1
|
Stewart S, White A, Ou W, Liu W, Nagashima J, Songsasen N, He X. Controlled Ice Nucleation With a Sand-PDMS Film Device Enhances Cryopreservation of Mouse Preantral Ovarian Follicles. J Med Device 2024; 18:041007. [PMID: 39465055 PMCID: PMC11500804 DOI: 10.1115/1.4066445] [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: 04/18/2024] [Revised: 08/28/2024] [Indexed: 10/29/2024] Open
Abstract
Ovarian follicle cryopreservation is a promising strategy for fertility preservation; however, cryopreservation protocols have room for improvement to maximize post-thaw follicle viability and quality. Current slow-freezing protocols use either manual ice-seeding in combination with expensive programmable-rate freezers or other clinically incompatible ice initiators to control the ice-seeding temperature in the extracellular solution, a critical parameter that impacts post-cryopreservation cell/tissue quality. Previously, sand has been shown to be an excellent, biocompatible ice initiator, and its use in cryopreservation of human induced pluripotent stem cells enables high cell viability and quality after cryopreservation. This study applies sand as an ice initiator to cryopreserve multicellular microtissue, preantral ovarian follicles, using a simple slow-freezing protocol in the mouse model. Ovarian follicles cryopreserved using the sand partially embedded in polydimethylsiloxane (PDMS) film to seed ice in the extracellular solution exhibit healthy morphology, high viability, and the ability to grow similarly to fresh follicles in culture post-thaw. This sand-based cryopreservation strategy can facilitate convenient ovarian follicle cryopreservation using simple equipment, and this study further demonstrates the translatability of this strategy to not only single cells but also multicellular tissues.
Collapse
Affiliation(s)
- Samantha Stewart
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
| | - Alisa White
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
| | - Wenquan Ou
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
| | - Wei Liu
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
| | - Jennifer Nagashima
- Center for Species Survival, Smithsonian National Zoo and Conservation Biology Institute, 1500 Remount Road, Front Royal, VA 22630
| | - Nucharin Songsasen
- Center for Species Survival, Smithsonian National Zoo and Conservation Biology Institute, 1500 Remount Road, Front Royal, VA 22630
| | - Xiaoming He
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742; Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD 20742; Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD 21201
| |
Collapse
|
2
|
McElhinney KL, Orr S, Gelarden IA, Laronda MM, Rowell EE. Is Routine Pathology Evaluation of Tissue Removed for Fertility Preservation Necessary? J Pediatr Surg 2024; 59:161632. [PMID: 39117537 PMCID: PMC11546292 DOI: 10.1016/j.jpedsurg.2024.07.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 07/10/2024] [Accepted: 07/14/2024] [Indexed: 08/10/2024]
Abstract
BACKGROUND For all fertility preservation (FP) cases at our institution, a biopsy is performed for routine pathology from all gonadal tissue removed. This is not standard at all centers. We reviewed our experience with biopsy for pathological evaluation of ovarian and testicular specimens in FP cases to determine clinical utility. METHODS The medical records of individuals who underwent ovarian tissue cryopreservation (OTC) or testicular tissue cryopreservation (TTC) between 2011 and 2023 were retrospectively reviewed under an IRB-approved study at a free-standing tertiary care children's hospital. Patient demographics, diagnosis, operative characteristics, and pathology results were collected. RESULTS One-hundred and eighty-three patients underwent OTC, and 134 patients underwent TTC. All patients had their gonadal tissue biopsied for routine pathology. Malignancy was identified in the biopsies of 4 OTC patients (2.2%) and 2 TTC patients (1.5%). Two OTC patients (1.1%) and 2 TTC patients (1.5%) did not have germ cells identified in their biopsy. All OTC and TTC patients and families elected to continue storing tissue for FP after discussion of pathology findings. CONCLUSIONS Pathology results provide another data point to help inform patients and their families when making decisions on ovarian or testicular tissue storage and on how tissue may be utilized in the future to restore fertility and/or hormones. There is a low rate of identifying malignancy in gonadal tissue biopsies taken from FP specimens even in patients with known malignancy. However, when malignancy was identified, it could be unexpected and alter the diagnosis and treatment plan significantly for patients. LEVEL OF EVIDENCE IV.
Collapse
Affiliation(s)
- Kathryn L McElhinney
- Fertility & Hormone Preservation & Restoration Program, Division of Pediatric Surgery, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA; Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA; Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| | - Sierra Orr
- Fertility & Hormone Preservation & Restoration Program, Division of Pediatric Surgery, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA; Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Ian A Gelarden
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Monica M Laronda
- Fertility & Hormone Preservation & Restoration Program, Division of Pediatric Surgery, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA; Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA; Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA; Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Erin E Rowell
- Fertility & Hormone Preservation & Restoration Program, Division of Pediatric Surgery, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA; Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA; Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
3
|
Dunlop CE, Anderson RA. Clinical dilemmas in ovarian tissue cryopreservation. Fertil Steril 2024; 122:559-564. [PMID: 38825305 DOI: 10.1016/j.fertnstert.2024.05.167] [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: 05/05/2024] [Revised: 05/26/2024] [Accepted: 05/29/2024] [Indexed: 06/04/2024]
Abstract
Ovarian tissue cryopreservation (OTC) is increasingly offered globally as a fertility preservation strategy for both postpubertal women and prepubertal girls, with subsequent reimplantation of cryopreserved ovarian cortex resulting in a rapidly growing number of live births. There remains very limited evidence of efficacy from tissue stored when the patient was prepubertal or from conditions affecting the ovary directly, e.g., Turner syndrome. Although OTC is becoming a more established practice, several clinical dilemmas remain from a practical and ethical standpoint. This review discusses the challenges regarding optimal patient selection for the procedure, the use of OTC in patients with a poor prognosis, the potential of reimplantation of tissue contaminated with malignant cells, and the role of OTC in those with an intrinsic ovarian disorder.
Collapse
Affiliation(s)
- Cheryl E Dunlop
- Obstetrics & Gynaecology Department, Edinburgh Royal Infirmary, Edinburgh, United Kingdom.
| | - Richard A Anderson
- Centre for Reproductive Health, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
4
|
Zhang J, Li X, Liang R, Duan S, Yang X, Hou Y, Tian L. Ovarian tissue cryopreservation after graft failure of allogeneic hematopoietic stem cell transplantation: first report and literature review. Front Endocrinol (Lausanne) 2024; 15:1367241. [PMID: 39253581 PMCID: PMC11382424 DOI: 10.3389/fendo.2024.1367241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 08/01/2024] [Indexed: 09/11/2024] Open
Abstract
Background Hematopoietic stem cell transplantation (HSCT) is an approach that has significantly improved the prognosis and survival of hematological patients. However, ovarian dysfunction and infertility following HSCT have gained increasing attention. Live births have been reported following ovarian tissue cryopreservation prior to HSCT and subsequent retransplantation of these tissues. Still, the feasibility of ovarian tissue cryopreservation (OTC) following graft failure (GF) of HSCT remains unknown. In this study, we report the first case of OTC following a GF of allogenic HSCT (allo-HSCT), as well as the cryopreservation of four MII oocytes via in vitro maturation with informed consent. Despite the lack of clinical outcomes after cryopreserved ovarian tissue retransplantation, we documented an interesting case in a woman after GF of allo-HSCT exhibiting functional ovaries and emphasized a clinical dilemma: whether OTC should be offered to women suffering from GF of HSCT. Case presentation A 22-year-old woman with severe aplastic anemia who had suffered GF of allo-HSCT from her sibling brother [HLA allele match (7/10)] with a reduced dose conditioning regimen including fludarabine, cyclophosphamide, and antithymocyte globulin came to our reproductive center for fertility preservation, as she was about to receive the second allo-HSCT. We evaluated the ovarian reserve of this patient. Hormone assessments showed an anti-Müllerian hormone level of 3.921 ng/mL, a follicle-stimulating hormone level of 5.88 IU/L, a luteinizing hormone level of 10.79 IU/L, and an estradiol level of 33.34 pg/mL. Antral follicle counts accessed transvaginally showed 12-15 follicles. All assessments indicated a well-protected ovarian reserve. Due to the urgency of the second allo-HSCT, the patient decided to undergo ovarian cryopreservation. Laparoscopic surgery proceeded. Ovarian tissues were successfully cryopreserved using vitrification technology, and histologic evaluation demonstrated a follicle density of 20 per 2 × 2 mm2 biopsy with good viability. Four MII oocytes were obtained via in vitro maturation technology and cryopreserved. After the second HSCT, the patient relieved from aplastic anemia but suffered iatrogenic premature ovarian failure as predicted. Conclusion OTC is applicable to fertility preservation in those undergoing GF of HSCT with benign hematological disorders and especially those who are about to receive the second HSCT.
Collapse
Affiliation(s)
- Jinghua Zhang
- Department of Obstetrics and Gynecology, Peking University People’s Hospital, Beijing, China
- Reproductive Medical Center, Peking University People’s Hospital, Beijing, China
| | - Xiaowei Li
- Department of Obstetrics and Gynecology, Peking University People’s Hospital, Beijing, China
| | - Rong Liang
- Department of Obstetrics and Gynecology, Peking University People’s Hospital, Beijing, China
- Reproductive Medical Center, Peking University People’s Hospital, Beijing, China
| | - Shengnan Duan
- Department of Obstetrics and Gynecology, Peking University People’s Hospital, Beijing, China
- Reproductive Medical Center, Peking University People’s Hospital, Beijing, China
| | - Xin Yang
- Department of Obstetrics and Gynecology, Peking University People’s Hospital, Beijing, China
| | - Yanru Hou
- Department of Obstetrics and Gynecology, Peking University People’s Hospital, Beijing, China
- Reproductive Medical Center, Peking University People’s Hospital, Beijing, China
| | - Li Tian
- Department of Obstetrics and Gynecology, Peking University People’s Hospital, Beijing, China
- Reproductive Medical Center, Peking University People’s Hospital, Beijing, China
| |
Collapse
|
5
|
Vitale F, Cacciottola L, Camboni A, Houeis L, Donnez J, Dolmans MM. Assessing the effect of adipose-tissue-derived stem cell conditioned medium on follicles and stromal cells in bovine ovarian tissue culture. Reprod Biomed Online 2024; 49:103938. [PMID: 38759499 DOI: 10.1016/j.rbmo.2024.103938] [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: 10/06/2023] [Revised: 01/31/2024] [Accepted: 03/05/2024] [Indexed: 05/19/2024]
Abstract
RESEARCH QUESTION Does adipose-tissue-derived stem cell conditioned medium (ASC-CM) supplementation enhance follicle and stromal cell outcomes in vitro? DESIGN Bovine ovaries (n = 8) were sectioned and cultured in vitro for 8 days in two different groups: (i) standard culture (OT Ctrl D8); and (ii) culture with ASC-CM supplementation (OT + CM D8). Half of the culture medium was replaced every other day, and stored to measure the production of oestradiol. Follicle classification was established using haematoxylin and eosin staining. Follicle and stromal cell DNA fragmentation was assessed by TUNEL assays, while growth differentiation factor-9 (GDF-9) staining served as a marker of follicle quality. Additionally, three factors, namely vascular endothelial growth factor (VEGF), interleukin 6 (IL-6) and transforming growth factor beta 1 (TGF-β1), were evaluated in ASC-CM in order to appraise the potential underlying mechanisms of action of ASC. RESULTS The OT + CM D8 group showed a significantly higher proportion of secondary follicles (P = 0.02) compared with the OT Ctrl D8 group. The OT + CM D8 group also demonstrated significantly lower percentages of TUNEL-positive follicles (P = 0.014) and stromal cells (P = 0.001) compared with the OT Ctrl D8 group. Furthermore, follicles in the OT + CM D8 group exhibited a significant increase (P = 0.002) in expression of GDF-9 compared with those in the OT Ctrl D8 group, and oestradiol production was significantly higher (P = 0.04) in the OT + CM D8 group. All studied factors were found to be present in ASC-CM. VEGF and IL-6 were the most widely expressed factors, while TGF-β1 showed the lowest expression. CONCLUSIONS Addition of ASC-CM to culture medium enhances follicle survival, development and oestradiol production, and promotes the viability of stromal cells. VEGF, IL-6 and TGF-β1 could be paracrine mediators underlying the beneficial effects.
Collapse
Affiliation(s)
- Francisco Vitale
- Gynaecology Research Unit, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Luciana Cacciottola
- Gynaecology Research Unit, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Alessandra Camboni
- Gynaecology Research Unit, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium; Pathology Department, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Lara Houeis
- Gynaecology Research Unit, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Jacques Donnez
- Société de Recherche pour l'Infertilité, Brussels, Belgium; Professor Em, Université Catholique de Louvain, Brussels, Belgium
| | - Marie-Madeleine Dolmans
- Gynaecology Research Unit, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium; Gynaecology Department, Cliniques Universitaires Saint-Luc, Brussels, Belgium.
| |
Collapse
|
6
|
McElhinney KL, Rowell EE, Laronda MM. Encapsulation of Bovine Primordial Follicles in Rigid Alginate Does Not Affect Growth Dynamics. Bioengineering (Basel) 2024; 11:734. [PMID: 39061816 PMCID: PMC11273846 DOI: 10.3390/bioengineering11070734] [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: 05/22/2024] [Revised: 07/02/2024] [Accepted: 07/07/2024] [Indexed: 07/28/2024] Open
Abstract
The only fertility preservation and subsequent restoration option for many patients facing gonadotoxic treatments is ovarian tissue cryopreservation and transplantation. While this process is successful for some, there is significant room for improvement to extend the life of the transplant and to make it safe for patients that may have metastatic disease within their ovarian tissue. We need a deeper understanding of how the physical properties of the ovarian microenvironment may affect folliculogenesis to engineer an environment that supports isolated follicles and maintains primordial follicle quiescence. Bovine ovaries were used here as a monovulatory model of folliculogenesis to examine the effects of primordial follicle activation and growth under different physical conditions. We found that there were no differences in activation, growth or survival when primordial follicles were cultured in isolation or in situ (remaining in the tissue) under two significantly differently rigid alginate gels. To determine if the extra rigid environment did not affect activation in isolated follicles due to an immediate activation event, we used 5-ethynyl-2'-deoxyuridine (EdU) to track follicle activation during the isolation process. We identified EdU incorporation in granulosa cells after primordial follicles were isolated from the surrounding extracellular matrix (ECM). These findings support that isolation of primordial follicles from the ECM is an activating event and that the differentially rigid environments assessed here had no effect on follicle growth. Further work is needed to suppress activation in primordial follicles to maintain the ovarian reserve and extend the life of an ovarian tissue transplant.
Collapse
Affiliation(s)
- Kathryn L. McElhinney
- Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA; (K.L.M.)
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Department of Pediatrics, Division of Endocrinology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Erin E. Rowell
- Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA; (K.L.M.)
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Monica M. Laronda
- Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA; (K.L.M.)
- Department of Pediatrics, Division of Endocrinology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
7
|
Chevillon F, Rebotier M, Dhédin N, Bruno B, Cacciatore C, Charbonier A, Joseph L, Le Bourgeois A, Talouarn M, Magro L, Barraud Lange V. [Fertility preservation and hematopoietic stem cell transplantation (SFGM-TC)]. Bull Cancer 2024:S0007-4551(24)00185-1. [PMID: 38918137 DOI: 10.1016/j.bulcan.2024.04.009] [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: 02/12/2024] [Revised: 04/20/2024] [Accepted: 04/22/2024] [Indexed: 06/27/2024]
Abstract
Conditioning regimen prior to hematopoietic stem cell transplantation have an impact on patient fertility through the use of gonadal irradiation and/or bifunctional alkylating agents. Their impact on fertility depends mainly on the dose used and, in women, on age at the time of treatment. All patients should benefit before treatment from a consultation informing them of the potential impact on fertility and of fertility preservation techniques. In the absence of contraindications, the major toxicity of myeloablative conditioning regimen justifies fertility preservation. There are few data concerning fertility after reduced-intensity conditioning. Despite lower theoretical gonadotoxicity, we also recommend fertility preservation, if possible before transplantation. The fertility preservation techniques used depend on the patient's age, pathology and conditioning. In the event of subsequent use of harvested gonadal tissue in the context of acute leukemia or aggressive lymphoma, it is advisable to assess the risk of reintroduction of tumor cells. Finally, it is recommended to assess gonadal function after transplant, especially after reduced conditioning. If there is persistent residual gonadal function, post-treatment fertility preservation should be discuss.
Collapse
Affiliation(s)
- Florian Chevillon
- Service hématologie adolescents jeunes adultes, hôpital Saint-Louis, AP-HP, 1, avenue Claude-Vellefaux, 75010 Paris, France.
| | - Marine Rebotier
- Service oncogynécologie, centre Leon-Berard et IHOPe, 28, promenade Léa et Napoléon Bullukian, 69008 Lyon, France
| | - Nathalie Dhédin
- Service hématologie adolescents jeunes adultes, hôpital Saint-Louis, AP-HP, 1, avenue Claude-Vellefaux, 75010 Paris, France
| | - Bénédicte Bruno
- Service hématologie pédiatrique, hôpital Jeanne-de-Flandre, avenue Eugène-Avinée, 59037 Lille, France
| | - Carlotta Cacciatore
- Service de médecine interne, hôpital Saint-Louis, AP-HP, 1, avenue Claude-Vellefaux, 75010 Paris, France
| | | | - Laure Joseph
- Service hématologie, département de biothérapie, hôpital Necker-enfants malades, AP-HP, 149, rue de Sèvres, 75015 Paris, France
| | - Amandine Le Bourgeois
- Service d'hématologie, CHU de Nantes, 1, place Alexis-Ricordeau, 44000 Nantes, France
| | - Marie Talouarn
- Service d'hématologie, hôpital Saint-Antoine, AP-HP, 184, rue du faubourg Saint-Antoine, 75012 Paris, France
| | - Leonardo Magro
- Service d'hématologie, CHU de Lille, 2, avenue Oscar-Lambret, 59000 Lille, France
| | - Virginie Barraud Lange
- Service hématologie adolescents jeunes adultes, hôpital Saint-Louis, AP-HP, 1, avenue Claude-Vellefaux, 75010 Paris, France; Service biologie de la reproduction, hôpital Cochin Port Royal, AP-HP, 123, boulevard de Port Royal, 75014 Paris, France
| |
Collapse
|
8
|
Jiang M, Zhang GH, Yu Y, Zhao YH, Liu J, Zeng Q, Feng MY, Ye F, Xiong DS, Wang L, Zhang YN, Yu L, Wei JJ, He LB, Zhi W, Du XR, Li NJ, Han CL, Yan HQ, Zhou ZT, Miao YB, Wang W, Liu WX. De novo design of a nanoregulator for the dynamic restoration of ovarian tissue in cryopreservation and transplantation. J Nanobiotechnology 2024; 22:330. [PMID: 38862987 PMCID: PMC11167790 DOI: 10.1186/s12951-024-02602-5] [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: 03/27/2024] [Accepted: 05/28/2024] [Indexed: 06/13/2024] Open
Abstract
The cryopreservation and transplantation of ovarian tissue underscore its paramount importance in safeguarding reproductive capacity and ameliorating reproductive disorders. However, challenges persist in ovarian tissue cryopreservation and transplantation (OTC-T), including the risk of tissue damage and dysfunction. Consequently, there has been a compelling exploration into the realm of nanoregulators to refine and enhance these procedures. This review embarks on a meticulous examination of the intricate anatomical structure of the ovary and its microenvironment, thereby establishing a robust groundwork for the development of nanomodulators. It systematically categorizes nanoregulators and delves deeply into their functions and mechanisms, meticulously tailored for optimizing ovarian tissue cryopreservation and transplantation. Furthermore, the review imparts valuable insights into the practical applications and obstacles encountered in clinical settings associated with OTC-T. Moreover, the review advocates for the utilization of microbially derived nanomodulators as a potent therapeutic intervention in ovarian tissue cryopreservation. The progression of these approaches holds the promise of seamlessly integrating nanoregulators into OTC-T practices, thereby heralding a new era of expansive applications and auspicious prospects in this pivotal domain.
Collapse
Affiliation(s)
- Min Jiang
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, 610045, China
| | - Guo-Hui Zhang
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, 610045, China
| | - Yuan Yu
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Yu-Hong Zhao
- School of Clinical Laboratory Medicine, Chengdu Medical College, Chengdu, 610083, China
| | - Jun Liu
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Qin Zeng
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, 610045, China
| | - Meng-Yue Feng
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Fei Ye
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, 610045, China
| | - Dong-Sheng Xiong
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, 610045, China
| | - Li Wang
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, 610045, China
| | - Ya-Nan Zhang
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, 610045, China
| | - Ling Yu
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, 610045, China
| | - Jia-Jing Wei
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, 610045, China
| | - Li-Bing He
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, 610045, China
| | - Weiwei Zhi
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, 610045, China
| | - Xin-Rong Du
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Ning-Jing Li
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Chang-Li Han
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - He-Qiu Yan
- School of Clinical Laboratory Medicine, Chengdu Medical College, Chengdu, 610083, China
| | - Zhuo-Ting Zhou
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Yang-Bao Miao
- Department of Haematology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610000, China.
| | - Wen Wang
- Department of Haematology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610000, China.
| | - Wei-Xin Liu
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China.
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, 610045, China.
| |
Collapse
|
9
|
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
|
10
|
Marco A, Gargallo M, Ciriza J, Shikanov A, Baquedano L, García Pérez-Llantada J, Malo C. Current Fertility Preservation Steps in Young Women Suffering from Cancer and Future Perspectives. Int J Mol Sci 2024; 25:4360. [PMID: 38673945 PMCID: PMC11050570 DOI: 10.3390/ijms25084360] [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: 03/07/2024] [Revised: 04/05/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
Childhood cancer incidence, especially in high-income countries, has led to a focus on preserving fertility in this vulnerable population. The common treatments, such as radiation and certain chemotherapeutic agents, though effective, pose a risk to fertility. For adult women, established techniques like embryo and egg freezing are standard, requiring ovarian stimulation. However, for prepubescent girls, ovarian tissue freezing has become the primary option, eliminating the need for hormonal preparation. This review describes the beginning, evolution, and current situation of the fertility preservation options for this young population. A total of 75 studies were included, covering the steps in the current fertility preservation protocols: (i) ovarian tissue extraction, (ii) the freezing method, and (iii) thawing and transplantation. Cryopreservation and the subsequent transplantation of ovarian tissue have resulted in successful fertility restoration, with over 200 recorded live births, including cases involving ovarian tissue cryopreserved from prepubescent girls. Despite promising results, challenges persist, such as follicular loss during transplantation, which is attributed to ischemic and oxidative damage. Optimizing ovarian tissue-freezing processes and exploring alternatives to transplantation, like in vitro systems for follicles to establish maturation, are essential to mitigating associated risks. Further research is required in fertility preservation techniques to enhance clinical outcomes in the future. Ovarian tissue cryopreservation appears to be a method with specific benefits, indications, and risks, which can be an important tool in terms of preserving fertility in younger women.
Collapse
Affiliation(s)
- Alicia Marco
- Faculty of Medicine, University of Zaragoza, 50018 Zaragoza, Spain;
| | - Marta Gargallo
- Institute for Health Research Aragón (IIS Aragón), 50009 Zaragoza, Spain; (M.G.); (J.C.)
| | - Jesús Ciriza
- Institute for Health Research Aragón (IIS Aragón), 50009 Zaragoza, Spain; (M.G.); (J.C.)
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain
| | - Ariella Shikanov
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA;
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI 48109, USA
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI 48109, USA
| | - Laura Baquedano
- Department of Gynecology, University Hospital Miguel Servat, 50009 Zaragoza, Spain;
| | | | - Clara Malo
- Institute for Health Research Aragón (IIS Aragón), 50009 Zaragoza, Spain; (M.G.); (J.C.)
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain
| |
Collapse
|
11
|
Tsui EL, McDowell HB, Laronda MM. Restoring Ovarian Fertility and Hormone Function: Recent Advancements, Ongoing Efforts and Future Applications. J Endocr Soc 2024; 8:bvae073. [PMID: 38698870 PMCID: PMC11065362 DOI: 10.1210/jendso/bvae073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Indexed: 05/05/2024] Open
Abstract
The last 20 years have seen substantial improvements in fertility and hormone preservation and restoration technologies for a growing number of cancer survivors. However, further advancements are required to fill the gaps for those who cannot use current technologies or to improve the efficacy and longevity of current fertility and hormone restoration technologies. Ovarian tissue cryopreservation (OTC) followed by ovarian tissue transplantation (OTT) offers those unable to undergo ovarian stimulation for egg retrieval and cryopreservation an option that restores both fertility and hormone function. However, those with metastatic disease in their ovaries are unable to transplant this tissue. Therefore, new technologies to produce good-quality eggs and restore long-term cyclic ovarian function are being investigated and developed to expand options for a variety of patients. This mini-review describes current and near future technologies including in vitro maturation, in vitro follicle growth and maturation, bioprosthetic ovaries, and stem cell applications in fertility restoration research by their proximity to clinical application.
Collapse
Affiliation(s)
- Elizabeth L Tsui
- Department of Pediatrics, Division of Endocrinology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| | - Hannah B McDowell
- Department of Pediatrics, Division of Endocrinology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| | - Monica M Laronda
- Department of Pediatrics, Division of Endocrinology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
12
|
Deligiannis SP, Kask K, Modhukur V, Boskovic N, Ivask M, Jaakma Ü, Damdimopoulou P, Tuuri T, Velthut-Meikas A, Salumets A. Investigating the impact of vitrification on bovine ovarian tissue morphology, follicle survival, and transcriptomic signature. J Assist Reprod Genet 2024; 41:1035-1055. [PMID: 38358432 PMCID: PMC11052753 DOI: 10.1007/s10815-024-03038-4] [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: 05/31/2023] [Accepted: 01/18/2024] [Indexed: 02/16/2024] Open
Abstract
PURPOSE Ovarian tissue cryopreservation is vital for fertility preservation, yet its effect on ovarian tissue follicle survival and transcriptomic signature requires further investigation. This study delves into the effects of vitrification on tissue morphology, function, and transcriptomic changes, helping to find possibilities for vitrification protocol improvements. METHODS Ovarian cortex from 19 bovine animals were used to conduct pre- and post-vitrification culture followed by histological assessment, immunohistochemistry, and TUNEL assay. Follicles' functionality was assessed for viability and growth within the tissue and in isolated cultures. RNA-sequencing of ovarian tissue was used to explore the transcriptomic alterations caused by vitrification. RESULTS Follicle density, cell proliferation, and DNA damage in ovarian stroma were unaffected by vitrification. However, vitrified cultured tissue exhibited reduced follicle density of primordial/primary and antral follicles, while freshly cultured tissue manifested reduction of antral follicles. Increased stromal cell proliferation and DNA damage occurred in both groups post-culture. Isolated follicles from vitrified tissue exhibited similar viability to fresh follicles until day 4, after which the survival dropped. RNA-sequencing revealed minor effects of vitrification on transcriptomic signatures, while culture induced significant gene expression changes in both groups. The altered expression of WNT and hormonal regulation pathway genes post-vitrification suggests the molecular targets for vitrification protocol refinement. CONCLUSION Vitrification minimally affects tissue morphology, follicle density, and transcriptomic signature post-thawing. However, culture revealed notable changes in vitrified tissue samples, including reduced follicle density, decreased isolated follicle survival, and alteration in WNT signalling and ovarian hormonal regulation pathways, highlighted them as possible limitations of the current vitrification protocol.
Collapse
Affiliation(s)
- Spyridon P Deligiannis
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Huddinge, 14186, Stockholm, Sweden.
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, 14186, Stockholm, Sweden.
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, 50406, Tartu, Estonia.
- Department of Obstetrics and Gynecology, University of Helsinki, 00290, Helsinki, Finland.
| | - Keiu Kask
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, 50406, Tartu, Estonia
- Competence Centre of Health Technologies, 50411, Tartu, Estonia
| | - Vijayachitra Modhukur
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, 50406, Tartu, Estonia
- Competence Centre of Health Technologies, 50411, Tartu, Estonia
| | - Nina Boskovic
- Department of Obstetrics and Gynecology, University of Helsinki, 00290, Helsinki, Finland
- Department of Biosciences and Nutrition, Karolinska Institutet, 14183, Huddinge, Sweden
| | - Marilin Ivask
- Department of Pathophysiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 50411, Tartu, Estonia
- Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, 51014, Tartu, Estonia
| | - Ülle Jaakma
- Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, 51014, Tartu, Estonia
| | - Pauliina Damdimopoulou
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Huddinge, 14186, Stockholm, Sweden
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, 14186, Stockholm, Sweden
| | - Timo Tuuri
- Department of Obstetrics and Gynecology, University of Helsinki, 00290, Helsinki, Finland
| | - Agne Velthut-Meikas
- Department of Chemistry and Biotechnology, Tallinn University of Technology, 12618, Tallinn, Estonia
| | - Andres Salumets
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Huddinge, 14186, Stockholm, Sweden.
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, 14186, Stockholm, Sweden.
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, 50406, Tartu, Estonia.
- Competence Centre of Health Technologies, 50411, Tartu, Estonia.
| |
Collapse
|
13
|
Li Y, Ruan X, Gu M, Du J, Jin F, Cheng J, Li Y, Jiang L, Wang Z, Yang Y, Zhang M, Mueck AO. Evaluating the safety and efficacy of cryopreserved ovarian tissue transplantation in leukemia patients with different bone marrow remission status using xenotransplantation. Front Endocrinol (Lausanne) 2024; 15:1364316. [PMID: 38590823 PMCID: PMC10999602 DOI: 10.3389/fendo.2024.1364316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 03/12/2024] [Indexed: 04/10/2024] Open
Abstract
Background Leukemia patients undergoing cryopreserved ovarian tissue transplantation (OTT) may carry a high risk of disease induction. Measurable residual disease (MRD) in bone marrow is linked to an elevated risk of relapse. It is controversial whether leukemia patients must be allowed to achieve measurable residual disease negative (MRD-negative) status instead of measurable residual disease positive (MRD-positive) status before ovarian tissue cryopreservation (OTC). Objective To explore the safety and efficacy of OTT in acute leukemia patients with different MRD status by using xenotransplantation. Method Cryopreserved ovarian tissue from 19 leukemia patients was thawed and xenotransplanted to ovariectomized BALB/C nude mice (n=36). The mice were divided into 2 groups based on the patient's MRD status before OTC: MRD-negative group (n=18) and MRD-positive group (n=18), additionally, a control group consisted of ovariectomized mice (n=9). Body weight was measured weekly and mortality, emaciation, and other abnormalities were recorded. Twenty-six weeks post-surgery, livers, spleens, uteruses, and ovarian grafts were removed for macroscopic and histological examinations to evaluate the efficacy of xenotransplantation and assess malignant cell contamination in mice. Results Follicle growth was visible in the ovarian grafts of the MRD-negative and MRD-positive groups. Compared with the ovariectomized group, a significant decrease in body weight (p<0.01) was noted, the uterine volume was notably larger, estradiol (E2) levels were significantly higher (p<0.01), and follicle-stimulating hormone (FSH) levels were significantly lower (p<0.001) in the other two groups. Mice in the MRD-positive group showed a significantly higher incidence of death (p<0.001) and emaciation (p<0.01), compared to the MRD-negative group. Histological observation revealed the presence of malignant cells in the grafts, livers, and spleens of 3 mice in the MRD-positive group. No abnormalities were observed in the mice from the MRD-negative group in both macroscopic and histological observations except one mouse was sacrificed for ascites unrelated to leukemia relapse. Conclusion For leukemia patients having ovarian tissue preserved in the first and only centralized human ovarian tissue cryobank in China, immunodeficient mice xenotransplantation can be a method to evaluate the safety and efficacy of OTT; the risk of malignant cell reimplantation due to OTT is higher in leukemia patients with MRD-positive status than those with MRD-negative status before OTC.
Collapse
Affiliation(s)
- Yanqiu Li
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Xiangyan Ruan
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
- Department for Women's Health, University Women's Hospital and Research Center for Women's Health, University of Tuebingen, Tuebingen, Germany
| | - Muqing Gu
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Juan Du
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Fengyu Jin
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Jiaojiao Cheng
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Yanglu Li
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Lingling Jiang
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Zecheng Wang
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Yu Yang
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Mingzhen Zhang
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Alfred O Mueck
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
- Department for Women's Health, University Women's Hospital and Research Center for Women's Health, University of Tuebingen, Tuebingen, Germany
| |
Collapse
|
14
|
Vitale F, Dolmans MM. Comprehensive Review of In Vitro Human Follicle Development for Fertility Restoration: Recent Achievements, Current Challenges, and Future Optimization Strategies. J Clin Med 2024; 13:1791. [PMID: 38542015 PMCID: PMC10970962 DOI: 10.3390/jcm13061791] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 03/11/2024] [Accepted: 03/19/2024] [Indexed: 11/11/2024] Open
Abstract
Ovarian tissue cryopreservation (OTC) and subsequent transplantation (OTT) is a fertility preservation technique widely offered to prepubertal girls and young fertile women who need to undergo oncological treatment but are at a high risk of infertility. However, OTT is not considered safe in patients with certain diseases like leukemia, Burkitt's lymphoma, and ovarian cancer because of the associated risk of malignant cell reintroduction. In vitro follicle development has therefore emerged as a promising means of obtaining mature metaphase II (MII) oocytes from the primordial follicle (PMF) pool contained within cryopreserved ovarian tissue, without the need for transplantation. Despite its significant potential, this novel approach remains highly challenging, as it requires replication of the intricate process of intraovarian folliculogenesis. Recent advances in multi-step in vitro culture (IVC) systems, tailored to the specific needs of each follicle stage, have demonstrated the feasibility of generating mature oocytes (MII) from early-stage human follicles. While significant progress has been made, there is still room for improvement in terms of efficiency and productivity, and a long way to go before this IVC approach can be implemented in a clinical setting. This comprehensive review outlines the most significant improvements in recent years, current limitations, and future optimization strategies.
Collapse
Affiliation(s)
- Francisco Vitale
- Gynecology Research Unit, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Avenue Mounier 52, 1200 Brussels, Belgium;
| | - Marie-Madeleine Dolmans
- Gynecology Research Unit, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Avenue Mounier 52, 1200 Brussels, Belgium;
- Gynecology Department, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| |
Collapse
|
15
|
Kourta D, Camboni A, Saussoy P, Kanbar M, Poels J, Wyns C. Evaluating testicular tissue for future autotransplantation: focus on cancer cell contamination and presence of spermatogonia in tissue cryobanked for boys diagnosed with a hematological malignancy. Hum Reprod 2024; 39:486-495. [PMID: 38227814 DOI: 10.1093/humrep/dead271] [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: 07/20/2023] [Revised: 11/21/2023] [Indexed: 01/18/2024] Open
Abstract
STUDY QUESTION What is the contamination rate by cancer cells and spermatogonia numbers in immature testicular tissue (ITT) harvested before the start of gonadotoxic therapy in boys with a hematological malignancy? SUMMARY ANSWER Among our cohort of boys diagnosed with acute lymphoblastic leukemia (ALL) and lymphomas, 39% (n = 11/28) had cancer cells identified in their tissues at the time of diagnosis and all patients appeared to have reduced spermatogonia numbers compared to healthy reference cohorts. WHAT IS KNOWN ALREADY Young boys affected by a hematological cancer are at risk of contamination of their testes by cancer cells but histological examination is unable to detect the presence of only a few cancer cells, which would preclude autotransplantation of cryobanked ITT for fertility restoration, and more sensitive detection techniques are thus required. Reduced numbers of spermatogonia in ITT in hematological cancer patients have been suggested based on results in a limited number of patients. STUDY DESIGN, SIZE, DURATION This retrospective cohort study included 54 pre- and peri-pubertal boys who were diagnosed with a hematological malignancy and who underwent a testicular biopsy for fertility preservation at the time of diagnosis before any gonadotoxic therapy between 2005 and 2021. PARTICIPANTS/MATERIALS, SETTING, METHODS Among the 54 patients eligible in our database, formalin-fixed paraffin-embedded (FFPE) testicular tissue was available for 28 boys diagnosed either with ALL (n = 14) or lymphoma (n = 14) and was used to evaluate malignant cell contamination. Hematoxylin and eosin (H&E) staining was performed for each patient to search for cancer cells in the tissue. Markers specific to each patient's disease were identified at the time of diagnosis on the biopsy of the primary tumor or bone marrow aspiration and an immunohistochemistry (IHC) was performed on the FFPE ITT for each patient to evidence his disease markers. PCR analyses on the FFPE tissue were also conducted when a specific gene rearrangement was available. MAIN RESULTS AND THE ROLE OF CHANCE The mean age at diagnosis and ITT biopsy of the 28 boys was 7.5 years (age range: 19 months-16 years old). Examination of ITT of the 28 boys on H&E stained sections did not detect malignant cells. Using IHC, we found contamination by cancerous cells using markers specific to the patient's disease in 10 of 28 boys, with a higher rate in patients diagnosed with ALL (57%, n = 8/14) compared with lymphoma (14%, n = 2/14) (P-value < 0.05). PCR showed contamination in three of 15 patients who had specific rearrangements identified on their bone marrow at the time of diagnosis; one of these patients had negative results from the IHC. Compared to age-related reference values of the number of spermatogonia per ST (seminiferous tubule) (Spg/ST) throughout prepuberty of healthy patients from a simulated control cohort, mean spermatogonial numbers appeared to be decreased in all age groups (0-4 years: 1.49 ± 0.54, 4-7 years: 1.08 ± 0.43, 7-11 years: 1.56 ± 0.65, 11-14 years: 3.37, 14-16 years: 5.44 ± 3.14). However, using a cohort independent method based on the Z-score, a decrease in spermatogonia numbers was not confirmed. LIMITATIONS, REASONS FOR CAUTION The results obtained from the biopsy fragments that were evaluated for contamination by cancer cells may not be representative of the entire cryostored ITT and tumor foci may still be present outside of the biopsy range. WIDER IMPLICATIONS OF THE FINDINGS ITT from boys diagnosed with a hematological malignancy could bear the risk for cancer cell reseeding in case of autotransplantation of the tissue. Such a high level of cancer cell contamination opens the debate of harvesting the tissue after one or two rounds of chemotherapy. However, as the safety of germ cells can be compromised by gonadotoxic treatments, this strategy warrants for the development of adapted fertility restoration protocols. Finally, the impact of the hematological cancer on spermatogonia numbers should be further explored. STUDY FUNDING/COMPETING INTEREST(S) The project was funded by a grant from the FNRS-Télévie (grant n°. 7.4533.20) and Fondation Contre le Cancer/Foundation Against Cancer (2020-121) for the research project on fertility restoration with testicular tissue from hemato-oncological boys. The authors declare that they have no conflict of interest. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Dhoha Kourta
- Laboratoire d'andrologie, Pôle de recherche en Physiologie de la Reproduction, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
- Department of Gynecology-Andrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Alessandra Camboni
- Pathology Department, Cliniques Universitaires Saint Luc, Brussels, Belgium
| | - Pascale Saussoy
- Department of Clinical Biology, Cliniques Universitaires Saint Luc, 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, Brussels, Belgium
- Department of Gynecology-Andrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Jonathan Poels
- Laboratoire d'andrologie, Pôle de recherche en Physiologie de la Reproduction, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, 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, Brussels, Belgium
- Department of Gynecology-Andrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| |
Collapse
|
16
|
Newman H, Hunger SP. Future of Treatment of Adolescents and Young Adults With ALL: A Vision for Collaboration and Equity. J Clin Oncol 2024; 42:665-674. [PMID: 37890130 DOI: 10.1200/jco.23.01351] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 08/03/2023] [Accepted: 08/30/2023] [Indexed: 10/29/2023] Open
Abstract
Over the past several decades, survival of children with ALL has improved dramatically with treatment regimens refined through cooperative group trials. Despite aggressive treatment and iterative therapy changes for adolescents and young adults (AYAs), improvement has not been as promising. Comparisons between pediatric and adult clinical trials have consistently demonstrated superior outcomes for AYAs treated on pediatric ALL protocols, leading to the implementation of pediatric-inspired ALL protocols by several groups worldwide and/or expansion of the age limit of pediatric trials to include the full spectrum of the AYA population. Despite these efforts, AYAs in both pediatric and adult settings continue to have inferior survival compared with younger children with ALL. Real-world data suggest that uptake of pediatric-style treatment is variable, and even with identical pediatric-style treatment, AYAs still fare worse than younger children. As we enter an era of immunotherapy and precision medicine for newly diagnosed ALL, now is an opportune time to consider how best to approach future therapy for AYA patients. Comparisons of pediatric and adult treatment approaches and subanalyses of AYA patients will help guide harmonization of treatment. The focus of the next stage of ALL therapy for AYA should not only involve novel treatment approaches but also standardization and optimization of supportive care measures, psychosocial support, adherence interventions, oncofertility treatment, and survivorship care. All these efforts should simultaneously work to address health disparities to ensure that a future of improved outcomes is experienced equitably for all AYA patients.
Collapse
Affiliation(s)
- Haley Newman
- Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA
- Division of Oncology, Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Stephen P Hunger
- Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA
- Division of Oncology, Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
17
|
Burns K, Loren AW. Fertility Preservation in Adolescents and Young Adults With Cancer: A Case-Based Review. J Clin Oncol 2024; 42:725-734. [PMID: 37976452 DOI: 10.1200/jco.23.01616] [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: 07/28/2023] [Revised: 09/08/2023] [Accepted: 10/03/2023] [Indexed: 11/19/2023] Open
Abstract
Adolescent and young adult (AYA) oncology patients are unique in many aspects of their care; fertility preservation (FP) is one of the most complex to address. In addition to the newly diagnosed AYA patient, there are growing numbers of AYA survivors of childhood cancer who present with concerns about their fertility. Emerging independence, emotional and intellectual growth, and development of an adult mindset are hallmarks of the AYA population; these transitions heighten the intrinsic medical, social, and financial challenges of a cancer diagnosis. FP is extraordinarily important in AYA oncology and can be addressed in many ways: experimental options as well as standard of care, with key differences on the basis of pubertal development, cancer diagnosis, and urgency of cancer-directed therapy. Options exist both at diagnosis and throughout the survivorship journey. It is imperative that oncologists recognize the challenges in this age group, as well as opportunities to pursue FP. The field has evolved significantly in the past 25 years and will continue to evolve as we incorporate more immune-based and targeted therapies into our treatment regimens. This case-based review will explore opportunities to preserve fertility in this unique patient population.
Collapse
Affiliation(s)
- Karen Burns
- University of Cincinnati College of Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Alison W Loren
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
18
|
Sönmezer M, Şükür YE, Saçıntı KG, Özkavukçu S, Kankaya D, Atabekoğlu CS, Cengiz Seval G, Oktay KH. Safety of ovarian cryopreservation and transplantation in patients with acute leukemia: a case series. Am J Obstet Gynecol 2024; 230:79.e1-79.e10. [PMID: 37666382 DOI: 10.1016/j.ajog.2023.08.032] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 08/14/2023] [Accepted: 08/29/2023] [Indexed: 09/06/2023]
Abstract
BACKGROUND With increased success, ovarian tissue cryopreservation has recently become a standard technique for fertility preservation. However, malignant cell introduction through ovarian tissue transplantation remains a major concern for patients with acute leukemias. OBJECTIVE This study aimed to investigate the safety of performing autologous ovarian tissue transplantation in survivors of acute leukemia. STUDY DESIGN Clinical, histopathological, and molecular data of 4 women with acute myeloid leukemia and 2 women with acute lymphoblastic leukemia who underwent ovarian tissue cryopreservation and transplantation were analyzed in this case series. Following cryopreservation of 66% to 100% of an ovarian cortex with a slow freezing method, all women received high-dose multiagent alkylating preconditioning chemotherapy for allogeneic hematopoietic stem cell transplantation. Before the ovarian tissue transplantation, (1) antral follicle counts, serum antimüllerian hormone and follicle-stimulating hormone levels were assessed to confirm primary ovarian insufficiency; (2) all recipients were cleared by their hematologist-oncologists; (3) representative cortical strips were screened for leukemia infiltration by histologic (hematoxylin and eosin staining), immunohistochemical (CD3, CD20, CD34, CD68, CD117, CD163, PAX-5, Tdt, lysozyme, and MPO), and molecular marker evaluation (BCR/ABL p190 and AML1/ETO) where appropriate. RESULTS The median age was 20 years (interquartile range, 15-32) at ovarian tissue cryopreservation. Before undergoing hematopoietic stem cell transplantation, all patients received induction or consolidation chemotherapy that included cytarabine + daunorubicin or Berlin-Frankfurt-Munich-95 protocol and were in remission. The mean serum antimüllerian hormone was 1.9±1.7 ng/mL before ovarian tissue cryopreservation. In all cases, ovarian tissue screening for leukemic cells was negative. Ovarian transplantation was performed laparoscopically with or without robotic assistance, after a median of 74.5 months (interquartile range, 41-120) after ovarian tissue cryopreservation. Ovarian function resumed in all patients after a median of 3.0 months (range, 2.5-4.0), and 2 women had 1 live birth each. The median graft longevity was 35.5 months (interquartile range, 18-57) after ovarian tissue transplantation. After a median follow-up of 51 months (interquartile range, 20-74), all patients remained relapse-free. In 1 patient, the graft was removed during cesarean delivery and was negative for immunochemical leukemia markers. CONCLUSION Our long-term follow-up demonstrated no evidence of disease relapse after ovarian tissue transplantation in patients with acute leukemia who received allogeneic hematopoietic stem cell transplantation. This safety profile may be explained by the fact that these patients are induced into remission by nongonadotoxic induction chemotherapy before undergoing ovarian tissue cryopreservation. We propose that ovarian tissue cryopreservation should not be excluded as a fertility preservation option for young women with leukemia who are due to receive preconditioning chemotherapy before allogeneic hematopoietic stem cell transplantation.
Collapse
Affiliation(s)
- Murat Sönmezer
- Faculty of Medicine, Departments of Obstetrics and Gynecology, Ankara University, Ankara, Turkey
| | - Yavuz Emre Şükür
- Faculty of Medicine, Departments of Obstetrics and Gynecology, Ankara University, Ankara, Turkey
| | - Koray Görkem Saçıntı
- Faculty of Medicine, Departments of Obstetrics and Gynecology, Ankara University, Ankara, Turkey; Faculty of Medicine, Division of Epidemiology, Department of Public Health, Hacettepe University, Ankara, Turkey
| | - Sinan Özkavukçu
- Assisted Conception Unit, Postgraduate Medicine, Ninewells Hospital, School of Medicine, University of Dundee, Dundee, United Kingdom
| | | | - Cem Somer Atabekoğlu
- Faculty of Medicine, Departments of Obstetrics and Gynecology, Ankara University, Ankara, Turkey
| | | | - Kutluk H Oktay
- Department of Obstetrics, Gynecology and Reproductive Sciences, Laboratory of Molecular Reproduction and Fertility Preservation, Yale University School of Medicine, New Haven, CT; Innovation Institute for Fertility Preservation, New Haven, CT, and New York, NY.
| |
Collapse
|
19
|
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
|
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: 5] [Impact Index Per Article: 5.0] [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
|
Silvestris E, Minoia C, De Palma G, Popescu O, Altavilla A, Guarini A, Pavone F, Loizzi V, Cormio G, Depalo R. Optimizing the Ovarian Tissue Cryopreservation in the 'Oncofertility' Institutional Program at an Italian National Cancer Institute. Healthcare (Basel) 2023; 11:2727. [PMID: 37893801 PMCID: PMC10606252 DOI: 10.3390/healthcare11202727] [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: 07/04/2023] [Revised: 10/01/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND The majority of female cancer patients undergoing anticancer treatments are at risk of experiencing 'cancer treatment-related infertility', which can result in permanent damage to their reproductive prospects. Among the fertility preservation methods, ovarian tissue cryopreservation (OTC) has emerged as an alternative for these patients. The Cancer Institute of Bari initiated a research program to assess the feasibility of OTC. This study compares the viability of ovarian cortical fragments cryopreserved using slow freezing (SF) and ultra-rapid freezing (URF) methods. METHODS Ovarian cortex biopsies were obtained from 11 fertile women enrolled in our oncofertility service between June 2022 and January 2023. After tissue collection, a histological assessment was performed before cryopreservation. OTC was carried out using both SF and URF methods. Six months later, thawed samples were evaluated for follicle counts and histological integrity. RESULTS No statistically significant difference was observed in the proportion of intact follicles (means of 31.5% and 73.0% in the SF and URF groups, respectively; p = 0.064). However, there was a significant difference in the number of follicles between the SF group (n = 149) and the URF group (n = 37) (p = 0.046). CONCLUSIONS We assessed the viability of ovarian cortex after freezing and thawing, focusing on the structural integrity of follicles. Our findings suggest that there are no significant differences between the SF and URF methods.
Collapse
Affiliation(s)
- Erica Silvestris
- Gynecologic Oncology Unit, IRCCS Istituto Tumori “Giovanni Paolo II” Bari, 70124 Bari, Italy; (V.L.); (G.C.)
| | - Carla Minoia
- Hematology Unit, IRCCS Istituto Tumori “Giovanni Paolo II” Bari, 70124 Bari, Italy; (C.M.); (A.G.); (F.P.)
| | - Giuseppe De Palma
- Institutional BioBank, Experimental Oncology and Biobank Management Unit, IRCCS Istituto Tumori “Giovanni Paolo II” Bari, 70124 Bari, Italy;
| | - Ondina Popescu
- Pathological Anatomy Unit, IRCCS Istituto Tumori “Giovanni Paolo II” Bari, 70124 Bari, Italy; (O.P.); (A.A.)
| | - Anna Altavilla
- Pathological Anatomy Unit, IRCCS Istituto Tumori “Giovanni Paolo II” Bari, 70124 Bari, Italy; (O.P.); (A.A.)
| | - Attilio Guarini
- Hematology Unit, IRCCS Istituto Tumori “Giovanni Paolo II” Bari, 70124 Bari, Italy; (C.M.); (A.G.); (F.P.)
| | - Fabio Pavone
- Hematology Unit, IRCCS Istituto Tumori “Giovanni Paolo II” Bari, 70124 Bari, Italy; (C.M.); (A.G.); (F.P.)
| | - Vera Loizzi
- Gynecologic Oncology Unit, IRCCS Istituto Tumori “Giovanni Paolo II” Bari, 70124 Bari, Italy; (V.L.); (G.C.)
- Department of Interdisciplinary Medicine (DIM), University of Bari “Aldo Moro”, 70121 Bari, Italy
| | - Gennaro Cormio
- Gynecologic Oncology Unit, IRCCS Istituto Tumori “Giovanni Paolo II” Bari, 70124 Bari, Italy; (V.L.); (G.C.)
- Department of Interdisciplinary Medicine (DIM), University of Bari “Aldo Moro”, 70121 Bari, Italy
| | - Raffaella Depalo
- Institutional BioBank, Experimental Oncology and Biobank Management Unit, IRCCS Istituto Tumori “Giovanni Paolo II” Bari, 70124 Bari, Italy;
| |
Collapse
|
22
|
Grubliauskaite M, van der Perk MEM, Bos AME, Meijer AJM, Gudleviciene Z, van den Heuvel-Eibrink MM, Rascon J. Minimal Infiltrative Disease Identification in Cryopreserved Ovarian Tissue of Girls with Cancer for Future Use: A Systematic Review. Cancers (Basel) 2023; 15:4199. [PMID: 37686475 PMCID: PMC10486797 DOI: 10.3390/cancers15174199] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/07/2023] [Accepted: 08/09/2023] [Indexed: 09/10/2023] Open
Abstract
BACKGROUND Ovarian tissue cryopreservation and transplantation are the only available fertility techniques for prepubertal girls with cancer. Though autotransplantation carries a risk of reintroducing malignant cells, it can be avoided by identifying minimal infiltrative disease (MID) within ovarian tissue. METHODS A broad search for peer-reviewed articles in the PubMed database was conducted in accordance with PRISMA guidelines up to March 2023. Search terms included 'minimal residual disease', 'cryopreservation', 'ovarian', 'cancer' and synonyms. RESULTS Out of 542 identified records, 17 were included. Ovarian tissues of at least 115 girls were evaluated and categorized as: hematological malignancies (n = 56; 48.7%), solid tumors (n = 42; 36.5%) and tumors of the central nervous system (n = 17; 14.8%). In ovarian tissue of 25 patients (21.7%), MID was detected using RT-qPCR, FISH or multicolor flow cytometry: 16 of them (64%) being ALL (IgH rearrangements with/without TRG, BCL-ABL1, EA2-PBX1, TEL-AML1 fusion transcripts), 3 (12%) Ewing sarcoma (EWS-FLI1 fusion transcript, EWSR1 rearrangements), 3 (12%) CML (BCR-ABL1 fusion transcript, FLT3) and 3 (12%) AML (leukemia-associated immunophenotypes, BCR-ABL1 fusion transcript) patients. CONCLUSION While the majority of malignancies were found to have a low risk of containing malignant cells in ovarian tissue, further studies are needed to ensure safe implementation of future fertility restoration in clinical practice.
Collapse
Affiliation(s)
- Monika Grubliauskaite
- Center for Pediatric Oncology and Hematology, Vilnius University Hospital Santaros Klinikos, Santariskiu Str. 4, LT-08406 Vilnius, Lithuania
- Life Sciences Center, Vilnius University, Sauletekio Ave. 7, LT-10257 Vilnius, Lithuania
- Department of Biobank, National Cancer Institute, Santariskiu Str. 1, LT-08406 Vilnius, Lithuania
| | | | - Annelies M. E. Bos
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands
- Department of Reproductive Medicine, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | | | - Zivile Gudleviciene
- Faculty of Medicine, Vilnius University, M. K. Ciurlionio Str. 21/27, LT-03101 Vilnius, Lithuania
| | - Marry M. van den Heuvel-Eibrink
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands
- Division of Child Health, UMCU-Wilhelmina Children’s Hospital, 3584 EA Utrecht, The Netherlands
| | - Jelena Rascon
- Center for Pediatric Oncology and Hematology, Vilnius University Hospital Santaros Klinikos, Santariskiu Str. 4, LT-08406 Vilnius, Lithuania
- Faculty of Medicine, Vilnius University, M. K. Ciurlionio Str. 21/27, LT-03101 Vilnius, Lithuania
| |
Collapse
|
23
|
Sheikh S, Lo BKM, Kaune H, Bansal J, Deleva A, Williams SA. Rescue of follicle development after oocyte-induced ovary dysfunction and infertility in a model of POI. Front Cell Dev Biol 2023; 11:1202411. [PMID: 37614224 PMCID: PMC10443433 DOI: 10.3389/fcell.2023.1202411] [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: 04/08/2023] [Accepted: 07/20/2023] [Indexed: 08/25/2023] Open
Abstract
The mechanisms and aetiology underlying the development of premature ovarian insufficiency (POI) are poorly understood. However, the oocyte clearly has a role as demonstrated by the Double Mutant (DM) mouse model where ovarian dysfunction (6 weeks) is followed by POI (3 months) due to oocyte-specific deletion of complex and hybrid N- and O-glycans. The ovaries of DM mice contain more primary follicles (3a stage) accompanied by fewer developing follicles, indicating a block in follicle development. To investigate this block, we first analysed early follicle development in postnatal (8-day), pre-pubertal (3-week) and post-pubertal (6-week and 3-month) DM (C1galt1 F/F Mgat1 F/F:ZP3Cre) and Control (C1galt1 F/F Mgat1 F/F) mice. Second, we investigated if transplantation of DM ovaries into a "normal" endocrine environment would restore follicle development. Third, we determined if replacing DM ovarian somatic cells would rescue development of DM oocytes. At 3-week, DM primary 3a follicles contain large oocytes accompanied by early development of a second GC layer and increased GC proliferation. At 6-week, DM primary 3a follicles contain abnormally large oocytes, accompanied with decreased GC proliferation. Transplantation of DM ovaries into a 'normal' endocrine environment did not restore normal follicle development. However, replacing somatic cells by generating reaggregated ovaries (ROs) did enable follicle development to progress and thus highlighted intra-ovarian factors were responsible for the onset of POI in DM females. Thus, these studies demonstrate oocyte-initiated altered communication between GCs and oocytes results in abnormal primary follicles which fail to progress and leads to POI.
Collapse
Affiliation(s)
| | | | | | | | | | - Suzannah A. Williams
- Nuffield Department of Women’s and Reproductive Health, Women’s Centre, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
24
|
Chen L, Dong Z, Chen X. Fertility preservation in pediatric healthcare: a review. Front Endocrinol (Lausanne) 2023; 14:1147898. [PMID: 37206440 PMCID: PMC10189781 DOI: 10.3389/fendo.2023.1147898] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 04/12/2023] [Indexed: 05/21/2023] Open
Abstract
Survival rates for children and adolescents diagnosed with malignancy have been steadily increasing due to advances in oncology treatments. These treatments can have a toxic effect on the gonads. Currently, oocyte and sperm cryopreservation are recognized as well-established and successful strategies for fertility preservation for pubertal patients, while the use of gonadotropin-releasing hormone agonists for ovarian protection is controversial. For prepubertal girls, ovarian tissue cryopreservation is the sole option. However, the endocrinological and reproductive outcomes after ovarian tissue transplantation are highly heterogeneous. On the other hand, immature testicular tissue cryopreservation remains the only alternative for prepubertal boys, yet it is still experimental. Although there are several published guidelines for navigating fertility preservation for pediatric and adolescent patients as well as transgender populations, it is still restricted in clinical practice. This review aims to discuss the indications and clinical outcomes of fertility preservation. We also discuss the probably effective and efficient workflow to facilitate fertility preservation.
Collapse
Affiliation(s)
- Lin Chen
- Reproductive Medical Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zirui Dong
- Department of Obstetrics and Gynecology, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - Xiaoyan Chen
- Maternal-Fetal Medicine Institute, Shenzhen Baoan Women’s and Children’s Hospital, Shenzhen University, Shenzhen, China
- The Fertility Preservation Research Center, Department of Obstetrics and Gynecology, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| |
Collapse
|
25
|
Shirai R, Osumi T, Keino D, Nakabayashi K, Uchiyama T, Sekiguchi M, Hiwatari M, Yoshida M, Yoshida K, Yamada Y, Tomizawa D, Takae S, Kiyokawa N, Matsumoto K, Yoshioka T, Hata K, Hori T, Suzuki N, Kato M. Minimal residual disease detection by mutation-specific droplet digital PCR for leukemia/lymphoma. Int J Hematol 2023; 117:910-918. [PMID: 36867356 DOI: 10.1007/s12185-023-03566-2] [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: 10/12/2022] [Revised: 02/13/2023] [Accepted: 02/14/2023] [Indexed: 03/04/2023]
Abstract
Minimal residual disease (MRD) is usually defined as the small number of cancer cells that remain in the body after treatment. The clinical significance of MRD kinetics is well recognized in treatment of hematologic malignancies, particularly acute lymphoblastic leukemia (ALL). Real time quantitative PCR targeting immunoglobulin (Ig) or T-cell receptor (TCR) rearrangement (PCR-MRD), as well as multiparametric flow cytometric analysis targeting antigen expression, are widely used in MRD detection. In this study, we devised an alternative method to detect MRD using droplet digital PCR (ddPCR), targeting somatic single nucleotide variants (SNVs). This ddPCR-based method (ddPCR-MRD) had sensitivity up to 1E-4. We assessed ddPCR-MRD at 26 time points from eight T-ALL patients, and compared it to the results of PCR-MRD. Almost all results were concordant between the two methods, but ddPCR-MRD detected micro-residual disease that was missed by PCR-MRD in one patient. We also measured MRD in stored ovarian tissue of four pediatric cancer patients, and detected 1E-2 of submicroscopic infiltration. Considering the universality of ddPCR-MRD, the methods can be used as a complement for not only ALL, but also other malignant diseases regardless of tumor-specific Ig/TCR or surface antigen patterns.
Collapse
Affiliation(s)
- Ryota Shirai
- Department of Pediatric Hematology and Oncology Research, National Research Institute for Child Health and Development, Tokyo, Japan.,Department of Pediatrics, Yokohama City University Graduate School of Medicine, Kanagawa, Japan
| | - Tomoo Osumi
- Department of Pediatric Hematology and Oncology Research, National Research Institute for Child Health and Development, Tokyo, Japan.,Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan
| | - Dai Keino
- Department of Pediatrics, St. Marianna University School of Medicine Hospital, Kawasaki, Japan.,Division of Hematology/Oncology, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Kazuhiko Nakabayashi
- Department of Maternal-Fetal Biology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Toru Uchiyama
- Department of Human Genetics, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Masahiro Sekiguchi
- Department of Pediatrics, the University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan
| | - Mitsuteru Hiwatari
- Department of Pediatrics, the University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan.,Department of Pediatrics, School of Medicine, Teikyo University, Tokyo, Japan
| | - Masanori Yoshida
- Department of Pediatric Hematology and Oncology Research, National Research Institute for Child Health and Development, Tokyo, Japan.,Department of Pediatrics, Yokohama City University Graduate School of Medicine, Kanagawa, Japan
| | - Kaoru Yoshida
- Department of Pediatric Hematology and Oncology Research, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Yuji Yamada
- Department of Pediatric Hematology and Oncology Research, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Daisuke Tomizawa
- Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan
| | - Seido Takae
- Department of Obstetrics and Gynecology, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Nobutaka Kiyokawa
- Department of Pediatric Hematology and Oncology Research, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Kimikazu Matsumoto
- Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan
| | - Takako Yoshioka
- Department of Pathology, National Center for Child Health and Development, Tokyo, Japan
| | - Kenichiro Hata
- Department of Maternal-Fetal Biology, National Research Institute for Child Health and Development, Tokyo, Japan.,Department of Human Molecular Genetics, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Toshinori Hori
- Department of Pediatrics, Aichi Medical University, Nagakute, Japan
| | - Nao Suzuki
- Department of Obstetrics and Gynecology, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Motohiro Kato
- Department of Pediatric Hematology and Oncology Research, National Research Institute for Child Health and Development, Tokyo, Japan. .,Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan. .,Department of Pediatrics, the University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan.
| |
Collapse
|
26
|
Wu M, Xue L, Guo Y, Dong X, Chen Z, Wei S, Yi X, Li Y, Zhang J, Zhou S, Wu M, Lou X, Dai J, Xia F, Wang S. Microenvironmentally Responsive Chemotherapeutic Prodrugs and CHEK2 Inhibitors Self-Assembled Micelles: Protecting Fertility and Enhancing Chemotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2210017. [PMID: 36528787 DOI: 10.1002/adma.202210017] [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: 10/30/2022] [Revised: 11/30/2022] [Indexed: 06/17/2023]
Abstract
Chemotherapy is a widely used and effective adjuvant treatment for cancer, and it has unavoidable damage to female fertility, with statistics showing 38% of women who have received chemotherapy are infertile. How to reduce fertility toxicity while enhancing the oncologic chemotherapy is a clinical challenge. Herein, co-delivery micelles (BML@PMP) are developed, which are composed of a reduction-sensitive paclitaxel prodrug (PMP) for chemotherapy and a CHEK2 inhibitor (BML277) for both fertility protection and chemotherapy enhancement. BML@PMP achieves fertility protection through three actions: (1) Due to the enhanced permeability and retention (EPR) effect, BML@PMP is more enriched in the tumor, while very little in the ovary (about 1/10th of the tumor). (2) Glutathione (GSH) triggers the release of PTX, and with low levels of GSH in the ovary, the amount of PTX released in the ovary is correspondingly reduced. (3) BML277 inhibits oocyte apoptosis by inhibiting the CHEK2-TAp63α pathway. Because of the different downstream targets of CHEK2 in tumor cells and oocytes, BML277 also enhances chemotherapeutic efficacy by reducing DNA damage repair which is activated through the CHEK2 pathway. This bidirectional effect of CHEK2 inhibitor-based co-delivery system represents a promising strategy for improving oncology treatment indices and preventing chemotherapy-associated fertility damage.
Collapse
Affiliation(s)
- Meng Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, 430030, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, 430030, China
| | - Liru Xue
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, 430030, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, 430030, China
| | - Yican Guo
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, 430030, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, 430030, China
| | - Xiaoqi Dong
- State Key Laboratory of Biogeology and Environmental Geology, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan, 430074, China
| | - Zhaojun Chen
- State Key Laboratory of Biogeology and Environmental Geology, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan, 430074, China
| | - Simin Wei
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, 430030, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, 430030, China
| | - Xiaoqing Yi
- Key Laboratory of Biomaterials and Biofabrication in Tissue Engineering of Jiangxi Province, Gannan Medical University, Ganzhou, 341000, China
| | - Yinuo Li
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, 430030, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, 430030, China
| | - Jinjin Zhang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, 430030, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, 430030, China
| | - Su Zhou
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, 430030, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, 430030, China
| | - Mingfu Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, 430030, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, 430030, China
| | - Xiaoding Lou
- State Key Laboratory of Biogeology and Environmental Geology, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan, 430074, China
| | - Jun Dai
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, 430030, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, 430030, China
| | - Fan Xia
- State Key Laboratory of Biogeology and Environmental Geology, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan, 430074, China
| | - Shixuan Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, 430030, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, 430030, China
| |
Collapse
|
27
|
Labrune E, Bianchetti S, Lepinasse O, Soignon G, Salle B, Lornage J. When to cryopreserve ovarian tissue: Determining the effects of chemotherapy on the ovarian reserve by studying follicular density and apoptosis. Cytopathology 2023; 34:146-153. [PMID: 36458472 PMCID: PMC10107618 DOI: 10.1111/cyt.13194] [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: 11/08/2022] [Accepted: 11/25/2022] [Indexed: 12/04/2022]
Abstract
OBJECTIVES Patients scheduled to receive chemotherapy should be counselled on fertility preservation. Known gonadotoxic chemotherapies such as alkylating agents have a high risk of altering ovarian reserve. In some cases, the urgency of treatment requires the use of chemotherapy before fertility preservation, which will be carried out at a later stage. Usually the ovarian tissue is cryopreserved. The aim of our study is to investigate the impact of chemotherapies on follicular density and the apoptosis of reserve follicles. METHODS We included 140 patients: 63 patients, mean age 18.8 years, were included in the group "no chemotherapy" (group A) and 77 patients, mean age 17.1 years, in the group "received chemotherapy before ovarian conservation" (group B). None of the patients had had pelvic radiotherapy prior to ovarian cryopreservation. The histological parameters studied were follicular density and the presence of malignant cells. We selected 12 patients from group A and 15 patients from group B, comparable in age and pathology, for whom we evaluated follicle apoptosis by immunostaining cleaved caspase-3. RESULTS We demonstrated an inverse relationship between follicular density and age (p < 0.0001), as well as a lack of effect of chemotherapy on follicular density (p = 0.87). We showed the impact of various chemotherapies, especially with alkylating agents, on the apoptosis of ovarian follicles (p < 0.0001). Three patients had ovarian tissue infiltration, two of which were malignant. CONCLUSION This work underlines the fact that conservation of ovarian tissue after chemotherapy remains possible.
Collapse
Affiliation(s)
- Elsa Labrune
- Hospices Civils de Lyon, Hôpital Mère Enfant, Service de Médecine de la Reproduction, Bron, France.,Université Claude Bernard, Faculté de Médecine Laennec, Lyon Cedex 08, France.,INSERM Unité 1208, Bron Cedex, France
| | - Serge Bianchetti
- Hospices Civils de Lyon, Hôpital Mère Enfant, Service de Médecine de la Reproduction, Bron, France.,Université Claude Bernard, Faculté de Médecine Laennec, Lyon Cedex 08, France.,INSERM Unité 1208, Bron Cedex, France
| | - Odile Lepinasse
- Hospices Civils de Lyon, Hôpital Mère Enfant, Service de Médecine de la Reproduction, Bron, France.,INSERM Unité 1208, Bron Cedex, France
| | - Gaelle Soignon
- Hospices Civils de Lyon, Hôpital Mère Enfant, Service de Médecine de la Reproduction, Bron, France
| | - Bruno Salle
- Hospices Civils de Lyon, Hôpital Mère Enfant, Service de Médecine de la Reproduction, Bron, France.,INSERM Unité 1208, Bron Cedex, France.,Université Claude Bernard, Faculté de Médecine Lyon Sud, Oullins Cedex, France
| | - Jacqueline Lornage
- Hospices Civils de Lyon, Hôpital Mère Enfant, Service de Médecine de la Reproduction, Bron, France.,INSERM Unité 1208, Bron Cedex, France.,Université Claude Bernard, Faculté de Médecine Lyon Sud, Oullins Cedex, France
| |
Collapse
|
28
|
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
|
29
|
Yildiz S, Bildik G, Benlioglu C, Turan V, Dilege E, Ozel M, Kim S, Oktem O. Breast cancer treatment and ovarian function. Reprod Biomed Online 2023; 46:313-331. [PMID: 36400663 DOI: 10.1016/j.rbmo.2022.09.014] [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: 06/13/2022] [Revised: 09/14/2022] [Accepted: 09/14/2022] [Indexed: 02/07/2023]
Abstract
The aim of this study was to provide an update on ovarian function and the mechanisms of gonadal damage after exposure to chemotherapy in breast cancer survivors. The alkylating agents are toxic to both primordial and growing follicles. However, anti-metabolite drugs are more likely to destroy preantral and antral follicles. Younger patients are more likely to have a higher ovarian reserve, and therefore, more likely to retain some residual ovarian function after exposure to gonadotoxic regimens. However, there can be significant variability in ovarian reserve among patients of the same age. Furthermore, patients with critically diminished ovarian reserve may continue to menstruate regularly. Therefore age and menstrual status are not reliable indicators of good ovarian reserve and might give a false sense of security and result in an adverse outcome if the patient is consulted without considering more reliable quantitative markers of ovarian reserve (antral follicle count and anti-Müllerian hormone) and fertility preservation is not pursued. In contrast to well-documented ovarian toxicity of older chemotherapy regimens, data for newer taxane-containing protocols have only accumulated in the last decade and data are still very limited regarding the impact of targeted therapies on ovarian function.
Collapse
Affiliation(s)
- Sule Yildiz
- The Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Koç University Hospital, Koç University School of Medicine, Istanbul, Turkey
| | - Gamze Bildik
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston TX 77054, USA
| | - Can Benlioglu
- Department of Obstetrics and Gynecology, Koç University Hospital, Istanbul, Turkey
| | - Volkan Turan
- Istanbul Tema Hospital, Assisted Reproduction Unit, Istanbul
| | - Ece Dilege
- Department of General Surgery, Koç University Hospital, Koç University School of Medicine, Istanbul, Turkey
| | - Melis Ozel
- Department of Gynecology and Obstetrics Klinikum Ingolstadt, Bavaria, Germany
| | - Samuel Kim
- Eden Centers for Advanced Fertility, Fullerton CA 92835, USA
| | - Ozgur Oktem
- The Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Koç University Hospital, Koç University School of Medicine, Istanbul, Turkey.
| |
Collapse
|
30
|
Babayev E, Xu M, Shea LD, Woodruff TK, Duncan FE. Follicle isolation methods reveal plasticity of granulosa cell steroidogenic capacity during mouse in vitro follicle growth. Mol Hum Reprod 2022; 28:6693628. [PMID: 36069625 PMCID: PMC9802420 DOI: 10.1093/molehr/gaac033] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 07/26/2022] [Indexed: 01/07/2023] Open
Abstract
Follicles are the functional unit of the ovary and several methods have been developed to grow follicles ex vivo, which recapitulate key events of oogenesis and folliculogenesis. Enzymatic digestion protocols are often used to increase the yield of follicles from the ovary. However, the impact of these protocols on the outermost theca and granulosa cells, and thereby follicle function, is not well defined. To investigate the impact of enzymatic digestion on follicle function, we collected preantral follicles from CD1 mice either by enzymatic digestion (Enzy-FL) or mechanical isolation (Mech-FL) and compared follicle growth, steroidogenesis and cell differentiation within an encapsulated in vitro follicle growth system which maintains the 3D architecture of the oocyte and its surrounding somatic cells. Follicles were encapsulated in 0.5% alginate and cultured for 8 days. Compared with Enzy-FL, Mech-FL grew more rapidly and produced significantly higher levels of androstenedione, estradiol and progesterone. The expression of theca-interstitial cell marker genes, Cyp17a1, which encodes 17-hydroxylase/17, 20-lyase and catalyzes the hydroxylation of pregnenolone and progesterone to 17-hydroxypregnenolone and 17-hydroxyprogesterone, and the conversion of these products into dehydroepiandrosterone and androstenedione, and Star, which encodes a transport protein essential for cholesterol entry into mitochondria, were also higher in Mech-FL than in Enzy-FL. Mech-FL maintained an intact theca-interstitial layer on the outer edge of the follicle that phenocopied in vivo patterns as confirmed by alkaline phosphatase staining, whereas theca-interstitial cells were absent from Enzy-FL from the onset of culture. Therefore, preservation of the theca cell layer at the onset of culture better supports follicle growth and function. Interestingly, granulosa cells in the outermost layers of Enzy-FL expressed CYP17A1 by Day 4 of culture while maintaining inhibin α-subunit expression and a cuboidal nucleus. Thus, in the absence of theca-interstitial cells, granulosa cells have the potential to differentiate into androgen-producing cells. This work may have implications for human follicle culture, where enzymatic isolation is required owing to the density of the ovarian cortex.
Collapse
Affiliation(s)
| | | | - Lonnie D Shea
- Member of the Oncofertility Consortium, Michigan State University, East Lansing, MI, USA,Institute of Bionanotechnology in Medicine, Northwestern University, Chicago, IL, USA,Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL, USA
| | - Teresa K Woodruff
- Correspondence address. Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, 303 E. Superior Street, Lurie 10-109, Chicago, IL 60611, USA. E-mail: (F.E.D.); Department of Obstetrics and Gynecology and Department of Biomedical Engineering, Michigan State University, 965 Wilson Road, Room A626B, East Lansing, MI 48824-1316, USA. E-mail: (T.K.W.)
| | - Francesca E Duncan
- Correspondence address. Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, 303 E. Superior Street, Lurie 10-109, Chicago, IL 60611, USA. E-mail: (F.E.D.); Department of Obstetrics and Gynecology and Department of Biomedical Engineering, Michigan State University, 965 Wilson Road, Room A626B, East Lansing, MI 48824-1316, USA. E-mail: (T.K.W.)
| |
Collapse
|
31
|
Arapaki A, Christopoulos P, Kalampokas E, Triantafyllidou O, Matsas A, Vlahos NF. Ovarian Tissue Cryopreservation in Children and Adolescents. CHILDREN (BASEL, SWITZERLAND) 2022; 9:1256. [PMID: 36010146 PMCID: PMC9406615 DOI: 10.3390/children9081256] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 07/29/2022] [Accepted: 08/12/2022] [Indexed: 11/16/2022]
Abstract
Cancer during childhood and adolescence remains a major public health issue, affecting a significant portion of this age group. Although newer anti-cancer treatments have improved survival rates, this comes at a cost in terms of gonadotoxic effects. As a result, the preservation of fertility is important. Ovarian tissue cryopreservation, one of the newest methods, has some advantages, especially for prepubertal patients: no need for ovarian stimulation, thus, no further risk for estrogen-sensitive cancer types, and preservation of more and better-quality primordial follicles of the ovarian cortex. The most frequent indications include treatment with alkylating agents, ovarian-focused radiotherapy, leukemias, lymphomas, brain and neurological tumors, as well as Turner syndrome and benign hemoglobinopathies. An expected survival exceeding 5 years, the absence of systematic disease and an overall risk of premature ovarian insufficiency over 50% are among the criteria that need to be fulfilled in order for a patient to undertake this method. Orthotopic transplantation is more frequently used, since it can allow both live birth and the recovery of endocrine function. Reimplantation of malignant cells is always a major risk and should always be taken into consideration. Histological analysis, as well as immunohistochemical and molecular methods, are needed in order to improve the search for malignant cells before transplantation. Ovarian tissue cryopreservation appears to be a method with specific benefits, indications and risks which can be an important tool in terms of preserving fertility in younger women.
Collapse
Affiliation(s)
| | - Panagiotis Christopoulos
- Second Department of Obstetrics and Gynecology, “Aretaieion” Hospital, Faculty of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | | | | | | | - Nikolaos F. Vlahos
- Second Department of Obstetrics and Gynecology, “Aretaieion” Hospital, Faculty of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece
| |
Collapse
|
32
|
Dolmans MM, Demeestere I, Anckaert E, De Vos M. Proceedings of the Oncofertility Congress of the "Freezing Ovarian Tissue and Oocytes" (FOTO) Consortium Brussels. J Assist Reprod Genet 2022; 39:1715-1725. [PMID: 35751830 PMCID: PMC9428079 DOI: 10.1007/s10815-022-02552-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 06/15/2022] [Indexed: 01/19/2023] Open
Affiliation(s)
- Marie-Madeleine Dolmans
- Gynecology Department, Cliniques Universitaires St-Luc, Avenue Hippocrate 10, 1200, Brussels, Belgium
- Gynecology Research Laboratory, Institut de Recherche Expérimentale Et Clinique, Université Catholique de Louvain, Avenue Mounier 52, bte B1.52.02, 1200, Brussels, Belgium
| | - Isabelle Demeestere
- Research Laboratory On Human Reproduction, Fertility Clinic, CUB-Hôpital Erasme, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Ellen Anckaert
- Follicle Biology Laboratory, Vrije Universiteit Brussel (VUB), 1090, Brussels, Belgium
| | - Michel De Vos
- Follicle Biology Laboratory, Vrije Universiteit Brussel (VUB), 1090, Brussels, Belgium.
- Brussels IVF, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Laarbeeklaan 101, 1090, Brussels, Belgium.
| |
Collapse
|
33
|
Schallmoser A, Einenkel R, Färber C, Sänger N. In vitro growth (IVG) of human ovarian follicles in frozen thawed ovarian cortex tissue culture supplemented with follicular fluid under hypoxic conditions. Arch Gynecol Obstet 2022; 306:1299-1311. [PMID: 35871693 PMCID: PMC9470640 DOI: 10.1007/s00404-022-06672-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 06/14/2022] [Indexed: 12/20/2022]
Abstract
Background Despite its clinical success rates, transplantation after ovarian tissue cryopreservation (OTC) remains a matter of concern. Certain cancer subtypes may lead to the transfer of malignant cells when transplantation of affected ovarian tissue is conducted. IVG and subsequent isolation of vital follicles obtained from frozen thawed ovarian tissue for further in vitro maturation (IVM) would expand current fertility protection techniques while reducing the risk of retransplanting malignant cells. Methods A total of 216 cortical biopsies from 3 patients were included in this study in 4 treatment groups. After freezing, thawing and 8 days of hypoxic tissue culture supplemented with different concentrations of human follicular fluid (HuFF) and follicle-stimulating hormone (FSH), follicles were isolated enzymatically and stained with calcein to determine follicular viability. Numbers and size of vital follicles were assessed by fluorescence microscopy (Ti2, Nikon) and specified by computer assisted, semi-automated measurement (NIS software, Nikon). To estimate the effect of in vitro culture on apoptosis, tissue sections were stained for nicked DNA (TUNEL) prior and after tissue culture. Results Analysing 3025 vital follicles, we observed significant differences [P < 0.01] regarding follicle size when hypoxic tissue culture was supplemented with HuFF compared with the control group on day 1, individual follicles reached sizes > 100 µm. Conclusions The results implicate that HuFF contains valuable factors contributing to significant IVG of follicles in human ovarian tissue and could be regarded as an additional tool in personalized fertility restoration prior to retransplantation of ovarian tissue.
Collapse
Affiliation(s)
- Andreas Schallmoser
- Department of Gynecological Endocrinology and Reproductive Medicine, University Hospital of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Rebekka Einenkel
- Department of Gynecological Endocrinology and Reproductive Medicine, University Hospital of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Cara Färber
- Department of Gynecological Endocrinology and Reproductive Medicine, University Hospital of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Nicole Sänger
- Department of Gynecological Endocrinology and Reproductive Medicine, University Hospital of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| |
Collapse
|
34
|
Effects of Chemotherapy on Fertility Preservation in Patients with Tumors of the Hematopoietic and Lymphoid Tissues. REPRODUCTIVE MEDICINE 2022. [DOI: 10.3390/reprodmed3020012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Fertility preservation is an important concern for young cancer patients. Oocyte or embryo cryopreservation prior to chemotherapy administration is desirable but often difficult for patients with hematopoietic and lymphoid tissue tumors. In this study, we examined the results of fertility preservation therapy in patients with hematopoietic and lymphoid tissue tumors. We retrospectively examined hematopoietic and lymphoid tissue tumors of five patients who underwent oocyte cryopreservation as a fertility preservation therapy after chemotherapy, at Showa University Hospital from February 2017 to September 2020. Eleven treatment cycles were administered (one of which was cancelled). The mean age of the patients was 28.6 years. The mean controlled ovarian stimulation duration for 10 cycles was 15.9 days, the mean total gonadotropin dose was 3705 IU, and the mean peak E2 was 502.8 (pg/mL). The mean number of eggs retrieved was 3.2, the mean number of mature oocytes was 2.1, and the mean maturation rate (mature oocytes/returned oocytes) was 70.7%. Fertility preservation procedures in the early period after chemotherapy may be viable because they allow for the acquisition of mature oocytes, even though the procedures may take longer and yield fewer oocytes.
Collapse
|
35
|
Leflon M, Rives-Feraille A, Letailleur M, Petrovic CH, Martin B, Marpeau L, Jardin F, Aziz M, Stamatoulas-Bastard A, Dumont L, Rondanino C, Rives N. Experience, gynaecological and reproductive health follow up of young adult women who have undergone ovarian tissue cryopreservation. Reprod Biomed Online 2022; 45:913-922. [DOI: 10.1016/j.rbmo.2022.06.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 06/14/2022] [Accepted: 06/17/2022] [Indexed: 11/17/2022]
|
36
|
Gomes FDR, de Brito DCC, de Sá NAR, Ñaupas LVS, Palomino GJQ, da Silva RF, Lopes ÉPF, Mbemya GT, Alves BG, Zelinski M, de Figueiredo JR, Rodrigues APR. Development of sheep secondary follicles and preservation of aromatase and metalloproteinases 2 and 9 after vitrification and in vitro culture. Cell Tissue Bank 2022; 23:247-259. [PMID: 34152507 DOI: 10.1007/s10561-021-09937-5] [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: 02/03/2021] [Accepted: 05/28/2021] [Indexed: 12/24/2022]
Abstract
The cryopreservation of secondary follicles (SF) is a promising alternative to preserve the reproductive potential both in humans and animals in situations in which the transplantation of ovarian tissue is not possible. The objective of the present study was cryopreserved SF isolated sheep. Beyond follicular morphology, viability and development, we investigated proteins related to steroidogenic function and basement membrane remodeling [metalloproteinases 2 (MMP-2) and 9 (MMP-9)] in fresh SF (FSF) and vitrified SF (VSF) followed by in vitro culture for 6 (D6) or 12 days (D12). The percentage of intact follicles, follicular and oocyte diameter of the VSF were lower than FSF on both days of culture (P < 0.05). The VSF viability was statistically reduced from D6 (95.5%) to D12 (77.3%) but did not differ from the FSF on both days (D6:96.2% to D12:86.5%). Antrum formation in the VSF (D6: 59.13%; D12: 79.56%) was significantly lower than the FSF (D6: 79.61%; D12: 92.23%). However, an increase in this percentage was observed from D6 to D12 in both groups. Aromatase showed stronger labeling on FSF D6 and VSF D12 compared to other treatments (P < 0.05). MMP-2 showed a similar pattern of labeling in FSF D6 and VSF D12, similarly to that observed in FSF D12 and VSF D6. MMP-9 was similar in FSF and VSF cultivated for 6 and 12 days. In conclusion, VSF are able to grow and develop during 12 days of in vitro culture and showed evidence of preservation of steroidogenic function and remodeling of the basement membrane.
Collapse
Affiliation(s)
- Francisco Denilson Rodrigues Gomes
- Laboratory of Manipulation of Oocytes and Ovarian Pre-Antral Follicles (LAMOFOPA), Faculty of Veterinary, State University of Ceará, Av. Dr. Silas Munguba, 1700, Fortaleza, CE, CEP: 60714-903, Brazil
| | - Danielle Cristina Calado de Brito
- Laboratory of Manipulation of Oocytes and Ovarian Pre-Antral Follicles (LAMOFOPA), Faculty of Veterinary, State University of Ceará, Av. Dr. Silas Munguba, 1700, Fortaleza, CE, CEP: 60714-903, Brazil
| | - Naíza Arcângela Ribeiro de Sá
- Laboratory of Manipulation of Oocytes and Ovarian Pre-Antral Follicles (LAMOFOPA), Faculty of Veterinary, State University of Ceará, Av. Dr. Silas Munguba, 1700, Fortaleza, CE, CEP: 60714-903, Brazil
| | - Lucy Vanessa Sulca Ñaupas
- Laboratory of Manipulation of Oocytes and Ovarian Pre-Antral Follicles (LAMOFOPA), Faculty of Veterinary, State University of Ceará, Av. Dr. Silas Munguba, 1700, Fortaleza, CE, CEP: 60714-903, Brazil
| | - Gaby Judith Quispe Palomino
- Laboratory of Manipulation of Oocytes and Ovarian Pre-Antral Follicles (LAMOFOPA), Faculty of Veterinary, State University of Ceará, Av. Dr. Silas Munguba, 1700, Fortaleza, CE, CEP: 60714-903, Brazil
| | - Renato Felix da Silva
- Laboratory of Manipulation of Oocytes and Ovarian Pre-Antral Follicles (LAMOFOPA), Faculty of Veterinary, State University of Ceará, Av. Dr. Silas Munguba, 1700, Fortaleza, CE, CEP: 60714-903, Brazil
| | - Éverton Pimentel Ferreira Lopes
- Laboratory of Manipulation of Oocytes and Ovarian Pre-Antral Follicles (LAMOFOPA), Faculty of Veterinary, State University of Ceará, Av. Dr. Silas Munguba, 1700, Fortaleza, CE, CEP: 60714-903, Brazil
| | - Gildas Tetaping Mbemya
- Laboratory of Manipulation of Oocytes and Ovarian Pre-Antral Follicles (LAMOFOPA), Faculty of Veterinary, State University of Ceará, Av. Dr. Silas Munguba, 1700, Fortaleza, CE, CEP: 60714-903, Brazil
| | | | - Mary Zelinski
- Oregon National Primate Research Center, Beaverton, OR, USA
| | - José Ricardo de Figueiredo
- Laboratory of Manipulation of Oocytes and Ovarian Pre-Antral Follicles (LAMOFOPA), Faculty of Veterinary, State University of Ceará, Av. Dr. Silas Munguba, 1700, Fortaleza, CE, CEP: 60714-903, Brazil
| | - Ana Paula Ribeiro Rodrigues
- Laboratory of Manipulation of Oocytes and Ovarian Pre-Antral Follicles (LAMOFOPA), Faculty of Veterinary, State University of Ceará, Av. Dr. Silas Munguba, 1700, Fortaleza, CE, CEP: 60714-903, Brazil.
| |
Collapse
|
37
|
Eijkenboom L, Saedt E, Zietse C, Braat D, Beerendonk C, Peek R. Strategies to safely use cryopreserved ovarian tissue to restore fertility after cancer: A systematic review. Reprod Biomed Online 2022; 45:763-778. [DOI: 10.1016/j.rbmo.2022.05.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 05/19/2022] [Accepted: 05/23/2022] [Indexed: 10/18/2022]
|
38
|
Cioffi R, Fais ML, Bergamini A, Vanni VS, Pagliardini L, Papaleo E, Mangili G, Candiani M. Ovarian failure risk in post-pubertal patients with cancer: a prognostic model. Future Oncol 2022; 18:2391-2400. [PMID: 35469452 DOI: 10.2217/fon-2022-0078] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: To develop a predictive model for ovarian failure (OF) after chemotherapy in young post-pubertal women with cancer. Methods: Retrospective, monocentric cohort study including 348 patients referring to the Oncofertility Unit of San Raffaele Hospital (Milan, Italy) from August 2011 to January 2020. A predictive model was constructed by multivariate logistic regression and receiver operating characteristic analysis. Results: Data about menstrual function resumption were available for 184 patients. The best predictive model for OF was identified by the combination of age; number of chemotherapy lines; vincristine, adriamycin, ifosphamide/adriamycin, ifosphamide; capecitabine; adriamycin, bleomycine, vinblastine, doxorubicin (area under the curve= 0.906, CI 95% 0.858-0.954, p = 0.0001). Conclusions: The model predicts the probability of loss of ovarian function at cancer diagnosis and with every change of treatment.
Collapse
Affiliation(s)
- Raffaella Cioffi
- Obstetrics & Gynecology Unit, IRCCS San Raffaele Scientific Institute, Milan, 20132, Italy
| | - Maria Luisa Fais
- Obstetrics & Gynecology Unit, University of Cagliari, Cagliari, 09124, Italy
| | - Alice Bergamini
- Obstetrics & Gynecology Unit, IRCCS San Raffaele Scientific Institute, Milan, 20132, Italy
| | - Valeria Stella Vanni
- Obstetrics & Gynecology Unit, IRCCS San Raffaele Scientific Institute, Milan, 20132, Italy
| | - Luca Pagliardini
- Division of Genetics & Cell Biology, Reproductive Sciences Laboratory, San Raffaele Scientific Institute, Milan, 20132, Italy
| | - Enrico Papaleo
- Obstetrics & Gynecology Unit, IRCCS San Raffaele Scientific Institute, Milan, 20132, Italy
| | - Giorgia Mangili
- Obstetrics & Gynecology Unit, IRCCS San Raffaele Scientific Institute, Milan, 20132, Italy
| | - Massimo Candiani
- Obstetrics & Gynecology Unit, IRCCS San Raffaele Scientific Institute, Milan, 20132, Italy
| |
Collapse
|
39
|
Assessment of female fertility preservation in Auvergne 3 years after implementation of the PREFERA platform (PREservation FERtilité Auvergne). J Gynecol Obstet Hum Reprod 2022; 51:102342. [PMID: 35181543 DOI: 10.1016/j.jogoh.2022.102342] [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: 10/27/2021] [Revised: 02/03/2022] [Accepted: 02/14/2022] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Fertility preservation (FP) in patients with cancer or pathology at risk of gonadotoxicity is now according to legislation, an integral part of the treatment protocol. for this reason, clinical-biological platforms have emerged with the aim of developing and improving this practice, such as the PREFERA platform (PREservation FERtilité Auvergne) MATERIAL ET METHOD: This is an observational cohort study to evaluate female fertility preservation activity in Auvergne at the AMP-CECOS center of the Clermont-Ferrand University Hospital from March 2013 to March 2019. This period covering 3 years before and after the creation of PREFERA in 2015. RESULTS 205 patients were referred for fertility preservation consultations, including 77 before the platform was set up and 128 after, corresponding to an increase of 66%. 190 patients (92.7%) referred were eligible for FP, of whom 169 (88.9%) received treatment. Thirty-nine patients underwent oocyte vitrification before the platform was set up and 74 after (+89.7%), twenty patients underwent ovarian cortex freezing before the platform was set up and 27 after (+35%). Only 54 patients (26.2%) were seen for follow-up with an increased number of consultations following the implementation of PREFERA. (8% vs 33%, p<0.001). CONCLUSION Creation of the PREFERA platform facilitated patient access and management of fertility preservation procedures. However, at the regional level, it is necessary to continue to raise awareness of fertility issues, particularly in the context of post-cancer follow-up, both among patients and health professionals.
Collapse
|
40
|
Ghezelayagh Z, Khoshdel-Rad N, Ebrahimi B. Human ovarian tissue in-vitro culture: primordial follicle activation as a new strategy for female fertility preservation. Cytotechnology 2022; 74:1-15. [PMID: 35185282 PMCID: PMC8816997 DOI: 10.1007/s10616-021-00510-2] [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: 05/29/2021] [Accepted: 11/18/2021] [Indexed: 02/03/2023] Open
Abstract
Cryopreservation and transplantation of ovarian tissue is the only fertility preservation option used for prepubertal girls and women who don't have a chance for embryo or oocyte vitrification. For women with aggressive cancer, hormone-responsive malignancies, autoimmune diseases, etc. ovary transplantation cannot be performed so an alternative technology called in-vitro follicle activation is thinkable. In this method, dormant primordial follicles are activated from the resting primordial pool by in-vitro culture and enter their growth phase. Different in-vitro culture media and supplements in addition to various culturing methods have been conducted for activating these dormant follicles. Furthermore, several signaling pathways such as Hippo, phosphatidylinositol-3-kinase, and mTOR influence follicle activation. Therefore, the addition of different activators of these signaling pathways can beneficially regulate this culture system. This review summarizes the findings on different aspects of human ovarian tissue culture strategies for in-vitro follicular activation, their medium, and different factors involved in this activation. Afterward, signaling pathways important for follicle activation and their clinical applications towards improving activation in culture are also reviewed.
Collapse
Affiliation(s)
- Zeinab Ghezelayagh
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
- Department of Developmental Biology, Faculty of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, ACECR, Tehran, Iran
| | - Niloofar Khoshdel-Rad
- Department of Developmental Biology, Faculty of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, ACECR, Tehran, Iran
- Department of Stem Cell and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Bita Ebrahimi
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| |
Collapse
|
41
|
Gharibeh N, Aghebati-Maleki L, Madani J, Pourakbari R, Yousefi M, Ahmadian Heris J. Cell-based therapy in thin endometrium and Asherman syndrome. Stem Cell Res Ther 2022; 13:33. [PMID: 35090547 PMCID: PMC8796444 DOI: 10.1186/s13287-021-02698-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 12/24/2021] [Indexed: 12/17/2022] Open
Abstract
Numerous treatment strategies have so far been proposed for treating refractory thin endometrium either without or with the Asherman syndrome. Inconsistency in the improvement of endometrial thickness is a common limitation of such therapies including tamoxifen citrate as an ovulation induction agent, acupuncture, long-term pentoxifylline and tocopherol or tocopherol only, low-dose human chorionic gonadotropin during endometrial preparation, aspirin, luteal gonadotropin-releasing hormone agonist supplementation, and extended estrogen therapy. Recently, cell therapy has been proposed as an ideal alternative for endometrium regeneration, including the employment of stem cells, platelet-rich plasma, and growth factors as therapeutic agents. The mechanisms of action of cell therapy include the cytokine induction, growth factor production, natural killer cell activity reduction, Th17 and Th1 decrease, and Treg cell and Th2 increase. Since cell therapy is personalized, dynamic, interactive, and specific and could be an effective strategy. Despite its promising nature, further research is required for improving the procedure and the safety of this strategy. These methods and their results are discussed in this article.
Collapse
Affiliation(s)
- Nastaran Gharibeh
- Student's Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Javad Madani
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ramin Pourakbari
- Student's Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Yousefi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Javad Ahmadian Heris
- Department of Allergy and Clinical Immunology, Pediatric Hospital, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
42
|
Zver T, Frontczak S, Poirot C, Rives-Feraille A, Leroy-Martin B, Koscinski I, Arbez-Gindre F, Garnache-Ottou F, Roux C, Amiot C. Minimal residual disease detection by multicolor flow cytometry in cryopreserved ovarian tissue from leukemia patients. J Ovarian Res 2022; 15:9. [PMID: 35042558 PMCID: PMC8767661 DOI: 10.1186/s13048-021-00936-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 12/13/2021] [Indexed: 12/26/2022] Open
Abstract
Background Cryopreservation of ovarian tissue is a fertility-preservation option for women before gonadotoxic treatments. However, cryopreserved ovarian tissue transplantation must be performed with caution in women with malignancies that may metastasize to the ovaries. For this purpose, detecting minimal residual disease (MRD) in the ovarian cortex using sensitive methods is a crucial step. We developed an automated ovarian tissue dissociation method to obtain ovarian cell suspensions. Results We assessed MRD by multicolor flow cytometry (MFC) in cryopreserved ovarian cortex of 15 leukemia patients: 6 with B-cell acute lymphoblastic leukemia (B-ALL), 2 with T-cell acute lymphoblastic leukemia (T-ALL) and 7 with acute myeloid leukemia (AML). Ovarian MRD was positive in 5 of the 15 leukemia patients (one T-ALL and 4 AML). No B-ALL patient was positive by MFC. Quantitative reverse-transcribed polymerase chain reaction was performed when a molecular marker was available, and confirmed the MFC results for 3 patients tested. Xenografts into immunodeficient mice were also performed with ovarian cortical tissue from 10 leukemia patients, with no evidence of leukemic cells after the 6-month grafting period. Conclusions In conclusion, this is the first study using MFC to detect MRD in ovarian cortical tissue from acute leukemia patients. MFC has been accepted in clinical practice for its ease of use, the large number of parameters available simultaneously, and high throughput analysis. We demonstrate here that MFC is a reliable method to detect MRD in cryopreserved ovarian tissue, with a view to controlling the oncological risk before ovarian tissue transplantation in leukemia patients. Supplementary Information The online version contains supplementary material available at 10.1186/s13048-021-00936-4.
Collapse
Affiliation(s)
- Tristan Zver
- CHU de Besançon, Service de Biologie et Médecine de la Reproduction, Cryobiologie, CECOS Bourgogne Franche-Comté, 25000, Besançon, France. .,Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, RIGHT Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire Et Génique, 25000, Besançon, France. .,INSERM CIC-1431, CHU Besançon, 25000, Besançon, France.
| | - Sophie Frontczak
- CHU de Besançon, Service de Biologie et Médecine de la Reproduction, Cryobiologie, CECOS Bourgogne Franche-Comté, 25000, Besançon, France.,Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, RIGHT Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire Et Génique, 25000, Besançon, France.,INSERM CIC-1431, CHU Besançon, 25000, Besançon, France
| | - Catherine Poirot
- Hôpital Saint-Louis, Service d'Hématologie, Unité AJA, 75010, Paris, France
| | | | - Brigitte Leroy-Martin
- CHU de Lille, Laboratoire de Biologie de la Reproduction, CECOS, Spermiologie, 59000, Lille, France
| | - Isabelle Koscinski
- CHRU de Nancy, Service de Biologie de la Reproduction, CECOS, 54035, Nancy, France
| | | | - Francine Garnache-Ottou
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, RIGHT Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire Et Génique, 25000, Besançon, France
| | - Christophe Roux
- CHU de Besançon, Service de Biologie et Médecine de la Reproduction, Cryobiologie, CECOS Bourgogne Franche-Comté, 25000, Besançon, France.,Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, RIGHT Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire Et Génique, 25000, Besançon, France.,INSERM CIC-1431, CHU Besançon, 25000, Besançon, France
| | - Clotilde Amiot
- CHU de Besançon, Service de Biologie et Médecine de la Reproduction, Cryobiologie, CECOS Bourgogne Franche-Comté, 25000, Besançon, France.,Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, RIGHT Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire Et Génique, 25000, Besançon, France.,INSERM CIC-1431, CHU Besançon, 25000, Besançon, France
| |
Collapse
|
43
|
Effect of Previous Alkylating Agent Exposure on Follicle Numbers in Cryopreserved Prepubertal and Young Adult Ovarian Tissue after Long-Term Xenografting. Cancers (Basel) 2022; 14:cancers14020399. [PMID: 35053561 PMCID: PMC8773859 DOI: 10.3390/cancers14020399] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/30/2021] [Accepted: 01/10/2022] [Indexed: 11/30/2022] Open
Abstract
Simple Summary Cryopreservation of ovarian tissue is a promising technique for fertility preservation in cancer patients at increased risk for subfertility. The International Guideline Harmonization Group recommends ovarian tissue cryopreservation for children and young adults before therapy with cumulative doses of alkylating agent at or above 6000–8000 mg/m2. A therapy that poses a high risk of subfertility is rarely the first-line therapy and many of the patients have already undergone several regimens of chemotherapy. The aim of our study was to assess the effects of chemotherapy exposures on the quality of cryopreserved ovarian tissue. We confirmed the harmful effects of alkylating agents on xenografted ovarian tissue and suggest that cumulative doses which are not regarded as an indication for fertility preservation in children and young adult may decrease the quality of cryopreserved follicles. Abstract Purpose and methods: To elucidate whether previous cancer treatment affects graft recovery and follicle numbers, morphology, and development in grafts, cryopreserved ovarian biopsies obtained from 18 cancer patients aged 1–24 years with and without exposure to chemotherapy were xenografted as 1 mm3 fragments to immunodeficient mice for 22 weeks with exogenous stimulation. Results: Graft recovery showed no association with chemotherapy exposure, pubertal stage, or leukemia contamination. Total follicle number per recovered graft varied between 0 and 1031 in the chemotherapy-exposed and between 0 and 502 in the non-chemotherapy-exposed group. Atretic follicles formed the largest proportion of the follicle pool in chemotherapy-exposed grafts. Increased atresia correlated with exposure to alkylating agents (mean ± SD 8866.2 ± 9316.3 mg/m2) but not with anthracyclines, pubertal stage, or leukemia contamination. Conclusion: The observation confirms the harmful effects of alkylating agents on ovarian tissue. Therapy at the median cumulative dose of 8866 mg/m2 leads to the decreased quality of cryopreserved ovarian follicles in children and young adults.
Collapse
|
44
|
Diaz AA, Kubo H, Handa N, Hanna M, Laronda MM. A Systematic Review of Ovarian Tissue Transplantation Outcomes by Ovarian Tissue Processing Size for Cryopreservation. Front Endocrinol (Lausanne) 2022; 13:918899. [PMID: 35774145 PMCID: PMC9239173 DOI: 10.3389/fendo.2022.918899] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 05/10/2022] [Indexed: 01/18/2023] Open
Abstract
UNLABELLED Ovarian tissue cryopreservation (OTC) is the only pre-treatment option currently available to preserve fertility for prepubescent girls and patients who cannot undergo ovarian stimulation. Currently, there is no standardized method of processing ovarian tissue for cryopreservation, despite evidence that fragmentation of ovaries may trigger primordial follicle activation. Because fragmentation may influence ovarian transplant function, the purpose of this systematic review was (1) to identify the processing sizes and dimensions of ovarian tissue within sites around the world, and (2) to examine the reported outcomes of ovarian tissue transplantation including, reported duration of hormone restoration, pregnancy, and live birth. A total of 2,252 abstracts were screened against the inclusion criteria. In this systematic review, 103 studies were included for analysis of tissue processing size and 21 studies were included for analysis of ovarian transplantation outcomes. Only studies where ovarian tissue was cryopreserved (via slow freezing or vitrification) and transplanted orthotopically were included in the review. The size of cryopreserved ovarian tissue was categorized based on dimensions into strips, squares, and fragments. Of the 103 studies, 58 fertility preservation sites were identified that processed ovarian tissue into strips (62%), squares (25.8%), or fragments (31%). Ovarian tissue transplantation was performed in 92 participants that had ovarian tissue cryopreserved into strips (n = 51), squares (n = 37), and fragments (n = 4). All participants had ovarian tissue cryopreserved by slow freezing. The pregnancy rate was 81.3%, 45.5%, 66.7% in the strips, squares, fragment groups, respectively. The live birth rate was 56.3%, 18.2%, 66.7% in the strips, squares, fragment groups, respectively. The mean time from ovarian tissue transplantation to ovarian hormone restoration was 3.88 months, 3.56 months, and 3 months in the strips, squares, and fragments groups, respectively. There was no significant difference between the time of ovarian function' restoration and the size of ovarian tissue. Transplantation of ovarian tissue, regardless of its processing dimensions, restores ovarian hormone activity in the participants that were reported in the literature. More detailed information about the tissue processing size and outcomes post-transplant are required to identify a preferred or more successful processing method. SYSTEMATIC REVIEW REGISTRATION [https://www.crd.york.ac.uk], identifier [CRD42020189120].
Collapse
Affiliation(s)
- Ashley A. Diaz
- Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL, United States
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Hana Kubo
- Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL, United States
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Nicole Handa
- Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL, United States
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Maria Hanna
- Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL, United States
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Monica M. Laronda
- Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL, United States
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- *Correspondence: Monica M. Laronda,
| |
Collapse
|
45
|
Chen J, Isachenko E, Wang W, Du X, Wang M, Rahimi G, Mallmann P, Isachenko V. Optimization of Follicle Isolation for Bioengineering of Human Artificial Ovary. Biopreserv Biobank 2021; 20:529-539. [PMID: 34936496 DOI: 10.1089/bio.2021.0060] [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: 12/26/2022] Open
Abstract
Background: A functional artificial ovary is a promising strategy to recover fertility and restore endocrine function in cancer patients. The aim of this study is to optimize the follicle isolation protocol for cryopreserved human ovarian tissues. Methods: Each of the cryopreserved human ovarian cortex pieces (OCPs) from 10 patients was cut into two equal parts and randomly distributed into two treatment groups. Group 1: OCPs digested with Tumor Dissociation Enzyme (TDE); Group 2: OCPs digested with Liberase Dispase High (DH). The efficiency of both groups were evaluated in terms of yield, viability, morphology, and a short-term in vitro culture (IVC) in alginate scaffolds. Results: The TDE can isolate more primordial follicles and smaller diameter of follicles than Liberase DH. The TDE also enabled the isolation of more bright red follicles, higher percent of viable follicles, more morphologically normal follicles, and lower oxidative stress levels compared with Liberase DH. After eight days of IVC, follicles in the TDE group had a higher growth rate from Day 0 to Day 8, and higher viability on Day 8 than the Liberase DH Group. Conclusion: The TDE can be considered an alternative to Liberase DH, enables the isolation of a higher number of healthy follicles from human OCPs, and improves follicle survival after IVC in contrast to Liberase DH.
Collapse
Affiliation(s)
- Jing Chen
- Research Group for Reproductive Medicine and IVF Laboratory, Department of Obstetrics and Gynecology, Cologne University, Cologne, Germany.,Reproductive Medical Center, The First Affiliated Hospital of Xiamen University, Fujian, People's Republic of China
| | - Evgenia Isachenko
- Research Group for Reproductive Medicine and IVF Laboratory, Department of Obstetrics and Gynecology, Cologne University, Cologne, Germany
| | - Wanxue Wang
- Research Group for Reproductive Medicine and IVF Laboratory, Department of Obstetrics and Gynecology, Cologne University, Cologne, Germany
| | - Xinxin Du
- Research Group for Reproductive Medicine and IVF Laboratory, Department of Obstetrics and Gynecology, Cologne University, Cologne, Germany
| | - Mengying Wang
- Research Group for Reproductive Medicine and IVF Laboratory, Department of Obstetrics and Gynecology, Cologne University, Cologne, Germany
| | - Gohar Rahimi
- Research Group for Reproductive Medicine and IVF Laboratory, Department of Obstetrics and Gynecology, Cologne University, Cologne, Germany
| | - Peter Mallmann
- Research Group for Reproductive Medicine and IVF Laboratory, Department of Obstetrics and Gynecology, Cologne University, Cologne, Germany
| | - Vladimir Isachenko
- Research Group for Reproductive Medicine and IVF Laboratory, Department of Obstetrics and Gynecology, Cologne University, Cologne, Germany
| |
Collapse
|
46
|
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: 7.0] [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
|
47
|
Al-Mahayri ZN, AlAhmad MM, Ali BR. Long-Term Effects of Pediatric Acute Lymphoblastic Leukemia Chemotherapy: Can Recent Findings Inform Old Strategies? Front Oncol 2021; 11:710163. [PMID: 34722258 PMCID: PMC8554193 DOI: 10.3389/fonc.2021.710163] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 09/30/2021] [Indexed: 02/05/2023] Open
Abstract
During the last few decades, pediatric acute lymphoblastic leukemia (ALL) cure rates have improved significantly with rates exceeding 90%. Parallel to this remarkable improvement, there has been mounting interest in the long-term health of the survivors. Consequently, modified treatment protocols have been developed and resulted in the reduction of many adverse long-term consequences. Nevertheless, these are still substantial concerns that warrant further mitigation efforts. In the current review, pediatric-ALL survivors’ late adverse events, including secondary malignant neoplasms (SMNs), cardiac toxicity, neurotoxicity, bone toxicity, hepatic dysfunction, visual changes, obesity, impact on fertility, and neurocognitive effects have been evaluated. Throughout this review, we attempted to answer a fundamental question: can the recent molecular findings mitigate pediatric-ALL chemotherapy’s long-term sequelae on adult survivors? For SMNs, few genetic predisposition factors have been identified including TP53 and POT1 variants. Other treatment-related risk factors have been identified such as anthracyclines’ possible association with breast cancer in female survivors. Cardiotoxicity is another significant and common adverse event with some germline variants been found, albeit with conflicting evidence, to increase the risk of cardiac toxicity. For peripheral neurotoxicity, vincristine is the primary neurotoxic agent in ALL regimens. Some germline genetic variants were found to be associated with the vincristine neurotoxic effect’s vulnerability. However, these were mainly detected with acute neuropathy. Moreover, the high steroid doses and prolonged use increase bone toxicity and obesity risk with some pharmacogenetic biomarkers were associated with increased steroid sensitivity. Therefore, the role of these biomarkers in tailoring steroid choice and dose is a promising research area. Future directions in pediatric ALL treatment should consider the various opportunities provided by genomic medicine. Understanding the molecular bases underlying toxicities will classify patients into risk groups and implement a closer follow-up to those at higher risk. Pharmacogenetic-guided dosing and selecting between alternative agents have proven their efficacy in the short-term management of childhood ALL. It is the right time to think about a similar approach for the life-long consequences on survivors.
Collapse
Affiliation(s)
- Zeina N Al-Mahayri
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Mohammad M AlAhmad
- Department of Clinical Pharmacy, College of Pharmacy, Al-Ain University, Al-Ain, United Arab Emirates
| | - Bassam R Ali
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates.,Zayed Center for Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| |
Collapse
|
48
|
Minimal residual disease quantification in ovarian tissue collected from patients in complete remission of acute leukemia. Blood 2021; 137:1697-1701. [PMID: 33171484 DOI: 10.1182/blood.2020007782] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 10/18/2020] [Indexed: 11/20/2022] Open
|
49
|
Kim S, Kim SW, Han SJ, Lee S, Park HT, Song JY, Kim T. Molecular Mechanism and Prevention Strategy of Chemotherapy- and Radiotherapy-Induced Ovarian Damage. Int J Mol Sci 2021; 22:ijms22147484. [PMID: 34299104 PMCID: PMC8305189 DOI: 10.3390/ijms22147484] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 07/12/2021] [Indexed: 12/14/2022] Open
Abstract
Fertility preservation is an emerging discipline, which is of substantial clinical value in the care of young patients with cancer. Chemotherapy and radiation may induce ovarian damage in prepubertal girls and young women. Although many studies have explored the mechanisms implicated in ovarian toxicity during cancer treatment, its molecular pathophysiology is not fully understood. Chemotherapy may accelerate follicular apoptosis and follicle reservoir utilization and damage the ovarian stroma via multiple molecular reactions. Oxidative stress and the radiosensitivity of oocytes are the main causes of gonadal damage after radiation treatment. Fertility preservation options can be differentiated by patient age, desire for conception, treatment regimen, socioeconomic status, and treatment duration. This review will help highlight the importance of multidisciplinary oncofertility strategies for providing high-quality care to young female cancer patients.
Collapse
Affiliation(s)
- Seongmin Kim
- Gynecologic Cancer Center, CHA Ilsan Medical Center, CHA University College of Medicine, 1205 Jungang-ro, Ilsandong-gu, Goyang-si 10414, Korea;
| | - Sung-Woo Kim
- Department of Obstetrics and Gynecology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 03080, Korea; (S.-W.K.); (S.-J.H.)
| | - Soo-Jin Han
- Department of Obstetrics and Gynecology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 03080, Korea; (S.-W.K.); (S.-J.H.)
| | - Sanghoon Lee
- Department of Obstetrics and Gynecology, Korea University College of Medicine, 73 Inchon-ro, Seongbuk-gu, Seoul 02841, Korea; (H.-T.P.); (J.-Y.S.); (T.K.)
- Correspondence: ; Tel.: +82-2-920-6773
| | - Hyun-Tae Park
- Department of Obstetrics and Gynecology, Korea University College of Medicine, 73 Inchon-ro, Seongbuk-gu, Seoul 02841, Korea; (H.-T.P.); (J.-Y.S.); (T.K.)
| | - Jae-Yun Song
- Department of Obstetrics and Gynecology, Korea University College of Medicine, 73 Inchon-ro, Seongbuk-gu, Seoul 02841, Korea; (H.-T.P.); (J.-Y.S.); (T.K.)
| | - Tak Kim
- Department of Obstetrics and Gynecology, Korea University College of Medicine, 73 Inchon-ro, Seongbuk-gu, Seoul 02841, Korea; (H.-T.P.); (J.-Y.S.); (T.K.)
| |
Collapse
|
50
|
Rodriguez-Wallberg KA, Milenkovic M, Papaikonomou K, Keros V, Gustafsson B, Sergouniotis F, Wikander I, Perot R, Borgström B, Ljungman P, Barbany G. Successful pregnancies after transplantation of ovarian tissue retrieved and cryopreserved at time of childhood acute lymphoblastic leukemia - A case report. Haematologica 2021; 106:2783-2787. [PMID: 34233451 PMCID: PMC8485665 DOI: 10.3324/haematol.2021.278828] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Indexed: 11/09/2022] Open
Abstract
Not available.
Collapse
Affiliation(s)
- Kenny A Rodriguez-Wallberg
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden; Department of Reproductive Medicine, Division of Gynecology and Reproduction, Karolinska University Hospital, Stockholm.
| | - Milan Milenkovic
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden; Department of Reproductive Medicine, Division of Gynecology and Reproduction, Karolinska University Hospital, Stockholm
| | - Kiriaki Papaikonomou
- Department of Women's and Children's Health, Division of Obstetrics and Gynecology, Karolinska Institutet, Stockholm
| | - Victoria Keros
- Department of Reproductive Medicine, Division of Gynecology and Reproduction, Karolinska University Hospital, Stockholm
| | - Britt Gustafsson
- Department of Women's and Children's Health, Division of Pediatric Oncology, Karolinska Institutet, Stockholm
| | - Fotios Sergouniotis
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden; Department of Reproductive Medicine, Division of Gynecology and Reproduction, Karolinska University Hospital, Stockholm
| | - Ida Wikander
- Department of Reproductive Medicine, Division of Gynecology and Reproduction, Karolinska University Hospital, Stockholm
| | - Ronak Perot
- Department of Gynecology, Division of Gynecology and Reproduction, Karolinska University Hospital, Stockholm
| | - Birgit Borgström
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm
| | - Per Ljungman
- Dept. of Cellular Therapy and Allogeneic Stem Cell Transplantation, Karolinska University Hospital and Division of Hematology, Department of Medicine Huddinge, Karolinska Institutet, Stockholm
| | - Gisela Barbany
- Department of Molecular Medicine and Surgery, Karolinska Institutet and Department of Clinical Genetics, Laboratory Division Karolinska University Hospital, Stockholm
| |
Collapse
|