EP2929041A1 - Omega-3-pentaensäure-zusammensetzungen und verfahren zur verwendung - Google Patents

Omega-3-pentaensäure-zusammensetzungen und verfahren zur verwendung

Info

Publication number
EP2929041A1
EP2929041A1 EP13861394.8A EP13861394A EP2929041A1 EP 2929041 A1 EP2929041 A1 EP 2929041A1 EP 13861394 A EP13861394 A EP 13861394A EP 2929041 A1 EP2929041 A1 EP 2929041A1
Authority
EP
European Patent Office
Prior art keywords
alternatively
dha
dpa
epa
omega
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.)
Withdrawn
Application number
EP13861394.8A
Other languages
English (en)
French (fr)
Other versions
EP2929041A4 (de
Inventor
George Bobotas
Abdel Aziz Fawzy
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 EP21170003.4A priority Critical patent/EP3888646A1/de
Publication of EP2929041A1 publication Critical patent/EP2929041A1/de
Publication of EP2929041A4 publication Critical patent/EP2929041A4/de
Withdrawn legal-status Critical Current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
    • A61K31/202Carboxylic 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
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23DEDIBLE OILS OR FATS, e.g. MARGARINES, SHORTENINGS, COOKING OILS
    • A23D9/00Other edible oils or fats, e.g. shortenings, cooking oils
    • A23D9/06Preservation of finished products
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23DEDIBLE OILS OR FATS, e.g. MARGARINES, SHORTENINGS, COOKING OILS
    • A23D9/00Other edible oils or fats, e.g. shortenings, cooking oils
    • 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
    • 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
    • A61K9/4875Compounds of unknown constitution, e.g. material from plants or animals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C11ANIMAL OR VEGETABLE OILS, FATS, FATTY SUBSTANCES OR WAXES; FATTY ACIDS THEREFROM; DETERGENTS; CANDLES
    • C11BPRODUCING, e.g. BY PRESSING RAW MATERIALS OR BY EXTRACTION FROM WASTE MATERIALS, REFINING OR PRESERVING FATS, FATTY SUBSTANCES, e.g. LANOLIN, FATTY OILS OR WAXES; ESSENTIAL OILS; PERFUMES
    • C11B5/00Preserving by using additives, e.g. anti-oxidants
    • C11B5/0007Organic substances
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/22Heterocyclic compounds, e.g. ascorbic acid, tocopherol or pyrrolidones

Definitions

  • the present invention relates to a method comprising administering omega- 3 fatty acid compositions for the reduction of fasting lipid parameters, such as triglycerides, total cholesterol, low density lipoprotein (LDL) cholesterol, free fatty acids, and other lipids.
  • the present invention also relates to a method comprising administration of omega-3 fatty acid compositions for the increase of high density lipoprotein (HDL) cholesterol.
  • fasting lipid parameters such as triglycerides, total cholesterol, low density lipoprotein (LDL) cholesterol, free fatty acids, and other lipids.
  • LDL low density lipoprotein
  • HDL high density lipoprotein
  • the methods of the present invention may be useful of the treatment of a condition selected from the group consisting of: hypertriglyceridemia; hypercholesterolemia; mixed dyslipidemia; coronary heart disease (CHD); vascular disease; cardiovascular disease; acute coronary syndrome; atherosclerotic disease and related conditions; heart failure; cardiac arrhythmias; coagulatory conditions associated with cardiac arrhythmias; ischemic dementia; vascular dementia; hypertension; coagulation related disorders; nephropathy; kidney or urinary tract disease; retinopathy; cognitive and other CNS disorders; autoimmune diseases; inflammatory diseases; asthma or other respiratory disease; dermatological disease; metabolic syndrome; diabetes, diabetes mellitis or other form of metabolic disease; liver disease; non-alcoholic fatty liver disease; disease of the gastrointestinal tract; disease of the male or female reproductive system or related secondary sexual organs; a cancer of any type, including lymphomas and myelomas; an infection caused by a virus, bacterium, fungus, protozoa or other organism; and the treatment and/
  • cholesterol and triglycerides are part of lipoprotein complexes in the bloodstream, and can be separated via ultracentrifugation into high-density lipoprotein (HDL), intermediate-density lipoprotein (IDL), low-density lipoprotein (LDL) and very-low-density lipoprotein (VLDL) fractions.
  • HDL high-density lipoprotein
  • IDL intermediate-density lipoprotein
  • LDL low-density lipoprotein
  • VLDL very-low-density lipoprotein
  • Total-C total cholesterol
  • LDL-cholesterol LDL-cholesterol
  • apolipoprotein B a membrane complex for LDL-cholesterol and VLDL-cholesterol, as well as IDL- cholesterol in rare individuals suffering from a disorder resulting in significant IDL- cholesterol levels
  • hypercholesterolemia Decreased levels of HDL-cholesterol and its transport complex, apolipoprotein A, as well as elevated levels of apolipoprotein C-lll and serum triglycerides (TG) are also associated with the development of atherosclerosis.
  • cardiovascular morbidity and mortality in humans can vary directly with the level of total-C, LDL-cholesterol and TG and inversely with the level of HDL- cholesterol.
  • non-HDL-cholesterol is an important indicator of hypertriglyceridemia (elevated triglycerides), vascular disease, atherosclerotic disease and related conditions. Therefore, non-HDL- cholesterol and fasting TG reduction has also been specified as a treatment objective in NCEP ATP III.
  • Fasting TG is commonly used as a key measure for TG in lipid management, because it minimizes the confounding factor of TG recently absorbed from meals, including the high variability of the content of meals and high variability of post-meal (post-prandial) spikes in TG.
  • fasting TG levels when we refer to triglycerides or TG.
  • HMG-CoA reductase inhibitors (“statins") as the primary treatment option for hypercholesterolemia.
  • LDL-cholesterol is the primary treatment parameter.
  • Patients with hypercholesteremia or mixed dyslipidemia often present with high blood levels of LDL-cholesterol (i.e. greater than 190 mg/dl) and TG (i.e. levels of 200 mg/dl or higher).
  • 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 Vll-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 500mg/dl_ or higher.
  • EPADEL® omega-3 fatty acid concentrate
  • This product is described as 98% EPA ethyl ester in Lancet (Vol.369; March 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 also consists almost entirely of EPA ethyl ester and is known under its developmental stage name AMR101 or its trade name VASCEPA®.
  • AMR101 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 III clinical development. Subsequently, AMR101 was entered in a development program for hypertriglyceridemia and mixed dyslipidemia.
  • EPANOVATM omega-3, long chain, polyunsaturated fatty acids from fish oil containing approximately 75% DHA and EPA as free fatty acids. 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 III 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.
  • Non-HDL-C -10.2 p ⁇ 0.05 -17.7 p ⁇ 0.0001 -8.1 p .0182
  • VLDL-C -20.3 p ⁇ 0.05 -24.4 p ⁇ 0.0001 -10.5 p 0.0093
  • Non-HDL-C -6.8 pO.0001 -13.6 p ⁇ 0.0001 -5.5 p 0.0054
  • LOVAZA® Under low fat meal conditions, LOVAZA® has only 15% AUC and 12% Cmax of the bioavailability versus LOVAZA® under high fat meal conditions, whereas EPANOVATM under low fat meal conditions has 78% AUC and 53% Cmax of the bioavailability versus LOVAZA® under high fat meal conditions. EPANOVATM under low fat meal conditions has 62% AUC and 46% Cmax of the bioavailability versus EPANOVATM under high fat meal conditions.
  • Omega-3 fatty acids are known to be "essential fatty acids".
  • EFAs 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. Patent 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 polyunsaturated 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. 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
  • omega-3 fatty acid concentrates Given the highly beneficial efficacy and side-effect profile of omega-3 fatty acid concentrates, these compositions are increasingly popular for the treatment of patients with dyslipidemias.
  • omega-3 fatty acid concentrates With the increased popularity of omega-3 fatty acid concentrates, there is an unmet medical need for omega-3 fatty acid containing compositions with improved bioavailability and a more optimal ratio of potency in reducing TG versus the resulting cholesterol profile.
  • agents with both a higher potency than AMR101/EPADEL® and lesser increase in LDL-C or further decrease in LDL-C and non-HDL-C than LOVAZA® are required.
  • Fasting triglyceride levels have been found to be correlated with the risk of cardiovascular diseases and conditions. For example, high fasting triglycerides levels have been associated with an increased risk of myocardial infarction. Gaziano et al. (Circulation, 1997; 96:2520-2525) discusses fasting triglyceride levels as a risk factor for coronary heart disease. Love-Osborne et al. (Pediatr Diabetes, 2006: 7:205-210) discloses the role of elevated fasting triglyceride levels in the development of type 2 diabetes mellitus.
  • the present invention provides 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; 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 DHA
  • the composition comprises 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
  • compositions comprise about 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 present invention also provides compositions having an improved profile versus very high purity EPA compositions which contain low levels of omega-6 fatty acids and low levels of omega-3 fatty acids other than EPA, ETA, HPA, TPA, and DPA.
  • the present invention provides methods of reducing lipid parameters, such as triglyceride levels, in a subject in need thereof, comprising administering to the subject an orally administrable composition comprising 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)
  • FIG. 1 shows the fasting plasma lipid values after seven days of dosing, relating to the study described in Example 34.
  • FIG. 2 shows the fasting plasma insulin levels after 28 days of administration, relating to the study described in Example 34.
  • FIG. 3 shows the relative liver gene expression following 28 days of administration, relating to the study described in Example 34.
  • 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
  • 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.
  • the composition comprises at least about 92% to about 99%, alteratively 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 55% and 100% of total fatty acids, alternatively between 60% and 100%, alternatively between 65% and 100%, alternatively between 70% and 100%, alternatively between 75% and 100%, alternatively between 80% and 100%, alternatively between 85% and 95%, alternatively between 85% and 97%, alternatively between 88% and 95%, alternatively between 88% and 97%, alternatively between 90% and 95%, alternatively between 90% and 97% of the total amount of fatty acids.
  • 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.
  • 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.
  • 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%; alternative
  • 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.
  • 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.
  • 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.
  • 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.
  • 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
  • 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%
  • 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, 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.
  • 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 alternativelybetween 1 % and 90%, alternatively alternativelybetween 1 % and 85%, alternatively between 1 % and 80%, alternatively alternativelybetween 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%,
  • the compositions comprise DPA in an amount between about 5% to about 15%, alternatively about 6% to about 12%, alternatively about 7% to about 1 1 %, alternatively about 8% to about 10% relative to the total amount of fatty acids present in the composition.
  • the compositions comprise DPA in an amount of about 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 of DPA.
  • compositions of the present invention comprise 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 :
  • 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 may be present in the compositions of this invention at a DHA:EPA ratio of less than 1 :99.
  • a relatively small amount of 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 20% DHA, alternatively no more than 15% DHA, alternatively no more than 12% DHA, alternatively no more than 10% 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 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 1 1 mg/g to about 13 mg/g, alternatively about 12 mg/g to about 13 mg/g of DHA.
  • 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 .
  • the ratio of EPA:HPA is about 85:1.
  • the ratio of EPA:HPA is about 30:1.
  • 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.
  • the ratio of DPA:HPA is about 8:1 .
  • 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 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 :5 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 :15 of DHA:DPA, alternatively no more than
  • 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
  • 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; C20:4-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 (altematively 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-HPA 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-HPA and non-DPA omega- 3 fatty acids, altematively 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, altematively no more than 5% non-EPA, non-ETA, non-HPA and non-DPA omega-3 fatty acids, alternatively no more
  • 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, altematively 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, altematively 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, altematively 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, altematively no more than 1 % of the sum of ALA, SDA and DHA relative to 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.
  • 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 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.
  • 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.
  • 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.
  • 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 are 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 than Tmax for LOVAZA® for EPA, DPA, DHA , EPA+DPA, EPA+DHA, 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 LOVAZA® for EPA, DPA, DHA , EPA+DPA, EPA+DHA, or total omega-3 fatty acids under either low fat meal, fasting, or both administration conditions.
  • Tmax for EPA, DPA, DHA , EPA+DPA, EPA+DHA, or total omega-3 fatty acids are equal or less than 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.
  • compositions of the present invention are better absorbed than LOVAZA® as measured by the maximum concentration (Cmax) in blood, serum or plasma of EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3-pentaenoic acids, or total omega-3 fatty acids.
  • Cmax for EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3-pentaenoic acids, or total omega-3 fatty acids under high fat meal administration conditions are preferably at least 1.1 x (110% of) Cmax for EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3- pentaenoic acids, or total omega-3 fatty acids for LOVAZA®, alternatively at least 1.2 x (120% of), alternatively at least 1.3 x (130% of), alternatively at least 1.4 x (140% of), alternatively at least 1.5 x (150% of) Cmax for EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3-pentaenoic acids, or total omega-3 fatty acids for LOVAZA®.
  • Cmax for EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3-pentaenoic acids, or total omega- 3 fatty acids under low fat meal administration conditions are preferably at least 1.5 x (150% of) Cmax for EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3- pentaenoic acids, or total omega-3 fatty acids for LOVAZA®, alternatively at least 2 x (200% of), alternatively at least 3 x (300% of), alternatively at least 4 x (400% of), alternatively at least 5 x (500% of), alternatively at least 6 x (600% of) Cmax for EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3-pentaenoic acids, or total omega-3 fatty acids for LOVAZA®.
  • Cmax for EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3-pentaenoic acids, or total omega- 3 fatty acids under fasting administration conditions are preferably at least 1.5 x (150% of) Cmax for EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3- pentaenoic acids, or total omega-3 fatty acids for LOVAZA®, alternatively at least 2 x (200% of), alternatively at least 3 x (300% of), alternatively at least 4 x (400% of), alternatively at least 5 x (500% of), alternatively at least 6 x (600% of), alternatively at least 7 x (700% of) Cmax for EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3-pentaenoic acids, or total omega-3 fatty acids for LOVAZA®.
  • compositions of the present invention are better absorbed than LOVAZA® as measured by the area under the concentration curve over time (AUC) in blood, serum or plasma of EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3-pentaenoic acids, or total omega-3 fatty acids.
  • AUC for EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3-pentaenoic acids, or total omega-3 fatty acids under high fat meal administration conditions are preferably at least 1.1 x (110% of) AUC for EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3- pentaenoic acids, or total omega-3 fatty acids for LOVAZA®, alternatively at least 1.2 x (120% of), alternatively at least 1.3 x (130% of), alternatively at least 1.4 x (140% of), alternatively at least 1.5 x (150% of) AUC for EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3-pentaenoic acids, or total omega-3 fatty acids for LOVAZA®.
  • AUC for EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3-pentaenoic acids, or total omega-3 fatty acids under low fat meal administration conditions are preferably at least 1.5 x (150% of) AUC for EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3- pentaenoic acids, or total omega-3 fatty acids for LOVAZA®, alternatively at least 2 x (200% of), alternatively at least 3 x (300% of), alternatively at least 4 x (400% of), alternatively at least 5 x (500% of), alternatively at least 6 x (600% of) AUC for EPA, DPA, DHA , EPA+DPA, EPA+DHA, or total omega-3-pentaenoic acids, or total omega-3 fatty acids for LOVAZA®.
  • AUC for EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3-pentaenoic acids, or total omega- 3 fatty acids under fasting administration conditions are preferably at least 1.5 x (150% of) AUC for EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3- pentaenoic acids, or total omega-3 fatty acids for LOVAZA®, alternatively at least 2 x (200% of), alternatively at least 3 x (300% of), alternatively at least 4 x (400% of), alternatively at least 5 x (500% of), alternatively at least 6 x (600% of), alternatively at least 7 x (700% of) AUC for EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3-pentaenoic acids, or total omega-3 fatty acids for LOVAZA®.
  • compositions of the present invention are better absorbed than AMR101 as measured by the maximum concentration (Cmax) in blood, serum or plasma of EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3-pentaenoic acids, or total omega-3 fatty acids.
  • Cmax for EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3-pentaenoic acids, or total omega-3 fatty acids under high fat meal administration conditions are preferably at least 1.1 x (110% of) Cmax for EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3- pentaenoic acids, or total omega-3 fatty acids for AMR10 , alternatively at least 1 .2 x (120% of), alternatively at least 1.3 x (130% of), alternatively at least 1 .4 x (140% of), alternatively at least 1.5 x (150% of) Cmax for EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3-pentaenoic acids, or total omega-3 fatty acids for AMR101.
  • Cmax for EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3-pentaenoic acids, or total omega- 3 fatty acids under low fat meal administration conditions are preferably at least 1 .5 x (150% of) Cmax for EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3- pentaenoic acids, or total omega-3 fatty acids for AMR101 , alternatively at least 2 x (200% of), alternatively at least 3 x (300% of), alternatively at least 4 x (400% of), alternatively at least 5 x (500% of), alternatively at least 6 x (600% of) Cmax for EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3-pentaenoic acids, or total omega-3 fatty acids for AMR101.
  • Cmax for EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3-pentaenoic acids, or total omega- 3 fatty acids under fasting administration conditions are preferably at least 1.5 x (150% of) Cmax for EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3- pentaenoic acids, or total omega-3 fatty acids for AMR101 , alternatively at least 2 x (200% of), alternatively at least 3 x (300% of), alternatively at least 4 x (400% of), alternatively at least 5 x (500% of), alternatively at least 6 x (600% of), alternatively at least 7 x (700% of) Cmax for EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3-pentaenoic acids, or total omega-3 fatty acids for AMR101 .
  • compositions of the present invention are better absorbed than AMR101 as measured by the area under the concentration curve over time (AUC) in blood, serum or plasma of EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3-pentaenoic acids, or total omega-3 fatty acids.
  • AUC for EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3-pentaenoic acids, or total omega-3 fatty acids under high fat meal administration conditions are preferably at least 1.1 x (1 10% of) AUC for EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3-pentaenoic acids, or total omega-3 fatty acids for AMR101 , alternatively at least 1.2 x (120% of), alternatively at least 1.3 x (130% of), alternatively at least 1 .4 x (140% of), alternatively at least 1 .5 x (150% of) AUC for EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3-pentaenoic acids, or total omega-3 fatty acids for AMR101.
  • AUC for EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3-pentaenoic acids, or total omega-3 fatty acids under low fat meal administration conditions are preferably at least 1.5 x (150% of) AUC for EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3- pentaenoic acids, or total omega-3 fatty acids for AMR101 , alternatively at least 2 x (200% of), alternatively at least 3 x (300% of), alternatively at least 4 x (400% of), alternatively at least 5 x (500% of), alternatively at least 6 x (600% of) AUC for EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3-pentaenoic acids, or total omega- 3 fatty acids for AMR101 .
  • AUC for EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3-pentaenoic acids, or total omega- 3 fatty acids under fasting administration conditions are preferably at least 1.5 x (150% of) AUC for EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3- pentaenoic acids, or total omega-3 fatty acids for AMR101 , alternatively at least 2 x (200% of), alternatively at least 3 x (300% of), alternatively at least 4 x (400% of), alternatively at least 5 x (500% of), alternatively at least 6 x (600% of), alternatively at least 7 x (700% of) AUC for EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3-pentaenoic acids, or total omega-3 fatty acids for AMR101 .
  • compositions of the present invention are better absorbed than EPANOVATM as measured by the maximum concentration (Cmax) in blood, serum or plasma of EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3-pentaenoic acids, or total omega-3 fatty acids.
  • Cmax for EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3-pentaenoic acids, or total omega-3 fatty acids under high fat meal administration conditions are preferably approximately 1.0 x (100% of) Cmax (or non-significant difference) for EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3-pentaenoic acids, or total omega-3 fatty acids for EPANOVATM, alternatively at least 1.05 x (105% of), alternatively at least 1.1 x (110% of), alternatively at least 1.2 x (120% of), alternatively at least 1.3 x (130% of) Cmax for EPA, DPA, DHA, EPA+DPA, EPA+DHA, total omega-3-pentaenoic acids, or total omega-3 fatty acids for EPANOVATM.
  • Cmax for EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3-pentaenoic acids, or total omega- 3 fatty acids under low fat meal administration conditions are preferably approximately 1.0 x (100% of) Cmax (or non-significant difference) for EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3-pentaenoic acids, or total omega-3 fatty acids for EPANOVATM, alternatively at least 1 .05 x (105% of), alternatively at least 1 .1 x (1 10% of), alternatively at least 1.2 x (120% of), alternatively at least 1.3 x (130% of) Cmax for EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3- pentaenoic acids, or total omega-3 fatty acids for EPANOVATM.
  • Cmax for EPA+DHA, total omega-3-pentaenoic acids, and total omega-3 fatty acids under fasting administration conditions are preferably approximately 1.0 x (100% of) Cmax (or non-significant difference) for EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3-pentaenoic acids, or total omega-3 fatty acids for EPANOVATM, alternatively at least 1.05 x (105% of), alternatively at least 1.1 x (110% of), alternatively at least 1.2 x (120% of), alternatively at least 1.3 x (130% of) Cmax for EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3-pentaenoic acids, or total omega-3 fatty acids for EPANOVATM.
  • compositions of the present invention are better absorbed than EPANOVATM as measured by the area under the concentration curve over time (AUC) in blood, serum or plasma of EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3-pentaenoic acids, or total omega-3 fatty acids.
  • AUC for EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3-pentaenoic acids, or total omega-3 fatty acids under high fat meal administration conditions are preferably approximately 1.0 x (100% of) AUC (or non-significant difference) for EPA, DPA, DHA , EPA+DPA, EPA+DHA, or total omega-3-pentaenoic acids, or total omega-3 fatty acids for EPANOVATM, alternatively at least 1 .05 x (105% of), alternatively at least 1.1 x (110% of), alternatively at least 1.2 x (120% of), alternatively at least 1.3 x (130% of) AUC for EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3-pentaenoic acids, or total omega-3 fatty acids for EPANOVATM.
  • AUC for EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3-pentaenoic acids, or total omega- 3 fatty acids under low fat meal administration conditions are preferably approximately 1.0 x (100% of) AUC (or non-significant difference) for EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3-pentaenoic acids, or total omega-3 fatty acids for EPANOVATM, alternatively at least 1.05 x (105% of), alternatively at least 1 .1 x (1 10% of), alternatively at least 1.2 x (120% of), alternatively at least 1.3 x (130% of) AUC for EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3- pentaenoic acids, or total omega-3 fatty acids for EPANOVATM.
  • AUC for EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3-pentaenoic acids, or total omega- 3 fatty acids under fasting administration conditions are preferably approximately 1.0 x (100% of) AUC (or non-significant difference) for EPA, DPA, DHA , EPA+DPA, EPA+DHA, or total omega-3-pentaenoic acids, or total omega-3 fatty acids for EPANOVATM, alternatively at least 1.05 x (105% of), alternatively at least 1.1 x (110% of), alternatively at least 1.2 x (120% of), alternatively at least 1 .3 x (130% of) AUC for EPA, DPA, DHA , EPA+DPA, EPA+DHA, total omega-3-pentaenoic acids, or total omega-3 fatty acids for EPANOVATM.
  • 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 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 symptoms associated with a disease or condition comprising administration of a composition of the present invention.
  • Exemplary diseases or conditions include, but are not limited to: hypertriglyceridemia (for example, by those skilled in the art typically established by assessing fasting triglyceride (TG) levels); hypertriglyceridemia with TG>500mg/dL (VHTG); hypertriglyceridemia with TG 200-499mg/dl_; hypertriglyceridemia with TG 200-499mg/dL while on statin treatment (HTG); hypercholesterolemia; mixed dyslipidemia; coronary heart disease (CHD); vascular disease; atherosclerotic disease and related conditions; heart failure; cardiac arrhythmias; blood coagulatory conditions associated with cardiac arrhytmias; hypertension; coagulation related disorders, including post-surgical deep vein thrombosis or other high risk thrombosis conditions; nephropathy; kidney or urinary tract disease; retinopathy; cognitive, psychiatric, neurological and other CNS disorders, including but not limited to schizophrenia, depression, bipolar
  • Cardiovascular and/or cardiac events may include, but are not limited to: myocardial infarction, ischemic cardiac attack, ischemic attack, acute angina, hospitalization due to acute angina, stroke, transient ischemic cerebral attack, cardiac revascularization, cardiac revascularization with stent placement, carotid artery revascularization, carotid artery revascularization with stent placement, peripheral artery revascularization, peripheral artery revascularization with stent placement, plaque rupture, death due to cardiovascular event, and hospitalization due to cardiovascular event. Cardiovascular and/or cardiac events may also include other events deemed to fall in such catergory by those skilled in the art.
  • the present invention also provides method for increasing high-lipoprotein (HDL) cholesterol levels.
  • HDL high-lipoprotein
  • Fasting lipid levels refer to levels of the lipids in the plasma of a subject after a fasting period, which is a period of about 8 to 12 hours without food.
  • the baseline fasting triglyceride level in the subject prior to administration of acomposition of the present invention is greater than 150 mg/dL. In some embodiments, the baseline fasting triglyceride level is 150 mg/dL to 199 mg/dl, alternatively 200-499 mg/dL, alternatively over 500 mg/dL.
  • the methods of the present invention relate to reducing or decreasing plasma lipid parameters in a subject in need thereof.
  • Lipid parameters include triglycerides, low-density lipoprotein (LDL) cholesterol, total cholesterol, free fatty acids, and non-high-lipoprotein cholesterol (non-HDL) cholesterol.
  • the lipid parameters may be measured in a fasting state or a fed state.
  • the methods comprise administration of EPA and/or DPA in the free fatty acid form or a salt, ester, or derivative form.
  • the methods comprise a reduction in triglyceride levels of at least 10%, alternatively at least 15%, alternatively at least 20%, alternatively at least 25%, alternatively at least 30%, alternatively at least 35%, alternatively at least 40%, alternatively at least 45%, and alternatively at least 50% compared to baseline.
  • the methods comprise a reduction in total cholesterol levels of at least 1 %, alternatively at least 2%, alternatively at least 3%, alternatively at least 4%, alternatively at least 5%, alternatively at least 6%, alternatively at least 7%, alternatively at least 8%, alternatively at least 9%, alternatively at least 10% compared to baseline.
  • the methods comprise a reduction in low-density lipoprotein (LDL) levels of at least 10%, alternatively at least 15%, alternatively at least 20%, alternatively at least 25%, alternatively at least 30%, alternatively at least 35%, alternatively at least 40%, alternatively at least 45%, and alternatively at least 50% compared to baseline.
  • the methods comprise a reduction in free fatty acid levels of at least 5%, alternatively at least 7%, alternatively at least 10%, alternatively at least 15%, alternatively at least 20% compared to baseline.
  • the methods comprise a reduction in non-HDL cholesterol levels of at least 1 %, alternatively at least 2%, alternatively at least 3%, alternatively at least 4%, alternatively at least 5%, alternatively at least 6%, alternatively at least 7%, alternatively at least 8%, alternatively at least 9%, alternatively at least 10%, alternatively at least 20%, alternatively at least 25%, alternatively at least 30% compared to baseline.
  • the methods comprise an increase in high density lipoprotein (HDL) cholesterol levels of at least 1 %, alternatively at least 2%, alternatively at least 3%, alternatively at least 4%, alternatively at least 5%, alternatively at least 6%, alternatively at least 7%, alternatively at least 8%, alternatively at least 9%, alternatively at least 10% compared to baseline.
  • HDL high density lipoprotein
  • this change in lipid parameters can be achieved after a period of daily administration, such as one week, alternatively one month, alternatively two months, alternatively three months or more.
  • the present invention provides methods of treatment for hypertriglyceridemia (either TG ⁇ 500mg/dL, TG>200mg/dL, TG>150mg/dL, TG200- 499mg/dl_, TG300-499mg/dl_, TG350-499mg/dL, or TG150-199mg/dl_), mixed dyslipidemia, or any other diseases or medical conditions as specified above, by dosing to a subject in need thereof omega-3 docosapentaenoic acid (DPA-n3) or its glycerol or ethyl esters.
  • DPA-n3 docosapentaenoic acid
  • Such method of treatment provide a dose of at least 60mg DPA-N3 per day, alternatively at least 80mg DPA-N3 per day, alternatively at least 90mg DPA-N3 per day, alternatively at least 120mg DPA-N3 per day, alternatively at least 150mg DPA-N3 per day, alternatively at least 160mg DPA-N3 per day, alternatively at least 180mg DPA-N3 per day, alternatively at least 200mg DPA-N3 per day, alternatively at least 250mg DPA-N3 per day, alternatively at least 300mg DPA-N3 per day, alternatively at least 350mg DPA-N3 per day, alternatively at least 400mg DPA-N3 per day, alternatively at least 500mg DPA-N3 per day, alternatively at least 600mg DPA-N3 per day, alternatively at least 800mg DPA-N3 or its glycerol or ethyl esters per day.
  • compositions of the present invention which may comprise significant amounts of omega-3 docosapentaenoic acid (DPA-n3) or its glycerol or ethyl esters, may be used for the treatment of hypertriglyceridemia (either TG>500mg/dL, TG>200mg/dL, TG ⁇ 150mg/dL, TG200-499mg/dL, TG300-499mg/dL, TG350-499mg/dL, or TG150-199mg/dL), mixed dyslipidemia, or any other diseases or medical conditions specified above.
  • DPA-n3 docosapentaenoic acid
  • ethyl esters may be used for the treatment of hypertriglyceridemia (either TG>500mg/dL, TG>200mg/dL, TG ⁇ 150mg/dL, TG200-499mg/dL, TG300-499mg/dL, TG350-4
  • Such method of treatment provides to a subject in need thereof a dose of at least 20mg DPA-N3 per day, alternatively at least 25mg DPA-N3 per day, alternatively at least 30mg DPA-N3 per day, alternatively at least 40mg DPA-N3 per day, alternatively at least 50mg DPA-N3 per day, alternatively at least 60mg DPA-N3 per day, alternatively at least 70mg DPA- N3 per day, alternatively at least 80mg DPA-N3 per day, alternatively at least 90mgDPA-N3 per day, alternatively at least 100mg DPA-N3 per day, alternatively at least 120mgDPA-N3 per day, alternatively at least 150mg DPA-N3 per day, alternatively at least 160mg DPA-N3 per day, alternatively at least 180mg DPA-N3 per day, alternatively at least 200mg DPA-N3 per day, alternatively at least 250mg DPA-N3 per day,
  • compositions of the present invention which may comprise significant amounts of omega-3 docosapentaenoic acid (DPA-n3) or its glycerol or ethyl esters and which comprise relatively small amounts of omega-3 docosahexaenoic acid (DHA-n3), may be used for the treatment of hypertriglyceridemia (either TG>500mg/dL, TG>200mg/dL, TG ⁇ 150mg/dl_, TG200- 499mg/dL, TG300-499mg/dL, TG350-499mg/dL, or TG150-199mg/dL), mixed dyslipidemia, or any other diseases or medical conditions specified above.
  • hypertriglyceridemia either TG>500mg/dL, TG>200mg/dL, TG ⁇ 150mg/dl_, TG200- 499mg/dL, TG300-499mg/dL, TG350-499mg/
  • Such method of treatment provides to a subject in need thereof a dose of at least 20mg DPA-N3 per day, alternatively at least 25mg DPA-N3 per day, alternatively at least 30mg DPA-N3 per day, alternatively at least 40mg DPA-N3 per day, alternatively at least 50mg DPA-N3 per day, alternatively at least 60mg DPA-N3 per day, alternatively at least 80mg DPA-N3 per day, alternatively at least 90mg DPA-N3 per day, alternatively at least 120mg DPA-N3 per day, alternatively at least 150mg DPA- N3 per day, alternatively at least 160mg DPA-N3 per day, alternatively at least 180mg DPA-N3 per day, alternatively at least 200mg DPA-N3 per day, alternatively at least 250mg DPA-N3 per day, alternatively at least 300mg DPA-N3 per day, alternatively at least 350mg DPA-N3 per day,
  • compositions of the present invention which may comprise significant amounts of omega-3 docosapentaenoic acid (DPA-n3) or its glycerol or ethyl esters and which comprise relatively small amounts of omega-3 docosahexaenoic acid (DHA-n3), may be used for the treatment of hypertriglyceridemia (either TG ⁇ 500mg/dL, TG ⁇ 200mg/dl_, TG>150mg/dL, TG200- 499mg/dL, TG300-499mg/dl_, TG350-499mg/dL, or TG150-199mg/dL), mixed dyslipidemia, or any other diseases or medical conditions specified above.
  • hypertriglyceridemia either TG ⁇ 500mg/dL, TG ⁇ 200mg/dl_, TG>150mg/dL, TG200- 499mg/dL, TG300-499mg/dl_, TG350-499m
  • Such method of treatment provides to a subject in need thereof a dose of at least 30mg DPA-N3 per day, alternatively at least 40mg DPA-N3 per day, alternatively at least 50mg DPA-N3 per day, alternatively at least 60mg DPA-N3 per day, alternatively at least 80mg DPA-N3 per day, alternatively at least 90mg DPA-N3 per day, alternatively at least 120mg DPA-N3 per day, alternatively at least 150mg DPA-N3 per day, alternatively at least 160mg DPA-N3 per day, alternatively at least 180mg DPA-N3 per day, alternatively at least 200mg DPA-N3 per day, alternatively at least 250mg DPA-N3 per day, alternatively at least 300mg DPA-N3 per day, alternatively at least 350mg DPA-N3 per day, alternatively at least 400mg DPA-N3 per day, alternatively at least 500mg DPA-N3 per day,
  • the present invention provides methods of reducing triglycerides and treating hypertriglyceridemia (either TG>500mg/dL, TG>200mg/dL, TG ⁇ 150mg/dl_, TG200-499mg/dl_, TG300-499mg/dL, TG350-499mg/dl_, or TG150-199mg/dl_), mixed dyslipidemia, or any other diseases or medical conditions as specified above, by dosing to a subject in need thereof omega-3 docosapentaenoic acid (HPA-n3) or its glycerol or ethyl esters.
  • HPA-n3 docosapentaenoic acid
  • Such method of treatment provides a dose of at least 10 mg HPA-N3 per day, alternatively at least 15 mg HPA-N3 per day, alternatively at least 20 mg HPA-N3 per day, alternatively at least 25 mg HPA-N3 per day, alternatively at least 30mg HPA-N3 per day, alternatively at least 40mg HPA-N3 per day, alternatively at least 50mg HPA-N3 per day, alternatively at least 60mg HPA-N3 per day, alternatively at least 70mg HPA-N3 per day, alternatively at least 80mg HPA-N3 per day, alternatively at least 90mg HPA-N3 per day, alternatively at least 100mg HPA-N3 per day, alternatively at least 120mg HPA-N3 per day, alternatively at least 150mg HPA-N3 per day, alternatively at least 160mg HPA-N3 per day, alternatively at least 180mg HPA-N3 per day, alternatively at least 200mg HPA-N
  • compositions of the present invention which may comprise significant amounts of omega-3 heneicosapentaenoic acid (HPA-n3) or its glycerol or ethyl esters, may be used for the treatment of hypertriglyceridemia (either TG>500mg/dL, TG>200mg/dL, TG ⁇ 150mg/dL, TG200-499mg/dl_, TG300- 499mg/dL, TG350-499mg/dL, or TG150-199mg/dL), mixed dyslipidemia, or any other diseases or medical conditions specified above.
  • hypertriglyceridemia either TG>500mg/dL, TG>200mg/dL, TG ⁇ 150mg/dL, TG200-499mg/dl_, TG300- 499mg/dL, TG350-499mg/dL, or TG150-199mg/dL
  • mixed dyslipidemia or any other diseases
  • Such method of treatment provides to a subject in need thereof a dose of at least 10 mg HPA-N3 per day, alternatively at least 15 mg HPA-N3 per day, alternatively at least 20 mg HPA-N3 per day, alternatively at least 25 mg HPA-N3 per day, alternatively at least 30 mg HPA- N3 per day, alternatively at least 40mg HPA-N3 per day, alternatively at least 50mg HPA-N3 per day, alternatively at least 60mg HPA-N3 per day, alternatively at least 70mg HPA-N3 per day, alternatively at least 80mg HPA-N3 per day, alternatively at least 90mg HPA-N3 per day, alternatively at least 100mg HPA-N3 per day, alternatively at least 120mg HPA-N3 per day, alternatively at least 150mg HPA-N3 per day, alternatively at least 160mg HPA-N3 per day, alternatively at least 180mg HPA-N3 per day, alternatively at least 200
  • compositions of the present invention which may comprise significant amounts of omega-3 docosapentaenoic acid (HPA-n3) or its glycerol or ethyl esters and which comprise relatively small amounts of omega-3 docosahexaenoic acid (DHA-n3), may be used for the treatment of hypertriglyceridemia (either TG ⁇ 500mg/dL, TG ⁇ 200mg/dL, TG ⁇ 150mg/dl_, TG200- 499mg/dl_, TG300-499mg/dL, TG350-499mg/dL, or TG150-199mg/dL), mixed dyslipidemia, or any other diseases or medical conditions specified above.
  • hypertriglyceridemia either TG ⁇ 500mg/dL, TG ⁇ 200mg/dL, TG ⁇ 150mg/dl_, TG200- 499mg/dl_, TG300-499mg/dL, TG350-499m
  • Such method of treatment provides to a subject in need thereof a dose of at least 20mg HPA-N3 per day, alternatively at least 25mg HPA-N3 per day, alternatively at least 30mg HPA-N3 per day, alternatively at least 40mg HPA-N3 per day, alternatively at least 50mg HPA-N3 per day, alternatively at least 60mg HPA-N3 per day, alternatively at least 70mg HPA-N3 per day, alternatively at least 80mg HPA-N3 per day, alternatively at least 90mg HPA-N3 per day, alternatively at least 100mg HPA- N3 per day, alternatively at least 120mg HPA-N3 per day, alternatively at least 150mg HPA-N3 per day, alternatively at least 160mg HPA-N3 per day, alternatively at least 180mg HPA-N3 per day, alternatively at least 200mg HPA-N3 per day, alternatively at least 250mg HPA-N3 per day,
  • compositions of the present invention which may comprise significant amounts of omega-3 heneicosapentaenoic acid (HPA-n3) or its glycerol or ethyl esters and which comprise relatively small amounts of omega- 3 docosahexaenoic acid (DHA-n3), may be used for the treatment of hypertriglyceridemia (either TG>500mg/dL, TG>200mg/dL, TG>150mg/dL, TG200- 499mg/dl_, TG300-499mg/dL, TG350-499mg/dL, or TG150-199mg/dl_), mixed dyslipidemia, or any other diseases or medical conditions specified above.
  • hypertriglyceridemia either TG>500mg/dL, TG>200mg/dL, TG>150mg/dL, TG200- 499mg/dl_, TG300-499mg/dL, TG350-4
  • Such method of treatment provides to a subject in need thereof a dose of at least 10mg HPA-N3 per day, alternatively at least 15mg HPA-N3 per day, alternatively at least 20mg HPA-N3 per day, alternatively at least 25mg HPA-N3 per day, alternatively at least 30mg HPA-N3 per day, alternatively at least 40mg HPA-N3 per day, alternatively at least 50mg HPA-N3 per day, alternatively at least 60mg HPA-N3 per day, alternatively at least 70mg HPA-N3 per day, alternatively at least 80mg HPA-N3 per day, alternatively at least 90mg HPA-N3 per day, alternatively at least 100mg HPA-N3 per day, alternatively at least 120mg HPA-N3 per day, alternatively at least 150mg HPA-N3 per day, alternatively at least 160mg HPA-N3 per day, alternatively at least 180mg HPA-N3 per day,
  • the present invention incorporates methods of treatment for hypertriglyceridemia (either TG>500mg/dL, TG>200mg/dL, TG>150mg/dL, TG200- 499mg/dL, TG300-499mg/dL, TG350-499mg/dL, or TG150-199mg/dL), mixed dyslipidemia, or any other diseases or medical conditions as specified above, by dosing to a subject in need thereof omega-3-pentaenoic acids or their glycerol or ethyl esters.
  • Such methods of treatment provides to a subject in need thereof a dose of at least 100mg omega-3- pentaenoic acids per day, alternatively at least 200mg omega-3-pentaenoic acids per day, alternatively at least 300mg omega-3-pentaenoic acids per day, alternatively at least 500mg omega-3-pentaenoic acids per day, alternatively at least 700mg omega-3-pentaenoic acids per day, alternatively at least 900mg omega-3- pentaenoic acids per day, alternatively at least 1000mg omega-3-pentaenoic acids per day, alternatively at least 1500mg omega-3-pentaenoic acids per day, alternatively at least 1900mg omega-3-pentaenoic acids per day, alternatively at least 2000mg omega-3-pentaenoic acids per day, alternatively at least 2500mg omega-3-pentaenoic acids per day, alternatively at least 2900mg omega-3- pentaenoic acids
  • compositions of the present invention which comprise significant amounts of omega-3-pentaenoic acids or their glycerol or ethyl esters and which comprise relatively small amounts of omega-3 docosahexaenoic acid (DHA-n3), may be used for the treatment of hypertriglyceridemia (either TG>500mg/dL, TG>200mg/dL, TG ⁇ 150mg/dl_, TG200-499mg/dL, TG300-499mg/dL, TG350-499mg/dL, or TG150-199mg/dL), mixed dyslipidemia, or any other diseases or medical conditions specified above.
  • hypertriglyceridemia either TG>500mg/dL, TG>200mg/dL, TG ⁇ 150mg/dl_, TG200-499mg/dL, TG300-499mg/dL, TG350-499mg/dL, or TG150-199mg/
  • Such method of treatment provides to a subject in need thereof a dose of at least 100mg omega-3-pentaenoic acids per day, alternatively at least 200mg omega-3-pentaenoic acids per day, alternatively at least 300mg omega-3-pentaenoic acids per day, alternatively at least 500mg omega-3- pentaenoic acids per day, alternatively at least 700mg omega-3-pentaenoic acids per day, alternatively at least 900mg omega-3-pentaenoic acids per day, alternatively at least 1000mg omega-3-pentaenoic acids per day, alternatively at least 1500mg omega-3-pentaenoic acids per day, alternatively at least 1900mg omega-3-pentaenoic acids per day, alternatively at least 2000mg omega-3- pentaenoic acids per day, alternatively at least 2500mg omega-3-pentaenoic acids per day, alternatively at least 2900mg omega-3-pentaenoic acids
  • compositions of the present invention which may comprise significant amounts of omega-3-pentaenoic acids or their glycerol or ethyl esters and which comprise relatively small amounts of omega-3 docosahexaenoic acid (DHA-n3), may be used for the reduction of triglycerides and treatment of hypertriglyceridemia (either TG ⁇ 500mg/dl_, TG>200mg/dL, TG>150mg/dL, TG200- 499mg/dL, TG300-499mg/dL, TG350-499mg/dL, or TG150-199mg/dl_), mixed dyslipidemia, or any other diseases or medical conditions specified above.
  • hypertriglyceridemia either TG ⁇ 500mg/dl_, TG>200mg/dL, TG>150mg/dL, TG200- 499mg/dL, TG300-499mg/dL, TG350-499mg
  • Such method of treatment provides to a subject in need thereof a dose of at least 100mg omega-3-pentaenoic acids per day, alternatively at least 200mg omega-3-pentaenoic acids per day, alternatively at least 300mg omega-3-pentaenoic acids per day, alternatively at least 500mg omega-3-pentaenoic acids per day, alternatively at least 700mg omega-3-pentaenoic acids per day, alternatively at least 900mg omega-3- pentaenoic acids per day, alternatively at least 1000mg omega-3-pentaenoic acids per day, alternatively at least 1500mg omega-3-pentaenoic acids per day, alternatively at least 1900mg omega-3-pentaenoic acids per day, alternatively at least 2000mg omega-3-pentaenoic acids per day, alternatively at least 2500mg omega-3-pentaenoic acids per day, alternatively at least 2900mg omega-3- pentaenoic acids
  • compositions of the present invention which may comprise significant amounts of omega-3-pentaenoic acids or their glycerol or ethyl esters and which comprise relatively small amounts of omega-3 docosahexaenoic acid (DHA-n3), may be used for the reduction of triglycerides and treatment of hypertriglyceridemia (either TG>500mg/dL, TG>200mg/dL, TG>150mg/dl_, TG200-499mg/dl_, TG300-499mg/dL, TG350-499mg/dL, or TG150- 199mg/dl_), mixed dyslipidemia, or any other diseases or medical conditions specified above.
  • hypertriglyceridemia either TG>500mg/dL, TG>200mg/dL, TG>150mg/dl_, TG200-499mg/dl_, TG300-499mg/dL, TG350-499m
  • Such method of treatment provides to a subject in need thereof a dose of at least 100mg omega-3-pentaenoic acids per day, alternatively at least 200mg omega-3-pentaenoic acids per day, alternatively at least 300mg omega-3- pentaenoic acids per day, alternatively at least 500mg omega-3-pentaenoic acids per day, alternatively at least 700mg omega-3-pentaenoic acids per day, alternatively at least 900mg omega-3-pentaenoic acids per day, alternatively at least 1000mg omega-3-pentaenoic acids per day, alternatively at least 1500mg omega-3- pentaenoic acids per day, alternatively at least 1900mg omega-3-pentaenoic acids per day, alternatively at least 2000mg omega-3-pentaenoic acids per day, alternatively at least 2500mg omega-3-pentaenoic acids per day, alternatively at least 2900mg omega-3-pentaenoic acids
  • the improved profile of the compositions of the present invention may be demonstrated upon treatment of a subject by differentially altering the ration 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.
  • the improved profile of the compositions of the present invention may be demonstrated upon treatment of a subject by differentially altering the surface charge of blood platelets and fragments thereof, either in resting state (non-activated platelets) or activated stage.
  • treatment of a subject or patient with compositions of the present invention affect the coagulatory cascade and differentially alter coagulation or bleeding times or platelet aggregation times and density.
  • treatment with compositions of the present invention improves the vascular healing process in response to atherogenic disease.
  • Such healing may be demonstrated by reduced stenosis and/or restenosis over time, reduced or lesser increase in intima-media thickness (IMT) of the arterial wall, larger lumen size and/or larger vascular diameter at vascular sites with stenosis or clot built-up, as determined by either by intravascular ultrasound (IVUS), radiographic, radiologic, non-invasive ultrasound, tomography, magnetic resonance interference (MRI), or other acceptable methods.
  • IVUS intravascular ultrasound
  • MRI magnetic resonance interference
  • such 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.
  • the improved profile resulting from treatment with the compositions of the present invention may also be demonstrated by a differentiated impact on blood/serum/plasma lipid and lipoprotein levels in a mammal; these include, but are not limited to: Triglycerides (TG), total-cholesterol, non-HDL-cholesterol, LDL- cholesterol, VLDL-cholesterol, apolipoprotein B, apolipoprotein A, apolipoprotein C- III, HDL-cholesterol, and Lp-PLA2.
  • TG Triglycerides
  • compositions of the present invention may also be used to provide a beneficial impact on the one or more of the following: apolipoprotein A-l (apo A-l), apolipoprotein B (apo B), apo A-l/apo B ratio, lipoprotein(a) (Lp[a]), lipoprotein-associated phospholipase A2 (Lp-PLA2), low density lipoprotein (LDL) particle number and size, oxidized LDL, C-reactive protein (CRP), high sensitivity C-reactive protein (HSCRP), intracellular adhesion molecule- 1 (ICAM-1 ), E-selectin, P-selectin, vascular cell adhesion molecule 1 (VCAM-1 ) or cluster of differentiation 106 (CD106), interleleukin -1 ⁇ (IL-1 ⁇ ), interleukin-2 (IL-2), interleukin-6 (IL-6), interleukin-8 (IL-8), interleukin-10 (IL-10), interleukin-12 (IL-12), interleuk
  • treatment with the compositions of the present invention is more potent than other omega-3 compositions known in the prior art (such as LOVAZA®, EPANOVATM or AMR101 ) in reducing as compared to placebo or baseline: TG levels, Total-cholesterol levels, non-HDL-cholesterol levels, VLDL- cholesterol levels, LDL-cholesterol levels, apolipoprotein B levels, apolipoprotein C- III levels, Lp-PLA2 levels, or any combinations thereof.
  • omega-3 compositions known in the prior art such as LOVAZA®, EPANOVATM or AMR101
  • such more potent effects in reducing these pararemeters are achieved in patients with baseline TG over 500 mg/dL, in patients on statin treatment with baseline TG in the 200-499 mg/dL range, in patients not on statin treatment with baseline LDL- cholesterol of 190 mg/dL or higher and with TG in the 300-700 mg/dL range, in patients not on statin treatment with baseline LDL-cholesterol of 190 mg/dL or higher and with TG in the 350-700 mg/dL range, in patients not on statin treatment with baseline LDL-cholesterol of 190 mg/dL or higher and with TG in the 300-750 mg/dL range, in patients not on statin treatment with baseline LDL-cholesterol of 190 mg/dL or higher and with TG in the 350-750 mg/dL range, or in patients not on statin treatment with baseline non-HDL-cholesterol of 200 mg/dL or higher and with TG in the 300-700 mg/dL range, or in patients not on statin treatment with baseline
  • treatment with the compositions of the present invention together with statin therapy is more potent than other omega-3 compositions known in the prior art (such as LOVAZA®, EPANOVATM or AMR101 ) in reducing as compared to placebo or baseline: TG levels, Total-cholesterol levels, non-HDL-cholesterol levels, VLDL-cholesterol levels, LDL-cholesterol levelsapolipoprotein B levels, apolipoprotein C-lll levels, Lp-PLA2 levels, or any combinations thereof.
  • omega-3 compositions known in the prior art such as LOVAZA®, EPANOVATM or AMR101
  • such more potent effects in reducing these pararemeters are achieved in patients with baseline TG over 500 mg/dL, in patients on statin treatment with baseline TG in the 200-499 mg/dL range, in patients not on baseline statin treatment with baseline LDL-cholesterol of 190 mg/dL or higher and with TG in the 300-700 mg/dL range, in patients not on baseline statin treatment with baseline LDL-cholesterol of 190 mg/dL or higher and with TG in the 350-700 mg/dL range, in patients not on baseline statin treatment with baseline LDL- cholesterol of 190 mg/dL or higher and with TG in the 300-750 mg/dL range, in patients not on baseline statin treatment with baseline LDL-cholesterol of 190 mg/dL or higher and with TG in the 350-750 mg/dL range, or in patients not on baseline statin treatment with baseline non-HDL-cholesterol of 200 mg/dL or higher and with TG in the 300-700 mg/dL range ,
  • the present invention also provides methods of reducing triglycerides levels in a subject, wherein the non-HDL cholesterol levels, such as LDL-cholesterol levels, of the subject are reduced or not significantly increased from, for example, baseline levels before treatment.
  • treatment with the compositions of the present invention results in a minor (less than 10% change from baseline, alternatively less than 5%) and/or non-significant change in non-HDL cholesterol levels (such as LDL-cholesterol levels) as compared to placebo in patients with baseline TG levels above 500 mg/dL.
  • treatment with the compositions of the present invention results in reductions of LDL-cholesterol levels as compared to placebo in patients with baseline TG levels above 500 mg/dL.
  • the methods involve coadministration of a statin.
  • treatment with the compositions of the present invention as compared to placebo does not increase LDL-cholesterol levels in patients with baseline TG levels of 200-499 mg/dL while on statin therapy.
  • treatment with the compositions of the present invention as compared to placebo results in significant reductions in LDL-cholesterol levels in patients with baseline TG levels of 200-499 mg/dL while on statin therapy.
  • compositions of the present invention as compared to placebo result in significant reductions in LDL-cholesterol levels in patients not on statin treatment with LDL-cholesterol of 190 mg/dL or higher and with TG in the 300-700 mg/dL range, in patients not on statin treatment with LDL- cholesterol of 190 mg/dL or higher and with TG in the 350-700 mg/dL range, in patients not on statin treatment with LDL-cholesterol of 190 mg/dL or higher and with TG in the 300-750 mg/dL range, or in patients not on statin treatment with LDL- cholesterol of 190 mg/dL or higher and with TG in the 350-750 mg/dL range.
  • compositions of the present invention as compared to placebo result in significant reductions in LDL-cholesterol levels in patients not on statin treatment with non-HDL-cholesterol of 200 mg/dL or higher and with TG in the 300-700 mg/dL range, in patients not on statin treatment with Non-HDL-cholesterol of 200 mg/dL or higher and with TG in the 350-700 mg/dL range, in patients not on statin treatment with non-HDL-cholesterol of 200 mg/dL or higher and with TG in the 300-750 mg/dL range, or in patients not on statin treatment with Non-HDL- cholesterol of 200 mg/dL or higher and with TG in the 350-750 mg/dL range.
  • treatment with the compositions of the present invention together with statin therapy results in significant reductions in LDL- cholesterol levels as compared to placebo in patients not on statin treatment at baseline with baseline LDL-cholesterol of 190 mg/dL or higher and with TG in the 300-700 mg/dL range, in patients not on baseline statin treatment with baseline LDL- cholesterol of 190 mg/dL or higher and with TG in the 350-700 mg/dL range, in patients not on baseline statin treatment with baseline LDL-cholesterol of 190 mg/dL or higher and with TG in the 300-750 mg/dL range, or in patients not on baseline statin treatment with baseline LDL-cholesterol of 190 mg/dL or higher and with TG in the 350-750 mg/dL range.
  • treatment with the compositions of the present invention together with statin therapy results in significant reductions in LDL- cholesterol levels as compared to placebo in patients not on baseline statin treatment with baseline non-HDL-cholesterol of 200 mg/dL or higher and with TG in the 300-700 mg/dL range, in patients not on baseline statin treatment with baseline non-HDL-cholesterol of 200 mg/dL or higher and with TG in the 350-700 mg/dL range, in patients not on baseline statin treatment with baseline non-HDL-cholesterol of 200 mg/dL or higher and with TG in the 300-750 mg/dL range, or in patients not on baseline statin treatment with baseline Non-HDL-cholesterol of 200 mg/dL or higher and with TG in the 350-750 mg/dL range.
  • compositions of the present invention are more potent than other omega-3 compositions known in the prior art (such as LOVAZA®, EPANOVATM or AMR101 ) in increasing as compared to placebo or baseline HDL- cholesterol levels, apolipoprotein-A levels, or a combination thereof.
  • compositions of the present invention are more potent than other omega-3 compositions known in the prior art (such as LOVAZA®, EPANOVATM or AMR101 ) in decreasing as compared to placebo or baseline Apolipoprotein-B (Apo-B) levels, Apolipoprotein-CIII levels, Lp-PLA2 levels or any combination thereof.
  • omega-3 compositions known in the prior art such as LOVAZA®, EPANOVATM or AMR101
  • compositions of the present invention as compared to placebo or baseline are more potent than other omega-3 compositions known in the prior art (such as LOVAZA®, EPANOVATM or AMR101 ) in reducing TG while causing a lesser increase in LDL-cholesterol, a lesser non-significant increase in LDL-cholesterol, no increase in LDL-cholesterol at all, or a more potent reduction in LDL-cholesterol at in patients with baseline TG levels above 500 mg/dL.
  • omega-3 compositions known in the prior art such as LOVAZA®, EPANOVATM or AMR101
  • the use of the compositions of the present invention may allow for a reduction in the dose of the statin required for a subject.
  • the coadministration of the composition of the present invention in a subject receiving statin therapy may allow for the reduction of the dose of the statin, compared to subject not being co-administered a composition of the present invention.
  • the dose of the statin may be reduced by at least 10%, alternatively at least 25%, alternatively at least 50%, or alternatively at least 75%.
  • the use of the compositions of the present invention may reduce the time needed for a subject to reach the recommended blood levels.
  • the administration of compositions of the present invention may allow a subject to reach goal lipid levels, for example, those described in the NCEP ATP III Guidelines, or any levels recommended by a health care practitioner.
  • the reduction of time is greater than 5%, alternatively greater than 15%, alternatively greater than 25%, alternatively greater than 50%, and alternatively greater than 75%.
  • compositions of the present invention are also useful to treat coronary heart disease (CHD), vascular disease, atherosclerotic disease or related conditions.
  • CHD coronary heart disease
  • the compositions of the present invention may also be use for the treatment and/or prevention and/or reduction of cardiac events and/or cardiovascular events and/or vascular events and/or symptoms. 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.
  • the present invention also relates to treatment of such conditions in with concomitant treatments regimes or combination products with other active pharmaceutical ingredients.
  • concomitant or fixed combination treatments may include a statin, an anticoagulant (such as aspirin or clopidogrel), 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 includes pharmaceutical compositions, for example, a unit dosage, comprising one or more HMG-CoA reductase inhibitors ("statins”) and the omega-3 fatty acid composition of the present invention.
  • statins HMG-CoA reductase inhibitors
  • the present invention may incorporate now known or future known statins in an amount generally recognized as safe.
  • statins There are currently seven statins that are widely available: atorvastatin, rosuvastatin, fluvastatin, lovastatin, pravastatin, pitavastatin, and simvastatin.
  • An eight 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.
  • statins are typically used at their common daily doses, which include, but are not limited to lovastatin 10mg, 20mg, 40mg; pravastatin 10mg, 20mg, 40mg, 80mg; simvastatin 5mg, 10mg, 20mg, 40mg, 80mg; fluvastatin 20mg, 40mg, 80mg; atorvastatin 10mg, 20mg, 40mg, 80mg; rosuvastatin 5mg, 10mg, 20mg, 40mg; and pitavastatin 1 mg, 2mg, 4mg, 8mg.
  • 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 present invention also includes methods of treatment, comprising dosing of one or more statins and the omega-3 fatty acid composition of the present invention, either as concomitant therapy or in a fixed dose combination product comprising both a statin and the composition of the present invention.
  • This method of treatment combines the administration of one or more statins at its common dose or an alternative dose with the composition of the present invention.
  • compositions of the present invention which comprise significant amounts of omega-3 docosapentaenoic acid (DPA-n3) or its glycerol or ethyl esters, together with a common or alternative statin dose, may be used for the treatment of hypertriglyceridemia (either TG ⁇ 500mg/dl_, TG ⁇ 200mg/dL, TG>150mg/dL, TG 200-499mg/dl_, TG300-499mg/dl_, TG350-499mg/dl_, or TG 150- 199mg/dL), mixed dyslipidemia, or any other diseases or medical conditions specified above.
  • DPA-n3 docosapentaenoic acid
  • ethyl esters ethyl esters
  • Such methods of treatment provide to a subject in need thereof a dose of at least 30 mg DPA-N3 per day, alternatively at least 40mg DPA-N3 per day, alternatively at least 50mg DPA-N3 per day, alternatively at least 60mg DPA-N3 per day, alternatively at least 70mg DPA-N3 per day, alternatively at least 80mg DPA-N3 per day, alternatively at least 90mg DPA-N3 per day, alternatively at least 100mg DPA-N3 per day, alternatively at least 120mg DPA-N3 per day, alternatively at least 150mg DPA-N3 per day, alternatively at least 160mg DPA-N3 per day, alternatively at least 180mg DPA-N3 per day, alternatively at least 200mg DPA-N3 per day, alternatively at least 250mgDPA-N3 per day, alternatively at least 300mg DPA-N3 per day, alternatively at least 350mg DPA-N3 per day, alternative
  • compositions of the present invention which comprise significant amounts of omega-3 docosapentaenoic acid (HPA-n3) or its glycerol or ethyl esters, together with a common or alternative statin dose, may be used for the treatment of hypertriglyceridemia (either TG ⁇ 500mg/dL, TG>200mg/dL, TG>150mg/dL, TG200- 499mg/dL, TG300-499mg/dl_, TG350-499mg/dl_, or TG150-199mg/dL), mixed dyslipidemia, or any other diseases or medical conditions specified above.
  • hypertriglyceridemia either TG ⁇ 500mg/dL, TG>200mg/dL, TG>150mg/dL, TG200- 499mg/dL, TG300-499mg/dl_, TG350-499mg/dl_, or TG150-199mg/dL
  • Such methods of treatment provide to a subject in need thereof a dose of at least 10mg HPA-N3 per day, alternatively at least 15mg HPA-N3 per day, alternatively at least 20mg HPA-N3 per day, alternatively at least 25mg HPA-N3 per day, alternatively at least 30mg HPA-N3 per day, alternatively at least 40mg HPA-N3 per day, alternatively at least 50mg HPA-N3 per day, alternatively at least 60mg HPA-N3 per day, alternatively at least 80mg HPA-N3 per day, alternatively at least 90mg HPA-N3 per day, alternatively at least 120mg HPA-N3 per day, alternatively at least 150mg HPA-N3 per day, alternatively at least 160mg HPA-N3 per day, alternatively at least 180mg HPA-N3 per day, alternatively at least 200mg HPA-N3 per day, alternatively at least 250mg HPA-N3 per day,
  • compositions of the present invention which comprise significant amounts of omega-3 docosapentaenoic acid (DPA-n3) or its glycerol or ethyl esters and which comprise relatively small amounts of omega-3 docosahexaenoic acid (DHA-n3), together with a common or alternative statin dose, may be used for the treatment of hypertriglyceridemia (either TG ⁇ 500mg/dL, TG ⁇ 200mg/dL, TG>150mg/dL, TG 200-499mg/dl_, TG300-499mg/dl_, TG350- 499mg/dL, or TG 150-199mg/dl_), mixed dyslipidemia, or any other diseases or medical conditions specified above.
  • hypertriglyceridemia either TG ⁇ 500mg/dL, TG ⁇ 200mg/dL, TG>150mg/dL, TG 200-499mg/dl_, TG300-499mg/
  • Such method of treatment provides to a subject in need thereof a common or alternative statin dose together with a dose of at least 30mg DPA-N3 per day, alternatively at least 40mg DPA-N3 per day, alternatively at least 50mg DPA-N3 per day, alternatively at least 60mg DPA-N3 per day, alternatively at least 80mg DPA-N3 per day, alternatively at least 90mg DPA-N3 per day, alternatively at least 120mg DPA-N3 per day, alternatively at least 150mg DPA- N3 per day, alternatively at least 160mg DPA-N3 per day, alternatively at least 180mg DPA-N3 per day, alternatively at least 200mg DPA-N3 per day, alternatively at least 250mg DPA-N3 per day, alternatively at least 300mg DPA-N3 per day, alternatively at least 350mg DPA-N3 per day, alternatively at least 400mg DPA-N3 per day, alternatively at least 500
  • compositions of the present invention which comprise significant amounts of omega-3 docosapentaenoic acid (HPA-n3) or its glycerol or ethyl esters and which comprise relatively small amounts of omega-3 docosahexaenoic acid (DHA-n3), together with a common or alternative statin dose, may be used for the treatment of hypertriglyceridemia (either TG ⁇ 500mg/dL, TG>200mg/dL, TG ⁇ 150mg/dL, TG 200-499mg/dL, TG300-499mg/dL, TG350- 499mg/dL, or TG 150-199mg/dL), mixed dyslipidemia, or any other diseases or medical conditions specified above.
  • hypertriglyceridemia either TG ⁇ 500mg/dL, TG>200mg/dL, TG ⁇ 150mg/dL, TG 200-499mg/dL, TG300-499mg/dL,
  • Such method of treatment provides to a subject in need thereof a common or alternative statin dose together with a dose of at least 10mg HPA-N3 per day, alternatively at least 15mg HPA-N3 per day, alternatively at least 20mg HPA-N3 per day, alternatively at least 25mg HPA-N3 per day, alternatively at least 30mg HPA-N3 per day, alternatively at least 40mg HPA-N3 per day, alternatively at least 50mg HPA-N3 per day, alternatively at least 60mg HPA-N3 per day, alternatively at least 80mg HPA-N3 per day, alternatively at least 90mg HPA-N3 per day, alternatively at least 120mg HPA-N3 per day, alternatively at least 150mg HPA-N3 per day, alternatively at least 160mg HPA-N3 per day, alternatively at least 180mg HPA-N3 per day, alternatively at least 200mg HPA-N3 per day, alternatively at least 250
  • compositions of the present invention which comprise significant amounts of omega-3 docosapentaenoic acid (DPA-n3) or its glycerol or ethyl esters and which comprise relatively small amounts of omega-3 docosahexaenoic acid (DHA-n3), together with a common or alternative statin dose, may be used for the treatment of hypertriglyceridemia (either TG ⁇ 500mg/dL, TG ⁇ 200mg/dl_, TG>150mg/dL, TG 200-499mg/dL, TG300-499mg/dL, TG350- 499mg/dL, or TG 150-199mg/dl_), mixed dyslipidemia, or any other diseases or medical conditions specified above.
  • hypertriglyceridemia either TG ⁇ 500mg/dL, TG ⁇ 200mg/dl_, TG>150mg/dL, TG 200-499mg/dL, TG300-499mg/d
  • Such method of treatment provides to a subject in need thereof a common or alternative statin dose together with a dose of at least 30mg DPA-N3 per day, alternatively at least 40mg DPA-N3 per day, alternatively at least 50mg DPA-N3 per day, alternatively at least 60mg DPA-N3 per day, alternatively at least 70mg DPA-N3 per day, alternatively at least 80mg DPA-N3 per day, alternatively at least 90mg DPA-N3 per day, alternatively at least 100mg DPA- N3 per day, alternatively at least 120mg DPA-N3 per day, alternatively at least 150mg DPA-N3 per day, alternatively at least 160mg DPA-N3 per day, alternatively at least 180mg DPA-N3 per day, alternatively at least 200mg DPA-N3 per day, alternatively at least 250mg DPA-N3 per day, alternatively at least 300mg DPA-N3 per day, alternatively at least 350
  • compositions of the present invention which comprise significant amounts of omega-3 docosapentaenoic acid (HPA-n3) or its glycerol or ethyl esters and which comprise relatively small amounts of omega-3 docosahexaenoic acid (DHA-n3), together with a common or alternative statin dose, may be used for the treatment of hypertriglyceridemia (either TG>500mg/dL, TG>200mg/dL, TG ⁇ 150mg/dL, TG 200-499mg/dL, TG300-499mg/dL, TG350- 499mg/dL, or TG 150-199mg/dL), mixed dyslipidemia, or any other diseases or medical conditions specified above.
  • hypertriglyceridemia either TG>500mg/dL, TG>200mg/dL, TG ⁇ 150mg/dL, TG 200-499mg/dL, TG300-499mg/dL,
  • Such method of treatment provides to a subject in need thereof a common or alternative statin dose together with a dose of at least 10mg HPA-N3 per day, alternatively at least 15mg HPA-N3 per day, alternatively at least 20mg HPA-N3 per day, alternatively at least 25mg HPA-N3 per day, alternatively at least 30mg HPA-N3 per day, alternatively at least 40mg HPA-N3 per day, alternatively at least 50mg HPA-N3 per day, alternatively at least 60mg HPA-N3 per day, alternatively at least 70mg HPA-N3 per day, alternatively at least 80mg HPA-N3 per day, alternatively at least 90mg HPA-N3 per day, alternatively at least 100mg HPA-N3 per day, alternatively at least 120mg HPA-N3 per day, alternatively at least 150mg HPA-N3 per day, alternatively at least 160mg HPA-N3 per day, alternatively at least 180
  • compositions of the present invention which comprise significant amounts of omega-3-pentaenoic acids or their glycerol or ethyl esters and which comprise relatively small amounts of omega-3 docosahexaenoic acid (DHA-n3), together with a common or alternative statin dose, may be used for the treatment of hypertriglyceridemia (either TG ⁇ 500mg/dl_, TG>200mg/dL, TG>150mg/dl_, TG 200-499mg/dL, TG300-499mg/dl_, TG350-499mg/dL, or TG 150- 199mg/dL), mixed dyslipidemia, or any other diseases or medical conditions specified above.
  • hypertriglyceridemia either TG ⁇ 500mg/dl_, TG>200mg/dL, TG>150mg/dl_, TG 200-499mg/dL, TG300-499mg/dl_, TG350-4
  • Such methods of treatment provides to a subject in need thereof with a common or alternative statin dose and a dose of at least 100mg omega-S- pentaenoic acids per day, alternatively at least 200mg omega-3-pentaenoic acids per day, alternatively at least 300mg omega-3-pentaenoic acids per day, alternatively at least 500mg omega-3-pentaenoic acids per day, alternatively at least 700mg omega-3-pentaenoic acids per day, alternatively at least 900mg omega-S- pentaenoic acids per day, alternatively at least 1000mg omega-3-pentaenoic acids per day, alternatively at least 1500mg omega-3-pentaenoic acids per day, alternatively at least 1900mg omega-3-pentaenoic acids per day, alternatively at least 2000mg omega-3-pentaenoic acids per day, alternatively at least 2500mg omega-3-pentaenoic acids per day, alternatively at least 2900
  • compositions of the present invention which comprise significant amounts of omega-3-pentaenoic acids including DPA or their glycerol or ethyl esters and which comprise relatively small amounts of omega-3 docosahexaenoic acid (DHA-n3), together with a common or alternative statin dose, may be used for the treatment of hypertriglyceridemia (either TG ⁇ 500mg/dL, TG>200mg/dL, TG>150mg/dL, TG 200-499mg/dL, TG300-499mg/dL, TG350- 499mg/dL, or TG 150-199mg/dL), mixed dyslipidemia, or any other diseases or medical conditions specified above.
  • hypertriglyceridemia either TG ⁇ 500mg/dL, TG>200mg/dL, TG>150mg/dL, TG 200-499mg/dL, TG300-499mg/dL, TG350- 499m
  • Such methods of treatment provides to a subject in need thereof with a common or alternative statin dose and a dose of at least 100mg omega-3-pentaenoic acids per day, alternatively at least 200mg omega-3-pentaenoic acids per day, alternatively at least 300mg omega-3-pentaenoic acids per day, alternatively at least 500mg omega-3-pentaenoic acids per day, alternatively at least 700mg omega-3-pentaenoic acids per day, alternatively at least 900mg omega-3-pentaenoic acids per day, alternatively at least 1000mg omega-3- pentaenoic acids per day, alternatively at least 1500mg omega-3-pentaenoic acids per day, alternatively at least 1900mg omega-3-pentaenoic acids per day, alternatively at least 2000mg omega-3-pentaenoic acids per day, alternatively at least 2500mg omega-3-pentaenoic acids per day, alternatively at least 2900mg
  • compositions of the present invention which comprise significant amounts of omega-3-pentaenoic acids including HPA or their glycerol or ethyl esters and which comprise relatively small amounts of omega-3 docosahexaenoic acid (DHA-n3), together with a common or alternative statin dose, may be used for the treatment of hypertriglyceridemia (either TG>500mg/dL, TG>200mg/dl_, TG ⁇ 150mg/dl_, TG 200-499mg/dL, TG300-499mg/dl_, TG350- 499mg/dl_, or TG 150-199mg/dl_), mixed dyslipidemia, or any other diseases or medical conditions specified above.
  • hypertriglyceridemia either TG>500mg/dL, TG>200mg/dl_, TG ⁇ 150mg/dl_, TG 200-499mg/dL, TG300-499mg/dl_
  • Such methods of treatment provides to a subject in need thereof with a common or alternative statin dose and a dose of at least 100mg omega-3-pentaenoic acids per day, alternatively at least 200mg omega-3-pentaenoic acids per day, alternatively at least 300mg omega-3-pentaenoic acids per day, alternatively at least 500mg omega-3-pentaenoic acids per day, alternatively at least 700mg omega-3-pentaenoic acids per day, alternatively at least 900mg omega-3-pentaenoic acids per day, alternatively at least l OOOmg omega-3- pentaenoic acids per day, alternatively at least 1500mg omega-3-pentaenoic acids per day, alternatively at least 1900mg omega-3-pentaenoic acids per day, alternatively at least 2000mg omega-3-pentaenoic acids per day, alternatively at least 2500mg omega-3-pentaenoic acids per day, alternatively at least 2
  • Such methods of treatment provides to a subject in need thereof with a common or alternative statin dose and a dose of at least 100mg omega-3- pentaenoic acids per day, alternatively at least 200mg omega-3-pentaenoic acids per day, alternatively at least 300mg omega-3-pentaenoic acids per day, alternatively at least 500mg omega-3-pentaenoic acids per day, alternatively at least 700mg omega-3-pentaenoic acids per day, alternatively at least 900mg omega-3- pentaenoic acids per day, alternatively at least 1000mg omega-3-pentaenoic acids per day, alternatively at least 1500mg omega-3-pentaenoic acids per day, alternatively at least 1900mg omega-3-pentaenoic acids per day, alternatively at least 2000mg omega-3-pentaenoic acids per day, alternatively at least 2500mg omega-3-pentaenoic acids per day, alternatively at least 2
  • 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.
  • 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 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 compostiions 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 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 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 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 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 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.
  • compositions of the present invention are preferably 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.
  • compositions of the present invention are preferably 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.
  • compositions of the present invention are preferably 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 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 1 15% 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.
  • 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%).
  • 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.
  • 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%).
  • 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%).
  • a composition according to the present invention isprepared 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.
  • Fatty Acid Ethyl Ester (FAEE GMP, approx. 3mmol/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.
  • Step 4 Repeat Step 4 several times ( ⁇ 2x) to remove ethanol and NaCI.
  • Example 3 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 6 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 100ml_ of this antioxidant preparation into 100 liters of the oil composition of Example 4 followed by thorough homogenization. The resulting pre-encapsulation formulated oil contains approximately 4mg/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 4mg/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 1000mg omega-3-pentaenoic-acids ethyl esters per capsule.
  • 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 100ml_ of this antioxidant preparation into 100 liters of the oil composition of Example 4 followed by thorough homogenization. The resulting pre-encapsulation formulated oil contains approximately 4mg/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 4mg/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, NC, 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 1000mg 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. 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 100ml_ of this antioxidant preparation into 100 liters of the oil composition of Example 4 followed by thorough homogenization. The resulting pre-encapsulation formulated oil contains approximately 4mg/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 4mg/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.Petersburg, FL, 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 1000mg 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 100ml_ 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 4mg/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 4mg/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, NC, 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 1000mg 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 severe hypertriglyceridemia (TG>500mg/dL). 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 (4g/d).
  • a patient is treated as per Example 15.
  • the treatment results in significant reduction of TG as well as non-HDL- and VLDL-cholesterol levels while the LDL- cholesterol level changes insignificantly.
  • a patient is treated as per Example 5.
  • the treatment results in significant reduction of TG as well as ⁇ -HDL-, LDL- and VLDL-cholesterol levels.
  • a patient already undergoing treatment with a statin is diagnosed with high triglycerides (TG between 200 and 500mg/dL). Thereupon, the patient is initiated on daily treatment with one of the encapsulated compositions according to Examples 10, 1 1 , 12 or 13. Four capsules per day are administered to this patient (4 g/d).
  • Example 19 A patient is treated as per Example 18. The treatment results in significant reduction of TG as well as ⁇ -HDL-, VLDL- and LDL-cholesterol levels.
  • a patient is diagnosed with mixed dyslipidemia (TG between 200 and 700 mg/dL and LDL-cholesterol above 190 mg/dL). Thereupon, the patient is initiated on concomitant daily treatment with a statin and one of the encapsulated compositions according to Examples 1 1 , 12, 13, or 14. Four capsules per day are administered to this patient (4 g/d).
  • a patient is treated as per Example 20.
  • the treatment results in significant reduction of TG as well as ⁇ -HDL-, VLDL- and LDL-cholesterol levels.
  • a patient is diagnosed with mixed dyslipidemia (TG between 200 and 700 mg/dL and non-HDL-cholesterol above 200 mg/dL). Thereupon, the patient is initiated on concomitant daily treatment with a statin and one of the encapsulated compositions according to Examples 1 1 , 12, 13, or 14. Four capsules per day are administered to this patient (4 g/d).
  • a patient is treated as per Example 22.
  • the treatment results in significant reduction of TG as well as ⁇ -HDL-, VLDL- and LDL-cholesterol levels.
  • a patient is diagnosed to be at significant risk for a cardiovascular event according to the NCEP guidelines and has TG levels above 150mg/dL. Thereupon, the patient is initiated on daily treatment with one of the encapsulated compositions according to Examples 1 1 , 12, 13, or 14. Four capsules per day are administered to this patient (4 g/d).
  • Example 25 A patient is treated as per Example 24. The treatment results in significant reduction of TG as well as ⁇ -HDL-, VLDL- and LDL-cholesterol levels.
  • a patient diagnosed as per Example 15, 18, 20, 22 or 24 is treated with 3 capsules per day (instead of 4) of one of the encapsulated compositions according to Examples 1 1 , 12, 13, or 14.
  • the treatment results in significant reduction of TG as well as non-HDL- and VLDL-cholesterol levels.
  • a patient diagnosed as per Example 15, 18, 20, 22 or 24 is treated with 3 capsules per day (instead of 4) of one of the encapsulated compositions according to Examples 11 , 12, 13, or 14.
  • the treatment results in significant reduction of TG as well as ⁇ -HDL-, VLDL- and LDL-cholesterol levels.
  • a patient diagnosed as per Example 15, 18, 20, 22 or 24 is treated with 2 capsules per day (instead of 3 or 4) of one of the encapsulated compositions according to Examples 11 , 12, 13, or 14.
  • the treatment results in significant reduction of TG as well as non-HDL- and VLDL-cholesterol levels.
  • a patient diagnosed as per Example 15, 18, 20, 22 or 24 is treated with 2 capsules per day (instead of 3 or 4) of one of the encapsulated compositions according to Examples 1 1 , 12, 13, or 14.
  • the treatment results in significant reduction of TG as well as ⁇ -HDL-, VLDL- and LDL-cholesterol levels.
  • DPA Docosapentaenoic acid 60 100 80
  • DHA Docosahexaenoic acid
  • DPA Docosapentaenoic acid
  • DHA Docosahexaenoic acid
  • 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.
  • DHA Docosahexaenoic acid
  • 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 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.
  • the DPA may be composed as a glyceride (such as triglyceride), an ester (such as ethyl ester), or a free fatty acid.
  • a mixture of DPA and EPA was prepared by combining 1 g DPA Ethyl Ester (SE-133-III) with 10g EPA Ethyl Ester, 914 mg/g (KD Pharma FM13001 ) in 150ml of 95% ethanol/water containing 35ml of 2M sodium hydroxide. This reaction mixture was stirred overnight at ambient temperature. Tic 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.
  • Corn oil was used as a diluent for the omega-3 compounds, and methylcellulose to prepare the statin (atorvastatin) for dosing.
  • statin atorvastatin
  • a separate group of animals receiving corn oil alone was used as the untreated control group. Animals received daily doses of respective solutions by oral gavage. The study was conducted in 2 phases. In the first phase, DPA and VASCEPA ⁇ solution is administered at 200 mg/kg, 400 mg/kg, and 1000 mg/kg. Animals were dosed daily for 14 days. For reference, a rat dose of 400 mg/kg would be equivalent to a human daily dose of approximately 4 grams (as shown in Reagan-Shaw et al. "Dose translation from animal to human studies revisited," FASEB J. 22, 659-661 (2007), which is incorporated by reference in its entirety).
  • the second phase was initiated on day 15, with the groups receiving DPA and VASCEPA ⁇ 400 mg/kg solution being co-administered statin at 10 mg/kg.
  • Another group previously dosed with corn oil vehicle, received atorvastatin to serve as an appropriate control.
  • This second phase consisted of 14 days of daily, oral administration.
  • Plasma total cholesterol, LDL, HDL, triglycerides and VLDL were measured in the fasting state on day 0, 7 and 14; and for those groups included in the second phase on days 21 and 28. Levels of lipid parameters were determined in a 96-well multiplexed system using standard clinical chemistry techniques. Non-HDL cholesterol was calculated by subtracting the HDL value from the total cholesterol value. In addition, for the groups included in the second phase, insulin levels were determined at day 28.
  • FIG. 1 shows the fasting plasma lipid values after seven days of dosing.
  • FIG. 2 shows the fasting plasma insulin levels after 28 days of administration.
  • HMGCoA 3-hydroxy-3-methylglutaryl-coenzyme A; key regulatory enzyme for new cholesterol biosynthesis
  • PCSK9 pro-protein convertase subtilisin kexin 9; associated with LDL receptor functioning and increased levels of LDL
  • SREB-2 sterol regulatory enhancing binding protein 2; regulates transcription of a wide variety of genes involved with new cholesterol synthesis
  • the mRNA messenger RNA
  • cDNA complementary DNA
  • FIG. 3 shows the relative liver gene expression following 28 days of administration.
  • Inclusion criteria for tested subjects include volunteers between ages 18-65, with a BMI of 30-35 (alternatively a BMI of 27-35) and triglyceride levels less than 350 mg/dL, who consume no more than 1 fish meal per week and who are not currently prescribed pharmaco-therapy for lowering triglycerides, including but not limited to fibrates, omega-3 agents, and niacin. Volunteers on stable anti-hypertensive, anti-diabetic and thyroid therapy re allowed for consideration. Any person on stable statin therapy is considered if their triglyceride levels are less than 350 mg/dL. However, the total composition of subjects in the study with this particular profile is limited to no more than 30%.
  • compound dosing is stopped for a 2- 4-week washout period prior to the initiation of the a similar cycle as above, now with COMPOSITION 1 a or 1 b and the reference compound are administered together with a meal.
  • Plasma levels of omega-3 fatty acids of interest from the study are determined utilizing an analytical LC/MS technique under GLP laboratory conditions in order to determine Cmax, Tmax and AUC for the omega-3 fatty acids of interest, including EPA, DPA, HPA, DHA, and other omega-3 fatty acids.
  • COMPOSITION 1 a or 1 b has a better bioavalability (as measured by AUC and Cmax) than the reference compound. This effect is seen under fasting and/or fed administration conditions.
  • this Study #2 is conducted with a certain dose level of COMPOSITION 2, COMPOSITION 3, or COMPOSITION 4 instead of
  • This Phase 3, multi-center study consists of a 6- to 8-week screening/washout period (to include a diet and lifestyle stabilization period), which includes a fasting triglyceride (TG) qualifying period of 2-3 weeks, followed by a 12- week double-blind treatment period.
  • TG fasting triglyceride
  • Patients on statin therapy (with or without ezetimibe) at screening are evaluated by the investigator as to whether this therapy could be safely discontinued at screening, or if it is to be continued.
  • Patients on any other dyslipidemia therapy need to discontinue these in order to qualify for the study. If statin therapy (with or without ezetimibe) is to be continued, dose(s) must be stable for >4 weeks prior to the fasting TG baseline qualifying measurements for randomization.
  • the screening visit is to occur at either 6 weeks before randomization for patients not on lipid-altering therapy at screening or for patients who do not need to discontinue their current dyslipidemia therapy, or at 8 weeks before randomization for patients who require washout of their current dyslipidemia therapy at screening.
  • the population for this study is men and women >18 years of age with a body mass index (BMI) ⁇ 45 kg/m2. Patients on lipid-lowering therapy and patients not on lipid-lowering therapy are eligible to enroll in the study. Patients had to have an average TG level >500 mg/dL and ⁇ 2000 mg/dL during the screening period to be eligible for randomization.
  • BMI body mass index
  • eligible patients will enter a 12-week randomized, double-blind treatment period.
  • patients will be randomly assigned to 1 of the following treatment groups: COMPOSITION 1 a or 1 b (approximately 2 g daily), COMPOSITION 1a or 1 b (approximately 3 g daily), COMPOSITION 1 a or 1 b (approximately 4 g daily), or placebo.
  • the daily dose may be taken as either a single dose or distributed over two doses per day.
  • COMPOSITION 1a or 1 b To determine the effect of COMPOSITION 1a or 1 b on lipid profiles, including total cholesterol (TC), non-high-density lipoprotein cholesterol (non- HDL-C) low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C), and very low-density lipoprotein cholesterol (VLDL-C);
  • TC total cholesterol
  • non-HDL-C non-high-density lipoprotein cholesterol
  • LDL-C low-density lipoprotein cholesterol
  • HDL-C high-density lipoprotein cholesterol
  • VLDL-C very low-density lipoprotein cholesterol
  • ICM-1 intracellular adhesion molecule-1
  • VCAM_1 vascular cell adhesion molecule 1
  • IL- ⁇ ⁇ interleukin-2
  • IL-6 interleukin-6
  • IL-8 interleukin-8
  • IL-10 interleukin-10
  • IL-12 interleukin-12
  • IL-15 interleukin-15
  • IL-18 interleukin-18
  • TNF-a tumor necrosis factor-alpha
  • TNF- ⁇ tumor necrosis factor-beta
  • PAI-1 plasminogen activator inhibitor- 1
  • the primary efficacy variable for the double-blind treatment period is percent change in fasting TG from baseline to the Week 12 endpoint.
  • the secondary efficacy variable for the double-blind treatment period includes the following: Percent changes in fasting Non-HDL-C, LDL-C, VLDL-C, HDL-C, Lp-PLA2, and apo B from baseline to Week 12 endpoint.
  • the primary and secondary efficacy analyses will be performed using an analysis of covariance (ANCOVA) model with treatment, gender, and the use of statin therapy at randomization as factors and baseline fasting TG value as a covariate.
  • ANCOVA analysis of covariance
  • this Study #3 is conducted with one or more dose levels of COMPOSITION 2, 3, or 4 instead of COMPOSITION 1 a or 1 b.
  • This multi-center study consists of a 4- to 6-week screening and washout period (to include a diet and lifestyle stabilization period, and to wash-out any non- statin/ezetimibe dyslipidemia medications), which also includes a 2-3 week fasting triglyceride (TG) level qualifying period, followed by a 6- to 12-week double-blind treatment period.
  • TG fasting triglyceride
  • statin therapy with or without ezetimibe
  • LDL low- density lipoprotein
  • statin therapy with or without ezetimibe
  • Dose(s) of statin therapy must be stable for >4 weeks prior to the TG baseline qualifying measurements for randomization.
  • the population for this study is men and women >18 years of age with a body mass index (BMI) ⁇ 45 kg/m2. Patients on lipid-lowering therapy and patients not on lipid-lowering therapy are eligible to enroll in the study. Patients had to have an average fasting TG level >200 mg/dL and ⁇ 499 mg/dL during the qualifying period to be eligible for randomization.
  • BMI body mass index
  • eligible patients will enter a 6- to 12-week randomized, double-blind treatment period.
  • patients will be randomly assigned to one of the following treatment groups: COMPOSITION 1 a or 1 b at an approximately 2 gram daily dose, COMPOSITION 1 a or 1 b at an approximately 3 gram daily dose, COMPOSITION 1 a or 1 b at an approximately 4 gram daily dose, or placebo.
  • the daily dose may be taken as either a single dose or distributed over two doses per day.
  • the primary objective of the study is to determine the efficacy of COMPOSITION 1 at approximately 2 grams daily, approximately 3 grams daily and approximately 4 grams daily, compared to placebo, in lowering fasting TG levels in statin-treated patients with fasting TG levels >200 mg/dL and ⁇ 499 mg/dL.
  • the secondary and exploratory objectives of the study may include but are not limited to the following objectives: [0293] 1. To determine the safety and tolerability of COMPOSITION 1 a or 1 b at approximately 2 g daily, approximately 3 g daily and approximately 4 g daily;
  • TC total cholesterol
  • non-HDL-C non-high-density lipoprotein cholesterol
  • LDL-C low-density lipoprotein cholesterol
  • HDL-C high-density lipoprotein cholesterol
  • VLDL-C very low-density lipoprotein cholesterol
  • ICM-1 intracellular adhesion molecule-1
  • VCAM_1 vascular cell adhesion molecule 1
  • IL-1 ⁇ interleukin-1 ⁇
  • IL-2 interleukin-2
  • IL-6 interleukin-6
  • IL-8 interleukin-8
  • IL-10 interleukin-10
  • IL-12 interleukin-12
  • IL-15 interleukin-15
  • IL-18 interleukin-18
  • TNF-a tumor necrosis factor-alpha
  • TNF- ⁇ tumor necrosis factor-beta
  • PAI-1 plasminogen activator inhibitor-1
  • the primary efficacy variable for the double-blind treatment period is percent change in fasting TG from baseline to the Week 6 to 12 endpoint.
  • the secondary efficacy variable for the double-blind treatment period include but are not limited to the following: Percent changes in fasting non-HDL-C, LDL-C, VLDL-C, HDL-C, Lp-PLA2, and apo B from baseline to Week 6 to 12 endpoint.
  • the primary and secondary efficacy analyses will be performed using an analysis of covariance (ANCOVA) model with treatment, gender, the type of statin therapy and diagnosis of diabetes at randomization as factors and baseline fasting TG value as a covariate.
  • ANCOVA analysis of covariance
  • this Study #4 is conducted with one or more dose levels of COMPOSITION 2, 3, or 4 instead of COMPOSITION 1 a or 1 b.
  • Study #4 is conducted, enrolling patients with a baseline triglyceride level of about 300 to 499 mg/dL or about 350 too 400 mg/dL, instead of 200 to 499 mg/dL.
  • Inclusion criteria for tested subjects include volunteers between ages 18-65, with a BMI of 30-35 (alternatively a BMI of 27-35) and triglyceride levels less than 350 mg/dL, who consume no more than 1 fish meal per week and who are not currently prescribed pharmaco-therapy for lowering triglycerides, including but not limited to fibrates, omega-3 agents, and niacin. Volunteers on stable antihypertensive, anti-diabetic and thyroid therapy re allowed for consideration. Any person on stable statin therapy is considered if his or her triglyceride levels are less than 350 mg/dL. However, the total composition of subjects in the study with this particular profile is limited to no more than 30%.
  • Volunteers self-administering omega-3 non-prescription dietary supplements are asked to refrain from their use 2 weeks prior to the initiation of the study until study completion.
  • Subjects using any other non-steroidal anti-inflammatory agents other than acetaminophen are asked to abstain and switch to acetaminophen for relief of pain, or are excluded from study consideration.
  • Subjects are excluded if they receive any type of hormone therapy, weight loss agents, HIV therapy, beta- blockers, or are diagnosed with known cardiovascular disease, including heart failure, arrhythmia, any incidence of acute coronary syndrome, myocardial infarct, coronary artery bypass graft surgery, and/or angioplasty.
  • COMPOSITION 1 a or 1 b or the reference compound are dosed at approximately 4 grams/day in the morning by administration of 4 capsules containing approximately 1 gram of each compound.
  • Baseline plasma levels of fasting serum triglycerides are determined on day 1 and just prior to initiation and completion of either the fasting or fed multi-dosing period. Additional lipid and other parameters (see below under “Results”) that are analyzed included total cholesterol, LDL, HDL, VLDL, non-HDL, and NEFA as previously described.
  • COMPOSITION 1 a or 1 b has a better fasting triglyceride lowering effect than the reference compound. This effect is seen under fasting and/or fed administration conditions.
  • Administration of COMPOSITION 1 a or 1 b have a beneficial effect, versus baseline and versus the reference compound, on other lipid parameters (such as HDL cholesterol, total cholesterol, non-HDL cholesterol, VLDL cholesterol), on platelet function, and one or more of the following: apolipoprotein A-l (apo A-l), apolipoprotein B (apo B), apo A-l/apo B ratio, lipoprotein(a) (Lp[a]), lipoprotein-associated phospholipase A2 (Lp-PLA2), low density lipoprotein (LDL) particle number and size, oxidized LDL, C-reactive protein (CRP), high sensitivity C-reactive protein (HSCRP), intracellular adhesion molecule- 1 (ICAM-1 ), E-selectin, P
  • COMPOSITION 1 has a beneficial impact, or a minimal impact, or no impact, on other non-HDL lipid parameters, such as LDL cholesterol versus baseline and the reference compound.
  • this Study #5 is conducted with a certain dose level of COMPOSITION 2, 3, pr 4 instead of COMPOSITION 1 a or 1 b.
  • 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 .
  • EPA eicosapentaenoic acid
  • DPA docosapentaenoic acid
  • 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 .
  • EPA eicosapentaenoic acid
  • DPA docosapentaenoic acid
  • 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.
  • EPA eicosapentaenoic acid
  • DPA docosapentaenoic acid
  • 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.
  • EPA eicosapentaenoic acid
  • DPA docosapentaenoic acid
  • 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.
  • EPA eicosapentaenoic acid
  • DPA docosapentaenoic acid
  • 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 .
  • EPA eicosapentaenoic acid
  • DPA docosapentaenoic acid
  • 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.
  • EPA eicosapentaenoic acid
  • DPA docosapentaenoic acid
  • 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.
  • EPA eicosapentaenoic acid
  • DPA docosapentaenoic acid
  • composition according to one of the preferred embodiments 1 through 8 comprising at least 2% docosapentaenoic acid (DPA; C22:5-n3).
  • DPA docosapentaenoic acid
  • composition according to one of the preferred embodiments 1 through 8 comprising at least 4% docosapentaenoic acid (DPA; C22:5-n3).
  • DPA docosapentaenoic acid
  • composition according to one of the preferred embodiments 1 through 8 comprising at least 5% docosapentaenoic acid (DPA; C22:5-n3).
  • DPA docosapentaenoic acid
  • composition according to one of the preferred embodiments 1 through 8 comprising at least 6% docosapentaenoic acid (DPA; C22:5-n3).
  • DPA docosapentaenoic acid
  • composition according to one of the preferred embodiments 1 through 8 comprising at least 7% docosapentaenoic acid (DPA; C22:5-n3).
  • DPA docosapentaenoic acid
  • composition according to one of the preferred embodiments 1 through 8 comprising at least 8% docosapentaenoic acid (DPA; C22:5-n3).
  • DPA docosapentaenoic acid
  • a composition according to one of the preferred embodiments 1 through 8 comprising at least 10% docosapentaenoic acid (DPA; C22:5-n3).
  • DPA docosapentaenoic acid
  • composition according to one of the preferred embodiments 1 through 8 comprising at least 12% docosapentaenoic acid (DPA; C22:5-n3).
  • DPA docosapentaenoic acid
  • composition according to one of the preferred embodiments 1 through 8 comprising at least 15% docosapentaenoic acid (DPA; C22:5-n3).
  • DPA docosapentaenoic acid
  • composition according to one of the preferred embodiments 1 through 17 comprising no more than 95% EPA.
  • a composition according to one of the preferred embodiments 1 through 17 comprising no more than 10% omega-6 fatty acids.
  • a composition according to one of the preferred embodiments 1 through 17 comprising no more than 7% omega-6 fatty acids.
  • composition according to one of the preferred embodiments 1 through 22, comprising no more than 5% arachidonic acid (C22:4-n6).
  • composition according to one of the preferred embodiments 1 through 22, comprising no more than 4% arachidonic acid (C22:4-n6).
  • composition according to one of the preferred embodiments 1 through 22, comprising no more than 3% arachidonic acid (C22:4-n6).
  • composition according to one of the preferred embodiments 1 through 22, comprising no more than 2% arachidonic acid (C22:4-n6).
  • composition according to one of the preferred embodiments 1 through 22, comprising no more than 1 % arachidonic acid (C22:4-n6).
  • composition according to one of the preferred embodiments 1 through 27, comprising at least 0.01 % heneicosapentaenoic acid (C21 :5-n3).
  • composition according to one of the preferred embodiments 1 through 27, comprising at least 0.3% heneicosapentaenoic acid (C21 :5-n3).
  • composition according to one of the preferred embodiments 1 through 27, comprising at least 0.5% heneicosapentaenoic acid (C21 :5-n3).
  • composition according to one of the preferred embodiments 1 through 27, comprising at least 1 % heneicosapentaenoic acid (C21 :5-n3).
  • composition according to one of the preferred embodiments 1 through 27, comprising at least 2% heneicosapentaenoic acid (C21 :5-n3).
  • composition according to one of the preferred embodiments 1 through 27, comprising at least 3% heneicosapentaenoic acid (C21 :5-n3).
  • composition according to one of the preferred embodiments 1 through 27, comprising at least 4% heneicosapentaenoic acid (C21 :5-n3).
  • 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.
  • 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.
  • 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.
  • 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.
  • 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.
  • 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.
  • composition according to one of the preferred embodiments through 44 wherein the EPA:DPA ratio is between 5:1 and 1 :1.
  • composition according to one of the preferred embodiments 1 through 44 comprising between 65% and 95% EPA.
  • composition according to one of the preferred embodiments 1 through 44 comprising between 75% and 95% EPA.
  • composition according to one of the preferred embodiments 1 through 44 comprising between 85% and 95% EPA.
  • a composition according to one of the preferred embodiments 1 through 44 comprising between 90% and 95% EPA.
  • composition according to one of the preferred embodiments 1 through 44 comprising between 2% and 10% DPA.
  • composition according to one of the preferred embodiments 1 through 44 comprising between 3% and 20% DPA.
  • composition according to one of the preferred embodiments 1 through 44 comprising between 3% and 30% DPA.
  • composition according to one of the preferred embodiments 1 through 44 comprising between 3% and 75% DPA.
  • composition according to one of the preferred embodiments 1 through 44 comprising between 3% and 90% DPA.
  • 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.
  • 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.
  • 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.
  • 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).
  • 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. .
  • 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.
  • 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.
  • 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.
  • 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.
  • 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.
  • 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.
  • 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.
  • 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.
  • 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.
  • 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.
  • 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 administration or treatment under low fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in a Tmax of 8 hours or less
  • a method of administration or treatment under low fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in a Tmax of 6 hours or less
  • a method of administration or treatment under low fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in a Tmax of 4 hours or less
  • a method of administration or treatment under low fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA Tmax less than the EPA+DPA+DHA Tmax for LOVAZA® under the same administration conditions
  • a method of administration or treatment under high fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax of at least 1 10% of the EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax for LOVAZA® under the same administration conditions
  • a method of administration or treatment under high fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax of at least 120% of the EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax for LOVAZA® under the same administration conditions
  • a method of administration or treatment under high fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax of at least 130% of the EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax for LOVAZA® under the same administration conditions
  • a method of administration or treatment under high fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax of at least 140% of the EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax for LOVAZA® under the same administration conditions
  • a method of administration or treatment under low fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax of at least 200% of the EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax for LOVAZA® under the same administration conditions
  • a method of administration or treatment under low fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax of at least 300% of the EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax for LOVAZA® under the same administration conditions
  • a method of administration or treatment under low fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax of at least 400% of the EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax for LOVAZA® under the same administration conditions
  • a method of administration or treatment under low fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax of at least 500% of the EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax for LOVAZA® under the same administration conditions
  • a method of administration or treatment under low fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax of at least 600% of the EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax for LOVAZA® under the same administration conditions
  • a method of administration or treatment under fasting conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax of at least 150% of the in EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax for LOVAZA® under the same administration conditions
  • a method of administration or treatment under fasting conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax of at least 200% of the EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax for LOVAZA® under the same administration conditions
  • a method of administration or treatment under fasting conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax of at least 300% of the EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax for LOVAZA® under the same administration conditions
  • a method of administration or treatment under fasting conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax of at least 400% of the EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax for LOVAZA® under the same administration conditions
  • a method of administration or treatment under fasting conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax of at least 500% of the EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax for LOVAZA® under the same administration conditions
  • a method of administration or treatment under fasting conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax of at least 600% of the EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax for LOVAZA® under the same administration conditions
  • a method of administration or treatment under fasting conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax of at least 700% of the EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax for LOVAZA® under the same administration conditions
  • a method of administration or treatment under high fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA AUC or Total Omega-3 FA AUC of at least 1 10% of the EPA+DPA+DHA AUC or Total Omega-3 FA AUC for LOVAZA® under the same administration conditions
  • a method of administration or treatment under high fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA AUC or Total Omega-3 FA AUC of at least 120% of the EPA+DPA+DHA AUC or Total Omega-3 FA AUC for LOVAZA® under the same administration conditions
  • a method of administration or treatment under high fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA AUC or Total Omega-3 FA AUC of at least 130% of the EPA+DPA+DHA AUC or Total Omega-3 FA AUC for LOVAZA® under the same administration conditions
  • a method of administration or treatment under high fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA AUC or Total Omega-3 FA AUC of at least 140% of the EPA+DPA+DHA AUC or Total Omega-3 FA AUC for LOVAZA® under the same administration conditions
  • a method of administration or treatment under high fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA AUC or Total Omega-3 FA AUC of at least 150% of the EPA+DPA+DHA AUC or Total Omega-3 FA AUC for LOVAZA® under the same administration conditions
  • a method of administration or treatment under low fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA AUC or Total Omega-3 FA AUC of at least 150% of the EPA+DPA+DHA AUC or Total Omega-3 FA AUC for LOVAZA® under the same administration conditions
  • a method of administration or treatment under low fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA AUC or Total Omega-3 FA AUC of at least 400% of the EPA+DPA+DHA AUC or Total Omega-3 FA AUC for LOVAZA® under the same administration conditions
  • a method of administration or treatment under low fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA AUC or Total Omega-3 FA AUC of at least 500% of the EPA+DPA+DHA AUC or Total Omega-3 FA AUC for LOVAZA® under the same administration conditions
  • a method of administration or treatment under low fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA AUC or Total Omega-3 FA AUC of at least 600% of the EPA+DPA+DHA AUC or Total Omega-3 FA AUC for LOVAZA® under the same administration conditions
  • a method of administration or treatment under fasting conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA AUC or Total Omega-3 FA AUC of at least 150% of the EPA+DPA+DHA AUC or Total Omega-3 FA AUC for LOVAZA® under the same administration conditions
  • a method of administration or treatment under fasting conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA AUC or Total Omega-3 FA AUC of at least 200% of the EPA+DPA+DHA AUC or Total Omega-3 FA AUC for LOVAZA® under the same administration conditions
  • a method of administration or treatment under fasting conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA AUC or Total Omega-3 FA AUC of at least 300% of the EPA+DPA+DHA AUC or Total Omega-3 FA AUC for LOVAZA® under the same administration conditions
  • a method of administration or treatment under fasting conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA AUC or Total Omega-3 FA AUC of at least 400% of the EPA+DPA+DHA AUC or Total Omega-3 FA AUC for LOVAZA® under the same administration conditions
  • a method of administration or treatment under fasting conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA AUC or Total Omega-3 FA AUC of at least 500% of the EPA+DPA+DHA AUC or Total Omega-3 FA AUC for LOVAZA® under the same administration conditions
  • a method of administration or treatment under fasting conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA AUC or Total Omega-3 FA AUC of at least 600% of the EPA+DPA+DHA AUC or Total Omega-3 FA AUC for LOVAZA® under the same administration conditions
  • a method of administration or treatment under fasting conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA AUC or Total Omega-3 FA AUC of at least 700% of the EPA+DPA+DHA AUC or Total Omega-3 FA AUC for LOVAZA® under the same administration conditions
  • a method of administration or treatment under high fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA Cmax or Total Omega-3 FA Cmax of at least 1 10% of the EPA+DPA Cmax or Total Omega-3 FA Cmax for AMR101 under the same administration conditions
  • a method of administration or treatment under high fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA Cmax or Total Omega-3 FA Cmax of at least 120% of the EPA+DPA Cmax or Total Omega-3 FA Cmax for AMR101 under the same administration conditions
  • a method of administration or treatment under high fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA Cmax or Total Omega-3 FA Cmax of at least 130% of the EPA+DPA Cmax or Total Omega-3 FA Cmax for AMR101 under the same administration conditions
  • a method of administration or treatment under high fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA Cmax or Total Omega-3 FA Cmax of at least 140% of the EPA+DPA Cmax or Total Omega-3 FA Cmax for AMR101 under the same administration conditions
  • a method of administration or treatment under high fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA Cmax or Total Omega-3 FA Cmax of at least 150% of the EPA+DPA Cmax or Total Omega-3 FA Cmax for AMR101 under the same administration conditions
  • a method of administration or treatment under low fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA Cmax or Total Omega-3 FA Cmax of at least 150% of the EPA+DPA Cmax or Total Omega-3 FA Cmax for AMR101 under the same administration conditions
  • a method of administration or treatment under low fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA Cmax or Total Omega-3 FA Cmax of at least 200% of the EPA+DPA Cmax or Total Omega-3 FA Cmax for AMR101 under the same administration conditions
  • a method of administration or treatment under low fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA Cmax or Total Omega-3 FA Cmax of at least 300% of the EPA+DPA Cmax or Total Omega-3 FA Cmax for AMR101 under the same administration conditions
  • a method of administration or treatment under low fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA Cmax or Total Omega-3 FA Cmax of at least 600% of the EPA+DPA Cmax or Total Omega-3 FA Cmax for AMR101 under the same administration conditions
  • a method of administration or treatment under fasting conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA Cmax or Total Omega-3 FA Cmax of at least 150% of the EPA+DPA Cmax or Total Omega-3 FA Cmax for AMR101 under the same administration conditions
  • a method of administration or treatment under fasting conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA Cmax or Total Omega-3 FA Cmax of at least 200% of the EPA+DPA Cmax or Total Omega-3 FA Cmax for AMR101 under the same administration conditions
  • a method of administration or treatment under fasting conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA Cmax or Total Omega-3 FA Cmax of at least 300% of the EPA+DPA Cmax or Total Omega-3 FA Cmax for AMR101 under the same administration conditions
  • a method of administration or treatment under fasting conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA Cmax or Total Omega-3 FA Cmax of at least 400% of the EPA+DPA Cmax or Total Omega-3 FA Cmax for AMR101 under the same administration conditions
  • a method of administration or treatment under fasting conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA Cmax or Total Omega-3 FA Cmax of at least 500% of the EPA+DPA Cmax or Total Omega-3 FA Cmax for AMR101 under the same administration conditions
  • a method of administration or treatment under fasting conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA Cmax or Total Omega-3 FA Cmax of at least 600% of the EPA+DPA Cmax or Total Omega-3 FA Cmax for AMR101 under the same administration conditions 207.
  • a method of administration or treatment under fasting conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA Cmax or Total Omega-3 FA Cmax of at least 700% of the EPA+DPA Cmax or Total Omega-3 FA Cmax for AMR101 under the same administration conditions
  • a method of administration or treatment under high fat meal conditions according to one of the preferred embodiments 03 through 105 resulting in an EPA+DPA AUC or Total Omega-3 FA AUC of at least 110% of the EPA+DPA AUG or Total Omega-3 FA AUC for AMR101 under the same administration conditions
  • a method of administration or treatment under high fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA AUC or Total Omega-3 FA AUC of at least 120% of the EPA+DPA AUC or Total Omega-3 FA AUC for AMR101 under the same administration conditions
  • a method of administration or treatment under high fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA AUC or Total Omega-3 FA AUC of at least 130% of the EPA+DPA AUC or Total Omega-3 FA AUC for AMR101 under the same administration conditions
  • a method of administration or treatment under high fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA AUC or Total Omega-3 FA AUC of at least 140% of the EPA+DPA AUC or Total Omega-3 FA AUC for AMR101 under the same administration conditions
  • a method of administration or treatment under high fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA AUC or Total Omega-3 FA AUC of at least 150% of the EPA+DPA AUC or Total Omega-3 FA AUC for AMR101 under the same administration conditions
  • a method of administration or treatment under low fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA AUC or Total Omega-3 FA AUC of at least 300% of the EPA+DPA AUC or Total Omega-3 FA AUC for AMR101 under the same administration conditions
  • a method of administration or treatment under low fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA AUC or Total Omega-3 FA AUC of at least 400% of the EPA+DPA AUC or Total Omega-3 FA AUC for AMR101 under the same administration conditions
  • a method of administration or treatment under low fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA AUC or Total Omega-3 FA AUC of at least 500% of the EPA+DPA AUC or Total Omega-3 FA AUC for AMR101 under the same administration conditions
  • a method of administration or treatment under low fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA AUC or Total Omega-3 FA AUC of at least 600% of the EPA+DPA AUC or Total Omega-3 FA AUC for AMR101 under the same administration conditions
  • a method of administration or treatment under fasting conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA AUC or Total Omega-3 FA AUC of at least 150% of the EPA+DPA AUC or Total Omega-3 FA AUC for AMR101 under the same administration conditions
  • a method of administration or treatment under fasting conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA AUC or Total Omega-3 FA AUC of at least 200% of the EPA+DPA AUC or Total Omega-3 FA AUC for AMR101 under the same administration conditions 221 .
  • a method of administration or treatment under fasting conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA AUC or Total Omega-3 FA AUC of at least 300% of the EPA+DPA AUC or Total Omega-3 FA AUC for AMR101 under the same administration conditions
  • a method of administration or treatment under fasting conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA AUC or Total Omega-3 FA AUC of at least 400% of the EPA+DPA AUC or Total Omega-3 FA AUC for AMR101 under the same administration conditions
  • a method of administration or treatment under fasting conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA AUC or Total Omega-3 FA AUC of at least 500% of the EPA+DPA AUC or Total Omega-3 FA AUC for AMR101 under the same administration conditions
  • a method of administration or treatment under fasting conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA AUC or Total Omega-3 FA AUC of at least 600% of the EPA+DPA AUC or Total Omega-3 FA AUC for AMR101 under the same administration conditions
  • a method of administration or treatment under fasting conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA AUC or Total Omega-3 FA AUC of at least 700% of the EPA+DPA AUC or Total Omega-3 FA AUC for AMR101 under the same administration conditions
  • a method of administration or treatment under high fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax of at least 100% of the EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax for EPANOVATM under the same administration conditions
  • a method of administration or treatment under high fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax of at least 105% of the EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax for EPANOVATM under the same administration conditions
  • a method of administration or treatment under high fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax of at least 1 10% of the EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax for EPANOVATM under the same administration conditions
  • a method of administration or treatment under high fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax of at least 120% of the EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax for EPANOVATM under the same administration conditions
  • a method of administration or treatment under high fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax of at least 130% of the EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax for EPANOVATM under the same administration conditions
  • a method of administration or treatment under high fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax of at least 140% of the EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax for EPANOVATM under the same administration conditions
  • a method of administration or treatment under low fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax of at least 100% of the EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax for EPANOVATM under the same administration conditions
  • a method of administration or treatment under low fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax of at least 105% of the EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax for EPANOVATM under the same administration conditions
  • a method of administration or treatment under low fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax of at least 1 10% of the EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax for EPANOVATM under the same administration conditions
  • a method of administration or treatment under low fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax of at least 120% of the EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax for EPANOVATM under the same administration conditions
  • a method of administration or treatment under low fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax of at least 130% of the EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax for EPANOVATM under the same administration conditions
  • a method of administration or treatment under low fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax of at least 140% of the EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax for EPANOVATM under the same administration conditions
  • a method of administration or treatment under fasting conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax of at least 100% of the EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax for EPANOVATM under the same administration conditions
  • a method of administration or treatment under fasting conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax of at least 105% of the EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax for EPANOVATM under the same administration conditions
  • a method of administration or treatment under fasting conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax of at least 1 10% of the EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax for EPANOVATM under the same administration conditions
  • a method of administration or treatment under fasting conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax of at least 120% of the EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax for EPANOVATM under the same administration conditions 242.
  • a method of administration or treatment under fasting conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax of at least 130% of the EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax for EPANOVATM under the same administration conditions
  • a method of administration or treatment under fasting conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax of at least 140% of the EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax for EPANOVATM under the same administration conditions
  • a method of administration or treatment under fasting conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax of at least 150% of the EPA+DPA+DHA Cmax or Total Omega-3 FA Cmax for EPANOVATM under the same administration conditions
  • a method of administration or treatment under high fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA AUC or Total Omega-3 FA AUC of at least 100% of the EPA+DPA+DHA AUC or Total Omega-3 FA AUC for EPANOVATM under the same administration conditions
  • a method of administration or treatment under high fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA AUC or Total Omega-3 FA AUC of at least 105% of the EPA+DPA+DHA AUC or Total Omega-3 FA AUC for EPANOVATM under the same administration conditions
  • a method of administration or treatment under high fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA AUC or Total Omega-3 FA AUC of at least 110% of the EPA+DPA+DHA AUC or Total Omega-3 FA AUC for EPANOVATM under the same administration conditions
  • a method of administration or treatment under high fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA AUC or Total Omega-3 FA AUC of at least 120% of the EPA+DPA+DHA AUC or Total Omega-3 FA AUC for EPANOVATM under the same administration conditions
  • a method of administration or treatment under high fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA AUC or Total Omega-3 FA AUC of at least 130% of the EPA+DPA+DHA AUC or Total Omega-3 FA AUC for EPANOVATM under the same administration conditions
  • a method of administration or treatment under high fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA AUC or Total Omega-3 FA AUC of at least 140% of the EPA+DPA+DHA AUC or Total Omega-3 FA AUC for EPANOVATM under the same administration conditions
  • a method of administration or treatment under low fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA AUC or Total Omega-3 FA AUC of at least 100% of the EPA+DPA+DHA AUC or Total Omega-3 FA AUC for EPANOVATM under the same administration conditions
  • a method of administration or treatment under low fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA AUC or Total Omega-3 FA AUC of at least 105% of the EPA+DPA+DHA AUC or Total Omega-3 FA AUC for EPANOVATM under the same administration conditions
  • a method of administration or treatment under low fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA AUC or Total Omega-3 FA AUC of at least 1 10% of the EPA+DPA+DHA AUC or Total Omega-3 FA AUC for EPANOVATM under the same administration conditions
  • a method of administration or treatment under low fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA AUC or Total Omega-3 FA AUC of at least 120% of the EPA+DPA+DHA AUC or Total Omega-3 FA AUC for EPANOVATM under the same administration conditions
  • a method of administration or treatment under low fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA AUC or Total Omega-3 FA AUC of at least 130% of the EPA+DPA+DHA AUC or Total Omega-3 FA AUC for EPANOVATM under the same administration conditions
  • a method of administration or treatment under low fat meal conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA AUC or Total Omega-3 FA AUC of at least 140% of the EPA+DPA+DHA AUC or Total Omega-3 FA AUC for EPANOVATM under the same administration conditions
  • a method of administration or treatment under fasting conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA AUC or Total Omega-3 FA AUC of at least 100% of the EPA+DPA+DHA AUC or Total Omega-3 FA AUC for EPANOVATM under the same administration conditions
  • a method of administration or treatment under fasting conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA AUC or Total Omega-3 FA AUC of at least 105% of the EPA+DPA+DHA AUC or Total Omega-3 FA AUC for EPANOVATM under the same administration conditions
  • a method of administration or treatment under fasting conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA AUC or Total Omega-3 FA AUC of at least 110% of the EPA+DPA+DHA AUC or Total Omega-3 FA AUC for EPANOVATM under the same administration conditions
  • a method of administration or treatment under fasting conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA AUC or Total Omega-3 FA AUC of at least 120% of the EPA+DPA+DHA AUC or Total Omega-3 FA AUC for EPANOVATM under the same administration conditions
  • a method of administration or treatment under fasting conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA AUC or Total Omega-3 FA AUC of at least 130% of the EPA+DPA+DHA AUC or Total Omega-3 FA AUC for EPANOVATM under the same administration conditions 262.
  • a method of administration or treatment under fasting conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA AUC or Total Omega-3 FA AUC of at least 140% of the EPA+DPA+DHA AUC or Total Omega-3 FA AUC for EPANOVATM under the same administration conditions
  • a method of administration or treatment under fasting conditions according to one of the preferred embodiments 103 through 105 resulting in an EPA+DPA+DHA AUC or Total Omega-3 FA AUC of at least 150% of the EPA+DPA+DHA AUC or Total Omega-3 FA AUC for EPANOVATM under the same administration conditions.
  • composition of claim 1 wherein the ratio of EPA to DPA (EPA:DPA) is between 15:1 to 8:1.
  • 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.
  • EPA eicosapentaenoic acid
  • DPA docosapentaenoic acid
  • DHA docosahexaenoic acid

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Epidemiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Oil, Petroleum & Natural Gas (AREA)
  • General Chemical & Material Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Diabetes (AREA)
  • Polymers & Plastics (AREA)
  • Food Science & Technology (AREA)
  • Wood Science & Technology (AREA)
  • Biochemistry (AREA)
  • Hematology (AREA)
  • Obesity (AREA)
  • Zoology (AREA)
  • Neurosurgery (AREA)
  • Cardiology (AREA)
  • Neurology (AREA)
  • Rheumatology (AREA)
  • Urology & Nephrology (AREA)
  • Pain & Pain Management (AREA)
  • Endocrinology (AREA)
  • Emergency Medicine (AREA)
  • Vascular Medicine (AREA)
  • Biomedical Technology (AREA)
  • Botany (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
EP13861394.8A 2012-12-06 2013-12-06 Omega-3-pentaensäure-zusammensetzungen und verfahren zur verwendung Withdrawn EP2929041A4 (de)

Priority Applications (1)

Application Number Priority Date Filing Date Title
EP21170003.4A EP3888646A1 (de) 2012-12-06 2013-12-06 Omega-3-pentaensäure-zusammensetzungen und verfahren zur verwendung

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201261734331P 2012-12-06 2012-12-06
US201361780948P 2013-03-13 2013-03-13
PCT/US2013/073701 WO2014089501A1 (en) 2012-12-06 2013-12-06 Omega-3 pentaenoic acid compositions and methods of use

Related Child Applications (1)

Application Number Title Priority Date Filing Date
EP21170003.4A Division EP3888646A1 (de) 2012-12-06 2013-12-06 Omega-3-pentaensäure-zusammensetzungen und verfahren zur verwendung

Publications (2)

Publication Number Publication Date
EP2929041A1 true EP2929041A1 (de) 2015-10-14
EP2929041A4 EP2929041A4 (de) 2016-06-08

Family

ID=50884050

Family Applications (3)

Application Number Title Priority Date Filing Date
EP21170003.4A Withdrawn EP3888646A1 (de) 2012-12-06 2013-12-06 Omega-3-pentaensäure-zusammensetzungen und verfahren zur verwendung
EP13861394.8A Withdrawn EP2929041A4 (de) 2012-12-06 2013-12-06 Omega-3-pentaensäure-zusammensetzungen und verfahren zur verwendung
EP13861305.4A Withdrawn EP2928462A4 (de) 2012-12-06 2013-12-06 Verfahren zur verabreichung von zusammensetzungen mit docosapentaensäure

Family Applications Before (1)

Application Number Title Priority Date Filing Date
EP21170003.4A Withdrawn EP3888646A1 (de) 2012-12-06 2013-12-06 Omega-3-pentaensäure-zusammensetzungen und verfahren zur verwendung

Family Applications After (1)

Application Number Title Priority Date Filing Date
EP13861305.4A Withdrawn EP2928462A4 (de) 2012-12-06 2013-12-06 Verfahren zur verabreichung von zusammensetzungen mit docosapentaensäure

Country Status (9)

Country Link
EP (3) EP3888646A1 (de)
JP (4) JP2016501249A (de)
KR (3) KR20150103671A (de)
CN (4) CN104937103A (de)
AU (7) AU2013354969A1 (de)
CA (2) CA2894366A1 (de)
IN (2) IN2015DE05009A (de)
MX (2) MX2015007085A (de)
WO (2) WO2014089501A1 (de)

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR101568665B1 (ko) 2015-07-21 2015-11-12 디와이오토 주식회사 모터의 맥동 전류를 증가시키는 브러시 구조
WO2018001844A1 (en) * 2016-06-28 2018-01-04 Nestec S.A. Biomarkers of blood-brain barrier dysfunction
JP2018168139A (ja) * 2016-10-21 2018-11-01 マルハニチロ株式会社 n−3系不飽和脂肪酸を含む中性脂肪値の低減または上昇抑制用の組成物、並びに該組成物の製造におけるn−3系不飽和脂肪酸の使用
JP7099821B2 (ja) * 2017-12-20 2022-07-12 ポッカサッポロフード&ビバレッジ株式会社 Pcsk9阻害剤及びコレステロール代謝改善用食品組成物
EP3586640A1 (de) 2018-06-21 2020-01-01 Nuseed Pty Ltd Dha-angereicherte mehrfach ungesättigte fettsäurezusammensetzungen

Family Cites Families (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB8819110D0 (en) 1988-08-11 1988-09-14 Norsk Hydro As Antihypertensive drug & method for production
GB9901809D0 (en) * 1999-01-27 1999-03-17 Scarista Limited Highly purified ethgyl epa and other epa derivatives for psychiatric and neurological disorderes
GB0016045D0 (en) 2000-06-29 2000-08-23 Laxdale Limited Therapeutic combinations of fatty acids
JP2002125618A (ja) * 2000-10-30 2002-05-08 Katsuyuki Wakatsuki 健康食品
EP1714564A1 (de) * 2005-04-21 2006-10-25 N.V. Nutricia Verfahren zur Behandlung oder Vorbehandlung von Lungeninsuffiziens
WO2007058523A1 (en) 2005-11-17 2007-05-24 N.V. Nutricia Composition with docosapentaenoic acid
NZ573719A (en) * 2006-07-05 2011-08-26 Photonz Corp Ltd Production of ultrapure eicosapentaenoic acid and polar lipids from largely heterotrophic culture of nitzschia laevis
KR100684642B1 (ko) * 2006-09-14 2007-02-22 주식회사 일신웰스 어유 유래 글리세라이드 유지 조성물 및 이의 제조방법
EP2405902B1 (de) * 2009-03-09 2021-07-21 Basf As Zusammensetzungen mit einer fettsäureölmischung und einem tensid sowie verfahren und verwendung dafür
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
CN102458109B (zh) * 2009-04-29 2015-02-11 阿马里纳制药公司 稳定的药物组合物和使用其的方法
KR102012111B1 (ko) * 2009-06-15 2019-08-19 아마린 파마, 인크. 병용 스타틴 요법을 받는 대상체에서 ldl-c 수준을 상승시키지 않으면서 트리글리세리드를 저하시키기 위한 조성물 및 방법
CA2827555A1 (en) * 2011-02-16 2012-08-23 Pivotal Therapeutics, Inc. Cholesterol absorption inhibitor (azetidinone) and omega 3 fatty acids (epa, dha, dpa) for the reduction of cholesterol and for the reduction of cardiovascular events
EP2675445A1 (de) * 2011-02-16 2013-12-25 Pivotal Therapeutics, Inc. Statin und omega-3-fettsäuren (epa, dha und dpa) zur verwendung bei kardiovaskulären erkrankungen
EP2861227A4 (de) * 2012-06-17 2016-01-27 Matinas Biopharma Inc Omega-3 pentaensäurezusammensetzungen und verfahren zur verwendung

Also Published As

Publication number Publication date
EP2928462A2 (de) 2015-10-14
MX2015007082A (es) 2016-01-12
AU2018229440A1 (en) 2018-10-04
CN104937103A (zh) 2015-09-23
CA2894366A1 (en) 2014-06-12
WO2014089511A2 (en) 2014-06-12
JP2021120407A (ja) 2021-08-19
JP2016501249A (ja) 2016-01-18
CN113274378A (zh) 2021-08-20
CN112107570A (zh) 2020-12-22
IN2015MU01505A (de) 2016-05-27
WO2014089511A3 (en) 2014-07-31
JP6881893B2 (ja) 2021-06-02
KR20210059779A (ko) 2021-05-25
CA2894183A1 (en) 2014-06-12
IN2015DE05009A (de) 2015-12-18
CN104902888A (zh) 2015-09-09
AU2013354969A1 (en) 2015-06-11
JP2016501248A (ja) 2016-01-18
AU2020203658A1 (en) 2020-06-25
MX2015007085A (es) 2016-01-12
KR20150103671A (ko) 2015-09-11
EP2929041A4 (de) 2016-06-08
AU2019216634A1 (en) 2019-09-05
EP3888646A1 (de) 2021-10-06
JP2019172697A (ja) 2019-10-10
AU2017232203A1 (en) 2017-10-12
KR20150103009A (ko) 2015-09-09
EP2928462A4 (de) 2016-06-01
AU2021201865A1 (en) 2021-04-22
WO2014089501A1 (en) 2014-06-12
AU2013354979A1 (en) 2015-06-11

Similar Documents

Publication Publication Date Title
US20210205254A1 (en) Omega-3 pentaenoic acid compositions and methods of use
AU2020203658A1 (en) Administering compositions comprising docosapentaenoic acid
WO2014179325A1 (en) Omega-3 fatty acid formulations for use as pharmaceutical treatment
WO2014179341A1 (en) Treatment with omega-3 fatty acid compositions
EP2968246A1 (de) Omega-3-pentaensäure-zusammensetzungen und verfahren zur verwendung
US20140194512A1 (en) Compositions comprising docosapentaenoic acid and methods of use

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20150602

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

DAX Request for extension of the european patent (deleted)
RA4 Supplementary search report drawn up and despatched (corrected)

Effective date: 20160506

RIC1 Information provided on ipc code assigned before grant

Ipc: A61K 31/40 20060101ALI20160429BHEP

Ipc: A61K 31/202 20060101ALI20160429BHEP

Ipc: A61K 31/225 20060101AFI20160429BHEP

Ipc: A23D 9/00 20060101ALI20160429BHEP

Ipc: C11B 5/00 20060101ALI20160429BHEP

Ipc: A61K 9/48 20060101ALI20160429BHEP

Ipc: A23D 9/06 20060101ALI20160429BHEP

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

17Q First examination report despatched

Effective date: 20200507

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20220701