Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
J Anim Physiol Anim Nutr (Berl) ; 101(1): 127-135, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27121903

RESUMO

The key biological active molecule of soya is the isoflavone daidzein, which possesses phytoestrogenic activity. The direct effect of soya and daidzein on ovarian cell functions is not known. This study examined the effect of daidzein on basic porcine ovarian granulosa cell functions and the response to follicle-stimulating hormone (FSH). We studied the effects of daidzein (0, 1, 10 and 100 µm), FSH (0, 0.01, 0.1, 1 IU/ml) and combinations of FSH (0, 0.01, 0.1, 1 IU/ml) + daidzein (50 µm) on proliferation, apoptosis and hormone release from cultured porcine ovarian granulosa cells and ovarian follicles. The expression of a proliferation-related peptide (PCNA) and an apoptosis-related peptide (Bax) was analysed using immunocytochemistry. The release of progesterone (P4) and testosterone (T) was detected using EIA. Leptin output was analysed using RIA. Daidzein administration increased granulosa cell proliferation, apoptosis and T and leptin release but inhibited P4 output. Daidzein also increased T release and decreased P4 release from cultured ovarian follicles. Follicle-stimulating hormone stimulated granulosa cell proliferation, apoptosis and P4, T and leptin release. The addition of daidzein promoted FSH-stimulated apoptosis (but not proliferation) but suppressed FSH-stimulated P4, T and leptin release. Our observations of FSH action confirm previous data on the stimulatory effect of FSH on ovarian cell proliferation, apoptosis and steroidogenesis and demonstrate for the first time the involvement of FSH in the upregulation of ovarian leptin release. Our observations of daidzein effects demonstrated for the first time that this soya isoflavone affected basic ovarian cell functions (proliferation, apoptosis and hormones release) and modified the effects of FSH. Daidzein promoted FSH action on ovarian cell proliferation and apoptosis and suppressed, and even inverted, FSH action on hormone release. The direct action of daidzein on basic ovarian cell functions and the ability of these cells to respond to FSH indicate the potential influence of soya-containing diets on female reproductive processes via direct action on the ovary.


Assuntos
Hormônio Foliculoestimulante/farmacologia , Células da Granulosa/efeitos dos fármacos , Isoflavonas/farmacologia , Suínos , Animais , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Interações Medicamentosas , Feminino , Hormônio Foliculoestimulante/administração & dosagem , Hormônio Foliculoestimulante/farmacocinética , Células da Granulosa/fisiologia , Isoflavonas/administração & dosagem , Isoflavonas/farmacocinética , Fitoestrógenos/administração & dosagem , Fitoestrógenos/farmacocinética , Fitoestrógenos/farmacologia
2.
Expert Opin Investig Drugs ; 31(11): 1255-1263, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36537742

RESUMO

OBJECTIVE: KN015 is a long-acting, recombinant human follicle-stimulating hormone Fc fusion protein that induces follicle development. This first-in-human study evaluated the effect of KN015 on healthy, pituitary-suppressed women and examined its pharmacokinetics, pharmacodynamics, and tolerability. METHODS: This phase I study was a double-blind, randomized, and placebo-controlled design with a single ascending dose (20, 40, and 60 µg, respectively). RESULTS: After subcutaneous administration of a single dose, the maximum serum KN015 concentrations reached 1.57, 2.78, and 3.62 ng/mL, respectively, after baseline adjustment. Over this dose range, the median Tmax occurred at 240-312 h, and the half-life (t½) was 752-1160 h. Dose proportionality was shown across the studied dose range. In most subjects, follicular growth was observed, and the number and diameter of the follicles increased with an increasing dose. In the 40-µg and 60-µg groups, the mean numbers of follicles with a diameter of ≥17 mm were 3 and 4, respectively. There was no significant difference in adverse events between the KN015 and placebo groups. KN015 antibody was not detected in any of the dosage groups. CONCLUSION: The administration of a single ascending dose of KN015 was tolerated and able to induce follicular growth. TRIAL REGISTRATION: This trial is registered at the Chinese Clinical Trials website (http://www.chinadrugtrials.org.cn/index.html # CTR20160741) and ClinicalTrials.gov (https://clinicaltrials.gov/ # NCT03192527).


Assuntos
Hormônio Foliculoestimulante Humano , Hormônio Foliculoestimulante , Humanos , Feminino , Hormônio Foliculoestimulante/farmacocinética , Hormônio Foliculoestimulante Humano/efeitos adversos , Proteínas Recombinantes , Meia-Vida , Método Duplo-Cego , Relação Dose-Resposta a Droga
3.
Front Endocrinol (Lausanne) ; 12: 636038, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33815292

RESUMO

It is well accepted that pituitary follitropin is secreted into the circulation as a mixture of variants, which differ not in primary structure but rather at the level of glycosylation. These glycosidic forms vary in the number of glycosylation sites filled, complexity of glycosidic chains, and sialylation and sulfation. It is generally agreed that high sialylation, 2,3 sialic acid capping of terminal N-acetyl galactosamine or galactose leads to longer circulating half-life, by blocking binding of asialoglycoprotein receptor (ASGPR) in the liver. In contrast, 2,6 sialic acid found in humans does not prevent recognition of galactose and N-acetyl galactosamine by ASGPR. Few studies on clinical outcomes comparing differences in sialylation of follitropin found in commercially available preparations are available. Thus, there is a clear need for a consortium of open data to address this unmet need. Recently, FSH glycosylation, primarily on the ß-subunit, which varies as women age, has emerged as a key modifier of follitropin action, with profound biological effects in vivo in animal models. To date, limited information of recombinant follitropin hormone preparations is available. Thus, most of the studies with FSH that is well characterized biochemically have been done in vitro, with engineered non gonadal host cells bearing recombinant receptors or in animal models. Since limited studies in human granulosa cells are available, a question is whether structural differences in glycosylation in commercially available follitropin affects biological function and clinical effect in humans. The presence of fucose, for example, has not been studied greatly even though, in the case of antibody therapy it has been shown to have a large effect on antibody targeting. This review on glycosidic variability of follitropin from the biochemical/structural point of view reflects on this question and presents an assessment in the context of available published data. If clinical differences are to be expected or not, the readers will have a better understanding of the evidence for and limitations of such expectations.


Assuntos
Receptor de Asialoglicoproteína/metabolismo , Hormônio Foliculoestimulante/administração & dosagem , Polissacarídeos/química , Acetilgalactosamina/metabolismo , Animais , Células CHO , Cricetulus , Feminino , Hormônio Foliculoestimulante/metabolismo , Hormônio Foliculoestimulante/farmacocinética , Hormônio Foliculoestimulante Humano/metabolismo , Glicoproteínas/metabolismo , Glicosilação , Humanos , Fígado/metabolismo , Masculino , Camundongos , Ácido N-Acetilneuramínico/química , Isoformas de Proteínas , Ratos , Proteínas Recombinantes/metabolismo
4.
J Clin Pharmacol ; 61(5): 700-713, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33274472

RESUMO

This study aimed to characterize an interactive and clinically applicable population pharmacokinetic-pharmacodynamic-pharmacodynamic (PK-PD-PD) model describing follicle-stimulating hormone (FSH)-inhibin B-oocyte relationship in women undergoing assisted reproduction with in vitro fertilization (IVF) or intracytoplasmic sperm injection (ICSI). The study was a prospective analysis of 25 healthy women undergoing IVF/ICSI using gonadotropin-releasing hormone (GnRH) antagonist protocol. The developed model used the FSH PK profiles to predict both inhibin B (first PD end point) and oocyte retrieval (second PD end point). The modeling framework involved 2 stages. First, the FSH-inhibin B model was developed by the simultaneous approach and applied to estimate the individual area under the inhibin B-time curve (AUCInhb ) at the end of stimulation cycles that varied in length in each woman. In the second stage, the estimated AUCInhb was introduced as a link covariate to predict oocyte retrieval and response category. The population FSH-inhibin B model was described as 3 submodels; PK (exogenous), endogenous, and inhibin B PD models. Weight was the main determinant of both endogenous and exogenous FSH exposures. GnRH antagonist therapy was a significant time-varying covariate when tested against the endogenous FSH production rate (P < .001). AUCInhb could be predicted with women's age and weight. Log-transformed AUCInhb was a significant covariate when tested against oocyte retrieval (P < .001). Simulations concluded a target AUCInhb of 144-303 ng·h/mL for optimal ovarian response. The GnRH antagonist was better started on day 7 of the cycle. Covariate-based dosing suggests lower recombinant follicle-stimulating hormone requirements in a thin and/or young population. An interactive web application "GonadGuide" was developed to facilitate the application in clinical practice.


Assuntos
Fertilização in vitro/efeitos dos fármacos , Hormônio Foliculoestimulante/farmacologia , Hormônio Liberador de Gonadotropina/antagonistas & inibidores , Inibinas/efeitos dos fármacos , Injeções de Esperma Intracitoplásmicas/efeitos dos fármacos , Adulto , Área Sob a Curva , Relação Dose-Resposta a Droga , Feminino , Fertilização in vitro/métodos , Hormônio Foliculoestimulante/administração & dosagem , Hormônio Foliculoestimulante/farmacocinética , Humanos , Recuperação de Oócitos/métodos , Estudos Prospectivos , Proteínas Recombinantes , Injeções de Esperma Intracitoplásmicas/métodos , Adulto Jovem
5.
Reprod Biomed Online ; 21(5): 593-601, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20843746

RESUMO

A single injection of corifollitropin alfa can replace seven daily injections of recombinant FSH (rFSH) using a gonadotrophin-releasing hormone antagonist protocol in ovarian stimulation prior to IVF or intracytoplasmic sperm injection. This double-blind randomized controlled trial assessed the pharmacokinetics and pharmacodynamics of 150µg corifollitropin alfa versus daily 200IU rFSH in 1509 patients. Comparative analyses were performed on serum concentrations of FSH immunoreactivity (pharmacokinetics), and the number and size of growing follicles, and inhibin B and oestradiol concentrations as biomarkers of ovarian response (pharmacodynamics). The rate of follicular development was similar in both treatment groups. By stimulation day 8, 33% of patients treated with corifollitropin alfa reached the criterion for human chorionic gonadotrophin (HCG) injection. The number of follicles ⩾11mm was slightly higher after corifollitropin alfa compared with daily rFSH at stimulation day 8 (difference, 1.2; 95% confidence interval (CI) 0.5-1.8; P<0.01) and on the day of HCG injection (difference, 2.1; 95% CI 1.4-2.8; P<0.01). The rise of inhibin B and oestradiol concentrations was similar in both treatment groups. Although the pharmacokinetics of corifollitropin alfa and rFSH are quite different their pharmacodynamic profiles at the dosages used are similar. A single injection of corifollitropin alfa can replace seven daily injections of recombinant FSH (rFSH) using a gonadotrophin-releasing hormone antagonist protocol in ovarian stimulation prior to IVF or intracytoplasmic sperm injection. The objective of this study was to compare the pharmacokinetics and pharmacodynamics of corifollitropin alfa versus daily rFSH. A total of 1509 patients were randomized in a double-blind, controlled trial to either a single injection of 150µg corifollitropin alfa or to daily injections of 200IU rFSH for the first 7 days of ovarian stimulation. Serum levels of FSH immunoreactivity were analysed (pharmacokinetic analysis), together with the number and size of growing follicles and serum inhibin B and oestradiol concentrations as biomarkers of the ovarian response (pharmacodynamic analysis). Serum FSH immunoreactivity levels were higher up to stimulation day 5 for corifollitropin alfa compared with the daily rFSH regimen but were similar from day 8 onwards, when patients started rFSH if the criteria for human chorionic gonadotrophin were not yet reached. Corifollitropin alfa treatment resulted in a similar growth rate of follicles though a slightly higher number of follicles were recruited compared with daily rFSH. It is concluded that the pharmacokinetics of corifollitropin alfa and rFSH are quite different but their induced pharmacodynamic effects at the dosages used are similar.


Assuntos
Hormônio Foliculoestimulante Humano/farmacocinética , Hormônio Foliculoestimulante Humano/uso terapêutico , Hormônio Foliculoestimulante/farmacocinética , Indução da Ovulação/métodos , Adulto , Gonadotropina Coriônica/administração & dosagem , Estradiol/sangue , Feminino , Fertilização in vitro , Hormônio Foliculoestimulante/administração & dosagem , Hormônio Foliculoestimulante/sangue , Humanos , Inibinas/sangue , Folículo Ovariano/efeitos dos fármacos , Folículo Ovariano/fisiologia , Gravidez , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/uso terapêutico
6.
Reprod Biol Endocrinol ; 7: 107, 2009 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-19807931

RESUMO

BACKGROUND: Follicle stimulating hormone (FSH) has been routinely used for ovulation induction. Because of rapid clearance of the hormone, FSH is commonly administered by daily intramuscular or subcutaneous injections in in-vitro fertilization (IVF). To reduce the number of visits to the clinic, an intermittent vaginal injection of rhFSH every 3 days employing the concepts of mesotherapy and uterine first-pass effect was invented and has successfully been applied in women receiving IVF treatment. This study was designed to monitor the pharmacokinetic pattern of rhFSH administered vaginally. METHODS: Twelve healthy women with regular ovulatory cycles were recruited. All volunteers received gonadotrophin-releasing hormone agonist to suppress pituitary function and were assigned to receive single dose recombinant human FSH (rhFSH, Puregon 300) either using conventional abdominal subcutaneous injection or vaginal subcutaneous injection in a randomized cross-over study. Serum samples were collected at pre- scheduled time intervals after injections of rhFSH to determine immunoreactive FSH levels. Pharmacokinetic parameters characterizing rate [maximal plasma concentrations (Cmax) and time of maximal plasma concentrations (tmax)] and extent [area under the plasma concentration-time curve (AUC) and clearance] of absorption of rhFSH were compared. RESULTS: Vaginal injection of rhFSH was well tolerated and no drug-related adverse reaction was noted. Our analysis revealed that tmax was significantly earlier (mean 6.67 versus 13.33 hours) and Cmax was significantly higher (mean 17.77 versus 13.96 IU/L) in vaginal versus abdominal injections. The AUC(0-infinity) was 1640 versus 1134 IU hour/L in vaginal and abdominal injections, respectively. Smaller plasma elimination rate constant (0.011 versus 0.016 hour-1), longer mean residence time (106.58 versus 70.47 hours), and slower total body clearance (292.2 versus 400.1 mL/hour) were also found in vaginal injection. CONCLUSION: The vaginal injection mode elicited a rapid and highly extended absorption of rhFSH injected compared to conventional abdominal injection. These data indicate that the rate and extent of FSH absorption from the injection site can vary depending on the route of the FSH administration.


Assuntos
Hormônio Foliculoestimulante/administração & dosagem , Hormônio Foliculoestimulante/farmacocinética , Vagina/metabolismo , Absorção , Administração Intravaginal , Adulto , Estudos Cross-Over , Feminino , Hormônio Foliculoestimulante/efeitos adversos , Hormônio Foliculoestimulante/sangue , Humanos , Injeções Subcutâneas , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/efeitos adversos , Proteínas Recombinantes/farmacocinética , Vagina/efeitos dos fármacos
7.
Theriogenology ; 112: 11-17, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-28888334

RESUMO

Assisted reproduction technologies are widely used in humans and domestic animals and often include follicle stimulating hormone (FSH) in the protocol. One limitation with most of the available FSH preparations is the relative short half-life in the circulation that dictates multiple daily injections for the desired follicle development and superovulation. The development of bioactive long-acting structurally modified FSH analogs is desirable for human and veterinary use. In addition, optimal preparations and/or formulations are expected to improve the regimen and efficiency of the treatment. This review briefly describes the approaches that have been explored to extend the half-life of FSH in the circulation. These include strategies to increase the mass and/or charge of FSH and to prevent the dissociation of the hormone to inactive subunits components. Most of these strategies, except one that led to a registered drug (Elonva) indicated for controlled ovarian stimulation protocols in humans, are still in experimental stage.


Assuntos
Hormônio Foliculoestimulante/análogos & derivados , Técnicas de Reprodução Assistida , Animais , Animais Domésticos , Desenho de Fármacos , Feminino , Hormônio Foliculoestimulante/administração & dosagem , Hormônio Foliculoestimulante/farmacocinética , Meia-Vida , Humanos , Indução da Ovulação/métodos , Polietilenoglicóis , Engenharia de Proteínas , Técnicas de Reprodução Assistida/veterinária , Superovulação
8.
Anim Reprod Sci ; 102(3-4): 328-34, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17418509

RESUMO

Extracts of anterior pituitary (AP) glands were infused i.v. into hypophysectomized male rats followed by sequential sampling of blood for 120 min. Determination of follicle-stimulating hormone (FSH) concentrations established that FSH from Chinese Meishan males decreased in the circulation of rats more slowly than FSH in extracts of AP from crossbred occidental pigs (P<0.003). Additionally, FSH from AP extracts of castrated males disappeared somewhat more slowly (P<0.06) than FSH from extracts of boars. Evaluation of FSH by bioassay and radioimmunoassay yielded similar concentrations in AP from Meishan and crossbred boars. Serum testosterone concentrations increased with time through 90 min after infusion of AP, but the rate of increase of testosterone was not related to amount of luteinizing hormone (LH) that was administered indicating LH receptor saturation. Unexpectedly, the rate of increase in testosterone was more rapid with AP extracts from boars than with extracts from castrated males. Observations from the current study imply structural alterations of FSH in the AP of Meishan males relative to crossbred males allowing sustained concentrations in the circulation, and this FSH possesses similar activation of the FSH receptor. The amount of LH in the AP extracts saturated the LH receptors of the hypophysectomized male rats, but some factor in extracts of boars differed from those of castrated males.


Assuntos
Hormônio Foliculoestimulante/farmacocinética , Hipofisectomia , Suínos , Animais , Hormônio Foliculoestimulante/análise , Hormônio Foliculoestimulante/sangue , Masculino , Taxa de Depuração Metabólica , Orquiectomia , Adeno-Hipófise/química , Adeno-Hipófise/metabolismo , Ratos , Ratos Sprague-Dawley , Especificidade da Espécie , Testosterona/sangue , Extratos de Tecidos/administração & dosagem , Extratos de Tecidos/química
9.
Theriogenology ; 66(4): 896-905, 2006 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-16580718

RESUMO

Embryo production is a useful tool for ex situ conservation of endangered species and breeds, despite a high variability in the ovarian response to superovulatory treatments. The current study evaluated the incidence and mechanisms of genetic factors in such variability, by determining the pharmacokinetics and pharmacodynamics of a standard treatment with ovine FSH (oFSH) in two endangered Spanish sheep breeds (Rubia del Molar, R, and Negra de Colmenar, N) in comparison to Manchega ewes (M, control group). In the first experiment, pharmacokinetics of an i.m. single dose of 1.32 mg of oFSH was evaluated in seven animals of each breed. Plasma FSH concentrations reached their maximum at 4h post-administration in all the ewes, but several of the kinetic parameters (plasma FSH concentration at 4h post-administration, maximum plasma FSH concentration, C(max), and both the area under the plasma concentration-time curve extrapolated to the infinite, AUC(inf), and to the last moment of sampling, AUC(last)) were higher in the N group. In the second trial, 10 animals of each breed were superovulated using eight decreasing doses of oFSH (3 x 1.32 mg, 2 x 1.10 mg, and 3 x 0.88 mg). The R group, when compared to N and M, showed both a higher number of corpora lutea (13.7+/-0.6 versus 10.0+/-0.4 in N and 9.8+/-0.6 in M, P<0.05 for both) and embryos (7.9+/-0.8 versus 4.3+/-0.4 in N, P<0.05, and 6.7+/-0.5 in M, n.s.). Evaluation of pharmacokinetic and dynamic parameters showed that, although there was a trend for a higher hormone availability in R sheep, mean FSH plasma concentrations were similar between breeds (0.54+/-0.08 ng/ml for R, 0.45+/-0.05 ng/ml for N and 0.35+/-0.05 ng/ml for M). However, differences were found in the number of preovulatory follicles growing in response to the FSH treatment between R (24.4+/-2.2), M (18.9+/-1.5, n.s.) and N sheep (14.1+/-1.4; P<0.01). Thus, differences in embryo yields between breeds would be related to differences in the pattern of follicular growth in response to FSH treatment.


Assuntos
Cruzamento , Hormônio Foliculoestimulante/sangue , Hormônio Foliculoestimulante/farmacocinética , Folículo Ovariano/efeitos dos fármacos , Ovinos/fisiologia , Superovulação/sangue , Animais , Tamanho Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Feminino , Hormônio Foliculoestimulante/administração & dosagem , Masculino , Folículo Ovariano/citologia , Folículo Ovariano/metabolismo , Ovinos/sangue , Especificidade da Espécie , Superovulação/metabolismo
10.
Clin Drug Investig ; 36(12): 1031-1044, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27638053

RESUMO

BACKGROUND AND OBJECTIVE: Highly purified human menotrophin and urofollitrophin preparations obtained from human urine via a novel patented purification method have been tested over a timeframe of 14 years in the studies presented in this article. The objective of the studies was to investigate the pharmacokinetics and the pharmacodynamics of follicle-stimulating hormone (FSH) after single subcutaneous and intramuscular doses and multiple subcutaneous doses of the tested preparations in healthy fertile pituitary-suppressed women. DESIGNS: We performed five open, randomised, crossover, single-dose bioequivalence and/or bioavailability studies and one open, multiple-dose, pharmacokinetics and pharmacodynamics study. STUDY SUBJECTS AND TREATMENTS: The six studies included 121 healthy fertile women taking their usual combined oral contraceptives for 3 months before the study: Study 1: 300 international units (IU) of highly purified menotrophin as single subcutaneous and intramuscular doses. Study 2: 300 IU of highly purified menotrophin (test formulation vs. comparator) as single subcutaneous doses. Study 3: 300 IU of highly purified urofollitrophin (hp-FSH) (test formulation vs. comparator) as single subcutaneous doses. Study 4: 300 IU (2 × 150 IU vs. 4 × 75 IU) of hp-FSH as single subcutaneous doses. Study 5: 225 and 445 IU of hp-FSH as single subcutaneous doses. Study 6: daily 225 IU of hp-FSH as subcutaneous doses for 5 consecutive days. MAIN OUTCOME MEASURES: The main outcome measures were the FSH pharmacokinetic parameters, estradiol concentrations, and the number and size of the follicles. RESULTS: FSH after single subcutaneous and intramuscular injections of menotrophin or urofollitrophin attained a systemic peak (maximum) concentration (C max) that was on average consistent throughout the first four studies and ranged from 4.98 to 7.50 IU/L. The area under the plasma concentration-time curve (AUC) from administration to the last observed concentration time t (AUCt) ranged from 409.71 to 486.16 IU/L·h and the elimination half-life (t ½) ranged from 39.02 to 53.63 h. After multiple doses of urofollitrophin (225 IU) for 5 days, FSH attained a mean C max of 14.93 ± 2.92 IU/L and had an AUC during the time interval τ between two consecutive doses at steady state (AUCτ) of 322.59 ± 57.92 IU/L·h, which was similar to the mean AUCt after a single subcutaneous dose of 225 IU of urofollitrophin in study 5 (306.82 ± 68.37 IU/L·h). CONCLUSIONS: In our studies, the intramuscular and subcutaneous routes of menotrophin were equivalent; both menotrophin and urofollitrophin were bioequivalent to their marketed reference; FSH kinetic parameters following injection of urofollitrophin were dose proportional and independent from the administered concentration; and multiple doses of FSH increased estradiol levels and enhanced growth of follicles with a good dose-response correlation. Local tolerability was excellent throughout the six studies.


Assuntos
Hormônio Foliculoestimulante/farmacocinética , Menotropinas/administração & dosagem , Urofolitropina/administração & dosagem , Adulto , Disponibilidade Biológica , Anticoncepcionais Orais Combinados , Estudos Cross-Over , Relação Dose-Resposta a Droga , Estradiol/sangue , Feminino , Meia-Vida , Humanos , Injeções Subcutâneas , Menotropinas/farmacocinética , Equivalência Terapêutica , Urofolitropina/farmacocinética
11.
Curr Med Res Opin ; 21(1): 121-5, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15881483

RESUMO

OBJECTIVE: The bioequivalence and tolerability of freeze-dried and liquid formulations of recombinant human follicle-stimulating hormone (r-hFSH) filled-by-mass were assessed in a crossover, open-label, randomised, single-centre, phase I bioequivalence study. METHODS: Following pituitary down-regulation with the gonadotrophin-releasing hormone agonist goserelin, healthy adult volunteers (18years-45years of age) received single subcutaneous injections of r-hFSH , 300 IU, from freeze-dried and liquid formulations in random order, separated by a 7-day washout period. Blood was obtained over 144 h for pharmacokinetic analysis. MAIN OUTCOME MEASURES: These were peak serum FSH concentrations (Cmax,), time to peak concentration (Tmax) and area under the concentration-time curve from zero to the last measurable concentration (AUCJ), local and systemic tolerability. RESULTS: Of 44 volunteers who underwent down-regulation, 39 (18 men, 21 women) completed the study. Cmax and AUClast were similar with the freeze-dried (mean 9.51 IU/L and 844 IU.h/L, respectively) and liquid (mean 8.99 IU/L and 841 IUh/L, respectively) formulations, whereas T was significantly higher with the liquid formulation (median 12h vs 15h, p = 0.0183). The 90% confidence intervals for the ratio of the treatment means for Cnw and AUC,=, were within the pre-defined bioequivalence range of 0.8-1.25. CONCLUSION: Both formulations were well tolerated with regard to both systemic and local adverse events. The freeze-dried and liquid formulations of r-hFSH are bioequivalent and show no significant differences in tolerability. Thus, the liquid formulation is expected to provide comparable efficacy and tolerability to the freeze-dried formulation in clinical use.


Assuntos
Hormônio Foliculoestimulante/farmacocinética , Adolescente , Adulto , Análise de Variância , Área Sob a Curva , Estudos Cross-Over , Feminino , Liofilização , Humanos , Masculino , Pessoa de Meia-Idade , Estatísticas não Paramétricas , Equivalência Terapêutica
12.
Treat Endocrinol ; 4(3): 155-65, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15898821

RESUMO

Anovulation is a common cause of female infertility. Treatment for women with anovulation is aimed at induction of ovulation. Ovulation induction with follicle-stimulating hormone (FSH) is indicated in women with WHO type II anovulation in whom treatment with clomifene citrate (clomifene) has failed. The majority of these women have polycystic ovary syndrome. The major disadvantages of ovulation induction with FSH are the risk of ovarian hyperstimulation syndrome and the risk of higher order multiple pregnancies. To reduce the rate of complications due to multiple follicular development, FSH should be administered using a chronic low-dose protocol with small dose increments. In women with WHO type I anovulation, an exogenous supply of luteinizing hormone (LH) is required to achieve an adequate follicular response to FSH treatment. Thus, ovulation induction with FSH is not the treatment of choice in these women. FSH is a hormone that stimulates follicle growth and oocyte maturation. Endogenous FSH is produced by the pituitary gland and exists as a family of isohormones exhibiting distinct oligosaccharide structures. FSH for exogenous administration is derived from urine or is produced as recombinant FSH. The commercially available FSH products all contain different mixtures of FSH isoforms. To determine the effectiveness of urofollitropin (urinary-derived FSH), a comparison with the other available gonadotropins was made (i.e. recombinant FSH and human menopausal gonadotropin). Urofollitropin and recombinant FSH appear to be equally effective and well tolerated for ovulation induction. Human menopausal gonadotropin is comparably effective to urofollitropin in terms of pregnancy outcomes. It remains unclear whether human menopausal gonadotropins have a higher risk of overstimulation and ovarian hyperstimulation syndrome compared to urofollitropin in women with polycystic ovary syndrome. In practice, recombinant products are more convenient to use but are also more expensive. Therefore, if availability is not an issue but costs are, there is still a place for the use of urofollitropins for ovulation induction.


Assuntos
Menotropinas/uso terapêutico , Indução da Ovulação/métodos , Anovulação/tratamento farmacológico , Custos de Medicamentos , Feminino , Hormônio Foliculoestimulante/química , Hormônio Foliculoestimulante/farmacocinética , Hormônio Foliculoestimulante/uso terapêutico , Humanos , Menotropinas/administração & dosagem , Proteínas Recombinantes
13.
Theriogenology ; 83(1): 30-7, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25277529

RESUMO

Follicular fluid meiosis-activating sterol (FF-MAS), an intermediate in the cholesterol biosynthetic pathway, has been identified as a compound that induces the resumption of meiosis in mammalian oocyte. Follicular fluid meiosis-activating sterol is converted to testis meiosis-activating sterol by a sterol Δ14-reductase. An inhibitor of Δ14-reductase and Δ7-reductase, AY9944 A-7, causes accumulation of FF-MAS by inhibiting its metabolism. The objective of this research was to investigate the specific contribution of AY9944 A-7 on gonadotropin-induced meiotic resumption and its interactive effects with FSH or LH on meiotic maturation of oocytes and preimplantation development of parthenogenetic embryo in sheep by addition of AY9944 A-7 during IVM to cause accumulation of FF-MAS. First, ovine cumulus-oocyte complexes (COCs) were cultured in the presence of FSH (10 µg/mL), LH (10 µg/mL), AY9944 A-7 (20 µmol/L), FSH (10 µg/mL)+AY9944 A-7 (20 µmol/L), or LH (10 µg/ml) + AY9944 A-7 (20 µmol/L) with an inhibitor hypoxanthine (Hx) to prevent spontaneous meiosis of oocytes. The resumption of meiosis was assessed by the frequency of germinal vesicle breakdown and the first polar body (PBI) extrusion. The kinetics of gonadotropin and AY9944 A-7-induced meiotic resumption in vitro was also evaluated in the study. The numbers of oocytes resuming meiosis and undergoing germinal vesicle breakdown were counted after the COCs were cultured for 2, 4, 8, 12, 16, 20, and 24 hours. Matured oocytes extruding PBI were selected for parthenogenetic activation, and the percentages developing to the two-cell stage and blastocyst stage were recorded as indicators of parthenogenetic embryo developmental competence. It was observed that FSH could induce the resumption of meiosis of ovine COCs cultured in the presence of Hx, but LH could not. AY9944 A-7 had a synergistic effect with FSH on nuclear maturation and developmental competence of embryos produced by parthenogenetic activation, whereas it had no added advantage on LH action. However, the kinetics of meiotic resumption after AY9944 A-7 stimulation was remarkably delayed when compared with FSH-induced maturation. In conclusion, the current study suggested that AY9944 A-7 supplementation in IVM medium optimized the beneficial effects of FSH on meiotic maturation of ovine oocytes and subsequent developmental competence of embryos produced by parthenogenetic activation. This work had important potential for developing a novel technique in IVM of ovine oocytes.


Assuntos
Desenvolvimento Embrionário/efeitos dos fármacos , Gonadotropinas/farmacologia , Meiose/efeitos dos fármacos , Oócitos/efeitos dos fármacos , Partenogênese/fisiologia , Ovinos/embriologia , Dicloridrato de trans-1,4-Bis(2-clorobenzaminometil)ciclo-hexano/farmacologia , Animais , Interações Medicamentosas , Técnicas de Cultura Embrionária/veterinária , Feminino , Hormônio Foliculoestimulante/farmacocinética , Hormônio Foliculoestimulante/farmacologia , Técnicas de Maturação in Vitro de Oócitos/métodos , Técnicas de Maturação in Vitro de Oócitos/veterinária , Hormônio Luteinizante/farmacocinética , Hormônio Luteinizante/farmacologia , Meiose/fisiologia , Oócitos/citologia , Oócitos/fisiologia , Oxirredutases/antagonistas & inibidores , Fatores de Tempo
14.
J Clin Endocrinol Metab ; 80(5): 1657-63, 1995 May.
Artigo em Inglês | MEDLINE | ID: mdl-7745014

RESUMO

Gonadotropin treatment of hypogonadotropic infertile men usually requires regular im administration of human urinary FSH (uFSH); however, testicular function is rarely normalized despite years of treatment. As the pharmacokinetics of standard FSH doses (75 IU, two or three times weekly) in gonadotropin-deficient men are poorly characterized, we studied 10 gonadotropin-deficient men by measuring plasma FSH levels with an ultrasensitive fluoroimmunoassay (Delfia, Pharmacia) in single dose and multidose studies. The single dose studies involved blood samples taken 15 min before and 0, 1, 2, 4, 6, 8, 10, 12, 15, 18, 21, 24, 48, 72, and 96 h after the injection of 75 IU uFSH in 1 mL diluent, either sc under the abdominal wall skin or im into the deltoid muscle, in a random sequence, cross-over design (n = 7 men) and after the injection of 150 IU, sc, with additional blood sampling at 120 and 168 h (n = 7 men). The multidose studies used a fixed ascending dose sequence, with blood sampled at 24-h intervals posttreatment after at least 1 month of regular administration of either 75 or 150 IU uFSH, sc, at injection intervals of 72, 48, and 24 h (n = 6 men). From the single dose studies, pharmacokinetic variables were estimated from a one-compartment open model fitted by a weighted polyexponential curve fit of plasma FSH over time. The bioavailability of uFSH via the sc route was high (mean area under the curve, 90% for 75 IU and 143% for 150 IU vs. 75 IU, im). Peak plasma FSH levels were later (21.1 vs. 7.1 h; P < 0.001) and lower (2.0 vs. 2.7 IU/L; P < 0.001) after sc compared with im administration of 75 IU due to a slower absorption half-time (6.1 h vs. 1.4 h; P < 0.001), whereas mean residence times and clearance half-times were similar. The pharmacokinetic features of the 150- and 75-IU doses sc were essentially identical, apart from expected dose-dependent increases in peak plasma FSH level (2.8 vs. 2.0 IU/L; P < 0.001) and area under the curve (206 vs. 129 IU.h/L; P < 0.05). Multidose simulations based on the single dose pharmacokinetic models predicted that during chronic sc administration of standard FSH doses, plasma FSH levels would be in the lower half of the eugonadal range and fluctuate less than with im administration. The multidose study confirmed empirically these predictions. These studies form a pharmacological basis for a more flexible, cost-effective, and convenient self-administered sc regimen.(ABSTRACT TRUNCATED AT 400 WORDS)


Assuntos
Hormônio Foliculoestimulante/farmacocinética , Gonadotropinas/deficiência , Adulto , Relação Dose-Resposta a Droga , Esquema de Medicação , Hormônio Foliculoestimulante/administração & dosagem , Hormônio Foliculoestimulante/urina , Humanos , Injeções Subcutâneas , Masculino , Pessoa de Meia-Idade
15.
J Clin Endocrinol Metab ; 73(4): 818-23, 1991 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-1909706

RESUMO

We measured the equilibrium MCR of purified human FSH via continuous infusion and its half-life by bolus injection in gonadotropin-deficient men. Serum FSH concentrations were determined by RIA, immunoradiometric assay, and an in vitro FSH bioassay (rat Sertoli cell). At steady state, the MCR of FSH averaged 5.4 +/- 0.4 mL/min.m2, which was not statistically different in the three assays at the three different infusion rates. The half-life of FSH after bolus injection averaged 274 +/- 45 min (4.6 +/- 0.75 h) when analyzed as a single exponential, and 1.8 and 10 h for biexponential kinetics. The distribution volume of FSH was 3.1 L (immunoradiometric assay) and 2.1 L (bioassay). In summary, the MCR of human FSH is invariant across a range of physiological gonadotropin concentrations and quantitatively similar in three different immunological/biological assays. These results support the hypothesis that removal of circulating FSH molecules proceeds in parallel for immunoreactive and biologically active glycoprotein hormone.


Assuntos
Hormônio Foliculoestimulante/farmacocinética , Células de Sertoli/metabolismo , Adulto , Animais , Bioensaio , Células Cultivadas , Hormônio Foliculoestimulante/administração & dosagem , Hormônio Foliculoestimulante/sangue , Humanos , Ensaio Imunorradiométrico , Técnicas In Vitro , Injeções Intravenosas , Masculino , Taxa de Depuração Metabólica , Radioimunoensaio , Ratos
16.
J Clin Endocrinol Metab ; 88(7): 3227-35, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12843169

RESUMO

FSH is a key component in assisted reproductive technologies. Because of rapid clearance of the hormone, patients have to be treated with daily injections. To address this problem, a long-acting FSH mutein was created by introduction of additional N-linked glycosylation into the molecule. New glycosylation sites were introduced by two different approaches: structure-aided, site-directed introduction of sites within the FSH molecule and addition of N-terminal extensions. A mutein with the extension sequence ANITVNITV at the N terminus of the alpha-chain (FSH1208) was efficiently glycosylated at both new sites. This resulted in a molecule with increased size and charge, factors known to reduce renal clearance of proteins. FSH1208 was found to have a 3- to 4-fold increased serum half-life, compared with wild-type recombinant FSH. Furthermore, in spite of a lower in vitro activity, FSH1208 had a markedly increased in vivo potency, as shown by increased ability to augment the ovarian weight and stimulate the serum estradiol levels in rats. These characteristics make FSH1208 a possible candidate for improved infertility treatment.


Assuntos
Hormônio Foliculoestimulante/genética , Hormônio Foliculoestimulante/farmacocinética , Animais , Células CHO , Cricetinae , Desenho de Fármacos , Estradiol/sangue , Feminino , Hormônio Foliculoestimulante/química , Expressão Gênica , Glicosilação , Humanos , Técnicas In Vitro , Infertilidade Feminina/tratamento farmacológico , Tamanho do Órgão , Ovário/anatomia & histologia , Ovário/efeitos dos fármacos , Estrutura Terciária de Proteína , Ratos , Ratos Sprague-Dawley
17.
J Clin Endocrinol Metab ; 89(10): 5204-12, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15472227

RESUMO

The effects of altering the number and type of additional carbohydrate moieties on the pharmacokinetic and pharmacodynamic properties of FSH were examined in this report. A series of single-chain follitropins, containing variable numbers of additional N- (or O-) linked carbohydrates, were designed and expressed in Chinese hamster ovary cells. Proper folding, efficient receptor binding, and signal transduction were confirmed by in vitro assays. Pharmacokinetic and pharmacodynamic parameters were evaluated in immature female Sprague Dawley rats. Increasing the number of glycosylation sites with either N- (or O-) linked moieties extended the elimination half-life as much as 2-fold compared with recombinant human FSH (rhFSH). However, there was a maximum elimination half-life such that further glycosylation provided no additional lengthening of the half-life. Conversely, biopotency, as assessed by inhibin A levels 74 h post injection, and follicle production were significantly higher for the N-linked analogs. Rats stimulated with the longest acting analogs (either N- or O-linked) showed significantly higher ovarian weights than rats receiving a single injection of rhFSH. The analog containing four additional N-linked sites (rhFSH-N4) had the greatest number of large, preovulatory follicles. Although the half-life of rhFSH-N4 displayed no further enhancement beyond the other longest acting analogs, this analog exhibited significantly increased biopotency in rats. This work provides the basis for the generation of a series of reagents potentially useful for therapeutic applications.


Assuntos
Hormônio Foliculoestimulante/análogos & derivados , Hormônio Foliculoestimulante/farmacocinética , Folículo Ovariano/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Células CHO , Cricetinae , Feminino , Hormônio Foliculoestimulante/genética , Glicosilação , Inibinas/metabolismo , Focalização Isoelétrica , Dados de Sequência Molecular , Tamanho do Órgão , Folículo Ovariano/citologia , Folículo Ovariano/metabolismo , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/farmacocinética
18.
J Endocrinol ; 182(1): 43-54, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15225130

RESUMO

The appropriate dosage regimen for equine FSH (eFSH) (dose, dosing interval) administration in a superovulation treatment in pony mares was determined by a kinetic approach using production rates and kinetic parameters of elimination of the hormone. Two dosage regimens were then tested in superovulation protocols. The eFSH production rates were determined by sampling four ovariectomized pony mares every 10 min for 8 h during the breeding season. Kinetic parameters were determined by administering four dose levels of a preparation of eFSH (4.4, 8.8, 17.6 and 35.2 micro g/kg) by the i.v. route to the same mares, in a randomized 4x4 Latin Square protocol. The overall mean plasma clearance was 0.256+/- 0.07 ml.kg(-1).min(-1), and was independent of the dose. The mean residence time ranged from 5.5 to 10.8 h and increased with the dose. The estimated FSH production rates were 8.6 to 15.3 micro g.kg(-1).day(-1) (i.e. 2.89 to 3.45 mg per day per mare). Two dosage regimens of eFSH were then tested in cyclic mares (ten treated mares in each trial): 3.45 mg per day (4.4 micro g/kg three times a day by the i.v. route), which corresponds to the maximal daily production rate of the native hormone in ovariectomized mares, and 1.72 mg per day (2.2 micro g/kg three times a day), which corresponds to half of that production rate. The dosage regimen of 2.2 micro g/kg three times a day gave satisfactory results in terms of efficacy (numbers of ovulations and embryos) with minimal unwanted effects (luteinized or anovulatory follicles).


Assuntos
Hormônio Foliculoestimulante/biossíntese , Cavalos , Indução da Ovulação/métodos , Indução da Ovulação/veterinária , Superovulação , Animais , Cruzamento , Relação Dose-Resposta a Droga , Feminino , Hormônio Foliculoestimulante/farmacocinética , Hormônio Foliculoestimulante/farmacologia , Ovariectomia
19.
J Endocrinol ; 141(1): 113-21, 1994 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-8014594

RESUMO

A dose-finding study was performed in adult male monkeys (Macaca fascicularis) to evaluate the pharmacokinetics and pharmacodynamics of a recombinant human FSH preparation (rhFSH). Groups of five monkeys were randomly assigned to receive single i.m. injections of 0.9% (w/v) NaCl (diluent), 6, 12 or 24 IU rhFSH/kg or 24 IU urinary human FSH/kg (uhFSH). The doses were based on an in vivo ovarian weight gain assay. Blood samples were collected 24 h before and immediately prior to injections, and 4, 8, 12, 24, 72 and 96 h after injections for determination of serum levels of immunoactive FSH by fluoroimmunoassay, bioactive FSH by an in vitro Sertoli cell assay, and inhibin and testosterone by radioimmunoassay. Inhibin was chosen as a marker for in vivo hFSH activity, since the secretion of inhibin in male monkeys is under the control of FSH. Administration of hFSH resulted in dose-related increases in serum hFSH concentrations. rhFSH and uhFSH exhibited similar pharmacokinetics. Comparable findings were obtained when serum samples were analysed for in vitro FSH bioactivity. Maximum serum hFSH levels were obtained 4-6 h after administration and the elimination half-life of hFSH was on average 18-22 h. The serum pharmacokinetics of rhFSH were linear within the dose range explored. Baseline inhibin concentrations varied significantly between groups. However, when the changes in inhibin concentrations were normalized to the baseline values (per cent change, area under curve and maximum inhibin level), a dose-dependent stimulatory effect of rhFSH on serum inhibin was evident.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Hormônio Foliculoestimulante/farmacocinética , Macaca fascicularis/metabolismo , Animais , Bioensaio , Relação Dose-Resposta a Droga , Fluorimunoensaio , Hormônio Foliculoestimulante/sangue , Hormônio Foliculoestimulante/farmacologia , Hormônio Foliculoestimulante/urina , Meia-Vida , Humanos , Inibinas/sangue , Masculino , Radioimunoensaio , Distribuição Aleatória , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/farmacologia , Testículo/efeitos dos fármacos , Testosterona/sangue
20.
J Endocrinol ; 158(3): 441-8, 1998 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9846174

RESUMO

We have previously described the preparation, purification and partial characterization of recombinant (rec) forms of rat luteinizing hormone (LH) and follicle-stimulating hormone (FSH). In the present study, the special functional features of these hormones were studied further, in vitro and in vivo, and compared with human recLH and recFSH, as well as with human urinary choriongonadotropin (hCG) and rat pituitary LH (NIDDK-RP3). In radioreceptor assay, the affinity of hCG binding to rat testis membranes was 5-fold higher than that of human recLH and 100-fold higher than that of rat recLH. In in vitro bioassay, using dispersed adult mouse interstitial cells or a mouse Leydig tumor cell line (BLT-1), hCG and human recLH were 10- to 20-fold more potent than rat recLH. Correspondingly, rat pituitary LH was about 10-fold less potent than rat recLH, and evoked a maximum testosterone response that was about half of that elicited by the other LH/CG preparations. Rat recFSH was about 10-fold less potent than human recFSH in stimulating cAMP production of a mouse Sertoli cell line (MSC-1) expressing the recombinant rat FSH receptor. The circulating half-times (T1/2) of rat and human rec hormones were assessed after i.v. injections into adult male rats rendered gonadotropin-deficient by treatment with a gonadotropin-releasing hormone antagonist. A novel immunometric assay was used for the rat FSH measurements. In the one-component model the T1/2 values of rat and human recLH were 18.2 +/- 1.9 min (n = 7) and 44.6 +/- 3.1 min (n = 7) respectively and those of rat and human recFSH were 88.4 +/- 10.7 min (n = 6) and 55.0 +/- 4.2 min (n = 6) respectively; the two-component models revealed similar differences between the rec hormone preparations. Collectively, rat recLH was eliminated significantly faster from the circulation than human recLH (P < 0.0001). In contrast, the elimination of rat recFSH was significantly slower than that of human recFSH (P = 0.02). In conclusion, rat recFSH and rat recLH display lower biopotencies per unit mass than the respective human hormones in vitro, and also in vivo for LH. This is paralleled by shorter T1/2 of rat recLH than the respective human hormone in the circulation, whereas human recFSH has a shorter T1/2 than human FSH. The special functional features of the rat rec gonadotropins emphasize the use of these preparations on studies of gonadotropin function in the rat, an important animal model for reproductive physiology.


Assuntos
Hormônio Foliculoestimulante/farmacologia , Hormônio Luteinizante/farmacologia , Animais , Bioensaio , Gonadotropina Coriônica/farmacologia , AMP Cíclico/biossíntese , Hormônio Foliculoestimulante/farmacocinética , Meia-Vida , Células Intersticiais do Testículo/efeitos dos fármacos , Células Intersticiais do Testículo/metabolismo , Hormônio Luteinizante/farmacocinética , Masculino , Camundongos , Camundongos Endogâmicos , Ensaio Radioligante , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/farmacologia , Células de Sertoli/efeitos dos fármacos , Células de Sertoli/metabolismo , Especificidade da Espécie , Testosterona/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA