Skip to main content Accessibility help
×
Hostname: page-component-76fb5796d-45l2p Total loading time: 0 Render date: 2024-04-29T05:14:28.421Z Has data issue: false hasContentIssue false

Section 2 - Assisted Reproductive Procedures

Published online by Cambridge University Press:  05 March 2021

Eliezer Girsh
Affiliation:
Barzilai Medical Center, Ashkelon
Get access

Summary

Image of the first page of this content. For PDF version, please use the ‘Save PDF’ preceeding this image.'
Type
Chapter
Information
Publisher: Cambridge University Press
Print publication year: 2021

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

References

Pandya, G, Cohen, MR. The effect of cis-isomer of clomiphene citrate (cis-clomiphene) on cervical mucus and vaginal cytology. J. Reprod. Med. 1972; 8:133138.Google Scholar
Kerin, JF, Liu, JH, Phillipou, G, Yen, SS. Evidence for a hypothalamic site of action of clomiphene citrate in women. J. Clin. Endocrinol. Metab. 1985; 61:265268.CrossRefGoogle ScholarPubMed
Kelly, JL, Adashi, EY. Ovulation induction. Obstet. Gynecol. Clin. North Am. 1987; 14:831864.Google Scholar
Palomba, S. Aromatase inhibitors for ovulation induction. J. Clin. Endocrinol. Metab. 2015; 100:17421747.Google Scholar
Deary, AJ, Seaton, JE, Prentice, A, et al. Single versus double insemination: a retrospective audit of pregnancy rates with two treatment protocols in donor insemination. Hum. Reprod. 1997; 12:14941496.CrossRefGoogle ScholarPubMed
van Rijswijk, J, Caanen, MR, Mijatovic, V, et al. Immobilization or mobilization after IUI: an RCT. Hum. Reprod. 2017; 32:22182224.Google Scholar
Kim, A, Young Lee, J, Il Ji, Y, et al. Do endometrial movements affect the achievement of pregnancy during intrauterine insemination? Int. J. Fertil. Steril. 2015; 8:339408.Google Scholar
Akino, N, Isono, W, Wada-Hiraike, O. Predicting suitable timing for artificial reproductive technology treatment in aged infertile women. Reprod. Med. Biol. 2016; 15:253259.Google Scholar
Cantineau, AE, Heineman, MJ, Cohlen, BJ. Single versus double intrauterine insemination in stimulated cycles for subfertile couples: a systematic review based on a Cochrane review. Hum. Reprod. 2003; 18:941946.Google Scholar
Abou-Setta, AM, Mansour, RT, Al-Inany, HG, et al. Intrauterine insemination catheters for assisted reproduction: a systematic review and meta-analysis. Hum. Reprod. 2006; 21:19611967.Google Scholar
Guzick, DS, Carson, SA, Coutifaris, C, et al. Efficacy of superovulation and intrauterine insemination in the treatment of infertility. National Cooperative Reproductive Medicine Network. N. Engl. J. Med. 1999; 340:177183.Google Scholar
Collins, J. Stimulated intra-uterine insemination is not a natural choice for the treatment of unexplained subfertility. Current best evidence for the advanced treatment of unexplained subfertility. Hum. Reprod. 2003; 18:907912.Google Scholar
Cèdrin-Durnerin, I, Bständig, B, Parneix, I, et al. Effects of oral contraceptive, synthetic progestogen or natural estrogen pre-treatments on the hormonal profile and the antral follicle cohort before GnRH antagonist protocol. Hum. Reprod. 2007; 22:109116.CrossRefGoogle ScholarPubMed
La Marca, A, Sunkara, SK. Individualization of controlled ovarian stimulation in IVF using ovarian reserve markers: from theory to practice. Hum. Reprod. Update 2014; 20:124140.CrossRefGoogle ScholarPubMed
Moraloğlu, Ö, Tonguc, EA, Özel, M, et al. The effects of peak and mid-luteal estradiol levels on in vitro fertilization outcome. Arch. Gynecol. Obstet. 2012; 285:857862.Google Scholar
Andersen, CY, Westergaard, LG, Sinosich, MJ, Byskov, AG. Human preovulatory follicular fluid: inhibin and free steroids related to optimal follicular maturation in ovarian stimulation regimes and possible function in ovulation. Hum. Reprod. 1992; 7:765769.Google Scholar
Ventura-Juncá, P, Irarrázaval, I, Rolle, AJ, et al. In vitro fertilization (IVF) in mammals: epigenetic and developmental alterations. Scientific and bioethical implications for IVF in humans. Biol. Res. 2015; 48:68.Google Scholar
Jiang, Z, Wang, Y, Lin, J, et al. Genetic and epigenetic risks of assisted reproduction. Best Pract. Res. Clin. Obstet. Gynaecol. 2017; 44:90104.CrossRefGoogle ScholarPubMed
Macklon, NS, Stouffer, RL, Giudice, LC, Fauser, BC. The science behind 25 years of ovarian stimulation for in vitro fertilization. Endocr. Rev. 2006; 27:170207.Google Scholar
Gonen, Y, Balakier, H, Powell, W, Casper, RF. Use of gonadotropin-releasing hormone agonist to trigger follicular maturation for in vitro fertilization. J. Clin. Endocrinol. Metab. 1990; 71:918922.Google Scholar
Albano, C, Smitz, J, Camus, M, et al. Comparison of different doses of gonadotropin-releasing hormone antagonist cetrorelix during controlled ovarian hyperstimulation. Fertil. Steril. 1997; 67:917922.Google Scholar
Tarlatzis, B, Fauser, B, Kilibianakis, E, Diedrich, K, Devroey, P. GnRH antagonists in ovarian stimulation for IVF. Hum. Reprod. Update 2006;12:333340.Google Scholar
