Skip to main content Accessibility help
×
Hostname: page-component-8448b6f56d-m8qmq Total loading time: 0 Render date: 2024-04-23T08:11:21.890Z Has data issue: false hasContentIssue false

Section 3 - Fertility Preservation in Cancer and Non-Cancer Patients

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. 67 - 126
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

Surveillance, Epidemiology and End Results Program, 1975–2015, Division of Cancer Control and Population Sciences, National Cancer Institute, 2018.Google Scholar
Larsen, EC, Müller, J, Schmiegelow, K et al. Reduced ovarian function in long-term survivors of radiation and chemotherapy-treated childhood cancer. J Clin Endocrinol Met, 2003;88:53075314.Google Scholar
Gidoni, Y, Holzer, H, Tulandi, T et al. Fertility preservation in patients with non-oncological conditions. Reprod Biomed Online, 2008;16:792800.CrossRefGoogle ScholarPubMed
Cobo, A, Bellver, J, Domingo, J et al. New options in assisted reproduction technology: the Cryotop method of oocyte vitrification. Reprod Biomed Online, 2008;17:6872.Google Scholar
Meseguer, M, Molina, N, García-Velasco, JA et al. Sperm cryopreservation in oncological patients: a 14-year follow-up study. Fertil Steril, 2006;85:640645.CrossRefGoogle ScholarPubMed
Kuwayama, M, Vajta, G, Kato, O et al. Highly efficient vitrification method for cryopreservation of human oocytes. Reprod Biomed Online, 2005;11:300308.Google Scholar
Silber, SJ, Derosa, M, Pineda, J et al. A series of monozygotic twins discordant for ovarian failure: ovary transplantation (cortical versus microvascular) and cryopreservation. Hum Reprod, 2008;23:15311537.CrossRefGoogle ScholarPubMed
Elgindy, E, Sibai, H, Abdelghani, A et al. Protecting ovaries during chemotherapy through gonad suppression: A systematic review and metaanalysis. Obstet Gynecol, 2015;126:187195.CrossRefGoogle ScholarPubMed
De Vos, M, Smitz, J, Woodruff, T. Fertility preservation in women with cancer. Lancet, 2014;384:13021310.Google Scholar
Tulandi, T, Huang, JY, Tan, SL. Preservation of female fertility: an essential progress. Obstet Gynecol, 2008;112:11601172.CrossRefGoogle ScholarPubMed
Bines, J, Oleske, DM, Cobleigh, MA. Ovarian function in premenopausal women treated with adjuvant chemotherapy for breast cancer. J Clin Oncol, 1996;14:17181729.CrossRefGoogle ScholarPubMed
Poniatowski, BC, Grimm, P, Cohen, G. Chemotherapy-induced menopause: A literature review. Cancer Invest, 2001;19:641648.Google Scholar
Meirow, D, Nugent, D. The effects of radiotherapy and chemotherapy on female reproduction. Hum Reprod Update, 2001;7:535543.Google Scholar
Meirow, D, Epstein, M, Lewis, H et al. Administration of cyclophosphamide at different stages of follicular maturation in mice: effects on reproductive performance and fetal malformations. Hum Reprod, 2001;16:632637.Google Scholar
Martinez, F. Update on fertility preservation from the Barcelona International Society for Fertility Preservation-ESHRE-ASRM 2015 expert meeting: indications, results and future perspectives. Fertil Steril, 2017;108:407415.Google Scholar
Sonmezer, M, Oktay, K. Fertility preservation in young women undergoing breast cancer therapy. The Oncologist, 2006;11:422434.Google Scholar
Warne, GL, Fairley, KF, Hobbs, JB et al. Cyclophosphamide-induced ovarian failure. N Engl J Med, 1973;289:11591162.CrossRefGoogle ScholarPubMed
Marcello, MF, Nuciforo, G, Romeo, R et al. Structural and ultrastructural study of the ovary in childhood leukemia after successful treatment. Cancer, 1990;66:20992104.3.0.CO;2-3>CrossRefGoogle ScholarPubMed
Familiari, G, Caggiati, A, Nottola, SA et al. Ultrastructure of human primordial follicles after combination chemotherapy for Hodgkin´s disease. Hum Reprod, 1993;8:20802087.CrossRefGoogle 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
Anderson, RA, Themmen, AP, Al-Qahtani, A et al. The effects of chemotherapy and long term gonadotrophin suppression on the ovarian reserve in premenopausal women with breast cancer. Hum Reprod, 2006;21:25832592.Google Scholar
Wallace, WH, Thomson, AB, Kelsey, TW. The radiosensitivity of human oocyte. Hum Reprod, 2003;18:117121.Google Scholar
Howell, SJ, Shalet, S. Gonadal damage from chemotherapy and radiotherapy. Endocrinol Metab Clin, 1998;27:927943.CrossRefGoogle ScholarPubMed
Patrick, K, Wallace, WH, Critchley, H. Late reproductive effects of cancer treatment in female survivors of childhood malignancy. Curr Obstet Gynecol, 2003;13:369372.CrossRefGoogle Scholar
Goynumer, G, Kayabasoglu, F, Aydogdu, S et al. The effect of tubal sterilization through electrocoagulation on the ovarian reserve. Contraception, 2009;80:9094.Google Scholar
Hernández-Díaz, S, Werler, MM, Walker, AM et al. Folic acid antagonists during pregnancy and the risk of birth defects. N Engl J Med, 2000;343:16081614.Google Scholar
Alonso, V, Linares, V, Bellés, M et al. Sulfasalazine induced oxidative stress: a possible mechanism of male infertility. Reprod Toxicol, 2009;27:3540.Google Scholar
Ostensen, , Khamashta, M, Lockshin, M et al. Anti-inflammatory and immunosuppressive drugs and reproduction. Arthritis Res Ther, 2006;8:209.CrossRefGoogle ScholarPubMed
Gutierrez, JC, Hwang, K. The toxicity of methotrexate in male fertility and paternal eratogenicity. Expert Opin Drug Metab Toxicol, 2017;1:5158.CrossRefGoogle Scholar
Martini, AC, Molina, RI, Tissera, AD et al. Analysis of semen from patients chronically treated with low or moderate doses of aspirin like drugs. Fertil Steril, 2003;80:221222.Google Scholar
Huang, JY, Tulandi, T, Holzer, H et al. Combining ovarian tissue cryobanking with retrieval of immature oocytes followed by in vitro maturation and vitrification: an additional strategy of fertility preservation. Fertil Steril, 2008;89:567572.Google Scholar
Tao, T, del Valle, A. Human oocyte and ovarian tissue cryopreservation and its application. J Assist Reprod Genet, 2008;25:287296.Google Scholar
Cobo, A, Kuwayama, M, Pérez, S et al. Comparison of concomitant outcome achieved with fresh and cryopreserved oocytes vitrified by the Cryotop method. Fertil Steril, 2008;89:16571664.Google Scholar
Chian, RC, Huang, JY, Tan, SL et al. Obstetric and perinatal outcome in 200 infants conceived from vitrified oocytes. Reprod Biomed Online, 2008;16:608610.Google Scholar
The Ethics Committee of the American Society for Reproductive Medicine. Fertility preservation and reproduction in cancer patients. Fertil Steril, 2005;83:16221628.Google Scholar
Nugent, D, Meirow, D, Brook, PF et al. Transplantation in reproductive medicine: previous experience, present knowledge and future prospects. Hum Reprod Update, 1997;3:267280.Google Scholar
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;32:11671170.Google Scholar
Van der Ven, H, Liebenthron, J, Beckmann, M et al. Ninety-five orthotropic transplantations in 74 women of ovarian tissue after cytotoxic treatment in a fertility preservation network: tissue activity, pregnancy and delivery rates. Hum Reprod, 2016;31:20312041.CrossRefGoogle Scholar
Diaz-Garcia, C, Domingo, J, Garcia-Velasco, JA et al. Oocyte vitrification versus ovarian cortex transplantation in fertility preservation for adult women undergoing gonadotoxic treatments: a prospective cohort study. Fertil Steril, 2018;109:478485.Google Scholar
Dolmans, MM, Luyckx, V, Donnez, J, Andersen, CY, Greve, T. Risk of transferring malignant cells with transplanted frozen-thawed ovarian tissue. Fertil Steril, 2013;99:15141522.Google Scholar
Shapira, M, Raanani, H, Barshack, I et al. First delivery in a leukemia survivor after transplantation of cryopreserved ovarian tissue, evaluated for leukemia cells contamination. Fertil Steril, 2018;109:4853.CrossRefGoogle Scholar
Committee, IP, Kim, SS, Donnez, J et al. Recommendations for fertility preservation in patients with lymphoma, leukemia, and breast cancer. J Assist Reprod Genet, 2012;29:465468.Google Scholar
Robertson, JA. Ethical issues in ovarian transplantation and donation. Fertil Steril, 2000;73:443446.Google Scholar
Blumenfeld, Z, Evron, A. Endocrine prevention of chemotherapy-induced ovarian failure. Curr Opin Obstet Gynecol, 2016;28:223229.Google Scholar
Del Mastro, L, Ceppi, M, Poggio, F et al. Gonadotropin-releasing hormone analogues for the prevention of chemotherapy-induced premature ovarian failure in cancer women: systematic review and meta-analysis of randomized trials. Cancer Treat Rev, 2014;40:675683.Google Scholar
Srkalovic, G, Schally, AV, Wittliff, JL et al. Presence and characteristics of receptors for [D-Trp6] luteinizing hormone releasing hormone and epidermal growth factor in human ovarian cancer. Int J Oncol, 1998;12:489498.Google ScholarPubMed
Emous, G, Grndker, C, Ganthert, AR et al. GnRH antagonists in the treatment of gynaecological and breast cancer. Endocr Relat Cancer, 2003;10:291299.Google Scholar
Blumenfeld, Z, Shapiro, D, Shteinberg, M et al. Preservation of fertility and ovarian function and minimizing gonadotoxicity in young women with systemic lupus erythematosus treated by chemotherapy. Lupus, 2000;9:401405.Google Scholar
Gougeon, A. Regulation of ovarian follicular development in primates: facts and hypotheses. Endocr Rev, 1996;17:121155.Google Scholar
Chian, RC, Huang, JY, Gilbert, L et al. Obstetric outcomes following vitrification of in vitro and in vivo matured oocytes. Fertil Steril, 2009;91:23912398.Google Scholar
Cohen, Y, St-Onge-St-Hilaire, A, Tannus, S et al. Decreased pregnancy and live birth rates after vitrification of in vitro matured oocytes. J Assist Reprod Genet. 2018;35(9):1683–1689.CrossRefGoogle Scholar
Buckett, WM, Chian, RC, Dean, NL et al. Pregnancy loss in pregnancies conceived after in vitro oocyte maturation, conventional in vitro fertilization and intracytoplasmic sperm injection. Fertil Steril, 2008;90:546550.Google Scholar
Bisharah, M, Tulandi, T. Laparoscopic preservation of ovarian function: an underused procedure. Am J Obstet Gynecol, 2003;188:367370.CrossRefGoogle ScholarPubMed
Chen, Y, Sun, J, Zou, K et al. Treatment for lupus nephritis: an overview of systematic reviews and meta-analyses. Rheumatol Int, 2017;37:10891099.Google Scholar
Bernatsky, S, Clarke, A, Suissa, S. Hematologic malignant neoplasms after drug exposure in rheumatoid arthritis. Arch Intern Med, 2008;168:378381.Google Scholar
Langford, C. Wegener’s granulomatosis: current and upcoming therapies. Arthritis Res Ther, 2003;5:180191.CrossRefGoogle ScholarPubMed
Schroeder, JO, Euler, H, Loffler, H. Synchronization of plasmapheresis and pulse cyclophosphamide in severe systemic lupus erythematosus. Ann Intern Med, 1987;107:344346.Google Scholar
Manger, K, Wildt, L, Kalden, JR et al. Prevention of gonadal toxicity and preservation of gonadal function and fertility in young women with systemic lupus erythematosus treated by cyclophosphamide: the PREGO-Study. Autoimmun Rev, 2006;5:269272.Google Scholar
Buyon, JP, Petri, MA, Kim, MY et al. The effect of combined estrogen and progesterone hormone replacement therapy on disease activity in systemic lupus erythematosus: a randomized trial. Ann Intern Med, 2005;142:953962.Google Scholar
Oktay, K, Hourvitz, A, Sahin, G et al. Letrozole reduces estrogen and gonadotropin exposure in women with breast cancer undergoing ovarian stimulation before chemotherapy. J Clin Endocrinol Metabol, 2006;91:38853890.CrossRefGoogle ScholarPubMed
Ahlstrom, A, Westin, C, Wikland, M et al. Prediction of live birth in frozen-thawed single blastocyst transfer cycles by pre-freeze and post-thaw morphology. Hum Reprod, 2013;28:11991209.Google Scholar
Goldman, RH, Racowsky, C, Farland, LV et al. Predicting the likelihood of live birth for elective oocyte cryopreservation: a counseling tool for physicians and patients. Hum Reprod, 2017;32:853859.CrossRefGoogle ScholarPubMed
Dondorp, WJ, De Wert, GM. Fertility preservation for healthy women: ethical aspects. Hum Reprod, 2009;24:17791785.CrossRefGoogle ScholarPubMed
Cobo, A, Garcia-Velasco, JA, Coello, A et al. Oocyte vitrification as an efficient option for elective fertility preservation. Fertil Steril, 2016;105:755–764.e8.Google Scholar
Cobo A, García-Velasco JA, Domingo J, Pellicer A, Remohí J. Elective and Onco-fertility preservation: factors related to IVF outcomes. Hum Reprod, 2018;33(12):2222–2231.Google Scholar
Somigliana, E, Ragni, G, Benedetti, F et al. Does laparoscopic excision of endometriotic ovarian cysts significantly affect ovarian reserve? Insights from IVF cycles. Hum Reprod, 2003;18:24502453.CrossRefGoogle ScholarPubMed
Carrillo, L, Seidman, D, Cittadini, E et al. The role of fertility preservation in patients with endometriosis. J Assist Reprod Genet, 2016;33:317323.CrossRefGoogle ScholarPubMed
García-Velasco, JA, Somigliana, E. Management of endometriomas in women requiring IVF: to touch or not to touch. Hum Reprod, 2009;24:496501.CrossRefGoogle ScholarPubMed
Busacca, M, Vignali, M. Endometrioma excision and ovarian reserve: a dangerous relation. J Minim Invasive Gynecol, 2009;16:142148.Google Scholar
Rauck, AM, Grouas, AC. Bone marrow transplantation in adolescents. Adolesc Med, 1999;10:445449.Google Scholar
Lau, NM, Huang, JY, MacDonald, S et al. Feasibility of fertility preservation in young females with Turner syndrome. Reprod Biomed Online, 2009;18:290295.Google Scholar
Pienkowski, C, Menendez, M, Cartault, A et al. Turner’s syndrome and procreation. Ovarian function and Turner’s syndrome. Gynecol Obstet Fertil, 2008;36:10301034.Google Scholar
Sénéchal, C, Rousset-Jablonski, C. Should a systematic fertility preservation be proposed to healthy women carrying a BRCA1/2 mutation? Gynecol Obstet Fertil, 2015;43:800805.Google Scholar
Derks-Smeets, IAP, van Tilborg, TC, van Montfoort, A et al. BRCA1 mutation carriers have a lower number of mature oocytes after ovarian stimulation for IVF/PGD. J Assist Reprod Genet, 2017;34:14751482.CrossRefGoogle ScholarPubMed
Cobo, A, Bellver, J, Domingo, J et al. New options in assisted reproduction technology: the Cryotop method of oocyte vitrification. Reprod Biomed Online, 2008;17:6872.Google Scholar
Bosch, E, Labarta, E, Crespo, J et al. Circulating progesterone levels and ongoing pregnancy rates in controlled ovarian stimulation cycles for in vitro fertilization: analysis of over 4000 cycles. Hum Reprod, 2010;25:20922100.Google Scholar

References

Wheeler, JM. Epidemiology of endometriosis-associated infertility. J Reprod Med, 1989;34(1):4146.Google Scholar
Giudice, LC, Kao, LC. Endometriosis. Lancet, 2004;364(9447):17891799.Google Scholar
Rogers, PA, D’Hooghe, TM, Fazleabas, A et al. Priorities for endometriosis research: recommendations from an international consensus workshop. Reprod Sci, 2009;16(4):335346.Google Scholar
Nisolle, M, Donnez, J. Peritoneal endometriosis, ovarian endometriosis, and adenomyotic nodules of the rectovaginal septum are three different entities. Fertil Steril, 1997;68(4):585596.Google Scholar
Seyhan, A, Ata, B, Uncu, G. The impact of endometriosis and its treatment on ovarian reserve. Semin Reprod Med, 2015;33(6):422428.Google Scholar
Giudice, LC. Clinical practice. Endometriosis. N Engl J Med, 2010;362(25):23892398.Google Scholar
Sanchez, AM, Vigano, P, Somigliana, E et al. The distinguishing cellular and molecular features of the endometriotic ovarian cyst: from pathophysiology to the potential endometrioma-mediated damage to the ovary. Hum Reprod Update, 2014;20(2):217230.Google Scholar
Kitajima, M, Defrere, S, Dolmans, MM et al. Endometriomas as a possible cause of reduced ovarian reserve in women with endometriosis. Fertil Steril, 2011;96(3):685691.CrossRefGoogle ScholarPubMed
Vercellini, P, Chapron, C, De Giorgi, O et al. Coagulation or excision of ovarian endometriomas? Am J Obstet Gynecol, 2003;188(3):606610.Google Scholar
Donnez, J, Chantraine, F, Nisolle, M. The efficacy of medical and surgical treatment of endometriosis-associated infertility: arguments in favour of a medico-surgical approach. Hum Reprod Update, 2002;8(1):8994.CrossRefGoogle Scholar
Exacoustos, C, Zupi, E, Amadio, A et al. Laparoscopic removal of endometriomas: sonographic evaluation of residual functioning ovarian tissue. Am J Obstet Gynecol, 2004;191(1):6872.Google Scholar
Muzii, L, Bellati, F, Bianchi, et al. Laparoscopic stripping of endometriomas: a randomized trial on different surgical techniques. Part II: pathological results. Hum Reprod, 2005;20(7):19871992.CrossRefGoogle ScholarPubMed
Hart, RJ, Hickey, M, Maouris, P, Buckett, W. Excisional surgery versus ablative surgery for ovarian endometriomata. Cochrane Database Syst Rev, 2008;2:CD004992.Google Scholar
Raffi, F, Metwally, M, Amer, S. The impact of excision of ovarian endometrioma on ovarian reserve: a systematic review and meta-analysis. J Clin Endocrinol Metab, 2012;97(9):31463154.Google Scholar
Meenakshi, M, McCluggage, WG. Vascular involvement in adenomyosis: report of a large series of a common phenomenon with observations on the pathogenesis of adenomyosis. Int J Gynecol Pathol, 2010;29(2):117121.Google Scholar
Donnez, J, Dolmans, MM. The ovary: from conception to death. Fertil Steril, 2017;108(4):594595.Google Scholar
Wallace, WH, Kelsey, TW. Human ovarian reserve from conception to the menopause. PLoS One, 2010;5(1):e8772.Google Scholar
Schubert, B, Canis, M, Darcha, C et al. Human ovarian tissue from cortex surrounding benign cysts: a model to study ovarian tissue cryopreservation. Hum Reprod, 2005;20(7):17861792.Google Scholar
Maneschi, F, Marasa, L, Incandela, S, Mazzarese, M, Zupi, E. Ovarian cortex surrounding benign neoplasms: a histologic study. Am J Obstet Gynecol, 1993; 169 (2 Pt 1): 388393.Google Scholar
Van Langendonckt, A, Donnez, J, Defrere, S, Dunselman, GA, Groothuis, PG. Antiangiogenic and vascular-disrupting agents in endometriosis: pitfalls and promises. Mol Hum Reprod, 2008;14(5):259268.CrossRefGoogle ScholarPubMed
Oktem, O, Urman, B. Understanding follicle growth in vivo. Hum Reprod, 2010;25(12):29442954.CrossRefGoogle ScholarPubMed
Skinner, MK. Regulation of primordial follicle assembly and development. Hum Reprod Update, 2005;11(5):461471.Google Scholar
Motta, PM, Nottola, SA, Familiari, G et al. Morphodynamics of the follicular-luteal complex during early ovarian development and reproductive life. Int Rev Cytol, 2003;223:177288.Google Scholar
Kitajima, M, Dolmans, MM, Donnez, O et al. Enhanced follicular recruitment and atresia in cortex derived from ovaries with endometriomas. Fertil Steril, 2014;101(4):10311037.CrossRefGoogle ScholarPubMed
Sanchez, AM, Papaleo, E, Corti, L et al. Iron availability is increased in individual human ovarian follicles in close proximity to an endometrioma compared with distal ones. Hum Reprod, 2014;29(3):577583.Google Scholar
Ferrero, S, Barra, F, Maggiore, ULR. Current and emerging therapeutics for the management of endometriosis. Drugs, 2018;78(10):9951012.Google Scholar
Dolmans, MM, Martinez-Madrid, B, Gadisseux, E et al. Short-term transplantation of isolated human ovarian follicles and cortical tissue into nude mice. Reproduction, 2007;134(2):253262.Google Scholar
Donnez, J, Lousse, JC, Jadoul, P, Donnez, O, Squifflet, J. Laparoscopic management of endometriomas using a combined technique of excisional (cystectomy) and ablative surgery. Fertil Steril, 2010;94(1):2832.Google Scholar
Working group of Esge, E, Wes, , Saridogan, E, Becker, CM et al. Recommendations for the surgical treatment of endometriosis-part 1: ovarian endometrioma. Gynecol Surg, 2017;14(1):27.Google Scholar
Donnez, J, Garcia-Solares, J, Dolmans, MM. Ovarian endometriosis and fertility preservation: a challenge in 2018. Minerva Ginecol, 2018;70(4):408414.Google Scholar
Donnez, J, Wyns, C, Nisolle, M. Does ovarian surgery for endometriomas impair the ovarian response to gonadotropin? Fertil Steril, 2001;76(4):662665.Google Scholar
Carmona, F, Martinez-Zamora, MA, Rabanal, A, Martinez-Roman, S, Balasch, J. Ovarian cystectomy versus laser vaporization in the treatment of ovarian endometriomas: a randomized clinical trial with a five-year follow-up. Fertil Steril, 2011;96(1):251254.Google Scholar
Dunselman, GA, Vermeulen, N, Becker, C et al. ESHRE guideline: management of women with endometriosis. Hum Reprod, 2014;29(3):400412.CrossRefGoogle ScholarPubMed
Falcone, T, Flyckt, R. Clinical management of endometriosis. Obstet Gynecol, 2018;131(3):557571.Google Scholar
Carrillo, L, Seidman, DS, Cittadini, E, Meirow, D. The role of fertility preservation in patients with endometriosis. J Assist Reprod Genet, 2016;33(3):317323.Google Scholar
Hirokawa, W, Iwase, A, Goto, M et al. The post-operative decline in serum anti-Mullerian hormone correlates with the bilaterality and severity of endometriosis. Hum Reprod, 2011;26(4):904910.Google Scholar
Aimetti, M, Manavella, V, Corano, L et al. Three-dimensional analysis of bone remodeling following ridge augmentation of compromised extraction sockets in periodontitis patients: A randomized controlled study. Clin Oral Implants Res, 2018;29(2):202214.Google Scholar
Roman, H. Endometriosis surgery and preservation of fertility, what surgeons should know. J Visc Surg, 2018;155(Suppl 1):S31S36.Google Scholar
Donnez, J. Reply of the authors to I. Brosens. Fertil Steril, 2010;94:e74.Google Scholar
Garavaglia, E, Sala, C, Taccagni, G et al. Fertility preservation in endometriosis patients: Anti-Mullerian hormone is a reliable marker of the ovarian follicle density. Front Surg, 2017;4:40.Google Scholar
Anderson, RA, Wallace, WH. Antimullerian hormone, the assessment of the ovarian reserve, and the reproductive outcome of the young patient with cancer. Fertil Steril, 2013;99(6):14691475.Google Scholar
Muzii, L, Luciano, AA, Zupi, E, Panici, PB. Effect of surgery for endometrioma on ovarian function: a different point of view. J Minim Invasive Gynecol, 2014;21(4):531533.Google Scholar
Shah, DK, Mejia, RB, Lebovic, DI. Effect of surgery for endometrioma on ovarian function. J Minim Invasive Gynecol, 2014;21(2):203209.Google Scholar
Practice Committee of the American Society for Reproductive M. Testing and interpreting measures of ovarian reserve: a committee opinion. Fertil Steril, 2012;98(6):14071415.Google Scholar
Somigliana, E, Berlanda, N, Benaglia, L et al. Surgical excision of endometriomas and ovarian reserve: a systematic review on serum antimullerian hormone level modifications. Fertil Steril, 2012;98(6):15311538.CrossRefGoogle ScholarPubMed
Goodman, LR, Goldberg, JM, Flyckt, RL et al. Effect of surgery on ovarian reserve in women with endometriomas, endometriosis and controls. Am J Obstet Gynecol, 2016;215(5):589 e1e6.Google Scholar
Turkcuoglu, I, Melekoglu, R. The long-term effects of endometrioma surgery on ovarian reserve: a prospective case-control study. Gynecol Endocrinol, 2018;34(7):612615.Google Scholar
Collinet, P, Fritel, X, Revel-Delhom, C et al. Management of endometriosis: CNGOF/HAS clinical practice guidelines – Short version. J Gynecol Obstet Hum Reprod, 2018;47(7):265274.Google Scholar
Dolmans, MM, Donnez, J. Indications for fertility preservation in women from malignant diseases to benign conditions to age-related fertility decline. Minerva Ginecol, 2018;70(4):402407.Google Scholar
Chen, C. Pregnancy after human oocyte cryopreservation. Lancet, 1986;1(8486):884886.Google Scholar
Cobo, A, Diaz, C. Clinical application of oocyte vitrification: a systematic review and meta-analysis of randomized controlled trials. Fertil Steril, 2011;96(2):277285.Google Scholar
Argyle, CE, Harper, JC, Davies, MC. Oocyte cryopreservation: where are we now? Hum Reprod Update, 2016;22(4):440449.Google Scholar
Levi-Setti, PE, Patrizio, P, Scaravelli, G. Evolution of human oocyte cryopreservation: slow freezing versus vitrification. Curr Opin Endocrinol Diabetes Obes, 2016;23(6):445450.Google Scholar
Rienzi, L, Gracia, C, Maggiulli, R et al. Oocyte, embryo and blastocyst cryopreservation in ART: systematic review and meta-analysis comparing slow-freezing versus vitrification to produce evidence for the development of global guidance. Hum Reprod Update, 2017;23(2):139155.Google Scholar
Cobo, A, Garcia-Velasco, JA, Coello, A et al. Oocyte vitrification as an efficient option for elective fertility preservation. Fertil Steril, 2016;105(3):755–764 e8.Google Scholar
Donnez, J, Dolmans, MM. Fertility preservation in women. N Engl J Med, 2017;377(17):16571665.Google Scholar
Donnez, J, Dolmans, MM. Fertility preservation in women. Nat Rev Endocrinol, 2013;9(12):735749.Google Scholar
Bedoschi, G, Oktay, K. Current approach to fertility preservation by embryo cryopreservation. Fertil Steril, 2013;99(6):14961502.Google Scholar
Donnez, J, Nisolle, M, Gillet, N et al. Large ovarian endometriomas. Hum Reprod, 1996;11(3):641646.Google Scholar
Practice Committee of American Society for Reproductive Medicine. Ovarian tissue cryopreservation: a committee opinion. Fertil Steril, 2014;101(5):12371243.Google Scholar
Practice Committee of the American Society for Reproductive Medicine. Endometriosis and infertility: a committee opinion. Fertil Steril, 2012;98(3):591598.Google Scholar
Cil, AP, Bang, H, Oktay, K. Age-specific probability of live birth with oocyte cryopreservation: an individual patient data meta-analysis. Fertil Steril, 2013;100(2):492–499 e3.Google 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(1):5863.Google Scholar
Silber, SJ, Grudzinskas, G, Gosden, RG. Successful pregnancy after microsurgical transplantation of an intact ovary. N Engl J Med, 2008;359(24):26172618.Google Scholar
Silber, SJ, DeRosa, M, Pineda, J et al. A series of monozygotic twins discordant for ovarian failure: ovary transplantation (cortical versus microvascular) and cryopreservation. Hum Reprod, 2008;23(7):15311537.Google Scholar
Silber, SJ, Gosden, RG. Ovarian transplantation in a series of monozygotic twins discordant for ovarian failure. N Engl J Med, 2007;356(13):13821384.Google Scholar
Donnez, J, Squifflet, J, Dolmans, MM et al. Orthotopic transplantation of fresh ovarian cortex: a report of two cases. Fertil Steril, 2005;84(4):1018.Google Scholar
Donnez, J, Martinez-Madrid, B, Jadoul, P et al. Ovarian tissue cryopreservation and transplantation: a review. Hum Reprod Update, 2006;12(5):519535.Google Scholar
Donnez, J, Dolmans, MM, Demylle, D et al. Livebirth after orthotopic transplantation of cryopreserved ovarian tissue. Lancet, 2004;364(9443):14051410.Google Scholar
Gellert, SE, Pors, SE, Kristensen, SG et al. 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;35(4):561570.Google Scholar
Ernst, E, Bergholdt, S, Jorgensen, JS, Andersen, CY. The first woman to give birth to two children following transplantation of frozen/thawed ovarian tissue. Hum Reprod, 2010;25(5):12801281.Google Scholar
Donnez, J, Dolmans, MM, Pellicer, A et al. Restoration of ovarian activity and pregnancy after transplantation of cryopreserved ovarian tissue: a review of 60 cases of reimplantation. Fertil Steril, 2013;99(6):15031513.Google Scholar
Jadoul, P, Guilmain, A, Squifflet, J et al. Efficacy of ovarian tissue cryopreservation for fertility preservation: lessons learned from 545 cases. Hum Reprod, 2017;32(5):10461054.Google Scholar
Meirow, D, Ra’anani, H, Shapira, M et al. Transplantations of frozen-thawed ovarian tissue demonstrate high reproductive performance and the need to revise restrictive criteria. Fertil Steril, 2016;106(2):467474.Google Scholar
Jensen, AK, Macklon, KT, Fedder, J et al. 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;34(3):325336.Google Scholar
Van der Ven, H, Liebenthron, J, Beckmann, M et al. 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;31(9):20312041.CrossRefGoogle Scholar
Demeestere, I, Simon, P, Emiliani, S, Delbaere, A, Englert, Y. Fertility preservation: successful transplantation of cryopreserved ovarian tissue in a young patient previously treated for Hodgkin’s disease. Oncologist, 2007;12(12):14371442.Google Scholar
Schmidt, KT, Nyboe Andersen, A, Greve, T. Fertility in cancer patients after cryopreservation of one ovary. Reprod Biomed Online, 2013;26(3):272279.Google 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.Google Scholar
Donnez, J, Dolmans, MM, Diaz, C, Pellicer, A. Ovarian cortex transplantation: time to move on from experimental studies to open clinical application. Fertil Steril, 2015;104(5):10971098.Google Scholar
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;32(8):11671170.Google Scholar
Donnez, J, Squifflet, J, Jadoul, P et al. Fertility preservation in women with ovarian endometriosis. Front Biosci (Elite Ed), 2012;4:16541662.Google Scholar
Dittrich, R, Lotz, L, Keck, G et al. Live birth after ovarian tissue autotransplantation following overnight transportation before cryopreservation. Fertil Steril, 2012;97(2):387390.Google Scholar

References

Noone, AM, Howlader, N, Noone, AM et al. SEER cancer statistics review, 1975–2015. National Cancer Institute 2018.Google Scholar
Phillips, SM, Padgett, LS, Leisenring, WM et al. Survivors of childhood cancer in the United States: prevalence and burden of morbidity. Cancer Epidemiol Biomarkers Prev, 2015;24:653663.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
Hudson, MM, Ness, KK, Gurney, JG et al. Clinical ascertainment of health outcomes among adults treated for childhood cancer. JAMA, 2013;309:23712381.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
Knapp, CA, Quinn, GP, Murphy, D. Assessing the reproductive concerns of children and adolescents with cancer: Challenges and potential solutions. J Adolesc Young Adult Oncol, 2011;1:3135.Google Scholar
Quinn, GP, Woodruff, TK, Knapp, CA. Expanding the oncofertility workforce: training allied health professionals to improve health outcomes for adolescents and young adults. J Adolesc Young Adult Oncol, 2016;5:292296.Google Scholar
Carlson, CA, Kolon, TF, Mattei, P. Developing a hospital-wide fertility reservation service for pediatric and young adult patients. J Adolesc Health, 2017;61:571576.Google Scholar
Sklar, C. Reproductive physiology and treatment-related loss of sex hormone production. Med Pediatr Oncol, 1999;33:28.Google Scholar
Kubota, H, Brinster, RL. Spermatogonial stem cells. Biol Reprod, 2018;99(1):5274.Google Scholar
Howell, SJ, Shalet, SM. Spermatogenesis after cancer treatment: damage and recovery. J Natl Cancer Inst Monogr, 2005;34:1217.Google Scholar
Wallace, WH. Oncofertility and preservation of reproductive capacity in children and young adults. Cancer, 2011;117:23012310.Google Scholar
Meistrich, ML. Effects of chemotherapy and radiotherapy on spermatogenesis in humans. Fertil Steril, 2013;100:11801186.Google Scholar
van Casteren, NJ, van der Linden, GH, Hakvoort-Cammel, FG et al. Effect of childhood cancer treatment on fertility markers in adult male long-term survivors. Pediatr Blood Cancer, 2009;52:108112.Google Scholar
Holoch, P, Wald, M. Current options for preservation of fertility in the male. Fertil Steril, 2011;96:286290.Google Scholar
Pryzant, RM, Meistrich, ML, Wilson, G. Long-term reduction in sperm count after chemotherapy with and without radiation therapy for non-Hodgkin’s lymphomas. J Clin Oncol, 1993;11:239247.Google Scholar
Meistrich, ML, Wilson, G, Brown, BW, da Cunha, MF, Lipshultz, LI. Impact of cyclophosphamide on long-term reduction in sperm count in men treated with combination chemotherapy for Ewing and soft tissue sarcomas. Cancer, 1992;70:27032712.Google Scholar
Kenney, LB, Laufer, MR, Grant, FD, Grier, H, Diller, L. High risk of infertility and long term gonadal damage in males treated with high dose cyclophosphamide for sarcoma during childhood. Cancer, 2001;91:613621.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
Viviani, S, Santoro, A, Ragni, G et al. Gonadal toxicity after combination chemotherapy for Hodgkin’s disease. Comparative results of MOPP vs ABVD. Eur J Cancer Clin Oncol, 1985;21:601605.Google Scholar
Marmor, D, Duyck, F. Male reproductive potential after MOPP therapy for Hodgkin’s disease: a long-term survey. Andrologia, 1995;27:99106.Google Scholar
Dohle, GR. Male infertility in cancer patients: Review of the literature. Int J Urol, 2010;17:327331.Google Scholar
Paoli, D, Rizzo, F, Fiore, G et al. Spermatogenesis in Hodgkin’s lymphoma patients: a retrospective study of semen quality before and after different chemotherapy regimens. Hum Reprod, 2016;31:263272.Google Scholar
Behringer, K, Mueller, H, Goergen, H et al. Gonadal function and fertility in survivors after Hodgkin lymphoma treatment within the German Hodgkin Study Group HD13 to HD15 trials. J Clin Oncol, 2013;31:231239.Google Scholar
Waring, AB, Wallace, WH. Subfertility following treatment for childhood cancer. Hosp Med, 2000;61:550557.Google Scholar
Chovanec, M, Abu Zaid, M, Hanna, N et al. Long-term toxicity of cisplatin in germ-cell tumor survivors. Ann Oncol, 2017;28:26702679.Google Scholar
Pont, J, Albrecht, W. Fertility after chemotherapy for testicular germ cell cancer. Fertil Steril, 1997;68:15.Google Scholar
Hansen, PV, Hansen, SW. Gonadal function in men with testicular germ cell cancer: the influence of cisplatin-based chemotherapy. Eur Urol, 1993;23:153156.Google Scholar
Meistrich, M, Chawla, SP, da Cunha, MF et al. Recovery of sperm production after chemotherapy for osteosarcoma. Cancer, 1989;63:21152123.Google Scholar
Brignardello, E, Felicetti, F, Castiglione, A et al. Gonadal status in long-term male survivors of childhood cancer. J Cancer Res Clin Oncol, 2016;142:11271132.CrossRefGoogle ScholarPubMed
Longhi, A, Macchiagodena, M, Vitali, G, Bacci, G. Fertility in male patients treated with neoadjuvant chemotherapy for osteosarcoma. J Pediatr Hematol Oncol, 2003;25:292296.Google Scholar
Janeway, KA, Grier, HE. Sequelae of osteosarcoma medical therapy: a review of rare acute toxicities and late effects. Lancet Oncol, 2010;11:670678.Google Scholar
Williams, D, Crofton, PM, Levitt, G. Does ifosfamide affect gonadal function? Pediatr Blood Cancer, 2008;50:347351.Google Scholar
Jacob, A, Barker, H, Goodman, A, Holmes, J. Recovery of spermatogenesis following bone marrow transplantation. Bone Marrow Transplant, 1998;22:277279.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
Ash, P. The influence of radiation on fertility in man. Br J Radiol, 1980;53:271278.Google Scholar
Kenney, LB, Cohen, LE, Shnorhavorian, M et al. Male reproductive health after childhood, adolescent, and young adult cancers: a report from the Children’s Oncology Group. J Clin Oncol, 2012;30:34083416.Google Scholar
Blatt, J, Sherins, RJ, Niebrugge, D, Bleyer, WA, Poplack, DG. Leydig cell function in boys following treatment for testicular relapse of acute lymphoblastic leukemia. J Clin Oncol, 1985;3:12271231.Google Scholar
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;18:185189.Google Scholar
Sklar, CA, Robison, LL, Nesbit, ME et al. Effects of radiation on testicular function in long-term survivors of childhood acute lymphoblastic leukemia: a report from the Children Cancer Study Group. J Clin Oncol, 1990;8:19811987.Google Scholar
Shapiro, E, Kinsella, TJ, Makuch, RW et al. Effects of fractionated irradiation of endocrine aspects of testicular function. J Clin Oncol, 1985;3:12321239.Google Scholar
Fraass, BA, Kinsella, TJ, Harrington, FS, Glatstein, E. Peripheral dose to the testes: the design and clinical use of a practical and effective gonadal shield. Int J Radiat Oncol Biol Phys, 1985;11:609615.Google Scholar
Socie, G, Salooja, N, Cohen, A et al. Nonmalignant late effects after allogeneic stem cell transplantation. Blood, 2003;101:33733385.Google Scholar
Sayan, M, Cassidy, RJ, Butker, EE et al. Gonadal shielding technique to preserve fertility in male pediatric patients treated with total body irradiation for stem cell transplantation. Bone Marrow Transplant, 2016;51:997998.Google Scholar
Speiser, B, Rubin, P, Casarett, G. Aspermia following lower truncal irradiation in Hodgkin’s disease. Cancer, 1973;32:692698.Google Scholar
Sandeman, TF. The effects of x irradiation on male human fertility. Br J Radiol, 1966;39:901907.Google Scholar
Djaladat, H, Burner, E, Parikh, PM, Beroukhim Kay, D, Hays, K. The association between testis cancer and semen abnormalities before orchiectomy: a systematic review. J Adolesc Young Adult Oncol, 2014;3:153159.Google Scholar
Kenney, LB, Antal, Z, Ginsberg, JP et al. Improving male reproductive health after childhood, adolescent, and young adult cancer: progress and future directions for survivorship research. J Clin Oncol, 2018;36:21602168.Google Scholar
Crowne, E, Gleeson, H, Benghiat, H, Sanghera, P, Toogood, A. Effect of cancer treatment on hypothalamic-pituitary function. Lancet Diabetes Endocrinol, 2015;3:568576.Google Scholar
Darzy, KH, Shalet, SM. Hypopituitarism following radiotherapy revisited. Endocr Dev, 2009;15:124.Google Scholar
Schmiegelow, M, Lassen, S, Poulsen, HS et al. Gonadal status in male survivors following childhood brain tumors. J Clin Endocrinol Metab, 2001;86:24462452.Google Scholar
Constine, LS, Woolf, PD, Cann, D et al. Hypothalamic-pituitary dysfunction after radiation for brain tumors. N Engl J Med, 1993;328:8794.Google Scholar
Ogle, SK, Hobbie, WL, Carlson, CA et al. Sperm banking for adolescents with cancer. J Pediatr Oncol Nurs, 2008;25:97101.Google Scholar
Klosky, JL, Lehmann, V, Flynn, JS et al. Patient factors associated with sperm cryopreservation among at-risk adolescents newly diagnosed with cancer. Cancer, 2018;124:35673575.Google Scholar
Ginsberg, JP, Ogle, SK, Tuchman, LK et al. Sperm banking for adolescent and young adult cancer patients: sperm quality, patient, and parent perspectives. Pediatr Blood Cancer, 2008;50:594598.Google Scholar
DiNofia, AM, Wang, X, Yannekis, G et al. Analysis of semen parameters in a young cohort of cancer patients. Pediatr Blood Cancer, 2017;64:381386.Google Scholar
Yu, G, Liu, Y, Zhang, H, Wu, K. Application of testicular spermatozoa cryopreservation in assisted reproduction. Int J Gynaecol Obstet, 2018;142:354358.Google Scholar
Adank, MC, van Dorp, W, Smit, M et al. Electroejaculation as a method of fertility preservation in boys diagnosed with cancer: a single-center experience and review of the literature. Fertil Steril, 2014;102:199–205 e191.Google Scholar
Jurewicz, M, Hillelsohn, J, Mehta, S, Gilbert, BR. Fertility preservation in pubertal and pre-pubertal boys with cancer. Pediatr Endocrinol Rev, 2018;15:234243.Google Scholar
Brinster, RL. Male germline stem cells: from mice to men. Science, 2007;316:404405.Google Scholar
Hermann, BP, Sukhwani, M, Winkler, F et al. Spermatogonial stem cell transplantation into rhesus testes regenerates spermatogenesis producing functional sperm. Cell Stem Cell, 2012;11:715726.Google Scholar
Bhang, DH, Kim, BJ, Kim, BG et al. Testicular endothelial cells are a critical population in the germline stem cell niche. Nat Commun, 2018;9:4379.Google Scholar
Ginsberg, JP, Carlson, CA, Lin, K et al. An experimental protocol for fertility preservation in prepubertal boys recently diagnosed with cancer: a report of acceptability and safety. Hum Reprod, 2010;25:3741.Google Scholar
Richards, JS. Perspective: the ovarian follicle–a perspective in 2001. Endocrinology, 2001;142:21842193.Google Scholar
Banerjee, S, Banerjee, S, Saraswat, G, Bandyopadhyay, SA, Kabir, SN. Female reproductive aging is master-planned at the level of ovary. PLoS One, 2014;9: e96210.Google Scholar
Mattison, DR, Plowchalk, DR, Meadows, MJ et al. Reproductive toxicity: male and female reproductive systems as targets for chemical injury. Med Clin North Am, 1990;74:391411.Google Scholar
Gargus, E, Deans, R, Anazodo, A, Woodruff, TK. Management of primary ovarian insufficiency symptoms in survivors of childhood and adolescent cancer. J Natl Compr Canc Netw, 2018;16:11371149.Google Scholar
Levine, JM, Whitton, JA, Ginsberg, JP et al. Nonsurgical premature menopause and reproductive implications in survivors of childhood cancer: a report from the childhood cancer survivor study. Cancer, 2018;124:10441052.Google Scholar
Chemaitilly, W, Li, Z, Krasin, MJ et al. Premature ovarian insufficiency in childhood cancer survivors: a report from the St. Jude lifetime cohort. J Clin Endocrinol Metab, 2017;102:22422250.Google Scholar
Oktem, O, Kim, SS, Selek, U, Schatmann, G, Urman, B. Ovarian and uterine functions in female survivors of childhood cancers. Oncologist, 2018;23:214224.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
Chemaitilly, W, Mertens, AC, Mitby, P et al. Acute ovarian failure in the childhood cancer survivor study. J Clin Endocrinol Metab, 2006;91:17231728.Google Scholar
Gracia, CR, Ginsberg, JP. Fertility risk in pediatric and adolescent cancers. Cancer Treat Res, 2007;138:5772.Google Scholar
Thomas-Teinturier, C, Allodji, RS, Svetlova, E et al. Ovarian reserve after treatment with alkylating agents during childhood. Hum Reprod, 2015;30:14371446.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
De Bruin, ML, Huisbrink, J, Hauptmann, M et al. Treatment-related risk factors for premature menopause following Hodgkin lymphoma. Blood, 2008;111:101108.Google Scholar
Sklar, CA, Mertens, AC, Mitby, P et al. Premature menopause in survivors of childhood cancer: a report from the childhood cancer survivor study. J Natl Cancer Inst, 2006;98:890896.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;36:21692180.Google Scholar
Overbeek, A, van den Berg, MH, van Leeuwen, FE et al. Chemotherapy-related late adverse effects on ovarian function in female survivors of childhood and young adult cancer: a systematic review. Cancer Treat Rev, 2017;53:1024.Google Scholar
Borgmann-Staudt, A, Rendtorff, R, Reinmuth, S et al. Fertility after allogeneic haematopoietic stem cell transplantation in childhood and adolescence. Bone Marrow Transplant, 2012;47:271276.Google Scholar
Thibaud, E, Rodriguez-Macias, K, Trivin, C et al. Ovarian function after bone marrow transplantation during childhood. Bone Marrow Transplant, 1998;21:287290.Google Scholar
Sanders, JE, Woolfrey, AE, Carpenter, PA et al. Late effects among pediatric patients followed for nearly 4 decades after transplantation for severe aplastic anemia. Blood, 2011;118:14211428.Google Scholar
Vatanen, A, Wilhelmsson, M, Borgstrom, B et al. Ovarian function after allogeneic hematopoietic stem cell transplantation in childhood and adolescence. Eur J Endocrinol, 2014;170:211218.Google Scholar
Nabhan, SK, Bitencourt, MA, Duval, M et al. Fertility recovery and pregnancy after allogeneic hematopoietic stem cell transplantation in Fanconi anemia patients. Haematologica, 2010;95:17831787.Google Scholar
Singhal, S, Powles, R, Treleaven, J et al. Melphalan alone prior to allogeneic bone marrow transplantation from HLA-identical sibling donors for hematologic malignancies: alloengraftment with potential preservation of fertility in women. Bone Marrow Transplant, 1996;18:10491055.Google Scholar
Panasiuk, A, Nussey, S, Veys, P et al. Gonadal function and fertility after stem cell transplantation in childhood: comparison of a reduced intensity conditioning regimen containing melphalan with a myeloablative regimen containing busulfan. Br J Haematol, 2015;170:719726.Google Scholar
Wallace, WH, Thomson, AB, Kelsey, TW. The radiosensitivity of the human oocyte. Hum Reprod, 2003;18:117121.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
Wallace, WH, Shalet, SM, Hendry, JH, Morris-Jones, PH, Gattamaneni, HR. Ovarian failure following abdominal irradiation in childhood: the radiosensitivity of the human oocyte. Br J Radiol, 1989;62:995998.Google Scholar
Beneventi, F, Locatelli, E, Giorgiani, G et al. Gonadal and uterine function in female survivors treated by chemotherapy, radiotherapy, and/or bone marrow transplantation for childhood malignant and non-malignant diseases. BJOG, 2014;121:856865; discussion 865.Google Scholar
Teh, WT, Stern, C, Chander, S, Hickey, M. The impact of uterine radiation on subsequent fertility and pregnancy outcomes. Biomed Res Int, 2014;2014:482968.Google Scholar
Koustenis, E, Pfitzer, C, Balcerek, M et al. Impact of cranial irradiation and brain tumor location on fertility: a survey. Klin Padiatr, 2013;225:320324.Google 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, 1927 e1.Google Scholar
Hamre, MR, Robison, LL, Nesbit, ME et al. Effects of radiation on ovarian function in long-term survivors of childhood acute lymphoblastic leukemia: a report from the Children’s Cancer Study Group. J Clin Oncol, 1987;5:17591765.Google Scholar
Wallace, WH, Shalet, SM, Tetlow, LJ, Morris-Jones, PH. Ovarian function following the treatment of childhood acute lymphoblastic leukaemia. Med Pediatr Oncol, 1993;21:333339.Google Scholar
DeWire, M, Green, DM, Sklar, CA et al. Pubertal development and primary ovarian insufficiency in female survivors of embryonal brain tumors following risk-adapted craniospinal irradiation and adjuvant chemotherapy. Pediatr Blood Cancer, 2015;62:329334.Google Scholar
Lester-Coll, NH, Morse, CB, Zhai, HA et al. Preserving fertility in adolescent girls and young women requiring craniospinal irradiation: a case report and discussion of options to be considered prior to treatment. J Adolesc Young Adult Oncol, 2014;3:9699.Google Scholar
Perez-Andujar, A, Newhauser, WD, Taddei, PJ, Mahajan, A, Howell, RM. The predicted relative risk of premature ovarian failure for three radiotherapy modalities in a girl receiving craniospinal irradiation. Phys Med Biol, 2013;58:31073123.Google Scholar
Jadoul, P, Anckaert, E, Dewandeleer, A et al. Clinical and biologic evaluation of ovarian function in women treated by bone marrow transplantation for various indications during childhood or adolescence. Fertil Steril, 2011;96(1):126133.Google Scholar
Sarafoglou, K, Boulad, F, Gillio, A, Sklar, C. Gonadal function after bone marrow transplantation for acute leukemia during childhood. J Pediatr, 1997;130:210216.Google Scholar
Jadoul, P, Donnez, J. How does bone marrow transplantation affect ovarian function and fertility? Curr Opin Obstet Gynecol, 2012;24:164171.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
Lee, SJ, Schover, LR, Partridge, AH et al. American Society of Clinical Oncology recommendations on fertility preservation in cancer patients. J Clin Oncol, 2006;24:29172931.Google Scholar
Oktay, K, Harvey, BE, Partridge, AH et al. Fertility preservation in patients with cancer: ASCO clinical practice guideline update. J Clin Oncol, 2018;36:19942001.Google Scholar
Druckenmiller, S, Goldman, KN, Labella, PA et al. Successful oocyte cryopreservation in reproductive-aged cancer survivors. Obstet Gynecol, 2016;127:474480.Google Scholar
Rienzi, L, Gracia, C, Maggiulli, R et al. Oocyte, embryo and blastocyst cryopreservation in ART: systematic review and meta-analysis comparing slow-freezing versus vitrification to produce evidence for the development of global guidance. Hum Reprod Update, 2017;23:139155.Google Scholar
Argyle, CE, Harper, JC, Davies, MC. Oocyte cryopreservation: where are we now? Hum Reprod Update, 2016;22:440449.Google Scholar
Donnez, J, Dolmans, MM, Diaz, C, Pellicer, A. Ovarian cortex transplantation: time to move on from experimental studies to open clinical application. Fertil Steril, 2015;104:10971098.Google Scholar
Pacheco, F, Oktay, K. Current success and efficiency of autologous ovarian transplantation: a meta-analysis. Reprod Sci, 2017;24:11111120.Google Scholar
Van der Ven, H, Liebenthron, J, Beckmann, M et al. 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;31:20312041.Google Scholar
Flyckt, R. Ovarian cortical biopsy for fertility preservation. 2019 Oncofertility Conference, Chicago, Illinois. Presented November 13, 2019.Google Scholar
Xiao, S, Zhang, J, Romero, MM et al. In vitro follicle growth supports human oocyte meiotic maturation. Sci Rep, 2015;5:17323.Google Scholar
Luyckx, V, Dolmans, MM, Vanacker, J et al. A new step toward the artificial ovary: survival and proliferation of isolated murine follicles after autologous transplantation in a fibrin scaffold. Fertil Steril, 2014;101:11491156.Google Scholar
Prasath, EB, Chan, ML, Wong, WH et al. First pregnancy and live birth resulting from cryopreserved embryos obtained from in vitro matured oocytes after oophorectomy in an ovarian cancer patient. Hum Reprod, 2014;29:276278.Google Scholar
Segers, I, Mateizel, I, Van Moer, E et al. In vitro maturation (IVM) of oocytes recovered from ovariectomy specimens in the laboratory: a promising “ex vivo” method of oocyte cryopreservation resulting in the first report of an ongoing pregnancy in Europe. J Assist Reprod Genet, 2015;32:12211231.Google Scholar
Moawad, NS, Santamaria, E, Rhoton-Vlasak, A, Lightsey, JL. Laparoscopic ovarian transposition before pelvic cancer treatment: ovarian function and fertility preservation. J Minim Invasive Gynecol, 2017;24:2835.Google Scholar
Fernandez-Pineda, I, Davidoff, AM, Lu, L et al. Impact of ovarian transposition before pelvic irradiation on ovarian function among long-term survivors of childhood Hodgkin lymphoma: A report from the St. Jude Lifetime Cohort Study. Pediatr Blood Cancer, 2018;65: e27232.Google Scholar
Oktay, K, Oktem, O. Fertility preservation medicine: a new field in the care of young cancer survivors. Pediatr Blood Cancer, 2009;53:267273.Google Scholar
Smith, KL, Gracia, C, Sokalska, A, Moore, H. Advances in fertility preservation for young women with cancer. Am Soc Clin Oncol Educ Book, 2018;38:2737.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 embrya, oocytes, or ovaries. Oncologist, 2007;12:10441054.Google Scholar
Blumenfeld, Z, Zur, H, Dann, EJ. Gonadotropin-releasing hormone agonist cotreatment during chemotherapy may increase pregnancy rate in survivors. Oncologist, 2015;20:12831289.Google Scholar
Practice Committees of American Society for Reproductive Medicine SfART. Mature oocyte cryopreservation: a guideline. Fertil Steril, 2013;99:3743.Google Scholar
Nightingale, CL, Quinn, GP, Shenkman, EA et al. Health-related quality of life of young adult survivors of childhood cancer: a review of qualitative studies. J Adolesc Young Adult Oncol, 2011;1:124132.Google Scholar
Ginsberg, JP, Li, Y, Carlson, CA et al. Testicular tissue cryopreservation in prepubertal male children: an analysis of parental decision-making. Pediatr Blood Cancer, 2014;61:16731678.Google Scholar
Sullivan-Pyke, CS, Carlson, CA, Prewitt, M, Gracia, CR, Ginsberg, JP. Ovarian tissue cryopreservation (OTC) in prepubertal girls and young women: an analysis of parents’ and patients’ decision-making. J Assist Reprod Genet, 2018;35:593600.Google Scholar
Nahata, L, Caltabellotta, NM, Yeager, ND et al. Fertility perspectives and priorities among male adolescents and young adults in cancer survivorship. Pediatr Blood Cancer, 2018;65: e27019.Google Scholar
Overbeek, A, van den Berg, M, Louwe, L et al. Practice, attitude and knowledge of Dutch paediatric oncologists regarding female fertility. Neth J Med, 2014;72:264270.Google Scholar
Ataman, LM, Rodrigues, JK, Marinho, RM et al. Creating a global community of practice for oncofertility. J Glob Oncol, 2016;2:8396.Google Scholar

References

Fitzmaurice, C, Akinyemiju, TF, Al Lami, FH et al. for the Global Burden of Disease Cancer Collaboration. Global, Regional, and National Cancer Incidence, Mortality, Years of Life Lost, Years Lived With Disability, and Disability-Adjusted Life-Years for 29 Cancer Groups, 1990 to 2016: A Systematic Analysis for the Global Burden of Disease Study. JAMA Oncol, 2018 June 2. DOI:10.1001/jamaoncol.2018.2706. [Epub ahead of print]Google Scholar
Engholm, G, Ferlay, J, Christensen, N et al. NORDCAN: Cancer Incidence, Mortality, Prevalence and Survival in the Nordic Countries. Association of the Nordic Cancer Registries. Danish Cancer Society 2014.Google Scholar
Surveillance, Epidemiology, and End Results (SEER) 5-year relative survival by site. SEER cancer statistics review 1975–2008. www.seer.cancer.govGoogle Scholar
Oktay, K, Harvey, BE, Partridge, AH et al. Fertility preservation in patients with cancer: ASCO clinical practice guideline update. J Clin Oncol, 2018 July 1;36(19):19942001.Google Scholar
Armuand, GM, Rodriguez-Wallberg, KA, Wettergren, L et al. Sex differences in fertility-related information received by young adult cancer survivors. J Clin Oncol, 2012;30(17):21472153.Google Scholar
Armuand, GM, Wettergren, L, Rodriguez-Wallberg, KA, Lampic, C. Women more vulnerable than men when facing risk for treatment-induced infertility: a qualitative study of young adults newly diagnosed with cancer. Acta Oncol, 2015 February;54(2):243552.Google Scholar
Fredholm, H, Eaker, S, Frisell, J et al. Breast cancer in young women: poor survival despite intensive treatment. PLoS One, 2009;4:e76957.Google Scholar
Curigliano, G, Burstein, HJ, Winer, EP et al.; Panel Members of the St. Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2017. Ann Oncol, 2018 May 3. DOI:10.1093/annonc/mdx806.[Epub ahead of print]Google Scholar
Rodriguez-Wallberg, KA. Principles of cancer treatment: impact on reproduction. Adv Exp Med Biol, 2012;732:18.Google Scholar
Sonmezer, M, Oktay, K. Fertility preservation in young women undergoing breast cancer therapy. Oncologist, 2006;11(5):422434.Google Scholar
Rodriguez-Wallberg, KA, Oktay, K. Fertility preservation and pregnancy in women with and without BRCA mutation-positive breast cancer. Oncologist, 2012;17:14091417.Google Scholar
Ethics Committee of the American Society for Reproductive Medicine. Fertility preservation and reproduction in patients facing gonadotoxic therapies: an Ethics Committee opinion. Fertil Steril, 2018 August;110(3):380386.Google Scholar
Lambertini, M, Fontana, V, Massarotti, C et al. Prospective study to optimize care and improve knowledge on ovarian function and/or fertility preservation in young breast cancer patients: results of the pilot phase of the PREgnancy and FERtility (PREFER) study. Breast, 2018 June 22;41:5156.Google Scholar
Rodriguez-Wallberg, KA, Tanbo, T, Tinkanen, H et al. Ovarian tissue cryopreservation and transplantation among alternatives for fertility preservation in the Nordic countries – compilation of 20 years of multicenter experience. Acta Obstet Gynecol Scand, 2016 September;95(9):10151026.Google Scholar
Oktay, K, Buyuk, E, Davis, O et al. Fertility preservation in breast cancer patients: IVF and embryo cryopreservation after ovarian stimulation with tamoxifen. Hum Reprod, 2003;18(1):9095.Google Scholar
Meirow, D, Raanani, H, Maman, E et al. Tamoxifen co-administration during controlled ovarian hyperstimulation for in vitro fertilization in breast cancer patients increases the safety of fertility-preservation treatment strategies. Fertil Steril, 2014 August;102(2):488495.Google Scholar
Fisk, NM, Templeton, AA, Papadopoulos, GC, Matlin, SA, Wu, ZY. Lack of effect of high-dose antioestrogen on the maturation and in-vitro fertilization of human oocytes. Hum Reprod, 1989 July;4(5):584587.Google Scholar
Tulandi, T, Martin, J, Al-Fadhli, R et al. Congenital malformations among 911 newborns conceived after infertility treatment with letrozole or clomiphene citrate. Fertil Steril, 2006;85(6):17611765.Google Scholar
Ata, B, Tulandi, T. Reassurance of safety of letrozole and suggested approaches in controlled ovarian hyperstimulation. Fertil Steril, 2009 July;92(1):e6.Google Scholar
Oktay, K, Hourvitz, A, Sahin, G et al. Letrozole reduces estrogen and gonadotropin exposure in women with breast cancer undergoing ovarian stimulation before chemotherapy. J Clin Endocrinol Metab, 2006;91(10):38853890.Google Scholar
Azim, AA, Costantini-Ferrando, M, Lostritto, K, Oktay, K. Relative potencies of anastrozole and letrozole to suppress estradiol in breast cancer patients undergoing ovarian stimulation before in vitro fertilization. J Clin Endocrinol Metab, 2007;92(6):21972200.Google Scholar
Oktay, K, Hourvitz, A, Sahin, G et al. Letrozole reduces estrogen and gonadotropin exposure in women with breast cancer undergoing ovarian stimulation before chemotherapy. J Clin Endocrinol Metab, 2006 October;91(10):38853890.Google Scholar
Oktay, K, Türkçüoglu, I, Rodriguez-Wallberg, KA. GnRH agonist trigger for women with breast cancer undergoing fertility preservation by aromatase inhibitor/FSH stimulation. Reprod Biomed Online, 2010;20(6):783788.Google Scholar
Sönmezer, M1, Türkçüoğlu, I, Coşkun, U, Oktay, K. Random-start controlled ovarian hyperstimulation for emergency fertility preservation in letrozole cycles. Fertil Steril, 2011 May;95(6):2125.e911.Google Scholar
Oktay, K, Buyuk, E, Rodriguez-Wallberg, KA, Sahin, G. In vitro maturation improves oocyte or embryo cryopreservation outcome in breast cancer patients undergoing ovarian stimulation for fertility preservation. Reprod Biomed Online, 2010 May;20(5):634638.Google Scholar
Gellert, SE, Pors, SE, Kristensen, SG et al. 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 April;35(4):561570.Google Scholar
Azim, AA, Costantini-Ferrando, M, Oktay, K. Safety of fertility preservation by ovarian stimulation with letrozole and gonadotropins in patients with breast cancer: a prospective controlled study. J Clin Oncol, 2008;26:26302635.Google Scholar
Kim, J, Turan, V, Oktay, K. Long-term safety of letrozone and gonadotropin stimulation for fertility preservation in women with breast cancer. J Clin Endocrinol Metab, 2016;101(4):13641371, jc20153878.Google Scholar
Rodriguez-Wallberg, KA, Eloranta, S, Krawiec, K et al. Safety of fertility preservation in breast cancer patients in a register-based matched cohort study. Breast Cancer Res Treat 2018 February;167 (3):761769.Google Scholar
Kroman, N, Jensen, MB, Wohlfahrt, J, Ejlertsen, B; Danish Breast Cancer Cooperative Group. Pregnancy after treatment of breast cancer–a population-based study on behalf of Danish Breast Cancer Cooperative Group. Acta Oncol, 2008;47(4):545549.Google Scholar
Azim, HA Jr, Santoro, L, Pavlidis, N et al. Safety of pregnancy following breast cancer diagnosis: a meta-analysis of 14 studies. Eur J Cancer, 2011 January;47(1):7483.Google Scholar
Hartman, EK, Eslick, GD. The prognosis of women diagnosed with breast cancer before, during and after pregnancy: a meta-analysis. Breast Cancer Res Treat, 2016 November;160(2):347360.Google Scholar
Sankila, R, Heinävaara, S, Hakulinen, T. Survival of breast cancer patients after subsequent term pregnancy: “healthy mother effect”. Am J Obstet Gynecol, 1994 March;170(3):818823.Google Scholar
Dalberg, K, Eriksson, J, Holmberg, L. Birth outcome in women with previously treated breast cancer–a population-based cohort study from Sweden. PLoS Med, 2006;3(9):e336.Google Scholar
Marklund, A, Nasiell, J, Berger, AS, Fagerberg, A, Rodriguez-Wallberg, KA. Pregnancy achieved using donor eggs in cancer survivors with treatment-induced ovarian failure: obstetric and perinatal outcome. J Womens Health (Larchmt), 2018 July;27(7):939945.Google Scholar

References

Oktem, O, Urman, B. Options of fertility preservation in female cancer patients. Obstet Gynecol Surv, 2010;65(8):531542.Google Scholar
Siegel, RL, Miller, KD, Jemal, A. Cancer statistics, 2018. CA Cancer J Clin, 2018;68(1):730.Google Scholar
Smith, A, Howell, D, Patmore, R, Jack, A, Roman, E. Incidence of haematological malignancy by sub-type: a report from the Haematological Malignancy Research Network. Br J Cancer, 2011;105(11):16841692.Google Scholar
Oktem, O, Kim, SS, Selek, U, Schatmann, G, Urman, B. Ovarian and uterine functions in female survivors of childhood cancers. Oncologist, 2018;23(2):214224.Google Scholar
Plowchalk, DR, Mattison, DR. Phosphoramide mustard is responsible for the ovarian toxicity of cyclophosphamide. Toxicol Appl Pharmacol, 1991;107(3):472481.Google Scholar
Oktem, O, Oktay, K. Quantitative assessment of the impact of chemotherapy on ovarian follicle reserve and stromal function. Cancer, 2007;110(10):22222229.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(6):16261633.Google Scholar
Bildik, G, Akin, N, Senbabaoglu, F et al. GnRH agonist leuprolide acetate does not confer any protection against ovarian damage induced by chemotherapy and radiation in vitro. Hum Reprod, 2015;30(12):29122925.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(185):185ra162.Google Scholar
Morgan, S, Lopes, F, Gourley, C, Anderson, RA, Spears, N. 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
Himelstein-Braw, R, Peters, H, Faber, M. Morphological study of the ovaries of leukaemic children. Br J Cancer, 1978;38(1):8287.Google Scholar
Epstein, RJ. Drug-induced DNA damage and tumor chemosensitivity. J Clin Oncol, 1990;8(12):20622084.Google Scholar
Antal, Z, Sklar, CA. Gonadal function and fertility among survivors of childhood cancer. Endocrinol Metab Clin North Am, 2015;44(4):739749.Google Scholar
Oktem, O, Oktay, K. The ovary: anatomy and function throughout human life. Ann N Y Acad Sci, 2008;1127:19.Google Scholar
Oktem, O, Oktay, K. Preservation of menstrual function in adolescent and young females. Ann N Y Acad Sci, 2008;1135:237243.Google Scholar
Bines, J, Oleske, DM, Cobleigh, MA. Ovarian function in premenopausal women treated with adjuvant chemotherapy for breast cancer. J Clin Oncol, 1996;14(5):17181729.Google Scholar
Wallace, WH, Thomson, AB, Kelsey, TW. The radiosensitivity of the human oocyte. Hum Reprod, 2003;18(1):117121.Google Scholar
Ashwood-Smith, MJ, Edwards, RG. DNA repair by oocytes. Mol Hum Reprod, 1996;2(1):4651.Google Scholar
Adriaens, I, Smitz, J, Jacquet, P. The current knowledge on radiosensitivity of ovarian follicle development stages. Hum Reprod Update, 2009;15(3):359377.Google Scholar
Jaroudi, S, Kakourou, G, Cawood, S et al. Expression profiling of DNA repair genes in human oocytes and blastocysts using microarrays. Hum Reprod, 2009;24(10):26492655.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(2):269275.Google Scholar
Oktem, O, Oktay K. Preservation of menstrual function in adolescent and young females. Ann N Y Acad Sci, 2008; 1135:237–243.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 Cli Oncol, 2009;27(16):2677–2685.Google Scholar
Critchley, HO, Wallace, WH. Impact of cancer treatment on uterine function. J Natl Cancer Inst Monogr, 2005;34:6468.Google Scholar
Sudour, H, Chastagner, P, Claude, L et al. Fertility and pregnancy outcome after abdominal irradiation that included or excluded the pelvis in childhood tumor survivors. Int J Radiat Oncol Biol Phys, 2010;76(3):867873.Google Scholar
Beneventi, F, Locatelli, E, Giorgiani, G et al. Gonadal and uterine function in female survivors treated by chemotherapy, radiotherapy, and/or bone marrow transplantation for childhood malignant and non-malignant diseases. BJOG, 2014;121(7):856865; discussion 865.Google Scholar
Hawkins, MM, Smith, RA. Pregnancy outcomes in childhood cancer survivors: probable effects of abdominal irradiation. Int J Cancer, 1989;43(3):399402.Google Scholar
Knopman, JM, Papadopoulos, EB, Grifo, JA et al. Surviving childhood and reproductive-age malignancy: effects on fertility and future parenthood. Lancet Oncol, 2010;11(5):490–498.Google Scholar
Critchley, HO, Bath, LE, Wallace, WH, Radiation damage to the uterus – review of the effects of treatment of childhood cancer. Hum Fertil (Camb), 2002;5(2):61–66.Google Scholar
Norwitz, ER, Stern, HM, Grier, H, Lee-Parritz, A. Placenta percreta and uterine rupture associated with prior whole body radiation therapy. Obstet Gynecol, 2001;98(5 Pt 2):929931.Google Scholar
Teh, WT et al. The impact of uterine radiation on subsequent fertility and pregnancy outcomes. Biomed Res Int, 2014;2014:482968.Google Scholar
Barton, SE 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(9):873881.Google Scholar
Gill, S, Blackstock, AW, Goldberg, RM, Colorectal cancer. Mayo Clin Proc, 2007; 82(1):114129.Google Scholar
Maindrault-Goebel, F, et al. Oxaliplatin added to the simplified bimonthly leucovorin and 5-fluorouracil regimen as second-line therapy for metastatic colorectal cancer (FOLFOX6). GERCOR. Eur J Cancer, 1999; 35(9):13381342.Google Scholar
Fuchs, CS et al. Randomized, controlled trial of irinotecan plus infusional, bolus, or oral fluoropyrimidines in first-line treatment of metastatic colorectal cancer: results from the BICCC Study. J Clin Oncol, 2007; 25(30):47794786.Google Scholar
Douillard, JY et al. Panitumumab-FOLFOX4 treatment and RAS mutations in colorectal cancer. N Engl J Med, 2013; 369(11):10231034.Google Scholar
Cercek, A et al. Incidence of chemotherapy-induced amenorrhea in premenopausal women treated with adjuvant FOLFOX for colorectal cancer. Clin Colorectal Cancer, 2013;12(3):163167.Google Scholar
Wan, J et al. Incidence of chemotherapy- and chemoradiotherapy-induced amenorrhea in premenopausal women with stage II/III colorectal cancer. Clin Colorectal Cancer, 2015;14(1):3134.Google Scholar
Utsunomiya, T et al. A novel molecular mechanism for anticancer drug-induced ovarian failure: Irinotecan HCl, an anticancer topoisomerase I inhibitor, induces specific FasL expression in granulosa cells of large ovarian follicles to enhance follicular apoptosis. Int J Oncol, 2008;32(5):9911000.Google Scholar
Tanaka, T et al. Irinotecan HCl, an anticancer topoisomerase I inhibitor, frequently induces ovarian failure in premenopausal and perimenopausal women. Oncol Rep, 2008;19(5):11231133.Google Scholar
Zamah, AM et al. Will imatinib compromise reproductive capacity? Oncologist, 2011;16(10):14221427.Google Scholar
Christopoulos, C, Dimakopoulou, V, Rotas, E. Primary ovarian insufficiency associated with imatinib therapy. N Engl J Med, 2008;358(10):10791080.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. Cisplatin-induced primordial follicle oocyte killing and loss of fertility are not prevented by imatinib. Nat Med, 2012;18(8):11701172; author reply 1172–1174.Google Scholar
Bildik, G C-Abl is not activated in DNA damage-induced and Tap63-mediated oocyte apoptosis in human ovary. Cell Death Dis, 2018;9(10):943.Google Scholar
Jou, E, Rajdev, L. Current and emerging therapies in unresectable and recurrent gastric cancer. World J Gastroenterol, 2016;22(20):48124823.Google Scholar
Berliere, M et al. Incidence of reversible amenorrhea in women with breast cancer undergoing adjuvant anthracycline-based chemotherapy with or without docetaxel. BMC Cancer, 2008;8:56.Google Scholar
Nabholtz, JPT, Mackey, J. Phase III trial comparing TAC (docetaxel, doxorubicin, cyclophosphamide) with FAC (5-fluorouracil, doxorubicin, cyclophosphamide) in the adjuvant treatment of node positive breast cancer (BC) patients: interim analysis of the BCIRG 001 study. Proc Annu Meet Am Soc Clin Oncol, 2002;141 (Abstract).Google Scholar
Hortobagyi, GN et al. Immediate and long-term toxicity of adjuvant chemotherapy regimens containing doxorubicin in trials at M.D. Anderson Hospital and Tumor Institute. NCI Monogr, 1986(1):105–109.Google Scholar
Gastrointestinal Tumor Study Group. Prolongation of the disease-free interval in surgically treated rectal carcinoma. N Engl J Med, 1985;312(23):14651472.Google Scholar
Krook, JE et al. Effective surgical adjuvant therapy for high-risk rectal carcinoma. N Engl J Med, 1991;324(11):709715.Google Scholar
Tveit, KM et al. Randomized controlled trial of postoperative radiotherapy and short-term time scheduled 5-fluorouracil against surgery alone in the treatment of Dukes B and C rectal cancer. Norwegian Adjuvant Rectal Cancer Project Group. Br J Surg, 1997;84(8):11301135.Google Scholar
O’Connell, MJ et al. Improving adjuvant therapy for rectal cancer by combining protracted-infusion fluorouracil with radiation therapy after curative surgery. N Engl J Med, 1994;331(8):502507.Google Scholar
Tepper, JE et al. Adjuvant postoperative fluorouracil-modulated chemotherapy combined with pelvic radiation therapy for rectal cancer: initial results of intergroup 0114. J Clin Oncol, 1997;15(5):20302039.Google Scholar
Tepper, JE et al. Adjuvant therapy in rectal cancer: analysis of stage, sex, and local control–final report of intergroup 0114. J Clin Oncol, 2002;20(7):17441750.Google Scholar
Fietkau, R, Rodel, C. [Postoperative radiochemotherapy of rectal carcinoma reduces the incidence of locoregional recurrences but does not improve the prognosis of patients–the results of a randomized study of the NSABP-R02. National Surgical Adjuvant Breast and Bowel Project Protocol]. Strahlenther Onkol, 2000;176(8):381382.Google Scholar
Wolmark, N et al. Randomized trial of postoperative adjuvant chemotherapy with or without radiotherapy for carcinoma of the rectum: National Surgical Adjuvant Breast and Bowel Project Protocol R-02. J Natl Cancer Inst, 2000;92(5):388396.Google Scholar
Lee, JH et al. Randomized trial of postoperative adjuvant therapy in stage II and III rectal cancer to define the optimal sequence of chemotherapy and radiotherapy: a preliminary report. J Clin Oncol, 2002;20(7):17511758.Google Scholar
Frykholm GJ, Glimelius B, Pahlman L. Preoperative or postoperative irradiation in adenocarcinoma of the rectum: final treatment results of a randomized trial and an evaluation of late secondary effects. Dis Colon Rectum, 1993;36(6):564–572.Google Scholar
Sauer, R et al. Preoperative versus postoperative chemoradiotherapy for rectal cancer. N Engl J Med, 2004;351(17):1731–1740.Google Scholar
Behringer, K et al. Gonadal function and fertility in survivors after Hodgkin lymphoma treatment within the German Hodgkin Study Group HD13 to HD15 trials. J Clin Oncol, 2013;31(2):231–239.Google Scholar
Jadoul P, Kim SS, IP Committee, Fertility considerations in young women with hematological malignancies. J Assist Reprod Genet, 2012;29(6):479–487.Google Scholar
Kiserud CE et al. Post-treatment parenthood in Hodgkin’s lymphoma survivors. Br J Cancer, 2007;96(9):1442–1449.Google Scholar
Hodgson DC et al. Fertility among female Hodgkin lymphoma survivors attempting pregnancy following ABVD chemotherapy. Hematol Oncol, 2007;25(1):11–15.Google Scholar
Behringer K et al. Fertility and gonadal function in female survivors after treatment of early unfavorable Hodgkin lymphoma (HL) within the German Hodgkin Study Group HD14 trial. Ann Oncol, 2012;23(7):1818–1825.Google Scholar
De Bruin ML et al. Treatment-related risk factors for premature menopause following Hodgkin lymphoma. Blood, 2008;111(1):101–108.Google Scholar
Donnez J et al. Children born after autotransplantation of cryopreserved ovarian tissue. a review of 13 live births. Ann Med, 2011;43(6):437–450.Google Scholar
Bittinger SE et al. Detection of Hodgkin lymphoma within ovarian tissue. Fertil Steril, 2011;95(2):803 e3–6.Google Scholar
Chorlton I, Norris HJ, King FM. Malignant reticuloendothelial disease involving the ovary as a primary manifestation: a series of 19 lymphomas and 1 granulocytic sarcoma. Cancer, 1974;34(2):397–407.Google Scholar
Meirow D et al. Ovarian tissue banking in patients with Hodgkin’s disease: is it safe? Fertil Steril, 1998;69(6):996–998.Google Scholar
Seshadri T et al. Lack of evidence of disease contamination in ovarian tissue harvested for cryopreservation from patients with Hodgkin lymphoma and analysis of factors predictive of oocyte yield. Br J Cancer, 2006;94(7):1007–1010.Google Scholar
Elis A et al. Fertility status among women treated for aggressive non-Hodgkin’s lymphoma. Leuk Lymphoma, 2006;47(4):623–627.Google Scholar
Seshadri T et al. The effect of the Hyper-CVAD chemotherapy regimen on fertility and ovarian function. Leuk Res, 2006;30(4):483–485.Google Scholar
Shaw JM et al. Fresh and cryopreserved ovarian tissue samples from donors with lymphoma transmit the cancer to graft recipients. Hum Reprod, 1996;11(8):1668–1673.Google Scholar
Dolmans MM et al. Reimplantation of cryopreserved ovarian tissue from patients with acute lymphoblastic leukemia is potentially unsafe. Blood, 2010;116(16):2908–2914.Google Scholar
Dolmans MM et al. Efficacy of in vitro fertilization after chemotherapy. Fertil Steril, 2005;83(4):897–901.Google Scholar
Gellert SE et al. 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;35(4):561–570.Google Scholar
Bastings L et al. Autotransplantation of cryopreserved ovarian tissue in cancer survivors and the risk of reintroducing malignancy: a systematic review. Hum Reprod Update, 2013;19(5):483–506.Google Scholar
Cuellar S et al. BCR-ABL1 tyrosine kinase inhibitors for the treatment of chronic myeloid leukemia. J Oncol Pharm Pract, 2018;24(6):433–452.Google Scholar
Carlsson IB et al. Kit ligand and c-Kit are expressed during early human ovarian follicular development and their interaction is required for the survival of follicles in long-term culture. Reproduction, 2006;131(4):641–649.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):987–997.Google Scholar
Tuppi M et al. Oocyte DNA damage quality control requires consecutive interplay of CHK2 and CK1 to activate p63. Nat Struct Mol Biol, 2018;25(3):261–269.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
×