Skip to main content Accessibility help
×
Hostname: page-component-77c89778f8-cnmwb Total loading time: 0 Render date: 2024-07-18T18:19:35.689Z Has data issue: false hasContentIssue false

Chapter 9 - Pediatric Cancer Therapy and Fertility

from 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

Summary

Each year, approximately 15,000 children between the ages of birth and 19 years are diagnosed with cancer [1]. Over the past five decades, there have been remarkable strides made in the modalities used to treat pediatric malignancies. As a result, nearly 80% of children diagnosed with cancer can expect to be cured. Recent estimates indicate that there are over 380,000 survivors of childhood cancer (diagnosed before the age of 20) in the United States. About 40% of these survivors are now between the ages of 20 and 40 and in their reproductive years [2].

Achieving cure is often associated with significant treatment-related sequelae, with approximately two-thirds of all pediatric cancer survivors experiencing at least one chronic medical condition [3]. Gonadal damage and infertility from chemotherapy, radiation, and surgery can be an unfortunate consequence of cancer therapy for both males and females. It is estimated that the cumulative prevalence at age 50 years in survivors of pediatric malignancies approaches 32% for primary ovarian failure and 31% for Leydig cell failure [4].

Type
Chapter
Information
Fertility Preservation
Principles and Practice
, pp. 90 - 105
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

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.CrossRefGoogle ScholarPubMed
Oeffinger, KC, Mertens, AC, Sklar, CA et al. Chronic health conditions in adult survivors of childhood cancer. N Engl J Med, 2006;355:15721582.CrossRefGoogle ScholarPubMed
Hudson, MM, Ness, KK, Gurney, JG et al. Clinical ascertainment of health outcomes among adults treated for childhood cancer. JAMA, 2013;309:23712381.CrossRefGoogle ScholarPubMed
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.CrossRefGoogle ScholarPubMed
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.CrossRefGoogle Scholar
Sklar, C. Reproductive physiology and treatment-related loss of sex hormone production. Med Pediatr Oncol, 1999;33:28.3.0.CO;2-7>CrossRefGoogle ScholarPubMed
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.CrossRefGoogle ScholarPubMed
Meistrich, ML. Effects of chemotherapy and radiotherapy on spermatogenesis in humans. Fertil Steril, 2013;100:11801186.CrossRefGoogle ScholarPubMed
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.CrossRefGoogle ScholarPubMed
Holoch, P, Wald, M. Current options for preservation of fertility in the male. Fertil Steril, 2011;96:286290.CrossRefGoogle ScholarPubMed
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;70:27032712.3.0.CO;2-X>CrossRefGoogle ScholarPubMed
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.CrossRefGoogle ScholarPubMed
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.CrossRefGoogle ScholarPubMed
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.CrossRefGoogle ScholarPubMed
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.CrossRefGoogle ScholarPubMed
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;32:692698.3.0.CO;2-I>CrossRefGoogle ScholarPubMed
Sandeman, TF. The effects of x irradiation on male human fertility. Br J Radiol, 1966;39:901907.CrossRefGoogle ScholarPubMed
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.CrossRefGoogle ScholarPubMed
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.CrossRefGoogle ScholarPubMed
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.CrossRefGoogle ScholarPubMed
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.CrossRefGoogle ScholarPubMed
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.CrossRefGoogle ScholarPubMed
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.CrossRefGoogle ScholarPubMed
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.CrossRefGoogle ScholarPubMed
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.CrossRefGoogle ScholarPubMed
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

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
×