Skip to main content Accessibility help
×
Hostname: page-component-76fb5796d-vvkck Total loading time: 0 Render date: 2024-04-25T18:07:54.745Z Has data issue: false hasContentIssue false

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
Type
Chapter
Information
Fertility Preservation
Principles and Practice
, pp. 323 - 380
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

Smitz, J, Cortvrindt, R. Oocyte in-vitro maturation and follicle culture: current clinical achievements and future directions. Hum Reprod, 1999;14(Suppl 1):145161.CrossRefGoogle ScholarPubMed
Rodrigues, P, Limback, D, McGinnis, LK, Plancha, CE, Albertini, DF. Oogenesis: prospects and challenges for the future. J Cell Physiol, 2008;216(2):355365.CrossRefGoogle ScholarPubMed
Rosendahl, M, Andersen, CY, Ernst, E et al. Ovarian function after removal of an entire ovary for cryopreservation of pieces of cortex prior to gonadotoxic treatment: a follow-up study. Hum Reprod, 2008;23(11):24752483.Google Scholar
Sanchez, M, Alama, P, Gadea, B et al. Fresh human orthotopic ovarian cortex transplantation: long-term results. Hum Reprod, 2007;22(3):786791.CrossRefGoogle ScholarPubMed
Senbon, S, Ishii, K, Fukumi, Y, Miyano, T. Fertilization and development of bovine oocytes grown in female SCID mice. Zygote, 2005;13(4):309315.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(1–2):117123.CrossRefGoogle ScholarPubMed
Van Den Hurk, R, Abir, R, Telfer, EE, Bevers, MM. Primate and bovine immature oocytes and follicles as sources of fertilizable oocytes. Hum Reprod Update, 2000;6(5):457474.Google Scholar
Hutt, KJ, Albertini, DF. An oocentric view of folliculogenesis and embryogenesis. Reprod Biomed Online, 2007;14(6):758764.Google Scholar
Jayawardana, BC, Shimizu, T, Nishimoto, H et al. Hormonal regulation of expression of growth differentiation factor-9 receptor type I and II genes in the bovine ovarian follicle. Reproduction, 2006;131(3):545553.Google Scholar
Walters, KA, Binnie, JP, Campbell, BK, Armstrong, DG, Telfer, EE. The effects of IGF-I on bovine follicle development and IGFBP-2 expression are dose and stage dependent. Reproduction, 2006;131(3):515523.Google Scholar
Blondin, P, Bousquet, D, Twagiramungu, H, Barnes, F, Sirard, MA. Manipulation of follicular development to produce developmentally competent bovine oocytes. Biol Reprod, 2002;66(1):28382843.CrossRefGoogle ScholarPubMed
Chian, RC, Chung, JT, Downey, BR, Tan, SL. Maturational and developmental competence of immature oocytes retrieved from bovine ovaries at different phases of folliculogenesis. Reprod Biomed Online, 2002;4(2):127132.Google Scholar
Fortune, JE, Kito, S, Byrd, DD. Activation of primordial follicles in vitro. J Reprod Fertil, 1999;54:439448.Google Scholar
Harada, M, Miyano, T, Matsumura, K et al. Bovine oocytes from early antral follicles grow to meiotic competence in vitro: effect of FSH and hypoxanthine. Theriogenology, 1997;48(5):743755.Google Scholar
Amorim, CA, Van, Langendonckt, A, David, A, Dolmans, MM, Donnez, J. Survival of human pre-antral follicles after cryopreservation of ovarian tissue, follicular isolation and in vitro culture in a calcium alginate matrix. Hum Reprod, 2009;24(1):9299.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
Johnson, LD, Albertini, DF, McGinnis, LK, Biggers, JD. Chromatin organization, meiotic status and meiotic competence acquisition in mouse oocytes from cultured ovarian follicles. J Reprod Fertil, 1995;104(2):277284.Google Scholar
Lenie, S, Cortvrindt, R, Adriaenssens, T, Smitz, J.A reproducible two-step culture system for isolated primary mouse ovarian follicles as single functional units. Biol Reprod, 2004;71(5):17301738.Google Scholar
Loret de, Mola JR, Barnhart, K, Kopf, GS et al. Comparison of two culture systems for the in-vitro growth and maturation of mouse preantral follicles. Clin Exp Obstet Gynecol, 2004;31(1):1519.Google Scholar
Vigo, D, Villani, S, Faustini, M et al. Follicle-like model by granulosa cell encapsulation in a barium alginate-protamine membrane. Tissue Eng, 2005;11(5–6):709714.Google Scholar
Heise, MK, Koepsel, R, McGee, EA, Russell, AJ. Dynamic oxygen enhances oocyte maturation in long-term follicle culture. Tissue Eng Part C Methods, 2009;15(3):323332.Google Scholar
Keros, V, Xella, S, Hultenby, K et al. Vitrification versus controlled-rate freezing in cryopreservation of human ovarian tissue. Hum Reprod, 2009;24(7):16701683.Google Scholar
Maman, E, Prokopis, K, Levron, J et al. Does controlled ovarian stimulation prior to chemotherapy increase primordial follicle loss and diminish ovarian reserve? An animal study. Hum Reprod, 2009;24(1):206210.Google Scholar
Bromfield, JJ, Coticchio, G, Hutt, K et al. Meiotic spindle dynamics in human oocytes following slow-cooling cryopreservation. Hum Reprod, 2009;24(9):21142123.Google Scholar
Fair, T, Hyttel, P, Greve, T. Bovine oocyte diameter in relation to maturational competence and transcriptional activity. Mol Reprod Dev, 1995;42(4):437442.Google Scholar
Humblot, P, Holm, P, Lonergan, P et al. Effect of stage of follicular growth during superovulation on developmental competence of bovine oocytes. Theriogenology, 2005;63(4):11491166.Google Scholar
Albertini, DF, Combelles, CM, Benecchi, E, Carabatsos, MJ. Cellular basis for paracrine regulation of ovarian follicle development. Reproduction, 2001;121(5):647653.Google Scholar
Feary, ES, Juengel, JL, Smith, P et al. Patterns of expression of messenger RNAs encoding GDF9, BMP15, TGFBR1, BMPR1B, and BMPR2 during follicular development and characterization of ovarian follicular populations in ewes carrying the Woodlands FecX2W mutation. Biol Reprod, 2007;77(6):990998.Google Scholar
Fouladi-Nashta, AA, Campbell, KH. Dissociation of oocyte nuclear and cytoplasmic maturation by the addition of insulin in cultured bovine antral follicles. Reproduction, 2006;131(3):449460.Google Scholar
Iwata, H, Hashimoto, S, Ohota, M et al. Effects of follicle size and electrolytes and glucose in maturation medium on nuclear maturation and developmental competence of bovine oocytes. Reproduction, 2004;127(2):159164.Google Scholar
Pavlok, A, Lapathitis, G, Cech, S et al. Simulation of intrafollicular conditions prevents GVBD in bovine oocytes: a better alternative to affect their developmental capacity after two-step culture. Mol Reprod Dev, 2005;71(2):197208.Google Scholar
Albertini, DF. Regulation of meiotic maturation in the mammalian oocyte: Inteplay between exogenous cues and the microtubule cytoskeleton. Bioessays, 1992;14(2):97103.Google Scholar
Allworth, AE, Albertini, DF. Meiotic maturation in cultured bovine oocytes is accompanied by remodeling of the cumulus cell cytoskeleton. Dev Biol, 1993;158(1):101112.Google Scholar
Combelles, CM, Carabatsos, MJ, Kumar, TR, Matzuk, MM, Albertini, DF. Hormonal control of somatic cell oocyte interactions during ovarian follicle development. Mol Reprod Dev, 2004;69(3):347355.Google Scholar
Berkholtz, CB, Shea, LD, Woodruff, TK. Extracellular matrix functions in follicle maturation. Semin Reprod Med, 2006;24(4):262269.Google Scholar
