WO2008046155A1 - Methods for regulating glucose homeostasis and agents therefor - Google Patents

Methods for regulating glucose homeostasis and agents therefor Download PDF

Info

Publication number
WO2008046155A1
WO2008046155A1 PCT/AU2007/001593 AU2007001593W WO2008046155A1 WO 2008046155 A1 WO2008046155 A1 WO 2008046155A1 AU 2007001593 W AU2007001593 W AU 2007001593W WO 2008046155 A1 WO2008046155 A1 WO 2008046155A1
Authority
WO
WIPO (PCT)
Prior art keywords
optionally substituted
alkyl
hydrogen
glucose
compound
Prior art date
Application number
PCT/AU2007/001593
Other languages
French (fr)
Inventor
Gregory Royce Collier
Ken Russell Walder
Subhadhcha Poonsatha
Original Assignee
Dia-B Tech Limited
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 Dia-B Tech Limited filed Critical Dia-B Tech Limited
Publication of WO2008046155A1 publication Critical patent/WO2008046155A1/en

Links

Classifications

    • 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
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H7/00Compounds containing non-saccharide radicals linked to saccharide radicals by a carbon-to-carbon bond
    • C07H7/06Heterocyclic radicals

Definitions

  • the present invention relates generally to the field of therapy.
  • the invention particularly relates to methods of regulating glucose homeostasis and to the therapeutic or prophylactic treatment of diseases and conditions, such as diabetes, syndrome X, obesity, hyperglycaemia, cardiovascular disease, vascular disease and kidney disease, in which impaired glucose uptake due to insulin resistance is involved or implicated.
  • diseases and conditions such as diabetes, syndrome X, obesity, hyperglycaemia, cardiovascular disease, vascular disease and kidney disease, in which impaired glucose uptake due to insulin resistance is involved or implicated.
  • the present invention further relates to compounds and agents and compositions thereof for use in the treatment methods.
  • Glucose is the body's preferred energy source. Once entered into the blood stream, it requires the assistance of the insulin to enter hepatic, muscle and adipose cells in order to be stored or utilised. In a healthy individual, glucose homeostasis is controlled primarily by insulin. As blood glucose levels rise, such as after eating, specialised ⁇ -cells within the pancreas release insulin which promotes glucose uptake, intracellular metabolism and glycogen synthesis by the body's target tissues. Thus, in healthy individuals, blood glucose concentrations are strictly controlled, typically in the range of 80-110 mg/dl. However, where the pancreas produces an inadequate insulin response, or the target cells do not respond appropriately to the insulin produced, the glucose cannot enter the body's cells. This results in a rapid accumulation of glucose in the blood stream (hyperglycemia).
  • diabetes Chronically elevated blood glucose levels (greater than about 126 mg/dl) from either inadequate insulin secretion and/or an inadequate response or sensitivity to insulin is referred to as diabetes, a disease which is now suffered by more than 10 percent of adults in the USA.
  • the disease may be characterised by persistent hyperglycemia, polyuria, polydipsia and/or hyperphagia, chronic microvascular complications such as retinopathy, nephropathy and neuropathy, and macrovascular complications, such as hyperlipidemia and hypertension which can lead to blindness, end-stage renal disease, limb amputation and myocardial infarction.
  • the three most common types of diabetes are type 1, type 2 and gestational.
  • Type 1 known as insulin dependent diabetes mellitus (IDDM), or juvenile-onset diabetes, occurs in 10-15% of all cases. It is most commonly diagnosed in children and adolescents but can occur in young adults as well. It is characterised by ⁇ -cell destruction resulting in a loss of insulin secretory function. Most cases relate to autoimmune destruction of the ⁇ - cells. Treatment is via insulin injection and must be continued indefinitely.
  • IDDM insulin dependent diabetes mellitus
  • type 2 diabetes known as non-insulin dependent diabetes mellitus (NIDDM) or late-onset diabetes
  • NIDDM non-insulin dependent diabetes mellitus
  • insulin resistance or insensitivity the pancreas begins to secrete excess insulin, however, in time the pancreas becomes less able to produce enough insulin resulting in chronic hyperglycaemia.
  • Initial symptoms are typically milder than for type 1 and the condition may go undiagnosed for years before more severe symptoms present. Lifestyle (poor diet and inactivity) is considered to be a major mediating factor of the condition, although a genetic predisposition increases the risk. Treatment focuses on lifestyle modification, insulin therapy and/or anti-diabetic agents such as insulin sensitisers and insulin releasers.
  • Gestational diabetes occurs in about 2-5% of all pregnancies. It is temporary, but if untreated may cause foetal complications. Most sufferers make a complete recovery after the birth. However, a proportion of women who develop gestational diabetes go on to develop type 2 diabetes.
  • diabetes causes of diabetes include genetic defects in ⁇ -cells, genetically related insulin resistance, diseases of the pancreas, hormonal defects, malnutrition and chemical or drug influences.
  • Impaired glucose tolerance and impaired fasting glucose are pre-type 2 diabetic states, closely related to type 2 and occur when the blood glucose level is higher than normal but not high enough to be classified as diabetes (about 110-126 mg/dl). As with type 2, the body produces insulin but in an insufficient amount or the target tissues are unresponsive to the insulin produced.
  • Insulin Resistance Syndrome X also known as Insulin Resistance Syndrome (IRS) is a cluster of risk factors for heart disease and is associated with insulin resistance. It presents symptoms or risk factors for the development of type 2 diabetes and heart disease including obesity, atheriosclerosis, hypertriglyceridemia, low HDL cholesterol, hyperinsulinemia, hyperglycaemia, hypertension, impaired glucose tolerance and impaired fasting glucose.
  • IRS Insulin Resistance Syndrome
  • High blood glucose levels and insulin resistance are also associated with fatty liver disease, which can progress to chronic inflammation, fibrosis and cirrhosis.
  • insulin resistance or insensitivity, can play a significant role in diabetic and other hyperglycemic-related conditions.
  • the latest WHO estimate (for the number of people with diabetes, worldwide, in 2003) is 194 million. This is expected to increase to at least 330 million by 2025 and it is estimated that there are around 4 million deaths per year are related to the disorder.
  • Insulin sensitisers resensitise cells to the action of insulin thereby reducing blood glucose levels and triglyceride levels.
  • the present invention is predicated on compounds related to strictosidine, a key intermediate in the biosynthesis of terpenoid indole alkaloids, as a new class of insulin sensitizers and provides new methods for modulating glucose uptake by a cell and treating insulin resistance and conditions associated with insulin resistance.
  • R 1 -R 4 are independently selected from hydrogen, hydroxy, thiol, halo, CO 2 H, carboxy ester, amino, nitro, cyano, amido, sulfoxide, sulfonamide, sulfonyl, sulfate, sulfonate, phosphate, phosphonate, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted carbocyclyl, optionally substituted heterocyclyl, optionally substituted acyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aryloxy, optionally substituted heteroaryloxy, optionally substituted carbocyclyloxy, optionally substituted heterocyclyloxy, optionally substituted aralkyloxy, optionally substituted acyloxy, optionally substituted alkylthi
  • R 6 is absent when " " - is a double bond
  • R 5 and R 6 independently are selected from hydrogen, optionally substituted alkyl, optionally substituted acyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted aralkyl, optionally substituted heterocyclyl, optionally substituted carbocyclyl, and amido;
  • R 7 is selected from hydrogen, optionally substituted alkyl, CO 2 H, carboxy ester, amido, optionally substituted heteroaryl and optionally substituted aryl;
  • R 8 is selected from hydrogen, optionally substituted alkyl, optionally substituted aryl, optionally substituted heteroaryl, CO 2 H, carboxy ester and SO 3 H;
  • R 7a and R 8a are independently selected from hydrogen, optionally substituted alkyl and optionally substituted aryl.
  • COX forms a group selected from optionally substituted acyl, CO 2 H, carboxy ester and amido
  • Y is selected from hydrogen, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclyl, optionally substituted carbocyclyl, optionally substituted acyl and a saccharide group,
  • Z is selected from optionally substituted at least C 2 alkyl, optionally substituted alkenyl and optionally substituted alkynyl,
  • the present invention thus provides a method of increasing glucose uptake by a cell comprising contacting said cell with a compound of Formula (I) or a pharmaceutically acceptable salt or prodrug thereof.
  • Methods for increasing glucose uptake by a cell for example adipocytes and muscle and hepatic cells, can be performed in vivo or in vitro.
  • a second aspect of the invention relates to a method for regulating glucose homeostasis in a subject in need thereof comprising the administration of a compound of Formula (I) or a pharmaceutically acceptable salt or prodrug thereof.
  • the present invention provides a method of treating insulin resistance in a subject in need thereof comprising the administration of a compound of Formula (I) or a pharmaceutically acceptable salt or prodrug thereof.
  • a further aspect of the invention relates to a method of lowering blood glucose levels in a subject in need thereof comprising the administration of a compound of Formula (I) or a pharmaceutically acceptable salt or prodrug thereof.
  • Yet a further aspect of the invention provides a method for treating a disease or condition, or symptom thereof, in which insulin resistance is involved comprising the administration of a compound of Formula (I) or a pharmaceutically acceptable salt or prodrug thereof to subject in need thereof.
  • the invention provides a compound of Formula (I) or a pharmaceutically acceptable salt or prodrug thereof provided that the compound is not (3R) or (3S) strictosidine, strictosidine ethyl ester and allyl ester, strictosidinic acid, 5- carboxystrictosidine, 3,4-dehydro-5-carboxystrictosidine, 3,4-dehydro5- carbomethoxystrictosidine or gluco-acetylated forms thereof.
  • a compound of Formula (I) or pharmaceutically acceptable salt or prodrug thereof for use in therapy, particularly for regulating glucose homeostasis, treating insulin resistance, lowering blood levels and/or treating a disease or condition, or one or more symptoms thereof, in which insulin resistance is involved.
  • the present invention also provides for agents and compositions comprising compounds of Formula (I), or pharmaceutically acceptable salt or prodrug thereof and the use of said compounds in treatment in the manufacture of medicaments for regulating glucose homeostasis, treating insulin resistance, lowering blood levels and/or treating a disease or condition, or one or more symptoms thereof, in which insulin resistance is involved.
  • said compositions and medicaments comprise the compound of Formula (I), or pharmaceutically acceptable salt or prodrug thereof, as at least 50%, such as at least 90 or 95% of the active component.
  • alkyl or "alk”, used either alone or in compound words denotes straight chain, branched or cyclic alkyl, preferably C 1-20 alkyl, eg C 1-10 or C 1-6.
  • straight chain and branched alkyl include methyl, ethyl, «-propyl, isopropyl, «-butyl, sec- butyl, t-butyl, n-pentyl, 1,2-dimethylpropyl, 1,1-dimethyl-propyl, hexyl, 4-methylpentyl, 1- methylpentyl, 2-methylpentyl, 3-methylpentyl, 1,1-dimethylbutyl, 2,2-dimethylbutyl, 3,3- dimethylbutyl, 1,2-dimethylbutyl, 1,3-dimethylbutyl, 1,2,2,-trimethyl ⁇ ropyl, 1,1,2- trimethylpropyl, heptyl, 5-methylhexyl
  • cyclic alkyl examples include mono- or polycyclic alkyl groups such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl, cyclodecyl and the like. Where an alkyl group is referred to generally as "propyl", butyl” etc, it will be understood that this can refer to any of straight, branched and cyclic isomers where appropriate. An alkyl group may be optionally substituted by one or more optional substitutents as herein defined.
  • alkenyl denotes groups formed from straight chain, branched or cyclic hydrocarbon residues containing at least one carbon to carbon double bond including ethylenically mono-, di- or poly-unsaturated alkyl or cycloalkyl groups as previously defined, preferably C 2-20 alkenyl (eg C 2-1O or C 2-6 )-
  • alkenyl include vinyl, allyl, 1-methylvinyl, butenyl, iso-butenyl, 3-methyl ⁇ 2-butenyl, 1-pentenyl, cyclopentenyl, 1-methyl-cyclopentenyl, 1-hexenyl, 3-hexenyl, cyclohexenyl, 1-heptenyl, 3-he ⁇ tenyl, 1-octenyl, cyclooctenyl, 1-nonenyl, 2-nonenyl, 3-nonenyl, 1-decenyl, 3- decenyl, 1,3-but
  • alkynyl denotes groups formed from straight chain, branched or cyclic hydrocarbon residues containing at least one carbon-carbon triple bond including ethynically mono-, di- or poly- unsaturated alkyl or cycloalkyl groups as previously defined. Unless the number of carbon atoms is specified the term preferably refers to C 2-2 O alkynyl (eg C 2-10 or C 2 . 6 ). Examples include ethynyl, 1-propynyl, 2-propynyl, and butynyl isomers, and pentynyl isomers. An alkynyl group may be optionally substituted by one or more optional substitutents as herein defined.
  • [group] oxy refer to a particular group when linked by oxygen, for example, the terms “alkoxy”, “alkenoxy”, . “alkynoxy” and “aryloxy” and “acyloxy” respectively denote alkyl, alkenyl, alkynyl, aryl and acyl groups as hereinbefore defined when linked by an oxygen atom.
  • [group]thio refer to a particular group when linked by sulfur, for example, the terms “alkylthio”, “alkenylthio", alkynylthio” and
  • arylthio respectively denote alkyl, alkenyl, alkynyl, aryl groups as hereinbefore defined when linked by a sulfur atom.
  • a term written as M [groupA]groupB" is intended to refer to a groupA when linked by a divalent form of groupB, for example, "hydroxyalkyl” is a hydroxy group when linked by an alkylene group.
  • halogen denotes fluorine, chlorine, bromine or iodine (fluoro, chloro, bromo or iodo).
  • aryl (or “carboaryl)" or the abbreviated form “ar” used in compound words such as “aralkyl” denotes any of single, polynuclear, conjugated and fused residues of aromatic hydrocarbon ring systems.
  • aryl include phenyl, biphenyl, terphenyl, quaterphenyl, naphthyl, tetrahydronaphthyl, anthracenyl, dihydroanthracenyl, benzanthracenyl, dibenzanthracenyl, phenanthrenyl, fluorenyl, pyrenyl, idenyl, azulenyl, chrysenyl.
  • Preferred aryl include phenyl and naphthyl.
  • An aryl group may be optionally substituted by one or more optional substituents as herein defined.
  • carbocyclyl includes any of non-aromatic monocyclic, polycyclic, fused or conjugated cyclic hydrocarbon residues, preferably C 3-20 (e.g. C 3 - J0 or C 3-8 ).
  • the rings may be saturated, for example cycloalkyl, or may possess one or more double bonds (cycloalkenyl) and/or one or more triple bonds (cycloalkynyl).
  • Particularly preferred carbocyclyl are 5-6-membered or bicyclic 9-10 membered ring systems.
  • Suitable examples include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl, cyclodecyl, cyclopentenyl, cyclohexenyl, cyclooctenyl, cyclopentadienyl, cyclohexadienyl, cyclooctatetraenyl, indanyl, decalinyl and indenyl.
  • a carbocyclyl group may be optionally substituted by one or more optional substituents as herein defined.
  • heterocyclyl when used alone or in compound words includes any of monocyclic, polycyclic, fused or conjugated hydrocarbon residues, preferably C 3-20 (e.g. C 3-10 or C 3-8 ) wherein one or more carbon atoms are replaced by a heteroatom so as to provide a non-aromatic residue.
  • Suitable heteroatoms include, O, N, S, P and Se 5 particularly O, N and S. Where two or more carbon atoms are replaced, this may be by two or more of the same heteroatom or by different heteroatoms.
  • the heterocyclyl group may be saturated or partially unsaturated, i.e. possess one or more double bonds. Particularly preferred heterocyclyl are 5-6 and bicyclic 9-10 membered heterocyclyl.
  • heterocyclyl groups may include azridinyl, oxiranyl, thiiranyl, azetidinyl, oxetanyl, thietanyl, 2H-pyrrolyl, pyrrolidinyl, pyrrolinyl, piperidyl, piperazinyl, morpholinyl, indolinyl, imidazolidinyl, imidazolinyl, pyrazolidinyl, thiomorpholinyl, dioxanyl, tetrahydrofuranyl, tetrahydropyranyl, tetrahydropyrrolyl, tetrahydrothiophenyl, pyrazolinyl, dioxalanyl, thiazolidinyl, isoxazolidinyl, dihydropyranyl, oxazinyl, thiazinyl, thiomorpholinyl, oxathianyl, dithi
  • heteroaryl includes any of monocyclic, polycyclic, fused or conjugated hydrocarbon residues, wherein one or more carbon atoms are replaced by a heteroatom so as to provide an aromatic residue.
  • Preferred heteroaryl have 3-20 ring atoms, e.g. 3-10.
  • Particularly preferred heteroaryl are 5-6 and 9-10 membered bicyclic ring systems.
  • Suitable heteroatoms include, O, N, S, P and Se, particularly O, N and S. Where two or more carbon atoms are replaced, this may be by two or more of the same heteroatom or by different heteroatoms.
  • heteroaryl groups may include pyridyl, pyrrolyl, thienyl, imidazolyl, furanyl, benzothienyl, isobenzothienyl, benzofuranyl, isobenzofuranyl, indolyl, isoindolyl, pyrazolyl, pyrazinyl, pyrimidinyl, pyridazinyl, indolizinyl, quinolyl, isoquinolyl, phthalazinyl, 1,5-naphthyridinyl, quinozalinyl, quinazolinyl, quinolinyl, oxazolyl, thiazolyl, isothiazolyl, isoxazolyl, triazolyl, oxadialzolyl, oxatriazolyl, triazinyl, tetrazolyl and furazanyl.
  • a heteroaryl group may be optionally substituted by one
  • Preferred acyl includes C(O)-R, wherein R is hydrogen or an alkyl, alkenyl, alkynyl, aryl, heteroaryl, carbocyclyl, or heterocyclyl residue.
  • R is hydrogen or an alkyl, alkenyl, alkynyl, aryl, heteroaryl, carbocyclyl, or heterocyclyl residue.
  • examples of acyl include formyl, straight chain or branched alkanoyl (eg.
  • C 1 ⁇ o such as, acetyl, propanoyl, butanoyl, 2-methylpropanoyl, pentanoyl, 2,2-dimethylpropanoyl, hexanoyl, heptanoyl, octanoyl, nonanoyl, decanoyl, undecanoyl, dodecanoyl, tridecanoyl, tetradecanoyl, pentadecanoyl, hexadecanoyl, heptadecanoyl, octadecanoyl, nonadecanoyl and icosanoyl; cycloalkylcarbonyl such as
  • aralkenoyl such as phenylalkenoyl (e.g. phenylpropenoyl, phenylbutenoyl, phenylmethacryloyl, phenylpentenoyl and phenylhexenoyl and naphthylalkenoyl (e.g.
  • aryloxyalkanoyl such as phenoxyacetyl and phenoxypropionyl
  • arylthiocarbanioyl such as phenylthiocarbamoyl
  • arylglyoxyloyl such as phenylglyoxyloyl and naphthylglyoxyloyl
  • arylsulfonyl such as phenylsulfonyl and napthylsulfonyl
  • heterocycliccarbonyl heterocyclicalkanoyl such as thienylacetyl, thienylpropanoyl, thienylbutanoyl, thienylpentanoyl, thienylhexanoyl, thiazolylacetyl, thiadiazolylacetyl and tetrazolylacetyl
  • a group may or may not be further substituted or fused (so as to form a condensed polycyclic group) with one, two, three or more of organic and inorganic groups, including those selected from: alkyl, alkenyl, alkynyl, carbocyclyl, aryl, heterocyclyl, heteroaryl, acyl, aralkyl, alkylaryl, alkylheterocyclyl, alkylheteroaryl, alkylcarbocyclyl, halo, haloalkyl, haloalkenyl, haloalkynyl, haloaryl, halocarbocyclyl, haloheterocyclyl, haloheteroaryl, haloacyl, haloaryalkyl, hydroxy, hydroxyalkyl, hydroxyalkenyl, hydroxyalkynyl, hydroxycarbocyclyl, hydroxy
  • optional substitutents include alkyl, (e.g. C 1-6 alkyl such as methyl, ethyl, propyl, butyl, cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl), hydroxyalkyl (e.g. hydroxymethyl, hydroxyethyl, hydroxypropyl), alkoxyalkyl (e.g. methoxymethyl, methoxyethyl, methoxypropyl, ethoxymethyl, ethoxy ethyl, ethoxypropyl etc) alkoxy (e.g.
  • alkyl e.g. C 1-6 alkyl such as methyl, ethyl, propyl, butyl, cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl
  • hydroxyalkyl e.g. hydroxymethyl, hydroxyethyl, hydroxypropyl
  • Ci -6 alkoxy such as methoxy, ethoxy, propoxy, butoxy, cyclopropoxy, cyclobutoxy), halo, trifluoromethyl, trichloromethyl, tribromomethyl, hydroxy, phenyl (which itself may be further substituted e.g., by one or more C 1-6 alkyl, halo, hydroxy, hydroxyCi -6 alkyl, C 1- 6 alkoxy, haloC 1-6 alkyl, cyano, nitro, OC(O)C ⁇ alkyl, and amino), benzyl (wherein benzyl itself may be further substituted e.g., by one or more of C 1-6 alkyl, halo, hydroxy, hydroxyd- ⁇ alkyl, C 1-6 alkoxy, haloCi -6 alkyl, cyano, nitro, OC(O)Ci -6 alkyl, and amino), phenoxy (wherein phenyl itself may be further substituted
  • C 1-6 alkyl such as methylamino, ethylamino, propylamino etc
  • dialkylamino e.g. C 1-6 alkyl, such as dimethylamino, diethylamino, dipropylamino
  • acylamino e.g.
  • phenylamino (wherein phenyl itself may be further substituted e.g., by one or more of C 1- 6 alkyl, halo, hydroxy hydroxyCi ⁇ alkyl, Ci -6 alkoxy, haloC 1-6 alkyl, cyano, nitro, OC(O)Ci- 6 alkyl, and amino), nitro, formyl, -C(O)-alkyl (e.g. C 1-6 alkyl, such as acetyl), O-C(O)- alkyl (e.g.
  • C ⁇ alkyl such as acetyloxy
  • benzoyl wherein benzyl itself may be further substituted e.g., by one or more of C 1-6 alkyl, halo, hydroxy hydroxyC 1-6 alkyl, C ⁇ alkoxy, haloC 1-6 alkyl, cyano, nitro, 0C(0)Ci- 6 alkyl, and amino
  • benzoyloxy wherein benzyl itself may be further substituted e.g., by one or more of C h alky!, halo, hydroxy hydroxyC ⁇ 6 alkyl, C 1 ⁇ aIkOXy, haloC t -galkyl, cyano, nitro, 0C(0)Ci- 6 alkyl, and amino
  • CO 2 H C0 2 alkyl (e.g.
  • C 1-6 alkyl such as methyl ester, ethyl ester, propyl ester, butyl ester
  • COaphenyl wherein phenyl itself may be further substituted e.g., by one or more of C 1- 6 alkyl, halo, hydroxy, hydroxyCi.
  • C 1-6 alkyl such as methyl ester, ethyl ester, propyl ester, butyl amide) CONHdialkyl (e.g. 6alkyl- and thioalkyl (e.g., HSd. 6 alkyl-), carboxyalkyl (e.g., H ⁇ 2 CC 1-6 alkyl-), carboxyesteralkyl (e.g., C 1-6 alkylO 2 CC 1-6 alkyl-), amidoalkyl (e.g., H 2 N(O)CC i- ⁇ alkyl-, H(C 1 . 6 alkyl)N(O)CC 1 .
  • CONHdialkyl e.g. 6alkyl- and thioalkyl (e.g., HSd. 6 alkyl-)
  • carboxyalkyl e.g., H ⁇ 2 CC 1-6 alkyl-
  • carboxyesteralkyl e.g., C 1-6 alkylO
  • sulfoxide refers to a group -S(O)R wherein R is selected from hydrogen, alkyl, alkenyl, alkynyl, aryl, heteroaryl, heterocyclyl, carbocyclyl, and aralkyl, each of which may be optionally substituted.
  • R include hydrogen, C 1-20 alkyl, phenyl and benzyl.
  • sulfonyl refers to a group S(O) 2 -R, wherein R is selected from hydrogen, alkyl, alkenyl, alkynyl, aryl, heteroaryl, heterocyclyl, carbocyclyl, acyl, and aralkyl, each of which may be optionally substituted.
  • R include hydrogen, C ⁇ oalkyl, phenyl and benzyl.
  • sulfonamide refers to a group S(O) 2 NRR wherein each R is independently selected from hydrogen, alkyl, alkenyl, alkynyl, aryl, heteroaryl, heterocyclyl, carbocyclyl, acyl, and aralkyl, each of which may be optionally substituted.
  • R include C 1-20 alkyl, phenyl and benzyl. In one embodiment at least one R is hydrogen. In another embodiment, both R are hydrogen.
  • a “sulfate” group refers to a group OS(O) 2 OR wherein each R is independently selected from hydrogen, alkyl, alkenyl, alkynyl, aryl, heteroaryl, heterocyclyl, carbocyclyl, acyl, and aralkyl, each of which may be optionally substituted.
  • R include hydrogen, C 1-2 oalkyl, phenyl and benzyl.
  • sulfonate refers to a group OS(O) 2 R wherein each R is independently selected from hydrogen, alkyl, alkenyl, alkynyl, aryl, heteroaryl, heterocyclyl, carbocyclyl, acyl, and aralkyl, each of which may be optionally substituted.
  • R include hydrogen, C 1-20 alkyl, phenyl and benzyl.
  • phosphonate refers to a group P(O)(OR 2 ) wherein R is independently selected from hydrogen, alkyl, alkenyl, alkynyl, aryl, heteroaryl, heterocyclyl, carbocyclyl, acyl, and aralkyl, each of which may be optionally substituted.
  • R include hydrogen, C ⁇ oalkyl, phenyl and benzyl.
  • phosphate refers to a group OP(O)(OR) 2 wherein R is independently selected from hydrogen, alkyl, alkenyl, alkynyl, aryl, heteroaryl, heterocyclyl, carbocyclyl, acyl, and aralkyl, each of which may be optionally substituted.
  • R include hydrogen, C 1-2 oalkyl, phenyl and benzyl.
  • thio is intended to include groups of the formula "SR" wherein R can be hydrogen (thiol), alkyl, alkenyl, alkynyl, aryl, carbocyclyl, heteroaryl, heterocyclyl, aralkyl, and acyl, each of which may be optionally substituted.
  • R include hydrogen, C 1-2 oalkyl, phenyl and benzyl.
  • amino is used here in its broadest sense as understood in the art and includes groups of the formula NR A R B wherein R A and R B may be any independently selected from hydrogen, hydroxy alkyl, alkoxyalkyl, alkenyl, alkynyl, aryl, carbocyclyl, heteroaryl, heterocyclyl, aralkyl, and acyl, each of which may be optionally substituted.
  • R A and R B together with the nitrogen to which they are attached, may also form a monocyclic, or polycyclic ring system e.g. a 3-10 membered ring, particularly, 5-6 and 9-10 membered systems.
  • amino examples include NH 2 , NHalkyl (e.g. Ci- 20 alkyl), NHalkoxyalkyl, NHaryl (e.g. NHphenyl), NHaralkyl (e.g. NHbenzyl), NHacyl (e.g. NHC(O)C 1-20 alkyl, NHC(O)phenyl), Ndialkyl (wherein each alkyl, for example C 1-2O , may be the same or different) and 5 or 6 membered rings, optionally containing one or more same or different heteroatoms (e.g. O, N and S).
  • Reference to groups written as "[group] amino" is intended to reflect the nature of the R A and R B groups.
  • alkylamino refers to NR A R B where one of R A or R B is alkyl.
  • Dialkylamino refers to NR A R B where R A and R B are each (independently) an alkyl group.
  • amido examples include C(O)NH 2 , C(O)NHalkyl (eg C 1-2O alkyl), C(O)NHaryl (eg
  • C(O)NHphenyl C(O)NHaralkyl (eg C(O)NHbenzyl), C(O)NHacyl (eg C(O)NHC(O)C 1 .
  • each alkyl for example C 1-20 , may be the same or different
  • 5 or 6 membered rings optionally containing one or more same or different heteroatoms (eg O, N and S).
  • -NR A R B may be the amino terminus of an amino acid or peptide molecule.
  • Such “amido” groups can be formed by coupling the amino terminus of an amino acid or peptide molecule with a precursor carboxylic acid group in the usual manner.
  • a peptide molecule comprises 2, 3, 4, 5 or more same or different amino acid molecules covalently linked by peptide or amide bonds formed between the amino terminus of one amino acid and the carboxylic acid terminus of another.
  • the amino acids contemplated include natural amino acids and non-natural amino acids, and modified and protected forms thereof.
  • carboxy ester is used here in its broadest sense as understood in the art and includes groups having the formula CO 2 R, wherein R may be selected from groups including alkyl, alkenyl, alkynyl, aryl, carbocyclyl, heteroaryl, heterocyclyl, aralkyl, and acyl, each of which may be optionally substituted.
  • Examples of carboxy ester include CQA-aoalkyl, CO 2 aryl (eg. CO 2 ⁇ henyl), CO 2 aralkyl (eg CO 2 benzyl).
  • saccharide groups contemplated herein include mono-, di- and tri-saccharides of pyranose and furanose groups. In preferred embodiments there are linked through Cl. Examples include, glucose, idose, allose, tallose, mannose, galactose, maltose, lactose, arabinose, glucosamine, ribose, xylose, cellobiose, maltotriose, cellotriose, isomaltotriose.
  • the saccharides are monosaccharides.
  • the saccharides are the pyranoses.
  • the saccharide group is glucose.
  • saccharide groups can exist in their "free hydroxyl” or “free amino” form or one or more, or all, of the hydroxyl and/or amino groups can be protected by a suitable protecting group (eg acetate groups).
  • a saccharide group may be removed and/or replaced by known methods in the art.
  • Compounds contemplated by the invention may be obtained by a variety of means including isolation from natural sources, synthetic methods for the preparation of isoquinolines/tetrahydro- ⁇ -carbolines and dehydro forms thereof, or by enzyme catalysed coupling.
  • naturally occurring compounds such as strictosidine (i), strictosidinic acid (ii), 5(S)- 5-carboxystrictosidine (iii) and 3,4-dehydro-5(S)-5-carboxystrictosidine (iv) (see below) can be isolated from their natural sources in accordance with described procedures (see for example, Kitajima, M. et al., 2002; Arbain, D. et al, 1993 and St ⁇ ckigt, J.
  • compounds where - represents a single bond can be prepared by acid- mediated or strictosidine synthase-catalysed coupling of an appropriately substituted/protected tryptamine compound and horrin, or derivative thereof, as depicted, for example in general Scheme I below.
  • appropriate tryptamine and Langanin compounds may be coupled under acidic conditions (Pictet Spengler reaction) such as described in US 6,720,331, Patthy- Lukats, A. et ah, 1999 and The Merck Index, Thirteenth Edition.
  • Compounds where ' • ' -"-" is a double bond referred to herein as 3,4-dehydro compounds, may be prepared by coupling an appropriate tryptamine compound and a modified horryde has been oxidized to a carboxylic acid group, followed by dehydrating ring closure in accordance with the methods described by US 6,350,757 and Kitajima, M. et al, 2002, and as described below in general Scheme III.
  • a suitable dehydrating agent such as DCC or POCl 3
  • 3,4-dehydro compounds may be prepared from the 3,4-dihydro compounds by appropriate oxidative treatment, such as treatment with DDQ (see for example US 6,350,757 or US 6,720,331).
  • Starting tryptamine compounds may be prepared by any one of the many methods known in the art for indole synthesis, or alternatively, may be obtained from commercial sources. Some examples of starting tryptamines contemplated by the present invention include:
  • Secologanin may be obtained commercially from Sigma (Fluka), USA, or isolated from natural sources (Galan et al, 2006). It may be modified prior to coupling, e.g. removal or replacement of the glucose moiety, protection of free OH groups, oxidation to the carboxylic acid (for synthesis of 3,4-dehydro compounds) or modification of the ester moiety. Alternatively, the vinyl group may be modified in accordance with the olefin cross metathesis methods described by Galan et al., 2006, to afford further horrin analogues.
  • R H or OH
  • R 1 H or protecting group
  • protecting group refers to an introduced functionality which temporarily renders a particular functional group inactive under certain conditions. Such protecting groups and methods for their installation and subsequent removal at an appropriate stage are described in Protective Groups in Organic Chemistry, 3 rd Edition, T.W.Greene and P. G.
  • exemplary forms of protected groups include: for amino (NH 2 ) - carbamates (such as Cbz, Boc, Fmoc), benzylamines, acetamides (e.g. acetamide, trifluoroacetamide); for carbonyl - acetals, ketals, dioxanes, dithianes, and hydrazones; for hydroxy - ethers (e.g.
  • alkyl ethers alkoxylalkyl ethers, allyl ethers, silyl ethers, benzyl ethers, tetrahydropyranyl ethers), carboxylic acid esters, acetals (e.g. acetonide and benzylidene acetal); for thio (SH) -ethers (e.g. alkyl ethers, benzyl ethers), esters for CO 2 H - esters (e.g. alkyl esters, benzyl esters).
  • SH thio
  • Such isomers may be prepared by asymmetric synthesis, for example using chiral intermediates or reagents, enzymes, or mixtures may be resolved by conventional methods, eg., chromatography, recrystallisation or use of a resolving agent.
  • the compounds of Formula (I) have the same stereochemistry at 1, 2, 3 or all 4 of the chiral centres depicted for strictosidinic acid (i), or I 5 2, 3 or all 4 chiral centres as depicted for 3,4-dehydro 5(S)-5-carboxystrictosidinic acid (iv).
  • R 1-4 are independently selected from hydrogen, alkyl, (e.g. C ⁇ alkyl such as methyl, ethyl, propyl, butyl, cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl), hydroxyalkyl (e.g. hydroxymethyl, hydroxyethyl, hydroxypropyl), alkoxyalkyl (e.g. methoxymethyl, methoxyethyl, methoxypropyl, ethoxymethyl, ethoxyethyl, ethoxypropyl etc) alkoxy (e.g.
  • alkyl e.g. C ⁇ alkyl such as methyl, ethyl, propyl, butyl, cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl
  • hydroxyalkyl e.g. hydroxymethyl, hydroxyethyl, hydroxypropyl
  • C 1-6 alkoxy such as methoxy, ethoxy, propoxy, butoxy, cyclopropoxy, cyclobutoxy
  • alkylthio e.g. MeS-, EtS-, PrS-, BuS-
  • halo trifluoromethyl, trichloromethyl, tribromomethyl, hydroxy, phenyl (which itself may be further substituted e.g., by Ci -6 alkyl, halo, hydroxy, hydroxyC 1-6 alkyl, C 1 .
  • benzyl (wherein benzyl itself may be further substituted e.g., by one or more of C h alky!, halo, hydroxy, hydroxyCi. ⁇ alkyl, C ⁇ alkoxy, haloCi. 6 alkyl, cyano, nitro, OC(O)C 1-6 alkyl, and amino), phenoxy (wherein phenyl itself may be further substituted e.g., by one or more of C 1 . 6 alkyl, halo, hydroxy, cyano, nitro, OC(O)C 1 .
  • benzyloxy (wherein benzyl itself may be further substituted e.g., by one or more of Ci -6 alkyl, halo, hydroxy, C 1-6 alkoxy, haloC 1-6 alkyl, cyano, nitro, OC(O)C 1 . 6 alkyl, and amino), amino, alkylamino (e.g. C 1-6 alkyl, such as methylamino, ethylamino, propylamino etc), dialkylamino (e.g. C 1-6 alkyl, such as dimethylamino, diethylamino, dipropylamino), acylamino (e.g.
  • NHC(O)CHs NHC(O)CHs
  • phenylamino wherein phenyl itself may be further substituted e.g., by one or more Of C 1 . 6 alkyl, halo, hydroxy hydroxyC ⁇ ealkyl, Ci -6 alkoxy, haloCi- ⁇ alkyl, cyano, nitro, OC(O)C 1 . 6 alkyl, and amino), nitro, formyl, -C(O)-alkyl (e.g. C 1-6 alkyl, such as acetyl), 0-C(O)- alkyl (e.g.
  • C ⁇ alkyl such as acetyloxy
  • benzoyl wherein benzyl itself may be further substituted e.g., by one or more of C ⁇ ancyl, halo, hydroxy hydroxyC 1-6 alkyl, C 1-6 alkoxy, halod.
  • C 1-6 alkyl such as methyl ester, ethyl ester, propyl ester, butyl ester
  • C ⁇ 2phenyl wherein phenyl itself may be further substituted e.g., by one or more of Ci- 6 alkyl, halo, hydroxy, hydroxyC t - ⁇ alkyl, C 1-6 alkoxy, haloCi -6 alkyl, cyano, nitro, OC(O)C 1 .
  • COibenzyl (wherein benzyl itself may be further substituted e.g., by one or more of C ⁇ alkyl, halo, hydroxy, hydroxyC 1-6 alkyl, C 1-6 alkoxy, haloC 1-6 alkyl, cyano, nitro, OC(O)C 1-6 alkyl, and amino), CONH 2 , CONHphenyl (wherein phenyl itself may be further substituted e.g., by one or more of C 1-6 alkyl, halo, hydroxy, hydroxyC ⁇ .
  • ⁇ alkyl C 1-6 alkoxy, haloC 1-6 alkyl, cyano, nitro, OC(O)C 1-6 alkyl, and amino
  • CONHbenzyl wherein benzyl itself may be further substituted e.g., by one or more of Ci- ⁇ alkyl, halo, hydroxy hydroxyC 1-6 alkyl, haloCi -6 alkyl, cyano, nitro, OC(O)C 1-6 alkyl, and amino
  • CONHalkyl e.g. C 1-6 alkyl such as methyl ester, ethyl ester, propyl ester, butyl amide
  • CONdialkyl e.g.
  • C 1-6 alkyl) aminoalkyl e.g., HNC 1-6 alkyl-, C 1-6 alkylHN-C 1-6 alkyl- and (C 1-6 alkyl) 2 N-C 1-6 alkyl-
  • thioalkyl e.g., HSC 1-6 alkyl-
  • carboxyalkyl e.g., HO 2 CCi- ⁇ alkyl-
  • carboxyesteralkyl e.g., C 1 . 6 alkyl0 2 CC 1-6 alkyl-
  • amidoalkyl e.g., H 2 N(O)CC 1- ealkyl-, H(C 1-6 alkyl)N(O)CC 1 .
  • alkyl- ⁇ alkyl-
  • formylalkyl e.g., OHCC ⁇ alkyl-
  • acylalkyl e.g., nitroalkyl (e.g., O 2 NC 1-6 alkyl-)
  • sulfoxidealkyl Cg 15 R(O)SC 1- ealkyl, such as C 1 . 6 alkyl(O)SC 1 .
  • sulfonylalkyl e.g., R(O) 2 SC i -6 alkyl- such as C 1- 6 alkyl(O) 2 SC 1-6 alkyl-
  • sulfonamidoalkyl e.g., 2 HN(O) 2 SC 1-6 alkyl, H(C 1 . 6 alkyl)N(O) 2 SCi- 6 alkyl-
  • 2 adjacent carbon atoms are substituted by one end each of a -O- (CH 2 ) n -0- or -NH-(CH 2 ) n -NH- group, wherein n is 1 or 2.
  • At least one of R 1 -R 4 is hydrogen, for example, at least 2 are hydrogen. In other embodiments, 3 or all 4 OfR 1 -R 4 are hydrogen.
  • R 5 and R 6 are selected from hydrogen, optionally substituted C 1-6 alkyl, optionally substituted C(O)C ⁇ alkyl, optionally substituted C(O)phenyl, optionally substituted C(0)beri2yl.
  • R 7 is selected from hydrogen, CO 2 H, CO 2 C 1-6 alkyl 5 e.g. methyl, ethyl, propyl or butyl), CONH 2 , CONHC 1-6 alkyl, CONHaryl. In particularly preferred embodiments, R 7 is selected from H and CO 2 H.
  • Rg is hydrogen
  • At least one of R 7a and Rg a is hydrogen, for example, both are hydrogen.
  • X include OH, OCi.ioalkyl, NHC ⁇ oalkyl, NHphenyl and NHbenzyl.
  • Y is H or a saccharide, such as glucose.
  • Some non-limiting examples of compounds contemplated by the invention include combinations of any two, three, four, five, six, seven or eight of the embodiments for R 1-4 , R 5 and R 6 , R 7 , R 8 , R 7a and Rg 8 , X, Y and Z described above.
  • the compound for use in the invention is strictosidinic acid.
  • Strictosidinic acid can be isolated from natural sources, in accordance with the procedures described in the literature, for example, described by Arbain et ah, 1993; Yamakazi et ah, 2003 and Geerlings et al. , 2000, and in accordance with the Examples described herein.
  • strictosidinic acid, or other compounds contemplated herein are used in extracted, isolated or substantially purified form (for example, at least 50, 75, 90, 95 or 99% pure).
  • regulating glucose homeostasis refers to the regulation or control of blood glucose levels to lower hyperglycaemic, or preferably achieve or maintain normal fasting state, blood glucose levels.
  • Normal fasting state blood glucose levels are typically less than 6.1 mmolL " (110 mgdL "1 ).
  • Hyperglycaemic levels refer to blood glucose levels greater than or equal to 6.1 mmolL '1 (110 mgdL "1 ).
  • Impaired fasting glycemia is characterised by a fasting plasma glucose concentration greater than or equal to 6.1 mmol (110 mgdL “1 ) but less than 7.0 mmolL “1 (126 mgdL “1 ) and a 2-h plasma glucose concentration during the oral glucose tolerance test (OGTT) (if measured) less than 7.8 mmolL “1 (140 mgdL “1 ).
  • Impaired glucose tolerance is characterised by a fasting plasma glucose concentration of less than 7.0 mmolL “1 (126 mgdL “1 ) and a 2-h plasma glucose concentration during the OGTT of greater than or equal to 7.8 mmolL '1 (140 mgdL “1 ) but less than 11.1 mmolL “1 (200 mgdL “1 ).
  • Diabetes is characterised by a fasting plasma glucose concentration of greater than or equal to 7.0 mmolL “1 (126 mgdL '1 ) or a 2-h plasma glucose concentration during the OGTT of greater than 11.1 mmolL '1 (200 mgdL "1 ).
  • the compounds of the invention may have utility in the therapeutic or prophylactic treatment of diseases and conditions, and /or their symptoms, in which insulin resistance in involved or implicated in a subject.
  • Any disease or condition, or symptom thereof in which insulin resistance or impaired glucose uptake by a cell or tissue can be attributed, or play a role or is manifested is contemplated herein.
  • Non-limiting examples include NIDDM, gestational diabetes, impaired glucose tolerance, impaired fasting glucose, Syndrome X, hyperglycemia, obesity, cardiovasular disease, atheriosclerosis, hypertriglyceridemia, dyslipidemia, hyperinsulinemia, nephropathy, neuropathy, retinopathy, ischemia, stroke and fatty liver disease.
  • Subjects to be treated include mammalian subjects: humans, primates, livestock animals (including cows, horses, sheep, pigs and goats), companion animals (including dogs, cats, rabbits, guinea pigs), and captive wild animals.
  • Laboratory animals such as rabbits, mice, rats, guinea pigs and hamsters are also contemplated as they may provide a convenient test system.
  • Non-mammalian species such as birds, amphibians and fish may also be contemplated in certain embodiments of the invention.
  • a particularly contemplated subject is a human subject.
  • the compounds of the invention are administered in an amount which, when administered according to the desired dosing regimen, at least partially attains the desired therapeutic effect, e.g. modulating glucose homeostasis, modulating cellular glucose uptake, treating insulin resistance, lowering blood glucose levels or treating a disease or condition, or symptom thereof, in which insulin resistance is involved.
  • treatment refers to therapeutic ameliorating or prophylactic treatment, including one or more of: alleviating or ameliorating the symptoms of, preventing or delaying the onset of, inhibiting the progression of, or halting or reversing altogether the onset or progression of the particular disorder or condition, or one or more symptoms thereof, being treated.
  • Suitable dosage amounts and dosing regimens can be determined by the attending physician and may depend on the particular condition being treated, the severity of the condition as well as the general age, health and weight of the subject, and may be in the range of from about 0.1 mg to about 1000 mg of active per day.
  • the active ingredient may be administered in a single dose or a series of doses.
  • Suitable dosages may contain about 1, 2.5, 5, 10, 20, 25, 50, 75, 100, 150, 200, 250 or 500 mg of active.
  • the active ingredient While it is possible for the active ingredient to be administered alone, it is preferable to present it as a composition, preferably as a pharmaceutical composition, with one or more pharmaceutically acceptable excipients or additives.
  • the present invention also relates to the use of a compound of formula (I) or a pharmaceutically acceptable salt or prodrug thereof in the manufacture of a medicament for modulating glucose homeostasis, modulating cellular glucose uptake, treating insulin resistance, or lowering blood glucose levels.
  • compositions are well known to those skilled in the art, see for example, Remington's Pharmaceutical Sciences, 18 th Edition, Mack Publishing, 1990.
  • the composition may contain any suitable carriers, diluents excipients or other additives.
  • compositions of the invention may also include other supplementary physiologically active agents.
  • compositions include those suitable for oral, rectal, nasal, topical (including dermal, buccal and sublingual), vaginal or parental (including subcutaneous, intramuscular, intravenous and intradermal) administration.
  • the compositions may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy. Such methods include the step of bringing into association the active ingredient with the carrier which constitutes one or more accessory ingredients. In general, the compositions are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both, and then if necessary shaping the product.
  • compositions of the present invention suitable for oral administration may be presented as discrete units such as capsules, sachets or tablets each containing a predetermined amount of the active ingredient; as a powder or granules; as a solution or a suspension in an aqueous or non-aqueous liquid; or as an oil-in-water liquid emulsion or a water-in-oil liquid emulsion.
  • a tablet may be made by compression or moulding, optionally with one or more accessory ingredients.
  • Compressed tablets may be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as a powder or granules, optionally mixed with a binder (e.g inert diluent), preservative disintegrant (e.g. sodium starch glycolate, cross-linked polyvinyl pyrrolidone, cross-linked sodium carboxymethyl cellulose) surface- active or dispersing agent.
  • a binder e.g inert diluent
  • preservative disintegrant e.g. sodium starch glycolate, cross-linked polyvinyl pyrrolidone, cross-linked sodium carboxymethyl cellulose
  • Moulded tablets may be made by moulding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
  • the tablets may optionally be coated or scored and may be formulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydroxypropylmethyl cellulose in varying proportions to provide the desired release profile. Tablets may optionally be provided with an enteric coating, to provide release in parts of the gut other than the stomach.
  • compositions suitable for topical administration in the mouth include lozenges comprising the active ingredient in a flavoured base, usually sucrose and acacia or tragacanth gum; pastilles comprising the active ingredient in an inert basis such as gelatin and glycerin, or sucrose and acacia gum; and mouthwashes comprising the active ingredient in a suitable liquid carrier.
  • Compositions suitable for topical administration to the skin may comprise the compounds dissolved or suspended in any suitable carrier or base and may be in the form of lotions, gel, creams, pastes, ointments and the like.
  • Suitable carriers include mineral oil, propylene glycol, polyoxyethylene, polyoxypropylene, emulsifying wax, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.
  • Transdermal patches may also be used to administer the compounds of the invention.
  • compositions for rectal administration may be presented as a suppository with a suitable base comprising, for example, cocoa butter, glycerin, gelatin or polyethylene glycol.
  • compositions suitable for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams or spray formulations containing in addition to the active ingredient such carriers as are known in the art to be appropriate.
  • compositions suitable for parenteral administration include aqueous and non-aqueous isotonic sterile injection solutions which may contain anti-oxidants, buffers, bactericides and solutes which render the composition isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents.
  • the compositions may be presented in unit-dose or multi-dose sealed containers, for example, ampoules and vials, and may be stored in a freeze-dried (lyophilised) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use.
  • Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described.
  • compositions of this invention are those containing a daily dose or unit, daily sub- dose, as herein above described, or an appropriate fraction thereof, of the active ingredient.
  • the compositions of this invention may include other agents conventional in the art having regard to the type of composition in question, for example, those suitable for oral administration may include such further agents as binders, sweeteners, thickeners, flavouring agents disintegrating agents, coating agents, preservatives, lubricants and/or time delay agents.
  • suitable sweeteners include sucrose, lactose, glucose, aspartame or saccharine.
  • Suitable disintegrating agents include corn starch, methylcellulose, polyvinylpyrrolidone, xanthan gum, bentonite, alginic acid or agar.
  • Suitable flavouring agents include peppermint oil, oil of wintergreen, cherry, orange or raspberry flavouring.
  • Suitable coating agents include polymers or copolymers of acrylic acid and/or methacrylic acid and/or their esters, waxes, fatty alcohols, zein, shellac or gluten.
  • Suitable preservatives include sodium benzoate, vitamin E, alpha-tocopherol, ascorbic acid, methyl paraben, propyl paraben or sodium bisulphite.
  • Suitable lubricants include magnesium stearate, stearic acid, sodium oleate, sodium chloride or talc.
  • Suitable time delay agents include glyceryl monostearate or glyceryl distearate.
  • the present invention also relates to prodrugs of formula (I). Any compound that is a prodrug of a compound of formula (I) is within the scope and spirit of the invention.
  • prodrug is used in its broadest sense and encompasses those derivatives that are converted in vivo, either enzymatically or hydrolytically, to the compounds of the invention. Such derivatives would readily occur to those skilled in the art, and include, for example, compounds where a free thiol or hydroxy group is converted into an ester, such as an acetate, or thioester or where a free amino group is converted into an amide.
  • acylating the compounds of the invention for example to prepare ester and amide prodrugs, are well known in the art and may include treatment of the compound with an appropriate carboxylic acid, anhydride or chloride in the presence of a suitable catalyst or base.
  • Esters of carboxylic acid (carboxy) groups are also contemplated. Suitable esters C 1-6 alkyl esters; C ⁇ ⁇ alkoxymethyl esters, for example methoxymethyl or ethoxymethyl; Ci -6 alkanoyloxymethyl esters, for example, pivaloyloxymethyl; phthalidyl esters; C 3 .
  • cycloalkoxycarbonylC 1-6 alkyl esters for example, 1- cyclohexylcarbonyloxyethyl; l,3-dioxolen-2-onylmethyl esters, for example, 5-methyl-l,3- dioxolen-2-onylmethyl; and Ci- ⁇ alkoxycarbonyloxyethyl esters, for example, 1- methoxycarbonyloxyethyl.
  • Prodrugs of amino functional groups include amides (see, for example, Adv. BioSci., 1979, 20, 369, Kyncl, J. et al), enamines (see, for example, J. Pharm. Sci., 1971, 60, 1810, Caldwell, H.
  • Suitable pharmaceutically acceptable salts of compounds of formula (I) include, but are not limited to salts of pharmaceutically acceptable inorganic acids such as hydrochloric, sulphuric, phosphoric nitric, carbonic, boric, sulfamic, and hydrobromic acids, or salts of pharmaceutically acceptable organic acids such as acetic, propionic, butyric, tartaric, maleic, hydroxymaleic, fumarie, maleic, citric, lactic, mucic, gluconic, benzoic, succinic, oxalic, phenylacetic, methanesulphonic, toluenesulphonic, benezenesulphonic, salicyclic sulphanilic, aspartic, glutamic, edetic, stearic, palmitic, oleic, lauric, pantothenic, tannic, ascorbic and valeric acids.
  • pharmaceutically acceptable inorganic acids such as hydrochloric, sulphuric, phosphoric
  • Base salts include, but are not limited to, those formed with pharmaceutically acceptable cations, such as sodium, potassium, lithium, calcium, magnesium, ammonium and alkylammonium.
  • Basic nitrogen-containing groups may be quarternised with such agents as lower alkyl halide, such as methyl, ethyl, propyl, and butyl chlorides, bromides and iodides; dialkyl sulfates like dimethyl and diethyl sulfate; and others.
  • solvate refers to a complex or aggregate formed by one or more molecules of a solute, ie compounds contemplated by the invention, and one or more molecules of a solvent.
  • Suitable solvents are well understood in the art and include for example, of water, ie to form hydrates, and common organic solvents such as alcohols (methanol, ethanol, isopropanol) and acetic acid. Methods of solvation are generally known within the art, for example, recrystallisation from an appropriate solvent.
  • Suitable agents may include insulin sensitisers, glucose resorption/uptake inhibitors and the classes and compounds identified in US 2005/0037981 , particularly Table 2, the contents of which are incorporated herein in their entirety.
  • compositions may also be presented for use in veterinary compositions. These may be prepared by any suitable means known in the art. Examples of such compositions include those adapted for:
  • oral administration eg drenches including aqueous and nonaqueous solutions or suspensions
  • external application eg drenches including aqueous and nonaqueous solutions or suspensions
  • tablets eg boluses, powders, granules, pellets for admixture with feedstuffs, pastes for application to the tongue
  • boluses eg drenches including aqueous and nonaqueous solutions or suspensions
  • parenteral administration eg subcutaneous, intramuscular or intravenous injection as a sterile solution or suspension
  • the bark and leaves of Alphitonia zizqphoides, Morinda citrifolia, Neisoperma oppositifolium, Pometia pinnata, Rhus taitensis, Syzygium malaccense, Terminalia catappa and Thespia populnea were dried and ground to form the plant material for subsequent extraction.
  • the plant extract used for both in vivo and in vitro studies was prepared as follows: lOOg of ground, dried plant material above was mixed thoroughly with IL of distilled water and maintained at 70°C for 24h. The mixture was then filtered and allowed to cool to room temperature. For all experiments the plant extract was prepared that day, and any unused plant extract was discarded at the end of the day.
  • Body weight and food intake were measured daily, while blood was collected from the tail vein for the measurement of glucose and insulin at the start and end of the 11 day experimental period.
  • Treatment with the plant extract tended to reduce plasma insulin concentration in the plant extract treated groups (by 22% in the lean, non-diabetic and by 31% in the obese, diabetic animals; Table 2), however these changes were not statistically significant.
  • 3T3-L1 adipocytes were differentiated and cultured in DMEM containing 5% fetal calf serum using standard procedures.
  • the cells were washed with Krebs Ringer Buffer Solution for 2h, and then insulin stimulated (0-10OnM) for 30min before addition of the plant extract at concentrations of 0.005-0.5% (v/v) and 14 C-2- deoxyglucose. After 15min the reaction was stopped with ice-cold PBS containing phloretin and SDS, and radioactivity counted immediately using a beta counter.
  • the plant extract stimulated lipogenesis in these cells at insulin concentrations of 0-1OnM (Table 4).
  • the plant extract had no discernible effect on lipogenesis in these cells, which was likely to be maximally stimulated at these concentrations of insulin.
  • the fteeze-dried plant extract (3g) was fractionated by HPLC/ESI-MS-ELSD under the conditions described in Table 5.
  • Table 5 Conditions for preparative chromatographic fractionation.
  • Preparative chromatographic (HPLC) fractionation resulted in 42 fractions of the plant extract. Each was collected in a 4-ml brown glass vial, and the water and buffer were removed online via solid phase extraction. The mass of material in each fraction ranged from 0.23 to 126.32 mg, and the total combined weight of all 42 fractions was approximately 1.7 g (accounting for ⁇ 56% of the 3 g used).
  • the plant extract fractions were examined in the lipogenesis (glucose incorporation into lipid) bioassay, measuring the amount of glucose converted into triglyceride using radioactive M C-glucose. Briefly, 3T3-L1 adipocytes were incubated in starvation media overnight followed by 30 minute incubation in DPBS buffer. Next, the DPBS buffer was removed, and treatment (freeze-dried plant extract, or its reconstituted fractions) was added to each well for 90 minutes in the presence of 1 nM insulin. Then, treatments were removed, and the cells were washed twice before being lysed with a lipid-extracting reagent.
  • lipogenesis glucose incorporation into lipid
  • the lipid extract was then transferred to 2 ml-tube containing water and heptane. Thereafter, the mixture was vigorously vortexed, and vaitrifuged to separate the lipid and water fractions.
  • the lipid fraction (heptane soluble layer, upper phase) was added to 4 ml scintillation fluid to count the radioactivity using the ⁇ -counter Tri-carb ® 2900TR Liquid Scintillation Analyser (Packard; Packard Bioscience B.V., Groningen, Netherlands).
  • Fraction 18 in particular had an effect on the incorporation of glucose into lipid in 3T3-L1 cells.
  • 3T3-L1 fibroblasts are grown in 10cm dishes and cultured in DMEM (4.5 g/L glucose) supplemented with 10% (v/v) heat-inactivated fetal bovine serum (FBS), 50 IU/ml pencillin and 50 ⁇ g/ml streptomycin.
  • FBS heat-inactivated fetal bovine serum
  • 50 IU/ml pencillin 50 IU/ml pencillin
  • streptomycin 50 IU/mlin
  • Cells for differentiation into adipocytes were maintained at post-confluence for 2 days, and then induced to differentiate by the addition of DMEM containing 10% FBS, 2 ⁇ g/ml insulin, 0.25 ⁇ M dexamethasone and 0.5 niM 3-isobutyl-l- methylxanthine.
  • the induction medium was replaced with fresh DMEM containing 10% FBS and 2 ⁇ g/ml insulin for another 3 days.
  • Adipocytes were maintained in DMEM with 5% FBS for 2 days thereafter.
  • Adipocytes were used 8-12 days after initiation of differentiation, after which time greater than 80% of fibroblasts had differentiated into mature adipocytes.
  • the reaction was stopped upon addition of ice-cold 80 ⁇ g/ml phloretin in PBS, pH 7.4 and cells solublised in 0.03% (w/v) SDS. Counts per minute (cpm) were measured by scintillator counter. Each measurement was performed in quadruplet in 1 assay, and each assay was repeated independently 3 times.
  • gene expression signatures were used as an endpoint for secondary in vitro screening studies. Specifically, microarray technology was used to characterise the effects of the plant extract fractions on gene expression in 3T3-L1 adipocytes.
  • Hybidisation involved indirect labelling of target cDNA and reference cDNA with fluorescent dyes Cy 5 and Cy3 respectively (Amersham Pharmacia Biotech).
  • Total RNA was extracted using the TRIzol method.
  • the RNA was purified using RNeasy midi columns (Qiagen). 20 ⁇ g of RNA per sample was processed using the Superscript Indirect cDNA labelling system (Invitrogen) as per manufacturer's instructions.
  • the cDNA was hybridised in a 40 ⁇ l reaction containing combined target and reference fluorescently labelled cDNAs, 5X SSC (Invitrogen), 16 ⁇ g PolydA (Amersham), 5X Denhardt's solution (Invitrogen), 4.8 ⁇ g yeast tRNA (SIGMA) and 0.01% Sodium dodecyl sulfate (Invitrogen).
  • the reaction was heated to 98 0 C for 2 min and maintained at 6O 0 C until used.
  • Microarrays were placed into a hybridisation chamber containing lOO ⁇ l of 2% SSC. A coverslip was placed on top of the microarray slide and the whole reaction mixture was loaded.
  • Hybridisation chambers were secured and placed in a waterbath within a hybridisation oven at 42 0 C for 16 hr. Following hybridisation, the array slides were washed with gentle agitation to ensure all unbound cDNA was removed. The washing procedure included gentle agitation for 2 minutes in solution 1 (0.5X SSC, 0.1% SDS) followed by 2 minutes in solution 2 (0.5X SSC, 0.01% SDS) and finally 2 minutes in solution 3 (0.06X SSC). Slides were centrifuged at 1500rpm for 1 minute to dry.
  • Microarrays were scanned using GenePix 4000B (Axon Instruments) scanner and microarray data was analysed using GenePix Pro 5.1 and Acuity 4.0 software (Axon Instruments). The data were analysed using linear discriminant analysis to identify a subset of genes that characterised the effects of the plant extract fractions on gene expression profiles ("gene expression signature"). Furthermore, genes were grouped according to function and assessment of trends within biological pathways was conducted.
  • Fraction 18 (cells treated with 0.5% w/v Fraction 18 for 24h) appeared to have significant effects on a number of genes associated with glucose metabolism, including those involved with adipogenesis (C/EBP ⁇ increased by 40%, SREBP2 increased by 22%), insulin signalling (PKC ⁇ increased by 54%, PIK3R2 increased by 32%, GSK3A increased by 10%, SORBSl increased by 24%).
  • Glycogen synthesis GYS 1 increased by 20%
  • glycolysis DLAT increased by 49%
  • PDK4 increased by 10%
  • GTT increased by 13%)
  • mitochondrial activity MFN2 increased by 20%
  • Fraction 18 appeared to contain a chemical entity with a m/z value of 517, and this was by far the most abundant entity observed following electrospray ionisation.
  • Partial linkage analysis revealed the presence of aromatic protons and a six membered aromatic carbon ring.
  • Using proprietary computer software a search of known structures was conducted for those that comprised a mass of -516, an empirical formula of 27C, 34H, 2N and 90 and which contain an aromatic ring.
  • 3T3-L1 fibroblasts are grown in 10cm dishes and cultured in DMEM (4.5 g/L glucose) supplemented with 10% (v/v) heat-inactivated fetal bovine serum (FBS), 50 IU/ml pencillin and 50 ⁇ g/ml streptomycin.
  • FBS heat-inactivated fetal bovine serum
  • 50 IU/ml pencillin 50 IU/ml pencillin
  • streptomycin 50 IU/mlin
  • Cells for differentiation into adipocytes were maintained at post-confluence for 2 days, and then induced to differentiate by the addition of DMEM containing 10% FBS, 2 ⁇ g/ml insulin, 0.25 ⁇ M dexamethasone and 0.5 mM 3-isobutyl-l- methylxanthine.
  • the induction medium was replaced with fresh DMEM containing 10% FBS and 2 ⁇ g/ml insulin for another 3 days.
  • Adipocytes were maintained in DMEM with 5% FBS for 2 days thereafter. Adipocytes were used 8-12 days after initiation of differentiation, after which time greater than 80% of fibroblasts had differentiated into mature adipocytes.
  • SA strictosidinic acid
  • the compound was administered once only by oral gavage on Day 0 of the study. Administration was performed at a dosing volume of 10 mL/kg, at dosing concentrations of 31, 62.5, 125, and 250 mg/kg.
  • Each animal's body weight was measured immediately prior to dosing each day.
  • the actual volume administered to each mouse was calculated and adjusted based on the body weight.
  • mice Starting with the lowest concentration (31 mg/kg, Group 1), one mouse was treated, followed by a 2 hour observation period. As no adverse effects were observed during this time, the remaining mice in the group were treated. Twenty four hours after the first concentration was administered, one mouse from the next dose concentration (62.5 mg/kg, Group 2) was treated. As no adverse effects were observed during a 2 hour observation period, the remaining mice in the group were treated. The process was continued until all dose concentrations were administered (125 mg/kg, Group 3 and 250 mg/kg, Group 4).
  • the acute tolerated dose is defined as the highest dose (administered as a single dose) that does not induce a weight loss in an individual animal in excess of 15% or cause death or severe morbidity, at any time during the study period.
  • mice Sixteen female Balb/c mice were randomised into 4 treatment groups, each with 4 mice. Each group had similar mean starting body weight. The groups were allocated a different treatment regimen of strictosidinic acid, administered once only, by oral gavage. The compound was administered at a volume of 10 mL/kg, at dosing concentrations of 31, 62.5, 125 and 250 mg/kg.
  • Body weight measurements were made daily from Day 0 of the study and were continued until 7 days after treatment was finished, and are detailed in Table A.
  • the ATD for strictosidinic acid administered by oral gavage was not achieved in this study, and is therefore determined to be >250 mg/kg (the highest dose concentration tested in the study).
  • Table A Body weight data for ATD Study: strictosidinic acid in Balb/c mice
  • mice Male C57B1/6J mice were obtained at 8 weeks of age from Animal Resources Centre (Canning Vale, Western Australia). Animals were individually housed and allowed free movement and ad libitum access to water and food. Animals were maintained on a 12 hour light (6 am-6 pm) and 12 hour dark (6 pm-6 am) cycle. After two weeks acclimatizing, mice were fed a high fat rich diet (Research Diets, Inc. [New Jersey, USA] with a total energy density of 4.7 kcal/g. Caloric distribution in the diet was 20% from protein, 35 % from carbohydrates, and 45 % from fat) for 11 weeks.
  • a high fat rich diet Research Diets, Inc. [New Jersey, USA] with a total energy density of 4.7 kcal/g. Caloric distribution in the diet was 20% from protein, 35 % from carbohydrates, and 45 % from fat
  • mice in groups of 6 were treated with doses selected from: vehicle, 25mg/kg, 50mg/kg, 100mg/kg and 175mg/kg of Strictosidinic acid in
  • Data represent the mean ⁇ SE of 4-6 animals per group.
  • Intraperitoneal Glucose Tolerance Test Blood glucose concentrations during ipGTT
  • Table C Effect of Strictosidinic acid treatment on area under plasma glucose curve during ipGTT in diet-induced obese mice
  • Intraperitoneal Glucose Tolerance Test Plasma insulin concentrations during ipGTT
  • Plasma insulin concentrations were determined using a commercially-available insulin ELISA kit (Crystal Chem) which was used according to the manufacturer's instructions. The results are presented in Table D.
  • Table D Effect of Strictosidinic acid treatment on plasma insulin concentration during ipGTT in diet-induced obese mice
  • Data represent the mean ⁇ SE of 5-6 animals per group. *unpaired ttest value vs vehicle at specified timepoint.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Diabetes (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Genetics & Genomics (AREA)
  • Biotechnology (AREA)
  • Epidemiology (AREA)
  • Emergency Medicine (AREA)
  • Endocrinology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Hematology (AREA)
  • Obesity (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention particularly relates to methods of regulating glucose homeostasis and to the therapeutic or prophylactic treatment of diseases and conditions, such as diabetes, syndrome X, obesity, hyperglycaemia, cardiovascular disease, vascular disease and kidney disease, in which impaired glucose uptake due to insulin resistance is involved or implicated. The present invention further relates to compounds and agents and compositions thereof for use in the treatment methods.

Description

METHODS FOR REGULATING GLUCOSE HOMEOSTASIS AND AGENTS THEREFOR
FIELD OF THE INVENTION
The present invention relates generally to the field of therapy. The invention particularly relates to methods of regulating glucose homeostasis and to the therapeutic or prophylactic treatment of diseases and conditions, such as diabetes, syndrome X, obesity, hyperglycaemia, cardiovascular disease, vascular disease and kidney disease, in which impaired glucose uptake due to insulin resistance is involved or implicated. The present invention further relates to compounds and agents and compositions thereof for use in the treatment methods.
DESCRIPTION OF THE PRIOR ART •
The reference in this specification to any prior publication (or information derived from it), or to any matter which is known, is not, and should not be taken as an acknowledgment or admission or any form of suggestion that that prior publication (or information derived from it) or known matter forms part of the common general knowledge in the field of endeavour to which this specification relates.
Bibliographic details of references provided in the subject specification are listed at the end of the specification.
Glucose is the body's preferred energy source. Once entered into the blood stream, it requires the assistance of the insulin to enter hepatic, muscle and adipose cells in order to be stored or utilised. In a healthy individual, glucose homeostasis is controlled primarily by insulin. As blood glucose levels rise, such as after eating, specialised β-cells within the pancreas release insulin which promotes glucose uptake, intracellular metabolism and glycogen synthesis by the body's target tissues. Thus, in healthy individuals, blood glucose concentrations are strictly controlled, typically in the range of 80-110 mg/dl. However, where the pancreas produces an inadequate insulin response, or the target cells do not respond appropriately to the insulin produced, the glucose cannot enter the body's cells. This results in a rapid accumulation of glucose in the blood stream (hyperglycemia).
High blood glucose levels over time may cause cardiovascular disease, retinal damage, renal failure, nerve damage, erectile disfunction and gangrene (with the risk of amputation). Furthermore, in the absence of available glucose, cells turn to fats as an alternative energy source. Resulting ketones, a product of fat hydrolysis, can accumulate in the blood stream instigating hypotension and shock, coma and even death.
Chronically elevated blood glucose levels (greater than about 126 mg/dl) from either inadequate insulin secretion and/or an inadequate response or sensitivity to insulin is referred to as diabetes, a disease which is now suffered by more than 10 percent of adults in the USA. The disease may be characterised by persistent hyperglycemia, polyuria, polydipsia and/or hyperphagia, chronic microvascular complications such as retinopathy, nephropathy and neuropathy, and macrovascular complications, such as hyperlipidemia and hypertension which can lead to blindness, end-stage renal disease, limb amputation and myocardial infarction. The three most common types of diabetes are type 1, type 2 and gestational.
Type 1, known as insulin dependent diabetes mellitus (IDDM), or juvenile-onset diabetes, occurs in 10-15% of all cases. It is most commonly diagnosed in children and adolescents but can occur in young adults as well. It is characterised by β-cell destruction resulting in a loss of insulin secretory function. Most cases relate to autoimmune destruction of the β- cells. Treatment is via insulin injection and must be continued indefinitely.
In contrast, in type 2 diabetes, known as non-insulin dependent diabetes mellitus (NIDDM) or late-onset diabetes, insulin levels are initially normal but the body's target cells lose their responsiveness to insulin. This is known as insulin resistance or insensitivity. By way of compensation, the pancreas begins to secrete excess insulin, however, in time the pancreas becomes less able to produce enough insulin resulting in chronic hyperglycaemia. Initial symptoms are typically milder than for type 1 and the condition may go undiagnosed for years before more severe symptoms present. Lifestyle (poor diet and inactivity) is considered to be a major mediating factor of the condition, although a genetic predisposition increases the risk. Treatment focuses on lifestyle modification, insulin therapy and/or anti-diabetic agents such as insulin sensitisers and insulin releasers.
Gestational diabetes occurs in about 2-5% of all pregnancies. It is temporary, but if untreated may cause foetal complications. Most sufferers make a complete recovery after the birth. However, a proportion of women who develop gestational diabetes go on to develop type 2 diabetes.
Other, less common, causes of diabetes include genetic defects in β-cells, genetically related insulin resistance, diseases of the pancreas, hormonal defects, malnutrition and chemical or drug influences.
Impaired glucose tolerance and impaired fasting glucose, are pre-type 2 diabetic states, closely related to type 2 and occur when the blood glucose level is higher than normal but not high enough to be classified as diabetes (about 110-126 mg/dl). As with type 2, the body produces insulin but in an insufficient amount or the target tissues are unresponsive to the insulin produced.
Syndrome X, also known as Insulin Resistance Syndrome (IRS) is a cluster of risk factors for heart disease and is associated with insulin resistance. It presents symptoms or risk factors for the development of type 2 diabetes and heart disease including obesity, atheriosclerosis, hypertriglyceridemia, low HDL cholesterol, hyperinsulinemia, hyperglycaemia, hypertension, impaired glucose tolerance and impaired fasting glucose.
High blood glucose levels and insulin resistance are also associated with fatty liver disease, which can progress to chronic inflammation, fibrosis and cirrhosis.
It is therefore apparent that insulin resistance, or insensitivity, can play a significant role in diabetic and other hyperglycemic-related conditions.
The latest WHO estimate (for the number of people with diabetes, worldwide, in 2003) is 194 million. This is expected to increase to at least 330 million by 2025 and it is estimated that there are around 4 million deaths per year are related to the disorder.
Given the prevalence of diabetes and other diseases and conditions with which insulin resistance is associated, and their potential consequences, there remains a continuing need for new therapeutic or preventative treatments for insulin resistance.
SUMMARY OF THE INVENTION
Insulin sensitisers resensitise cells to the action of insulin thereby reducing blood glucose levels and triglyceride levels. The present invention is predicated on compounds related to strictosidine, a key intermediate in the biosynthesis of terpenoid indole alkaloids, as a new class of insulin sensitizers and provides new methods for modulating glucose uptake by a cell and treating insulin resistance and conditions associated with insulin resistance.
The compounds contemplated by the invention include those of Formula (I):
Figure imgf000006_0001
wherein,
is a single or a double bond;
R1-R4 are independently selected from hydrogen, hydroxy, thiol, halo, CO2H, carboxy ester, amino, nitro, cyano, amido, sulfoxide, sulfonamide, sulfonyl, sulfate, sulfonate, phosphate, phosphonate, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted carbocyclyl, optionally substituted heterocyclyl, optionally substituted acyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aryloxy, optionally substituted heteroaryloxy, optionally substituted carbocyclyloxy, optionally substituted heterocyclyloxy, optionally substituted aralkyloxy, optionally substituted acyloxy, optionally substituted alkylthio, optionally substituted alkenylthio, optionally substituted alkynylthio, optionally substituted arylthio, optionally substituted carbocyclylthio, optionally substituted aralkylthio, optionally substituted heteroarylthio, optionally substituted heterocyclylthio, optionally substituted acylthio or any 2 adjacent Ri-R4 together form a -0-(CHi)n-O- or -NR-(CH2)n-NR-group wherein n is 1 or 2 and each R is independently H or C1-6alkyl;
R6 is absent when ""- is a double bond;
R5 and R6 (when present) independently are selected from hydrogen, optionally substituted alkyl, optionally substituted acyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted aralkyl, optionally substituted heterocyclyl, optionally substituted carbocyclyl, and amido;
R7 is selected from hydrogen, optionally substituted alkyl, CO2H, carboxy ester, amido, optionally substituted heteroaryl and optionally substituted aryl;
R8 is selected from hydrogen, optionally substituted alkyl, optionally substituted aryl, optionally substituted heteroaryl, CO2H, carboxy ester and SO3H;
R7a and R8a are independently selected from hydrogen, optionally substituted alkyl and optionally substituted aryl.
COX forms a group selected from optionally substituted acyl, CO2H, carboxy ester and amido, Y is selected from hydrogen, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclyl, optionally substituted carbocyclyl, optionally substituted acyl and a saccharide group,
Z is selected from optionally substituted at least C2alkyl, optionally substituted alkenyl and optionally substituted alkynyl,
and pharmaceutically acceptable salts and prodrugs thereof.
In a first aspect, the present invention thus provides a method of increasing glucose uptake by a cell comprising contacting said cell with a compound of Formula (I) or a pharmaceutically acceptable salt or prodrug thereof.
Methods for increasing glucose uptake by a cell, for example adipocytes and muscle and hepatic cells, can be performed in vivo or in vitro.
A second aspect of the invention relates to a method for regulating glucose homeostasis in a subject in need thereof comprising the administration of a compound of Formula (I) or a pharmaceutically acceptable salt or prodrug thereof.
In another aspect, the present invention provides a method of treating insulin resistance in a subject in need thereof comprising the administration of a compound of Formula (I) or a pharmaceutically acceptable salt or prodrug thereof.
A further aspect of the invention relates to a method of lowering blood glucose levels in a subject in need thereof comprising the administration of a compound of Formula (I) or a pharmaceutically acceptable salt or prodrug thereof.
Yet a further aspect of the invention provides a method for treating a disease or condition, or symptom thereof, in which insulin resistance is involved comprising the administration of a compound of Formula (I) or a pharmaceutically acceptable salt or prodrug thereof to subject in need thereof.
In yet another aspect, the invention provides a compound of Formula (I) or a pharmaceutically acceptable salt or prodrug thereof provided that the compound is not (3R) or (3S) strictosidine, strictosidine ethyl ester and allyl ester, strictosidinic acid, 5- carboxystrictosidine, 3,4-dehydro-5-carboxystrictosidine, 3,4-dehydro5- carbomethoxystrictosidine or gluco-acetylated forms thereof.
Further aspects of the invention provide a compound of Formula (I), or pharmaceutically acceptable salt or prodrug thereof for use in therapy, particularly for regulating glucose homeostasis, treating insulin resistance, lowering blood levels and/or treating a disease or condition, or one or more symptoms thereof, in which insulin resistance is involved.
The present invention also provides for agents and compositions comprising compounds of Formula (I), or pharmaceutically acceptable salt or prodrug thereof and the use of said compounds in treatment in the manufacture of medicaments for regulating glucose homeostasis, treating insulin resistance, lowering blood levels and/or treating a disease or condition, or one or more symptoms thereof, in which insulin resistance is involved. Advantageously, in certain embodiments, said compositions and medicaments comprise the compound of Formula (I), or pharmaceutically acceptable salt or prodrug thereof, as at least 50%, such as at least 90 or 95% of the active component.
DETAILED DESCRIPTION OF THE INVENTION
Throughout this specification and the claims which follow, unless the context requires otherwise, the word "comprise" and variations such as "comprises" and "comprising" will be understood to imply the inclusion of a stated integer or step or group of integers but not the exclusion of any other integer or step or group of integers.
The singular forms "a", "an" and "the" include plural aspects unless the context clearly dictates otherwise.
As used herein, the term "alkyl" or "alk", used either alone or in compound words denotes straight chain, branched or cyclic alkyl, preferably C1-20 alkyl, eg C1-10 or C1-6. Examples of straight chain and branched alkyl include methyl, ethyl, «-propyl, isopropyl, «-butyl, sec- butyl, t-butyl, n-pentyl, 1,2-dimethylpropyl, 1,1-dimethyl-propyl, hexyl, 4-methylpentyl, 1- methylpentyl, 2-methylpentyl, 3-methylpentyl, 1,1-dimethylbutyl, 2,2-dimethylbutyl, 3,3- dimethylbutyl, 1,2-dimethylbutyl, 1,3-dimethylbutyl, 1,2,2,-trimethylρropyl, 1,1,2- trimethylpropyl, heptyl, 5-methylhexyl, 1-methylhexyl, 2,2-dimethylpentyl, 3,3- dimethylpentyl, 4,4-dimethylpentyl, 1,2-dimethylpentyl, 1,3-dimethylpentyl, 1,4- dimethyl-pentyl, 1,2,3-trimethylbutyl, 1,1,2-trimethylbutyl, 1,1,3-trimethylbutyl, octyl, 6- methylheptyl, 1-methylheptyl, 1,1,3,3-tetramethylbutyl, nonyl, 1-, 2-, 3-, 4-, 5-, 6- or 7- methyl-octyl, 1-, 2-, 3-, 4- or 5-ethylheptyl, 1-, 2- or 3-propylhexyl, decyl, 1-, 2-, 3-, 4-, 5-, 6-, 7- and 8-methylnonyl, 1-, 2-, 3-, 4-, 5- or 6-ethyloctyl, 1-, 2-, 3- or 4-propylheptyl, undecyl, 1-, 2-, 3-, A-, 5-, 6-, 7-, 8- or 9-methyldecyl, 1-, 2-, 3-, A-, 5-, 6- or 7-ethylnonyl, 1-, 2-, 3-, 4- or 5-propylocytl, 1-, 2- or 3-butylheptyl, 1-pentylhexyl, dodecyl, 1-, 2-, 3-, A-, 5-, 6-, 7-, 8-, 9- or 10-methylundecyl, 1-, 2-, 3-, A-, 5-, 6-, 7- or 8-ethyldecyl, 1-, 2-, 3-, A-, 5- or 6-propylnonyl, 1-, 2-, 3- or 4-butyloctyl, 1-2-pentylheptyl and the like. Examples of cyclic alkyl include mono- or polycyclic alkyl groups such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl, cyclodecyl and the like. Where an alkyl group is referred to generally as "propyl", butyl" etc, it will be understood that this can refer to any of straight, branched and cyclic isomers where appropriate. An alkyl group may be optionally substituted by one or more optional substitutents as herein defined.
The term "alkenyl" as used herein denotes groups formed from straight chain, branched or cyclic hydrocarbon residues containing at least one carbon to carbon double bond including ethylenically mono-, di- or poly-unsaturated alkyl or cycloalkyl groups as previously defined, preferably C2-20 alkenyl (eg C2-1O or C2-6)- Examples of alkenyl include vinyl, allyl, 1-methylvinyl, butenyl, iso-butenyl, 3-methyl~2-butenyl, 1-pentenyl, cyclopentenyl, 1-methyl-cyclopentenyl, 1-hexenyl, 3-hexenyl, cyclohexenyl, 1-heptenyl, 3-heρtenyl, 1-octenyl, cyclooctenyl, 1-nonenyl, 2-nonenyl, 3-nonenyl, 1-decenyl, 3- decenyl, 1,3-butadienyl, l-4,pentadienyl, 1,3-cyclopentadienyl, 1,3-hexadienyl, 1,4- hexadienyl, 1,3-cyclohexadienyl, 1,4-cyclohexadienyl, 1,3-cycloheρtadienyl, 1,3,5- cycloheptatrienyl and 1,3,5,7-cyclooctatetraenyl. An alkenyl group may be optionally substituted by one or more optional substitutents as herein defined.
As used herein the term "alkynyl" denotes groups formed from straight chain, branched or cyclic hydrocarbon residues containing at least one carbon-carbon triple bond including ethynically mono-, di- or poly- unsaturated alkyl or cycloalkyl groups as previously defined. Unless the number of carbon atoms is specified the term preferably refers to C2-2O alkynyl (eg C2-10 or C2.6). Examples include ethynyl, 1-propynyl, 2-propynyl, and butynyl isomers, and pentynyl isomers. An alkynyl group may be optionally substituted by one or more optional substitutents as herein defined.
Terms written as " [group] oxy" refer to a particular group when linked by oxygen, for example, the terms "alkoxy", "alkenoxy", . "alkynoxy" and "aryloxy" and "acyloxy" respectively denote alkyl, alkenyl, alkynyl, aryl and acyl groups as hereinbefore defined when linked by an oxygen atom. Terms written as "[group]thio" refer to a particular group when linked by sulfur, for example, the terms "alkylthio", "alkenylthio", alkynylthio" and
"arylthio" respectively denote alkyl, alkenyl, alkynyl, aryl groups as hereinbefore defined when linked by a sulfur atom. Similarly, a term written as M[groupA]groupB" is intended to refer to a groupA when linked by a divalent form of groupB, for example, "hydroxyalkyl" is a hydroxy group when linked by an alkylene group.
The term "halogen" ("halo") denotes fluorine, chlorine, bromine or iodine (fluoro, chloro, bromo or iodo).
The term "aryl" (or "carboaryl)" or the abbreviated form "ar" used in compound words such as "aralkyl" denotes any of single, polynuclear, conjugated and fused residues of aromatic hydrocarbon ring systems. Examples of aryl include phenyl, biphenyl, terphenyl, quaterphenyl, naphthyl, tetrahydronaphthyl, anthracenyl, dihydroanthracenyl, benzanthracenyl, dibenzanthracenyl, phenanthrenyl, fluorenyl, pyrenyl, idenyl, azulenyl, chrysenyl. Preferred aryl include phenyl and naphthyl. An aryl group may be optionally substituted by one or more optional substituents as herein defined.
The term "carbocyclyl" includes any of non-aromatic monocyclic, polycyclic, fused or conjugated cyclic hydrocarbon residues, preferably C3-20 (e.g. C3-J0 or C3-8). The rings may be saturated, for example cycloalkyl, or may possess one or more double bonds (cycloalkenyl) and/or one or more triple bonds (cycloalkynyl). Particularly preferred carbocyclyl are 5-6-membered or bicyclic 9-10 membered ring systems. Suitable examples include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl, cyclodecyl, cyclopentenyl, cyclohexenyl, cyclooctenyl, cyclopentadienyl, cyclohexadienyl, cyclooctatetraenyl, indanyl, decalinyl and indenyl. A carbocyclyl group may be optionally substituted by one or more optional substituents as herein defined.
The term "heterocyclyl" when used alone or in compound words includes any of monocyclic, polycyclic, fused or conjugated hydrocarbon residues, preferably C3-20 (e.g. C3-10 or C3-8) wherein one or more carbon atoms are replaced by a heteroatom so as to provide a non-aromatic residue. Suitable heteroatoms include, O, N, S, P and Se5 particularly O, N and S. Where two or more carbon atoms are replaced, this may be by two or more of the same heteroatom or by different heteroatoms. The heterocyclyl group may be saturated or partially unsaturated, i.e. possess one or more double bonds. Particularly preferred heterocyclyl are 5-6 and bicyclic 9-10 membered heterocyclyl. Suitable examples of heterocyclyl groups may include azridinyl, oxiranyl, thiiranyl, azetidinyl, oxetanyl, thietanyl, 2H-pyrrolyl, pyrrolidinyl, pyrrolinyl, piperidyl, piperazinyl, morpholinyl, indolinyl, imidazolidinyl, imidazolinyl, pyrazolidinyl, thiomorpholinyl, dioxanyl, tetrahydrofuranyl, tetrahydropyranyl, tetrahydropyrrolyl, tetrahydrothiophenyl, pyrazolinyl, dioxalanyl, thiazolidinyl, isoxazolidinyl, dihydropyranyl, oxazinyl, thiazinyl, thiomorpholinyl, oxathianyl, dithianyl, trioxanyl, thiadiazinyl, dithiazinyl, trithianyl, azepinyl, oxepinyl, thiepinyl, indenyl, indanyl, 3H-indolyl, isoindolinyl, 4H-quinolazinyl, chromenyl, chromanyl, isochromanyl, pyranyl and dihydropyranyl. A heterocyclyl group may be optionally substituted by one or more optional substituents as herein defined.
The term "heteroaryl" includes any of monocyclic, polycyclic, fused or conjugated hydrocarbon residues, wherein one or more carbon atoms are replaced by a heteroatom so as to provide an aromatic residue. Preferred heteroaryl have 3-20 ring atoms, e.g. 3-10. Particularly preferred heteroaryl are 5-6 and 9-10 membered bicyclic ring systems. Suitable heteroatoms include, O, N, S, P and Se, particularly O, N and S. Where two or more carbon atoms are replaced, this may be by two or more of the same heteroatom or by different heteroatoms. Suitable examples of heteroaryl groups may include pyridyl, pyrrolyl, thienyl, imidazolyl, furanyl, benzothienyl, isobenzothienyl, benzofuranyl, isobenzofuranyl, indolyl, isoindolyl, pyrazolyl, pyrazinyl, pyrimidinyl, pyridazinyl, indolizinyl, quinolyl, isoquinolyl, phthalazinyl, 1,5-naphthyridinyl, quinozalinyl, quinazolinyl, quinolinyl, oxazolyl, thiazolyl, isothiazolyl, isoxazolyl, triazolyl, oxadialzolyl, oxatriazolyl, triazinyl, tetrazolyl and furazanyl. A heteroaryl group may be optionally substituted by one or more optional substituents as herein defined.
The term "acyl" either alone or in compound words denotes a group containing the moiety C=O (and not being a carboxylic acid, ester or amide) Preferred acyl includes C(O)-R, wherein R is hydrogen or an alkyl, alkenyl, alkynyl, aryl, heteroaryl, carbocyclyl, or heterocyclyl residue. Examples of acyl include formyl, straight chain or branched alkanoyl (eg. C1^o) such as, acetyl, propanoyl, butanoyl, 2-methylpropanoyl, pentanoyl, 2,2-dimethylpropanoyl, hexanoyl, heptanoyl, octanoyl, nonanoyl, decanoyl, undecanoyl, dodecanoyl, tridecanoyl, tetradecanoyl, pentadecanoyl, hexadecanoyl, heptadecanoyl, octadecanoyl, nonadecanoyl and icosanoyl; cycloalkylcarbonyl such as
• cyclopropylcarbonyl cyclobutylcarbonyl, cyclopentylcarbonyl and cyclohexylcarbonyl; aroyl such as benzoyl, toluoyl and naphthoyl; aralkanoyl such as phenylalkanoyl (e.g. phenylacetyl, phenylpropanoyl, phenylbutanoyl, phenylisobutylyl, phenylpentanoyl and phenylhexanoyl) and naphthylalkanoyl (e.g. naphthylacetyl, naphthylpropanoyl and naphthylbutanoyl]; aralkenoyl such as phenylalkenoyl (e.g. phenylpropenoyl, phenylbutenoyl, phenylmethacryloyl, phenylpentenoyl and phenylhexenoyl and naphthylalkenoyl (e.g. naphthylpropenoyl, naphthylbutenoyl and naphthylpentenoyl); aryloxyalkanoyl such as phenoxyacetyl and phenoxypropionyl; arylthiocarbanioyl such as phenylthiocarbamoyl; arylglyoxyloyl such as phenylglyoxyloyl and naphthylglyoxyloyl; arylsulfonyl such as phenylsulfonyl and napthylsulfonyl; heterocycliccarbonyl; heterocyclicalkanoyl such as thienylacetyl, thienylpropanoyl, thienylbutanoyl, thienylpentanoyl, thienylhexanoyl, thiazolylacetyl, thiadiazolylacetyl and tetrazolylacetyl; heterocyclicalkenoyl such as heterocyclicpropenoyl, heterocyclicbutenoyl, heterocyclicpentenoyl and heterocyclichexenoyl; and heterocyclicglyoxyloyl such as thiazolyglyoxyloyl and thienylglyoxyloyl. The R residue may be optionally substituted as described herein.
In this specification "optionally substituted" is taken to mean that a group may or may not be further substituted or fused (so as to form a condensed polycyclic group) with one, two, three or more of organic and inorganic groups, including those selected from: alkyl, alkenyl, alkynyl, carbocyclyl, aryl, heterocyclyl, heteroaryl, acyl, aralkyl, alkylaryl, alkylheterocyclyl, alkylheteroaryl, alkylcarbocyclyl, halo, haloalkyl, haloalkenyl, haloalkynyl, haloaryl, halocarbocyclyl, haloheterocyclyl, haloheteroaryl, haloacyl, haloaryalkyl, hydroxy, hydroxyalkyl, hydroxyalkenyl, hydroxyalkynyl, hydroxycarbocyclyl, hydroxyaryl, hydroxyheterocyclyL hydroxyheteroaryl, hydroxyacyl, hydroxyaralkyl, alkoxyalkyl, alkoxyalkenyl, alkoxyalkynyl, alkoxycarbocyclyl, alkoxyaryl, alkoxyheterocyclyl, alkoxyheteroaryl, alkoxyacyl, alkoxy aralkyl, alkoxy, alkenyloxy, alkynyloxy, aryloxy, carbocyclyloxy, aralkyloxy, heteroaryloxy, heterocyclyloxy, acyloxy, haloalkoxy, haloalkenyloxy, haloalkynyloxy, haloaryloxy, halocarbocyclyloxy, haloaralkyloxy, haloheteroaryloxy, haloheterocyclyloxy, haloacyloxy, nitro, nitroalkyl, nitroalkenyl, nitroalkynyl, nitroaryl, nitroheterocyclyl, nitroheteroaryl, nitrocarbocyclyl, nitroacyl, nitroaralkyl, amino (NH2), alkylamino, dialkylamino, alkenylamino, alkynylamino, arylamino, diarylamino, aralkylamino, diaralkylamino, acylamino, diacylamino, heterocyclamino, heteroarylamino, carboxy, carboxyester, amido, alkylsulphonyloxy, arylsulphenyloxy, alkylsulphenyl, arylsulphenyl, thio, alkylthio, alkenylthio, alkynylthio, arylthio, aralkylthio, carbocyclylthio, heterocyclylthio, heteroarylthio, acylthio, sulfoxide, sulfonyl, sulfonamide, aminoalkyl, aminoalkenyl, aminoalkynyl, aminocarbocyclyl, aminoaryl, aminoheterocyclyl, aminoheteroaryl, aminoacyl, aminoaralkyl, thioalkyl, thioalkenyl, thioalkynyl, thiocarbocyclyl, thioaryl, thioheterocyclyl, thioheteroaryl, thioacyl, thioaralkyl, carboxyalkyl, carboxyalkenyl, carboxyalkynyl, carboxycarbocyclyl, carboxyaryl, carboxyheterocyclyl, carboxyheteroaryl, carboxyacyl, carboxyaralkyl, carboxyesteralkyl, carboxyesteralkenyl, carboxyesteralkynyl, carboxyestercarbocyclyl, carboxyesteraryl, carboxyesterheterocyclyl, carboxyesterheteroaryl, carboxyesteracyl, carboxyesteraralkyl, amidoalkyl, amidoalkenyl, amidoalkynyl, amidocarbocyclyl, amidoaryl, amidoheterocyclyl, amidoheteroaryl, amidoacyl, amidoaralkyl, formylalkyl, formylalkenyl, formylalkynyl, formylcarbocyclyl, formylaryl, formylheterocyclyl, formylheteroaryl, formylacyl, formylaralkyl, acylalkyl, acylalkenyl, acylalkynyl, acylcarbocyclyl, acylaryl, acylheterocyclyl, acylheteroaryl, acylacyl, acylaralkyl, sulfoxidealkyl, sulfoxidealkenyl, sulfoxidealkynyl, sulfoxidecarbocyclyl, sulfoxidearyl, sulfoxideheterocyclyl, sulfoxideheteroaryl, sulfoxideacyl, sulfoxidearalkyl, sulfonylalkyl, sulfonylalkenyl, sulfonylalkynyl, sulfonylcarbocyclyl, sulfonylaryl, sulfonylheterocyclyl, sulfonylheteroaryl, sulfonylacyl, sulfonylaralkyl, sulfonamidoalkyl, sulfonamidoalkenyl, sulfonamidoalkynyl, sulfonamidocarbocyclyl, sulfonamidoaryl, sulfonamidoheterocyclyl, sulfonamidoheteroaryl, sulfonamidoacyl, sulfonamidoaralkyl, nitroalkyl, nitroalkenyl, nitroalkynyl, nitrocarbocyclyl, nitroaryl, nitroheterocyclyl, nitroheteroaryl, nitroacyl, nitroaralkyl, cyano, sulfate, sulfonate, phosphonate and phosphate groups. Optional substitution may also be taken to refer to where a CH2 group in a chain or ring is replaced by a carbonyl group (C=O) or where 2 adjacent carbon atoms are substituted by one end each of a -O-(CH2)n-O- or -NH-(CH2)n-NH- group, wherein n is 1 or 2.
Examples of optional substitutents include alkyl, (e.g. C1-6alkyl such as methyl, ethyl, propyl, butyl, cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl), hydroxyalkyl (e.g. hydroxymethyl, hydroxyethyl, hydroxypropyl), alkoxyalkyl (e.g. methoxymethyl, methoxyethyl, methoxypropyl, ethoxymethyl, ethoxy ethyl, ethoxypropyl etc) alkoxy (e.g. Ci-6 alkoxy such as methoxy, ethoxy, propoxy, butoxy, cyclopropoxy, cyclobutoxy), halo, trifluoromethyl, trichloromethyl, tribromomethyl, hydroxy, phenyl (which itself may be further substituted e.g., by one or more C1-6alkyl, halo, hydroxy, hydroxyCi-6alkyl, C1- 6alkoxy, haloC1-6alkyl, cyano, nitro, OC(O)C μβalkyl, and amino), benzyl (wherein benzyl itself may be further substituted e.g., by one or more of C1-6alkyl, halo, hydroxy, hydroxyd-βalkyl, C1-6alkoxy, haloCi-6alkyl, cyano, nitro, OC(O)Ci-6alkyl, and amino), phenoxy (wherein phenyl itself may be further substituted e.g., by one or more of C1- βalkyl, halo, hydroxy, hydroxyC1-6alkyl, C1-OaIkOXy, haloC^alkyl, cyano, nitro, OC(O)Ci- 6alkyl, and amino), benzyloxy (wherein benzyl itself may be further substituted e.g., by one or more of C1-6alkyl, halo, hydroxy, hydroxyC1-6alkyl, C1-6alkoxy, haloCi-βalkyl, cyano, nitro, OC(O)C 1-6alkyl, and amino), amino, alkylamino (e.g. C1-6alkyl, such as methylamino, ethylamino, propylamino etc), dialkylamino (e.g. C1-6alkyl, such as dimethylamino, diethylamino, dipropylamino), acylamino (e.g. NHC(O)CH3), phenylamino (wherein phenyl itself may be further substituted e.g., by one or more of C1- 6alkyl, halo, hydroxy hydroxyCi^alkyl, Ci-6alkoxy, haloC1-6alkyl, cyano, nitro, OC(O)Ci- 6alkyl, and amino), nitro, formyl, -C(O)-alkyl (e.g. C1-6 alkyl, such as acetyl), O-C(O)- alkyl (e.g. C^alkyl, such as acetyloxy), benzoyl (wherein benzyl itself may be further substituted e.g., by one or more of C1-6alkyl, halo, hydroxy hydroxyC1-6alkyl, C^alkoxy, haloC1-6alkyl, cyano, nitro, 0C(0)Ci-6alkyl, and amino), benzoyloxy (wherein benzyl itself may be further substituted e.g., by one or more of Chalky!, halo, hydroxy hydroxyCμ 6alkyl, C1^aIkOXy, haloCt-galkyl, cyano, nitro, 0C(0)Ci-6alkyl, and amino), CO2H, C02alkyl (e.g. C1-6 alkyl such as methyl ester, ethyl ester, propyl ester, butyl ester), COaphenyl (wherein phenyl itself may be further substituted e.g., by one or more of C1- 6alkyl, halo, hydroxy, hydroxyCi.6alkyl, C1-6alkoxy, haloCi-6alkyl, cyano, nitro, OC(O)C1- 6alkyL and amino), CC^benzyl (wherein benzyl itself may be further substituted e.g., by one or more of C1-6alkyl, halo, hydroxy, hydroxyC^ealkyl, Ci-6alkoxy, haloC1-6alkyl, cyano, nitro, 0C(O)C1-6alkyl, and amino), CONH2, CONHphenyl (wherein phenyl itself may be further substituted e.g., by one or more of C1-6alkyl, halo, hydroxy, hydroxyd, 6alkyl, C1-6alkoxy, haloC^alkyl, cyano, nitro, OC(O)Ci_6alkyl5 and amino), CONHbenzyl (wherein benzyl itself may be further substituted e.g., by one or more of C1-6alkyl, halo, hydroxy hydroxyC1-6alkyl, C1-6alkoxy, haloC1-6alkyl, cyano, nitro, OC(O)C 1-6alkyl, and amino), CONHalkyl (e.g. C1-6 alkyl such as methyl ester, ethyl ester, propyl ester, butyl amide) CONHdialkyl (e.g.
Figure imgf000017_0001
6alkyl- and
Figure imgf000017_0002
thioalkyl (e.g., HSd.6alkyl-), carboxyalkyl (e.g., Hθ2CC1-6alkyl-), carboxyesteralkyl (e.g., C1-6alkylO2CC1-6alkyl-), amidoalkyl (e.g., H2N(O)CC i-βalkyl-, H(C1.6alkyl)N(O)CC1.6alkyl-), formylalkyl (e.g., OHCCi-6alkyl-), acylalkyl (e.g., C1-6alkyl(O)CC1-6alkyl-), nitroalkyl (e.g., O2NCι.6a\kyl-), sulfoxidealkyl (e.g., R(O)Sd.6alkyl, such as C1-6alkyl(O)SC1-6alkyl-), sulfonylalkyl (e.g., R(O)2SC1- 6alkyl- such as C1-6alkyl(O)2SC1-6alkyl-), sulfonamidoalkyl (e.g., H2N(O)SC 1-6alkyl, H(Ci- 6alkyl)NS(O)C1-6alkyl-), replacement of CH2 with C=O, and where 2 adjacent carbon atoms are substituted by one end each of a -0-(CHa)n-O- or -NH-(CH2)n-NH- group, wherein n is 1 or 2.
The term "sulfoxide", either alone or in a compound word, refers to a group -S(O)R wherein R is selected from hydrogen, alkyl, alkenyl, alkynyl, aryl, heteroaryl, heterocyclyl, carbocyclyl, and aralkyl, each of which may be optionally substituted. Examples of R include hydrogen, C1-20alkyl, phenyl and benzyl.
The term "sulfonyl", either alone or in a compound word, refers to a group S(O)2-R, wherein R is selected from hydrogen, alkyl, alkenyl, alkynyl, aryl, heteroaryl, heterocyclyl, carbocyclyl, acyl, and aralkyl, each of which may be optionally substituted. Examples of R include hydrogen, C^oalkyl, phenyl and benzyl.
The term "sulfonamide", either alone or in a compound word, refers to a group S(O)2NRR wherein each R is independently selected from hydrogen, alkyl, alkenyl, alkynyl, aryl, heteroaryl, heterocyclyl, carbocyclyl, acyl, and aralkyl, each of which may be optionally substituted. Examples of R include C1-20alkyl, phenyl and benzyl. In one embodiment at least one R is hydrogen. In another embodiment, both R are hydrogen.
A "sulfate" group refers to a group OS(O)2OR wherein each R is independently selected from hydrogen, alkyl, alkenyl, alkynyl, aryl, heteroaryl, heterocyclyl, carbocyclyl, acyl, and aralkyl, each of which may be optionally substituted. Examples of R include hydrogen, C1-2oalkyl, phenyl and benzyl.
The term "sulfonate" refers to a group OS(O)2R wherein each R is independently selected from hydrogen, alkyl, alkenyl, alkynyl, aryl, heteroaryl, heterocyclyl, carbocyclyl, acyl, and aralkyl, each of which may be optionally substituted. Examples of R include hydrogen, C1-20alkyl, phenyl and benzyl.
The term "phosphonate" refers to a group P(O)(OR2) wherein R is independently selected from hydrogen, alkyl, alkenyl, alkynyl, aryl, heteroaryl, heterocyclyl, carbocyclyl, acyl, and aralkyl, each of which may be optionally substituted. Examples of R include hydrogen, C^oalkyl, phenyl and benzyl.
The term "phosphate" refers to a group OP(O)(OR)2 wherein R is independently selected from hydrogen, alkyl, alkenyl, alkynyl, aryl, heteroaryl, heterocyclyl, carbocyclyl, acyl, and aralkyl, each of which may be optionally substituted. Examples of R include hydrogen, C1-2oalkyl, phenyl and benzyl.
The term "thio" is intended to include groups of the formula "SR" wherein R can be hydrogen (thiol), alkyl, alkenyl, alkynyl, aryl, carbocyclyl, heteroaryl, heterocyclyl, aralkyl, and acyl, each of which may be optionally substituted. Examples of R include hydrogen, C1-2oalkyl, phenyl and benzyl.
The term, "amino" is used here in its broadest sense as understood in the art and includes groups of the formula NRARB wherein RA and RB may be any independently selected from hydrogen, hydroxy alkyl, alkoxyalkyl, alkenyl, alkynyl, aryl, carbocyclyl, heteroaryl, heterocyclyl, aralkyl, and acyl, each of which may be optionally substituted. RA and RB, together with the nitrogen to which they are attached, may also form a monocyclic, or polycyclic ring system e.g. a 3-10 membered ring, particularly, 5-6 and 9-10 membered systems. Examples of "amino" include NH2, NHalkyl (e.g. Ci-20alkyl), NHalkoxyalkyl, NHaryl (e.g. NHphenyl), NHaralkyl (e.g. NHbenzyl), NHacyl (e.g. NHC(O)C 1-20alkyl, NHC(O)phenyl), Ndialkyl (wherein each alkyl, for example C1-2O, may be the same or different) and 5 or 6 membered rings, optionally containing one or more same or different heteroatoms (e.g. O, N and S). Reference to groups written as "[group] amino" is intended to reflect the nature of the RA and RB groups. For example, "alkylamino" refers to NRARB where one of RA or RB is alkyl. "Dialkylamino" refers to NRARB where RA and RB are each (independently) an alkyl group.
The term "amido" is used here in its broadest sense as understood in the art and includes groups having the formula C(O)NRARB, wherein RA and RB are as defined as above.
Examples of amido include C(O)NH2, C(O)NHalkyl (eg C1-2Oalkyl), C(O)NHaryl (eg
C(O)NHphenyl), C(O)NHaralkyl (eg C(O)NHbenzyl), C(O)NHacyl (eg C(O)NHC(O)C1.
2Oalkyl, C(O)NHC(O)phenyl), C(O)Nalkylalkyl (wherein each alkyl, for example C1-20, may be the same or different) and 5 or 6 membered rings, optionally containing one or more same or different heteroatoms (eg O, N and S).
In a further form of "amido", particularly when R7 is such, -NRARB may be the amino terminus of an amino acid or peptide molecule. Such "amido" groups can be formed by coupling the amino terminus of an amino acid or peptide molecule with a precursor carboxylic acid group in the usual manner. As referred to herein a peptide molecule comprises 2, 3, 4, 5 or more same or different amino acid molecules covalently linked by peptide or amide bonds formed between the amino terminus of one amino acid and the carboxylic acid terminus of another. The amino acids contemplated include natural amino acids and non-natural amino acids, and modified and protected forms thereof. The term "carboxy ester" is used here in its broadest sense as understood in the art and includes groups having the formula CO2R, wherein R may be selected from groups including alkyl, alkenyl, alkynyl, aryl, carbocyclyl, heteroaryl, heterocyclyl, aralkyl, and acyl, each of which may be optionally substituted. Examples of carboxy ester include CQA-aoalkyl, CO2aryl (eg. CO2ρhenyl), CO2aralkyl (eg CO2 benzyl).
"Saccharide" groups contemplated herein include mono-, di- and tri-saccharides of pyranose and furanose groups. In preferred embodiments there are linked through Cl. Examples include, glucose, idose, allose, tallose, mannose, galactose, maltose, lactose, arabinose, glucosamine, ribose, xylose, cellobiose, maltotriose, cellotriose, isomaltotriose. In one embodiment, the saccharides are monosaccharides. In a preferred embodiment, the saccharides are the pyranoses. In a particularly preferred embodiment, the saccharide group is glucose. It will be understood that the saccharide groups can exist in their "free hydroxyl" or "free amino" form or one or more, or all, of the hydroxyl and/or amino groups can be protected by a suitable protecting group (eg acetate groups). A saccharide group may be removed and/or replaced by known methods in the art.
Reference to the following numbering system will be used throughout:
Figure imgf000020_0001
Compounds contemplated by the invention may be obtained by a variety of means including isolation from natural sources, synthetic methods for the preparation of isoquinolines/tetrahydro-β-carbolines and dehydro forms thereof, or by enzyme catalysed coupling. Thus, naturally occurring compounds such as strictosidine (i), strictosidinic acid (ii), 5(S)- 5-carboxystrictosidine (iii) and 3,4-dehydro-5(S)-5-carboxystrictosidine (iv) (see below) can be isolated from their natural sources in accordance with described procedures (see for example, Kitajima, M. et al., 2002; Arbain, D. et al, 1993 and Stδckigt, J. et al, 1977). Once isolated, further chemical manipulations may be performed, e.g. acylation, alkylation, arylation or benzylation of some or all of the free hydroxy or amino groups, amidation or esterification of carboxylic acid groups, de-esterification of ester groups, reduction of carboxylic ester or acid groups to form aldehydes or removal of the glucosyl group and optionally replacing it with another saccharide moiety or other non-H Y group as defined herein. Methods for performing such chemical manipulations or transformations are known in the art and are extensively described, for example, Larock, R.E., 1989.
Figure imgf000021_0001
(i) R = H5 R' = OMe (iv) (ii) R = H, R1 = OH (iii) R = CO2H5 R' = OMe
Alternatively, compounds where - represents a single bond can be prepared by acid- mediated or strictosidine synthase-catalysed coupling of an appropriately substituted/protected tryptamine compound and secologanin, or derivative thereof, as depicted, for example in general Scheme I below.
Figure imgf000022_0001
Scheme I
Compounds of Formula (I), wherein "" is a single bond can be prepared in a stereoselective manner (at C3 of the resulting carbinol) by strictosidane synthase coupling of (1) and (2) in an analogous manner to that described by Patthy-Lukats, A. et ah, 1999 and Treimer, J.F. et ah, 1979, where, in the presence of strictosidine synthase, the coupling of tryptamine and secologanin afforded strictosidine with complete stereoselectivity (Scheme II) :
Figure imgf000022_0002
Scheme II
Alternatively, appropriate tryptamine and secologanin compounds may be coupled under acidic conditions (Pictet Spengler reaction) such as described in US 6,720,331, Patthy- Lukats, A. et ah, 1999 and The Merck Index, Thirteenth Edition. Compounds where ''-"-" is a double bond, referred to herein as 3,4-dehydro compounds, may be prepared by coupling an appropriate tryptamine compound and a modified secologanin compound, wherein the aldehyde has been oxidized to a carboxylic acid group, followed by dehydrating ring closure in accordance with the methods described by US 6,350,757 and Kitajima, M. et al, 2002, and as described below in general Scheme III. Thus, coupling of (1) and (2) followed by ring closure of (3) with a suitable dehydrating agent such as DCC or POCl3, yields the 3,4-dehydro compounds (4).
Figure imgf000023_0001
Alternatively, 3,4-dehydro compounds may be prepared from the 3,4-dihydro compounds by appropriate oxidative treatment, such as treatment with DDQ (see for example US 6,350,757 or US 6,720,331).
Starting tryptamine compounds may be prepared by any one of the many methods known in the art for indole synthesis, or alternatively, may be obtained from commercial sources. Some examples of starting tryptamines contemplated by the present invention include:
Figure imgf000024_0001
Figure imgf000024_0002
Figure imgf000025_0001
Figure imgf000026_0001
Figure imgf000027_0001
Figure imgf000028_0001
Secologanin may be obtained commercially from Sigma (Fluka), USA, or isolated from natural sources (Galan et al, 2006). It may be modified prior to coupling, e.g. removal or replacement of the glucose moiety, protection of free OH groups, oxidation to the carboxylic acid (for synthesis of 3,4-dehydro compounds) or modification of the ester moiety. Alternatively, the vinyl group may be modified in accordance with the olefin cross metathesis methods described by Galan et al., 2006, to afford further secologanin analogues. In some particularly preferred embodiments obtained thereby, Z is selected from (trans)CH=CH-R or (CHa)2R wherein R is Cnoalkyl, for example t-butyl, n-butyl, n- pentyl, and n-heptyl.
In one embodiment of the invention, a preferred compound for coupling to tryptamine compounds is secologanin itself (X = OMe, Y = GIc, Z = CH=CH2). (optionally protected) or the oxidised carboxylic acid form, e.g.
Figure imgf000029_0001
R = H or OH
R1 = H or protecting group
Other preferred secologanin-type compounds for coupling include the aglycone form (or protected form thereof) of those described above.
It will be recognised that in some circumstances it may be appropriate to perform any chemical manipulations on the tryptamine and/or secologanin compounds prior to coupling. In others, for example, where the modification could hinder or block the coupling, the appropriate modification can be performed after coupling.
It will be recognised that during the processes for the preparation of compounds contemplated by the present invention, it may be necessary or desirable to protect certain functional groups which may be reactive or sensitive to the reaction or transformation conditions undertaken (eg OH (including diols), NH2, CO2H, SH, C=O). Suitable protecting groups for such functional groups are known in the art and may be used in accordance with standard practice. As used herein, the term "protecting group", refers to an introduced functionality which temporarily renders a particular functional group inactive under certain conditions. Such protecting groups and methods for their installation and subsequent removal at an appropriate stage are described in Protective Groups in Organic Chemistry, 3rd Edition, T.W.Greene and P. G. Wutz, John Wiley and Sons, 1999, the entire contents of which are incorporated herein by reference. Exemplary forms of protected groups include: for amino (NH2) - carbamates (such as Cbz, Boc, Fmoc), benzylamines, acetamides (e.g. acetamide, trifluoroacetamide); for carbonyl - acetals, ketals, dioxanes, dithianes, and hydrazones; for hydroxy - ethers (e.g. alkyl ethers, alkoxylalkyl ethers, allyl ethers, silyl ethers, benzyl ethers, tetrahydropyranyl ethers), carboxylic acid esters, acetals (e.g. acetonide and benzylidene acetal); for thio (SH) -ethers (e.g. alkyl ethers, benzyl ethers), esters for CO2H - esters (e.g. alkyl esters, benzyl esters).
It will also be recognised that certain compounds of formula (I) may possess asymmetric centres and are therefore capable of existing in more than one stereoisomeric form, such as enantiomers and diastereomers. The invention thus also relates to compounds in substantially pure (e.g. about 95%, preferably at least 97%, or more preferably at least
99%) isomeric form at one or more asymmetric centres, as well as stereoisomeric mixtures.
Such isomers may be prepared by asymmetric synthesis, for example using chiral intermediates or reagents, enzymes, or mixtures may be resolved by conventional methods, eg., chromatography, recrystallisation or use of a resolving agent. In some embodiments of the invention, the compounds of Formula (I) have the same stereochemistry at 1, 2, 3 or all 4 of the chiral centres depicted for strictosidinic acid (i), or I5 2, 3 or all 4 chiral centres as depicted for 3,4-dehydro 5(S)-5-carboxystrictosidinic acid (iv).
In some embodiments of the invention, R1-4 are independently selected from hydrogen, alkyl, (e.g. C^alkyl such as methyl, ethyl, propyl, butyl, cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl), hydroxyalkyl (e.g. hydroxymethyl, hydroxyethyl, hydroxypropyl), alkoxyalkyl (e.g. methoxymethyl, methoxyethyl, methoxypropyl, ethoxymethyl, ethoxyethyl, ethoxypropyl etc) alkoxy (e.g. C1-6 alkoxy such as methoxy, ethoxy, propoxy, butoxy, cyclopropoxy, cyclobutoxy), alkylthio (e.g. MeS-, EtS-, PrS-, BuS-), halo, trifluoromethyl, trichloromethyl, tribromomethyl, hydroxy, phenyl (which itself may be further substituted e.g., by Ci-6alkyl, halo, hydroxy, hydroxyC1-6alkyl, C1. 6alkoxy, haloC1-6alkyl, cyano, nitro OC(O)C 1-6alkyl, and amino), benzyl (wherein benzyl itself may be further substituted e.g., by one or more of Chalky!, halo, hydroxy, hydroxyCi.δalkyl, Cμβalkoxy, haloCi.6alkyl, cyano, nitro, OC(O)C1-6alkyl, and amino), phenoxy (wherein phenyl itself may be further substituted e.g., by one or more of C1. 6alkyl, halo, hydroxy,
Figure imgf000031_0001
cyano, nitro, OC(O)C1. 6alkyl, and amino), benzyloxy (wherein benzyl itself may be further substituted e.g., by one or more of Ci-6alkyl, halo, hydroxy,
Figure imgf000031_0002
C1-6alkoxy, haloC1-6alkyl, cyano, nitro, OC(O)C1.6alkyl, and amino), amino, alkylamino (e.g. C1-6alkyl, such as methylamino, ethylamino, propylamino etc), dialkylamino (e.g. C1-6alkyl, such as dimethylamino, diethylamino, dipropylamino), acylamino (e.g. NHC(O)CHs), phenylamino (wherein phenyl itself may be further substituted e.g., by one or more Of C1. 6alkyl, halo, hydroxy hydroxyC^ealkyl, Ci-6alkoxy, haloCi-βalkyl, cyano, nitro, OC(O)C1. 6alkyl, and amino), nitro, formyl, -C(O)-alkyl (e.g. C1-6 alkyl, such as acetyl), 0-C(O)- alkyl (e.g. C^alkyl, such as acetyloxy), benzoyl (wherein benzyl itself may be further substituted e.g., by one or more of C^ancyl, halo, hydroxy hydroxyC1-6alkyl, C1-6alkoxy, halod.6alkyl, cyano, nitro, OC(O)C μβalkyl, and amino), benzoyloxy (wherein benzyl itself may be further substituted e.g., by one or more of C1-6alkyl, halo, hydroxy hydroxyCi- 6alkyl, C1-6alkoxy, haloC1-6alkyl, cyano, nitro, 0C(0)d-6alkyl, and amino), CO2H, Cθ2alkyl (e.g. C1-6alkyl such as methyl ester, ethyl ester, propyl ester, butyl ester), Cθ2phenyl (wherein phenyl itself may be further substituted e.g., by one or more of Ci- 6alkyl, halo, hydroxy, hydroxyCt-δalkyl, C1-6alkoxy, haloCi-6alkyl, cyano, nitro, OC(O)C1. 6alkyl, and amino), COibenzyl (wherein benzyl itself may be further substituted e.g., by one or more of C^alkyl, halo, hydroxy, hydroxyC1-6alkyl, C1-6alkoxy, haloC1-6alkyl, cyano, nitro, OC(O)C 1-6alkyl, and amino), CONH2, CONHphenyl (wherein phenyl itself may be further substituted e.g., by one or more of C1-6alkyl, halo, hydroxy, hydroxyCϊ. βalkyl, C1-6alkoxy, haloC1-6alkyl, cyano, nitro, OC(O)C1-6alkyl, and amino), CONHbenzyl (wherein benzyl itself may be further substituted e.g., by one or more of Ci-βalkyl, halo, hydroxy hydroxyC1-6alkyl,
Figure imgf000032_0001
haloCi-6alkyl, cyano, nitro, OC(O)C 1-6alkyl, and amino), CONHalkyl (e.g. C1-6 alkyl such as methyl ester, ethyl ester, propyl ester, butyl amide) CONdialkyl (e.g. C1-6alkyl) aminoalkyl (e.g., HNC1-6alkyl-, C1-6alkylHN-C1-6alkyl- and (C1-6alkyl)2N-C1-6alkyl-), thioalkyl (e.g., HSC1-6alkyl-), carboxyalkyl (e.g., HO2CCi- βalkyl-), carboxyesteralkyl (e.g., C1.6alkyl02CC1-6alkyl-), amidoalkyl (e.g., H2N(O)CC1- ealkyl-, H(C1-6alkyl)N(O)CC1.όalkyl-), formylalkyl (e.g., OHCC^alkyl-), acylalkyl (e.g.,
Figure imgf000032_0002
nitroalkyl (e.g., O2NC1-6alkyl-), sulfoxidealkyl (Cg15 R(O)SC1- ealkyl, such as C1.6alkyl(O)SC1.6alkyl-), sulfonylalkyl (e.g., R(O)2SC i-6alkyl- such as C1- 6alkyl(O)2SC1-6alkyl-), sulfonamidoalkyl (e.g., 2HN(O)2SC1-6alkyl, H(C1.6alkyl)N(O)2SCi- 6alkyl-), and where 2 adjacent carbon atoms are substituted by one end each of a -O- (CH2)n-0- or -NH-(CH2)n-NH- group, wherein n is 1 or 2.
In some embodiments of the invention, at least one of R1-R4 is hydrogen, for example, at least 2 are hydrogen. In other embodiments, 3 or all 4 OfR1-R4 are hydrogen.
In some embodiments of the invention, R5 and R6 are selected from hydrogen, optionally substituted C1-6alkyl, optionally substituted C(O)C ^alkyl, optionally substituted C(O)phenyl, optionally substituted C(0)beri2yl.
In some embodiments of the invention, R7 is selected from hydrogen, CO2H, CO2C1-6alkyl5 e.g. methyl, ethyl, propyl or butyl), CONH2, CONHC1-6alkyl, CONHaryl. In particularly preferred embodiments, R7 is selected from H and CO2H.
In some embodiments of the invention, Rg is hydrogen.
In some embodiments of the invention, at least one of R7a and Rga is hydrogen, for example, both are hydrogen. In some embodiments of the invention, X include OH, OCi.ioalkyl, NHC^oalkyl, NHphenyl and NHbenzyl.
In some embodiments of the invention, Y is H or a saccharide, such as glucose.
In some embodiments of the invention, Z is CH2=CH2 or C^oalkyl.
Some non-limiting examples of compounds contemplated by the invention include combinations of any two, three, four, five, six, seven or eight of the embodiments for R1-4, R5 and R6, R7, R8, R7a and Rg8, X, Y and Z described above.
In a particular embodiment, the compound for use in the invention is strictosidinic acid.
Strictosidinic acid can be isolated from natural sources, in accordance with the procedures described in the literature, for example, described by Arbain et ah, 1993; Yamakazi et ah, 2003 and Geerlings et al. , 2000, and in accordance with the Examples described herein. Thus, in certain embodiments, strictosidinic acid, or other compounds contemplated herein are used in extracted, isolated or substantially purified form (for example, at least 50, 75, 90, 95 or 99% pure).
As used herein, "regulating glucose homeostasis" refers to the regulation or control of blood glucose levels to lower hyperglycaemic, or preferably achieve or maintain normal fasting state, blood glucose levels. Normal fasting state blood glucose levels are typically less than 6.1 mmolL" (110 mgdL"1). Hyperglycaemic levels refer to blood glucose levels greater than or equal to 6.1 mmolL'1 (110 mgdL"1).
Impaired fasting glycemia (IFG) is characterised by a fasting plasma glucose concentration greater than or equal to 6.1 mmol (110 mgdL"1) but less than 7.0 mmolL"1 (126 mgdL"1) and a 2-h plasma glucose concentration during the oral glucose tolerance test (OGTT) (if measured) less than 7.8 mmolL"1 (140 mgdL"1). Impaired glucose tolerance (IGT) is characterised by a fasting plasma glucose concentration of less than 7.0 mmolL"1 (126 mgdL"1) and a 2-h plasma glucose concentration during the OGTT of greater than or equal to 7.8 mmolL'1 (140 mgdL"1) but less than 11.1 mmolL"1 (200 mgdL"1). Diabetes is characterised by a fasting plasma glucose concentration of greater than or equal to 7.0 mmolL"1 (126 mgdL'1) or a 2-h plasma glucose concentration during the OGTT of greater than 11.1 mmolL'1 (200 mgdL"1).
The compounds of the invention may have utility in the therapeutic or prophylactic treatment of diseases and conditions, and /or their symptoms, in which insulin resistance in involved or implicated in a subject. Any disease or condition, or symptom thereof in which insulin resistance or impaired glucose uptake by a cell or tissue can be attributed, or play a role or is manifested is contemplated herein. Non-limiting examples include NIDDM, gestational diabetes, impaired glucose tolerance, impaired fasting glucose, Syndrome X, hyperglycemia, obesity, cardiovasular disease, atheriosclerosis, hypertriglyceridemia, dyslipidemia, hyperinsulinemia, nephropathy, neuropathy, retinopathy, ischemia, stroke and fatty liver disease.
Subjects to be treated include mammalian subjects: humans, primates, livestock animals (including cows, horses, sheep, pigs and goats), companion animals (including dogs, cats, rabbits, guinea pigs), and captive wild animals. Laboratory animals such as rabbits, mice, rats, guinea pigs and hamsters are also contemplated as they may provide a convenient test system. Non-mammalian species such as birds, amphibians and fish may also be contemplated in certain embodiments of the invention. A particularly contemplated subject is a human subject.
The compounds of the invention are administered in an amount which, when administered according to the desired dosing regimen, at least partially attains the desired therapeutic effect, e.g. modulating glucose homeostasis, modulating cellular glucose uptake, treating insulin resistance, lowering blood glucose levels or treating a disease or condition, or symptom thereof, in which insulin resistance is involved. As used herein, treatment refers to therapeutic ameliorating or prophylactic treatment, including one or more of: alleviating or ameliorating the symptoms of, preventing or delaying the onset of, inhibiting the progression of, or halting or reversing altogether the onset or progression of the particular disorder or condition, or one or more symptoms thereof, being treated.
Suitable dosage amounts and dosing regimens can be determined by the attending physician and may depend on the particular condition being treated, the severity of the condition as well as the general age, health and weight of the subject, and may be in the range of from about 0.1 mg to about 1000 mg of active per day. The active ingredient may be administered in a single dose or a series of doses. Suitable dosages may contain about 1, 2.5, 5, 10, 20, 25, 50, 75, 100, 150, 200, 250 or 500 mg of active.
While it is possible for the active ingredient to be administered alone, it is preferable to present it as a composition, preferably as a pharmaceutical composition, with one or more pharmaceutically acceptable excipients or additives. Thus, the present invention also relates to the use of a compound of formula (I) or a pharmaceutically acceptable salt or prodrug thereof in the manufacture of a medicament for modulating glucose homeostasis, modulating cellular glucose uptake, treating insulin resistance, or lowering blood glucose levels.
The formulation of such compositions is well known to those skilled in the art, see for example, Remington's Pharmaceutical Sciences, 18th Edition, Mack Publishing, 1990. The composition may contain any suitable carriers, diluents excipients or other additives.
These include all conventional solvents, dispersion media, fillers, solid carriers, coatings, antifungal and antibacterial agents, dermal penetration agents, surfactants, isotonic and absorption agents and the like. It will be understood that the compositions of the invention may also include other supplementary physiologically active agents.
The carrier must be pharmaceutically acceptable in the sense of being compatible with the other ingredients of the composition and not injurious to the subject. Compositions include those suitable for oral, rectal, nasal, topical (including dermal, buccal and sublingual), vaginal or parental (including subcutaneous, intramuscular, intravenous and intradermal) administration. The compositions may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy. Such methods include the step of bringing into association the active ingredient with the carrier which constitutes one or more accessory ingredients. In general, the compositions are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both, and then if necessary shaping the product.
Compositions of the present invention suitable for oral administration may be presented as discrete units such as capsules, sachets or tablets each containing a predetermined amount of the active ingredient; as a powder or granules; as a solution or a suspension in an aqueous or non-aqueous liquid; or as an oil-in-water liquid emulsion or a water-in-oil liquid emulsion.
A tablet may be made by compression or moulding, optionally with one or more accessory ingredients. Compressed tablets may be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as a powder or granules, optionally mixed with a binder (e.g inert diluent), preservative disintegrant (e.g. sodium starch glycolate, cross-linked polyvinyl pyrrolidone, cross-linked sodium carboxymethyl cellulose) surface- active or dispersing agent. Moulded tablets may be made by moulding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent. The tablets may optionally be coated or scored and may be formulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydroxypropylmethyl cellulose in varying proportions to provide the desired release profile. Tablets may optionally be provided with an enteric coating, to provide release in parts of the gut other than the stomach.
Compositions suitable for topical administration in the mouth include lozenges comprising the active ingredient in a flavoured base, usually sucrose and acacia or tragacanth gum; pastilles comprising the active ingredient in an inert basis such as gelatin and glycerin, or sucrose and acacia gum; and mouthwashes comprising the active ingredient in a suitable liquid carrier. Compositions suitable for topical administration to the skin may comprise the compounds dissolved or suspended in any suitable carrier or base and may be in the form of lotions, gel, creams, pastes, ointments and the like. Suitable carriers include mineral oil, propylene glycol, polyoxyethylene, polyoxypropylene, emulsifying wax, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water. Transdermal patches may also be used to administer the compounds of the invention.
Compositions for rectal administration may be presented as a suppository with a suitable base comprising, for example, cocoa butter, glycerin, gelatin or polyethylene glycol.
Compositions suitable for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams or spray formulations containing in addition to the active ingredient such carriers as are known in the art to be appropriate.
Compositions suitable for parenteral administration include aqueous and non-aqueous isotonic sterile injection solutions which may contain anti-oxidants, buffers, bactericides and solutes which render the composition isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents. The compositions may be presented in unit-dose or multi-dose sealed containers, for example, ampoules and vials, and may be stored in a freeze-dried (lyophilised) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described.
Preferred unit dosage compositions are those containing a daily dose or unit, daily sub- dose, as herein above described, or an appropriate fraction thereof, of the active ingredient. It should be understood that in addition to the active ingredients particularly mentioned above, the compositions of this invention may include other agents conventional in the art having regard to the type of composition in question, for example, those suitable for oral administration may include such further agents as binders, sweeteners, thickeners, flavouring agents disintegrating agents, coating agents, preservatives, lubricants and/or time delay agents. Suitable sweeteners include sucrose, lactose, glucose, aspartame or saccharine. Suitable disintegrating agents include corn starch, methylcellulose, polyvinylpyrrolidone, xanthan gum, bentonite, alginic acid or agar. Suitable flavouring agents include peppermint oil, oil of wintergreen, cherry, orange or raspberry flavouring. Suitable coating agents include polymers or copolymers of acrylic acid and/or methacrylic acid and/or their esters, waxes, fatty alcohols, zein, shellac or gluten. Suitable preservatives include sodium benzoate, vitamin E, alpha-tocopherol, ascorbic acid, methyl paraben, propyl paraben or sodium bisulphite. Suitable lubricants include magnesium stearate, stearic acid, sodium oleate, sodium chloride or talc. Suitable time delay agents include glyceryl monostearate or glyceryl distearate.
The present invention also relates to prodrugs of formula (I). Any compound that is a prodrug of a compound of formula (I) is within the scope and spirit of the invention. The term "prodrug" is used in its broadest sense and encompasses those derivatives that are converted in vivo, either enzymatically or hydrolytically, to the compounds of the invention. Such derivatives would readily occur to those skilled in the art, and include, for example, compounds where a free thiol or hydroxy group is converted into an ester, such as an acetate, or thioester or where a free amino group is converted into an amide. Procedures for acylating the compounds of the invention, for example to prepare ester and amide prodrugs, are well known in the art and may include treatment of the compound with an appropriate carboxylic acid, anhydride or chloride in the presence of a suitable catalyst or base. Esters of carboxylic acid (carboxy) groups are also contemplated. Suitable esters C1-6alkyl esters; Cμδalkoxymethyl esters, for example methoxymethyl or ethoxymethyl; Ci-6alkanoyloxymethyl esters, for example, pivaloyloxymethyl; phthalidyl esters; C3.8cycloalkoxycarbonylC1-6alkyl esters, for example, 1- cyclohexylcarbonyloxyethyl; l,3-dioxolen-2-onylmethyl esters, for example, 5-methyl-l,3- dioxolen-2-onylmethyl; and Ci-βalkoxycarbonyloxyethyl esters, for example, 1- methoxycarbonyloxyethyl. Prodrugs of amino functional groups include amides (see, for example, Adv. BioSci., 1979, 20, 369, Kyncl, J. et al), enamines (see, for example, J. Pharm. Sci., 1971, 60, 1810, Caldwell, H. et al), Schiff bases (see, for example, US Patent No 2,923,661 and Antimicrob. Agents Chemother., 1981, 19, 1004, Smyth, R. et al), oxazolidines (see, for example, J. Pharm. Sci, 1983, 72, 1294, Johansen, M. et al), Mannich bases (see, for example, J. Pharm. Sci. 1980, 69, 44, Bundgaard, H. et al and J. Am. Chem. Soc, 1959, 81, 1198, Gottstein, W. et al), hydroxymethyl derivatives (see, for example, J. Pharm. Sci, 1981, 70, 855, Bansal, P. et al) and N-(acyloxy)alkyl derivatives and carbamates (see, for example, J. Med. Chem., 1980, 23, 469, Bodor, N. et al, J. Med. Chem., 1984, 27, 1037, Firestone, R. et al, J. Med. Chem., 1967, 10, 960, Kreiger, M. et al, US Patent No 5,684,018 and J. Med. Chem., 1988, 31, 318-322, Alexander, J. et al). Other conventional procedures for the selection and preparation of suitable prodrugs are known in the art and are described, for example, in WO 00/23419; Design of Prodrugs, H. Bundgaard, Ed., Elsevier Science Publishers, 1985; Methods in Enzymology, 42: 309-396, K. Widder, Ed, Academic Press, 1985; A Textbook of Drug Design and Development, Krogsgaard-Larsen and H. Bundgaard, Eds, Chapter 5, pi 13-191 (1991); Advanced Drug Delivery Reviews, 8; 1-38 (1992); Journal of Pharmaceutical Sciences, 77;285 (1988), H. Bundgaard, et al; Chem Pharm Bull, 32692 (1984), N. Kakeya et al and The Organic Chemistry of Drug Desig and Drug Action, Chapter 8, pp352-401, Academic press, Inc., 1992.
Suitable pharmaceutically acceptable salts of compounds of formula (I) include, but are not limited to salts of pharmaceutically acceptable inorganic acids such as hydrochloric, sulphuric, phosphoric nitric, carbonic, boric, sulfamic, and hydrobromic acids, or salts of pharmaceutically acceptable organic acids such as acetic, propionic, butyric, tartaric, maleic, hydroxymaleic, fumarie, maleic, citric, lactic, mucic, gluconic, benzoic, succinic, oxalic, phenylacetic, methanesulphonic, toluenesulphonic, benezenesulphonic, salicyclic sulphanilic, aspartic, glutamic, edetic, stearic, palmitic, oleic, lauric, pantothenic, tannic, ascorbic and valeric acids. Base salts include, but are not limited to, those formed with pharmaceutically acceptable cations, such as sodium, potassium, lithium, calcium, magnesium, ammonium and alkylammonium. Basic nitrogen-containing groups may be quarternised with such agents as lower alkyl halide, such as methyl, ethyl, propyl, and butyl chlorides, bromides and iodides; dialkyl sulfates like dimethyl and diethyl sulfate; and others.
The compounds of the invention may be in crystalline form either as the free compounds or as solvates and it is intended that both forms are within the scope of the present invention. The term "solvate" refers to a complex or aggregate formed by one or more molecules of a solute, ie compounds contemplated by the invention, and one or more molecules of a solvent. Suitable solvents are well understood in the art and include for example, of water, ie to form hydrates, and common organic solvents such as alcohols (methanol, ethanol, isopropanol) and acetic acid. Methods of solvation are generally known within the art, for example, recrystallisation from an appropriate solvent.
The methods, compounds and compositions of the invention may be utilised in conjunction with other anti-diabetic or anti-hyperglycaemic agents and may be administered, simultaneously (separately or as a combination) or sequentially in accordance with a suitable dosing and treatment regime as determined by the attending physician. Suitable agents may include insulin sensitisers, glucose resorption/uptake inhibitors and the classes and compounds identified in US 2005/0037981 , particularly Table 2, the contents of which are incorporated herein in their entirety.
The compounds of the invention may also be presented for use in veterinary compositions. These may be prepared by any suitable means known in the art. Examples of such compositions include those adapted for:
(a) oral administration, external application (eg drenches including aqueous and nonaqueous solutions or suspensions), tablets, boluses, powders, granules, pellets for admixture with feedstuffs, pastes for application to the tongue;
(b) parenteral administration, eg subcutaneous, intramuscular or intravenous injection as a sterile solution or suspension;
(c) topical application eg creams, ointments, gels, lotions etc. The invention will now be described with reference to the following examples which are provided for the purpose of illustrating certain embodiments of the invention and are not to be construed as limiting the generality hereinbefore described.
EXAMPLES
Example 1: Effect of Plant Extract
Plant Extract Preparation
The bark and leaves of Alphitonia zizqphoides, Morinda citrifolia, Neisoperma oppositifolium, Pometia pinnata, Rhus taitensis, Syzygium malaccense, Terminalia catappa and Thespia populnea were dried and ground to form the plant material for subsequent extraction.
The plant extract used for both in vivo and in vitro studies was prepared as follows: lOOg of ground, dried plant material above was mixed thoroughly with IL of distilled water and maintained at 70°C for 24h. The mixture was then filtered and allowed to cool to room temperature. For all experiments the plant extract was prepared that day, and any unused plant extract was discarded at the end of the day.
In Vivo Studies in Psammomys obesus
The effects of the plant extract were tested in both lean, non-diabetic and obese, type 2 diabetic P. obesus. Animals from within each group (n=l 0 each) were randomly assigned to plant extract treatment or control groups. All animals used in these studies were male and 18 weeks of age. The characteristics of the animals are given in Table 1.
Table 1: Characteristics of P. obesus
Figure imgf000043_0001
For the plant extract treatment group, 500ml of plant extract was mixed thoroughly with 1.5L dH2O and 5kg ground rodent chow. The chow, now containing 1% (w/w) plant extract, was re-pelleted and dried. After a 7-day baseline period, animals in the plant extract treatment group received ad libitum access to the chow containing 1% plant extract for 11 days, while control animals received ad libitum access to chow re-pelleted and dried as above after being mixed with άϊlzO only.
Body weight and food intake were measured daily, while blood was collected from the tail vein for the measurement of glucose and insulin at the start and end of the 11 day experimental period. Treatment with the plant extract resulted in a significant reduction in blood glucose concentration in both lean, non-diabetic (by 10.5%; p=0.001) and obese, diabetic P. obesus (by 33.7%; p=0.024; Table 2). Treatment with the plant extract tended to reduce plasma insulin concentration in the plant extract treated groups (by 22% in the lean, non-diabetic and by 31% in the obese, diabetic animals; Table 2), however these changes were not statistically significant. Treatment with the plant extract for 11 days had a significant effect on the change in body weight throughout the study in both lean, non- diabetic and obese, diabetic P. obesus (Table 2). In the lean, non-diabetic animals, the plant extract treated group lost 1.6 + 2.8 g during the study period, while the control animals gained 7.4 ± 2.2 g (p=0.035). In obese, diabetic P. obesus, the plant extract treated group lost 9.4 + 4.1 g during the study period, while the control animals gained 5.6 + 3.1 g (p=0.019). The effects of the plant extract on body weight appeared to be independent of effects on food intake, as there was no detectable difference between food intake during the baseline and experimental periods.
Table 2: Effects of plant extract on blood glucose and plasma insulin
Figure imgf000044_0001
In summary, treatment with the plant extract for 11 days reduced blood glucose concentration in both lean, non-diabetic and obese, diabetic P. obesus. Three of the 5 obese, diabetic animals treated with the plant extract became non-diabetic within the 11 day study period, while in the other two animals the severity of hyperglycemia was substantially reduced. In addition, there was a tendency for reductions in plasma insulin concentrations in the plant extract treated animals, suggesting improvement in insulin action after treatment. The plant extract treated animals also exhibited reduced body weight gain during the study period relative to control animals, which may have contributed to their improved insulin sensitivity. Interestingly, this altered body weight gain was independent of food intake, which was not affected by the treatment (which also prevents the possibility of conditioned taste aversion). It is therefore assumed that the plant extract may have affected energy expenditure and/or substrate utilisation in P. obesus.
Overall these data suggest that the plant extract used in these studies may contain an insulin sensitising agent, and has potential therefore as an antidiabetic treatment. In Vitro Studies in Cultured 3T3-L1 Adipocytes
Glucose Uptake
3T3-L1 adipocytes were differentiated and cultured in DMEM containing 5% fetal calf serum using standard procedures. For the glucose uptake assay, the cells were washed with Krebs Ringer Buffer Solution for 2h, and then insulin stimulated (0-10OnM) for 30min before addition of the plant extract at concentrations of 0.005-0.5% (v/v) and 14C-2- deoxyglucose. After 15min the reaction was stopped with ice-cold PBS containing phloretin and SDS, and radioactivity counted immediately using a beta counter.
The effects of the plant extract at concentrations of 0.005, 0.05, 0.125, 0.25 and 0.5% were tested at various concentrations of insulin (0, 0.1, 0.5, 1, 5, 10 and 10OnM). The glucose uptake in plant extract treated cells was calculated relative to the corresponding dose of insulin without plant extract, and the results presented as percentages of the effects of insulin alone. Table 3 shows the results obtained at the various insulin concentrations. At each insulin concentration, the ability of insulin to stimulate glucose uptake was tested (data not shown) and results only accepted if insulin stimulation of glucose uptake was apparent.
The plant extract clearly stimulated glucose uptake in these cells at insulin concentrations of 0-5nM (Table 3). The rate of glucose uptake was increased by 3-fold at the highest plant extract concentration (0.5%) when no insulin was present (p<0.001), and by 2-fold in the presence of InM insulin (p=0.009). At insulin concentrations of 10 and 10OnM the plant extract had no discernible effect on glucose uptake in these cells, which was likely to be maximally stimulated at these concentrations of insulin. Table 3: Effects of plant extract on glucose uptake in 3T3-L1 cells (mean ± SEM, n=4-8)
Figure imgf000046_0001
*p<0.05, p<0.01, #ρ<0.001 compared with 0 plant extract.
Lipogenesis
A similar protocol was used to measure incorporation of radioactive glucose into lipid in differentiated 3T3-L1 adipocytes, a measure of lipogenesis. For this assay, the cells were serum-starved overnight, then incubated in DPBS containing insulin (0-100OnM), plant extract at concentrations of 0.005-0.5% (v/v) and wC-2-deoxyglucose. After 3h the reaction was stopped with ice-cold PBS and the lipid fraction extracted by addition of isopropanol:heptane:H2SO4 (4:1 : ;0.1) for 20 min followed by centrifugation and removal of the upper phase, in which radioactivity was counted immediately using a beta counter.
The plant extract stimulated lipogenesis in these cells at insulin concentrations of 0-1OnM (Table 4). The rate of lipogenesis was increased by more than 2-fold at the highest plant extract concentration (0.5%) when no insulin was present (p<0.001), by 1.5-fold in the presence of InM insulin (p<0.001), and by 1.2-fold at an insulin concentration of 1OnM (p=0.036). At higher insulin concentrations the plant extract had no discernible effect on lipogenesis in these cells, which was likely to be maximally stimulated at these concentrations of insulin.
Figure imgf000047_0001
*p<0.05, Λp<0.01, #p<0.001 compared to 0 plant extract.
To summarise the initial in vitro data, the plant extract stimulated lipogenesis and glucose uptake in differentiated 3T3-L1 adipocytes in the presence of 0-1OnM insulin. These data support the contention that the plant extract used in these studies may contain an insulin- sensitising agent, and suggest that this agent can directly affect glucose metabolism in adipocytes.
Example 2: Fractionation, Identification and In Vitro Effect of Strictosidinic Acid
Fractionation of plant extract.
The fteeze-dried plant extract (3g) was fractionated by HPLC/ESI-MS-ELSD under the conditions described in Table 5. Table 5: Conditions for preparative chromatographic fractionation.
Figure imgf000048_0001
Preparative chromatographic (HPLC) fractionation resulted in 42 fractions of the plant extract. Each was collected in a 4-ml brown glass vial, and the water and buffer were removed online via solid phase extraction. The mass of material in each fraction ranged from 0.23 to 126.32 mg, and the total combined weight of all 42 fractions was approximately 1.7 g (accounting for ~ 56% of the 3 g used).
Table 6: Weights of fractions collected after preparative chromatographic (HPLC) fractionation
Fraction Weight (mg) Fraction Weight (mg)
FOO 126.32 F21 0.83
FOl 0.57 F22 0.59
F02 0.74 F23 1.05
F03 1.58 F24 0.59
F04 2.09 F25 0.36
F05 5.21 F26 1.42
F06 7.33 F27 0.61
F07 2.24 F28 0.38
F08 1.01 F29 0.29
F09 1.1 F30 0.23
FlO 1.09 F31 0.26
FI l Ll F32 0.24
F12 0.7 F33 0.31
F13 1.26 F34 0.41
F14 1.62 F35 0.42
F15 1.31 F36 0.45
F16 1.12 F37 0.8
F17 1.28 F38 0.4
F18 1.45 F39 0.3
F19 0.94 F40 0.25
F20 0.83 F41 1.82
Screening of fractions 1) Lipogenesis bioassay (3T3-L1 adipocytes)
The plant extract fractions were examined in the lipogenesis (glucose incorporation into lipid) bioassay, measuring the amount of glucose converted into triglyceride using radioactive MC-glucose. Briefly, 3T3-L1 adipocytes were incubated in starvation media overnight followed by 30 minute incubation in DPBS buffer. Next, the DPBS buffer was removed, and treatment (freeze-dried plant extract, or its reconstituted fractions) was added to each well for 90 minutes in the presence of 1 nM insulin. Then, treatments were removed, and the cells were washed twice before being lysed with a lipid-extracting reagent. The lipid extract was then transferred to 2 ml-tube containing water and heptane. Thereafter, the mixture was vigorously vortexed, and ceiitrifuged to separate the lipid and water fractions. The lipid fraction (heptane soluble layer, upper phase) was added to 4 ml scintillation fluid to count the radioactivity using the β-counter Tri-carb® 2900TR Liquid Scintillation Analyser (Packard; Packard Bioscience B.V., Groningen, Netherlands).
In each experiment, 2 plant extract fractions were tested at 0.05, 0.5, and 1% (w/v) in triplicate, together with insulin and the IMO 14 extract as positive control. To obtain uniformity and ease of comparison, results (amount of glucose incorporated into lipid) were converted to percent change relative to control (1 nM insulin).
Fraction 18 in particular had an effect on the incorporation of glucose into lipid in 3T3-L1 cells.
Effect of Fraction 18
1. Glucose Uptake in 3T3 -Ll adipocytes Cell culture
3T3-L1 fibroblasts are grown in 10cm dishes and cultured in DMEM (4.5 g/L glucose) supplemented with 10% (v/v) heat-inactivated fetal bovine serum (FBS), 50 IU/ml pencillin and 50 μg/ml streptomycin. Cells for differentiation into adipocytes were maintained at post-confluence for 2 days, and then induced to differentiate by the addition of DMEM containing 10% FBS, 2 μg/ml insulin, 0.25 μM dexamethasone and 0.5 niM 3-isobutyl-l- methylxanthine. After 3 days, the induction medium was replaced with fresh DMEM containing 10% FBS and 2 μg/ml insulin for another 3 days. Adipocytes were maintained in DMEM with 5% FBS for 2 days thereafter. Adipocytes were used 8-12 days after initiation of differentiation, after which time greater than 80% of fibroblasts had differentiated into mature adipocytes. Treatment with Fraction 18
3T3-L1 adipocytes were re-seeded into gelatin-coated clear- wall clear-bottom 96-well plates (2 x 10cm dishes : 1 x 96-well plate) and maintained in DMEM with 5% FBS. After 24 hrs, adipocytes were treated with 0.2%, 0.5%, 1% or 2% (w/v) Fraction 18 in DMEM (4.5 g/L glucose) containing 0.2% BSA for 16 to 18 hr (overnight). Each dose of SA was added to 4 wells and the following day, [3H] -2-deoxy glucose uptake was then measured (n=4). Fraction 18 stock solution was prepared as either 10% (w/v) (54 μg/ml) or 50% (w/v) (270 μg/ml) in milliQH20 and stored at 4°C.
Glucose Uptake
Cells were washed twice in Dulbecco's PBS, pH 7.4 (Gibco) containing 0.2% (w/v) RIA- grade BSA, 0.5 rnM MgCl2 and 0.5 mM CaCl2. Insulin at 0, 0.1, 1 and 10 nM is added for 30 min at 37°C. Uptake of 50 μM 2-deoxy glucose and 0.5 μCi 2-deoxy- [U-3H] glucose (NEN, PerkinElmer Life Sciences) per well was measured over the final 10 min of insulin stimulation (0, 0.5 and 1OnM). The reaction was stopped upon addition of ice-cold 80 μg/ml phloretin in PBS, pH 7.4 and cells solublised in 0.03% (w/v) SDS. Counts per minute (cpm) were measured by scintillator counter. Each measurement was performed in quadruplet in 1 assay, and each assay was repeated independently 3 times.
Following the screening, glucose uptake assays were utilised in 3T3-L1 adipocytes to confirm the activity of the selected fractions. As shown in Table 7 below, Fraction 18 caused a dose-dependent increase in glucose uptake in 3T3-L1 cells. At a dose of 0.5% (w/v) Fraction 18 caused an increase in glucose uptake of 17%. When the dose of Fraction 18 was increased to 1% (w/v), the increase in glucose uptake was 53%. These data strongly support the proposition that Fraction 18 increased the uptake of glucose into differentiated 3T3-L1 adipocytes. Table 7: Effects of Fraction 18 on glucose uptake in 3T3-L1 adipocytes.
Treatment Dose Glucose uptake % increase (pmol/min/well)
Insulin O nM 360 ± 44 0
I nM 1201 ± 131 234
1O nM 1673 ± 87 364
Fraction 18 0.5% (w/v) 420 ± 10 17
1.0% (w/v) 550 ± 99 53
Gene expression signature analysis
In addition to the bioassays above, gene expression signatures were used as an endpoint for secondary in vitro screening studies. Specifically, microarray technology was used to characterise the effects of the plant extract fractions on gene expression in 3T3-L1 adipocytes.
Hybidisation involved indirect labelling of target cDNA and reference cDNA with fluorescent dyes Cy 5 and Cy3 respectively (Amersham Pharmacia Biotech). Total RNA was extracted using the TRIzol method. The RNA was purified using RNeasy midi columns (Qiagen). 20μg of RNA per sample was processed using the Superscript Indirect cDNA labelling system (Invitrogen) as per manufacturer's instructions. The cDNA was hybridised in a 40μl reaction containing combined target and reference fluorescently labelled cDNAs, 5X SSC (Invitrogen), 16μg PolydA (Amersham), 5X Denhardt's solution (Invitrogen), 4.8μg yeast tRNA (SIGMA) and 0.01% Sodium dodecyl sulfate (Invitrogen). The reaction was heated to 980C for 2 min and maintained at 6O0C until used. Microarrays were placed into a hybridisation chamber containing lOOμl of 2% SSC. A coverslip was placed on top of the microarray slide and the whole reaction mixture was loaded. Hybridisation chambers were secured and placed in a waterbath within a hybridisation oven at 420C for 16 hr. Following hybridisation, the array slides were washed with gentle agitation to ensure all unbound cDNA was removed. The washing procedure included gentle agitation for 2 minutes in solution 1 (0.5X SSC, 0.1% SDS) followed by 2 minutes in solution 2 (0.5X SSC, 0.01% SDS) and finally 2 minutes in solution 3 (0.06X SSC). Slides were centrifuged at 1500rpm for 1 minute to dry.
Microarrays were scanned using GenePix 4000B (Axon Instruments) scanner and microarray data was analysed using GenePix Pro 5.1 and Acuity 4.0 software (Axon Instruments). The data were analysed using linear discriminant analysis to identify a subset of genes that characterised the effects of the plant extract fractions on gene expression profiles ("gene expression signature"). Furthermore, genes were grouped according to function and assessment of trends within biological pathways was conducted.
Pathway analysis revealed that Fraction 18 (cells treated with 0.5% w/v Fraction 18 for 24h) appeared to have significant effects on a number of genes associated with glucose metabolism, including those involved with adipogenesis (C/EBPβ increased by 40%, SREBP2 increased by 22%), insulin signalling (PKCΘ increased by 54%, PIK3R2 increased by 32%, GSK3A increased by 10%, SORBSl increased by 24%). Glycogen synthesis (GYS 1 increased by 20%), glycolysis (DLAT increased by 49%, HK2 increased by TA, PDK4 increased by 10%), glucose transport (OGT increased by 13%) and mitochondrial activity (MFN2 increased by 20%). Taken together, these data provide significant support to the hypothesis that Fraction 18 had effects on processes that regulate glucose homeostasis.
Mass spectrometry analysis of Fraction 18
Low resolution LC-MS analysis of Fraction 18 was conducted. Fraction 18 appeared to contain a chemical entity with a m/z value of 517, and this was by far the most abundant entity observed following electrospray ionisation.
The fraction was then analysed by high resolution mass spectrometry and gave an ion + believed to be a protonated species [M+H ] with a mass of 517.2179090. This mass was compared with a library of possible combinations of atoms that would match this exact molecular weight. This identified a combination of 33H, 26C, 2N and 90 with a mass of
517.2180571 which differed from the fraction 18 species by only 1.921 x 10 mass units.
1 13
H and C NMR analysis of Fraction 18
The solid material was dissolved in D3OD and gave the following H and 13, C NMR data.
Figure imgf000054_0001
C26H32N2O9 Exact Mass: 516.2108 MoI. Wt: 516.5403 C, 60.46; H, 6.24; N, 5.42; 0, 27.1
Figure imgf000054_0002
Figure imgf000055_0001
Identification of strictosidinic acid
Partial linkage analysis revealed the presence of aromatic protons and a six membered aromatic carbon ring. Using proprietary computer software, a search of known structures was conducted for those that comprised a mass of -516, an empirical formula of 27C, 34H, 2N and 90 and which contain an aromatic ring. This identified strictosidinic acid (SA), a monoterpenoid indole alkaloid (empirical formula C27H34N2O9; CA Index Name 2H- Pyran-5-carboxylic acid, 3-ethenyl-2-(β-D-glucopyranosyloxy)-3,4-dihydro-4-[[(lS)- 2,3,4,9-tetrahydro-lH-pyrido[3,4-b]indol-l-yl]methyl]-, methyl ester, (2S,3R,4S)-(9C1)). It is the acid form of strictosidine, a glycosylated alkaloid intermediate that is a common precursor for some 2200 alkaloids. The 1H and 13C nmr data correlated well with that reported (Arbain, D. et al, 1993). Analysis of the MS and NMR data indicated that strictosidinic acid was the only chemical entity present in Fraction 18.
Effects of strictosidinic acid in bioassays
1. Glucose Uptake in 3T3-L1 adipocytes
Cell culture
3T3-L1 fibroblasts are grown in 10cm dishes and cultured in DMEM (4.5 g/L glucose) supplemented with 10% (v/v) heat-inactivated fetal bovine serum (FBS), 50 IU/ml pencillin and 50 μg/ml streptomycin. Cells for differentiation into adipocytes were maintained at post-confluence for 2 days, and then induced to differentiate by the addition of DMEM containing 10% FBS, 2 μg/ml insulin, 0.25 μM dexamethasone and 0.5 mM 3-isobutyl-l- methylxanthine. After 3 days, the induction medium was replaced with fresh DMEM containing 10% FBS and 2 μg/ml insulin for another 3 days. Adipocytes were maintained in DMEM with 5% FBS for 2 days thereafter. Adipocytes were used 8-12 days after initiation of differentiation, after which time greater than 80% of fibroblasts had differentiated into mature adipocytes.
Strictosidinic Acid Treatments 3T3-L1 adipocytes were re-seeded into gelatin-coated clear-wall clear-bottom 96-well plates (2 x 10cm dishes : 1 x 96-well plate) and maintained in DMEM with 5% FBS. After 24 hrs, adipocytes were treated with 0.2%, 0.5%, 1% or 2% (w/v) strictosidinic acid (SA) in DMEM (4.5 g/L glucose) containing 0.2% BSA for 16 to 18 hr (overnight). Each dose of SA was added to 4 wells and the following day, [3H] -2-deoxy glucose uptake was then measured (n=4). Strictosidinic acid stock solution was prepared as either 10% (w/v) (54 μg/ml) or 50% (w/v) (270 μg/ml) in milliQH20 and stored at 40C.
Glucose Uptake
Cells were washed twice in Dulbecco's PBS, pH 7.4 (Gibco) containing 0.2% (w/v) RIA- grade BSA, 0.5 mM MgCl2 and 0.5 mM CaCl2. Insulin at 0, 0.1, 1 and 10 nM is added for
30 min at 37°C. Uptake of 50 μM 2-deoxy glucose and 0.5 μCi 2-deoxy- [U-3H] glucose (NEN, PerkinElmer Life Sciences) per well was measured over the final 10 min of insulin stimulation (0, 0.5 and 1OnM). The reaction was stopped upon addition of ice-cold 80 μg/ml phloretin in PBS, pH 7.4 and cells solublised in 0.03% (w/v) SDS. Counts per minute (cpm) were measured by scintillator counter. Each measurement was performed in quadruplet in 1 assay, and each assay was repeated independently 3 times.
Results
Administration of strictosidinic acid caused a statistically significant, dose-dependent increase in glucose uptake by 3T3-L1 adipocytes. As shown in the table below, at doses of 0.5% and 1% (w/v) strictosidinic acid increased glucose uptake by up to 26%.
Figure imgf000057_0001
These results demonstrate that strictosidinic acid increases glucose uptake in 3T3-L1 adipocytes. Example 3: In Vivo Effects of Strictosidinic Acid
Compound Preparation and Administration
Dosing solutions of the test article, strictosidinic acid, were prepared fresh on each dosing day and stored at room temperature, protected from light. The compound was formulated in sterile NMP:PEG300:Saline (1:2:17, v/v). Stock solutions were prepared by dissolving the compound in NMP:PEG300 (1:2, v/v). Saline was added to the stock solution prior to administration of the compound. The solutions were mixed by vortexing immediately prior to dosing to ensure a homogeneous suspension of the compound. "Vehicle" corresponds to a sterile solution of NMP:PEG300:Saline (1:2:17, v/v).
Acute Tolerated Dose of Strictosidinic Acid in Balh/c mice
The compound was administered once only by oral gavage on Day 0 of the study. Administration was performed at a dosing volume of 10 mL/kg, at dosing concentrations of 31, 62.5, 125, and 250 mg/kg.
Each animal's body weight was measured immediately prior to dosing each day. The actual volume administered to each mouse was calculated and adjusted based on the body weight.
Starting with the lowest concentration (31 mg/kg, Group 1), one mouse was treated, followed by a 2 hour observation period. As no adverse effects were observed during this time, the remaining mice in the group were treated. Twenty four hours after the first concentration was administered, one mouse from the next dose concentration (62.5 mg/kg, Group 2) was treated. As no adverse effects were observed during a 2 hour observation period, the remaining mice in the group were treated. The process was continued until all dose concentrations were administered (125 mg/kg, Group 3 and 250 mg/kg, Group 4).
Results The acute tolerated dose (ATD) is defined as the highest dose (administered as a single dose) that does not induce a weight loss in an individual animal in excess of 15% or cause death or severe morbidity, at any time during the study period.
Sixteen female Balb/c mice were randomised into 4 treatment groups, each with 4 mice. Each group had similar mean starting body weight. The groups were allocated a different treatment regimen of strictosidinic acid, administered once only, by oral gavage. The compound was administered at a volume of 10 mL/kg, at dosing concentrations of 31, 62.5, 125 and 250 mg/kg.
Body weight measurements were made daily from Day 0 of the study and were continued until 7 days after treatment was finished, and are detailed in Table A.
None of the mice lost body weight in excess of the defined parameters of the study, and no adverse effects were observed in the mice during the study period. The ATD for strictosidinic acid administered by oral gavage was not achieved in this study, and is therefore determined to be >250 mg/kg (the highest dose concentration tested in the study).
Table A: Body weight data for ATD Study: strictosidinic acid in Balb/c mice
Figure imgf000059_0001
Effect of Strictosidinic acid on fasting blood glucose levels in diet-induced obese mice (Mus Musculus)
Animals Male C57B1/6J mice were obtained at 8 weeks of age from Animal Resources Centre (Canning Vale, Western Australia). Animals were individually housed and allowed free movement and ad libitum access to water and food. Animals were maintained on a 12 hour light (6 am-6 pm) and 12 hour dark (6 pm-6 am) cycle. After two weeks acclimatizing, mice were fed a high fat rich diet (Research Diets, Inc. [New Jersey, USA] with a total energy density of 4.7 kcal/g. Caloric distribution in the diet was 20% from protein, 35 % from carbohydrates, and 45 % from fat) for 11 weeks.
Treatment
During the last week, animals in groups of 6 were treated with doses selected from: vehicle, 25mg/kg, 50mg/kg, 100mg/kg and 175mg/kg of Strictosidinic acid in
NMP:PEG300:Saline (l:2:17, v/v) by oral gavage (5 μl per gram of body weight) using feeding tubes 18ga (1.2mm)*38mm from Instech Solomon (Plymouth, USA), while control animals were administered with vehicle only. It became clear early in the study that SA was not fully soluble at the concentration required for the 175 mg/kg dose, so this group was discontinued and not included in the analysis.
Fasting Blood Glucose Levels
Animals were fasted overnight and blood samples collected from the tail at 10am on days 0 and 9 of the study. The results are presented in Table B. Table B: Effect of Strictosidinic acid treatment on fasting blood glucose concentration in diet-induced obese mice
Figure imgf000061_0001
Data represent the mean ± SE of 4-6 animals per group.
Intraperitoneal Glucose Tolerance Test: Blood glucose concentrations during ipGTT
Animals were subjected to an intraperitoneal glucose tolerance test (ipGTT) prior to commencement of dosing, and on day 7 after final dosing. Animals were fasted overnight and then administered glucose (50%w/v in saline: Pharmalab) by intraperitoneal injection at a dose of 2g/kg. Blood glucose levels were monitored at t = Omin, 15min, 30min, 60min and 90min after the glucose dose using commercially available glucometers (Accuchek Advantage, Roche) based on the electric current generated when a blood drop is spotted on a test strip resulting in conversion of the glucose present in the sample to gluconolactone by the glucose dehydrogenase enzyme, in the presence of the coenzyme (PQQ). The results are depicted in Table C.
Table C: Effect of Strictosidinic acid treatment on area under plasma glucose curve during ipGTT in diet-induced obese mice
Figure imgf000061_0002
Data represent the mean ± SE of 5-6 animals per group. Intraperitoneal Glucose Tolerance Test: Plasma insulin concentrations during ipGTT
Plasma insulin concentrations were determined using a commercially-available insulin ELISA kit (Crystal Chem) which was used according to the manufacturer's instructions. The results are presented in Table D.
Table D: Effect of Strictosidinic acid treatment on plasma insulin concentration during ipGTT in diet-induced obese mice
Figure imgf000062_0001
Data represent the mean ± SE of 5-6 animals per group. *unpaired ttest value vs vehicle at specified timepoint.
Determination of Epididymal Fat as a Percentage of Body Weight
Animals were euthenized by cervical dislocation, and their weights were independently recorded. The animals were then dissected and their livers and epididymal fat pads were surgically excised, and their individual weights recorded. The results are presented in Table E. Table E: Effect of Strictosidinic acid treatment epididymal fat mass as % of body weight in diet-induced obese mice
Figure imgf000063_0001
Data represent the mean ± SE of 5-6 animals per group.
BIBLIOGRAPHY
Arbain, D. et al., Amt. J. Chem. 1993, 46: 977-985. Galan, M. C. et al. , Tetrahedron Lett. , 2006, 1563- 1565. Geerlings et al., J. Biol Chem., 2000, 275: 3051-3056.
Govers et al, MCB, 2004, 29: 6456-6466.
Kitajima, M. et al, Chem. Pharm. Bull. 2002, 50(10): 1376-1378.
Larock, R.E., Comprehensive Organic Transformations, VCH Publishers, 1989. The Merck Index, Thirteenth Edition, ONR-82, 310. Morita et al, Gene Therapy, 2000, 7: 1063-1066.
Patthy-Lukats, A. et al, J. Nat. Prod. 1999, 62: 1492-1499.
QvLon et aL, PNAS, 1994, 91: 5587-5591.
Stδckigt, J. et al, J. Chem. Soc. Chem. Commun. 1977, 646-648. Treimer, J. F. et al, Eur. J. Biochem. 1979, 81 : 225-233. Yamazaki et al, Phytochemistry, 2003, 62: 461-470.
United States Patent No. 6,720,331
United States Patent No. 6,350,757.

Claims

THE CLAIMS DEFINING THE INVENTION ARE AS FOLLOWS:
1. A method of increasing glucose uptake by a cell comprising contacting said cell with a compound of Formula (I):
Figure imgf000065_0001
wherein,
is a single or a double bond;
Ri-R4 are independently selected from hydrogen, hydroxy, thiol, halo, CO2H, carboxy ester, amino, nitro, cyano, amido, sulfoxide, sulfonamide, sulfonyl, sulfate, sulfonate, phosphate, phosphonate, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted carbocyclyl, optionally substituted heterocyclyl, optionally substituted acyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aryloxy, optionally substituted heteroaryloxy, optionally substituted carbocyclyloxy, optionally substituted heterocyclyloxy, optionally substituted aralkyloxy, optionally substituted acyloxy, optionally substituted alkylthio, optionally substituted alkenylthio, optionally substituted alkynylthio, optionally substituted arylthio, optionally substituted carbocyclylthio, optionally substituted aralkylthio, optionally substituted heteroarylthio, optionally substituted heterocyclylthio, optionally substituted acylthio or any 2 adjacent R1-R4 together form a -0-(CHa)n-O- or -NR-(CH2)n-NR-group wherein n is 1 or 2 and each R is independently H or Ci-6alkyl;
Re is absent when is a double bond;
R5 and R6 (when present) independently are selected from hydrogen, optionally substituted alkyl, optionally substituted acyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted aralkyl, optionally substituted heterocyclyl, optionally substituted carbocyclyl, and amido;
R7 is selected from hydrogen, optionally substituted alkyl, CO2H, carboxy ester, amido, optionally substituted heteroaryl and optionally substituted aryl;
Rs is selected from hydrogen, optionally substituted alkyl, optionally substituted aryl, optionally substituted heteroaryl, CO2H, carboxy ester and SO3H;
R7a and R8a are independently selected from hydrogen, optionally substituted alkyl and optionally substituted aryl.
COX forms a group selected from optionally substituted acyl, CO2H, carboxy ester and amido,
Y is selected from hydrogen, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclyl, optionally substituted carbocyclyl, optionally substituted acyl and a saccharide group, Z is selected from optionally substituted at least C2alkyl, optionally substituted alkenyl and optionally substituted alkynyl,
or a pharmaceutically acceptable salt or prodrug thereof.
2. A method for regulating glucose homeostasis in a subject in need thereof comprising the administration of a compound of Formula (I) as defined in claim 1 or a pharmaceutically acceptable salt or prodrug thereof.
3. A method of treating insulin resistance in a subject in need thereof comprising the administration of a compound of Formula (I) as defined in claim 1 or a pharmaceutically acceptable salt or prodrug thereof.
4. A method of lowering blood glucose levels in a subject in need thereof comprising the administration of a compound of Formula (I) as defined in claim 1 or a pharmaceutically acceptable salt or prodrug thereof.
5. A method for treating a disease or condition, or symptom thereof, in which insulin resistance is involved comprising the administration of a compound of Formula (I) as. defined in claim 1 or a pharmaceutically acceptable salt or prodrug thereof to subject in need thereof.
6. A method according to claim 5 wherein the disease or condition is NIDDM, gestational diabetes, impaired glucose tolerance, impaired fasting glucose or syndrome X.
7. A method according to any one of claims 1 to 6, wherein:
R1-4 are independently selected from hydrogen, alkyl, alkoxyalkyl, alkoxy, alkylthio, halo, trifluoromethyl, trichloromethyl, tribromomethyl, hydroxy, phenyl (which itself may be further substituted), benzyl (wherein benzyl itself may be further substituted), phenoxy (wherein phenyl itself may be further substituted), benzyloxy (wherein benzyl itself may be further substituted), amino, alkylamino, dialkylamino, acylamino, phenylamino (wherein phenyl itself may be further substituted), nitro, formyl, -C(O)-alkyl, O-C(O)-alkyl, benzoyl (wherein benzyl itself may be further substituted, benzoyloxy (wherein benzyl itself may be further substituted, COjH5 CCtølkyl, Cθ2phenyl, C02benzyl (wherein benzyl itself may be further substituted, CONH2, CONHphenyl (wherein phenyl itself may be further substituted, CONHbenzyl (wherein benzyl itself may be further substituted, CONHalkyl, CONdialkyl, aminoalkyl, thioalkyl, carboxyalkyl, carboxyesteralkyl, amidoalkyl, formylalkyl, acylalkyl, nitroalkyl, sulfoxidealkyl, sulfonylalkyl, sulfonamidoalkyl, and where 2 adjacent carbon atoms are substituted by one end each of a -O-(CH2)n-O- or -NH-(CH2)n-NH- group, wherein n is 1 or 2;
R5 and R6 are selected from hydrogen, optionally substituted C1-6alkyl, optionally substituted C(O)C1-6alkyl, optionally substituted C(O)phenyl, and optionally substituted C(O)benzyl;
R7 is selected from hydrogen, CO2H, CO2C1-6alkyl, CONH2, CONHC1-6alkyl and CONHaryl;
Rg is hydrogen;
R7a and R8a are hydrogen;
X is selected from OH, OCi-I0alkyl, NHC1-loalkyl, NHphenyl and NHbenzyl;
Y is H or a saccharide;
Z is CH2=CH2 or C2-10alkyl
8. A method according to any one of claims 1 to 6 wherein the compound is strictosidinic acid.
9. A method according to claim 8 wherein the compound is in substantially pure or isolated form.
10. A compound of Formula (I):
Figure imgf000069_0001
wherein,
is a single or a double bond;
R1-R4 are independently selected from hydrogen, hydroxy, thiol, halo, CO2H, carboxy ester, amino, nitro, cyano, amido, sulfoxide, sulfonamide, sulfonyl, sulfate, sulfonate, phosphate, phosphonate, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted carbocyclyl, optionally substituted heterocyclyl, optionally substituted acyl, optionally substituted alkoxy, optionally substituted alkenyloxy, optionally substituted alkynyloxy, optionally substituted aryloxy, optionally substituted heteroaryloxy, optionally substituted carbocyclyloxy, optionally substituted heterocyclyloxy, optionally substituted aralkyloxy, optionally substituted acyloxy, optionally substituted alkylthio, optionally substituted alkenylthio, optionally substituted alkynylthio, optionally substituted arylthio, optionally substituted carbocyclylthio, optionally substituted aralkylthio, optionally substituted heteroarylthio, optionally substituted heterocyclylthio, optionally substituted acylthio or any 2 adjacent R1-R4 together form a -O-(CH2)n-O- or -NR-(CH2)n-NR-group wherein n is 1 or 2 and each R is independently H or C1-6alkyl;
R6 is absent when "■ is a double bond;
R5 and R6 (when present) independently are selected from hydrogen, optionally substituted alkyl, optionally substituted acyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted aralkyl, optionally substituted heterocyclyl, optionally substituted carbocyclyl, and amido;
R7 is selected from hydrogen, optionally substituted alkyl, CO2H, carboxy ester, amido, optionally substituted heteroaryl and optionally substituted aryl;
R8 is selected from hydrogen, optionally substituted alkyl, optionally substituted aryl, optionally substituted heteroaryl, CO2H, carboxy ester and SO3H;
R7a and R8a are independently selected from hydrogen, optionally substituted alkyl and optionally substituted aryl.
COX forms a group selected from optionally substituted acyl, CO2H, carboxy ester and amido,
Y is selected from hydrogen, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclyl, optionally substituted carbocyclyl, optionally substituted acyl and a saccharide group, Z is selected from optionally substituted at least C2alkyl, optionally substituted alkenyl and optionally substituted alkynyl,
or a pharmaceutically acceptable salt or ester thereof, provided that the compound is not (3R) or (3S) strictosidine, strictosidine ethyl ester or allyl ester, strictosidinic acid, 5- carboxystrictosidine, 3,4-dehydro-5-carboxystrictosidine, 3,4-dehydro5- carbomethoxystrictosidine or gluco-acetylated forms thereof.
11. A composition comprising a compound of Formula (I), as defined in claim 1, or a pharmaceutically acceptable salt or prodrug thereof, together with a pharmaceutically acceptable excipient.
PCT/AU2007/001593 2006-10-20 2007-10-19 Methods for regulating glucose homeostasis and agents therefor WO2008046155A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US85335306P 2006-10-20 2006-10-20
US60/853,353 2006-10-20

Publications (1)

Publication Number Publication Date
WO2008046155A1 true WO2008046155A1 (en) 2008-04-24

Family

ID=39313518

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/AU2007/001593 WO2008046155A1 (en) 2006-10-20 2007-10-19 Methods for regulating glucose homeostasis and agents therefor

Country Status (1)

Country Link
WO (1) WO2008046155A1 (en)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8629158B2 (en) 2009-07-01 2014-01-14 Albany Molecular Research, Inc. Azabicycloalkane-indole and azabicycloalkane-pyrrolo-pyridine MCH-1 antagonists, methods of making, and use thereof
US8637501B2 (en) 2009-07-01 2014-01-28 Albany Molecular Research, Inc. Azinone-substituted azepino[b]indole and pyrido-pyrrolo-azepine MCH-1 antagonists, methods of making, and use thereof
US8697700B2 (en) 2010-12-21 2014-04-15 Albany Molecular Research, Inc. Piperazinone-substituted tetrahydro-carboline MCH-1 antagonists, methods of making, and uses thereof
US8716308B2 (en) 2008-01-11 2014-05-06 Albany Molecular Research, Inc. (1-azinone)-substituted pyridoindoles
US8993765B2 (en) 2010-12-21 2015-03-31 Albany Molecular Research, Inc. Tetrahydro-azacarboline MCH-1 antagonists, methods of making, and uses thereof
US9073925B2 (en) 2009-07-01 2015-07-07 Albany Molecular Research, Inc. Azinone-substituted azabicycloalkane-indole and azabicycloalkane-pyrrolo-pyridine MCH-1 antagonists, methods of making, and use thereof
US10112915B2 (en) 2015-02-02 2018-10-30 Forma Therapeutics, Inc. 3-aryl bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10183934B2 (en) 2015-02-02 2019-01-22 Forma Therapeutics, Inc. Bicyclic [4,6,0] hydroxamic acids as HDAC inhibitors
US10555935B2 (en) 2016-06-17 2020-02-11 Forma Therapeutics, Inc. 2-spiro-5- and 6-hydroxamic acid indanes as HDAC inhibitors
CN111592537A (en) * 2019-11-13 2020-08-28 江西中医药大学 Sinomenium majus indole alkaloid for reducing hyperuricemia and application thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
HUT46032A (en) * 1987-03-26 1988-09-28 Gyogynoeveny Kutato Intezet Kv Process for producing semisynthetic glycolalkaloids with indole skeleton, particularly new n under b-substituted vincosides and n under b-substituted strictosidines

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
HUT46032A (en) * 1987-03-26 1988-09-28 Gyogynoeveny Kutato Intezet Kv Process for producing semisynthetic glycolalkaloids with indole skeleton, particularly new n under b-substituted vincosides and n under b-substituted strictosidines

Non-Patent Citations (50)

* Cited by examiner, † Cited by third party
Title
CHEMICAL & PHARMACEUTICAL BULLETIN, vol. 40, no. 9, 1992, pages 2588 - 2590 *
CHEMICAL COMMUNICATIONS, no. 18, 1972, pages 1006 - 1007 *
CHEMICAL COMMUNICATIONS, no. 21, 1968, pages 1282 - 1284 *
DATABASE CA [online] AIMI ET AL.: "Synthesis of lyaloside, a prototypal b-carboline gluco indole alkaloid in rubiaceous plants", Database accession no. (118:124854) *
DATABASE CA [online] BATTERSBY ET AL.: "Alkaloid biosynthesis. XV. Partial synthesis and isolation of vincoside and isovincoside. Biosynthesis of the three major classes of indole alkaloids from vincoside", Database accession no. (71:10344) *
DATABASE CA [online] BATTERSBY ET AL.: "Partial synthesis and isolation of vincoside and isovincoside: Biosynthesis of the three major classes of indole alkaloids from the b-carboline system", Database accession no. (70:29243) *
DATABASE CA [online] BLACKSTOCK ET AL.: "Adina alkaloids. Deoxycordifoline, 3a,5a- and 3b,5a-tetrahydrodeoxycordifoline lactam", Database accession no. (77:152412) *
DATABASE CA [online] BRANDT ET AL.: "Main glucosidase conversion products of the gluco-alkaloids dolichantoside and palicoside", Database accession no. (135:208208) *
DATABASE CA [online] BROWN ET AL.: "Adina alkaloids. Structure of 5-oxostrictosidine", Database accession no. (83:43561) *
DATABASE CA [online] BROWN ET AL.: "Alternative cyclization of 3a- and 3b-indole alkaloid glucosides", Database accession no. (83:59132) *
DATABASE CA [online] BROWN ET AL.: "Biomimetic conversion of vincoside into heteroyohimbine alkaloids", Database accession no. (82:73279) *
DATABASE CA [online] BROWN ET AL.: "Biomimetic synthesis of indole alkaloids. Dihydromancunine", Database accession no. (82:73275) *
DATABASE CA [online] BROWN ET AL.: "Conversion of secologanin into Corynanthe-type alkaloids", Database accession no. (80:108730) *
DATABASE CA [online] CHUNG ET AL.: "Effect of precursors on growt and alkaloid production in Chinchona ledgeriana leaf-shoot organ cultures", Database accession no. (109:187425) *
DATABASE CA [online] COUNE ET AL.: "Minor alkaloids from Strychnos gossweileri", Database accession no. (93:217906) *
DATABASE CA [online] Database accession no. (111:154290) *
DATABASE CA [online] FERRARI ET AL.: "Constituents of Guettarda platypoda", Database accession no. (106:172925) *
DATABASE CA [online] GERASIMENKO ET AL.: "Heterologous expression of a Rauvolfia cDNA encoding strictosidine glucosidase, a biosynthetic key to over 2000 monoterpenoid indole alkaloids", Database accession no. (137:306556) *
DATABASE CA [online] HECKENDORF ET AL.: "Biosynthesis of camptothecin. II. Confirmation that isovincoside, not vincoside, is the penultimate biosynthetic precursor of indole alkaloids", Database accession no. (88:133299) *
DATABASE CA [online] MATTES ET AL.: "Absolute configuration of vincoside", Database accession no. (83:193574) *
DATABASE CA [online] OHMORI ET AL.: "The correlation between CD spectra and C-3 stereochemistries of some monoterpenoid glucoindole alkaloids", Database accession no. (130:223464) *
DATABASE CA [online] PATTHY-LUKATS ET AL.: "Configurative Correlation and Conformational Analysis of Strictosidine and Vincoside Derivatives", Database accession no. (131:337256) *
DATABASE CA [online] PAUL ET AL.: "Novel bis(monoterpenoid)indole alkaloids from Psychotria bahiensis", Database accession no. (139:82036) *
DATABASE CA [online] SILVESTRINI ET AL.: "Effects of alkaloid precursor feeding on a Camptotheca acuminata cell line", Database accession no. (138:103683) *
DATABASE CA [online] SMITH ET AL.: "5a-Carboxystrictosidine", Database accession no. (75:130069) *
DATABASE CA [online] STOECKIGT ET AL.: "Strictosidine (isovincoside): the key intermediate in the biosynthesis of monoterpenoid indole alkaloids", Database accession no. (88:100432) *
DATABASE CA [online] TAKAYAMA ET AL.: "Hunterioside B, a disaccharide carrying monoterpenoid indole alkaloid, from Hunteria zeylanica", Database accession no. (128:306176) *
DATABASE CA CARDOSO ET AL.: "Indole glucoalkaloids from Chimarrhis turbinate and their evaluation as antioxidant agents and acetylcholinesterase inhibitors" *
DATABASE CA ITOH ET AL.: "Two chromone-secoiridoid glycosides and three indole alkaloid glycosides from Neonauclea sessilifolia" *
EUROPEAN JOURNAL OF BIOCHEMISTRY, vol. 269, no. 8, 2002, pages 2204 - 2213 *
EXPERIENTIA, vol. 31, no. 5, 1975, pages 505 - 507 *
HERBA HUNGARICA, vol. 19, no. 1, 1980, pages 189 - 193 *
HETEROCYCLES, vol. 47, no. 1, 1998, pages 87 - 90 *
INTERNATIONAL CONGRESS SERIES, vol. 1157, 1998, pages 573 - 582 *
JOURNAL OF NATURAL PRODUCTS, vol. 49, no. 6, 1986, pages 1150 - 1151 *
JOURNAL OF NATURAL PRODUCTS, vol. 62, no. 11, 1999, pages 1492 - 1499 *
JOURNAL OF NATURAL PRODUCTS, vol. 66, no. 6, 2003, pages 752 - 754 *
JOURNAL OF NATURAL PRODUCTS, vol. 67, no. 11, 2004, pages 1882 - 1885 *
JOURNAL OF THE AMERICAN CHEMICAL SOCIETY, vol. 97, no. 21, 1975, pages 6270 - 6271 *
JOURNAL OF THE CHEMICAL SOCIETY SECTION C: ORGANIC, no. 8, 1969, pages 1193 - 1200 *
JOURNAL OF THE CHEMICAL SOCIETY SECTION D: CHEMICAL COMMUNICATIONS, no. 16, 1971, pages 908 - 909 *
JOURNAL OF THE CHEMICAL SOCIETY, CHEMICAL COMMUNICATIONS, no. 18, 1974, pages 740 - 742 *
JOURNAL OF THE CHEMICAL SOCIETY, CHEMICAL COMMUNICATIONS, no. 18, 1974, pages 756 - 757 *
JOURNAL OF THE CHEMICAL SOCIETY, CHEMICAL COMMUNICATIONS, no. 18, 1977, pages 646 - 648 *
JOURNAL OF THE CHEMICAL SOCIETY, CHEMICAL COMMUNICATIONS, no. 22, 1973, pages 886 - 888 *
JOURNAL OF THE CHEMICAL SOCIETY, CHEMICAL COMMUNICATIONS, no. 8, 1975, pages 295 - 296 *
PHYTOCHEMISTRY, vol. 57, no. 5, 2001, pages 653 - 659 *
PHYTOCHEMISTRY, vol. 62, no. 3, 2003, pages 359 - 369 *
PLANT PHYSIOLOGY AND BIOCHEMISTRY, vol. 40, no. 9, 2002, pages 749 - 753 *
PLANTA MEDICA, vol. 54, no. 3, 1988, pages 206 - 210 *

Cited By (47)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8716308B2 (en) 2008-01-11 2014-05-06 Albany Molecular Research, Inc. (1-azinone)-substituted pyridoindoles
US9296743B2 (en) 2008-01-11 2016-03-29 Albany Molecular Research, Inc. (1-azinone)-substituted pyridoindoles
US9650378B2 (en) 2008-01-11 2017-05-16 Albany Molecular Research, Inc. (1-azinone)-substituted pyridoindoles
US8629158B2 (en) 2009-07-01 2014-01-14 Albany Molecular Research, Inc. Azabicycloalkane-indole and azabicycloalkane-pyrrolo-pyridine MCH-1 antagonists, methods of making, and use thereof
US8637501B2 (en) 2009-07-01 2014-01-28 Albany Molecular Research, Inc. Azinone-substituted azepino[b]indole and pyrido-pyrrolo-azepine MCH-1 antagonists, methods of making, and use thereof
US9073925B2 (en) 2009-07-01 2015-07-07 Albany Molecular Research, Inc. Azinone-substituted azabicycloalkane-indole and azabicycloalkane-pyrrolo-pyridine MCH-1 antagonists, methods of making, and use thereof
US8697700B2 (en) 2010-12-21 2014-04-15 Albany Molecular Research, Inc. Piperazinone-substituted tetrahydro-carboline MCH-1 antagonists, methods of making, and uses thereof
US8993765B2 (en) 2010-12-21 2015-03-31 Albany Molecular Research, Inc. Tetrahydro-azacarboline MCH-1 antagonists, methods of making, and uses thereof
US10457652B2 (en) 2015-02-02 2019-10-29 Forma Therapeutics, Inc. 3-alkyl-4-amido-bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10494352B2 (en) 2015-02-02 2019-12-03 Forma Therapeutics, Inc. 3-aryl-bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10214501B2 (en) 2015-02-02 2019-02-26 Forma Therapeutics, Inc. 3-alkyl bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10214500B2 (en) 2015-02-02 2019-02-26 Forma Therapeutics, Inc. 3-alkyl-4-amido-bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10239845B2 (en) 2015-02-02 2019-03-26 Forma Therapeutics, Inc. 3-aryl-4-amido-bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10377726B2 (en) 2015-02-02 2019-08-13 Forma Therapeutics, Inc. 3-aryl-4-amido-bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10407418B2 (en) 2015-02-02 2019-09-10 Forma Therapeutics, Inc. Bicyclic [4,6,0] hydroxamic acids as HDAC inhibitors
US10414738B2 (en) 2015-02-02 2019-09-17 Forma Therapeutics, Inc. 3-alkyl bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10421731B2 (en) 2015-02-02 2019-09-24 Forma Therapeutics, Inc. 3-alkyl bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10421732B2 (en) 2015-02-02 2019-09-24 Forma Therapeutics, Inc. 3-alkyl-4-amido-bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10428031B2 (en) 2015-02-02 2019-10-01 Forma Therapeutics, Inc. 3-alkyl bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10442776B2 (en) 2015-02-02 2019-10-15 Forma Therapeutics, Inc. 3-alkyl-4-amido-bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10450284B2 (en) 2015-02-02 2019-10-22 Forma Therapeutics, Inc. 3-aryl-4-amido-bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10450283B2 (en) 2015-02-02 2019-10-22 Forma Therapeutics, Inc. 3-alkyl bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10112915B2 (en) 2015-02-02 2018-10-30 Forma Therapeutics, Inc. 3-aryl bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10464909B2 (en) 2015-02-02 2019-11-05 Forma Therapeutics, Inc. 3-alkyl-4-amido-bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10464910B2 (en) 2015-02-02 2019-11-05 Forma Therapeutics, Inc. 3-alkyl-4-amido-bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10472337B2 (en) 2015-02-02 2019-11-12 Forma Therapeutics, Inc. 3-aryl-4-amido-bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10479772B2 (en) 2015-02-02 2019-11-19 Forma Therapeutics, Inc. 3-aryl-4-amido-bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10183934B2 (en) 2015-02-02 2019-01-22 Forma Therapeutics, Inc. Bicyclic [4,6,0] hydroxamic acids as HDAC inhibitors
US10494354B2 (en) 2015-02-02 2019-12-03 Forma Therapeutics, Inc. 3-aryl-bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10494351B2 (en) 2015-02-02 2019-12-03 Forma Therapeutics, Inc. 3-aryl-bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10494353B2 (en) 2015-02-02 2019-12-03 Forma Therapeutics, Inc. 3-aryl-4-amido-bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10501424B2 (en) 2015-02-02 2019-12-10 Forma Therapeutics, Inc. 3-aryl-bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10513501B2 (en) 2015-02-02 2019-12-24 Forma Therapeutics, Inc. 3-alkyl bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US11891365B2 (en) 2015-02-02 2024-02-06 Valo Health, Inc. 3-alkyl-4-amido-bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US11702412B2 (en) 2015-02-02 2023-07-18 Valo Health, Inc. Bicyclic [4,6,0] hydroxamic acids as HDAC inhibitors
US10822316B2 (en) 2015-02-02 2020-11-03 Valo Early Discovery, Inc. 3-aryl-bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10829462B2 (en) 2015-02-02 2020-11-10 Valo Early Discovery, Inc. 3-alkyl bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10829461B2 (en) 2015-02-02 2020-11-10 Valo Early Discovery, Inc. 3-aryl-4-amido-bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10870645B2 (en) 2015-02-02 2020-12-22 Valo Early Discovery, Inc. Bicyclic [4,6,0] hydroxamic acids as HDAC inhibitors
US11279681B2 (en) 2015-02-02 2022-03-22 Valo Health, Inc. 3-alkyl bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10988450B2 (en) 2015-02-02 2021-04-27 Valo Early Discovery, Inc. 3-alkyl-4-amido-bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US11274084B2 (en) 2015-02-02 2022-03-15 Valo Health, Inc. 3-aryl-4-amido-bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US11274085B2 (en) 2015-02-02 2022-03-15 Valo Health, Inc. 3-aryl-bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors
US10874649B2 (en) 2016-06-17 2020-12-29 Valo Early Discovery, Inc. 2-spiro-5- and 6-hydroxamic acid indanes as HDAC inhibitors
US11730721B2 (en) 2016-06-17 2023-08-22 Valo Health, Inc. 2-spiro-5- and 6-hydroxamic acid indanes as HDAC inhibitors
US10555935B2 (en) 2016-06-17 2020-02-11 Forma Therapeutics, Inc. 2-spiro-5- and 6-hydroxamic acid indanes as HDAC inhibitors
CN111592537A (en) * 2019-11-13 2020-08-28 江西中医药大学 Sinomenium majus indole alkaloid for reducing hyperuricemia and application thereof

Similar Documents

Publication Publication Date Title
WO2008046155A1 (en) Methods for regulating glucose homeostasis and agents therefor
Tronci et al. BDNF over-expression induces striatal serotonin fiber sprouting and increases the susceptibility to L-DOPA-induced dyskinesia in 6-OHDA-lesioned rats
US8377963B2 (en) Insulin-regulated aminopeptidase (IRAP) inhibitors and uses thereof
US11834663B2 (en) Fibrotic treatment
EP1789079B1 (en) Insulin-regulated aminopeptidase (IRAP) inhibitors and uses thereof
Daniele et al. Human neural stem cell aging is counteracted by α-glycerylphosphorylethanolamine
Zhang et al. Akt3 deletion in mice impairs spatial cognition and hippocampal CA 1 long long‐term potentiation through downregulation of mTOR
WO2018100536A1 (en) Methods of treating cancer
Udumula et al. Pharmacological evaluation of novel PKR inhibitor indirubin-3-hydrazone in-vitro in cardiac myocytes and in-vivo in wistar rats
Hu et al. Regulation of M1-receptor mRNA stability by smilagenin and its significance in improving memory of aged rats
JP6305422B2 (en) Method for inhibiting SHC-1 / P66 to overcome aging-related diseases
AU2020358184B2 (en) Preparation of drug for treating Alzheimer&#39;s disease
KR20200112898A (en) Use of caramycin or its active ingredient
CN114478464B (en) Inflammatory corpuscle selective inhibitor and synthetic method and application thereof
AU2005282229B2 (en) Enzyme inhibitors and uses thereof
JP2011126791A (en) Substances for inhibiting expression of genes for sensitivity to allergic disorders
JP2021063127A (en) Nurr1/RXR ACTIVATOR FOR TREATMENT OR PREVENTION OF NEURODEGENERATIVE DISEASES
TWI484953B (en) Uses of polymer composition for the manufacture of a medicament to modulate functions of shc-1/p66 gene
KR20040034655A (en) A therapeutic agent containing celastrol, celaphanol, sesquiterpene ester or an extract of celastrus obiculatus as an effective ingredient for the treatment of inflammatory disease, immune disorders or cancers
KR20100023681A (en) Use in anti-cancer agent of 2,3-dihydro-5-methoxy-2-phenyl-4h-1-benzopyran-4-on

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 07815398

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 07815398

Country of ref document: EP

Kind code of ref document: A1