Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 810
Filtrar
1.
Clin Lab ; 69(9)2023 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-37702693

RESUMO

BACKGROUND: Only a few epidemiological studies have illuminated the association between pesticide exposure and female infertility. However, evidence of the available data is restricted and also controversial. Vitamin D supplement was considered as having a beneficial effect on fertility. So, the purpose of our study is to assess the effect of dietary vitamin D consumption on the relationship between pesticide exposure in home and female infertility. METHODS: There were a total of 2,968 subjects from the National Health and Nutrition Examination Survey (NHANES), 2011 - 2018. The daily vitamin D intake was divided into two groups high intake (≥ 6 µg/d) and low intake (< 6 µg/d). Multi-variable logistic regression analysis was used to assess the relationship among vitamin D intake, pesticide exposure, and female infertility. RESULTS: We found a significant association between household pesticide exposure and infertility on a basis of a fully-adjusted model (OR 1.61; 95% CI 1.1 - 2.37). Furthermore, the relationship between pesticide exposure and in-fertility differed from low vitamin D intake group (OR 3.96; 95% CI 1.77 - 8.86) and high intake group (OR 1.36, 95% CI: 0.86 - 2.16), and p for interaction is 0.043 stratified by vitamin D intake. CONCLUSIONS: A significant association of female infertility with pesticide exposure in home is modified by dietary vitamin D consumption. This was the first study to demonstrate that dietary vitamin D may alter associations of human female infertility with pesticide exposure in home.


Assuntos
Infertilidade Feminina , Praguicidas , Feminino , Humanos , Infertilidade Feminina/induzido quimicamente , Infertilidade Feminina/epidemiologia , Inquéritos Nutricionais , Estado Nutricional , Vitamina D , Vitaminas , Praguicidas/efeitos adversos
2.
Mol Biol Rep ; 50(8): 7069-7088, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37402067

RESUMO

INTRODUCTION:  According to the World Health Organization, infertility is a public health problem that affects around 48 million couples and 186 million individuals worldwide. Endocrine disruptors are one of the causes that raise more concern, given that it is a problem that has evolved with the progress of society. Many chemicals are used by food industry, entering food chain, and directly affecting human health. Endocrine disruptors have the capacity of interfering with the normal hormonal action, metabolism, and biosynthesis, which can lead to a variation of the normal hormonal homeostasis. Some of these endocrine disruptors are highly associated with diseases that are positively correlated with female infertility, such as polycystic ovary syndrome, endometriosis, irregular menstrual cycle and also disturbances on processes as steroidogenesis and development of the ovarian follicles. RESULTS: The present literature review covers various aspects of the possible relationship between endocrine disruptors and female infertility. Bisphenol A and its metabolites, phthalates, dioxins, organochlorine, and organophosphate compounds are groups of chemicals considered to have the capacity to disrupt endocrine activity and herein addressed. The results reported in in vivo studies and in clinical trials addressing endocrine disruptors and female infertility were discussed as well as their possible mechanism of action. CONCLUSIONS: Large, double-blind, placebo-controlled randomized clinical trials are needed to better understand the mechanisms of action of endocrine disruptors in female infertility, as well as the doses and frequency of exposure responsible for it.


Assuntos
Disruptores Endócrinos , Endometriose , Poluentes Ambientais , Infertilidade Feminina , Humanos , Feminino , Infertilidade Feminina/induzido quimicamente , Poluentes Ambientais/toxicidade , Disruptores Endócrinos/toxicidade , Folículo Ovariano , Ensaios Clínicos Controlados Aleatórios como Assunto
3.
J Clin Oncol ; 41(12): 2281-2292, 2023 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-36888938

RESUMO

PURPOSE: To review the complex concerns of oncofertility created through increased cancer survivorship and the long-term effects of cancer treatment in young adults. DESIGN: Review chemotherapy-induced ovarian dysfunction, outline how fertility may be addressed before treatment initiation, and discuss barriers to oncofertility treatment and guidelines for oncologists to provide this care to their patients. CONCLUSION: In women of childbearing potential, ovarian dysfunction resulting from cancer therapy has profound short- and long-term implications. Ovarian dysfunction can manifest as menstrual abnormalities, hot flashes, night sweats, impaired fertility, and in the long term, increased cardiovascular risk, bone mineral density loss, and cognitive deficits. The risk of ovarian dysfunction varies between drug classes, number of received lines of therapy, chemotherapy dosage, patient age, and baseline fertility status. Currently, there is no standard clinical practice to evaluate patients for their risk of developing ovarian dysfunction with systemic therapy or means to address hormonal fluctuations during treatment. This review provides a clinical guide to obtain a baseline fertility assessment and facilitate fertility preservation discussions.


Assuntos
Preservação da Fertilidade , Infertilidade Feminina , Neoplasias , Adulto Jovem , Humanos , Feminino , Preservação da Fertilidade/métodos , Fertilidade , Infertilidade Feminina/induzido quimicamente , Infertilidade Feminina/prevenção & controle , Neoplasias/tratamento farmacológico
4.
Taiwan J Obstet Gynecol ; 62(2): 256-263, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36965892

RESUMO

OBJECTIVE: In women, agents used in chemotherapy treatment have side effects such as accelerating follicular depletion and early menopause. Thus, cytotoxic treatments may cause various effects ranging from partial damage to the ovary to premature ovarian failure (POI) and infertility. This study aimed to investigate the protective effect of carvacrol on cisplatin (CIS)-induced reproductive toxicity in female rats. MATERIALS AND METHODS: The animals were divided to four groups; a healthy group (HG), administered only cisplatin 2.5 mg/kg (CIS); cisplatin 2.5 mg/kg + carvacrol mg/kg (CC-50), and cisplatin 2.5 mg/kg + carvacrol 100 mg/kg (CC-100). In this study, the CC-50 and CC-100 groups were injected with carvacrol at 50 and 100 mg/kg intraperitoneally (IP). The CIS and HG groupswere administered normal saline as a solvent in the same way. One hour afterwardthe CC-50 and CC-100 groups were injected with cisplatin at 2.5 mg/kg IP. This procedure was continued once a day for 14 days. At the end of this period, six rats from each group were euthanized with high-dose anaesthesia. Biochemical (oxidant-antioxidant and proinflammatory cytokines) and histopathological examinations were performed on the right ovarian tissue removed from the dead rats. The remaining (n = 6 in each group) animals were kept in the laboratory with mature male rats for two months for breeding. Rats that didn't give birth within two months were considered infertile. A one-way ANOVA test was used for the biochemical analysis, the a Kruskal Wallis test was used for the histopathological analysis. RESULTS: It has been observed that cisplatine causes oxidative stress and inflammatory damage in the ovarian tissue of animals and ultimately causes infertility due to this oxidative stress. While carvacrol significantly suppressed cisplatin-related oxidative stress in ovarian tissue at the 50 and 100 mg/kg doses, it could suppress proinflammatory cytokine increase only at thecytokine increase only at the 100 mg/kg dose. In addition, carvacrol significantly reduced the development of cisplatin-related infertility (from 0 to 83.3%) at a dose of 100 mg/kg. CONCLUSION: These findings suggest that carvacrol at high doses can reduce the harmful effects of cisplatin on the ovary and improve ovarian reserve in rats.


Assuntos
Infertilidade Feminina , Insuficiência Ovariana Primária , Humanos , Ratos , Feminino , Masculino , Animais , Cisplatino/uso terapêutico , Citocinas/efeitos adversos , Estresse Oxidativo , Insuficiência Ovariana Primária/induzido quimicamente , Insuficiência Ovariana Primária/tratamento farmacológico , Fertilidade , Infertilidade Feminina/induzido quimicamente , Infertilidade Feminina/tratamento farmacológico
5.
Reprod Biol Endocrinol ; 21(1): 17, 2023 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-36737817

RESUMO

BACKGROUND: Low vitamin D status has been associated with an increased risk for infertility. Recent evidence regarding the efficacy of vitamin D supplementation in improving reproductive outcomes is inconsistent. Therefore, this systematic review was conducted to investigate whether vitamin D supplementation could improve the reproductive outcomes of infertile patients and evaluate how the parameters of vitamin D supplementation affected the clinical pregnancy rate. METHODS: We searched seven electronic databases (CNKI, Cqvip, Wanfang, PubMed, Medline, Embase, and Cochrane Library) up to March 2022. Randomized and cohort studies were collected to assess the reproductive outcomes difference between the intervention (vitamin D) vs. the control (placebo or none). Mantel-Haenszel random effects models were used. Effects were reported as odds ratio (OR) and their 95% confidence interval (CI). PROSPERO database registration number: CRD42022304018. RESULTS: Twelve eligible studies (n = 2352) were included: 9 randomized controlled trials (RCTs, n = 1677) and 3 cohort studies (n = 675). Pooled results indicated that infertile women treated with vitamin D had a significantly increased clinical pregnancy rate compared with the control group (OR: 1.70, 95% CI: 1.24-2.34; I2 = 63%, P = 0.001). However, the implantation, biochemical pregnancy, miscarriage, and multiple pregnancy rates had no significant difference (OR: 1.86, 95% CI: 1.00-3.47; I2 = 85%, P = 0.05; OR: 1.49; 0.98-2.26; I2 = 63%, P = 0.06; OR: 0.98, 95% CI: 0.63-1.53; I2 = 0%, P = 0.94 and OR: 3.64, 95% CI: 0.58-11.98; I2 = 68%, P = 0.21). The improvement of clinical pregnancy rate in the intervention group was influenced by the vitamin D level of patients, drug type, the total vitamin D dosage, the duration, administration frequency, and daily dosage of vitamin D supplementation. The infertile women (vitamin D level < 30 ng/mL) treated with the multicomponent drugs including vitamin D (10,000-50,000 IU or 50,000-500,000 IU), or got vitamin D 1000-10,000 IU daily, lasting for 30-60 days could achieve better pregnancy outcome. CONCLUSION: To the best of our knowledge, this is the first meta-analysis systematically investigated that moderate daily dosing of vitamin D supplementation could improve the clinical pregnancy rate of infertile women and reported the effects of vitamin D supplementation parameters on pregnancy outcomes. A larger sample size and high-quality RCTs are necessary to optimize the parameters of vitamin D supplementation to help more infertile patients benefit from this therapy.


Assuntos
Infertilidade Feminina , Feminino , Humanos , Gravidez , Infertilidade Feminina/tratamento farmacológico , Infertilidade Feminina/induzido quimicamente , Vitaminas/uso terapêutico , Vitamina D/uso terapêutico , Taxa de Gravidez , Suplementos Nutricionais
6.
Fertil Steril ; 119(6): 976-984, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36805437

RESUMO

OBJECTIVE: To assess the impact of 3 different ovarian stimulation protocols on surrogate biomarkers of coagulation. DESIGN: Observational multicenter cohort study. SETTING: The study was conducted in assisted reproductive technology (ART) units. PATIENTS: Infertile women undergoing ART in 2017-2019 were included. INTERVENTIONS: None. MAIN OUTCOME MEASURE(S): Our primary outcome was the endogenous thrombin potential (ETP) assessed by the calibrated automated thrombogram. The ETP was measured at baseline (T1), on the day of ovulation triggering (T2), and 7 days after triggering (T3). Three protocols were prescribed according to the standards used and without hormonal before treatment: agonist protocol with human chorionic gonadotropin (hCG) trigger (ag-hCG), antagonist protocol with hCG trigger (atg-hCG), or GnRH agonist trigger. The evolution of ETP was compared among groups using a mixed-effects linear regression model. RESULT(S): Sixty-four women with a mean age of 37.8 years participated in the study: of which 24, 16, 24 received ag-hCG, atg-hCG, and GnRH agonist triggers, respectively. As expected, the mean serum estradiol levels in GnRH agonist trigger were statistically higher at T2 and lower at T3 than that for both ag-hCG and atg-hCG. Overall, the ETP evolution over time was statistically different between the groups. Values were similar between groups at T1 and increased at T2 in each group. The greatest difference occurred between T2 and T3 in each group. The ETP continued to increase at T3 in ag-hCG (+110 nM/L × min) and atg-hCG (+171 nM/L × min), but it remained stable in GnRH agonist trigger (-2 nM/L × min). Sex hormone-binding globulin showed persistent increase at T3 despite the fall in estradiol levels, particularly in the GnRH agonist trigger group. CONCLUSION(S): The ag-hCG and atg-hCG groups were associated with a higher hypercoagulable state at T3 than the GnRH agonist trigger group. However, our results show the persistence of a hypercoagulable state after the GnRH agonist triggering despite a sharp drop in estradiol levels. These findings may support the use of GnRH agonist trigger protocol in patients with high thrombotic risk and gives new insight into the fact that coagulation parameters could be disturbed for long time periods. CLINICAL TRIAL REGISTRATION NUMBER: NCT04188444.


Assuntos
Infertilidade Feminina , Síndrome de Hiperestimulação Ovariana , Gravidez , Humanos , Feminino , Adulto , Síndrome de Hiperestimulação Ovariana/etiologia , Síndrome de Hiperestimulação Ovariana/induzido quimicamente , Fertilização In Vitro , Infertilidade Feminina/diagnóstico , Infertilidade Feminina/terapia , Infertilidade Feminina/induzido quimicamente , Taxa de Gravidez , Hormônio Liberador de Gonadotropina , Estudos de Coortes , Indução da Ovulação/métodos , Gonadotropina Coriônica/efeitos adversos , Estradiol
7.
Clin Breast Cancer ; 23(3): 241-248, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36710145

RESUMO

In patients with early breast cancer, the combination of different systemic treatment strategies, including chemotherapy, endocrine therapy, targeted therapy, and more recently also immunotherapy has demonstrated to significantly improve their survival outcomes. However, this gain is often obtained at the cost of higher toxicity calling for the need of increased attention toward survivorship-related issues, including fertility preservation in young women. According to available guidelines, health care providers should offer oncofertility counseling to all patients with cancer diagnosed at reproductive age. Counselling should focus on the risk of gonadotoxicity of anticancer treatments and on the access to fertility preservation techniques. However, several surveys have demonstrated suboptimal implementation of these recommendations. This review aims at summarizing the available evidence on oncofertility to guide health care providers involved in the management of young women with breast cancer. Available and effective options for fertility preservation include oocyte/embryo cryopreservation or ovarian tissue cryopreservation. Patient, disease, and treatment characteristics should be carefully considered when offering these strategies. Ovarian function preservation with gonadotrophin-releasing hormone agonists during chemotherapy should be discussed and offered to every premenopausal woman concerned about developing premature ovarian insufficiency and independently of her wish to preserve fertility. Current available data confirm that pregnancy occurring after proper treatment for breast cancer is safe, both in terms of long-term clinical outcomes and for the babies. Fertility preservation and pregnancy desire should be pivotal components of the multimodal management of breast cancer in young women, and require a multidisciplinary approach based on close collaborations between oncologists and fertility specialists.


Assuntos
Neoplasias da Mama , Preservação da Fertilidade , Infertilidade Feminina , Gravidez , Feminino , Humanos , Neoplasias da Mama/tratamento farmacológico , Criopreservação , Fertilidade , Infertilidade Feminina/induzido quimicamente , Infertilidade Feminina/prevenção & controle
8.
Ecotoxicol Environ Saf ; 249: 114414, 2023 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-36516626

RESUMO

BACKGROUND: Based on self-report questionnaires, two previous epidemiological studies investigated the association between the exposure of women to antibiotics and their fertility. However, biomonitoring studies on low-dose antibiotic exposure, mainly from food and water, and its relation to the risk of infertility are missing. METHODS: Based on a case-control study design, 302 women with infertility (144 primary infertility, 158 secondary infertility) and 302 women with normal fertility, all aged 20-49 years, were recruited from Anhui Province, China, in 2020 and 2021. A total of 41 common antibiotics and two antibiotic metabolites in urine samples were determined by liquid chromatography-triple quadrupole tandem mass spectrometry (LC-QqQ-MS/MS). RESULTS: Twenty-eight antibiotics with detection rates from 10% to 100% in both cases (median concentration: ∼2.294 ng/mL) and controls (∼1.596 ng/mL) were included in the analysis. Logistic regression analysis revealed that after controlling for confounding factors, high concentrations of eight individual antibiotics (sulfamethoxazole, sulfaclozine, sulfamonomethoxine, penicillin G, chlorotetracycline, ofloxacin, norfloxacin, and cyadox) and four antibiotic classes (sulfonamides, tetracyclines, quinoxalines, and veterinary antibiotics) were related to a high risk of female infertility, with odds ratios (ORs) ranging from 1.30 to 2.86, except for chlorotetracycline (OR = 6.34), while another nine individual antibiotics (sulfamethazine, azithromycin, cefaclor, amoxicillin, oxytetracycline, pefloxacin, sarafloxacin, enrofloxacin, and florfenicol) and classes of chloramphenicol analogs and human antibiotics were related to a reduced risk of infertility, with ORs ranging from 0.70 to 0.20. Based on restricted cubic spline models after controlling for confounding factors, we observed that the relationship between all of the above protective antibiotics and infertility was nonlinear: A certain concentration could reduce the risk of female infertility while exceeding a safe dose could increase the risk of infertility. CONCLUSION: These results provide preliminary evidence that the effects of antibiotics on female fertility vary based on the active ingredient and usage and imply the importance of exposure dose. Future studies are needed to verify these results by controlling for multiple confounding factors.


Assuntos
Clortetraciclina , Infertilidade Feminina , Humanos , Feminino , Antibacterianos/análise , Espectrometria de Massas em Tandem/métodos , Clortetraciclina/análise , Infertilidade Feminina/induzido quimicamente , Infertilidade Feminina/epidemiologia , Estudos de Casos e Controles , China/epidemiologia
9.
JBRA Assist Reprod ; 26(4): 627-630, 2022 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-35916450

RESUMO

Infertility is a major problem in modern society that affects a significant number of couples around the world. Heavy metals and a number of other factors have been causally linked to infertility. The aim of this study was to determine the effect of heavy metals lead, cadmium, and copper on the epidemiology of male and female infertility. Searches for articles published from 1982 to 2020 using related keywords such as male and female infertility and heavy metals were performed in scientific databases PubMed, Google Scholar, Science Direct, and others. The results showed that, in recent years, the number of infertile individuals has increased. Various environmental, occupational, and genetic factors have been described as potential causes. Heavy metals lead, cadmium, and copper cause infertility in couples through various mechanisms, such as changes in sperm motility factors, decreased semen quality, or effects on the egg. Exposure to physical phenomena such as radiation (ionized or microwave) and heat; stress and mental disorders; chemicals from cigarettes, respiratory pollutants (lead), insecticides and pesticides; anesthetic gases; and mercury and cytotoxic drugs may also contribute to the onset of infertility.


Assuntos
Infertilidade Feminina , Infertilidade Masculina , Metais Pesados , Masculino , Feminino , Humanos , Cádmio/toxicidade , Análise do Sêmen , Cobre , Infertilidade Feminina/induzido quimicamente , Infertilidade Feminina/epidemiologia , Motilidade dos Espermatozoides , Metais Pesados/toxicidade , Infertilidade Masculina/induzido quimicamente , Infertilidade Masculina/epidemiologia
10.
Reprod Biomed Online ; 45(2): 191-201, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35725534

RESUMO

RESEARCH QUESTION: Polycystic ovary syndrome (PCOS) alters oocyte developmental competence and so the treatment of PCOS patients with assisted reproductive techniques is a major challenge for an infertility specialist. Does ovulation induction therapy with clomiphene citrate, metformin and flutamide affect abnormal PCOS follicle gene expression, its quality, and nuclear and cytoplasmic maturation in the oocyte matured in vitro? DESIGN: Fifty NMRI mice (7-8 weeks old) were randomly divided into five groups, including non-PCOS, PCOS and PCOS groups treated with clomiphene citrate (18 mg/kg body weight for 2 days), metformin (50 mg/100 g body weight for 30 days) and flutamide (10 mg/kg body weight injection for 15 days). The oocytes were collected for quantitative real-time polymerase chain reaction analysis (the evaluation of genes associated with oocyte maturation), transmission electron microscopy (the evaluation of cytoplasmic maturation) and in-vitro maturation (IVM) and IVF outcomes. RESULTS: The dysregulated expression of some genes of insulin-like growth factor (IGF) families involved in oocyte competence and steroid metabolism (e.g. Cyp19a1 and Cyp11a1), transforming growth factor beta (TGF-ß) family (e.g. Tgfb1, Gdf9, Bmp15, Inhbb and Bmpr1a), and oestrogen receptor signalling (e.g. Ncoa3, Pgr) was, to some extent, restored in the PCOS follicles following their with clomiphene citrate, metformin and flutamide. The improved cytoplasmic maturation was observed particularly in the PCOS mice treated with clomiphene citrate. The better IVM-IVF outcomes were also observed in the clomiphene citrate and metformin groups compared with the PCOS group. CONCLUSIONS: This study opens up a new perspective on knowing the effect of ovulation induction therapy on not only nuclear maturation but also ultrastructural characteristics, IVM-IVF outcomes and PCOS oocyte developmental competence.


Assuntos
Infertilidade Feminina , Metformina , Síndrome do Ovário Policístico , Animais , Peso Corporal , Clomifeno/farmacologia , Feminino , Fármacos para a Fertilidade Feminina/farmacologia , Fármacos para a Fertilidade Feminina/uso terapêutico , Flutamida/farmacologia , Humanos , Infertilidade Feminina/induzido quimicamente , Metformina/farmacologia , Camundongos , Oócitos , Indução da Ovulação/métodos , Síndrome do Ovário Policístico/tratamento farmacológico
11.
Toxicology ; 465: 153031, 2022 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-34774661

RESUMO

Per- and poly-fluoroalkyl substances (PFAS) are widespread environmental contaminants frequently detected in drinking water supplies worldwide that have been linked to a variety of adverse reproductive health outcomes in women. Compared to men, reproductive health effects in women are generally understudied while global trends in female reproduction rates are declining. Many factors may contribute to the observed decline in female reproduction, one of which is environmental contaminant exposure. PFAS have been used in home, food storage, personal care and industrial products for decades. Despite the phase-out of some legacy PFAS due to their environmental persistence and adverse health effects, alternative, short-chain and legacy PFAS mixtures will continue to pollute water and air and adversely influence women's health. Studies have shown that both long- and short-chain PFAS disrupt normal reproductive function in women through altering hormone secretion, menstrual cyclicity, and fertility. Here, we summarize the role of a variety of PFAS and PFAS mixtures in female reproductive tract dysfunction and disease. Since these chemicals may affect reproductive tissues directly or indirectly through endocrine disruption, the role of PFAS in breast, thyroid, and hypothalamic-pituitary-gonadal axis function are also discussed as the interplay between these tissues may be critical in understanding the long-term reproductive health effects of PFAS in women. A major research gap is the need for mechanism of action data - the targets for PFAS in the female reproductive and endocrine systems are not evident, but the effects are many. Given the global decline in female fecundity and the ability of PFAS to negatively impact female reproductive health, further studies are needed to examine effects on endocrine target tissues involved in the onset of reproductive disorders of women.


Assuntos
Disruptores Endócrinos/efeitos adversos , Doenças do Sistema Endócrino/induzido quimicamente , Exposição Ambiental/efeitos adversos , Poluentes Ambientais/efeitos adversos , Fertilidade/efeitos dos fármacos , Hidrocarbonetos Fluorados/efeitos adversos , Ciclo Menstrual/efeitos dos fármacos , Reprodução/efeitos dos fármacos , Doenças do Sistema Endócrino/metabolismo , Doenças do Sistema Endócrino/fisiopatologia , Feminino , Humanos , Infertilidade Feminina/induzido quimicamente , Infertilidade Feminina/metabolismo , Infertilidade Feminina/fisiopatologia , Gravidez , Complicações na Gravidez/induzido quimicamente , Complicações na Gravidez/metabolismo , Complicações na Gravidez/fisiopatologia , Prognóstico , Medição de Risco , Fatores de Risco
12.
Clin Pharmacol Ther ; 111(2): 382-390, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-33826770

RESUMO

We reviewed the available animal and human reproductive function studies of recently approved noncytotoxic oncology drugs. We reviewed the oncofertility information in the prescribing information for the US Food and Drug Administration (FDA)-approved products and/or the product information and consumer medicine information for Therapeutic Goods Administration (TGA)-approved drugs of 32 novel oncology drugs approved between 2014 and 2018 in the United States and/or Australia supplemented by a literature review for additional reproductive effects. No human studies were available on the reproductive effects of all 32 drugs. A systematic literature review of animal reproductive toxicity studies provided only very limited data with nine drugs displaying impaired male fertility, three impaired female fertility, and nine producing impaired fertility in both male and female animals. Two drugs in the study are reported to have no demonstrable impact on fertility in animal reproductive toxicity studies and nine are reported to have unknown effects on fertility. Of the 32 newly listed drugs, only 4 had recommendations regarding potential human fertility risks and accordingly advised clinicians about fertility preservation procedures for patients. The lack of human data and limited animal reproductive toxicity data raises concerns about the potential impact of these novel oncology drugs on human fertility and reproductive function. Consequently, adequate oncofertility recommendations, including for fertility preservation procedures, counselling for psychological or cost implications, and future prognosis for fertility are hindered by this paucity of relevant data. More data on human reproductive effects of novel oncology drugs is urgently required to facilitate effective use of the growing array of oncofertility care options available.


Assuntos
Antineoplásicos/efeitos adversos , Preservação da Fertilidade , Fertilidade/efeitos dos fármacos , Infertilidade Feminina/induzido quimicamente , Infertilidade Masculina/induzido quimicamente , Animais , Feminino , Humanos , Infertilidade Feminina/fisiopatologia , Infertilidade Feminina/terapia , Infertilidade Masculina/fisiopatologia , Infertilidade Masculina/terapia , Masculino , Medição de Risco , Fatores de Risco
13.
J Endocrinol ; 252(1): 15-30, 2021 11 24.
Artigo em Inglês | MEDLINE | ID: mdl-34647523

RESUMO

Immunotherapy has emerged at the forefront of cancer treatment. Checkpoint inhibitor pembrolizumab (KEYTRUDA), a chimeric antibody which targets programmed cell death protein 1 (PD-1), has been approved by the Food and Drug Administration (FDA) for use in pediatric patients with relapsed or refractory classical Hodgkin's lymphoma. However, there is currently no published data regarding the effects of pembrolizumab on the ovary of female pediatric patients. In this study, prepubertal immunocompetent and immunodeficient female mice were injected with pembrolizumab or anti-mouse PD-1 antibody. The number of primordial follicles significantly decreased post-injection of both pembrolizumab and anti-mouse PD-1 antibody in immunocompetent mice. However, no changes in follicle numbers were observed in immunodeficient nude mice. Superovulation test and vaginal opening experiments suggest that there is no difference in the number of cumulus-oocyte complexes (COCs) and the timing of puberty onset between the control and anti-mouse PD-1 antibody treatment groups, indicating that there is no effect on short-term fertility. Elevation of pro-inflammatory cytokine TNF-α following COX-2 upregulation was observed in the ovary. CD3+ T-cell infiltration was detected within some ovarian follicles and between stromal cells of the ovaries in mice following treatment with anti-mouse PD-1 antibody. Thus, PD-1 immune checkpoint blockade affects the ovarian reserve through a mechanism possibly involving inflammation following CD3+ T-cell infiltration.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Folículo Ovariano/efeitos dos fármacos , Maturidade Sexual/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Anticorpos Monoclonais Humanizados/efeitos adversos , Antineoplásicos Imunológicos/efeitos adversos , Antineoplásicos Imunológicos/farmacologia , Contagem de Células , Feminino , Inibidores de Checkpoint Imunológico/efeitos adversos , Inibidores de Checkpoint Imunológico/farmacologia , Infertilidade Feminina/induzido quimicamente , Infertilidade Feminina/patologia , Camundongos , Camundongos Nus , Oócitos/citologia , Oócitos/efeitos dos fármacos , Reserva Ovariana/efeitos dos fármacos , Ovário/efeitos dos fármacos , Ovário/fisiologia , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptor de Morte Celular Programada 1/imunologia
14.
J Assist Reprod Genet ; 38(11): 3047-3055, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34495476

RESUMO

PURPOSE: To develop an innovative machine learning (ML) model that predicts personalized risk of primary ovarian insufficiency (POI) after chemotherapy for reproductive-aged women. Currently, individualized prediction of a patient's risk of POI is challenging. METHODS: Authors of published studies examining POI after gonadotoxic therapy were contacted, and six authors shared their de-identified data (N = 435). A composite outcome for POI was determined for each patient and validated by 3 authors. The primary dataset was partitioned into training and test sets; random forest binary classifiers were trained, and mean prediction scores were computed. Institutional data collected from a cross-sectional survey of cancer survivors (N = 117) was used as another independent validation set. RESULTS: Our model predicted individualized risk of POI with an accuracy of 88% (area under the ROC 0.87, 95% CI: 0.77-0.96; p < 0.001). Mean prediction scores for patients who developed POI and who did not were 0.60 and 0.38 (t-test p < 0.001), respectively. Highly weighted variables included age, chemotherapy dose, prior treatment, smoking, and baseline diminished ovarian reserve. CONCLUSION: We developed an ML-based model to estimate personalized risk of POI after chemotherapy. Our web-based calculator will be a user-friendly decision aid for individualizing risk prediction in oncofertility consultations.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Infertilidade Feminina/diagnóstico , Modelos Estatísticos , Neoplasias/tratamento farmacológico , Doenças Ovarianas/diagnóstico , Medicina de Precisão , Insuficiência Ovariana Primária/diagnóstico , Adulto , Sobreviventes de Câncer , Estudos Transversais , Feminino , Humanos , Infertilidade Feminina/induzido quimicamente , Infertilidade Feminina/epidemiologia , Neoplasias/patologia , Doenças Ovarianas/induzido quimicamente , Doenças Ovarianas/epidemiologia , Insuficiência Ovariana Primária/induzido quimicamente , Insuficiência Ovariana Primária/epidemiologia , Medição de Risco/métodos , Inquéritos e Questionários , Estados Unidos/epidemiologia
15.
Front Endocrinol (Lausanne) ; 12: 689532, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34557157

RESUMO

Background: Whilst the ability of AMH to induce the regression of the Müllerian ducts in the male fetus is well appreciated, AMH has additional biological actions in relation to steroid biosynthesis and ovarian follicle dynamics. An understanding of the physiology of AMH illuminates the potential therapeutic utility of AMH to protect the ovarian reserve during chemotherapy and in the treatment of female malignancies. The translation of the biological actions of AMH into clinical applications is an emerging focus of research, with promising preliminary results. Objective and Rationale: Studies indicate AMH restrains primordial follicle development, thus administration of AMH during chemotherapy may protect the ovarian reserve by preventing the mass activation of primordial follicles. As AMH induces regression of tissues expressing the AMH receptor (AMHRII), administration of AMH may inhibit growth of malignancies expressing AMHR II. This review evaluates the biological actions of AMH in females and appraises human clinical applications. Search Methods: A comprehensive search of the Medline and EMBASE databases seeking articles related to the physiological functions and therapeutic applications of AMH was conducted in July 2021. The search was limited to studies published in English. Outcomes: AMH regulates primordial follicle recruitment and moderates sex steroid production through the inhibition of transcription of enzymes in the steroid biosynthetic pathway, primarily aromatase and 17α-hydroxylase/17,20-lyase. Preliminary data indicates that administration of AMH to mice during chemotherapy conveys a degree of protection to the ovarian reserve. Administration of AMH at the time of ovarian tissue grafting has the potential to restrain uncontrolled primordial follicle growth during revascularization. Numerous studies demonstrate AMH induced regression of AMHR II expressing malignancies. As this action occurs via a different mechanism to traditional chemotherapeutic agents, AMH has the capacity to inhibit proliferation of chemo-resistant ovarian cancer cells and cancer stem cells. Wider Implications: To date, AMH has not been administered to humans. Data identified in this review suggests administration of AMH would be safe and well tolerated. Administration of AMH during chemotherapy may provide a synchronistic benefit to women with an AMHR II expressing malignancy, protecting the ovarian reserve whilst the cancer is treated by dual mechanisms.


Assuntos
Hormônio Antimülleriano/fisiologia , Antineoplásicos/efeitos adversos , Preservação da Fertilidade/métodos , Infertilidade Feminina/induzido quimicamente , Neoplasias/tratamento farmacológico , Reserva Ovariana , Feminino , Humanos
16.
Ann Hematol ; 100(11): 2831-2841, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34536088

RESUMO

Since the survival rates of pediatric patients undergoing cancer treatment or hematopoietic stem cell transplantation (HSCT) have increased rapidly in recent decades, the late effects of treatment are now an important focus of patient care. Access to fertility preservation (FP) procedures as well as their financing differs considerably across Europe. However, some countries in Europe have recently changed the legal basis for financing FP procedures; therefore, the implementation of structures is mandatory to give patients access to FP. In this prospective cohort study, we characterized the process for establishing pediatric fertility counseling, including the development of an in-house standard procedure for recommendations regarding FP with potentially gonadotoxic treatment and valuating data from all FP counseling sessions. All data concerning patient characteristics (pubertal status, disease group) and recommendation of FP measures were prospectively collected and adoption of FP measures analyzed. Prior to the establishment of a structured process for FP in our pediatric oncology and stem cell transplantation center, there was no standardized FP counseling. We demonstrate that with the establishment of an inhouse standard procedure, it is possible to give consistent yet individualized FP counseling to approximately 90% of our patients facing gonadotoxic treatment, counseling over 200 patients between 2017 and 2019. This pilot study could potentially be adapted in other pediatric hematology, oncology, and stem cell transplantation centers to allow a more standardized handling of FP counseling for all patients facing gonadotoxic treatment.


Assuntos
Aconselhamento/métodos , Preservação da Fertilidade/métodos , Adolescente , Antineoplásicos/efeitos adversos , Criança , Pré-Escolar , Criopreservação , Feminino , Preservação da Fertilidade/economia , Preservação da Fertilidade/normas , Transplante de Células-Tronco Hematopoéticas , Humanos , Lactente , Infertilidade Feminina/induzido quimicamente , Infertilidade Feminina/etiologia , Infertilidade Feminina/prevenção & controle , Infertilidade Masculina/induzido quimicamente , Infertilidade Masculina/etiologia , Infertilidade Masculina/prevenção & controle , Masculino , Neoplasias/terapia , Recuperação de Oócitos , Ovário/transplante , Estudos Prospectivos , Puberdade , Lesões por Radiação/prevenção & controle , Radioterapia/efeitos adversos , Preservação do Sêmen , Condicionamento Pré-Transplante/efeitos adversos , Adulto Jovem
17.
J Assist Reprod Genet ; 38(10): 2713-2721, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34370210

RESUMO

PURPOSE: To characterize female pediatric and adolescent patients seen for fertility preservation consultation at an academic medical center and to describe the association between demographic or clinical factors and the use of fertility preservation treatment (FPT). METHODS: This is a retrospective chart analysis of female pediatric and adolescent patients seen for fertility preservation consultation at an academic fertility center over a 14-year period from 2005 to 2019. RESULTS: One hundred six females aged 3-21 years were seen for fertility preservation consultation with a mean age of 16.6 years. Diagnoses included hematologic malignancies (41.5%), gynecologic malignancies (9.4%), other malignancies (31.1%), non-malignant hematologic disease (14.2%), and non-malignant conditions (3.8%). Overall, 64.2% of subjects pursued fertility preservation, including oocyte cryopreservation (35.8%) and ovarian tissue cryopreservation (23.6%). Overall, age, minority race, diagnosis, time since diagnosis, and median household income were not significantly associated with odds of completing an FPT procedure. Among all patients, those who underwent gonadotoxic therapy prior to consultation had a lower odds of receiving FPT (OR= 0.24, 95% CI 0.10-0.55). Among patients without chemotherapy exposure, no factors were associated with FPT. CONCLUSIONS: Among pediatric and adolescent patients at an academic center undergoing a fertility preservation consultation, there were no socioeconomic or clinical barriers to FPT use in those who had not yet undergone gonadotoxic therapy. The only factor that was negatively associated with odds of pursuing FPT was prior chemotherapy exposure.


Assuntos
Antineoplásicos/efeitos adversos , Fármacos para a Fertilidade Feminina/administração & dosagem , Preservação da Fertilidade/métodos , Infertilidade Feminina/terapia , Neoplasias/tratamento farmacológico , Ovário/efeitos dos fármacos , Adolescente , Adulto , Criança , Pré-Escolar , Feminino , Humanos , Infertilidade Feminina/induzido quimicamente , Neoplasias/patologia , Estudos Retrospectivos , Adulto Jovem
18.
Front Endocrinol (Lausanne) ; 12: 672532, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34305812

RESUMO

Glyphosate base herbicides (GBHs) are the most widely applied pesticides in the world and are mainly used in association with GBH-tolerant crop varieties. Indiscriminate and negligent use of GBHs has promoted the emergence of glyphosate resistant weeds, and consequently the rise in the use of these herbicides. Glyphosate, the active ingredient of all GBHs, is combined with other chemicals known as co-formulants that enhance the herbicide action. Nowadays, the safety of glyphosate and its formulations remain to be a controversial issue, as evidence is not conclusive whether the adverse effects are caused by GBH or glyphosate, and little is known about the contribution of co-formulants to the toxicity of herbicides. Currently, alarmingly increased levels of glyphosate have been detected in different environmental matrixes and in foodstuff, becoming an issue of social concern. Some in vitro and in vivo studies have shown that glyphosate and its formulations exhibit estrogen-like properties, and growing evidence has indicated they may disrupt normal endocrine function, with adverse consequences for reproductive health. Moreover, multigenerational effects have been reported and epigenetic mechanisms have been proved to be involved in the alterations induced by the herbicide. In this review, we provide an overview of: i) the routes and levels of human exposure to GBHs, ii) the potential estrogenic effects of glyphosate and GBHs in cell culture and animal models, iii) their long-term effects on female fertility and mechanisms of action, and iv) the consequences on health of successive generations.


Assuntos
Exposição Ambiental/efeitos adversos , Glicina/análogos & derivados , Herbicidas/toxicidade , Infertilidade Feminina/induzido quimicamente , Reprodução/efeitos dos fármacos , Feminino , Glicina/toxicidade , Humanos
19.
Psychopharmacol Bull ; 51(2): 131-148, 2021 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-34092827

RESUMO

Infertility, the inability to establish a clinical pregnancy after 12 months of regular unprotected sexual intercourse, is caused by a wide variety of both male and female factors. Infertility is estimated to affect between 8-12% of couples trying to conceive globally. Female factor infertility can be subdivided into the following broad categories: ovulatory dysfunction, fallopian tubal disease, uterine causes, and oocyte quality. Hyperprolactinemia causes ovulary dysfunction along with other hormonal abnormalities, such as decreased estrogen, which can lead to infertility. In this regard, antipsychotics are commonly used for both schizophrenia and bipolar disorder. The use of these medications can be associated with hyperprolactinemia and hyperprolactinemia associated infertility. Antipsychotic-induced hyperprolactinemia occurs through blockade of D2 receptors on lactotroph cells of the anterior pituitary gland. Discontinuation of the hyperprolactinemia-inducing antipsychotic is an option, but this may worsen the patient's psychosis or mood. If antipsychotics are determined to be the culprit of infertility, the degree of hyperprolactinemia symptoms, length of treatment with the antipsychotic, and risk of relapse should be assessed prior to discontinuation, reduction, or switching of antipsychotic medications. The treatment of a women's mental health and her desire to have children should always be considered as treatment may influence fertility while on the medication.


Assuntos
Antipsicóticos , Transtorno Bipolar , Hiperprolactinemia , Infertilidade Feminina/induzido quimicamente , Esquizofrenia , Antipsicóticos/efeitos adversos , Transtorno Bipolar/tratamento farmacológico , Feminino , Humanos , Hiperprolactinemia/induzido quimicamente , Esquizofrenia/tratamento farmacológico
20.
Lupus ; 30(9): 1509-1514, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34053364

RESUMO

Systemic lupus erythematosus (SLE) often affects females of reproductive age and Cyclophosphamide, an alkylating agent leading to premature ovarian insufficiency (POF) and labelled category D for pregnancy is used as induction therapy for severe manifestations of lupus. There have been multiple case series reflecting variable outcomes of pregnancies after cyclophosphamide use for cancers and autoimmune diseases. With increasing maternal age, we have an increasing population of lupus patients who may wish to conceive after having received cyclophosphamide therapy. The objective of our study was to improve our understanding of the impact of cyclophosphamide exposure on fertility and pregnancy outcomes in patients with SLE.We retrospectively reviewed the charts of all patients who had received intravenous cyclophosphamide at our academic institute in the time period from 2000-2018 and identified 440 patients which included 157 female patients of reproductive age. There were 37 documented pregnancies after the cyclophosphamide infusion, of which 23 patients had successful outcomes; 4 elective abortion and 10 miscarriages. There were 17 patients who developed POF, of which 7 also had end stage renal disease. The average cumulative dose of cyclophosphamide in the patients who had successful pregnancy was 4080.37 mg compared to 2806.25 mg in those who had a miscarriage (p 0.164) and 5526.47 mg in those who developed POF (p 0.046). Using multiple regressions to evaluate risk factors impacting pregnancy outcomes, when taken as a set, the predictors including race, serological profile, exposure to steroids and Mycophenolate mofetil, age at cyclophosphamide infusion, age at pregnancy, and cumulative cyclophosphamide dose accounted for 46.29% of the variance in outcome of pregnancy (p 0.23) and 39.58% of the variance in development of premature ovarian failure (p 0.008). We noted statistical significance in the impact of maternal age at time of pregnancy (p 0.04) and duration of time between the last infusions to subsequent pregnancy (p 0.02) to pregnancy outcome.Our findings suggest that a longer time interval between the last cyclophosphamide infusion and subsequent pregnancy was favorable for a successful outcome and higher cumulative cyclophosphamide dose is more likely to be associated with premature ovarian failure.


Assuntos
Ciclofosfamida/efeitos adversos , Imunossupressores/efeitos adversos , Infertilidade Feminina/induzido quimicamente , Lúpus Eritematoso Sistêmico/tratamento farmacológico , Insuficiência Ovariana Primária/induzido quimicamente , Administração Intravenosa , Adulto , Ciclofosfamida/administração & dosagem , Relação Dose-Resposta a Droga , Feminino , Fertilidade/efeitos dos fármacos , Humanos , Imunossupressores/administração & dosagem , Gravidez , Resultado da Gravidez , Estudos Retrospectivos , Fatores de Tempo , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...