US20170029368A1 - Novel sulphur containing lipids for use as food supplement or as medicament - Google Patents

Novel sulphur containing lipids for use as food supplement or as medicament Download PDF

Info

Publication number
US20170029368A1
US20170029368A1 US15/040,260 US201615040260A US2017029368A1 US 20170029368 A1 US20170029368 A1 US 20170029368A1 US 201615040260 A US201615040260 A US 201615040260A US 2017029368 A1 US2017029368 A1 US 2017029368A1
Authority
US
United States
Prior art keywords
group
ethyl
mmol
acid
icosa
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US15/040,260
Inventor
Anne Kristin Holmeide
Ragnar Hovland
Morten Braendvang
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Pronova Biopharma Norge AS
Original Assignee
Pronova Biopharma Norge AS
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=40419454&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20170029368(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Pronova Biopharma Norge AS filed Critical Pronova Biopharma Norge AS
Priority to US15/040,260 priority Critical patent/US20170029368A1/en
Publication of US20170029368A1 publication Critical patent/US20170029368A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C323/00Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups
    • C07C323/50Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and carboxyl groups bound to the same carbon skeleton
    • C07C323/61Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and carboxyl groups bound to the same carbon skeleton having the sulfur atom of at least one of the thio groups bound to a carbon atom of a ring other than a six-membered aromatic ring of the carbon skeleton
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23LFOODS, FOODSTUFFS, OR NON-ALCOHOLIC BEVERAGES, NOT COVERED BY SUBCLASSES A21D OR A23B-A23J; THEIR PREPARATION OR TREATMENT, e.g. COOKING, MODIFICATION OF NUTRITIVE QUALITIES, PHYSICAL TREATMENT; PRESERVATION OF FOODS OR FOODSTUFFS, IN GENERAL
    • A23L33/00Modifying nutritive qualities of foods; Dietetic products; Preparation or treatment thereof
    • A23L33/10Modifying nutritive qualities of foods; Dietetic products; Preparation or treatment thereof using additives
    • A23L33/115Fatty acids or derivatives thereof; Fats or oils
    • A23L33/12Fatty acids or derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/095Sulfur, selenium, or tellurium compounds, e.g. thiols
    • A61K31/10Sulfides; Sulfoxides; Sulfones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/215Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids
    • A61K31/22Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acyclic acids, e.g. pravastatin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C313/00Sulfinic acids; Sulfenic acids; Halides, esters or anhydrides thereof; Amides of sulfinic or sulfenic acids, i.e. compounds having singly-bound oxygen atoms of sulfinic or sulfenic groups replaced by nitrogen atoms, not being part of nitro or nitroso groups
    • C07C313/02Sulfinic acids; Derivatives thereof
    • C07C313/04Sulfinic acids; Esters thereof
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C317/00Sulfones; Sulfoxides
    • C07C317/44Sulfones; Sulfoxides having sulfone or sulfoxide groups and carboxyl groups bound to the same carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C323/00Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups
    • C07C323/50Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and carboxyl groups bound to the same carbon skeleton
    • C07C323/51Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and carboxyl groups bound to the same carbon skeleton having the sulfur atoms of the thio groups bound to acyclic carbon atoms of the carbon skeleton
    • C07C323/52Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and carboxyl groups bound to the same carbon skeleton having the sulfur atoms of the thio groups bound to acyclic carbon atoms of the carbon skeleton the carbon skeleton being acyclic and saturated
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C323/00Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups
    • C07C323/50Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and carboxyl groups bound to the same carbon skeleton
    • C07C323/51Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and carboxyl groups bound to the same carbon skeleton having the sulfur atoms of the thio groups bound to acyclic carbon atoms of the carbon skeleton
    • C07C323/54Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and carboxyl groups bound to the same carbon skeleton having the sulfur atoms of the thio groups bound to acyclic carbon atoms of the carbon skeleton the carbon skeleton being acyclic and unsaturated
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C323/00Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups
    • C07C323/50Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and carboxyl groups bound to the same carbon skeleton
    • C07C323/51Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and carboxyl groups bound to the same carbon skeleton having the sulfur atoms of the thio groups bound to acyclic carbon atoms of the carbon skeleton
    • C07C323/55Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and carboxyl groups bound to the same carbon skeleton having the sulfur atoms of the thio groups bound to acyclic carbon atoms of the carbon skeleton the carbon skeleton being unsaturated and containing rings other than six-membered aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C323/00Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups
    • C07C323/50Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and carboxyl groups bound to the same carbon skeleton
    • C07C323/51Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and carboxyl groups bound to the same carbon skeleton having the sulfur atoms of the thio groups bound to acyclic carbon atoms of the carbon skeleton
    • C07C323/56Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and carboxyl groups bound to the same carbon skeleton having the sulfur atoms of the thio groups bound to acyclic carbon atoms of the carbon skeleton the carbon skeleton containing six-membered aromatic rings
    • C07C2101/04
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2601/00Systems containing only non-condensed rings
    • C07C2601/04Systems containing only non-condensed rings with a four-membered ring

Definitions

  • the present invention relates to lipid compounds of the general formula (I):
  • the invention also relates to pharmaceutical compositions and lipid compositions comprising such compounds, and to such compounds for use as medicaments or for use in therapy, in particular for the treatment of diseases related to the cardiovascular, metabolic and inflammatory disease area.
  • dietary polyunsaturated fatty acids have been shown to regulate plasma lipid levels, cardiovascular and immune functions, insulin action, and neuronal development and visual function.
  • Tetradecylthioacetic acid is a modified fatty acid which has a number of powerful effects demonstrable both in-vivo and in-vitro.
  • TTA has properties very similar to natural fatty acids, the main difference being that it cannot be oxidised by the mitochondrial ⁇ -oxidation, but significantly increases the oxidation of other fatty acids. Despite the fact that TTA is not able to undergo ⁇ -oxidation, it is metabolised in most ways as a normal saturated fatty acid.
  • TTA affects oxidative status at different levels by having the potential of changing the antioxidant defense system, in addition to being an antioxidant itself through its free radical scavenging capacity.
  • TTA may prevent the oxidative modification of low-density lipoprotein (LDL) particles in plasma and reduce the generation of lipid peroxides.
  • LDL low-density lipoprotein
  • novel fatty acid derivatives represented by the general formula (I) have higher affinities for the receptors PPAR ⁇ and PPAR ⁇ compared to TTA and (all-Z)-3-thia-6,9,12,15-octadecatetraenoic acid.
  • Fatty acid derivatives represented by the general formula (I) also reduced triglyceride, cholesterol and free fatty acids levels in a dyslipidemic mice model to a greater extent than TTA and (all-Z)-3-thia-6,9,12,15-octadecatetraenoic acid.
  • FIG. 1 Results of PPAR activation in PPAR ⁇ , PPAR ⁇ , and PPAR ⁇ luciferase reporter cell lines by compounds according to the present disclosure compared to PPAR ⁇ , PPAR ⁇ , and PPAR ⁇ activity of GW7647, L-165041, and BRL49653, respectively.
  • FIG. 2 Plasma triglyceride levels and plasma cholesterol levels in APOE*3Leiden mice after administration of compounds according to the present disclosure and unsubstituted reference substances.
  • FIG. 3 Plasma glucose levels in ob/ob mice after administration of 2-((5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaenylthio)butanoic acid, pioglitazone, and a placebo.
  • FIG. 4 Plasma insulin levels in ob/ob mice after administration of 2-((5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaenylthio)butanoic acid, pioglitazone, and a placebo.
  • FIG. 5 Whole blood HbA1c levels in ob/ob mice after administration of 2-((5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaenylthio)butanoic acid, pioglitazone, and a placebo.
  • FIG. 6 Body weight differences in ob/ob mice after administration of 2-((5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaenylthio)butanoic acid, pioglitazone, and a placebo.
  • One object of the present invention is to provide lipid compounds having improved biological activity compared to 3-thia fatty acids. This object is achieved by a lipid compound of formula (I)
  • R 1 is selected from a C 10 -C 22 alkyl, a C 10 -C 22 alkenyl having 1-6 double bonds, and a C 10 -C 22 alkynyl having 1-6 triple bonds;
  • said alkyl group may be selected from the group consisting of methyl, ethyl, n-propyl, isopropyl, n-butyl, sec.-butyl, and n-hexyl
  • said alkenyl group may be selected from the group consisting of allyl, 2-butenyl, and 3-hexenyl
  • said alkynyl group may be selected from the group consisting of propargyl, 2-butynyl, and 3-hexynyl
  • said halogen atom may be selected from the group consisting of fluorine, chlorine, bromine, and iodine
  • said alkoxy group may be selected from the group consisting of methoxy, ethoxy, propoxy, isopropoxy, sec.-butoxy, phenoxy, benzyloxy, OCH 2 CF 3 , and OCH 2 CH 2 OCH 3
  • said acyloxy group may be selected from acetoxy, propionoxy, and
  • one of the substituents R 2 and R 3 of the compound of formula (I) is hydrogen and the other one is selected from a group of substituents consisting of a hydroxy group, an alkyl group, a halogen atom, an alkoxy group, an acyloxy group, an acyl group, an alkenyl group, an alkynyl group, an aryl group, an alkylthio group, an alkoxycarbonyl group, a carboxy group, an alkylsulfinyl group, an alkylsulfonyl group, an amino group, and an alkylamino group.
  • R 2 and R 3 are independently selected from a hydrogen atom, an alkyl group, an alkoxy group or an aryl group; or R 2 and R 3 can be connected in order to form a cycloalkane.
  • R 2 and R 3 are independently selected from a hydrogen atom, an alkyl group, or a methoxy group or an ethoxy group.
  • R 2 and R 3 are independently selected from a hydrogen atom, an ethyl, methoxy or ethoxy group, phenyl; or R 2 and R 3 are connected to form a cyclobutane group.
  • the substituents R 2 and R 3 of the compound of formula (I) are the same or different and may be selected from a group of substituents consisting of a hydroxy group, an alkyl group, a halogen atom, an alkoxy group, an acyloxy group, an acyl group, an alkenyl group, an alkynyl group, an aryl group, an alkylthio group, an alkoxycarbonyl group, a carboxy group, an alkylsulfinyl group, an alkylsulfonyl group, an amino group.
  • R 2 and R 3 are alkyl groups selected from methyl, ethyl, n-propyl, or isopropyl, more preferably selected from methyl or ethyl, and most preferably R 2 and R 3 are ethyl.
  • the substituent R 1 of the compound of formula (I) is a C 10 -C 22 alkyl, and the said compound is derived from a saturated fatty acid.
  • the substituents R 2 and R 3 of the compound of formula (I) are the same or different and may be selected from a group of substituents as mentioned above, and the substituent R 1 is a C 10 -C 22 alkyl, and the said compound is derived from a saturated fatty acid.
  • R 1 When derived from a polyunsaturated fatty acid, R 1 is typically a C 10 -C 22 alkenyl with 2-6 double bonds, e.g 3-6 double bounds, e.g. 3-6 methylene interrupted double bonds in Z configuration.
  • R 1 is: a C 15 alkenyl with 4 methylene interrupted double bonds in Z-configuration a C 18 alkenyl with 3-5 double bonds, e.g.
  • a C 18 alkenyl with 5 methylene interrupted double bonds in Z configuration a C 14 -C 22 alkenyl group with at least one double bond, having Z configuration, and having the first double bond at the third carbon-carbon bond from the omega (w) end of the carbon chain a C 20 alkenyl with 5 methylene interrupted double bonds in Z-configuration a C 22 alkenyl with 6 methylene interrupted double bonds in Z-configuration.
  • R 1 may be a C 10 -C 22 alkynyl, e.g. a C 16 -C 22 alkynyl with 1-6 triple bonds.
  • the substituent Y of the compound of formula (I) is sulfur. In another embodiment of the invention, the substituent Y of the compound of formula (I) is sulfoxide. In still another embodiment of the invention, the substituent Y of the compound of formula (I) is sulfone.
  • the substituent X of the compound of formula (I) is a carboxylic acid in the form of an ester, a free acid, a triglyceride or a phospholipid.
  • the substituent X is a carboxylic acid in the form of an ester, or a free acid, and more preferably X is a carboxylic acid in the form of a free acid.
  • the substituent R 1 is a C 10 -C 22 alkyl, and the lipid compound being derived from a saturated fatty acid;
  • R 2 and R 3 are the same or different and may be selected from a group of substituents consisting of a hydroxy group, an alkyl group, a halogen atom, an alkoxy group, an acyloxy group, an acyl group, an alkenyl group, an alkynyl group, an aryl group, an alkylthio group, an alkoxycarbonyl group, a carboxy group, an alkylsulfinyl group, an alkylsulfonyl group, an amino group; preferably R 2 and R 3 are alkyl groups; and X is a carboxylic acid in the form of a free acid.
  • the invention also relates to salts of the compound of formula (I). Such salts may be represented by
  • Z 2+ is selected from the group consisting of Mg 2+ , Ca 2+ ,
  • X is COO ⁇
  • Z n+ is a polyvalent cation such as
  • the compounds of formula (I) are capable of existing in stereoisomeric forms. It will be understood that the invention encompasses all optical isomers of the compounds of formula (I) and mixtures thereof. Hence, compounds of formula (I) being present as diastereomers, racemates and enantiomers are included.
  • the present invention also relates to a lipid compound according of formula (I) for use as a medicament.
  • the present invention provides a food supplement, a food additive, or a neutraceutical preparation comprising a lipid compound of formula (I).
  • Such a food supplement may be produced for administration through any route of administration.
  • the food supplement may be administered as a liquid nutritional or as a beverage.
  • the food supplement may be in the form of a capsule, e.g. a gelatin capsule, and the capsule may be flavoured.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of formula (I), preferably together with one or more pharmaceutically acceptable carriers or excipients.
  • novel lipid compounds and compositions of the invention may be formulated in conventional oral administration forms, e.g. tablets, coated tablets, capsules, powders, granulates, solutions, dispersions, suspensions, syrups, emulsions, sprays, etc using conventional excipients, e.g.
  • solvents diluents, binders, sweeteners, aromas, pH modifiers, viscosity modifiers, antioxidants, corn starch, lactose, glucose, microcrystalline cellulose, magnesium stearate, polyvinylpyrrolidone, citric acid, tartaric acid, water, ethanol, glycerol, sorbitol, polyethylene glycol, propylene glycol, cetylstearyl alcohol, carboxymethylcellulose or fatty substances such as hard fat or suitable mixtures thereof etc.
  • Conventional formulation techniques well known in the art, may be used.
  • compositions may likewise be administered by conventional administration routes, i.e. orally.
  • administration routes i.e. orally.
  • orally administrable compositions e.g. tablets, coated tablets, capsules, syrups, etc is especially preferred.
  • a suitable daily dosage of the compound according to formula (I) is 1 mg to 10 g of said compound; 50 mg to 1 g of said compound, or 50 mg to 200 mg of said compound.
  • the pharmaceutical composition according to the invention may be used as a medicament.
  • the present invention also relates to lipid composition
  • lipid composition comprising a lipid compound according to formula (I).
  • lipid compound according to formula (I)
  • at least 60% by weight, or at least 80% by weight of the lipid composition is comprised of said compound.
  • the lipid composition may further comprise a pharmaceutically acceptable antioxidant, e.g. tocopherol.
  • a pharmaceutically acceptable antioxidant e.g. tocopherol.
  • the present invention relates to a lipid composition for use as a medicament.
  • the present invention relates to the use of a lipid compound according to formula (I) for use in:
  • the invention also relates to lipid compounds according to formula (I) for the treatment of the above mentioned conditions, and to methods for the treatment and/or prevention of the conditions listed above, comprising administering to a mammal in need thereof a pharmaceutically active amount of a compound according to formula (I).
  • the present invention encompasses methods for manufacturing lipid compounds according to formula (I).
  • the raw material may e.g. originate from a vegetable, a microbial and/or an animal source, such as a marine fish oil.
  • a marine oil or a krill oil is used.
  • lipid compound relates to fatty acid analogues derived from e.g. saturated fatty acids, monounsaturated fatty acids, polyunsaturated fatty acids and lipids comprising 1-6 triple bonds.
  • a “pharmaceutically active amount” relates to an amount that will lead to the desired pharmacological and/or therapeutic effects, i.e. an amount of the combination product which is effective to achieve its intended purpose. While individual patient needs may vary, determination of optimal ranges for effective amounts of the combination product is within the skill of the art. Generally, the dosage regimen for treating a condition with the combination product of this invention is selected in accordance with a variety of factors, including the type, age, weight, sex, diet and medical condition of the patient.
  • a pharmaceutical composition is meant a lipid compound according to the invention in any form suitable to be used for a medical purpose.
  • Treatment includes any therapeutic application that can benefit a human or non-human mammal. Both human and veterinary treatments are within the scope of the present invention. Treatment may be in respect of an existing condition or it may be prophylactic.
  • Fatty acids are straight chain hydrocarbons possessing a carboxyl (COOH) group at one end ( ⁇ ) and (usually) a methyl group at the other ( ⁇ ) end. In chemistry, the numbering of the carbon atoms starts from the ⁇ end.
  • the ⁇ carbon refers to the first carbon after the carbon that attaches to the functional group, and the second carbon is the ⁇ carbon.
  • methylene interrupted double bonds relates to the case when a methylene group is located between to separate double bonds in a carbon chain of a lipid compound.
  • the inventors have surprisingly found that the following lipid compound shown in categories A-E, are particularly preferable.
  • lipid compounds according to the invention are:
  • the compounds of categories A-E above were R 2 and R 3 are different, are capable of existing in stereoisomeric forms, i.e. all optical isomers of the compounds and mixtures thereof are encompassed. Hence, the said compounds may be present as diastereomers, racemates and enantiomers.
  • the alcohols described in method I and II may be prepared directly from the carboxylic esters of, for example, naturally occurring fatty acids; e.g. alpha-linolenic acid, conjugated linoleic acid, eicosapentaenoic acid (EPA), etc. by reduction with a reducing agent like lithium aluminiumhydride or diisobultylaluminiumhydride at ⁇ 10 to 0° C.
  • the alcohols can also be prepared by degradation of the polyunsaturated fatty acids EPA and DHA, as described by Holmeide et al. ( J. Chem. Soc., Perkin Trans. 1, 2000, 2271). In this case one can start with purified EPA or DHA, but it is also possible to start with fish oil containing EPA and DHA in mixture.
  • the leaving group (LG) present in compounds of formula (XI) may, for example, be mesylate, tosylate or a suitable halogen, such as bromine.
  • the resulting alcohols can be converted, using functional group interconversion, by methods familiar to persons skilled in the art (step I), to compounds where the terminal hydroxy group have been transformed into a suitable leaving group (LG).
  • Suitable leaving groups include bromine, mesylate and tosylate.
  • the alcohols can be converted to the corresponding thiols (step IV) by methods familiar to persons skilled in the art.
  • the thiols can then be reacted further (step V) in a substitution reaction with compounds of formula (XI), in the presence of base in an appropriate solvent system.
  • the corresponding sulfoxides and sulfones can be prepared by oxidation of the thioethers (Y ⁇ S) with a suitable oxidising agent (step III).
  • suitable oxidising agents are m-chloro-perbenzoic acid (MCPBA), hydrogen peroxide (H 2 O 2 ) and oxone (potassium peroxymonosulfate).
  • an esterifying group such as a methyl of an ethyl group may be removed, for example, by alkaline hydrolysis using a base such as an alkali metal hydroxide, for example LiOH, NaOH or KOH or by using an organic base, for example Et 3 N together with an inorganic salt, for example LiCl in an appropriate solvent system.
  • a tert-butyl group may be removed, for example, by treatment with an acid, for example an organic acid such as trifluoroacetic acid or formic acid in an appropriate solvent system.
  • An arylmethyl group such as a benzyl group may be removed, for example, by hydrogenation over a catalyst such as palladium-on-carbon in an appropriate solvent system.
  • the compounds of formula (I) wherein X is a carboxylic acid and in the form of a phospholipid can be prepared through the following processes.
  • Sn-Glycero-3-phosphocholine, as cadmium (II) adduct can also be reacted with the imidazolide activated fatty acid in the presence of DBU (1,8-diazabicyclo[5.4.0]undec-7-ene) to prepare the phosphatidylcholine of the respective fatty acid (International application number PCT/GB2003/002582).
  • Enzymatic transphosphatidylation can effect the transformation of phosphatidylcholine to phosphatidyletanolamine (Wang et al, J. Am. Chem. Soc., 1993, 115, 10487).
  • Phospholipids may also be prepared by enzymatic esterification and transesterification of phospholipids or enzymatic transphosphatidylation of phospholipids. (Hosokawa, J. Am. Oil Chem. Soc. 1995, 1287, Lilja-Hallberg, Biocatalysis, 1994, 195).
  • the compounds of formula (I) wherein X is a carboxylic acid in the form of a triglyceride can be prepared through the following process. Excess of the fatty acid can be coupled to glycerol using dimethylaminopyridine (DMAP) and 2-(1H-benzotriazol-1-yl)-N,N,N′,N′-tetramethyluroniumhexafluorophosphate (HBTU).
  • DMAP dimethylaminopyridine
  • HBTU 2-(1H-benzotriazol-1-yl)-N,N,N′,N′-tetramethyluroniumhexafluorophosphate
  • the compounds of formula (I) wherein X is a carboxylic acid in the form of a diglyceride can be prepared by reaction of the fatty acid (2 equivalents) with glycerol (1 equivalent) in the presence of 1,3-dicyclohexylcarbondiimide (DCC) and 4-dimethylaminopyridine (DMAP).
  • DCC 1,3-dicyclohexylcarbondiimide
  • DMAP 4-dimethylaminopyridine
  • the compounds of formula (I) wherein X is a carboxylic acid and in the form of a monoglyceride can be prepared through the following processes.
  • One method utilizes esterification of the fatty acid with glycidol in the presence of 1-(3-dimethylaminopropyI)-3-ethylcarbodiimidehydrochloride (EDC) and 4-dimethylaminopyridine (DMAP) to produce a glycidyl derivative.
  • EDC 1-(3-dimethylaminopropyI)-3-ethylcarbodiimidehydrochloride
  • DMAP 4-dimethylaminopyridine
  • TFAA trifluoroacetic anhydride
  • a 1,3-regiospecific lipase from the fungus Mucor miehei can be used to produce triglycerides or diglycerides from polyunsaturated fatty acids and glycerol.
  • a different lipase, the non-regiospecific yeast lipase from Candida antartica is highly efficient in generating triglycerides from polyunsaturated fatty acids (Haraldsson, Pharmazie, 2000, 3).
  • Triphenylphosphine (PPh 3 ) (41.7 g, 159 mmol) was dissolved in dry tetrahydrofurane (THF) (250 mL) at 0° C. under inert atmosphere and added diisopropyl azodicarboxylate (DIAD) (30.8 mL, 159 mmol). The mixture was stirred at 0° C. for 40 minutes and then dropwise added a solution of (all-Z)-3,6,9,12-pentadecatetraenol (17.5 g, 79.4 mmol) and thioacetic acid (11.4 mL, 159 mmol) in dry THF (150 mL).
  • THF dry tetrahydrofurane
  • DIAD diisopropyl azodicarboxylate
  • Triphenylphosphine (21.0 g, 80 mmol) was dissolved in dry THF (170 mL) at 0° C. under inert atmosphere and added DIAD (15.8 mL, 80 mmol) dropwise. After 40 minutes at 0° C. the white suspension was added dropwise to a solution of (5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaen-1-ol (11.5 g, 40 mmol) and thioacetic acid (5.7 mL, 80 mmol) in dry THF (50 mL) during 15 minutes. The resulting turbid mixture was stirred at 0° C. for 30 minutes, followed by ambient temperature for 1.5 hour.
  • Ethyl 2-((3Z,6Z,9Z,12Z)-pentadeca-3,6,9,12-tetraenylthio)butanoate (2.7 g, 7.7 mmol) was dissolved in dry CHCl 3 (40 mL) and the solution was cooled down to ⁇ 20° C. under inert atmosphere.
  • meta-Chloroperoxybenzoic acid (mCPBA) ( ⁇ 77%, 4.0 g, 18 mmol) dissolved in dry CHCl 3 (10 mL) was added dropwise and the resulting solution was stirred at ⁇ 20° C. for 30 minutes, allowed to slowly reach ambient temperature and then stirred over night.
  • the assay was carried out in-vitro in three stable reporter cell lines, PPAR ⁇ , PPAR ⁇ or PPAR ⁇ , expressing respectively a chimeric protein containing the ligand binding domain (LBD) of human PPAR ⁇ , human PPAR ⁇ or human PPAR ⁇ fused to the yeast transactivator GAL4 DNA binding domain (DBD).
  • LBD ligand binding domain
  • DBD yeast transactivator GAL4 DNA binding domain
  • the luciferase (Luc) reporter gene is driven by a pentamer of the GAL4 recognition sequence in front of a 3-globin promoter.
  • the use of GAL4-PPAR ⁇ , GAL4-PPAR ⁇ and GAL4-PPAR ⁇ chimeric receptors allows for elimination of background activity from endogenous receptors and quantitation of relative activity across the three PPAR subtypes with the same reporter gene.
  • the PPAR selectivity of the substances was determined by comparison to known drug references (1 ⁇ M GW7647 for PPAR ⁇ , 1 ⁇ M L-165041 for PPAR ⁇ and 1 ⁇ M BRL49653 for PPAR ⁇ ) set of 100% activity.
  • the assay was carried out in-vitro using mammalian-one-hybrid assays (M1H) comprising GAL4-DNA binding domain-PPAR ⁇ -LBD fusion constructs in conjunction with 5 ⁇ GAL4-sites driven Photinus pyralis luciferase reporter construct in transiently transfected HEK293 cells.
  • M1H mammalian-one-hybrid assays
  • This animal model has proven to be representative for the human situation regarding plasma lipoprotein levels, lipoprotein profiles, its responsiveness to hypolipidemic drugs (like statins, fibrates etc.) and nutrition.
  • hypolipidemic drugs like statins, fibrates etc.
  • APOE*3Leiden mice develop atherosclerotic lesions in the aorta resembling those found in humans with respect to cellular composition and morphological and immunohistochemical characteristics.
  • test substances were administered orally as admix to the Western-type diet.
  • sunflower oil was added to a total oil volume of 10 mL/kg diet.
  • mice were fasted overnight (o/n) and blood samples were taken to measure plasma ketone bodies and free fatty acids.
  • blood samples were taken after a 4 hour-fast period to measure plasma cholesterol and triglycerides.
  • Ob/ob mice can be used as a model for type II diabetes.
  • the mice are homozygous for the obese spontaneous mutation (Lee) leading to leptin deficiency.
  • Lee obese spontaneous mutation
  • ob/ob mice may reach three times the normal body weight of wild type controls
  • ob/ob mice exhibit a diabetes type II-like syndrome of hyperglycemia, glucose intolerance, elevated plasma insulin, infertility, impaired wound healing, and an increase in hormone production from both pituitary and adrenal glands.
  • All compounds were administered orally as admix to AM II diet.
  • sunflower oil was added to a total oil volume of 10 ml/kg diet.

Abstract

The present disclosure relates to lipid compounds of the general formula (I): (I) wherein R1 is selected from a C10-C22 alkyl, a C10-C22 alkenyl having 1-6 double bonds, and a C10-C22 alkynyl having 1-6 triple bonds; R2 and R3 are the same or different substituents; Y is selected from sulphur, sulfoxide, and sulfone; and X represents a carboxylic acid or a derivative thereof, a carboxylic ester, a carboxylic anhydride or a carboxamide; or a pharmaceutically acceptable salt, complex or solvate thereof. The disclosure also relates to pharmaceutical compositions and lipid compositions comprising such compounds, and to such compounds for use as medicaments or for use in therapy, in particular for the treatment of diseases related to the cardiovascular, metabolic and inflammatory disease area.

Description

  • This application is a continuation of U.S. application Ser. No. 14/263,793, filed Apr. 28, 2014, a continuation of U.S. application Ser. No. 13/054,212, filed Apr. 13, 2011, now U.S. Pat. No. 8,759,558, which is the U.S. national stage application of International Application No. PCT/NO2009/000262, filed Jul. 13, 2009, which claims benefit of priority of U.S. Provisional Application No. 61/080,804 and European Application No. 08160450.6, which were filed Jul. 15, 2008, all of which are incorporated herein by reference.
  • TECHNICAL FIELD
  • The present invention relates to lipid compounds of the general formula (I):
  • Figure US20170029368A1-20170202-C00001
  • wherein
      • R1 is selected from a C10-C22 alkyl, a C10-C22 alkenyl having 1-6 double bonds, and a C10-C22 alkynyl having 1-6 triple bonds;
      • R2 and R3 are the same or different and may be selected from a group of substituents consisting of a hydrogen atom, a hydroxy group, an alkyl group, a halogen atom, an alkoxy group, an acyloxy group, an acyl group, an alkenyl group, an alkynyl group, an aryl group, an alkylthio group, an alkoxycarbonyl group, a carboxy group, an alkylsulfinyl group, an alkylsulfonyl group, an amino group, and an alkylamino group, provided that R2 and R3 cannot both be a hydrogen atom; or
      • R2 and R3 can be connected in order to form a cycloalkane like cyclopropane, cyclobutane, cyclopentane or cyclohexane,
      • Y is selected from sulphur, sulfoxide, and sulfone,
      • X represents a carboxylic acid or a derivative thereof, a carboxylic ester or a carboxamide, or a pharmaceutically acceptable salt, solvate, solvate of such salt or a prodrug thereof.
  • In those cases were R2 and R3 are different, the compounds of formula (I) are capable of existing in stereoisomeric forms. It will be understood that the invention encompasses all optical isomers of the compounds of formula (I) and mixtures thereof.
  • The invention also relates to pharmaceutical compositions and lipid compositions comprising such compounds, and to such compounds for use as medicaments or for use in therapy, in particular for the treatment of diseases related to the cardiovascular, metabolic and inflammatory disease area.
  • BACKGROUND OF THE INVENTION
  • Up to date, there has been a lot of research on fatty acid analogues and their effects on diverse physiological processes impacting normal health and chronic diseases.
  • For example, dietary polyunsaturated fatty acids (PUFAs) have been shown to regulate plasma lipid levels, cardiovascular and immune functions, insulin action, and neuronal development and visual function.
  • Tetradecylthioacetic acid (TTA) is a modified fatty acid which has a number of powerful effects demonstrable both in-vivo and in-vitro.
  • TTA has properties very similar to natural fatty acids, the main difference being that it cannot be oxidised by the mitochondrial β-oxidation, but significantly increases the oxidation of other fatty acids. Despite the fact that TTA is not able to undergo β-oxidation, it is metabolised in most ways as a normal saturated fatty acid.
  • Figure US20170029368A1-20170202-C00002
  • TTA affects oxidative status at different levels by having the potential of changing the antioxidant defense system, in addition to being an antioxidant itself through its free radical scavenging capacity.
  • Addition of TTA may prevent the oxidative modification of low-density lipoprotein (LDL) particles in plasma and reduce the generation of lipid peroxides.
  • Several polyunsaturated fatty acid derivatives with sulfur in 3-position have been prepared (Flock et al, Acta Chemica Scand., 1999, 53, 436). Methyl (all-Z)-3-thia-6,9,12,15-octadecatetraenoate was tested in a Wistar rat model, and the effects were compared to the effects of TTA. The results suggest that both the saturated and the unsaturated fatty acids lowered plasma triglycerides to a similar extent (Willumsen et al, J. Lipid Mediators Cell Signaling, 1997, 17, 115)
  • It has surprisingly been found that novel fatty acid derivatives represented by the general formula (I) have higher affinities for the receptors PPARα and PPARγ compared to TTA and (all-Z)-3-thia-6,9,12,15-octadecatetraenoic acid. Fatty acid derivatives represented by the general formula (I) also reduced triglyceride, cholesterol and free fatty acids levels in a dyslipidemic mice model to a greater extent than TTA and (all-Z)-3-thia-6,9,12,15-octadecatetraenoic acid.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1: Results of PPAR activation in PPARα, PPARδ, and PPARγ luciferase reporter cell lines by compounds according to the present disclosure compared to PPARα, PPARδ, and PPARγ activity of GW7647, L-165041, and BRL49653, respectively.
  • FIG. 2: Plasma triglyceride levels and plasma cholesterol levels in APOE*3Leiden mice after administration of compounds according to the present disclosure and unsubstituted reference substances.
  • FIG. 3: Plasma glucose levels in ob/ob mice after administration of 2-((5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaenylthio)butanoic acid, pioglitazone, and a placebo.
  • FIG. 4: Plasma insulin levels in ob/ob mice after administration of 2-((5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaenylthio)butanoic acid, pioglitazone, and a placebo.
  • FIG. 5: Whole blood HbA1c levels in ob/ob mice after administration of 2-((5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaenylthio)butanoic acid, pioglitazone, and a placebo.
  • FIG. 6: Body weight differences in ob/ob mice after administration of 2-((5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaenylthio)butanoic acid, pioglitazone, and a placebo.
  • SUMMARY OF THE INVENTION
  • One object of the present invention is to provide lipid compounds having improved biological activity compared to 3-thia fatty acids. This object is achieved by a lipid compound of formula (I)
  • Figure US20170029368A1-20170202-C00003
  • In particular, the present invention relates to compounds of formula (I), wherein:
  • R1 is selected from a C10-C22 alkyl, a C10-C22 alkenyl having 1-6 double bonds, and a C10-C22 alkynyl having 1-6 triple bonds;
      • R2 and R3 are the same or different and may be selected from a group of substituents consisting of a hydrogen atom, a hydroxy group, an alkyl group, a halogen atom, an alkoxy group, an acyloxy group, an acyl group, an alkenyl group, an alkynyl group, an aryl group, an alkylthio group, an alkoxycarbonyl group, a carboxy group, an alkylsulfinyl group, an alkylsulfonyl group, an amino group, and an alkylamino group, provided that R2 and R3 cannot both be a hydrogen atom, or
      • R2 and R3 can be connected in order to form a cycloalkane like cyclopropane, cyclobutane, cyclopentane or cyclohexane,
      • Y is selected from sulphur, sulfoxide, and sulfone,
      • X represents a carboxylic acid or a derivative thereof, a carboxylic ester or a carboxamide,
        or a pharmaceutically acceptable salt, solvate, solvate of such salt or a prodrug thereof.
  • In a compound according to the invention, said alkyl group may be selected from the group consisting of methyl, ethyl, n-propyl, isopropyl, n-butyl, sec.-butyl, and n-hexyl, said alkenyl group may be selected from the group consisting of allyl, 2-butenyl, and 3-hexenyl, said alkynyl group may be selected from the group consisting of propargyl, 2-butynyl, and 3-hexynyl, said halogen atom may be selected from the group consisting of fluorine, chlorine, bromine, and iodine; said alkoxy group may be selected from the group consisting of methoxy, ethoxy, propoxy, isopropoxy, sec.-butoxy, phenoxy, benzyloxy, OCH2CF3, and OCH2CH2OCH3, said acyloxy group may be selected from acetoxy, propionoxy, and butyroxy, said aryl group is a phenyl group; said alkylthio group may be selected from the group consisting of methylthio, ethylthio, isopropylthio, and phenylthio, said alkoxycarbonyl group may be selected from the group consisting of methoxycarbonyl, ethoxycarbonyl, propoxycarbonyl, and butoxycarbonyl, said alkylsulfinyl group may be selected from the group consisting of methanesulfinyl, ethanesulfinyl, and isopropanesulfinyl, said alkylsulfonyl group may be selected from the group consisting of methanesulfonyl, ethanesulfonyl, and isopropanesulfonyl, said alkylamino group may be selected from the group consisting of methylamino, dimethylamino, ethylamino, and diethylamino, said carboxylate group may be selected from the group consisting of ethyl carboxylate, methyl carboxylate, n-propyl carboxylate, isopropyl carboxylate, n-butyl carboxylate, sec.-butyl carboxylate, and n-hexyl carboxylate, said carboxamide group may be selected from the group consisting of carboxamide such as N-methyl carboxamide, N,N-dimethyl carboxamide, N-ethyl carboxamide and N,N-diethyl carboxamide.
  • In one embodiment of the invention, one of the substituents R2 and R3 of the compound of formula (I) is hydrogen and the other one is selected from a group of substituents consisting of a hydroxy group, an alkyl group, a halogen atom, an alkoxy group, an acyloxy group, an acyl group, an alkenyl group, an alkynyl group, an aryl group, an alkylthio group, an alkoxycarbonyl group, a carboxy group, an alkylsulfinyl group, an alkylsulfonyl group, an amino group, and an alkylamino group.
  • In a preferred embodiment R2 and R3 are independently selected from a hydrogen atom, an alkyl group, an alkoxy group or an aryl group; or R2 and R3 can be connected in order to form a cycloalkane.
  • In another preferred embodiment R2 and R3 are independently selected from a hydrogen atom, an alkyl group, or a methoxy group or an ethoxy group.
  • In yet another preferred embodiment R2 and R3 are independently selected from a hydrogen atom, an ethyl, methoxy or ethoxy group, phenyl; or R2 and R3 are connected to form a cyclobutane group.
  • In another embodiment of the invention, the substituents R2 and R3 of the compound of formula (I) are the same or different and may be selected from a group of substituents consisting of a hydroxy group, an alkyl group, a halogen atom, an alkoxy group, an acyloxy group, an acyl group, an alkenyl group, an alkynyl group, an aryl group, an alkylthio group, an alkoxycarbonyl group, a carboxy group, an alkylsulfinyl group, an alkylsulfonyl group, an amino group. Preferably R2 and R3 are alkyl groups selected from methyl, ethyl, n-propyl, or isopropyl, more preferably selected from methyl or ethyl, and most preferably R2 and R3 are ethyl.
  • In one embodiment of the invention the substituent R1 of the compound of formula (I) is a C10-C22 alkyl, and the said compound is derived from a saturated fatty acid.
  • Preferably, the substituents R2 and R3 of the compound of formula (I) are the same or different and may be selected from a group of substituents as mentioned above, and the substituent R1 is a C10-C22 alkyl, and the said compound is derived from a saturated fatty acid.
  • When derived from a polyunsaturated fatty acid, R1 is typically a C10-C22 alkenyl with 2-6 double bonds, e.g 3-6 double bounds, e.g. 3-6 methylene interrupted double bonds in Z configuration. For example, R1 is: a C15 alkenyl with 4 methylene interrupted double bonds in Z-configuration a C18 alkenyl with 3-5 double bonds, e.g. a C18 alkenyl with 5 methylene interrupted double bonds in Z configuration a C14 -C 22 alkenyl group with at least one double bond, having Z configuration, and having the first double bond at the third carbon-carbon bond from the omega (w) end of the carbon chain a C20 alkenyl with 5 methylene interrupted double bonds in Z-configuration a C22 alkenyl with 6 methylene interrupted double bonds in Z-configuration.
  • Furthermore, R1 may be a C10-C22 alkynyl, e.g. a C16 -C 22 alkynyl with 1-6 triple bonds.
  • In one embodiment of the invention, the substituent Y of the compound of formula (I) is sulfur. In another embodiment of the invention, the substituent Y of the compound of formula (I) is sulfoxide. In still another embodiment of the invention, the substituent Y of the compound of formula (I) is sulfone.
  • In one embodiment of the invention, the substituent X of the compound of formula (I) is a carboxylic acid in the form of an ester, a free acid, a triglyceride or a phospholipid.
  • Preferably, the substituent X is a carboxylic acid in the form of an ester, or a free acid, and more preferably X is a carboxylic acid in the form of a free acid.
  • In another embodiment of the invention, the substituent R1 is a C10-C22 alkyl, and the lipid compound being derived from a saturated fatty acid; R2 and R3 are the same or different and may be selected from a group of substituents consisting of a hydroxy group, an alkyl group, a halogen atom, an alkoxy group, an acyloxy group, an acyl group, an alkenyl group, an alkynyl group, an aryl group, an alkylthio group, an alkoxycarbonyl group, a carboxy group, an alkylsulfinyl group, an alkylsulfonyl group, an amino group; preferably R2 and R3 are alkyl groups; and X is a carboxylic acid in the form of a free acid.
  • The invention also relates to salts of the compound of formula (I). Such salts may be represented by
  • Figure US20170029368A1-20170202-C00004
  • wherein X is COO,
    • Z+ is selected from the group consisting of Li+, Na+, K+, NH4 +,
  • Figure US20170029368A1-20170202-C00005
  • wherein X=COO, Z2+ is selected from the group consisting of Mg2+, Ca2+,
  • Figure US20170029368A1-20170202-C00006
  • Another representative salt is
  • Figure US20170029368A1-20170202-C00007
  • wherein X is COO, Zn+ is a polyvalent cation such as
  • Figure US20170029368A1-20170202-C00008
  • In the case the compounds of formula (I) is in the form of a phospholipid, such compounds may be represented by the following formulas (II-IV),
  • Figure US20170029368A1-20170202-C00009
  • wherein Z is:
  • Figure US20170029368A1-20170202-C00010
  • wherein Z is:
  • Figure US20170029368A1-20170202-C00011
  • wherein Z is:
  • Figure US20170029368A1-20170202-C00012
  • Compounds of formula (I), wherein X is a carboxylic acid in the form of a triglyceride, a 1,2-diglyceride, a 1,3 diglyceride, a 1-monoglyceride and a 2-monoglyceride, are also included in the present invention. These are hereinafter represented by the formulas (V), (VI), (VII), (VIII) and (IX), respectively.
  • Figure US20170029368A1-20170202-C00013
  • The compounds of formula (I) are capable of existing in stereoisomeric forms. It will be understood that the invention encompasses all optical isomers of the compounds of formula (I) and mixtures thereof. Hence, compounds of formula (I) being present as diastereomers, racemates and enantiomers are included.
  • In a preferred embodiment of the invention the compound of formula (I) is
  • Figure US20170029368A1-20170202-C00014
  • ethyl 2-((5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaenylthio)butanoate.
  • In another preferred embodiment of the invention the compound of formula (I) is
  • Figure US20170029368A1-20170202-C00015
  • ethyl 1-((5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaenylthio)cyclobutanecarboxylate.
  • The present invention also relates to a lipid compound according of formula (I) for use as a medicament.
  • In a further aspect, the present invention provides a food supplement, a food additive, or a neutraceutical preparation comprising a lipid compound of formula (I).
  • Such a food supplement may be produced for administration through any route of administration. For example, the food supplement may be administered as a liquid nutritional or as a beverage.
  • The food supplement may be in the form of a capsule, e.g. a gelatin capsule, and the capsule may be flavoured.
  • In still a further aspect, the present invention provides a pharmaceutical composition comprising a compound of formula (I), preferably together with one or more pharmaceutically acceptable carriers or excipients.
  • The novel lipid compounds and compositions of the invention may be formulated in conventional oral administration forms, e.g. tablets, coated tablets, capsules, powders, granulates, solutions, dispersions, suspensions, syrups, emulsions, sprays, etc using conventional excipients, e.g. solvents, diluents, binders, sweeteners, aromas, pH modifiers, viscosity modifiers, antioxidants, corn starch, lactose, glucose, microcrystalline cellulose, magnesium stearate, polyvinylpyrrolidone, citric acid, tartaric acid, water, ethanol, glycerol, sorbitol, polyethylene glycol, propylene glycol, cetylstearyl alcohol, carboxymethylcellulose or fatty substances such as hard fat or suitable mixtures thereof etc. Conventional formulation techniques, well known in the art, may be used.
  • The compositions may likewise be administered by conventional administration routes, i.e. orally. The use of orally administrable compositions, e.g. tablets, coated tablets, capsules, syrups, etc is especially preferred.
  • A suitable daily dosage of the compound according to formula (I) is 1 mg to 10 g of said compound; 50 mg to 1 g of said compound, or 50 mg to 200 mg of said compound.
  • The pharmaceutical composition according to the invention may be used as a medicament.
  • The present invention also relates to lipid composition comprising a lipid compound according to formula (I). Suitably, at least 60% by weight, or at least 80% by weight of the lipid composition is comprised of said compound.
  • The lipid composition may further comprise a pharmaceutically acceptable antioxidant, e.g. tocopherol.
  • Further, the present invention relates to a lipid composition for use as a medicament.
  • Additionally, the present invention relates to the use of a lipid compound according to formula (I) for use in:
      • activation or modulation of at least one of the human peroxisome proliferator-activated receptor (PPAR) isoforms α, γ or δ, wherein said compound e.g. is a pan-agonist or modulator
      • the prevention and/or treatment of a dyslipidemic condition, e.g. hypertriglyceridemia (HTG)
      • the prevention and/or treatment of elevated triglyceride levels, LDL cholesterol levels, and/or VLDL cholesterol levels
      • the treatment and/or the prevention of obesity or an overweight condition
      • the reduction of body weight and/or for preventing body weight gain
      • the treatment and/or the prevention of a fatty liver disease, e.g. non-alcoholic fatty liver disease (NAFLD).
      • the treatment and/or the prevention of atherosclerosis
      • the prevention of myocardial infarction
      • the treatment and/or the prevention of peripheral insulin resistance and/or a diabetic condition
      • the treatment and/or prevention of type 2 diabetes
      • the reduction of plasma insulin, blood glucose and/or serum triglycerides
      • the treatment and/or the prevention of an inflammatory disease or condition.
  • The invention also relates to lipid compounds according to formula (I) for the treatment of the above mentioned conditions, and to methods for the treatment and/or prevention of the conditions listed above, comprising administering to a mammal in need thereof a pharmaceutically active amount of a compound according to formula (I).
  • In addition, the present invention encompasses methods for manufacturing lipid compounds according to formula (I). The raw material may e.g. originate from a vegetable, a microbial and/or an animal source, such as a marine fish oil. Preferably a marine oil or a krill oil is used.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present inventors have found that compounds of formula (I) as presented above, have remarkably good pharmaceutical activity.
  • As used herein, the term “lipid compound” relates to fatty acid analogues derived from e.g. saturated fatty acids, monounsaturated fatty acids, polyunsaturated fatty acids and lipids comprising 1-6 triple bonds.
  • A “pharmaceutically active amount” relates to an amount that will lead to the desired pharmacological and/or therapeutic effects, i.e. an amount of the combination product which is effective to achieve its intended purpose. While individual patient needs may vary, determination of optimal ranges for effective amounts of the combination product is within the skill of the art. Generally, the dosage regimen for treating a condition with the combination product of this invention is selected in accordance with a variety of factors, including the type, age, weight, sex, diet and medical condition of the patient.
  • By “a pharmaceutical composition” is meant a lipid compound according to the invention in any form suitable to be used for a medical purpose.
  • “Treatment” includes any therapeutic application that can benefit a human or non-human mammal. Both human and veterinary treatments are within the scope of the present invention. Treatment may be in respect of an existing condition or it may be prophylactic.
  • Nomenclature and Terminology
  • Fatty acids are straight chain hydrocarbons possessing a carboxyl (COOH) group at one end (α) and (usually) a methyl group at the other (ω) end. In chemistry, the numbering of the carbon atoms starts from the α end.
  • Figure US20170029368A1-20170202-C00016
  • The α carbon refers to the first carbon after the carbon that attaches to the functional group, and the second carbon is the β carbon.
  • As used herein, the expression “methylene interrupted double bonds” relates to the case when a methylene group is located between to separate double bonds in a carbon chain of a lipid compound.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The inventors have surprisingly found that the following lipid compound shown in categories A-E, are particularly preferable.
  • Category A
      • derived from saturated fatty acids
      • R1 is a C10-C22 alkyl
      • X represents a carboxylic acid or a derivative thereof, a carboxylic ester or a carboxamide
    EXAMPLE i
    • R1═C14, Y═S
  • Figure US20170029368A1-20170202-C00017
  • Category B
      • derived from monounsaturated fatty acids
      • R1 is a C10-C22 alkenyl having 1 double bond
      • X represents a carboxylic acid or a derivative thereof, a carboxylic ester or a carboxamide
    EXAMPLE ii
    • R1═C18, Y═S
  • Figure US20170029368A1-20170202-C00018
  • EXAMPLE iii
    • R1═C14, Y═S
  • Figure US20170029368A1-20170202-C00019
  • Category C
      • derived from polyunsaturated fatty acids
      • R1 is a C10-C22 alkenyl having 2-6 double bonds
      • X represents a carboxylic acid or a derivative thereof, a carboxylic ester or a carboxamide
    EXAMPLE iv
    • R1═C20 with 5 methylene interrupted double bonds in Z-configuration, Y═S
  • Figure US20170029368A1-20170202-C00020
  • EXAMPLE v
    • R1═C22 with 6 methylene interrupted double bonds in Z-configuration, Y═S
  • Figure US20170029368A1-20170202-C00021
  • EXAMPLE vi
    • R1═C15 with 3 methylene interrupted double bonds in Z-configuration, Y═S
  • Figure US20170029368A1-20170202-C00022
  • EXAMPLE vii
    • R1═C15 with 4 methylene interrupted double bonds in Z-configuration, Y═S
  • Figure US20170029368A1-20170202-C00023
  • EXAMPLE viii
    • R1═C15 with 3 methylene interrupted double bonds in Z-configuration and 1 double bond in E-configuration, Y═S
  • Figure US20170029368A1-20170202-C00024
  • EXAMPLE ix
    • R1═C18 with 5 methylene interrupted double bonds in Z-configuration, Y═S
  • Figure US20170029368A1-20170202-C00025
  • EXAMPLE x
    • R1═C18 with 4 methylene interrupted double bonds in Z-configuration and 1 double bond in E-configuration, Y═S
  • Figure US20170029368A1-20170202-C00026
  • Category D
      • derived from lipids containing 1-6 triple bonds
      • R1 is a C10-C22 alkynyl
      • X represents a carboxylic acid or a derivative thereof, a carboxylic ester or a carboxamide
    EXAMPLE xi
      • R1═C14 with 1 triple bond, Y═S
  • Figure US20170029368A1-20170202-C00027
  • Category E
      • R1 is selected from a C10-C22 alkyl, a C10-C22 alkenyl having 1-6 double bonds, and a C10-C22 alkynyl having 1-6 triple bonds
      • X represents a carboxylic acid or a derivative thereof, a carboxylic ester or a carboxamide
      • Y is sulfoxide or sulfone
    EXAMPLE xii
    • R1═C15 with 4 methylene interrupted double bonds in Z-configuration, Y═SO
  • Figure US20170029368A1-20170202-C00028
  • EXAMPLE xiii
    • R1═C15 with 4 methylene interrupted double bonds in Z-configuration, Y═SO2
  • Figure US20170029368A1-20170202-C00029
  • Specific examples of preferred lipid compounds according to the invention are:
  • Category A—Saturated Fatty Acids
  • Figure US20170029368A1-20170202-C00030
    • 2-(tetradecylthio)butanoic acid (1)
    • R1═C14H29, R2=ethyl, R3=a hydrogen atom, Y═S and X═COOH
  • Figure US20170029368A1-20170202-C00031
    • 2-methoxy-2-(tetradecylthio)acetic acid (2)
    • R1═C14H29, R2=methoxy, R3=a hydrogen atom, Y═S and X═COOH
  • Figure US20170029368A1-20170202-C00032
    • 2-(icosylthio)butanoic acid (3)
    • R1═C20H41, R2=ethyl, R3=a hydrogen atom, Y═S and X═COOH
  • Figure US20170029368A1-20170202-C00033
    • 2-ethyl-2-(tetradecylthio)butanoic acid (4)
    • R1═C14H29, R2═R3=ethyl, Y═S and X═COOH
    Category B—Monounsaturated Fatty Acids
  • Figure US20170029368A1-20170202-C00034
    • 2-ethyl-3-thia-12Z-heneicosaenoic acid (5)
    • R1═C18H35, R2=ethyl, R3=a hydrogen atom, Y═S and X═COOH
  • Figure US20170029368A1-20170202-C00035
    • (Z)-2-ethyl-2-(octadec-9-enylthio)butanoic acid (6)
    • R1C 181-135, R2═R3=ethyl, Y═S and X═COOH
    Category C—Polyunsaturated Fatty Acid Derivatives
  • Figure US20170029368A1-20170202-C00036
    • 2-((3Z,6Z,9Z,12Z)-pentadeca-3,6,9,12-tetraenylthio)butanoic acid (7)
    • R1═C15H23, R2=ethyl, R3=a hydrogen atom, Y═S and X═COOH
  • Figure US20170029368A1-20170202-C00037
    • 2-ethyl-2-((3Z,6Z,9Z,12Z)-pentadeca-3,6,9,12-tetraenylthio)butanoic acid (8)
    • R1═C15H23, R2═R3=ethyl, Y═S and X═COOH
  • Figure US20170029368A1-20170202-C00038
    • 2-((5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaenylthio)propanoic acid (9)
    • R1═C20H31, R2=methyl, R3=a hydrogen atom, Y═S and X═COOH
  • Figure US20170029368A1-20170202-C00039
    • 2-((5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaenylthio)butanoic acid (10)
    • R1═C20H31, R2=ethyl, R3=a hydrogen atom, Y═S and X═COOH
  • Figure US20170029368A1-20170202-C00040
    • 2-((5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaenylthio)-2-methylpropanoic acid (11)
    • R1═C20H31, R2=methyl, R3=methyl, Y═S and X═COOH
  • Figure US20170029368A1-20170202-C00041
    • 2-ethyl-2-((5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaenylthio)butanoic acid (12)
    • R1═C20H31, R2═R3=ethyl, Y═S and X═COOH
  • Figure US20170029368A1-20170202-C00042
    • 1-((5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaenylthio)cyclobutanecarboxylic acid (13)
    • R1═C20H31, R2 and R3 combines to form cyclobutane ring, Y═S and X═COOH
  • Figure US20170029368A1-20170202-C00043
    • 2-((5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaenylthio)-2-phenylacetic acid (14)
    • R1═C20H31, R2=phenyl, R3=a hydrogen atom, Y═S and X═COOH
  • Figure US20170029368A1-20170202-C00044
    • 2-((5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaenylthio)-2-methoxyacetic acid (15)
    • R1═C20H31, R2=methoxy, R3=a hydrogen atom, Y═S and X═COOH
  • Figure US20170029368A1-20170202-C00045
    • 2-((4Z,7Z,10Z,13Z,16Z,19Z)-docosa-4,7,10,13,16,19-hexaenylthio)butanoic acid (16)
    • R1═C22H33, R2=ethyl, R3=a hydrogen atom, Y═S and X═COOH
  • Figure US20170029368A1-20170202-C00046
    • 2-((4Z,7Z,10Z,13Z,16Z,19Z)-docosa-4,7,10,13,16,19-hexaenylthio)-2-ethylbutanoic acid (17)
    • R1═C22H33, R2═R3=ethyl, Y═S and X═COOH
  • Figure US20170029368A1-20170202-C00047
    • 2-((9Z,12Z,15Z)-octadeca-9,12,15-trienylthio)butanoic acid (18)
    • R1═C18H31, R2=ethyl, R3=a hydrogen atom, Y═S and X═COOH
  • Figure US20170029368A1-20170202-C00048
    • 2-ethyl-2-((9z,12Z,15Z)-octadeca-9,12,15-trienylthio)butanoic acid (19)
    • R1═C18H31, R2═R3=ethyl, Y═S and X═COOH
  • Figure US20170029368A1-20170202-C00049
    • propane-1,2,3-triyl tris(2-((5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaenylthio)butanoate) (20)
    • R1═C20H31, R2=ethyl, R3=a hydrogen atom, Y═S and X=a carboxylic acid in the form of a triglyceride
    Category D—Triple Bond Containing Fatty Acids
  • Figure US20170029368A1-20170202-C00050
    • 2-(tetradec-12-ynylthio)butanoic acid (21)
    • R1═C14H25, R2=ethyl, R3=a hydrogen atom, Y═S and X═COOH
  • Figure US20170029368A1-20170202-C00051
    • 2-ethyl-2-(tetradec-12-ynylthio)butanoic acid (22)
    • R1═C14H25, R2═R3=ethyl, Y═S and X═COOH
  • Figure US20170029368A1-20170202-C00052
    • 2-methoxy-2-(tetradec-12-ynylthio)acetic acid (23)
    • R1═C14H25, R2=methoxy, R3=a hydrogen atom, Y═S and X═COOH
    Category E—Sulfones and Sulfoxides
  • Figure US20170029368A1-20170202-C00053
    • 2-((3Z,6Z,9Z,12Z)-pentadeca-3,6,9,12-tetraenylsulfinyl)butanoic acid (24)
    • R1═C15H23, R2=ethyl, R3=a hydrogen atom, Y═SO and X═COOH
  • Figure US20170029368A1-20170202-C00054
    • 2-((3Z,6Z,9Z,12Z)-pentadeca-3,6,9,12-tetraenylsulfonyl)butanoic acid (25)
    • R1═C15H23, R2=ethyl, R3=a hydrogen atom, Y═SO2 and X═COOH
  • Figure US20170029368A1-20170202-C00055
    • 2-((5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaenylsulfinyl)butanoic acid (26)
    • R1═C20H31, R2=ethyl, R3=a hydrogen atom, Y═SO and X═COOH
  • Figure US20170029368A1-20170202-C00056
    • 2-((5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaenylsulfonyl)butanoic acid (27)
    • R1═C20H31, R2=ethyl, R3=a hydrogen atom, Y═SO2 and X═COOH
  • Figure US20170029368A1-20170202-C00057
    • 2-ethyl-2-((5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaenylsulfinyl)butanoic acid (28)
    • R1═C20H31, R2═R3=ethyl, Y═SO and X═COOH
  • Figure US20170029368A1-20170202-C00058
    • 2-ethyl-2-((5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaenylsulfonyl)butanoic acid (29)
    • R1═C20H31, R2═R3=ethyl, Y═SO2 and X═COOH
  • The compounds of categories A-E above, were R2 and R3 are different, are capable of existing in stereoisomeric forms, i.e. all optical isomers of the compounds and mixtures thereof are encompassed. Hence, the said compounds may be present as diastereomers, racemates and enantiomers.
  • General Synthetic Methods for the Compounds Described Herein
  • The compounds of general formula (I) can be prepared by the following general procedures:
  • Method I
  • Figure US20170029368A1-20170202-C00059
  • Method II
  • Figure US20170029368A1-20170202-C00060
  • The alcohols described in method I and II may be prepared directly from the carboxylic esters of, for example, naturally occurring fatty acids; e.g. alpha-linolenic acid, conjugated linoleic acid, eicosapentaenoic acid (EPA), etc. by reduction with a reducing agent like lithium aluminiumhydride or diisobultylaluminiumhydride at −10 to 0° C. The alcohols can also be prepared by degradation of the polyunsaturated fatty acids EPA and DHA, as described by Holmeide et al. (J. Chem. Soc., Perkin Trans. 1, 2000, 2271). In this case one can start with purified EPA or DHA, but it is also possible to start with fish oil containing EPA and DHA in mixture.
  • Compounds of formula (X) and (XI) are commercially available, or they are known in the literature, or they are prepared by standard processes known in the art. The leaving group (LG) present in compounds of formula (XI) may, for example, be mesylate, tosylate or a suitable halogen, such as bromine.
  • Using method I, the resulting alcohols can be converted, using functional group interconversion, by methods familiar to persons skilled in the art (step I), to compounds where the terminal hydroxy group have been transformed into a suitable leaving group (LG). Suitable leaving groups include bromine, mesylate and tosylate. These compounds can be reacted further (step II) in a substitution reaction with the appropriately substituted thiol acetic acid derivatives (X), in the presence of base.
  • Using method II, the alcohols can be converted to the corresponding thiols (step IV) by methods familiar to persons skilled in the art. The thiols can then be reacted further (step V) in a substitution reaction with compounds of formula (XI), in the presence of base in an appropriate solvent system.
  • The corresponding sulfoxides and sulfones (Y═SO or SO2) can be prepared by oxidation of the thioethers (Y═S) with a suitable oxidising agent (step III). Examples of oxidising agents are m-chloro-perbenzoic acid (MCPBA), hydrogen peroxide (H2O2) and oxone (potassium peroxymonosulfate). By using 1 equivivalent or less of the oxidising agent, the main product will be the sulfoxide. By using an excess oxidising agent, the main product will be the sulfone.
  • If the acid derivatives used are carboxylic esters, hydrolysis can be performed to obtain the free fatty acids. An esterifying group such as a methyl of an ethyl group may be removed, for example, by alkaline hydrolysis using a base such as an alkali metal hydroxide, for example LiOH, NaOH or KOH or by using an organic base, for example Et3N together with an inorganic salt, for example LiCl in an appropriate solvent system. A tert-butyl group may be removed, for example, by treatment with an acid, for example an organic acid such as trifluoroacetic acid or formic acid in an appropriate solvent system. An arylmethyl group such as a benzyl group may be removed, for example, by hydrogenation over a catalyst such as palladium-on-carbon in an appropriate solvent system.
  • The preparation of compounds of formula I, according to method I or II, may result in mixtures of stereoisomers. If required, these isomers may be separated by means of chiral resolving agents and/or by chiral column chromatography through methods known to the person skilled in the art.
  • Method III
  • The compounds of formula (I) wherein X is a carboxylic acid and in the form of a phospholipid can be prepared through the following processes.
  • Figure US20170029368A1-20170202-C00061
    • Acylation of sn-glycero-3-phosphocholine (GPC) with an activated fatty acid, such as fatty acid imidazolides, is a standard procedure in phosphatidylcholine synthesis. It is usually carried out in the presence of DMSO anion with DMSO as solvent
  • (Hermetter, Chemistry and Physics of lipids, 1981, 28, 111). Sn-Glycero-3-phosphocholine, as cadmium (II) adduct can also be reacted with the imidazolide activated fatty acid in the presence of DBU (1,8-diazabicyclo[5.4.0]undec-7-ene) to prepare the phosphatidylcholine of the respective fatty acid (International application number PCT/GB2003/002582). Enzymatic transphosphatidylation can effect the transformation of phosphatidylcholine to phosphatidyletanolamine (Wang et al, J. Am. Chem. Soc., 1993, 115, 10487).
  • Phospholipids may also be prepared by enzymatic esterification and transesterification of phospholipids or enzymatic transphosphatidylation of phospholipids. (Hosokawa, J. Am. Oil Chem. Soc. 1995, 1287, Lilja-Hallberg, Biocatalysis, 1994, 195).
  • Method IV
  • The compounds of formula (I) wherein X is a carboxylic acid in the form of a triglyceride can be prepared through the following process. Excess of the fatty acid can be coupled to glycerol using dimethylaminopyridine (DMAP) and 2-(1H-benzotriazol-1-yl)-N,N,N′,N′-tetramethyluroniumhexafluorophosphate (HBTU).
  • Method V
  • The compounds of formula (I) wherein X is a carboxylic acid in the form of a diglyceride can be prepared by reaction of the fatty acid (2 equivalents) with glycerol (1 equivalent) in the presence of 1,3-dicyclohexylcarbondiimide (DCC) and 4-dimethylaminopyridine (DMAP).
  • Method VI
  • The compounds of formula (I) wherein X is a carboxylic acid and in the form of a monoglyceride can be prepared through the following processes.
  • Figure US20170029368A1-20170202-C00062
  • Acylation of 1,2-O-isopropylidene-sn-glycerol with a fatty acid using DCC and DMAP in chloroform gives a monodienoylglycerol. Deprotection of the isopropylidene group can be done by treating the protected glycerol with an acidic (HCl, acetic acid etc.) (O'Brian, J. Org. Chem., 1996, 5914).
  • There are several synthetic methods for the preparation of monoglycerides with the fatty acid in 2-position. One method utilizes esterification of the fatty acid with glycidol in the presence of 1-(3-dimethylaminopropyI)-3-ethylcarbodiimidehydrochloride (EDC) and 4-dimethylaminopyridine (DMAP) to produce a glycidyl derivative. Treatment of the glycidyl derivative with trifluoroacetic anhydride (TFAA) prior to trans-esterification the monoglyceride is obtained (Parkkari et al, Bioorg. Med. Chem. Lett. 2006, 2437).
  • Figure US20170029368A1-20170202-C00063
  • Further methods for the preparation of mono-, di- and tri-glycerides of fatty acid derivatives are described in international patent application, PCT/FRO2/02831.
  • It is also possible to use enzymatic processes (lipase reactions) for the transformation of a fatty acid to a mono-, di-, tri-glyceride. A 1,3-regiospecific lipase from the fungus Mucor miehei can be used to produce triglycerides or diglycerides from polyunsaturated fatty acids and glycerol. A different lipase, the non-regiospecific yeast lipase from Candida antartica is highly efficient in generating triglycerides from polyunsaturated fatty acids (Haraldsson, Pharmazie, 2000, 3).
  • Preparation, Characterisation and Biological Testing of Specific Fatty Acid Derivatives of Formula (I)
  • The invention will now be further described by the following non-limiting examples, in which standard techniques known to the skilled chemist and techniques analogous to those described in these examples may be used where appropriate. Unless otherwise stated:
      • evaporations were carried out by rotary evaporation in vacuo,
      • all reactions were carried out at room temperature, typically in the range between 18-25° C. with solvents of HPLC grade under anhydrous conditions;
      • column chromatography were performed by the flash procedure on silica gel 40-63 μm (Merck) or by an Armen Spotflash using the pre-packed silica gel columns “MiniVarioFlash”, “SuperVarioFlash”, “SuperVarioPrep” or “EasyVarioPrep” (Merck);
      • yields are given for illustration only and are not necessarily the maximum attainable;
      • the nuclear magnetic resonance (NMR) shift values were recorded on a Bruker Avance DPX 200 or 300 instrument, and the peak multiplicities are shown as follows: s, singlet; d, doublet; dd, double doublet; t, triplet; q, quartet; p, pentet, m, multiplett, br, broad;
      • the mass spectra were recorded with a LC/MS spectrometer. Separation was performed using a Agilent 1100 series module on a Eclipse XDB-018 2.1×150 mm column with gradient elution. As eluent were used a gradient of 5-95% acetonitrile in buffers containing 0.01% trifluoroacetic acid or 0.005% sodium formate. The mass spectra were recorded with a G 1956 A mass spectrometer (electrospray, 3000 V) switching positive and negative ionization mode.
    Preparation of Intermediates EXAMPLE 1 Preparation of S-(3Z,6Z,9Z,12Z)-pentadeca-3,6,9,12-tetraenyl ethanethioate
  • Figure US20170029368A1-20170202-C00064
  • Triphenylphosphine (PPh3) (41.7 g, 159 mmol) was dissolved in dry tetrahydrofurane (THF) (250 mL) at 0° C. under inert atmosphere and added diisopropyl azodicarboxylate (DIAD) (30.8 mL, 159 mmol). The mixture was stirred at 0° C. for 40 minutes and then dropwise added a solution of (all-Z)-3,6,9,12-pentadecatetraenol (17.5 g, 79.4 mmol) and thioacetic acid (11.4 mL, 159 mmol) in dry THF (150 mL). The resulting turbid mixture was stirred at 0° C. for 40 minutes, then at ambient temperature for two hours. Heptane was added (300 mL), the mixture was stirred for ten minutes and the precipitated white solid was removed by filtration. This procedure was repeated twice and finally the residue after concentration was stirred in heptane (100 mL) for 16 hours. Filtration and purification of the residue by flash chromatography (1% EtOAc in heptane) provided 13.7 g (62% yield) of the title compound as an oil.
  • 1H-NMR (200 MHz, CDCl3): δ 0.96 (t, 3H), 2.05 (m, 2H), 2.31 (s+m, 5H), 2.76-2.92 (m, 8H), 5.32-5.45 (m, 8H).
  • EXAMPLE 2 Preparation of (3Z,6Z,9Z,12Z)-pentadeca-3,6,9,12-tetraene-1-thiol
  • Figure US20170029368A1-20170202-C00065
  • LiAlH4 (2.05 g, 54.1 mmol) was suspended in dry diethyl ether (100 mL) at 0° C. under inert atmosphere. To this suspension was added dropwise a solution of 5-(3Z,6Z,9Z,12Z)-pentadeca-3,6,9,12-tetraenyl ethanethioate (13.7 g, 49.2 mmol) in dry diethyl ether (50 mL) and the resulting grey mixture was stirred at 0° C. for ten minutes and then at ambient temperature for 30 minutes. The mixture was cooled to −5° C., added 1M HCl until pH=2 and filtrated through a short pad of celite. The pad was washed with water and diethyl ether, the phases were separated and the aqueous phase was extracted twice with diethyl ether (100 mL each). The combined organic extracts were dried (Na2SO4), filtered and concentrated under reduced pressure to afford 7.8 g (67% yield) of the title compound as oil.
  • 1H-NMR (200 MHz, CDCl3): δ 0.96 (t, 3H), 2.06 (m, 2H), 2.39 (m, 2H), 2.51 (m, 2H), 2.81 (m, 6H), 5.28-5.54 (m, 8H), MS (ESI): 235 [M−H].
  • EXAMPLE 3 Preparation of S-(5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaenyl ethanethioate
  • Figure US20170029368A1-20170202-C00066
  • Triphenylphosphine (21.0 g, 80 mmol) was dissolved in dry THF (170 mL) at 0° C. under inert atmosphere and added DIAD (15.8 mL, 80 mmol) dropwise. After 40 minutes at 0° C. the white suspension was added dropwise to a solution of (5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaen-1-ol (11.5 g, 40 mmol) and thioacetic acid (5.7 mL, 80 mmol) in dry THF (50 mL) during 15 minutes. The resulting turbid mixture was stirred at 0° C. for 30 minutes, followed by ambient temperature for 1.5 hour. Heptane was added (200 mL), the mixture was stirred for ten minutes and the precipitated white solid removed by filtration and rinsed with heptane (150 mL). The residue was concentrated to remove most of the THF and stirred at ambient for 18 hours. The mixture was filtered, concentrated and added heptane (200 mL). The resulting mixture was stirred for 2 hours, filtered and evaporated. The residue was purified by flash chromatography on silica gel, using EtOAc:Heptane (2:98), followed by EtOAc:Heptane (4:96) and finally EtOAc:Heptane (5:95). Concentration of the appropriate fractions provided 11.0 g (79% yield) of the title compound as oil.
  • 1H-NMR (300 MHz, CDCl3): δ 0.95 (t, 3H, J=7.5 Hz), 1.40 (m, 2H), 1.58 (m, 2H), 2.06 (m, 4H), 2.29 (s, 3H), 2.77 2.87 (m, 10H), 5.255.42 (m, 10H), MS (Cl (CH4)): 387 [M+C3H5]+, 375 [M+C2H5]+, 347 [M+H]+, 333 [M−CH2]+, 305 [R−SH]+.
  • EXAMPLE 4 Preparation of (5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaene-1-thiol
  • Figure US20170029368A1-20170202-C00067
  • S-(5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaenyl ethanethioate (7.00 g, 20.2 mmol) was dissolved in MeOH (100 mL) by stirring 10 minutes until the droplets of oil dissolved, before anhydrous potassium carbonate, K2CO3 (2.79 g, 20.2 mmol) was added in one portion. The mixture was stirred for 1 hour and 20 minutes at ambient temperature and quenched by addition of 1 M HCl (50 mL) and water (150 mL). The white cloudy mixture was added Et2O (250 mL) and the phases were separated. The water phase was extracted with Et2O (2×250 mL). The combined organic phases were washed with brine (250 mL) and dried (MgSO4). Filtration and evaporation gave the title compound as oil (5.99 g, 97% yield), which was used without further purification.
  • 1H-NMR (300 MHz, CDCl3): δ 0.96 (t, 3H, J=7.5 Hz), 1.31 (t, 1H, J=7.8 Hz), 1.44 (m, 2H), 1.61 (m, 2H), 2.06 (m, 4H), 2.51 (m, 2H), 2.77 2.85 (m, 8H), 5.28 5.41 (m, 10H), MS (Cl(CH4)): 345 [M+C3H5]+, 333 [M+C2H5]+, 305 [M+H]+, 271 [M−SH]+.
  • EXAMPLE 5 Preparation of (5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaenyl methanesulfonate
  • Figure US20170029368A1-20170202-C00068
  • Et3N (1.50 mL, 10.8 mmol) and methanesulfonyl chloride (402 μL, 5.20 mmol) was added to a solution of (5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaen-1-ol (1.15g, 4.0 mmol) in CH2Cl2 (40 mL) held at 0° C. under nitrogen. The mixture was stirred at 0° C. for one hour, and poured into ice-water (100 g) and the water phase extracted with Et2O (50 mL). The combined organic extracts were added 0.5 M H2SO4 (35 mL), the organic phase washed with NaHCO3 (sat. aq.) (25 mL), before dried (Mg2SO4, 10 gram). Filtration and concentration in vacuo afforded 1.24 gram of crude oil. Purification on Armen, SVP D26 column packed with 30 gram of 15-40 μm Merck silica, flow 20 mL/min, UV 210 nm and collecting 15 mL fraction, was performed using gradient elution: (starting heptane:EtOAc (100:0) and increasing during 10 min. to 10% EtOAc, then increasing 5 min. to 20% EtOAc (hold 10 min.), then increasing in 5 min. to 40% EtOAc (hold 0 min.). Fractions 6-14 afforded 1.16 g (79% yield) of the title compound as oil.
  • 1H-NMR (300 MHz, CDCl3): δ 0.97 (t, 3H), 1.50 (m, 2H), 1.75 (m, 2H), 2.03-2.15 (m, 4H), 2.76-2.86 (m, 8H), 2.99 (s, 3H), 4.22 (t, 2H), 5.27-5.40 (m, 10H), MS (electrospray): 389.2 [M+Na]+.
  • EXAMPLE 6 Preparation of (4S,5R)-3-((S)-2-((5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaenylthio)butanoyl)-4-methyl-5-phenyloxazolidin-2-one and (4S,5R)-3-((R)-2-((5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaenylthio)butanoyl)-4-methyl-5-phenyloxazolidin-2-one
  • Figure US20170029368A1-20170202-C00069
  • A mixture of 2-((5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaenylthio)butanoic acid (3.0 g, 7.9 mmol) in dry dichloromethane (40 mL) held at 0° C. under nitrogen was added DMAP (1.0 g, 9.5 mmol) and 1,3-dicyclohexylcarbodiimide (DCC) (1.8 g, 8.7 mmol). The resulting mixture was stirred at 0° C. for 20 minutes, (4S,5R)-4-methyl-5-phenyl-2-oxazolidinone (1.7 g, 9.5 mmol) was added and the resulting turbid mixture was stirred at ambient temperature for 24 hours. The mixture was filtrated and concentrated under reduced pressure to give a crude product containing the desired product as a mixture of two diastereomers. The residue was purified by flash chromatography on Armen Spotflash instrument on silica gel using 2% ethyl acetate in heptane as eluent. The two diastereomers were separated and the appropriate fractions were concentrated. (4S,5R)-3-((R)-2-((5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaenylthio)butanoyl)-4-methyl-5-phenyloxazolidin-2-one eluted first and was obtained in 0.95 g (47% yield) as an oil. 1.47 g (67% yield) of (4S,5R)-3-((S)-2-((5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaenylthio)butanoyl)-4-methyl-5-phenyloxazolidin-2-one was obtained as an oil.
  • (4S,5R)-3-((R)-2-((5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaenylthio)butanoyl)-4-methyl-5-phenyloxazolidin-2-one (E1):
    • 1H-NMR (300 MHz, CDCl3): δ 0.93-1.06 (m, 9H), 1.45-1.60 (m, 4H), 1.75-1.85 (m, 1H), 2.05-2.15 (m, 5H), 2.55-2.70 (m, 2H), 2.87 (m, 8H), 4.69 (t, 1H), 4.79 (p, 1H), 5.30-5.45 (m, 10H), 5.72 (d, 1H), 7.32 (m, 2H), 7.43 (m, 3H).
  • (4S,5R)-3-((S)-2-((5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaenylthio)butanoyl)-4-methyl-5-phenyloxazolidin-2-one:
  • 1H-NMR (300 MHz, CDCl3): δ 0.93 (d, 3H), 0.99 (t, 3H), 1.05 (t, 3H), 1.40-1.56 (m, 4H), 1.50-1.75 (m, 1H), 2.00-2.15 (m, 5H), 2.47-2.65 (m, 2H), 2.83 (m, 8H), 4.62 (t, 1H), 4.85 (p, 1H), 5.25-5.45 (m, 10H), 5.70 (d, 1H), 7.32 (m, 2H), 7.43 (m, 3H).
  • Preparation of Target Molecules EXAMPLE 7 Preparation of ethyl 2-((3Z,6Z,9Z,12Z)-pentadeca-3,6,9,12-tetraenylthio)butanoate (30)
  • Figure US20170029368A1-20170202-C00070
  • A solution of 3Z,6Z,9Z,12Z)-pentadeca-3,6,9,12-tetraene-1-thiol (9.80 g, 41.5 mmol) in dry dimethylformamide (DMF) (70 mL) at 0° C. under inert atmosphere was added NaH (60% in mineral oil, 1.82 g, 45.6 mmol) and stirred at this temperature for ten minutes. Ethyl bromobutyrate (6.39 mL, 43.5 mmol) was added and the mixture was stirred at ambient temperature for 30 minutes. The mixture was partitioned between saturated NH4Cl (150 mL) and heptane (150 mL). The aqueous layer was extracted twice with heptane (100 mL each) and the combined organic extract were washed with water (100 mL) and brine (100 mL). The organic layer was dried (Na2SO4), filtrated and concentrated. The residue was purification by flash chromatography on silica gel (heptane:EtOAc 99:1 then 95:5). Concentration of the appropriate fractions afforded 14.1 g (97% yield) of the title compound as oil.
  • 1H-NMR (200 MHz, CDCl3): δ 0.92-1.01 (2×t, 6H), 1.27 (t, 3H), 1.60-1.80 (m,1H), 1.80-1.95 (m, 1H), 2.00-2.15 (m, 2H) 2.25-2.45 (m, 2H), 2.60-2.75 (m, 2H), 2.80 (m, 6H), 3.15 (t, 1H), 4.17 (q, 2H), 5.31-5.43 (m, 8H), MS (ESI): 373 [M+Na]+.
  • EXAMPLE 8 Preparation of ethyl 2-((3Z,6Z,9Z,12Z)-pentadeca-3,6,9,12-tetraenylsulfonyl)butanoate (31).
  • Figure US20170029368A1-20170202-C00071
  • Ethyl 2-((3Z,6Z,9Z,12Z)-pentadeca-3,6,9,12-tetraenylthio)butanoate (2.7 g, 7.7 mmol) was dissolved in dry CHCl3 (40 mL) and the solution was cooled down to −20° C. under inert atmosphere. meta-Chloroperoxybenzoic acid (mCPBA) (˜77%, 4.0 g, 18 mmol) dissolved in dry CHCl3 (10 mL) was added dropwise and the resulting solution was stirred at −20° C. for 30 minutes, allowed to slowly reach ambient temperature and then stirred over night. The solvents were evaporated in vacuo, the residue was added heptane (30 mL) and the resulting white precipitate was removed by filtration. The filtrate was concentrated in vacuo and the residue was added heptane (10 mL). The resulting white precipitate was again removed by filtration. The filtrate was concentrated in vacuo and and the residue was purified by flash chromatography on silica gel (heptane:EtOAc 4:1). Concentration of the appropriate fractions afforded 0.37 g (13% yield) of the title compound as an oil.
  • 1H NMR (300 MHz, CDCl3): δ 0.96 (t, 3H), 1.03 (t, 3H), 1.31 (t, 3H), 2.02-2.15 (m, 4H), 2.62 (m, 2H), 2.82 (m, 6H), 3.05 (m, 1H), 3.20 (m, 1H), 3.70 (dd, J=10.3 Hz, J=4.7 Hz, 1H), 4.28 (q, 2H), 5.26-5.41 (m, 7H), 5.46-5.52 (m, 1H), MS (electrospray): 405.2 [M+Na]+
  • EXAMPLE 9 Preparation of 2-((3Z,6Z,9Z,12Z)-pentadeca-3,6,9,12-tetraenylthio)butanoic acid (7)
  • Figure US20170029368A1-20170202-C00072
  • Ethyl 2-((3Z,6Z,9Z,12Z)-pentadeca-3,6,9,12-tetraenylthio)butanoate (14.1 g, 40.2 mmol) was dissolved in ethanol (200 mL) and added a solution of LiOH×H2O (13.5 g, 322 mmol) in water (50 mL). The resulting turbid solution was stirred at 70° C. under inert atmosphere for 90 minutes, cooled, added water (100 mL) and 3M HCl until pH=2. The mixture was extracted three times with heptane (100 mL each). The combined organic extracts were dried (Na2SO4), filtered and concentrated under reduced pressure to afford 11.8 g (91% yield) of the title compound as oil.
  • 1H-NMR (200 MHz, CDCl3): δ 0.91-1.06(2×t, J=7.2 Hz, J=7.5 Hz, 6H), 1.60-1.80 (m, 1H), 1.80-1.95 (m, 1H), 2.05 (p, J=7.2 Hz, 2H), 2.35 (m, 2H), 2.60-2.75 (m, 2H), 2.75-2.90 (m, 6H), 3.14 (t, J=7.1 Hz, 1H), 5.31-5.47 (m, 8H), MS (ESI): 321 [M−H]+.
  • EXAMPLE 10 Preparation of 2-((3Z,6Z,9Z,12Z)-pentadeca-3,6,9,12-tetraenylsulfinyl)butanoic acid (24)
  • Figure US20170029368A1-20170202-C00073
  • 2-((3Z,6Z,9Z,12Z)-Pentadeca-3,6,9,12-tetraenylthio)butanoic acid (0.20 g, 0.62 mmol) was dissolved in dry CHCl3 (10 mL) and the solution was cooled down to −20° C. under inert atmosphere. mCPBA (˜77%, 0.15 g, 0.68 mmol) dissolved in dry CHCl3 (2 mL) was added dropwise and the resulting solution was stirred at −20° C. for 35 minutes. The solvents were evaporated in vacuo, the residue was added heptane (10 mL) and the resulting white precipitate was removed by filtration. The filtrate was concentrated in vacuo and the residue was added heptane (10 mL). The resulting white precipitate was again removed by filtration. The filtrate was concentrated in vacuo and the residue was purified by flash chromatography on silica gel (heptane:EtOAc+w/1% HCOOH 4:1-1:1). Concentration of the appropriate fractions afforded 100 mg (48% yield) of the title compound as an oil.
  • 1H NMR (200 MHz, CDCl3): δ 0.95 (t, 3H), 1.10 (2 x q, 3H), 1.70-1.80(m, 1H), 2.05 (m, 3.5H), 2.20-2-40 (m, 0.5H), 2.60 (m, 2H), 2.81 (m, 7H), 2.90-3.00 (m, 0.5H), 3.10-3.25 (m, 1H), 3.70 (dd, 0.5H), 5.25-5.55 (m, 8H),L MS (electrospray): 337.1 [M−H].
  • EXAMPLE 11 Preparation of 2-((3Z,6Z,9Z,12Z)-pentadeca-3,6,9,12-tetraenylsulfonyl)butanoic acid (25)
  • Figure US20170029368A1-20170202-C00074
  • Ethyl 2-((3Z,6Z,9Z,12Z)-pentadeca-3,6,9,12-tetraenylsulfonyl)butanoate (370 mg, 0.97 mmol) was dissolved in ethanol (10 mL) and added a solution of LiOH in H2O (1 M, 3.9 mL, 3.9 mmol). The resulting mixture was stirred at 60° C. for three hours, cooled, added 0.1 M HCl until pH=2 and extracted twice with diethyl ether (15 mL each). The combined organic layer was washed with brine (15 mL), dried, filtrated, concentrated in vacuo and purified by flash chromatography on silica gel (heptane:EtOAc w/5% HCOOH 4:1). Concentration of the appropriate fractions afforded 250 mg (73% yield) of the title compound as an oil.
  • 1H NMR (300 MHz, CDCl3): δ 0.96 (t, 3H), 1.09 (t, 3H), 2.02-2.25 (m, 4H), 2.65 (m, 2H), 2.82 (m, 6H), 3.10 (m, 1H), 3.20 (m, 1H),
  • EXAMPLE 12 Preparation of ethyl 2-((5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaenylthio)propanoate (32)
  • Figure US20170029368A1-20170202-C00075
  • (5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaene-1-thiol (305 mg, 1.00 mmol) was added to a solution of NaH (60% in mineral oil, 44 mg, 1.10 mmol) in dry DMF (10 mL) held at 0° C. under inert atmosphere. After ten minutes ethyl bromopropionate (136 μL, 1.05 mmol) was added and the mixture was stirred for 1.5 hour at 0° C. The reaction mixture was added sat. aq. NH4Cl (20 mL) and heptane (50 mL). The phases were separated and the water phase extracted with heptane (2×25 mL). The combined organics were washed with brine (25 mL), dried (MgSO4), filtered and evaporated to give 376 mg of title compound as crude oil. Purification by flash chromatography on silica gel using gradient elution (starting pure heptane and increasing stepwise to heptane:EtOAc 95:5) afforded 318 mg (79% yield) of the title compound as oil.
  • 1H-NMR (300 MHz, CDCI3): δ 0.95 (t, 3H), 1.25 (t, 3H), 1.41 (d, 3H), 1.44 (m, 2H), 1.58 (m, 2H), 2.06 (m, 4H), 2.60 (m, 2H), 2.71 2.85 (m, 8H), 3.36 (d, 1H), 4.17 (m, 2H), 5.25-5.40 (m, 10H), MS (Cl (CH4)): 445 [M+C3H5]+, 433 [M+C2H5]+, 405 [M+H]+, 359 [M−OEt]+, 331 [M−CO2Et]+, 303 [R−S]•+.
  • EXAMPLE 13 Preparation of ethyl 2-((5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaenylthio)butanoate (33)
  • Figure US20170029368A1-20170202-C00076
  • To a solution of (5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaene-1-thiol (305 mg, 1.00 mmol) in dry DMF (10 mL) at 0° C. under inert atmosphere was added NaH (60% in mineral oil, 44 mg, 1.1 mmol). After fifteen minutes ethyl bromobutyrate (154 μL, 1.05 mmol) was added. The mixture was stirred for 1 hour at 0° C. Sat. aq. NH4Cl (20 mL), water (20 mL) and heptane (50 mL) were added. The phases were separated and the water phase was extracted with heptane (2×25 mL). The combined organics were washed with water (25 mL) and brine (25 mL), dried (MgSO4), filtered and evaporated to give 379 mg of the title compound as a crude oil. Purification by flash chromatography on silica gel using gradient elution (starting pure heptane and increasing stepwise to heptane:EtOAc 95:5) afforded 345 mg (82% yield) of the title compound as oil.
  • 1H-NMR (300 MHz, CDCl3): δ 0.931.00 (m, 6H), 1.25 (t, 3H), 1.44 (m, 2H), 1.59 (m, 2H), 1,68 (m, 1H), 1.87 (m, 1H), 2.07 (m, 4H), 2.57 (m, 2H), 2.73 2.88 (m, 8H), 3.12 (m, 1H), 4.17 (m, 2H), 5.275.46 (m, 10H), MS (Cl (CH4)): 459 [M+C3H5]+, 447 [M+C2H5]+, 419 [M+H]+, 373 [M−OEt]+, 345 [M−CO2Et]+, 303 [R−S]•+.
  • EXAMPLE 14 Preparation of 2-((5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaenylthio)butanoic acid (10)
  • Figure US20170029368A1-20170202-C00077
  • Ethyl 2-((5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaenylthio)butanoate (209 mg, 0.50 mmol) was dissolved in ethanol (2.5 mL) and added to a solution of LiOH×H2O (168 mg, 4.0 mmol) in water (2.5 mL). The resulting turbid solution was stirred at 70° C. under inert atmosphere for 2 hours, cooled and added water (10 mL) and 1 M HCl (5 mL) to pH=1-2. The mixture was extracted with heptane (2×20 mL) and diethyl ether (20 mL). The combined organic extracts were dried (MgSO4), filtered and concentrated under reduced pressure to give 154 mg of the title compound as crude oil. Purification by flash chromatography on silica gel using gradient elution (starting with pure heptane and increasing stepwise to heptane:EtOAc (with 5% HOAc) 80:20) afforded 151 mg (77% yield) of the title compound as oil.
  • 1H-NMR (300 MHz, CDCl3): δ 0.95 (t, 3H), 1.02 (t, 3H), 1.46 (m, 2H), 1.52-1.78 (m, 3H), 1.90 (m, 1H), 2.05 (m, 4H), 2.63 (m, 2H), 2.75-2.90 (m, 8H), 3.14 (t, 1H) (m, 1H), 4.17 (m, 2H), 5.27-5.46 (m, 10H).
  • EXAMPLE 15 Preparation of (S)-2-((5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaenylthio)butanoic acid (34)
  • Figure US20170029368A1-20170202-C00078
  • Hydrogen peroxide (30% in water, 0.71 mL, 6.91 mmol) and lithium hydroxide monohydrate (0.15 g, 3.46 mmol) was added to a solution of (4S,5R)-3-((S)-2-((5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaenylthio)butanoyl)-4-methyl-5-phenyloxazolidin-2-one (0.95 g, 1.73 mmol) in tetrahydrofuran (12 mL) and water (4 mL) held at 0° C. under nitrogen. The reaction mixture was stirred at 0° C. for 30 minutes. 10% Na2SO3 (aq) (30 mL) was added, the pH was adjusted to ˜2 with 5M HCl and the mixture was extracted twice with heptane (30 mL). The combined organic extract was dried (Na2SO4), filtered and concentrated. The residue was subjected to flash chromatography on silica gel using increasingly polar mixtures of heptane and ethyl acetate (98:8→1:1) as eluent. Concentration of the appropriate fractions afforded 0.15 g (17% yield) of the title product as an oil.
  • 1H-NMR (300 MHz, CDCl3): δ 1.00 (t, 3H), 1.07 (t, 3H), 1.46 (m, 2H), 1.60-1.75 (m, 3H), 1.85 (m, 1H), 2.10 (m, 4H), 2.66 (m, 2H), 2.80-2.90 (m, 8H), 3.21 (t, 1H), 5.35-5.45 (m, 10H), MS (electrospray): 389.3 [M−H]; [α]D−49° (c=0.12, ethanol).
  • EXAMPLE 16 Preparation of (R)-2-((5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaenylthio)butanoic acid (35)
  • Figure US20170029368A1-20170202-C00079
  • Hydrogen peroxide (30% in water, 1.04 mL, 10.2 mmol) and lithium hydroxide monohydrate (0.21 g, 5.09 mmol) was added to a solution of (4S,5R)-3-((R)-2-((5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaenylthio)butanoyl)-4-methyl-5-phenyloxazolidin-2-one (1.40 g, 2.55 mmol) in tetrahydrofuran (15 mL) and water (5 mL) held at 0° C. under nitrogen. The reaction mixture was stirred at 0° C. for 45 minutes. 10% Na2SO3 (aq) (35 mL) was added, pH was adjusted to ˜2 with 5M HCl and the mixture was extracted twice with heptane (35 mL). The combined organic extract was dried (Na2SO4), filtered and concentrated. The residue was subjected to flash chromatography on silica gel using increasingly polar mixtures of heptane and ethyl acetate (98:8→1:1) as eluent. Concentration of the appropriate fractions afforded 0.17 g (22% yield) of the title product as an oil. 1H-NMR (300 MHz, CDCl3): δ 1.00 (t, 3H), 1.07 (t, 3H), 1.46 (m, 2H), 1.60-1.75 (m, 3H), 1.85 (m, 1H), 2.10 (m, 4H), 2.66 (m, 2H), 2.80-2.90 (m, 8H), 3.21 (t, 1H), 5.35-5.45 (m, 10H), MS (electrospray): 389.3 [M−H]; [α]D+50° (c=0.14, ethanol).
  • EXAMPLE 17 Preparation of ethyl 2-((5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaenylthio)-2-methylpropanoate (36)
  • Figure US20170029368A1-20170202-C00080
  • To a solution of (5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaene-1-thiol (305 mg, 1.00 mmol) in dry DMF (10 mL) at 0° C. under inert atmosphere was added NaH (60% in mineral oil, 44 mg, 1.1 mmol). After fifteen minutes ethyl 2-bromo-2-methylbutyrate (154 μL, 1.05 mmol) was added and the mixture was stirred for 1.5 hour at 0° C. The reaction mixture was quenched by addition of sat. aq. NH4Cl (20 mL). Water (20 mL) and heptane (50 mL) were added and the phases were separated. The water phase was extracted with heptane (2×25 mL). The combined organics were washed with water (25 mL) and brine (2×25 mL), dried (MgSO4), filtered and evaporated to give 377 mg of the title compound as a crude oil. Purification by flash chromatography on silica gel using isocratic elution (heptane:EtOAc 98:2) afforded 307 mg (77% yield) of the title compound as oil.
  • 1H-NMR (300 MHz, CDCl3): δ 0.95 (t, 3H), 1.28 (t, 3H), 1.42 (m, 2H), 1.48 (s, 6H), 1.54 (m, 2H), 2.06 (m, 4H), 2.58 (m, 2H), 2.71 2.85 (m, 8H), 4.15 (m, 2H), 5.22-5.48 (m, 10H), MS (Cl (CH4)): 459 [M+C3H5]+, 447 [M+C2H5]+, 419 [M+H]+, 373 [M−OEt]+, 345 [M−CO2Et]+, 303 [R−S]•+.
  • EXAMPLE 18 Preparation of 2-((5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaenylthio)-2-methylpropanoic acid (11)
  • Figure US20170029368A1-20170202-C00081
  • Ethyl 2-((5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaenylthio)-2-methylpropanoate (209 mg, 0.50 mmol) was dissolved in ethanol (2.5 mL) and added to a solution of LiOH×H2O (168 mg, 4.0 mmol) in water (2.5 mL). The resulting turbid solution was stirred at 70° C. under inert atmosphere for 2 hours, cooled and added water (10 mL) and 1 M HCl (5 mL) to pH=1-2. The mixture was extracted three times with heptane (3×20 mL). The combined organic extracts were dried (MgSO4), filtered and concentrated under reduced pressure to give 101 mg of the title compound as crude oil. Purification by flash chromatography on silica gel using gradient elution (starting with pure heptane and increasing stepwise to heptane:EtOAc (with 5% HOAc) 80:20) afforded 78 mg (40%) of the title compound as oil.
  • 1H-NMR (300 MHz, CDCl3): δ 0.95 (t, 3H), 1.35 1.66 (m, 4H), 1.50 (s, 6H), 2.07 (m, 4H), 2.63 (t, 3H), 2.70 2.92 (m, 8H), 5.135.50 (m, 10H).
  • EXAMPLE 19 Preparation of ethyl 1-((5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaenylthio)cyclobutanecarboxylate (37)
  • Figure US20170029368A1-20170202-C00082
  • To a solution of (5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaene-1-thiol (305 mg, 1.00 mmol) in dry DMF (10 mL) at 0° C. under inert atmosphere was added NaH (60% in mineral oil, 44 mg, 1.1 mmol). After fifteen minutes ethyl 2-bromo-carboxylate (170 μL, 1.05 mmol) was added and the mixture was stirred for 1.5 hour at 0° C. The reaction was quenched by addition of sat. aq. NH4Cl (20 mL). Heptane (50 mL) was added, and the phases were separated. The water phase was extracted with heptane (2×25 mL). The combined organics were washed with water (25 mL) and brine (25 mL), dried (MgSO4), filtered and evaporated to give 409 mg of the title compound as a crude oil. Purification by flash chromatography on silica gel using isocratic elution (heptane:acetone 98:2) afforded 243 mg (56% yield) of the title compound as oil.
  • 1H-NMR (300 MHz, CDCl3): δ 0.95 (t, 3H), 1.27 (t, 3H), 1.42 (d, 3H), 1.54 (m, 2H), 1.84 (m, 1H), 1.96 2.23 (m, 7H), 2.51 (m, 2H), 2.60 (m, 2H), 2.73 2.90 (m, 8H), 4.18 (m, 2H), 5.235.43 (m, 10H), MS (Cl (CH4)): 471 [M+C3H5]+, 459 [M+C2H5]+, 431 [M+H]+, 385 [M−OEt]+, 357 [M−CO2Et]+, 303 [R−S]•+.
  • EXAMPLE 20 Preparation of 2-ethyl-2-((5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaenylthio)butanoic acid (12)
  • Figure US20170029368A1-20170202-C00083
  • NaOEt (21 wt. % in EtOH, 0.37 mL, 0.98 mmol) was added dropwise to a solution of 2-mercapto-2-ethyl butyric acid (0.08 g, 0.49 mmol) in dry EtOH (7 mL) held at 0° C. under inert atmosphere. The resulting mixture was stirred at 0° C. for 30 minutes before a solution of (5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaenyl methanesulfonate (0.15 g, 0.41 mmol) in dry EtOH (3 mL) was added dropwise. The resulting turbid mixture was stirred at ambient temperature for 24 hours, poured into NH4Cl (sat.) (aq.) (15 mL), added 3M HCl to pH ˜2 before extracted twice with EtOAc (2×20 mL). The combined organic extracts were washed with brine (10 mL), dried (MgSO4), filtrated and evaporated in vacuo. The residue was purified by flash chromatography on silica gel using a gradient of 10-25% ethyl acetate in heptane as eluent. Concentration of the appropriate fractions afforded 0.12 g (70% yield) of the title compound as oil.
  • 1H-NMR (300 MHz, CDCl3): δ 0.88-1.02 (m, 9H), 1.45-1.58 (2×m, 4H), 1.72 (m, 2H), 1.82 (m, 2H) 2.09 (m, 4H), 2.53 (t, 2H), 2.76-2.86 (m, 8H), 5.29-5.39 (m, 10H. MS (electrospray): 417.3 [M−H]−;
  • EXAMPLE 21 Preparation of ethyl ethyl 2-((5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaenylthio)-2-phenylacetate (38)
  • Figure US20170029368A1-20170202-C00084
  • To a solution of (5Z,8Z,11Z,14Z,17Z)-icosa-5,8,11,14,17-pentaene-1-thiol (305 mg, 1.00 mmol) in dry DMF (10 mL) at 0° C. under inert atmosphere was added NaH (60% in mineral oil, 44 mg, 1.1 mmol). After fifteen minutes ethyl 2-bromo-2-phenyl acetate (255 mg, 1.05 mmol) was added and the mixture stirred for 1.5 hour at 0° C. The reaction mixture was quenched by addition of sat. aq. NH4Cl (25 mL). Heptane (50 mL) was added and the phases were separated. The water phase was extracted with heptane (2×25 mL). The combined organics were washed with water (25 mL) and brine (25 mL), dried (MgSO4), filtered and evaporated to give 453 mg of title compound as crude oil. Purification by flash chromatography on silica gel using isocratic elution (heptane:EtOAc 98:2) afforded 177 mg (38% yield) of the title compound as oil.
  • 1H-NMR (300 MHz, CDCl3): δ 0.95 (t, 3H), 1.24 (t, 3H), 1.41 (m, 2H), 1.56 (m, 2H), 2.05 (m, 2H), 2.51 (m, 2H), 2.60 (m, 2H), 2.67 2.92 (m, 8H), 4.17 (m, 2H), 4.53 (s, 1H), 5.21 5.46 (m, 10H), 7.27 7.35 (m, 3H), 7.43 7.46 (m, 2H), MS (Cl (CH4)): 507 [M+C3H5]+, 495 [M+C2H5]+, 467 [M+H]+, 421 [M−OEt]+, 393 [M−CO2Et]+, 303 [R−S]•+.
  • Biological Testing EXAMPLE 22 Evaluation of PPAR Activation In-Vitro
  • The assay was carried out in-vitro in three stable reporter cell lines, PPARα, PPARδ or PPARγ, expressing respectively a chimeric protein containing the ligand binding domain (LBD) of human PPARα, human PPARδ or human PPARγ fused to the yeast transactivator GAL4 DNA binding domain (DBD).
  • The luciferase (Luc) reporter gene is driven by a pentamer of the GAL4 recognition sequence in front of a 3-globin promoter. The use of GAL4-PPARα, GAL4-PPARδ and GAL4-PPARγ chimeric receptors allows for elimination of background activity from endogenous receptors and quantitation of relative activity across the three PPAR subtypes with the same reporter gene.
  • Two unsubstituted reference substances, Reference 1 and 2, and five test substances, (7), (10), (11), (24) and (25) were tested in a concentration of 10 μM. The structural formulae of the tested substances are as show below:
  • Figure US20170029368A1-20170202-C00085
  • The PPAR selectivity of the substances was determined by comparison to known drug references (1 μM GW7647 for PPARα, 1 μM L-165041 for PPARδ and 1 μM BRL49653 for PPARγ) set of 100% activity.
  • The results are presented in FIG. 1.
  • EXAMPLE 23 Evaluation of PPARα Activation In-Vitro (Concentration Response Data)
  • The assay was carried out in-vitro using mammalian-one-hybrid assays (M1H) comprising GAL4-DNA binding domain-PPARα-LBD fusion constructs in conjunction with 5×GAL4-sites driven Photinus pyralis luciferase reporter construct in transiently transfected HEK293 cells.
  • Compound (12) and positive control (GW7647) were tested at different concentrations. The results are presented in Table 1.
  • TABLE 1
    PPARα
    Compound EC50 (nM) Efficacy (%)
    GW7647 0.45 100
    (12) 286 84
  • EXAMPLE 24 Evaluation of the Effects on Lipid Metabolism In-Vivo in a Dyslipidemic Model (APOE*3Leiden Transgenic Mice)
  • This animal model has proven to be representative for the human situation regarding plasma lipoprotein levels, lipoprotein profiles, its responsiveness to hypolipidemic drugs (like statins, fibrates etc.) and nutrition. In addition, depending on the level of plasma cholesterol APOE*3Leiden mice develop atherosclerotic lesions in the aorta resembling those found in humans with respect to cellular composition and morphological and immunohistochemical characteristics.
  • Female APOE*3Leiden mice were put on a semi-synthetic Western-type diet (WTD, 15% cocoa butter, 40% sucrose and 0.25% cholesterol; all w/w). With this diet the plasma cholesterol level reached mildly elevated levels of about 12-15 mmol/l. After a 4 weeks run-in period the mice were sub-divided into groups of 10 mice each, matched for plasma cholesterol, triglycerides and body weight (t=0).
  • The test substances were administered orally as admix to the Western-type diet. To facilitate the mixing of the compounds sunflower oil was added to a total oil volume of 10 mL/kg diet.
  • After three weeks of treatment (t=3 weeks) mice were fasted overnight (o/n) and blood samples were taken to measure plasma ketone bodies and free fatty acids. At t=0 and 4 weeks blood samples were taken after a 4 hour-fast period to measure plasma cholesterol and triglycerides.
  • Two unsubstituted reference substances, Reference 3 and 2, and three test substances, (7), (10) and (12), were dosed at 0.3 mmol/kg bw/day. The structural formulae of the tested substances are as show below:
  • Figure US20170029368A1-20170202-C00086
  • The results are shown in FIG. 2.
  • EXAMPLE 25 Evaluation of the Effects on Glucose Metabolism in a Diabetes Type-II Model (Male Ob/Ob Mice)
  • Ob/ob mice can be used as a model for type II diabetes. The mice are homozygous for the obese spontaneous mutation (Lee) leading to leptin deficiency. In addition to obesity (ob/ob mice may reach three times the normal body weight of wild type controls), ob/ob mice exhibit a diabetes type II-like syndrome of hyperglycemia, glucose intolerance, elevated plasma insulin, infertility, impaired wound healing, and an increase in hormone production from both pituitary and adrenal glands.
  • Male ob/ob mice were put on a normal low-fat diet for a few weeks for acclimatization. After the acclimatization period the mice were sub-divided into three groups of 10 mice each, matched for body weight, plasma glucose and insulin (t=0).
  • All compounds were administered orally as admix to AM II diet. To facilitate the mixing of the compounds, sunflower oil was added to a total oil volume of 10 ml/kg diet.
  • At t=0, 2 and 4 weeks body weight and food intake was measured. At t=0, 2 and 4 weeks blood samples were taken after a 4 hour-fast period to measure whole blood HbA1c and plasma glucose, insulin, cholesterol and triglycerides.
  • Pioglitazone was used as reference (15 mg/kg bw/day). Compound (10) was dosed at 0.6 mmol/kg bw/day. The results (t=4) are shown in FIGS. 3-6.

Claims (2)

1. A lipid compound of formula (I):
Figure US20170029368A1-20170202-C00087
wherein
R1 is selected from a C10 -C22 alkyl group, a C10 -C22 alkenyl group having 1-6 double bonds, and a C10-C22 alkynyl group having 1-6 triple bonds;
R2 and R3 are the same or different and may be selected from a group of substituents consisting of a hydrogen atom, a hydroxy group, an alkyl group, a halogen atom, an alkoxy group, an acyloxy group, an acyl group, an alkenyl group, an alkynyl group, an aryl group, an alkylthio group, an alkoxycarbonyl group, a carboxy group, an alkylsulfinyl group, an alkylsulfonyl group, an amino group, and an alkylamino group, provided that R2 and R3 cannot both be a hydrogen atom; or
R2 and R3 can be connected in order to form a cycloalkane like cyclopropane, cyclobutane, cyclopentane or cyclohexane;
Y is selected from sulphur, sulfoxide, and sulfone;
X represents a carboxylic acid or a derivative thereof, a carboxylic ester or a carboxamide;
or a pharmaceutically acceptable salt, solvate, solvate of such salt or a prodrug thereof.
2-86. (canceled)
US15/040,260 2008-07-15 2016-02-10 Novel sulphur containing lipids for use as food supplement or as medicament Abandoned US20170029368A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US15/040,260 US20170029368A1 (en) 2008-07-15 2016-02-10 Novel sulphur containing lipids for use as food supplement or as medicament

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
US8080408P 2008-07-15 2008-07-15
EP08160450.6 2008-07-15
EP08160450A EP2147910A1 (en) 2008-07-15 2008-07-15 Novel lipid compounds
PCT/NO2009/000262 WO2010008299A1 (en) 2008-07-15 2009-07-13 Novel sulphur containing lipids for use as food supplement or as medicament
US201113054212A 2011-04-13 2011-04-13
US14/263,793 US20140316002A1 (en) 2008-07-15 2014-04-28 Novel sulphur containing lipids for use as food supplement or as medicament
US15/040,260 US20170029368A1 (en) 2008-07-15 2016-02-10 Novel sulphur containing lipids for use as food supplement or as medicament

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US14/263,793 Continuation US20140316002A1 (en) 2008-07-15 2014-04-28 Novel sulphur containing lipids for use as food supplement or as medicament

Publications (1)

Publication Number Publication Date
US20170029368A1 true US20170029368A1 (en) 2017-02-02

Family

ID=40419454

Family Applications (3)

Application Number Title Priority Date Filing Date
US13/054,212 Active 2030-12-12 US8759558B2 (en) 2008-07-15 2009-07-13 Sulphur containing lipids for use as food supplement or as medicament
US14/263,793 Abandoned US20140316002A1 (en) 2008-07-15 2014-04-28 Novel sulphur containing lipids for use as food supplement or as medicament
US15/040,260 Abandoned US20170029368A1 (en) 2008-07-15 2016-02-10 Novel sulphur containing lipids for use as food supplement or as medicament

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US13/054,212 Active 2030-12-12 US8759558B2 (en) 2008-07-15 2009-07-13 Sulphur containing lipids for use as food supplement or as medicament
US14/263,793 Abandoned US20140316002A1 (en) 2008-07-15 2014-04-28 Novel sulphur containing lipids for use as food supplement or as medicament

Country Status (29)

Country Link
US (3) US8759558B2 (en)
EP (2) EP2147910A1 (en)
JP (1) JP5628164B2 (en)
KR (2) KR101901836B1 (en)
CN (1) CN102159200B (en)
AU (1) AU2009271791B2 (en)
BR (1) BRPI0916456B8 (en)
CA (1) CA2730958C (en)
CL (2) CL2011000090A1 (en)
CO (1) CO6341548A2 (en)
DK (1) DK2313090T3 (en)
EA (1) EA022593B1 (en)
ES (1) ES2573977T3 (en)
HK (1) HK1161078A1 (en)
HR (1) HRP20160557T1 (en)
HU (1) HUE029195T2 (en)
IL (1) IL210673A (en)
IN (1) IN2011CH01039A (en)
MA (1) MA33083B1 (en)
MX (1) MX2011000556A (en)
MY (1) MY153620A (en)
NZ (2) NZ601501A (en)
PH (1) PH12011500113A1 (en)
PL (1) PL2313090T3 (en)
SG (1) SG192525A1 (en)
UA (1) UA109252C2 (en)
VN (1) VN28144A1 (en)
WO (1) WO2010008299A1 (en)
ZA (1) ZA201100666B (en)

Families Citing this family (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2217224B1 (en) 2007-11-09 2019-05-08 Basf As Lipid compounds for use in cosmetic products, as food supplement or as a medicament
EP2147910A1 (en) 2008-07-15 2010-01-27 Pronova BioPharma Norge AS Novel lipid compounds
DK2427415T3 (en) 2009-05-08 2019-06-11 Basf As Polyunsaturated fatty acids for the treatment of diseases of cardiovascular, metabolic and inflammatory disease areas
EP2571974B1 (en) 2010-05-21 2021-06-23 Coors Brewing Company Preparation of hop acids and their derivatives
US8530413B2 (en) 2010-06-21 2013-09-10 Sanofi Heterocyclically substituted methoxyphenyl derivatives with an oxo group, processes for preparation thereof and use thereof as medicaments
TW201215387A (en) 2010-07-05 2012-04-16 Sanofi Aventis Spirocyclically substituted 1,3-propane dioxide derivatives, processes for preparation thereof and use thereof as a medicament
TW201221505A (en) 2010-07-05 2012-06-01 Sanofi Sa Aryloxyalkylene-substituted hydroxyphenylhexynoic acids, process for preparation thereof and use thereof as a medicament
TW201215388A (en) 2010-07-05 2012-04-16 Sanofi Sa (2-aryloxyacetylamino)phenylpropionic acid derivatives, processes for preparation thereof and use thereof as medicaments
CN103298469A (en) 2010-08-31 2013-09-11 首尔大学校产学协力财团 Use of the fetal reprogramming of a PPAR delta agonist
GB201014633D0 (en) 2010-09-02 2010-10-13 Avexxin As Rheumatoid arthritis treatment
AU2011324909B2 (en) * 2010-11-05 2016-09-08 Pronova Biopharma Norge As Methods of treatment using lipid compounds
WO2013037390A1 (en) 2011-09-12 2013-03-21 Sanofi 6-(4-hydroxy-phenyl)-3-styryl-1h-pyrazolo[3,4-b]pyridine-4-carboxylic acid amide derivatives as kinase inhibitors
WO2013045413A1 (en) 2011-09-27 2013-04-04 Sanofi 6-(4-hydroxy-phenyl)-3-alkyl-1h-pyrazolo[3,4-b]pyridine-4-carboxylic acid amide derivatives as kinase inhibitors
KR101390589B1 (en) * 2012-05-30 2014-04-30 가천대학교 산학협력단 Composition for preventing or treating diabetes comprising diamines
JP6537980B2 (en) 2013-02-28 2019-07-03 プロノヴァ・バイオファーマ・ノルゲ・アーエスPronova BioPharma Norge AS Composition comprising lipid compound, triglyceride and surfactant, and method of use thereof
GB201313238D0 (en) 2013-07-24 2013-09-04 Avexxin As Process for the preparation of a polyunsaturated ketone compound
GB201501144D0 (en) * 2015-01-23 2015-03-11 Avexxin As Process for the preparation of a polyunsaturated ketone compound
WO2016173923A1 (en) 2015-04-28 2016-11-03 Pronova Biopharma Norge As Use of structurally enhanced fatty acids containing sulphur for preventing and/or treating non-alcoholic steatohepatitis
GB201604316D0 (en) 2016-03-14 2016-04-27 Avexxin As Combination therapy
WO2018055062A1 (en) 2016-09-21 2018-03-29 Avexxin As Pharmaceutical composition
KR20200075815A (en) * 2017-10-23 2020-06-26 에피트래커, 인코포레이티드 Fatty acid analogs and their use in the treatment of metabolic syndrome related conditions
MX2020005621A (en) 2017-12-06 2020-08-20 Basf As Fatty acid derivatives for treating non-alcoholic steatohepatitis.
CA3099561A1 (en) * 2018-05-16 2019-11-21 Epitracker, Inc. Compositions and methods for diagnosis and treatment of conditions related to aging
JP2021526552A (en) * 2018-05-23 2021-10-07 ノースシー セラピューティクス ベスローテン フェンノートシャップ Structurally modified fatty acids for improving glycemic control and treating inflammatory bowel disease
EP3972693A1 (en) * 2019-05-20 2022-03-30 T Omega AS Fatty acid compounds for prevention and treatment of neurodegenerative disorders
WO2021124272A1 (en) * 2019-12-19 2021-06-24 Liminal Biosciences Limited Cycloalkyl-containing carboxylic acids and uses thereof

Family Cites Families (69)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2909554A (en) * 1956-07-23 1959-10-20 Monsanto Chemicals Process for the manufacture of (alkylmercapto) alkyl sulfates
GB1038723A (en) 1962-03-26
JPS4839001B1 (en) * 1970-11-09 1973-11-21
US4040781A (en) * 1974-06-06 1977-08-09 Lever Brothers Company Novel 2-(alkylsulfinyl)ethyl sulfates and compositions employing same
US4032564A (en) * 1974-09-09 1977-06-28 Zoecon Corporation Esters of cyclopropylalkanols
GB1523276A (en) 1974-09-20 1978-08-31 Lafon Labor Sulphur-containing amino compounds
US4009211A (en) * 1975-07-29 1977-02-22 Gulf Research & Development Company Beta,beta-dialkylethylmercaptoethoxylate as new compounds
US4209410A (en) * 1976-04-28 1980-06-24 Phillips Petroleum Company Lubricants
EP0002007B1 (en) 1977-11-11 1982-07-28 Ciba-Geigy Ag Pyridine dicarboxylic acid derivatives, their mixtures with metallic stabilisers and their use in stabilising chlorine-containing thermoplasts
JPS5570841A (en) * 1978-11-24 1980-05-28 Konishiroku Photo Ind Co Ltd Forming method of dye image
US4368190A (en) * 1980-04-17 1983-01-11 Merck & Co., Inc. Immunologically active dipeptidyl 4-O-,6-O-acyl-2-amino-2-deoxy-D-glucose derivatives and methods for their preparation
EP0050327B1 (en) 1980-10-21 1984-06-20 Roche Diagnostics GmbH Phospholipids that contain sulphur, process for their preparation and medicines containing these compounds
US4411808A (en) * 1982-08-04 1983-10-25 Exxon Research & Engineering Co. Multifunctional additive for power transmission shift fluids
US4775223A (en) 1984-09-20 1988-10-04 Canon Kabushiki Kaisha Lactic acid derivative, liquid crystal composition containing same and liquid crystal device
CA2010000A1 (en) 1989-04-07 1990-10-07 Paul B. Merkel Photographic recording material containing a cyan dye-forming coupler
JPH0451149A (en) * 1990-06-19 1992-02-19 Fuji Photo Film Co Ltd Silver halide color photographic sensitive material
FR2663928B1 (en) 1990-06-27 1994-04-08 Norsolor NOVEL SULFUR ACRYLIC COMPOUNDS, A PROCESS FOR THEIR PREPARATION AND THEIR APPLICATION TO THE SYNTHESIS OF NEW POLYMERS.
JPH0543529A (en) * 1991-08-10 1993-02-23 Taisho Pharmaceut Co Ltd Alkaneamide ammonium compound
US5328952A (en) * 1992-02-14 1994-07-12 Rohm And Haas Company Multi-stage polymer latex cement modifier and process of making
JP2793458B2 (en) * 1992-03-19 1998-09-03 三井化学株式会社 Polyamide resin composition for connector and connector
JP2755279B2 (en) * 1992-03-19 1998-05-20 三井化学株式会社 Thermoplastic resin composition and molded article thereof
DE69431596D1 (en) * 1993-06-10 2002-11-28 Wake Forest University Winston (PHOSPHO) LIPIDS TO COMBAT HEPATITIS B INFECTION
JP3110918B2 (en) * 1993-06-18 2000-11-20 富士写真フイルム株式会社 Silver halide photographic material
GB2290789B (en) * 1994-07-01 1998-09-16 Ciba Geigy Ag Titanium and zirconium complexes of carboxylic acids as corrosion inhibitors
KR100463709B1 (en) * 1994-10-13 2005-08-04 위민스 앤드 칠드런스 호스피털 애드레이드 Modified polyunsaturated fatty acids
US7517858B1 (en) * 1995-06-07 2009-04-14 The Regents Of The University Of California Prodrugs of pharmaceuticals with improved bioavailability
FR2741619B1 (en) * 1995-11-28 1998-02-13 Pf Medicament NOVEL 2,3,5-TRIMETHYL-4-HYDROXY-ANILIDES DERIVATIVES, THEIR PREPARATION AND THEIR THERAPEUTIC APPLICATION
JP2000508645A (en) 1996-04-12 2000-07-11 ペプテック リミテッド Method for treating immune diseases using polyunsaturated fatty acids
US6060515A (en) * 1997-01-24 2000-05-09 The Regents Of The University Of California Treatment of skin conditions by use of PPARα activators
WO1999058120A1 (en) * 1998-05-08 1999-11-18 Rolf Berge USE OF NON-β-OXIDIZABLE FATTY ACID ANALOGUES FOR TREATMENT OF SYNDROME-X CONDITIONS
FR2786187B1 (en) * 1998-11-19 2001-11-09 Univ Paris Curie 2-ACYLAMINO-2-DEOXY-GLUCONO-1,5-LACTONE TYPE COMPOUNDS, PROCESS FOR OBTAINING SAME, COMPOSITIONS COMPRISING SAME AND USES THEREOF
FR2792312B1 (en) * 1999-04-15 2001-06-08 Oreal THIA-ALCYNOIC (POLY) COMPOUNDS AND DERIVATIVES THEREOF, COMPOSITIONS COMPRISING SAME AND THEIR USE
JP2003500459A (en) 1999-06-01 2003-01-07 ザ・ユニバーシティ・オブ・テキサス・サウスウエスタン・メディカル・センター Substituted biaryl ether compounds
US6723717B1 (en) * 1999-06-01 2004-04-20 The University Of Texas Southwestern Medical Center Sulfur-containing thyroxane derivatives and their use as hair growth promotors
NO328803B1 (en) * 2000-03-03 2010-05-18 Thia Medica New fatty acid analogues
UA75083C2 (en) 2000-06-22 2006-03-15 Тераванс, Інк. Derivatives of glycopeptidephosphonates
FR2828487B1 (en) 2001-08-09 2005-05-27 Genfit S A NOVEL COMPOUNDS DERIVED FROM FATTY ACIDS, PREPARATION AND USES
EP1458354A1 (en) * 2001-12-21 2004-09-22 3M Innovative Properties Company Medicinal aerosol formulations comprising ion pair complexes
GB2383355A (en) * 2001-12-22 2003-06-25 Schlumberger Holdings An aqueous viscoelastic fluid containing hydrophobically modified polymer and viscoelastic surfactant
AU2003206762C1 (en) * 2002-01-31 2009-01-15 Tfl Ledertechnik Gmbh Compositions and its use for imparting water repellency to leather or furskins, textiles and other fibrous materials
US7273852B2 (en) * 2002-06-13 2007-09-25 The Research Foundation Of The City University Of New York Synthetic C-glycolipid and its use for treating cancer, infectious diseases and autoimmune diseases
KR20050061400A (en) * 2002-06-20 2005-06-22 아이씨 벡 리미티드 Sulfur-containing phospholipid derivatives
FR2845991B1 (en) * 2002-10-16 2005-02-04 Pf Medicament ALPHA-PHENYL ACETANILIDE DERIVATIVES AND THEIR USE IN HUMAN THERAPEUTICS
US8372430B2 (en) * 2002-12-17 2013-02-12 The Procter & Gamble Company Compositions, methods, and kits useful for the alleviation of gastrointestinal effects
WO2004076404A1 (en) * 2003-02-28 2004-09-10 Kaneka Corpration Processes for the production of optically active compounds having substituents at the 2-position
DE10326303A1 (en) * 2003-06-11 2004-12-30 Celares Gmbh Reagents for modifying biopharmaceuticals, their manufacture and use
AU2005209331A1 (en) 2004-01-30 2005-08-11 Peplin Biolipids Pty Ltd Therapeutic and carrier molecules
JP4563114B2 (en) 2004-08-30 2010-10-13 出光興産株式会社 Additive for lubricant
US20090123507A1 (en) 2005-03-08 2009-05-14 Reinhold Ohrlein Metal Oxide Nanoparticles Coated With Specific N-Acylaminomethylene Phosphonates
EP1888727B1 (en) * 2005-05-04 2015-04-15 Pronova BioPharma Norge AS New dha derivatives and their use as medicaments
EP2004169B1 (en) 2006-04-12 2012-08-08 Unilever PLC Oral composition comprising a polyunsaturated fatty acid and salicylic acid for obtaining an antiinflammatory effect in skin
EP1849449A1 (en) * 2006-04-26 2007-10-31 3M Innovative Properties Company Filler containing composition and process for production and use thereof
US7763607B2 (en) * 2006-04-27 2010-07-27 Solvay Pharmaceuticals Gmbh Pharmaceutical compositions comprising CBx cannabinoid receptor modulators and potassium channel modulators
US20080075692A1 (en) * 2006-05-09 2008-03-27 Perrine Susan P Methods for treating blood disorders
JP5552313B2 (en) 2006-11-01 2014-07-16 プロノヴァ・バイオファーマ・ノルゲ・アーエス Lipid compounds
WO2008053340A1 (en) * 2006-11-03 2008-05-08 Pronova Biopharma Norge As A combination product comprising at least one lipid substituted in the alpha position and at least one hypoglycemic agent
CN101225064A (en) 2007-01-19 2008-07-23 上海汇瑞生物科技有限公司 New method for preparing neta-thia-alpha-alkyl fatty acid
DE102007017179A1 (en) 2007-04-12 2008-10-23 Clariant International Ltd. Process for the preparation of Alkylpolyglykolcarbonsäuren and Polyglykoldicarbonsäuren by direct oxidation
EP2217224B1 (en) * 2007-11-09 2019-05-08 Basf As Lipid compounds for use in cosmetic products, as food supplement or as a medicament
WO2009149496A1 (en) 2008-06-10 2009-12-17 Central Northern Adelaide Health Service Treatment of diabetes and complications thereof and related disorders
FR2933006B1 (en) 2008-06-27 2010-08-20 Inst Francais Du Petrole ABSORBENT SOLUTION CONTAINING SULFUR DEGRADATION INHIBITOR WITH CARBOXYL GROUPING AND METHOD FOR LIMITING THE DEGRADATION OF AN ABSORBENT SOLUTION
CA2729186C (en) 2008-07-08 2018-01-16 Catabasis Pharmaceuticals, Inc. Fatty acid acetylated salicylates and their uses
EP2147910A1 (en) 2008-07-15 2010-01-27 Pronova BioPharma Norge AS Novel lipid compounds
DK2427415T3 (en) * 2009-05-08 2019-06-11 Basf As Polyunsaturated fatty acids for the treatment of diseases of cardiovascular, metabolic and inflammatory disease areas
US8304551B2 (en) * 2009-09-01 2012-11-06 Catabasis Pharmaceuticals, Inc. Fatty acid niacin conjugates and their uses
WO2011089529A1 (en) 2010-01-20 2011-07-28 Pronova Biopharma Norge As Salicylate fatty acid derivatives
AU2011324909B2 (en) 2010-11-05 2016-09-08 Pronova Biopharma Norge As Methods of treatment using lipid compounds
WO2012115695A1 (en) 2011-02-25 2012-08-30 Catabasis Pharmaceuticals, Inc. Bis-fatty acid conjugates and their uses
WO2013016531A2 (en) 2011-07-26 2013-01-31 Purdue Research Foundation Compounds and methods for use in treating neoplasia and cancer

Also Published As

Publication number Publication date
UA109252C2 (en) 2015-08-10
NZ590645A (en) 2012-08-31
AU2009271791A1 (en) 2010-01-21
IL210673A (en) 2015-02-26
HK1161078A1 (en) 2012-08-24
JP5628164B2 (en) 2014-11-19
MX2011000556A (en) 2011-03-15
US20110190395A1 (en) 2011-08-04
EP2313090A1 (en) 2011-04-27
CL2018001111A1 (en) 2018-07-20
ZA201100666B (en) 2011-10-26
EA201170193A1 (en) 2011-08-30
SG192525A1 (en) 2013-08-30
CL2011000090A1 (en) 2012-03-30
EP2313090A4 (en) 2012-01-18
MY153620A (en) 2015-02-27
AU2009271791B2 (en) 2014-06-26
IN2011CH01039A (en) 2011-12-30
US8759558B2 (en) 2014-06-24
JP2011528350A (en) 2011-11-17
HRP20160557T1 (en) 2016-06-17
US20140316002A1 (en) 2014-10-23
ES2573977T3 (en) 2016-06-13
EP2313090B1 (en) 2016-03-30
HUE029195T2 (en) 2017-02-28
WO2010008299A1 (en) 2010-01-21
CN102159200B (en) 2015-06-24
CA2730958C (en) 2016-07-05
EA022593B1 (en) 2016-01-29
KR20110031497A (en) 2011-03-28
AU2009271791A2 (en) 2011-02-24
PH12011500113A1 (en) 2010-01-21
KR101901836B1 (en) 2018-09-27
BRPI0916456A2 (en) 2018-06-12
CA2730958A1 (en) 2010-01-21
CO6341548A2 (en) 2011-11-21
EP2147910A1 (en) 2010-01-27
IL210673A0 (en) 2011-03-31
CN102159200A (en) 2011-08-17
BRPI0916456B8 (en) 2021-05-25
BRPI0916456B1 (en) 2020-12-29
DK2313090T3 (en) 2016-06-13
PL2313090T3 (en) 2016-09-30
MA33083B1 (en) 2012-03-01
NZ601501A (en) 2014-03-28
VN28144A1 (en) 2011-12-26
KR20170093253A (en) 2017-08-14

Similar Documents

Publication Publication Date Title
US20170029368A1 (en) Novel sulphur containing lipids for use as food supplement or as medicament
US8735436B2 (en) Polyunsaturated fatty acids for the treatment of diseases related to cardiovascular, metabolic and inflammatory disease areas
US7550613B2 (en) Compounds
US8741966B2 (en) Lipid compounds for use in cosmetic products, as food supplement or as a medicament
US20100240616A1 (en) Novel lipid compounds
US20100267828A1 (en) dha derivatives and their use as medicaments
EP2248798A1 (en) Novel lipid compounds
AU2013205154A1 (en) Polyunsaturated fatty acids for the treatment of diseases related to cardiovascular, metabolic and inflammatory disease areas

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION