EP3897677A1 - Composition à utiliser dans une méthode visant à abaisser le taux plasmatique de cholestérol lbd - Google Patents

Composition à utiliser dans une méthode visant à abaisser le taux plasmatique de cholestérol lbd

Info

Publication number
EP3897677A1
EP3897677A1 EP19829701.2A EP19829701A EP3897677A1 EP 3897677 A1 EP3897677 A1 EP 3897677A1 EP 19829701 A EP19829701 A EP 19829701A EP 3897677 A1 EP3897677 A1 EP 3897677A1
Authority
EP
European Patent Office
Prior art keywords
cholesterol
ubiquinone
bacteriopheophytin
spirilloxanthin
geranyl
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Withdrawn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
EP19829701.2A
Other languages
German (de)
English (en)
Inventor
Ruddy Wattiez
Felice MASTROLEO
Robertus Christiaan Josephus Suters
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.)
Ezcol BV
Original Assignee
Ezcol BV
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 Ezcol BV filed Critical Ezcol BV
Publication of EP3897677A1 publication Critical patent/EP3897677A1/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • 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
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/045Hydroxy compounds, e.g. alcohols; Salts thereof, e.g. alcoholates
    • 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
    • A61K31/085Ethers or acetals having an ether linkage to aromatic ring nuclear carbon
    • A61K31/09Ethers or acetals having an ether linkage to aromatic ring nuclear carbon having two or more such linkages
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/12Ketones
    • A61K31/122Ketones having the oxygen directly attached to a ring, e.g. quinones, vitamin K1, anthralin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/409Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil having four such rings, e.g. porphine derivatives, bilirubin, biliverdine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • A61K31/52Purines, e.g. adenine
    • A61K31/522Purines, e.g. adenine having oxo groups directly attached to the heterocyclic ring, e.g. hypoxanthine, guanine, acyclovir
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/74Bacteria
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics

Definitions

  • the present invention relates to a composition
  • bacteriopheophytins, carotenoids and quinones in particular compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10 and a pharmaceutical composition comprising a bacteriopheophytin, carotenoid and quinone in particular a composition comprising said compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin
  • the present invention relates to compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4- rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10 and a pharmaceutical composition comprising compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinol
  • the present invention also relates to bacteriopheophytins, carotenoids and quinones such as compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4- spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10 and to a pharmaceutical composition comprising a bacteriopheophytin, carotenoid and quinone in particular said compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rho
  • the invention also relates to a food supplement with cholesterol-lowering properties, comprising compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10.
  • the invention relates to a foodstuff comprising a food supplement, wherein the food supplement has cholesterol-lowering properties and comprises compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4- rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10.
  • the proviso is that the (pharmaceutical) composition is not, and the compounds are not provided as, a petroleum ether extract of Rhodospirillum rubrum obtainable by extraction of Rhodospirillum rubrum cells with a mixture of petroleum ether with a boiling point of between 60°C and 80°C and methanol comprising sodium chloride, wherein the extract is obtained by extraction for between 10 minutes and 48 hours at between 8°C and 37°C, while mixing the cells with the mixture, and wherein the Rhodospirillum rubrum cells are extracted with the mixture in a volume ratio of between 10:1 and 1 :10 of petroleum ether with a boiling point of between 60°C and 80°C and methanol comprising sodium chloride.
  • the proviso is that the composition and the composition for use of the invention is not a preparation comprising the membrane fraction of Rhodospirillum spp. and/or the membrane fraction of Phaeospirillum spp., and not a preparation or pharmaceutical preparation of Rhodospirillum spp. for use in lowering plasma cholesterol, said preparation or pharmaceutical preparation comprising dead Rhodospirillum spp. , or freeze-dried Rhodospirillum spp.
  • CVDs Cardiovascular diseases
  • CVDs are responsible for over 17.3 million deaths per year and are the leading causes of death in the world, according to the World Health Organization.
  • CVDs include diseases of the heart, vascular diseases of the brain and diseases of blood vessels.
  • the different types of CVDs are: CVDs due to atherosclerosis, which are ischaemic heart disease or coronary artery disease (e.g. heart attack); cerebrovascular disease (e.g. stroke); diseases of the aorta and arteries, including hypertension and peripheral vascular disease, and other CVDs, i.e. congenital heart disease; rheumatic heart disease; cardiomyopathies; and cardiac arrhythmias.
  • CVD is caused by a number of synergistic factors, the most important being a too high blood cholesterol level.
  • Cholesterol is an essential building block for animal and human cells, since it is a component of cell membranes. Human cells can synthesize their own cholesterol, but cholesterol is also assimilated from food. Both processes play an important part in cholesterol metabolism.
  • cardiovascular disease Apart from its essential biological role as a building block for cellular membranes, cholesterol also has negative effects on human health, as a cause of cardiovascular disease (such as, for instance, myocardial infarction, stroke, and peripheral vascular disease), more specifically in relation to the occurrence of atherosclerotic lesions in the blood vessel wall.
  • An elevated plasma cholesterol level is the most important predictive risk factor for the occurrence of cardiovascular disease and atherosclerosis.
  • VLDL very-low-density lipoproteins
  • IDL intermediate-density lipoproteins
  • LDL low-density lipoproteins
  • HDL high-density lipoproteins
  • the pro-atherogenic cholesterol such as the level of LDL-cholesterol
  • the anti-atherogenic cholesterol the HDL-cholesterol
  • Drugs that are used to inhibit cholesterol synthesis are often inhibitors of the enzyme hydroxymethyl- glutaryl-coenzyme A reductase (HMGCoA reductase), the rate-limiting enzyme in the cholesterol synthesis pathway.
  • HMGCoA reductase hydroxymethyl- glutaryl-coenzyme A reductase
  • statins are molecules that inhibit enzyme action. Examples are simvastatin, pravastatin and atorvastatin.
  • Statins are generally chemically-synthetized derivatives of naturally- occurring fungal metabolites.
  • Extended release niacin has been reported to lower LDL-cholesterol with 17%. Fenofibrate has been reported to lower LDL-cholesterol levels with about 20%.
  • Ezetimibe is an intestinal cholesterol absorption inhibitor which reduces LDL-cholesterol with 18%.
  • Colesevelam is a bile acid sequestrant which reduces LDL-cholesterol with 18%.
  • Mipomersen is an inhibitor of apolipoprotein B-100 synthesis and was shown to reduce LDL-cholesterol levels with about 25% in patients with homozygous familial hypercholesterolemia.
  • Lomitapide is an inhibitor of microsomal triglyceride transfer protein for example for the treatment of patients with homozygous familial hypercholesterolemia.
  • the lomitapide reduced LDL-cholesterol levels with 50% in those patients.
  • Proprotein convertase subtilisin / kexin type 9 inhibitor (PCSK9 inhibitor) molecules and gene-silencing approaches are under development.
  • Inhibition of PCSK9 in a subject may enhance the LDL-cholesterol lowering activity of statins.
  • Combined treatment of subjects with an antibody against PCSK9 (REGN727 / SAR236553) and statin atorvastatin resulted in a reduction in LDL-cholesterol levels of about 39% to 61 %.
  • the small molecule ETC-1002 modulates adenosine triphosphate-citrate lyase as well as adenosine monophosphate-activated protein kinase.
  • LDL-cholesterol levels In patients suffering from hypercholesterolemia (LDL-cholesterol levels of 130-220 mg/dL), LDL- cholesterol levels were reduced with about 18% to about 27%, when treated with increasing doses of ETC-1002.
  • Cholesteryl ester transfer protein (CETP) inhibitors raise HDL-cholesterol levels and decrease LDL-cholesterol levels. Examples of such a CETP inhibitors are anacetrapib and evacetrapib. Anacetrapib and evacetrapib have been shown in clinical trials with human subjects to increase HDL- cholesterol levels with respectively about 138% and about 129%, and to lower LDL-cholesterol levels with respectively about 40% and about 36%.
  • WAY-252623 is an activator of the beta-isoform of the liver X receptors.
  • WAY-252623 reduced LDL-cholesterol with 70%-77%.
  • a bile acid-adsorbing resin can be used (for example cholestyramine). Because of the adsorption of bile acids to the resin, their secretion in the stool is increased, and their reabsorption from the gut into the blood is reduced, resulting in a relative loss of bile acids from the body. Consequently, the liver increases the conversion of cholesterol into bile acids, resulting in a net increase in the secretion of cholesterol (metabolites) from the body. Because bile acids (by solubilizing cholesterol) are essential for the uptake of cholesterol from the lumen into the intestinal tissue, a reduction in bile acid content in the intestinal lumen will also result in a decreased cholesterol uptake.
  • Patent EP1569667 discloses a preparation of Rhodospirillum cells for use as a medicament, for use in a food supplement, for use in a foodstuff, wherein said use is for use in a method for the lowering of plasma cholesterol in a subject.
  • An aspect of the invention relates to a composition
  • a composition comprising at least one carotenoid, pheophytin such as a bacteriopheophytin or quinone for use in lowering plasma cholesterol, under the proviso that the composition is not a petroleum ether extract of Rhodospirillum rubrum obtainable by extraction of Rhodospirillum rubrum cells with a mixture of petroleum ether with a boiling point of between 60°C and 80°C and methanol comprising sodium chloride, wherein the extract is obtained by extraction for between 10 minutes and 48 hours at between 8°C and 37°C, while mixing the cells with the mixture, and wherein the Rhodospirillum rubrum cells are extracted with the mixture in a volume ratio of between 10:1 and 1 :10 of petroleum ether with a boiling point of between 60°C and 80°C and methanol comprising sodium chloride.
  • An aspect of the invention relates to a composition
  • a composition comprising at least one carotenoid, pheophytin such as a bacteriopheophytin or quinone for use in the lowering of LDL-cholesterol in blood plasma of a subject, under the proviso that the composition is not a petroleum ether extract of Rhodospirillum rubrum obtainable by extraction of Rhodospirillum rubrum cells with a mixture of petroleum ether with a boiling point of between 60°C and 80°C and methanol comprising sodium chloride, wherein the extract is obtained by extraction for between 10 minutes and 48 hours at between 8°C and 37°C, while mixing the cells with the mixture, and wherein the Rhodospirillum rubrum cells are extracted with the mixture in a volume ratio of between 10:1 and 1 :10 of petroleum ether with a boiling point of between 60°C and 80°C and methanol comprising sodium chloride.
  • An aspect of the invention relates to a composition
  • a composition comprising at least one carotenoid, pheophytin, such as a bacteriopheophytin, or quinone for use in a method for the lowering of LDL-cholesterol in blood plasma of a subject, under the proviso that the composition is not a petroleum ether extract of Rhodospirillum rubrum obtainable by extraction of Rhodospirillum rubrum cells with a mixture of petroleum ether with a boiling point of between 60°C and 80°C and methanol comprising sodium chloride, wherein the extract is obtained by extraction for between 10 minutes and 48 hours at between 8°C and 37°C, while mixing the cells with the mixture, and wherein the Rhodospirillum rubrum cells are extracted with the mixture in a volume ratio of between 10:1 and 1 :10 of petroleum ether with a boiling point of between 60°C and 80°C and methanol comprising sodium chloride.
  • An aspect of the invention relates to a composition
  • a composition comprising at least one carotenoid, pheophytin such as a bacteriopheophytin or quinone for use in the treatment or prophylaxis of a cardiovascular disease (CVD) such as coronary heart disease, or of risk factors associated with development of a CVD or for example atherosclerosis, under the proviso that the composition is not a petroleum ether extract of Rhodospirillum rubrum obtainable by extraction of Rhodospirillum rubrum cells with a mixture of petroleum ether with a boiling point of between 60°C and 80°C and methanol comprising sodium chloride, wherein the extract is obtained by extraction for between 10 minutes and 48 hours at between 8°C and 37°C, while mixing the cells with the mixture, and wherein the Rhodospirillum rubrum cells are extracted with the mixture in a volume ratio of between 10:1 and 1 :10 of petroleum ether with a boiling point of between 60°C
  • An aspect of the invention relates to a composition
  • a composition comprising at least one carotenoid such as a xanthophyll for use in the treatment or prophylaxis of a cardiovascular disease (CVD) such as coronary heart disease, or of risk factors associated with development of a CVD or for example atherosclerosis, under the proviso that the composition is not a petroleum ether extract of Rhodospirillum rubrum obtainable by extraction of Rhodospirillum rubrum cells with a mixture of petroleum ether with a boiling point of between 60°C and 80°C and methanol comprising sodium chloride, wherein the extract is obtained by extraction for between 10 minutes and 48 hours at between 8°C and 37°C, while mixing the cells with the mixture, and wherein the Rhodospirillum rubrum cells are extracted with the mixture in a volume ratio of between 10:1 and 1 :10 of petroleum ether with a boiling point of between 60°C and 80°C and methanol comprising sodium chloride.
  • An aspect of the invention relates to a composition
  • a composition comprising at least one pheophytin such as a bacteriopheophytin for use in the treatment or prophylaxis of a cardiovascular disease (CVD) such as coronary heart disease, or of risk factors associated with development of a CVD or for example atherosclerosis, under the proviso that the composition is not a petroleum ether extract of Rhodospirillum rubrum obtainable by extraction of Rhodospirillum rubrum cells with a mixture of petroleum ether with a boiling point of between 60°C and 80°C and methanol comprising sodium chloride, wherein the extract is obtained by extraction for between 10 minutes and 48 hours at between 8°C and 37°C, while mixing the cells with the mixture, and wherein the Rhodospirillum rubrum cells are extracted with the mixture in a volume ratio of between 10:1 and 1 :10 of petroleum ether with a boiling point of between 60°C and 80°C and methanol compris
  • the pheophytin(s) is/are for example natural pheophytin(s) and are for example isolated from a natural source or are chemically synthesized.
  • An aspect of the invention relates to a composition comprising at least one quinone or ubiquinone or ubiquinol or ubiquinone derivative for use in the treatment or prophylaxis of a cardiovascular disease (CVD) such as coronary heart disease, or of risk factors associated with development of a CVD or for example atherosclerosis.
  • CVD cardiovascular disease
  • the quinone or ubiquinone or ubiquinol or ubiquinone derivative is/are for example natural quinone, ubiquinone or ubiquinol or a naturally occurring ubiquinone derivative such as rhodoquinone, and are for example isolated from a natural source or are chemically synthesized.
  • the ubiquinol is for example any one or more of naturally occurring and/or chemically synthesized ubiquinol-5, ubiquinol-6, ubiquinol-7, ubiquinol-8, ubiquinol-9, ubiquinol-10, ubiquinol-1 1 , ubiquinol-12, ubiquinol-13, ubiquinol-14, or is for example any one or more of naturally occurring and/or chemically synthesized ubiquinol-6, ubiquinol-7, ubiquinol-8, ubiquinol-9, ubiquinol-10, ubiquinol-1 1 , ubiquinol-12, ubiquinol-13, or is for example any one or more of naturally occurring and/or chemically synthesized ubiquinol-7, ubiquinol-8, ubiquinol-9, ubiquinol-10, ubiquinol-1 1 , ubiquinol-12, or is for example any one or more of naturally occurring and/or chemically synthesized ubiquinol-8, ubiquinol-9, ubiquinol-10, ubiquinol-1 1
  • the ubiquinone is for example any one or more of naturally occurring and/or chemically synthesized ubiquinone-5, ubiquinone-6, ubiquinone-7, ubiquinone-8, ubiquinone-9, ubiquinone-10, ubiquinone-1 1 , ubiquinone-12, ubiquinone-13, ubiquinone-14, or is for example any one or more of naturally occurring and/or chemically synthesized ubiquinone-6, ubiquinone-7, ubiquinone-8, ubiquinone-9, ubiquinone-10, ubiquinone-1 1 , ubiquinone-12, ubiquinone-13, or is for example any one or more of naturally occurring and/or chemically synthesized ubiquinone-7, ubiquinone-8, ubiquinone- 9, ubiquinone-10, ubiquinone-1 1 , ubiquinone-12, or is for example any one or more of naturally occurring and/or chemically synthe
  • the pheophytin is for example a chlorophyll derivative lacking the central Mg 2+ ion and is for example a bacteriopheophytin a, for example a bacteriopheophytin a comprising a phytyl group or comprising a geranyl-geranyl group.
  • the carotenoid(s) is/are for example natural carotenoid(s) and are for example isolated from a natural source or are chemically synthesized.
  • the carotenoid(s) is/are preferably tetraterpenoids such as a xanthophyll and a carotene, though xanthophylls are preferred.
  • the carotenoids are preferably polar carotenoids such as xanthophylls, and xanthophylls are preferred.
  • the carotenoids are preferably oxygenated carotenoids.
  • the carotenoids are for example xanthophylls based on the precursor phytoene.
  • the carotenoids are for example xanthophylls based on the precursor xanthophyll rhodopin.
  • the xanthophyll is for example an alcohol, an ether, or has both a hydroxyl group and an ether group.
  • the xanthophyll is for example rhodovibrin hydroxyspirilloxanthin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin or 3,4-spirilloxanthin.
  • An aspect of the invention relates to a composition
  • a composition comprising at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, at least eleven, at least twelve, preferably all thirteen, of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4- spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10 for use in the treatment or prophylaxis of a cardiovascular disease (CVD), under the proviso that the composition is not a petroleum ether extract of Rhodospirillum rubrum obtainable by extraction of Rho
  • the treatment or prevention of a CVD in a human subject or in the treatment or prophylaxis of risk factors associated with development of a CVD or for example with development of atherosclerosis are for example isolated from a natural source, such as cultured R.rubrum bacteria cells, or are chemically synthesized, under the proviso that the isolated geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a
  • An aspect of the invention relates to a composition
  • a composition comprising two or more, in particular three, four, five, six, seven, eight, nine, ten, eleven, twelve or thirteen, preferably thirteen compounds of geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10 for use in a method for the lowering of LDL- cholesterol in blood plasma of a human subject, wherein the composition is not a petroleum ether extract of Rhodospirillum rubrum obtainable by extraction of Rhodospirillum rubrum cells with a mixture of petroleum ether with
  • An embodiment of the invention relates to a composition for use according to the invention comprising all of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10.
  • An aspect of the invention relates to a composition
  • a composition comprising at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, at least eleven, at least twelve, preferably all thirteen, of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10 for use as an LDL-cholesterol level lowering medicament, under the proviso that the composition is not a petroleum ether extract of Rhodospirillum rubrum obtainable by extraction of Rhodospirillum
  • An embodiment is the composition comprising at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, at least eleven, at least twelve, preferably all thirteen, of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10 for use as an LDL-cholesterol level lowering medicament, wherein the HDL- cholesterol level in the plasma of a human subject to whom the composition is administered, remains essentially unaltered, increases when compared to the level prior to administration of the composition, or decreases to a lesser
  • a further aspect of the invention relates to a composition
  • a composition comprising at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, at least eleven, at least twelve, preferably all thirteen, of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10 for use in the treatment or prophylaxis of atherosclerosis, dyslipidemia, arteriosclerosis, hypercholesterolemia, familial hypercholesterolemia, hyperlipidemia, an LDL- cholesterol plasma level of at least
  • the plasma LDL-cholesterol level of the treated human subject reduces to a level relating to lower risk for CVD and/or any of the listed diseases and health problems, such as a level of less than 200 mg/dL LDL-cholesterol, or less than 160 mg/dL, or less than 140 mg/dL, or less than 120 mg/dL, or less than 100 mg/dL, or less than 80 mg/dL, such as between 45 mg/dL and 75 mg/dL LDL-cholesterol.
  • the level of HDL-cholesterol in plasma of a human subject to whom the composition is administered essentially remains unaltered, increases, or decreases to a limited extent such as with about 5-20%.
  • An aspect of the invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a pharmaceutically effective amount of at least one, at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, at least eleven, at least twelve, preferably all thirteen of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10 and optionally a pharmaceutically acceptable excipient for use in lowering plasma cholesterol.
  • the pharmaceutical composition comprises at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, at least eleven, at least twelve, preferably all thirteen of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4- spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10.
  • the pharmaceutical composition comprises all of geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10, under the proviso that the pharmaceutical composition is not, or does not comprise a petroleum ether extract of Rhodospirillum rubrum obtainable by extraction of Rhodospirillum rubrum cells with a mixture of petroleum ether with a boiling point of between 60°C and 80°C and methanol comprising sodium chloride, wherein the extract is obtained by extraction for between 10 minutes and 48 hours at between 8°C and 37°C, while mixing the cells with the mixture, and
  • An aspect of the invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a pharmaceutically effective amount of at least one, preferably at least two, more preferably all, of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10and optionally a pharmaceutically acceptable excipient for use in the lowering of LDL- cholesterol in blood plasma of a subject, under the proviso that the pharmaceutical composition is not, or does not comprise a petroleum ether extract of Rhodospirillum rubrum obtainable by extraction of Rhodospirillum rubrum cells with
  • An aspect of the invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a pharmaceutically effective amount of at least one, preferably at least two, more preferably all, of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4- spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10 and optionally a pharmaceutically acceptable excipient for use in a method for the lowering of LDL-cholesterol in blood plasma of a subject, under the proviso that the pharmaceutical composition is not, or does not comprise a petroleum ether extract of Rhodospirillum rubrum obtainable by extraction of Rhodospir
  • a further aspect of the invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a pharmaceutically effective amount of at least one, preferably at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, at least eleven, at least twelve, more preferably all thirteen, of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10 and optionally a pharmaceutically acceptable excipient, for use in a method for the treatment or prophylaxis of any one or more of cardiovascular disease, athe
  • a petroleum ether extract of Rhodospirillum rubrum comprises compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10 and reduces the LDL-cholesterol plasma level while maintaining the plasma HDL- cholesterol level essentially unaltered when the compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodo
  • the at least one preferably at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, at least eleven, at least twelve, more preferably all thirteen, of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10is/are either the sole active pharmaceutical ingredient(s) in the pharmaceutical composition, orthe pharmaceutical composition is administered to a subject to whom at least one further active pharmaceutical ingredient
  • a statin and a PCSK9 inhibitor are administered to a subject in need thereof, and the pharmaceutical composition according to the invention is administered to said subject.
  • the at least one further active pharmaceutical ingredient is administered to a subject in need thereof simultaneously, separately or sequentially with the pharmaceutical composition for use in the method for the treatment or prophylaxis of e.g. CVD.
  • An aspect of the invention relates to a method for treating or preventing a cardiovascular disease, atherosclerosis, dyslipidemia, arteriosclerosis, hypercholesterolemia, familial hypercholesterolemia, hyperlipidemia, an LDL-cholesterol plasma level of at least 70 mg/dL, an LDL- cholesterol plasma level of at least 100 mg/dL, an LDL-cholesterol plasma level of at least 140 mg/dL, an LDL-cholesterol plasma level of at least 200 mg/dL, a total plasma cholesterol level of at least 200 mg/dL, an Lp(a) level of at least 14 mg/dL, inflammation, inflammatory disease, ischemia, infection, the method comprising administering in a subject in need thereof an effective amount of at least one, preferably at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, at least eleven, at least twelve, more preferably all thirteen, of compounds geranyl-geranyl bacterioph
  • a further aspect of the invention relates to a method of treating a patient suffering from an LDL- cholesterol concentration in the plasma of said patient of at least 70 mg/dl_ such as above 100 mg/dL or above 150 mg/dL by administering an effective amount of at least one, preferably at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, at least eleven, at least twelve, more preferably all thirteen, of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4- rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10 to the patient
  • An aspect of the invention relates to a method of treating a patient suffering from or having an increased risk for any one or more of cardiovascular disease, atherosclerosis, dyslipidemia, arteriosclerosis, hypercholesterolemia, familial hypercholesterolemia, hyperlipidemia, an LDL- cholesterol plasma level of at least 70 mg/dL, an LDL-cholesterol plasma level of at least 100 mg/dL, an LDL-cholesterol plasma level of at least 140 mg/dL, an LDL-cholesterol plasma level of at least 200 mg/dL, a total plasma cholesterol level of at least 200 mg/dL, an Lp(a) level of at least 14 mg/dL, inflammation, inflammatory disease, ischemia, infection, by administering an effective amount of at least one, preferably at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, at least eleven, at least twelve, more preferably all thirteen, of compounds geranyl-geranyl bacteri
  • the effective amount of at least one, preferably at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, at least eleven, at least twelve, more preferably all thirteen, of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10 is administered to a patient in need thereof, said patient suffering from or having an increased risk for any one or more of said aforementioned diseases or risk factors.
  • the CVDs include diseases of the heart, vascular diseases of the brain and diseases of blood vessels.
  • the CVDs are: CVDs due to atherosclerosis, which are ischaemic heart disease or coronary artery disease (e.g. heart attack), coronary heart disease; cerebrovascular disease (e.g. stroke); diseases of the aorta and arteries, including hypertension and peripheral vascular disease, and other CVDs, i.e. congenital heart disease; rheumatic heart disease; cardiomyopathies; and cardiac arrhythmias.
  • the invention also relates to a food supplement with cholesterol-lowering properties, comprising at least one, preferably at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, at least eleven, at least twelve, more preferably all thirteen, of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4- spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10, under the proviso that these one or more compounds is/are not provided as a petroleum ether extract of Rhodospirillum rubrum obtainable by extraction of Rhodospirill
  • the invention relates to a foodstuff comprising a food supplement, wherein the food supplement has cholesterol-lowering properties and comprises at least one, preferably at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, at least eleven, at least twelve, more preferably all thirteen, of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4- spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10, under the proviso that these one or more compounds is/are not provided as a petroleum ether extract of Rhodospirillum rubrum
  • the proviso is that the composition and the composition for use of the invention is not a preparation comprising the membrane fraction of Rhodospirillum spp. and/or the membrane fraction of Phaeospirillum spp., and not a preparation or pharmaceutical preparation of Rhodospirillum spp. for use in lowering plasma cholesterol, said preparation or pharmaceutical preparation comprising dead Rhodospirillum spp. , or freeze-dried Rhodospirillum spp.
  • the compounds are not provided as a preparation comprising the membrane fraction of Rhodospirillum spp.
  • Rhodospirillum spp. and/or the membrane fraction o Phaeospirillum spp., and not as a preparation or pharmaceutical preparation of Rhodospirillum spp. for use in lowering plasma cholesterol, said preparation or pharmaceutical preparation comprising dead Rhodospirillum spp. , or freeze-dried Rhodospirillum spp.
  • the current invention provides a composition
  • a composition comprising compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4- rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10, under the proviso that the composition is not a petroleum ether extract of Rhodospirillum rubrum obtainable by extraction of Rhodospirillum rubrum cells with a mixture of petroleum ether with a boiling point of between 60°C and 80°C and methanol comprising sodium chloride, wherein the extract is obtained by extraction for between 10 minutes and 48 hours at between 8°C and 37°C, while mixing the cells with the mixture,
  • Rhodospirillum spp. is not a preparation or pharmaceutical preparation of Rhodospirillum spp. for use in lowering plasma cholesterol, said preparation or pharmaceutical preparation comprising dead Rhodospirillum spp. , or freeze-dried Rhodospirillum spp. , according to the invention.
  • FIG 1 This figure displays the scope of Figure 2 of WO 2004/052380 A1 , displaying the lipoprotein pattern in plasma of Wistar rats that were fed a normal chow diet (‘Controls’), and from Wistar rats that were fed a chow diet containing 10% (w/w) R. rubrum (‘10% R.rub’), as explained in detail in Example 2 of WO 2004/052380 A1 .
  • Controls normal chow diet
  • R. rubrum ‘10% R.rubrum’
  • FIG 2 This figure displays the scope of Figure 5 of WO 2004/052380 A1 , showing the lipoprotein pattern in plasma of C57BI/6 mice that were fed a hypercholesterolaemic“Western-type” diet, and a hypercholesterolaemic“Western-type” diet containing 10% (w/w) R.rubrum, as outlined in detail in Example 4 of WO 2004/052380 A1 . Under influence of consumed R.rubrum cells the plasma LDL-cholesterol levels in the mice decreased, whereas the plasma HDL-cholesterol levels remained essentially unaltered.
  • Figure 3 Shown is the plasma cholesterol lowering effect with regard to LDL-cholesterol in an in vivo mouse model, while HDL-cholesterol levels and total cholesterol levels essentially remain unaltered upon treatment of the mice.
  • Mice were fed control feed (‘control’) or feed enriched with an extract of bacterium Rhodospirillum rubrum comprising geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10(‘Extract 1.1’).
  • This extract is an example of a composition
  • a composition comprising geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4- rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10.
  • a cholesterol-lowering property is herein defined as the capability of a compound or a composition, such as a carotenoid, bacteriopheophytin or quinone, such as compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4- rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10, a composition comprising geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxant
  • petroleum ether with a boiling point of between 60°C and 80°C has its regular scientific meaning and here refers to a petroleum fraction consisting of aliphatic hydrocarbons and boiling in the range of between 60°C and 80°C.
  • LDL-cholesterol level and the term“LDL-cholesterol concentration” have their regular scientific meaning throughout the description and in the claims, and here refer to the amount of cholesterol that is associated with and transported by LDL in the body of the subject, e.g. a human subject.
  • the LDL-cholesterol level is measured directly, as outlined in the Examples section. Where indicated, the LDL-cholesterol concentration is calculated from the measured total cholesterol concentration, the measured HDL-cholesterol concentration and the measured tri-acyl glycerol concentration in a sample such as a plasma sample of a human subject, and based on these measured data, the LDL-cholesterol level is calculated.
  • a compound or a composition comprising said compound, which is one or more of i) safe at a dose suitable for administering to a subject in need thereof, wherein‘safe’ refers to an acceptable extent, if occurring at all, of adverse side events in a subject to whom the compound or composition is administered, such as a human subject with a plasma LDL-cholesterol level of 70 mg/dL or higher, ii) active when lowering plasma LDL-cholesterol level in a subject is considered, and iii) capable of maintaining plasma HDL-cholesterol essentially unaltered or increasing HDL-cholesterol concentration in plasma in an absolute manner or relative manner when compared to plasma LDL-cholesterol level, when the compound or the composition comprising the compound is administered to a (human) subject.
  • safe refers to an acceptable extent, if occurring at all, of adverse side events in a subject to whom the compound or composition is administered, such as a human subject with a plasma LDL-cholesterol level of 70 mg/dL or higher,
  • At least one of the above objectives is achieved by at least some embodiments of the invention, providing at least one, preferably at least two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, more preferably all, of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10 for use in a method for treating or preventing a disease or a health problem such as CVD and atherosclerosis, related to a plasma LDL-cholesterol level of 70 mg/dL or higher in a subject, for example a human subject, such as for example 100 mg
  • At least one of the above objectives is also achieved by providing a composition comprising two or more compounds, such as three, four, five, six, seven, eight, nine, ten, eleven, twelve, preferably all thirteen of geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4- spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10 for use in a method for the lowering of LDL-cholesterol in blood plasma of a human subject, wherein the composition is not a petroleum ether extract of Rhodospirillum rubrum obtainable by extraction of Rhodospirillum rubrum cells with a mixture of petroleum ether with
  • said composition for use according to the invention comprises all of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10.
  • a pharmaceutical composition comprising a pharmaceutically effective amount of at least one, preferably at least two, more preferably all of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10 and optionally a pharmaceutically acceptable excipient, for use in a method for the treatment or prophylaxis of any one or more of cardiovascular disease, atherosclerosis, dyslipidemia, arteriosclerosis, hypercholesterolemia, familial hypercholesterolemia, hyperlipidemia, an LDL-cholesterol plasma level of at
  • An aspect of the invention relates to a composition
  • a composition comprising one, two or more, preferably all of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4- spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10 for use as a medicament, under the proviso that the composition and the one or more compounds is/are not provided as a petroleum ether extract of Rhodospirillum rubrum obtainable by extraction of Rhodospirillum rubrum cells with a mixture of petroleum ether with a boiling point of between 60°C and 80°C and methanol comprising sodium chloride, wherein the
  • An aspect of the invention relates to the use of a composition
  • a composition comprising one, two or more, preferably all of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10 for the manufacture of a medicament for the treatment of high LDL-cholesterol in blood plasma of a subject, such as a human subject, under the proviso that the composition and the one or more compounds is/are not provided as a petroleum ether extract of Rhodospirillum rubrum obtainable by extraction of Rhodospirillum rubrum cells with a mixture of petroleum
  • the term“high LDL-cholesterol in blood plasma” should be read as a plasma LDL-cholesterol concentration of at least 70 mg/dL, such as more than 160 mg/dL or more than 120 mg/dL.
  • An aspect of the invention relates to the use of at least one carotenoid, pheophytin such as a bacteriopheophytin or quinone for the manufacture of a medicament for the lowering of LDL- cholesterol in blood plasma of a subject, such as a human subject, such as a human subject having a plasma LDL-cholesterol level of higher than 80 mg/dL, under the proviso that the one or more carotenoids is/are not provided as a petroleum ether extract of Rhodospirillum rubrum obtainable by extraction of Rhodospirillum rubrum cells with a mixture of petroleum ether with a boiling point of between 60°C and 80°C and methanol comprising sodium chloride, wherein the extract is obtained by extraction for between 10 minutes
  • An aspect of the invention relates to a composition
  • a composition comprising one, two or more, preferably all of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10 for use in lowering plasma cholesterol, under the proviso that the composition and the one or more compounds is/are not provided as a petroleum ether extract of Rhodospirillum rubrum obtainable by extraction of Rhodospirillum rubrum cells with a mixture of petroleum ether with a boiling point of between 60°C and 80°C and methanol comprising sodium chloride, wherein the
  • An aspect of the invention relates to a composition
  • a composition comprising one, two or more, preferably all of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10, for use in the lowering of LDL-cholesterol in blood plasma of a subject, under the proviso that the composition and the one or more compounds is/are not provided as a petroleum ether extract of Rhodospirillum rubrum obtainable by extraction of Rhodospirillum rubrum cells with a mixture of petroleum ether with a boiling point of between 60°C and 80°
  • An aspect of the invention relates to a composition
  • a composition comprising one, two or more, preferably all of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4- rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10, for use in a method for the lowering of LDL-cholesterol in blood plasma of a subject, under the proviso that the composition and the one or more compounds is/are not provided as a petroleum ether extract of Rhodospirillum rubrum obtainable by extraction of Rhodospirillum rubrum cells with a mixture of petroleum ether with a boiling point of between 60
  • An aspect of the invention relates to a composition
  • a composition comprising one, two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, or more, preferably all thirteen of compouns geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4- rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10, for use in the treatment or prophylaxis of a cardiovascular disease such as coronary heart disease, under the proviso that the composition and the one or more compounds is/are not provided as a petroleum ether extract of Rhodospirillum rubrum obtainable by extraction of Rhod
  • An aspect of the invention relates to a composition
  • a composition comprising compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10 for use in the treatment or prophylaxis of a cardiovascular disease, under the proviso that the composition is not provided as a petroleum ether extract of Rhodospirillum rubrum obtainable by extraction of Rhodospirillum rubrum cells with a mixture of petroleum ether with a boiling point of between 60°C and 80°C and methanol comprising sodium chloride, wherein the extract is obtained by extraction for between 10 minutes
  • the CVDs include diseases of the heart, vascular diseases of the brain and diseases of blood vessels.
  • the CVDs include: CVDs due to atherosclerosis, which are ischaemic heart disease or coronary artery disease (e.g. heart attack); cerebrovascular disease (e.g. stroke); diseases of the aorta and arteries, including hypertension and peripheral vascular disease, and other CVDs, i.e. congenital heart disease; rheumatic heart disease; cardiomyopathies; and cardiac arrhythmias.
  • a further aspect of the invention relates to a composition
  • a composition comprising one, two or more, preferably all of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10, for use in the treatment or prophylaxis of atherosclerosis, dyslipidemia, arteriosclerosis, hypercholesterolemia, familial hypercholesterolemia, hyperlipidemia, an LDL-cholesterol plasma level of at least 70 mg/dL, an LDL-cholesterol plasma level of at least 100 mg/dL, an LDL-cholesterol plasma level of at least 140 mg
  • a further aspect of the invention relates to a composition
  • a composition comprising compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10 for use in the treatment or prophylaxis of atherosclerosis, dyslipidemia, arteriosclerosis, hypercholesterolemia, familial hypercholesterolemia, hyperlipidemia, an LDL plasma level of at least 70 mg/dL, an LDL plasma level of at least 100 mg/dL, an LDL plasma level of at least 140 mg/dL, an LDL plasma level of at least 200 mg/dL, a total cholesterol level
  • An aspect of the invention relates to a composition
  • a composition comprising two, three, four, five, six-twelve or more, preferably all of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10 for use in a method for the lowering of LDL-cholesterol in blood plasma of a human subject, wherein the composition is not a petroleum ether extract of Rhodospirillum rubrum obtainable by extraction of Rhodospirillum rubrum cells with a mixture of petroleum ether with a boiling point of between 60°C and 80°
  • An embodiment is the composition for use according to the invention comprising all of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4- spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10.
  • An aspect of the current invention relates to a composition
  • a composition comprising at least one, preferably at least two, more preferably all of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10, for use in a method for the treatment or prophylaxis of a cardiovascular risk factor and/or a risk factor for developing atherosclerosis, wherein the composition is not a petroleum ether extract of Rhodospirillum rubrum obtainable by extraction of Rhodospirillum rubrum cells with a mixture of petroleum ether with a boiling point of between
  • a risk factor for developing cardiovascular disease, or a cardiovascular risk factor, and/or a risk factor for developing atherosclerosis is for example a total blood cholesterol level of 200 mg/dL - 239 mg/dL or 240 mg/dL and higher.
  • a further risk factor is for example a plasma LDL-cholesterol level of 100 mg/dL or higher, such as 100 mg/dL - 129 mg/dL, 130 mg/dL - 159 mg/dL, 160 mg/dL - 189 mg/dL, or 190 mg/dL and higher.
  • a further risk factor is for example a plasma HDL-cholesterol level in a human subject of lower than 60 mg/dL, or less than 40 mg/dL.
  • lowering the LDL-cholesterol level in plasma / serum of a subject can be the consequence of inhibition of the absorption of cholesterol and/or cholesterol esters from the gastrointestinal tract, and/or the consequence of inhibition of the synthesis of cholesterol in e.g. the liver, and/or the consequence of an upregulated density and number of LDL-receptors on the plasma membranes of organ cells such as the liver cells, facilitating clearance of circulating LDL-cholesterol.
  • a portion of cholesterol in the body and in the circulation originates from dietary intake, and in addition a portion of the cholesterol present in a subject originates from de novo synthesis in amongst others and predominantly the liver.
  • Cholesterol biosynthesized in the liver is in part transported and stored in the gall bladder, in the bile.
  • the bile with the cholesterol is excreted to the gastrointestinal tract when required, i.e. when the subject consumes fat-rich food products, etc. It has been established that a large portion of bile cholesterol is excreted with the feces, with the remainder of the bile cholesterol being taken up from the intestine together with a portion of dietary cholesterol.
  • triacyl glycerides and cholesterol from the food intake, together with the cholesterol transferred from the gall bladder to the intestine with the bile are solubilized first as emulsion particles and then in bile salt micelles.
  • intestinal mucosal cells absorp the mixed bile salt micelles comprising the cholesterol.
  • the cholesterol is transferred into the lymphatic circulation as part of chylomicrons and also as part of very low density lipoproteins (VLDLs), which are transferred to the blood circulation.
  • Relocation and transfer of cholesterol comprising particles is based on passive simple diffusion and/or based on scavenger receptor mediated transfer, for example with involvement of scavenger receptor B-l and/or CD36.
  • the cholesterol-loaded chylomicrons transfer (in part) into chylomicron remnants carrying cholesterol, which are taken up from the circulation by the liver.
  • the VLDL-cholesterol particles are degraded into intermediate density lipoproteins (IDLs) comprising the cholesterol, and further into low- density lipoprotein-cholesterol upon discarding triacyl glycerol molecules from the IDL particles.
  • IDLs intermediate density lipoproteins
  • the LDL-cholesterol particles are bound by LDL-receptor molecules on the surface of organ cells such as the liver. Upon binding, the LDL-cholesterol is taken up by these LDL-receptor carrying cells.
  • Cholesterol taken up by liver cells is in part relocated to the gall bladder and becomes part of the stored bile, to be secreted into the intestine after food intake. Cholesterol taken up by liver cells is also in part transferred to the endoplasmic reticulum during assembly of VLDL particles comprising cholesterol.
  • the cholesterol in the VLDL-cholesterol particles originate in part from de novo cholesterol synthesis by the liver cells and in part from dietary cholesterol if present in the diet of the subject.
  • the liver cells excrete the VLDL-cholesterol into the blood circulation. In the blood circulation, the VLDL- cholesterol are transformed into IDL-cholesterol, which (partly) further transforms into LDL-cholesterol.
  • Carotenoids and (bacterio)chlorophyll analogues such as a bacteriopheophytin are not biosynthesized by humans.
  • Carotenoids and bacteriopheophytins present in the body thus originate from e.g. dietary intake, food supplements, etc. It has been established that uptake of polar carotenoids such as xanthophylls, and bacteriopheophtyins such as bacteriopheophytin a, from the ingested food into the blood circulation and into e.g. liver cells, essentially is established via the same transport route and mechanism applied by the body for cholesterol intake from food.
  • the polar carotenoids and the bacteriopheophytins are dissolved in emulsion particles and then in mixed bile salt micelles, facilitating transferto the lymphatic circulation as part of chylomicrons, and further into the blood stream.
  • Chylomicrons, once transferred into chylomicron remnants, are taken up by the liver.
  • Quinones are both biosynthesized by humans and by other organisms that are at least in part subject to human consumption and/or a fraction is part of the human diet, for example when prepared as part of a food product or food supplement. Quinones present in the body can thus originate from e.g. dietary intake, food supplements, etc.
  • the inventors now propose that the LDL-lowering effect of the carotenoids, bacteriopheophytins and quinones is based on the following principle of inhibition of cholesterol uptake into the circulation and inhibition of VLDL-cholesterol release from (liver) cells into the blood circulation.
  • dietary polar carotenoids, bacteriopheophytins as well as quinones compete with cholesterol during the process of formation of the lipid emulsion, such that uptake of cholesterol by the lipid emulsion is inhibited at the expense of increased uptake of the polar carotenoid, bacteriopheophytin or quinone by the lipid emulsion.
  • the liver takes up the chylomicron remnants comprising the carotenoids, bacteriopheophytins and quinones originating from the chylomicrons, and therewith the carotenoids, bacteriopheophytins and quinones become part of the pool of fat soluble compounds present and stored in liver cells, together with de novo synthesized cholesterol by the liver cells and cholesterol taken up by liver cells. In liver cells assembly of VLDL-cholesterol particles occurs.
  • the carotenoid, bacteriopheophtyin or quinone again can compete with the cholesterol for incorporation in the VLDL particle. Less cholesterol is secreted into the blood circulation with the VLDL, as a consequence. Further, also less IDL-cholesterol can be formed from VLDL, and then also less LDL- cholesterol. Indeed, polar carotenoids and quinones are determined in the blood plasma of human subjects, as part of LDL-particles.
  • bacteriopheophtyins are also in the blood plasma of human subjects, as part of LDL-particles. Furthermore, a lower chylomicron-cholesterol level in the blood due to inhibition of cholesterol uptake from the intestine by the presence of polar carotenoids, bacteriopheophtyins and quinones in the intestine, induces upregulation of LDL-receptor on hepatocytes, which as a result increases uptake of circulating LDL-cholesterol by the liver cells. Increased uptake of LDL-cholesterol by the hepatocytes additionally contributes to a lower level of blood LDL-cholesterol.
  • the mode of action may in part resemble the mode of action of ezetimibe, for the cholesterol-absorption inhibitory activity.
  • bacteriopheophytins and quinoneswhen lowering plasma level of LDL-cholesterol is considered, also (or solely) inhibition of enzymes and proteins involved in cholesterol synthesis and transport such as Niemann-Pick C1 Like 1 , ATP-binding cassette transporters such as ABCG5 and ABCG8, and 3-hydroxy-3-methylglutaryl-CoA reductase, by the geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10
  • a composition comprising at least one of, preferably at least two of, more preferably all of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10 or a pharmaceutical composition comprising at least one, preferably at least two, more preferably all of geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rho
  • the carotenoid, bacteriopheophytin and/or quinone is taken orally at the start of e.g. a cholesterol-comprising meal, snack, etc., during consumption thereof, and/or shortly thereafter.
  • a composition comprising at least one, preferably at least two, more preferably all of geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10 or a pharmaceutical composition comprising at least one, preferably at least two, more preferably all of geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhod
  • the subject is administered the composition or pharmaceutical composition at any time during the day at which a cholesterol-rich product is consumed.
  • the (human) subject to whom the composition comprising at least one, preferably at least two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, more preferably all thirteen of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4- spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10 or the pharmaceutical composition comprising at least one, preferably at least two, more preferably all of compounds geranyl- geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin,
  • the human subject has an LDL-cholesterol blood level of 2,94-4,71 mmol/L or 2,50-4,00 mmol/L.
  • the LDL-cholesterol plasma level of a human subject is at least 95 mg/dL such as at least 100 mg/dL, and typically lower than 240 mg/dL, such as lower than 200 mg/dL, typically lower than 190 mg/dL or lower than 159 mg/dL.
  • a composition comprising at least one, preferably at least two, most preferably all of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10, or the pharmaceutical composition comprising at least one, preferably at least two, more preferably all of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxant
  • composition comprising at least one, preferably at least two, most preferably all of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4- rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10, or the pharmaceutical composition comprising at least one, preferably at least two, more preferably all of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin,
  • the geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10 is/are the sole active pharmaceutical ingredients) administered to the human subject, or the geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10,
  • the lUPAC name of the carotenoid rhodovibrin is (6E,8E,10E,12E,14E,16E,18E,20E,22E,24E,26E,28E)-31 -methoxy-2,6,10,14,19,23,27,31 - octamethyldotriaconta-6,8,10,12,14,16,18,20,22,24,26,28-dodecaen-2-ol; the molecular weight of rhodovibrin is 584.929 g/mol.
  • Rhodovibrin is isolated from for example R.rubmm bacterium cells (See also Example 2-4, here below), and rhodovibrin is synthesized (See for example Example 5, here below).
  • the carotenoid is lipophilic and is suitably dissolved in a solvent for dissolving lipophilic agents, such as a fat or an oil or a lipid or lipid composition. That is to say, the rhodovibrin can be mixed with for example an oil or a lipid composition.
  • the oil or lipids then act as a carrier, supporting the ease of (orally) administering the carotenoid to a subject such as a human subject, such as a patient in need thereof, such as a human subject suffering from a CVD or a plasma LDL-cholesterol level of at least 90 mg/dl_.
  • the lUPAC name of the carotenoid 1 -hydroxy-spirilloxanthin is (4E,6E,8E,10E,12E,14E,16E,18E,20E,22E,24E,26E,28E)-31 -methoxy-2,6,10,14,19,23,27,31 - octamethyldotriaconta-4,6,8,10,12,14,16,18,20,22,24,26,28-tridecaen-2-ol; the molecular weight of 1 - hydroxy-spirilloxanthin is 582.913 g/mol.
  • 1 -hydroxy-spirilloxanthin is isolated from for example R.rubmm bacterium cells (See also the Examples 2-4, here below).
  • the carotenoid is lipophilic and is suitably dissolved in a solvent for dissolving lipophilic agents, such as a fat or an oil or a lipid or lipid composition. That is to say, the 1 -hydroxy-spirilloxanthin can be mixed with for example an oil or a lipid composition.
  • the oil or lipids then act as a carrier, supporting the ease of (orally) administering the carotenoid to a subject such as a human subject, such as a patient in need thereof, such as a human subject suffering from a CVD or a plasma LDL-cholesterol level of at least 90 mg/dL.
  • the lUPAC name of the carotenoid 3,4-didehydro-rhodopin (also referred to as 3,4-dehydro- rhodopin) is (4E,6E,8E,10E,12E,14E,16E,18E,20E,22E,24E,26E)-2,6,10,14,19,23,27,31 - octamethyldotriaconta-4,6,8,10,12,14,16,18,20,22,24,26,30-tridecaen-2-ol; the molecular weight of 3,4- dehydro-rhodopin is 552.887 g/mol.
  • 3,4-dehydro-rhodopin is isolated from for example R.rubmm bacterium cells (See also Example 2-4, here below), and 3,4-dehydro-rhodopin is synthesized (See for example Example 5, here below).
  • the carotenoid is lipophilic and is suitably dissolved in a solvent for dissolving lipophilic agents, such as a fat or an oil or a lipid or lipid composition. That is to say, the 3,4- dehydro-rhodopin can be mixed with for example an oil or a lipid composition.
  • the oil or lipids then act as a carrier, supporting the ease of (orally) administering the carotenoid to a subject such as a human subject, such as a patient in need thereof, such as a human subject suffering from a CVD or a plasma LDL-cholesterol level of at least 90 mg/dL.
  • the lUPAC name of the carotenoid chloroxanthin is (6E,8E,10E,12E,14E,16E,18E,20E,22E,26E)-2,6,10,14,19,23,27,31 -octamethyldotriaconta-
  • Chloroxanthin is isolated from for example R.rubrum bacterium cells (See also Example 2-4, here below), and chloroxanthin is chemically synthesized (See for example Example 5, here below). Chloroxanthin is also referred to as hydroxyneurosporene.
  • the carotenoid is lipophilic and is suitably dissolved in a solvent for dissolving lipophilic agents, such as a fat or an oil or a lipid or lipid composition. That is to say, the chloroxanthin can be mixed with for example an oil or a lipid composition.
  • the oil or lipids then act as a carrier, supporting the ease of (orally) administering the carotenoid to a subject such as a human subject, such as a patient in need thereof, such as a human subject suffering from a CVD or a plasma LDL-cholesterol level of at least 90 mg/dL.
  • the lUPAC name of the carotenoid rhodopin is (6E,8E,10E,12E,14E,16E,18E,20E,22E,24E,26E)-2,6,10,14,19,23,27,31 -octamethyldotriaconta-
  • Rhodopin is isolated from for example R.rubrum bacterium cells (See also Example 2-4, here below), and rhodopin is synthesized (See for example Example 5, here below).
  • the carotenoid is lipophilic and is suitably dissolved in a solvent for dissolving lipophilic agents, such as a fat or an oil or a lipid or lipid composition. That is to say, the rhodopin can be mixed with for example an oil or a lipid composition.
  • the oil or lipids then act as a carrier, supporting the ease of (orally) administering the carotenoid to a subject such as a human subject, such as a patient in need thereof, such as a human subject suffering from a CVD or a plasma LDL-cholesterol level of at least 90 mg/dL.
  • the lUPAC name of the carotenoid spirilloxanthin is (4E,6E,8E,10E,12E,14E,16E,18E,20E,22E,24E,26E,28E)-2,31 -dimethoxy-2,6,10,14,19,23,27,31 - octamethyldotriaconta-4,6,8,10,12,14,16,18,20,22,24,26,28-tridecaene; the molecular weight of spirilloxanthin is 596.94 g/mol.
  • Spirilloxanthin also referred to as rhodoviolascin
  • rhodoviolascin is isolated from for example R.rubrum bacterium cells (See also Example 2-4, here below), and spirilloxanthin is chemically synthesized (See for example Example 5, here below).
  • the carotenoid is lipophilic and is suitably dissolved in a solvent for dissolving lipophilic agents, such as a fat or an oil or a lipid or lipid composition. That is to say, the spirilloxanthin can be mixed with for example an oil or a lipid composition.
  • the oil or lipids then act as a carrier, supporting the ease of (orally) administering the carotenoid to a subject such as a human subject, such as a patient in need thereof, such as a human subject suffering from a CVD or a plasma LDL-cholesterol level of at least 90 mg/dL.
  • the lUPAC name of the carotenoid 3,4-dihyd ro-spirilloxanthin is (4E,6E,8E,10E,12E,14E,16E,18E,20E,22E,24E,26E)-2,31 -dimethoxy-2,6,10,14,19,23,27,31 - octamethyldotriaconta-4,6,8,10,12,14,16,18,20,22,24,26-dodecaene; the molecular weight of rhodopin is 598.956 g/mol.
  • 3,4-dihydro-spirilloxanthin is isolated from for example R.rubrum bacterium cells (See also Example 2-4, here below).
  • the carotenoid is lipophilic and is suitably dissolved in a solvent for dissolving lipophilic agents, such as a fat or an oil or a lipid or lipid composition. That is to say, the 3,4- dihyd ro-spirilloxanthin can be mixed with for example an oil or a lipid composition.
  • the oil or lipids then act as a carrier, supporting the ease of (orally) administering the carotenoid to a subject such as a human subject, such as a patient in need thereof, such as a human subject suffering from a CVD or a plasma LDL-cholesterol level of at least 90 mg/dL.
  • Geranyl-geranyl bacteriopheophytin a is isolated from for example R.rubrum bacterium cells (See also Example 2-4, here below).
  • the pheophytin, a bacteriochlorophyll derivative lacking the Mg 2+ ion, is lipophilic and is suitably dissolved in a solvent for dissolving lipophilic agents, such as a fat or an oil or a lipid or lipid composition. That is to say, the geranyl-geranyl bacteriopheophytin a can be mixed with for example an oil or a lipid composition.
  • the oil or lipids then act as a carrier, supporting the ease of (orally) administering the pheophytin to a subject such as a human subject, such as a patient in need thereof, such as a human subject suffering from a CVD or a plasma LDL-cholesterol level of at least 90 mg/dL.
  • Phytyl bacteriopheophytin a is isolated from for example R.rubrum bacterium cells (See also Example 2-4, here below).
  • the pheophytin, a bacteriochlorophyll derivative lacking the Mg 2+ ion, is lipophilic and is suitably dissolved in a solvent for dissolving lipophilic agents, such as a fat or an oil or a lipid or lipid composition. That is to say, the phytyl bacteriopheophytin a can be mixed with for example an oil or a lipid composition.
  • the oil or lipids then act as a carrier, supporting the ease of (orally) administering the pheophytin to a subject such as a human subject, such as a patient in need thereof, such as a human subject suffering from a CVD or a plasma LDL-cholesterol level of at least 90 mg/dL.
  • the lUPAC name of the quinone ubiquinol-10 is 2-[(2E,6E,10E,14E,18E,22E,26E,30E,34E)- 3,7,1 1 ,15,19,23,27,31 ,35,39-decamethyltetraconta-2,6,10,14,18,22,26,30,34,38-decaenyl]-5,6- dimethoxy-3-methyl benzene-1 ,4-diol; the molecular weight of ubiquinol-10 is 865.381 g/mol.
  • Ubiquinol- 10 is isolated from for example R.rubrum bacterium cells (See also Example 2-4, here below), or ubiquinol-10 is synthesized.
  • the ubiquinol is lipophilic and is suitably dissolved in a solvent for dissolving lipophilic agents, such as a fat or an oil or a lipid or lipid composition. That is to say, the ubiquinol-10 can be mixed with for example an oil or a lipid composition.
  • the oil or lipids then act as a carrier, supporting the ease of (orally) administering the ubiquinol to a subject such as a human subject, such as a patient in need thereof, such as a human subject suffering from a CVD or a plasma LDL-cholesterol level of at least 90 mg/dL.
  • the lUPAC name of the quinone ubiquinone-9 is 2,3-diniethoxy-5-niethyl-6- [(2E,6E,1 QE,14E,1 SE,22E,26E,3GE)-3,7,1 1 ,15,19,23,27,31 ,35-nonaniethylhe.xatriaconta- 2,6,10,14,1 S,22,28,3Q,34-nonaenyi]cyclobexa-2,5-diene-1 ,4-dione; the molecular weight of ubiquinone- 9 is 795.246 g/mol.
  • Ubiquinone-9 is isolated from for example R.rubrum bacterium cells (See also Example 2-4, here below), or ubiquinone-9 is synthesized (See Examples section, here below).
  • the ubiquinone is lipophilic and is suitably dissolved in a solvent for dissolving lipophilic agents, such as a fat or an oil or a lipid or lipid composition. That is to say, the ubiquinone-9 can be mixed with for example an oil or a lipid composition.
  • the oil or lipids then act as a carrier, supporting the ease of (orally) administering the ubiquinone to a subject such as a human subject, such as a patient in need thereof, such as a human subject suffering from a CVD or a plasma LDL-cholesterol level of at least 90 mg/dL.
  • the lUPAC name of the quinone ubiquinone-9 is 2,3-dimethoxy-5-methyl-6- [(2E,6E,1 GE,14E,18E,22E,26E,3QE)-3,7,1 1 ,15,19,23,27,31 ,35-nonamethyihexatriaconta- 2,6,10 14,18,22,26 30 34-nonaenyi]eyciohexa-2 : 5-diene-1 ,4-dione; the molecular weight of ubiquinone- 9 is 795 246 g/mol.
  • Ubiquinone-9 is isolated from for example R.rubrum bacterium cells (See also Example 2-4, here below), or ubiquinone-9 is synthesized (See Examples section, here below).
  • the ubiquinone is lipophilic and is suitably dissolved in a solvent for dissolving lipophilic agents, such as a fat or an oil or a lipid or lipid composition. That is to say, the ubiquinone-9 can be mixed with for example an oil or a lipid composition.
  • the oil or lipids then act as a carrier, supporting the ease of (orally) administering the ubiquinone to a subject such as a human subject, such as a patient in need thereof, such as a human subject suffering from a CVD or a plasma LDL-cholesterol level of at least 90 mg/dL.
  • the lUPAC name of the ubiquinone ubiquinone-10 is 2- [(2E,6E,10E,14E,18E,22E,26E,30E,34E)-3,7,1 1 ,15,19,23,27,31 ,35,39-Decamethyltetraconta-
  • ubiquinone-10 is isolated from for example R.rubrum bacterium cells (See also Example 2-4, here below), or ubiquinone-10 is synthesized (See Examples section, here below).
  • the ubiquinone is lipophilic and is suitably dissolved in a solvent for dissolving lipophilic agents, such as a fat or an oil or a lipid or lipid composition.
  • the ubiquinone-10 can be mixed with for example an oil or a lipid composition.
  • the oil or lipids then act as a carrier, supporting the ease of (orally) administering the ubiquinone to a subject such as a human subject, such as a patient in need thereof, such as a human subject suffering from a CVD or a plasma LDL-cholesterol level of at least 90 mg/dL.
  • the lUPAC name of the ubiquinone derivative rhodoquinone-10 is 2-amino-5- [(2E,6E,10E,14E,18E,22E,26E,30E,34E)-3,7,1 1 ,15,19,23,27,31 ,35,39-decamethyltetraconta-
  • Rhodoquinone-10 is isolated from for example R.rubrum bacterium cells (See also Example 2-4, here below), or rhodoquinone-10 is synthesized (See Examples section, here below).
  • the ubiquinone derivative is lipophilic and is suitably dissolved in a solvent for dissolving lipophilic agents, such as a fat or an oil or a lipid or lipid composition.
  • the rhodoquinone-10 can be mixed with for example an oil or a lipid composition.
  • the oil or lipids then act as a carrier, supporting the ease of (orally) administering the ubiquinone derivative to a subject such as a human subject, such as a patient in need thereof, such as a human subject suffering from a CVD or a plasma LDL-cholesterol level of at least 90 mg/dL.
  • the at least one carotenoid, pheophytin such as a bacteriopheophytin or quinone, e.g. rhodovibrin, hydroxyspirilloxanthin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4- spirilloxanthin, geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10, preferably a combination of odovibrin, hydroxyspirilloxanthin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, geranyl-geranyl bacteriopheophytin a,
  • the subject is for example a human subject, such as a patient in need of treatment of a CVD or a plasma LDL-cholesterol level of over e.g. 1 10 mg/dL.
  • a plasma LDL- cholesterol level of higher than 100 mg/dL is associated with an increased risk for developing cardiovascular disease.
  • a subject for example a human subject, having a plasma LDL- cholesterol level of higher than 100 mg/dL has an increased risk for developing for example atherosclerosis.
  • the subject to whom the carotenoid, pheophytin such as a bacteriopheophtyin or quinone is administered is for example a subject suffering from a CVD or being at risk for developing a CVD or atherosclerosis.
  • the human subject has for example a plasma LDL-cholesterol level of on average 75 mg/dL, or 90 mg/dL or higher, or 100-250 mg/dL such as 120-220 mg/dL or 140-200 mg/dL, 160-180 mg/dL, related to an increased risk for developing a disease or health problem such as a CVD, atherosclerosis, arteriosclerosis, a total cholesterol level of at least 200 mg/dL, an Lp(a) level of at least 14 mg/dL, ischemia.
  • a plasma LDL-cholesterol level of on average 75 mg/dL, or 90 mg/dL or higher, or 100-250 mg/dL such as 120-220 mg/dL or 140-200 mg/dL, 160-180 mg/dL, related to an increased risk for developing a disease or health problem such as a CVD, atherosclerosis, arteriosclerosis, a total cholesterol level of at least 200 mg/dL, an Lp(a) level of at least 14 mg/d
  • a composition comprising at least one, preferably at least two, more preferably all of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10 or pharmaceutical composition comprising at least one, preferably at least two, more preferably all of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spir
  • a carotenoid pheophytin such as a bacteriopheophtyin and/or quinone
  • the at least one further active pharmaceutical ingredient and the carotenoid, pheophytin such as a bacteriopheophytin and/or quinone are administered simultaneously, separately, or sequentially.
  • the combination of) the geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10 is/are co-administered, preferably separately, with a statin.
  • the composition comprising the thirteen compounds is administered sequentially with a PCSK9 inhibitor.
  • the composition comprising rodovibrin, hydroxyspirilloxanthin, 3,4- rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, ubiquinol-10, ubiquinone-9, ubiquinone- 10 and rhodoquinone-10 is co-administered with a statin and a PCSK9 inhibitor.
  • the rodovibrin, hydroxyspirilloxanthin, 3,4- rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, ubiquinol-10, ubiquinone-9, ubiquinone- 10 and rhodoquinone-10 are the sole active pharmaceutical active ingredients in a composition comprising rodovibrin, hydroxyspirilloxanthin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4- spirilloxanthin, geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriophe
  • the at least one further active pharmaceutical ingredient is administered to a subject in need thereof simultaneously, separately or sequentially with the (pharmaceutical) composition
  • the (pharmaceutical) composition comprising carotenoid(s), bacteriopheophytin(s) and/or quinone(s), such as geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10, for use in the treatment or prophylaxis of e.g.
  • the at least one carotenoid, bacteriopheophytin and/or quinone is geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4- rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and/or rhodoquinone-10.
  • At least two, more preferably all of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10 are used.
  • all of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10 are used.
  • the at least one further active pharmaceutical ingredient such as a statin or anacetrapib are combined with a composition
  • a composition comprising at least one, preferably at least two, preferably all of geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10 as a single dosage form, e.g. combined in a capsule, tablet, granulate, sachet comprising powdered compounds, etc.
  • the composition or pharmaceutical composition according to the invention and further active pharmaceutical ingredient(s) are administered orally or are administered using a different route, although oral administration is preferred.
  • composition or pharmaceutical composition is also used in a method for the treatment or the prophylaxis of cardiovascular disease, wherein administering at least one, preferably at least two, more preferably all of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10 to a (human) subject replaces previous administration of an active pharmaceutical ingredient that was administered for the same purpose of lowering the plasma LDL-cholesterol level in said subject.
  • composition or pharmaceutical composition for use according to the invention replaces for example any pharmaceutical composition comprising a statin, niacin, fenofibrate, ezetimibe, colesevelam, mipomersen, lomitapide, a PCSK9 inhibitor, alirocumab, evolocumab, ETC-1002, a CETP inhibitor, anacetrapib, evacetrapib, WAY-252623, a blood-pressure lowering compound, hydrochlorothiazide.
  • a statin niacin, fenofibrate, ezetimibe, colesevelam, mipomersen, lomitapide, a PCSK9 inhibitor, alirocumab, evolocumab, ETC-1002, a CETP inhibitor, anacetrapib, evacetrapib, WAY-252623, a blood-pressure lowering compound, hydrochlorothiazide.
  • composition or pharmaceutical composition comprising at least one, preferably at least two, more preferably all of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10 for use according to the invention, wherein the composition or pharmaceutical composition is administered orally to a subject, preferably a human subject.
  • alternative routes of administration are equally preferred if applicable for administering a sufficient amount of the compound(s), composition or pharmaceutical composition to a subject in need thereof, such as administration using a parenteral route, e.g. by injection intradermally or intravenously, or by infusion such as intravenously, wherein the administered amount of the compound(s), composition or pharmaceutical composition is sufficient to induce a plasma LDL-cholesterol level lowering effect in the (human) subject to whom the compound(s), composition or pharmaceutical composition is/are administered.
  • the compound(s), composition or pharmaceutical composition is/are administered to a human subject in need thereof with the aim of lowering the plasma LDL-cholesterol level to a level of 100 mg/dL or lower such as 70 mg/dL, or lower, e.g. 60 mg/dL or lower.
  • geranyl-geranyl bacteriopheophytin a phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4- spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10 and in some embodiments at least one further active pharmaceutical ingredient, is for example a human patient suffering from ischemia, a CVD, a high plasma LDL-cholesterol level of at least 150 mg/dL.
  • composition or pharmaceutical composition according to the invention preferably comprises administering the carotenoids, bacteriopheophtyins and quinones to a subject, said administering resulting in lowering of the plasma LDL-cholesterol concentration in the subject, preferably to a plasma concentration of less than 3,34 mmol/L, preferably less than 2,59 mmol/L, more preferably to a plasma LDL-cholesterol concentration of less than 1 ,8 mmol/L.
  • composition or pharmaceutical composition for use according to the invention wherein administering (a pharmaceutically effective dose of) the at least one, preferably at least two, preferably all of geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4- spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10 to a subject (in need thereof) results in a decrease of the LDL-cholesterol concentration in the plasma of said subject, wherein the plasma HDL-cholesterol concentration remains essentially unaltered or decreases to a smaller extent than the decrease in the plasma LDL-cholesterol concentration (HDL-cholesterol level increases in a relative sense),
  • the plasma HDL- cholesterol level is in an embodiment at least 40 mg/dL, or at least 60 mg/dL, in the plasma of a (human) subject to whom the composition or pharmaceutical composition is administered. Desired is a plasma HDL-cholesterol level of 60 mg/dL or higher.
  • the decrease of the LDL-cholesterol concentration is expressed as the percentage of the plasma LDL-cholesterol concentration prior to the administration of (an effective dose of) (a composition comprising) the carotenoid(s), bacteriopheophtin(s) and/or quinone(s), such as geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4- spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10, to said (human) subject (in need of cholesterol-lowering treatment), and the change of the HDL-cholesterol concentration is expressed as a percentage of the plasma HDL-cholesterol concentration prior to the administration of the composition
  • the subject is for example a human subject, and the human subject is preferably a human subject in need of a reduction of the plasma LDL-cholesterol concentration in view of treating or prophylaxis of a CVD or one or more of the further diseases or risk factors for developing a disease, as here above listed.
  • the human subject in need thereof is for example a human subject suffering from any one or more of CVD, atherosclerosis, dyslipidemia, arteriosclerosis, hypercholesterolemia, familial hypercholesterolemia, hyperlipidemia, an LDL-cholesterol plasma level of at least 70 mg/dL, an LDL-cholesterol plasma level of at least 100 mg/dL, an LDL-cholesterol plasma level of at least 140 mg/dL, an LDL-cholesterol plasma level of at least 200 mg/dL, a total plasma cholesterol level of at least 200 mg/dL, an Lp(a) level of at least 14 mg/dL, ischemia.
  • the human subject in need of treatment with an effective amount of at least one carotenoid, bacteriopheophytin or quinone e.g. a composition comprising geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4- rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10, is for example a human subject having an increased cardiovascular risk factor and/or an increased atherosclerosis risk factor, such as a plasma LDL- cholesterol level of 100 mg/dL or higher and/or a total plasma cholesterol level of 200 mg/dL or higher, such as 200 mg/dL - 239 mg/dL or 240 mg
  • the human subject is administered at least two, preferably all of geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10.
  • the human subject is administered all of geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10.
  • An embodiment is the composition or pharmaceutical composition for use according to the invention, wherein administering the at least one carotenoid, bacteriopheophytin or quinone, e.g. all of geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10, to a subject results in a decrease of the plasma LDL-cholesterol concentration in said subject with at least 5%, preferably at least 10%, more preferably at least 20%, most preferably at least 30%, based on the plasma LDL-cholesterol concentration prior to the administration of the carotenoid, bacterioph
  • the administered amount of carotenoid (s), bacteriopheophytin(s) and/or quinone(s), such as geranyl- geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10, to a human subject in need thereof is an amount sufficient to lower the plasma LDL-cholesterol concentration in said human subject to a level of lower than 200 mg/dL such as lower than 160 mg/dL, or lower than 145 mg/dL, lower than 120 mg/dL, lower than 100 mg/dL, preferably 70 mg/dL or lower.
  • composition or pharmaceutical composition is for example administered to a human subject suffering from a risk factor for developing cardiovascular disease or atherosclerosis, such as a plasma LDL-cholesterol level of about 100 mg/dL or about 200 mg/dL, wherein the administering of the carotenoid results in a decrease of the plasma LDL-cholesterol level to about 70 mg/dL or lower, or to about 140 mg/dL or lower, respectively.
  • a risk factor for developing cardiovascular disease or atherosclerosis such as a plasma LDL-cholesterol level of about 100 mg/dL or about 200 mg/dL
  • the administering of the carotenoid results in a decrease of the plasma LDL-cholesterol level to about 70 mg/dL or lower, or to about 140 mg/dL or lower, respectively.
  • administering the composition or pharmaceutical composition comprising at least one carotenoid, bacteripheophytin and/or ubiquinone, e.g. in a pharmaceutically effective amount, to a subject such as a human subject in need thereof results in a decrease of the plasma LDL-cholesterol concentration to, or maintenance of the plasma LDL-cholesterol concentration at, a plasma LDL- cholesterol concentration of less than 200 mg/dL, preferably less than 159 mg/dL, more preferably less than 129 mg/dL, most preferably less than 100 mg/dL, such as less than 70 mg/dL, or even lower such as less than 60 mg/dL.
  • the carotenoid(s), bacteriopheophytin(s) and/or quinone(s) such as
  • the composition or pharmaceutical composition comprising at least one, preferably at least two, preferably all of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10 is formulated as an oral dosage form, preferably a solid oral dosage form or a liquid oral dosage form, preferably a liquid oral dosage form comprising an oil and/or at least one lipid such as a phospholipid and/or a fat, preferably comprising omega-3 fatty acids and/or omega-6 fatty acids.
  • An oral dosage form is for example self-administered conveniently by the patient in need of LDL-cholesterol lowering treatment.
  • the oral dosage form is for example provided as a single capsule or tablet, or as multiple capsules or tablets for (self-)administration once daily or during more than one occasion during a day, such as twice or thrice daily or before or after a meal.
  • the composition or pharmaceutical composition for use according to the invention is in an embodiment formulated as a tablet or a capsule or a powder or a granulate.
  • composition or pharmaceutical composition comprising at least one, preferably at least two, more preferably all compounds of geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10 for use according to the invention, wherein the daily dose of the at least one, preferably at least two, more preferably all of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chlorox
  • a daily dose of the active pharmaceutical ingredient of less than 10 microgram per day or more than 150 mg per day.
  • a daily dose of the active pharmaceutical ingredient of less than 10 microgram per day or more than 30 g per day.
  • the daily dose of rhodovibrin is 10 mg/kg - 500 mg/kg, such as 25 mg/kg - 250 mg/kg, or 50 mg/kg - 200 mg/kg, based on the weight of the human subject.
  • the effective daily dose is the dose administered to the patient in need thereof, that results in the desired lowering of plasma LDL-cholesterol level aimed for.
  • the carotenoid such as rhodovibrin is administered to a human subject resulting in the LDL- cholesterol level in the blood plasma lowering from any initial value before treatment of 120 mg/dL - 159 mg/dL, to a value lower than said initial value, such as a LDL-cholesterol level of between 60 mg/dL and 140 mg/dL, or lower than 120 mg/dL.
  • the at least one carotenoid, bacteriopheophytin and/or quinone such as geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4- spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10, is administered to a human subject resulting in the LDL-cholesterol level in the blood plasma lowering from any initial value before treatment of 95 mg/dL - 190 mg/dL, or 95 mg/dL - 155 mg/dL, to a value lower than said initial value, such as a LDL-cholesterol level of between 60 mg/dL and 140 mg/dL
  • plasma LDL-cholesterol levels in a human subject are lower than 100 mg/dL, such as lower than 70 mg/dL, and that the plasma LDL-cholesterol levels are maintained at such low values, therewith diminishing the risk for developing a cardiovascular disease and/or any of atherosclerosis, dyslipidemia, arteriosclerosis, inflammation, inflammatory disease, ischemia, infection.
  • An aspect of the invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a pharmaceutically effective amount of at least one carotenoid, pheophtyin such as a bacteriopheophytin and/or a quinone, such as geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4- spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10, and optionally a pharmaceutically acceptable excipient for use in lowering plasma cholesterol, wherein the pharmaceutical composition is not, or does not comprise, a petroleum ether extract of Rhodospirillum rubrum obtainable by extraction of Rhod
  • the pharmaceutical composition comprises at least two, preferably all of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10.
  • the pharmaceutical composition comprises all of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4- spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10.
  • An aspect of the invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a pharmaceutically effective amount of at least one carotenoid, pheophytin such as a bacteriopheophytin and/or quinone, such as geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4- spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10, and optionally a pharmaceutically acceptable excipient for use in the lowering of LDL-cholesterol in blood plasma of a subject, wherein the pharmaceutical composition is not, or does not comprise, a petroleum ether extract of Rhodospirill
  • the pharmaceutical composition comprises at least two, preferably all of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4- rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10.
  • the pharmaceutical composition comprises all of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10.
  • An aspect of the invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a pharmaceutically effective amount of at least one carotenoid, pheophytin such as a bacteriopheophytin and/or quinone, such as geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4- spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10, and optionally a pharmaceutically acceptable excipient for use in a method for the lowering of LDL-cholesterol in blood plasma of a subject, wherein the pharmaceutical composition is not, or does not comprise, a petroleum ether extract of Rhod
  • the pharmaceutical composition comprises at least two, preferably all of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10.
  • the pharmaceutical composition comprises all of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10.
  • An aspect of the invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a pharmaceutically effective amount of at least one carotenoid, pheophytin such as a bacteriopheophytin and/or quinone, such as geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4- spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10, and optionally a pharmaceutically acceptable excipient, for use in a method for the treatment or the prophylaxis of any one or more of cardiovascular disease, such as coronary heart disease, atherosclerosis, dyslipidemia, arteriosclerosis, hyper
  • the pharmaceutical composition comprises at least two of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4- spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10.
  • the pharmaceutical composition comprises all of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4- rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10.
  • an embodiment is the pharmaceutical composition comprising a pharmaceutically effective amount of at least one carotenoid, pheophytin such as a bacteriopheophytin and/or quinone and optionally a pharmaceutically acceptable excipient, for use in a method for the treatment or the prophylaxis of a symptom or a risk factor of any one or more of a cardiovascular disease, atherosclerosis, dyslipidemia, arteriosclerosis, hypercholesterolemia, familial hypercholesterolemia, hyperlipidemia, inflammation, inflammatory disease, ischemia, infection.
  • pheophytin such as a bacteriopheophytin and/or quinone
  • a pharmaceutically acceptable excipient for use in a method for the treatment or the prophylaxis of a symptom or a risk factor of any one or more of a cardiovascular disease, atherosclerosis, dyslipidemia, arteriosclerosis, hypercholesterolemia, familial hypercholesterolemia, hyperlipidemia, inflammation, inflammatory disease
  • the at least one carotenoid, pheophytin such as a bacteriopheophytin and/or quinone, e.g.
  • geranyl-geranyl bacteriopheophytin a phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10, is/are either the sole active pharmaceutical ingredient(s) in the pharmaceutical composition, or the pharmaceutical composition is administered to a subject to whom at least one further active pharmaceutical ingredient is administered, such as an active pharmaceutical ingredient selected from a statin, niacin, fenofibrate, ezetimibe, colesevelam, mipomersen, lomitapide, a PCSK9 inhibitor, alirocumab, evolocumab, ETC-1002, a CETP inhibitor, anacetrapi
  • An aspect of the invention relates to a method for treating or preventing a cardiovascular disease, atherosclerosis, dyslipidemia, arteriosclerosis, hypercholesterolemia, familial hypercholesterolemia, hyperlipidemia, an LDL-cholesterol plasma level of at least 70 mg/dL, an LDL- cholesterol plasma level of at least 100 mg/dL, an LDL-cholesterol plasma level of at least 140 mg/dL, an LDL-cholesterol plasma level of at least 200 mg/dL, a total plasma cholesterol level of at least 200 mg/dL, an Lp(a) level of at least 14 mg/dL, inflammation, inflammatory disease, ischemia, infection, comprising administering in a subject in need thereof an effective amount of a composition comprising at least one carotenoid, pheophytin such as a bacteriopheophytin and/or quinone, such as geranyl- geranyl bacteriopheophytin a, phytyl derivative of bacteri
  • the composition comprises at least two of compounds geranyl- geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10.
  • the composition comprises all of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10.
  • the invention also relates to a method for treating any symptom or risk factor related to any one or more of these health problems and diseases.
  • the pharmaceutical composition for use wherein (an effective dose of) the pharmaceutical composition is administered to a (human) subject (in need thereof) suffering from an LDL-cholesterol plasma level of at least 70 mg/dL, an LDL-cholesterol plasma level of at least 100 mg/dL, an LDL-cholesterol plasma level of at least 140 mg/dL, an LDL-cholesterol plasma level of at least 200 mg/dL, a CVD.
  • an effective dose of the pharmaceutical composition is administered to a (human) subject (in need thereof) suffering from an LDL-cholesterol plasma level of at least 70 mg/dL, an LDL-cholesterol plasma level of at least 100 mg/dL, an LDL-cholesterol plasma level of at least 140 mg/dL, an LDL-cholesterol plasma level of at least 200 mg/dL, a CVD.
  • Equally preferred is the pharmaceutical composition for use, wherein the pharmaceutical composition is administered to a subject suffering from a total plasma cholesterol level of 200 mg/dL or higher and/or
  • An aspect of the invention relates to a method of treating a patient suffering from an LDL- cholesterol concentration in the plasma of said patient of above 100 mg/dL by administering a composition comprising an effective amount of at least one carotenoid, pheophytin such as a bacteriopheophytin and/or quinone, such as geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10 to the patient, wherein the composition is not, or does not comprise, a petroleum ether extract of Rhodospirillum rubrum obtainable by extraction of Rh
  • the composition comprises at least two of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10.
  • the composition comprises all of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10.
  • An aspect of the invention relates to a method of treating a patient suffering from or having an increased risk for any one or more of cardiovascular disease, atherosclerosis, dyslipidemia, arteriosclerosis, hypercholesterolemia, familial hypercholesterolemia, hyperlipidemia, an LDL- cholesterol plasma level of at least 70 mg/dL, an LDL-cholesterol plasma level of at least 100 mg/dL, an LDL-cholesterol plasma level of at least 140 mg/dL, an LDL-cholesterol plasma level of at least 200 mg/dL, a total plasma cholesterol level of at least 200 mg/dL, an Lp(a) level of at least 14 mg/dL, inflammation, inflammatory disease, ischemia, infection, by administering a composition comprising an effective amount of at least one carotenoid, pheophytin such as a bacteriopheophytin and/or quinone, such as geranyl-geranyl bacteriopheophytin a, phytyl derivative of
  • the composition comprises at least two, preferably all of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10.
  • the composition comprises all of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4- rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10.
  • the subject is suffering from a CVD and/or the subject is at risk for developing a CVD.
  • the CVD being due to atherosclerosis, relating to any of ischaemic heart disease or coronary artery disease (e.g. heart attack), cerebrovascular disease (e.g. stroke), diseases of the aorta and arteries, including hypertension and peripheral vascular disease, and/or the CVD is congenital heart disease, rheumatic heart disease, cardiomyopathies, or cardiac arrhythmias.
  • An aspect of the current invention relates to a food supplement or a feed supplement with LDL- cholesterol lowering properties, comprising at least one, two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, preferably thirtheen of a carotenoid, pheophytin such as a bacteriopheophytin and/or quinone, such as geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10.
  • pheophytin such as a bacteriopheophytin
  • a food supplement and a feed supplement is defined as a formulation that is consumed in addition to a normal diet and that contains compounds or components that do not occur in a normal diet, or that occur in low amounts or in insufficient amounts, while sufficient or increased consumption of these components is desired.
  • a food supplement according to the invention is composed such that it is suitable for human consumption. Consequently, a food supplement as defined in the present invention should preferably have a texture, taste and smell, but also a nutritional value, that makes the supplement suitable for human consumption.
  • a feed supplement according to the invention is composed such that it is suitable for animal consumption, such as consumption by poultry such as laying hens, chicken, cow, pig, goat, horse, sheep, dog, cat, rabbit, etc. Consequently, a feed supplement as defined in the present invention should preferably have a texture, taste and smell, but also a nutritional value, that makes the supplement suitable for animal consumption.
  • the food supplement or feed supplement comprises at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, at least eleven, at least twelve or at least thirteen, preferably all thirteen of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4- rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10.
  • the food or feed supplement comprises all of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10.
  • a food supplement or a feed supplement according to the invention preferably contains between 0.01 % and 99.9% (w/w) of at least one carotenoid, pheophytin such as a bacteriopheophtyin and/or quinone, such as geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4- spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10, for example 0.05%- 50% by weight based on the total weight of the food supplement or the feed supplement.
  • pheophytin such as a bacteriopheophtyin and/or quinone
  • a food supplement or a feed supplement contains between 10% and 90% (w/w), or between 30% and 75% (w/w), of a preparation of at least one carotenoid, pheophytin such as a bacteriopheophytin and/or quinone, such as geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4- spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10.
  • components are preferably added to improve, for instance, texture, taste or smell.
  • a food supplement or a feed supplement preferably comprises (additional) sources of protein, carbohydrate and fat, and vitamins, minerals, electrolytes, trace elements, and other suitable components, so that the food supplement or the feed supplement itself is suitable for use as a nourishing food.
  • each and every protein that is suitable for use in nutritional formulations, and mixtures of these, are preferably used in a food supplement or a feed supplement according to the invention.
  • This type of proteins encompasses for instance animal proteins such as whey proteins, whey protein concentrates, whey powder, egg protein, egg albumin, casein, or milk albumin, and plant proteins such as soy protein, soy meal, or proteins from soy milk.
  • animal proteins such as whey proteins, whey protein concentrates, whey powder, egg protein, egg albumin, casein, or milk albumin
  • plant proteins such as soy protein, soy meal, or proteins from soy milk.
  • the biological value of a protein may constitute an important criterion.
  • Caseinate including calcium caseinate, but also whey, milk albumin, egg albumin, and total egg proteins, for instance, are proteins with a very high biological value, because they contain a large proportion of essential amino acids.
  • Suitable carbohydrates to be used in a food supplement or a feed supplement according to the invention are, for instance, preferably simple short-chain carbohydrates such as mono- and disaccharides, but also polysaccharides, or a combination of both.
  • a carbohydrate is preferably selected because of its suitable organoleptic properties, according to the invention.
  • a complex carbohydrate is suitably used as a food fiber, according to the invention.
  • a food supplement or a feed supplement according to the invention preferably contains, in some embodiments, combinations of both simple and complex carbohydrates.
  • a food supplement or a feed supplement according to the invention preferably contains, in some embodiments, a fat selected from all edible oils and edible fats.
  • Vitamins and minerals are preferably added to a preparation according to the invention, in conformity with the rules of the regulatory health authorities, and preferably encompasses all vitamins and minerals endorsed by the above authorities, for instance vitamin A, B1 , B2, B12, C, D, B, and K, and folic acid, niacin, pantothenic acid, and biotin.
  • vitamins and minerals endorsed by the above authorities for instance vitamin A, B1 , B2, B12, C, D, B, and K, and folic acid, niacin, pantothenic acid, and biotin.
  • minerals for instance iron, zinc, iodine, calcium, magnesium, chromium, and selenium are preferably added to a preparation according to the invention.
  • Electrolytes such as the ions of sodium, potassium, and chloride, and trace elements and other additives do preferably also form part of a food supplement or a feed supplement according to the invention.
  • Such components are, if present, preferably used in the recommended concentrations.
  • a food supplement or a feed supplement according to the invention preferably contains components improving its texture, colorings and flavorings, aromatic substances, spices, fillers, emulsifiers, stabilizing compounds, preservatives, antioxidants, fibers, and other supplements such as amino acids, choline, lecithin, fatty acids, etc.
  • the choice of such components depends upon formulation, design, and preferences.
  • Emulsifiers are preferably added to stabilize the final product of the invention.
  • acceptable emulsifiers are lecithin (e.g . , derived from soy or from egg), and/or mono- and di-glycerides, according to the invention.
  • As stabilizers, carob, guar or carrageenan are, for instance, preferably used, according to the invention.
  • Preservatives are preferably added to increase the shelf life of the product of the invention.
  • preservatives such as sodium sorbate, potassium sorbate, potassium benzoate, sodium benzoate, or calcium disodium EDTA are used in a preparation of the invention.
  • natural or synthetic sweeteners such as saccharides, cyclamates, aspartame, acesulfame potassium, and/or sorbitol, are preferably added to the food supplement or to the feed supplement, according to the invention.
  • the amounts of food supplement or of feed supplement of the invention to be consumed are varying in size, and are not necessarily restricted to the dosages mentioned in the dosages advised.
  • the term“food supplement” is not meant to be restricted to a specified weight, or to a specified dose of the food supplement.
  • feed supplement is not meant to be restricted to a specified weight, or to a specified dose of the feed supplement.
  • composition of a food supplement or a feed supplement according to the invention takes in principle any form that is suitable for human or animal consumption, according to the invention.
  • the food supplement or the feed supplement is a dry powder that is suitable to be suspended, dispersed or emulsified in an aqueous solution such as water, milk, coffee, tea, broth, and fruit juice.
  • an aqueous solution such as water, milk, coffee, tea, broth, and fruit juice.
  • the powder is preferably supplied in a dispenser according to the invention.
  • the food supplement or the feed supplement is formulated, starting from dry powder, as a tablet or as a granulate.
  • the composition of a food supplement or a feed supplement according to the invention is suitably supplied with fillers such as microcrystalline cellulose (MCC) and mannitol, binders such as hydroxylpropyl-cellulose (HPC), lubricants such as stearic acid, and other excipients.
  • MCC microcrystalline cellulose
  • HPC hydroxylpropyl-cellulose
  • lubricants such as stearic acid
  • a food supplement or a feed supplement according to the invention is in one embodiment preferably supplied as a fluid, in which the solid components have been suspended, dispersed or emulsified.
  • Such a composition of the invention is preferably directly mixed into a foodstuff or a feedstuff, or is preferably for instance extruded and formatted into granules or other forms.
  • a food supplement or a feed supplement is preferably formulated in a solid form, such as a bar, a biscuit, or a roll.
  • a food supplement or a feed supplement of the invention is preferably formulated for oral consumption, preferably in combination with an acceptable carrier such as a capsule, a tablet, a granulate, a water-miscible powder, or another form acceptable for administration.
  • a food supplement of the invention is preferably processed into a foodstuff, according to the invention.
  • a feed supplement of the invention is preferably processed into a feedstuff, according to the invention.
  • the present invention relates to a foodstuff comprising a food supplement according to the invention.
  • the food supplement comprising at least one carotenoid, pheophytin such as a bacteriopheophytin and/or quinone, such as one or more, preferably all of geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4- rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10.
  • One aspect of the present invention relates to a feedstuff comprising a feed supplement according to the invention.
  • the feed supplement comprising at least one, preferably at least two, more preferably all of compoundsgeranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10.
  • the food supplement comprised in the foodstuff comprises at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, at least eleven, at least twelve or at least thirteen, preferably all of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4- rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10.
  • the food supplement comprised in the the foodstuff comprises all of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4- spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10.
  • a food supplement or a feed supplement may suitably be used to reduce intestinal cholesterol absorption, thus reducing the cholesterol level of blood plasma, in particular the plasma LDL-cholesterol concentration, while preferably leaving the HDL-cholesterol concentration essentially unaltered or raising the plasma HDL-cholesterol concentration in a subject, such that the plasma levels of HDL- cholesterol increase in absolute sense or relative to the plasma LDL-cholesterol concentration.
  • the subject being a human subject or an animal subject such as a chicken or laying hen.
  • the invention relates to a food supplement with cholesterol-lowering properties, comprising at least one, preferably at least two, more preferably all compounds of geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10.
  • Feeding an animal such as a broiler, hen, pig, cow, duck, goat, goose, turkey, bovine calf, sheep, in particular a pig, a broiler and a laying hen, with the feed supplement or the feed stuff comprising the feed supplement results in the lowering of the cholesterol level in the blood serum (plasma) of said animal and/or the lowering of the cholesterol content in the meat of said animal and/or in other parts or products derived from said animal and/or in the lowering of the cholesterol content in the eggs laid by the e.g. chicken, duck, goose, turkey, etc.
  • This lowering of the cholesterol content, in particular the LDL- cholesterol content provides for e.g.
  • the human diet comprises less cholesterol, in particular less LDL-cholesterol, when the consumed meat and/or eggs by the human subject are derived from animals fed with the feed supplement or the feedstuff of the invention. As a consequence, less cholesterol is transported from the intestine to the blood circulation, the liver and further organs of the human subject.
  • a food supplement of the invention is applied in a foodstuff with cholesterollowering properties.
  • a feed supplement of the invention is applied in a feedstuff with cholesterol-lowering properties.
  • a method to prepare a cholesterol-lowering foodstuff or feedstuff of the invention involves the production of a foodstuff or feedstuff, respectively, incorporating a food supplement or a feed supplement according to the invention.
  • Such a method preferably involves a step in which a foodstuff or a feedstuff is first prepared in the normal way, followed by the addition of at least one carotenoid, pheophtyin such as a bacteriopheophytin and/or quinone, such as preferably all of geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4- rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and
  • At least two of compounds as geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4- spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10 is added.
  • a foodstuff with cholesterol-lowering properties according to the invention or a feedstuff with cholesterol-lowering properties according to the invention contains typically between 0.1 % and 20% (w/w), preferably between 1 % and 10% (w/w), of the food supplement or feed supplement according to the invention and described above.
  • the feedstuff is for example chicken feed or feed for laying hen.
  • the invention relates to a foodstuff comprising a food supplement, wherein the food supplement has cholesterol-lowering properties and comprises at least one, preferably at least two, preferably all of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4- spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10.
  • the carotenoid(s) is/are for example natural carotenoid(s) and is/are for example isolated from a natural source or is/are chemically synthesized.
  • the pheophytin(s) is/are for example natural pheophytin(s) and are for example isolated from a natural source or are chemically synthesized.
  • the quinone, ubiquinone or ubiquinol or ubiquinone derivative such as rhodoquinone is/are for example natural quinone, ubiquinone or ubiquinol or a naturally occurring ubiquinone derivative such as rhodoquinone, and are for example isolated from a natural source or are chemically synthesized.
  • the pheophytin is for example a chlorophyll derivative lacking the central Mg 2+ ion and is for example a bacteriopheophytin a, for example a bacteriopheophytin a comprising a phytyl group or comprising a geranyl-geranyl group.
  • the carotenoid(s) is/are preferably tetraterpenoids such as a xanthophyll and a carotene, though xanthophylls are preferred.
  • the carotenoids are preferably polar carotenoids such as xanthophylls, and xanthophylls are preferred.
  • the carotenoids are for example xanthophylls based on the precursor phytoene.
  • the carotenoids are for example xanthophylls based on the precursor xanthophyll rhodopin.
  • the xanthophyll is for example an alcohol, an ether, or has both a hydroxyl group and an ether group.
  • the proviso is that for the composition according to the invention and for the composition for use according to the invention and for the compounds in e.g. the foodstuff of the invention and the food supplement of the invention or the feed of the invention, the composition and the composition for use of the invention and and the compounds in e.g. the foodstuff of the invention and the food supplement of the invention or the feed of the invention, is/are not a preparation comprising the membrane fraction of Rhodospirillum spp.
  • the compounds are not provided as a preparation comprising the membrane fraction of Rhodospirillum spp. and/or the membrane fraction of Phaeospirillum spp., and not as a preparation or pharmaceutical preparation of Rhodospirillum spp.
  • the preparation or pharmaceutical preparation comprising dead Rhodospirillum spp. , or freeze-dried Rhodospirillum spp.
  • the (pharmaceutical) composition of the invention does not comprise or consist of whole R.rubrum cells and is not or does not comprise a preparation comprising the membrane fraction of Rhodospirillum spp. and/or Phaeospirillum spp., and is not or does not comprise a preparation of Rhodospirillum spp. for use in lowering plasma cholesterol, said preparation comprising dead Rhodospirillum spp. , or freeze-dried Rhodospirillum spp. , according to the invention.
  • the current invention provides a composition
  • a composition comprising compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10, under the proviso that the composition is not a petroleum ether extract of Rhodospirillum rubrum obtainable by extraction of Rhodospirillum rubrum cells with a mixture of petroleum ether with a boiling point of between 60°C and 80°C and methanol comprising sodium chloride, wherein the extract is obtained by extraction for between 10 minutes and 48 hours at between 8°C and 37°C, while mixing the cells with the mixture, and
  • Rhodospirillum spp. is not a preparation or pharmaceutical preparation of Rhodospirillum spp. for use in lowering plasma cholesterol, said preparation or pharmaceutical preparation comprising dead Rhodospirillum spp., or freeze-dried Rhodospirillum spp., according to the invention.
  • the measurement of total plasma cholesterol in the mouse plasma was conducted using the in vitro kit “cholesterol” from Beckman Coulter Nederland B.V. (Woerden, Netherlands, Datasheet from 09-2011 , coded BAOSR6x16.02; catalogue No. OSR6116) intended for the quantitative determination of Cholesterol concentrations in serum, EDTA plasma, heparinized plasma on Beckman Coulter AU analyzers. Reference is made to the protocol used as provided by Beckman Coulter reagent kit.
  • Measurements of cholesterol are used primarily in the diagnosis and treatment of disorders involving excess cholesterol in the blood, and lipid and lipoprotein metabolism disorders.
  • Total serum or plasma cholesterol analysis has proven useful in the diagnosis of hyperlipoproteinemia, atherosclerosis, hepatic and thyroid diseases.
  • Total and HDL cholesterols, in conjunction with a triglyceride determination, provide valuable information for the prediction of coronary heart disease.
  • the system reagent of the kit (OSR6116 / OSR6216 /OSR6516) was obtained from Beckman Coulter Nederland B.V. and consists of:
  • the reagent is brought into contact with the plasma in aBeckman Coulter analyzer, which causes the cholesterol esters in the serum to be hydrolyzed by the cholesterol esterase (CHE).
  • the free cholesterol produced in this reaction is oxidized by cholesterol oxidase (CHO) to cholest-4-en-3-one with the simultaneous production of hydrogen peroxide (H2O2), which oxidatively couples with the 4- aminoantipyrine and phenol in the presence of the peroxidase to yield a chromophore.
  • H2O2 hydrogen peroxide
  • the red quinoneimine dye formed through this reaction can be measured spectrophotometrically at 540/600 nm as an increase in absorbance.
  • the measurement of plasma triglyceride levels in the mouse plasma was conducted using the in vitro kit“tryglycerides” from Beckman Coulter Nederland B.V. (Woerden, Netherlands, Datasheet from 1 1 - 2010, coded BAOSR6x1 18.02; catalogue No. OSR601 18) intended for the quantitative determination of Triglyceride concentrations in human serum, EDTA, or heparinized plasma samples on Beckman Coulter All analyzers. Reference is made to the protocol used as provided by Beckman Coulter reagent kit.
  • Triglycerides are the major form of fat found in nature and their primary function is to provide energy for the cell. Measurements of triglyceride are used in the diagnosis and treatment of patients with diabetes mellitus, nephrosis, liver obstruction, other diseases involving lipid metabolism, or various endocrine disorders. Clinically, triglyceride assays are used to help classify the various genetic and metabolic lipoprotein disorders and in the assessment of risk factors for atherosclerosis and coronary artery disease.
  • the system reagent of the kit (OSR601 18 / OSR61 1 18 / OSR661 18) was obtained from Beckman Coulter Nederland B.V. and consists of:
  • Glycerol kinase Bacillus stearothermophiius - 3 0.5 kU/L (8.3 pkat/L)
  • Glycerol phosphate oxidase Pseudomonas ) - > 1 .5 kU/L (25 pkat/L)
  • Ascorbate oxidase Curcubita species ) - > 1 .5 kll/L (25 pkat/L)
  • the Triglyceride Reagents are ready for use. No preparation was required.
  • OSR661 18 the pipe supplied had to be inserted into the 180 ml_ reagent vial before use on the analyzer. The pipe was for single use only.
  • This triglyceride procedure is based on a series of coupled enzymatic reactions.
  • the triglycerides in the sample are hydrolyzed by a combination of microbial lipases to give glycerol and fatty acids.
  • the glycerol is phosphorylated by adenosine triphosphate (ATP) in the presence of glycerol kinase (GK) to produce glycerol-3-phosphate.
  • GK glycerol kinase
  • the glycerol-3-phosphate is oxidized by molecular oxygen in the presence of GPO (glycerol phosphate oxidase) to produce hydrogen peroxide (H2O2) and dihydroxyacetone phosphate.
  • GPO glycerol phosphate oxidase
  • H2O2 reacts with 4-aminophenazone and N,N-bis(4-sulfobutyl)-3,5- dimethylaniline, disodium salt (MADB) in the presence of peroxidase (POD) to produce a chromophore, which is read at 660/800nm.
  • MADB N,N-bis(4-sulfobutyl)-3,5- dimethylaniline, disodium salt
  • POD peroxidase
  • the increase in absorbance at 660/800 nm is proportional to the triglyceride content of the sample.
  • LDL-cholesterol from Beckman Coulter Nederland B.V. (Woerden, Netherlands, Datasheet from 08- 2009, coded BAOSR6x96.01 ; catalogue No. OSR6196) intended for the quantitative determination of LDL-Cholesterol concentrations in human serum, EDTA, or heparinized plasma samples on Beckman Coulter AU analyzers. Reference is made to the protocol used as provided by Beckman Coulter reagent kit.
  • NBD coronary heart disease
  • NCEP-ATP National Cholesterol Education Program Adult Treatment Panel
  • the system reagent of the kit (OSR6196 / OSR6296) was obtained from Beckman Coulter Nederland B.V. and consists of:
  • the Cholesterol reagents are ready for use. No preparation was required.
  • the LDL-Cholesterol test is a two reagent homogenous system.
  • the assay is comprised of two distinct phases. In phase one a unique detergent solubilizes cholesterol from non-LDL- lipoprotein particles. This cholesterol is consumed by cholesterol esterase, cholesterol oxidase, peroxidase and 4- aminoantipyrine to generate a colorless end product.
  • phase two a second detergent in the reagent releases cholesterol from the LDL - lipoproteins.
  • This cholesterol reacts with cholesterol esterase, cholesterol oxidase and a chromogen system to yield a blue color complex which can be measured bichromatically at 540/660nm.
  • the resulting increase in absorbance is directly proportional to the LDL-C concentration in the sample.
  • NCEP-ATP III classify LDL - Cholesterol levels as follows, when a human subject is considered:
  • HDL-cholesterol from Beckman Coulter Nederland B.V. (Woerden, Netherlands, Datasheet from 08- 2009, coded BAOSR6x95.01 ; catalogue No. OSR6195) intended for the quantitative determination of HDL-Cholesterol concentrations in human serum, EDTA, or heparinized plasma samples on Beckman Coulter AU analyzers. Reference is made to the protocol used as provided by Beckman Coulter reagent kit.
  • Measurements of cholesterol are used primarily in the diagnosis and treatment of disorders involving excess cholesterol in the blood, and lipid and lipoprotein metabolism disorders.
  • the system reagent of the kit (OSR6195 / OSR6295) was obtained from Beckman Coulter Nederland B.V. and consists of:
  • the Cholesterol reagents are ready for use. No preparation was required.
  • the HDL-Cholesterol test is a two reagent homogenous system for the selective measurement of serum or plasma HDL-Cholesterol in the presence of other lipoprotein particles.
  • the assay is comprised of two distinct phases. In phase one, free cholesterol in non-HDL-lipoproteins is solubilized and consumed by cholesterol oxidase, peroxidase, and DSBmT to generate a colorless end product.
  • phase two a unique detergent selectively solubilizes HDL- lipoproteins.
  • the HDL cholesterol is released for reaction with cholesterol esterase, cholesterol oxidase and a chromogen system to yield a blue color complex which can be measured bichromatically at 600/700nm.
  • the resulting increase in absorbance is directly proportional to the HDL-C concentration in the sample.
  • NCEP ATP 2 The guidelines (NCEP ATP 2) classify HDL- C levels as follows, when a human subject is considered:
  • Rhodospirillum rubrum bacteria 25 g, were purchased from Algosource Technologies (Saint- Nazaire, France) and stored at 4°C until use.
  • mice Eight-week old male mice (21 -27 g) were of the C57BL/6J strain that is well known for their high cholesterol level upon exposure to a high fat (a so-called Western) diet. The mice were obtained from Charles River Laboratories (France).
  • mice The feed for the mice was obtained from Altromin Spezialfutter GmbH (Lage, Germany). Hydrogels were obtained from ClearH20 (Westbrook, USA) and contained approximately 65 gram of 97% water in a gel.
  • the 65 g hydrogel was heated for 30 minutes at 70°C and then 1 .5 g control diet (in the pretest diet and in the diet for the control group), or 1 .5 g dried bacteria (the diet for the treated group) was gently mixed into the liquefied hydrogel with a spatula. Subsequently, 13.5 g high fat diet was also mixed into the hydrogel. The mixing was continued until a smooth homogenous product was obtained.
  • the hydrogel was weighed before and after mixing with the diets and also just prior to placement into the animal cages. The hydrogel-diets were stored in the dark at 4°C for a maximum of 3 days.
  • mice were placed in cages with litter underneath a mesh. Enrichment was provided in the form of a polymer shelter and a piece of paper towel. The mice were provided the hydrogel-diet cups as their only source of food and water.
  • mice Upon reception from the supplier the mice were quarantined together for 7 days. After individually housing, the mice were provided the feed-hydrogel mix in cups containing 60 gr hydrogel, 13.5 gr high fat diet and 1 .5 g control diet for 7 days. The feed-hydrogel product was refreshed 3 times over that period.
  • mice and the remaining hydrogel-diet cup were weighed to assess the effect of the two feeds on the mice weight development and their food consumption.
  • mice were weighed and anaesthetized with 0.05 ml of 200 mg/ml Nembutal (Kela, Lot 26344A13). After sufficient sedation the chest cavity was opened, and blood was taken with a 24 G needle on a 1 ml syringe (both heparin-treated) and stored in a 2 ml Vacutainer EDTA tube on ice.
  • the blood was centrifuged for 10 minutes at 1500 rpm at 4°C to separate the cells from the plasma.
  • the plasma was stored at 4°C overnight.
  • the cholesterol was measured in the plasma by company SynLab (Mons, Belgium). In this study subcontractor SynLab was chosen to perform the cholesterol analysis using an ELISA-based method that required less plasma for their measurements.
  • mice The goal was to examine the effect of administering Rhodospirillum rubrum (GEPEA/Algosolis) on the blood cholesterol level in mice.
  • GEPEA/Algosolis Rhodospirillum rubrum
  • mice The weekly food consumption by the mice showed no difference between the consumption of the groups in the second week or as compared to the first week (pre-test: all 8 mice).
  • LDL cholesterol levels of the treated mice that were administered R.rubrum cells were 39% lower than those of the control group with a p value of 0.044 (statistically significant; p-values below 0.05 when compared to the corresponding control parameter).
  • Non-HDL cholesterol levels (mostly comprised of LDL and VLDL) of the treated mice were 32% lower than those of the control group with a p value of 0.053.
  • feeding mice with feed comprising Rhodospirillum rubrum bacteria cells produced by GEPEA/Algosolis had a statistically significant reductive effect on the total plasma cholesterol levels (-6%) and on the LDL cholesterol levels (-39%) of C57BL/6J mice placed on a high fat diet.
  • the species of bacterium is from the genus Rhodospirillum, or is a mixture of different Rhodospirillum spp. selected for example from Rhodospirillum rubrum, Rhodospirillum centenum, Rhodospirillum photometricum, Rhodospirillum oryzae, Rhodospirillum sulfurexigens, Rhodospirillum salexigens, Rhodospirillum salinarum, Rhodospirillum sodomense, and Rhodospirillum mur.
  • the Rhodospirillum is Rhodospirillum rubrum.
  • composition comprising carotenoids, bacteriopheophytins and quinones on plasma LDL-cholesterol levels
  • Rhodospirillum rubrum strain S1 H was stored in liquid nitrogen in a 10% w/w sucrose- 0.85% w/w saline solution. To regrow the strain, the cells were taken out the liquid nitrogen and thawed for 30 minutes at room temperature. Cells were streaked on a sistrom succinate agar plate and a rich Luria Bertani (LB) medium to grow colony forming units. The agar plates were incubated at 30°C in dark and aerobic conditions for up to 4 days.
  • LB Luria Bertani
  • 30 g of bacterial pellet of Rhodospirillum rubrum strain S1 H cells are mixed using 440 ml of a biphasic mixture of petroleum ether (boiling point 60-80°C) and methanolic saline during 2 h at room temperature (biphasic mixture: 220 ml containing 20 ml of NaCI 0.3% by mass and 200 ml of methanol + 220 ml of the petroleum ether (boiling point 60-80°C)). After centrifugation at 5000 RPM during 20 minutes at room temperature, the upper phase (the petroleum ether phase) was removed and stored at room temperature.
  • Lower phase (the methanolic saline solution) was optionally submitted to a second extraction with an additional 220 ml of petroleum ether for 2 h at room temperature.
  • the second upper phase was mixed with the first one and dried using a rotavapor system.
  • the resulting viscous liquid was named“FRACTION 1.1”, or“fl .1 or“Extract 1.1” in Figure 3.
  • This fl .1 is an example of a composition
  • a composition comprising geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4- spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10.
  • Matrix-assisted laser desorption ionization time-of-flight (MALDI-ToF) mass spectrum was recorded using a QToF Premier mass spectrometer equipped with a Nd:YAG laser, operating at 355 nm with a output frequency of 50 Hz. Time-of-flight mass analyses were performed in reflection mode at a resolution of about 10.000. Samples of fl .1 were analyzed using (DCTB)trans-2-[3-(4-tertbutylphenyl)- 2-methylprop-2-enylidene] malononitrile. This matrix was prepared as a 40 mg/mL solution in CHCI3. The matrix solution (1 mg/mL) was applied to a stainless steel target and air dried.
  • the samples were dissolved in THF and I microliter aliquots of this solution were applied onto the target area already bearing the matrix crystals and air dried.
  • the quadrupole rf-only mode
  • the quadrupole was set to pass ions from 200 to 2500 Th, and all ions were transmitted into the pusher region of the time-of-flight analyzer where they were mass analyzed with 1 s integration time.
  • Samples of fl.1 were diluted in 0.1 % formic acid in acetonitrile, centrifuged at room temperature during 5 min at 13.000 RPM and the supernatants were infused directly in the Mass spectrometer (flow rate : 89 microliter/hour) using nano-esi source.
  • the acquisition parameters were: ion source gas1 : 4; Curtain gas 15; ionspray Voltage floating 2.300, heater temperature 150°C; Polarity : positive; ToF mass range : 100-2.000.
  • the carotenoids rhodovibrin, 1 -hydroxyspirilloxanthin, 3,4-dehdyro-rhodopin, chloroxanthin, rhodopin, spirilloxanthin and 3,4-dihyd ro-spirilloxanthin were also identified in the fl.1 extract when applying nano-ESI Q-ToF analysis. Ten mI of the fraction 1.1 was dried in presence of matrix.
  • the pheophytins geranyl-geranyl bacteriopheophytin a and phytyl bacteriopheophytin a were also identified in the fl.1 extract when applying nano-ESI Q-ToF analysis. Ten mI of the fraction 1.1 was dried in presence of matrix.
  • the quinones ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10 were also identified in the fl.1 extract when applying nano-ESI Q-ToF analysis. Ten mI of the fraction 1.1 was dried in presence of matrix.
  • NanoESI MS spectra were obtained for the fraction 1.1 with a triple tof mass spectrometer (ABSCIEX) using acetonitrile 99%, 1 % HCOOC as organic solvent.
  • Q-tOF 5600 ABSCIEX
  • peaks were revealed at the following m/z values (approximate relative intensity in brackets): 647.6 (2.5); 651 .6 (3.3); 881 .5 (2); 927.5 (1).
  • the 1.1 fraction comprises all of geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10.
  • the inventors have herewith found an example of a composition comprising all of the above-mentioned carotenoids, bacteriopheophytins and quinones.
  • mice test for testing the influence of a diet comprising fl .1 with carotenoids, bacteriopheophytins and quinoneson plasma cholesterol level was performed at SCKOEN animalarium (BE) following 2 weeks acclimation of 40 C57BL/6 male mice. After initial weighing of the food and the mice, they were placed in individual ventilated cage. Food consumption was checked every day and hydrogel weighed every 2 days. Based on previously performed preliminary palatability tests, the bacterial extracts were resuspended in sunflower oil, and 5% regular sugar was added to the chow (Cafetaria-diet) to ensure high palatability.
  • SB SCKOEN animalarium
  • mice received the cafetaria Diet + sunflower oil replacing R. rubrum extract fl.1 with carotenoids, bacteriopheophytins and quinones ad libitum while the second group of mice received the cafetaria Diet + the control diet replacing R. rubrum extract fl.1 ad libitum.
  • feeding control feed or feed comprising fl.1 as a composition comprising geranyl- geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10 on cholesterol levels in plasma is detailed below.
  • mice End of mice test After 2 weeks of testing, the mice where weighted and euthanized using intraperitoneal pentobarbital injection prior to dissection. Whole-blood was removed in EDTA-tubes, centrifuged to obtain plasma and placed at 4°C for further analysis.
  • Figure 3 shows the results of the cholesterol-, HDL- and LDL analysis in the blood of the treated mice.
  • Extract 1.1 (which is Fraction 1.1 , fl.1 ) as an example of a composition comprising geranyl- geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10 has a significant effect on the plasma LDL- cholesterol concentration since the Extract 1.1 , as a composition comprising geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4- rhodopin, chloro
  • Total cholesterol in plasma was 4,39 pg/mI for the fl .1 group of mice compared to 4,46 pg/mI for the control group, HDL-cholesterol was 2,93 mg/ml for the fl.1 group and 2,72 mr/mI for the control group, and LDL-cholesterol was 0,36 mg/ml for the fl .1 group (p ⁇ 0.001) and 0,67 mg/ml for the control group, respectively.
  • the extract fl.1 as an example of a composition with geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4- rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10has a beneficial effect on lowering LDL-cholesterol level to a large extent, while at the same time keeping the HDL-cholesterol level essentially unaltered when the extract comprising geranyl-geranyl bacteriopheophytin
  • composition comprising the geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10, has an LDL-cholesterol lowering effect.
  • the extract fraction fl.1 of R. rubrum, with rhodovibrin, and obtained as described here above in Example 2, is submitted to analytical chromatography for defining suitable conditions for the separation and isolation of the rhodovibrin present in the fraction Fl.1 .
  • the choice of the chromatography support and approach e.g . a hydrophobic chromatography using silica and two organic solvents
  • rhodovibrin will be analyzed by mass spectrometry.
  • rhodovibrin is isolated from the fraction fl.1 by applying preparative (HPLC) chromatography based on hydrophobic column material.
  • HPLC is coupled to a mass spectrometer for analysis of the fractions comprising the carotenoid.
  • the carotenoid is separated from the remainder of fraction fl.1 to an extent as close as possible to homogeneity.
  • the isolated rhodovibrin obtained with preparative chromatography is tested in a mice activity test performed substantially as described here above in Example 1 and Example 2. A minimum of 5 mice is included in each group.
  • a positive control group of mice consisting of mice fed with active extract fl.1 , is included in the test in order to be able to evaluate quantitatively the cholesterol lowering effect of the carotenoid compound. See also Example 1 and 2 for the LDL-cholesterol lowering effect in plasma of mice, of the fraction 1.1 of R.rubrum.
  • the isolated carotenoid rhodovibrin with cholesterol-lowering activity is further characterized by applying methods including for example mass spectrometry, UV spectroscopy, Infrared spectroscopy, NMR spectroscopy.
  • the extract fraction fl.1 of R.rubrum, with 1 -hydroxy-spirilloxanthin, and obtained as described here above in Example 2, is submitted to analytical chromatography for defining suitable conditions for the separation and isolation of the 1 -hydroxy-spirilloxanthin present in the fraction Fl.1 .
  • the choice of the chromatography support and approach e.g. a hydrophobic chromatography using silica and two organic solvents
  • 1 -hydroxy-spirilloxanthin will be analyzed by mass spectrometry.
  • 1 -hydroxy-spirilloxanthin is isolated from the fraction fl.1 by applying preparative (HPLC) chromatography based on hydrophobic column material.
  • the HPLC is coupled to a mass spectrometer for analysis of the fractions comprising the carotenoid.
  • the carotenoid is separated from the remainder of fraction fl .1 to an extent as close as possible to homogeneity.
  • the isolated 1 -hydroxy-spirilloxanthin obtained with preparative chromatography is tested in a mice activity test performed substantially as described here above in Example 1 and Example 2. A minimum of 5 mice is included in each group.
  • a positive control group of mice consisting of mice fed with active extract fl.1 , is included in the test in order to be able to evaluate quantitatively the cholesterol lowering effect of the carotenoid compound. See also Example 1 and 2 for the LDL-cholesterol lowering effect in plasma of mice, of the fraction 1.1 of R.rubrum.
  • the isolated carotenoid 1 -hydroxy-spirilloxanthin with cholesterol-lowering activity is further characterized by applying methods including for example mass spectrometry, UV spectroscopy, Infrared spectroscopy, NMR spectroscopy.
  • the extract fraction fl.1 of R.rubrum, with 3,4-dehydro-rhodopin, and obtained as described here above in Example 2, is submitted to analytical chromatography for defining suitable conditions for the separation and isolation of the 3,4-dehydro-rhodopin present in the fraction Fl.1 .
  • the choice of the chromatography support and approach e.g. a hydrophobic chromatography using silica and two organic solvents
  • 3,4-dehydro-rhodopin will be analyzed by mass spectrometry.
  • 3,4-dehydro-rhodopin is isolated from the fraction fl.1 by applying preparative (HPLC) chromatography based on hydrophobic column material.
  • the HPLC is coupled to a mass spectrometer for analysis of the fractions comprising the carotenoid.
  • the carotenoid is separated from the remainder of fraction fl.1 to an extent as close as possible to homogeneity.
  • the isolated 3,4-dehydro-rhodopin obtained with preparative chromatography is tested in a mice activity test performed substantially as described here above in Example 1 and Example 2. A minimum of 5 mice is included in each group.
  • a positive control group of mice consisting of mice fed with active extract fl.1 , is included in the test in order to be able to evaluate quantitatively the cholesterol lowering effect of the carotenoid compound. See also Example 1 and 2 for the LDL- cholesterol lowering effect in plasma of mice, of the fraction 1.1 of R.rubrum.
  • the isolated carotenoid 3,4-dehydro-rhodopin with cholesterol-lowering activity is further characterized by applying methods including for example mass spectrometry, UV spectroscopy, Infrared spectroscopy, NMR spectroscopy.
  • the extract fraction fl.1 of R.rubrum, with chloroxanthin, and obtained as described here above in Example 2, is submitted to analytical chromatography for defining suitable conditions for the separation and isolation of the chloroxanthin present in the fraction Fl.1 .
  • the choice of the chromatography support and approach e.g. a hydrophobic chromatography using silica and two organic solvents
  • the analytically separated chloroxanthin will be analyzed by mass spectrometry.
  • chloroxanthin is isolated from the fraction fl .1 by applying preparative (HPLC) chromatography based on hydrophobic column material.
  • HPLC preparative chromatography
  • the HPLC is coupled to a mass spectrometer for analysis of the fractions comprising the carotenoid.
  • the carotenoid is separated from the remainder of fraction fl.1 to an extent as close as possible to homogeneity.
  • the isolated chloroxanthin obtained with preparative chromatography is tested in a mice activity test performed substantially as described here above in Example 1 and Example 2. A minimum of 5 mice is included in each group.
  • a positive control group of mice consisting of mice fed with active extract fl .1 , is included in the test in order to be able to evaluate quantitatively the cholesterol lowering effect of the carotenoid compound. See also Example 1 and 2 for the LDL-cholesterol lowering effect in plasma of mice, of the fraction 1.1 of R.rubrum.
  • the isolated carotenoid chloroxanthin with cholesterol-lowering activity is further characterized by applying methods including for example mass spectrometry, UV spectroscopy, Infrared spectroscopy, NMR spectroscopy.
  • the extract fraction fl .1 of R.rubrum, with rhodopin, and obtained as described here above in Example 2, is submitted to analytical chromatography for defining suitable conditions for the separation and isolation of the rhodopin present in the fraction Fl.1 .
  • the choice of the chromatography support and approach e.g. a hydrophobic chromatography using silica and two organic solvents
  • rhodopin will be analyzed by mass spectrometry.
  • rhodopin is isolated from the fraction fl.1 by applying preparative (HPLC) chromatography based on hydrophobic column material.
  • HPLC is coupled to a mass spectrometer for analysis of the fractions comprising the carotenoid.
  • the carotenoid is separated from the remainder of fraction fl.1 to an extent as close as possible to homogeneity.
  • the isolated rhodopin obtained with preparative chromatography is tested in a mice activity test performed substantially as described here above in Example 1 and Example 2. A minimum of 5 mice is included in each group.
  • a positive control group of mice consisting of mice fed with active extract fl.1 , is included in the test in order to be able to evaluate quantitatively the cholesterol lowering effect of the carotenoid compound. See also Example 1 and 2 for the LDL-cholesterol lowering effect in plasma of mice, of the fraction 1.1 of R.rubrum.
  • the isolated carotenoid rhodopin with cholesterol-lowering activity is further characterized by applying methods including for example mass spectrometry, UV spectroscopy, Infrared spectroscopy, NMR spectroscopy.
  • the extract fraction fl.1 of R.rubrum, with spirilloxanthin, and obtained as described here above in Example 2, is submitted to analytical chromatography for defining suitable conditions for the separation and isolation of the spirilloxanthin present in the fraction FI.1 .
  • the choice ofthe chromatography support and approach e.g. a hydrophobic chromatography using silica and two organic solvents
  • spirilloxanthin will be analyzed by mass spectrometry.
  • spirilloxanthin is isolated from the fraction fl .1 by applying preparative (HPLC) chromatography based on hydrophobic column material.
  • HPLC is coupled to a mass spectrometer for analysis of the fractions comprising the carotenoid.
  • the carotenoid is separated from the remainder of fraction fl.1 to an extent as close as possible to homogeneity.
  • the isolated spirilloxanthin obtained with preparative chromatography is tested in a mice activity test performed substantially as described here above in Example 1 and Example 2. A minimum of 5 mice is included in each group.
  • a positive control group of mice consisting of mice fed with active extract fl .1 , is included in the test in order to be able to evaluate quantitatively the cholesterol lowering effect of the carotenoid compound. See also Example 1 and 2 for the LDL-cholesterol lowering effect in plasma of mice, of the fraction 1.1 of R.rubrum.
  • the isolated carotenoid spirilloxanthin with cholesterol-lowering activity is further characterized by applying methods including for example mass spectrometry, UV spectroscopy, Infrared spectroscopy, NMR spectroscopy.
  • the extract fraction fl .1 of R.rubrum, with 3,4-dihydro-spirilloxanthin, and obtained as described here above in Example 2, is submitted to analytical chromatography for defining suitable conditions for the separation and isolation of the 3,4-dihydro-spirilloxanthin present in the fraction Fl.1 .
  • the choice of the chromatography support and approach e.g. a hydrophobic chromatography using silica and two organic solvents
  • 3,4-dihydro- spirilloxanthin will be analyzed by mass spectrometry.
  • 3,4-dihydro-spirilloxanthin is isolated from the fraction fl.1 by applying preparative (HPLC) chromatography based on hydrophobic column material.
  • the HPLC is coupled to a mass spectrometer for analysis of the fractions comprising the carotenoid.
  • the carotenoid is separated from the remainder of fraction fl.1 to an extent as close as possible to homogeneity.
  • mice activity test performed substantially as described here above in Example 1 and Example 2.
  • a minimum of 5 mice is included in each group.
  • a positive control group of mice, consisting of mice fed with active extract fl.1 is included in the test in order to be able to evaluate quantitatively the cholesterol lowering effect of the carotenoid compound. See also Example 1 and 2 for the LDL-cholesterol lowering effect in plasma of mice, of the fraction 1.1 of R.rubrum.
  • the isolated carotenoid 3,4-dihydro-spirilloxanthin with cholesterol-lowering activity is further characterized by applying methods including for example mass spectrometry, UV spectroscopy, Infrared spectroscopy, NMR spectroscopy.
  • the extract fraction fl.1 of R.rubrum, with geranyl-geranyl bacteriopheophytin a, and obtained as described here above in Example 2, is submitted to analytical chromatography for defining suitable conditions for the separation and isolation of the geranyl-geranyl bacteriopheophytin a present in the fraction Fl.1 .
  • the choice of the chromatography support and approach e.g. a hydrophobic chromatography using silica and two organic solvents
  • geranyl-geranyl bacteriopheophytin a is isolated from the fraction fl.1 by applying preparative (HPLC) chromatography based on hydrophobic column material.
  • HPLC preparative chromatography
  • the HPLC is coupled to a mass spectrometer for analysis of the fractions comprising the pheophytin.
  • the pheophytin is separated from the remainder of fraction fl.1 to an extent as close as possible to homogeneity.
  • the isolated geranyl-geranyl bacteriopheophytin a obtained with preparative chromatography is tested in a mice activity test performed substantially as described here above in Example 1 and Example 2. A minimum of 5 mice is included in each group. A positive control group of mice, consisting of mice fed with active extract fl.1 , is included in the test in order to be able to evaluate quantitatively the cholesterol lowering effect of the pheophytin compound. See also Example 1 and 2 for the LDL-cholesterol lowering effect in plasma of mice, of the fraction 1.1 of R.rubrum.
  • the isolated pheophytin geranyl-geranyl bacteriopheophytin a with cholesterol-lowering activity is further characterized by applying methods including for example mass spectrometry, UV spectroscopy, Infrared spectroscopy, NMR spectroscopy.
  • the retention time and mass of phytyl bacteriopheophytin a are depicted in Table 1 of Bona-Lovasz et al. (2013), and these are 26,75 minutes and 888,5 g/mol respectively.
  • the isolated phytyl bacteriopheophytin a obtained according to the method of Bona-Lovasz et al. (2013) is tested in a mice activity test performed substantially as described here above in Example 1 and Example 2. A minimum of 5 mice is included in each group.
  • a positive control group of mice, consisting of mice fed with active extract fl.1 is included in the test in order to be able to evaluate quantitatively the cholesterol lowering effect of the pheophytin compound.
  • Negative control is a group of mice fed regular diet omitting the pheophytin and omitting the R.rubrum fraction.
  • the extract fraction fl.1 of R.rubrum, with ubiquinol-10, and obtained as described here above in Example 2, is submitted to analytical chromatography for defining suitable conditions for the separation and isolation of the ubiquinol-10 present in the fraction Fl.1 .
  • the choice of the chromatography support and approach e.g . a hydrophobic chromatography using silica and two organic solvents
  • the analytically separated ubiquinol-10 will be analyzed by mass spectrometry.
  • ubiquinol-10 is isolated from the fraction fl.1 by applying preparative (HPLC) chromatography based on hydrophobic column material.
  • the HPLC is coupled to a mass spectrometer for analysis of the fractions comprising the ubiquinol.
  • the ubiquinol is separated from the remainder of fraction fl.1 to an extent as close as possible to homogeneity.
  • the isolated ubiquinol-10 obtained with preparative chromatography is tested in a mice activity test performed substantially as described here above in Example 1 and Example 2. A minimum of 5 mice is included in each group.
  • a positive control group of mice consisting of mice fed with active extract fl.1 , is included in the test in order to be able to evaluate quantitatively the cholesterol lowering effect of the ubiquinol compound. See also Example 1 and 2 for the LDL-cholesterol lowering effect in plasma of mice, of the fraction 1.1 of R.rubrum.
  • the isolated ubiquinol ubiquinol-10 with cholesterol-lowering activity is further characterized by applying methods including for example mass spectrometry, UV spectroscopy, Infrared spectroscopy, NMR spectroscopy.
  • the extract fraction fl.1 of R.rubrum, with ubiquinone-9, and obtained as described here above in Example 2, is submitted to analytical chromatography for defining suitable conditions for the separation and isolation of the ubiquinone-9 present in the fraction Fl.1 .
  • the choice of the chromatography support and approach e.g. a hydrophobic chromatography using silica and two organic solvents
  • the analytically separated ubiquinone-9 will be analyzed by mass spectrometry.
  • ubiquinone-9 is isolated from the fraction fl.1 by applying preparative (HPLC) chromatography based on hydrophobic column material.
  • HPLC preparative chromatography
  • the HPLC is coupled to a mass spectrometer for analysis of the fractions comprising the ubiquinone.
  • the ubiquinone is separated from the remainder of fraction fl.1 to an extent as close as possible to homogeneity.
  • mice activity test performed substantially as described here above in Example 1 and Example 2.
  • a minimum of 5 mice is included in each group.
  • a positive control group of mice, consisting of mice fed with active extract fl.1 is included in the test in order to be able to evaluate quantitatively the cholesterol lowering effect of the ubiquinone compound. See also Example 1 and 2 for the LDL-cholesterol lowering effect in plasma of mice, of the fraction 1.1 of R.rubrum.
  • the isolated ubiquinone ubiquinone-9 with cholesterol-lowering activity is further characterized by applying methods including for example mass spectrometry, UV spectroscopy, Infrared spectroscopy, NMR spectroscopy.
  • the extract fraction fl.1 of R.rubrum, with ubiquinone-10, and obtained as described here above in Example 2, is submitted to analytical chromatography for defining suitable conditions for the separation and isolation of the ubiquinone-10 present in the fraction Fl.1 .
  • the choice of the chromatography support and approach e.g. a hydrophobic chromatography using silica and two organic solvents
  • ubiquinone-10 isolation See Example 4, here below, referring to Bona-Lovasz et al. (2013), page 925, last paragraph - page 926, first paragraph).
  • the analytically separated ubiquinone-10 will be analyzed by mass spectrometry.
  • ubiquinone-10 is isolated from the fraction fl.1 by applying preparative (HPLC) chromatography based on hydrophobic column material.
  • HPLC is coupled to a mass spectrometer for analysis of the fractions comprising the ubiquinone.
  • the ubiquinone is separated from the remainder of fraction fl.1 to an extent as close as possible to homogeneity.
  • the isolated ubiquinone-10 obtained with preparative chromatography is tested in a mice activity test performed substantially as described here above in Example 1 and Example 2. A minimum of 5 mice is included in each group. A positive control group of mice, consisting of mice fed with active extract fl.1 , is included in the test in order to be able to evaluate quantitatively the cholesterol lowering effect of the ubiquinone compound. See also Example 1 and 2 for the LDL-cholesterol lowering effect in plasma of mice, of the fraction 1.1 of R.rubrum.
  • the isolated ubiquinone ubiquinone-10 with cholesterol-lowering activity is further characterized by applying methods including for example mass spectrometry, UV spectroscopy, Infrared spectroscopy, NMR spectroscopy.
  • the extract fraction fl.1 of R.rubrum, with rhodoquinone-10, and obtained as described here above in Example 2, is submitted to analytical chromatography for defining suitable conditions for the separation and isolation of the rhodoquinone-10 present in the fraction Fl.1 .
  • the choice of the chromatography support and approach e.g. a hydrophobic chromatography using silica and two organic solvents
  • rhodoquinone-10 will be analyzed by mass spectrometry.
  • rhodoquinone-10 is isolated from the fraction fl .1 by applying preparative (HPLC) chromatography based on hydrophobic column material.
  • the HPLC is coupled to a mass spectrometer for analysis of the fractions comprising the ubiquinone derivative.
  • the ubiquinone derivative is separated from the remainder of fraction fl.1 to an extent as close as possible to homogeneity.
  • the isolated rhodoquinone-10 obtained with preparative chromatography is tested in a mice activity test performed substantially as described here above in Example 1 and Example 2. A minimum of 5 mice is included in each group.
  • a positive control group of mice consisting of mice fed with active extract fl.1 , is included in the test in order to be able to evaluate quantitatively the cholesterol lowering effect of the ubiquinone derivative compound. See also Example 1 and 2 for the LDL- cholesterol lowering effect in plasma of mice, of the fraction 1.1 of R. rubrum.
  • the isolated ubiquinone derivative rhodoquinone-10 with cholesterol-lowering activity is further characterized by applying methods including for example mass spectrometry, UV spectroscopy, Infrared spectroscopy, NMR spectroscopy.
  • a composition comprising at least one, preferably at least two, preferably all of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4-rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10 can be made.
  • Phase separation was achieved by adding 1 ml of water to the mixture, followed by vortexing for 1 minute and centrifugation for 20 minutes. The upper supernatant phase was then used directly for separation of rhodovibrin using HPLC-MS.
  • HPLC a Spherisorb C18 column with a mobile phase consisting of acetone-water and a non-linear gradient of 50%-100% acetone, was applied (see: abstract on page 912 of Bona-Lovasz et al. (2013)). The retention time and mass of rhodovibrin are depicted in Table 1 of Bona-Lovasz et al. (2013), and these are 21 .43 minutes and 584.5 g/mol respectively.
  • the isolated rhodovibrin obtained according to the method of Bona-Lovasz et al. (2013) is tested in a mice activity test performed substantially as described here above in Example 1 and Example 2. A minimum of 5 mice is included in each group.
  • a positive control group of mice, consisting of mice fed with active extract fl.1 is included in the test in order to be able to evaluate quantitatively the cholesterol lowering effect of the carotenoid compound.
  • Negative control is a group of mice fed regular diet omitting the carotenoid and omitting the R.rubrum fraction.
  • Rhodovibrin isolated according to these procedures is subjected to plasma LDL-cholesterol level lowering tests, as herein described.
  • Phase separation was achieved by adding 1 ml of water to the mixture, followed by vortexing for 1 minute and centrifugation for 20 minutes. The upper supernatant phase was then used directly for separation of 1 -hydroxy-spirilloxanthin using HPLC-MS.
  • HPLC a Spherisorb C18 column with a mobile phase consisting of acetone-water and a non-linear gradient of 50%-100% acetone, was applied (see: abstract on page 912 of Bona-Lovasz et al. (2013)). The retention time and mass of 1 -hydroxy- spirilloxanthin are depicted in Table 1 of Bona-Lovasz et al. (2013), and these are 20,85 minutes and 582.5 g/mol, respectively.
  • the isolated 1 -hydroxy-spirilloxanthin obtained according to the method of Bona-Lovasz et al. (2013) is tested in a mice activity test performed substantially as described here above in Example 1 and Example 2. A minimum of 5 mice is included in each group. A positive control group of mice, consisting of mice fed with active extract fl.1 , is included in the test in order to be able to evaluate quantitatively the cholesterol lowering effect of the carotenoid compound. Negative control is a group of mice fed regular diet omitting the carotenoid and omitting the R.rubrum fraction.
  • Phase separation was achieved by adding 1 ml of water to the mixture, followed by vortexing for 1 minute and centrifugation for 20 minutes. The upper supernatant phase was then used directly for separation of 3,4-dehydro-rhodopin using HPLC-MS.
  • HPLC a Spherisorb C18 column with a mobile phase consisting of acetone-water and a non-linear gradient of 50%-100% acetone, was applied (see: abstract on page 912 of Bona-Lovasz et al. (2013)).
  • the retention time and mass of 3,4-dehydro- rhodopin are depicted in Table 1 of Bona-Lovasz et al. (2013), and these are 22,07 minutes and 552,5 g/mol respectively.
  • the isolated 3,4-dehydro-rhodopin obtained according to the method of Bona-Lovasz et al. (2013) is tested in a mice activity test performed substantially as described here above in Example 1 and Example 2. A minimum of 5 mice is included in each group. A positive control group of mice, consisting of mice fed with active extract fl .1 , is included in the test in order to be able to evaluate quantitatively the cholesterol lowering effect of the carotenoid compound. Negative control is a group of mice fed regular diet omitting the carotenoid and omitting the R.rubrum fraction.
  • Phase separation was achieved by adding 1 ml of water to the mixture, followed by vortexing for 1 minute and centrifugation for 20 minutes. The upper supernatant phase was then used directly for separation of chloroxanthin using HPLC-MS.
  • HPLC a Spherisorb C18 column with a mobile phase consisting of acetone-water and a non-linear gradient of 50%-100% acetone, was applied (see: abstract on page 912 of Bona-Lovasz et al. (2013)). The retention time and mass of chloroxanthin are depicted in Table 1 of Bona-Lovasz et al. (2013), and these are 24.61 minutes and 556.5 g/mol respectively.
  • the isolated chloroxanthin obtained according to the method of Bona-Lovasz et al. (2013) is tested in a mice activity test performed substantially as described here above in Example 1 and Example 2. A minimum of 5 mice is included in each group.
  • a positive control group of mice, consisting of mice fed with active extract fl.1 is included in the test in order to be able to evaluate quantitatively the cholesterol lowering effect of the carotenoid compound.
  • Negative control is a group of mice fed regular diet omitting the carotenoid and omitting the R.rubrum fraction.
  • chloroxanthin is also outlined in the Main List, entry No. 100 of the Carotenoids Handbook, edited by Britton et al. (2004). Chloroxanthin isolated according to these procedures is subjected to plasma LDL-cholesterol level lowering tests, as herein described.
  • the isolated rhodopin obtained according to the method of Bona-Lovasz et al. (2013) is tested in a mice activity test performed substantially as described here above in Example 1 and Example 2. A minimum of 5 mice is included in each group.
  • a positive control group of mice, consisting of mice fed with active extract fl.1 is included in the test in order to be able to evaluate quantitatively the cholesterol lowering effect of the carotenoid compound.
  • Negative control is a group of mice fed regular diet omitting the carotenoid and omitting the R.rubrum fraction.
  • Rhodopin isolated according to these procedures is subjected to plasma LDL-cholesterol level lowering tests, as herein described.
  • Phase separation was achieved by adding 1 ml of water to the mixture, followed by vortexing for 1 minute and centrifugation for 20 minutes. The upper supernatant phase was then used directly for separation of spirilloxanthin using HPLC-MS.
  • HPLC a Spherisorb C18 column with a mobile phase consisting of acetone-water and a non-linear gradient of 50%-100% acetone, was applied (see: abstract on page 912 of Bona-Lovasz et al. (2013)). The retention time and mass of spirilloxanthin are depicted in Table 1 of Bona-Lovasz et al. (2013), and these are 23.87 minutes and 596.4 g/mol respectively.
  • the isolated spirilloxanthin obtained according to the method of Bona-Lovasz et al. (2013) is tested in a mice activity test performed substantially as described here above in Example 1 and Example 2. A minimum of 5 mice is included in each group.
  • a positive control group of mice, consisting of mice fed with active extract fl.1 is included in the test in order to be able to evaluate quantitatively the cholesterol lowering effect of the carotenoid compound.
  • Negative control is a group of mice fed regular diet omitting the carotenoid and omitting the R.rubrum fraction.
  • Phase separation was achieved by adding 1 ml of water to the mixture, followed by vortexing for 1 minute and centrifugation for 20 minutes. The upper supernatant phase was then used directly for separation of 3,4-dihydro-spirilloxanthin using HPLC-MS.
  • HPLC a Spherisorb C18 column with a mobile phase consisting of acetone-water and a non-linear gradient of 50%-100% acetone, was applied (see: abstract on page 912 of Bona-Lovasz et al. (2013)). The retention time and mass of 3,4-dihydro-spirilloxanthin are depicted in Table 1 of Bona-Lovasz et al. (2013), and these are 24.46 minutes and 598.5 g/mol respectively.
  • mice activity test performed substantially as described here above in Example 1 and Example 2.
  • a minimum of 5 mice is included in each group.
  • a positive control group of mice consisting of mice fed with active extract fl .1 , is included in the test in order to be able to evaluate quantitatively the cholesterol lowering effect of the carotenoid compound.
  • Negative control is a group of mice fed regular diet omitting the carotenoid and omitting the R.rubrum fraction.
  • 3,4-dihydro-spirilloxanthin is also outlined in the Main List, entry No. 168 of the Carotenoids Handbook, edited by Britton et al. (2004). 3,4-dihydro-spirilloxanthin isolated according to these procedures is subjected to plasma LDL-cholesterol level lowering tests, as herein described.
  • the isolated geranyl-geranyl bacteriopheophytin a obtained according to the method of Bona- Lovasz et al. (2013) is tested in a mice activity test performed substantially as described here above in Example 1 and Example 2. A minimum of 5 mice is included in each group. A positive control group of mice, consisting of mice fed with active extract fl.1 , is included in the test in order to be able to evaluate quantitatively the cholesterol lowering effect of the pheophytin compound. Negative control is a group of mice fed regular diet omitting the pheophytin and omitting the R.rubrum fraction.
  • the resulting suspension was diluted with 150 ml of ether and filtered through a slurry of 12 g of Celite in ether using a glass suction filter G3, such that a clear extract was obtained.
  • the filtered methanol extract was saved.
  • the filter residue was washed 3 times with 30 ml of ether and the resulting combined filtrate was diluted with 150 ml pentane.
  • the separated water/methanol phase was shaken with 60 ml ether/pentane (1 :1) and 5 ml of NaCI solution was added. Both organic solutions were first washed with 30 ml buffer with pH 7 and then with 30 ml of NaCI solution.
  • This crude extract contains geranyl-geranyl bacteriopheophytin a and geranyl-geranyl bacteriochlorophyll a.
  • the crude extract is dried over Na 2 SC> 4 after addition of 20 ml methanol in a rotary evaporator at 30°. Five ml of methanol is then poured overthe crude product four times, while the solution is pipetted off, filtered and evaporated after every round. The residue that does not dissolve in methanol was dissolved in ether. After addition of methanol, the solvent was evaporated.
  • the geranyl-geranyl bacteriopheophytin a was obtained. This was dissolved in 2 ml methylene chloride, then diluted with 2 ml methanol, and left for 16 hr at room temperature in a not completely closed container. The precipitate was filtered and obtained, and was washed with 8 ml methanol, and then dried (yield was 1 10 mg).
  • the geranyl-geranyl bacteriopheophytin a was purified by two cycles of: dissolving the geranyl-geranyl bacteriopheophytin a in methylene chloride; diluting with methanol; evaporating the solvents with a rotary evaporator; washing the solid geranyl-geranyl bacteriopheophytin a with 6 ml methanol. Isolating the bacteriopheophytin phytyl bacteriopheophytin a from R. rubrum bacteria cells
  • the isolated phytyl bacteriopheophytin a obtained according to the method of Bona-Lovasz et al. (2013) is tested in a mice activity test performed substantially as described here above in Example 1 and Example 2. A minimum of 5 mice is included in each group. A positive control group of mice, consisting of mice fed with active extract fl .1 , is included in the test in order to be able to evaluate quantitatively the cholesterol lowering effect of the pheophytin compound. Negative control is a group of mice fed regular diet omitting the pheophytin and omitting the R.rubrum fraction.
  • Phase separation was achieved by adding 1 ml of water to the mixture, followed by vortexing for 1 minute and centrifugation for 20 minutes. The upper supernatant phase was then used directly for separation of ubiquinol-10 using HPLC-MS.
  • HPLC a Spherisorb C18 column with a mobile phase consisting of acetone-water and a non-linear gradient of 50%-100% acetone, was applied (see: abstract on page 912 of Bona-Lovasz et al. (2013)). The retention time and mass of ubiquinol-10 are depicted in Table 1 of Bona-Lovasz et al. (2013), and these are 31 .08 minutes and 846.7 g/mol respectively.
  • the isolated ubiquinol-10 obtained according to the method of Bona-Lovasz et al. (2013) is tested in a mice activity test performed substantially as described here above in Example 1 and Example 2. A minimum of 5 mice is included in each group. A positive control group of mice, consisting of mice fed with active extract fl.1 , is included in the test in order to be able to evaluate quantitatively the cholesterol lowering effect of the ubiquinol compound. Negative control is a group of mice fed regular diet omitting the ubiquinol and omitting the R.rubrum fraction.
  • Phase separation was achieved by adding 1 ml of water to the mixture, followed by vortexing for 1 minute and centrifugation for 20 minutes. The upper supernatant phase was then used directly for separation of ubiquinone-9 using HPLC-MS.
  • HPLC a Spherisorb C18 column with a mobile phase consisting of acetone-water and a non-linear gradient of 50%-100% acetone, was applied (see: abstract on page 912 of Bona-Lovasz et al. (2013)).
  • the retention time and mass of ubiquinone-9 are depicted in Table 1 of Bcma-Lovasz et al. (2013), and these are 31 .43 minutes and 794.6 g/mol respectively.
  • the isolated ubiquinone-9 obtained according to the method of Bona-Lovasz et al. (2013) is tested in a mice activity test performed substantially as described here above in Example 1 and Example 2. A minimum of 5 mice is included in each group. A positive control group of mice, consisting of mice fed with active extract fl.1 , is included in the test in order to be able to evaluate quantitatively the cholesterol lowering effect of the ubiquinone compound. Negative control is a group of mice fed regular diet omitting the ubiquinone and omitting the R.rubrum fraction.
  • Phase separation was achieved by adding 1 ml of water to the mixture, followed by vortexing for 1 minute and centrifugation for 20 minutes. The upper supernatant phase was then used directly for separation of ubiquinone-10 using HPLC-MS.
  • HPLC a Spherisorb C18 column with a mobile phase consisting of acetone-water and a non-linear gradient of 50%-100% acetone, was applied (see: abstract on page 912 of Bona-Lovasz et al. (2013)).
  • the retention time and mass of ubiquinone-10 are depicted in Table 1 of Bcma-Lovasz et al. (2013), and these are 32.81 minutes and 862.7 g/mol, respectively.
  • the isolated ubiquinone-10 obtained according to the method of Bona-Lovasz et al. (2013) is tested in a mice activity test performed substantially as described here above in Example 1 and Example 2. A minimum of 5 mice is included in each group. A positive control group of mice, consisting of mice fed with active extract fl.1 , is included in the test in order to be able to evaluate quantitatively the cholesterol lowering effect of the ubiquinone compound. Negative control is a group of mice fed regular diet omitting the ubiquinone and omitting the R.rubrum fraction.
  • the cells were extracted with 4 litre portions of boiling absolute ethanol and evaporated under vacuum to dryness yielding 102.9 g of tarry residue.
  • the hexane extract was purified by precipitation of impurities and chromatography to give 249 mg of yellow oil containing the C0Q10 (66%).
  • the ethanol extract was purified in the same manner and yielded 600 mg of material containing C0Q10.
  • Phase separation was achieved by adding 1 ml of water to the mixture, followed by vortexing for 1 minute and centrifugation for 20 minutes. The upper supernatant phase was then used directly for separation of rhodoquinone-10 using HPLC-MS.
  • HPLC a Spherisorb C18 column with a mobile phase consisting of acetone-water and a non-linear gradient of 50%-100% acetone, was applied (see: abstract on page 912 of Bona-Lovasz et at. (2013)).
  • the retention time and mass of rhodoquinone-10 are depicted in Table 1 of Bona-Lovasz et al. (2013), and these are 32.24 minutes and 847.7 g/mol, respectively.
  • the isolated rhodoquinone-10 obtained according to the method of Bona-Lovasz et al. (2013) is tested in a mice activity test performed substantially as described here above in Example 1 and Example 2. A minimum of 5 mice is included in each group.
  • a positive control group of mice, consisting of mice fed with active extract fl .1 is included in the test in order to be able to evaluate quantitatively the cholesterol lowering effect of the ubiquinone derivative compound.
  • Negative control is a group of mice fed regular diet omitting the ubiquinone derivative and omitting the R.rubrum fraction.
  • Rhodoquinone-10 Isolation of rhodoquinone-10 from a natural source is performed using Rhodospirillum rubrum and is detailed by Parson & Rudney (1965; p 1856, left column, 2 nd paragraph). Reference is made to the protocol outlined in Parson & Rudney (1965), and in brief, first, R. rubrum is grown and the cells harvested, centrifuged, washed with water and lyophilized. The lyophilized cells were then extracted with three 70 ml portions of ether-ethanol (3:1). The extract was evaporated, redissolved in 25 ml of petroleum-ether and filtered. This petroleum-ether solution was then extracted four times with 5 ml portions of 95% methanol. Rhodoquinone-10 was eluted with 16% ether in petroleum ether.
  • Surmatis et al. (1966) provided rhodovibrin by reacting crocetindialdehyde with (7-hydroxy- 3,7-dimethyl-2-octenyl)triphenylphosphonium bromid in a benzene-methanol solution. After purification by chromatography on grade I alumina, recrystallization was carried out with benzene to yield 42.3% of 1 -hydroxy-1 ,2-dihydroapo-3-lycopenal as a dark red crystalline solid.
  • the synthesized rhodovibrin obtained according to the method of Surmatis et al. (1966) is tested in a mice activity test performed substantially as described here above in Example 1 and Example 2. A minimum of 5 mice is included in each group.
  • a positive control group of mice, consisting of mice fed with active extract fl.1 is included in the test in order to be able to evaluate quantitatively the cholesterol lowering effect of the carotenoid compound.
  • Negative control is a group of mice fed regular diet omitting the carotenoid and omitting the R.rubrum fraction.
  • rhodovibrin The synthesis of rhodovibrin is also outlined in Carotenoids Volume 2 Synthesis (1996), page 142-143, page 297. The synthesis of rhodovibrin is also outlined in the Main List, entry No. 167 of the Carotenoids Handbook, edited by Britton et al. (2004). Rhodovibrin synthesized according to these procedures is subjected to plasma LDL-cholesterol level lowering tests, as herein described.
  • Lithium derivatives of methylpent-4-yn-2-ol and 2-methoxy-2-methylpent-4-yne reacted with methyl vinyl ketone to give the 2,6-dimethyloct-7-en-4ybe-2,6 diol and 7-methoxy-3,7-dimethyloct-1 -en-4-yn-3-ol, respectively. These were reduced with lithium aluminium hybride to give 6,6-dimethylocta-4,7-diene- 2,6-diol and 7-methoxy-3,7-dimethylocta-1 ,4-dien-3-ol.
  • mice activity test performed substantially as described here above in Example 1 and Example 2.
  • a minimum of 5 mice is included in each group.
  • a positive control group of mice consisting of mice fed with active extract fl .1 , is included in the test in order to be able to evaluate quantitatively the cholesterol lowering effect of the carotenoid compound.
  • Negative control is a group of mice fed regular diet omitting the carotenoid and omitting the R.rubrum fraction.
  • 3,4-dehydro-rhodopin is also outlined in Carotenoids Volume 2 Synthesis (1996), page 141 (compound 90).
  • the synthesis of 3,4-dehydro-rhodopin is also referred to in the Main List, entry No. 90 of the Carotenoids Handbook, edited by Britton et al. (2004).
  • 3,4-dehydro-rhodopin synthesized according to these procedures is subjected to plasma LDL-cholesterol level lowering tests, as herein described.
  • Barber et al. (1966) provided chloroxanthin following the following procedure. Sodium methoxide in methanol was added dropwise to a stirred solution of 7,8-dihydro-apo-8’-lycopenal and (7- hydrocy-3,7-dimethyloct-2-enyl)triphenylphosphonium bromide in dry methanol. The solution was stirred, diluted with water and extracted with benzene. The crude product was purified by repeated chromatography on alumina using benzene as eluant.
  • the synthesized chloroxanthin obtained according to the method of Barber et al. (1966) is tested in a mice activity test performed substantially as described here above in Example 1 and Example 2. A minimum of 5 mice is included in each group. A positive control group of mice, consisting of mice fed with active extract fl.1 , is included in the test in order to be able to evaluate quantitatively the cholesterol lowering effect of the carotenoid compound. Negative control is a group of mice fed regular diet omitting the carotenoid and omitting the R.rubrum fraction.
  • chloroxanthin (compound 100) is also outlined in Carotenoids Volume 2 Synthesis (1996), page 144-145. The synthesis of chloroxanthin is also outlined in the Main List, entry No. 100 of the Carotenoids Handbook, edited by Britton et al. (2004). Chloroxanthin synthesized according to these procedures is subjected to plasma LDL-cholesterol level lowering tests, as herein described.
  • the synthesized rhodopin obtained according to the method of Surmatis et al. (1966) is tested in a mice activity test performed substantially as described here above in Example 1 and Example 2. A minimum of 5 mice is included in each group.
  • a positive control group of mice, consisting of mice fed with active extract fl.1 is included in the test in order to be able to evaluate quantitatively the cholesterol lowering effect of the carotenoid compound.
  • Negative control is a group of mice fed regular diet omitting the carotenoid and omitting the R.rubmm fraction.
  • rhodopin The synthesis of rhodopin is also outlined in Carotenoids Volume 2 Synthesis (1996), page 144- 145. The synthesis of rhodopin is also outlined in the Main List, entry No. 93 of the Carotenoids Handbook, edited by Britton et al. (2004). Rhodopin synthesized according to these procedures is subjected to plasma LDL-cholesterol level lowering tests, as herein described.
  • Surmatis et al. (1963) prepared 6-methoxy-6-methyl-2-heptanone by stirring 6-methylhept- 5-en-2-one in a solution of sulfuric acid and methyl alcohol for 24 hrs at room temperature. Condensation of 6-methoxy-6-methyl-2-heptanone with ethyl bromoacetate by the Reformatsky reaction, followed by dehydration with phosphorus oxychloride in pyridine yielded 7-methoxy-3,7-dimethyl-2-octenoic acid ethyl ester.
  • Surmatis et al. (1963) describe a second method to chemically synthesize spirilloxanthin. Briefly summarized, Surmatis et al. (1963) outlined the ethynylation of 6-methoxy-6-methyl-2-heptanone with sodium acetylide in liquid ammonia to yield 7-methoxy-3,7-dimethyl-1 -octyn-3-ol. Selective hydrogenation of 7-methoxy-3,7-dimethyl-1 -octyn-3-ol resulted in 7-methoxy-3,7-dimethyl-1 -octen-3-ol.
  • Lithium derivatives of methylpent-4-yn-2-ol and 2-methoxy-2-methylpent-4-yne reacted with methyl vinyl ketone to give the 2,6-dimethyloct-7-en-4ybe-2,6 diol and 7-methoxy-3,7- dimethyloct-1 -en-4-yn-3-ol, respectively. These were reduced with lithium aluminium hybride to give 6,6- dimethylocta-4,7-diene-2,6-diol and 7-methoxy-3,7-dimethylocta-1 ,4-dien-3-ol.
  • the synthesized spirilloxanthin obtained according to the method(s) of Surmatis et al. (1963) and/or Schneider et al. (1967) is tested in a mice activity test performed substantially as described here above in Example 1 and Example 2. A minimum of 5 mice is included in each group. A positive control group of mice, consisting of mice fed with active extract fl.1 , is included in the test in order to be able to evaluate quantitatively the cholesterol lowering effect of the carotenoid compound. Negative control is a group of mice fed regular diet omitting the carotenoid and omitting the R.rubrum fraction.
  • spirilloxanthin is also referred to in Carotenoids Volume 2 Synthesis (1996), page 142- 143.
  • the synthesis of spirilloxanthin is also outlined in the Main List, entry No. 166 of the Carotenoids Handbook, edited by Britton et al. (2004). Spirilloxanthin synthesized according to these procedures is subjected to plasma LDL-cholesterol level lowering tests, as herein described.
  • C0Q10 is chemically synthesized as detailed by Ravada et al. (2009). Reference is made to the detailed protocol in Ravada et al. (2009), and in brief, the synthesis is carried out in nine steps and makes use of isoprenol as a pre-cursor.
  • the method starts with prenylation of 2,3-dimethoxy-5- methylhydroquinone using isoprenol in presence of a Lewis acid, followed by selective oxidation of the trans methyl group of the isoprenyl side chain and subsequent allylic bromination.
  • a chemical synthesis method for ubiquinone-10 is outlined by Moore & Folkers (1965). Details of the chemical synthesis of rhodoquinone-10 are outlined in Moore & Folkers (1965) and in brief, synthetic rhodoquinone-10 is synthesised from precursor Coenzyme Qio by ammonolysis. Treating C0Q10 with ammonium hydroxide in a solvent of diethyl ether-ethanol (1 :1) gives synthetic rhodoquinone-10 (See for further details: Moore & Folkers, 1965; p. 1410, left column, 1 st paragraph).
  • This Example has shown that the synthesis of several carotenoids and quinones is possible, to create a composition comprising at least one, preferably two, more preferably all of compounds geranyl-geranyl bacteriopheophytin a, phytyl derivative of bacteriopheophytin a, hydroxyspirilloxanthin, rhodovibrin, 3,4- rhodopin, chloroxanthin, rhodopin, spirilloxanthin, 3,4-spirilloxanthin, ubiquinol-10, ubiquinone-9, ubiquinone-10 and rhodoquinone-10.
  • R.rubrum fraction 1.1 was prepared as here above described for Example 2. Preparing R.rubrum fraction 1.1 from 60 g of biomass (bacterial cells) resulted in a yield of 8 ml (approx. 7 g) of oily petroleum ether extract Fraction 1.1 (after removal of the petroleum ether).
  • HPLC method was initially based on Indriatmokoa et at. (2015) Procedia Chemistry 14, 202-210 “Separation of Photosynthetic Pigments by High-Performance Liquid Chromatography: Comparison of Column Performance, Mobile Phase, and Temperature”.
  • the current column material was similar to the column described in this paper: a C18-type column material, but now functionalized to be more specific for aromatic compounds.
  • THF was used as the solvent for the R.rubrum extract Fraction 1.1 and a gradient elution of THF in isopropanol was used (flow was 0,5 ml/min.), as well as, alternatively, a gradient elution of THF in methanol (flow was 0,5 ml/min.).
  • the fraction 1.1 of R.rubrum dissolves very well in THF. Solutions of 1 mg/ml and 10 mg/ml were prepared. Samples of 50 mg fraction 1.1 are injectable in this semi-preparative HPLC approach. Elution of the compounds from the column resulted in two compound peaks (230 nm detection).
  • Coenzyme Q XX Isolation of coenzymes Q9 and Q10 from two basidiomycetes. Archives of biochemistry and biophysics. 90; 314-317.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Molecular Biology (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Diabetes (AREA)
  • Hematology (AREA)
  • Obesity (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

La présente invention concerne une composition (pharmaceutique) qui comprend de la géranyl-géranyl-bactériophéophytine a, un dérivé phytyl de la bactériophéophytine a, de l'hydroxy-spirilloxanthine, de la rhodovibrine, de la 3,4-rhodopine, de la chloroxanthine, de la rhodopine, de la spirilloxanthine, de la 3,4-spirilloxanthine, de l'ubiquinol-10, de l'ubiquinone-9, de l'ubiquinone-10 et de la rhodoquinone-10, et qui est destinée à être utilisée dans une méthode visant à abaisser le cholestérol LBD chez le patient qui en a besoin. De plus, l'invention concerne un aliment qui comprend un complément alimentaire, le complément alimentaire possédant des propriétés d'abaissement du cholestérol et comprenant les composés suivant : de la géranyl-géranyl-bactériophéophytine a, un dérivé phytyl de la bactériophéophytine a, de l'hydroxy-spirilloxanthine, de la rhodovibrine, de la 3,4-rhodopine, de la chloroxanthine, de la rhodopine, de la spirilloxanthine, de la 3,4-spirilloxanthine, de l'ubiquinol-10, de l'ubiquinone-9, de l'ubiquinone-10 et de la rhodoquinone-10. Pour la composition pharmaceutique, la composition et le complément alimentaire, la condition est que la composition (pharmaceutique) ne soit pas, et que les composés ne soient pas fournis comme étant, un extrait d'éther de pétrole de Rhodospirillum rubrum pouvant être obtenu par extraction de cellules de Rhodospirillum rubrum avec un mélange d'éther de pétrole à point d'ébullition compris entre 60 °C et 80 °C et de méthanol comportant du chlorure de sodium, l'extrait étant obtenu par extraction pendant 10 minutes à 48 heures à une température comprise entre 8 °C et 37 °C, tout en mélangeant les cellules avec le mélange, les cellules de Rhodospirillum rubrum étant extraites avec le mélange dans un rapport de volume compris entre 10 / 1 et 1 / 10 d'éther de pétrole à point d'ébullition compris entre 60 °C et 80 °C et de méthanol comportant du chlorure de sodium. De plus, la composition (pharmaceutique) et les composés ne comprennent pas ou ne sont pas fournis en tant que cellules entières de R. rubrum ou en tant que fraction de membrane de R. rubrum.
EP19829701.2A 2018-12-18 2019-12-17 Composition à utiliser dans une méthode visant à abaisser le taux plasmatique de cholestérol lbd Withdrawn EP3897677A1 (fr)

Applications Claiming Priority (14)

Application Number Priority Date Filing Date Title
NL2022227 2018-12-18
NL2022225 2018-12-18
NL2022229 2018-12-18
NL2022224 2018-12-18
NL2022233 2018-12-18
NL2022234 2018-12-18
NL2022232 2018-12-18
NL2022235 2018-12-18
NL2022231 2018-12-18
NL2022236 2018-12-18
NL2022230 2018-12-18
NL2022226 2018-12-18
NL2022228 2018-12-18
PCT/NL2019/050847 WO2020130813A1 (fr) 2018-12-18 2019-12-17 Composition à utiliser dans une méthode visant à abaisser le taux plasmatique de cholestérol lbd

Publications (1)

Publication Number Publication Date
EP3897677A1 true EP3897677A1 (fr) 2021-10-27

Family

ID=69063848

Family Applications (1)

Application Number Title Priority Date Filing Date
EP19829701.2A Withdrawn EP3897677A1 (fr) 2018-12-18 2019-12-17 Composition à utiliser dans une méthode visant à abaisser le taux plasmatique de cholestérol lbd

Country Status (2)

Country Link
EP (1) EP3897677A1 (fr)
WO (1) WO2020130813A1 (fr)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113143932B (zh) * 2021-04-15 2022-10-04 东莞市人民医院 依塞曲匹作为抗结直肠癌药物的制药用途
WO2023081725A1 (fr) * 2021-11-02 2023-05-11 Whitehead Institute For Biomedical Research Utilisations de rhodoquinone pour le traitement d'une maladie

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
NL1022153C2 (nl) 2002-12-12 2004-06-15 Tno Cholesterol verlagend preparaat, voedingssupplement en voedingsmiddel en werkwijzen voor de bereiding daarvan.
NL2000548C1 (nl) * 2007-03-19 2008-09-22 Ipstar B V Rhodospirillum en Phaeospirillum in diervoeder voor het verkrijgen van voedingsproducten met een verlaagd cholesterolgehalte.
JP2020524500A (ja) * 2017-06-15 2020-08-20 エズコル ビー.ヴイ. Ldl−コレステロールを低下させる細胞抽出物および食品サプリメント

Also Published As

Publication number Publication date
WO2020130813A1 (fr) 2020-06-25

Similar Documents

Publication Publication Date Title
Fki et al. Hypocholesterolemic effects of phenolic-rich extracts of Chemlali olive cultivar in rats fed a cholesterol-rich diet
Hayek et al. Reduced progression of atherosclerosis in apolipoprotein E–deficient mice following consumption of red wine, or its polyphenols quercetin or catechin, is associated with reduced susceptibility of LDL to oxidation and aggregation
Breithaupt et al. Plasma response to a single dose of dietary β-cryptoxanthin esters from papaya (Carica papaya L.) or non-esterified β-cryptoxanthin in adult human subjects: a comparative study
Jensen et al. All-rac-α-tocopherol acetate is a better vitamin E source than all-rac-α-tocopherol succinate for broilers
Gil-Ramírez et al. Modulation of cholesterol-related gene expression by ergosterol and ergosterol-enriched extracts obtained from Agaricus bisporus
Bodas et al. The liver antioxidant status of fattening lambs is improved by naringin dietary supplementation at 0.15% rates but not meat quality
KR20080110444A (ko) 안트로디아 캄포라타로부터 얻은 시클로헥세논 유형의 간보호 화합물
US20210275601A1 (en) Ldl-cholesterol-lowering cell extract and food supplement
CZ1199A3 (cs) Způsob vyhodnocování rizika kardiovaskulárních chorob a jiných poruch a prostředek založený na fytosterolu, vhodný pro prevenci a léčení kardiovaskulárních chorob a jiných poruch
EP3897677A1 (fr) Composition à utiliser dans une méthode visant à abaisser le taux plasmatique de cholestérol lbd
Lee et al. Grain sorghum whole kernel oil lowers plasma and liver cholesterol in male hamsters with minimal wax involvement
JP2013520444A (ja) 高コレステロール血症を処置するための薬剤及び機序
Fujita et al. Extract of black tea (pu‐ehr) inhibits postprandial rise in serum cholesterol in mice, and with long term use reduces serum cholesterol and low density lipoprotein levels and renal fat weight in rats
JPH09510446A (ja) ペリリルアルコール及びペリラ酸メチルエステルの使用
Hansen et al. Supplementation of laying-hen feed with annatto tocotrienols and impact of α-tocopherol on tocotrienol transfer to egg yolk
US5665359A (en) Method and compositions for lowering blood lipids
Ruocco et al. Molecular and metabolic effects of extra-virgin olive oil on the cardiovascular gene signature in rodents
WO2005063233A1 (fr) Composition pour la prevention et le traitement d'hepatome
NL2024271B1 (en) Rhodospirillum rubrum cells that lower plasma cholesterol while leaving other serum protein levels unaffected
NL2026903B1 (en) Rhodospirillum rubrum cells that lower plasma cholesterol while leaving other serum protein levels unaffected
EP4260849A1 (fr) Composé pour la réduction du tissu adipeux blanc et traitement du surpoids et de l'obésité
EP3142660B1 (fr) Composition comprenant du 7-hydroxymatairésinol
KR100302024B1 (ko) 혈중콜레스테롤저하용홍화씨추출물및그제조방법
KR20120001150A (ko) 명란젓 또는 명란을 유효성분으로 함유하는 비만 또는 고지혈증 및 동맥경화성 혈관계 질환의 예방 및 치료용 조성물
Keller et al. Nanoscale and customary non‐esterified sitosterols are equally enriched in different body compartments of the guinea pig

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: UNKNOWN

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

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

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

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

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20210713

AK Designated contracting states

Kind code of ref document: A1

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

DAV Request for validation of the european patent (deleted)
DAX Request for extension of the european patent (deleted)
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

17Q First examination report despatched

Effective date: 20220714

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: 20230125