Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
AMINO ACID SALTS OF SATURATED FATTY ACIDS
Document Type and Number:
WIPO Patent Application WO/2017/079391
Kind Code:
A1
Abstract:
The present invention provides compounds of Formula I and Formula II, and related compositions and methods.

Inventors:
MATHIAS GARY (US)
SCIAVOLINO FRANK C (US)
Application Number:
PCT/US2016/060256
Publication Date:
May 11, 2017
Filing Date:
November 03, 2016
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
THETIS PHARMACEUTICALS LLC (US)
International Classes:
C07C229/76; A61K31/16; A61P25/08; C07K5/068
Domestic Patent References:
WO2014011895A22014-01-16
Other References:
PISHAN CHANG ET AL: "Seizure control by ketogenic diet-associated medium chain fatty acids", NEUROPHARMACOLOGY, vol. 69, 1 June 2013 (2013-06-01), pages 105 - 114, XP055075887, ISSN: 0028-3908, DOI: 10.1016/j.neuropharm.2012.11.004
CHANG ET AL., NEUROPHARMACOLOGY, vol. 69, 2013, pages 105 - 114
Attorney, Agent or Firm:
LIBERTO, Muriel et al. (US)
Download PDF:
Claims:
What is claimed is:

A compound of Formula I, or an enantiomer, polymorph, solvate, or hydrate thereof:

Formula I

wherein

Ri is H when A is absent, or absent when A is present,

R2 is H or absent,

Xi and X2 may be the same or different and are each the side chain of an amino acid residue,

A+ is present or absent and is a monovalent metal cation, or a non-metal molecule having at least one basic function, and

B- is a saturated fatty acid molecule and n is 1 or 2.

The compound of claim 1, wherein A is absent and Ri is H.

3. The compound of claim 1, wherein Ri is absent, and A+ is a monovalent metal cation, or a monovalent amine-based cation, and n is i .

4. The compound of claim 3, wherein the monovalent metal cation is selected from Na+ and K+.

5. The compound of claim 2, wherein Xi and X2 are each the side chain of lysine and n is 2.

6. The compound of any one of claims 1-5, wherein the fatty acid is a medium chain or a long chain saturated fatty acid.

7. The compound of claim 6, wherein the medium chain saturated fatty acid is a (C5- Cii) fatty acid.

8. The compound of claim 6, wherein the long chain saturated fatty acid is a (C12-C24) fatty acid.

9. The compound of claim 8, wherein the long chain saturated fatty acid is a (Ci6-C24) fatty acid selected from the stearic acid and palmitic acid.

10. A composition comprising a compound as defined in any one of claims 1-9, or mixtures thereof.

11. The composition of claim 10, wherein the composition is a pharmaceutical composition, optionally comprising a pharmaceutically acceptable carrier or excipient.

12. A unit dosage form comprising a compound or composition as defined in any of claims 1-9, or a mixture of two or more of the compounds, wherein the unit dosage form comprises from about 0.05 g to 12 g of total fatty acids in the fatty acid component of the compound or mixture.

13. The pharmaceutical composition of claim 12, wherein the composition further comprises one or more additional active pharmaceutical agents (APIs).

14. The pharmaceutical composition of claim 13, wherein the one or more additional APIs is selected from the group consisting of an antihyperlipidemic agent, an anti-diabetic agent, an anti -epileptic agent, and an anti-inflammatory agent, and combinations thereof.

15. A method for delivering saturated fatty acids in ionic form to a subject, the method comprising administering to the subject a compound as defined in any one of claims 1-9, or mixtures thereof.

16. A method for treating a disease or disorder responsive to treatment with a saturated fatty acid, the method comprising administering to a subject in need thereof a compound as defined in any one of claims 1-9, or mixtures thereof.

17. The method of claim 16, wherein the disease or disorder is selected from epilepsy or a metabolic disease or disorder.

18. The method of claim 17, wherein the metabolic disease or disorder is a fatty acid metabolism disorder, a long-chain fatty acid oxidation disorder, or a lipid metabolism disorder.

19. The method of claim 17, wherein the metabolic disease or disorder is abnormal glucose metabolism manifesting in diabetes, including type 2 diabetes, or pre-diabetes, insulin resistance, abnormal lipid metabolism manifesting as hypertriglyceridemia, i.e., elevated triglycerides, mixed dyslipidemia, hypercholesterolemia, fatty liver, and combined abnormal glucose and lipid metabolism manifesting in obesity; or a dyslipidemic disorder selected from hypertriglyceridemia, hypercholesterolemia and mixed dyslipidemias.

20. The method of claim 17, wherein the metabolic disease or disorder is

hypertriglyceridemia, severe hypertriglyceridemia, hypercholesterolemia, pre-diabetes, fatty liver disease, or obesity.

21. A compound of Formula II, or an enantiomer, polymorph, solvate, or hydrate thereof:

Formula II

wherein

M2+ is a divalent metal cation,

Xi and X2 may be the same or different and are each the side chain of an amino acid residue having a basic function,

A- and B- may be the same or different and are each a saturated fatty acid molecule. The compound of claim 21, wherein M+ is Mg+2, Ca+2, or Zn+2.

23. The compound of claim 22, wherein Xi and X2 are the same and are the side chain of lysine.

24. The compound of claim 23, wherein the fatty acid is a medium chain or a long chain saturated fatty acid.

25. The compound of claim 24, wherein the medium chain saturated fatty acid is a (C5- Cii) fatty acid.

26. The compound of claim 24, wherein the long chain saturated fatty acid is a (Ci2-C24) fatty acid.

27. A composition comprising a compound as defined in any one of claims 21-26, or mixtures thereof.

28. The composition of claim 27, wherein the composition is a pharmaceutical composition, optionally comprising a pharmaceutically acceptable carrier or excipient.

29. A unit dosage form comprising a compound or composition as defined in any of claims 21-26, or a mixture of two or more of the compounds, wherein the unit dosage form comprises from about 0.05 g to 12 g of total fatty acids in the fatty acid component of the compound or mixture.

30. The pharmaceutical composition of claim 28, wherein the composition further comprises one or more additional active pharmaceutical agents (APIs).

31. The pharmaceutical composition of claim 30, wherein the one or more additional APIs is selected from the group consisting of an antihyperlipidemic agent, an anti-diabetic agent, an anti -epileptic agent, and an anti-inflammatory agent, and combinations thereof.

32. A method for delivering saturated fatty acids in ionic form to a subject, the method comprising administering to the subject a compound as defined in any one of claims 21-26, or mixtures thereof.

33. A method for treating a disease or disorder responsive to treatment with a saturated fatty acid, the method comprising administering to a subject in need thereof a compound as defined in any one of claims 21-26, or mixtures thereof.

34. The method of claim 33, wherein the disease or disorder is selected from epilepsy or a metabolic disease or disorder.

35. The method of claim 34, wherein the metabolic disease or disorder is a fatty acid metabolism disorder, a long-chain fatty acid oxidation disorder, or a lipid metabolism disorder.

36. The method of claim 34, wherein the metabolic disease or disorder is abnormal glucose metabolism manifesting in diabetes, including type 2 diabetes, or pre-diabetes, insulin resistance, abnormal lipid metabolism manifesting as hypertriglyceridemia, i.e., elevated triglycerides, mixed dyslipidemia, hypercholesterolemia, fatty liver, and combined abnormal glucose and lipid metabolism manifesting in obesity; or a dyslipidemic disorder selected from hypertriglyceridemia, hypercholesterolemia and mixed dyslipidemias.

37. The method of claim 34, wherein the metabolic disease or disorder is

hypertriglyceridemia, severe hypertriglyceridemia, hypercholesterolemia, pre-diabetes, fatty liver disease, or obesity.

Description:
AMINO ACID SALTS OF SATURATED FATTY ACIDS

CROSS-REFERENCE TO RELATED APPLICATION

[01] The current application claims priority to U.S. Provisional patent application serial number 62/250,668, filed on November 4, 2015 and entitled "Amino Acid Salts of Saturated Fatty Acids," the contents of which are incorporated by reference herein in its entirety.

FIELD OF THE INVENTION

[02] The present invention relates to compounds which are amino acid and peptide salts of saturated fatty acids, compositions comprising same, and methods for their manufacture and use.

BACKGROUND

[03] Medium and long length fatty acids show promise in the treatment of diseases and disorders such as epilepsy and long-chain fatty acid oxidation disorders (LC-FAOD). LC- FAODs are a group of autosomal recessive genetic disorders characterized by metabolic deficiencies in which the body is unable to convert long-chain fatty acids into energy. The inability to produce energy from fat can lead to severe depletion of glucose in the body, and serious liver, muscle, and heart disease, which can lead to hospitalizations or early death.

[04] Certain medium length fatty acids may be useful as a substitute for a ketogenic diet for the treatment of drug resistant epilepsy. See Chang et al, Neuropharmacology 69 (2013): 105-114.

[05] LC-FAOD is currently treated with the avoidance of fasting, low-fat/high

carbohydrate diets, carnitine, and medium-chain triglyceride (MCT) oils. In addition, triheptanoin is a synthetic triglyceride in development for LC-FAOD.

[06] There is a need to develop new compositions able to deliver fatty acids at much higher plasma levels than is possible using the currently available free fatty acid, sodium salt, or ester forms, in order to fulfill the therapeutic and nutritional promise of these compounds and translate the many promising in vitro and cellular pharmacology observations into clinical and general health benefits. The present invention addresses these needs. SUMMARY OF THE INVENTION

[07] The disclosure provides compounds of Formula I and Formula II, including enantiomers, polymorphs, solvates, and hydrates thereof, and mixtures thereof, as well as methods of making the compounds, compositions comprising same, and methods for their use. The compounds are chemically and physically stable solid materials, e.g., powders. In embodiments, the compounds also have low hygroscopicity and are highly water soluble. The compounds described here offer improved means for delivering fatty acids to a subject. In embodiments, the compounds may be useful for delivering medium chain fatty acids for treatment of epilepsy as a substitute to the ketogenic diet, including e.g., caprylic acid, lauric acid, and valproic acid. In embodiments, the compounds may be useful for delivering medium chain fatty acids, including odd-chain fatty acids, for the treatment of LC-FAOD.

Compounds of Formula I have the structure:

Formula I

wherein

Ri is H when A is absent, or absent when A is present;

R 2 is H or absent;

X 1 and X 2 may be the same or different and are each the side chain of an amino acid residue,

A+ is present or absent and is a monovalent metal cation, or a non-metal molecule having at least one basic functionality, and

B- is a saturated fatty acid molecule and n is 1 or 2.

[09] A compound of Formula I consists of at least (i) a dipeptide component and (ii) a fatty acid component (B-), with an optional basic moiety (A+). In embodiments, B- may be present as a single fatty acid molecule, n=l, or two fatty acid molecules, n=2. The dipeptide component contains X 1 and X 2 which are each the side chain of an amino acid residue. In embodiments, the amino acid is selected from lysine, arginine, histidine, aspartate, glutamate, serine, threonine, asparagine, glutamine, cysteine, glycine, proline, alanine, valine, isoleucine, leucine, methionine, phenylalanine, tyrosine, and tryptophan. In embodiments, X 1 and X 2 are the same. In embodiments, at least one of Xi or X 2 contains a basic function. In embodiments, X 1 and X 2 are the same and each is the side chain of lysine.

[10] A compound of Formula I may also optionally contain a molecule (A + ) having at least one basic function which forms an ionic bond with the terminal carboxy of the amino acid component. In embodiments, A + is a monovalent metal cation, e.g., Na + , K + , or a molecule having at least one basic functionality, such as a monovalent amine-based cation, e.g., tri- ethanolamine, or tri-ethylamine, or a basic pharmaceutical compound.

[11] As described in more detail below, the compounds of Formula I encompass simple salts of dipeptides and a fatty acid (Formula IA), simple metal salts of the dipeptides and a fatty acid with a monovalent metal (Formula IB), and simple non-metal salts of the dipeptides and a fatty acid with a non-metal molecule having at least one basic functionality (Formula IC).

[12] Compounds of Formula II have the structure:

Formula II

wherein

M is a divalent metal cation,

X 1 and X 2 may be the same or different and are each the side chain of an amino acid residue having a basic function,

A- and B- may be the same or different and are each a saturated fatty acid molecule.

[13] Compounds of Formula II have two amino acids coordinated around a divalent metal cation as the amino acid component and two fatty acid molecules as the fatty acid component. In embodiments, the divalent metal cation is Mg +2 , Ca +2 , or Zn +2 . In embodiments, the amino acid component comprises or consists of lysine.

[14] The fatty acid component of the compounds described here may be selected from a naturally occurring, non-naturally occurring, branched or unbranched saturated fatty acid having from about 5 carbon atoms to about 24 carbon atoms. In embodiments, the fatty acid is a medium chain fatty acid, which in this context refers to a C 5 - Cn carbon chain, or a long chain fatty acid, which refers to a C 12 - C 24 carbon chain.

[15] In embodiments, the fatty acid component is a medium chain fatty acid such as valproic acid, caproic acid (hexanoic acid), caprylic acid (octanoic acid), or capric acid (decanoic acid), or a long chain fatty acid such as lauric acid (dodecanoic acid), or a derivative thereof. Derivatives may include odd-chain fatty acids such as triheptanoin. In embodiments, the fatty acid component comprises or consists of palmitic acid or stearic acid.

[16] The disclosure also provides pharmaceutical compositions and dosage forms comprising or consisting of any of the compounds described herein, and mixtures thereof. In one embodiment, the dosage form is a solid dosage form. In embodiments, the solid dosage form is a powder, a tablet, a capsule, or a caplet. In embodiments, the solid dosage form, for example a powder, may be suitable for reconstitution in an aqueous vehicle, for example to be administered intraperitoneally.

[17] In embodiments, the pharmaceutical compositions or dosage forms further comprise one or more optional excipients as described infra. In embodiments, the pharmaceutical composition or dosage form further comprises one or more additional active pharmaceutical agents (APIs). In embodiments, the one or more additional APIs is selected from the group consisting of an anti-hyperlipidemic agent, an anti-diabetic agent, an anti-epileptic agent, and an anti-inflammatory agent, and combinations thereof.

[18] The disclosure also provides a food additive or dietary supplement comprising a compound described herein, and mixtures thereof, optionally comprising a carrier suitable for administration to a human or non-human animal. In embodiments, the food additive or dietary supplement further comprises one or more additional biologically active agents. In embodiments, the one or more additional biologically active agents is selected from the group consisting of a vitamin, a mineral, an amino acid, a carbohydrate, an antioxidant, a flavonoid, a carotenoid, a phytoseterol, an herb, an enzyme, a botanical extract or concentrate, and a botanical compound, and combinations thereof. In embodiments, the one or more additional biologically active agents is selected from the group consisting of vitamin A, vitamin Bl, vitamin B 12, vitamin B6, vitamin C, vitamin D, vitamin E, vitamin K, calcium, carnitine, chromium, chondroitin, coenzyme Q10 (ubiquinone), folate, glucosamine, metafolin, riboflavin, biotin, iodine, iron, magnesium, selenium, thiamin, and zinc, and combinations thereof. [19] The disclosure also provides unit dosage forms comprising the compounds or mixtures of the compounds described here. In embodiments, the unit dosage form comprises from about 0.05 g to 12 g of fatty acids total as the fatty acid component of the compound or mixture of compounds in the unit dosage form.

[20] The disclosure also provides pharmaceutical and non-pharmaceutical uses of the compounds and mixtures of compounds described herein. In embodiments, a compound or composition comprising same is useful for delivering the fatty acids of the fatty acid component as free fatty acids, or mixtures of two or more different free fatty acids, in ionic form to a subject. In embodiments, a compound or composition of the invention is also useful for delivering, along with the free fatty acids, or mixture of free fatty acids in ionic form, at least one additional API or biologically active agent to a subject. In embodiments, the free fatty acids delivered are medium or long chain fatty acids as described above and infra. In embodiments, the at least one additional API or biologically active agent is as described above and infra.

[21] In embodiments, a compound or composition of the disclosure is useful for treating a disease or disorder responsive to treatment with a saturated fatty acid. In embodiments, at least 50 wt%, at least 60 wt%, at least 70 wt%, or at least 90 wt% of the fatty acid component of the compound or composition consists of one or more saturated fatty acids selected from valproic acid, caproic acid (hexanoic acid), caprylic acid (octanoic acid), or capric acid (decanoic acid), and lauric acid (dodecanoic acid), or a derivative thereof, including e.g., triheptanoin .

[22] In embodiments, the disease or disorder is selected from epilepsy, pediatric epilepsy, and epilepsy or pediatric epilepsy that is resistant to standard treatment, such as treatment with valproic acid.

[23] In embodiments, the disease or disorder is a fatty acid metabolism disorder, a long- chain fatty acid oxidation disorder, or a lipid metabolism disorder.

[24] In embodiments, the disease or disorder is a metabolic disorder selected from the group consisting of abnormal glucose metabolism manifesting in diabetes or pre-diabetes, abnormal lipid metabolism manifesting as hypertriglyceridemia, i.e., elevated triglycerides, mixed dyslipidemia, fatty liver, and combined abnormal glucose and lipid metabolism manifesting in obesity. In one embodiment, a composition of the invention is used in a method for treating a disease or disorder selected from diabetes, pre-diabetes,

hypertriglyceridemia, dyslipidemia, fatty liver, and obesity. [25] In embodiments, the disclosure provides a method for treating epilepsy or epileptic syndrome, the method comprising administering to a subject in need of such treatment, a compound described herein, or a composition comprising same. In embodiments, the fatty acid component consists of a saturated fatty acid selected from valproic acid, caprylic acid, and 1 auric acid.

