Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Clin Pharmacokinet ; 61(11): 1571-1583, 2022 11.
Article in English | MEDLINE | ID: mdl-36173592

ABSTRACT

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.


Subject(s)
Estrogens, Conjugated (USP) , Postmenopause , Animals , Female , Humans , China , Chromatography, Liquid/methods , Estrogens/metabolism , Estrogens, Conjugated (USP)/pharmacokinetics , Horses , Tandem Mass Spectrometry/methods
2.
Menopause ; 25(8): 877-882, 2018 08.
Article in English | MEDLINE | ID: mdl-29738412

ABSTRACT

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.


Subject(s)
Estradiol/pharmacokinetics , Estrogen Replacement Therapy/methods , Estrogens, Conjugated (USP)/pharmacokinetics , Liver-Specific Organic Anion Transporter 1/drug effects , Pharmacogenomic Variants/genetics , Biological Availability , Estrone/analogs & derivatives , Estrone/pharmacokinetics , Female , Genotype , Hot Flashes/drug therapy , Humans , Middle Aged , Postmenopause/genetics
3.
J Steroid Biochem Mol Biol ; 142: 16-29, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24176763

ABSTRACT

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'.


Subject(s)
Estrogens, Conjugated (USP)/therapeutic use , Alzheimer Disease/prevention & control , Antioxidants/therapeutic use , Cardiovascular Diseases/prevention & control , Estrogens, Conjugated (USP)/pharmacokinetics , Female , Humans , Male , Osteoporosis, Postmenopausal/prevention & control
4.
Fertil Steril ; 94(6): 2365-8, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20466363

ABSTRACT

In this open-label, randomized, multiple-dose, two-treatment crossover study, 24 postmenopausal women with moderate to severe atrophic vaginitis received 0.3 mg conjugated estrogens daily for 14 days: 7 days orally (0.3 mg tablet) and 7 days vaginally (0.5 g cream). Steady-state plasma concentrations of E2 and estrone were one-third lower after vaginal versus oral administration of conjugated estrogens.


Subject(s)
Estradiol/blood , Estrogens, Conjugated (USP)/administration & dosage , Vagina/pathology , Vaginitis/drug therapy , Administration, Intravaginal , Administration, Oral , Aged , Atrophy/blood , Atrophy/drug therapy , Atrophy/metabolism , Cross-Over Studies , Drug Administration Schedule , Estrogens, Conjugated (USP)/blood , Estrogens, Conjugated (USP)/pharmacokinetics , Estrone/blood , Female , Humans , Middle Aged , Osmolar Concentration , Vagina/drug effects , Vaginal Creams, Foams, and Jellies , Vaginitis/blood , Vaginitis/metabolism
5.
Eur J Pharm Sci ; 37(3-4): 378-86, 2009 Jun 28.
Article in English | MEDLINE | ID: mdl-19491029

ABSTRACT

Carriers may mediate the permeation across enterocytes for drug substances being organic anions. Carrier mediated permeation for the organic anions estrone-3-sulfate (ES) and glipizide across Caco-2 cells were investigated kinetically, and interactions on involved carriers evaluated. Initial uptakes (P(UP)) at apical and basolateral membranes, apparent permeabilities (P(APP)) and corresponding intracellular end-point accumulations (P(EPA)) of radioactive labeled compounds were studied. Possible effects of other anionic compounds were investigated. Apical P(UP) and absorptive P(APP) for ES were inhibited and its absorptive P(EPA) prevented in presence of the investigated organic anions and apical P(UP) was saturable with K(m) 23microM. Basolateral P(UP) and exsorptive P(APP) were inhibited, its exsorptive P(EPA) was prevented, and basolateral P(UP) and exsorptive P(APP) were saturable with K(m) 44microM and 38microM, respectively. BCRP inhibition affected both absorptive an exsorptive P(EPA) and P(APP) for ES. Glipizide apical P(UP) and absorptive P(APP) were not inhibitable. Basolateral P(UP) for glipizide was inhibitable, its P(EPA) prevented, and P(UP) was saturable with K(m) 56microM, but exsorptive P(APP) was not affected. Carrier mediated exsorption kinetics for ES are seen at both apical and basolateral membranes, resulting in predominant exsorption despite presence of absorptive carrier(s). Carrier mediated basolateral P(UP) for glipizide was observed, but glipizide P(APP) was not described by carrier kinetics. However, glipizide is affecting exsorption for ES, due to interactions on basolateral carrier. The study confirms that estrone-3-sulfate can be used to characterize anionic carrier kinetics. Furthermore it is suggested that estrone-3-sulfate may be used to identify compounds which may interact on anionic carriers.


Subject(s)
Estrogens, Conjugated (USP)/pharmacokinetics , Estrone/analogs & derivatives , Glipizide/pharmacokinetics , Hypoglycemic Agents/pharmacokinetics , Absorption , Administration, Oral , Caco-2 Cells , Cell Membrane Permeability , Estrogens, Conjugated (USP)/administration & dosage , Estrone/administration & dosage , Estrone/pharmacokinetics , Glipizide/administration & dosage , Humans , Hydrogen-Ion Concentration , Hypoglycemic Agents/administration & dosage
6.
Menopause ; 16(1): 30-6, 2009.
Article in English | MEDLINE | ID: mdl-18820592

ABSTRACT

OBJECTIVE: Approximately 50% of postmenopausal women suffer from vaginal atrophy, and a large proportion of them choose intravaginal estrogen preparations administered for local action to avoid systemic exposure to estrogens and its associated risk of breast and uterine cancer. The primary objective of this study was the evaluation of the systematic bioavailability of estradiol and estrone and the pharmacokinetics of two of the most frequently used intravaginal estrogen preparations, namely Vagifem and Premarin cream. DESIGN: While immunobased assays could not previously provide accurate measurement of serum estrogen concentrations in postmenopausal women, we have used validated mass spectrometry assays to measure the pharmacokinetics of serum estradiol and estrone during the 24 hours following the seventh daily application of 25 microg estradiol (Vagifem) and 1 g (0.625 mg) conjugated estrogens (Premarin) cream in 10 postmenopausal women in each group. RESULTS: Serum estradiol was increased on average by 5.4-fold from 3 to 17 pg/mL during the 24-hour period after daily administration of 25 microg estradiol or 1 g (0.625 mg) conjugated estrogens cream. Serum estrone, conversely, increased 150% with Vagifem and 500% with Premarin cream. CONCLUSIONS: The present data using validated, accurate, and sensitive mass spectrometry assays of estrogens show that the Vagifem pill and Premarin cream, after 1 week of daily treatment, cause an approximately fivefold increase in serum estradiol in postmenopausal women, thus indicating that the effects are unlikely to be limited to the vagina and that systemic actions are expected after application of these intravaginal estrogen preparations.


Subject(s)
Estrogens/administration & dosage , Estrogens/blood , Postmenopause , Administration, Intravaginal , Adult , Aged , Estradiol/administration & dosage , Estradiol/blood , Estradiol/pharmacokinetics , Estrogens/pharmacokinetics , Estrogens, Conjugated (USP)/administration & dosage , Estrogens, Conjugated (USP)/blood , Estrogens, Conjugated (USP)/pharmacokinetics , Estrone/blood , Female , Gas Chromatography-Mass Spectrometry , Humans , Middle Aged , Prospective Studies , Sensitivity and Specificity
7.
Endocrinology ; 149(10): 4857-70, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18599548

ABSTRACT

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.


Subject(s)
Estrogen Receptor alpha/chemistry , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/chemistry , Estrogen Receptor beta/metabolism , Estrogens, Conjugated (USP)/chemistry , Estrogens, Conjugated (USP)/pharmacokinetics , Carcinoma, Hepatocellular , Cell Line, Tumor , Estradiol/chemistry , Estradiol/pharmacokinetics , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/genetics , Genes, Reporter , Humans , Liver Neoplasms , Protein Structure, Quaternary , Protein Structure, Tertiary , Structure-Activity Relationship , Transfection , Tritium
8.
J Reprod Med ; 53(2): 97-101, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18357800

ABSTRACT

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.


Subject(s)
Estrogens, Conjugated (USP)/pharmacokinetics , Estrogens/pharmacokinetics , Postmenopause/drug effects , Administration, Oral , Adult , Aged , Estrogens/administration & dosage , Estrogens/blood , Estrogens, Conjugated (USP)/administration & dosage , Estrogens, Conjugated (USP)/blood , Female , Humans , Middle Aged
9.
Expert Opin Investig Drugs ; 16(10): 1663-72, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17922629

ABSTRACT

Bazedoxifene acetate (WAY-140424; TSE-424) is an investigational non-steroidal indole-based selective estrogen receptor modulator (SERM) - also classified as an estrogen agonist/antagonist - that is being developed as a daily oral drug for the prevention and treatment of postmenopausal osteoporosis (PMO). Clinical studies have shown favorable effects on the skeleton, with prevention of bone loss in postmenopausal women without osteoporosis and reduction in vertebral fracture risk in women with PMO, without stimulation of endometrium or breast. Bazedoxifene combined with conjugated estrogens is an investigational tissue-selective estrogen complex, the first in a new class of therapeutic agents that pairs a selective estrogen receptor modulator with estrogens. Clinical trials with bazedoxifene/conjugated estrogens in postmenopausal women have shown skeletal benefit with improvement in menopausal vasomotor symptoms and little or no stimulation of endometrial or breast tissue. Bazedoxifene/conjugated estrogens is a potential agent for the prevention of PMO and control of menopausal symptoms.


Subject(s)
Estrogens, Conjugated (USP)/administration & dosage , Estrogens/administration & dosage , Indoles/administration & dosage , Osteoporosis, Postmenopausal/drug therapy , Selective Estrogen Receptor Modulators/administration & dosage , Animals , Bone Density/drug effects , Clinical Trials as Topic , Drug Therapy, Combination , Estrogens/pharmacokinetics , Estrogens/therapeutic use , Estrogens, Conjugated (USP)/pharmacokinetics , Estrogens, Conjugated (USP)/therapeutic use , Female , Humans , Indoles/chemistry , Indoles/pharmacokinetics , Indoles/therapeutic use , Selective Estrogen Receptor Modulators/chemistry , Selective Estrogen Receptor Modulators/pharmacokinetics , Selective Estrogen Receptor Modulators/therapeutic use
10.
J Steroid Biochem Mol Biol ; 85(2-5): 473-82, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12943738

ABSTRACT

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.


Subject(s)
Alzheimer Disease/drug therapy , Estrogens, Conjugated (USP)/therapeutic use , Menopause/physiology , Neurodegenerative Diseases/prevention & control , Alzheimer Disease/prevention & control , Animals , Estrogens, Conjugated (USP)/pharmacokinetics , Female , Humans , Menopause/drug effects , Metabolic Clearance Rate , Neurodegenerative Diseases/drug therapy
12.
Ceska Gynekol ; 68(2): 117-21, 2003 Mar.
Article in Czech | MEDLINE | ID: mdl-12749182

ABSTRACT

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.


Subject(s)
Beverages , Estradiol/analogs & derivatives , Estradiol/pharmacokinetics , Estrogens, Conjugated (USP)/pharmacokinetics , Food-Drug Interactions , Progesterone/pharmacokinetics , Administration, Oral , Biological Availability , Citrus paradisi , Estradiol/administration & dosage , Estrogens, Conjugated (USP)/administration & dosage , Female , Humans , Middle Aged , Progesterone/administration & dosage
13.
J Clin Pharmacol ; 42(3): 332-41, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11865971

ABSTRACT

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.


Subject(s)
Estradiol Congeners/administration & dosage , Estradiol Congeners/pharmacokinetics , Estrogen Replacement Therapy , Estrogens, Conjugated (USP)/administration & dosage , Estrogens, Conjugated (USP)/pharmacokinetics , Adult , Aged , Area Under Curve , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/pharmacokinetics , Double-Blind Method , Equilin/blood , Estradiol/blood , Estradiol Congeners/blood , Estrogen Replacement Therapy/statistics & numerical data , Estrogens, Conjugated (USP)/blood , Estrone/blood , Female , Humans , Middle Aged , Postmenopause/blood , Tablets
14.
Am J Obstet Gynecol ; 184(3): 255-63, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11228470

ABSTRACT

In the next decade many women will turn to the medical community for advice on maintaining or improving health after menopause. Estrogen replacement therapy, with or without progestins, alleviates menopausal symptoms, prevents or manages osteoporosis, and reduces the increased cardiovascular disease risk that results from estrogen deficiency caused by ovarian decline. Although several estrogen replacement products are available, the pharmacokinetics and efficacy of these products may vary depending on either the estrogen formulation or the route of administration, or both. For example, oral estrogens, which elicit a marked hepatic response, induce greater beneficial effects on serum lipoproteins than transdermal estrogens, which circumvent first-pass liver metabolism. Oral conjugated estrogens and transdermal estradiol increase bone density and prevent bone loss. This article summarizes the studies comparing estrogen formulations and discusses the differential effects of various estrogen products that promote postmenopausal health.


Subject(s)
Estrogen Replacement Therapy/methods , Estrogens, Conjugated (USP)/pharmacokinetics , Estrogens/pharmacokinetics , Postmenopause/physiology , Aged , Estrogen Replacement Therapy/adverse effects , Estrogen Replacement Therapy/standards , Estrogens/adverse effects , Estrogens/therapeutic use , Estrogens, Conjugated (USP)/therapeutic use , Female , Humans , Middle Aged , Postmenopause/drug effects , Progestins/pharmacokinetics , Progestins/therapeutic use , Therapeutic Equivalency
15.
Endocrinology ; 142(3): 1228-33, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11181539

ABSTRACT

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.


Subject(s)
Bone and Bones/drug effects , Estradiol/administration & dosage , Estradiol/pharmacology , Estrogens, Conjugated (USP) , Ovariectomy , Prodrugs , Animals , Aspartic Acid/analogs & derivatives , Bone Matrix/chemistry , Estradiol/analogs & derivatives , Estrogens, Conjugated (USP)/metabolism , Estrogens, Conjugated (USP)/pharmacokinetics , Estrogens, Conjugated (USP)/pharmacology , Female , Mice , Prodrugs/metabolism , Prodrugs/pharmacokinetics , Prodrugs/pharmacology , Proteins/genetics , RNA, Messenger/metabolism , Receptors, Estrogen/metabolism
16.
J Soc Gynecol Investig ; 7(3): 175-83, 2000.
Article in English | MEDLINE | ID: mdl-10865186

ABSTRACT

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.


Subject(s)
Estrogens, Conjugated (USP)/pharmacokinetics , Postmenopause , Adult , Cross-Over Studies , Equilin/analogs & derivatives , Equilin/blood , Estradiol/blood , Estrogen Replacement Therapy , Estrone/blood , Female , Humans , Middle Aged , Therapeutic Equivalency
17.
Rinsho Byori ; 48(11): 1059-63, 2000 Nov.
Article in Japanese | MEDLINE | ID: mdl-11132560

ABSTRACT

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.


Subject(s)
Estradiol/blood , Estrogen Replacement Therapy , Estrogens, Conjugated (USP)/pharmacokinetics , Radioimmunoassay/methods , Biomarkers/blood , Cross Reactions , Estrogens, Conjugated (USP)/administration & dosage , Female , Humans , Male , Monitoring, Physiologic/methods , Reagent Kits, Diagnostic , Sensitivity and Specificity
18.
Rev. chil. obstet. ginecol ; 65(1): 1-8, 2000. tab, graf
Article in Spanish | LILACS | ID: lil-267674

ABSTRACT

Determinar el efecto sobre la sintomatología clínica, perfil lipídico y niveles plasmáticos de estradiol y estrona frente a la administración de estrógenos conjugados equinos y estrógenos conjugados genéricos formulados localmente. Estudio prospectivo, controlado y randomizado, comparando cuatro grupos de 20 pacientes cada uno con formulaciones diferentes de estrógenos conjugados (EC) contra placebo por un período de 6 meses. No hubo diferencias significativas en la respuesta clínica ni en los niveles plasmático de estradiol y estrona entre los grupos con EC. Todos los grupos mostraron mejorías en el perfil lipídico al compararse contra placebo, se encontró significación sólo en dos de ellos. La respuesta a la sintomatología clínica así como los niveles plasmáticos de estradiol son comparables entre los grupos de EC. Las diferencias significativas entre estrógenos conjugados equinos y los genéricos sobre algunas fracciones de perfil lipídico deber ser precisadas con estudios complementarios para precisar su real significado


Subject(s)
Female , Estrogens, Conjugated (USP)/pharmacokinetics , Premenopause/drug effects , Estrogen Replacement Therapy/methods , Estradiol Congeners/pharmacokinetics , Estradiol/blood , Estrone/blood , Lipids/blood , Placebos/pharmacokinetics
19.
J Steroid Biochem Mol Biol ; 67(1): 33-40, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9780027

ABSTRACT

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.


Subject(s)
Amnion/physiology , Chorion/physiology , Estrone/analogs & derivatives , Animals , Biological Transport , Estrogens, Conjugated (USP)/pharmacokinetics , Estrone/pharmacokinetics , Female , Gestational Age , Guinea Pigs , In Vitro Techniques , Kinetics , Permeability , Pregnancy , Sulfur Radioisotopes , Tritium
20.
Arzneimittelforschung ; 48(9): 941-7, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9793623

ABSTRACT

A randomized, single-dose cross-over study in 32 postmenopausal women was performed to demonstrate bioequivalence of two estradiol valerate containing formulations (first sequence of Klimonorm as test preparation). The serum levels of estradiol, free and conjugated estrone were measured until 48 h after an oral dosage of 4 mg estradiol valerate (CAS 979-32-8). The mean AUC(0-48) of estradiol was calculated as 1006.6 +/- 479.4 h x pg x ml-1 (Test) and 1015.2 +/- 555.2 h x pg x ml-1 (Reference). The corresponding (AUC(0-48) of the active metabolite, free estrone, exceeded that of estradiol at 3578.3 h x pg x ml-1 (Test) and 3485.1 h x pg x ml-1 (Reference). Much higher was the AUC(0-48) for conjugated estrone at 132.4 h x ng x ml-1 (Test) and 133.6 h x ng x ml-1 (Reference). Mean estradiol Cmax values of 39.8 +/- 17.7 pg/ml (Test) and 42.9 +/- 21.0 pg/ml (Reference) were attained 8.2 +/- 4.5 h (Test) and 10.0 +/- 5.9 h (Reference) after the administration of 4 mg estradiol valerate. Maximal free estrone concentrations of 163 pg/ml (Test) and 174.3 pg/ml (Reference) were reached after 7.2 h (Test) and 7.5 h (Reference). Maximal conjugated estrone concentrations of 15.5 ng/ml (Test) and 16.2 ng/ml (Reference) were reached after 2.4 h (Test) and 2.0 h (Reference). The terminal elimination half-life of estradiol was calculated at 16.9 +/- 6.0 h (Test) and 15.0 +/- 4.8 h (Reference), that of free estrone at 16.3 h (Test) and 13.5 h (Reference), that of conjugated estrone at 11.8 h (Test) and 10.6 h (Reference). After logarithmic transformation, the 90% confidence intervals of the AUC(0-48) and Cmax ratios for estradiol and also for the metabolites (free and conjugated estrone) were within the acceptance ranges for bioequivalence. Therefore the test preparation and the reference preparation are bioequivalent.


Subject(s)
Estradiol/analogs & derivatives , Estrogens, Conjugated (USP)/pharmacokinetics , Postmenopause/metabolism , Administration, Oral , Area Under Curve , Cross-Over Studies , Estradiol/administration & dosage , Estradiol/blood , Estradiol/pharmacokinetics , Estrogens, Conjugated (USP)/administration & dosage , Estrone/blood , Female , Half-Life , Humans , Middle Aged , Radioimmunoassay , Reproducibility of Results , Therapeutic Equivalency
SELECTION OF CITATIONS
SEARCH DETAIL
...