US20010016590A1 - Use of hydroxymatairesinol for prevention of cancers, non-cancer, hormone dependent diseases and cardiovascular diseases by hydroxymatairesinol, and a pharmaceutical preparation, food additive and food product comprising hydroxymatairesinol - Google Patents

Use of hydroxymatairesinol for prevention of cancers, non-cancer, hormone dependent diseases and cardiovascular diseases by hydroxymatairesinol, and a pharmaceutical preparation, food additive and food product comprising hydroxymatairesinol Download PDF

Info

Publication number
US20010016590A1
US20010016590A1 US09/829,944 US82994401A US2001016590A1 US 20010016590 A1 US20010016590 A1 US 20010016590A1 US 82994401 A US82994401 A US 82994401A US 2001016590 A1 US2001016590 A1 US 2001016590A1
Authority
US
United States
Prior art keywords
cancer
hydroxymatairesinol
food
hmr
food product
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US09/829,944
Inventor
Markku Ahotupa
Christer Eckerman
Lauri Kangas
Sari Makela
Niina Saarinen
Risto Santti
Anni Warri
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.)
Individual
Original Assignee
Individual
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 Individual filed Critical Individual
Priority to US09/829,944 priority Critical patent/US20010016590A1/en
Publication of US20010016590A1 publication Critical patent/US20010016590A1/en
Priority to US09/972,850 priority patent/US6689809B2/en
Priority to US10/639,530 priority patent/US7005447B2/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/365Lactones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/34Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide
    • A61K31/341Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide not condensed with another ring, e.g. ranitidine, furosemide, bufetolol, muscarine
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23LFOODS, FOODSTUFFS, OR NON-ALCOHOLIC BEVERAGES, NOT COVERED BY SUBCLASSES A21D OR A23B-A23J; THEIR PREPARATION OR TREATMENT, e.g. COOKING, MODIFICATION OF NUTRITIVE QUALITIES, PHYSICAL TREATMENT; PRESERVATION OF FOODS OR FOODSTUFFS, IN GENERAL
    • A23L33/00Modifying nutritive qualities of foods; Dietetic products; Preparation or treatment thereof
    • A23L33/10Modifying nutritive qualities of foods; Dietetic products; Preparation or treatment thereof using additives
    • A23L33/105Plant extracts, their artificial duplicates or their derivatives
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/02Drugs for disorders of the urinary system of urine or of the urinary tract, e.g. urine acidifiers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/10Drugs for disorders of the urinary system of the bladder
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/08Drugs for genital or sexual disorders; Contraceptives for gonadal disorders or for enhancing fertility, e.g. inducers of ovulation or of spermatogenesis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P39/00General protective or antinoxious agents
    • A61P39/06Free radical scavengers or antioxidants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/24Drugs for disorders of the endocrine system of the sex hormones
    • A61P5/30Oestrogens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/24Drugs for disorders of the endocrine system of the sex hormones
    • A61P5/32Antioestrogens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23VINDEXING SCHEME RELATING TO FOODS, FOODSTUFFS OR NON-ALCOHOLIC BEVERAGES AND LACTIC OR PROPIONIC ACID BACTERIA USED IN FOODSTUFFS OR FOOD PREPARATION
    • A23V2002/00Food compositions, function of food ingredients or processes for food or foodstuffs

Definitions

  • HMR Based on the structure of HMR one had expected that 7-hydroxyenterolactone were the main metabolite of HMR, but this was not the case. This hydroxyl group is eliminated in the metabolism.
  • test compounds were added to incubation mixtures in a small volume diluted in ethanol or dimethylsulphoxide (2% of incubation volume), and the lipid peroxidation potency was compared to that of the vehicle (ethanol or dimethyl sulphoxide).
  • the reaction was initiated by 0.05 ml of 0.9 mM t-BuOOH at 33° C. Chemiluminescence was measured for about 45 min at 1 min cycles, and the area under curve (integral) was calculated. Chemiluminescence measurements were carried out using a Bio-Orbit 1251 Luminometer.(Bio-Orbit, Turku, Finland) connected to a personal computer using dedicated software for the assays.

Abstract

This invention relates to methods for prevention of cancers, certain non-cancer, hormone dependent diseases and/or cardiovascular diseases in a person, based on administering of hydroxymatairesinol to said person. The invention also concerns a method for increasing the level of enterolactone or another metabolite of hydroxymatairesinol in a person's serum thereby causing prevention of a cancer or a certain non-cancer, hormone dependent disease in a person, based on administering of hydroxymatairesinol to said person. Furthermore, this invention relates to pharmaceutical preparations, food additives and food products comprising hydroxymatairesinol.

Description

    FIELD OF THE INVENTION
  • This invention relates to methods for prevention of cancers, certain non-cancer, hormone dependent diseases and/or cardiovascular diseases in a person, based on administering of hydroxymatairesinol to said person. The invention also concerns a method for increasing the level of enterolactone or another metabolite of hydroxymatairesinol in a person's serum thereby causing prevention of a cancer or a certain non-cancer, hormone dependent disease in a person, based on administering of hydroxymatairesinol to said person. Furthermore, this invention relates to pharmaceutical preparations, food additives and food products comprising hydroxymatairesinol. [0001]
  • BACKGROUND OF THE INVENTION
  • The publications and other materials used herein to illuminate the background of the invention, and in particular, cases to provide additional details respecting the practice, are incorporated by reference. Lignans are defined as a class of phenolic compounds possessing a 2,3-dibenzylbutane skeleton. They are formed by coupling of monomeric units called precursors such as cinnamic acid, caffeic, ferulic, coumaric, and gallic acids (Ayres and Loike, 1990). Lignans are widely distributed in plants. They can be found in different parts (roots, leafs, stem, seeds, fruits) but mainly in small amounts. In many sources (seeds, fruits) lignans are found as glycosidic conjugates associated with fiber component of plants. The most common dietary sources of mammalian lignan precursors are unrefined grain products. The highest concentrations in edible plants have been found in flaxseed, followed by unrefined grain products, particularly rye. Mammalian lignan production from different plant food are given in Table 1. [0002]
  • Considerable amounts of lignans are also found in coniferous trees. The type of lignans differs in different species and the amounts of lignans vary in different parts of the trees. The typical lignans in heart wood of spruce (Picea abies) are hydroxymatairesinol (HMR), α-conidendrin, conidendrinic acid, matairesinol, isolariciresinol, secoisolariciresinol, liovile, picearesinol, lariciresinol and pinoresinol (Elknan 1979). The far most abundant single component of lignans in spruce is HMR, about 60 per cent of total lignans, which occurs mainly in unconjugated free form. Lignan concentration in thick roots is 2-3 per cent. Abundance of lignans occur in the heart wood of branches (5 - 10 per cent) and twists and especially in the knots, where the amount of lignans may be higher than 10 per cent (Ekman, 1976 and 1979). These concentrations are about hundred-fold compared to ground flax powder known as lignan-rich material. [0003]
  • The chemical structure of hydroxymatairesinol is [0004]
    Figure US20010016590A1-20010823-C00001
  • Lignans can be isolated e.g. from compression-wood fiber. These fibers originate from compression wood of stems and knots (oversize chip fraction) worsen the quality of paper (Ekman, 1976). [0005]
  • Plant lignans such as matairesinol and secoisolariciresinol, are converted by gut microflora to mammalian lignans, enterolactone and enterodiol, correspondingly (Axelson et al. 1982). They undergo an enterohepatic circulation and are excreted in the urine as glucuronide conjugates (Axelson and Setchell, 1981). As an experimental evidence for the chemopreventive actions of lignans, supplementation of a high-fat diet with lignan-rich flaxseed flour (5% or 10%) or flaxseed lignans (secoisolariciresinol-diglycoside, SDG) prevented the development of antiestrogen-sensitive DMBA-induced breast cancer in the rat (Serraino and Thompson 1991 and 1992; Thompson et al. 1996a and 1996b). They reduced the epithelial cell proliferation, nuclear aberrations, the growth of tumors, and the development of new tumors. High lignan intake may also protect against experimental prostate and colon cancers. Dietary rye (containing lignans), prevented at early stages the growth of transplanted Dunning R3327 prostatic adenocarcinomas in rats (Zhang et al. 1997; Landstrom et al. 1998). The percentage of animals bearing palpable tumors, the tumor volume, and the growth rate were significantly lower. Further, flaxseed or SDG supplementation inhibited the formnation of chemically induced aberrant crypts in rat colon (Serraino and Thompson 1992; Jenab and Thompson 1996). The antitumor action may therefore be due to weak estrogen-antiestrogen-like properties and/or other mechanisms, which are not well understood. [0006]
  • Urinary excretion and serum concentrations of enterolactone are low in women diagnosed with breast cancer (Ingram et al. 1997; Hulten et al. 1998) suggesting that lignans are chemopreventive. Mammalian lignans (enterolactone and enterodiol) have been hypothesized to modulate hormone-related cancers, such as breast cancer, because of their structural similarities to the estrogens. Enterolactone had weak estrogenic potency in MCF-7 cells (Mousavi and Adlercreutz 1992), but had no estrogenic response in mouse uterine weight (Setchell et al. 1981). As a sign of estrogen-like activity, SDG feeding during pregnancy and lactation to rats increased the uterine weight at weaning but the effect was not evident at later stages (Tou et al. 1998). Possible antitumor effects have also been associated with their antiestrogenic actions (Waters and Knowler, 1982). The inhibition of aromatase by mammalian lignan, enterolactone, would suggest a mechanism by which consumption of lignan- rich plant food might contribute to reduction of estrogen-dependent diseases, such as breast cancer (Adlercreutz et al. 1993, Wang et al. 1994). The potential antioxidant activity of lignans could also represent a mechanism associated with the preventive action of lignans in the development of cancers. Further, mammalian lignans have shown to inhibit the conversion of testosterone to 5a-dihydrotestosterone (DHT), the potent intracellular androgen, at the concentrations which are achievable in humans (Evans et al. 1995). The reduction in DHT concentration would modify the risk of prostate cancer (PC) and benign prostatic hyperplasia (BPH). [0007]
  • It is possible that lignans as precursors of enterolactone could also alleviate lower urinary tract symptoms (LUTS) and gynecomastia. On the basis of the results obtained in the animal model, we have suggested that estrogens play an essential role in the development of the muscular dysfunction involved in urethral dyssynergia seen as bladder neck dyssynergia or external sphincter pseudodyssynergia (Streng et al. unpublished observations). Such neuromuscular changes are at least partially reversed by an aromatase inhibitor (MPV-2213ad) indicating the role of estrogens. Further, gynecomastia, which is induced by exposure to estrogens or in the presence of increased ratio of estrogen to androgens. Gynecomastia can be successfully treated with an aromatase inhibitor. The capability of lignans to inhibit 5α-reductase and/or aromatase combined with their potential antioxidant activity may represent mechanisms associated with the preventive action of lignans in the development of hormone-related diseases in male organism. [0008]
  • No data is available on the possible effects of lignans in humans. The current theories about lignan action in humans have been derived from studies on the effects of diets supplemented with flaxseed products (and thus lignans). Flaxseed in human female diet caused changes in menstrual cycle (Phipps et al. 1993). The subjects, all normally cycling women, showed a longer mean length of luteal phase and higher progesterone/17β-estradiot ration in serum during the luteal phase when they took 10 g of flax seed powder/day in addition to their habitual diets (Phipps et al. 1993). No significant differences between flax and control cycles or concentrations of either estrone or 17β-estradiol were found. Neither there were any significant differences between flax and control groups for concentrations of serum estrogens in postmenopausal women (Brzezinski et al. 1997). Flaxseed supplementation increased SHBG (protein which binds estradiol with high capacity) concentration in serum. This is a typical estrogenic effect in the liver tissue. Increased SHBG concentration on the other hand reduces bioavailability of endogenous estrogens. In healthy young men, the short-term (6 weeks) flaxseed supplementation of the diet (10 g/d in muffins) had no significant effect on plasma testosterone concentrations (Shultz et al. 1991) indicating a lack of estrogenicity in the male organism. All together, these studies indicate that lignans may have weak hormonal (estrogenic and antiestrogenic) effects, but the mechanism of their action cannot be fully described by the hormonal effects. [0009]
  • In conclusion, isolated mammalian lignans have not been available earlier in sufficient amounts to be used in animal experiments or clinical trials, and the only possibility to increase lignan intake has been to increase the consumption of fiber-rich food items such as flaxseed. HMR or any other lignan that is efficiently converted to enterolactone, and can be produced/isolated in large quantities would be valuable in the development of pharmaceutical preparations and food products such as functional foods for chemoprevention of cancer and other hormone-related diseases and cardiovascular diseases. [0010]
  • SUMMARY OF THE INVENTION
  • According to one aspect, this invention concerns a method for prevention of a cancer, a certain non-cancer, hormone dependent disease and/or a cardiovascular disease in a person comprising administering to said person an effective amount of hydroxymatairesinol or a geometric isomer or a stereoisomer thereof. [0011]
  • According to an other aspect, the invention concerns a method for increasing the levels of enterolactone or another metabolite of hydroxymatairesinol in a person's serum thereby causing prevention of a cancer or a certain non-cancer, hormone dependent disease in a person comprising administering to said person an effective amount of hydroxymatairesinol or a geometric isomer or a stereoisomer thereof. [0012]
  • According to a third aspect, the invention concerns a pharmaceutical preparation comprising an effective amount of hydroxymatairesinol or a geometric isomer or a stereoisomer thereof in combination with a pharmaceutically acceptable carrier. According to a fourth aspect, the invention concerns a product comprising a liquid or solid material enriched with hydroxymatairesinol or a geometric isomer or a stereoisomer thereof, for use as additive to a food product. [0013]
  • According to a fifth aspect, the invention concerns a food product comprising an effective amount of hydroxymatairesinol or a geometric isomer or a stereoisomer thereof. [0014]
  • According to still one aspect, the invention concerns a method for increasing the stability of a food product comprising the addition to said food product of an effective amount of hydroxymatairesinol or a geometric isomer or a stereoisomer thereof [0015]
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 shows the concentration-related inhibition of aromatase by lignans in JEG-3 cells. [0016]
  • FIG. 2 shows the proliferation of MCF-7 cells in the presence and absence of HMR. [0017]
  • FIG. 3 shows the uterine wet weight of immature rats treated with HMR or with an aromatase inhibitor. [0018]
  • FIG. 4 shows the antitumor activity of HMR against DMBA-induced mammary gland tumors in female rats. [0019]
  • FIG. 5 shows the excretion of enterolactone in the urine of rats treated with different doses of HMR. [0020]
  • DETAILED DESCRIPTION OF THE INVENTION
  • This invention relates to the use of a lignan, hydroxymatairesinol (HMR), for the prevention of cancer, non-cancer, hormone dependent diseases and cardiovascular diseases by adding said HMR into food or by using it as a pharmaceutical preparation Surprisingly, HMR is metabolized in vivo to enterolactone, which is assumed to account at least partly for the antitumor properties of the lignans. Antioxidative activity of HMR in vitro is strong and this property indicates that HMR can also prevent cardiovascular diseases through the protective effect against damaging free oxygen species in the body. The invention relates also to the use of HMR as a food additive to increase the food stability (i.e. inhibit lipid and pigment oxidations and vitamin losses which cause loss of nutritional value and development of off-flavors in food). [0021]
  • The method according to this invention is particularly effective in the prevention of cancers such as breast cancer, prostate cancer and colon cancer, non-cancer, hormonal dependent diseases such as lower urinary tract symptoms, urethral dyssynergia, bladder instability, bladder outlet obstruction, benign prostatic hyperplasia, and gynecomastia in men, and cardiovascular diseases resulting from oxidized LDL in serum. [0022]
  • The pharmaceutical preparation according to this invention is preferably an oral formulation . The required amount of the active compound (HMR) will vary with the particular condition to be prevented. A typical dose ranges from about 10 to about 100 mg per day and adult person. [0023]
  • In the food additive of the invention, the material to be enriched with hydroxymatairesinol can be any edible, non-toxic solid or liquid material suitable to be admixed with HMR without affecting the properties of HMR. The role of the material is mainly to make the exact dosage of HMR easier. A suitable concentration is for example 100 mg to 1 g of HMR per 100 g of enriched material. [0024]
  • The food product according to this invention is especially a functional food, a nutritional supplement, a nutrient, a pharmafood, a nutraceutical, a health food, a designer food or any food product. A suitable concentration of HMR in the food product is, for example, 1 to 20 mg of HMR per 100 g of food product. [0025]
  • The functional food according to this invention can, for example be in the form of butter, margarin, biscuits, bread, cake, candy, confectionery, yogurt or an other fermented milk product, or cereal such as muesli. [0026]
  • The addition of hydroxymatairesinol is particularly useful to increase food stability in the meaning of inhibitition of lipid, vitamin and pigment oxidations, which cause loss of nutritional value and development of off-flavors in food. A suitable concentration of HMR for this purpose is, for example, about 0.1%. [0027]
  • Isolation of HMR for use in this invention can be made from oversize chip fraction (containing branches, twists and knots) of compression wood and use of HMR in the prevention of diseases such as cancer and cardiovascular diseases. [0028]
  • The properties of HMR were studied by seven different assays: [0029]
  • 1. Measurement of antioxidant capacity in vitro [0030]
  • 2. Measurement of aromatase inhibiting capacity in JEG-3 cells [0031]
  • 3. Measurement of estrogenic and antiestrogenic activity in MCF-7 cell cultures [0032]
  • 4. Evaluation of estrogenic and antiestrogenic activity by uterine growth bioassay [0033]
  • 5. Measurement of estrogenic and antiestrogenic activity in adult male rats [0034]
  • 6. Investigating the antitumor activity in rat DMBA-induced mammary cancer model [0035]
  • 7. Analysis of metabolites from rat urine after different doses of HMR [0036]
  • The isolation and purification of HMR in sufficient amounts for biological tests has been impossible earlier because it is a component of wood lignans, which have been relatively poorly characterized. Understanding the distribution of HMR in different parts of spruce (Ekman 1976 and 1979) has given the opportunity to study lignans and especially HMR in detail. [0037]
  • A linear correlation was found between the doses of HMR and the amounts of urinary enterolactone. Enterolactone is a well known mammalian lignan formed by intestinal bacteria from matairesinol or by oxidation of enterodiol (Axelson and Setchell 1981; Axelson et al. 1982). Only minute amounts of unmetabolized HMR and other metabolites (enterodiol and 7-hydroxyenterolactone) were found in urine. Their amounts remained unchanged when the daily dose of HMR was increased. These findings suggest that HMR was metabolized to enterolactone, and, further, enterolactone derived from HMR through demethylation and dehydroxylation steps is not converted to enterodiol. Based on the structure of HMR one had expected that 7-hydroxyenterolactone were the main metabolite of HMR, but this was not the case. This hydroxyl group is eliminated in the metabolism. The metabolism of HMR differs from that of SDG. SDG is metabolized to enterodiol which is partly oxidized to enterolactone (Rickard et al. 1996; Lampe et al. 1994). HMR thus offers an advantage over SDG as a direct precursor of enterolactone. [0038]
  • HMR had weak if any estrogenic action in rat uterus or in the male organism. It exerted weak but not significant estrogen-like activity in MCF-7 cells. No antiestrogenic activity was demonstrated for HMR. Therefore, it is surprising that it had highly significant antitumor activity in DMBA-induced tumor model in rats as shown in FIG. 2. The activity of HMR may be due to HMR itself or to enterolactone. However, no dose-dependence was found in the chemopreventive action of HMR when given in two different doses (3 and 15 mg/kg) to rats after DMBA-treatrient. Thus HMR needs not to be converted to enterolactone to have an antitumor effect or smaller doses of these lignans are sufficient to accomplish the maximal chemopreventive effects. [0039]
  • HMR is very effective antioxidant as shown in Tables 2 and 3. It is one of the most potent known inhibitors of lipid peroxidation and excellent inhibitor of LDL oxidation. Inhibition of LDL oxidation is considered to be of special importance in humans as the concentration of oxidized LDL in serum is considered to be one of the best predictors of cardiovascular diseases such as atherosclerosis. HMR may serve as a food additive to increase the food stability (i.e. inhibit vitamin, lipid and pigment oxidations which cause loss of nutritional value and development of off-flavors in food), because HMR was much better superoxide anion scavenging and peroxyl radical scavenging agent than well known antioxidants butylated hydroxyanisol (BHA) and butylated hydroxytoluene (BHT), which are commonly used for increasing the food stability. [0040]
  • EXPERIMENTS
  • Chemicals [0041]
  • Various lignans were tested in vitro for their estrogenicity, antiestrogenicity, capability to inhibit aromatization and for their antioxidative properties. The test compounds were purchased from the following sources: enterodiol and enterolactone from Plantech, London, UK, and 7-hydroxyenterolactone containing two 7-OH enantiomers was a generous gift from Dr. Kristina Wahadla, Department of Applied Chemistry, University of Helsinki, Finland. [0042]
  • Extraction of HMR from wood [0043]
  • HMR extracts- were isolated from Norway spruce (Picea abies) as described by Ekman, 1976 and Ekman 1979. Shortly, freeze-dryed ground heartwood was Soxhlet-extracted in with hexane to remove non-polar lipophilic extractive. The wood sample was re-extracted in the same apparatus with acetone/water (9:1 v/v) to give crude lignans. Hydroxymatairesinol (HMR) and its isomer were isolated and re- chromatographed with XAD-resin for further purification. [0044]
  • Measurement of antioxidant capacity in vitro [0045]
  • The antioxidative capacity of lignans was estimated by four different methods: 1) inhibition of lipid peroxidation, 2) inhibition of low density lipoprotein (LDL) oxidation, 3) superoxide anion scavenging and 4) peroxyl radical scavenging assays. [0046]
  • Inhibiton of lipid peroxidation was evaluated on the basis of their potency to inhibit tert-butylhydroperoxide-induced lipid peroxidation (t-BuOOH-LP) in rat liver microsomes in vitro (Ahotupa et al. 1997). The test for the t-BuOOH-LP was carried out as follows: The buffer (50 mM sodium carbonate, pH 10.2, with 0.1 mM EDTA) was pipetted in a volume of 0.8 ml in the luminometer cuvette. Twenty microliters of diluted liver microsomes, final concentration 1.5 mg protein/ml, was added, followed by 6 ml of luminol (0.5 mg/ml) and test chemicals. The test compounds were added to incubation mixtures in a small volume diluted in ethanol or dimethylsulphoxide (2% of incubation volume), and the lipid peroxidation potency was compared to that of the vehicle (ethanol or dimethyl sulphoxide). The reaction was initiated by 0.05 ml of 0.9 mM t-BuOOH at 33° C. Chemiluminescence was measured for about 45 min at 1 min cycles, and the area under curve (integral) was calculated. Chemiluminescence measurements were carried out using a Bio-Orbit 1251 Luminometer.(Bio-Orbit, Turku, Finland) connected to a personal computer using dedicated software for the assays. [0047]
  • Inhibition of LDL oxidation was estimated as described by Ahotupa et al, 1996. Shortly: LDL was isolated by precipitation with buffered heparin. After resuspendation in phosphate buffer, 20 mM CuCl[0048] 2 was added and the mixture was incubated for 3 hrs at +37° C. After this, LDL lipids were extracted with chloroform-methanol, dried under nitrogen, redissolved in cyclohexane and analyzed spectrophotometrically at 234 nm. The intensity of absorbance is indicative of LDL oxidation. To test the ability of different compounds to prevent LDL oxidation, the compounds were added to the incubation mixture prior addition of CuCl2. Possible interference of test compounds with the assay procedure was excluded by measuring the absorption at 234 nm before and after the incubation period. For those compounds which showed antioxidative potency at the starting concentration (0,1 mM), IC-50 values (i.e. concentrations at which test compound inhibited LDL oxidation by 50%) was determined.
  • Superoxide anion scavenging method was based on the superoxide anion produced in controlled conditions by xanthine-xanthine oxidase system and detection of the generated reactive oxygen species by luminometer (Ahotupa et al., 1997). The ability of test compounds to decrease the chemiluminescence was evaluated. IC-50 concentration (concentration which prevented the chemiluminescence by 50%) was calculated. [0049]
  • Peroxyl radiocal scavenging assay was based on generation of peroxyl radicals by thermal decomposition of 2,2′-azobis(2-amidinopropane)HCl and their detection by chemiluminescence (Ahotupa et al., 1997). The results were calculated as the stochiometric factor, i.e. how many moles of peroxyl radicals can be scavenged by one mole of the test compound. [0050]
  • Measurement of aromatase inhibiting capacity in JEG-3 cells [0051]
  • The effects of HMR and structurally related lignans (enterolactone, enterodiol and 7-hydroxyenterolactone) were studied on formation of [0052] 3H-17β-estradiol from 3H-andostenedione in JEG-3 cells, human choriocarcinoma cell line. The JEG-3 choriocarcinoma cells are a useful aromatase model enabling the study of aromatase inhibition in vitro (Krekels et al 1991). Cells were maintained in DMEM containing 10% fetal calf serum (FCS). The incubation mixture contained 50 μl 3H-androst-4-ene, 3,17-dione (0.5 nM), 50 μl unlabelled androstenedione (0.5 nM), 100 μl test compounds (10 mM) and 800 μl cell suspension (1 million cells). After the incubation for 4 h, unlabelled carriers (androstenedione, testosterone, 17β-estradiol and estrone) were added. The steroids were extracted twice with 3.0 ml dichloromethane. HPLC was used for separation and quantification of the radiolabelled 3H-17β-estradiol as previously described (Makela et al. 1995). The column system consisted of a guard column followed by a C18 150×3.9 mm ID analytical column (Technopak 10 C18 HPLC Technology; Wellington House, Cheshire, UK). The mobile phase was acetonitrile /water (35/65) and the flow rate was 1.2 ml/min. For in-line detection of the radioactive metabolites, the eluent of the HPCL column was continuously mixed with liquid scintillant and then monitored with in-line radioactivity detector.
  • Measurement of estrogenic and antiestrogenic activity in MCF-7 cell cultures [0053]
  • The MCF-7 cell line (human breast cancer cells) stock cultures were grown in phenol red free RPMI medium supplemented with 5 % FCS, 100 U/ml penicillin and 100 μg/ml streptomycin, 10 μg/ml insulin and 1 nM 17μ-estradiol in T-75 cell culture bottles. The medium was replaced with fresh ones three times per week. The stock cultures were harvested by trypsinization and suspended in 10 ml phenol red free versene solution and centrifuged for 5 [0054] min 800 rpm. The cell pellet was carefully resuspended into RPMI medium supplemented with 5% dextran charcoal stripped FCS (dcFCS) and seeded on 6 well plates 50 000 cells/ 3.0 ml medium/ well. On the second day of culture the medium was changed and test compounds were added. To test the estrogenicity of the lignan compounds, they were diluted in ethanol and added to cell cultures in final concentration of 1.0 M. In each proliferation assay 1.0 nM 1 7β-estradiol solution in ethanol was used as a positive control for estrogenic response. Equal amounts of ethanol were added to control wells. To test the antiestrogenicity both 17β-estradiol and lignan solutions were added to cell cultures. The cells were cultured for 5 to 7 days in the presence of test compounds, and the medium was changed every second day. Cell proliferation was quantified by counting the released nuclei with Coulter counter.
  • Evaluation of estrogenic and antiestrogenic activity in immature rat uterotropic test [0055]
  • The estrogenicity HMR was evaluated by the uterotropic assay in immature rats which was performed as described earlier (Jordan et al. 1977), with the exception of treatment time which was 7 days instead of 3 days in the reference study. The treatment time was longer because of the expected weak estrogenicity of the test compound. The treatment of immature rats with an aromatase inhibitor (MPV-2213ad), which prevents biosynthesis of estradiol, was used as a methodological control for non-estrogen-stimulated uterus. [0056]
  • Evaluation of estrogenic and antiestrogenic activity in adult male rats [0057]
  • Estrogenic (anfiandrogenic) and antiestrogenic effects of HMR were studied in intact and hypoandrogenic Noble strain male rats (age 6-9 month), correspondingly. The chronic hypoandrogenic state with both structural and functional changes in the male reproductive tract was induced by neonatal estrogenization (diethylstilbestrol, 10.0 μg/kg body weight in rape oil s.c. on postnatal days 1-5). These changes are known to be partly reversible by aromatase inhibitor treatment consisting daily dose of MPV-2213 ad 10-30 mg/kg body weight (Streng et al. unpublished observations). [0058]
  • Animals were fed the soy-free basal diet (SDS, Whitham Essex, England) and they had a free access to water. Twelve of both intact and hypoandrogenic animals were cavaged in daily dosage of [0059] HMR 50 mg/kg body weight in rape oil. Another twelve animals from both animal models were cavaged with rape oil only as a placebo treatment. After four-week treatment the animals were sacrificed. The weights of testis and accessory sex glands (ventral prostate, seminal vesicles and coagulating gland) were measured. Serum and testis testosterone and pituitary and serum luteinizing hormone (LH) levels were measured by immunoassays (Haavisto et al. 1993).
  • Investigating the antitumor activity in rat DMBA-induced mammary cancer model [0060]
  • Antitumor activity of HMR in rat mammary cancer was studied as described earlier (Kangas et al. 1986). Fifty-day-old female Sprague-Dawley rats were given 12.0 mg DMBA (dimethylbentz[a]anthracene) by cavage. After approximately 6 weeks palpable tumors could be detected, whereafter the width (w) and the length (1) of the tumors were measured once a week to determine the tumor volumes according to a formula V=(πw[0061] 21)/12. The rats were also weighed once a week. The rats were allocated in 3 different groups so that the total number of tumors in the beginning of the experiment was similar in each group: (1) Control group 8 animals, (2) HMR 3.0 mg/kg 7 animals, and (3) HMR 15.0 mg/kg 7 animals, one of which had to be killed before the end of the experiment.
  • HMR was given per os starting 9 weeks after the DMBA-induction, i.e. 3 weeks after palpable tumors appeared, and was given daily for 7.5 weeks. At the end of the experiment the tumors were classified in groups according to their growth pattern: 1. Growing tumors (PD=progressive disease); 2. Non-growing, stabilized tumors (SD=stabilized disease, no change in tumor volume or regression less than 75%; 3. Regressing tumors (PR=partial response, regression of tumor volume more than 75%); 4. Disappeared tumors (CR=complete response, no palpable tumor). [0062]
  • Analysis of metabolites from rat urine receiving different doses of HMR [0063]
  • Ten Sprague—Dawley male rats ([0064] age 4 month) were used to study the metabolism of HMR in vivo. Animals were housed in pairs with 12 h light:dark cycle and had free access to water and soy-free basal diet (SDS, Whitham Essex, England) during the metabolism study. Rats were cavaged with HMR dissolved in 10% ethanol in PEG in doses 3, 15, 25 and 50 mg/kg body weight once a day for two days. After second cavaging the 24 hour urine was collected in metabolic cages in collection jars containing 120 μ0,56 M ascorbic acid and 120 μl 0,15 M Na-azide as preservatives. The centrifuged urine volumes were measured and stored in −20° C. For pretreatment 750 μl 0.2 M acetate buffer (pH 4.0±0.1) was added to 3.0 ml thawed urine aliquots. Sep-Pak C18 columns (100 mg silica based resin/column) were used for urine extractions. Columns were preconditioned with 3.0 ml H2O, 3.0 ml methanol and 3.0 ml acetate buffer. After urine had filtered through the column and washed with 3.0 ml of acetate buffer polyphenolics were eluted with 3.0 ml methanol. The eluate was evaporated to dryness under nitrogen in +45° C. water bath and dryed residues were redissolved in 3.0 ml of 0.2 M acetate buffer. 30 μl Helix pomatia enzyme mix was added and the solutions were incubated in +37° C. to hydrolyze both glucuronides and sulfates. 300 μl of flavone stock solution (100 μg/ml in EtOH) was added into hydrolyzed samples. The samples were extracted in C-18 columns and evaporated to dryness as described above and stored in −20° C. until analyzed with GC-MS.
  • The evaporated urine samples were dissolved in pyridine, and silylated by adding BSTFA:TMCS (10:1) silylation reagent. The GC-MS analyses of the silylated samples were performed with an HP 6890-5973 GC-MS instrument. The GC column was an HP-1 crosslinked methyl polysiloxane column (15 m×0,25 mm i.d., 0,25 μm film thickness). Helium was used as carrier gas at a flow of 1 ml/min. The GC-oven was temperature programmed from 60° C. to 290° C., at 8OC/min heating rate. The GC-injector was set in split-mode at a split ratio of 1:15. The injector temperature was 250° C. Compound identifications were based on mass spectra. The quantitative calculations were based on uncorrected peak areas of target components relative to the internal standard. [0065]
  • RESULTS
  • Assessment of antioxidant capacity in vitro [0066]
  • HMR had stronger lipid peroxidation capacity than any other lignan or flavonoid in our tests (Table 2). HMR was compared to well known antioxidants TROLOX, which is a water soluble vitamin E derivative, and BHA and BHT in the ability of inhibiting lipid peroxidation, inhibition of LDL oxidation, and scavenging superoxide and peroxyl radicals (Table 3). HMR was as a whole the strongest antioxidant, more effective than BHA or BHT in all assays, and stronger than TROLOX in all assays except for lipid peroxidation inhibition assay, where the compounds were almost equally active. [0067]
  • Aromatase inhibiting capacity in JEG-3 cells [0068]
  • The inhibition of [0069] 3H- 17β-estradiol formation from 3H-androstenedione in JEG-3 cells was tested at different concentrations of HMR. The inhibitory capacity of HMR was compared to enterolactone, 7-hydroxyenterolactone and enterodiol. Enterolactone caused a dose-dependent inhibition of aromatization within the concentration range of 1.0 to 10.0 μM. It was further shown that enterodiol was noninhibitory indicating that the lactone ring is critical for the inhibition. 7-hydroxyenterolactone and hydroxymatairesinol had no inhibitory effects (FIG. 1) indicating the importance of the number and location hydroxyl groups in the lignan molecule for the aromatase inhibition.
  • Estrogenic and antiestogenic activity in MCF-7 cell cultures [0070]
  • HMR had very weak, not statistically significant estrogenic or antiestrogenic activi-,,t in MCF-7 cell proliferation assays as shown in FIG. 2. [0071]
  • Evaluation of estrogenic and antiestrogenic activity in immature rat uterotropic test [0072]
  • FIG. 3 illustrates the effects of HMR on the uterine growth of the immature rats. HMR had no significant estrogenic effect on the uterine weight gain of the immature rats. Neither did HMR reduce the weight gains indicating no antiestrogenic effect. Aromatase inhibitor prevented the increase of uterine weight, as expected, indicating that the method for the measurement of the aromatase inhibitors was adequate. [0073]
  • Evaluation of estrogenic and antiestrogenic activity in adult male rats [0074]
  • After a 4-week treatment with HMR, no significant changes in the weights of accessory sex glands and testis were observed in control and hypoandrogenic animals (Table 4). There were no significant changes in testosterone or LH concentrations, either (Table 5). These results indicate, that HMR is not a full estrogen agonist in male organism, because it does not exert the typical estrogenic activity on hypothalamus—hypophysis—gonad—axis (inhibition of LH and androgen secretion). Neither is HMR an antiestrogen because it does not reverse the changes induced by neonatal estrogenization in the male rat. [0075]
  • Investigating the antitumor activity in rat DMBA induced mammary cancer model [0076]
  • Number of growing (PD) versus stable (SD) tumors, regressing (PR) tumors and disappeared (CR) tumors is presented in FIG. 4. The antitumor effect of HMR was found to be statistically very significant. There was no clear dose-dependency of antitumor action in this model. Both antioxidative and tumor growth regressing properties of HMR may therefore be connected with the in vivo antitumor activity. The mechanism of antitumor activity of HMR in vivo is still unknown. [0077]
  • Analysis of metabolites from rat urine after different doses of HMR [0078]
  • FIG. 5 illustrates that the main excreting metabolite of HMR in rats is enterolactone, which may be the biologically active compound. This is surprising taking into account the chemical structure of HMR, because one would expect hydroxyenterolactone to be the main metabolite. The metabolism of HMR to enterolactone may be catalyzed by bacterial intestinal flora rather than by the rat liver. [0079]
  • CONCLUSIONS
  • Hydroxymatairesinol (HMR) has antitumor activity either as unchanged compound and/or after conversion to enterolactone in DMBA induced breast cancer model. HMR has therefore a potential to have beneficial effects in humans who are at risk of developing breast cancer (BC), prostate cancer (PC), colon cancer or benign prostatic hyperplasia (BPH). HMR is metabolized to enterolactone which inhibits aromatization in vitro. HMR may as a precursor of aromatase inhibitor also prevent the development of lower urinary tract symptoms (LUTS), bladder instability, bladder outlet obstruction, urethral dyssynergia, and gynecomastia. HMR has also strong antioxidative activity and may therefore be used as food additive (antioxidant). HMR as pharmaceutical product or dietary supplement may have advantageous cardiovascular effects in humans. Addition of HMR to food to make innovative new functional food, nutraceutical, health food, pharmafood, designer food or novel food is feasible. [0080]
  • It will be appreciated that the methods of the present invention can be incorporated in the form of a variety of embodiments, only a few of which are disclosed herein. It will be apparent for the specialist in the field that other embodiments exist and do not depart from the spirit of the invention. Thus, the described embodiments are illustrative and should not be construed as restrictive. [0081]
    TABLE 1
    Production of mammalian lignans from different plant food
    by in vitro fermentation with human fecal flora.
    μg/100 g
    FLAXSEED FLOUR 68000
    SOYBEAN  170
    CEREAL BRANS:
    WHEAT  570
    OAT  650
    WHOLE CEREALS:
    RYE  160
    POTATO   80
    CARROT  350
    ONION  110
  • [0082]
    TABLE 2
    ANTIOXIDANT PROPERTIES OF LIGNANS AND SOME
    RELATED FLAVONOIDS IN VITRO BY LIPID
    PEROXIDATION INHIBITION TEST.
    Antioxidative
    capacity
    (t-BuOOH-LP)
    Compound IC50 (μM)
    Flavonoids
    kaempferol 0.9
    (3,4′,5,7-tetrahydroxy-
    flavone)
    quercetin 0.4
    (3,3′,4′,5,7-pentahydroxy-
    flavone)
    kaempferide 0.5
    (3,5,7-trihydroxy-4′-methoxy-
    flavone)
    Lignans
    enterolactone 15.9
    2,3-bis-(3’-hydroxybenzyl)-butyrolactone
    enterodiol 12.7
    2,3-bis-(3’-hydroxybenzyl)-butane-1,4-diol
    hydroxymatairesinol 0.08
  • [0083]
    TABLE 3
    COMPARISON OF ANTIOXIDATIVE EFFECTS OF HMR AND
    KNOWN ANTIOXIDANTS IN VITRO. IC-50 concentrations
    have been presented, except for peroxyl radical scavenging
    assay where the stochiometric factor (i.e. how many moles
    of peroxyl radical one mole of test compound can scavenge).
    HMR1 TROLOX2 BHA3 BHT4
    Inhibition of lipid 0,06 μM 0,02 μM 1,1 μM 15,3 μM
    perioxidation
    Inhibition of LDL 2,0 μM 2,7 μM not determined
    oxidation
    Superoxide anion
    5,6 μM 25 μM 15 μM >1 mM
    scavenging
    Peroxyl radical 1:4 1:2 not determined
    scavenging
  • [0084]
    TABLE 4
    The effect of four week exposure to HMR on male rat reproductive organ relative weights1
    Ventral Seminal Coaculating
    Body weight Testis Prostate vesicle gland
    Treatment n g mg/kg body weight
    Intact animals Placebo 12 426 ± 28 4362 ± 170 909 ± 146 412 ± 43 223 ± 49
    HMR 12 447 ± 38 4223 ± 304 938 ± 148 419 ± 59 204 ± 48
    50 mg/kg
    Hypoandrogenic Placebo 12 481 ± 29 3340 ± 509 333 ± 188 249 ± 63  69 ± 49
    animals HMR 12 455 ± 36 3276 ± 327 378 ± 198 266 ± 49  70 ± 30
    50 mg/kg
  • [0085]
    TABLE 5
    The effect of four week exposure to HMR on male
    rat testosterone and LH concentrations1
    Testis
    testos- Serum
    terone testos- Pituitary
    (ng/ terone LH Serum LH
    Treatment n testis) (ng/ml) (μg/pit) (ng/ml)
    Intact Placebo 12  97,6 ± 2,405 ± 6,747 ± 1,804 ±
    animals  46,3 1,122 2,479 1,294
    50 mg/kg 12 112,9 ± 2,770 ± 6,838 ± 1,088 ±
    HMR  58,5 1,421 2,061 0,352
    Hypoan- Placebo 12  63,5 ± 1,197 ± 8 673 ± 0,712 ±
    drogenic  25,9 0,663 2,224 0,371
    animals 50 mg/kg 12  48,0 ± 0,939 ± 7,530 ± 0,854 ±
    HMR  15,2 0,431 2,286 0,333
  • REFERENCES
  • Adlercreutz H, Bannwart C, Wähälä K, Makela T, Brunow G, Hase T, Arosemena P J, Kellis J T, and Vickery L E: Inhibition of human aromatase by mammalian lignans and isoflavonoid phytoestrogens. J Steroid Biochem Mol Biol, 44: 147-153, 1993. [0086]
  • Ahotupa M, Ruutu M, and Mäntylä E: Simple methods of quantifying oxidation products and antioxidant potential of low density lipoproteins. Clin Biochem, 29: 139-144, 1996. [0087]
  • Ahotupa M, Mäntylä E, and Kangas L: Antioxidant properties of the triphenylethylene antiestrogen drug toremifene. Naunyn-Schmiedeberg's Arch Pharmacol, 356: 297-302, 1997. [0088]
  • Axelson M and Setchell KDR: The excretion of lignans in rats—evidence for an intestinal bacterial source for this new group of compounds. FEBS lett, 123: 337-342, 1981. [0089]
  • Axelson M, Sjövall J, Gustafsson BE and Setchell KDR: Origin of lignans in mammals and identification of a precursor from plants. Nature, 298: 659-660, 1982. [0090]
  • Ayres D, and Loike, J. Lignans: Chemical, bilogical and clinical properties. Cambridge university press, 1990. [0091]
  • Brzezinski A, Adlercreutz H, Shaoul R, Rosler A, Shmueli A, Tanos V and Schenker J G: Short-term effects of phytoestrogen-rich diet on postmenopausal women. Menopause (The Journal of the North American Menopause Society), 4:89-94, 1997. [0092]
  • Ekman R: Analysis of lignans in Norway spruce by combined gas chromatography-mass spectrometry. Holzforschung, 30:79-85, 1976. [0093]
  • Ekman R: Distribution of lignans in Norway spruce. Acta Academiae Aboensis, Ser B, 39:1-6, 1979. [0094]
  • Evans B A, Griffiths K and Morton M S. Inhibition of 5α-reductase in genital skin fibroblasts and prostate tissue by dietary lignans and isoflavonoids. J Endocrinol, 147: 295-302, 1995. [0095]
  • Haavisto A-M, Petterson K, Bergendahl M, Perheentupa A, Roser J F, and Huhtaniemi I A. Supersensitive immunofluorometric assay for rat luteinizing hormone. Endocrinology, 132: 1687-1691,1993. [0096]
  • Hultén K, Adlercreutz H, Winkvist A, Lenner P, Hallmans G and Agren A. Low levels of phyto-estrogens in blood as risk factor for breast cancer. In: COST 916 Workshop ‘Phyto-oestrogens: exposure, bioavailability, health benefits and safety concerns’, 1998 [0097]
  • Ingram D, Sanders K. Kolybaba M and Lopez D. Case-control study of phyto-oestrogens and breast cancer. Lancet, [0098] Oct 4;350(9083): 990-994, 1997.
  • Jenab M and Thompson L U. The influence of flaxseed and lignans on colon carcinogenesis and beta-glucuronidase activity. Carcinogenesis, Jun; 17(6):1343-1348, 1996 [0099]
  • Jordan V C, Collins M M, Rowsby L, Prestwich G: A monohydroxylated metabolite of tamoxifen with potent antiestrogenic activity. J Endocrinol, 75: 305-316, 1977. [0100]
  • Kangas L, Nieminen A-L, Blanco G, Grönroos M, Kallio S, Ka jalainen A, Perilä M, Södervall M and Toivola R: A new triphenylethylene compound, Fc-1157a. II Antitumor effects. Cancer Chemother Pharmacol, 17:109-113, 1986. [0101]
  • Lampe J W, Martini M C, Kurzer M S, Adlercreutz H and Slavin J L: Urinary lignan and isoflavonoid excretion in premenopausal women consuming flaxseed powder. Am J Clin Nutr, 60:122-8, 1994. [0102]
  • Landstrom M, Zhang J X, Hallmans G, Aman P, Bergh A, Damber J E, Mazur W, Wähälä K and Adlercreutz H. Inhibitory effects of soy and rye diets on the development of Dunning R3327 prostateadenocarcinoma in rats. Prostate, [0103] Aug 1; 36(3): 151-161, 1998
  • Mousavi Y and Adlercreutz H: Enterolactone and estradiol inhibit each other's proliferative effect on MCF-7 breast cancer cells in culture. J Steroid Biochem Mol Biol, 41: 615-619, 1992. [0104]
  • Mattinen J, Sjöholm R and Ekman R. NMR-spectroscopic study of hydroxymatairesinol, the major lignan in Norway spruce (Picea abies) heartwood. ACH models in chemistry, 135(4):583-590, 1998. [0105]
  • Makela S, Poutanen M, Lehtimaki J, Kostian M-L, Santti R and Vihko R. Estrogen-specific 170-hydroxysteroid oxidoreductase type 1 (E.C. 1.1.1.62) as a possible target for the action of phytoestrogens. P.S.E.B.M., 208:51-59, 1995. [0106]
  • Phipps W R, Martini M C, Lampe J W, Slavin J L and Kurzer M S. Effect of flax seed ingestion on the menstrual cycle. J Clin Endocrinol Metab, 77(5):1215-1219, 1993. [0107]
  • Rickard S E, Orcheson L J, Seidl M M, Luyengi L, Fong H H S and Thompson L U: Dose-dependent production of mammalian lignans in rats and in vitro from the purified precursor secoisolariciresinol diglycoside in flaxseed. J Nutr, 126: 2012-2019, 1996. [0108]
  • Shultz T D, Bonorden W R and Seaman W R. Effect of short-time flaxseed consumption on lignan and sex hormone metabolism in men. Nutrition Research, 11:1089-110, 1991. [0109]
  • Serraino M and Thompson L U: The effect of flaxseed supplementation on early risk markers for mammary carcinogenesis. Cancer Letters, 60: 135-142, 1991. [0110]
  • Serraino M and Thompson L U: The effect of flaxseed supplementation on the initiation and promotional stages of mammary tumorigenesis. Nutr Cancer, 17:153-159, 1992. [0111]
  • Setchell K D R, Borriello S P, Gordon H, Lawson A M, Harkness R and Morgan D M L. Lignan formation in man—microbial involvement and possible roles in relation to cancer. Lancet, 4:4-7, 1981 [0112]
  • Streng T, Talo A and Santti R. Unpublished observations. [0113]
  • Thompson L U, Robb P, Serraino M and Cheung F. Mammalian lignan production from various foods. Nutr Cancer, 16: 43-52, 1991 [0114]
  • Thompson L U, Seidl, M M, Rickard S E, Orcheson, L J, and Fong H H S: Antitumorigenic effect of a mammalian lignan precursors from flaxseed. Nutr Cancer, 26: 159-165, 1996a. [0115]
  • Thompson L U, Rickard S E, Orcheson L J and Seidl M M: Flaxseed and its lignan and oil components reduce mammary tumor growth at a late stage of carcinogenesis. Carcinogenesis, 17: 1373-1376, 1996b. [0116]
  • Tou J C L, Chen J and Thompson U. Flaxseed and its lignan precursor, secoisolariciresinol diglycoside, affect pregnancy outcome and reproductive development in rats. J Nutr, 128: 1861-1868, 1998. [0117]
  • Wang C, Mäkelä T, Hase T, Adlercreutz H and Kurzer M S: Lignans and flavonoids inhibit aromatase enzyme in human adipocytes. J Steroid Biochem Molec Biol, 50: 205-212, 1994. [0118]
  • Waters A P and Knowler J T. Effect of a lignan (HPMF) on RNA synthesis in the rat uterus. J Reprod. Fert, 66:379-381, 1982. [0119]
  • Zhang J-X, Hallmans G, Landström M, Bergh A, Damber J-E, Åman P and Adlercreutz H. soy and rye diets inhibit the development of Dunning R3327 prostatic adenocarcinoma in rats. Cancer Letters, 114: 313-314, 1997. [0120]

Claims (16)

1. A method for prevention of a cancer, a certain non-cancer, hormone dependent disease and/or a cardiovascular disease in a person comprising administering to said person an effective amount of hydroxymatairesinol or a geometric isomer or a stereoisomer thereof.
2. The method according to
claim 1
wherein said cancer is selected from the group consisting of breast cancer, prostate cancer and colon cancer.
3. The method according to
claim 1
wherein said non-cancer, hormonal dependent disease is selected from the group consisting of lower urinary tract symptoms, urethral dyssynergia, bladder instability, bladder outlet obstruction, benign prostatic hyperplasia, and gynecomastia in men.
4. The method according to
claim 1
wherein said cardiovascular disease is resulted from oxidized LDL in serum.
5. A method for increasing the level of enterolactone or another metabolite of hydroxymatairesinol in a person's serum thereby causing prevention of a cancer or a certain non-cancer, hormone dependent disease in a person comprising administering to said person an effective amount of hydroxymatairesinol or a geometric isomer or a stereoisomer thereof.
6. The method according to
claim 5
wherein said cancer is selected from the group consisting of breast cancer, prostate cancer and colon cancer.
7. The method according to
claim 5
wherein said non-cancer, hormonal dependent disease is selected from the group consisting of lower urinary tract symptoms, urethral dyssynergia, benign prostatic hyperplasia, and gynecomastia in men.
8. A pharmaceutical preparation comprising an effective amount of hydroxymatairesinol or a geometric isomer or a stereoisomer thereof in combination with a pharmaceutically acceptable carrier.
9. A product comprising a liquid or solid material enriched with hydroxymatairesinol or a geometric isomer or a stereoisomer thereof, for use as additive to a food product.
10. The product according to
claim 9
, wherein said food product is selected from the group consisting of a functional food, a nutritional supplement, a nutrient, a pharmafood, a nutraceutical, a health food, a designer food or any food product.
11. The product according to
claim 9
, wherein said food product is a functional food in the form of butter, margarin, biscuits, bread, cake, candy, confectionery, yogurt or an other fermented milk product, or cereal such as muesli.
12. A food product comprising an effective amount of hydroxymatairesinol or a geometric isomer or a stereoisomer thereof.
13. The food product according to
claim 12
wherein said food product is selected from the group consisting of a functional food, a nutritional supplement, a nutrient, a pharmafood, a nutraceutical, a health food, a designer food or any food product.
14. The food product according to
claim 13
, wherein said food product is a functional food in the form of butter, margarin, biscuits, bread, cake, candy, confectionery, yogurt or an other fermented milk product, or cereal such as muesli.
15. A method for increasing the stability of a food product comprising the addition to said food product of an effective amount of hydroxymatairesinol or a geometric isomer or a stereoisomer thereof.
16. The method according to
claim 15
where the increasing of food stability comprises the inhibitition of lipid, vitamin and pigment oxidations, which cause loss of nutritional value and development of off-flavors in food.
US09/829,944 1999-03-30 2001-04-11 Use of hydroxymatairesinol for prevention of cancers, non-cancer, hormone dependent diseases and cardiovascular diseases by hydroxymatairesinol, and a pharmaceutical preparation, food additive and food product comprising hydroxymatairesinol Abandoned US20010016590A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US09/829,944 US20010016590A1 (en) 1999-03-30 2001-04-11 Use of hydroxymatairesinol for prevention of cancers, non-cancer, hormone dependent diseases and cardiovascular diseases by hydroxymatairesinol, and a pharmaceutical preparation, food additive and food product comprising hydroxymatairesinol
US09/972,850 US6689809B2 (en) 1999-03-30 2001-10-10 Food additive or product or a pharmaceutical preparation, comprising hydroxymatairesinol
US10/639,530 US7005447B2 (en) 1999-03-30 2003-08-13 Food product comprising hydroxymatairesinol

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US09/281,094 US6451849B1 (en) 1999-03-30 1999-03-30 Use of hydroxymatairesinol for prevention of cancers, non-cancer, hormone dependent diseases and cardiovascular diseases by hydroxymatairesinol, and a pharmaceutical preparation, food additive and food product comprising hydroxymatairesinol
US09/829,944 US20010016590A1 (en) 1999-03-30 2001-04-11 Use of hydroxymatairesinol for prevention of cancers, non-cancer, hormone dependent diseases and cardiovascular diseases by hydroxymatairesinol, and a pharmaceutical preparation, food additive and food product comprising hydroxymatairesinol

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/281,094 Continuation US6451849B1 (en) 1999-03-30 1999-03-30 Use of hydroxymatairesinol for prevention of cancers, non-cancer, hormone dependent diseases and cardiovascular diseases by hydroxymatairesinol, and a pharmaceutical preparation, food additive and food product comprising hydroxymatairesinol

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US09/972,850 Continuation-In-Part US6689809B2 (en) 1999-03-30 2001-10-10 Food additive or product or a pharmaceutical preparation, comprising hydroxymatairesinol

Publications (1)

Publication Number Publication Date
US20010016590A1 true US20010016590A1 (en) 2001-08-23

Family

ID=23075932

Family Applications (2)

Application Number Title Priority Date Filing Date
US09/281,094 Expired - Lifetime US6451849B1 (en) 1999-03-30 1999-03-30 Use of hydroxymatairesinol for prevention of cancers, non-cancer, hormone dependent diseases and cardiovascular diseases by hydroxymatairesinol, and a pharmaceutical preparation, food additive and food product comprising hydroxymatairesinol
US09/829,944 Abandoned US20010016590A1 (en) 1999-03-30 2001-04-11 Use of hydroxymatairesinol for prevention of cancers, non-cancer, hormone dependent diseases and cardiovascular diseases by hydroxymatairesinol, and a pharmaceutical preparation, food additive and food product comprising hydroxymatairesinol

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US09/281,094 Expired - Lifetime US6451849B1 (en) 1999-03-30 1999-03-30 Use of hydroxymatairesinol for prevention of cancers, non-cancer, hormone dependent diseases and cardiovascular diseases by hydroxymatairesinol, and a pharmaceutical preparation, food additive and food product comprising hydroxymatairesinol

Country Status (25)

Country Link
US (2) US6451849B1 (en)
EP (1) EP1165537B1 (en)
JP (2) JP4852685B2 (en)
KR (2) KR100741723B1 (en)
CN (1) CN1146553C (en)
AT (1) ATE231500T1 (en)
AU (1) AU767691B2 (en)
BG (1) BG65380B1 (en)
BR (1) BR0007187A (en)
CA (2) CA2650297C (en)
CZ (2) CZ302730B6 (en)
DE (1) DE60001271T2 (en)
DK (1) DK1165537T3 (en)
EE (1) EE200100507A (en)
ES (1) ES2189738T3 (en)
HK (1) HK1045992B (en)
HU (1) HUP0200530A3 (en)
MX (1) MXPA01009714A (en)
NO (4) NO330135B1 (en)
NZ (1) NZ512099A (en)
PL (1) PL199892B1 (en)
RU (1) RU2241453C2 (en)
SK (2) SK287001B6 (en)
WO (1) WO2000059946A1 (en)
ZA (1) ZA200104440B (en)

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003066556A1 (en) * 2002-02-05 2003-08-14 Hormos Medical Corporation Lignan derivatives
US20030162224A1 (en) * 2001-11-12 2003-08-28 Arnon Chait Characterization of molecules
US20030180436A1 (en) * 2002-03-22 2003-09-25 Pizzey Glenn Roy High lignan flaxseed product and product by process
US20040229375A1 (en) * 2001-08-16 2004-11-18 Analiza, Inc. Method for measuring solubility
US20050249857A1 (en) * 2002-08-29 2005-11-10 Tomi Jarvinen Lignan complexes
US20060210691A1 (en) * 2005-03-15 2006-09-21 Pizzey Glenn R Methods of increasing flaxseed hull recovery and resultant flax products
US20070117864A1 (en) * 2003-12-26 2007-05-24 Tokyo University Of Agr. & Tech. Tlo Co., Ltd. Composition for preventing and treating hepatoma
US20080050831A1 (en) * 2003-06-12 2008-02-28 Analiza, Inc. Systems and methods for characterization of molecules
US20080124414A1 (en) * 2003-11-12 2008-05-29 Willfoer Stefan Use Of Knotwood Extracts
US20090143483A1 (en) * 2004-02-03 2009-06-04 Kotosugi Inc. Therapeutic or preventive drug for osteoporosis comprising isotaxiresinol derived from taxus yunnanensis
US8437964B2 (en) 2005-12-19 2013-05-07 Analiza, Inc. Systems and methods involving data patterns such as spectral biomarkers
US9678076B2 (en) 2014-06-24 2017-06-13 Analiza, Inc. Methods and devices for determining a disease state
US10613087B2 (en) 2012-08-10 2020-04-07 Analiza, Inc. Methods and devices for analyzing species to determine diseases
US11796544B1 (en) 2021-10-28 2023-10-24 Analiza, Inc. Partitioning systems and methods for determining multiple types of cancers

Families Citing this family (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6271257B1 (en) * 2000-04-17 2001-08-07 Hormos Nutraceutical Oy Ltd. Decreasing the intracellular level of β-catenin by administering hydroxymatairesinol
WO2002034277A1 (en) * 2000-10-23 2002-05-02 Council Of Scientific And Industrial Research Pharmaceutical composition comprising wikstromol and/or matairesinol, its use as hepatoprotectant and process for their isolation from cedrus deodara
FI111638B (en) 2001-11-23 2003-08-29 Hormos Nutraceutical Oy Ltd Method for production of a hydroxymatairesinol complex from wood
US7008666B2 (en) * 2001-11-26 2006-03-07 Hormos Nutraceutical Oy Ltd. Method of inhibiting overactivity of phagocytes or lymphocytes in an individual
US20030144216A1 (en) * 2002-01-25 2003-07-31 Mikko Unkila Method for prevention of diseases in coeliac patients
FI20021184A (en) 2002-06-19 2003-12-20 Hormos Nutraceutical Oy Ltd Lignaanivalmisteita
FR2851919A1 (en) * 2003-03-03 2004-09-10 Lmd Use of new and known lignans and neolignans as cathepsin inhibitors, for treatment of cancers, hepatic protectors, preservation of human and animal organs, Alzheimer's, and cosmetic anti-aging preparations
FI116727B (en) 2003-11-12 2006-02-15 Arbonova Ab Oy New use of branch knot extracts
US20080057140A1 (en) * 2005-01-10 2008-03-06 Mikko Unkila Use of a Lignan for the Manufacture of a Composition for Preventing or Alleviating of Symptoms Relating to Estrogen Deficiency
DE102006008772A1 (en) * 2006-02-22 2007-08-23 Beiersdorf Ag Use of hydroxymatairesinol to prepare cosmetic or dermatological formulation for the protection of sensitive and dry skin, and to increase cerium amide biosynthesis and strengthen the barrier function of the skin
DE102006019044A1 (en) * 2006-04-25 2007-10-31 Merck Patent Gmbh antioxidants
FI20106293A0 (en) * 2010-12-06 2010-12-06 Emilia Peuhu New pharmaceutical compositions
EP2517574B1 (en) 2011-04-29 2015-11-11 Symrise AG Specific vanillyl lignanes and their use as taste enhancers
RU2510268C1 (en) * 2012-11-14 2014-03-27 ОБЩЕСТВО С ОГРАНИЧЕННОЙ ОТВЕТСТВЕННОСТЬЮ "Медресурс" Agent for treating oestrogen-dependent cancer
WO2015173345A1 (en) 2014-05-15 2015-11-19 Linnea S.A. Composition comprising 7-hydroxymatairesinol
CZ2015262A3 (en) * 2015-04-20 2016-03-16 Výzkumný ústav potravinářský Praha, v.v.i. Treatment process of branch knots with exactly regulated structure of crushed material for the production of lignans and apparatus for making the same used in food production
US9669006B2 (en) 2015-07-28 2017-06-06 U.S. Nutraceuticals, LLC Composition and method to treat and alleviate symptoms of hot flashes in a female subject
EP3386524A1 (en) * 2015-12-09 2018-10-17 Oy Granula Ab Ltd The use of a composition for lowering cholesterol level in a mammal, a method for its preparation, a composition and a method for preparing food additive comprising said composition
IT201700050994A1 (en) * 2017-05-11 2018-11-11 Linnea Sa Use of a Composition comprising 7-Hydroxymato-salinol

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB8628228D0 (en) * 1986-11-26 1986-12-31 Inst Biolog Morya Dalnevostoch Composition inhibiting pathological addiction to alcohol
US4808674A (en) * 1987-08-24 1989-02-28 General Electric Company Aryl ester-grafted polyphenylene ethers and phenylene ether-amide graft copolymers prepared therefrom
DE4317466A1 (en) 1993-05-26 1994-12-01 Degussa Improved process for bleaching wood pulp
EP0797564B1 (en) 1995-10-18 2001-06-27 Kanoldt Arzneimittel Gmbh Lignans, a process for their production and pharmaceutical compositions and uses thereof
WO1997032593A2 (en) 1996-03-08 1997-09-12 Energiser Plc Composition containing iso-flavonoids and lignans
US5846944A (en) 1996-04-04 1998-12-08 The University Of Saskatchewan Purified SDG as an antioxidant
JP2987365B1 (en) * 1998-10-29 1999-12-06 かどや製油株式会社 Antioxidant
CN1893939B (en) * 2003-12-26 2011-12-28 农工大Tlo株式会社 Composition for preventing and treating hepatoma

Cited By (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040229375A1 (en) * 2001-08-16 2004-11-18 Analiza, Inc. Method for measuring solubility
US8932862B2 (en) 2001-08-16 2015-01-13 Analiza, Inc. Method for measuring solubility
US7968350B2 (en) 2001-11-12 2011-06-28 Analiza, Inc. Characterization of molecules
US20030162224A1 (en) * 2001-11-12 2003-08-28 Arnon Chait Characterization of molecules
US8211714B2 (en) 2001-11-12 2012-07-03 Analiza, Inc. Characterization of molecules
US20110218738A1 (en) * 2001-11-12 2011-09-08 Analiza, Inc. Characterization of molecules
US20050101541A1 (en) * 2002-02-05 2005-05-12 Patrik Eklund Lignan derivatives
WO2003066556A1 (en) * 2002-02-05 2003-08-14 Hormos Medical Corporation Lignan derivatives
US7528166B2 (en) 2002-02-05 2009-05-05 Hormos Medical Corporation Lignan derivatives
US20030180436A1 (en) * 2002-03-22 2003-09-25 Pizzey Glenn Roy High lignan flaxseed product and product by process
US7048960B2 (en) 2002-03-22 2006-05-23 Glenn Roy Pizzey High lignan flaxseed product and product by process
US20050249857A1 (en) * 2002-08-29 2005-11-10 Tomi Jarvinen Lignan complexes
US8099242B2 (en) * 2003-06-12 2012-01-17 Analiza, Inc. Systems and methods for characterization of molecules
US20080050831A1 (en) * 2003-06-12 2008-02-28 Analiza, Inc. Systems and methods for characterization of molecules
US11422135B2 (en) 2003-06-12 2022-08-23 Analiza, Inc. Systems and methods for characterization of molecules
US9354229B2 (en) 2003-06-12 2016-05-31 Analiza, Inc. Systems and methods for characterization of molecules
US7976877B2 (en) * 2003-11-12 2011-07-12 Oy Arbonova Ab Use of knotwood extracts
US20080124414A1 (en) * 2003-11-12 2008-05-29 Willfoer Stefan Use Of Knotwood Extracts
US20070117864A1 (en) * 2003-12-26 2007-05-24 Tokyo University Of Agr. & Tech. Tlo Co., Ltd. Composition for preventing and treating hepatoma
US20090143483A1 (en) * 2004-02-03 2009-06-04 Kotosugi Inc. Therapeutic or preventive drug for osteoporosis comprising isotaxiresinol derived from taxus yunnanensis
US7666913B2 (en) 2004-02-03 2010-02-23 Kotosugi Inc. Method of treating or preventing osteoporosis using isotaxiresinol derived from Taxus yunnanensis
US7595078B2 (en) 2005-03-15 2009-09-29 Glanbia Nutritionals Ireland Limited Methods of increasing flaxseed hull recovery and resultant flax products
US20060210691A1 (en) * 2005-03-15 2006-09-21 Pizzey Glenn R Methods of increasing flaxseed hull recovery and resultant flax products
US8437964B2 (en) 2005-12-19 2013-05-07 Analiza, Inc. Systems and methods involving data patterns such as spectral biomarkers
US10613087B2 (en) 2012-08-10 2020-04-07 Analiza, Inc. Methods and devices for analyzing species to determine diseases
US9678076B2 (en) 2014-06-24 2017-06-13 Analiza, Inc. Methods and devices for determining a disease state
US11796544B1 (en) 2021-10-28 2023-10-24 Analiza, Inc. Partitioning systems and methods for determining multiple types of cancers

Also Published As

Publication number Publication date
NZ512099A (en) 2004-01-30
MXPA01009714A (en) 2003-06-24
SK13262001A3 (en) 2002-02-05
CN1146553C (en) 2004-04-21
NO330135B1 (en) 2011-02-21
HUP0200530A3 (en) 2002-10-28
NO332110B1 (en) 2012-06-25
AU767691B2 (en) 2003-11-20
CN1345315A (en) 2002-04-17
NO20111291L (en) 2001-09-25
KR100741723B1 (en) 2007-07-23
NO20101701L (en) 2001-09-25
WO2000059946A1 (en) 2000-10-12
ES2189738T3 (en) 2003-07-16
NO331188B1 (en) 2011-10-31
HK1045992A1 (en) 2002-12-20
BG105856A (en) 2002-04-30
NO20014639D0 (en) 2001-09-25
EE200100507A (en) 2002-12-16
NO332251B1 (en) 2012-08-06
JP4852685B2 (en) 2012-01-11
SK287749B6 (en) 2011-08-04
HUP0200530A2 (en) 2002-07-29
JP2002541158A (en) 2002-12-03
ZA200104440B (en) 2002-07-30
CA2371839A1 (en) 2000-10-12
EP1165537A1 (en) 2002-01-02
CZ20013486A3 (en) 2002-02-13
KR20010108434A (en) 2001-12-07
DE60001271T2 (en) 2003-07-31
SK287001B6 (en) 2009-09-07
AU3169200A (en) 2000-10-23
CA2650297A1 (en) 2000-10-12
PL350367A1 (en) 2002-12-02
EP1165537B1 (en) 2003-01-22
JP2011052024A (en) 2011-03-17
DK1165537T3 (en) 2003-05-12
RU2241453C2 (en) 2004-12-10
ATE231500T1 (en) 2003-02-15
KR100741724B1 (en) 2007-07-23
WO2000059946A8 (en) 2001-01-25
CA2371839C (en) 2011-10-04
CZ301725B6 (en) 2010-06-02
KR20070027770A (en) 2007-03-09
NO20014639L (en) 2001-09-25
BR0007187A (en) 2002-02-19
BG65380B1 (en) 2008-05-30
DE60001271D1 (en) 2003-02-27
CZ302730B6 (en) 2011-10-05
CA2650297C (en) 2012-05-15
US6451849B1 (en) 2002-09-17
NO20101067L (en) 2001-09-25
PL199892B1 (en) 2008-11-28
HK1045992B (en) 2005-02-25

Similar Documents

Publication Publication Date Title
US6689809B2 (en) Food additive or product or a pharmaceutical preparation, comprising hydroxymatairesinol
US6451849B1 (en) Use of hydroxymatairesinol for prevention of cancers, non-cancer, hormone dependent diseases and cardiovascular diseases by hydroxymatairesinol, and a pharmaceutical preparation, food additive and food product comprising hydroxymatairesinol
Saarinen et al. Hydroxymatairesinol, a novel enterolactone precursor with antitumor properties from coniferous tree (Picea abies)
Adlercreutz Lignans and human health
Setchell et al. Mammalian Lignans and Phytooestrogens Recent Studies on their Formation, Metabolism and Biological Role in Health and
US8450364B2 (en) Use of equol for treating androgen mediated diseases
Kangas et al. Antioxidant and antitumor effects of hydroxymatairesinol (HM-3000, HMR), a lignan isolated from the knots of spruce
Andlauer et al. Does cereal reduce the risk of cancer?
Saarinen et al. Structural determinants of plant lignans for growth of mammary tumors and hormonal responses in vivo
Adlercreutz Lignans and Breast Cancer
Saarinen et al. Anticancer effects of lignans

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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