WO2000001716A2 - Vasculoprotector - Google Patents

Vasculoprotector Download PDF

Info

Publication number
WO2000001716A2
WO2000001716A2 PCT/GB1999/002157 GB9902157W WO0001716A2 WO 2000001716 A2 WO2000001716 A2 WO 2000001716A2 GB 9902157 W GB9902157 W GB 9902157W WO 0001716 A2 WO0001716 A2 WO 0001716A2
Authority
WO
WIPO (PCT)
Prior art keywords
agonist
vasculoprotective
effect
erβ
subject
Prior art date
Application number
PCT/GB1999/002157
Other languages
French (fr)
Other versions
WO2000001716A3 (en
Inventor
Pekka Hayry
Sari Makela
Jan-Ake Gustafsson
Original Assignee
Karo Bio Ab
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 Karo Bio Ab filed Critical Karo Bio Ab
Priority to EP99929579A priority Critical patent/EP1094806A2/en
Priority to CA002331885A priority patent/CA2331885A1/en
Priority to AU46364/99A priority patent/AU4636499A/en
Priority to JP2000558117A priority patent/JP2002519435A/en
Priority to KR1020017000141A priority patent/KR20010079495A/en
Publication of WO2000001716A2 publication Critical patent/WO2000001716A2/en
Publication of WO2000001716A3 publication Critical patent/WO2000001716A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/35Heterocyclic 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
    • 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 
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis

Abstract

The invention relates to a method of inducing a vasculoprotective effect, particularly relating to restonosis, angioplasty, chronic allograft rejection, diabetic angiopathy, autoimmune angiopathy, ateriosclerosis and atherosclerosis in a subject, the method comprising treating the subject with an ERbeta agonist. The preferred Estrogen Receptor beta agonist is genistein.

Description

VASCULOPROTECTOR
This invention relates to the use of ligands of the second estrogen receptor, ERβ, or compounds which affect ERβ for vasculoprotection, that is to say in inducing protective effects m the vascular wall particularly in fibroproliferative disorders (such as atherosclerosis, ateπosclerosis, diabetic and autoimmune angiopathies), after injury (such as restenosis after angioplasty and bypass surgery) and m chronic allograft rejection.
Recently, a second estrogen receptor, ERβ, has been revealed (WO97/09348)
Vascular mtimal dysplasia and remodelling are characteπstic features of remjury following balloon angioplasty, coronary bypass surgery (Holmes, D. et al. Am J Cardiol (1984); 53: 77C-81C, Holmes, D et al J Am Coll Cardiol (1988); 22 1 149-55), and in chronic allograft rejection (Lemstrom KB, Koskinen PK Circulation (1997); 96' 1240-1249, Hayry, P et al. Immunol Rev (1993) Aug; 134: 33-81, Hayry P et al. Faseb J (1993); 7: (11) 1055-60) The initial response to vascular injury is inflammatory and involves the attraction of lymphocytes, macrophages and thrombocytes to the site of injury and the secretion of cytokines, eikosanoids and growth factors (Ross. R. Nature (1993); 362(6423)' 801-9). Under the influence of growth factors and cytokines, smooth muscle cells (SMC) proliferate and migrate from the media into the intima and contπbute to mtimal hyperplasia and stenosis
Estrogen has several protective effects on the vascular wall (Farhat MY et al. J Pharmacol Exp Ther (1996), 276- 652-7). Some of these are rapid, presumably direct membrane effects, whereas others require transcπptional activation of genes (Farhat MY. et al. Biochem Pharmacol (1996); 51(5)- 571-6). The inhibitory effect of estrogen on the replication, migration and extracellular matrix deposition by vascular smooth muscle cells, the key event m vascular fibroproliferative dysplasias, is presumably a genomic effect mediated through a vaπety of mechanisms including regulation of several growth factors and/or their receptors and possibly by a direct antiprohferative effect of estrogen on smooth muscle cells (Farhat MY, et al. Faseb J (1996); 10(5): 615-24) The vasculoprotective effect of estrogen was first demonstrated in population studies in humans, where estrogen replacement therapy demonstrated a protective effect on atherosclerotic vascular disease in post menopausal women (Stampfer M J et al (1991) N Engl J Med 325 756-762, Grady D et al (1992) Ann Intern Med 111 1016-1037), as later confirmed in monkeys (Wagner JD et al Metabolism (1997), 46(6) 698-705) Later, the vasculoprotective effect has been documented m more detail in animal models and in vitro Estrogen has been found to inhibit the intimal thickening after mechanical carotid balloon injury in rabbits (Foegh ML et al J Vase Surg (1994), 19(4) 722-6), rats (Chen SJ et al Circulation (1996), 93(3) 577-84) and in mice (Sullivan TJ et al J Clin Invest (1995), 96 2482-8), as well as the immunologically-mduced vascular fibroproliferative dysplasia in rabbit aorta (Cheng LP, et al Transplantation (1991), 52(6) 967-72) and heart (Foegh ML et al Transplant Proc (1987) 90-5) allogratts In vitro, it has been demonstrated that estrogen inhibits migration and replication of vascular smooth muscle cells (Akishita M et al Atherosclerosis (1997), 130(1-2) 1-10, Kolodgie FD, et al Am J Pathol (1996), 148(3) 969-76, Morev AK, et al Endocπnology (1997), 138(8) 3330-9, Suzuki A, et al Cardiovasc Res (1996), 32(3) 516-23) These observations are consistant with findings in reporter gene assays that functional estrogen receptors are expressed in vascular smooth muscle cells of bovine (Balica M et al Circulation (1997), 95(7) 1954-60, rat ((Bayard F et al Endocrinology (1995), 136(4) 1523-9 Bayard F et al Ciba Found Symp (1995), 191(122) 122-32. Bei M et al J Steπod Biochem Mol Biol (1996), 58(1) 83-8), guinea pig (Bhalla RC, et al Am J Physiol (1997) H1996-2003), and human (Karas RH. et al Febs Lett (1995), 377(2) 103-8) origin
In addition to being anti-prohferative and anti-migratory to smooth muscle cells, estrogen may also display vasculoprotective effects via the vascular wall endothehum Functional estrogen receptors have been demonstrated in endothehal cells (Venkov CD et al Circulation (1996), 94 727-33) Estrogen downregulates cytokine-induced adhesion molecule expression in human endothehum in vitro (Cauhn GT et al J Chn Invest (1996), 98(1) 36-42), it is anti-apoptotic to endothehal cells (Spyπdopoulos, I et al Circulation (1997), 95(6) 1505-14) and it enhances functional endothehal recovery after denudation assay in vivo (Krasmski K et al Circulation (1997), 95(7) 1768-72) There are also additional, indirect pathways whereby estrogen may mediate vasculoprotective effects In addition to being directly anti-prohferative to smooth muscle cells and protective to the vascular endothehal cells, the estrogen effect may be mediated indirectly via hpoprotem metabolism and via promoting vasodilation by stimulating prostacyclm and nitric oxide synthesis and via regulation of the cell membrane voltage-dependent calcium channels resulting in inhibition of extracellular calcium mobilization and flux (see Farhat MY et al. J Pharmacol Exp Ther (1996); 276(2): 652-7, Farhat MY. et al Biochem Pharmacol (1996); 51(5). 571-6) None of these mechanisms alone explain the beneficial effect of estrogen. For example, although the endothehal nitπc oxide synthetase (e-NOS) mRNA and protein are upregulated in vascular endothehum by estrogen in vitro (MacRitchie AN, et al. Circ Res ( 1997), 81(3) 355-62), the effect is only partially inhibited by the e-NOS inhibitor NAME (Holm P et al. J C n Invest (1997); 100(4) 821-8)
The development of vasculoprotective drug therapies based on the protective effect of estrogen has been difficult, as it has not been possible to differentiate the desired vasculoprotective effect of estrogen from its undesirable effects on the reproductive system - e.g its utero trophic effect
Recent work on the vasculoprotective effect of estrogen m ERα-deficient mice by Iafrati and coworkers (Iafrati MD et al Nat Med (1997); 3(5) 545-8). suggests that ERα may not be responsible for the vasculoprotective estrogemc response
The inventors have unexpectedly managed to differentiate the vasculoprotective effect of estrogen from its uterotrophic effect using ligands with different binding affinity to ERα and ERβ. The inventors have discovered that the genistem, an lsoflavonic phytoestrogen, which shows approximately 20 x higher binding affinity to ERβ compared its binding affinity for ERα displays a full vasculoprotective effect but is devoid of any uterotrophic effect. In addition, the inventors have demonstrated that ERβ is strongly upregulated in the vascular wall as a consequence of injury, whereas ERα remains expressed at a constitutive background level only The discovery of a second estrogen receptor ERβ, and the recent finding that disruption of the "classical" ERα gene in mice preserves the vasculoprotective effect of estrogen, offer better targeting of estrogen in vasculoprotective drug therapies The inventors have unexpectedly demonstrated that, after endothehal denudation injury of rat carotid artery, ERα mRNA (and protein) are constitutively expressed at a low level in the vascular wall, whereas the expression of ERβ mRNA increases >30-fold after injury In in situ hybridization, the ERβ mRNA co-localizes with the replicating and migrating SMC m the media and in the neointima Treatment of ovariectomized female rats on a soybean deficient diet with the lsoflavonic phytoestrogen genistein, which shows approximately 20x higher binding affinity to ERβ than to ERα, and with 17β -estradiol, which does not differentiate between the two receptor subtypes, at a dose range of 0 00250 through 2 50 mg/kg/d, provides on both occasions a dose-dependent vasculoprotective effect However, treatment with 17β-estradιol only, but not with genistein, is accompanied by a dose-dependent uterotrophic effect Thus the vaculoprotective effect of estrogen has been for the first time differentiated from the uterotrophic effect using ligands with different binding affinity to ERβ vs ERα The in vivo experiments used genistein doses of less than 2 mg/kg/d. which is well known below the dose level (50 - 100 mM) where the protein tyrosine kinase inhibitory effect of genistein and lsoflavones have been demonstrated As genistein at this dose range is likely to function via the ERβ, and is devoid of inhibitory effect on protein tyrosine kmases, the vasculoprotective effect of genistein is therefore mediated by ERβ
The colocahzation of ERβ mRNA in in situ hybridization on the replicating/migratory smooth muscle cells dunng the rephcative and migratory bursts after endothehal trauma, is of relevance in explaining the results obtained First, on day 7 after denudation injury the endothehal regrowth from both ends of the vessel is only at its very beginning (Clowes AW, et al Lab Invest (1983); 49(3): 327-33) and the dose dependent inhibitory effect with both ligands may be discussed in terms of inhibition of smooth muscle cell replication, only Although the binding affinity of genistein to ER-β is only 20-fold better than 17β-estradιol, (the best pair of ligands at the moment to differentiate between the two receptors -Kuiper. G et al Endocrinology (1997), 138(3) 863-70), the anti-pro ferative effect and the effect on mtimal thickening vs. the uterotrophic effect were clearly different.
In regard to mtimal thickening and in vivo replication after endothehal denudation, the linear plots show a slight advantage for genistein vs. 17-β-estradιol This advantage is also seen m the in vitro vascular smooth muscle cell inhibition studies However, at the tested dose range genistein displayed no uterogenic effect in vivo, whereas the uterogenic effect of 17-β-estradιol was clearly visible Thus, it can be calculated that the vasculoprotective effect of the estrogen is most likely mediated via ERβ This is clearly supported by the observation of Iafrati et al Nat Med (1997); 3(5): 545-8 where selective disruption of the ERα gene m mice did not have an effect on the vasculoprotective effect of estrogen
Therefore, in summary, the inventors have demonstrated that ERβ is strongly upregulated in the vascular wall as a consequence of injury, whereas ERα remains expressed at constitutive low background level, only In situ hybridization demonstrated that ERβ mRNA co-localizes on the replicating and migrating vascular smooth muscle cells in the media and neointima suggesting that both genes are transcnbed and expressed in functional form This was tested by using two different ligands with approximately 20-fold affinity difference for ERα and ERβ, 17-β-estradιol and genistein These two ligands clearly differentiated the vasculoprotective vs uterogenic effect Withm the dose range tested both of the ligands demonstrated a dose-dependent vasculoprotective effect, whereas only 17-β-estradιol but not genistein demonstrated a dose-dependent uterotrophic effect Finally, the anti-prohferative effect of these two ligands on vascular smooth muscle cell cultures deriving from rat aorta was confirmed in vitro, a dose-dependent inhibition of smooth muscle cell proliferation was again observed.
Taken together, the results presented in this study and previous observations strongly suggest that the vascular protective effect of estrogen is mediated predominantly or exclusively by ERβ. These results will enable the generation of vasculoprotective estrogen mimetics without classical uterotrophic side-effects. A first aspect of the invention provides a vasculoprotective composition comprising an ERβ hgand, preferably an agonist, although the compound may be Erβ antagonist
In particular, post menopausal women suffer from an increased risk of vascular disease and heart disease and are therefore a target population for treatment with agonists of ERβ
This was unexpected because it was possible that the vasculoprotective effect operates via the recently-discovered ERβ or by another hitherto unknown ER subtype Another possibility was that the desired vasculoprotective effect is obtained via modification of the signalling to the response elements of "vasculoprotective" genes via intermediary transcription factors, such as SRC-1, TIF-2, A1B1, or via ER-interactmg proteins, such as RIP 140, RIP 160, TIF 1, etc
According to another aspect of the invention, there is provided a pharmaceutical composition useful for the treatment of vasculopathies compπsing an ERβ agonist
According to another aspect of the invention, there is provided the use of an ERβ agonist in the treament of vasculopathies
According to further aspects of the invention there are provided methods of treating, or preventing, a vasculopathy comprising treating a subject with an ERβ agonist, preferably an ERβ-selective agonist The vasculopathy may be a fibroproliferative condition For example, it may be a fibroproliferative condition such as restenosis, angioplasty, chronic allograft rejection, diabetic angiopathy, autoimmune angiopathy, atreπosclerosis, and atherosclerosis
According to another aspect of the invention there is provided a method of inducing a vasculoprotective effect in a subject, the method compπsing contacting the subject with an ERβ agonist Alternatively, the vasculoprotective effect may be induced in cells, tissues such as blood vessels or organs. The cells tissues on which may have been produced in vitro, may than be placed in a subject. For example in a vasculoprotective effect may be induced and in vitro generated or seeded vascular grafts pπor to implantation in humans. The gene may be inserted by a virus.
The vasculoprotective effect may be, for example, reducing intimal thickness.
Preferably, the ERβ agonist is selective for ERβ. For example, it may have a binding affinity for ERβ which is at least 10 times, preferably at least 20 times, greater than for ERα
According to a further aspect of the invention there is provided a method of producing artificial tissues or organs the method including the step of treating the tissue or organ with an ERβ agonist For example it is known to produce blood vessels by seeding cells, usually from the patient to receive the blood vessel about a former tube and growing those cells The invention embraces articificial tissues or organs obtainable by such a method.
Methods and compositions m accordance with the invention will now be described, by way of example only, with reference to the accompanying drawings Figs 1 to 5. in which;
Fig. 1 shows the results of in situ expression of ERα and ERβ at 15 mm and 7 days after injury Sense controls (not shown) were negative;
Fig. 2 shows the results of ERα and ERβ expression at different time points, as quantitated as number of grams/400 μm. separately for media and neointima;
Fig. 3 shows dose response plots of 17α estradiol and genistein on a 7 day denudation injury, as quantiated as number of intima nuclei, and on the uterotrophic effect, as quantitated as the 7 day weight of rat uteruses; Fig 4 shows a dose response plot of the effect of estradiol and genistein on the prohferative response of rat vascular smooth muscle cells after serum starvation and PDGF-stimulation in vitro, and
Fig 5 gives details of antibodies used in Examples
1. Expression and localization of ERα and ERβ after carotid denudation trauma in male rats
Expression and localization of ERα and ERβ was investigated by in situ hybπdisation from paraformaldehyde-fixed parrafm embedded specimens with sense controls
Carotid denudations were made to Wistar rats purchased from the Laboratory Animal Center, University of Helsinki, Finland The rats were anaesthetized with 240 mg/kg chloral hydrate I p Buprenorphine (Temgesic, Reckitt Coleman, Hull, England) was given for pen- and postoperative pain relief Male and ovaπectomized female rats weighing 300-400g were used for all experiments
