Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 135
Filtrar
1.
Front Endocrinol (Lausanne) ; 14: 1201547, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37766684

RESUMO

Introduction: The pharmacological target for progesterone, different progestins, and Selective Progesterone Receptor Modulators (SPRMs) is the nuclear progesterone receptor (PR). EC313 is a new member of a subgroup of SPRMs, mesoprogestins, which combine especially PR- agonistic and PR-antagonistic activities in one molecule. Methods: The suitable in vivo-model for the differentiation of SPRMs from the subgroup of mesoprogestins is the estrogen-primed juvenile rabbit endometrium assay (McPhail Assay). Remarkably, in contrast to other well-known SPRMs with no agonistic effects in this test, EC313 shows clear partial PR-agonistic effects that are higher than that of the well-known mesoprogestin Asoprisnil which already demonstrated remarkable clinical effectiveness for the treatment of uterine fibroids and endometriosis. The findings from the guinea pig studies presented here can be the impetus for further preclinical development of EC313. This model shows the same features for the termination of pregnancy by antiprogestins such as Mifepristone and Ulipristal acetate (UPA) in humans. Moreover, it is possible to distinguish between progestational and anti-progestational activities in the same experiment. Results: The EC313 treatment reveals PR dominance in the genital tract and inhibits unopposed estrogenic effects. In very high doses (30.0 mg/animal/day subcutaneously (s.c.)) given twice on pregnancy days 43 and 44, no premature labor was induced (in contrast to UPA, dosed at 10.0 and 30. mg/animal/day s.c.). The anti-ovulatory activity of EC313 exceeds that of Ulipristal acetate or Mifepristone. EC313 binds to the steroid receptors in vitro with a similar affinity as the natural ligand progesterone. At the glucocorticoid receptor (GR) EC313 acts as a weak inhibitor. Minor activities at the human androgen receptor (AR) and mineralocorticoid receptor (MR) are considered negligible. No binding to the estradiol receptor was detected. In contrast to some in vitro-receptor findings, estrogenic, anti-estrogenic, androgenic, anti-androgenic, glucocorticoid, and anti-glucocorticoid actions were absent in vivo. The tissue selectivity of EC313 was demonstrated previously by reducing the growth and proliferation of uterine fibroids in animal models (lowest effective dosage 0.1 mg/kg/day s.c.).. As shown in this article, the anti-fibroid activity of EC313 was confirmed with a 10 times lower dosage (0.01 mg/kg/day s.c.). It was also shown that EC313 reduces the growth of endometriotic lesions in a human xenograft immune-deficient (NOD-SCID) mice model with a comparatively very low dosage range. In the aforementioned EC313 activity model, UPA was tested as the reference compound, the clinical effectiveness of which has already been demonstrated. Discussion: For an explanation of these findings, the possibility is discussed that the mixed agonistic/antagonistic feature of EC313 is tissue target-specific based on its super-additive synergism characteristic for active bifunctional agents. In conclusion, the specific pharmacodynamic profile of this compound opens the possibility for the development of a drug with a distinct pharmaco-endocrinological profile against uterine fibroids, endometriosis, and other PR-dependent gynecological diseases.


Assuntos
Endometriose , Receptores de Progesterona , Camundongos , Feminino , Gravidez , Humanos , Animais , Cobaias , Coelhos , Camundongos Endogâmicos NOD , Camundongos SCID , Progesterona , Mifepristona/farmacologia , Progestinas , Estrogênios
2.
Eur J Paediatr Dent ; 20(4): 274-279, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31850768

RESUMO

AIM: The relationship between mouth breathing and dental caries, gingival inflammation, and halitosis in children is contentious with studies reporting positive and negative associations; this study aimed at investigating the effect of mouth breathing on dental, gingival health status, and halitosis. MATERIALS AND METHODS: An observational cross-sectional study was carried out involving 785 randomly selected children and adolescents between the ages of 10 and 15 in the city of Leipzig, Germany (LIFE Child cohort). Caries levels and gingival health status for the upper-right and the lower-left central incisors were assessed by evaluating ICDAS scores and CPI scores, respectively. A standardised questionnaire was used to assess self-reported mouth-breathing habit and halitosis. RESULTS: This study showed a statistically significant association between halitosis and mouth breathing (OR=3.0; 95% CI: 1.5-6.2), and a significant increase in mouth breathing habit in males compared to females (59.7% vs. 40.3%; p<0.05). There were no statistically significant differences in ICDAS scores, orthodontic treatment, CPI scores, or socioeconomic status between the mouth and nasal-breathing groups. CONCLUSION: Mouth breathing habit has no effect on the prevalence of caries or gingivitis based on examining the upper-right central incisor (11) and the lower-left central incisor. However, mouth breathers showed a significant increase in halitosis compared to nasal-breathing individuals.


Assuntos
Cárie Dentária , Halitose , Adolescente , Criança , Estudos Transversais , Feminino , Humanos , Masculino , Respiração Bucal , Saúde Bucal
3.
Hum Reprod Open ; 2019(4): hoz027, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31777761

RESUMO

STUDY QUESTION: What is the safety and efficacy profile during long-term (12-24 months) uninterrupted treatment with the selective progesterone receptor modulator asoprisnil, 10 and 25 mg in women with heavy menstrual bleeding (HMB) associated with uterine fibroids? SUMMARY ANSWER: Uninterrupted treatment with asoprisnil should be avoided due to endometrial safety concerns and unknown potential long-term consequences. WHAT IS KNOWN ALREADY: Asoprisnil was well tolerated in shorter-term studies and effectively suppressed HMB and reduced fibroid volume. STUDY DESIGN SIZE DURATION: Women with uterine fibroids who had previously received placebo (n = 87) or asoprisnil 10 mg (n = 221) or 25 mg (n = 215) for 12 months in two double-blind studies entered this randomized uncontrolled extension study and received up to 12 additional months of treatment followed by 6 months of post-treatment follow-up. Women who previously received placebo were re-randomized to either asoprisnil 10 or 25 mg for the extension study. This report focuses on the 436 women who received asoprisnil in the double-blind studies and this extension study. Results for women who previously received placebo in the double-blind studies are not described. PARTICIPANTS/MATERIALS SETTING METHODS: Women ≥18 years of age who completed a 12-month, double-blind, placebo-controlled study, had estradiol levels indicating that they were not menopausal and had no endometrial hyperplasia or other significant endometrial pathology were eligible. The safety endpoints were focused on endometrial assessments. The composite primary efficacy endpoint was the proportion of women who demonstrated a response to treatment by meeting all three of the following criteria at the final month for participants who prematurely discontinued or at month 12 for those who completed the study: a reduction from initial baseline to final visit of ≥50% in the menstrual pictogram score, hemoglobin concentration ≥11 g/dl or an increase of ≥1 g/dl from initial baseline at the final visit, and no surgical or invasive intervention for uterine fibroids. Other efficacy endpoints included rates for amenorrhea and suppression of bleeding, changes in fibroid and uterine volume and changes in hematologic parameters. No statistical tests were planned or performed for this uncontrolled study. MAIN RESULTS AND ROLE OF CHANCE: Imaging studies revealed a progressive increase in endometrial thickness and cystic changes that frequently prompted invasive diagnostic procedures. Endometrial biopsy results were consistent with antiproliferative effects of asoprisnil. Two cases of endometrial cancer were diagnosed. At the final month of this extension study (total duration of uninterrupted treatment up to 24 months), the primary efficacy endpoint was achieved in 86 and 92% of women in the asoprisnil 10- and 25-mg groups, respectively. During each month of treatment, amenorrhea was observed in the majority of women (up to 77 and 94% at 10 and 25 mg, respectively). There was a progressive, dose-dependent decrease in the volume of the primary fibroid with asoprisnil 10 and 25 mg (-55.7 and -75.2% median decrease, respectively, from baseline [i.e. the beginning of the placebo-controlled study] to month 12 [cumulative months 12-24] of this extension study). These effects were associated with improvements in quality of life measures. LIMITATIONS REASONS FOR CAUTION: This study was uncontrolled, which limits the interpretation of safety and efficacy findings. The study also had multiple protocol amendments with the addition of diagnostic procedures and, because no active comparator was included, the potential place of asoprisnil in comparison to therapies such as GnRH agonists and surgery cannot be determined. WIDER IMPLICATIONS OF THE FINDINGS: Long-term, uninterrupted treatment with asoprisnil leads to prominent cystic endometrial changes that are consistent with the 'late progesterone receptor modulator' effects, which prompted invasive diagnostic procedures, although treatment efficacy is maintained. Although endometrial cancers were uncommon during both treatment and follow-up, these findings raise concerns regarding endometrial safety during uninterrupted long-term treatment with asoprisnil. This study shows that uninterrupted treatment with asoprisnil should be avoided due to safety concerns and unknown potential long-term consequences. STUDY FUNDING/COMPETING INTERESTS: AbbVie Inc. (prior sponsor, TAP Pharmaceutical Products Inc.) sponsored the study and contributed to the design and conduct of the study, data management, data analysis, interpretation of the data and the preparation and approval of the manuscript. Financial support for medical writing and editorial assistance was provided by AbbVie Inc. M. P. Diamond received research funding for the conduct of the study paid to the institution and is a consultant to AbbVie. He is a stockholder and board and director member of Advanced Reproductive Care. He has also received funding for study conduct paid to the institution for Bayer and ObsEva. E. A. Stewart participated as a site investigator in the phase 2 study of asoprisnil and served as a consultant to TAP Pharmaceuticals during the time of design and conduct of the studies while on the faculty of Harvard Medical School and Brigham and Women's Hospital, Boston, MA. In the last 3 years, she has received support from National Institutes of Health grants HD063312, HS023418 and HD074711. She has served as a consultant for AbbVie Inc., Allergan, Bayer HealthCare AG and Myovant for consulting related to uterine leiomyoma and to Welltwigs for consulting related to infertility. She has received royalties from UpToDate and the Med Learning Group. A.R.W. Williams has acted as a consultant for TAP Pharmaceutical Products Inc. and Repros Therapeutics Inc. He has current consultancies with PregLem SA, Gedeon Richter, HRA Pharma and Bayer. B.R. Carr has served as consultant and received research funding from AbbVie Inc. and Synteract (Medicines360). E.R. Myers has served as consultant for AbbVie Inc., Allergan and Bayer. R.A. Feldman received compensation for serving as a principal investigator and participating in the conduct of the trial. W. Elger was a co-inventor of several patents related to asoprisnil.C. Mattia-Goldberg is a former employee of AbbVie Inc. and owns AbbVie stock or stock options. B.M. Schwefel and K. Chwalisz are employees of AbbVie Inc. and own AbbVie stock or stock options. TRIAL REGISTRATION NUMBER: NCT00156195 at clinicaltrials.gov.

4.
Hum Reprod ; 34(4): 623-634, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30865281

RESUMO

STUDY QUESTION: Can asoprisnil, a selective progesterone receptor modulator, provide clinically meaningful improvements in heavy menstrual bleeding (HMB) associated with uterine fibroids with an acceptable safety profile? SUMMARY ANSWER: Uninterrupted treatment with asoprisnil for 12 months effectively controlled HMB and reduced fibroid and uterine volume with few adverse events. WHAT IS KNOWN ALREADY: In a 3-month study, asoprisnil (5, 10 and 25 mg) suppressed uterine bleeding, reduced fibroid and uterine volume, and improved hematological parameters in a dose-dependent manner. STUDY DESIGN, SIZE, DURATION: In two Phase 3, double-blind, randomized, placebo-controlled, multicentre studies, women received oral asoprisnil 10 mg, asoprisnil 25 mg or placebo (2:2:1) once daily for up to 12 months. PARTICIPANTS/MATERIALS, SETTING, METHODS: Premenopausal women ≥18 years of age in North America with HMB associated with uterine fibroids were included (N = 907). The primary efficacy endpoint was the percentage of women who met all three predefined criteria at 12 months or the final month for patients who prematurely discontinued: (1) ≥50% reduction in monthly blood loss (MBL) by menstrual pictogram, (2) hemoglobin concentration ≥11 g/dL or an increase of ≥1 g/dL, and (3) no interventional therapy for uterine fibroids. Secondary efficacy endpoints included changes in other menstrual bleeding parameters, volume of the largest fibroids, uterine volume and health-related quality of life (HRQL). MAIN RESULTS AND THE ROLE OF CHANCE: In all, 90% and 93% of women in the asoprisnil 10-mg and 25-mg groups, respectively, and 35% of women in the placebo group met the primary endpoint (P < 0.001). Similar results were observed at month 6 (P < 0.001). The percentage of women who achieved amenorrhea in any specified month ranged from 66-78% in the asoprisnil 10-mg group and 83-93% in the asoprisnil 25-mg group, significantly higher than with placebo (3-12%, P < 0.001). Hemoglobin increased rapidly (by month 2) with asoprisnil treatment and was significantly higher versus placebo throughout treatment. The primary fibroid and uterine volumes were significantly reduced from baseline through month 12 with asoprisnil 10 mg (median changes up to -48% and -28%, respectively) and 25 mg (median changes up to -63% and -39%, respectively) versus placebo (median changes up to +16% and +13%, respectively; all P < 0.001). Dose-dependent, significant improvements in HRQL (Uterine Fibroid Symptom and Quality of Life instrument) were observed with asoprisnil treatment. Asoprisnil was generally well tolerated. Endometrial biopsies indicated dose- and time-dependent decreases in proliferative patterns and increases in quiescent or minimally stimulated endometrium at month 12 of treatment. Although not statistically significantly different at month 6, mean endometrial thickness at month 12 increased by ~2 mm in both asoprisnil groups compared with placebo (P < 0.01). This effect was associated with cystic changes in the endometrium on MRI and ultrasonography, which led to invasive diagnostic and therapeutic procedures in some asoprisnil-treated women. LIMITATIONS, REASONS FOR CAUTION: Most study participants were black; few Asian and Hispanic women participated. The study duration may have been insufficient to fully characterize the endometrial effects. WIDER IMPLICATIONS OF THE FINDINGS: Daily uninterrupted treatment with asoprisnil was highly effective in controlling menstrual bleeding, improving anemia, reducing fibroid and uterine volume, and increasing HRQL in women with HMB associated with uterine fibroids. However, this treatment led to an increase in endometrial thickness and invasive diagnostic and therapeutic procedures, with potential unknown consequences. STUDY FUNDING/COMPETING INTEREST(S): This trial was funded by AbbVie Inc. (prior sponsors: TAP Pharmaceutical Products Inc., Abbott Laboratories). E.A. Stewart was a site investigator in the Phase 2 study of asoprisnil and consulted for TAP during the design and conduct of these studies while at Harvard Medical School and Brigham and Women's Hospital. She received support from National Institutes of Health grants HD063312, HS023418 and HD074711 and research funding, paid to Mayo Clinic for patient care costs related to an NIH-funded trial from InSightec Ltd. She consulted for AbbVie, Allergan, Bayer HealthCare AG, Gynesonics, and Welltwigs. She received royalties from UpToDate and the Med Learning Group. M.P. Diamond received research funding for the conduct of the studies paid to the institution and consulted for AbbVie. He is a stockholder and board and director member of Advanced Reproductive Care. He has also received funding for study conduct paid to the institution from Bayer and ObsEva. A.R.W. Williams consulted for TAP and Repros Therapeutics Inc. He has current consultancies with PregLem SA, Gedeon Richter, HRA Pharma and Bayer. B.R. Carr consulted for and received research funding from AbbVie. E.R. Myers consulted for AbbVie, Allergan and Bayer. R.A. Feldman received compensation for serving as a principal investigator and participating in the conduct of the trial. W. Elger was co-inventor of several patents related to asoprisnil. C. Mattia-Goldberg is a former employee of AbbVie and may own AbbVie stock or stock options. B.M. Schwefel and K. Chwalisz are employees of AbbVie and may own AbbVie stock or stock options. TRIAL REGISTRATION NUMBER: NCT00152269, NCT00160381 (clinicaltrials.gov). TRIAL REGISTRATION DATE: 7 September 2005; 8 September 2005. DATE OF FIRST PATIENT'S ENROLMENT: 12 September 2002; 6 September 2002.


Assuntos
Estrenos/efeitos adversos , Estrenos/uso terapêutico , Leiomioma/tratamento farmacológico , Menorragia/tratamento farmacológico , Oximas/efeitos adversos , Oximas/uso terapêutico , Receptores de Progesterona/efeitos dos fármacos , Neoplasias Uterinas/tratamento farmacológico , Administração Oral , Adulto , Método Duplo-Cego , Endométrio/efeitos dos fármacos , Estrenos/administração & dosagem , Feminino , Seguimentos , Humanos , Leiomioma/complicações , Menorragia/complicações , Pessoa de Meia-Idade , Oximas/administração & dosagem , Medidas de Resultados Relatados pelo Paciente , Pré-Menopausa , Qualidade de Vida , Resultado do Tratamento , Carga Tumoral/efeitos dos fármacos , Neoplasias Uterinas/complicações
5.
J Steroid Biochem Mol Biol ; 183: 152-158, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29902513

RESUMO

A guinea pig model for new HEC methods is proposed. Two targets for HEC (Hormonal Emergency Contraception), ovulation and conception (post-mating study), were investigated using adjusted PRM treatments: (a) Ovulation inhibition study: Injections on cycle days 10-17, study of ovarian histology on day 18; (b) post-mating study: Injections on cycle days 1 and 2; rate of pregnant females was recorded at autopsy on day 18. P plasma levels permitted assessment of effects on ovulation in non-conceiving animals. RESULTS: (a) All controls had recently ovulated. Statistically significant anti-ovulatory effects (p < 0.05, Fisher's Exact Test) were seen at 10 mg UPA (ulipristal acetate, CDB2914) and ≥0.3 mg EC317; 100% inhibition was found for EC317 at 10, 3, and 1 mg/day. No dosage of UPA was 100% effective. (b) In post-mating studies, 16 of 30 controls were pregnant. Both PRMs (progesterone receptor modulator) exerted inhibitory effects on conception, none on imminent ovulation; 1 of 10 animals had living conceptuses after 10 mg UPA, none following 10 and 1 mg EC317/day, respectively. At pairwise comparison with controls, 10 mg was the lowest effective dosage for UPA (p < 0.05), and 1 mg for EC317 (p < 0.01). P plasma levels: Significantly lower P (p < 0.05) in subsequently pregnant vs non-pregnant controls was found on cycle day 3 or 4; this difference disappeared on day 8 or 9. This stage thus appears vulnerable to hormonal constellations and possibly PRM effects. HEC model: Effects on ovulation and conception were seen at the same dose levels of both PRM. Superior and more consistent effects of EC317 vs UPA (factor ≥10) suggest higher efficacy using EC317 for HEC.


Assuntos
Anticoncepção Pós-Coito/métodos , Anticoncepcionais Femininos/farmacologia , Modelos Animais , Norpregnadienos/farmacologia , Ovulação/efeitos dos fármacos , Animais , Feminino , Cobaias , Masculino , Gravidez , Receptores de Progesterona/metabolismo
6.
J Steroid Biochem Mol Biol ; 165(Pt B): 305-311, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27449818

RESUMO

Oral compared to parenteral estrogen administration is characterized by reduced systemic but prominent hepatic estrogenic effects on lipids, hemostatic factors, GH-/IGF I axis, angiotensinogen. In order to avoid such adverse metabolic effects of oral treatment, estradiol (E2) prodrugs (EP) were designed which bypass the liver tissue as inactive molecules. Carbone17-OH sulfonamide [-O2-NH2] substituted esters of E2 (EC508, others) were synthesized and tested for carbonic anhydrase II (CA-II) binding. CA II in erythrocytes is thought to oppose extraction of EP from portal vein blood during liver passage. Ovariectomized (OVX, day minus 14) rats were orally treated once daily from day 1-3. Sacrifice day 4. Uteri were dissected and weighed. Cholesterol fractions and angiotensinogen were determined in plasma. Oral E2 and ethinyl estradiol (EE) generated dose related uterine growth and important hepatic estrogenic effects. EP induced uterine growth at about hundred-fold lower doses. This was possible with almost absent effects on plasma cholesterol or angiotensinogen. Preliminary pharmacokinetic studies with EC508 used intravenous and oral administration in male rats. Resulting blood levels revealed complete oral bioavailability. Further high blood- but low plasma concentrations indicated erythrocyte binding of EC508 in vivo as potential mechanism of low extraction at liver passage. Very high systemic estrogenicity combined with markedly lower or absent adverse hepatic estrogenic effects is evidence for a systemic release of E2 from sulfonamide EP. In conclusion, tested oral EP bypass the liver in erythrocytes furnishing systemic estradiol at hydrolysis. This mechanism avoids the hepatic estrogenic impact of conventional oral estrogen therapy.


Assuntos
Estradiol/farmacologia , Estrogênios/administração & dosagem , Fígado/metabolismo , Pró-Fármacos/farmacologia , Administração Oral , Angiotensinogênio/sangue , Animais , Disponibilidade Biológica , Anidrase Carbônica II/metabolismo , Colesterol/sangue , Eritrócitos/citologia , Eritrócitos/metabolismo , Ésteres/química , Feminino , Humanos , Hidrólise , Fígado/efeitos dos fármacos , Masculino , Ovariectomia , Ratos , Ratos Sprague-Dawley , Especificidade da Espécie , Sulfonamidas/química , Tromboembolia , Útero/efeitos dos fármacos
9.
J Steroid Biochem Mol Biol ; 84(1): 51-6, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12648524

RESUMO

The present study investigated the influence of the endogenous estradiol metabolite 2-methoxyestradiol (2ME) on the growth of methyl-nitroso-urea (MNU)-induced mammary carcinoma in the rat. 2ME was administered by means of subcutaneously implanted osmotic pumps for a period of 4 weeks. The dosages of 2ME were 1 and 5mg/kg per day, the control animals received saline. At the low dosage of 2ME a stimulation of tumor growth was observed, whereas at the high dosage an inhibition was found. The urinary excretion of 15 estradiol metabolites revealed that 2ME triggered strong changes in estrogen metabolism in the organism. Our data show that 2ME may elicit both stimulation and inhibition of tumor growth depending on the dosage used, a fact which should be considered in case of therapeutic use.


Assuntos
Carcinógenos , Estradiol/farmacologia , Neoplasias Mamárias Animais/induzido quimicamente , Metilnitrosoureia , Neoplasias Experimentais/tratamento farmacológico , 2-Metoxiestradiol , Animais , Estradiol/análogos & derivados , Feminino , Neoplasias Mamárias Animais/tratamento farmacológico , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
10.
Toxicol Lett ; 128(1-3): 129-44, 2002 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-11869824

RESUMO

Interactions of 27 steroids, among them 17 derivatives such as ethers, sulfates and amidosulfonates derived from 17 beta- and 17 alpha-estradiol, from testosterone and alpha- and beta-dihydrotesosterone and from dehydroepiandrosterone with rat liver microsomal cytochromes P450 (P450) were investigated in vitro by assessing binding to P450 and effects on P450 mediated monooxygenase functions as measured by different model reactions: ethoxyresorufin O-deethylation (EROD), ethoxycoumarin O-deethylation (ECOD) and ethylmorphine N-demethylation (EMND). With the exception of 17 alpha-estradiol-3-dimethylamidosulfonate, estrone, its -3-methylether and -3-amidosulfonate and testosterone, all other steroids displayed type I or reverse type I binding to P450. All steroids inhibited EROD activity in micromolar concentrations. An additional strong inhibition of ECOD and EMND activities was only demonstrated for the androgens and progestins. Estriol, estrone and mestranol displayed less inhibitory actions on the model reactions than estradiol. No major differences in comparison to the parent compounds were noted with the other derivatives. The only exceptions were 17 beta-(8,9-dehydro-14 alpha,15 alpha-methylene)estradiol, which displayed stronger effects than estradiol, and dehydroepiandrosterone-3-sulfate, which was less effective than dehydroepiandrosterone. Possible antioxidant properties of the steroids were examined by the stimulated lipid peroxidation (LPO), H2O2 production, and lucigenin (LC) and luminol (LM) amplified chemiluminescence (CL) using rat liver microsomes. Additionally, the influence on rat whole blood chemiluminescence (WB-CL) was assessed. All the estrogens, but not their methylethers and amidosulfonates inhibited LPO in micromolar concentrations. The effects on the other oxidase model reactions or on WB-CL were less distinct. Only ethinylestradiol and 17 beta-(8,9-dehydro-14 alpha,15 alpha-methylene)estradiol displayed a strong inhibitory action on all model reactions. With the exception of dehydroepiandrosterone-3-sulfate, which in general had only weak effects, the androgen and progestin derivatives, in contrast, strongly decreased H2O2 formation and LM- and LC-CL, but were mostly ineffective on LPO and WB-CL.


Assuntos
Androstenodiona/análogos & derivados , Sistema Enzimático do Citocromo P-450/metabolismo , Desidroepiandrosterona/análogos & derivados , Estradiol/análogos & derivados , Microssomos Hepáticos/efeitos dos fármacos , O-Dealquilase 7-Alcoxicumarina/metabolismo , Androstenodiona/metabolismo , Androstenodiona/farmacologia , Animais , Citocromo P-450 CYP1A1/metabolismo , Desidroepiandrosterona/metabolismo , Desidroepiandrosterona/farmacologia , Estradiol/metabolismo , Estradiol/farmacologia , Etilmorfina-N-Demetilasa/metabolismo , Fígado/metabolismo , Medições Luminescentes , Masculino , Microssomos Hepáticos/enzimologia , Microssomos Hepáticos/metabolismo , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Testosterona/análogos & derivados , Testosterona/metabolismo , Testosterona/farmacologia
11.
Theriogenology ; 56(4): 601-11, 2001 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-11572441

RESUMO

Effective and reversible control of reproduction in bears is highly desirable for conservation and management programs in zoos to establish genetically variable ex-situ populations of bears within the constraints of limited space in captivity. The reproductive physiology of bears is characterized by two main traits--seasonality and delayed implantation, which is progesterone dependent. This offers the opportunity to interrupt early pregnancy by short-term administration of antiprogestins. The aim of the present study was to investigate the pharmacological characteristics of antiprogestin J956 to establish an efficient contraceptive protocol for administration of J956 in captive bears. The J956 binds to the uterine progesterone receptor of bears (n = 2) with almost the same relative binding affinity (1.25) as progesterone. The blood serum level of J956 after oral (on four consecutive days) and single parenteral administration was determined by a modified progestin receptor assay. The relative bioavailability of J956 after oral administration was approximately 10% of the parenteral administration. The estimated half-life was 12 to 16 hours after oral administration. Parenteral treatment of J956 (10 mg/kg body mass) led to sustain plasma concentrations (6.4 +/- 1.3 ng/mL) in one black bear and in five brown bears. The plasma level lasted for almost 2 months. Oral and low dosage parenteral (1 mg/kg body mass) administration of J956 had no effect on ongoing pregnancies in bears. Whereas single parenteral administration with higher dosages of J956 (7.5 to 10 mg/kg body mass) efficiently prevented implantation of early embryos in eight female captive bears.


Assuntos
Anticoncepcionais Femininos/farmacologia , Progestinas/antagonistas & inibidores , Ursidae/fisiologia , Administração Oral , Animais , Disponibilidade Biológica , Anticoncepcionais Femininos/administração & dosagem , Anticoncepcionais Femininos/sangue , Feminino , Injeções Intramusculares/veterinária , Gravidez , Receptores de Progesterona/antagonistas & inibidores , Receptores de Progesterona/metabolismo , Ursidae/sangue , Ursidae/metabolismo
12.
J Steroid Biochem Mol Biol ; 78(1): 83-8, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11530288

RESUMO

The majority of data regarding biological effects of estrogens is based on studies in male rats or ovariectomized (Ovx) female rats. Therefore, in this study, the effects of estradiol treatment on the regulation of the hepatic estrogen receptor and the level of circulating angiotensinogen were examined in the intact female rat. The data were compared with that of the hypophysectomized (Hx) rat. Animals were treated with either low (physiological) or high (pharmacological) doses of estrogen. In intact rats, the hepatic estrogen receptor (ER) level increased with increasing doses of estrogen. This was in contrast to the Hx rats where growth hormone (GH) and dexametasone (Dex) in combination were the sole modulators of the estrogen receptor. The angiotensinogen level increased in normal rats after estrogen administration in a dose dependent manner, regardless of the mode of administration. The pure antiestrogen ICI 182 780 efficiently blocked the increase in circulating angiotensinogen. The conclusion is that in the normal female, estrogens are important modulators of the serum angiotensinogen level.


Assuntos
Angiotensinas/sangue , Estrogênios/farmacologia , Fígado/metabolismo , Receptores de Estrogênio/biossíntese , Angiotensinogênio/sangue , Animais , Dexametasona/farmacologia , Relação Dose-Resposta a Droga , Estradiol/análogos & derivados , Estradiol/farmacologia , Antagonistas de Estrogênios/farmacologia , Receptor alfa de Estrogênio , Feminino , Fulvestranto , Glucocorticoides/farmacologia , Hormônio do Crescimento/farmacologia , Técnicas Imunoenzimáticas , Hibridização In Situ , RNA Mensageiro/metabolismo , Radioimunoensaio , Ratos , Ratos Sprague-Dawley , Receptores de Estrogênio/metabolismo
13.
J Reprod Fertil Suppl ; 57: 249-54, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11787158

RESUMO

The aim of this study was to establish new methods for controlling reproduction in bears. Anti-progestins were used to interrupt pregnancies. In two consecutive years, the anti-progestin J956 was administered to 11 female bears (nine Ursus arctos, one Ursus tibethanus, one Tremarctos ornatus) living in zoos. The anti-progestin J956 was given orally (n = 4) or parenterally (n = 12). The anti-progestin was administered alone or in combination with ethinyloestradiol, and before or after embryo implantation. The effects of anti-progestin treatment were determined using ultrasonographic examination of the urogenital tract and by monitoring progesterone concentrations in the blood and faeces. Oral administration of anti-progestin was not successful (successful in 0 of 4); however, in contrast, none of the parenteral treated animals remained pregnant (successful in 12 of 12). Parenteral treatment with J956, with or without ethinyloestradiol, was effective in disrupting pregnancy before implantation (successful in 6 of 6) and after implantation (successful in 6 of 6), but administration one month after implantation (n = 2) resulted in incomplete resorption of the fetuses. In conclusion, the administration of anti-progestins may be a useful method for preventing embryo implantation in captive bears.


Assuntos
Abortivos/administração & dosagem , Aborto Induzido/veterinária , Aborto Animal , Animais de Zoológico , Oximas/administração & dosagem , Ocitócicos/administração & dosagem , Progesterona/antagonistas & inibidores , Ursidae , Administração Oral , Animais , Implantação do Embrião/efeitos dos fármacos , Implantação Tardia do Embrião/efeitos dos fármacos , Estrenos , Etinilestradiol/administração & dosagem , Feminino , Infusões Parenterais , Gravidez , Ultrassonografia Pré-Natal/veterinária
14.
Reprod Fertil Dev ; 13(4): 297-305, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11800168

RESUMO

Sulfamate substitution (-O-SO2-NH2) at carbon atom 3 of the steroid skeleton leads to orally active prodrugs of estrogens with much higher systemic, but lower hepatic, estrogenic activity than their parent steroids. This dissociation is achieved by first passage through the liver in erythrocytes, followed by systemic hydrolysis which releases the 'parent' estrogen. In the rat, orally administered tritiated estradiol sulfamate, unlike estradiol, appears in the circulation at high concentrations. At Cmax, approximately one third of the administered dose forms a depot in the circulation (98% in erythrocytes, 2% in plasma). Significant estradiol, estrone and estrone sulfate concentrations were recorded in plasma during depletion of the red blood cell pool. Estradiol sulfamate (J995) has no estrogen receptor affinity per se or estrogenic activity in vitro ( i.e. without hydrolysis). Its oral uterotropic activity in rats is approximately 100 times greater than that of estradiol, however, its hepatotropic activity is only marginally elevated. These functions include bile secretion, the secretion of angiotensinogen, lipoproteins (total and high-density lipoprotein cholesterol) and insulin-like growth factor I (IGF-I). Orally administred estradiol sulfamate led to systemic estrogenic effects without significant hepatic responses, whereas estradiol and other 'conventional' estrogens exerted parallel systemic and hepatic estrogenic effects. Sulfamate technology represents an approach to the use of natural estrogens for fertility control and hormone replacement therapy in both genders. In this context, reduced effects on hemostatic factors, angiotensinogen, bile and IGF-I secretion seem the most important aspects. In addition, blood concentrations of estrogens are less variable than with conventional estrogens.


Assuntos
Estradiol/farmacologia , Estrogênios/farmacologia , Sulfonamidas/farmacologia , Administração Oral , Animais , Bile/química , Bile/metabolismo , Estradiol/administração & dosagem , Estradiol/análogos & derivados , Estradiol/sangue , Estradiol/farmacocinética , Estrogênios/administração & dosagem , Estrogênios/farmacocinética , Estrona/sangue , Feminino , Fígado/efeitos dos fármacos , Perfusão , Pró-Fármacos , Ratos , Ratos Wistar , Receptores de Estrogênio/metabolismo , Sulfonamidas/administração & dosagem , Sulfonamidas/farmacocinética , Células Tumorais Cultivadas
15.
Pharmazie ; 56(11): 843-9, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11817166

RESUMO

To improve the ratio of non-hormonal to hormonal activity, estrogens 3 and 4 were modified at various molecule positions. Isomerization of the 14 alpha,15 alpha-methylene bridge, controlled 3-methoxy group cleavage with respect to the 14 alpha,15 alpha-methylene bridge stereochemistry, reduction of the 8-double bond, and substitution of the oxyfunctionality at C-17 by a methylene and a difluoromethylene moiety were in the focus. As a result of in vivo and in vitro tests, compounds 27 and 29 were selected as potential follow-up candidates of lead 3.


Assuntos
Congêneres do Estradiol/síntese química , Estradiol/análogos & derivados , Antioxidantes/química , Ciclopropanos/química , Estradiol/síntese química , Indicadores e Reagentes , Isomerismo , Espectroscopia de Ressonância Magnética , Conformação Molecular
16.
Steroids ; 65(10-11): 713-23, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-11108882

RESUMO

Studies were conducted to assess progesterone antagonists (PAs) and progesterone receptor modulators (PRMs) with respect to PR agonistic and antagonistic activities in vivo. These properties are not always adequately reflected in transactivation in vitro models. Studies were performed in pregnant rats, estrogen-primed rabbits (McPhail -Test), and cycling and pregnant guinea pigs. Tested compounds included mifepristone (RU486), onapristone, J867, J956, J1042, and ZK137316. J-compounds induced sub-maximum endometrial transformation and, paradoxically, inhibited effects of progesterone in rabbits. Mifepristone, onapristone, and ZK137316 behaved as 'pure' antagonists in this species. Inhibition of uterine PGF(2alpha) secretion and inhibition of luteolysis in cycling guinea pigs were more sensitive parameters of PR-agonistic and antagonistic properties. 'Pure' PAs inhibited uterine PGF(2alpha) secretion and luteal regression completely. The PR agonist R5020 reversed both effects which demonstrates a PR mediation. Agonistic PRMs (J-substances and mifepristone) showed no or blunted antiluteolytic effects compared to the 'pure' PR antagonist onapristone. When tested in pregnant guinea pigs for their labor-inducing potential, PR agonistic PRMs had much reduced or abolished abortifacient activity compared to mifepristone (mifepristone > J956 > J867/J912 > J1042). However, in cycling animals, superior antiovulatory and antiproliferative properties of the J-substances were seen. Antiovulatory effects of 'pure' and agonistic PRMs are probably due to different mechanisms. The relevance of rodent studies for antiovulatory and uterine antiproliferative effects for the human is still uncertain. The non-abortifacient PRM J1042 induced stromal compaction and inhibition of endometrial proliferation in monkeys, but this effect was not stronger than that of the 'purer' PAs. 'Pure' PAs are important pharmacological tools analogous to PRKO models to study the role of PR in the menstrual cycle and in pregnancy.


Assuntos
Sistema Endócrino/efeitos dos fármacos , Progesterona/antagonistas & inibidores , Receptores de Progesterona , Aborto Induzido , Animais , Avaliação Pré-Clínica de Medicamentos , Feminino , Cobaias , Modelos Animais , Gravidez , Taxa de Gravidez , Progesterona/sangue , Coelhos , Receptores de Progesterona/agonistas , Receptores de Progesterona/antagonistas & inibidores , Receptores de Progesterona/metabolismo
17.
Steroids ; 65(10-11): 741-51, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-11108885

RESUMO

Progesterone antagonists (PAs, antiprogestins) can modulate estrogenic effects in various estrogen-dependent tissues. These modulatory effects are complex and depend on species, tissue, type of compound, dose, and duration of treatment. In non-human primates, PAs, including mifepristone, ZK 137 316 and ZK 230 211, inhibit endometrial proliferation and induce amenorrhea. When administered chronically at relatively low doses, these compounds block the mitotic activity of endometrial epithelium and induce stromal compaction in a dose-dependent manner in both spayed and intact monkeys at high estradiol concentrations. These effects were accompanied by an atrophy of spiral arteries. The antiproliferative effects were endometrium-specific, since the estrogenic effects in the oviduct and vagina were not inhibited by PAs. Similar endometrial antiproliferative effects were also found after treatment with the progesterone receptor modulator (PRM), mesoprogestin J1042. The endometrial antiproliferative effects of PAs, particularly within the endometrial glands, were also observed in spayed rabbits. In spayed rats, however, the PAs did not inhibit, but rather enhanced, various estrogen responses, including endometrial proliferation, pointing to species-specific differences. In conclusion, our studies indicate that both pure PAs and PRMs selectively inhibit estrogen-dependent endometrial proliferation in the primate endometrium without affecting estrogenic response in other estrogen-dependent tissues or inducing unscheduled bleeding. Our studies indicate that the spiral arteries, which are unique to the primate endometrium, are the primary targets that are damaged or inhibited by PAs and PRMs. The damage to these unique vessels may underlay the paradoxical, endometrium-specific, antiproliferative effects of these compounds. Hence, the properties of PAs and PRMs (mesoprogestins) open up new applications in gynecological therapy and hormone replacement therapy.


Assuntos
Endométrio/efeitos dos fármacos , Antagonistas de Hormônios/farmacologia , Progesterona/antagonistas & inibidores , Animais , Vasos Sanguíneos/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Endométrio/irrigação sanguínea , Endométrio/citologia , Feminino , Humanos , Receptores de Estrogênio/efeitos dos fármacos , Receptores de Estrogênio/metabolismo , Receptores de Progesterona/efeitos dos fármacos , Receptores de Progesterona/metabolismo
18.
J Steroid Biochem Mol Biol ; 74(3): 99-107, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11086229

RESUMO

Estradiol sulfamate (J995), estradiol-17beta with a substituted sulfamate group in position 3, has much higher systemic estrogenic activity after oral administration than 17beta-estradiol (E2) due to reduced hepatic metabolism of the drug. The lower dose necessary for achievement of adequate systemic estrogenic effects results in a substantial reduction of otherwise commonly observed hepatic side-effects. This makes J995 a strong candidate as an estrogen suitable for oral administration. The present study was performed to examine and compare the effects of J995 and E2 on the uterus after oral or subcutaneous administration to ovariectomized or ovariectomized+hypophysectomized female rats, in particular on the levels of the estrogen receptor (ER) (alpha+beta), ERalpha mRNA and insulin-like growth factor-I (IGF-I) mRNA. The ER levels were determined using a ligand binding assay and the mRNA levels using solution hybridization. The doses of J995 or E2 were chosen to achieve comparable uterotrophic activity. The rats were treated with hormones for 7 days and the treatment was initiated 14 days after surgery. We conclude that there are no major differences in the uterine response to treatment with J995 or E2 with respect to the effects on ER and ERalpha mRNA levels. The IGF-I mRNA level though, is more affected by J995 than by E2 after 7 days of treatment, indicating a prolonged effect of J995.


Assuntos
Estradiol/farmacologia , Hipofisectomia , Ovariectomia , Sulfonamidas/farmacologia , Útero/efeitos dos fármacos , Administração Oral , Animais , Relação Dose-Resposta a Droga , Estradiol/administração & dosagem , Estradiol/análogos & derivados , Estradiol/sangue , Estradiol/química , Receptor alfa de Estrogênio , Feminino , Injeções Subcutâneas , Fator de Crescimento Insulin-Like I/genética , Tamanho do Órgão/efeitos dos fármacos , RNA Mensageiro/análise , RNA Mensageiro/genética , Radioimunoensaio , Ratos , Ratos Wistar , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Sulfonamidas/administração & dosagem , Sulfonamidas/química , Útero/metabolismo
19.
Steroids ; 64(7): 460-71, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10443902

RESUMO

The title compound 17 has been synthesized for the use as hapten in the development of a competitive enzyme immunoassay for estrogen sulfamates. The synthesis started from estradiol diacetate 2. Oxyfunctionalization at C-11 to give 11alpha-hydroxy steroid 8 was accomplished by hydroboration/alkaline hydrogen peroxide oxidation of the 9(11)-dehydro derivative 7, which was obtained from compound 2 via 9-hydroxylation with dimethyldioxirane. After transformation of compound 8 into the allyl ether 9, the side chain was thio-functionalized at the omega-position affording the thioate 11 in two steps. Selective silylether deprotection at position 3 followed by sulfamoylation gave the sulfamate 19, which in turn was demasked at position 17 and treated with sodium borohydride/aluminum chloride to liberate the side chain thiol. Alternatively, title compound 17 was synthesized via the disulfides 13-16. For the preparation of the immunogen the title compound 17 was coupled to bovine gamma globulin in a two-step procedure using an amine and thiol specific bifunctional crosslinker. The immunization of rabbits resulted in the formation of antibodies which clearly discriminated the sulfamoylated estrogens from the non-esterified estrogens. The use of a biotinylated hapten derivative as a tracer in combination with a streptavidin-peroxidase-tetramethylbenzidine based detection system allowed the measurement of estradiol 3-sulfamate (1) in the range of about 1 to 1000 pg/well.


Assuntos
Estrenos/química , Haptenos/química , Técnicas Imunoenzimáticas/métodos , Animais , Bovinos , Estrenos/análise , Haptenos/análise , Espectroscopia de Ressonância Magnética , Masculino , Espectrometria de Massas , Estrutura Molecular , Coelhos , Ácidos Sulfônicos
20.
Exp Toxicol Pathol ; 50(4-6): 458-64, 1998 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9784023

RESUMO

The strong hepatic estrogenic actions of ethinylestradiol (EE) are very likely to be the cause of the cardiovascular morbidity related to the use of combined oral contraceptives (COCs). This survey presents results of EE replacement in COCs with natural 17beta-estradiol (E2) in the following stages: reduction of EE to daily doses of 0.01 mg and concomitant replacement with E2 (as valerate, EV), complete replacement of EE with E2 using a novel multiphasic combination containing EV and the progestin dienogest (DNG), and the use of natural E2 to develop estrogen sulfamates (J 995) showing sufficient dissociation of uterine from liver estrogenicity. Recent data from preclinical and clinical studies show that these approaches seem to be promising.


Assuntos
Anticoncepcionais Orais Combinados/administração & dosagem , Congêneres do Estradiol/administração & dosagem , Estradiol/administração & dosagem , Etinilestradiol/administração & dosagem , Animais , Ensaios Clínicos como Assunto , Anticoncepcionais Orais Combinados/efeitos adversos , Esquema de Medicação , Estradiol/efeitos adversos , Estradiol/análogos & derivados , Congêneres do Estradiol/efeitos adversos , Etinilestradiol/efeitos adversos , Feminino , Humanos , Ciclo Menstrual/efeitos dos fármacos , Ciclo Menstrual/fisiologia , Folículo Ovariano/efeitos dos fármacos , Folículo Ovariano/fisiologia , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...