1
|
Tanaka Y, Amano T, Nakamura A, Takahashi A, Takebayashi A, Hanada T, Tsuji S, Murakami T. Balancing Fertility Preservation and Treatment Efficacy in (Neo)adjuvant Therapy for Adolescent and Young Adult Breast Cancer Patients: a Narrative Review. Curr Oncol Rep 2024; 26:1563-1574. [PMID: 39499484 DOI: 10.1007/s11912-024-01615-5] [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] [Accepted: 10/11/2024] [Indexed: 11/07/2024]
Abstract
PURPOSE OF REVIEW Adolescent and young adult (AYA) breast cancer survivors face a significant risk of infertility due to the gonadotoxic effects of (neo)adjuvant therapy, which complicates their ability to conceive post-treatment. While (neo)adjuvant therapy primarily aims to improve recurrence-free and overall survival, fertility preservation strategies should also be considered for young patients. This narrative review explores recent advancements in fertility preservation techniques, such as oocyte, embryo, and ovarian tissue cryopreservation, and evaluates the feasibility of modifying breast cancer (neo)adjuvant therapy to preserve fertility without compromising survival outcomes. RECENT FINDINGS Our review highlights that clinical trials with co-primary endpoints of oncological safety and fertility preservation are limited, and substituting standard treatment regimens solely for fertility preservation is currently not recommended. Nevertheless, new clinical studies have emerged that either exclude highly ovarian-toxic agents, such as cyclophosphamide, or omit adjuvant therapy altogether, even if fertility preservation is not their primary endpoint. Unfortunately, many of these trials have not evaluated ovarian toxicity. Notably, since 2020, major oncology organizations, including the American Society of Clinical Oncology (ASCO), the European Society of Medical Oncology (ESMO) have advocated for the routine assessment of ovarian toxicity in all clinical trials. The review underscores the importance of incorporating ovarian toxicity as a standard endpoint in future trials involving premenopausal breast cancer patients to identify treatment regimens that can effectively balance fertility preservation with treatment efficacy.
Collapse
Affiliation(s)
- Yuji Tanaka
- Department of Obstetrics and Gynaecology, Shiga University of Medical Science, 520-2192/Seta Tsukinowa-Cho, Otsu, Shiga, Japan.
| | - Tsukuru Amano
- Department of Obstetrics and Gynaecology, Shiga University of Medical Science, 520-2192/Seta Tsukinowa-Cho, Otsu, Shiga, Japan
| | - Akiko Nakamura
- Department of Obstetrics and Gynaecology, Shiga University of Medical Science, 520-2192/Seta Tsukinowa-Cho, Otsu, Shiga, Japan
| | - Akimasa Takahashi
- Department of Obstetrics and Gynaecology, Shiga University of Medical Science, 520-2192/Seta Tsukinowa-Cho, Otsu, Shiga, Japan
| | - Akie Takebayashi
- Department of Obstetrics and Gynaecology, Shiga University of Medical Science, 520-2192/Seta Tsukinowa-Cho, Otsu, Shiga, Japan
| | - Tetsuro Hanada
- Department of Obstetrics and Gynaecology, Shiga University of Medical Science, 520-2192/Seta Tsukinowa-Cho, Otsu, Shiga, Japan
| | - Shunichiro Tsuji
- Department of Obstetrics and Gynaecology, Shiga University of Medical Science, 520-2192/Seta Tsukinowa-Cho, Otsu, Shiga, Japan
| | - Takashi Murakami
- Department of Obstetrics and Gynaecology, Shiga University of Medical Science, 520-2192/Seta Tsukinowa-Cho, Otsu, Shiga, Japan
| |
Collapse
|
2
|
Bhattacharya A, Alam K, Roy NS, Kaur K, Kaity S, Ravichandiran V, Roy S. Exploring the interaction between extracellular matrix components in a 3D organoid disease model to replicate the pathophysiology of breast cancer. J Exp Clin Cancer Res 2023; 42:343. [PMID: 38102637 PMCID: PMC10724947 DOI: 10.1186/s13046-023-02926-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 12/06/2023] [Indexed: 12/17/2023] Open
Abstract
In vitro models are necessary to study the pathophysiology of the disease and the development of effective, tailored treatment methods owing to the complexity and heterogeneity of breast cancer and the large population affected by it. The cellular connections and tumor microenvironments observed in vivo are often not recapitulated in conventional two-dimensional (2D) cell cultures. Therefore, developing 3D in vitro models that mimic the complex architecture and physiological circumstances of breast tumors is crucial for advancing our understanding of the illness. A 3D scaffold-free in vitro disease model mimics breast cancer pathophysiology by allowing cells to self-assemble/pattern into 3D structures, in contrast with other 3D models that rely on artificial scaffolds. It is possible that this model, whether applied to breast tumors using patient-derived primary cells (fibroblasts, endothelial cells, and cancer cells), can accurately replicate the observed heterogeneity. The complicated interactions between different cell types are modelled by integrating critical components of the tumor microenvironment, such as the extracellular matrix, vascular endothelial cells, and tumor growth factors. Tissue interactions, immune cell infiltration, and the effects of the milieu on drug resistance can be studied using this scaffold-free 3D model. The scaffold-free 3D in vitro disease model for mimicking tumor pathophysiology in breast cancer is a useful tool for studying the molecular basis of the disease, identifying new therapeutic targets, and evaluating treatment modalities. It provides a more physiologically appropriate high-throughput platform for screening large compound library in a 96-384 well format. We critically discussed the rapid development of personalized treatment strategies and accelerated drug screening platforms to close the gap between traditional 2D cell culture and in vivo investigations.
Collapse
Affiliation(s)
- Anamitra Bhattacharya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India
| | - Kamare Alam
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India
| | - Nakka Sharmila Roy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India
| | - Kulwinder Kaur
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine a Health Sciences, Dublin, Ireland
- Tissue Engineering Research Group, Department of Anatomy & Regenerative Medicine, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Santanu Kaity
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India
| | - Velayutham Ravichandiran
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India
| | - Subhadeep Roy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India.
| |
Collapse
|
3
|
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: 48] [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
|
4
|
Nguyen TYT, Cacciottola L, Camboni A, Ravau J, De Vos M, Demeestere I, Donnez J, Dolmans MM. Ovarian tissue cryopreservation and transplantation in patients with central nervous system tumours. Hum Reprod 2021; 36:1296-1309. [PMID: 33394011 DOI: 10.1093/humrep/deaa353] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 11/24/2020] [Indexed: 11/12/2022] Open
Abstract
STUDY QUESTION Is there a possibility of reseeding cancer cells potentially present in frozen ovarian tissue from patients with central nervous system (CNS) tumours? SUMMARY ANSWER Malignancy reseeding in cryopreserved ovarian tissue from 20 patients with CNS tumours was not detected by histology, immunohistochemistry (IHC), molecular biology or xenotransplantation. WHAT IS KNOWN ALREADY Ovarian metastasis potential has been documented in patients with leukaemia, borderline ovarian tumours, advanced breast cancer and Ewing sarcoma. However, data on the safety of transplanting frozen-thawed ovarian tissue from cancer patients with CNS tumours are still lacking. STUDY DESIGN, SIZE, DURATION This prospective experimental study was conducted in an academic gynaecology research laboratory using cryopreserved ovarian cortex from 20 patients suffering from CNS tumours. Long-term (5 months) xenografting was performed in immunodeficient mice. PARTICIPANTS/MATERIALS, SETTING, METHODS Subjects enrolled in the study were suffering from one of six types of CNS tumours including medulloblastoma, ependymoma, primitive neuroectodermal tumours, astrocytoma, glioblastoma and germinoma. The presence of malignant cells was investigated with disease-specific markers for each patient in cryopreserved and xenografted ovarian tissue by histology, IHC via expression of neuron-specific enolase (NSE) and glial fibrillary acidic protein (GFAP), and reverse transcription droplet digital polymerase chain reaction (RT-ddPCR) for quantification of GFAP and ENO2 gene amplification. MAIN RESULTS AND THE ROLE OF CHANCE Serial sections of cryopreserved and xenografted ovarian tissue from 20 patients showed no malignant cells by histology. All samples were negative for NSE and GFAP, although these neural markers were expressed extensively in the patients' primary tumours. Analysis by RT-ddPCR revealed no cancer cells detected in cryopreserved and xenografted ovarian fragments from subjects with astrocytoma, ependymoma, glioblastoma or medulloblastoma. Taken together, the study found no evidence of malignancy seeding in frozen-thawed and xenotransplanted ovarian tissue from patients affected by CNS cancers. LIMITATIONS, REASONS FOR CAUTION This analysis cannot guarantee complete elimination of disseminated disease from all cryopreserved ovarian cortex, since we are unable to examine the fragments used for transplantation. WIDER IMPLICATIONS OF THE FINDINGS This is the first study to be conducted in patients with CNS cancers undergoing ovarian tissue cryopreservation and transplantation, and clearly demonstrates no tumour seeding in their frozen-thawed and xenografted tissue. This information is vital for doctors to provide patients with meaningful and accurate advice on the possibilities and risks of ovarian tissue reimplantation. STUDY FUNDING/COMPETING INTEREST(S) This study was supported by grants from the Fonds National de la Recherche Scientifique de Belgique-the Excellence of Science (FNRS-EOS), number 30443682 awarded to M.-M.D. and T.Y.T.N., FNRS grant number 5/4/150/5 and FNRS-PDR Convention grant number T.0077.14 awarded to M.-M.D., grant 2018-042 from the Foundation Against Cancer awarded to A.C., and private donations (Ferrero, de Spoelberch). The authors declare no competing financial interests. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Thu Yen Thi Nguyen
- Pôle de Gynécologie, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
| | - Luciana Cacciottola
- Pôle de Gynécologie, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
| | - Alessandra Camboni
- Pôle de Gynécologie, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium.,Service d'Anatomie Pathologique, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Joachim Ravau
- Laboratory of Pediatric Hepatology and Cell Therapy, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
| | - Michel De Vos
- Centre for Reproductive Medicine, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium.,Follicle Biology Laboratory (FOBI), UZ Brussel, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Isabelle Demeestere
- Research Laboratory in Human Reproduction, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Jacques Donnez
- Société de Recherche pour l'Infertilité (SRI), Brussels, Belgium
| | - Marie-Madeleine Dolmans
- Pôle de Gynécologie, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium.,Gynecology Department, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| |
Collapse
|
5
|
Chen J, Todorov P, Isachenko E, Rahimi G, Mallmann P, Isachenko V. Construction and cryopreservation of an artificial ovary in cancer patients as an element of cancer therapy and a promising approach to fertility restoration. HUM FERTIL 2021; 25:651-661. [PMID: 33648431 DOI: 10.1080/14647273.2021.1885756] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The proportion of cancer patients that survive is increasing because of improvements in cancer therapy. However, some cancer treatments, such as chemo- and radio-therapies, can cause considerable damage to reproductive function. The issue of fertility is paramount for women of childbearing age once they are cured from cancer. For those patients with prepubertal or haematogenous cancer, the possibilities of conventional fertility treatments, such as oocyte or embryo cryopreservation and transplantation, are limited. Moreover, ovarian tissue cryopreservation as an alternative to fertility preservation has limitations, with a risk of re-implanting malignant cells in patients who have recovered from potentially fatal malignant disease. One possible way to restore fertility in these patients is to mimic artificially the function of the natural organ, the ovary, by grafting isolated follicles embedded in a biological scaffold to their native environment. Construction and cryopreservation of an artificial ovary might offer a safer alternative option to restore fertility for those who cannot benefit from traditional fertility preservation techniques. This review considers the protocols for constructing an artificial ovary, summarises advances in the field with potential clinical application, and discusses future trends for cryopreservation of these artificial constructions.
Collapse
Affiliation(s)
- Jing Chen
- University Maternal Hospital, Research Group for Reproductive Medicine and IVF-Laboratory, Department of Obstetrics and Gynaecology, Cologne University, Cologne, Germany
| | - Plamen Todorov
- Institute of Biology and Immunology of Reproduction, Sofia, Bulgaria
| | - Evgenia Isachenko
- University Maternal Hospital, Research Group for Reproductive Medicine and IVF-Laboratory, Department of Obstetrics and Gynaecology, Cologne University, Cologne, Germany
| | - Gohar Rahimi
- University Maternal Hospital, Research Group for Reproductive Medicine and IVF-Laboratory, Department of Obstetrics and Gynaecology, Cologne University, Cologne, Germany
| | - Peter Mallmann
- University Maternal Hospital, Research Group for Reproductive Medicine and IVF-Laboratory, Department of Obstetrics and Gynaecology, Cologne University, Cologne, Germany
| | - Vladimir Isachenko
- University Maternal Hospital, Research Group for Reproductive Medicine and IVF-Laboratory, Department of Obstetrics and Gynaecology, Cologne University, Cologne, Germany
| |
Collapse
|
6
|
Anderson RA, Amant F, Braat D, D'Angelo A, Chuva de Sousa Lopes SM, Demeestere I, Dwek S, Frith L, Lambertini M, Maslin C, Moura-Ramos M, Nogueira D, Rodriguez-Wallberg K, Vermeulen N. ESHRE guideline: female fertility preservation. Hum Reprod Open 2020; 2020:hoaa052. [PMID: 33225079 PMCID: PMC7666361 DOI: 10.1093/hropen/hoaa052] [Citation(s) in RCA: 320] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 10/02/2020] [Indexed: 12/15/2022] Open
Abstract
STUDY QUESTION What is the recommended management for women and transgender men with regards to fertility preservation (FP), based on the best available evidence in the literature? SUMMARY ANSWER The ESHRE Guideline on Female Fertility Preservation makes 78 recommendations on organization of care, information provision and support, pre-FP assessment, FP interventions and after treatment care. Ongoing developments in FP are also discussed. WHAT IS KNOWN ALREADY The field of FP has grown hugely in the last two decades, driven by the increasing recognition of the importance of potential loss of fertility as a significant effect of the treatment of cancer and other serious diseases, and the development of the enabling technologies of oocyte vitrification and ovarian tissue cryopreservation (OTC) for subsequent autografting. This has led to the widespread, though uneven, provision of FP for young women. STUDY DESIGN SIZE DURATION The guideline was developed according to the structured methodology for development of ESHRE guidelines. After formulation of key questions by a group of experts, literature searches and assessments were performed. Papers published up to 1 November 2019 and written in English were included in the review. PARTICIPANTS/MATERIALS SETTING METHODS Based on the collected evidence, recommendations were formulated and discussed until consensus was reached within the guideline group. A stakeholder review was organized after finalization of the draft. The final version was approved by the guideline group and the ESHRE Executive Committee. MAIN RESULTS AND THE ROLE OF CHANCE This guideline aims to help providers meet a growing demand for FP options by diverse groups of patients, including those diagnosed with cancer undergoing gonadotoxic treatments, with benign diseases undergoing gonadotoxic treatments or those with a genetic condition predisposing to premature ovarian insufficiency, transgender men (assigned female at birth), and women requesting oocyte cryopreservation for age-related fertility loss.The guideline makes 78 recommendations on information provision and support, pre-FP assessment, FP interventions and after treatment care, including 50 evidence-based recommendations-of which 31 were formulated as strong recommendations and 19 as weak-25 good practice points and 3 research only recommendations. Of the evidence-based recommendations, 1 was supported by high-quality evidence, 3 by moderate-quality evidence, 17 by low-quality evidence and 29 by very low-quality evidence. To support future research in the field of female FP, a list of research recommendations is provided. LIMITATIONS REASONS FOR CAUTION Most interventions included are not well studied in FP patients. As some interventions, e.g. oocyte and embryo cryopreservation, are well established for treatment of infertility, technical aspects, feasibility and outcomes can be extrapolated. For other interventions, such as OTC and IVM, more evidence is required, specifically pregnancy outcomes after applying these techniques for FP patients. Such future studies may require the current recommendations to be revised. WIDER IMPLICATIONS OF THE FINDINGS The guideline provides clinicians with clear advice on best practice in female FP, based on the best evidence currently available. In addition, a list of research recommendations is provided to stimulate further studies in FP. STUDY FUNDING/COMPETING INTERESTS The guideline was developed and funded by ESHRE, covering expenses associated with the guideline meetings, with the literature searches and with the dissemination of the guideline. The guideline group members did not receive payment. R.A.A. reports personal fees and non-financial support from Roche Diagnostics, personal fees from Ferring Pharmaceuticals, IBSA and Merck Serono, outside the submitted work; D.B. reports grants from Merck Serono and Goodlife, outside the submitted work; I.D. reports consulting fees from Roche and speaker's fees from Novartis; M.L. reports personal fees from Roche, Novartis, Pfizer, Lilly, Takeda, and Theramex, outside the submitted work. The other authors have no conflicts of interest to declare. DISCLAIMER This guideline represents the views of ESHRE, which were achieved after careful consideration of the scientific evidence available at the time of preparation. In the absence of scientific evidence on certain aspects, a consensus between the relevant ESHRE stakeholders has been obtained. Adherence to these clinical practice guidelines does not guarantee a successful or specific outcome, nor does it establish a standard of care. Clinical practice guidelines do not replace the need for application of clinical judgment to each individual presentation, nor variations based on locality and facility type. ESHRE makes no warranty, express or implied, regarding the clinical practice guidelines and specifically excludes any warranties of merchantability and fitness for a particular use or purpose. (Full disclaimer available at www.eshre.eu/guidelines.) †ESHRE Pages content is not externally peer reviewed. The manuscript has been approved by the Executive Committee of ESHRE.
Collapse
Affiliation(s)
| | - Richard A Anderson
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK
| | - Frédéric Amant
- Department of Gynaecological Oncology, Academic Medical Centres Amsterdam, Amsterdam, The Netherlands.,Department of Gynaecology, Antoni van Leeuwenhoek-Netherlands Cancer Institute, Amsterdam, The Netherlands.,Department of Oncology, Catholic University Leuven, Leuven, Belgium
| | - Didi Braat
- Department of Obstetrics and Gynaecology, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Arianna D'Angelo
- Wales Fertility Institute, Swansea Bay Health Board, University Hospital of Wales, Cardiff University, Cardiff, UK
| | | | - Isabelle Demeestere
- Fertility Clinic, CUB-Hôpital Erasme and Research Laboratory on Human Reproduction, Université Libre de Bruxelles, Brussels, Belgium
| | | | - Lucy Frith
- Institute of Population Health, University of Liverpool, Liverpool, UK
| | - Matteo Lambertini
- Department of Medical Oncology, U.O.C Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy.,Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
| | | | - Mariana Moura-Ramos
- Reprodutive Medicine Unit, Unit of Clinical Psychology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.,University of Coimbra, Center for Research in Neuropsychology and Cognitive and Behavioral Intervention, Coimbra, Portugal
| | - Daniela Nogueira
- Laboratory of Reproductive Biology, INOVIE Fertilité Clinique Croix du Sud, Toulouse, France
| | - Kenny Rodriguez-Wallberg
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.,Division of Gynaecology and Reproduction, Department of Reproductive Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Nathalie Vermeulen
- European Society of Human Reproduction and Embryology, Central Office, Grimbergen, Belgium
| |
Collapse
|
7
|
Poulain M, Vandame J, Tran C, Koutchinsky S, Pirtea P, Ayoubi JM. Fertility preservation in borderline ovarian tumor patients and survivors. Horm Mol Biol Clin Investig 2020; 43:179-186. [PMID: 32628631 DOI: 10.1515/hmbci-2019-0072] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 05/02/2020] [Indexed: 12/24/2022]
Abstract
Borderline ovarian tumors (BOTs) represent around 15% of all epithelial ovarian cancer. Around one third of those patients is under 40 and has not completed childbearing when the tumor is diagnosed. Cancer survivors are more and more concerned about their future fertility since a large proportion of those with BOTs are young. Whatever the tumor stage, information regarding future fertility after treatment and fertility preservation (FP) options must be delivered to all patients before treatment. A multidisciplinary team will discuss and propose personalized treatment and FP strategies. Nowadays, the FP options offered to patients with BOT are the followings: i) minimal invasive conservative surgery, ii) oocyte cryopreservation after controlled ovarian stimulation (COS) or in vitro maturation (IVM) and iii) ovarian tissue cryopreservation. Generally, the most common strategy to preserve future fertility is represented by minimal invasive conservative surgery. However, with the remarkable success and evolution of assisted reproductive technologies (ART) - notably progress and efficiency in COS and oocyte vitrification - have led to offer another potential approach for FP consisting in oocyte cryopreservation. Several COS protocols, such as random start or dual stimulation associating tamoxifen or aromatase inhibitors with gonadotropins provide similar results when compared to standard protocols while providing safety by minimizing the risk of high estrogen exposure. When COS is contraindicated, oocyte cryopreservation can still be possible throw IVM. Even though, oocyte competence after IVM is lower than that obtained after COS. A less used approach is cryopreservation of ovarian tissue, consisting in freezing ovarian cortex fragments for a future thawing and graft. Some concerns and limitations regard the ovarian cortex graft and the risk of reintroducing malignant cells once performed. Nonetheless, the latter it is the only option in prepubertal patients.
Collapse
Affiliation(s)
- Marine Poulain
- FOCH Hospital, Gynecology Obstetric and Reproductive Medical Unit Department, Suresnes, France
- Université Paris Saclay, UVSQ, INRAE, BREED, 78350, Jouy-en-Josas, France - ENVA, BREED, 94700, Maison-Alfort, France
| | - Jessica Vandame
- FOCH Hospital, Gynecology Obstetric and Reproductive Medical Unit Department, Suresnes, France
| | - Chloé Tran
- FOCH Hospital, Gynecology Obstetric and Reproductive Medical Unit Department, Suresnes, France
| | - Sonia Koutchinsky
- FOCH Hospital, Gynecology Obstetric and Reproductive Medical Unit Department, Suresnes, France
| | - Paul Pirtea
- FOCH Hospital, Gynecology Obstetric and Reproductive Medical Unit Department, Suresnes, France
- Université Paris Saclay, UVSQ, INRAE, BREED, 78350, Jouy-en-Josas, France - ENVA, BREED, 94700, Maison-Alfort, France
| | - Jean-Marc Ayoubi
- FOCH Hospital, Gynecology Obstetric and Reproductive Medical Unit Department, Suresnes, France
- Université Paris Saclay, UVSQ, INRAE, BREED, 78350, Jouy-en-Josas, France - ENVA, BREED, 94700, Maison-Alfort, France
| |
Collapse
|
8
|
Kirillova A, Kovalskaya E, Brovkina O, Ekimov A, Bunyaeva E, Gordiev M, Mishieva N, Nazarenko T, Abubakirov A, Sukikh G. Cryopreservation of euploid blastocysts obtained after fertilization of in vitro matured ovarian tissue oocytes: a case report. J Assist Reprod Genet 2020; 37:905-911. [PMID: 32206960 PMCID: PMC7183014 DOI: 10.1007/s10815-020-01729-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 02/24/2020] [Indexed: 12/18/2022] Open
Abstract
With the increased rate of stable remission after gonadotoxic cancer treatment, new methods of fertility preservation are required in order to provide the best possible care for oncological patients. Here, we report an original case of euploid blastocyst cryopreservation after in vitro maturation of ovarian tissue oocytes (OTO IVM). Thirty-three oocytes were obtained from the ovarian tissue after ovariectomy in the breast cancer patient. Six out of 12 matured oocytes fertilized successfully and 3 blastocysts were formed. Genetic investigation for mutations associated with this type of malignancy found that the patient is not a carrier. Preimplantation genetic testing was performed only for aneuploidies and found all 3 blastocysts to be euploid and suitable for embryo transfer. Our study showed that the ovarian tissue oocytes matured in vitro have the potential for euploid blastocyst formation after ICSI which could be screened for aneuploidies and inherited mutations and then be vitrified in order to provide the best fertility preservation strategy for women with cancer.
Collapse
Affiliation(s)
- Anastasia Kirillova
- National Medical Research Center for Obstetrics, Gynecology and Perinatology named after Academician V.I.Kulakov, of the Ministry of Healthcare of Russian Federation, Moscow, Russia.
| | - Evgeniya Kovalskaya
- National Medical Research Center for Obstetrics, Gynecology and Perinatology named after Academician V.I.Kulakov, of the Ministry of Healthcare of Russian Federation, Moscow, Russia
| | - Olga Brovkina
- Federal Research and Clinical Center, FMBA of Russia, Moscow, Russia
| | - Aleksey Ekimov
- National Medical Research Center for Obstetrics, Gynecology and Perinatology named after Academician V.I.Kulakov, of the Ministry of Healthcare of Russian Federation, Moscow, Russia
| | - Ekaterina Bunyaeva
- National Medical Research Center for Obstetrics, Gynecology and Perinatology named after Academician V.I.Kulakov, of the Ministry of Healthcare of Russian Federation, Moscow, Russia
| | | | - Nona Mishieva
- National Medical Research Center for Obstetrics, Gynecology and Perinatology named after Academician V.I.Kulakov, of the Ministry of Healthcare of Russian Federation, Moscow, Russia
| | - Tatiana Nazarenko
- National Medical Research Center for Obstetrics, Gynecology and Perinatology named after Academician V.I.Kulakov, of the Ministry of Healthcare of Russian Federation, Moscow, Russia
| | - Aydar Abubakirov
- National Medical Research Center for Obstetrics, Gynecology and Perinatology named after Academician V.I.Kulakov, of the Ministry of Healthcare of Russian Federation, Moscow, Russia
| | - Gennady Sukikh
- National Medical Research Center for Obstetrics, Gynecology and Perinatology named after Academician V.I.Kulakov, of the Ministry of Healthcare of Russian Federation, Moscow, Russia
| |
Collapse
|
9
|
Lotz L, Dittrich R, Hoffmann I, Beckmann MW. Ovarian Tissue Transplantation: Experience From Germany and Worldwide Efficacy. CLINICAL MEDICINE INSIGHTS. REPRODUCTIVE HEALTH 2019; 13:1179558119867357. [PMID: 31431803 PMCID: PMC6685107 DOI: 10.1177/1179558119867357] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 07/12/2019] [Indexed: 12/29/2022]
Abstract
Extraction of ovarian tissue prior to oncologic therapy and subsequent transplantation is being performed increasingly often to preserve fertility in women. The procedure can be performed at any time of the cycle and, therefore, generally does not lead to any delay in oncological therapy. Success rates with transplantation of cryopreserved ovarian tissue have reached promising levels. More than 130 live births have been reported worldwide with the aid of cryopreserved ovarian tissue and the estimated birth rate is currently approximately 30%. In Germany, Austria, and Switzerland, the FertiPROTEKT consortium has successfully achieved 21 pregnancies and 17 deliveries generated after 95 ovarian tissue transplantations by 2015, one of the largest case series worldwide confirming that ovarian tissue cryopreservation and transplantation are successful. Approximately, more than 400 ovarian tissue cryopreservation procedures are performed each year in the FertiPROTEKT consortium, and the request and operations for ovarian tissue transplantation have increased in recent years. Therefore, recommendations for managing transplantation of ovarian tissue to German-speaking reproductive medicine centers were developed. In this overview, these recommendations and our experience in ovarian tissue transplantation are presented and discussed with international procedures.
Collapse
Affiliation(s)
- Laura Lotz
- Laura Lotz, Department of Obstetrics and Gynecology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nuremberg, Universitätsstrasse 21–23, D-91054 Erlangen, Germany.
| | | | | | | |
Collapse
|
10
|
Duniic. M, Krstic D, Stanisic S, Mirkovic M, Dunjic M, Milicevic N, Dunjic K, Dodic L. Ovarian Carcinoma Occurring After Breast Carcinoma Detection of Risk Factors Using Bdort Test. ACUPUNCTURE ELECTRO 2019. [DOI: 10.3727/036012919x15549226100437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Breast cancer is the most frequently diagnosed malignancy among women worldwide. There are many risk factors for breast carcinoma, such as genetics, Electro Magnetic Field Radiation (EMFs), the presence of human papillomaviruses (HPV), BRCA mutations, low vitamin D level, and toxins.
The risk of developing ovarian metastases increases with the passage of time after the breast cancer diagnosis, and there are significant links between the tumor stage and the development of ovarian metastases (expressed Mammoglobin B). Because there is no screening test for ovarian cancer
proven to be effective, women need to learn about their personal risk for developing ovarian cancer. This is especially true for the women already diagnosed with breast cancer, who should make sure if they are at risk for ovarian cancer. By using BiDigital O-Ring Test (BDORT), originated by
Prof. Omura, we can detect a lot of risk factors contributing to the risk for ovarian cancer, such as the presence ofHPV in the ovary, exposure to EMFs, food intolerance, low level oftelomeres, the presence of Borrelia Burgdorferi (BB), Tuberculosis (TB) etc. A high level of HPV 16 in the
ovary that is located on the same side as breast cancer could be detected, in addition to a high level of Integrin aSpl, as a marker of carcinogenesis. The cervix region infected by HPV is connected with the presence of HPV on both breasts, while the infection of one ovary (either left or
right) is connected with the diseased breast (with cancer) on the same side. We examined 38 patients with confirmed diagnosis of breast cancer, by using indirect BDORT and in all patients we found out one ovary with a high level of HPV infection as with high level of Integrin aSpl . There
are some mutual factors for the carcinogenesis of breast cancer and ovary cancer (HPV infection, BB, TB etc., toxins, EMFs). All patients reported being exposed to EMFs. 29 out ofa total of 38 (76.32%) patients had the infection of one ovary and the same side breast where exist breast carcinoma.
In the same time on same side ovary we detected high level of HPV 16 and 18 and high level of Integrin aSpl. In three of38 patients we found out increased ovary tumor markers. In study group we found out 29 out ofa total of38 patients have a risk of ovary carcinoma. What we can learn from
related literature is that ovarian carcinoma occurring after breast carcinoma is usually the metastasis of breast carcinoma; however, after this research, we believe that, in most cases, ovarian and breast carcinoma develop simultaneously, stimulated by the same or similar factors, and possibly
by the same carcinoma stem cell.
Collapse
|
11
|
Immuno-Isolating Dual Poly(ethylene glycol) Capsule Prevents Cancer Cells from Spreading Following Mouse Ovarian Tissue Auto-Transplantation. ACTA ACUST UNITED AC 2019; 2019. [PMID: 33969303 PMCID: PMC8101948 DOI: 10.20900/rmf20190006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
For female cancer survivors, premature ovarian insufficiency (POI) is a common complication of anticancer treatments. Ovarian tissue cryopreservation before treatment, followed by auto-transplantation after remission is a promising option to restore fertility and ovarian endocrine function. However, auto-transplantation is associated with the risk of re-introducing malignant cells harbored in the stroma of the ovarian autograft. To mitigate this risk, we investigated in this pilot study whether an immuno-isolating dual-layered poly(ethylene glycol)(PEG) capsule can retain cancer cells, while supporting folliculogenesis. The dual PEG capsule loaded with 1000 4T1 cancer cells retained 100% of the encapsulated cells in vitro for 21 days of culture. However, a greater cell load of 10,000 cells/capsule led to capsule failure and cells’ release. To assess the ability of the capsule to retain cancer cells, prevent metastasis, and support folliculogenesis in vivo we co-encapsulated cancer cells with ovarian tissue in the dual PEG capsule and implanted subcutaneously in mice. Control mice implanted with 2000 non-encapsulated cancer cells had tumors formed within 14 days and metastasis to the lungs. In contrast, no tumor mass formation or metastasis to the lungs was observed in mice with the same number of cancer cells encapsulated in the capsule. Our findings suggest that the immuno-isolating capsule may prevent the escape of the malignant cells potentially harbored in ovarian allografts and, in the future, improve the safety of ovarian tissue auto-transplantation in female cancer survivors.
Collapse
|
12
|
Tian W, Zhou Y, Wu M, Yao Y, Deng Y. Ovarian metastasis from breast cancer: a comprehensive review. Clin Transl Oncol 2018; 21:819-827. [DOI: 10.1007/s12094-018-02007-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 12/04/2018] [Indexed: 12/20/2022]
|
13
|
Fleury A, Pirrello O, Maugard C, Mathelin C, Linck C. Breast cancer and ovarian tissue cryopreservation: Review of the literature. J Gynecol Obstet Hum Reprod 2018; 47:351-357. [PMID: 29793036 DOI: 10.1016/j.jogoh.2018.05.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 05/10/2018] [Accepted: 05/17/2018] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Ovarian tissue cryopreservation is a modern technique of fertility preservation, useful before using ovariotoxic therapies in the treatment of breast cancer. The aim of our literature review was to study ovarian cryopreservation experiences for women with breast cancer, to identify guidelines, constraints and results in the oncological and obstetrical fields. METHODS We searched articles through the PubMed/Medline database, including all French and English references from January 2000 to October 2017. The combination of key words "breast cancer" and "ovarian tissue cryopreservation" allowed us to select 50 articles. We kept 18 publications which matched our subject. RESULTS Sixteen cases of ovarian transplants among patients treated for breast cancer were published with 14 pregnancies, 11 births and 3 failures. Two cases of breast recurrences were published after ovarian grafting. However, the hindsight in this technique is limited, with a first transplant published in 2004 and only a low number of cases. PERSPECTIVES A national census and comprehensive gathering of data among the patients treated for breast cancer using ovarian tissue cryopreservation would make it possible to better evaluate the occurrence of pregnancies and the carcinological risk of this technique.
Collapse
Affiliation(s)
- Audrey Fleury
- Unité de Sénologie, CHRU, Hôpitaux Universitaires de Strasbourg, Hôpital Hautepierre, 1 avenue Molière, 67200 Strasbourg, France.
| | - Olivier Pirrello
- CMCO - Centre Médico-Chirurgical et Obstétrical, 19 rue Louis Pasteur, 67303 Schiltigheim, France
| | - Christine Maugard
- Unité d'Oncogénétique Moléculaire, Hôpitaux Universitaires de Strasbourg, 1 place de l'Hôpital, 67091 Strasbourg, France; Unité d'Oncogénétique Clinique: Evaluation familiale et suivi, Hôpitaux Universitaires de Strasbourg, 1 place de l'Hôpital, 67091 Strasbourg, France
| | - Carole Mathelin
- Unité de Sénologie, CHRU, Hôpitaux Universitaires de Strasbourg, Hôpital Hautepierre, 1 avenue Molière, 67200 Strasbourg, France; IGBMC, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Biologie du Cancer, CNRS UMR 7104, INSERM U964, Université de Strasbourg, 67400 Illkirch, France
| | - Christelle Linck
- Unité de Sénologie, CHRU, Hôpitaux Universitaires de Strasbourg, Hôpital Hautepierre, 1 avenue Molière, 67200 Strasbourg, France
| |
Collapse
|
14
|
Comparison of the Oocyte Quality Derived from Two-Dimensional Follicle Culture Methods and Developmental Competence of In Vitro Grown and Matured Oocytes. BIOMED RESEARCH INTERNATIONAL 2018; 2018:7907092. [PMID: 29850567 PMCID: PMC5904821 DOI: 10.1155/2018/7907092] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 01/30/2018] [Accepted: 02/28/2018] [Indexed: 01/26/2023]
Abstract
In vitro follicle growth (IVFG) is an emerging fertility preservation technique, which can obtain fertilizable oocytes from an in vitro culture system in female. This study aimed to compare efficiency of the most widely used two-dimensional follicle culture methods [with or without oil layer (O+ or O- group)]. Preantral follicles were isolated from mice and randomly assigned. Follicles were cultured for 10 days and cumulus-oocyte complexes harvested 16-18 hours after hCG treatment. Follicle and oocyte growth, hormones in spent medium, meiotic spindle localization, expression of reactive oxygen species (ROS), mitochondrial activity, and gene expression were evaluated. In follicle growth, survival, pseudoantral cavity formation, ovulation, and oocyte maturation were also significantly higher in O+ group than O- group. Hormone production was significantly higher in follicles cultured in O+ than O-. There were no significant differences in mRNA expression related to development. On the other hand, the level of ROS was increased while the mitochondrial activity of in vitro grown matured oocyte was less than in vivo matured oocytes. In conclusion, follicle culture with O+ group appears to be superior to the culture in O- group in terms of follicle growth, development, oocyte growth, maturation, and microorganelles in oocyte.
Collapse
|
15
|
Masciangelo R, Bosisio C, Donnez J, Amorim CA, Dolmans MM. Safety of ovarian tissue transplantation in patients with borderline ovarian tumors. Hum Reprod 2017; 33:212-219. [DOI: 10.1093/humrep/dex352] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 11/03/2017] [Indexed: 11/13/2022] Open
Affiliation(s)
- Rossella Masciangelo
- Pôle de Recherche en Gynécologie, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Avenue Mounier 52, bte. B1.52.02, 1200 Brussels, Belgium
| | - Chiara Bosisio
- Pôle de Recherche en Gynécologie, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Avenue Mounier 52, bte. B1.52.02, 1200 Brussels, Belgium
| | - Jacques Donnez
- Société de Recherche pour l’Infertilité, Avenue Grandchamp 143, 1150 Brussels, Belgium
| | - Christiani A Amorim
- Pôle de Recherche en Gynécologie, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Avenue Mounier 52, bte. B1.52.02, 1200 Brussels, Belgium
| | - Marie-Madeleine Dolmans
- Pôle de Recherche en Gynécologie, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Avenue Mounier 52, bte. B1.52.02, 1200 Brussels, Belgium
- Département de Gynécologie, Cliniques Universitaires St. Luc, Avenue Hippocrate 10, 1200 Brussels, Belgium
| |
Collapse
|
16
|
Schüring AN, Fehm T, Behringer K, Goeckenjan M, Wimberger P, Henes M, Henes J, Fey MF, von Wolff M. Practical recommendations for fertility preservation in women by the FertiPROTEKT network. Part I: Indications for fertility preservation. Arch Gynecol Obstet 2017; 297:241-255. [PMID: 29177593 PMCID: PMC5762797 DOI: 10.1007/s00404-017-4594-3] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 11/10/2017] [Indexed: 12/11/2022]
Abstract
Purpose Most guidelines about fertility preservation are predominantly focused on scientific evidence, but are less practically orientated. Therefore, practically oriented recommendations are needed to support the clinician in daily practice. Methods A selective literature search was performed based on the clinical and scientific experience of the authors, focussing on the most relevant diseases and gynaecological cancers. This article (Part I) provides information on topics that are essential for the fertility preservation indication, such as disease prognosis, disease therapy and its associated risks to fertility, recommending disease-specific fertility preservation measures. Part II specifically focusses on fertility preservation techniques. Results In breast cancer patients, fertility preservation such as ovarian tissue and oocyte cryopreservation is especially recommended in low-stage cancer and in women < 35 years of age. In Hodgkin’s lymphoma, the indication is mainly based on the chemotherapy regime as some therapies have very low, others very high gonadotoxicity. In borderline ovarian tumours, preservation of fertility usually is achieved through fertility sparing surgery, ovarian stimulation may also be considered. In cervical cancer, endometrial cancer, rheumatic diseases and other malignancies such as Ewing sarcoma, colorectal carcinoma, non-Hodgkin lymphoma, leukaemia etc., several other factors must be considered to enable an individual, stage-dependent decision. Conclusion The decision for or against fertility preservation depends on the prognosis, the risks to fertility and individual factors such as prospective family planning.
Collapse
Affiliation(s)
- A N Schüring
- UKM Kinderwunschzentrum, Department of Gynaecology and Obstetrics, University Hospital of Münster, Albert-Schweitzer Campus 1, D-11, 48149, Münster, Germany.
| | - T Fehm
- Department of Gynaecology and Obstetrics, University Hospital of Düsseldorf, Düsseldorf, Germany
| | - K Behringer
- Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany
| | - M Goeckenjan
- Department of Gynaecology and Obstetrics, TU Dresden, Dresden, Germany
| | - P Wimberger
- Department of Gynaecology and Obstetrics, TU Dresden, Dresden, Germany
| | - M Henes
- Department of Women's Health, University of Tübingen, Tübingen, Germany
| | - J Henes
- Centre for Interdisciplinary Clinical Immunology, Rheumatology and Auto-inflammatory Diseases and Department of Internal Medicine II (Oncology, Hematology, Immunology, Rheumatology, Pulmology), University of Tübingen, Tübingen, Germany
| | - M F Fey
- Department of Medical Oncology, Inselspital and University of Berne, Berne, Switzerland
| | - M von Wolff
- Division of Gynaecological Endocrinology and Reproductive Medicine, University Women's Hospital, Berne, Switzerland
| |
Collapse
|
17
|
Srikanthan A, Amir E, Bedard P, Giuliani M, Hodgson D, Laframboise S, Prica A, Yee K, Greenblatt E, Lewin J, Gupta A. Fertility preservation in post-pubescent female cancer patients: A practical guideline for clinicians. Mol Clin Oncol 2017; 8:153-158. [PMID: 29387409 DOI: 10.3892/mco.2017.1486] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 10/30/2017] [Indexed: 11/06/2022] Open
Abstract
Increasing accessibility of fertility preservation (FP) options has permitted women to retain fertility following anticancer therapies. Several published guidelines have made recommendations for FP however their implementation into practice is currently unknown. In this review, we aim to provide oncology clinicians practical information about FP options for post-pubescent female cancer patients and recommendations for care delivery in order to answer preliminary questions and help triage whether FP referral is appropriate. Herein, we present a resource for oncology providers to guide them with FP discussions. Key points that are discussed in this critical review include: i) All cancer patients beginning a new plan of care should be informed of potential fertility risk. ii) If a woman requests further information on FP interventions, referral to a FP clinic should be made. iii) Given the evolving technologies in this area, patients should be informed of those which are proven and unproven, with oocyte and embryo preservation recognized as standard practice. iv) Random start (independent of menstrual cycle day) techniques are available to minimize oncologic treatment delays. v) Specific protocols for ovarian stimulation may be center-specific. vi) There is unlikely an increased cancer recurrence risk as a result of stimulation protocols in women with hormone-sensitive cancers. vii) Lastly, given the absence of consensus in the literature, routine use of GnRH analogs is not recommended for all cancer patients, however may be considered in select cases, such as high-risk women in whom definitive FP is not possible or feasible.
Collapse
Affiliation(s)
- Amirrtha Srikanthan
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON M5G 2M9, Canada
| | - Eitan Amir
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON M5G 2M9, Canada
| | - Philippe Bedard
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON M5G 2M9, Canada
| | - Meredith Giuliani
- Department of Radiation Oncology, Princess Margaret Cancer Centre, Toronto, ON M5G 2M9, Canada
| | - David Hodgson
- Department of Radiation Oncology, Princess Margaret Cancer Centre, Toronto, ON M5G 2M9, Canada
| | - Stephanie Laframboise
- Department of Gynecologic Oncology, Princess Margaret Cancer Centre, Toronto, ON M5G 2M9, Canada
| | - Anca Prica
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON M5G 2M9, Canada
| | - Karen Yee
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON M5G 2M9, Canada
| | - Ellen Greenblatt
- Mount Sinai Centre for Fertility and Reproductive Health, Mount Sinai Hospital, Toronto, ON M5T 2Z5, Canada
| | - Jeremy Lewin
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON M5G 2M9, Canada
| | - Abha Gupta
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON M5G 2M9, Canada.,Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| |
Collapse
|
18
|
Abstract
BACKGROUND Isolated adipose stem cells have been reported to encourage migration and early metastasis of breast cancer. Mimicking a surgical situation, the authors developed a human breast cancer model to evaluate in vivo whether human adipose tissue promotes tumor growth and invasion. METHODS Human adipose tissue was obtained from four patients. The MDA-MB-468 cell line was cultured with a lentiviral vector encoding a puromycin resistance gene and mCherry fluorescent protein. Virus-infected cells were selected. Animals were injected in the left renal capsule and divided into three experimental groups: group A, MDA-MB-468 cells (n = 4); group B, MDA-MB-468 cells/human adipose tissue (n = 4); and group C, Dulbecco's Modified Eagle Medium/F-12 medium (negative control, n = 4). Metastatic development was monitored using an in vivo imaging system. Small breast epithelial mucin (SBEM), human hypoxanthine-guanine phosphoribosyltransferase (HPRTh), and murine hypoxanthine-guanine phosphoribosyltransferase (HPRTm) expression were analyzed by real-time polymerase chain reaction to detect multifocal metastases in right/left renal capsule, liver, spleen, and pancreas. RESULTS Metastasis was observed between postinjection days 37 and 44. No significant differences were found in survival rates between groups (group A, 157 ± 42.60 days; group B, 169 ± 40.17 days). All samples expressed HPRTm. HPRTh and SBEM were expressed in left renal capsules from all group A and B mice, whereas in spleen, liver, pancreas, and right renal capsule the HPRTm and SBEM expression was not constant in all samples of group A and B mice. Differences were found between groups in HPRTh and SBEM expression but were not statistically significant. CONCLUSION Human adipose tissue used to restore breast defects after oncologic resection did not increase metastasis development risk when there were residual breast cancer cells in proximity.
Collapse
|
19
|
Peters ITA, van der Steen MA, Huisman BW, Hilders CGJM, Smit VTHBM, Vahrmeijer AL, Sier CFM, Trimbos JB, Kuppen PJK. Morphological and phenotypical features of ovarian metastases in breast cancer patients. BMC Cancer 2017; 17:206. [PMID: 28327103 PMCID: PMC5361796 DOI: 10.1186/s12885-017-3191-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 03/11/2017] [Indexed: 12/23/2022] Open
Abstract
Background Autotransplantation of frozen-thawed ovarian tissue is a method to preserve ovarian function and fertility in patients undergoing gonadotoxic therapy. In oncology patients, the safety cannot yet be guaranteed, since current tumor detection methods can only exclude the presence of malignant cells in ovarian fragments that are not transplanted. We determined the need for a novel detection method by studying the distribution of tumor cells in ovaries from patients with breast cancer. Furthermore, we examined which cell-surface proteins are suitable as a target for non-invasive tumor-specific imaging of ovarian metastases from invasive breast cancer. Methods Using the nationwide database of the Dutch Pathology Registry (PALGA), we identified a cohort of 46 women with primary invasive breast cancer and ovarian metastases. The localization and morphology of ovarian metastases were determined on hematoxylin-and-eosin-stained sections. The following cell-surface markers were immunohistochemically analyzed: E-cadherin, epithelial membrane antigen (EMA), human epidermal growth receptor type 2 (Her2/neu), carcinoembryonic antigen (CEA), αvβ6 integrin and epithelial cell adhesion molecule (EpCAM). Results The majority of ovarian metastases (71%) consisted of a solitary metastasis or multiple distinct nodules separated by uninvolved ovarian tissue, suggesting that ovarian metastases might be overlooked by the current detection approach. Combining the targets E-cadherin, EMA and Her2/neu resulted in nearly 100% detection of ductal ovarian metastases, whereas the combination of EMA, Her2/neu and EpCAM was most suitable to detect lobular ovarian metastases. Conclusions Examination of the actual ovarian transplants is recommended. A combination of targets is most appropriate to detect ovarian metastases by tumor-specific imaging. Electronic supplementary material The online version of this article (doi:10.1186/s12885-017-3191-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Inge T A Peters
- Department of Gynecology, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Bertine W Huisman
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Vincent T H B M Smit
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Cornelis F M Sier
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - J Baptist Trimbos
- Department of Gynecology, Leiden University Medical Center, Leiden, the Netherlands
| | - Peter J K Kuppen
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
20
|
Bergamini ML, Maugard CM, Mathelin C. [Do controlled ovarian hyperstimulations and cryopreservations promote recurrences after breast cancer?]. ACTA ACUST UNITED AC 2017; 45:172-179. [PMID: 28259703 DOI: 10.1016/j.gofs.2017.01.008] [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: 10/31/2016] [Accepted: 12/26/2016] [Indexed: 11/18/2022]
Abstract
The objective of this review was to identify recurrences (ipsilateral, contralateral, metastases and deaths) occurring after controlled ovarian hyperstimulation (COH) or cryopreservation of ovarian tissue (CPTO) for patients treated for a breast cancer. METHODS We performed a bibliographical research through the Pubmed/Medline database, including all the references from January 2006 until September 2016, in French or in English, after exclusion of animal studies. The keywords association "breast neoplasms", "fertility preservation", "reproductive techniques", "ovarian cryopreservation" and "in vitro fertilization" allowed the selection of 852 publications among which only 6 were selected because they included data on recurrence and long term follow up. Four publications involved HSC (3 before breast cancer treatment and 1 after) and 2 concerned CPTO with re-implantation. RESULTS This analysis has not shown increasing of breast recurrences after HSC and CPTO. However, results were not statistically significant, due to several biases in particular heterogeneousness of the groups of patients. CONCLUSION A survey of patients who used fertility preservation or assisted reproductive technologies after breast cancer would be helpful to better estimate their oncological risk.
Collapse
Affiliation(s)
- M-L Bergamini
- Unité de sénologie, hôpital Hautepierre, hôpitaux universitaires de Strasbourg, CHRU, 1, avenue Molière, 67200 Strasbourg, France.
| | - C M Maugard
- Unité d'oncogénétique moléculaire, hôpitaux universitaires de Strasbourg, 1, place de l'Hôpital, 67091 Strasbourg, France; Unité d'oncogénétique clinique : évaluation familiale et suivi, hôpitaux universitaires de Strasbourg, 1, place de l'Hôpital, 67091 Strasbourg, France
| | - C Mathelin
- Unité de sénologie, hôpital Hautepierre, hôpitaux universitaires de Strasbourg, CHRU, 1, avenue Molière, 67200 Strasbourg, France; IGBMC, institut de génétique et de biologie moléculaire et cellulaire, biologie du cancer, CNRS UMR 7104, Inserm U964, université de Strasbourg, 67400 Illkirch, France
| |
Collapse
|
21
|
Prevalence and Risk Factors of Ovarian Metastases in Breast Cancer Patients < 41 Years of Age in the Netherlands: A Nationwide Retrospective Cohort Study. PLoS One 2017; 12:e0168277. [PMID: 28125710 PMCID: PMC5268771 DOI: 10.1371/journal.pone.0168277] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 11/29/2016] [Indexed: 11/22/2022] Open
Abstract
Purpose Breast cancer is one of the primary indications for cryopreservation and subsequent autotransplantation of ovarian tissue. The safety of this fertility preservation method remains questionable, as the presence of disseminated breast tumor cells cannot yet be excluded in the ovarian autografts. We explored the prevalence of ovarian metastases among young breast cancer patients and determined risk factors for the development of ovarian metastases. Methods Using the nationwide database of the Dutch Pathology Registry (PALGA), we identified a cohort of 2648 women with primary invasive breast cancer at age < 41 years in the period 2000–2010 in the Netherlands who subsequently underwent an oophorectomy. From this source population, all cases who had histologically confirmed ovarian metastases were included. For each case of whom clinical data were available, one control without ovarian metastases who matched the time interval between breast cancer diagnosis and oophorectomy was selected. Data were collected on patient characteristics, diagnosis, treatment and follow-up. Results Ovarian metastases were found in 63 out of 2648 patients who met the inclusion criteria. The risk of developing ovarian metastases increased with time passed since breast cancer diagnosis. Multivariate logistic regression analyses showed significant association between tumor stage and the development of ovarian metastases (p = 0.024). Conclusions The prevalence of ovarian metastases was 2.4% among young breast cancer patients. Early ovary removal may reduce the risk of developing ovarian metastases. In breast cancer patients with tumors > 5 cm and/or inflammatory carcinoma, we recommend a cautious approach to ovarian tissue autotransplantation.
Collapse
|
22
|
Peters ITA, Hilders CGJM, Sier CFM, Vahrmeijer AL, Smit VTHBM, Baptist Trimbos J, Kuppen PJK. Identification of cell-surface markers for detecting breast cancer cells in ovarian tissue. Arch Gynecol Obstet 2016; 294:385-93. [PMID: 26946151 PMCID: PMC4937067 DOI: 10.1007/s00404-016-4036-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 02/03/2016] [Indexed: 02/07/2023]
Abstract
PURPOSE The safety of ovarian tissue autotransplantation in oncology patients cannot be ensured, as current tumor-detection methods compromise the ovarian tissue viability. Although non-destructive methods (for instance near-infrared fluorescence imaging) can discriminate malignant from healthy tissues while leaving the examined tissues unaffected, they require specific cell-surface tumor markers. We determined which tumor markers are suitable targets for tumor-specific imaging to exclude the presence of breast cancer cells in ovarian tissue. METHODS Immunohistochemistry was performed on formalin-fixed, paraffin-embedded specimens of ten ovaries from premenopausal patients. Additionally, we screened a tissue microarray containing tumor tissue cores from 24 breast cancer patients being eligible for ovarian tissue cryopreservation. The following cell-surface tumor markers were tested: E-cadherin, EMA (epithelial membrane antigen), Her2/neu (human epidermal growth factor receptor type 2), αvβ6 integrin, EpCAM (epithelial cell adhesion molecule), CEA (carcinoembryonic antigen), FR-α (folate receptor-alpha), and uPAR (urokinase-type plasminogen activator receptor). For each tumor, the percentage of positive breast tumor cells was measured. RESULTS None of the ten ovaries were positive for any of the markers tested. However, all markers (except CEA and uPAR) were present on epithelial cells of inclusion cysts. E-cadherin was present in the majority of breast tumors: ≥90 % of tumor cells were positive for E-cadherin in 17 out of 24 tumors, and 100 % of tumor cells were positive in 5 out of 24 tumors. CONCLUSIONS Of the markers tested, E-cadherin is the most suitable marker for a tumor-specific probe in ovarian tissue. Methods are required to distinguish inclusion cysts from breast tumor cells.
Collapse
Affiliation(s)
- Inge T A Peters
- Department of Gynecology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Cornelis F M Sier
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Vincent T H B M Smit
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - J Baptist Trimbos
- Department of Gynecology, Leiden University Medical Center, Leiden, The Netherlands
| | - Peter J K Kuppen
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
23
|
Abstract
PURPOSE OF REVIEW This article aims to carefully evaluate a number of critical points related to ovarian tissue freezing and presents factual data in terms of live birth rates and risks. RECENT FINDINGS Reimplantation of frozen-thawed ovarian tissue remains an experimental procedure according to the American Society for Reproductive Medicine, despite almost 40 live births reported in the literature. Recent literature on the topic has focused on the risk of reimplanting malignant cells, so the present review assesses the risks according to disease. SUMMARY This manuscript emphasizes the crucial importance of not only preserving fertility in young women but also clearly explaining to patients the different available options and their respective success rates. Some previously published reviews have reported inaccurate reimplantation success rates. In this review, we report the true picture, with a live birth rate of 25%. Ovarian tissue freezing may be combined with pickup of immature oocytes (at the time of ovarian biopsy and tissue removal) or mature oocytes (if chemotherapy can be delayed).
Collapse
|
24
|
Kniazeva E, Hardy AN, Boukaidi SA, Woodruff TK, Jeruss JS, Shea LD. Primordial Follicle Transplantation within Designer Biomaterial Grafts Produce Live Births in a Mouse Infertility Model. Sci Rep 2015; 5:17709. [PMID: 26633657 PMCID: PMC4668556 DOI: 10.1038/srep17709] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 11/04/2015] [Indexed: 11/09/2022] Open
Abstract
The gonadotoxic effects of chemotherapy and radiation may result in premature ovarian failure in premenopausal oncology patients. Although autotransplantation of ovarian tissue has led to successful live births, reintroduction of latent malignant cells inducing relapse is a significant concern. In this report, we investigated the design of biomaterial grafts for transplantation of isolated ovarian follicles as a means to preserve fertility. Primordial and primary ovarian follicles from young female mice were extracted and encapsulated into biomaterials for subsequent transplantation into adult mice. Among the formulations tested, aggregated follicles encapsulated within fibrin had enhanced survival and integration with the host tissue following transplantation relative to the fibrin-alginate and fibrin-collagen composites. All mice transplanted with fibrin-encapsulated follicles resumed cycling, and live births were achieved only for follicles transplanted within VEGF-loaded fibrin beads. The extent to which these procedures reduce the presence of metastatic breast cancer cells among the isolated follicles was evaluated, with significantly reduced numbers of cancer cells present relative to intact ovaries. This ability to obtain live births by transplanting isolated primordial and primary follicles, while also reducing the risk of re-seeding disease relative to ovarian tissue transplantation, may ultimately provide a means to preserve fertility in premenopausal oncology patients.
Collapse
Affiliation(s)
- E Kniazeva
- Department of Chemical and Biological Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL 60208, USA.,Department of Obstetrics and Gynecology, Institute for Women's Health Research, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - A N Hardy
- Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - S A Boukaidi
- Department of Obstetrics and Gynecology and Reproductive Medicine, CHU de Nice, Archet 2 Hospital, Nice, France
| | - T K Woodruff
- Department of Obstetrics and Gynecology, Institute for Women's Health Research, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - J S Jeruss
- Department of Obstetrics and Gynecology, Institute for Women's Health Research, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.,Department of Surgery, University of Michigan, Ann Arbor, MI 48109, USA.,Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - L D Shea
- Department of Chemical and Biological Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL 60208, USA.,Department of Obstetrics and Gynecology, Institute for Women's Health Research, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.,Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
25
|
Rodríguez-Iglesias B, Novella-Maestre E, Herraiz S, Díaz-García C, Pellicer N, Pellicer A. New methods to improve the safety assessment of cryopreserved ovarian tissue for fertility preservation in breast cancer patients. Fertil Steril 2015; 104:1493-502.e1-2. [DOI: 10.1016/j.fertnstert.2015.08.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 08/06/2015] [Accepted: 08/06/2015] [Indexed: 12/16/2022]
|
26
|
Chiti MC, Dolmans MM, Orellana R, Soares M, Paulini F, Donnez J, Amorim CA. Influence of follicle stage on artificial ovary outcome using fibrin as a matrix. Hum Reprod 2015; 31:427-35. [PMID: 26628641 DOI: 10.1093/humrep/dev299] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 11/06/2015] [Indexed: 01/01/2023] Open
Abstract
STUDY QUESTION Do primordial-primary versus secondary follicles embedded inside a fibrin matrix have different capabilities to survive and grow after isolation and transplantation? SUMMARY ANSWER Mouse primordial-primary follicles showed a lower recovery rate than secondary follicles, but both were able to grow. WHAT IS KNOWN ALREADY Fresh isolated mouse follicles and ovarian stromal cells embedded in a fibrin matrix are capable of surviving and developing after short-term autografting. STUDY DESIGN, SIZE, DURATION In vivo experimental model using 11 donor Naval Medical Research Institute (NMRI) mice and 11 recipient severe combined immunodeficiency (SCID) mice. Both ovaries from all NMRI mice were mechanically disrupted and primordial-primary and secondary follicles were isolated with ovarian stromal cells. They were then encapsulated in a fibrin matrix composed of 12.5 mg/ml of fibrinogen (F12.5) and 1 IU/ml of thrombin (T1) (F12.5/T1), and grafted to the inner part of the peritoneum of SCID mice for 2 and 7 days. PARTICIPANTS/MATERIALS, SETTING, METHODS This study was conducted at the Gynecology Research Unit, Université Catholique de Louvain. All materials were used to conduct histological (H-E staining) and immunohistochemical (Ki67, TUNEL) analyses. MAIN RESULTS AND THE ROLE OF CHANCE Although all grafted fibrin clots were recovered, the follicle recovery rate on day 2 was 16 and 40% for primordial-primary and secondary follicles respectively, while on day 7, it was 6 and 28%. The secondary group showed a significantly higher recovery rate than the primordial-primary group (23%, P-value <0.001). Follicles found in both groups were viable, as demonstrated by live/dead assays, and no difference was observed in the apoptosis rate between groups, as evidenced by TUNEL. Their growth to further stages was confirmed by Ki67 immunostaining. LIMITATIONS, REASONS FOR CAUTION As demonstrated by our results, secondary follicles appear to be more likely to survive and develop than primordial-primary follicles in a fibrin matrix after both periods of grafting. These findings may also be attributed to the specific features of the fibrin matrix, which could benefit larger follicles, but not smaller follicles. WIDER IMPLICATIONS OF THE FINDINGS This study is essential to understanding possible impairment caused by factors such as the isolation procedure or fibrin matrix composition to the survival and development of different follicle stages. It therefore provides the basis for further investigations with longer periods of grafting. STUDY FUNDING/COMPETING INTERESTS This study was supported by grants from the Fonds National de la Recherche Scientifique de Belgique (grant Télévie No. 7.4578.14 and 7.4627.13, grant 5/4/150/5 awarded to Marie-Madeleine Dolmans), Fonds Spéciaux de Recherche, Fondation St Luc, the Foundation Against Cancer, and the Region Wallone (Convention N°6519-OVART) and donations from Mr Pietro Ferrero, Baron Frère and Viscount Philippe de Spoelberch. None of the authors have any competing interests to declare.
Collapse
Affiliation(s)
- M C Chiti
- Pôle de Recherche en Gynécologie, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Avenue Mounier 52, bte. B1.52.02, 1200 Brussels, Belgium
| | - M M Dolmans
- Pôle de Recherche en Gynécologie, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Avenue Mounier 52, bte. B1.52.02, 1200 Brussels, Belgium Gynecology Department, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - R Orellana
- Pôle de Recherche en Gynécologie, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Avenue Mounier 52, bte. B1.52.02, 1200 Brussels, Belgium
| | - M Soares
- Pôle de Recherche en Gynécologie, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Avenue Mounier 52, bte. B1.52.02, 1200 Brussels, Belgium Gynecology Department, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - F Paulini
- Pôle de Recherche en Gynécologie, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Avenue Mounier 52, bte. B1.52.02, 1200 Brussels, Belgium
| | - J Donnez
- Society for Research into Infertility, Brussels, Belgium
| | - C A Amorim
- Pôle de Recherche en Gynécologie, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Avenue Mounier 52, bte. B1.52.02, 1200 Brussels, Belgium
| |
Collapse
|
27
|
Bockstaele L, Boulenouar S, Van Den Steen G, Dechène J, Tsepelidis S, Craciun L, Noël JC, Demeestere I. Evaluation of quantitative polymerase chain reaction markers for the detection of breast cancer cells in ovarian tissue stored for fertility preservation. Fertil Steril 2015; 104:410-7.e4. [DOI: 10.1016/j.fertnstert.2015.04.036] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 04/24/2015] [Accepted: 04/28/2015] [Indexed: 12/20/2022]
|
28
|
Soares M, Sahrari K, Chiti M, Amorim C, Ambroise J, Donnez J, Dolmans MM. The best source of isolated stromal cells for the artificial ovary: medulla or cortex, cryopreserved or fresh? Hum Reprod 2015; 30:1589-98. [DOI: 10.1093/humrep/dev101] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 04/20/2015] [Indexed: 01/28/2023] Open
|
29
|
Dittrich R, Lotz L, Fehm T, Krüssel J, von Wolff M, Toth B, van der Ven H, Schüring AN, Würfel W, Hoffmann I, Beckmann MW. Xenotransplantation of cryopreserved human ovarian tissue--a systematic review of MII oocyte maturation and discussion of it as a realistic option for restoring fertility after cancer treatment. Fertil Steril 2015; 103:1557-65. [PMID: 25881879 DOI: 10.1016/j.fertnstert.2015.03.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Revised: 02/28/2015] [Accepted: 03/02/2015] [Indexed: 12/25/2022]
Abstract
OBJECTIVE To systematically review the reporting of MII (MII) oocyte development after xenotransplantation of human ovarian tissue. DESIGN Systematic review in accordance with the guidelines of the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA). SETTING Not applicable. PATIENT(S) Not applicable. INTERVENTION(S) Formation of MII oocytes after xenotransplantation of human ovarian tissue. MAIN OUTCOME MEASURE(S) Any outcome reported in Pubmed. RESULT(S) Six publications were identified that report on formation of MII oocytes after xenotransplantation of human ovarian tissue. CONCLUSION(S) Xenografting of human ovarian tissue has proved to be a useful model for examining ovarian function and follicle development in vivo. With human follicles that have matured through xenografting, the possibility of cancer transmission and relapse can also be eliminated, because cancer cells are not able to penetrate the zona pellucida. The reported studies have demonstrated that xenografted ovarian tissue from a range of species, including humans, can produce antral follicles that contain mature (MII) oocytes, and it has been shown that mice oocytes have the potential to give rise to live young. Although some ethical questions remain unresolved, xenotransplantation may be a promising method for restoring fertility. This review furthermore describes the value of xenotransplantation as a tool in reproductive biology and discusses the ethical and potential safety issues regarding ovarian tissue xenotransplantation as a means of recovering fertility.
Collapse
Affiliation(s)
- Ralf Dittrich
- Department of Obstetrics and Gynecology, Erlangen University Hospital, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany.
| | - Laura Lotz
- Department of Obstetrics and Gynecology, Erlangen University Hospital, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Tanja Fehm
- Department of Obstetrics and Gynecology, Düsseldorf University Hospital, Düsseldorf, Germany
| | - Jan Krüssel
- Department of Obstetrics and Gynecology, Düsseldorf University Hospital, Düsseldorf, Germany
| | - Michael von Wolff
- Division of Gynecologic Endocrinology and Reproductive Medicine, University Women's Hospital, Berne, Switzerland
| | - Bettina Toth
- Department of Gynecologic Endocrinology and Fertility Disorders, Ruprecht-Karls University Hospital, Heidelberg, Germany
| | - Hans van der Ven
- Department of Obstetrics and Gynecology, Bonn University Hospital, Bonn, Germany
| | - Andreas N Schüring
- Department of Obstetrics and Gynecology, UKM Kinderwunschzentrum, Münster University Hospital, Münster, Germany
| | | | - Inge Hoffmann
- Department of Obstetrics and Gynecology, Erlangen University Hospital, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Matthias W Beckmann
- Department of Obstetrics and Gynecology, Erlangen University Hospital, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
30
|
Fertilitätsprotektion bei Mammakarzinom. GYNAKOLOGISCHE ENDOKRINOLOGIE 2015. [DOI: 10.1007/s10304-014-0652-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
31
|
Hoekman EJ, Smit VTHBM, Fleming TP, Louwe LA, Fleuren GJ, Hilders CGJM. Searching for metastases in ovarian tissue before autotransplantation: a tailor-made approach. Fertil Steril 2014; 103:469-77. [PMID: 25497447 DOI: 10.1016/j.fertnstert.2014.11.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Revised: 11/03/2014] [Accepted: 11/03/2014] [Indexed: 11/27/2022]
Abstract
OBJECTIVE To exclude minimal residual disease in remaining ovarian tissue after harvesting the ovarian cortex for cryopreservation, by means of a tailor-made approach. DESIGN Retrospective case series. SETTING Hospital laboratory. PATIENT(S) We evaluated the ovarian and tubal tissue from 47 cancer patients (breast cancer, [non-]Hodgkin lymphoma; osteo-, Ewing, myxoid lipo-, and oropharyngeal synovial sarcoma; cervical, rectal, and esophageal cancer), who had stored ovarian tissue for fertility preservation. INTERVENTION(S) Immunohistochemistry (IHC) with tumor-related antibodies and genetic mutation analysis were performed to detect micrometastases by multiple sectioning at three levels of the paraffin-embedded formalin-fixed material. Molecular assays were performed with the use of tissue between these three levels of sectioning. MAIN OUTCOME MEASURE(S) Detection of micrometastases in ovaries. RESULT(S) We analyzed 847 ovarian slides to detect isolated tumor cells (ITCs) or micrometastases by IHC. In only one case (1/47) were ITCs detected in the fallopian tube. That patient had an intra-abdominal metastatic esophageal carcinoma. Additional DNA analyses of breast and rectal cancer, Ewing sarcoma, and human papilloma virus in cervical patients did not show evidence of micrometastases in the ovarian tissue. CONCLUSION(S) The tailor-made approach consisted of patient-specific tumor markers which were used to search for ovarian micrometastases. We found evidence of metastatic disease within the fallopian tube of a patient with intraperitoneal metastatic esophageal adenocarcinoma.
Collapse
Affiliation(s)
- Ellen J Hoekman
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands; Department of Gynecology, Leiden University Medical Center, Leiden, the Netherlands.
| | - Vincent T H B M Smit
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | - Timothy P Fleming
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Leonie A Louwe
- Department of Gynecology, Leiden University Medical Center, Leiden, the Netherlands
| | - Gert Jan Fleuren
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | - Carina G J M Hilders
- Department of Gynecology, Leiden University Medical Center, Leiden, the Netherlands; Department of Gynecology, Reinier de Graaf Gasthuis Delft, Delft, the Netherlands
| |
Collapse
|
32
|
Abstract
In women, ∼10% of cancers occur in those <45 years old. Chemotherapy, radiotherapy and bone marrow transplantation can cure >90% of girls and young women with diseases that require such treatments. However, these treatments can result in premature ovarian failure, depending on the follicular reserve, the age of the patient and the type and dose of drugs used. This article discusses the different fertility preservation strategies: medical therapy before chemotherapy; ovarian transposition; embryo cryopreservation; oocyte vitrification; and ovarian tissue cryopreservation. The indications, results and risks of these options are discussed. Whether medical therapy should be used to protect the gonads during chemotherapy remains a source of debate. Fertility preservation needs to be completed before chemotherapy and/or irradiation is started and might take 2-3 weeks with established techniques such as embryo or oocyte cryopreservation. Further studies are needed in patients with cancer to confirm the excellent outcomes obtained in patients without cancer or in egg donation programmes. For prepubertal girls or cases where immediate therapy is required, cryopreservation of ovarian tissue is the only available option. Finally, possible future approaches are reviewed, including in vitro maturation of nonantral follicles, the artificial ovary, oogonial stem cells and drugs to prevent follicle loss.
Collapse
Affiliation(s)
- Jacques Donnez
- Société de Recherche pour l'Infertilité, Avenue Grandchamp, 143, B-1150 Brussels, Belgium
| | | |
Collapse
|