Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Clin Pharmacokinet ; 61(11): 1571-1583, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36173592

RESUMO

BACKGROUND AND OBJECTIVE: Postmenopausal women often require estrogen supplementation to improve menopausal and postmenopausal vasomotor symptoms and maintain hormonal balance. Conjugated equine estrogens extracted from the urine of pregnant mares are commonly used to provide this estrogen replacement therapy. The complex composition of this mixture of animal sulfated metabolites makes its bioanalysis challenging such that its detailed pharmacokinetics has not been fully characterized. The purpose of this work is to reveal the pharmacokinetic behavior of conjugated equine estrogens in healthy Chinese postmenopausal women by a parallel two-column LC-MS/MS method. METHODS: An open-label study was carried out in 35 Chinese healthy postmenopausal women who received a single dose of Premarin® 0.625 mg. A high-throughput column-switching liquid chromatography-tandem mass spectrometry method was developed to determine four conjugated estrogens and two unconjugated estrogens formed by hydrolysis in vivo. The method multiplexes two high-performance liquid chromatography systems into one mass spectrometer and incorporates the positive/negative ion switching acquisition mode of mass spectrometry to significantly increase analysis efficiency. Pharmacokinetics was determined using non-compartmental methods. RESULTS: Both conjugated and unconjugated estrogens can be analyzed simultaneously in a single run with an analysis time of 13.0 minutes in the column-switching liquid chromatography-tandem mass spectrometry method as opposed to 23.0 minutes in a single-column liquid chromatography-tandem mass spectrometry system. The exposures (maximum concentration and area under the curve) of estrone and equilin in Chinese women were higher than those in the North American women. CONCLUSIONS: The fully validated assay was successfully applied to a pharmacokinetic study in healthy postmenopausal Chinese women after oral administration of a conjugated equine estrogen tablet. This study suggests that Chinese postmenopausal women achieve the same level of unconjugated estrogens in plasma at a lower dose of conjugated equine estrogens than North American women.


Assuntos
Estrogênios Conjugados (USP) , Pós-Menopausa , Animais , Feminino , Humanos , China , Cromatografia Líquida/métodos , Estrogênios/metabolismo , Estrogênios Conjugados (USP)/farmacocinética , Cavalos , Espectrometria de Massas em Tandem/métodos
2.
Endocrinology ; 149(10): 4857-70, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18599548

RESUMO

The human estrogen receptors (ERs) alpha and beta interact with 17beta-estradiol (17beta-E2), estrone, 17alpha-estradiol, and the ring B unsaturated estrogens, equilin, 17beta-dihydroequilin, 17alpha-dihydroequilin, equilenin, 17beta-dihydroequilenin, 17alpha-dihydroequilenin, Delta8-estrone, and Delta8, 17beta-E2 with varying affinities. In comparison to 17beta-E2, the relative binding affinities of most ring B unsaturated estrogens were 2- to 8-fold lower for ERalpha and ERbeta, however, some of these unique estrogens had two to four times greater affinity for ERbeta than ERalpha. The transcriptional activity of these estrogens in HepG2 cells transfected with ERalpha or ERbeta, or both, and the secreted-alkaline phosphatase gene showed that all estrogens were functionally active. 17beta-E2 induced the activity of secreted-alkaline phosphatase by ERalpha to a level higher than any other estrogen. Activity of other estrogens was 12-17% that of 17beta-E2. In contrast, 17beta-E2 stimulated the activity of ERbeta to a 5-fold lower level than that with ERalpha, whereas the activity of other estrogens was 66-290% that of 17beta-E2, with equilenin being the most active. The presence of both ER subtypes did not alter the functional activity of 17beta-E2, although it further enhanced the activity of 17beta-dihydroequilin (200%), 17beta-dihydroequilenin (160%), and Delta8, 17beta-E2 (130%). Except for 17beta-E2, no correlation was observed between the functional activities and their binding affinities for ER. In conclusion, our results show that the effects of ring B unsaturated estrogens are mainly mediated via ERbeta and that the presence of both ER subtypes further enhances their activity. It is now possible to develop hormone replacement therapy using selective ring B unsaturated estrogens for target tissues where ERbeta is the predominant ER.


Assuntos
Receptor alfa de Estrogênio/química , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/química , Receptor beta de Estrogênio/metabolismo , Estrogênios Conjugados (USP)/química , Estrogênios Conjugados (USP)/farmacocinética , Carcinoma Hepatocelular , Linhagem Celular Tumoral , Estradiol/química , Estradiol/farmacocinética , Receptor alfa de Estrogênio/genética , Receptor beta de Estrogênio/genética , Genes Reporter , Humanos , Neoplasias Hepáticas , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Relação Estrutura-Atividade , Transfecção , Trítio
3.
J Reprod Med ; 53(2): 97-101, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18357800

RESUMO

OBJECTIVE: To determine the steady-state exposure of conjugated and unconjugated estrogen components following oral administration of conjugated equine estrogens (2 0.625-mg tablets). STUDY DESIGN: A prospective, open-label, single-treatment study conducted at 1 clinical site with 12 healthy, postmenopausal women. Each subject received 7 daily doses of 2 conjugated equine estrogen (0.625-mg) tablets, and blood samples were taken on the last day of dosing for pharmacokinetic analysis of estrogen components. RESULTS: The major estrogen components after estrogen dosing (as determined by steady-state plasma concentration-time curves) were estrone (100 ng x h/mL), equilin (43.1 ng x h/mL) and delta8,9-dehydroestrone (13.6 ng x h/mL). Several 17beta-reduced forms of estrogen also had consistent plasma concentrations during a steady-state dosing interval. Mean t(max) values ranged from 6.2 to 9.0 hours after dosing, and the 24-hour profiles of the various plasma estrogen concentrations at steady state showed limited fluctuations. CONCLUSION: Oral dosing of conjugated equine estrogen at steady state resulted in consistent concentrations of estrogen components during a dosing interval.


Assuntos
Estrogênios Conjugados (USP)/farmacocinética , Estrogênios/farmacocinética , Pós-Menopausa/efeitos dos fármacos , Administração Oral , Adulto , Idoso , Estrogênios/administração & dosagem , Estrogênios/sangue , Estrogênios Conjugados (USP)/administração & dosagem , Estrogênios Conjugados (USP)/sangue , Feminino , Humanos , Pessoa de Meia-Idade
4.
Menopause ; 25(8): 877-882, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29738412

RESUMO

OBJECTIVE: Response to menopausal hormone therapy (MHT) shows individual variation. SLCO1B1 encodes the OATP1B1 transporter expressed in the liver that transports many endogenous substances, including estrone sulfate, from the blood into hepatocytes. This study evaluated the relationship between genetic variation in SLCO1B1 and response to MHT in women enrolled in the Kronos Early Estrogen Prevention Study (KEEPS) at Mayo Clinic, Rochester, MN. METHODS: KEEPS participants were randomized to oral conjugated equine estrogen (n = 33, oCEE), transdermal 17ß-estradiol (n = 33, tE2), or placebo (n = 34) for 48 months. Menopausal symptoms (hot flashes, night sweats, insomnia, palpitations) were self-reported before treatment and at 48 months. Estrone (E1), E2, and sulfated conjugates (E1S, E2S) were measured using high-performance liquid chromatography-tandem mass spectrometry. SLCO1B1 rs4149056 (c.521T>C, p.Val174Ala) was genotyped using a TaqMan assay. RESULTS: After adjusting for treatment, there was a significant association between the SLCO1B1 rs4149056 TT genotype (encoding normal function transporter) and lower E1S, E1S/E1, and E2S (P = 0.032, 0.010, and 0.008, respectively) compared with women who were heterozygous (TC) or homozygous (CC) for the reduced function allele. The interactions between genotype, treatment, and E2S concentration were stronger in women assigned to tE2 (P = 0.013) than the women taking oCEE (P = 0.056). Among women assigned to active treatment, women with the CT genotype showed a significantly greater decrease in night sweats (P = 0.041) than those with the TT genotype. CONCLUSIONS: Individual variation in sulfated estrogens is explained, in part, by genetic variation in SLCO1B1. Bioavailability of sulfated estrogens may contribute to relief of night sweats.


Assuntos
Estradiol/farmacocinética , Terapia de Reposição de Estrogênios/métodos , Estrogênios Conjugados (USP)/farmacocinética , Transportador 1 de Ânion Orgânico Específico do Fígado/efeitos dos fármacos , Variantes Farmacogenômicos/genética , Disponibilidade Biológica , Estrona/análogos & derivados , Estrona/farmacocinética , Feminino , Genótipo , Fogachos/tratamento farmacológico , Humanos , Pessoa de Meia-Idade , Pós-Menopausa/genética
5.
Endocrinology ; 142(3): 1228-33, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11181539

RESUMO

We have developed a novel osteotropic prodrug of estradiol (E(2)) conjugated with L-Asp-hexapeptide (E(2).3D(6)), which has very low affinity for estrogen receptors, and in this study, we examined its pharmacokinetic behavior and pharmacological potential. After a single iv injection of E(2) x 3D(6) to mice, the half-time for elimination from plasma was about 100 min; however, E(2) was selectively delivered to the bone and eliminated very slowly, declining to the endogenous level at about 7 days. After a single iv injection of E(2), the half-time in plasma was about 70 min, whereas E(2) was highly distributed to the uterus, and the bone concentration of E(2) was only slightly increased at 6 h. When E(2) (0.37 micromol/kg, sc, every third day) or E(2) x 3D(6) (0.11 to 1.1 micromol/kg, sc, every seventh day) was administered to OVX mice for 4 weeks, E(2) increased the bone mineral density (BMD) together with weights of liver and uterus, whereas E(2) x 3D(6) increased only the BMD, in a dose-dependent manner. E(2) x 3D(6) enhanced the expression of messenger RNAs of bone matrix proteins (osteopontin, bone sialoprotein, type I collagen alpha) of OVX mice at 4 h after administration, but E(2) did very slightly. These results indicate that the E(2) prodrug was delivered to the bone, where it gradually released E(2), thereby ameliorating bone loss. This acidic oligopeptide appears to be a good candidate for selective drug delivery to bone.


Assuntos
Osso e Ossos/efeitos dos fármacos , Estradiol/administração & dosagem , Estradiol/farmacologia , Estrogênios Conjugados (USP) , Ovariectomia , Pró-Fármacos , Animais , Ácido Aspártico/análogos & derivados , Matriz Óssea/química , Estradiol/análogos & derivados , Estrogênios Conjugados (USP)/metabolismo , Estrogênios Conjugados (USP)/farmacocinética , Estrogênios Conjugados (USP)/farmacologia , Feminino , Camundongos , Pró-Fármacos/metabolismo , Pró-Fármacos/farmacocinética , Pró-Fármacos/farmacologia , Proteínas/genética , RNA Mensageiro/metabolismo , Receptores de Estrogênio/metabolismo
6.
J Clin Endocrinol Metab ; 78(1): 197-204, 1994 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-8288704

RESUMO

The MCRs of 17 beta-dihydroequilin sulfate and 17 beta-dihydroequilin were determined in normal postmenopausal women by single iv injection of either 17 beta-[3H]dihydroequilin sulfate ([3H]17 beta-EqS) or 17 beta-[3H]dihydroequilin ([3H]17 beta-Eq). After the administration of [3H]17 beta-EqS, blood was drawn at various time intervals, and the plasma obtained was fractionated into the unconjugated, sulfate, and glucuronide fractions. The bulk of radioactivity was present in the sulfate fraction, and from this [3H]17 beta-EqS, [3H]equilin sulfate, [3H]equilenin sulfate, and 17 beta-[3H]dihydroequilenin sulfate were isolated and purified, and their concentrations were measured. The disappearance of [3H]17 beta-EqS from plasma can be described as a function of two exponentials. The half-life of the initial fast component was 5 +/- 0.2 min; this component represents the distribution and transfer from a space, with a mean volume (V1) of 6 +/- 0.5 L. The value for the rate constant (k) of total removal from this space was 300 +/- 20 U/day, of which 35 +/- 2% was irreversible. The mean half-life of the slower component of 17 beta-EqS was 147 +/- 15 min, and the mean MCR was 376 +/- 93 L/day.m2. Similarly, after the administration of [3H]17 beta-Eq, the disappearance of radioactivity as 17 beta-Eq from plasma also had two components. The half-lives of the fast and slow component were 5.5 +/- 0.8 and 45 +/- 2.0 min, respectively. The MCR of 17 beta-Eq was 1252 +/- 103 L/day.m2. From both series of experiments, unconjugated and sulfate-conjugated equilin, equilenin, and 17 beta-dihydroequilenin were isolated and purified, and their concentrations were measured. No 17 alpha-reduced metabolites were detected. These results indicate that 17 beta-EqS is cleared twice as fast as equilin sulfate (MCR, 176 L/day.m2), whereas the more potent estrogen 17 beta-Eq is cleared 2 times slower than equilin. The slower elimination and greater estrogenic activity of 17 beta-Eq support the hypothesis that the major in vivo activity of equilin sulfate present in conjugated equine estrogen preparations is expressed via its metabolites 17 beta-EqS and 17 beta-Eq.


Assuntos
Equilina/análogos & derivados , Pós-Menopausa , Equilina/farmacocinética , Estrogênios Conjugados (USP)/farmacocinética , Feminino , Humanos , Taxa de Depuração Metabólica , Pessoa de Meia-Idade , Valores de Referência
7.
J Steroid Biochem Mol Biol ; 67(1): 33-40, 1998 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9780027

RESUMO

The possible role of fetal membrane deconjugating activity in the movement of a charged steroid conjugate between fetal and maternal compartments was investigated. The ability of amnion and chorion laeve to transfer [3H]estrone-[35S]sulfate was assessed in both orientations of guinea pig tissue at 45 days and near parturition. While early amnion was impermeable, late tissue transferred approximately 50% (w/w) of the substrate in a bidirectional process that was non-saturable and independent of either deconjugation or ATP. Transfer across early chorion was similar to late amnion. Saturation curves from each tissue were superimposable, as were those of the time course. Transfer across both early and late chorion proceeded in the absence of deconjugation, with no effect of tissue orientation or ATP depletion. However, late chorion exhibited a decrease in estrone-sulfate transfer, as verified by concentration dependency and time course analyses, though transport across the tissue remained non-saturable. The results in amnion were congruous with the presence and absence of tight junctions in the epithelium of early and late tissue, respectively. However, sulfoconjugate transfer across early chorion proceeded in the presence of a paracellular barrier, suggesting specialized regulation of the transport process which extended late into gestation.


Assuntos
Âmnio/fisiologia , Córion/fisiologia , Estrona/análogos & derivados , Animais , Transporte Biológico , Estrogênios Conjugados (USP)/farmacocinética , Estrona/farmacocinética , Feminino , Idade Gestacional , Cobaias , Técnicas In Vitro , Cinética , Permeabilidade , Gravidez , Radioisótopos de Enxofre , Trítio
8.
J Steroid Biochem Mol Biol ; 67(2): 119-31, 1998 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9877212

RESUMO

Recently a tenth equine estrogen, identified as the sulfate ester of delta8-estrone has been reported to be present in Premarin (a conjugated equine estrogen preparation), and because of its unique ring B unsaturated structure (conjugated double bond in the B ring), we have, in the present study, determined its pharmacokinetics in postmenopausal women and men, its interaction with uterine estrogen receptors and its uterotropic activity. After the administration of [14C]delta8-estrone, blood was drawn at various time intervals, and the plasma fractionated into the unconjugated sulfate and glucuronide fractions. The disappearance of radioactivity as delta8-estrone from plasma can be described as a function of two exponentials. The half-lives of the first and second components were 5+/-0.2 and 40.4 min, respectively. The mean metabolic clearance rate calculated (MCR), was 1711+/-252 l/d m2. From the unconjugated fraction, delta8-17beta-estradiol was also isolated and identified. From the sulfate conjugated fraction, delta8-estrone sulfate and delta8-17beta-estradiol sulfate were isolated in almost equal amounts. No other metabolites of delta8-estrone was detectable in the plasma. Both delta8-estrone and delta8-17beta-estradiol bind with human endometrial and rat uterine estrogen receptors with high affinity. The binding affinities of delta8-17beta-estradiol for human endometrial and rat uterine cytoplasmic receptors were 4 and 25 times higher than those of the parent estrogen delta8-estrone, respectively. Administration of delta8-estrone and delta8-17beta-estradiol (2 microg/100 g body weight) to immature rats significantly (P< 0.05) increased the uterine weight compared to the controls. These data demonstrate that delta8-estrone has estrogenic activity, and that it is further metabolized in man to a single more potent estrogen, delta8-17beta-estradiol. The extent of this activation by 17beta-reduction appears to be greater than that observed with other estrogens. Both estrogens circulate as sulfate conjugates and are very slowly eliminated from the circulation. These data further suggest that delta8-estrone and its major metabolite delta8-17beta-estradiol can contribute to the overall in vivo biological effects of Premarin.


