Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 104
Filtrar
1.
Hum Reprod Open ; 2019(4): hoz027, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31777761

RESUMEN

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.

2.
Hum Reprod ; 34(4): 623-634, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30865281

RESUMEN

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.


Asunto(s)
Estrenos/efectos adversos , Estrenos/uso terapéutico , Leiomioma/tratamiento farmacológico , Menorragia/tratamiento farmacológico , Oximas/efectos adversos , Oximas/uso terapéutico , Receptores de Progesterona/efectos de los fármacos , Neoplasias Uterinas/tratamiento farmacológico , Administración Oral , Adulto , Método Doble Ciego , Endometrio/efectos de los fármacos , Estrenos/administración & dosificación , Femenino , Estudios de Seguimiento , Humanos , Leiomioma/complicaciones , Menorragia/complicaciones , Persona de Mediana Edad , Oximas/administración & dosificación , Medición de Resultados Informados por el Paciente , Premenopausia , Calidad de Vida , Resultado del Tratamiento , Carga Tumoral/efectos de los fármacos , Neoplasias Uterinas/complicaciones
3.
Reprod Sci ; 22(5): 551-5, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25296695

RESUMEN

Endometriosis affects up to 10% of women of reproductive age and 176 million women worldwide. The prevalence in women with infertility is between 30% and 50% but may be higher in women with pelvic pain, interstitial cystitis, or irritable bowel syndrome. Cytokeratin 19 has been suggested as a potential biomarker in urine for the diagnosis of this condition. The objective of this study was to prospectively determine the accuracy and the performance of a urinary cytokeratin 19 (uCYFRA 21-1) test for diagnosing endometriosis. Ninety-eight consecutive women who underwent laparoscopy had a urinary sample obtained before surgery and were included in the study. Endometriosis was diagnosed by laparoscopy and pathology in 64.3% (63 of 98 women). The estimates and 95% confidence intervals for sensitivity, specificity, positive and negative predictive values, and likelihood ratios were 11.1% (4.5%-21.5%), 94.3% (80.8%-99.3%), 77.7% (39.9-97.1), 37% (27-47.9), 1.94 (0.43-8.86), and 0.94 (0.84-1.06), respectively. Despite the high specificity, the uCYFRA 21-1 test has limited value for clinical practice to discriminate between women with and without endometriosis.


Asunto(s)
Antígenos de Neoplasias/orina , Endometriosis/diagnóstico , Endometriosis/orina , Queratina-19/orina , Adulto , Área Bajo la Curva , Biomarcadores/orina , Endometriosis/cirugía , Femenino , Humanos , Laparoscopía , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Estudios Prospectivos , Curva ROC , Reproducibilidad de los Resultados , Urinálisis , Adulto Joven
4.
Prostate Cancer Prostatic Dis ; 15(1): 93-9, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22025196

RESUMEN

BACKGROUND: This open-label study evaluated the efficacy and safety of a new leuprolide acetate 45 mg 6-month depot formulation in 151 men with prostate cancer who received 2 intramuscular injections administered 24 weeks apart. METHODS: The primary efficacy measurement was the proportion of patients achieving suppression of serum testosterone to ≤ 50 ng dl(-1) from week 4 through week 48. Adverse events (AEs) and hormonal and safety laboratory values were monitored. RESULTS: The primary efficacy end point was achieved in 93.4% of subjects (95% confidence interval (89.2%, 97.6%)). There were nine escapes from testosterone suppression during the study, none of which were accompanied by a rise in PSA. By week 4, mean testosterone concentration was suppressed below castrate levels to 15.9 ng dl(-1); suppression was maintained for the entire 24-week duration of each depot injection. No mean increase in testosterone was observed after the second injection. Mean PSA levels were maintained below 3 ng ml(-1) from week 14 through the 48-week treatment period. The most frequent AE was flushing (58.3%). Injection site reactions were reported in 24.5% of patients. CONCLUSIONS: Leuprolide acetate 45 mg 6-month depot demonstrated rapid and sustained testosterone suppression through 12 months and was well tolerated. This 6-month leuprolide acetate depot will decrease the number of annual injections in the treatment of prostate cancer.


Asunto(s)
Antagonistas de Andrógenos/uso terapéutico , Antineoplásicos Hormonales/uso terapéutico , Leuprolida/uso terapéutico , Neoplasias de la Próstata/tratamiento farmacológico , Anciano , Anciano de 80 o más Años , Antagonistas de Andrógenos/efectos adversos , Antagonistas de Andrógenos/farmacocinética , Anemia/sangre , Anemia/inducido químicamente , Antineoplásicos Hormonales/efectos adversos , Antineoplásicos Hormonales/farmacocinética , Biomarcadores de Tumor/sangre , Glucemia , Química Farmacéutica , Hemoglobinas/metabolismo , Humanos , Leuprolida/efectos adversos , Leuprolida/farmacocinética , Hormona Luteinizante/sangre , Masculino , Persona de Mediana Edad , Antígeno Prostático Específico/sangre , Neoplasias de la Próstata/sangre , Testosterona/sangre , Resultado del Tratamiento
5.
J Med Econ ; 14(1): 115-23, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21222565

RESUMEN

OBJECTIVE: This descriptive study assessed the rate and costs of surgical procedures among newly diagnosed endometriosis patients. METHODS: Utilizing the Medstat MarketScan database, commercially insured women aged 18-45 with endometriosis newly diagnosed during 2006-2007 were identified. Each endometriosis patient was matched to four women without endometriosis (population controls) based on age and region of residence. Surgical procedures received during the 12 months post-diagnosis were assessed. Costs of surgical procedures were the amount paid by the insurance companies. RESULTS: This study identified 15,891 women with newly diagnosed endometriosis and 63,564 population controls. More than 65% of endometriosis patients received an endometriosis-related surgical procedure within 1 year of the initial diagnosis. The most common procedure was therapeutic laparoscopy (31.6%), followed by abdominal hysterectomy (22.1%) and vaginal hysterectomy (6.8%). Prevalence and type of surgery performed varied by patient age, including a hysterectomy rate of approximately 16% in patients younger than 35 and 37% among patients aged 35-45 years. Average costs ranged from $4,289 (standard deviation [SD]: $3,313) for diagnostic laparoscopy to $11,397 (SD: $8,749) for abdominal hysterectomy. LIMITATIONS: Diagnosis of endometriosis cannot be validated against medical records, and information on the severity of endometriosis-related symptoms is not available in administrative claims data. CONCLUSIONS: Over 65% of patients had endometriosis-related surgical procedures, including hysterectomy, within 1 year of being diagnosed with endometriosis. The cost of surgical procedures related to endometriosis places a significant financial burden on the healthcare system.


Asunto(s)
Endometriosis/cirugía , Procedimientos Quirúrgicos Operativos/economía , Adolescente , Adulto , Comorbilidad , Costos y Análisis de Costo , Bases de Datos Factuales , Endometriosis/diagnóstico , Femenino , Humanos , Auditoría Médica , Persona de Mediana Edad , Estados Unidos , Adulto Joven
6.
Hum Reprod ; 24(5): 1036-44, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19176543

RESUMEN

BACKGROUND: The selective progesterone receptor modulator asoprisnil suppresses uterine bleeding and decreases leiomyoma volume while maintaining follicular phase estrogen concentrations. For safety of potential clinical applications, any proliferative effect of asoprisnil on uterine tissues, particularly endometrium, needs to be established. METHODS: In a double-blind, randomized, placebo-controlled study (continuation of previously published trial No. NCT00150644 (Williams et al., 2007 and Wilkens et al., 2008)), 33 patients with symptomatic uterine leiomyomata received placebo, 10 or 25 mg asoprisnil daily for 12 weeks before hysterectomy. Proliferation markers Ki-67 and anti-phospho-histone H3 (PH3) were immunolocalized in endometrium, myometrium and leiomyoma tissue. Endometrial PTEN (phosphatase and tensin homologue, a tumour suppressor gene) expression was also assessed by immunohistochemistry. PH3-positive glandular and stromal cells were counted per measured endometrial area. Endometrial Ki-67 expression was assessed using stereological methods. Stained myometrial and leiomyoma cells were counted per 10 fields (x250). PTEN immunostaining was quantified using a histoscore. Each asoprisnil group was compared with placebo (secretory phase) with significance at 0.05 level. RESULTS: Endometrial epithelial proliferation and PTEN expression were not significantly different between placebo and asoprisnil groups. Decreased stromal Ki-67 expression (P < 0.05) suggested any effect of asoprisnil on endometrial proliferation to be inhibitory. Immunolocalization of PTEN expression was not different between treatment groups in any tissue compartments. Myometrial Ki-67 expression decreased following asoprisnil 25 mg (P < 0.05). CONCLUSIONS: Asoprisnil does not induce proliferation of uterine tissues and does not suppress endometrial PTEN expression.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Estrenos/farmacología , Miometrio/efectos de los fármacos , Oximas/farmacología , Fosfohidrolasa PTEN/genética , Útero/efectos de los fármacos , Adulto , Endometrio/efectos de los fármacos , Endometrio/metabolismo , Femenino , Expresión Génica/efectos de los fármacos , Marcadores Genéticos , Humanos , Inmunohistoquímica , Antígeno Ki-67/metabolismo , Leiomioma/metabolismo , Leiomioma/patología , Persona de Mediana Edad , Miometrio/metabolismo , Fosfohidrolasa PTEN/metabolismo , Útero/metabolismo , Útero/patología
7.
Hum Reprod ; 22(6): 1696-704, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17339234

RESUMEN

BACKGROUND: Asoprisnil is a selective progesterone receptor modulator with mixed progesterone agonist/antagonist activity which controls uterine bleeding via an endometrial effect. This study examined full-thickness endometrial, leiomyoma and myometrial morphology in hysterectomy specimens from patients with uterine leiomyomata, after treatment with asoprisnil for 3 months. METHODS: In this double-blind, randomized, placebo-controlled study, 33 subjects with uterine leiomyomata were randomized to receive asoprisnil 10, 25 mg or placebo for an average of 95 days prior to hysterectomy. Samples of endometrium, myometrium and leiomyoma tissue were subjected to systematic morphological assessment with quantification of mitotic activity. RESULTS: In patients treated with 10 or 25 mg asoprisnil, a unique pattern called 'non-physiologic secretory effect' was evident in endometrium, recognizable through partially developed secretory glandular appearances and stromal changes. Endometrial thickness was decreased, and there were low levels of mitotic activity in endometrial glands and stroma. Unusual thick-walled muscular arterioles and prominent aggregations of thin-walled vessels were present in endometrial stroma, but not in myometrium or non-endometrial vascular beds. Mitotic activity was decreased in leiomyomata. CONCLUSIONS: Asoprisnil induces unique morphological changes and is associated with low levels of glandular and stromal proliferation in endometrium, and in leiomyomata. These changes are likely to contribute to the amenorrhoea experienced after exposure to the medication.


Asunto(s)
Estrenos/efectos adversos , Leiomioma/patología , Oximas/efectos adversos , Receptores de Progesterona/efectos de los fármacos , Neoplasias Uterinas/patología , Útero/efectos de los fármacos , Útero/patología , Adulto , Endometrio/efectos de los fármacos , Endometrio/patología , Estrenos/administración & dosificación , Femenino , Humanos , Leiomioma/tratamiento farmacológico , Persona de Mediana Edad , Miometrio/efectos de los fármacos , Miometrio/patología , Oximas/administración & dosificación , Placebos , Neoplasias Uterinas/tratamiento farmacológico
8.
Hum Reprod ; 21(12): 3081-90, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16936297

RESUMEN

BACKGROUND: Clinicians routinely prescribe progestins along with estrogens during menopausal hormone therapy (HT) to block estrogen-dependent endometrial proliferation. Breakthrough bleeding (BTB) can negate the utility of this treatment. Because progestin antagonists also inhibit estrogen-dependent endometrial proliferation in women and macaques, we used a menopausal macaque model to determine whether a potent progestin antagonist (ZK 230 211, Schering AG; ZK) combined with estrogen would provide a novel mode of HT. METHOD: Ovariectomized rhesus macaques were treated for 5 months with either estradiol (E(2)) alone, E(2) + progesterone (two doses) or E(2) + ZK (0.01, 0.05 or 0.25 mg/kg). RESULTS: In the E(2) + progesterone groups, progesterone suppressed endometrial proliferation and induced a thick decidualized endometrium. In the E(2) + ZK 230 211 groups, all doses of ZK blocked endometrial proliferation and induced endometrial atrophy. In all ZK-treated groups, the atrophied endometrium contained some dilated glands lined by an inactive, flattened, non-mitotic epithelium. BTB was much lower in the E(2) + ZK groups (17 days of spotting, all groups) than in the E(2) and E(2) + progesterone groups (155 bleeding days, all groups). ZK suppressed E(2) effects in the cervix, but not in the vagina, oviduct or mammary glands. All serum chemistry and lipid profiles were normal. CONCLUSION: The ability of ZK to block estrogen-dependent endometrial proliferation, induce endometrial atrophy and suppress BTB in a menopausal macaque model indicates that progestin antagonists may provide a novel mode of HT.


Asunto(s)
Endometrio/efectos de los fármacos , Estradiol/administración & dosificación , Estrenos/administración & dosificación , Antagonistas de Hormonas/administración & dosificación , Menopausia/efectos de los fármacos , Metrorragia/prevención & control , Progesterona/antagonistas & inhibidores , Animales , Proliferación Celular , Modelos Animales de Enfermedad , Quimioterapia Combinada , Endometrio/patología , Estradiol/sangre , Femenino , Genitales Femeninos/efectos de los fármacos , Macaca mulatta , Glándulas Mamarias Animales/efectos de los fármacos
9.
Hum Reprod ; 18(1): 69-76, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12525443

RESUMEN

BACKGROUND: Administration of low doses of an antiprogestin to rhesus monkeys permits ovarian/menstrual cyclicity, suppresses endometrial proliferation and prevents pregnancy without adverse or toxic side-effects after 5-6 months of daily treatment. The purpose of this study was to test the reversibility with respect to restoration of fertility after 1 year of low-dose antiprogestin treatment. METHODS: This experiment included a daily 1 year vehicle- or antiprogestin-treatment interval followed by a 9 month post-treatment interval for adult, female rhesus monkeys (n = 5/group) of proven fertility and exhibiting regular menstrual cycles. Co-habitation occurred with a male of proven fertility and vaginal swabs were taken to identify the presence of sperm during the treatment (antiprogestin females) and post-treatment intervals (vehicle and antiprogestin females). RESULTS: Mating and vaginal sperm were evident in all antiprogestin females during, and, in both groups, after treatment. Based on ultrasonography, none of the antiprogestin-treated females became pregnant during the treatment interval. However, upon cessation of treatment, pregnancy rates were similar between antiprogestin-treated (3/5) relative to vehicle-treated (4/5) females with live, healthy infants born in both groups. There were no differences between groups in fetal measurements, gestation lengths, live birth rates and infant weights. CONCLUSIONS: The reversal of the anti-fertility effects of chronic, low-dose antiprogestin treatment supports the clinical feasibility of potent and selective antiprogestins as potential contraceptives for women.


Asunto(s)
Anticonceptivos Femeninos/administración & dosificación , Antagonistas de Hormonas/administración & dosificación , Progestinas/antagonistas & inhibidores , Esteroides/administración & dosificación , Animales , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Femenino , Fertilidad/efectos de los fármacos , Inyecciones Intramusculares , Masculino , Ciclo Menstrual/efectos de los fármacos , Embarazo , Índice de Embarazo
10.
Mol Hum Reprod ; 8(5): 465-74, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-11994544

RESUMEN

The interaction between nitric oxide (NO), progesterone and the MAPkinase signalling pathway involved in decidualization was studied using immunohistochemistry during implantation in the rat. Early pregnant rats were treated with the inhibitor of nitric oxide synthesizing enzyme iNOS, aminoguanidine, either alone or in combination with the low dose antiprogestin, onapristone. The combined treatment was most effective on days 7 and 9 post coitum leading to a complete loss of embryos. The expression pattern of activated MAPkinases, Erk1/2 and iNOS appeared to be associated with the differentiation process of decidualization. A maximum staining of both enzymes was observed on day 9 post coitum in the mesometrial decidua. In addition, Erk1/2 and iNOS were highly coexpressed around the mesometrial sinusoids. Combined treatment with aminoguanidine and onapristone for 3 days led to a transient suppression of Erk1/2 and abolished Cox2 expression. Concomitantly, angiogenesis was reduced and dilated sinusoids were missing in the mesometrial decidua. In conclusion, our study suggests that (i) the member of the mitogen-activated protein kinase (MAPK) family, Erk1/2, is activated during implantation and may play an important role during the decidualization process, and (ii) this enzyme may be regulated by both progesterone and NO.


Asunto(s)
Decidua/fisiología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Óxido Nítrico/metabolismo , Progesterona/metabolismo , Animales , Decidua/irrigación sanguínea , Decidua/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Femenino , Gonanos/farmacología , Guanidinas/farmacología , Proteína Quinasa 1 Activada por Mitógenos/efectos de los fármacos , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa/efectos de los fármacos , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo II , Embarazo , Resultado del Embarazo , Preñez , Progesterona/farmacología , Ratas , Ratas Wistar , Transducción de Señal
11.
J Med Primatol ; 31(6): 330-9, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12519211

RESUMEN

The expression of endometrial beta3 integrin and insulin-like growth factor binding protein-1 (IGFBP-1) was studied in cycling and pregnant vervet monkeys. There were clear changes of beta3 integrin expression during the menstrual cycle, with the strongest immunostaining observed on day 26. Moderate to strong expression was observed during pregnancy. The expression of IGFBP-1 during the menstrual cycle was weak but upregulated during pregnancy with moderate to strong staining. The administration of a single dose of onapristone at 10 mg/kg on days 17, 21 and 22 of the menstrual cycle, followed by a biopsy on days 22, 22 and 26, respectively, and during pregnancy (34-44 days menstrual age) 24 h before the biopsy, disrupted and desynchronized the endometrium. However, no effect on beta3 integrin expression could be observed and staining reflected the untreated patterns. The same applied to IGFBP-1 except that during pregnancy the expression of this protein was reduced or abolished. The results suggest that beta3 integrin is associated with endometrial receptivity in vervet monkeys and that IGFBP-1 plays an important role during pregnancy in this species. The administration of onapristone appeared to only influence IGFBP-1 expression. To our knowledge, this is the first time that these endometrial proteins have been investigated in vervet monkeys. This study should therefore contribute to improving our understanding of the reproductive function of this species.


Asunto(s)
Chlorocebus aethiops/fisiología , Endometrio/fisiología , Proteína 1 de Unión a Factor de Crecimiento Similar a la Insulina/biosíntesis , Integrina beta3/biosíntesis , Ciclo Menstrual/fisiología , Preñez/fisiología , Progesterona/metabolismo , Animales , Biopsia/veterinaria , Chlorocebus aethiops/anatomía & histología , Chlorocebus aethiops/metabolismo , Endometrio/anatomía & histología , Endometrio/metabolismo , Femenino , Gonanos/farmacología , Antagonistas de Hormonas/farmacología , Inmunohistoquímica/veterinaria , Proteína 1 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Integrina beta3/metabolismo , Masculino , Embarazo , Preñez/metabolismo , Progesterona/antagonistas & inhibidores
12.
Hum Reprod ; 16(8): 1562-74, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11473944

RESUMEN

BACKGROUND: A reliable means of menstrual suppression would greatly improve the quality of life for women. Information is lacking on the direct endometrial effects and appropriate dosages of new antiprogestins that may be useful for this purpose. METHODS: The current work evaluated three different systems in macaque monkeys. First, the range of doses of two relatively new antiprogestins, ZK 137 316 and ZK 230 211, that would block progesterone action directly on the endometrium in artificially cycled, spayed rhesus macaques; second, the direct endometrial effects of ZK 230 211, a type III antiprogestin; and third, investigation of whether endometrial-suppressive doses administered chronically to intact, cycling monkeys could be used for reversible, menstrual suppression. RESULTS: The results in naturally cycling animals showed that ZK 137 316 blocked menstruation in all animals, but doses of 0.05 mg/kg blocked ovulation in 55.5% of animals and doses of 0.1 mg/kg blocked ovulation in 66.6% of the animals. However, all doses of ZK 230 211 that blocked menstruation also blocked ovulation. All progesterone antagonist (PA)-treated animals, regardless of dose, maintained normal follicular phase concentrations of oestradiol and returned to normal menstrual cyclicity within 15--41 days post-treatment. Therefore ZK 137 316, depending on dose, can allow ovulation but block menstruation, while ZK 230 211, a much more potent PA, blocks both ovulation and menstruation at all effective doses. Both PAs block unopposed oestrogenic action on the endometrium through their antiproliferative effects. CONCLUSIONS: Reversible amenorrhoea can be achieved with these two PAs, and they can protect the endometrium from the effects of unopposed oestrogen whether or not ovulation is blocked. Chronic, low dose PA treatment may provide a new option for women who wish to suppress their menstrual periods.


Asunto(s)
Antagonistas de Hormonas/farmacología , Menstruación/efectos de los fármacos , Progesterona/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Endometrio/anatomía & histología , Endometrio/química , Endometrio/efectos de los fármacos , Estradiol/sangre , Estradiol/farmacología , Estrenos/administración & dosificación , Estrenos/farmacología , Receptor alfa de Estrógeno , Trompas Uterinas/efectos de los fármacos , Femenino , Antígeno Ki-67/análisis , Macaca mulatta , Ovariectomía , Ovulación/efectos de los fármacos , Progesterona/sangre , Progesterona/farmacología , Receptores de Estrógenos/análisis , Receptores de Progesterona/análisis , Esteroides/administración & dosificación , Esteroides/farmacología
13.
J Clin Endocrinol Metab ; 86(6): 2668-79, 2001 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11397870

RESUMEN

Antiprogestins (APs) inhibit estradiol (E(2))-stimulated endometrial growth in women and nonhuman primates, but the mechanism of this "antiestrogenic" action is unknown. Here, we report that APs up-regulate endometrial androgen receptor (AR) in both women and macaques, an effect that might play a role in the antiproliferative effects of APs on the primate endometrium. In addition, because there are discrepancies in the literature on the regulation and localization of AR in the primate endometrium, we used both in situ hybridization and immunocytochemistry to evaluate hormonal influences on endometrial AR in women and macaques. In ovariectomized macaques, the following treatments were given for 4 weeks each: E(2) alone, E(2) + progesterone (P), E(2) + mifepristone (RU 486), and E(2) + P + RU 486. In women, samples were obtained during the normal menstrual cycle and after treatment with either RU 486 for 30 days at 2 mg/day, or after a single oral administration of 200 mg RU 486 on cycle day LH + 2. In macaques, E(2) significantly increased AR expression above vehicle controls; E(2) + RU 486 increased binding further; E(2) + P decreased AR binding; and E(2) + P + RU 486 treatment caused an intermediate elevation in AR binding. In macaques treated with E(2) alone, stromal AR staining was predominant, and P treatment suppressed that staining. E(2) + RU 486 or E(2) + P + RU 486 treatment produced a striking up-regulation of glandular epithelial AR staining and enhanced the stromal AR signal. In situ hybridization analyses confirmed the immunocytochemistry data. Similar induction of glandular AR staining and enhanced stromal AR staining were obtained in macaques treated with ZK 137 316 and ZK 230 211. During the natural cycle in women, stromal AR staining predominated and was greater in the proliferative than the late secretory phase. RU 486 treatment of women up-regulated glandular epithelial AR staining after either daily treatment for 30 days with 2 mg/day or after a single oral dose of 200 mg. In summary, endometrial AR was highest in the stroma during the human proliferative phase (or during E(2) treatment in macaques) and lowest during the late secretory phase in women (or after E(2) + P treatment in macaques). In both species, RU 486 induced AR expression in the glands and enhanced AR expression in stromal cells. Because androgens can antagonize E(2) action, enhanced endometrial AR expression induced by APs could play a role in the antiproliferative, "antiestrogenic" effects of APs in primates.


Asunto(s)
Endometrio/efectos de los fármacos , Endometrio/metabolismo , Antagonistas de Hormonas/farmacología , Macaca mulatta/metabolismo , Mifepristona/farmacología , Progesterona/antagonistas & inhibidores , Receptores Androgénicos/metabolismo , Animales , Sinergismo Farmacológico , Estradiol/farmacología , Femenino , Humanos , Inmunohistoquímica , Hibridación in Situ , Ovariectomía , Progesterona/farmacología , Regulación hacia Arriba
14.
Steroids ; 65(10-11): 713-23, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-11108882

RESUMEN

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.


Asunto(s)
Sistema Endocrino/efectos de los fármacos , Progesterona/antagonistas & inhibidores , Receptores de Progesterona , Aborto Inducido , Animales , Evaluación Preclínica de Medicamentos , Femenino , Cobayas , Modelos Animales , Embarazo , Índice de Embarazo , Progesterona/sangre , Conejos , Receptores de Progesterona/agonistas , Receptores de Progesterona/antagonistas & inhibidores , Receptores de Progesterona/metabolismo
15.
Steroids ; 65(10-11): 763-71, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-11108887

RESUMEN

The influences of the synthetic progestin, medroxyprogesterone acetate (MPA), the progesterone receptor modulator J867, and the antagonist ZK137316 were studied in vitro on isolated endometrial epithelial cells, as well as endometrial fibroblasts. We evaluated the expression of estrogen receptor alpha (ER) and the progesterone receptor (PR) by RT-PCR. ER and PR were strongly expressed in the fibroblasts and epithelial cells under treatment with 10(-8) M 17beta-estradiol (E(2)). Treatment with 10(-6) M J867 or ZK137316 upregulated the PR expression as did E(2), in contrast to treatment with 10(-6) M MPA, which caused a downregulation of PR in epithelial cells, but not in fibroblasts. In addition, the vascular endothelial growth factor (VEGF) release into the cell culture medium was analyzed by a VEGF-ELISA. VEGF which plays an important role in angiogenesis, is regulated by steroid hormones as well as hypoxia. E(2) stimulates VEGF release into the medium in both cell types. MPA reduces VEGF release significantly in the fibroblast cell culture, but increases it in the epithelial cell culture. ZK137316, in the presence or absence of E(2), reduces VEGF release in fibroblast cell culture. J867 increases the VEGF production in fibroblasts only in the presence of E(2). Both compounds show no significant effects, compared to E(2), in epithelial cell culture. The different results for the epithelial cells and fibroblasts indicate that the pharmacological effects of PR modulators (PRMs) and progesterone antagonists (PAs) may be cell specific and depend on the presence or absence of partial progestagenic agonistic activities. This observation opens up new perspectives for various clinical applications.


Asunto(s)
Endometrio/citología , Factores de Crecimiento Endotelial/metabolismo , Linfocinas/metabolismo , Congéneres de la Progesterona/farmacología , Receptores de Progesterona/efectos de los fármacos , Técnicas de Cultivo de Célula , Endometrio/química , Endometrio/metabolismo , Células Epiteliales/química , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Estradiol/farmacología , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Antagonistas de Hormonas/farmacología , Humanos , Inmunohistoquímica , Linfocinas/efectos de los fármacos , Acetato de Medroxiprogesterona/farmacología , Progesterona/antagonistas & inhibidores , ARN Mensajero/efectos de los fármacos , Receptores de Estrógenos/efectos de los fármacos , Receptores de Estrógenos/genética , Receptores de Progesterona/genética , Esteroides/farmacología , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
16.
Steroids ; 65(10-11): 741-51, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-11108885

RESUMEN

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.


Asunto(s)
Endometrio/efectos de los fármacos , Antagonistas de Hormonas/farmacología , Progesterona/antagonistas & inhibidores , Animales , Vasos Sanguíneos/efectos de los fármacos , División Celular/efectos de los fármacos , Endometrio/irrigación sanguínea , Endometrio/citología , Femenino , Humanos , Receptores de Estrógenos/efectos de los fármacos , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/efectos de los fármacos , Receptores de Progesterona/metabolismo
17.
Steroids ; 65(10-11): 807-15, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-11108892

RESUMEN

Both progesterone receptor modulators (PRMs) as well as pure progesterone antagonists (PAs) have numerous proven and potential therapeutic applications in female health care. Mifepristone, a PRM with only marginal agonistic activity, together with a prostaglandin can terminate pregnancies of less than 9 weeks duration; mifepristone is also used in the preparation of women at later gestational stages whose pregnancies are terminated with prostaglandins or surgery. Mifepristone causes expulsion of the uterine contents following intrauterine fetal death and promotes dilation of the non-pregnant primigravid uterus. It is also effective in the treatment of missed abortion. Together with methotrexate, mifepristone can be used in the medical treatment of ectopic pregnancy. Both PAs and PRMs display antiproliferative effects on the endometrium. Because of this, they have application in the treatment of endometriosis, an estrogen-dependent condition. They may also be utilized to reduce myoma size, acting as both a PA and antiproliferative agent. Unlike GnRH agonists, long-term use in endometriosis and myoma is not associated with loss of bone and hypoestrogenism. PRMs may also be useful in IVF programs to prevent a premature LH surge and to delay the emergence of the implantation window. Some PRMs have potential use as hormone replacement therapy in women during menopause or in those with dysfunctional uterine bleeding.


Asunto(s)
Antagonistas de Hormonas/uso terapéutico , Progesterona/antagonistas & inhibidores , Abortivos no Esteroideos/farmacología , Abortivos no Esteroideos/uso terapéutico , Animales , Femenino , Enfermedades de los Genitales Femeninos/tratamiento farmacológico , Antagonistas de Hormonas/farmacología , Humanos , Mifepristona/farmacología , Mifepristona/uso terapéutico , Embarazo , Receptores de Progesterona/agonistas , Receptores de Progesterona/antagonistas & inhibidores
18.
Hum Reprod ; 15 Suppl 3: 96-111, 2000 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11041226

RESUMEN

Nitric oxide (NO) is a major paracrine mediator of various biological processes, including vascular functions and inflammation. In blood vessels, NO is produced by the low-input constitutive endothelial NO synthase (eNOS) and is a potent vasodilator and platelet aggregation inhibitor. The inducible NOS isoform (iNOS) is capable of producing NO at high concentrations which have pro-inflammatory properties. Immunohistochemical and molecular studies of endometrial NOS expression, as well as animal experiments with NOS inhibitors, indicate that NO plays an important role in endometrial functions such as endometrial receptivity, implantation and menstruation. In rodents, both iNOS and eNOS are highly up-regulated in the implantation sites, and NOS inhibitors show synergistic effects with antiprogestins in inhibiting the establishment of pregnancy. In the human endometrium, eNOS have been localized in the glandular epithelium and in endometrial microvascular endothelium, primarily during the luteal phase. iNOS has been found in the endometrial epithelium during menstruation, in immunocompetent endometrial cells, and in decidualized stromal cells. In primates, NO may be involved in the initiation and maintenance of menstrual bleeding by inducing tissue breakdown and vascular relaxation as well as by inhibiting platelet aggregation. Endometrium-derived NO may also play a role in myometrial relaxation during menstruation. These studies open up new applications for NO-donating and -inhibiting agents in uterine disorders. NO donors may be useful in the treatment of dysmenorrhoea and for promoting fertility. Antiprogestins, progesterone receptor modulators and iNOS inhibitors may find applications in the treatment and prevention of abnormal uterine bleeding.


Asunto(s)
Implantación del Embrión/fisiología , Menstruación/fisiología , Óxido Nítrico/fisiología , Animales , Endometrio/enzimología , Estrógenos/fisiología , Femenino , Humanos , Óxido Nítrico Sintasa/fisiología , Embarazo , Progesterona/fisiología , Hemorragia Uterina , Útero/inmunología
19.
J Clin Endocrinol Metab ; 85(9): 3442-52, 2000 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10999847

RESUMEN

Several reports indicate that vascular endothelial growth factor (VEGF) expression is increased in endometrial glands and stroma during the menstrual phase in the human endometrium. Here we report that VEGF receptor type 2 (KDR), normally expressed only in the vascular endothelium, was dramatically up-regulated in the stromal cells of the superficial endometrial zones during the premenstrual phase in both human and macaque endometrium. This increase was detectable by Northern analysis, in situ hybridization, and immunocytochemistry and was cell specific, zone specific, cycle phase specific, and VEGF receptor type specific. That is, it only occurred during the premenstrual/menstrual phase, did not occur in glandular epithelium, endothelium, or stromal cells of the deepest endometrial zones, and was not observed for VEGF receptor type 1. The upregulation of stromal KDR was induced by progesterone (P) withdrawal in both women and macaques, and adding back P 24 h after P withdrawal in macaques blocked stromal, but not vascular, endothelial KDR expression. Promatrix metalloproteinase-1 (MMP-1) was coordinately up-regulated in the same stromal cell population by P withdrawal. Because of reports that VEGF can enhance MMP expression, we hypothesize that VEGF-KDR interactions may influence MMP expression in the superficial zones of the primate endometrium during the premenstrual phase, and that these interactions play a role in the induction of menstruation.


Asunto(s)
Endometrio/metabolismo , Menstruación/metabolismo , Progesterona/efectos adversos , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Receptores de Factores de Crecimiento/biosíntesis , Síndrome de Abstinencia a Sustancias/metabolismo , Adulto , Animales , Especificidad de Anticuerpos , Northern Blotting , Hipoxia de la Célula , Endometrio/citología , Femenino , Humanos , Inmunohistoquímica , Hibridación in Situ , Macaca mulatta , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Proto-Oncogénicas/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Proteínas Tirosina Quinasas Receptoras/genética , Receptores de Factores de Crecimiento/genética , Receptores de Factores de Crecimiento Endotelial Vascular , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Esteroides/sangre , Células del Estroma/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular
20.
Biol Reprod ; 63(3): 820-5, 2000 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10952926

RESUMEN

Signals from the developing mammalian blastocyst rescue the corpus luteum (CL) and modulate the uterine environment in preparation for implantation and early pregnancy. Our previous studies demonstrated that both short- and long-term administration of chorionic gonadotropin (CG) markedly alters the morphology and the biochemical activity of the receptive endometrium. Because the effects of CG were superimposed on a progesterone-primed endometrium, this study was undertaken to determine if the inhibition of progesterone action by progesterone receptor antagonists (PRa) in intact and ovariectomized baboons would alter the action of CG on the endometrium at the time of uterine receptivity. In the short-term hCG-treated baboons, the PRa reduced the epithelial plaque reaction, completely inhibited alpha-smooth muscle actin (alphaSMA) expression in stromal fibroblasts, and induced the reappearance of the progesterone (PR) and estrogen (ERalpha) receptors in epithelial cells. However, this treatment protocol had no effect on the expression of glycodelin in the glandular epithelium. In contrast, glycodelin expression in addition to alphaSMA was suppressed in the ovariectomized animals. In the long-term hCG-treated baboons, the PRa had a similar effect on both alphaSMA, PR, and ER. In addition, this treatment also resulted in an inhibition of glycodelin expression in the glandular epithelium. These results indicate that blocking the action of progesterone on the endometrium even for a short period of time has a profound effect on the hCG-induced response in stromal fibroblasts. In contrast, for the diminution of glandular epithelial function in the presence of an ovary requires prolonged inhibition of progesterone action, suggesting a potential paracrine effect on the endometrium from the CL in response to hCG.


Asunto(s)
Gonadotropina Coriónica/farmacología , Antagonistas de Hormonas/farmacología , Receptores de Progesterona/antagonistas & inhibidores , Útero/efectos de los fármacos , Actinas/genética , Animales , Western Blotting , Endometrio/metabolismo , Epitelio/efectos de los fármacos , Epitelio/metabolismo , Femenino , Fibroblastos/metabolismo , Expresión Génica/efectos de los fármacos , Glicoproteínas/genética , Ovariectomía , Papio , Proteínas Gestacionales/genética , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...