Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Reprod Biomed Online ; 49(2): 103978, 2024 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-38805862

RESUMO

Patients with haematologic malignancies represent one of the most common groups referred for fertility preservation before gonadotoxic oncological treatment. The aim of this systematic review and meta-analysis was to evaluate the effect of haematologic cancer on ovarian reserve and response to ovarian stimulation compared with healthy controls. A total of eight observative studies were included in the final quantitative analysis. Despite a younger age (mean difference -4.17, 95% CI -6.20 to -2.14; P < 0.0001), patients with haematologic malignancy had lower serum anti-Müllerian hormone levels compared with the control group (MD -1.04, 95% CI -1.80 to -0.29; P = 0.007). The marginally higher total recombinant FSH dose (MD 632.32, 95% CI -187.60 to 1452.24; P = 0.13) and significantly lower peak oestradiol serum level (MD -994.05, 95% CI -1962.09 to -26.02; P = 0.04) were demonstrated in the study group compared with the healthy controls. A similar number of retrieved oocytes were achieved in both groups (MD 0.20, 95% CI -0.80 to 1.20; P = 0.69). In conclusion, haematologic malignancies may detrimentally affect ovarian function manifesting in decreased AMH serum levels despite a younger age compared with healthy controls. This effect can be overcome by the application of relevant IVF protocols and stimulation doses to achieve an adequate oocyte yield.

2.
Int J Mol Sci ; 24(22)2023 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-38003481

RESUMO

Dormant primordial follicles (PMF), which constitute the ovarian reserve, are recruited continuously into the cohort of growing follicles in the ovary throughout female reproductive life. Gonadotoxic chemotherapy was shown to diminish the ovarian reserve pool, to destroy growing follicle population, and to cause premature ovarian insufficiency (POI). Three primary mechanisms have been proposed to account for this chemotherapy-induced PMF depletion: either indirectly via over-recruitment of PMF, by stromal damage, or through direct toxicity effects on PMF. Preventative pharmacological agents intervening in these ovotoxic mechanisms may be ideal candidates for fertility preservation (FP). This manuscript reviews the mechanisms that disrupt follicle dormancy causing depletion of the ovarian reserve. It describes the most widely studied experimental inhibitors that have been deployed in attempts to counteract these affects and prevent follicle depletion.


Assuntos
Antineoplásicos , Reserva Ovariana , Humanos , Feminino , Folículo Ovariano , Ovário , Apoptose , Antineoplásicos/efeitos adversos
3.
Hum Reprod ; 38(6): 1086-1098, 2023 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-37015102

RESUMO

STUDY QUESTION: To what extent and how does combined administration of the follicle activation pathway suppressive agents temsirolimus (Tem) and c-terminus recombinant anti-Müllerian hormone (rAMH) protect against chemotherapy-induced ovarian reserve loss? SUMMARY ANSWER: Combined administration of Tem and rAMH completely prevents cyclophosphamide (Cy)-induced follicle depletion and protects the ovarian reserve in mice, primarily via primordial follicle (PMF) suppression of activation and to a lesser degree by reducing apoptosis. WHAT IS KNOWN ALREADY: There is conflicting evidence regarding the contributory roles of apoptosis and follicle activation in chemotherapy-induced PMF loss. Tem, a mammalian target of rapamycin (mTOR) inhibitor, reduces activity of the phosphoinositide 3-kinases-phosphatase and tensin homolog (PI3K-PTEN) pathway which provides intrinsic regulation of PMF activation. Anti-Müllerian hormone (AMH), secreted by early growing follicles, is an extrinsic regulator of PMF activation. STUDY DESIGN, SIZE, DURATION: Whole ovaries of 12-day-old mice were cultured ex vivo for 7 days in the presence of Cy ± rAMH or Tem. Eight-week-old mice were randomized into eight treatment groups: vehicle control/rAMH/Tem/Cy/Tem + rAMH/Cy + Tem/Cy + rAMH/Cy + Tem + rAMH. Twelve hours after treatment, ovaries were removed for DNA damage analysis, and 24 h after treatment either for analysis of PI3K pathway proteins or to be fixed and immunostained for analyses of proliferation and apoptosis. Three or 21 days following treatment, ovaries were fixed and sectioned for follicle counting. PARTICIPANTS/MATERIALS, SETTING, METHODS: Hematoxylin and eosin staining was used for differential follicle counts of primordial, primary, and secondary follicles in ex vivo (n = 16-18 ovaries per group) and in vivo ovaries (n = 8 mice per group). Histological analyses were carried out to measure proliferation by quantifying Ki-67-positive granulosa cells in primary follicles (n = 4 mice per group). DNA damage and apoptosis were measured by quantification of phosphorylated form of histone 2AX (γH2AX) and cleaved poly (ADP-ribose) polymerase (cPARP)-positive PMF oocytes, respectively (n = 8 mice per group). Protein extracts from whole ovaries were analyzed by western blotting. MAIN RESULTS AND THE ROLE OF CHANCE: In vivo experiments show that treatment with Cy alone caused significant loss of PMF reserve (32 ± 2.12 versus 144 ± 2.8 in control, P < 0.001), and this was significantly attenuated by treatment with either Tem (P < 0.001) or rAMH (P < 0.001). Combined cotreatment with Cy + Tem + rAMH provided complete protection of the PMF reserve, with no significant difference in numbers of PMF versus untreated animals. Similar results were demonstrated in the ex vivo experiments. Proliferation marker Ki-67 staining was significantly reduced in granulosa cells of primary follicles in the Cy + Tem + rAMH group compared with Cy alone group (after 24 h in vivo administration of Cy, 16% versus 65%, respectively; P < 0.001). Protein analysis demonstrated not significant increased phosphorylation of follicle activation proteins rpS6 and mTOR with in vivo administration of Cy alone (1.9 and 1.4 times the control ovaries, respectively), and this was reduced to below control levels in the Cy + Tem + rAMH group (P < 0.01). The Cy + Tem + rAMH combined cotreatment protected the follicle reservoir via inhibition of Cy-induced upregulation of the PI3K signaling pathway, together with replacement of AMH suppression of PMF activation with rAMH, implying a complementary effect of the two inhibitors. The DNA damage marker γH2AX was highly positive in PMF oocytes from Cy-treated ovaries 12 h after treatment, compared with controls (94% versus 59%, respectively, P < 0.001) and was significantly reduced to (69%) in Cy + Tem + rAMH cotreated ovaries (P < 0.001). However, only 22% of PMF oocytes of the Cy group showed apoptosis at 24 h, and this was significantly reduced (12%) in ovaries after treatment with Cy + Tem + rAMH (P < 0.01). This suggests that it is not possible to equate DNA damage with oocyte death, and also indicates that less than one-third of the total PMF loss can be attributed to apoptosis, implying that most of the PMF depletion results from PMF activation but that both mechanisms play a significant role. LARGE SCALE DATA: N/A. LIMITATIONS, REASONS FOR CAUTION: The experimental design was limited by the selection of one time point for analysis of PMF activation and apoptosis (i.e. 24 h after Cy administration), although DNA damage was measured at 12 h after Cy administration and any impact on short-term follicle dynamics at 3 days after treatment. Protein analysis was conducted on whole ovary lysates therefore the protein changes identified cannot be localized to specific cells within the ovary. However, this complementary assay showed that there was activation in the ovary through massive reduction in the phosphorylation of key proteins in the PI3K cascade (rpS6 and mTOR), which is consistent with the sequence of events after Cy administration. WIDER IMPLICATIONS OF THE FINDINGS: Understanding the complementary nature of different follicle activation pathways and the impact of their suppression in prevention of chemotherapy-induced ovotoxic damage, as well as their involvement in DNA damage inhibition, provides an interesting direction for future research, and the potential for noninvasive pharmacological fertility preservation. STUDY FUNDING/COMPETING INTEREST(S): This work was supported by a grant from the Morris Kahn Foundation. The authors declare no conflicts of interest.


Assuntos
Antineoplásicos , Reserva Ovariana , Feminino , Animais , Camundongos , Reserva Ovariana/fisiologia , Hormônio Antimülleriano/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Antígeno Ki-67 , Ciclofosfamida , Serina-Treonina Quinases TOR , Mamíferos/metabolismo
4.
J Assist Reprod Genet ; 39(12): 2827-2834, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36418617

RESUMO

PURPOSE: To assess the feasibility, effectiveness, and reproductive outcomes of transplantation of tiny cryopreserved ovarian pieces through a pipelle cannula during laparoscopic surgery. METHODS: A retrospective study of patients who underwent ovarian tissue transplantation for fertility restoration between 2004 and 2022. The "pipelle group" had their ovarian cortex cut into tiny pieces of ~ 1-2 mm3 before cryopreservation. The pieces were too small to be handled and transplanted via standard laparoscopic tools. Transplantation was performed using a pipelle cannula during laparoscopic surgery. The "control group" underwent transplants of ovarian cortex pieces 1-2 mm thick, measuring approximately 25-50 mm2 pieces, using standard procedures. RESULTS: The pipelle group consisted of 4 patients aged 19, 21, 27, and 28 years old at ovarian tissue cryopreservation (OTC). The control group consisted of 14 patients aged 21-30 years old. All pipelle patients restored their endocrine activity, and all of them conceived. FSH levels dropped during the first 3 months following the pipelle transplant. IVF cycle outcomes were similar for both groups. All patients from the pipelle group conceived, resulting in 5 pregnancies and 4 live births (one patient had 2 deliveries, and one additional pregnancy is ongoing), compared to the control group, where 8 patients achieved a total of 20 pregnancies and 18 live births. CONCLUSION: Pipelle transplantation for tiny cryopreserved ovarian pieces is feasible and effective. This study opens a door for patients who had their ovaries cut into small pieces and may even simplify the procedure in some instances, making ovarian transplant more accessible. TRIAL REGISTRATION: (#6531-19-SMC) [18/09/2019].


Assuntos
Preservação da Fertilidade , Gravidez , Feminino , Humanos , Adulto Jovem , Adulto , Preservação da Fertilidade/métodos , Estudos Retrospectivos , Ovário/transplante , Criopreservação/métodos , Nascido Vivo
5.
Hum Reprod Update ; 28(2): 296-311, 2022 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-35043201

RESUMO

BACKGROUND: BReast CAncer (BRCA) genes are extensively studied in the context of fertility and reproductive aging. BRCA proteins are part of the DNA repair Fanconi anemia (FA)/BRCA pathway, in which more than 20 proteins are implicated. According to which gene is mutated and which interactions are lost owing to the mutation, carriers and patients with monoallelic or biallelic FA/BRCA mutations exhibit very different phenotypes, from overt FA to cancer predisposition or no pathological implications. The effect of the so far neglected non-BRCA FA mutations on fertility also deserves consideration. OBJECTIVE AND RATIONALE: As improved treatments allow a longer life expectancy in patients with biallelic FA mutations and overt FA, infertility is emerging as a predominant feature. We thus reviewed the mechanisms for such a manifestation, as well as whether they also occur in monoallelic carriers of FA non-BRCA mutations. SEARCH METHODS: Electronic databases PUBMED, EMBASE and CENTRAL were searched using the following term: 'fanconi' OR 'FANC' OR 'AND' 'fertility' OR 'pregnancy' OR 'ovarian reserve' OR 'spermatogenesis' OR 'hypogonadism'. All pertinent reports in the English-language literature were retrieved until May 2021 and the reference lists were systematically searched in order to identify any potential additional studies. OUTCOMES: Biallelic FA mutations causing overt FA disease are associated with premature ovarian insufficiency (POI) occurring in the fourth decade in women and with primary non-obstructive azoospermia (NOA) in men. Hypogonadism in FA patients seems mainly associated with a defect in primordial germ cell proliferation in fetal life. In recent small, exploratory whole-exome sequencing studies, biallelic clinically occult mutations in the FA complementation group A (Fanca) and M (Fancm) genes were found in otherwise healthy patients with isolated NOA or POI, and also monoallelic carrier status for a loss-of-function mutation in Fanca has been implicated as a possible cause for POI. In those patients with known monoallelic FA mutations undergoing pre-implantation genetic testing, poor assisted reproduction outcomes are reported. However, the mechanisms underlying the repeated failures and the high miscarriage rates observed are not fully known. WIDER IMPLICATIONS: The so far 'neglected' members of the FA/BRCA family will likely emerge as a relevant focus of investigation in the genetics of reproduction. Several (rather than a single) non-BRCA genes might be implicated. State-of-the-art methods, such as whole-genome/exome sequencing, and further exploratory studies are required to understand the prevalence and mechanisms for occult FA mutations in infertility and recurrent miscarriage.


Assuntos
Azoospermia , Neoplasias da Mama , Anemia de Fanconi , Hipogonadismo , Insuficiência Ovariana Primária , DNA Helicases/genética , Anemia de Fanconi/genética , Feminino , Humanos , Masculino , Mutação , Reprodução
6.
Fertil Steril ; 115(5): 1239-1249, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33485607

RESUMO

OBJECTIVE: To investigate mechanisms of primordial follicle (PMF) loss in vivo in human ovaries shortly after alkylating agent (AA) chemotherapy. DESIGN: Cohort study. SETTING: Tertiary university medical center. PATIENT(S): Ninety-six women aged 15-39 years who underwent ovarian tissue cryopreservation for fertility preservation. INTERVENTION(S): Fresh ovarian tissue samples were harvested from women treated with AA (n = 24) or non-AA (n = 24) chemotherapy <6 months after treatment and age-matched untreated women (n = 48). MAIN OUTCOME MEASURE(S): Differential follicle counts, time from chemotherapy exposure, immunostaining for apoptosis (cleaved caspase-3) and FOXO3A on tissue harvested within ultrashort time intervals (4-12 days), collagen (Sirius red) and neovascularization (CD34). RESULT(S): AA-treated ovaries had significant loss of PMFs, and significant increase in absolute numbers of growing follicles compared with untreated control ovaries. The number of growing follicles was inversely correlated with time from chemotherapy. Representative staining for FOXO3A observed decreased nuclear localization in PMF oocytes in AA-treated ovaries removed within the ultrashort time interval compared with untreated ovaries. Neither significant loss of PMFs, increase in growing follicles, nor decrease in nuclear FOXO3A were observed in non-AA-treated ovaries. No increased expression of cleaved caspase-3 was seen in PMFs within the ultrashort time interval after AA or non-AA chemotherapy. Significant stromal fibrosis and neovascularization were observed in AA-treated ovaries only after follicle loss had already occurred (4-6 months). CONCLUSION(S): Follicle activation occurs in vivo in ovaries of patients treated with AA, indicating a pathologic mechanism which may contribute to chemotherapy-induced follicle loss.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Apoptose/efeitos dos fármacos , Folículo Ovariano/efeitos dos fármacos , Ovário/efeitos dos fármacos , Adolescente , Adulto , Apoptose/fisiologia , Estudos de Casos e Controles , Estudos de Coortes , Criopreservação , Feminino , Preservação da Fertilidade , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Oócitos/efeitos dos fármacos , Oócitos/fisiologia , Folículo Ovariano/fisiologia , Ovário/patologia , Ovário/fisiologia , Adulto Jovem
7.
Fertil Steril ; 114(2): 388-397, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32605799

RESUMO

OBJECTIVE: To report ovarian tissue autotransplantation (AT) results and describe the relationship between technical and clinical factors and outcomes. DESIGN: Multicenter retrospective cohort study. SETTING: Tertiary medical centers. PATIENT(S): Infertile patients who had stored ovarian tissue before sterilizing treatment and returned for AT with the aim of conceiving. INTERVENTIONS(S): Ovarian tissue cryopreservation (OTC) and AT, endocrine monitoring, in vitro fertilization. MAIN OUTCOME MEASURE(S): Endocrine performance, pregnancy and live-birth rates. RESULT(S): From 2004 to 2018, 70 patients underwent 87 ATs. Sixty patients undergoing 70 ATs met the inclusion criteria. After AT, menses returned in 94% of patients and median FSH dropped from 68 to 19 IU/mL. Fifty pregnancies and 44 deliveries were attained, with 50% of women achieving at least one pregnancy and 41.6% at least one delivery. Twelve patients underwent AT more than once and had their endocrine activity restored in case menses recurred after the first transplantation. Repeated transplantations yielded five live births in three patients, two of whom had already given birth after the first transplantation. Preharvesting chemotherapy was not associated with inferior outcomes. Of seven patients whose pelvis was exposed to radiation before AT, four conceived and delivered. Neither tissue dimensions nor surgical approach affected fertility outcomes. CONCLUSION(S): OTC is highly effective at restoring fertility in sterilized patients, and prior exposure to chemotherapy should not be considered a contraindication. Repeated AT should be contemplated in case of graft malfunction, especially if previous transplantation was successful. In selected cases, conception and delivery may be feasible after pelvic exposure to radiation.


Assuntos
Preservação da Fertilidade , Infertilidade Feminina/terapia , Ovário/transplante , Técnicas de Reprodução Assistida , Adulto , Bélgica , Sobreviventes de Câncer , Feminino , Fertilidade , Preservação da Fertilidade/efeitos adversos , Humanos , Infertilidade Feminina/diagnóstico , Infertilidade Feminina/fisiopatologia , Israel , Nascido Vivo , Missouri , Ovário/fisiopatologia , Gravidez , Taxa de Gravidez , Técnicas de Reprodução Assistida/efeitos adversos , Estudos Retrospectivos , Fatores de Tempo , Transplante Autólogo , Resultado do Tratamento , Adulto Jovem
8.
Reproduction ; 158(5): F35-F44, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31394506

RESUMO

Ovarian tissue cryopreservation and transplantation (OTCP-TP) has progressed over the past decade from a revolutionary experimental procedure to a well-accepted treatment in many centers for young patients with a high risk of ovarian failure after cancer treatment. The procedure is remarkably successful, with studies reporting return of ovarian function in up to 95% of graft recipients and pregnancy rates of between 30 and 50%. The most significant limitation of OTCP-TP is the massive loss of follicles that occurs following transplantation, which is primarily attributed to ischemic damage and follicle activation. We review the current approaches to reducing follicle loss and maximizing graft lifespan via pharmacological agents which reduce ischemic damage and follicle activation. We further discuss the value and disadvantage of inducing follicle activation in the graft as a means of generating mature follicles in the immediate short term.


Assuntos
Criopreservação/métodos , Preservação da Fertilidade/efeitos adversos , Folículo Ovariano/citologia , Folículo Ovariano/transplante , Reserva Ovariana/fisiologia , Ovário , Contagem de Células , Feminino , Preservação da Fertilidade/métodos , Humanos , Ovário/transplante , Gravidez , Taxa de Gravidez
9.
J Assist Reprod Genet ; 36(9): 1793-1803, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31250176

RESUMO

PURPOSE: To determine whether pharmacological administration of recombinant human anti-Mullerian hormone (rAMH) protects the ovarian reserve and preserves fertility without interfering with anti-tumoural cytotoxic action of chemotherapy. METHODS: Intraperitoneal delivery of rAMH and ovarian post-receptor activity were assessed with immunohistochemistry and western blot. Differential follicle counts and reproductive outcomes were assessed after cyclophosphamide (Cy) administration, with/without concurrent administration of rAMH. Interference of rAMH with Cy chemotoxicity was assessed on a human breast cancer cell line and an in vivo mouse model of human leukaemia. RESULTS: rAMH reached the ovary after intraperitoneal injection and demonstrated post-receptor bioactivity. Cy administration in mice caused primordial follicle activation, as shown by a decrease in primordial follicle population accompanied by an increase in early growing follicles and granulosa cell proliferation. Co-administration of rAMH reduced follicle activation, thereby protecting the primordial follicle reserve, and improving long-term fertility and reproductive outcomes. rAMH co-administration did not interfere with the cytotoxic actions of Cy in vitro on breast cancer cell line or in vivo in a model of human leukaemia. CONCLUSION: This study demonstrates that rAMH is bioactive in the ovary for a limited time, and that pharmacological administration of rAMH during chemotherapy treatment reduces follicle activation and primordial follicle loss and significantly improves reproductive outcomes in a mouse model, and does not interfere with the therapeutic actions of the treatment. Further investigation is necessary to determine whether it has similar protective effects in the human ovary.


Assuntos
Hormônio Antimülleriano/farmacologia , Ciclofosfamida/farmacologia , Preservação da Fertilidade/métodos , Reserva Ovariana/efeitos dos fármacos , Animais , Hormônio Antimülleriano/genética , Antineoplásicos Alquilantes/efeitos adversos , Antineoplásicos Alquilantes/farmacologia , Linhagem Celular Tumoral , Ciclofosfamida/efeitos adversos , Feminino , Humanos , Leucemia Experimental/tratamento farmacológico , Masculino , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos NOD , Folículo Ovariano/efeitos dos fármacos , Folículo Ovariano/patologia , Reserva Ovariana/fisiologia , Gravidez , Taxa de Gravidez , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacologia
10.
Reprod Biomed Soc Online ; 5: 82-92, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30014045

RESUMO

Medical egg freezing (MEF) is being recommended increasingly for women at risk of losing their reproductive ability due to cancer chemotherapy or other fertility-threatening medical conditions. This first, binational, ethnographic study of women who had undergone MEF sought to explore women's experiences under two different funding systems: (i) the USA, where the cost of MEF is rarely covered by private or state health insurance; and (ii) Israel, where the cost of MEF is covered by national health insurance. Women were recruited from four American and two Israeli in-vitro fertilization clinics where MEF is offered. In-depth, semi-structured interviews were conducted with 45 women (33 Americans, 12 Israelis) who had completed at least one cycle of MEF. All of the Israeli women had cancer diagnoses, but were not faced with the additional burden of funding an MEF cycle. In marked contrast, the American women - 23 with cancer diagnoses and 10 with other fertility-threatening medical conditions - struggled, along with their families, to 'piece together' MEF funding, which added significant financial pressure to an already stressful situation. Given the high priority that both American and Israeli women in this study placed on survival and future motherhood, it is suggested that insurance funding for MEF should be mandated in the USA, as it is in Israel. This article concludes by describing new state legislative efforts in this regard.

11.
Minerva Ginecol ; 70(4): 456-464, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29696942

RESUMO

An ongoing increase in survival rates among young leukemia patients is accompanied by a growing attention to possible long-term complications such as chemotherapy induced ovarian insufficiency and infertility. Therefore, an important element of the management of these patients is fertility preservation, which is challenged by considerations specific for leukemia. Such considerations include prepuberty, poor medical condition on presentation and the need for urgent chemotherapy, all of which may preclude the use of conventional assisted reproductive technologies. A restrictive approach towards utilizing cryopreserved ovarian tissue due to the risk for malignancy reintroduction on transplantation, further minimize the options that currently exist for these patients. This paper aims to provide up-to-date knowledge on gonadotoxicity associated with therapy regimens currently used in the treatment of leukemia. Different fertility preservation techniques are discussed, with an emphasis on efficacy, applicability and limitations in the context of leukemia.


Assuntos
Preservação da Fertilidade/métodos , Leucemia/tratamento farmacológico , Ovário/transplante , Antineoplásicos/administração & dosagem , Antineoplásicos/efeitos adversos , Criopreservação/métodos , Feminino , Humanos , Infertilidade Feminina/etiologia , Ovário/patologia , Insuficiência Ovariana Primária/etiologia , Técnicas de Reprodução Assistida
12.
J Assist Reprod Genet ; 35(5): 851-856, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29574609

RESUMO

PURPOSE: Few clinical options for fertility preservation are available to females with cancer, and data about clinical outcomes is limited. Potential supplementary approaches to fertility preservation include retrieval of immature oocytes followed by in vitro maturation (IVM) and storage. The aim of this study was to evaluate post-thawing outcomes of immature oocytes collected both by transvaginal aspiration and from excised ovarian tissue. METHODS: We conducted a retrospective cohort study of patients treated in a single tertiary center. We reviewed the records of 119 cancer patients who underwent ovarian tissue cryopreservation and immature oocyte harvesting for fertility preservation. All embryos and oocytes that were frozen and thawed were included in the study. Post-thawing outcomes were evaluated. RESULTS: Thirty-five stored embryos from eight patients were thawed. Twenty-nine embryos survived (82% survival rate) and were transferred. Six oocytes were thawed, two oocytes survived, and no oocytes were fertilized. Only one PCOS patient became pregnant, resulting in the normal delivery of a healthy baby. CONCLUSIONS: Although a relatively high number of mature oocytes and embryos can be stored with the combined procedure, the limited rate of pregnancies represents a poor reproductive outcome. Therefore, this approach should be reserved for special groups with limited options.


Assuntos
Preservação da Fertilidade/métodos , Neoplasias , Recuperação de Oócitos/métodos , Ovário/citologia , Adolescente , Adulto , Criança , Criopreservação , Transferência Embrionária/métodos , Feminino , Humanos , Técnicas de Maturação in Vitro de Oócitos/métodos , Ovário/fisiologia , Gravidez , Estudos Retrospectivos , Coleta de Tecidos e Órgãos , Resultado do Tratamento
13.
Fertil Steril ; 109(1): 48-53, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29198847

RESUMO

OBJECTIVE: To describe a successful autologous ovarian tissue re-transplantation in a sterile leukemia survivor after evaluation for minimal residual disease and provide a review of the current literature. DESIGN: Presentation of a carefully designed workup taken to evaluate tissue for minimal residual disease, its limitations, and applicability to other patients. To date, there have not been any publications of auto-transplantations in leukemia survivors, owing to an estimated high risk for malignancy induction. SETTING: Large tertiary hospital. PATIENT(S): A 19-year-old acute myeloid leukemia patient underwent ovarian tissue cryopreservation during complete remission before bone marrow transplantation. After prolonged amenorrhea, the patient desired pregnancy. Laboratory tests showed antimüllerian hormone <0.1 ng/mL and FSH 116 mIU/mL. Ultrasound revealed no ovarian follicles. INTERVENTION(S): Ovarian tissue cryopreservation and auto-transplantation. Histology, immunohistochemistry, FISH, next-generation sequencing, and xenotransplantation were done to evaluate thawed tissue samples for the presence of leukemia cells. MAIN OUTCOME MEASURE(S): Evidence for leukemia cells in thawed ovarian tissue, reproductive outcomes and live birth after transplantation, and leukemia-free survival. RESULT(S): Histology was negative for leukemia cells. Three severe combined immunodeficiency mice, grafted with tissue fragments, were followed for 6 months and showed no macroscopic/microscopic signs for leukemia. Fluorescence in situ hybridization for disease-specific gene rearrangement resulted in a read below the probe's cut-off. A next-generation sequencing panel of genes implicated in myeloproliferative disorders did not reveal any significant molecular event. Transplantation was performed, followed by ovarian stimulation and IVF, resulting in the delivery of healthy newborn. More than 2 years have elapsed since transplantation, and the patient is leukemia free. CONCLUSION(S): Harvesting during complete remission, combined with intense tissue evaluation before transplantation, allowed a safe, successful transplantation in an acute myeloid leukemia survivor.


Assuntos
Transplante de Medula Óssea/efeitos adversos , Sobreviventes de Câncer , Criopreservação , Preservação da Fertilidade/métodos , Infertilidade Feminina/terapia , Leucemia Mieloide Aguda/cirurgia , Ovário/transplante , Adulto , Animais , Biomarcadores Tumorais/genética , Biópsia , Feminino , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Hibridização in Situ Fluorescente , Infertilidade Feminina/diagnóstico , Infertilidade Feminina/etiologia , Infertilidade Feminina/fisiopatologia , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patologia , Nascido Vivo , Camundongos SCID , Transplante de Neoplasias , Ovário/patologia , Gravidez , Transplante Autólogo , Resultado do Tratamento , Adulto Jovem
14.
J Assist Reprod Genet ; 35(1): 49-59, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29124460

RESUMO

PURPOSE: This binational qualitative study of medical egg freezing (MEF) examined women's motivations and experiences, including their perceived needs for patient-centered care in the midst of fertility- and life-threatening diagnoses. METHODS: Forty-five women who had undertaken MEF were interviewed in the USA (33 women) and in Israel (12 women) between June 2014 and August 2016. Interviews lasted approximately 1 h and were conducted by two senior medical anthropologists, one in each country. Women were recruited from four American IVF clinics (two academic, two private) and two Israeli clinics (both academic) where MEF is being offered to cancer patients and women with other fertility-threatening medical conditions. RESULTS: Women who undertake MEF view their fertility and future motherhood as important components of their identities and recovery and, thus, are grateful for the opportunity to pursue fertility preservation. However, women who undergo MEF have special needs, given that they tend to be a "vulnerable" population of young (age < 30), unmarried, resource-constrained women, who are facing not only fertility loss but also the "double jeopardy" of cancer. Through in-depth, qualitative interviews, these women's MEF stories reveal 10 dimensions of care important to fertility preservation, including five "system factors" (information, coordination and integration, accessibility, physical comfort, cost) and five "human factors" (adolescent issues, male partner involvement, family involvement, egg disposition decisions, emotional support). Together, these dimensions of care constitute an important framework that can be best described as "patient-centered MEF." CONCLUSIONS: Women pursuing MEF have special medical needs and concerns, which require particular forms of patient-centered care. This study outlines 10 dimensions of patient-centered fertility preservation that are appropriate for MEF patients. This approach may help IVF clinics to be better prepared for delivering top-quality care to mostly young, single women facing the daunting prospect of fertility loss and life-threatening medical diagnoses.


Assuntos
Preservação da Fertilidade , Congelamento , Oócitos , Assistência Centrada no Paciente/métodos , Adolescente , Adulto , Atitude Frente a Saúde , Criopreservação , Feminino , Preservação da Fertilidade/psicologia , Preservação da Fertilidade/estatística & dados numéricos , Necessidades e Demandas de Serviços de Saúde , Humanos , Entrevistas como Assunto , Assistência Centrada no Paciente/normas , Adulto Jovem
15.
Breast ; 35: 203-217, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28822332

RESUMO

The 3rd International Consensus Conference for Breast Cancer in Young Women (BCY3) took place in November 2016, in Lugano, Switzerland organized by the European School of Oncology (ESO) and the European Society of Medical Oncologists (ESMO). Consensus recommendations for the management of breast cancer in young women were updated from BCY2 with incorporation of new evidence to inform the guidelines, and areas of research priorities were identified. This manuscript summarizes the ESO-ESMO international consensus recommendations, which are also endorsed by the European Society of Breast Specialists (EUSOMA).


Assuntos
Neoplasias da Mama/terapia , Consenso , Oncologia/normas , Guias de Prática Clínica como Assunto/normas , Adulto , Neoplasias da Mama/prevenção & controle , Gerenciamento Clínico , Feminino , Humanos , Sociedades Médicas/normas , Suíça , Adulto Jovem
16.
Fertil Steril ; 106(2): 467-74, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27181924

RESUMO

OBJECTIVE: To report the single-center results of orthotopic retransplantations of cryopreserved ovarian tissue in cancer survivors and evaluate the validity of commonly accepted procedure limitations. DESIGN: Prospective cohort study. SETTING: Tertiary university-affiliated assisted reproduction technology (ART) and oncology centers. PATIENT(S): Twenty cancer survivors who underwent ovarian transplantation of frozen-thawed ovarian tissue with the aim to conceive. INTERVENTION(S): Ovarian tissue cryopreservation (OTCP) and transplantation, endocrine monitoring, in vitro fertilization (IVF). MAIN OUTCOME MEASURE(S): Endocrine profile, IVF, pregnancies, live births. RESULT(S): The patient ages at tissue harvesting ranged from 14 to 39 years. Fifteen women had hematologic malignancies, and two had leukemia (chronic myelogenous leukemia and acute myelogenous leukemia). Ten patients were exposed to nonsterilizing chemotherapy before OTCP. After transplantation, the endocrine recovery rate was 93%. Fourteen patients underwent IVF treatments with a fertilization rate of 58%. Sixteen pregnancies were achieved (10 after IVF, 6 spontaneous), resulting in 10 live births, two (twins) after harvesting from the mother at the age of 37. Two pregnancies are currently ongoing. After transplantation, 53% of patients conceived, and 32% delivered at least once. One patient conceived four times. Preharversting chemotherapy exposure was not associated with inferior outcomes. All patients, including two leukemia survivors, remained cancer free. CONCLUSION(S): Orthotopic transplantation of thawed ovarian tissue is a highly effective measure to restore fertility in sterilized cancer patients. Chemotherapy exposure before harvesting and age >35 is a realistic option in selected patients. Retransplantation in leukemic patients is possible after application of maximal safety measures. These results have led the national ethical and professional authorities to decide for the first time not to consider OTCP as an experimental modality for fertility preservation. CLINICAL TRIAL REGISTRATION NUMBER: NCT02659592.


Assuntos
Antineoplásicos/efeitos adversos , Criopreservação , Preservação da Fertilidade/métodos , Infertilidade Feminina/terapia , Neoplasias/tratamento farmacológico , Ovário/transplante , Reimplante , Sobreviventes , Centros Médicos Acadêmicos , Adolescente , Adulto , Feminino , Preservação da Fertilidade/efeitos adversos , Fertilização in vitro , Humanos , Infertilidade Feminina/diagnóstico , Infertilidade Feminina/etiologia , Infertilidade Feminina/fisiopatologia , Israel , Nascido Vivo , Idade Materna , Paridade , Gravidez , Taxa de Gravidez , Estudos Prospectivos , Técnicas de Reprodução Assistida/efeitos adversos , Fatores de Risco , Fatores de Tempo , Resultado do Tratamento , Adulto Jovem
17.
Fertil Steril ; 105(1): 20-9, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26677788

RESUMO

Recent advances in our understanding of the mechanisms underlying the impact of cytotoxic drugs on the ovary have opened up new directions for the protection of the ovary from chemotherapy-induced damage. These advances have spurred the investigation of pharmacological agents to prevent ovarian damage at the time of treatment. Prevention of ovarian damage and follicle loss would provide significant advantages over existing fertility preservation techniques. This manuscript reviews new methods for the prevention of chemotherapy-induced ovarian damage, including agents that act on the PI3K/PTEN/Akt follicle activation pathway, apoptotic pathways, the vascular system, and other potential methods of reducing chemotherapy-induced ovotoxicity.


Assuntos
Antineoplásicos/efeitos adversos , Fertilidade/efeitos dos fármacos , Infertilidade Feminina/prevenção & controle , Ovário/efeitos dos fármacos , Substâncias Protetoras/farmacologia , Animais , Antineoplásicos/metabolismo , Apoptose/efeitos dos fármacos , Citoproteção , Desenho de Fármacos , Feminino , Humanos , Infertilidade Feminina/induzido quimicamente , Infertilidade Feminina/metabolismo , Infertilidade Feminina/patologia , Infertilidade Feminina/fisiopatologia , Terapia de Alvo Molecular , Folículo Ovariano/efeitos dos fármacos , Folículo Ovariano/metabolismo , Folículo Ovariano/patologia , Ovário/metabolismo , Ovário/patologia , Ovário/fisiopatologia , Transdução de Sinais/efeitos dos fármacos
18.
Fertil Steril ; 104(5): 1162-7, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26335130

RESUMO

OBJECTIVE: To evaluate the association between carriage of BRCA1/2 mutations and ovarian performance, as demonstrated by in vitro fertilization (IVF) outcomes. DESIGN: Retrospective cohort study. SETTING: Two tertiary IVF centers. PATIENT(S): BRCA mutation carriers undergoing IVF for preimplantation genetic diagnosis (PGD) or fertility preservation were compared with non-BRCA PGD or fertility preservation patients, matched by age, IVF protocol, IVF center, and cancer disease status. INTERVENTION(S): In vitro fertilization cycles for PGD and fertility preservation. MAIN OUTCOME MEASURE(S): Outcome of IVF: oocyte yield, poor response rate, number of zygotes, pregnancy rates. RESULT(S): A total of 62 BRCA mutation carriers and 62 matched noncarriers were included; 42 were fertility preservation breast cancer patients, and 82 were PGD non-cancer patients. Mean (± SD) age of patients was 32 ± 3.58 years. Number of stimulation days and total stimulation dose were comparable between carriers and noncarriers. Their cycles resulted in comparable oocyte yield (13.75 vs. 14.75) and low response rates (8.06% vs. 6.45%). Number of zygotes, fertilization rates, and conception rates were also comparable. CONCLUSION(S): Both healthy and cancer-affected BRCA mutation carriers demonstrated normal ovarian response in IVF cycles.


Assuntos
Proteína BRCA1/genética , Proteína BRCA2/genética , Fertilização in vitro , Heterozigoto , Infertilidade Feminina/terapia , Mutação , Ovário/fisiopatologia , Adulto , Análise Mutacional de DNA , Transferência Embrionária , Feminino , Preservação da Fertilidade , Triagem de Portadores Genéticos , Predisposição Genética para Doença , Humanos , Infertilidade Feminina/diagnóstico , Infertilidade Feminina/genética , Infertilidade Feminina/fisiopatologia , Israel , Recuperação de Oócitos , Fenótipo , Valor Preditivo dos Testes , Gravidez , Taxa de Gravidez , Diagnóstico Pré-Implantação , Estudos Retrospectivos , Fatores de Risco , Centros de Atenção Terciária , Resultado do Tratamento
19.
Fertil Steril ; 103(5): 1305-12, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25792249

RESUMO

OBJECTIVE: To determine whether BRCA mutation carriers who undergo fertility treatments are at increased risk of developing invasive epithelial ovarian cancer (IEOC). DESIGN: Historical cohort study. SETTING: Tertiary university-affiliated medical center and the National Cancer Registry. PATIENT(S): A total of 1,073 Jewish Israeli BRCA mutation carriers diagnosed in a single institution between 1995 and 2013, including 164 carriers (15.2%) who had fertility treatments that included clomiphene citrate (n = 82), gonadotropin (n = 69), in vitro fertilization (IVF) (n = 66), or a combination (n = 50), and 909 carriers not treated for infertility. INTERVENTION(S): None. MAIN OUTCOME MEASURE(S): Odds ratios (OR) and 95% confidence intervals (CI) for IEOC association with fertility treatments and other hormone and reproductive variables. RESULT(S): In 175 (16.3%) mutation carriers, IEOC was diagnosed; 139 women carried BRCA1, 33 carried BRCA2, and 3 had unknown mutations. Fertility treatments were not associated with IEOC risk (age-adjusted OR 0.63; 95% CI, 0.38-1.05) regardless of treatment type (with clomiphene citrate, OR 0.87; 95% CI, 0.46-1.63; with gonadotropin, OR 0.59; 95% CI, 0.26-1.31; with IVF, OR 1.08, 95% CI, 0.57-2.06). Multivariate analysis indicated an increased risk of IEOC with hormone-replacement therapy (OR 2.22; 95% CI, 1.33-3.69) and a reduced risk with oral contraceptives (OR 0.19; 95% CI, 0.13-0.28) in both BRCA1 and BRCA2 mutation carriers. Parity was a risk factor for IEOC by univariate but not multivariate analysis. CONCLUSION(S): According to our results, treatments for infertile BRCA mutation carriers should not be contraindicated or viewed as risk modifiers for IEOC. Parity as a risk factor in BRCA mutation carriers warrants further investigation.


Assuntos
Proteína BRCA1/genética , Proteína BRCA2/genética , Heterozigoto , Infertilidade/terapia , Judeus/genética , Mutação , Neoplasias Epiteliais e Glandulares/genética , Neoplasias Ovarianas/genética , Técnicas de Reprodução Assistida/efeitos adversos , Centros Médicos Acadêmicos , Adulto , Idoso , Idoso de 80 Anos ou mais , Carcinoma Epitelial do Ovário , Feminino , Humanos , Infertilidade/fisiopatologia , Israel/epidemiologia , Pessoa de Meia-Idade , Análise Multivariada , Invasividade Neoplásica , Neoplasias Epiteliais e Glandulares/etnologia , Neoplasias Epiteliais e Glandulares/patologia , Razão de Chances , Neoplasias Ovarianas/etnologia , Neoplasias Ovarianas/patologia , Paridade , Fenótipo , Estudos Prospectivos , Sistema de Registros , Medição de Risco , Fatores de Risco , Centros de Atenção Terciária , Adulto Jovem
20.
Acta Haematol ; 132(3-4): 400-13, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25228566

RESUMO

Impaired reproductive function and possible infertility are major concerns in long-term survivors of hematological malignancies. The ongoing increase in the survival rates of these patients is therefore accompanied with a growing demand for effective, safe and specifically tailored fertility preservation options. When approaching patients facing hematological malignancy, an individual evaluation of potential infertility risks and possible preventive or preserving measures should be performed. This review aims to provide up-to-date knowledge on female reproductive risks, and ovarian, uterine and genital injuries associated with therapy regimens currently used in hemato-oncological disorders. Recent progress in fertility preservation methods including ovarian tissue cryopreservation and transplantation, egg and embryo freezing, ovarian transposition and their specific role in hematological disorders are presented. The efficacy of these methods, possible risks and future challenges are critically discussed.


Assuntos
Antineoplásicos/efeitos adversos , Preservação da Fertilidade , Raios gama/efeitos adversos , Insuficiência Ovariana Primária/etiologia , Antineoplásicos/uso terapêutico , Transplante de Medula Óssea/efeitos adversos , Feminino , Neoplasias Hematológicas/tratamento farmacológico , Neoplasias Hematológicas/patologia , Neoplasias Hematológicas/radioterapia , Humanos , Doenças Uterinas/etiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA