1
|
Bonomi M, Cangiano B, Cianfarani S, Garolla A, Gianfrilli D, Lanfranco F, Rastrelli G, Sbardella E, Corona G, Isidori AM, Rochira V. "Management of andrological disorders from childhood and adolescence to transition age: guidelines from the Italian Society of Andrology and Sexual Medicine (SIAMS) in collaboration with the Italian Society for Pediatric Endocrinology and Diabetology (SIEDP)-Part-1". J Endocrinol Invest 2025; 48:1-22. [PMID: 39126560 DOI: 10.1007/s40618-024-02435-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024]
Abstract
PURPOSE Andrological pathologies in the adulthood are often the results of conditions that originate during childhood and adolescence and sometimes even during gestation and neonatal period. Unfortunately, the reports in the literature concerning pediatric andrological diseases are scares and mainly concerning single issues. Furthermore, no shared position statement are so far available. METHODS The Italian Society of Andrology and Sexual Medicine (SIAMS) commissioned an expert task force involving the Italian Society of Pediatric Endocrinology and Diabetology (SIEDP) to provide an updated guideline on the diagnosis and management of andrological disorders from childhood and adolescence to transition age. Derived recommendations were based on the grading of recommendations, assessment, development, and evaluation (GRADE) system. RESULTS A literature search of articles in English for the term "varicoceles", "gynecomastia", "fertility preservation", "macroorchidism", "precocious puberty" and "pubertal delay" has been performed. Three major aspects for each considered disorder were assessed including diagnosis, clinical management, and treatment. Recommendations and suggestions have been provided for each of the mentioned andrological disorders. CONCLUSIONS These are the first guidelines based on a multidisciplinary approach that involves important societies related to the field of andrological medicine from pediatric to transition and adult ages. This fruitful discussion allowed for a general agreement on several recommendations and suggestions to be reached, which can support all stakeholders in improving andrological and general health of the transitional age.
Collapse
Affiliation(s)
- M Bonomi
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy.
- Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Piazzale Brescia 20, 20149, Milan, Italy.
| | - B Cangiano
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
- Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Piazzale Brescia 20, 20149, Milan, Italy
| | - S Cianfarani
- Endocrinology and Diabetes Unit, Bambino Gesù Children's Hospital, Rome, Italy
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
- Department of Women's and Children's Health, Karolinska Institute, Stockholm, Sweden
| | - A Garolla
- Unit of Andrology and Reproductive Medicine, Department of Medicine, University of Padova, Padua, Italy
| | - D Gianfrilli
- Section of Medical Pathophysiology and Endocrinology, Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
- Centre for Rare Diseases (Endo-ERN Accredited), Policlinico Umberto I, Rome, Italy
| | - F Lanfranco
- Division of Endocrinology, Andrology and Metabolism, Department of Medical Sciences, Humanitas Gradenigo, University of Turin, Turin, Italy
| | - G Rastrelli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Careggi Hospital, University of Florence, Florence, Italy
| | - E Sbardella
- Section of Medical Pathophysiology and Endocrinology, Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
- Centre for Rare Diseases (Endo-ERN Accredited), Policlinico Umberto I, Rome, Italy
| | - G Corona
- Endocrinology Unit, Medical Department, Maggiore-Bellaria Hospital, Azienda Usl, Bologna, Italy
| | - A M Isidori
- Section of Medical Pathophysiology and Endocrinology, Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
- Centre for Rare Diseases (Endo-ERN Accredited), Policlinico Umberto I, Rome, Italy
| | - V Rochira
- Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy.
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria Di Modena Policlinico Di Modena, Ospedale Civile Di Baggiovara, Via Giardini 1355, 41126, Modena, Italy.
| |
Collapse
|
2
|
Tholeti P, Koulmane Laxminarayana SL, Lakshmi VR, Bhat VK, Kumar P V, Uppangala S, Kalthur G, Spears N, Adiga SK. Spermatogonial quantity in prepubertal boys undergoing fertility preservation is comparable between haematological and non-haematological cancers. HUM FERTIL 2024; 27:2362980. [PMID: 38842163 DOI: 10.1080/14647273.2024.2362980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 05/26/2024] [Indexed: 06/07/2024]
Abstract
Fertility restoration potential of immature testicular tissue (ITT) depends on the number of spermatogonial cells in the retrieved tissue prior to cryopreservation in oncofertility programme. There are limited data on the association between type of malignancy and testicular germ cell population. Hence, this study is aimed to investigate the spermatogonial and Sertoli cell population in ITT retrieved from 14 pre-pubertal boys who opted for fertility preservation. Histopathological and immunochemical analysis of seminiferous tubules from haematological (N = 7) and non-haematological (N = 7) malignant patients revealed 3.43 ± 2.92 and 1.71 ± 1.81 spermatogonia per tubular cross section (S/T), respectively. The Sertoli cell number was comparable between haematological and non-haematological group (18.42 ± 3.78 and 22.03 ± 10.43). Spermatogonial quantity in ITT did not vary significantly between haematological and non-haematological cancers. This observation, though preliminary, would contribute to the limited literature on paediatric male oncofertility.
Collapse
Affiliation(s)
- Prathima Tholeti
- Centre for Fertility Preservation, Centre of Excellence in Clinical Embryology, Department of Reproductive Science, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | | | - Vani R Lakshmi
- Department of Data Science, Prasanna School of Public Health, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Vasudeva K Bhat
- Department of Pediatric Hematology and Oncology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Vijay Kumar P
- Department of Pediatric Surgery, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Shubhashree Uppangala
- Division of Reproductive Genetics, Department of Reproductive Science, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Guruprasad Kalthur
- Division of Reproductive Biology, Department of Reproductive Science, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Norah Spears
- Biomedical Sciences, Hugh Robson Building, University of Edinburgh, Edinburgh, United Kingdom
| | - Satish Kumar Adiga
- Centre for Fertility Preservation, Centre of Excellence in Clinical Embryology, Department of Reproductive Science, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|
3
|
Damyanova KB, Nixon B, Johnston SD, Gambini A, Benitez PP, Lord T. Spermatogonial stem cell technologies: applications from human medicine to wildlife conservation†. Biol Reprod 2024; 111:757-779. [PMID: 38993049 PMCID: PMC11473898 DOI: 10.1093/biolre/ioae109] [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: 05/01/2024] [Revised: 07/05/2024] [Accepted: 07/10/2024] [Indexed: 07/13/2024] Open
Abstract
Spermatogonial stem cell (SSC) technologies that are currently under clinical development to reverse human infertility hold the potential to be adapted and applied for the conservation of endangered and vulnerable wildlife species. The biobanking of testis tissue containing SSCs from wildlife species, aligned with that occurring in pediatric human patients, could facilitate strategies to improve the genetic diversity and fitness of endangered populations. Approaches to utilize these SSCs could include spermatogonial transplantation or testis tissue grafting into a donor animal of the same or a closely related species, or in vitro spermatogenesis paired with assisted reproduction approaches. The primary roadblock to progress in this field is a lack of fundamental knowledge of SSC biology in non-model species. Herein, we review the current understanding of molecular mechanisms controlling SSC function in laboratory rodents and humans, and given our particular interest in the conservation of Australian marsupials, use a subset of these species as a case-study to demonstrate gaps-in-knowledge that are common to wildlife. Additionally, we review progress in the development and application of SSC technologies in fertility clinics and consider the translation potential of these techniques for species conservation pipelines.
Collapse
Affiliation(s)
- Katerina B Damyanova
- Discipline of Biological Sciences, The University of Newcastle, Callaghan, NSW 2308, Australia
- Infertility and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| | - Brett Nixon
- Discipline of Biological Sciences, The University of Newcastle, Callaghan, NSW 2308, Australia
- Infertility and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| | - Stephen D Johnston
- School of Environment, The University of Queensland, Gatton, QLD 4343, Australia
- School of Veterinary Science, The University of Queensland, Gatton, QLD 4343, Australia
| | - Andrés Gambini
- School of Veterinary Science, The University of Queensland, Gatton, QLD 4343, Australia
- School of Agriculture and Food Science, The University of Queensland, Gatton, QLD 4343, Australia
| | - Patricio P Benitez
- School of Agriculture and Food Science, The University of Queensland, Gatton, QLD 4343, Australia
| | - Tessa Lord
- Discipline of Biological Sciences, The University of Newcastle, Callaghan, NSW 2308, Australia
- Infertility and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| |
Collapse
|
4
|
Sockel K, Neu A, Goeckenjan M, Ditschkowski M, Hilgendorf I, Kröger N, Ayuk FA, Stoelzel F, Middeke JM, Eder M, Bethge W, Finke J, Bertz H, Kobbe G, Kaufmann M, Platzbecker U, Beverungen D, Schmid C, von Bonin M, Egger-Heidrich K, Heberling L, Trautmann-Grill K, Teipel R, Bug G, Tischer J, Fraccaroli A, Fante M, Wolff D, Luft T, Winkler J, Schäfer-Eckart K, Scheid C, Holtick U, Klein S, Blau IW, Burchert A, Wulf G, Hasenkamp J, Schwerdtfeger R, Kaun S, Junghanss C, Wortmann F, Winter S, Neidlinger H, Theuser C, Beyersmann J, Bornhaeuser M, Schmeller S, Schetelig J. Hope for motherhood: pregnancy after allogeneic hematopoietic cell transplantation (a national multicenter study). Blood 2024; 144:1532-1542. [PMID: 39007722 DOI: 10.1182/blood.2024024342] [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: 03/12/2024] [Revised: 06/10/2024] [Accepted: 06/16/2024] [Indexed: 07/16/2024] Open
Abstract
ABSTRACT Improved long-term survival rates after allogeneic hematopoietic cell transplantation (alloHCT) make family planning for young adult cancer survivors an important topic. However, treatment-related infertility risk poses challenges. To assess pregnancy and birth rates in a contemporary cohort, we conducted a national multicenter study using data from the German Transplant Registry, focusing on adult women aged 18 to 40 years who underwent alloHCT between 2003 and 2018. Of 2654 women who underwent transplantation, 50 women experienced 74 pregnancies, occurring at a median of 4.7 years after transplant. Fifty-seven of these resulted in live births (77%). The annual first birth rate among HCT recipients was 0.45%, which is >6 times lower than in the general population. The probability of a live birth 10 years after HCT was 3.4%. Factors associated with an increased likelihood of pregnancy were younger age at alloHCT, nonmalignant transplant indications, no total body irradiation or a cumulative dose of <8 Gy, and nonmyeloablative/reduced-intensity conditioning. Notably, 72% of pregnancies occurred spontaneously, with assisted reproductive technologies used in the remaining cases. Preterm delivery and low birth weight were more common than in the general population. This study represents the largest data set reporting pregnancies in a cohort of adult female alloHCT recipients. Our findings underscore a meaningful chance of pregnancy in alloHCT recipients. Assisted reproductive technologies techniques are important and funding should be made available. However, the potential for spontaneous pregnancies should not be underestimated, and patients should be informed of the possibility of unexpected pregnancy despite reduced fertility. Further research is warranted to understand the impact of conditioning decisions on fertility preservation.
Collapse
Affiliation(s)
- Katja Sockel
- Department of Internal Medicine I, University Hospital Dresden, Technical University Dresden, Dresden, Germany
| | - Annika Neu
- Altona Children's Hospital, Hamburg, Germany
| | - Maren Goeckenjan
- Department for Gynaecology and Obstetrics, University Hospital Dresden, Technical University Dresden, Dresden, Germany
| | - Markus Ditschkowski
- Department of Hematology and Stem Cell Transplantation, University of Duisburg-Essen, University Hospital Essen, Essen, Germany
| | - Inken Hilgendorf
- Universitätsklinikum Jena, Klinik für Innere Medizin II, Abteilung für Hämatologie und Internistische Onkologie, Jena, Germany
| | - Nicolaus Kröger
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Francis A Ayuk
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Friedrich Stoelzel
- Division of Stem Cell Transplantation and Cellular Immunotherapy, Department of Internal Medicine II, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Jan Moritz Middeke
- Department of Internal Medicine I, University Hospital Dresden, Technical University Dresden, Dresden, Germany
| | - Matthias Eder
- Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Wolfgang Bethge
- Department of Medicine II, Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Tübingen, Germany
| | - Jürgen Finke
- Department of Hematology, Oncology, and Stem Cell Transplantation, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Hartmut Bertz
- Department of Hematology, Oncology, and Stem Cell Transplantation, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Guido Kobbe
- Department of Hematology, Oncology and Clinical Immunology, University Hospital Düsseldorf, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Martin Kaufmann
- 2nd Department of Internal Medicine, Oncology, and Hematology, Robert Bosch Hospital, Stuttgart, Germany
| | - Uwe Platzbecker
- Department of Hematology, Cellular Therapy and Hemostaseology, University Hospital Leipzig, Leipzig, Germany
| | - David Beverungen
- Department of Hematology, Cellular Therapy and Hemostaseology, University Hospital Leipzig, Leipzig, Germany
| | - Christoph Schmid
- Augsburg University Hospital and Medical Faculty and Bavarian Cancer Research Center, Augsburg, Germany
| | - Malte von Bonin
- Department of Internal Medicine I, University Hospital Dresden, Technical University Dresden, Dresden, Germany
| | - Katharina Egger-Heidrich
- Department of Internal Medicine I, University Hospital Dresden, Technical University Dresden, Dresden, Germany
| | - Lisa Heberling
- Department of Internal Medicine I, University Hospital Dresden, Technical University Dresden, Dresden, Germany
| | - Karolin Trautmann-Grill
- Department of Internal Medicine I, University Hospital Dresden, Technical University Dresden, Dresden, Germany
| | - Raphael Teipel
- Department of Internal Medicine I, University Hospital Dresden, Technical University Dresden, Dresden, Germany
| | - Gesine Bug
- Department of Medicine, Hematology, and Oncology, Goethe University Frankfurt, University Hospital, Frankfurt, Germany
| | - Johanna Tischer
- Department of Medicine III, Ludwig Maximilian University, University Hospital of Munich, Munich, Germany
| | - Alessia Fraccaroli
- Department of Medicine III, Ludwig Maximilian University, University Hospital of Munich, Munich, Germany
| | - Matthias Fante
- Department of Internal Medicine III, Hematology, and Oncology, University Hospital Regensburg, Regensburg, Germany
| | - Daniel Wolff
- Department of Internal Medicine III, Hematology, and Oncology, University Hospital Regensburg, Regensburg, Germany
| | - Thomas Luft
- Department of Internal Medicine V (Hematology/Oncology/Rheumatology), Heidelberg University Hospital, Heidelberg, Germany
| | - Julia Winkler
- Department of Internal Medicine 5, University Hospital Erlangen, Erlangen, Germany
| | - Kerstin Schäfer-Eckart
- Department of Internal Medicine V, Nuremberg Hospital North, Paracelsus Medical University, Nuremberg, Germany
| | - Christof Scheid
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Udo Holtick
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Stefan Klein
- Department of Hematology and Oncology, University Hospital Mannheim, Heidelberg University, Mannheim, Germany
| | - Igor Wolfgang Blau
- Department of BMT, Clinic of Hematology, Oncology, and Tumor Immunology, Charite-University Medicine Berlin, Berlin, Germany
| | - Andreas Burchert
- Department of Hematology, Oncology, and Immunology, Carreras Leukemia Center, Philips University Marburg and University Hospital Gießen and Marburg, Marburg, Germany
| | - Gerald Wulf
- Hematology and Medical Oncology, University Medicine Göttingen, Göttingen, Germany
| | - Justin Hasenkamp
- Hematology and Medical Oncology, University Medicine Göttingen, Göttingen, Germany
| | - Rainer Schwerdtfeger
- Center for Hematopoietic Cell Transplantation, Deutsche Klinik für Diagnostik Helios Klinik, Wiesbaden, Germany
| | - Stephan Kaun
- Hematology, Oncology und Infectiology, Clinic Bremen-Mitte, Bremen, Germany
| | - Christian Junghanss
- Department of Medicine, Clinic III, Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Rostock, Germany
| | - Friederike Wortmann
- Department of Hematology and Oncology, Medical Center, University of Schleswig-Holstein, Lübeck, Germany
| | - Susann Winter
- Department of Internal Medicine I, University Hospital Dresden, Technical University Dresden, Dresden, Germany
| | - Helga Neidlinger
- German Registry of Stem Cell Transplantation (DRST), Ulm, Germany
| | - Catrin Theuser
- Department of Internal Medicine I, University Hospital Dresden, Technical University Dresden, Dresden, Germany
| | | | - Martin Bornhaeuser
- Department of Internal Medicine I, University Hospital Dresden, Technical University Dresden, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, Dresden, Germany
- National Center for Tumor Diseases, Partner Site Dresden, Dresden, Germany
| | | | - Johannes Schetelig
- Department of Internal Medicine I, University Hospital Dresden, Technical University Dresden, Dresden, Germany
| |
Collapse
|
5
|
Alsuliman T, Rojas RMM, Moukalled N, Brissot E, Quarez-Blaise L, Marjanovic Z, Blaise D, Murphy D, Logue M, Savani BN, Mohty M. Sexual health and emotional wellbeing of patients with haematological malignancies: general review. Lancet Haematol 2024; 11:e770-e779. [PMID: 39312924 DOI: 10.1016/s2352-3026(24)00208-4] [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: 12/25/2023] [Revised: 06/08/2024] [Accepted: 06/24/2024] [Indexed: 09/25/2024]
Abstract
Sexual health is an important aspect of a person's life. Many patients and haematologists believe that intimacy and sexuality issues are substantial during cancer treatment. The haematological cancer disease, diagnosis, shock of the announcement, treatment, and follow-up appointments, can all have negative effects on the quality of life of patients, their partners, other family members, and friends. Addressing the intimate aspects of patients' lives not only enhances their wellbeing but also contributes to the quality of their survivorship. Progress has been made in the management of sexual life-related complications; however, novel strategies in coordination with a multidisciplinary team need to be implemented. New and comprehensive approaches must be developed on a multidisciplinary scale. In this Series paper, we discuss the factors affecting the sexual life of patients with haematological malignancies, different methods to assess sexual function, as well as management approaches of sexual wellbeing among patients with haematological cancers.
Collapse
Affiliation(s)
- Tamim Alsuliman
- Sorbonne University, Paris, France; Department of Hematology and Cellular Therapy, Saint-Antoine Hospital, Assistance Publique- Hôpitaux de Paris, Paris, France; INSERM, Unité Mixte de Recherche 938, Paris, France.
| | - Reyes María Martín Rojas
- Department of Hematology and Cellular Therapy, Saint-Antoine Hospital, Assistance Publique- Hôpitaux de Paris, Paris, France
| | - Nour Moukalled
- Division of Hematology and Oncology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Eolia Brissot
- Sorbonne University, Paris, France; Department of Hematology and Cellular Therapy, Saint-Antoine Hospital, Assistance Publique- Hôpitaux de Paris, Paris, France; INSERM, Unité Mixte de Recherche 938, Paris, France
| | | | - Zora Marjanovic
- Sorbonne University, Paris, France; Department of Hematology and Cellular Therapy, Saint-Antoine Hospital, Assistance Publique- Hôpitaux de Paris, Paris, France; INSERM, Unité Mixte de Recherche 938, Paris, France
| | - Didier Blaise
- Transplant and Cellular Immunotherapy Program, Department of Hematology, Institute Paoli-Calmettes, Marseille, France
| | - Danielle Murphy
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Melissa Logue
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Bipin N Savani
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Mohamad Mohty
- Sorbonne University, Paris, France; Department of Hematology and Cellular Therapy, Saint-Antoine Hospital, Assistance Publique- Hôpitaux de Paris, Paris, France; INSERM, Unité Mixte de Recherche 938, Paris, France
| |
Collapse
|
6
|
Alon I, Bussod I, Golan OC, Ravitsky V. Mapping ethical, legal, and social implications (ELSI) of fertility preservation. J Assist Reprod Genet 2024; 41:2495-2514. [PMID: 39141169 PMCID: PMC11405582 DOI: 10.1007/s10815-024-03210-w] [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: 05/23/2024] [Accepted: 07/17/2024] [Indexed: 08/15/2024] Open
Abstract
RESEARCH QUESTION The study examines the ethical, legal, and social implications of fertility preservation, highlighting its importance across oncofertility, elective egg freezing, and posthumous assisted reproduction, as well as its impact on transgender individuals undergoing gender-affirming surgeries. DESIGN A comprehensive analysis of 600 articles, focusing on a diverse range of disciplines, including bioethics, psychology, and sociology, to explore public and healthcare professionals' knowledge, patient experiences, and regulatory constraints. RESULTS The body of literature is growing, indicating increasing recognition of FP's significance. Key themes included the centrality of counseling and informed decision-making, especially in oncofertility and EEF, and ethical debates surrounding informed consent and the autonomy of involved individuals. The analysis underscored a western-centric bias in current research, emphasizing the need for more inclusive and culturally sensitive studies. CONCLUSIONS The study calls for a nuanced understanding of FP, advocating for policies that consider ethical, cultural, and social dimensions. It suggests the necessity for interdisciplinary research to address identified gaps, particularly in understanding non-Western perspectives and ensuring equitable access to FP services globally. Moreover, the review emphasizes the importance of integrating patient-centric approaches and ethical frameworks to guide FP practices and policies, ensuring they respect diverse values and meet individuals' needs.
Collapse
Affiliation(s)
- Ido Alon
- Department of Development Economics, Autonomous University of Madrid, Madrid, Spain.
- University of Montreal, Montreal, Canada.
| | | | - Orit Cherny Golan
- University of Montreal, Montreal, Canada
- Yezreel Valley College, Yezreel Valley, Israel
| | | |
Collapse
|
7
|
Lin J, Yang T, Li L, Sun X, Li H. Analysis of assisted reproductive outcomes for gynecologic cancer survivors: a retrospective study. Reprod Biol Endocrinol 2024; 22:97. [PMID: 39107798 PMCID: PMC11301938 DOI: 10.1186/s12958-024-01272-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 07/31/2024] [Indexed: 08/10/2024] Open
Abstract
OBJECTIVE To examine the reproductive outcomes of assisted reproductive technology (ART) in gynecologic cancer patients and to assess maternal and neonatal complications. METHODS Women diagnosed with gynecologic cancer who underwent their first in vitro fertilization/intracytoplasmic sperm injection (IVF/ICSI) treatment between 2013 and 2021 at Shanghai Ji Ai Genetics and IVF Institute were included in this study. Infertile women without any history of cancer were matched to the cancer group. The primary outcome was the cumulative live birth rate. Baseline and follow-up data were compared between groups using Student's t-tests for normally distributed variables and with Chi-square test for categorical variables. A propensity score-based patient-matching approach was adopted to ensure comparability between individuals with and without specific cancer type. RESULTS A total of 136 patients with a history of gynecologic cancer and 241 healthy infertile controls were included in this study. Endometrial cancer constituted 50.70% of the cases and cervical cancer constituted 34.60% of the cases. The cancer group exhibited significantly shorter duration of stimulation, lower levels of estradiol, lower number of retrieved oocytes, day-3 embryos, and blastocysts compared to the control group (P < 0.05). The cumulative live birth rate of the gynecologic cancer group was significantly lower than that of the control group (36.10% vs. 60.50%, P < 0.001). Maternal and neonatal complications did not significantly differ between the groups (P > 0.05). The endometrial cancer and cervical cancer groups showed significantly lower cumulative live birth rates than their matched controls (38.60% vs. 64.50%, P = 0.011 and 24.20% vs. 68.60%, P < 0.001, respectively). CONCLUSIONS These findings highlight the decreased occurrence of pregnancy and live birth in female gynecologic cancer patients undergoing ART, particularly in endometrial cancers and cervical cancers. These findings have important implications for counseling and managing gynecologic cancer patients undergoing ART.
Collapse
Affiliation(s)
- Jing Lin
- Center for Reproductive Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tianying Yang
- Shanghai Ji Ai Genetics and IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Lu Li
- Shanghai Ji Ai Genetics and IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Xiaoxi Sun
- Shanghai Ji Ai Genetics and IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China.
- Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China.
| | - He Li
- Shanghai Ji Ai Genetics and IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
8
|
Foster KL, Lee DJ, Witchel SF, Gordon CM. Ovarian Insufficiency and Fertility Preservation During and After Childhood Cancer Treatment. J Adolesc Young Adult Oncol 2024; 13:377-388. [PMID: 38265460 DOI: 10.1089/jayao.2023.0111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2024] Open
Abstract
Premature ovarian insufficiency (POI) is one of many potential long-term consequences of childhood cancer treatment in females. Causes of POI in this patient population can include chemotherapy, especially alkylating agents, and radiation therapy. Rarely, ovarian tumors lead to ovarian dysfunction. POI can manifest as delayed pubertal development, irregular menses or amenorrhea, and infertility. This diagnosis often negatively impacts emotional health due to the implications of impaired ovarian function after already enduring treatment for a primary malignancy. The emerging adult may be challenged by the impact on energy level, quality of life, and fertility potential. POI can also lead to low bone density and compromised skeletal strength. This review discusses the health consequences of POI in childhood cancer survivors (CCS). We also explore the role of fertility preservation for CCS, including ovarian tissue cryopreservation and other available options. Lastly, knowledge gaps are identified that will drive a future research agenda.
Collapse
Affiliation(s)
- Kayla L Foster
- Texas Children's Cancer and Hematology Center, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas, USA
| | - Danielle J Lee
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Selma F Witchel
- Division of Pediatric Endocrinology, Department of Pediatrics, UPMC Children's Hospital, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Catherine M Gordon
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
9
|
Silvestris E, D’Oronzo S, Petracca EA, D’Addario C, Cormio G, Loizzi V, Canosa S, Corrado G. Fertility Preservation in the Era of Immuno-Oncology: Lights and Shadows. J Pers Med 2024; 14:431. [PMID: 38673058 PMCID: PMC11050999 DOI: 10.3390/jpm14040431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 04/05/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
In recent years, immuno-oncology has revolutionized the cancer treatment field by harnessing the immune system's power to counteract cancer cells. While this innovative approach holds great promise for improving cancer outcomes, it also raises important considerations related to fertility and reproductive toxicity. In fact, most young females receiving gonadotoxic anti-cancer treatments undergo iatrogenic ovarian exhaustion, resulting in a permanent illness that precludes the vocation of motherhood as a natural female sexual identity. Although commonly used, oocyte cryopreservation for future in vitro fertilization and even ovarian cortex transplantation are considered unsafe procedures in cancer patients due to their oncogenic risks; whereas, ovarian stem cells might support neo-oogenesis, providing a novel stemness model of regenerative medicine for future fertility preservation programs in oncology. Recent scientific evidence has postulated that immune checkpoint inhibitors (ICIs) might in some way reduce fertility by inducing either primary or secondary hypogonadism, whose incidence and mechanisms are not yet known. Therefore, considering the lack of data, it is currently not possible to define the most suitable FP procedure for young patients who are candidates for ICIs. In this report, we will investigate the few available data concerning the molecular regulation of ICI therapy and their resulting gonadal toxicity, to hypothesize the most suitable fertility preservation strategy for patients receiving these drugs.
Collapse
Affiliation(s)
- Erica Silvestris
- Gynecologic Oncology Unit, IRCCS Istituto Tumori “Giovanni Paolo II” Bari, 70124 Bari, Italy; (E.A.P.); (G.C.); (V.L.)
| | - Stella D’Oronzo
- Department of Interdisciplinary Medicine (DIM), University of Bari “Aldo Moro”, 70121 Bari, Italy;
- Division of Medical Oncology, A.O.U. Consorziale Policlinico di Bari, 70124 Bari, Italy
| | - Easter Anna Petracca
- Gynecologic Oncology Unit, IRCCS Istituto Tumori “Giovanni Paolo II” Bari, 70124 Bari, Italy; (E.A.P.); (G.C.); (V.L.)
| | - Claudia D’Addario
- Department of Interdisciplinary Medicine (DIM), University of Bari “Aldo Moro”, 70121 Bari, Italy;
- Division of Medical Oncology, A.O.U. Consorziale Policlinico di Bari, 70124 Bari, Italy
| | - Gennaro Cormio
- Gynecologic Oncology Unit, IRCCS Istituto Tumori “Giovanni Paolo II” Bari, 70124 Bari, Italy; (E.A.P.); (G.C.); (V.L.)
- Department of Interdisciplinary Medicine (DIM), University of Bari “Aldo Moro”, 70121 Bari, Italy;
| | - Vera Loizzi
- Gynecologic Oncology Unit, IRCCS Istituto Tumori “Giovanni Paolo II” Bari, 70124 Bari, Italy; (E.A.P.); (G.C.); (V.L.)
- Department of Translational Biomedicine and Neuroscience (DiBraiN), University of Bari “Aldo Moro”, 70124 Bari, Italy
| | - Stefano Canosa
- IVIRMA, Global Research Alliance, LIVET, 10126 Turin, Italy;
| | - Giacomo Corrado
- Gynecologic Oncology Unit, Department of Woman, Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00136 Roma, Italy;
| |
Collapse
|
10
|
Affdal AO, Salama M, Ravitsky V. Ethical, legal, social, and policy issues of ovarian tissue cryopreservation in prepubertal girls: a critical interpretive review. J Assist Reprod Genet 2024; 41:999-1026. [PMID: 38430324 PMCID: PMC11052756 DOI: 10.1007/s10815-024-03059-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 02/07/2024] [Indexed: 03/03/2024] Open
Abstract
PURPOSE Despite the increasing number of childhood cancer survivors, significant advances in ovarian tissue cryopreservation (OTC) technique and medical societies' recommendations, fertility preservation (FP) and FP discussions are not always offered as a standard of care in the pediatric context. The aim of this literature review is to understand what ethical, legal, social, and policy issues may influence the provision of FP by OTC in prepubertal girls with cancer. METHODS A critical interpretive review of peer-reviewed papers published between 2000 and January 2023 was conducted, guided by the McDougall's version of the critical interpretive synthesis (Dixon-Woods), to capture recurring concepts, principles, and arguments regarding FP by OTC for prepubertal girls. RESULTS Of 931 potentially relevant papers, 162 were included in our analysis. Data were grouped into seven thematic categories: (1) risks of the procedure, (2) unique decision-making issues in pediatric oncofertility, (3) counseling, (4) cultural and cost issues, and (5) disposition of cryopreserved reproductive tissue. CONCLUSION This first literature review focusing on ethical, legal, social, and policy issues surrounding OTC in prepubertal girls highlights concerns in the oncofertility debate. Although OTC is no longer experimental as of December 2019, these issues could limit its availability and the child's future reproductive autonomy. This review concludes that specific actions must be provided to enable the offer of FP, such as supporting families' decision-making in this unique and complex context, and providing pediatric patients universal and full access to free or highly subsidized OTC.
Collapse
Affiliation(s)
| | | | - Vardit Ravitsky
- University of Montreal, Montreal, Canada
- The Hastings Center, Garrison, USA
- Harvard Medical School, Boston, USA
| |
Collapse
|
11
|
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
|
12
|
Rosario R, Stewart HL, Spears N, Telfer EE, Anderson RA. Anti-Mullerian hormone attenuates both cyclophosphamide-induced damage and PI3K signalling activation, while rapamycin attenuates only PI3K signalling activation, in human ovarian cortex in vitro. Hum Reprod 2024; 39:382-392. [PMID: 38070496 PMCID: PMC10833070 DOI: 10.1093/humrep/dead255] [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: 05/05/2023] [Revised: 11/13/2023] [Indexed: 02/02/2024] Open
Abstract
STUDY QUESTION What are the effects of cyclophosphamide exposure on the human ovary and can anti-Mullerian hormone (AMH) and rapamycin protect against these? SUMMARY ANSWER Exposure to cyclophosphamide compromises the health of primordial and transitional follicles in the human ovarian cortex and upregulates PI3K signalling, indicating both direct damage and increased follicular activation; AMH attenuates both of these chemotherapy-induced effects, while rapamycin attenuates only PI3K signalling upregulation. WHAT IS KNOWN ALREADY Studies primarily in rodents demonstrate that cyclophosphamide causes direct damage to primordial follicles or that the primordial follicle pool is depleted primarily through excessive initiation of follicle growth. This increased follicular activation is mediated via upregulated PI3K signalling and/or reduced local levels of AMH production due to lost growing follicles. Furthermore, while rodent data show promise regarding the potential benefits of inhibitors/protectants alongside chemotherapy treatment to preserve female fertility, there is no information about the potential for this in humans. STUDY DESIGN, SIZE, DURATION Fresh ovarian cortical biopsies were obtained from 17 healthy women aged 21-41 years (mean ± SD: 31.8 ± 4.9 years) at elective caesarean section. Biopsies were cut into small fragments and cultured for 24 h with either vehicle alone (DMSO), the active cyclophosphamide metabolite 4-hydroperoxycyclophosphamide (4-HC) alone, 4-HC + rapamycin or 4-HC+AMH. Two doses of 4-HC were investigated, 0.2 and 2 μM in separate experiments, using biopsies from seven women (aged 27-41) and six women (aged 21-34), respectively. Biopsies from four women (aged 28-38) were used to investigate the effect of rapamycin or AMH only. PARTICIPANTS/MATERIALS, SETTING, METHODS Histological analysis of ovarian tissue was undertaken for follicle staging and health assessment. Western blotting and immunostaining were used to assess activation of PI3K signalling by measuring phosphorylation of AKT and phosphorylated FOXO3A staining intensity, respectively. MAIN RESULTS AND THE ROLE OF CHANCE Exposure to either dose of 4-HC caused an increase in the proportion of unhealthy primordial (P < 0.0001, both doses) and transitional follicles (P < 0.01 for low dose and P < 0.01 for high dose) compared to vehicle. AMH significantly reduced follicle damage by approximately half in both of the investigated doses of 4-HC (P < 0.0001), while rapamycin had no protective effect on the health of the follicles. Culture with AMH or rapamycin alone had no effect on follicle health. Activation of PI3K signalling following 4-HC exposure was demonstrated by both Western blotting data showing that 4-HC increased in AKT phosphorylation and immunostaining showing increased phosphorylated FOXO3A staining of non-growing oocytes. Treatment with rapamycin reduced the activation of PI3K signalling in experiments with low doses of 4-HC while culture with AMH reduced PI3K activation (both AKT phosphorylation and phosphorylated FOXO3A staining intensity) across both doses investigated. LIMITATIONS, REASONS FOR CAUTION These in vitro studies may not replicate in vivo exposures. Furthermore, longer experiment durations are needed to determine whether the effects observed translate into irreparable deficits of ovarian follicles. WIDER IMPLICATIONS OF THE FINDINGS These data provide a solid foundation on which to explore the efficacy of AMH in protecting non-growing ovarian follicles from gonadotoxic chemotherapies. Future work will require consideration of the sustained effects of chemotherapy treatment and potential protectants to ensure these agents do not impair the developmental competence of oocytes or lead to the survival of oocytes with accumulated DNA damage, which could have adverse consequences for potential offspring. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by grants from TENOVUS Scotland, the Academy of Medical Sciences (to R.R.), the Medical Research Council (G1100357 to R.A.A., MR/N022556/1 to the MRC Centre for Reproductive Health), and Merck Serono UK (to R.A.A.). R.R., H.L.S., N.S., and E.E.T. declare no conflicts of interest. R.A.A. reports grants and personal fees from Roche Diagnostics and Ferring Pharmaceuticals, and personal fees from IBSA and Merck outside the submitted work. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Roseanne Rosario
- Biomedical Sciences, University of Edinburgh, Edinburgh, UK
- Centre for Reproductive Health, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Hazel L Stewart
- Centre for Reproductive Health, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Norah Spears
- Biomedical Sciences, University of Edinburgh, Edinburgh, UK
| | - Evelyn E Telfer
- Biomedical Sciences, University of Edinburgh, Edinburgh, UK
- Institute of Cell Biology, University of Edinburgh, Edinburgh, UK
| | - Richard A Anderson
- Centre for Reproductive Health, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
13
|
Liakath Ali F, Park HS, Beckman A, Eddy AC, Alkhrait S, Ghasroldasht MM, Al-Hendy A, Raheem O. Fertility Protection, A Novel Concept: Umbilical Cord Mesenchymal Stem Cell-Derived Exosomes Protect against Chemotherapy-Induced Testicular Cytotoxicity. Int J Mol Sci 2023; 25:60. [PMID: 38203232 PMCID: PMC10779299 DOI: 10.3390/ijms25010060] [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] [Revised: 12/15/2023] [Accepted: 12/15/2023] [Indexed: 01/12/2024] Open
Abstract
Currently, there is no viable option for fertility preservation in prepubertal boys. Experimentally, controlled vitrification of testicular tissue has been evaluated and found to cause potential structural damage to the spermatogonial stem cell (SSC) niche during cryopreservation. In this report, we leveraged the regenerative effect of human umbilical cord-derived Mesenchymal stem cell exosomes (h-UCMSC-Exo) to protect against testicular damage from the cytotoxic effects of polychemotherapy (CTX). A chemotherapy-induced testicular dysfunctional model was established by CTX treatment with cyclophosphamide and Busulfan in vitro (human Sertoli cells) and in prepubescent mice. We assessed the effects of the exosomes by analyzing cell proliferation assays, molecular analysis, immunohistochemistry, body weight change, serum hormone levels, and fertility rate. Our data indicates the protective effect of h-UCMSC-Exo by preserving the SSC niche and preventing testicular damage in mice. Interestingly, mice that received multiple injections of h-UCMSC-Exo showed significantly higher fertility rates and serum testosterone levels (p < 0.01). Our study demonstrates that h-UCMSC-Exo can potentially be a novel fertility protection approach in prepubertal boys triaged for chemotherapy treatment.
Collapse
Affiliation(s)
- Farzana Liakath Ali
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL 60637, USA; (F.L.A.)
| | - Hang-Soo Park
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL 60637, USA; (F.L.A.)
| | - Analea Beckman
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL 60637, USA; (F.L.A.)
| | - Adrian C. Eddy
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL 60637, USA; (F.L.A.)
| | - Samar Alkhrait
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL 60637, USA; (F.L.A.)
| | | | - Ayman Al-Hendy
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL 60637, USA; (F.L.A.)
| | - Omer Raheem
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL 60637, USA; (F.L.A.)
- Department of Surgery, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
14
|
Ruan X. Expert consensus on fertility preservation in hematopoietic stem cell transplantation in girls in China. Gynecol Endocrinol 2023; 39:2146671. [PMID: 36403607 DOI: 10.1080/09513590.2022.2146671] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Aims: Preconditioning before hematopoietic stem cell transplantation (HSCT) seriously damages the ovarian function and causes female infertility. This consensus focuses on the fertility preservation(FP) for girls needing HSCT, aim to make doctors in different disciplines aware of the importance, necessity and technique of ovarian protection.Materials and methods: Summarizing relevant literature and organizing multidisciplinary experts, including obstetrics and gynecology, reproductive medicine, oncology, pediatrics and hematology for full discussion.Results: Individuals exposed to HSCT in childhood are at higher risk of loss of fertility. Considering the high risk of premature ovarian insufficiency (POI) after conditioning and negative impact of POI on fertility, physical and mental health, it is absolutely necessary to protect fertility before HSCT conditioning. Ovarian tissue cryopreservation is the main fertility preservation option for these population.Conclusions: Fertility preservation before HSCT conditioning is crucial. Ovarian tissue cryopreservation is often the only option for these population.
Collapse
Affiliation(s)
- X Ruan
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University. Beijing Maternal and Child Health Care Hospital, Beijing, China
| |
Collapse
|
15
|
Huang Y, Zhang Q, Cao W, Zhang Q, Wang L, Lai D. TNF-α and IFN-γ prestimulation enhances the therapeutic efficacy of human amniotic epithelial stem cells in chemotherapy-induced ovarian dysfunction. Inflamm Regen 2023; 43:57. [PMID: 37993924 PMCID: PMC10664537 DOI: 10.1186/s41232-023-00309-y] [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] [Accepted: 11/11/2023] [Indexed: 11/24/2023] Open
Abstract
BACKGROUND Exposure to a harsh ovarian microenvironment induced by chemotherapeutic agents seriously affects the remodeling of ovarian function and follicular development, leading to premature ovarian failure or insufficiency (POF/POI). For decades, the effectiveness of stem cell therapies in POI animal models has been intensively studied; however, strategies to enhance the therapeutic effect of stem cells remain challenging. METHODS In this study, we first observed the pathological changes of the ovaries at different time points during chemotherapy, including the number of follicles, granulosa cell proliferation, oxidative stress damage, ovarian fibrosis, and inflammatory reaction. Moreover, we investigated whether activated hAECs stimulated by the proinflammatory cytokines tumor necrosis factor-α (TNF-α) and interferon-γ (IFN-γ) were more effective than native hAECs in repairing ovarian injury induced by chemotherapy. RESULTS The inhibitory effect of chemotherapy drugs on ovarian granulosa cells (GCs) in growing follicles mainly occurred on day 3 after chemotherapy in a mouse model. Then, continued ovarian injury, including oxidative damage and cell death cascades, resulted in the depletion of follicular reserves and inflammation-related ovarian fibrosis. Cytokine array demonstrated that activated hAECs secreted high levels of paracrine cytokines related to extracellular matrix (ECM) remodeling, angiogenesis, and immunomodulation. An in vivo study showed that the engraftment rate of activated hAECs in damaged ovaries was higher than that of native hAECs. Furthermore, activated hAECs in damaged ovaries had significantly upregulated expression of the antioxidant proteins thioredoxin1/2. In addition, activated hAECs had increased numbers of mature follicles and ameliorated the ovarian microenvironment by promoting angiogenesis and reducing ovarian fibrosis. CONCLUSIONS These results indicated that secondary ovarian damage induced by chemotherapy, including oxidative stress damage, chronic inflammatory response, and ovarian tissue fibrosis should be attended. Prestimulation with the proinflammatory factors TNF-α and IFN-γ could enhance the therapeutic efficacy of hAECs against chemotherapy-induced ovarian dysfunction, which may become a new feasible strategy to improve the therapeutic potential of hAECs in regenerative medicine.
Collapse
Affiliation(s)
- Yating Huang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qiuwan Zhang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
- Shanghai Key Laboratory of Embryo Original Diseases, 145, Guang-Yuan Road, Shanghai, 200030, China.
| | - Wenjiao Cao
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qinyu Zhang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lulu Wang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Dongmei Lai
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
- Shanghai Key Laboratory of Embryo Original Diseases, 145, Guang-Yuan Road, Shanghai, 200030, China.
| |
Collapse
|
16
|
Antunes MB, Cardeal SP, Magalhães M, Vale-Fernandes E, Barreiro M, Sá R, Sousa M. Preservation of fertility in female patients with hematologic diseases. Blood Rev 2023; 62:101115. [PMID: 37562987 DOI: 10.1016/j.blre.2023.101115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 06/23/2023] [Accepted: 07/10/2023] [Indexed: 08/12/2023]
Abstract
Recent developments of assisted reproduction techniques turned possible to avoid the infertility consequences of oncologic treatments, but fertility preservation (FP) has been somewhat neglected in women with hematologic diseases undergoing gonadotoxic treatments. For these specific cases, the current options for FP include the cryopreservation of embryos, mature oocytes and ovarian tissue, and oocyte in-vitro maturation. We intend to make patients and clinicians aware of this important and relevant issue, and provide hematologists, assisted reproduction physicians and patients, with updated tools to guide decisions for FP. The physicians of the units responsible for female FP should always be available to decide on the best-individualized FP option in strict collaboration with hematologists. With a wide range of options for FP tailored to each case, a greater level of training and information is needed among clinicians, so that patients proposed to gonadotoxic treatments can be previously advised for FP techniques in hematological conditions. ABBREVIATED ABSTRACT: Recent developments of assisted reproduction techniques turned possible to preserve the fertility of women with hematologic diseases undergoing gonadotoxic treatments. Current options for fertility preservation in women with hematologic diseases are presented. It is imperative to offer fertility preservation to all women before starting any gonadotoxic treatment and in some cases after treatment. Fertility preservation methods enable to later achieve the desired pregnancy.
Collapse
Affiliation(s)
- Marika Bini Antunes
- Department of Clinical Hematology, Centro Hospitalar Universitário do Porto, Largo do Professor Abel Salazar, 4099-001 Porto, Portugal; UMIB-Unit for Multidisciplinary Research in Biomedicine/ITR-Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal.
| | - Sara Pinto Cardeal
- Laboratory of Cell Biology, Department of Microscopy, ICBAS-School of Medicine and Biomedical Sciences, University of Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| | - Manuel Magalhães
- UMIB-Unit for Multidisciplinary Research in Biomedicine/ITR-Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal; Department of Oncology, Centro Hospitalar Universitário do Porto, Largo do Professor Abel Salazar, 4099-001 Porto, Portugal
| | - Emídio Vale-Fernandes
- UMIB-Unit for Multidisciplinary Research in Biomedicine/ITR-Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal; Centro de Procriação Medicamente Assistida, Centro Materno-Infantil do Norte, Centro Hospitalar Universitário do Porto, Largo da Maternidade, 4050-371, Porto, Portugal
| | - Márcia Barreiro
- UMIB-Unit for Multidisciplinary Research in Biomedicine/ITR-Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal; Centro de Procriação Medicamente Assistida, Centro Materno-Infantil do Norte, Centro Hospitalar Universitário do Porto, Largo da Maternidade, 4050-371, Porto, Portugal.
| | - Rosália Sá
- UMIB-Unit for Multidisciplinary Research in Biomedicine/ITR-Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal; Laboratory of Cell Biology, Department of Microscopy, ICBAS-School of Medicine and Biomedical Sciences, University of Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| | - Mário Sousa
- UMIB-Unit for Multidisciplinary Research in Biomedicine/ITR-Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal; Laboratory of Cell Biology, Department of Microscopy, ICBAS-School of Medicine and Biomedical Sciences, University of Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| |
Collapse
|
17
|
Mattioli R, Ilari A, Colotti B, Mosca L, Fazi F, Colotti G. Doxorubicin and other anthracyclines in cancers: Activity, chemoresistance and its overcoming. Mol Aspects Med 2023; 93:101205. [PMID: 37515939 DOI: 10.1016/j.mam.2023.101205] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 07/15/2023] [Accepted: 07/17/2023] [Indexed: 07/31/2023]
Abstract
Anthracyclines have been important and effective treatments against a number of cancers since their discovery. However, their use in therapy has been complicated by severe side effects and toxicity that occur during or after treatment, including cardiotoxicity. The mode of action of anthracyclines is complex, with several mechanisms proposed. It is possible that their high toxicity is due to the large set of processes involved in anthracycline action. The development of resistance is a major barrier to successful treatment when using anthracyclines. This resistance is based on a series of mechanisms that have been studied and addressed in recent years. This work provides an overview of the anthracyclines used in cancer therapy. It discusses their mechanisms of activity, toxicity, and chemoresistance, as well as the approaches used to improve their activity, decrease their toxicity, and overcome resistance.
Collapse
Affiliation(s)
- Roberto Mattioli
- Dept. Biochemical Sciences A. Rossi Fanelli, Sapienza University of Rome, Rome, Italy
| | - Andrea Ilari
- Institute of Molecular Biology and Pathology, Italian National Research Council IBPM-CNR, Rome, Italy
| | - Beatrice Colotti
- Dept. Biochemical Sciences A. Rossi Fanelli, Sapienza University of Rome, Rome, Italy
| | - Luciana Mosca
- Dept. Biochemical Sciences A. Rossi Fanelli, Sapienza University of Rome, Rome, Italy
| | - Francesco Fazi
- Department of Anatomical, Histological, Forensic & Orthopaedic Sciences, Section of Histology and Medical Embryology, Sapienza University of Rome, Rome, Italy
| | - Gianni Colotti
- Institute of Molecular Biology and Pathology, Italian National Research Council IBPM-CNR, Rome, Italy.
| |
Collapse
|
18
|
Mincheva M, Fraire-Zamora JJ, Liperis G, Ammar OF, Duffin K, Kanbar M, Mitchell RT, Moura-Ramos M, Massarotti C. Going back to the start: do cancer and haematological disorders affect germ cells in prepubertal boys? Hum Reprod 2023; 38:1856-1860. [PMID: 37328443 DOI: 10.1093/humrep/dead128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/18/2023] Open
Affiliation(s)
| | | | - George Liperis
- Westmead Fertility Centre, Institute of Reproductive Medicine, University of Sydney, Westmead, NSW, Australia
| | - Omar F Ammar
- Biomaterials Cluster, Bernal Institute, University of Limerick, Limerick, Ireland
- School of Engineering, Faculty of Science and Engineering, University of Limerick, Limerick, Ireland
| | | | - Marc Kanbar
- Andrology Lab, Pôle de recherche en Physiopathologie de la Reproduction (REPR), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
- Department of Gynecology-Andrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Rod T Mitchell
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Mariana Moura-Ramos
- Clinical Psychology Unit, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- Center for Research in Neuropsychology and Cognitive Behavioral Intervention, University of Coimbra, Coimbra, Portugal
| | - Claudia Massarotti
- Academic Unit of Obstetrics and Gynecology, DINOGMI Department, University of Genoa, Genoa, Italy
- Physiopathology of Human Reproduction Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| |
Collapse
|
19
|
Lopez J, Hohensee G, Liang J, Sela M, Johnson J, Kallen AN. The Aging Ovary and the Tales Learned Since Fetal Development. Sex Dev 2023; 17:156-168. [PMID: 37598664 PMCID: PMC10841896 DOI: 10.1159/000532072] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 07/13/2023] [Indexed: 08/22/2023] Open
Abstract
BACKGROUND While the term "aging" implies a process typically associated with later life, the consequences of ovarian aging are evident by the time a woman reaches her forties, and sometimes earlier. This is due to a gradual decline in the quantity and quality of oocytes which occurs over a woman's reproductive lifespan. Indeed, the reproductive potential of the ovary is established even before birth, as the proper formation and assembly of the ovarian germ cell population during fetal life determines the lifetime endowment of oocytes and follicles. In the ovary, sophisticated molecular processes have been identified that regulate the timing of ovarian aging and these are critical to ensuring follicular maintenance. SUMMARY The mechanisms thought to contribute to overall aging have been summarized under the term the "hallmarks of aging" and include such processes as DNA damage, mitochondrial dysfunction, telomere attrition, genomic instability, and stem cell exhaustion, among others. Similarly, in the ovary, molecular processes have been identified that regulate the timing of ovarian aging and these are critical to ensuring follicular maintenance. In this review, we outline critical processes involved in ovarian aging, highlight major achievements for treatment of ovarian aging, and discuss ongoing questions and areas of debate. KEY MESSAGES Ovarian aging is recognized as what may be a complex process in which age, genetics, environment, and many other factors contribute to the size and depletion of the follicle pool. The putative hallmarks of reproductive aging outlined herein include a diversity of plausible processes contributing to the depletion of the ovarian reserve. More research is needed to clarify if and to what extent these putative regulators do in fact govern follicle and oocyte behavior, and how these signals might be integrated in order to control the overall pattern of ovarian aging.
Collapse
Affiliation(s)
- Jesus Lopez
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Gabe Hohensee
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Jing Liang
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Meirav Sela
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Joshua Johnson
- Department of Obstetrics and Gynecology, University of Colorado Denver, Aurora, CO, USA
| | - Amanda N. Kallen
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
20
|
Delgouffe E, Braye A, Vloeberghs V, Mateizel I, Ernst C, Ferster A, Devalck C, Tournaye H, Gies I, Goossens E. Spermatogenesis after gonadotoxic childhood treatment: follow-up of 12 patients. Hum Reprod Open 2023; 2023:hoad029. [PMID: 37547664 PMCID: PMC10403430 DOI: 10.1093/hropen/hoad029] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 07/13/2023] [Indexed: 08/08/2023] Open
Abstract
STUDY QUESTION What is the long-term impact of presumed gonadotoxic treatment during childhood on the patient's testicular function at adulthood? SUMMARY ANSWER Although most patients showed low testicular volumes and some degree of reproductive hormone disruption 12.3 (2.3-21.0) years after gonadotoxic childhood therapy, active spermatogenesis was demonstrated in the semen sample of 8 out of the 12 patients. WHAT IS KNOWN ALREADY In recent decades, experimental testicular tissue banking programmes have been set up to safeguard the future fertility of young boys requiring chemo- and/or radiotherapy with significant gonadotoxicity. Although the risk of azoospermia following such therapies is estimated to be high, only limited long-term data are available on the reproductive potential at adulthood. STUDY DESIGN SIZE DURATION This single-centre prospective cohort study was conducted between September 2020 and February 2023 and involved 12 adult patients. PARTICIPANTS/MATERIALS SETTING METHODS This study was carried out in a tertiary care centre and included 12 young adults (18.1-28.3 years old) who had been offered testicular tissue banking prior to gonadotoxic treatment during childhood. All patients had a consultation and physical examination with a fertility specialist, a scrotal ultrasound to measure the testicular volumes and evaluate the testicular parenchyma, a blood test for assessment of reproductive hormones, and a semen analysis. MAIN RESULTS AND THE ROLE OF CHANCE Testicular tissue was banked prior to the gonadotoxic treatment for 10 out of the 12 included patients. Testicular volumes were low for 9 patients, and 10 patients showed some degree of reproductive hormone disruption. Remarkably, ongoing spermatogenesis was demonstrated in 8 patients at a median 12.3 (range 2.3-21.0) years post-treatment. LIMITATIONS REASONS FOR CAUTION This study had a limited sample size, making additional research with a larger study population necessary to verify these preliminary findings. WIDER IMPLICATIONS OF THE FINDINGS These findings highlight the need for multicentric research with a larger study population to establish universal inclusion criteria for immature testicular tissue banking. STUDY FUNDING/COMPETING INTERESTS This study was conducted with financial support from the Research Programme of the Research Foundation-Flanders (G010918N), Kom Op Tegen Kanker, and Scientific Fund Willy Gepts (WFWG19-03). The authors declare no competing interests. TRIAL REGISTRATION NUMBER NCT04202094; https://clinicaltrials.gov/ct2/show/NCT04202094?id=NCT04202094&draw=2&rank=1 This study was registered on 6 December 2019, and the first patient was enrolled on 8 September 2020.
Collapse
Affiliation(s)
- E Delgouffe
- Correspondence address. Department of Reproduction, Genetics and Regenerative Medicine (RGRG), Biology of the Testis (BITE), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussels, Belgium. Tel: +32-(0)2-477-46-44; E-mail: https://orcid.org/0000-0001-5611-2173
| | - A Braye
- Department of Reproduction, Genetics and Regenerative Medicine (RGRG), Biology of the Testis (BITE), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - V Vloeberghs
- Brussels IVF, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - I Mateizel
- Brussels IVF, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - C Ernst
- Division of Paediatric Radiology, Department of Radiology, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - A Ferster
- Department of Hemato-Oncology, Hôpital Universitaire des Enfants Reine Fabiola (HUDERF), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - C Devalck
- Department of Hemato-Oncology, Hôpital Universitaire des Enfants Reine Fabiola (HUDERF), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - H Tournaye
- Brussels IVF, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
- Department of Obstetrics, Gynaecology, Perinatology and Reproduction, Institute of Professional Education, Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| | - I Gies
- Division of Paediatric Endocrinology, Department of Paediatrics, Universitair Ziekenhuis Brussel (UZ Brussel), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - E Goossens
- Department of Reproduction, Genetics and Regenerative Medicine (RGRG), Biology of the Testis (BITE), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| |
Collapse
|
21
|
Stukaite-Ruibiene E, van der Perk MEM, Vaitkeviciene GE, Bos AME, Bumbuliene Z, van den Heuvel-Eibrink MM, Rascon J. Evaluation of oncofertility care in childhood cancer patients: the EU-Horizon 2020 twinning project TREL initiative. Front Pediatr 2023; 11:1212711. [PMID: 37565239 PMCID: PMC10411952 DOI: 10.3389/fped.2023.1212711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 07/14/2023] [Indexed: 08/12/2023] Open
Abstract
Background The 5-year survival rate of childhood cancer exceeds 80%, however, many survivors develop late effects including infertility. The aim of this study was to evaluate the current status of oncofertility care at Vilnius University Hospital Santaros Klinikos (VULSK) within the framework of the EU-Horizon 2020 TREL project. Methods All parents or patients aged 12-17.9 years treated from July 1, 2021 until July 1, 2022 were invited to complete an oncofertility-care-evaluation questionnaire. After completing the questionnaire, patients were triaged to low-risk (LR) or high-risk (HR) of gonadal damage using a risk stratification tool (triage). Data was assessed using descriptive statistics. Results Questionnaires were completed by 48 parents and 13 children triaged as 36 (59%) LR and 25 (41%) HR patients. Most HR respondents (21/25, 84%) were not counseled by a fertility specialist. Six boys (4 HR, 2 LR) were counseled, none of the girls was counseled. Three HR boys underwent sperm cryopreservation. Only 17 (27.9%, 9 HR, 8 LR) respondents correctly estimated their risk. All counseled boys (n = 6) agreed the risk for fertility impairment had been mentioned as compared to 49.1% (n = 27) of uncounseled. All counseled respondents agreed they knew enough about fertility (vs. 42%). Conclusions Respondents counseled by a fertility specialist were provided more information on fertility than uncounseled. HR patients were not sufficiently counseled by a fertility specialist. Based on the current experience oncofertility care at VULSK will be improved.
Collapse
Affiliation(s)
| | | | - Goda Elizabeta Vaitkeviciene
- Faculty of Medicine, Vilnius University, Vilnius, Lithuania
- Center for Pediatric Oncology and Hematology, Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
| | | | - Zana Bumbuliene
- Faculty of Medicine, Vilnius University, Vilnius, Lithuania
- Center of Obstetrics and Gynecology, Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
| | | | - Jelena Rascon
- Faculty of Medicine, Vilnius University, Vilnius, Lithuania
- Center for Pediatric Oncology and Hematology, Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
| |
Collapse
|
22
|
Slonim M, Peate M, Merigan K, Lantsberg D, Anderson RA, Stern K, Gook D, Jayasinghe Y. Ovarian stimulation and oocyte cryopreservation in females and transgender males aged 18 years or less: a systematic review. Front Endocrinol (Lausanne) 2023; 14:1146476. [PMID: 37404308 PMCID: PMC10315913 DOI: 10.3389/fendo.2023.1146476] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 05/15/2023] [Indexed: 07/06/2023] Open
Abstract
Background Fertility preservation is an important healthcare focus in the paediatric and adolescent population when gonadotoxic treatments are required. Ovarian stimulation (OS) resulting in oocyte cryopreservation is a well-established fertility preservation option in the adult population. It's utility, however, is little known in young patients. The purpose of this review was to synthesise the available literature on OS in patients ≤18 years old, to identify gaps in current research and provide suggestions for future research directions. Methods Using PRISMA guidelines, a systematic review of the literature was performed for all relevant full-text articles published in English in Medline, Embase, the Cochrane Library and Google Scholar databases. The search strategy used a combination of subject headings and generic terms related to the study topic and population. Two reviewers independently screened studies for eligibility, extracted data and assessed the risk of bias. Characteristics of the studies, objectives and key findings were extracted and summarised in a narrative synthesis. Results Database search and manual review identified 922 studies, 899 were eliminated based on defined exclusion criteria. Twenty-three studies were included and comprised 468 participants aged ≤18 years who underwent OS (median 15.2, range 7-18 years old). Only three patients were premenarchal, and four patients were on treatment to suppress puberty. Patients had OS for a broad range of indications including oncology treatment, transgender care and Turner syndrome. A total of 488 cycles of OS were completed, with all but 18 of these cycles (96.3%) successfully resulting in cryopreserved mature oocytes (median 10 oocytes, range 0-35). Fifty-three cycles (9.8%) were cancelled. Complications were rare (<1%). One pregnancy was reported from a female who had OS aged 17 years old. Conclusion This systematic review demonstrates that OS and oocyte cryopreservation is achievable in young females however there are only a few cases in the literature describing OS in premenarcheal children or those who have suppressed puberty. There is little proof that OS can lead to pregnancy in adolescents, and no proof that this can be achieved in premenarchal girls. Therefore it should be regarded as an innovative procedure for adolescents and experimental for premenarcheal girls. Systematic review registration https://www.crd.york.ac.uk/prospero/display_record.php?RecordID=265705, identifier CRD42021265705.
Collapse
Affiliation(s)
- Marnie Slonim
- Oncofertility Program and Department of Gynaecology, Royal Children’s Hospital, Melbourne, VIC, Australia
| | - Michelle Peate
- Department of Obstetrics and Gynaecology, Royal Women's Hospital, The University of Melbourne, Melbourne, VIC, Australia
| | - Kira Merigan
- Department of Obstetrics and Gynaecology, Royal Women’s Hospital, Melbourne, VIC, Australia
| | | | - Richard A. Anderson
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, United Kingdom
| | - Kate Stern
- Department of Obstetrics and Gynaecology, Royal Women’s Hospital, Melbourne, VIC, Australia
- Melbourne IVF, Melbourne, VIC, Australia
| | - Debra Gook
- Department of Obstetrics and Gynaecology, Royal Women's Hospital, The University of Melbourne, Melbourne, VIC, Australia
- Melbourne IVF, Melbourne, VIC, Australia
| | - Yasmin Jayasinghe
- Oncofertility Program and Department of Gynaecology, Royal Children’s Hospital, Melbourne, VIC, Australia
- Department of Obstetrics and Gynaecology, Royal Women's Hospital, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
23
|
Calanni-Pileri M, Michaelis M, Langhammer M, Tognetti PR, Weitzel JM. The Imitation of the Ovarian Fatty Acid Profile of Superfertile Dummerstorf Mouse Lines during IVM of Control Line Oocytes Could Influence Their Maturation Rates. Biomedicines 2023; 11:biomedicines11051439. [PMID: 37239110 DOI: 10.3390/biomedicines11051439] [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: 03/24/2023] [Revised: 05/08/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
Declining human fertility worldwide is an attractive research target for the search for "high fertility" genes and pathways to counteract this problem. To study these genes and pathways for high fertility, the superfertile Dummerstorf mouse lines FL1 and FL2 are two unique model organisms representing an improved fertility phenotype. A direct reason for this remarkable characteristic of increased litter size, which reaches >20 pups/litter in both FLs, is the raised ovulation rate by approximately 100%, representing an impressive record in this field. Dummerstorf high-fertility lines incarnate extraordinary and singular models of high-fertility for other species, mostly farm animals, with the aim of improving production and reducing costs. Our main goal is to describe the genetic and molecular pathways to reach their phenotypical excellence, and to reproduce them using the control population. The large litter size and ovulation rate in Dummerstorf lines are mostly due to an increase in the quality of their oocytes, which receive a different intake of fat and are composed of different types and concentrations of fatty acids. As the follicular microenvironment plays a fundamental role during the oocytes development, in the present manuscript, we tried to improve the in vitro maturation technique by mimicking the fatty acid profile of FLs oocytes during the IVM of control oocytes. Currently, the optimization of the IVM system is fundamental mostly for prepubertal girls and oncological patients whose main source of gametes to restore fertility may be their maturation in vitro. Our data suggest that the specific fatty acid composition of FLs COCs can contribute to their high-fertility phenotype. Indeed, COCs from the control line matured in IVM-medium supplemented with C14:0 (high in FL2 COCs) or with C20:0, C21:0, C22:0, and C23:0 (high in FL1 COCs), but also control oocytes without cumulus, whose concentration in long-chain FAs are "naturally" higher, showing a slightly higher maturation rate. These findings represent an important starting point for the optimization of the IVM system using FA supplementation.
Collapse
Affiliation(s)
- Michela Calanni-Pileri
- Institute of Reproductive Biology, Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
| | - Marten Michaelis
- Institute of Reproductive Biology, Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
| | - Martina Langhammer
- Service Group Lab Animal Facility, Institute of Genetics and Biometry, Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
| | - Paolo Rosellini Tognetti
- Institute of Reproductive Biology, Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
- Service Group Lab Animal Facility, Institute of Genetics and Biometry, Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
| | - Joachim M Weitzel
- Institute of Reproductive Biology, Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
| |
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: 2.5] [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
|
Clasen NHZ, van der Perk MEM, Neggers SJCMM, Bos AME, van den Heuvel-Eibrink MM. Experiences of Female Childhood Cancer Patients and Survivors Regarding Information and Counselling on Gonadotoxicity Risk and Fertility Preservation at Diagnosis: A Systematic Review. Cancers (Basel) 2023; 15:cancers15071946. [PMID: 37046607 PMCID: PMC10093478 DOI: 10.3390/cancers15071946] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/12/2023] [Accepted: 03/21/2023] [Indexed: 03/29/2023] Open
Abstract
Background: Childhood cancer patients and their families are increasingly offered oncofertility care including information regarding their risk of gonadal damage by paediatric oncologists, fertility counselling by fertility specialists and fertility preservation options. However, experiences regarding oncofertility care are underreported. We aimed to summarize the available evidence of experiences of female childhood cancer patients and survivors regarding oncofertility care. Methods: Manuscripts were systematically identified using the PubMed and Embase database. From, respectively, 1256 and 3857 manuscripts, 7 articles were included and assessed, including risk of bias assessment. Outcome measures included data describing experiences of female childhood cancer patients and survivors, regarding fertility information, counselling and/or preservation. Results: Female patients and survivors are variably satisfied with fertility information, report challenges in communication with healthcare professionals and prefer to receive general information at diagnosis and detailed fertility information later. Regrets after fertility counselling are underreported, but are associated with refusing fertility preservation. Lastly, regardless of counselling, female patients and survivors report fertility concerns about their future children’s health and effect on relationships. Conclusion: Currently, the satisfaction with oncofertility care varies and female patients or survivors report regrets and concerns regardless of receiving fertility information or counselling. These results may help to improve the content of fertility information, communication skills of healthcare professionals and timing of counselling.
Collapse
Affiliation(s)
- Nikita H Z Clasen
- Princess Máxima Center for Pediatric Oncology, 3584CS Utrecht, The Netherlands
- Faculty of Medicine, Vrije Universiteit Amsterdam, 1081CX Amsterdam, The Netherlands
| | | | - Sebastian J C M M Neggers
- Department of Internal Medicine, Section Endocrinology, Erasmus Medical Center, 3015GD Rotterdam, The Netherlands
| | - Annelies M E Bos
- Princess Máxima Center for Pediatric Oncology, 3584CS Utrecht, The Netherlands
| | - Marry M van den Heuvel-Eibrink
- Princess Máxima Center for Pediatric Oncology, 3584CS Utrecht, The Netherlands
- Division of Child Health, Wilhelmina Children's Hospital, University Medical Center Utrecht, 3584EA Utrecht, The Netherlands
| |
Collapse
|
26
|
Horvath-Pereira BDO, Almeida GHDR, da Silva Júnior LN, do Nascimento PG, Horvath Pereira BDO, Fireman JVBT, Pereira MLDRF, Carreira ACO, Miglino MA. Biomaterials for Testicular Bioengineering: How far have we come and where do we have to go? Front Endocrinol (Lausanne) 2023; 14:1085872. [PMID: 37008920 PMCID: PMC10060902 DOI: 10.3389/fendo.2023.1085872] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 02/24/2023] [Indexed: 03/18/2023] Open
Abstract
Traditional therapeutic interventions aim to restore male fertile potential or preserve sperm viability in severe cases, such as semen cryopreservation, testicular tissue, germ cell transplantation and testicular graft. However, these techniques demonstrate several methodological, clinical, and biological limitations, that impact in their results. In this scenario, reproductive medicine has sought biotechnological alternatives applied for infertility treatment, or to improve gamete preservation and thus increase reproductive rates in vitro and in vivo. One of the main approaches employed is the biomimetic testicular tissue reconstruction, which uses tissue-engineering principles and methodologies. This strategy pursues to mimic the testicular microenvironment, simulating physiological conditions. Such approach allows male gametes maintenance in culture or produce viable grafts that can be transplanted and restore reproductive functions. In this context, the application of several biomaterials have been proposed to be used in artificial biological systems. From synthetic polymers to decellularized matrixes, each biomaterial has advantages and disadvantages regarding its application in cell culture and tissue reconstruction. Therefore, the present review aims to list the progress that has been made and the continued challenges facing testicular regenerative medicine and the preservation of male reproductive capacity, based on the development of tissue bioengineering approaches for testicular tissue microenvironment reconstruction.
Collapse
Affiliation(s)
| | | | | | - Pedro Gabriel do Nascimento
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | | | | | | | - Ana Claudia Oliveira Carreira
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
- Centre for Natural and Human Sciences, Federal University of ABC, São Paulo, Brazil
| | - Maria Angelica Miglino
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
27
|
Reduced-Intensity Conditioning Mitigates Risk for Primary Ovarian Insufficiency but Does Not Decrease Risk for Infertility in Pediatric and Young Adult Survivors of Hematopoietic Stem Cell Transplantation. Transplant Cell Ther 2023; 29:130.e1-130.e8. [PMID: 36323400 DOI: 10.1016/j.jtct.2022.10.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 09/17/2022] [Accepted: 10/20/2022] [Indexed: 11/21/2022]
Abstract
Hematopoietic stem cell transplantation (HSCT) is a curative therapy for many pediatric malignant and nonmalignant conditions. Gonadal insufficiency or infertility is present in almost all HSCT survivors who received a myeloablative conditioning (MAC) regimen. Reduced-intensity conditioning (RIC) regimens are being increasingly used in medically fragile patients or in patients with nonmalignant diagnoses to limit the toxicities associated with HSCT; however, the short-term and long-term gonadal toxicity of RIC regimens in pediatric and young adult survivors remains unknown. In this study, we compared the prevalence of gonadal insufficiency and infertility among pubertal and postpubertal pediatric and young adult survivors of HSCT who received a RIC regimen versus those who received a MAC regimen. Twenty-three females (RIC, n = 8; MAC, n = 15) and 35 males (RIC, n = 19; MAC, n = 16) were included in this single-center, retrospective cross-sectional study. Eligible patients were those with available laboratory results who were ≥1 year post-HSCT, age <40 years, and pubertal or postpubertal as assessed by an endocrinologist. Follicle-stimulating hormone (FSH), luteinizing hormone (LH), estradiol, and anti-Müllerian hormone (AMH) levels were measured in females, and FSH, LH, total testosterone, and inhibin B (InhB) levels were measured in males. Twenty-one males (RIC, n = 11; MAC, n = 10) underwent semen analysis through a separate consent. Parametric and nonparametric analyses were undertaken to compare the RIC and MAC groups. Female patients who received RIC were less likely than those who received MAC to develop primary ovarian insufficiency, as demonstrated by elevated FSH (P = .02) and low estradiol (P = .01) or elevated LH (P = .09). Most females in the RIC (75%) and MAC (93%) groups had low AMH levels, indicating low or absent ovarian reserve, with no significant difference between the groups (P = .53). In males, there were no significant differences between the 2 groups in the prevalence of abnormal FSH, LH, testosterone, or InhB levels. Ten of 11 RIC males (91%) and 10 of 10 MAC males (100%) had azoospermia or oligospermia, at a median time to semen analysis from HSCT of 3.7 years (range, 1.3 to 12.2 years). RIC may pose less risk than MAC for primary ovarian insufficiency among female survivors of HSCT; however, both female and male recipients of either RIC or MAC regimens are at high risk for infertility. In the largest reported series of semen analyses of pediatric and young adult male recipients of RIC, azoospermia or oligospermia was found in nearly all (91%) RIC survivors. All patients undergoing HSCT should receive counseling about the high risk of gonadal toxicity, and efforts should be made to preserve fertility in patients undergoing either RIC or MAC.
Collapse
|
28
|
Effects and Mechanisms Activated by Treatment with Cationic, Anionic and Zwitterionic Liposomes on an In Vitro Model of Porcine Pre-Pubertal Sertoli Cells. Int J Mol Sci 2023; 24:ijms24021201. [PMID: 36674712 PMCID: PMC9865246 DOI: 10.3390/ijms24021201] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/28/2022] [Accepted: 01/03/2023] [Indexed: 01/11/2023] Open
Abstract
Liposomes have been successfully used as drug-delivery vehicles, but there are no clinical studies on improved fertility and the few reported experimental studies have been performed in animal models far from humans. The aim of this paper was to study the effects of treatment with cationic, anionic and zwitterionic liposomes on our superior mammalian model of porcine prepubertal Sertoli cells (SCs) to find a carrier of in vitro test drugs for SCs. Porcine pre-pubertal SCs cultures were incubated with different liposomes. Viability, apoptosis/necrosis status (Annexin-V/Propidium iodide assay), immunolocalisation of β-actin, vimentin, the phosphorylated form of AMP-activated protein Kinase (AMPK)α and cell ultrastructure (Transmission Electron Microscopy, TEM) were analysed. Zwitterionic liposomes did not determine changes in the cell cytoplasm. The incubation with anionic and cationic liposomes modified the distribution of actin and vimentin filaments and increased the levels of the phosphorylated form of AMPKα. The Annexin/Propidium Iodide assay suggested an increase in apoptosis. TEM analysis highlighted a cytoplasmic vacuolisation. In conclusion, these preliminary data indicated that zwitterionic liposomes were the best carrier to use in an in vitro study of SCs to understand the effects of molecules or drugs that could have a clinical application in the treatment of certain forms of male infertility.
Collapse
|
29
|
Zhao J, Tang M, Tang H, Wang M, Guan H, Tang L, Zhang H. Sphingosine 1-phosphate alleviates radiation-induced ferroptosis in ovarian granulosa cells by upregulating glutathione peroxidase 4. Reprod Toxicol 2023; 115:49-55. [PMID: 36503164 DOI: 10.1016/j.reprotox.2022.12.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 12/01/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022]
Abstract
Ferroptosis is a form of cell death caused by the accumulation of lipid peroxidation products due to abnormal iron metabolism. However, it remains unknown whether ferroptosis participates in the process of radiation-induced ovarian injury. Sphingosine-1-phosphate (S1P) is an important bioactive sphingolipid that has a protective effect on ovarian injury. The present study aims to determine whether X-ray radiation induces ferroptosis in the ovarian granulosa KGN cell line, and explore the potential effect of S1P and its mechanism in radiation-induced ferroptosis. The results indicated that irradiation reduced the viability of KGN cells, altered the mitochondrial morphology, induced the intracellular accumulation of iron ions, increased oxidative stress, and induced lipid peroxidation. Furthermore, the radiation exposure triggered the ferroptosis in KGN cells. S1P can alleviate radiation-induced ferroptosis. Furthermore, the protective effect of S1P was reversed after the application of siRNA to interfere with the glutathione peroxidase 4 expression. Ferroptosis might be pervasive in radiation-induced ovarian injury, and S1P may serve as a potential therapeutic approach to protect against the toxic effect of radiation in female gonads by inhibiting ferroptosis.
Collapse
Affiliation(s)
- Jiahui Zhao
- Department of Reproductive Medicine, Lianyungang Maternal and Child Health Hospital
| | - Mingyan Tang
- Department of Reproductive Medicine, the Second Affiliated Hospital of Soochow University
| | - Huaiyun Tang
- Department of Reproductive Medicine, Lianyungang Maternal and Child Health Hospital
| | - Mei Wang
- Department of Reproductive Medicine, Lianyungang Maternal and Child Health Hospital
| | - Huijuan Guan
- Department of Reproductive Medicine, Lianyungang Maternal and Child Health Hospital
| | - Lisha Tang
- Department of Reproductive Medicine, Lianyungang Maternal and Child Health Hospital
| | - Hong Zhang
- Department of Reproductive Medicine, the Second Affiliated Hospital of Soochow University.
| |
Collapse
|
30
|
Ovarian Reserve Disorders, Can We Prevent Them? A Review. Int J Mol Sci 2022; 23:ijms232315426. [PMID: 36499748 PMCID: PMC9737352 DOI: 10.3390/ijms232315426] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022] Open
Abstract
The ovarian reserve is finite and begins declining from its peak at mid-gestation until only residual follicles remain as women approach menopause. Reduced ovarian reserve, or its extreme form, premature ovarian insufficiency, stems from multiple factors, including developmental, genetic, environmental exposures, autoimmune disease, or medical/surgical treatment. In many cases, the cause remains unknown and resulting infertility is not ultimately addressed by assisted reproductive technologies. Deciphering the mechanisms that underlie disorders of ovarian reserve could improve the outcomes for patients struggling with infertility, but these disorders are diverse and can be categorized in multiple ways. In this review, we will explore the topic from a perspective that emphasizes the prevention or mitigation of ovarian damage. The most desirable mode of fertoprotection is primary prevention (intervening before ablative influence occurs), as identifying toxic influences and deciphering the mechanisms by which they exert their effect can reduce or eliminate exposure and damage. Secondary prevention in the form of screening is not recommended broadly. Nevertheless, in some instances where a known genetic background exists in discrete families, screening is advised. As part of prenatal care, screening panels include some genetic diseases that can lead to infertility or subfertility. In these patients, early diagnosis could enable fertility preservation or changes in family-building plans. Finally, Tertiary Prevention (managing disease post-diagnosis) is critical. Reduced ovarian reserve has a major influence on physiology beyond fertility, including delayed/absent puberty or premature menopause. In these instances, proper diagnosis and medical therapy can reduce adverse effects. Here, we elaborate on these modes of prevention as well as proposed mechanisms that underlie ovarian reserve disorders.
Collapse
|
31
|
Zhang J, Wei L, Deng X, Luo C, Zhu Q, Lu S, Mao C. Current status and reflections on fertility preservation in China. J Assist Reprod Genet 2022; 39:2835-2845. [PMID: 36322229 PMCID: PMC9790826 DOI: 10.1007/s10815-022-02648-0] [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] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 10/20/2022] [Indexed: 11/07/2022] Open
Abstract
PURPOSE With the progress of medical technology and renovated conception of fertility, the prospective studies and practice of fertility preservation are drawing more and more attention from medical workers. With the largest population of over 1.4 billion, China makes the experience accumulated in fertility preservation efforts even more relevant. This article summarizes China's experience and shares it with the world to promote the healthy development of fertility preservation. METHODS This study was based on multiple Chinese expert consensuses on fertility preservation issued in 2021 and the current national regulations and principles, compared with the latest advice and guidelines issued by global reproductive authorities such as the ASRM and ESHRE. Summarize the experience and reflection of Chinese scholars in the process of fertility preservation. RESULTS This study reports on the current situation of fertility preservation in China, sharing the Chinese experience gained in the process of development, and offering Chinese reflections on worrying issues. CONCLUSION Fertility preservation is a medical and social issue of reproductive health security, which is conducive to the sound development of the world population and social production.
Collapse
Affiliation(s)
- Jiakai Zhang
- Reproductive Medicine Center, First Affiliated Hospital of Soochow University, 899 Pinghai Rd, Suzhou Suzhou, Jiangsu, 215000 China
- Marxism Research Institute, Soochow University, Suzhou, Jiangsu, 215123 China
- Suzhou High School Affiliated to Xi’an Jiaotong University, Suzhou, Jiangsu, 215000 China
| | - Lun Wei
- Reproductive Medicine Center, First Affiliated Hospital of Soochow University, 899 Pinghai Rd, Suzhou Suzhou, Jiangsu, 215000 China
| | - Xiaoling Deng
- Reproductive Medicine Center, First Affiliated Hospital of Soochow University, 899 Pinghai Rd, Suzhou Suzhou, Jiangsu, 215000 China
| | - Chao Luo
- Reproductive Medicine Center, First Affiliated Hospital of Soochow University, 899 Pinghai Rd, Suzhou Suzhou, Jiangsu, 215000 China
| | - Qianmeng Zhu
- Reproductive Medicine Center, First Affiliated Hospital of Soochow University, 899 Pinghai Rd, Suzhou Suzhou, Jiangsu, 215000 China
| | - Shucheng Lu
- Marxism Research Institute, Soochow University, Suzhou, Jiangsu, 215123 China
| | - Caiping Mao
- Reproductive Medicine Center, First Affiliated Hospital of Soochow University, 899 Pinghai Rd, Suzhou Suzhou, Jiangsu, 215000 China
| |
Collapse
|
32
|
A predictive model of the effect of therapeutic radiation on the human ovary. PLoS One 2022; 17:e0277052. [PMID: 36399448 PMCID: PMC9674157 DOI: 10.1371/journal.pone.0277052] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 10/18/2022] [Indexed: 11/19/2022] Open
Abstract
Radiation to the female pelvis as part of treatment for cancer predisposes young women to develop Premature Ovarian Insufficiency (POI). As the human female is born with their full complement of non-growing follicles which decline in an exponential fashion until the menopause, the age at which POI occurs is dependent on the age of the patient at treatment and the dose received by the ovary. A model that predicts the age at which POI occurs for a known dose at a known age will aid counselling patients on their fertility risk. Patients deemed to be at high risk of POI may be considered to be good candidates for established fertility preservation techniques. An updated and externally validated model of the age-related decline in human ovarian reserve was combined with the best available estimate of the median lethal dose LD50 for the human ovary. Using known age at diagnosis and posited radiotherapy treatment plan to estimate the dose to the least-affected ovary, we use an age-related model of the decline in ovarian reserve to generate a personalized age prediction of premature ovarian insufficiency. Our algorithm is available as an online calculator which graphs model outputs to inform discussions around survivor fertility. We report four example cases across different ages and diagnoses, each with two carefully designed photon and proton treatment plans. The treatment options are compared in terms of remaining fertile lifespan for the survivor. International oncology guidelines now mandate the consideration of later fertility when reviewing treatment options for children diagnosed with cancer. Our calculator (https://sites.cs.st-andrews.ac.uk/radiosensitivity), and the underlying algorithm and models, allow detailed predictions of the impact of various radiotherapy plans on fertility. These patient-specific data enhance pre-treatment discussions around post-treatment fertility and fertility preservation.
Collapse
|
33
|
Claude F, Ubertini G, Szinnai G. Endocrine Disorders in Children with Brain Tumors: At Diagnosis, after Surgery, Radiotherapy and Chemotherapy. CHILDREN (BASEL, SWITZERLAND) 2022; 9:1617. [PMID: 36360345 PMCID: PMC9688119 DOI: 10.3390/children9111617] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 10/12/2022] [Accepted: 10/20/2022] [Indexed: 10/28/2023]
Abstract
INTRODUCTION Brain tumors are the second most frequent type of all pediatric malignancies. Depending on their localization, patients with brain tumors may present neurological or ophthalmological symptoms, but also weight anomalies and endocrine disorders ranging from growth hormone deficiency, anomalies of puberty, diabetes insipidus to panhypopituitarism. Immediately at diagnosis, all patients with brain tumors require a complete assessment of the hypothalamic-pituitary function in order to address eventual endocrine disorders. Moreover, children and adolescents undergoing brain surgery must receive peri- and postoperative hydrocortisone stress therapy. Post-operative disorders of water homeostasis are frequent, ranging from transient diabetes insipidus, as well as syndrome of inappropriate antidiuretic hormone secretion to persistent diabetes insipidus. Late endocrine disorders may result from surgery near or within the hypothalamic-pituitary region. Pituitary deficits are frequent after radiotherapy, especially growth hormone deficiency. Thyroid nodules or secondary thyroid cancers may arise years after radiotherapy. Gonadal dysfunction is frequent after chemotherapy especially with alkylating agents. CONCLUSION Early detection and treatment of specific endocrine disorders at diagnosis, perioperatively, and during long-term follow-up result in improved general and metabolic health and quality of life.
Collapse
Affiliation(s)
- Fabien Claude
- Department of Pediatric Endocrinology and Diabetology, University Children’s Hospital Basel, University of Basel, 4056 Basel, Switzerland
| | - Graziamaria Ubertini
- Department of Pediatric Endocrinology, Bambino Gesù Children’s Hospital, 00165 Rome, Italy
| | - Gabor Szinnai
- Department of Pediatric Endocrinology and Diabetology, University Children’s Hospital Basel, University of Basel, 4056 Basel, Switzerland
- Department of Clinical Research, University Hospital Basel, University of Basel, 4056 Basel, Switzerland
| |
Collapse
|
34
|
van der Perk MEM, van der Kooi ALLF, Bos AME, Broer SL, Veening MA, van Leeuwen J, van Santen HM, van Dorp W, van den Heuvel-Eibrink MM. Oncofertility Perspectives for Girls with Cancer. J Pediatr Adolesc Gynecol 2022; 35:523-526. [PMID: 35358705 DOI: 10.1016/j.jpag.2022.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 03/03/2022] [Accepted: 03/18/2022] [Indexed: 11/17/2022]
Abstract
Infertility is a serious early, as well as late, effect of childhood cancer treatment. If addressed in a timely manner at diagnosis, fertility preservation measures can be taken, preferably before the start of cancer treatment. However, pediatric oncologists might remain reluctant to offer counseling on fertility-preservation methods, although infrastructure to freeze ovarian tissue has become available and is currently considered standard care for pre- and postpubertal girls at high risk of gonadal damage. More importantly, risk factors have been identified for cancer treatment-related impairment of gonadal function, and the first successful pregnancies have been reported after autotransplanted ovarian tissue, which has been harvested from children. Additionally, great progress has been made in the field of ex vivo maturation of oocytes in frozen ovarian tissue, which provides opportunities for those at risk of ovarian micrometastasis. Hence, it is time to counsel girls at risk and make every effort to cryopreserve their ovarian tissue, now more than ever before.
Collapse
Affiliation(s)
| | | | - Annelies M E Bos
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Department of Reproductive Medicine and Gynecology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Simone L Broer
- Department of Reproductive Medicine and Gynecology, University Medical Center Utrecht, Utrecht, the Netherlands
| | | | - Jeanette van Leeuwen
- Department of Reproductive Medicine and Gynecology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Hanneke M van Santen
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Department of Pediatric Endocrinology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Wendy van Dorp
- Department of Obstetrics and Gynecology, IJsselland Ziekenhuis, Rotterdam, the Netherlands
| | | |
Collapse
|
35
|
van Iersel L, Mulder RL, Denzer C, Cohen LE, Spoudeas HA, Meacham LR, Sugden E, Schouten-van Meeteren AYN, Hoving EW, Packer RJ, Armstrong GT, Mostoufi-Moab S, Stades AM, van Vuurden D, Janssens GO, Thomas-Teinturier C, Murray RD, Di Iorgi N, Neggers SJCMM, Thompson J, Toogood AA, Gleeson H, Follin C, Bardi E, Torno L, Patterson B, Morsellino V, Sommer G, Clement SC, Srivastava D, Kiserud CE, Fernandez A, Scheinemann K, Raman S, Yuen KCJ, Wallace WH, Constine LS, Skinner R, Hudson MM, Kremer LCM, Chemaitilly W, van Santen HM. Hypothalamic-Pituitary and Other Endocrine Surveillance Among Childhood Cancer Survivors. Endocr Rev 2022; 43:794-823. [PMID: 34962573 DOI: 10.1210/endrev/bnab040] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Indexed: 12/12/2022]
Abstract
Endocrine disorders in survivors of childhood, adolescent, and young adult (CAYA) cancers are associated with substantial adverse physical and psychosocial effects. To improve appropriate and timely endocrine screening and referral to a specialist, the International Late Effects of Childhood Cancer Guideline Harmonization Group (IGHG) aims to develop evidence and expert consensus-based guidelines for healthcare providers that harmonize recommendations for surveillance of endocrine disorders in CAYA cancer survivors. Existing IGHG surveillance recommendations for premature ovarian insufficiency, gonadotoxicity in males, fertility preservation, and thyroid cancer are summarized. For hypothalamic-pituitary (HP) dysfunction, new surveillance recommendations were formulated by a guideline panel consisting of 42 interdisciplinary international experts. A systematic literature search was performed in MEDLINE (through PubMed) for clinically relevant questions concerning HP dysfunction. Literature was screened for eligibility. Recommendations were formulated by drawing conclusions from quality assessment of all evidence, considering the potential benefits of early detection and appropriate management. Healthcare providers should be aware that CAYA cancer survivors have an increased risk for endocrine disorders, including HP dysfunction. Regular surveillance with clinical history, anthropomorphic measures, physical examination, and laboratory measurements is recommended in at-risk survivors. When endocrine disorders are suspected, healthcare providers should proceed with timely referrals to specialized services. These international evidence-based recommendations for surveillance of endocrine disorders in CAYA cancer survivors inform healthcare providers and highlight the need for long-term endocrine follow-up care in subgroups of survivors and elucidate opportunities for further research.
Collapse
Affiliation(s)
- Laura van Iersel
- Department of Pediatric Endocrinology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Renee L Mulder
- Princess Máxima Center for Pediatric Oncology, Department of Neuro-oncology, Utrecht, The Netherlands
| | - Christian Denzer
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics & Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - Laurie E Cohen
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, USA.,Dana Farber/Boston Children's Cancer and Blood Disorder Center, Boston, MA, USA.,Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Helen A Spoudeas
- The London Centre for Pediatric Endocrinology & Diabetes, London, UK.,Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK.,The London Centre for Pediatric Endocrinology and Diabetes, University College London Hospital, London, UK
| | - Lillian R Meacham
- Emory University School of Medicine; Atlanta, GA, USA.,Aflac Cancer and Blood Disorders Center of Children's Healthcare of Atlanta, Atlanta, GA,USA
| | | | | | - Eelco W Hoving
- Princess Máxima Center for Pediatric Oncology, Department of Neuro-oncology, Utrecht, The Netherlands
| | - Roger J Packer
- The Brain Tumor Institute, Center for Neuroscience and Behavioral Medicine, Children's National Health System, Washington, DC, USA
| | - Gregory T Armstrong
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis TN, USA
| | - Sogol Mostoufi-Moab
- Department of Pediatrics, The Children's Hospital of Philadelphia, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA,USA
| | - Aline M Stades
- Department of Endocrinology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Dannis van Vuurden
- Princess Máxima Center for Pediatric Oncology, Department of Neuro-oncology, Utrecht, The Netherlands
| | - Geert O Janssens
- Princess Máxima Center for Pediatric Oncology, Department of Neuro-oncology, Utrecht, The Netherlands.,Department of Radiation Oncology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Cécile Thomas-Teinturier
- Radiation Epidemiology Group, Center for Research in Epidemiology and Population Health (CESP), Université Paris-Sud XI, Villejuif, France.,Department of Pediatric Endocrinology, APHP, Hôpitaux Paris-Sud, Site Bicetre, Le Kremlin-Bicetre, France
| | - Robert D Murray
- Department of Endocrinology, Leeds Centre for Diabetes & Endocrinology, Leeds Teaching Hospitals NHS Trust, Leeds, UK.,Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Natascia Di Iorgi
- Department of Pediatrics, Istituto Giannina Gaslini, University of Genova, Genova, Italy
| | - Sebastian J C M M Neggers
- Department of Internal Medicine, Endocrinology Section, Pituitary Center Rotterdam, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Joel Thompson
- Division of Hematology/Oncology/BMT, Children's Mercy Hospitals and Clinics, Kansas City, Missouri, USA
| | - Andrew A Toogood
- Department of Endocrinology, Queen Elizabeth Hospital Birmingham, Birmingham, UK
| | - Helena Gleeson
- Department of Endocrinology, Queen Elizabeth Hospital Birmingham, Birmingham, UK
| | - Cecilia Follin
- Department of Oncology, Skåne University Hospital, Lund, Sweden
| | - Edit Bardi
- Department of Pediatrics and Adolescent Medicine, Kepler Universitätsklinikum, Linz, Austria.,St Anna Childrens Hospital, Vienna, Austria
| | - Lilibeth Torno
- Division of Pediatric Oncology, CHOC Children's Hospital/University of California, Orange, CA, USA
| | - Briana Patterson
- Emory University School of Medicine; Atlanta, GA, USA.,Aflac Cancer and Blood Disorders Center of Children's Healthcare of Atlanta, Atlanta, GA,USA
| | - Vera Morsellino
- DOPO Clinic, Division of Pediatric Hematology/Oncology, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Grit Sommer
- Swiss Childhood Cancer Registry, Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland.,Pediatric Endocrinology, Diabetology and Metabolism, Department of Pediatrics, Bern University Hospital, University of Bern, Switzerland
| | - Sarah C Clement
- Department of Pediatrics, Amsterdam University Medical Center, location VU University Medical Center, Amsterdam, The Netherlands
| | - Deokumar Srivastava
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis TN, USA
| | - Cecilie E Kiserud
- Department of Oncology, National Advisory Unit on Late Effects after Cancer Treatment, Oslo University Hospital, Oslo, Norway
| | - Alberto Fernandez
- Endocrinology Department, Hospital Universitario de Mostoles, Madrid, Spain
| | - Katrin Scheinemann
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Kantonsspital Aarau, Aarau, Switzerland.,Division of Pediatric Hematology/Oncology, University Children's Hospital Basel and University of Basel, Basel, Switzerland.,Division of Pediatric Hematology/Oncology, McMaster Children's Hospital and McMaster University, Hamilton, ON, Canada
| | - Sripriya Raman
- Division of Pediatric Endocrinology and Diabetes, Children's Hospital of Pittsburgh, Pittsburgh, PA,USA
| | - Kevin C J Yuen
- Department of Neuroendocrinology and Neurosurgery, Barrow Pituitary Center, Barrow Neurological Institute, University of Arizona College of Medicine, Phoenix, AZ, USA
| | - W Hamish Wallace
- Department of Paediatric Oncology, Royal Hospital for Sick Children, Edinburgh, UK
| | - Louis S Constine
- Departments of Radiation Oncology and Pediatrics, University of Rochester Medical Center, Rochester, NY, USA
| | - Roderick Skinner
- Department of Paediatric and Adolescent Haematology/Oncology, Great North Children's Hospital and Newcastle University Centre for Cancer, Newcastle upon Tyne, UK
| | - Melissa M Hudson
- Department of Epidemiology and Cancer Control, and Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Leontien C M Kremer
- Princess Máxima Center for Pediatric Oncology, Department of Neuro-oncology, Utrecht, The Netherlands.,Department of Pediatric Oncology, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Wassim Chemaitilly
- Division of Endocrinology and Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Hanneke M van Santen
- Department of Pediatric Endocrinology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
36
|
Li Y, Cai X, Dong B, Wang Q, Yang X, Yu A, Wei H, Ke Z, Sun P, Zheng B, Sun Y. The Impact of Malignancy on Assisted Reproductive Outcomes for Cancer Survivors: A Retrospective Case–Control Study. Front Oncol 2022; 12:941797. [PMID: 36185197 PMCID: PMC9523265 DOI: 10.3389/fonc.2022.941797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 08/30/2022] [Indexed: 12/24/2022] Open
Abstract
BackgroundRelated studies have shown that it is safe for cancer patients to undergo assisted reproduction. However, studies on whether a history of cancer affects long-term reproductive outcomes in women who undergo assisted reproductive technology (ART) are scarce. In this study, we evaluated the long-term reproductive outcomes of patients with malignant tumors undergoing ART treatment and explored the impact of malignancy history on ART outcomes.MethodsThis retrospective study analyzed the clinical outcomes of patients with malignant tumors undergoing their first in vitro fertilization/intracytoplasmic sperm injection (IVF/ICSI) cycles compared with those of age-matched healthy infertile women at Fujian Maternity and Child Health Hospital between January 2003 and October 2020. We evaluated ovarian stimulation outcome, the pregnancy rate, the live birth rate, the risk of adverse obstetric outcomes and birth outcomes.ResultsThis study included 59 patients in the cancer group for data analysis who had a history of malignancy. By matching, a total of 118 healthy infertile women were included in the control group. No statistically significant association was found in terms of age, duration of infertility, BMI, or insemination type between the two groups of patients. Thyroid cancer(45.8%) and gynecologic malignancies (44.07%) were the major cancer types in this study. There were statistically significant differences in the antral follicle count (AFC) (12.00 ± 7.86 vs. 14.90 ± 8.71, P=0.033), length of ovarian stimulation (9.98 ± 2.68 vs. 11.42 ± 2.43, P=0.033) and endometrial thickness on the trigger day (10.16 ± 3.11 vs. 10.84 ± 2.17, P<0.001) between the two groups. The total gonadotropin dose, number of oocytes retrieved, fertilization rate, cleavage rate, high-quality embryo rate, blastocyst rate and first-time embryo-transfer (ET) implantation rate were nonsignificantly lower in the cancer group than in the control group (P>0.05). There were no significant differences in the clinical pregnancy rate per ET cycle (32% vs. 40.39%, P=0.156), live birth rate per ET cycle (27% vs. 35.96%, P=0.119), miscarriage rate per ET cycle (5% vs. 4.43%, P=0.779), or preterm delivery rate per ET cycle (11.11% vs. 17.80%, P=0.547) between the two groups. Additionally, regression analysis showed that a history of malignancy was not a risk factor for reproductive outcomes.ConclusionsOverall, it is feasible for women with a history of cancer to conceive using ART is feasible and their long-term reproductive outcomes are similar to these of healthy infertile women. A history of cancer does not decrease the number of retrieved oocytes, increase the risk of adverse obstetric outcomes or affect birth outcomes.
Collapse
Affiliation(s)
- Yuehong Li
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Xuefen Cai
- Center of Reproductive Medicine, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Binhua Dong
- Laboratory of Gynecologic Oncology, Department of Gynecology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Women and Children’s Critical Diseases Research, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Qi Wang
- Fujian Province Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, China
| | - Xiaohui Yang
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Aili Yu
- Center of Reproductive Medicine, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Huijuan Wei
- Center of Reproductive Medicine, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Zhanghong Ke
- Center of Reproductive Medicine, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Pengming Sun
- Laboratory of Gynecologic Oncology, Department of Gynecology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Women and Children’s Critical Diseases Research, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
- *Correspondence: Pengming Sun, ; Beihong Zheng, ; Yan Sun,
| | - Beihong Zheng
- Center of Reproductive Medicine, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
- *Correspondence: Pengming Sun, ; Beihong Zheng, ; Yan Sun,
| | - Yan Sun
- Center of Reproductive Medicine, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
- *Correspondence: Pengming Sun, ; Beihong Zheng, ; Yan Sun,
| |
Collapse
|
37
|
Matilionyte G, Rimmer MP, Spears N, Anderson RA, Mitchell RT. Cisplatin Effects on the Human Fetal Testis - Establishing the Sensitive Period for (Pre)Spermatogonial Loss and Relevance for Fertility Preservation in Pre-Pubertal Boys. Front Endocrinol (Lausanne) 2022; 13:914443. [PMID: 35909565 PMCID: PMC9330899 DOI: 10.3389/fendo.2022.914443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 06/07/2022] [Indexed: 11/18/2022] Open
Abstract
Background Exposure to chemotherapy during childhood can impair future fertility. Studies using in vitro culture have shown exposure to platinum-based alkylating-like chemotherapy reduces the germ cell number in the human fetal testicular tissues. We aimed to determine whether effects of exposure to cisplatin on the germ cell sub-populations are dependent on the gestational age of the fetus and what impact this might have on the utility of using human fetal testis cultures to model chemotherapy exposure in childhood testis. Methods We utilised an in vitro culture system to culture pieces of human fetal testicular tissues (total n=23 fetuses) from three different gestational age groups (14-16 (early), 17-19 (mid) and 20-22 (late) gestational weeks; GW) of the second trimester. Tissues were exposed to cisplatin or vehicle control for 24 hours, analysing the tissues 72 and 240 hours post-exposure. Number of germ cells and their sub-populations, including gonocytes and (pre)spermatogonia, were quantified. Results Total germ cell number and number of both germ cell sub-populations were unchanged at 72 hours post-exposure to cisplatin in the testicular tissues from fetuses of the early (14-16 GW) and late (20-22 GW) second trimester. In the testicular tissues from fetuses of mid (17-19 GW) second trimester, total germ cell and gonocyte number were significantly reduced, whilst (pre)spermatogonial number was unchanged. At 240 hours post-exposure, the total number of germ cells and that of both sub-populations was significantly reduced in the testicular tissues from fetuses of mid- and late-second trimester, whilst germ cells in early-second trimester tissues were unchanged at this time-point. Conclusions In vitro culture of human fetal testicular tissues can be a useful model system to investigate the effects of chemotherapy-exposure on germ cell sub-populations during pre-puberty. Interpretation of the results of such studies in terms of relevance to later (infant and pre-pubertal) developmental stages should take into account the changes in germ cell composition and periods of germ cell sensitivity in the human fetal testis.
Collapse
Affiliation(s)
- Gabriele Matilionyte
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Michael P Rimmer
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Norah Spears
- Biomedical Sciences, The University of Edinburgh, Edinburgh, United Kingdom
| | - Richard A Anderson
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Rod T Mitchell
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
- Department of Paediatric Diabetes and Endocrinology, Royal Hospital for Children & Young People, Edinburgh, United Kingdom
| |
Collapse
|
38
|
Cioffi R, Cervini L, Taccagni G, Papaleo E, Pagliardini L, Bergamini A, Ferrari S, Mangili G, Candiani M. A prospective, observational study of chemotherapy-induced ovarian damage on follicular reserve and maturation. Arch Gynecol Obstet 2022; 306:1723-1729. [PMID: 35833992 DOI: 10.1007/s00404-022-06692-0] [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: 02/24/2022] [Accepted: 06/26/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Chemotherapy negatively affects gonadal function, often resulting in premature ovarian failure (POF) due to ovarian reserve depletion. Mechanisms of gonadotoxicity, such as primordial follicle overactivation and "burnout", remain to be established. Ovarian tissue cryopreservation (OTC) before treatment plays an important role in safeguarding fertility. METHODS This is a prospective observational study that aims to evaluate the feasibility of OTC after chemotherapeutic treatment initiation. Patients were divided into 2 groups depending on whether they received chemotherapy before the harvesting procedure (Group 1) or not (Group 2). The main outcomes of this study are serum anti-Mullerian hormone (AMH) levels and histological follicular counts on ovarian tissue biopsies. RESULTS Between 2012 and 2020, 79 patients underwent OTC at our Hospital. Follicular counts from the ovarian biopsies of 30 post-pubertal patients and respective serum AMH levels were included in the analysis. AMH levels did not significantly differ between the 2 groups (P = 0.70) as well as the number of primordial follicles (P = 0.73). Ovarian biopsies of patients from Group 1 showed a higher number of primary follicles (P = 0.04) and atretic follicles (P = 0.05) with respect to Group 2. CONCLUSIONS In conclusion, OTC appears to be feasible even after the start of chemotherapeutic treatment, since in treated patients, the main ovarian reserve indicators (number of primordial follicles and serum AMH levels) were not significantly reduced compared to untreated patients. The "burnout" theory of chemotherapeutic damage to the ovary seems to be supported by the higher number of primary follicles found in the ovaries of patients who received chemotherapy before OTC.
Collapse
Affiliation(s)
- Raffaella Cioffi
- Obstetrics and Gynecology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy.
| | - Laura Cervini
- Obstetrics and Gynecology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Gianluca Taccagni
- Pathology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Enrico Papaleo
- Obstetrics and Gynecology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Luca Pagliardini
- Division of Genetics and Cell Biology, Reproductive Sciences Laboratory, San Raffaele Scientific Institute, Milan, Italy
| | - Alice Bergamini
- Obstetrics and Gynecology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Stefano Ferrari
- Obstetrics and Gynecology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Giorgia Mangili
- Obstetrics and Gynecology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Massimo Candiani
- Obstetrics and Gynecology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| |
Collapse
|
39
|
Ghidei L, Sullivan J, Valero Carrion RJ, Schammel J, Lipshultz L, McKenzie LJ. Current Gaps in Fertility Preservation for Men: How Can We do Better? J Clin Oncol 2022; 40:2524-2529. [PMID: 35724344 DOI: 10.1200/jco.21.02714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Luwam Ghidei
- Division of Reproductive Endocrinology and Infertility, Baylor College of Medicine, One Baylor Plaza, Houston, TX
| | - John Sullivan
- Department of Urology, Baylor College of Medicine, One Baylor Plaza, Houston, TX
| | | | - Joshua Schammel
- Department of Urology, Baylor College of Medicine, One Baylor Plaza, Houston, TX
| | - Larry Lipshultz
- Department of Urology, Baylor College of Medicine, One Baylor Plaza, Houston, TX
| | - Laurie J McKenzie
- Division of Reproductive Endocrinology and Infertility, Baylor College of Medicine, One Baylor Plaza, Houston, TX.,Department of Gynecology Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
40
|
Whelan EC, Yang F, Avarbock MR, Sullivan MC, Beiting DP, Brinster RL. Reestablishment of spermatogenesis after more than 20 years of cryopreservation of rat spermatogonial stem cells reveals an important impact in differentiation capacity. PLoS Biol 2022; 20:e3001618. [PMID: 35536782 PMCID: PMC9089916 DOI: 10.1371/journal.pbio.3001618] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 04/04/2022] [Indexed: 12/22/2022] Open
Abstract
Treatment of cancer in children is increasingly successful but leaves many prepubertal boys suffering from infertility or subfertility later in life. A current strategy to preserve fertility in these boys is to cryopreserve a testicular biopsy prior to treatment with the expectation of future technologies allowing for the reintroduction of stem cells and restoration of spermatogenesis. Spermatogonial stem cells (SSCs) form the basis of male reproduction, differentiating into all germ cell types, including mature spermatozoa and can regenerate spermatogenesis following transplantation into an infertile testis. Here, we demonstrate that rat SSCs frozen for more than 20 years can be transplanted into recipient mice and produce all differentiating germ cell types. However, compared with freshly isolated cells or those frozen for a short period of time, long-frozen cells do not colonize efficiently and showed reduced production of spermatids. Single-cell RNA sequencing revealed similar profiles of gene expression changes between short- and long-frozen cells as compared with fresh immediately after thawing. Conversely, following transplantation, long-frozen samples showed enhanced stem cell signaling in the undifferentiated spermatogonia compartment, consistent with self-renewal and a lack of differentiation. In addition, long-frozen samples showed fewer round spermatids with detectable protamine expression, suggesting a partial block of spermatogenesis after meiosis resulting in a lack of elongating spermatids. These findings strongly suggest that prolonged cryopreservation can impact the success of transplantation to produce spermatogenesis, which may not be revealed by analysis of the cells immediately after thawing. Our analysis uncovered persistent effects of long-term freezing not found in other cryopreservation studies that lacked functional regeneration of the tissue and this phenomenon must be accounted for any future therapeutic application. This study shows that spermatogonial stem cells frozen for more than 20 years can repopulate the niche when transplanted into an infertile host, but with lower efficiency and displaying abnormal spermatogenesis compared to fresh or short-term frozen cells. Single-cell RNA sequencing reveals details of the dysregulated signaling.
Collapse
Affiliation(s)
- Eoin C. Whelan
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Fan Yang
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Department of Histology and Embryology, Medical College, Yangzhou University, Yangzhou, China
| | - Mary R. Avarbock
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Megan C. Sullivan
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Daniel P. Beiting
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Ralph L. Brinster
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
41
|
van der Perk MEM, Stukaitė-Ruibienė E, Bumbulienė Ž, Vaitkevičienė GE, Bos AME, van den Heuvel-Eibrink MM, Rascon J. Development of a questionnaire to evaluate female fertility care in pediatric oncology, a TREL initiative. BMC Cancer 2022; 22:450. [PMID: 35468746 PMCID: PMC9036799 DOI: 10.1186/s12885-022-09450-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 03/11/2022] [Indexed: 11/28/2022] Open
Abstract
Background Currently the five-year survival of childhood cancer is up to 80% due to improved treatment modalities. However, the majority of childhood cancer survivors develop late effects including infertility. Survivors describe infertility as an important and life-altering late effect. Fertility preservation options are becoming available to pre- and postpubertal patients diagnosed with childhood cancer and fertility care is now an important aspect in cancer treatment. The use of fertility preservation options depends on the quality of counseling on this important and delicate issue. The aim of this manuscript is to present a questionnaire to determine the impact of fertility counseling in patients suffering from childhood cancer, to improve fertility care and evaluate what patients and their parents or guardians consider good fertility care. Methods Within the framework of the EU-Horizon 2020 TREL project, a fertility care evaluation questionnaire used in the Netherlands was made applicable for international multi-center use. The questionnaire to be used at least also in Lithuania, incorporates patients’ views on fertility care to further improve the quality of fertility care and counseling. Results evaluate fertility care and will be used to improve current fertility care in a national specialized pediatric oncology center in the Netherlands and a pediatric oncology center in Lithuania. Conclusion An oncofertility-care-evaluation questionnaire has been developed for pediatric oncology patients and their families specifically. Results of this questionnaire may contribute to enhancement of fertility care in pediatric oncology in wider settings and thus improve quality of life of childhood cancer patients and survivors. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09450-2.
Collapse
Affiliation(s)
| | | | - Žana Bumbulienė
- Vilnius University, Faculty of Medicine, Vilnius, Lithuania.,Center of Obstetrics and Gynaecology, Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
| | - Goda Elizabeta Vaitkevičienė
- Vilnius University, Faculty of Medicine, Vilnius, Lithuania.,Center for Pediatric Oncology and Hematology, Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
| | - Annelies M E Bos
- University Medical Center Utrecht, Reproductive Medicine and Gynaecology, Utrecht, The Netherlands
| | | | - Jelena Rascon
- Vilnius University, Faculty of Medicine, Vilnius, Lithuania.,Center for Pediatric Oncology and Hematology, Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
| |
Collapse
|
42
|
Sanou I, van Maaren J, Eliveld J, Lei Q, Meißner A, de Melker AA, Hamer G, van Pelt AMM, Mulder CL. Spermatogonial Stem Cell-Based Therapies: Taking Preclinical Research to the Next Level. Front Endocrinol (Lausanne) 2022; 13:850219. [PMID: 35444616 PMCID: PMC9013905 DOI: 10.3389/fendo.2022.850219] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 03/07/2022] [Indexed: 01/15/2023] Open
Abstract
Fertility preservation via biobanking of testicular tissue retrieved from testicular biopsies is now generally recommended for boys who need to undergo gonadotoxic treatment prior to the onset of puberty, as a source of spermatogonial stem cells (SSCs). SSCs have the potential of forming spermatids and may be used for therapeutic fertility approaches later in life. Although in the past 30 years many milestones have been reached to work towards SSC-based fertility restoration therapies, including transplantation of SSCs, grafting of testicular tissue and various in vitro and ex vivo spermatogenesis approaches, unfortunately, all these fertility therapies are still in a preclinical phase and not yet available for patients who have become infertile because of their treatment during childhood. Therefore, it is now time to take the preclinical research towards SSC-based therapy to the next level to resolve major issues that impede clinical implementation. This review gives an outline of the state of the art of the effectiveness and safety of fertility preservation and SSC-based therapies and addresses the hurdles that need to be taken for optimal progression towards actual clinical implementation of safe and effective SSC-based fertility treatments in the near future.
Collapse
Affiliation(s)
- Iris Sanou
- Reproductive Biology Laboratory, Center for Reproductive Medicine, Amsterdam University Medical Center (UMC), Amsterdam Reproduction and Development Research Institute, University of Amsterdam, Amsterdam, Netherlands
| | - Jillis van Maaren
- Reproductive Biology Laboratory, Center for Reproductive Medicine, Amsterdam University Medical Center (UMC), Amsterdam Reproduction and Development Research Institute, University of Amsterdam, Amsterdam, Netherlands
| | - Jitske Eliveld
- Reproductive Biology Laboratory, Center for Reproductive Medicine, Amsterdam University Medical Center (UMC), Amsterdam Reproduction and Development Research Institute, University of Amsterdam, Amsterdam, Netherlands
| | - Qijing Lei
- Reproductive Biology Laboratory, Center for Reproductive Medicine, Amsterdam University Medical Center (UMC), Amsterdam Reproduction and Development Research Institute, University of Amsterdam, Amsterdam, Netherlands
| | - Andreas Meißner
- Reproductive Biology Laboratory, Center for Reproductive Medicine, Amsterdam University Medical Center (UMC), Amsterdam Reproduction and Development Research Institute, University of Amsterdam, Amsterdam, Netherlands
- Department of Urology, Center for Reproductive Medicine, Amsterdam University Medical Center (UMC), Amsterdam Reproduction and Development Research Institute, University of Amsterdam, Amsterdam, Netherlands
| | - Annemieke A de Melker
- Reproductive Biology Laboratory, Center for Reproductive Medicine, Amsterdam University Medical Center (UMC), Amsterdam Reproduction and Development Research Institute, University of Amsterdam, Amsterdam, Netherlands
| | - Geert Hamer
- Reproductive Biology Laboratory, Center for Reproductive Medicine, Amsterdam University Medical Center (UMC), Amsterdam Reproduction and Development Research Institute, University of Amsterdam, Amsterdam, Netherlands
| | - Ans M M van Pelt
- Reproductive Biology Laboratory, Center for Reproductive Medicine, Amsterdam University Medical Center (UMC), Amsterdam Reproduction and Development Research Institute, University of Amsterdam, Amsterdam, Netherlands
| | - Callista L Mulder
- Reproductive Biology Laboratory, Center for Reproductive Medicine, Amsterdam University Medical Center (UMC), Amsterdam Reproduction and Development Research Institute, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
43
|
van der Perk MEM, Cost NG, Bos AME, Brannigan R, Chowdhury T, Davidoff AM, Daw NC, Dome JS, Ehrlich P, Graf N, Geller J, Kalapurakal J, Kieran K, Malek M, McAleer MF, Mullen E, Pater L, Polanco A, Romao R, Saltzman AF, Walz AL, Woods AD, van den Heuvel-Eibrink MM, Fernandez CV. White paper: Onco-fertility in pediatric patients with Wilms tumor. Int J Cancer 2022; 151:843-858. [PMID: 35342935 PMCID: PMC9541948 DOI: 10.1002/ijc.34006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/11/2022] [Accepted: 02/17/2022] [Indexed: 11/18/2022]
Abstract
The survival of childhood Wilms tumor is currently around 90%, with many survivors reaching reproductive age. Chemotherapy and radiotherapy are established risk factors for gonadal damage and are used in both COG and SIOP Wilms tumor treatment protocols. The risk of infertility in Wilms tumor patients is low but increases with intensification of treatment including the use of alkylating agents, whole abdominal radiation or radiotherapy to the pelvis. Both COG and SIOP protocols aim to limit the use of gonadotoxic treatment, but unfortunately this cannot be avoided in all patients. Infertility is considered one of the most important late effects of childhood cancer treatment by patients and their families. Thus, timely discussion of gonadal damage risk and fertility preservation options is important. Additionally, irrespective of the choice for preservation, consultation with a fertility preservation (FP) team is associated with decreased patient and family regret and better quality of life. Current guidelines recommend early discussion of the impact of therapy on potential fertility. Since most patients with Wilms tumors are prepubertal, potential FP methods for this group are still considered experimental. There are no proven methods for FP for prepubertal males (testicular biopsy for cryopreservation is experimental), and there is just a single option for prepubertal females (ovarian tissue cryopreservation), posing both technical and ethical challenges. Identification of genetic markers of susceptibility to gonadotoxic therapy may help to stratify patient risk of gonadal damage and identify patients most likely to benefit from FP methods.
Collapse
Affiliation(s)
| | - Nicholas G Cost
- Department of Surgery, Division of Urology, University of Colorado School of Medicine and the Surgical Oncology Program of the Children's Hospital Colorado, Aurora, CO, USA
| | - Annelies M E Bos
- University Medical Center Utrecht, Reproductive Medicine and Gynaecology, Utrecht, Netherlands
| | - Robert Brannigan
- Department of Urology, Northwestern University, Chicago, Illinois, USA
| | - Tanzina Chowdhury
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Andrew M Davidoff
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, USA
| | - Najat C Daw
- Department of Pediatrics - Patient Care, MD Anderson Cancer Center, Houston, TX, USA
| | - Jeffrey S Dome
- Division of Oncology at Children's National Hospital, Washington, DC, USA
| | - Peter Ehrlich
- University of Michigan, C.S. Mott Children's Hospital Section of Pediatric Surgery, Ann Arbor, MI, USA
| | - Norbert Graf
- Department for Pediatric Oncology and Hematology, Saarland University Medical Center, Homburg, Germany
| | - James Geller
- Division of Pediatric Oncology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
| | - John Kalapurakal
- Department of Radiation Oncology, Northwestern University, Chicago, Illinois, USA
| | - Kathleen Kieran
- Department of Urology, University of Washington, and Division of Urology, Seattle Children's Hospital, Seattle, USA
| | - Marcus Malek
- Division of Pediatric General and Thoracic Surgery, UPMC Children's Hospital of Pittsburgh, Pittsburgh, USA
| | - Mary F McAleer
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Elizabeth Mullen
- Department of Pediatric Oncology, Children's Hospital Boston/Dana-Farber Cancer Institute, Boston, MA, USA
| | - Luke Pater
- Department of Radiation Oncology, University of Cincinnati, Cincinnati, Ohio, USA
| | - Angela Polanco
- National Cancer Research Institute Children's Group Consumer Representative, London, UK
| | - Rodrigo Romao
- Departments of Surgery and Urology, IWK Health Centre, Dalhousie University, Halifax, Canada
| | | | - Amy L Walz
- Division of Hematology, Oncology, Neuro-Oncology, and Stem Cell Transplant, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, USA
| | - Andrew D Woods
- Children's Cancer Therapy Development Institute, Beaverton, Oregon, USA
| | | | - Conrad V Fernandez
- Department of Pediatric Hematology/Oncology, IWK Health Centre and Dalhousie University, Halifax, Canada
| |
Collapse
|
44
|
Menstrual blood-derived endometrial stem cells ameliorate the viability of ovarian granulosa cells injured by cisplatin through activating autophagy. Reprod Toxicol 2022; 110:39-48. [PMID: 35346788 DOI: 10.1016/j.reprotox.2022.03.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 03/16/2022] [Accepted: 03/22/2022] [Indexed: 01/10/2023]
Abstract
Although the cancer incidence showed a yearly increasing trend, the long-term survival rate of cancer patients significantly increased with the continuous improvements in cancer diagnosis and treatment. Therefore, recent strategies for cancer treatment not only focus on improving the survival rate of patients but also simultaneously consider the life quality of cancer patients, especially for those with fertility requirements. Stem cell-based therapies have exhibited promising improvement in various disease treatments, and provide hope for diseases without effective treatment. Menstrual blood-derived endometrial stem cells (MenSCs) can be noninvasively and periodically obtained from discarded menstrual blood samples and exhibit high proliferative capacity, low immunogenicity and autologous transplantation. As expected, MenSCs treatment effectively improved the viability of cisplatin-injured ovarian granulosa cells (GCs) and significantly upregulated their antiapoptotic capacity. Further results demonstrated that MenSCs treatment significantly upregulated autophagy activity in cisplatin-injured ovarian GCs, and the degree of autophagy activation was positively correlated with the viability improvement of ovarian GCs, while autophagy inhibitors significantly impaired MenSC-promoted viability improvement of cisplatin-injured ovarian GCs. Additionally, MenSCs treatment can also significantly promote the proliferation of normal GCs by activating the PI3K/Akt signaling pathway. Conclusively, MenSCs treatment not only enhanced the antiapoptotic capacity and survival of cisplatin-injured ovarian GCs by upregulating autophagy activity but also improved the viability of normal ovarian GCs by activating the PI3K/Akt signal pathway. These results provide a theoretical and experimental foundation for the clinical application of MenSCs in improving chemotherapy-induced ovarian injury and delaying ovarian senescence.
Collapse
|
45
|
Matilionyte G, Tharmalingam MD, Sanou I, Lopes F, Lane S, Stukenborg JB, Spears N, Anderson RA, Mitchell RT. Maintenance of Sertoli Cell Number and Function in Immature Human Testicular Tissues Exposed to Platinum-Based Chemotherapy-Implications for Fertility Restoration. FRONTIERS IN TOXICOLOGY 2022; 4:825734. [PMID: 35387428 PMCID: PMC8977418 DOI: 10.3389/ftox.2022.825734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 02/17/2022] [Indexed: 11/18/2022] Open
Abstract
Background: Retrospective studies in adult survivors of childhood cancer show long-term impacts of exposure to alkylating chemotherapy on future fertility. We recently demonstrated germ cell loss in immature human testicular tissues following exposure to platinum-based chemotherapeutic drugs. This study investigated the effects of platinum-based chemotherapy exposure on the somatic Sertoli cell population in human fetal and pre-pubertal testicular tissues. Methods: Human fetal (n = 23; 14-22 gestational weeks) testicular tissue pieces were exposed to cisplatin (0.5 or 1.0 μg/ml) or vehicle for 24 h in vitro and analysed 24-240 h post-exposure or 12 weeks after xenografting. Human pre-pubertal (n = 10; 1-12 years) testicular tissue pieces were exposed to cisplatin (0.5 μg/ml), carboplatin (5 μg/ml) or vehicle for 24 h in vitro and analysed 24-240 h post-exposure; exposure to carboplatin at 10-times the concentration of cisplatin reflects the relative clinical doses given to patients. Immunohistochemistry was performed for SOX9 and anti-Müllerian hormone (AMH) expression and quantification was carried out to assess effects on Sertoli cell number and function respectively. AMH and inhibin B was measured in culture medium collected post-exposure to assess effects on Sertoli cell function. Results: Sertoli cell (SOX9+ve) number was maintained in cisplatin-exposed human fetal testicular tissues (7,647 ± 459 vs. 7,767 ± 498 cells/mm2; p > 0.05) at 240 h post-exposure. No effect on inhibin B (indicator of Sertoli cell function) production was observed at 96 h after cisplatin (0.5 and 1.0 μg/ml) exposure compared to control (21 ± 5 (0.5 μg/ml cisplatin) vs. 23 ± 7 (1.0 μg/ml cisplatin) vs. 25 ± 7 (control) ng/ml, p > 0.05). Xenografting of cisplatin-exposed (0.5 μg/ml) human fetal testicular tissues had no long-term effect on Sertoli cell number or function (percentage seminiferous area stained for SOX9 and AMH, respectively), compared with non-exposed tissues. Sertoli cell number was maintained in human pre-pubertal testicular tissues following exposure to either 0.5 μg/ml cisplatin (6,723 ± 1,647 cells/mm2) or 5 μg/ml carboplatin (7,502 ± 627 cells/mm2) compared to control (6,592 ± 1,545 cells/mm2). Conclusions: This study demonstrates maintenance of Sertoli cell number and function in immature human testicular tissues exposed to platinum-based chemotherapeutic agents. The maintenance of a functional Sertoli cell environment following chemotherapy exposure suggests that fertility restoration by spermatogonial stem cell (SSC) transplant may be possible in boys facing platinum-based cancer treatment.
Collapse
Affiliation(s)
- Gabriele Matilionyte
- MRC Centre for Reproductive Health, Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Melissa D. Tharmalingam
- MRC Centre for Reproductive Health, Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
- KK Women’s and Children’s Hospital, Edinburgh, Singapore
| | - Iris Sanou
- MRC Centre for Reproductive Health, Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Federica Lopes
- MRC Centre for Reproductive Health, Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
- Medical School, University of Dundee, Dundee, United Kingdom
| | - Sheila Lane
- Department of Womens and Reproductive Health, University of Oxford and Oxford University Hospitals NHS Foundation Trust, Edinburgh, United Kingdom
| | - Jan-Bernd Stukenborg
- NORDFERTIL Research Lab Stockholm, Childhood Cancer Research Unit, Department of Women’s and Children’s Health, Karolinska Institutet, Karolinska University Hospital, Solna, Sweden
| | - Norah Spears
- Biomedical Sciences, The University of Edinburgh, Edinburgh, United Kingdom
| | - Richard A. Anderson
- MRC Centre for Reproductive Health, Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Rod T. Mitchell
- MRC Centre for Reproductive Health, Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
- Royal Hospital for Children and Young People, Edinburgh, United Kingdom
| |
Collapse
|
46
|
Lugat A, Drui D, Baron S, Thebaud E, Supiot S, Jouglar E, Doré M. Effets secondaires endocriniens de la radiothérapie : diagnostic, prévention et traitements. Cancer Radiother 2022; 26:1078-1089. [DOI: 10.1016/j.canrad.2021.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 11/19/2021] [Accepted: 12/24/2021] [Indexed: 11/30/2022]
|
47
|
Newton HL, Picton HM, Friend AJ, Hayden CM, Brougham M, Cox R, Grandage V, Kwok-Williams M, Lane S, Mitchell RT, Skinner R, Wallace WH, Yeomanson D, Glaser AW. Inconsistencies in fertility preservation for young people with cancer in the UK. Arch Dis Child 2022; 107:265-270. [PMID: 34544694 DOI: 10.1136/archdischild-2021-321873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 06/29/2021] [Indexed: 11/04/2022]
Abstract
OBJECTIVE To assess the utilisation of and funding structure for fertility preservation for children diagnosed with cancer in the UK. DESIGN Survey of paediatric oncologists/haematologists. Questionnaires were sent electronically with reminder notifications to non-responders. SETTING UK Paediatric Oncology Principal Treatment Centres (PTCs). PARTICIPANTS Paediatric oncologists/haematologists with an interest in the effects of treatment on fertility representing the 20 PTCs across the UK. MAIN OUTCOME MEASURES Referral practices, sources and length of funding for storage of gametes or gonadal tissue for children diagnosed with cancer in the preceding 12 months. RESULTS Responses were received from 18 PTCs (90%) with responses to 98.3% of questions. All centres had referred patients for fertility preservation: ovarian tissue collection/storage 100% (n=18 centres), sperm banking 100% (n=17; one centre was excluded due to the age range of their patients), testicular tissue storage 83% (n=15), mature oocyte collection 35% (n=6; one centre was excluded due to the age range of their patients). All centres with knowledge of their funding source reported sperm cryopreservation was NHS funded. Only 60% (n=9) centres reported the same for mature oocyte storage. Of the centres aware of their funding source, half reported that ovarian and testicular tissue storage was funded by charitable sources; this increased in England compared with the rest of the UK. CONCLUSIONS Inequality exists in provision of fertility preservation for children with cancer across the UK. There is lack of formalised government funding to support international guidelines, with resultant geographical variation in care. Centralised funding of fertility preservation for children and young adults is needed alongside establishment of a national advisory panel to support all PTCs.
Collapse
Affiliation(s)
- Hannah L Newton
- University of Leeds Leeds Institute of Cardiovascular and Metabolic Medicine, Leeds, UK.,Obstetrics & Gynaecology, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Helen M Picton
- University of Leeds Leeds Institute of Cardiovascular and Metabolic Medicine, Leeds, UK
| | - Amanda Jane Friend
- Leeds Institute of Medical Research, University of Leeds, Leeds, UK.,Paediatric and Adolescent Oncology, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | | | - Mark Brougham
- Department of Haematology and Oncology, Royal Hospital for Sick Children, Edinburgh, UK
| | - Rachel Cox
- Paediatric Oncology, Bristol Royal Hospital for Children, Bristol, UK
| | | | | | - Sheila Lane
- Paediatric Oncology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Rod Thomas Mitchell
- MRC Centrre for Reproductive Health, University of Edinburgh, Edinburgh, UK.,Department of Diabetes and Endocrinology, Royal Hospital for Sick Children, Edinburgh, UK
| | - Roderick Skinner
- Paediatric Haematology, Great North Children's Hospital, Newcastle upon Tyne, UK.,Translational and Clinical Research Institute, University of Newcastle upon Tyne, Newcastle upon Tyne, UK
| | - W Hamish Wallace
- The University of Edinburgh MRC Centre for Reproductive Health, Edinburgh, UK.,Royal Hospital for Sick Children, Edinburgh, UK
| | - Daniel Yeomanson
- Paediatric Oncology, Sheffield Children's Hospital, Sheffield, UK
| | - Adam W Glaser
- Leeds Institute of Medical Research, University of Leeds, Leeds, UK .,Paediatric and Adolescent Oncology, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | | |
Collapse
|
48
|
Jiang M, Wang J, Yu R, Hu R, Li J. A narrative review on the research progress of gonadal function protection in children with cancer. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:374. [PMID: 35434006 PMCID: PMC9011244 DOI: 10.21037/atm-22-681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 03/04/2022] [Indexed: 11/06/2022]
Abstract
Background and Objective The global incidence of malignant tumors in children (0-14 years) and adolescents (15-19 years) ranges between 95 per 1 million and 255 per 1 million, which seriously affects the survival of patients. In the past 30 years, with the application of comprehensive treatments (including surgery, chemotherapy, radiotherapy, and bone marrow transplantation), great progress has been made in the treatment of malignant tumors in children and adolescents. The 5-year survival rate now exceeds 80%, and most patients can smoothly enter adolescence or the reproductive period. However, due to the particular age of patients with malignant tumors in children and adolescents, treatment may cause abnormal growth of the patient's height, bones, and some vital organs (such as the pituitary gland and reproductive organs). Treatment may also cause abnormal secretion of growth hormones, thyroid hormones, and sex hormones. These complications seriously affect the quality of life of tumor patients. In the past ten years, countries have established long-term follow-up specifications for children with tumors. These programs have found that, in adulthood, 67% to 75% of children who survived having tumors have at least one treatment-related complication. Among patients receiving chemotherapy, gonadal dysfunction is the most common related endocrine dysfunction. Methods This paper reviews the literature on fertility protection services for cancer patients in foreign countries was conducted to provide a reference for developing gonadal protection services for cancer patients and for establishing consensus or guidelines on gonadal protection in China. Key Content and Findings In the treatment of childhood cancer, the assistance of reproductive technology can effectively reduce the occurrence of complications from treatment. Conclusions Therefore, minimizing the effects of radiotherapy and chemotherapy on the growth and endocrine of children and adolescents while treating tumors is a new challenge for oncologists.
Collapse
Affiliation(s)
- Mingyan Jiang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Jialing Wang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Ruixin Yu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Ruolan Hu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Jinrong Li
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| |
Collapse
|
49
|
Di Mattei VE, Perego G, Taranto P, Rancoita PMV, Maglione M, Notarianni L, Mangili G, Bergamini A, Cioffi R, Papaleo E, Candiani M. Factors Associated With a High Motivation to Undergo Fertility Preservation in Female Cancer Patients. Front Psychol 2022; 12:782073. [PMID: 34975672 PMCID: PMC8716366 DOI: 10.3389/fpsyg.2021.782073] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 11/30/2021] [Indexed: 12/27/2022] Open
Abstract
Objective: Fertility loss due to cancer treatment can be a devastating experience for women and the couple. Undergoing fertility preservation can be a complex decision from both a medical and emotional point of view. The aim of the present study was to evaluate which socio-demographic and psychological factors predict a high motivation to undergo fertility preservation. Methods: Fifty-eight female cancer patients who accessed an Oncofertility Unit completed: a questionnaire to collect socio-demographic characteristics and the level of motivation, the Beck-Depression Inventory-II, the State-Trait Anxiety Inventory-Y, and the Fertility Problem Inventory. Results: Almost half of the sample (44.8%) declared a high motivation. At multiple logistic regression analysis only the “Need for parenthood” subscale of the FPI predicted a high motivation. We alternatively evaluated as possible predictor the construct “Representations about the importance of parenthood” (i.e., the sum of the “Need for Parenthood” and “Rejection of childfree lifestyle” subscales) in place of the two separate subscales. At multiple logistic regression analysis, only this variable predicted a high motivation to undergo fertility preservation. Conclusion: The most important predictor of a high motivation to undergo fertility preservation is the individual desire for parenthood. This implies that, regardless of socio-demographic characteristics, any woman of childbearing age should receive an appropriate counseling about fertility preservation.
Collapse
Affiliation(s)
- Valentina Elisabetta Di Mattei
- School of Psychology, Vita-Salute San Raffaele University, Milan, Italy.,Clinical and Health Psychology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Gaia Perego
- Department of Psychology, University of Milano-Bicocca, Milan, Italy
| | - Paola Taranto
- Clinical and Health Psychology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Paola M V Rancoita
- University Centre for Statistics in the Biomedical Sciences (CUSSB), Vita-Salute San Raffaele University, Milan, Italy.,School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
| | - Mariangela Maglione
- Clinical and Health Psychology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Lisa Notarianni
- Clinical and Health Psychology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giorgia Mangili
- Obstetrics and Gynecology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Alice Bergamini
- Obstetrics and Gynecology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Raffaella Cioffi
- Obstetrics and Gynecology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Enrico Papaleo
- Obstetrics and Gynecology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Massimo Candiani
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy.,Obstetrics and Gynecology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
50
|
Oktay K, Marin L, Bedoschi G, Pacheco F, Sugishita Y, Kawahara T, Taylan E, Acosta C, Bang H. Ovarian transplantation with robotic surgery and a neovascularizing human extracellular matrix scaffold: a case series in comparison to meta-analytic data. Fertil Steril 2022; 117:181-192. [PMID: 34801235 PMCID: PMC8863399 DOI: 10.1016/j.fertnstert.2021.08.034] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 08/17/2021] [Accepted: 08/19/2021] [Indexed: 01/03/2023]
Abstract
OBJECTIVE To report our experience with robot-assisted (RA) autologous cryopreserved ovarian tissue transplantation (ACOTT) with the use of a neovascularizing extracellular matrix scaffold. DESIGN Case series with meta-analytic update. SETTING Academic. PATIENT(S) Seven recipients of RA-ACOTT. INTERVENTION(S) Before or shortly after initiating chemotherapy, ovarian tissue was cryopreserved from 7 women, who then underwent RA-ACOTT 9.9 ± 1.8 years (range, 7-12 years) later. Perioperatively, they received transdermal estrogen and low-dose aspirin to enhance graft vascularization. Ovarian cortical pieces were thawed and sutured on an extracellular matrix scaffold, which was then robotically anastomosed to the bivalved remaining ovary in 6 cases and retroperitoneally (heterotopic) to the lower abdomen in 1 case. MAIN OUTCOME MEASURE(S) Ovarian function return, the number of oocytes/embryos, aneuploidy %, live births, and neonatal outcomes were recorded. Graft longevity was compared with the mean from the meta-analytic data. RESULT(S) Ovarian function returned 13.9 ± 2.7 weeks (11-16.2 weeks) after ACOTT, and oocytes were retrieved in all cases with 12.3 ± 6.9 embryos generated. In contrast to orthotopic, the heterotopic ACOTT demonstrated low embryo quality and an 80% aneuploidy rate. A recipient did not attempt to conceive and 2 needed a surrogate, whereas 4 of 4 delivered 6 healthy children, compared with 115 of 460 (25% pregnancy rate) from the meta-analytic data (n = 79). The mean graft longevity (43.2 ± 23.6/47.4 ± 22.8 months with/without sensitivity analysis) trended longer than the meta-analytic mean (29.4 ± 22.7), even after matching age at cryopreservation. CONCLUSION(S) In this series, RA-ACOTT resulted in extended graft longevity, with ovarian functions restored in all cases, even when the tissues were cryopreserved after chemotherapy exposure.
Collapse
Affiliation(s)
- Kutluk Oktay
- Laboratory of Molecular Reproduction and Fertility Preservation, Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT 06510, USA,Innovation Institute for Fertility Preservation, New York, NY 10028, USA
| | - Loris Marin
- Laboratory of Molecular Reproduction and Fertility Preservation, Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT 06510, USA,Department of Women’s and Children’s Health, University of Padua, Padua, PD 35100, Italy
| | - Giuliano Bedoschi
- Laboratory of Molecular Reproduction and Fertility Preservation, Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT 06510, USA,Division of Reproductive Medicine, Department of Gynecology & Obstetrics, Faculty of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP 14049-900, Brazil
| | - Fernanda Pacheco
- Innovation Institute for Fertility Preservation, New York, NY 10028, USA,Classiclínica, Porto Alegre, Rio Grande do Sul, 90000-000, Brazil
| | - Yodo Sugishita
- Laboratory of Molecular Reproduction and Fertility Preservation, Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT 06510, USA,St Marianna University, Yokohama, Japan
| | - Tai Kawahara
- Laboratory of Molecular Reproduction and Fertility Preservation, Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT 06510, USA,St Marianna University, Yokohama, Japan
| | - Enes Taylan
- Laboratory of Molecular Reproduction and Fertility Preservation, Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Carlo Acosta
- Innovation Institute for Fertility Preservation, New York, NY 10028, USA
| | - Heejung Bang
- Division of Biostatistics, Department of Public Health Sciences, University of California, Davis, CA 95616, USA
| |
Collapse
|