Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Zhonghua Nan Ke Xue ; 29(9): 810-814, 2023 Sep.
Artigo em Chinês | MEDLINE | ID: mdl-38639593

RESUMO

OBJECTIVE: To evaluate the therapeutic effects of Xuanju compound capsule combined with urofollitropin (uFSH) in the treatment of idiopathic oligoasthenozoospermia. METHODS: From June 2022 to June 2023, patients with idiopathic oligoastheospermia were enrolled in this study, and divided into trail group (Xuanju compound capsule combined with urofollitropin tablets, n=53) and control group (urofollitropin tablets, n=61) according to the difference in treatment. Treatment methods: Xuanju compound capsule, 3 pills, three times a day; Urofollitropin, 75IU, one times three day. The curses of treatments for control group and trail group is 12 weeks. In order to evaluate the therapeutic effects of control group and trial group, semen volume, sperm concentration, progressive sperm ratio (PR), peripheral serum sex hormone, liver functions were analyzed before and after treatment for two times. RESULTS: Compared with the baseline, the semen volume and liver function were not significantly changed after the treatment in control group and trial group. However, sperm concentration, PR, testosterone (T) levels, follicle stimulating hormone (FSH) levels, and luteinizing hormone (LH) levels were significantly unregulated after the treatment in control group and trial group. More importantly, compared to control group, sperm concentration, PR, T leves, FSH levels, LH levels, and T/E2 ratio of trial group were further enhanced after the treatment, which were statistically significant (P < 0.05). CONCLUSIONS: Xuanju compound capsule combined with urofollitropin tablets could significantly improve the semen quality, up-regulate the testosterone levels and T/E2 ratio in patients with idiopathic oligoasthenozoospermia.


Assuntos
Urofolitropina , Humanos , Masculino , Hormônio Foliculoestimulante , Sêmen , Análise do Sêmen , Contagem de Espermatozoides , Testosterona , Resultado do Tratamento , Urofolitropina/uso terapêutico
2.
Int J Mol Sci ; 21(19)2020 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-32992491

RESUMO

Different Follicle Stimulating Hormone (FSH) formulation and Luteinizing Hormone (LH) are used in Assisted Reproductive Technology (ART) to induce follicles development and oocytes maturation, but it is still under debate which protocol is to be preferred. In the present study, the different effects on cumulus cells (CCs) of three controlled ovarian stimulation (COS) protocols, based on urinary FSH, recombinant FSH, or human Menopausal Gonadotropin (hMG) administration, were assessed. CCs were obtained from 42 normal-responders women undergoing COS, randomly divided into three groups according to the used gonadotropin formulation. Differences were found in the expression of genes belonging to the endocannabinoid system (the receptors CNR1, CNR2 and TRPV1, and the enzymes involved in the metabolisms of anandamide, NAPE-PLD and FAAH, and 2-acylglycerol, DAGL and MAGL); consistently, changes in lipid (PPARα, and FASN) and carbohydrate (GLUT1 and GLUT9) metabolisms, in CCs' macromolecules composition (highlighted by Fourier Transform Infrared Microspectroscopy, FTIRM), and in the number of retrieved oocytes were found. For the first time, statistically significant evidence on the differences related to each COS protocol on the endocannabinoid system, metabolism and macromolecular composition of CCs was found, representing a proof of concept to be further confirmed in a larger cohort of patients.


Assuntos
Células do Cúmulo/efeitos dos fármacos , Células do Cúmulo/metabolismo , Endocanabinoides/metabolismo , Hormônio Foliculoestimulante Humano/farmacologia , Menotropinas/farmacologia , Indução da Ovulação/métodos , Transdução de Sinais/efeitos dos fármacos , Urofolitropina/farmacologia , Adulto , Ácidos Araquidônicos/genética , Ácidos Araquidônicos/metabolismo , Células Cultivadas , Estudos de Coortes , Endocanabinoides/genética , Feminino , Expressão Gênica/efeitos dos fármacos , Humanos , Recuperação de Oócitos , Alcamidas Poli-Insaturadas/metabolismo , Proteínas Recombinantes/farmacologia , Espectroscopia de Infravermelho com Transformada de Fourier
3.
Endocrine ; 66(2): 381-385, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31240459

RESUMO

INTRODUCTION: A reduction of testicular volume (TV) represents an important clinical sign, which may hide sperm abnormalities and predispose to hypogonadism. AIM: The primary purpose of this study was to evaluate the serum levels of total testosterone after treatment with urofollitropin in selected patients with male infertility and idiopathic mild reduction of testicular volume. METHODS: In this 1-year-long prospective design, patients with abnormal sperm parameters, mild reduction in TV (8-12 mL) and normal gonadotropin, and total testosterone (TT) serum levels were recruited in this study. Patients treated for 4 months with urofollitropin were included in group A, those treated with intracytoplasmatic sperm injection due to a female-factor infertility were included in group B. Hormone values, sperm parameters, and TV were detected at baseline (T0), after 4 (T1) and 12 months (T2) in group A and at T0 and T2 in group B. RESULTS: Group A (n = 80) showed increased follicle-stimulating hormone (FSH) at T1 and sperm morphology at T1 and T2 compared to T0 (all p < 0.05). Group B (n = 50) had lower TT and higher FSH levels at T2 compared to T0 (all p < 0.05). At T2, TT, VT, total sperm count, progressive motility, total motility, and sperm morphology were higher in group A compared to group B (all p < 0.05). CONCLUSION: Reduced TV may predispose to infertility and hypogonadism. FSH treatment may improve Sertoli and Leydig cell function and prevent the development of hypogonadism.


Assuntos
Infertilidade Masculina/tratamento farmacológico , Motilidade dos Espermatozoides/efeitos dos fármacos , Espermatozoides/efeitos dos fármacos , Testículo/diagnóstico por imagem , Testosterona/sangue , Urofolitropina/uso terapêutico , Adulto , Hormônio Foliculoestimulante/sangue , Humanos , Infertilidade Masculina/sangue , Infertilidade Masculina/diagnóstico por imagem , Estudos Longitudinais , Hormônio Luteinizante/sangue , Masculino , Tamanho do Órgão , Estudos Prospectivos , Contagem de Espermatozoides , Ultrassonografia , Urofolitropina/administração & dosagem
4.
Urologia ; 85(1): 19-21, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28799634

RESUMO

INTRODUCTION: Recent evidences demonstrated that male factor alone is responsible for about 30% cases of infertility. Human follicle-stimulating hormone (hFSH) has been introduced to increase sperm concentration, spermatogonial population, or both natural or assisted pregnancy rates (PRs) in oligozoospermic subjects with normal concentrations of gonadotropins. METHODS: Fifty infertile men affected by idiopathic azoospermia were enrolled in this study, after undergoing medical history, physical and clinical examination, baseline semen parameters and hormonal plasma concentrations. Inclusion criteria were infertility for at least 2 years, idiopathic azoospermia, FSH <12 mIU/ml. Twenty-five patients were allocated to treatment with hFSH three times/week per 3 months (Fostimon), and 25 patients underwent just testicular sperm extraction (TESE) without medical treatment. All patients underwent, after 3 months, assisted reproduction techniques (ARTs) with TESE. The primary outcome was represented by the differences in the sperm retrieval rate (SRR) between groups, while the secondary outcomes were the differences in PR and fertilization rate (FR). RESULTS: We observed a PR of 15% (3/25) and 28% (7/25) in control and treated group, respectively. SRR after medical treatment and ART was 24% (6/25), while in the control group was 12.5% (2/25). The sperm in the ejaculate of five patients (20%) after medical treatment exhibited a mean concentration of 0.9 million/ml and a mean motility of 12%. The FR was significantly greater in the treatment group with respect to the control group, 30% and 20%, respectively. CONCLUSIONS: FSH treatment showed greater efficacy rather than control by increasing the rate of PR and FR in azoospermic patients who underwent TESE.


Assuntos
Azoospermia/tratamento farmacológico , Técnicas de Reprodução Assistida , Recuperação Espermática , Urofolitropina/uso terapêutico , Adulto , Azoospermia/diagnóstico , Estudos de Casos e Controles , Feminino , Humanos , Masculino , Gravidez , Taxa de Gravidez
5.
Arch Gynecol Obstet ; 295(1): 233-238, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27604242

RESUMO

PURPOSE: Severe pre-eclampsia affects maternal health with long-term consequences. It is postulated that during the process of implantation and cell differentiation, embryos resulting from the fertilization of ageing oocytes produce malfunctioning trophoectoderm leading to placental dysfunction. Therefore, severe pre-eclampsia may be associated with a decreased ovarian reserve. The objective of this study was to compare serum markers of ovarian reserve and function between women who had severe pre-eclampsia and those who had normal pregnancies. METHODS: Twenty women who had severe pre-eclampsia (PE) and 20 who had uncomplicated pregnancies (controls) matched for age and body mass index were included in the study. Fasting blood samples were taken during the follicular phase (day 5) of the menstrual cycle 6 months to 5 years after the delivery. Serum was separated and frozen at -70 °C until analyzed for anti-Mϋllerian hormone (AMH), total and free testosterone (TT), free-androgen index (FAI), luteinizing hormone (LH), and follicle-stimulating hormone (FSH) hormone to evaluate ovarian reserve and function, and the results were compared between two groups. RESULTS: The median AMH was 0.91 ng/mL in PE group compared to 0.72 ng/mL in controls (p = 0.995). No significant differences were found between the two groups in the levels of LH (5.65 vs. 5.4 IU/L, respectively, p = 0.897) and FSH (4.95 vs. 5.1 IU/L, respectively, p = 0.523). However, total and free-TT levels as well as FAI were significantly lower in the PE group compared to controls (p = 0.017, p = 0.006, and p = 0.011, respectively). CONCLUSIONS: Ovarian reserve and function are not altered significantly in women with a previous history of pre-eclampsia compared with women who had an uncomplicated pregnancy.


Assuntos
Oócitos , Reserva Ovariana/fisiologia , Pré-Eclâmpsia/epidemiologia , Adulto , Hormônio Antimülleriano/sangue , Estudos de Casos e Controles , Feminino , Hormônio Foliculoestimulante/sangue , Humanos , Hormônio Luteinizante/sangue , Ciclo Menstrual , Gravidez , Estudos Prospectivos , Testosterona/sangue , Urofolitropina/sangue
6.
J Heart Lung Transplant ; 36(1): 59-63, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27773451

RESUMO

BACKGROUND: Serum lactate dehydrogenase (LDH) is the standard measure for detection of hemolysis and thus surveillance for device thrombosis in patients on continuous-flow left ventricular assist device (CF-LVAD) support. Significant hemolysis has been defined as LDH ≥600 IU/L. However, LDH testing requires phlebotomy, precluding frequent home monitoring. Simple dipstick urinalysis (UA) for urine hemoglobin (U-Hb) overcomes this limitation. This study correlated U-Hb and LDH levels and evaluated the performance of UA for detection of significant hemolysis in patients with CF-LVADs. METHODS: U-Hb and LDH were measured concurrently 956 times in 221 patients with CF-LVADs. Statistics were computed to determine accuracy of UA in detecting LDH ≥600 IU/L, with a positive result being any detected U-Hb. All analyses were performed with and without excluding for 1) conditions associated with tissue damage, which are known to increase LDH, and 2) suspected or confirmed urinary tract infections or hematuria, which are known to cause hemoglobinuria for reasons other than hemolysis. RESULTS: Mean LDH for absent/mild/severe U-Hb was 360 IU/L/467 IU/L IU/L/777 IU/L without exclusions, 354 IU/L/444 IU/L IU/L/651 IU/L after excluding non-hemolytic LDH elevations, 370 IU/L/513 IU/L IU/L/1,357 IU/L after excluding urinary tract infections and hematuria, and 367 IU/L/470 IU/L IU/L/1,217 IU/L when both exclusions applied (all p < 0.001). Absent U-Hb had a negative predictive value for LDH ≥600 IU/L of >90% for all analyses. CONCLUSIONS: Serum LDH is significantly associated with U-Hb levels. Absence of U-Hb appears to efficiently exclude significant hemolysis in patients with CF-LVADs. Because it can be performed by patients at home, hemoglobinuria monitoring may enable more intense surveillance and earlier diagnosis of device thrombosis.


Assuntos
Insuficiência Cardíaca/terapia , Coração Auxiliar/efeitos adversos , Hemoglobinas/metabolismo , Hemólise/fisiologia , Trombose/urina , Biomarcadores/sangue , Biomarcadores/urina , Falha de Equipamento , Feminino , Insuficiência Cardíaca/mortalidade , Insuficiência Cardíaca/urina , Humanos , Incidência , Estimativa de Kaplan-Meier , L-Lactato Desidrogenase/sangue , Masculino , Pessoa de Meia-Idade , New York/epidemiologia , Reprodutibilidade dos Testes , Estudos Retrospectivos , Fatores de Risco , Taxa de Sobrevida/tendências , Trombose/diagnóstico , Trombose/etiologia , Urinálise , Urofolitropina
7.
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
8.
Fertil Steril ; 104(6): 1419-25, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26361207

RESUMO

OBJECTIVE: To compare the outcomes of controlled ovarian stimulation/in vitro fertilization cycles using 450 IU and 600 IU gonadotropin per day in women at risk of poor ovarian response. DESIGN: Prospective randomized controlled nonblinded study. SETTING: University-affiliated private IVF center. PATIENT(S): Women considered to be at risk of poor ovarian response: aged <41 years with basal FSH >10 IU/L, antimüllerian hormone <1 ng/mL, antral follicle count ≤ 8, or a previous IVF cycle with ≥ 300 IU/d gonadotropin that resulted in a cancellation, <8 follicles, or <5 oocytes. INTERVENTION(S): A total of 356 patients underwent a microdose GnRH agonist flare-up IVF/intracytoplasmic sperm injection protocol with a fixed daily dose of either 450 IU FSH (n = 176) or 600 IU FSH (n = 180) equally divided between Menopur and Bravelle. MAIN OUTCOME MEASURE(S): Number of mature oocytes retrieved. RESULT(S): The two groups were similar in terms of age, ovarian reserve, cause of infertility, duration of stimulation, and cycle cancellation rate. There were no significant differences in the number of metaphase II oocytes retrieved (4 [range 0-6] vs. 4 [range 2-7]), fertilization rate (62.4% vs. 57.0%), biochemical pregnancy rate (20.5% vs. 22.9%), clinical pregnancy rate (16.4% vs. 18.3%), and implantation rate (29.8% vs. 30.4%) between the 450 IU and 600 IU groups, respectively. CONCLUSION(S): Gonadotropin of 600 IU/d does not improve outcome of IVF cycles compared with 450 IU/d in women at risk of poor ovarian response. CLINICAL TRIAL REGISTRATION NUMBER: NCT00971152.


Assuntos
Fármacos para a Fertilidade Feminina/administração & dosagem , Hormônio Foliculoestimulante/administração & dosagem , Infertilidade/terapia , Menotropinas/administração & dosagem , Ovário/efeitos dos fármacos , Indução da Ovulação/métodos , Ovulação/efeitos dos fármacos , Urofolitropina/administração & dosagem , Adulto , Implantação do Embrião , Transferência Embrionária , Feminino , Fármacos para a Fertilidade Feminina/efeitos adversos , Fertilização In Vitro , Hormônio Foliculoestimulante/efeitos adversos , Humanos , Infertilidade/diagnóstico , Infertilidade/fisiopatologia , Menotropinas/efeitos adversos , Recuperação de Oócitos , Ovário/fisiopatologia , Indução da Ovulação/efeitos adversos , Gravidez , Taxa de Gravidez , Estudos Prospectivos , Quebeque , Injeções de Esperma Intracitoplásmicas , Resultado do Tratamento , Urofolitropina/efeitos adversos
9.
J Clin Endocrinol Metab ; 100(6): 2449-55, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25825944

RESUMO

CONTEXT: Gonadotropin therapy using a human chorionic gonadotropin (hCG) and FSH preparation is an effective regimen in inducing masculinization and spermatogenesis in men with idiopathic hypogonadotropic hypogonadism (IHH). However, the high cost of medication and frequent injections affect compliance. OBJECTIVE: The aim of this study was to determine the efficacy of sequential use of highly purified urinary FSH (uFSH)/hCG in men with IHH. DESIGN AND SETTING: A randomized, open-label, prospective, controlled noninferiority trial with an 18-month follow-up was conducted in 9 tertiary hospitals. PATIENTS AND INTERVENTION: A total of 67 Chinese men with IHH were randomly allocated into group A receiving continual uFSH (75 U, 3 times a week) and hCG (2000 U, twice a week) injection and group B receiving sequential uFSH (75 U, 3 times a week every other 3 months) and hCG (2000 U, twice a week) injection. MAIN OUTCOME MEASURE: The primary outcome was the proportion of subjects with a sperm concentration of ≥ 1.0 × 10(6)/mL during the 18 months. The efficacy between groups A and B was compared for noninferiority. RESULTS: Of the patients, 17/33 (51.5%) receiving continual uFSH/hCG and 19/34 (55.9%) receiving sequential uFSH/hCG achieved sperm concentrations of ≥ 1.0 × 10(6)/mL. The efficacy in the sequential uFSH/hCG group was not inferior to that in the continual uFSH/hCG group (noninferiority, P = .008) by intention-to-treat analysis. CONCLUSIONS: The efficacy of the sequential uFSH/hCG regimen is not inferior to that of the continual uFSH/hCG regimen in inducing spermatogenesis and masculinization of patients with IHH.


Assuntos
Gonadotropina Coriônica/administração & dosagem , Hipogonadismo/tratamento farmacológico , Urofolitropina/administração & dosagem , Adolescente , Adulto , Gonadotropina Coriônica/isolamento & purificação , Gonadotropina Coriônica/urina , Esquema de Medicação , Quimioterapia Combinada , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Contagem de Espermatozoides , Espermatogênese/efeitos dos fármacos , Testículo/efeitos dos fármacos , Urofolitropina/isolamento & purificação , Adulto Jovem
10.
Reprod Biomed Online ; 29(4): 509-15, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25131556

RESUMO

The human androgen receptor (AR) gene contains a highly polymorphic CAG repeat sequence within exon 1. In-vitro studies have shown a relationship between CAG repeats in the AR gene and its transactivation potential. This variation in length may play a role in anovulatory infertility. The objective of this study was to investigate whether CAG polymorphism of the AR gene has a predictive value for ovarian reserve, response and cycle outcome in an egg donor programme. CAG length of the AR gene was determined in 147 oocyte donors. All donors underwent ovarian stimulation with a gonadotrophin-releasing hormone antagonist protocol (n = 355). No differences were reported in days of stimulation, gonadotrophin doses, and number of oocytes retrieved. Clinical outcomes were not affected by the CAG repeat length of the AR gene; the primary end-point, antral follicle count, was significantly affected (P < 0.05). In conclusion, in a population of fertile egg donors AR gene CAG polymorphism does not affect ovarian response to gonadotrophins. Antral follicle count was associated with the CAG polymorphism genotype. This suggests that genetic factors may increase susceptibility to poor ovarian reserve, and that AR gene genotype could play a role in the natural ovarian ageing process.


Assuntos
Fármacos para a Fertilidade Feminina/farmacologia , Reserva Ovariana , Ovário/efeitos dos fármacos , Indução da Ovulação , Polimorfismo Genético , Receptores Androgênicos/genética , Expansão das Repetições de Trinucleotídeos , Adolescente , Adulto , Feminino , Estudos de Associação Genética , Hormônio Liberador de Gonadotropina/análogos & derivados , Hormônio Liberador de Gonadotropina/farmacologia , Humanos , Infertilidade Feminina/genética , Infertilidade Feminina/patologia , Doação de Oócitos , Ovário/citologia , Ovário/diagnóstico por imagem , Ovário/patologia , Receptores Androgênicos/química , Receptores Androgênicos/metabolismo , Estudos Retrospectivos , Espanha , Pamoato de Triptorrelina/farmacologia , Ultrassonografia , Urofolitropina/farmacologia , Adulto Jovem
11.
Arch Gynecol Obstet ; 288(4): 901-4, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23545834

RESUMO

PURPOSE: In the GnRH-antagonist protocol, ovarian stimulation with gonadotropins typically commences on cycle day 2 or 3. Initiation of ovarian stimulation with a spontaneously occurring menstruation, however, poses significant organizational challenges for treatment centres and patients alike. It has previously been demonstrated in the context of fertility preservation that initiation of stimulation in the luteal phase is feasible in terms of retrieval of mature oocytes for cryopreservation. Herein, we report the extension of this concept to a routine IVF setting with the aim of establishing an ovarian stimulation protocol, which can be utilized independent of menstruation. Because of asynchrony of endometrium and embryo in such a setting, all fertilized oocytes have to be cryopreserved for a later transfer. METHODS: This was a prospective, case-control study (trial registration: NCT00795041) on the feasibility of starting ovarian stimulation in a GnRH-antagonist protocol in the luteal phase. Inclusion criteria were: IVF or ICSI; 18-36 years; ≤3 previous IVF/ICSI attempts; BMI 20-30 kg/m(2); regular cycle (28-35 days); luteal phase progesterone >7 ng/ml at initiation of stimulation. Exclusion criteria were: PCOS, endometriosis ≥AFS III°, unilateral ovary, expected poor response. Stimulation was performed with highly purified uFSH (Bravelle®) 300 IU/day and 0.25 mg/day GnRH-antagonist starting on cycle day 19-21 of a spontaneous menstrual cycle and commencing until hCG administration when three follicles ≥17 mm were present. All 2PN stage oocytes were vitrified for later transfers in programmed cycles. Feasibility was defined as the achievement of ongoing pregnancies progressing beyond the 12th gestational week in at least 2/10 study subjects (primary outcome). Secondary outcomes were gonadotropin consumption per oocyte obtained, stimulation duration, and fertilization rates. Study subjects were matched in a 1:3 ratio with concomitantly treated control cases of similar age, BMI, and duration of infertility who were treated in a conventional GnRH-antagonist protocol with 150-225 rFSH or HP-HMG/day. RESULTS: The study group consisted of ten subjects, mean age 31.4 years, BMI 25.4 kg/m(2), of which one had fertilization failure. Mean stimulation duration was 11.7 (SD 1.6) vs. 9.1 (SD 1.3) days, mean cumulative FSH dose was 3,495.0 (SD 447.5) vs. 2,040.5 (SD 576.2) IU, and mean number of oocytes was 8.8 (SD 5.0) vs. 10.0 (SD 5.4) in study vs. control group, respectively. Per follicle ≥10 mm, the cumulative FSH dose was 274.5 (SD 130.8) IU vs. 245.2 (SD 232.3) IU in study and control groups, respectively. Cumulative ongoing pregnancy rates were 1/10 (10 %) and 6/30 (20.0 %) in study and control group, respectively (difference: 10 %, 95 % confidence interval of the difference: -29.2-22.2 %, p = 0.47). Fertilization rate was similar between groups, with 63.5 % (SD 32.9) in the study and 61.3 % (SD 26.7) in the control group, respectively. Serum estradiol levels were significantly lower on the day of triggering final oocyte maturation with 1,005.3 (SD 336.2) vs. 1,977.4 pg/ml (SD 1,106.5) in study and control group, respectively. Similarly, peak estradiol biosynthesis per growing follicle ≥10 mm was lower in the study group (134 pg/ml, SD 158.4 vs. 186.7 pg/ml, SD 84.7). CONCLUSIONS: Per retrieved oocyte, a nearly threefold higher dose of FSH had to be administered when ovarian stimulation had been initiated in the luteal phase. Furthermore, the present study casts doubt on the efficacy of initiating ovarian stimulation in the luteal phase in terms of pregnancy achievement. Thus, this concept is currently not feasible for routine use, and it should also be explored further before using it at larger scale in the context of emergency stimulation for fertility preservation.


Assuntos
Fertilização In Vitro/métodos , Hormônio Liberador de Gonadotropina/antagonistas & inibidores , Infertilidade Feminina/terapia , Indução da Ovulação/métodos , Urofolitropina/administração & dosagem , Adolescente , Adulto , Esquema de Medicação , Quimioterapia Combinada , Estudos de Viabilidade , Feminino , Humanos , Fase Luteal , Recuperação de Oócitos , Gravidez , Taxa de Gravidez , Estudos Prospectivos , Resultado do Tratamento , Adulto Jovem
12.
Gynecol Endocrinol ; 28(2): 94-101, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21591977

RESUMO

BACKGROUND: Randomized trials (RCTs) and systematic reviews have challenged the claim for superiority of recombinant follicle-stimulating hormone (recFSH) compared with human-derived FSH (hFSH). Even so, much of the evidence comes from unavailable products. If the efficacy of the currently available Fostimon(®) is superior, the off-market Metrodin-HP(®), then data from the latter should not be used, gauge of the former. METHODS: Electronic/hand searches were performed to identify RCTs comparing Fostimon(®) vs. Metrodin-HP(®) or either product with recFSH. Primary outcomes were live-birth rate (LBR), ongoing pregnancy rate (OPR), and OPR/LBR. Secondary outcomes were clinical pregnancy rate (CPR), multiple pregnancy rate (MPR), ovarian hyperstimulation syndrome (OHSS), abortion rates, and cycle demographics. Data were extracted, allowed for an intention-to-treat analysis, and meta-analyzed using combined direct/adjusted indirect methods. RESULTS: Twenty-two RCTs met the inclusion criteria: Fostimon(®) vs. Metrodin-HP(®) (n = 2); Fostimon(®) vs. recFSH (n = 8); and Metrodin-HP(®) vs. recFSH (n = 12). LBR (odds ratio = 1.72; 95% confidence interval = 1.05-2.80), OPR/LBR, and CPR were all significantly higher favoring Fostimon(®). OPR, MPR, OHSS, and miscarriage rates were not significantly different. Pooled results for cycle demographics were not reported due to high heterogeneity. Conclusions. Fostimon(®) is superior to Metrodin-HP(®) regarding clinical outcomes. Therefore, care should be taken not to assume that all hFSH products have the same efficacy.


Assuntos
Hormônio Foliculoestimulante Humano/normas , Feminino , Fertilização In Vitro , Humanos , Gravidez , Resultado da Gravidez , Taxa de Gravidez , Ensaios Clínicos Controlados Aleatórios como Assunto , Injeções de Esperma Intracitoplásmicas , Resultado do Tratamento , Urofolitropina/normas
13.
Gynecol Endocrinol ; 28(5): 345-50, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22115012

RESUMO

An observational, matched, case-control study was carried out to compare the efficacy of recombinant human luteinizing hormone (r-hLH) supplementation with that of urinary human menopausal gonadotrophin (u-hMG)-based LH activity during controlled ovarian stimulation (COS) for assisted reproductive technology (ART) using a long gonadotrophin-releasing hormone (GnRH)-agonist protocol. A total of 4719 women, 1573 per group, matched by age, body mass index, indication and number of previous ART cycles, were treated with either recombinant human follicle-stimulating hormone (r-hFSH) and r-hLH in a fixed 2:1 ratio or u-hMG, either alone or in combination with r-hFSH, after down-regulation in a long GnRH-agonist protocol. Compared with the two u-hMG groups (u-hMG alone or in combination with r-hFSH, respectively), r-hFSH consumption was significantly lower (p < 0.001; p < 0.001), and pregnancy rates per cycle (p = 0.006; p = 0.022) and per embryo transfer (p = 0.025; p = 0.008), and implantation rate per embryo transferred (p < 0.001; p < 0.001) were significantly higher in the group treated with the fixed combination of r-hFSH and r-hLH. In COS protocols with r-hFSH, supplementation with r-hLH appears to be more effective than supplementation with u-hMG using the long GnRH-agonist protocol for ART.


Assuntos
Hormônio Foliculoestimulante Humano/administração & dosagem , Hormônio Luteinizante/administração & dosagem , Indução da Ovulação/métodos , Urofolitropina/administração & dosagem , Adulto , Estudos de Casos e Controles , Feminino , Hormônio Liberador de Gonadotropina/agonistas , Humanos , Masculino , Gravidez , Taxa de Gravidez , Proteínas Recombinantes/administração & dosagem
14.
Fertil Steril ; 94(6): 2332-4, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20188364

RESUMO

In a randomized study comparing purified urinary FSH with recombinant FSH for IVF/intracytoplasmic sperm injection in patients with polycystic ovary syndrome, there was no significant difference between the mean total dose of FSH used, duration of stimulation, number of retrieved oocytes, number of mature oocytes, number of embryos transferred, or the ongoing pregnancy rate between the two groups. However, there were significantly more fertilized oocytes, a higher fertilization rate, more top-quality embryos, and more cryopreserved embryos in the urinary FSH group.


Assuntos
Fertilização In Vitro/métodos , Hormônio Foliculoestimulante/uso terapêutico , Infertilidade Feminina/tratamento farmacológico , Síndrome do Ovário Policístico/tratamento farmacológico , Urofolitropina/uso terapêutico , Adulto , Feminino , Hormônio Foliculoestimulante/efeitos adversos , Humanos , Infertilidade Feminina/etiologia , Síndrome de Hiperestimulação Ovariana/prevenção & controle , Indução da Ovulação/métodos , Síndrome do Ovário Policístico/complicações , Gravidez , Taxa de Gravidez , Proteínas Recombinantes/efeitos adversos , Proteínas Recombinantes/uso terapêutico , Injeções de Esperma Intracitoplásmicas/métodos , Resultado do Tratamento , Urofolitropina/efeitos adversos , Urofolitropina/isolamento & purificação , Adulto Jovem
15.
Mol Hum Reprod ; 16(3): 200-6, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19720661

RESUMO

The aim of the study was to investigate the effects of urinary follicle-stimulating hormone (FSH) compounds on the electrical activity of myometrium using signal-processing techniques. Thirty animals were involved in the experiment. After two successive normal estrous cycles, 15 of these animals were put into three equal subgroups. Group 1 was the control; animals were given solvent. Groups 2 and 3 were treated with Urofollitropin and Menotropin, respectively. The other 15 animals were ovariectomized and subjected to the same protocol. Their uterine myoelectrical signals were recorded over a period of at least 3 min at a sampling frequency of 500 Hz, and analyzed through software assisted signal processing. The results show the power and some characteristic spectral components of myoelectrical signal were differentially reduced with the administration of highly purified urinary FSH and human menopausal FSH but significant differences were not detected between their histology. In conclusion, uterine myoelectrical signals change with administration of urinary FSH preparations. Human menopausal FSH and more precisely highly purified FSH suppress the spectral components and modify the power of the myoelectrical signals which provides uterine quiescence.


Assuntos
Hormônio Foliculoestimulante/farmacologia , Miométrio/efeitos dos fármacos , Miométrio/fisiologia , Animais , Eletrofisiologia , Feminino , Humanos , Menotropinas/farmacologia , Ovariectomia , Ratos , Ratos Wistar , Urofolitropina/farmacologia
16.
J Reprod Med ; 54(8): 459-66, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19769189

RESUMO

OBJECTIVE: To understand the properties of each available gonadotropin preparation, especially in terms of the differences between urinary-derived and recombinant preparations. STUDY DESIGN: Human menopausal gonadotropin (hMG), highly purified urinary-derived follicle-stimulating hormone (uFSH-HP) and recombinant FSH (rFSH) were subjected to 2-dimensional gel electrophoresis (2-DE), and protein spots were visualized by silver-staining procedures. Major spots were analyzed by mass spectrometry. Fluorescent-labeled preparations were also subjected to 2-DE to evaluate the quantities of FSH isohormones contained in each preparation. RESULTS: 2-DE and mass spectrometry analyses of hMG identified many extracellular proteins as major impurities and several plasma membrane proteins including prion proteins. Both uFSH-HP and rFSH demonstrated slight impurities and showed several alpha and beta subunit isohormones. rFSH contained higher amounts of the basic isohormones of the alpha subunit than uFSH-HP, whereas the predominance of the basic isohormones was less significant in the beta subunit. CONCLUSION: Proteomic analyses demonstrated the detailed protein profiles of each preparation. Differences in the quantities of alpha subunit isohormones may contribute to the variations in FSH activity observed between recombinant and urinary-derived FSH preparations.


Assuntos
Subunidade beta do Hormônio Folículoestimulante/análise , Subunidade alfa de Hormônios Glicoproteicos/análise , Menotropinas/química , Urofolitropina/química , Eletroforese em Gel Bidimensional , Feminino , Humanos , Espectrometria de Massas , Proteínas Recombinantes/química
17.
Fertil Steril ; 91(4): 1005-11, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18367182

RESUMO

OBJECTIVE: To compare the efficacy of highly purified human urinary follicle stimulating hormone (HP-hFSH) versus human recombinant follitropin-alpha (rFSH) in volunteers undergoing controlled ovarian stimulation for IVF. DESIGN: A randomized, controlled, investigator-blind trial. SETTING: Four assisted reproductive technology centers. PATIENT(S): One hundred fifty-two IVF patients. INTERVENTION(S): Volunteers, aged 18-39, were randomized to HP-hFSH (n = 76) versus rFSH (n = 76) at a starting dose of 300 IU in down-regulated cycles. MAIN OUTCOME MEASURE(S): Number of oocytes, clinical pregnancy rate, and live birth rate with HP-hFSH versus rFSH. RESULT(S): The total IU of gonadotropin used did not differ between the two groups. There was no difference in number of oocytes retrieved with HP-hFSH (mean = 16.3) compared with rFSH (mean = 17.1), confidence interval (CI) of difference = -3.79 to +2.18. Clinical pregnancy rate, as defined by the presence of a gestational sac, was 48.7% (CI = 37.0%-60.4%) with HP-hFSH versus 44.7% (CI = 33.3%-56.6%) with rFSH (CI of difference = -11.9% to +19.8%). Live birth rate was 38.2% (29 of 76) in both groups (CI = 27.2%-50.0%), for a difference between groups of 0.0% (CI of the difference = -15.4% to +15.4%). CONCLUSION(S): There were no statistically significant differences in mean oocyte number, clinical pregnancy rate, or live birth rate between HP-hFSH versus rFSH.


Assuntos
Fertilização In Vitro/métodos , Hormônio Foliculoestimulante/uso terapêutico , Indução da Ovulação/métodos , Proteínas Recombinantes/uso terapêutico , Urofolitropina/uso terapêutico , Adolescente , Adulto , Algoritmos , Feminino , Experimentação Humana , Humanos , Infertilidade/terapia , Gravidez , Taxa de Gravidez , Método Simples-Cego , Resultado do Tratamento , Urofolitropina/isolamento & purificação , Adulto Jovem
18.
Fertil Steril ; 91(4): 1012-7, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18371963

RESUMO

OBJECTIVE: To assess the efficacy and safety of a vaginal progesterone (P(4)) insert (Endometrin) for luteal support for assisted reproductive technology (ART). DESIGN: Multicenter, randomized, open-label (assessor-blinded) phase III clinical trial. SETTING: Twenty-five U.S. ART centers. PATIENT(S): A total of 1,211 ART patients randomized to three groups: Endometrin 100 mg twice daily (n = 404), Endometrin 100 mg three times daily (n = 404), and P(4) 90 mg 8% gel daily (n = 403). INTERVENTION(S): In vitro fertilization and ET were performed according to site-specific protocols. The day after oocyte retrieval, Endometrin or vaginal P(4) gel was begun for luteal support and continued for up to 10 weeks of pregnancy. MAIN OUTCOME MEASURE(S): Biochemical, clinical, and ongoing pregnancy and live birth rates. RESULT(S): Pregnancy rates were high and similar in all treatment groups, with biochemical rates exceeding 50%, clinical and ongoing rates >or=40%, and live birth rates at 35%-38%. The adverse event profiles were similar across groups. CONCLUSION(S): Pregnancy rates and live birth rates for Endometrin (twice daily and three times daily) were high and similar to those for P(4) gel. The adverse event profiles for both were similar to that for P(4) gel and primarily due to IVF stimulation and oocyte retrieval. Endometrin was safe and well tolerated.


Assuntos
Fertilização In Vitro/métodos , Dispositivos Intrauterinos Medicados , Fase Luteal/efeitos dos fármacos , Menotropinas/administração & dosagem , Indução da Ovulação/métodos , Progesterona/administração & dosagem , Urofolitropina/administração & dosagem , Adolescente , Adulto , Combinação de Medicamentos , Feminino , Fármacos para a Fertilidade Feminina/administração & dosagem , Fármacos para a Fertilidade Feminina/efeitos adversos , Humanos , Dispositivos Intrauterinos Medicados/efeitos adversos , Menotropinas/efeitos adversos , Indução da Ovulação/efeitos adversos , Gravidez , Taxa de Gravidez , Progesterona/efeitos adversos , Progesterona/uso terapêutico , Método Simples-Cego , Resultado do Tratamento , Urofolitropina/efeitos adversos , Adulto Jovem
19.
Am J Health Syst Pharm ; 65(12): 1148-53, 2008 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-18541685

RESUMO

PURPOSE: A prospective observational study was conducted to identify risk factors for ovarian hyperstimulation syndrome (OHSS) in Thai patients using gonadotropins for in vitro fertilization. METHODS: Outpatients receiving a short and a long protocol of ovarian stimulation with a recombinant follicle-stimulating hormone (rec FSH) at a Bangkok hospital were enrolled. Patients in the short protocol received rec FSH and the gonadotropin-releasing-hormone agonist buserelin acetate by subcutaneous injection after blood sampling on day 2 of the patient's cycle. When one or more follicles reached a diameter of 18 mm as determined by ultrasonography and the serum estradiol (E(2)) concentration exceeded 400 pg/mL, patients received human chorionic gonadotropin (hCG); oocytes were retrieved 34-36 hours later. The long protocol differed only in that patients received nasal buserelin acetate and rec FSH 10 days before the cycle started and on day 2 of the cycle. Serum levels of E(2), luteinizing hormone, and FSH were monitored at baseline. In addition to E(2) concentration and follicle size and number, patients' weight, waist circumference, complete blood counts, and C-reactive protein (CRP) levels were monitored at intervals. Patients were instructed to notify the hospital of any symptoms of OHSS. RESULTS: Of 117 patients enrolled, 13 had moderate OHSS and 1 had severe OHSS; all recovered. Patients with OHSS were significantly younger, had significantly lower body mass index, and had significantly higher E(2) before hCG injection, total number of follicles, total number of oocytes retrieved, and white blood cell and neutrophil counts after hCG injection. Patients with polycystic ovary syndrome were significantly more likely to have OHSS. CRP levels were higher in OHSS patients, but the difference was not significant. Multiple logistic regression showed that the combination of serum E(2) peak concentration of > or =4500 pg/mL and total number of oocytes retrieved of > or =15 predicted the occurrence of moderate-to-severe OHSS; 12 of 14 study patients who met these criteria had OHSS. CONCLUSION: Serum E(2) peak concentrations of > or =4500 pg/mL and total number of oocytes > or =15 may be useful indicators for identifying patients at high risk for moderate-to-severe OHSS.


Assuntos
Fertilização In Vitro/métodos , Gonadotropinas/uso terapêutico , Síndrome de Hiperestimulação Ovariana/etiologia , Adulto , Assistência Ambulatorial , Busserrelina , Feminino , Fármacos para a Fertilidade Feminina , Gonadotropinas/administração & dosagem , Gonadotropinas/farmacologia , Humanos , Observação , Síndrome de Hiperestimulação Ovariana/diagnóstico , Síndrome de Hiperestimulação Ovariana/epidemiologia , Síndrome de Hiperestimulação Ovariana/fisiopatologia , Estudos Prospectivos , Fatores de Risco , Tailândia/epidemiologia , Urofolitropina
20.
Prog. obstet. ginecol. (Ed. impr.) ; 51(5): 281-300, mayo 2008. tab
Artigo em Espanhol | IBECS | ID: ibc-139883

RESUMO

En el mercado español se dispone de diversas gonadotropinas utilizadas en los programas de reproducción asistida. El objetivo de esta revisión es determinar las diferencias entre ellas y establecer las ventajas y los inconvenientes de cada una en base a su origen, seguridad y eficacia clínica. Desde el punto de vista técnico, las gonadotropinas recombinantes presentan ventajas técnicas y mayor pureza, actividad específica y homogeneidad entre lotes. Desde el punto de vista de la seguridad, aunque hay diferencias claras en el origen, y por tanto en los riesgos de transmisión de enfermedad infecciosa, todas las gonadotropinas disponibles han mostrado ser seguras. Es desde el punto de vista de la eficacia donde es más difícil establecer diferencias. Muchos de los estudios disponibles son pequeños y no siempre los parámetros de evaluación han sido comparables. La mayoría de los estudios no han podido establecer diferencias, por lo que existen en la literatura científica diversos metaanálisis que tratan de responder básicamente a dos aproximaciones: saber si las FSH recombinantes (FSHr) son mejores que las urinarias y si la hormona recombinante es mejor que la gonadotropina menopáusica humana. Los datos no permiten demostrar que las urinarias o la hMG, solas o en combinación, sean más eficaces que la FSHr, mientras que, por el contrario, las recombinantes han mostrado ser más eficaces que las urinarias purificadas agrupadas (AU)


In Spain, several different gonadotrophins are available for assisted reproductive techniques. The present review aims to determine the differences between these gonadotrophins and to establish the advantages and limitations of each in terms of their origin, safety, and efficacy. From the technical point of view, recombinant gonadotrophins have enhanced purity, specific activity and greater consistency. From the safety point of view, there are clear differences in the origin and manufacturing process and consequently in the risk of infectious diseases; however, to date, all gonadotropins have been demonstrated to be safe. Differences in efficacy are more difficult to establish. Many trials have compared preparations but, because of their small size and variations in study design, the results have been variable. Most of the studies have failed to detect any differences and consequently several meta-analyses have aimed to determine whether recombinant follicle- stimulating hormone (FSH) gonadotrophins are preferable to urinary-derived FSH and whether recombinant hormone is superior to human menopausal gonadotropin (hMG). The published results do not demonstrate that urinary-derived FSH or hMG, alone or when data are pooled, are more effective than recombinant FSH. In contrast, recombinant gonadotrophins have been shown to be more effective than urinary- derived gonadotrophins as a whole (AU)


Assuntos
Adulto , Feminino , Humanos , Indução da Ovulação , Técnicas de Reprodução Assistida , Gonadotropinas/uso terapêutico , Menotropinas/uso terapêutico , Hormônio Luteinizante/uso terapêutico , Hormônio Foliculoestimulante Humano/uso terapêutico , Hormônio Liberador de Gonadotropina/uso terapêutico , Gonadotropina Coriônica/uso terapêutico , Clomifeno/uso terapêutico , Urofolitropina/uso terapêutico , Técnicas de Reprodução Assistida/história , Gonadotropinas/história , Medicina Reprodutiva/tendências , Infertilidade/etiologia , Doenças Transmissíveis
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...