Assuntos
Estradiol/farmacocinética , Estrogênios Conjugados (USP)/farmacocinética , Estrona/análogos & derivados , Pós-Menopausa , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Estradiol/análogos & derivados , Estrona/farmacocinética , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Ratos , Ratos Sprague-Dawley
9.
J Steroid Biochem Mol Biol ; 55(2): 271-8, 1995 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-7495708

RESUMO

The metabolism of equilin sulfate was determined in female dogs receiving 2.5 mg/kg of [3H]equilin sulfate alone or in a preparation that contained all the components that are present in the conjugated equine estrogen product Premarin. The pharmacokinetic parameters of total radioactivity indicated that the drug is rapidly absorbed and it has a moderate half-life in plasma. The total radioactivity in plasma following administration of [3H]equilin sulfate as part of a mixture of conjugated equine estrogens had significantly lower peak concentration (Cmax), a lower area under the curve (AUC), a longer terminal half-life (t1/2) and a longer mean residence time (MRT) than when [3H]equilin sulfate was given alone, indicating that the other components in the conjugated equine estrogen preparation altered the pharmacokinetics of equilin sulfate. An average of 26.7 +/- 4.4% of the administered radioactive dose was excreted in urine of dogs receiving [3H]equilin sulfate. Again, a significantly lower percentage (21.4 +/- 6.3%, P = 0.023) was eliminated in urine of dogs receiving [3H]equilin sulfate in the conjugated equine estrogen preparation, indicating that the absorption of equilin sulfate was perhaps altered by the other components in the conjugated equine estrogen preparation. Metabolite profiles of plasma and urine were similar. Equilin, equilenin, 17 beta-dihydroequilenin, 17 beta-dihydroequilin, 17 alpha-dihydroequilenin and 17 alpha-dihydroequilin were present in both matrices. 17 beta-Dihydroequilin and equilin were the two major chromatographic peaks in plasma samples. 17 beta-Dihydroequilenin and 17 beta-dihydroequilin were the major metabolites in urine. In conclusion, following oral administration of [3H]equilin sulfate to dogs, the radioactivity is rapidly absorbed. The disposition of equilin sulfate is altered by the other components that are present in the conjugated equine estrogen preparation Premarin. The reduction of the 17-keto group and aromatization of ring-B are the major metabolic pathways of equilin in the dog.


Assuntos
Equilina/análogos & derivados , Estrogênios Conjugados (USP)/metabolismo , Animais , Biotransformação , Cromatografia Líquida de Alta Pressão , Cães , Combinação de Medicamentos , Equilina/sangue , Equilina/metabolismo , Equilina/farmacocinética , Estrogênios Conjugados (USP)/sangue , Estrogênios Conjugados (USP)/farmacocinética , Estrogênios Conjugados (USP)/farmacologia , Feminino , Cromatografia Gasosa-Espectrometria de Massas , Meia-Vida , Taxa de Depuração Metabólica , Técnica de Diluição de Radioisótopos , Trítio
10.
J Steroid Biochem Mol Biol ; 85(2-5): 473-82, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12943738

RESUMO

Menopause marks the start of a new phase in a woman's life that is associated with a decrease in circulating estrogen levels. Although the average age of women has increased from 50 to nearly 85 years, the average age at menopause has remained essentially constant at 50 years. Thus, women now spend nearly a third of their lives in an estrogen deficient state. This normal aging process in women is associated with increasing health problems such as osteoporosis, cardiovascular disease, neurodegenerative diseases, and cancer. Estrogen replacement therapy (ERT) has been shown to play an important beneficial role in the health and well being of postmenopausal women. Several estrogen preparations are available and among these conjugated equine estrogens (CEE) are most frequently used. The drug CEE, is a complex natural urinary extract of pregnant mare's urine and contains at least 10 estrogens in their sulfate ester form and these are the ring B saturated estrogens: estrone (E(1)), 17beta-estradiol (17beta-E(2)), 17alpha-estradiol (17alpha-E(2)), and the ring B unsaturated estrogens equilin (Eq), 17beta-dihydroequilin (17beta-Eq), 17alpha-dihydroequilin (17alpha-Eq), equilenin (Eqn), 17beta-dihydroequilenin (17beta-Eqn), 17alpha-dihydroequilenin (17alpha-Eqn), and Delta(8)-estrone (Delta(8)-E(1)). All of these estrogens in their unconjugated form are biologically active and can interact with recombinant human estrogen receptor alpha (ERalpha) and beta (ERbeta) with 17beta-estradiol and 17beta-dihydroequilin having the highest affinity for both receptors. A number of the ring B unsaturated estrogens had nearly twofold higher affinity for the ERbeta. The pharmacokinetics of these estrogens in postmenopausal women indicate that the unconjugated estrogens compared to their sulfated forms are cleared more rapidly. The 17-keto estrogens are metabolized to the more potent 17beta-reduced products which are cleared at a slower rate. In postmenopausal women, the extent of 17beta-activation is much higher with the ring B unsaturated estrogens than with ring B saturated estrogens. Oxidized LDL and oxidative stress are thought to contribute to both atherosclerosis and neurodegenerative disorders. Neurons in particular are at a high risk from damage resulting from oxidative stress. In vivo and in vitro studies indicate that the oxidation of LDL isolated from postmenopausal women was inhibited differently by various estrogens and other antioxidants. The unique ring B unsaturated estrogens were the most potent while the red wine component t-resveratrol was the least potent. Studies were designed to explore the cellular and molecular mechanisms that may be involved in the neuroprotective effects of CEE components. The data indicate that the neurotoxic effects of oxidized LDL and glutamate can be inhibited by various estrogens, with the ring B unsaturated estrogens being the most active. These effects are involved in the inhibition of DNA fragmentation and up-regulation of anti-apoptotic protein Bcl-2 and down-regulation of pro-apoptotic protein Bax. These combined data suggest that some of the neuroprotective benefits associated with long-term estrogen therapy may occur by the above mechanism(s). Because estrogens such as the Delta(8)-estrogens are relatively less feminizing than the classical estrogen 17beta-estradiol, they may be important in the development of more neuro-specific estrogens that will be useful in the prevention of neurodegenerative diseases, such as Alzheimer's and Parkinson disease, in both men and women.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Estrogênios Conjugados (USP)/uso terapêutico , Menopausa/fisiologia , Doenças Neurodegenerativas/prevenção & controle , Doença de Alzheimer/prevenção & controle , Animais , Estrogênios Conjugados (USP)/farmacocinética , Feminino , Humanos , Menopausa/efeitos dos fármacos , Taxa de Depuração Metabólica , Doenças Neurodegenerativas/tratamento farmacológico
11.
Obstet Gynecol ; 89(3): 340-5, 1997 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9052581

RESUMO

OBJECTIVE: To investigate the pharmacokinetic profiles of different doses of micronized 17 beta-estradiol administered by oral or sublingual routes. METHODS: Single doses of micronized 17 beta-estradiol were administered orally (1 mg, 0.5 mg) or sublingually (1 mg, 0.5 mg, 0.25 mg) to six postmenopausal women in a randomized clinical trial. We calculated pharmacokinetic parameters for estradiol (E2) and estrone (E1) of maximum serum concentration, time to maximum serum concentration, terminal half-life, area under the concentration curve, and oral clearance. Serum levels of E1 sulfate also were compared at 4, 12, and 24 hours after dosing. RESULTS: Sublingual administration resulted in rapid absorption with significantly higher E2 levels than did comparable oral dosing. Estrone levels did not vary with route of administration but correlated with the dosage administered. Estrone sulfate levels correlated with the dosage administered and also tended to be higher with sublingual administration. Sublingual administration resulted in a significantly lower E1 to E2 ratio during the 24 hours than did oral administration. CONCLUSION: Sublingual administration of micronized 17 beta-estradiol results in a rapid, burst-like absorption into the systemic circulation, yielding high E2 levels that fall rapidly over the first 6 hours.


Assuntos
Estradiol/administração & dosagem , Estradiol/farmacocinética , Estrogênios Conjugados (USP)/administração & dosagem , Estrogênios Conjugados (USP)/farmacocinética , Estrona/análogos & derivados , Estrona/administração & dosagem , Estrona/farmacocinética , Administração Oral , Administração Sublingual , Idoso , Feminino , Humanos , Pessoa de Meia-Idade , Fatores de Tempo
12.
Obstet Gynecol ; 84(2): 215-8, 1994 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-8041532

RESUMO

OBJECTIVE: To test the hypothesis tha a very-low-dose regimen of vaginal estrogen would provide effective relief from atrophic vaginitis without endometrial proliferation. METHODS: Twenty postmenopausal women with symptoms, signs, and cytologic evidence of atrophic vaginitis were enrolled. Each subject was treated with 0.3 mg of conjugated estrogens, administered vaginally 3 nights per week for 6 months. We examined the following outcomes: symptoms, vaginal cellular (cytologic) maturity, endometrial histology, sonographic evaluation of endometrial thickness, Doppler measures of uterine artery blood flow, and serum levels of estrone and estradiol. Pre- and post-treatment data were compared for each subject. RESULTS: Satisfactory relief of symptoms occurred in 19 of 20 cases. Vaginal cellular maturation improved significantly with therapy (P < .01). There were no significant changes in endometrial thickness, uterine artery blood flow, or serum estrogen levels. Endometrial proliferation was observed in one case. CONCLUSIONS: Relief from atrophic vaginitis can be achieved with 0.3 mg of conjugated estrogens administered vaginally three times per week. Endometrial proliferation may occur at this low dose, albeit rarely.


Assuntos
Endométrio/efeitos dos fármacos , Estrogênios Conjugados (USP)/administração & dosagem , Vaginite/tratamento farmacológico , Administração Intravaginal , Idoso , Atrofia , Biópsia , Velocidade do Fluxo Sanguíneo/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Endométrio/irrigação sanguínea , Endométrio/patologia , Endométrio/fisiopatologia , Estradiol/sangue , Estrogênios Conjugados (USP)/farmacocinética , Estrona/sangue , Feminino , Humanos , Pessoa de Meia-Idade , Pós-Menopausa , Reologia , Fatores de Tempo , Vaginite/sangue , Vaginite/patologia , Vaginite/fisiopatologia
13.
J Clin Pharmacol ; 42(3): 332-41, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11865971

RESUMO

A multiple-dose, placebo-controlled, randomized pharmacokinetic study was performed in 15 early (i.e., 1-3 years) postmenopausal women to evaluate the single and steady-state pharmacokinetics of 0.625 mg Cenestin (Synthetic Conjugated Estrogens, A) tablets, administered once daily for 90 days. Plasma concentration-time profiles for both total (conjugated and unconjugated) estrone and equilin, two major estrogens in Cenestin, were nearly superimposable between Day 1 (single dose) and Day 90 (multiple dose), indicating equivalent drug exposure from one dose to the next. For total estrone, the mean estimates of Cmax and AUC0-24 were 2.5 ng/ml and 35.0 ng x h/ml for Day 1 and 3.0 ng/ml and 39.8 ng x h/ml for Day 90, respectively. Similarly, Cmax and AUC0-24 mean values for total equilin were 1.4 ng/ml and 17.4 ng x h/ml after Day 1 and 1.5 ng/ml and 17.3 ng x h/ml after Day 90, respectively. The mean tmax value for total estrone was 8.3 hours on Day 1 and 8.6 hours on Day 90, indicating a slower rate of absorption. The average estimate for observed drug accumulation index for the 24-hour dosing interval was calculated to be 1.1 for total estrone and 1.0 for total equilin. These data, taken together, indicate a slow and steady rate of absorption, minimal drug accumulation at steady state, and consistent drug exposure between Cenestin doses.


Assuntos
Congêneres do Estradiol/administração & dosagem , Congêneres do Estradiol/farmacocinética , Terapia de Reposição de Estrogênios , Estrogênios Conjugados (USP)/administração & dosagem , Estrogênios Conjugados (USP)/farmacocinética , Adulto , Idoso , Área Sob a Curva , Preparações de Ação Retardada/administração & dosagem , Preparações de Ação Retardada/farmacocinética , Método Duplo-Cego , Equilina/sangue , Estradiol/sangue , Congêneres do Estradiol/sangue , Terapia de Reposição de Estrogênios/estatística & dados numéricos , Estrogênios Conjugados (USP)/sangue , Estrona/sangue , Feminino , Humanos , Pessoa de Meia-Idade , Pós-Menopausa/sangue , Comprimidos
14.
J Soc Gynecol Investig ; 7(3): 175-83, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10865186

RESUMO

OBJECTIVE: To compare the pharmacokinetics and relative bioavailabilities of key estrogen components of Premarin (Wyeth-Ayerst, Canada) with those of a generic conjugated estrogen preparation, C.E.S. (synthetic mixture of estrogens; ICN, Montreal, Canada) in healthy postmenopausal women. METHODS: We conducted a randomized, single-dose, two-treatment, three-period crossover study in 41 postmenopausal women. After an oral dose (2 x 0.625 mg) of Premarin or C.E.S., plasma concentrations of unconjugated and total estrone (E(1)), equilin (Eq), 17beta-estradiol (17beta-E(2)), 17beta-dihydroequilin (17beta-Eq), Delta(8)-esterone (Delta(8)-E(1)) and Delta(8),17beta-estradiol (Delta(8),17beta-E(2)) were measured over 72 hours using gas chromatography and mass spectroscopy. RESULTS: After administration of C.E.S., E(1), Eq, and 17beta-Eq appeared in blood at a significantly faster rate (lower t(max)) than after Premarin. The rapid appearance of estrogens after C.E.S. was associated with significantly higher (14-61%) C(max) values. In contrast to the high C(max) values, the area under the curve (AUC)(infinity) of unconjugated and total Eq, and 17beta-Eq were significantly lower after C.E.S., whereas those of E(1) were significantly higher. Although, the t(max) values for 17beta-E(2) were lower and the C(max) values higher after C.E.S., only the C(max) of unconjugated 17beta-E(2) was significantly different after Premarin. Unconjugated and total Delta(8)-E(1) and its main metabolite, Delta(8),17beta-E(2), were detectable in plasma only after administration of Premarin. The geometric mean ratio (GMR) (C. E.S./Premarin) of bioavailability parameters indicated that all C(max) and t(max) values for the unconjugated and total E(1), Eq, 17beta-E(2), and 17beta-Eq fell outside the regulatory requirement that the 90% confidence intervals of GMRs of two products be within 80% and 125%. Similarly, with the exception of total E(1) and total Eq, none of the AUC(t) or AUC(alpha) of the remaining estrogens meets the required regulatory standards of bioequivalence. CONCLUSIONS: C.E.S. is not bioequivalent to Premarin. Because C.E.S. also is not pharmaceutically equivalent to Premarin, it cannot be assumed to be therapeutically equivalent. Until long-term clinical trials with C.E.S. demonstrate its efficacy, extrapolation of the long-term benefits described for Premarin to C.E.S. would be risky and questionable.


Assuntos
Estrogênios Conjugados (USP)/farmacocinética , Pós-Menopausa , Adulto , Estudos Cross-Over , Equilina/análogos & derivados , Equilina/sangue , Estradiol/sangue , Terapia de Reposição de Estrogênios , Estrona/sangue , Feminino , Humanos , Pessoa de Meia-Idade , Equivalência Terapêutica
15.
Steroids ; 59(4): 270-3, 1994 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-8079382

RESUMO

In order to study the metabolism of estrone sulfate in the adult female rhesus monkey, we gave four monkeys a pulse injection of [3H]estrone sulfate and obtained blood samples at 15, 30, 60, 120, 300, and 1,440 minutes after the pulse. All blood samples were analyzed for [3H]estrone sulfate. The mean +/- SE for the initial volume of distribution was 4.6 +/- 0.9 L. The mean metabolic clearance rate (MCR) was 42.0 +/- 2.9 L/day. In addition, 2 monkeys were infused for 240 minutes with [3H]estrone sulfate and 5 monkeys were infused for 240 minutes with [3H]estrone sulfate/[14C]estrone. Three blood samples were obtained during the last hour of the infusions and analyzed for radioactivity as estrone sulfate, estrone, and estradiol, and MCRs, conversion ratios, and [rho]BB values were calculated. The mean +/- SE for the MCR was 67.5 +/- 8.3 L/day, and for the conversion ratio, estradiol to estrone sulfate was 0.054 +/- 0.016. The mean [rho]BB value for estrone sulfate conversion to estrone was 43.6 +/- 3.4% and for estrone conversion to estrone sulfate was 33.5 +/- 6.6%. Thus estrone sulfate is cleared slowly and is converted to both estrone and estradiol. The hydrolysis of estrone sulfate to estrone is not significantly different than the conversion of estrone to estrone sulfate.


Assuntos
Estrogênios Conjugados (USP)/farmacocinética , Estrona/análogos & derivados , Animais , Estrona/farmacocinética , Feminino , Humanos , Macaca mulatta , Taxa de Depuração Metabólica
16.
Maturitas ; 12(3): 171-97, 1990 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-2170822

RESUMO

There are large inter- and intra-individual variations in the serum concentrations of natural and synthetic sex steroids irrespective of the route of administration. Oral ingestion of steroids has a stronger effect on hepatic metabolism than parenteral administration, as the local concentration in liver sinusoids are 4-5 times higher during the first liver passage. Oestradiol and oestrone are interconvertible, dependent on the local concentrations in liver and target organs, and oestrone sulphate serves as a large reservoir. The oestrone/oestradiol ratio has no physiological significance, as oestrone is only a weak oestrogen. Oestrone is both a precursor and a metabolite of oestradiol. Oestriol is extensively conjugated after oral administration. Therefore, the oestriol serum levels are similar after oral intake of 10 mg and after vaginal application of 0.5 mg oestriol resulting in similar systemic effectiveness. Conjugated oestrogens can easily enter the hepatocytes but are hormonally active only after hydrolyzation into the parent steroids. Ethinylestradiol which exerts strong effects on hepatic metabolism and inhibits metabolizing enzymes, should not be used for hormone replacement therapy. Among the progestogens, the progesterone derivatives have less effects on liver metabolism than the norethisterone derivatives (13-methyl-gonanes and 13-ethyl-gonanes). The highly potent 13-ethyl-gonanes are effective at very low doses, because of a slow inactivation and elimination rate due to the ethinyl group.


Assuntos
Estrogênios/farmacocinética , Congêneres da Progesterona/farmacocinética , Estrogênios/química , Estrogênios/farmacologia , Estrogênios Conjugados (USP)/química , Estrogênios Conjugados (USP)/farmacocinética , Estrogênios Conjugados (USP)/farmacologia , Feminino , Humanos , Progesterona/metabolismo , Congêneres da Progesterona/química , Congêneres da Progesterona/farmacologia
17.
J Pharm Pharmacol ; 50(8): 857-64, 1998 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9751449

RESUMO

The pharmacokinetics and enterohepatic cycling of oestradiol have been studied after three oral, single-dose administrations of equimolar doses of oestradiol alone, oestradiol plus desogestrel and oestradiol valerate, in a 3-way cross-over mode in 18 healthy postmenopausal women. Oestradiol was readily absorbed and metabolized to oestrone, which reached much higher serum concentrations (140pgmL(-1)) than its parent compound (35pgmL(-1)). All three formulations had the same kinetic profile and were bioequivalent on testing. Noticeable first and second absorption phases were apparent from the oestradiol and oestrone serum concentration-time curves for all oestradiol formulations. The mean serum concentration-time curves of the metabolite oestrone (corrected for endogenous oestrone) showed a second maximum at approximately 25h. By means of line feathering, serum concentration-time curves were constructed which belonged to the first, second and third phases of absorption. The maximum serum concentration, Cmax, of the second absorption or recirculation of oestrone was 20% that of the first, and the Cmax of the third circulation was 50% that of the second. The areas under the serum-concentration-time curves (AUC) for the second and third recirculations were similar-each comprised 12-13% of the total AUC. The oral clearance values of the recirculations were constant (590Lh(-1)). Enterohepatic recirculation of endogenous compounds is aimed at maintaining a steady-state serum concentration for immediate use and hydrolysis in the target organs. It is concluded that exogenously added oestradiol and its metabolites follow the recirculation pathways of the endogenous oestrogen pool.


Assuntos
Circulação Êntero-Hepática , Estradiol/farmacocinética , Pós-Menopausa , Idoso , Estudos Cross-Over , Desogestrel/farmacocinética , Combinação de Medicamentos , Estradiol/administração & dosagem , Estradiol/análogos & derivados , Estradiol/metabolismo , Terapia de Reposição de Estrogênios , Estrogênios/farmacocinética , Estrogênios Conjugados (USP)/farmacocinética , Estrogênios Esterificados (USP) , Estrona/metabolismo , Feminino , Humanos , Pessoa de Meia-Idade , Proteínas da Gravidez/farmacocinética , Congêneres da Progesterona/farmacocinética , Valores de Referência
18.
Ceska Gynekol ; 68(2): 117-21, 2003 Mar.
Artigo em Tcheco | MEDLINE | ID: mdl-12749182

RESUMO

OBJECTIVE: To verify if and to which extent the interaction with grapefruit juice can increase bioavailability of orally administered sexual steroids. DESIGN: Pilot pharmacokinetics study. SETTING: Department of Obstetrics and Gynecology and Institute of Pharmacology, Medical Faculty, Palacký University, Olomouc; Department of Nuclear Medicine, University Hospital, Olomouc. METHODS: 2 mg of estradiol valerate and 100 mg of micronized progesterone were given to eight healthy postmenopausal volunteers. Blood samples were collected at time 0, 2, 3, 5 and 24 hours after tablets application. The same trial was repeated a week later but tablets were swallowed with 200 ml of grapefruit juice. Serum levels of estradiol and progesterone were measured by RIA. Results were statistically evaluated using the Wilcoxon's nonparametric paired test. RESULTS: Though grapefruit juice on average slightly increased serum levels of estradiol (E2) and progesterone, this increase reached statistical significance only for the E2 level 24 hours after application of tablets. The mean area under curve (AUC) of estradiol rose significantly to 117%. The even greater increase in the mean AUC of progesterone (to 125%) was not statistically significant because of marked individual variability of response. CONCLUSIONS: Our results suggest that grapefruit juice may increase bioavailability of orally administered estradiol and progesterone. The response varies markedly between individuals. This observation may be of some importance also for users of OC and HRT.


Assuntos
Bebidas , Estradiol/análogos & derivados , Estradiol/farmacocinética , Estrogênios Conjugados (USP)/farmacocinética , Interações Alimento-Droga , Progesterona/farmacocinética , Administração Oral , Disponibilidade Biológica , Citrus paradisi , Estradiol/administração & dosagem , Estrogênios Conjugados (USP)/administração & dosagem , Feminino , Humanos , Pessoa de Meia-Idade , Progesterona/administração & dosagem
19.
Rinsho Byori ; 48(11): 1059-63, 2000 Nov.
Artigo em Japonês | MEDLINE | ID: mdl-11132560

RESUMO

To investigate problems associated with the measurement of estradiol 17 beta(E2) in hormone replacement therapy(HRT), five commercial immunoassay methods(Coat-A-Count E2 as the conventional method; Immulyze E2, Immuno 1 E2, Vitros E2, and HRT-E2 as the comparative methods) were used to assay E2 concentrations. Samples were obtained from 21 women who had been receiving HRT, 99 nonmedicated women, and 10 healthy men volunteers. No significant difference between the Coat-A-Count E2 and the comparative method was observed in the nonmedicated women. However, we found that the serum E2 concentration from patients taking Premarin showed a large discrepancy between the Coat-A-Count E2 method, which showed considerably higher values, and the other four methods. The reason for our conflicting results from patients with HRT was probably because the Coat-A-Count E2 detected circulating estrogen conjugates. The experimental addition of Premarin for the in vitro cross-reactivity was done. The cross-reactivity was low because a similar E2 steroid exists independently. However, the E2 serum value of the ten male volunteers after taking Premarin was elevated. The reason for this result was due to the high cross-reactivity between anti E2 polyclonal antibody and the various metabolic products of Premarin. In conclusion, the influence of Premarin should be taken into consideration when measuring estradiol concentration to monitor HRT.


Assuntos
Estradiol/sangue , Terapia de Reposição de Estrogênios , Estrogênios Conjugados (USP)/farmacocinética , Radioimunoensaio/métodos , Biomarcadores/sangue , Reações Cruzadas , Estrogênios Conjugados (USP)/administração & dosagem , Feminino , Humanos , Masculino , Monitorização Fisiológica/métodos , Kit de Reagentes para Diagnóstico , Sensibilidade e Especificidade
20.
J Steroid Biochem Mol Biol ; 142: 16-29, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24176763

RESUMO

Oral conjugated equine estrogens (CEE) are the most used estrogen formulation for postmenopausal hormone therapy either alone or in combination with a progestin. CEE is most commonly used for the management of early menopausal symptoms such as hot flashes, vaginitis, insomnia, and mood disturbances. Additionally, if used at the start of the menopausal phase (age 50-59 years), CEE prevents osteoporosis and may in some women reduce the risk of cardiovascular disease (CVD) and Alzheimer's disease (AD). There appears to be a common mechanism through which estrogens can protect against CVD and AD. CEE is a natural formulation of an extract prepared from pregnant mares' urine. The product monogram lists the presence of only 10 estrogens consisting of the classical estrogens, estrone and 17ß-estradiol, and a group of unique ring B unsaturated estrogens such as equilin and equilenin. The ring B unsaturated estrogens are formed by an alternate steroidogenic pathway in which cholesterol is not an obligatory intermediate. Both the route of administration and structure of these estrogens play a role in the overall pharmacology of CEE. In contrast to 17ß-estradiol, ring B unsaturated estrogens express their biological effects mainly mediated by the estrogen receptor ß and not the estrogen receptor α. All estrogen components of CEE are antioxidants, and some ring B unsaturated estrogens have several fold greater antioxidant activity than estrone and 17ß-estradiol. The cardioprotective and neuroprotective effects of CEE appear to be, to some extent, due to its ability to prevent the formation of oxidized LDL and HDL, and by inhibiting or modulating some of the key proteases involved in programmed cell death (apoptosis) induced by the excess neurotransmitter glutamate and other neurotoxins. Selective combinations of ring B unsaturated estrogens have the potential of being developed as novel therapeutic agents for the prevention of cardiovascular disease and Alzheimer's disease in both aging women and men. This article is part of a Special Issue entitled 'Menopause'.


Assuntos
Estrogênios Conjugados (USP)/uso terapêutico , Doença de Alzheimer/prevenção & controle , Antioxidantes/uso terapêutico , Doenças Cardiovasculares/prevenção & controle , Estrogênios Conjugados (USP)/farmacocinética , Feminino , Humanos , Masculino , Osteoporose Pós-Menopausa/prevenção & controle
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA