WO1999066929A1 - Compositions et methodes de traitement d'une cholesterolemie elevee - Google Patents

Compositions et methodes de traitement d'une cholesterolemie elevee Download PDF

Info

Publication number
WO1999066929A1
WO1999066929A1 PCT/US1999/013887 US9913887W WO9966929A1 WO 1999066929 A1 WO1999066929 A1 WO 1999066929A1 US 9913887 W US9913887 W US 9913887W WO 9966929 A1 WO9966929 A1 WO 9966929A1
Authority
WO
WIPO (PCT)
Prior art keywords
geranylgeranyl
geranylgeraniol
hmg
coa reductase
reductase inhibitor
Prior art date
Application number
PCT/US1999/013887
Other languages
English (en)
Inventor
Edward M. Scolnick
Original Assignee
Merck & Co., Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from GBGB9817167.1A external-priority patent/GB9817167D0/en
Application filed by Merck & Co., Inc. filed Critical Merck & Co., Inc.
Priority to EP99930451A priority Critical patent/EP1089731A4/fr
Priority to JP2000555615A priority patent/JP2002518448A/ja
Priority to AU46989/99A priority patent/AU754767B2/en
Priority to CA002335366A priority patent/CA2335366A1/fr
Publication of WO1999066929A1 publication Critical patent/WO1999066929A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/215Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids
    • A61K31/22Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acyclic acids, e.g. pravastatin
    • A61K31/221Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acyclic acids, e.g. pravastatin with compounds having an amino group, e.g. acetylcholine, acetylcarnitine
    • 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
    • A61K31/35Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom
    • A61K31/351Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom not condensed with another ring
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • 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
    • A61K31/403Heterocyclic 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 condensed with carbocyclic rings, e.g. carbazole
    • A61K31/404Indoles, e.g. pindolol
    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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

Definitions

  • compositions and methods for treating elevated blood cholesterol in a mammal while counteracting potential adverse side effects such as myopathy comprise the combination of a pharmaceutically effective amount of a 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitor (hereafter "HMG-CoA reductase inhibitor”) and a geranylgeraniol compound to a mammal in need thereof.
  • HMG-CoA reductase inhibitor 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitor
  • geranylgeraniol compound a mammal in need thereof.
  • LDL low density lipoprotein
  • HDL high density lipoprotein
  • this therapy is not easy to administer or tolerate and was therefore often unsuccessful except in specialist lipid clinics.
  • the fibrates produce a moderate reduction in LDL cholesterol accompanied by increased HDL cholesterol and a substantial reduction in triglycerides, and because they are well tolerated these drugs have been more widely used.
  • Probucol produces only a small reduction in LDL cholesterol and also reduces HDL cholesterol, which, because of the strong inverse relationship between HDL cholesterol level and CHD risk, is generally considered undesirable.
  • lovastatin the first inhibitor of HMG-CoA reductase to become available for prescription in 1987, for the first time physicians were able to obtain large reductions in plasma cholesterol with very few adverse effects.
  • HMG-CoA reductase inhibitors can cause potentially adverse side effects such as myopathy and related disorders in a small percentage of patients.
  • Myopathy is characterized by muscle pain and weakness.
  • Geranylgeraniol and its derivatives belong to a class of naturally- occurring compounds known as terpenes. Terpenes are constructed of multiples of five-carbon isoprene units. See Lehninger, A.L., Biochemistry, 1975, pp. 296 and 682-683, which is incorporated by reference herein in its entirety.
  • geranylgeraniol is a linear terpene containing four isoprene units, corresponding to the following chemical structure.
  • the geranylgeraniol derivative, geranylgeranyl pyrophosphate is an intermediate in the cholesterol biosynthetic pathway and is a substrate in the prenylation of proteins. See J.A. Glomset et 1., Geranylgeranylated proteins, Biochem-Soc-Trans., 1992 May, 20(2): 479-484, which is incorporated by reference herein in its entirety. Certain of these proteins, for example the small GTPases Rac, Rho, and Cdc42, regulate cytoskeletal function.
  • geranylgeraniol In cell cultures, geranylgeraniol is found to block apoptosis, i.e. programmed cell death that can be induced by an HMG-CoA reductase inhibitor.
  • geranylgeraniol and its derivatives have not previously been investigated either in vitro or in vivo for their ability to mitigate the potentially adverse myopathy side effects that can be associated with HMG-CoA reductase inhibitor therapy for treating or preventing elevated blood cholesterol.
  • the combination of an HMG- CoA reductase inhibitor and a geranylgeraniol compound is effective for treating or preventing elevated blood cholesterol while mitigating the potentially adverse myopathy side effects that can be associated with the therapy.
  • the combination has the advantage of providing increased safety and better patient compliance, which should maximize therapeutic efficacy.
  • the geranylgeraniol compound blocks the potentially harmful effect of the HMG- CoA reductase inhibitor on muscle cells.
  • the geranylgeraniol compound is believed to interfere with apoptosis, or functional impair due to reduced geranylgeranylation, which can potentially be induced in muscle cells by the HMG- CoA reductase inhibitor.
  • compositions comprising the combination of an HMG-CoA reductase inhibitor and a geranylgeraniol compound. It is another object of the present invention to provide methods for treating or preventing elevated blood cholesterol in a mammal, particularly wherein said mammal is a human.
  • the present invention relates to a pharmaceutical composition comprising an HMG-CoA reductase inhibitor and a geranylgeraniol compound.
  • the present invention relates to a pharmaceutical composition comprising a pharmaceutically-effective amount of an HMG-CoA reductase inhibitor and an amount of a geranylgeraniol compound effective to counteract HMG-CoA reductase inhibitor-associated myopathy.
  • the present invention relates to a method for treating or preventing elevated blood cholesterol in a mammal in need thereof comprising administering an HMG-CoA reductase inhibitor and a geranylgeraniol compound.
  • the present invention relates to a method for treating or preventing elevated blood cholesterol in a mammal in need thereof comprising sequentially administering a geranylgeraniol compound and an HMG-CoA reductase inhibitorn.
  • the present invention relates to the use of a composition in the manufacture of a medicament for treating or preventing elevated blood cholesterol in a mammal in need thereof, said composition comprising an HMG- Co A reductase inhibitor and a geranylgeraniol compound.
  • the present invention relates to the use of a composition comprising an HMG-CoA reductase inhibitor and a geranylgeraniol compound for treating or preventing elevated blood cholesterol in a mammal in need thereof. All percentages and ratios used herein, unless otherwise indicated, are by weight.
  • the invention hereof can comprise, consist of, or consist essentially of the essential as well as optional ingredients, components, and methods described herein.
  • Figure 1 shows that activation of Mstl cleavage by 10 ⁇ M lovastatin is blocked by geranylgeraniol.
  • Osteoclast-like cells are purified from cocultures by sequential treatment of culture dishes with collagenase and EDTA. Cells are then treated for 17 hours with lovastatin. Cell lysates are made and then analyzed by an in- gel kinase assay using myelin basic protein as a substrate.
  • Lane 1 is a no-treatment control.
  • Lane 2 shows treatment with 10 ⁇ M lovastatin.
  • Lane 3 shows treatment with the combination of 10 ⁇ M Lovastatin and 10 ⁇ M geranylgeraniol.
  • the present invention relates to compositions and methods for treating or preventing elevated blood cholesterol in a mammal in need of such treatment, while counteracting the occurence of adverse myopathy effects.
  • the compositions comprise a pharmaceutically effective amount of an HMG-CoA reductase inhibitor and a pharmaceutically effective amount of a geranylgeraniol compound.
  • pharmaceutically effective amount means that amount of the HMG-CoA reductase inhibitor or geranylgeraniol compound that will elicit the desired therapeutic effect or response or provide the desired benefit when administered in accordance with the desired treatment regimen.
  • a prefered pharmaceutically effective amount of the HMG-CoA reductase inhibitor is an amount that is effective for treating or preventing elevated blood cholesterol.
  • a preferred pharmaceutically effective amount of the geranylgeraniol compound is an amount that will block or mitigate the occurrence of adverse myopathy effects, while not blocking, or only minimally blocking, the therapeutic blood cholesterol effects of the HMG-CoA reductase inhibitor.
  • counteracting the occurence of adverse myopathy effects means preventing, decreasing, or lessening the occurrence of unwanted side effects in the muscular effects, relative to treatment with a HMG-CoA reductase inhibitor alone.
  • the term "until the desired therapeutic effect is achieved and/or maintained”, as used herein, means that the therapeutic agent or agents are continuously administered, according to the dosing schedule chosen, up to the time that the clinical or medical effect sought for the disease or condition being treated is observed by the clinician or researcher.
  • the pharmaceutical composition is continuously administered until the desired change in blood cholesterol is observed. In such instances, achieving a decrease in blood cholesterol is a desried objective.
  • the pharmaceutical composition is continuously administered for as long as necessary to prevent the undesired condition. In such instances, maintenance of blood cholesterol level is often an objective as well as prevention of or reducing the risk of developing atherosclerotic disease or cardiovascular disorders such as heart attack and stroke.
  • compositions of the present invention are provided.
  • compositions of the present invention comprise a pharmaceutically effective amount of an HMG-CoA reductase inhibitor and a pharmaceutically effective amount of a geranylgeraniol compound. These compositions are useful for treating or preventing elevated blood cholesterol in a mammal in need thereof while counteracting the potentially adverse effects, such as myopathy, that can be associated with the administration of the HMG-CoA reductase inhibitor.
  • compositions herein comprise a compound which inhibits the enzyme, HMG-CoA reductase.
  • Compounds which have inhibitory activity for HMG- CoA reductase can be readily identified by using assays well-known in the art. See U.S. Patent No. 4,231,938, to Monoghan et al., issued November 4, 1980 and U.S. Patent No. 5,354,772, to Kathawal, issued October 11, 1994, both of which are incorporated by reference herein in their entirety.
  • HMG-CoA reductase inhibitors examples include but are not limited to lovastatin (MEVACOR®; see U.S. Patent No.
  • simvastatin ZOCOR®; see U.S. Patent No. 4,444,784, to Hoffman et al., issued April 24, 1984
  • pravastatin PRAVACHOL®; see U.S. Patent No. 4,346,227, to Terahara et al., issued August 24, 1982
  • fluvastatin LESCOL®; see U.S. Patent No. 5,354,772, already cited above and incorporated by reference herein
  • atorvastatin LIPITOR®; see U.S. Patent No. 5,273,995, to Roth, issued December 28, 1993
  • cerivastatin also known as rivastatin; see U.S. Patent No.
  • HMG-CoA reductase inhibitor is intended to include all pharmaceutically acceptable lactone and open acid (that is where the lactone ring is opened to form the free acid), as well as salt and ester forms of compounds which have HMG-CoA reductase inhibitory activity, and therefor the use of such lactone, open acid, salt, and ester forms is included within the scope of this invention.
  • the HMG-CoA reductase inhibitor is selected from the group consisting of lovastatin, simvastatin, pravastatin, fluvastatin, atorvastatin, cerivastatin, mevastatin, and the pharmaceutically acceptable lactones, open acids, salts, and esters thereof, and mixtures thereof.
  • the HMG-CoA reductase inhibitor is selected from the group consisting of lovastatin, simvastatin, pravastatin, fluvastatin, atorvastatin, cerivastatin, and the pharmaceutically acceptable lactones, open acids, salts, and esters thereof, and mixtures thereof. More preferably, the HMG-CoA reductase inhibitor is selected from the group consisting of lovastatin, simvastatin, and the pharmaceutically acceptable lactones, open acids, salts, and esters thereof, and mixtures thereof.
  • HMG-CoA reductase inhibitors can be represented by the chemical formula
  • Z is selected from the group consisting of: a)
  • R2 is selected from the group consisting of C1-C3 alkyl, hydroxy, oxo, and C1-C3 hydroxy substituted alkyl,
  • R3 is selected from the group consisting of hydrogen, hydroxy, C1-C3 alkyl, and Ci- C3 hydroxy substituted alkyl, a, b, c, and d are all single bonds, or a and c are double bonds, or b and d are double bonds, or one of a, b, c, and d is a double bond, and n is O, 1, or 2; b)
  • X is selected from the group consisting of N[CH(CH3)2] and CH(CH2)3CH3
  • R4 and R5 are each independently selected from the group consisting of hydrogen, fluorine, chlorine, bromine, iodine, C1-C4 alkyl, C1-C4 alkoxy, and trifluoromethyl
  • R6, R7, R8, and R9 are each independently selected from the group consisting of hydrogen, fluorine, chlorine, bromine, iodine, C1-C4 alkyl, and C1-C4 alkoxy.
  • pharmaceutically acceptable salts as used herein in referring to the HMG-CoA reductase inhibitors shall mean non-toxic salts of the compounds employed in this invention which are generally prepared by reacting the free acid with a suitable organic or inorganic base.
  • salt forms of HMG-CoA reductase inhibitors include, but are not limited to, acetate, benzenesulfonate, benzoate, bicarbonate, bisulfate, bitartrate, borate, bromide, calcium, calcium edetate, camsylate, carbonate, chloride, clavulanate, citrate, dihydrochloride, edetate, edisylate, estolate, esylate, fumarate, gluceptate, gluconate, glutamate, glycollylarsanilate, hexylresorcinate, hydrabamine, hydrobromide, hydrochloride, hydroxynapthoate, iodide, isothionate, lactate, lactobionate, laurate, malate, maleate, mandelate, mesylate, methylbromide, methylnitrate, methylsulfate, mucate, napsylate, nitrate, ole
  • esters as used herein in referring to the HMG-CoA reductase inhibitors is used in its standard meaning to denote the condensation product of a carboxylic acid and an alcohol. Ester derivatives of the described compounds can function as prodrugs which, when absorbed into the bloodstream of a warm-blooded animal, can cleave in such a manner as to release the drug form and permit the drug to afford improved therapeutic efficacy.
  • lactones is used herein in referring to the HMG-CoA reductase inhibitors is used in its standard meaning to denote a cyclic condensation product of a carboxylic acid and an alcohol, i.e. a cyclic ester.
  • open acid is used herein in referring to the HMG-CoA reductase inhibitors to denote that the lactone ring is open, i.e. uncyclized, to form the free acid. It is recognized that mixtures of two or more HMG-CoA reductase inhibitors can be utilized.
  • the dosage regimen utilizing a HMG-CoA reductase inhibitor is selected in accordance with a variety of factors including type, species, age, weight, sex and medical condition of the patient; the severity of the condition to be treated; the route of administration; the renal and hepatic function of the patient; and the particular compound or salt or ester thereof employed. A consideration of these factors is well within the purview of the ordinarily skilled clinician for the purpose of determining the therapeutically effective or prophylactically effective dosage amounts needed to prevent, counter, or arrest the progress of the condition.
  • patient includes mammals, especially humans, who take an HMG-CoA reductase inhibitor or combination for any of the uses described herein.
  • Administering of the drug or drugs to the patient includes both self-administration and administration to the patient by another person.
  • the precise dosage of the HMG-CoA reductase inhibitor will vary with the dosing schedule, the particular compound chosen, the age, size, sex and condition of the mammal or human, the nature and severity of the disorder to be treated, and other relevant medical and physical factors.
  • a precise pharmaceutically effective amount cannot be specified in advance and can be readily determined by the caregiver or clinician. Appropriate amounts can be determined by routine experimentation from animal models and human clinical studies.
  • the amounts of the HMG-CoA reductase inhibitor can be the same or similar to those amounts which are employed for anti- hypercholesterolemic treatment and which are described in the Physicians' Desk Reference (PDR), 52 nd Ed. of the PDR, 1998 (Medical Economics Co), which is incorporated by reference herein in its entirety.
  • the doses can be the same or similar to those amounts which are known in the art.
  • the HMG-CoA reductase inhibitors can be administered via a wide variety of routes including oral administration, intravenous administration, intranasal administration, injections, ocular administration, and the like.
  • a preferred route of delivery is oral administration.
  • Oral dosage amounts of the HMG-CoA reductase inhibitor are from about 1 to 200 mg/day, and more preferably from about 5 to 160 mg/day. However, dosage amounts will vary depending on the potency of the specific HMG-CoA reductase inhibitor used as well as other factors as noted above.
  • An HMG-CoA reductase inhibitor which has sufficiently greater potency may be given in sub- milligram daily dosages.
  • the HMG-CoA reductase inhibitor may be administered from 1 to 4 times per day, and preferably once per day.
  • the daily dosage amount for simvastatin can be selected from 5 mg, 10 mg, 20 mg, 40 mg, and 80 mg; for lovastatin, 10 mg, 20 mg, 40 mg and 80 mg; for fluvastatin sodium, 20 mg, 40 mg and 80 mg; for pravastatin sodium, 10 mg, 20 mg, and 40 mg; and for atorvastatin calcium, 10 mg, 20 mg, and 40 mg.
  • compositions of the present invention comprise a pharmaceutically effective amount of a geranylgeraniol compound.
  • RlO is selected from the group consisting of H (i.e. geranylgeraniol), C1-C30 alkyl (including straight, branched, and cyclic alkyl), C2-C30 alkenyl (including straight, branched, and cyclic alkenyl), C2-C30 alkynyl (including straight, branched, and cyclic alkynyl), C5-C14 aryl, PO3H2 (i.e. geranylgeranyl phosphate), P2O7H3
  • RU is selected from the group consisting of H, C1-C10 alkyl (including straight, branched, and cyclic alkyl), C2-C10 alkenyl (including straight, branched, and cyclic alkenyl), C2-C10 alkynyl (including straight, branched, and cyclic alkynyl), C2-C10 hydroxy-substituted alkyl (including straight, branched, and cyclic), C2-C10 amino-substituted alkyl
  • n is an integer from 0 to 3.
  • aryl refers to a monocyclic or polycyclic system comprising at least one aromatic ring, wherein the monocyclic or polycyclic system contains 0, 1, 2, 3, or 4 heteroatoms chosen from N, O, or S, and wherein the monocyclic or polycyclic system is either unsubstituted or substituted with one or more groups independently selected from hydrogen, halogen, Ci-io alkyl, C3-8 cycloalkyl, aryl, aryl Ci-8 alkyl, amino, amino Ci-8 alkyl, Ci-3 acylamino, C ⁇ _3 acylamino Ci -8 alkyl, Ci-6 alkylamino, Ci-6 alkylamino Ci-8 alkyl, Ci-6 dialkylamino, Ci-6 dialkylamino-Ci-8 alkyl, C1.4 alkoxy, Ci-4 alkoxy Ci-6 alkyl, hydroxycarbonyl, hydroxycarbonyl Ci-6 alkyl, Ci-5 alkoxycarbonyl, Ci-3 al
  • aryl examples include, but are not limited to, phenyl, naphthyl, pyridyl, pyrazinyl, pyrimidinyl, imidazolyl, benzimidazolyl, indolyl, thienyl, furyl, dihydrobenzofuryl, benzo(l,3) dioxolane, oxazolyl, isoxazolyl and thiazolyl.
  • esters are also intended to encompass esters of substituted acids such as lactic acid, amino acids, and other complex acids, and mono and higher esters of di and higher carboxylic acids such as succinic acid and glutaric acid.
  • Preferred geranylgerniol compounds are selected from the group consisting of geranylgeraniol, geranylgeranyl ethyl ether, geranylgeranyl phosphate, geranylgeranyl pyrophosphate, geranylgeranyl acetate, geranylgeranyl propionate, geranylgeranyl benzoate, geranylgeranyl lactate, geranylgeranyl succinate, geranylgeranyl glutarate, and mixtures thereof.
  • geranylgeraniol compounds can be utilized.
  • the precise dosage of the geranylgeraniol compounds will also vary with the dosing schedule, the particular compound chosen, the age, size, sex and condition of the mammal or human, the nature and severity of the disorder to be treated, and other relevant medical and physical factors.
  • a precise pharmaceutically effective amount cannot be specified in advance and can be readily determined by the caregiver or clinician. Appropriate amounts can be determined by routine experimentation from animal models and human clinical studies.
  • an appropriate amount is chosen to counteract the potential adverse myopathy effects of the HMG-CoA reductase inhibitor. The amount should be below that level which will inhibit the desired cholesterol lowering effect of the HMG-CoA reductase inhibitor.
  • an effective oral dose of the geranylgernaiol compound is typically chosen so as to provide a concentration in the blood stream from about 1 ⁇ M to about 100 ⁇ M, preferably about 10 ⁇ M, although other ranges can be used.
  • Nonlimiting exemplary doses are about 1 ug/kg to about 100 ug/kg, preferably about 10 ug/kg, for a human subject.
  • human doses which can be administered are generally in the range of about 0.1 mg/day to about 10 mg/day, preferably from about 0.25 mg/day to about 5 mg/day, and more preferably from about 0.5 mg/day to about 1.5 mg/day, based on a geranylgeraniol active weight basis.
  • a typical nonlimiting dosage amount would be about 0.75 mg/day.
  • the pharmaceutical compositions herein comprise from about 0.1 mg to about 10 mg, preferably from about 0.25 mg to about 5 mg, and more preferably from about 0.5 mg to about 1.5 mg of the geranylgeraniol compound.
  • a typical nonlimiting amount for is about 0.75 mg.
  • the HMG-CoA reductase inhibitor and the geranylgeraniol compound are typically administered in admixture with suitable pharmaceutical diluents, excipients, or carriers, collectively referred to herein as "carrier materials”, suitably selected with respect to oral administration, i.e. tablets, capsules, elixirs, syrups, powders, and the like, and consistent with conventional pharmaceutical practices.
  • carrier materials suitably selected with respect to oral administration, i.e. tablets, capsules, elixirs, syrups, powders, and the like, and consistent with conventional pharmaceutical practices.
  • the active ingredient can be combined with an oral, non-toxic, pharmaceutically acceptable inert carrier such as lactose, starch, sucrose, glucose, methyl cellulose, magnesium stearate, mannitol, sorbitol, croscarmellose sodium and the like;
  • an oral, non-toxic, pharmaceutically acceptable inert carrier such as lactose, starch, sucrose, glucose, methyl cellulose, magnesium stearate, mannitol, sorbitol, croscarmellose sodium and the like
  • the oral drug components can be combined with any oral, non-toxic, pharmaceutically acceptable inert carrier such as ethanol, glycerol, water and the like.
  • suitable binders, lubricants, disintegrating agents and coloring agents can also be incorporated.
  • Suitable binders can include starch, gelatin, natural sugars such a glucose, anhydrous lactose, free-flow lactose, beta-lactose, and corn sweeteners, natural and synthetic gums, such as acacia, guar, tragacanth or sodium alginate, carboxymethyl cellulose, polyethylene glycol, waxes, and the like.
  • Lubricants used in these dosage forms include sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride and the like.
  • the compounds used in the present method can also be coupled with soluble polymers as targetable drug carriers.
  • Such polymers can include polyvinylpyrrolidone, pyran copolymer, polyhydroxylpropyl-methacrylamide, and the like.
  • the present invention comprises methods for treating or preventing elevated blood cholesterol in mammals.
  • the mammal is a human.
  • compositions and methods of the present invention are administered and caried out until the desired therapeutic effect is achieved.
  • the HMG-CoA reductase inhibitor and the geranylgeraniol compound are generally administered concurrently.
  • the HMG-CoA reductase inhibitor and the geranylgeraniol compound can be adminsitered sequentially.
  • the germanaylgeraniol compound is administered first.
  • Bone marrow cells are harvested from 6-week-old male Balb/C mice by flushing marrow spaces of freshly isolated long bones (tibiae and femora) with ⁇ -MEM (minimal essential media) containing penicillin/streptomycin (100 1.U./ml of each and 20 mM Hepes buffer).
  • the bone marrow cells are suspended in ⁇ -MEM and the cells are filtered through an approximately 70 ⁇ m cell strainer. The filtrate is centrifuged at about 300 x g for about 7 minutes. The resulting pellet is resuspended in ⁇ -MEM supplemented with fetal calf serum (10 % v/v) and 10 nM 1, 25-(OH)2 vitamin D3.
  • osteoblastic MB 1.8 cells are added to sub-confluent monolayers of osteoblastic MB 1.8 cells in cell culture plates and cultured for 7 days at 37DC in the presence of 5% CO2- Culture media is replenished ever other day. Fusion of the osteoclast precursor cells from bone marrow (with each other) to form multinucleated osteoclast-like cells typically occurs after about 7 days. Osteoclast-like cells are enriched by sequential treatment with collagenase (1 mg/rnL in phosphate buffered saline) for one hour at 37°C and EDTA (0.2 g/L in phosphate buffered saline) for 20 min at 37°C.
  • Non-adherent cells are rinsed away by washing with phosphate buffered saline.
  • Osteoclast-like cells which are resistant to the sequential treatments are present at about 95% purity and are maintained in ⁇ -MEM supplemented with fetal calf serum (10 % v/v), 10 nM 1,25- (OH)2 vitamin D3, macrophage-colony-stimulating factor (5 ng/mL).
  • the compounds to be evaluated are prepared as a solution of the desired concentration in ⁇ -MEM.
  • examples of compounds that can be evaluated include HMG-CoA reductase inhibitors such lovastatin, simvastatin, pravastatin, fluvastatin, atorvastatin, cerivastsin, mevastatin, and the pharmaceutically acceptable salts, esters, and lactones thereof, as well as compounds that block the effects of these HMG-CoA reductase inhibitors, such as geranylgeraniol compounds, for example, geranylgeraniol, geranylgeranyl ethyl ether, geranylgeranyl phosphate, geranylgeranyl pyrophosphate, geranylgeranyl acetate, geranylgeranyl propionate, geranylgeranyl benzoate, geranylgeranyl lactate, geranylgeranyl succinate, geranylgeranyl glutarate.
  • Combinations of compounds can also be evaluated.
  • the solutions of the compounds to be evaluated are added to the cultures for a time period of 17-24 hours.
  • No treatment controls (controls not treated with comcpounds) are prepared by adding equivalent volumes of ⁇ -MEM to the control dishes.
  • HEPES N-(2- hydroxyethyl)piperazine-N'-(2-ethansulfonic acid) or Tris buffer containing the following: ⁇ -glycerophosphate (50 mM); Na3VO4 (lmM); NaF (ImM); Microcystin LR (1 ⁇ M); leupeptin (10 ⁇ g/ml); aprotinin (10 ⁇ g/ml); phenylmethyl sulfonylfluoride (1 mM).
  • HEPES N-(2- hydroxyethyl)piperazine-N'-(2-ethansulfonic acid
  • Protein concentrations are determined for each lysate and 5-20 ⁇ g are loaded into each lane of a SDS-PAGE (sodium dodecyl sulfate-polyacrylamide gel electrophoresis) gel containing Myelin Basic Protein, or another kinase substrate, which has been polymerized into the gel at a concentration between 50-400 ⁇ g/ml.
  • SDS-PAGE sodium dodecyl sulfate-polyacrylamide gel electrophoresis
  • Myelin Basic Protein or another kinase substrate, which has been polymerized into the gel at a concentration between 50-400 ⁇ g/ml.
  • Molecular weight standards are also loaded into one or more lanes of the gels.
  • In-gel kinase assays are run according to a standard procedure based on Kameshita and Fujisawa, 1989 (Anal. Biochem. 183:139-143) and of Gotoh et al., 1990 (Eur
  • the proteins are electrophoresed in the above gels.
  • the gels are then successively soaked in 50 mM HEPES, pH 7.6; 5 mM 2-mercaptoethanol and each of the following (for each wash): (a) 20% isopropanol; (b) no additions; (c) urea (6 M); (d) Urea (3 M); (e) Urea (0.75 M); and Tween 20 (0.05% vol:vol).
  • Kinase reactions are then run by first soaking the gels in 20 mM HEPES, pH 7.6; 20 mM MgCl2; 2 mM DTT and then in the same buffer containing 0.02 M ATP (non-radioactive) with ca. 1000 cpm/pmol 32p_ ⁇ _ATP.
  • the gels are then washed six times with 5% trichloroacetic acid and 1% pyrophosphate.
  • the gels are then stained with Coomassie brilliant blue dye (0.125%) in 50% methanol, 10% acetic acid; destained with 30% methanol, 10% acetic acid; soaked in 2% glycerol; and dried using a gel dryer.
  • the gels are then exposed to autoradiography film for times ranging from several hours to weeks.
  • the bands observed in the autoradiographs representing the gels reflect kinase activities.
  • Mst 1 (apparent molecular weight about 59 kDa)
  • Mst 2 (apparent molecular weight about 60 kDa)
  • a 34 kDa Mst kinase fragment are observed and identified by their migration as compared to the migration of molecular weight standards.
  • the band intensities on the autoradiography film are quantitated by densitometry and comparisons between bands from untreated controls and bands from echistatin-treated cells provide the basis for the analyses.
  • All the ingredients except magnesium stearate are blended together in a suitable mixer.
  • the powder mixture is then granulated with adequate quantities of granulating solvent(s), e.g. water.
  • the wet granulated mass is dried in a suitable dryer.
  • the dried granulation is sized through a suitable screen.
  • the sized granulation is mixed with magnesium stearate before tableting.
  • the tablets may be coated if deemed necessary.
  • Additional ingredients that may be added to the above include suitable color and mixtures of colors.
  • composition is useful for treating or preventing elevated blood cholesterol.
  • the simvastatin is replaced by an HMG-CoA reductase inhibitor selected from lovastatin, pravastatin, fluvastatin, atorvastatin, cerivastatin, or mevastatin
  • the geranylgeraniol is replaced by geranylgeranyl phosphate, geranylgeranyl pyrophosphate, geranylgeranyl acetate, geranylgeranyl propionate, geranylgeranyl benzoate, geranylgeranyl lactate, geranylgeranyl succinate, or geranylgeranyl glutarate
  • the ingredients are combined and blended together and are compressed using conventional tableting techniques.
  • composition is useful for treating or preventing elevated blood cholesterol.
  • the simvastatin is replaced by an HMG-CoA reductase inhibitor selected from lovastatin, pravastatin, fluvastatin, atorvastatin, cerivastatin, or mevastatin
  • the geranylgeraniol is replaced by geranylgeranyl phosphate, geranylgeranyl pyrophosphate, geranylgeranyl acetate, geranylgeranyl propionate, geranylgeranyl benzoate, geranylgeranyl lactate, geranylgeranyl succinate, or geranylgeranyl glutarate
  • Hard gelatin capsule The dry ingredients are combined and blended together and encapsulated in a gelatin coating using standard manufacturing techniques.
  • composition is useful for treating or preventing elevated blood cholesterol.
  • simvastatin is replaced by an HMG-CoA reductase inhibitor selected from lovastatin, pravastatin, fluvastatin, atorvastatin, cerivastatin, or mevastatin
  • the geranylgeraniol is replaced by geranylgeranyl phosphate, geranylgeranyl pyrophosphate, geranylgeranyl acetate, geranylgeranyl propionate, geranylgeranyl benzoate, geranylgeranyl lactate, geranylgeranyl succinate, or geranylgeranyl glutarate
  • An oral suspension is prepared by combining the ingredients using standard formulation techniques.
  • the composition is useful for treating or preventing elevated blood cholesterol.
  • the simvastatin is replaced by an HMG-CoA reductase inhibitor selected from lovastatin, pravastatin, fluvastatin, atorvastatin, cerivastatin, or mevastatin
  • the geranylgeraniol is replaced by geranylgeranyl phosphate, geranylgeranyl pyrophosphate, geranylgeranyl acetate, geranylgeranyl propionate, geranylgeranyl benzoate, geranylgeranyl lactate, geranylgeranyl succinate, or geranylgeranyl glutarate EXAMPLE 6
  • the ingredients are combined using standard formulation techniques.
  • composition is useful for treating or preventing elevated blood cholesterol.
  • the simvastatin is replaced by an HMG-CoA reductase inhibitor selected from lovastatin, pravastatin, fluvastatin, atorvastatin, cerivastatin, or mevastatin
  • the geranylgeraniol is replaced by geranylgeranyl phosphate, geranylgeranyl pyrophosphate, geranylgeranyl acetate, geranylgeranyl propionate, geranylgeranyl benzoate, geranylgeranyl lactate, geranylgeranyl succinate, or geranylgeranyl glutarate

Abstract

L'invention concerne des compositions et des méthodes de traitement d'une cholestérolémie élevée chez un mammifère tout en évitant l'apparition d'effets secondaires indésirables tels que la myopathie. Ces compositions contiennent une combinaison d'une dose pharmaceutiquement efficace d'un inhibiteur de la 3-hydroxy-3-méthyl-glutaryle coenzyme A (HMG-CoA) réductase et d'un composé de géranylgéraniol. Ces compositions sont destinées à être administrées à un mammifère nécessitant un tel traitement.
PCT/US1999/013887 1998-06-24 1999-06-21 Compositions et methodes de traitement d'une cholesterolemie elevee WO1999066929A1 (fr)

Priority Applications (4)

Application Number Priority Date Filing Date Title
EP99930451A EP1089731A4 (fr) 1998-06-24 1999-06-21 Compositions et methodes de traitement d'une cholesterolemie elevee
JP2000555615A JP2002518448A (ja) 1998-06-24 1999-06-21 高血中コレステロールを治療する組成物および方法
AU46989/99A AU754767B2 (en) 1998-06-24 1999-06-21 Compositions and methods for treating elevated blood cholesterol
CA002335366A CA2335366A1 (fr) 1998-06-24 1999-06-21 Compositions et methodes de traitement d'une cholesterolemie elevee

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US9052798P 1998-06-24 1998-06-24
US60/090,527 1998-06-24
GB9817167.1 1998-08-06
GBGB9817167.1A GB9817167D0 (en) 1998-08-06 1998-08-06 Compositions and methods for treating elevated blood cholesterol

Publications (1)

Publication Number Publication Date
WO1999066929A1 true WO1999066929A1 (fr) 1999-12-29

Family

ID=26314174

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1999/013887 WO1999066929A1 (fr) 1998-06-24 1999-06-21 Compositions et methodes de traitement d'une cholesterolemie elevee

Country Status (5)

Country Link
EP (1) EP1089731A4 (fr)
JP (1) JP2002518448A (fr)
AU (1) AU754767B2 (fr)
CA (1) CA2335366A1 (fr)
WO (1) WO1999066929A1 (fr)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6350453B1 (en) 1999-05-24 2002-02-26 American River Nutrition, Inc. Tocotrienols and geranylgeraniol from Bixa orellana byproducts
US7229982B2 (en) 2002-07-26 2007-06-12 Moore William D Pharmaceutical formulation
WO2010126579A1 (fr) * 2009-04-27 2010-11-04 Beth Israel Deaconess Medical Center Méthodes et compositions utilisées pour le traitement et le diagnostic des myopathies provoquées par les statines
EP2465516A1 (fr) 2003-04-08 2012-06-20 Barrie Tan Compositions d'extrait de rocou, y compris les géraniols géranyl et procédés d'utilisation
US20120270933A1 (en) * 2009-02-24 2012-10-25 Madeira Therapeutics Liquid statin formulation
WO2013130654A1 (fr) * 2012-02-29 2013-09-06 Coyote Pharmaceuticals, Inc. Gga et dérivés de gga, compositions de ceux-ci et procédés pour le traitement de maladies neurodégénératives, comprenant une paralysie, les comprenant
WO2014055440A1 (fr) * 2012-10-01 2014-04-10 Coyote Pharmaceuticals, Inc. Préparation de gga et de leurs dérivés et leur co-cristallisation avec l'urée ou la thio-urée
US9119808B1 (en) 2012-10-08 2015-09-01 Coyote Pharmaceuticals, Inc. Treating neurodegenerative diseases with GGA or a derivative thereof

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4933165A (en) * 1989-01-18 1990-06-12 Merck & Co., Inc. Coenzyme Q10 with HMG-CoA reductase inhibitors
US5316765A (en) * 1989-09-07 1994-05-31 Karl Folkers Foundation For Biomedical And Clinical Research Use of coenzyme Q10 in combination with HMG-CoA reductase inhibitor therapies
US5447959A (en) * 1989-10-13 1995-09-05 Medafor Method of using derivatives of long chain fatty alcohols to treat neuronal degradation
US5574025A (en) * 1994-10-26 1996-11-12 Merck & Co., Inc. Inhibitors of prenyl-protein transferases
US5639653A (en) * 1993-07-19 1997-06-17 Albert Einstein College Of Medicine Of Yeshiva University, A Division Of Yeshiva Universtiy Method for proliferating Vγ2Vδ2 T cells
US5763646A (en) * 1997-03-13 1998-06-09 Ranbaxy Laboratories, Ltd. Process for manufacturing simvastatin from lovastatin or mevinolinic acid

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES8105781A1 (es) * 1979-07-27 1981-06-16 Sankyo Co Un procedimiento para la preparacion de sales y esteres, de monacolina k utiles como agentes antitlipercolesteremicos.
US5622985A (en) * 1990-06-11 1997-04-22 Bristol-Myers Squibb Company Method for preventing a second heart attack employing an HMG CoA reductase inhibitor
US6369103B1 (en) * 1994-01-18 2002-04-09 Bristol-Myers Squibb Company Method for preventing or reducing risk of onset of cardiovascular events employing an HMG CoA reductase inhibitor
IL112639A0 (en) * 1994-03-11 1995-05-26 Bristol Myers Squibb Co A pharmaceutical composition containing pravastin
IT1276162B1 (it) * 1995-11-23 1997-10-27 Baldacci Lab Spa Geranilgeranil-derivati,procedimento per la loro preparazione e relative composizioni farmaceutiche
JPH1087480A (ja) * 1996-09-13 1998-04-07 Eisai Co Ltd 抗動脈硬化治療剤

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4933165A (en) * 1989-01-18 1990-06-12 Merck & Co., Inc. Coenzyme Q10 with HMG-CoA reductase inhibitors
US5316765A (en) * 1989-09-07 1994-05-31 Karl Folkers Foundation For Biomedical And Clinical Research Use of coenzyme Q10 in combination with HMG-CoA reductase inhibitor therapies
US5447959A (en) * 1989-10-13 1995-09-05 Medafor Method of using derivatives of long chain fatty alcohols to treat neuronal degradation
US5639653A (en) * 1993-07-19 1997-06-17 Albert Einstein College Of Medicine Of Yeshiva University, A Division Of Yeshiva Universtiy Method for proliferating Vγ2Vδ2 T cells
US5574025A (en) * 1994-10-26 1996-11-12 Merck & Co., Inc. Inhibitors of prenyl-protein transferases
US5763646A (en) * 1997-03-13 1998-06-09 Ranbaxy Laboratories, Ltd. Process for manufacturing simvastatin from lovastatin or mevinolinic acid

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP1089731A4 *

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6350453B1 (en) 1999-05-24 2002-02-26 American River Nutrition, Inc. Tocotrienols and geranylgeraniol from Bixa orellana byproducts
US7229982B2 (en) 2002-07-26 2007-06-12 Moore William D Pharmaceutical formulation
US7718643B2 (en) 2002-07-26 2010-05-18 Schering Corporation Pharmaceutical formulation
EP2465516A1 (fr) 2003-04-08 2012-06-20 Barrie Tan Compositions d'extrait de rocou, y compris les géraniols géranyl et procédés d'utilisation
EP1624880B1 (fr) * 2003-04-08 2018-12-19 Barrie Tan Compositions d'extrait de rocou contenant des geranylgeraniols pour utilisation dans la diminution de niveau sanguin de triglyceride
US20160310463A1 (en) * 2009-02-24 2016-10-27 Peter Joiner Liquid statin formulation
US20120270933A1 (en) * 2009-02-24 2012-10-25 Madeira Therapeutics Liquid statin formulation
WO2010126579A1 (fr) * 2009-04-27 2010-11-04 Beth Israel Deaconess Medical Center Méthodes et compositions utilisées pour le traitement et le diagnostic des myopathies provoquées par les statines
US20120171286A1 (en) * 2009-04-27 2012-07-05 Beth Isreal Deaconess Medical Center Methods and compositions for the treatment and diagnosis of statin-induced myopathy
US9045403B2 (en) 2012-02-29 2015-06-02 Coyote Pharmaceuticals, Inc. Geranyl geranyl acetone (GGA) derivatives and compositions thereof
WO2013130654A1 (fr) * 2012-02-29 2013-09-06 Coyote Pharmaceuticals, Inc. Gga et dérivés de gga, compositions de ceux-ci et procédés pour le traitement de maladies neurodégénératives, comprenant une paralysie, les comprenant
WO2014055440A1 (fr) * 2012-10-01 2014-04-10 Coyote Pharmaceuticals, Inc. Préparation de gga et de leurs dérivés et leur co-cristallisation avec l'urée ou la thio-urée
US9119808B1 (en) 2012-10-08 2015-09-01 Coyote Pharmaceuticals, Inc. Treating neurodegenerative diseases with GGA or a derivative thereof

Also Published As

Publication number Publication date
EP1089731A1 (fr) 2001-04-11
JP2002518448A (ja) 2002-06-25
CA2335366A1 (fr) 1999-12-29
EP1089731A4 (fr) 2003-06-18
AU754767B2 (en) 2002-11-21
AU4698999A (en) 2000-01-10

Similar Documents

Publication Publication Date Title
EP1919466B9 (fr) Formulations pour le traitement des anormalites liees aux lipoproteines comprenant une statine et un derive de la methylnicotinamide
EP1660066B1 (fr) Composition pharmaceutique comprenant une combinaison de metformine et une statine
PT97806A (pt) Processo para a preparacao de composicoes farmaceuticas contendo pravastatina utilizadas para prevenir a manifestacao de reestenose apos angioplastia
US20070166321A1 (en) Compositions and Methods for Reducing Cholesterol and Inflammation
US20150272944A1 (en) Novel triglyceride reducing agent
AU754767B2 (en) Compositions and methods for treating elevated blood cholesterol
ES2926725T3 (es) Formulación médica para tratar la hipercolesterolemia
EP0373507A1 (fr) Combinaison d'un inhibiteur de HMG-CoA réductase et autres agents réduisant le cholestérol du sérum et méthode pour diminuer le cholestérol du sérum utilisant celle combinaison
US9642860B2 (en) Combinations of corroles and statins
JPH06500537A (ja) アテローム硬化症の治療のためのアリールヒドロキシ尿素化合物の使用
US20050182036A1 (en) Medicinal composition containing an HMG-CoA reductase inhibitor
EP1089732A1 (fr) Compositions et methodes de traitement d'une cholesterolemie elevee
US20040023919A1 (en) Blood lipid ameliorant composition
KR101086040B1 (ko) 간섬유화 및 간경화 치료 효과를 갖는 아시아트산 유도체
KR20060124634A (ko) 유기 화합물의 용도
Jurkowski Statins regulate IL-1β-induced RANKL and OPG production by human gingival fibroblasts
CA2494801A1 (fr) Composition medicamenteuse contenant un inhibiteur de hmg-coa reductase
RU2008144912A (ru) Комбинации активатора (активаторов) рецептора, активируемого пролифератором пероксисом (рапп), и ингибитора (ингибиторов) всасывания стерина и лечение заболеваний сосудов

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AL AM AU AZ BA BB BG BR BY CA CN CU CZ EE GD GE HR HU ID IL IN IS JP KG KR KZ LC LK LR LT LV MD MG MK MN MX NO NZ PL RO RU SG SI SK TJ TM TR TT UA US UZ VN YU ZA

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW SD SL SZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 1999930451

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 46989/99

Country of ref document: AU

ENP Entry into the national phase

Ref document number: 2335366

Country of ref document: CA

ENP Entry into the national phase

Ref country code: JP

Ref document number: 2000 555615

Kind code of ref document: A

Format of ref document f/p: F

WWP Wipo information: published in national office

Ref document number: 1999930451

Country of ref document: EP

WWG Wipo information: grant in national office

Ref document number: 46989/99

Country of ref document: AU

WWW Wipo information: withdrawn in national office

Ref document number: 1999930451

Country of ref document: EP