Lyttle Schumacher, MB, Mersereau, JE, Steiner, AZ. Cycle day, estrogen level, and lead follicle size: analysis of 27,790 in vitro fertilization cycles to determine optimal start criteria for gonadotropin-releasing hormone antagonist. Fertil. Steril. 2018; 109:633637.Google Scholar
Blunt, SM, Butt, WR. Pulsatile GnRH therapy for the induction of ovulation in hypogonadotropic hypogonadism. Acta Endocrinol. Suppl. (Copenh.) 1988; 288:5865.Google Scholar
Nargund, G, Hutchison, L, Scaramuzzi, R, Campbell, S. Low-dose HCG is useful in preventing OHSS in high-risk women without adversely affecting the outcome of IVF cycles. RBM Online 2007; 14:682685.Google Scholar
Wang, W, Zhang, X-H, Wang, W-H, et al. The time interval between hCG priming and oocyte retrieval in ART program: a meta-analysis. J. Assist. Reprod. Genet. 2011; 28:901910.Google Scholar
Bosch, E, Labarta, E, Kolibianakis, E, Roen, M, Meldrum, D. Regimen of ovarian stimulation affects oocyte quality and therefore embryo quality. Fertil. Steril. 2016; 105:560570.Google Scholar
Itskovitz-Eldor, J, Kol, S, Mannaerts, B. Use of a single bolus of GnRH agonist triptorelin to trigger ovulation after GnRH antagonist ganirelix treatment in women undergoing ovarian stimulation for assisted reproduction, with special reference to the prevention of ovarian hyperstimulation syndrome: preliminary report: short communication. Hum. Reprod. 2000; 15:19651968.Google Scholar
Kolibianakis, EM, Schultze-Mosgau, A, Schroer, A, et al. A lower ongoing pregnancy rate can be expected when GnRH agonist is used for triggering final oocyte maturation instead of HCG in patients undergoing IVF with GnRH antagonists. Hum. Reprod. 2005; 20:28872892.Google Scholar
Choi, JH, Gilks, CB, Auersperg, N, Leung, PC. Immunolocalization of gonadotropin-releasing hormone (GnRH)-I, GnRH-II, and type I GnRH receptor during follicular development in the human ovary. J. Clin. Endocrinol. Metab. 2006; 91:45624570.CrossRefGoogle ScholarPubMed
Laprise, SL. Implications of epigenetics and genomic imprinting in assisted reproductive technologies. Mol. Reprod. Dev. 2009; 76:10061018.Google Scholar
Rubio, C, Mercader, A, Alamá, P, et al. Prospective cohort study in high responder oocyte donors using two hormonal stimulation protocols: impact on embryo aneuploidy and development. Hum. Reprod. 2010; 25:22902297.Google Scholar
van der Gaast, MH, Eijkemans, MJ, van der Net, JB, et al. Optimum number of oocytes for a successful first IVF treatment cycle. RBM Online 2006; 13:476480.Google Scholar
Steward, RG, Lan, L, Shah, AA, et al. Oocyte number as a predictor for ovarian hyperstimulation syndrome and live birth: an analysis of 256,381 in vitro fertilization cycles. Fertil. Steril. 2014; 101:967973.Google Scholar
Vermey, BG, Chua, SJ, Zafarmand, MH, et al. Is there an association between oocyte number and embryo quality? A systematic review and meta-analysis. RBM Online 2019; 39:751763.Google Scholar
Haaf, T, Lambrecht, A, Grossmann, B, et al. A high oocyte yield for intracytoplasmic sperm injection treatment is associated with an increased chromosome error rate. Fertil. Steril. 2009; 91:733738.Google Scholar
ESHRE Capri Workshop Group. Health and fertility in World Health Organization group 2 anovulatory women. Hum. Reprod. Update 2012; 18:586599.CrossRefGoogle Scholar
Homburg, R, Ray, A, Bhide, P, et al. The relationship of serum anti-Mullerian hormone with polycystic ovarian morphology and polycystic ovary syndrome: a prospective cohort study. Hum. Reprod. 2013; 28:10771083.Google Scholar
Dumesic, DA, Padmanabhan, V, Abbott, DH. Polycystic ovary syndrome and oocyte developmental competence. Obstet. Gynecol. Surv. 2008; 63:3948.CrossRefGoogle ScholarPubMed
Wood, JR, Dumesic, DA, Abbott, DH, Strauss, JF 3rd. Molecular abnormalities in oocytes from women with polycystic ovary syndrome revealed by microarray analysis. J. Clin. Endocrinol. Metab. 2007; 92:705713.CrossRefGoogle ScholarPubMed
Amato, G, Conte, M, Mazziotti, G, et al. Serum and follicular fluid cytokines in polycystic ovary syndrome during stimulated cycles. Obstet. Gynecol. 2003; 101:11771182.Google ScholarPubMed
Gallinelli, A, Ciaccio, I, Giannella, L, et al. Correlations between concentrations of interleukin-12 and interleukin-13 and lymphocyte subsets in the follicular fluid of women with and without polycystic ovary syndrome. Fertil. Steril. 2003; 79:13651372.CrossRefGoogle ScholarPubMed
Wu, CH, Winkel, CA. The effect of therapy initiation day on clomiphene citrate therapy. Fertil. Steril. 1989; 52:564568.Google Scholar
Dickey, RP, Taylor, SN, Lu, PY, et al. Effect of diagnosis, age, sperm quality, and number of preovulatory follicles on the outcome of multiple cycles of clomiphene citrate-intrauterine insemination. Fertil. Steril. 2002; 78:10881095.Google Scholar
Veltman-Verhulst, SM, Fauser, BC, Eijkemans, MJ. High singleton live birth rate confirmed after ovulation induction in women with anovulatory polycystic ovary syndrome: validation of a prediction model for clinical practice. Fertil. Steril. 2012; 98:761768.CrossRefGoogle ScholarPubMed
Mathur, R, Alexander, CJ, Yano, J, Trivax, B, Azziz, R. Use of metformin in polycystic ovary syndrome. Am. J. Obstet. Gynecol. 2008; 199:596609.Google Scholar
