Skip to main content Accessibility help
×
Hostname: page-component-8448b6f56d-jr42d Total loading time: 0 Render date: 2024-04-18T05:53:13.641Z Has data issue: false hasContentIssue false

Section 1 - Introduction

Published online by Cambridge University Press:  27 March 2021

Jacques Donnez
Affiliation:
Catholic University of Louvain, Brussels
S. Samuel Kim
Affiliation:
University of Kansas School of Medicine
Get access
Type
Chapter
Information
Fertility Preservation
Principles and Practice
, pp. 1 - 34
Publisher: Cambridge University Press
Print publication year: 2021

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

References

Bavister, BD. Early history of in vitro fertilization. Reprod, 2002;124:181196.Google Scholar
Clarke, GN. A.R.T. and history. Hum Reprod, 2006;21:16451650.CrossRefGoogle ScholarPubMed
Yanagimachi, R, Chang, MC. Fertilization of hamster eggs in vitro. Nature, 1963;200:281282.CrossRefGoogle ScholarPubMed
Edwards, RG. Maturation in vitro of human ovarian oocytes. Lancet, 1965;2:926.Google Scholar
Steptoe, PC. Laparoscopy in Gynaecology. Edinburgh: E & S Livingstone, 1967.Google Scholar
Steptoe, PC. Laparoscopy and ovulation. Lancet, 1968;2:913.CrossRefGoogle ScholarPubMed
Edwards, RG, Bavister, BD, Steptoe, PC. Early stages of fertilization in vitro of human oocytes matured in vitro. Nature, 1969;221:632635.Google Scholar
Bavister, BD, Edwards, RG, Steptoe, PC. Identification of the midpiece and tail of the spermatozoon during fertilization of human eggs in vitro. J Reprod Fert, 1969;20:169170.CrossRefGoogle ScholarPubMed
Steptoe, PC, Edwards, RG. Laparoscopic recovery of pre-ovulatory oocytes after priming of ovaries with gonadotrophins. Lancet, 1970;1:683689.CrossRefGoogle Scholar
Steptoe, PC, Edwards, RG. Reimplantation of a human embryo with subsequent tubal pregnancy. Lancet, 1976;1:880882.Google Scholar
Steptoe, PC, Edwards, RG. Birth after the reimplantation of a human embryo. Lancet, 1978;2:366.CrossRefGoogle ScholarPubMed
Trounson, A, Leeton, J, Wood, J et al. Pregnancies in the human by fertilization in vitro and embryo transfer in the controlled ovulatory cycle. Science, 1981;212:681682.Google Scholar
Jones, HW, Jones, GS, Andrews, MC, et al. The program for in vitro fertilization at Norfolk. Fertil Steril, 1982;381:1421.Google Scholar
Steptoe, PC, Edwards, RG. Observations on 767 clinical pregnancies and 500 live births after human in-vitro fertilization. Hum Reprod, 1986;1:8994.Google Scholar
Trounson, A, Mohr, L. Human pregnancy following cryopreservation thawing and transfer of an eight-cell embryo. Nature, 1983;305:707709.CrossRefGoogle ScholarPubMed
Trounson, A, Leeton, J, Besanko, M et al. Pregnancy established in an infertile patient following transfer of a donated embryo fertilized in vitro. Br Med J, 1983;286:835838.Google Scholar
Utian, WF, Goldfarb, JM, Kiwi, R et al. Preliminary experience with in vitro fertilization-surrogate gestational pregnancy. Fertil Steril, 1989;52:633638.Google Scholar
Lenz, S, Lauritsen, JG. Ultrasonically guided percutaneous aspiration of human follicles under local anaesthesia: a new method of collecting oocytes for in vitro fertilization. Fertil Steril, 1982;38:673677.Google Scholar
Gleicher, N, Friberg, J, Fullan, N et al. Egg retrieval for in vitro fertilisation by sonographically controlled vaginal culdocentesis. Lancet, 1983;2:508509.Google Scholar
Wikland, M, Hamberger, L, Enk, L. Transvesical and transvaginal approaches for aspiration of follicles by use of ultrasound. Ann NY Acad Sci, 1985;442:683689.Google Scholar
Porter, RN, Smith, W, Craft, IL et al. Induction of ovulation for in vitro fertilization using buserelin and gonadotrophins. Lancet, 1984;2:12841285.CrossRefGoogle Scholar
Frydman, R, Cornel, C, de Zeigler, D et al. Prevention of premature luteinising hormone and progesterone rise with a GnRH antagonist Nal-Glu in controlled ovarian hyperstimulation. Fertil Steril, 1991;56:921927.CrossRefGoogle ScholarPubMed
Germond, M, Dessole, S, Senn, A et al. Successful in vitro fertilisation and embryo transfer after treatment with recombinant human FSH. Lancet, 1992;339:1170.CrossRefGoogle ScholarPubMed
Devroey, P, Mannaerts, B, Smitz, J et al. First established pregnancy and birth after ovarian stimulation with recombinant follicle stimulating hormone (Org 32489). Hum Reprod, 1993;8:863865.Google Scholar
Laws-King, A, Trounsen, A, Sathananthan, H et al. Fertilization of human oocytes by microinjection of a single spermatozoon under the zona pellucida. Fertil Steril, 1987;48:637642.Google Scholar
Palermo, G, Joris, H, Devroey, P, Van Steirteghem, A. Pregnancies after intracytoplasmic sperm injection of single spermatozoon into an oocyte. Lancet, 1992;2:1718.Google Scholar
Handyside, AH, Kontogianni, EH, Hardy, K et al. Pregnancies from biopsied human pre-implantation embryos sexed by Y-specific DNA amplification. Nature, 1990;344:768770.Google Scholar
Verlinsky, Y, Ginsberg, N, Lifchez, A et al. Analysis of the first polar body: preconception genetic diagnosis. Hum Reprod, 1990;5:826829.Google Scholar
Cha, KY, Yoon, SH, Ko, JJ et al. Pregnancy after in vitro fertilization of human follicular oocytes collected from non-stimulated cycle, their culture in vitro and their transfer according to the donor oocyte program. Fertil Steril, 1989. Abstract O-00: In Proceedings of Annual Meeting of the American Fertility Society.Google Scholar
Silber, SJ, Nagy, Z, Liu, J, Tournaye, H et al. The use of epididymal and testicular spermatozoa for intracytoplasmic sperm injection: the genetic implications for male infertility. Hum Reprod, 1995;10:20312043.CrossRefGoogle ScholarPubMed
Reijo, R, Lee, T-Y, Salo, P et al. Diverse spermatogenic defects in humans caused by Y chromosome deletions encompassing a novel RNA-binding protein gene. Nat Gene, 1995;10:383393.CrossRefGoogle ScholarPubMed
Picton, HM, Wyns, C, Anderson, RA et al. A European perspective on testicular tissue cryopreservation for fertility preservation in prepubertal and adolescent boys. Hum Reprod, 2015;30:24632475.Google Scholar
Gosden, RG. Restoration of fertility in sterilized mice by transferring primordial ovarian follicles. Hum Reprod, 1990;5:499504.CrossRefGoogle Scholar
Gosden, RG, Baird, DT, Wade, JC et al. Restoration of fertility in oophorectomized sheep by ovarian autographs stored at −196°C. Hum Reprod, 1994;9:597603.CrossRefGoogle Scholar
Oktay, K, Economos, K, Kan, M et al. Endocrine function and oocyte retrieval after autologous transplantation of ovarian cortical strips to the forearm. J Am Med Ass, 2001;286:14901493.Google Scholar
Donnez, J, Dolmans, M, Demylle, D et al. Livebirth after orthotopic transplantation of cryopreserved ovarian tissue. Lancet, 2004;364:14051410.Google Scholar
Anderson, RA, Wallace, WHB, Telfer, EE et al. Ovarian tissue cryopreservation for fertility preservation: Clinical and research perspectives. Hum Reprod Open, 2017;1. https://doi.org/10.1093/hropen/hox001Google Scholar
Silber, SJ, Lenahan, KM, Levine, DJ et al. Ovarian transplantation between monozygotic twins discordant for premature ovarian failure. N Engl J Med, 2005;353:5863.CrossRefGoogle ScholarPubMed
Nyboe, Andersen A, Goossens, V, Bhattacharya, S et al. Assisted reproductive technology and intrauterine inseminations in Europe, 2005: results generated from European registers by ESHRE. Hum Reprod, 2009;24:12671287.Google Scholar
De Geyter, C, Calhaz-Jorge, C, Wyns, C et al. The European IVF monitoring consortium (EIM) for the European Society of Human Reproduction and Embryology (ESHRE). Hum Reprod, 2018;1–16. Advance access: https://academic.oup.com/humrep/advance-article-abstract/doi/10.1093/humrep/dey242/50Google Scholar
Code of Practice 9th Edition. Human Fertilisation and Embryology Authority, London 2018 (Draft: For publication October 1, 2018). www.hfea.june-2018-code-of-practice-9th-edition-draftGoogle Scholar
International Federation of Fertility Societies (IFFS). IFFS surveillance 2016. Global Reproductive Health Open, 2016;1:1143.Google Scholar
Fauser, BC, Edwards, RG. The early days of IVF [editorial]. Hum Reprod Update, 2005;11:439459.Google Scholar

Further Recommended Reading

Edwards, RG, Sharpe, DJ. Social values and research in human embryology. Nature, 1971;231:8791.Google Scholar
Steptoe, PC, Edwards, RG. Birth after reimplantation of a human embryo. Lancet, 1978;2:366.CrossRefGoogle ScholarPubMed
Edwards, RG, Steptoe, PC. A Matter of Life. London: Hutchinson, 1980.Google Scholar
Jones, HW Jr. The ethics of in vitro fertilization – 1981. In Edwards RG, Purdy JM (eds.) Human Conception in Vitro. London: Academic Press. 351357.Google Scholar
Jones, HW, Jones, GS, Andrews, MC et al. The programme for in vitro fertilization at Norfolk. Fertil Steril, 1982;38:1421.Google Scholar
Wood, C, Trounson, AO. Historical perspectives of IVF. 2000. In Trounson AO, Gardner DK (eds.) Handbook of in Vitro Fertilization, 2nd edn.Google Scholar
Cohen, J, Trounson, A, Dawson, J et al. The early days of IVF outside the UK. Hum Rep Update, 2005;11:439459.Google Scholar
Edwards, RG. Introduction: the beginnings of human in-vitro fertilization. In Gardner, DK. et al. (eds.) Textbook of Assisted Reproductive Techniques. Informa UK Ltd. 2009, 1730.Google Scholar
Martinez, F et al. Update on fertility preservation from the Barcelona International Society for Fertility Preservation – ESHRE – ASRM 2015 expert meeting: Indications, results and future perspectives. Hum Reprod, 2017;32(9);18021811. https://doi.org/10.1093/humrep/dex2180Google Scholar
Brown, L. My Life as the World’s First Test-Tube Baby. Bristol: Bristol Books CIC, 2015.Google Scholar
Kovacs, G, Brinsden, PR, DeCherney, A. In-Vitro Fertilization: The Pioneers’ History. Cambridge: Cambridge University Press, 2018.Google Scholar

References

Meirow, D, Nugent, D. The effects of radiotherapy and chemotherapy on female reproduction. Hum Reprod Update, 2001;7(6):535543.Google Scholar
Whitehead, E et al. The effect of combination chemotherapy on ovarian function in women treated for Hodgkin’s disease. Cancer, 1983;52(6):988993.Google Scholar
Himelstein-Braw, R, Peters, H, Faber, M. Morphological study of the ovaries of leukaemic children. Br J Cancer, 1978;38(1):8287.CrossRefGoogle ScholarPubMed
Familiari, G et al. Ultrastructure of human ovarian primordial follicles after combination chemotherapy for Hodgkin’s disease. Hum Reprod, 1993;8(12):20802087.Google Scholar
Sklar, CA et al. Premature menopause in survivors of childhood cancer: a report from the childhood cancer survivor study. J Natl Cancer Inst, 2006;98(13):890896.Google Scholar
Chemaitilly, W et al. Acute ovarian failure in the childhood cancer survivor study. J Clin Endocrinol Metab, 2006;91(5):17231728.Google Scholar
Nielsen, SN et al. A 10-year follow up of reproductive function in women treated for childhood cancer. Reprod Biomed Online, 2013;27(2):192200.Google Scholar
Charpentier, AM et al. Anti-Mullerian hormone screening to assess ovarian reserve among female survivors of childhood cancer. J Cancer Surviv, 2014;8(4):548554.Google Scholar
Thomas-Teinturier, C et al. Ovarian reserve after treatment with alkylating agents during childhood. Hum Reprod, 2015;30(6):14371446.Google Scholar
Luke, B et al. Assisted reproductive technology use and outcomes among women with a history of cancer. Hum Reprod, 2016;31(1):183189.Google Scholar
Meirow, D et al. Subclinical depletion of primordial follicular reserve in mice treated with cyclophosphamide: clinical importance and proposed accurate investigative tool. Hum Reprod, 1999;14(7):19031907.Google Scholar
Meirow, D et al. Toxicity of chemotherapy and radiation on female reproduction. Clin Obstet Gynecol, 2010;53(4):727739.Google Scholar
Fan, W. Possible mechanisms of paclitaxel-induced apoptosis. Biochem Pharmacol, 1999;57(11):12151221.Google Scholar
Stumm, S et al. Paclitaxel treatment of breast cancer cell lines modulates Fas/Fas ligand expression and induces apoptosis which can be inhibited through the CD40 receptor. Oncology, 2004;66(2):101111.Google Scholar
Roti Roti, EC et al. Acute doxorubicin insult in the mouse ovary is cell- and follicle-type dependent. PLoS One, 2012;7(8):e42293.Google Scholar
Lopes, F et al. Docetaxel induces moderate ovarian toxicity in mice, primarily affecting granulosa cells of early growing follicles. Mol Hum Reprod, 2014;20(10):948959.Google Scholar
Kalich-Philosoph, L et al. Cyclophosphamide triggers follicle activation and “burnout”; AS101 prevents follicle loss and preserves fertility. Sci Transl Med, 2013;5(185):185ra62.Google Scholar
Chen, XY et al. Follicle loss and apoptosis in cyclophosphamide-treated mice: What’s the matter? Int J Mol Sci, 2016;17(6)836.Google Scholar
Ben-Aharon, I et al. Doxorubicin-induced ovarian toxicity. Reprod Biol Endocrinol, 2010;8:20.Google Scholar
Lopez, SG, Luderer, U. Effects of cyclophosphamide and buthionine sulfoximine on ovarian glutathione and apoptosis. Free Radic Biol Med, 2004;36(11):13661377.Google Scholar
Anderson, RA et al. The effects of chemotherapy and long-term gonadotrophin suppression on the ovarian reserve in premenopausal women with breast cancer. Hum Reprod, 2006;21(10):25832592.Google Scholar
Rosendahl, M et al. Dynamics and mechanisms of chemotherapy-induced ovarian follicular depletion in women of fertile age. Fertil Steril, 2010 June;94(1):156–66, epub March 31.Google Scholar
Kalich-Philosoph, L, Roness, H, Carmely, A et al. Cyclophosphamide triggers follicle activation causing ovarian reserve “burn out”; AS101 prevents follicle loss and preserves fertility. Sci Transl Med, 2013;15(5):185.Google Scholar
Lande, Y et al. Short-term exposure of human ovarian follicles to cyclophosphamide metabolites seems to promote follicular activation in vitro. Reprod Biomed Online, 2017;34(1):104114.Google Scholar
Chang, EM et al. Cisplatin induces overactivation of the dormant primordial follicle through PTEN/AKT/FOXO3a pathway which leads to loss of ovarian reserve in mice. PLoS One, 2015;10(12):e0144245.Google Scholar
Jang, H et al. Melatonin prevents cisplatin-induced primordial follicle loss via suppression of PTEN/AKT/FOXO3a pathway activation in the mouse ovary. J Pineal Res, 2016;60(3):336347.Google Scholar
Durlinger, AL, Visser, JA, Themmen, AP. Regulation of ovarian function: the role of anti-Mullerian hormone. Reproduction, 2002;124(5):601609.Google Scholar
Adhikari, D, Liu, K. Molecular mechanisms underlying the activation of mammalian primordial follicles. Endocr Rev, 2009;30(5):438464.Google Scholar
Kawamura, K et al. Hippo signaling disruption an Akt stimulation of ovarian follicles for infertility treatment. Proc Natl Acad Sci U S A, 2013;110(43):1747417479.CrossRefGoogle ScholarPubMed
Li, J et al. Activation of dormant ovarian follicles to generate mature eggs. Proc Natl Acad Sci U S A, 2010;107(22):1028010284.Google Scholar
McLaughlin, M et al. Inhibition of phosphatase and tensin homologue (PTEN) in human ovary in vitro results in increased activation of primordial follicles but compromises development of growing follicles. Mol Hum Reprod, 2014;20(8):736744.CrossRefGoogle ScholarPubMed
Durlinger, AL et al. Anti-Mullerian hormone inhibits initiation of primordial follicle growth in the mouse ovary. Endocrinology, 2002;143(3):10761084.CrossRefGoogle ScholarPubMed
Durlinger, AL et al. Control of primordial follicle recruitment by anti-Mullerian hormone in the mouse ovary. Endocrinology, 1999;140(12):57895796.Google Scholar
Marcello, MF et al. Structural and ultrastructural study of the ovary in childhood leukemia after successful treatment. Cancer, 1990;66(10):20992104.Google Scholar
Meirow, D et al. Cortical fibrosis and blood-vessels damage in human ovaries exposed to chemotherapy. Potential mechanisms of ovarian injury. Hum Reprod, 2007;22(6):1626–1633.Google Scholar
Nicosia, SV, Matus-Ridley, M, Meadows, AT. Gonadal effects of cancer therapy in girls. Cancer, 1985;55(10):23642372.Google Scholar
Soleimani, R, Heytens, E, Oktay, K. Enhancement of neoangiogenesis and follicle survival by sphingosine-1-phosphate in human ovarian tissue xenotransplants. PLoS One, 2011;6(4):e19475.Google Scholar
Ben-Aharon, I et al. Chemotherapy-induced ovarian failure as a prototype for acute vascular toxicity. Oncologist, 2012;17(11):13861393.CrossRefGoogle ScholarPubMed
Bar-Joseph, H et al. In vivo bioimaging as a novel strategy to detect doxorubicin-induced damage to gonadal blood vessels. PLoS One, 2011;6(9):e23492.Google Scholar
Sanders, JE et al. Pregnancies following high-dose cyclophosphamide with or without high-dose busulfan or total-body irradiation and bone marrow transplantation. Blood, 1996;87:30453052.Google Scholar
Green, DM et al. Fertility of female survivors of childhood cancer: a report from the childhood cancer survivor study. J Clin Oncol, 2009;27(16):26772685.CrossRefGoogle ScholarPubMed
Mackie, EJ, Radford, M, Shalet, SM. Gonadal function following chemotherapy for childhood Hodgkin’s disease. Med Pediatr Oncol, 1996;27(2):7478.Google Scholar
Behringer, K et al. Secondary amenorrhea after Hodgkin’s lymphoma is influenced by age at treatment, stage of disease, chemotherapy regimen, and the use of oral contraceptives during therapy: a report from the German Hodgkin’s Lymphoma Study Group. J Clin Oncol, 2005;23(30):75557564.Google Scholar
Brusamolino, E et al. Treatment of early-stage Hodgkin’s disease with four cycles of ABVD followed by adjuvant radio-therapy: analysis of efficacy and long-term toxicity. Haematologica, 2000;85(10):10321039.Google Scholar
De Bruin, ML et al. Treatment-related risk factors for premature menopause following Hodgkin lymphoma. Blood, 2008;111(1):101108.Google Scholar
Hodgson, DC et al. Fertility among female Hodgkin lymphoma survivors attempting pregnancy following ABVD chemotherapy. Hematol Oncol, 2006;25:1115.Google Scholar
Gershenson, DM. Management of ovarian germ cell tumors. J Clin Oncol, 2007;25(20):29382943.Google Scholar
Gershenson, DM. Menstrual and reproductive function after treatment with combination chemotherapy for malignant ovarian germ cell tumors. J Clin Oncol, 1988;6(2):270275.Google Scholar
Park, MC et al. Risk of ovarian failure and pregnancy outcome in patients with lupus nephritis treated with intravenous cyclophosphamide pulse therapy. Lupus, 2004; 13(8):569574.Google Scholar
Blumenfeld, Z et al. Prevention of irreversible chemotherapy-induced ovarian damage in young women with lymphoma by a gonadotrophin-releasing hormone agonist in parallel to chemotherapy. Hum Reprod, 1996;11(8):1620–1626.Google Scholar
Blumenfeld, Z. How to preserve fertility in young women exposed to chemotherapy? The role of GnRH agonist cotreatment in addition to cryopreservation of embryo, oocytes, or ovaries. Oncologist, 2007;12(9):10441054.Google Scholar
Elgindy, EA et al. Gonadatrophin suppression to prevent chemotherapy-induced ovarian damage: a randomized controlled trial. Obstet Gynecol, 2013;121(1):7886.Google Scholar
Gerber, B et al. Effect of luteinizing hormone-releasing hormone agonist on ovarian function after modern adjuvant breast cancer chemotherapy: the GBG 37 ZORO study. J Clin Oncol, 2011;29(17):23342341.CrossRefGoogle ScholarPubMed
Munster, PN et al. Randomized trial using gonadotropin-releasing hormone agonist triptorelin for the preservation of ovarian function during (neo)adjuvant chemotherapy for breast cancer. J Clin Oncol, 2012;30(5):533538.Google Scholar
Sverrisdottir, A et al. Adjuvant goserelin and ovarian preservation in chemotherapy treated patients with early breast cancer: results from a randomized trial. Breast Cancer Res Treat, 2009;117(3):561567.Google Scholar
Del Mastro, L et al. Effect of the gonadotropin-releasing hormone analogue triptorelin on the occurrence of chemotherapy-induced early menopause in premenopausal women with breast cancer: a randomized trial. JAMA, 2011;306(3):269276.Google Scholar
Badawy, A et al. Gonadotropin-releasing hormone agonists for prevention of chemotherapy-induced ovarian damage: prospective randomized study. Fertil Steril, 2009;91(3):694697.Google Scholar
Moore, HC et al. Goserelin for ovarian protection during breast-cancer adjuvant chemotherapy. N Engl J Med, 2015;372(10):923932.Google Scholar
Song, G, Gao, H, Yuan, Z. Effect of leuprolide acetate on ovarian function after cyclophosphamide-doxorubicin-based chemotherapy in premenopausal patients with breast cancer: results from a phase II randomized trial. Med Oncol, 2013;30(3):667.Google Scholar
Gonfloni, S et al. Inhibition of the c-Abl-TAp63 pathway protects mouse oocytes from chemotherapy-induced death. Nat Med, 2009;15(10):11791185.Google Scholar
Kerr, JB et al. DNA damage-induced primordial follicle oocyte apoptosis and loss of fertility require TAp63-mediated induction of Puma and Noxa. Mol Cell, 2012 November 9;48(3):343352.Google Scholar
Morgan, S et al. Cisplatin and doxorubicin induce distinct mechanisms of ovarian follicle loss; imatinib provides selective protection only against cisplatin. PLoS One, 2013;8(7):e70117.Google Scholar
Kim, SY et al. Rescue of platinum-damaged oocytes from programmed cell death through inactivation of the p53 family signaling network. Cell Death Differ, 2013;20(8):987997.Google Scholar
Hancke, K et al. Sphingosine 1-phosphate protects ovaries from chemotherapy-induced damage in vivo. Fertil Steril, 2007;87(1):172177.Google Scholar
Li, F et al. Sphingosine-1-phosphate prevents chemotherapy-induced human primordial follicle death. Hum Reprod, 2014;29(1):107113.Google Scholar
Zhou, L et al. Rapamycin prevents cyclophosphamide-induced over-activation of primordial follicle pool through PI3K/Akt/mTOR signaling pathway in vivo. J Ovarian Res, 2017;10(1):56.Google Scholar
Goldman, KN et al. mTORC1/2 inhibition preserves ovarian function and fertility during genotoxic chemotherapy. Proc Natl Acad Sci U S A, 2017;114(12):31863191.Google Scholar
Sonigo, C et al. AMH prevents primordial ovarian follicle loss and fertility alteration in cyclophosphamide-treated mice. FASEB J, 2018;33(1):12781287,fj201801089R.Google Scholar
Kano, M et al. AMH/MIS as a contraceptive that protects the ovarian reserve during chemotherapy. Proc Natl Acad Sci U S A, 2017;114(9):E1688E1697.Google Scholar
Meirow, D, Dor, J. Epidemiology and infertility in cancer patients. In Tulandi, T, Gosden, R (eds.)Preservation of Fertility. Abingdon: Taylor and Francis. 2004, 2138.Google Scholar

References

SEER. Surveillance, Epidemiology, and End Results (SEER) Program (www.seer.cancer.gov). SEER*Stat Database: Incidence, Survival & Mortality, November 2017 ed. 2018.Google Scholar
Mertens, AC, Yong, J, Dietz, AC, et al. Conditional survival in pediatric malignancies: analysis of data from the Childhood Cancer Survivor Study and the Surveillance, Epidemiology, and End Results Program. Cancer. 2015;121(7):11081117. DOI:10.1002/cncr.29170Google Scholar
Landier, W, Armenian, S, Bhatia, S. Late effects of childhood cancer and its treatment. Pediatr Clin North Am, 2015;62(1):275300. DOI:10.1016/j.pcl.2014.09.017Google Scholar
Anderson, RA, Brewster, DH, Wood, R et al. The impact of cancer on subsequent chance of pregnancy: a population-based analysis. Hum Reprod, 2018;33(7):12811290. DOI:10.1093/humrep/dey216Google Scholar
Wallace, WH, Kelsey, TW, Anderson, RA. Fertility preservation in pre-pubertal girls with cancer: the role of ovarian tissue cryopreservation. Fertil Steril, 2016;105(1):612. DOI:10.1016/j.fertnstert.2015.11.041Google Scholar
Gerstl, B, Sullivan, E, Chong, S et al. Reproductive outcomes after a childhood and adolescent young adult cancer diagnosis in female cancer survivors: a systematic review and meta-analysis. J Adolesc Young Adult Oncol, 2018. DOI:10.1089/jayao.2018.0036CrossRefGoogle Scholar
Depmann, M, Faddy, MJ, van der Schouw, YT et al. The relationship between variation in size of the primordial follicle pool and age at natural menopause. J Clin Endocrinol Metab, 2015;100(6):E845E851. DOI:10.1210/jc.2015-1298Google Scholar
Kelsey, TW, Anderson, RA, Wright, P, Nelson, SM, Wallace, WH. Data-driven assessment of the human ovarian reserve. Mol Hum Reprod, 2012;18(2):7987. DOI:10.1093/molehr/gar059Google Scholar
Wallace, WH, Kelsey, TW. Human ovarian reserve from conception to the menopause. PLoS One, 2010;5(1):e8772. DOI:10.1371/journal.pone.0008772Google Scholar
Johnson, J, Canning, J, Kaneko, T, Pru, JK, Tilly, JL. Germline stem cells and follicular renewal in the postnatal mammalian ovary. Nature, 2004;428(6979):145150. DOI:10.1038/nature02316CrossRefGoogle ScholarPubMed
Lee, HJ, Selesniemi, K, Niikura, Y et al. Bone marrow transplantation generates immature oocytes and rescues long-term fertility in a preclinical mouse model of chemotherapy-induced premature ovarian failure. J Clin Oncol, 2007;25(22):31983204. DOI:10.1200/JCO.2006.10.3028Google Scholar
Johnson, J, Skaznik-Wikiel, M, Lee, HJ et al. Setting the record straight on data supporting postnatal oogenesis in female mammals. Cell Cycle, 2005;4(11):14711477. DOI:10.4161/cc.4.11.2186Google Scholar
Zou, K, Yuan, Z, Yang, Z et al. Production of offspring from a germline stem cell line derived from neonatal ovaries. Nat Cell Biol, 2009;11(5):631636. DOI:10.1038/ncb1869CrossRefGoogle ScholarPubMed
Hummitzsch, K, Anderson, RA, Wilhelm, D et al. Stem cells, progenitor cells, and lineage decisions in the ovary. Endocr Rev, 2015;36(1):6591. DOI:10.1210/er.2014-1079Google Scholar
McLaughlin, M, Kelsey, TW, Wallace, WH, Anderson, RA, Telfer, EE. Non-growing follicle density is increased following adriamycin, bleomycin, vinblastine and dacarbazine (ABVD) chemotherapy in the adult human ovary. Hum Reprod, 2017;32(1):165174. DOI:10.1093/humrep/dew260Google Scholar
Jadoul, P, Dolmans, MM, Donnez, J. Fertility preservation in girls during childhood: is it feasible, efficient and safe and to whom should it be proposed? Hum Reprod Update. 2010;16(6):617630. DOI:10.1093/humupd/dmq010Google Scholar
Gao, W, Liang, JX, Yan, Q. Exposure to radiation therapy is associated with female reproductive health among childhood cancer survivors: a meta-analysis study. J Assist Reprod Genet, 2015;32(8):11791186. DOI:10.1007/s10815-015-0490-6Google Scholar
Meirow, D, Nugent, D. The effects of radiotherapy and chemotherapy on female reproduction. Hum Reprod Update, 2001;7(6):535543.Google Scholar
Chow, EJ, Stratton, KL, Leisenring, WM et al. Pregnancy after chemotherapy in male and female survivors of childhood cancer treated between 1970 and 1999: a report from the Childhood Cancer Survivor Study cohort. Lancet Oncol, 2016;17(5):567576. DOI:10.1016/S1470-2045(16)00086-3Google Scholar
Wallace, WH, Thomson, AB, Kelsey, TW. The radiosensitivity of the human oocyte. Hum Reprod, 2003;18(1):117121.Google Scholar
Salooja, N, Szydlo, RM, Socie, G et al. Pregnancy outcomes after peripheral blood or bone marrow transplantation: a retrospective survey. Lancet, 2001;358(9278):271276.Google Scholar
Sanders, JE, Hawley, J, Levy, W et al. Pregnancies following high-dose cyclophosphamide with or without high-dose busulfan or total-body irradiation and bone marrow transplantation. Blood, 1996;87(7):30453052.Google Scholar
Wallace, WH, Shalet, SM, Crowne, EC, Morris-Jones, PH, Gattamaneni, HR. Ovarian failure following abdominal irradiation in childhood: natural history and prognosis. Clin Oncol (R Coll Radiol), 1989;1(2):7579.Google Scholar
Anderson, RA, Mitchell, RT, Kelsey, TW et al. Cancer treatment and gonadal function: experimental and established strategies for fertility preservation in children and young adults. Lancet Diabetes Endocrinol, 2015;3(7):556567. DOI:10.1016/S2213-8587(15)00039-XGoogle Scholar
Wallace, WH, Thomson, AB, Saran, F, Kelsey, TW. Predicting age of ovarian failure after radiation to a field that includes the ovaries. Int J Radiat Oncol Biol Phys, 2005;62(3):738744. DOI:10.1016/j.ijrobp.2004.11.038Google Scholar
Davies, HA, Didcock, E, Didi, M et al. Growth, puberty and obesity after treatment for leukaemia. Acta Paediatr Suppl, 1995;411:4550; discussion 1.Google Scholar
Bath, LE, Anderson, RA, Critchley, HO, Kelnar, CJ, Wallace, WH. Hypothalamic-pituitary-ovarian dysfunction after prepubertal chemotherapy and cranial irradiation for acute leukaemia. Hum Reprod, 2001;16(9):18381844.Google Scholar
Constine, LS, Woolf, PD, Cann, D et al. Hypothalamic-pituitary dysfunction after radiation for brain tumors. N Engl J Med, 1993;328(2):8794. DOI:10.1056/NEJM199301143280203Google Scholar
Merchant, TE, Rose, SR, Bosley, C et al. Growth hormone secretion after conformal radiation therapy in pediatric patients with localized brain tumors. J Clin Oncol, 2011;29(36):47764780. DOI:10.1200/JCO.2011.37.9453Google Scholar
Green, DM, Nolan, VG, Kawashima, T et al. Decreased fertility among female childhood cancer survivors who received 22–27 Gy hypothalamic/pituitary irradiation: a report from the Childhood Cancer Survivor Study. Fertil Steril, 2011;95(6):19221927, 7 e1. DOI:10.1016/j.fertnstert.2011.02.002Google Scholar
Green, DM, Kawashima, T, Stovall, M et al. Fertility of female survivors of childhood cancer: a report from the childhood cancer survivor study. J Clin Oncol, 2009;27(16):26772685. DOI:10.1200/JCO.2008.20.1541Google Scholar
Marci, R, Mallozzi, M, Di Benedetto, L et al. Radiations and female fertility. Reprod Biol Endocrinol, 2018;16(1):112. DOI:10.1186/s12958-018-0432-0Google Scholar
Bath, LE, Critchley, HO, Chambers, SE et al. Ovarian and uterine characteristics after total body irradiation in childhood and adolescence: response to sex steroid replacement. Br J Obstet Gynaecol, 1999;106(12):12651272.Google Scholar
Larsen, EC, Schmiegelow, K, Rechnitzer, C et al. Radiotherapy at a young age reduces uterine volume of childhood cancer survivors. Acta Obstet Gynecol Scand, 2004;83(1):96102.Google Scholar
Wo, JY, Viswanathan, AN. Impact of radiotherapy on fertility, pregnancy, and neonatal outcomes in female cancer patients. Int J Radiat Oncol Biol Phys, 2009;73(5):13041312. DOI:10.1016/j.ijrobp.2008.12.016Google Scholar
Zheng, Y, Lei, Q, Jongejan, A et al. The influence of retinoic acid-induced differentiation on the radiation response of male germline stem cells. DNA Repair (Amst), 2018;70:5566. DOI:10.1016/j.dnarep.2018.08.027CrossRefGoogle ScholarPubMed
Shalet, SM, Tsatsoulis, A, Whitehead, E, Read, G. Vulnerability of the human Leydig cell to radiation damage is dependent upon age. J Endocrinol, 1989;120(1):161165.CrossRefGoogle ScholarPubMed
Castillo, LA, Craft, AW, Kernahan, J, Evans, RG, Aynsley-Green, A. Gonadal function after 12-Gy testicular irradiation in childhood acute lymphoblastic leukaemia. Med Pediatr Oncol, 1990;18(3):185189.Google Scholar
Grundy, RG, Leiper, AD, Stanhope, R, Chessells, JM. Survival and endocrine outcome after testicular relapse in acute lymphoblastic leukaemia. Arch Dis Child, 1997;76(3):190196.Google Scholar
Leiper, AD, Stanhope, R, Lau, T et al. The effect of total body irradiation and bone marrow transplantation during childhood and adolescence on growth and endocrine function. Br J Haematol, 1987;67(4):419426.Google Scholar
Choi, M, Hayes, JP, Mehta, MP et al. Using intensity-modulated radiotherapy to spare the kidney in a patient with seminoma and a solitary kidney: a case report. Tumori, 2013;99(2):e38e42. DOI:10.1700/1283.14205Google Scholar
Jonska-Gmyrek, J, Peczkowski, P, Michalski, W, et al. Radiotherapy in testicular germ cell tumours – a literature review. Contemp Oncol (Pozn), 2017;21(3):203208. DOI:10.5114/wo.2017.69592Google ScholarPubMed

References

Argyle, CE, Harper, JC, Davies, MC. Oocyte cryopreservation: where are we now? Hum Reprod Update, 2016;22:440449.Google Scholar
Devroey, P, Van Steirteghem, A. A review of ten years experience of ICSI. Hum Reprod Update, 2004;10:1928.Google Scholar
Ward, E, DeSantis, C, Robbins, A, Kohler, B, Jemal, A. Childhood and adolescent cancer statistics. CA Cancer J Clin, 2014;64:83103.Google Scholar
Oeffinger, KC, Mertens, AC, Sklar, CA et al. Chronic health conditions in adult survivors of childhood cancer. N Engl J Med, 2006;355:15721582.Google Scholar
van Dorp, W, Haupt, R, Anderson, RA et al. Reproductive function and outcomes in female survivors of childhood, adolescent, and young adult cancer: a review. J Clin Oncol, 2018:JCO2017763441.Google Scholar
Peate, M, Meiser, B, Hickey, M, Friedlander, M. The fertility-related concerns, needs and preferences of younger women with breast cancer: a systematic review. Breast Cancer Res Treat, 2009;116:215223.Google Scholar
Shandley, LM, Spencer, JB, Fothergill, A et al. Impact of tamoxifen therapy on fertility in breast cancer survivors. Fertil Steril, 2017;107:243252 e245.Google Scholar
Lambertini, M, Kroman, N, Ameye, L et al. Long-term safety of pregnancy following breast cancer according to estrogen receptor status. J Natl Cancer Inst, 2018;110:426429.Google Scholar
Frobisher, C, Lancashire, ER, Winter, DL, Jenkinson, HC, Hawkins, MM. Long-term population-based marriage rates among adult survivors of childhood cancer in Britain. Int J Cancer, 2007;121:846855.Google Scholar
Koch, SV, Kejs, AM, Engholm, G et al. Marriage and divorce among childhood cancer survivors. J Pediatr Hematol Oncol, 2011;33:500505.Google Scholar
Ganz, PA, Rowland, JH, Desmond, K, Meyerowitz, BE, Wyatt, GE. Life after breast cancer: understanding women’s health-related quality of life and sexual functioning. J Clin Oncol, 1998;16:501514.Google Scholar
Howard-Anderson, J, Ganz, PA, Bower, JE, Stanton, AL. Quality of life, fertility concerns, and behavioral health outcomes in younger breast cancer survivors: a systematic review. J Natl Cancer Inst, 2012;104:386405.Google Scholar
Himelstein-Braw, R, Peters, H, Faber, M. Influence of irradiation and chemotherapy on the ovaries of children with abdominal tumours. Br J Cancer, 1977;36:269275.Google Scholar
Chapman, RM, Sutcliffe, SB, Malpas, JS. Cytotoxic-induced ovarian failure in women with Hodgkin’s disease. I. Hormone function. JAMA, 1979;242:18771881.CrossRefGoogle ScholarPubMed
Morgan, S, Anderson, RA, Gourley, C, Wallace, WH, Spears, N. How do chemotherapeutic agents damage the ovary? Hum Reprod Update, 2012;18:525535.Google Scholar
Kalich-Philosoph, L, Roness, H, Carmely, A et al. Cyclophosphamide triggers follicle activation and “burnout”; AS101 prevents follicle loss and preserves fertility. Sci Transl Med, 2013;5:185ra162.Google Scholar
Meirow, D, Dor, J, Kaufman, B et al. Cortical fibrosis and blood-vessels damage in human ovaries exposed to chemotherapy. Potential mechanisms of ovarian injury. Hum Reprod, 2007;22:16261633.Google Scholar
Oktem, O, Oktay, K. Quantitative assessment of the impact of chemotherapy on ovarian follicle reserve and stromal function. Cancer, 2007;110:22222229.Google Scholar
Roti Roti, EC, Leisman, SK, Abbott, DH, Salih, SM. Acute doxorubicin insult in the mouse ovary is cell- and follicle-type dependent. PLoS One, 2012;7:e42293.Google Scholar
Anderson, RA, Remedios, R, Kirkwood, AA et al. Chemotherapy regimen, age and pretreatment ovarian reserve as determinants of ovarian function after response-adapted therapy in patients with Hodgkin lymphoma: a prospective cohort study. Lancet Oncol, 2018 October;19(10):13281337.Google Scholar
Meistrich, ML. Male gonadal toxicity. Pediatr Blood Cancer, 2009;53:261266.Google Scholar
van Alphen, MM, van de Kant, HJ, de Rooij, DG. Repopulation of the seminiferous epithelium of the rhesus monkey after X irradiation. Radiat Res, 1988;113:487500.Google Scholar
Poganitsch-Korhonen, M, Masliukaite, I, Nurmio, M et al. Decreased spermatogonial quantity in prepubertal boys with leukaemia treated with alkylating agents. Leukemia, 2017;31:14601463.Google Scholar
Stukenborg, JB, Alves-Lopes, JP, Kurek, M et al. Spermatogonial quantity in human prepubertal testicular tissue collected for fertility preservation prior to potentially sterilizing therapy. Hum Reprod, 2018;33:16771683.Google Scholar
Green, DM, Liu, W, Kutteh, WH et al. Cumulative alkylating agent exposure and semen parameters in adult survivors of childhood cancer: a report from the St Jude Lifetime Cohort Study. Lancet Oncol, 2014;15:12151223.Google Scholar
Loren, AW, Mangu, PB, Beck, LN et al. Fertility preservation for patients with cancer: American Society of Clinical Oncology clinical practice guideline update. J Clin Oncol, 2013;31:25002510.Google Scholar
Mitchell, RT, Stukenborg, JB, Jahnukainen, K. Male hypogonadism due to cancer treatment. In Winters, SJ, Huhtaniemi, IT (eds.) Male Hypogonadism: Basic, Clinical, and Therapeutic Principles. Springer: Humana Press, 2017.Google Scholar
Marmor, D, Grob-Menendez, F, Duyck, F, Delafontaine, D. Very late return of spermatogenesis after chlorambucil therapy: case reports. Fertil Steril, 1992;58:845846.Google Scholar
Imai, A, Ichigo, S, Matsunami, K, Takagi, H, Kawabata, I. Ovarian function following targeted anti-angiogenic therapy with bevacizumab. Mol Clin Oncol, 2017;6:807810.Google Scholar
Wallace, WH, Thomson, AB, Saran, F, Kelsey, TW. Predicting age of ovarian failure after radiation to a field that includes the ovaries. Int J Radiat Oncol Biol Phys, 2005;62:738744.Google Scholar
Sanders, JE, Hawley, J, Levy, W et al. Pregnancies following high-dose cyclophosphamide with or without high-dose busulfan or total-body irradiation and bone marrow transplantation. Blood, 1996;87:30453052.Google Scholar
Salooja, N, Szydlo, RM, Socie, G et al. Pregnancy outcomes after peripheral blood or bone marrow transplantation: a retrospective survey. Lancet, 2001;358:271276.Google Scholar
Signorello, LB, Cohen, SS, Bosetti, C et al. Female survivors of childhood cancer: preterm birth and low birth weight among their children. J Natl Cancer Inst, 2006;98:14531461.Google Scholar
Reulen, RC, Bright, CJ, Winter, DL et al. Pregnancy and labor complications in female survivors of childhood cancer: the British childhood cancer survivor study. J Natl Cancer Inst, 2017;109. DOI:10.1093/jnci/djx1056Google Scholar
Bath, LE, Critchley, HOD, Chambers, SE et al. Ovarian and uterine characteristics after total body irradiation in childhood and adolescence: response to sex steroid replacement. Brit J Obstet Gynaecol, 1999;106:12651272.Google Scholar
Kelsey, TW, Ginbey, E, Chowdhury, MM et al. A validated normative model for human uterine volume from birth to age 40 years. PLoS One, 2016;11:e0157375.Google Scholar
Bath, LE, Anderson, RA, Critchley, HOD, Kelnar, CJH, Wallace, WHB. Hypothalamic–pituitary-ovarian dysfunction after prepubertal chemotherapy and cranial irradiation for acute leukaemia. Hum Reprod, 2001;16:18381844.Google Scholar
Ogilvy-Stuart, AL, Shalet, SM. Effect of radiation on the human reproductive system. Environ Health Perspect, 1993;101(Suppl 2):109116.Google Scholar
van Dorp, W, Mulder, RL, Kremer, LC et al. Recommendations for premature ovarian insufficiency surveillance for female survivors of childhood, adolescent, and young adult cancer: a report from the international late effects of childhood cancer guideline harmonization group in collaboration with the PanCareSurFup consortium. J Clin Oncol, 2016;34:34403450.Google Scholar
Green, DM, Kawashima, T, Stovall, M et al. Fertility of female survivors of childhood cancer: a report from the childhood cancer survivor study. J Clin Oncol, 2009;27:26772685.Google Scholar
Green, DM, Sklar, CA, Boice, JD, Jr. et al. Ovarian failure and reproductive outcomes after childhood cancer treatment: results from the Childhood Cancer Survivor Study. J Clin Oncol, 2009;27:23742381.Google Scholar
Chow, EJ, Stratton, KL, Leisenring, WM et al. Pregnancy after chemotherapy in male and female survivors of childhood cancer treated between 1970 and 1999: a report from the Childhood Cancer Survivor Study cohort. Lancet Oncol, 2016;17:567576.Google Scholar
Barton, SE, Najita, JS, Ginsburg, ES et al. Infertility, infertility treatment, and achievement of pregnancy in female survivors of childhood cancer: a report from the Childhood Cancer Survivor Study cohort. Lancet Oncol, 2013;14:873881.Google Scholar
Letourneau, JM, Ebbel, EE, Katz, PP et al. Acute ovarian failure underestimates age-specific reproductive impairment for young women undergoing chemotherapy for cancer. Cancer, 2012;118:19331939.Google Scholar
Bramswig, JH, Riepenhausen, M, Schellong, G. Parenthood in adult female survivors treated for Hodgkin’s lymphoma during childhood and adolescence: a prospective, longitudinal study. Lancet Oncol, 2015;16:667675.Google Scholar
Behringer, K, Breuer, K, Reineke, T et al. Secondary amenorrhea after Hodgkin’s lymphoma is influenced by age at treatment, stage of disease, chemotherapy regimen, and the use of oral contraceptives during therapy: a report from the German Hodgkin’s Lymphoma Study Group. J Clin Oncol, 2005;23:75557564.Google Scholar
Swerdlow, AJ, Cooke, R, Bates, A et al. Risk of premature menopause after treatment for Hodgkin’s lymphoma. J Natl Cancer Inst, 2014;106(pii):dju207.Google Scholar
Petrek, JA, Naughton, MJ, Case, LD et al. Incidence, time course, and determinants of menstrual bleeding after breast cancer treatment: a prospective study. J Clin Oncol, 2006;24:10451051.Google Scholar
Bath, LE, Wallace, WH, Shaw, MP, Fitzpatrick, C, Anderson, RA. Depletion of ovarian reserve in young women after treatment for cancer in childhood: detection by anti-Mullerian hormone, inhibin B and ovarian ultrasound. Hum Reprod, 2003;18:23682374.Google Scholar
Jayasinghe, YL, Wallace, WHB, Anderson, RA. Ovarian function, fertility and reproductive lifespan in cancer patients. Expert Rev Endocrinol Metab, 2018;13:125136.Google Scholar
Anderson, RA, Themmen, APN, Al Qahtani, A, Groome, NP, Cameron, DA. The effects of chemotherapy and long-term gonadotrophin suppression on the ovarian reserve in premenopausal women with breast cancer. Human Reprod, 2006;21:25832592.Google Scholar
Chai, J, Howie, AF, Cameron, DA, Anderson, RA. A highly-sensitive anti-Mullerian hormone assay improves analysis of ovarian function following chemotherapy for early breast cancer. Eur J Cancer, 2014;50:23672374.Google Scholar
Dezellus, A, Barriere, P, Campone, M et al. Prospective evaluation of serum anti-Mullerian hormone dynamics in 250 women of reproductive age treated with chemotherapy for breast cancer. Eur J Cancer, 2017;79:7280.Google Scholar
Su, HC, Haunschild, C, Chung, K et al. Prechemotherapy antimullerian hormone, age, and body size predict timing of return of ovarian function in young breast cancer patients. Cancer, 2014;120:36913698.Google Scholar
Anderson, RA, Cameron, DA. Pretreatment serum anti-mullerian hormone predicts long-term ovarian function and bone mass after chemotherapy for early breast cancer. J Clin Endocrinol Metab, 2011;96:13361343.Google Scholar
Hagen, CP, Vestergaard, S, Juul, A et al. Low concentration of circulating antimullerian hormone is not predictive of reduced fecundability in young healthy women: a prospective cohort study. Fertil Steril, 2012;98:16021608 e1602.Google Scholar
Steiner, AZ, Pritchard, D, Stanczyk, FZ et al. Association between biomarkers of ovarian reserve and infertility among older women of reproductive age. JAMA, 2017;318:13671376.Google Scholar
Anderson, RA, Brewster, DH, Wood, R et al. The impact of cancer on subsequent chance of pregnancy: a population-based analysis. Hum Reprod, 2018;33:12811290.Google Scholar
van Beek, RD, Smit, M, van den Heuvel-Eibrink, MM et al. Inhibin B is superior to FSH as a serum marker for spermatogenesis in men treated for Hodgkin’s lymphoma with chemotherapy during childhood. Hum Reprod, 2007;22:32153222.Google Scholar
Green, DM, Zhu, L, Zhang, N et al. Lack of specificity of plasma concentrations of inhibin B and follicle-stimulating hormone for identification of azoospermic survivors of childhood cancer: a report from the St Jude lifetime cohort study. J Clin Oncol, 2013;31:13241328.Google Scholar
Kelsey, TW, McConville, L, Edgar, AB et al. Follicle Stimulating Hormone is an accurate predictor of azoospermia in childhood cancer survivors. PLoS One, 2017;12:e0181377.Google Scholar
Wallace, WH, Smith, AG, Kelsey, TW, Edgar, AE, Anderson, RA Fertility preservation for girls and young women with cancer: population-based validation of criteria for ovarian tissue cryopreservation. Lancet Oncol, 2014;15:11291136.Google Scholar
Jones, GL, Hughes, J, Mahmoodi, N et al. Observational study of the development and evaluation of a fertility preservation patient decision aid for teenage and adult women diagnosed with cancer: the Cancer, Fertility and Me research protocol. BMJ Open, 2017;7: e013219.Google Scholar
Peate, M, Meiser, B, Cheah, BC et al. Making hard choices easier: a prospective, multicentre study to assess the efficacy of a fertility-related decision aid in young women with early-stage breast cancer. Br J Cancer, 2012;106:10531061.Google Scholar
Anderson, RA, Mitchell, RT, Kelsey, TW et al. Cancer treatment and gonadal function: experimental and established strategies for fertility preservation in children and young adults. Lancet Diabetes Endocrinol, 2015;3:556567.Google Scholar

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×