. References and . .. Bibliographiques,

. .. Annexes,

P. Reddy, W. Zheng, and K. Liu, Mechanisms maintaining the dormancy and survival of mammalian primordial follicles, Trends Endocrinol. Metab. TEM, vol.21, pp.96-103, 2010.

D. Adhikari and K. Liu, Molecular mechanisms underlying the activation of mammalian primordial follicles, Endocr. Rev, vol.30, pp.438-464, 2009.

A. J. Hsueh, K. Kawamura, Y. Cheng, and B. C. Fauser, Intraovarian control of early folliculogenesis, Endocr. Rev, vol.36, pp.1-24, 2015.

J. A. Visser and A. P. Themmen, Role of anti-Müllerian hormone and bone morphogenetic proteins in the regulation of FSH sensitivity, Mol. Cell. Endocrinol, vol.382, pp.460-465, 2014.

A. L. Durlinger, P. Kramer, B. Karels, F. H. De-jong, J. T. Uilenbroek et al., Control of primordial follicle recruitment by antiMüllerian hormone in the mouse ovary, Endocrinology, vol.140, pp.5789-5796, 1999.

I. B. Carlsson, J. E. Scott, J. A. Visser, O. Ritvos, A. P. Themmen et al., Anti-Müllerian hormone inhibits initiation of growth of human primordial ovarian follicles in vitro, Hum. Reprod. Oxf. Engl, vol.21, pp.2223-2227, 2006.

M. Y. Yang, R. A. Cushman, and J. E. Fortune, Anti-Müllerian hormone inhibits activation and growth of bovine ovarian follicles in vitro and is localized to growing follicles, Mol. Hum. Reprod, pp.1-10, 2017.

E. Nilsson, N. Rogers, and M. K. Skinner, Actions of anti-Mullerian hormone on the ovarian transcriptome to inhibit primordial to primary follicle transition, Reprod. Camb. Engl, vol.134, pp.209-221, 2007.

A. L. Durlinger, M. J. Gruijters, P. Kramer, B. Karels, H. A. Ingraham et al., , 2002.

, Anti-Müllerian hormone inhibits initiation of primordial follicle growth in the mouse ovary, Endocrinology, vol.143, pp.1076-1084

I. Gigli, R. A. Cushman, C. M. Wahl, and J. E. Fortune, Evidence for a role for anti-Mullerian hormone in the suppression of follicle activation in mouse ovaries and bovine ovarian cortex grafted beneath the chick chorioallantoic membrane, Mol. Reprod. Dev, vol.71, pp.480-488, 2005.

M. De-vos, P. Devroey, and B. C. Fauser, Primary ovarian insufficiency, Lancet Lond. Engl, vol.376, pp.911-921, 2010.

J. Donnez, B. Martinez-madrid, P. Jadoul, A. Van-langendonckt, D. Demylle et al., Ovarian tissue cryopreservation and transplantation: a review, Hum. Reprod. Update, vol.12, pp.519-535, 2006.

G. Bedoschi, P. A. Navarro, and K. Oktay, Chemotherapy-induced damage to ovary: mechanisms and clinical impact, Future Oncol. Lond. Engl, vol.12, pp.2333-2344, 2016.

H. Roness, L. Kalich-philosoph, and D. Meirow, Prevention of chemotherapyinduced ovarian damage: possible roles for hormonal and non-hormonal attenuating agents, Hum. Reprod. Update, vol.20, pp.759-774, 2014.

S. Morgan, R. A. Anderson, C. Gourley, W. H. Wallace, and N. Spears, How do chemotherapeutic agents damage the ovary?, Hum. Reprod. Update, vol.18, pp.525-535, 2012.

J. Assi, J. Santos, T. Bonetti, P. C. Serafini, E. L. Motta et al., Psychosocial benefits of fertility preservation for young cancer patients, J. Assist. Reprod. Genet, 2018.

, Fertility preservation in patients undergoing gonadotoxic therapy or gonadectomy: a committee opinion, Practice Committee of American Society for Reproductive Medicine, vol.100, pp.1214-1223, 2013.

W. H. Wallace and T. W. Kelsey, Human ovarian reserve from conception to the menopause

, PloS One, vol.5, p.8772, 2010.

P. Reddy, W. Zheng, and K. Liu, Mechanisms maintaining the dormancy and survival of mammalian primordial follicles, Trends Endocrinol. Metab. TEM, vol.21, pp.96-103, 2010.

D. Monniaux, The ovarian reserve of primordial follicles and the dynamic reserve of antral growing follicles: what is the link?, Biol. Reprod, vol.90, p.85, 2014.
URL : https://hal.archives-ouvertes.fr/hal-00961546

J. L. Tilly, Ovarian follicle counts--not as simple as 1, 2, 3, Reprod. Biol. Endocrinol. RBE, vol.1, p.11, 2003.

M. Myers, K. L. Britt, N. G. Wreford, F. J. Ebling, and J. B. Kerr, Methods for quantifying follicular numbers within the mouse ovary, Reprod. Camb. Engl, vol.127, pp.569-580, 2004.

C. Casari, Accelerated uptake of VWF/platelet complexes in macrophages contributes to VWD type 2B-associated thrombocytopenia, Blood, vol.122, pp.2893-2902, 2013.

H. Sharma, N. Zerbe, I. Klempert, O. Hellwich, and P. Hufnagl, Deep convolutional neural networks for automatic classification of gastric carcinoma using whole slide images in digital histopathology, Comput. Med. Imaging Graph. Off. J. Comput. Med. Imaging Soc, 2017.

M. E. Vandenberghe, Relevance of deep learning to facilitate the diagnosis of HER2 status in breast cancer, Sci. Rep, vol.7, p.45938, 2017.

Y. Lecun, Y. Bengio, and G. Hinton, Deep learning, Nature, vol.521, pp.436-444, 2015.

T. Araújo, Classification of breast cancer histology images using Convolutional Neural Networks, PloS One, vol.12, p.177544, 2017.

D. C. Cire?an, A. Giusti, L. M. Gambardella, and J. Schmidhuber, Mitosis detection in breast cancer histology images with deep neural networks, Med. Image Comput. Comput.-Assist

. Interv and . Int, Conf. Med. Image Comput. Comput.-Assist. Interv, vol.16, pp.411-418, 2013.

H. Su, Robust Cell Detection and Segmentation in Histopathological Images Using Sparse Reconstruction and Stacked Denoising Autoencoders, Med. Image Comput. Comput.-Assist. Interv. MICCAI Int. Conf. Med. Image Comput. Comput.-Assist. Interv, vol.9351, pp.383-390, 2015.

K. Sirinukunwattana, Locality Sensitive Deep Learning for Detection and Classification of Nuclei in Routine Colon Cancer Histology Images, IEEE Trans. Med. Imaging, vol.35, pp.1196-1206, 2016.

T. Pedersen and H. Peters, Proposal for a classification of oocytes and follicles in the mouse ovary, J. Reprod. Fertil, vol.17, pp.555-557, 1968.

T. A. Welch, A Technique for High-Performance Data Compression, Computer, vol.17, pp.8-19, 1984.

I. Goodfellow, Y. Bengio, and A. Courville, Deep Learning (Adaptive Computation and Machine Learning series), 2016.

S. Hochreiter, Y. Bengio, P. Frasconi, and J. Schmidhuber, Gradient Flow in Recurrent Nets: the Difficulty of Learning Long-Term Dependencies, 2001.

K. Simonyan and A. Zisserman, Very Deep Convolutional Networks for Large-Scale Image Recognition, 2014.

A. Krizhevsky, I. Sutskever, and G. E. Hinton, ImageNet Classification with Deep Convolutional Neural Networks, Advances in Neural Information Processing Systems, vol.25, pp.1097-1105, 2012.

R. Girshick, J. Donahue, T. Darrell, and J. Malik, Rich Feature Hierarchies for Accurate Object Detection and Semantic Segmentation, Proceedings of the 2014 IEEE Conference on Computer Vision and Pattern Recognition, pp.580-587, 2014.

S. Ren, K. He, R. Girshick, J. Sun, and . Faster-r-cnn, Towards Real-Time Object Detection with Region Proposal Networks. ArXiv150601497 Cs, 2015.

M. D. Zeiler, ADADELTA: An Adaptive Learning Rate Method, 2012.

N. Srivastava, G. Hinton, A. Krizhevsky, I. Sutskever, and R. Salakhutdinov, Dropout: A Simple Way to Prevent Neural Networks from Overfitting, J. Mach. Learn. Res, vol.15, pp.1929-1958, 2014.

F. Chollet and . Keras, , 2015.

M. Abadi, Large-Scale Machine Learning on Heterogeneous Distributed Systems. ArXiv160304467 Cs, 2016.

G. Bradski, The OpenCV Library. Dr. Dobb's Available at, p.16, 2018.

E. Evans, Domain-Driven Design: Tackling Complexity in the Heart of Software, 2013.

S. Suzuki and K. Be, Topological structural analysis of digitized binary images by border following, Comput. Vis. Graph. Image Process, vol.30, pp.32-46, 1985.

L. Perez and J. Wang, The Effectiveness of Data Augmentation in Image Classification using Deep Learning, 2017.

P. F. Felzenszwalb, R. B. Girshick, D. Mcallester, and D. Ramanan, Object Detection with Discriminatively Trained Part-Based Models, IEEE Trans. Pattern Anal. Mach. Intell, vol.32, pp.1627-1645, 2010.

N. Dalal and B. Triggs, Histograms of oriented gradients for human detection, IEEE Computer Society Conference on Computer Vision and Pattern Recognition (CVPR'05), vol.1, pp.886-893, 2005.
URL : https://hal.archives-ouvertes.fr/inria-00548512

H. Wang, Mitosis detection in breast cancer pathology images by combining handcrafted and convolutional neural network features, J. Med. Imaging Bellingham Wash, vol.1, p.34003, 2014.

T. J. Bucci, B. Bolon, A. R. Warbritton, J. J. Chen, and J. J. Heindel, Influence of sampling on the reproducibility of ovarian follicle counts in mouse toxicity studies, Reprod. Toxicol. Elmsford N, vol.11, pp.689-696, 1997.

P. B. Miller, J. S. Charleston, D. E. Battaglia, N. A. Klein, and M. R. Soules, An accurate, simple method for unbiased determination of primordial follicle number in the primate ovary, Biol

. Reprod, , vol.56, pp.909-915, 1997.

J. S. Charleston, Estimating human ovarian non-growing follicle number: the application of modern stereology techniques to an old problem, Hum. Reprod. Oxf. Engl, vol.22, pp.2103-2110, 2007.

L. Guzy and I. Demeestere, Assessment of ovarian reserve and fertility preservation strategies in children treated for cancer, Minerva Ginecol, 2016.

J. Assi, Psychosocial benefits of fertility preservation for young cancer patients, J. Assist. Reprod. Genet, 2018.

J. Donnez and M. Dolmans, Fertility Preservation in Women. N. Engl. J. Med, vol.377, pp.1657-1665, 2017.

, REFERENCES BIBLIOGRAPHIQUES

G. P. Adams, J. Singh, and A. R. Baerwald, Large animal models for the study of ovarian follicular dynamics in women, Theriogenology, vol.78, pp.1733-1748, 2012.

D. Adhikari and K. Liu, Molecular mechanisms underlying the activation of mammalian primordial follicles, Endocr Rev, vol.30, pp.438-464, 2009.

A. Akdemir, B. Zeybek, L. Akman, A. M. Ergenoglu, A. O. Yeniel et al., Granulocyte-colony stimulating factor decreases the extent of ovarian damage caused by cisplatin in an experimental rat model, J Gynecol Oncol, vol.25, pp.328-333, 2014.

A. Gougeon, The early stages of follicular growth, Biol Pathol Oocyte Role Infertil Reprod Med, p.cambridge, 2013.

M. S. Albamonte, M. A. Willis, M. I. Albamonte, F. Jensen, M. B. Espinosa et al., The developing human ovary: immunohistochemical analysis of germ-cell-specific VASA protein

, /BAX expression balance and apoptosis, Hum Reprod Oxf Engl, vol.23, pp.1895-1901, 2008.

A. Mercadal, B. Imbert, R. Demeestere, I. Gervy, C. et al., AMH mutations with reduced in vitro bioactivity are related to premature ovarian insufficiency, Hum Reprod Oxf Engl, vol.30, pp.1196-1202, 2015.

C. Anders, P. K. Marcom, B. Peterson, L. Gu, S. Unruhe et al., A pilot study of predictive markers of chemotherapy-related amenorrhea among premenopausal women with early stage breast cancer, Cancer Invest, vol.26, pp.286-295, 2008.

R. A. Anderson, Can Ovarian Reserve Assessment Predict Fertility following Breast Cancer Treatment?, Womens Health, vol.7, pp.623-625, 2011.

R. A. Anderson and D. A. Cameron, Pretreatment serum anti-müllerian hormone predicts long-term ovarian function and bone mass after chemotherapy for early breast cancer, J Clin Endocrinol Metab, vol.96, pp.1336-1343, 2011.

J. Assi, J. Santos, T. Bonetti, P. C. Serafini, E. Motta et al., Psychosocial benefits of fertility preservation for young cancer patients, J Assist Reprod Genet, 2018.

A. A. Azim, M. Costantini-ferrando, and K. Oktay, Safety of fertility preservation by ovarian stimulation with letrozole and gonadotropins in patients with breast cancer: a prospective controlled study, J Clin Oncol Off J Am Soc Clin Oncol, vol.26, pp.2630-2635, 2008.

H. A. Azim, N. Kroman, M. Paesmans, S. Gelber, N. Rotmensz et al., Prognostic impact of pregnancy after breast cancer according to estrogen receptor status: a multicenter retrospective study, J Clin Oncol Off J Am Soc Clin Oncol, vol.31, pp.73-79, 2013.

H. A. Azim, F. A. Peccatori, . Azambuja-e-de, and M. J. Piccart, Motherhood after breast cancer: searching for la dolce vita, Expert Rev Anticancer Ther, vol.11, pp.287-298, 2011.

W. M. Baarends, J. T. Uilenbroek, P. Kramer, J. W. Hoogerbrugge, A. P. Leeuwen-ec-van,-themmen et al., Anti-müllerian hormone and anti-müllerian hormone type II receptor messenger ribonucleic acid expression in rat ovaries during postnatal development, the estrous cycle, and gonadotropin-induced follicle growth, Endocrinology, vol.136, pp.4951-4962, 1995.

A. R. Baerwald, G. P. Adams, and R. A. Pierson, A new model for ovarian follicular development during the human menstrual cycle, Fertil Steril, vol.80, pp.116-122, 2003.

A. Baillet and B. Mandon-pepin, Mammalian ovary differentiation -a focus on female meiosis, Mol Cell Endocrinol, vol.356, pp.13-23, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01000904

T. G. Baker, . Study, . Germ, . In, and . Ovaries,

, Proc R Soc Lond B Biol Sci, vol.158, pp.417-433, 1963.

J. N. Bakkum-gamez, G. Aletti, K. A. Lewis, G. L. Keeney, B. M. Thomas et al., Müllerian inhibiting substance type II receptor (MISIIR): a novel, tissue-specific target expressed by gynecologic cancers, Gynecol Oncol, vol.108, pp.141-148, 2008.

H. Bar-joseph, I. Ben-aharon, M. Tzabari, G. Tsarfaty, S. M. Stemmer et al., In vivo bioimaging as a novel strategy to detect doxorubicin-induced damage to gonadal blood vessels, PloS One, vol.6, p.23492, 2011.

S. E. Barton, S. A. Missmer, K. F. Berry, and E. S. Ginsburg, Female cancer survivors are low responders and have reduced success compared with other patients undergoing assisted reproductive technologies, Fertil Steril, vol.97, pp.381-386, 2012.

M. W. Beckmann, R. Dittrich, S. Findeklee, and L. Lotz, Surgical Aspects of Ovarian Tissue Removal and Ovarian Tissue Transplantation for Fertility Preservation, Geburtshilfe Frauenheilkd, vol.76, pp.1057-1064, 2016.

G. Bedoschi, P. A. Navarro, and K. Oktay, Chemotherapy-induced damage to ovary: mechanisms and clinical impact, Future Oncol Lond Engl, vol.12, pp.2333-2344, 2016.

R. R. Behringer, M. J. Finegold, and R. L. Cate, Müllerian-inhibiting substance function during mammalian sexual development, Cell, vol.79, pp.415-425, 1994.

I. Ben-aharon, I. Meizner, T. Granot, S. Uri, N. Hasky et al.,

, Chemotherapy-induced ovarian failure as a prototype for acute vascular toxicity, The Oncologist, vol.17, pp.1386-1393, 2012.

E. R. Bertone-johnson, J. E. Manson, A. C. Purdue-smithe, A. Z. Steiner, A. H. Eliassen et al., Anti-Müllerian hormone levels and incidence of early natural menopause in a prospective study, Hum Reprod Oxf Engl, 2018.

J. Bézard, B. Vigier, D. Tran, P. Mauléon, and N. Josso, Immunocytochemical study of anti-Müllerian hormone in sheep ovarian follicles during fetal and post-natal development, J Reprod Fertil, vol.80, pp.509-516, 1987.

L. J. Blakely, A. U. Buzdar, J. A. Lozada, S. A. Shullaih, E. Hoy et al., Effects of pregnancy after treatment for breast carcinoma on survival and risk of recurrence, Cancer, vol.100, pp.465-469, 2004.

J. Bouilly, A. Bachelot, I. Broutin, P. Touraine, and N. Binart, Novel NOBOX loss-of-function mutations account for 6.2% of cases in a large primary ovarian insufficiency cohort, Hum Mutat, vol.32, pp.1108-1113, 2011.

, Références bibliographiques, p.127

J. Bouilly, F. Roucher-boulez, A. Gompel, H. Bry-gauillard, K. Azibi et al., New NOBOX mutations identified in a large cohort of women with primary ovarian insufficiency decrease KIT-L expression, J Clin Endocrinol Metab, vol.100, pp.994-1001, 2015.

J. Bouilly, R. A. Veitia, and N. Binart, NOBOX is a key FOXL2 partner involved in ovarian folliculogenesis
URL : https://hal.archives-ouvertes.fr/hal-01132566

, J Mol Cell Biol, vol.6, pp.175-177, 2014.

M. Braem, M. Voorhuis, Y. T. Schouw, . Van-der, P. Peeters et al., Onland-Moret NC. Interactions between genetic variants in AMH and AMHR2 may modify age at natural menopause, PloS One, vol.8, p.59819, 2013.

F. J. Broekmans, M. R. Soules, and B. C. Fauser, Ovarian aging: mechanisms and clinical consequences

, Endocr Rev, vol.30, pp.465-493, 2009.

S. L. Broer, F. Broekmans, J. Laven, and B. Fauser, Anti-Müllerian hormone: ovarian reserve testing and its potential clinical implications, Hum Reprod Update, vol.20, pp.688-701, 2014.

S. L. Broer, K. A. Disseldorp-j-van,-broeze, M. Dolleman, B. C. Opmeer, P. Bossuyt et al., Added value of ovarian reserve testing on patient characteristics in the prediction of ovarian response and ongoing pregnancy: an individual patient data approach, Hum Reprod Update, vol.19, pp.26-36, 2013.

H. Bry-gauillard, F. Larrat-ledoux, J. Levaillant, N. Massin, L. Maione et al., Anti-Mullerian hormone and ovarian morphology in women with isolated hypogonadotropic hypogonadism/Kallmann syndrome. Effects of recombinant human FSH, J Clin Endocrinol Metab, 2017.

M. J. Carabatsos, J. Elvin, M. M. Matzuk, and D. F. Albertini, Characterization of oocyte and follicle development in growth differentiation factor-9-deficient mice, Dev Biol, vol.204, p.373, 1998.

I. B. Carlsson, J. E. Scott, J. A. Visser, O. Ritvos, A. Themmen et al., Anti-Müllerian hormone inhibits initiation of growth of human primordial ovarian follicles in vitro, Hum Reprod Oxf Engl, vol.21, pp.2223-2227, 2006.

D. H. Castrillon, L. Miao, R. Kollipara, J. W. Horner, and R. A. Depinho, Suppression of ovarian follicle activation in mice by the transcription factor Foxo3a, Science, vol.301, pp.215-218, 2003.

E. M. Chang, E. Lim, S. Yoon, K. Jeong, S. Bae 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, vol.10, p.144245, 2015.

H. Chang, J. Qiao, and P. Leung, Oocyte-somatic cell interactions in the human ovary-novel role of bone morphogenetic proteins and growth differentiation factors, Hum Reprod Update, vol.23, pp.1-18, 2016.

A. Chassot, I. Gillot, and M. Chaboissier, R-spondin1, WNT4, and the CTNNB1 signaling pathway: strict control over ovarian differentiation, Reprod Camb Engl, vol.148, pp.97-110, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01421167

C. Vizcaíno, M. A. Corchado, A. R. Cuadri, M. Comadran, M. G. Brassesco et al., The effects of letrozole on ovarian stimulation for fertility preservation in cancer-affected women

, Reprod Biomed Online, vol.24, pp.606-610, 2012.

X. Chen, H. Xia, H. Guan, B. Li, and W. Zhang, Follicle Loss and Apoptosis in CyclophosphamideTreated Mice: What's the Matter?, Int J Mol Sci, vol.17, 2016.

Y. Chen, W. N. Jefferson, R. R. Newbold, E. Padilla-banks, and M. E. Pepling, Estradiol, progesterone, and genistein inhibit oocyte nest breakdown and primordial follicle assembly in the neonatal mouse ovary in vitro and in vivo, Endocrinology, vol.148, pp.3580-3590, 2007.

Y. Choi, D. J. Ballow, Y. Xin, and A. Rajkovic, Lim homeobox gene, lhx8, is essential for mouse oocyte differentiation and survival, Biol Reprod, vol.79, pp.442-449, 2008.

I. Cimino, F. Casoni, X. Liu, A. Messina, J. Parkash et al., Novel role for anti-Müllerian hormone in the regulation of GnRH neuron excitability and hormone secretion, Nat Commun, vol.7, p.10055, 2016.

, Références bibliographiques, p.129

. Clemente-n-di, S. Ghaffari, R. B. Pepinsky, C. Pieau, N. Josso et al., A quantitative and interspecific test for biological activity of anti-müllerian hormone: the fetal ovary aromatase assay, Dev Camb Engl, vol.114, pp.721-727, 1992.

A. Cobo, J. A. García-velasco, A. Coello, J. Domingo, A. Pellicer et al., Oocyte vitrification as an efficient option for elective fertility preservation, Fertil Steril, vol.105, pp.755-764, 2016.

M. Conti, Signaling networks in somatic cells and oocytes activated during ovulation, Ann Endocrinol, vol.71, pp.189-190, 2010.

M. Conti, M. Hsieh, J. Park, and Y. Su, Role of the epidermal growth factor network in ovarian follicles

, Mol Endocrinol Baltim Md, vol.20, pp.715-723, 2006.

H. Creux, P. Monnier, W. Son, and W. Buckett, Thirteen years' experience in fertility preservation for cancer patients after in vitro fertilization and in vitro maturation treatments, J Assist Reprod Genet, 2018.

L. Crisponi, M. Deiana, A. Loi, F. Chiappe, M. Uda et al., The putative forkhead transcription factor FOXL2 is mutated in blepharophimosis/ptosis/epicanthus inversus syndrome, Nat Genet, vol.27, pp.159-166, 2001.

. Cuevas-m-de, M. A. Lilly, and A. C. Spradling, Germline cyst formation in Drosophila, Annu Rev Genet, vol.31, pp.405-428, 1997.

R. A. Cushman, C. M. Wahl, and J. E. Fortune, Bovine ovarian cortical pieces grafted to chick embryonic membranes: a model for studies on the activation of primordial follicles, Hum Reprod Oxf Engl, vol.17, pp.48-54, 2002.

Â. M. D'avila, V. Biolchi, E. Capp, and E. Corleta-h-von, Age, anti-müllerian hormone, antral follicles count to predict amenorrhea or oligomenorrhea after chemotherapy with cyclophosphamide, J Ovarian Res, vol.8, p.82, 2015.

D. Vos, M. Devroey, P. Fauser, and B. , Primary ovarian insufficiency, Lancet Lond Engl, vol.376, pp.911-921, 2010.

I. Demeestere, P. Brice, F. A. Peccatori, A. Kentos, J. Dupuis et al., No Evidence for the Benefit of Gonadotropin-Releasing Hormone Agonist in Preserving Ovarian Function and Fertility in Lymphoma Survivors Treated With Chemotherapy: Final Long-Term

, J Clin Oncol Off J Am Soc Clin Oncol, vol.34, pp.2568-2574, 2016.

I. Demeestere, P. Simon, L. Dedeken, F. Moffa, S. Tsépélidis et al., Live birth after autograft of ovarian tissue cryopreserved during childhood, Hum Reprod Oxf Engl, vol.30, pp.2107-2109, 2015.

L. Detti, R. A. Uhlmann, J. Zhang, M. P. Diamond, G. M. Saed et al., Goserelin fosters bone elongation but does not prevent ovarian damage in cyclophosphamidetreated prepubertal mice, Fertil Steril, vol.101, pp.1157-1164, 2014.

D. Dewailly, C. Y. Andersen, A. Balen, F. Broekmans, N. Dilaver et al., The physiology and clinical utility of anti-Mullerian hormone in women, Hum Reprod Update, vol.20, pp.370-385, 2014.

D. Dewailly, H. Gronier, E. Poncelet, G. Robin, M. Leroy et al., Diagnosis of polycystic ovary syndrome (PCOS): revisiting the threshold values of follicle count on ultrasound and of the serum AMH level for the definition of polycystic ovaries

, Hum Reprod Oxf Engl, vol.26, pp.3123-3129, 2011.

D. Dewailly, G. Robin, M. Peigne, C. Decanter, P. Pigny et al., Interactions between androgens, FSH, anti-Müllerian hormone and estradiol during folliculogenesis in the human normal and polycystic ovary, Hum Reprod Update, vol.22, pp.709-724, 2016.

A. Dezellus, P. Barriere, M. Campone, C. Lemanski, L. Vanlemmens et al., Prospective evaluation of serum anti-Müllerian hormone dynamics in 250 women of reproductive age treated with chemotherapy for breast cancer

, Eur J Cancer Oxf Engl, vol.79, pp.72-80, 1990.

P. K. Donahoe, T. Clarke, J. Teixeira, S. Maheswaran, and D. T. Maclaughlin, Enhanced purification and production of Müllerian inhibiting substance for therapeutic applications, Mol Cell Endocrinol, vol.211, pp.37-42, 2003.

J. Donnez and M. Dolmans, Ovarian cortex transplantation: 60 reported live births brings the success and worldwide expansion of the technique towards routine clinical practice, J Assist Reprod Genet, vol.32, pp.1167-1170, 2015.

J. Donnez and M. Dolmans, Fertility Preservation in Women, N Engl J Med, vol.377, pp.1657-1665, 2017.

J. Donnez, M. M. Dolmans, D. Demylle, P. Jadoul, C. Pirard et al., Livebirth after orthotopic transplantation of cryopreserved ovarian tissue, Lancet Lond Engl, vol.364, pp.1405-1410, 2004.

J. Donnez, B. Martinez-madrid, P. Jadoul, A. Van-langendonckt, D. Demylle et al., Ovarian tissue cryopreservation and transplantation: a review, Hum Reprod Update, vol.12, pp.519-535, 2006.

M. A. Driancourt, K. Reynaud, R. Cortvrindt, and J. Smitz, Roles of KIT and KIT LIGAND in ovarian function, Rev Reprod, vol.5, pp.143-152, 2000.

C. E. Dunlop and R. A. Anderson, Uses of anti-Müllerian hormone (AMH) measurement before and after cancer treatment in women, Maturitas, vol.80, pp.245-250, 2015.

A. L. Durlinger, M. J. Gruijters, P. Kramer, B. Karels, T. R. Kumar et al., Anti-Müllerian hormone attenuates the effects of FSH on follicle development in the mouse ovary, Endocrinology, vol.142, pp.4891-4899, 2001.

A. L. Durlinger, P. Kramer, B. Karels, F. H. Jong, . De et al., Control of primordial follicle recruitment by anti-Müllerian hormone in the mouse ovary, Endocrinology, vol.140, pp.5789-5796, 1999.

A. Durlinger, M. Gruijters, P. Kramer, B. Karels, H. A. Ingraham et al., Anti-Müllerian hormone inhibits initiation of primordial follicle growth in the mouse ovary, Endocrinology, vol.143, pp.1076-1084, 2002.

, Références bibliographiques, p.132

A. Durlinger, J. A. Visser, and A. Themmen, Regulation of ovarian function: the role of anti-Müllerian hormone, Reprod Camb Engl, vol.124, pp.601-609, 2002.

M. A. Edson, A. K. Nagaraja, and M. M. Matzuk, The mammalian ovary from genesis to revelation, Endocr Rev, vol.30, pp.624-712, 2009.

M. Elzaiat, A. Todeschini, S. Caburet, and R. A. Veitia, The genetic make-up of ovarian development and function: the focus on the transcription factor FOXL2, Clin Genet, vol.91, pp.173-182, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01498857

, Ethics Committee of American Society for Reproductive Medicine. Fertility preservation and reproduction in patients facing gonadotoxic therapies: a committee opinion, Fertil Steril, vol.100, pp.1224-1231, 2013.

M. M. Ezzati, M. D. Baker, H. D. Saatcioglu, G. M. Aloisio, C. G. Pena et al., Regulation of FOXO3 subcellular localization by Kit ligand in the neonatal mouse ovary, J Assist Reprod Genet, vol.32, pp.1741-1747, 2015.

G. Familiari, A. Caggiati, S. A. Nottola, M. Ermini, D. Benedetto et al., Ultrastructure of human ovarian primordial follicles after combination chemotherapy for Hodgkin's disease, Hum Reprod Oxf Engl, vol.8, pp.2080-2087, 1993.

R. Fanchin, L. M. Schonäuer, C. Righini, J. Guibourdenche, R. Frydman et al., Serum anti-Müllerian hormone is more strongly related to ovarian follicular status than serum inhibin B, estradiol, FSH and LH on day 3, Hum Reprod Oxf Engl, vol.18, pp.323-327, 2003.

C. Feng, J. Bowles, and P. Koopman, Control of mammalian germ cell entry into meiosis, Mol Cell Endocrinol, vol.382, pp.488-497, 2014.

B. Fisch and R. Abir, Female fertility preservation: past, present and future, Reprod Camb Engl, 2018.

S. M. Galloway, K. P. Mcnatty, L. M. Cambridge, M. P. Laitinen, J. L. Juengel et al., Mutations in an oocyte-derived growth factor gene (BMP15) cause increased ovulation rate and infertility in a dosage-sensitive manner, Nat Genet, vol.25, pp.279-283, 2000.

Z. Gavish, G. Peer, H. Roness, Y. Cohen, and D. Meirow, Follicle activation and "burn-out" contribute to post-transplantation follicle loss in ovarian tissue grafts: the effect of graft thickness, Hum Reprod Oxf Engl, vol.30, p.1003, 2015.

Z. Gavish, I. Spector, G. Peer, S. Schlatt, J. Wistuba et al., Follicle activation is a significant and immediate cause of follicle loss after ovarian tissue transplantation, J Assist Reprod Genet, vol.35, pp.61-69, 2018.

T. R. Gawriluk, A. N. Hale, J. A. Flaws, C. P. Dillon, D. R. Green et al., Autophagy is a cell survival program for female germ cells in the murine ovary, Reprod Camb Engl, vol.141, pp.759-765, 2011.

B. Gerstl, E. Sullivan, A. Ives, C. Saunders, H. Wand et al., Pregnancy Outcomes After a Breast Cancer Diagnosis: A Systematic Review and Meta-analysis, Clin Breast Cancer, vol.18, pp.79-88, 2018.

F. Ghafari, C. G. Gutierrez, and G. M. Hartshorne, Apoptosis in mouse fetal and neonatal oocytes during meiotic prophase one, BMC Dev Biol, vol.7, p.87, 2007.

I. Gigli, R. A. Cushman, C. M. Wahl, and J. E. Fortune, Evidence for a role for anti-Mullerian hormone in the suppression of follicle activation in mouse ovaries and bovine ovarian cortex grafted beneath the chick chorioallantoic membrane, Mol Reprod Dev, vol.71, pp.480-488, 2005.

K. N. Goldman, D. Chenette, R. Arju, F. E. Duncan, D. L. Keefe et al., mTORC1/2 inhibition preserves ovarian function and fertility during genotoxic chemotherapy, Proc Natl Acad Sci U S A, vol.114, pp.3186-3191, 2017.

R. H. Goldman, C. Racowsky, L. V. Farland, S. Munné, L. Ribustello et al., Predicting the likelihood of live birth for elective oocyte cryopreservation: a counseling tool for physicians and patients, Hum Reprod Oxf Engl, vol.32, pp.853-859, 2017.

S. Gonfloni, D. Tella, L. Caldarola, S. Cannata, S. M. Klinger et al., Inhibition of the c-Abl-TAp63 pathway protects mouse oocytes from chemotherapy-induced death, Nat Med, vol.15, pp.1179-1185, 2009.

A. Gougeon, Regulation of ovarian follicular development in primates: facts and hypotheses

, Endocr Rev, vol.17, pp.121-155, 1996.

A. Gougeon, Human ovarian follicular development: from activation of resting follicles to preovulatory maturation, Ann Endocrinol, vol.71, pp.132-143, 2010.
URL : https://hal.archives-ouvertes.fr/inserm-00504499

A. Gremeau, N. Andreadis, M. Fatum, J. Craig, K. Turner et al., In vitro maturation or in vitro fertilization for women with polycystic ovaries? A case-control study of 194 treatment cycles, Fertil Steril, vol.98, pp.355-360, 2012.

K. J. Grive and R. N. Freiman, The developmental origins of the mammalian ovarian reserve, Dev Camb Engl, vol.142, pp.2554-2563, 2015.

M. L. Grøndahl, M. E. Nielsen, M. B. Dal-canto, R. Fadini, I. A. Rasmussen et al., Anti-Müllerian hormone remains highly expressed in human cumulus cells during the final stages of folliculogenesis, Reprod Biomed Online, vol.22, pp.389-398, 2011.

M. P. Grossman, S. T. Nakajima, M. E. Fallat, and Y. Siow, Müllerian-inhibiting substance inhibits cytochrome P450 aromatase activity in human granulosa lutein cell culture, Fertil Steril, vol.89, pp.1364-1370, 2008.

M. Grynberg, E. Hachem, H. Bantel, A. De, J. Benard et al., In vitro maturation of oocytes: uncommon indications, Fertil Steril, vol.99, pp.1182-1188, 2013.

M. Grynberg, A. Pierre, R. Rey, A. Leclerc, N. Arouche et al., Differential regulation of ovarian anti-müllerian hormone (AMH) by estradiol through ?-and ?-estrogen receptors, J Clin Endocrinol Metab, vol.97, pp.1649-1657, 2012.

X. Guéripel, V. Brun, and A. Gougeon, Oocyte bone morphogenetic protein 15, but not growth differentiation factor 9, is increased during gonadotropin-induced follicular development in the immature mouse and is associated with cumulus oophorus expansion, Biol Reprod, vol.75, pp.836-843, 2006.

C. J. Guigon and S. Magre, Contribution of germ cells to the differentiation and maturation of the ovary: insights from models of germ cell depletion, Biol Reprod, vol.74, pp.450-458, 2006.

C. P. Hagen, K. Sørensen, R. A. Anderson, and A. Juul, Serum levels of antimüllerian hormone in early maturing girls before, during, and after suppression with GnRH agonist, Fertil Steril, vol.98, pp.1326-1330, 2012.

A. Hamy, R. Porcher, C. Cuvier, S. Giacchetti, M. Schlageter et al., Ovarian reserve in breast cancer: assessment with antiMüllerian hormone, Reprod Biomed Online, vol.29, pp.573-580, 2014.

A. Hamy, R. Porcher, S. Eskenazi, C. Cuvier, S. Giacchetti et al., Anti-Müllerian hormone in breast cancer patients treated with chemotherapy: a retrospective evaluation of subsequent pregnancies, Reprod Biomed Online, vol.32, pp.299-307, 2016.

K. Hancke, O. Strauch, C. Kissel, H. Göbel, W. Schäfer et al., Sphingosine 1-phosphate protects ovaries from chemotherapy-induced damage in vivo, Fertil Steril, vol.87, pp.172-177, 2007.

K. R. Hansen, G. M. Hodnett, N. Knowlton, and L. B. Craig, Correlation of ovarian reserve tests with histologically determined primordial follicle number, Fertil Steril, vol.95, pp.170-175, 2011.

N. Hasky, S. Uri-belapolsky, K. Goldberg, I. Miller, H. Grossman et al., Gonadotrophin-releasing hormone agonists for fertility preservation: unraveling the enigma?, Hum Reprod Oxf Engl, vol.30, pp.1089-1101, 2015.

A. Hassanpour, S. Yousefian, M. Askaripour, F. Sharififar, and M. Ezzatabadipour, Ovarian protection in cyclophosphamide-treated mice by fennel, Toxicol Rep, vol.4, pp.160-164, 2017.

E. Hayes, V. Kushnir, X. Ma, A. Biswas, H. Prizant et al., Intra-cellular mechanism of AntiMüllerian hormone (AMH) in regulation of follicular development, Mol Cell Endocrinol, vol.433, pp.56-65, 2016.

S. G. Hillier, L. E. Reichert, V. Hall, and E. V. , Control of preovulatory follicular estrogen biosynthesis in the human ovary, J Clin Endocrinol Metab, vol.52, pp.847-856, 1981.

R. Himelstein-braw, H. Peters, and M. Faber, Morphological study of the ovaries of leukaemic children

, Br J Cancer, vol.38, pp.82-87, 1978.

S. Hirobe, W. W. He, M. L. Gustafson, D. T. Maclaughlin, and P. K. Donahoe, Müllerian inhibiting substance gene expression in the cycling rat ovary correlates with recruited or graafian follicle selection, Biol Reprod, vol.50, pp.1238-1243, 1994.

S. Hirobe, W. W. He, M. M. Lee, and P. K. Donahoe, Mullerian inhibiting substance messenger ribonucleic acid expression in granulosa and Sertoli cells coincides with their mitotic activity, Endocrinology, vol.131, pp.854-862, 1992.

F. Horicks, G. Van-den-steen, C. Gervy, H. J. Clarke, and I. Demeestere, Both in vivo FSH depletion and follicular exposure to Gonadotrophin-releasing hormone analogues in vitro are not effective to prevent follicular depletion during chemotherapy in mice, Mol Hum Reprod, 2018.

R. Hu, F. Wang, L. Yu, Y. Luo, X. Wu et al., Antimüllerian hormone regulates stem cell factor expression in human granulosa cells, Fertil Steril, vol.102, pp.1742-1750, 2014.

K. Iino, I. Kaori, A. Tarakida, A. T. Abe, K. Kazuhiro et al., Role of antimüllerian hormone as a biomarker of the menopausal transition

N. Menopause, , vol.20, pp.218-222, 2013.

A. Iwase, T. Nakamura, S. Osuka, S. Takikawa, M. Goto et al., Anti-Müllerian hormone as a marker of ovarian reserve: What have we learned, and what should we know, Reprod Med Biol, vol.15, pp.127-136, 2016.

A. Iwase, S. Osuka, M. Goto, T. Murase, T. Nakamura et al., Clinical application of serum anti-Müllerian hormone as an ovarian reserve marker: A review of recent studies

, J Obstet Gynaecol Res, 2018.

K. Jagarlamudi, L. Liu, D. Adhikari, P. Reddy, A. Idahl et al., Oocyte-specific deletion of Pten in mice reveals a stage-specific function of PTEN/PI3K signaling in oocytes in controlling follicular activation, PloS One, vol.4, p.6186, 2009.

H. Jang, O. Lee, Y. Lee, H. Yoon, E. M. Chang et al., Melatonin prevents cisplatin-induced primordial follicle loss via suppression of PTEN/AKT/FOXO3a pathway activation in the mouse ovary, J Pineal Res, vol.60, pp.336-347, 2016.

H. Jang, Y. Na, K. Hong, S. Lee, S. Moon et al., Synergistic effect of melatonin and ghrelin in preventing cisplatin-induced ovarian damage via regulation of FOXO3a phosphorylation and binding to the p27Kip1promoter in primordial follicles, J Pineal Res, vol.63, 2017.

J. V. Jeppesen, R. A. Anderson, T. W. Kelsey, S. L. Christiansen, S. G. Kristensen et al., Which follicles make the most anti-Mullerian hormone in humans? Evidence for an abrupt decline in AMH production at the time of follicle selection, Mol Hum Reprod, vol.19, pp.519-527, 2013.

G. B. John, T. D. Gallardo, L. J. Shirley, and D. H. Castrillon, Foxo3 is a PI3K-dependent molecular switch controlling the initiation of oocyte growth, Dev Biol, vol.321, pp.197-204, 2008.

J. Johnson, J. Bagley, M. Skaznik-wikiel, H. Lee, G. B. Adams et al., Oocyte generation in adult mammalian ovaries by putative germ cells in bone marrow and peripheral blood, Cell, vol.122, pp.303-315, 2005.

N. Josso, Professor Alfred Jost: the builder of modern sex differentiation, Sex Dev Genet Mol Biol Evol Endocrinol Embryol Pathol Sex Determ Differ, vol.2, pp.55-63, 2008.

N. Josso, N. Di-clemente, and L. Gouédard, Anti-Müllerian hormone and its receptors, Mol Cell Endocrinol, vol.179, pp.25-32, 2001.

L. Kalich-philosoph, H. Roness, A. Carmely, M. Fishel-bartal, H. Ligumsky et al., Cyclophosphamide triggers follicle activation and "burnout"; AS101 prevents follicle loss and preserves fertility, Sci Transl Med, vol.5, pp.185-62, 2013.

M. Kano, A. E. Sosulski, L. Zhang, H. D. Saatcioglu, D. Wang et al., AMH/MIS as a contraceptive that protects the ovarian reserve during chemotherapy, Proc Natl Acad Sci, vol.114, pp.1688-1697, 2017.

K. Kawamura, Y. Cheng, N. Suzuki, M. Deguchi, Y. Sato et al., Hippo signaling disruption and Akt stimulation of ovarian follicles for infertility treatment, Proc Natl Acad Sci U S A, vol.110, pp.17474-17479, 2013.

H. Kaya, R. Desdicioglu, M. Sezik, E. Ulukaya, O. Ozkaya et al., Does sphingosine-1-phosphate have a protective effect on cyclophosphamide-and irradiation-induced ovarian damage in the rat model?, Fertil Steril, vol.89, pp.732-735, 2008.

T. W. Kelsey, P. Wright, S. M. Nelson, R. A. Anderson, and W. Wallace, A validated model of serum antimüllerian hormone from conception to menopause, PloS One, vol.6, p.22024, 2011.

J. B. Kerr, K. J. Hutt, E. M. Michalak, M. Cook, C. J. Vandenberg et al., DNA damage-induced primordial follicle oocyte apoptosis and loss of fertility require TAp63-mediated induction of Puma and Noxa, Mol Cell, vol.48, pp.343-352, 2012.

N. Kersual, V. Garambois, T. Chardès, J. Pouget, I. Salhi et al., The human Müllerian inhibiting substance type II receptor as immunotherapy target for ovarian cancer. Validation using the mAb 12G4, mAbs, vol.6, pp.1314-1326, 2014.

M. E. Kevenaar, M. F. Meerasahib, P. Kramer, . Lang-born, . Bmn-van-de et al., Serum anti-mullerian hormone levels reflect the size of the primordial follicle pool in mice, Endocrinology, vol.147, pp.3228-3234, 2006.

P. Kezele and M. K. Skinner, Regulation of ovarian primordial follicle assembly and development by estrogen and progesterone: endocrine model of follicle assembly, Endocrinology, vol.144, pp.3329-3337, 2003.

, Références bibliographiques, p.139

J. Kim, V. Turan, and K. Oktay, Long-Term Safety of Letrozole and Gonadotropin Stimulation for Fertility Preservation in Women With Breast Cancer, J Clin Endocrinol Metab, vol.101, pp.1364-1371, 2016.

J. H. Kim, D. T. Maclaughlin, and P. K. Donahoe, Müllerian inhibiting substance/anti-Müllerian hormone: A novel treatment for gynecologic tumors, Obstet Gynecol Sci, vol.57, pp.343-357, 2014.

S. Kim, M. H. Cordeiro, V. A. Serna, K. Ebbert, L. M. Butler et al., Rescue of platinum-damaged oocytes from programmed cell death through inactivation of the p53 family signaling network, Cell Death Differ, vol.20, pp.987-997, 2013.

E. Kishk, M. Ali, and M. H. , Effect of a gonadotropin-releasing hormone analogue on cyclophosphamide-induced ovarian toxicity in adult mice, Arch Gynecol Obstet, vol.287, pp.1023-1029, 2013.

M. Kitajima, M. Dolmans, O. Donnez, H. Masuzaki, M. Soares et al., Enhanced follicular recruitment and atresia in cortex derived from ovaries with endometriomas, Fertil Steril, vol.101, pp.1031-1037, 2014.

P. G. Knight and C. Glister, TGF-beta superfamily members and ovarian follicle development, Reprod Camb Engl, vol.132, pp.191-206, 2006.

H. S. Kong, S. K. Kim, J. Lee, H. W. Youm, J. R. Lee et al., Effect of Exogenous Anti-Müllerian Hormone Treatment on Cryopreserved and Transplanted Mouse Ovaries, Reprod Sci Thousand Oaks Calif, vol.23, pp.51-60, 2016.

L. L. Kujjo, T. Laine, R. Pereira, W. Kagawa, H. Kurumizaka et al., Enhancing survival of mouse oocytes following chemotherapy or aging by targeting Bax and Rad51

, PloS One, vol.5, p.9204, 2010.

V. A. Kushnir, D. B. Seifer, D. H. Barad, A. Sen, and N. Gleicher, Potential therapeutic applications of human anti-Müllerian hormone (AMH) analogues in reproductive medicine, J Assist Reprod Genet, vol.34, pp.1105-1113, 2017.

L. Marca, A. Ferraretti, A. P. Palermo, R. Ubaldi, and F. M. , The use of ovarian reserve markers in IVF clinical practice: a national consensus, Gynecol Endocrinol Off J Int Soc Gynecol Endocrinol, vol.32, pp.1-5, 2016.

L. Marca, A. Sunkara, and S. K. , Individualization of controlled ovarian stimulation in IVF using ovarian reserve markers: from theory to practice, Hum Reprod Update, vol.20, pp.124-140, 2014.

L. Marca, A. Volpe, and A. , The Anti-Mullerian hormone and ovarian cancer, Hum Reprod Update, vol.13, pp.265-273, 2007.

M. Lambertini, D. Mastro, L. Pescio, M. C. Andersen, C. Y. Azim et al., Cancer and fertility preservation: international recommendations from an expert meeting, BMC Med, vol.14, p.1, 2016.

K. Lan, C. Sy, F. Huang, H. Lin, C. Lin et al., Analysis of androgen receptor and anti-Müllerian hormone pathways in human granulosa cells under luteinizing hormone treatment, Reprod Biol Endocrinol RBE, vol.11, p.11, 2013.

Y. Lande, B. Fisch, A. Tsur, J. Farhi, R. Prag-rosenberg et al., Short-term exposure of human ovarian follicles to cyclophosphamide metabolites seems to promote follicular activation in vitro, Reprod Biomed Online, vol.34, pp.104-114, 2017.

H. Lee, R. Grosse, R. Ranganath, and A. Y. Ng, Unsupervised learning of hierarchical representations with convolutional deep belief networks, Commun ACM, vol.54, pp.95-103, 2011.

F. Li, V. Turan, S. Lierman, C. Cuvelier, D. Sutter et al., Sphingosine-1-phosphate prevents chemotherapy-induced human primordial follicle death, Hum Reprod Oxf Engl, vol.29, pp.107-113, 2014.

H. Li, R. A. Anderson, W. Yeung, P. C. Ho, and E. Ng, Evaluation of serum antimullerian hormone and inhibin B concentrations in the differential diagnosis of secondary oligoamenorrhea

, Fertil Steril, vol.96, pp.774-779, 2011.

J. Li, K. Kawamura, Y. Cheng, S. Liu, C. Klein et al., Activation of dormant ovarian follicles to generate mature eggs, Proc Natl Acad Sci U S A, vol.107, pp.10280-10284, 2010.

L. Li, X. Zhou, X. Wang, J. Wang, W. Zhang et al., A dominant negative mutation at the ATP binding domain of AMHR2 is associated with a defective anti-Müllerian hormone signaling pathway, Mol Hum Reprod, vol.22, pp.669-678, 2016.

C. Liu, J. Peng, M. M. Matzuk, and H. Yao, Lineage specification of ovarian theca cells requires multicellular interactions via oocyte and granulosa cells, Nat Commun, vol.6, p.6934, 2015.

H. Liu, Y. Tao, P. Luo, C. Deng, Y. Gu et al., Preventive Effects of a Novel Polysaccharide from Sepia esculenta Ink on Ovarian Failure and Its Action Mechanisms in Cyclophosphamide-Treated Mice, J Agric Food Chem, vol.64, pp.5759-5766, 2016.

L. Schumacher, B. Grover, N. Mesen, T. Steiner, A. Mersereau et al., Modeling of live-birth rates and cost-effectiveness of oocyte cryopreservation for cancer patients prior to high-and lowrisk gonadotoxic chemotherapy, Hum Reprod Oxf Engl, vol.32, pp.2049-2055, 2017.

E. Maiani, D. Bartolomeo, C. Klinger, F. G. Cannata, S. M. Bernardini et al., Reply to: Cisplatin-induced primordial follicle oocyte killing and loss of fertility are not prevented by imatinib, Nat Med, vol.18, pp.1172-1174, 2012.

E. Maiani, M. Diederich, and S. Gonfloni, DNA damage response: the emerging role of c-Abl as a regulatory switch?, Biochem Pharmacol, vol.82, pp.1269-1276, 2011.

A. Margolskee and J. F. Selgrade, A lifelong model for the female reproductive cycle with an antimüllerian hormone treatment to delay menopause, J Theor Biol, vol.326, pp.21-35, 2013.

F. Martinez, International Society for Fertility Preservation-ESHRE-ASRM Expert Working Group. Update on fertility preservation from the Barcelona International Society for Fertility Preservation-ESHRE-ASRM 2015 expert meeting: indications, results and future perspectives, vol.108, pp.407-415, 2017.

A. Martinez-royo, J. J. Jurado, J. P. Smulders, J. I. Martí, J. L. Alabart et al., A deletion in the bone morphogenetic protein 15 gene causes sterility and increased prolificacy in Rasa Aragonesa sheep, Anim Genet, vol.39, pp.294-297, 2008.

P. T. Masiakos, D. T. Maclaughlin, S. Maheswaran, J. Teixeira, A. F. Fuller et al., Human ovarian cancer, cell lines, and primary ascites cells express the human Mullerian inhibiting substance (MIS) type II receptor, bind, and are responsive to MIS, Clin Cancer Res Off J Am Assoc Cancer Res, vol.5, pp.3488-3499, 1999.

J. Massagué and Y. G. Chen, Controlling TGF-beta signaling, Genes Dev, vol.14, pp.627-644, 2000.

M. M. Matzuk, K. H. Burns, M. M. Viveiros, and J. J. Eppig, Intercellular communication in the mammalian ovary: oocytes carry the conversation, Science, vol.296, pp.2178-2180, 2002.

E. A. Mcgee and R. S. Raj, Regulators of ovarian preantral follicle development, Semin Reprod Med, vol.33, pp.179-184, 2015.

M. Mclaughlin, H. L. Kinnell, R. A. Anderson, and E. E. Telfer, 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, vol.20, pp.736-744, 2014.

K. P. Mcnatty, J. L. Juengel, K. L. Reader, S. Lun, S. Myllymaa et al., Bone morphogenetic protein 15 and growth differentiation factor 9 co-operate to regulate granulosa cell function, Reprod Camb Engl, vol.129, pp.473-480, 2005.

G. Meduri, N. Charnaux, F. Spyratos, K. Hacene, H. Loosfelt et al., Luteinizing hormone receptor status and clinical, pathologic, and prognostic features in patients with breast carcinomas, Cancer, vol.97, pp.1810-1816, 2003.

D. Meirow, G. Assad, J. Dor, and J. Rabinovici, The GnRH antagonist cetrorelix reduces cyclophosphamide-induced ovarian follicular destruction in mice, Hum Reprod Oxf Engl, vol.19, pp.1294-1299, 2004.

, Références bibliographiques, p.143

D. Meirow, J. Dor, B. Kaufman, A. Shrim, J. Rabinovici et al., Cortical fibrosis and blood-vessels damage in human ovaries exposed to chemotherapy. Potential mechanisms of ovarian injury, Hum Reprod Oxf Engl, vol.22, pp.1626-1633, 2007.

D. Meirow, H. Lewis, D. Nugent, and M. Epstein, Subclinical depletion of primordial follicular reserve in mice treated with cyclophosphamide: clinical importance and proposed accurate investigative tool, Hum Reprod Oxf Engl, vol.14, pp.1903-1907, 1999.

D. Meirow, H. Roness, S. G. Kristensen, and C. Y. Andersen, Optimizing outcomes from ovarian tissue cryopreservation and transplantation; activation versus preservation, Hum Reprod Oxf Engl, vol.30, pp.2453-2456, 2015.

Z. Merhi, M. Irani, A. D. Doswell, and J. Ambroggio, Follicular Fluid Soluble Receptor for Advanced Glycation End-Products (sRAGE): A Potential Indicator of Ovarian Reserve, J Clin Endocrinol Metab, 2013.

N. Mizushima, T. Yoshimori, and Y. Ohsumi, The role of Atg proteins in autophagosome formation, Annu Rev Cell Dev Biol, vol.27, pp.107-132, 2011.

P. Monget and C. Bondy, Importance of the IGF system in early folliculogenesis, Mol Cell Endocrinol, vol.163, pp.89-93, 2000.

D. Monniaux, Driving folliculogenesis by the oocyte-somatic cell dialog: Lessons from genetic models, Theriogenology, vol.86, pp.41-53, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01529484

D. Monniaux, F. Clément, R. Dalbiès-tran, A. Estienne, S. Fabre et al., The ovarian reserve of primordial follicles and the dynamic reserve of antral growing follicles: what is the link?, Biol Reprod, vol.90, p.85, 2014.
URL : https://hal.archives-ouvertes.fr/hal-00961546

H. Moore, J. M. Unger, K. Phillips, F. Boyle, E. Hitre et al., Goserelin for ovarian protection during breast-cancer adjuvant chemotherapy, N Engl J Med, vol.372, pp.923-932, 2015.

Y. Morel, F. Roucher, D. Mallet, and I. Plotton, Genetic of gonadal determination, Ann Endocrinol, vol.75, pp.32-39, 2014.

, Références bibliographiques 144

S. Morgan, R. A. Anderson, C. Gourley, W. H. Wallace, and N. Spears, How do chemotherapeutic agents damage the ovary?, Hum Reprod Update, vol.18, pp.525-535, 2012.

S. Morgan, F. Lopes, C. Gourley, R. A. Anderson, and N. Spears, Cisplatin and doxorubicin induce distinct mechanisms of ovarian follicle loss; imatinib provides selective protection only against cisplatin, PloS One, vol.8, p.70117, 2013.

P. Mulsant, F. Lecerf, S. Fabre, L. Schibler, P. Monget et al., Mutation in bone morphogenetic protein receptor-IB is associated with increased ovulation rate in Booroola Mérino ewes, Proc Natl Acad Sci U S A, vol.98, pp.5104-5109, 2001.

P. N. Munster, A. P. Moore, R. Ismail-khan, C. E. Cox, M. Lacevic 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 Off J Am Soc Clin Oncol, vol.30, pp.533-538, 2012.

M. Myers, K. L. Britt, N. Wreford, F. Ebling, and J. B. Kerr, Methods for quantifying follicular numbers within the mouse ovary, Reprod Camb Engl, vol.127, pp.569-580, 2004.

L. Nicol, S. C. Bishop, R. Pong-wong, C. Bendixen, L. Holm et al., Homozygosity for a single base-pair mutation in the oocyte-specific GDF9 gene results in sterility in Thoka sheep, Reprod Camb Engl, vol.138, pp.921-933, 2009.

E. Nilsson, N. Rogers, and M. K. Skinner, Actions of anti-Mullerian hormone on the ovarian transcriptome to inhibit primordial to primary follicle transition, Reprod Camb Engl, vol.134, pp.209-221, 2007.

R. J. Norman, D. Dewailly, R. S. Legro, and T. E. Hickey, Polycystic ovary syndrome, Lancet Lond Engl, vol.370, pp.685-697, 2007.

S. Ogura-nose, O. Yoshino, Y. Osuga, J. Shi, H. Hiroi et al., Anti-Mullerian hormone (AMH) is induced by bone morphogenetic protein (BMP) cytokines in human granulosa cells, Eur J Obstet Gynecol Reprod Biol, vol.164, pp.44-47, 2012.

K. Oktay, E. Taylan, K. A. Rodriguez-wallberg, G. Bedoschi, V. Turan et al., Goserelin Does Not Preserve Ovarian Function Against Chemotherapy-Induced Damage, Ann Oncol Off J Eur Soc Med Oncol, 2017.

O. Oktem and K. Oktay, A novel ovarian xenografting model to characterize the impact of chemotherapy agents on human primordial follicle reserve, Cancer Res, vol.67, pp.10159-10162, 2007.

F. Otsuka, Z. Yao, T. Lee, S. Yamamoto, G. F. Erickson et al., Bone morphogenetic protein-15. Identification of target cells and biological functions, J Biol Chem, vol.275, pp.39523-39528, 2000.

O. Ustaalioglu, B. B. Bilici, A. Kefeli, U. Seker, M. Salepci et al., A retrospective analysis of women's chances to become pregnant after completion of chemotherapy: a single center experience, J BUON Off J Balk Union Oncol, vol.16, pp.349-352, 2011.

T. D. Pache, J. W. Wladimiroff, F. H. Jong, . De, W. C. Hop et al., Growth patterns of nondominant ovarian follicles during the normal menstrual cycle, Fertil Steril, vol.54, pp.638-642, 1990.

A. Pacheco, M. Cruz, G. Velasco, and J. A. , Impact of very low anti-Müllerian hormone on pregnancy success, Curr Opin Obstet Gynecol, vol.29, pp.131-135, 2017.

S. A. Pangas, Regulation of the ovarian reserve by members of the transforming growth factor beta family, Mol Reprod Dev, vol.79, pp.666-679, 2012.

S. A. Pangas, Y. Choi, D. J. Ballow, Y. Zhao, H. Westphal et al., Oogenesis requires germ cell-specific transcriptional regulators Sohlh1 and Lhx8, Proc Natl Acad Sci, vol.103, pp.8090-8095, 2006.

M. Pannetier, A. Chassot, M. Chaboissier, and E. Pailhoux, Involvement of FOXL2 and RSPO1 in Ovarian Determination, Development, and Maintenance in Mammals, Sex Dev Genet Mol Biol Evol Endocrinol Embryol Pathol Sex Determ Differ, vol.10, pp.167-184, 2016.

E. Papaleo, S. Zaffagnini, M. Munaretto, V. S. Vanni, G. Rebonato et al., Clinical application of a nomogram based on age, serum FSH and AMH to select the FSH starting dose in IVF/ICSI cycles: a retrospective two-centres study, Eur J Obstet Gynecol Reprod Biol, vol.207, pp.94-99, 2016.

A. Paredes, C. Garcia-rudaz, B. Kerr, V. Tapia, G. A. Dissen et al., Loss of synaptonemal complex protein-1, a synaptonemal complex protein, contributes to the initiation of follicular assembly in the developing rat ovary, Endocrinology, vol.146, pp.5267-5277, 2005.

J. Park, Y. Su, M. Ariga, E. Law, J. Conti et al., EGF-like growth factors as mediators of LH action in the ovulatory follicle, Science, vol.303, pp.682-684, 2004.

N. Pascuali, L. Scotti, D. Pietro, M. Oubiña, G. Bas et al., Ceramide-1-phosphate has protective properties against cyclophosphamide

, Hum Reprod Oxf Engl, 2018.

T. Pedersen, Determination of follicle growth rate in the ovary of the immature mouse, J Reprod Fertil, vol.21, pp.81-93, 1970.

T. Pedersen and H. Peters, Proposal for a classification of oocytes and follicles in the mouse ovary, J Reprod Fertil, vol.17, pp.555-557, 1968.

M. Peigné and C. Decanter, Serum AMH level as a marker of acute and long-term effects of chemotherapy on the ovarian follicular content: a systematic review, Reprod Biol Endocrinol RBE, vol.12, p.26, 2014.

E. Pelosi, S. Omari, M. Michel, J. Ding, T. Amano et al., Constitutively active Foxo3 in oocytes preserves ovarian reserve in mice, Nat Commun, vol.4, p.1843, 2013.

M. E. Pepling, Follicular assembly: mechanisms of action, Reprod Camb Engl, vol.143, pp.139-149, 2012.

M. E. Pepling and A. C. Spradling, Mouse ovarian germ cell cysts undergo programmed breakdown to form primordial follicles, Dev Biol, vol.234, pp.339-351, 2001.

N. Pereira, K. Hancock, C. N. Cordeiro, J. P. Lekovich, G. L. Schattman et al., Comparison of
URL : https://hal.archives-ouvertes.fr/hal-00795586

, Off J Int Soc Gynecol Endocrinol, vol.32, pp.823-826, 2016.

L. Persani, R. Rossetti, D. Pasquale, E. Cacciatore, C. Fabre et al., The fundamental role of bone morphogenetic protein 15 in ovarian function and its involvement in female fertility disorders, Hum Reprod Update, vol.20, pp.869-883, 2014.

J. Piasecka-srader, F. F. Blanco, D. H. Delman, D. A. Dixon, J. L. Geiser et al., Tamoxifen prevents apoptosis and follicle loss from cyclophosphamide in cultured rat ovaries, Biol Reprod, vol.92, p.132, 2015.

A. Pierre, A. Estienne, C. Racine, J. Picard, R. Fanchin et al., The Bone Morphogenetic Protein 15 Up-Regulates the Anti-Müllerian Hormone Receptor Expression in Granulosa Cells, J Clin Endocrinol Metab, vol.101, pp.2602-2611, 2016.

A. Pierre, M. Peigné, M. Grynberg, N. Arouche, J. Taieb et al., Loss of LH-induced down-regulation of anti-Müllerian hormone receptor expression may contribute to anovulation in women with polycystic ovary syndrome, Hum Reprod Oxf Engl, vol.28, pp.762-769, 2013.

A. Pierre, J. Taieb, F. Giton, M. Grynberg, S. Touleimat et al., Dysregulation of the Anti-Müllerian Hormone System by Steroids in Women With Polycystic Ovary Syndrome, J Clin Endocrinol Metab, vol.102, pp.3970-3978, 2017.

P. Pigny, S. Jonard, Y. Robert, and D. Dewailly, Serum anti-Mullerian hormone as a surrogate for antral follicle count for definition of the polycystic ovary syndrome, J Clin Endocrinol Metab, vol.91, pp.941-945, 2006.

C. Poirot, F. Abirached, M. Prades, C. Coussieu, F. Bernaudin et al., Induction of puberty by autograft of cryopreserved ovarian tissue, Lancet Lond Engl, vol.379, p.588, 2012.

, Références bibliographiques 148

D. H. Poole, O. M. Ocón-grove, and A. L. Johnson, Anti-Müllerian hormone (AMH) receptor type II expression and AMH activity in bovine granulosa cells, Theriogenology, vol.86, pp.1353-1360, 2016.

E. B. Prasath, M. Chan, W. Wong, C. Lim, M. D. Tharmalingam 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 Oxf Engl, vol.29, pp.276-278, 2014.

Y. Qin, X. Jiao, R. Dalgleish, S. Vujovic, J. Li et al., Novel variants in the SOHLH2 gene are implicated in human premature ovarian failure, Fertil Steril, vol.101, pp.1104-1109, 2014.

M. M. Quinn, C. Kao, A. K. Ahmad, D. J. Haisenleder, N. Santoro et al., Age-stratified thresholds of anti-Müllerian hormone improve prediction of polycystic ovary syndrome over a population-based threshold, Clin Endocrinol (Oxf), vol.87, pp.733-740, 2017.

S. Rajareddy, P. Reddy, C. Du, L. Liu, K. Jagarlamudi et al., p27kip1 (cyclin-dependent kinase inhibitor 1B) controls ovarian development by suppressing follicle endowment and activation and promoting follicle atresia in mice, Mol Endocrinol Baltim Md, vol.21, pp.2189-2202, 2007.

A. Rajkovic, S. A. Pangas, D. Ballow, N. Suzumori, and M. M. Matzuk, NOBOX deficiency disrupts early folliculogenesis and oocyte-specific gene expression, Science, vol.305, pp.1157-1159, 2004.

. Rapport_synthese_france_ia_,

P. Reddy, D. Adhikari, W. Zheng, S. Liang, T. Hämäläinen et al., PDK1 signaling in oocytes controls reproductive aging and lifespan by manipulating the survival of primordial follicles, Hum Mol Genet, vol.18, pp.2813-2824, 2009.

P. Reddy, L. Liu, D. Adhikari, K. Jagarlamudi, S. Rajareddy et al., Oocyte-specific deletion of Pten causes premature activation of the primordial follicle pool, Science, vol.319, pp.611-613, 2008.

P. Reddy, W. Zheng, and K. Liu, Mechanisms maintaining the dormancy and survival of mammalian primordial follicles, Trends Endocrinol Metab TEM, vol.21, pp.96-103, 2010.

E. J. Renaud, D. T. Maclaughlin, E. Oliva, B. R. Rueda, and P. K. Donahoe, Endometrial cancer is a receptormediated target for Mullerian Inhibiting Substance, Proc Natl Acad Sci U S A, vol.102, pp.111-116, 2005.

S. Rice, K. Ojha, S. Whitehead, and H. Mason, Stage-specific expression of androgen receptor, folliclestimulating hormone receptor, and anti-Müllerian hormone type II receptor in single, isolated, human preantral follicles: relevance to polycystic ovaries, J Clin Endocrinol Metab, vol.92, pp.1034-1040, 2007.

J. S. Richards and S. A. Pangas, The ovary: basic biology and clinical implications, J Clin Invest, vol.120, pp.963-972, 2010.

H. Roness, O. Kashi, and D. Meirow, Prevention of chemotherapy-induced ovarian damage, Fertil Steril, vol.105, pp.20-29, 2016.

V. Rossi, M. Lispi, S. Longobardi, M. Mattei, F. D. Rella et al., LH prevents cisplatin-induced apoptosis in oocytes and preserves female fertility in mouse, Cell Death Differ, vol.24, pp.72-82, 2017.

R. Roti, E. C. Ringelstetter, A. K. Kropp, J. Abbott, D. H. Salih et al., Bortezomib prevents acute doxorubicin ovarian insult and follicle demise, improving the fertility window and pup birth weight in mice, PloS One, vol.9, p.108174, 2014.

, Revised 2003 consensus on diagnostic criteria and long-term health risks related to polycystic ovary syndrome (PCOS), Rotterdam ESHRE/ASRM-Sponsored PCOS consensus workshop group, vol.19, pp.41-47, 2004.

, Références bibliographiques, p.150

D. O. Saleh and D. F. Mansour, Ovario-protective effects of genistein against cyclophosphamide toxicity in rats: Role of anti-müllerian hormone and oestradiol, Eur J Pharmacol, vol.789, pp.163-171, 2016.

H. S. Saleh, O. E. Froemming, G. Said, and R. M. , Tocotrienol preserves ovarian function in cyclophosphamide therapy, Hum Exp Toxicol, vol.34, pp.946-952, 2015.

S. Scalercio, A. B. Brito, S. Domingues, R. R. Santos, and C. A. Amorim, Immunolocalization of growth, inhibitory, and proliferative factors involved in initial ovarian folliculogenesis from adult common squirrel monkey (Saimiri collinsi), Reprod Sci Thousand Oaks Calif, vol.22, pp.68-74, 2015.

D. Schmidt, C. E. Ovitt, K. Anlag, S. Fehsenfeld, L. Gredsted et al., The murine wingedhelix transcription factor Foxl2 is required for granulosa cell differentiation and ovary maintenance, Dev Camb Engl, vol.131, pp.933-942, 2004.

K. Schmidt, N. Kryger-baggesen, A. G. Byskov, and C. Y. Andersen, Anti-Müllerian hormone initiates growth of human primordial follicles in vitro, Mol Cell Endocrinol, vol.234, pp.87-93, 2005.

A. Schneider, X. Zhi, F. Moreira, T. Lucia, R. G. Mondadori et al., Primordial follicle activation in the ovary of Ames dwarf mice, J Ovarian Res, vol.7, p.120, 2014.

I. Segers, I. Mateizel, E. Van-moer, J. Smitz, H. Tournaye 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, vol.32, pp.1221-1231, 2015.

D. L. Segev, Y. Hoshiya, A. E. Stephen, M. Hoshiya, T. T. Tran et al., Mullerian inhibiting substance regulates NFkappaB signaling and growth of mammary epithelial cells in vivo, J Biol Chem, vol.276, pp.26799-26806, 2001.

D. B. Seifer and Z. Merhi, Is AMH a regulator of follicular atresia?, J Assist Reprod Genet, vol.31, pp.1403-1407, 2014.

A. Seroka-vanhove, C. Sonigo, C. Roche, and M. Grynberg,

, J Gynecol Obstet Biol Reprod (Paris), vol.43, pp.559-571, 2014.

L. M. Shandley, A. Fothergill, J. B. Spencer, A. C. Mertens, H. N. Cottrell et al., Impact of cancer treatment on risk of infertility and diminished ovarian reserve in women with polycystic ovary syndrome, Fertil Steril, 2018.

Y. Shin, Y. Ren, H. Suzuki, K. J. Golnoski, H. W. Ahn et al., Transcription factors SOHLH1 and SOHLH2 coordinate oocyte differentiation without affecting meiosis I, J Clin Invest, vol.127, pp.2106-2117, 2017.

R. L. Siegel, K. D. Miller, and A. Jemal, Cancer statistics, CA Cancer J Clin, vol.68, pp.7-30, 2018.

M. E. Skaznik-wikiel, M. M. Mcguire, M. Sukhwani, J. Donohue, T. Chu et al.,

, Granulocyte colony-stimulating factor with or without stem cell factor extends time to premature ovarian insufficiency in female mice treated with alkylating chemotherapy

, Fertil Steril, vol.99, pp.2045-2054, 2013.

R. Soleimani, E. Heytens, Z. Darzynkiewicz, and K. Oktay, Mechanisms of chemotherapy-induced human ovarian aging: double strand DNA breaks and microvascular compromise, Aging, vol.3, pp.782-793, 2011.

Z. Song, H. Yu, P. Wang, G. Mao, W. Liu et al., Germ cell-specific Atg7 knockout results in primary ovarian insufficiency in female mice

, Cell Death Dis, vol.6, p.1589, 2015.

C. Sonigo, N. Sermondade, J. Benard, A. Benoit, J. Shore et al., The past, present and future of fertility preservation in cancer patients, Future Oncol Lond Engl, 2015.

M. Sönmezer, I. Türkçüo?lu, U. Co?kun, and K. Oktay, Random-start controlled ovarian hyperstimulation for emergency fertility preservation in letrozole cycles, Fertil Steril, vol.95, pp.2125-2134, 2011.

K. Staley and H. Scharfman, A woman's prerogative, Nat Neurosci, 2005.

A. Z. Steiner, D. Pritchard, F. Z. Stanczyk, J. S. Kesner, J. W. Meadows et al., Association Between Biomarkers of Ovarian Reserve and Infertility Among Older Women of Reproductive Age, JAMA, vol.318, pp.1367-1376, 2017.

H. I. Su, K. Maas, P. M. Sluss, R. J. Chang, J. E. Hall et al., The impact of depot GnRH agonist on AMH levels in healthy reproductive-aged women, J Clin Endocrinol Metab, vol.98, pp.1961-1966, 2013.

P. Sukumvanich, L. D. Case, K. Van-zee, S. E. Singletary, E. D. Paskett et al., Incidence and time course of bleeding after long-term amenorrhea after breast cancer treatment: a prospective study, Cancer, vol.116, pp.3102-3111, 2010.

Y. Sun, X. Sun, P. W. Dyce, W. Shen, and H. Chen, The role of germ cell loss during primordial follicle assembly: a review of current advances, Int J Biol Sci, vol.13, pp.449-457, 2017.

Y. Sun, Y. Wang, X. Sun, S. Cheng, L. Li et al., The role of autophagy during murine primordial follicle assembly, Aging, vol.10, pp.197-211, 2018.

M. I. Taskin, A. Yay, E. Adali, E. Balcioglu, and U. Inceboz, Protective effects of sildenafil citrate administration on cisplatin-induced ovarian damage in rats, Gynecol Endocrinol Off J Int Soc Gynecol Endocrinol, vol.31, pp.272-277, 2015.

F. R. Tehrani, M. Solaymani-dodaran, M. Tohidi, M. R. Gohari, and F. Azizi, Modeling age at menopause using serum concentration of anti-mullerian hormone, J Clin Endocrinol Metab, vol.98, pp.729-735, 2013.

J. Teixeira, S. Maheswaran, and P. K. Donahoe, Müllerian inhibiting substance: an instructive developmental hormone with diagnostic and possible therapeutic applications, Endocr Rev, vol.22, pp.657-674, 2001.

M. Teletin, N. Vernet, N. B. Ghyselinck, and M. Mark, Roles of Retinoic Acid in Germ Cell Differentiation, Curr Top Dev Biol, vol.125, pp.191-225, 2017.

Y. Tham, K. Sexton, H. Weiss, R. Elledge, L. C. Friedman et al., The rates of chemotherapyinduced amenorrhea in patients treated with adjuvant doxorubicin and cyclophosphamide followed by a taxane, Am J Clin Oncol, vol.30, pp.126-132, 2007.

, Références bibliographiques, p.153

Y. Tham, K. Sexton, H. Weiss, R. Elledge, L. C. Friedman et al., The rates of chemotherapyinduced amenorrhea in patients treated with adjuvant doxorubicin and cyclophosphamide followed by a taxane, Am J Clin Oncol, vol.30, pp.126-132, 2007.

J. L. Tilly, Ovarian follicle counts--not as simple as 1, 2, 3, Reprod Biol Endocrinol RBE, vol.1, p.11, 2003.

A. Y. Ting and B. K. Petroff, Tamoxifen decreases ovarian follicular loss from experimental toxicant DMBA and chemotherapy agents cyclophosphamide and doxorubicin in the rat, J Assist Reprod Genet, vol.27, pp.591-597, 2010.

C. Tingen, A. Kim, and T. K. Woodruff, The primordial pool of follicles and nest breakdown in mammalian ovaries, Mol Hum Reprod, vol.15, pp.795-803, 2009.

M. Uda, C. Ottolenghi, L. Crisponi, J. E. Garcia, M. Deiana et al., Foxl2 disruption causes mouse ovarian failure by pervasive blockage of follicle development, Hum Mol Genet, vol.13, pp.1171-1181, 2004.

P. S. Uzelac, A. A. Delaney, G. L. Christensen, H. Bohler, and S. T. Nakajima, Live birth following in vitro maturation of oocytes retrieved from extracorporeal ovarian tissue aspiration and embryo cryopreservation for 5 years, Fertil Steril, vol.104, pp.1258-1260, 2015.

A. Vaiarelli, R. Venturella, D. Vizziello, F. Bulletti, and F. M. Ubaldi, Dual ovarian stimulation and random start in assisted reproductive technologies: from ovarian biology to clinical application

, Curr Opin Obstet Gynecol, vol.29, pp.153-159, 2017.

A. C. Van-erp, D. Hoeksma, R. A. Rebolledo, P. J. Ottens, I. Jochmans et al., The Crosstalk between ROS and Autophagy in the Field of Transplantation Medicine, Oxid Med Cell Longev, p.7120962, 2017.

K. A. Vendola, J. Zhou, O. O. Adesanya, S. J. Weil, and C. A. Bondy, Androgens stimulate early stages of follicular growth in the primate ovary, J Clin Invest, vol.101, pp.2622-2629, 1998.

B. Vigier, M. G. Forest, B. Eychenne, J. Bézard, O. Garrigou et al., Anti-Müllerian hormone produces endocrine sex reversal of fetal ovaries, Proc Natl Acad Sci U S A, vol.86, pp.3684-3688, 1989.

B. Vigier, D. Tran, L. Legeai, J. Bézard, and N. Josso, Origin of anti-Müllerian hormone in bovine freemartin fetuses, J Reprod Fertil, vol.70, pp.473-479, 1984.

D. Vinatier, P. Dufour, N. Tordjeman-rizzi, J. F. Prolongeau, S. Depret-moser et al., Immunological aspects of ovarian function: role of the cytokines, Eur J Obstet Gynecol Reprod Biol, vol.63, pp.155-168, 1995.

J. A. Visser, F. H. De-jong, J. Laven, and A. Themmen, Anti-Müllerian hormone: a new marker for ovarian function, Reprod Camb Engl, vol.131, pp.1-9, 2006.

J. A. Visser, A. Durlinger, I. Peters, E. R. Heuvel, . Van-den et al., Increased oocyte degeneration and follicular atresia during the estrous cycle in antiMüllerian hormone null mice, Endocrinology, vol.148, pp.2301-2308, 2007.

J. A. Visser, I. Schipper, J. Laven, and A. Themmen, Anti-Müllerian hormone: an ovarian reserve marker in primary ovarian insufficiency, Nat Rev Endocrinol, vol.8, pp.331-341, 2012.

J. A. Visser and A. Themmen, Anti-Müllerian hormone and folliculogenesis, Mol Cell Endocrinol, vol.234, pp.81-86, 2005.

M. Voorhuis, F. J. Broekmans, B. Fauser, N. C. Onland-moret, Y. T. Schouw et al., Genes involved in initial follicle recruitment may be associated with age at menopause, J Clin Endocrinol Metab, vol.96, pp.473-479, 2011.

C. Weenen, J. Laven, V. Bergh, A. Cranfield, M. Groome et al., Anti-Müllerian hormone expression pattern in the human ovary: potential implications for initial and cyclic follicle recruitment, Mol Hum Reprod, vol.10, pp.77-83, 2004.

S. Wesselborg and B. Stork, Autophagy signal transduction by ATG proteins: from hierarchies to networks, Cell Mol Life Sci CMLS, vol.72, pp.4721-4757, 2015.

T. Wilson, X. Y. Wu, J. L. Juengel, I. K. Ross, J. M. Lumsden et al., Highly prolific Booroola sheep have a mutation in the intracellular kinase domain of bone morphogenetic protein IB receptor (ALK-6) that is expressed in both oocytes and granulosa cells, Biol Reprod, vol.64, pp.1225-1235, 2001.

N. Winkler, O. Bukulmez, D. B. Hardy, and B. R. Carr, Gonadotropin releasing hormone antagonists suppress aromatase and anti-Müllerian hormone expression in human granulosa cells

, Fertil Steril, vol.94, pp.1832-1839, 2010.

A. L. Winship, J. M. Stringer, S. H. Liew, and K. J. Hutt, The importance of DNA repair for maintaining oocyte quality in response to anti-cancer treatments, environmental toxins and maternal ageing

, Hum Reprod Update, 2018.

. Wolff-m-von, A. Germeyer, J. Liebenthron, M. Korell, and F. Nawroth, Practical recommendations for fertility preservation in women by the FertiPROTEKT network. Part II: fertility preservation techniques, Arch Gynecol Obstet, vol.297, pp.257-267, 2018.

T. Xia, Y. Fu, H. Gao, Z. Zhao, L. Zhao et al., Recovery of ovary function impaired by chemotherapy using Chinese herbal medicine in a rat model, Syst Biol Reprod Med, vol.60, pp.293-303, 2014.

C. Yan, P. Wang, J. Demayo, F. J. Demayo, J. A. Elvin et al., Synergistic roles of bone morphogenetic protein 15 and growth differentiation factor 9 in ovarian function, Mol Endocrinol Baltim Md, vol.15, pp.854-866, 2001.

M. Y. Yang, R. A. Cushman, and J. E. Fortune, Anti-Müllerian hormone inhibits activation and growth of bovine ovarian follicles in vitro and is localized to growing follicles, Mol Hum Reprod, pp.1-10, 2017.

M. Y. Yang and J. E. Fortune, The capacity of primordial follicles in fetal bovine ovaries to initiate growth in vitro develops during mid-gestation and is associated with meiotic arrest of oocytes

, Biol Reprod, vol.78, pp.1153-1161, 2008.

Z. Yang and R. Chian, Development of in vitro maturation techniques for clinical applications, Fertil Steril, vol.108, pp.577-584, 2017.

J. Zhai, G. Yao, F. Dong, Z. Bu, Y. Cheng et al., In Vitro Activation of Follicles and Fresh Tissue Auto-transplantation in Primary Ovarian Insufficiency Patients, J Clin Endocrinol Metab, vol.101, pp.4405-4412, 2016.

, Références bibliographiques, p.156

X. Zhang, L. Li, J. Xu, N. Wang, W. Liu et al., Rapamycin preserves the follicle pool reserve and prolongs the ovarian lifespan of female rats via modulating mTOR activation and sirtuin expression, Gene, vol.523, pp.82-87, 2013.

W. Zheng, G. Nagaraju, Z. Liu, and K. Liu, Functional roles of the phosphatidylinositol 3-kinases (PI3Ks) signaling in the mammalian ovary, Mol Cell Endocrinol, vol.356, pp.24-30, 2012.

J. Zhou, T. R. Kumar, M. M. Matzuk, and C. Bondy, Insulin-like growth factor I regulates gonadotropin responsiveness in the murine ovary, Mol Endocrinol Baltim Md, vol.11, pp.1924-1933, 1997.

L. Zhou, Y. Xie, S. Li, Y. Liang, Q. Qiu et al.,

R. L. Siegel, K. D. Miller, and A. Jemal, Cancer statistics, 2015. CA, Cancer J. Clin, vol.65, issue.1, pp.5-29, 2015.

. Isfp-practice-committee, S. S. Kim, and J. Donnez, Recommendations for fertility preservation in patients with lymphoma, leukemia, and breast cancer

, J. Assist. Reprod. Genet, vol.29, issue.6, pp.465-468, 2012.

, Ethics Committee of American Society for Reproductive Medicine. Fertility preservation and reproduction in patients facing gonadotoxic therapies: a committee opinion, Fertil. Steril, vol.100, issue.5, pp.1224-1231, 2013.

, Epub ahead of print), Future Oncol

?. , Latest recommendation from the American Society for Reproductive Medicine society

A. W. Loren, P. B. Mangu, and L. N. Beck, Fertility preservation for patients with cancer: American Society of Clinical Oncology clinical practice guideline update, J. Clin. Oncol, vol.31, issue.19, pp.2500-2510, 2013.

?. Latest,

K. A. Rodriguez-wallberg and K. Oktay, Fertility preservation during cancer treatment: clinical guidelines, Cancer Manag. Res, vol.6, pp.105-117, 2014.

D. H. Williams, E. Karpman, J. C. Sander, P. E. Spiess, L. L. Pisters et al., Pretreatment semen parameters in men with cancer, J. Urol, vol.181, issue.2, pp.736-740, 2009.

N. J. Van-casteren, W. Boellaard, J. C. Romijn, and G. R. Dohle, Gonadal dysfunction in male cancer patients before cytotoxic treatment, Int. J. Androl, vol.33, issue.1, pp.73-79, 2010.

W. Wallace, R. A. Anderson, and D. S. Irvine, Fertility preservation for young patients with cancer: who is at risk and what can be offered?, Lancet Oncol, vol.6, issue.4, pp.209-218, 2005.

H. Tournaye, G. R. Dohle, and C. Barratt, Fertility preservation in men with cancer, Lancet, vol.384, issue.9950, pp.1295-1301, 2014.

B. E. Oosterhuis, T. Goodwin, M. Kiernan, M. M. Hudson, and G. V. Dahl, Concerns about infertility risks among pediatric oncology patients and their parents. Pediatr, Blood Cancer, vol.50, issue.1, pp.85-89, 2008.

R. G. Bunge and J. K. Sherman, Fertilizing capacity of frozen human spermatozoa, Nature, vol.172, issue.4382, pp.767-768, 1953.

G. Horne, A. D. Atkinson, E. Pease, J. P. Logue, D. R. Brison et al., Live birth with sperm cryopreserved for 21 years prior to cancer treatment: case report, Hum. Reprod. Oxf. Engl, vol.19, issue.6, pp.1448-1449, 2004.

I. D. Morris, Sperm DNA damage and cancer treatment, Int. J. Androl, vol.25, issue.5, pp.255-261, 2002.

A. K. Nangia, S. A. Krieg, and S. S. Kim, Clinical guidelines for sperm cryopreservation in cancer patients, Fertil. Steril, vol.100, issue.5, pp.1203-1209, 2013.

B. M. Berookhim and J. P. Mulhall, Outcomes of operative sperm retrieval strategies for fertility preservation among males scheduled to undergo cancer treatment, Fertil. Steril, vol.101, issue.3, pp.805-811, 2014.

P. Bizet, J. Saias-magnan, and J. E. , Sperm cryopreservation before cancer treatment: a 15-year monocentric experience, Reprod. Biomed. Online, vol.24, issue.3, pp.321-330, 2012.

F. Abdelhafez, M. Bedaiwy, S. A. El-nashar, E. Sabanegh, and N. Desai, Techniques for cryopreservation of individual or small numbers of human spermatozoa: a systematic review, Hum. Reprod. Update, vol.15, issue.2, pp.153-164, 2009.

A. Stein, Y. Shufaro, S. Hadar, B. Fisch, and H. Pinkas, Successful use of the Cryolock device for cryopreservation of scarce human ejaculate and testicular spermatozoa, Andrology, vol.3, issue.2, pp.220-224, 2015.

K. Nysom, J. L. Pedersen, and M. Jørgensen, Spermaturia in two normal boys without other signs of puberty, Acta Paediatr. Oslo Nor, vol.83, issue.5, pp.520-521, 1994.

M. Daudin, N. Rives, and M. Walschaerts, Sperm cryopreservation in adolescents and young adults with cancer: results of the French national sperm banking network (CECOS)
URL : https://hal.archives-ouvertes.fr/hal-01657933

, Fertil. Steril, vol.103, issue.2, pp.478-486, 2015.

I. Hagenäs, N. Jørgensen, and C. Rechnitzer, Clinical and biochemical correlates of successful semen collection for cryopreservation from 12-18-year-old patients: a single-center study of 86 adolescents, Hum. Reprod. Oxf. Engl, vol.25, issue.8, pp.2031-2038, 2010.

G. Bahadur, K. Ling, and R. Hart, Semen quality and cryopreservation in adolescent cancer patients, Hum. Reprod. Oxf. Engl, vol.17, issue.12, pp.3157-3161, 2002.

Z. Laron, Age at first ejaculation (spermarche)-the overlooked milestone in male development, Pediatr. Endocrinol. Rev, vol.7, issue.3, pp.256-257, 2010.

M. C. Adank, W. Van-dorp, and M. Smit, Electroejaculation as a method of fertility preservation in boys diagnosed with cancer: a single-center experience and review of the literature, Fertil. Steril, vol.102, issue.1, 2014.

I. Gat, A. Toren, and A. Hourvitz, Sperm preservation by electroejaculation in adolescent cancer patients. Pediatr, Blood Cancer, vol.61, issue.2, pp.286-290, 2014.

E. Goossens, D. Van-saen, and H. Tournaye, Spermatogonial stem cell preservation and transplantation: from research to clinic, Hum. Reprod. Oxf. Engl, vol.28, issue.4, pp.897-907, 2013.

Y. Hayashi, M. Saitou, and S. Yamanaka, Germline development from human pluripotent stem cells toward disease modeling of infertility, Fertil. Steril, vol.97, issue.6, pp.1250-1259, 2012.

J. Poels, A. Van-langendonckt, M. Many, F. Wese, and C. Wyns, Vitrification preserves proliferation capacity in human spermatogonia, Hum. Reprod. Oxf. Engl, vol.28, issue.3, pp.578-589, 2013.

E. Goossens, M. Geens, D. Block, G. Tournaye, and H. , Spermatogonial survival in long-term human prepubertal xenografts, Fertil. Steril, vol.90, issue.5, pp.2019-2022, 2008.

D. Van-saen, I. Gies, D. Schepper, J. Tournaye, H. Goossens et al., Can pubertal boys with Klinefelter syndrome benefit from spermatogonial stem cell banking?, Hum. Reprod. Oxf. Engl, vol.27, issue.2, pp.323-330, 2012.

T. Shinohara, K. Inoue, and N. Ogonuki, Birth of offspring following transplantation of cryopreserved immature testicular pieces and in-vitro microinsemination, Hum. Reprod. Oxf. Engl, vol.17, issue.12, pp.3039-3045, 2002.

M. Nakai, H. Kaneko, and T. Somfai, Production of viable piglets for the first time using sperm derived from ectopic testicular xenografts, Reprod. Camb. Engl, vol.139, issue.2, pp.331-335, 2010.

R. L. Brinster and J. W. Zimmermann, Spermatogenesis following male germ-cell transplantation, Proc. Natl Acad. Sci. USA, vol.91, issue.24, pp.11298-11302, 1994.

E. Goossens, V. Frederickx, D. Block, G. , V. Steirteghem et al., Reproductive capacity of sperm obtained after germ cell transplantation in a mouse model, Hum. Reprod. Oxf. Engl, vol.18, issue.9, pp.1874-1880, 2003.

B. P. Hermann, M. Sukhwani, and F. Winkler, Spermatogonial stem cell transplantation into rhesus testes regenerates spermatogenesis producing functional sperm, Cell Stem Cell, vol.11, issue.5, pp.715-726, 2012.

H. Sadri-ardekani, M. A. Akhondi, F. Van-der-veen, S. Repping, and A. Van-pelt, In vitro propagation of human prepubertal spermatogonial stem cells, JAMA, vol.305, issue.23, pp.2416-2418, 2011.

M. Geens, E. Goossens, and H. Tournaye, Cell selection by selective matrix adhesion is not sufficiently efficient for complete malignant cell depletion from contaminated human testicular cell suspensions, Fertil. Steril, vol.95, issue.2, pp.787-791, 2011.

T. Sato, K. Katagiri, and A. Gohbara, In vitro production of functional sperm in cultured neonatal mouse testes, Nature, vol.471, issue.7339, pp.504-507, 2011.

H. Mahmoud, Concise review: spermatogenesis in an artificial threedimensional system, Stem Cells Dayt. Ohio, vol.30, issue.11, pp.2355-2360, 2012.

N. Irie, L. Weinberger, and W. Tang, SOX17 Is a critical specifier of human primordial germ cell fate, Cell, vol.160, issue.1-2, pp.253-268, 2015.

J. Donnez, B. Martinez-madrid, P. Jadoul, A. V. Langendonckt, and D. Demylle,

. Review-sonigo, . Sermondade, and . Benard, future science group M-M. Ovarian tissue cryopreservation and transplantation: a review, vol.12, pp.519-535, 2006.

H. Roness, L. Kalich-philosoph, and D. Meirow, Prevention of chemotherapy-induced ovarian damage: possible roles for hormonal and non-hormonal attenuating agents, Hum. Reprod. Update, vol.20, issue.5, pp.759-774, 2014.

, ?? An excellent review with all developed molecules to reduce, in vivo

D. Meirow, H. Biederman, R. A. Anderson, and W. Wallace, Toxicity of chemotherapy and radiation on female reproduction, Clin. Obstet. Gynecol, vol.53, issue.4, pp.727-739, 2010.

C. A. Sklar, A. C. Mertens, and P. Mitby, Premature menopause in survivors of childhood cancer: a report from the childhood cancer survivor study, J. Natl Cancer Inst, vol.98, issue.13, pp.890-896, 2006.

R. A. Anderson and W. Wallace, Antimüllerian hormone, the assessment of the ovarian reserve, and the reproductive outcome of the young patient with cancer, Fertil. Steril, vol.99, issue.6, pp.1469-1475, 2013.

K. Gubbala, A. Laios, I. Gallos, P. Pathiraja, K. Haldar et al., Outcomes of ovarian transposition in gynaecological cancers; a systematic review and meta-analysis

, J. Ovarian Res, vol.7, p.69, 2014.

K. Ataya, L. V. Rao, E. Lawrence, and R. Kimmel, Luteinizing hormone-releasing hormone agonist inhibits cyclophosphamide-induced ovarian follicular depletion in rhesus monkeys, Biol. Reprod, vol.52, issue.2, pp.365-372, 1995.

M. A. Edson, A. K. Nagaraja, and M. M. Matzuk, The mammalian ovary from genesis to revelation, Endocr. Rev, vol.30, issue.6, pp.624-712, 2009.

D. Meirow, J. Dor, and B. Kaufman, Cortical fibrosis and blood-vessels damage in human ovaries exposed to chemotherapy. Potential mechanisms of ovarian injury, Hum. Reprod. Oxf. Engl, vol.22, issue.6, pp.1626-1633, 2007.

Z. Blumenfeld, How to preserve fertility in young women exposed to chemotherapy? The role of GnRH agonist cotreatment in addition to cryopreservation of embrya, oocytes, or ovaries, Oncologist, vol.12, issue.9, pp.1044-1054, 2007.

N. Hasky, S. Uri-belapolsky, and K. Goldberg, Gonadotrophin-releasing hormone agonists for fertility preservation: unraveling the enigma?, Hum. Reprod. Oxf. Engl, vol.30, issue.5, pp.1089-1101, 2015.

H. Moore, J. M. Unger, and K. Phillips, Goserelin for ovarian protection during breast-cancer adjuvant chemotherapy, N. Engl. J. Med, vol.372, issue.10, pp.923-932, 2015.

V. Wolff, M. Montag, M. Dittrich, R. Denschlag, D. Nawroth et al., Fertility preservation in women -a practical guide to preservation techniques and therapeutic strategies in breast cancer, Hodgkin's lymphoma and borderline ovarian tumours by the fertility preservation network FertiPROTEKT, Arch. Gynecol. Obstet, vol.284, issue.2, pp.427-435, 2011.

R. Ronn and H. Holzer, Oncofertility in Canada: gonadal protection and fertilitysparing strategies, Curr. Oncol, vol.20, issue.6, pp.602-607, 2013.

A. Cobo, V. Serra, N. Garrido, I. Olmo, A. Pellicer et al., Obstetric and perinatal outcome of babies born from vitrified oocytes, Fertil. Steril, vol.102, issue.4, 2014.

Z. Li, Y. A. Wang, W. Ledger, D. H. Edgar, and E. A. Sullivan, Clinical outcomes following cryopreservation of blastocysts by vitrification or slow freezing: a population-based cohort study, Hum. Reprod. Oxf. Engl, vol.29, issue.12, pp.2794-2801, 2014.

F. F. Abdelhafez, N. Desai, A. M. Abou-setta, T. Falcone, and J. Goldfarb, Slow freezing, vitrification and ultra-rapid freezing of human embryos: a systematic review and metaanalysis, Reprod. Biomed. Online, vol.20, issue.2, pp.209-222, 2010.

V. Wolff, M. Thaler, C. J. Frambach, and T. , Ovarian stimulation to cryopreserve fertilized oocytes in cancer patients can be started in the luteal phase, Fertil. Steril, vol.92, issue.4, pp.1360-1365, 2009.

V. Turan, G. Bedoschi, F. Moy, and K. Oktay, Safety and feasibility of performing two consecutive ovarian stimulation cycles with the use of letrozole-gonadotropin protocol for fertility preservation in breast cancer patients, Fertil. Steril, vol.100, issue.6, 2013.

K. Oktay, A. Hourvitz, and G. Sahin, Letrozole reduces estrogen and gonadotropin exposure in women with breast cancer undergoing ovarian stimulation before chemotherapy

, J. Clin. Endocrinol. Metab, vol.91, issue.10, pp.3885-3890, 2006.

D. Meirow, H. Raanani, and E. Maman, Tamoxifen co-administration during controlled ovarian hyperstimulation for in vitro fertilization in breast cancer patients increases the safety of fertility-preservation treatment strategies, Fertil. Steril, vol.102, issue.2, 2014.

, Practice Committees of American Society for Reproductive Medicine, Society for Assisted Reproductive Technology, Fertil. Steril, vol.99, issue.1, pp.37-43, 2013.

A. P. Cil, H. Bang, and K. Oktay, Age-specific probability of live birth with oocyte cryopreservation: an individual patient data meta-analysis, Fertil. Steril, vol.100, issue.2, pp.492-499, 2013.

D. Yang, S. E. Brown, K. Nguyen, V. Reddy, C. Brubaker et al., Live birth after the transfer of human embryos developed from cryopreserved oocytes harvested before cancer treatment, Fertil. Steril, vol.87, issue.6, pp.1469-1470, 2007.

E. Porcu, S. Venturoli, and G. Damiano, Healthy twins delivered after oocyte cryopreservation and bilateral ovariectomy for ovarian cancer, Reprod. Biomed. Online, vol.17, issue.2, pp.265-267, 2008.

M. K. Kim, D. R. Lee, and J. E. Han, Live birth with vitrified-warmed oocytes of a chronic myeloid leukemia patient nine years after allogenic bone marrow transplantation

, J. Assist. Reprod. Genet, vol.28, issue.12, pp.1167-1170, 2011.

J. A. Garcia-velasco, J. Domingo, A. Cobo, M. Martínez, L. Carmona et al., Five years' experience using oocyte vitrification to preserve fertility for medical and nonmedical indications, Fertil. Steril, vol.99, issue.7, pp.1994-1999, 2013.

M. Alvarez, M. Solé, and M. Devesa, Live birth using vitrified -warmed oocytes in invasive ovarian cancer: case report and literature review, Reprod. Biomed. Online, vol.28, issue.6, pp.663-668, 2014.

E. Da-motta, M. Bonavita, J. R. Alegretti, M. Chehin, and P. Serafini, Live birth after 6 years of oocyte vitrification in a survivor with breast cancer, J. Assist. Reprod. Genet, vol.31, issue.10, pp.1397-1400, 2014.

M. Sánchez-serrano, J. Crespo, and V. Mirabet, Twins born after transplantation of ovarian cortical tissue and oocyte vitrification, Fertil. Steril, vol.93, issue.1, pp.11-13, 2010.

S. K. Sunkara, V. Rittenberg, N. Raine-fenning, S. Bhattacharya, J. Zamora et al., Association between the number of eggs and live birth in IVF treatment: an analysis of 400 135 treatment cycles, Hum. Reprod. Oxf. Engl, vol.26, issue.7, pp.1768-1774, 2011.

E. Maman, D. Meirow, M. Brengauz, H. Raanani, J. Dor et al., Luteal Phase oocyte retrieval and in vitro maturation is an optional procedure for urgent fertility preservation, Fertil. Steril, vol.95, issue.1, pp.64-67, 2011.

R. Chian, J. Huang, and L. Gilbert, Obstetric outcomes following vitrification of in vitro and in vivo matured oocytes, Fertil. Steril, vol.91, issue.6, pp.2391-2398, 2009.

, Epub ahead of print), Future Oncol

M. Grynberg, E. Hachem, H. De-bantel, A. Benard, J. Le-parco et al., In vitro maturation of oocytes: uncommon indications, Fertil. Steril, vol.99, issue.5, pp.1182-1188, 2013.

G. Fasano, F. Moffa, J. Dechène, Y. Englert, and I. Demeestere, Vitrification of in vitro matured oocytes collected from antral follicles at the time of ovarian tissue cryopreservation, Reprod. Biol. Endocrinol, vol.9, p.150, 2011.

E. B. Prasath, M. Chan, and W. Wong, First pregnancy and live birth resulting from cryopreserved embryos obtained from in vitro matured oocytes after oophorectomy in an ovarian cancer patient, Hum. Reprod. Oxf. Engl, vol.29, issue.2, pp.276-278, 2014.

R. Fadini, M. Renzini, M. Guarnieri, and T. , Comparison of the obstetric and perinatal outcomes of children conceived from in vitro or in vivo matured oocytes in in vitro maturation treatments with births from conventional ICSI cycles, Hum. Reprod. Oxf. Engl, vol.27, issue.12, pp.3601-3608, 2012.

G. Pliushch, E. Schneider, and T. Schneider, In vitro maturation of oocytes is not associated with altered deoxyribonucleic acid methylation patterns in children from in vitro fertilization or intracytoplasmic sperm injection, Fertil. Steril, vol.103, issue.3, pp.720-727, 2015.

A. Gremeau, N. Andreadis, and M. Fatum, In vitro maturation or in vitro fertilization for women with polycystic ovaries? A case-control study of 194 treatment cycles, Fertil. Steril, vol.98, issue.2, pp.355-360, 2012.

C. X. Yeo, R. B. Gilchrist, J. G. Thompson, and M. Lane, Exogenous growth differentiation factor 9 in oocyte maturation media enhances subsequent embryo development and fetal viability in mice, Hum. Reprod. Oxf. Engl, vol.23, issue.1, pp.67-73, 2008.

F. K. Albuz, M. Sasseville, M. Lane, D. T. Armstrong, J. G. Thompson et al., Simulated physiological oocyte maturation (SPOM): a novel in vitro maturation system that substantially improves embryo yield and pregnancy outcomes, Hum. Reprod. Oxf. Engl, vol.25, issue.12, pp.2999-3011, 2010.

M. De-vos, J. Smitz, and T. K. Woodruff, Fertility preservation in women with cancer, Lancet, vol.384, issue.9950, pp.1302-1310, 2014.

K. T. Schmidt, N. Andersen, A. Greve, T. Ernst, E. Loft et al., Fertility in cancer patients after cryopreservation of one ovary, Reprod. Biomed. Online, vol.26, issue.3, pp.272-279, 2013.

J. Donnez and M. Dolmans, Transplantation of ovarian tissue, Best Pract. Res. Clin. Obstet. Gynaecol, vol.28, issue.8, pp.1188-1197, 2014.

J. Donnez, M. Dolmans, and A. Pellicer, Restoration of ovarian activity and pregnancy after transplantation of cryopreserved ovarian tissue: a review of 60 cases of reimplantation, Fertil. Steril, vol.99, issue.6, pp.1503-1513, 2013.

F. Sauvat, J. Bouilly, and C. Capito, Ovarian function is restored after grafting of cryopreserved immature ovary in ewes, FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol, vol.27, issue.4, pp.1511-1518, 2013.

C. Poirot, F. Abirached, M. Prades, C. Coussieu, F. Bernaudin et al., Induction of puberty by autograft of cryopreserved ovarian tissue, Lancet, vol.379, issue.9815, p.588, 2012.

S. S. Kim, Assessment of long term endocrine function after transplantation of frozenthawed human ovarian tissue to the heterotopic site: 10 year longitudinal follow-up study, J. Assist. Reprod. Genet, vol.29, issue.6, pp.489-493, 2012.

C. A. Amorim, S. Jacobs, and R. V. Devireddy, Successful vitrification and autografting of baboon (Papio anubis) ovarian tissue, Hum. Reprod. Oxf. Engl, vol.28, issue.8, pp.2146-2156, 2013.

S. Herraiz, E. Novella-maestre, and B. Rodríguez, Improving ovarian tissue cryopreservation for oncologic patients: slow freezing versus vitrification, effect of different procedures and devices, Fertil. Steril, vol.101, issue.3, pp.775-784, 2014.

K. Kawamura, Y. Cheng, and N. Suzuki, Hippo signaling disruption and Akt stimulation of ovarian follicles for infertility treatment, Proc. Natl Acad. Sci. USA, vol.110, issue.43, pp.17474-17479, 2013.

N. Suzuki, N. Yoshioka, and S. Takae, Successful fertility preservation following ovarian tissue vitrification in patients with primary ovarian insufficiency, Hum. Reprod. Oxf. Engl, vol.30, issue.3, pp.608-615, 2015.

Z. Gavish, G. Peer, H. Roness, Y. Cohen, and D. Meirow, Follicle activation and "burn-out" contribute to post-transplantation follicle loss in ovarian tissue grafts: the effect of graft thickness, Hum. Reprod. Oxf. Engl, vol.30, issue.4, p.1003, 2015.

M. Dolmans, V. Luyckx, J. Donnez, C. Y. Andersen, and T. Greve, Risk of transferring malignant cells with transplanted frozenthawed ovarian tissue, Fertil. Steril, vol.99, issue.6, pp.1514-1522, 2013.

J. Donnez and M. Dolmans, Fertility preservation in women, Nat. Rev. Endocrinol, vol.9, issue.12, pp.735-749, 2013.

T. Zver, M. Alvergnas-vieille, F. Garnache-ottou, C. Ferrand, C. Roux et al., Minimal residual disease detection in cryopreserved ovarian tissue by multicolor flow cytometry in acute myeloid leukemia, Haematologica, vol.99, issue.12, pp.249-252, 2014.

J. Vanacker, V. Luyckx, and M. Dolmans, Transplantation of an alginate-matrigel matrix containing isolated ovarian cells: first step in developing a biodegradable scaffold to transplant isolated preantral follicles and ovarian cells, Biomaterials, vol.33, issue.26, pp.6079-6085, 2012.

V. Luyckx, M. Dolmans, J. Vanacker, S. R. Scalercio, J. Donnez et al., First step in developing a 3D biodegradable fibrin scaffold for an artificial ovary, J. Ovarian Res, vol.6, issue.1, p.83, 2013.

V. Luyckx, M. Dolmans, and J. Vanacker, A new step toward the artificial ovary: survival and proliferation of isolated murine follicles after autologous transplantation in a fibrin scaffold, Fertil. Steril, vol.101, issue.4, pp.1149-1156, 2014.

J. J. Eppig and M. J. O'brien, Development in vitro of mouse oocytes from primordial follicles, Biol. Reprod, vol.54, issue.1, pp.197-207, 1996.

M. J. O'brien, J. K. Pendola, and J. J. Eppig, A revised protocol for in vitro development of mouse oocytes from primordial follicles dramatically improves their developmental competence, Biol. Reprod, vol.68, issue.5, pp.1682-1686, 2003.

E. E. Telfer, M. Mclaughlin, C. Ding, and K. J. Thong, A two-step serum-free culture system supports development of human oocytes from primordial follicles in the presence of activin, Hum. Reprod. Oxf. Engl, vol.23, issue.5, pp.1151-1158, 2008.

E. E. Telfer and M. B. Zelinski, Ovarian follicle culture: advances and challenges for human and nonhuman primates, Fertil. Steril, vol.99, issue.6, pp.1523-1533, 2013.

R. A. Anderson, M. Mclaughlin, W. Wallace, D. F. Albertini, and E. E. Telfer, The immature human ovary shows loss of abnormal follicles and increasing follicle developmental competence through childhood and adolescence, Hum. Reprod. Oxf. Engl, vol.29, issue.1, pp.97-106, 2014.

J. Johnson, J. Canning, T. Kaneko, J. K. Pru, and J. L. Tilly, Germline stem cells and follicular renewal in the postnatal mammalian ovary, Nature, vol.428, issue.6979, pp.145-150, 2004.

Y. White, D. C. Woods, Y. Takai, O. Ishihara, H. Seki et al., Oocyte formation by mitotically active germ cells purified from ovaries of reproductive-age women, Nat. Med, vol.18, issue.3, pp.413-421, 2012.

K. Hayashi, S. Ogushi, K. Kurimoto, S. Shimamoto, H. Ohta et al.,

. Review-sonigo, . Sermondade, and . Benard,

C. E. Dunlop, E. E. Telfer, and R. A. Anderson, Ovarian stem cells -potential roles in infertility treatment and fertility preservation, Maturitas, vol.76, issue.3, pp.279-283, 2013.

M. Brännström, L. Johannesson, and H. Bokström, Livebirth after uterus transplantation, Lancet, vol.385, issue.9968, pp.607-616, 2015.

M. Brännström, L. Johannesson, and P. Dahmkähler, First clinical uterus transplantation trial: a six-month report, Fertil. Steril, vol.101, issue.5, pp.1228-1236, 2014.

R. M. Farrell and T. Falcone, Uterine transplant: new medical and ethical considerations, Lancet, vol.385, issue.9968, pp.581-582, 2015.

M. Hellström, R. R. El-akouri, and C. Sihlbom, Towards the development of a bioengineered uterus: comparison of different protocols for rat uterus decellularization, Acta Biomater, vol.10, issue.12, pp.5034-5042, 2014.

L. Kalich-philosoph, H. Roness, and A. Carmely, Cyclophosphamide triggers follicle activation and "burnout" AS101 prevents follicle loss and preserves fertility, Sci. Transl. Med, vol.5, issue.185, pp.185-62, 2013.

H. Kaya, R. Desdicioglu, and M. Sezik, Does sphingosine-1-phosphate have a protective effect on cyclophosphamide-and irradiationinduced ovarian damage in the rat model?, Fertil. Steril, vol.89, issue.3, pp.732-735, 2008.

M. B. Zelinski, M. K. Murphy, and M. S. Lawson, In vivo delivery of FTY720 prevents radiation-induced ovarian failure and infertility in adult female nonhuman primates, Fertil. Steril, vol.95, issue.4, pp.1440-1445, 2011.

S. Gonfloni, D. Tella, L. Caldarola, and S. , Inhibition of the c-Abl-TAp63 pathway protects mouse oocytes from chemotherapyinduced death, Nat. Med, vol.15, issue.10, pp.1179-1185, 2009.

M. E. Ochalski, J. J. Shuttleworth, T. Chu, and K. E. Orwig, Thalidomide treatment attenuates chemotherapy-induced gonadal toxicity, Fertil. Steril, vol.95, issue.2, pp.819-822, 2011.

A. Y. Ting and B. K. Petroff, Tamoxifen decreases ovarian follicular loss from experimental toxicant DMBA and chemotherapy agents cyclophosphamide and doxorubicin in the rat

, J. Assist. Reprod. Genet, vol.27, issue.11, pp.591-597, 2010.

Y. F. Mahran, E. El-demerdash, A. S. Nada, and A. A. Ali, Abdel-Naim AB. Insights into the protective mechanisms of tamoxifen in radiotherapy-induced ovarian follicular loss: impact on insulin-like growth factor 1, Endocrinology, vol.154, issue.10, pp.3888-3899, 2013.

R. W. Ahn, S. L. Barrett, and M. R. Raja, Nano-encapsulation of arsenic trioxide enhances efficacy against murine lymphoma model while minimizing its impact on ovarian reserve in vitro and in vivo, PLoS ONE, vol.8, issue.3, p.58491, 2013.

A. Linkeviciute, G. Boniolo, L. Chiavari, and F. A. Peccatori, Fertility preservation in cancer patients: the global framework, Cancer Treat. Rev, vol.40, issue.8, pp.1019-1027, 2014.

S. J. Lee, L. R. Schover, and A. H. Partridge, American Society of Clinical Oncology recommendations on fertility preservation in cancer patients, J. Clin. Oncol, issue.18, pp.2917-2931, 2006.

, Epub ahead of print), Future Oncol

K. Fiedler and D. Ezcurra, Predicting and preventing ovarian hyperstimulation syndrome (OHSS): the need for individualized not standardized treatment, Reprod Biol Endocrinol RBE, vol.10, p.32, 2012.

C. Boothroyd, S. Karia, N. Andreadis, L. Rombauts, N. Johnson et al., Consensus statement on prevention and detection of ovarian hyperstimulation syndrome, Aust N Z J Obstet Gynaecol, vol.55, pp.523-557, 2015.

N. Panagiotopoulou, H. Byers, W. G. Newman, and K. Bhatia, Spontaneous ovarian hyperstimulation syndrome: case report, pathophysiological classification and diagnostic algorithm, Eur J Obstet Gynecol Reprod Biol, vol.169, pp.143-151, 2013.

A. D. Miras, J. T. Mogford, J. Wright, N. N. Mendoza, P. Xekouki et al., Ovarian hyperstimulation from ectopic hypersecretion of follicle stimulating hormone, Lancet Lond Engl, vol.385, p.392, 2015.

G. Smits, O. Olatunbosun, A. Delbaere, R. Pierson, G. Vassart et al., Ovarian hyperstimulation syndrome due to a mutation in the follicle-stimulating hormone receptor, N Engl J Med, vol.349, pp.760-766, 2003.

A. De-leener, G. Caltabiano, S. Erkan, M. Idil, G. Vassart et al., Identification of the first germline mutation in the extracellular domain of the follitropin receptor responsible for spontaneous ovarian hyperstimulation syndrome, Hum Mutat, vol.29, pp.91-99, 2008.

A. De-leener, L. Montanelli, J. Van-durme, C. H. Smits, G. Vassart et al., Presence and absence of follicle-stimulating hormone receptor mutations provide some insights into spontaneous ovarian hyperstimulation syndrome physiopathology, J Clin Endocrinol Metab, vol.91, pp.555-62, 2006.

C. Vasseur, P. Rodien, I. Beau, A. Desroches, C. Gérard et al., A chorionic gonadotropin-sensitive mutation in the follicle-stimulating hormone

, Engl J Med, vol.349, pp.753-762, 2003.

L. Montanelli, A. Delbaere, D. Carlo, C. Nappi, C. Smits et al., A mutation in the follicle-stimulating hormone receptor as a cause of familial spontaneous ovarian hyperstimulation syndrome, J Clin Endocrinol Metab, vol.89, pp.1255-1263, 2004.

S. S. Desai, B. S. Roy, and S. D. Mahale, Mutations and polymorphisms in FSH receptor: functional implications in human reproduction, Reprod Camb Engl, vol.146, pp.235-248, 2013.

M. S. Bramble, E. H. Goldstein, A. Lipson, T. Ngun, A. Eskin et al., A novel follicle-stimulating hormone receptor mutation causing primary ovarian failure: a fertility application of whole exome sequencing, Hum Reprod Oxf Engl, vol.31, pp.905-919, 2016.

B. Vannier, H. Loosfelt, G. Meduri, C. Pichon, and E. Milgrom, Anti-human FSH receptor monoclonal antibodies: immunochemical and immunocytochemical characterization of the receptor, Biochemistry (Mosc), vol.35, pp.1358-66, 1996.

I. Beau, P. Touraine, G. Meduri, A. Gougeon, A. Desroches et al., A novel phenotype related to partial loss of function mutations of the follicle stimulating hormone receptor, J Clin Invest, vol.102, pp.1352-1361, 1998.

I. Beau, M. T. Groyer-picard, L. Bivic, A. Vannier, B. Loosfelt et al., The basolateral localization signal of the follicle-stimulating hormone receptor, J Biol Chem, vol.273, pp.18610-18616, 1998.

E. T. Siegel, H. Kim, H. K. Nishimoto, and L. C. Layman, The molecular basis of impaired follicle-stimulating hormone action: evidence from human mutations and mouse models, Reprod Sci Thousand Oaks Calif, vol.20, pp.211-244, 2013.

D. Carlo, C. Savoia, F. Fabozzi, A. Gargano, V. Nappi et al., A case of ovarian torsion in a patient carrier of a FSH receptor gene mutation previously affected by spontaneous ovarian hyperstimulation syndrome, Gynecol Endocrinol Off J Int Soc Gynecol Endocrinol, vol.31, pp.105-113, 2015.

C. Timossi, C. Ortiz-elizondo, D. B. Pineda, J. A. Dias, P. M. Conn et al., Functional significance of the BBXXB motif reversed present in the cytoplasmic domains of the human follicle-stimulating hormone receptor, Mol Cell Endocrinol, vol.223, pp.17-26, 2004.

A. Ulloa-aguirre, A. Uribe, T. Zariñán, I. Bustos-jaimes, M. A. Pérez-solis et al., Role of the intracellular domains of the human FSH receptor in G(alphaS) protein coupling and receptor expression, Mol Cell Endocrinol, pp.153-62, 2007.

A. A. Banerjee and S. D. Mahale, Role of the Extracellular and Intracellular Loops of Follicle-Stimulating Hormone Receptor in Its Function, Front Endocrinol, vol.6, p.110, 2015.

M. T. Duvernay, F. Zhou, and G. Wu, A conserved motif for the transport of G proteincoupled receptors from the endoplasmic reticulum to the cell surface, J Biol Chem, vol.279, pp.30741-50, 2004.

T. Zariñán, M. A. Perez-solís, M. , G. Casas-gonzález, P. Conn et al., Dominant negative effects of human follicle-stimulating hormone receptor expression-deficient mutants on wild-type receptor cell surface expression. Rescue of oligomerization-dependent defective receptor expression by using cognate decoys, Mol Cell Endocrinol, vol.321, pp.112-134, 2010.

S. Uchida, H. Uchida, T. Maruyama, T. Kajitani, H. Oda et al., Molecular analysis of a mutated FSH receptor detected in a patient with spontaneous ovarian hyperstimulation syndrome, PLoS One, vol.8, p.75478, 2013.