Sinawat, S, Buppasiri, P, Lumbiganon, P, Pattanittum, P. Long versus short course treatment with metformin and clomiphene citrate for ovulation induction in women with PCOS. Cochrane Database Syst. Rev. 2012; 10:CD006226. doi:10.1002/14651858.CD006226.pub3.Google Scholar
Gleicher, N, Barad, DH. Dehydroepiandrosterone (DHEA) supplementation in diminished ovarian reserve (DOR). Reprod. Biol. Endocrinol. 2011; 9:67, doi:10.1186/1477-7827-9-67.CrossRefGoogle ScholarPubMed
Narkwichean, A, Maalouf, W, Campbell, BK, Jayaprakasan, K. Efficacy of dehydroepiandrosterone to improve ovarian response in women with diminished ovarian reserve: a meta-analysis. Reprod. Biol. Endocrinol. 2013; 11:44, doi:10.1186/1477-7827-11-44.Google Scholar
Yeung, T, Chai, J, Li, R, et al. A double-blind randomised trial on the effect of dehydroepiandrosterone on ovarian reserve markers, ovarian response and number of oocyte in anticipated normal ovarian responders. BJOG 2016; 123:10971105.Google Scholar
Gómez, R, Soares, SR, Busso, C, et al. Physiology and pathology of ovarian hyperstimulation syndrome. Sem. Reprod. Med. 2010; 28:448457.Google Scholar
Kasum, M, Danolic, D, Oreskovic, S, et al. Thrombosis following ovarian hyperstimulation syndrome. Gynecol. Endocrinol. 2014; 30:764768.CrossRefGoogle ScholarPubMed
Practice Committee of the American Society for Reproductive Medicine. Prevention and treatment of moderate and severe ovarian hyperstimulation syndrome. Fertil. Steril. 2016; 106:16341647.Google Scholar

References

Jabbour, HN, Kelly, RW, Fraser, HM, Critchley, HO. Endocrine regulation of menstruation. Endocr. Rev. 2006; 27:1746.Google Scholar
Garcia, J, Jones, GS, Acosta, AA, Wright, GL. Corpus luteum fuction after follicle aspiration for oocyte retrieval. Fertil. Steril. 1981; 36:565572.Google Scholar
Fatemi, HM, Popovic-Todorovic, B, Papanikolaou, E, Donoso, P, Devroey, P. An update of luteal phase support in stimulated IVF cycles. Hum. Reprod. Update 2007; 13:581590.Google Scholar
O’Neill, C, Ferrier, AJ, Vaughan, J, Sinosich, MJ, Saunders, DM. Causes of implantation failure after in-vitro fertilization and embryo transfer. Lancet 1985; 2:615.Google Scholar
The ESHRE Capri Workshop Group. Anovulatory infertility. Hum. Reprod. 1995; 10:15491553.CrossRefGoogle Scholar
Duncan, WC. A guide to understanding polycystic ovary syndrome (PCOS). J. Fam. Plann. Reprod. Health Care 2014; 40:217225.Google Scholar
Mackens, S, Santos-Ribeiro, S, van de Vijver, A, et al. Frozen embryo transfer: a review on the optimal endometrial preparation and timing. Hum. Reprod. 2017; 32:22342242.Google Scholar
Kutlusoy, F, Guler, I, Erdem, M, et al. Luteal phase support with estrogen in addition to progesterone increases pregnancy rates in in-vitro fertilization cycles with poor response to gonadotropins. Gynecol. Endocrinol. 2014; 30:363366.Google Scholar
Ho, CH, Chen, SU, Peng, FS, Chang, CY, Yang, YS. Luteal support for IVF/ICSI cycles with Crinone 8% (90 mg) twice daily results in higher pregnancy rates than with intramuscular progesterone. J. Chin. Med. Assoc. 2008; 71:386391.Google Scholar
Barbosa, MW, Silva, LR, Navarro, PA, et al. Dydrogesterone vs progesterone for luteal-phase support: systematic review and meta-analysis of randomized controlled trials. Ultrasound Obstet. Gynecol. 2016; 48:161170.Google Scholar
Saccone, G, Khalifeh, A, Elimian, A, et al. Vaginal progesterone vs intramuscular 17α-hydroxyprogesterone caproate for prevention of recurrent spontaneous preterm birth in singleton gestations: systematic review and meta-analysis randomized controlled trials. Ultrasound Obstet. Gynecol. 2017; 49:315321.Google Scholar
Child, T, Leonard, SA, Evans, JS, Lass, A. Systematic review of the clinical efficacy of vaginal progesterone for luteal phase support in assisted reproductive technology cycles. RBM Online 2018; 36:630645.Google Scholar
Tournaye, H, Sukhikh, G, Kuhler, E, Griesinger, G. A phase III randomized controlled trial comparing the efficacy, safety and tolerability of oral dydrogesterone versus micronized vaginal progesterone for luteal support in in vitro fertilization. Hum. Reprod. 2017; 32:10191027.Google Scholar
Baker, V, Jones, C, Doody, K, et al. A randomized controlled trial comparing the efficacy and safety of aqueous subcutaneous progesterone with vaginal progesterone for luteal phase support of in vitro fertilization. Hum. Reprod. 2014; 29:22102220.CrossRefGoogle ScholarPubMed
Kleinstein, J. Efficacy and tolerability of vaginal progesterone capsules (Utrogest 200) compared with progesterone gel (Crinone 8%) for luteal phase support during assisted reproduction. Fertil. Steril. 2005; 83:16411649.Google Scholar
Penzias, A. Luteal phase support. Fertil. Steril. 2002; 77:318323.Google Scholar
van der Linden, M, Buckingham, K, Farquhar, C, Kremer, JA, Metwally, M. Luteal phase support for assisted reproduction cycles. Cochrane Database Syst. Rev. 2015; CD009154. doi:10.1002/14651858.CD009154.Google Scholar
Hutchison, JS, Zeleznik, AJ. The corpus luteum of the primate menstrual cycle is capable of recovering from a transient withdrawal of pituitary gonadotropin support. Endocrinology 1985; 117:10431049.Google Scholar
Liu, X, Mu, H, Shi, Q, Xiao, X, Qi, H. The optimal duration of progesterone supplementation in pregnant women after IVF/ICSI: a meta-analysis. Reprod. Biol. Endocrinol. 2012; 10:107115.Google Scholar
Thomsen, LH, Kesmodel, US, Erb, K, et al. The impact of luteal serum progesterone levels on live birth rates-a prospective study of 602 IVF/ICSI cycles. Hum. Reprod. 2018; 33:15061516. doi:10.1093/humrep/dey226.Google Scholar
Silver, RI. Endocrine abnormalities in boys with hypospadias. Adv. Exp. Med. Biol. 2004; 545:4572.CrossRefGoogle ScholarPubMed
Vaisbuch, E, de Ziegler, D, Leong, M, Weissman, A, Shoham, Z. Luteal-phase support in assisted reproduction treatment: real-life practices reported worldwide by an updated website-based survey. RBM Online 2014; 28:330335.Google Scholar

References

Lopata, A, Johnston, WIH, Leeton, J, et al. Collection of human oocytes by laparotomy and laparoscopy. Fertil. Steril. 1974; 25:10301038.Google Scholar
Wood, C, Leeton, J, Talbot, M, Trounson, AO. Technique for collecting mature human oocytes for in-vitro fertilization. Br. J. Obstet. Gynaecol. 1981; 88:756760.Google Scholar
Renou, P, Trounson, A, Wood, C, Leeton, JF. The collection of human oocytes for in-vitro fertilization. An instrument for maximizing oocyte recovery rate. Fertil. Steril. 1981; 35:409412.Google Scholar
Dellenbach, P, Nisand, I, Moreau, L, et al. Transvaginal, sonographically controlled ovarian follicle puncture for egg retrieval. Lancet 1984; 1:1467.Google Scholar
Dellenbach, P, Nisand, I, Moreau, L, et al. Transvaginal sonographically controlled ovarian follicle puncture for egg retrieval. Fertil. Steril. 1985; 44:656662.Google Scholar
Grimbizis, GF, Di Spiezio Sardo, A, Saravelos, SH, et al. The Thessaloniki ESHRE/ESGE consensus on diagnosis of female genital anomalies. Hum. Reprod. 2016; 31:27.Google Scholar
Stelling, JR, Chapman, ET, Frankfurter, D, et al. Subcutaneous versus intramuscular administration of human chorionic gonadotropin during an in vitro fertilization cycle. Fertil. Steril. 2003; 79:881885.Google Scholar
Weiss, A, Neril, R, Geslevich, J, et al. Lag time from ovulation trigger to oocyte aspiration and oocyte maturity in assisted reproductive technology cycles: a retrospective study. Fertil. Steril. 2014; 102:419423.Google Scholar
Matorras, R, Aparicio, V, Corcostegui, B, et al. Failure of intrauterine insemination as rescue treatment in low responders with adequate HCG timing with no oocytes retrieved. RBM Online 2014; 29:634639.Google Scholar
Hershlag, A, Feng, HL, Scholl, GS. Betadine (povidone-iodine) is toxic to murine embryogenesis. Fertil. Steril. 2003; 79:12491250.Google Scholar
Jeffcoate, N. Infertility and assisted reproductive technologies. In: Kumar, P, Malhotra, N, eds., Jeffcoate’s Principles of Gynaecology. New Delhi: Jaypee. 2008; 721723.Google Scholar
Bennett, SJ, Waterstone, JJ, Cheng, WC, Parsons, J. Complications of transvaginal ultrasound-directed follicle aspiration: a review of 2670 consecutive procedures. J. Assist. Reprod. Genet. 1993; 10:7277.Google Scholar
Mansour, RT, Aboulghar, MA, Serour, GI. Study of the optimum time for human chorionic gonadotropin-ovum pickup interval in in vitro fertilization. J. Assist. Reprod. Genet. 1994; 11:478481.Google Scholar
Nargund, G, Reid, F, Parsons, J. Human chorionic gonadotropin-to-oocyte collection interval in a superovulation IVF program. A prospective study. J. Assist. Reprod. Genet. 2001; 18:8790.Google Scholar
Raziel, A, Schachter, M, Strassburger, D, et al. In vivo maturation of oocytes by extending the interval between human chorionic gonadotropin administration and oocyte retrieval. Fertil. Steril. 2006; 86:583587.Google Scholar
Jamieson, ME, Fleming, R, Kader, S, et al. In vivo and in vitro maturation of human oocytes: effects on embryo development and polyspermic fertilization. Fertil. Steril. 1991; 56:9397.Google Scholar
Garor, R, Shufaro, Y, Kotler, N, et al. Prolonging oocyte in vitro culture and handling time does not compensate for a shorter interval from human chorionic gonadotropin administration to oocyte pickup. Fertil. Steril. 2015; 103:7275.Google Scholar
Fisch, B, Kaplan-Kraicer, R, Amit, S, Ovadia, J, Tadir, Y. The effect of preinsemination interval upon fertilization of human oocytes in vitro. Hum. Reprod. 1989; 4:495496.Google Scholar
Bokal, EV, Vrtovec, HM, Virant Klun, I, Verdenik, I. Prolonged HCG action affects angiogenic substances and improves follicular maturation, oocyte quality and fertilization competence in patients with polycystic ovarian syndrome. Hum. Reprod. 2005; 20:15621568.Google Scholar
Fauque, P, Guibert, J, Jouannet, P, Patrat, C. Successful delivery after the transfer of embryos obtained from a cohort of incompletely in vivo matured oocytes at retrieval time. Fertil. Steril. 2008; 89:991.e1–991.e4.Google Scholar
Van de Velde, H, De Vos, A, Joris, H, Nagy, ZP, Van Steirteghem, AC. Effect of timing of oocyte denudation and micro-injection on survival, fertilization and embryo quality after intracytoplasmic sperm injection. Hum. Reprod. 1998; 13:31603164.Google Scholar
Dozortsev, D, Nagy, P, Abdelmassih, S, et al. The optimal time for intracytoplasmic sperm injection in the human is from 37 to 41 hours after administration of human chorionic gonadotropin. Fertil. Steril. 2004; 82:14921496.Google Scholar
Isiklar, A, Mercan, R, Balaban, B, et al. Impact of oocyte pre-incubation time on fertilization, embryo quality and pregnancy rate after intracytoplasmic sperm injection. RBM Online 2004; 8:682686.Google Scholar
Stevenson, T, Lashen, H. Empty follicle syndrome: the reality of a controversial syndrome, a systematic review. Fertil. Steril. 2008; 90:691698.CrossRefGoogle ScholarPubMed
Awonuga, A, Govindbhai, J, Zierke, S, Schnauffer, K. Continuing the debate on empty follicle syndrome: can it be associated with normal bioavailability of β-human chorionic gonadotrophin on the day of oocyte recovery? Hum. Reprod. 1998; 13:12811284.Google Scholar
van Heusden, AM, van Santbrink, EJ, de Jong, D. The empty follicle syndrome is dead. Fertil. Steril. 2008; 89:746.CrossRefGoogle ScholarPubMed
Bustillo, M. Unsuccessful oocyte retrieval: technical artifact or genuine empty follicle syndrome? RBM Online 2004; 8:5967.Google Scholar
Tsuiki, A, Rose, BI, Hung, TT. Steroid profiles of follicular fluids from a patient with the empty follicle syndrome. Fertil. Steril. 1988; 49:104107.Google Scholar
Eppig, JJ. Oocyte control of ovarian follicular development and function in mammals. Reproduction 2001; 122:829838.Google Scholar
Feuerstein, P, Cadoret, V, Dalbies-Tran, R, et al. Gene expression in human cumulus cells: one approach to oocyte competence. Hum. Reprod. 2007; 22:30693077.Google Scholar
Hamel, M, Dufort, I, Robert, C, et al. Identification of differentially expressed markers in human follicular cells associated with competent oocytes. Hum. Reprod. 2008; 23:11181127.CrossRefGoogle ScholarPubMed
Adriaenssens, T, Wathlet, S, Segers, I, et al. Cumulus cell gene expression is associated with oocyte developmental quality and influenced by patient and treatment characteristics. Hum. Reprod. 2010; 25:12591270.Google Scholar
Assidi, M, Montag, M, van der Ven, K, Sirard, MA. Biomarkers of human oocyte developmental competence expressed in cumulus cells before ICSI: a preliminary study. J. Assist. Reprod. Genet. 2011; 28:173188.Google Scholar
Granot, I, Dekel, N. Cell-to-cell communication in the ovarian follicle: developmental and hormonal regulation of the expression of connexin-43. Hum. Reprod. 1998; 13:8597.Google Scholar
Onalan, G, Pabuccu, R, Onalan, R, Ceylaner, S, Selam, B. Empty follicle syndrome in two sisters with three cycles: case report. Hum. Reprod. 2003; 18:18641867.Google Scholar
Yariz, KO, Walsh, T, Uzak, A, et al. Inherited mutation of the luteinizing hormone/choriogonadotropin receptor (LHCGR) in empty follicle syndrome. Fertil. Steril. 2011; 96:E125E130.Google Scholar
Girsh, E, Makovski Lev-Tov, E, Umansky, N, et al. Empty follicle syndrome-oocyte could be retrieved in consecutive cycle. JFIV Reprod. Med. Genet. 2016; 4:4. doi:10.4172/2375-4508.1000193.Google Scholar
Dai, C, Chen, Y, Hu, L, et al. ZP1 mutations are associated with empty follicle syndrome: evidence for the existence of an intact oocyte and a zona pellucida in follicles up to the early antral stage. A case report. Hum. Reprod. 2019; 34:22012207. doi:10.1093/humrep/dez174.Google Scholar

References

Bjorndahl, L, Mortimer, D, Barratt, CLR, et al. Sperm preparation. In: A Practical Guide to Basic Laboratory Andrology, 1st ed. New York: Cambridge University Press. 2010; 167187.Google Scholar
Levitas, E, Lunenfeld, E, Weiss, N, et al. Relationship between the duration of sexual abstinence and semen quality: analysis of 9,489 semen samples. Fertil. Steril. 2005; 83:16801683.Google Scholar
Pons, I, Cercas, R, Villas, C, Braña, C, Fernández-Shaw, S. One abstinence day decreases sperm DNA fragmentation in 90 % of selected patients J. Assist. Reprod. Genet. 2013; 30:12111218.Google Scholar
Bahadur, G, Almossawia, O, Zeirideen Zaid, R, et al. Semen characteristics in consecutive ejaculates with short abstinence in subfertile males. RBM Online 2016; 32:323328.Google Scholar
Henkel, RR, Schill, WB. Sperm preparation for ART. Reprod. Biol. Endocrinol. 2003; 108:122.Google Scholar
Makker, K, Agarwal, A, Sharma, R. Oxidative stress and male infertility. Indian J. Med. Res. 2009; 129:357367.Google Scholar
Oshio, S, Kaneko, S, Iizuka, R, Mohri, H. Effects of gradient centrifugation on human sperm. Arch. Androl. 1987; 19:8593.Google Scholar
Bourne, H, Edgar, DH, Baker, HWG. Sperm preparation techniques. In: Gardner, DK, Weissman, A, Howles, CM, Shoham, Z, eds., Textbook of Assisted Reproductive Techniques: Laboratory and Clinical Perspectives, 2nd ed. USA: Informa Healthcare. 2004; 7991.Google Scholar
Sakkas, D, Manicardi, GC, Tomlinson, M, et al. The use of two density gradient centrifugation techniques and the swim-up method to separate spermatozoa with chromatin and nuclear DNA anomalies. Hum. Reprod. 2000; 15:11121116.Google Scholar
Allamaneni, SS, Agarwal, A, Rama, S, Ranganathan, P, Sharma, RK. Comparative study on density gradients and swim-up preparation techniques utilizing neat and cryopreserved spermatozoa. Asian J. Androl. 2005; 7:8692.Google Scholar
May-Panloup, P, Chrétien, MF, Savagner, F, et al. Increased sperm mitochondrial DNA content in male infertility. Hum. Reprod. 2003; 18:550556.Google Scholar
Jackson, RE, Bormann, CL, Hassun, PA, et al. Effects of semen storage and separation techniques on sperm DNA fragmentation. Fertil. Steril. 2010; 94:26262630.Google Scholar
Menkveld, R, Stander, FS, Kotze, TJ, Kruger, TF, van Zyl, JA. The evaluation of morphological characteristics of human spermatozoa according to stricter criteria. Hum. Reprod. 1990; 5:586592.CrossRefGoogle ScholarPubMed
Bonde, JP, Ernst, E, Jensen, TK, et al. Relation between semen quality and fertility: a population-based study of 430 first-pregnancy planners. Lancet 1998; 352:11721177.Google Scholar
Kruger, TF, Menkveld, R, Stander, FS, et al. Sperm morphologic features as a prognostic factor in in vitro fertilization. Fertil. Steril. 1986; 46:11181123.Google Scholar
Jacobs, M, Stolwijk, AM, Wetzels, AM. The effect of insemination/injection time on the results of IVF and ICSI. Hum. Reprod. 2001; 16:17081713.Google Scholar
Dozortsev, D, Nagy, P, Abdelmassih, S, et al. The optimal time for intracytoplasmic sperm injection in the human is from 37 to 41 hours after administration of human chorionic gonadotropin. Fertil. Steril. 2004; 82:14921496.Google Scholar
Isiklar, A, Mercan, R, Balaban, B, et al. Impact of oocyte pre-incubation time on fertilization, embryo quality and pregnancy rate after intracytoplasmic sperm injection. RBM Online 2004; 8:682686.Google Scholar
Garor, R, Shufaro, Y, Kotler, N, et al. Prolonging oocyte in vitro culture and handling time does not compensate for a shorter interval from human chorionic gonadotropin administration to oocyte pickup. Fertil. Steril. 2015; 103:7275.Google Scholar
Naji, O, Moska, N, Dajani, Y, et al. Early oocyte denudation does not compromise ICSI cycle outcome: a large retrospective cohort study. RBM Online 2018; 37:1824.Google Scholar
de Moura, BR, Gurgel, MC, Machado, SP, et al. Low concentration of hyaluronidase for oocyte denudation can improve fertilization rates and embryo quality. JBRA Assist. Reprod. 2017; 21: 2730.Google Scholar
Rienzi, L, Ubaldi, F. Oocyte retrieval and selection. In: Gardner, DK, Weissman, A, Howles, CM, Shoham, Z, eds., Textbook of Assisted Reproductive Technologies: Laboratory and Clinical Perspectives, 3rd ed. London: Informa Healthcare, 2009; 5101.Google Scholar
Rienzi, L, Vajta, G, Ubaldi, F. Predictive value of oocyte morphology in human IVF: a systematic review of the literature. Hum. Reprod. Update 2011; 17:3445.Google Scholar
Lazzaroni-Tealdi, E, Barad, DH, Yu, Y, et al. Oocyte scoring system with better predictability of clinical IVF pregnancies than currently practiced embryo quality assessment. PLoS One 2015; 10: e0143632.CrossRefGoogle Scholar
Kilani, S, Cooke, S, Chapman, M. Time course of meiotic spindle development in MII oocytes. Zygote 2011; 19:5562.Google Scholar
Alvarez, C, García-Garrido, C, Taronger, R, González de Merlo, G. In vitro maturation, fertilization, embryo development & clinical outcome of human metaphase-I oocytes retrieved from stimulated intracytoplasmic sperm injection cycles. Indian J. Med. Res. 2013; 137:331338.Google Scholar
Fauque, P, Guibert, J, Jouannet, P, Patrat, C. Successful delivery after the transfer of embryos obtained from a cohort of incompletely in vivo matured oocytes at retrieval time. Fertil. Steril. 2008; 89:991.e1–991.e4.Google Scholar
Hyun, C-S, Cha, J-H, Son, W-Y, et al. Optimal ICSI timing after the first polar body extrusion in in vitro matured human oocytes. Hum. Reprod. 2007; 22:19911995.Google Scholar
Ebner, T, Moser, M, Sommergruber, M, Shebl, O, Tews, G. Incomplete denudation of oocytes prior to ICSI enhances embryo quality and blastocyst development. Hum. Reprod. 2006; 21:29722977.Google Scholar
Parikh, FR, Nadkarni, SG, Naik, NJ, Naik, DJ, Uttamchandani, SA. Cumulus coculture and cumulus-aided embryo transfer increases pregnancy rates in patients undergoing in vitro fertilization. Fertil. Steril. 2006; 86:839847.Google Scholar
Cohen, MR. Intrauterine insemination. Int. J. Fertil. 1962; 7:235240.Google Scholar
Sakhel, K, Schwarck, S, Ashraf, M, Abuzeid, M. Semen parameters as determinants of success in 1662 cycles of intrauterine insemination after controlled ovarian hyperstimulation. Fertil. Steril. 2005; 84:248249.Google Scholar
Papillon-Smith, J, Baker, SE, Agbo, C, Dahan, MH. Pregnancy rates with intrauterine insemination: comparing 1999 and 2010 World Health Organization semen analysis norms. RBM Online 2015; 30:392400.Google Scholar
Marshburn, PB, Alanis, M, Matthews, ML, et al. A short period of ejaculatory abstinence before intrauterine insemination is associated with higher pregnancy rates. Fertil. Steril. 2010; 93:286288.Google Scholar
Merviel, P, Heraud, MH, Grenier, N, et al. Predictive factors for pregnancy after intrauterine insemination (IUI): an analysis of 1038 cycles and a review of the literature. Fertil. Steril. 2010; 93:7988.CrossRefGoogle Scholar
Zhang, E, Tao, X, Xing, W, Cai, L, Zhang, B. Effect of sperm count on success of intrauterine insemination in couples diagnosed with male factor infertility. Mater Sociomed 2014; 26:321323.Google Scholar
Jurema, MW, Vieira, AD, Bankowski, B, et al. Effect of ejaculatory abstinence period on the pregnancy rate after intrauterine insemination. Fertil. Steril. 2005; 84:678681.Google Scholar
Speyer, BE, Abramov, B, Saab, W, et al. Factors influencing the outcome of intrauterine insemination (IUI): age, clinical variables and significant thresholds. J. Obstet. Gynaecol. 2013; 33:697700.Google Scholar
van Noord-Zaadstra, BM, Looman, CW, Alsbach, H, et al. Delaying childbearing: effect of age on fecundity and outcome of pregnancy. BMJ 1991; 302:13611365.CrossRefGoogle ScholarPubMed
Goldman, MB, Thornton, KL, Ryley, D, et al. A randomized clinical trial to determine optimal infertility treatment in older couples: the Forty and Over Treatment Trial (FORT-T). Fertil. Steril. 2014; 101:15741581.Google Scholar
Tomlinson, MJ, Amissah-Arthur, JB, Thompson, KA, Kasraie, JL, Bentick, B. Prognostic indicators for intrauterine insemination (IUI): statistical model for IUI success. Hum. Reprod. 1996; 11:18921896.Google Scholar
Honda, T, Tsutsumi, M, Komoda, F, Tatsumi, K. Acceptable pregnancy rate of unstimulated intrauterine insemination: a retrospective analysis of 17,830 cycles. Reprod. Med. Biol. 2015; 14: 2732.Google Scholar
Tonguc, E, Var, T, Onalan, G, et al. Comparison of the effectiveness of single versus double intrauterine insemination with three different timing regimens. Fertil. Steril. 2010; 94:12671270.Google Scholar
Khalifa, Y, Redgment, CJ, Tsirigotis, M, Grudzinskas, JG, Craft, IL. The value of single versus repeated insemination in intra-uterine donor insemination cycles. Hum. Reprod. 1995; 10:153154.Google Scholar
Trounson, AO. The choice of the most appropriate fertilization technique for human male factor infertility. Reprod. Fertil. Dev. 1994; 6:3743.Google Scholar
Fiorentino, A, Magli, MC, Fortini, D, et al. Sperm:oocyte ratios in an in vitro fertilization (IVF) program. J. Assist. Reprod. Genet. 1994; 2:97103.Google Scholar
Ron-el, R, Nachum, H, Herman, A, et al. Delayed fertilization and poor embryonic development associated with impaired semen quality. Fertil. Steril. 1991; 55:338344.Google Scholar
Gianaroli, L, Tosti, E, Magli, MC, et al. Fertilization current in the human oocyte. Mol. Reprod. Dev. 1994; 3:209214.Google Scholar
Bedford, JM, Kim, HH. Cumulus oophorus as a sperm sequestering device in vivo. J. Exp. Zool. 1993; 265:321328.Google Scholar
Gianaroli, L, Fiorentino, A, Magli, MC, Ferraretti, AP, Montanaro, N. Prolonged sperm-oocyte exposure and high sperm concentration affect human embryo viability and pregnancy rate. Hum. Reprod. 1996; 11:25072511.Google Scholar
Malter, HE, Cohen, J. Partial zona dissection of the human oocyte: a nontraumatic method using micromanipulation to assist zona pellucida penetration. Fertil. Steril. 1989; 51:139148.Google Scholar
Laws-King, A, Trounson, A, Sathananthan, AH, Kola, I. Fertilisation of human oocytes by micro-injection of a single spermatozoon under the zona pellucida. Fertil. Steril. 1987; 48:637642.Google Scholar
Ng, SC, Bongso, A, Ratnam, SS. Microinjection of human oocytes: a technique for severe oligoasthenoteratozoospermia. Fertil. Steril. 1991; 56:11171123.Google Scholar
Sakkas, D, Gianaroli, L, Diotallevi, L, et al. IVF treatment of moderate male factor infertility: a comparison of mini-Percoll, partial zona dissection and sub-zonal sperm insertion techniques. Hum. Reprod. 1993; 8:587591.Google Scholar
Levran, D, Bider, D, Yonesh, M, et al. A randomized study of intracytoplasmic sperm injection (ICSI) versus subzonal insemination (SUZI) for the management of severe male factor infertility. J. Assist. Reprod. Genet. 1995; 12:319321.Google Scholar
Palermo, G, Joris, H, Devroey, P, Van Steirteghem, AC. Pregnancies after intra-cytoplasmic injection of single spermatozoon into an oocyte. Lancet 1992; 340:1718.Google Scholar
Boulet, SL, Mehta, A, Kissin, DM, et al. Trends in use of and reproductive outcomes associated with intracytoplasmic sperm injection. JAMA 2015; 313:255263.Google Scholar
Wang, J, Sauer, MV. In vitro fertilization (IVF): a review of 3 decades of clinical innovation and technological advancement. Ther. Clin. Risk Manag. 2006; 2:355364.Google Scholar
Tsai, MY, Huang, FJ, Kung, FT, et al. Influence of polyvinylpyrrolidone on the outcome of intracytoplasmic sperm injection. J. Reprod. Med. 2000; 45:115120.Google Scholar
Dozortsev, D, Rybouchkin, A, De Sutter, P, Dhont, M. Sperm plasma membrane damage prior to intracytoplasmic sperm injection: a necessary condition for sperm nucleus decondensation. Hum. Reprod. 1995; 10:29602964.Google Scholar
Rienzi, L, Ubaldi, F, Martinez, F, et al. Relationship between meiotic spindle location with regard to the polar body position and oocyte developmental potential after ICSI. Hum. Reprod. 2003; 18:12891293.Google Scholar
Palermo, GD. ICSI: technical aspects. In: Gardner, DK, Weissman, A, Howles, CM, Shoham, Z, eds., Textbook of Assisted Reproductive Techniques: Laboratory and Clinical Perspectives. Boca Raton: CRC Press. 2001; 147157.Google Scholar