Dong, J, Albertini, DF, Nishimori, K et al. Growth differentiation factor-9 is required during early ovarian folliculogenesis. Nature, 1996;383(6600):531535.CrossRefGoogle ScholarPubMed
Carabatsos, MJ, Elvin, J, Matzuk, MM, Albertini, DF. Characterization of oocyte and follicle development in growth differentiation factor-9-deficient mice. Dev Biol, 1998;204(2):373384.CrossRefGoogle ScholarPubMed
Albertini, DF. Oocyte–granulosa cell interactions. In Blerkom, JV (ed.) Essential IVF: Reviews of Topical Issues in Clinical In Vitro Fertilization. Boston: Kluwer Academic Publishers. 2002, 4358.Google Scholar
Carabatsos, MJ, Sellitto, C, Goodenough, DA, Albertini, DF. Oocyte–granulosa cell heterologous gap junctions are required for the coordination of nuclear and cytoplasmic meiotic competence. Dev Biol, 2000;226(2):167.Google Scholar
Mattson, BA, Albertini, DF. Oogenesis: chromatin and microtubule dynamics during meiotic prophase. Mol Reprod Dev, 1990;25(4):374383.Google Scholar
McGinnis, LK, Kinsey, WH, Albertini, DF. Functions of Fyn kinase in the completion of meiosis in mouse oocytes. Dev Biol, 2009;327(2):280287.Google Scholar
Parrott, JA, Skinner, MK. Direct actions of kit-ligand on theca cell growth and differentiation during follicle development. Endocrinology, 1997;138(9):38193827.Google Scholar
Rodrigues, P, Limback, D, McGinnis, LK, Plancha, CE, Albertini, DF. Multiple mechanisms of germ cell loss in the perinatal mouse ovary. Reproduction, 2009;137(4):709720.Google Scholar
Hanoux, V, Pairault, C, Bakalska, M, Habert, R, Livera, G. Caspase-2 involvement during ionizing radiation-induced oocyte death in the mouse ovary. Cell Death Differ, 2007;14(4):671681.Google Scholar

References

Dolmans, MM, Manavella, DD. Recent advances in fertility preservation. J Obstet Gynaecol Res, 2019;45:266279.Google Scholar
Anderson, RA, Mitchell, RT, Kelsey, TW et al. Cancer treatment and gonadal function: experimental and established strategies for fertility preservation in children and young adults. Lancet Diabetes Endocrinol, 2015;3:556567.Google Scholar
Anderson, RA, Wallace, WHB, Telfer, EE. Ovarian tissue cryopreservation for fertility preservation: clinical and research perspectives. Hum Reprod Open, March 29 2017;2017(1).Google Scholar
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.CrossRefGoogle 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.Google Scholar
Eppig, JJ, O’Brien, MJ. Development in vitro of mouse oocytes from primordial follicles. Biol Reprod, 1996;54:197207.Google Scholar
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.CrossRefGoogle ScholarPubMed
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.CrossRefGoogle ScholarPubMed
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.Google Scholar
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.CrossRefGoogle ScholarPubMed
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.CrossRefGoogle ScholarPubMed
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.Google Scholar
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.Google Scholar
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.CrossRefGoogle ScholarPubMed
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.Google Scholar
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.CrossRefGoogle ScholarPubMed
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.Google Scholar
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.CrossRefGoogle ScholarPubMed
Spinelli, L, Lindsay, YE, Leslie, NR. PTEN inhibitors: an evaluation of current compounds. Adv Biol Regul, 2015;57:102111.CrossRefGoogle ScholarPubMed
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.Google Scholar
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.Google Scholar
Skory, RM, Xu, Y, Shea, LD, Woodruff, TK. Engineering the ovarian cycle using in vitro follicle culture. Hum Reprod, 2015;30:13861395.CrossRefGoogle ScholarPubMed
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.CrossRefGoogle ScholarPubMed
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.CrossRefGoogle ScholarPubMed
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.CrossRefGoogle ScholarPubMed
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.Google 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.Google Scholar
Anckaert, E, De, Rycke, M, Smitz, J. Culture of oocytes and risk of imprinting defects. Hum Reprod Update, 2013;19:5266.CrossRefGoogle ScholarPubMed
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.CrossRefGoogle ScholarPubMed
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.CrossRefGoogle ScholarPubMed
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

References

West, ER, Shea, LD, Woodruff, TK. Engineering the follicle microenvironment. Semin Reprod Med, 2007;25(4):287299.CrossRefGoogle ScholarPubMed
Woodruff, TK. The emergence of a new interdiscipline: oncofertility. Cancer Treat Res, 2007;138:311.Google Scholar
De Vos, M, Smitz, J, Woodruff, TK. Fertility preservation in women with cancer. Lancet (London, England), 2014;384(9950):13021310.Google Scholar
Goetsch, AL, Kimelman, D, Woodruff, TK. Fertility Preservation and Restoration for Patients with Complex Medical Conditions. Switzerland: Springer Nature, 2017.Google Scholar
Jeruss, JS, Woodruff, TK. Preservation of fertility in patients with cancer. N Engl J Med, 2009 February 26;360(9):902911.Google Scholar
Donnez, J, Dolmans, M-M, Diaz, C, Pellicer, A. Ovarian cortex transplantation: time to move on from experimental studies to open clinical application. Fertil Steril, 2015 November;104(5):10971098.Google Scholar
Skory, RM, Xu, Y, Shea, LD, Woodruff, TK. Engineering the ovarian cycle using in vitro follicle culture. Hum Reprod, 2015;30(6):13861395.Google Scholar
Xiao, S, Zhang, J, Romero, MM et al. In vitro follicle growth supports human oocyte meiotic maturation. Sci Rep, 2015 December 27;5(1):17323.Google Scholar
Hornick, JE, Duncan, FE, Shea, LD, Woodruff, TK. Isolated primate primordial follicles require a rigid physical environment to survive and grow in vitro. Hum Reprod, 2012;27(6):18011810.Google Scholar
Hornick, JE, Duncan, FE, Shea, LD, Woodruff, TK. Multiple follicle culture supports primary follicle growth through paracrine-acting signals. Reproduction, 2013 January;145(1):1932.Google Scholar
Tingen, CM, Kiesewetter, SE, Jozefik, J et al. A macrophage and theca cell-enriched stromal cell population influences growth and survival of immature murine follicles in vitro. Reproduction, 2011 June 1;141(6):809820.Google Scholar
Tagler, D, Tu, T, Smith, RM et al. Embryonic fibroblasts enable the culture of primary ovarian follicles within alginate hydrogels. Tissue Eng Part A, 2012 June;18(11–12):12291238.Google Scholar
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 May 1;23(5):11511158.Google Scholar
Eppig, JJ. Development in vitro of mouse oocytes from primordial follicles. Biol Reprod, 1996 January 1;54(1):197207.Google Scholar
Baird, DT, Webb, R, Campbell, BK, Harkness, LM, Gosden, RG. Long-term ovarian function in sheep after ovariectomy and transplantation of autografts Stored at −196 C. Endocrinology, 1999 January;140(1):462471.Google Scholar
Cushman, RA, Wahl, CM, Fortune, JE. Bovine ovarian cortical pieces grafted to chick embryonic membranes: a model for studies on the activation of primordial follicles. Hum Reprod, 2002 January;17(1):4854.Google Scholar
Picton, HM, Gosden, RG. In vitro growth of human primordial follicles from frozen-banked ovarian tissue. Mol Cell Endocrinol, 2000 August;166(1):2735.Google Scholar
Laronda, MM, Duncan, FE, Hornick, JE et al. Alginate encapsulation supports the growth and differentiation of human primordial follicles within ovarian cortical tissue. J Assist Reprod Genet, 2014;31(8):10131028.Google Scholar
Jakus, AE, Laronda, MM, Rashedi, AS et al. “Tissue Papers” from organ-specific decellularized extracellular matrices. Adv Funct Mater, 2017;1700992:114.Google Scholar
Magoffin, DA, Magarelli, PC. Preantral follicles stimulate luteinizing hormone independent differentiation of ovarian theca-interstitial cells by an intrafollicular paracrine mechanism. Endocrine, 1995;3(2):107112.Google Scholar
Magoffin, DA. Ovarian theca cell. Int J Biochem Cell Biol, 2005 July;37(7):13441349.Google Scholar
Magarelli, PC, Zachow, RJ, Magoffin, DA. Developmental and hormonal regulation of rat theca-cell differentiation factor secretion in ovarian follicles. Biol Reprod, 1996 August;55(2):416420.Google Scholar
Huang, CTF, Weitsman, SR, Dykes, BN, Magoffin, DA. Stem cell factor and insulin-like growth factor-I stimulate luteinizing hormone-independent differentiation of rat ovarian theca cells. Biol Reprod, 2001 February 1;64(2):451456.Google Scholar
Erickson, GF, Magoffin, DA, Dyer, CA, Hofeditz, C. The ovarian androgen producing cells: A review of structure/function relationships. Endocr Rev, 1985 July;6(3):371399.Google Scholar
Dennis, JE, Charbord, P. Origin and differentiation of human and murine stroma. Stem Cells, 2002 May;20(3):205214.Google Scholar
Reeves, G. Specific stroma in the cortex and medulla of the ovary. Cell types and vascular supply in relation to follicular apparatus and ovulation. Obstet Gynecol, 1971 June;37(6):832844.Google Scholar
Orisaka, M, Tajima, K, Mizutani, T et al. Granulosa cells promote differentiation of cortical stromal cells into theca cells in the bovine ovary. Biol Reprod, 2006 November 1;75(5):734740.Google Scholar
Jabara, S, Christenson, LK, Wang, CY et al. Stromal cells of the human postmenopausal ovary display a distinctive biochemical and molecular phenotype. J Clin Endocrinol Metab, 2003 January;88(1):484492.Google Scholar
Hummitzsch, K, Irving-Rodgers, HF, Hatzirodos, N et al. A new model of development of the mammalian ovary and follicles. PLoS One, 2013;8(2):e55578.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(May):110.Google Scholar
Knight, PG, Glister, C. TGF superfamily members and ovarian follicle development. Reproduction, 2006 August 1;132(2):191206.Google Scholar
Lee, W-S, Yoon, S-J, Yoon, T-K et al. Effects of bone morphogenetic protein-7 (BMP-7) on primordial follicular growth in the mouse ovary. Mol Reprod Dev, 2004 October;69(2):159163.Google Scholar
Dubreuil, G. Sur l’existence d’une substance inductrice a action limitee et locale pour la metaplasie theca des cellules du stroma cortical ovarien. Ann Endocrinol, 1948;434442.Google Scholar
Gurava, S. Biology of the Ovarian Follicle. New York: Springer-Verlag, 1985.CrossRefGoogle Scholar
Parrott, JA, Skinner, MK. Kit ligand actions on ovarian stromal cells: Effects on theca cell recruitment and steroid production. Mol Reprod Dev, 2000 January;55(1):5564.Google Scholar
Nilsson, EE, Skinner, MK. Kit ligand and basic fibroblast growth factor interactions in the induction of ovarian primordial to primary follicle transition. Mol Cell Endocrinol, 2004 February 12;214(1–2):1925.Google Scholar
Hillier, SG, Miró, F. Inhibin, activin, and follistatin. Potential roles in ovarian physiology. Ann N Y Acad Sci, 1993 May 28(687):2938.Google Scholar
Hayashi, M, McGee, EA, Min, G et al. Recombinant Growth Differentiation Factor-9 (GDF-9) enhances growth and differentiation of cultured early ovarian follicles. Endocrinology, 1999 March;140(3):12361244.Google Scholar
Lee, WS, Otsuka, F, Moore, RK, Shimasaki, S. Effect of bone morphogenetic protein-7 on folliculogenesis and ovulation in the rat. Biol Reprod, 2001 October;65(4):994999.Google Scholar
Nilsson, EE, Skinner, MK. Bone morphogenetic protein-4 acts as an ovarian follicle survival factor and promotes primordial follicle development. Biol Reprod, 2003 May 28;69(4):12651272.CrossRefGoogle ScholarPubMed
Cortvrindt, R, Smitz, J, Van Steirteghem, AC. In-vitro maturation, fertilization and embryo development of immature oocytes from early preantral follicles from prepuberal mice in a simplified culture system. Hum Reprod, 1996 December;11(12):26562666.Google Scholar
Picton, HM, Harris, SE, Muruvi, W, Chambers, EL. The in vitro growth and maturation of follicles. Reproduction, 2008 December 1;136(6):703715.Google Scholar
West, ER, Zelinski, MB, Kondapalli, LA, et al. Preserving female fertility following cancer treatment: current options and future possibilities. Pediatr Blood Cancer. 2009 August;53(2):289295.Google Scholar
Eppig, JJ. Mouse oocyte development in vitro with various culture systems. Dev Biol, 1977 October 15;60(2):371388.Google Scholar
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 May 1;68(5):16821686.Google Scholar
Kreeger, PK, Deck, JW, Woodruff, TK, Shea, LD. The in vitro regulation of ovarian follicle development using alginate-extracellular matrix gels. Biomaterials, 2006 February;27(5):714723.Google Scholar
Eppig, JJ, Schroeder, AC. Capacity of mouse oocytes from preantral follicles to undergo embryogenesis and development to live young after growth, maturation, and fertilization in vitro. Biol Reprod, 1989 Aug;41(2):268276.Google Scholar
Gomes, JE, Correia, SC, Gouveia-Oliveira, A, Cidadão, AJ, Plancha, CE. Three-dimensional environments preserve extracellular matrix compartments of ovarian follicles and increase FSH-dependent growth. Mol Reprod Dev, 1999 October 1;54(2):163172.Google Scholar
Cain, L, Chatterjee, S, Collins, TJ. In vitro folliculogenesis of rat preantral follicles. Endocrinology, 1995 August;136(8):33693377.Google Scholar
Nayudu, PL, Osborn, SM. Factors influencing the rate of preantral and antral growth of mouse ovarian follicles in vitro. Reproduction, 1992 July 1;95(2):349362.Google Scholar
Rowghani, NM, Heise, MK, McKeel, D et al. Maintenance of morphology and growth of ovarian follicles in suspension culture. Tissue Eng, 2004 March;10(3–4):545552.Google Scholar
Boland, NI, Humpherson, PG, Leese, HJ, Gosden, RG. Pattern of lactate production and steroidogenesis during growth and maturation of mouse ovarian follicles in vitro. Biol Reprod, 1993 April 1;48(4):798806.Google Scholar
Nation, A, Selwood, L. The production of mature oocytes from adult ovaries following primary follicle culture in a marsupial. Reproduction, 2009 August 1;138(2):247255.Google Scholar
Tibbitt, MW, Anseth, KS. Hydrogels as extracellular matrix mimics for 3D cell culture. Biotechnol Bioeng, 2009 July 1;103(4):655663.Google Scholar
Petersen, OW, Rønnov-Jessen, L, Howlett, AR, Bissell, MJ. Interaction with basement membrane serves to rapidly distinguish growth and differentiation pattern of normal and malignant human breast epithelial cells. Proc Natl Acad Sci U S A, 1992 October 1;89(19):90649068.Google Scholar
Weaver, VM, Fischer, AH, Peterson, OW, Bissell, MJ. The importance of the microenvironment in breast cancer progression: recapitulation of mammary tumorigenesis using a unique human mammary epithelial cell model and a three-dimensional culture assay. Biochem Cell Biol, 1996;74(6):833851.Google Scholar
Tanaka, H, Murphy, CL, Murphy, C et al. Chondrogenic differentiation of murine embryonic stem cells: Effects of culture conditions and dexamethasone. J Cell Biochem, 2004 October 15;93(3):454462.Google Scholar
Engler, AJ, Sen, S, Sweeney, HL, Discher, DE. Matrix elasticity directs stem cell lineage specification. Cell, 2006 August 25;126(4):677689.Google Scholar
Joo, S, Oh, SH, Sittadjody, S et al. The effect of collagen hydrogel on 3D culture of ovarian follicles. Biomed Mater, 2016 November 11;11(6):65009.Google Scholar
Xu, J, Lawson, MS, Yeoman, RR et al. Fibrin promotes development and function of macaque primary follicles during encapsulated three-dimensional culture. Hum Reprod, 2013 August 1;28(8):21872200.Google Scholar
Desai, N, Abdelhafez, F, Calabro, A, Falcone, T. Three dimensional culture of fresh and vitrified mouse pre-antral follicles in a hyaluronan-based hydrogel: a preliminary investigation of a novel biomaterial for in vitro follicle maturation. Reprod Biol Endocrinol, 2012 June 13;10(1):29.Google Scholar
Shikanov, A, Smith, RM, Xu, M, Woodruff, TK, Shea, LD. Hydrogel network design using multifunctional macromers to coordinate tissue maturation in ovarian follicle culture. Biomaterials, 2011;32(10):25242531.Google Scholar
Abir, R, Roizman, P, Fisch, B et al. Pilot study of isolated early human follicles cultured in collagen gels for 24 hours. Hum Reprod, 1999;14(5):12991301.Google Scholar
Torrance, C, Telfer, E, Gosden, RG. Quantitative study of the development of isolated mouse pre-antral follicles in collagen gel culture. J Reprod Fertil, 1989 September 1;87(1):367374.Google Scholar
Sharma, GT, Dubey, PK, Meur, SK. Survival and developmental competence of buffalo preantral follicles using three-dimensional collagen gel culture system. Anim Reprod Sci, 2009 August 1;114(1–3):115124.Google Scholar
Carroll, J, Whittingham, DG, Wood, MJ. Effect of gonadotrophin environment on growth and development of isolated mouse primary ovarian follicles. J Reprod Fertil, 1991 September;93(1):7179.Google Scholar
Hirao, Y, Nagai, T, Kubo, M et al. In vitro growth and maturation of pig oocytes. J Reprod Fertil, 1994 March;100(2):333339.Google Scholar
Rodgers, RJ, Irving-Rodgers, HF, Russell, DL. Extracellular matrix of the developing ovarian follicle. Reproduction, 2003 October;126(4):415424.Google Scholar
Pangas, Sa, Saudye, H, Shea, LD, Woodruff, TK. Novel approach for the three-dimensional culture of granulosa cell-oocyte complexes. Tissue Eng, 2003;9(5):10131021.Google Scholar
Xu, M, West, E, Shea, LD, Woodruff, TK. Identification of a stage-specific permissive in vitro culture environment for follicle growth and oocyte development. Biol Reprod, 2006 December 1;75(September):916923.Google Scholar
Erin, R, West Teresa, K, Woodruff, et al. Physical properties of alginate hydrogels and their effects on in vitro follicle development. Biomaterials, 2007;28(30):44394448.Google Scholar
Xu, M, West-Farrell, ER, Stouffer, RL et al. Encapsulated three-dimensional culture supports development of nonhuman primate secondary follicles. Biol Reprod, 2009 September 1;81(3):587594.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 October 1;24(10):25312540.Google Scholar
Xu, M, Kreeger, PK, Shea, LD, Woodruff, TK. Tissue-engineered follicles produce live, fertile offspring. Tissue Eng, 2006 October;12(10):27392746.Google Scholar
Shikanov, A, Xu, M, Woodruff, TK, Shea, LD. Interpenetrating fibrin–alginate matrices for in vitro ovarian follicle development. Biomaterials, 2009 October;30(29):54765485.Google Scholar
Augst, AD, Kong, HJ, Mooney, DJ. Alginate hydrogels as biomaterials. Macromol Biosci, 2006 August 7;6(8):623633.Google Scholar
Rowley, JA, Madlambayan, G, Mooney, DJ. Alginate hydrogels as synthetic extracellular matrix materials. Biomaterials, 1999 January;20(1):4553.Google Scholar
Skory, RM, Xu, Y, Shea, LD, Woodruff, TK. Engineering the ovarian cycle using in vitro follicle culture. Hum Reprod, 2015;30(6):13861395.Google Scholar
Silva, GM, Rossetto, R, Chaves, RN et al. In vitro development of secondary follicles from pre-pubertal and adult goats cultured in two-dimensional or three-dimensional systems. Zygote, 2015 August 26;23(4):475484.Google Scholar
Sadeghnia, S, Akhondi, MM, Hossein, G et al. Development of sheep primordial follicles encapsulated in alginate or in ovarian tissue in fresh and vitrified samples. Cryobiology, 2016 April 1;72(2):100105.Google Scholar
Songsasen, N, Woodruff, TK, Wildt, DE. In vitro growth and steroidogenesis of dog follicles are influenced by the physical and hormonal microenvironment. Reproduction, 2011 July;142(1):113122.Google Scholar
Jin, SY, Lei, L, Shikanov, A, Shea, LD, Woodruff, TK. A novel two-step strategy for in vitro culture of early-stage ovarian follicles in the mouse. Fertil Steril, 2010 May 15;93(8):26332639.Google Scholar
Amorim, CA, Van Langendonckt, A, David, A, Dolmans, M-M, Donnez, J. Survival of human pre-antral follicles after cryopreservation of ovarian tissue, follicular isolation and in vitro culture in a calcium alginate matrix. Hum Reprod, 2008 October 14;24(1):9299.Google Scholar
Osborn, SM, Gook, DA, Stern, K, Speirs, AL. The isolation and culture of human primordial follicles from fresh ovarian tissue. Hum Reprod, 1997 June 1;12(Suppl 2):192193.CrossRefGoogle Scholar
Itoh, T, Hoshi, H. Efficient isolation and long-term viability of bovine small preantral follicles in vitro. Vitr Cell Dev Biol – Anim, 2000 April;36(4):235.Google Scholar
Wu, MF, Huang, WT, Tsay, C et al. The stage-dependent inhibitory effect of porcine follicular cells on the development of preantral follicles. Anim Reprod Sci, 2002;73(1–2):7388.Google Scholar
Ramesh, HS, Gupta, PSP, Nandi, S et al. Co-culture of buffalo large preantral follicles with ovarian somatic cells. Adv Biol Res (Rennes), 2007;1(2):2933.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
Scarrit, ME, Pashos, NC, Bunnell, BA. A review of cellularization strategies for tissue engineering of whole organs. Front Bioeng Biotechnol, 2015;3:43.Google Scholar
Atala, A, Bauer, SB, Soker, S, Yoo, JJ, Retik, AB. Tissue-engineered autologous bladders for patients needing cystoplasty. Lancet (London, England), 2006 April 15;367(9518):12411246.Google Scholar
Hendriks, J, Riesle, J, van Blitterswijk, CA. Co-culture in cartilage tissue engineering. J Tissue Eng Regen Med, 2007 May;1(3):170178.Google Scholar
Soto-Gutiérrez, A, Navarro-Álvarez, N, Zhao, D et al. Differentiation of mouse embryonic stem cells to hepatocyte-like cells by co-culture with human liver nonparenchymal cell lines. Nat Protoc, 2007 February 1;2(2):347356.Google Scholar
Harimoto, M, Yamato, M, Hirose, M et al. Novel approach for achieving double-layered cell sheets co-culture: overlaying endothelial cell sheets onto monolayer hepatocytes utilizing temperature-responsive culture dishes. J Biomed Mater Res, 2002 December 5;62(3):464470.Google Scholar
Robinson, SN, Ng, J, Niu, T et al. Superior ex vivo cord blood expansion following co-culture with bone marrow-derived mesenchymal stem cells. Bone Marrow Transplant, 2006 February 9;37(4):359366.Google Scholar
Luk, JM, Wang, PP, Lee, CK, Wang, JH, Fan, ST. Hepatic potential of bone marrow stromal cells: Development of in vitro co-culture and intra-portal transplantation models. J Immunol Methods, 2005 October 20;305(1):3947.Google Scholar
Houchin-Ray, T, Zelivyanskaya, M, Huang, A, Shea, LD. Non-viral gene delivery transfection profiles influence neuronal architecture in an in vitro co-culture model. Biotechnol Bioeng, 2009 August 1;103(5):10231033.Google Scholar
Dominguez, F, Gadea, B, Mercader, A et al. Embryologic outcome and secretome profile of implanted blastocysts obtained after coculture in human endometrial epithelial cells versus the sequential system. Fertil Steril, 2010 February;93(3):774–782.e1.Google Scholar
Godard, NM, Pukazhenthi, BS, Wildt, DE, Comizzoli, P. Paracrine factors from cumulus-enclosed oocytes ensure the successful maturation and fertilization in vitro of denuded oocytes in the cat model. Fertil Steril, 2009 May;91(5):20512060.Google Scholar
Reubinoff, BE, Pera, MF, Fong, C-Y, Trounson, A, Bongso, A. Embryonic stem cell lines from human blastocysts: somatic differentiation in vitro. Nat Biotechnol, 2000 April 1;18(4):399404.Google Scholar
Thomson, JA, Itskovitz-Eldor, J, Shapiro, SS et al. Embryonic stem cell lines derived from human blastocysts. Science, 1998 November 6;282(5391):11451147.Google Scholar

References

Delvigne, A, Rozenberg, S. Review of clinical course and treatment of ovarian hyperstimulation syndrome (OHSS). Hum Reprod Update, 2003;9:7796.Google Scholar
Delvigne, A, Rozenberg, S. Epidemiology and prevention of ovarian hyperstimulation syndrome (OHSS): a review. Hum Reprod Update, 2002;8:559577.Google Scholar
Child, TJ, Abdul-Falil, AK, Gulekli, B et al. In vitro maturation and fertilization of oocytes from unstimulated normal ovaries, polycystic ovaries, and women with polycystic ovary syndrome. Fertil Steril, 2001;76:936942.Google Scholar
Engmann, L, DiLuigi, A, Schmidt, D et al. The use of gonadotropin-releasing hormone (GnRH) agonist to induce oocyte maturation after cotreatment with GnRH antagonist in high-risk patients undergoing in vitro fertilization prevents the risk of ovarian hyperstimulation syndrome: a prospective randomized controlled study. Fertil Steril, 2008;89:8491.Google Scholar
Trounson, A, Wood, C, Kausche, A. In vitro maturation and the fertilization and developmental competence of oocytes recovered from untreated polycystic ovarian patients. Fertil Steril, 1994;62:353362.Google Scholar
Ho, VNA, Pham, TD, Le, AH, Ho, TM, Vuong, LN. Live birth rate after human chorionic gonadotropin priming in vitro maturation in women with polycystic ovary syndrome. J Ovarian Res, 2018 August 27;11(1):767–711.Google Scholar
Smitz, J, Picton, HM, Platteau, P et al. Principal findings from a multicenter trial investigating the safety of follicular-fluid meiosis-activating sterol for in vitro maturation of human cumulus-enclosed oocytes. Fertil Steril, 2007;87:949964.Google Scholar
Banwell, KM, Thompson, JG. In vitro maturation of mammalian oocytes: outcomes and consequences. Semin Reprod Med, 2008;26:162174.Google Scholar
Filali, M, Hesters, L, Franchin, R et al. Retrospective comparison of two media for in vitro maturation of oocytes. Reprod Biomed Online, 2008;16:250256.Google Scholar
Fadini, R, Dal Canto, MB, Mignini Renzini, M et al. Effect of different gonadotrophin priming on IVM of oocytes from women with normal ovaries: a prospective randomized study. Reprod Biomed Online, 2009;19:343351.Google Scholar
Mikkelsen, AL, Smith, S, Lindenberg, S. Possible factors affecting the development of oocytes in in vitro maturation. Hum Reprod, 2000;15:1117.Google Scholar
Nogueira, D, Ron-El, R, Friedler, S et al. Meiotic arrest in vitro by phosphodiesterase 3-inhibitor enhances maturation capacity of human oocytes and allows subsequent embryonic development. Biol Reprod, 2006;74:177184.Google Scholar
Albuz, FK, Sasseville, M, Lane, M et al. Simulated physiological oocyte maturation (SPOM): a novel in vitro maturation system that substantially improves embryo yield and pregnancy outcomes. Human Reproduction, 2010 December;25(12):29993011.Google Scholar
Sanchez, F, Lolicato, F, Romero, S et al. An improved IVM method for cumulus-oocyte complexes from small follicles in polycystic ovary syndrome patients enhances oocyte competence and embryo yield. Hum Reprod, 2017 August 30;32(10):20562068.Google Scholar
Park, JY, Su, YQ, Ariga, M et al. EGF-like growth factors as mediators of LH action in the ovulatory follicle. Science, 2004;303:682684.Google Scholar
Pincus, G, Enzmann, EV. The comparative behaviour of mammalian eggs in vitro and in vivo.II The activation of tubal eggs in the rabbit. J Exp Med, 1935;62:665675.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.Google Scholar
Chian, RC, Gulekli, B, Buckett, WM et al. Priming with human chorionic gonadotropin before retrieval of immature oocytes in women with infertility due to the polycystic ovary syndrome. N Engl J Med, 1999;341:16241626.Google Scholar
Chian, RC, Buckett, WM, Tan, SL. In-vitro maturation of human oocytes. Reprod Biomed Online, 2004;8:148166.Google Scholar
Suikkari, AM, Söderström-Anttila, V. In-vitro maturation of eggs: is it really useful? Best Pract Res Clin Obst Gyn, 2007;21:145155.Google Scholar
de Paula Martins, W, dos Reis, RM, Ferriani, RA et al. Endometrial preparation for in vitro oocyte maturation: early use of estrogen increases endometrial tissue and requires lower daily dosage: a cross over trial in ‘mock’ cycles. J Assist Reprod Genet, 2006;23:241246.Google Scholar
Walls, ML, Hunter, T, Ryan, JP et al. In vitro maturation as an alternative to standard in vitro fertilization for patients diagnosed with polycystic ovaries: a comparative analysis of fresh, frozen and cumulative cycle outcomes. Hum Reprod, 2015;30:8896.Google Scholar
De Vos, M, Smitz, J, Thompson, JG, Gilchrist, RB. The definition of IVM is clear—variations need defining. Hum Reprod, 2016 October 21;31(11):24112415.Google Scholar
Gilchrist, RB, Thompson, JG. Oocyte maturation: emerging concepts and technologies to improve developmental potential in vitro. Theriogenology, 2007;67:615.Google Scholar
Edwards, RG. Maturation in vitro of human ovarian oocytes. Lancet, 1965;2 926929.Google Scholar
Eppig, JJ. The participation of cyclic adenosine monophosphate (cAMP) in the regulation of meiotic maturation of oocytes in the laboratory mouse. J Reprod Fertil Suppl, 1989;38:38.Google Scholar
Dekel, N. Regulation of oocyte maturation. The role of cAMP. Ann NY Acad Sci, 1988;541:211216.Google Scholar
Thomas, RE, Armstrong, DT, Gilchrist, RB. Bovine cumulus cell-oocyte gap junctional communication during in vitro maturation in response to manipulation of cell-specific cyclic adenosine 30, 50-monophosophate levels. Biol Reprod, 2004;70:548556.Google Scholar
35. Thomas, RE, Thompson, JG, Armstrong, DT et al. Effect of specific phosphodiesterase isoenzyme inhibitors during in vitro maturation of bovine oocytes on meiotic and developmental capacity. Biol Reprod, 2004;71:11421149.Google Scholar
Luciano, AM, Modina, S, Vassena, R et al. Role of intracellular cyclic adenosine 30, 50- monophosphate concentration and oocyte-cumulus cells communications on the acquisition of the developmental competence during in vitro maturation of bovine oocyte. Biol Reprod, 2004;70: 465472.Google Scholar
Mottershead, DG, Sugimura, S, Al-Musawi, SL et al. Cumulin, an oocyte-secreted heterodimer of the transforming growth factor-β family, is a potent activator of granulosa cells and improves oocyte quality. J Biol Chem, 2015 September 25;290(39):2400724020.Google Scholar
Zeng, H-T, Ren, Z, Guzman, L et al. Heparin and cAMP modulators interact during pre-in vitro maturation to affect mouse and human oocyte meiosis and developmental competence. Hum Reprod,. 2013 June;28(6):15361545.Google Scholar
Sanchez, F, Lolicato, F, Romero, S et al. An improved IVM method for cumulus-oocyte complexes from small follicles in polycystic ovary syndrome patients enhances oocyte competence and embryo yield. Hum Reprod, 2017 October 1;32(10):20562068.Google Scholar
Grynberg, M, Poulain, M, le Parco, S et al. Similar in vitromaturation rates of oocytes retrieved during the follicular or luteal phase offer flexible options for urgent fertility preservation in breast cancer patients. Hum Reprod, 2016;31(3):623629.Google Scholar
Creux, H, Monnier, P, Son, W-Y, Buckett, W. Thirteen years’ experience in fertility preservation for cancer patients after in vitro fertilization and in vitro maturation treatments. J Assist Reprod Genet, 2018 March;2(97):110.Google Scholar
Revel, A, Koler, M, Simon, A et al. Oocyte collection during cryopreservation of the ovarian cortex. Fertil Steril, 2003;79:12371239.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
Uzelac, PS, Delaney, AA, Christensen, GL et al. Live birth following in vitro maturation of oocytes retrieved from extracorporeal ovarian tissue aspiration and embryo cryopreservation for 5 years. Fertil Steril, 2015;104:12581260.Google Scholar
Donnez, J, Dolmans, M-M. Fertility preservation in women. N Engl J Med, 2017 October 26;377(17):16571665.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
Rosendahl, M, Greve, T, Andersen, CY. The safety of transplanting cryo-preserved ovarian tissue in cancer patients: a review of the literature. J Assist Reprod Genet, 2013;30:1124.Google Scholar
Anderson, RA, McLaughlin, M, Wallace, WHB et al. The immature human ovary shows loss of abnormal follicles and increasing follicle developmental competence through childhood and adolescence. Hum Reprod, 2014;29:97106.Google Scholar
Demeestere, I, Simon, P, Dedeken, L et al. Live birth after autograft of ovarian tissue cryopreserved during childhood. Hum Reprod, 2015;30:21072109.Google Scholar
Rose, BI. Approaches to oocyte retrieval for advanced reproductive technology cycles planning to utilize in vitro maturation: a review of the many choices to be made. J Assist Reprod Genet, 2014;31:14091419.Google Scholar
Huang, JY, Buckett, WM, Gilbert, L Tan SL, Chian RC, Retrieval of immature oocytes followed by in vitro maturation and vitrification: A case report on a new strategy of fertility preservation in women with borderline ovarian malignancy. Gynecol Oncol, 2007;105:542544.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
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.Google Scholar
Telfer, EE, Zelinski, MB. Ovarian follicle culture: advances and challenges for human and nonhuman primates. Fertil Steril, 2013;99:15231533.Google Scholar
Garcia-Velasco, JA, Domingo, J, Cobo, A et al. Five years’ experience using oocyte vitrification to preserve fertility for medical and non-medical indications. Fertil Steril, 2013;99:19941999.Google Scholar
Johnson, RH, Chien, FL, Bleyer, A. Incidence of breast cancer with distant involvement among women in the United States, 1976 to 2009. Jama, 2013;309(8):800805. DOI:10.1001/jama.2013.776Google Scholar
Merlo, DF, Ceppi, M, Filiberti, R et al. Breast cancer incidence trends in European women aged 20–39 years at diagnosis. Breast Cancer Res Treat, 2012;134(1):363370.Google Scholar
Peccatori, FA, Azim, HA, Orecchia, R, et al. Cancer, pregnancy and fertility: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Annals of Oncology: Official Journal of the European Society for Medical Oncology / ESMO, 2013;Vol. 24(Suppl 6):vi160vi170.Google Scholar
Gilchrist, R, Rizk BRMB, Falcone T. Current status and future trends of the clinical practice of human oocyte in vitro maturation. In Gardner D, Smitz JEJ, Thompson JG (eds.) Human Assisted Reproductive Technology: Future Trends in Laboratory and Clinical Practice. Cambridge: Cambridge University Press. 2011, 186–198.Google Scholar
Sinclair, KD, Young, LE, Wilmut, I et al. In-utero overgrowth in ruminants following embryo culture: lessons from mice and a warning to men. Hum Reprod, 2000;5:6886.Google Scholar
Kuhtz, J, Romero, S, De Vos, M et al. Human in vitro oocyte maturation is not associated with increased imprinting error rates at LIT1, SNRPN, PEG3 and GTL2. Hum Reprod, 2014;29:19952005.Google Scholar
Spits, C, Guzman, L, Mertzanidou, A et al. Chromosome constitution of human embryos generated after in vitro maturation including 3-isobutyl-1-methylxanthine in the oocyte collection medium. Hum Reprod, 2015;30:653663.Google Scholar

References

The ESHRE Working Group on Oocyte Cryopreservation in Europe, Shenfield, F, de Mouzon, J, Scaravelli, G et al. Oocyte and ovarian tissue cryopreservation in European countries: statutory background, practice, storage and use. Hum Reprod Open, 2017;2017:hox003.Google Scholar
Depalo, R, Nappi, L, Loverro, G et al. Evidence of apoptosis in human primordial and primary follicles. Hum Reprod, 2003;18:26782682.Google Scholar
McLaughlin, EA, McIver, SC. Awakening the oocyte: controlling primordial follicle development. Reproduction, 2009;137:111.Google Scholar
Otala, M, Erkkilä, K, Tuuri, T et al. Cell death and its supression in human ovarian tissue culture. Mol Hum Reprod, 2002;8:228236.CrossRefGoogle Scholar
Isachenko, V, Montag, M, Isachenko, E et al. Effective method for in-vitro culture of cryopreserved human ovarian tissue. Reprod Biomed Online, 2006;13:228234.Google Scholar
Sadeu, JC, Smitz, J. Growth differentiation factor-9 and anti-Müllerian hormone expression in cultured human follicles from frozen–thawed ovarian tissue. Reprod Biomed Online, 2008;17:537548.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
Hovatta, O, Silye, R, Abir, R, Krausz, T, Winston, RML. 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
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
Hovatta, O, Wright, C, Krausz, T, Hardy, K, Winston, RML. Human primordial, primary and secondary ovarian follicles in long-term culture: effect of partial isolation. Hum Reprod, 1999;14:25192524.Google Scholar
Louhio, H, Hovatta, O, Sjöberg, J, Tuuri, T. The effects of insulin, and insulin-like growth factors I and II on human ovarian follicles in long-term culture. Mol Hum Reprod, 2000;6:694698.Google Scholar
Rahimi, G, Isachenko, E, Sauer, H et al. Measurement of apoptosis in long-term cultures of human ovarian tissue. Reproduction, 2001;122:657663.Google Scholar
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.Google Scholar
Isachenko, E, Isachenko, V, Rahimi, G, Nawroth, F. Cryopreservation of human ovarian tissue by direct plunging into liquid nitrogen. Eur J Obstet Gynecol Reprod Biol, 2003;108:186193.Google Scholar
Biron-Shental, T, Fisch, B, Van Den Hurk, R et al. Survival of frozen-thawed human ovarian fetal follicles in long-term organ culture. Fertil Steril, 2004;81:716719.Google Scholar
Otala, M, Mäkinen, S, Tuuri, T et al. Effects of testosterone, dihydrotestosterone, and 17β-estradiol on human ovarian tissue survival in culture. Fertil Steril, 2004;82:10771085.Google Scholar
Scott, JE, Carlsson, IB, Bavister, BD, Hovatta, O. Human ovarian tissue cultures: extracellular matrix composition, coating density and tissue dimensions. Reprod Biomed Online, 2004;9:287293.Google Scholar
Scott, JE, Zhang, P, Hovatta, O. benefits of 8-bromo-guanosine 3’,5’-cyclic monophosphate (8-br-cGMP) in human ovarian cortical tissue culture. Reprod Biomed Online, 2004;8:319324.Google Scholar
Zhang, P, Louhio, H, Tuuri, T et al. In vitro effect of cyclic adenosine 3’,5’-monophosphate (cAMP) on early human ovarian follicles. J Assist Reprod Genet, 2004;21:301306.Google Scholar
Schmidt, KLT, Kryger-Baggasen, N, Byskov, AG, Andersen, CY. Anti-Müllerian hormone initiates growth of human primordial follicles in vitro. Mol Cell Endocrinol, 2005;234:8793.Google Scholar
Carlsson, IB, Laitinen, MPE, Scott, JE 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;131:641649.Google Scholar
Carlsson, IB, Scott, JE, Visser, JA et al. Anti-Müllerian hormone inhibits initiation of growth of human primordial ovarian follicles in vitro. Hum Reprod, 2006;21:22232227.Google Scholar
Sadeu, JC, Cortvrindt, R, Ron-El, R, Kasterstein, E, Smitz, J. Morphological and ultrastructural evaluation of cultured frozen-thawed human fetal ovarian tissue. Fertil Steril, 2006;85:11301141.Google Scholar
Isachenko, V, Isachenko, E, Reinsberg, J et al. Cryopreservation of human ovarian tissue: comparison of rapid and conventional freezing. Cryobiology, 2007;55:261268.Google Scholar
Morimoto, Y, Oku, Y, Sonoda, M et al. High oxygen atmosphere improves human follicle development in organ cultures of ovarian cortical tissues in vitro. Hum Reprod, 2007;22:31703177.Google Scholar
Telfer, EE, McLaughlin, M, Ding, C, Joo Thong, K. 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
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
Kedem, A, Hourvitz, A, Fisch, B et al. Alginate scaffold for organ culture of cryopreserved-thawed human ovarian cortical follicles. J Assist Reprod Genet, 2011;28:761769.Google Scholar
Kedem, A, Fisch, B, Garor, R et al. Development of human primordial follicles in vitro, with seemingly more beneficial effects of GDF9. J Clin Endocrinol Metab, 2011;96:E1246-E1254.Google Scholar
McLaughlin, M, Patrizio, P, Kayisli, U et al. mTOR kinase inhibition results in oocyte loss characterized by empty follicles in human ovarian cortical strips cultured in vitro. Fertil Steril, 2011;96:11541159.Google Scholar
Lerer-Serfaty, G, Samara, N, Risch, B et al. Attempted application of bioengineered/biosynthetic supporting matrices with phosphatidylinositol-trisphosphate-enhancing substances to organ culture of human primordial follicles. J Assist Reprod Genet, 2013;30:12791288.Google Scholar
Anderson, RA, McLaughlin, M, Wallace, WHB, 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.Google Scholar
Laronda, MM, Duncan, FE, Hornick, JE et al. Alginate encapsulation supports the growth and differentiation of human primordial follicles within ovarian cortical tissue. J Assist Reprod Genet, 2014;31:10131028.Google Scholar
Lande, Y, Fisch, B, Tsur, A et al. Short-term exposure of human ovarian follicles to cyclophosphamide metabolites seems to promote follicular activation in vitro. Reprod Biomed Online, 2017;34:104114.Google Scholar
Younis, AJ, Lerer-Serfaty, G, Stav, D et al. Extracellular-like matrices and leukemia inhibitory factor for in vitro culture of human primordial follicles. Reprod Fertil Dev, 2017;29:19821994.Google Scholar
Talevi, R, Sudhakaran, S, Barbato, V et al. Is oxygen availability a limiting factor for in vitro folliculogenesis? PLoS One, 2018;13:e0192501.Google Scholar
Schubert, B, Smitz, J. In vitro culture of human primordial follicles. In Chian, RC, Quinn, P (eds.) Fertility Cryopreservation. Cambridge: Cambridge University. 2010, 200212.Google Scholar
Da Silva-Buttkus, P, Marcelli, G, Franks, S, Stark, J, Hardy, K. Inferring biological mechanisms from spatial analysis: prediction of a local inhibitor in the ovary. Proc Natl Acad Sci, 2009;106:456461.Google Scholar
Chiti, MC, Dolmans, MM, Mortiaux, L et al. A novel fibrin-based artificial ovary prototype resembling human ovarian tissue in terms of architecture and rigidity. J Assist Reprod Genet, 2018;35:4148.Google Scholar
Schmidt, KL, Byskov, AG, Nyboe, Andersen A, Müller, J, Yding, Andersen C. Density and distribution of primordial follicles in single pieces of cortex from 21 patients and in individual pieces of cortex from three entire human ovaries. Hum Reprod, 2003;18:11581164.Google Scholar
Chambers, EL, Gosden, RG, Yap, C, Picton, HM. In situ identification of follicles in ovarian cortex as a tool for quantifying follicle density, viability and developmental potential in strategies to preserve female fertility. Hum Reprod, 2010;25:25592568.Google Scholar
Peters, ITA, Stegehuis, PL, Peek, R et al. Noninvasive detection of metastases and follicle density in ovarian tissue using full-field optical coherence tomography. Clin Cancer Res, 2016;22:55065513.Google Scholar
Guérard, M, Zeller, A, Singer, T, Gocke, E. In vitro genotoxicity of neutral red after photo-activation and metabolic activation in the Ames test, the micronucleous test and the comet assay. Mutat Res, 2012;746:1520.Google Scholar
Fischer, BB, Krieger-Liszkay, A, Eggen, RIL. Photosensitizers neutral red (type I) and rose bengal (type II) cause light-dependent toxicity in Chlamydomonas reinhardtii and induce the Gpxh gene via increased singlet oxygen formation. Environ Sci Technol, 2004;38:63076313.Google Scholar
Langbeen, A, Jorssen, EPA, Granata, N et al. Effects of neutral red assisted viability assessment on the cryotolerance of isolated bovine preantral follicles. J Assist Reprod Genet, 2014;31:17271736.Google Scholar
Bulgarelli, DL, Ting, AY, Gordon, BJ, de Sá Rosa-e-Silva, ACJ, Zelinski, MB. Development of macaque secondary follicles exposed to neutral red prior to 3-dimensional culture. J Assist Reprod Genet, 2018;35:7179.Google Scholar
Abir, R, Fisch, B, Nitke, S et al. Morphological study of fully and partially isolated early human follicles. Fertil Steril, 2001;75:141146.Google Scholar
Amorim, CA, Van Langendonckt, A, David, A, Dolmans, MM, Donnez, J. Survival of human pre-antral follicles after cryopreservation of ovarian tissue, follicular isolation and in vitro culture in a calcium alginate matrix. Hum Reprod, 2009;24:9299.Google Scholar
Vanacker, J, Camboni, A, Dath, C et al. Enzymatic isolation of human primordial and primary ovarian follicles with Liberase DH: protocol for application in a clinical setting. Fertil Steril, 2011;96:379–383.e3.Google Scholar
Camboni, A, Van Langendonckt, A, Donnez, J et al. Alginate beads as a tool to handle, cryopreserve and culture isolated human primordial/primary follicles. Cryobiology, 2013;67:6469.Google Scholar
Vanacker, J, Luyckx, V, Amorim, C et al. Should we isolate human preantral follicles before or after cryopreservation of ovarian tissue? Fertil Steril, 2013;99:13631368.e2.Google Scholar
Abir, R, Roizman, P, Fisch, B et al. Pilot study of isolated early human follicles cultured in collagen gels for 24 hours. Hum Reprod, 1999;14:12991301.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.Google Scholar
Schmidt, VM, Isachenko, V, Rappl, G et al. Comparison of the enzymatic efficiency of Liberase TM and tumor dissociation enzyme: effect on the viability of cells digested from fresh and cryopreserved human ovarian cortex. Reprod Biol Endocrinol, 2018;16:PMC5985056.Google Scholar
Chiti, MC, Dolmans, MM, Hobeika, M et al. A modified and tailored human follicle isolation procedure improves follicle recovery and survival. J Ovarian Res, 2017;10:71.Google Scholar
Roy, SK, Treacy, BJ. Isolation and long-term culture of human preantral follicles. Fertil Steril, 1993;59:783790.Google Scholar
Fabbri, R, Pasquinelli, G, Keane, D et al. Culture of cryopreserved ovarian tissue: state of the art in 2008. Fertil Steril, 2009;91:16191629.Google Scholar
Paulini, F, Vilela, JM, Chiti, MC et al. Survival and growth of human preantral follicles after cryopreservation of ovarian tissue, follicle isolation and short-term xenografting. Reprod Biomed Online, 2016;33:425432.Google Scholar
Kim, CH, Cheon, YP, Lee, YJ et al. The effect of fibroblast co-culture on in vitro maturation of mouse preantral follicles. Dev Reprod, 2013;17:269274.Google Scholar
Tagler, D, Tu, T, Smith, RM et al. Embryonic fibroblasts enable the culture of primary ovarian follicles within alginate hydrogels. Tissue Eng Part A, 2012;18:12291238.Google Scholar
Tingen, CM, Kiesewetter, SE, Jozefik, J et al. A macrophage and theca cell-enriched stromal cell population influences growth and survival of immature murine follicles in vitro. Reproduction, 2011;141:809820.Google Scholar
Hornick, JE, Duncan, FE, Shea, LD, Woodruff, TK. Multiple follicle culture supports primary follicle growth through paracrine-acting signals. Reproduction, 2013;145:1932.Google Scholar
Guzel, Y, Oktem, O. Understanding follicle growth in vitro: Are we getting closer to obtaining mature oocytes from in vitro-grown follicles in human? Mol Reprod Dev, 2017;84:544559.Google Scholar
Picton, HM, Gosden, RG. In vitro growth of human primordial follicles from frozen-banked ovarian tissue. Mol Cell Endocrinol, 2000;166:2735.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.Google Scholar
Amorim, CA, Shikanov, A. The artificial ovary: current status and future perspectives. Future Oncol, 2016;12:23232332.Google Scholar
Ouni, E, Vertommen, D, Chiti, MC, Dolmans, MM, Amorim, CA. A draft map of the human ovarian proteome for tissue engineering and clinical applications. Mol Cell Proteomics, 2019;18(1):S 159–S173 DOI:10.1074/mcp.RA117.000469.Google Scholar
Caliari, SR, Burdick, JA. A practical guide to hydrogels for cell culture. Nat Methods, 2016;13:405414.Google Scholar
Sun, J, Tan, H. Alginate-based biomaterials for regenerative medicine applications. Materials, 2013;6:12851309.Google Scholar
Stein, H, Wilensky, M, Tsafrir, Y et al. Production of bioactive, post-translationally modified, heterotrimeric, human recombinant type-I collagen in transgenic tobacco. Biomacromolecules, 2009;10:26402645.Google Scholar
Peled, E, Boss, J, Bejar, J, Zinman, C, Seliktar, D. A novel poly(ethylene glycol)-fibrinogen hydrogel for tibial segmental defect repair in a rat model. J Biomed Mater Res A, 2007;80:874884.Google Scholar
Smitz, J, Dolmans, MM, Donnez, J et al. Current achievements and future research directions in ovarian tissue culture, in vitro follicle development and transplantation: implications for fertility preservation. Hum Reprod Update, 2010;16:395414.Google Scholar
Gigli, I, Byrd, DD, Fortune, JE. Effects of oxygen tension and supplements to the culture medium on activation and development of bovine follicles in vitro. Theriogenology, 2006;66:344353.Google Scholar
Meyvantsson, I, Beebe, DJ. Cell culture in microfluidic systems. Annu Rev Anal Chem, 2008;1:423449.Google Scholar
Mehling, M, Tay, S. Microfluidic cell culture. Curr Opin Biotechnol, 2014;25:95102.Google Scholar
Nagashima, JB, El Assal, R, Songsasen, N, Demirci, U. Evaluation of an ovary-on-a-chip in large mammalian models: Species specificity and influence of follicle isolation status. J Tissue Eng Regen Med, 2018;12:e1926e1935.Google Scholar
Aziz, AUR, Fu, M, Deng, J et al. A microfluidic device for culturing an encapsulated ovarian follicle. Micromachines, 2017;8:335.Google Scholar
Navis, AR. Hanging drop tissue culture. Embryo Project Encyclopedia (2007–11-08). ISSN: 1940–5030 http://embryo.asu.edu/handle/10776/1720Google Scholar
Choi, JK, Agarwal, P, He, X. In vitro culture of early secondary preantral follicles in hanging drop of ovarian cell-conditioned medium to obtain MII oocytes from outbred deer mice. Tissue Eng Part A, 2013;19:26262637.Google Scholar
Millet, LJ, Gillette, MU. Over a century of neuron culture: from the hanging drop to microfluidic devices. Yale J Biol Med, 2012;85:501521.Google Scholar
Wycherley, G, Downey, D, Kane, MT, Hynes, AC. A novel follicle culture system markedly increases follicle volume, cell number and oestradiol secretion. Reproduction, 2004;127:669677.Google Scholar
He, X, Toth, TL. In vitro culture of ovarian follicles from Peromyscus. Semin Cell Dev Biol, 2017;61:140149.Google Scholar
Wang, W, Tang, Y, Ni, L et al. A modified protocol for in vitro maturation of mouse oocytes from secondary preantral follicles. Adv Biosci Biotechnol, 2012;3:17194.Google Scholar
Peroglio, M, Gaspar, D, Zeugolis, DI, Alini, M. Relevance of bioreactors and whole tissue cultures for the translation of new therapies to humans. J Orthop Res, 2018;36:1021.Google Scholar
Zanotelli, MR, Henningsen, JD, Hopkins, PM et al. An ovarian bioreactor for in vitro culture of the whole bovine ovary: a preliminary report. J Ovarian Res, 2016;9:47.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
×