Skip to main content Accessibility help
×
Hostname: page-component-7479d7b7d-rvbq7 Total loading time: 0 Render date: 2024-07-13T00:58:53.996Z Has data issue: false hasContentIssue false

Chapter 2 - The Effect of Chemotherapy on the Human Reproductive System

from Section 1 - Introduction

Published online by Cambridge University Press:  27 March 2021

Jacques Donnez
Affiliation:
Catholic University of Louvain, Brussels
S. Samuel Kim
Affiliation:
University of Kansas School of Medicine
Get access

Summary

Chemotherapy induces ovarian damage in a drug- and dose-dependent manner, and is related to age at the time of treatment, with progressively smaller doses required to produce ovarian failure with increasing age [1, 2].

The stockpile of primordial follicles found in the cortex of the ovaries represents the ovarian reserve. Histological studies of human ovaries have shown chemotherapy to cause ovarian atrophy and global loss of primordial follicles [3, 4]. A reduced follicular reserve may result in premature ovarian failure (POF) and menopause many years posttreatment, even in patients undergoing chemotherapy at a very young age [5]. The odds ratio of POF is as high as 3.8 in childhood cancer survivors of Hodgkin’s disease and 3.2 for non-Hodgkin’s lymphoma [6]. In addition, women who do not undergo POF and retain ovarian functionality after chemotherapy may still have reduced fertility.

Type
Chapter
Information
Fertility Preservation
Principles and Practice
, pp. 11 - 17
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

Meirow, D, Nugent, D. The effects of radiotherapy and chemotherapy on female reproduction. Hum Reprod Update, 2001;7(6):535543.Google Scholar
Whitehead, E et al. The effect of combination chemotherapy on ovarian function in women treated for Hodgkin’s disease. Cancer, 1983;52(6):988993.Google Scholar
Himelstein-Braw, R, Peters, H, Faber, M. Morphological study of the ovaries of leukaemic children. Br J Cancer, 1978;38(1):8287.Google Scholar
Familiari, G et al. Ultrastructure of human ovarian primordial follicles after combination chemotherapy for Hodgkin’s disease. Hum Reprod, 1993;8(12):20802087.Google Scholar
Sklar, CA et al. Premature menopause in survivors of childhood cancer: a report from the childhood cancer survivor study. J Natl Cancer Inst, 2006;98(13):890896.CrossRefGoogle ScholarPubMed
Chemaitilly, W et al. Acute ovarian failure in the childhood cancer survivor study. J Clin Endocrinol Metab, 2006;91(5):17231728.Google Scholar
Nielsen, SN et al. A 10-year follow up of reproductive function in women treated for childhood cancer. Reprod Biomed Online, 2013;27(2):192200.Google Scholar
Charpentier, AM et al. Anti-Mullerian hormone screening to assess ovarian reserve among female survivors of childhood cancer. J Cancer Surviv, 2014;8(4):548554.Google Scholar
Thomas-Teinturier, C et al. Ovarian reserve after treatment with alkylating agents during childhood. Hum Reprod, 2015;30(6):14371446.Google Scholar
Luke, B et al. Assisted reproductive technology use and outcomes among women with a history of cancer. Hum Reprod, 2016;31(1):183189.CrossRefGoogle ScholarPubMed
Meirow, D et al. Subclinical depletion of primordial follicular reserve in mice treated with cyclophosphamide: clinical importance and proposed accurate investigative tool. Hum Reprod, 1999;14(7):19031907.Google Scholar
Meirow, D et al. Toxicity of chemotherapy and radiation on female reproduction. Clin Obstet Gynecol, 2010;53(4):727739.Google Scholar
Fan, W. Possible mechanisms of paclitaxel-induced apoptosis. Biochem Pharmacol, 1999;57(11):12151221.Google Scholar
Stumm, S et al. Paclitaxel treatment of breast cancer cell lines modulates Fas/Fas ligand expression and induces apoptosis which can be inhibited through the CD40 receptor. Oncology, 2004;66(2):101111.Google Scholar
Roti Roti, EC et al. Acute doxorubicin insult in the mouse ovary is cell- and follicle-type dependent. PLoS One, 2012;7(8):e42293.CrossRefGoogle ScholarPubMed
Lopes, F et al. Docetaxel induces moderate ovarian toxicity in mice, primarily affecting granulosa cells of early growing follicles. Mol Hum Reprod, 2014;20(10):948959.Google Scholar
Kalich-Philosoph, L et al. Cyclophosphamide triggers follicle activation and “burnout”; AS101 prevents follicle loss and preserves fertility. Sci Transl Med, 2013;5(185):185ra62.CrossRefGoogle ScholarPubMed
Chen, XY et al. Follicle loss and apoptosis in cyclophosphamide-treated mice: What’s the matter? Int J Mol Sci, 2016;17(6)836.Google Scholar
Ben-Aharon, I et al. Doxorubicin-induced ovarian toxicity. Reprod Biol Endocrinol, 2010;8:20.Google Scholar
Lopez, SG, Luderer, U. Effects of cyclophosphamide and buthionine sulfoximine on ovarian glutathione and apoptosis. Free Radic Biol Med, 2004;36(11):13661377.CrossRefGoogle ScholarPubMed
Anderson, RA et al. The effects of chemotherapy and long-term gonadotrophin suppression on the ovarian reserve in premenopausal women with breast cancer. Hum Reprod, 2006;21(10):25832592.Google Scholar
Rosendahl, M et al. Dynamics and mechanisms of chemotherapy-induced ovarian follicular depletion in women of fertile age. Fertil Steril, 2010 June;94(1):156–66, epub March 31.Google Scholar
Kalich-Philosoph, L, Roness, H, Carmely, A et al. Cyclophosphamide triggers follicle activation causing ovarian reserve “burn out”; AS101 prevents follicle loss and preserves fertility. Sci Transl Med, 2013;15(5):185.Google Scholar
Lande, Y et al. Short-term exposure of human ovarian follicles to cyclophosphamide metabolites seems to promote follicular activation in vitro. Reprod Biomed Online, 2017;34(1):104114.Google Scholar
Chang, EM et al. Cisplatin induces overactivation of the dormant primordial follicle through PTEN/AKT/FOXO3a pathway which leads to loss of ovarian reserve in mice. PLoS One, 2015;10(12):e0144245.Google Scholar
Jang, H et al. Melatonin prevents cisplatin-induced primordial follicle loss via suppression of PTEN/AKT/FOXO3a pathway activation in the mouse ovary. J Pineal Res, 2016;60(3):336347.Google Scholar
Durlinger, AL, Visser, JA, Themmen, AP. Regulation of ovarian function: the role of anti-Mullerian hormone. Reproduction, 2002;124(5):601609.Google Scholar
Adhikari, D, Liu, K. Molecular mechanisms underlying the activation of mammalian primordial follicles. Endocr Rev, 2009;30(5):438464.Google Scholar
Kawamura, K et al. Hippo signaling disruption an Akt stimulation of ovarian follicles for infertility treatment. Proc Natl Acad Sci U S A, 2013;110(43):1747417479.Google Scholar
Li, J et al. Activation of dormant ovarian follicles to generate mature eggs. Proc Natl Acad Sci U S A, 2010;107(22):1028010284.Google Scholar
McLaughlin, M et al. Inhibition of phosphatase and tensin homologue (PTEN) in human ovary in vitro results in increased activation of primordial follicles but compromises development of growing follicles. Mol Hum Reprod, 2014;20(8):736744.Google Scholar
Durlinger, AL et al. Anti-Mullerian hormone inhibits initiation of primordial follicle growth in the mouse ovary. Endocrinology, 2002;143(3):10761084.CrossRefGoogle ScholarPubMed
Durlinger, AL et al. Control of primordial follicle recruitment by anti-Mullerian hormone in the mouse ovary. Endocrinology, 1999;140(12):57895796.Google Scholar
Marcello, MF et al. Structural and ultrastructural study of the ovary in childhood leukemia after successful treatment. Cancer, 1990;66(10):20992104.Google Scholar
Meirow, D et al. Cortical fibrosis and blood-vessels damage in human ovaries exposed to chemotherapy. Potential mechanisms of ovarian injury. Hum Reprod, 2007;22(6):1626–1633.Google Scholar
Nicosia, SV, Matus-Ridley, M, Meadows, AT. Gonadal effects of cancer therapy in girls. Cancer, 1985;55(10):23642372.Google Scholar
Soleimani, R, Heytens, E, Oktay, K. Enhancement of neoangiogenesis and follicle survival by sphingosine-1-phosphate in human ovarian tissue xenotransplants. PLoS One, 2011;6(4):e19475.Google Scholar
Ben-Aharon, I et al. Chemotherapy-induced ovarian failure as a prototype for acute vascular toxicity. Oncologist, 2012;17(11):13861393.Google Scholar
Bar-Joseph, H et al. In vivo bioimaging as a novel strategy to detect doxorubicin-induced damage to gonadal blood vessels. PLoS One, 2011;6(9):e23492.Google Scholar
Sanders, JE et al. Pregnancies following high-dose cyclophosphamide with or without high-dose busulfan or total-body irradiation and bone marrow transplantation. Blood, 1996;87:30453052.Google Scholar
Green, DM et al. Fertility of female survivors of childhood cancer: a report from the childhood cancer survivor study. J Clin Oncol, 2009;27(16):26772685.Google Scholar
Mackie, EJ, Radford, M, Shalet, SM. Gonadal function following chemotherapy for childhood Hodgkin’s disease. Med Pediatr Oncol, 1996;27(2):7478.3.0.CO;2-Q>CrossRefGoogle ScholarPubMed
Behringer, K et al. Secondary amenorrhea after Hodgkin’s lymphoma is influenced by age at treatment, stage of disease, chemotherapy regimen, and the use of oral contraceptives during therapy: a report from the German Hodgkin’s Lymphoma Study Group. J Clin Oncol, 2005;23(30):75557564.Google Scholar
Brusamolino, E et al. Treatment of early-stage Hodgkin’s disease with four cycles of ABVD followed by adjuvant radio-therapy: analysis of efficacy and long-term toxicity. Haematologica, 2000;85(10):10321039.Google Scholar
De Bruin, ML et al. Treatment-related risk factors for premature menopause following Hodgkin lymphoma. Blood, 2008;111(1):101108.Google Scholar
Hodgson, DC et al. Fertility among female Hodgkin lymphoma survivors attempting pregnancy following ABVD chemotherapy. Hematol Oncol, 2006;25:1115.Google Scholar
Gershenson, DM. Management of ovarian germ cell tumors. J Clin Oncol, 2007;25(20):29382943.Google Scholar
Gershenson, DM. Menstrual and reproductive function after treatment with combination chemotherapy for malignant ovarian germ cell tumors. J Clin Oncol, 1988;6(2):270275.Google Scholar
Park, MC et al. Risk of ovarian failure and pregnancy outcome in patients with lupus nephritis treated with intravenous cyclophosphamide pulse therapy. Lupus, 2004; 13(8):569574.Google Scholar
Blumenfeld, Z et al. Prevention of irreversible chemotherapy-induced ovarian damage in young women with lymphoma by a gonadotrophin-releasing hormone agonist in parallel to chemotherapy. Hum Reprod, 1996;11(8):1620–1626.Google Scholar
Blumenfeld, Z. How to preserve fertility in young women exposed to chemotherapy? The role of GnRH agonist cotreatment in addition to cryopreservation of embryo, oocytes, or ovaries. Oncologist, 2007;12(9):10441054.Google Scholar
Elgindy, EA et al. Gonadatrophin suppression to prevent chemotherapy-induced ovarian damage: a randomized controlled trial. Obstet Gynecol, 2013;121(1):7886.CrossRefGoogle ScholarPubMed
Gerber, B et al. Effect of luteinizing hormone-releasing hormone agonist on ovarian function after modern adjuvant breast cancer chemotherapy: the GBG 37 ZORO study. J Clin Oncol, 2011;29(17):23342341.Google Scholar
Munster, PN et al. Randomized trial using gonadotropin-releasing hormone agonist triptorelin for the preservation of ovarian function during (neo)adjuvant chemotherapy for breast cancer. J Clin Oncol, 2012;30(5):533538.Google Scholar
Sverrisdottir, A et al. Adjuvant goserelin and ovarian preservation in chemotherapy treated patients with early breast cancer: results from a randomized trial. Breast Cancer Res Treat, 2009;117(3):561567.Google Scholar
Del Mastro, L et al. Effect of the gonadotropin-releasing hormone analogue triptorelin on the occurrence of chemotherapy-induced early menopause in premenopausal women with breast cancer: a randomized trial. JAMA, 2011;306(3):269276.Google Scholar
Badawy, A et al. Gonadotropin-releasing hormone agonists for prevention of chemotherapy-induced ovarian damage: prospective randomized study. Fertil Steril, 2009;91(3):694697.Google Scholar
Moore, HC et al. Goserelin for ovarian protection during breast-cancer adjuvant chemotherapy. N Engl J Med, 2015;372(10):923932.Google Scholar
Song, G, Gao, H, Yuan, Z. Effect of leuprolide acetate on ovarian function after cyclophosphamide-doxorubicin-based chemotherapy in premenopausal patients with breast cancer: results from a phase II randomized trial. Med Oncol, 2013;30(3):667.Google Scholar
Gonfloni, S et al. Inhibition of the c-Abl-TAp63 pathway protects mouse oocytes from chemotherapy-induced death. Nat Med, 2009;15(10):11791185.Google Scholar
Kerr, JB et al. DNA damage-induced primordial follicle oocyte apoptosis and loss of fertility require TAp63-mediated induction of Puma and Noxa. Mol Cell, 2012 November 9;48(3):343352.Google Scholar
Morgan, S et al. Cisplatin and doxorubicin induce distinct mechanisms of ovarian follicle loss; imatinib provides selective protection only against cisplatin. PLoS One, 2013;8(7):e70117.CrossRefGoogle ScholarPubMed
Kim, SY et al. Rescue of platinum-damaged oocytes from programmed cell death through inactivation of the p53 family signaling network. Cell Death Differ, 2013;20(8):987997.Google Scholar
Hancke, K et al. Sphingosine 1-phosphate protects ovaries from chemotherapy-induced damage in vivo. Fertil Steril, 2007;87(1):172177.Google Scholar
Li, F et al. Sphingosine-1-phosphate prevents chemotherapy-induced human primordial follicle death. Hum Reprod, 2014;29(1):107113.Google Scholar
Zhou, L et al. Rapamycin prevents cyclophosphamide-induced over-activation of primordial follicle pool through PI3K/Akt/mTOR signaling pathway in vivo. J Ovarian Res, 2017;10(1):56.Google Scholar
Goldman, KN et al. mTORC1/2 inhibition preserves ovarian function and fertility during genotoxic chemotherapy. Proc Natl Acad Sci U S A, 2017;114(12):31863191.Google Scholar
Sonigo, C et al. AMH prevents primordial ovarian follicle loss and fertility alteration in cyclophosphamide-treated mice. FASEB J, 2018;33(1):12781287,fj201801089R.Google Scholar
Kano, M et al. AMH/MIS as a contraceptive that protects the ovarian reserve during chemotherapy. Proc Natl Acad Sci U S A, 2017;114(9):E1688E1697.Google Scholar
Meirow, D, Dor, J. Epidemiology and infertility in cancer patients. In Tulandi, T, Gosden, R (eds.)Preservation of Fertility. Abingdon: Taylor and Francis. 2004, 2138.Google Scholar

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
×