US20140107199A1 - Omega-3 pentaenoic acid compositions and methods of use - Google Patents

Omega-3 pentaenoic acid compositions and methods of use Download PDF

Info

Publication number
US20140107199A1
US20140107199A1 US14/109,254 US201314109254A US2014107199A1 US 20140107199 A1 US20140107199 A1 US 20140107199A1 US 201314109254 A US201314109254 A US 201314109254A US 2014107199 A1 US2014107199 A1 US 2014107199A1
Authority
US
United States
Prior art keywords
alternatively
dpa
epa
dha
fatty acids
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
US14/109,254
Inventor
Abdel Aziz Fawzy
George Bobotas
Roelof RONGEN
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.)
Matinas Biopharma Inc
Original Assignee
Matinas Biopharma Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Matinas Biopharma Inc filed Critical Matinas Biopharma Inc
Priority to US14/109,254 priority Critical patent/US20140107199A1/en
Assigned to MATINAS BIOPHARMA INC. reassignment MATINAS BIOPHARMA INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BOBOTAS, GEORGE, FAWZY, ABDEL AZIZ, RONGEN, ROELOF
Publication of US20140107199A1 publication Critical patent/US20140107199A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/20Carboxylic acids, e.g. valproic acid having a carboxyl group bound to a chain of seven or more carbon atoms, e.g. stearic, palmitic, arachidic acids
    • A61K31/201Carboxylic acids, e.g. valproic acid having a carboxyl group bound to a chain of seven or more carbon atoms, e.g. stearic, palmitic, arachidic acids having one or two double bonds, e.g. oleic, linoleic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/20Carboxylic acids, e.g. valproic acid having a carboxyl group bound to a chain of seven or more carbon atoms, e.g. stearic, palmitic, arachidic acids
    • A61K31/202Carboxylic acids, e.g. valproic acid having a carboxyl group bound to a chain of seven or more carbon atoms, e.g. stearic, palmitic, arachidic acids having three or more double bonds, e.g. linolenic
    • 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
    • A61K31/23Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acyclic acids, e.g. pravastatin of acids having a carboxyl group bound to a chain of seven or more carbon atoms
    • A61K31/232Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acyclic acids, e.g. pravastatin of acids having a carboxyl group bound to a chain of seven or more carbon atoms having three or more double bonds, e.g. etretinate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/4816Wall or shell material
    • A61K9/4825Proteins, e.g. gelatin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/4841Filling excipients; Inactive ingredients
    • A61K9/4858Organic compounds
    • 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
    • A61P9/00Drugs for disorders of the cardiovascular system

Definitions

  • the present invention relates to omega-3 fatty acid compositions, and methods of treating, preventing, reducing the occurrence of, and improving symptoms associated with coronary heart disease (CHD), vascular disease, atherosclerotic disease or related conditions.
  • CHD coronary heart disease
  • the present invention also provides methods for the treatment and/or prevention and/or reduction of cardiac events and/or cardiovascular events and/or vascular events and/or symptoms.
  • Marine oils also commonly referred to as fish oils, are a good source of the two main omega-3 fatty acids, eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), which have been found to regulate lipid metabolism.
  • Omega-3 fatty acids have been found to have beneficial effects on the risk factors for cardiovascular diseases, especially mild hypertension, hypertriglyceridemia and on the coagulation factor VII phospholipid complex activity.
  • Omega-3 fatty acids lower serum triglycerides (TG), increase serum HDL-cholesterol, lower systolic and diastolic blood pressure and the pulse rate, and lower the activity of the blood coagulation factor VII-phospholipid complex.
  • omega-3 fatty acids seem to be well tolerated, without giving rise to any severe side effects.
  • omega-3 fatty acids is a concentrate of omega-3, long chain, polyunsaturated fatty acids from fish oil containing DHA ethyl esters, EPA ethyl esters as well as ethyl esters of other omega-3 fatty acids (described in USP35 for LOVAZA®) and is sold under the trademarks OMACOR® and LOVAZA®.
  • omega-3 fatty acid comprises at least 90% omega-3 fatty acids of which at least 80% EPA+DHA (in a ratio of 1.2:1) and is described, for example, in U.S. Pat. Nos. 5,502,077, 5,656,667 and 5,698,594.
  • LOVAZA® (omega-3-acid ethyl esters) is indicated for the treatment of patients with hypertriglyceridemia with TG levels of 500 mg/dL or higher.
  • EPADEL® omega-3 fatty acid concentrate
  • This product is described as 98% EPA ethyl ester in Lancet (Vol. 369; Mar. 31, 2007; 1090-1098) reporting on a large outcome study with EPADEL®.
  • EPADEL® is known to contain less than 1% of any fatty acid other than EPA.
  • omega-3 fatty acid concentrate Similar to EPADEL®, another form of omega-3 fatty acid concentrate also consists almost entirely of EPA ethyl ester and is known under its developmental stage name AMR101 or its trade name VASCEPA®. This product is described in US patent application 2010/0278879 as comprising at least 95% EPA (typically referred to as 97% or at least 96% in company releases and references) and less than 1% of any other fatty acid.
  • AMR101 was previously under development for the treatment of Huntingdon's Disease but failed in phase Ill clinical development. Subsequently, AMR101 was entered in a development program for hypertriglyceridemia and mixed dyslipidemia.
  • omega-3, long chain, polyunsaturated fatty acids from fish oil containing approximately 75% DHA and EPA as free fatty acids is known under its developmental stage name EPANOVATM. This product is described as comprising approximately 55% EPA and 20% DHA. EPANOVATM was previously under development for the treatment of Crohn's Disease but failed in phase Ill clinical development. Subsequently, EPANOVATM was entered in a development program for hypertriglyceridemia and mixed dyslipidemia.
  • omega-3 fatty acid compositions are dose dependent, i.e., the higher the dose, the greater the therapeutic affect and bioavailability.
  • the effect of each specific omega-3 fatty acid composition may be different, and therefore the level of therapeutic effect of one composition at a given dose cannot necessarily be inferred from the level of therapeutic effects of other omega-3 fatty acid compositions at the same or similar dose.
  • Omega-3 fatty acids are known to be “essential fatty acids”.
  • the main EFAs in the diet are linoleic acid of the omega-6 series and alpha-linolenic acid of the omega-3 series. However, to fulfill most of their biological effects these “parent” EFAs must be metabolised to the other longer chain fatty acids. Each fatty acid probably has a specific role in the body.
  • n-6 series dihomo-gammalinolenic acid (DGLA, 20:3-n6) and arachidonic acid (ARA, 20:4-n6)
  • DGLA dihomo-gammalinolenic acid
  • ARA arachidonic acid
  • EPA eicosapentaenoic acid
  • DHA docosahexaenoic acid
  • U.S. Pat. No. 6,479,544 describes an invention in which it is found that ARA is highly desirable rather than undesirable and it may be helpful to administer ARA in association with EPA.
  • This invention provides pharmaceutical formulations containing eicosapentaenoic acid or any appropriate derivative (hereinafter collectively referred to as EPA) and arachidonic acid (ARA), as set out in the granted claims for this patent.
  • EPA eicosapentaenoic acid or any appropriate derivative
  • ARA arachidonic acid
  • ARA may be replaced by one or more of its precursors, DGLA or GLA.
  • the ratio of EPA to ARA is preferably between 1:1 and 20:1.
  • Patent application PCT/GB 2004/000242 describes the treatment or prevention of psoriasis with a formulation comprising more than 95% EPA and less than 2% DHA.
  • the EPA is replaced with DPA.
  • Patent application PCT/NL 2006/050291 (WO/2007/058538, GB 0301701.9) describes combinations of idigestible oligosaccharides and long chain poly-unsaturated fatty acids such as ARA, EPA, DA, and combinations thereof to improve intestinal barrier integrity, improving barrier function, stimulating gut maturation and/or reducing intestinal barrier permeability.
  • Holub et al. Lipids, 2011, 46:399-407 discloses a study assessing the effect of oral supplementation with docosapentaenoic acid (DPA) on levels of serum and tissue lipid classes and their fatty acid compositions in rat liver, heart, and kidney.
  • DPA docosapentaenoic acid
  • Cardiovascular disease includes numerous problems, many of which are related to a process called atherosclerosis.
  • Atherosclerosis is a condition that develops when plaque builds up in the walls of the arteries. This buildup narrows the arteries, making it harder for blood to flow through. If a blood clot forms, it can stop the blood flow. This can cause lead to conditions or event such as myocardial infarction, stroke, heart failure, arrhythmias, valve dysfunction, and death.
  • the present invention provides omega-3 fatty acid compositions and methods of administering these compositions.
  • the present invention provides a pharmaceutical composition
  • fatty acids comprising fatty acids, wherein at least 50% by weight of the fatty acids comprise omega-3 fatty acids, salts, esters, or derivatives thereof, wherein the omega-3 fatty acids comprise eicosapentaenoic acid (EPA) and docosapentaenoic acid (DPA) and wherein the ratio of docosahexaenoic acid to DHA to EPA (DHA:EPA) is less than 1:10, and wherein the ratio of DHA to DPA (DHA:DPA) is less than 2:1.
  • omega-3 fatty acids comprise eicosapentaenoic acid (EPA) and docosapentaenoic acid (DPA) and wherein the ratio of docosahexaenoic acid to DHA to EPA (DHA:EPA) is less than 1:10, and wherein the ratio of DHA to DPA (DHA:DPA) is less than 2:1.
  • EPA eicosap
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising eicosapentaenoic acid (EPA) in an amount between about 70% to about 95% of the total amount of fatty acids and docosapentaenoic acid (DPA), wherein the composition comprises no more than 5% docosahexaenoic acid (DHA) of the total amount of fatty acids, and wherein the ratio of DHA:DPA is 1:1 or lower.
  • EPA eicosapentaenoic acid
  • DPA docosapentaenoic acid
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising pharmaceutical composition comprising eicosapentaenoic acid (EPA) in an amount between about 750 mg/g to about 950 mg/g, and docosapentaenoic acid (DPA), wherein the composition comprises no more than 5% DHA of the total amount of fatty acids, and wherein the ratio of DHA:DPA is 1:1 or lower.
  • EPA eicosapentaenoic acid
  • DPA docosapentaenoic acid
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising eicosapentaenoic acid (EPA) in a daily dosage amount of between about 1000 mg to about 5000 mg, and further comprising docosapentaenoic acid (DPA) and docosahexaenoic acid (DHA), wherein the composition comprises no more than 5% DHA of the total amount of fatty acids, and wherein the ratio of DHA:DPA is 1:1 or lower.
  • EPA eicosapentaenoic acid
  • DPA docosapentaenoic acid
  • DHA docosahexaenoic acid
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising eicosapentaenoic acid (EPA) and docosapentaenoic acid (DPA), wherein the amount of EPA and DPA is about 55% or more by weight of the total amount of fatty acids, and wherein the ratio of DHA:DPA is no more than 1:1.
  • EPA eicosapentaenoic acid
  • DPA docosapentaenoic acid
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising: docosapentaenoic acid (DPA) in an amount between about 50% to about 80% of the total amount of fatty acids, docosahexaenoic acid (DHA) in an amount between about 25% to about 40% of the total amount of fatty acids, and optionally eicosapentaenoic acid (EPA) in an amount less than about 10% of the total amount of fatty acids.
  • DPA docosapentaenoic acid
  • DHA docosahexaenoic acid
  • EPA eicosapentaenoic acid
  • compositions of the present invention comprise additional fatty acids, such as heneicosapentaenoic acid (HPA), arachidonic acid (ARA), and omega-6-docosapentaenoic acid (n-6 DPA), tetracosapentaenoic acid (TPA), and/or gamma-linoleic acid (GLA).
  • HPA heneicosapentaenoic acid
  • ARA arachidonic acid
  • n-6 DPA omega-6-docosapentaenoic acid
  • TPA tetracosapentaenoic acid
  • GLA gamma-linoleic acid
  • the present invention provides methods comprising administering the compositions.
  • the present invention provides a method of treating, preventing, reducing the occurrence of, and improving symptoms associated with coronary heart disease (CHD), vascular disease, atherosclerotic disease or related conditions, comprising administering the compositions of the present invention to a subject in need thereof.
  • CHD coronary heart disease
  • the present invention also provides methods of treatment of patients by administering an effective amount of such compositions to a subject or a subject in need thereof, such as a subject prone to or afflicted with a vascular/cardiovascular disease or condition, a subject at risk of vascular/cardiovascular events, or a subject in need of treatment for a vascular/cardiovascular disease or condition.
  • the present invention provides an orally administrable composition
  • fatty acids wherein at least 50% by weight of the fatty acids comprise omega-3-fatty acids, salts, esters, or derivatives thereof, wherein the omega-3 fatty acids comprise eicosapentaenoic acid (EPA; C20:5-n3), docosapentaenoic acid (DPA; C22:5-n3), and docosahexaenoic acid (DHA; C22:6-n3), wherein the ratio of DHA to EPA (DHA:EPA) is less than 1:20, and wherein the ratio of DHA to DPA (DHA:DPA) is less than 2:1.
  • omega-3 fatty acids comprise eicosapentaenoic acid (EPA; C20:5-n3), docosapentaenoic acid (DPA; C22:5-n3), and docosahexaenoic acid (DHA; C22:6-n3), wherein the ratio of
  • the present invention provides a pharmaceutical composition
  • fatty acids comprising fatty acids, wherein at least 50% by weight of the fatty acids comprise omega-3 fatty acids, salts, esters, or derivatives thereof, wherein the omega-3 fatty acids comprise eicosapentaenoic acid (EPA) and docosapentaenoic acid (DPA) and wherein the ratio of docosahexaenoic acid to DHA to EPA (DHA:EPA) is less than 1:10, and wherein the ratio of DHA to DPA (DHA:DPA) is less than 2:1, and methods of using this composition.
  • the composition comprises EPA in an amount between about 70% and about 95% of the total amount of fatty acids.
  • the composition comprises less than about 5% of DHA of the total amount of fatty acids. In some embodiments, the composition comprises DPA in an amount of between about 5% and about 15% of the total amount of fatty acids. In some embodiments, the ratio of EPA to DPA (EPA:DPA) is less than about 1:1. In some embodiments. the composition further comprises heneicosapentaenoic acid (HPA) in an amount of at least 1% of the total amount of fatty acids
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising eicosapentaenoic acid (EPA) in an amount between about 70% to about 95% of the total amount of fatty acids and docosapentaenoic acid (DPA), wherein the composition comprises no more than 5% docosahexaenoic acid (DHA) of the total amount of fatty acids, and wherein the ratio of DHA:DPA is 1:1 or lower, and methods of using the composition.
  • the composition comprises DPA in an amount of between about 5% and about 15% of the total amount of fatty acids.
  • the composition further comprises heneicosapentaenoic acid (HPA) in an amount of at least 1% of the total amount of fatty acids.
  • the ratio of EPA to DPA EPA:DPA
  • the ratio of DHA:EPA is less than about 1:10.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising pharmaceutical composition comprising eicosapentaenoic acid (EPA) in an amount between about 750 mg/g to about 950 mg/g, and docosapentaenoic acid (DPA), wherein the composition comprises no more than 5% DHA of the total amount of fatty acids, and wherein the ratio of DHA:DPA is 1:1 or lower, and methods of using the composition.
  • the composition comprises about 60 mg/g to about 120 mg/g of DPA.
  • the ratio of DHA:EPA is less than 1:10.
  • the ratio of EPA:DPA is less than about 1:1.
  • the composition further comprises heneicosapentaenoic acid (HPA) in an amount of at least 1% of the total amount of fatty acids.
  • HPA heneicosapentaenoic acid
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising eicosapentaenoic acid (EPA) in a daily dosage amount of between about 1000 mg to about 5000 mg, and further comprising docosapentaenoic acid (DPA) and docosahexaenoic acid (DHA), wherein the composition comprises no more than 5% DHA of the total amount of fatty acids, and wherein the ratio of DHA:DPA is 1:1 or lower, and methods of using the composition.
  • the ratio of DHA:EPA is less than about 1:10.
  • the ratio of EPA:DPA is less than about 1:1.
  • the composition further comprises heneicosapentaenoic acid (HPA) in an amount of at least 1% of the total amount of fatty acids.
  • HPA heneicosapentaenoic acid
  • the present invention provides a pharmaceutical composition
  • EPA eicosapentaenoic acid
  • DPA docosapentaenoic acid
  • the composition comprises docosahexaenoic acid (DHA) in an amount of less than about 30% of the total amount of fatty acids.
  • the composition comprises a daily dosage of DPA of greater than about 120 mg/day.
  • the composition comprises omega-6 fatty acids in an amount of no more than 6% of total amount of fatty acids.
  • the composition comprises DPA in an amount of at least 6% of the total amount of fatty acids.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising: docosapentaenoic acid (DPA) in an amount between about 50% to about 80% of the total amount of fatty acids, docosahexaenoic acid (DHA) in an amount between about 25% to about 40% of the total amount of fatty acids, and optionally eicosapentaenoic acid (EPA) in an amount less than about 10% of the total amount of fatty acids, and methods of using the composition.
  • the composition comprises docosapentaenoic acid (DPA) in an amount between about 50% to 75%, alternatively about 50% to about 70%, alternatively about 50% to about 65%, or alternatively about 50% to about 60%, of the total amount of fatty acids.
  • the composition comprises docosahexaenoic acid (DHA) in an amount between about 25% to about 38%, alternatively about 25% to about 35%, alternatively about 30% to about 35% of the total amount of fatty acids.
  • the composition comprises eicosapentaenoic acid (EPA) in an amount less than about 9%, alternatively less than about 8%, alternatively less than about 7%, alternatively less than about 6%, alternatively less than about 5%, of the total amount of fatty acids.
  • DHA docosahexaenoic acid
  • EPA eicosapentaenoic acid
  • compositions of the present invention comprise at least 50% omega-3 fatty acids, alternatively at least 55%, alternatively at least 60%, alternatively at least 65%, alternatively at least 70%, alternatively at least 75%, alternatively at least 80%, alternatively at least 85%, alternatively at least 95%, most preferably at least 90% omega-3 fatty acids of the total amount of fatty acids.
  • the composition comprises at least about 92% to about 99%, alternatively about 93% to about 98%, alternatively about 94% to about 98%, omega-3 fatty acids of the total amount of fatty acids.
  • EPA and DPA are jointly present in the compositions of the present invention at between about 55% and about 100% of total fatty acids, alternatively between about 60% and about 100%, alternatively between about 65% and about 100%, alternatively between about 70% and about 100%, alternatively between about 75% and about 100%, alternatively between about 80% and about 100%, alternatively between about 85% and about 95%, alternatively about 85% to about 100%, alternatively between about 85% and about 97%, alternatively between about 88% and about 95%, alternatively between about 88% and about 97%, alternatively about 88% to about 100%, alternatively between about 90% and about 95%, alternatively between about 90% and about 97%, alternatively about 90% to about 100%, alternatively about 95% to about 100%, alternatively about 97% to about 100% of the total amount of fatty acids.
  • the compositions of the present invention may be provided in a dose of between 100 mg and 10,100 mg/day, alternatively between 200 mg and 8,100 mg/day, alternatively between 300 mg and 6,100 mg/day, alternatively between 400 mg and 5,100 mg/day, alternatively between 500 mg and 4,100 mg/day.
  • the compositions and methods of the present invention may be provided in a dose of between 100 mg and 10,100 mg/day, alternatively between 200 mg and 8,100 mg/day, alternatively between 300 mg and 6,100 mg/day, alternatively between 400 mg and 5,100 mg/day, alternatively between 500 mg and 4,100 mg/day.
  • the methods and compositions of the present invention may be provided in a dose of between 100 mg and 10,100 mg/day, alternatively between 200 mg and 8,100 mg/day, alternatively between 300 mg and 6,100 mg/day, alternatively between 400 mg and 5,100 mg/day, alternatively between 500 mg and 4,100 mg/day.
  • the fatty acids such as EPA and DPA
  • the fatty acids may be present in free fatty acid form, or as a salt, ester, or derivative.
  • the fatty acids are preferably composed as a triglyceride, an ester (such as an ethyl ester) or free fatty acid.
  • Other forms of the fatty acids which may be useful include salts, esters of any type, amides, mono-, di- or triglycerides, phospholipids or any other form which can lead to metabolization of the fatty acids (such as EPA and/or DPA), or the incorporation of the fatty acids (such as EPA and/or DPA) into body fluids, tissues or organs.
  • Omega-3 fatty acids may be grouped by the number of double bonds contained in the fatty acid chain.
  • HTA hexadecatrienoic acid
  • alpha-linolenic acid (ALA) and eicosatrienoic acid (ETE) are omega-3-trienoic acids
  • SDA stearidonic acid
  • ETA eicosatetraenoic acid
  • EPA heneicosapentaenoic acid
  • DPA and tetracosapentaenoic acid TPA
  • DHA and tetracosahexaenoic acid (THA) are omega-3-hexaenoic acids.
  • omega-3-pentaenoic acids will refer to a mixture of at least two omega-3 pentaenoic acids in a ratio of at least 1:25, more preferably in a ratio of at least 1:50, more preferably in a ratio of at least 1:75, more preferably in a ratio of at least 1:100, more preferably in a ratio of at least 1:125, more preferably in a ratio of at least 1:150, more preferably in a ratio of at least 1:200.
  • the ratio refers to the ratio of the least prevalent omega-3 pentaenoic acid in the mixture to the most prevalent omega-3 pentaenoic acid in the mixture.
  • compositions of the present invention comprise EPA, HPA, DPA and TPA, alternatively EPA and DPA, and alternatively the compositions of the present invention comprise EPA, HPA and DPA.
  • the omega-3-pentaenoic acids in the compositions of the present invention comprise no more than 99.5% of a single omega-3-pentaenoic acid, alternatively no more than 99%; alternatively no more than 98.5%; alternatively no more than 98%; alternatively no more than 97.5%; alternatively no more than 96%; alternatively no more than 95%; alternatively no more than 94%; alternatively no more than 93%; alternatively no more than 92%; alternatively no more than 91%; alternatively no more than 90%; alternatively no more than 88%; alternatively no more than 85%; alternatively no more than 80%; alternatively no more than 75%; alternatively no more than 70%; alternatively no more than 65%; alternatively no more than 60%; alternatively no more than 55%; alternatively no more than 50%; alternatively no more than 45%; alternatively no more than 40%; alternatively no more than 30%.
  • compositions of the present invention wherein at least 10%, alternatively at least 20%, alternatively at least 25%, alternatively at least 35%, alternatively at least 50%, alternatively at least 60%, alternatively at least 65%, alternatively at least 70%, alternatively at least 75%, by weight of the fatty acids comprise omega-3-pentaenoic acids, salts, esters, or derivatives thereof.
  • compositions and methods comprise significant amounts of omega-3-pentaenoic acids or their glycerol or ethyl esters may be used in the methods of the present invention.
  • the compositions and methods comprise at least 100 mg omega-3-pentaenoic acids per day, alternatively at least 200 mg omega-3-pentaenoic acids per day, alternatively at least 300 mg omega-3-pentaenoic acids per day, alternatively at least 500 mg omega-3-pentaenoic acids per day, alternatively at least 700 mg omega-3-pentaenoic acids per day, alternatively at least 900 mg omega-3-pentaenoic acids per day, alternatively at least 1000 mg omega-3-pentaenoic acids per day, alternatively at least 1500 mg omega-3-pentaenoic acids per day, alternatively at least 1900 mg omega-3-pentaenoic acids per day, alternatively at least 2000 mg omega-3-pentaenoic acids per day, alternatively at least 2500
  • the compositions provide a DHA as compared to the amount of omega-3-pentaenoic acids (N3-5enoicFA) such that the DHA:N3-5enoicFA ratio is no more than 15:1 of DHA:N3-5enoicFA, alternatively no more than 12:1 of DHA:N3-5enoicFA, alternatively no more than 10:1 of DHA:N3-5enoicFA, alternatively no more than 8:1 of DHA:N3-5enoicFA, alternatively no more than 5:1 of DHA:N3-5enoicFA, alternatively no more than 3:1 of DHA:N3-5enoicFA, alternatively no more than 2:1 of DHA:N3-5enoicFA, alternatively no more than 1:1 of DHA:N3-5enoicFA, alternatively no more than 1:2 of DHA:N3-5enoicFA, alternatively no more than 1:3 of DHA:N3-5enoicFA, alternatively no more than 1:5 of DHA:N3-5enoicFA, alternatively no more
  • the compositions of the present invention comprise at least 0.01% HPA of total fatty acids in the composition, alternatively at least 0.05% HPA, alternatively at least 0.10% HPA, alternatively at least 0.15% HPA, alternatively at least 0.2% HPA, alternatively at least 0.3% HPA, alternatively at least 0.4% HPA, alternatively at least 0.5% HPA, alternatively at least 0.75% HPA, alternatively at least 1% HPA, alternatively at least 1.5% HPA, alternatively at least 2% HPA, alternatively at least 2.5% HPA, alternatively at least 3% HPA, alternatively at least 3.5% HPA, alternatively at least 4% HPA, alternatively at least 4.5% HPA, alternatively at least 5% HPA, alternatively at least 6% HPA, alternatively at least 7% HPA, alternatively the compositions of the present invention comprise at least 9% HPA of total fatty acids in the composition.
  • the compositions of the present invention comprise no more than 20% HPA of total fatty acids in the composition, alternatively no more than 15% HPA, alternatively no more than 12% HPA, alternatively no more than 10% HPA, alternatively no more than 8% HPA, alternatively no more than 7% HPA, alternatively no more than 6% HPA, alternatively no more than 5% HPA, alternatively no more than 4% HPA, alternatively no more than 3% HPA, alternatively no more than 2% HPA, alternatively no more than 1.5% HPA, alternatively the compositions of the present invention comprise at least 1% HPA of total fatty acids in the composition. In some embodiments, the compositions of the present invention comprise 1% to 20% HPA of the total fatty acids in the composition.
  • compositions of the present invention comprise about 1% to about 6% HPA, alternatively about 2% to about 5% HPA, alternatively about 3% to about 4% HPA, relative to the total amount of fatty acids in the composition. In some embodiments, the compositions of the present invention comprise about 10 mg/g to about 50 mg/g HPA, alternatively about 15 mg/g to about 45 mg/g, alternatively about 20 mg/g to about 40 mg/g, alternatively about 25 mg/g to about 35 mg/g, alternatively about 30 mg/g HPA.
  • the present invention provides compositions and methods which comprise significant amounts of omega-3 heneicosapentaenoic acid (HPA) or its glycerol or ethyl esters.
  • the methods of treatment may provide to a subject in need thereof a dose of at least 10 mg HPA per day, alternatively at least 15 mg HPA per day, alternatively at least 20 mg HPA per day, alternatively at least 25 mg HPA per day, alternatively at least 30 mg HPA per day, alternatively at least 40 mg HPA per day, alternatively at least 50 mg HPA per day, alternatively at least 60 mg HPA per day, alternatively at least 70 mg HPA per day, alternatively at least 80 mg HPA per day, alternatively at least 90 mg HPA per day, alternatively at least 100 mg HPA per day, alternatively at least 120 mg HPA per day, alternatively at least 150 mg HPA per day, alternatively at least 160 mg HPA per day, alternatively at least 180 mg HPA per day, alternatively at least 200 mg
  • compositions of the present invention comprise no more than 10% omega-3 fatty acids that are not omega-3-pentaenoic acids, alternatively no more than 9%, alternatively no more than 8%, alternatively no more than 7%, alternatively no more than 6%, alternatively no more than 5%, alternatively no more than 4.5%, alternatively no more than 4%, alternatively no more than 3.5%, alternatively no more than 3%, alternatively no more than 2.5%, alternatively no more than 2%, alternatively no more than 1.5%, alternatively no more than 1.25%, alternatively no more than 1%, alternatively no more than 0.75%, alternatively no more than 0.5%, alternatively no more than 0.4%, alternatively no more than 0.3%, alternatively no more than 0.2%, alternatively the compositions of the present invention comprise no more than 0.1% omega-3 fatty acids that are not omega-3-pentaenoic acids.
  • compositions comprise EPA and DPA in an EPA:DPA ratio between 99:1 and 1:99 EPA:DPA, alternatively between 90:1 and 1:90, alternatively between 60:1 and 1:60, alternatively between 60:1 and 1:20, alternatively between 60:1 and 1:4, alternatively between 40:1 and 1:20, alternatively between 30:1 and 1:20, alternatively between 30:1 and 1:10, alternatively between 30:1 and 1:5, alternatively between 40:1 and 1:4, alternatively between 30:1 and 1:4, alternatively between 30:1 and 1:2, alternatively between 30:1 and 1:1, alternatively between 30:1 and 2:1, alternatively between 30:1 and 5:1, alternatively between 20:1 and 1:20, alternatively between 20:1 and 1:10, alternatively between 20:1 and 1:5, alternatively between 20:1 and 1:2, alternatively between 20:1 and 1:1, alternatively between 20:1 and 2:1, alternatively between 20:1 and 5:1, alternatively between 20:1 and 2:1, alternatively between 20:
  • the ratio of EPA:DPA is greater than 1:1, preferably greater than 2:1, and more preferably greater than 5:1. In some embodiments, the ratio of EPA:DPA is 1:1 to 25:1, preferably 5:1 to 20:1, more preferably 8:1 to 15:1, even more preferably 9:1 to 13:1, even more most preferably about 10:1 to 11:1, and most preferably about 10:1.
  • the compositions comprise EPA in an amount between 55% and 95% relative to the total amount of fatty acids present in the composition, alternatively between 60% and 95%, alternatively between 65% and 95%, alternatively between 70% and 95%, alternatively between 75% and 95%, alternatively between 90% and 95%, alternatively between 80% and 95%, alternatively between 90% and 95%, alternatively between 55% and 90%, alternatively between 60% and 90%, alternatively between 65% and 90%, alternatively between 70% and 90%, alternatively between 75% and 90%, alternatively between 80% and 90%, alternatively between 85% and 90%, alternatively between 55% and 92%, alternatively between 60% and 92%, alternatively between 65% and 92%, alternatively between 70% and 92%, alternatively between 75% and 92%, alternatively between 80% and 92%, alternatively between 85% and 92%, alternatively between 55% and 93%, alternatively between 60% and 93%, alternatively between 65% and 93%, alternatively between 70% and 93%, alternatively between 75% and 93%, alternatively between 60% and 93%,
  • the compositions comprise about 70% to about 95%, 80% to about 90%, alternatively about 81% to about 88%, alternatively about 82% to about 88%, alternatively about 83% to about 87%, alternatively about 84% to about 86%, alternatively about 85% EPA relative to the total amount of fatty acids present in the composition.
  • the compositions comprise about 750 mg/g to about 950 mg/g, alternatively about 800 mg/g to about 900 mg/g, alternatively about 830 mg/g to about 870 mg/g, alternatively about 840 mg/g to about 870 mg/g, alternatively 845 mg/g to about 865 mg/g, alternatively 846 mg/g to about 860 mg/g, alternatively 847 mg/g to about 859 mg/g, alternatively about 848 mg/g to about 858 mg/g, alternatively about 849 mg/g to about 857 mg/g, alternatively about 850 mg/g to about 856 mg/g, alternatively about 851 mg/g to about 855 mg/g, alternatively about 852 mg/g to about 854 mg/g, alternatively about 853 mg/g EPA.
  • compositions of the present invention are preferably provided in a dose of between 100 mg and 10,000 mg/day, alternatively between 200 mg and 8,000 mg/day, alternatively between 300 mg and 6,000 mg/day, alternatively between 400 mg and 5,000 mg/day, alternatively between 500 mg and 4,000 mg/day.
  • compositions and methods of the present invention are provided in a dose of between about 1000 mg/day to about 5000 mg/day, alternatively about 1200 mg/day to about 3000 mg/day, alternatively about 1500 mg/day to about 2500 mg/day, alternatively 1600 mg/day to about 1950 mg/day, alternatively about 1735 mg/day to about 1855 mg/day, alternatively about 1740 mg/day to about 1840 mg/day, alternatively about 1745 mg/day to about 1820 mg/day, alternatively about 1750 mg/day to about 1800 mg/day, alternatively about 1755 mg/day to about 1790 mg/day, alternatively about 1760 mg/day to about 1780 mg/day, alternatively about 1770 mg/day of EPA.
  • the compostions of the present invention are provided in a dose of between about 2300 mg/day to about 3000 mg/day, alternatively about 2400 mg/day to about 2800 mg/day, alternatively about 2520 mg/day to about 2780 mg/day, alternatively about 2600 mg/day to about 2700 mg/day, alternatively about 2610 mg/day to about 2680 mg/day, alternatively about 2620 mg/day to about 2670 mg/day, alternatively about 2630 mg/day to about 2665 mg/day, alternatively about 2640 mg/day to about 2660 mg/day, alternatively about 2650 mg/day of EPA.
  • the compostions of the present invention are provided in a dose of between about 3200 mg/day to about 3900 mg/day, alternatively 3300 mg/day to about 3800 mg/day, alternatively 3360 mg/day to about 3710 mg/day, alternatively about 3400 mg/day to about 3700 mg/day, alternatively about 3450 mg/day to about 3650 mg/day, alternatively about 3500 mg/day to about 3600 mg/day, alternatively about 3530 mg/day to about 3580 mg/day, alternatively about 3540 mg/day to about 3560 mg/day, alternatively about 3550 mg/day of EPA.
  • compositions of the present invention are provided in a dose of between about 1650 mg/day to about 2050 mg/day, alternatively about 1700 mg/day to about 2000 mg/day, alternatively about 1750 mg/day to about 1950 mg/day, alternatively about 1775 mg/day to about 1925 mg/day, alternatively about 1800 mg/day to about 1900 mg/day, alternatively about 1800 mg/day to about 2000 mg/day, alternatively about 1820 mg/day to about 1880 mg/day, alternatively about 1830 mg/day to about 1870 mg/day, alternatively about 1840 mg/day to about 1860 mg/day, alternatively about 1850 mg/day of EPA.
  • the compostions of the present invention are provided in a dose of between about 2500 mg/day to about 3100 mg/day, alternatively about 2600 mg/day to about 2000 mg/day, alternatively about 2650 mg/day to about 2950 mg/day, alternatively about 2700 mg/day to about 2900 mg/day, alternatively about 2725 mg/day to about 2875 mg/day, alternatively about 2750 mg/day to about 2850 mg/day, alternatively about 2780 mg/day to about 2820 mg/day, alternatively about 2790 mg/day to about 2810 mg/day, alternatively about 2800 mg/day of EPA.
  • the compostions of the present invention are provided in a dose of between about 3300 mg/day to about 4000 mg/day, alternatively about 3400 mg/day to about 3900 mg/day, alternatively about 3500 mg/day to about 3900 mg/day, alternatively about 3550 mg/day to about 3850 mg/day, alternatively about 3600 mg/day to about 3800 mg/day, alternatively about 3650 mg/day to about 3750 mg/day, alternatively about 3680 mg/day to about 3725 mg/day, alternatively about 3690 mg/day to about 3710 mg/day, alternatively about 3700 mg/day of EPA.
  • compositions of the present invention are provided in a dose of between about 1500 mg/day to about 2500 mg/day, alternatively about 1750 mg/day to about 2300 mg/day, alternatively about 1800 mg/day to about 2200 mg/day, alternatively about 1900 mg/day to about 2100 mg/day, alternatively about 1950 mg/day to about 2050 mg/day, alternatively about 1975 mg/day to about 2025 mg/day, alternatively about 2000 mg/day of EPA.
  • compositions of the present invention are provided in a dose of between about 2500 mg/day to about 3500 mg/day, alternatively about 2700 mg/day to about 3300 mg/day alternatively about 2750 mg/day to about 3300 mg/day, alternatively about 2800 mg/day to about 3200 mg/day, alternatively about 2900 mg/day to about 3100 mg/day, alternatively about 2950 mg/day to about 3050 mg/day, alternatively about 2975 mg/day to about 3025 mg/day, alternatively about 3000 mg/day of EPA.
  • compositions of the present invention are provided in a dose of between about 3500 mg/day to about 4500 mg/day, alternatively about 3700 mg/day to about 4300 mg/day, alternatively about 3750 mg/day to about 4300 mg/day, alternatively about 3800 mg/day to about 4200 mg/day, alternatively about 3900 mg/day to about 4100 mg/day, alternatively about 3950 mg/day to about 4050 mg/day, alternatively about 3975 mg/day to about 4025 mg/day, alternatively about 4000 mg/day of EPA.
  • the compositions comprise DPA in an amount between 1% and 99% relative to the total amount of fatty acids present in the composition, alternatively between 1% and 95%, alternatively alternatively between 1% and 90%, alternatively between 1% and 85%, alternatively between 1% and 80%, alternatively between 1% and 75%, alternatively between 1% and 70%, alternatively between 1% and 65%, alternatively between 1% and 60%, alternatively between 1% and 55%, alternatively between 1% and 50%, alternatively between 1% and 45%, alternatively between 1% and 40%, alternatively between 1% and 35%, alternatively between 1% and 30%, alternatively between 1% and 25%, alternatively between 1% and 20%, alternatively between 1% and 15%, alternatively between 1% and 10%, alternatively between 1% and 5%, alternatively between 2% and 99%, alternatively between 2% and 95%, alternatively between 2% and 90%, alternatively between 2% and 85%, alternatively between 2% and 80%, alternatively between 2% and 75%, alternatively between 1% and 95%,
  • compositions comprise docosapentaenoic acid (DPA) in an amount between about 5% to about 15%, alternatively about 6% to about 12%, alternatively about 7% to about 11%, alternatively about 8% to about 10% relative to the total amount of fatty acids present in the composition.
  • DPA docosapentaenoic acid
  • the composition comprises at least about 4% or at least about 5% or at least about 6% or at least about 7% or at least about 8% or at least about 9% or at least about 10% or at least about 15% or at least about 20% or at least about 25% or at least about 30% or at least about 35% or at least about 40% or at least about 45% or at least about 50% or at least about 55% or at least about 60% or at least about 65% or at least about 70% or at least about 75% or at least about 80% or at least about 85% or at least about 90% or at least about 95% of docosapentaenoic acid (DPA).
  • DPA docosapentaenoic acid
  • compositions comprise docosapentaenoic acid (DPA) in an amount of about 60 mg/g to about 120 mg/g, alternatively about 70 mg/g to about 100 mg/g, 75 mg/g to about 90 mg/g, alternatively about 77 mg/g to about 85 mg/g, alternatively about 78 mg/g to about 84 mg/g, alternatively about 79 mg/g to about 83 mg/g, alternatively about 80 mg/g to about 82 mg/g, alternatively about 81 mg/g to about 82 mg/g.
  • DPA docosapentaenoic acid
  • compositions comprise docosapentaenoic acid (DPA) in an daily dosage amount of at least about 20 mg/day, alternatively at least about 25 mg/day, alternatively at least about 30 mg/day, alternatively at least about 40 mg/day, alternatively at least about 50 mg/day, alternatively at least about 60 mg/day alternatively, at least about 70 mg/day alternatively at least about 75 mg/day, alternatively at least about 80 mg/day, alternatively at least about 90 mg/day, alternatively at least about 100 mg/day, alternatively at least about 120 mg/day, alternatively at least about 150 mg/day, alternatively at least about 160 mg/day, alternatively at least about 180 mg/day, alternatively at least about 200 mg/day, alternatively at least about 250 mg/day, alternatively at least about 300 mg/day, alternatively at least about 350 mg/day, or alternatively at least about 400 mg/day, alternatively at least about 500 mg/day, alternatively at least about 600 mg/day, alternatively at least about 800 mg
  • DPA do
  • the composition comprises DPA in a daily dosage of about 120 mg/day to about 150 mg/day, alternatively about 150 mg/day to about 200 mg/day, alternatively about 200 mg/day to about 250 mg/day, alternatively about 250 mg/day to about 300 mg/day, alternatively about 300 mg/day to about 400 mg/day, alternatively about 400 mg/day to about 600 mg/day, alternatively about 600 mg/day to about 1000 mg/day.
  • the method of treatment provides a dose of at least about 1 mg/kg of docosapentaenoic acid (DPA) per day, alternatively about 2 mg/kg of DPA per day, alternatively about 3 mg/kg of DPA per day, alternatively about 4 mg/kg of DPA per day, alternatively about 6 mg/kg of DPA per day, alternatively about 8 mg/kg of DPA per day, alternatively about 10 mg/kg of DPA per day, alternatively about 20 mg/kg of DPA per day, alternatively about 30 mg/kg of DPA per day, and alternatively about 40 mg/kg of DPA per day, alternatively about 50 mg/kg of DPA per day, alternatively about 75 mg/kg of DPA per day, and alternatively about 100 mg/kg of DPA per day.
  • DPA docosapentaenoic acid
  • compositions of the present invention comprise no more than 1:1 of DHA:EPA, alternatively no more than 1:2, alternatively no more than 1:3, alternatively no more than 1:3, alternatively no more than 1:4, alternatively no more than 1:5 of DHA:EPA, alternatively no more than 1:6 of DHA:EPA, alternatively no more than 1:7 of DHA:EPA, alternatively no more than 1:8 of DHA:EPA, alternatively no more than 1:9 of DHA:EPA, alternatively no more than 1:10 of DHA:EPA, alternatively no more than 1:12 of DHA:EPA, alternatively no more than 1:15 of DHA:EPA, alternatively no more than 1:20 of DHA:EPA, alternatively no more than 1:25 of DHA:EPA, alternatively no more than 1:30 of DHA:EPA, alternatively no more than 1:40 of DHA:EPA, alternatively
  • DHA may be present in the compositions of this invention at a relative amount of ratio less than 1% than the amount of EPA.
  • DHA docosahexaenoic acid
  • DHA may be present in the compositions of this invention at a DHA:EPA ratio of less than 1:99.
  • a relatively small amount of docosahexaenoic acid (DHA) relative to the total amount of fatty acids present in the composition is present.
  • the compositions of the present invention comprise no more than 30% DHA, alternatively no more than 20% DHA, alternatively no more than 15% DHA, alternatively no more than 12% DHA, alternatively no more than 10% DHA, alternatively no more than 9% DHA, alternatively no more than 8% DHA, alternatively no more than 7% DHA, alternatively no more than 6% DHA, alternatively no more than 5% DHA, alternatively no more than 4% DHA, alternatively no more than 3% DHA, alternatively no more than 2% DHA, alternatively no more than 1% DHA relative to the total amount of fatty acids present in the composition.
  • the compositions and methods comprise less than 1500 mg of DHA, alternatively less than 1200 mg of DHA, alternatively less than 1000 mg of DHA, alternatively less than 800 mg of DHA, alternatively less than 700 mg of DHA, alternatively less than 600 mg of DHA, alternatively less than 500 mg of DHA, alternatively less than 400 mg of DHA, alternatively less than 350 mg of DHA, alternatively less than 300 mg of DHA, alternatively less than 250 mg of DHA, alternatively less than 200 mg of DHA alternatively less than 150 mg of DHA, alternatively less than 120 mg of DHA, alternatively less than 100 mg of DHA, alternatively less than 80 mg of DHA, alternatively less than 60 mg of DHA, alternatively less than 40 mg of DHA, alternatively less than 30 mg of DHA, alternatively less than 25 mg of DHA, alternatively less than 20 mg of DHA or its glycerol or ethyl esters as a totally daily dose.
  • the composition comprises about 5 mg/g to about 20 mg/g, alternatively about 8 mg/g to about 18 mg/g, alternatively about 9 mg/g to about 15 mg/g, alternatively about 10 mg/g to about 14 mg/g, alternatively about 11 mg/g to about 13 mg/g, alternatively about 12 mg/g to about 13 mg/g of docosahexaenoic acid (DHA).
  • DHA docosahexaenoic acid
  • the ratio of EPA:HPA is about 1500:1 to 25:1, alternatively 1000:1 to 50:1, alternatively 800:1 to 60:1, alternatively 500:1 to 60:1, alternatively 250:1 to 75:1, and alternatively 100:1 to 80:1. In some preferred embodiments, the ratio of EPA:HPA is about 85:1. In some preferred embodiments, the ratio of EPA:HPA is about 30:1. In some embodiments, the ratio of DPA:HPA is about 250:1 to 1:1, alternatively 200:1 to 2:1, alternatively 150:1 to 3:1, alternatively 100:1 to 4:1, alternatively 50:1 to 5:1, alternatively 25:1 to 6:1, and alternatively 10:1 to 7:1. In some preferred embodiments, the ratio of DPA:HPA is about 8:1. In some embodiments, the ratio of DPA:HPA is about 3:0.
  • compositions of the present invention comprise no more than 15:1 of DHA:DPA, alternatively no more than 12:1 of DHA:DPA, alternatively no more than 10:1 of DHA:DPA, alternatively no more than 8:1 of DHA:DPA, alternatively no more than 5:1 of DHA:DPA, alternatively no more than 4:1 of DHA:DPA, alternatively no more than 3:1 of DHA:DPA, alternatively no more than 2:1 of DHA:DPA, alternatively no more than 1:1 of DHA:DPA, alternatively no more than 1:2 of DHA:DPA, alternatively no more than 1:3 of DHA:DPA, alternatively no more than 1:4 of DHA:DPA, alternatively no more than 1:5 of DHA:DPA, alternatively no more than 1:6 of DHA:DPA, alternatively no more than 1:7 of DHA:DPA, alternatively no more than 1:
  • compositions of the present invention comprise no more than 15:1 of DHA:HPA, alternatively no more than 12:1 of DHA:HPA, alternatively no more than 10:1 of DHA:HPA, alternatively no more than 8:1 of DHA:HPA, alternatively no more than 5:1 of DHA:HPA, alternatively no more than 3:1 of DHA:HPA, alternatively no more than 2:1 of DHA:HPA, alternatively no more than 1:1 of DHA:HPA, alternatively no more than 1:2 of DHA:HPA, alternatively no more than 1:3 of DHA:HPA, alternatively no more than 1:5 of DHA:HPA, alternatively no more than 1:8 of DHA:HPA, alternatively no more than 1:10 of DHA:HPA, alternatively no more than 1:15 of DHA:HPA, alternatively no more than 1:20 of DHA:HPA, alternatively no more than 1:
  • compositions of the present invention comprise no more than 10% omega-6 fatty acids relative to the total amount of fatty acids, alternatively no more than 9%, alternatively no more than 8%, alternatively no more than 7%, alternatively no more than 6%, alternatively no more than 5%, alternatively no more than 4.5%, alternatively no more than 4%, alternatively no more than 3.5%, alternatively no more than 3%, alternatively no more than 2.5%, alternatively no more than 2%, alternatively no more than 1.7%, alternatively no more than 1.5%, alternatively no more than 1.2%, alternatively no more than 1%, alternatively no more than 0.5% omega-6 fatty acids versus the total amount of fatty acids comprised by the compositions of the present invention.
  • Omega-6 fatty acids include, but are not limited to: linoleic acid (LA; C18:2-n6); gamma-linoleic acid (GLA; C18:3-n6); eicosadienoic acid (C20:2-n6); dihomo-gamma-linoleic acid (DGLA; C20:3-n6); arachiconic acid (ARA; C20:4-n6); and omega-6 docosapentaenoic acid (DPA; C22:5-n6).
  • LA linoleic acid
  • GLA gamma-linoleic acid
  • C20:2-n6 eicosadienoic acid
  • DGLA dihomo-gamma-linoleic acid
  • ARA arachiconic acid
  • DPA omega-6 docosapentaenoic acid
  • compositions of the present invention comprise no more than 10% omega-6 fatty acids relative to the total amount of omega-3 fatty acids plus omega-6 fatty acids, alternatively no more than 9%, alternatively no more than 8%, alternatively no more than 7%, alternatively no more than 6%, alternatively no more than 5%, alternatively no more than 4.5%, alternatively no more than 4%, alternatively no more than 3.5%, alternatively no more than 3%, alternatively no more than 2.5%, alternatively no more than 2%, alternatively no more than 1.7%, alternatively no more than 1.5%, alternatively no more than 1.2%, alternatively no more than 1%, alternatively no more than 0.5% omega-6 fatty acids versus the total amount of omega-3 fatty acids plus omega-6 fatty acids comprised by the compositions of the present invention.
  • compositions of the present invention comprise no more than 8% arachidonic acid (ARA; C20:4-n6) relative to the total amount of omega-3 fatty acids plus omega-6 fatty acids, alternatively no more than 7%, alternatively no more than 6%, alternatively no more than 5%, alternatively no more than 4.5%, alternatively no more than 4%, alternatively no more than 3.5%, alternatively no more than 3%, alternatively no more than 2.5%, alternatively no more than 2%, alternatively no more than 1.7%, alternatively no more than 1.5%, alternatively no more than 1.2%, alternatively no more than 1%, alternatively no more than 0.5% arachidonic acid (ARA; 020:4-n6) versus the total amount of omega-3 fatty acids plus omega-6 fatty acids comprised by the compositions of the present invention.
  • ARA arachidonic acid
  • a relatively small amount of omega-3 fatty acids in aggregate other than EPA, ETA, HPA and DPA (alternatively indicated as non-EPA, non-ETA, non-HPA and non-DPA omega-3 fatty acids in aggregate) relative to the total amount of fatty acids present in the composition is present.
  • compositions of the present invention comprise no more than 20% non-EPA, non-ETA, non-H PA and non-DPA omega-3 fatty acids, alternatively no more than 15% non-EPA, non-ETA, non-HPA and non-DPA omega-3 fatty acids, alternatively no more than 12% non-EPA, non-ETA, non-H PA and non-DPA omega-3 fatty acids, alternatively no more than 10% non-EPA, non-ETA, non-HPA and non-DPA omega-3 fatty acids, alternatively no more than 8% non-EPA, non-ETA, non-HPA and non-DPA omega-3 fatty acids, alternatively no more than 7% non-EPA, non-ETA, non-HPA and non-DPA omega-3 fatty acids, alternatively no more than 6% non-EPA, non-ETA, non-HPA and non-DPA omega-3 fatty acids, alternatively no more than 5% non-EPA, non-ETA, non-HPA and non-DPA omega-3 fatty acids, alternatively no more than 4%
