1
|
Mottram R, Feltbower RG, Jones G, Gelcich S, Rostron H, Allen M, Glaser AW. From Storage to Survivorship: A Scoping Review of Young Adult Cancer Survivors' Experiences and Preferences in Reproductive Survivorship Care After Fertility Tissue Preservation. J Pediatr Adolesc Gynecol 2024:S1083-3188(24)00273-0. [PMID: 39197581 DOI: 10.1016/j.jpag.2024.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 07/25/2024] [Accepted: 08/14/2024] [Indexed: 09/01/2024]
Abstract
Despite improved survival rates for childhood cancer, around 60% of survivors suffer lifelong health problems due to their treatment, including fertility issues which account for one third of these problems. Ovarian or testicular tissue cryopreservation can be offered to patients whose cancer treatment puts them at high risk of subsequent subfertility, but it presents unique challenges compared to standard methods of fertility preservation. We report the available information on the experiences of cancer survivors who preserved tissue for future fertility, to support the development of survivorship care informed by recipients' perspectives and experiences to identify future research priorities. We conducted a scoping review following the recommendations of the Joanna Briggs Institute and the Systematic Reviews and Meta-Analyses Extension for Scoping Review (PRISMA-ScR). From 1956 unique records, 5 met our inclusion criteria. No literature was found reporting on the experiences of people who stored testicular tissue. We found that young women who had stored ovarian tissue faced complex emotional and ethical dilemmas in reproductive decisions post cancer and strongly desired their own biological children. This scoping review is the first to report beyond clinical outcomes by focusing specifically on the self-reported outcomes of patients who preserved ovarian tissue in early life after a cancer diagnosis. Separate consideration of the needs of patients with stored fertility tissue is needed to enable personalized survivorship care. Patient-reported outcomes are also needed from individuals with stored testicular tissue, parents who consented to their child's tissue storage, and the healthcare professionals involved in their care.
Collapse
Affiliation(s)
- Rebecca Mottram
- School of Medicine, University of Leeds, Leeds, United Kingdom.
| | - Richard G Feltbower
- Leeds Institute for Data Analytics, School of Medicine, University of Leeds, Leeds, United Kingdom
| | | | - Sarah Gelcich
- School of Medicine, University of Leeds, Leeds, United Kingdom
| | - Heather Rostron
- Leeds Teaching Hospitals NHS Trust and School of Medicine, University of Leeds, Leeds, United Kingdom
| | - Majorie Allen
- Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
| | - Adam W Glaser
- Leeds Teaching Hospitals NHS Trust and School of Medicine, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
2
|
Rooda I, Hassan J, Hao J, Wagner M, Moussaud-Lamodière E, Jääger K, Otala M, Knuus K, Lindskog C, Papaikonomou K, Gidlöf S, Langenskiöld C, Vogt H, Frisk P, Malmros J, Tuuri T, Salumets A, Jahnukainen K, Velthut-Meikas A, Damdimopoulou P. In-depth analysis of transcriptomes in ovarian cortical follicles from children and adults reveals interfollicular heterogeneity. Nat Commun 2024; 15:6989. [PMID: 39168975 PMCID: PMC11339373 DOI: 10.1038/s41467-024-51185-0] [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: 11/16/2023] [Accepted: 08/01/2024] [Indexed: 08/23/2024] Open
Abstract
The ovarian cortical reserve of follicles is vital for fertility. Some medical treatments are toxic to follicles, leading to premature ovarian insufficiency. Ovarian tissue cryopreservation is an established method to preserve fertility in adults and even applied in prepuberty despite unproven efficacy. Here, we analyze transcriptomes of 120 cortical follicles from children and adults for detailed comparison. We discover heterogeneity with two main types of follicles in both age groups: one with expected oocyte-granulosa profiles and another with predicted role in signaling. Transcriptional changes during growth to the secondary stage are similar overall in children and adults, but variations related to extracellular matrix, theca cells, and miRNA profiles are found. Notably, cyclophosphamide dose correlates with interferon signaling in child follicles. Additionally, morphology alone is insufficient for follicle categorization suggesting a need for additional markers. Marker genes for early follicle activation are determined. These findings will help refine follicular classification and fertility preservation techniques across critical ages.
Collapse
Affiliation(s)
- Ilmatar Rooda
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Huddinge, Stockholm, Sweden.
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm, Sweden.
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia.
| | - Jasmin Hassan
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Huddinge, Stockholm, Sweden
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Jie Hao
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Huddinge, Stockholm, Sweden
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm, Sweden
- Department of Reproductive Medicine, Xiangya Hospital, Central South University, Changsha, PR China
| | - Magdalena Wagner
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Huddinge, Stockholm, Sweden
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Elisabeth Moussaud-Lamodière
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Huddinge, Stockholm, Sweden
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Kersti Jääger
- Institute of Computer Science, University of Tartu, Tartu, Estonia
- Competence Centre on Health Technologies, Tartu, Estonia
| | - Marjut Otala
- Department of Obstetrics and Gynecology, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Katri Knuus
- Department of Obstetrics and Gynecology, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Cecilia Lindskog
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Kiriaki Papaikonomou
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm, Sweden
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Sebastian Gidlöf
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Huddinge, Stockholm, Sweden
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm, Sweden
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Cecilia Langenskiöld
- Department of Pediatric Oncology, Queen Silvia Children's Hospital, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Hartmut Vogt
- Crown Princess Victoria Children's Hospital, and Division of Children's and Women's Health, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Per Frisk
- Department of Women's and Children's Health, Uppsala University Children's Hospital, Uppsala, Sweden
| | - Johan Malmros
- Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Timo Tuuri
- Department of Obstetrics and Gynecology, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Andres Salumets
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Huddinge, Stockholm, Sweden
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm, Sweden
- Competence Centre on Health Technologies, Tartu, Estonia
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
| | - Kirsi Jahnukainen
- Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Women's and Children's Health, NORDFERTIL Research Lab Stockholm, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Agne Velthut-Meikas
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| | - Pauliina Damdimopoulou
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Huddinge, Stockholm, Sweden.
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
3
|
Cui Y, Harteveld F, Ba Omar HAM, Yang Y, Bjarnason R, Romerius P, Sundin M, Norén Nyström U, Langenskiöld C, Vogt H, Henningsohn L, Frisk P, Vepsäläinen K, Petersen C, Mitchell RT, Guo J, Alves-Lopes JP, Jahnukainen K, Stukenborg JB. Prior exposure to alkylating agents negatively impacts testicular organoid formation in cells obtained from childhood cancer patients. Hum Reprod Open 2024; 2024:hoae049. [PMID: 39188568 PMCID: PMC11346771 DOI: 10.1093/hropen/hoae049] [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: 08/28/2023] [Revised: 07/15/2024] [Indexed: 08/28/2024] Open
Abstract
STUDY QUESTION Can human pre- and peri-pubertal testicular cells obtained from childhood cancer patients, previously treated with chemotherapy, form testicular organoids (TOs)? SUMMARY ANSWER Organoid formation from testicular tissue collected from childhood cancer patients positively correlates with SRY-Box transcription factor 9 (SOX9) expression in Sertoli cells, which in turn negatively correlates with previous exposure to alkylating chemotherapy. WHAT IS KNOWN ALREADY Pre- and peri-pubertal boys exposed to highly gonadotoxic therapies can only safeguard their fertility potential through testicular tissue cryopreservation. Today, there is no established clinical tool to restore fertility using these testicular samples. Organoids hold promise in providing fundamental early insights in creating such platforms. However, the generation of TOs that closely resemble the innate testis, to enable a thorough monitoring of the necessary steps for germ cell differentiation and somatic functionalities, remains a challenge. STUDY DESIGN SIZE DURATION We used a Matrigel-based three-layer gradient culture system to generate human TOs and to reveal whether chemotherapy exposure affects TO formation capacity and the functionality of pre- and peri-pubertal testicular somatic cells. Testicular cells of 11 boys (aged 7.7 ± 4.1 (mean ± SD) years) were assessed for TO formation in relation to previous chemotherapy exposure and SOX9 expression in histological sections of paraffin-embedded testicular tissue samples collected on the day of biopsy and compared with testicular tissue samples obtained from 28 consecutive patients (aged 6.9 ± 3.8 (mean ± SD) years). All 39 patients were part of the fertility preservation project NORDFERTIL; an additional 10 samples (from boys aged 5.5 ± 3.5 (mean ± SD) years, without an underlying pathology) in an internal biobank collection were used as controls. PARTICIPANTS/MATERIALS SETTING METHODS We obtained 49 testicular tissue samples from boys aged 0.8-13.4 years. Fresh samples (n = 11) were dissociated into single-cell suspensions and applied to a three-layer gradient culture system for organoid formation. Histological sections of another 28 samples obtained as part of the fertility preservation project NORDFERTIL, and 10 samples from a sample collection of a pathology biobank were used to evaluate the effects of prior exposure to alkylating agents on testicular samples. Testicular organoid formation was defined based on morphological features, such as compartmentalized structures showing cord formation, and protein expression of testicular cell-specific markers for germ and somatic cells was evaluated via immunohistochemical staining. Hormone secretion was analysed by specific enzyme-linked immunosorbent assays for testosterone and anti-Müllerian hormone (AMH) production. MAIN RESULTS AND THE ROLE OF CHANCE Our results revealed that 4 out of 11 prepubertal testicular samples formed TOs that showed compartmentalized cord-like structures surrounded by interstitial-like areas and increasing levels of both testosterone as well as AMH over a 7-day culture period. We observed that SOX9 expression was correlated positively with TO formation. Moreover, exposure to alkylating agents before biopsy was inversely correlated with SOX9 expression (P = 0.006). LARGE SCALE DATA N/A. LIMITATIONS REASONS FOR CAUTION Due to the limited amount of material available, only 11 out of the 39 pre- and peri-pubertal testicular tissue samples could be used for the organoid formation experiments. The testicular tissue samples obtained from a sample collection of the internal biobank of Department of Pathology, Karolinska University Hospital were considered normal and included in the study if no testicular pathology was reported. However, detailed information regarding previous medical treatments and/or testicular volumes of the patients included in this biobank was not available. WIDER IMPLICATIONS OF THE FINDINGS Our observations suggest that SOX9 expression may serve as a putative indicator of TO formation, indicating a critical role of Sertoli cells in promoting organoid formation, seminiferous tubule integrity, and testicular function in pre- and peri-pubertal testicular tissue. STUDY FUNDING/COMPETING INTERESTS This study was supported by grants from the Swedish Childhood Cancer Foundation (PR2019-0123; PR2022-0115; TJ2020-0023) (J.-B.S.), Finnish Cancer Society (K.J.), Finnish Foundation for Paediatric Research (K.J.), Swedish Research Council (2018-03094; 2021-02107) (J.-B.S.), and Birgitta and Carl-Axel Rydbeck's Research Grant for Paediatric Research (2020-00348; 2020-00335; 2021-00073; 2022-00317) (J.-B.S. and K.J.). Y.C. and Y.Y. received a scholarship from the Chinese Scholarship Council. J.P.A-L. was supported by a Starting Grant in Medicine and Health (2022-01467) from the Swedish Research Council. R.T.M. was supported by a UKRI Future Leaders Fellowship (MR/S017151/1). The MRC Centre for Reproductive Health was supported by an MRC Centre Grant (MR/N022556/1). The authors declare no competing interests.
Collapse
Affiliation(s)
- Yanhua Cui
- NORDFERTIL Research Lab Stockholm, Childhood Cancer Research Unit, Department of Women’s and Children’s Health, Karolinska Institutet, Karolinska University Hospital, Solna, Sweden
| | - Femke Harteveld
- NORDFERTIL Research Lab Stockholm, Childhood Cancer Research Unit, Department of Women’s and Children’s Health, Karolinska Institutet, Karolinska University Hospital, Solna, Sweden
| | - Hajar Ali Mohammed Ba Omar
- NORDFERTIL Research Lab Stockholm, Childhood Cancer Research Unit, Department of Women’s and Children’s Health, Karolinska Institutet, Karolinska University Hospital, Solna, Sweden
| | - Yifan Yang
- NORDFERTIL Research Lab Stockholm, Childhood Cancer Research Unit, Department of Women’s and Children’s Health, Karolinska Institutet, Karolinska University Hospital, Solna, Sweden
| | - Ragnar Bjarnason
- Children’s Medical Center, Landspítali University Hospital, Reykjavik, Iceland
- Department of Paediatrics, Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Patrik Romerius
- Department of Paediatric Oncology and Haematology, Clinical Sciences, Lund University, Barn-och Ungdomssjukhuset Lund, Skånes Universitetssjukhus, Lund, Sweden
| | - Mikael Sundin
- Division of Paediatrics, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Huddinge, Sweden
- Pediatric Blood Disorders, Immunodeficiency and Stem Cell Transplantation Unit, Astrid Lindgren Children’s Hospital, Karolinska University Hospital, Huddinge, Sweden
| | | | - Cecilia Langenskiöld
- Department of Paediatric Oncology, The Queen Silvia Children’s Hospital, Gothenburg, Sweden
| | - Hartmut Vogt
- Department of Biomedical and Clinical Science, H.R.H Crown Princess Victoria’s Children’s and Youth Hospital, Linköping University and University Hospital, Linköping, Sweden
| | - Lars Henningsohn
- Division of Urology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Huddinge, Sweden
| | - Per Frisk
- Pediatric Hematology & Oncology, Children’s University Hospital, Uppsala, Sweden
| | - Kaisa Vepsäläinen
- Department of Paediatrics, Kuopio University Hospital, Kuopio, Finland
| | - Cecilia Petersen
- NORDFERTIL Research Lab Stockholm, Childhood Cancer Research Unit, Department of Women’s and Children’s Health, Karolinska Institutet, Karolinska University Hospital, Solna, Sweden
| | - Rod T Mitchell
- MRC Centre for Reproductive Health, Institute for Regeneration and Repair, Edinburgh BioQuarter, The University of Edinburgh, Edinburgh, UK
- Royal Hospital for Children and Young People, Edinburgh, UK
| | - Jingtao Guo
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - João Pedro Alves-Lopes
- NORDFERTIL Research Lab Stockholm, Childhood Cancer Research Unit, Department of Women’s and Children’s Health, Karolinska Institutet, Karolinska University Hospital, Solna, Sweden
| | - Kirsi Jahnukainen
- NORDFERTIL Research Lab Stockholm, Childhood Cancer Research Unit, Department of Women’s and Children’s Health, Karolinska Institutet, Karolinska University Hospital, Solna, Sweden
- New Children’s Hospital, Paediatric Research Centre, Department of Paediatrics, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Jan-Bernd Stukenborg
- NORDFERTIL Research Lab Stockholm, Childhood Cancer Research Unit, Department of Women’s and Children’s Health, Karolinska Institutet, Karolinska University Hospital, Solna, Sweden
| |
Collapse
|
4
|
Ramirez T, Pavone M. Exploring the Frontiers of Ovarian Tissue Cryopreservation: A Review. J Clin Med 2024; 13:4513. [PMID: 39124779 PMCID: PMC11312752 DOI: 10.3390/jcm13154513] [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: 05/28/2024] [Revised: 07/15/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024] Open
Abstract
Objective: This paper serves as an up-to-date narrative review of the most effective methods and outcomes of ovarian tissue cryopreservation (OTC) with new data comparing this method to oocyte and embryo cryopreservation as well as its utility in restoration of endocrine function. Background: Data on OTC are becoming more available as more patients are achieving cancer remission and choosing to use their cryopreserved tissue to conceive or restore endocrine function. With OTC only recently becoming a non-experimental method of fertility preservation, it is important to evaluate, compare, and optimize current practices to improve live birth outcomes. Methods: A literature search of meta-analyses, systematic reviews, case series, retrospective studies, and randomized control trials was performed using the PubMed database with multiple search terms. Discussion: Current practices and outcomes of OTC remain heterogeneous, though they are becoming more streamlined with the emerging data on successful live births. Multiple aspects of OTC have been studied to optimize protocols, particularly methods of cryopreserving, in vitro maturation, and transplantation. In vitro follicle maturation is a novel application with emerging data on methods and outcomes. OTC is a versatile method not only for fertility preservation but also for hormone restoration as well. With wider usage of OTC, ethical dilemmas will need to be addressed. Conclusions: OTC can be used as fertility preservation for a variety of patients. Recent studies suggest it may be comparable to embryo cryopreservation, but with growing data on live births, comparative studies should continue to be performed. In vitro follicle maturation (IVFM) is a promising application of ovarian tissue harvesting. Data are lacking on cost-effectiveness, patient satisfaction, and morbidity associated with OTC.
Collapse
Affiliation(s)
- Tia Ramirez
- McGaw Medical Center, Northwestern University, Chicago, IL 60611, USA
| | - MaryEllen Pavone
- Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
5
|
Steinmann M, Rietschin A, Pagano F, Karrer T, Kollár A, Weidlinger S, von Wolff M. Systematic Review of the Gonadotoxicity and Risk of Infertility of Soft Tissue Sarcoma Chemotherapies in Pre- and Postpubertal Females and Males. J Adolesc Young Adult Oncol 2024. [PMID: 38995852 DOI: 10.1089/jayao.2024.0057] [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: 07/14/2024] Open
Abstract
Increasing awareness of gonadotoxicity in cancer treatments and infertility risk is essential for counseling young cancer patients. While fertility preservation options are available in many countries, limited data on gonadotoxicity hinder recommendations, especially for soft tissue cancers. This review, part of the FertiTOX project (www.fertitox.com), organized by FertiPROTEKT (www.fertiprotekt.com), aims to address this knowledge gap to improve fertility preservation guidance. We performed a systematic literature search on gonadotoxicity in soft tissue sarcoma (STS) cancer treatments. Only patients without metastases or recurrent disease were considered. "Suspected infertility" was defined based on low ovarian reserve parameters, low inhibin B levels, high gonadotropin concentration, gonadal dysfunction, amenorrhea, oligomenorrhea, azoospermia, or oligozoospermia due to limited infertility data. The study quality was assessed using the Newcastle-Ottawa Scale. The search yielded 3309 abstracts, with 138 undergoing full-text analysis. Eight studies on STS were included. Suspected infertility was observed in 20 of 28 females (71.4%, range 0-100%) and 38 of 63 males (60.3%, range 34.8-100%) with STS. Six of the eight studies received high-quality scores on the NOS, while two received a fair score. Our data suggest a high risk of infertility from chemotherapy in pre- and postpubertal STS survivors. This underscores the importance of considering fertility preservation measures when counseling these patients.
Collapse
Affiliation(s)
- Marcel Steinmann
- Division of Gynecological Endocrinology and Reproductive Medicine, Wome's Hospital, Lucerne Cantonal Hospital, Lucerne, Switzerland
| | - Anita Rietschin
- Division of Gynecological Endocrinology and Reproductive Medicine, University Womeńs Hospital, Inselspital Bern, University of Bern, Bern, Switzerland
| | - Flavia Pagano
- Division of Gynecological Endocrinology and Reproductive Medicine, University Womeńs Hospital, Inselspital Bern, University of Bern, Bern, Switzerland
| | - Tanya Karrer
- Medical Library, University Library Bern, University of Bern, Bern, Switzerland
| | - Attila Kollár
- Department of Medical Oncology, Inselspital Bern, University of Bern, Bern, Switzerland
| | - Susanna Weidlinger
- Division of Gynecological Endocrinology and Reproductive Medicine, University Womeńs Hospital, Inselspital Bern, University of Bern, Bern, Switzerland
| | - Michael von Wolff
- Division of Gynecological Endocrinology and Reproductive Medicine, University Womeńs Hospital, Inselspital Bern, University of Bern, Bern, Switzerland
| |
Collapse
|
6
|
Lau LS, Allingham C, Anazodo A, Sullivan M, Peate M, Hunter S, Stern C, Ryan AL, Super L, Orme LM, McCarthy M, Gook D, Lewin J, Marino J, Ryan J, Downie P, Manudhane R, Winstanley M, Pettit T, Lantsberg D, Irving H, Reid S, Heath JA, Kabalan-Baeza P, Wanaguru D, Moore L, Gillam L, Zacharin M, Assis M, Rozen G, Hunter T, Julania S, Sharwood E, Ameratunga D, Jayasinghe Y. The Australian New Zealand Consortium in Children, Adolescents, and Young Adults Oncofertility action plan. Pediatr Blood Cancer 2024; 71:e31041. [PMID: 38715224 DOI: 10.1002/pbc.31041] [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: 10/12/2023] [Revised: 03/25/2024] [Accepted: 04/12/2024] [Indexed: 05/24/2024]
Abstract
International and national oncofertility networks, including the US-led Oncofertility Consortium, FertiProtekt, and the Danish Network, have played pivotal roles in advancing the discipline of oncofertility over the last decade. Many other countries lack a shared approach to pediatric oncofertility health service delivery. This study aims to describe baseline oncofertility practices at Australian New Zealand Children's Haematology/Oncology Group centers in 2019-2021, describe binational priorities for care, and propose a 5-year action plan for best practice to be implemented by the newly formed Australian New Zealand Consortium in Children, Adolescents, and Young Adults (CAYA) Oncofertility (ANZCO).
Collapse
Affiliation(s)
- Lei Shong Lau
- Department of Obstetrics, Gynaecology and Newborn Health, Royal Women's Hospital, University of Melbourne, Parkville, Melbourne, Victoria, Australia
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Catherine Allingham
- Department of Obstetrics, Gynaecology and Newborn Health, Royal Women's Hospital, University of Melbourne, Parkville, Melbourne, Victoria, Australia
| | - Antoinette Anazodo
- Kids Cancer Centre, Sydney Children's Hospital, Sydney, New South Wales, Australia
- School of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Michael Sullivan
- Children's Cancer Centre, Royal Children's Hospital, Melbourne, Victoria, Australia
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia
- Department of Pediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Michelle Peate
- Department of Obstetrics, Gynaecology and Newborn Health, Royal Women's Hospital, University of Melbourne, Parkville, Melbourne, Victoria, Australia
| | - Sarah Hunter
- Starship Blood and Cancer Centre, Starship Hospital Te Whatu Ora-Health New Zealand, Auckland, New Zealand
| | - Catharyn Stern
- Department of Reproductive Services, Royal Women's Hospital, Melbourne, Victoria, Australia
| | - Anne Louise Ryan
- Department of Oncology and Haematology, Perth Children's Hospital, Perth, Western Australia, Australia
- Telethon Kids Institute, University of Western Australia, Perth, Western Australia, Australia
| | - Leanne Super
- Children's Cancer Centre, Royal Children's Hospital, Melbourne, Victoria, Australia
- Children's Cancer Centre, Monash Children's Hospital, Melbourne, Victoria, Australia
- Monash University, Melbourne, Victoria, Australia
| | - Lisa M Orme
- Children's Cancer Centre, Royal Children's Hospital, Melbourne, Victoria, Australia
- Department of Pediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Maria McCarthy
- Children's Cancer Centre, Royal Children's Hospital, Melbourne, Victoria, Australia
- Department of Pediatrics, University of Melbourne, Melbourne, Victoria, Australia
- Clinical Sciences, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Debra Gook
- Department of Obstetrics, Gynaecology and Newborn Health, Royal Women's Hospital, University of Melbourne, Parkville, Melbourne, Victoria, Australia
- Department of Reproductive Services, Royal Women's Hospital, Melbourne, Victoria, Australia
| | - Jeremy Lewin
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Victorian Adolescent & Young Adult Cancer Service, Melbourne, Victoria, Australia
| | - Jennifer Marino
- Department of Obstetrics, Gynaecology and Newborn Health, Royal Women's Hospital, University of Melbourne, Parkville, Melbourne, Victoria, Australia
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia
- Department of Pediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Jessica Ryan
- Cancer Centre for Children, The Children's Hospital at Westmead, Sydney, New South Wales, Australia
| | - Peter Downie
- Children's Cancer Centre, Royal Children's Hospital, Melbourne, Victoria, Australia
- Children's Cancer Centre, Monash Children's Hospital, Melbourne, Victoria, Australia
- Monash University, Melbourne, Victoria, Australia
| | - Rebecca Manudhane
- Michael Rice Centre for Haematology and Oncology, Women's and Children's Hospital, Adelaide, South Australia, Australia
| | - Mark Winstanley
- Starship Blood and Cancer Centre, Starship Hospital Te Whatu Ora-Health New Zealand, Auckland, New Zealand
| | - Tristan Pettit
- Children's Haematology Oncology Centre, Waipapa Hospital, Christchurch, New Zealand
| | - Daniel Lantsberg
- Department of Obstetrics, Gynaecology and Newborn Health, Royal Women's Hospital, University of Melbourne, Parkville, Melbourne, Victoria, Australia
- Department of Reproductive Services, Royal Women's Hospital, Melbourne, Victoria, Australia
- Melbourne IVF, Melbourne, Victoria, Australia
| | - Helen Irving
- Oncology Services Group, Children's Health Queensland Hospital and Health Service, University of Queensland, Brisbane, Queensland, Australia
| | - Sally Reid
- University of Adelaide, Adelaide, South Australia, Australia
| | - John A Heath
- School of Medicine, University of Tasmania, Hobart, Tasmania, Australia
- Department of Pediatrics, Royal Hobart Hospital, Hobart, Tasmania, Australia
| | - Paola Kabalan-Baeza
- Hunter and Northern New South Wales Youth Cancer Service, John Hunter Children's Hospital, New Lambton Heights, New South Wales, Australia
| | - Dylan Wanaguru
- School of Medicine, University of New South Wales, Sydney, New South Wales, Australia
- Department of Pediatric Surgery, Sydney Children's Hospital, Sydney, New South Wales, Australia
| | - Lyndal Moore
- Hunter and Northern New South Wales Youth Cancer Service, Calvary Mater Hospital, Waratah, New South Wales, Australia
| | - Lynn Gillam
- Department of Pediatrics, University of Melbourne, Melbourne, Victoria, Australia
- Children's Bioethics Centre, Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Margaret Zacharin
- Department of Endocrinology & Diabetes, The Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Michael Assis
- Department of Obstetrics, Gynaecology and Newborn Health, Royal Women's Hospital, University of Melbourne, Parkville, Melbourne, Victoria, Australia
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia
- Oncofertility Service, Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Genia Rozen
- Department of Reproductive Services, Royal Women's Hospital, Melbourne, Victoria, Australia
| | - Tamara Hunter
- Perth Children's Hospital, Perth, Western Australia, Australia
| | - Shital Julania
- Department of Oncology and Haematology, Perth Children's Hospital, Perth, Western Australia, Australia
| | - Erin Sharwood
- Department of Endocrinology and Diabetes, Queensland Children's Hospital, Brisbane, Queensland, Australia
- Faculty of Medicine, University of Queensland, Herston, Queensland, Australia
| | - Devini Ameratunga
- Gynaecology and Women's Service, Royal Brisbane and Women's Hospital, Herston, Queensland, Australia
| | - Yasmin Jayasinghe
- Department of Obstetrics, Gynaecology and Newborn Health, Royal Women's Hospital, University of Melbourne, Parkville, Melbourne, Victoria, Australia
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia
- Oncofertility Service, Royal Children's Hospital, Melbourne, Victoria, Australia
| |
Collapse
|
7
|
Sriram S, Macedo T, Mavinkurve‐Groothuis A, van de Wetering M, Looijenga LHJ. Alkylating agents-induced gonadotoxicity in prepubertal males: Insights on the clinical and preclinical front. Clin Transl Sci 2024; 17:e13866. [PMID: 38965809 PMCID: PMC11224131 DOI: 10.1111/cts.13866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/26/2024] [Accepted: 06/06/2024] [Indexed: 07/06/2024] Open
Abstract
Rising cure rates in pediatric cancer patients warrants an increased attention toward the long-term consequences of the diagnosis and treatment in survivors. Chemotherapeutic agents can be gonadotoxic, rendering them at risk for infertility post-survival. While semen cryopreservation is an option that can be provided for most (post)pubertal boys before treatment, this is unfortunately not an option prepubertal in age, simply due to the lack of spermatogenesis. Over the last couple of years, studies have thus focused on better understanding the testis niche in response to various chemotherapeutic agents that are commonly administered and their direct and indirect impact on the germ cell populations. These are generally compounds that have a high risk of infertility and have been classified into risk categories in curated fertility guidelines. However, with it comes the lack of evidence and the challenge of using informative models and conditions most reflective of the physiological scenario, in short, the appropriate study designs for clinically relevant outcomes. Besides, the exact mechanism(s) of action for many of these "risk" compounds as well as other agents is unclear. Understanding their behavior and effect on the testis niche will pave the way for incorporating new strategies to ultimately combat infertility. Of the various drug classes, alkylating agents pose the highest risk of gonadotoxicity as per previously established studies as well as risk stratification guidelines. Therefore, this review will summarize the findings in the field of male fertility concerning gonadotoxicity of akylating agents as a result of chemotherapy exposure.
Collapse
Affiliation(s)
- Sruthi Sriram
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
| | - Tiago Macedo
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
| | | | | | | |
Collapse
|
8
|
Pasten González A, Salvador Alarcón C, Mora J, Martín Gimenez MP, Carrasco Torrents R, Krauel L. Current Status of Fertility Preservation in Pediatric Oncology Patients. CHILDREN (BASEL, SWITZERLAND) 2024; 11:537. [PMID: 38790532 PMCID: PMC11120648 DOI: 10.3390/children11050537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 04/25/2024] [Accepted: 04/29/2024] [Indexed: 05/26/2024]
Abstract
Cancer poses significant emotional challenges for children and adolescents, despite improvements in survival rates due to new therapies. However, there is growing concern about the long-term effects, including fertility issues. This review examines recent advancements and future directions in fertility preservation within a pediatric population subjected to oncological therapies. Worldwide, there is variability in the availability of fertility preservation methods, influenced by factors like development status and governmental support. The decision to pursue preservation depends on the risk of gonadotoxicity, alongside factors such as diagnosis, treatment, clinical status, and prognosis. Currently, options for preserving fertility in prepubertal boys are limited compared to girls, who increasingly have access to ovarian tissue preservation. Adolescents and adults have more options available, but ethical considerations remain complex and diverse.
Collapse
Affiliation(s)
- Albert Pasten González
- Pediatric Surgical Oncology Unit, Department of Pediatric Surgery, SJD Barcelona Children’s Hospital, Universitat de Barcelona, 08950 Barcelona, Spain; (A.P.G.); (M.P.M.G.); (R.C.T.)
- Pediatric Cancer Center Barcelona, SJD Barcelona Children’s Hospital, Universitat de Barcelona, 08950 Barcelona, Spain;
| | - Cristina Salvador Alarcón
- Department of Obstetrics and Gynecology, SJD Barcelona Children’s Hospital, Universitat de Barcelona, 08950 Barcelona, Spain;
| | - Jaume Mora
- Pediatric Cancer Center Barcelona, SJD Barcelona Children’s Hospital, Universitat de Barcelona, 08950 Barcelona, Spain;
| | - Marta P. Martín Gimenez
- Pediatric Surgical Oncology Unit, Department of Pediatric Surgery, SJD Barcelona Children’s Hospital, Universitat de Barcelona, 08950 Barcelona, Spain; (A.P.G.); (M.P.M.G.); (R.C.T.)
- Pediatric Cancer Center Barcelona, SJD Barcelona Children’s Hospital, Universitat de Barcelona, 08950 Barcelona, Spain;
| | - Rosalia Carrasco Torrents
- Pediatric Surgical Oncology Unit, Department of Pediatric Surgery, SJD Barcelona Children’s Hospital, Universitat de Barcelona, 08950 Barcelona, Spain; (A.P.G.); (M.P.M.G.); (R.C.T.)
- Pediatric Cancer Center Barcelona, SJD Barcelona Children’s Hospital, Universitat de Barcelona, 08950 Barcelona, Spain;
| | - Lucas Krauel
- Pediatric Surgical Oncology Unit, Department of Pediatric Surgery, SJD Barcelona Children’s Hospital, Universitat de Barcelona, 08950 Barcelona, Spain; (A.P.G.); (M.P.M.G.); (R.C.T.)
- Pediatric Cancer Center Barcelona, SJD Barcelona Children’s Hospital, Universitat de Barcelona, 08950 Barcelona, Spain;
| |
Collapse
|
9
|
Ying H, Shi L, Zhang S. Research progress on mechanism of follicle injury after ovarian tissue transplantation and protective strategies. Zhejiang Da Xue Xue Bao Yi Xue Ban 2024; 53:321-330. [PMID: 38562041 PMCID: PMC11348700 DOI: 10.3724/zdxbyxb-2023-0566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 03/29/2024] [Indexed: 04/04/2024]
Abstract
Ovarian tissue cryopreservation and transplantation is the only way to preserve fertility for female cancer patients in prepubertal ages and those who cannot delay radiotherapy or chemotherapy. However, the success rate of cryopreservation and transplantation of ovarian tissue is still low at present due to the risk of ischemia and hypoxia of the grafted tissues. Abnormal activation of primordial follicles and ischemia-reperfusion injury after blood supply recovery also cause massive loss of follicles in grafted ovarian tissues. Various studies have explored the use of different drugs to reduce the damage of follicles during freezing and transplantation as well as to extend the duration of endocrine and reproductive function in patients with ovarian transplantation. For example, melatonin, N-acetylcysteine, erythropoietin or other antioxidants have been used to reduce oxidative stress; mesenchymal stem cells derived from different tissues, basic fibroblast growth factor, vascular endothelial growth factor, angiopoietin 2 and gonadotropin have been used to promote revascularization; anti-Müllerian hormone and rapamycin have been used to reduce abnormal activation of primordial follicles. This article reviews the research progress on the main mechanisms of follicle loss after ovarian tissue transplantation, including hypoxia, ischemia-reperfusion injury and associated cell death, and abnormal activation of follicles. The methods for reducing follicle loss in grafted ovarian tissues are further explored to provide a reference for improving the efficiency of ovarian tissue cryopreservation and transplantation.
Collapse
Affiliation(s)
- Hanqi Ying
- Reproductive Medicine Center, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China.
| | - Libing Shi
- Reproductive Medicine Center, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Songying Zhang
- Reproductive Medicine Center, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China.
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou 310016, China.
| |
Collapse
|
10
|
Affdal AO, Salama M, Ravitsky V. Ethical, legal, social, and policy issues of ovarian tissue cryopreservation in prepubertal girls: a critical interpretive review. J Assist Reprod Genet 2024; 41:999-1026. [PMID: 38430324 PMCID: PMC11052756 DOI: 10.1007/s10815-024-03059-z] [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: 08/29/2023] [Accepted: 02/07/2024] [Indexed: 03/03/2024] Open
Abstract
PURPOSE Despite the increasing number of childhood cancer survivors, significant advances in ovarian tissue cryopreservation (OTC) technique and medical societies' recommendations, fertility preservation (FP) and FP discussions are not always offered as a standard of care in the pediatric context. The aim of this literature review is to understand what ethical, legal, social, and policy issues may influence the provision of FP by OTC in prepubertal girls with cancer. METHODS A critical interpretive review of peer-reviewed papers published between 2000 and January 2023 was conducted, guided by the McDougall's version of the critical interpretive synthesis (Dixon-Woods), to capture recurring concepts, principles, and arguments regarding FP by OTC for prepubertal girls. RESULTS Of 931 potentially relevant papers, 162 were included in our analysis. Data were grouped into seven thematic categories: (1) risks of the procedure, (2) unique decision-making issues in pediatric oncofertility, (3) counseling, (4) cultural and cost issues, and (5) disposition of cryopreserved reproductive tissue. CONCLUSION This first literature review focusing on ethical, legal, social, and policy issues surrounding OTC in prepubertal girls highlights concerns in the oncofertility debate. Although OTC is no longer experimental as of December 2019, these issues could limit its availability and the child's future reproductive autonomy. This review concludes that specific actions must be provided to enable the offer of FP, such as supporting families' decision-making in this unique and complex context, and providing pediatric patients universal and full access to free or highly subsidized OTC.
Collapse
Affiliation(s)
| | | | - Vardit Ravitsky
- University of Montreal, Montreal, Canada
- The Hastings Center, Garrison, USA
- Harvard Medical School, Boston, USA
| |
Collapse
|
11
|
De Windt S, Kourta D, Kanbar M, Wyns C. Optimized Recovery of Immature Germ Cells after Prepubertal Testicular Tissue Digestion and Multi-Step Differential Plating: A Step towards Fertility Restoration with Cancer-Cell-Contaminated Tissue. Int J Mol Sci 2023; 25:521. [PMID: 38203691 PMCID: PMC10779385 DOI: 10.3390/ijms25010521] [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: 10/25/2023] [Revised: 12/19/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024] Open
Abstract
Undifferentiated germ cells, including the spermatogonial stem cell subpopulation required for fertility restoration using human immature testicular tissue (ITT), are difficult to recover as they do not easily adhere to plastics. Due to the scarcity of human ITT for research, we used neonatal porcine ITT. Strategies for maximizing germ cell recovery, including a comparison of two enzymatic digestion protocols (P1 and P2) of ITT fragment sizes (4 mm3 and 8 mm3) and multi-step differential plating were explored. Cellular viability and yield, as well as numbers and proportions of DDX4+ germ cells, were assessed before incubating the cell suspensions overnight on uncoated plastics. Adherent cells were processed for immunocytochemistry (ICC) and floating cells were further incubated for three days on Poly-D-Lysine-coated plastics. Germ cell yield and cell types using ICC for SOX9, DDX4, ACTA2 and CYP19A1 were assessed at each step of the multi-step differential plating. Directly after digestion, cell suspensions contained >92% viable cells and 4.51% DDX4+ germ cells. Pooled results for fragment sizes revealed that the majority of DDX4+ cells adhere to uncoated plastics (P1; 82.36% vs. P2; 58.24%). Further incubation on Poly-D-Lysine-coated plastics increased germ cell recovery (4.80 ± 11.32 vs. 1.90 ± 2.07 DDX4+ germ cells/mm2, respectively for P1 and P2). The total proportion of DDX4+ germ cells after the complete multi-step differential plating was 3.12%. These results highlight a reduced proportion and number of germ cells lost when compared to data reported with other methods, suggesting that multi-step differential plating should be considered for optimization of immature germ cell recovery. While Poly-D-Lysine-coating increased the proportions of recovered germ cells by 16.18% (P1) and 28.98% (P2), future studies should now focus on less cell stress-inducing enzymatic digestion protocols to maximize the chances of fertility restoration with low amounts of cryo-banked human ITT.
Collapse
Affiliation(s)
- Sven De Windt
- Laboratoire d’andrologie, Pôle de Recherche en Physiologie de la Reproduction, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, 1200 Brussels, Belgium; (S.D.W.); (D.K.); (M.K.)
| | - Dhoha Kourta
- Laboratoire d’andrologie, Pôle de Recherche en Physiologie de la Reproduction, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, 1200 Brussels, Belgium; (S.D.W.); (D.K.); (M.K.)
- Department of Gynecology-Andrology, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Marc Kanbar
- Laboratoire d’andrologie, Pôle de Recherche en Physiologie de la Reproduction, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, 1200 Brussels, Belgium; (S.D.W.); (D.K.); (M.K.)
- Department of Gynecology-Andrology, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Christine Wyns
- Laboratoire d’andrologie, Pôle de Recherche en Physiologie de la Reproduction, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, 1200 Brussels, Belgium; (S.D.W.); (D.K.); (M.K.)
- Department of Gynecology-Andrology, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| |
Collapse
|
12
|
Albamonte MI, Vitullo AD. Preservation of fertility in female and male prepubertal patients diagnosed with cancer. J Assist Reprod Genet 2023; 40:2755-2767. [PMID: 37770817 PMCID: PMC10656407 DOI: 10.1007/s10815-023-02945-2] [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: 07/04/2023] [Accepted: 09/12/2023] [Indexed: 09/30/2023] Open
Abstract
Over the past two decades, the importance of fertility preservation has grown not only in the realm of medical and clinical patient care, but also in the field of basic and applied research in human reproduction. With advancements in cancer treatments resulting in higher rates of patient survival, it is crucial to consider the quality of life post-cure. Therefore, fertility preservation must be taken into account prior to antitumor treatments, as it can significantly impact a patient's future fertility. For postpubertal patients, gamete cryopreservation is the most commonly employed preservation strategy. However, for prepubertal patients, the situation is more intricate. Presently, ovarian tissue cryopreservation is the standard practice for prepubertal girls, but further scientific evidence is required in several aspects. Testicular tissue cryopreservation, on the other hand, is still experimental for prepubertal boys. The primary aim of this review is to address the strategies available for possible fertility preservation in prepubertal girls and boys, such as ovarian cryopreservation/transplantation, in vitro follicle culture and meiotic maturation, artificial ovary, transplantation of cryopreserved spermatogonia, and cryopreservation/grafting of immature testicular tissue and testicular organoids.
Collapse
Affiliation(s)
- María Itatí Albamonte
- Centro de Estudios Biomédicos Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Hidalgo 775, C1405BCK, Buenos Aires, Argentina
| | - Alfredo D Vitullo
- Centro de Estudios Biomédicos Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Hidalgo 775, C1405BCK, Buenos Aires, Argentina.
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.
| |
Collapse
|
13
|
Romualdez-Tan MV. Modelling in vitro gametogenesis using induced pluripotent stem cells: a review. CELL REGENERATION (LONDON, ENGLAND) 2023; 12:33. [PMID: 37843621 PMCID: PMC10579208 DOI: 10.1186/s13619-023-00176-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 08/28/2023] [Indexed: 10/17/2023]
Abstract
In vitro gametogenesis (IVG) has been a topic of great interest in recent years not only because it allows for further exploration of mechanisms of germ cell development, but also because of its prospect for innovative medical applications especially for the treatment of infertility. Elucidation of the mechanisms underlying gamete development in vivo has inspired scientists to attempt to recapitulate the entire process of gametogenesis in vitro. While earlier studies have established IVG methods largely using pluripotent stem cells of embryonic origin, the scarcity of sources for these cells and the ethical issues involved in their use are serious limitations to the progress of IVG research especially in humans. However, with the emergence of induced pluripotent stem cells (iPSCs) due to the revolutionary discovery of dedifferentiation and reprogramming factors, IVG research has progressed remarkably in the last decade. This paper extensively reviews developments in IVG using iPSCs. First, the paper presents key concepts from groundwork studies on IVG including earlier researches demonstrating that IVG methods using embryonic stem cells (ESCs) also apply when using iPSCs. Techniques for the derivation of iPSCs are briefly discussed, highlighting the importance of generating transgene-free iPSCs with a high capacity for germline transmission to improve efficacy when used for IVG. The main part of the paper discusses recent advances in IVG research using iPSCs in various stages of gametogenesis. In addition, current clinical applications of IVG are presented, and potential future applications are discussed. Although IVG is still faced with many challenges in terms of technical issues, as well as efficacy and safety, novel IVG methodologies are emerging, and IVG using iPSCs may usher in the next era of reproductive medicine sooner than expected. This raises both ethical and social concerns and calls for the scientific community to cautiously develop IVG technology to ensure it is not only efficacious but also safe and adheres to social and ethical norms.
Collapse
Affiliation(s)
- Maria Victoria Romualdez-Tan
- Present Address: Repro Optima Center for Reproductive Health, Inc., Ground Floor JRDC Bldg. Osmena Blvd. Capitol Site, Cebu City, 6000, Philippines.
- Cebu Doctors University Hospital, Cebu City, Philippines.
| |
Collapse
|
14
|
Chabut M, Schneider P, Courbiere B, Saultier P, Bertrand Y, Tabone MD, Pochon C, Ducassou S, Paillard C, Gandemer V, Kanold J, Dalle JH, Poiree M, Plat G, Thouvenin S, Plantaz D, Sirvent N, Weinhard S, Berbis J, Baruchel A, Leverger G, Hamidou Z, Auquier P, Michel G. Ovarian Function and Spontaneous Pregnancy After Hematopoietic Stem Cell Transplantation for Leukemia Before Puberty: An L.E.A. Cohort Study. Transplant Cell Ther 2023:S2666-6367(23)01130-2. [PMID: 36849077 DOI: 10.1016/j.jtct.2023.02.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 02/17/2023] [Accepted: 02/19/2023] [Indexed: 02/27/2023]
Abstract
Ovarian function impairment and infertility are among the most frequent late effects after hematopoietic stem cell transplantation (HSCT). The aim of this study was to evaluate ovarian function, occurrence of premature ovarian insufficiency (POI), and spontaneous pregnancy in a large cohort of adult survivor women who had undergone HSCT for leukemia before puberty. We conducted a retrospective observational study in women from the national cohort L.E.A., the long-term French follow-up program after childhood leukemia. The median follow-up duration was 18 years (14.2-23.3) after HSCT. Among 178 women, 106 (60%) needed pubertal induction with hormone substitution treatment, whereas 72 (40%) had spontaneous menarche. After spontaneous menarche, 33 (46%) developed POI, mostly within 5 years of HSCT. Older age at time of HSCT and cryopreservation of ovarian tissue appeared as significant risk factors for POI. More than 65% of patients who underwent HSCT before the age of 4.8 years had spontaneous menarche, and almost 50% didn't have POI at last evaluation, whereas more than 85% with HSCT after the age of 10.9 years didn't have spontaneous menarche and needed induction of puberty with hormone replacement therapy. Twenty-two women (12%) had at least one spontaneous pregnancy, with 17 live-births, 14 miscarriages, 4 legal abortions, and 2 therapeutic abortions. These results add supplementary data to better counsel patients and their families on the chances of ovarian residual function and pregnancy after HSCT, as well as on the potential interest of fertility preservation.
Collapse
Affiliation(s)
- Mathilde Chabut
- Department of Pediatrics, University Hospital of Rouen, Rouen, France.
| | - Pascale Schneider
- Department of Pediatric Hematology and Oncology, University Hospital of Rouen, Rouen, France
| | - Blandine Courbiere
- Department of Gynecology-Obstetric and Reproductive Medicine, AP-HM, Hôpital La Conception/Aix-Marseille Université, IMBE, CNRS, IRD, Avignon Université, Marseille, France
| | - Paul Saultier
- Department of Pediatric Hematology, Immunology and Oncology, APHM, La Timone Children's Hospital, Marseille, France; Aix Marseille Univ, INSERM, INRAe, C2VN, Marseille, France
| | - Yves Bertrand
- Department of Pediatric Hematology and Oncology, University Hospital of Lyon, Lyon, France
| | - Marie-Dominique Tabone
- Department of Pediatric Hematology and Oncology, Armand Trousseau Hospital, AP-HP, Sorbonne University, Paris, France
| | - Cécile Pochon
- Department of Pediatric Hematology and Oncology, Children's Hospital of Brabois, Brabois,France
| | - Stéphane Ducassou
- Department of Pediatric Hematology and Oncology, University Hospital of Bordeaux, Bordeaux, France
| | - Catherine Paillard
- Department of Pediatric Hematology-Oncology, University Hospital, Strasbourg, France
| | - Virginie Gandemer
- Department of Pediatric Hematology and Oncology, University Hospital of Rennes, Rennes, France
| | - Justyna Kanold
- Department of Pediatric Hematology and Oncology, University Hospital of Clermont-Ferrand, Clermont-Ferrand, France
| | - Jean-Hugues Dalle
- Department of Pediatric Onco-Hematology, University Hospital of Saint Louis, Paris, France; Department of Hematology and immunology, Hôpital Robert-Debré, GHU APHP-Nord Université de Paris-Cité, Paris, France
| | - Maryline Poiree
- Department of Pediatric Hematology and Oncology, University Hospital L'Archet, Nice, France
| | - Geneviève Plat
- Department of Pediatric Hematology and Oncology, University Hospital of Toulouse, Toulouse, France
| | - Sandrine Thouvenin
- Department of Pediatric Onco-hematology, CHU de Saint-Etienne, Saint-Etienne, France
| | - Dominique Plantaz
- Department of Pediatric Hematology and Oncology, University Hospital of Grenoble, Grenoble, France
| | - Nicolas Sirvent
- Department of Pediatric Hematology and Oncology, University Hospital of Montpellier, Montpellier, France
| | - Sara Weinhard
- Department of Pediatric Hematology and Oncology, Children's Hospital of Brabois, Brabois,France
| | - Julie Berbis
- CEReS Research Unit EA 3279 and Department of Public Health, Hôpital de la Timone, Marseille, France
| | - André Baruchel
- Department of Hematology and immunology, Hôpital Robert-Debré, GHU APHP-Nord Université de Paris-Cité, Paris, France
| | - Guy Leverger
- Department of Pediatric Hematology and Oncology, Armand Trousseau Hospital, AP-HP, Sorbonne University, Paris, France
| | - Zeinab Hamidou
- CEReS Research Unit EA 3279 and Department of Public Health, Hôpital de la Timone, Marseille, France
| | - Pascal Auquier
- CEReS Research Unit EA 3279 and Department of Public Health, Hôpital de la Timone, Marseille, France
| | - Gérard Michel
- Department of Pediatric Hematology, Immunology and Oncology, APHM, La Timone Children's Hospital, Marseille, France; CEReS Research Unit EA 3279 and Department of Public Health, Hôpital de la Timone, Marseille, France
| |
Collapse
|
15
|
Aden NL, Bleeke M, Kordes UR, Brunne B, Holstermann B, Biemann R, Ceglarek U, Soave A, Salzbrunn A, Schneider SW, von Kopylow K. Germ Cell Maintenance and Sustained Testosterone and Precursor Hormone Production in Human Prepubertal Testis Organ Culture with Tissues from Boys 7 Years+ under Conditions from Adult Testicular Tissue. Cells 2023; 12:cells12030415. [PMID: 36766757 PMCID: PMC9913959 DOI: 10.3390/cells12030415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/20/2023] [Accepted: 01/24/2023] [Indexed: 01/28/2023] Open
Abstract
Human prepubertal testicular tissues are rare, but organ culture conditions to develop a system for human in vitro-spermatogenesis are an essential option for fertility preservation in prepubertal boys subjected to gonadotoxic therapy. To avoid animal testing in line with the 3Rs principle, organ culture conditions initially tested on human adult testis tissue were applied to prepubertal samples (n = 3; patient ages 7, 9, and 12 years). Tissues were investigated by immunostaining and transmission electron microscopy (TEM), and the collected culture medium was profiled for steroid hormones by liquid chromatography-tandem mass spectrometry (LC-MS/MS). Culture conditions proved suitable for prepubertal organ culture since SSCs and germ cell proliferation could be maintained until the end of the 3-week-culture. Leydig cells (LCs) were shown to be competent for steroid hormone production. Three additional testis tissues from boys of the same age were examined for the number of germ cells and undifferentiated spermatogonia (SPG). Using TEM micrographs, eight tissues from patients aged 1.5 to 13 years were examined, with respect to the sizes of mitochondria (MT) in undifferentiated SPG and compared with those from two adult testicular tissues. Mitochondrial sizes were shown to be comparable between adults and prepubertal boys from approximately 7 years of age, which suggests the transition of SSCs from normoxic to hypoxic metabolism at about or before this time period.
Collapse
Affiliation(s)
- Neels Lennart Aden
- Clinic and Polyclinic for Dermatology and Venerology, Andrological Section, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Matthias Bleeke
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Uwe R. Kordes
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Bianka Brunne
- Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Barbara Holstermann
- Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Ronald Biemann
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University of Leipzig, 04103 Leipzig, Germany
| | - Uta Ceglarek
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University of Leipzig, 04103 Leipzig, Germany
| | - Armin Soave
- Department of Urology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Andrea Salzbrunn
- Clinic and Polyclinic for Dermatology and Venerology, Andrological Section, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Stefan W. Schneider
- Clinic and Polyclinic for Dermatology and Venerology, Andrological Section, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Kathrein von Kopylow
- Clinic and Polyclinic for Dermatology and Venerology, Andrological Section, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Correspondence:
| |
Collapse
|
16
|
Comparison of Anti-Mullerian Hormone Levels Pre- and Post-Hematopoietic Cell Transplantation in Pediatric and Adolescent Females with Sickle Cell Disease. Transplant Cell Ther 2022; 28:770.e1-770.e6. [DOI: 10.1016/j.jtct.2022.08.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 07/23/2022] [Accepted: 08/09/2022] [Indexed: 11/18/2022]
|
17
|
Delessard M, Stalin L, Rives-Feraille A, Moutard L, Saulnier J, Dumont L, Rives N, Rondanino C. Achievement of complete in vitro spermatogenesis in testicular tissues from prepubertal mice exposed to mono- or polychemotherapy. Sci Rep 2022; 12:7407. [PMID: 35523907 PMCID: PMC9076692 DOI: 10.1038/s41598-022-11286-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 04/18/2022] [Indexed: 02/08/2023] Open
Abstract
The assessment of the impact of chemotherapies on in vitro spermatogenesis in experimental models is required before considering the application of this fertility restoration strategy to prepubertal boys who received these treatments before testicular tissue cryopreservation. The present work investigated the effects of exposure of prepubertal mice to mono- (vincristine or cyclophosphamide) and polychemotherapy (a combination of vincristine and cyclophosphamide) on the first wave of in vitro spermatogenesis. When testicular tissue exposed to monochemotherapy was preserved, polychemotherapy led to severe alterations of the seminiferous epithelium and increased apoptosis in prepubertal testes prior in vitro maturation, suggesting a potential additive gonadotoxic effect. These alterations were also found in the testicular tissues of polychemotherapy-treated mice after 30 days of organotypic culture and were associated with a reduction in the germ cell/Sertoli cell ratio. The different treatments neither altered the ability of spermatogonia to differentiate in vitro into spermatozoa nor the yield of in vitro spermatogenesis. However, more spermatozoa with morphological abnormalities and fragmented DNA were produced after administration of polychemotherapy. This work therefore shows for the first time the possibility to achieve a complete in vitro spermatogenesis after an in vivo exposure of mice to a mono- or polychemotherapy before meiotic entry.
Collapse
Affiliation(s)
- Marion Delessard
- INSERM, U1239, Team Adrenal and Gonadal Pathophysiology, Laboratory of Neuroendocrine Endocrine and Germinal Differentiation and Communication, Rouen University Hospital, Rouen Normandy University, 76000, Rouen, France
| | - Laura Stalin
- INSERM, U1239, Team Adrenal and Gonadal Pathophysiology, Laboratory of Neuroendocrine Endocrine and Germinal Differentiation and Communication, Rouen University Hospital, Rouen Normandy University, 76000, Rouen, France
| | - Aurélie Rives-Feraille
- INSERM, U1239, Team Adrenal and Gonadal Pathophysiology, Laboratory of Neuroendocrine Endocrine and Germinal Differentiation and Communication, Rouen University Hospital, Rouen Normandy University, 76000, Rouen, France
| | - Laura Moutard
- INSERM, U1239, Team Adrenal and Gonadal Pathophysiology, Laboratory of Neuroendocrine Endocrine and Germinal Differentiation and Communication, Rouen University Hospital, Rouen Normandy University, 76000, Rouen, France
| | - Justine Saulnier
- INSERM, U1239, Team Adrenal and Gonadal Pathophysiology, Laboratory of Neuroendocrine Endocrine and Germinal Differentiation and Communication, Rouen University Hospital, Rouen Normandy University, 76000, Rouen, France
| | - Ludovic Dumont
- INSERM, U1239, Team Adrenal and Gonadal Pathophysiology, Laboratory of Neuroendocrine Endocrine and Germinal Differentiation and Communication, Rouen University Hospital, Rouen Normandy University, 76000, Rouen, France
| | - Nathalie Rives
- INSERM, U1239, Team Adrenal and Gonadal Pathophysiology, Laboratory of Neuroendocrine Endocrine and Germinal Differentiation and Communication, Rouen University Hospital, Rouen Normandy University, 76000, Rouen, France
| | - Christine Rondanino
- INSERM, U1239, Team Adrenal and Gonadal Pathophysiology, Laboratory of Neuroendocrine Endocrine and Germinal Differentiation and Communication, Rouen University Hospital, Rouen Normandy University, 76000, Rouen, France.
| |
Collapse
|
18
|
Li Y, Wei L, Meinsohn MC, Suliman R, Chauvin M, Berstler J, Hartland K, Jensen MM, Sicher NA, Nagykery N, Donahoe PK, Pepin D. A screen of repurposed drugs identifies AMHR2/MISR2 agonists as potential contraceptives. Proc Natl Acad Sci U S A 2022; 119:e2122512119. [PMID: 35380904 PMCID: PMC9169708 DOI: 10.1073/pnas.2122512119] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 02/22/2022] [Indexed: 11/18/2022] Open
Abstract
We identified the anti-Mullerian hormone (also known as Müllerian inhibiting substance or MIS) as an inhibitory hormone that induces long-term contraception in mammals. The type II receptor to this hormone, AMHR2 (also known as MISR2), represents a promising druggable target for the modulation of female reproduction with a mechanism of action distinct from steroidal contraceptives. We designed an in vitro platform to screen and validate small molecules that can activate MISR2 signaling and suppress ovarian folliculogenesis. Using a bone morphogenesis protein (BMP)–response element luciferase reporter cell–based assay, we screened 5,440 compounds from a repurposed drug library. Positive hits in this screen were tested for specificity and potency in luciferase dose–response assays, and biological activity was tested in ex vivo Mullerian duct regression bioassays. Selected candidates were further evaluated in ex vivo follicle/ovary culture assays and in vivo in mice and rats. Here, we report that SP600125, CYC-116, gandotinib, and ruxolitinib can specifically inhibit primordial follicle activation and repress folliculogenesis by stimulating the MISR2 pathway.
Collapse
Affiliation(s)
- Yi Li
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA 02114
- Department of Surgery, Harvard Medical School, Boston, MA 02115
| | - Lina Wei
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA 02114
- Department of Surgery, Harvard Medical School, Boston, MA 02115
| | - Marie-Charlotte Meinsohn
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA 02114
- Department of Surgery, Harvard Medical School, Boston, MA 02115
| | - Rana Suliman
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA 02114
- Department of Surgery, Harvard Medical School, Boston, MA 02115
| | - Maeva Chauvin
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA 02114
- Department of Surgery, Harvard Medical School, Boston, MA 02115
| | - Jim Berstler
- Center for the Development of Therapeutics (CDoT), Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142
| | - Kate Hartland
- Center for the Development of Therapeutics (CDoT), Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142
| | - Mark M Jensen
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA 02114
- Department of Surgery, Harvard Medical School, Boston, MA 02115
| | - Natalie A Sicher
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA 02114
- Department of Surgery, Harvard Medical School, Boston, MA 02115
| | - Nicholas Nagykery
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA 02114
- Department of Surgery, Harvard Medical School, Boston, MA 02115
| | - Patricia K Donahoe
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA 02114
- Department of Surgery, Harvard Medical School, Boston, MA 02115
| | - David Pepin
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA 02114
- Department of Surgery, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
19
|
Delgouffe E, Braye A, Goossens E. Testicular Tissue Banking for Fertility Preservation in Young Boys: Which Patients Should Be Included? Front Endocrinol (Lausanne) 2022; 13:854186. [PMID: 35360062 PMCID: PMC8960265 DOI: 10.3389/fendo.2022.854186] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 02/11/2022] [Indexed: 12/11/2022] Open
Abstract
Due to the growing number of young patients at risk of germ cell loss, there is a need to preserve spermatogonial stem cells for patients who are not able to bank spermatozoa. Worldwide, more and more clinics are implementing testicular tissue (TT) banking programs, making it a novel, yet indispensable, discipline in the field of fertility preservation. Previously, TT cryopreservation was predominantly offered to young cancer patients before starting gonadotoxic chemo- or radiotherapy. Nowadays, most centers also bank TT from patients with non-malignant conditions who need gonadotoxic conditioning therapy prior to hematopoietic stem cell (HSCT) or bone marrow transplantation (BMT). Additionally, some centers include patients who suffer from genetic or developmental disorders associated with prepubertal germ cell loss or patients who already had a previous round of chemo- or radiotherapy. It is important to note that the surgical removal of TT is an invasive procedure. Moreover, TT cryopreservation is still considered experimental as restoration methods are not yet clinically available. For this reason, TT banking should preferably only be offered to patients who are at significant risk of becoming infertile. In our view, TT cryopreservation is recommended for young cancer patients in need of high-risk chemo- and/or radiotherapy, regardless of previous low-risk treatment. Likewise, TT banking is advised for patients with non-malignant disorders such as sickle cell disease, beta-thalassemia, and bone marrow failure, who need high-risk conditioning therapy before HSCT/BMT. TT retrieval during orchidopexy is also proposed for patients with bilateral cryptorchidism. Since patients with a medium- to low-risk treatment generally maintain their fertility, TT banking is not advised for this group. Also for Klinefelter patients, TT banking is not recommended as it does not give better outcomes than a testicular sperm extraction later in life.
Collapse
|
20
|
Female Oncofertility: Current Understandings, Therapeutic Approaches, Controversies, and Future Perspectives. J Clin Med 2021; 10:jcm10235690. [PMID: 34884393 PMCID: PMC8658080 DOI: 10.3390/jcm10235690] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 11/25/2021] [Accepted: 12/02/2021] [Indexed: 12/14/2022] Open
Abstract
Recent advances in early detection and oncological therapies have ameliorated the survival rate of young cancer patients. Yet, ovarian impairment induced by chemotherapy and radiotherapy is still a challenging issue. This review, based on clinical and lab-based studies, summarizes the evidence of gonadotoxicity of chemoradiotherapy, the recent approaches, ongoing controversies, and future perspectives of fertility preservation (FP) in female patients who have experienced chemo- or radio-therapy. Existing data indicate that chemotherapeutic agents induce DNA alterations and massive follicle activation via the phosphoinositide 3-kinase (PI3K)/Akt signaling pathway. Meanwhile, the radiation causes ionizing damage, leading to germ cell loss. In addition to the well-established methods, numerous therapeutic approaches have been suggested, including minimizing the follicle loss in cryopreserved ovarian grafts after transplantation, in vitro activation or in vitro growing of follicles, artificial ovarian development, or fertoprotective adjuvant to prevent ovarian damage from chemotherapy. Some reports have revealed positive outcomes from these therapies, whereas others have demonstrated conflictions. Future perspectives are improving the live birth rate of FP, especially in patients with adverse ovarian reserve, eliminating the risk of malignancy reintroducing, and increasing society’s awareness of FP importance.
Collapse
|