs: 2, 4, 6, 8, 10, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32 and 34 and fragments thereof or the complement thereof.
  • In other embodiments, there is provided a nucleic acid encoding a polypeptide comprising a sequence at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% percent identity to any one of the sequences set forth in SEQ ID NOs: 1, 3, 5, 7, 9, 13, 15, 17, 19, 21, 23, 25, 27, 29, 30, 31 and 33 and fragments thereof. A worker skilled in the art would readily appreciate that overall sequence identity or similarity may be less than 50% but regions of the enzyme (such as the catalytic site or areas adjacent to the catalytic site) may have conserved amino acids. For example, there are conserved amino acids at the opening adjacent to the UDPG catalytic site. In particular, a leucine at position 379 of UGT76G1 is conserved. In certain embodiments, the nucleic acid encodes an UDP-glucosyltransferase having the sequence SDFGLDQ at a position corresponding to amino acid residues 375 to 381 of the UGT76G1 set forth in SEQ ID NO:1.
  • In certain embodiments, fragments are at least 10, at least 20, at least 50 nucleotides in length. The fragments may be used, for example, as primers or probes.
  • Also provided are nucleic acids that hybridize to the nucleic acids of the present invention or the complement thereof. In certain embodiments, there is provided a nucleic acid that hybridizes to any one of the sequences set forth in SEQ ID NOs: 2, 4, 6, 8, 10, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32 and 34 or the complement thereof under conditions of low, moderate or high stringency. A worker skilled in the art readily appreciates that hybridization and the strength of hybridization (i.e., the strength of the association between the nucleic acids) is impacted by such factors as the degree of complementary between the nucleic acids, stringency of the conditions involved, the Tm of the formed hybrid, and the G:C ratio within the nucleic acids. Such a worker could readily determine appropriate stringent (see, for example, Sambrook, et al., Molecular Cloning: A Laboratory Manual, 2nd ed., Cold Spring Harbor Laboratory Press, New York (1989) pp. 9.50-51, 11.48-49 and 11.2-11.3).
  • Typically under high stringency conditions only highly similar sequences will hybridize under these conditions (typically >95% identity). With moderate stringency conditions typically those sequence having greater than 80% identity will hybridize and with low stringency conditions those sequences having greater than 50% identity will hybridize.
  • A non-limiting example of “high stringency conditions” when used in reference to nucleic acid hybridization comprise conditions equivalent to binding or hybridization at 42° C. in a solution consisting of 5×SSPE (43.8 g/I NaCl, 6.9 g/I NaH2PO4H2O and 1.85 g/I EDTA, pH adjusted to 7.4 with NaOH), 0.5% SDS, 5×Denhardt's reagent and 100 μg/ml denatured salmon sperm DNA followed by washing in a solution comprising 0.1×SSPE, 1.0% SDS at 42° C. when a probe of about 500 nucleotides in length is employed. A non-limiting example of “medium stringency conditions” when used in reference to nucleic acid hybridization comprise conditions equivalent to binding or hybridization at 42° C. in a solution consisting of 5×SSPE (43.8 g/I NaCl, 6.9 g/I NaH2PO4H2O and 1.85 g/I EDTA, pH adjusted to 7.4 with NaOH), 0.5% SDS, 5×Denhardt's reagent and 100 μg/ml denatured salmon sperm DNA followed by washing in a solution comprising 1.0×SSPE, 1.0% SDS at 42° C. when a probe of about 500 nucleotides in length is employed. A non-limiting example “Low stringency conditions” when used in reference to nucleic acid hybridization comprise conditions equivalent to binding or hybridization at 42.degree. C. in a solution consisting of 5×SSPE (43.8 g/I NaCl, 6.9 g/I NaH2PO4H2O and 1.85 g/I EDTA, pH adjusted to 7.4 with NaOH), 0.5% SDS, 5×Denhardt's reagent and 100 μg/ml denatured salmon sperm DNA followed by washing in a solution comprising 5×SSPE, 0.1% SDS at 42° C. when a probe of about 500 nucleotides in length is employed.
  • The polynucleotides include the coding sequence polypeptide, in isolation, in combination with additional coding sequences (e.g., a purification tag, a localization signal, as a fusion-protein, as a pre-protein, or the like), in combination with non-coding sequences (e.g., introns or inteins, regulatory elements such as promoters (including inducible promoters, tissue-specific promoters (such as root-specific or leaf specific promoters), enhancers, terminators, and the like), and/or in a vector or host environment in which the polynucleotide encoding a transcription factor or transcription factor homologue polypeptide is an endogenous or exogenous gene.
  • Appropriate additional coding sequences (e.g., a purification tag, a localization signal, as a fusion-protein, as a pre-protein, or the like), non-coding sequences (e.g. regulatory elements such as promoters (including inducible promoters, tissue-specific promoters (such as root-specific or leaf specific promoters), enhancers, terminators, and the like), and vectors for use in prokaryotic such as E. coli and eukaryotic cells, including but not limited to yeast and plant cells are known in the art.
  • Polypeptides
  • The present invention provides for glycosyltransferases. The glycosyltransferases of the present invention are capable of primary, secondary and/or tertiary glycosylations. In certain embodiments, the glycosyltransferases are capable of primary, secondary and tertiary glycosylations. In other embodiments, the glycosyltransferases are capable of secondary and/or tertiary glycosylations. In certain embodiments, the glycosyltransferases is a glucosyltransferase, including but not limited to a UDP-glycotransferase. The glucosyltransferases include but are not limited to a Stevia rebaudiana UDP-glucosyltransferase, such as UGT76G1 or UGT74G1 or an Oryza sativa glucosyltrasferase, such as Os03g0702000. In other embodiments, the invention provides for a cyclodextrin glucanotransferase. Also provided are sucrose synthases.
  • In certain embodiments, there is provided an UGT76G1 or UGT76G1-like glucosyltransferase. UGT76G1-like glucosyltransferase include for example, other members of the UGT76G1 clade such as UGT76G2 or UGT76H1. Accordingly, in certain embodiments, there is provided an UGT76G1 comprising the amino acid sequence as set forth in any one of SEQ ID NOs: 1, 3, 5 and 7 or fragments and variants thereof. In certain embodiments, there is provided an UGT76G1 encoded by the nucleic acid molecule comprising the sequence as set forth in any one of SEQ ID NOs: 2, 4, 6 and 8.
  • In certain embodiments, there is provided an UGT76G2 comprising the amino acid sequence as set forth in SEQ ID NO: 27 or fragments and variants thereof. In certain embodiments, there is provided an UGT76G1 encoded by the nucleic acid molecule comprising the sequence as set forth in SEQ ID NO: 28.
  • In certain embodiments, there is provided an UGT76H1 comprising the amino acid sequence as set forth in SEQ ID NO: 29 or fragments and variants thereof. In certain embodiments, there is provided an UGT76G1 encoded by the nucleic acid molecule comprising the sequence as set forth in SEQ ID NO: 30.
  • In certain embodiments, there is provided an Os03g0702000 or Os03g0702000-like glucosyltransferase. Os03g0702000-like glucosyltransferase include for example, other members of the UGT91clade such as UGT91D1 or UGT91D2. Accordingly, in certain embodiments, there is provided an Os03g0702000 comprising an amino acid sequence as set forth in SEQ ID NO: 9 or fragments and variants thereof. In certain embodiments, there is provided an Os03g0702000 encoded by the nucleic acid molecule comprising the sequence as set forth in SEQ ID NO: 10.
  • In certain embodiments, there is provided an UGT91 D1 comprising the amino acid sequence as set forth in SEQ ID NO: 31 or fragments and variants thereof. In certain embodiments, there is provided an UGT91 D1 encoded by the nucleic acid molecule comprising the sequence as set forth in SEQ ID NO: 32.
  • In certain embodiments, there is provided an UGT91 D2 comprising the amino acid sequence as set forth in SEQ ID NO: 33 or fragments and variants thereof. In certain embodiments, there is provided an UGT76G1 encoded by the nucleic acid molecule comprising the sequence as set forth in SEQ ID NO: 34.
  • In certain embodiments, there is provided a Stevia rebaudiana UGT74G1. Accordingly, in certain embodiments, the UGT74G1 comprises the amino acid sequence as set forth in SEQ ID NO: 13 or fragments and variants thereof. In certain embodiments, the UGT74G1 is encoded by the nucleic acid molecule comprising the sequence as set forth in SEQ ID NO: 14.
  • In other embodiments, the invention provides for a cyclodextrin glucanotransferase. Cyclodextrin-glucanotransferase is commercially available (CGTase, Toruzyme 3.0L, trademark of Novozymes Inc.).
  • In certain embodiments, there is provided sucrose synthase. Accordingly, in certain embodiments, the sucrose synthase comprises the amino acid sequence as set forth in SEQ ID NO: 15, 17, 19, 21, 23 or 25 or fragments and variants thereof. In certain embodiments, the polypeptide comprises an amino acid sequence encoded by the nucleic acid molecule comprises comprising the sequence as set forth in SEQ ID NO: 16, 18, 20, 22, 24 or 26.
  • In other embodiments, there is provided a polypeptide comprising a sequence at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% percent identity to any one of the sequences set forth in SEQ ID NOs: 1, 3, 5, 7, 9, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31 and 33 and fragments thereof. A worker skilled in the art would readily appreciate that overall sequence identity or similarity of related enzymes may be less than 50% but regions of the enzyme (such as the catalytic site or areas adjacent to the catalytic site) may have conserved amino acids and therefore the related enzymes have similar activity. For example, there are conserved amino acids at the opening adjacent to the UDPG catalytic site. In particular, a leucine at position 379 of UGT76G1 is conserved. In certain embodiments, the nucleic acid encodes an UDP-glucosyltransferase having the sequence SDFGLDQ at a position In certain embodiments, fragments are at least 10, at least 20, at least 50 amino acids in length. In certain embodiments, the polypeptide sequences contain heterologous sequences including but not limited to purification tags such as a HIS tag. In a certain embodiments, there is provided a polypeptide comprising a 6×HIS tag at the N-terminus. In other embodiments, there is provided a polypeptide comprising a 6×HIS tag at the C-terminus.
  • Methods for screening the activity of glycosyltransferases including glucosyltransferases and cyclodextrin glucanotransferases are known in the art. As such, a worker skilled in the art could readily determine if the glycosyltransferases are capable of primary, secondary and/or tertiary glycosylations (see, for example Dewitte et al., J Biotechnol. 2016 Sep. 10; 233:49-55. doi: 10.1016/j.jbiotec.2016.06.034; Grubb et al., Plant J. (2014) 79, 92-105; Richman et al., Plant J. (2005) 41, 56-67; Tanaka et al., Plant Cell Rep. (1996) 15, 819-823; Tanaka et al., J. Nat. Prod (1993) 56(12), 2068-2072. In addition, methods for screening the activity of sucrose synthase are also known in the art. (Baroja-Fernandez et al., PNAS. (2012) 109(1), 321-326. doi: 10.1073/pnas.1117099109; Barratt et al., Plant Physiol. (2001) 127, 655-664; Huber and Akazawa, Plant Physiol. (1986) 81, 1008-1013.
  • Cells and Plants
  • The present invention further provides cells and plants which express one or more of the polypeptides of the present invention. The cells and plants may naturally express one or more of the polypeptides of the present invention or have been modified to express one or more the polypeptides of the present invention. The cells may be prokaryotic or eukaryotic cells and include but are not limited to, E. coli, yeast such as Pichia pastoris, Stevia rebaudiana, Phytolacca Americana, Cannabis including but not limited to Cannabis sativa, Cannabis indica and Cannabis ruderalis.
  • In certain embodiments, there is provided a cell which expresses an UGT76G1 or UGT76G1-like glucosyltransferase (such as UGT76G2 and UGT76H1). Accordingly, in certain embodiments, there is provided a cell which expresses an UGT76G1 glucosyltransferase comprising a sequence encoding the amino acid sequence as set forth in any one of SEQ ID NOs: 1, 3, 5 and 7. In certain embodiments, there is provided a cell which expresses an UGT76G1-like glucosyltransferase comprising a sequence encoding the amino acid sequence as set forth in SEQ ID NO: 27 or 29. The cell may further express further glucosyltransferases, such as Os03g0702000 or Os03g0702000-like glucosyltransferase (such as UGT91 D1 and UGT91 D2) and/or a sucrose synthase, such as the sucrose synthase comprising the sequence as set forth in SEQ ID NO: 15, 17, 19, 21, 23 or 25.