[26] In embodiments, the disclosure provides a method for treating a fatty acid metabolism disorder, a long-chain fatty acid oxidation disorder, or a lipid metabolism disorder, the method comprising administering to a subject in need of such treatment, a compound of described herein, or a composition comprising same. In embodiments, the fatty acid component consists of a medium chain fatty acid.

[27] The disclosure also provides for the use of a compound of described herein as a food additive or dietary supplement. In embodiments, the use is to counter a dietary deficiency or nutritional disorder in a subject, or in a method for maintaining, promoting, or improving the general health of a subject. Accordingly, the invention provides methods of countering a dietary deficiency or nutritional disorder in a subject, as well as methods for maintaining, promoting, or improving the general health of a subject, the methods comprising

administering to the subject a compound of Formula I, or a composition or unit dosage form comprising same, or a composition comprising a mixture of two or more of said compounds. In embodiments, the composition comprises from 50 mg to 6 g of total fatty acids, as the fatty acid component of the compounds in the composition.

DETAILED DESCRIPTION OF THE INVENTION

[28] The disclosure provides compounds of Formula I and Formula II and related compositions as well as methods for making the compounds and methods for their use. As described in more detail in the following sections, the disclosure provides compositions comprising one or more of the compounds described herein, and mixtures thereof. Also provided are compositions further comprising a second active agent. In embodiments the second active agent is a biologically active agent or an active pharmaceutical ingredient (API), as described herein.

[29] In embodiments, the composition is a pharmaceutical composition. In other embodiments, the composition is a dietary additive or supplement. In embodiments, mixtures of two or more of the compounds described here are prepared by physical admixture of the compounds in the desired proportions. [30] The disclosure also provides a package or kit comprising a unit dosage form of a compound or composition described herein, at least one container for holding the unit dosage form, and instructions for use.

[31] The compounds of Formula I each contain at least (i) an amino acid component, and (ii) a fatty acid component (B-). The compounds may also optionally contain a molecule (A+) having at least one basic function. In embodiments A+ is a monovalent metal cation, or a non-metal molecule having at least one basic function. In embodiments the non-metal compound is a primary amine, a secondary amine, a tertiary amine, or a guanidine. In one embodiment, the non-metal compound is tri-ethanolamine. The non-metal compound may also be a therapeutic agent having a basic functionality.

[32] The compounds described here advantageously provide the fatty acid component in the form of a chemically and physically stable solid material, such as a powder. The fatty acid component of the compounds described herein is both water soluble and stable against oxidative degradation. These compounds thus provide considerable advantages over fatty acid compositions which are in the physical form of an oily liquid which is relatively difficult to formulate and chemically susceptible to degradation, especially oxidative degradation. The physical properties of the compounds of the present disclosure are described in more detail infra.

Compounds of Formula I

[33] In embodiments, the invention provides a compound of Formula I, and enantiomers, polymorphs, solvates, and hydrates thereof:

Formula I

wherein

Ri is H when A is absent, or absent when A is present;

R 2 is H or absent;

X 1 and X 2 may be the same or different and are each the side chain of an amino acid residue, A+ is present or absent and is a monovalent metal cation, or a non-metal molecule having at least one basic function, and

B- is a saturated fatty acid molecule and n is 1 or 2.

[34] A compound of Formula I consists of at least (i) a dipeptide component and (ii) a fatty acid component (B-), with an optional basic moiety (A+). In embodiments, B- may be present as a single fatty acid molecule, n=l, or two fatty acid molecules, n=2. The dipeptide component contains X 1 and X 2 which are each the side chain of an amino acid residue. In embodiments, the amino acid is selected from lysine, arginine, histidine, aspartate, glutamate, serine, threonine, asparagine, glutamine, cysteine, glycine, proline, alanine, valine, isoleucine, leucine, methionine, phenylalanine, tyrosine, and tryptophan. In embodiments, X 1 and X 2 are the same. In embodiments, X 1 and X 2 are the same and each is the side chain of lysine.

[35] A compound of Formula I may also optionally contain a molecule (A + ) having at least one basic function which forms an ionic bond with the terminal carboxy of the amino acid component. In embodiments, A + is a monovalent metal cation, e.g., Na + , K + , or a molecule having at least one basic functionality, such as a monovalent amine-based cation, e.g., tri- ethanolamine, or tri-ethylamine, or a basic pharmaceutical compound.

[36] As described in more detail below, the compounds of Formula I encompass simple salts of dipeptides and a fatty acid (Formula IA), simple metal salts of the dipeptides and a fatty acid with a monovalent metal (Formula IB), and simple non-metal salts of the dipeptides and a fatty acid with a non-metal molecule having at least one basic functionality (Formula IC).

[37] The following non-limiting examples of compounds of Formula I is provided to illustrate the nature of the compounds described and is not intended to limit the disclosure to the particular compounds depicted.

A compound of Formula I wherein

Ri and R 2 are each H,

X 1 and X 2 are each H,

A+ is absent,

B- is a saturated fatty acid,

and enantiomers, polymorphs, solvates, and hydrates thereof:

Formula IA-1 (Gly-Gly-FA).

[38] A compound of Formula I wherein

Ri and R 2 are each H,

X 1 is the side chain of lysine (butylamine),

X 2 is the side chain of valine (isopropyl),

A+ is absent,

B- is a saturated fatty acid,

and enantiomers, polymorphs, solvates, and hydrates thereof:

Formula IA-2 (Lys-Val-FA).

[39] A compound of Formula I wherein

Ri and R 2 are each H,

Xi is the side chain of lysine (butylamine),

X 2 is the side chain of serine,

A+ is absent,

B- is a saturated fatty acid,

and enantiomers, polymorphs, solvates, and hydrates thereof: Formula ΙΑ-3 (Lys-Ser-FA).

[40] A compound of Formula I wherein

Ri and R 2 are each H,

X 1 is the side chain of lysine (butylamine),

X 2 is the side chain of glycine

A+ is absent,

B- is a saturated fatty acid,

and enantiomers, polymorphs, solvates, and hydrates thereof:

Formula IA-4 (Lys-Gly-FA).

[41] A compound of Formula I wherein

Ri and R 2 are each H,

X 1 is the side chain of lysine (butylamine),

X 2 is the side chain of leucine (isobutyl)

A+ is absent,

B- is a saturated fatty acid,

and enantiomers, polymorphs, solvates, and hydrates thereof: Formula IA-5 (Lys-Leu-FA).

[42] A compound of Formula I wherein

Ri and R 2 are each H,

X 1 is the side chain of lysine (butylamine),

X 2 is the side chain of histidine (imidazole)

A+ is absent,

B- is a saturated fatty acid,

nd enantiomers, polymorphs, solvates, and hydrates thereof:

[43] In embodiments, A+ is a monovalent metal cation such as Na + or K + (Formula IB). Accordingly, the disclosure provides compounds of Formula IB including the following:

A compound of Formula I wherein

Ri is absent,

R 2 is H,

Xi and X 2 are each H,

A+ is Na + ,

B- is a saturated fatty acid Formula IB-1 (Na+ -Gly-Gly-FA)

[44] In embodiments, A+ is a non-metal molecule having at least one basic functionality, such as a monovalent amine-based cation, e.g., tri-ethanolamine, or tri-ethylamine or a basic pharmaceutical compound. Accordingly, the disclosure provides compounds of Formula IC including the following:

[45] A compound of Formula I wherein

Ri is absent,

R 2 is H,

Xi and X 2 are each H,

A+ is a non-metal and a molecule having at least one basic functionality,

B- is a saturated fatty acid,

and enantiomers, polymorphs, solvates, and hydrates thereof:

IC-1 (A+ -Gly-Gly-FA).

[46] A compound of Formula I wherein

Ri is absent,

R 2 is H,

Xi and X 2 are each H,

A+ is trienthanolamine,

B- is a saturated fatty acid,

and enantiomers, polymorphs, solvates, and hydrates thereof:

Formula IC-2 (Triethanolamine -Gly-Gly-FA).

[47] A compound of Formula I wherein

Ri is absent,

R 2 is H,

X 1 and X 2 are each H,

A+ is metformin,

B- is a saturated fatty acid,

and enantiomers, polymorphs, solvates, and hydrates thereof:

Formula IC-3 (Metformin -Gly-Gly-FA).

Compounds of Formula II

[48] In embodiments, the invention provides a compound of Formula II, and enantiomers, polymorphs, solvates, and hydrates thereof:

Formula II

wherein

M is a divalent metal cation, X 1 and X 2 may be the same or different and are each the side chain of an amino acid residue having a basic function,

A- and B- may be the same or different and are each a saturated fatty acid molecule.

[49] The following non-limiting example of a compound of Formula II is provided to illustrate the nature of the compounds described and is not intended to limit the disclosure to the particular compounds depicted.

A compound of Formula II wherein

Xi and X 2 are each the side chain of ly

Formula II- 1

The Fatty Acid Component of Formula I and Formula II Compounds

[50] The term "fatty acid" is used to describe a carboxylic acid with a long aliphatic carbon chain of from about 4 to 28 carbon atoms, which is either saturated or unsaturated, referring to whether the carbon chain contains one or more double bonds between the carbon atoms (unsaturated). In one embodiment, the fatty acid is a saturated fatty acid. In one embodiment, the saturated fatty acid is a long-chain saturated fatty acid having 12 to 24 carbon atoms (C 12 -C 24 ), or 20 to 22 carbon atoms (C 20 -C 22 ). In one embodiment, the polyunsaturated fatty acid is a medium-chain polyunsaturated fatty acid having 5 to 11 carbon atoms (C 5 -Cn), or 8 to 11 carbon atoms (C 8 -Cn).

[51] Exemplary fatty acids for use as the fatty acid component of the compounds described here include valproic acid, caproic acid (hexanoic acid), caprylic acid (octanoic acid), or capric acid (decanoic acid), lauric acid (dodecanoic acid), or a derivative thereof, including e.g., synthetic odd-chain fatty acids such as triheptanoin. In embodiments, the fatty acid component comprises or consists of palmitic acid or stearic acid.

Physical Properties

[52] Generally, the compounds of the disclosure are physically and chemically stable solid materials, e.g., powders. In embodiments, the compounds are solid, free flowing powders suitable for formulation into solid dosage forms such as powders, tablets, capsules or caplets. The solid, free-flowing character of the compounds also provides for ease of their formulation in physical admixture with each other and with other active agents in the same solid dosage form. In embodiments, the compounds are non-hygroscopic or have low hygroscopicity. In embodiments, the compounds are highly water soluble.

[53] The compounds of the disclosure generally possess superior chemical and physical stability of the fatty acid component, for example as compared to fatty acid compounds based upon the oil form of the fatty acid, e.g., the free fatty acids or the ethyl ester or glyceryl ester forms of the fatty acids. Since the compounds described here are solids (not oils) they are very stable against oxidative degradation of the fatty acid component, particularly when compared to the free fatty acid or fatty acid ester forms of the fatty acids, which are highly susceptible to oxidative degradation in their liquid forms and consequently tend to degrade when exposed to air or humidity. In contrast, the compounds described here, are, for example, relatively more stable to air, oxygen, and humidity compared to compounds and compositions comprising the free fatty acids or the ethyl ester or glyceryl ester forms of the fatty acids.

[54] The compounds described here are also expected to provide improved bioavailability of the fatty acid component, due to improved pharmacokinetics, compared to other dosage forms comprising, for example, the oily free fatty acids and esters of the fatty acids.

Compositions

[55] The present disclosure provides compositions comprising one or more of the compounds described herein, including compositions comprising mixtures of two or more different compounds described herein. Also provided are compositions comprising one or more of the compounds described herein, or mixtures of same, along with a second active agent. In embodiments the second active agent is a biologically active agent or an active pharmaceutical ingredient (API).

[56] The compositions comprising one or more compounds of the invention may be formulated as a solid dosage form selected from a powder, tablet, capsule, or caplet. In one embodiment, the solid dosage form is adapted for oral delivery. In one embodiment, the solid dosage form is adapted for once a day delivery. In another embodiment, the solid dosage form is adapted for delivery twice a day. In one embodiment, the dosage form is an oral dosage form. The oral dosage form may be in the form of a solid, such as a tablet, a capsule containing particulates, liquids, or powders, a lozenge (including liquid-filled), a gum, or a gel. In one embodiment, the dosage form is a solid oral dosage form. [57] In one embodiment, the composition is a powder suitable for reconstitution in an aqueous liquid. Such powders may be used, for example, to prepare a liquid suitable for intraperitoneal delivery of the compounds described here.

[58] In one embodiment, the composition is a pharmaceutical composition and the carrier is acceptable for administration to humans or non-human animals, as described in more detail infra.

[59] In one embodiment, the composition is a dietary supplement or additive and the carrier is acceptable for administration to humans or non-human animals, as described in more detail infra.

[60] A composition may be in the form of a unit dose. The unit dose may be, for example, in the form of a tablet or capsule. In one embodiment, the composition comprises a compound of Formula I or II. In one embodiment, a unit dose of the composition contains from about 0.05 g to 12 g of total fatty acids. In one embodiment, the unit dose contains from about 0.05 g, 1 g, 2 g, 3 g, 4 g, 5 g, or 6 g of total fatty acids.

[61] In embodiments, a composition of the invention contains from about 50 mg to 6 g of fatty acids in the fatty acid counter ion component. In one embodiment, a unit dose of such a composition comprises from about 50 mg to 6 g, or from about 500 mg to 6 g, or at least 200 mg, at least 300 mg, at least 400 mg, at least 500 mg, or at least 1 g of fatty acids.

In one embodiment, the fatty acid component of the composition consists of at least 70%, at least 80%), or at least 90% by weight of one or more saturated fatty acids, or from about 20% to 90%, from 30% to 90%, from 40% to 90%, from 50% to 90%, from 60% to 90%, or from 70%) to 90%) by weight of one or more saturated fatty acids.

[62] The compounds described here may be formulated alone or in combination with one or more additional active pharmaceutical ingredients (API) or biologically active agents. In one embodiment, a compound described here is formulated with one or more additional APIs or biologically active agents in a single dosage form. In one embodiment, the dosage form is a solid dosage form. In one embodiment, the solid dosage form is a powder suitable for reconstitution in aqueous media.

[63] Depending on the nature of the compounds and excipients making up the

composition, the composition may be suitable for pharmaceutical or veterinary use, or for use a dietary additive or supplement, or any combination of these uses. The various compositions are discussed in the following sections as "pharmaceutical compositions" and "additives and supplements" but these terms are not meant to be limiting, only descriptive. [64] The compositions of the invention may be formulated using one or more suitable excipients or carriers. A suitable excipient or carrier is one suitable for human or animal use. The term "excipient" refers to an additive that serves some purpose in the composition other than a carrier, for example as a stabilizer, taste masking agent (e.g., a sweetener), solubilizing agent, or suspending agent. Often, a carrier will serve a dual purpose as a simple carrier or diluent and an excipient. Examples of pharmaceutically acceptable excipients may thus include carriers. Non-limiting examples of excipients for use in the compositions of the invention include sterile liquids, water, buffered saline, ethanol, polyols (for example, glycerol, propylene glycol, liquid polyethylene glycol and the like), oils, detergents, suspending agents, carbohydrates (e.g., glucose, lactose, sucrose or dextran), antioxidants (e.g., ascorbic acid or glutathione), chelating agents, low molecular weight proteins, and suitable mixtures thereof.

[65] A suitable excipient or carrier is typically a pharmaceutically acceptable carrier or excipient for use in animals or humans (or both). The term "pharmaceutically acceptable" indicates approval by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia such as the European Pharmacopeia, for use in animals, and more particularly in humans. In the context of the pharmaceutical compositions of the invention, a "carrier" refers to, for example, a solvent, a diluent, or vehicle with which the ionic salt of the invention is formulated for delivery.

Examples of pharmaceutically acceptable carriers for use in the compositions of the invention include, without limitation, sterile aqueous and non-aqueous liquids, water, buffered saline, ethanol, polyols (for example, glycerol, propylene glycol, liquid polyethylene glycol and the like), and oils, for liquid dosage forms; or carbohydrates (e.g., glucose, lactose, sucrose or dextran) for solid dosage forms.

[66] The compounds of the invention may be formulated in any suitable form and for any suitable intended route of administration. Typically, the dosage form is at least in part determined by the intended route of administration.

[67] In one embodiment, the dosage form is a liquid suitable for administration to the eye. The formulation may be a solution, suspension, or gel suitable for ocular administration, e.g., suitable for topical administration to the eye, also referred to as an ophthalmic formulation.

[68] In one embodiment, the ophthalmic formulation is an aqueous formulation. In one embodiment, the ophthalmic formulation comprises one or more of glycerin, hypromellose, propylene glycol or polyethylene glycol. In one embodiment, the ophthalmic formulation further comprises one or more of polysorbate 80, carbomer copolymer type A, purified water, sodium hydroxide, ascorbic acid, benzalkonium chloride, boric acid, dextrose, disodium phosphate, glycine, magnesium chloride, potassium chloride, sodium borate, sodium chloride, sodium citrate, sodium lactate, edetate disodium, hydrochloric acid, sodium hydroxide, aminornethylpropanol, hydroxypropyl guar, polyquaternium-I, or sorbitol.

