Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-8bljj Total loading time: 0 Render date: 2024-06-30T09:42:06.905Z Has data issue: false hasContentIssue false

Chapter 29 - In Vitro Growth of Human Oocytes

from Section 8 - In Vitro Follicle Culture

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

Patients with cancer who desire to preserve their future reproductive potential but require immediate gonadotoxic treatments (chemo and/or radiotherapy) are left with few options for fertility preservation. These options include (a) cryopreservation of ovarian tissues as cortical strips; (b) dual cryopreservation of both ovarian cortical tissue and cryopreservation, after in vitro maturation, of immature oocytes extracted from the small antral follicles visible within the ovarian cortex at the time of the harvest; and (c) cryopreservation of one whole ovary [1–9]. At the time of this writing, each of these options is still considered experimental by the American Society of Reproductive Medicine (thus requiring institutional review board approval and patient’s informed consent), although it is expected that soon ovarian tissue cryopreservation will no longer be considered experimental.

Type
Chapter
Information
Fertility Preservation
Principles and Practice
, pp. 332 - 340
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

Dolmans, MM, Manavella, DD. Recent advances in fertility preservation. J Obstet Gynaecol Res, 2019;45:266279.CrossRefGoogle ScholarPubMed
Anderson, RA, Mitchell, RT, Kelsey, TW et al. Cancer treatment and gonadal function: experimental and established strategies for fertility preservation in children and young adults. Lancet Diabetes Endocrinol, 2015;3:556567.CrossRefGoogle ScholarPubMed
Anderson, RA, Wallace, WHB, Telfer, EE. Ovarian tissue cryopreservation for fertility preservation: clinical and research perspectives. Hum Reprod Open, March 29 2017;2017(1).CrossRefGoogle ScholarPubMed
Anderson, RA, Baird, DT. The development of ovarian tissue cryopreservation in Edinburgh: translation from a rodent model through validation in a large mammal and then into clinical practice. Acta Obstet Gynecol Scand, 2019;98(5):545549.Google Scholar
Donnez, J, Dolmans, MM. Fertility preservation in women. N Engl J Med, 2017;377:16571665.Google Scholar
Telfer, EE, Zelinski, MB. Ovarian follicle culture: advances and challenges for human and nonhuman primates. Fertil Steril, 2013;99:15231533.CrossRefGoogle ScholarPubMed
Eppig, JJ, O’Brien, MJ. Development in vitro of mouse oocytes from primordial follicles. Biol Reprod, 1996;54:197207.CrossRefGoogle ScholarPubMed
O’Brien, MJ, Pendola, JK, Eppig, JJ. A revised protocol for in vitro development of mouse oocytes from primordial follicles dramatically improves their developmental competence. Biol Reprod, 2003;68:16821686.Google Scholar
Herta, AC, Lolicato, F, Smitz, JEJ. In vitro follicle culture in the context of IVF. Reproduction, 2018;156:F59F73.Google Scholar
Anderson, RA, Telfer, EE. Being a good egg in the 21st century. Br Med Bull, 2018;127:8389.Google Scholar
McLaughlin, M, Albertini, DF, Wallace, WHB, Anderson, RA, Telfer, EE. Metaphase II oocytes from human unilaminar follicles grown in a multi-step culture system. Mol Hum Reprod, 2018;24:135142.Google Scholar
Oktem, O, Oktay, K. The ovary: anatomy and function throughout human life. Ann N Y Acad Sci, 2008;1127:19.Google Scholar
Hsueh, AJ, Kawamura, K, Cheng, Y, Fauser, BC. Intraovarian control of early folliculogenesis. Endocr Rev, 2015;36:124.Google Scholar
Sanfins, A, Rodrigues, P, Albertini, DF. GDF-9 and BMP-15 direct the follicle symphony. J Assist Reprod Genet, 2018;35:17411750.CrossRefGoogle ScholarPubMed
Zhang, J, Xu, Y, Liu, H, Pan, Z. MicroRNAs in ovarian follicular atresia and granulosa cell apoptosis. Reprod Biol Endocrinol, 2019;17:9.Google Scholar
Shah, JS, Sabouni, R, Cayton Vaught, KC et al. Biomechanics and mechanical signaling in the ovary: a systematic review. J Assist Reprod Genet, 2018;35:11351148.Google Scholar
Sirard, MA. Resumption of meiosis: mechanism involved in meiotic progression and its relation with developmental competence. Theriogenology, 2001;55:12411254.Google Scholar
Matsuda, F, Inoue, N, Manabe, N, Ohkura, S. Follicular growth and atresia in mammalian ovaries: regulation by survival and death of granulosa cells. J Reprod Dev, 2012;58:4450.Google Scholar
Li, R, Albertini, DF. The road to maturation: somatic cell interaction and self-organization of the mammalian oocyte. Nat Rev Mol Cell Biol, 2013;14:141152.Google Scholar
McGinnis, LK, Limback, SD, Albertini, DF. Signaling modalities during oogenesis in mammals. Curr Top Dev Biol, 2013;102:227242.CrossRefGoogle ScholarPubMed
Martin, JH, Aitken, RJ, Bromfield, EG, Nixon, B. DNA damage and repair in the female germline: contributions to ART. Hum Reprod Update, 2019;25:180201.Google Scholar
Winship, AL, Stringer, JM, Liew, SH, Hutt, KJ. The importance of DNA repair for maintaining oocyte quality in response to anti-cancer treatments, environmental toxins and maternal ageing. Hum Reprod Update, 2018;24(2):119–134.Google Scholar
Hikabe, O, Hamazaki, N, Nagamatsu, G et al. Reconstitution in vitro of the entire cycle of the mouse female germ line. Nature, 2016;539:299303.Google Scholar
Morohaku, K, Tanimoto, R, Sasaki, K et al. Complete in vitro generation of fertile oocytes from mouse primordial germ cells. Proc Natl Acad Sci U S A, 2016;113:90219026.CrossRefGoogle ScholarPubMed
Garor, R, Abir, R, Erman, A et al. Effects of basic fibroblast growth factor on in vitro development of human ovarian primordial follicles. Fertil Steril, 2009;91:19671975.Google Scholar
Hovatta, O, Silye, R, Abir, R, Krausz, T, Winston, RM. Extracellular matrix improves survival of both stored and fresh human primordial and primary ovarian follicles in long-term culture. Hum Reprod, 1997;12:10321036.Google Scholar
Hovatta, O, Wright, C, Krausz, T, Hardy, K, Winston, RM. Human primordial, primary and secondary ovarian follicles in long-term culture: effect of partial isolation. Hum Reprod, 1999;14:25192524.Google Scholar
Picton, HM, Gosden, RG. In vitro growth of human primordial follicles from frozen-banked ovarian tissue. Mol Cell Endocrinol, 2000;166:2735.CrossRefGoogle ScholarPubMed
Telfer, EE, McLaughlin, M, Ding, C, Thong, KJ. A two-step serum-free culture system supports development of human oocytes from primordial follicles in the presence of activin. Hum Reprod, 2008;23:11511158.Google Scholar
Wright, CS, Hovatta, O, Margara, R et al. Effects of follicle-stimulating hormone and serum substitution on the in-vitro growth of human ovarian follicles. Hum Reprod, 1999;14:15551562.Google Scholar
Anderson, RA, McLaughlin, M, Wallace, WH, Albertini, DF, Telfer, EE. The immature human ovary shows loss of abnormal follicles and increasing follicle developmental competence through childhood and adolescence. Hum Reprod, 2014;29:97106.CrossRefGoogle ScholarPubMed
Hreinsson, JG, Scott, JE, Rasmussen, C et al. Growth differentiation factor-9 promotes the growth, development, and survival of human ovarian follicles in organ culture. J Clin Endocrinol Metab, 2002;87:316321.CrossRefGoogle ScholarPubMed
Younis, AJ, Lerer-Serfaty, G, Stav, D et al. Extracellular-like matrices and leukaemia inhibitory factor for in vitro culture of human primordial follicles. Reprod Fertil Dev, 2017;29:19821994.Google Scholar
Kallen, A, Polotsky, AJ, Johnson, J. Untapped reserves: Controlling primordial follicle growth activation. Trends Mol Med, 2018;24:319331.Google Scholar
Grosbois, J, Demeestere, I. Dynamics of PI3 K and Hippo signaling pathways during in vitro human follicle activation. Hum Reprod, 2018;33:17051714.Google Scholar
Li, J, Kawamura, K, Cheng, Y et al. Activation of dormant ovarian follicles to generate mature eggs. Proc Natl Acad Sci U S A, 2010;107:1028010284.Google Scholar
McLaughlin, M, Kinnell, HL, Anderson, RA, Telfer, EE. 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;20:736744.Google Scholar
Reddy, P, Liu, L, Adhikari, D et al. Oocyte-specific deletion of Pten causes premature activation of the primordial follicle pool. Science, 2008;319:611613.Google Scholar
Adhikari, D, Liu, K. mTOR signaling in the control of activation of primordial follicles. Cell Cycle, 2010;9:16731674.Google Scholar
Kawamura, K, Cheng, Y, Suzuki, N et al. Hippo signaling disruption and Akt stimulation of ovarian follicles for infertility treatment. Proc Natl Acad Sci U S A, 2013;110:1747417479.Google Scholar
Zhao, B, Tumaneng, K, Guan, KL. The Hippo pathway in organ size control, tissue regeneration and stem cell self-renewal. Nat Cell Biol, 2011;13:877883.Google Scholar
Maidarti, M, Clarkson, YL, McLaughlin, M, Anderson, RA, Telfer, EE. Inhibition of PTEN activates bovine non-growing follicles in vitro but increases DNA damage and reduces DNA repair response. Hum Reprod, 2019;34:297307.Google Scholar
Spinelli, L, Lindsay, YE, Leslie, NR. PTEN inhibitors: an evaluation of current compounds. Adv Biol Regul, 2015;57:102111.Google Scholar
Stringer, JM, Winship, A, Liew, SH, Hutt, K. The capacity of oocytes for DNA repair. Cell Mol Life Sci, 2018;75:27772792.Google Scholar
McLaughlin, M, Telfer, EE. Oocyte development in bovine primordial follicles is promoted by activin and FSH within a two-step serum-free culture system. Reproduction, 2010;139:971978.Google Scholar
Telfer, EE, Binnie, JP, McCaffery, FH, Campbell, BK. In vitro development of oocytes from porcine and bovine primary follicles. Mol Cell Endocrinol, 2000;163:117123.Google Scholar
Dolmans, MM, Michaux, N, Camboni, A et al. Evaluation of Liberase, a purified enzyme blend, for the isolation of human primordial and primary ovarian follicles. Hum Reprod, 2006;21:413420.CrossRefGoogle ScholarPubMed
Rice, S, Ojha, K, Mason, H. Human ovarian biopsies as a viable source of pre-antral follicles. Hum Reprod, 2008;23:600605.Google Scholar
Jones, ASK, Shikanov, A. Follicle development as an orchestrated signaling network in a 3D organoid. J Biol Eng, 2019;13:2.Google Scholar
Rajabi, Z, Aliakbari, F, Yazdekhasti, H. Female fertility preservation, clinical and experimental options. J Reprod Infertil, 2018;19:125132.Google Scholar
Shea, LD, Woodruff, TK, Shikanov, A. Bioengineering the ovarian follicle microenvironment. Annu Rev Biomed Eng, 2014;16:2952.Google Scholar
Xu, M, Barrett, SL, West-Farrell, E et al. In vitro grown human ovarian follicles from cancer patients support oocyte growth. Hum Reprod, 2009;24:25312540.CrossRefGoogle ScholarPubMed
Skory, RM, Xu, Y, Shea, LD, Woodruff, TK. Engineering the ovarian cycle using in vitro follicle culture. Hum Reprod, 2015;30:13861395.Google Scholar
Heise, M, Koepsel, R, Russell, AJ, McGee, EA. Calcium alginate microencapsulation of ovarian follicles impacts FSH delivery and follicle morphology. Reprod Biol Endocrinol, 2005;3: 47.Google Scholar
West-Farrell, ER, Xu, M, Gomberg, MA et al. The mouse follicle microenvironment regulates antrum formation and steroid production: alterations in gene expression profiles. Biol Reprod, 2009;80:432439.Google Scholar
Laronda, MM, Jakus, AE, Whelan, KA et al. Initiation of puberty in mice following decellularized ovary transplant. Biomaterials, 2015;50:2029.Google Scholar
Laronda, MM, Rutz, AL, Xiao, S et al. A bioprosthetic ovary created using 3D printed microporous scaffolds restores ovarian function in sterilized mice. Nat Commun, 2017;8:15261.Google Scholar
Liverani, L, Raffel, N, Fattahi, A et al. Electrospun patterned porous scaffolds for the support of ovarian follicles growth: a feasibility study. Sci Rep, 2019;9:1150.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
Chian, RC, Uzelac, PS, Nargund, G. In vitro maturation of human immature oocytes for fertility preservation. Fertil Steril, 2013;99:11731181.Google Scholar
Nogueira, D, Sadeu, JC, Montagut, J. In vitro oocyte maturation: current status. Semin Reprod Med, 2012;30:199213.Google Scholar
Edwards, RG, Bavister, BD, Steptoe, PC. Early stages of fertilization in vitro of human oocytes matured in vitro. Nature, 1969;221:632635.Google Scholar
Cha, KY, Koo, JJ, Ko, JJ et al. Pregnancy after in vitro fertilization of human follicular oocytes collected from nonstimulated cycles, their culture in vitro and their transfer in a donor oocyte program. Fertil Steril, 1991;55:109113.CrossRefGoogle Scholar
Shirasawa, H, Terada, Y. In vitro maturation of human immature oocytes for fertility preservation and research material. Reprod Med Biol, 2017;16:258267.Google Scholar
Barrett, SL, Albertini, DF. Cumulus cell contact during oocyte maturation in mice regulates meiotic spindle positioning and enhances developmental competence. J Assist Reprod Genet, 2010;27:2939.Google Scholar
Coticchio, G, Guglielmo, MC, Dal Canto, M et al. Mechanistic foundations of the metaphase II spindle of human oocytes matured in vivo and in vitro. Hum Reprod, 2013;28:32713282.CrossRefGoogle ScholarPubMed
Anckaert, E, De, Rycke, M, Smitz, J. Culture of oocytes and risk of imprinting defects. Hum Reprod Update, 2013;19:5266.Google Scholar
Picton, HM, Harris, SE, Muruvi, W, Chambers, EL. The in vitro growth and maturation of follicles. Reproduction, 2008;136:703715.Google Scholar
Telfer, EE, McLaughlin, M. In vitro development of ovarian follicles. Semin Reprod Med, 2011;29:1523.Google Scholar
Gook, DA, McCully, BA, Edgar, DH, McBain, JC. Development of antral follicles in human cryopreserved ovarian tissue following xenografting. Hum Reprod, 2001;16:417422.Google Scholar
Fisch, B, Abir, R. Female fertility preservation: past, present and future. Reproduction, 2018;156:F11F27.Google Scholar
Shi, Q, Xie, Y, Wang, Y, Li, S. Vitrification versus slow freezing for human ovarian tissue cryopreservation: a systematic review and meta-anlaysis. Sci Rep, 2017;7:8538.Google Scholar
Corkum, KS, Rhee, DS, Wafford, QE et al. Fertility and hormone preservation and restoration for female children and adolescents receiving gonadotoxic cancer treatments: A systematic review. J Pediatr Surg, 2019;54(11):22002209.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
×