  • Accordingly, in certain embodiments, there is provided a cell which expresses UGT76G1 glucosyltransferase comprising a sequence encoding the amino acid sequence as set forth in any one of SEQ ID NOs: 1, 3, 5 and 7 and Os03g0702000 glucosyltransferase comprising the sequence as set forth in SEQ ID NO:10. The cell may further express a sucrose synthase comprising the sequence as set forth in SEQ ID NO: 15, 17, 19, 21, 23 or 25.
  • In certain embodiments, there is provided a cell which expresses an Os03g0702000 or Os03g0702000-like glucosyltransferase. Accordingly, in certain embodiments, there is provided a cell which expresses Os03g0702000 glucosyltransferase comprising a sequence encoding the amino acid sequence as set forth in SEQ ID NO: 10. The cell may further express a sucrose synthase, such as the sucrose synthase comprising the sequence as set forth in SEQ ID NO: 15, 17, 19, 21, 23 or 25.
  • Transgenic cells and plants (including plant cells, or plant explants, or plant tissues) can be produced by a variety of well established techniques. Following construction of a vector, most typically an expression cassette, including a polynucleotide of the invention, standard techniques can be used to introduce the polynucleotide into cell or a plant. Optionally, the plant cell, explant or tissue can be regenerated to produce a transgenic plant.
  • In a certain embodiments, there is provided Cannabis plants genetically engineered to express one or more of the proteins of the invention. A worker skilled in the art would readily appreciate appropriate vectors and promoters for genetically engineering Cannabis plats. For example, a tissue specific promoter, such as a secretory trichomes specific promoter may be used such that the proteins of the invention are expressed in the same tissue that cannabinoids are produced in, namely the secretory trichomes of the plant. Suitable promoter elements include the promoter for the cytosolic O-acetylserine(thiol)lyase (OASA1) enzyme from Arabidopsis thaliana (Gutierrez-Alcala 2005).
  • Transformation and regeneration of plant cells is now routine, and the selection of the most appropriate transformation technique will be determined by the practitioner. Suitable methods can include, but are not limited to: electroporation of plant protoplasts; liposome-mediated transformation; polyethylene glycol (PEG) mediated transformation; transformation using viruses; micro-injection of plant cells; micro-projectile bombardment of plant cells; vacuum infiltration; and Agrobacterium tumeficiens mediated transformation. Transformation means introducing a nucleotide sequence into a plant in a manner to cause stable or transient expression of the sequence.
  • Successful examples of the modification of plant characteristics by transformation with cloned sequences which serve to illustrate the current knowledge in this field of technology, and which are herein incorporated by reference, include: U.S. Pat. Nos. 5,571,706; 5,677,175; 5,510,471; 5,750,386; 5,597,945; 5,589,615; 5,750,871; 5,268,526; 5,780,708; 5,538,880; 5,773,269; 5,736,369 and 5,610,042.
  • Following transformation, plants may be selected using a dominant selectable marker incorporated into the transformation vector. Typically, such a marker will confer antibiotic or herbicide resistance on the transformed plants, and selection of transformants can be accomplished by exposing the plants to appropriate concentrations of the antibiotic or herbicide.
  • Methods
  • The present invention further provides methods for the production of cannabinoid glycoside prodrugs and the cannabinoid glycosides prodrugs produced by the methods. The methods may be in vitro or in vivo (in a cell system or in planta). In certain embodiments, there is provided a method of producing cannabinoid glycoside prodrugs, said method comprising incubating a cannabinoid aglycone with one or more sugar donors in the presence of one or more glycosyltransferases.
  • The aglycones include but are not limited to: cannabinoids, including but not limited to cannabidiol, cannabidivarin, cannabigerol, tetrahydrocannabinol, cannabinol and cannabidiolic acid, endocannabinoids including but not limited to arachidonoylethanolamide (anandamide, AEA), 2-arachidonoylethanolamide (2-AG), 1-arachidonoylethanolamide (1-AG), and docosahexaenoyl ethanolamide (DHEA, synaptamide); and vanilloids including but not limited to vanillin, curcumin, and capsaicin.
  • A worker skilled in the art would readily appreciate that the one or more sugar donors will be dependent on the one or more glycosyltransferases used in the method and/or the desired end products. For example, for UDP-glucosyltransferases, the sugar donors include but are not limited to UDP-glucose, UDP-glucuronic acid, UDP-mannose, UDP-fructose, UDP-xylose, UDP-fluorodeoxyglucose, and UDP-rhamnose. For cyclodextrin glucanotransferase, the sugar donor includes maltodextrin.
  • In certain embodiments, there is provided a method of producing a cannabinoid glycoside, said method comprising incubating an aglycone with a sugar donor in the presence of a glycosyltransferase. Also provided are the cannabinoid glycosides produced by the above method. In specific embodiments, there is provided a method of producing a cannabinoid glycoside, said method comprising incubating an aglycone with UDP-glucose, in the presence of a UGT76G1 or UGT76G1-like glucosyltransferase under conditions that allow for glycosylation. In other specific embodiments, there is provided a method of producing a glycoside prodrug, said method comprising incubating an aglycone with maltodextrin, in the presence of a cyclodextrin glucanotransferase under conditions that allow for glycosylation.
  • An exemplary method for producing cannabinoid-glycosides comprises incubating a cannabinoid, with UDP-glucose in the presence of a UGT76G1 or UGT76G1-like glucosyltransferase under conditions which allow for glycosylation. Also provided are cannabinoid-glycosides produced by the above method.
  • A further exemplary method for producing cannabinoid-glycosides comprises incubating a cannabinoid with maltodextrin in the presence of a cyclodextrin glucanotransferase under conditions which allow for glycosylation. Also provided are cannabinoid-glycosides produced by the above method.
  • In certain embodiments, there is provided a method of producing a cannabinoid glycoside, said method comprising incubating an aglycone with one or more sugar donors in the presence of a first glycosyltransferase and a second glycosyltransferase under conditions which allow for glycosylation. Also provided are cannabinoid glycosides produced by the above method.
  • A worker skilled in the art would readily appreciate that the first glycosyltransferase and a second glycosyltransferase may be provided concurrently or added sequentially. In addition, if more than one sugar donor is used, the sugar donors may be provided concurrently or added sequentially. Such a worker would further appreciate that the structure of the resulting cannabinoid glycoside may be dependent on the order the glycosyltransferases are provided. In addition, the ratio of first to second glycosyltransferase may impact the resulting products. A worker skilled in the art would further appreciate that the activity levels of the glycosyltransferases may dictate the ratios and the ratios could be readily determined by a worker skilled in the art. For example, the ratios first to second glycosyltransferase include but are not limited to 1:1, 1:2, 1:10, 1:50 and vice versa.
  • In specific embodiments, there is provided a method of producing a cannabinoid glycoside, said method comprising incubating an aglycone with UDP-glucose in the presence of a UGT76G1 or UGT76G1-like glucosyltransferase and Os03g0702000 or Os03g0702000-like glucosyltransferase under conditions which allow for glycosylation. In alternative specific embodiments, there is provided a method of producing a cannabinoid glycoside, said method comprising incubating an aglycone with UDP-glucose and maltodextrin in the presence of a UGT76G1 or UGT76G1-like glucosyltransferase and cyclodextrin glucanotransferase under conditions which allow for glycosylation. Also provided are cannabinoid glycosides produced by the above methods.
  • An exemplary method for producing cannabinoid-glycosides comprises incubating cannabinoid, including but not limited to cannabidiol, cannabidivarin, cannabigerol, tetrahydrocannabinol, cannabinol and cannabidiolic acid, with UDP-glucose in the presence of a UGT76G1 or UGT76G1-like glucosyltransferase and Os03g0702000 or Os03g0702000-like glucosyltransferase under conditions which allow for glycosylation. Also provided are cannabinoid-glycosides produced by the above method.
  • A further exemplary method for producing cannabinoid-glycosides comprises incubating cannabinoids with UDP-glucose and maltodextrin in the presence of a UGT76G1 or UGT76G1-like glucosyltransferase and and cyclodextrin glucanotransferase under conditions which allow for glycosylation. Also provided are cannabinoid-glycosides produced by the above method.
  • It is within the scope of the present invention that each of the above described glycosylation methods may be applied to a lower order cannabinoid glycoside to form a higher order cannabinoid glycoside. For example, a cannabinoid monoglycoside may be glycosylated using any of the glycosylation methods of the present invention to form a diglycoside, or a cannabinoid diglycoside may be glycosylated to form a triglycoside, etc.
  • Methods of purifying the cannabinoid glycosides are known in the art and include for example solid phase extraction, such as column purification.
  • The invention also provides cell culture and in planta methods for the production of cannabinoid glycosides. The methods comprise expressing one or more of the glycosyltransferases in a cell or plant which produces the aglycone and isolating the cannabinoid glycosides. In certain embodiments, one or more sucrose synthases are also expressed. Appropriate vectors and genetic engineering methods are known in the art.
  • The invention also provides methods for the conversion of UDP to UDPG utilizing the sucrose synthases of the present invention. Accordingly, in certain embodiments of the methods of producing cannabinoid glycosides which utilize UDP-glucose as a sugar donor, the methods further comprise the use of sucrose synthase to recycle UDP. In certain embodiments, there is provided a method of producing a cannabinoid glycoside, said method comprising incubating aglycone with UDP-glucose, in the presence of a UGT76G1 glucosyltransferase and a sucrose synthase under conditions that allow for glycosylation.
  • The invention will now be described with reference to specific examples. It will be understood that the following examples are intended to describe embodiments of the invention and are not intended to limit the invention in any way.
  • EXAMPLES Example 1: Conversion of Cannabinoids to Cannabinoid Glycoside Prodrugs
  • Glycosylation reactions consisted of 50 mM KPO4 pH 7.2, 3 mM MgCl2, 0.005% CBD, 2.5% UGT76G1 purified enzyme preparation, and 2.5 mM UDP-glucose. Buffers were degassed and tubes were purged with nitrogen, reactions were protected from light and incubated at 28° C. with 180 rpm agitation for 18 hours. Reactions were then extracted 3× with an equal volume of ethyl acetate, evaporated to dryness, and dissolved in a half volume of HPLC grade methanol. 50 microliters was injected on a reverse phase C18 column and eluted with a gradient of acetonitrile starting at 10% and increasing to 99%. UGT76G1 was produced through expression in Pichia pastoris and purified through standard molecular biology techniques. The UGT76G1 enzyme was found to glycosylate CBD in a UDP-glucose dependent manner. This activity was also proportional to the amount of UDP-glucose present. Incubation temperature was 28° C., and an acceptable range would be 20° C. to 30° C. as high temperatures can cause significant degradation of CBD. Reactions were carried out in the dark to prevent photo-degradation of the substrates. Gentle agitation from 120 to 200 rpm were used to mix the reactions in an inert atmosphere.
  • Substrate CBD in the reactions was replaced with Δ9THC and CBDV and performed in an identical fashion with similar results. Enzyme combinations needed to create various products are listed in Table 4 for CBD-glycosides, Table 5 for CBDV-glycosides, and Table 6 for Δ9THC-glycosides.
  • Other enzymes screened for activity towards CBD were the Stevia rebaudiana UGT74G1, UGT85C2, UGPase, E. coli Maltodextrin phosphotransferase (MalP), and O. sativa Os03g0702000 (SEQ ID NO. 9). No primary glycosylation activity was seen with any other tested enzyme other than UGT76G1.
  • Example 2: 2-0 Glycosylation of of CBD-Monoglycoside
  • Enzymatic reactions are performed as described in Example 1 but with the inclusion of recombinant Os03g0702000 enzyme at a 1:2 ratio relative to UGT76G1. Samples were extracted and analyzed as in Example 1. Recombinant Os03g0702000 enzyme was codon optimized and expressed in E. coli BL21-DE3 cells and purified by immobilized metal ion chromatography.
  • Example 3: Conversion of CBD to Alpha-Glycoside Linked CBD Compounds
  • Recombinant cyclodextrin glucanotransferase (CGTase, Toruzyme 3.0L trade name, Novozymes Inc.) was added to reactions as indicated in Example 1 but without UDPG or UGT76G1. Maltodextrin was used at 0.05% final concentration, and Toruzyme 3.0L was used at 0.1%. Samples were extracted and analyzed as in example 1. Additionally, reactions from Example 1 were carried out to convert cannabinoids to cannabinoid-glycosides, and then CGTase and maltodextrin were added and given adequate time to incubate with the cannabinoid-glycosides. The resulting products contain a β-glycosylation on the cannabinoid backbone, and α-glycosylations emanating from the primary sugar. This additional treatment created a new category of compounds termed β-primed, α-glycosylated cannabinoids.
  • Example 4: Purification of Cannabinoid Glycosides
  • Glycoside products were generated through the aforementioned biocatalytic reactions and purified to homogeneity by C18 solid phase extraction. 100 mg Hypersep C18 columns (Thermo) were hydrated in methanol, rinsed with 50% methanol in water, rinsed with water, glycosylation reaction passed through the column, washed with water, washed with 10%, 20%, and 30% methanol, and the glycoside products were eluted with 45 and 60% methanol in water. Eluates were dried and extracted with ethyl acetate, and dried to completion to yield >95% pure cannabinoid-glycosides for further analysis and testing.
  • Example 5: HPLC Analysis of Cannabinoid Glycoside Prodrugs
  • The HPLC line traces of the reaction products of glycosylation reactions of the cannabinoid aglycones CBD, CBDV, Δ9-THC, CBN, 1-AG and 2-AG, DHEA, AEA, capsaicin, and vanillin, are provided in FIGS. 16 to 24, respectively. Enzymatic reactions were performed as described in Example 1. The solid lines indicate the elution profile of the starting aglycone and the dashed lines indicate the elution profile of the glycosylation reaction product mixture.
  • In FIG. 16, the CBD aglycone retention time is 13.65 minutes, and product peaks are observed at 8.87, 9.02, 9.97, 10.33, and 10.37 min.
  • In FIG. 17, the CBDV aglycone retention time is 12.75 minutes, and product peaks are observed at 8.53, 9.70, and 10.01 min.
  • In FIG. 18, the THC aglycone retention time is 14.45 minutes, and product peaks are observed at 9.46, 10.67, 10.97, 11.28, 11.67, and 12.49 min.
  • In FIG. 19, the CBN aglycone retention time is 14.32 minutes, and product peaks are observed at 10.87, 11.50, and 12.25 min.
  • In FIG. 20, the 1-AG aglycone retention time is 14.18 minutes and the 2-AG aglycone retention time is 14.32 minutes, and product peaks are observed at 11.40, 11.78, 11.83, 11.97, 12.53, 12.92, 13.07, and 13.35 min.
  • In FIG. 21, the DHEA aglycone retention time is 13.78 minutes, and product peaks are observed at 10.09 and 12.43 min.
  • In FIG. 22, the AEA aglycone retention time is 13.87 minutes, and product peaks are observed at 12.47 min.
  • In FIG. 23, the vanillin aglycone retention time is 1.95 minutes and product peaks are observed from 1.25 to 1.35 min.
  • In FIG. 24, the capsaicin aglycone retention time is 11.73 minutes, and product peaks are observed at 10.23 min.
  • Example 6A: LCMS Analysis of CBD Glycosides
  • As shown in the HPLC linetrace of FIG. 16, input CBD aglycone (VB101, 13.65′) has been depleted to 5% of original quantity after +65 hours of incubation time. The CBD-glycosides elute off the HPLC column at 8.87, 9.02, 9.97, 10.33, and 10.37 min. The glycosylated products were identified by LCMS analysis. The glycosylated product “g1” is a monoglycoside, “g2” is a diglycoside, “g3” is a triglycoside, and “g4” is a tetraglycoside. LC-LRMS was performed on a Shimadzu LC-MS 2010 EV instrument. The LC column used was a Silia Chrom XDB C18 Sum, 150A, 4.6×50 mm. The method was 12 min 5 to 95 H2O:ACN gradient. For LRMS electrospray ionization (ESI) was performed in positive mode.
  • VB101 (CBD aglycone) MS data: LC/ESI-LRMS. [M+H]+ (C21H31O2). Calcd: m/z=315. Found: m/z=315.
  • (CBDg1) MS data: LC/ESI-LRMS. [M+H]+ (C27H41O7). Calcd: m/z=477. Found: m/z=477.
  • VB104 (CBDg2) MS data: LC/ESI-LRMS. [M+H]+ (C33H51O12). Calcd: m/z=639. Found: m/z=639.
  • VB110 (CBDg2) MS data: LC/ESI-LRMS. [M+H]+ (C33H51O12). Calcd: m/z=639. Found: m/z=639.
  • (CBDg3) MS data: LC/ESI-LRMS. [M+H]+ (C39H61O17). Calcd: m/z=801. Found: m/z=801. [M+K+H]+ (C39H61O17K). Calcd: m/z=420. Found: m/z=420. [M+ACN+H2O+H]+ (C41H63NO17). Calcd: m/z=860. Found: m/z=860.
  • (CBDg4) MS data: LC/ESI-LRMS. [M+H]+ (C45H71O22). Calcd: m/z=964. Found: m/z=964. [M+H2O+H]+ (C45H73O18). Calcd: m/z=983. Found: m/z=983.
  • (CBDg3) MS data: LC/ESI-LRMS. [M+H]+ (C39H61O17). Calcd: m/z=801. Found: m/z=801. [M+Na]+ (C39H60O17Na). Calcd: m/z=823. Found: m/z=823. [M+K+H]2+ (C39H61O17K). Calcd: m/z=420. Found: m/z=420.
  • Example 6B: LCMS Analysis of Δ9-THC Glycosides
  • In a manner similar to that carried out in Example 6A, the products of the glycosylation reaction of Δ9-THC (shown in the HPLC linetrace of FIG. 18) were identified by LCMS analysis.
  • VB301 (THC aglycone) MS data: LC/ESI-LRMS. [M+H]+ (C21H31O2). Calcd: m/z=315. Found: m/z=315. [M+3ACN+2H]2+ (C27H41N3O2). Calcd: m/z=314. Found: m/z=314.
  • VB304 (THCg2) MS data: LC/ESI-LRMS. [M+H]+ (C33H51O12). Calcd: m/z=639. Found: m/z=639.
  • VB308 (THCg3) MS data: LC/ESI-LRMS. [M+H]+ (C39H61O17). Calcd: m/z=801. Found: m/z=801. [M+Na]+ (C39H60O17Na). Calcd: m/z=823. Found: m/z=823. [M+K+H]+ (C39H61O17K). Calcd: m/z=420. Found: m/z=420.
  • Example 7: NMR Analysis of Cannabinoid Glycosides
  • FIG. 27 depicts the 1NMR spectra of isolated VB104 and FIG. 28 depicts the 1H MR spectra of isolated VB110. Each of these products was isolated from the reaction mixture produced by the glycosylation reaction of CBD. The 1H NMR spectra of 10 mg/ml solutions of each compound prepared in CD30D were obtained on a Bruker Avance II 400 MHz instrument using TopSpin acquisition and processing software.
  • Example 8: Solubility Analysis
  • C18 retention times were empirically determined on a linear ramp of increasing acetonitrile on a Phenomenex Kinetex 2.6u 100A C18 column, on a Dionex HPLC equipped with Diode Array Detector. C Log P values in Table A were predicted by ChemDraw (CambridgeSoft). Reference cannabinoids were analyzed by HPLC and established log P values (http://pubchem.ncbi.nlm.nih.gov/) and used to create a calibration line as depicted in FIG. 29. The predicted c Log P values correlated with the reference calibration line. C18 reverse phase HPLC retention times were plotted against the c Log P values presented in Table A, as depicted in FIG. 29. Data point numbering correlates with table numbering. Open diamonds indicate novel cannabinoid glycosides, filled diamonds indicate reference cannabinoids and derivatives. C log P values were predicted by ChemDraw (CambridgeSoft). Linear regression was performed on all data points (R2=0.9455).
  • TABLE A
    CLogP values for select cannabinoid
    glycosides and reference cannabinoids
    # Compound Retention Time ClogP
    1 VB110 8.967 3.4
    2 11-COOH- 9.347 3.7
    Tetrahydrocannabinol Glucuronide
    3 VB104 10.720 4.3
    4 VB304 11.250 4.7
    5 VB302 11.688 5.7
    6 11-COOH-Tetrahydrocannabinol 12.910 5.7
    7 Cannabidivarin 13.017 5.6
    8 11-OH-Tetrahydrocannabinol 13.037 5.9
    9 Cannabidiol 13.647 6.6
    10 Cannabinol 14.178 7.3
    11 Tetrahydrocannabinol 14.487 7.2
  • Example 9: Bioavailability Assay
  • In order to investigate the effectiveness of glycosylation to effect site-specific drug delivery, VB110 was administered to three mice by oral gavage and the animals sacrificed at 30, 60, and 90 minutes. Eight week old male Swiss mice were fasted for 12 hours prior to administration of 120 mg/kg VB110 in 10% Ethanol USP, 10% Propylene Glycol USP, 0.05% Sodium Deoxycholate USP, 79.95% Saline USP. Following termination and tissue harvest, the intestinal contents were then extracted and analyzed by C18 reverse phase HPLC. As shown in FIG. 30A, the small intestinal contents showed intact VB110, but no decoupled CBD. As shown in FIG. 30B, the large intestinal contents contained both VB110 and CBD in the 60 and 90 minute time points. This decoupling of VB110 is consistent with the large intestinal decoupling seen for sennoside beta-glycosides, and is the result of secreted beta-glycosidases from the large intestinal microflora.
  • Example 10: Analysis of Large Intestine Contents Upon Administration of CBD and CBD Glycosides
  • In order to investigate the metabolism and decoupling of CBD-glycosides in the large intestine, an aqueous solution of a mixture of CBD-glycosides was administered to a mouse by oral gavage. As a control, a solution of CBD in cremophor, ethanol, and saline was administered to a second mouse. The animals were each sacrificed at 2 hours. Following termination and tissue harvest, the intestinal contents were then extracted and analyzed by C18 reverse phase HPLC. The mice employed in this example were eight week old male Swiss mice fasted for 12 hours prior to administration of the solutions.
  • The resulting extracts were analyzed by LCMS performed using a Shimadzu LC-MS 2010 EV. LC separation was carried out using a Silia Chrom XDB C18 5 um, 150A, 4.6×50 mm. The method was 12 min, 5 to 95 H2O:ACN gradient elution. Low resolution MS was performed in negative mode via electrospray ionization (ESI). Acetic acid and formic acid were used as sample additives during analysis, and the injection volume was 20 μl.
  • Analysis of the large intestinal contents of animals administered a mixture of oral CBD-glycosides indicated that both aglycone and glycosides were present, along with hydroxy metabolites of each:
  • [CBD−H], [2CBD−H] and [CBD*2OH+Formic acid−H] MS data: LC/ESI-LRMS. [M−H] (C21H29O2). Calcd: m/z=313. Found: m/z=313. [2M−H] (C42H59O4). Calcd: m/z=627. Found: m/z=627. [M*2OH+Formic acid−H] (C22H31O6 ). Calcd: m/z=391. Found: m/z=391.
    [CBDg1−H], [CBDg1+Cl] and [2CBDg1−H] MS data: LC/ESI-LRMS. [Mg1−H] (C27H39O7). Calcd: m/z=475. Found: m/z=475. [Mg1+Cl] (C27H40O7Cl). Calcd: m/z=511. Found: m/z=511. [2Mg1−H] (C54H79O14). Calcd: m/z=951. Found: m/z=951.
    [CBDg2−H] and [CBDg2+Acetic acid−H] MS data: LC/ESI-LRMS. [Mg2−H] (C33H49O12). Calcd: m/z=637. Found: m/z=637. [Mg2+Acetic acid−H] (C35H53O14 ). Calcd: m/z=697. Found: m/z=697.
    [CBDg3−H], [CBDg3*OH−H] and [CBDg3*OH−2H] MS data: LC/ESI-LRMS. [Mg3−H] (C39H59O17). Calcd: m/z=799. Found: m/z=799. [Mg3*OH−H] (C39H59O18). Calcd: m/z=815. Found: m/z=815. [Mg3*OH−2H]−2 (C39H58O18). Calcd: m/z=407. Found: m/z=407.
  • Analysis of the large intestinal contents of animals administered oral CBD indicated that hydroxy metabolites of CBD were present:
  • [CBD*20H+Formic acid−H] and [2CBD*3OH+Acetic acid−H] MS data: LC/ESI-LRMS. [M*2OH+Formic acid−H] (C22H31O6 ). Calcd: m/z=391. Found: m/z=391. [2M*3OH+Acetic acid−H] (C44H63O12 ). Calcd: m/z=783.9. Found: m/z=784.
  • The plasma and brains from the same animals were also extracted and analyzed by HPLC for the presence of CBD-glycosides and CBD. CBD was only present in the control animal that received CBD aglycone (data not shown). The contents of the small intestines from the same animals were also extracted and analyzed by HPLC for the presence of CBD-glycosides and CBD, but no CBD aglycone was present in the small intestines (data not shown, consistent with THC decoupling data shown in example 11). The presence of the CBD aglycone in the large intestinal contents indicates the successful delivery of CBD-glycosides, and the subsequent hydrolysis of the glycosides by beta-glycosidase enzymes only present in the large intestine. The presence of decoupled CBD in the large intestine, but not in the small intestine, indicates that glycoside decoupling only occurs upon transit to the large intestine. The presence of CBD detoxification metabolite CBD-2OH is also consistent with delivery of CBD and absorption into the intestinal epithelium where CBD begins to be metabolized. This example illustrates the potential to administer CBD-glycosides, safely transit the CBD-glycosides through the small intestine without absorption, transit to the large intestine where the sugars can be decoupled to release CBD locally, avoiding systemic absorption and delivery of the CBD to other tissues where it can have unwanted effects.
  • Example 11: Analysis of Large Intestine Contents Upon Administration of THC-Glycosides
  • In order to investigate the metabolism and decoupling of THC-glycosides in the large intestine, an aqueous solution of a mixture of THC-glycosides was administered to two mice by oral gavage. The first animal was sacrificed at 2 hours and the second animal was sacrificed at 4 hours. Following termination and tissue harvest, the intestinal contents were then extracted and analyzed by C18 reverse phase HPLC. The mice employed in this example were eight week old male Swiss mice fasted for 12 hours prior to administration of the solutions.
  • The resulting extracts were analyzed by LCMS under the same conditions employed in Example 10.
  • Analysis of the large intestinal contents from mice administered THC glycosides after 2 hours indicated that both THC aglycone and THC glycosides were present, along with hydroxy metabolites of each:
  • [THC−H], [THC*OH−H], [2THC*3OH+Acetic acid−H] and [THC*2OH+Formic acid−H] MS data: LC/ESI-LRMS. [M−H] (C21H29O2). Calcd: m/z=313. Found: m/z=313. [M*OH−H] (C21H29O3). Calcd: m/z=329. Found: m/z=329. [2M*3OH+Acetic acid−H] (C44H63O12 ). Calcd: m/z=783.9. Found: m/z=783. [M*2OH+Formic acid−H] (C22H31O). Calcd: m/z=391. Found: m/z=391.
    [THCg1+Cl], [THCg1+Acetic acid−H], [2THCg1−H], and [2THCg1+Acetic acid−H] MS data: LC/ESI-LRMS. [Mg1+Cl] (C27H40O7Cl). Calcd: m/z=511. Found: m/z=511. [Mg1+Acetic acid−H] (C29H43O9). Calcd: m/z=535. Found: m/z=535. [2Mg1−H] (C54H79O14). Calcd: m/z=951. Found: m/z=951. [2Mg1+Acetic acid−H] (C56H83O16 ). Calcd: m/z=1011. Found: m/z=1011.
    [THCg2−H], [THCg2+Acetic acid−H] and [THCg2*OH+Formic acid−H] MS data: LC/ESI-LRMS. [Mg2−H] (C33H49O12). Calcd: m/z=637. Found: m/z=637. [Mg2+Acetic acid−H] (C35H53O14). Calcd: m/z=697. Found: m/z=697. [Mg2*OH+Acetic acid−H] (C34H51O15 ). Calcd: m/z=699. Found: m/z=699.
    [THCg3−H], [THCg3+Acetic acid−H], [CBDg3*OH−H] and [CBDg3*OH−2H] MS data: LC/ESI-LRMS. [Mg3−H] (C39H59O17). Calcd: m/z=799. Found: m/z=799. [Mg3+Acetic acid−H] (C41H63O19). Calcd: m/z=859. Found: m/z=859. [Mg3*OH−H] (C39H59O18 ). Calcd: m/z=815. Found: m/z=815. [Mg3*OH−2H]−2 (C39H58O18 2−). Calcd: m/z=407. Found: m/z=407.
  • Analysis of the THC glycosides mixture extract after 4 hours indicated that both THC aglycone and THC glycosides were confirmed, along with hydroxy metabolites of each:
  • [THC−H], [THC*OH+Acetic acid−H], [2THC*30H+Acetic acid−H] and [THC*2OH+Formic acid−H] MS data: LC/ESI-LRMS. [M−H] (C21H29O2). Calcd: m/z=313. Found: m/z=313. [M*OH+Acetic acid−H] (C23H33O5). Calcd: m/z=389. Found: m/z=389. [2M*3OH+Acetic acid−H] (C44H63O12 ). Calcd: m/z=783.9. Found: m/z=784. [M*2OH+Formic acid−H] (C22H31O). Calcd: m/z=391. Found: m/z=391.
    [THCg1+Cl], [THCg1+Acetic acid−H], [2THCg1−H], and [2THCg1+Acetic acid−H] MS data: LC/ESI-LRMS. [Mg1+Cl] (C27H40O7Cl). Calcd: m/z=511. Found: m/z=511. [Mg1+Acetic acid−H] (C29H43O9). Calcd: m/z=535. Found: m/z=535. [2Mg1−H] (C54H79O14). Calcd: m/z=951. Found: m/z=951. [2Mg1+Acetic acid−H] (C56H83O16 ). Calcd: m/z=1011. Found: m/z=1011.
    [THCg2−H] and [THCg2+Acetic acid−H] MS data: LC/ESI-LRMS. [Mg2−H] (C33H49O12). Calcd: m/z=637. Found: m/z=637. [Mg2+Acetic acid−H] (C35H53O14 ). Calcd: m/z=697. Found: m/z=697.
    [THCg3−H], [THCg3+Acetic acid−H], [CBDg3*OH−H], [CBDg3*OH−2H] and [CBDg3*OH+Acetic acid−2H] MS data: LC/ESI-LRMS. [Mg3−H]−(C39H59O17). Calcd: m/z=799. Found: m/z=799. [Mg3+Acetic acid−H] (C41H63O19). Calcd: m/z=859. Found: m/z=859. [Mg3*OH−H] (C39H59O18 ). Calcd: m/z=815. Found: m/z=815. [Mg3*OH−2H]−2 (C39H58O18 2−). Calcd: m/z=407. Found: m/z=407. [Mg3*OH+Acetic acid−2H]−2 (C41H62O20 2−). Calcd: m/z=467. Found: m/z=467.
  • The plasma and brains from the same animals were also extracted and analyzed by HPLC for the presence of THC-glycosides and THC, but neither compound was seen in these tissues (data not shown). The contents of the small intestines from the same animals were also extracted and analyzed by HPLC for the presence of THC-glycosides and THC, but no THC aglycone was observed (data not shown, consistent with CBD decoupling data shown in Example 10). The presence of the THC aglycone in the large intestinal contents at 2 and 4 hours indicates the successful delivery of THC-glycosides, and their subsequent hydrolysis of the glycosides by beta-glycosidases in the large intestine. The presence of decoupled THC in the large intestine, but not in the small intestine, indicates that glycoside decoupling only occurs upon transit to the large intestine. The presence of THC detoxification metabolites in the large intestine is further proof that the THC aglycone is present and being absorbed by the intestinal epithelium where it begins to be metabolized. This example illustrates the potential to administer THC-glycosides orally, transit the THC-glycosides through the small intestine without absorption, transit to the large intestine where the sugars can be decoupled to release THC locally, avoiding systemic absorption and delivery of the THC to the central nervous system where it can have unwanted psychoactivity.
  • Example 12: Discovery of Novel Sucrose Synthase Isoforms from Stevia rebaudiana
  • A number of research groups have utilized simple UDP to UDPG recycling systems to decrease the amount of UDPG needed for product formation (Hardin 2004, Bungarang 2013). These studies have characterized the primary sucrose synthase isoforms found in leaf tissue, which presumably carry out the synthesis of sucrose by reacting fructose with UDPG, producing sucrose and spent UDP.
  • As plants are known to contain numerous isoforms of the sucrose synthase enzyme, identification of alternative SUS enzymes from the Stevia rebaudiana plant with enhanced activity for the back reaction of UDP+sucrose→UDPG+fructose was carried out. As steviol glycosides occur at a high level in Stevia leaves, it was postulated that a sucrose synthase from the leaves of Stevia would have improved ability to catalyze the back reaction that recycles UDP to UDPG. Six sucrose synthase isoforms were identified within the stevia transcriptome, all having similar homology to the 6 isoforms found in Arabidopsis thaliana and named in conjunction with their homologues. These transcripts were cloned as described in materials and methods with the corresponding sequence ID information listed herein.
  • Enzymatic activities were tested and assayed for their ability to enhance UGT reactions with decreased UDPG input. The best isoform, SrSUS4, was capable of recycling UDP to UDPG with sucrose, in concert with the steviol 19-O-glucosyltransferase SrUGT74G1 mediated glycosylation of steviol bioside to stevioside.
  • Targeted mutagenesis was performed to mutate a serine residue at the N-terminus that is commonly phosphorylated in planta to prevent dimerization (Hardin 2004). SrSUS1-S13D mutants were created by mutating serine at position 13 to an aspartic acid residue (S13D), thus forming a phospho-mimetic protein. Additionally, the creation of SrSus1-S13R,L14I was created to replace the serine with an arginine, a large charged residue, also to prevent dimerization and inactivation of the enzyme. Sucrose synthase mutants showed improved UDPG production activity compared to their native counterparts. SrSUS5 (SEQ ID NOs. 19 and 20) was identified in the Stevia transcriptome and primers designed (SEQ ID NOs. 67 and 68), but was not able to be amplified from cDNA. SrSus4 showed an impressive UDPG recycling activity with a 20% improvement over the activity seen in SrSus1. It is proposed that SrSus4 is the ideal isoform for carrying out the back reaction of converting of UDP to UDPG in the presence of sucrose. For midi-scale purification of cannabinoid glycosides the use of C18 flash chromatography columns were employed. Biotage flash C18 columns with 33 g of resin were washed, loaded, washed, and eluted using peristaltic pumps to achieve the similar separation and purification as the gravity fed Hypersep columns listed previously.
  • Relative activity for UDPG production with SUS isoforms is as follows:
  • SrSus4>SrSus1-Untagged>SrSus6>SrSus2>SrSus1>6×His-SrSus1>SrSus3
  • Example 13: Improved In Vitro Catalysis of Cannabinoid-Glycosides
  • As the formation of cannabinoid glycosides via UGT enzyme requires the nucleotide sugar donor UDPG in stoichiometric amounts, it is advantageous to recycle or recapture the spent UDP following a glycosylation reaction. Utilizing the SUS4 isoform from Stevia rebaudiana, cannabinoid glycosides were successfully produced using only UMP as the input nucleotide.
  • A two step reaction took place, first to produce UDP from UMP, and second to produce UDPG from the UDP in tandem with the UGT reaction. First, a 5L reaction containing 50 mM KPO4 pH7.2, 200 mM UMP disodium salt, 200 mM ATP disodium salt, 1M MgCl2, 10% UMPK recombinant enzyme in 50% glycerol was prepared. The reaction was incubated at 28 C with stirring for >24 hours. The 5L reaction 1 was filtered at 0.45 microns to remove precipitate then applied to a 50 L reaction containing 50 mM KPO4 pH7.2, 50 mM MgCl2, 300 mM Sucrose, 200 mg of CBD in 200 ml DMSO, 5L UGT76G1 in 50% glycerol, 2.5L SrSUS4 in 50% glycerol. The main 50 L reaction was then mixed and allowed to react. An additional 200 mg of CBD in 200 ml DMSO was added after the reaction went to completion, and allowed to continue incubating at the same conditions. After the remaining CBD was consumed by the reaction, the mixture was filtered by tangential flow filtration with a ultrafiltration membrane at 5 kDa to remove enzymes and particulate, and then concentrated using nanofiltration membrane at 500 Da. The nanofiltration retentate containing the cannabosides was then applied to hydrated C18 flash columns, washed with 10-30% methanol, and eluted with 40-65% methanol. The eluate was then concentrated by rotary evaporation to remove all solvent, shell-frozen in a vacuum beaker and lyophilized to dryness. The powdered cannabosides produced were then collected and stored at −20 C in sealed vials. Sucrose should be sterile filtered to avoid carmelization or sugar breakdown, as autoclaving sucrose stock solutions greatly decreases reaction activity.
  • TABLE 1
    Cannabidiol-glycoside compositions by R-group
    R-group location is as depicted in FIG. 1B
    VB # 1° 1-O Position 2° 2-O- 2° 3-O- 3° 3-O- 1° Position 2° 2-O- 2° 3-O-
    VB101 R1 = H R2 = H
    VB102 R1 = β-D-glucose H R2 = H
    VB103 R1 = β-D-glucose β-D-glucose R2 = H
    VB104 R1 = β-D-glucose H β-D-glucose R2 = H
    VB105 R1 = β-D-glucose β-D-glucose β-D-glucose R2 = H
    VB106 R1 = H R2 = β-D-glucose
    VB107 R1 = H R2 = β-D-glucose β-D-glucose
    VB108 R1 = H R2 = β-D-glucose H β-D-glucose
    VB109 R1 = H R2 = β-D-glucose β-D-glucose β-D-glucose
    VB110 R1 = β-D-glucose R2 = β-D-glucose
    VB111 R1 = β-D-glucose β-D-glucose R2 = β-D-glucose
    VB112 R1 = β-D-glucose H β-D-glucose R2 = β-D-glucose
    VB113 R1 = β-D-glucose β-D-glucose β-D-glucose R2 = β-D-glucose
    VB114 R1 = β-D-glucose R2 = β-D-glucose β-D-glucose
    VB115 R1 = β-D-glucose β-D-glucose R2 = β-D-glucose β-D-glucose
    VB116 R1 = β-D-glucose H β-D-glucose R2 = β-D-glucose β-D-glucose
    VB117 R1 = β-D-glucose β-D-glucose β-D-glucose R2 = β-D-glucose β-D-glucose
    VB118 R1 = β-D-glucose R2 = β-D-glucose H β-D-glucose
    VB119 R1 = β-D-glucose H β-D-glucose R2 = β-D-glucose H β-D-glucose
    VB120 R1 = β-D-glucose β-D-glucose R2 = β-D-glucose H β-D-glucose
    VB121 R1 = β-D-glucose β-D-glucose β-D-glucose R2 = β-D-glucose H β-D-glucose
    VB122 R1 = β-D-glucose R2 = β-D-glucose β-D-glucose β-D-glucose
    VB123 R1 = β-D-glucose β-D-glucose R2 = β-D-glucose β-D-glucose β-D-glucose
    VB124 R1 = β-D-glucose H β-D-glucose R2 = β-D-glucose β-D-glucose β-D-glucose
    VB125 R1 = β-D-glucose β-D-glucose β-D-glucose R2 = β-D-glucose β-D-glucose β-D-glucose
    VB126 R1 = α-D-glucose R2 = H
    VB127 R1 = H R2 = α-D-glucose
    VB128 R1 = α-D-glucose R2 = α-D-glucose
    VB129 R1 = β-D-glucose α-D-glucose R2 = H
    VB130 R1 = H R2 = β-D-glucose α-D-glucose
    VB131 R1 = β-D-glucose α-D-glucose R2 = β-D-glucose α-D-glucose
    VB132 R1 = β-D-glucose β-D-glucose β-D-glucose R2 = H
    VB133 R1 = H R2 = β-D-glucose H β-D-glucose
    VB134 R1 = β-D-glucose H β-D-glucose β-D-glucose R2 = β-D-glucose H β-D-glucose
    VB # 3° 3-O- Name 1° Enzyme 2° Enzyme
    VB101 Cannabidiol (CBD)
    VB102 CBD-1-O-glucopyranoside UGT76G1
    VB103 CBD-1-O-(2-1)-diglucopyranoside UGT76G1 Os03g0702000
    VB104 CBD-1-O-(3-1)-diglucopyranoside UGT76G1
    VB105 CBD-1-O-(2-1,3-1)-triglucopyranoside UGT76G1 Os03g0702000
    VB106 CBD-2-O-glucopyranoside UGT76G1
    VB107 CBD-2-O-(2-1)-diglucopyranoside UGT76G1 Os03g0702000
    VB108 CBD-2-O-(3-1)-diglucopyranoside UGT76G1
    VB109 CBD-1-O-(2-1,3-1)-triglucopyranoside UGT76G1 Os03g0702000
    VB110 CBD-1,2-O-diglucopyranoside UGT76G1
    VB111 CBD-1-O-(2-1), 2-O-triglucopyranoside UGT76G1 Os03g0702000
    VB112 CBD-1-O-(3-1), 2-O-triglucopyranoside UGT76G1
    VB113 CBD-1-O-(2-1,3-1), 2-O-tetraglucopyranoside UGT76G1 Os03g0702000
    VB114 CBD-1-O, 2-O-(2-1)-triglucopyranoside UGT76G1 Os03g0702000
    VB115 CBD-1-O-(2-1), 2-O-(2-1)-tetraglucopyranoside UGT76G1 Os03g0702000
    VB116 CBD-1-O-(3-1), 2-O-(2-1)-tetraglucopyranoside UGT76G1 Os03g0702000
    VB117 CBD-1-O-(2-1,3-1), 2-O-(2-1)-pentaglucopyranoside UGT76G1 Os03g0702000
    VB118 CBD-1-O, 2-O-(3-1)-triglucopyranoside UGT76G1
    VB119 CBD-1-O-(3-1), 2-O-(3-1)-tetraglucopyranoside UGT76G1
    VB120 CBD-1-O-(2-1), 2-O-(3-1)-tetraglucopyranoside UGT76G1 Os03g0702000
    VB121 CBD-1-O-(2-1,3-1), 2-O-(3-1)-pentaglucopyranoside UGT76G1 Os03g0702000
    VB122 CBD-1-O, 2-O-(2-1,3-1)-tetraglucopyranoside UGT76G1 Os03g0702000
    VB123 CBD-1-O-(2-1), 2-O-(2-1,3-1)-pentaglucopyranoside UGT76G1 Os03g0702000
    VB124 CBD-1-O-(3-1), 2-O-(2-1,3-1)-pentaglucopyranoside UGT76G1 Os03g0702000
    VB125 CBD-1-O-(2-1,3-1), 2-O-(2-1,3-1)-hexaglucopyranoside UGT76G1 Os03g0702000
    VB126 CBD-1-O-α-glucopyranoside CGTase
    VB127 CBD-2-O-α-glucopyranoside CGTase
    VB128 CBD-1,2-O-α-glucopyranoside CGTase
    VB129 CBD-1-O-β-primed-α-diglucopyranoside UGT76G1 CGTase
    VB130 CBD-2-O-β-primed-α-diglucopyranoside UGT76G1 CGTase
    VB131 CBD-1,2-O-β-primed-α-diglucopyranoside UGT76G1 CGTase
    VB132 CBD-1-O-(3-1,3-1)-triglucopyranoside UGT76G1
    VB133 β-D-glucose CBD-2-O-(3-1,3-1)-triglucopyranoside UGT76G1
    VB134 β-D-glucose CBD-1,2-O-(3-1,3-1)-hexaglucopyranoside UGT76G1
  • TABLE 2
    Cannabidivarin-glycoside compositions by R-group
    R-group location is as depicted in FIG. 1B
    VB # 1° 1-O Position 2° 2-O- 2° 3-O- 3° 3-O- 1° Position 2° 2-O- 2° 3-O-
    VB201 R1 = H R2 = H
    VB202 R1 = β-D-glucose R2 = H
    VB203 R1 = β-D-glucose β-D-glucose R2 = H
    VB204 R1 = β-D-glucose H β-D-glucose R2 = H
    VB205 R1 = β-D-glucose β-D-glucose β-D-glucose R2 = H
    VB206 R1 = H R2 = β-D-glucose
    VB207 R1 = H R2 = β-D-glucose β-D-glucose
    VB208 R1 = H R2 = β-D-glucose H β-D-glucose
    VB209 R1 = H R2 = β-D-glucose β-D-glucose β-D-glucose
    VB210 R1 = β-D-glucose R2 = β-D-glucose
    VB211 R1 = β-D-glucose β-D-glucose R2 = β-D-glucose
    VB212 R1 = β-D-glucose H β-D-glucose R2 = β-D-glucose
    VB213 R1 = β-D-glucose β-D-glucose β-D-glucose R2 = β-D-glucose
    VB214 R1 = β-D-glucose R2 = β-D-glucose β-D-glucose
    VB215 R1 = β-D-glucose β-D-glucose R2 = β-D-glucose β-D-glucose
    VB216 R1 = β-D-glucose H β-D-glucose R2 = β-D-glucose β-D-glucose
    VB217 R1 = β-D-glucose β-D-glucose β-D-glucose R2 = β-D-glucose β-D-glucose
    VB218 R1 = β-D-glucose R2 = β-D-glucose H β-D-glucose
    VB219 R1 = β-D-glucose H β-D-glucose R2 = β-D-glucose H β-D-glucose
    VB220 R1 = β-D-glucose β-D-glucose R2 = β-D-glucose H β-D-glucose
    VB221 R1 = β-D-glucose β-D-glucose β-D-glucose R2 = β-D-glucose H β-D-glucose
    VB222 R1 = β-D-glucose R2 = β-D-glucose β-D-glucose β-D-glucose
    VB223 R1 = β-D-glucose β-D-glucose R2 = β-D-glucose β-D-glucose β-D-glucose
    VB224 R1 = β-D-glucose H β-D-glucose R2 = β-D-glucose β-D-glucose β-D-glucose
    VB225 R1 = β-D-glucose β-D-glucose β-D-glucose R2 = β-D-glucose β-D-glucose β-D-glucose
    VB226 R1 = α-D-glucose R2 = H
    VB227 R1 = H R2 = α-D-glucose
    VB228 R1 = α-D-glucose R2 = α-D-glucose
    VB229 R1 = β-D-glucose α-D-glucose R2 = H
    VB230 R1 = H R2 = β-D-glucose α-D-glucose
    VB231 R1 = β-D-glucose α-D-glucose R2 = β-D-glucose α-D-glucose
    VB232 R1 = β-D-glucose H β-D-glucose β-D-glucose R2 = H
    VB233 R1 = H R2 = β-D-glucose H β-D-glucose
    VB234 R1 = β-D-glucose H β-D-glucose β-D-glucose R2 = β-D-glucose H β-D-glucose
    VB # 3° 3-O- Name 1° Enzyme 2° Enzyme
    VB201 Cannabidivarin (CBDV)
    VB202 CBDV-1-O-glucopyranoside UGT76G1
    VB203 CBDV-1-O-(2-1)-diglucopyranoside UGT76G1 Os03g0702000
    VB204 CBDV-1-O-(3-1)-diglucopyranoside UGT76G1
    VB205 CBDV-1-O-(2-1,3-1)-triglucopyranoside UGT76G1 Os03g0702000
    VB206 CBDV-2-O-glucopyranoside UGT76G1
    VB207 CBDV-2-O-(2-1)-diglucopyranoside UGT76G1 Os03g0702000
    VB208 CBDV-2-O-(3-1)-diglucopyranoside UGT76G1
    VB209 CBDV-1-O-(2-1,3-1)-triglucopyrancoside UGT76G1 Os03g0702000
    VB210 CBDV-1,2-O-diglucopyranoside UGT76G1
    VB211 CBDV-1-O-(2-1), 2-O-triglucopyranoside UGT76G1 Os03g0702000
    VB212 CBDV-1-O-(3-1), 2-O-triglucopyranoside UGT76G1
    VB213 CBDV-1-O-(2-1,3-1), 2-O-tetraglucopyranoside UGT76G1 Os03g0702000
    VB214 CBDV-1-O, 2-O-(2-1)-triglucopyranoside UGT76G1 Os03g0702000
    VB215 CBDV-1-O-(2-1), 2-O-(2-1)-tetraglucopyranoside UGT76G1 Os03g0702000
    VB216 CBDV-1-O-(3-1), 2-O-(2-1)-tetraglucopyranoside UGT76G1 Os03g0702000
    VB217 CBDV-1-O-(2-1,3-1), 2-O-(2-1)-pentaglucopyranoside UGT76G1 Os03g0702000
    VB218 CBDV-1-O, 2-O-(3-1)-triglucopyranoside UGT76G1
    VB219 CBDV-1-O-(3-1), 2-O-(3-1)-tetraglucopyranoside UGT76G1
    VB220 CBDV-1-O-(2-1), 2-O-(3-1)-tetraglucopyranoside UGT76G1 Os03g0702000
    VB221 CBDV-1-O-(2-1,3-1), 2-O-(3-1)-pentaglucopyranoside UGT76G1 Os03g0702000
    VB222 CBDV-1-O, 2-O-(2-1,3-1)-tetraglucopyranoside UGT76G1 Os03g0702000
    VB223 CBDV-1-O-(2-1), 2-O-(2-1,3-1)-pentaglucopyranoside UGT76G1 Os03g0702000
    VB224 CBDV-1-O-(3-1), 2-O-(2-1,3-1)-pentaglucopyranoside UGT76G1 Os03g0702000
    VB225 CBDV-1-O-(2-1,3-1), 2-O-(2-1,3-1)-hexaglucopyranoside UGT76G1 Os03g0702000
    VB226 CBDV-1-O-α-glucopyranoside CGTase
    VB227 CBDV-2-O-α-glucopyranoside CGTase
    VB228 CBDV-1,2-O-α-glucopyranoside CGTase
    VB229 CBDV-1-O-β-primed-α-diglucopyranoside UGT76G1 CGTase
    VB230 CBDV-2-O-β-primed-α-diglucopyranoside UGT76G1 CGTase
    VB231 CBDV-1,2-O-β-primed-α-diglucopyranoside UGT76G1 CGTase
    VB232 CBDV-1-O-(3-1,3-1)-triglucopyranoside UGT76G1
    VB233 β-D-glucose CBDV-2-O-(3-1,3-1)-triglucopyranoside UGT76G1
    VB234 β-D-glucose CBDV-1,2-O-(3-1,3-1)-hexaglucopyranoside UGT76G1
  • TABLE 3
    Δ9-Tetrahydrocannabinol-glycoside compositions by R-group
    R-group location is as depicted in FIG. 1B
    VB#
    1° 1-O Position 2° 2-O- 2° 3-O- 3° 3-O- Name Enzyme 2° Enzyme
    VB301 R1 = H Δ9-Tetrahydrocannabinol
    VB302 R1 = β-D-glucose Δ9THC-1-O-glucopyranoside UGT76G1
    VB303 R1 = β-D-glucose β-D-glucose Δ9THC-1-O-(2-1)-diglucopyranoside UGT76G1 Os03g0702000
    VB304 R1 = β-D-glucose H β-D-glucose Δ9THC-1-O-(3-1)-diglucopyraonside UGT76G1
    VB305 R1 = β-D-glucose β-D-glucose β-D-glucose Δ9THC-1-O-(2-1,3-1)-triglucopyranoside UGT76G1 Os03g0702000
    VB306 R1 = α-D-glucose Δ9THC-1-O-α-glucopyranoside CGTase
    VB307 R1 = β-D-glucose α-D-glucose Δ9THC-1-O-β-primed-α-diglucopyranoside UGT76G1 CGTase
    VB308 R1 = β-D-glucose H β-D-glucose β-D-glucose Δ9THC-1-O-(3-1,3-1)-triglucopyranoside UGT76G1
  • TABLE 4
    -Cannabinol-glycoside compositions by R-group
    R-group location is as depicted in FIG. 1B
    VB#
    1° 1-O Position 2° 2-O- 2° 3-O- 3° 3-O- Name Enzyme 2° Enzyme
    VB401 R1 = H Cannabinol
    VB402 R1 = β-D-glucose CBN-1-O-glucopyranoside UGT76G1
    VB403 R1 = β-D-glucose β-D-glucose CBN-1-O-(2-1)-diglucopyranoside UGT76G1 Os03g0702000
    VB404 R1 = β-D-glucose H β-D-glucose CBN-1-O-(3-1)-diglucopyranoside UGT76G1
    VB405 R1 = β-D-glucose β-D-glucose β-D-glucose CBN-1-O-(2-1,3-1)-triglucopyranoside UGT76G1 Os03g0702000
    VB406 R1 = α-D-glucose CBN-1-O-α-glucopyranoside CGTase
    VB407 R1 = β-D-glucose α-D-glucose CBN-1-O-β-primed-α-diglucopyranoside UGT76G1 CGTase
    VB408 R1 = β-D-glucose H β-D-glucose β-D-glucose CBN-1-O-(3-1,3-1)-triglucopyranoside UGT76G1
  • TABLE 5
    Anandamide (AEA) glycoside Compositions by R-group
    R-group location is as depicted in FIG. 1B
    VB#
    1° 1-O Position 2° 2-O- 2° 3-O- 3° 3-O- Name Enzyme 2° Enzyme
    VB501 R1 = H Anandamide (AEA)
    VB502 R1 = β-D-glucose AEA-1-O-glucopyranoside UGT76G1
    VB503 R1 = β-D-glucose H β-D-glucose AEA-1-O-(3-1)-diglucopyranoside UGT76G1
    VB504 R1 = β-D-glucose β-D-glucose AEA-1-O-(2-1)-diglucopyranoside UGT76G1 Os03g0702000
    VB505 R1 = β-D-glucose H β-D-glucose β-D-glucose AEA-1-O-(3-1,3-1)-triglucopyranoside UGT76G1
    VB506 R1 = β-D-glucose β-D-glucose β-D-glucose AEA-1-O-(3-1,2-1)-triglucopyranoside UGT76G1 Os03g0702000
    VB507 R1 = α-D-glucose AEA-1-O-α-glucopyranoside CGTase
    VB508 R1 = β-D-glucose α-D-glucose AEA-1-O-β-primed-α-diglucopyranoside UGT76G1 CGTase
  • TABLE 6
    2-Arachidonoylglycerol (2-AG)-glycoside compositions by R-group
    R-group location is as depicted in FIG. 1B
    VB# 1° 1-O Position 2° 2-O- 2° 3-O- 3° 3-O- 2° Position 2° 2-O- 2° 3-O-
    VB601 R1 = H R2 = H
    VB602 R1 = β-D-glucose R2 = H
    VB603 R1 = H R2 = β-D-glucose
    VB604 R1 = β-D-glucose β-D-glucose R2 = H
    VB605 R1 = β-D-glucose H β-D-glucose R2 = H
    VB606 R1 = H R2 = β-D-glucose β-D-glucose
    VB607 R1 = H R2 = β-D-glucose H β-D-glucose
    VB608 R1 = β-D-glucose R2 = β-D-glucose
    VB609 R1 = β-D-glucose R2 = β-D-glucose H β-D-glucose
    VB610 R1 = β-D-glucose H β-D-glucose R2 = β-D-glucose
    VB611 R1 = β-D-glucose R2 = β-D-glucose β-D-glucose
    VB612 R1 = β-D-glucose H β-D-glucose β-D-glucose R2 = H
    VB613 R1 = H R2 = β-D-glucose H β-D-glucose
    VB614 R1 = β-D-glucose β-D-glucose R2 = β-D-glucose
    VB615 R1 = β-D-glucose H β-D-glucose R2 = β-D-glucose H β-D-glucose
    VB616 R1 = β-D-glucose β-D-glucose R2 = β-D-glucose H β-D-glucose
    VB617 R1 = β-D-glucose H β-D-glucose R2 = β-D-glucose β-D-glucose
    VB618 R1 = β-D-glucose β-D-glucose R2 = β-D-glucose β-D-glucose
    VB619 R1 = α-D-glucose R2 = H
    VB620 R1 = β-D-glucose α-D-glucose R2 = H
    VB621 R1 = H R2 = α-D-glucose
    VB622 R1 = H R2 = β-D-glucose α-D-glucose
    VB623 R1 = α-D-glucose R2 = α-D-glucose
    VB# 3° 3-O- Name 1° Enzyme 2° Enzyme
    VB601 2-Arachidonoylglycerol (2AG)
    VB602 2-AG-1-O-glucopyranoside UGT76G1
    VB603 2-AG-2-O-glucopyranoside UGT76G1
    VB604 2-AG-1-O-(3-1)-diglucopyranoside UGT76G1
    VB605 2-AG-1-O-(2-1)-diglucopyranoside UGT76G1 Os03g0702000
    VB606 2-AG-2-O-(3-1)-diglucopyranoside UGT76G1
    VB607 2-AG-2-O-(2-1)-diglucopyranoside UGT76G1 Os03g0702000
    VB608 2-AG-1-O,2-O-diglucopyranoside UGT76G1
    VB609 2-AG-1-O, 2-O-(3-1)-triglucopyranoside UGT76G1
    VB610 2-AG-1-O-(3-1), 2-O-triglucopyranoside UGT76G1
    VB611 2-AG-1-O, 2-O-(2-1)-triglucopyranoside UGT76G1 Os03g0702000
    VB612 2-AG-1-O-(3-1, 3-1)-triglucopyranoside UGT76G1
    VB613 β-D-glucose 2-AG-2-O-(3-1, 3-1)-triglucopyranoside UGT76G1
    VB614 2-AG-1-O-(2-1), 2-O-triglucopyranoside UGT76G1 Os03g0702000
    VB615 2-AG-1-O-(3-1), 2-O-(3-1)-tetraglucopyranoside UGT76G1
    VB616 2-AG-1-O-(2-1), 2-O-(3-1)-tetraglucopyranoside UGT76G1 Os03g0702000
    VB617 2-AG-1-O-(3-1), 2-O-(2-1)-tetraglucopyranoside UGT76G1 Os03g0702000
    VB618 2-AG-1-O-(2-1), 2-O-(2-1)-tetraglucopyranoside UGT76G1 Os03g0702000
    VB619 2-AG-1-O-α-glucopyranoside CGTase
    VB620 2-AG-1-O-β-primed-α-diglucopyranoside UGT76G1 CGTase
    VB621 2-AG-2-O-α-glucopyranoside CGTase
    VB622 2-AG-2-O-β-primed-α-diglucopyranoside UGT76G1 CGTase
    VB623 2-AG-1,2-O-α-diglucopyranoside CGTase
  • TABLE 7
    1-Arachidonoylglycerol (1-AG)-glycoside compositions by R-group
    R-group location is as depicted in FIG. 1B
    VB# 1° 1-O Position 2° 2-O- 2° 3-O- 3° 3-O- 2° Position 2° 2-O- 2° 3-O-
    VB701 R1 = H R2 = H
    VB702 R1 = β-D-glucose R2 = H
    VB703 R1 = H R2 = β-D-glucose
    VB704 R1 = β-D-glucose β-D-glucose R2 = H
    VB705 R1 = β-D-glucose H β-D-glucose R2 = H
    VB706 R1 = H R2 = β-D-glucose β-D-glucose
    VB707 R1 = H R2 = β-D-glucose H β-D-glucose
    VB708 R1 = β-D-glucose R2 = β-D-glucose
    VB709 R1 = β-D-glucose R2 = β-D-glucose H β-D-glucose
    VB710 R1 = β-D-glucose H β-D-glucose R2 = β-D-glucose
    VB711 R1 = β-D-glucose R2 = β-D-glucose β-D-glucose
    VB712 R1 = β-D-glucose β-D-glucose R2 = β-D-glucose
    VB713 R1 = β-D-glucose H β-D-glucose β-D-glucose R2 = H
    VB714 R1 = H R2 = β-D-glucose H β-D-glucose
    VB715 R1 = β-D-glucose β-D-glucose R2 = β-D-glucose H β-D-glucose
    VB716 R1 = β-D-glucose β-D-glucose R2 = β-D-glucose H β-D-glucose
    VB717 R1 = β-D-glucose H β-D-glucose R2 = β-D-glucose β-D-glucose
    VB718 R1 = β-D-glucose β-D-glucose R2 = β-D-glucose β-D-glucose
    VB719 R1 = α-D-glucose R2 = H
    VB720 R1 = β-D-glucose α-D-glucose R2 = H
    VB721 R1 = H R2 = α-D-glucose
    VB722 R1 = H R2 = β-D-glucose α-D-glucose
    VB723 R1 = α-D-glucose R2 = α-D-glucose
    VB# 3° 3-O- Name 1° Enzyme 2° Enzyme
    VB701 1-Arachidonoylglycerol (1-AG)
    VB702 1-AG-1-O-glucopyranoside UGT76G1
    VB703 1-AG-2-O-glucopyranoside UGT76G1
    VB704 1-AG-1-O-(2-1)-diglucopyranoside UGT76G1
    VB705 1-AG-1-O-(3-1)-diglucopyranoside UGT76G1 Os03g0702000
    VB706 1-AG-2-O-(2-1)-diglucopyranoside UGT76G1
    VB707 1-AG-2-O-(3-1)-diglucopyranoside UGT76G1 Os03g0702000
    VB708 1-AG-1-O,2-O-diglucopyranoside UGT76G1
    VB709 1-AG-1-O, 2-O-(3-1)-triglucopyranoside UGT76G1
    VB710 1-AG-1-O-(3-1), 2-O-triglucopyranoside UGT76G1
    VB711 1-AG-1-O, 2O-(2-1)-triglucopyranoside UGT76G1 Os03g0702000
    VB712 1-AG-1-O-(2-1), 2-O-triglucopyranoside UGT76G1 Os03g0702000
    VB713 1-AG-1-O-(3-1, 3-1)-triglucopyranoside UGT76G1
    VB714 β-D-glucose 1-AG-2-O-(3-1, 3-1)-triglucopyranoside UGT76G1
    VB715 1-AG-1-O-(3-1), 2-O-(3-1)-tetraglucopyranoside UGT76G1
    VB716 1-AG-1-O-(2-1), 2-O-(3-1)-tetraglucopyranoside UGT76G1 Os03g0702000
    VB717 1-AG-1-O-(3-1), 2-O-(2-1)-tetraglucopyranoside UGT76G1 Os03g0702000
    VB718 1-AG-1-O-(2-1), 2-O-(2-1)-tetraglucopyranoside UGT76G1 Os03g0702000
    VB719 1-AG-1-O-α-glucopyranoside CGTase
    VB720 1-AG-1-O-β-primed-α-diglucopyranoside UGT76G1 CGTase
    VB721 1-AG-2-O-α-glucopyranoside CGTase
    VB722 1-AG-2-O-β-primed-α-diglucopyranoside UGT76G1 CGTase
    VB723 1-AG-1,2-O-α-diglucopyranoside CGTase
  • TABLE 8
    Docosahexaenoyl ethanoloamide (DHEA) glycoside compositions by R-group
    R-group location is as depicted in FIG. 1B
    VB#
    1° 1-O Position 2° 2-O- 2° 3-O- 3° 3-O- Name Enzyme 2° Enzyme
    VB801 R1 = H Docosahexaenoyl ethanoloamide (DHEA)
    VB802 R1 = β-D-glucose DHEA-1-O-glucopyranoside UGT76G1
    VB803 R1 = β-D-glucose H β-D-glucose DHEA-1-O-(3-1)-diglucopyranoside UGT76G1
    VB804 R1 = β-D-glucose β-D-glucose DHEA-1-O-(2-1)-diglucopyranoside UGT76G1 Os03g0702000
    VB805 R1 = β-D-glucose H β-D-glucose β-D-glucose DHEA-1-O-(3-1,3-1)-triglucopyranoside UGT76G1
    VB806 R1 = α-D-glucose β-D-glucose β-D-glucose DHEA-1-O-(3-1,2-1)-triglucopyranoside UGT76G1 Os03g0702000
    VB807 R1 = α-D-glucose DHEA-1-O-α-glucopyranoside CGTase
    VB808 R1 = β-D-glucose α-D-glucose DHEA-1-O-β-primed-α-diglucopyranoside UGT76G1 CGTase
  • TABLE 9
    Capsiacin glycoside compositions by R-group
    R-group location is as depicted in FIG. 1B
    VB#
    1° 1-O Position 2° 2-O- 2° 3-O- 3° 3-O- Name Enzyme 2° Enzyme
    VB901 R1 = H Capsalcin
    VB902 R1 = β-D-glucose Capsalcin-1-O-glucopyranoside UGT76G1
    VB903 R1 = β-D-glucose H β-D-glucose Capsalcin-1-O-(3-1)-diglcopyranoside UGT76G1
    VB904 R1 = β-D-glucose β-D-glucose Capsalcin-1-O-(2-1)-diglcopyranoside UGT76G1 Os03g0702000
    VB905 R1 = β-D-glucose H β-D-glucose β-D-glucose Capsalcin-1-O-(3-1, 3-1)- UGT76G1
    triglucopyranoside
    VB906 R1 = β-D-glucose β-D-glucose β-D-glucose Capsalcin-1-O-(2-1,3-1)- UGT76G1 Os03g0702000
    triglucopyranoside
    VB907 R1 = α-D-glucose Capslacin-1-O-α-glucopyranoside CGTase
    VB908 R1 = β-D-glucose α-D-glucose Capslacin-1-O-β-primed-α- UGT76G1 CGTase
    diglucopyranoside
  • TABLE 10
    Vanillin glycoside compositions by R-group
    R-group location is as depicted in FIG. 1B
    VB#
    1° 1-O Position 2° 2-O- 2° 3-O- 3° 3-O- Name Ezyme 2° Enzyme
    VB1001 R1 = H Vanillin
    VB1002 R1 = β-D-glucose Vanillin-1-O-glucopyranoside UGT76G1
    VB1003 R1 = β-D-glucose H β-D-glucose Vanillin-1-O-(3-1)-diglcopyranoside UGT76G1
    VB1004 R1 = β-D-glucose β-D-glucose Vanillin-1-O-(2-1)-diglcopyranoside UGT76G1 Os03g0702000
    VB1005 R1 = β-D-glucose β-D-glucose β-D-glucose Vanillin-1-O-(2-1,3-1)-triglucopyranoside UGT76G1 Os03g0702000
    VB1006 R1 = β-D-glucose H β-D-glucose β-D-glucose Vanillin-1-O-(3-1,3-1)-triglucopyranoside UGT76G1
    VB1007 R1 = α-D-glucose Vanillin-1-O-α-glucopyranoside CGTase
    VB1008 R1 = β-D-glucose α-D-glucose Vanillin-1-O-β-primed-α-diglucopyranoside UGT76G1 CGTase
  • TABLE 11
    Curcumin glycoside compositions by R-group
    R-group location is as depicted in FIG. 1B
    VB# 1° 1-O Postition 2° 2-O- 2° 3-O- 3° 3-O- 2° Position 2° 2-O- 2° 3-O-
    VB1101 R1 = H R2 = H
    VB1102 R1 = β-D-glucose R2 = H
    VB1103 R1 = H R2 = β-D-glucose
    VB1104 R1 = β-D-glucose H β-D-glucose R2 = H
    VB1105 R1 = β-D-glucose β-D-glucose R2 = H
    VB1106 R1 = H R2 = β-D-glucose H β-D-glucose
    VB1107 R1 = H R2 = β-D-glucose β-D-glucose
    VB1108 R1 = β-D-glucose R2 = β-D-glucose
    VB1109 R1 = β-D-glucose R2 = β-D-glucose H β-D-glucose
    VB1110 R1 = β-D-glucose H β-D-glucose R2 = β-D-glucose H
    VB1111 R1 = β-D-glucose H R2 = β-D-glucose β-D-glucose
    VB1112 R1 = β-D-glucose β-D-glucose R2 = β-D-glucose
    VB1113 R1 = β-D-glucose β-D-glucose β-D-glucose R2 = H
    VB1114 R1 = H R2 = β-D-glucose β-D-glucose β-D-glucose
    VB1115 R1 = H R2 = β-D-glucose H β-D-glucose
    VB1116 R1 = β-D-glucose H β-D-glucose β-D-glucose R2 = H
    VB1117 R1 = β-D-glucose H β-D-glucose R2 = β-D-glucose H β-D-glucose
    VB1118 R1 = β-D-glucose β-D-glucose R2 = β-D-glucose H β-D-glucose
    VB1119 R1 = β-D-glucose H β-D-glucose R2 = β-D-glucose β-D-glucose
    VB1120 R1 = β-D-glucose β-D-glucose R2 = β-D-glucose β-D-glucose
    VB1121 R1 = β-D-glucose H β-D-glucose β-D-glucose R2 = β-D-glucose
    VB1122 R1 = β-D-glucose β-D-glucose β-D-glucose R2 = β-D-glucose
    VB1123 R1 = β-D-glucose R2 = β-D-glucose H β-D-glucose
    VB1124 R1 = β-D-glucose R2 = β-D-glucose β-D-glucose β-D-glucose
    VB1125 R1 = β-D-glucose H β-D-glucose R2 = β-D-glucose H β-D-glucose
    VB1126 R1 = β-D-glucose H β-D-glucose R2 = β-D-glucose β-D-glucose H
    VB1127 R1 = β-D-glucose H β-D-glucose β-D-glucose R2 = β-D-glucose H β-D-glucose
    VB1128 R1 = β-D-glucose β-D-glucose H β-D-glucose R2 = β-D-glucose H β-D-glucose
    VB1129 R1 = β-D-glucose H β-D-glucose β-D-glucose R2 = β-D-glucose H β-D-glucose
    VB1130 R1 = β-D-glucose β-D-glucose H β-D-glucose R2 = β-D-glucose H β-D-glucose
    VB1131 R1 = β-D-glucose H β-D-glucose β-D-glucose R2 = β-D-glucose β-D-glucose H
    VB1132 R1 = α-D-glucose R2 = H
    VB1133 R1 = β-D-glucose α-D-glucose R2 = H
    VB1134 R1 = H R2 = α-D-glucose
    VB1135 R1 = H R2 = β-D-glucose α-D-glucose
    VB1136 R1 = α-D-glucose R2 = α-D-glucose
    VB# 3° 3-O- Name 1° Enzyme 2° Enzyme
    VB1101 Curcumin
    VB1102 Curcimun-1-O-glucopyranoside UGT76G1
    VB1103 Curcumin-2-O-glucopyranoside UGT76G1
    VB1104 Curcumin-1-O-(3-1)-diglucopyranoside UGT76G1
    VB1105 Curcumin-1-O-(2-1)-diglucopyranoside UGT76G1 Os03g0702000
    VB1106 Curcumin-2-O-(3-1)-diglucopyranoside UGT76G1
    VB1107 Curcumin-2-O-(2-1)-diglucopyranoside UGT76G1 Os03g0702000
    VB1108 Curcumin-1-O, 2-O-diglucopyranoside UGT76G1
    VB1109 Curcumin-1-O, 2-O-(3-1)-triglucopyranoside UGT76G1
    VB1110 Curcumin-1-O-(3-1), 2-O-triglucopyranoside UGT76G1
    VB1111 Curcumin-1-O, 2-O-(2-1)-triglucopyranoside UGT76G1 Os03g0702000
    VB1112 Curcumin-1-O-(2-1), 2-O-triglucopyranoside UGT76G1 Os03g0702000
    VB1113 Curcumin-1-O-(2-1, 3-1)-triglucopyranoside UGT76G1 Os03g0702000
    VB1114 Curcumin-2-O-(2-1, 3-1)-triglucopyranoside UGT76G1 Os03g0702000
    VB1115 β-D-glucose Curcumin-2-O-(3-1, 3-1)-triglucopyranoside UGT76G1
    VB1116 Curcumin-1-O-(3-1, 3-1)-triglucopyranoside UGT76G1
    VB1117 Curcumin-1-O-(3-1), 2-O-(3-1)-tetraglucopyranoside UGT76G1
    VB1118 Curcumin-1-O-(2-1), 2-O-(3-1)-tetraglucopyranoside UGT76G1 Os03g0702000
    VB1119 Curcumin-1-O-(3-1), 2-O-(2-1)-tetraglucopyranoside UGT76G1 Os03g0702000
    VB1120 Curcumin-1-O-(2-1), 2-O-(2-1)-tetraglucopyranoside UGT76G1 Os03g0702000
    VB1121 Curcumin-1-O-(3-1, 3-1), 2-O-tetraglucopyranoside UGT76G1
    VB1122 Curcumin-1-O-(2-1, 3-1), 2-O-tetraglucopyranoside UGT76G1 Os03g0702000
    VB1123 β-D-glucose Curcumin-1-O, 2O-(3-1, 3-1)-tetraglucopyranoside UGT76G1
    VB1124 Curcumin-1-O, 2-O-(2-1, 3-1)-tetraglucopyranoside UGT76G1 Os03g0702000
    VB1125 β-D-glucose Curcumin-1-O-(3-1), 2-O-(3-1, 3-1)-pentaglucopyranoside UGT76G1
    VB1126 β-D-glucose Curcumin-1-O-(3-1), 2-O-(2-1, 3-1)-pentaglucopyranoside UGT76G1 Os03g0702000
    VB1127 Curcumin-1-O-(3-1, 3-1), 2-O-(3-1)-pentaglucopyranoside UGT76G1
    VB1128 Curcumin-1-O-(2-1,3-1), 2-O-(3-1)-pentaglucopyranoside UGT76G1 Os03g0702000
    VB1129 Curcumin-1-O-(3-1, 3-1), 2-O-(3-1, 3-1)-hexaglucopyranoside UGT76G1
    VB1130 β-D-glucose Curcumin-1-O-(2-1, 3-1), 2-O-(3-1, 3-1)-hexaglucopyranoside UGT76G1 Os03g0702000
    VB1131 β-D-glucose Curcumin-1-O-(3-1, 3-1), 2-O-(2-1, 3-1)-hexaglucopyranoside UGT76G1 Os03g0702000
    VB1132 β-D-glucose Curcumin-1-O-α-glucopyranoside CGTase
    VB1133 Curcumin-1-O-β-primed-α-diglucopyranoside UGT76G1 CGTase
    VB1134 Curcumin-2-O-α-glucopyranoside CGTase
    VB1135 Curcumin-2-O-β-primed-α-diglucopyranoside UGT76G1 CGTase
    VB1136 Curcumin-1,2-O-α-glucopyranoside CGTase
  • It is obvious that the foregoing embodiments of the invention are examples and can be varied in many ways. Such present or future variations are not to be regarded as a departure from the spirit and scope of the invention, and all such modifications as would be obvious to one skilled in the art are intended to be included within the scope of the following claims.
  • REFERENCES
    • Bartzokis G. (2004). Age-related myelin breakdown: a developmental model of cognitive decline and Alzheimer's disease. Neurobiology of Aging. 25:5-18.
    • Bisogno T, et al. (2001) Molecular targets for cannabidiol and its synthetic analogues: effect on vanilloid VR1 receptors and on the cellular uptake and enzymatic hydrolysis of anandamide. British Journal of Pharmacology. 134, 845-852.
    • Chen Q, et al. (2009). Synthesis, in vitro and in vivo characterization of glycosyl derivatives of ibuprofen as novel prodrugs for brain drug delivery. J Drug Targeting. 17(4):318-328. Conchie J., Findlay J., Levvy G A. (1958). Mammalian Glycosidases, Distribution in the body. Biochem J. 71(2):318-325.
    • De Petrocellis L, et al. (2011) Effects of cannabinoids and cannabinoid-enriched Cannabis extracts on TRP channels and endocannabinoid metabolic enzymes. British Journal of Pharmacology. 163, 1479-1494.
    • Dewitte G, et al. (2016) Screening of Recombinant Glycosyltransferases Reveals the Broad acceptor Specificity of Stevia UGT-76G1. Journal of Biotechnology. Accepted Manuscript, DOI: http://dx.doi.org/doi:10.1016/j.jbiotec.2016.06.034.
    • Friend D R., Chang G W. (1984). A Colon-Specific Drug-Delivery System Based on Drug Glycosides and the Glycosidases of the Colonic Bacteria. J Med Chem. 27:261-266. Friend D R., Chang G W. (1985). Drug Glycosides: Potential Prodrugs for Colon-Specific Drug Delivery. J Med Chem. 28:51-57.
    • Gomez O., Arevalo-Martin A., Garcia-Ovejero D., Ortega-Gutierrez S., Cisneros J A., Almazan G, Sanchez-Rodriguez M A., Molina-Holgado F., Molina-Holgado E. (2010). The Constitutive Production of the Endocannabinoid 2-Arachidonoylglycerol Participates in Oligodendrocyte Differentiation. Glia. 58:1913-1927.
    • Iuvone T., Esposito G., De Filippis D., Scuderi C., Steardo L. (2009). Cannabidiol: a promising drug for neurodegenerative disorders? CNS Neurosci Ther. 15(1):65-75.
    • Jarh, P., Pate D W., Brenneisen R., Jarvinen T. (1998). Hydroxypropyl-beta-cyclodextrin and its combination with hydroxypropyl-methylcellulose increases aqueous solubility of delta9-tetrahydrocannabinol. Life Sci. 63(26):PL381-384.
    • Jiang R, et al. (2011) Identification of cytochrome P450 enzymes responsible for metabolism of cannabidiol by human liver microsomes. Life Sciences. 89, 165-170.
    • Kren V (2008) Glycoside vs. Aglycon: The Role of Glycosidic Residue in Biologic Activity. Glycoscience. pp 2589-2644.
    • Kren V, Rezanka T (2008) Sweet antibiotics—the role of glycosidic residues in antibiotic and antitumor activity and their randomization. FEMS Microbiol Rev. 32, 858-889.
    • Li S., Li W., Xiao Q., Xia Y. (2012). Transglycosylation of stevioside to improve the edulcorant quality by lower substitution using cornstarch hydrolyzate and CGTase. J Food Chem. 138(2013):2064-2069.
    • Mazur A., et al. (2009). Characterization of Human Hepatic and Extrahepatic UDP-Glucuronosyltransferase Enzymes Involved in the Metabolism of Classic Cannabinoids. Drug Metabolism and Disposition. 37(7):1496-1504.
    • Mecha M., Torrao A S., Mestre L., Carrillo-Salinas F J., Mechoulam R., Guaza C. (2012). Cannabidiol protects oligodendrocyte progenitor cells from inflammation-induced apoptosis by attenuating endoplasmic reticulum stress. Cell Death and Disease. 3(e331).
    • Mechoulam R., Parker L A., Gallily R. (2002). Cannabidiol: An Overview of Some Pharmacological Aspects. 42(S1):11S-19S.
    • Mighdoll M I., Tao R., Kleinman J E., Hyde T M. (2015). Myelin, myelin-related disorders, and psychosis. Schizophr Res. 161(1):85-93.
    • Molina-Holgado E., Vela J M., Arevalo-Martin A., Almazan G., Molina-Holgado F., Borrell J., Guaza C. (2002). Cannabinoids Promote Oligodendrocyte Progenitor Survival: Involvement of Cannavinoid Receptors and Phosphatidylinositol-3-Kinase/Akt Signaling. J. Neurosci. 22(22):9742-9753.
    • Noguchi A, et al. (2009). Identification of an inducible glucosyltransferase from Phytolacca americana L. cells that are capable of glucosylating capsaicin. Plant Biotechnology. 26, 285-292.
    • Pacher P, et al. (2006) The endocannabinoid system as an emerging target of pharmacotherapy. Pharmacology Review. 58(3), 389-462.
    • Richman A., Swanso, A., Humphrey T., Chapman R., McGarvey B., Pocs R., Brandle J. (2005). Functional genomics uncovers three glucosyltransferases involved in the synthesis of the major sweet glucosides of Stevia rebaudiana. Plant J. 41(1):56-67.
    • Russo E., Guy, G W. (2006) A tale of two cannabinoids: the therapeutic rationale for combining tetrahydrocannabinol and cannabidiol. Medical Hypotheses. 66(2):234-46.
    • Tanaka H., et al. (1993). Cannabis, 21.1 Biotransformation of cannabinol to its glycosides by in vitro plant tissue. Journal of Natural Products. 56(12):2068-2072.
    • Tanaka H., et al. (1996). Cannabis 25, biotransformation of cannabidiol and cannabidiolic acid by Pinellia ternata tissue segments. Plant Cell Reports. 15:819-823.
    • Terao J., Murota K., Kawai Y. (2011). Conjugated quercetin glucuronides as bioactive metabolites and precursors of aglycone in vivo. Food Function. 2:11-17.
    • Thomas A., et al. (2007) Cannabidiol displays unexpectedly high potency as an antagonist of CB1 and CB2 receptor agonists in vitro. British Journal of Pharmacology. 150, 613-623.
    • U.S. Pat. No. 8,410,064 B2. 2013. Classical cannabinoid metabolites and methods of use thereof.
    • U.S. Pat. No. 8,227,627 B2. 2012. Prodrugs of tetrahydrocannabinol, compositions comprising prodrugs of tetrahyrocannabinol and methods of using the same.
    • Watanabe K, et al. (1998) Distribution and characterization of anandamide amidohydrolase in mouse brain and liver. Life Sciences. 62(14), 1223-1229.
    • WO2009018389 A4. 2009. Prodrugs of cannabidiol, compositions comprising prodrugs of cannabidiol and methods of using the same.
    • WO2012011112 A1. 2011. Non psychoactive cannabinoids and uses thereof.
    • WO 2014108899 A1. 2014. Fluorinated CBD compounds, compositions and uses thereof. Yamaori S, et al. (2011) Potent inhibition of human cytochrome P450 3A isoforms by cannabidiol: Role of phenolic hydroxyl groups in the resorcinol moiety. Life Sciences, 88, 730-736.
    • Zuardi A W, et al. (2012). A Critical Review of the Antipyschotic Effects of Cannabidiol: 30 Years of a Translational Investigation. Current Pharmaceutical Design, 18, 5131-5140.

Claims (33)

1. A cannabinoid glycoside prodrug compound having formula (I):
Figure US20220168428A1-20220602-C00036
wherein
R is H, β-D-glucopyranosyl, or 3-O-β-D-glucopyranosyl-β-D-glucopyranosyl;
R′ is H or β-D-glucopyranosyl, or 3-O-β-D-glucopyranosyl-β-D-glucopyranosyl; and
A is an aglycone moiety formed through reaction of a hydroxyl group on a cannabinoid compound, an endocannabinoid compound, or a vanilloid compound,
or a pharmaceutically compatible salt thereof.
2. A compound according to claim 1, wherein A is A′, A″ or A′″;
wherein A′ is:
Figure US20220168428A1-20220602-C00037
wherein A″ is:
Figure US20220168428A1-20220602-C00038
and
wherein A′″ is:
Figure US20220168428A1-20220602-C00039
wherein G is H, β-D-glucopyranosyl, 3-O-β-D-glucopyranosyl-β-D-glucopyranosyl, or β-D-glucopyranosyl-(1→3)-3-D-glucopyranosyl-(1→3)-D-glucopyranosyl.
3. A compound according to claim 2, wherein A is A′.
4.-11. (canceled)
12. A compound according to claim 2, wherein A is A″.
13.-20. (canceled)
21. A compound according to claim 2, wherein A is A′″.
22.-28. (canceled)
29. A pharmaceutical composition comprising a compound as defined in claim 1 and a pharmaceutically acceptable carrier, diluent, excipient, or adjuvant.
30. A method for the site-specific delivery of a cannabinoid drug to a subject, comprising the step of administering a cannabinoid glycoside prodrug as defined in claim 1 to a subject in need thereof.
31. The method of claim 30, wherein the cannabinoid glycoside prodrug is formulated for oral, parenteral, or transdermal administration.
32. (canceled)
33. (canceled)
34. A method for the site-specific delivery of a cannabinoid drug to a subject, comprising the step of administering a pharmaceutical composition as defined in claim 29 to a subject in need thereof.
35. The method of claim 34, wherein the pharmaceutical composition is formulated for oral, parenteral, or transdermal administration.
36. (canceled)
37. (canceled)
38. A method for facilitating the transport of a cannabinoid drug across the blood brain barrier of a subject comprising administering a cannabinoid glycoside prodrug as defined in claim 1 to a subject in need thereof.
39. An antimicrobial agent comprising an effective amount of a cannabinoid glycoside prodrug as defined in claim 1.
40. (canceled)
41. A detersive agent comprising an effective amount of a cannabinoid glycoside prodrug as defined in claim 1.
42. (canceled)
43. A method of producing a cannabinoid glycoside, comprising incubating a cannabinoid aglycone with one or more sugar donors in the presence of one or more glycosyltransferases, wherein the one or more glycosyltransferases is a UGT76G1 or UGT76G1-like glucosyltransferase.
44. (canceled)
45. The method of claim 43, wherein the one or more glycosyltransferases further comprise a Os03g0702000 or Os03g0702000-like glucosyltransferase.
46. The method of claim 43, wherein the one or more sugar donors are selected from the group consisting of UDP-glucose, UDP-glucuronic acid, UDP-mannose, UDP-fructose, UDP-xylose, UDP-rhamnose, UDP-fluoro-deoxyglucose and combinations thereof.
47.-52. (canceled)
53. The method of claim 43, wherein the cannabinoid glycoside produced by the method is a compound of the Formula (I):
Figure US20220168428A1-20220602-C00040
wherein
R is H, β-D-glucopyranosyl, or 3-O-β-D-glucopyranosyl-β-D-glucopyranosyl;
R′ is H or β-D-glucopyranosyl, or 3-O-β-D-glucopyranosyl-β-D-glucopyranosyl; and
A is an aglycone moiety formed through reaction of a hydroxyl group on a cannabinoid compound, an endocannabinoid compound, or a vanilloid compound,
or a pharmaceutically compatible salt thereof.
54. A method of producing a cannabinoid glycoside prodrug of claim 1, comprising incubating a cannabinoid aglycone with maltodextrin, in the presence of a cyclodextrin glucanotransferase under conditions that allow for glycosylation.
55. A method of producing a cannabinoid glycoside prodrug of claim 1, comprising incubating a cannabinoid aglycone with UDP-glucose and maltodextrin in the presence of a UGT76G1 or UGT76G1-like glucosyltransferase and cyclodextrin glucanotransferase under conditions which allow for glycosylation.
56.-59. (canceled)
60. A method producing a cannabinoid glycoside prodrug of claim 1, comprising expressing one or more of the glycosyltransferases in a cell or plant which produces a cannabinoid aglycone and isolating the cannabinoid glycoside.
61.-79. (canceled)
US17/527,685 2015-09-22 2021-11-16 Cannabinoid glycoside prodrugs and methods of synthesis Pending US20220168428A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US17/527,685 US20220168428A1 (en) 2015-09-22 2021-11-16 Cannabinoid glycoside prodrugs and methods of synthesis
US18/182,618 US20230346952A1 (en) 2015-09-22 2023-03-13 Cannabinoid glycoside prodrugs and methods of synthesis

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US201562222144P 2015-09-22 2015-09-22
US201562245928P 2015-10-23 2015-10-23
US201662363808P 2016-07-18 2016-07-18
PCT/US2016/053122 WO2017053574A1 (en) 2015-09-22 2016-09-22 Cannabinoid glycoside prodrugs and methods of synthesis
US201815762180A 2018-03-22 2018-03-22
US17/527,685 US20220168428A1 (en) 2015-09-22 2021-11-16 Cannabinoid glycoside prodrugs and methods of synthesis

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
US15/762,180 Division US11207414B2 (en) 2015-09-22 2016-09-22 Cannabinoid glycoside prodrugs and methods of synthesis
PCT/US2016/053122 Division WO2017053574A1 (en) 2015-09-22 2016-09-22 Cannabinoid glycoside prodrugs and methods of synthesis

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US18/182,618 Continuation-In-Part US20230346952A1 (en) 2015-09-22 2023-03-13 Cannabinoid glycoside prodrugs and methods of synthesis

Publications (1)

Publication Number Publication Date
US20220168428A1 true US20220168428A1 (en) 2022-06-02

Family

ID=58387412

Family Applications (2)

Application Number Title Priority Date Filing Date
US15/762,180 Active US11207414B2 (en) 2015-09-22 2016-09-22 Cannabinoid glycoside prodrugs and methods of synthesis
US17/527,685 Pending US20220168428A1 (en) 2015-09-22 2021-11-16 Cannabinoid glycoside prodrugs and methods of synthesis

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US15/762,180 Active US11207414B2 (en) 2015-09-22 2016-09-22 Cannabinoid glycoside prodrugs and methods of synthesis

Country Status (8)

Country Link
US (2) US11207414B2 (en)
EP (1) EP3352855A4 (en)
JP (1) JP2018528977A (en)
CN (1) CN108136208B (en)
AU (1) AU2016326518B2 (en)
BR (1) BR112018005639A2 (en)
CA (1) CA2999764A1 (en)
WO (1) WO2017053574A1 (en)

Families Citing this family (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021173130A1 (en) * 2020-02-26 2021-09-02 Vitality Biopharma, Inc. Novel cannabinoid glycosides and uses thereof
WO2017053574A1 (en) 2015-09-22 2017-03-30 Vitality Biopharma, Inc. Cannabinoid glycoside prodrugs and methods of synthesis
US10239808B1 (en) 2016-12-07 2019-03-26 Canopy Holdings, LLC Cannabis extracts
AU2018239671A1 (en) 2017-03-24 2019-10-10 Trait Biosciences, Inc. High level In vivo biosynthesis and isolation of water-soluble cannabinoids in plant systems
US11207291B2 (en) 2017-05-09 2021-12-28 Graphium Biosciences, Inc. Antimicrobial compositions comprising cannabinoids and methods of using the same
US11946059B2 (en) * 2017-07-11 2024-04-02 Trait Biosciences, Inc. In vivo generation of water-soluble cannabinoids in plant cell suspension cultures
WO2019014395A1 (en) 2017-07-11 2019-01-17 Trait Biosciences, Inc. Generation of water-soluble cannabinoid compounds in yeast and plant cell suspension cultures and compositions of matter
US11905543B2 (en) 2017-07-11 2024-02-20 Trait Biosceinces, Inc. In vivo generation of water-soluble acetylated cannabinoid glycoside compounds in plant cell suspension cultures
WO2019082171A1 (en) * 2017-10-27 2019-05-02 Alvit Pharma Oral cannabinoid compositions with improved bioavailability
KR20200104352A (en) * 2017-12-21 2020-09-03 파마시틱스 비.브이. How to improve the oral bioavailability of drugs
US11202771B2 (en) 2018-01-31 2021-12-21 Treehouse Biotech, Inc. Hemp powder
EP3856172A4 (en) * 2018-09-28 2022-10-05 Visceral Therapeutics Inc. Pharmaceutically active cannabis-based compositions and methods of use for treating gastrointestinal conditions
US11040932B2 (en) 2018-10-10 2021-06-22 Treehouse Biotech, Inc. Synthesis of cannabigerol
WO2020167751A1 (en) * 2019-02-11 2020-08-20 Chancey John Methods of making and using phytocannabinoids complexed with a protein, peptide, amino acid, polysaccharide, disaccharide, ormonosaccharide
CN109943547B (en) * 2019-04-18 2022-07-19 安徽农业大学 Tea tree sucrose synthase CsSUS587, preparation method and application
WO2020239784A1 (en) 2019-05-27 2020-12-03 Octarine Bio Ivs Genetically modified host cells producing glycosylated cannabinoids.
CN110201019A (en) * 2019-07-17 2019-09-06 李卫 It is a kind of for treating the composition and preparation method thereof containing cannabidiol of muscle cramp
CN110693027A (en) * 2019-10-14 2020-01-17 桂林莱茵生物科技股份有限公司 Water-dispersible cannabidiol product and preparation method thereof
WO2021146687A1 (en) * 2020-01-16 2021-07-22 Cannabis Global, Inc. A cannaboside composition and method to produce
CN112010924B (en) * 2020-09-04 2022-09-09 中国药科大学 Novel Nosiheptide glycosylated derivative and preparation method and application thereof
CA3197361A1 (en) * 2020-11-07 2022-05-12 Mathias Schuetz Production of glycosylated cannabinoids
CN114573648A (en) * 2020-11-30 2022-06-03 云南汉盟制药有限公司 Cannabinoid glycosides and methods of making the same
WO2022126028A1 (en) * 2020-12-11 2022-06-16 Graphium Biosciences, Inc. Continuous enzymatic perfusion reactor system
CA3233367A1 (en) * 2021-10-01 2023-04-06 Anindya GOSWAMI Cannabinoids c- and o-glycosides possessing anti-proliferative and anti-metastatic properties and process for preparation thereof
WO2023240221A2 (en) * 2022-06-11 2023-12-14 Trait Biosciences, Inc. System and methods for sequential desorption of cannabidiol (cbd) glycoside species
DE102022004596A1 (en) * 2022-12-08 2024-06-13 Biosynth Gmbh Novel cannabinoid oligosaccharides

Family Cites Families (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5292899A (en) 1991-11-27 1994-03-08 Synthetic Technology Corporation Synthesis of 11-nor-Δ-9-tetrahydrocannabinol-9-carboxylic acid glucuronide
US5627270A (en) * 1991-12-13 1997-05-06 Trustees Of Princeton University Glycosylated steroid derivatives for transport across biological membranes and process for making and using same
DK2176208T3 (en) 2007-07-30 2015-04-27 Zynerba Pharmaceuticals Inc Prodrugs of cannabidiol, compositions containing prodrugs of cannabidiol and methods of use thereof
DK2215071T3 (en) 2007-11-30 2015-10-05 Zynerba Pharmaceuticals Inc Prodrugs of tetrahydocannabinol, compositions containing prodrugs of tetrahydrocannabinol, as well as methods of use thereof
WO2009158499A2 (en) * 2008-06-25 2009-12-30 University Of North Texas Health Science Center At Fort Worth Prevention of bacterial growth and biofilm formation by ligands that act on cannabinoidergic systems
MX2011011445A (en) 2009-04-29 2011-11-18 Univ Kentucky Res Found Cannabinoid-containing compositions and methods for their use.
US8410064B2 (en) * 2009-08-24 2013-04-02 The Board Of Trustees Of The University Of Arkansas Classical cannabinoid metabolites and methods of use thereof
WO2012011112A1 (en) 2010-07-22 2012-01-26 Yissum Research Development Company Of The Hebrew University Of Jerusalem, Ltd. Non psychoactive cannabinoids and uses thereof
IT1402018B1 (en) 2010-10-11 2013-08-28 Indena Spa FORMULATIONS FOR THE TREATMENT OF THE AFFECTIONS OF THE FIRST RESPIRATORY ROUTES.
ES2690550T3 (en) 2011-01-13 2018-11-21 Austin Research Labs Corp. High load dispersions for the treatment of infections
MX2015008766A (en) 2013-01-08 2016-03-07 Yissum Res Dev Co Fluorinated cbd compounds, compositions and uses thereof.
EP2954058B1 (en) * 2013-02-06 2021-03-31 Evolva SA Methods for improved production of rebaudioside d and rebaudioside m
US10441617B2 (en) 2013-03-15 2019-10-15 Biotech Institute, Llc Breeding, production, processing and use of medical cannabis
WO2015074137A1 (en) * 2013-11-20 2015-05-28 Mary Lynch Compositions and methods for treatment of ocular inflammation and pain
US9497299B2 (en) 2014-09-18 2016-11-15 Blackberry Limited Configuring a discard timer
US20160374958A1 (en) 2015-06-23 2016-12-29 Axim Biotechnologies, Inc. Anti-microbial composition comprising cannabinoids
WO2017053574A1 (en) 2015-09-22 2017-03-30 Vitality Biopharma, Inc. Cannabinoid glycoside prodrugs and methods of synthesis
CA3021139A1 (en) 2016-04-15 2017-10-19 Teewinot Technologies Limited Biosynthesis of cannabinoid prodrugs
CA3068806A1 (en) 2016-07-14 2018-01-18 Therapix Biosciences Ltd. Compositions and methods of potentiating antimicrobials
AU2018239671A1 (en) 2017-03-24 2019-10-10 Trait Biosciences, Inc. High level In vivo biosynthesis and isolation of water-soluble cannabinoids in plant systems
US11207291B2 (en) 2017-05-09 2021-12-28 Graphium Biosciences, Inc. Antimicrobial compositions comprising cannabinoids and methods of using the same
US20200046639A1 (en) 2017-07-11 2020-02-13 Trait Biosciences Inc. Consumable water-soluble cannabinoid food and beverage additive having enhanced stability

Also Published As

Publication number Publication date
AU2016326518B2 (en) 2022-03-24
US20180264122A1 (en) 2018-09-20
EP3352855A4 (en) 2019-05-22
CA2999764A1 (en) 2017-03-30
CN108136208B (en) 2022-07-15
US11207414B2 (en) 2021-12-28
EP3352855A1 (en) 2018-08-01
WO2017053574A1 (en) 2017-03-30
BR112018005639A2 (en) 2018-10-09
CN108136208A (en) 2018-06-08
AU2016326518A1 (en) 2018-04-26
JP2018528977A (en) 2018-10-04

Similar Documents

Publication Publication Date Title
US20220168428A1 (en) Cannabinoid glycoside prodrugs and methods of synthesis
Hultin Bioactive C-glycosides from bacterial secondary metabolism
US8598132B2 (en) Glycosylated indolocarbazoles, method for obtaining same and uses thereof
Hardman et al. Cannabinoid glycosides: In vitro production of a new class of cannabinoids with improved physicochemical properties
Cui et al. Self-resistance during muraymycin biosynthesis: a complementary nucleotidyltransferase and phosphotransferase with identical modification sites and distinct temporal order
Chiu et al. Diversity of sugar acceptor of glycosyltransferase 1 from Bacillus cereus and its application for glucoside synthesis
US20220227726A1 (en) Cannabinoid glycosides and uses thereof
Aly et al. Synthesis, antimicrobial and cytotoxicity evaluation of new cholesterol congeners
Shen et al. Structural and pharmacological diversity of 1, 4-naphthoquinone glycosides in recent 20 years
US9605015B2 (en) Polyene compound, method for preparing the same, and antifungal drug comprising novel polyene compound as active ingredient
Parajuli et al. Regiospecific biosynthesis of tamarixetin derivatives in Escherichia coli
Liu et al. Recent advances in methods of puerarin biotransformation
CN112830949B (en) Antifungal compound produced by marine aspergillus and preparation method thereof
Wu et al. Design, synthesis, and anti-tobacco mosaic virus (TMV) activity of glycoconjugates of phenanthroindolizidines alkaloids
Shimoda et al. Glycosylation of daidzein by the Eucalyptus cell cultures
US20230346952A1 (en) Cannabinoid glycoside prodrugs and methods of synthesis
US20230192748A1 (en) Etoposide Glycosides, Methods Of Making, And Uses Thereof As An Anti-Cancer Drug
Hoang et al. Biochemical characterization of recombinant UDP-glucose: sterol 3-O-glycosyltransferase from Micromonospora rhodorangea ATCC 31603 and enzymatic biosynthesis of sterol-3-O-β-glucosides
KR101575295B1 (en) Novel Epothilone Glycosides and Method for Preparing the Same
Hu et al. Directed biosynthesis of iso-aclacinomycins with improved anticancer activity
KR101704706B1 (en) Method for Preparing Sterol Glucosides Using Sterol Glucosyltransferase
Ponrasu et al. Syntheses of α-tocopheryl glycosides by glucosidases
KR101966882B1 (en) New Glycoside Derivatives of Genistein, Pharmaceutical Compositions for Treating Cancer Diseases Containing The Same and Method for Preparing the Same
CN111205343B (en) Nitrogen acetyl glucoside or galactoside compound of epothilone B, and enzymatic preparation and application thereof
KR102141656B1 (en) Method for bioconversion of corticosterone by glucosyltransferase and corticosterone glucoside produced thereby

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION