1
|
Houeis L, van der Plancke G, Wen JY, Dupuy L, Kara E, Cacciottola L, Maurel MC, Donnez J, Dolmans MM. Chemotherapy-induced diminished murine ovarian reserve model and impact of low-dose chemotherapy on fertility. F&S SCIENCE 2025; 6:177-185. [PMID: 39800215 DOI: 10.1016/j.xfss.2025.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 01/07/2025] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
OBJECTIVE To establish a murine model of chemotherapy-induced diminished ovarian reserve (DOR) and investigate residual fertility after chemotherapy exposure. DESIGN Two different chemotherapy protocols were tested to establish a valid DOR model by comparing follicle densities in mice given either protocol or physiological solution. An ovarian stimulation protocol was then selected from among different gonadotropins by counting the number of day 2 embryos obtained from normal mice. Finally, DOR mice were stimulated 5 and 8 weeks after chemotherapy with the chosen gonadotropin protocols, and day 2 embryos were recovered after mating, as was ovarian tissue for further immunohistologic analyses. SUBJECTS Seventy-two Naval Medical Research Institute mice. EXPOSURE Two different chemotherapy protocols. MAIN OUTCOME MEASURES This study compared day 2 embryo counts in both normal and chemotherapy-induced DOR mice. Ovarian histology and morphology were also investigated by follicle counting and classification, as was immunostaining for apoptosis (cleaved caspase-3), activation (phospho-Akt), and proliferation (Ki67). RESULTS A dose of 12 mg/kg of busulfan (Bu) + 120 mg/kg of cyclophosphamide (Cy) was chosen to establish the DOR model as it significantly reduced the ovarian reserve compared to both control mice (physiological solution) and the 1.2 mg/kg of Bu + 12 mg/kg of Cy protocol, without depleting it completely. When stimulated with 3.75 IU of Menopur, normal mice produced significantly more embryos than DOR mice given 12 mg/kg of Bu + 120 mg/kg of Cy (41.40 ± 14.74 vs. 23.67 ± 15.55 day 2 embryos). Although the follicle count was statistically diminished after single-dose chemotherapy administration, the remaining follicles did not display any difference in terms of apoptosis, activation, or proliferation rates. CONCLUSION We successfully established a chemotherapy-induced DOR model using 12 mg/kg of Bu + 120 mg/kg of Cy, as evidenced by lower, but not completely depleted, follicle numbers and fewer retrieved embryos. Histologic study of ovarian tissue exposed to DOR-inducing chemotherapy revealed that surviving follicles were of the similar quality as tissue not exposed to chemotherapy.
Collapse
Affiliation(s)
- Lara Houeis
- Gynecology Research Unit, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Graziella van der Plancke
- Gynecology Research Unit, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Jen-Yu Wen
- Gynecology Research Unit, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Laurence Dupuy
- IGYXOS Biotherapeutics, Centre INRAe Val de Loire, Nouzilly, France
| | - Elodie Kara
- IGYXOS Biotherapeutics, Centre INRAe Val de Loire, Nouzilly, France
| | - Luciana Cacciottola
- Gynecology Research Unit, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | | | - Jacques Donnez
- Society for Research Into Infertility, Brussels, Belgium
| | - Marie-Madeleine Dolmans
- Gynecology Research Unit, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium; Gynecology Department, Cliniques Universitaires Saint-Luc, Brussels, Belgium.
| |
Collapse
|
2
|
Ronchetti L, Terrenato I, Lobascio AM, Iacobelli M, Ferretti M, Setti Boubaker N, Sperati F, Mandoj C, Mancini E, Carosi M, Vizza E, Corrado G. Role of inflammatory blood parameters from complete blood count in predicting ovarian follicular density in cancer patients undergoing ovarian tissue cryopreservation. J Ovarian Res 2025; 18:89. [PMID: 40307845 PMCID: PMC12042306 DOI: 10.1186/s13048-025-01670-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 04/11/2025] [Indexed: 05/02/2025] Open
Abstract
BACKGROUND Inflammation is a key feature of neoplastic diseases, especially cancer. Predicting follicular density (FD) in ovarian cortical tissue is essential for evaluating ovarian tissue cryopreservation (OTC) outcomes in fertility preservation. However, to date, no studies have explored the role of inflammatory markers in predicting FD in OTC patients. This study aims to investigate the relationship between blood inflammatory parameters and FD in this population. METHODS We conducted a retrospective observational study on 101 OTC patients. The primary goal was to assess whether parameters from Complete Blood Count (CBC) that include White Blood Cells (WBC), absolute neutrophil count, absolute lymphocyte count, Neutrophil/Lymphocyte Ratio (NLR), Mean Platelet Volume (MPV), Platelet Count (PC), MPV/PC and the Platelet/Lymphocyte Ratio (PLR) could predict FD. We also evaluated the impact of factors such as oncological diagnosis, smoking, Body Mass Index (BMI), and germline BRCA mutations. Spearman's correlation coefficient and the Mann-Whitney test were used for analysis. RESULTS Significant correlations were found in patients aged between 27 and 31. In this group, NLR was inversely correlated with FD (Rho = -0.374, p = 0.032), while lymphocyte count (Rho = 0.371, p = 0.034) and MPV/PC (Rho = 0.365, p = 0.037) were positively correlated with FD. An inverse correlation was also found between PLR and FD (Rho = -0.38, p = 0.028). CONCLUSIONS Our findings suggest that NLR, lymphocyte count, MPV/PC and PLR may be useful in predicting FD in a subgroup of OTC patients. Larger studies are needed to confirm these results.
Collapse
Affiliation(s)
- Livia Ronchetti
- Pathology Unit, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, Rome, 00144, Italy.
| | - Irene Terrenato
- Biostatistics & Bioinformatics, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Anna Maria Lobascio
- Gynecologic Oncology Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Marcello Iacobelli
- Gynecologic Oncology Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | | | | | - Francesca Sperati
- UOSD Clinical Trial Center and Biostatistics & Bioinformatics, San Gallicano Dermatological Institute IRCCS, Rome, Italy
| | - Chiara Mandoj
- Clinical Pathology Unit and Cancer Biobank, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Emanuela Mancini
- Gynecologic Oncology Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Mariantonia Carosi
- Pathology Unit, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, Rome, 00144, Italy
| | - Enrico Vizza
- Gynecologic Oncology Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Giacomo Corrado
- Department of Women and Child Health, Division of Gynecologic Oncology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.
| |
Collapse
|
3
|
Hatekar P, Tsiartas P, Gómez LDM, Mateoiu C, Sehic E, Hellström M, Patrizio P, Akouri R. Ovarian function and response to gonadotropins after prolonged perfusion of whole ewe ovaries in a bioreactor. J Assist Reprod Genet 2025:10.1007/s10815-025-03432-6. [PMID: 40009267 DOI: 10.1007/s10815-025-03432-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
PURPOSE Fertility preservation for pre-pubertal girls undergoing gonadotoxic cancer treatments and women with systemic cancers at high risk for ovarian metastasis remains limited. Current options, such as ovarian cortex transplantation, risk reintroducing malignant cells. This study presents a novel approach focusing on ex vivo folliculogenesis and mature oocyte retrieval for cryopreservation, mitigating this risk. METHODS This experimental study optimized an ex vivo ovarian perfusion system in sheep, refining gonadotropin stimulation to yield mature oocytes. Eleven ovaries were divided into two experimental subgroups: Group 1 (n = 5) and Group 2 (n = 6). Both groups were perfused in a bioreactor for 4 to 8 days under distinct perfusion protocols, differing in gonadotropin administration overnight-Group 1 did not receive overnight gonadotropin stimulation, whereas Group 2 received basal gonadotropin stimulation overnight. Assessments included follicular proliferation, oocyte maturity, apoptosis, ovarian function-related gene expression, and the levels of hormones, metabolites, and electrolytes in the culture medium, compared across subgroups. RESULTS The protocol without overnight ovarian stimulation yielded mature MII oocytes, despite fewer secondary follicles and overexpression of the pro-apoptotic BAX gene. Conversely, ovaries with overnight stimulation yielded mostly GV-MI oocytes and exhibited reduced secondary follicle proliferation and higher HIF1A expression. Hormone levels, metabolites, and electrolytes remained stable across groups and time. CONCLUSIONS This study is the first to report the successful harvesting of MII oocytes following extended ex vivo perfusion of intact ewe ovaries, highlighting the potential of the perfusion model to support advanced follicular development. Further investigations are warranted to elucidate underlying mechanisms and refine protocol efficiency.
Collapse
Affiliation(s)
- Prajakta Hatekar
- Laboratory for Transplantation and Regenerative Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Institute of Clinical Sciences, Department of Obstetrics and Gynecology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Panagiotis Tsiartas
- Karolinska Institute, Department of Clinical Science, Intervention and Technology: Karolinska Institutet, Institutionen För Klinisk Vetenskap, Intervention Och Teknik, Stockholm, Sweden
- Stockholm IVF-EUGIN, Stockholm, Sweden
| | - Lucía de Miguel Gómez
- Laboratory for Transplantation and Regenerative Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Institute of Clinical Sciences, Department of Obstetrics and Gynecology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Claudia Mateoiu
- Department of Pathology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Edina Sehic
- Laboratory for Transplantation and Regenerative Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Institute of Clinical Sciences, Department of Obstetrics and Gynecology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Mats Hellström
- Laboratory for Transplantation and Regenerative Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Institute of Clinical Sciences, Department of Obstetrics and Gynecology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Health Innovation Labs By Sahlgrenska Science Park, Gothenburg, Sweden
| | - Pasquale Patrizio
- Miller School of Medicine, Division of Reproductive Endocrinology & Infertility, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Miami, Miami, USA
| | - Randa Akouri
- Institute of Clinical Sciences, Department of Obstetrics and Gynecology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
- Department of Obstetrics and Gynecology, Sahlgrenska University Hospital, Gothenburg, Sweden.
| |
Collapse
|
4
|
Chung N, Yang C, Yang H, Shin J, Song CY, Min H, Kim JH, Lee K, Lee JR. Local delivery of platelet-derived factors mitigates ischemia and preserves ovarian function through angiogenic modulation: A personalized regenerative strategy for fertility preservation. Biomaterials 2025; 313:122768. [PMID: 39232332 DOI: 10.1016/j.biomaterials.2024.122768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/11/2024] [Accepted: 08/20/2024] [Indexed: 09/06/2024]
Abstract
As the most prominent and ideal modality in female fertility preservation, ovarian tissue cryopreservation, and transplantation often confront the challenge of ischemic damage and follicular loss from avascular transplantation. To surmount this impediment, we engineered a novel platelet-derived factors-encapsulated fibrin hydrogel (PFH), a paradigmatic biomaterial. PFH encapsulates autologous platelet-derived factors, utilizing the physiological blood coagulation cascade for precise local delivery of bioactive molecules. In our study, PFH markedly bolstered the success of avascular ovarian tissue transplantation. Notably, the quantity and quality of follicles were preserved with improved neovascularization, accompanied by decreased DNA damage, increased ovulation, and superior embryonic development rates under a Low-concentration Platelet-rich plasma-derived factors encapsulated fibrin hydrogel (L-PFH) regimen. At a stabilized point of tissue engraftment, gene expression analysis mirrored normal ovarian tissue profiles, underscoring the effectiveness of L-PFH in mitigating the initial ischemic insult. This autologous blood-derived biomaterial, inspired by nature, capitalizes on the blood coagulation cascade, and combines biodegradability, biocompatibility, safety, and cost-effectiveness. The adjustable properties of this biomaterial, even in injectable form, extend its potential applications into the broader realm of personalized regenerative medicine. PFH emerges as a promising strategy to counter ischemic damage in tissue transplantation, signifying a broader therapeutic prospect. (197 words).
Collapse
Affiliation(s)
- Nanum Chung
- Department of Translational Medicines, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea; Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, 13620, Republic of Korea
| | - Chungmo Yang
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, 13620, Republic of Korea; Program in Nanoscience and Technology, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, Republic of Korea
| | - Heeseon Yang
- Department of Translational Medicines, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea; Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, 13620, Republic of Korea
| | - Jungwoo Shin
- Department of Translational Medicines, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea; Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, 13620, Republic of Korea
| | - Chae Young Song
- Department of Translational Medicines, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea; Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, 13620, Republic of Korea
| | - Hyewon Min
- Department of Translational Medicines, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea; Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, 13620, Republic of Korea
| | - Ji Hyang Kim
- Department of Obstetrics and Gynecology, Fertility Center of CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, 13496, Republic of Korea.
| | - Kangwon Lee
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, Republic of Korea.
| | - Jung Ryeol Lee
- Department of Translational Medicines, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea; Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, 13620, Republic of Korea; Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
| |
Collapse
|
5
|
Dadashzadeh A, Moghassemi S, Amorim CA. Bioprinting of a Liposomal Oxygen-Releasing Scaffold for Ovary Tissue Engineering. Tissue Eng Part A 2025; 31:69-78. [PMID: 38534964 DOI: 10.1089/ten.tea.2024.0003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/13/2024] Open
Abstract
This study addresses a critical challenge in bioprinting for regenerative medicine, specifically the issue of hypoxia compromising cell viability in engineered tissues. To overcome this hurdle, a novel approach using a microfluidic bioprinter is used to create a two-layer structure resembling the human ovary. This structure incorporates a liposomal oxygen-releasing system to enhance cell viability. The bioprinting technique enables the simultaneous extrusion of two distinct bioinks, namely, bioink A (comprising alginate 1% and 5 mg/mL PEGylated fibrinogen in a 20:1 molar ratio) and bioink B (containing alginate 0.5%). In addition, liposomal catalase and hydrogen peroxide (H2O2) are synthesized and incorporated into bioinks A and B, respectively. The liposomes are prepared using thin film hydration with a monodisperse size (140-160 nm) and high encapsulation efficiency. To assess construct functionality, isolated human ovarian cells are added to bioink A. The bioprinted constructs, with or without liposomal oxygen-releasing systems, are cultured under hypoxic and normoxic conditions for 3 days. Live/Dead assay results demonstrate that liposomal oxygen-releasing systems effectively preserve cell viability in hypoxic conditions, resembling viability under normoxic conditions without liposomes. PrestoBlue assay reveals significantly higher mitochondrial activity in constructs with liposomal oxygen delivery systems under both hypoxic and normoxic conditions. The evaluation of apoptosis status through annexin V immunostaining shows that liposomal oxygen-releasing scaffolds successfully protect cells from hypoxic stress, exhibiting a proportion of apoptotic cells similar to normoxic conditions. In contrast, constructs lacking liposomes in hypoxic conditions exhibit a higher incidence of cells in early-stage apoptosis. In conclusion, the study demonstrates the promising potential of bioprinted oxygen-releasing liposomal scaffolds to protect ovarian stromal cells in hypoxic environments. These innovative scaffolds not only offer protection but also recapitulate the mechanical differences between the medulla and the cortex in the normal ovary structure. This opens new avenues for advanced ovary tissue engineering and transplantation strategies.
Collapse
Affiliation(s)
- Arezoo Dadashzadeh
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Saeid Moghassemi
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Christiani A Amorim
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
6
|
Houeis L, van der Plancke G, Poirot C, Cacciottola L, Camboni A, Brocheriou I, Donnez J, Dolmans MM. Low doses of alkylating agents do not harm human ovarian tissue destined for cryopreservation. Fertil Steril 2024:S0015-0282(24)02447-6. [PMID: 39701360 DOI: 10.1016/j.fertnstert.2024.12.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 12/12/2024] [Accepted: 12/12/2024] [Indexed: 12/21/2024]
Abstract
OBJECTIVE To investigate the impact of nongonadotoxic doses of alkylating agents on human ovarian cortex. DESIGN Retrospective study. SETTING Academic research center. PATIENTS Biopsies from 78 patients who had undergone ovarian tissue cryopreservation were retrieved and analyzed. Among them, 42 had previously been treated with chemotherapy (alkylating agents, dose <3,400 mg/m2), making up the chemotherapy group, whereas 36 had not been given any chemotherapy, constituting the control group. MEAN OUTCOME MEASURES Follicle count and classification, morphology study, immunostaining for apoptosis (cleaved caspase-3), immunostaining for activation (phospho-Akt), fibrosis (Masson's trichrome), and vascularization (von Willebrand factor and smooth muscle actin). RESULTS In the prepubertal group, 271 follicles/mm3 were detected in control patients and 501 follicles/mm3 in chemotherapy-exposed subjects. In the adult group, 4,916 follicles/mm3 were found in control patients and 6,570 follicles/mm3 in chemotherapy-exposed patients. No difference in follicle density was observed between the 2 groups in any age category. Neither did we encounter any significant difference in follicle viability according to chemotherapy exposure or age. Proportions of nongrowing follicles were >76% in all age groups, irrespective of chemotherapy exposure, and higher, although not significantly, in the chemotherapy group compared with the control group. There were significantly fewer secondary follicles in the adult chemotherapy group than in the adult control group. Concerning apoptosis, no significant difference was observed between control and chemotherapy subjects in any age groups. Numbers of activated follicles were systematically higher in all age categories in the chemotherapy group than the control group. Areas of atypical follicles were noted in 4 out of 14 prepubertal patients in the chemotherapy group. In these areas, follicle density was 84,570 ± 8,837 follicles/mm3 and all follicles appeared nonviable but showed no sign of apoptosis. CONCLUSION Low-dose chemotherapy had no major impact on ovarian tissue, suggesting that ovarian tissue exposed to some chemotherapy before cryopreservation is comparable with ovarian tissue free of any chemotherapy, as clinically demonstrated by high pregnancy rates after ovarian tissue transplantation in women exposed to chemotherapy. Previous chemotherapy should therefore no longer be a contraindication to ovarian tissue cryopreservation.
Collapse
Affiliation(s)
- Lara Houeis
- Gynecology Research Unit, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Graziella van der Plancke
- Gynecology Research Unit, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Catherine Poirot
- Department of Hematology, AYA Unit, Saint Louis Hospital, Paris, France; Médecine Sorbonne Université, Site Pitié Salpêtrière, Paris, France
| | - Luciana Cacciottola
- Gynecology Research Unit, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Alessandra Camboni
- Gynecology Research Unit, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium; Anatomopathology Department, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Isabelle Brocheriou
- Médecine Sorbonne Université, Site Pitié Salpêtrière, Paris, France; Department of Pathology, Pitié-Salpêtrière Hospital, APHP, Paris, France
| | - Jacques Donnez
- Society for Research into Infertility, Brussels, Belgium
| | - Marie-Madeleine Dolmans
- Gynecology Research Unit, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium; Gynecology Department, Cliniques Universitaires Saint-Luc, Brussels, Belgium.
| |
Collapse
|
7
|
Di Nisio V, Daponte N, Messini C, Anifandis G, Antonouli S. Oncofertility and Fertility Preservation for Women with Gynecological Malignancies: Where Do We Stand Today? Biomolecules 2024; 14:943. [PMID: 39199331 PMCID: PMC11353009 DOI: 10.3390/biom14080943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 07/19/2024] [Accepted: 08/01/2024] [Indexed: 09/01/2024] Open
Abstract
Oncofertility is a growing medical and research field that includes two main areas: oncology and reproductive medicine. Nowadays, the percentage of patients surviving cancer has exponentially increased, leading to the need for intervention for fertility preservation in both men and women. Specifically, gynecological malignancies in women pose an additional layer of complexity due to the reproductive organs being affected. In the present review, we report fertility preservation options with a cancer- and stage-specific focus. We explore the drawbacks and the necessity for planning fertility preservation applications during emergency statuses (i.e., the COVID-19 pandemic) and comment on the importance of repro-counseling for multifaceted patients during their oncological and reproductive journey.
Collapse
Affiliation(s)
- Valentina Di Nisio
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Huddinge, 14186 Stockholm, Sweden;
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Huddinge, 14186 Stockholm, Sweden
| | - Nikoletta Daponte
- Department of Obstetrics and Gynaecology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41500 Larisa, Greece; (N.D.); (C.M.); (G.A.)
| | - Christina Messini
- Department of Obstetrics and Gynaecology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41500 Larisa, Greece; (N.D.); (C.M.); (G.A.)
| | - George Anifandis
- Department of Obstetrics and Gynaecology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41500 Larisa, Greece; (N.D.); (C.M.); (G.A.)
| | - Sevastiani Antonouli
- Department of Obstetrics and Gynaecology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41500 Larisa, Greece; (N.D.); (C.M.); (G.A.)
| |
Collapse
|
8
|
Zaniker EJ, Hashim PH, Gauthier S, Ankrum JA, Campo H, Duncan FE. Three-Dimensionally Printed Agarose Micromold Supports Scaffold-Free Mouse Ex Vivo Follicle Growth, Ovulation, and Luteinization. Bioengineering (Basel) 2024; 11:719. [PMID: 39061801 PMCID: PMC11274170 DOI: 10.3390/bioengineering11070719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Ex vivo follicle growth is an essential tool, enabling interrogation of folliculogenesis, ovulation, and luteinization. Though significant advancements have been made, existing follicle culture strategies can be technically challenging and laborious. In this study, we advanced the field through development of a custom agarose micromold, which enables scaffold-free follicle culture. We established an accessible and economical manufacturing method using 3D printing and silicone molding that generates biocompatible hydrogel molds without the risk of cytotoxicity from leachates. Each mold supports simultaneous culture of multiple multilayer secondary follicles in a single focal plane, allowing for constant timelapse monitoring and automated analysis. Mouse follicles cultured using this novel system exhibit significantly improved growth and ovulation outcomes with comparable survival, oocyte maturation, and hormone production profiles as established three-dimensional encapsulated in vitro follicle growth (eIVFG) systems. Additionally, follicles recapitulated aspects of in vivo ovulation physiology with respect to their architecture and spatial polarization, which has not been observed in eIVFG systems. This system offers simplicity, scalability, integration with morphokinetic analyses of follicle growth and ovulation, and compatibility with existing microphysiological platforms. This culture strategy has implications for fundamental follicle biology, fertility preservation strategies, reproductive toxicology, and contraceptive drug discovery.
Collapse
Affiliation(s)
- Emily J. Zaniker
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (E.J.Z.); (P.H.H.); (S.G.)
| | - Prianka H. Hashim
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (E.J.Z.); (P.H.H.); (S.G.)
| | - Samuel Gauthier
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (E.J.Z.); (P.H.H.); (S.G.)
| | - James A. Ankrum
- Roy J. Carver Department of Biomedical Engineering, Pappajohn Biomedical Institute, University of Iowa, Iowa City, IA 52245, USA;
| | - Hannes Campo
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (E.J.Z.); (P.H.H.); (S.G.)
| | - Francesca E. Duncan
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (E.J.Z.); (P.H.H.); (S.G.)
| |
Collapse
|
9
|
Rich BS, McCracken K, Nagel C, Allen L, Aldrink JH. The Shared Ovary: A Multidisciplinary Discussion With Pediatric and Adolescent Gynecology. J Pediatr Surg 2024; 59:1349-1354. [PMID: 38614951 DOI: 10.1016/j.jpedsurg.2024.03.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 03/04/2024] [Indexed: 04/15/2024]
Abstract
Pediatric and adolescent ovarian lesions are common and are frequently managed by both pediatric surgeons and pediatric and adolescent gynecologists. During the 2023 American Academy of Pediatric Section on Surgery meeting, an educational symposium was delivered focusing on various aspects of management of pediatric and adolescent benign and malignant masses, borderline lesions, and fertility options for children and adolescents undergoing cancer therapies. This article highlights the discussion during this symposium.
Collapse
Affiliation(s)
- Barrie S Rich
- Division of Pediatric Surgery, Northwell Health, Cohen Children's Medical Center, New Hyde Park, NY, USA
| | - Kate McCracken
- Section of Pediatric and Adolescent Gynecology, Division of Gynecology, Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
| | - Christa Nagel
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, The Ohio State University College of Medicine, The James Cancer Hospital and Solove Research Institute, Columbus, OH, USA
| | - Lisa Allen
- Division of Pediatric Gynecology, Department of Obstetrics and Gynecology, Sick Kids Hospital, University of Toronto, Toronto, Canada
| | - Jennifer H Aldrink
- Division of Pediatric Surgery, Department of Surgery, Nationwide Children's Hospital, The Ohio State University College of Medicine, Columbus, OH, USA.
| |
Collapse
|
10
|
Zaninović L, Bašković M, Ježek D, Habek D, Pogorelić Z, Katušić Bojanac A, Elveđi Gašparović V, Škrgatić L. Enhancement of Vascularization and Ovarian Follicle Survival Using Stem Cells in Cryopreserved Ovarian Tissue Transplantation-A Systematic Review. BIOLOGY 2024; 13:342. [PMID: 38785824 PMCID: PMC11117700 DOI: 10.3390/biology13050342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/12/2024] [Accepted: 05/13/2024] [Indexed: 05/25/2024]
Abstract
The increase in cancer survival rates has put a focus on ensuring fertility preservation procedures for cancer patients. Ovarian tissue cryopreservation presents the only option for prepubertal girls and patients who require immediate start of treatment and, therefore, cannot undergo controlled ovarian stimulation. We aimed to provide an assessment of stem cells' impact on cryopreserved ovarian tissue grafts in regard to the expression of growth factors, angiogenesis promotion, tissue oxygenation, ovarian follicle survival and restoration of endocrine function. For this systematic review, we searched the Scopus and PubMed databases and included reports of trials using murine and/or human cryopreserved ovarian tissue for transplantation or in vitro culture in combination with mesenchymal stem cell administration to the grafting site. Of the 1201 articles identified, 10 met the criteria. The application of stem cells to the grafting site has been proven to support vascular promotion and thereby shorten the period of tissue hypoxia, which is reflected in the increased number of remaining viable follicles and faster recovery of ovarian endocrine function. Further research is needed before implementing the use of stem cells in OT cryopreservation and transplantation procedures in clinical practice. Complex ethical dilemmas make this process more difficult.
Collapse
Affiliation(s)
- Luca Zaninović
- Scientific Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, Šalata 3, 10 000 Zagreb, Croatia
- Department of Obstetrics and Gynecology, University Hospital Centre Zagreb, Petrova ulica 13, 10 000 Zagreb, Croatia
- School of Medicine, University of Zagreb, Šalata 3, 10 000 Zagreb, Croatia
| | - Marko Bašković
- Scientific Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, Šalata 3, 10 000 Zagreb, Croatia
- School of Medicine, University of Zagreb, Šalata 3, 10 000 Zagreb, Croatia
- Department of Pediatric Surgery, Children’s Hospital Zagreb, Ulica Vjekoslava Klaića 16, 10 000 Zagreb, Croatia
- Croatian Academy of Medical Sciences, Kaptol 15, 10 000 Zagreb, Croatia
| | - Davor Ježek
- Scientific Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, Šalata 3, 10 000 Zagreb, Croatia
- School of Medicine, University of Zagreb, Šalata 3, 10 000 Zagreb, Croatia
- Department of Histology and Embryology, School of Medicine, University of Zagreb, Šalata 3, 10 000 Zagreb, Croatia
- Department of Transfusion Medicine and Transplantation Biology, University Hospital Centre Zagreb, Kišpatićeva ulica 12, 10 000 Zagreb, Croatia
| | - Dubravko Habek
- School of Medicine, University of Zagreb, Šalata 3, 10 000 Zagreb, Croatia
- Croatian Academy of Medical Sciences, Kaptol 15, 10 000 Zagreb, Croatia
- Department of Obstetrics and Gynecology, Clinical Hospital Merkur, Zajčeva ulica 19, 10 000 Zagreb, Croatia
- School of Medicine, Catholic University of Croatia, Ilica 242, 10 000 Zagreb, Croatia
| | - Zenon Pogorelić
- Department of Pediatric Surgery, University Hospital of Split, Spinčićeva ulica 1, 21 000 Split, Croatia;
- School of Medicine, University of Split, Šoltanska ulica 2a, 21 000 Split, Croatia
| | - Ana Katušić Bojanac
- Scientific Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, Šalata 3, 10 000 Zagreb, Croatia
- School of Medicine, University of Zagreb, Šalata 3, 10 000 Zagreb, Croatia
- Department of Medical Biology, School of Medicine, University of Zagreb, Šalata 3, 10 000 Zagreb, Croatia
| | - Vesna Elveđi Gašparović
- Department of Obstetrics and Gynecology, University Hospital Centre Zagreb, Petrova ulica 13, 10 000 Zagreb, Croatia
- School of Medicine, University of Zagreb, Šalata 3, 10 000 Zagreb, Croatia
| | - Lana Škrgatić
- Scientific Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, Šalata 3, 10 000 Zagreb, Croatia
- Department of Obstetrics and Gynecology, University Hospital Centre Zagreb, Petrova ulica 13, 10 000 Zagreb, Croatia
- School of Medicine, University of Zagreb, Šalata 3, 10 000 Zagreb, Croatia
| |
Collapse
|
11
|
Silvestris E, Cormio G, Loizzi V, Corrado G, Arezzo F, Petracca EA. Fertility Preservation in BRCA1/2 Germline Mutation Carriers: An Overview. Life (Basel) 2024; 14:615. [PMID: 38792636 PMCID: PMC11122448 DOI: 10.3390/life14050615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 04/28/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
BRCA1 and BRCA2 mutations are responsible for a higher incidence of breast and ovarian cancer (from 55% up to 70% vs. 12% in the general population). If their functions have been widely investigated in the onset of these malignancies, still little is known about their role in fertility impairment. Cancer patients treated with antineoplastic drugs can be susceptible to their gonadotoxicity and, in women, some of them can induce apoptotic program in premature ovarian follicles, progressive depletion of ovarian reserve and, consequently, cancer treatment-related infertility (CTRI). BRCA variants seem to be associated with early infertility, thus accelerating treatment impairment of ovaries and making women face the concrete possibility of an early pregnancy. In this regard, fertility preservation (FP) procedures should be discussed in oncofertility counseling-from the first line of prevention with risk-reducing salpingo-oophorectomy (RRSO) to the new experimental ovarian stem cells (OSCs) model as a new way to obtain in vitro-differentiated oocytes, several techniques may represent a valid option to BRCA-mutated patients. In this review, we revisit knowledge about BRCA involvement in lower fertility, pregnancy feasibility, and the fertility preservation (FP) options available.
Collapse
Affiliation(s)
- Erica Silvestris
- Gynecologic Oncology Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy; (G.C.); (V.L.); (F.A.); (E.A.P.)
| | - Gennaro Cormio
- Gynecologic Oncology Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy; (G.C.); (V.L.); (F.A.); (E.A.P.)
- Department of Interdisciplinary Medicine (DIM), University of Bari “Aldo Moro”, 70121 Bari, Italy
| | - Vera Loizzi
- Gynecologic Oncology Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy; (G.C.); (V.L.); (F.A.); (E.A.P.)
- Department of Interdisciplinary Medicine (DIM), University of Bari “Aldo Moro”, 70121 Bari, Italy
| | - Giacomo Corrado
- Department of Woman, Child Health and Public Health, Gynecologic Oncology Unit, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00136 Rome, Italy;
| | - Francesca Arezzo
- Gynecologic Oncology Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy; (G.C.); (V.L.); (F.A.); (E.A.P.)
| | - Easter Anna Petracca
- Gynecologic Oncology Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy; (G.C.); (V.L.); (F.A.); (E.A.P.)
| |
Collapse
|
12
|
Keckstein P, Dittrich R, Bleisinger N, Hoffmann I, Beckmann MW, Gebhardt A, Schmid B, Keckstein S. Survival and hormone production of isolated mouse follicles in three-dimensional artificial scaffolds after stimulation with bpV(HOpic). Arch Gynecol Obstet 2024; 309:2127-2136. [PMID: 38472502 PMCID: PMC11018681 DOI: 10.1007/s00404-024-07419-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 02/05/2024] [Indexed: 03/14/2024]
Abstract
PURPOSE To preserve fertility before gonadotoxic therapy, ovarian tissue can be removed, cryopreserved, and transplanted back again after treatment. An alternative is the artificial ovary, in which the ovarian follicles are extracted from the tissue, which reduces the risk of reimplantation of potentially remaining malignant cells. The PTEN inhibitor bpV(HOpic) has been shown to activate human, bovine and alpacas ovarian follicles, and it is therefore considered a promising substance for developing the artificial ovary. The purpose of this study was to examine the impact of different scaffolds and the vanadate derivative bpV(HOpic) on mice follicle survival and hormone secretion over 10 days. METHODS A comparative analysis was performed, studying the survival rates (SR) of isolated mice follicle in four different groups that differed either in the scaffold (polycaprolactone scaffold versus polyethylene terephthalate membrane) or in the medium-bpV(HOpic) versus control medium. The observation period of the follicles was 10 days. On days 2, 6, and 10, the viability and morphology of the follicles were checked using fluorescence or confocal microscopy. Furthermore, hormone levels of estrogen (pmol/L) and progesterone (nmol/L) were determined. RESULTS When comparing the SR of follicles among the four groups, it was observed that on day 6, the study groups utilizing the polycaprolactone scaffold with bpV(HOpic) in the medium (SR: 0.48 ± 0.18; p = 0.004) or functionalized in the scaffold (SR: 0.50 ± 0.20; p = 0.003) exhibited significantly higher survival rates compared to the group using only the polyethylene terephthalate membrane (SR: 0). On day 10, a significantly higher survival rate was only noted when comparing the polycaprolactone scaffold with bpV(HOpic) in the medium to the polyethylene terephthalate membrane group (SR: 0.38 ± 0.20 versus 0; p = 0.007). Higher levels of progesterone were only significantly associated with better survival rates in the group with the polycaprolactone scaffold functionalized with bpV(HOpic) (p = 0.017). CONCLUSION This study demonstrates that three-dimensional polycaprolactone scaffolds improve the survival rates of isolated mice follicles in comparison with a conventional polyethylene terephthalate membrane. The survival rates slightly improve with added bpV(HOpic). Furthermore, higher rates of progesterone were also partly associated with improved survival.
Collapse
Affiliation(s)
- Philip Keckstein
- Department of Gynecology and Obstetrics, Erlangen University Hospital, Friedrich-Alexander-University of Erlangen-Nürnberg, Comprehensive Cancer Center ER-EMN, Erlangen, Germany.
| | - Ralf Dittrich
- Department of Gynecology and Obstetrics, Erlangen University Hospital, Friedrich-Alexander-University of Erlangen-Nürnberg, Comprehensive Cancer Center ER-EMN, Erlangen, Germany
| | - Nathalie Bleisinger
- Department of Gynecology and Obstetrics, Erlangen University Hospital, Friedrich-Alexander-University of Erlangen-Nürnberg, Comprehensive Cancer Center ER-EMN, Erlangen, Germany
| | - Inge Hoffmann
- Department of Gynecology and Obstetrics, Erlangen University Hospital, Friedrich-Alexander-University of Erlangen-Nürnberg, Comprehensive Cancer Center ER-EMN, Erlangen, Germany
| | - Matthias W Beckmann
- Department of Gynecology and Obstetrics, Erlangen University Hospital, Friedrich-Alexander-University of Erlangen-Nürnberg, Comprehensive Cancer Center ER-EMN, Erlangen, Germany
| | - Albrecht Gebhardt
- Department of Statistics, University of Klagenfurt, Klagenfurt, Austria
| | - Benjamin Schmid
- Optical Imaging Center Erlangen (OICE), University of Erlangen-Nürnberg, Erlangen, Germany
| | - Simon Keckstein
- Department of Obstetrics and Gynecology, LMU University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
13
|
Barrenetxea G, Celis R, Barrenetxea J, Martínez E, De Las Heras M, Gómez O, Aguirre O. Intraovarian platelet-rich plasma injection and IVF outcomes in patients with poor ovarian response: a double-blind randomized controlled trial. Hum Reprod 2024; 39:760-769. [PMID: 38423539 DOI: 10.1093/humrep/deae038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 02/03/2024] [Indexed: 03/02/2024] Open
Abstract
STUDY QUESTION Does platelet-rich plasma (PRP) intraovarian injection increase the number of retrieved oocytes in successive ovarian punctions among patients with poor ovarian reserve (POR)? SUMMARY ANSWER The injection of PRP increases the number of retrieved oocytes without increasing the quality of developed blastocysts. WHAT IS KNOWN ALREADY Management of women with reduced ovarian response to stimulation is one of the significant challenges in reproductive medicine. Recently, PRP treatment has been proposed as an adjunct in assisted reproduction technology, with controversial results. STUDY DESIGN, SIZE, DURATION This placebo-controlled, double-blind, randomized trial included 60 patients with POR stratified according to the POSEIDON classification groups 3 and 4. It was conducted to explore the efficacy and safety of intraovarian PRP injection. Patients were proposed to undergo three consecutive ovarian stimulations to accumulate oocytes and were randomized to receive either PRP or placebo during their first oocyte retrieval. Randomization was performed using computer-generated randomization codes. Double blinding was ensured so that neither the participant nor the investigators knew of the treatment allotted. All patients underwent three ovarian stimulations and egg retrieval procedures. ICSI was performed after a third ovarian puncture. The primary endpoint was the number of mature oocytes retrieved after PRP or placebo injection in successive ovarian punctures. PARTICIPANTS/MATERIALS, SETTING, METHODS Sixty women (30-42 years) fulfilling inclusion criteria were randomized in equal proportions to the treatment or control groups. MAIN RESULTS AND THE ROLE OF CHANCE The baseline demographic and clinical characteristics [age, BMI, anti-Müllerian hormone (AMH) levels] were comparable between the groups. Regarding the primary endpoint, the cumulative number (mean ± SEM) of retrieved mature oocytes was slightly higher in the treatment group: 10.45 ± 0.41 versus 8.91 ± 0.39 in the control group, respectively (95% CI of the difference 0.42-2.66; P = 0,008). The number of mature oocytes obtained among all patients increased in successive egg retrievals: 2.61 ± 0.33 (mean ± SEM) in punction 1 (P1), 3.85 ± 0.42 in P2, and 4.73 ± 0.44 in P3. However, the increase was higher among patients receiving the assessed PRP treatment. In P2, the number of retrieved mature oocytes was 4.18 ± 0.58 versus 3.27 ± 0.61 in controls (95% CI of the difference: -0.30 to 2.12; P = 0.138) and in P3, 5.27 ± 0.73 versus 4.15 ± 0.45 (95% CI of the difference: 0.12-2.12; P = 0.029). The mean ± SEM number of developed and biopsied blastocysts was 2.43 ± 0.60 in the control group and 1.90 ± 0.32 in the treatment group, respectively (P = 0.449). The mean number of euploid blastocysts was 0.81 ± 0.24 and 0.81 ± 0.25 in the control and treatment groups, respectively (P = 1.000). The percentages of patients with euploid blastocysts were 53.33% (16 out of 30) and 43.33% (13 out of 30) for patients in the control and treatment groups, respectively (Fisher's exact test P = 0.606). The overall pregnancy rate per ITT was 43% (26 out of 60 patients). However, the percentage of clinical pregnancies was higher in the control group (18 out of 30, 60%) than in the treatment group (8 out of 30, 27%) (P = 0.018). There was also a trend toward poorer outcomes in the treatment group when considering full-term pregnancies (P = 0.170). There were no differences between control and treatment groups regarding type of delivery, and sex of newborns. LIMITATIONS, REASONS FOR CAUTION The mechanism of the potential beneficial effect of PRP injection on the number of retrieved oocytes is unknown. Either delivered platelet factors or a mechanical effect could be implicated. Further studies will be needed to confirm or refute the data presented in this trial and to specify the exact mechanism of action, if any, of PRP preparations. WIDER IMPLICATIONS OF THE FINDINGS The increasing number of women with a poor response to ovarian stimulation supports the exploration of new areas of research to know the potential benefits of therapies capable of increasing the number of oocytes available for fertilization and improving the quality of developed blastocysts. An increase in the retrieved oocytes in both arms of the trial suggests that, beyond the release of growth factor from platelets, a mechanical effect can play a role. However, neither improvement in euploid blastocyst development nor pregnancy rates have been demonstrated. STUDY FUNDING/COMPETING INTEREST(S) This trial was supported by Basque Government and included in HAZITEK program, framed in the new Euskadi 2030 Science and Technology Plan (PCTI 2030). These aids are co-financed by the European Regional Development Fund (FEDER). The study funders had no role in the study design, implementation, analysis, manuscript preparation, or decision to submit this article for publication. No competing interests are declared by all the authors. TRIAL REGISTRATION NUMBER Clinical Trial Number EudraCT 2020-000247-32. TRIAL REGISTRATION DATE 3 November 2020. DATE OF FIRST PATIENT’S ENROLLMENT 16 January 2021.
Collapse
Affiliation(s)
- G Barrenetxea
- Reproducción Bilbao Assisted Reproduction Center, Bilbao, Spain
- Departamento de Especialidades Médico-Quirúrgicas, Universidad del País Vasco/Euskal Herriko Unibertsitatea (UPV/EHU), Leioa, Spain
| | - R Celis
- Reproducción Bilbao Assisted Reproduction Center, Bilbao, Spain
| | - J Barrenetxea
- Reproducción Bilbao Assisted Reproduction Center, Bilbao, Spain
- Osakidetza/Servicio Vasco de Salud, Hospital de Urduliz Alfredo Espinosa, Urduliz, Spain
| | - E Martínez
- Reproducción Bilbao Assisted Reproduction Center, Bilbao, Spain
| | - M De Las Heras
- Reproducción Bilbao Assisted Reproduction Center, Bilbao, Spain
| | - O Gómez
- Reproducción Bilbao Assisted Reproduction Center, Bilbao, Spain
| | - O Aguirre
- Reproducción Bilbao Assisted Reproduction Center, Bilbao, Spain
| |
Collapse
|
14
|
Kocsuta V, Shah S, Lawson AK, Pavone ME. Do Fertility Preservation Outcomes in Patients Diagnosed with Lymphoma Differ Based on Cancer Stage? J Adolesc Young Adult Oncol 2024; 13:300-306. [PMID: 36809174 DOI: 10.1089/jayao.2022.0069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023] Open
Abstract
Purpose: This study aims to investigate whether oocyte stimulation outcomes in fertility preservation (FP) vary in patients with different stages of lymphoma. Methods: This is a retrospective cohort study conducted at Northwestern Memorial Hospital (NMH). Between 2006 and 2017, 89 patients were identified with a diagnosis of lymphoma who contacted the FP navigator at NMH. Anti-müllerian hormone (AMH) levels and FP ovarian stimulation outcomes were collected for analysis. The data were analyzed using chi-squared and analysis of variance tests. A regression analysis was also done to adjust for potential confounding variables. Results: Of the 89 patients who contacted the FP navigator, there were 12 patients (13.5%) with stage 1 lymphoma, 43 patients (48.3%) with stage 2, 13 patients (14.6%) with stage 3, 13 patients (14.6%) with stage 4, and 8 patients (9.0%) where staging information was not available. Forty-five of the patients proceeded with ovarian stimulation before initiating cancer treatment. Patients who underwent ovarian stimulation had a mean AMH level of 2.62 and median peak estradiol levels of 1772.0 pg/mL. Median oocytes retrieved was 16.77, mature oocytes were 11.00 and median oocytes frozen after completing FP was 8.00. These measures were also stratified by stage of lymphoma. Conclusion: We found no significant difference in number of retrieved, mature or vitrified oocytes between different cancer stages. There was also no difference in AMH levels in the different cancer stage groups. This suggests that even in higher stages of lymphoma, many patients respond to ovarian stimulation techniques and have a successful stimulation cycle.
Collapse
Affiliation(s)
- Victoria Kocsuta
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Shriya Shah
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Angela K Lawson
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Mary Ellen Pavone
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
15
|
Wang XJ, Chen MX, Ruan LL, Tan L, Geng LH, Yang HJ, Fu LJ, Zhong ZH, Lv XY, Ding YB, Wan Q. Study on the optimal time limit of frozen embryo transfer and the effect of a long-term frozen embryo on pregnancy outcome. Medicine (Baltimore) 2024; 103:e37542. [PMID: 38552082 PMCID: PMC10977551 DOI: 10.1097/md.0000000000037542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 02/19/2024] [Indexed: 04/02/2024] Open
Abstract
In this retrospective study conducted at Sichuan Jinxin Xinan Women and Children's Hospital spanning January 2015 to December 2021, our objective was to investigate the impact of embryo cryopreservation duration on outcomes in frozen embryo transfer. Participants, totaling 47,006 cycles, were classified into 3 groups based on cryopreservation duration: ≤1 year (Group 1), 1 to 6 years (Group 2), and ≥6 years (Group 3). Employing various statistical analyses, including 1-way ANOVA, Kruskal-Wallis test, chi-square test, and a generalized estimating equation model, we rigorously adjusted for confounding factors. Primary outcomes encompassed clinical pregnancy rate and Live Birth Rate (LBR), while secondary outcomes included biochemical pregnancy rate, multiple pregnancy rate, ectopic pregnancy rate, early and late miscarriage rates, preterm birth rate, neonatal birth weight, weeks at birth, and newborn sex. Patient distribution across cryopreservation duration groups was as follows: Group 1 (40,461 cycles), Group 2 (6337 cycles), and Group 3 (208 cycles). Postcontrolling for confounding factors, Group 1 exhibited a decreased likelihood of achieving biochemical pregnancy rate, clinical pregnancy rate, and LBR (OR < 1, aOR < 1, P < .05). Furthermore, an elevated incidence of ectopic pregnancy was observed (OR > 1, aOR > 1), notably significant after 6 years of freezing time [aOR = 4.141, 95% confidence intervals (1.013-16.921), P = .05]. Cryopreservation exceeding 1 year was associated with an increased risk of early miscarriage and preterm birth (OR > 1, aOR > 1). No statistically significant differences were observed in birth weight or sex between groups. However, male infant birth rates were consistently higher than those of female infants across all groups. In conclusion, favorable pregnancy outcomes align with embryo cryopreservation durations within 1 year, while freezing for more than 1 year may diminish clinical pregnancy and LBRs, concurrently elevating the risk of ectopic pregnancy and preterm birth.
Collapse
Affiliation(s)
- Xue-Jiao Wang
- Sichuan Jinxin Xinan Women and Children’s Hospital, Chengdu, China
| | - Ming-Xing Chen
- Department of Obstetrics and Gynecology, Women and Children’s Hospital of Chongqing Medical University, Chongqing, China
- Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing, China
| | - Ling-Ling Ruan
- Department of Obstetrics and Gynecology, Women and Children’s Hospital of Chongqing Medical University, Chongqing, China
- Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing, China
| | - Li Tan
- Sichuan Jinxin Xinan Women and Children’s Hospital, Chengdu, China
| | - Li-Hong Geng
- Sichuan Jinxin Xinan Women and Children’s Hospital, Chengdu, China
| | - Hui-Jun Yang
- Assisted Reproductive Center, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, China
| | - Li-Juan Fu
- Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing, China
- Department of Pharmacology, Academician Workstation, Changsha Medical University, Changsha, China
| | - Zhao-Hui Zhong
- Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing, China
| | - Xing-Yu Lv
- Sichuan Jinxin Xinan Women and Children’s Hospital, Chengdu, China
| | - Yu-Bin Ding
- Department of Obstetrics and Gynecology, Women and Children’s Hospital of Chongqing Medical University, Chongqing, China
- Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing, China
| | - Qi Wan
- Department of Obstetrics and Gynecology, West China Second Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| |
Collapse
|
16
|
Finkelstein T, Zhang Y, Vollenhoven B, Rolnik DL, Horta F. Successful pregnancy rates amongst patients undergoing ovarian tissue cryopreservation for non-malignant indications: A systematic review and meta-analysis. Eur J Obstet Gynecol Reprod Biol 2024; 292:30-39. [PMID: 37952490 DOI: 10.1016/j.ejogrb.2023.11.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 10/15/2023] [Accepted: 11/04/2023] [Indexed: 11/14/2023]
Abstract
BACKGROUND Ovarian tissue cryopreservation (OTC) is a fertility preservation method that has been clinically applied for almost 30 years. Studies specifically evaluating patients presenting with non-malignant indications for OTC and their subsequent pregnancy rates are limited. OBJECTIVE To summarise the evidence on the rates of successful pregnancy amongst women who have undergone OTC for non-malignant indications. METHODS A systematic review with meta-analysis (PROSPERO registration CRD42022307925) was conducted to investigate the pregnancy outcomes of patients who have undergone ovarian tissue cryopreservation for non-malignant indications. Articles published in EMBASE and Ovid MEDLINE before October 2022 were screened for inclusion based on the following criteria: original human studies pertaining to OTC with a defined non-malignant cohort and pregnancy outcomes. The successful pregnancy rates were pooled with a random-effects model of double-arcsine transformed proportions. Sensitivity analysis involved pooling the results of studies with a low risk of bias after being assessed with NIH tools. RESULTS The database search retrieved 3,225 results, of which 16 were included in the meta-analysis. The pooled successful pregnancy rate was 23.52 % (16 studies, 95 % CI 6.48 to 44.79 %). When subgroup analysis of study types was performed, the successful pregnancy rate was higher amongst case series (47.02 %, 9 studies, 95 % CI 6.98 to 89.00 %) than cohort studies (14.64 %, 7 studies, 95 % CI 3.59 to 29.78 %). Sensitivity analysis limited to studies at low risk of bias revealed a similar pooled successful pregnancy rate of 23.35 % (12 studies, 95 % CI 2.50 to 51.96 %). CONCLUSIONS Approximately one quarter of women who underwent OTC for non-malignant indications had a successful pregnancy. These findings are clinically important for fertility preservation counselling by providing greater evidence for more informed care.
Collapse
Affiliation(s)
- T Finkelstein
- Department of Obstetrics and Gynaecology, Monash University, Clayton, Australia.
| | - Y Zhang
- Department of Obstetrics and Gynaecology, Monash University, Clayton, Australia
| | - B Vollenhoven
- Department of Obstetrics and Gynaecology, Monash University, Clayton, Australia; Women's and Newborn, Monash Health, Australia; Monash IVF Melbourne, Australia
| | - D L Rolnik
- Department of Obstetrics and Gynaecology, Monash University, Clayton, Australia; Women's and Newborn, Monash Health, Australia
| | - F Horta
- Department of Obstetrics and Gynaecology, Monash University, Clayton, Australia; Monash Data Future Institute, Monash University, Clayton, Australia; City Fertility, Australia
| |
Collapse
|
17
|
Zhang L, Chi M, Cheng Y, Chen Z, Cao Y, Zhao G. Static magnetic field assisted thawing improves cryopreservation of mouse whole ovaries. Bioeng Transl Med 2024; 9:e10613. [PMID: 38193129 PMCID: PMC10771557 DOI: 10.1002/btm2.10613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 09/19/2023] [Accepted: 10/01/2023] [Indexed: 01/10/2024] Open
Abstract
Ovarian tissue cryopreservation is considered to be the only means to preserve fertility for prepubertal girls and women whose cancer treatment cannot be postponed. However, ovarian tissues are inevitably damaged by oxidative stress during cryopreservation, which threatens follicle survival and development, and thus affects female fertility. Therefore, reducing tissue oxidative stress injury is one of the major challenges to achieving efficient cryopreservation of ovarian tissues, especially for whole ovaries. Here, we proposed a new method to improve the antioxidant capacity of whole ovaries during cryopreservation, static magnetic field assisted thawing. The results demonstrated that the antioxidant capacity of the ovarian tissue was significantly improved by static magnetic field treatment. In addition, ovarian tissue allograft transplantation was carried out, which successfully achieved vascular regeneration and maintained follicular development. The findings of this study not only provide a new reference for the preservation of female fertility, but also is a major step forward in the cryopreservation of tissues and organs. It will have good application prospects in the field of assisted reproduction and cryo-biomedicine.
Collapse
Affiliation(s)
- Liyuan Zhang
- School of Basic MedicineAnhui Medical UniversityHefeiChina
| | - Mengqiao Chi
- School of Basic MedicineAnhui Medical UniversityHefeiChina
| | - Yue Cheng
- School of Biomedical EngineeringAnhui Medical UniversityHefeiChina
| | - Zhongrong Chen
- School of Biomedical EngineeringAnhui Medical UniversityHefeiChina
| | - Yunxia Cao
- Department of Obstetrics and GynecologyReproductive Medicine Center, The First Affiliated Hospital of Anhui Medical UniversityHefeiChina
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University)HefeiChina
| | - Gang Zhao
- School of Basic MedicineAnhui Medical UniversityHefeiChina
- School of Biomedical EngineeringAnhui Medical UniversityHefeiChina
- Department of Electronic Engineering and Information ScienceUniversity of Science and Technology of ChinaHefeiChina
| |
Collapse
|
18
|
Zhang S, Yahaya BH, Pan Y, Liu Y, Lin J. Menstrual blood-derived endometrial stem cell, a unique and promising alternative in the stem cell-based therapy for chemotherapy-induced premature ovarian insufficiency. Stem Cell Res Ther 2023; 14:327. [PMID: 37957675 PMCID: PMC10644549 DOI: 10.1186/s13287-023-03551-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 10/27/2023] [Indexed: 11/15/2023] Open
Abstract
Chemotherapy can cause ovarian dysfunction and infertility since the ovary is extremely sensitive to chemotherapeutic drugs. Apart from the indispensable role of the ovary in the overall hormonal milieu, ovarian dysfunction also affects many other organ systems and functions including sexuality, bones, the cardiovascular system, and neurocognitive function. Although conventional hormone replacement therapy can partly relieve the adverse symptoms of premature ovarian insufficiency (POI), the treatment cannot fundamentally prevent deterioration of POI. Therefore, effective treatments to improve chemotherapy-induced POI are urgently needed, especially for patients desiring fertility preservation. Recently, mesenchymal stem cell (MSC)-based therapies have resulted in promising improvements in chemotherapy-induced ovary dysfunction by enhancing the anti-apoptotic capacity of ovarian cells, preventing ovarian follicular atresia, promoting angiogenesis and improving injured ovarian structure and the pregnancy rate. These improvements are mainly attributed to MSC-derived biological factors, functional RNAs, and even mitochondria, which are directly secreted or indirectly translocated with extracellular vesicles (microvesicles and exosomes) to repair ovarian dysfunction. Additionally, as a novel source of MSCs, menstrual blood-derived endometrial stem cells (MenSCs) have exhibited promising therapeutic effects in various diseases due to their comprehensive advantages, such as periodic and non-invasive sample collection, abundant sources, regular donation and autologous transplantation. Therefore, this review summarizes the efficacy of MSCs transplantation in improving chemotherapy-induced POI and analyzes the underlying mechanism, and further discusses the benefit and existing challenges in promoting the clinical application of MenSCs in chemotherapy-induced POI.
Collapse
Affiliation(s)
- Shenghui Zhang
- Stem Cell and Biotherapy Technology Research Center, Henan Joint International Research Laboratory of Stem Cell Medicine, Xinxiang Medical University, East of JinSui Road, Xinxiang, Henan, China
- Lung Stem Cell and Gene Therapy Group, Regenerative Medicine Cluster, Advanced Medical and Dental Institute (IPPT), Universiti Sains Malaysia, Penang, Malaysia
| | - Badrul Hisham Yahaya
- Lung Stem Cell and Gene Therapy Group, Regenerative Medicine Cluster, Advanced Medical and Dental Institute (IPPT), Universiti Sains Malaysia, Penang, Malaysia
| | - Ying Pan
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, , China
| | - Yanli Liu
- Stem Cell and Biotherapy Technology Research Center, Henan Joint International Research Laboratory of Stem Cell Medicine, Xinxiang Medical University, East of JinSui Road, Xinxiang, Henan, China.
| | - Juntang Lin
- Stem Cell and Biotherapy Technology Research Center, Henan Joint International Research Laboratory of Stem Cell Medicine, Xinxiang Medical University, East of JinSui Road, Xinxiang, Henan, China.
| |
Collapse
|
19
|
Antunes MB, Cardeal SP, Magalhães M, Vale-Fernandes E, Barreiro M, Sá R, Sousa M. Preservation of fertility in female patients with hematologic diseases. Blood Rev 2023; 62:101115. [PMID: 37562987 DOI: 10.1016/j.blre.2023.101115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 06/23/2023] [Accepted: 07/10/2023] [Indexed: 08/12/2023]
Abstract
Recent developments of assisted reproduction techniques turned possible to avoid the infertility consequences of oncologic treatments, but fertility preservation (FP) has been somewhat neglected in women with hematologic diseases undergoing gonadotoxic treatments. For these specific cases, the current options for FP include the cryopreservation of embryos, mature oocytes and ovarian tissue, and oocyte in-vitro maturation. We intend to make patients and clinicians aware of this important and relevant issue, and provide hematologists, assisted reproduction physicians and patients, with updated tools to guide decisions for FP. The physicians of the units responsible for female FP should always be available to decide on the best-individualized FP option in strict collaboration with hematologists. With a wide range of options for FP tailored to each case, a greater level of training and information is needed among clinicians, so that patients proposed to gonadotoxic treatments can be previously advised for FP techniques in hematological conditions. ABBREVIATED ABSTRACT: Recent developments of assisted reproduction techniques turned possible to preserve the fertility of women with hematologic diseases undergoing gonadotoxic treatments. Current options for fertility preservation in women with hematologic diseases are presented. It is imperative to offer fertility preservation to all women before starting any gonadotoxic treatment and in some cases after treatment. Fertility preservation methods enable to later achieve the desired pregnancy.
Collapse
Affiliation(s)
- Marika Bini Antunes
- Department of Clinical Hematology, Centro Hospitalar Universitário do Porto, Largo do Professor Abel Salazar, 4099-001 Porto, Portugal; UMIB-Unit for Multidisciplinary Research in Biomedicine/ITR-Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal.
| | - Sara Pinto Cardeal
- Laboratory of Cell Biology, Department of Microscopy, ICBAS-School of Medicine and Biomedical Sciences, University of Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| | - Manuel Magalhães
- UMIB-Unit for Multidisciplinary Research in Biomedicine/ITR-Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal; Department of Oncology, Centro Hospitalar Universitário do Porto, Largo do Professor Abel Salazar, 4099-001 Porto, Portugal
| | - Emídio Vale-Fernandes
- UMIB-Unit for Multidisciplinary Research in Biomedicine/ITR-Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal; Centro de Procriação Medicamente Assistida, Centro Materno-Infantil do Norte, Centro Hospitalar Universitário do Porto, Largo da Maternidade, 4050-371, Porto, Portugal
| | - Márcia Barreiro
- UMIB-Unit for Multidisciplinary Research in Biomedicine/ITR-Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal; Centro de Procriação Medicamente Assistida, Centro Materno-Infantil do Norte, Centro Hospitalar Universitário do Porto, Largo da Maternidade, 4050-371, Porto, Portugal.
| | - Rosália Sá
- UMIB-Unit for Multidisciplinary Research in Biomedicine/ITR-Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal; Laboratory of Cell Biology, Department of Microscopy, ICBAS-School of Medicine and Biomedical Sciences, University of Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| | - Mário Sousa
- UMIB-Unit for Multidisciplinary Research in Biomedicine/ITR-Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal; Laboratory of Cell Biology, Department of Microscopy, ICBAS-School of Medicine and Biomedical Sciences, University of Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| |
Collapse
|
20
|
Canosa S, Revelli A, Gennarelli G, Cormio G, Loizzi V, Arezzo F, Petracca EA, Carosso AR, Cimadomo D, Rienzi L, Vaiarelli A, Ubaldi FM, Silvestris E. Innovative Strategies for Fertility Preservation in Female Cancer Survivors: New Hope from Artificial Ovary Construction and Stem Cell-Derived Neo-Folliculogenesis. Healthcare (Basel) 2023; 11:2748. [PMID: 37893822 PMCID: PMC10606281 DOI: 10.3390/healthcare11202748] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/05/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
Recent advances in anticancer treatment have significantly improved the survival rate of young females; unfortunately, in about one third of cancer survivors the risk of ovarian insufficiency and infertility is still quite relevant. As the possibility of becoming a mother after recovery from a juvenile cancer is an important part of the quality of life, several procedures to preserve fertility have been developed: ovarian surgical transposition, induction of ovarian quiescence by gonadotropin-releasing hormone agonists (GnRH-a) treatment, and oocyte and/or ovarian cortical tissue cryopreservation. Ovarian tissue cryostorage and allografting is a valuable technique that applies even to prepubertal girls; however, some patients cannot benefit from it due to the high risk of reintroducing cancer cells during allograft in cases of ovary-metastasizing neoplasias, such as leukemias or NH lymphomas. Innovative techniques are now under investigation, as in the construction of an artificial ovary made of isolated follicles inserted into an artificial matrix scaffold, and the use of stem cells, including ovarian stem cells (OSCs), to obtain neo-folliculogenesis and the development of fertilizable oocytes from the exhausted ovarian tissue. This review synthesizes and discusses these innovative techniques, which potentially represent interesting strategies in oncofertility programs and a new hope for young female cancer survivors.
Collapse
Affiliation(s)
- Stefano Canosa
- IVIRMA, Global Research Alliance, LIVET, 10126 Turin, Italy; (A.R.); (G.G.)
| | - Alberto Revelli
- IVIRMA, Global Research Alliance, LIVET, 10126 Turin, Italy; (A.R.); (G.G.)
- Gynecology and Obstetrics 2U, Department of Surgical Sciences, S. Anna Hospital, University of Turin, 10126 Turin, Italy
| | - Gianluca Gennarelli
- IVIRMA, Global Research Alliance, LIVET, 10126 Turin, Italy; (A.R.); (G.G.)
- Gynecology and Obstetrics 1U, Physiopathology of Reproduction and IVF Unit, Department of Surgical Sciences, S. Anna Hospital, University of Turin, 10126 Turin, Italy;
| | - Gennaro Cormio
- Gynecologic Oncology Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy; (G.C.); (V.L.); (E.A.P.); (E.S.)
- Department of Interdisciplinary Medicine (DIM), University of Bari “Aldo Moro”, 70121 Bari, Italy
| | - Vera Loizzi
- Gynecologic Oncology Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy; (G.C.); (V.L.); (E.A.P.); (E.S.)
- Department of Interdisciplinary Medicine (DIM), University of Bari “Aldo Moro”, 70121 Bari, Italy
| | - Francesca Arezzo
- Obstetrics and Gynecology Unit, Department of Biomedical Sciences and Human Oncology, University of “Aldo Moro”, 70124 Bari, Italy
| | - Easter Anna Petracca
- Gynecologic Oncology Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy; (G.C.); (V.L.); (E.A.P.); (E.S.)
| | - Andrea Roberto Carosso
- Gynecology and Obstetrics 1U, Physiopathology of Reproduction and IVF Unit, Department of Surgical Sciences, S. Anna Hospital, University of Turin, 10126 Turin, Italy;
| | - Danilo Cimadomo
- IVIRMA, Global Research Alliance, GENERA, Clinica Valle Giulia, 00197 Rome, Italy; (D.C.); (L.R.); (A.V.); (F.M.U.)
| | - Laura Rienzi
- IVIRMA, Global Research Alliance, GENERA, Clinica Valle Giulia, 00197 Rome, Italy; (D.C.); (L.R.); (A.V.); (F.M.U.)
- Department of Biomolecular Sciences, University of Urbino “Carlo Bo”, 61029 Urbino, Italy
| | - Alberto Vaiarelli
- IVIRMA, Global Research Alliance, GENERA, Clinica Valle Giulia, 00197 Rome, Italy; (D.C.); (L.R.); (A.V.); (F.M.U.)
| | - Filippo Maria Ubaldi
- IVIRMA, Global Research Alliance, GENERA, Clinica Valle Giulia, 00197 Rome, Italy; (D.C.); (L.R.); (A.V.); (F.M.U.)
| | - Erica Silvestris
- Gynecologic Oncology Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy; (G.C.); (V.L.); (E.A.P.); (E.S.)
| |
Collapse
|
21
|
Rodrigues AQ, Silva IM, Goulart JT, Araújo LO, Ribeiro RB, Aguiar BA, Ferreira YB, Silva JKO, Bezerra JLS, Lucci CM, Paulini F. Effects of erythropoietin on ischaemia-reperfusion when administered before and after ovarian tissue transplantation in mice. Reprod Biomed Online 2023; 47:103234. [PMID: 37524029 DOI: 10.1016/j.rbmo.2023.05.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 05/04/2023] [Accepted: 05/10/2023] [Indexed: 08/02/2023]
Abstract
RESEARCH QUESTION Is the optimal timing for administering erythropoietin to minimize ischaemic injury in ovarian tissue transplantation before ovary removal for cryopreservation and subsequent transplantation or after transplantation? DESIGN Thirty Swiss mice (nu/nu) were divided into three groups: treatment control group (n = 10); erythropoietin before harvesting group (EPO-BH) (n = 10) and erythropoietin after transplantation group (EPO-AT) (n = 10). Animals underwent bilateral ovariohysterectomy and their hemiovaries were cryopreserved by slow freezing. At the same time, previously cryopreserved hemiovaries were transplanted subcutaneously in the dorsal region. Erythropoietin (250 IU/kg) and sterile 0.9% saline solution were administered every 12/12 h over 5 consecutive days in the EPO-AT and EPO-BH groups, respectively. RESULTS Administration of erythropoietin in the EPO-AT group improved the viability of ovarian follicles, reducing degeneration and increasing the number of morphologically normal growing follicles at 14 days after transplantation compared with the EPO-BH group (P = 0.002). This group also showed higher percentages of proliferative follicles at 7 days after transplantation (P ≤ 0.03), increased blood vessel count (P ≤ 0.03) and greater tissue area occupied by blood vessels at days 7 and 14 after transplantation (P ≤ 0.03), compared with hormone administration before cryopreservation (EPO-BH group) and the treatment control group. Additionally, treatment with erythropoietin before or after transplantation reduced fibrotic areas at 7 days after transplantation (P = 0.004). CONCLUSION Erythropoietin treatment after transplantation reduced ischaemic damage in transplanted ovarian tissue, increased angiogenesis, maintenance of ovarian follicle proliferation and reduced fibrosis areas in the grafted tissue.
Collapse
Affiliation(s)
- Aline Q Rodrigues
- University of Brasilia, Institute of Biological Sciences, Department of Physiological Sciences, Brasilia-DF, 70910-900, Brazil
| | - Isabella Mg Silva
- University of Brasilia, Institute of Biological Sciences, Department of Physiological Sciences, Brasilia-DF, 70910-900, Brazil
| | - Jair T Goulart
- University of Brasilia, Institute of Biological Sciences, Department of Physiological Sciences, Brasilia-DF, 70910-900, Brazil
| | - Luane O Araújo
- University of Brasilia, Institute of Biological Sciences, Department of Physiological Sciences, Brasilia-DF, 70910-900, Brazil
| | - Rayane B Ribeiro
- University of Brasilia, Institute of Biological Sciences, Department of Physiological Sciences, Brasilia-DF, 70910-900, Brazil
| | - Beatriz A Aguiar
- University of Brasilia, Institute of Biological Sciences, Department of Physiological Sciences, Brasilia-DF, 70910-900, Brazil
| | - Yasmin B Ferreira
- University of Brasilia, Institute of Biological Sciences, Department of Physiological Sciences, Brasilia-DF, 70910-900, Brazil
| | - Jessyca Karoline O Silva
- University of Brasilia, Institute of Biological Sciences, Department of Physiological Sciences, Brasilia-DF, 70910-900, Brazil
| | - Julliene Larissa S Bezerra
- University of Brasilia, Institute of Biological Sciences, Department of Physiological Sciences, Brasilia-DF, 70910-900, Brazil
| | - Carolina M Lucci
- University of Brasilia, Institute of Biological Sciences, Department of Physiological Sciences, Brasilia-DF, 70910-900, Brazil
| | - Fernanda Paulini
- University of Brasilia, Institute of Biological Sciences, Department of Physiological Sciences, Brasilia-DF, 70910-900, Brazil.
| |
Collapse
|
22
|
Latif S, Martins Da Silva S, Davies M, Mavrelos D, Foo X, Sangster P, Lane S, Yasmin E. Fertility preservation provision in the NHS: a national assessment of care policies. HUM FERTIL 2023; 26:433-438. [PMID: 35400285 DOI: 10.1080/14647273.2022.2045519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 01/16/2022] [Indexed: 11/04/2022]
Abstract
Fertility preservation has gained momentum in recent years. As cancer survival rates improve, late effects of loss of gonadal function have increased the need to consider fertility preservation. NICE recommends offering cryopreservation of gametes or embryos to patients undergoing gonadotoxic therapy, highlighting that this should be extrapolated to those with non-malignant conditions that pose a risk to fertility. We investigated whether variation in fertility preservation provision exists across the United Kingdom, with a view to identifying equitable models of provision. In England, cryopreservation of gametes and embryos is funded for all patients undergoing treatment for cancer, but eligibility criteria and duration of storage funding vary widely. In Scotland, a national policy is applied, with health boards equitably providing funding for cryopreservation of gametes, embryos, and ovarian and testicular tissue for those undergoing treatment for benign and malignant conditions which impair fertility, including gender incongruence. In Wales and Northern Ireland, cryopreservation of gametes and embryos is funded for those undergoing treatment likely to make them infertile, but ovarian tissue cryopreservation is not funded. Funding criteria for fertility preservation in England, Wales, and Northern Ireland deviates from NICE guidance. Standardization of fertility preservation policies is needed to provide equity of access for patients.
Collapse
Affiliation(s)
- Sania Latif
- Reproductive Medicine Unit, University College London Hospital, London, UK
- Institute for Women's Health, University College London, London, UK
| | | | - Melanie Davies
- Reproductive Medicine Unit, University College London Hospital, London, UK
| | - Dimitrios Mavrelos
- Reproductive Medicine Unit, University College London Hospital, London, UK
| | - Xulin Foo
- Reproductive Medicine Unit, University College London Hospital, London, UK
| | - Philippa Sangster
- Reproductive Medicine Unit, University College London Hospital, London, UK
| | - Sheila Lane
- Department of Paediatric Oncology and Haematology, Oxford University Hospital, Oxford, UK
| | - Ephia Yasmin
- Reproductive Medicine Unit, University College London Hospital, London, UK
| |
Collapse
|
23
|
Ma H, Wang Y, Liu G, Hu Q, Zhu J, Dai Y. Ovarian scaffolds promoted mouse ovary recovery from cyclophosphamide damage. J Reprod Immunol 2023; 157:103950. [PMID: 37079974 DOI: 10.1016/j.jri.2023.103950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 04/08/2023] [Accepted: 04/12/2023] [Indexed: 04/22/2023]
Abstract
There is growing evidence to suggest that scaffold of tissue can promote the tissue reparation. In this study, we investigate the effects of ovarian scaffolds on the reparation of cyclophosphamide (CPA) damaged mice ovaries. The mice were first administered with CPA, was then either transplanted an ovarian scaffold into each ovarian bursa for the experimental group (EG) or underwent sham surgery as the control (CG). To evaluate the extent of ovarian damage caused by CPA, a third group which did not undergo any treatment was included for the normal control (NG). Their ovaries were harvested for examination at day 30, 60, and 90 post CPA injection. We found that in EG, the number of all types of follicles in the ovaries remained almost the same throughout. The numbers of follicles were not significantly different from CG, except at day 60, where in CG the numbers of each type of follicle decreased to basal levels. The decrease in the number of ovarian follicles at day 60 in CG was mirrored by the significant increase in the number of apoptotic granulosa cells in the follicles, and was corroborated further by the basal levels of serum estradiol. Furthermore, we observed a significant decrease in collagen composition preceded by macrophage polarization, and elevation of inflammatory cytokine expression in the ovaries of the EG compared to the CG at day 60. We concluded that ovarian scaffolds can effectively protect primordial follicles from CPA-damage and promote the reparation of CPA-damaged ovaries. This research establishes a proof of concept for the future treatment of chemo-damaged ovaries.
Collapse
Affiliation(s)
- Hongmeng Ma
- College of Biological Sciences, Inner Mongolia University, 235 West Univ. Road, Hohhot, 010021, Inner Mongolia, China
| | - Yuxing Wang
- College of Biological Sciences, Inner Mongolia University, 235 West Univ. Road, Hohhot, 010021, Inner Mongolia, China
| | - Gang Liu
- Key Laboratory of Medical Cell Biology, Clinical Medicine Research Center, Affiliated Hospital of Inner Mongolia Medical University, 1 Tongdao North Street, Hohhot, 010050, Inner Mongolia, China
| | - Qike Hu
- College of Biological Sciences, Inner Mongolia University, 235 West Univ. Road, Hohhot, 010021, Inner Mongolia, China
| | - Jie Zhu
- College of Biological Sciences, Inner Mongolia University, 235 West Univ. Road, Hohhot, 010021, Inner Mongolia, China
| | - Yanfeng Dai
- College of Biological Sciences, Inner Mongolia University, 235 West Univ. Road, Hohhot, 010021, Inner Mongolia, China.
| |
Collapse
|
24
|
Zhang Y, Wang H, Pan X. Live birth in woman with premature ovarian insufficiency and 46, XY karyotype after chemotherapy and bone marrow transplant: a case report. BMC Pregnancy Childbirth 2023; 23:170. [PMID: 36915065 PMCID: PMC10012704 DOI: 10.1186/s12884-023-05464-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 02/22/2023] [Indexed: 03/15/2023] Open
Abstract
BACKGROUND Premature ovarian insufficiency (POI) is a clinical syndrome defined by loss of ovarian function before the age of 40 years, characterized by elevated serum gonadotropin levels and decreased estrogen levels with menstrual disturbance. POI can be natural or iatrogenic such as after chemotherapy, radiotherapy and surgery. CASE PRESENTATION In this study, we describe a successful live birth in a 31-year-old woman with POI and 46, XY Karyotype after being treated with chemotherapy and bone marrow transplant (BMT) for acute non-lymphocytic leukemia when she was 17 years old. With amenorrhea or oligomenorrhea for 11 years, her serum level of FSH was up to 35.0 IU/L and 53.0 IU/L taken 4 weeks apart, which can be diagnosed as POI. After controlled ovarian stimulation treatment for three cycles with different protocols and frozen-thawed embryo transfer (FET), she finally got a successful pregnancy and had a live birth later. CONCLUSIONS This case report serves as a reminder that karyotype of peripheral blood may mislead the diagnosis as disorders of sex development (DSD). It also demonstrates that it is possible for a woman with chemotherapy and bone marrow transplant induced POI can have successful pregnancy and live birth with appropriate therapy. Furthermore, as age may plays a predominant role in fertility rather than residual ovarian reserve, active treatment may be concerned for women with POI at younger age.
Collapse
Affiliation(s)
- Yaojia Zhang
- Department of Assisted Reproduction Center, Huzhou Maternity and Child Health Care Hospital, Huzhou, China
| | - Haiyan Wang
- Department of Assisted Reproduction Center, Huzhou Maternity and Child Health Care Hospital, Huzhou, China
| | - Xiaoqin Pan
- Department of Assisted Reproduction Center, Huzhou Maternity and Child Health Care Hospital, Huzhou, China.
| |
Collapse
|
25
|
Raimondo D, Raspollini A, Vicenti R, Renzulli F, Magnani V, Franceschini C, Raffone A, Mollo A, Casadio P, Seracchioli R. The use of near-infrared imaging with indocyanine green in the ovarian tissue transplantation: a case report. Facts Views Vis Obgyn 2022; 14:353-356. [PMID: 36724430 PMCID: PMC10364327 DOI: 10.52054/fvvo.14.4.051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The request for fertility preservation has consistently increased in recent years. To our knowledge this case report is the first to describe the application of near-infrared intraoperative imaging using indocyanine green (NIR-ICG) during ovarian tissue transplantation (OTT), to assist surgeon choosing the site of implantation of ovarian fragments. OTT was performed in a 42-year-old woman using NIR-ICG to evaluate the vascularisation of peritoneal area as the site of implantation for the ovarian graft. we believe this new approach could be useful in identifying the best reimplantation site.
Collapse
|
26
|
Development and Survival of Human Ovarian Cells in Chitosan Hydrogel Micro-Bioreactor. Medicina (B Aires) 2022; 58:medicina58111565. [DOI: 10.3390/medicina58111565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/20/2022] [Accepted: 10/22/2022] [Indexed: 11/06/2022] Open
Abstract
Background and Objectives: To test the long-term ability of human ovarian cortex cells to develop in unconventional culture conditions. Materials and Methods. Ovarian cortex cells from fetuses aged 23 to 39 weeks gestation were cultured for 90 days in hollow chitosan hydrogel micro-bioreactors and concurrently in traditional wells. Various cell-type counts were considered. Results: With intact follicles as a denominator, the percentage of growing intact follicles at Day 0 varied widely between ovaries (0 to 31.7%). This percentage tended to increase or stay relatively constant in bioreactor as in control cultures; it tended more toward an increase over time in bioreactor vs. control cultures. Modeled percentages showed differences (though not significant) in favor of bioreactor cultures (16.12% difference at D50 but only 0.12% difference at D90). With all follicles present as a denominator, the percentage of growing primary and secondary follicles at D0 varied widely between ovaries (0 to 29.3%). This percentage tended to increase over time in bioreactor cultures but to decrease in control cultures. Modeled percentages showed significant differences in favor of bioreactor cultures (8.9% difference at D50 and 11.1% difference at D90). At D50 and D90, there were only few and sparse apoptotic cells in bioreactor cultures vs. no apoptotic cells in control cultures. Conclusions: Over three months, bioreactor folliculogenesis outperformed slightly traditional culture. This is an interesting perspective for follicle preservation and long-term toxicological studies.
Collapse
|
27
|
Tong Y, Cheng N, Jiang X, Wang K, Wang F, Lin X, Wang F. The Trends and Hotspots in Premature Ovarian Insufficiency Therapy from 2000 to 2022. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph191811728. [PMID: 36142002 PMCID: PMC9517308 DOI: 10.3390/ijerph191811728] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 09/08/2022] [Accepted: 09/12/2022] [Indexed: 05/08/2023]
Abstract
This study aims to map the knowledge structure and themes trends of primary ovarian insufficiency (POI) therapy to help researchers rapidly master the hotspots and prospects of POI therapy from the increasing number of publications. The literature search and bibliometric analyses were performed by using Web of Science Core Collection and VOSviewer. Annual publications from 2000 to 2022 continued to increase with some fluctuations. The most productive country, organization, and journal were the USA, Shanghai Jiao Tong University, and Human Reproduction, respectively. Harvard University was the organization with the highest citation. Fertility and Sterility and Nelson, L.M. were the most influential journal and author, respectively. Seven clusters separated by keywords association showed the extensive scope of POI therapy. The hotspots of POI therapy were hormone replacement therapy and fertility preservation, and the innovative treatment strategies including in vitro activation and mesenchymal stem cells had development potential. In addition, our result showed that the high-cited articles were published in journals with high impact factors. The paper provides a comprehensive overview of the development and hotspots of POI therapy, allowing researchers to recognize the current status and future directions of POI therapy.
Collapse
Affiliation(s)
- Yan Tong
- Department of Literature and Information of Library, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Nan Cheng
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xinran Jiang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Kai Wang
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Fei Wang
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xinxin Lin
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Fang Wang
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, China
- Correspondence:
| |
Collapse
|
28
|
Su H, Zhou X, Zhao Y, Lu Y, Liu D, Zhang J, Yang X. Gynecological complications in long-term survivors after allogeneic hematopoietic cell transplantation—a single-center real-life cross-sectional study. Front Med (Lausanne) 2022; 9:956867. [PMID: 36186762 PMCID: PMC9521594 DOI: 10.3389/fmed.2022.956867] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 08/22/2022] [Indexed: 12/02/2022] Open
Abstract
Background and objectives Hematopoietic stem cell transplantation (HCT) is a treatment for hematopoietic diseases. However, most cured female patients may suffer from premature ovarian insufficiency (POI) after HCT, which is mainly caused by the pre-HCT conditioning regimen. Hence, this study aims to explore the impact of HCT treatment on reproductive and ovarian functions in female survivors. Methods A total of 55 female participants under the age of 40, who underwent HCT and met the inclusion criteria were enrolled. Data related to blood disease, menstruation, and fertility in the 3 years following HCT were collected. Results The involved patients received transplantation at different age stages, ranging from 8 to 37. All patients, except those with aplastic anemia (AA; 5/55), received a myeloablative conditioning regimen, usually modified total body irradiation/cyclophosphamide (TBI/Cy; 25/55) or modified Busulfan/cyclophosphamide (Bu/Cy; 23/55). Among women (42/55) who menstruated before HCT, 16.67% (7/42) had a spontaneous menstrual relapse and 83.3% (35/42) had amenorrhea after HCT. 72.7% (40/55) could be regarded as having POI. This proportion included 100% (25/25) of women aged 21–40 at the time of HCT, 62.5% (15/24) of those aged 11–20, and 0% (0/6) of those ≤10 years old. Patients with AML were more likely to have POI (95.7%). Patients aged ≤10 years (0%) or 11–20 years (16.7%) at the time of HCT were less likely to have moderate to severe menopause than those 21–40 years old (44%). Conclusion The prevalence of POI following HCT was high and POI was associated with age, conditioning regimen, and type of blood disease.
Collapse
Affiliation(s)
- Huina Su
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing, China
| | - Xinyu Zhou
- Department of Obstetrics and Gynecology, Beijing Tsinghua Changgung Hospital, Beijing, China
| | - Yanli Zhao
- Department of Bone Marrow Transplantation, Hebei Yanda Lu Daopei Hospital, Langfang, China
- *Correspondence: Yanli Zhao
| | - Yue Lu
- Department of Bone Marrow Transplantation, Hebei Yanda Lu Daopei Hospital, Langfang, China
| | - DeYan Liu
- Department of Bone Marrow Transplantation, Hebei Yanda Lu Daopei Hospital, Langfang, China
| | - Janping Zhang
- Department of Bone Marrow Transplantation, Hebei Yanda Lu Daopei Hospital, Langfang, China
| | - Xin Yang
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing, China
- Xin Yang
| |
Collapse
|
29
|
Transplantation of Refrozen Ovarian Cortical Strips Retrieved from a Cryopreserved Whole Ovary: Proof of Feasibility. J Clin Med 2022; 11:jcm11174942. [PMID: 36078872 PMCID: PMC9456442 DOI: 10.3390/jcm11174942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 08/11/2022] [Accepted: 08/20/2022] [Indexed: 11/17/2022] Open
Abstract
We report successful clinical outcomes after transplantation of refrozen-rethawed cortical strips from a cryopreserved whole ovary in a patient diagnosed with stage IIIb rectal adenocarcinoma. Whole ovary cryopreservation was proposed as a fertility preservation strategy in 2006 prior to radiotherapy, chemotherapy and oncological surgery. To allow for minimal residual disease screening before ovarian reimplantation, the whole ovary was thawed and dissected into cortical strips. While awaiting the results, the majority of the cortical strips were refrozen. These refrozen-rethawed cortical strips were laparoscopically grafted to 2 sites: the previously irradiated pelvic cavity and the non-irradiated extrapelvic cavity. Ovarian function resumption was assessed by recovery of menses, hormone levels, ultrasound and oocyte pick-up following controlled ovarian stimulation (COS). Restoration of ovarian function occurred 6 months after reimplantation, with recovery of menses and estradiol secretion. A total of 12 cycles were followed by the IVF department. A second reimplantation was performed 1.5 years later, since the grafts were found to have stopped functioning for >3 consecutive months. Overall, 3 fertilizable oocytes were retrieved transabdominally from the extrapelvic graft following COS, yielding 2 embryos and culminating in one fresh embryo transfer, but no pregnancy. Concerning the reimplantation site, no ovarian activity was detected in the graft placed in the previously irradiated pelvic cavity. Indeed, only fibrotic-looking tissue was observed in the pelvic site at second laparoscopy 1.5 years later, while ovarian activity was noted in the extrapelvic graft, showing a large antral follicle. All in all, transplantation of refrozen-rethawed cortical strips from a cryopreserved whole ovary can lead to ovarian function resumption and embryo development if grafted to a non-irradiated field.
Collapse
|
30
|
Li B, Wang W, Huang Y, Han L, Li J, Zheng N, Wu Z, Zhang X, Li X, Deng L, Lin M, Chen X, Zhang M. Lithium treatment promotes the activation of primordial follicles through PI3K/Akt signaling. Biol Reprod 2022; 107:1059-1071. [PMID: 35871551 DOI: 10.1093/biolre/ioac150] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 04/20/2022] [Accepted: 07/17/2022] [Indexed: 11/12/2022] Open
Abstract
Abstract
In mammals, dormant primordial follicles represent the ovarian reserve throughout reproductive life. In vitro activation of dormant primordial follicles has been used to treat patients with premature ovarian insufficiency (POI). However, there remains a lack of effective strategies to stimulate follicle activation in vivo. In this study, we used an in vitro ovarian culture system and intraperitoneal injection to study the effect of lithium treatment on primordial follicle activation. Lithium increased the number of growing follicles in cultured mouse ovaries and promoted pre-granulosa cell proliferation. Furthermore, lithium significantly increased the levels of phosphorylated protein kinase B (Akt) and the number of oocytes with forkhead Box O3a (FOXO3a) nuclear export. Inhibition of the phosphatidylinositol 3 kinase (PI3K)/Akt pathway by LY294002 reversed lithium-promoted mouse primordial follicle activation. These results suggest that lithium promotes mouse primordial follicle activation by the PI3K/Akt signaling. Lithium also promoted primordial follicle activation and increased the levels of p-Akt in mouse ovaries in vivo and in human ovarian tissue cultured in vitro. Taken together, lithium promotes primordial follicle activation in mice and humans by the PI3K/Akt signaling. Lithium might be a potential oral drug for treating infertility in POI patients with residual dormant primordial follicles.
Collapse
Affiliation(s)
- Biao Li
- State Key Laboratory for Agrobiotechnology , College of Biological Sciences, China Agricultural University, Beijing 100193 , China
- Division of Cell , Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou 510006 , China
| | - Weiyong Wang
- Division of Cell , Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou 510006 , China
| | - Yingying Huang
- Division of Cell , Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou 510006 , China
| | - Lincheng Han
- Division of Cell , Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou 510006 , China
| | - Jia Li
- State Key Laboratory for Agrobiotechnology , College of Biological Sciences, China Agricultural University, Beijing 100193 , China
- Division of Cell , Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou 510006 , China
| | - Nana Zheng
- State Key Laboratory for Agrobiotechnology , College of Biological Sciences, China Agricultural University, Beijing 100193 , China
- Division of Cell , Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou 510006 , China
| | - Zhanying Wu
- State Key Laboratory for Agrobiotechnology , College of Biological Sciences, China Agricultural University, Beijing 100193 , China
- Division of Cell , Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou 510006 , China
| | - Xiaodan Zhang
- State Key Laboratory for Agrobiotechnology , College of Biological Sciences, China Agricultural University, Beijing 100193 , China
- Division of Cell , Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou 510006 , China
| | - Xuelan Li
- The Center for Reproductive Medicine , Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan 528300, Guangdong , China
| | - Ling Deng
- The Center for Reproductive Medicine , Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan 528300, Guangdong , China
| | - Min Lin
- The Center for Reproductive Medicine , Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan 528300, Guangdong , China
| | - Xin Chen
- The Center for Reproductive Medicine , Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan 528300, Guangdong , China
| | - Meijia Zhang
- Division of Cell , Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou 510006 , China
| |
Collapse
|
31
|
Xu W, Wu C, Zhu X, Wu J, Zhang Z, Wei Z, Cao Y, Zhou P, Wang J. UC-MSCs promote frozen-thawed ovaries angiogenesis via activation of the Wnt/β-catenin pathway in vitro ovarian culture system. Stem Cell Res Ther 2022; 13:296. [PMID: 35841074 PMCID: PMC9284710 DOI: 10.1186/s13287-022-02989-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 04/07/2022] [Indexed: 12/01/2022] Open
Abstract
Background Ovarian tissue cryopreservation and transplantation are novel therapeutic approaches for fertility preservation. However, follicle loss caused by ischemic and hypoxic damage is one of the issues after frozen-thawed ovarian tissue transplantation. Promoting angiogenesis in grafts is the key to restore cryopreserved ovarian function. Mesenchymal stem cells (MSCs) have been reported to facilitate angiogenesis in the cryopreserved ovarian tissue transplantation. However, the risk of embolization, immunogenic effect and tumorigenesis hinders the clinical application of MSCs to human organ transplantation. In this study, we established an in vitro ovarian culture system to restore frozen-thawed ovarian function before transplantation with the application of umbilical cord mesenchymal stem cells (UC-MSCs), and explored the effects of UC-MSCs on frozen-thawed ovaries in vitro ovarian culture system and the mechanisms of UC-MSCs on the angiogenesis of frozen-thawed ovaries. Methods A simple in vitro three dimensional (3D) ovarian culture system using Matrigel was established to support to an ideal niche, and ovary was alone cultured in the 24-well plate as a control. We also evaluated the effects of UC-MSCs treatment on ovarian function with or without Matrigel support. All thawed ovaries were randomly divided into control group (Matrigel−/UC-MSCs−), Matrigel group (Matrigel+/UC-MSCs−), UC-MSCs group (Matrigel−/UC-MSCs+) and UC-MSCs + Matrigel group (Matrigel+/UC-MSCs+). HE staining was used to detect the histological structure of follicles and TUNEL staining was used to detect cell apoptosis. The number of microvessels was counted to evaluate neovascularization. The mRNA expression of VEGFA, IGF1 and ANGPT2 were detected by RT-PCR. Western blotting was used to measure the expression of GSK-3β, β-catenin and p-β-catenin. Results In the absence of UC-MSCs, 3D culture system supported by Matrigel showed significantly improved follicular development and microvascular number. Additionally, UC-MSCs were also found to effectively improve follicular development and microvascular number regardless of the culture condition used. However, alleviated follicular apoptosis, increased mRNA expression of angiogenesis-related gene and activated Wnt/β-catenin pathway occurred only in the UC-MSCs + Matrigel group. Besides, with the application of IWP-2 in UC-MSCs + Matrigel group, Wnt//β-catenin pathway could be blocked by IWP-2 serving as one of Wnt/β-catenin pathway inhibitors. Conclusions This in vitro study showed the beneficial effects of UC-MSCs on thawed ovaries and explored a potential mechanism inducing angiogenesis. In particular, 3D ovarian culture system supported by Matrigel further improved UC-MSCs treatment. The in vitro culture system using Matrigel and UC-MSCs may provide a potential treatment strategy for improving the success rate of thawed ovaries transplantation. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-02989-8.
Collapse
Affiliation(s)
- Wenjuan Xu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, 230022, Anhui, China.,Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China.,NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Caiyun Wu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, 230022, Anhui, China.,Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China.,NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Xiaoqian Zhu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, 230022, Anhui, China.,Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China.,NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Jingjing Wu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, 230022, Anhui, China.,Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China.,NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Zhiguo Zhang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, 230022, Anhui, China.,Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China.,NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Zhaolian Wei
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, 230022, Anhui, China.,Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China.,NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Yunxia Cao
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, 230022, Anhui, China.,Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China.,NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Ping Zhou
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, 230022, Anhui, China. .,Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China. .,NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China.
| | - Jianye Wang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, 230022, Anhui, China. .,Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China. .,NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China.
| |
Collapse
|
32
|
Zhang XR, Ouyang YC, Meng TG, Zhang HY, Yue W, Yan FZ, Xue Y, Schatten H, Wang ZB, Sun QY. OTSSP167 leads to follicular dysplasia and negatively affects oocyte quality in mice. Toxicology 2022; 476:153243. [PMID: 35760214 DOI: 10.1016/j.tox.2022.153243] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 06/11/2022] [Accepted: 06/23/2022] [Indexed: 11/16/2022]
Abstract
OTSSP167 is an anti-tumor drug significantly inhibiting tumor growth in xenotransplantation studies using mouse breast, lung, prostate, and pancreatic cancer cell lines. Its phase I clinical trial has been completed, indicating its great potential for future treatment of solid tumors. However, its drug-related adverse effects on reproductive systems have not yet been reported. In this study, we evaluated the effects of OTSSP167 on reproduction of female mice by determining oocyte quality and follicular development. We selected four-week-old female ICR mice for a 21-day intraperitoneal injection of OTSSP167 at a dose of 5mg/kg/d. We found that OTSSP167 could block the meiotic process of oocytes, leading to a decrease in oocyte maturation and ovulated oocyte numbers, as well as a decrease in the quality of oocytes. The results showed that OTSSP167 treatment caused disordered spindle assembly, decreased mitochondria membrane potential, and increased accumulation of reactive oxygen species in oocytes. Further investigation showed that OTSSP167 induced DNA double-strand breaks, as indicated by increased levels of γH2AX in oocytes of primordial follicles and granulosa cells of growing follicles, which induced follicular atresia and decreased the numbers of follicles at various growing stages. Our study suggests that OTSSP167 treatment may have serious effects on the ovary and consequences for female cancer patients, providing strong evidence for the necessity of protecting female fertility in clinical OTSSP167 trials.
Collapse
Affiliation(s)
- Xin-Ran Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China; College of Life Science, University of Chinese Academy of Science, 100101, Beijing
| | - Ying-Chun Ouyang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
| | - Tie-Gang Meng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
| | - Hong-Yong Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
| | - Wei Yue
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China; College of Life Science, University of Chinese Academy of Science, 100101, Beijing
| | - Feng-Ze Yan
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China; College of Life Science, University of Chinese Academy of Science, 100101, Beijing
| | - Yue Xue
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
| | - Heide Schatten
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, 65211, USA
| | - Zhen-Bo Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China; College of Life Science, University of Chinese Academy of Science, 100101, Beijing
| | - Qing-Yuan Sun
- Fertility Preservation Lab, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China.
| |
Collapse
|
33
|
Kim HY, Kim SW. History of fertility preservation. JOURNAL OF THE KOREAN MEDICAL ASSOCIATION 2022. [DOI: 10.5124/jkma.2022.65.6.322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Background: Fertility preservation refers to a procedure performed to maintain the ability to become pregnant before receiving treatment with a risk of fertility loss, such as chemo- or radiation therapy. Examples of fertility-preserving procedures include freezing, sperm freezing, embryo freezing through in vitro fertilization, and ovarian tissue freezing.Current Concepts: Until the late 1990s, awareness of fertility preservation among clinicians and patients was relatively low, and the only way to preserve and restore fertility in women with cancer was the cryopreservation of embryos. However, as the survival rate of cancer patients increased and the treatment results of various diseases improved, interest in quality of life such as pregnancy and childbirth after treatment gradually increased, and became a driving force for the development of fertility preservation. In the 2000s, several centers began cryopreserving ovarian tissue, including primordial follicles from young patients before chemotherapy. Currently, ovarian tissue cryopreservation can be used in combination with in vitro maturation and egg vitrification techniques. Novel methods to improve follicle survival after transplantation are currently being investigated. Methods to improve follicle survival after transplantation and new ovarian protective agents to protect the ovaries from cytotoxic agents are currently being studied.Discussion and Conclusion: Advances in fertility-preserving technologies in the future will contribute to the delivery of healthy children by providing tailored treatments and more individualized fertility-preserving strategies to patients whose fertility is at risk.
Collapse
|
34
|
Papageorgiou D, Diakosavvas M, Angelou K, Kathopoulis N, Voros C, Zachariou E, Papadatou K, Papapanagiotou IK, Papakonstantinou K. Fertility outcomes after treatment with intraperitoneal chemotherapy. J OBSTET GYNAECOL 2022; 42:1626-1634. [PMID: 35612269 DOI: 10.1080/01443615.2022.2055453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Intraperitoneal chemotherapy (IPC) administration has been added to the therapeutic protocols of cancers, confined to the abdominal cavity. Since, a survival benefit in patients treated with adjuvant IPC has been demonstrated, fertility questions are raised in these patients. A comprehensive search of the English literature of PubMed/MEDLINE, EmBase and Google Scholar databases was conducted, from their inception until April 2021, following the MOOSE guidelines. Twelve out of 473 studies were included in the final review and analysis. After treatment with IPC for peritoneal carcinomatosis, 19 women, who underwent fertility-sparing surgery, with 20 successful pregnancies were reported. The mean interval time between IPC and pregnancy was 38.4 months (range 9-168 months). In 16 cases, conception was spontaneous, three required in vitro fertilisation, whereas one pregnancy was achieved through intrauterine insemination. Mean disease-free survival was 76 months (range 24-177 months). Childbearing is a feasible approach in selected patients, after treatment with IPC. When future pregnancy is desirable, a multidisciplinary team of surgeons, gynaecologists, oncologists and reproductive specialists is required, to inform the patient thoroughly about the fertility preservation alternatives, without endangering patient's survival.
Collapse
Affiliation(s)
- Dimitrios Papageorgiou
- Department of Gynecology, Athens Naval and Veterans Hospital, Athens, Greece.,1st Department of Obstetrics and Gynecology, Alexandra Maternity Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Michail Diakosavvas
- 1st Department of Obstetrics and Gynecology, Alexandra Maternity Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Kyveli Angelou
- 1st Department of Obstetrics and Gynecology, Alexandra Maternity Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolaos Kathopoulis
- 1st Department of Obstetrics and Gynecology, Alexandra Maternity Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Charalambos Voros
- 1st Department of Obstetrics and Gynecology, Alexandra Maternity Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Eleftherios Zachariou
- 1st Department of Obstetrics and Gynecology, Alexandra Maternity Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Konstantina Papadatou
- 1st Department of Obstetrics and Gynecology, Alexandra Maternity Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioannis K Papapanagiotou
- 1st Department of Obstetrics and Gynecology, Alexandra Maternity Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | | |
Collapse
|
35
|
Di Tucci C, Galati G, Mattei G, Chinè A, Fracassi A, Muzii L. Fertility after Cancer: Risks and Successes. Cancers (Basel) 2022; 14:2500. [PMID: 35626104 PMCID: PMC9139810 DOI: 10.3390/cancers14102500] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/08/2022] [Accepted: 05/16/2022] [Indexed: 12/22/2022] Open
Abstract
The incidence of cancer in reproductive-aged women is 7%, but, despite the increased number of cancer cases, advances in early diagnosis and treatment have raised the survival rate. Furthermore, in the last four decades, there has been a rising trend of delaying childbearing. There has been an increasing number of couples referred to Reproductive Medicine Centers for infertility problems after one partner has been treated for cancer. In these cases, the main cause of reduced fertility derives from treatments. In this review, we describe the effects and the risks of chemotherapy, radiotherapy, and surgery in women with cancer, and we will focus on available fertility preservation techniques and their efficacy in terms of success in pregnancy and live birth rates.
Collapse
Affiliation(s)
- Chiara Di Tucci
- Department of Obstetrics and Gynecology, “Sapienza” University, 00185 Rome, Italy; (G.G.); (G.M.); (A.C.); (A.F.); (L.M.)
| | | | | | | | | | | |
Collapse
|
36
|
Khattak H, Malhas R, Craciunas L, Afifi Y, Amorim CA, Fishel S, Silber S, Gook D, Demeestere I, Bystrova O, Lisyanskaya A, Manikhas G, Lotz L, Dittrich R, Colmorn LB, Macklon KT, Hjorth IMD, Kristensen SG, Gallos I, Coomarasamy A. Fresh and cryopreserved ovarian tissue transplantation for preserving reproductive and endocrine function: a systematic review and individual patient data meta-analysis. Hum Reprod Update 2022; 28:400-416. [PMID: 35199164 PMCID: PMC9733829 DOI: 10.1093/humupd/dmac003] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 12/29/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Ovarian tissue cryopreservation involves freezing and storing of surgically retrieved ovarian tissue in liquid or vapour nitrogen below -190°C. The tissue can be thawed and transplanted back with the aim of restoring fertility or ovarian endocrine function. The techniques for human ovarian tissue freezing and transplantation have evolved over the last 20 years, particularly in the context of fertility preservation in pre-pubertal cancer patients. Fresh ovarian tissue transplantation, using an autograft or donor tissue, is a more recent development; it has the potential to preserve fertility and hormonal function in women who have their ovaries removed for benign gynaecological conditions. The techniques of ovarian tissue cryopreservation and transplantation have progressed rapidly since inception; however, the evidence on the success of this intervention is largely based on case reports and case series. OBJECTIVE AND RATIONALE The aim of this study was to systematically review the current evidence by incorporating study-level and individual patient-level meta-analyses of women who received ovarian transplants, including frozen-thawed transplant, fresh or donor graft. SEARCH METHODS The review protocol was registered with PROSPERO (CRD42018115233). A comprehensive literature search was performed using MEDLINE, EMBASE, CINAHL and Cochrane Central Register of Controlled Trials from database inception to October 2020. Authors were also contacted for individual patient data if relevant outcomes were not reported in the published manuscripts. Meta-analysis was performed using inverse-variance weighting to calculate summary estimates using a fixed-effects model. OUTCOMES The review included 87 studies (735 women). Twenty studies reported on ≥5 cases of ovarian transplants and were included in the meta-analysis (568 women). Fertility outcomes included pregnancy, live birth and miscarriage rates, and endocrine outcomes included oestrogen, FSH and LH levels. The pooled rates were 37% (95% CI: 32-43%) for pregnancy, 28% (95% CI: 24-34%) for live birth and 37% (95% CI: 30-46%) for miscarriage following frozen ovarian tissue transplantation. Pooled mean for pre-transplant oestrogen was 101.6 pmol/l (95% CI: 47.9-155.3), which increased post-transplant to 522.4 pmol/l (95% CI: 315.4-729; mean difference: 228.24; 95% CI: 180.5-276). Pooled mean of pre-transplant FSH was 66.4 IU/l (95% CI: 52.8-84), which decreased post-transplant to 14.1 IU/l (95% CI: 10.9-17.3; mean difference 61.8; 95% CI: 57-66.6). The median time to return of FSH to a value <25 IU/l was 19 weeks (interquartile range: 15-26 weeks; range: 0.4-208 weeks). The median duration of graft function was 2.5 years (interquartile range: 1.4-3.4 years; range: 0.7-5 years). The analysis demonstrated that ovarian tissue cryopreservation and transplantation could restore reproductive and hormonal functions in women. Further studies with larger samples of well-characterized populations are required to define the optimal retrieval, cryopreservation and transplantation processes. WIDER IMPLICATIONS Ovarian tissue cryopreservation and transplantation may not only be effective in restoring fertility but also the return of reproductive endocrine function. Although this technology was developed as a fertility preservation option, it may have the scope to be considered for endocrine function preservation.
Collapse
Affiliation(s)
- Hajra Khattak
- Tommy’s National Centre for Miscarriage Research, Institute of Metabolism and
Systems Research, University of Birmingham, Birmingham, UK
| | - Rosamund Malhas
- Birmingham Women’s and Children’s NHS Foundation Trust,
Birmingham, UK
| | - Laurentiu Craciunas
- Population Health Sciences Institute, Newcastle University, Newcastle upon
Tyne, UK
| | - Yousri Afifi
- Birmingham Women’s and Children’s NHS Foundation Trust,
Birmingham, UK
| | - Christiani A Amorim
- Pôle de Recherche en Gynécologie, Institut de Recherche Expérimentale et
Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Simon Fishel
- CARE Fertility Group, Nottingham, UK
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores
University, Liverpool, UK
| | | | - Debra Gook
- Reproductive Services/Melbourne IVF, The Royal Women’s Hospital,
Parkville, VIC, Australia
| | - Isabelle Demeestere
- Research Laboratory on Human Reproduction, Faculty of Medicine, Université
Libre de Bruxelles (ULB), Brussels, Belgium
| | - Olga Bystrova
- AVA-PETER Fertility Clinic, Saint-Petersburg, Russia
| | - Alla Lisyanskaya
- Division of Gynecologic Oncology, Saint-Petersburg City Oncology
Clinic, Saint-Petersburg, Russia
| | - Georgy Manikhas
- Department of Oncology of the First Pavlov State Medical University of
Saint-Petersburg, Saint-Petersburg, Russia
| | - Laura Lotz
- Department of Obstetrics and Gynecology, Erlangen University Hospital,
Friedrich Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Ralf Dittrich
- Department of Obstetrics and Gynecology, Erlangen University Hospital,
Friedrich Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Lotte Berdiin Colmorn
- The Fertility Clinic, University Hospital of Copenhagen,
Rigshospitalet, Copenhagen, Denmark
| | - Kirsten Tryde Macklon
- The Fertility Clinic, University Hospital of Copenhagen,
Rigshospitalet, Copenhagen, Denmark
| | | | - Stine Gry Kristensen
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women,
Children and Reproduction, University Hospital of Copenhagen,
Rigshospitalet, Copenhagen, Denmark
| | - Ioannis Gallos
- Tommy’s National Centre for Miscarriage Research, Institute of Metabolism and
Systems Research, University of Birmingham, Birmingham, UK
| | - Arri Coomarasamy
- Tommy’s National Centre for Miscarriage Research, Institute of Metabolism and
Systems Research, University of Birmingham, Birmingham, UK
| |
Collapse
|
37
|
Khattak H, Gallos I, Coomarasamy A, Topping AE. Why are women considering ovarian tissue cryopreservation to preserve reproductive and hormonal ovarian function? A qualitative study protocol. BMJ Open 2022; 12:e051288. [PMID: 35418423 PMCID: PMC9013984 DOI: 10.1136/bmjopen-2021-051288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 03/28/2022] [Indexed: 11/26/2022] Open
Abstract
INTRODUCTION Current fertility preservation options available to women are oocyte cryopreservation (egg freezing) or embryo cryopreservation. A newer procedure, ovarian tissue cryopreservation (OTC), has become available in some centres, which offers another option for women and girls considering fertility preservation. These procedures are commonly offered to women about to undergo treatments for cancer. OTC involves removing sections of ovarian tissue and cryopreserving it for future reimplantation, often several years later. OTC offers girls and women who may become infertile with optionality and the possibility of pregnancy. OTC has potential for other applications, including restoring ovarian endocrine function beyond biological menopause. This is not without controversy but has led to some women considering undergoing the procedure for purposes of ovarian hormonal preservation (conservation of ovarian endocrine function). OTC is invasive, involves two surgical procedures with concomitant risks and can be costly. Understanding why women may consider and ultimately undergo OTC is timely, so that evidence-based and women-centred care can be provided. METHODS A pragmatic narrative qualitative design will be used. A purposive sample of women aged 18-45 who are considering, or have sought, OTC will be recruited over 1-year period. Potential participants will be approached via a clinic that offers OTC on a private basis or via social media. ANALYSIS Participant interviews will be audio and, if consented, video recorded. These will be conducted face-to-face or virtually. The recordings will be transcribed verbatim and analysed using a thematic analysis approach supported by NVivo software. ETHICS AND DISSEMINATION Ethical approval has been granted by the Institutional Ethical Review ERN_19-1578A. We expect to disseminate the findings of this study through journal articles, conference presentations and multimedia to public.
Collapse
Affiliation(s)
- Hajra Khattak
- Tommy's National Centre for Miscarriage Research, Institute of Metabolism and Systems Research, University of Birmingham College of Medical and Dental Sciences, Birmingham, UK
| | - Ioannis Gallos
- Tommy's National Centre for Miscarriage Research, Institute of Metabolism and Systems Research, University of Birmingham College of Medical and Dental Sciences, Birmingham, UK
| | - Arri Coomarasamy
- Tommy's National Centre for Miscarriage Research, Institute of Metabolism and Systems Research, University of Birmingham College of Medical and Dental Sciences, Birmingham, UK
| | - A E Topping
- Institute of Clinical Sciences, School of Nursing, University Hospitals Birmingham NHS Foundation Trust, University of Birmingham, Birmingham, UK
| |
Collapse
|
38
|
Lipovac M, Aschauer J, Imhof H, Herrmann C, Sima M, Weiß P, Imhof M. The effect of micronutrient supplementation on serum anti-Mullerian hormone levels: a retrospective pilot study. Gynecol Endocrinol 2022; 38:310-313. [PMID: 35147056 DOI: 10.1080/09513590.2022.2028770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
BACKGROUND The anti-Müllerian-hormone (AMH) is secreted by the granulosa cells of the oocytes and can be used as a marker of the ovarian reserve; helpful to estimate female fertility or the menopause onset. Although various factors may influence AMH levels, the correlation with nutritional factors needs more research. OBJECTIVE To evaluate the effect of a micronutrient supplementation on female AMH levels. METHODS This retrospective analysis includes a total of 244 women, who attended the Karl Landsteiner Institute, Korneuburg, Austria from January 2013 to June 2019 due to an unfulfilled desire for a child. All women were treated with an oral micronutrient preparation consisting the dosage of one soft capsule and one tablet per day for 3 months. The soft capsule contains omega-3 fatty acids and the tablet is a standardized combination of coenzyme Q10, vitamin E, folic acid, selenium, catechins from green tea extract, and glycyrrhizin from licorice extract. Serum AMH levels before and after 3 months were compared. In addition, available clinical data such as ovulation frequency, endometrium thickness, and luteal phase duration were analyzed. RESULTS The mean age of the women was 37.3 ± 1.8 years, the mean body mass index of 24.3 ± 4.6 k/m2. The mean serum AMH levels and endometrial thickness values were significantly higher after micronutrient supplementation as compared to baseline (1.42 ± 0.86 versus 1.86 ± 0.82 ng/mL and 6.10 ± 1.76 versus 7.29 ± 1.65 mm, respectively). In addition, ovulation frequency and luteal phase duration significantly improved in more than 60%. CONCLUSION Proposed micronutrient supplementation had a positive effect on serum AMH levels, endometrial thickness, ovulation frequency, and luteal phase duration. It could be a simple, risk-free therapeutic option to improve female fertility. More research is warranted to prove this effect.
Collapse
Affiliation(s)
- Markus Lipovac
- IMI Fertility Center, Vienna, Austria
- Karl Landsteiner Institute for Cell-Based Therapy in Gynecology, Korneuburg, Austria
| | | | | | | | | | | | - Martin Imhof
- IMI Fertility Center, Vienna, Austria
- Karl Landsteiner Institute for Cell-Based Therapy in Gynecology, Korneuburg, Austria
| |
Collapse
|
39
|
Kim YY, Yun JW, Kim SW, Kim H, Kang BC, Ku SY. Synergistic Promoting Effects of X-Linked Inhibitor of Apoptosis Protein and Matrix on the In Vitro Follicular Maturation of Marmoset Follicles. Tissue Eng Regen Med 2022; 19:93-103. [PMID: 34741748 PMCID: PMC8782987 DOI: 10.1007/s13770-021-00387-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 08/02/2021] [Accepted: 08/16/2021] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND In vitro follicular maturation (IVFM) of ovarian follicles is an emerging option for fertility preservation. Many paracrine factors and two-dimensional or three-dimensional (3D) environments have been used for optimization. However, since most studies were conducted using the murine model, the physiological differences between mice and humans limit the interpretation and adaptation of the results. Marmoset monkey is a non-human primate (NHPs) with more similar reproductive physiology to humans. In this study, we attempted to establish a 3D matrix (Matrtigel)-based IVFM condition for marmoset ovarian follicles in combination with anti-apoptotic factor, X-linked inhibitor of apoptosis protein (XIAP). METHODS Marmoset follicles were isolated as individual follicles and cultured in a single drop with the addition of 0, 10, and 100 μg/mL of XIAP molecules. Matured oocytes and granulosa cells from mature follicles were collected and analyzed. The average number of isolated follicles was less than 100, and primordial and antral follicles were abundant in the ovaries. RESULTS IVFM of marmoset follicles in 3D matrix conditions with XIAP increased the rates of survival and in vitro follicle development. Furthermore, oocytes from the 3D cultures were successfully fertilized and developed in vitro. The addition of XIAP increased the secretion of estradiol and aromatase. Furthermore, expression of granulosa-specific genes, such as bone morphogenetic protein 15, Oct4, and follicle-stimulating hormone receptor were upregulated in the in vitro-matured follicles than in normal, well-grown, and atretic follicles. Apoptosis-related B-cell lymphoma-2 was highly expressed in the atretic follicles than in the XIAP-treated follicles, and higher caspase-3 was localized in the XIAP-treated follicles. CONCLUSION In this study, we attempted to establish a 3D-matrix-based marmoset IVFM condition and demonstrated the synergistic effects of XIAP. The use of a 3D matrix may be applied as an optimal culture condition for marmoset ovarian follicles.
Collapse
Affiliation(s)
- Yoon Young Kim
- Department of Obstetrics and Gynecology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Korea
- Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Korea
| | - Jun-Won Yun
- Department of Medical and Biological Sciences, The Catholic University of Korea, 327 Sosa-ro, Bucheon, 14662, Korea
| | - Sung Woo Kim
- Department of Obstetrics and Gynecology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Korea
- Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Korea
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Korea
| | - Hoon Kim
- Department of Obstetrics and Gynecology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Korea
- Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Korea
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Korea
| | - Byeong-Cheol Kang
- Department of Medicine, Seoul National University College of Medicine, 103 Daehak ro, Jongno-gu, Seoul, 03080, Korea
| | - Seung-Yup Ku
- Department of Obstetrics and Gynecology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Korea.
- Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Korea.
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Korea.
| |
Collapse
|
40
|
Dolmans MM, Hossay C, Nguyen TYT, Poirot C. Fertility Preservation: How to Preserve Ovarian Function in Children, Adolescents and Adults. J Clin Med 2021; 10:jcm10225247. [PMID: 34830528 PMCID: PMC8621487 DOI: 10.3390/jcm10225247] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/04/2021] [Accepted: 11/08/2021] [Indexed: 01/15/2023] Open
Abstract
Chemotherapy, pelvic radiotherapy and ovarian surgery have known gonadotoxic effects that can lead to endocrine dysfunction, cessation of ovarian endocrine activity and early depletion of the ovarian reserve, causing a risk for future fertility problems, even in children. Important determinants of this risk are the patient’s age and ovarian reserve, type of treatment and dose. When the risk of premature ovarian insufficiency is high, fertility preservation strategies must be offered to the patient. Furthermore, fertility preservation may sometimes be needed in conditions other than cancer, such as in non-malignant diseases or in patients seeking fertility preservation for personal reasons. Oocyte and/or embryo vitrification and ovarian tissue cryopreservation are the two methods currently endorsed by the American Society for Reproductive Medicine, yielding encouraging results in terms of pregnancy and live birth rates. The choice of one technique above the other depends mostly on the age and pubertal status of the patient, and personal and medical circumstances. This review focuses on the available fertility preservation techniques, their appropriateness according to patient age and their efficacy in terms of pregnancy and live birth rates.
Collapse
Affiliation(s)
- Marie-Madeleine Dolmans
- Gynecology Research Unit, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Av. Mounier 52, 1200 Brussels, Belgium; (C.H.); (T.Y.T.N.)
- Department of Gynecology, Cliniques Universitaires Saint-Luc, Av. Hippocrate 10, 1200 Brussels, Belgium
- Correspondence:
| | - Camille Hossay
- Gynecology Research Unit, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Av. Mounier 52, 1200 Brussels, Belgium; (C.H.); (T.Y.T.N.)
| | - Thu Yen Thi Nguyen
- Gynecology Research Unit, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Av. Mounier 52, 1200 Brussels, Belgium; (C.H.); (T.Y.T.N.)
| | - Catherine Poirot
- Department of Hematology, AYA Unit, Saint Louis Hospital AP-HP, 1 Avenue Claude Vellefaux, 75010 Paris, France;
- Médecine Sorbonne Université, Site Pitié Salpêtrière, 91 Bd de l’Hôpital, 75013 Paris, France
- Department of Reproductive Biology, Cochin Hospital AP-HP, 123 Bd de Port Royal, 75014 Paris, France
| |
Collapse
|
41
|
Rodrigues AQ, Picolo VL, Goulart JT, Silva IMG, Ribeiro RB, Aguiar BA, Ferreira YB, Oliveira DM, Lucci CM, de Bem AF, Paulini F. Metabolic activity in cryopreserved and grafted ovarian tissue using high-resolution respirometry. Sci Rep 2021; 11:21517. [PMID: 34728762 PMCID: PMC8563867 DOI: 10.1038/s41598-021-01082-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 10/19/2021] [Indexed: 11/09/2022] Open
Abstract
Cryopreservation of ovarian tissue followed by transplantation represents a strategy to restore ovarian function and fertility. Stress from cryopreservation-thawing processes can lead to alterations and/or damage to mitochondrial structure and functionality. High resolution respirometry and histological analysis were used to evaluate the effect of cryopreservation and transplantation on ovarian tissue. Four different conditions were performed: Fresh non-transplanted tissue, Fresh transplanted tissue, Cryopreserved non-transplanted tissue and Cryopreserved transplanted tissue. All groups were able to respond to the substrates-uncoupler-inhibitor protocol. We found a dramatic decrease in general oxygen consumption in hemi-ovaries submitted to cryopreservation and/or transplantation. The effect of cryopreservation on mitochondrial metabolism was less intense than effect of transplantation, since the transplantation affected all of the mitochondrial states. A total of 2644 follicles were analyzed. Of these, 2198 were classified as morphologically normal. The percentage of morphologically normal follicles was significantly lower in the Cryopreserved transplanted group when compared to the Cryopreserved non-transplanted group and the Fresh transplanted group (p-value < 0.05). Despite decreased follicular viability and mitochondrial activity, the cryopreservation followed by transplantation of ovarian tissue proved feasible for attempts to restore ovarian function.
Collapse
Affiliation(s)
- Aline Q Rodrigues
- Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, DF, 70910-900, Brazil
| | - Victor L Picolo
- Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, DF, 70910-900, Brazil
| | - Jair T Goulart
- Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, DF, 70910-900, Brazil
| | - Isabella M G Silva
- Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, DF, 70910-900, Brazil
| | - Rayane B Ribeiro
- Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, DF, 70910-900, Brazil
| | - Beatriz A Aguiar
- Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, DF, 70910-900, Brazil
| | - Yasmin B Ferreira
- Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, DF, 70910-900, Brazil
| | - Daniela M Oliveira
- Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasília, Brasília, 70910-900, Brazil
| | - Carolina M Lucci
- Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, DF, 70910-900, Brazil
| | - Andreza F de Bem
- Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, DF, 70910-900, Brazil
| | - Fernanda Paulini
- Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, DF, 70910-900, Brazil.
| |
Collapse
|
42
|
Dadashzadeh A, Moghassemi S, Shavandi A, Amorim CA. A review on biomaterials for ovarian tissue engineering. Acta Biomater 2021; 135:48-63. [PMID: 34454083 DOI: 10.1016/j.actbio.2021.08.026] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/26/2021] [Accepted: 08/18/2021] [Indexed: 12/19/2022]
Abstract
Considerable challenges in engineering the female reproductive tissue are the follicle's unique architecture, the need to recapitulate the extracellular matrix, and tissue vascularization. Over the years, various strategies have been developed for preserving fertility in women diagnosed with cancer, such as embryo, oocyte, or ovarian tissue cryopreservation. While autotransplantation of cryopreserved ovarian tissue is a viable choice to restore fertility in prepubertal girls and women who need to begin chemo- or radiotherapy soon after the cancer diagnosis, it is not suitable for all patients due to the risk of having malignant cells present in the ovarian fragments in some types of cancer. Advances in tissue engineering such as 3D printing and ovary-on-a-chip technologies have the potential to be a translational strategy for precisely recapitulating normal tissue in terms of physical structure, vascularization, and molecular and cellular spatial distribution. This review first introduces the ovarian tissue structure, describes suitable properties of biomaterials for ovarian tissue engineering, and highlights recent advances in tissue engineering for developing an artificial ovary. STATEMENT OF SIGNIFICANCE: The increase of survival rates in young cancer patients has been accompanied by a rise in infertility/sterility in cancer survivors caused by the gonadotoxic effect of some chemotherapy regimens or radiotherapy. Such side-effect has a negative impact on these patients' quality of life as one of their main concerns is generating biologically related children. To aid female cancer patients, several research groups have been resorting to tissue engineering strategies to develop an artificial ovary. In this review, we discuss the numerous biomaterials cited in the literature that have been tested to encapsulate and in vitro culture or transplant isolated preantral follicles from human and different animal models. We also summarize the recent advances in tissue engineering that can potentially be optimal strategies for developing an artificial ovary.
Collapse
|
43
|
Eshghifar N, Dehghan BK, Do AA, Koukhaloo SZ, Habibi M, Pouresmaeili F. Infertility cell therapy and epigenetic insights. Hum Antibodies 2021; 29:17-26. [PMID: 33554898 DOI: 10.3233/hab-200438] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Recent advances in assisted reproductive technology (ART) have allowed couples with severe infertility to conceive, but the methods are not effective for all cases. Stem cells as undifferentiated cells which are found in different stages of embryonic, fetal and adult life are known to be capable of forming different cell types, tissues, and organs. Due to their unlimited resources and the incredible power of differentiation are considered as potential new therapeutic biological tools for treatment of infertility. For reproductive medicine, stem cells are stimulated in vitro to develop various specialized functional cells including male and female gametes. The epigenetic patterns can be modified in the genome under certain drugs exposure or lifestyle alterations. Therefore, epigenetics-related disorders may be treated if the nature of the modifications is completely admissible. It is proved that our understanding of epigenetic processes and its association with infertility would help us not only to understand the etiological factors but also to treat some type of male infertilities. Exploration of both genetic and epigenetic variations in the disease development could help in the identification of the interaction patterns between these two phenomena and possible improvement of therapeutic methods.
Collapse
Affiliation(s)
- Nahal Eshghifar
- Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Behnam Kamali Dehghan
- Department of Medical Genetics, National Institute of Medical Engineering and Biotechnology (NIGEB), Tehran, Iran.,Medical Genetics, Jiroft University of Medical Sciences and Health Services, Jiroft, Kerman, Iran.,Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Atieh Abedin Do
- Groupe de Recherche en Écologie Buccale, Faculté de Médecine Dentaire, Université Laval, Quebec, Canada
| | | | - Mohsen Habibi
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farkhondeh Pouresmaeili
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
44
|
Hao X, Anastácio A, Rodriguez-Wallberg KA. Feasibility of Secondary Follicle Isolation, Culture and Achievement of In-Vitro Oocyte Maturation from Superovulated Ovaries: An Experimental Proof-of-Concept Study Using Mice. J Clin Med 2021; 10:jcm10132757. [PMID: 34201725 PMCID: PMC8268951 DOI: 10.3390/jcm10132757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 06/08/2021] [Accepted: 06/18/2021] [Indexed: 11/16/2022] Open
Abstract
Fertility preservation through ovarian stimulation, aiming at cryopreserving mature oocytes or embryos, is sometimes unsuccessful. This clinical situation deserves novel approaches to overcome infertility following cancer treatment in patients facing highly gonadotoxic treatment. In this controlled experimental study, we investigated the feasibility of in-vitro culturing secondary follicles isolated from superovulated ovaries of mice recently treated with gonadotropins. The follicle yields of superovulated ovaries were 45.9% less than in unstimulated controls. Follicles from superovulated ovaries showed faster growth pace during the initial 7 days of culture and secreted more 17β-estradiol by the end of culture vs controls. Parameters reflecting the outcome of follicular development and oocyte maturation competence in vitro were similar between superovulated and control groups, with a similar follicle size at the end of culture and around 70% survival. Nearly half of cultured follicles met the criteria for in-vitro maturation in both groups and approximately 60% of those achieved a mature MII oocyte, similarly in both groups. Over 60% of obtained MII oocytes displayed normal-looking spindle and chromosome configurations, without significant differences between the groups. Using a validated follicle culture system, we demonstrated the feasibility of secondary follicle isolation, in-vitro culture and oocyte maturation with normal spindle and chromosome configurations obtained from superovulated mice ovaries.
Collapse
Affiliation(s)
- Xia Hao
- Department of Oncology and Pathology, Karolinska Institutet, Solna, SE-171 76 Stockholm, Sweden; (X.H.); (A.A.)
- Laboratory of Translational Fertility Preservation, BioClinicum, SE-171 64 Stockholm, Sweden
| | - Amandine Anastácio
- Department of Oncology and Pathology, Karolinska Institutet, Solna, SE-171 76 Stockholm, Sweden; (X.H.); (A.A.)
- Laboratory of Translational Fertility Preservation, BioClinicum, SE-171 64 Stockholm, Sweden
| | - Kenny A. Rodriguez-Wallberg
- Department of Oncology and Pathology, Karolinska Institutet, Solna, SE-171 76 Stockholm, Sweden; (X.H.); (A.A.)
- Laboratory of Translational Fertility Preservation, BioClinicum, SE-171 64 Stockholm, Sweden
- Department of Reproductive Medicine, Division of Gynecology and Reproduction, Karolinska University Hospital, Novumhuset Plan 4, SE-141 86 Stockholm, Sweden
- Correspondence:
| |
Collapse
|
45
|
Çil N, Mete GA. The effect of adipose-derived mesenchymal stem cell treatment on mTOR and p-mTOR expression in ovarian damage due to cyclophosphomide. Reprod Toxicol 2021; 103:71-78. [PMID: 34098046 DOI: 10.1016/j.reprotox.2021.06.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/29/2021] [Accepted: 06/01/2021] [Indexed: 12/20/2022]
Abstract
Our aim is to investigate the effect of the Mesenchymal stem cell (MSC) administration on the release of Mammalian Target of Rapamycin (mTOR) and Phosphorylated- mTOR(p-mTOR) in Cyclophosphomide (CTX) induced ovarian damage. Rats divided into three groups. The first group was categorized as the control(C group;n = 6), the second group as CTX-administered group (CTX group;n = 6), and the third group as CTX and MSC-administered group (CTX + SC group;n = 6). CTX was injected intraperitoneally at 50 mg/kg on the first day and at 8 mg/kg during the following 13 days. In Group 3, adipose-derived MSCs (5 × 104) were injected locally into the ovary. Both ovaries were removed at the end of the 8th week. The follicle count was made. The expression of mTOR and p-mTOR was analyzed immunohistochemically. The follicles in the ovary of Group C were observed in normal structures. Degeneration was evident in the CTX group. In the CTX + SC group, the degenerative appearance monitored in the CTX group vanished in most areas, and fibrosis was greatly reduced. The number of follicles in the CTX group was lower than that of both C and CTX + SC groups (p < 005). In the C group, mTOR showed strong positive staining while mTOR and p-mTOR expression was negative in all follicles in the CTX group. Both mTOR and p-mTOR revealed moderate positive expression in the CTX + SC group. MSC therapy rescued the damage ovarian function created by CTX, reducing follicle loss. MSCs were shown to inhibit the loss of mTOR and p-mTOR signaling, which is key to meiosis in oocytes.
Collapse
Affiliation(s)
- Nazlı Çil
- Department of Histology and Embryology, Faculty of Medicine, Pamukkale University, Denizli, Turkey.
| | - Gülçin Abban Mete
- Department of Histology and Embryology, Faculty of Medicine, Pamukkale University, Denizli, Turkey.
| |
Collapse
|
46
|
Labrune E, Jaeger P, Santamaria C, Fournier C, Benchaib M, Rabilloud M, Salle B, Lornage J. Cellular and Molecular Impact of Vitrification Versus Slow Freezing on Ovarian Tissue. Tissue Eng Part C Methods 2021; 26:276-285. [PMID: 32323623 DOI: 10.1089/ten.tec.2020.0063] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Objective: To evaluate a vitrification protocol from histology to gene expression to slow freezing. Methods: Ovaries from 12 prepubertal ewes. The same ovary was cut into fragments, studied fresh, frozen, and vitrified. Follicle morphology by hematoxylin-eosin-safran staining, vitality by Trypan Blue, and apoptosis by marking cleaved caspase-3 were studied. The expression of gene: anti-Müllerian hormone (AMH), cytochrome p450 family 11 subfamily A member 1 (CYP11A), and steroidogenic acute regulatory protein (STAR; granulosa cells); growth differentiation factor 9 (GDF9) and zona pellucida glycoprotein 3 (ZP3; oocytes); and cyclin D2 (CCND2) and cyclin-dependent kinase inhibitor 1A (CDKN1A; cell cycle regulation), was evaluated by reverse transcription quantitative polymerase chain reaction. Results: The slow freezing protocol had a significant negative impact on intact primordial follicles compared with fresh tissue (37.6% vs. 62.5%, p = 0.003). More intact follicles after vitrification were observed compared with slow freezing (p = 0.037). The apoptotic primordial follicles were similar after slow freezing and vitrification (12.6% vs. 13.9%). Concerning granulosa cell genes, slow freezing led to a trend toward overexpression of AMH messenger RNA (mRNA; p = 0.07); while vitrification led to a significant overexpression of CYP11A mRNA (p = 0.003), and a trend toward an overexpression of STAR mRNA (p = 0.06). Concerning oocyte genes, both techniques did not lead to a difference of GDF9 and ZP3 mRNA. Concerning cell cycle genes, slow freezing led to a significant underexpression of CCND2 (p = 0.04); while vitrification did not lead to a difference for CCND2 and CDKN1A mRNA. Conclusion: Vitrification preserved follicular morphology better than slow freezing and led to gene overexpressed, while slow freezing led to gene underexpressed. Impact statement The preservation of female fertility and in particular the cryopreservation of ovarian tissue (OT) is a major public health issue aimed at improving the quality of life of patients after gonadotoxic treatments. The use of slow freezing of this OT, which is the reference technique, is not optimal due to tissue alteration. The alternative would be vitrification. This study compares these two techniques. We have highlighted that vitrification preserved follicular morphology better than slow freezing and led to gene overexpressed, while slow freezing led to gene underexpressed.
Collapse
Affiliation(s)
- Elsa Labrune
- Service de Médecine de la Reproduction, Hôpital Mère Enfant, Hospices Civils de Lyon, Bron, France.,Faculté de Médecine Laennec, Université Claude Bernard, Lyon, France.,INSERM Unité 1208, Bron, France
| | - Pauline Jaeger
- Service de Médecine de la Reproduction, Hôpital Mère Enfant, Hospices Civils de Lyon, Bron, France.,Faculté de Médecine Laennec, Université Claude Bernard, Lyon, France.,INSERM Unité 1208, Bron, France
| | | | - Cyrielle Fournier
- Faculté de Médecine Laennec, Université Claude Bernard, Lyon, France.,INSERM Unité 1208, Bron, France
| | - Mehdi Benchaib
- Service de Médecine de la Reproduction, Hôpital Mère Enfant, Hospices Civils de Lyon, Bron, France.,Faculté de Médecine Laennec, Université Claude Bernard, Lyon, France.,INSERM Unité 1208, Bron, France
| | - Muriel Rabilloud
- Service de Biostatistique et Bioinformatique, Hospices Civils de Lyon, Lyon, France.,Laboratoire Biostatistique Santé, UMR 5558, Lyon, France
| | - Bruno Salle
- Service de Médecine de la Reproduction, Hôpital Mère Enfant, Hospices Civils de Lyon, Bron, France.,Faculté de Médecine Laennec, Université Claude Bernard, Lyon, France.,Faculté de Médecine Lyon Sud, Université Claude Bernard, Oullins, France
| | - Jacqueline Lornage
- Service de Médecine de la Reproduction, Hôpital Mère Enfant, Hospices Civils de Lyon, Bron, France.,Faculté de Médecine Laennec, Université Claude Bernard, Lyon, France.,INSERM Unité 1208, Bron, France
| |
Collapse
|
47
|
Nguyen TYT, Cacciottola L, Camboni A, Ravau J, De Vos M, Demeestere I, Donnez J, Dolmans MM. Ovarian tissue cryopreservation and transplantation in patients with central nervous system tumours. Hum Reprod 2021; 36:1296-1309. [PMID: 33394011 DOI: 10.1093/humrep/deaa353] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 11/24/2020] [Indexed: 11/12/2022] Open
Abstract
STUDY QUESTION Is there a possibility of reseeding cancer cells potentially present in frozen ovarian tissue from patients with central nervous system (CNS) tumours? SUMMARY ANSWER Malignancy reseeding in cryopreserved ovarian tissue from 20 patients with CNS tumours was not detected by histology, immunohistochemistry (IHC), molecular biology or xenotransplantation. WHAT IS KNOWN ALREADY Ovarian metastasis potential has been documented in patients with leukaemia, borderline ovarian tumours, advanced breast cancer and Ewing sarcoma. However, data on the safety of transplanting frozen-thawed ovarian tissue from cancer patients with CNS tumours are still lacking. STUDY DESIGN, SIZE, DURATION This prospective experimental study was conducted in an academic gynaecology research laboratory using cryopreserved ovarian cortex from 20 patients suffering from CNS tumours. Long-term (5 months) xenografting was performed in immunodeficient mice. PARTICIPANTS/MATERIALS, SETTING, METHODS Subjects enrolled in the study were suffering from one of six types of CNS tumours including medulloblastoma, ependymoma, primitive neuroectodermal tumours, astrocytoma, glioblastoma and germinoma. The presence of malignant cells was investigated with disease-specific markers for each patient in cryopreserved and xenografted ovarian tissue by histology, IHC via expression of neuron-specific enolase (NSE) and glial fibrillary acidic protein (GFAP), and reverse transcription droplet digital polymerase chain reaction (RT-ddPCR) for quantification of GFAP and ENO2 gene amplification. MAIN RESULTS AND THE ROLE OF CHANCE Serial sections of cryopreserved and xenografted ovarian tissue from 20 patients showed no malignant cells by histology. All samples were negative for NSE and GFAP, although these neural markers were expressed extensively in the patients' primary tumours. Analysis by RT-ddPCR revealed no cancer cells detected in cryopreserved and xenografted ovarian fragments from subjects with astrocytoma, ependymoma, glioblastoma or medulloblastoma. Taken together, the study found no evidence of malignancy seeding in frozen-thawed and xenotransplanted ovarian tissue from patients affected by CNS cancers. LIMITATIONS, REASONS FOR CAUTION This analysis cannot guarantee complete elimination of disseminated disease from all cryopreserved ovarian cortex, since we are unable to examine the fragments used for transplantation. WIDER IMPLICATIONS OF THE FINDINGS This is the first study to be conducted in patients with CNS cancers undergoing ovarian tissue cryopreservation and transplantation, and clearly demonstrates no tumour seeding in their frozen-thawed and xenografted tissue. This information is vital for doctors to provide patients with meaningful and accurate advice on the possibilities and risks of ovarian tissue reimplantation. STUDY FUNDING/COMPETING INTEREST(S) This study was supported by grants from the Fonds National de la Recherche Scientifique de Belgique-the Excellence of Science (FNRS-EOS), number 30443682 awarded to M.-M.D. and T.Y.T.N., FNRS grant number 5/4/150/5 and FNRS-PDR Convention grant number T.0077.14 awarded to M.-M.D., grant 2018-042 from the Foundation Against Cancer awarded to A.C., and private donations (Ferrero, de Spoelberch). The authors declare no competing financial interests. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Thu Yen Thi Nguyen
- Pôle de Gynécologie, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
| | - Luciana Cacciottola
- Pôle de Gynécologie, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
| | - Alessandra Camboni
- Pôle de Gynécologie, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium.,Service d'Anatomie Pathologique, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Joachim Ravau
- Laboratory of Pediatric Hepatology and Cell Therapy, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
| | - Michel De Vos
- Centre for Reproductive Medicine, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium.,Follicle Biology Laboratory (FOBI), UZ Brussel, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Isabelle Demeestere
- Research Laboratory in Human Reproduction, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Jacques Donnez
- Société de Recherche pour l'Infertilité (SRI), Brussels, Belgium
| | - Marie-Madeleine Dolmans
- Pôle de Gynécologie, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium.,Gynecology Department, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| |
Collapse
|
48
|
Human Mesenchymal Stem Cell Therapy and Other Novel Treatment Approaches for Premature Ovarian Insufficiency. Reprod Sci 2021; 28:1688-1696. [PMID: 33956339 PMCID: PMC8144118 DOI: 10.1007/s43032-021-00528-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 03/01/2021] [Indexed: 12/11/2022]
Abstract
Premature ovarian insufficiency (POI) is a condition characterized by amenorrhea, hypergonadotropic hypogonadism, estrogen deficiency, and reduced follicle counts leading to infertility under the age of 40. POI occurs in approximately 1-3% of women in the general population. Evaluation is warranted when the diagnosis of POI is made to rule out underlying etiologies, which could be multifactorial. This review serves to cover the novel treatment approaches reported in the literature.
Collapse
|
49
|
Chronopoulou E, Raperport C, Sfakianakis A, Srivastava G, Homburg R. Elective oocyte cryopreservation for age-related fertility decline. J Assist Reprod Genet 2021; 38:1177-1186. [PMID: 33608838 PMCID: PMC7894970 DOI: 10.1007/s10815-021-02072-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Accepted: 01/11/2021] [Indexed: 12/27/2022] Open
Abstract
PURPOSE Women who pursue fertility at an advanced age are increasingly common. Family planning and sexual education have traditionally focused on contraception and prevention of sexually transmitted diseases. A focus should now also be placed on fertility awareness and fertility preservation. This manuscript aims to give an update on the existing evidence around elective oocyte cryopreservation, also highlighting the need for fertility education and evidence-based, individualized counselling. METHODS A thorough electronic search was performed from the start of databases to March 2020 aiming to summarize the existing evidence around elective egg freezing, the logic behind its use, patient counselling and education, success rates and risks involved, regulation, cost-effectiveness, current status and future perspectives. RESULTS Clinician-led counselling regarding reproductive aging and fertility preservation is often overlooked. Elective oocyte cryopreservation is not a guarantee of live birth, and the answer regarding cost-effectiveness needs to be individualized. The existing studies on obstetric and perinatal outcomes following the use of egg freezing are, until now, reassuring. Constant monitoring of short-term and long-term outcomes, uniform regulation and evidence-based, individualized counselling is of paramount importance. CONCLUSIONS Elective oocyte cryopreservation is one of the most controversial aspects of the world of assisted reproduction, and a lot of questions remain unanswered. However, women today do have this option which was not available in the past. Elective oocyte cryopreservation for age-related fertility decline should be incorporated in women's reproductive options to ensure informed decisions and reproductive autonomy.
Collapse
Affiliation(s)
- E Chronopoulou
- Homerton Fertility Centre, Homerton University Hospital, Homerton Row, Clapton, London, E9 6SR, UK.
| | - C Raperport
- Homerton Fertility Centre, Homerton University Hospital, Homerton Row, Clapton, London, E9 6SR, UK
- Queen Mary University of London, Mile End Road, Bethnal Green, London, E1 4NS, UK
| | - A Sfakianakis
- Homerton Fertility Centre, Homerton University Hospital, Homerton Row, Clapton, London, E9 6SR, UK
| | - G Srivastava
- Homerton Fertility Centre, Homerton University Hospital, Homerton Row, Clapton, London, E9 6SR, UK
| | - R Homburg
- Homerton Fertility Centre, Homerton University Hospital, Homerton Row, Clapton, London, E9 6SR, UK
| |
Collapse
|
50
|
Transplantation of cryopreserved ovarian tissue in a series of 285 women: a review of five leading European centers. Fertil Steril 2021; 115:1102-1115. [DOI: 10.1016/j.fertnstert.2021.03.008] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 02/19/2021] [Accepted: 03/04/2021] [Indexed: 01/18/2023]
|