EP1341543A2 - Procedes faisant appels a des phospholipides oxydes definis, ainsi que compositions contenant ces derniers, pour la prevention et le traitement de l'atherosclerose - Google Patents

Procedes faisant appels a des phospholipides oxydes definis, ainsi que compositions contenant ces derniers, pour la prevention et le traitement de l'atherosclerose

Info

Publication number
EP1341543A2
EP1341543A2 EP01997274A EP01997274A EP1341543A2 EP 1341543 A2 EP1341543 A2 EP 1341543A2 EP 01997274 A EP01997274 A EP 01997274A EP 01997274 A EP01997274 A EP 01997274A EP 1341543 A2 EP1341543 A2 EP 1341543A2
Authority
EP
European Patent Office
Prior art keywords
group
phosphatidyl
alle
compound
disease
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Withdrawn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
EP01997274A
Other languages
German (de)
English (en)
Other versions
EP1341543A4 (fr
Inventor
Dror Harats
Jacob George
Gideon Halperin
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.)
Notable Labs Ltd
Original Assignee
Vascular Biogenics Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=22956589&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=EP1341543(A2) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Vascular Biogenics Ltd filed Critical Vascular Biogenics Ltd
Priority to EP11189562A priority Critical patent/EP2425841A1/fr
Publication of EP1341543A2 publication Critical patent/EP1341543A2/fr
Publication of EP1341543A4 publication Critical patent/EP1341543A4/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic Table
    • C07F9/02Phosphorus compounds
    • C07F9/06Phosphorus compounds without P—C bonds
    • C07F9/08Esters of oxyacids of phosphorus
    • C07F9/09Esters of phosphoric acids
    • C07F9/10Phosphatides, e.g. lecithin
    • C07F9/106Adducts, complexes, salts of phosphatides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic Table
    • C07F9/02Phosphorus compounds
    • C07F9/06Phosphorus compounds without P—C bonds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/075Ethers or acetals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/075Ethers or acetals
    • A61K31/08Ethers or acetals acyclic, e.g. paraformaldehyde
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/11Aldehydes
    • 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
    • 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
    • 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
    • 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
    • A61K31/235Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids having an aromatic ring attached to a carboxyl group
    • A61K31/24Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids having an aromatic ring attached to a carboxyl group having an amino or nitro group
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/683Diesters of a phosphorus acid with two hydroxy compounds, e.g. phosphatidylinositols
    • A61K31/685Diesters of a phosphorus acid with two hydroxy compounds, e.g. phosphatidylinositols one of the hydroxy compounds having nitrogen atoms, e.g. phosphatidylserine, lecithin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P41/00Drugs used in surgical methods, e.g. surgery adjuvants for preventing adhesion or for vitreum substitution
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/08Vasodilators for multiple indications
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C43/00Ethers; Compounds having groups, groups or groups
    • C07C43/02Ethers
    • C07C43/03Ethers having all ether-oxygen atoms bound to acyclic carbon atoms
    • C07C43/14Unsaturated ethers
    • C07C43/178Unsaturated ethers containing hydroxy or O-metal groups
    • C07C43/1785Unsaturated ethers containing hydroxy or O-metal groups having more than one ether bound
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C43/00Ethers; Compounds having groups, groups or groups
    • C07C43/02Ethers
    • C07C43/03Ethers having all ether-oxygen atoms bound to acyclic carbon atoms
    • C07C43/14Unsaturated ethers
    • C07C43/178Unsaturated ethers containing hydroxy or O-metal groups
    • C07C43/1787Unsaturated ethers containing hydroxy or O-metal groups containing six-membered aromatic rings and having unsaturation outside the aromatic rings

Definitions

  • the present invention relates to defined, oxidized LDL (oxLDL) components for prevention and treatment of atherosclerosis and related disease and, more particularly, to methods and compositions employing oxidized phospholipids effective in inducing mucosal tolerance and inhibiting inflammatory processes contributing to atheromatous vascular disease and sequalae.
  • oxLDL oxidized LDL
  • Cardiovascular disease is a major health risk throughout the industrialized world.
  • Atherosclerosis the most prevalent of cardiovascular diseases, is the principal cause of heart attack, stroke, and gangrene of the extremities, and as such, the principle cause of death in the United States.
  • Atherosclerosis is a complex disease involving many cell types and molecular factors (for a detailed review, see Ross, 1993, Nature 362: 801-809).
  • SMCs smooth muscle cells
  • the advanced lesions of atherosclerosis may occlude the artery concerned, and result from an excessive inflammatory-fibroproliferative response to numerous different forms of insult.
  • shear stresses are thought to be responsible for the frequent occurrence of atherosclerotic plaques in regions of the circulatory system where turbulent blood flow occurs, such as branch points and irregular structures.
  • the first observable event in the formation of an atherosclerotic plaque occurs when inflammatory cells such as monocyte-derived macrophages adhere to the vascular endothelial layer and transmigrate through to the sub-endothelial space. Elevated plasma LDL levels lead to lipid engorgement of the vessel walls, with adjacent endothelial cells producing oxidized low density lipoprotein (LDL). In addition, lipoprotein entrapment by the extracellular matrix leads to progressive oxidation of LDL by lipoxygenases, reactive oxygen species, peroxynitrite and/or myeloperoxidase. These oxidized LDL's are then taken up in large amounts by vascular cells through scavenger receptors expressed on their surfaces.
  • LDL low density lipoprotein
  • Lipid-f ⁇ lled monocytes and smooth-muscle derived cells are called foam cells, and are the major constituent of the fatty streak. Interactions between foam cells and the endothelial and smooth muscle cells surrounding them produce a state of chronic local inflammation which can eventually lead to activation of endothelial cells, increased macrophage apoptosis, smooth muscle cell proliferation and migration, and the formation of a fibrous plaque (Hajjar, DP and Haberland, ME, J.Biol Chem 1997 Sep 12; 272(37):22975-78). Such plaques occlude the blood vessels concerned and thus restrict the flow of blood, resulting in ischemia, a condition characterized by a lack of oxygen supply in tissues of organs due to inadequate perfusion.
  • Oxidized LDL has been implicated in the pathogenesis of atherosclerosis and atherothrombosis, by it's action on monocytes and smooth muscle cells, and by inducing endothelial cell apoptosis, impairing anticoagulant balance in the endothelium.
  • Oxidized LDL also inhibits anti-atherogenic HDL-associated breakdown of oxidized phospholipids (Mertens, A and Holvoet, P, FASEB J 2001 Oct; 15(12):2073-84). This association is also supported by many studies demonstrating the presence of oxidized LDL in the plaques in various animal models of atherogenesis; the retardation of atherogenesis through inhibition of oxidation by pharmacological and/or genetic manipulations; and the promising results of intervention trials with anti-oxidant vitamins (see, for example, Witztum J and Steinberg, D, Trends Cardiovasc Med 2001 Apr- May; 11 (3-4): 93- 102 for a review of current literature). Indeed, oxidized LDL and malondialdehyde
  • MDA-modif ⁇ ed LDL have been recently proposed as accurate blood markers for 1 st and 2 nd stages of coronary artery disease (US Pat. Nos. 6,309,888 to Holvoet et. al. and 6,255,070 to Witztum, et al).
  • autoimmune diseases the immune system recognizes and attacks normally non-antigenic body components (autoantigens), in addition to attacking invading foreign antigens.
  • autoantigens normally non-antigenic body components
  • the autoimmune diseases are classified as auto- (or self-) antibody mediated or cell mediated diseases.
  • Typical autoantibody mediated autoimmune diseases are myasthenia gravis and idiopathic thrombocytopenic purpura (ITP), while typical cell mediated diseases are Hashimoto's thyroiditis and type I (Juvenile) Diabetes.
  • lymphocytes and macrophages which include a predominant population of CD4+ cells (the remainder being CD8+ cells) were found to be more abundant over macrophages in early lesions, as compared witlj the more advanced lesions, in which this ratio tends to reverse.
  • MHC class II HLA-DR and interleukin (IL) receptor as well as leukocyte common antigen (CD45R0) and the very late antigen 1 (VLA-1) integrin.
  • the on-going inflammatory reaction in the early stages of the atherosclerotic lesion may either be the primary initiating event leading to the production of various cytokines by the local cells (i.e endothelial cells, macrophages, smooth muscle cells and inflammatory cells), or it may be that this reaction is a form of the body's defense immune system towards the hazardous process.
  • cytokines which have been shown to be upregulated by the resident cells include TNF- ⁇ , IL-1, IL-2, IL-6, IL-8, IFN- ⁇ and monocyte chemoattractant peptide- 1 (MCP-1).
  • Platelet derived growth factor (PDGF) and insulin-like growth factor (ILGF) which are expressed by all cellular constituents within atherosclerotic plaques have also been shown to be overexpressed, thus possibly intensifying the preexisting inflammatory reaction by a co-stimulatory support in the form of a mitogenic and chemotactic factor.
  • PDGF Platelet derived growth factor
  • ILGF insulin-like growth factor
  • IL-12 - a T-cell growth factor produced primarily by activated monocytes and a selective inducer of Thl cytokine pattern, was found to be overexpressed within lesions as manifested by the abundance of its major heterodimer form p70 and p40 (its dominant inducible protein) mRNA.
  • Atherosclerosis is not a classical autoimmune disease, although some of its manifestations such as the production of the plaque which obstructs the blood vessels may be related to aberrant immune responsiveness.
  • classical autoimmune disease one can often define very clearly the sensitizing autoantigen attacked by the immune system and the component(s) of the immune system which recognize the autoantigen (humoral, i.e. autoantibody or cellular, i.e. lymphocytes). Above all, one can show that by passive transfer of these components of the immune system the disease can be induced in healthy animals, or in the case of humans the disease may be transferred from a sick pregnant mother to her offspring. Many of the above are not prevailing in atherosclerosis. In addition, the disease definitely has common risk factors such as hypertension, diabetes, lack of physical activity, smoking and others, the disease affects elderly people and has a different genetic preponderance than in classical autoimmune diseases.
  • Treatment of autoimmune inflammatory disease may be directed towards supression or reversal of general and/or disease-specific immune reactivity.
  • Aiello for example (US Pat. Nos. 6,034,102 and 6,114,395) discloses the use of estrogen-like compounds for treatment and prevention of atherosclerosis and atherosclerotic lesion progression by inhibition of inflammatory cell recruitment.
  • Medford et al (US Pat. No. 5,846,959) disclose methods for the prevention of formation of oxidized PUFA, for treatment of cardiovascular and non-cardiovascular inflammatory diseases mediated by the cellular adhesion molecule VCAM-1.
  • Falb US Pat. No. 6,156,500 designates a number of cell signaling and adhesion molecules abundant in atherosclerotic plaque and disease as potential targets of anti-inflammatory therapies.
  • Ox LDL is chemotactic for T-cells and monocytes. Ox LDL and its byproducts are also known to induce the expression of factors such as monocyte chemotactic factor 1, secretion of colony stimulating factor and platelet activating properties, all of which are potent growth stimulants.
  • monocyte chemotactic factor 1 secretion of colony stimulating factor and platelet activating properties, all of which are potent growth stimulants.
  • the active involvement of the cellular immune response in atherosclerosis has recently been substantiated (Stemme S, et al, Proc Natl Acad Sci USA 1995; 92: 3893-97), who isolated CD4+ within plaques clones responding to Ox LDL as stimuli. The clones corresponding to Ox LDL (4 out of 27) produced principally interferon- ⁇ rather than IL-4. It remains to be seen whether the above T-cell clones represent mere contact with the cellular immune system with the inciting strong immunogen (Ox LDL) or that this reaction provides means of combat
  • Ox LDL Antibodies to Ox LDL have been hypothesized as playing an active role in lipoprotein metabolism. Thus, it is known that immune complexes of Ox LDL and its corresponding antibodies are taken up more efficiently by macrophages in suspension as compared with Ox LDL. No conclusions can be drawn from this consistent finding on the pathogenesis of atherosclerosis since the question of whether the accelerated uptake of Ox LDL by the macrophages is beneficial or deleterious has not yet been resolved. Important data as to the significance of the humoral immune system in atherogenesis comes from animal models.
  • Intravenous administration of autoantigens has been found to induce immune suppression through a mechanism called clonal anergy.
  • Clonal anergy causes deactivation of only immune attack T-cells specific to a particular antigen, the result being a significant reduction in the immune response to this antigen.
  • the autoimmune response-promoting T-cells specific to an autoantigen once anergized, no longer proliferate in response to that antigen.
  • This reduction in proliferation also reduces the immune reactions responsible for autoimmune disease symptoms (such as neural tissue damage that is observed in MS).
  • oral administration of autoantigens (or immunodominant fragments) in a single dose and in substantially larger amounts than those that trigger "active suppression” may also induce tolerance through anergy (or clonal deletion).
  • a method of treatment has also been disclosed that proceeds by active suppression. Active suppression functions via a different mechanism from that of clonal anergy. This method, discussed extensively in PCT Application PCT/US93/01705, involves oral or mucosal administration of antigens specific to the tissue under autoimmune attack. These are called “bystander antigens”.
  • This treatment causes regulatory (suppressor) T-cells to be induced in the gut-associated lymphoid tissue (GALT), or bronchial associated lymphoid tissue (BALT), or most generally, mucosa associated lymphoid tissue (MALT) (MALT includes GALT and BALT).
  • T-cells elicited by the bystander antigen are targeted to the locus of autoimmune attack where they mediate the local release of certain immunomodulatory factors and cytokines, such as transforming growth factor beta (TGF- ⁇ ), interleukin-4 (IL-4), and/or interleukin-10 (IL-10).
  • TGF- ⁇ transforming growth factor beta
  • IL-4 interleukin-4
  • IL-10 interleukin-10
  • IL-4 and IL-10 are also antigen-nonspecific immunoregulatory cytokines.
  • IL-4 in particular enhances (T helper Th2) Th 2 response, i.e., acts on T-cell precursors and causes them to differentiate preferentially into Th 2 cells at the expense of Th 1 responses.
  • Th2 enhances (T helper Th2) Th 2 response, i.e., acts on T-cell precursors and causes them to differentiate preferentially into Th 2 cells at the expense of Th 1 responses.
  • IL-4 also indirectly inhibits Thi exacerbation.
  • IL-10 is a direct inhibitor of Thi responses.
  • a Boston based company called Autoimmune has carried out several human experiments for preventing diabetes, multiple sclerosis, rheumatoid arthritis and uveitis.
  • the results of the human experiments have been less impressive than the non-human ones, however there has been some success with the prevention of arthritis.
  • immunogenic compounds intended for mucosal as well as intravenous or intraperitoneal administration should optimally be adaptable to nasal and other membranous routes of administration.
  • phospholipids bearing lipid-soluble active compounds may be incorporated into compositions for trans-dermal and trans-membranal application (US Pat. No. 5,985,292 to Fournerou et al) and phospholipid derivatives can be incorporated into liposomes and biovectors for drug delivery (see, for example US Pat. Nos. 6,261,597 and 6,017,513 to Kurtz and Betbeder, et al, respectively).
  • Modified phospholipid derivatives mimicing platelet activation factor (PAF) structure are known to be pharmaceutically active in variety of disorders and diseases, effecting such functions as vascular permeability, blood pressure, heart function inhibition etc.
  • PAF platelet activation factor
  • Oxidation of phospholipids occurs in vivo through the action of free radicals and enzymatic reactions abundant in atheromatous plaque.
  • preparation of oxidized phospholipids usually involves simple chemical oxidation of a native LDL or LDL phospholipid component.
  • Investigators studying the role of oxidized LDL have employed, for example, ferrous ions and ascorbic acid (Itabe, H, et al, J.Biol. Chem. 1996; 271 :33208-217) and copper sulfate (George, J. et al, Atherosclerosis. 1998; 138:147-152; Ameli, S.
  • 5,561,052 discloses a method of producing oxidized lipids and phospholipids using copper sulfate and superoxide dismutase to produce oxidized arachadonic or linoleic acids and oxidized LDL for diagnostic use.
  • the oxidation techniques employed are non-specific, yielding a variety of oxidized products, necessitating either further purification or use of impure antigenic compounds. This is of even greater concern with native LDL, even if purified.
  • X is a C ! - 2 chain
  • Y is selected from the group consisting of:
  • R 3 is selected from the group consisting of H, acyl, alkyl, phosphatidyl choline, phosphatidyl ethanolamine, phosphatidyl serine, phosphatidyl cardiolipin and phosphatidyl inisitol.
  • R 3 is a non-phosphatidyl moeity, and as such the compound is a diglyceride.
  • at least one of Ri and R2 is: wherein X is a C 1 - 24 chain, Y is selected from the group consisting of:
  • Z is selected from the group consisting of:
  • compositions for prevention and/or treatment of atherosclerosis, cardiovascular disease, cerebrovascular disease, peripheral vascular disease, stenosis, restenosis and/or in-stent- stenosis in a subject in need thereof comprising, as an active ingredient, a therapeutically effective amount of a compound selected from the group having a formula:
  • X is C ⁇ . 2
  • Y is selected from the group consisting of:
  • R 3 is selected from the group consisting of H, acyl, alkyl, phosphatidyl choline, phosphatidyl ethanolamine, phosphatidyl serine, phosphatidyl cardiolipin and phosphatidyl inisitol and a pharmaceutically acceptable carrier.
  • R 3 is a non-phosphatidyl moeity, and as such the compound is a diglyceride.
  • At least one of Rj and R 2 is:
  • X is a C ⁇ chain
  • Z is selected from the group consisting of:
  • At least one of Ai and A 2 is CH 2 .
  • this molecule is known as ALLE.
  • the pharmaceutical composition is designed for inducing tolerance to oxidized LDL via mucosal administration.
  • the pharmaceutical composition is designed for nasal, oral, subcutaneous or intra- peritoneal administration, alone or in combination with additional routes of immunomodulation.
  • the compound reduces immune reactivity to oxidized LDL in said subject.
  • the pharmaceutical composition is packaged and identified for use in the prevention and/or treatment of at least one disorder selected from the group consisting of atherosclerosis, cardiovascular disease, cerebrovascular disease, peripheral vascular disease, stenosis, restenosis and/or in-stent-stenosis.
  • the pharmaceutical composition further comprises a therapeutically effective amount of at least one additional compound selected from the group consisting of HMG CoA reductase inhibitors (Statins) mucosal adjuvants, corticosteroids, anti-inflammatory compounds, analgesics, growth factors, toxins, and additional tolerizing antigens.
  • a pharmaceutical composition for prevention and/or treatment of a disease, syndrome or condition selected from the group consisting of atherosclerosis, cardiovascular disease, cerebrovascular disease, peripheral vascular disease, stenosis, restenosis and/or in-stent-stenosis in a subject in need thereof comprising, as an active ingredient, a therapeutically effective amount of a synthetic LDL derivative, or pharmaceutically acceptable salts thereof, the composition further comprising a pharmaceutically acceptable carrier.
  • a method of prevention and/or treatment of atherosclerosis, cardiovascular disease, cerebrovascular disease, peripheral vascular disease, stenosis, restenosis and/or in-stent-stenosis in a subject in need thereof comprising administering a therapeutically effective amount of a compound, said compound selected from the group having a formula:
  • Aj and A 2 are independently selected from the group consisting of CH 2 and CO; and (ii) Rj or R 2 are each independently selected from the group consisting of an alkyl chain 1-27 carbons in length and:
  • X is a Cj_ 2 chain
  • Y is selected from a group consisting of:
  • Z is selected from the group consisting of:
  • R 3 is selected from the group consisting of H, acyl, alkyl, phosphatidyl choline, phosphatidyl ethanolamine, phosphatidyl serine, phosphatidyl cardiolipin and phosphatidyl inisitol and a pharmaceutically acceptable carrier.
  • At least one of Rj and R 2 is:
  • X is a Cj. 24 chain
  • Z is selected from the group consisting of:
  • At least one of A and A 2 is CH 2.
  • the compound is l-hexadecyl-2-(5'-oxo-pentanyl)- ⁇ «- glycero-3-phosphocholine ester, 3- hexadecyl-2-(5'-oxo-pentanyl)-5 , n- glycero-1-phosphocholine and racemic mixtures thereof (ALLE).
  • the compound is administered via mucosal administration.
  • the administration of the compound is nasal, oral, subcutaneous or intra- peritoneal administration, alone or in combination with additional routes of immunomodulation.
  • the administration of the compound reduces immune reactivity to oxidized LDL in said subject.
  • the compound is administered in addition to a therapeutically effective amount of at least one additional compound selected from the group consisting of HMG CoA reductase inhibitors (Statins), mucosal adjuvants, corticosteroids, anti-inflammatory compounds, analgetics, growth factors, toxins, and additional tolerizing antigens.
  • HMG CoA reductase inhibitors Statins
  • mucosal adjuvants corticosteroids
  • anti-inflammatory compounds analgetics
  • growth factors growth factors
  • toxins and additional tolerizing antigens.
  • a method of synthesizing an oxidized phospholipid comprising: (a) providing a phospholipid backbone including two fatty acid side chains, wherein at least one of the fatty acid side chains is a mono-unsaturated fatty acid; and (b) oxidizing the double bond of the mono-unsaturated fatty acid to thereby generate the oxidized phospholipid.
  • the phospholipid backbone further includes a moiety selected from the group consisting of H, acyl, alkyl, phosphatidyl choline, phosphatidyl ethanolamine, phosphatidyl serine, phosphatidyl cardiolipin and phosphatidyl inisitol.
  • the mono unsaturated fatty acid is C 2 - 15 .
  • the oxidized phospholipid is l-palmitoyl-2-oxovaleroyl-sn-glycero-3-phosphocholine, (POVPC), and the mono-unsaturated fatty acid is 5-hexenoic acid.
  • POVPC l-palmitoyl-2-oxovaleroyl-sn-glycero-3-phosphocholine
  • the mono-unsaturated fatty acid is 5-hexenoic acid.
  • FIG. 1 is a flow chart depicting the synthesis of 2,5' Aldehyde lecitin ether, l-hexadecyl-2-(5'-oxo-pentanyl)-sn-glycero-3-phosphocholine ether (for D-ALLE) or 3-hexadecyl-2-(5 ' -oxo-pentanyl)-sn-glycero- 1 -phosphocholine ether (for L-ALLE) . (ALLE);
  • FIG. 2 is a flow chart depicting the synthesis of POVPC
  • FIG. 3 is a graphic representation demonstrating inhibition of early atherogenesis in apoE-deficient mice by intra peritoneal immunization with mixed D- and L- isomers of ALLE.
  • Atherogenesis is expressed as the area of atheromatous lesions in the aortic sinus 6 weeks following the 4 immunization;
  • FIG. 4 is a graphic representation demonstrating inhibition of early atherogenesis in apoE-deficient mice by oral tolerance induced by feeding ALLE.
  • Atherogenesis is expressed as the area of atheromatous lesions in the aortic sinus 8 weeks after the last feeding;
  • FIG. 5 is a graphic representation demonstrating inhibition of early atherogenesis in apoE-deficient mice by mucosal tolerance induced by exposure to L-ALLE.
  • Atherogenesis is expressed as the area of atheromatous lesions in the aortic sinus 8 weeks after the last oral or nasal exposure;
  • FIG. 6 is a graphic representation demonstrating suppression of immune reactivity to atheroslerotic plaque antigens induced by oral exposure to synthetic oxidized phospholipids L-ALLE and POVPC.
  • L-ALLE lmg/mouse L-ALLE
  • POVPC POVPC
  • FIG. 7 is a graphic representation demonstrating inhibition of progression of late-stage atherogenesis in apoE-deficient mice by oral tolerance induced by synthetic oxidized phospholipids D-ALLE, L-ALLE or POVPC.
  • Atherogenesis is expressed as the area of atheromatous lesions in the aortic sinus 12 weeks after the first feeding, as compared to the lesion scores of untreated 24.5 week old mice before feeding (Time 0); FIG.
  • FIG. 8 is a graphic representation demonstrating reduction of triglyceride content of VLDL in 24.5 week old apoE-deficient mice induced by feeding synthetic oxidized phospholipids D-ALLE, L-ALLE or POVPC.
  • Triglyceride content (Tg, mg/ml) was measured by enzymatic colorimetric method in the VLDL fractions following separation of pooled blood samples on FPLC, as described in the materials and methods section that follows;
  • FIG. 9 is a graphic representation demonstrating reduction of cholesterol content of VLDL in 24.5 week old apoE-deficient mice induced by feeding synthetic oxidized phospholipids D-ALLE, L-ALLE or POVPC.
  • Cholesterol content (Cholesterol, mg/ml) was measured by enzymatic colorimetric method in the VLDL fractions following separation of pooled blood samples on FPLC, as described in the materials and methods section that follows.
  • the present invention is of methods and compositions employing synthetic oxidized phospholipids effective in inducing mucosal tolerance and inhibiting inflammatory processes contributing to atheromatous vascular disease and sequalae.
  • oxidized LDL and LDL components have been the targets of numerous therapies for prevention and treatment of heart disease, cerebral-vascular disease and peripheral vascular disease.
  • a method of inducing immune tolerance to oxidized LDL in a subject such as a human being.
  • immune tolerance can be used in the prevention and/or treatment of disorders associated with plaque formation, including but not limited to atherosclerosis, atherosclerotic cardiovascular disease, cerebrovascular disease, peripheral vascular disease, stenosis, restenosis and in-stent-stenosis.
  • atherosclerotic cardiovascular disease are myocardial infarction, coronary arterial disease, acute coronary syndromes, congestive heart failure, angina pectoris and myocardial ischemia.
  • peripheral vascular disease are gangrene, diabetic vasculopathy, ischemic bowel disease, thrombosis, diabetic retinopathy and diabetic nephropathy.
  • cerebrovascular disease are stroke, cerebrovascular inflammation, cerebral hemorrhage and vertebral arterial insufficiency.
  • Stenosis is occlusive disease of the vasculature, commonly caused by atheromatous plaque and enhanced platelet activity, most critically affecting the coronary vasculature. Restenosis is the progressive re-occlusion often following reduction of occlusions in stenotic vasculature. In cases where patency of the vasculature requires the mechanical support of a stent, in-stent-stenosis may occur, re-occluding the treated vessel.
  • the method is effected by administering to the subject a therapeutically effective amount of a synthetic ester derivatives of oxidized phospholipid (hereinafter also referred to as an esterified oxidized phospholipid).
  • a synthetic ester derivatives of oxidized phospholipid hereinafter also referred to as an esterified oxidized phospholipid.
  • ester derivatives of oxidized phospholipids suitable for use by the present method include POVPC (see Examples section below for full formula), POVPC, PGPC (see Watson, AD et al J. Biol. Chem. 1997 272:13597-607) and derivatives having the general formula:
  • Rj or R 2 are each independently selected from the group consisting of an alkyl chain 1-27 carbons in length;
  • X is Cj. 2
  • Y is selected from the group consisting of:
  • Z is selected from the group consisting of:
  • R 3 is selected from the group consisting of H, acyl, alkyl, phosphatidyl choline, phosphatidyl ethanolamine, phosphatidyl serine, phosphatidyl cardiolipin and phosphatidyl inisitol.
  • C j_ n is defined as a carbon backbone consisting of a carbon atom covalently bonded to a chain of n-1 carbons.
  • R 3 is selected from a group consisting of non-phosphatidyl moieties, and as such the resultant compound is not a phospholipid, rather a diglyceride compound.
  • Such diglyceride compounds retain similar structure characteristics and as such in all probability would posses antigenicity and immune tolerizing activity.
  • these compounds can also be used in prevention and/or treatment of atherosclerosis related disorders, and employed and applied similarly to the oxidized phospholipid derivatives described herein.
  • phospholipids and phospholipid metabolites have been clearly implicated in the pathogenesis, and therefore potential treatment of additional, non-atherosclerosis-related diseases.
  • diseases and syndromes include oxidative stress of aging (Onorato JM, et al, Annal N Y Acad Sci 1998 Nov 20;854:277-90), rheumatoid arthritis (RA)(Paimela L, et al.
  • the method of the invention may be used for prevention and/or treatment of non-atherosclerosis related diseases such as aging, RA, juvenile RA, IBD and cancer.
  • the synthetic esterified oxidized phospholipids can be synthesized using prior art approaches (Ou Z., Ogamo A., Guo L., Konda Y., Harigaya Y. and Nakagawa Y. Anal. Biochem. 227: 289-294, 1995).
  • the synthetic ester derivatives of an oxidized phospholipid utilized by this aspect of the present invention are synthesized using a novel synthesis method.
  • a method of synthesizing an esterified oxidized phospholipid is effected by first providing a phospholipid backbone including two fatty acid side chains, at least one of the fatty acid side chains being a mono-unsaturated fatty (preferably C2-I5) acid followed by oxidizing the double bond of the mono-unsaturated fatty acid thereby generating the esterified oxidized phospholipid.
  • Examples of phospholipid backbones suitable for synthesis of an esterified oxidized phospholipid according to the teachings of the present invention include, but are not limited to lecithin, which includes two fatty acid side chains, and lysolecithin which includes a single side chain and as such must undergo an additional synthesis step of adding an additional fatty acid side chain prior to oxidation.
  • the phospholipid backbone When utilized to synthesize POVPC, the phospholipid backbone includes 5-hexenoic acid as the mono-unsaturated fatty acid side chain.
  • Such a reaction provides specific, desired oxidized phospholipids, traversing the need to perform complicated separations and purification.
  • immune tolerance can also be established by using an oxidized phospholipid ether (hereinafter also referred to as an etherified oxidized phospholipid).
  • a method of inducing immune tolerance to oxidized LDL which method is effected by administering to a subject an oxidized phospholipid ether.
  • ALLE (preferably a mixture of 1 -hexadecyl-2-(5 ' -oxo-pentanyl)-.s?z-glycero-3 -phosphocholine ether
  • the immune tolerance inducing compounds described herein can be administered per se, or in a pharmaceutical composition where it is mixed with suitable carriers or excipients.
  • a pharmaceutical composition refers to a preparation of one or more of the active ingredients described herein with other chemical components such as physiologically suitable carriers and excipients.
  • the purpose of a pharmaceutical composition is to facilitate administration of a compound to an organism.
  • active ingredient refers to the compounds (e.g.,
  • physiologically acceptable carrier and “pharmaceutically acceptable carrier” which may be interchangeably used refer to a carrier or a diluent that does not cause significant irritation to an organism and does not abrogate the biological activity and properties of the administered compound.
  • An adjuvant is included under these phrases.
  • excipient refers to an inert substance added to a pharmaceutical composition to further facilitate administration of an active ingredient.
  • excipients include calcium carbonate, calcium phosphate, various sugars and types of starch, cellulose derivatives, gelatin, vegetable oils and polyethylene glycols.
  • Suitable routes of administration may, for example, include oral, rectal, transmucosal, especially transnasal, intestinal or parenteral delivery, including intramuscular, subcutaneous and intramedullary injections as well as intrathecal, direct intraventricular, intravenous, inrtaperitoneal, intranasal, or intraocular injections.
  • compositions of the present invention may be manufactured by processes well known in the art, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or lyophilizing processes.
  • compositions for use in accordance with the present invention thus may be formulated in conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries, which facilitate processing of the active ingredients into preparations which, can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen.
  • the active ingredients of the pharmaceutical composition may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hank's solution, Ringer's solution, or physiological salt buffer.
  • physiologically compatible buffers such as Hank's solution, Ringer's solution, or physiological salt buffer.
  • penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art.
  • the pha ⁇ naceutical • composition can be formulated readily by combining the active compounds with pharmaceutically acceptable carriers well known in the art. Such carriers enable the pharmaceutical composition to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions, and the like, for oral ingestion by a patient.
  • Pharmacological preparations for oral use can be made using a solid excipient, optionally grinding the resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries if desired, to obtain tablets or dragee cores.
  • Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carbomethylcellulose; and/or physiologically acceptable polymers such as polyvinylpyrrolidone (PVP).
  • disintegrating agents may be added, such as cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
  • Dragee cores are provided with suitable coatings.
  • suitable coatings For this purpose, concentrated sugar solutions may be used which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, titanium dioxide, lacquer solutions and suitable organic solvents or solvent mixtures.
  • Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
  • compositions which can be used orally include push- fit capsules made of gelatin as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol.
  • the push-fit capsules may contain the active ingredients in admixture with filler such as lactose, binders such as starches, lubricants such as talc or magnesium stearate and, optionally, stabilizers.
  • the active ingredients may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols.
  • stabilizers may be added. All formulations for oral administration should be in dosages suitable for the chosen route of administration.
  • the compositions may take the form of tablets or lozenges formulated in conventional manner.
  • the active ingredients for use according to the present invention are conveniently delivered in the form of an aerosol spray presentation from a pressurized pack or a nebulizer with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichloro-tetrafluoroethane or carbon dioxide.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichloro-tetrafluoroethane or carbon dioxide.
  • the dosage unit may be determined by providing a valve to deliver a metered amount.
  • Capsules and cartridges of, e.g., gelatin for use in a dispenser may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.
  • compositions described herein may be formulated for parenteral administration, e.g., by bolus injection or continuos infusion.
  • Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multidose containers with optionally, an added preservative.
  • the compositions may be suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • compositions for parenteral administration include aqueous solutions of the active preparation in water-soluble form. Additionally, suspensions of the active ingredients may be prepared as appropriate oily or water based injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acids esters such as ethyl oleate, triglycerides or liposomes. Aqueous injection suspensions may contain substances, which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol or dextran.
  • the suspension may also contain suitable stabilizers or agents which increase the solubility of the active ingredients to allow for the preparation of highly concentrated solutions.
  • the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile, pyrogen-free water based solution, before use.
  • a suitable vehicle e.g., sterile, pyrogen-free water based solution
  • the pharmaceutical composition of the present invention may also be formulated in rectal compositions such as suppositories or retention enemas, using, e.g., conventional suppository bases such as cocoa butter or other glycerides.
  • compositions suitable for use in context of the present invention include compositions wherein the active ingredients are contained in an amount effective to achieve the intended purpose. More specifically, a therapeutically effective amount means an amount of active ingredients effective to prevent, alleviate or ameliorate symptoms of a disorder (e.g., atherosclerosis) or prolong the survival of the subject being treated. Determination of a therapeutically effective amount is well within the capability of those skilled in the art, especially in light of the detailed disclosure provided herein.
  • the therapeutically effective amount or dose can be estimated initially from in vitro and cell culture assays.
  • a dose can be formulated in animal models to achieve a desired concentration or titer. Such information can be used to more accurately determine useful doses in humans.
  • Toxicity and therapeutic efficacy of the active ingredients described herein can be determined by standard pharmaceutical procedures in vitro, in cell cultures or experimental animals.
  • the data obtained from these in vitro and cell culture assays and animal studies can be used in formulating a range of dosage for use in human.
  • the dosage may vary depending upon the dosage form employed and the route of administration utilized.
  • the exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient's condition. (See e.g., Fingl, et al., 1975, in "The Pharmacological Basis of Therapeutics", Ch. 1 p.l).
  • Dosage amount and interval may be adjusted individually to provide plasma or brain levels of the active ingredient are sufficient to induce or suppress angiogenesis (minimal effective concentration, MEC).
  • MEC minimum effective concentration
  • the MEC will vary for each preparation, but can be estimated from in vitro data. Dosages necessary to achieve the MEC will depend on individual characteristics and route of administration. Detection assays can be used to determine plasma concentrations.
  • dosing can be of a single or a plurality of administrations, with course of treatment lasting from several days to several weeks or until cure is effected or diminution of the disease state is achieved.
  • compositions to be administered will, of course, be dependent on the subject being treated, the severity of the affliction, the manner of administration, the judgment of the prescribing physician, etc.
  • compositions of the present invention may, if desired, be presented in a pack or dispenser device, such as an FDA approved kit, which may contain one or more unit dosage forms containing the active ingredient.
  • the pack may, for example, comprise metal or plastic foil, such as a blister pack.
  • the pack or dispenser device may be accompanied by instructions for administration.
  • the pack or dispenser may also be accommodated by a notice associated with the container in a form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals, which notice is reflective of approval by the agency of the form of the compositions or human or veterinary administration. Such notice, for example, may be of labeling approved by the U.S. Food and Drug Administration for prescription drugs or of an approved product insert.
  • Compositions comprising a preparation of the invention formulated in a compatible pharmaceutical carrier may also be prepared, placed in an appropriate container, and labeled for treatment of an indicated condition, as if further detailed above.
  • the present invention illustrates for the first time that synthetic derivatives of oxidized phospholipids can be used to prevent and treat atherosclerosis in a subject, while being devoid of limitations inherent to treatments utilizing biologically derived forms of oxidized LDL.
  • the present invention also provides a novel approach for synthesizing esterified oxidized phopholipids.
  • the present invention also provides a novel form of an oxidized phospholipid ether, utilizable for treatment of atherosclerosis and related disorders.
  • Apo-E deficient mice used in our experiments are from the atherosclerosis prone strain C57BL/6J-Apoe talunc . Mice homozygous for the
  • Apoe 1 " 11 TM 0 mutations show a marked increase in total plasma cholesterol levels which is unaffected by age or sex. Fatty streaks in the proximal aorta are found at 3 months of age. The lesions increase with age and progress to lesions with less lipid but more elongated cells, typical of a more advanced stage of pre-atherosclerotic lesion.
  • the Apoe talunc mutant strain was developed in the laboratory of Dr. Nobuyo Maeda at University of North Carolina at Chapel Hill.
  • the 129-derived E14Tg2a ES cell line was used.
  • the plasmid used is designated as pNMC109 and the founder line is T-89.
  • the C57BL/6J strain was produced by backcrossing the Apoe 1 " 111 " 10 mutation 10 times to C57BL/6J mice (11,12).
  • mice were maintained at the Sheba Hospital Animal Facility (Tel-Hashomer, Israel) on a 12-hour light/dark cycle, at 22-24°C and fed a no ⁇ nal fat diet of laboratory chow (Purina Rodent Laboratory Chow No. 5001) containing 0.027%) cholesterol, approximately 4.5% total fat, and water, ad libitum.
  • laboratory chow Purina Rodent Laboratory Chow No. 5001
  • ALLE D+L The phospholipid ether analog (ALLE D+L) was coupled to purified protein derivative from tuberculin (PPD) at a ratio of 1.6/1.
  • the stock solution of ALLE (D+L) was dissolved in ethanol (99 mg/ml).
  • 5 mg ALLE (D+L) 50.5 ⁇ l from stock solution was diluted to 5mg/ml with 0.25M phosphate buffer, pH 7.2, by stirring on ice.
  • 1.5 mg of D- and L- ALLE in 300 ⁇ l of phosphate buffer were added to 0.6 mg PPD dissolved in 300 ⁇ l of phosphate buffer.
  • l-Ethyl-3- (3-dimethylaminopropyI)-carbodiimid-HCl (5 mg; Sigma, St.Louis, MO) dissolved in 50 ⁇ l of water was added by stirring at 4° for 20 min. The remaining active sites were blocked with lOO ⁇ l of 1M glycine. Coupled compounds were dialyzed against phosphate-buffered saline (PBS), adjusted to 3ml with PBS and stored at 4°. Immunization with 0.3ml (150 ⁇ g) antigen per mouse was performed intraperitoneally 4 times every 2 weeks.
  • PBS phosphate-buffered saline
  • Subcutaneous immunization with Human oxidized LDL Human oxidized LDL was prepared from human plasma pool (d-1.019 to 1.063g/ml by ultracentrifugation) and was Cu-oxidized overnight (by adding 15 ⁇ l ImM CuS0 4 to each ml of LDL previously diluted to lmg/ml concentration). The oxidized LDL was dialyzed against saline-Tris-EDTA and filtrated. For immunization, oxidized LDL was dissolved in PBS and mixed with equal volumes of Freund's incomplete adjuvant. Immunizations were performed by single subcutaneous injection of 50 ⁇ g antigen/mouse in 0.2ml volume. One to three days following the last oral administration the mice received one immunization, and were sacrificed 7-10 days post immunization.
  • FPLC Analysis Fast Protein Liquid Chromatography analysis of cholesterol and lipid content of lipoproteins was performed using Superose 6 HR 10/30 column (Amersham Pharmacia Biotech, Inc, Peapack, NJ) on a FPLC system (Pharmacia LKB. FRAC-200, Pharmacia, Peapack, NJ). A minimum sample volume of 300 ⁇ l (blood pooled from 3 mice was diluted 1 :2 and filtered before loading) was required in the sampling vial for the automatic sampler to completely fill the 200 ⁇ l sample loop. Fractions 10-40 were collected, each fraction contained 0.5 ml.
  • a 250 ⁇ l sample from each fraction was mixed with freshly prepared cholesterol reagent or triglyceride reagent respectively, incubated for 5 minutes at 37°C and assayed spectrophotometrically at 500nm.
  • Assessment of Atherosclerosis Quantification of atherosclerotic fatty streak lesions was done by calculating the lesion size in the aortic sinus as previously described (16) and by calculating the lesion size in the aorta. Briefly, after perfusion with saline Tris EDTA, the heart and the aorta were removed from the animals and the peripheral fat cleaned carefully.
  • the upper section of the heart was embedded in OCT medium (10.24%> w/w polyvinyl alcohol; 4.26% w/w polyethylene glycol; 85.50%) w/w nonreactive ingredients) and frozen. Every other section (10 ⁇ m thick) throughout the aortic sinus (400 ⁇ m) was taken for analysis. The distal portion of the aortic sinus was recognized by the three valve cusps that are the junctions of the aorta to the heart. Sections were evaluated for fatty streak lesions after staining with oil-red O. Lesion areas per section were scored on a grid (17) by an observer counting unidentified, numbered specimens. The aorta was dissected from the heart and surrounding adventitious tissue was removed.
  • Proliferation assays Mice were fed ALLE, POVPC or PBS as described for assessment of atherosclerosis, and then immunized one day following the last feeding with oxidized LDL prepared from purified human LDL as described above. Proliferation was assayed eight days after immunization with the oxidized LDL as follows: Spleens or lymph nodes were prepared by meshing the tissues on 100 mesh screens. (Lymph nodes where immunization was performed, and spleens where no immunization performed). Red blood cells were lysed with cold sterile double distilled water (6ml) for 30 seconds and 2ml of NaCl 3.5% was added.
  • mice were fed with ALLE or PBS and immunized with oxidized LDL as described above, one day following the last fed dose. Draining inguinal lymph nodes (taken 8 days after immunization) were collected from 3 mice from each of the groups, for the proliferation studies, lxl 0 6 cells per ml were incubated in triplicates for 72 h in 0.2 ml of culture medium in microtiter wells in the presence 10 ⁇ g/ml oxidized LDL. Proliferation was measured by the incorporation of [ H] thymidine into DNA during the final 12 h of incubation. The results are expressed as the stimulation index (S.I.): the ratio of the mean radioactivity (cpm) of the antigen to the mean background (cpm) obtained in the absence of the antigen. Standard deviation was always ⁇ 10% of the mean cpm.
  • S.I. stimulation index
  • Hexadecyl-glycerol ether D-Acetone-glycerol (4g) for synthesis of L-ALLE or L- Acetone glycerol for synthesis of D-ALLE, powdered potassium hydroxide (approximately lOg) and hexadecyl bromide (9.3g) in benzene (100ml) were stirred and refluxed for 5 hours, removing the water formed by azeotropic distillation (compare W.J. Baumann and H.K. Mangold, J. Org. Chem. 29: 3055, 1964 and F. Paltauf, Monatsh. 99:1277, 1968).
  • the volume of the solvent was gradually reduced to about 20ml and the cooled mixture was dissolved in ether (100ml). The solution was washed with water (50ml) twice, and the solvent was removed in vacuo. The residue was refluxed in 100ml of methanol :water: concentrated hydrochloric acid 90:10:5 for 10 min. The product was extracted with ether (200ml) washed with water (50ml), 10% sodium hydroxide (20ml) and again with water (volumes of 20ml) until neutral.
  • 5-hexenyl-methane sulfonate 5-Hexedecenol (1.9ml) and dry pyridine (5ml) were mixed in a culture tube fitted with teflon-protected screw cap and cooled to between -4 and -10 °C in an ice-salt bath. Methanesulfonyl chloride (10ml) was added in portions of 1ml over 60 minutes, and the mixture was kept at 4°C for 48 hours. Ice (20g) was added, the mixture was allowed to stand for 30 minutes, and the product was extracted with ether (200ml). The organic phase was washed with water (20ml), 10%> hydrochloric acid, 10% sodium bicarbonate (20ml) and again with water (20ml).
  • the mixture was heated in an oil bath at 100°C for 12 hours. After cooling, 300ml of ether and 150ml of ice-cold water were added, and the reaction mixture was transferred to a separatory funnel. The separated ether phase was washed consecutively with 50ml of ice water, 1% potassium carbonate solution (until basic) and 50ml of water, then dried over anhydrous sodium sulfate. A second extraction of the combined aqueous phase increased the yield slightly. The solvent was evaporated, the residue was treated with 150ml of warm petroleum ether and the solution was cooled at 4° over night.
  • 2-Bromoethyl dichlorophosphate was prepared by dropwise addition of freshly distilled 2-bromoethanol (0.5M, prepared as described in Gilman Org. Synth. 12:117, 1926) to an ice-cooled solution of freshly distilled phosphorous oxychloride (0.5M) in dry chloroform over a one hour period, followed by 5 hours reflux and vacuum distillation (bp 66-68° at 0.4-0.5mm Hg). The reagent was stored (-20°) under nitrogen in small sealed ampoules prior to use (Hansen W.H et al. Lipids 17(6):453-459, 1982). l-Hexadecanoyl-2-(5'-oxo)-pentanoyl-sn-3-glycerophosphocholine (POVPC)
  • the mixture was directly chromatographed on silica gel 60 (40g) and the product l-hexadecanoyl-2-(5'-hexenoyl)-sn-3-glycerophosphocholine (compound II, Figure 2) was eluted with chlorofor ⁇ r.methanol 25:75 (2.8g).
  • the eluent was dissolved in 30% hydrogen peroxide: formic acid 4:15 and kept overnight at room temperature. Water (50ml) was added and the product was extracted with chloroform :methanol 2:1 (100ml) and the organic phase was washed again with water.
  • mice 19 female 5-7 weeks old Apo E/C 57 mice were divided into 3 groups.
  • group A the mice were immunized intreperitoneally, as described in Materials and Methods section above, with 150 ⁇ g/mouse L-ALLE + D-ALLE once every 2 weeks (0.3 ml/mouse) for 8 weeks.
  • group B the mice were immunized with tuberculin toxin Purified Protein Derivative (PPD) once every 2 weeks (0.3 ml/mouse).
  • mice from all three groups were bled prior to immunization (Time 0), and at one week after the second immunization for determination of anti-ox LDL antibodies, anti-ALLE antibodies and lipid profile. Mice in which the antibody response did not plateau were bled at successive 2 week intervals until plateau was reached. Atherosclerosis assessment was performed as described above, and spleens collected 6 weeks after the fourth immunization. The mice from all groups were weighed at 2 week intervals throughout the experiment. All mice were fed normal chow-diet containing 4.5%> fat by weight (0.02% cholesterol) and water ad libitum.
  • mice 34 male 8-10 week old Apo E/C 57 mice were divided into three groups.
  • oral tolerance was induced by administration by gavage of L-ALLE + D-ALLE suspended in PBS (5 mg/ml, 1 mg/mouse) for 5 days every other day.
  • group B mice received 10 ⁇ g/mouse L-ALLE + D-ALLE suspended in PBS for 5 days every other day. (0.2 ml/mouse).
  • mice in group C received PBS (containing the same volume of ethanol as in groups A+B). Mice were bled prior to feeding (Time 0) and at the conclusion of the experiment (End) for determination of lipid profile. Atherosclerosis was assessed in heart, aorta, and serum as described above 8 weeks after the last feeding. Mice were weighed every 2 weeks during the experiment. All mice were fed normal chow-diet containing 4.5% fat by weight (0.02% cholesterol) and water ad libitum.
  • Weight is weight in grams; “Choi” is serum cholesterol and “TG” is serum triglycerides, expressed in mg/dL.
  • the results depicted in Table 2 demonstrate a striking attenuation of atherosclerotic progression measured in the tissues of mice fed low doses (10 ⁇ g - 1 mg/ mouse) of ALLE, compared to unexposed control mice. No significant effect is apparent on other parameters measured, such as weight gain, triglyceride or cholesterol blood levels, or immune competence, as measured by the levels of the immunosuppressive cytokine TGF- ⁇ .
  • the synthetic oxidized LDL component ALLE is a potent inducer of oral tolerance, conferring significant (>50%) protection from atherosclerosis in these genetically susceptible apoE-deficient mice, similar to the protection achieved with peritoneal immunization (see Figure 1).
  • mice in group C received PBS administered orally and nasally (containing the same volume of ethanol as in groups A+B). Mice were bled prior to feeding (Time 0) and at the conclusion of the experiment (End) for determination of lipid profile.
  • Atherosclerosis was assessed in heart and aorta as described above, 8 weeks after the last feeding. Mice were weighed every 2 weeks during the experiment. All mice were fed normal chow-diet containing 4.5% fat by weight
  • Weight is weight in grams; “Choi” is serum cholesterol and “TG” is serum triglycerides, expressed in mg/dL.
  • the results depicted in Table 3 demonstrate effective (as effective as oral tolerance) inhibition of atherogenesis measured in the tissues of mice receiving nasal exposure to low doses (10 ⁇ g/ mouse) of ALLE, compared to unexposed control mice.
  • Induction of nasal tolerance like oral tolerance, had no significant effect on other parameters measured, such as weight gain, triglyceride or cholesterol blood levels, or immune competence, as measured by the levels of the immunosuppressive cytokine TGF- ⁇ .
  • the synthetic oxidized LDL component ALLE is a potent inducer of nasal as well as oral tolerance, conferring significant (approximately 50%) protection from atherogenesis in these genetically susceptible apoE-deficient mice, similar to the protection achieved induction of oral tolerance alone.
  • Example V Suppression of specific anti- ox LDL immune reactivity in genetically predisposed (apoE-deficient) mice by oral administration of L-ALLE and
  • POVPC l-Hexadecanoyl-2-(5'-oxo-pentanoyl)- OT-glycerophosphocholine
  • ALLE is susceptible to breakdown in the liver.
  • Lymphocyte proliferation in response to oral exposure to both POVPC and the more stable analog ALLE was measured in apoE-deficient mice. 8 male, 6 week old Apo E/C 57 mice were divided into 3 groups.
  • mice in group A received oral administration of 200 ⁇ l PBS every other day for 5 days.
  • Immune reactivity was stimulated by immunization with Human oxidized LDL as described above in the Materials and Methods section, one day after the last feeding.
  • lymph nodes were collected for assay of proliferation. All mice were fed normal chow-diet containing 4.5% fat by weight (0.02% cholesterol) and water ad libitum.
  • oral administration of the synthetic oxidized LDL component L-ALLE is an effective method of attenuating the cellular immune response to immunogenic and atherogenic plaque components in these genetically susceptible apoE-deficient mice.
  • Figure 4 also demonstrates a similar, if less effective inhibition of proliferation with oral administration of the less stable synthetic oxidized LDL component POVPC.
  • group B oral tolerance was induced with 1 mg/mouse D-ALLE suspended in 0.2 ml PBS, administered per os, as described above, every other day for 5 days.
  • group C oral tolerance was induced with 1 mg/mouse POVPC suspended in 0.2 ml PBS, administered by gavage, as described above, every other day for 5 days.
  • Control group D received oral administration of PBS (containing the same volume of ethanol as in groups A,B,C). Oral administration of the tested antigens took place every 4 weeks (5 oral feedings; every other day) starting at 24.5 weeks age, during 12 weeks (3 sets of feedings).
  • mice were bled prior to feeding (Time 0), after the 2 nd set of feeding and at the conclusion of the experiment (End) for determination of lipid profile, lipid fractionation and plasma collection.
  • Atherosclerosis was assessed as described above in the heart and aorta and spleens collected for proliferation assay 12 weeks after the first feeding. Weight was recorded every 2 weeks throughout the experiment. All mice were fed normal chow-diet containing 4.5% fat by weight (0.02% cholesterol) and water ad libitum.
  • Weight is weight in grams; “Cholesterol” is serum cholesterol and “Triglyceride” is serum triglycerides, expressed in mg dL.
  • ALLE and POVPC are individually potent inducers of oral tolerance, conferring nearly complete protection from atheromatous progression (as compared with lesion scores at 24.5 weeks) in these genetically susceptible apoE-deficient mice.
  • atherogenesis is accompanied by a significant reduction in VLDL cholesterol and triglycerides, as measured by FPLC
  • mice are a model of lipoprotein oxidation in atherogenesis. Demonstration of oxidation-specific epitopes in lesions and high titers of autoantibodies to malondialdehyde-lysine in serum. Arterioscler Thromb 1994; 14: 605-616

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Immunology (AREA)
  • Emergency Medicine (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Urology & Nephrology (AREA)
  • Vascular Medicine (AREA)
  • Transplantation (AREA)
  • Surgery (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

L'invention concerne de nouvelles formes synthétiques de phospholipides oxydés éthérifiés ainsi que des procédés mettant en oeuvre ces derniers, pour prévenir et traiter l'athérosclérose et d'autres troubles connexes. En outre, l'invention concerne des procédés de synthèse des phospholipides oxydés estérifiés et d'utilisation de ces derniers pour prévenir et traiter l'athérosclérose et d'autres troubles connexes.
EP01997274A 2000-11-24 2001-11-22 Procedes faisant appels a des phospholipides oxydes definis, ainsi que compositions contenant ces derniers, pour la prevention et le traitement de l'atherosclerose Withdrawn EP1341543A4 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
EP11189562A EP2425841A1 (fr) 2000-11-24 2001-11-22 Procédés utilisant et compositions contenant des phospholipides oxydés définis pour la prévention et le traitement de l'athérosclérose

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US25257400P 2000-11-24 2000-11-24
US252574P 2000-11-24
PCT/IL2001/001080 WO2002041827A2 (fr) 2000-11-24 2001-11-22 Procedes faisant appels a des phospholipides oxydes definis, ainsi que compositions contenant ces derniers, pour la prevention et le traitement de l'atherosclerose

Publications (2)

Publication Number Publication Date
EP1341543A2 true EP1341543A2 (fr) 2003-09-10
EP1341543A4 EP1341543A4 (fr) 2009-09-02

Family

ID=22956589

Family Applications (2)

Application Number Title Priority Date Filing Date
EP01997274A Withdrawn EP1341543A4 (fr) 2000-11-24 2001-11-22 Procedes faisant appels a des phospholipides oxydes definis, ainsi que compositions contenant ces derniers, pour la prevention et le traitement de l'atherosclerose
EP11189562A Withdrawn EP2425841A1 (fr) 2000-11-24 2001-11-22 Procédés utilisant et compositions contenant des phospholipides oxydés définis pour la prévention et le traitement de l'athérosclérose

Family Applications After (1)

Application Number Title Priority Date Filing Date
EP11189562A Withdrawn EP2425841A1 (fr) 2000-11-24 2001-11-22 Procédés utilisant et compositions contenant des phospholipides oxydés définis pour la prévention et le traitement de l'athérosclérose

Country Status (9)

Country Link
EP (2) EP1341543A4 (fr)
JP (3) JP4162486B2 (fr)
KR (1) KR100865142B1 (fr)
CN (1) CN100577174C (fr)
AU (2) AU1846102A (fr)
CA (1) CA2429817C (fr)
IL (1) IL156015A0 (fr)
MX (1) MXPA03004517A (fr)
WO (1) WO2002041827A2 (fr)

Families Citing this family (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR100865142B1 (ko) * 2000-11-24 2008-10-24 바스큘라 바이오제닉스 리미티드 죽상 경화증의 예방 및 치료를 위해 정의된 산화 인지질을사용하는 방법 및 이를 함유하는 조성물
US6838452B2 (en) * 2000-11-24 2005-01-04 Vascular Biogenics Ltd. Methods employing and compositions containing defined oxidized phospholipids for prevention and treatment of atherosclerosis
AU2003243806A1 (en) * 2002-07-15 2004-02-02 Bernd Binder Lipid oxidation products for inhibiting inflammation
KR101201935B1 (ko) * 2004-07-09 2012-11-16 바스큘라 바이오제닉스 리미티드 산화 인지질류의 개선된 제조 방법
US7807847B2 (en) 2004-07-09 2010-10-05 Vascular Biogenics Ltd. Process for the preparation of oxidized phospholipids
AU2007200090B2 (en) * 2004-07-09 2011-09-22 Vascular Biogenics Ltd. Improved Process for the Preparation of Oxidized Phospholipids
US8569529B2 (en) 2007-01-09 2013-10-29 Vascular Biogenics Ltd. High-purity phospholipids
US9006217B2 (en) 2007-01-09 2015-04-14 Vascular Biogenics Ltd. High-purity phospholipids
WO2009116750A2 (fr) * 2008-03-17 2009-09-24 이화여자대학교 산학협력단 Composition pour le revêtement d'endoprothèse assurant une prévention contre la resténose et endoprothèse fabriquée au moyen de cette composition
JP5833922B2 (ja) 2008-08-12 2015-12-16 ジンファンデル ファーマシューティカルズ インコーポレイテッド 疾患危険因子を同定する方法
US8846315B2 (en) 2008-08-12 2014-09-30 Zinfandel Pharmaceuticals, Inc. Disease risk factors and methods of use
US8999960B2 (en) 2008-10-08 2015-04-07 Vascular Biogenics Ltd. Oxidized thiophospholipid compounds and uses thereof
EP2348866B1 (fr) * 2008-11-06 2015-01-07 Vascular Biogenics Ltd. Composés lipidiques oxydés et leurs utilisations
WO2011083469A1 (fr) * 2010-01-05 2011-07-14 Vascular Biogenics Ltd. Traitement avec le vb-201
EP2665479B1 (fr) 2011-01-10 2019-06-26 Zinfandel Pharmaceuticals, Inc. Procédés et produits médicamenteux destinés au traitement de la maladie d'alzheimer
AU2012301602B2 (en) 2011-09-01 2015-09-03 Vascular Biogenics Ltd. Formulations and dosage forms of oxidized phospholipids
MX2014015197A (es) * 2012-06-11 2015-11-16 Cleveland Clinic Foundation Tratamiento y prevencion de la enfermedad cardiovascular y trombosis.
US9771385B2 (en) 2014-11-26 2017-09-26 Vascular Biogenics Ltd. Oxidized lipids
CA2968790A1 (fr) 2014-11-26 2016-06-02 Vascular Biogenics Ltd. Lipides oxydes et traitement ou prevention de la fibrose

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0121088A1 (fr) * 1983-03-05 1984-10-10 A. Nattermann & Cie. GmbH O-Acyl-alcanediol-phospholipides, procédé pour les préparer et compositions pharmaceutiques les contenant
EP0142333A2 (fr) * 1983-11-08 1985-05-22 Ono Pharmaceutical Co., Ltd. Dérivés du glycérol
WO1987005904A1 (fr) * 1986-03-24 1987-10-08 The University Of Sydney Analogues antigeniques du facteur d'activation des thrombocytes (paf)
JPS6354386A (ja) * 1986-08-26 1988-03-08 Takeda Chem Ind Ltd リン脂質およびその用途
JPH01258691A (ja) * 1988-04-06 1989-10-16 Nippon Oil & Fats Co Ltd リン脂質誘導体及びその製造方法
ES2019552A6 (es) * 1990-04-11 1991-06-16 Menarini Lab Procedimiento para la preparacion de glicerofosfolipidos.
WO1995023592A1 (fr) * 1994-03-04 1995-09-08 The University Of British Columbia Compositions a base de liposomes et procedes de traitement de l'atherosclerose

Family Cites Families (39)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
NL154600B (nl) 1971-02-10 1977-09-15 Organon Nv Werkwijze voor het aantonen en bepalen van specifiek bindende eiwitten en hun corresponderende bindbare stoffen.
NL154598B (nl) 1970-11-10 1977-09-15 Organon Nv Werkwijze voor het aantonen en bepalen van laagmoleculire verbindingen en van eiwitten die deze verbindingen specifiek kunnen binden, alsmede testverpakking.
NL154599B (nl) 1970-12-28 1977-09-15 Organon Nv Werkwijze voor het aantonen en bepalen van specifiek bindende eiwitten en hun corresponderende bindbare stoffen, alsmede testverpakking.
US3901654A (en) 1971-06-21 1975-08-26 Biological Developments Receptor assays of biologically active compounds employing biologically specific receptors
US3853987A (en) 1971-09-01 1974-12-10 W Dreyer Immunological reagent and radioimmuno assay
US3867517A (en) 1971-12-21 1975-02-18 Abbott Lab Direct radioimmunoassay for antigens and their antibodies
NL171930C (nl) 1972-05-11 1983-06-01 Akzo Nv Werkwijze voor het aantonen en bepalen van haptenen, alsmede testverpakkingen.
US3850578A (en) 1973-03-12 1974-11-26 H Mcconnell Process for assaying for biologically active molecules
US3935074A (en) 1973-12-17 1976-01-27 Syva Company Antibody steric hindrance immunoassay with two antibodies
US3996345A (en) 1974-08-12 1976-12-07 Syva Company Fluorescence quenching with immunological pairs in immunoassays
US4034074A (en) 1974-09-19 1977-07-05 The Board Of Trustees Of Leland Stanford Junior University Universal reagent 2-site immunoradiometric assay using labelled anti (IgG)
US3984533A (en) 1975-11-13 1976-10-05 General Electric Company Electrophoretic method of detecting antigen-antibody reaction
US4098876A (en) 1976-10-26 1978-07-04 Corning Glass Works Reverse sandwich immunoassay
CH642665A5 (en) 1979-02-08 1984-04-30 Rudolf Berchtold Process for the preparation of 1-(omega-carboxyalkyl)-2-alkyl- glycero-3-phosphatides
US4329302A (en) * 1980-06-27 1982-05-11 Board Of Regents, The University Of Texas System Synthetic phosphoglycerides possessing platelet activating properties
US4879219A (en) 1980-09-19 1989-11-07 General Hospital Corporation Immunoassay utilizing monoclonal high affinity IgM antibodies
US4614796A (en) * 1983-03-25 1986-09-30 Nippon Shinyaku Co., Ltd. Liposome and method of manufacture therefor
JPS60104066A (ja) * 1983-11-10 1985-06-08 Ono Pharmaceut Co Ltd グリセリン誘導体
US5869534A (en) 1992-05-21 1999-02-09 The Picower Institute For Medical Research Glycosylation of lipids and lipid-containing particles, and diagnostic and therapeutic methods and materials derived therefrom
US5011771A (en) 1984-04-12 1991-04-30 The General Hospital Corporation Multiepitopic immunometric assay
US4827011A (en) * 1984-12-10 1989-05-02 American Cyanamid Company Antihypertensive phosphate derivatives
DE3663522D1 (en) 1985-11-29 1989-06-29 Takeda Chemical Industries Ltd Phospholipid derivatives, their production and use
US5561052A (en) 1992-06-18 1996-10-01 Koike; Katsumasa Process for detecting oxidized lipids and process for forming oxidized lipids
US5281521A (en) 1992-07-20 1994-01-25 The Trustees Of The University Of Pennsylvania Modified avidin-biotin technique
US5807884A (en) 1992-10-30 1998-09-15 Emory University Treatment for atherosclerosis and other cardiovascular and inflammatory diseases
FR2714382B1 (fr) 1993-12-27 1996-02-02 Roussel Uclaf Phospholipides vecteur de molécule active, leur préparation et leur utilisation dans des compositions cosmétiques ou dermatologiques.
ATE277176T1 (de) 1994-01-31 2004-10-15 Cancer Res Inst Royal Faltende proteine
JP3364313B2 (ja) * 1994-03-22 2003-01-08 株式会社トクヤマ ポルフィリン/インジウム錯体及び陰イオン感応膜
US5660855A (en) * 1995-02-10 1997-08-26 California Institute Of Technology Lipid constructs for targeting to vascular smooth muscle tissue
US6156500A (en) 1995-02-10 2000-12-05 Millennium Pharmaceuticals, Inc. Methods for the treatment and diagnosis of cardiovascular disease
US6261597B1 (en) 1995-08-31 2001-07-17 Seymour J. Kurtz Method for treating periodontal disease
US6114395A (en) 1996-11-15 2000-09-05 Pfizer Inc. Method of treating atherosclerosis
US6034102A (en) 1996-11-15 2000-03-07 Pfizer Inc Atherosclerosis treatment
US6096291A (en) 1996-12-27 2000-08-01 Biovector Therapeutics, S.A. Mucosal administration of substances to mammals
US5935577A (en) 1998-01-23 1999-08-10 Autoimmune Inc. Treatment of autoimmune disease using tolerization in combination with methotrexate
US5945308A (en) 1998-04-03 1999-08-31 Incyte Pharmaceuticals, Inc. Human oxidized LDL receptor
US6309888B1 (en) 1998-09-04 2001-10-30 Leuven Research & Development Vzw Detection and determination of the stages of coronary artery disease
US6701176B1 (en) 1998-11-04 2004-03-02 Johns Hopkins University School Of Medicine Magnetic-resonance-guided imaging, electrophysiology, and ablation
KR100865142B1 (ko) * 2000-11-24 2008-10-24 바스큘라 바이오제닉스 리미티드 죽상 경화증의 예방 및 치료를 위해 정의된 산화 인지질을사용하는 방법 및 이를 함유하는 조성물

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0121088A1 (fr) * 1983-03-05 1984-10-10 A. Nattermann & Cie. GmbH O-Acyl-alcanediol-phospholipides, procédé pour les préparer et compositions pharmaceutiques les contenant
EP0142333A2 (fr) * 1983-11-08 1985-05-22 Ono Pharmaceutical Co., Ltd. Dérivés du glycérol
WO1987005904A1 (fr) * 1986-03-24 1987-10-08 The University Of Sydney Analogues antigeniques du facteur d'activation des thrombocytes (paf)
JPS6354386A (ja) * 1986-08-26 1988-03-08 Takeda Chem Ind Ltd リン脂質およびその用途
JPH01258691A (ja) * 1988-04-06 1989-10-16 Nippon Oil & Fats Co Ltd リン脂質誘導体及びその製造方法
ES2019552A6 (es) * 1990-04-11 1991-06-16 Menarini Lab Procedimiento para la preparacion de glicerofosfolipidos.
WO1995023592A1 (fr) * 1994-03-04 1995-09-08 The University Of British Columbia Compositions a base de liposomes et procedes de traitement de l'atherosclerose

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
DEIGNER H P ET AL: "Effect of platelet activating factor on the kinetics of LDL oxidation in vitro" FEBS LETTERS, ELSEVIER, AMSTERDAM, NL, vol. 317, no. 3, 15 February 1993 (1993-02-15), pages 202-206, XP025615837 ISSN: 0014-5793 [retrieved on 1993-02-15] *
ITABE H ET AL: "Oxidized phosphatidylcholines that modify proteins. Analysis by monoclonal antibody against oxidized low density lipoprotein." THE JOURNAL OF BIOLOGICAL CHEMISTRY 27 DEC 1996, vol. 271, no. 52, 27 December 1996 (1996-12-27), pages 33208-33217, XP002532604 ISSN: 0021-9258 *
ITABE H ET AL: "Preparation of radioactive aldehyde-containing phosphatidylcholine." ANALYTICAL BIOCHEMISTRY 1 OCT 2000, vol. 285, no. 1, 1 October 2000 (2000-10-01), pages 151-155, XP002532603 ISSN: 0003-2697 *
KAMIDO H ET AL: "LIPID ESTER-BOUND ALDEHYDES AMONG COPPER-CATALYZED PEROXIDATION PRODUCTS OF HUMA PLASMA LIPOPROTEINS" JOURNAL OF LIPID RESEARCH, BETHESDA, MD, US, vol. 36, 1 January 1995 (1995-01-01), pages 1876-1886, XP002951008 ISSN: 0022-2275 *
KERN H ET AL: "STIMULATION MONOCYTTS AND PLATELETS BY SHORT-CHAIN PHOSPHATIDYLCHOLINES WITH AND WITHOUT TERMINAL CARBOXYL GROUP" BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH, ELSEVIER SCIENCE PUBLISHERS, AMSTERDAM, NL, vol. 1394, no. 1, 1 January 1998 (1998-01-01), pages 33-42, XP000938800 ISSN: 0167-4889 *
LEITINGER N ET AL: "STRUCTURALLY SIMILAR OXIDIZED PHOSPHOLIPIDS DIFFERENTIALLY REGULATE ENDOTHELIAL BINDING OF MONOCYTES AND NEUTROPHILS" PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF USA, NATIONAL ACADEMY OF SCIENCE, WASHINGTON, DC.; US, vol. 96, no. 21, 12 October 1999 (1999-10-12), pages 12010-12015, XP002951991 ISSN: 0027-8424 *
See also references of WO0241827A2 *
SUBBANAGOUNDER G ET AL: "DETERMINANTS OF BIOACTIVITY OF OXIDIZED PHOSPHOLIPIDS SPECIFIC OXIDIZED FATTY ACYL GROUPS AT THE SN-2 POSITION" ARTERIOSCLEROSIS, THROMBOSIS, AND VASCULAR BIOLOGY, LIPPINCOTT WILLIAMS & WILKINS, no. 20, 1 January 2000 (2000-01-01), pages 2248-2254, XP002951986 ISSN: 1079-5642 *

Also Published As

Publication number Publication date
EP2425841A1 (fr) 2012-03-07
WO2002041827A3 (fr) 2002-10-10
KR20030077543A (ko) 2003-10-01
JP2004537498A (ja) 2004-12-16
WO2002041827A9 (fr) 2003-05-30
WO2002041827A2 (fr) 2002-05-30
JP2008222726A (ja) 2008-09-25
JP2012149075A (ja) 2012-08-09
IL156015A0 (en) 2003-12-23
AU1846102A (en) 2002-06-03
CA2429817C (fr) 2013-02-12
AU2002218461B2 (en) 2006-09-07
MXPA03004517A (es) 2004-03-26
EP1341543A4 (fr) 2009-09-02
JP5001906B2 (ja) 2012-08-15
CA2429817A1 (fr) 2002-05-30
KR100865142B1 (ko) 2008-10-24
CN100577174C (zh) 2010-01-06
CN1529605A (zh) 2004-09-15
JP4162486B2 (ja) 2008-10-08

Similar Documents

Publication Publication Date Title
US6838452B2 (en) Methods employing and compositions containing defined oxidized phospholipids for prevention and treatment of atherosclerosis
JP5001906B2 (ja) アテローム性動脈硬化を防止および処置するための規定された酸化型リン脂質を含有する組成物
US7279459B2 (en) Methods employing and compositions containing plaque associated molecules for prevention and treatment of atherosclerosis
AU2002225301A1 (en) Methods employing and compositions containing plaque associated molecules for prevention and treatment of atherosclerosis
IL176976A (en) Use of oxygenated phospholipids defined for the preparation of a medicinal product for the treatment of atherosclerosis and other vascular diseases
IL156770A (en) Compositions containing plaque associated molecules and uses thereof for preparing medicaments for prevention and treatment of atherosclerosis

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: 20030530

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LI LU MC NL PT SE TR

AX Request for extension of the european patent

Extension state: AL LT LV MK RO SI

RIC1 Information provided on ipc code assigned before grant

Ipc: A61P 9/10 20060101ALI20090618BHEP

Ipc: C07C 59/235 20060101ALI20090618BHEP

Ipc: C07F 9/02 20060101ALI20090618BHEP

Ipc: A61K 31/08 20060101ALI20090618BHEP

Ipc: A61K 31/075 20060101ALI20090618BHEP

Ipc: A61K 31/11 20060101ALI20090618BHEP

Ipc: A61K 31/12 20060101ALI20090618BHEP

Ipc: A61K 31/20 20060101ALI20090618BHEP

Ipc: A61K 31/19 20060101ALI20090618BHEP

Ipc: A61K 31/24 20060101ALI20090618BHEP

Ipc: A61K 31/215 20060101ALI20090618BHEP

Ipc: A61K 31/685 20060101AFI20021028BHEP

A4 Supplementary search report drawn up and despatched

Effective date: 20090803

17Q First examination report despatched

Effective date: 20091109

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: 20130522

REG Reference to a national code

Ref country code: HK

Ref legal event code: WD

Ref document number: 1056688

Country of ref document: HK