[69] In one embodiment, the ophthalmic formulation comprises one or more of surfactants, tonicity agents, buffers, preservatives, co-solvents and viscosity building agents. Various tonicity agents may be employed to adjust the tonicity of the composition, preferably to that of natural tears for ophthalmic compositions. For example, sodium chloride, potassium chloride, magnesium chloride, calcium chloride, dextrose and/or mannitol may be added to the composition to approximate physiological tonicity. Preferably, the tonicity agent is present in an amount sufficient to cause the final composition to have an ophthalmically acceptable osmolality (generally about 150-450 mOsm, preferably 250-350 mOsm). An appropriate buffer system (e.g., sodium phosphate, sodium acetate, sodium citrate, sodium borate or boric acid) may be added to the compositions to prevent pH drift under storage conditions. The particular concentration will vary, depending on the agent employed.

Preferably, however, the buffer will be chosen to maintain a target pH within the range of pH 6-7.5.

[70] Methods of preparing these formulations or compositions include the step of bringing into association a compound of the present invention with the carrier and, optionally, one or more accessory ingredients. In general, the formulations are prepared by uniformly and intimately bringing into association a compound of the present invention with liquid carriers.

Pharmaceutical Compositions

[71] In embodiments, the composition is a pharmaceutical composition comprising a compound of Formula I or Formula II, and optionally a pharmaceutically acceptable carrier and/or excipient. In embodiments, the composition further comprises an additional active agent, such as an API, as described below.

[72] In one embodiment is provided a solid dosage form comprising a compound of the invention in physical admixture with one or more additional active pharmaceutical ingredients (APIs). In embodiments, the one or more additional APIs is an antihyperlipidemic agent, an anti-diabetic agent, an anti-epileptic agent, or an anti-inflammatory agent. In one embodiment the API is an antihyperlipidemic agent or an anti-diabetic agent. In one embodiment, the antihyperlipidemic agent is selected from the group consisting of an HMG CoA enzyme inhibitor (e.g., a statin), a cholesterol absorption inhibitor, and a cholesterol esterase transfer protein (CETP) inhibitor. In one embodiment, the antihyperlipidemic agent is a statin. In one embodiment, the statin is selected from the group consisting of

atorvastatin, risuvostatin, simvastatin, pravastatin, and pharmaceutically acceptable salts or prodrugs thereof. In one embodiment, the statin is present in an amount ranging from 5 mg to 100 mg. In one embodiment, the statin is pravastatin. In one embodiment, the

antihyperlipidemic agent is a cholesterol absorption inhibitor. In one embodiment, the cholesterol absorption inhibitor is ezetimibe, also known as Zetia. In one embodiment, the antihyperlipidemic agent is a CETP inhibitor. In one embodiment, the CETP inhibitor is anacetrapib, or a hydrate, or solvate thereof.

[73] In embodiments, the pharmaceutical composition comprises an amount of one or more of the compounds described here effective to treat epilepsy, pediatric epilepsy, and epilepsy or pediatric epilepsy that is resistant to standard treatment, such as treatment with valproic acid.

[74] In embodiments, the pharmaceutical composition comprises an amount of one or more of the compounds described here effective to treat a fatty acid metabolism disorder, a long-chain fatty acid oxidation disorder, or a lipid metabolism disorder.

[75] In embodiments, the pharmaceutical composition comprises an amount of one or more of the compounds described here effective to treat a metabolic disorder selected from the group consisting of abnormal glucose metabolism manifesting in diabetes or pre-diabetes, abnormal lipid metabolism manifesting as hypertriglyceridemia, i.e., elevated triglycerides, mixed dyslipidemia, fatty liver, and combined abnormal glucose and lipid metabolism manifesting in obesity. In one embodiment, a composition of the invention is used in a method for treating a disease or disorder selected from diabetes, pre-diabetes,

hypertriglyceridemia, dyslipidemia, fatty liver, and obesity.

[76] Particular compounds for use in treating the various diseases and disorders referred to here are described in more detail infra.

Non-Pharmaceutical Compositions

[77] The compounds of the invention may also be formulated with one or more additional non-pharmaceutical agents, for example beneficial biologically active agents, such as a nutrient or nutraceutical compounds, including e.g., vitamins, minerals, botanical extracts, etc., in the same dosage form, along with any suitable excipients or carriers. In one embodiment, the one or more additional biologically active agents is selected from the group consisting of a vitamin, a mineral, an amino acid, a carbohydrate, an antioxidant, a flavonoid, a carotenoid, a phytoseterol, an herb, an enzyme, a botanical extract or concentrate, and a botanical compound. In one embodiment, the one or more additional biologically active agents is selected from the group consisting of vitamin A, vitamin Bl, vitamin B12, vitamin B6, vitamin C, vitamin D, vitamin E, vitamin K, calcium, carnitine, chromium, chondroitin, coenzyme Q10 (ubiquinone), folate, glucosamine, metafolin, riboflavin, biotin, iodine, iron, magnesium, selenium, thiamin, and zinc. In one embodiment, the one or more additional biologically active agents is selected from the group consisting of coenzyme Q10, L- carnitine, an antioxidant, a phytoseterol, and a flavonoid. In one embodiment, the antioxidant is a polyphenol. In one embodiment, the polyphenol is selected from lycopene, resveratrol, and epigallocatechingallate.

[78] In one embodiment, a compound described here is useful as a dietary supplement or nutraceutical additive. In an embodiment, a compound of Formula I or Formula II is formulated as a nutraceutical additive or supplement, either alone or in combination with one or more additives or supplements and any suitable excipients. In one embodiment, the nutraceutical additive or supplement is in the form of a powder. In one embodiment, the nutraceutical additive or supplement is in the form of a liquid. In one embodiment, the nutraceutical additive or supplement is in the form of a mouth wash, a dentifrice, chewing gum, a candy, a tablet, a capsule, a mouth spray, or a film.

[79] In one embodiment, the nutraceutical additive forms part of a food or drink product suitable for human consumption. There is no specific limitation on the foods/drinks to which a nutraceutical additive of the invention can be incorporated. Examples of such foods/drinks include processed foods based on meat, poultry meat, fish/shellfish and the like; soup;

seasonings including sweetener and the like; rice seasonings; instant foods; frozen foods; snacks; various types of functional foods such as supplements, nutritional drinks and the like; canned foods; dairy products; confectionery such as chewing gum, candy, gummy candy, chocolate, baked sweets and the like; ice cream; soft drinks such as tea, coffee, cocoa, fruit juice, sports drink, carbonated drink, vegetable drink and the like; liquors; soya milk; lactic acid bacteria beverages; and chlorophyll juice.

[80] The amount of the nutraceutical additive of the invention incorporated into the food or drink varies in accordance with the type of food or drink and the amount that one wishes to supplement a diet with one or more omega-3 fatty acids. In one embodiment, the

nutraceutical additive is incorporated into the food or drink so as to provide an amount of the omega-3 fatty acid that is about 0.000001 to 20% by weight, based on total weight of the food or drink product, and more preferably in an amount of about 0.00001 to 10% by weight.

Methods of Use

[81] The compositions of the invention are useful in methods of treating various diseases and disorders that are responsive to treatment with fatty acids. In addition, the compositions of the invention may have non-pharmaceutical uses, for example as dietary supplements or additives. These uses are described in more detail infra.

[82] In one embodiment, the methods relate to diseases and disorders that are responsive to treatment with fatty acids, especially saturated fatty acids. In embodiments, the disease or disorder is selected from epilepsy, pediatric epilepsy, and epilepsy or pediatric epilepsy that is resistant to standard treatment, such as treatment with valproic acid; a fatty acid

metabolism disorder, a long-chain fatty acid oxidation disorder, or a lipid metabolism disorder; or a metabolic disorder selected from the group consisting of abnormal glucose metabolism manifesting in diabetes or pre-diabetes, abnormal lipid metabolism manifesting as hypertriglyceridemia, i.e., elevated triglycerides, mixed dyslipidemia, fatty liver, and combined abnormal glucose and lipid metabolism manifesting in obesity; diabetes, prediabetes, hypertriglyceridemia, dyslipidemia, fatty liver, and obesity.

[83] In accordance with any of the following embodiments the fatty acid component of a compound described herein may be selected from valproic acid, caproic acid (hexanoic acid), caprylic acid (octanoic acid), or capric acid (decanoic acid), lauric acid (dodecanoic acid), or a derivative thereof, including e.g., synthetic odd-chain fatty acids such as triheptanoin.

[84] In embodiments, the fatty acid component of the compounds administered according to the methods described herein comprises a long-chain saturated fatty acid, a medium chain saturated fatty acid, or a mixture of medium and long chain saturated fatty acids.

[85] In the context of any of the methods described here, a compound described herein may be formulated as a pharmaceutical composition, or as a food additive or supplement, meaning that the compound itself and any additives or excipients in the formulation are suitable for administration to humans or animals.

[86] In the context of the methods described here, the term "treating" may refer to the amelioration or stabilization of one or more symptoms associated with the disease or disorder being treated. The term "treating" may also encompass the management of a disease or disorder, referring to the beneficial effects that a subject derives from a therapy which does not result in a cure of the underlying disease or disorder. The compositions of the invention can also be used in the prevention of certain diseases, disorders, and conditions. In this context, the term "prevention" refers to preventing the recurrence, development, progression or onset of one or more symptoms of the disease, disorder, or condition.

[87] In accordance with the methods described here, a therapeutically effective amount of a compound described herein is administered to a subject, the therapeutically effective amount being an amount of the compound (or mixture of two or more compounds) sufficient to achieve a desired therapeutic outcome, for example the amelioration or stabilization of one or more symptoms of the disease or disorder being treated, or in the context of prevention, the amount sufficient to achieve prevention of the recurrence, development, progression or onset of one or more symptoms of the disease, disorder, or condition.

[88] For administration to human patients, the total daily dose of a compound of the invention is typically in the range 50 mg to 12 g depending, of course, on the route of administration. In one embodiment the total daily dose is in the range of from about 100 mg to 500 mg, about 500 mg to 1 g, about 1 g to 2 g, about 2 g to 5 g, or about 5 g to 10 g. In another embodiment the total daily dose is in the range 4 g to 8 g and in yet another embodiment the total daily dose is in the range 1 g to 2 g. The total daily dose may be administered in single or divided doses.

[89] These dosages are based on an average human subject having a weight of about 65 kg to 70 kg. The physician will readily be able to determine doses for subjects whose weight falls outside this range, such as infants and the elderly.

[90] In one embodiment, a therapeutically effective amount is the amount required to achieve at least an equivalent therapeutic effect compared to a standard therapy.

[91] In the context of any of the methods of the present invention, the subject may be a human or a non-human mammal. The non-human mammal may be, for example, a non- human primate, a dog, cat, a rodent (e.g., a mouse, a rat, a rabbit), a horse, a cow, a sheep, a goat, a bird, a chicken, or any other non-human mammal. Preferably, the subject is a human.

[92] In one embodiment, the subject is a human subject. In one embodiment, the human is an adult human, a pediatric human, or a geriatric human, as those terms are understood by the medical practitioner, for example as defined by the U.S. Food and Drug Administration.

[93] The compounds or compositions described here can be used as monotherapy or adjunctive therapy. The compositions of the invention can be administered alone or in combination with one or more additional therapeutic agents (i.e., additional APIs) or therapies, for example as part of a therapeutic regimen that includes, e.g., aspects of diet and exercise. In certain embodiments, the methods of the invention include administration of a composition of the invention as the primary therapy. In other embodiments, the administration of a composition of the invention is an adjuvant therapy. In either case, the methods of the invention contemplate the administration of a composition of the invention in combination with one or more additional therapeutic agents and/or therapies for the treatment or prevention of a disease or disorder. The terms "therapy" and "therapies" refer to any method, protocol and/or agent that can be used in the prevention, treatment, management or amelioration of a disease or disorder, or one or more symptoms thereof.

[94] The compounds or compositions described here can also be used in combination therapy. As used herein, "combination therapy" or "co-therapy" includes the administration of a therapeutically effective amount of one or more of the compounds described here as part of a specific treatment regimen intended to provide the beneficial effect from the co-action of the one or more compounds and an additional active agent, for example an additional API or active biological agent as described above. The beneficial effect of the combination includes, but is not limited to, pharmacokinetic or pharmacodynamic co-action resulting from the combination. The beneficial effect of the combination may also relate to the mitigation of a toxicity, side effect, or adverse event associated with another agent in the combination.

"Combination therapy" is not intended to encompass the administration of two or more compounds as part of separate monotherapy regimens that incidentally and arbitrarily result in a beneficial effect that was not intended or predicted.

Epilepsy

[95] In one embodiment, the invention provides methods of treating a epilepsy, pediatric epilepsy, and epilepsy or pediatric epilepsy that is resistant to standard treatment, such as treatment with valproic acid, in a subject in need thereof, the method comprising

administering to the subject, preferably a human subject, an effective amount of a

composition comprising a compound described herein, or mixtures thereof.

Metabolic Disorders

[96] In one embodiment, the invention provides methods of treating a metabolic disorder in a subject in need thereof, the method comprising administering to the subject, preferably a human subject, an effective amount of a composition comprising a compound described herein, or mixtures thereof.

[97] In embodiments the metabolic disorder is selected from a fatty acid metabolism disorder, a long-chain fatty acid oxidation disorder, or a lipid metabolism disorder; a metabolic disorder selected from the group consisting of abnormal glucose metabolism manifesting in diabetes or pre-diabetes, abnormal lipid metabolism manifesting as hypertriglyceridemia, i.e., elevated triglycerides, mixed dyslipidemia, fatty liver, and combined abnormal glucose and lipid metabolism manifesting in obesity; or diabetes, prediabetes, hypertriglyceridemia, dyslipidemia, fatty liver, and obesity.

[98] In embodiments the metabolic disorder is selected from the group consisting of abnormal glucose metabolism manifesting in diabetes or pre-diabetes, abnormal lipid metabolism manifesting as hypertriglyceridemia, i.e., elevated triglycerides, mixed dyslipidemia, hypercholesterolemia, fatty liver, and combined abnormal glucose and lipid metabolism manifesting in obesity. In one embodiment the metabolic disorder is a dyslipidemic disorder selected from hypertriglyceridemia, hypercholesterolemia and mixed dyslipidemias. In one embodiment, the metabolic disorder is selected from the group consisting of pre-diabetes, type 2 diabetes, obesity, fatty liver disease, and insulin resistance.

[99] In one embodiment, the methods comprise administering a therapeutically effective amount, which amount is effective to reduce plasma triglycerides in an adult human subject by at least about 0.5 mmol/L, about 1 mmol/L, or about 2 mmol/L.

[100] In one embodiment, the subject is a human subject having severe

hypertriglyceridemia characterized by serum triglyceride levels of from 500 to 2,000 mg/dl.

Combination Therapies

[101] In the context of combination therapies, a composition of the invention may be administered together with at least one additional API or separately from the additional API. Where delivery is together, a composition of the invention may be delivered in the same dosage form as the additional API, or in a different dosage form. One of the advantages of the present invention, as discussed above, is the ease of formulating the compositions described herein with additional APIs and excipients in a single solid dosage form due to their form as a free flowing powder that is chemically and physically stable (as opposed to the relatively unstable oily liquid form of free fatty acids and their esters).

[102] In one embodiment, a composition of the invention is formulated in a single solid dosage form with an antihyperlipidemic agent or an anti-diabetic agent. Antihyperlipidemic agents that may be used include HMG CoA enzyme inhibitors (e.g., statins), cholesterol absorption inhibitors, and cholesterol esterase transfer protein (CETP) inhibitors. In one embodiment, the antihyperlipidemic agent is selected from a statin, a cholesterol absorption inhibitor, a CETP inhibitor, and pharmaceutically-acceptable salts and prodrugs of any of the foregoing. The pharmaceutically acceptable salt may be selected from the group consisting of a propionate, decanoate, caprylate, acrylate, formate, isobutyrate, caprate, heptanoate, propiolate, oxalate, malonate, succinate, suberate, sebacate, fumarate, maleate, butyne-1,4- dioate, hexyne-l,6-dioate, benzoate, chlorobenzoate, methylbenzoate, dinitrobenzoate, hydroxybenzoate, methoxybenzoate, phthalate, terephathal ate, sulfonate, xylenesulfonate, phenyl acetate, phenylpropionate, phenylbutyrate, citrate, lactate, p- hydroxybutyrate, glycolate, tartrate, methanesulfonate, propanesulfonates, naphthalene- 1 -sulfonate, naphthalene-2-sulfonate, mandelate, hippurate, gluconate, and lactobionate salt.

[103] In one embodiment, the antihyperlipidemic agent is a statin. In one embodiment, the statin is selected from the group consisting of atorvastatin, risuvostatin, simvastatin, pravastatin, and pharmaceutically acceptable salts and prodrugs of any of the foregoing. In one embodiment, the statin is present in an amount ranging from 5 mg to 100 mg. In one embodiment, the statin is pravastatin.

[104] In one embodiment, the antihyperlipidemic agent is a cholesterol absorption inhibitor.

In one embodiment, the cholesterol absorption inhibitor is ezetimibe, also known as Zetia.

[105] In one embodiment, the antihyperlipidemic agent is a CETP inhibitor. In one embodiment, the CETP inhibitor is anacetrapib, or a hydrate, or solvate thereof.

[106] In one embodiment, a composition of the invention is formulated in a single solid dosage form with an anti-epileptic agent or an inhibitor of neuropathic pain such as gabapentin, or a pharmaceutically acceptable salt and prodrug thereof.

[107] The invention is further described in the following examples, which do not limit the scope of the invention described in the claims.

Non-Pharmaceutical Uses

[108] In one embodiment, the invention provides compositions, particularly compositions of Formula I or Formula II, and mixtures of same, for a non-pharmaceutical use, e.g., for use as a dietary supplement or additive. In accordance with any of the embodiments described herein, the method may comprise administering to the subject an effective amount of a composition comprising a compound described herein. In embodiments, the effective amount is effective to maintain, promote, or improve the general health of the subject.

[109] In one embodiment, the composition may be used in a method to counter a dietary deficiency or nutritional disorder in a subject. In one embodiment, the composition may be used in a method for maintaining, promoting, or improving the general health of a subject. [110] The invention is further described and exemplified by the following non-limiting examples.

EXAMPLES

Example 1 : Preparation of L-lysyl-L-lysine bis stearic acid

Step 1: Benzyl N6-((benzyloxy)carbonyl)-N2-(N2,N6-bis((benzyloxy)carbonyl)- L-lysyl)- L-lysinate

CbzHN

HCbz

[111] A solution/suspension of H-Lys(Z)-OBzl hydrochloride (16.3g, 40mmol) and Z- Lys(Z)-OH (16.6g, 40mmol) in anhydrous dichloromethane (50mL) under nitrogen was cooled on ice and treated with HOBT hydrate (9.2g, 60mmol) and triethylamine (16.8mL, 120mmol), and the clear solution was stirred for 15min. EDC hydrochloride (9.6g, 50mmol) was added, and the stirred mixture allowed to warm to room temperature and stirred 20h. The product mixture was diluted to 500mL total volume with dichloromethane, then washed successively with 5% citric acid, water, saturated aqueous sodium bicarbonate, and brine (200mL each), and dried (Na 2 S0 4 ). The solution was added directly to a silica gel column (~500cc) and eluted with dichloromethane, then 2: 1 dichloromethane/ethyl acetate to afford 29.71g (97%) of subject material as a white solid. MP 145-6°C. NMR (CDC1 3 ): δ 7.15-7.30 (m, 20H), 6.50-6.65 (m, 1H), 5.40-5.55 (m, 1H), 5.10 (d, 1H, J=l 1.5Hz), 4.90-5.10 (m, 8H), 4.45-4.55 (m, 1H), 4.05-4.15 (m, 1H), 2.90-3.15 (m, 4H), 1.70-1.80 (m, 2H), 1.55-1.65 (m, 2H), 1.10-1.45 (m, 8H). 13 C NMR (151 MHz, cdcl 3 ) δ 171.94, 156.68, 156.59, 156.33, 136.59, 136.50, 136.15, 135.23, 128.60, 128.59, 128.50, 128.48, 128.47, 128.37, 128.34, 128.16, 128.09, 128.04, 76.82, 67.21, 67.05, 66.69, 66.60, 54.50, 52.14, 40.22, 40.17, 32.06, 31.25, 29.17, 22.14, 22.04.

Step 2: L-lysyl-L-lysine

[112] A stirred solution/suspension of (L,L)-Z3-lysyllisine, benzyl ester (5.37g, 7.0mmol) in methanol (120mL) under nitrogen was treated with 10% Pd/C (0.50g), then evacuated and purged several times with hydrogen via balloon. The mixture was stirred for 42h under hydrogen, then the flask evacuated with nitrogen and carefully filtered through Celite with water rinse. The filtrate was concentrated in vacuo to afford a white foam. The foam was transferred to a 50mL pear shaped flask by dissolving in hot methanol, then concentrated in vacuo and the residual foam stirred with acetonitrile for 30min, filtered, collected, and dried in vacuo to afford 1.92g (100%) as a white powder. MP 88-90°C. NMR (D 2 0): δ 4.08 (dd, 1H, J=5Hz,8Hz), 3.31 (t, 1H, J=7Hz), 2.75-2.95 (m, 4H), 1.45-1.80 (m, 8H), 1.20-1.40 (m, 4H). 13 C MR (151 MHz, d 2 o) δ 178.75, 176.84, 54.77, 54.26, 40.95, 39.40, 33.72, 31.12, 27.66, 27.11, 22.14, 21.80.

Step 3: L-lysyl-L-lysine bis stearic acid

[113] While under nitrogen, a solution of (L,L)-Lysyllisine (0.29 g, 1.08 mmol) in methanol (8 mL) was warmed to 50 °C and treated with a combined solution of stearic acid (0.71 g, 2.48 mmol) and alpha-D-tocopherol (15 mg pre-dissolved in 0.3 mL of ethyl acetate and added to the stearic acid solution ) in methanol (7 mL). After stirring for 20 min, the mixture is cooled to room temperature and concentrated. The resulting foam is suspended in acetonitrile, stirred for 2 hours, collected by filtration, washed with acetonitrile and dried overnight in the vacuum oven to afford 0.89 g (98%) of (L,L)-Lysyllisine bis stearic acid as a white solid. 1H MR (400 MHz, d4-MeOH): δ 4.23 (dd, 1H, J=5Hz,8Hz) 3.63 (t, 1H, J=6.5Hz) 2.92-2.89 (m, 4H) 2.21 (t, 4H, J=7.5Hz) 1.87-1.44 (m, 16H) 1.37-1.22 (m, 56H) 0.89 (t, 6H, J=7.5Hz)

Example 2: Preparation of L-lysyl-L-lysine bis palmitic acid

[114] While under nitrogen, a solution of (L,L)-Lysyllisine (0.27 g, 0.97 mmol) in methanol (8 mL) was warmed to 50 °C and treated with a combined solution of palmitic acid (0.57 g, 2.23 mmol) and alpha-D-tocopherol (15 mg pre-dissolved in 0.3 mL of ethyl acetate and added to the palmitic acid solution ) in methanol (7 mL). After stirring for 20 min, the mixture is cooled to room temperature and concentrated. The resulting foam is suspended in acetonitrile, stirred for 2 hours, collected by filtration, washed with acetonitrile and dried overnight in the vacuum oven to afford 0.69 g (90%) of (L,L)-Lysyllisine bis palmitic acid as a white solid. 1H MR (400 MHz, d4-MeOH): δ 4.23 (dd, 1H, J=5Hz,8Hz) 3.60 (t, 1H, J=7 Hz) 2.93-2.89 (m, 4H) 2.21 (t, 4H, J=7.5Hz) 1.89-1.42 (m, 16H) 1.37-1.22 (m, 48H) 0.89 (t, 6H, J=7.5Hz)

Example 3 : Preparation of L-lysyl-L-lysine bis lauric acid

[115] While under nitrogen, a solution/suspension of L-lysyl-L-lysine (0.30 g, 1.09 mmol) in ethanol (10 mL) was warmed to 60 °C and treated with a combined solution of lauric acid (0.50 g, 2.51 mmol) and alpha-D-tocopherol (15 mg pre-dissolved in 0.3 mL of ethyl acetate and added to the lauric acid solution) in ethanol (5 mL). After stirring for 20 min, the mixture is cooled to room temperature and concentrated. The resulting foam is suspended in acetonitrile, stirred for 2 hours, collected by filtration, washed with acetonitrile and dried overnight in the vacuum oven to afford 0.68 g (92%) of L-lysyl-L-lysine bis lauric acid as a white solid. 1H MR (400 MHz, d4-MeOH): δ 4.26 (dd, 1H, J=5Hz,8Hz) 3.62 (t, 1H, J=6.5Hz) 2.95-2.90 (m, 4H) 2.21 (t, 4H, J=7.5Hz) 1.91-1.83 (m, 1H) 1.80-1.56 (m, 11H) 1.52-1.45 (m, 4H) 1.35-1.22 (m, 32H) 0.90 (t, 6H, J=7Hz).

Example 4: Preparation of L-lysyl-L-lysine mono lauric acid

[116] While under nitrogen, a solution/suspension of L-lysyl-L-lysine (0.53 g, 1.93 mmol) in ethanol (10 mL) was warmed to 60 °C and treated with a combined solution of lauric acid (0.40 g, 2.02 mmol) and alpha-D-tocopherol (14 mg pre-dissolved in 0.3 mL of ethyl acetate and added to the lauric acid solution) in ethanol (5 mL). After stirring for 20 min, the mixture is cooled to room temperature and concentrated. The resulting foam is suspended in acetonitrile, stirred for 2 hours, collected by filtration, washed with acetonitrile and dried overnight in the vacuum oven to afford 0.82 g (89%) of L-lysyl-L-lysine mono lauric acid as a white solid. 1H NMR (400 MHz, d4-MeOH): δ 4.26 (dd, 1H, J=5Hz,8Hz) 3.42 (t, 1H, J=6.5Hz) 2.91 (t, 4H, J=7Hz) 2.17 (t, 2H, J=7.5Hz) 1.90-1.82 (m, 1H) 1.73-1.56 (m, 9H) 1.48-1.41 (m, 4H) 1.34-1.24 (m, 16H) 0.90 (t, 3H, J=7Hz).

Example 5: Preparation of L-lysyl-L-lysine bis caprylic acid

[117] While under nitrogen, a solution/suspension of L-lysyl-L-lysine (0.33 g, 1.20 mmol) in ethanol (10 mL) was warmed to 60 °C and treated with a combined solution of caprylic acid (0.40 g, 2.76 mmol) and alpha-D-tocopherol (14 mg pre-dissolved in 0.3 mL of ethyl acetate and added to the caprylic acid solution) in ethanol (5 mL). After stirring for 20 min, the mixture is cooled to room temperature and concentrated. The resulting foam is suspended in acetonitrile, stirred for 2 hours, collected by filtration, washed with acetonitrile and dried overnight in the vacuum oven to afford 0.63 g (93%) of L-lysyl-L-lysine bis caprylic acid as a white solid. 1H NMR (400 MHz, d4-MeOH): δ 4.26 (dd, 1H, J=5Hz,8Hz) 3.58 (t, 1H, J=6.5Hz) 2.94-2.90 (m, 4H) 2.20 (t, 4H, J=7.5Hz) 1.89-1.83 (m, 1H) 1.77-1.55 (m, 11H) 1.50-1.42 (m, 4H) 1.35-1.22 (m, 16H) 0.90 (t, 6H, J=7Hz).

Example 6: Preparation of L-lysyl-L-lysine mono caprylic acid

[118] While under nitrogen, a solution/suspension of L-lysyl-L-lysine (0.51 g, 1.86 mmol) in ethanol (10 mL) was warmed to 60 °C and treated with a combined solution of caprylic acid (0.28 g, 1.95 mmol) and alpha-D-tocopherol (15 mg pre-dissolved in 0.3 mL of ethyl acetate and added to the caprylic acid solution) in ethanol (5 mL). After stirring for 20 min, the mixture is cooled to room temperature and concentrated. The resulting foam is suspended in acetonitrile, stirred for 2 hours, collected by filtration, washed with acetonitrile and dried overnight in the vacuum oven to afford 0.68 g (87%) of L-lysyl-L-lysine mono caprylic acid aass aa wwhhiittee ssoolliidd.. 1H1H NNMMRR ((440000 MMHHzz,, dd44--MMeeOOHH)):: δδ 44..2266 ((dddd,, 11HH,, JJ==55HHzz,,88HHzz)) 33..4422 ((tt,, 11HH,, JJ==66..55HHzz)) 22..9911 ((tt,, 44HH,, JJ==77HHzz)) 22..1166 ((tt,, 22HH,, JJ==77..55HHzz)) 11..8899--11..8833 ((mm,, 11HH)) 11..7744--11..5533 ((mm,, 99HH)) 11..5500--11..4422 ((mm,, 44HH)) 11..3355--11..2244 ((mm,, 88HH)) 00..9900 ((tt,, 33HH,, JJ==77HHzz))..

EExxaammppllee 77:: PPrreeppaarraattiioonn ooff LL--llyyssyyll--LL--llyyssiinnee bbiiss ccaappriricc aacciidd

OO ccoo 22 -- *

[119] While under nitrogen, a solution/suspension of L-lysyl-L-lysine (0.33 g, 1.20 mmol) in ethanol (10 mL) was warmed to 60 °C and treated with a combined solution of capric acid (0.48 g, 2.76 mmol) and alpha-D-tocopherol (15 mg pre-dissolved in 0.3 mL of ethyl acetate and added to the capric acid solution) in ethanol (5 mL). After stirring for 20 min, the mixture is cooled to room temperature and concentrated. The resulting foam is suspended in acetonitrile, stirred for 2 hours, collected by filtration, washed with acetonitrile and dried overnight in the vacuum oven to afford 0.65 g (87%) of L-lysyl-L-lysine bis capric acid as a white solid. 1H NMR (400 MHz, d4-MeOH): δ 4.26 (dd, 1H, J=5Hz,8Hz) 3.61 (t, 1H, J=6.5Hz) 2.94-2.90 (m, 4H) 2.20 (t, 4H, J=7.5Hz) 1.91-1.84 (m, 1H) 1.80-1.55 (m, 11H) 1.52-1.44 (m, 4H) 1.39-1.25 (m, 24H) 0.91 (t, 6H, J=7Hz).

Example 8: Preparation of L-lysyl-L-lysine mono capric acid

[120] While under nitrogen, a solution/suspension of L-lysyl-L-lysine (0.36 g, 1.30 mmol) in ethanol (10 mL) was warmed to 60 °C and treated with a combined solution of capric acid (0.24 g, 1.37 mmol) and alpha-D-tocopherol (12 mg pre-dissolved in 0.3 mL of ethyl acetate and added to the capric acid solution) in ethanol (5 mL). After stirring for 20 min, the mixture is cooled to room temperature and concentrated. The resulting foam is suspended in acetonitrile, stirred for 2 hours, collected by filtration, washed with acetonitrile and dried overnight in the vacuum oven to afford 0.54 g (92%) of L-lysyl-L-lysine mono capric acid as a white solid. 1H MR (400 MHz, d4-MeOH): δ 4.26 (dd, 1H, J=5Hz,8Hz) 3.43 (t, 1H, J=6.5Hz) 2.91 (t, 4H, J=7Hz) 2.17 (t, 2H, J=7.5Hz) 1.91-1.82 (m, 1H) 1.75-1.55 (m, 9H) 1.49-1.42 (m, 4H) 1.37-1.25 (m, 12H) 0.89 (t, 3H, J=7Hz).

Example 9: Preparation of L-lysyl-L-lysine bis caproic acid

[121] While under nitrogen, a solution/suspension of L-lysyl-L-lysine (0.28 g, 1.02 mmol) in ethanol (10 mL) was warmed to 60 °C and treated with a combined solution of caproic acid (0.27 g, 2.3 mmol) and alpha-D-tocopherol (12 mg pre-dissolved in 0.3 mL of ethyl acetate and added to the caproic acid solution) in ethanol (5 mL). After stirring for 20 min, the mixture is cooled to room temperature and concentrated. The resulting foam is suspended in acetonitrile, stirred for 2 hours, collected by filtration, washed with acetonitrile and dried overnight in the vacuum oven to afford 0.46 g (89%) of L-lysyl-L-lysine bis caproic acid as a white solid. 1H MR (400 MHz, d4-MeOH): δ 4.26 (dd, 1H, J=5Hz,8Hz) 3.59 (t, 1H, J=6.5Hz) 2.94-2.90 (m, 4H) 2.19 (t, 4H, J=7.5Hz) 1.92-1.83 (m, 1H) 1.80-1.57 (m, 11H) 1.54-1.43 (m, 4H) 1.36-1.26 (m, 8H) 0.91 (t, 6H, J=7Hz).

Example 10: Preparation of L-lysyl-L-lysine mono caproic acid

[122] While under nitrogen, a solution/suspension of L-lysyl-L-lysine (0.35 g, 1.28 mmol) in ethanol (10 mL) was warmed to 60 °C and treated with a combined solution of caproic acid (0.15 g, 1.34 mmol) and alpha-D-tocopherol (13 mg pre-dissolved in 0.3 mL of ethyl acetate and added to the caproic acid solution) in ethanol (5 mL). After stirring for 20 min, the mixture is cooled to room temperature and concentrated. The resulting foam is suspended in acetonitrile, stirred for 2 hours, collected by filtration, washed with acetonitrile and dried overnight in the vacuum oven to afford 0.42 g (84%) of L-lysyl-L-lysine mono caproic acid as a white solid. 1H MR (400 MHz, d4-MeOH): δ 4.26 (dd, 1H, J=5Hz,8Hz) 3.42 (t, 1H, J=6.5Hz) 2.91 (t, 4H, J=7Hz) 2.16 (t, 2H, J=7.5Hz) 1.90-1.83 (m, 1H) 1.77-1.56 (m, 9H) 1.54-1.41 (m, 4H) 1.37-1.26 (m, 4H) 0.91 (t, 3H, J=7Hz).

Equivalents

[123] Those skilled in the art will recognize or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following claims.

[124] All references cited herein are incorporated herein by reference in their entirety and for all purposes to the same extent as if each individual publication or patent or patent application was specifically and individually indicated to be incorporated by reference in its entirety for all purposes.

[125] The present invention is not to be limited in scope by the specific embodiments described herein. Indeed, various modifications of the invention in addition to those described herein will become apparent to those skilled in the art from the foregoing description and accompanying figures. Such modifications are intended to fall within the scope of the appended claims.