A transverse incision of the neck was performed A full exposure of the carotid system was made by cleaving of the ventral edge of the left stemomastoid muscle and omohyoid muscles The proximal and distal control of the carotid artery was obtained with a l l mm micro vascular clip A 2-I,rench Fogarty balloon catheter (Baxter Healthcare Corp, Santa Ana, CA) was introduced into the common carotid artery through the left external carotid artery and inflated with 0 2 ml air To produce adequate vessel injury the catheter was passed 3 times, balloon inflated, through the common carotid artery The external carotid artery was hgated after removal of the catheter and the wound was closed
Evaluation of histological changes was made from midcarotid sections at 0, 15 mm, 3 days, 7 days, 14 days and 30 days after denudation injury The carotids were removed "en block" and fixed in paraformaldehyde
Histological specimens were fixed in 3% paraphormaldehyde solution for 4 hours, transferred to saline and embedded in paraffin The number of cell nuclei in the adventia, media and intima was quantitated from paraffin sections stained with Meyer's hematoxylm-eosin using 400x magnification
For in situ hybridization, the left carotid of male rats was denuded of endothehum and the rats were sacπfied at 15 mm, 3 days, 7 days, 14 days and 28 days after injury, with a minimum 3 of rats per time point. The specimens of different time points and the non-denuded control specimen were placed on a single organosilane-treated microscopy slide and in situ hybridization was performed as descπbed below.
2. In situ hybridization
After deparaffinization and rehydration. sections were denatured in 0 2 M HC1, heat-denatured in 2x Sahne-Socmm Citrate (SSC) at 70°C and treated with proteinase K (1 μg/ml) Sections were then post-fixed with 4% paraformaldehyde, acetylated with 0 25% acetic anhydride in 0 1 M tnethanolamine, dehydrated and air-dπed. Slides were hybπdized with antisense or sense RNA probes (described below) overnight at 60°C, washed in 4xSSC, treated with RNAse A (20 μg/ml) and washed sequentially in SSC solutions (2xSSC, lxSSC, 0.5xSSC, all at room temperature; O. lxSCC at 50°C; O. lxSCC at RT) with ImM DTT Finally, the slides were πnsed in O. lxSSC (with 1 mM DTT), dehydrated in graded ethanols and air-dried Slides were dipped into autography emulsion (NBT 3, Kodak), exposed for 7-14 days, developed, counterstained, dehydrated and mounted with Permount
3. Probe preparation
The complementary RNA probes were synthesized according to manufacturer's directions (Promega, Madison, WI) in the presence of 35S-UTP (Amersham, International. Willshire, UK) using following cDNA fragments as templates. For ERβ ; a 400 bp EcoRI-AccI fragment (from the 5'UTR region) of the rat ERβ cDNA subcloned in a pBluescπpt KS vector was linearized with EcoRI or Accl enzymes for the production of antisense and sense transcripts, respectively. For ERα, a 200 bp BstxI-EcoRI fragment (from the 3' UTR, F-domain region) of the rat ERα cDNA subcloned to a Bluescπpt vector was hneaπzed with Sad or EcoRI enzymes pπor to synthesis of antisense and sense probes, respectively. RNA probes transcπpted from opposite strands of the same plasmid template, yielding antisense and sense probes, were adjusted to the same specific radioactivity (minimum 10 000 cpm μl).
Control non-denuded carotids were compared to denuded carotids removed at 15 mm, and
3, 7, 14, and 30 days post denudation. To ensure that the expression levels between different specimens were comparable, all tissue specimens were placed on one slide and hybπdized in identical conditions either for ERα or β.
ERα and β mRNAs were expressed constitutively at low level in the vascular tunica media in normal non-denuded cartotids The level of expression of ERα mRNA remained unaltered throughout the experiment as can be seen from Figs 1 and 2
Figure 1 shows the in situ expression of ERα and ERβ m rat carotid seven days past denudation. Antisense RNA probes were used with the Lumen (L) facing up. Compared to specimens obtained 15 minutes post denudation, ERβ expression is strongly enhanced in the media (MED) and particularly in the vascular mtima (INT) whereas the level of ERα expression was not elevated
Figure 2 shows the time course of the events. Male rats were denuded as previously descπbed, and the animals were sacrificed at the same points, I e 15 minutes, 3, 7, and 14 days post injury Three-fold up-regulation of ERβ mRNA was observed three days after denudation in the media and the level of expression in the media increased to 8-fold on day 7, whereafter it declined Even more prominent changes in the expression levels were observed in the hyperplastic mtima/neomtima. Whereas the ERα expression in the intima remained at the level observed in the control vessel media, or at most doubled, the level of ERβ expression in the mtima increased nearly 40-fold on day 7 (Fig. 2), whereafter it declined but remained elevated even after 14 days post injury
4. Dose responses to 17β-E2 and genistein on post denudation carotid trauma and on uterine weight in female rats. Female adult rats were ovariectomized on day -7 and carotid denudation was performed on day 0 and the animals were killed on day 7 (at the end of the experiment also the uterus was removed, weighed and histology was performed).
Female rats were ovariectomized, placed on a soy-bean free diet (Special Diet Services, UK) (to eliminate effects of phytoestrogens from the diet) for 7 days and both carotids were denuded. One group of animals received 17β-estradiol (17β-E2) (Sigma, St Louis, MO) and the other group genistein (kindly donated by Dr. William Helferich, Michigan State University, or purchased from Plantech, UK) at reducing doses from 2.5 mg/kg/d s.c. downwards, whilst the third group received vehicle only and served as control. 17β-E2 and genistein were dissolved in dimethylsulphoxide (Sigma). Animals were weighed daily, and both drugs were administered subcutaneously (s.c.) using the following doses: 2.5, 0.25, 0.025 and 0.0025 mg/kg in one s.c. injection per day. The animals were killed at 7 days post injury, the uterus and both carotids were removed, the uterus was weighed and both organs were processed for histology as previously described.
Ten denuded carotids of rats receiving only vehicle (DMSO 200 ml/kg/day) were compared to denuded carotids of rats receiving 17-β estradiol and to carotids of rats receiving genistein at escalating doses of 0.0025, 0.025, 0.25 and 2.5 mg/kg/d, three to five carotids at each dose level.
Both 17β-E2 and genistein had a dose-dependent effect on nuclear number in mtima, but no measurable effect on the number of nuclei in the media (not shown). (Fig. 3). The line plots indicate that genistein might have been slightly more efficacious (r2 0.838 vs 0.746) in its vasculoprotective effect.
On the other hand, in the dose range employed, only 17-β estradiol displayed a dose-dependent stimulatory effect on uterine weight (r2 0.954) while genistein had no effect (r2 0.96) (Figure 3). 5. Effect on estradiol vs genistein on SMC replication in vivo
An aqueous solution of 5-bromo-2'-deoxyuridine and 5-fluoro-2'deoxyuridine (Zymed Laboratories, Inc, San Francisco,CA) was used for labelling of proliferating cells after denudation. For "pulse labelling" a dose of 400 μl of labelling suspension was injected i.v. according to manufacturer's instructions, and the rats were killed exactly 3 h after the pulse. The carotids were fixed as described above and processed for paraffin embedding. BrdU stainings of cross sections were performed using a mouse primary antibody (Bu20a, Dako, A/S, Denmark) and Vectastain Elite ABC kit (Vector Laboratories, Burlingame, CA). Sections were deparaffinized and microwave-treated at 500 W for 2 x 5 min in 0.1 M citrate buffer, pH 6, followed by treatment in 95% formamide in 0.15 M tri-sodium citrate at 70°C for 45 min. Antibody dilutions were made according to manufacturer's instructions. Sections were counterstained with Mayers' haematoxylin and eosin, and the number of positive cells was counted separately from the intimal, medial and adventitial layers.
Both of these ligands also reduced, dose dependently, the replication rate in the intima, as quantitated by the number of BrdU incorporating cells after pulse labelling of the rat.
Table 1 shows the effect of E2 and genistein on the number of proliferating (BrdU-incorporating) cells in the vascular intima seven days after denudation injury.#
No of BrdU incorporating cells
Drug dose (mg/kg/d)
17β estradiol genistein
Nil 36.7 + 7.5
0.0025 27.3 + 12.5 32.3 + 12.
0.025 6.0 + 3.8 6.0 + 3.8
2.5 10.5 + 2.9 12.3 + 3.7
# Animals received BrdU pulse 3 hours before sacrifice. 6. Dose-responses to 17β-E2 and genistein on vascular smooth muscle cell proliferation in vitro.
As the results regarding the in vivo responses of genistem vs 17β-E2 to vascular trauma suggested a marginally better efficacy of genistem, this possibility was investigated further in vitro in the proliferation assays of vascular smooth muscle cells Rat thoracic aorta smooth muscle cells at 10-12 passage were plated in 96 well tissue culture plates on day -2, left to attach phenyl-red free RPMI 1640 and depπved from serum for additional two days On day 0, the cells were stimulated by 20 ng/ml Platelet-Deπved Growth Factor-B (PDGF-B) (Sigma), or left non-stimulated Genistein and 17-β estradiol were added to the cultures at the indicated concentrations on day -1, and all cultures were harvested on day 1 after a 24 hour 3H-thymιdme (Η-TdR) pulse on day 1
As seen in Figure 4, both E2 and genistem displayed a dose-dependent anti-prohferative effect on baby rat smooth muscle cells in culture
7. Expression and localization of ERα AND ERβ in primates after carotid denudation trauma
Because species differences may exist m gene expression, we wanted to confirm the rat results descπbed above in subhuman pπmates and also to extend the observations to other types of vasculopathies, particularly to allograft fibroproliferative vascular disease in a rat cardiac transplant model and to human cardiac transplantation
Baboons are a particularly useful animal model as they have 98% sequence identity with the human genome
Carotid/iliac denudations were performed to baboons Balloon catheter denudation of carotid artenes was performed in 8 male baboons (Papio ursmus) weighing 16-18 kg Additional 4 baboons served as non-operated controls.The animals were purchased from the Animal Laboratory of the Medical Faculty, University of Stellenbosch, South Africa. The baboons were sedated with ketamine hydrochloπde (5mg/kg EM) and anesthesia was induced with thiopental sodιum(5mg/kg IV) and maintained with inhaled halothane. Prophylactic cefazohn sodium (25mg/kg IM,Elι Lilly) was administered and the left common carotid artery was explored by vertical neck incision following the anteπor border of sternocleidomastoid muscle The carotid system was exposed in the carotid tπangle and proximal and distal control of the common carotid artery was obtained with small vascular clamps, just proximal to its bifurcation No hepaπn was administered. Through a small artenotomy, a 4-Fr Fogarty balloon catheter (Baxter Healthcare Corp, Santa Ana , CA) was introduced into the distal common carotid artery It was passed retrograde into the aortic arch and inflated with 1 7 ml of air resulting m a 1.5 lbs pull force and a balloon size of 9 mm when inflated The inflated balloon was then retπeved under tension while rotating the shaft of the catheter to produce uniform injury This was repeated three times to ensure sufficient arteπal denudation All procedures were done by the same individual The catheter was then removed; the artenotomy closed with interrupted 7-0 monofilament polypropylene sutures, and flow restored The wound was then closed in layers and buprenorphine (Temgesic, Reckitt, Coleman, Hull, England) 0.25mg/kg IV was given as required, for postoperative pam relief One baboon was sacπficed at each time point by administering a overdose of pentobarbitol (100 mg kg IV) and TV potassium chloπde. Three minutes before extermination, standard hepaπn (300U/kg IV) was given Both carotid arteries were then removed and evaluation of histological changes was madefrom midcarotid sections at 0,15 mm and at 2,3,4,7,14 and 28 days post injury
Pieces of rat uterus were obtained for ER-α specificity controls
All animals received humane care in compliance with guidelines set forth by the National Institutes of Health, publication No.86-23,Guιde for the Care and Use of Laboratory Animals, and the project was approved by the Ethical Committee of the Faculty of Medicine of the University of Stellenbosch, South Africa
Immunohistochemistry was made from paraffin cross sections using a mouse or rabbit pπmary antibody and Vectastain Elite ABC kit (Vector Laboratoπes, Burhngame, CA). Sections were deparaffinized and microwave-treated at 500 W for 2 x 5 mm in 0 1 M citrate buffer, pH 6, followed by treatment in 95%> formamide in 0.15 M tπ-sodium citrate at +70°C for 45 min. Antibody dilutions were made according to manufacturers instructions. Sections were counterstained with Mayers haematoxylin and eosin, and the total number of positive cells was counted separately from the intimal, medial and adventitial layers using 400x magnification. At least 5 sections were investigated separately of each carotid and the specimen with median intensity of intimal changes was counted.
In the baboon carotid/iliac denudation model 4 baboons were sacrificed w/o injury and at least 1 baboon was sacrificed at 15 min, 2, 3, 4, 7, 14 and 28 d time points post injury. ERβ was found as exclusive receptor in baboon arteries and ERα in baboon uterus. After injury, the intensity of immunohistochemical staining with all three commercial antibodies for ERβ increased considerably and staining colocalized with the vascular SMC. There was no staining with ERα antibodies though the control uterus stained strongly.
ERβ was the only ER located in the baboon ateries found by immunohistochemistry. Specifically tissue samples were contacted with commercially-available ERβ or ERα-selective antibodies.
The staining colocalized with vascular SMC.
8. Analysis of ERα and ERβ expression in rat and human allograft vessels.
In rat and human heart allograft vessels ERβ is the exclusive receptor and ERα in uterus. During acute rejection, the ERβ was shown to be strongly upregulated (same panel of antibodies) and ERα remain non-existent.
Human specimens were obtained from routine enbdomyocardial biopsies performed for diagnostic purposes to U of Helsinki Hospital heart transplant receipients. The biopsies represented no rejection, acute rejections of different histological intensities and of chronic rejection, least 10 specimens per group. Rat heart transplants were made to abdominal vessels as descnbed (Lemstrom, K., Sihvola, R., Bruggeman C, Hayry, P., and Koskmen, P. abolished by DHPG prophylaxis m the rat. Circulation 1997:95:2614-2616).
Specimens for immunohistochemistry were obtained as descnbed above in relation to baboons.
and following antibodies shown in Fig. 5 were employed, with consistent results:
In rat and human heart allograft vessels ERβ was also found to be the exclusive receptor and ERα dominated in uterus. Duπng acute rejection, the staining for ERβ was strongly upregulated (same panel of antibodies) and the ERα reactivity remain non-existent
Taken together, the expression patterns of ERβ vs ERα in subhuman pnmates and in human and rat allograft models entirely agree with the rat carotid results descπbed in the application and demonstrate ERβ as the exclusive estrogen receptor in arteπal tissue and vascular SMC making this a ideal target for drug therapies

Claims

Claims
1. A vasculoprotective composition comprising an ER╬▓ ligand.
2. A vasculoprotective composition according to claim 1 wherein the ER╬▓ ligand is an ER╬▓ agonist.
3. A vasculoprotective composition according to claim 1 wherein the ER╬▓ ligand is an ER╬▓ antagonist
4. A vasculoprotective composition according to claim 1 or claim 2 comprising an ER╬▓-selective agonist.
5. A pharmaceutical composition useful for the treatment of vasculopathies comprising an ER╬▓ agonist.
6. A pharmaceutical composition according to claim 5 comprising an ER╬▓-selective agonist.
7. A composition according to claim 4 or 6 in which the binding affinity of the ER╬▓ agonist to ER╬▓ is at least 10 times greater than the binding affinity to ER╬▒.
8. A composition according to claim 7 in which the binding affinity of the agonist to ER╬▓ is at least 20 times greater than to ER╬▒.
9. The use of an ER╬▓ agonist in the treatment of vasculopathies.
10. The use of an ER╬▓-selective agonist in the treatment of vasculopathies.
11. The use according to claim 10 in which the vasculopathy is a fibroproliferative condition.
12. The use according to claim 11 in which the fibroproliferative vasculopathy is selected from restenosis, angioplasty, chronic allograft rejection, diabetic angiopathy, autoimmune angiopathy, arteriosclerosis, and atherosclerosis.
13. A method of inducing a vasculoprotective effect in a subject, the method comprising treating the subject with an ER╬▓ agonist.
14. A method of inducing a vasculoprotective effect according to claim 13 in which the ER╬▓ agonist has a higher affinity for ER╬▓ than ER╬▒.
15. A method of inducing a vasculoprotective effect in a subject according to claim 14 in which the binding affinity of the agonist to ER╬▓ is at least 10 times greater than to ER╬▒.
16. A method of inducing a vasculoprotective effect in a subject according to claim 15 in which the binding affinity of the agonist to ER╬▓ is at least 20 times greater than to ER╬▒.
17. A method of inducing a vasculoprotective effect in which the effect is decrease of intimal thickness.
18. A method according to any one of claims 13 to 17 in which the vasculoprotective effect is induced to treat a fibroproliferative vasculopathy.
19. A method according to claim 18 in which the fibroproliferative vasculopathy is selected from restenosis, angioplasty, chronic allograft rejection, diabetic angiopathy, autoimmune angiopathy, ateriosclerosis and atherosclerosis.
20. A composition, use or method according to any preceding claim in which the ER╬▓ selective agonist is genistein or a chemical derivative or structural analogue thereof.
21. A use or method according to any one of claims 9 to 20 in which uterotrophic effects are minimised or do not result.
22. A method according to any one of claims 13 to 21 in which the subject is a mammal.
23. A method according to claim 22 in which the mammal is a primate.
24. A method according to claim 23 in which the mammal is human.
25. A method according to claim 22, 23 or 24 in which the mammal is female.
26. A method according to claim 25 in which the female is post-menopausal.
27. A method of producing artificial tissues or organs the method including the step of treating the tissue or organ with an ER╬▓ agonist.
28. A method according to claim 27 in which the tissue or organ is a blood vessel.
29. Articificial tissues or organs obtainable by a method according to claim 27 or 28.
PCT/GB1999/002157 1998-07-06 1999-07-06 Vasculoprotector WO2000001716A2 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
EP99929579A EP1094806A2 (en) 1998-07-06 1999-07-06 Vasculoprotector
CA002331885A CA2331885A1 (en) 1998-07-06 1999-07-06 Vasculoprotector
AU46364/99A AU4636499A (en) 1998-07-06 1999-07-06 Vasculoprotector
JP2000558117A JP2002519435A (en) 1998-07-06 1999-07-06 Vascular protective agent
KR1020017000141A KR20010079495A (en) 1998-07-06 1999-07-06 Vasculoprotector

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GB9814620.2 1998-07-06
GBGB9814620.2A GB9814620D0 (en) 1998-07-06 1998-07-06 Vasculoprotector

Publications (2)

Publication Number Publication Date
WO2000001716A2 true WO2000001716A2 (en) 2000-01-13
WO2000001716A3 WO2000001716A3 (en) 2000-10-05

Family

ID=10835023

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB1999/002157 WO2000001716A2 (en) 1998-07-06 1999-07-06 Vasculoprotector

Country Status (7)

Country Link
EP (1) EP1094806A2 (en)
JP (1) JP2002519435A (en)
KR (1) KR20010079495A (en)
AU (1) AU4636499A (en)
CA (1) CA2331885A1 (en)
GB (1) GB9814620D0 (en)
WO (1) WO2000001716A2 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000016759A2 (en) * 1998-09-17 2000-03-30 Guido Schnyder Genistein for the treatment of coronary heart disease and for the prevention of restenosis after percutaneous transluminal coronary angioplasty
WO2002046168A1 (en) * 2000-12-07 2002-06-13 Astrazeneca Ab Therapeutic benzimidazole compounds
US6794403B2 (en) 2001-12-05 2004-09-21 Wyeth Substituted benzoxazoles as estrogenic agents
US6835745B2 (en) 2002-01-15 2004-12-28 Wyeth Phenyl substituted thiophenes as estrogenic agents
US7354927B2 (en) 2004-09-07 2008-04-08 Wyeth 6H-[1]benzopyrano[4,3-b]quinolines and their use as estrogenic agents
US7674783B2 (en) 2002-11-22 2010-03-09 Dimera Inc. Estrogen beta receptor agonists to prevent or reduce the severity of cardiovascular disease

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2008174463A (en) * 2007-01-16 2008-07-31 Ankhs:Kk TGF-beta SIGNAL TRANSMISSION MECHANISM INHIBITOR, SMAD DEGRADATION INDUCTOR, SMAD UBIQUITIN FORMATION PROMOTER, SMURF DEGRADATION INDUCTOR, SMURF UBIQUITIN FORMATION PROMOTER, METHOD FOR INHIBITING TGF-beta SIGNAL TRANSMISSION MECHANISM, METHOD FOR INDUCING SMAD DEGRADATION, METHOD FOR PROMOTING SMAD UBIQUITIN FORMATION, METHOD FOR INDUCING SMURF DEGRADATION, AND METHOD FOR PROMOTING SMURF UBIQUITIN FORMATION

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1993023090A1 (en) * 1992-05-20 1993-11-25 Sherwood Medical Company Artificial blood vessel
WO1997009348A2 (en) * 1995-09-08 1997-03-13 Karo Bio Ab Orphan receptor
WO1998008503A1 (en) * 1996-08-30 1998-03-05 Novogen Research Pty. Ltd. Therapeutic methods and compositions involving isoflavones
US5919813A (en) * 1998-03-13 1999-07-06 Johns Hopkins University, School Of Medicine Use of a protein tyrosine kinase pathway inhibitor in the treatment of diabetic retinopathy

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP3476604B2 (en) * 1995-08-22 2003-12-10 鐘淵化学工業株式会社 Method for manufacturing stent with drug attached / coated
JP2829388B2 (en) * 1996-09-27 1998-11-25 農林水産省四国農業試験場長 Composition for promoting cell differentiation into fat cells
JPH11139974A (en) * 1997-11-07 1999-05-25 Kureha Chem Ind Co Ltd Tissue fibrosis suppressant

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1993023090A1 (en) * 1992-05-20 1993-11-25 Sherwood Medical Company Artificial blood vessel
WO1997009348A2 (en) * 1995-09-08 1997-03-13 Karo Bio Ab Orphan receptor
WO1998008503A1 (en) * 1996-08-30 1998-03-05 Novogen Research Pty. Ltd. Therapeutic methods and compositions involving isoflavones
US5919813A (en) * 1998-03-13 1999-07-06 Johns Hopkins University, School Of Medicine Use of a protein tyrosine kinase pathway inhibitor in the treatment of diabetic retinopathy
US5919813C1 (en) * 1998-03-13 2002-01-29 Univ Johns Hopkins Med Use of a protein tyrosine kinase pathway inhibitor in the treatment of diabetic retinopathy

Non-Patent Citations (15)

* Cited by examiner, † Cited by third party
Title
BALARAM S.K. ET AL: "Cell adhesion molecules and insulin-like growth factor-1 in vascular disease." JOURNAL OF VASCULAR SURGERY, (1997) 25/5 (866-876)., XP002126538 *
BECKELER, KLAUS ET AL: "Differential effects of immunosuppressive cyclosporine A and inhibitors of protein kinases on cytokine induced cell proliferation of umbilical cord artery smooth muscle cells: The role of endothelial supernatants on TNF-alpha effects." EUROPEAN JOURNAL OF CELL BIOLOGY, (1996) VOL. 69, NO. SUPPL. 42, PP. 126. MEETING INFO.: 21ST ANNUAL MEETING OF THE GERMAN SOCIETY FOR CELL BIOLOGY HAMBURG, GERMANY MARCH 24-28, 1996, XP002126534 *
CLARKSON T.B. ET AL: "The nonhuman primate model of the relationship between gonadal steroids and coronary heart disease." PROGRESS IN CARDIOVASCULAR DISEASES, (1995) 38/3 (189-198)., XP002126540 *
DATABASE WPI Section Ch, Week 199720 Derwent Publications Ltd., London, GB; Class A96, AN 1997-215838 XP002126543 & JP 09 056807 A (KANEBUCHI KAGAKU KOGYO KK), 4 March 1997 (1997-03-04) *
DATABASE WPI Section Ch, Week 199826 Derwent Publications Ltd., London, GB; Class B02, AN 1998-292041 XP002126542 & JP 10 101561 A (SHIKOKU NOGYO SHIKENJO), 21 April 1998 (1998-04-21) *
DATABASE WPI Section Ch, Week 199932 Derwent Publications Ltd., London, GB; Class B02, AN 1999-374524 XP002126544 & JP 11 139974 A (KUREHA CHEM IND CO LTD), 25 May 1999 (1999-05-25) *
HONORE E K ET AL: "Soy isoflavones enhance coronary vascular reactivity in atherosclerotic female macaques." FERTILITY AND STERILITY, (1997 JAN) 67 (1) 148-54., XP002126539 *
KAPIOTIS S ET AL: "Genistein, the dietary-derived angiogenesis inhibitor, prevents LDL oxidation and protects endothelial cells from damage by atherogenic LDL." ARTERIOSCLEROSIS, THROMBOSIS, AND VASCULAR BIOLOGY, (1997 NOV) 17 (11) 2868-74., XP002126537 *
KUIPER ET AL.: "Comparison of the ligand Binding Specificity and Transcript Distribution of Estrogen Receptors alpha and beta" ENDOCRINOLOGY, vol. 138, no. 3, 1997, pages 863-870, XP002126536 cited in the application *
LINDNER, VOLKHARD ET AL: "Increased expression of estrogen receptor -. beta. mRNA in male blood vessels after vascular injury" CIRC. RES. (1998), 83(2), 224-229, XP000856530 *
M S ANTHONY ET AL: "Soybean isoflavones improve cardiovascular risk factors without affecting the reproductive system of peripubertal rhesus monkeys" JOURNAL OF NUTRITION,XX,XX, vol. 126, no. 1, page 43-50 XP002095625 ISSN: 0022-3166 *
ST CLAIR R.W.: "Estrogens and atherosclerosis: Phytoestrogens and selective estrogen receptor modulators." CURRENT OPINION IN LIPIDOLOGY, (1998) 9/5 (457-463)., XP002126541 *
TAKAMURA, HIROYUKI: "Immunosuppressive effects of a tyrosine kinase specific inhibitors ( genistein ) on the rejection of rat allogeneic small bowel transplantation" KANAZAWA DAIGAKU JUZEN IGAKKAI ZASSHI (1995), 104(6), 719-731, XP002126533 *
TROVATI M. ET AL: "Insulin increases cyclic nucleotide content in human vascular smooth muscle cells: A mechanism potentially involved in insulin-induced modulation of vascular tone." DIABETOLOGIA, (1995) 38/8 (936-941)., XP002126535 *
WILLIAMS J.K. ET AL: "Estrogens, progestins, and coronary artery reactivity in atherosclerotic monkeys." JOURNAL OF STEROID BIOCHEMISTRY AND MOLECULAR BIOLOGY, (1998) 65/1-6 (219-224)., XP002126532 *

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000016759A2 (en) * 1998-09-17 2000-03-30 Guido Schnyder Genistein for the treatment of coronary heart disease and for the prevention of restenosis after percutaneous transluminal coronary angioplasty
WO2000016759A3 (en) * 1998-09-17 2000-05-25 Guido Schnyder Genistein for the treatment of coronary heart disease and for the prevention of restenosis after percutaneous transluminal coronary angioplasty
WO2002046168A1 (en) * 2000-12-07 2002-06-13 Astrazeneca Ab Therapeutic benzimidazole compounds
US6794403B2 (en) 2001-12-05 2004-09-21 Wyeth Substituted benzoxazoles as estrogenic agents
US7129258B2 (en) 2001-12-05 2006-10-31 Wyeth Substituted benzoxazoles as estrogenic agents
US7148247B2 (en) 2001-12-05 2006-12-12 Wyeth Substituted benzoxazoles as estrogenic agents
US7531564B2 (en) 2001-12-05 2009-05-12 Wyeth Substituted benzoxazoles as estrogenic agents
US6835745B2 (en) 2002-01-15 2004-12-28 Wyeth Phenyl substituted thiophenes as estrogenic agents
US7674783B2 (en) 2002-11-22 2010-03-09 Dimera Inc. Estrogen beta receptor agonists to prevent or reduce the severity of cardiovascular disease
US7354927B2 (en) 2004-09-07 2008-04-08 Wyeth 6H-[1]benzopyrano[4,3-b]quinolines and their use as estrogenic agents

Also Published As

Publication number Publication date
AU4636499A (en) 2000-01-24
CA2331885A1 (en) 2000-01-13
EP1094806A2 (en) 2001-05-02
GB9814620D0 (en) 1998-09-02
JP2002519435A (en) 2002-07-02
KR20010079495A (en) 2001-08-22
WO2000001716A3 (en) 2000-10-05

Similar Documents

Publication Publication Date Title
Fu et al. HIF-1α-BNIP3-mediated mitophagy in tubular cells protects against renal ischemia/reperfusion injury
Barha et al. Progesterone treatment normalizes the levels of cell proliferation and cell death in the dentate gyrus of the hippocampus after traumatic brain injury
McEwen et al. The brain as a target for steroid hormone action
Fan et al. 17β-estradiol on the expression of G-protein coupled estrogen receptor (GPER/GPR30) mitophagy, and the PI3K/Akt signaling pathway in ATDC5 chondrocytes in vitro
Mathews et al. Paradoxical hypermasculinization of the zebra finch song system by an antiestrogen
Wolf et al. Characterization of tamoxifen stimulated MCF-7 tumor variants grown in athymic mice
Benoit et al. Estetrol, a fetal selective estrogen receptor modulator, acts on the vagina of mice through nuclear estrogen receptor α activation
JP2000510460A (en) Treatment of epithelial malignancies using squalamine in combination with other anticancer drugs
JP2001506638A (en) How to prevent or control cataracts
Holder et al. Methamphetamine-enhanced female sexual motivation is dependent on dopamine and progesterone signaling in the medial amygdala
DiMusto et al. Increased JNK in males compared with females in a rodent model of abdominal aortic aneurysm
Storment et al. Estrogen augments the vasodilatory effects of vascular endothelial growth factor in the uterine circulation of the rat
JP2003509456A (en) Local delivery of 17-β estradiol to prevent intimal thickening and improve vascular endothelial function after vascular injury
Turi et al. Estrogen downregulates the number of caveolae and the level of caveolin in uterine smooth muscle
Takaoka et al. Oestrogen protects against ischaemic acute renal failure in rats by suppressing renal endothelin-1 overproduction
EP1094806A2 (en) Vasculoprotector
Xu et al. Pharmacology-based molecular docking of 4-methylcatechol and its role in RANKL-mediated ROS/Keap1/Nrf2 signalling axis and osteoclastogenesis
Gérard et al. Estetrol: from preclinical to Clinical Pharmacology and advances in the understanding of the molecular mechanism of action
Rudzitis-Auth et al. Inhibition of cyclooxygenase-2 suppresses the recruitment of endothelial progenitor cells in the microvasculature of endometriotic lesions
TW200845989A (en) Medicament for the treatment of endometriosis
Chen et al. Basic fibroblast growth factor protects against liver ischemia-reperfusion injury via the Nrf2/Hippo signaling pathway
CN107007611B (en) Application of nomegestrol acetate in preparation of medicine for treating endometrial cancer
Love et al. Tamoxifen therapy in breast cancer control worldwide.
de Azevedo Camin et al. Effects of maternal exposure to the galactagogue Sulpiride on reproductive parameters in female rats
AU2004200635A1 (en) Vasculoprotector

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AU CA JP KR NZ US

AL Designated countries for regional patents

Kind code of ref document: A2

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

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)
AK Designated states

Kind code of ref document: A3

Designated state(s): AU CA JP KR NZ US

AL Designated countries for regional patents

Kind code of ref document: A3

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

WWE Wipo information: entry into national phase

Ref document number: 1999929579

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 46364/99

Country of ref document: AU

ENP Entry into the national phase

Ref document number: 2331885

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 1020017000141

Country of ref document: KR

ENP Entry into the national phase

Ref country code: JP

Ref document number: 2000 558117

Kind code of ref document: A

Format of ref document f/p: F

WWP Wipo information: published in national office

Ref document number: 1999929579

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 09719658

Country of ref document: US

WWP Wipo information: published in national office

Ref document number: 1020017000141

Country of ref document: KR

WWW Wipo information: withdrawn in national office

Ref document number: 1020017000141

Country of ref document: KR

WWW Wipo information: withdrawn in national office

Ref document number: 1999929579

Country of ref document: EP