Development of Ovarian Tissue Autograft to Restore Ovarian Function: Protocol for a French Multicenter Cohort Study

Jean-Baptiste Pretalli, Sophie Frontczak Franck, Lionel Pazart, Christophe Roux, Clotilde Amiot, DATOR Group, Frédérique Dijoud, Germain Agnani, Pierre-Simon Rorhlich, Christophe Piguet, Brigitte Mignot, Eric Deconink, Christophe Borg, Francine Arbez Gindre, Paul Barrière, Frédérique Carre Pigeon, Blandine Courbière, Christine Decanter, Marie-Catherine Diligent, Olivier Graesslin, Aurore Guennifey, Sylviane Hennebicq, Claude Hocke, Pascale Hoffmann, Isabelle Koscinski, Florence Leperlier, Brigitte Leroy, Loïc Marpeau, Catherine Metzler Guillemain, Aline Papaxanthos, Jean Parinaud, Jean-Christophe Pech, Anne Perdrix, Olivier Pirrello, Marius Teletin, Pascal Piver, Benoît Resch, Nathalie Rives, Jacques Villière, Denis Vinatier, Annie Zaccabri, Roger Leandri, Carole Fajau Prevot, Florence Eustache, Michael Grynberg, Rajeev Ramanah, Florence Brugnon, Anne-Sophie Gremeau, Elodie Adda Herzog, Valérie Scarabin Carre, Nelly Achour Frydman, Pascale Lagre, Beatrice Delepine, Ghassan Harika, Rachel Levy, Nathalie Chabbert Buffet, Marcos Ballester, Cécile Mezan de Malartic, Bénédicte Paillusson, Florence Boitrelle, Jean-Baptiste Pretalli, Sophie Frontczak Franck, Lionel Pazart, Christophe Roux, Clotilde Amiot, DATOR Group, Frédérique Dijoud, Germain Agnani, Pierre-Simon Rorhlich, Christophe Piguet, Brigitte Mignot, Eric Deconink, Christophe Borg, Francine Arbez Gindre, Paul Barrière, Frédérique Carre Pigeon, Blandine Courbière, Christine Decanter, Marie-Catherine Diligent, Olivier Graesslin, Aurore Guennifey, Sylviane Hennebicq, Claude Hocke, Pascale Hoffmann, Isabelle Koscinski, Florence Leperlier, Brigitte Leroy, Loïc Marpeau, Catherine Metzler Guillemain, Aline Papaxanthos, Jean Parinaud, Jean-Christophe Pech, Anne Perdrix, Olivier Pirrello, Marius Teletin, Pascal Piver, Benoît Resch, Nathalie Rives, Jacques Villière, Denis Vinatier, Annie Zaccabri, Roger Leandri, Carole Fajau Prevot, Florence Eustache, Michael Grynberg, Rajeev Ramanah, Florence Brugnon, Anne-Sophie Gremeau, Elodie Adda Herzog, Valérie Scarabin Carre, Nelly Achour Frydman, Pascale Lagre, Beatrice Delepine, Ghassan Harika, Rachel Levy, Nathalie Chabbert Buffet, Marcos Ballester, Cécile Mezan de Malartic, Bénédicte Paillusson, Florence Boitrelle

Abstract

Background: Sterility is a major late effect of radiotherapy and chemotherapy treatments. Iatrogenic sterility is often permanent and greatly impacts long-term quality of life. Ovarian tissue cryopreservation (OTC) performed before gonadotoxic treatments with subsequent autograft is a method of fertility preservation available for girls and women. Its application in prepubertal girls is of particular value as it is the only possible approach in this patient group. In addition, it does not require a delay in cancer therapy and no ovarian stimulation is needed.

Objective: The primary aim of this protocol is to help increase the implementation of ovarian tissue autografting in France. Knowledge is still lacking regarding the efficacy of ovarian transplantation in restoring ovarian function and regarding the safety of this procedure, especially the risk of cancer cell reseeding in certain types of cancer. A secondary aim of this study is to generate data to improve our understanding of these two essential aspects.

Methods: The DATOR (Development of Ovarian Tissue Autograft in Order to Restore Ovarian Function) study is ongoing in 17 university hospitals. The DATOR protocol includes the autograft of ovarian cortex fragments. Candidates are identified from an observational prospective cohort (called the Prospective Cohort of Patients Candidates for Ovarian Tissue Autograft [PERIDATOR]) of patients who have undergone OTC. Enrollment in the study is initiated at the patient's request and must be validated by the center's multidisciplinary team and by the study steering committee. The DATOR study begins with a total medical checkup. Ovarian tissue qualification and residual disease detection, if required, are performed.

Results: The study is ongoing. Currently, 38 patients have provided informed consent and have been entered into the DATOR study. Graft has been performed for 34 of these patients. An interim analysis was conducted on the first 25 patients for whom the period of at least 1 year posttransplantation was achieved. Out of these 25 patients, 11 women succeeded in becoming pregnant (pregnancy rate=44% [11/25]; delivery rate=40% [10/25]). Among these, 6 women conceived twice, and 1 pregnancy led to a miscarriage.

Conclusions: Our preliminary analysis appears to be coherent with the accumulating body of evidence indicating the potential utility of ovarian tissue autograft for patients with premature ovarian failure. All these elements justify the pursuit of our study.

Trial registration: ClinicalTrials.gov NCT02846064; https://ichgcp.net/clinical-trials-registry/NCT02846064.

International registered report identifier (irrid): DERR1-10.2196/12944.

Keywords: cohort study; cryopreservation; fertility preservation; live birth rate; ovarian tissue; pregnancy rate.

Conflict of interest statement

Conflicts of Interest: None declared.

©Jean-Baptiste Pretalli, Sophie Frontczak Franck, Lionel Pazart, Christophe Roux, Clotilde Amiot, DATOR Group. Originally published in JMIR Research Protocols (http://www.researchprotocols.org), 30.09.2019.

Figures

https://www.ncbi.nlm.nih.gov/pmc/articles/instance/6802486/bin/resprot_v8i9e12944_fig1.jpg
Study flowchart.
https://www.ncbi.nlm.nih.gov/pmc/articles/instance/6802486/bin/resprot_v8i9e12944_fig2.jpg
Flowchart of the preliminary analysis. DATOR: Development of Ovarian Tissue Autograft in Order to Restore Ovarian Function.
https://www.ncbi.nlm.nih.gov/pmc/articles/instance/6802486/bin/resprot_v8i9e12944_fig3.jpg
Box plots of monthly follicle-stimulating hormone levels during the year after autograft for the no pregnancy (blue) and the pregnancy group (red). For each group, the curves represent the monthly follicle-stimulating hormone means for the no pregnancy (blue curve) and for the pregnancy group (red curve), respectively. FSH: follicle-stimulating hormone.

References

    1. Croucher C, Whelan JS, Møller H, Davies EA. Trends in the incidence and survival of cancer in teenagers and young adults: regional analysis for South East England 1960-2002. Clin Oncol (R Coll Radiol) 2009 Jun;21(5):417–24. doi: 10.1016/j.clon.2009.02.006.
    1. Aben KK, van Gaal C, van Gils NA, van der Graaf WT, Zielhuis GA. Cancer in adolescents and young adults (15-29 years): a population-based study in the Netherlands 1989-2009. Acta Oncol. 2012 Sep;51(7):922–33. doi: 10.3109/0284186X.2012.705891.
    1. Cancer Research UK. [2018-10-24]. Young People's Cancers Statistics .
    1. Desandes E, Lacour B, Sommelet D, Danzon A, Delafosse P, Grosclaude P, Mace-Lesech J, Maarouf N, Marr A, Raverdy-Bourdon N, Tretarre B, Velten M, Brugieres L. Cancer survival among adolescents in France. Eur J Cancer. 2006 Feb;42(3):403–9. doi: 10.1016/j.ejca.2005.07.035.
    1. Jemal A, Siegel R, Ward E, Hao Y, Xu J, Murray T, Thun MJ. Cancer statistics, 2008. CA Cancer J Clin. 2008;58(2):71–96. doi: 10.3322/CA.2007.0010. doi: 10.3322/CA.2007.0010.
    1. Ries LA, Melbert D, Krapcho M, Stinchcomb DG, Howlader N, Horner MJ, Mariotto A, Miller BA, Feuer EJ, Altekruse SF, Lewis DR, Clegg L, Eisner MP, Reichman M, Edwards BK. SEER - National Cancer Institute. 2008. [2018-11-13]. SEER Cancer Statistics Review, 1975-2005
    1. de Angelis R, Sant M, Coleman MP, Francisci S, Baili P, Pierannunzio D, Trama A, Visser O, Brenner H, Ardanaz E, Bielska-Lasota M, Engholm G, Nennecke A, Siesling S, Berrino F, Capocaccia R. Cancer survival in Europe 1999-2007 by country and age: results of EUROCARE--5-a population-based study. Lancet Oncol. 2014 Jan;15(1):23–34. doi: 10.1016/S1470-2045(13)70546-1.
    1. Levine JM, Kelvin JF, Quinn GP, Gracia CR. Infertility in reproductive-age female cancer survivors. Cancer. 2015 May 15;121(10):1532–9. doi: 10.1002/cncr.29181. doi: 10.1002/cncr.29181.
    1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA Cancer J Clin. 2019 Jan;69(1):7–34. doi: 10.3322/caac.21551. doi: 10.3322/caac.21551.
    1. Meirow D, Nugent D. The effects of radiotherapy and chemotherapy on female reproduction. Hum Reprod Update. 2001;7(6):535–43. doi: 10.1093/humupd/7.6.535.
    1. Larsen EC, Müller J, Schmiegelow K, Rechnitzer C, Andersen AN. Reduced ovarian function in long-term survivors of radiation- and chemotherapy-treated childhood cancer. J Clin Endocrinol Metab. 2003 Nov;88(11):5307–14. doi: 10.1210/jc.2003-030352.
    1. Donnez J, Martinez-Madrid B, Jadoul P, van Langendonckt A, Demylle D, Dolmans MM. Ovarian tissue cryopreservation and transplantation: a review. Hum Reprod Update. 2006;12(5):519–35. doi: 10.1093/humupd/dml032.
    1. Lie Fong S, Laven JS, Hakvoort-Cammel FG, Schipper I, Visser JA, Themmen AP, de Jong FH, van den Heuvel-Eibrink MM. Assessment of ovarian reserve in adult childhood cancer survivors using anti-Müllerian hormone. Hum Reprod. 2009 Apr;24(4):982–90. doi: 10.1093/humrep/den487.
    1. Thomas-Teinturier C, Allodji RS, Svetlova E, Frey M, Oberlin O, Millischer A, Epelboin S, Decanter C, Pacquement H, Tabone M, Sudour-Bonnange H, Baruchel A, Lahlou N, de Vathaire F. Ovarian reserve after treatment with alkylating agents during childhood. Hum Reprod. 2015 Jun;30(6):1437–46. doi: 10.1093/humrep/dev060.
    1. Donnez J, Bassil S. Indications for cryopreservation of ovarian tissue. Hum Reprod Update. 1998;4(3):248–59. doi: 10.1093/humupd/4.3.248.
    1. Charpentier AM, Chong AL, Gingras-Hill G, Ahmed S, Cigsar C, Gupta AA, Greenblatt E, Hodgson DC. Anti-Müllerian hormone screening to assess ovarian reserve among female survivors of childhood cancer. J Cancer Surviv. 2014 Dec;8(4):548–54. doi: 10.1007/s11764-014-0364-4.
    1. Schover LR. Sexuality and fertility after cancer. Hematology Am Soc Hematol Educ Program. 2005;(1):523–7. doi: 10.1182/asheducation-2005.1.523.
    1. Molina JR, Barton DL, Loprinzi CL. Chemotherapy-induced ovarian failure: manifestations and management. Drug Saf. 2005;28(5):401–16. doi: 10.2165/00002018-200528050-00004.
    1. Gracia CR, Sammel MD, Freeman E, Prewitt M, Carlson C, Ray A, Vance A, Ginsberg JP. Impact of cancer therapies on ovarian reserve. Fertil Steril. 2012 Jan;97(1):134–40.e1. doi: 10.1016/j.fertnstert.2011.10.040.
    1. Bedoschi G, Navarro PA, Oktay K. Chemotherapy-induced damage to ovary: mechanisms and clinical impact. Future Oncol. 2016 Oct;12(20):2333–44. doi: 10.2217/fon-2016-0176.
    1. Revel A, Laufer N. Protecting female fertility from cancer therapy. Mol Cell Endocrinol. 2002 Feb 22;187(1-2):83–91. doi: 10.1016/s0303-7207(01)00705-5.
    1. Mattle V, Behringer K, Engert A, Wildt L. Female fertility after cytotoxic therapy--protection of ovarian function during chemotherapy of malignant and non-malignant diseases. Eur J Haematol Suppl. 2005 Jul;(66):77–82. doi: 10.1111/j.1600-0609.2005.00459.x.
    1. Behringer K, Wildt L, Mueller H, Mattle V, Ganitis P, van den Hoonaard B, Ott HW, Hofer S, Pluetschow A, Diehl V, Engert A, Borchmann P. No protection of the ovarian follicle pool with the use of GnRH-analogues or oral contraceptives in young women treated with escalated BEACOPP for advanced-stage Hodgkin lymphoma. Final results of a phase II trial from the German Hodgkin study group. Ann Oncol. 2010 Oct;21(10):2052–60. doi: 10.1093/annonc/mdq066.
    1. Blumenfeld Z, Evron A. Preserving fertility when choosing chemotherapy regimens - the role of gonadotropin-releasing hormone agonists. Expert Opin Pharmacother. 2015 May;16(7):1009–20. doi: 10.1517/14656566.2015.1031654.
    1. Lambertini M, Ceppi M, Poggio F, Peccatori FA, Azim HA, Ugolini D, Pronzato P, Loibl S, Moore HC, Partridge AH, Bruzzi P, del Mastro L. Ovarian suppression using luteinizing hormone-releasing hormone agonists during chemotherapy to preserve ovarian function and fertility of breast cancer patients: a meta-analysis of randomized studies. Ann Oncol. 2015 Dec;26(12):2408–19. doi: 10.1093/annonc/mdv374.
    1. Salama M, Isachenko V, Isachenko E, Rahimi G, Mallmann P. Updates in preserving reproductive potential of prepubertal girls with cancer: systematic review. Crit Rev Oncol Hematol. 2016 Jul;103:10–21. doi: 10.1016/j.critrevonc.2016.04.002.
    1. Demeestere I, Brice P, Peccatori FA, Kentos A, Dupuis J, Zachee P, Casasnovas O, van den Neste E, Dechene J, de Maertelaer V, Bron D, Englert Y. No evidence for the benefit of gonadotropin-releasing hormone agonist in preserving ovarian function and fertility in lymphoma survivors treated with chemotherapy: final long-term report of a prospective randomized trial. J Clin Oncol. 2016 Aug 1;34(22):2568–74. doi: 10.1200/JCO.2015.65.8864.
    1. Senra JC, Roque M, Talim MC, Reis FM, Tavares RL. Gonadotropin-releasing hormone agonists for ovarian protection during cancer chemotherapy: systematic review and meta-analysis. Ultrasound Obstet Gynecol. 2018 Jan;51(1):77–86. doi: 10.1002/uog.18934. doi: 10.1002/uog.18934.
    1. Sonigo C, Beau I, Grynberg M, Binart N. AMH prevents primordial ovarian follicle loss and fertility alteration in cyclophosphamide-treated mice. FASEB J. 2019 Jan;33(1):1278–87. doi: 10.1096/fj.201801089R.
    1. Kano M, Sosulski AE, Zhang L, Saatcioglu HD, Wang D, Nagykery N, Sabatini ME, Gao G, Donahoe PK, Pépin D. AMH/MIS as a contraceptive that protects the ovarian reserve during chemotherapy. Proc Natl Acad Sci U S A. 2017 Feb 28;114(9):E1688–97. doi: 10.1073/pnas.1620729114.
    1. Lee HN, Chang EM. Primordial follicle activation as new treatment for primary ovarian insufficiency. Clin Exp Reprod Med. 2019 Jun;46(2):43–9. doi: 10.5653/cerm.2019.46.2.43.
    1. Carter J, Rowland K, Chi D, Brown C, Abu-Rustum N, Castiel M, Barakat R. Gynecologic cancer treatment and the impact of cancer-related infertility. Gynecol Oncol. 2005 Apr;97(1):90–5. doi: 10.1016/j.ygyno.2004.12.019.
    1. Logan S, Perz J, Ussher JM, Peate M, Anazodo A. A systematic review of patient oncofertility support needs in reproductive cancer patients aged 14 to 45 years of age. Psychooncology. 2018 Feb;27(2):401–9. doi: 10.1002/pon.4502.
    1. Logan S, Perz J, Ussher JM, Peate M, Anazodo A. Systematic review of fertility-related psychological distress in cancer patients: informing on an improved model of care. Psychooncology. 2019 Jan;28(1):22–30. doi: 10.1002/pon.4927.
    1. Samuelsson A, Fuchs T, Simonsson B, Björkholm M. Successful pregnancy in a 28-year-old patient autografted for acute lymphoblastic leukemia following myeloablative treatment including total body irradiation. Bone Marrow Transplant. 1993 Dec;12(6):659–60.
    1. Schimmer AD, Quatermain M, Imrie K, Ali V, McCrae J, Stewart AK, Crump M, Derzko C, Keating A. Ovarian function after autologous bone marrow transplantation. J Clin Oncol. 1998 Jul;16(7):2359–63. doi: 10.1200/JCO.1998.16.7.2359.
    1. Logan S, Perz J, Ussher J, Peate M, Anazodo A. Clinician provision of oncofertility support in cancer patients of a reproductive age: a systematic review. Psychooncology. 2018 Mar;27(3):748–56. doi: 10.1002/pon.4518.
    1. Anderson RA, Wallace WH, Baird DT. Ovarian cryopreservation for fertility preservation: indications and outcomes. Reproduction. 2008 Dec;136(6):681–9. doi: 10.1530/REP-08-0097.
    1. Peccatori FA, Azim HA, Orecchia R, Hoekstra HJ, Pavlidis N, Kesic V, Pentheroudakis G, ESMO Guidelines Working Group Cancer, pregnancy and fertility: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2013 Oct;24(Suppl 6):vi160–70. doi: 10.1093/annonc/mdt199.
    1. Köhler TS, Kondapalli LA, Shah A, Chan S, Woodruff TK, Brannigan RE. Results from the survey for preservation of adolescent reproduction (SPARE) study: gender disparity in delivery of fertility preservation message to adolescents with cancer. J Assist Reprod Genet. 2011 Mar;28(3):269–77. doi: 10.1007/s10815-010-9504-6.
    1. Lambertini M, del Mastro L, Pescio MC, Andersen CY, Azim HA, Peccatori FA, Costa M, Revelli A, Salvagno F, Gennari A, Ubaldi FM, la Sala GB, de Stefano C, Wallace WH, Partridge AH, Anserini P. Cancer and fertility preservation: international recommendations from an expert meeting. BMC Med. 2016 Jan 4;14:1. doi: 10.1186/s12916-015-0545-7.
    1. Condorelli M, Lambertini M, del Mastro L, Boccardo F, Demeestere I, Bober SL. Fertility, sexuality and cancer in young adult women. Curr Opin Oncol. 2019 Jul;31(4):259–67. doi: 10.1097/CCO.0000000000000540.
    1. Anazodo A, Laws P, Logan S, Saunders C, Travaglia J, Gerstl B, Bradford N, Cohn R, Birdsall M, Barr R, Suzuki N, Takae S, Marinho R, Xiao S, Qiong-Hua C, Mahajan N, Patil M, Gunasheela D, Smith K, Sender L, Melo C, Almeida-Santos T, Salama M, Appiah L, Su I, Lane S, Woodruff TK, Pacey A, Anderson RA, Shenfield F, Ledger W, Sullivan E. How can we improve oncofertility care for patients? A systematic scoping review of current international practice and models of care. Hum Reprod Update. 2019 Mar 1;25(2):159–79. doi: 10.1093/humupd/dmy038.
    1. Oktay K, Harvey BE, Partridge AH, Quinn GP, Reinecke J, Taylor HS, Wallace WH, Wang ET, Loren AW. Fertility preservation in patients with cancer: ASCO clinical practice guideline update. J Clin Oncol. 2018 Jul 1;36(19):1994–2001. doi: 10.1200/JCO.2018.78.1914.
    1. Winkler-Crepaz K, Böttcher B, Toth B, Wildt L, Hofer-Tollinger S. What is new in 2017? Update on fertility preservation in cancer patients. Minerva Endocrinol. 2017 Dec;42(4):331–9. doi: 10.23736/S0391-1977.17.02633-5.
    1. Anderson RA, Wallace WH, Telfer EE. Ovarian tissue cryopreservation for fertility preservation: clinical and research perspectives. Hum Reprod Open. 2017;2017(1):hox001. doi: 10.1093/hropen/hox001.
    1. Donnez J, Dolmans MM. Preservation of fertility in females with haematological malignancy. Br J Haematol. 2011 Jul;154(2):175–84. doi: 10.1111/j.1365-2141.2011.08723.x.
    1. Poirot C, Martelli H, Lichtblau I, Dhedin N, Abirached F, Faraguet C, Vacher-Lavenu M. [Female fertility preservation before sterilizing treatment: contribution of ovarian tissue cryopreservation] J Gynecol Obstet Biol Reprod (Paris) 2002 Dec;31(8):717–23.
    1. Gosden RG, Baird DT, Wade JC, Webb R. Restoration of fertility to oophorectomized sheep by ovarian autografts stored at -196 degrees C. Hum Reprod. 1994 Apr;9(4):597–603. doi: 10.1093/oxfordjournals.humrep.a138556.
    1. Fauque P, Ben Amor A, Joanne C, Agnani G, Bresson JL, Roux C. Use of trypan blue staining to assess the quality of ovarian cryopreservation. Fertil Steril. 2007 May;87(5):1200–7. doi: 10.1016/j.fertnstert.2006.08.115.
    1. Kagawa N, Silber S, Kuwayama M. Successful vitrification of bovine and human ovarian tissue. Reprod Biomed Online. 2009 Apr;18(4):568–77. doi: 10.1016/S1472-6483(10)60136-8.
    1. Silber S, Kagawa N, Kuwayama M, Gosden R. Duration of fertility after fresh and frozen ovary transplantation. Fertil Steril. 2010 Nov;94(6):2191–6. doi: 10.1016/j.fertnstert.2009.12.073.
    1. Nikiforov D, Russo V, Nardinocchi D, Bernabò N, Mattioli M, Barboni B. Innovative multi-protectoral approach increases survival rate after vitrification of ovarian tissue and isolated follicles with improved results in comparison with conventional method. J Ovarian Res. 2018 Aug 7;11(1):65. doi: 10.1186/s13048-018-0437-5.
    1. Abir R, Nitke S, Ben-Haroush A, Fisch B. In vitro maturation of human primordial ovarian follicles: clinical significance, progress in mammals, and methods for growth evaluation. Histol Histopathol. 2006 Aug;21(8):887–98. doi: 10.14670/HH-21.887.
    1. Smitz JE, Thompson JG, Gilchrist RB. The promise of in vitro maturation in assisted reproduction and fertility preservation. Semin Reprod Med. 2011 Jan;29(1):24–37. doi: 10.1055/s-0030-1268701.
    1. Telfer EE, McLaughlin M. Strategies to support human oocyte development in vitro. Int J Dev Biol. 2012;56(10-12):901–7. doi: 10.1387/ijdb.130001et.
    1. Telfer EE, Zelinski MB. Ovarian follicle culture: advances and challenges for human and nonhuman primates. Fertil Steril. 2013 May;99(6):1523–33. doi: 10.1016/j.fertnstert.2013.03.043.
    1. Zhai J, Yao G, Dong F, Bu Z, Cheng Y, Sato Y, Hu L, Zhang Y, Wang J, Dai S, Li J, Sun J, Hsueh AJ, Kawamura K, Sun Y. In vitro activation of follicles and fresh tissue auto-transplantation in primary ovarian insufficiency patients. J Clin Endocrinol Metab. 2016 Nov;101(11):4405–12. doi: 10.1210/jc.2016-1589.
    1. Khalili MA, Shahedi A, Ashourzadeh S, Nottola SA, Macchiarelli G, Palmerini MG. Vitrification of human immature oocytes before and after in vitro maturation: a review. J Assist Reprod Genet. 2017 Nov;34(11):1413–26. doi: 10.1007/s10815-017-1005-4.
    1. Fasano G, Dechène J, Antonacci R, Biramane J, Vannin AS, van Langendonckt A, Devreker F, Demeestere I. Outcomes of immature oocytes collected from ovarian tissue for cryopreservation in adult and prepubertal patients. Reprod Biomed Online. 2017 Jun;34(6):575–82. doi: 10.1016/j.rbmo.2017.03.007.
    1. Bertoldo MJ, Walters KA, Ledger WL, Gilchrist RB, Mermillod P, Locatelli Y. In-vitro regulation of primordial follicle activation: challenges for fertility preservation strategies. Reprod Biomed Online. 2018 May;36(5):491–9. doi: 10.1016/j.rbmo.2018.01.014.
    1. McLaughlin M, Albertini DF, Wallace WH, Anderson RA, Telfer EE. Metaphase II oocytes from human unilaminar follicles grown in a multi-step culture system. Mol Hum Reprod. 2018 Mar 1;24(3):135–42. doi: 10.1093/molehr/gay002.
    1. Mouloungui E, Zver T, Roux C, Amiot C. A protocol to isolate and qualify purified human preantral follicles in cases of acute leukemia, for future clinical applications. J Ovarian Res. 2018 Jan 5;11(1):4. doi: 10.1186/s13048-017-0376-6.
    1. Chiti MC, Donnez J, Amorim CA, Dolmans MM. From isolation of human ovarian follicles to the artificial ovary: tips and tricks. Minerva Ginecol. 2018 Aug;70(4):444–55. doi: 10.23736/S0026-4784.18.04231-4.
    1. Paulini F, Vilela JM, Chiti MC, Donnez J, Jadoul P, Dolmans MM, Amorim CA. Survival and growth of human preantral follicles after cryopreservation of ovarian tissue, follicle isolation and short-term xenografting. Reprod Biomed Online. 2016 Sep;33(3):425–32. doi: 10.1016/j.rbmo.2016.05.003.
    1. Amorim CA, Shikanov A. The artificial ovary: current status and future perspectives. Future Oncol. 2016 Oct;12(20):2323–32. doi: 10.2217/fon-2016-0202.
    1. Cho E, Kim YY, Noh K, Ku SY. A new possibility in fertility preservation: the artificial ovary. J Tissue Eng Regen Med. 2019 May 6; doi: 10.1002/term.2870. (epub ahead of print)
    1. Jensen AK, Kristensen SG, Macklon KT, Jeppesen JV, Fedder J, Ernst E, Andersen CY. Outcomes of transplantations of cryopreserved ovarian tissue to 41 women in Denmark. Hum Reprod. 2015 Dec;30(12):2838–45. doi: 10.1093/humrep/dev230.
    1. Hudson JN, Stanley NB, Nahata L, Bowman-Curci M, Quinn GP. New promising strategies in oncofertility. Expert Rev Qual Life Cancer Care. 2017;2(2):67–78. doi: 10.1080/23809000.2017.1308808.
    1. Demeestere I, Simon P, Emiliani S, Delbaere A, Englert Y. Orthotopic and heterotopic ovarian tissue transplantation. Hum Reprod Update. 2009;15(6):649–65. doi: 10.1093/humupd/dmp021.
    1. Kondapalli LA. Ovarian tissue cryopreservation and transplantation. In: Gracia C, Woodruff TK, editors. Oncofertility Medical Practice: Clinical Issues and Implementation. New York: Springer; 2012. pp. 63–75.
    1. Soares M, Dolmans MM, Donnez J. Heterotopic ovarian tissue transplantation. In: Suzuki N, Donnez J, editors. Gonadal Tissue Cryopreservation in Fertility Preservation. Tokyo, Japan: Springer; 2016. pp. 105–23.
    1. Donfack NJ, Alves KA, Araújo VR, Cordova A, Figueiredo JR, Smitz J, Rodrigues AP. Expectations and limitations of ovarian tissue transplantation. Zygote. 2017 Aug;25(4):391–403. doi: 10.1017/S0967199417000338.
    1. Oktay K, Karlikaya G. Ovarian function after transplantation of frozen, banked autologous ovarian tissue. N Engl J Med. 2000 Jun 22;342(25):1919. doi: 10.1056/NEJM200006223422516.
    1. Radford JA, Lieberman BA, Brison DR, Smith AR, Critchlow JD, Russell SA, Watson AJ, Clayton JA, Harris M, Gosden RG, Shalet SM. Orthotopic reimplantation of cryopreserved ovarian cortical strips after high-dose chemotherapy for Hodgkin's lymphoma. Lancet. 2001 Apr 14;357(9263):1172–5. doi: 10.1016/s0140-6736(00)04335-x.
    1. Oktay K, Buyuk E, Veeck L, Zaninovic N, Xu K, Takeuchi T, Opsahl M, Rosenwaks Z. Embryo development after heterotopic transplantation of cryopreserved ovarian tissue. Lancet. 2004 Mar 13;363(9412):837–40. doi: 10.1016/S0140-6736(04)15728-0.
    1. Donnez J, Dolmans MM, Demylle D, Jadoul P, Pirard C, Squifflet J, Martinez-Madrid B, van Langendonckt A. Livebirth after orthotopic transplantation of cryopreserved ovarian tissue. Lancet. 2004;364(9443):1405–10. doi: 10.1016/S0140-6736(04)17222-X.
    1. Donnez J, Dolmans MM. Fertility preservation in women. N Engl J Med. 2017 Oct 26;377(17):1657–65. doi: 10.1056/NEJMra1614676.
    1. Jensen AK, Macklon KT, Fedder J, Ernst E, Humaidan P, Andersen CY. 86 successful births and 9 ongoing pregnancies worldwide in women transplanted with frozen-thawed ovarian tissue: focus on birth and perinatal outcome in 40 of these children. J Assist Reprod Genet. 2017 Mar;34(3):325–36. doi: 10.1007/s10815-016-0843-9.
    1. Donnez J, Dolmans MM, Pellicer A, Diaz-Garcia C, Serrano MS, Schmidt KT, Ernst E, Luyckx V, Andersen CY. Restoration of ovarian activity and pregnancy after transplantation of cryopreserved ovarian tissue: a review of 60 cases of reimplantation. Fertil Steril. 2013 May;99(6):1503–13. doi: 10.1016/j.fertnstert.2013.03.030.
    1. Stoop D, Cobo A, Silber S. Fertility preservation for age-related fertility decline. Lancet. 2014 Oct 4;384(9950):1311–9. doi: 10.1016/S0140-6736(14)61261-7.
    1. Donnez J, Dolmans MM. Ovarian cortex transplantation: 60 reported live births brings the success and worldwide expansion of the technique towards routine clinical practice. J Assist Reprod Genet. 2015 Aug;32(8):1167–70. doi: 10.1007/s10815-015-0544-9.
    1. Dittrich R, Hackl J, Lotz L, Hoffmann I, Beckmann MW. Pregnancies and live births after 20 transplantations of cryopreserved ovarian tissue in a single center. Fertil Steril. 2015 Feb;103(2):462–8. doi: 10.1016/j.fertnstert.2014.10.045.
    1. Meirow D, Ra'anani H, Shapira M, Brenghausen M, Chaim SD, Aviel-Ronen S, Amariglio N, Schiff E, Orvieto R, Dor J. Transplantations of frozen-thawed ovarian tissue demonstrate high reproductive performance and the need to revise restrictive criteria. Fertil Steril. 2016 Aug;106(2):467–74. doi: 10.1016/j.fertnstert.2016.04.031.
    1. van der Ven H, Liebenthron J, Beckmann M, Toth B, Korell M, Krüssel J, Frambach T, Kupka M, Hohl MK, Winkler-Crepaz K, Seitz S, Dogan A, Griesinger G, Häberlin F, Henes M, Schwab R, Sütterlin M, von Wolff M, Dittrich R. Ninety-five orthotopic transplantations in 74 women of ovarian tissue after cytotoxic treatment in a fertility preservation network: tissue activity, pregnancy and delivery rates. Hum Reprod. 2016 Sep;31(9):2031–41. doi: 10.1093/humrep/dew165.
    1. Jadoul P, Guilmain A, Squifflet J, Luyckx M, Votino R, Wyns C, Dolmans MM. Efficacy of ovarian tissue cryopreservation for fertility preservation: lessons learned from 545 cases. Hum Reprod. 2017 May 1;32(5):1046–54. doi: 10.1093/humrep/dex040.
    1. Diaz-Garcia C, Domingo J, Garcia-Velasco JA, Herraiz S, Mirabet V, Iniesta I, Cobo A, Remohí J, Pellicer A. Oocyte vitrification versus ovarian cortex transplantation in fertility preservation for adult women undergoing gonadotoxic treatments: a prospective cohort study. Fertil Steril. 2018 Mar;109(3):478–85.e2. doi: 10.1016/j.fertnstert.2017.11.018.
    1. Gellert SE, Pors SE, Kristensen SG, Bay-Bjørn A M, Ernst E, Andersen CY. Transplantation of frozen-thawed ovarian tissue: an update on worldwide activity published in peer-reviewed papers and on the Danish cohort. J Assist Reprod Genet. 2018 Apr;35(4):561–70. doi: 10.1007/s10815-018-1144-2.
    1. Roux C, Amiot C, Agnani G, Aubard Y, Rohrlich PS, Piver P. Live birth after ovarian tissue autograft in a patient with sickle cell disease treated by allogeneic bone marrow transplantation. Fertil Steril. 2010 May 1;93(7):2413.e15–9. doi: 10.1016/j.fertnstert.2009.12.022.
    1. Accueil | Légifrance, Le Service Public De La Diffusion Du Droit. 2017. [2019-08-22]. Arrêté Du 30 Juin 2017 Modifiant L'arrêté Du 11 Avril 2008 Modifié Relatif Aux Règles De Bonnes Pratiques Cliniques Et Biologiques D'assistance Médicale À La Procréation .
    1. ISFP Practice Committee. Kim SS, Donnez J, Barri P, Pellicer A, Patrizio P, Rosenwaks Z, Nagy P, Falcone T, Andersen C, Hovatta O, Wallace H, Meirow D, Gook D, Kim SH, Tzeng CR, Suzuki S, Ishizuka B, Dolmans MM. Recommendations for fertility preservation in patients with lymphoma, leukemia, and breast cancer. J Assist Reprod Genet. 2012 Jun;29(6):465–8. doi: 10.1007/s10815-012-9786-y.
    1. Dolmans MM, Masciangelo R. Risk of transplanting malignant cells in cryopreserved ovarian tissue. Minerva Ginecol. 2018 Aug;70(4):436–43. doi: 10.23736/S0026-4784.18.04233-8.
    1. Meirow D, Hardan I, Dor J, Fridman E, Elizur S, Ra'anani H, Slyusarevsky E, Amariglio N, Schiff E, Rechavi G, Nagler A, Yehuda DB. Searching for evidence of disease and malignant cell contamination in ovarian tissue stored from hematologic cancer patients. Hum Reprod. 2008 May;23(5):1007–13. doi: 10.1093/humrep/den055.
    1. Greve T, Clasen-Linde E, Andersen MT, Andersen MK, Sørensen SD, Rosendahl M, Ralfkiaer E, Andersen CY. Cryopreserved ovarian cortex from patients with leukemia in complete remission contains no apparent viable malignant cells. Blood. 2012 Nov 22;120(22):4311–6. doi: 10.1182/blood-2012-01-403022.
    1. Asadi-Azarbaijani B, Sheikhi M, Nurmio M, Tinkanen H, Juvonen V, Dunkel L, Hovatta O, Oskam IC, Jahnukainen K. Minimal residual disease of leukemia and the quality of cryopreserved human ovarian tissue in vitro. Leuk Lymphoma. 2016;57(3):700–7. doi: 10.3109/10428194.2015.1065980.
    1. Amiot C, Angelot-Delettre F, Zver T, Alvergnas-Vieille M, Saas P, Garnache-Ottou F, Roux C. Minimal residual disease detection of leukemic cells in ovarian cortex by eight-color flow cytometry. Hum Reprod. 2013 Aug;28(8):2157–67. doi: 10.1093/humrep/det126.
    1. Zver T, Alvergnas-Vieille M, Garnache-Ottou F, Roux C, Amiot C. A new method for evaluating the risk of transferring leukemic cells with transplanted cryopreserved ovarian tissue. J Assist Reprod Genet. 2015 Aug;32(8):1263–6. doi: 10.1007/s10815-015-0512-4.
    1. Lotz L, Montag M, van der Ven H, von Wolff M, Mueller A, Hoffmann I, Wachter D, Beckmann MW, Dittrich R. Xenotransplantation of cryopreserved ovarian tissue from patients with ovarian tumors into SCID mice--no evidence of malignant cell contamination. Fertil Steril. 2011 Jun 30;95(8):2612–4.e1. doi: 10.1016/j.fertnstert.2011.05.003.
    1. Meyer LH, Debatin KM. Diversity of human leukemia xenograft mouse models: implications for disease biology. Cancer Res. 2011 Dec 1;71(23):7141–4. doi: 10.1158/0008-5472.CAN-11-1732.
    1. Accueil | Légifrance, Le Service Public De La Diffusion Du Droit. 2019. [2019-08-22]. Arrêté Du 11 Avril 2008 Relatif Aux Règles De Bonnes Pratiques Cliniques Et Biologiques D'assistance Médicale À La Procréation .
    1. Campana D, Coustan-Smith E. Detection of minimal residual disease in acute leukemia by flow cytometry. Cytometry. 1999 Aug 15;38(4):139–52. doi: 10.1002/cyto.b.21766.

Source: PubMed

3
Iratkozz fel