26
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Spermatogonial quantity in human prepubertal testicular tissue collected for fertility preservation prior to potentially sterilizing therapy

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          STUDY QUESTION

          Does chemotherapy exposure (with or without alkylating agents) or primary diagnosis affect spermatogonial quantity in human prepubertal testicular tissue?

          SUMMARY ANSWER

          Spermatogonial quantity is significantly reduced in testes of prepubertal boys treated with alkylating agent therapies or with hydroxyurea for sickle cell disease.

          WHAT IS KNOWN ALREADY

          Cryopreservation of spermatogonial stem cells, followed by transplantation into the testis after treatment, is a proposed clinical option for fertility restoration in children. The key clinical consideration behind this approach is a sufficient quantity of healthy cryopreserved spermatogonia. However, since most boys with malignancies start therapy with agents that are not potentially sterilizing, they will have already received some chemotherapy before testicular tissue cryopreservation is considered.

          STUDY DESIGN, SIZE, DURATION

          We examined histological sections of prepubertal testicular tissue to elucidate whether chemotherapy exposure or primary diagnosis affects spermatogonial quantity. Quantity of spermatogonia per transverse tubular cross-section (S/T) was assessed in relation to treatment characteristics and normative reference values in histological sections of paraffin embedded testicular tissue samples collected from 32 consecutive boy patients (aged 6.3 ± 3.8 [mean ± SD] years) between 2014 and 2017, as part of the NORDFERTIL study, and in 14 control samples (from boys aged 5.6 ± 5.0 [mean ± SD] years) from an internal biobank.

          PARTICIPANTS/MATERIALS, SETTING, METHODS

          Prepubertal boys in Sweden, Finland and Iceland who were facing treatments associated with a very high risk of infertility, were offered the experimental procedure of testicular cryopreservation. Exclusion criteria were testicular volumes >10 ml and high bleeding or infection risk. There were 18 patients with a diagnosis of malignancy and 14 patients a non-malignant diagnosis. While 20 patients had the testicular biopsy performed 1–45 days after chemotherapy, 12 patients had not received any chemotherapy. In addition, 14 testicular tissue samples of patients with no reported testicular pathology, obtained from the internal biobank of the Department of Pathology at Karolinska University Hospital, were included as control samples in addition to reference values obtained from a recently published meta-analysis. The quantity of spermatogonia was assessed by both morphological and immunohistochemical analysis.

          MAIN RESULTS AND THE ROLE OF CHANCE

          The main finding was a significant reduction in spermatogonial cell counts in boys treated with alkylating agents or with hydroxyurea for sickle cell disease. The mean S/T values in boys exposed to alkylating agents (0.2 ± 0.3, n = 6) or in boys with sickle cell disease and exposed to hydroxyurea (0.3 ± 0.6, n = 6) were significantly lower ( P = 0.003 and P = 0.008, respectively) than in a group exposed to non-alkylating agents or in biobank control samples (1.7 ± 1.0, n = 8 and 4.1 ± 4.6, n = 14, respectively). The mean S/T values of the testicular tissue samples included in the biobank control group and the patient group exposed to non-alkylating agents were within recently published normative reference values.

          LIMITATIONS, REASONS FOR CAUTION

          Normal testicular tissue samples included in this study were obtained from the internal biobank of Karolinska University Hospital. Samples were considered normal and included in the study if no testicular pathology was reported in the analysed samples. However, detailed information regarding previous medical treatments and testicular volumes of patients included in this biobank were not available.

          WIDER IMPLICATIONS OF THE FINDINGS

          This study summarizes, for the first time, spermatogonial quantity in a prepubertal patient cohort just before and after potentially sterilizing treatments. Boys facing cancer and cytotoxic therapies are regarded as the major group who will benefit from novel fertility preservation techniques. There are no previous reports correlating spermatogonial quantity to cumulative exposure to alkylating agents and anthracyclines (non-alkylating agents) and no information about the timing of cytotoxic exposures among this particular patient cohort. For prepubertal boys in whom fertility preservation is indicated, testicular tissue should be obtained before initiation of chemotherapy with alkylating agents, whilst for those with sickle cell disease and treated with hydroxyurea, this approach to fertility preservation may not be feasible.

          STUDY FUNDING/COMPETING INTEREST(S)

          This study was supported by grants from The Swedish Childhood Cancer Foundation (PR2016-0124; TJ2016-0093; PR2015-0073, TJ2015-0046) (J.-B.S. and K.J.), the Jane and Dan Olssons Foundation (2016-33) (J.-B.S.), the Finnish Cancer Society (K.J.), the Foundation for Paediatric Research (J.-B.S.), Kronprinsessan Lovisas Förening För Barnasjukvård/ Stiftelsen Axel Tielmans Minnesfond, Samariten Foundation (J.-B.S.), the Väre Foundation for Paediatric Cancer Research (K.J.) and the Swedish Research Council (2012-6352) (O.S.). R.T.M. was supported by a Wellcome Trust Fellowship (09822). J.P.A.-L. and M.K. were supported by the ITN Marie Curie program ‘Growsperm’ (EU-FP7-PEOPLE-2013-ITN 603568). The authors declare no conflicts of interest.

          TRIAL REGISTRATION NUMBER

          N/A.

          Related collections

          Most cited references22

          • Record: found
          • Abstract: found
          • Article: not found

          A European perspective on testicular tissue cryopreservation for fertility preservation in prepubertal and adolescent boys.

          What clinical practices, patient management strategies and experimental methods are currently being used to preserve and restore the fertility of prepubertal boys and adolescent males?
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Cancer treatment and gonadal function: experimental and established strategies for fertility preservation in children and young adults.

            Preservation of gonadal function is an important priority for the long-term health of cancer survivors of both sexes and all ages at treatment. Loss of opportunity for fertility is a prime concern in both male and female cancer survivors, but endocrine effects of gonadal damage are likewise central to long-term health and wellbeing. Some fertility preservation techniques, such as semen and embryo cryopreservation, are established and successful in adults, and development of oocyte vitrification has greatly improved the potential to cryopreserve unfertilised oocytes. Despite being recommended for all pubertal male patients, sperm banking is not universally practised in paediatric oncology centres, and very few adolescent-friendly facilities exist. All approaches to fertility preservation have specific challenges in children and teenagers, including ethical, practical, and scientific issues. For young women, cryopreservation of ovarian cortical tissue with later replacement has resulted in at least 40 livebirths, but is still regarded as experimental in most countries. For prepubertal boys, testicular biopsy cryopreservation is offered in some centres, but how that tissue might be used in the future is unclear, and so far no evidence suggests that fertility can be restored. For both sexes, these approaches involve an invasive procedure and have an uncertain risk of tissue contamination in haematological and other malignancies. Decision making for all these approaches needs assessment of the individual's risk of fertility loss, and is made at a time of emotional distress. Development of this specialty needs better provision of information for patients and their medical teams, and improvements in service provision, to match technical and scientific advances.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Testicular organoid generation by a novel in vitro three-layer gradient system.

              A system that models the testicular microenvironment and spermatogonial stem-cell (SSC) niche in vitro has not been produced yet. Here, we developed and characterized a novel three-dimensional multilayer model, the Three-Layer Gradient System (3-LGS), which permits the generation of rat testicular organoids with a functional blood-testis barrier (BTB) and germ cell establishment and proliferation. The model is unique as regards the formation of cellular organizations that more closely represent the in vivo germ-to-somatic cell associations in vitro. Moreover, we also verified the roles of retinoic acid (RA), IL-1α, TNFα and RA inhibitors in germ cell maintenance and BTB organization in vitro. Treatment with RA was beneficial for germ cell maintenance, while IL-1α and TNFα were observed to impair the formation of testicular organoids and germ cell maintenance. Taking in account our characterization and validation results, we propose the 3-LGS as a new platform to investigate the SSC niche in vitro and to search for novel unknown factors involved in germ cell proliferation and differentiation. Moreover, we suggest that this model can be used in other scientific fields to study organogenesis and development by the generation of organoids.
                Bookmark

                Author and article information

                Journal
                Hum Reprod
                Hum. Reprod
                humrep
                Human Reproduction (Oxford, England)
                Oxford University Press
                0268-1161
                1460-2350
                September 2018
                25 July 2018
                25 July 2018
                : 33
                : 9
                : 1677-1683
                Affiliations
                [1 ]NORDFERTIL Research Lab Stockholm, Department of Women's and Children's Health, Karolinska Institutet and University Hospital, Stockholm, Sweden
                [2 ]Pediatric Endocrinology Unit, Department of Women’s and Children’s Health, Karolinska Institutet and University Hospital, Stockholm, Sweden
                [3 ]Sultan Qaboos University, College of Medicine and Health Sciences, Muscat, Oman
                [4 ]Reproductive Medicine, Department of Obstetrics and Gynaecology, Karolinska University Hospital, Stockholm, Sweden
                [5 ]Department of Medicine, Karolinska Institutet, Stockholm, Sweden
                [6 ]Clinic and University, Children's Medical Center, Landspítali University Hospital, Reykjavik, Iceland
                [7 ]Faculty of Medicine, University of Iceland, Reykjavik, Iceland
                [8 ]Department of Paediatric Oncology and Haematology, Clinical Sciences, Lund University, Lund, Sweden
                [9 ]Division of Paediatrics, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
                [10 ]Pediatric Blood Disorders, Immunodeficiency and Stem Cell Transplantation, Astrid Lindgren Children’s Hospital, Karolinska University Hospital, Stockholm, Sweden
                [11 ]Clinical Sciences, Paediatrics, Umeå University, Umeå, Sweden
                [12 ]Department of Paediatric Oncology, The Queen Silvia Children’s Hospital, Gothenburg, Sweden
                [13 ]Department of Paediatrics, Faculty of Health Sciences, Linköping University, Linköping, Sweden
                [14 ]Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping, Sweden
                [15 ]Division of Urology, Institution for Clinical Science Intervention and Technology, Karolinska Institutet, Huddinge, Stockholm, Sweden
                [16 ]MRC Centre for Reproductive Health, The Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, UK
                [17 ]The Edinburgh Royal Hospital for Sick Children, Edinburgh, UK
                [18 ]Department of Women’s and Children’s Health, Paediatric Oncology Unit, Karolinska Institutet, Stockholm, Sweden
                [19 ]University Hospital, Stockholm, Sweden
                [20 ]Division of Haematology-Oncology and Stem Cell Transplantation, Children´s Hospital, University of Helsinki, Helsinki University Central Hospital, Helsinki, Finland
                [21 ]Department of Development and Regeneration, Organ System Cluster, Group of Biomedical Sciences, KU Leuven, Herestraat 49, Leuven, Belgium
                Author notes
                Correspondence address. Department of Women’s and Children’s Health, NORDFERTIL Research Lab Stockholm, Paediatric Endocrinology Unit, Visionsgatan 4, J9:30, Karolinska Institutet and University Hospital, SE-171 64 Solna, Sweden. Tel: +46-8-517-72507; E-mail: jan-bernd.stukenborg@ 123456ki.se

                C. Petersen and K. Jahnukainen contributed equally to this work.

                Author information
                http://orcid.org/0000-0002-2839-1870
                http://orcid.org/0000-0003-4650-3765
                Article
                dey240
                10.1093/humrep/dey240
                6112575
                30052981
                9a32c3d9-0b59-4874-afde-f094c1370965
                © The Author(s) 2018. Published by Oxford University Press on behalf of the European Society of Human Reproduction and Embryology.

                This is an Open Access article distributed under the terms of the Creative Commons Attribution License ( http://creativecommons.org/licenses/by/4.0/), which permits unrestricted reuse, distribution, and reproduction in any medium, provided the original work is properly cited.

                History
                : 01 June 2018
                : 15 June 2018
                Page count
                Pages: 11
                Funding
                Funded by: Swedish Childhood Cancer Foundation
                Award ID: PR2016-0124
                Award ID: TJ2016-0093
                Award ID: PR2015-0073
                Award ID: TJ2015-0046
                Funded by: Jane and Dan Olssons Foundation
                Award ID: 2016-33
                Funded by: Finnish Cancer Society
                Funded by: Foundation for Paediatric Research
                Funded by: Kronprinsessan Lovisas Förening För Barnasjukvård/ Stiftelsen Axel Tielmans Minnesfond, Samariten Foundation
                Funded by: Väre Foundation for Paediatric Cancer Research
                Funded by: Swedish Research Council
                Award ID: 2012-6352
                Funded by: RTM
                Funded by: Wellcome Trust Fellowship
                Award ID: 09822
                Funded by: ITN Marie Curie program ‘Growsperm’
                Award ID: EU-FP7-PEOPLE-2013-ITN 603568
                Categories
                Original Article
                Infertility

                Human biology
                fertility preservation,childhood cancer,sickle cell disease,alkylating agents,spermatogonial quantity

                Comments

                Comment on this article