US20160193175A1 - Use of Docosahexaenoic Glycerides For The Treatment Of Tumorous Diseases - Google Patents

Use of Docosahexaenoic Glycerides For The Treatment Of Tumorous Diseases Download PDF

Info

Publication number
US20160193175A1
US20160193175A1 US15/005,181 US201615005181A US2016193175A1 US 20160193175 A1 US20160193175 A1 US 20160193175A1 US 201615005181 A US201615005181 A US 201615005181A US 2016193175 A1 US2016193175 A1 US 2016193175A1
Authority
US
United States
Prior art keywords
dha
pharmaceutical composition
oil
fatty acids
enriched
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US15/005,181
Other versions
US20170333381A9 (en
Inventor
Joan Carles Domingo Pedrol
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.)
Brudy Technology SL
Original Assignee
Brudy Technology SL
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 Brudy Technology SL filed Critical Brudy Technology SL
Priority to US15/005,181 priority Critical patent/US20170333381A9/en
Publication of US20160193175A1 publication Critical patent/US20160193175A1/en
Publication of US20170333381A9 publication Critical patent/US20170333381A9/en
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/215Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids
    • A61K31/22Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acyclic acids, e.g. pravastatin
    • A61K31/23Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acyclic acids, e.g. pravastatin of acids having a carboxyl group bound to a chain of seven or more carbon atoms
    • A61K31/232Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acyclic acids, e.g. pravastatin of acids having a carboxyl group bound to a chain of seven or more carbon atoms having three or more double bonds, e.g. etretinate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/20Carboxylic acids, e.g. valproic acid having a carboxyl group bound to a chain of seven or more carbon atoms, e.g. stearic, palmitic, arachidic acids
    • A61K31/202Carboxylic acids, e.g. valproic acid having a carboxyl group bound to a chain of seven or more carbon atoms, e.g. stearic, palmitic, arachidic acids having three or more double bonds, e.g. linolenic
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/107Emulsions ; Emulsion preconcentrates; Micelles
    • A61K9/1075Microemulsions or submicron emulsions; Preconcentrates or solids thereof; Micelles, e.g. made of phospholipids or block copolymers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • the present invention relates to use of an acid enriched in docosahexaenoic acid for manufacturing a drug for the treatment of a tumorous disease.
  • omega-3 fatty acids and especially the long-chain polyunsaturated fatty acids (PUFAs) such as eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), present in fish and in oils derived therefrom inhibit carcinogenesis and show potential anti-tumour activity.
  • PUFAs long-chain polyunsaturated fatty acids
  • EPA eicosapentaenoic acid
  • DHA docosahexaenoic acid
  • the omega-3 PUFAs principally DHA, reduce the growth of various types of tumorous cells, including breast, bowel, pancreas, and in chronic myeloid leukaemia and melanoma.
  • omega-3 PUFAs selectively inhibit the proliferation of tumorous cells, being less toxic towards normal cells. This different sensitivity to the omega-3 PUFAs cannot be explained simply by differences in the take-up of fatty acids.
  • Various mechanisms have been proposed by which the omega-3 fatty acids might alter the carcinogenic process, notably the following: suppression of biosynthesis of eicosanoids deriving from arachidonic acid (ARA); alterations in the activity of transcription factors, regulation of gene expression and intracellular marking; alteration of oestrogen metabolism; alteration of the generation of free radicals and reactive oxygen species and modifications in membrane fluidity.
  • ARA arachidonic acid
  • the ARA-derived eicosanoids have been associated with tumour development.
  • the omega-3 PUFAs compete with the omega-6 PUFAs as substrates of the desaturases and elongases, the omega-3 PUFAs having greater affinity for said enzymes.
  • the omega-3 fatty acids inhibit cycloxygenase-2 at transcriptional level and compete with the omega-6 fatty acids as substrates of the cycloxygenases in formation of the eicosanoids.
  • omega-3 PUFAs and their metabolytes can exercise some of their anti-tumour effects by affecting the expression of various genes or the activities of the signal-transmission molecules involved in controlling growth, differentiation, cellular apoptosis, angiogenesis and metastasis.
  • the most important are the activated receptor of peroxysomal proliferation, that of nuclear transcription factor ⁇ B, the ras oncogene, protein kinase C, co-enzyme-A-3-hydroxyl-3-methylglutaril reductase, cycloxygenase-2, lipoxygenases and the nitric oxide synthase. It has been shown that the treatment of colon carcinoma cells with DHA alters the characteristics of the cellular membrane and reduce its metastasic capacity.
  • tumour cells have a deficit of anti-oxidant defence systems in comparison with the healthy cells and are thus more susceptible to oxidation damage.
  • the PUFAs are the main intracellular substrates in lipid peroxidation, whether it be by causing damage to the cell membranes, altering cellular composition or the assembly of the cytoskeleton, altering the membrane transport systems or the activity of their enzymes, or inhibiting the reactions of the polymerase. It is therefore reasonable to consider the DHA-enriched cells of the tumour as being more susceptible to oxidative damage.
  • omega-3 fatty acids have an effect on the cell cycle.
  • DHA In vitro treatment with DHA leads to a stoppage in the G 1 /S or G 2 /M phase during the cell cycle in tumorous cells of breast and melanoma.
  • the administration of fish oil rich in omega-3 to rats implanted with a breast tumorous line can prolong replication of the DNA of the tumorous cells thereby delaying progression through the synthesis phase.
  • omega-3 PUFAs can increase the cytotoxicity of several anti-neoplasic agents and the anti-carcinogenic effects of radiotherapy. These effects are possibly mediated by incorporation of the fatty acids into the tumour cell membranes, thus altering physical and functional characteristics.
  • the therapeutic efficacy achieved depends on several factors, such as the bioavailability of the fatty acid, which is in turn related with the chemical structure of which it forms part, the type of omega-3 PUFA used (ALA, EPA or DHA) and the interaction availability between the PUFA and the target cell.
  • omega-3 PUFA used ALA, EPA or DHA
  • DHA is the omega-3 PUFA that provides most effective anti-tumour protection, exceeding that of EPA. This result has also been confirmed in combined EPA+DHA anti-tumour treatments, where the presence of EPA has been observed to diminish the efficacy of the DHA.
  • intratumour administration is the preferred route for the treatment of gliomas. It is essential in this respect that the PUFAs be administered to the patients in such a way as to be easily taken up by the tumorous cells.
  • parenteral administration for example, which is suitable for the treatment of hepatomas, it is essential to have a carrier system such as an emulsion with the additional objective of limiting bonding of the PUFAs to the serum albumin that suppresses their tumour cytotoxicity.
  • a carrier system such as an emulsion with the additional objective of limiting bonding of the PUFAs to the serum albumin that suppresses their tumour cytotoxicity.
  • oral administration suitable for the treatment of lymphomas, following processing and intestinal absorption the PUFAs are transported to the target tissue incorporated into the kilomicrons in the form of triglycerides.
  • the present invention concerns the unexpected discovery that oils with a high content of DHA incorporated into a glyceride, in particular with a content exceeding 50% by weight, present an anti-tumour efficacy greater than the same concentration of DHA in any other chemical form.
  • An object of the present invention is therefore the use of an oil enriched in docosahexaenoic acid (hereinafter also referred to as “DHA”) which is incorporated into a glyceride, for manufacturing a pharmaceutical composition for the treatment of a tumorous disease, with said oil being enriched by a concentration of up to 70% by weight in relation to the total weight of said pharmaceutical composition and with said docosahexaenoic acid being incorporated into a glyceride in a percentage of at least 50% by weight in relation to the total fatty acids in said oil.
  • DHA docosahexaenoic acid
  • oil enriched in docosahexaenoic acid means natural or synthetic derivatives of glycerol containing 50-100% by weight of the docasohexaenoyl group based on the total content of fatty acids.
  • docosahexaenoic acid incorporated into a glyceride is taken to mean a glycerol with the three positions esterified with docosahexaenoic acid.
  • the expression “up to 70% by weight of said enriched oil” means that 70% of the total weight of the pharmaceutical composition corresponds to the DHA-enriched oil.
  • the expression “at least 50% in relation to the total fatty acids” means that the DHA represents at least 50% by weight of the total fatty acids of said enriched oil.
  • the inventors of the present invention have found that using an amount of at least 50% of DHA doubles or even quintuples the anti-tumour effect of said DHA, thereby achieving a high specific tumourous cytotoxic activity without the existence of side-effects.
  • said pharmaceutical composition is in emulsion form.
  • the emulsions can be prepared under apyrogenic and sterile conditions with sufficient physical and chemical stability for administration thereof, including parenteral administration, by means of methods well-known to an expert in the art.
  • the mean diameter of the microemulsion is less than 200 nm.
  • this mean diameter permits parenteral application, thereby administering an effective dose of DHA greater than that obtained by oral administration, and therefore increasing the bioavailability of said acid. This is due to avoiding the loss inherent to intestinal absorption. Furthermore, high concentrations of DHA can be administered by the parenteral route.
  • said pharmaceutical composition in emulsion form is administered by parenteral route.
  • the doses to be administered depend on the type and severity of the pathology to be treated, and there are no dietary restrictions (interactions with foods).
  • the emulsions of the invention can also be administered orally, sublingually, intravenously, intramuscularly, topically, subcutaneously, rectally or even simply by bringing the active ingredient of the emulsion of the invention into contact with the olfactory organs situated at the entrance to the airways in liquid or vapour form. Administration can thus be carried out by spraying, nebulising or atomising the emulsions or by inhalation.
  • said pharmaceutical composition is administered by intramuscular injection.
  • said DHA-enriched oil also includes ecosapentaenoic acid (also referred to as EPA) in a percentage by weight of up to 30% in relation to the total fatty acids in said oil.
  • ecosapentaenoic acid also referred to as EPA
  • the concentration of DHA-enriched oil is in the range of 10-70%, preferably in the range of 10-30% in relation to the total weight of the pharmaceutical composition.
  • the percentage by weight of DHA in relation to the total weight of fatty acids of said enriched oil ranges between 50-100%, preferably between 70 and 90%, and more preferably said percentage by weight of DHA is 70%.
  • the inventors of the present invention have further found that when a pharmaceutical composition in accordance with the present invention is administered, there are no interactions with the components of the anti-neoplasic regime being administered to the patient, since said composition is not metabolised in pathways common to those of metabolisation of the anti-neoplasic drugs.
  • said composition is administered concomitantly with at least one anti-neoplasic drug.
  • anti-neoplasic drug is taken to mean an active ingredient or drug designed for the treatment of a pathology of tumorous or cancerous origin.
  • the invention relates to a stable, non-detergent composition in microemulsion form for administration to human beings, comprising:
  • the initial emulsion is homogenised repeatedly at high pressure to a suitable size and the pH is adjusted to a physiological value (between 6.5-7) with sodium hydroxide (Sigma & Aldrich). Once adjusted to the final volume, the microemulsion is sterilised by filtration (0.22 ⁇ m, Millipore) in its definitive glass receptacle.
  • KG-1a cells derived from an acute myeloid leukaemia which are moreover MDR+, ATCC CCL-246.1
  • Jurkat cells derived from an acute type-T lymphoma, TIB-152
  • HeLa and KB3.1 cells derived from a human epithelial carcinoma, CCL-2
  • HT-29 cells derived from a human colon tumour, HTB-38
  • 435 cells derived from a human breast tumour, MDA-MB-435
  • SH-SY5Y cells derived from a human neuroblastoma, CRL-2266
  • NP-18 cells derived from a human pancreal tumour.
  • the non-tumorous model used was Foreskin cells (non-differentiated epidermis fibroblasts, CRL-1635) and REPTC cells (renal proximal tubule cells, DPK-KTEC-H). All the cellular lines were obtained from the American Type Culture Collection, except for the NP-18 cells which were ceded by the Merck Pharm and Chemistry Bioresearch Laboratory and the REPTCs which were acquired from Dominion Pharmakine. The cell cultures were kept under suitable growth conditions: temperature (37° C.), CO 2 concentration (5%) and humidity (95%) in a special incubator for this purpose. The cells were kept growing in culture bottles, but were transferred to 96-well plates to allow the experiment to be carried out.
  • This method consists of adding the MTT reagent (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide), Sigma & Aldrich), soluble in aqueous medium, to the incubation medium.
  • the viable cells metabolise this compound and it is converted into formazan salt.
  • This salt is a colorimetric compound insoluble in aqueous medium, soluble in DMSO and usable as a measure of cell viability.
  • the method consists of adding 20 ⁇ l per well of an MTT solution of 7.5 mg/ml (in excess). It is incubated for one hour at 37° C.
  • IC 50 defined as the concentration of DHA necessary to reduce the viability of the culture to 50% in relation to control.
  • the first aspect to be studied to determine the action of the DHA as an anti-tumour agent is the specificity of its cytotoxic activity.
  • the model used was little-differentiated Foreskin cells and human renal proximal tubule cells (RPTEC), and the results obtained are shown in Table 1.
  • the cytotoxicity of the product resides in the presence of DHA, since a reduction of the proportion of said acid reduces the cytotoxic power (compare the results of TG-70, TG-50 and TG-20), with the threshold concentration necessary to obtain a therapeutic development being estimated as at least a 50% of content in DHA, although an optimum therapeutic effect is achieved with a DHA concentration of 70%.
  • the second aspect studied was the applicability spectrum of the DHA as an anti-tumour drug, with its chemical structure and the nature of the pathological process at which it is directed being deemed as variables, and maintaining the DHA content at 70%.
  • the results obtained are shown in Table 2.
  • cytotoxic potency which follows the order TG>FA>EE in all the models used, interdependently of their origin. From the results obtained we might stress, firstly, the greater sensitivity to the DHA (5-8 times greater) for tumorous processes of a haematological nature (Jurkat and KG-1a) as against the solid tumours (the rest). As an exception that confirms the rule, great sensitivity is observed in cells derived from a pancreatic tumour (NP18).
  • cytotoxicity of the microemulsions is very much greater in a tumorous line (HeLa cells) than in a normal one (Foreskin cells), depending directly on the concentration of DHA in the glyceride. Moreover, the toxicity observed is attributable solely to the DHA and not to the carrier system (emulsion), since an emulsion of the same characteristics prepared with an oleic acid triglyceride is entirely innocuous.
  • the cytotoxic efficacy of the DHA microemulsions against the different tumorous lines was also analysed, and the results obtained are shown in Table 4, using once again as control a microemulsion of oleic acid prepared under the same conditions as the DHA microemulsion.
  • the results obtained confirm that the emulsions of DHA are at least as effective in their anti-tumour activity as DHA incorporated into a free glyceride.
  • the efficacy is 1.5-3 times greater.
  • the non-toxicity of the carrier system (emulsion) is likewise confirmed, since an emulsion of the same characteristics prepared with an oleic acid triglyceride is entirely innocuous.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Epidemiology (AREA)
  • Emergency Medicine (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Biophysics (AREA)
  • Molecular Biology (AREA)
  • Dispersion Chemistry (AREA)
  • Dermatology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Medicinal Preparation (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The present invention relates to the use of an oil enriched in docosahexaenoic acid, which is conjugated to triglyceride, for manufacturing a pharmaceutical composition for the treatment of a tumorous disease, said oil being enriched in a concentration of up to 70% by weight in relation to the total weight of said pharmaceutical composition, and said docosahexaenoic acid being in a percentage of at least 50% in relation to the total fatty acids in said oil.

Description

    FIELD OF THE INVENTION
  • The present invention relates to use of an acid enriched in docosahexaenoic acid for manufacturing a drug for the treatment of a tumorous disease.
  • BACKGROUND
  • Various evidence from studies on animals and in vitro studies indicate that the omega-3 fatty acids, and especially the long-chain polyunsaturated fatty acids (PUFAs) such as eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), present in fish and in oils derived therefrom inhibit carcinogenesis and show potential anti-tumour activity. In vitro studies with human tumorous lines have shown convincingly that the omega-3 PUFAs, principally DHA, reduce the growth of various types of tumorous cells, including breast, bowel, pancreas, and in chronic myeloid leukaemia and melanoma. Epidemiological data on the association between fish consumption, as a marker for the take-up of omega-3 fatty acids, and the risk of cancer are nevertheless rather less consistent. It has only proved possible to establish that the ingestion of DHA and EPA reduces the growth of tumours in rodents, including tumours of the mammary gland, the colon, the prostate, the liver and the pancreas.
  • Moreover, various studies have shown that the omega-3 PUFAs selectively inhibit the proliferation of tumorous cells, being less toxic towards normal cells. This different sensitivity to the omega-3 PUFAs cannot be explained simply by differences in the take-up of fatty acids. Various mechanisms have been proposed by which the omega-3 fatty acids might alter the carcinogenic process, notably the following: suppression of biosynthesis of eicosanoids deriving from arachidonic acid (ARA); alterations in the activity of transcription factors, regulation of gene expression and intracellular marking; alteration of oestrogen metabolism; alteration of the generation of free radicals and reactive oxygen species and modifications in membrane fluidity.
  • The ARA-derived eicosanoids have been associated with tumour development. Various mechanisms exist by which the omega-3 fatty acids can reduce the biosynthesis of ARA-derived eicosanoids. Firstly, the omega-3 fatty acids are taken into the phospholipids of the membrane, where they partially replace the omega-6 fatty acids. Secondly, the omega-3 PUFAs compete with the omega-6 PUFAs as substrates of the desaturases and elongases, the omega-3 PUFAs having greater affinity for said enzymes. Finally, the omega-3 fatty acids inhibit cycloxygenase-2 at transcriptional level and compete with the omega-6 fatty acids as substrates of the cycloxygenases in formation of the eicosanoids.
  • Furthermore, the omega-3 PUFAs and their metabolytes can exercise some of their anti-tumour effects by affecting the expression of various genes or the activities of the signal-transmission molecules involved in controlling growth, differentiation, cellular apoptosis, angiogenesis and metastasis. The most important are the activated receptor of peroxysomal proliferation, that of nuclear transcription factor κB, the ras oncogene, protein kinase C, co-enzyme-A-3-hydroxyl-3-methylglutaril reductase, cycloxygenase-2, lipoxygenases and the nitric oxide synthase. It has been shown that the treatment of colon carcinoma cells with DHA alters the characteristics of the cellular membrane and reduce its metastasic capacity.
  • The generation of free radicals and reactive oxygen species appears to be involved in the initiation of apoptosis and in natural defences against the transformed cells. Thus the inhibitory effects of the long-chain omega-3 PUFAs on the growth of tumour cells can be explained, at least partly, by the formation of oxidation products, which leads to growth of the cell being halted and to the onset of the apoptosis process. It has been suggested that the tumour cells have a deficit of anti-oxidant defence systems in comparison with the healthy cells and are thus more susceptible to oxidation damage. The PUFAs are the main intracellular substrates in lipid peroxidation, whether it be by causing damage to the cell membranes, altering cellular composition or the assembly of the cytoskeleton, altering the membrane transport systems or the activity of their enzymes, or inhibiting the reactions of the polymerase. It is therefore reasonable to consider the DHA-enriched cells of the tumour as being more susceptible to oxidative damage.
  • There is a certain amount of evidence that the omega-3 fatty acids have an effect on the cell cycle. In vitro treatment with DHA leads to a stoppage in the G1/S or G2/M phase during the cell cycle in tumorous cells of breast and melanoma. In vivo, the administration of fish oil rich in omega-3 to rats implanted with a breast tumorous line can prolong replication of the DNA of the tumorous cells thereby delaying progression through the synthesis phase.
  • Preclinical studies have nevertheless shown that the omega-3 PUFAs can increase the cytotoxicity of several anti-neoplasic agents and the anti-carcinogenic effects of radiotherapy. These effects are possibly mediated by incorporation of the fatty acids into the tumour cell membranes, thus altering physical and functional characteristics.
  • On the other hand, the therapeutic efficacy achieved depends on several factors, such as the bioavailability of the fatty acid, which is in turn related with the chemical structure of which it forms part, the type of omega-3 PUFA used (ALA, EPA or DHA) and the interaction availability between the PUFA and the target cell.
  • Data resulting from a study in which the bioavailability of three concentrated omega-3 PUFAs in the form of ethyl esters, free fatty acids and triglycerides were compared following oral administration showed that the re-esterified triglycerides presented greater bioavailability than the other two preparations.
  • It was also shown in a multi-organ model of carcinogenesis that in a monotherapy treatment DHA is the omega-3 PUFA that provides most effective anti-tumour protection, exceeding that of EPA. This result has also been confirmed in combined EPA+DHA anti-tumour treatments, where the presence of EPA has been observed to diminish the efficacy of the DHA.
  • Finally, the administration route is an important aspect on which the efficacy of the system depends. For example, intratumour administration is the preferred route for the treatment of gliomas. It is essential in this respect that the PUFAs be administered to the patients in such a way as to be easily taken up by the tumorous cells. For parenteral administration, for example, which is suitable for the treatment of hepatomas, it is essential to have a carrier system such as an emulsion with the additional objective of limiting bonding of the PUFAs to the serum albumin that suppresses their tumour cytotoxicity. In the case of oral administration, suitable for the treatment of lymphomas, following processing and intestinal absorption the PUFAs are transported to the target tissue incorporated into the kilomicrons in the form of triglycerides.
  • DESCRIPTION OF THE INVENTION
  • The present invention concerns the unexpected discovery that oils with a high content of DHA incorporated into a glyceride, in particular with a content exceeding 50% by weight, present an anti-tumour efficacy greater than the same concentration of DHA in any other chemical form.
  • An object of the present invention is therefore the use of an oil enriched in docosahexaenoic acid (hereinafter also referred to as “DHA”) which is incorporated into a glyceride, for manufacturing a pharmaceutical composition for the treatment of a tumorous disease, with said oil being enriched by a concentration of up to 70% by weight in relation to the total weight of said pharmaceutical composition and with said docosahexaenoic acid being incorporated into a glyceride in a percentage of at least 50% by weight in relation to the total fatty acids in said oil.
  • In the present invention, the expression “oil enriched in docosahexaenoic acid” means natural or synthetic derivatives of glycerol containing 50-100% by weight of the docasohexaenoyl group based on the total content of fatty acids.
  • In the present invention, “docosahexaenoic acid incorporated into a glyceride” is taken to mean a glycerol with the three positions esterified with docosahexaenoic acid.
  • In the present invention, the expression “up to 70% by weight of said enriched oil” means that 70% of the total weight of the pharmaceutical composition corresponds to the DHA-enriched oil.
  • In the present invention, the expression “at least 50% in relation to the total fatty acids” means that the DHA represents at least 50% by weight of the total fatty acids of said enriched oil.
  • Surprisingly, the inventors of the present invention have found that using an amount of at least 50% of DHA doubles or even quintuples the anti-tumour effect of said DHA, thereby achieving a high specific tumourous cytotoxic activity without the existence of side-effects.
  • In a preferred embodiment, said pharmaceutical composition is in emulsion form.
  • It has been found in practice that when a pharmaceutical composition that contains an oil enriched in docosahexaenoic acid incorporated into a glyceride, as described above, is administered to a patient in emulsion form, a tumorous cytotoxic activity of the order of ten times greater is achieved (as emerges from the examples included below).
  • The emulsions can be prepared under apyrogenic and sterile conditions with sufficient physical and chemical stability for administration thereof, including parenteral administration, by means of methods well-known to an expert in the art.
  • In another preferred embodiment, the mean diameter of the microemulsion is less than 200 nm.
  • Advantageously, this mean diameter permits parenteral application, thereby administering an effective dose of DHA greater than that obtained by oral administration, and therefore increasing the bioavailability of said acid. This is due to avoiding the loss inherent to intestinal absorption. Furthermore, high concentrations of DHA can be administered by the parenteral route.
  • Due to all the foregoing, in a preferred embodiment of the present invention said pharmaceutical composition in emulsion form is administered by parenteral route. The doses to be administered depend on the type and severity of the pathology to be treated, and there are no dietary restrictions (interactions with foods).
  • The emulsions of the invention can also be administered orally, sublingually, intravenously, intramuscularly, topically, subcutaneously, rectally or even simply by bringing the active ingredient of the emulsion of the invention into contact with the olfactory organs situated at the entrance to the airways in liquid or vapour form. Administration can thus be carried out by spraying, nebulising or atomising the emulsions or by inhalation.
  • In another preferred embodiment, said pharmaceutical composition is administered by intramuscular injection.
  • In another preferred embodiment, said DHA-enriched oil also includes ecosapentaenoic acid (also referred to as EPA) in a percentage by weight of up to 30% in relation to the total fatty acids in said oil.
  • In a preferred embodiment, the concentration of DHA-enriched oil is in the range of 10-70%, preferably in the range of 10-30% in relation to the total weight of the pharmaceutical composition.
  • In another preferred embodiment, the percentage by weight of DHA in relation to the total weight of fatty acids of said enriched oil ranges between 50-100%, preferably between 70 and 90%, and more preferably said percentage by weight of DHA is 70%.
  • The inventors of the present invention have further found that when a pharmaceutical composition in accordance with the present invention is administered, there are no interactions with the components of the anti-neoplasic regime being administered to the patient, since said composition is not metabolised in pathways common to those of metabolisation of the anti-neoplasic drugs.
  • Therefore, in another preferred embodiment of the invention, said composition is administered concomitantly with at least one anti-neoplasic drug.
  • In the present invention, “anti-neoplasic drug” is taken to mean an active ingredient or drug designed for the treatment of a pathology of tumorous or cancerous origin.
  • Advantageously, it has been observed that joint administration of a DHA-enriched oil according to the invention together with an anti-neoplasic drug boosts the anti-tumour activity of said treatment and in turn inhibits the side-effects of the anti-tumour treatment.
  • There follow some examples by way of non-restrictive illustration of the present invention.
  • EXAMPLES Example 1 Formulation of a Pharmaceutical Composition in Accordance with the Invention
  • The invention relates to a stable, non-detergent composition in microemulsion form for administration to human beings, comprising:
      • between 1 and 70% by weight of a glyceride that contains DHA as the active ingredient in at least 50% of its fatty acids (Proyecto Empresarial Brudy);
      • between 1 and 1.5% by weight of phospholipids of soya (Lipoid E80, Lipoid);
      • 2.25% by weight of glycerol (Sigma & Aldrich); and
      • up to 100% of water USP (ADESCO).
  • The initial emulsion is homogenised repeatedly at high pressure to a suitable size and the pH is adjusted to a physiological value (between 6.5-7) with sodium hydroxide (Sigma & Aldrich). Once adjusted to the final volume, the microemulsion is sterilised by filtration (0.22 μm, Millipore) in its definitive glass receptacle.
  • Example 2 Evaluation of a DHA Preparation in Different Tumorous Models
  • This study used as an experimental tumorous model KG-1a cells (derived from an acute myeloid leukaemia which are moreover MDR+, ATCC CCL-246.1), Jurkat cells (derived from an acute type-T lymphoma, TIB-152), HeLa and KB3.1 cells (derived from a human epithelial carcinoma, CCL-2), HT-29 cells (derived from a human colon tumour, HTB-38), 435 cells (derived from a human breast tumour, MDA-MB-435), SH-SY5Y cells (derived from a human neuroblastoma, CRL-2266) and NP-18 cells (derived from a human pancreal tumour). The non-tumorous model used was Foreskin cells (non-differentiated epidermis fibroblasts, CRL-1635) and REPTC cells (renal proximal tubule cells, DPK-KTEC-H). All the cellular lines were obtained from the American Type Culture Collection, except for the NP-18 cells which were ceded by the Merck Pharm and Chemistry Bioresearch Laboratory and the REPTCs which were acquired from Dominion Pharmakine. The cell cultures were kept under suitable growth conditions: temperature (37° C.), CO2 concentration (5%) and humidity (95%) in a special incubator for this purpose. The cells were kept growing in culture bottles, but were transferred to 96-well plates to allow the experiment to be carried out. Work was carried out at all times with DHA from fish oil with a variable percentage of DHA in the fatty acids composition (20, 50 or 70%) and incorporated into glycerides (TG), ethyl esters (EE) or as free fatty acid (FA).
  • Cell-viability studies were carried out to evaluate the cytotoxic effect of the different samples. This method consists of adding the MTT reagent (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide), Sigma & Aldrich), soluble in aqueous medium, to the incubation medium. The viable cells metabolise this compound and it is converted into formazan salt. This salt is a colorimetric compound insoluble in aqueous medium, soluble in DMSO and usable as a measure of cell viability. The method consists of adding 20 μl per well of an MTT solution of 7.5 mg/ml (in excess). It is incubated for one hour at 37° C. so that the viable cells metabolise the compound and produce the formazan salt, while the non-viable cells do not metabolise. Following one hour of incubation the cells are precipitated and 100 μl of DMSO (Sigma & Aldrich) is added, which will dissolve the formazan salt. Finally, the absorbency at 550 nm is measured in a plate reader. The viability results are expressed as a percentage of optical density in relation to the controls, deeming the latter to possess 100% viability. Cell viability curves were drawn up on 96-well plates by sowing about 20,000 cells per well (following an analysis of the right number of cells in the light of their growth ratio) with an approximate volume of 200 μl of medium per well. The product-efficiency study was carried out following exposure of the cells to the product for 72 h over a sufficiently wide interval of concentrations to determine the IC50 value, defined as the concentration of active ingredient necessary to reduce cell growth/viability to 50% in relation to control.
  • The experimental results were adjusted to the Hill equation using SigmaPlot 8.0 software in order to determine the IC50, defined as the concentration of DHA necessary to reduce the viability of the culture to 50% in relation to control.
  • Results
  • The first aspect to be studied to determine the action of the DHA as an anti-tumour agent is the specificity of its cytotoxic activity. In this respect we studied the effect of the DHA on the cellular proliferation/viability of non-immortalised cells with a metabolism similar to that of a normal cell. The model used was little-differentiated Foreskin cells and human renal proximal tubule cells (RPTEC), and the results obtained are shown in Table 1.
  • As can be observed from the different structural variants of the DHA studied (triglyceride (TG), free fatty acid (FA) and ethyl ester (EE)), the triglyceride is the one that presents lower cytotoxicity in both models of normal cell. Under these same conditions, but in a tumorous model (HeLa cells), the different DHA variants show a very significant toxicity, with an IC50 of less than 100 μM in all cases. The cytotoxic power remains of the order of TG>FA=EE with the same DHA content (70%).
  • It is also revealed that the cytotoxicity of the product resides in the presence of DHA, since a reduction of the proportion of said acid reduces the cytotoxic power (compare the results of TG-70, TG-50 and TG-20), with the threshold concentration necessary to obtain a therapeutic development being estimated as at least a 50% of content in DHA, although an optimum therapeutic effect is achieved with a DHA concentration of 70%.
  • TABLE 1
    Effect of chemical structure and concentration on the toxicity
    of the DHA in normal and tumorous cellular lines
    COMPOSITION IC50 (μM)
    % CELLULAR LINE
    STRUCTURE DHA Foreskin RPTEC HeLa
    FA 70 123.1 ± 9.0 n.a. 72.2 ± 5.8
    EE 70  566.3 ± 53.9 72.6 ± 8.3 71.0 ± 9.7
    TG 70  716.7 ± 41.9 392.7 ± 37.5 58.6 ± 8.8
    TG 50 1956.0 ± 27.8 413.6 ± 25.6 564.0 ± 69.6
    TG 20 2267.6 ± 27.9 3116.0 ± 158.2 1379.4 ± 286.4
  • The second aspect studied was the applicability spectrum of the DHA as an anti-tumour drug, with its chemical structure and the nature of the pathological process at which it is directed being deemed as variables, and maintaining the DHA content at 70%. The results obtained are shown in Table 2.
  • TABLE 2
    Effect of chemical structure on the cellular toxicity of the
    DHA on cellular lines of different pathological origin
    CELLULAR LINE IC50 (μM)
    FA TG EE
    KG-1a 88.4 ± 8.0 27.1 ± 3.2 120.8 ± 15.1
    Jurkat 61.8 ± 4.5 22.1 ± 1.2 104.6 ± 12.6
    HT-29 158.2 ± 19.6 115.8 ± 17.3 n.a.
    435P 193.1 ± 20.9 129.7 ± 14.9 n.a.
    NP18 n.a. 18.8 ± 3.0 n.a.
    KB3.1 n.a. 116.2 ± 15.7 n.a.
    HeLa 72.2 ± 5.8 58.6 ± 8.8 71.0 ± 9.7
    SH-SY5Y n.a. 86.8 ± 6.8 n.a.
  • The most notable feature is the retention of the cytotoxic potency, which follows the order TG>FA>EE in all the models used, interdependently of their origin. From the results obtained we might stress, firstly, the greater sensitivity to the DHA (5-8 times greater) for tumorous processes of a haematological nature (Jurkat and KG-1a) as against the solid tumours (the rest). As an exception that confirms the rule, great sensitivity is observed in cells derived from a pancreatic tumour (NP18).
  • On the basis of these results it was decided to use as active ingredient a triglyceride with a proportion of DHA equal to or exceeding 70% of the total fatty acids, since this is the one that maintains an optimum relationship between tumorous cytotoxicity and innocuousness to the normal cell.
  • Taking account of these considerations, an emulsion was prepared as described in the preceding section, with a proportion by weight of oil of 10% and containing various proportions of DHA, and a comparative study of its cytotoxic specificity in relation to triglyceride of free DHA was conducted. The results obtained are shown in Table 3, using as control a microemulsion of oleic acid prepared under the same conditions as the DHA microemulsion.
  • A behaviour similar to that described for the free triglyceride was observed. The cytotoxicity of the microemulsions is very much greater in a tumorous line (HeLa cells) than in a normal one (Foreskin cells), depending directly on the concentration of DHA in the glyceride. Moreover, the toxicity observed is attributable solely to the DHA and not to the carrier system (emulsion), since an emulsion of the same characteristics prepared with an oleic acid triglyceride is entirely innocuous.
  • The cytotoxic efficacy of the DHA microemulsions against the different tumorous lines was also analysed, and the results obtained are shown in Table 4, using once again as control a microemulsion of oleic acid prepared under the same conditions as the DHA microemulsion.
  • The results obtained confirm that the emulsions of DHA are at least as effective in their anti-tumour activity as DHA incorporated into a free glyceride. For cells derived from solid tumours or cells of epithelial origin the efficacy is 1.5-3 times greater. The non-toxicity of the carrier system (emulsion) is likewise confirmed, since an emulsion of the same characteristics prepared with an oleic acid triglyceride is entirely innocuous.
  • TABLE 3
    Effect of the content in DHA on the comparative cytotoxicity
    of a glyceride in free form or incorporated into a microemulsion
    on normal and tumorous cellular lines
    IC50 (μM)
    COMPOSITION CELLULAR LINE
    % DHA STRUCTURE FORESKIN HELA
    70 TG  716.7 ± 41.9 58.6 ± 8.8
    μEMULSIÓN  639.3 ± 27.2 19.5 ± 2.0
    50 TG 1956.0 ± 27.8 564.0 ± 69.6
    μEMULSIÓN >5000 545.8 ± 68.5
    20 TG 2267.6 ± 27.9 1379.4 ± 286.4
    μEMULSIÓN >5000 2135.6 ± 189.3
    0 (OLEIC ACID) μEMULSIÓN >5000 >5000
  • TABLE 4
    Study of the comparative cellular cytotoxicity of DHA incorporated
    into a glyceride in free form or in microemulsions on cellular
    lines of different pathological origin
    IC50 (μM)
    free TG 10% microemulsion
    CELLULAR LINE DHA OLEIC ACID
    KG-1a 27.1 ± 3.2 49.5 ± 2.7 >5000
    Jurkat 22.1 ± 1.2 37.2 ± 1.3 >5000
    HT29 115.8 ± 17.3 86.9 ± 3.5 >5000
    435P 129.7 ± 14.9 61.2 ± 2.5 >5000
    HeLa 58.6 ± 8.8 19.5 ± 2.0 >5000
    NP18 18.8 ± 3.0 10.8 ± 1.9 >5000

Claims (21)

1. A method for treating a tumor which comprises administering to a patient a pharmaceutical composition comprising an oil enriched in DHA triglyceride, and wherein said DHA triglyceride being in a percentage of at least 50% in relation to the total fatty acids in said oil.
2. The method according to claim 1, characterized in that said pharmaceutical composition is in emulsion form.
3. The method according to claim 1, wherein said enriched oil comprises eicosapentaenoic acid in a percentage by weight of up to 30% in relation to the total fatty acids of said oil.
4. The method according to claim 1, wherein the concentration of DHA triglyceride is in a percentage of 70%-90% by weight in relation to the total fatty acids of said oil.
5. The method according to claim 1, wherein said DHA triglyceride is in a percentage of 70% by weight in relation to the total fatty acids in said oil.
6. The method according to claim 2, wherein the mean diameter of the emulsion is less than 200 nm.
7. The method according to claim 1, wherein said pharmaceutical composition is administered by parenteral route.
8. The method according to claim 1, wherein said pharmaceutical composition is administered by intratumor injection.
9. The method according to claim 1, wherein said pharmaceutical composition is administered to a patient who is receiving concomitantly at least one antineoplastic drug.
10. The method according to claim 1, wherein the tumor to be treated is one of the group that includes: lung, prostate, bowel, breast, pancreas, brain, central and peripheral nervous system, and melanoma.
11. The method according to claim 1 wherein the tumor to be treated is of a haematological nature.
12. A method for treating a tumor which comprises administering to a patient a pharmaceutical composition comprising an oil enriched in DHA triglyceride, wherein said oil comprises at least 50% by weight of a DHA triglyceride, and wherein said DHA triglyceride being in a percentage of at least 50% in relation to the total fatty acids in said oil.
13. The method according to claim 12, characterized in that said pharmaceutical composition is in emulsion form.
14. The method according to claim 13, wherein said enriched oil comprises eicosapentaenoic acid.
15. The method according to claim 12, wherein the enriched oil comprises at least 10% of the total weight of the pharmaceutical composition.
16. The method according to claim 12, wherein the enriched oil comprises between 50% to 100% of the total weight of the pharmaceutical composition.
17. The method according to claim 12, wherein the enriched oil comprises between 70% to 90% of the total weight of the pharmaceutical composition.
18. The method according to claim 12, wherein the DHA triglyceride is in a percentage of 70% to 90% in relation to the total fatty acids of said oil.
19. The method according to claim 12, wherein the pharmaceutical composition is administered by parenteral route.
20. The method according to claim 12, wherein the pharmaceutical composition is administered by intratumor injection.
21. The method of claim 12, wherein the tumor to be treated is selected from the group consisting of: lung, prostate, bowel, breast, pancreas, brain, central and peripheral nervous system, and melanoma.
US15/005,181 2005-05-12 2016-01-25 Use of Docosahexaenoic Glycerides For The Treatment Of Tumorous Diseases Abandoned US20170333381A9 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US15/005,181 US20170333381A9 (en) 2005-05-12 2016-01-25 Use of Docosahexaenoic Glycerides For The Treatment Of Tumorous Diseases

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
ESP-200501141 2005-05-12
ES200501141A ES2264886B1 (en) 2005-05-12 2005-05-12 USE OF DOCOSAHEXAENOIC ACID FOR THE TREATMENT OF TUMOR DISEASES.
PCT/EP2006/061833 WO2006120120A1 (en) 2005-05-12 2006-04-26 Use of docosahexaenoic glycerides for the treatment of tumorous diseases
US91399407A 2007-11-09 2007-11-09
US12/550,780 US9271954B2 (en) 2005-05-12 2009-08-31 Use of docosahexaenoic glycerides for the treatment of tumorous diseases
US15/005,181 US20170333381A9 (en) 2005-05-12 2016-01-25 Use of Docosahexaenoic Glycerides For The Treatment Of Tumorous Diseases

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US12/550,780 Continuation US9271954B2 (en) 2005-05-12 2009-08-31 Use of docosahexaenoic glycerides for the treatment of tumorous diseases

Publications (2)

Publication Number Publication Date
US20160193175A1 true US20160193175A1 (en) 2016-07-07
US20170333381A9 US20170333381A9 (en) 2017-11-23

Family

ID=36809521

Family Applications (3)

Application Number Title Priority Date Filing Date
US11/913,994 Abandoned US20080207947A1 (en) 2005-05-12 2006-04-26 Use Of Docosahexaenoic Glycerides For The Treatment Of Tumorous Diseases
US12/550,780 Active US9271954B2 (en) 2005-05-12 2009-08-31 Use of docosahexaenoic glycerides for the treatment of tumorous diseases
US15/005,181 Abandoned US20170333381A9 (en) 2005-05-12 2016-01-25 Use of Docosahexaenoic Glycerides For The Treatment Of Tumorous Diseases

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US11/913,994 Abandoned US20080207947A1 (en) 2005-05-12 2006-04-26 Use Of Docosahexaenoic Glycerides For The Treatment Of Tumorous Diseases
US12/550,780 Active US9271954B2 (en) 2005-05-12 2009-08-31 Use of docosahexaenoic glycerides for the treatment of tumorous diseases

Country Status (13)

Country Link
US (3) US20080207947A1 (en)
EP (1) EP1881824B1 (en)
JP (2) JP5893235B2 (en)
CA (1) CA2614192C (en)
DK (1) DK1881824T3 (en)
ES (2) ES2264886B1 (en)
HK (1) HK1113319A1 (en)
IL (1) IL187274A (en)
NO (1) NO336953B1 (en)
NZ (1) NZ563301A (en)
PL (1) PL1881824T3 (en)
PT (1) PT1881824E (en)
WO (1) WO2006120120A1 (en)

Families Citing this family (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2264886B1 (en) * 2005-05-12 2008-02-01 Proyecto Empresarial Brudy, S.L. USE OF DOCOSAHEXAENOIC ACID FOR THE TREATMENT OF TUMOR DISEASES.
AU2006269405B2 (en) * 2005-07-08 2013-01-17 Dsm Ip Assets B.V. Polyunsaturated fatty acids for treatment of dementia and pre-dementia-related conditions
EP1962825B1 (en) * 2005-12-21 2014-04-02 Brudy Technology, S.L. Use of dha for treating a pathology associated with cellular oxidative damage
ES2277557B1 (en) 2005-12-21 2008-07-01 Proyecto Empresarial Brudy, S.L. USE OF DOCOSAHEXAENOIC ACID FOR THE TREATMENT OF OXIDATIVE CELL DAMAGE.
US8343753B2 (en) 2007-11-01 2013-01-01 Wake Forest University School Of Medicine Compositions, methods, and kits for polyunsaturated fatty acids from microalgae
NZ586249A (en) * 2007-12-19 2012-05-25 Prometic Biosciences Inc A combination of medium-chain length fatty acids, salts or triglycerides, gemcitabine, and optionally erlotinib, for the treatment of pancreatic cancer
US20110200644A1 (en) * 2010-02-18 2011-08-18 Martek Biosciences Corporation DHA Ester Emulsions
US20110200645A1 (en) * 2010-02-18 2011-08-18 Martek Biosciences Corporation DHA Free Fatty Acid Emulsions
WO2011103514A1 (en) * 2010-02-18 2011-08-25 Martek Biosciences Corporation Dha triglyceride emulsions
JP5847475B2 (en) * 2010-07-26 2016-01-20 富士フイルム株式会社 Oil-containing composition and oral preparation containing the same
PL2991507T3 (en) 2013-04-22 2020-02-28 Smartfish As Use of a composition comprising fish oil and juice for the treatment and/or post treatment of cancer
KR101665253B1 (en) 2015-06-19 2016-10-25 순천향대학교 산학협력단 Composition for Treating and Prevention of endometrial cancer Including docosahexaenoic acid and triacsin C

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH05331105A (en) * 1991-07-24 1993-12-14 Agency Of Ind Science & Technol Docosahexaenoic acid triglyceride and its production
JPH0860181A (en) * 1994-08-22 1996-03-05 Asahi Chem Ind Co Ltd Highly unsaturated fatty acid-containing oil and fat

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE3721137A1 (en) * 1987-06-26 1989-01-05 Dietl Hans Fat emulsion for intravenous use
US5147854A (en) * 1990-05-22 1992-09-15 Hoffmann-La Roche Inc. Tgf-b compositions and method
JPH05139968A (en) * 1991-11-14 1993-06-08 Sagami Chem Res Center Agent for promoting medullasin activity
JPH0616548A (en) * 1992-06-30 1994-01-25 Sagami Chem Res Center Tissue abnormal growth inhibitor
JPH06116585A (en) * 1992-10-09 1994-04-26 Kanegafuchi Chem Ind Co Ltd Method for purifying fat and oil
JPH1057086A (en) * 1996-08-20 1998-03-03 Asahi Chem Ind Co Ltd Production of highly concentrated docosahexaenoic acid monoglyceride
WO2001017524A1 (en) * 1999-09-09 2001-03-15 Efa Sciences Llc. Methods for treating cell proliferative disorders including cancer
JP2002180082A (en) * 2000-12-11 2002-06-26 Maruha Corp Intake material
US20040052837A1 (en) * 2002-06-27 2004-03-18 William Stillwell Lipid conjugated anti-cancer drugs and methods of use thereof
ES2264886B1 (en) * 2005-05-12 2008-02-01 Proyecto Empresarial Brudy, S.L. USE OF DOCOSAHEXAENOIC ACID FOR THE TREATMENT OF TUMOR DISEASES.

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH05331105A (en) * 1991-07-24 1993-12-14 Agency Of Ind Science & Technol Docosahexaenoic acid triglyceride and its production
JPH0860181A (en) * 1994-08-22 1996-03-05 Asahi Chem Ind Co Ltd Highly unsaturated fatty acid-containing oil and fat

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Machine Translation of JP 2572692, 1997, issuing from JP 05-331105, Google Patents Index *

Also Published As

Publication number Publication date
NO20076382L (en) 2008-02-08
US9271954B2 (en) 2016-03-01
JP2008540484A (en) 2008-11-20
PL1881824T3 (en) 2015-06-30
NZ563301A (en) 2010-08-27
IL187274A0 (en) 2008-06-05
ES2264886A1 (en) 2007-01-16
JP2014208677A (en) 2014-11-06
JP5893235B2 (en) 2016-03-23
HK1113319A1 (en) 2008-10-03
US20080207947A1 (en) 2008-08-28
CA2614192A1 (en) 2006-11-16
NO336953B1 (en) 2015-11-30
IL187274A (en) 2011-11-30
WO2006120120A1 (en) 2006-11-16
CA2614192C (en) 2014-03-18
JP6038841B2 (en) 2016-12-07
US20090318553A1 (en) 2009-12-24
EP1881824A1 (en) 2008-01-30
PT1881824E (en) 2015-03-26
EP1881824B1 (en) 2014-12-24
DK1881824T3 (en) 2015-02-23
ES2531599T3 (en) 2015-03-17
ES2264886B1 (en) 2008-02-01
US20170333381A9 (en) 2017-11-23

Similar Documents

Publication Publication Date Title
US9271954B2 (en) Use of docosahexaenoic glycerides for the treatment of tumorous diseases
EP1275399B1 (en) Method and preparation for preventing and/or treating vascular disorders and secondary disorders associated therewith
KR101833772B1 (en) Use of derivatives of polyunsaturated fatty acids as medicaments
JP4205943B2 (en) Bioavailable compositions of natural and synthetic HCA
RU2272620C2 (en) Method for decreasing cell proliferation based upon (3r,4r)- △8-tetrahydrocannabinol-11-onic acids (hemp-obtained acids)
SK10802001A3 (en) Essential fatty acids in the prevention of cardiovascular events
US11819486B2 (en) Composition for external use on skin for inflammatory diseases
US9597312B2 (en) Treatment of cognitive disorders associated with abnormal dendritic spines using PKC activators
EP1909800B1 (en) Acylglycerophospholipids for treating symptoms concomitant with cancer
WO2006054316A1 (en) Method(s) of preparation, stabilization, composition, and administration of gamma-linolenic acid for brain tumors

Legal Events

Date Code Title Description
AS Assignment

Owner name: BRUDY TECHNOLOGY S.L., SPAIN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:DOMINGO PEDROL, JOAN CARLES;REEL/FRAME:038110/0245

Effective date: 20160223

STPP Information on status: patent application and granting procedure in general

Free format text: ADVISORY ACTION MAILED

STCV Information on status: appeal procedure

Free format text: NOTICE OF APPEAL FILED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

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