EP1467738A1 - Utilisation d'inhibiteurs de la prostaglandine e synthase ou d'antagonistes du recepteur ep2 ou ep4 dans le traitement d'une affection de l'uterus - Google Patents

Utilisation d'inhibiteurs de la prostaglandine e synthase ou d'antagonistes du recepteur ep2 ou ep4 dans le traitement d'une affection de l'uterus

Info

Publication number
EP1467738A1
EP1467738A1 EP02765100A EP02765100A EP1467738A1 EP 1467738 A1 EP1467738 A1 EP 1467738A1 EP 02765100 A EP02765100 A EP 02765100A EP 02765100 A EP02765100 A EP 02765100A EP 1467738 A1 EP1467738 A1 EP 1467738A1
Authority
EP
European Patent Office
Prior art keywords
cox
methyl
pathological condition
prostaglandin
pge
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Ceased
Application number
EP02765100A
Other languages
German (de)
English (en)
Inventor
Henry N. MRC Human Repro. Sciences Unit JABBOUR
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Medical Research Council
Original Assignee
Medical Research Council
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 GBGB0124124.9A external-priority patent/GB0124124D0/en
Application filed by Medical Research Council filed Critical Medical Research Council
Publication of EP1467738A1 publication Critical patent/EP1467738A1/fr
Ceased legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/557Eicosanoids, e.g. leukotrienes or prostaglandins
    • A61K31/559Eicosanoids, e.g. leukotrienes or prostaglandins having heterocyclic rings containing hetero atoms other than oxygen
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • 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
    • A61K31/352Heterocyclic 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 condensed with carbocyclic rings, e.g. methantheline 
    • A61K31/3533,4-Dihydrobenzopyrans, e.g. chroman, catechin
    • 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
    • A61K31/357Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having two or more oxygen atoms in the same ring, e.g. crown ethers, guanadrel
    • 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
    • A61K31/401Proline; Derivatives thereof, e.g. captopril
    • 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4151,2-Diazoles
    • 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41961,2,4-Triazoles
    • 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • A61K31/427Thiazoles not condensed and containing further heterocyclic rings
    • 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
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • 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
    • A61K31/54Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame
    • A61K31/5415Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame ortho- or peri-condensed with carbocyclic ring systems, e.g. phenothiazine, chlorpromazine, piroxicam
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/557Eicosanoids, e.g. leukotrienes or prostaglandins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • the present invention relates to methods of treatment, and in particular methods of treating uterine pathological conditions.
  • Pathological conditions of the uterus represent a serious health problem in women, particularly women of the Western world.
  • pathological conditions include uterine carcinoma, and endometrial or myometrial pathological conditions such as endometriosis (endometrial) and fibroids (myometrial).
  • Cyclooxygenase (COX) enzymes also called prostaglandin endoperoxide synthase, (PGHS), catalyse the rate limiting step in the conversion of arachidonic acid to prostaglandin H 2 (PGH 2 ).
  • PGH 2 serves as a substrate for specific prostaglandin synthase enzymes that synthesise the natural prostaglandins.
  • prostaglandin D 2 is synthesised by prostaglandin-D- synthase
  • prostaglandin E 2 PGE 2
  • prostaglandin-E-synthase PGES
  • prostaglandin F 2 ⁇ by prostaglandin-F-synthase.
  • COX-1 is constitutively expressed in many tissues and cell types and generates prostaglandins for normal physiological function (Herschman, 1996).
  • COX-2 is rapidly induced following stimulation of quiescent cells by growth factors, oncogenes, carcinogens and tumour-promoting phorbol esters (Herschman, 1996; Subbaramaiah el al., 1996).
  • two isoforms of PGES have been isolated; a microsomal glutathione-dependent inducible PGES (mPGES) and a constitutive cytosolic glutathione dependent PGES (Jakobsson et al., 1999; Tanioka et al., 2000).
  • mPGES microsomal glutathione-dependent inducible PGES
  • cytosolic glutathione dependent PGES Jakobsson et al., 1999; Tanioka et al., 2000.
  • COX-2 and possibly PGE 2 are involved in neoplastic transformation of certain epithelial cells and subsequently carcinogenesis.
  • COX-2 and PGE 2 synthesis in rat intestinal epithelial cells increases their proliferation rate, resistance to apoptosis, and their invasiveness by suppressing the transcription of target genes that may be involved in cellular growth transformation and adhesion (Tsujii & DuBois, 1995).
  • COX-2 and PGE 2 promote cancer development and invasiveness by mediating the transcription of angiogenic factors that induce both migration of endothelial cells and their arrangement into tubular structures (Tsujii et al., 1998; Jones et al, 1999b).
  • PGE 2 mediates its effect on target cells through interaction with different isoforms of seven transmembrane G protein coupled receptors (GPCR) which belong to the rhodopsin family of serpentine receptors.
  • GPCR transmembrane G protein coupled receptors
  • EP1, EP2, EP3 and EP4 Four main PGE 2 receptor subtypes have been identified (EP1, EP2, EP3 and EP4) which utilise alternate and in some cases opposing intracellular signalling pathways (Coleman et al., 1994). This diversity of receptors with opposing action may confer a homeostatic control on the action of PGE 2 that is released in high concentrations close to its site of synthesis (Ashby, 1998).
  • Endometrial carcinoma is the most common gynaecologic malignancy. Endometrial cancer can arise from several cell types but the glandular epithelium is the most common progenitor (adenocarcinomas account for 80-90% of uterine tumours). Endometrial cancer is predominantly a post-menopausal disease where incidence is uncommon below the age of forty and peaks by about seventy years of age. The incidence of endometrial cancer has been increasing steadily in the Western world during the last fifty years and this has been attributed largely to increased life expectancy and improved detection methods (Gordon & Ireland, 1994; Mant & Vessey, 1994).
  • a first aspect of the invention provides a method of treating a pathological condition of the uterus in an individual the method comprising administering to the individual any one or more of an inhibitor of prostaglandin E synthase (PGES), or an EP2 or EP4 receptor antagonist.
  • PGES prostaglandin E synthase
  • the pathological condition of the uterus may be any pathological condition wherein synthesis of PGE2 is found, and wherein expression of EP2 and EP4 receptors is found.
  • the pathological condition of the uterus is any one of uterine carcinoma, an endometrial pathological condition such as endometriosis including adenomyosis, or a myometrial pathological condition such as fibroids (leiomyomas) or leiomyosarcomas which are fibroids which have become malignant.
  • the uterine pathological condition is one which is associated with abnormal growth of cells of the myometrium or endometrium.
  • Endometriosis is the ectopic implantation and growth of endometrium and can therefore be considered as abnormal growth cells of the endometrium as defined.
  • Adenomyosis is a form of endometriosis where the ectopic endometrium is implanted in the myometrium.
  • the method of the invention is used to treat endometrial carcinoma.
  • the individual is administered an inhibitor of PGES. It has been reported by Thoren & Jakobsson (2000) Eur. J Biochem. 267, 6428-6434 (incorporated herein by reference) that NS-398, sulindac sulphide and leukotriene C 4 inhibit PGES activity with IC 50 values of 20 ⁇ M, 80 ⁇ M and 5 ⁇ M, respectively.
  • the individual is administered an antagonist of an EP2 receptor or an antagonist of an EP4 receptor.
  • the prostaglandin EP2 receptor antagonist may be any suitable EP2 receptor antagonist.
  • the prostaglandin EP4 receptor antagonist may be any suitable EP4 receptor antagonist.
  • suitable we mean that the antagonist is one which may be administered to the patient.
  • the receptor antagonists are molecules which bind to their respective receptors, compete with the natural ligand (PGE 2 ) and inhibit the initiation of the specific receptor-mediated signal transduction pathways.
  • the receptor antagonists are typically selective to the particular receptor and typically have a higher binding affinity to the receptor than the natural ligand. Although antagonists with a higher affinity for the receptor than the natural ligand are preferred, antagonists with a lower affinity may also be used, but it may be necessary to use these at higher concentrations.
  • the antagonists bind reversibly to their cognate receptor.
  • antagonists are selective for a particular receptor and do not affect the other receptor; thus, typically, an EP2 receptor antagonist binds the EP2 receptor but does not substantially bind the EP4 receptor, whereas an EP4 receptor antagonist binds the EP4 receptor but does not substantially bind the EP2 receptor.
  • the EP2 or EP4 receptor antagonist is selective for the particular receptor subtype.
  • the antagonist has a binding affinity for the particular receptor subtype which is at least ten- fold higher than for at least one of the other EP receptor subtypes.
  • selective EP4 receptor antagonists have at least a ten-fold higher affinity for the EP4 receptor than any of the EP1, EP2 or EP3 receptor subtypes.
  • the EP2 or EP4 receptor antagonist is selective for its cognate receptor.
  • the EP2 or EP4 receptor antagonists are typically administered in an effective amount to combat the pathological condition of the uterus.
  • the antagonists may be used to alleviate symptoms (ie are used palliatively) or may be used to treat the condition.
  • the antagonist may be administered prophylactically (and by "treating" we include prophylactic treatment).
  • the antagonist may be administered by any suitable route, and in any suitable form. It is desirable to administer an amount of the EP2 or EP4 receptor antagonist that is effective in preventing or alleviating or ameliorating or curing the pathological condition of the uterus.
  • EP2 receptor antagonists include AH6809 (Pelletier et al (2001) Br. J. Pharmacol. 132, 999-1008).
  • EP4 receptor antagonists include AH23848B (developed by Glaxo) and AH22921X (Pelletier et al (2001) Br. J. Pharmacol. 132, 999-1008.
  • the chemical name for AH23848B is ([lalpha(z), 2beta5alpha]-(+/-)-7-[5- [[(l,l'-biphenyl)-4-yl]methoxy]-2-(4-morph olinyl)-3-oxo-cyclopentyl]-4- heptenoic acid) (see Hillock & Crankshaw (1999) Eur. J. Pharmacol. 28, 99-108).
  • EP4RA (Li (2000) Endocrinology 141, 2054-61) is an EP(4) - selective ligand (Machwate et al (2001) Mol. Pharmacol. 60: 36-41).
  • the omega-substituted prostaglandin E derivatives described in WO 00/15608 (EP 1 114 816) (Ono Pharm Co Ltd) bind EP4 receptors selectively and may be EP4 receptor antagonists.
  • Peptides described in WO 01/42281 eg: IFTSYLECL, IFASYECL, IFTSAECL, IFTSYEAL, ILASYECL, IFTSTDCL, TSYEAL (with 4-biphenyl alanine), TSYEAL (with homophenyl alanine) are also described as EP4 receptor antagonists, as are some of the compounds described in WO 00/18744 (Fujisawa Pharm Co Ltd).
  • the 5-thia-prostaglandin E derivatives described in WO 00/03980 EP 1 097 922) (Ono Pharm Co Ltd) may be EP4 receptor antagonists.
  • EP4 receptor antagonists are also described in WO 01/10426 (Glaxo), 5 WO 00/21532 (Merck) and GB 2 330 307 (Glaxo).
  • EP4 receptor antagonists WO 00/21532 describes the following as EP4 receptor antagonists:
  • GB 2 330 307 describes [l ⁇ (Z), 2 ⁇ ,5 ⁇ ]-( ⁇ )-7-[5-[[(l,l '-biphenyl)-4- yl]methoxy]-2-(4-mo ⁇ holinyl)-3-oxocyclopentyl]-4-heptenoic acid and [lR[l ⁇ (z),2 ⁇ ,5 ⁇ ]]-(-)-7-[5-[[(l,l '-biphenyl)-4-yl]methoxy]-2-(4- 0 mo ⁇ holinyl)-3-oxocyclopentyl]-4-heptenoic acid.
  • WO 00/18405 (Pharmagene) describes the EP4 receptor antagonists AH22921 and AH23848 (which are also described in GB 2 028 805 and US 4, 342, 756).
  • WO 01/72302 (Pharmagene) describes further EP4 receptor antagonists, for example those described by reference to, and included in the general formula (I) shown on page 8 et seq.
  • EP2 receptor antagonists may be administered to the patient. It will also be appreciated that a combination of one or more EP2 or EP4 receptor antagonists may be administered to the patient.
  • the individual is additionally administered a COX-2 inhibitor.
  • the inhibitor is selective for COX-2.
  • the compound may selectively inhibit COX-2 function at any level.
  • the compound selectively inhibits COX-2 enzyme activity.
  • COX-2 enzyme activity we mean that the compound preferably inhibits COX-2 in preference to other cyclo-oxygenase enzymes, in particular in preference to cyclo-oxygenase-1 (COX-1).
  • COX-1 gene and the sequence of its polypeptide product are described in Yokoyama and Tanabe (1989) Biochem. Biophys. Res. Comm. 165, 888-894 incorporated herein by reference.
  • COX-1 is also called PGHS-1.
  • the COX-2 gene and the sequence of its polypeptide product are described in O'Banion et al (1991) J Biol. Chem. 266, 23261-23267 incorporated herein by reference.
  • COX-2 is also called PGHS-2.
  • the compound which selectively inhibits COX-2 enzyme activity is at least ten times better at inhibiting COX-2 than COX-1; preferably it is at least fifty times better; preferably it is at least one hundred times better; still more preferably it is at least one thousand times better and in greater preference it is at least ten thousand times better.
  • the compound has substantially no inhibitory activity against the COX-1 enzyme.
  • the compound selectively inhibits COX-2 enzyme production.
  • the compound may, for example, selectively prevent transcription of the COX-2 or it may selectively prevent translation of the COX-2 message.
  • selectively inhibits COX-2 enzyme production we mean that the compound preferably inhibits the production of COX-2 in preference to other cyclo-oxygenases, in particular in preference to the production of COX-1.
  • the compound which selectively inhibits COX-2 enzyme production is at least ten times better at inhibiting COX-2 production than COX-1 production; preferably it is at least fifty times better; more preferably it is at least one hundred times better; more preferably still it is at least one thousand times better; and in greater preference it is at least ten thousand times better. It is most preferred if the compound has substantially no inhibitory activity against COX-1 enzyme production.
  • a particularly preferred embodiment is wherein the compound is any one of nimesulide, 4-hydroxynimesulide, flosulide, and meloxicam.
  • Nimesulide is N-(4-nitro-2-phenoxyphenyl) methanesulfonamide (also called 4-nifro-2-phenoxymethanesulfonanilide). Nimesulide is 100-fold more specific for COX-2 inhibition than for COX-1 inhibition. Nimesulide is manufactured by Boehringer.
  • Flosulide is 6-(2,4-difluorophenoxy)-5-methyl sulphonylamino-1-indanone (also known as N-6-(2,4-difluorophenoxy)-l-oxo-indan-5-yl methane-sulphonamide). Flosulide is 1000-fold more specific for COX-2 inhibition than for COX-1 inhibition. Flosulide is manufactured by Ciba Geigy.
  • Meloxicam is 4-hydroxy-2-methyl-N-(5-methyl-2-thiazolyl)-2H- l,2-benzothiazine-3-carboxamide 1,1-dioxide. Meloxicam is 1000-fold more specific for COX-2 inhibition than for COX-1 inhibition. Meloxicam is manufactured by Boehringer.
  • COX-2-specific inhibitors which may be useful in the practice of the invention include: L 475 L337 which is 500-fold more specific for COX-2 inhibition than for COX-1 inhibition. This is manufactured by Merck Frost.
  • Vioxx sold by Merck, is also a suitable COX-2 inhibitor.
  • Celecoxib which is 100-fold more specific for COX-2 inhibition than for COX-1 inhibition. Celecoxib is manufactured by Searle.
  • DuP 697 which is COX-2-selective and is manufactured by DuPont.
  • Nimesulide, flosulide and meloxicam are COX-2 enzyme inhibitors, probably competitive inhibitors.
  • the treatment agents are administered in an effective amount to combat the undesired pathological condition of the uterus.
  • the treatment agents may be used to alleviate symptoms (ie are used palliatively) or may be used to treat the condition or may be used prophylactically to prevent the condition.
  • the treatment agent may be administered by any suitable route, and in any suitable form.
  • the aforementioned treatment agents for use in the invention or a formulation thereof may be administered by any conventional method including oral and parenteral (eg subcutaneous or intramuscular) injection.
  • the treatment may consist of a single dose or a plurality of doses over a period of time.
  • the dose to be administered is determined upon consideration of age, body weight, mode of administration, duration of the treatment and pharmacokinetic and toxicological properties of the treatment agent or agents.
  • the treatment agents are administered at a dose (or in multiple doses) which produces a beneficial therapeutic effect in the patient.
  • the treatment agents are administered at a dose the same as or similar to that used when the treatment agent is used for another medical indication.
  • the dose suitable for treatment of a patient may be determined by the physician.
  • a treatment agent of the invention Whilst it is possible for a treatment agent of the invention to be administered alone or in combination with other said treatment agents, it is preferable to present it or them as a pharmaceutical formulation, together with one or more acceptable carriers.
  • the carrier(s) must be "acceptable” in the sense of being compatible with the treatment agent of the invention and not deleterious to the recipients thereof.
  • the carriers will be water or saline which will be sterile and pyrogen free.
  • the formulations may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. Such methods include the step of bringing into association the treatment agent or agents with the carrier which constitutes one or more accessory ingredients. In general the formulations are prepared by uniformly and intimately bringing into association the active ingredient (ie treatment agent or agents) with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product.
  • Formulations in accordance with the present invention suitable for oral administration may be presented as discrete units such as capsules, cachets or tablets, each containing a predetermined amount of the active ingredient; as a powder or granules; as a solution or a suspension in an aqueous liquid or a non-aqueous liquid; or as an oil-in-water liquid emulsion or a water-in-oil liquid emulsion.
  • the active ingredient may also be presented as a bolus, electuary or paste.
  • a tablet may be made by compression or moulding, optionally with one or more accessory ingredients.
  • Compressed tablets may be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as a powder or granules, optionally mixed with a binder (eg povidone, gelatin, hydroxypropylmethyl cellulose), lubricant, inert diluent, preservative, disintegrant (eg sodium starch glycolate, cross-linked povidone, cross-linked sodium carboxymethyl cellulose), surface-active or dispersing agent.
  • Moulded tablets may be made by moulding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
  • the tablets may optionally be coated or scored and may be formulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydroxypropylmethylcellulose in varying proportions to provide desired release profile.
  • Formulations suitable for topical administration in the mouth include lozenges comprising the active ingredient in a flavoured basis, usually sucrose and acacia or tragacanth; pastilles comprising the active ingredient in an inert basis such as gelatin and glycerin, or sucrose and acacia; and mouth-washes comprising the active ingredient in a suitable liquid carrier.
  • buccal administration is also preferred.
  • Formulations suitable for parenteral administration include aqueous and non- aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents.
  • the formulations may be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze-dried (lyophilised) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use.
  • Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described.
  • Preferred unit dosage formulations are those containing a daily dose or unit, daily sub-dose or an appropriate fraction thereof, of an active ingredient.
  • formulations of this invention may include other agents conventional in the art having regard to the type of formulation in question, for example those suitable for oral administration may include flavouring agents.
  • EP2 and EP4 receptor antagonists are proteins or peptides. Proteins and peptides may be delivered using an injectable sustained-release drug delivery system. These are designed specifically to reduce the frequency of injections.
  • An example of such a system is Nutropin Depot which encapsulates recombinant human growth hormone (rhGH) in biodegradable microspheres that, once injected, release rhGH slowly over a sustained period.
  • the protein and peptide can be administered by a surgically implanted device that releases the drug directly to the required site.
  • Vitrasert releases ganciclovir directly into the eye to treat CMV retinitis.
  • Elecfroporation therapy (EPT) systems can also be employed for the administration of proteins and peptides.
  • a device which delivers a pulsed electric field to cells increases the permeability of the cell membranes to the drug, resulting in a significant enhancement of intracellular drag delivery.
  • Proteins and peptides can be delivered by electroinco ⁇ oration (El).
  • El occurs when small particles of up to 30 microns in diameter on the surface of the skin experience electrical pulses identical or similar to those used in elecfroporation. In El, these particles are driven through the stratum corneum and into deeper layers of the skin.
  • the particles can be loaded or coated with drugs or genes or can simply act as "bullets" that generate pores in the skin through which the drugs can enter.
  • ReGel injectable system An alternative method of protein and peptide delivery is the ReGel injectable system that is thermo-sensitive. Below body temperature, ReGel is an injectable liquid while at body temperature it immediately forms a gel reservoir that slowly erodes and dissolves into known, safe, biodegradable polymers. The EP2 or EP4 receptor antagonist is delivered over time as the biopolymers dissolve.
  • Protein and peptide pharmaceuticals can also be delivered orally.
  • the process employs a natural process for oral uptake of vitamin B 12 in the body to co-deliver proteins and peptides. By riding the vitamin B 12 uptake system, the protein or peptide can move through the intestinal wall.
  • Complexes are synthesised between vitamin B 12 analogues and the drug that retain both significant affinity for intrinsic factor (IF) in the vitamin B 12 portion of the complex and significant bioactivity of the drug portion of the complex.
  • IF intrinsic factor
  • Proteins and polypeptides can be introduced to cells by "Trojan peptides". These are a class of polypeptides called penetratins which have translocating properties and are capable of carrying hydrophilic compounds across the plasma membrane. This system allows direct targetting of oligopeptides to the cytoplasm and nucleus, and may be non-cell type specific and highly efficient. See Derossi et al (1998), Trends Cell Biol 8, 84-87.
  • the treatment agent (or agents) is administered orally. It is further preferred if the treatment agent (or agents) is administered to the female reproductive system.
  • the treatment agent or agents may suitably be administered intravaginally using, for example, a gel or cream or vaginal ring or tampon.
  • the treatment agent may also advantageously be administered by intrauterine delivery, for example using methods well known in the art such as an intrauterine device.
  • the gel or cream is one which is formulated for administration to the vagina. It may be oil based or water based.
  • the treatment agent or agents is present in the cream or gel in a sufficient concentration so that an effective amount is administered in a single (or in repeated) application.
  • the vaginal ring comprises a polymer which formed into a "doughnut" shape which fits within the vagina.
  • the treatment agent (or agents) is present within the polymer, typically as a core, which may dissipate through the polymer and into the vagina and/or cervix in a controlled fashion.
  • Vaginal rings are known in the art.
  • the tampon is impregnated with the treatment agent (or agents) and that a sufficient amount of the treatment agent (or agents) is present in the tampon.
  • the intrauterine device is for placing in the uterus over extended periods of time, such as between one and five years.
  • the intrauterine device comprises a plastic frame, often in the shape of a "T” and contains sufficient antagonist to be released over the period of use.
  • the antagonist is generally present within or encompassed by a slow-release polymer which forms part of the device, such as in the form of a "sausage" of antagonist which wraps around the long arm of the "T" which is typically covered with a controlled-release membrane.
  • Intrauterine devices are known in the art.
  • the individual to be treated may be any female individual who would benefit from such treatment.
  • the individual to be treated is a human female.
  • the methods of the invention may be used to treat female mammals, such as the females of the following species: cows, horses, pigs, sheep, cats and dogs.
  • the methods have uses in both human and veterinary medicine.
  • a second aspect of the invention provides use of any one or more of an inhibitor of prostaglandin E synthase (PGES), or an EP2 or EP4 receptor antagonist in the manufacture of a medicament for treating or preventing a pathological condition of the uterus.
  • PGES prostaglandin E synthase
  • EP2 or EP4 receptor antagonist in the manufacture of a medicament for treating or preventing a pathological condition of the uterus.
  • the medicament may usefully further comprise an inhibitor of COX-2.
  • the medicament is for administration to a patient who is administered a COX-2 inhibitor and/or an inhibitor of PGES and/or an EP2 or EP4 receptor antagonist.
  • the patient may have been administered one of the treatment agent or agents previously, or is administered them simultaneously or is administered them after the treatment agent or agents present in the medicament.
  • FIG. 1 Ribonuclease protection assay conducted using 10 ⁇ g of total RNA extracted from normal secretory phase endometrium (N) and well (W), moderately (M) and poorly (P) differentiated endometrial adenocarcinoma tissue. COX-2 expression was detected using a 381 homologous cRNA probe. The integrity of the RNA and the relative amount of total RNA in each reaction were determined using a ribosomal 18S cDNA probe.
  • Figure 3 mPGES expression is detected in epithelial cells of poorly ( Figure a), moderately ( Figure b) and well (Figure c) differentiated endometrial adenocarcinoma. Minimal immunostaining for mPGES was detected in post- menopausal uterus and secretory phase endometrium ( Figure d and e respectively). Inset in Figure c is a section that was stained with mPGES pre-adsorbed serum (negative control). Scale bar is 100 ⁇ m.
  • Figure 4. COX-2 ( Figure a), mPGES ( Figure b), PGE 2 ( Figure c) and EP4 ( Figure d) are detected in endothelial cells of all carcinoma tissues.
  • Vascular endothelial cells in endometrial adenocarcinoma were localised using antibodies raised against the human CD34 endothelial cell marker (Figure e).
  • the inset in Figure e is a section that was stained with non- immune goat serum (CD34 negative control). Negative controls for the other antibodies are presented in Figures 2, 3 and 5B. Scale bar is 50 ⁇ m.
  • Figure 5 A. Relative expression of EP2 and EP4 receptors in endometrial adenocarcinoma of different grades of differentiation and in healthy secretory phase endometrium collected from fertile women with normal menstrual cycles.
  • the inset in Figure c is a section that was stained with immune serum that had been pre-adsorbed with the blocking peptide. Scale bar is 50 ⁇ m.
  • Example 1 Expression of COX-2 and PGE synthase and synthesis of PGE 2 in endometrial adenocarcinoma: a possible autocrine/paracrine regulation of neoplastic cell function via EP2 EP4 receptors
  • COX-2 cyclooxygenase-2
  • PGE 2 prostaglandin E 2
  • COX-2 RNA expression was confirmed in various grades of adenocarcinoma by ribonuclease protection assay.
  • COX-2 and microsomal glutathione-dependent prostaglandin E synthase (mPGES) expression and PGE 2 synthesis were localised to the neoplastic epithelial cells and endothelial cells.
  • Endometrial adenocarcinoma tissue was collected from women undergoing hysterectomy and who had been pre- diagnosed to have adenocarcinoma of the uterus. All women with endometrial adenocarcinoma were post-menopausal. To provide control tissue, normal secretory phase (Days 18-25 of the menstrual cycle) endometrial tissue was collected with a pipelle suction curette (Pipelle; Laboratoire CCD, Paris, France) from fertile women with regular menstrual cycles, undergoing gynecological procedures for benign conditions. Biopsies were dated from the patient's last menstrual period (LMP) and histological dating was consistent with date of LMP.
  • LMP menstrual period
  • the tissue was either snap frozen in dry ice and stored at -70°C (for RNA extraction), fixed in Neutral buffered formalin and wax embedded (for immunohistochemical analyses) or placed in RPMI 1640 (containing 2mmol/L L-glutamine, 100 U penicillin and 100 ⁇ g/mL streptomycin) and transported to the laboratory for in vitro culture.
  • RPMI 1640 containing 2mmol/L L-glutamine, 100 U penicillin and 100 ⁇ g/mL streptomycin
  • archival tissue blocks of healthy post-menopausal uterus were obtained from The Department of Pathology (The University of Edinburgh Medical School) and utilised for immunohistochemical analyses.
  • Written informed consent was obtained prior to tissue collection and ethical approval was received from the Lothian Research Ethics Committee. The data in this study were analysed by ANOVA using StatView 5.0.
  • a homologous 381 bp COX-2 cDNA probe was generated by PCR from a clone containing the human COX-2 cDNA (the clone was a gift from Dr S Prescott, University of Utah) and primers at base pair position 950-971 (COX2A: 5'-CAAGCAGGCTAATACTGATAGG- 3') and 1310-1331 (COX2B: 5'-ATCTGCCTGCTCTGGTCAATGG-3').
  • the amplified PCR product was subcloned into pCRII and its identity and orientation confirmed following sequencing using the Applied Biosystems 373A DNA sequencer and the ABI prism DNA sequencing kit (Applied Biosystems, Cheshire, UK).
  • an antisense cRNA probe was prepared from Hindlll linearised pCRII plasmid containing the 381 bp cDNA fragment of the human COX-2.
  • the RPA was conducted using the Ambion RPA II kit (AMS Biotechnology Europe, Oxfordshire, UK) as previously reported (Jabbour et al., 1998). Briefly, radiolabelled cRNA was generated using the linearised plasmid, T7 RNA polymerase and ⁇ 32p. ⁇ jTP (800 Ci/mmol; Amersham, Buckinghamshire, UK).
  • Five-micron paraffin wax-embedded tissue sections were dewaxed in xylene, rehydrated in graded ethanol and washed in water followed by TBS (50 mM Tris-HCl, 150 mM NaCl pH 7.4) and blocked for endogenous peroxidase (3% H2O2 in methanol).
  • tissue sections were incubated with polyclonal goat anti-COX-2 antibody (sc-1745; Autogenbioclear, Wilts, UK) at a dilution of 1:400, polyclonal rabbit anti-mPGES antibody (catalogue number 160140; Cayman Chemical, Alexis Co ⁇ oration-Europe, Nottingham, UK) at a dilution of 1 :50, polyclonal rabbit anti-EP4 receptor (catalogue number 101770; Cayman Chemical) at a dilution of 1 :500, polyclonal rabbit anti- PGE 2 antibody (kindly supplied by Professor RW Kelly, MRC Human Reproductive Sciences Unit, Edinburgh, UK) at a dilution of 1:100 or monoclonal mouse anti-human CD34 primary antibody (mca-547; Serotec
  • Control tissue was incubated with either 5% non-immune antisera (CD34), goat anti-COX-2 antibody pre-adsorbed to blocking peptide (sc-1745p; Autogenbioclear), rabbit anti-mPGES antibody pre-adsorbed to blocking peptide (catalogue number 360140; Cayman Chemical), rabbit anti-EP4 pre-adsorbed to blocking peptide (catalogue number 101780; Caman Chemical) or rabbit anti-PGE 2 antibody pre-adsorbed to 10-fold excess PGE 2 (Sigma).
  • tissue sections probed with the goat anti-human COX-2, rabbit anti-mPGES, rabbit anti- EP4 and rabbit anti-PGE 2 primary antibodies were incubated with biotinylated rabbit anti-goat secondary IgG antibody (for COX-2; Dako, Bucks, UK) or swine anti-rabbit secondary IgG antibody (for mPGES, EP4 and PGE 2 ; Dako) at a dilution of 1 :500 for 40 min at 25°C. Thereafter the tissue sections were incubated with streptavidin-biotin peroxidase complex (Dako) for 20 min at 25°C. Tissue sections probed with the mouse anti- human CD34 antibody were developed using a Mouse EnVision Kit (Dako) as instructed by the manufacturer. Colour reaction was developed by incubation with 3.3'-diaminobenzidine (Dako).
  • RNA samples were reverse transcribed using MgCl 2 (5.5 mM), dNTPs (0.5 mM each), random hexamers (2.5 ⁇ M), RNAase inhibitor (0.4 U/ ⁇ l) and multiscribe reverse transcriptase (1.25 U/ ⁇ l; all from PE Biosystems, Warrington, UK). The mix was ali quoted into individual tubes (16 ⁇ l/tube) and template RNA was added (4 ⁇ l/tube of 100 ng/ ⁇ l RNA). Samples were incubated for 60 minutes at 25°C, 45 minutes at 48°C and then at 95°C for 5 minutes.
  • a reaction mix was made containing Taqman buffer (5.5 mM MgCl 2 , 200 ⁇ M dATP, 200 ⁇ M dCTP, 200 ⁇ M dGTP, 400 ⁇ M dUTP), ribosomal 18S forward and reverse primers and probe (all at 50 nM), forward and reverse primers for EP receptor (300 nM), EP receptor probe (200 nM), AmpErase UNG (0.01 U/ ⁇ l) and AmpliTaq Gold DNA Polymerase (0.025 U/ ⁇ l; all from PE Biosystems).
  • a volume of 48 ⁇ l of reaction mix was aliquoted into separate tubes for each cDNA sample and 2 ⁇ l/replicate of cDNA was added.
  • EP receptor primers and probe for quantitative PCR were designed using the PRIMER express program (PE Biosystems). The sequence of the EP2 receptor primers and probe were as follows; Forward: 5'-GAC CGC TTA CCT GCA GCT GTA C-3'; Reverse: 5'-TGA AGT TGC AGG CGA GCA-3'; Probe (FAM labelled): 5'-CCA CCC TGC TGC TGC TTC TCA TTG TCT-3'.
  • the sequence of the EP4 receptor primers and probe were as follows; Forward: 5'-ACG CCG CCT ACT CCT AC A TG-3'; Reverse: 5'-AGA GGA CGG TGG CGA GAA T-3'; Probe (FAM labelled): 5'-ACG CGG GCT TCA GCT CCT TCC T-3'.
  • the ribosomal 18S primers and probe sequences were as follows; Forward: 5' -CGG CTA CCA CAT CCA AGG AA-3'; Reverse: 5'-GCT GGA ATT ACC GCG GCT-3'; Probe (VIC labelled): 5'-TGC TGG CAC CAG ACT TGC CCT C-3'.
  • IBMX IBMX
  • This phase of the menstrual cycle was chosen as minimal proliferative activity is detected in the endometrium during the secretory phase (Ferenczy et al., 1979). This would be comparable to the absence of proliferative activity predicted in healthy post-menopausal endometrium.
  • EP2 and EP4 The expression of two subtypes of PGE 2 receptors, namely EP2 and EP4, was investigated by real-time quantitative PCR in carcinoma tissue and normal secretory phase endometrium. Expression of both receptors was significantly up-regulated in adenocarcinoma tissues compared with normal secretory endometrium (P ⁇ 0.01). No differences in the level of expression of EP2 or EP4 receptors were detected between poorly, moderately or well differentiated adenocarcinomas (Figure 5 A). Overall, the fold induction of EP2 and EP4 receptor expression in adenocarcinoma tissue (mean fold induction of all carcinoma samples) compared with normal secretory endometrium was 28.0 + 7.4 and 52.5 + 10.1 for EP2 and EP4 receptors respectively. Using immunohistochemistry, EP4 receptor expression was localised to neoplastic epithelial cells of carcinoma tissues of all grades of differentiation (Figure 5B) and also in endothelial cells of the microvasculature ( Figure 4d).
  • cAMP generation was measured following short term in vitro culture with or without PGE 2 ( Figure 6). Comparable cAMP turnover in response to PGE 2 was observed in all carcinoma tissue. The fold induction of cAMP generation in response to PGE 2 was significantly higher in carcinoma tissue compared with secretory phase endometrium (3.42 + 0.46 vs 1.15 + 0.05 respectively; PO.001).
  • COX-2 enzyme observed in endometrial adenocarcinomas resembles that reported for a number of other carcinomas including colon, lung, bladder, stomach, pancreas, prostate and cervix (Gupta et al., 2000; Mohammed et al., 1999; Ratnasinghe et al., 1999; Sales et al., 2001; Tsujii et al., 1997; Tucker et al., 1999; Wolff et al., 1998).
  • the exact intracellular signalling pathways that lead to up-regulation in COX-2 expression in carcinomas remain to be elucidated.
  • COX-2 expression and PGE 2 synthesis can induce neoplastic changes in epithelial cells through a number of biological pathways. These include promotion of cellular proliferation, inhibition of apoptosis, increasing metastatic potential of neoplastic cells and promoting angiogenesis (Rolland et al., 1980; Tsuji et al., 1996; Tsujii & DuBois, 1995). Over-expression of COX-2 enzyme in rat intestinal epithelial cells results in enhanced secretion of PGE 2 which is associated with increased cellular proliferation and resistance to apoptosis (Tsujii & DuBois, 1995).
  • E-Cadherin cell adhesion molecules
  • Expression of E-Cadherin is down regulated in a number of solid tumours and is closely and inversely related to enhanced invasion of neoplastically transformed cells (Mayer et al, 1993; Schipper et al., 1991).
  • COX-2 and PGE 2 are strongly linked with regulation of the angiogenic process during tumour development (Masferrer et al., 2000). Over-expression of COX-2 and increased production of PGE 2 in epithelial cells enhances the expression of angiogenic factors which act in a paracrine manner to induce endothelial cell migration and microvascular tube formation (Tsujii et al., 1998). Similarly, COX-2 and PGE 2 may influence angiogenesis directly by acting on endothelial cells.
  • PGE 2 acts on target cells through interaction with seven transmembrane G- protein coupled receptors. Different forms of the membrane bound receptors have been cloned which utilise alternate intracellular signalling pathways. In this study we investigated the expression of two of the membrane bound PGE 2 receptors, namely EP2 and EP4, which mediate their effect on target cells via the PKA pathway by activating adenylate cyclase and increasing intracellular cAMP (Coleman et al., 1994).
  • EP2 and EP4 receptors are up-regulated in comparison with normal secretory phase endometrium and expression of at least the EP4 receptor is localised to neoplastic epithelial cells and the endothelium of the microvasculature. It was not possible to conduct parallel studies to localise EP2 receptors in the carcinoma tissues as no commercial antibodies are available for this receptor. Hence it remains to be established whether these receptors are co-expressed in the same cell type. However, using in situ hybridisation techniques, EP2 and EP4 receptor expression have been recently co-localised to epithelial and endothelial cells of the normal human endometrium (Milne et al., 2001).
  • COX-2/mPGES and PGE 2 may mediate proliferation of epithelial and/or endothelial cells.
  • the proliferating cells within endometrial adenocarcinomas are detected predominantly in post-menopausal women at a time when the healthy endometrium is expected to atrophy and display minimal cellular proliferation or angiogenesis.
  • healthy post-menopausal uterus and normal secretory endometrium both of which have minimal proliferative or angiogenic activity, display negligible COX- 2/mPGES/EP2/EP4 receptor expression and minimal cAMP generation in response to treatment with PGE 2 .
  • PGE 2 in proliferation has already been established in a number of cell types including endothelial cells and it has been suggested that this effect is mediated via cAMP and induction of expression of mitogenic growth factors such as vascular endothelial growth factor and basic fibroblast growth factor (Cheng et al., 1998; Hoper et ah, 1997). Future studies will elucidate the exact role of PGE 2 and its associated receptors on proliferation and neoplastic differentiation of epithelial/endothelial cells in endometrial adenocarcinomas .
  • Example 2 Treatment of uterine cancer with an EP2 receptor antagonist
  • a patient suffering from uterine cancer is administered AH6809.
  • the patient is also administered meloxicam.
  • a patient suffering from fibroids is administered AH23848B.
  • a patient suffering from endometriosis is administered AH22921X.
  • the patient is also administered nimesulide.
  • Pharmacology classification of prostanoid receptors properties, distribution, and structure of the receptors and their subtypes.
  • HGF HGF triggers activation of the COX-2 gene in rat gastric epithelial cells: action mediated through the ERK2 signaling pathway. Faseb J 13: 2186-2194.
  • Prostaglandin in human breast cancer Evidence suggesting that an elevated prostaglandin production is a marker of high metastatic potential for neoplastic cells. JNatl Cancer Inst 64: 1061-1070. Sales, KJ, Katz, AA, Davis, M, Hinz, S, Soeters, RP, Hofmeyr, MD, Millar, RP, Jabbour, HN. (2001) Cyclooxygenase-2 Expression and Prostaglandin E 2 Synthesis Are Up- Regulated in Carcinomas of the
  • Tsujii, M, Kawano, S, DuBois, RN (1997) Cyclooxygenase-2 expression in human colon cancer cells increases metastatic potential. Proc Natl Acad Sci USA 94: 3336-3340. Tsujii, M, Kawano, S, Tsuji, S, Sawaoka, H, Hori, M, DuBois, RN. (1998) Cyclooxygenase regulates angiogenesis induced by colon cancer cells. Cell 93: 705-716.

Abstract

La présente invention concerne une méthode de traitement ou de prévention d'une affection de l'utérus chez un individu, ladite méthode consistant à administrer à l'individu au moins un inhibiteur de la prostaglandine E synthase (PGES), ou un antagoniste du récepteur EP2 ou EP4. Eventuellement, on administre un inhibiteur de COX-2 au patient. Généralement, l'affection est un cancer de l'utérus, des fibromes ou une endométriose.
EP02765100A 2001-10-08 2002-10-08 Utilisation d'inhibiteurs de la prostaglandine e synthase ou d'antagonistes du recepteur ep2 ou ep4 dans le traitement d'une affection de l'uterus Ceased EP1467738A1 (fr)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
GBGB0124124.9A GB0124124D0 (en) 2001-10-08 2001-10-08 Methods of treatment
GB0124124 2001-10-08
US33356201P 2001-11-27 2001-11-27
US333562P 2001-11-27
PCT/GB2002/004549 WO2003030911A1 (fr) 2001-10-08 2002-10-08 Utilisation d'inhibiteurs de la prostaglandine e synthase ou d'antagonistes du recepteur ep2 ou ep4 dans le traitement d'une affection de l'uterus

Publications (1)

Publication Number Publication Date
EP1467738A1 true EP1467738A1 (fr) 2004-10-20

Family

ID=26246623

Family Applications (1)

Application Number Title Priority Date Filing Date
EP02765100A Ceased EP1467738A1 (fr) 2001-10-08 2002-10-08 Utilisation d'inhibiteurs de la prostaglandine e synthase ou d'antagonistes du recepteur ep2 ou ep4 dans le traitement d'une affection de l'uterus

Country Status (2)

Country Link
EP (1) EP1467738A1 (fr)
WO (1) WO2003030911A1 (fr)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2149554A1 (fr) 2008-07-30 2010-02-03 Bayer Schering Pharma Aktiengesellschaft Indolylamides en tant que modulateurs du récepteur EP2
EP2149552A1 (fr) 2008-07-30 2010-02-03 Bayer Schering Pharma AG Dérivés de benzamide 5,6 substitués en tant que modulateurs du récepteur EP2
EP2149551A1 (fr) 2008-07-30 2010-02-03 Bayer Schering Pharma AG Dérivés de N-(indol-3-ylalkyl)-(hétéro)arylamide en tant que modulateurs du récepteur EP2
DE102009049662A1 (de) 2009-10-13 2011-04-14 Bayer Schering Pharma Aktiengesellschaft 2,5-Disubstituierte 2H-Indazole als EP2-Rezeptor-Antagonisten
WO2013079425A1 (fr) 2011-11-28 2013-06-06 Bayer Intellectual Property Gmbh Nouveaux 2h-indazoles en tant qu'antagonistes du récepteur ep2

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1900731A1 (fr) * 2006-09-07 2008-03-19 Bayer Schering Pharma Aktiengesellschaft N-(1-Phthalazin-1-yl-piperidin-4-yl)-amides comme modulateurs du récepteur EP2
US20110287112A1 (en) 2009-01-30 2011-11-24 Ono Pharmaceutical Co., Ltd. Prostate cancer progression inhibitor and progression inhibition method
GB202212749D0 (en) * 2022-09-01 2022-10-19 Gesynta Pharma Ab New uses

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TWI249520B (en) * 1998-07-15 2006-02-21 Ono Pharmaceutical Co 5-Thia-omega-substituted phenyl prostaglandin E derivatives, method for producing the same and medicines containing the same as the active ingredient
WO2000015608A1 (fr) * 1998-09-14 2000-03-23 Ono Pharmaceutical Co., Ltd. Derives e de phenyl-prostaglandine a substitution-φ et medicaments les contenant comme ingredient actif
CA2346038A1 (fr) * 1998-10-15 2000-04-20 Merck & Co., Inc. Inhibition de la resorption osseuse
MXPA02005626A (es) * 1999-12-06 2004-09-10 Pital Sainte Justine H Composiciones para el tratamiento de filtracion glomerular, conductos arteriosos y osteoporosis del paciente.

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
"Table of Contents", BRITISH JOURNAL OF CANCER, vol. 85, no. 7, 2 October 2001 (2001-10-02), pages 937 - 1983, Retrieved from the Internet <URL:http://www.nature.com/bjc/journal/v85/n7/index.html> [retrieved on 20090423] *
DATABASE MEDLINE [online] 10 October 2001 (2001-10-10), JABBOUR H.N. ET AL.: "Expression of COX-2 and PGE synthase and synthesis of PGE2 in endometrial adenocarcinoma: A possible autocrine/paracrine regulation of neoplastic cell receptors", XP003004332, accession no. ncbi Database accession no. 11592775
DATABASE PUBMED [online] 28 September 2001 (2001-09-28), JABBOUR ET AL.: "Expression of COX-s and PGE synthase and synthesis of PBE(2) in endometrial adenocarcinoma:a possible autocrine/paracrine regulation of neoplastic cell function via EP2/EP4 receptors", Database accession no. 11592775 *
See also references of WO03030911A1 *

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2149554A1 (fr) 2008-07-30 2010-02-03 Bayer Schering Pharma Aktiengesellschaft Indolylamides en tant que modulateurs du récepteur EP2
EP2149552A1 (fr) 2008-07-30 2010-02-03 Bayer Schering Pharma AG Dérivés de benzamide 5,6 substitués en tant que modulateurs du récepteur EP2
EP2149551A1 (fr) 2008-07-30 2010-02-03 Bayer Schering Pharma AG Dérivés de N-(indol-3-ylalkyl)-(hétéro)arylamide en tant que modulateurs du récepteur EP2
DE102009049662A1 (de) 2009-10-13 2011-04-14 Bayer Schering Pharma Aktiengesellschaft 2,5-Disubstituierte 2H-Indazole als EP2-Rezeptor-Antagonisten
WO2013079425A1 (fr) 2011-11-28 2013-06-06 Bayer Intellectual Property Gmbh Nouveaux 2h-indazoles en tant qu'antagonistes du récepteur ep2

Also Published As

Publication number Publication date
WO2003030911A1 (fr) 2003-04-17

Similar Documents

Publication Publication Date Title
US20100035868A1 (en) Methods of treatment of uterine pathological conditions
Jabbour et al. Expression of COX-2 and PGE synthase and synthesis of PGE2 in endometrial adenocarcinoma: a possible autocrine/paracrine regulation of neoplastic cell function via EP2/EP4 receptors
Soares Etiology of OHSS and use of dopamine agonists
Koki et al. Potential utility of COX-2 inhibitors in chemoprevention and chemotherapy
Fosslien Biochemistry of cyclooxygenase (COX)-2 inhibitors and molecular pathology of COX-2 in neoplasia
Fujita et al. Blockade of angiotensin AT1a receptor signaling reduces tumor growth, angiogenesis, and metastasis
Amano et al. Host prostaglandin E2-EP3 signaling regulates tumor-associated angiogenesis and tumor growth
Cherukuri et al. The EP4 receptor antagonist, L-161,982, blocks prostaglandin E2-induced signal transduction and cell proliferation in HCA-7 colon cancer cells
US6239137B1 (en) Salts of aminoimidazole carboxamide and 5 amino or substituted amino 1,2,3-triazole, induce apoptosis, inhibit DNA synthesis and control cyclooxygenase activity
Banno et al. Progestin therapy for endometrial cancer: the potential of fourth-generation progestin
JP2020019780A (ja) B−Raf阻害薬、EGFR阻害薬及び場合によってはPI3K−α阻害薬を含む組合せ医薬
Sookvanichsilp et al. Anti-implantation effects of indomethacin and celecoxib in rats
CA2357525A1 (fr) Traitement de cancers associe a la surexpression de her-2/neu
JP2008546754A (ja) プロテインキナーゼモジュレーターとしてのピラゾール誘導体を含む医薬組み合わせ
US20190231735A1 (en) Methods for treating cancer
EP1450813B1 (fr) Antagonistes de recepteurs de prostaglandine ep2 et/ou ep4 pour le traitement de la menorragie
WO2003030911A1 (fr) Utilisation d&#39;inhibiteurs de la prostaglandine e synthase ou d&#39;antagonistes du recepteur ep2 ou ep4 dans le traitement d&#39;une affection de l&#39;uterus
US20070004620A1 (en) Fp receptor antagonists or pgf2 alpha antagonists for treating pathological conditions of the uterus
Divvela et al. Pathogenic role of cyclooxygenase-2 in cancer
US20060171945A1 (en) Ip receptor antagonists for the treatment of pathological uterine conditions
Half et al. Anti-EGFR and ErbB-2 antibodies attenuate cyclooxygenase-2 expression and cooperatively inhibit survival of human colon cancer cells
US20030220266A1 (en) Method of treating a disease
Perchick et al. Cyclooxygenase-2 overexpression inhibits cathepsin D-mediated cleavage of plasminogen to the potent antiangiogenic factor angiostatin
US20060166872A1 (en) Fp receptor antagonists or pgf2 alpha antagonists for treating menorrhagia
Cahlin et al. The effects of non-selective, preferential-selective and selective COX-inhibitors on the growth of experimental and human tumors in mice related to prostanoid receptors

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20040812

AK Designated contracting states

Kind code of ref document: A1

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

AX Request for extension of the european patent

Extension state: AL LT LV MK RO SI

17Q First examination report despatched

Effective date: 20050203

17Q First examination report despatched

Effective date: 20050203

TPAC Observations filed by third parties

Free format text: ORIGINAL CODE: EPIDOSNTIPA

TPAC Observations filed by third parties

Free format text: ORIGINAL CODE: EPIDOSNTIPA

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

Free format text: STATUS: THE APPLICATION HAS BEEN REFUSED

18R Application refused

Effective date: 20090805