Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
J Assist Reprod Genet ; 39(2): 409-416, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35066698

RESUMO

PURPOSE: Growth hormone (GH) supplementation in association with in vitro fertilization (IVF) is worldwide again increasing, even though study outcomes have been discrepant. Since GH acts via insulin-like growth factor-1 (IGF-1), its utilization in IVF would only seem to make sense with low IGF-1. We, therefore, determined whether IGF-I levels affect IVF outcomes. METHODS: Retrospectively, 302 consecutive first fresh, non-donor IVF cycles were studied, excluding patients on GH supplementation. Patients were divided into 3 subgroups: IGF-1 in lower 25th percentile (group A, < 132 ng/mL, n = 64); 25th-75th percentile (B, 133-202 ng/mL, n = 164), and upper 25th percentile (C, > 202 ng/mL, n = 74). IGF-1 was tested immunochemiluminometric with normal range at 78-270 ng/mL. Because of the study patients' adverse selection and low pregnancy chances, the main outcome measure for the study was cycle cancellation. Secondary outcomes were oocyte numbers, embryos transferred, pregnancies, and live births. RESULTS: Group A was significantly older than B and C (P = 0.019). IGF-1 decreased with increasing age per year by 2.2 ± 0.65 ng/mL (P = 0.0007). FSH was best in group B and worst in A (trend, P = 0.085); AMH was best in B and worst in A (N.S.). Cycle cancellations were lowest in C (11.6%) and highest in A (25.0%; P = 0.042). This significance further improved with age adjustment (P = 0.021). Oocytes, embryo numbers, pregnancies, and live birth rates did not differ, though oocyte numbers trended highest in B. CONCLUSIONS: Here presented results support the hypothesis that IGF-1 levels affect IVF outcomes. GH treatments, therefore, may be effective only with low IGF-1.


Assuntos
Hormônio do Crescimento , Fator de Crescimento Insulin-Like I , Suplementos Nutricionais , Feminino , Fertilização in vitro , Humanos , Nascido Vivo , Indução da Ovulação , Gravidez , Taxa de Gravidez , Estudos Retrospectivos
2.
Endocrine ; 72(1): 260-267, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33009651

RESUMO

BACKGROUND: Mediated via the androgen receptor on granulosa cells, models of small growing follicle stages demonstrate dependence on testosterone. Androgen deficiency reduces ovarian response to follicle stimulation hormone (FSH), granulosa cell mass and estradiol (E2) production falls and FSH, therefore, rises. Though potentially of adrenal and/or ovarian origin, androgen deficiency in association with female infertility is almost universally primarily of adrenal origin, raising the possibility that women with presumptive diagnosis of primary ovarian insufficiency (POI), also called primary ovarian failure (POF) may actually suffer from secondary ovarian insufficiency (SOI) due to adrenal hypoandrogenism that leads to follicular arrest at small-growing follicle stages. METHODS: This retrospective cohort study was performed in a private, academically affiliated infertility center in New York City. We searched the center's anonymized electronic research data bank for consecutive patients who presented with a diagnosis of POI, defined by age <41 year, FSH > 40.0 mIU/mL, amenorrhea for at least 6 month, and low testosterone (T), defined as total T (TT) in the lowest age-specific quartile of normal range. This study did not include patients with oophoritis. Since dehydroepiandrosterone sulfate (DHEAS) is the only androgen almost exclusively produce by adrenals, adrenal hypoandrogenism was defined by DHEAS < 100ug/dL. Thirteen of 78 presumed POI women (16.67%) qualified and represented the original study population. POI patients are usually treated with third-party egg donation; 6/13, however, rejected egg donation for personal or religious reasons and insisted on undergoing at least one last IVF cycle attempt (final study population). In preparation, they were supplemented with DHEA 25 mg TID and CoQ10 333 mg TID for at least 6 weeks prior to ovarian stimulation for IVF with FSH and human menopausal gonadotropins (hMG). Since POI patients are expected to be resistant to ovarian stimulation, primary outcome for the study was ovarian response, while secondary outcome was pregnancy/delivery. RESULTS: Though POI/POF patients usually are completely unresponsive to ovarian stimulation, to our surprise, 5/6 (83.3%) patients demonstrated an objective follicle response. In addition, 2/6 (33.3%) conceived spontaneously between IVF cycles, while on DHEA and CoQ10 supplementation and delivered healthy offspring. One of those is currently in treatment for a second child. CONCLUSIONS: This preliminary report suggests that a surprising portion of young women below age 41, tagged with a diagnosis of POI/POF, due to adrenal hypoandrogenism actually suffer from a form of SOI, at least in some cases amenable to treatment by androgen supplementation. Since true POI/POF usually requires third-party egg donation, correct differentiation between POI and SOI in such women appears of great importance and may warrant a trial stimulation after androgen pre-supplementation for at least 6 weeks.


Assuntos
Infertilidade Feminina , Insuficiência Ovariana Primária , Adulto , Criança , Feminino , Hormônio Foliculoestimulante , Humanos , Infertilidade Feminina/diagnóstico , Infertilidade Feminina/etiologia , Cidade de Nova Iorque , Indução da Ovulação , Gravidez , Insuficiência Ovariana Primária/complicações , Insuficiência Ovariana Primária/diagnóstico , Estudos Retrospectivos
3.
Endocrine ; 63(3): 632-638, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30311171

RESUMO

PURPOSE: To investigate the effects of dehydroepiandrosterone (DHEA) supplementation on female sexual function in premenopausal infertile women of advanced ages. METHODS: This observational study was conducted in an academically affiliated private fertility center. Patients included 87 premenopausal infertile women, 50 of whom completed the study including the Female Sexual Function Index (FSFI) questionnaires and comprehensive endocrine evaluation before and 4-8 weeks after initiating 25 mg of oral micronized DHEA TID. RESULTS: Age of patients was 41.1 ± 4.2 years, BMI 24.4 ± 6.1 kg/m2, 86% were married, and 42% were parous. Following supplementation with DHEA, all serum androgen levels increased (each P < 0.0001), while FSH levels decreased by 2.6 ± 4.4 from a baseline of 10.3 ± 5.4 mIU/mL (P = 0.009). The FSFI score for the whole study group increased by 7% (from 27.2 ± 6.9 to 29.2 ± 5.6; P = 0.0166). Domain scores for desire increased by 17% (P = 0.0004) and by 12% for arousal (P = 0.0122); lubrication demonstrated an 8% trend towards improvement (P = 0.0551), while no changes in domain scores for orgasm, satisfaction, or pain were observed. Women in the lowest starting FSFI score quartile (<25.7), experienced a 6.1 ± 8.0 (34%) increase in total FSFI score following DHEA supplementation. Among these women, improvements in domain categories were noted for desire (40%), arousal (46%), lubrication (33%), orgasm (54%), satisfaction (24%), and pain (25%). CONCLUSIONS: This uncontrolled observational study implies that supplementation with DHEA improves sexual function in older premenopausal women with low baseline FSFI scores.


Assuntos
Desidroepiandrosterona/uso terapêutico , Infertilidade Feminina/tratamento farmacológico , Comportamento Sexual/efeitos dos fármacos , Adulto , Desidroepiandrosterona/sangue , Desidroepiandrosterona/farmacologia , Feminino , Humanos , Pessoa de Meia-Idade , Pré-Menopausa
4.
Reprod Biomed Online ; 36(1): 12-19, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29223475

RESUMO

'Poor responders' is a term used to describe a subpopulation of IVF patients who do not respond well to ovarian stimulation with gonadotrophins. While there is no standard definition of a poor responder, these patients tend to be of advanced maternal age (≥40 years), have a history of poor ovarian response with conventional stimulation protocols, and/or have low ovarian reserve. Despite the heterogeneity of this patient group, there are characteristics and needs common to many poor responders that can be addressed through a holistic approach. Stimulation during the earlier stages of follicle maturation may help synchronize follicle development for improved response to later gonadotrophin stimulation, and supplementation with dehydroepiandrosterone or human growth hormone may promote early follicle development in poor responders. IVF protocols should be specifically tailored to poor responders to complement the patient's natural cycle. Because poor responders tend to have high levels of stress and anxiety, patients should receive psychological counselling and support, both prior to and during IVF cycles, to ensure optimal outcomes and improve patients' experience. It is important to set realistic expectations with poor responders and their partners to help patients make informed decisions and better manage their distress and anxiety.


Assuntos
Hormônios/administração & dosagem , Folículo Ovariano/efeitos dos fármacos , Indução da Ovulação/métodos , Animais , Terapias Complementares , Feminino , Fertilização in vitro , Humanos , Indução da Ovulação/psicologia , Falha de Tratamento
5.
J Steroid Biochem Mol Biol ; 167: 144-152, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27993552

RESUMO

How anti-Müllerian hormone (AMH) and testosterone (T) interrelate in infertile women is currently largely unknown. We, therefore, in a retrospective cohort study investigated how infertile women with high-AMH (AMH ≥75th quantile; n=144) and with normal-AMH (25th-75th quantile; n=313), stratified for low-T (total testosterone ≤19.0ng/dL), normal-T (19.0-29.0ng/dL) and high-T (>29.0ng/dL) phenotypically behaved. Patient age, follicle stimulating hormone (FSH), dehyroepiandrosterone (DHEA), DHEA sulphate (DHEAS), cortisol (C), adrenocorticotrophic hormone (ACTH), IVF outcomes, as well as inflammatory and immune panels were then compared between groups, with AMH and T as variables. We identified a previously unknown infertile PCOS-like phenotype, characterized by high-AMH but, atypically, low-T, with predisposition toward autoimmunity. It presents with incompatible high-AMH and low-T (<19.0ng/dL), is restricted to lean PCOS-like patients, presenting delayed for tertiary fertility services. Since also characterized by low DHEAS, low-T is likely of adrenal origina, and consequence of autoimmune adrenal insufficiency since also accompanied by low-C and evidence of autoimmunity. DHEA supplementation in such patients equalizes low- to normal-T and normalizes IVF cycle outcomes. Once recognized, this high-AMH/low-T phenotype is surprisingly common in tertiary fertility centers but, currently, goes unrecognized. Its likely adrenal autoimmune etiology offers interesting new directions for investigations of adrenals control over ovarian function via adrenal androgen production.


Assuntos
Glândulas Suprarrenais/patologia , Androgênios/metabolismo , Hormônio Antimülleriano/metabolismo , Infertilidade Feminina/metabolismo , Síndrome do Ovário Policístico/metabolismo , Glândulas Suprarrenais/embriologia , Insuficiência Adrenal/complicações , Insuficiência Adrenal/diagnóstico , Adulto , Autoimunidade/imunologia , Feminino , Fertilização in vitro , Humanos , Pessoa de Meia-Idade , Ovário/metabolismo , Fenótipo , Síndrome do Ovário Policístico/complicações , Síndrome do Ovário Policístico/diagnóstico , Prognóstico , Estudos Retrospectivos , Testosterona/metabolismo , Glândula Tireoide/metabolismo , Resultado do Tratamento , Zona Reticular/metabolismo
6.
J Endocrinol ; 226(3): 167-80, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26264981

RESUMO

Why IVF pregnancy rates decline sharply after age 43 is unknown. In this study, we compared granulosa cell (GC) function in young oocyte donors (n=31, ages 21-29), middle-aged (n=64, ages 30-37) and older infertile patients (n=41, ages 43-47). Gene expressions related to gonadotropin activity, steroidogenesis, apoptosis and luteinization were examined by real-time PCR and western blot in GCs collected from follicular fluid. FSH receptor (FSHR), aromatase (CYP19A1) and 17ß-hydroxysteroid dehydrogenase (HSD17B) expression were found down regulated with advancing age, while LH receptor (LHCGR), P450scc (CYP11A1) and progesterone receptor (PGR) were up regulated. Upon in vitro culture, GCs were found to exhibit lower proliferation and increased apoptosis with aging. While FSH supplementation stimulated GCs growth and prevented luteinization in vitro. These observations demonstrate age-related functional declines in GCs, consistent with premature luteinization. To avoid premature luteinization in women above age 43, we advanced oocyte retrieval by administering human chorionic gonadotropin at maximal leading follicle size of 16  mm (routine 19-21  mm). Compared to normal cycles in women of similar age, earlier retrieved patients demonstrated only a marginal increase in oocyte prematurity, yet exhibited improved embryo numbers as well as quality and respectable clinical pregnancy rates. Premature follicular luteinization appears to contribute to rapidly declining IVF pregnancy chances after age 43, and can be avoided by earlier oocyte retrieval.


Assuntos
Envelhecimento/fisiologia , Células da Granulosa/fisiologia , Luteinização/fisiologia , Recuperação de Oócitos/métodos , 17-Hidroxiesteroide Desidrogenases/genética , 17-Hidroxiesteroide Desidrogenases/metabolismo , Adulto , Fatores Etários , Aromatase/genética , Aromatase/metabolismo , Regulação para Baixo , Feminino , Regulação da Expressão Gênica , Humanos , Pessoa de Meia-Idade , Gravidez , Receptores do FSH/genética , Receptores do FSH/metabolismo , Receptores do LH/genética , Receptores do LH/metabolismo , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Regulação para Cima , Adulto Jovem
7.
Nat Rev Endocrinol ; 11(7): 429-41, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25942654

RESUMO

Hypoandrogenism in women with low functional ovarian reserve (LFOR, defined as an abnormally low number of small growing follicles) adversely affects fertility. The androgen precursor dehydroepiandrosterone (DHEA) is increasingly used to supplement treatment protocols in women with LFOR undergoing in vitro fertilization. Due to differences in androgen metabolism, however, responses to DHEA supplementation vary between patients. In addition to overall declines in steroidogenic capacity with advancing age, genetic factors, which result in altered expression or enzymatic function of key steroidogenic proteins or their upstream regulators, might further exacerbate variations in the conversion of DHEA to testosterone. In this Review, we discuss in vitro studies and animal models of polymorphisms and gene mutations that affect the conversion of DHEA to testosterone and attempt to elucidate how these variations affect female hormone profiles. We also discuss treatment options that modulate levels of testosterone by targeting the expression of steroidogenic genes. Common variants in genes encoding DHEA sulphotransferase, aromatase, steroid 5α-reductase, androgen receptor, sex-hormone binding globulin, fragile X mental retardation protein and breast cancer type 1 susceptibility protein have been implicated in androgen metabolism and, therefore, can affect levels of androgens in women. Short of screening for all potential genetic variants, hormonal assessments of patients with low testosterone levels after DHEA supplementation facilitate identification of underlying genetic defects. The genetic predisposition of patients can then be used to design individualized fertility treatments.


Assuntos
Androgênios/deficiência , Desidroepiandrosterona/metabolismo , Infertilidade Feminina/genética , Testosterona/metabolismo , 3-Oxo-5-alfa-Esteroide 4-Desidrogenase/genética , Animais , Aromatase/genética , Desidroepiandrosterona/uso terapêutico , Modelos Animais de Doenças , Feminino , Proteína do X Frágil da Deficiência Intelectual/genética , Genes BRCA1 , Humanos , Reserva Ovariana , Polimorfismo Genético , Receptores Androgênicos/genética , Globulina de Ligação a Hormônio Sexual/genética , Sulfotransferases/genética
8.
Reprod Biol Endocrinol ; 13: 38, 2015 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-25943720

RESUMO

BACKGROUND: Low functional ovarian reserve (LFOR) has been associated with hypoandrogenemia and increased embryo aneuploidy, while androgen supplementation has been reported to improve aneuploidy rates. We, therefore, assessed whether in infertile women undergoing in vitro fertilization (IVF) androgen concentrations are associated with aneuploidy rates. METHODS: This study was performed in 2 academically affiliated fertility centers in New York City and an academically affiliated steroid chemistry laboratory in Utah. Androgen concentrations were measured in blinded fashion from 84 infertile women (age 40.3+/-2.4 years) at New York University (NYU), using a validated LC-MS/MS method, in cryopreserved serum samples of patients who had undergone IVF with concomitant preimplantation genetic screening (PGS), utilizing a 24-chromosome platform. The Center for Human Reproduction (CHR) provided plasma samples of 100 historical controls (ages 38.6+/-5.0 years) undergoing IVF without PGS. Statistical comparisons were made of androgen concentrations, and of associations between androgen concentrations and embryo aneuploidy. RESULTS: Women undergoing IVF+PGS at NYU revealed no association between embryo aneuploidy and androgen concentrations but demonstrated significantly lower androgen concentrations than the 100 control patients from CHR, CONCLUSIONS: Though this study revealed no association between androgen levels and embryo ploidy, the extremely low androgen levels in the NYU study group raise the possibility of a threshold effect below which testosterone no longer affects aneuploidy. Before an androgen effect on embryo ploidy can be completely ruled out, a patient population with more normal androgen levels has to be investigated.


Assuntos
Aneuploidia , Infertilidade Feminina/metabolismo , Reserva Ovariana , Fatores Etários , Androstenodiona/sangue , Hormônio Antimülleriano/sangue , Desidroepiandrosterona/sangue , Feminino , Hormônio Foliculoestimulante/sangue , Humanos , Idade Materna , Diagnóstico Pré-Implantação , Fatores de Risco , Testosterona/sangue
10.
Reprod Biol Endocrinol ; 12: 68, 2014 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-25048047

RESUMO

BACKGROUND: In various animal models androgens have been demonstrated to enhance follicle stimulating hormone (FSH) activity on granulosa cells during small growing follicle stages. To assess whether similar synergism may also exist in humans we investigated women on androgen (dehydroepiandrosterone, DHEA) supplementation with varying concomitant FSH exposure. METHODS: In a case controlled cohort study we determine if time interval between IVF cycles of IVF treatment with FSH had an effect on ovarian response to ovulation induction in women supplemented with DHEA. Among 85 women with known low functional ovarian reserve (LFOR), supplemented with DHEA, and undergoing at least 3 consecutive IVF cycles, 68 demonstrated short (<120 days) intervals between repeated cycles (Group 1) and were, therefore, considered to have consistent FSH exposure. In contrast 17 women (Group 2) demonstrated long (>=120 days) intervals between repeated cycles and, therefore, were considered to demonstrate inconsistent FSH exposure. Trends in oocyte yields were compared between these groups, utilizing mixed model repeated measures ANOVA, adjusted for initial age and FSH dose. RESULTS: Only women in Group I demonstrated a linear increase in oocyte yields across their three cycles of treatments (F=7.92; df 1, 68.6; p=0.017). Moreover, the analysis revealed a significant interaction between the two patient groups and cycle number for retrieved oocytes (F=6.32, df=2, 85.9, p=0.003). CONCLUSIONS: This study offers preliminary confirmatory evidence that repeated short interval exposure to androgens in combination with FSH improves human FOR. A higher level of evidence will require prospectively randomized studies.


Assuntos
Androgênios/farmacologia , Desidroepiandrosterona/farmacologia , Fármacos para a Fertilidade Feminina/farmacologia , Hormônio Foliculoestimulante/farmacologia , Infertilidade Feminina/terapia , Oogênese/efeitos dos fármacos , Indução da Ovulação , Adulto , Estudos de Casos e Controles , Estudos de Coortes , Sinergismo Farmacológico , Feminino , Fertilização in vitro , Hormônio Foliculoestimulante/sangue , Humanos , Infertilidade Feminina/sangue , Infertilidade Feminina/etiologia , Infertilidade Feminina/patologia , Infertilidade Masculina , Masculino , Recuperação de Oócitos , Folículo Ovariano/citologia , Folículo Ovariano/efeitos dos fármacos , Folículo Ovariano/patologia , Reserva Ovariana , Insuficiência Ovariana Primária/fisiopatologia , Estudos Retrospectivos , Índice de Gravidade de Doença
11.
Hum Reprod ; 28(4): 1084-91, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23427231

RESUMO

STUDY QUESTION: Is diminished functional ovarian reserve (DFOR) associated with low androgen levels? SUMMARY ANSWER: Low androgen levels are associated with DFOR at all ages. WHAT IS KNOWN ALREADY: Androgen supplementation via dehydroepiandrosterone (DHEA) has been reported to improve functional ovarian reserve (FOR); pregnancy rates in IVF cycles are associated with how well DHEA converts to testosterone (T); and androgen effects through the androgen receptor have been demonstrated in mice to beneficially affect early stages of follicle maturation. STUDY DESIGN, SIZE, DURATION: In a controlled cohort study we investigated consecutive women presenting to our center with two forms of DFOR, premature ovarian aging/occult primary ovarian insufficiency (POA/OPOI) and physiologic diminished ovarian reserve (DOR). As controls for POA/OPOI patients, infertile women with normal age-specific FOR were recruited. PARTICIPANTS/MATERIALS, SETTINGS, METHODS: The study involved 140 women with POA/OPOI, defined as age <38 years and abnormally low FOR by age-specific FSH and/or anti-Müllerian hormone (AMH), 166 women with DOR, defined as women age >40 years. Forty-nine control patients <38 years demonstrated normal FOR by FSH and/or AMH. In all three patient groups dehydroepiandrosterone (DHEA), DHEA-sulfate (DHEAS), total testosterone (TT) and free testosterone (FT) at the time of initial presentation to our fertility center were assessed. In a small subgroup of women early morning cortisol levels were also assessed. MAIN RESULTS AND THE ROLE OF CHANCE: DHEAS marginally varied between the three groups (P = 0.04), with older women with DOR actually demonstrating higher levels than controls (P = 0.03). TT differed between the three groups more profoundly (P = 0.005), with women with POA/OPOI demonstrating significantly lower levels than controls (P = 0.009). Adjustment for body mass index, age and race in principle maintained observed differences in TT between groups, while adjustment for FMR1 (fragile X mental retardation 1) genotypes/sub-genotypes eliminated all differences. All three patient groups demonstrated low morning cortisol levels. LIMITATIONS, REASONS FOR CAUTION: While results support lower androgen levels in women with DOR, and even more so in women with POA/OPOI, presented data should be viewed as preliminary, considering the known variability of androgen levels and the small number of women in whom morning cortisol levels were available. WIDER IMPLICATIONS OF THE FINDINGS: Especially at young ages DFOR appears associated with significant hypoandrogenism (low T) in comparison with young control patients with normal FOR, raising the question whether this hypoandrogenism originates in adrenals or ovaries. POA/OPOI, thus, phenotypically mimics the polycystic ovary syndrome, where similar questions arise, though in regard to hyperandrogenism. STUDY FUNDING/COMPETING INTEREST(S): This research was supported by the Foundation for Reproductive Medicine, a not-for-profit medical research foundation and intramural funds from the Center for Human Reproduction. N.G. and D.H.B are members of the Board of the Foundation for Reproductive Medicine. N.G., A.W. and D.H.B. received research support, lecture fees and travel support from a variety of pharmaceutical and medical device companies, none in any way related to the issues discussed in this manuscript. N.G. and D.H.B. are listed as co-inventors on two, already granted US user patents, which claim therapeutic benefits from DHEA supplementation in women with DFOR and DOR: both authors are also listed on additional pending patents in regard to DHEA supplementation and on pending patents, claiming diagnostic and therapeutic benefits from the determination of CGG repeats on the FMR1 gene. N.G. is the owner of the Center for Human Reproduction, where this research was performed.


Assuntos
Androgênios/sangue , Hormônio Antimülleriano/sangue , Ovário/fisiologia , Adulto , Fatores Etários , Estudos de Coortes , Desidroepiandrosterona/sangue , Sulfato de Desidroepiandrosterona/sangue , Feminino , Hormônio Foliculoestimulante/sangue , Humanos , Hidrocortisona/sangue , Testosterona/sangue
12.
J Assist Reprod Genet ; 30(1): 49-62, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23212832

RESUMO

PURPOSE: To investigate whether androgen conversion rates after supplementation with dehydroepiandrosterone (DHEA) differ, and whether differences between patients with diminished ovarian reserve (DOR) are predictive of pregnancy chances in association with in vitro fertilization (IVF). METHODS: In a prospective cohort study we investigated 213 women with DOR, stratified for age (≤ 38 or >38 years) and ovarian FMR1 genotypes/sub-genotypes. All women were for at least 6 weeks supplemented with 75 mg of DHEA daily prior to IVF, between initial presentation and start of 1st IVF cycles. Levels of DHEA, DHEA-sulfate (DHEAS), total T (TT) and free T (FT) at baseline ((BL)) and IVF cycle start ((CS)) were then compared between conception and non-conception cycles. RESULTS: Mean age for the study population was 41.5 ± 4.4 years. Forty-seven IVF cycles (22.1 %) resulted in clinical pregnancy. Benefits of DHEA on pregnancy rates were statistically associated with efficiency of androgen conversion from DHEA to T and amplitude of T gain. Younger women converted significantly more efficiently than older females, and selected FMR1 genotypes/sub-genotypes converted better than others. FSH/androgen and AMH/androgen ratios represent promising new predictors of IVF pregnancy chances in women with DOR. CONCLUSIONS: DOR at all ages appears to represent an androgen-deficient state, benefitting from androgen supplementation. Efficacy of androgen supplementation with DHEA, however, varies depending on female age and FMR1 genotype/sub-genotype. Further clarification of FMR1 effects should lead to better individualization of androgen supplementation, whether via DHEA or other androgenic compounds.


Assuntos
Androgênios/administração & dosagem , Infertilidade Feminina/terapia , Ovário/metabolismo , Taxa de Gravidez , Testosterona/sangue , Adulto , Fatores Etários , Androgênios/deficiência , Hormônio Antimülleriano/sangue , Desidroepiandrosterona/administração & dosagem , Desidroepiandrosterona/sangue , Feminino , Fertilização in vitro/métodos , Hormônio Foliculoestimulante/sangue , Proteína do X Frágil da Deficiência Intelectual/genética , Genótipo , Humanos , Infertilidade Feminina/diagnóstico , Modelos Logísticos , Ciclo Menstrual , Pessoa de Meia-Idade , Ovário/efeitos dos fármacos , Indução da Ovulação/métodos , Valor Preditivo dos Testes , Gravidez , Estudos Prospectivos , Resultado do Tratamento
13.
Hum Reprod ; 27(11): 3287-93, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22811313

RESUMO

BACKGROUND: For decades androgens have been considered detrimental to follicle maturation. Animal studies now suggest that they are essential for normal folliculogenesis. Especially in women with premature ovarian aging (POA), recent IVF data in humans are supportive. The literature also suggests an association between recently reported ovarian genotypes of the FMR1 gene and ovarian aging patterns. We, therefore, attempted to determine a potential difference in androgen concentrations and androgen interactions in women with POA who do or do not become pregnant while undergoing androgen supplementation, and whether androgen concentrations and pregnancy chances are affected by FMR1 genotypes. METHODS: We longitudinally assessed androgen metabolism in 91 women with POA, following pre-supplementation with micronized dehydroepiandrosterone (DHEA) prior to IVF. IVF outcomes were assessed based on androgen levels and ovarian FMR1 genotypes. RESULTS: The mean age of the women was 39.8 ± 4.4 years; the clinical pregnancy rate was 25.3%. Total androgen concentrations were not associated with pregnancy; however, in women with abnormal FMR1 genotypes, but not those with the normal genotype, free testosterone significantly affected clinical pregnancy potential (ß = 1.101, SE ± 0.508, P = 0.03). At the start of the IVF cycle, interactions of DHEA with total and free testosterone also significantly affected subsequent pregnancy rates (ß = -0.058, SE ± 0.023, P = 0.01 and ß = -0.496, SE ± 0.197, P = 0.012). CONCLUSIONS: Androgen interactions significantly influence IVF pregnancy rates in women with POA, with the impact of total androgens on cycle outcomes varying according to FMR1 genotypes. These observations suggest that the effectiveness of androgen supplementation in women with POA varies based on FMR1 genotypes, and defines androgen deficiency as a subset of diminished ovarian reserve.


Assuntos
Androgênios/uso terapêutico , Desidroepiandrosterona/uso terapêutico , Suplementos Nutricionais , Proteína do X Frágil da Deficiência Intelectual/genética , Oogênese , Polimorfismo Genético , Insuficiência Ovariana Primária/dietoterapia , Adulto , Androgênios/química , Androgênios/deficiência , Androgênios/metabolismo , Estudos de Coortes , Desidroepiandrosterona/química , Feminino , Fertilização in vitro , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Estudos de Associação Genética , Humanos , Infertilidade Feminina/etiologia , Infertilidade Feminina/terapia , Estudos Longitudinais , Cidade de Nova Iorque , Folículo Ovariano/metabolismo , Folículo Ovariano/fisiopatologia , Gravidez , Taxa de Gravidez , Insuficiência Ovariana Primária/genética , Insuficiência Ovariana Primária/metabolismo , Insuficiência Ovariana Primária/fisiopatologia , Estudos Retrospectivos
14.
Reprod Biol Endocrinol ; 9: 67, 2011 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-21586137

RESUMO

BACKGROUND: With infertility populations in the developed world rapidly aging, treatment of diminished ovarian reserve (DOR) assumes increasing clinical importance. Dehydroepiandrosterone (DHEA) has been reported to improve pregnancy chances with DOR, and is now utilized by approximately one third of all IVF centers world-wide. Increasing DHEA utilization and publication of a first prospectively randomized trial now warrants a systematic review. METHODS: PubMed, Cochrane and Ovid Medline were searched between 1995 and 2010 under the following strategy: [ and ]. Bibliographies of relevant publications were further explored for additional relevant citations. Since only one randomized study has been published, publications, independent of evidence levels and quality assessment, were reviewed. RESULTS: Current best available evidence suggests that DHEA improves ovarian function, increases pregnancy chances and, by reducing aneuploidy, lowers miscarriage rates. DHEA over time also appears to objectively improve ovarian reserve. Recent animal data support androgens in promoting preantral follicle growth and reduction in follicle atresia. DISCUSSION: Improvement of oocyte/embryo quality with DHEA supplementation potentially suggests a new concept of ovarian aging, where ovarian environments, but not oocytes themselves, age. DHEA may, thus, represent a first agent beneficially affecting aging ovarian environments. Others can be expected to follow.


Assuntos
Desidroepiandrosterona/uso terapêutico , Insuficiência Ovariana Primária/tratamento farmacológico , Animais , Contagem de Células , Feminino , Fármacos para a Fertilidade Feminina/uso terapêutico , Terapia de Reposição Hormonal , Humanos , Ovário/efeitos dos fármacos , Ovário/patologia , Gravidez , Insuficiência Ovariana Primária/patologia , Ensaios Clínicos Controlados Aleatórios como Assunto/estatística & dados numéricos
15.
Reprod Biol Endocrinol ; 9: 23, 2011 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-21299886

RESUMO

Though a widely utilized term and clinical concept, ovarian reserve (OR) has been only inadequately defined. Based on Medline and PubMed searches we here define OR in its various components, review genetic control of OR, with special emphasis on the FMR1 gene, and discuss whether diminished OR (DOR) is treatable. What is generally referred to as OR reflects only a small portion of total OR (TOR), a pool of growing (recruited) follicles (GFs) at different stages of maturation. Functional OR (FOR) depends on size of the follicle pool at menarche and the follicle recruitment rate. Both vary between individuals and, at least partially, are under genetic control. The FMR1 gene plays a role in defining FOR at all ages. Infertility treatments have in the past almost exclusively only centered on the last two weeks of folliculogenesis, the gonadotropin-sensitive phase. Expansions of treatments into earlier stages of maturation will offer opportunity to significantly improve ovarian stimulation protocols, especially in women with DOR. Dehydroepiandrosterone (DHEA) may represent a first such intervention. Data generated in DHEA-supplemented women, indeed, suggest a new ovarian aging concept, based on aging of ovarian environments and not, as currently is believed, aging oocytes.


Assuntos
Envelhecimento/fisiologia , Ovário/fisiologia , Adulto , Desidroepiandrosterona/uso terapêutico , Feminino , Proteína do X Frágil da Deficiência Intelectual/genética , Humanos , Pessoa de Meia-Idade , Folículo Ovariano/efeitos dos fármacos , Folículo Ovariano/fisiologia , Folículo Ovariano/fisiopatologia , Ovário/fisiopatologia , Indução da Ovulação/métodos
16.
Reprod Biol Endocrinol ; 8: 140, 2010 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-21067609

RESUMO

BACKGROUND: Dehydroepiandrosterone (DHEA) has been reported to improve pregnancy chances in women with diminished ovarian reserve (DOR), and to reduce miscarriage rates by 50-80%. Such an effect is mathematically inconceivable without beneficial effects on embryo ploidy. This study, therefore, assesses effects of DHEA on embryo aneuploidy. METHODS: In a 1:2, matched case control study 22 consecutive women with DOR, supplemented with DHEA, underwent preimplantation genetic screening (PGS) of embryos during in vitro fertilization (IVF) cycles. Each was matched by patient age and time period of IVF with two control IVF cycles without DHEA supplementation (n = 44). PGS was performed for chromosomes X, Y, 13, 16, 18, 21 and 22, and involved determination of numbers and percentages of aneuploid embryos. RESULTS: DHEA supplementation to a significant degree reduced number (P = 0.029) and percentages (P < 0.001) of aneuploid embryos, adjusted for relevant covariates. Short term supplementation (4-12 weeks) resulted in greatest reduction in aneuploidy (21.6%, 95% CI -2.871-46.031). DISCUSSION: Beneficial DHEA effects on DOR patients, at least partially, are the likely consequence of lower embryo aneuploidy. DHEA supplementation also deserves investigation in older fertile women, attempting to conceive, where a similar effect, potentially, could positively affect public health.


Assuntos
Aneuploidia , Transtornos Cromossômicos/prevenção & controle , Desidroepiandrosterona/uso terapêutico , Ovário/patologia , Diagnóstico Pré-Implantação , Aborto Espontâneo , Adulto , Estudos de Casos e Controles , Feminino , Fertilização in vitro , Testes Genéticos , Humanos , Gravidez , Taxa de Gravidez , Estudos Retrospectivos
17.
Reprod Biomed Online ; 21(3): 360-5, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20638339

RESUMO

Dehydroepiandrosterone (DHEA) has been reported to improve oocyte/embryo yields and oocyte/embryo quality in women with diminished ovarian reserve. Whether DHEA objectively improves ovarian reserve is, however, unknown. This study investigated 120 consecutive patients with diminished ovarian reserve, supplemented for 30-120 days (mean 73+/-27) with DHEA (25mg three times daily). Anti-Müllerian hormone (AMH) concentrations were determined in relationship to DHEA supplementation using linear regression and, longitudinally, by examining interaction between days of DHEA treatment and pregnancy success in respect to changes in AMH. AMH concentrations significantly improved after DHEA supplementation over time (P=0.002). Women under age 38 years demonstrated higher AMH concentrations and improved AMH concentrations more than older females. AMH improved longitudinally by approximately 60% (P<0.0002). Women reaching IVF experienced a 23.64% clinical pregnancy rate and conceiving women showed significantly improved AMH concentrations compared with those who did not (P=0.001). DHEA supplementation, thus, significantly improved ovarian reserve in parallel with longer DHEA use and was more pronounced in younger women.


Assuntos
Desidroepiandrosterona/uso terapêutico , Infertilidade Feminina/tratamento farmacológico , Ovário/efeitos dos fármacos , Adulto , Hormônio Antimülleriano/sangue , Estudos Transversais , Desidroepiandrosterona/administração & dosagem , Estradiol/sangue , Feminino , Fertilização in vitro , Hormônio Foliculoestimulante/sangue , Humanos , Infertilidade Feminina/fisiopatologia , Infertilidade Feminina/terapia , Estudos Longitudinais , Ovário/fisiopatologia , Gravidez , Estudos Retrospectivos
18.
Reprod Biol Endocrinol ; 7: 108, 2009 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-19811650

RESUMO

BACKGROUND: Dehydroepinadrosterone (DHEA) supplementation improves pregnancy chances in women with diminished ovarian reserve (DOR), by possibly reducing aneuploidy. Since a large majority of spontaneous miscarriages are associated with aneuploidy, one can speculate that DHEA supplementation may also reduce miscarriage rates. METHODS: We retroactively compared, utilizing two independent statistical models, miscarriage rates in 73 DHEA supplemented pregnancies at two independent North American infertility centers, age-stratified, to miscarriages reported in a national U.S. in vitro fertilization (IVF) data base. RESULTS: After DHEA supplementation the miscarriage rate at both centers was 15.1% (15.0% and 15.2%, respectively). For DHEA supplementation Mantel-Hänszel common odds ratio (and 95% confidence interval), stratified by age, was significantly lower, relative to odds of miscarriage in the general IVF control population [0.49 (0.25-0.94; p = 0.04)]. Miscarriage rates after DHEA were significantly lower at all ages but most pronounced above age 35 years. DISCUSSION: Since DOR patients in the literature are reported to experience significantly higher miscarriage rates than average IVF patients, the here observed reduction in miscarriages after DHEA supplementation exceeds, however, all expectations. Miscarriage rates after DHEA not only were lower than in an average national IVF population but were comparable to rates reported in normally fertile populations. Low miscarriage rates, comparable to those of normal fertile women, are statistically impossible to achieve in DOR patients without assumption of a DHEA effect on embryo ploidy. Beyond further investigations in infertile populations, these data, therefore, also suggest the investigations of pre-conception DHEA supplementation in normal fertile populations above age 35 years.


Assuntos
Aborto Espontâneo/induzido quimicamente , Aborto Espontâneo/epidemiologia , Desidroepiandrosterona/efeitos adversos , Ovário/patologia , Adulto , Estudos de Casos e Controles , Contagem de Células , Feminino , Fertilização in vitro , Humanos , Infertilidade Feminina/epidemiologia , Infertilidade Feminina/patologia , Infertilidade Feminina/terapia , Oócitos/patologia , Ovário/efeitos dos fármacos , Gravidez , Ensaios Clínicos Controlados Aleatórios como Assunto/estatística & dados numéricos , Estudos Retrospectivos
19.
J Assist Reprod Genet ; 24(12): 629-34, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18071895

RESUMO

OBJECTIVE: We assessed the role of DHEA supplementation on pregnancy rates in women with diminished ovarian function. DESIGN: This is a case control study of 190 women with diminished ovarian function. The study group includes 89 patients who used supplementation with 75 mg daily of oral, micronized DHEA for up to 4 months prior to entry into in vitro fertilization (IVF). The control group is composed of 101 couples who received infertility treatment, but did not use DHEA. The primary outcome was clinical pregnancy after the patient's initial visit. We developed a Cox proportional hazards model to compare the proportional hazards of pregnancy among women using DHEA with the controls group. RESULTS: Cumulative clinical pregnancy rates were significantly higher in the study group (25 pregnancies; 28.4% vs. 11 pregnancies; 11.9%; relative hazard of pregnancy in study group (HR 3.8; 95% CI 1.2-11.8; p < 0.05). CONCLUSIONS: DHEA treatment resulted in significantly higher cumulative pregnancy rates. These data support a beneficial effect of DHEA supplementation among women with diminished ovarian function.


Assuntos
Desidroepiandrosterona/uso terapêutico , Infertilidade Feminina/tratamento farmacológico , Indução da Ovulação/métodos , Pré-Medicação/métodos , Insuficiência Ovariana Primária/tratamento farmacológico , Adulto , Estudos de Casos e Controles , Estudos de Coortes , Feminino , Humanos , Infertilidade Feminina/etiologia , Menopausa Precoce/efeitos dos fármacos , Ovário/efeitos dos fármacos , Ovário/fisiologia , Gravidez , Taxa de Gravidez , Pré-Medicação/tendências , Insuficiência Ovariana Primária/complicações , Estudos Retrospectivos
20.
Hum Reprod ; 21(11): 2845-9, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16997936

RESUMO

BACKGROUND: The aim of this study was to investigate the effect of treatment with dehydroepiandrosterone (DHEA) on fertility outcomes among women with diminished ovarian reserve. MATERIALS AND METHODS: This is a case-control study in an academically affiliated private infertility centre. Twenty-five women with significantly diminished ovarian reserve had one IVF cycle before and after DHEA treatment, with otherwise identical hormonal stimulation. Women received 75 mg of DHEA daily (25 mg three times daily) for an average of 17.6 +/- 2.13 weeks. We performed a comparison of IVF outcome parameters, before and after DHEA treatment, including peak estradiol (E(2)) levels, oocyte and embryo numbers, oocyte and embryo quality and embryo transfer statistics. RESULTS: Paired analysis of IVF cycle outcomes in 25 patients, who underwent cycles both before and after DHEA supplementation, demonstrated significant increases in fertilized oocytes (P < 0.001), normal day 3 embryos (P = 0.001), embryos transferred (P = 0.005) and average embryo scores per oocyte (P < 0.001) after DHEA treatment. CONCLUSION: This study confirms the previously reported beneficial effects of DHEA supplementation on ovarian function in women with diminished ovarian reserve.


Assuntos
Desidroepiandrosterona/farmacologia , Fertilização in vitro , Fertilização/fisiologia , Oócitos/fisiologia , Transferência Embrionária , Estradiol/sangue , Feminino , Fertilização/efeitos dos fármacos , Humanos , Oócitos/efeitos dos fármacos , Gravidez
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA