WO2001072699A1 - Inhibition of angiogenesis and tumor growth - Google Patents

Inhibition of angiogenesis and tumor growth Download PDF

Info

Publication number
WO2001072699A1
WO2001072699A1 PCT/US2001/009756 US0109756W WO0172699A1 WO 2001072699 A1 WO2001072699 A1 WO 2001072699A1 US 0109756 W US0109756 W US 0109756W WO 0172699 A1 WO0172699 A1 WO 0172699A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
cis
cyclohexyl
trans
covalent bond
Prior art date
Application number
PCT/US2001/009756
Other languages
English (en)
French (fr)
Inventor
Dale L. Boger
David A. Cheresh
Original Assignee
The Scripps Research Institute
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 The Scripps Research Institute filed Critical The Scripps Research Institute
Priority to AU5101801A priority Critical patent/AU5101801A/xx
Priority to EP01924359A priority patent/EP1276713A4/en
Priority to AU2001251018A priority patent/AU2001251018B2/en
Priority to US10/240,141 priority patent/US6803383B2/en
Priority to SK1484-2002A priority patent/SK14842002A3/sk
Priority to JP2001570612A priority patent/JP2003528850A/ja
Priority to CA2403871A priority patent/CA2403871C/en
Priority to HU0301797A priority patent/HUP0301797A3/hu
Priority to MXPA02009504A priority patent/MXPA02009504A/es
Publication of WO2001072699A1 publication Critical patent/WO2001072699A1/en
Priority to NO20024578A priority patent/NO328969B1/no

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/215Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids
    • A61K31/216Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acids having aromatic rings, e.g. benactizyne, clofibrate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/192Carboxylic acids, e.g. valproic acid having aromatic groups, e.g. sulindac, 2-aryl-propionic acids, ethacrynic acid 
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • 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
    • A61P9/14Vasoprotectives; Antihaemorrhoidals; Drugs for varicose therapy; Capillary stabilisers

Definitions

  • This invention relates to compositions for inhibiting angiogenesis and tumor growth. More particularly, the invention relates to compositions that bind to integrin ⁇ v ⁇ 3 and block the interaction of integrin ⁇ v ⁇ 3 with matrix metalloprotemase 2 (MMP2). The invention also relates to methods of inhibiting angiogenesis and tumor growth utilizing selective inhibitors of the binding of integrin ⁇ v ⁇ 3 with MMP2.
  • MMP2 matrix metalloprotemase 2
  • MMP2 matrix metalloprotemase 2
  • MT1- MMP activator membrane type 1 -matrix metalloprotemase
  • MMP function in general, may be required for many processes in the adult organism, active site inhibition of enzymatic function is likely to have far reaching effects on various biological processes involving tissue remodeling, such as wound healing.
  • therapies with broad spectrum MMP inhibitors in clinical studies of various cancer types cause severe side effects, including inflammatory tendinitis, polyarthritis, and muscoskeletal pain syndromes, which are dose limiting and often persist after discontinuation of therapy.
  • integrin ⁇ v ⁇ 3 in adult organisms, however, one would predict that targeting the interaction between MMP2 and ⁇ v ⁇ 3 to the areas of neovascularization or cellular invasion should correspondingly limit the effects of such treatment-related toxicities.
  • the present invention provides novel compounds useful as inhibitors of angiogenesis and tumor growth.
  • the invention also provides a method for the inhibition of the interaction of MMP2 with integrin ⁇ v ⁇ 3 and a method for inhibition of angiogenesis in cells containing integrin ⁇ v ⁇ 3 . Further, the invention provides a method for inhibition of tumor growth by administration of MMP2- ⁇ v ⁇ 3 interaction inhibitors.
  • the compounds of the present invention are represented by Formula (I) and include glycyl lysine derivatives chemically attached to a linking group:
  • R 2 and R 3 are each independently H, Cj - C 4 alkyl, phenyl or benzyl; X 2 and X 3 are each independently halo, nitro, C t - C 4 alkoxy, - C 4 alkyl, or - C 4 perfluoroalkyl; A is H or a covalent bond; and t is an integer having a value of 0 or 1; with the proviso that when A is H, t is 0 and when A is a covalent bond, t is 1.
  • the glycyl lysine derivative moieties may be attached to the benzene linking group in the ortho, meta or para position.
  • angiogenesis and tumor growth inhibiting compounds of the present invention are thus useful therapeutic agents for the treatment of patients with tumors or angiogenic disorders. Because the present compounds bind to ⁇ v ⁇ 3 , these compounds can also be used to suppress inflammatory events.
  • the compounds. of the present invention may be formulated in suitable pharmaceutically acceptable media to afford pharmaceutical compositions useful for the treatment of tumors and other disorders involving undesired angiogenesis.
  • compositions containing the compounds of Formulas (I) and (II) are prepared by formulating the compound in a pharmaceutically acceptable matrix.
  • the pharmaceutical compositions of the active compounds are administered to a patient with a tumor to reduce or eliminate tumor growth.
  • the active compounds can be administered parenterally by injection or by gradual infusion over time, or by any other method suitable for the particular dosage form.
  • FIG. 1 is a schematic illustration depicting MMP2 interaction with integrin ⁇ v ⁇ 3 and its role in angiogenesis as well as the inhibition of MMP2 interaction with ⁇ v ⁇ 3 by an antagonist such as the compounds of the present invention.
  • FIG. 2A graphically illustrates the effect of inhibitor compounds of Formula (I) on MMP2 interaction with integrin ⁇ v ⁇ 3 in a solid phase binding assay.
  • FIG. 2B illustrates the binding of compounds of Formula (I) on with integrin ⁇ v ⁇ 3 and 5 ⁇
  • FIG. 2C compares the effect of a compound of the present invention with a control compound on binding to ⁇ v ⁇ 3 .
  • FIG. 2D compares the effect of amino acid residues RGD on binding of a present compound to v ⁇ 3 with the effect of RGD on binding of bVN to ⁇ v ⁇ 3 .
  • FIG. 3A graphically illustrates proteinase activity in ⁇ 3 positive cells and in ⁇ 3 negative cells.
  • FIG. 3B illustrates MMP2 binding in ⁇ 3 positive cells and ⁇ 3 negative cells.
  • FIG. 4A is a microscopic depiction of angiogenesis inhibition in Chick CAM tissue.
  • FIG. 4B is a graphical illustration of angiogenesis inhibition in Chick CAM tissue.
  • FIG. 4C depicts the levels of MMP2 in treated and untreated cells.
  • FIG. 5 A is a microscopic depiction of tumor growth and vasculature in Chick CAM tissue.
  • FIG. 5B is a microscopic depiction of blood vessel density in Chick CAM tissue.
  • FIG. 5C is graphical illustration of tumor weight in Chick CAM tissue.
  • FIG. 5D is a graphical illustration of vascularization in Chick
  • FIG. 5D is a microscopic depiction of tumor cell density in Chick CAM tissue.
  • the binding of MMP2 to integrin ⁇ v ⁇ 3 is an important mechanism in the process of angiogenesis. Specific inhibition of this binding interaction results in a reduction in vascularization in growing tissues such as tumors, and thus retards tumor growth.
  • the interaction of MMP2 with integrin ⁇ v ⁇ 3 is illustrated pictorially in FIG. 1.
  • a new class of angiogenesis and tumor growth inhibitors are small molecule antagonists described below, that specifically interfere with the binding of MMP2 to integrin oc v ⁇ 3 , thus affording an important new treatment tool.
  • Certain compounds of this invention may possess one or more asymmetric centers and may exist in optically active forms. Additional asymmetric centers may be present in a substituent group, such as an alkyl group.
  • alkoxy means an oxygen atom linked by an ether bond to an alkyl group, as defined below, of the size indicated. Examples of alkoxy groups are methoxy, ethoxy, t-butoxy, and the like.
  • alkyl means a straight- or branched-chain carbon radical of the size indicated.
  • alkyl radicals are methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl, t-butyl, 2-ethylhexyl, n-octyl, 2,4-dimethylpentyl, and the like.
  • hydroxyalkyl means an alkyl group, as defined above, of the size indicated, attached to a hydroxyl group. Examples include hydroxymethyl, 2-hydroxyethyl, 3-hydroxy-l-propyl, 2-hydroxy-l-propyl, 4-hydroxybutyl, and the like.
  • perfluoroalkyl refers to a alkyl group of the size indicated, as defined below, bearing f ⁇ uoro substituents in place of each hydrogen, for example trifluoromethyl and pentafluoroethyl.
  • halo or halogen refer to bromo, chloro, fluoro and iodo.
  • the compounds of the present invention are represented by Formula (I) and include glycyl lysine derivatives chemically attached to a linking group:
  • G 1 and G 2 are each independently -NH-C ⁇ -O-R 1 , -NH-C ⁇ -O- ⁇ H ⁇ .- ⁇ H ⁇ -X 1 , -NH-C(O)-NH-(CH 2 ) v -(C 6 H 4 )-X 1 , -O-C ⁇ -NH- CH ⁇ H ⁇ -X 1 , -O-C O-O- CH ⁇ H ⁇ -X 1 , or -NH-C(O)-CH 2 -(C 6 H 4 )-X 1 ;
  • Y 1 and Y 2 are each independently OH, C r C 4 alkyl, C r C 4 hydroxyalkyl, C r C 4 alkoxy, phenyl, benzyl, or -NH 2 ;
  • R 1 is C r C 4 alkyl;
  • X 1 is halo, nitro, C r C 4 alkyl, C r C 4 alkoxy, or C r C 4 perfluoroal
  • G 1 and G 2 are -NH-C(O)-O-(CH 2 ) v -(C 6 H 4 )-X ⁇ Y 1 and Y 2 are OH, and m and n are 1.
  • X 1 is C r C 4 perfluroalkyl, most preferably trifluoromethyl.
  • Preferred compounds within the purview of structural Formula (I) are represented in structural Formula (II) and include glycyl lysine derivatives attached to a benzene linking group in either the ortho, meta or para orientation:
  • R 2 and R 3 are each independently H, C l - C 4 alkyl, phenyl or benzyl; X 2 and X 3 are each independently halo, nitro, C j - C 4 alkoxy, - C 4 alkyl, or C x - C 4 perfluoroalkyl; A is H or a covalent bond; and t is an integer having a value of 0 or 1; with the proviso that when A is H, t is 0 and when A is a covalent bond, t is 1.
  • the glycyl lysine derivative moieties may be attached to the benzene linking group in the ortho, meta or para position.
  • the substituents X 2 and X 3 are attached to the phenyl ring of the benzyl moiety in the 4- position relative to the benzylic CH 2 (i.e. para substituent).
  • the preferred X 2 and X 3 groups are C, to C 4 perfluoroalkyl, most preferred is para-trifluoromethyl.
  • the preferred R 2 and R 3 groups are hydrogen and methyl.
  • the substituents X 2 and X 3 may be the same or different, and the substituents R 2 and R 3 may also be the same or different.
  • a particularly active member of the family of compounds represented by Formula (II), wherein A is a covalent bond and t is 1, is the compound represented by Compound 1 below:
  • the compounds of Formulas (I) and (II) may be synthesized from readily available materials in a moderate number of synthetic steps.
  • Scheme 1 shows the synthesis of Compound 1, and is illustrative of a general method of producing compounds of Formula (II) wherein A is covalent bond, R 2 and R 3 are the same and X 2 and X 3 are the same.
  • Scheme 1 further illustrates the synthesis of compounds of Formula (II), wherein X 2 and X 3 are both para-trifluoromethyl, and R 2 and R 3 are both either methyl or hydrogen.
  • Scheme 3 illustrates the synthesis of Compound 12, an inactive analog of Compound 1, in which the p-trifluoromethylbenzyloxycarbonyl group of Formula (II) is replaced by a benzoyl amide.
  • compositions comprising the active compounds of Formulas (I) and (II) are administered to a host with a tumor to reduce or eliminate tumor growth.
  • the active compounds can be administered parenterally by injection, or by gradual infusion over time.
  • tissue to be treated is most often treated by intraperitoneal or subcutaneous administration, the active compounds can also be administered intraocularly, intravenously, intramuscularly, intrasynovially, intracavity, or transdermally, and can be delivered by peristaltic means as well.
  • administration refers to systemic use as when taken orally, parenterally, by inhalation spray, by nasal, rectal or buccal routes, or topically in dosage form unit formulations containing conventional nontoxic pharmaceutically acceptable carriers, adjuvants and vehicles as desired.
  • parenteral as used herein includes intravenous, intramuscular, intraperitoneal, intrasternal, subcutaneous and intraarticular injection and infusion techniques.
  • pharmaceutically acceptable it is meant those salts, amides and esters which are, within the scope of sound medical judgement, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio, effective for their intended use in the treatment of tumors and angiogenic-related disorders.
  • Pharmaceutically acceptable salts are well known in the art. For example, S. M Berge, et al. describe pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences, 66: 1-19 (1977).
  • Representative acid addition salts include hydrochloride, hydrobromide, sulfate, bisulfate, acetate, oxalate, valerate, oleate, palmitate, stearate, laurate, borate, benzoate, lactate, phosphate, toluenesulfonate, methanesulfonate, citrate, maleate, fumarate, succinate, tartrate, ascorbate, glucoheptonate, lactobionate, lauryl sulfate salts and the like.
  • Representative alkali or alkaline earth metal salts include sodium, calcium, potassium, magnesium salts and the like.
  • the term "pharmaceutically acceptable carriers” means a non-toxic, inert solid, semi-solid or liquid filler, diluent, encapsulating material or formulation auxiliary of any type.
  • Some examples of the materials that can serve as pharmaceutically acceptable carriers are sugars, such as lactose, glucose and sucrose; starches such as corn starch and potato starch; cellulose and its derivatives such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients such as cocoa butter and suppository waxes; oils such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; glycols, such as propylene glycol; polyols such as glycerin, sorbitol, mannitol and polyethylene glycol; esters such as ethyl oleate and ethyl laur
  • wetting agents such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, releasing agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the composition, according to the judgement of the formulator.
  • antioxidants examples include water soluble antioxidants such as ascorbic acid, cysteine hydrochloride, sodium bisulfite, sodium metabisulfite, sodium sulfite, and the like; oil soluble antioxidants such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxy toluene (BHT), lecithin, propyl gallate, alpha-tocopherol and the like; and the metal chelating agents such as citric acid, ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid and the like.
  • water soluble antioxidants such as ascorbic acid, cysteine hydrochloride, sodium bisulfite, sodium metabisulfite, sodium sulfite, and the like
  • oil soluble antioxidants such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxy toluene (BHT), lecithin, propy
  • a “therapeutically effective amount” of the inventive agent or compound is meant a sufficient amount of the compound to treat tumors and angiogenic-related disorders at a reasonable benefit/risk ratio applicable to any medical treatment. It will be understood, however, that the total daily usage of the compounds and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgement.
  • the specific therapeutically effective dose level for any particular patient will depend upon a variety of factors including the disorder being treated and the severity of the disorder; activity of the specific compound employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidently with the specific compound employed; and like factors well known in the medical arts.
  • compositions in unit dosage forms comprising a therapeutically effective amount of a compound (or compounds) of this invention in combination with a conventional pharmaceutical carrier.
  • injectable preparations for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution, suspension or emulsion in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol.
  • acceptable vehicles and solvents that may be employed are water, Ringer's solution, U.S. P. and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil can be employed including synthetic mono- or digly cerides.
  • fatty acids such as oleic acid are used in the preparation of injectables.
  • the injectable formulation can be sterilized, for example, by filtration through a bacteria-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium just prior to use.
  • sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium just prior to use.
  • Injectable depot forms can also be made by forming microcapsule matrices of drugs and biodegradable polymers such as polylactide-polyglycolide. Depending on the ratio of drug to polymer and the composition of the polymer, the rate of drug release can be controlled. Examples of other biodegradable polymers include poly-orthoesters and polyanhydrides.
  • the depot injectables can also be made by entrapping the drug in liposomes or microemulsions which are compatible with body tissues.
  • Suppositories for rectal administration of the drug can be prepared by mixing the drug with a suitable nonirritating excipient such as cocoa butter and polyethylene glycol which are solid at ordinary temperature but liquid at the rectal temperature and will therefore melt in the rectum and release the drug.
  • Solid dosage forms for oral administration may include capsules, tablets, pills, powders, prills and granules. In such solid dosage forms the active compound may be admixed with at least one inert diluent such as sucrose, lactose or starch.
  • Such dosage forms may also comprise, as is normal practice, additional substances other than inert diluents, e.g., tableting lubricants and other tableting aids such as magnesium stearate and microcrystalline cellulose.
  • additional substances other than inert diluents e.g., tableting lubricants and other tableting aids such as magnesium stearate and microcrystalline cellulose.
  • the dosage forms may also comprise buffering agents. Tablets and pills can additionally be prepared with enteric coatings and other release-controlling coatings.
  • Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like.
  • Liquid dosage forms for oral administration may include pharmaceutically acceptable lo emulsions, microemulsions, solutions, suspensions, syrups and elixirs containing inert diluents commonly used in the art such as water. Such compositions may also comprise adjuvants, such as wetting agents; emulsifying and suspending agents; sweetening, flavoring and perfuming agents. If desired, the compounds of the present invention can be incorporated into slow release or targeted delivery systems such as polymer matrices, liposomes and microspheres.
  • the active compounds may be sterilized, for example, by filtration through a bacteria-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can dissolve in sterile water, or some other sterile injectable medium immediately before use.
  • the active compounds can also be in micro-encapsulated form with one or more excipients as noted above.
  • the solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferably, in a certain part of the intestinal tract, optionally in a delayed manner. Examples of embedding compositions which can be used include polymeric substances and waxes. Dosage forms for topical or transdermal administration of a compound of this invention further include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants or patches.
  • the active component is admixed under sterile conditions with a pharmaceutically acceptable carrier and any needed preservatives or buffers as may be required.
  • Ophthalmic formulations, ear drops, eye ointments, powders and solutions are also contemplated as being within the scope of this invention.
  • the ointments, pastes, creams and gels may contain, in addition to an active compound of this invention, excipients such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.
  • Powders and sprays can contain, in addition to the compounds of this invention, excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and poly amide powder, or mixmres of these substances.
  • Sprays can additionally contain customary propellants such as chlorofluorohydrocarbons.
  • Transdermal patches have the added advantage of providing controlled delivery of a compound to the body. Such dosage forms can be made by dissolving or dispersing the compound in the proper medium. Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate can be controlled by either providing a rate controlling membrane or by dispersing the compound in a polymer matrix or gel.
  • compositions containing the active compounds are administered in a manner compatible with the dosage formulation and in a therapeutically effective amount.
  • the quantity to be administered and the timing of administration depend on the host to be treated, capacity of the host's system to utilize the active ingredient, and degree of therapeutic effect desired. Precise amounts of the active ingredient required to be administered depend on the judgment of the practitioner, and are peculiar to each individual.
  • Suitable dosage ranges for systemic application are disclosed herein and depend on the route of administration. Suitable regimes for administration are also variable, but are typified by an initial administration, followed by repeated doses at one or more predetermined intervals by a subsequent injection or other route of administration.
  • the present invention also provides a pharmaceutical composition useful for practicing the therapeutic methods described herein.
  • the compositions contain an active compound described hereinabove, together with a pharmaceutically acceptable carrier.
  • Preparations for parental administration of the present compounds or compositions include sterile aqueous or non-aqueous solutions, suspensions, and emulsions.
  • non-aqueous solvents are propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and injectable organic esters such as ethyl oleate.
  • Aqueous carriers include water, alcoholic/aqueous solutions, emulsions or suspensions, including saline and buffered media.
  • Parental vehicles include sodium chloride solution, Ringer's dextrose, dextrose and sodium chloride, lactated Ringer's, or fixed oils.
  • Intravenous vehicles include fluid and nutrient replenishers, electrolyte replenishers (such as those based on Ringer's dextrose), and the like. Preservatives and other additives may also be present, such as, for example, antimicrobials, antioxidants, chelating agents, inert gases, and the like.
  • Another aspect of the present invention provides a method for inhibiting MMP2 interaction with ⁇ v ⁇ 3 and thus angiogenesis in a tumor tissue.
  • the inhibiting method comprises administering to the host a composition comprising an angiogenesis-inhibiting amount of a compound described hereinabove. MMP2 interaction with v ⁇ 3 is inhibited by contacting ⁇ v ⁇ 3 with a compound of the present invention. .
  • Angiogenesis is the formation of a neovascular network from preexisting host vessels and is required for tumor growth beyond 1-2 mm 3 .
  • angiogenesis is inhibited as long as angiogenesis and the disease symptoms mediated by angiogenesis are ameliorated.
  • the dosage ranges for the administration to a host of the active compound depend upon the particular active compound and its potency to a particular tumor or integrin. One skilled in the art can readily determine the proper dosage for a particular active compound without undue experimentation.
  • the host can be any mammal.
  • the dosage should be large enough to produce the desired therapeutic effect in which angiogenesis and the disease symptoms mediated by angiogenesis are ameliorated, and is usually an amount sufficient to maintain a plasma level of the active compound in the range of about 0.01 to about 100 micromolar ( ⁇ M), preferably about 0.2 to about 20 ⁇ M, more preferably about 1 to about 10 ⁇ M.
  • the dosage should not be so large as to cause adverse side effects, however.
  • the dosage per kilogram (kg) of body weight can vary from 1 to 20 mg per dose, in one or more dose administrations daily, for one or several days or indefinitely.
  • the therapeutically effective amount is an amount of active compound sufficient to produce a measurable inhibition of angiogenesis in the tissue being treated, i.e., an angiogenesis-inhibiting amount or an MMP2 - ⁇ v ⁇ 3 interaction inhibiting amount.
  • Inhibition of angiogenesis can be measured in situ by immunohistochemistry, as described herein, or by other methods known to one skilled in the art.
  • the present invention additionally provides pharmaceutical compositions useful for practicing the therapeutic methods described herein.
  • the compositions contain an active compound defined hereinabove together pharmaceutically acceptable carrier.
  • the present invention also provides a method of inducing apoptosis in tumor cells. This method comprises administering to the host a therapeutically effective amount of an active compound sufficient to initiate tumor cell apoptosis.
  • tumor cell apoptosis is induced if an increased tumor cell apoptosis is observed in the target tumor being treated.
  • Tumor cell apoptosis can be measured by methods described herein or commonly known in the art. The following non-limiting examples are provided to illustrate various aspects of the present invention.
  • CS-1 hamster melanoma cells and CS-1 cells transfected with the human ⁇ 3 -integrin subunit ( ⁇ 3 CS-l cells) were described previously ⁇ Cell, 85, 683-93 (1996); Cell, 92, 391-400 (1998)).
  • the horseradish peroxidase (HRP)-conjugated monoclonal antibodies anti-biotin mAb BN-34 and anti-actin mAb AC-40 were obtained from Sigma (St. Louis, MO).
  • Anti-von Willebrand Factor (vWF) polyclonal antibodies (pAb) were obtained from DAKO (Glostrup, Denmark).
  • the cyclic peptides cRGDfV and cRADfV and integrin- ⁇ v ⁇ 3 were generously provided by Merck KGaA (Darmstadt, Germany).
  • Purified proMMP2 and integrin- ⁇ v ⁇ 3 were provided by Chemicon International (Temecula, CA).
  • Purified active MMP2 was obtained from Calbiochem (La Jolla, CA).
  • Basic fibroblast growth factor (bFGF) was kindly provided by Scios (Mountain View, CA).
  • Purified integrins were adsorbed overnight onto microtiter wells (1- 5 ⁇ g/ml, 50 ⁇ g/well) prior to blocking with Caseinblocker (Pierce, Rockford, IL).
  • Purified biotinylated MMP2 (bMMP2, 3-5 nM) in binding buffer (50 mM Tris, pH 8, 150 mM NaCl, 1 mM MgCl 2 , 0.5 mM MnCl 2 ) was added to the wells in the presence or absence of Compound 1,.
  • Compound 12 cyclic RGD or RAD peptides, or buffer vehicle alone. Control wells received no integrin.
  • Biotinylated vitronectin (bVN, 1 ⁇ g/ml) was used as a reference. Bound protein was detected with HRP-anti-biotin mAb and quantitated at 450 nm with 3,3',5,5'- tetramethylbenzidine solution (TMB; a substrate for the peroxidase) (BioRad,
  • v ⁇ 3 and ⁇ 5 ⁇ ! (10 ⁇ g/ml, 50 ⁇ l/well) were coated onto Immulon-4 microtiter wells (Dynatech Laboratories, Chantilly, NA), which were substantially blocked and incubated with titration of [ 14 C]-Compound 1 prior to the addition of 150 ⁇ l of binding buffer containing 0.1% Tween-20 and aspiration of all liquid. Dried wells were separated and immersed in BetaMax liquid scintillation cocktail (IC ⁇ Biochemicals, Costa Mesa, CA) for quantitation.
  • Example 2 MMP2 Cell-Binding and [ 3 H " l-Colla en IN Degradation Assays.
  • FBM adhesion buffer fibroblast basal medium
  • BSA bovine serum albumin
  • MnCl 2 0.5% bovine serum albumin
  • 10 ⁇ g/ml aprotinin aprotinin containing either 4 nM purified active MMP2 alone, or in combination with 10 ⁇ M Compound 1 or Compound 12 for 45 minutes at 37 °C prior to washing and addition to the [ 3 H]-collagen IV- coated wells.
  • Angiogenesis was assessed essentially as described previously ⁇ Cell, 85, 683-93 (1996); Cell, 92, 391-400 (1998)).
  • bFGF basic fibroblast growth factor
  • chick embryo CAMs were treated with 20 ⁇ l of 3 ⁇ M Compound 1 or Compound 12.
  • the CAMs were quantitated in a blind evaluation. CAMs from each group were pooled, minced, and extracted with 50 mM Tris, 150 nM NaCl, 0.1 % Triton X-100 containing COMPLETE-brand protease inhibitor cocktail without EDTA (Boehringer, Mannheim, Germany) prior to analysis by zymography.
  • Immunoblotting Equal quantities of protein were separated by SDS-PAGE under reducing conditions and electroblotted to an Immobilon-P membrane (Millipore, Bedford, MD). The membrane was blocked and immobilized proteins were detected by incubation with an antigen-specific primary antibody, followed by an HRP-conjugated secondary antibody as required. Bands were visualized the chemiluminescent substrate PS-3 (Lumigen, Inc., Southfield, MI).
  • Chick CAM lysates were prepared as described above and equal quantities of protein were separated in the absence of reducing agents or boiling on polyacrylamide gels embedded with 0.2% gelatin. The gels were washed with 2% Triton X-100, followed by extensive washing with water prior to overnight incubation at 37 °C in collagenase buffer (50 mM Tris 7.4, 200 mM NaCl 10 mM CaC12). Gelatinolytic activity was visualized by staining the gels with 0.5%) Coomassie blue.
  • Example 5 Tumor Growth Assay. Primary tumors were grown on CAMs of 9-day embryos by implantation 5 x 10 6 CS-1 cells and incubation for 7 days. At this point, 50 mg sections of these tumors were subcultured onto fresh 9-day CAMs and allowed to implant for 24 hours before a single intravenous (IV) injection with 100 ⁇ l of 100 ⁇ M of test compounds in Hank'sBalanced Saline Solution (HBSS). Buffer alone was used as control. Tumors were incubated for a total of 10 days, harvested and trimmed free of excess stromal tissue before determining wet weight and processing for histology.
  • IV intravenous
  • HBSS Hank'sBalanced Saline Solution
  • Example 6 Immunofluorescence Assays Snap-frozen CS-1 tumor sections were fixed with 4% paraformaldehyde and permeablized with 0.1% Triton X-100. Sections were blocked with 5% bovine serum albumin (BSA) in phosphate buffered saline (PBS) prior to staining with an anti-vWF pAb and visualization with an Alexa 568- conjugated anti-rabbit secondary antibody. Samples were analyzed on a MRC1024 confocal microscope (BioRad, Hercules, CA). Blood vessel density was quantitated with a 20X objective on four fields per section and four tumors per condition.
  • BSA bovine serum albumin
  • PBS phosphate buffered saline
  • Example 7 (SVMethyl 6-(((tert-butyloxy)carbonyl)amino')-2-((4-trifluoromethyl)- benzyloxycarbonyPhexanoate (2 .
  • Example 8 (SVMethyl 6-r2-(((tert-b ⁇ tyloxy carbonyl)amino ' )acetamido1-2-[(4- trifluoromethyDbenzyloxycarbonyllhexanoate (3) .
  • Compound 1 binds directly to integrin v ⁇ 3 and not to MMP2.
  • additional solid phase receptor binding assays were performed with immobilized ⁇ v ⁇ 3 and [ 14 C]-labeled Compound 1, or biotinylated VN as a control.
  • Compound 1 bound directly to integrin ⁇ v ⁇ 3 in a solid phase receptor binding assay. This interaction was dose-dependent, saturable and specific, demonstrating minimal interaction of Compound 1 with the unrelated control integrin 5 ⁇ (FIG. 2B). Indeed, negligible binding to integrin ⁇ 5 ⁇ was observed at higher concentrations of compound (data not shown).
  • FIG. 3 A To demonstrate that the reduced cell-mediated collagen degradation observed in FIG. 3 A was the result of inhibition of MMP2 interaction with integrin ⁇ v ⁇ 3 by Compound 1 on the cell surface, CS-1 cells and their ⁇ v ⁇ 3 -bearing counterpart were examined in a biotinylated MMP2 binding assay. As expected, the ⁇ 3 -negative CS-1 cells were capable of binding some level of MMP2, however their capacity to do so was not diminished by the presence of either compound (FIG. 3B). In contrast, ⁇ 3 CS-l cells bound significantly greater quantities of MMP2, and this enhanced MMP2 binding was specifically suppressed by Compound 1.
  • Compound 1 when corrected for the loading of the lanes as demonstrated by staining with an anti-actin mAb, Compound 1 effectively reduced the binding of MMP2 to the ⁇ 3 CS-l cells to the level observed in the absence ⁇ v ⁇ 3 (i.e. parental CS-1 cells)(FIG. 3B, lane 2). Compound 1 disrupts angiogenesis in vivo without suppressing
  • Compound 1 abrogates tumor growth in vivo. Disruption of angiogenesis has been shown to inhibit tumor growth in numerous systems. As a result, blocking the invasive properties of endothelial cells by inhibiting MMPs suppresses angiogenesis and tumor growth in animal models as well. In fact, a number of MMP inhibitors have shown promise as anti-angiogenic agents in man.

Landscapes

  • Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Epidemiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Vascular Medicine (AREA)
  • Emergency Medicine (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Hydrogenated Pyridines (AREA)
PCT/US2001/009756 2000-03-27 2001-03-27 Inhibition of angiogenesis and tumor growth WO2001072699A1 (en)

Priority Applications (10)

Application Number Priority Date Filing Date Title
AU5101801A AU5101801A (en) 2000-03-27 2001-03-27 Inhibition of angiogenesis and tumor growth
EP01924359A EP1276713A4 (en) 2000-03-27 2001-03-27 INHIBITION OF ANGIOGENESIS AND TUMOR GROWTH
AU2001251018A AU2001251018B2 (en) 2000-03-27 2001-03-27 Inhibition of angiogenesis and tumor growth
US10/240,141 US6803383B2 (en) 2000-03-27 2001-03-27 Inhibition of angiogenesis and tumor growth
SK1484-2002A SK14842002A3 (sk) 2000-03-27 2001-03-27 Inhibícia angiogenézy a rastu tumoru
JP2001570612A JP2003528850A (ja) 2000-03-27 2001-03-27 血管形成および腫瘍成長の阻害
CA2403871A CA2403871C (en) 2000-03-27 2001-03-27 Inhibition of angiogenesis and tumor growth
HU0301797A HUP0301797A3 (en) 2000-03-27 2001-03-27 Inhibition of angiogenesis and tumor growth
MXPA02009504A MXPA02009504A (es) 2000-03-27 2001-03-27 Inhibicion de angiogenesis y de crecimiento de tumor.
NO20024578A NO328969B1 (no) 2000-03-27 2002-09-24 Forbindelser, sammensetninger og anvendelser derav

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US19226000P 2000-03-27 2000-03-27
US60/192,260 2000-03-27

Publications (1)

Publication Number Publication Date
WO2001072699A1 true WO2001072699A1 (en) 2001-10-04

Family

ID=22708924

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/US2001/009756 WO2001072699A1 (en) 2000-03-27 2001-03-27 Inhibition of angiogenesis and tumor growth
PCT/US2001/009785 WO2001072297A1 (en) 2000-03-27 2001-03-27 Methods for inhibiting angiogenesis and tumor growth

Family Applications After (1)

Application Number Title Priority Date Filing Date
PCT/US2001/009785 WO2001072297A1 (en) 2000-03-27 2001-03-27 Methods for inhibiting angiogenesis and tumor growth

Country Status (15)

Country Link
EP (2) EP1272173A4 (no)
JP (2) JP2003528850A (no)
KR (2) KR100767616B1 (no)
CN (2) CN1229339C (no)
AU (4) AU5101801A (no)
CA (3) CA2659030A1 (no)
CZ (2) CZ20023510A3 (no)
HU (2) HUP0301621A3 (no)
MX (2) MXPA02009510A (no)
NO (2) NO328969B1 (no)
PL (2) PL358272A1 (no)
RU (2) RU2276133C2 (no)
SK (2) SK14852002A3 (no)
WO (2) WO2001072699A1 (no)
ZA (2) ZA200208628B (no)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2315721T5 (es) 2003-10-23 2012-08-24 Otsuka Pharmaceutical Co., Ltd. Formulación de aripiprazol inyectable estéril de liberación controlada y procedimiento
UA97286C2 (ru) 2007-07-31 2012-01-25 Оцука Фармасьютикал Ко., Лтд. Способ изготовления суспензии арипипразола и лиофилизированного состава

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4511504A (en) * 1983-04-26 1985-04-16 G.D. Searle & Co. Carboxyalkyl peptide derivatives
WO1998003573A1 (en) * 1996-07-17 1998-01-29 Biomolecular Research Institute Ltd. Angiogenic inhibitory compounds
WO1998009957A1 (en) * 1996-09-04 1998-03-12 Warner-Lambert Company Compounds for and a method of inhibiting matrix metalloproteinases
WO1999049856A2 (en) * 1998-03-27 1999-10-07 Genentech, Inc. Antagonists for treatment of cd11/cd18 adhesion receptor mediated disorders

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH05125029A (ja) 1991-11-06 1993-05-21 Yamanouchi Pharmaceut Co Ltd 新規なアミド化合物又はその塩
GB9308695D0 (en) * 1993-04-27 1993-06-09 Celltech Ltd Peptidyl derivatives
GB9405076D0 (en) * 1994-03-16 1994-04-27 Inst Of Ophtalmology A medical use of matrix metalloproteinase inhibitors
EP0968423A4 (en) * 1997-02-07 2002-10-30 Scripps Research Inst CONVERGENT SYNTHESIS OF COMBINATORIAL BANKING

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4511504A (en) * 1983-04-26 1985-04-16 G.D. Searle & Co. Carboxyalkyl peptide derivatives
WO1998003573A1 (en) * 1996-07-17 1998-01-29 Biomolecular Research Institute Ltd. Angiogenic inhibitory compounds
WO1998009957A1 (en) * 1996-09-04 1998-03-12 Warner-Lambert Company Compounds for and a method of inhibiting matrix metalloproteinases
WO1999049856A2 (en) * 1998-03-27 1999-10-07 Genentech, Inc. Antagonists for treatment of cd11/cd18 adhesion receptor mediated disorders

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP1276713A4 *

Also Published As

Publication number Publication date
CA2403630C (en) 2009-05-26
JP2003528850A (ja) 2003-09-30
RU2276133C2 (ru) 2006-05-10
CA2659030A1 (en) 2001-10-04
EP1272173A4 (en) 2006-05-03
CN1245967C (zh) 2006-03-22
EP1272173A1 (en) 2003-01-08
AU2001251018B2 (en) 2005-12-01
MXPA02009504A (es) 2003-05-21
AU4949901A (en) 2001-10-08
CZ20023510A3 (cs) 2003-03-12
KR20020091156A (ko) 2002-12-05
KR20020084258A (ko) 2002-11-04
AU2001249499B2 (en) 2005-04-21
CA2403630A1 (en) 2001-10-04
AU5101801A (en) 2001-10-08
NO20024578L (no) 2002-11-20
CZ20023509A3 (cs) 2003-06-18
SK14852002A3 (sk) 2003-06-03
CA2403871C (en) 2010-05-11
HUP0301797A3 (en) 2010-09-28
ZA200208626B (en) 2004-02-16
NO20024576L (no) 2002-11-20
CA2403871A1 (en) 2001-10-04
CN1229339C (zh) 2005-11-30
PL358272A1 (en) 2004-08-09
RU2269339C2 (ru) 2006-02-10
CN1429106A (zh) 2003-07-09
CN1441777A (zh) 2003-09-10
PL366316A1 (en) 2005-01-24
KR100767616B1 (ko) 2007-10-18
NO328969B1 (no) 2010-06-28
SK14842002A3 (sk) 2003-04-01
KR100776185B1 (ko) 2007-11-16
JP2003528140A (ja) 2003-09-24
HUP0301621A2 (hu) 2003-09-29
NO20024576D0 (no) 2002-09-24
NO20024578D0 (no) 2002-09-24
WO2001072297A1 (en) 2001-10-04
EP1276713A1 (en) 2003-01-22
HUP0301621A3 (en) 2009-06-29
RU2002128751A (ru) 2004-03-10
HUP0301797A2 (hu) 2003-09-29
EP1276713A4 (en) 2006-05-03
PL205134B1 (pl) 2010-03-31
ZA200208628B (en) 2004-02-12
MXPA02009510A (es) 2003-05-21

Similar Documents

Publication Publication Date Title
KR100194242B1 (ko) 눈의 염증성 질환을 치료하기 위한 3-벤조일페닐아세트산 유도체를 함유하는 국소투여가능한 조성물
JP2020125355A (ja) 併用療法
MXPA03011448A (es) Derivados de n-arilfenilacetamida y composicion farmaceutica que los contienen.
US7368478B2 (en) Methods for inhibiting angiogenesis and tumor growth
US7696235B2 (en) EP2 receptor agonists for treating glaucoma
EP1284265A1 (en) Benzothiophene derivatives and medicinal use thereof
CA2403871C (en) Inhibition of angiogenesis and tumor growth
US6803383B2 (en) Inhibition of angiogenesis and tumor growth
AU2001251018A1 (en) Inhibition of angiogenesis and tumor growth
AU2001249499A1 (en) Methods for inhibiting angiogenesis and tumor growth
CA3162689A1 (en) Combinations of tie-2 activators and prostaglandins and uses thereof
JP2002505281A (ja) 炎症細胞抑制剤
AU2013200669A1 (en) EP2 receptor agonists for treating glaucoma

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ 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 TR 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)
REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWE Wipo information: entry into national phase

Ref document number: 2403871

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2001 570612

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 1020027012720

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: PA/a/2002/009504

Country of ref document: MX

Ref document number: 10240141

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 14842002

Country of ref document: SK

WWE Wipo information: entry into national phase

Ref document number: PV2002-3510

Country of ref document: CZ

Ref document number: 2001251018

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2002/08628

Country of ref document: ZA

Ref document number: 200208628

Country of ref document: ZA

WWE Wipo information: entry into national phase

Ref document number: 2001924359

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2002 2002128751

Country of ref document: RU

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 018097448

Country of ref document: CN

WWP Wipo information: published in national office

Ref document number: 1020027012720

Country of ref document: KR

WWP Wipo information: published in national office

Ref document number: 2001924359

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: PV2002-3510

Country of ref document: CZ

WWG Wipo information: grant in national office

Ref document number: 2001251018

Country of ref document: AU