  • a relatively small amount of the sum of ALA, SDA and DHA relative to the total amount of fatty acids present in the composition is present, while at the same time large amounts of the sum of EPA, DPA-n3, HPA and ETA are present.
  • compositions of the present invention comprise no more than 20% of the sum of ALA, SDA and DHA, alternatively no more than 15% of the sum of ALA, SDA and DHA, alternatively no more than 12% of the sum of ALA, SDA and DHA, alternatively no more than 10% of the sum of ALA, SDA and DHA, alternatively no more than 8% of the sum of ALA, SDA and DHA, alternatively no more than 7% of the sum of ALA, SDA and DHA, alternatively no more than 6% of the sum of ALA, SDA and DHA, alternatively no more than 5% of the sum of ALA, SDA and DHA, alternatively no more than 4% of the sum of ALA, SDA and DHA, alternatively no more than 3% of the sum of ALA, SDA and DHA, alternatively no more than 2% of the sum of ALA, SDA and DHA, alternatively no more than 1% of the sum of ALA, SDA and DHA relative to the total amount of fatty
  • compositions of the present invention comprise no more than 8% arachidonic acid (ARA; C20:4-n6) relative to the total amount of fatty acids, alternatively no more than 7%, alternatively no more than 6%, alternatively no more than 5%, alternatively no more than 4.5%, alternatively no more than 4%, alternatively no more than 3.5%, alternatively no more than 3%, alternatively no more than 2.5%, alternatively no more than 2%, alternatively no more than 1.7%, alternatively no more than 1.5%, alternatively no more than 1.2%, alternatively no more than 1%, alternatively no more than 0.5% arachidonic acid (ARA; C20:4-n6) relative the total amount of fatty acids comprised by the compositions of the present invention.
  • ARA arachidonic acid
  • compositions of the present invention comprise no more than 2.5% arachidonic acid (ARA; C20:4-n6), no more than 0.4% omega-6-docosapentaenoic acid (DPA; C22:5-n6) and no more than 0.2% gamma-linoleic acid (GLA; C18:3-n6) relative the total amount of fatty acids comprised by the compositions of the present invention.
  • ARA arachidonic acid
  • DPA omega-6-docosapentaenoic acid
  • GLA gamma-linoleic acid
  • fatty acid compositions comprising no more than 2.5% arachidonic acid (ARA; C20:4-n6), no more than 0.3% omega-6 docosapentaenoic acid (DPA; C22:5-n6) and no more than 0.1% gamma-linoleic acid (GLA; C18:3-n6) relative the total amount of fatty acids comprised, by the compositions of the present invention.
  • ARA arachidonic acid
  • DPA docosapentaenoic acid
  • GLA gamma-linoleic acid
  • the composition of the present invention further comprises TPA at concentration of at least 0.05%.
  • the TPA concentration is about 0.01% to about 5%, alternatively about 0.05% to about 2%, alternatively about 0.1% to about 1%, alternatively about 0.2% to about 0.8%, alternatively about 0.4% to about 0.6%, alternatively about 0.5%.
  • compositions of the present invention may also be taken as a general nutritional supplement.
  • the active ingredient of the formulations of the present invention consists essentially wholly of the EPA and DPA or precursors thereof (ethyl ester, triglyceride, or any other pharmaceutically acceptable salt or derivative thereof).
  • no large amounts preferably less than 15%, alternatively less than 12%, alternatively less than 10%, alternatively less than 9%, alternatively less than 8%, alternatively less than 7%, alternatively less than 6%, alternatively less than 5%, alternatively less than 4%, alternatively less than 3%, alternatively less than 2%, alternatively less than 1%, alternatively less than 0.5%, alternatively less than 0.25%
  • any other fatty acids are present.
  • the active ingredient of the formulations of the present invention consists essentially wholly of omega-3-pentaenoic acids or precursors thereof (ethyl ester, triglyceride, or any other pharmaceutically acceptable salt or derivative thereof).
  • no large amounts preferably less than 15%, alternatively less than 12%, alternatively less than 10%, alternatively less than 9%, alternatively less than 4%, alternatively less than 4%, alternatively less than 4%, alternatively less than 4%, alternatively less than 4%, alternatively less than 4%, alternatively less than 4%, alternatively less than 3%, alternatively less than 2%, alternatively less than 1%, alternatively less than 0.5%, alternatively less than 0.25%
  • any other fatty acids are present.
  • the fatty acid percentage is determined on a weight/weight, mol/mol, or chromatography area percent basis relative to all fatty acids present in the composition as determined by methods such as disclosed in the European Pharmacopeia monograph for omega-3 fatty acid concentrates, European Pharmacopeia monograph for omega-3-acid ethyl esters 90%, or European Pharmacopeia monograph method 2.4.29, USP monograph for fish oil dietary supplements, USP 35 omega-3-acid ethyl esters (LOVAZA®) monograph, or any essentially equivalent methods (whether by gas chromatography, HPLC, FPLC or any other chromatographic method).
  • the fatty acid percentage is determined not as a percentage of all fatty acids present in the composition but as a specific type of fatty acid ethyl esters as percentage of all fatty acid ethyl esters present in the composition, thus excluding from the fatty acid percentage determination such fatty acids present as, for instance: free fatty acids; mono-, di-, and tri-glycerides; or fatty acids present in phospholipids (such as phosphatidylserine or phosphatidylcholine) or polysorbates (such as Tween 80, Tween 20, or polysorbate 40).
  • phospholipids such as phosphatidylserine or phosphatidylcholine
  • polysorbates such as Tween 80, Tween 20, or polysorbate 40.
  • the fatty acid percentage is determined not as a percentage of all fatty acids present in the composition but as a specific type of free fatty acid as percentage of all free fatty acids present in the composition, thus excluding from the fatty acid percentage determination such fatty acids present as, for instance: fatty acid ethyl esters; mono-, di-, and tri-glycerides; or fatty acids present in phospholipids (such as phosphatidylserine or phosphatidylcholine) or polysorbates (such as Tween 80, Tween 20, or polysorbate 40).
  • fatty acids present for instance: fatty acid ethyl esters; mono-, di-, and tri-glycerides; or fatty acids present in phospholipids (such as phosphatidylserine or phosphatidylcholine) or polysorbates (such as Tween 80, Tween 20, or polysorbate 40).
  • the fatty acid percentage is determined not as a percentage of all fatty acids present in the composition but as a specific type of glycerol fatty acid ester as percentage of all glycerol fatty acid esters present in the composition, thus excluding from the fatty acid percentage determination such fatty acids present as, for instance: fatty acid ethyl esters; free fatty acids; or fatty acids present in phospholipids (such as phosphatidylserine or phosphatidylcholine) or polysorbates (such as Tween 80, Tween 20, or polysorbate 40).
  • fatty acids present for instance: fatty acid ethyl esters; free fatty acids; or fatty acids present in phospholipids (such as phosphatidylserine or phosphatidylcholine) or polysorbates (such as Tween 80, Tween 20, or polysorbate 40).
  • the fatty acid percentage is determined not as a percentage of all fatty acids present in the composition but as di- or tri-fatty acid esters with glycerol as percentage of all glycerol di- and tri-fatty acid esters present in the composition, thus excluding from the fatty acid percentage determination such fatty acids present as, for instance: glycerol-mono-fatty acid esters; fatty acid ethyl esters; free fatty acids; or fatty acids present in phospholipids (such as phosphatidylserine or phosphatidylcholine) or polysorbates (such as Tween 80, Tween 20, or polysorbate 40).
  • phospholipids such as phosphatidylserine or phosphatidylcholine
  • polysorbates such as Tween 80, Tween 20, or polysorbate 40.
  • the fatty acid percentage is determined not as a percentage of all fatty acids present in the composition but as a tri-fatty acid esters with glycerol as percentage of all glycerol tri-fatty acid esters present in the composition, thus excluding from the fatty acid percentage determination such fatty acids present as, for instance: mono- and di-fatty acid esters of glycerol; fatty acid ethyl esters; free fatty acids; or fatty acids present in phospholipids (such as phosphatidylserine or phosphatidylcholine) or polysorbates (such as Tween 80, Tween 20, or polysorbate 40).
  • phospholipids such as phosphatidylserine or phosphatidylcholine
  • polysorbates such as Tween 80, Tween 20, or polysorbate 40.
  • the EPA, HPA, DPA, or omega-3-pentaenoic acids may be derived from any appropriate source including plant seed oils, microbial oils from algae or fungal or marine oils from fish or other marine animals. Certain species are a particular good source of oils containing DPA, for example seal oil. They may be used in the form of the natural oil, if that oil meets the required purity requirements of the present invention, or may be purified to give products containing the fatty acid composition of the present invention.
  • compositions of the present invention may be produced through a range of the methods.
  • Such methods may include: distillation, including short path distillation; urea precipitation; enzymatic conversion concentration; conventional chromatography; HPLC/FPLC; supercritical carbondioxide extraction; supercritical carbondioxide chromatography; simulated moving bed chromatography; supercritical carbondioxide simulated moving bed chromatography; or chemical conversion methods such as iodolactonization.
  • distillation including short path distillation; urea precipitation; enzymatic conversion concentration; conventional chromatography; HPLC/FPLC; supercritical carbondioxide extraction; supercritical carbondioxide chromatography; simulated moving bed chromatography; supercritical carbondioxide simulated moving bed chromatography; or chemical conversion methods such as iodolactonization.
  • Such methods are generally known to those skilled in the art of purifying and isolating omega-3 fatty acids.
  • the omega-3 fatty acid concentration/purification process is initiated by esterifying the fatty acids comprised by the marine oil raw material (such as crude fish oil) with ethanol (to form fatty acid ethyl esters) in order to separate omega-3 fatty acids from other fatty acids covalently bound together in the natural triglyceride molecules of the source oil. Subsequently, the material may be distilled once or several times to achieve omega-3-acid ethyl ester concentrations above 60%-70%. Alternatively, enzymatic concentration, urea precipitation or supercritical extraction may be used alone or in conjunction with distillation to reach omega-3 levels above 70%-90%.
  • chromatography In order to prepare a highly pure concentrate of a single omega-3 fatty acid, methods such as chromatography, supercritical chromatography, simulated moving bed chromatography, supercritical simulated moving bed chromatography, or chemical conversion methods such as iodolactolization are typically most practical to reach levels above 50%, alternatively above 60%, alternatively above 70%, alternatively above 80%, alternatively above 90%, alternatively above 95%, of a single omega-3 fatty acid such as ETA, EPA, HPA, DPA, TPA, or DHA.
  • a single omega-3 fatty acid such as ETA, EPA, HPA, DPA, TPA, or DHA.
  • DPA and HPA are present at much lower concentrations.
  • DPA or HPA may be concentrated and purified from fish or other marine oils according to the methods referred to above, either alone or DPA combined with EPA and/or HPA.
  • the DPA or HPA may be chemically prepared from a high purity EPA concentrate by elongation of the EPA fatty-acid chain with two or one hydrogen-saturated carbons (C2-elongation or C1-elongation) on the carboxyl side of the molecule (for instance with a method similar to or alternate methods with equivalent results such as described by Kuklev DV and Smith WL in Chem Phys Lipids, 2006; 144(2): 172-177).
  • a high purity EPA concentrate may be partially converted to DPA (or HPA) using a method for C2-elongation (or C1-elongation) of EPA similar to those described above, thus directly yielding compositions of the present invention or intermediates therefore.
  • oils containing one or more of the desired fatty acids may be blended to give the desirable relative amounts of EPA, DPA, HPA, DHA, TPA, other omega-3 fatty acids and omega-6 fatty acids to obtain the compositions of the present invention described in detail above.
  • Fish oils may also contain by-products and contaminants such as pesticides, chlorinated or brominated hydrocarbons, heavy metals, cholesterol and vitamins. During the production of the concentrate, the concentrations of these components are significantly reduced compared to untreated fish oils. Such reduction is inherent due to the nature of purification methods and their ability to concentrate of several or specific omega-3 fatty acids, thus removing other compounds.
  • Triglycerides comprising more than 60% of the omega-3 fatty acids in the composition may be produced from ethyl esters and glycerol by well known, published, or alternative chemical synthetic or enzymatic procedures.
  • the free acids may be produced from ethyl esters by well known hydrolization or saponification procedures. Methods for converting ethyl esters to triglycerides, free fatty acids, and other molecular forms comprising fatty acids, are generally known to those skilled in the art chemically or enzymatically converting omega-3 fatty acids from one form to another.
  • compositions of the present invention have improved pharmacological features as demonstrated by improved bioavailability in a mammal of EPA, HPA, DPA, DHA, EPA+DHA, EPA+DPA or EPA+HPA+DPA combined, total omega-3-pentaenoic acids, or of total omega-3 fatty acids.
  • Key parameters for determining bioavailability are maximum concentration of a therapeutic compound or a metabolite thereof (Cmax); the time from administration to maximum concentration (Tmax); and the area under the concentration curve over time (AUC). Such parameters may be determined under single dose or multiple dose administration regimens. Methods to determine comparative bioavailability in mammals are generally known to those skilled in the art.
  • Meal conditions during administration to a subject of omega-3 fatty acid compositions or omega-3 fatty acid formulations can be of special significance for absorption and bioavailability of omega-3 fatty acids.
  • the meal conditions typically considered are: fasting (no food at all prior for 8-12 hours prior to administration and 2-3 hours post administration of the treatment); a low fat meat (a meal typically containing less than 25 gram of fat [350-600 Kcal] consumed just before or after the administration of the treatment; typically within a 15-30 minute range); or a high fat meat (a meal containing 40 gram to 75 gram of fat [700-1000 Kcal] consumed just before or after the administration of the treatment; typically within a 15-30 minute range).
  • compositions of the present invention are more rapidly absorbed as measured by the time to reach the maximum concentration (Tmax) in blood, serum or plasma of EPA, DPA, DHA, EPA+DPA, EPA+DHA, total omega-3-pentaenoic acids, or total omega-3 fatty acids.
  • Tmax under high fat meal administration conditions is less than 8 hours, alternatively less than 6 hours, alternatively approximately 5 hours, alternatively 4 hours or less.
  • Tmax under low fat meal administration conditions is less than 8 hours, alternatively less than 6 hours, alternatively approximately 5 hours, alternatively 4 hours or less.
  • Tmax under fasting administration conditions is less than 8 hours, alternatively less than 6 hours, alternatively approximately 5 hours, alternatively 4 hours or less.
  • Tmax for EPA, DPA, DHA, EPA+DPA, EPA+DHA, or total omega-3 fatty acids are equal or less than Tmax for AMR101 for EPA, DPA, DHA EPA+DPA, EPA+DHA, total omega-3-pentaenoic acids, or total omega-3 fatty acids under high fat meat, low fat meal, and fasting administration conditions.
  • Tmax for EPA, DPA, DHA, EPA+DPA, EPA+DHA, or total omega-3 fatty acids are less than Tmax for AMR101 for EPA+DHA and to EPA, DPA, DHA, EPA+DPA, EPA+DHA, or total omega-3 fatty acids under either low fat meal, fasting, or both administration conditions.
  • the improved bioavailability features described above are apparent upon single dose administration, while in other embodiments the improved bioavailability features described above are apparent after multiple dose administration of formulations according to the present invention as compared to referenced comparator products above or substantial equivalent forms thereof.
  • compositions of the present invention are more potent and effective than other omega-3 compositions known in the prior art (such as LOVAZA®, EPANOVATM or VASCEPA®).
  • the formulation may be a single daily dose preparation to give in one dose the above intakes, or may be in convenient divided doses, for example, a daily dose formed of two to four soft gelatin or other dosage forms, each containing 300-1500 mg of EPA, EPA+DPA, EPA+DPA+HPA, or omega-3-pentaenoic acids in any form embodied in the present invention.
  • Flavourants or emulsifiers may be included, for instance, to make the preparation palatable.
  • Other conventional additives, diluents and excipients may be present.
  • the preparation for ingestion may be in the form of a capsule, a dry powder, a tablet, a solution, an oil, an emulsion or any other appropriate form.
  • the capsules may be hard or soft gelatin capsules, agar capsules, or any other appropriate capsule.
  • the omega-3 fatty acid composition optionally includes chemical antioxidants, such as alpha tocopherol, which are administered in pure form or suspended in a vegetable oil, such as soybean oil or corn oil.
  • chemical antioxidants such as alpha tocopherol
  • the blended fatty acid compositions may then be incorporated into any appropriate dosage form for oral, enteral, parenteral, rectal, vaginal, dermal or other route of administration.
  • Soft or hard gelatin capsules, flavoured oil blends, emulsifiers or other liquid forms, and microencapsulate powders or other dry form vehicles are all appropriate ways of administering the products.
  • the formulated final drug product containing the omega-3 fatty acid composition may be administered to a mammal or patient in need thereof in a capsule, a tablet, a powder that can be dispersed in a beverage, or another solid oral dosage form, a liquid, a soft gel capsule or other convenient dosage form such as oral liquid in a capsule, as known in the art.
  • the capsule comprises a hard gelatin.
  • the combination product may also be contained in a liquid suitable for injection or infusion.
  • Example pharmaceutical grade finished dosage forms (a) Soft or hard gelatin capsules each containing 500 mg or 1000 mg of a mix 20 parts of EPA as a free fatty acid to 1 parts of DPA as a free fatty acid; (b) As in (a) but where the EPA and DPA free fatty acids are replaced with the fatty acids in any other appropriate bioassimilable form such as the ethyl esters; (c) As in (a)-(b) but where the material is in the form of a microencapsulated powder which can be used as a powder or compressed into tablets.
  • Such powders may be prepared by a variety of technologies known to those skilled in the art; (d) As in (a)-(b) but where the formulation is a liquid or emulsion, appropriately flavoured for palatable oral administration; (e) As in (a)-(b) but where the material is formulated into a pharmaceutically acceptable vehicle appropriate for topical application such as a cream or ointment.
  • the omega-3 compositions of the present invention may also be administered with a combination of one or more non-active pharmaceutical ingredients (also known generally herein as “excipients”).
  • Non-active ingredients serve to solubilize, suspend, thicken, dilute, emulsify, stabilize, preserve, protect, color, flavor, and fashion the active ingredients into an applicable and efficacious preparation that is safe, convenient, and otherwise acceptable for use.
  • the non-active ingredients may include colloidal silicon dioxide, crospovidone, lactose monohydrate, lecithin, microcrystalline cellulose, polyvinyl alcohol, povidone, sodium lauryl sulfate, sodium stearyl fumarate, talc, titanium dioxide and xanthum gum.
  • pharmaceutically acceptable vehicle includes any of the following: a solution where the first API and optional other ingredients are wholly dissolved in a solubilizer (e.g., a pharmaceutically acceptable solvent or mixture of solvents), wherein the solution remains in clear liquid form at about room temperature; a suspension; an oil; or a semi-solid, wherein the first API and optionally other ingredients are dissolved wholly or partially in a solubilizer (an emulsion, cream, etc.).
  • a solubilizer e.g., a pharmaceutically acceptable solvent or mixture of solvents
  • a “pharmaceutical grade finished dosage form” as used herein may be construed as a unit dose form suitable for administration to, for example, human or animal subjects, and having content uniformity acceptable to regulatory authorities.
  • a pharmaceutical grade finished dosage form should have an amount of API within the range of 85% to 115% of the desired dosage and an RSD less than or equal to 6.0%.
  • a pharmaceutical grade finished dosage form must be stable (i.e., have a “shelf life”) for a pharmaceutically acceptable duration of time, preferably at least six months, alternatively at least one year, or at least two years, when stored at room temperature (about 23 degree Celcius to 27 degree Celcius, preferably about 25 degree Celcius) and 60% relative humidity.
  • stability is determined by physical appearance and/or chemical modification of the ingredients, in accordance with standards well-known in the pharmaceutical arts, including those documented in ICH guidelines.
  • the omega-3 fatty acid dosage form optionally includes chemical antioxidants, such as alpha tocopherol, oils, such as soybean oil and partially hydrogenated vegetable oil, and lubricants such as fractionated coconut oil, lecithin and a mixture of the same.
  • chemical antioxidants such as alpha tocopherol, oils, such as soybean oil and partially hydrogenated vegetable oil
  • lubricants such as fractionated coconut oil, lecithin and a mixture of the same.
  • compositions of the present invention may be used for the treatment of patients by administering an effective amount of such compositions to a subject in need thereof, such as a subject prone to or afflicted with a disease or condition or in need of treatment for a disease or condition.
  • the present invention provides methods of treating, preventing, and reducing the symptoms, pathology or events associated with a disease or condition comprising administration of any of the compositions of the present invention.
  • the present invention provides methods of treating, preventing, reducing the occurrence of, and reducing symptoms associated with coronary heart disease (CHD), vascular disease, atherosclerotic disease, or related conditions.
  • CHD coronary heart disease
  • vascular disease vascular disease
  • atherosclerotic disease or related conditions.
  • the present invention also provides methods of treating, preventing, reducing the occurrence of, and reducing the symptoms, pathology or events associated with vascular-related diseases or conditions.
  • vascular diseases and conditions include, but are not limited to: atherosclerosis, atherosclerosis associated with hypercholesterolemia, hypertriglyceridemia or mixed dyslipidemia, coronary heart disease (CHD), vascular disease, peripheral artery disease (PAD), heart failure, cardiac arrhythmias, blood coagulatory conditions associated with cardiac arrhythmias, hypertension, coagulation related disorders (including but not limited to post-surgical deep vein thrombosis or other high risk thrombosis conditions), nephropathy and other kidney diseases, renal impairment, retinopathy, cognitive impairment, dementia (including but not limited to ischemic dementia and vascular dementia), and metabolic syndrome.
  • compositions of the present invention may be used for the treatment of patients by administering an effective amount of such compositions to a subject or a subject in need thereof, such as a subject prone to or afflicted with a vascular/cardiovascular disease or condition, a subject at risk of vascular/cardiovascular events, or a subject in need of treatment for a vascular/cardiovascular disease or condition.
  • the present invention provides for the prevention of death (including all cause mortality, cardiovascular mortality, and cardiac death); the treatment and/or prevention of cardiac events, cardiovascular events and/or vascular events and/or symptoms. In some embodiments, the present invention also provides for the reduction of number or frequency of such events, as well as a reduction or amelioration of symptoms associated with such events.
  • Vascular, cardiovascular and/or cardiac events may include, but are not limited to: death due to cardiovascular event (including cardiovascular death and cardiac death), sudden cardiac death, myocardial infarction (fatal or non-fatal), ischemic cardiac attack, ischemic attack, angina, stroke, transient ischemic cerebral attack, thrombosis, deep vein thrombosis, pulmonary embolism, coronary revascularization, coronary revascularization with stent placement, carotid artery revascularization, carotid artery revascularization with stent placement, peripheral artery revascularization, peripheral artery revascularization with stent placement, major coronary event, coronary artery bypass grafting, other blood vessel grafting, treatment of aneurism, plaque rupture, and hospitalization due to cardiovascular event (including angina, acute angina and unstable angina).
  • cardiovascular event including angina, acute angina and unstable angina.
  • Cardiovascular and/or cardiac events may also include other events deemed to fall in such category by those skilled in the art.
  • Study endpoints may comprise individual events or a composite of several of the events described directly above (for the latter the term Major Adverse Cardiovascular Event or MACE is often used).
  • the present invention provides methods of retarding the atherosclerotic disease process or improves the vascular healing process in response to presence of atherogenic disease.
  • retardation or healing may be demonstrated by reduced stenosis and/or restenosis at specific vascular sites over time, reduced or lesser increase in intima-media thickness (IMT) of the arterial wall over time, larger lumen size and/or larger vascular diameter at vascular sites with stenosis or clot built-up over time.
  • IMT intima-media thickness
  • Such retardation or healing may be determined by intravascular ultrasound (IVUS), radiographic, echocardiographic, radiologic, non-invasive ultrasound, tomography, magnetic resonance interference (MRI), or other acceptable methods.
  • IVUS intravascular ultrasound
  • MRI magnetic resonance interference
  • such retardation or improved healing may be demonstrated by the vascular wall composition, such as a reduced foam cell presence or fibrillated tissue in the vessel wall.
  • such improved vascular healing is demonstrated by improved inflammatory markers in the vascular wall or specific types of staining of vascular tissue.
  • compositions of the present invention may differentially alter the ratio between blood platelets and fragments thereof (also known as platelet microparticles). Such fragments may be evaluated as a whole or examined and described as fragment sub-categories.
  • compositions of the present invention may differentially alter the surface charge of blood platelets and fragments thereof, either in resting state (non-activated platelets) or activated stage.
  • compositions of the present invention may affect the coagulatory cascade and differentially alter coagulation or bleeding times or platelet aggregation times and density.
  • compositions of the present invention may be useful for any subjects and they may be especially useful in certain patient populations, including but not limited to the following: subjects having receiving treatment for or in need of treatment for hypertriglyceridemia, hypercholesterolemia, Frederickson dyslipidemia, and other comorbidities.
  • the present invention provides for methods of treatment of subjects with no previous cardiovascular events, or subjects with one or more previous cardiovascular events.
  • the subjects to be treated with or administered the compositions of the present invention are selected based on their dyslipidemic profile.
  • Subjects may be selected for treatment/administration from the following categories: hypertriglyceridemia (such as TG ⁇ 750 mg/dL, TG ⁇ 500 mg/dL, TG ⁇ 200 mg/dL, TG ⁇ 150 mg/dL, TG500-2000 mg/dL, TG500-1500 mg/dL, TG750-2000 mg/dL, TG750-1500 mg/dL, TG200-499 mg/dL, TG300-499 mg/dL, TG350-499 mg/dL, TG300-750 mg/dL, TG300-700 mg/dL, TG200-499 mg/dL, or TG150-199 mg/dL); hypercholesterolemia (such as LDL-C ⁇ 70 mg/dL, LDL-C ⁇ 70 mg/dL, LDL-C ⁇ 100 mg/dL, LDL-C ⁇ 130/dL, LDL-C ⁇ 160 mg/dL, LDL-C190 mg/dL, LDL-C 70-100 mg/dL
  • the subjects to be treated with or administered the compositions of the present invention are selected based on the presence of certain comorbidities.
  • comorbidities may include, but are not limited to the following: renal disease, nephropathy, IgA nephropathy, renal impairment, renal failure (also kidney failure or renal insufficiency), renal insufficiency requiring dialysis, renal insufficiency without dialysis treatment, chronic analgesic nephritis, polycystic kidney disease, proteinuria, hypertension, thrombotic microangiopathy, renal failure (acute renal failure, chronic renal failure), uremic pericarditis, uremia, renal artery stenosis, renal ischemia, hypertensive nephropathy, renovascular hypertension, renal osteodystrophy, nephroptosis, renal cortical necrosis, glomerulitis, metabolic syndrome, diabetes, or pre-diabetes.
  • the methods of administering the compositions of the present invention are useful in subjects having cardiopathy, coronary ischemia, cardiac decompensation, or diabetic pathology with cardiopathy, and subjects with previous myocardial infarction, stroke, recent myocardial infarction (post-MI), Acute Coronary Syndrome (ACS), ACS without ST-segment elevation (NSTEMI), ACS with ST-segment elevation (STEMI), or any other major cardiovascular event.
  • the compositions of the present invention may reduce the reoccurrence of such cardiovascular events.
  • treatment with the compositions of the present invention results in clinical improvement of such comorbidities.
  • administration with the compositions of the present invention may reduce the time necessary to achieve clinical improvement and/or attain treatment goals.
  • the administration of compositions can result in clinical improvements in clinical markers, including, but not limited to: glomerular filtration rate, serum creatinine, serum urea, serum phosphate levels, serum potassium levels, systolic or diastolic blood pressure, progression to dialysis, progression to renal failure, progression to need for renal transplant, fasting serum glucose levels, postprandial serum glucose levels, serum fructosamine levels, therapeutic insulin utilization, progression to diabetes, hypo-glucose events, hyper-glucose events, or hemoglobin A1C.
  • compositions of the present invention may be co-administered with one or more other therapeutic agents.
  • clinical benefits resulting from the administration or treatment of subjects with the compositions of the present invention may be improved with concomitant use or in combination with other therapeutic agents.
  • concomitant or fixed combination treatments may include coadministration with one or more of the following: an HMG-CoA reductase inhibitor (“statin”), an anti-platelet agent (such as aspirin or clopidogrel), an anti-coagulant (such as warfarin) an antihypertensive (such as a diuretic, beta-blocker, calcium channel blocker, ACE-inhibitor, angiotensin II receptor (ARB) antagonist), or other treatments for cardiovascular diseases.
  • statin HMG-CoA reductase inhibitor
  • an anti-platelet agent such as aspirin or clopidogrel
  • an anti-coagulant such as warfarin
  • an antihypertensive such as a diuretic, beta-blocker
  • the present invention also provides pharmaceutical compositions, for example, a unit dosage, comprising one or more HMG-CoA reductase inhibitors (“statins”) and an omega-3 fatty acid composition of the present invention.
  • statins HMG-CoA reductase inhibitors
  • the present invention provides a fixed dose combination product comprising both a statin and the composition of the present invention.
  • the present invention may incorporate currently known or future known statins in an amount generally recognized as safe and effective. There are currently seven statins that are widely available: atorvastatin, rosuvastatin, fluvastatin, lovastatin, pravastatin, pitavastatin, and simvastatin. An eighth statin, cerivastatin, has been removed from the U.S. market at the time of this writing.
  • cerivastatin may be used in conjunction with some embodiments of the present invention if cerivastatin is ultimately determined to be safe and effective in certain treatment regimens.
  • Such statins are typically used at their common daily doses, which include, but are not limited to lovastatin 10 mg, 20 mg, 40 mg; pravastatin 10 mg, 20 mg, 40 mg, 80 mg; simvastatin 5 mg, 10 mg, 20 mg, 40 mg, 80 mg, fluvastatin 20 mg, 40 mg, 80 mg; atorvastatin 10 mg, 20 mg, 40 mg, 80 mg; rosuvastatin 5 mg, 10 mg, 20 mg, 40 mg; and pitavastatin 1 mg, 2 mg, 4 mg, 8 mg.
  • statins are dose dependent, i.e., the higher the dose, the greater the therapeutic affect.
  • the effect of each statin is different, and therefore the level of therapeutic effect of one statin cannot be necessarily be directly correlated to the level of therapeutic effects of other statins.
  • bioavailability varies widely among the statins. Specifically, it has been shown that simvastatin is less than 5% bioavailable, while fluvastatin is approximately 24% bioavailable. Statins are absorbed at rates ranging from about 30% with lovastatin to 98% with fluvastatin. First-pass metabolism occurs in all statins except pravastatin.
  • Pravastatin is also the least protein-bound of the statins (about 50%), compared with the others, which are more than 90% protein-bound. Accordingly, the statins possess distinct properties from one another.
  • the combination products of this invention involving each statin or a plurality of statins are also distinct.
  • the method of treatment may combine the administration of one or more statins at its common dose or an alternative dose with the composition of the present invention
  • a composition according to the present invention is prepared by mixing and homogenizing in a ratio of 98:2 the intermediates MEGAPEX E90D00EE (90% EPA ethyl ester,) and MAXOMEGA DPA95 FFA ( ⁇ 95% DPA synthetic fatty acid produced from EPA ethyl ester concentrate) converted to ethyl ester, respectively.
  • MEGAPEX E90D00EE 90% EPA ethyl ester,
  • MAXOMEGA DPA95 FFA ⁇ 95% DPA synthetic fatty acid produced from EPA ethyl ester concentrate
  • the resulting novel composition comprises 89.10% EPA, 1.95% DPA, 0.19% HPA, 91.24% omega-3-pentaenoic acids, less than 0.01% DHA, 91.24% omega-3-pentaenoic acids, 93.09% total omega-3 fatty acids, 3.15% ARA and 3.57% omega-6 fatty acids (all Area %).
  • a composition according to the present prevention is prepared by mixing and homogenizing in a ratio of 96:4 the intermediates MEGAPEX E90D00EE (90% EPA ethyl ester,) and MAXOMEGA DPA95 FFA (95% DPA synthetic fatty acid produced from EPA ethyl ester concentrate), converted to ethyl ester, respectively.
  • MEGAPEX E90D00EE 90% EPA ethyl ester
  • MAXOMEGA DPA95 FFA 95% DPA synthetic fatty acid produced from EPA ethyl ester concentrate
  • MAXOMEGA DPA95 FFA
  • the resulting novel composition comprises 87.28% EPA, 3.89% DPA, 0.18% HPA, 91.35% omega-3-pentaenoic acids, less than 0.01% DHA, 93.17% total omega-3 fatty acids and 3.49% omega-6 fatty acids (all Area %).
  • a composition according to the present invention is prepared by mixing and homogenizing in a ratio of 94:6 the intermediates MEGAPEX E90D00EE (90% EPA ethyl ester,) and MAXOMEGA DPA95 FFA ( ⁇ 95% DPA synthetic fatty acid produced from EPA ethyl ester concentrate) converted to ethyl ester, respectively.
  • MEGAPEX E90D00EE 90% EPA ethyl ester,
  • MAXOMEGA DPA95 FFA ⁇ 95% DPA synthetic fatty acid produced from EPA ethyl ester concentrate
  • the resulting novel composition comprises 85.46% EPA, 5.84% DPA, 0.18% HPA, 91.48% omega-3-pentaenoic acids, less than 0.01% DHA, 93.26% total omega-3 fatty acids, 3.02% ARA, and 3.42% omega-6 fatty acids (all Area %).
  • a composition according to the present invention is prepared by mixing and homogenizing in a ratio of 75:25 the intermediates MEGAPEX E90D00EE (90% EPA ethyl ester,) and MAXOMEGA DPA95 FFA ( ⁇ 95% DPA synthetic fatty acid produced from EPA ethyl ester concentrate, converted to ethyl ester, respectively.
  • MEGAPEX E90D00EE 90% EPA ethyl ester,
  • MAXOMEGA DPA95 FFA ⁇ 95% DPA synthetic fatty acid produced from EPA ethyl ester concentrate, converted to ethyl ester, respectively.
  • These intermediates were prepared and commercially offered for sale by Chemport Korea (MEGAPEX) and Equateq Ltd from Scotland, UK (MAXOMEGA).
  • the relative amounts of fatty acids present in the starting intermediates and in the resulting novel composition is listed in table 4 below.
  • the resulting novel composition comprises 68.10% EPA, 24.32% DPA, 0.19% HPA, 92.65% omega-3-pentaenoic acids, less than 0.01% DHA, 94.07% total omega-3 fatty acids, 2.41% ARA and 2.73% omega-6 fatty acids (all Area %).
  • a composition according to the present invention is prepared by mixing and homogenizing in a ratio of 60:40 the intermediates KD-PharmaKD-PUR 900EE and MAXOMEGA DPA95 FFA converted to ethyl ester, respectively. These intermediates were prepared and commercially offered for sale by KD-Pharma Germany (KD-Pharma) and Equateq Ltd from Scotland, UK (MAXOMEGA). The relative amounts of fatty acids present in the starting intermediates and in the resulting novel composition is listed in table 5 below. The resulting novel composition comprises 55.74% EPA, 39.26% DPA, 2.39% HPA, 97.44% omega-3-pentaenoic acids, and 98.06% total omega-3 fatty acids (all Area %).
  • a composition according to the present invention is prepared by mixing and homogenizing in a ratio of 96:4 the intermediates KD-PUR 900EE KD-Pharma and MAXOMEGA DPA95 FFA converted to ethyl ester, respectively. These intermediates were prepared and commercially offered for sale by KD-Pharma Germany (KD-Pharma) and Equateq Ltd from Scotland, UK (MAXOMEGA). The relative amounts of fatty acids present in the starting intermediates and in the resulting novel composition is listed in table 6 below.
  • the resulting novel composition comprises 89.27% EPA, 4.45% DPA, 3.82% HPA, 97.54% omega-3-pentaenoic acids, and 98.54% total omega-3 fatty acids (all Area %).
  • a composition according to the present invention is prepared by mixing and homogenizing in a ratio of 91.8:8.2 the intermediates KD-PUR 910EE KD-Pharma and DPA95 FFA converted to ethyl ester, respectively.
  • the relative amounts of fatty acids present in the starting intermediates and in the resulting novel composition is listed in table 7 below.
  • the ethyl ester composition of Example 4 may be converted into a free fatty acid composition with essentially the same fatty acid composition according to “Conversion Method EE to FFA” below. This method is indiscriminate with respect to the type, degree of saturation or length of fatty acid if performed for an adequate amount of time under the described conditions.
  • the ethyl ester composition of Example 3 is converted into a free fatty acid composition with essentially the same fatty acid composition according to “Conversion Method EE to FFA” above. This method is indiscriminate with respect to the type, degree of saturation or length of fatty acid if performed for an adequate amount of time under the described conditions.
  • the ethyl ester composition of Example 6 is converted into a free fatty acid composition with essentially the same fatty acid composition according to “Conversion Method EE to FFA” above. This method is indiscriminate with respect to the type, degree of saturation or length of fatty acid if performed for an adequate amount of time under the described conditions.
  • Example 4 The composition of Example 4 is formulated into a soft gelatin capsule. Prior to encapsulation, an anti-oxidant preparation (composed of 4000 mg alpha-D-tocopherol in one liter of corn oil; corn oil is a triglyceride low in omega-3) is added to the composition of Example 4, by mixing and homogenizing 100 mL of this anti-oxidant preparation into 100 liters of the oil composition of Example 4 followed by thorough homogenization. The resulting pre-encapsulation formulated oil contains approximately 4 mg/gram alpha-D-tocopherol.
  • an anti-oxidant preparation composed of 4000 mg alpha-D-tocopherol in one liter of corn oil; corn oil is a triglyceride low in omega-3
  • the resulting pre-encapsulation formulated oil contains approximately 4 mg/gram alpha-D-tocopherol.
  • the formulated oil is encapsulated into soft gelatin capsules with printed logo according to general methods typically used by Accucaps in Canada for fish oils or by any other documented and operational encapsulation method.
  • the fill mass of the oil is approximately 1.08 gram/capsule, providing a dose of approximately 1000 mg omega-3-pentaenoic-acids ethyl esters per capsule.
  • the capsules are bottled in HDPE bottles with induction seal and child resistant cap.
  • Example 9 The composition of Example 9 is formulated into a soft gelatin capsule. Prior to encapsulation, an anti-oxidant preparation (composed of 4000 mg alpha-D-tocopherol in one liter of corn oil; corn oil is a triglyceride low in omega-3) is added to the composition of Example 4, by mixing and homogenizing 100 mL of this anti-oxidant preparation into 100 liters of the oil composition of Example 4 followed by thorough homogenization. The resulting pre-encapsulation formulated oil contains approximately 4 mg/gram alpha-D-tocopherol.
  • an anti-oxidant preparation composed of 4000 mg alpha-D-tocopherol in one liter of corn oil; corn oil is a triglyceride low in omega-3
  • the resulting pre-encapsulation formulated oil contains approximately 4 mg/gram alpha-D-tocopherol.
  • the formulated oil is encapsulated into soft gelatin capsules with printed logo according to general methods typically used by Banner in High Point, N.C., for fish oils or by any other documented and operational encapsulation method.
  • the fill mass of the oil is approximately 1.09 gram/capsule, providing a dose of approximately 1000 mg omega-3-pentaenoic-acids per capsule.
  • the capsules are bottled in HDPE bottles with induction seal and child resistant cap.
  • Example 5 The composition of Example 5 is formulated into a soft gelatin capsule.
  • an anti-oxidant preparation (composed of 4000 mg alpha-D-tocopherol in one liter of corn oil; corn oil is a triglyceride low in omega-3) is added to the composition of Example 4, by mixing and homogenizing 100 mL of this anti-oxidant preparation into 100 liters of the oil composition of Example 4 followed by thorough homogenization.
  • the resulting pre-encapsulation formulated oil contains approximately 4 mg/gram alpha-D-tocopherol.
  • the formulated oil is encapsulated into soft gelatin capsules with printed logo according to general methods typically used by Catalent in St.
  • the fill mass of the oil is approximately 1.05 gram/capsule, providing a dose of approximately 1000 mg omega-3-pentaenoic-acids ethyl esters per capsule.
  • the capsules are bottled in HDPE bottles with induction seal and child resistant cap.
  • Example 10 The composition of Example 10 is formulated into a soft gelatin capsule. Prior to encapsulation, an anti-oxidant preparation (composed of 4000 mg alpha-D-tocopherol in one liter of corn oil; corn oil is a triglyceride low in omega-3) is added to the composition of Example 4, by mixing and homogenizing 100 mL of this anti-oxidant preparation into 100 liters of the oil composition of Example 4 followed by thorough homogenization. The resulting pre-encapsulation formulated oil contains approximately 4 mg/gram alpha-D-tocopherol.
  • an anti-oxidant preparation composed of 4000 mg alpha-D-tocopherol in one liter of corn oil; corn oil is a triglyceride low in omega-3
  • the resulting pre-encapsulation formulated oil contains approximately 4 mg/gram alpha-D-tocopherol.
  • the formulated oil is encapsulated into soft gelatin capsules with printed logo according to general methods typically used by Banner in High Point, N.C., for fish oils or by any other documented and operational encapsulation method.
  • the fill mass of the oil is 1.06 gram/capsule, providing a dose of approximately 1000 mg omega-3-pentaenoic-acids per capsule.
  • the capsules are bottled in HDPE bottles with induction seal and child resistant cap.
  • a patient is diagnosed with vascular disease. Thereupon, the patient may be initiated on daily treatment with one of the encapsulated compositions according to Examples 10, 11, 12 or 13. Four capsules per day are administered to this patient (4 g/d).
  • a patient is treated as per Example 15.
  • the treatment results in significant reduction of the occurrence of MACE.
  • COMPOSITION 1b Minimum Maximum Target Composition (mg/g) (mg/g) (mg/g) Omega-3 pentaenoic acid 870 990 920 Eicosapentaenoic acid (EPA) 750 950 830 Heneicosapentaenoic acid (HPA) 5 70 40 Docosapentaenoic acid (DPA) 50 130 90 Docosahexaenoic acid (DHA) 40 20
  • the EPA:HPA ratio is between 13 and 190
  • the EPA:DPA ratio is between 8 and 15, the HPA:DPA ration between 0.05 and 1, the DPA:DHA ratio more than 2.4, preferably more than 4, more preferably more than 6, most preferably more than 10, and the EPA:DHA ratio more than 32, preferably more than 38, more preferably more than 80, most preferably more than 95.
  • the EPA, HPA, DPA and DHA may be composed as a glyceride (such as a g
  • COMPOSITION 2 Minimum Maximum Target Composition (mg/g) (mg/g) (mg/g) Omega-3 pentaenoic acid 900 980 940 Eicosapentaenoic acid (EPA) 15 60 30 Heneicosapentaenoic acid (HPA) 5 60 30 Docosapentaenoic acid (DPA) 800 950 880 Docosahexaenoic acid (DHA) 25 ⁇ 10
  • the EPA:HPA ratio is between 0.25 and 12
  • the DPA:EPA ratio is between 13 and 63
  • the EPA, HPA, DPA and DHA may be composed as a glyceride (such
  • DPA n-3 Minimum Maximum Target Composition (mg/g) (mg/g) (mg/g) (mg/g)
  • the DPA may be composed as a glyceride (such as triglyceride), an ester (such as ethyl ester), or a free fatty acid.
  • COMPOSITION 4 Minimum Maximum Target Composition (mg/g) (mg/g) (mg/g) Omega-3 pentaenoic acid 930 1000 966 Eicosapentaenoic acid (EPA) 840 870 853 Heneicosapentaenoic acid (HPA) 20 40 30 Docosapentaenoic acid (DPA) 60 100 81 Docosahexaenoic acid (DHA) 5 20 12
  • a mixture of DPA and EPA was prepared by combining 1 g DPA Ethyl Ester (SE-133-III) with 10 g EPA Ethyl Ester, 914 mg/g (KD Pharma FM13001) in 150 ml of 95% ethanol/water containing 35 ml of 2M sodium hydroxide. This reaction mixture was stirred overnight at ambient temperature. Tlc analysis showed complete conversion of the ethyl esters to the corresponding acids. The reaction mixture was cooled in an ice bath, acidified with 6N hydrochloric acid and concentrated on a rotavap under reduced pressure. Water and ethyl acetate were added, the phases separated and the aqueous residue extracted with ethyl acetate.

Abstract

Orally administrable composition comprising fatty acids comprising omega-3-fatty acids, salts or derivatives thereof are provided. These compositions can be used for the treatment or prophylaxis of dyslipidemic, cardiovascular, CNS, inflammatory, and other diseases/conditions or risk factors therefore.

Description

    RELATED APPLICATIONS
  • This application is a continuation-in-part (CIP) application of PCT International Application No. PCT/US13/46176, filed on Jun. 17, 2013, which claims the benefit of U.S. Provisional Patent Application No. 61/660,757, filed Jun. 17, 2012, U.S. Provisional Patent Application No. 61/734,331, filed Dec. 6, 2012, and U.S. Provisional Patent Application No. 61/780,948, filed Mar. 13, 2013, the contents of which are incorporated herein by reference.
  • FIELD OF INVENTION
  • The present invention relates to omega-3 fatty acid compositions, and methods of treating, preventing, reducing the occurrence of, and improving symptoms associated with coronary heart disease (CHD), vascular disease, atherosclerotic disease or related conditions. The present invention also provides methods for the treatment and/or prevention and/or reduction of cardiac events and/or cardiovascular events and/or vascular events and/or symptoms.
  • BACKGROUND OF THE INVENTION
  • Marine oils, also commonly referred to as fish oils, are a good source of the two main omega-3 fatty acids, eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), which have been found to regulate lipid metabolism. Omega-3 fatty acids have been found to have beneficial effects on the risk factors for cardiovascular diseases, especially mild hypertension, hypertriglyceridemia and on the coagulation factor VII phospholipid complex activity. Omega-3 fatty acids lower serum triglycerides (TG), increase serum HDL-cholesterol, lower systolic and diastolic blood pressure and the pulse rate, and lower the activity of the blood coagulation factor VII-phospholipid complex. Further, omega-3 fatty acids seem to be well tolerated, without giving rise to any severe side effects.
  • The table directly below lists the most common omega-3 fatty acids, including their 3-letter abbreviation code. In this application, the use of any of the 3-letter abbreviations shall refer to the omega-3 fatty acid, unless otherwise indicated (e.g. DPA or DPA 22:5 (n-3) or DPA 22:5-n3 or DPA 22:5n3 or DPA-n3, which all refer to the omega-3 isomer of docosapentaenoic acid).
  • Common Name for Omega-3 Fatty Acid (+abbreviation) Codified Lipid Name Chemical Name
    Hexadecatrienoic acid (HTA) 16:3 (n-3) all-cis-7,10,13-hexadecatrienoic acid
    α-Linolenic acid (ALA) 18:3 (n-3) all-cis-9,12,15-octadecatrienoic acid
    Stearidonic acid (SDA) 18:4 (n-3) all-cis-6,9,12,15-octadecatetraenoic acid
    Eicosatrienoic acid (ETE) 20:3 (n-3) all-cis-11,14,17-eicosatrienoic acid
    Eicosatetraenoic acid (ETA) 20:4 (n-3) all-cis-8,11,14,17-eicosatetraenoic acid
    Eicosapentaenoic acid (EPA) 20:5 (n-3) all-cis-5,8,11,14,17-eicosapentaenoic acid
    Heneicosapentaenoic acid (HPA) 21:5 (n-3) all-cis-6,9,12,15,18-heneicosapentaenoic acid
    Docosapentaenoic acid (DPA) or Clupanodonic acid 22:5 (n-3) all-cis-7,10,13,16,19-docosapentaenoic acid
    Docosahexaenoic acid (DHA) 22:6 (n-3) all-cis-4,7,10,13,16,19-docosahexaenoic acid
    Tetracosapentaenoic acid (TPA) 24:5 (n-3) all-cis-9,12,15,18,21-tetracosapentaenoic acid
    Tetracosahexaenoic acid (THA) or Nisinic acid 24:6 (n-3) all-cis-6,9,12,15,18,21-tetracosahexaenoic acid
  • One form of omega-3 fatty acids is a concentrate of omega-3, long chain, polyunsaturated fatty acids from fish oil containing DHA ethyl esters, EPA ethyl esters as well as ethyl esters of other omega-3 fatty acids (described in USP35 for LOVAZA®) and is sold under the trademarks OMACOR® and LOVAZA®. Such a form of omega-3 fatty acid comprises at least 90% omega-3 fatty acids of which at least 80% EPA+DHA (in a ratio of 1.2:1) and is described, for example, in U.S. Pat. Nos. 5,502,077, 5,656,667 and 5,698,594. LOVAZA® (omega-3-acid ethyl esters) is indicated for the treatment of patients with hypertriglyceridemia with TG levels of 500 mg/dL or higher.
  • Another form of omega-3 fatty acid concentrate is sold under the trademark EPADEL® ® for the treatment of dyslipidemia. This product is described as 98% EPA ethyl ester in Lancet (Vol. 369; Mar. 31, 2007; 1090-1098) reporting on a large outcome study with EPADEL®. EPADEL® is known to contain less than 1% of any fatty acid other than EPA.
  • Similar to EPADEL®, another form of omega-3 fatty acid concentrate also consists almost entirely of EPA ethyl ester and is known under its developmental stage name AMR101 or its trade name VASCEPA®. This product is described in US patent application 2010/0278879 as comprising at least 95% EPA (typically referred to as 97% or at least 96% in company releases and references) and less than 1% of any other fatty acid. AMR101 was previously under development for the treatment of Huntingdon's Disease but failed in phase Ill clinical development. Subsequently, AMR101 was entered in a development program for hypertriglyceridemia and mixed dyslipidemia.
  • Yet another concentrate of omega-3, long chain, polyunsaturated fatty acids from fish oil containing approximately 75% DHA and EPA as free fatty acids is known under its developmental stage name EPANOVA™. This product is described as comprising approximately 55% EPA and 20% DHA. EPANOVA™ was previously under development for the treatment of Crohn's Disease but failed in phase Ill clinical development. Subsequently, EPANOVA™ was entered in a development program for hypertriglyceridemia and mixed dyslipidemia.
  • Generally, the bioavailability and therapeutic effect of omega-3 fatty acid compositions is dose dependent, i.e., the higher the dose, the greater the therapeutic affect and bioavailability. However, the effect of each specific omega-3 fatty acid composition may be different, and therefore the level of therapeutic effect of one composition at a given dose cannot necessarily be inferred from the level of therapeutic effects of other omega-3 fatty acid compositions at the same or similar dose.
  • Omega-3 fatty acids are known to be “essential fatty acids”. There are two series of essential fatty acids (EFAs) in humans. They are termed “essential” because they cannot be synthesized de novo in mammals. These fatty acids can be interconverted within a series, but the omega-6 (n-6) series cannot be converted to the omega-3 series nor can the omega-3 (n-3) series be converted to the omega-6 series in humans. The main EFAs in the diet are linoleic acid of the omega-6 series and alpha-linolenic acid of the omega-3 series. However, to fulfill most of their biological effects these “parent” EFAs must be metabolised to the other longer chain fatty acids. Each fatty acid probably has a specific role in the body. The scientific literature suggests that particularly important in the n-6 series are dihomo-gammalinolenic acid (DGLA, 20:3-n6) and arachidonic acid (ARA, 20:4-n6), while particularly important in the n-3 series are eicosapentaenoic acid (EPA, 20:5-n3) and docosahexaenoic acid (DHA, 22:6-n3).
  • U.S. Pat. No. 6,479,544 describes an invention in which it is found that ARA is highly desirable rather than undesirable and it may be helpful to administer ARA in association with EPA. This invention provides pharmaceutical formulations containing eicosapentaenoic acid or any appropriate derivative (hereinafter collectively referred to as EPA) and arachidonic acid (ARA), as set out in the granted claims for this patent. ARA may be replaced by one or more of its precursors, DGLA or GLA. In this reference, the ratio of EPA to ARA is preferably between 1:1 and 20:1.
  • Patent application PCT/GB 2004/000242 describes the treatment or prevention of psoriasis with a formulation comprising more than 95% EPA and less than 2% DHA. In another embodiment of this invention the EPA is replaced with DPA.
  • Patent application PCT/NL 2006/050291 (WO/2007/058538, GB 0301701.9) describes combinations of idigestible oligosaccharides and long chain poly-unsaturated fatty acids such as ARA, EPA, DA, and combinations thereof to improve intestinal barrier integrity, improving barrier function, stimulating gut maturation and/or reducing intestinal barrier permeability.
  • Lindeborg et al. (Prostag Leukotr Ess, 2013, 88:313-319) discloses a study evaluating postprandial metabolism of docosapentaenoic acid (DPA) and eicosapentaenoic acid (EPA) in humans.
  • Holub et al. (Lipids, 2011, 46:399-407) discloses a study assessing the effect of oral supplementation with docosapentaenoic acid (DPA) on levels of serum and tissue lipid classes and their fatty acid compositions in rat liver, heart, and kidney.
  • Cardiovascular disease includes numerous problems, many of which are related to a process called atherosclerosis. Atherosclerosis is a condition that develops when plaque builds up in the walls of the arteries. This buildup narrows the arteries, making it harder for blood to flow through. If a blood clot forms, it can stop the blood flow. This can cause lead to conditions or event such as myocardial infarction, stroke, heart failure, arrhythmias, valve dysfunction, and death.
  • All references cited herein are incorporated by reference in their entirety.
  • SUMMARY OF THE INVENTION
  • The present invention provides omega-3 fatty acid compositions and methods of administering these compositions.
  • The present invention provides a pharmaceutical composition comprising fatty acids, wherein at least 50% by weight of the fatty acids comprise omega-3 fatty acids, salts, esters, or derivatives thereof, wherein the omega-3 fatty acids comprise eicosapentaenoic acid (EPA) and docosapentaenoic acid (DPA) and wherein the ratio of docosahexaenoic acid to DHA to EPA (DHA:EPA) is less than 1:10, and wherein the ratio of DHA to DPA (DHA:DPA) is less than 2:1.
  • The present invention provides a pharmaceutical composition comprising eicosapentaenoic acid (EPA) in an amount between about 70% to about 95% of the total amount of fatty acids and docosapentaenoic acid (DPA), wherein the composition comprises no more than 5% docosahexaenoic acid (DHA) of the total amount of fatty acids, and wherein the ratio of DHA:DPA is 1:1 or lower.
  • The present invention provides a pharmaceutical composition comprising pharmaceutical composition comprising eicosapentaenoic acid (EPA) in an amount between about 750 mg/g to about 950 mg/g, and docosapentaenoic acid (DPA), wherein the composition comprises no more than 5% DHA of the total amount of fatty acids, and wherein the ratio of DHA:DPA is 1:1 or lower.
  • The present invention provides a pharmaceutical composition comprising eicosapentaenoic acid (EPA) in a daily dosage amount of between about 1000 mg to about 5000 mg, and further comprising docosapentaenoic acid (DPA) and docosahexaenoic acid (DHA), wherein the composition comprises no more than 5% DHA of the total amount of fatty acids, and wherein the ratio of DHA:DPA is 1:1 or lower.
  • The present invention provides a pharmaceutical composition comprising eicosapentaenoic acid (EPA) and docosapentaenoic acid (DPA), wherein the amount of EPA and DPA is about 55% or more by weight of the total amount of fatty acids, and wherein the ratio of DHA:DPA is no more than 1:1.
  • The present invention provides a pharmaceutical composition comprising: docosapentaenoic acid (DPA) in an amount between about 50% to about 80% of the total amount of fatty acids, docosahexaenoic acid (DHA) in an amount between about 25% to about 40% of the total amount of fatty acids, and optionally eicosapentaenoic acid (EPA) in an amount less than about 10% of the total amount of fatty acids.
  • In some embodiments, the compositions of the present invention comprise additional fatty acids, such as heneicosapentaenoic acid (HPA), arachidonic acid (ARA), and omega-6-docosapentaenoic acid (n-6 DPA), tetracosapentaenoic acid (TPA), and/or gamma-linoleic acid (GLA).
  • The present invention provides methods comprising administering the compositions. The present invention provides a method of treating, preventing, reducing the occurrence of, and improving symptoms associated with coronary heart disease (CHD), vascular disease, atherosclerotic disease or related conditions, comprising administering the compositions of the present invention to a subject in need thereof. The present invention also provides methods of treatment of patients by administering an effective amount of such compositions to a subject or a subject in need thereof, such as a subject prone to or afflicted with a vascular/cardiovascular disease or condition, a subject at risk of vascular/cardiovascular events, or a subject in need of treatment for a vascular/cardiovascular disease or condition.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention provides an orally administrable composition comprising fatty acids, wherein at least 50% by weight of the fatty acids comprise omega-3-fatty acids, salts, esters, or derivatives thereof, wherein the omega-3 fatty acids comprise eicosapentaenoic acid (EPA; C20:5-n3), docosapentaenoic acid (DPA; C22:5-n3), and docosahexaenoic acid (DHA; C22:6-n3), wherein the ratio of DHA to EPA (DHA:EPA) is less than 1:20, and wherein the ratio of DHA to DPA (DHA:DPA) is less than 2:1.
  • The present invention provides a pharmaceutical composition comprising fatty acids, wherein at least 50% by weight of the fatty acids comprise omega-3 fatty acids, salts, esters, or derivatives thereof, wherein the omega-3 fatty acids comprise eicosapentaenoic acid (EPA) and docosapentaenoic acid (DPA) and wherein the ratio of docosahexaenoic acid to DHA to EPA (DHA:EPA) is less than 1:10, and wherein the ratio of DHA to DPA (DHA:DPA) is less than 2:1, and methods of using this composition. In some embodiments the composition comprises EPA in an amount between about 70% and about 95% of the total amount of fatty acids. In some embodiments, the composition comprises less than about 5% of DHA of the total amount of fatty acids. In some embodiments, the composition comprises DPA in an amount of between about 5% and about 15% of the total amount of fatty acids. In some embodiments, the ratio of EPA to DPA (EPA:DPA) is less than about 1:1. In some embodiments. the composition further comprises heneicosapentaenoic acid (HPA) in an amount of at least 1% of the total amount of fatty acids
  • The present invention provides a pharmaceutical composition comprising eicosapentaenoic acid (EPA) in an amount between about 70% to about 95% of the total amount of fatty acids and docosapentaenoic acid (DPA), wherein the composition comprises no more than 5% docosahexaenoic acid (DHA) of the total amount of fatty acids, and wherein the ratio of DHA:DPA is 1:1 or lower, and methods of using the composition. In some embodiments, the composition comprises DPA in an amount of between about 5% and about 15% of the total amount of fatty acids. In some embodiments, the composition further comprises heneicosapentaenoic acid (HPA) in an amount of at least 1% of the total amount of fatty acids. In some embodiments, the ratio of EPA to DPA (EPA:DPA) is less than about 1:1. In some embodiments, the ratio of DHA:EPA is less than about 1:10.
  • The present invention provides a pharmaceutical composition comprising pharmaceutical composition comprising eicosapentaenoic acid (EPA) in an amount between about 750 mg/g to about 950 mg/g, and docosapentaenoic acid (DPA), wherein the composition comprises no more than 5% DHA of the total amount of fatty acids, and wherein the ratio of DHA:DPA is 1:1 or lower, and methods of using the composition. In some embodiments, the composition comprises about 60 mg/g to about 120 mg/g of DPA. In some embodiments, the ratio of DHA:EPA is less than 1:10. In some embodiments, the ratio of EPA:DPA is less than about 1:1. In some embodiments, the composition further comprises heneicosapentaenoic acid (HPA) in an amount of at least 1% of the total amount of fatty acids.
  • The present invention provides a pharmaceutical composition comprising eicosapentaenoic acid (EPA) in a daily dosage amount of between about 1000 mg to about 5000 mg, and further comprising docosapentaenoic acid (DPA) and docosahexaenoic acid (DHA), wherein the composition comprises no more than 5% DHA of the total amount of fatty acids, and wherein the ratio of DHA:DPA is 1:1 or lower, and methods of using the composition. In some embodiments, the ratio of DHA:EPA is less than about 1:10. In some embodiments, the ratio of EPA:DPA is less than about 1:1. In some embodiments, the composition further comprises heneicosapentaenoic acid (HPA) in an amount of at least 1% of the total amount of fatty acids.
  • The present invention provides a pharmaceutical composition comprising eicosapentaenoic acid (EPA) and docosapentaenoic acid (DPA), wherein the amount of EPA and DPA is about 55% or more by weight of the total amount of fatty acids, and wherein the ratio of DHA:DPA is no more than 1:1, and methods of using the composition. In some embodiments, the composition comprises docosahexaenoic acid (DHA) in an amount of less than about 30% of the total amount of fatty acids. In some embodiments, the composition comprises a daily dosage of DPA of greater than about 120 mg/day. In some embodiments, the composition comprises omega-6 fatty acids in an amount of no more than 6% of total amount of fatty acids. In some embodiments, the composition comprises DPA in an amount of at least 6% of the total amount of fatty acids.
  • The present invention provides a pharmaceutical composition comprising: docosapentaenoic acid (DPA) in an amount between about 50% to about 80% of the total amount of fatty acids, docosahexaenoic acid (DHA) in an amount between about 25% to about 40% of the total amount of fatty acids, and optionally eicosapentaenoic acid (EPA) in an amount less than about 10% of the total amount of fatty acids, and methods of using the composition. In some embodiments, the composition comprises docosapentaenoic acid (DPA) in an amount between about 50% to 75%, alternatively about 50% to about 70%, alternatively about 50% to about 65%, or alternatively about 50% to about 60%, of the total amount of fatty acids. In some embodiments, the composition comprises docosahexaenoic acid (DHA) in an amount between about 25% to about 38%, alternatively about 25% to about 35%, alternatively about 30% to about 35% of the total amount of fatty acids. In some embodiments, the composition comprises eicosapentaenoic acid (EPA) in an amount less than about 9%, alternatively less than about 8%, alternatively less than about 7%, alternatively less than about 6%, alternatively less than about 5%, of the total amount of fatty acids.
  • In some embodiments, the compositions of the present invention comprise at least 50% omega-3 fatty acids, alternatively at least 55%, alternatively at least 60%, alternatively at least 65%, alternatively at least 70%, alternatively at least 75%, alternatively at least 80%, alternatively at least 85%, alternatively at least 95%, most preferably at least 90% omega-3 fatty acids of the total amount of fatty acids. In some embodiments, the composition comprises at least about 92% to about 99%, alternatively about 93% to about 98%, alternatively about 94% to about 98%, omega-3 fatty acids of the total amount of fatty acids.
  • In other embodiments, EPA and DPA are jointly present in the compositions of the present invention at between about 55% and about 100% of total fatty acids, alternatively between about 60% and about 100%, alternatively between about 65% and about 100%, alternatively between about 70% and about 100%, alternatively between about 75% and about 100%, alternatively between about 80% and about 100%, alternatively between about 85% and about 95%, alternatively about 85% to about 100%, alternatively between about 85% and about 97%, alternatively between about 88% and about 95%, alternatively between about 88% and about 97%, alternatively about 88% to about 100%, alternatively between about 90% and about 95%, alternatively between about 90% and about 97%, alternatively about 90% to about 100%, alternatively about 95% to about 100%, alternatively about 97% to about 100% of the total amount of fatty acids.
  • On a EPA+DPA daily dose basis, the compositions of the present invention may be provided in a dose of between 100 mg and 10,100 mg/day, alternatively between 200 mg and 8,100 mg/day, alternatively between 300 mg and 6,100 mg/day, alternatively between 400 mg and 5,100 mg/day, alternatively between 500 mg and 4,100 mg/day. In some embodiments, on a EPA+HPA+DPA daily dose basis, the compositions and methods of the present invention may be provided in a dose of between 100 mg and 10,100 mg/day, alternatively between 200 mg and 8,100 mg/day, alternatively between 300 mg and 6,100 mg/day, alternatively between 400 mg and 5,100 mg/day, alternatively between 500 mg and 4,100 mg/day. In some embodiments, on an omega-3-pentaenoic acid daily dose basis, the methods and compositions of the present invention may be provided in a dose of between 100 mg and 10,100 mg/day, alternatively between 200 mg and 8,100 mg/day, alternatively between 300 mg and 6,100 mg/day, alternatively between 400 mg and 5,100 mg/day, alternatively between 500 mg and 4,100 mg/day.
  • The fatty acids, such as EPA and DPA, may be present in free fatty acid form, or as a salt, ester, or derivative. The fatty acids are preferably composed as a triglyceride, an ester (such as an ethyl ester) or free fatty acid. Other forms of the fatty acids which may be useful include salts, esters of any type, amides, mono-, di- or triglycerides, phospholipids or any other form which can lead to metabolization of the fatty acids (such as EPA and/or DPA), or the incorporation of the fatty acids (such as EPA and/or DPA) into body fluids, tissues or organs.
  • Omega-3 fatty acids may be grouped by the number of double bonds contained in the fatty acid chain. For instance, hexadecatrienoic acid (HTA), alpha-linolenic acid (ALA) and eicosatrienoic acid (ETE) are omega-3-trienoic acids; stearidonic acid (SDA) and eicosatetraenoic acid (ETA) are omega-3-tetraenoic acids; EPA, heneicosapentaenoic acid (HPA), DPA and tetracosapentaenoic acid (TPA) are omega-3-pentaenoic acids; and DHA and tetracosahexaenoic acid (THA) are omega-3-hexaenoic acids. In some preferred embodiments, the term omega-3-pentaenoic acids will refer to a mixture of at least two omega-3 pentaenoic acids in a ratio of at least 1:25, more preferably in a ratio of at least 1:50, more preferably in a ratio of at least 1:75, more preferably in a ratio of at least 1:100, more preferably in a ratio of at least 1:125, more preferably in a ratio of at least 1:150, more preferably in a ratio of at least 1:200. In some embodiments, the ratio refers to the ratio of the least prevalent omega-3 pentaenoic acid in the mixture to the most prevalent omega-3 pentaenoic acid in the mixture.
  • In some embodiments, the compositions of the present invention comprise EPA, HPA, DPA and TPA, alternatively EPA and DPA, and alternatively the compositions of the present invention comprise EPA, HPA and DPA.
  • In some embodiments, the omega-3-pentaenoic acids in the compositions of the present invention comprise no more than 99.5% of a single omega-3-pentaenoic acid, alternatively no more than 99%; alternatively no more than 98.5%; alternatively no more than 98%; alternatively no more than 97.5%; alternatively no more than 96%; alternatively no more than 95%; alternatively no more than 94%; alternatively no more than 93%; alternatively no more than 92%; alternatively no more than 91%; alternatively no more than 90%; alternatively no more than 88%; alternatively no more than 85%; alternatively no more than 80%; alternatively no more than 75%; alternatively no more than 70%; alternatively no more than 65%; alternatively no more than 60%; alternatively no more than 55%; alternatively no more than 50%; alternatively no more than 45%; alternatively no more than 40%; alternatively no more than 30%.
  • In some embodiments, the compositions of the present invention wherein at least 10%, alternatively at least 20%, alternatively at least 25%, alternatively at least 35%, alternatively at least 50%, alternatively at least 60%, alternatively at least 65%, alternatively at least 70%, alternatively at least 75%, by weight of the fatty acids comprise omega-3-pentaenoic acids, salts, esters, or derivatives thereof.
  • In some embodiments, compositions and methods comprise significant amounts of omega-3-pentaenoic acids or their glycerol or ethyl esters may be used in the methods of the present invention. In some embodiments, the compositions and methods comprise at least 100 mg omega-3-pentaenoic acids per day, alternatively at least 200 mg omega-3-pentaenoic acids per day, alternatively at least 300 mg omega-3-pentaenoic acids per day, alternatively at least 500 mg omega-3-pentaenoic acids per day, alternatively at least 700 mg omega-3-pentaenoic acids per day, alternatively at least 900 mg omega-3-pentaenoic acids per day, alternatively at least 1000 mg omega-3-pentaenoic acids per day, alternatively at least 1500 mg omega-3-pentaenoic acids per day, alternatively at least 1900 mg omega-3-pentaenoic acids per day, alternatively at least 2000 mg omega-3-pentaenoic acids per day, alternatively at least 2500 mg omega-3-pentaenoic acids per day, alternatively at least 2900 mg omega-3-pentaenoic acids per day, alternatively at least 3000 mg omega-3-pentaenoic acids per day, alternatively at least 3500 mg omega-3-pentaenoic acids per day, alternatively at least 3900 mg omega-3-pentaenoic acids per day, alternatively at least 4000 mg omega-3-pentaenoic acids per day, alternatively at least 4100 mg omega-3-pentaenoic acids per day, alternatively at least 4500 mg omega-3-pentaenoic acids per day, alternatively at least 5000 mg omega-3-pentaenoic acids per day, alternatively at least 5500 mg omega-3-pentaenoic acids per day, alternatively at least 6000 mg omega-3-pentaenoic acids per day, alternatively at least 6100 mg omega-3-pentaenoic acids per day or their glycerol or ethyl esters.
  • In some embodiments, the compositions provide a DHA as compared to the amount of omega-3-pentaenoic acids (N3-5enoicFA) such that the DHA:N3-5enoicFA ratio is no more than 15:1 of DHA:N3-5enoicFA, alternatively no more than 12:1 of DHA:N3-5enoicFA, alternatively no more than 10:1 of DHA:N3-5enoicFA, alternatively no more than 8:1 of DHA:N3-5enoicFA, alternatively no more than 5:1 of DHA:N3-5enoicFA, alternatively no more than 3:1 of DHA:N3-5enoicFA, alternatively no more than 2:1 of DHA:N3-5enoicFA, alternatively no more than 1:1 of DHA:N3-5enoicFA, alternatively no more than 1:2 of DHA:N3-5enoicFA, alternatively no more than 1:3 of DHA:N3-5enoicFA, alternatively no more than 1:5 of DHA:N3-5enoicFA, alternatively no more than 1:8 of DHA:N3-5enoicFA, alternatively no more than 1:10 of DHA:N3-5enoicFA, alternatively no more than 1:15 of DHA:N3-5enoicFA, alternatively a relative amount of no more than 1:20 of DHA:N3-5enoicFA.
  • In some embodiments, the compositions of the present invention comprise at least 0.01% HPA of total fatty acids in the composition, alternatively at least 0.05% HPA, alternatively at least 0.10% HPA, alternatively at least 0.15% HPA, alternatively at least 0.2% HPA, alternatively at least 0.3% HPA, alternatively at least 0.4% HPA, alternatively at least 0.5% HPA, alternatively at least 0.75% HPA, alternatively at least 1% HPA, alternatively at least 1.5% HPA, alternatively at least 2% HPA, alternatively at least 2.5% HPA, alternatively at least 3% HPA, alternatively at least 3.5% HPA, alternatively at least 4% HPA, alternatively at least 4.5% HPA, alternatively at least 5% HPA, alternatively at least 6% HPA, alternatively at least 7% HPA, alternatively the compositions of the present invention comprise at least 9% HPA of total fatty acids in the composition. In some embodiments, the compositions of the present invention comprise no more than 20% HPA of total fatty acids in the composition, alternatively no more than 15% HPA, alternatively no more than 12% HPA, alternatively no more than 10% HPA, alternatively no more than 8% HPA, alternatively no more than 7% HPA, alternatively no more than 6% HPA, alternatively no more than 5% HPA, alternatively no more than 4% HPA, alternatively no more than 3% HPA, alternatively no more than 2% HPA, alternatively no more than 1.5% HPA, alternatively the compositions of the present invention comprise at least 1% HPA of total fatty acids in the composition. In some embodiments, the compositions of the present invention comprise 1% to 20% HPA of the total fatty acids in the composition. In some embodiments, the compositions of the present invention comprise about 1% to about 6% HPA, alternatively about 2% to about 5% HPA, alternatively about 3% to about 4% HPA, relative to the total amount of fatty acids in the composition. In some embodiments, the compositions of the present invention comprise about 10 mg/g to about 50 mg/g HPA, alternatively about 15 mg/g to about 45 mg/g, alternatively about 20 mg/g to about 40 mg/g, alternatively about 25 mg/g to about 35 mg/g, alternatively about 30 mg/g HPA.
  • In some embodiments, the present invention provides compositions and methods which comprise significant amounts of omega-3 heneicosapentaenoic acid (HPA) or its glycerol or ethyl esters. In some embodiments, the methods of treatment may provide to a subject in need thereof a dose of at least 10 mg HPA per day, alternatively at least 15 mg HPA per day, alternatively at least 20 mg HPA per day, alternatively at least 25 mg HPA per day, alternatively at least 30 mg HPA per day, alternatively at least 40 mg HPA per day, alternatively at least 50 mg HPA per day, alternatively at least 60 mg HPA per day, alternatively at least 70 mg HPA per day, alternatively at least 80 mg HPA per day, alternatively at least 90 mg HPA per day, alternatively at least 100 mg HPA per day, alternatively at least 120 mg HPA per day, alternatively at least 150 mg HPA per day, alternatively at least 160 mg HPA per day, alternatively at least 180 mg HPA per day, alternatively at least 200 mg HPA per day, alternatively at least 250 mg HPA per day, alternatively at least 300 mg HPA per day, alternatively at least 350 mg HPA per day, alternatively at least 400 mg HPA per day, alternatively at least 500 mg HPA per day, alternatively at least 600 mg HPA per day, alternatively at least 800 mg HPA or its glycerol or ethyl esters per day.
  • In some embodiments, the compositions of the present invention comprise no more than 10% omega-3 fatty acids that are not omega-3-pentaenoic acids, alternatively no more than 9%, alternatively no more than 8%, alternatively no more than 7%, alternatively no more than 6%, alternatively no more than 5%, alternatively no more than 4.5%, alternatively no more than 4%, alternatively no more than 3.5%, alternatively no more than 3%, alternatively no more than 2.5%, alternatively no more than 2%, alternatively no more than 1.5%, alternatively no more than 1.25%, alternatively no more than 1%, alternatively no more than 0.75%, alternatively no more than 0.5%, alternatively no more than 0.4%, alternatively no more than 0.3%, alternatively no more than 0.2%, alternatively the compositions of the present invention comprise no more than 0.1% omega-3 fatty acids that are not omega-3-pentaenoic acids.
  • In the embodiments of the present invention, the compositions comprise EPA and DPA in an EPA:DPA ratio between 99:1 and 1:99 EPA:DPA, alternatively between 90:1 and 1:90, alternatively between 60:1 and 1:60, alternatively between 60:1 and 1:20, alternatively between 60:1 and 1:4, alternatively between 40:1 and 1:20, alternatively between 30:1 and 1:20, alternatively between 30:1 and 1:10, alternatively between 30:1 and 1:5, alternatively between 40:1 and 1:4, alternatively between 30:1 and 1:4, alternatively between 30:1 and 1:2, alternatively between 30:1 and 1:1, alternatively between 30:1 and 2:1, alternatively between 30:1 and 5:1, alternatively between 20:1 and 1:20, alternatively between 20:1 and 1:10, alternatively between 20:1 and 1:5, alternatively between 20:1 and 1:2, alternatively between 20:1 and 1:1, alternatively between 20:1 and 2:1, alternatively between 20:1 and 5:1, alternatively between 20:1 and 10:1, alternatively between 20:1 and 10:1, alternatively between 30:1 and 10:1, alternatively between 60:1 and 10:1, alternatively comprise EPA and DPA in an EPA:DPA ratio between 40:1 and 10:1. In some embodiments, the ratio of EPA:DPA is greater than 1:1, preferably greater than 2:1, and more preferably greater than 5:1. In some embodiments, the ratio of EPA:DPA is 1:1 to 25:1, preferably 5:1 to 20:1, more preferably 8:1 to 15:1, even more preferably 9:1 to 13:1, even more most preferably about 10:1 to 11:1, and most preferably about 10:1.
  • In some embodiments of the present invention, the compositions comprise EPA in an amount between 55% and 95% relative to the total amount of fatty acids present in the composition, alternatively between 60% and 95%, alternatively between 65% and 95%, alternatively between 70% and 95%, alternatively between 75% and 95%, alternatively between 90% and 95%, alternatively between 80% and 95%, alternatively between 90% and 95%, alternatively between 55% and 90%, alternatively between 60% and 90%, alternatively between 65% and 90%, alternatively between 70% and 90%, alternatively between 75% and 90%, alternatively between 80% and 90%, alternatively between 85% and 90%, alternatively between 55% and 92%, alternatively between 60% and 92%, alternatively between 65% and 92%, alternatively between 70% and 92%, alternatively between 75% and 92%, alternatively between 80% and 92%, alternatively between 85% and 92%, alternatively between 55% and 93%, alternatively between 60% and 93%, alternatively between 65% and 93%, alternatively between 70% and 93%, alternatively between 75% and 93%, alternatively between 80% and 93%, alternatively between 85% and 93%, alternatively more than 85%, alternatively more than 85%, alternatively between 85% and 95% EPA relative to the total amount of fatty acids present in the composition. In some embodiments, the compositions comprise about 70% to about 95%, 80% to about 90%, alternatively about 81% to about 88%, alternatively about 82% to about 88%, alternatively about 83% to about 87%, alternatively about 84% to about 86%, alternatively about 85% EPA relative to the total amount of fatty acids present in the composition. In some embodiments, the compositions comprise about 750 mg/g to about 950 mg/g, alternatively about 800 mg/g to about 900 mg/g, alternatively about 830 mg/g to about 870 mg/g, alternatively about 840 mg/g to about 870 mg/g, alternatively 845 mg/g to about 865 mg/g, alternatively 846 mg/g to about 860 mg/g, alternatively 847 mg/g to about 859 mg/g, alternatively about 848 mg/g to about 858 mg/g, alternatively about 849 mg/g to about 857 mg/g, alternatively about 850 mg/g to about 856 mg/g, alternatively about 851 mg/g to about 855 mg/g, alternatively about 852 mg/g to about 854 mg/g, alternatively about 853 mg/g EPA.
  • On a EPA daily dose basis, the compositions of the present invention are preferably provided in a dose of between 100 mg and 10,000 mg/day, alternatively between 200 mg and 8,000 mg/day, alternatively between 300 mg and 6,000 mg/day, alternatively between 400 mg and 5,000 mg/day, alternatively between 500 mg and 4,000 mg/day. In some embodiments, the compositions and methods of the present invention are provided in a dose of between about 1000 mg/day to about 5000 mg/day, alternatively about 1200 mg/day to about 3000 mg/day, alternatively about 1500 mg/day to about 2500 mg/day, alternatively 1600 mg/day to about 1950 mg/day, alternatively about 1735 mg/day to about 1855 mg/day, alternatively about 1740 mg/day to about 1840 mg/day, alternatively about 1745 mg/day to about 1820 mg/day, alternatively about 1750 mg/day to about 1800 mg/day, alternatively about 1755 mg/day to about 1790 mg/day, alternatively about 1760 mg/day to about 1780 mg/day, alternatively about 1770 mg/day of EPA. In some embodiments, the compostions of the present invention are provided in a dose of between about 2300 mg/day to about 3000 mg/day, alternatively about 2400 mg/day to about 2800 mg/day, alternatively about 2520 mg/day to about 2780 mg/day, alternatively about 2600 mg/day to about 2700 mg/day, alternatively about 2610 mg/day to about 2680 mg/day, alternatively about 2620 mg/day to about 2670 mg/day, alternatively about 2630 mg/day to about 2665 mg/day, alternatively about 2640 mg/day to about 2660 mg/day, alternatively about 2650 mg/day of EPA. In some embodiments, the compostions of the present invention are provided in a dose of between about 3200 mg/day to about 3900 mg/day, alternatively 3300 mg/day to about 3800 mg/day, alternatively 3360 mg/day to about 3710 mg/day, alternatively about 3400 mg/day to about 3700 mg/day, alternatively about 3450 mg/day to about 3650 mg/day, alternatively about 3500 mg/day to about 3600 mg/day, alternatively about 3530 mg/day to about 3580 mg/day, alternatively about 3540 mg/day to about 3560 mg/day, alternatively about 3550 mg/day of EPA.
  • In some embodiments, the compositions of the present invention are provided in a dose of between about 1650 mg/day to about 2050 mg/day, alternatively about 1700 mg/day to about 2000 mg/day, alternatively about 1750 mg/day to about 1950 mg/day, alternatively about 1775 mg/day to about 1925 mg/day, alternatively about 1800 mg/day to about 1900 mg/day, alternatively about 1800 mg/day to about 2000 mg/day, alternatively about 1820 mg/day to about 1880 mg/day, alternatively about 1830 mg/day to about 1870 mg/day, alternatively about 1840 mg/day to about 1860 mg/day, alternatively about 1850 mg/day of EPA. In some embodiments, the compostions of the present invention are provided in a dose of between about 2500 mg/day to about 3100 mg/day, alternatively about 2600 mg/day to about 2000 mg/day, alternatively about 2650 mg/day to about 2950 mg/day, alternatively about 2700 mg/day to about 2900 mg/day, alternatively about 2725 mg/day to about 2875 mg/day, alternatively about 2750 mg/day to about 2850 mg/day, alternatively about 2780 mg/day to about 2820 mg/day, alternatively about 2790 mg/day to about 2810 mg/day, alternatively about 2800 mg/day of EPA. In some embodiments, the compostions of the present invention are provided in a dose of between about 3300 mg/day to about 4000 mg/day, alternatively about 3400 mg/day to about 3900 mg/day, alternatively about 3500 mg/day to about 3900 mg/day, alternatively about 3550 mg/day to about 3850 mg/day, alternatively about 3600 mg/day to about 3800 mg/day, alternatively about 3650 mg/day to about 3750 mg/day, alternatively about 3680 mg/day to about 3725 mg/day, alternatively about 3690 mg/day to about 3710 mg/day, alternatively about 3700 mg/day of EPA.
  • In some embodiments, the compositions of the present invention are provided in a dose of between about 1500 mg/day to about 2500 mg/day, alternatively about 1750 mg/day to about 2300 mg/day, alternatively about 1800 mg/day to about 2200 mg/day, alternatively about 1900 mg/day to about 2100 mg/day, alternatively about 1950 mg/day to about 2050 mg/day, alternatively about 1975 mg/day to about 2025 mg/day, alternatively about 2000 mg/day of EPA. In some embodiments, the compositions of the present invention are provided in a dose of between about 2500 mg/day to about 3500 mg/day, alternatively about 2700 mg/day to about 3300 mg/day alternatively about 2750 mg/day to about 3300 mg/day, alternatively about 2800 mg/day to about 3200 mg/day, alternatively about 2900 mg/day to about 3100 mg/day, alternatively about 2950 mg/day to about 3050 mg/day, alternatively about 2975 mg/day to about 3025 mg/day, alternatively about 3000 mg/day of EPA. In some embodiments, the compositions of the present invention are provided in a dose of between about 3500 mg/day to about 4500 mg/day, alternatively about 3700 mg/day to about 4300 mg/day, alternatively about 3750 mg/day to about 4300 mg/day, alternatively about 3800 mg/day to about 4200 mg/day, alternatively about 3900 mg/day to about 4100 mg/day, alternatively about 3950 mg/day to about 4050 mg/day, alternatively about 3975 mg/day to about 4025 mg/day, alternatively about 4000 mg/day of EPA.
  • In other embodiments of the present invention, the compositions comprise DPA in an amount between 1% and 99% relative to the total amount of fatty acids present in the composition, alternatively between 1% and 95%, alternatively alternatively between 1% and 90%, alternatively between 1% and 85%, alternatively between 1% and 80%, alternatively between 1% and 75%, alternatively between 1% and 70%, alternatively between 1% and 65%, alternatively between 1% and 60%, alternatively between 1% and 55%, alternatively between 1% and 50%, alternatively between 1% and 45%, alternatively between 1% and 40%, alternatively between 1% and 35%, alternatively between 1% and 30%, alternatively between 1% and 25%, alternatively between 1% and 20%, alternatively between 1% and 15%, alternatively between 1% and 10%, alternatively between 1% and 5%, alternatively between 2% and 99%, alternatively between 2% and 95%, alternatively between 2% and 90%, alternatively between 2% and 85%, alternatively between 2% and 80%, alternatively between 2% and 75%, alternatively between 2% and 70%, alternatively between 2% and 65%, alternatively between 2% and 60%, alternatively between 2% and 55%, alternatively between 2% and 50%, alternatively between 2% and 45%, alternatively between 2% and 40%, alternatively between 2% and 35%, alternatively between 2% and 30%, alternatively between 2% and 25%, alternatively between 2% and 20%, alternatively between 2% and 15%, alternatively between 2% and 10%, alternatively between 2% and 5%, alternatively between 3% and 99%, alternatively between 3% and 95%, alternatively between 3% and 90%, alternatively between 3% and 85%, alternatively between 3% and 80%, alternatively between 3% and 75%, alternatively between 3% and 70%, alternatively between 3% and 65%, alternatively between 3% and 60%, alternatively between 3% and 55%, alternatively between 3% and 50%, alternatively between 3% and 45%, alternatively between 3% and 40%, alternatively between 3% and 35%, alternatively between 3% and 30%, alternatively between 3% and 25%, alternatively between 3% and 20%, alternatively between 3% and 15%, alternatively between 3% and 10%, alternatively between 3% and 5%, alternatively between 4% and 99%, alternatively between 4% and 95%, alternatively between 4% and 90%, alternatively between 4% and 85%, alternatively between 4% and 80%, alternatively between 4% and 75%, alternatively between 4% and 70%, alternatively between 4% and 65%, alternatively between 4% and 60%, alternatively between 4% and 55%, alternatively between 4% and 50%, alternatively between 4% and 45%, alternatively between 4% and 40%, alternatively between 4% and 35%, alternatively between 4% and 30%, alternatively between 4% and 25%, alternatively between 4% and 20%, alternatively between 4% and 15%, alternatively between 4% and 10%, alternatively between 4% and 5%, alternatively between 5% and 99%, alternatively between 5% and 95%, alternatively between 5% and 90%, alternatively between 5% and 85%, alternatively between 5% and 80%, alternatively between 5% and 75%, alternatively between 5% and 70%, alternatively between 5% and 65%, alternatively between 5% and 60%, alternatively between 5% and 55%, alternatively between 5% and 50%, alternatively between 5% and 45%, alternatively between 5% and 40%, alternatively between 5% and 35%, alternatively between 5% and 30%, alternatively between 5% and 25%, alternatively between 5% and 20%, alternatively between 5% and 15%, alternatively between 5% and 12%, alternatively between 5% and 10%, alternatively between 6% and 99%, alternatively between 6% and 95%, alternatively between 6% and 90%, alternatively between 6% and 85%, alternatively between 6% and 80%, alternatively between 6% and 75%, alternatively between 6% and 70%, alternatively between 6% and 65%, alternatively between 6% and 60%, alternatively between 6% and 55%, alternatively between 6% and 50%, alternatively between 6% and 45%, alternatively between 6% and 40%, alternatively between 6% and 35%, alternatively between 6% and 30%, alternatively between 6% and 25%, alternatively between 6% and 20%, alternatively between 6% and 15%, alternatively between 6% and 12%, alternatively between 6% and 11%, alternatively between 6% and 10%, alternatively between 7% and 99%, alternatively between 7% and 95%, alternatively between 7% and 90%, alternatively between 7% and 85%, alternatively between 7% and 80%, alternatively between 7% and 75%, alternatively between 7% and 70%, alternatively between 7% and 65%, alternatively between 7% and 60%, alternatively between 7% and 55%, alternatively between 7% and 50%, alternatively between 7% and 45%, alternatively between 7% and 40%, alternatively between 7% and 35%, alternatively between 7% and 30%, alternatively between 7% and 25%, alternatively between 7% and 20%, alternatively between 7% and 15%, alternatively between 7% and 12%, alternatively between 7% and 11%, alternatively between 7% and 10%, alternatively between 8% and 99%, alternatively between 8% and 95%, alternatively between 8% and 90%, alternatively between 8% and 85%, alternatively between 8% and 80%, alternatively between 8% and 75%, alternatively between 8% and 70%, alternatively between 8% and 65%, alternatively between 8% and 60%, alternatively between 8% and 55%, alternatively between 8% and 50%, alternatively between 8% and 45%, alternatively between 8% and 40%, alternatively between 8% and 35%, alternatively between 8% and 30%, alternatively between 8% and 25%, alternatively between 8% and 20%, alternatively between 8% and 15%, alternatively between 8% and 12%, alternatively between 9% and 95%, alternatively between 9% and 90%, alternatively between 9% and 85%, alternatively between 9% and 80%, alternatively between 9% and 75%, alternatively between 9% and 70%, alternatively between 9% and 65%, alternatively between 9% and 60%, alternatively between 9% and 55%, alternatively between 9% and 50%, alternatively between 9% and 45%, alternatively between 9% and 40%, alternatively between 9% and 35%, alternatively between 9% and 30%, alternatively between 9% and 25%, alternatively between 9% and 20%, alternatively between 9% and 15%, alternatively between 9% and 12%, relative to the total amount of fatty acids present in the composition. In some embodiments, the compositions comprise docosapentaenoic acid (DPA) in an amount between about 5% to about 15%, alternatively about 6% to about 12%, alternatively about 7% to about 11%, alternatively about 8% to about 10% relative to the total amount of fatty acids present in the composition. In some alternative embodiments, the composition comprises at least about 4% or at least about 5% or at least about 6% or at least about 7% or at least about 8% or at least about 9% or at least about 10% or at least about 15% or at least about 20% or at least about 25% or at least about 30% or at least about 35% or at least about 40% or at least about 45% or at least about 50% or at least about 55% or at least about 60% or at least about 65% or at least about 70% or at least about 75% or at least about 80% or at least about 85% or at least about 90% or at least about 95% of docosapentaenoic acid (DPA).
  • In some embodiments, the compositions comprise docosapentaenoic acid (DPA) in an amount of about 60 mg/g to about 120 mg/g, alternatively about 70 mg/g to about 100 mg/g, 75 mg/g to about 90 mg/g, alternatively about 77 mg/g to about 85 mg/g, alternatively about 78 mg/g to about 84 mg/g, alternatively about 79 mg/g to about 83 mg/g, alternatively about 80 mg/g to about 82 mg/g, alternatively about 81 mg/g to about 82 mg/g. In some embodiments, the compositions comprise docosapentaenoic acid (DPA) in an daily dosage amount of at least about 20 mg/day, alternatively at least about 25 mg/day, alternatively at least about 30 mg/day, alternatively at least about 40 mg/day, alternatively at least about 50 mg/day, alternatively at least about 60 mg/day alternatively, at least about 70 mg/day alternatively at least about 75 mg/day, alternatively at least about 80 mg/day, alternatively at least about 90 mg/day, alternatively at least about 100 mg/day, alternatively at least about 120 mg/day, alternatively at least about 150 mg/day, alternatively at least about 160 mg/day, alternatively at least about 180 mg/day, alternatively at least about 200 mg/day, alternatively at least about 250 mg/day, alternatively at least about 300 mg/day, alternatively at least about 350 mg/day, or alternatively at least about 400 mg/day, alternatively at least about 500 mg/day, alternatively at least about 600 mg/day, alternatively at least about 800 mg/day, or alternatively at least about 1000 mg/day, alternatively at least about 1200 mg/day, alternatively at least about 1500 mg/day, or alternatively at least about 2000 mg/day, or alternatively at least about 3000 mg/day, or alternatively at least about 3500 mg/day, or alternatively at least about 4000 mg/day, or alternatively at least about 4250 mg/day. In some embodiments, the composition comprises DPA in a daily dosage of about 120 mg/day to about 150 mg/day, alternatively about 150 mg/day to about 200 mg/day, alternatively about 200 mg/day to about 250 mg/day, alternatively about 250 mg/day to about 300 mg/day, alternatively about 300 mg/day to about 400 mg/day, alternatively about 400 mg/day to about 600 mg/day, alternatively about 600 mg/day to about 1000 mg/day. In some embodiments, the method of treatment provides a dose of at least about 1 mg/kg of docosapentaenoic acid (DPA) per day, alternatively about 2 mg/kg of DPA per day, alternatively about 3 mg/kg of DPA per day, alternatively about 4 mg/kg of DPA per day, alternatively about 6 mg/kg of DPA per day, alternatively about 8 mg/kg of DPA per day, alternatively about 10 mg/kg of DPA per day, alternatively about 20 mg/kg of DPA per day, alternatively about 30 mg/kg of DPA per day, and alternatively about 40 mg/kg of DPA per day, alternatively about 50 mg/kg of DPA per day, alternatively about 75 mg/kg of DPA per day, and alternatively about 100 mg/kg of DPA per day.
  • In some embodiments, a relatively small amount of docosahexaenoic acid (DHA) as compared to EPA is present. In some embodiments, the compositions of the present invention comprise no more than 1:1 of DHA:EPA, alternatively no more than 1:2, alternatively no more than 1:3, alternatively no more than 1:3, alternatively no more than 1:4, alternatively no more than 1:5 of DHA:EPA, alternatively no more than 1:6 of DHA:EPA, alternatively no more than 1:7 of DHA:EPA, alternatively no more than 1:8 of DHA:EPA, alternatively no more than 1:9 of DHA:EPA, alternatively no more than 1:10 of DHA:EPA, alternatively no more than 1:12 of DHA:EPA, alternatively no more than 1:15 of DHA:EPA, alternatively no more than 1:20 of DHA:EPA, alternatively no more than 1:25 of DHA:EPA, alternatively no more than 1:30 of DHA:EPA, alternatively no more than 1:40 of DHA:EPA, alternatively no more than 1:50 of DHA:EPA, alternatively no more than 1:75 of DHA:EPA, alternatively no more than 1:90 of DHA:EPA, alternatively no more than 1:99 of DHA:EPA. Alternatively, DHA may be present in the compositions of this invention at a relative amount of ratio less than 1% than the amount of EPA. Alternatively, docosahexaenoic acid (DHA) may be present in the compositions of this invention at a DHA:EPA ratio of less than 1:99.
  • In some embodiments, a relatively small amount of docosahexaenoic acid (DHA) relative to the total amount of fatty acids present in the composition is present. In some embodiments, the compositions of the present invention comprise no more than 30% DHA, alternatively no more than 20% DHA, alternatively no more than 15% DHA, alternatively no more than 12% DHA, alternatively no more than 10% DHA, alternatively no more than 9% DHA, alternatively no more than 8% DHA, alternatively no more than 7% DHA, alternatively no more than 6% DHA, alternatively no more than 5% DHA, alternatively no more than 4% DHA, alternatively no more than 3% DHA, alternatively no more than 2% DHA, alternatively no more than 1% DHA relative to the total amount of fatty acids present in the composition. In some embodiments, the compositions and methods comprise less than 1500 mg of DHA, alternatively less than 1200 mg of DHA, alternatively less than 1000 mg of DHA, alternatively less than 800 mg of DHA, alternatively less than 700 mg of DHA, alternatively less than 600 mg of DHA, alternatively less than 500 mg of DHA, alternatively less than 400 mg of DHA, alternatively less than 350 mg of DHA, alternatively less than 300 mg of DHA, alternatively less than 250 mg of DHA, alternatively less than 200 mg of DHA alternatively less than 150 mg of DHA, alternatively less than 120 mg of DHA, alternatively less than 100 mg of DHA, alternatively less than 80 mg of DHA, alternatively less than 60 mg of DHA, alternatively less than 40 mg of DHA, alternatively less than 30 mg of DHA, alternatively less than 25 mg of DHA, alternatively less than 20 mg of DHA or its glycerol or ethyl esters as a totally daily dose.
  • In some embodiments, the composition comprises about 5 mg/g to about 20 mg/g, alternatively about 8 mg/g to about 18 mg/g, alternatively about 9 mg/g to about 15 mg/g, alternatively about 10 mg/g to about 14 mg/g, alternatively about 11 mg/g to about 13 mg/g, alternatively about 12 mg/g to about 13 mg/g of docosahexaenoic acid (DHA).
  • In some embodiments, the ratio of EPA:HPA is about 1500:1 to 25:1, alternatively 1000:1 to 50:1, alternatively 800:1 to 60:1, alternatively 500:1 to 60:1, alternatively 250:1 to 75:1, and alternatively 100:1 to 80:1. In some preferred embodiments, the ratio of EPA:HPA is about 85:1. In some preferred embodiments, the ratio of EPA:HPA is about 30:1. In some embodiments, the ratio of DPA:HPA is about 250:1 to 1:1, alternatively 200:1 to 2:1, alternatively 150:1 to 3:1, alternatively 100:1 to 4:1, alternatively 50:1 to 5:1, alternatively 25:1 to 6:1, and alternatively 10:1 to 7:1. In some preferred embodiments, the ratio of DPA:HPA is about 8:1. In some embodiments, the ratio of DPA:HPA is about 3:0.
  • In other embodiments, a relatively small amount of DHA as compared to DPA is present. In these embodiments, the compositions of the present invention comprise no more than 15:1 of DHA:DPA, alternatively no more than 12:1 of DHA:DPA, alternatively no more than 10:1 of DHA:DPA, alternatively no more than 8:1 of DHA:DPA, alternatively no more than 5:1 of DHA:DPA, alternatively no more than 4:1 of DHA:DPA, alternatively no more than 3:1 of DHA:DPA, alternatively no more than 2:1 of DHA:DPA, alternatively no more than 1:1 of DHA:DPA, alternatively no more than 1:2 of DHA:DPA, alternatively no more than 1:3 of DHA:DPA, alternatively no more than 1:4 of DHA:DPA, alternatively no more than 1:5 of DHA:DPA, alternatively no more than 1:6 of DHA:DPA, alternatively no more than 1:7 of DHA:DPA, alternatively no more than 1:8 of DHA:DPA, alternatively no more than 1:10 of DHA:DPA, alternatively no more than 1:12 of DHA:DPA, alternatively no more than 1:15 of DHA:DPA, alternatively no more than 1:20 of DHA:DPA, alternatively no more than 1:25 of DHA:DPA, alternatively no more than 1:50 of DHA:DPA, alternatively no more than 1:75 of DHA:DPA, alternatively no more than 1:90 of DHA:DPA, alternatively no more than 1:95 of DHA:DPA, alternatively no more than 1:100 of DHA:DPA. In some embodiments, the ratio of DHA:DPA is preferably less than 2:1.
  • In other embodiments, a relatively small amount of DHA as compared to HPA is present. In these embodiments, the compositions of the present invention comprise no more than 15:1 of DHA:HPA, alternatively no more than 12:1 of DHA:HPA, alternatively no more than 10:1 of DHA:HPA, alternatively no more than 8:1 of DHA:HPA, alternatively no more than 5:1 of DHA:HPA, alternatively no more than 3:1 of DHA:HPA, alternatively no more than 2:1 of DHA:HPA, alternatively no more than 1:1 of DHA:HPA, alternatively no more than 1:2 of DHA:HPA, alternatively no more than 1:3 of DHA:HPA, alternatively no more than 1:5 of DHA:HPA, alternatively no more than 1:8 of DHA:HPA, alternatively no more than 1:10 of DHA:HPA, alternatively no more than 1:15 of DHA:HPA, alternatively no more than 1:20 of DHA:HPA, alternatively no more than 1:25 of DHA:HPA, alternatively no more than 1:50 of DHA:HPA, alternatively no more than 1:75 of DHA:HPA, alternatively no more than 1:90 of DHA:HPA, alternatively no more than 1:95 of DHA:HPA, alternatively no more than 1:100 of DHA:HPA.
  • In yet other embodiments, the compositions of the present invention comprise no more than 10% omega-6 fatty acids relative to the total amount of fatty acids, alternatively no more than 9%, alternatively no more than 8%, alternatively no more than 7%, alternatively no more than 6%, alternatively no more than 5%, alternatively no more than 4.5%, alternatively no more than 4%, alternatively no more than 3.5%, alternatively no more than 3%, alternatively no more than 2.5%, alternatively no more than 2%, alternatively no more than 1.7%, alternatively no more than 1.5%, alternatively no more than 1.2%, alternatively no more than 1%, alternatively no more than 0.5% omega-6 fatty acids versus the total amount of fatty acids comprised by the compositions of the present invention.
  • Omega-6 fatty acids include, but are not limited to: linoleic acid (LA; C18:2-n6); gamma-linoleic acid (GLA; C18:3-n6); eicosadienoic acid (C20:2-n6); dihomo-gamma-linoleic acid (DGLA; C20:3-n6); arachiconic acid (ARA; C20:4-n6); and omega-6 docosapentaenoic acid (DPA; C22:5-n6).
  • In further embodiments, the compositions of the present invention comprise no more than 10% omega-6 fatty acids relative to the total amount of omega-3 fatty acids plus omega-6 fatty acids, alternatively no more than 9%, alternatively no more than 8%, alternatively no more than 7%, alternatively no more than 6%, alternatively no more than 5%, alternatively no more than 4.5%, alternatively no more than 4%, alternatively no more than 3.5%, alternatively no more than 3%, alternatively no more than 2.5%, alternatively no more than 2%, alternatively no more than 1.7%, alternatively no more than 1.5%, alternatively no more than 1.2%, alternatively no more than 1%, alternatively no more than 0.5% omega-6 fatty acids versus the total amount of omega-3 fatty acids plus omega-6 fatty acids comprised by the compositions of the present invention.
  • In yet other embodiments, the compositions of the present invention comprise no more than 8% arachidonic acid (ARA; C20:4-n6) relative to the total amount of omega-3 fatty acids plus omega-6 fatty acids, alternatively no more than 7%, alternatively no more than 6%, alternatively no more than 5%, alternatively no more than 4.5%, alternatively no more than 4%, alternatively no more than 3.5%, alternatively no more than 3%, alternatively no more than 2.5%, alternatively no more than 2%, alternatively no more than 1.7%, alternatively no more than 1.5%, alternatively no more than 1.2%, alternatively no more than 1%, alternatively no more than 0.5% arachidonic acid (ARA; 020:4-n6) versus the total amount of omega-3 fatty acids plus omega-6 fatty acids comprised by the compositions of the present invention.
  • In some embodiments, a relatively small amount of omega-3 fatty acids in aggregate other than EPA, ETA, HPA and DPA (alternatively indicated as non-EPA, non-ETA, non-HPA and non-DPA omega-3 fatty acids in aggregate) relative to the total amount of fatty acids present in the composition is present. In some embodiments, the compositions of the present invention comprise no more than 20% non-EPA, non-ETA, non-H PA and non-DPA omega-3 fatty acids, alternatively no more than 15% non-EPA, non-ETA, non-HPA and non-DPA omega-3 fatty acids, alternatively no more than 12% non-EPA, non-ETA, non-H PA and non-DPA omega-3 fatty acids, alternatively no more than 10% non-EPA, non-ETA, non-HPA and non-DPA omega-3 fatty acids, alternatively no more than 8% non-EPA, non-ETA, non-HPA and non-DPA omega-3 fatty acids, alternatively no more than 7% non-EPA, non-ETA, non-HPA and non-DPA omega-3 fatty acids, alternatively no more than 6% non-EPA, non-ETA, non-HPA and non-DPA omega-3 fatty acids, alternatively no more than 5% non-EPA, non-ETA, non-HPA and non-DPA omega-3 fatty acids, alternatively no more than 4% non-EPA, non-ETA, non-HPA and non-DPA omega-3 fatty acids, alternatively no more than 3% non-EPA, non-ETA, non-HPA and non-DPA omega-3 fatty acids, alternatively no more than 2% non-EPA, non-ETA, non-HPA and non-DPA omega-3 fatty acids, alternatively no more than 1% non-EPA, non-ETA, non-HPA and non-DPA omega-3 fatty acids in aggregate relative to the total amount of fatty acids present in the composition.
  • In some embodiments, a relatively small amount of the sum of ALA, SDA and DHA relative to the total amount of fatty acids present in the composition is present, while at the same time large amounts of the sum of EPA, DPA-n3, HPA and ETA are present. In some embodiments, the compositions of the present invention comprise no more than 20% of the sum of ALA, SDA and DHA, alternatively no more than 15% of the sum of ALA, SDA and DHA, alternatively no more than 12% of the sum of ALA, SDA and DHA, alternatively no more than 10% of the sum of ALA, SDA and DHA, alternatively no more than 8% of the sum of ALA, SDA and DHA, alternatively no more than 7% of the sum of ALA, SDA and DHA, alternatively no more than 6% of the sum of ALA, SDA and DHA, alternatively no more than 5% of the sum of ALA, SDA and DHA, alternatively no more than 4% of the sum of ALA, SDA and DHA, alternatively no more than 3% of the sum of ALA, SDA and DHA, alternatively no more than 2% of the sum of ALA, SDA and DHA, alternatively no more than 1% of the sum of ALA, SDA and DHA relative to the total amount of fatty acids present in the composition, while at the same time contain more than 40% the sum of EPA, DPAn-3, HPA and ETA, alternatively more than 50% the sum of EPA, DPAn-3, HPA and ETA, alternatively more than 60% the sum of EPA, DPAn-3, HPA and ETA, alternatively more than 70% the sum of EPA, DPAn-3, HPA and ETA, alternatively more than 75% the sum of EPA, DPAn-3, HPA and ETA, alternatively more than 80% the sum of EPA, DPAn-3, HPA and ETA, alternatively more than 85% the sum of EPA, DPAn-3, HPA and ETA, alternatively more than 90% the sum of EPA, DPAn-3, HPA and ETA, alternatively more than 95% the sum of EPA, DPAn-3, HPA and ETA, alternatively between 80% and 98% the sum of EPA, DPAn-3, HPA and ETA, alternatively between 80% and 96% the sum of EPA, DPAn-3, HPA and ETA, alternatively between 85% and 98% the sum of EPA, DPAn-3, HPA and ETA, alternatively between 85% and 96% the sum of EPA, DPAn-3, HPA and ETA, alternatively between 90% and 98% the sum of EPA, DPAn-3, HPA and ETA, alternatively between 90% and 97% the sum of EPA, DPAn-3, HPA and ETA, alternatively between 90% and 96% the sum of EPA, DPAn-3, HPA and ETA, alternatively between 90% and 95% the sum of EPA, DPAn-3, HPA and ETA, relative to the total amount of fatty acids present in the composition is present.
  • In further embodiments, the compositions of the present invention comprise no more than 8% arachidonic acid (ARA; C20:4-n6) relative to the total amount of fatty acids, alternatively no more than 7%, alternatively no more than 6%, alternatively no more than 5%, alternatively no more than 4.5%, alternatively no more than 4%, alternatively no more than 3.5%, alternatively no more than 3%, alternatively no more than 2.5%, alternatively no more than 2%, alternatively no more than 1.7%, alternatively no more than 1.5%, alternatively no more than 1.2%, alternatively no more than 1%, alternatively no more than 0.5% arachidonic acid (ARA; C20:4-n6) relative the total amount of fatty acids comprised by the compositions of the present invention.
  • In other embodiments, the compositions of the present invention comprise no more than 2.5% arachidonic acid (ARA; C20:4-n6), no more than 0.4% omega-6-docosapentaenoic acid (DPA; C22:5-n6) and no more than 0.2% gamma-linoleic acid (GLA; C18:3-n6) relative the total amount of fatty acids comprised by the compositions of the present invention.
  • Further embodiments provide fatty acid compositions comprising no more than 2.5% arachidonic acid (ARA; C20:4-n6), no more than 0.3% omega-6 docosapentaenoic acid (DPA; C22:5-n6) and no more than 0.1% gamma-linoleic acid (GLA; C18:3-n6) relative the total amount of fatty acids comprised, by the compositions of the present invention.
  • In some embodiments, the composition of the present invention further comprises TPA at concentration of at least 0.05%. In some embodiments, the TPA concentration is about 0.01% to about 5%, alternatively about 0.05% to about 2%, alternatively about 0.1% to about 1%, alternatively about 0.2% to about 0.8%, alternatively about 0.4% to about 0.6%, alternatively about 0.5%.
  • The compositions of the present invention may also be taken as a general nutritional supplement.
  • In yet other embodiments, the active ingredient of the formulations of the present invention consists essentially wholly of the EPA and DPA or precursors thereof (ethyl ester, triglyceride, or any other pharmaceutically acceptable salt or derivative thereof). In that case, no large amounts (preferably less than 15%, alternatively less than 12%, alternatively less than 10%, alternatively less than 9%, alternatively less than 8%, alternatively less than 7%, alternatively less than 6%, alternatively less than 5%, alternatively less than 4%, alternatively less than 3%, alternatively less than 2%, alternatively less than 1%, alternatively less than 0.5%, alternatively less than 0.25%) of any other fatty acids are present.
  • In further embodiments, the active ingredient of the formulations of the present invention consists essentially wholly of omega-3-pentaenoic acids or precursors thereof (ethyl ester, triglyceride, or any other pharmaceutically acceptable salt or derivative thereof). In that case, no large amounts (preferably less than 15%, alternatively less than 12%, alternatively less than 10%, alternatively less than 9%, alternatively less than 4%, alternatively less than 4%, alternatively less than 4%, alternatively less than 4%, alternatively less than 4%, alternatively less than 3%, alternatively less than 2%, alternatively less than 1%, alternatively less than 0.5%, alternatively less than 0.25%) of any other fatty acids are present.
  • The fatty acid percentage is determined on a weight/weight, mol/mol, or chromatography area percent basis relative to all fatty acids present in the composition as determined by methods such as disclosed in the European Pharmacopeia monograph for omega-3 fatty acid concentrates, European Pharmacopeia monograph for omega-3-acid ethyl esters 90%, or European Pharmacopeia monograph method 2.4.29, USP monograph for fish oil dietary supplements, USP 35 omega-3-acid ethyl esters (LOVAZA®) monograph, or any essentially equivalent methods (whether by gas chromatography, HPLC, FPLC or any other chromatographic method).
  • In some embodiments, the fatty acid percentage is determined not as a percentage of all fatty acids present in the composition but as a specific type of fatty acid ethyl esters as percentage of all fatty acid ethyl esters present in the composition, thus excluding from the fatty acid percentage determination such fatty acids present as, for instance: free fatty acids; mono-, di-, and tri-glycerides; or fatty acids present in phospholipids (such as phosphatidylserine or phosphatidylcholine) or polysorbates (such as Tween 80, Tween 20, or polysorbate 40).
  • In other embodiments, the fatty acid percentage is determined not as a percentage of all fatty acids present in the composition but as a specific type of free fatty acid as percentage of all free fatty acids present in the composition, thus excluding from the fatty acid percentage determination such fatty acids present as, for instance: fatty acid ethyl esters; mono-, di-, and tri-glycerides; or fatty acids present in phospholipids (such as phosphatidylserine or phosphatidylcholine) or polysorbates (such as Tween 80, Tween 20, or polysorbate 40).
  • In yet other embodiments, the fatty acid percentage is determined not as a percentage of all fatty acids present in the composition but as a specific type of glycerol fatty acid ester as percentage of all glycerol fatty acid esters present in the composition, thus excluding from the fatty acid percentage determination such fatty acids present as, for instance: fatty acid ethyl esters; free fatty acids; or fatty acids present in phospholipids (such as phosphatidylserine or phosphatidylcholine) or polysorbates (such as Tween 80, Tween 20, or polysorbate 40).
  • In further embodiments, the fatty acid percentage is determined not as a percentage of all fatty acids present in the composition but as di- or tri-fatty acid esters with glycerol as percentage of all glycerol di- and tri-fatty acid esters present in the composition, thus excluding from the fatty acid percentage determination such fatty acids present as, for instance: glycerol-mono-fatty acid esters; fatty acid ethyl esters; free fatty acids; or fatty acids present in phospholipids (such as phosphatidylserine or phosphatidylcholine) or polysorbates (such as Tween 80, Tween 20, or polysorbate 40).
  • In yet other embodiments, the fatty acid percentage is determined not as a percentage of all fatty acids present in the composition but as a tri-fatty acid esters with glycerol as percentage of all glycerol tri-fatty acid esters present in the composition, thus excluding from the fatty acid percentage determination such fatty acids present as, for instance: mono- and di-fatty acid esters of glycerol; fatty acid ethyl esters; free fatty acids; or fatty acids present in phospholipids (such as phosphatidylserine or phosphatidylcholine) or polysorbates (such as Tween 80, Tween 20, or polysorbate 40).
  • The EPA, HPA, DPA, or omega-3-pentaenoic acids may be derived from any appropriate source including plant seed oils, microbial oils from algae or fungal or marine oils from fish or other marine animals. Certain species are a particular good source of oils containing DPA, for example seal oil. They may be used in the form of the natural oil, if that oil meets the required purity requirements of the present invention, or may be purified to give products containing the fatty acid composition of the present invention.
  • The compositions of the present invention may be produced through a range of the methods. Such methods may include: distillation, including short path distillation; urea precipitation; enzymatic conversion concentration; conventional chromatography; HPLC/FPLC; supercritical carbondioxide extraction; supercritical carbondioxide chromatography; simulated moving bed chromatography; supercritical carbondioxide simulated moving bed chromatography; or chemical conversion methods such as iodolactonization. Such methods are generally known to those skilled in the art of purifying and isolating omega-3 fatty acids.
  • Typically, the omega-3 fatty acid concentration/purification process is initiated by esterifying the fatty acids comprised by the marine oil raw material (such as crude fish oil) with ethanol (to form fatty acid ethyl esters) in order to separate omega-3 fatty acids from other fatty acids covalently bound together in the natural triglyceride molecules of the source oil. Subsequently, the material may be distilled once or several times to achieve omega-3-acid ethyl ester concentrations above 60%-70%. Alternatively, enzymatic concentration, urea precipitation or supercritical extraction may be used alone or in conjunction with distillation to reach omega-3 levels above 70%-90%. In order to prepare a highly pure concentrate of a single omega-3 fatty acid, methods such as chromatography, supercritical chromatography, simulated moving bed chromatography, supercritical simulated moving bed chromatography, or chemical conversion methods such as iodolactolization are typically most practical to reach levels above 50%, alternatively above 60%, alternatively above 70%, alternatively above 80%, alternatively above 90%, alternatively above 95%, of a single omega-3 fatty acid such as ETA, EPA, HPA, DPA, TPA, or DHA.
  • Those skilled in the art will be able to design processes suited to prepare a certain omega-3 fatty acid composition as desired, based on the methods described above. Such processes are flexible enough to affect the relative proportions between the long chain C18, C20, C21 and C22 fatty acids which occur naturally in available fish oil raw materials and other marine oils. It provides not only for the concentration of the individual omega-3 fatty acids, but the ratio between them will remain within a pattern of variation caused by variations in nature. However, suitable methods compensate for sometimes extreme variations which may occur naturally. Thus, for those skilled in the art, it will be possible to make a product with a constant and predetermined composition.
  • EPA is relatively abundant in fish oils or other marine oils and can be relatively easy obtained through the application of concentration and purification technologies from such fish or marine oils. DPA and HPA are present at much lower concentrations. In order to prepare the compositions of the present invention, DPA or HPA may be concentrated and purified from fish or other marine oils according to the methods referred to above, either alone or DPA combined with EPA and/or HPA. Alternatively, the DPA or HPA may be chemically prepared from a high purity EPA concentrate by elongation of the EPA fatty-acid chain with two or one hydrogen-saturated carbons (C2-elongation or C1-elongation) on the carboxyl side of the molecule (for instance with a method similar to or alternate methods with equivalent results such as described by Kuklev DV and Smith WL in Chem Phys Lipids, 2006; 144(2): 172-177). In another alternative approach, a high purity EPA concentrate may be partially converted to DPA (or HPA) using a method for C2-elongation (or C1-elongation) of EPA similar to those described above, thus directly yielding compositions of the present invention or intermediates therefore.
  • Once the oils containing one or more of the desired fatty acids have been obtained, and purified as necessary, these oils may be blended to give the desirable relative amounts of EPA, DPA, HPA, DHA, TPA, other omega-3 fatty acids and omega-6 fatty acids to obtain the compositions of the present invention described in detail above.
  • Fish oils may also contain by-products and contaminants such as pesticides, chlorinated or brominated hydrocarbons, heavy metals, cholesterol and vitamins. During the production of the concentrate, the concentrations of these components are significantly reduced compared to untreated fish oils. Such reduction is inherent due to the nature of purification methods and their ability to concentrate of several or specific omega-3 fatty acids, thus removing other compounds.
  • Triglycerides comprising more than 60% of the omega-3 fatty acids in the composition may be produced from ethyl esters and glycerol by well known, published, or alternative chemical synthetic or enzymatic procedures. The free acids may be produced from ethyl esters by well known hydrolization or saponification procedures. Methods for converting ethyl esters to triglycerides, free fatty acids, and other molecular forms comprising fatty acids, are generally known to those skilled in the art chemically or enzymatically converting omega-3 fatty acids from one form to another.
  • In some embodiments, the compositions of the present invention have improved pharmacological features as demonstrated by improved bioavailability in a mammal of EPA, HPA, DPA, DHA, EPA+DHA, EPA+DPA or EPA+HPA+DPA combined, total omega-3-pentaenoic acids, or of total omega-3 fatty acids. Key parameters for determining bioavailability are maximum concentration of a therapeutic compound or a metabolite thereof (Cmax); the time from administration to maximum concentration (Tmax); and the area under the concentration curve over time (AUC). Such parameters may be determined under single dose or multiple dose administration regimens. Methods to determine comparative bioavailability in mammals are generally known to those skilled in the art.
  • Meal conditions during administration to a subject of omega-3 fatty acid compositions or omega-3 fatty acid formulations can be of special significance for absorption and bioavailability of omega-3 fatty acids. The meal conditions typically considered are: fasting (no food at all prior for 8-12 hours prior to administration and 2-3 hours post administration of the treatment); a low fat meat (a meal typically containing less than 25 gram of fat [350-600 Kcal] consumed just before or after the administration of the treatment; typically within a 15-30 minute range); or a high fat meat (a meal containing 40 gram to 75 gram of fat [700-1000 Kcal] consumed just before or after the administration of the treatment; typically within a 15-30 minute range).
  • In some embodiments of the present invention, compositions of the present invention are more rapidly absorbed as measured by the time to reach the maximum concentration (Tmax) in blood, serum or plasma of EPA, DPA, DHA, EPA+DPA, EPA+DHA, total omega-3-pentaenoic acids, or total omega-3 fatty acids. In preferred embodiments of the present invention, Tmax under high fat meal administration conditions is less than 8 hours, alternatively less than 6 hours, alternatively approximately 5 hours, alternatively 4 hours or less. In other preferred embodiments of the present invention, Tmax under low fat meal administration conditions is less than 8 hours, alternatively less than 6 hours, alternatively approximately 5 hours, alternatively 4 hours or less. In yet other preferred embodiments of the present invention, Tmax under fasting administration conditions is less than 8 hours, alternatively less than 6 hours, alternatively approximately 5 hours, alternatively 4 hours or less.
  • In yet other embodiments of the present invention, Tmax for EPA, DPA, DHA, EPA+DPA, EPA+DHA, or total omega-3 fatty acids are equal or less than Tmax for AMR101 for EPA, DPA, DHA EPA+DPA, EPA+DHA, total omega-3-pentaenoic acids, or total omega-3 fatty acids under high fat meat, low fat meal, and fasting administration conditions. Finally, in other embodiments of the present invention, Tmax for EPA, DPA, DHA, EPA+DPA, EPA+DHA, or total omega-3 fatty acids are less than Tmax for AMR101 for EPA+DHA and to EPA, DPA, DHA, EPA+DPA, EPA+DHA, or total omega-3 fatty acids under either low fat meal, fasting, or both administration conditions.
  • In some embodiments, the improved bioavailability features described above are apparent upon single dose administration, while in other embodiments the improved bioavailability features described above are apparent after multiple dose administration of formulations according to the present invention as compared to referenced comparator products above or substantial equivalent forms thereof.
  • In another embodiment, the compositions of the present invention are more potent and effective than other omega-3 compositions known in the prior art (such as LOVAZA®, EPANOVA™ or VASCEPA®).
  • The formulation may be a single daily dose preparation to give in one dose the above intakes, or may be in convenient divided doses, for example, a daily dose formed of two to four soft gelatin or other dosage forms, each containing 300-1500 mg of EPA, EPA+DPA, EPA+DPA+HPA, or omega-3-pentaenoic acids in any form embodied in the present invention.
  • Flavourants or emulsifiers may be included, for instance, to make the preparation palatable. Other conventional additives, diluents and excipients may be present. The preparation for ingestion may be in the form of a capsule, a dry powder, a tablet, a solution, an oil, an emulsion or any other appropriate form. The capsules may be hard or soft gelatin capsules, agar capsules, or any other appropriate capsule.
  • Use of the formulations of the invention in the manufacture of a medicament for the treatment or prevention of any disease or disorder, including those mentioned above, is included in the present invention.
  • The omega-3 fatty acid composition optionally includes chemical antioxidants, such as alpha tocopherol, which are administered in pure form or suspended in a vegetable oil, such as soybean oil or corn oil.
  • The blended fatty acid compositions may then be incorporated into any appropriate dosage form for oral, enteral, parenteral, rectal, vaginal, dermal or other route of administration. Soft or hard gelatin capsules, flavoured oil blends, emulsifiers or other liquid forms, and microencapsulate powders or other dry form vehicles are all appropriate ways of administering the products.
  • The formulated final drug product containing the omega-3 fatty acid composition may be administered to a mammal or patient in need thereof in a capsule, a tablet, a powder that can be dispersed in a beverage, or another solid oral dosage form, a liquid, a soft gel capsule or other convenient dosage form such as oral liquid in a capsule, as known in the art. In some embodiments, the capsule comprises a hard gelatin. The combination product may also be contained in a liquid suitable for injection or infusion.
  • Example pharmaceutical grade finished dosage forms: (a) Soft or hard gelatin capsules each containing 500 mg or 1000 mg of a mix 20 parts of EPA as a free fatty acid to 1 parts of DPA as a free fatty acid; (b) As in (a) but where the EPA and DPA free fatty acids are replaced with the fatty acids in any other appropriate bioassimilable form such as the ethyl esters; (c) As in (a)-(b) but where the material is in the form of a microencapsulated powder which can be used as a powder or compressed into tablets. Such powders may be prepared by a variety of technologies known to those skilled in the art; (d) As in (a)-(b) but where the formulation is a liquid or emulsion, appropriately flavoured for palatable oral administration; (e) As in (a)-(b) but where the material is formulated into a pharmaceutically acceptable vehicle appropriate for topical application such as a cream or ointment.
  • The omega-3 compositions of the present invention may also be administered with a combination of one or more non-active pharmaceutical ingredients (also known generally herein as “excipients”). Non-active ingredients, for example, serve to solubilize, suspend, thicken, dilute, emulsify, stabilize, preserve, protect, color, flavor, and fashion the active ingredients into an applicable and efficacious preparation that is safe, convenient, and otherwise acceptable for use. Thus, the non-active ingredients may include colloidal silicon dioxide, crospovidone, lactose monohydrate, lecithin, microcrystalline cellulose, polyvinyl alcohol, povidone, sodium lauryl sulfate, sodium stearyl fumarate, talc, titanium dioxide and xanthum gum.
  • The term “pharmaceutically acceptable vehicle,” as used herein, includes any of the following: a solution where the first API and optional other ingredients are wholly dissolved in a solubilizer (e.g., a pharmaceutically acceptable solvent or mixture of solvents), wherein the solution remains in clear liquid form at about room temperature; a suspension; an oil; or a semi-solid, wherein the first API and optionally other ingredients are dissolved wholly or partially in a solubilizer (an emulsion, cream, etc.).
  • A “pharmaceutical grade finished dosage form” as used herein may be construed as a unit dose form suitable for administration to, for example, human or animal subjects, and having content uniformity acceptable to regulatory authorities. For example, under the USP requirements for content uniformity, a pharmaceutical grade finished dosage form should have an amount of API within the range of 85% to 115% of the desired dosage and an RSD less than or equal to 6.0%. In addition, a pharmaceutical grade finished dosage form must be stable (i.e., have a “shelf life”) for a pharmaceutically acceptable duration of time, preferably at least six months, alternatively at least one year, or at least two years, when stored at room temperature (about 23 degree Celcius to 27 degree Celcius, preferably about 25 degree Celcius) and 60% relative humidity. Typically, stability is determined by physical appearance and/or chemical modification of the ingredients, in accordance with standards well-known in the pharmaceutical arts, including those documented in ICH guidelines.
  • The omega-3 fatty acid dosage form optionally includes chemical antioxidants, such as alpha tocopherol, oils, such as soybean oil and partially hydrogenated vegetable oil, and lubricants such as fractionated coconut oil, lecithin and a mixture of the same.
  • The compositions of the present invention may be used for the treatment of patients by administering an effective amount of such compositions to a subject in need thereof, such as a subject prone to or afflicted with a disease or condition or in need of treatment for a disease or condition. The present invention provides methods of treating, preventing, and reducing the symptoms, pathology or events associated with a disease or condition comprising administration of any of the compositions of the present invention. The present invention provides methods of treating, preventing, reducing the occurrence of, and reducing symptoms associated with coronary heart disease (CHD), vascular disease, atherosclerotic disease, or related conditions. The present invention also provides methods of treating, preventing, reducing the occurrence of, and reducing the symptoms, pathology or events associated with vascular-related diseases or conditions. Examples of vascular diseases and conditions include, but are not limited to: atherosclerosis, atherosclerosis associated with hypercholesterolemia, hypertriglyceridemia or mixed dyslipidemia, coronary heart disease (CHD), vascular disease, peripheral artery disease (PAD), heart failure, cardiac arrhythmias, blood coagulatory conditions associated with cardiac arrhythmias, hypertension, coagulation related disorders (including but not limited to post-surgical deep vein thrombosis or other high risk thrombosis conditions), nephropathy and other kidney diseases, renal impairment, retinopathy, cognitive impairment, dementia (including but not limited to ischemic dementia and vascular dementia), and metabolic syndrome. The compositions of the present invention may be used for the treatment of patients by administering an effective amount of such compositions to a subject or a subject in need thereof, such as a subject prone to or afflicted with a vascular/cardiovascular disease or condition, a subject at risk of vascular/cardiovascular events, or a subject in need of treatment for a vascular/cardiovascular disease or condition.
  • Determination of such cardiovascular diseases/conditions and prevention of events/symptoms in mammals and methods to determine treatment and preventative/therapeutic effects therefore are generally know to those skilled in the art.
  • Administration to or treatment of subject with the compositions of the present inventions results in significant clinical improvements, including but not limited to the following. In some embodiments, the present invention provides for the prevention of death (including all cause mortality, cardiovascular mortality, and cardiac death); the treatment and/or prevention of cardiac events, cardiovascular events and/or vascular events and/or symptoms. In some embodiments, the present invention also provides for the reduction of number or frequency of such events, as well as a reduction or amelioration of symptoms associated with such events. Vascular, cardiovascular and/or cardiac events may include, but are not limited to: death due to cardiovascular event (including cardiovascular death and cardiac death), sudden cardiac death, myocardial infarction (fatal or non-fatal), ischemic cardiac attack, ischemic attack, angina, stroke, transient ischemic cerebral attack, thrombosis, deep vein thrombosis, pulmonary embolism, coronary revascularization, coronary revascularization with stent placement, carotid artery revascularization, carotid artery revascularization with stent placement, peripheral artery revascularization, peripheral artery revascularization with stent placement, major coronary event, coronary artery bypass grafting, other blood vessel grafting, treatment of aneurism, plaque rupture, and hospitalization due to cardiovascular event (including angina, acute angina and unstable angina). Cardiovascular and/or cardiac events may also include other events deemed to fall in such category by those skilled in the art. Study endpoints may comprise individual events or a composite of several of the events described directly above (for the latter the term Major Adverse Cardiovascular Event or MACE is often used).
  • In some embodiments, the present invention provides methods of retarding the atherosclerotic disease process or improves the vascular healing process in response to presence of atherogenic disease. Such retardation or healing may be demonstrated by reduced stenosis and/or restenosis at specific vascular sites over time, reduced or lesser increase in intima-media thickness (IMT) of the arterial wall over time, larger lumen size and/or larger vascular diameter at vascular sites with stenosis or clot built-up over time. Such retardation or healing may be determined by intravascular ultrasound (IVUS), radiographic, echocardiographic, radiologic, non-invasive ultrasound, tomography, magnetic resonance interference (MRI), or other acceptable methods. In other embodiments, such retardation or improved healing may be demonstrated by the vascular wall composition, such as a reduced foam cell presence or fibrillated tissue in the vessel wall. In yet other embodiments, such improved vascular healing is demonstrated by improved inflammatory markers in the vascular wall or specific types of staining of vascular tissue.
  • In some embodiments, the compositions of the present invention may differentially alter the ratio between blood platelets and fragments thereof (also known as platelet microparticles). Such fragments may be evaluated as a whole or examined and described as fragment sub-categories.
  • In some embodiments, the compositions of the present invention may differentially alter the surface charge of blood platelets and fragments thereof, either in resting state (non-activated platelets) or activated stage.
  • In some embodiments, the compositions of the present invention may affect the coagulatory cascade and differentially alter coagulation or bleeding times or platelet aggregation times and density.
  • The compositions of the present invention may be useful for any subjects and they may be especially useful in certain patient populations, including but not limited to the following: subjects having receiving treatment for or in need of treatment for hypertriglyceridemia, hypercholesterolemia, Frederickson dyslipidemia, and other comorbidities. The present invention provides for methods of treatment of subjects with no previous cardiovascular events, or subjects with one or more previous cardiovascular events. In some embodiments, the subjects to be treated with or administered the compositions of the present invention, are selected based on their dyslipidemic profile. Subjects may be selected for treatment/administration from the following categories: hypertriglyceridemia (such as TG≧750 mg/dL, TG≧500 mg/dL, TG≧200 mg/dL, TG≧150 mg/dL, TG500-2000 mg/dL, TG500-1500 mg/dL, TG750-2000 mg/dL, TG750-1500 mg/dL, TG200-499 mg/dL, TG300-499 mg/dL, TG350-499 mg/dL, TG300-750 mg/dL, TG300-700 mg/dL, TG200-499 mg/dL, or TG150-199 mg/dL); hypercholesterolemia (such as LDL-C<70 mg/dL, LDL-C≧70 mg/dL, LDL-C≧100 mg/dL, LDL-C≧130/dL, LDL-C≧160 mg/dL, LDL-C190 mg/dL, LDL-C 70-100 mg/dL, LDL-C 100-130 mg/dL, LDL-C 130-160 mg/dL, LDL-C 160-190 mg/dL, LDL-C 130-190 mg/dL, LDL-C 100-160 mg/dL, LDL-C 70-130 mg/dL, LDL-C 70-160 mg/dL, LDL-C 100-190 mg/dL, LDL-C 70-190 mg/dL, NON-HDL-C<100 mg/dL, NON-HDL-C≧100 mg/dL, NON-HDL-C≧130 mg/dL, NON-HDL-C≧160/dL, NON-HDL-C≧190 mg/dL, NON-HDL-C 100-130 mg/dL, NON-HDL-C 130-160 mg/dL, NON-HDL-C 160-190 mg/dL, NON-HDL-C 130-190 mg/dL, NON-HDL-C 100-160 mg/dL, or NON-HDL-C 100-190 mg/dL); or Frederickson dyslipidemia type (either, Type I, Type IIa, Type IIb, Type III, Type IV, or Type V). In some embodiments, the subjects to be treated with or administered the compositions of the present invention, are selected based on the presence of certain comorbidities. Such comorbidities may include, but are not limited to the following: renal disease, nephropathy, IgA nephropathy, renal impairment, renal failure (also kidney failure or renal insufficiency), renal insufficiency requiring dialysis, renal insufficiency without dialysis treatment, chronic analgesic nephritis, polycystic kidney disease, proteinuria, hypertension, thrombotic microangiopathy, renal failure (acute renal failure, chronic renal failure), uremic pericarditis, uremia, renal artery stenosis, renal ischemia, hypertensive nephropathy, renovascular hypertension, renal osteodystrophy, nephroptosis, renal cortical necrosis, glomerulitis, metabolic syndrome, diabetes, or pre-diabetes. In some embodiments, the methods of administering the compositions of the present invention are useful in subjects having cardiopathy, coronary ischemia, cardiac decompensation, or diabetic pathology with cardiopathy, and subjects with previous myocardial infarction, stroke, recent myocardial infarction (post-MI), Acute Coronary Syndrome (ACS), ACS without ST-segment elevation (NSTEMI), ACS with ST-segment elevation (STEMI), or any other major cardiovascular event. In some embodiments, the compositions of the present invention may reduce the reoccurrence of such cardiovascular events.
  • In some embodiments, treatment with the compositions of the present invention results in clinical improvement of such comorbidities. In some embodiments, administration with the compositions of the present invention may reduce the time necessary to achieve clinical improvement and/or attain treatment goals. In some embodiments, the administration of compositions can result in clinical improvements in clinical markers, including, but not limited to: glomerular filtration rate, serum creatinine, serum urea, serum phosphate levels, serum potassium levels, systolic or diastolic blood pressure, progression to dialysis, progression to renal failure, progression to need for renal transplant, fasting serum glucose levels, postprandial serum glucose levels, serum fructosamine levels, therapeutic insulin utilization, progression to diabetes, hypo-glucose events, hyper-glucose events, or hemoglobin A1C.
  • The compositions of the present invention may be co-administered with one or more other therapeutic agents. In some embodiments, clinical benefits resulting from the administration or treatment of subjects with the compositions of the present invention may be improved with concomitant use or in combination with other therapeutic agents. Examples of such concomitant or fixed combination treatments may include coadministration with one or more of the following: an HMG-CoA reductase inhibitor (“statin”), an anti-platelet agent (such as aspirin or clopidogrel), an anti-coagulant (such as warfarin) an antihypertensive (such as a diuretic, beta-blocker, calcium channel blocker, ACE-inhibitor, angiotensin II receptor (ARB) antagonist), or other treatments for cardiovascular diseases.
  • The present invention also provides pharmaceutical compositions, for example, a unit dosage, comprising one or more HMG-CoA reductase inhibitors (“statins”) and an omega-3 fatty acid composition of the present invention. In some embodiments, the present invention provides a fixed dose combination product comprising both a statin and the composition of the present invention. The present invention may incorporate currently known or future known statins in an amount generally recognized as safe and effective. There are currently seven statins that are widely available: atorvastatin, rosuvastatin, fluvastatin, lovastatin, pravastatin, pitavastatin, and simvastatin. An eighth statin, cerivastatin, has been removed from the U.S. market at the time of this writing. However, it is conceivable to one skilled in the art that cerivastatin may be used in conjunction with some embodiments of the present invention if cerivastatin is ultimately determined to be safe and effective in certain treatment regimens. Such statins are typically used at their common daily doses, which include, but are not limited to lovastatin 10 mg, 20 mg, 40 mg; pravastatin 10 mg, 20 mg, 40 mg, 80 mg; simvastatin 5 mg, 10 mg, 20 mg, 40 mg, 80 mg, fluvastatin 20 mg, 40 mg, 80 mg; atorvastatin 10 mg, 20 mg, 40 mg, 80 mg; rosuvastatin 5 mg, 10 mg, 20 mg, 40 mg; and pitavastatin 1 mg, 2 mg, 4 mg, 8 mg. Generally, the effect of statins is dose dependent, i.e., the higher the dose, the greater the therapeutic affect. However, the effect of each statin is different, and therefore the level of therapeutic effect of one statin cannot be necessarily be directly correlated to the level of therapeutic effects of other statins. For example, bioavailability varies widely among the statins. Specifically, it has been shown that simvastatin is less than 5% bioavailable, while fluvastatin is approximately 24% bioavailable. Statins are absorbed at rates ranging from about 30% with lovastatin to 98% with fluvastatin. First-pass metabolism occurs in all statins except pravastatin. Pravastatin is also the least protein-bound of the statins (about 50%), compared with the others, which are more than 90% protein-bound. Accordingly, the statins possess distinct properties from one another. The combination products of this invention involving each statin or a plurality of statins are also distinct. The method of treatment may combine the administration of one or more statins at its common dose or an alternative dose with the composition of the present invention
  • EXAMPLES Example 1
  • A composition according to the present invention is prepared by mixing and homogenizing in a ratio of 98:2 the intermediates MEGAPEX E90D00EE (90% EPA ethyl ester,) and MAXOMEGA DPA95 FFA (≧95% DPA synthetic fatty acid produced from EPA ethyl ester concentrate) converted to ethyl ester, respectively. These intermediates were prepared and commercially offered for sale by Chemport Korea (MEGAPEX) and Equateq Ltd from Scotland, UK (MAXOMEGA). The relative amounts of fatty acids present in the starting intermediates and in the resulting novel composition are listed in Table 1 below. The resulting novel composition comprises 89.10% EPA, 1.95% DPA, 0.19% HPA, 91.24% omega-3-pentaenoic acids, less than 0.01% DHA, 91.24% omega-3-pentaenoic acids, 93.09% total omega-3 fatty acids, 3.15% ARA and 3.57% omega-6 fatty acids (all Area %).
  • TABLE 1
    Fatty acid Composition (Area %) of intermediates and
    novel composition according to Example 1
    98.0% 2.0%
    Megapex Maxomega Novel
    Fatty Acid E90D00EE DPA95FFA => EE Composition
    c18:0 0.05 0 0.05
    c18:1n9 0.06 0 0.06
    c18:1n7 0.02 0 0.02
    c18:2n6 0.01 0 0.01
    c18:3n6 0.02 0 0.02
    c18:3n3 0.03 0 0.03
    c18:4n3 0.42 0 0.41
    c18:4n1 0.07 0 0.07
    c20:0 0 0 0.00
    c20:1n11 0 0 0.00
    c20:1n9 0 0 0.00
    c20:1n7 0 0 0.00
    c20:2n6 0.25 0 0.25
    c20:3n9 0 0 0.00
    c20:3n6 0.15 0 0.15
    c21:0 0 0 0.00
    c20:4n6 3.21 0 3.15
    c20:3n3 0 0 0.00
    c20:4n3 1.44 0 1.41
    c20:5n3 90.92 0 89.10
    c22:0 0.3 0 0.29
    c22:1n11 0.07 0 0.07
    c22:1n9 0.18 0 0.18
    c22:1n7 0.19 0 0.19
    c21:5n3 0.19 0 0.19
    c22:5n6 0 0 0.00
    c22:5n3 0 97.27 1.95
    c22:6n3 0 0 0.00
    c24:0 0 0.33 0.01
    OTHER 2.42 2.4 2.42
    100 100 100
  • Example 2
  • A composition according to the present prevention is prepared by mixing and homogenizing in a ratio of 96:4 the intermediates MEGAPEX E90D00EE (90% EPA ethyl ester,) and MAXOMEGA DPA95 FFA (95% DPA synthetic fatty acid produced from EPA ethyl ester concentrate), converted to ethyl ester, respectively. These intermediates were prepared and commercially offered for sale by Chemport Korea (MEGAPEX) and Equateq Ltd from Scotland, UK (MAXOMEGA). The relative amounts of fatty acids present in the starting intermediates and in the resulting novel composition is listed in Table 2 below. The resulting novel composition comprises 87.28% EPA, 3.89% DPA, 0.18% HPA, 91.35% omega-3-pentaenoic acids, less than 0.01% DHA, 93.17% total omega-3 fatty acids and 3.49% omega-6 fatty acids (all Area %).
  • TABLE 2
    Fatty acid Composition (Area %) of intermediates and
    novel composition according to Example 2
    96.0% 4.0%
    Megapex Maxomega Novel
    Fatty Acid E90D00EE DPA95FFA => EE Composition
    c18:0 0.05 0 0.05
    c18:1n9 0.06 0 0.06
    c18:1n7 0.02 0 0.02
    c18:2n6 0.01 0 0.01
    c18:3n6 0.02 0 0.02
    c18:3n3 0.03 0 0.03
    c18:4n3 0.42 0 0.40
    c18:4n1 0.07 0 0.07
    c20:0 0 0 0.00
    c20:1n11 0 0 0.00
    c20:1n9 0 0 0.00
    c20:1n7 0 0 0.00
    c20:2n6 0.25 0 0.24
    c20:3n9 0 0 0.00
    c20:3n6 0.15 0 0.14
    c21:0 0 0 0.00
    c20:4n6 3.21 0 3.08
    c20:3n3 0 0 0.00
    c20:4n3 1.44 0 1.38
    c20:5n3 90.92 0 87.28
    c22:0 0.3 0 0.29
    c22:1n11 0.07 0 0.07
    c22:1n9 0.18 0 0.17
    c22:1n7 0.19 0 0.18
    c21:5n3 0.19 0 0.18
    c22:5n6 0 0 0.00
    c22:5n3 0 97.27 3.89
    c22:6n3 0 0 0.00
    c24:0 0 0.33 0.01
    OTHER 2.42 2.4 2.42
    100 100 100
  • Example 3
  • A composition according to the present invention is prepared by mixing and homogenizing in a ratio of 94:6 the intermediates MEGAPEX E90D00EE (90% EPA ethyl ester,) and MAXOMEGA DPA95 FFA (≧95% DPA synthetic fatty acid produced from EPA ethyl ester concentrate) converted to ethyl ester, respectively. These intermediates were prepared and commercially offered for sale by Chemport Korea (MEGAPEX) and Equateq Ltd from Scotland, UK (MAXOMEGA). The relative amounts of fatty acids present in the starting intermediates and in the resulting novel composition are listed in table 3 below. The resulting novel composition comprises 85.46% EPA, 5.84% DPA, 0.18% HPA, 91.48% omega-3-pentaenoic acids, less than 0.01% DHA, 93.26% total omega-3 fatty acids, 3.02% ARA, and 3.42% omega-6 fatty acids (all Area %).
  • TABLE 3
    Fatty acid Composition (Area %) of intermediates and
    novel composition according to Example 3
    94.0% 6.0%
    Megapex Maxomega Novel
    Fatty Acid E90D00EE DPA95FFA => EE Composition
    c18:0 0.05 0 0.05
    c18:1n9 0.06 0 0.06
    c18:1n7 0.02 0 0.02
    c18:2n6 0.01 0 0.01
    c18:3n6 0.02 0 0.02
    c18:3n3 0.03 0 0.03
    c18:4n3 0.42 0 0.39
    c18:4n1 0.07 0 0.07
    c20:0 0 0 0.00
    c20:1n11 0 0 0.00
    c20:1n9 0 0 0.00
    c20:1n7 0 0 0.00
    c20:2n6 0.25 0 0.24
    c20:3n9 0 0 0.00
    c20:3n6 0.15 0 0.14
    c21:0 0 0 0.00
    c20:4n6 3.21 0 3.02
    c20:3n3 0 0 0.00
    c20:4n3 1.44 0 1.35
    c20:5n3 90.92 0 85.46
    c22:0 0.3 0 0.28
    c22:1n11 0.07 0 0.07
    c22:1n9 0.18 0 0.17
    c22:1n7 0.19 0 0.18
    c21:5n3 0.19 0 0.18
    c22:5n6 0 0 0.00
    c22:5n3 0 97.27 5.84
    c22:6n3 0 0 0.00
    c24:0 0 0.33 0.02
    OTHER 2.42 2.4 2.42
    100 100 100
  • Example 4
  • A composition according to the present invention is prepared by mixing and homogenizing in a ratio of 75:25 the intermediates MEGAPEX E90D00EE (90% EPA ethyl ester,) and MAXOMEGA DPA95 FFA (≧95% DPA synthetic fatty acid produced from EPA ethyl ester concentrate, converted to ethyl ester, respectively. These intermediates were prepared and commercially offered for sale by Chemport Korea (MEGAPEX) and Equateq Ltd from Scotland, UK (MAXOMEGA). The relative amounts of fatty acids present in the starting intermediates and in the resulting novel composition is listed in table 4 below. The resulting novel composition comprises 68.10% EPA, 24.32% DPA, 0.19% HPA, 92.65% omega-3-pentaenoic acids, less than 0.01% DHA, 94.07% total omega-3 fatty acids, 2.41% ARA and 2.73% omega-6 fatty acids (all Area %).
  • TABLE 4
    Fatty acid Composition (Area %) of intermediates and
    novel composition according to Example 4
    75.0% 25.0%
    Megapex Maxomega Novel
    Fatty Acid E90D00EE DPA95FFA => EE Composition
    c18:0 0.05 0 0.04
    c18:1n9 0.06 0 0.05
    c18:1n7 0.02 0 0.02
    c18:2n6 0.01 0 0.01
    c18:3n6 0.02 0 0.02
    c18:3n3 0.03 0 0.02
    c18:4n3 0.42 0 0.32
    c18:4n1 0.07 0 0.05
    c20:0 0 0 0.00
    c20:1n11 0 0 0.00
    c20:1n9 0 0 0.00
    c20:1n7 0 0 0.00
    c20:2n6 0.25 0 0.19
    c20:3n9 0 0 0.00
    c20:3n6 0.15 0 0.11
    c21:0 0 0 0.00
    c20:4n6 3.21 0 2.41
    c20:3n3 0 0 0.00
    c20:4n3 1.44 0 1.08
    c20:5n3 90.92 0 68.19
    c22:0 0.3 0 0.23
    c22:1n11 0.07 0 0.05
    c22:1n9 0.18 0 0.14
    c22:1n7 0.19 0 0.14
    c21:5n3 0.19 0 0.14
    c22:5n6 0 0 0.00
    c22:5n3 0 97.27 24.32
    c22:6n3 0 0 0.00
    c24:0 0 0.33 0.08
    OTHER 2.42 2.4 2.42
    100 100 100
  • Example 5
  • A composition according to the present invention is prepared by mixing and homogenizing in a ratio of 60:40 the intermediates KD-PharmaKD-PUR 900EE and MAXOMEGA DPA95 FFA converted to ethyl ester, respectively. These intermediates were prepared and commercially offered for sale by KD-Pharma Germany (KD-Pharma) and Equateq Ltd from Scotland, UK (MAXOMEGA). The relative amounts of fatty acids present in the starting intermediates and in the resulting novel composition is listed in table 5 below. The resulting novel composition comprises 55.74% EPA, 39.26% DPA, 2.39% HPA, 97.44% omega-3-pentaenoic acids, and 98.06% total omega-3 fatty acids (all Area %).
  • TABLE 5
    Fatty acid Composition (Area %) of intermediates and
    novel composition according to Example 5
    40.0%
    60.0% Maxomega Novel
    Fatty Acid KD-Pur 900EE DPA95FFA => EE Composition
    c18:0 0 0 0.00
    c18:1n9 0 0 0.00
    c18:1n7 0 0 0.00
    c18:2n6 0 0 0.00
    c18:3n6 0 0 0.00
    c18:3n3 0 0 0.00
    c18:4n3 0 0 0.00
    c18:4n1 0 0 0.00
    c20:0 0 0 0.00
    c20:1n11 0 0 0.00
    c20:1n9 0 0 0.00
    c20:1n7 0 0 0.00
    c20:2n6 0 0 0.00
    c20:3n9 0 0 0.00
    c20:3n6 0 0 0.00
    c21:0 0 0 0.00
    c20:4n6 0 0 0.00
    c20:3n3 0 0 0.00
    c20:4n3 1.04 0 0.62
    c20:5n3 92.99 0 55.79
    c22:0 0 0 0.00
    c22:1n11 0 0 0.00
    c22:1n9 0 0 0.00
    c22:1n7 0 0 0.00
    c21:5n3 3.98 0 2.39
    c22:5n6 0 0 0.00
    c22:5n3 0.58 97.27 39.26
    c22:6n3 0 0 0.00
    c24:0 0 0.33 0.13
    OTHER 1.41 2.4 1.81
    100.00 100 100.00
  • Example 6
  • A composition according to the present invention is prepared by mixing and homogenizing in a ratio of 96:4 the intermediates KD-PUR 900EE KD-Pharma and MAXOMEGA DPA95 FFA converted to ethyl ester, respectively. These intermediates were prepared and commercially offered for sale by KD-Pharma Germany (KD-Pharma) and Equateq Ltd from Scotland, UK (MAXOMEGA). The relative amounts of fatty acids present in the starting intermediates and in the resulting novel composition is listed in table 6 below. The resulting novel composition comprises 89.27% EPA, 4.45% DPA, 3.82% HPA, 97.54% omega-3-pentaenoic acids, and 98.54% total omega-3 fatty acids (all Area %).
  • TABLE 6
    Fatty acid Composition (Area %) of intermediates and
    novel composition according to Example 6
    4.0%
    96.0% Maxomega Novel
    Fatty Acid KD-Pur 900EE DPA95FFA => EE Composition
    c18:0 0 0 0.00
    c18:1n9 0 0 0.00
    c18:1n7 0 0 0.00
    c18:2n6 0 0 0.00
    c18:3n6 0 0 0.00
    c18:3n3 0 0 0.00
    c18:4n3 0 0 0.00
    c18:4n1 0 0 0.00
    c20:0 0 0 0.00
    c20:1n11 0 0 0.00
    c20:1n9 0 0 0.00
    c20:1n7 0 0 0.00
    c20:2n6 0 0 0.00
    c20:3n9 0 0 0.00
    c20:3n6 0 0 0.00
    c21:0 0 0 0.00
    c20:4n6 0 0 0.00
    c20:3n3 0 0 0.00
    c20:4n3 1.04 0 1.00
    c20:5n3 92.99 0 89.27
    c22:0 0 0 0.00
    c22:1n11 0 0 0.00
    c22:1n9 0 0 0.00
    c22:1n 7 0 0 0.00
    c21:5n3 3.98 0 3.82
    c22:5n6 0 0 0.00
    c22:5n3 0.58 97.27 4.45
    c22:6n3 0 0 0.00
    c24:0 0 0.33 0.01
    OTHER 1.41 2.4 1.45
    100.00 100 100.00
  • Example 7
  • A composition according to the present invention is prepared by mixing and homogenizing in a ratio of 91.8:8.2 the intermediates KD-PUR 910EE KD-Pharma and DPA95 FFA converted to ethyl ester, respectively. The relative amounts of fatty acids present in the starting intermediates and in the resulting novel composition is listed in table 7 below.
  • TABLE 7
    Fatty acid Composition (Area %) of intermediates and
    novel composition according to Example 7
    91.8% 8.2%
    KD-Pur DPA- Novel
    Fatty Acid EPA910EE 95% Est composition N6 N3
    c18:0 0 0 0.00
    c18:1n9 0 0 0.00
    c18:1n7 0 0 0.00
    c18:2n6 0 0 0.00 0.00
    c18:3n6 0 0 0.00 0.00
    c18:3n3 0 0 0.00 0.00
    c18:4n3 0 0 0.00 0.00
    c18:4n1 0 0 0.00
    c20:0 0 0 0.00
    c20:1n11 0.1 0 0.09
    c20:1n9 0 0 0.00
    c20:1n7 0 0 0.00
    c20:2n6 0 0 0.00 0.00
    c20:2n9 0 0.2 0.02
    c20:3n9 0 0 0.00
    c20:3n6 0 0 0.00 0.00
    c21:0 0 0 0.00
    c20:4n6 0.3 0 0.28 0.28
    c20:3n3 0 0 0.00 0.00
    c20:4n3 1.2 0.3 1.13 1.13
    c20:5n3 92.5 0 85.34 85.34
    c22:0 0.2 0 0.18
    c22:1n11 0 0 0.00
    c22:1n9 0 0 0.00
    c22:1n7 0 0 0.00
    c22:4n3 0 1.9 0.16 0.16
    c21:5n3 3.3 0.1 3.08 3.08
    c22:5n6 0 0 0.00 0.00
    c22:5n3 0.2 97 8.16 8.16
    c22:6n3 1.5 0 1.25 1.25
    c24:0 0 0 0.00
    OTHER 0.7 0.5 0.68
    100.00 100 100.36 0.28 99.11
  • Example 8
  • The ethyl ester composition of Example 4 may be converted into a free fatty acid composition with essentially the same fatty acid composition according to “Conversion Method EE to FFA” below. This method is indiscriminate with respect to the type, degree of saturation or length of fatty acid if performed for an adequate amount of time under the described conditions.
  • Conversion Method EE to FFA
      • 1. Fatty Acid Ethyl Ester (FAEE GMP, approx. 3 mmol/g) oil is brought into a closed heated/cooled reaction chamber under nitrogen atmosphere (preferably with pressure control), and heated to 50-60 degree Celcius under stirring.
      • 2. 2M NaOH solution in water is added under firm stirring to ensure phase mixing (est. 2-3×FAEE w/w) and stir until no ethyl ester is presence (est. 2-4 hrs). Test ethyl ester presence at lab scale/in process with TLC (hexanes/EtOAc 9:1) and with EP GC method to confirm reaction completion under GMP.
      • 3. Under cooling (keep mixture below 70 degree Celcius), add 6M HCl in water (est. <1 hr) until slightly acid (˜pH3-4). It may be necessary to control pressure to prevent excessive foaming. Then halt stirring, give time to let phases separate, and remove water phase from bottom (keep oil protected from oxygen, apply nitrogen atmosphere blanket).
      • 4. Add demineralized water (est. 2-3×FAEE w/w) and wash out NaCl and ethanol from oil under firm stirring (est. ˜1 hr). Halt stirring, give time to let phases separate, and remove water phase from bottom (keep oil protected from oxygen, apply nitrogen atmosphere blanket).
      • 5. Repeat Step 4 several times (˜2×) to remove ethanol and NaCl.
      • 6. Remove water and remaining ethanol [determine in-process controls], confirm under GMP with USP residual solvent method (target: ethanol <100 ppm) by stirring oil while applying vacuum 10-50 mbar (with solvent trap) and heat oil (70-80 degree celcius) until water/ethanol target is met (est. 2-4 hrs).
      • 7. Add anti-oxidants (i.e. alpha-D-tocopherol, USP, target 4 mg/g) and/or other excipients.
      • 8. All reagents and excipients USP grade.
    Example 9
  • The ethyl ester composition of Example 3 is converted into a free fatty acid composition with essentially the same fatty acid composition according to “Conversion Method EE to FFA” above. This method is indiscriminate with respect to the type, degree of saturation or length of fatty acid if performed for an adequate amount of time under the described conditions.
  • Example 10
  • The ethyl ester composition of Example 6 is converted into a free fatty acid composition with essentially the same fatty acid composition according to “Conversion Method EE to FFA” above. This method is indiscriminate with respect to the type, degree of saturation or length of fatty acid if performed for an adequate amount of time under the described conditions.
  • Example 11
  • The composition of Example 4 is formulated into a soft gelatin capsule. Prior to encapsulation, an anti-oxidant preparation (composed of 4000 mg alpha-D-tocopherol in one liter of corn oil; corn oil is a triglyceride low in omega-3) is added to the composition of Example 4, by mixing and homogenizing 100 mL of this anti-oxidant preparation into 100 liters of the oil composition of Example 4 followed by thorough homogenization. The resulting pre-encapsulation formulated oil contains approximately 4 mg/gram alpha-D-tocopherol. Subsequently, the formulated oil is encapsulated into soft gelatin capsules with printed logo according to general methods typically used by Accucaps in Canada for fish oils or by any other documented and operational encapsulation method. The fill mass of the oil is approximately 1.08 gram/capsule, providing a dose of approximately 1000 mg omega-3-pentaenoic-acids ethyl esters per capsule. Finally, the capsules are bottled in HDPE bottles with induction seal and child resistant cap.
  • Example 12
  • The composition of Example 9 is formulated into a soft gelatin capsule. Prior to encapsulation, an anti-oxidant preparation (composed of 4000 mg alpha-D-tocopherol in one liter of corn oil; corn oil is a triglyceride low in omega-3) is added to the composition of Example 4, by mixing and homogenizing 100 mL of this anti-oxidant preparation into 100 liters of the oil composition of Example 4 followed by thorough homogenization. The resulting pre-encapsulation formulated oil contains approximately 4 mg/gram alpha-D-tocopherol. Subsequently, the formulated oil is encapsulated into soft gelatin capsules with printed logo according to general methods typically used by Banner in High Point, N.C., for fish oils or by any other documented and operational encapsulation method. The fill mass of the oil is approximately 1.09 gram/capsule, providing a dose of approximately 1000 mg omega-3-pentaenoic-acids per capsule. Finally, the capsules are bottled in HDPE bottles with induction seal and child resistant cap.
  • Example 13
  • The composition of Example 5 is formulated into a soft gelatin capsule. Prior to encapsulation, an anti-oxidant preparation (composed of 4000 mg alpha-D-tocopherol in one liter of corn oil; corn oil is a triglyceride low in omega-3) is added to the composition of Example 4, by mixing and homogenizing 100 mL of this anti-oxidant preparation into 100 liters of the oil composition of Example 4 followed by thorough homogenization. The resulting pre-encapsulation formulated oil contains approximately 4 mg/gram alpha-D-tocopherol. Subsequently, the formulated oil is encapsulated into soft gelatin capsules with printed logo according to general methods typically used by Catalent in St. Petersburg, Fla., for fish oils or by any other documented and operational encapsulation method. The fill mass of the oil is approximately 1.05 gram/capsule, providing a dose of approximately 1000 mg omega-3-pentaenoic-acids ethyl esters per capsule. Finally, the capsules are bottled in HDPE bottles with induction seal and child resistant cap.
  • Example 14
  • The composition of Example 10 is formulated into a soft gelatin capsule. Prior to encapsulation, an anti-oxidant preparation (composed of 4000 mg alpha-D-tocopherol in one liter of corn oil; corn oil is a triglyceride low in omega-3) is added to the composition of Example 4, by mixing and homogenizing 100 mL of this anti-oxidant preparation into 100 liters of the oil composition of Example 4 followed by thorough homogenization. The resulting pre-encapsulation formulated oil contains approximately 4 mg/gram alpha-D-tocopherol. Subsequently, the formulated oil is encapsulated into soft gelatin capsules with printed logo according to general methods typically used by Banner in High Point, N.C., for fish oils or by any other documented and operational encapsulation method. The fill mass of the oil is 1.06 gram/capsule, providing a dose of approximately 1000 mg omega-3-pentaenoic-acids per capsule. Finally, the capsules are bottled in HDPE bottles with induction seal and child resistant cap.
  • Example 15
  • A patient is diagnosed with vascular disease. Thereupon, the patient may be initiated on daily treatment with one of the encapsulated compositions according to Examples 10, 11, 12 or 13. Four capsules per day are administered to this patient (4 g/d).
  • Example 16
  • A patient is treated as per Example 15. The treatment results in significant reduction of the occurrence of MACE.
  • Example 17
  • The following are examples of preferred embodiments of the present invention.
  • COMPOSITION 1a
    Minimum Maximum Target
    Composition (mg/g) (mg/g) (mg/g)
    Omega-3 pentaenoic acid 880 980 930
    Eicosapentaenoic acid (EPA) 800 950 850
    Heneicosapentaenoic acid (HPA) 5 60 30
    Docosapentaenoic acid (DPA) 60 100 80
    Docosahexaenoic acid (DHA) 25 <10
  • COMPOSITION 1b
    Minimum Maximum Target
    Composition (mg/g) (mg/g) (mg/g)
    Omega-3 pentaenoic acid 870 990 920
    Eicosapentaenoic acid (EPA) 750 950 830
    Heneicosapentaenoic acid (HPA) 5 70 40
    Docosapentaenoic acid (DPA) 50 130 90
    Docosahexaenoic acid (DHA) 40 20

    In COMPOSITIONS 1a and 1b, the EPA:HPA ratio is between 13 and 190, the EPA:DPA ratio is between 8 and 15, the HPA:DPA ration between 0.05 and 1, the DPA:DHA ratio more than 2.4, preferably more than 4, more preferably more than 6, most preferably more than 10, and the EPA:DHA ratio more than 32, preferably more than 38, more preferably more than 80, most preferably more than 95. The EPA, HPA, DPA and DHA may be composed as a glyceride (such as triglyceride), an ester (such as ethyl ester), or a free fatty acid.
  • Example 18
  • The following is an example of a preferred embodiment of the present invention.
  • COMPOSITION 2
    Minimum Maximum Target
    Composition (mg/g) (mg/g) (mg/g)
    Omega-3 pentaenoic acid 900 980 940
    Eicosapentaenoic acid (EPA) 15 60 30
    Heneicosapentaenoic acid (HPA) 5 60 30
    Docosapentaenoic acid (DPA) 800 950 880
    Docosahexaenoic acid (DHA) 25 <10

    In COMPOSITION 2, the EPA:HPA ratio is between 0.25 and 12, the DPA:EPA ratio is between 13 and 63, the DPA:HPA ration between 13 and 190, the DPA:DHA ratio more than 32, preferably more than 38, more preferably more than 80, most preferably more than 95, and the EPA:DHA ratio more than 00.6, preferably more than 1.5, more preferably more than 2.4, most preferably more than 6. The EPA, HPA, DPA and DHA may be composed as a glyceride (such as triglyceride), an ester (such as ethyl ester), or a free fatty acid.
  • Example 19
  • The following is an example of an embodiment of the present invention.
  • COMPOSITION 3
    Minimum Maximum Target
    Composition (mg/g) (mg/g) (mg/g)
    Docosapentaenoic acid (DPA n-3) 800 990 920

    The DPA may be composed as a glyceride (such as triglyceride), an ester (such as ethyl ester), or a free fatty acid.
  • Example 20
  • The following is an example of an embodiment of the present invention.
  • COMPOSITION 4
    Minimum Maximum Target
    Composition (mg/g) (mg/g) (mg/g)
    Omega-3 pentaenoic acid 930 1000 966
    Eicosapentaenoic acid (EPA) 840 870 853
    Heneicosapentaenoic acid (HPA) 20 40 30
    Docosapentaenoic acid (DPA) 60 100 81
    Docosahexaenoic acid (DHA) 5 20 12
  • Example 21
  • A mixture of DPA and EPA was prepared by combining 1 g DPA Ethyl Ester (SE-133-III) with 10 g EPA Ethyl Ester, 914 mg/g (KD Pharma FM13001) in 150 ml of 95% ethanol/water containing 35 ml of 2M sodium hydroxide. This reaction mixture was stirred overnight at ambient temperature. Tlc analysis showed complete conversion of the ethyl esters to the corresponding acids. The reaction mixture was cooled in an ice bath, acidified with 6N hydrochloric acid and concentrated on a rotavap under reduced pressure. Water and ethyl acetate were added, the phases separated and the aqueous residue extracted with ethyl acetate. The ethyl acetate extracts were combined, dried over sodium sulfate and concentrated to dryness on a rotavap under reduced pressure. Yield: 9.83 g. The ethyl ester mixture was then converted to the free fatty acids as described in Example 8.
  • A representative sample of this ethyl ester composition was analysed using split inject by capillary gas chromatography by a 30 meter×0.25 mm Restek Stabil wax column using temperature programming.
  • DESCRIPTION OF EMBODIMENTS OF THE PRESENT INVENTION
    • 1. A fatty acid composition comprising at least 50% omega-3-fatty acids, salts or derivatives thereof, while comprising eicosapentaenoic acid (EPA; C20:5-n3) and docosapentaenoic acid (DPA; C22:5-n3) and wherein the EPA:DHA ratio is higher than 20:1.
    • 2. A fatty acid composition comprising at least 60% omega-3-fatty acids, salts or derivatives thereof, while comprising eicosapentaenoic acid (EPA; C20:5-n3) and docosapentaenoic acid (DPA; C22:5-n3) and wherein the EPA:DHA ratio is higher than 20:1.
    • 3. A fatty acid composition comprising at least 70% omega-3-fatty acids, salts or derivatives thereof, while comprising eicosapentaenoic acid (EPA; C20:5-n3) and docosapentaenoic acid (DPA; C22:5-n3) and wherein the EPA:DHA ratio is higher than 20:1.
    • 4. A fatty acid composition comprising at least 75% omega-3-fatty acids, salts or derivatives thereof, while comprising eicosapentaenoic acid (EPA; C20:5-n3) and docosapentaenoic acid (DPA; C22:5-n3) and wherein the EPA:DHA ratio is higher than 20:1.
    • 5. A fatty acid composition comprising at least 80% omega-3-fatty acids, salts or derivatives thereof, while comprising eicosapentaenoic acid (EPA; C20:5-n3) and docosapentaenoic acid (DPA; C22:5-n3) and wherein the EPA:DHA ratio is higher than 20:1.
    • 6. A fatty acid composition comprising at least 85% omega-3-fatty acids, salts or derivatives thereof, while comprising eicosapentaenoic acid (EPA; C20:5-n3) and docosapentaenoic acid (DPA; C22:5-n3) and wherein the EPA:DHA ratio is higher than 20:1.
    • 7. A fatty acid composition comprising at least 90% omega-3-fatty acids, salts or derivatives thereof, while comprising eicosapentaenoic acid (EPA; C20:5-n3) and docosapentaenoic acid (DPA; C22:5-n3) and wherein the EPA:DHA ratio is higher than 20:1.
    • 8. A fatty acid composition comprising at least 95% omega-3-fatty acids, salts or derivatives thereof, while comprising eicosapentaenoic acid (EPA; C20:5-n3) and docosapentaenoic acid (DPA; C22:5-n3) and wherein the EPA:DHA ratio is higher than 20:1.
    • 9. A composition according to one of the preferred embodiments 1 through 8, comprising at least 2% docosapentaenoic acid (DPA; C22:5-n3).
    • 10. A composition according to one of the preferred embodiments 1 through 8, comprising at least 4% docosapentaenoic acid (DPA; C22:5-n3).
    • 11. A composition according to one of the preferred embodiments 1 through 8, comprising at least 5% docosapentaenoic acid (DPA; C22:5-n3).
    • 12. A composition according to one of the preferred embodiments 1 through 8, comprising at least 6% docosapentaenoic acid (DPA; C22:5-n3).
    • 13. A composition according to one of the preferred embodiments 1 through 8, comprising at least 7% docosapentaenoic acid (DPA; C22:5-n3).
    • 14. A composition according to one of the preferred embodiments 1 through 8, comprising at least 8% docosapentaenoic acid (DPA; C22:5-n3).
    • 15. A composition according to one of the preferred embodiments 1 through 8, comprising at least 10% docosapentaenoic acid (DPA; C22:5-n3).
    • 16. A composition according to one of the preferred embodiments 1 through 8, comprising at least 12% docosapentaenoic acid (DPA; C22:5-n3).
    • 17. A composition according to one of the preferred embodiments 1 through 8, comprising at least 15% docosapentaenoic acid (DPA; C22:5-n3).
    • 18. A composition according to one of the preferred embodiments 1 through 17, comprising no more than 95% EPA.
    • 19. A composition according to one of the preferred embodiments 1 through 17, comprising no more than 10% omega-6 fatty acids.
    • 20. A composition according to one of the preferred embodiments 1 through 17, comprising no more than 7% omega-6 fatty acids.
    • 21. A composition according to one of the preferred embodiments 1 through 17, comprising no more than 5% omega-6 fatty acids.
    • 22. A composition according to one of the preferred embodiments 1 through 17, comprising no more than 3% omega-6 fatty acids.
    • 23. A composition according to one of the preferred embodiments 1 through 22, comprising no more than 5% arachidonic acid (C22:4-n6).
    • 24. A composition according to one of the preferred embodiments 1 through 22, comprising no more than 4% arachidonic acid (C22:4-n6).
    • 25. A composition according to one of the preferred embodiments 1 through 22, comprising no more than 3% arachidonic acid (C22:4-n6).
    • 26. A composition according to one of the preferred embodiments 1 through 22, comprising no more than 2% arachidonic acid (C22:4-n6).
    • 27. A composition according to one of the preferred embodiments 1 through 22, comprising no more than 1% arachidonic acid (C22:4-n6).
    • 28. A composition according to one of the preferred embodiments 1 through 27, also comprising heneicosapentaenoic acid (C21:5-n3).
    • 29. A composition according to one of the preferred embodiments 1 through 27, comprising at least 0.01% heneicosapentaenoic acid (C21:5-n3).
    • 30. A composition according to one of the preferred embodiments 1 through 27, comprising at least 0.1% heneicosapentaenoic acid (C21:5-n3).
    • 31. A composition according to one of the preferred embodiments 1 through 27, comprising at least 0.3% heneicosapentaenoic acid (C21:5-n3).
    • 32. A composition according to one of the preferred embodiments 1 through 27, comprising at least 0.5% heneicosapentaenoic acid (C21:5-n3).
    • 33. A composition according to one of the preferred embodiments 1 through 27, comprising at least 1% heneicosapentaenoic acid (C21:5-n3).
    • 34. A composition according to one of the preferred embodiments 1 through 27, comprising at least 2% heneicosapentaenoic acid (C21:5-n3).
    • 35. A composition according to one of the preferred embodiments 1 through 27, comprising at least 3% heneicosapentaenoic acid (C21:5-n3).
    • 36. A composition according to one of the preferred embodiments 1 through 27, comprising at least 4% heneicosapentaenoic acid (C21:5-n3).
    • 37. A composition according to one of the preferred embodiments 1 through 27, comprising at least 5% heneicosapentaenoic acid (C21:5-n3).
    • 38. A composition according to one of the preferred embodiments 1 through 37, comprising no more than 5% omega-3 fatty acids that are not omega-3-pentaenoic acids.
    • 39. A composition according to one of the preferred embodiments 1 through 37, comprising no more than 4% omega-3 fatty acids that are not omega-3-pentaenoic acids.
    • 40. A composition according to one of the preferred embodiments 1 through 37, comprising no more than 3% omega-3 fatty acids that are not omega-3-pentaenoic acids.
    • 41. A composition according to one of the preferred embodiments 1 through 37, comprising no more than 2% omega-3 fatty acids that are not omega-3-pentaenoic acids.
    • 42. A composition according to one of the preferred embodiments 1 through 37, comprising no more than 1.5% omega-3 fatty acids that are not omega-3-pentaenoic acids.
    • 43. A composition according to one of the preferred embodiments 1 through 37, comprising no more than 1.25% omega-3 fatty acids that are not omega-3-pentaenoic acids.
    • 44. A composition according to one of the preferred embodiments 1 through 37, comprising no more than 1% omega-3 fatty acids that are not omega-3-pentaenoic acids.
    • 45. A composition according to one of the preferred embodiments 1 through 44, wherein the EPA:DPA ratio is between 99:1 and 1:99.
    • 46. A composition according to one of the preferred embodiments 1 through 44, wherein the EPA:DPA ratio is between 60:1 and 1:60.
    • 47. A composition according to one of the preferred embodiments 1 through 44, wherein the EPA:DPA ratio is between 50:1 and 1:10.
    • 48. A composition according to one of the preferred embodiments 1 through 44, wherein the EPA:DPA ratio is between 40:1 and 1:3.
    • 49. A composition according to one of the preferred embodiments 1 through 44, wherein the EPA:DPA ratio is between 40:1 and 1:2.
    • 50. A composition according to one of the preferred embodiments 1 through 44, wherein the EPA:DPA ratio is between 40:1 and 1:1.
    • 51. A composition according to one of the preferred embodiments 1 through 44, wherein the EPA:DPA ratio is between 30:1 and 1:1.
    • 52. A composition according to one of the preferred embodiments 1 through 44, wherein the EPA:DPA ratio is between 20:1 and 1:1.
    • 53. A composition according to one of the preferred embodiments 1 through 44, wherein the EPA:DPA ratio is between 10:1 and 1:1.
    • 54. A composition according to one of the preferred embodiments 1 through 44, wherein the EPA:DPA ratio is between 5:1 and 1:1.
    • 55. A composition according to one of the preferred embodiments 1 through 44, wherein the EPA:DPA ratio is between 10:1 and 2:1.
    • 56. A composition according to one of the preferred embodiments 1 through 44, wherein the EPA:DPA ratio is between 20:1 and 2:1.
    • 57. A composition according to one of the preferred embodiments 1 through 44, wherein the EPA:DPA ratio is between 30:1 and 2:1.
    • 58. A composition according to one of the preferred embodiments 1 through 44, wherein the EPA:DPA ratio is between 40:1 and 2:1.
    • 59. A composition according to one of the preferred embodiments 1 through 44, wherein the EPA:DPA ratio is between 50:1 and 2:1.
    • 60. A composition according to one of the preferred embodiments 1 through 44, wherein the EPA:DPA ratio is between 10:1 and 3:1.
    • 61. A composition according to one of the preferred embodiments 1 through 44, wherein the EPA:DPA ratio is between 20:1 and 3:1.
    • 62. A composition according to one of the preferred embodiments 1 through 44, wherein the EPA:DPA ratio is between 30:1 and 3:1.
    • 63. A composition according to one of the preferred embodiments 1 through 44, wherein the EPA:DPA ratio is between 40:1 and 3:1.
    • 64. A composition according to one of the preferred embodiments 1 through 44, wherein the EPA:DPA ratio is between 50:1 and 3:1.
    • 65. A composition according to one of the preferred embodiments 1 through 44, wherein the EPA:DPA ratio is between 60:1 and 3:1.
    • 66. A composition according to one of the preferred embodiments 1 through 44, wherein the EPA:DPA ratio is between 10:1 and 5:1.
    • 67. A composition according to one of the preferred embodiments 1 through 44, wherein the EPA:DPA ratio is between 20:1 and 5:1.
    • 68. A composition according to one of the preferred embodiments 1 through 44, wherein the EPA:DPA ratio is between 30:1 and 5:1.
    • 69. A composition according to one of the preferred embodiments 1 through 44, wherein the EPA:DPA ratio is between 40:1 and 5:1.
    • 70. A composition according to one of the preferred embodiments 1 through 44, wherein the EPA:DPA ratio is between 50:1 and 5:1.
    • 71. A composition according to one of the preferred embodiments 1 through 44, wherein the EPA:DPA ratio is between 60:1 and 5:1.
    • 72. A composition according to one of the preferred embodiments 1 through 44, wherein the EPA:DPA ratio is between 20:1 and 10:1.
    • 73. A composition according to one of the preferred embodiments 1 through 44, wherein the EPA:DPA ratio is between 30:1 and 10:1.
    • 74. A composition according to one of the preferred embodiments 1 through 44, wherein the EPA:DPA ratio is between 40:1 and 10:1.
    • 75. A composition according to one of the preferred embodiments 1 through 44, wherein the EPA:DPA ratio is between 50:1 and 10:1.
    • 76. A composition according to one of the preferred embodiments 1 through 44, wherein the EPA:DPA ratio is between 60:1 and 10:1.
    • 77. A composition according to one of the preferred embodiments 1 through 44, wherein the EPA:DPA ratio is between 100:1 and 10:1.
    • 78. A composition according to one of the preferred embodiments 1 through 44, comprising between 55% and 95% EPA.
    • 79. A composition according to one of the preferred embodiments 1 through 44, comprising between 60% and 95% EPA.
    • 80. A composition according to one of the preferred embodiments 1 through 44, comprising between 65% and 95% EPA.
    • 81. A composition according to one of the preferred embodiments 1 through 44, comprising between 70% and 95% EPA.
    • 82. A composition according to one of the preferred embodiments 1 through 44, comprising between 75% and 95% EPA.
    • 83. A composition according to one of the preferred embodiments 1 through 44, comprising between 80% and 95% EPA.
    • 84. A composition according to one of the preferred embodiments 1 through 44, comprising between 85% and 95% EPA.
    • 85. A composition according to one of the preferred embodiments 1 through 44, comprising between 90% and 95% EPA.
    • 86. A composition according to one of the preferred embodiments 1 through 44, comprising between 1% and 3% DPA.
    • 87. A composition according to one of the preferred embodiments 1 through 44, comprising between 1% and 5% DPA.
    • 88. A composition according to one of the preferred embodiments 1 through 44, comprising between 2% and 10% DPA.
    • 89. A composition according to one of the preferred embodiments 1 through 44, comprising between 3% and 20% DPA.
    • 90. A composition according to one of the preferred embodiments 1 through 44, comprising between 3% and 30% DPA.
    • 91. A composition according to one of the preferred embodiments 1 through 44, comprising between 3% and 50% DPA.
    • 92. A composition according to one of the preferred embodiments 1 through 44, comprising between 3% and 75% DPA.
    • 93. A composition according to one of the preferred embodiments 1 through 44, comprising between 3% and 90% DPA.
    • 94. A fatty acid composition according to one of the preferred embodiments 1 through 93, in which the fatty acids are present as ethyl esters.
    • 95. A fatty acid composition according to one of the preferred embodiments 1 through 93, in which the fatty acids are present as free fatty acids.
    • 96. A fatty acid composition according to one of the preferred embodiments 1 through 93, in which the fatty acids are present as esters in di-glyceride form.
    • 97. A fatty acid composition according to one of the preferred embodiments 1 through 93, in which the fatty acids are present as esters in triglyceride form.
    • 98. A fatty acid composition according to one of the preferred embodiments 94 through 97, also comprising a suitable anti-oxidant in a concentration sufficient to protect the fatty acids of the composition from oxidation.
    • 99. A pharmaceutically suitable formulation comprising one of the compositions according to preferred embodiments 94 through 98, in which the amount of eicosapentaenoic acid plus docosapentaenoic acid is present in an amount between 100 and 10,000 mg.
    • 100. A pharmaceutically suitable formulation or dosage form comprising one of the compositions according to preferred embodiments 94 through 98, in which the amount of eicosapentaenoic acid plus docosapentaenoic acid is present in an amount between 250 and 1,250 mg.
    • 101. A pharmaceutically suitable formulation or dosage form comprising one of the compositions according to preferred embodiments 94 through 98, in which the amount of eicosapentaenoic acid plus docosapentaenoic acid is present in an amount between 500 and 1,100 mg.
    • 102. A pharmaceutically suitable formulation or dosage form comprising one of the compositions according to preferred embodiments 94 through 98, in which the amount of eicosapentaenoic acid plus docosapentaenoic acid is present in an amount between 100 and 10,000 mg.
    • 103. A method of administration or treatment to a subject of a formulation or dosage form according to one of the preferred embodiments 94 through 102 at a daily dose between 100 and 10,000 mg.
    • 104. A method of administration or treatment to a subject of a formulation or dosage form according to one of the preferred embodiments 94 through 102 at a daily dose between 500 and 5,000 mg.
    • 105. A method of administration or treatment to a subject of a formulation or dosage form according to one of the preferred embodiments 94 through 102 at a daily dose between 1,500 and 4,100 mg.
    • 106. A method of treatment according to preferred embodiments 103 through 105, in which the subject is a patient diagnosed with very high triglycerides (equal or more than 500 mg/dL).
    • 107. A method of treatment according to preferred embodiments 103 through 105, in which the subject is a patient diagnosed with high triglycerides (equal to or more than 200 mg/dL but less than 500 mg/dL).
    • 108. A method of treatment according to preferred embodiments 103 through 105, in which the subject is a patient already undergoing treatment with a statin and then diagnosed with high triglycerides (equal to or more than 200 mg/dL but less than 500 mg/dL).
    • 109. A method of treatment according to preferred embodiments 103 through 105, in which the subject is a patient diagnosed with mixed dyslipidemia with TG 200-499 mg/dL and LDL-cholesterol equal to or more than 190 mg/dL.
    • 110. A method of treatment according to preferred embodiments 103 through 105, in which the subject is a patient diagnosed with mixed dyslipidemia with TG 300-700 mg/dL and LDL-cholesterol equal to or more than 190 mg/dL.
    • 111. A method of treatment according to preferred embodiments 103 through 105, in which the subject is a patient diagnosed with mixed dyslipidemia with TG 200-499 mg/dL and non-HDL-cholesterol equal to or more than 200 mg/dL.
    • 112. A method of treatment according to preferred embodiments 103 through 105, in which the subject is a patient diagnosed with mixed dyslipidemia with TG 300-700 mg/dL and non-HDL-cholesterol equal to or more than 200 mg/dL.
    • 113. A method of treatment according to preferred embodiments 103 through 105, in which the subject is a patient diagnosed with mixed dyslipidemia with TG 200-499 mg/dL and LDL-cholesterol equal to or more than 160 mg/dL.
    • 114. A method of treatment according to preferred embodiments 103 through 105, in which the subject is a patient diagnosed with mixed dyslipidemia with TG 300-700 mg/dL and LDL-cholesterol equal to or more than 160 mg/dL.
    • 115. A method of treatment according to preferred embodiments 103 through 105, in which the subject is a patient diagnosed with mixed dyslipidemia with TG 200-499 mg/dL and non-HDL-cholesterol equal to or more than 160 mg/dL.
    • 116. A method of treatment according to preferred embodiments 103 through 105, in which the subject is a patient diagnosed with mixed dyslipidemia with TG 300-700 mg/dL and non-HDL-cholesterol equal to or more than 160 mg/dL.
    • 117. A method of treatment according to preferred embodiments 103 through 105, in which the subject is a patient diagnosed with mixed dyslipidemia with TG 200-499 mg/dL and LDL-cholesterol equal to or more than 130 mg/dL.
    • 118. A method of treatment according to preferred embodiments 103 through 105, in which the subject is a patient diagnosed with mixed dyslipidemia with TG 300-700 mg/dL and LDL-cholesterol equal to or more than 130 mg/dL.
    • 119. A method of treatment according to preferred embodiments 103 through 105, in which the subject is a patient diagnosed with mixed dyslipidemia with TG 200-499 mg/dL and non-HDL-cholesterol equal to or more than 130 mg/dL.
    • 120. A method of treatment according to preferred embodiments 103 through 105, in which the subject is a patient diagnosed with mixed dyslipidemia with TG 300-700 mg/dL and non-HDL-cholesterol equal to or more than 130 mg/dL.
    • 121. A method of treatment according to preferred embodiments 103 through 105, in which the subject is a patient diagnosed/assessed to be at substantially elevated risk for cardiovascular events.
    • 122. A method of treatment according to preferred embodiments 103 through 105, in which the subject is a patient diagnosed with diabetes.
    • 123. A method of treatment according to preferred embodiments 103 through 105, in which the subject is a patient diagnosed with pre-diabetes or metabolic syndrome.
    • 124. A method of treatment according to one of the preferred embodiments 103 through 123, in which the treatment results in significant reduction of blood, serum or plasma triglyceride levels.
    • 125. A method of treatment according to one of the preferred embodiments 103 through 123, in which the treatment results in significant reduction of blood, serum or plasma triglyceride levels while not significantly increasing blood, serum or plasma LDL-cholesterol levels.
    • 126. A method of treatment according to one of the preferred embodiments 103 through 123, in which the treatment results in significant reduction of blood, serum or plasma total-cholesterol levels.
    • 127. A method of treatment according to one of the preferred embodiments 103 through 123, in which the treatment results in significant reduction of blood, serum or plasma non-HDL-cholesterol levels.
    • 128. A method of treatment according to one of the preferred embodiments 103 through 123, in which the treatment results in significant reduction of blood, serum or plasma LDL-cholesterol levels.
    • 129. A method of treatment according to one of the preferred embodiments 103 through 123, in which the treatment results in significant reduction of blood, serum or plasma VLDL-cholesterol levels.
    • 130. A method of treatment according to one of the preferred embodiments 103 through 123, in which the treatment results in significant reduction of blood, serum or plasma VLDL-cholesterol levels while not significantly increasing blood, serum or plasma LDL-cholesterol levels.
    • 131. A method of treatment according to one of the preferred embodiments 103 through 123, in which the treatment results in significant reduction of blood, serum or plasma apo-B levels.
    • 132. A method of treatment according to one of the preferred embodiments 103 through 123, in which the treatment results in significant reduction of blood, serum or plasma apo-C-III levels.
    • 133. A method of treatment according to one of the preferred embodiments 103 through 123, in which the treatment results in significant reduction of blood, serum or plasma LP-PLA2 levels.
    • 134. A method of treatment according to one of the preferred embodiments 103 through 123, in which the treatment results in significant reduction of blood, serum or plasma hs-CRP levels.
    • 135. A method of treatment according to one of the preferred embodiments 103 through 123, in which the treatment results in significant increase of blood, serum or plasma HDL-cholesterol levels.
    • 136. A method of treatment according to one of the preferred embodiments 103 through 123, in which the treatment results in significant increase of blood, serum or plasma apo-A levels.
    • 137. A method of treatment according to one of the preferred embodiments 103 through 123, in which the treatment results in significant reduction of the risk of suffering certain cardiovascular events.
    • 138. The composition of claim 1, wherein the ratio of EPA to DPA (EPA:DPA) is between 15:1 to 8:1.
    • 139. An orally administrable composition comprising fatty acids, wherein at least 50% by weight of the fatty acids comprise omega-3-pentaenoic acids, salts, esters, or derivatives thereof, wherein the composition comprises eicosapentaenoic acid (EPA), docosapentaenoic acid (DPA), and docosahexaenoic acid (DHA), and wherein the ratio of DHA to EPA (DHA:EPA) is less than 1:20, and wherein the ratio of DHA to DPA (DHA:DPA) is less than 2:1.

Claims (63)

What is claimed:
1. A method of treating, preventing and/or reducing and improving symptoms associated with the occurrence of MACE, coronary heart disease, vascular disease, and/or atherosclerotic disease in a subject in need thereof, comprising administering to the subject a composition comprising fatty acids, wherein at least 50% by weight of the fatty acids comprise omega-3 fatty acids, salts, esters, or derivatives thereof, wherein the omega-3 fatty acids comprise eicosapentaenoic acid (EPA) and docosapentaenoic acid (DPA) and wherein the ratio of docosahexaenoic acid to DHA to EPA (DHA:EPA) is less than 1:10, and wherein the ratio of DHA to DPA (DHA:DPA) is less than 2:1.
2. The method of claim 1, wherein the subject also suffers from another morbidity selected from the group consisting of: atherosclerosis, hypertriglyceridemia, mixed dyslipidemia, hypercholesterolemia, peripheral artery disease (PAD), recent myocardial infarction (post-MI), Acute Coronary Syndrome (ACS), ACS without ST-segment elevation (NSTEMI), ACS with ST-segment elevation (STEMI), heart failure, cardiac arrhythmias, blood coagulatory conditions associated with cardiac arrhythmias, hypertension, coagulation related disorders, nephropathy, renal impairment, renal insufficiency requiring dialysis, renal insufficiency without dialysis, retinopathy, cognitive impairment, dementia, and metabolic syndrome.
3. The method of claim 1, wherein the ratio of DHA:EPA is less than 1:20.
4. The method of claim 1, wherein the ratio of DHA:DPA is less than 1:1.
5. The method of claim 1, wherein the ratio of EPA to DPA (EPA:DPA) is between 30:1 and 1:1.
6. The method of claim 1, wherein ratio of DHA:EPA is less than 1:10.
7. The method of claim 1, wherein the composition comprises DHA in an amount less than 5% of the total amount of fatty acids.
8. The method of claim 1, wherein the composition comprises EPA in an amount between about 80% and about 90% of the total amount of fatty acids.
9. The method of claim 1, wherein the composition comprises DPA in an amount between about 5% and about 15% of the total amount of fatty acids.
10. The method of claim 1, wherein the composition comprises DPA free fatty acid or a salt, ester or derivative of DPA.
11. The method of claim 1, wherein composition comprises EPA free fatty acid or a salt, ester or derivative of EPA.
12. A method of treating, preventing and/or reducing and improving symptoms associated with the occurrence of MACE, coronary heart disease, vascular disease, and/or atherosclerotic disease in a subject in need thereof, comprising administering to the subject a composition comprising eicosapentaenoic acid (EPA) in an amount between about 750 mg/g to about 950 mg/g,
and wherein the composition comprises no more than 5% DHA of the total amount of fatty acids, and
and wherein the ratio of DHA:DPA is 1:1 or lower.
13. The method of claim 12, wherein the subject also suffers from another morbidity selected from the group consisting of: atherosclerosis, hypertriglyceridemia, mixed dyslipidemia, hypercholesterolemia, peripheral artery disease (PAD), recent myocardial infarction (post-MI), Acute Coronary Syndrome (ACS), ACS without ST-segment elevation (NSTEMI), ACS with ST-segment elevation (STEMI), heart failure, cardiac arrhythmias, blood coagulatory conditions associated with cardiac arrhythmias, hypertension, coagulation related disorders, nephropathy, renal impairment, renal insufficiency requiring dialysis, renal insufficiency without dialysis, retinopathy, cognitive impairment, dementia, and metabolic syndrome.
14. The method of claim 12, wherein the composition comprises eicosapentaenoic acid (EPA) in an amount between about 800 mg/g to about 900 mg/g,
and wherein the composition comprises no more than 5% DHA of the total amount of fatty acids, and
and wherein the ratio of DHA:DPA is 1:1 or lower.
15. The method of claim 12, wherein the composition comprises eicosapentaenoic acid (EPA) in an amount between about 830 mg/g to about 870 mg/g,
and wherein the composition comprises no more than 5% DHA of the total amount of fatty acids, and
and wherein the ratio of DHA:DPA is 1:1 or lower.
16. The method of claim 12, wherein the composition comprises docosapentaenoic acid (DPA) is an amount between about 60 mg/g to about 120 mg/g.
17. The method of claim 1, wherein the composition comprises docosapentaenoic acid (DPA) is an amount between about 70 mg/g to about 100 mg/g.
18. A method of treating, preventing and/or reducing and improving symptoms associated with the occurrence of MACE, coronary heart disease, vascular disease, and/or atherosclerotic disease in a subject in need thereof, comprising administering to the subject a composition comprising:
eicosapentaenoic acid (EPA) in an amount between about 70% to about 95% of the total amount of fatty acids and
docosapentaenoic acid (DPA),
wherein the composition comprises no more than 5% docosahexaenoic acid (DHA) of the total amount of fatty acids, and
wherein the ratio of DHA:DPA is 1:1 or lower.
19. The method of claim 18, wherein the subject also suffers from another morbidity selected from the group consisting of: atherosclerosis, hypertriglyceridemia, mixed dyslipidemia, hypercholesterolemia, peripheral artery disease (PAD), recent myocardial infarction (post-MI), Acute Coronary Syndrome (ACS), ACS without ST-segment elevation (NSTEMI), ACS with ST-segment elevation (STEMI), heart failure, cardiac arrhythmias, blood coagulatory conditions associated with cardiac arrhythmias, hypertension, coagulation related disorders, nephropathy, renal impairment, renal insufficiency requiring dialysis, renal insufficiency without dialysis, retinopathy, cognitive impairment, dementia, and metabolic syndrome.
20. The method of claim 18, wherein the composition comprises eicosapentaenoic acid (EPA) in an amount between about 80% to about 90% of the total amount of fatty acids.
21. The method of 18, wherein the composition comprises eicosapentaenoic acid (EPA) in an amount between about 82% to about 88% of the total amount of fatty acids.
22. The method of 18, wherein the composition comprises docosapentaenoic acid (DPA) in amount between about 5% and about 15% of the total amount of fatty acids.
23. The method of 18, wherein the composition comprises docosapentaenoic acid (DPA) in an amount between about 6% to about 12% of the total amount of fatty acids.
24. The method of claim 18, wherein the composition comprises EPA free fatty acid or a salt, ester or derivative of EPA.
25. A method of treating, preventing and/or reducing and improving symptoms associated with the occurrence of MACE, coronary heart disease, vascular disease, and/or atherosclerotic disease in a subject in need thereof, comprising administering to the subject a composition comprising eicosapentaenoic acid (EPA) in a daily dosage amount of between about 1000 mg to about 5000 mg,
and further comprising docosapentaenoic acid (DPA) and docosahexaenoic acid (DHA),
wherein the composition comprises no more than 5% DHA of the total amount of fatty acids, and wherein the ratio of DHA:DPA is 1:1 or lower.
26. The method of claim 25, wherein the subject also suffers from another morbidity selected from the group consisting of: atherosclerosis, hypertriglyceridemia, mixed dyslipidemia, hypercholesterolemia, peripheral artery disease (PAD), recent myocardial infarction (post-MI), Acute Coronary Syndrome (ACS), ACS without ST-segment elevation (NSTEMI), ACS with ST-segment elevation (STEMI), heart failure, cardiac arrhythmias, blood coagulatory conditions associated with cardiac arrhythmias, hypertension, coagulation related disorders, nephropathy, renal impairment, renal insufficiency requiring dialysis, renal insufficiency without dialysis, retinopathy, cognitive impairment, dementia, and metabolic syndrome.
27. The method of claim 25, wherein the composition comprises eicosapentaenoic acid (EPA) in a daily dosage amount selected from the group consisting of: about 1735 mg to about 1855 mg, about 2520 mg to about 2780 mg, and about 3360 mg to about 3710 mg.
28. The method of claim 25, wherein the composition comprises eicosapentaenoic acid (EPA) in a daily dosage amount selected from the group consisting of: about 1750 mg to about 1950 mg, about 1800 mg to about 2000 mg about 2650 mg to about 2950 mg, and about 3500 mg to about 3900 mg.
29. The method of claim 25, wherein the composition comprises eicosapentaenoic acid (EPA) in daily dosage amount selected from the group consisting of: about 1900 mg to about 2100 mg, about 2700 mg to about 3300 mg, and about 3700 mg to about 4300 mg.
30. The method of claim 25, wherein the composition comprises EPA free fatty acid or a salt, ester or derivative of EPA.
31. The method of claim 25, wherein the composition comprises DPA free fatty acid or a salt, ester or derivative of DPA.
32. A method of treating, preventing and/or reducing and improving symptoms associated with the occurrence of MACE, coronary heart disease, vascular disease, and/or atherosclerotic disease in a subject in need thereof, comprising administering to the subject a composition comprising eicosapentaenoic acid (EPA) and docosapentaenoic acid (DPA), wherein the amount of EPA and DPA is about 55% or more by weight of the total amount of fatty acids, and wherein the ratio of DHA:DPA is no more than 1:1.
33. The method of claim 32, wherein the subject also suffers from another morbidity selected from the group consisting of: atherosclerosis, hypertriglyceridemia, mixed dyslipidemia, hypercholesterolemia, peripheral artery disease (PAD), recent myocardial infarction (post-MI), Acute Coronary Syndrome (ACS), ACS without ST-segment elevation (NSTEMI), ACS with ST-segment elevation (STEMI), heart failure, cardiac arrhythmias, blood coagulatory conditions associated with cardiac arrhythmias, hypertension, coagulation related disorders, nephropathy, renal impairment, renal insufficiency requiring dialysis, renal insufficiency without dialysis, retinopathy, cognitive impairment, dementia, and metabolic syndrome.
34. The method of claim 32, wherein the composition comprises a daily dosage of about 120 mg/day to about 150 mg/day.
35. The method of claim 32, wherein the composition comprises a daily dosage of DPA of about 150 mg/day to about 200 mg/day.
36. The method of claim 32, wherein the composition comprises a daily dosage of DPA of about 200 mg/day to about 250 mg/day.
37. The method of claim 32, wherein the composition comprises a daily dosage of DPA of about 250 mg/day to about 300 mg/day.
38. The method of claim 32, wherein the composition comprises a daily dosage of DPA of about 300 mg/day to about 400 mg/day.
39. The method of claim 32, wherein the composition comprises a daily dosage of DPA of about 400 mg/day to about 600 mg/day.
40. The method of claim 32, wherein the composition comprises a daily dosage of DPA of about 600 mg/day to about 1000 mg/day.
41. The method of claim 32, wherein the composition further comprises eicosapentaenoic acid (EPA) and, wherein the amount of EPA and DPA is about 55% or more by weight of the total amount of fatty acids, and wherein the ratio of DHA:DPA is no more than 1:1.
42. The method of claim 32, wherein the amount of EPA and DPA is selected from the group consisting of about 60% or more, about 65% or more, about 70% or more, about 75% or more, about 80% or more, about 85% or more, and about 90% or more by weight of the total amount of fatty acids.
43. The method of claim 32, wherein the composition comprises further omega-6 fatty acids in an amount of no more than 6% of total amount of fatty acids.
44. The method of claim 32, wherein the composition comprises no more than about 30% docosahexaenoic acid (DHA) by weight of fatty acids present in the composition.
45. The method of claim 32, wherein the composition comprises no more than about 10% docosahexaenoic acid (DHA) by weight of fatty acids present in the composition.
46. The method of claim 32, wherein the composition comprises no more than about 5% docosahexaenoic acid (DHA) by weight of fatty acids present in the composition.
47. The method of claim 32, wherein the composition comprises DPA in ethyl ester form.
48. The method of claim 32, wherein the composition comprises DPA in free fatty acid form.
49. The method of claim 32, wherein the composition further comprises docosahexaenoic acid (DHA), and the ratio of DHA:DPA is no more than 2:1.
50. The method of claim 32, wherein the composition further comprises docosahexaenoic acid (DHA), and the ratio of DHA:DPA is no more than 1:1.
51. The method of claim 32, wherein the composition further comprises docosahexaenoic acid (DHA), and the ratio of DHA:DPA is no more than 1:2.
52. The method of claim 32, wherein the composition further comprises docosahexaenoic acid (DHA), and the ratio of DHA:DPA is no more than 1:4.
53. The method of claim 32, wherein the composition comprises at least about 6% docosapentaenoic acid (DPA) by weight of fatty acids present in the composition.
54. The method of claim 32, wherein the composition comprises at least about 20% docosapentaenoic acid (DPA) by weight of fatty acids present in the composition.
55. The method of claim 32, wherein the composition comprises at least about 50% docosapentaenoic acid (DPA) by weight of fatty acids present in the composition.
56. A method of treating, preventing and/or reducing and improving symptoms associated with the occurrence of MACE, coronary heart disease, vascular disease, and/or atherosclerotic disease in a subject in need thereof, comprising administering to the subject a composition comprising: docosapentaenoic acid (DPA) in an amount between about 50% to about 80% of the total amount of fatty acids, docosahexaenoic acid (DHA) in an amount between about 25% to about 40% of the total amount of fatty acids, and optionally eicosapentaenoic acid (EPA) in an amount less than about 10% of the total amount of fatty acids.
57. The method of claim 56, wherein the subject also suffers from another morbidity selected from the group consisting of: atherosclerosis, hypertriglyceridemia, mixed dyslipidemia, hypercholesterolemia, peripheral artery disease (PAD), recent myocardial infarction (post-MI), Acute Coronary Syndrome (ACS), ACS without ST-segment elevation (NSTEMI), ACS with ST-segment elevation (STEMI), heart failure, cardiac arrhythmias, blood coagulatory conditions associated with cardiac arrhythmias, hypertension, coagulation related disorders, nephropathy, renal impairment, renal insufficiency requiring dialysis, renal insufficiency without dialysis, retinopathy, cognitive impairment, dementia, and metabolic syndrome.
58. The method of claim 56, wherein the composition comprises docosapentaenoic acid (DPA) in an amount between about 50% to 75% of the total amount of fatty acids.
59. The method of claim 56, wherein the composition comprises docosapentaenoic acid (DPA) in an amount between about 50% to 65% of the total amount of fatty acids.
60. The method of claim 56, wherein the composition comprises docosahexaenoic acid (DHA) in an amount between about 25% to about 35%, of the total amount of fatty acids.
61. The method of claim 56, wherein the composition comprises docosahexaenoic acid (DHA) in an amount between about 30% to about 35% of the total amount of fatty acids.
62. The method of claim 56, wherein the composition comprises eicosapentaenoic acid (EPA) in an amount less than about 8% of the total amount of fatty acids.
63. The method of claim 56, wherein the composition comprises eicosapentaenoic acid (EPA) in an amount less than about 5% of the total amount of fatty acids.
US14/109,254 2012-06-17 2013-12-17 Omega-3 pentaenoic acid compositions and methods of use Abandoned US20140107199A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US14/109,254 US20140107199A1 (en) 2012-06-17 2013-12-17 Omega-3 pentaenoic acid compositions and methods of use

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US201261660757P 2012-06-17 2012-06-17
US201261734331P 2012-12-06 2012-12-06
US201361780948P 2013-03-13 2013-03-13
PCT/US2013/046176 WO2013192109A1 (en) 2012-06-17 2013-06-17 Omega-3 pentaenoic acid compositions and methods of use
US14/109,254 US20140107199A1 (en) 2012-06-17 2013-12-17 Omega-3 pentaenoic acid compositions and methods of use

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2013/046176 Continuation-In-Part WO2013192109A1 (en) 2012-06-17 2013-06-17 Omega-3 pentaenoic acid compositions and methods of use

Publications (1)

Publication Number Publication Date
US20140107199A1 true US20140107199A1 (en) 2014-04-17

Family

ID=49769263

Family Applications (13)

Application Number Title Priority Date Filing Date
US14/101,198 Abandoned US20140100275A1 (en) 2012-06-17 2013-12-09 Methods of administering compositions comprising docosapentaenoic acid
US14/101,114 Abandoned US20140100272A1 (en) 2012-06-17 2013-12-09 Methods of administering compositions comprising docosapentaenoic acid
US14/101,154 Abandoned US20140100281A1 (en) 2012-06-17 2013-12-09 Omega-3 pentaenoic acid compositions and methods of use
US14/101,188 Abandoned US20140107198A1 (en) 2012-06-17 2013-12-09 Omega-3 pentaenoic acid compositions and methods of use
US14/101,173 Expired - Fee Related US10058521B2 (en) 2012-06-17 2013-12-09 Omega-3 pentaenoic acid compositions and methods of use
US14/101,146 Abandoned US20140107205A1 (en) 2012-06-17 2013-12-09 Omega-3 pentaenoic acid compositions and methods of use
US14/101,165 Expired - Fee Related US8906964B2 (en) 2012-06-17 2013-12-09 Methods of administering compositions comprising docosapentaenoic acid
US14/101,175 Abandoned US20140100274A1 (en) 2012-06-17 2013-12-09 Methods of administering compositions comprising docosapentaenoic acid
US14/109,194 Abandoned US20140107206A1 (en) 2012-06-17 2013-12-17 Omega-3 pentaenoic acid compositions and methods of use
US14/109,254 Abandoned US20140107199A1 (en) 2012-06-17 2013-12-17 Omega-3 pentaenoic acid compositions and methods of use
US14/109,338 Abandoned US20140107200A1 (en) 2012-06-17 2013-12-17 Omega-3 pentaenoic acid compositions and methods of use
US14/279,155 Abandoned US20140249226A1 (en) 2012-06-17 2014-05-15 Methods of administering compositions comprising docosapentaenoic acid
US17/205,915 Pending US20210205254A1 (en) 2012-06-17 2021-03-18 Omega-3 pentaenoic acid compositions and methods of use

Family Applications Before (9)

Application Number Title Priority Date Filing Date
US14/101,198 Abandoned US20140100275A1 (en) 2012-06-17 2013-12-09 Methods of administering compositions comprising docosapentaenoic acid
US14/101,114 Abandoned US20140100272A1 (en) 2012-06-17 2013-12-09 Methods of administering compositions comprising docosapentaenoic acid
US14/101,154 Abandoned US20140100281A1 (en) 2012-06-17 2013-12-09 Omega-3 pentaenoic acid compositions and methods of use
US14/101,188 Abandoned US20140107198A1 (en) 2012-06-17 2013-12-09 Omega-3 pentaenoic acid compositions and methods of use
US14/101,173 Expired - Fee Related US10058521B2 (en) 2012-06-17 2013-12-09 Omega-3 pentaenoic acid compositions and methods of use
US14/101,146 Abandoned US20140107205A1 (en) 2012-06-17 2013-12-09 Omega-3 pentaenoic acid compositions and methods of use
US14/101,165 Expired - Fee Related US8906964B2 (en) 2012-06-17 2013-12-09 Methods of administering compositions comprising docosapentaenoic acid
US14/101,175 Abandoned US20140100274A1 (en) 2012-06-17 2013-12-09 Methods of administering compositions comprising docosapentaenoic acid
US14/109,194 Abandoned US20140107206A1 (en) 2012-06-17 2013-12-17 Omega-3 pentaenoic acid compositions and methods of use

Family Applications After (3)

Application Number Title Priority Date Filing Date
US14/109,338 Abandoned US20140107200A1 (en) 2012-06-17 2013-12-17 Omega-3 pentaenoic acid compositions and methods of use
US14/279,155 Abandoned US20140249226A1 (en) 2012-06-17 2014-05-15 Methods of administering compositions comprising docosapentaenoic acid
US17/205,915 Pending US20210205254A1 (en) 2012-06-17 2021-03-18 Omega-3 pentaenoic acid compositions and methods of use

Country Status (5)

Country Link
US (13) US20140100275A1 (en)
EP (1) EP2861227A4 (en)
AU (1) AU2013277441B2 (en)
CA (1) CA2916208A1 (en)
WO (1) WO2013192109A1 (en)

Cited By (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9050308B2 (en) 2012-01-06 2015-06-09 Omthera Pharmaceuticals, Inc. DPA-enriched compositions of omega-3 polyunsaturated fatty acids in free acid form
US9492545B2 (en) 2012-05-07 2016-11-15 Omthera Pharmaceuticals Inc. Compositions of statins and omega-3 fatty acids
US20180333383A1 (en) * 2017-05-19 2018-11-22 Amarin Pharmaceuticals Ireland Limited Compositions and Methods for Lowering Triglycerides in a Subject Having Reduced Kidney Function
US10449172B2 (en) 2009-04-29 2019-10-22 Amarin Pharmaceuticals Ireland Limited Stable pharmaceutical composition and methods of using same
US10493058B2 (en) 2009-09-23 2019-12-03 Amarin Pharmaceuticals Ireland Limited Pharmaceutical composition comprising omega-3 fatty acid and hydroxy-derivative of a statin and methods of using same
US10537544B2 (en) 2011-11-07 2020-01-21 Amarin Pharmaceuticals Ireland Limited Methods of treating hypertriglyceridemia
US10555925B1 (en) 2012-06-29 2020-02-11 Amarin Pharmaceuticals Ireland Limited Methods of reducing the risk of a cardiovascular event in a subject at risk for cardiovascular disease
US10561631B2 (en) 2014-06-11 2020-02-18 Amarin Pharmaceuticals Ireland Limited Methods of reducing RLP-C
US10610508B2 (en) 2013-02-06 2020-04-07 Amarin Pharmaceuticals Ireland Limited Methods of reducing apolipoprotein C-III
US10624870B2 (en) 2009-04-29 2020-04-21 Amarin Pharmaceuticals Ireland Limited Methods of treating mixed dyslipidemia
US10668042B2 (en) 2018-09-24 2020-06-02 Amarin Pharmaceuticals Ireland Limited Methods of reducing the risk of cardiovascular events in a subject
US10722485B2 (en) 2013-10-10 2020-07-28 Amarin Pharmaceuticals Ireland Limited Compositions and methods for lowering triglycerides without raising LDL-C levels in a subject on concomitant statin therapy
US10842768B2 (en) 2009-06-15 2020-11-24 Amarin Pharmaceuticals Ireland Limited Compositions and methods for lowering triglycerides
US10842765B2 (en) 2016-03-15 2020-11-24 Amarin Pharmaceuticals Ireland Limited Methods of reducing or preventing oxidation of small dense ldl or membrane polyunsaturated fatty acids
US10851374B2 (en) 2013-02-13 2020-12-01 Amarin Pharmaceuticals Ireland Limited Compositions comprising eicosapentaenoic acid and mipomersen and methods of use thereof
US10888539B2 (en) 2013-09-04 2021-01-12 Amarin Pharmaceuticals Ireland Limited Methods of treating or preventing prostate cancer
US10966968B2 (en) 2013-06-06 2021-04-06 Amarin Pharmaceuticals Ireland Limited Co-administration of rosiglitazone and eicosapentaenoic acid or a derivative thereof
US10973796B2 (en) 2012-01-06 2021-04-13 Amarin Pharmaceuticals Ireland Limited Compositions and methods for lowering levels of high-sensitivity C-reactive protein (hs-CRP) in a subject
US11058661B2 (en) 2018-03-02 2021-07-13 Amarin Pharmaceuticals Ireland Limited Compositions and methods for lowering triglycerides in a subject on concomitant statin therapy and having hsCRP levels of at least about 2 mg/L
US11141399B2 (en) 2012-12-31 2021-10-12 Amarin Pharmaceuticals Ireland Limited Methods of treating or preventing nonalcoholic steatohepatitis and/or primary biliary cirrhosis
US11179362B2 (en) 2012-11-06 2021-11-23 Amarin Pharmaceuticals Ireland Limited Compositions and methods for lowering triglycerides without raising LDL-C levels in a subject on concomitant statin therapy
US11291643B2 (en) 2011-11-07 2022-04-05 Amarin Pharmaceuticals Ireland Limited Methods of treating hypertriglyceridemia
US11446269B2 (en) 2014-06-16 2022-09-20 Amarin Pharmaceuticals Ireland Limited Methods of reducing or preventing oxidation of small dense LDL or membrane polyunsaturated fatty acids
US11547710B2 (en) 2013-03-15 2023-01-10 Amarin Pharmaceuticals Ireland Limited Pharmaceutical composition comprising eicosapentaenoic acid and derivatives thereof and a statin
US11712429B2 (en) 2010-11-29 2023-08-01 Amarin Pharmaceuticals Ireland Limited Low eructation composition and methods for treating and/or preventing cardiovascular disease in a subject with fish allergy/hypersensitivity
US11712428B2 (en) 2010-11-29 2023-08-01 Amarin Pharmaceuticals Ireland Limited Low eructation composition and methods for treating and/or preventing cardiovascular disease in a subject with fish allergy/hypersensitivity

Families Citing this family (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB9901809D0 (en) * 1999-01-27 1999-03-17 Scarista Limited Highly purified ethgyl epa and other epa derivatives for psychiatric and neurological disorderes
EP3578177A1 (en) 2008-09-02 2019-12-11 Amarin Pharmaceuticals Ireland Limited Pharmaceutical composition comprising eicosapentaenoic acid and nicotinic acid and methods of using same
US20120178813A1 (en) 2011-01-12 2012-07-12 Thetis Pharmaceuticals Llc Lipid-lowering antidiabetic agent
EP2861227A4 (en) * 2012-06-17 2016-01-27 Matinas Biopharma Inc Omega-3 pentaenoic acid compositions and methods of use
US9382187B2 (en) 2012-07-10 2016-07-05 Thetis Pharmaceuticals Llc Tri-salt form of metformin
CA2894183A1 (en) * 2012-12-06 2014-06-12 Matinas Biopharma, Inc. Omega-3 pentaenoic acid compositions and methods of use
US9814733B2 (en) 2012-12-31 2017-11-14 A,arin Pharmaceuticals Ireland Limited Compositions comprising EPA and obeticholic acid and methods of use thereof
US9662307B2 (en) 2013-02-19 2017-05-30 The Regents Of The University Of Colorado Compositions comprising eicosapentaenoic acid and a hydroxyl compound and methods of use thereof
WO2014158256A1 (en) * 2013-03-13 2014-10-02 Matinas Biopharma Inc. Omega-3 pentaenoic acid compositions and methods of use
US9283201B2 (en) 2013-03-14 2016-03-15 Amarin Pharmaceuticals Ireland Limited Compositions and methods for treating or preventing obesity in a subject in need thereof
US10808521B2 (en) 2013-05-31 2020-10-20 Conocophillips Company Hydraulic fracture analysis
EP3140316A1 (en) 2014-05-05 2017-03-15 Thetis Pharmaceuticals LLC Compositions and methods relating to ionic salts of peptides
US9242008B2 (en) 2014-06-18 2016-01-26 Thetis Pharmaceuticals Llc Mineral amino-acid complexes of fatty acids
AU2015277509A1 (en) 2014-06-18 2017-01-05 Thetis Pharmaceuticals Llc Mineral amino-acid complexes of active agents
CA2961148A1 (en) * 2014-09-25 2016-03-31 Astrazeneca Ab Use of a combination of an omega-3 fatty acid and an sglt-2 inhibitor for the treatment of non-alcoholic liver disease
WO2016138959A1 (en) 2015-03-05 2016-09-09 Volvo Car Corporation Estimation of battery parameters
WO2016150936A1 (en) * 2015-03-26 2016-09-29 Tiberio Bruzzese Purified compositions of polyunsaturated fatty acids, their preparation method and their use
US10890058B2 (en) 2016-03-09 2021-01-12 Conocophillips Company Low-frequency DAS SNR improvement
EP3454907B1 (en) 2016-06-03 2020-07-22 Thetis Pharmaceuticals LLC Compositions and methods relating to salts of specialized pro-resolving mediators of inflammation
US11255997B2 (en) 2017-06-14 2022-02-22 Conocophillips Company Stimulated rock volume analysis
EP3619560B1 (en) 2017-05-05 2022-06-29 ConocoPhillips Company Stimulated rock volume analysis
WO2019079481A2 (en) * 2017-10-17 2019-04-25 Conocophillips Company Low frequency distributed acoustic sensing hydraulic fracture geometry
WO2019191106A1 (en) 2018-03-28 2019-10-03 Conocophillips Company Low frequency das well interference evaluation
US11021934B2 (en) 2018-05-02 2021-06-01 Conocophillips Company Production logging inversion based on DAS/DTS
EP3947905A4 (en) 2019-03-25 2022-11-30 ConocoPhillips Company Machine-learning based fracture-hit detection using low-frequency das signal
AU2022310512A1 (en) 2021-07-16 2024-01-25 Conocophillips Company Passive production logging instrument using heat and distributed acoustic sensing

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080085911A1 (en) * 2006-10-10 2008-04-10 Reliant Pharmaceuticals, Inc. Statin and omega-3 fatty acids for reduction of apo-b levels
US20110034555A1 (en) * 2009-06-15 2011-02-10 Amarin Pharma , Inc. Compositions and methods for lowering triglycerides without raising ldl-c levels in a subject on concomitant statin therapy

Family Cites Families (63)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB154073A (en) 1919-12-03 1920-11-25 Bertram Harold Cope A new or improved jig or appliance for use in the attachment of cylinder and the like locks and latches
GB8819110D0 (en) 1988-08-11 1988-09-14 Norsk Hydro As Antihypertensive drug & method for production
GB9217781D0 (en) 1992-08-21 1992-10-07 Efamol Holdings Fatty acid treatment
CA2119000A1 (en) * 1993-03-19 1994-09-20 David Frederick Horrobin Formulation for use in smokers
MY118354A (en) * 1995-05-01 2004-10-30 Scarista Ltd 1,3-propane diol derivatives as bioactive compounds
US5861399A (en) 1996-07-17 1999-01-19 Heart Care Partners Methods and compositions for the rapid and enduring relief of inadequate myocardial function
EP0843972B1 (en) * 1996-11-20 2002-07-31 N.V. Nutricia Nutritional composition comprising fats for the treatment of the metabolic syndrome
US6107334A (en) * 1998-02-23 2000-08-22 Wake Forest University Dietary control of arachidonic acid metabolism
GB0016045D0 (en) 2000-06-29 2000-08-23 Laxdale Limited Therapeutic combinations of fatty acids
ITMI20020731A1 (en) * 2002-04-08 2003-10-08 Ibsa Inst Biochimique Sa PHARMACEUTICAL COMPOSITIONS FOR ACETYLSALICYLIC ACID AND OMEGA-3 OILS
GB0301701D0 (en) 2003-01-24 2003-02-26 Ensay Ltd Psoriasis and Eicosapentaenoic acid
WO2004098311A1 (en) 2003-05-05 2004-11-18 Denofa As Fish oils with an altered fatty acid profile, method of producing same and their use
JP4870430B2 (en) 2003-06-20 2012-02-08 持田製薬株式会社 Composition for prevention and treatment of varicose veins
EP1656839A1 (en) 2004-11-11 2006-05-17 N.V. Nutricia Nutrition containing lipid blend
JP2008520739A (en) 2004-11-19 2008-06-19 マーテック・バイオサイエンシーズ・コーポレーション Oxylipins derived from long-chain polyunsaturated fatty acids and methods for their preparation and use
FR2878747B1 (en) 2004-12-03 2007-03-30 Pierre Fabre Medicament Sa USE OF OMEGA-3 FATTY ACID (S) FOR THE TREATMENT OF HYPERCHOLESTEROLEMIA CAUSED BY ANTI-RETROVIRAL TREATMENT IN HIV INFECTED PATIENTS
CN101098690A (en) 2004-12-06 2008-01-02 瑞莱恩特医药品有限公司 Omega-3 fatty acids and dyslipidemic agent for lipid therapy
US20060188529A1 (en) 2004-12-06 2006-08-24 George Bobotas Stable compositions of fenofibrate with fatty acid esters
US20070191467A1 (en) * 2004-12-06 2007-08-16 Reliant Pharmaceutical, Inc. Statin and omega-3 fatty acids for lipid therapy
US20060211762A1 (en) * 2004-12-06 2006-09-21 Rongen Roelof M Omega-3 fatty acids and dyslipidemic agent for lipid therapy
US20090239927A1 (en) 2004-12-06 2009-09-24 George Bobotas Statin and Omega-3 Fatty Acids For Lipid Therapy
US20080200707A1 (en) 2005-01-04 2008-08-21 Mochida-Pharmaceuticals Pharmaceutical Co., Ltd. Lipotoxicity Relieving Agent
EA200701913A1 (en) 2005-03-08 2008-08-29 Релайэнт Фармасьютикалз, Инк. TREATMENT WITH STATIN, OMEGA-3 FATTY ACIDS AND THEIR COMBINED PRODUCT
EP1714564A1 (en) * 2005-04-21 2006-10-25 N.V. Nutricia Method for treatment and prevention of respiratory insufficiency
EP1888727B1 (en) 2005-05-04 2015-04-15 Pronova BioPharma Norge AS New dha derivatives and their use as medicaments
TWI412361B (en) 2005-07-08 2013-10-21 Mochida Pharm Co Ltd Composition for preventing onset of cardiovascular events
AU2006270047A1 (en) 2005-07-18 2007-01-25 Reliant Pharmaceuticals, Inc. Treatment with azetidinone-based cholesterol absorption inhibitors and omega-3 fatty acids and a combination product thereof
JP2009502950A (en) 2005-07-28 2009-01-29 レリアント ファーマスーティカルズ インコーポレイテッド Therapies using dihydropyridine calcium channel blockers and ω3 fatty acids, and mixed products thereof
US20070104779A1 (en) 2005-11-07 2007-05-10 Rongen Roelof M Treatment with omega-3 fatty acids and products thereof
WO2007058523A1 (en) 2005-11-17 2007-05-24 N.V. Nutricia Composition with docosapentaenoic acid
US20090286879A1 (en) 2005-12-02 2009-11-19 Kiyoshi Hashizume Preventive or Therapeutic Drug for Alzheimer-Type Dementia
US20070265340A1 (en) 2006-01-05 2007-11-15 Shalwitz Robert A Treatment of fatty liver
EP2526937A1 (en) 2006-02-07 2012-11-28 Mochida Pharmaceutical Co., Ltd. Composition for prevention of recurrence of stroke
WO2008004900A1 (en) 2006-07-05 2008-01-10 Photonz Corporation Limited Production of ultrapure epa and polar lipids from largely heterotrophic culture
KR100684642B1 (en) * 2006-09-14 2007-02-22 주식회사 일신웰스 Glyceride oil composition from fish oil and preparation method thereof
CA2667211A1 (en) 2006-11-01 2008-05-08 Pronova Biopharma Norge As Alpha-substituted omega-3 lipids that are activators or modulators of the peroxisome proliferators-activated receptor (ppar)
WO2008113177A1 (en) 2007-03-20 2008-09-25 Centre De Recherche Sur Les Biotechnologies Marines Compositions comprising polyunsaturated fatty acid monoglycerides or derivatives thereof and uses thereof
WO2008115529A1 (en) * 2007-03-20 2008-09-25 Reliant Pharmaceuticals, Inc. Compositions comprising omega-3 fatty acids and cetp inhibitors
US20090264520A1 (en) 2008-04-21 2009-10-22 Asha Lipid Sciences, Inc. Lipid-containing compositions and methods of use thereof
JPWO2009142242A1 (en) 2008-05-20 2011-09-29 持田製薬株式会社 Composition for preventing cardiovascular events in high-risk patients
US20100021573A1 (en) 2008-07-22 2010-01-28 Michael J Gonzalez Compositions and methods for the prevention of cardiovascular disease
PL2395991T3 (en) 2009-02-10 2013-11-29 Amarin Pharmaceuticals Ie Ltd Use of eicosapentaenoic acid ethyl ester for treating hypertriglyceridemia
CA2754860C (en) * 2009-03-09 2020-02-25 Pronova Biopharma Norge As Compositions comprising a fatty acid oil mixture and a surfactant, and methods and uses thereof
US20110071090A1 (en) * 2009-03-11 2011-03-24 Stable Solutions Llc Method of mitigating adverse drug events using omega-3-fatty acids as a parenteral therapeutic drug vehicle
CN106822080A (en) * 2009-04-29 2017-06-13 阿马里纳药物爱尔兰有限公司 Pharmaceutical composition containing EPA and cardiovascular agents and use its method
EP3181131A1 (en) 2009-05-22 2017-06-21 Mochida Pharmaceutical Co., Ltd. Self-emulsifying compositin of 3 fatty acid
US20110177061A1 (en) 2009-07-10 2011-07-21 Martek Biosciences Corporation Methods of treating and preventing neurological disorders using docosahexaenoic acid
NZ600167A (en) * 2009-10-29 2014-06-27 Acasti Pharma Inc Concentrated therapeutic phospholipid compositions
WO2011060492A1 (en) * 2009-11-18 2011-05-26 The University Of Sydney Combination for treating metabolic disorders
US20110178105A1 (en) * 2010-01-15 2011-07-21 E.I. Du Pont De Nemours And Company Clinical benefits of eicosapentaenoic acid in humans
GB201001345D0 (en) 2010-01-27 2010-03-17 Equateq Ltd Process for preparing and purifying fatty acids
EP2542060B1 (en) * 2010-03-04 2019-10-02 Amarin Pharmaceuticals Ireland Limited Compositions for treating and/or preventing cardiovascular disease
ES2710540T5 (en) 2010-06-30 2022-08-26 Mochida Pharm Co Ltd Omega-3 fatty acid compound preparation
US20120302639A1 (en) 2011-02-16 2012-11-29 Pivotal Therapeutics Inc. Omega 3 formulations for treatment of risk factors for cardiovascular disease and protection against sudden death
US8951514B2 (en) 2011-02-16 2015-02-10 Pivotal Therapeutics Inc. Statin and omega 3 fatty acids for reduction of apolipoprotein-B levels
JP2014505732A (en) 2011-02-16 2014-03-06 ピヴォタル セラピューティクス インコーポレイテッド Statins and omega-3 fatty acids (EPA, DHA and DPA) for use in cardiovascular disease
US20120264824A1 (en) 2011-04-15 2012-10-18 Mochida Pharmaceutical Co., Ltd. Compositions and methods for treating non-alcoholic steatohepatitis
US8846604B2 (en) 2011-09-02 2014-09-30 Artic Nutrition AS Lipid compositions with high DHA content
US20130095179A1 (en) 2011-09-15 2013-04-18 Omthera Pharmaceuticals, Inc. Methods and compositions for treating, reversing, inhibiting or preventing resistance to antiplatelet therapy
US20150005382A1 (en) * 2011-10-04 2015-01-01 Wake Forest University Health Sciences (WFUHS) Methods for Identifying and Treating an Individual with an Inflammatory Disease using Fatty Acid-Based Therapies
RU2018116572A (en) 2012-01-06 2018-10-23 Омтера Фармасьютикалс, Инк. DPA Enriched Omega-3 Polyunsaturated Fatty Acids Free Acid Formulations
EP2861227A4 (en) * 2012-06-17 2016-01-27 Matinas Biopharma Inc Omega-3 pentaenoic acid compositions and methods of use
CN102755350B (en) * 2012-08-01 2015-07-01 浙江金诺康生物制药有限公司 Refined seal oil and application thereof to preparation of medicament for treating non-alcoholic fatty liver

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080085911A1 (en) * 2006-10-10 2008-04-10 Reliant Pharmaceuticals, Inc. Statin and omega-3 fatty acids for reduction of apo-b levels
US20110034555A1 (en) * 2009-06-15 2011-02-10 Amarin Pharma , Inc. Compositions and methods for lowering triglycerides without raising ldl-c levels in a subject on concomitant statin therapy

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
http://www.hopkinsvasculitis.org/types-vasculitis/buergers-disease/ *
National Institute of Arthritis and Musculoskeletal and Skin diseases, 2016(http://www.niams.nih.gov/health_info/raynauds_phenomenon/raynauds_phenomenon_ff.pdf) *

Cited By (64)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11103477B2 (en) 2009-04-29 2021-08-31 Amarin Pharmaceuticals Ireland Limited Stable pharmaceutical composition and methods of using same
US11033523B2 (en) 2009-04-29 2021-06-15 Amarin Pharmaceuticals Ireland Limited Pharmaceutical compositions comprising EPA and a cardiovascular agent and methods of using the same
US10987331B2 (en) 2009-04-29 2021-04-27 Amarin Pharmaceuticals Ireland Limited Methods of treating mixed dyslipidemia
US11147787B2 (en) 2009-04-29 2021-10-19 Amarin Pharmaceuticals Ireland Limited Stable pharmaceutical composition and methods of using same
US11154526B2 (en) 2009-04-29 2021-10-26 Amarin Pharmaceuticals Ireland Limited Methods of treating mixed dyslipidemia
US10449172B2 (en) 2009-04-29 2019-10-22 Amarin Pharmaceuticals Ireland Limited Stable pharmaceutical composition and methods of using same
US10940131B2 (en) 2009-04-29 2021-03-09 Amarin Pharmaceuticals Ireland Limited Methods of treating mixed dyslipidemia
US11213504B2 (en) 2009-04-29 2022-01-04 Amarin Pharmaceuticals Ireland Limited Stable pharmaceutical composition and methods of using same
US10888537B2 (en) 2009-04-29 2021-01-12 Amarin Pharmaceuticals Ireland Limited Pharmaceutical compositions comprising omega-3 fatty acids
US10881632B2 (en) 2009-04-29 2021-01-05 Amarin Pharmaceuticals Ireland Limited Stable pharmaceutical composition and methods of using same
US10842766B2 (en) 2009-04-29 2020-11-24 Amarin Pharmaceuticals Ireland Limited Stable pharmaceutical composition and methods of using same
US10792267B2 (en) 2009-04-29 2020-10-06 Amarin Pharmaceuticals Ireland Limited Methods of treating mixed dyslipidemia
US11400069B2 (en) 2009-04-29 2022-08-02 Amarin Pharmaceuticals Ireland Limited Methods of treating mixed dyslipidemia
US11690820B2 (en) 2009-04-29 2023-07-04 Amarin Pharmaceuticals Ireland Limited Methods of treating mixed dyslipidemia
US10624870B2 (en) 2009-04-29 2020-04-21 Amarin Pharmaceuticals Ireland Limited Methods of treating mixed dyslipidemia
US11439618B2 (en) 2009-06-15 2022-09-13 Amarin Pharmaceuticals Ireland Limited Compositions and methods for lowering triglycerides
US11464757B2 (en) 2009-06-15 2022-10-11 Amarin Pharmaceuticals Ireland Limited Compositions and methods for lowering triglycerides
US10842768B2 (en) 2009-06-15 2020-11-24 Amarin Pharmaceuticals Ireland Limited Compositions and methods for lowering triglycerides
US10493058B2 (en) 2009-09-23 2019-12-03 Amarin Pharmaceuticals Ireland Limited Pharmaceutical composition comprising omega-3 fatty acid and hydroxy-derivative of a statin and methods of using same
US11007173B2 (en) 2009-09-23 2021-05-18 Amarin Pharmaceuticals Ireland Limited Pharmaceutical composition comprising omega-3 fatty acid and hydroxy-derivative of a statin and methods of using same
US11712428B2 (en) 2010-11-29 2023-08-01 Amarin Pharmaceuticals Ireland Limited Low eructation composition and methods for treating and/or preventing cardiovascular disease in a subject with fish allergy/hypersensitivity
US11712429B2 (en) 2010-11-29 2023-08-01 Amarin Pharmaceuticals Ireland Limited Low eructation composition and methods for treating and/or preventing cardiovascular disease in a subject with fish allergy/hypersensitivity
US10632094B2 (en) 2011-11-07 2020-04-28 Amarin Pharmaceuticals Ireland Limited Methods of treating hypertriglyceridemia
US10537544B2 (en) 2011-11-07 2020-01-21 Amarin Pharmaceuticals Ireland Limited Methods of treating hypertriglyceridemia
US11291643B2 (en) 2011-11-07 2022-04-05 Amarin Pharmaceuticals Ireland Limited Methods of treating hypertriglyceridemia
US9050309B2 (en) 2012-01-06 2015-06-09 Omthera Pharmaceuticals, Inc. DPA-enriched compositions of omega-3 polyunsaturated fatty acids in free acid form
US10117844B2 (en) 2012-01-06 2018-11-06 Omthera Pharmaceuticals, Inc. DPA-enriched compositions of omega-3 polyunsaturated fatty acids in free acid form
US10973796B2 (en) 2012-01-06 2021-04-13 Amarin Pharmaceuticals Ireland Limited Compositions and methods for lowering levels of high-sensitivity C-reactive protein (hs-CRP) in a subject
US9050308B2 (en) 2012-01-06 2015-06-09 Omthera Pharmaceuticals, Inc. DPA-enriched compositions of omega-3 polyunsaturated fatty acids in free acid form
US9492545B2 (en) 2012-05-07 2016-11-15 Omthera Pharmaceuticals Inc. Compositions of statins and omega-3 fatty acids
US10555924B2 (en) 2012-06-29 2020-02-11 Amarin Pharmaceuticals Ireland Limited Methods of reducing the risk of a cardiovascular event in a subject at risk for cardiovascular disease
US10894028B2 (en) 2012-06-29 2021-01-19 Amarin Pharmaceuticals Ireland Limited Methods of reducing the risk of a cardiovascular event in a subject at risk for cardiovascular disease
US10555925B1 (en) 2012-06-29 2020-02-11 Amarin Pharmaceuticals Ireland Limited Methods of reducing the risk of a cardiovascular event in a subject at risk for cardiovascular disease
US10792270B2 (en) 2012-06-29 2020-10-06 Amarin Pharmaceuticals Ireland Limited Methods of reducing the risk of a cardiovascular event in a subject at risk for cardiovascular disease
US10568861B1 (en) 2012-06-29 2020-02-25 Amarin Pharmaceuticals Ireland Limited Methods of reducing the risk of a cardiovascular event in a subject at risk for cardiovascular disease
US10576054B1 (en) 2012-06-29 2020-03-03 Amarin Pharmaceuticals Ireland Limited Methods of reducing the risk of a cardiovascular event in a subject at risk for cardiovascular disease
US11179362B2 (en) 2012-11-06 2021-11-23 Amarin Pharmaceuticals Ireland Limited Compositions and methods for lowering triglycerides without raising LDL-C levels in a subject on concomitant statin therapy
US11229618B2 (en) 2012-11-06 2022-01-25 Amarin Pharmaceuticals Ireland Limited Compositions and methods for lowering triglycerides without raising LDL-C levels in a subject on concomitant statin therapy
US11141399B2 (en) 2012-12-31 2021-10-12 Amarin Pharmaceuticals Ireland Limited Methods of treating or preventing nonalcoholic steatohepatitis and/or primary biliary cirrhosis
US10675263B2 (en) 2013-02-06 2020-06-09 Amarin Pharmaceuticals Ireland Limited Methods of reducing apolipoprotein C-III
US10610508B2 (en) 2013-02-06 2020-04-07 Amarin Pharmaceuticals Ireland Limited Methods of reducing apolipoprotein C-III
US10973797B2 (en) 2013-02-06 2021-04-13 Amarin Pharmaceuticals Ireland Limited Methods of reducing apolipoprotein c-III
US11185525B2 (en) 2013-02-06 2021-11-30 Amarin Pharmaceuticals Ireland Limited Methods of reducing apolipoprotein C-III
US10851374B2 (en) 2013-02-13 2020-12-01 Amarin Pharmaceuticals Ireland Limited Compositions comprising eicosapentaenoic acid and mipomersen and methods of use thereof
US11547710B2 (en) 2013-03-15 2023-01-10 Amarin Pharmaceuticals Ireland Limited Pharmaceutical composition comprising eicosapentaenoic acid and derivatives thereof and a statin
US10966968B2 (en) 2013-06-06 2021-04-06 Amarin Pharmaceuticals Ireland Limited Co-administration of rosiglitazone and eicosapentaenoic acid or a derivative thereof
US10888539B2 (en) 2013-09-04 2021-01-12 Amarin Pharmaceuticals Ireland Limited Methods of treating or preventing prostate cancer
US10722485B2 (en) 2013-10-10 2020-07-28 Amarin Pharmaceuticals Ireland Limited Compositions and methods for lowering triglycerides without raising LDL-C levels in a subject on concomitant statin therapy
US11285127B2 (en) 2013-10-10 2022-03-29 Amarin Pharmaceuticals Ireland Limited Compositions and methods for lowering triglycerides without raising LDL-C levels in a subject on concomitant statin therapy
US11052063B2 (en) 2014-06-11 2021-07-06 Amarin Pharmaceuticals Ireland Limited Methods of reducing RLP-C
US10561631B2 (en) 2014-06-11 2020-02-18 Amarin Pharmaceuticals Ireland Limited Methods of reducing RLP-C
US11446269B2 (en) 2014-06-16 2022-09-20 Amarin Pharmaceuticals Ireland Limited Methods of reducing or preventing oxidation of small dense LDL or membrane polyunsaturated fatty acids
US10842765B2 (en) 2016-03-15 2020-11-24 Amarin Pharmaceuticals Ireland Limited Methods of reducing or preventing oxidation of small dense ldl or membrane polyunsaturated fatty acids
US10966951B2 (en) * 2017-05-19 2021-04-06 Amarin Pharmaceuticals Ireland Limited Compositions and methods for lowering triglycerides in a subject having reduced kidney function
US20180333383A1 (en) * 2017-05-19 2018-11-22 Amarin Pharmaceuticals Ireland Limited Compositions and Methods for Lowering Triglycerides in a Subject Having Reduced Kidney Function
US11058661B2 (en) 2018-03-02 2021-07-13 Amarin Pharmaceuticals Ireland Limited Compositions and methods for lowering triglycerides in a subject on concomitant statin therapy and having hsCRP levels of at least about 2 mg/L
US11298333B1 (en) 2018-09-24 2022-04-12 Amarin Pharmaceuticals Ireland Limited Methods of reducing the risk of cardiovascular events in a subject
US11369582B2 (en) 2018-09-24 2022-06-28 Amarin Pharmaceuticals Ireland Limited Methods of reducing the risk of cardiovascular events in a subject
US10786478B2 (en) 2018-09-24 2020-09-29 Amarin Pharmaceuticals Ireland Limited Methods of reducing the risk of cardiovascular events in a subject
US10668042B2 (en) 2018-09-24 2020-06-02 Amarin Pharmaceuticals Ireland Limited Methods of reducing the risk of cardiovascular events in a subject
US11000499B2 (en) 2018-09-24 2021-05-11 Amarin Pharmaceuticals Ireland Limited Methods of reducing the risk of cardiovascular events in a subject
US11116743B2 (en) 2018-09-24 2021-09-14 Amarin Pharmaceuticals Ireland Limited Methods of reducing the risk of cardiovascular events in a subject
US11116742B2 (en) 2018-09-24 2021-09-14 Amarin Pharmaceuticals Ireland Limited Methods of reducing the risk of cardiovascular events in a subject
US11717504B2 (en) 2018-09-24 2023-08-08 Amarin Pharmaceuticals Ireland Limited Methods of reducing the risk of cardiovascular events in a subject

Also Published As

Publication number Publication date
CA2916208A1 (en) 2013-12-27
US20140094520A1 (en) 2014-04-03
US20140107206A1 (en) 2014-04-17
US20140100273A1 (en) 2014-04-10
US20140100275A1 (en) 2014-04-10
US20210205254A1 (en) 2021-07-08
US20140249226A1 (en) 2014-09-04
WO2013192109A1 (en) 2013-12-27
US8906964B2 (en) 2014-12-09
US20140107198A1 (en) 2014-04-17
US20140107200A1 (en) 2014-04-17
US20140107205A1 (en) 2014-04-17
US20140100272A1 (en) 2014-04-10
AU2013277441A1 (en) 2015-01-29
EP2861227A1 (en) 2015-04-22
US20140100281A1 (en) 2014-04-10
US10058521B2 (en) 2018-08-28
US20140100274A1 (en) 2014-04-10
AU2013277441B2 (en) 2017-07-06
EP2861227A4 (en) 2016-01-27

Similar Documents

Publication Publication Date Title
US20210205254A1 (en) Omega-3 pentaenoic acid compositions and methods of use
US20140322314A1 (en) Omega-3 Fatty Acid Formulations for Use as Pharmaceutical Treatment
AU2020203658A1 (en) Administering compositions comprising docosapentaenoic acid
CA2905671A1 (en) Omega-3 pentaenoic acid compositions and methods of use
WO2014179341A1 (en) Treatment with omega-3 fatty acid compositions
US20140194512A1 (en) Compositions comprising docosapentaenoic acid and methods of use

Legal Events

Date Code Title Description
AS Assignment

Owner name: MATINAS BIOPHARMA INC., NEW JERSEY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:FAWZY, ABDEL AZIZ;BOBOTAS, GEORGE;RONGEN, ROELOF;SIGNING DATES FROM 20131216 TO 20131217;REEL/FRAME:031824/0441

STCB Information on status: application discontinuation

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