Sousa, M, Tesarik, J. Ultrastructural analysis of fertilization failure after intracytoplasmic sperm injection. Hum. Reprod. 1994; 9:23742380.Google Scholar
Flaherty, SP, Payne, D, Swann, NJ, Mattews, CD. Aetiology of failed and abnormal fertilization after intracytoplasmic sperm injection. Hum. Reprod. 1995; 10:26232629.Google Scholar
Ebner, T, Moser, M, Sommergruber, M. Jesacher, K, Tews, G. Complete oocyte activation failure after ICSI can be overcome by a modified injection technique. Hum. Reprod. 2004; 19:18371841.Google Scholar
Bosco, L, Ruvolo, G, Morici, G, et al. Apoptosis in human unfertilized oocytes after intracytoplasmic sperm injection. Fertil. Steril. 2005; 84:14171423.Google Scholar
Bárcena, P, Obradors, A, Vernaeve, V, Vassena, R. Should we worry about the clock? Relationship between time to ICSI and reproductive outcomes in cycles with fresh and vitrified oocytes. Hum. Reprod. 2016; 31:11821191.Google Scholar
Van de Velde, H, De Vos, A, Joris, H, Nagy, ZP, Van Steirteghem, AC. Effect of timing of oocyte denudation and micro-injection on survival, fertilization and embryo quality after intracytoplasmic sperm injection. Hum. Reprod. 1998; 13:31603164.Google Scholar
Reynier, P, May-Panloup, P, Chretien, MF, et al. Mitochondrial DNA content affects the fertilizability of human oocytes. Mol. Hum. Reprod. 2001; 7:425429.Google Scholar
Celik-Ozenci, C, Jakab, A, Kovacs, T, et al. Sperm selection for ICSI: shape properties do not predict the absence or presence of numerical chromosomal aberrations. Hum. Reprod. 2004; 19:20522059.Google Scholar
Viville, S, Mollard, R, Bach, ML, et al. Do morphological anomalies reflect chromosomal aneuploidies?: case report. Hum. Reprod. 2000; 15:25632566.Google Scholar
Zeyneloglu, HB, Baltaci, V, Duran, HE, Erdemli, E, Batioglu, S. Achievement of pregnancy in globozoospermia with Y chromosome microdeletion after ICSI. Hum. Reprod. 2002; 17:18331836.Google Scholar
Stalf, T, Sánchez, R, Köhn, FM, et al. Pregnancy and birth after intracytoplasmic sperm injection with spermatozoa from a patient with tail stump syndrome. Hum. Reprod. 1995; 10:21122114.Google Scholar
Okada, H, Fujioka, H, Tatsumi, N, et al. Assisted reproduction for infertile patients with 9 + 0 immotile spermatozoa associated with autosomal dominant polycystic kidney disease. Hum. Reprod. 1999; 14:110113.Google Scholar
Tesarik, J, Rolet, F, Brami, C, et al. Spermatid injection into human oocytes. II. Clinical application in the treatment of infertility due to non-obstructive azoospermia. Hum. Reprod. 1996; 11:780783.Google Scholar
Terriou, P, Hans, E, Giorgetti, C, et al. Pentoxifylline initiates motility in spontaneously immotile epididymal and testicular spermatozoa and allows normal fertilization, pregnancy, and birth after intracytoplasmic sperm injection. J. Assist. Reprod. Genet. 2000; 17:194199.Google Scholar
Aktan, TM, Montag, M, Duman, S, et al. Use of a laser to detect viable but immotile spermatozoa. Andrologia 2004; 36:366369.Google Scholar
Said, TM, Agarwal, A, Grunewald, S, et al. Evaluation of sperm recovery following annexin V magnetic-activated cell sorting separation. RBM Online 2006;13:336339.Google Scholar
Bartoov, B, Berkovitz, A, Eltes, F, et al. Real-time fine morphology of motile human sperm cells is associated with IVF–ICSI outcome. J. Androl. 2002; 23:18.Google Scholar
Parmegiani, L, Cognigni, GE, Bernardi, S, et al. “Physiologic ICSI”: hyaluronic acid (HA) favors selection of spermatozoa without DNA fragmentation and with normal nucleus, resulting in improvement of embryo quality. Fertil. Steril. 2010; 93:598604.Google Scholar
Huszar, G, Ozkavukcu, S, Jakab, A, et al. Hyaluronic acid binding ability of human sperm reflects cellular maturity and fertilizing potential: selection of sperm for intracytoplasmic sperm injection. Curr. Opin. Obstet. Gynecol. 2003; 18:260267.Google Scholar
Yagci, A, Murk, W, Stronk, J, Huszar, G. Spermatozoa bound to solid state hyaluronic acid show chromatin structure with high DNA chain integrity: an acridine orange fluorescence study. J. Androl. 2010; 31:566572.Google Scholar
Chiou Fen, C, Ni Lee, S, Nee Lim, M, Ling Yu, S. Relationship between sperm hyaluronan-binding assay (HBA) scores on embryo development, fertilisation, and pregnancy rate in patients undergoing intra-cytoplasmic sperm injection (ICSI). Proc. Singapore Healthc. 2013; 22:120124.Google Scholar
Morozumi, K, Shikano, T, Miyazaki, S, Yanagimachi, R. Simultaneous removal of sperm plasma membrane and acrosome before intracytoplasmic sperm injection improves oocyte activation/embryonic development. Proc. Natl. Acad. Sci. U. S. A. 2006; 103:1766117666.Google Scholar
Ramalho-Santos, J, Sutovsky, P, Simerly, C, et al. ICSI choreography: fate of sperm structures after monospermic rhesus ICSI and first cell cycle implications. Hum. Reprod. 2000; 15:26102620.Google Scholar
Rawe, VY, Olmedo, SB, Nodar, FN, et al. Cytoskeletal organization defects and abortive activation in human oocytes after IVF and ICSI failure. Mol. Hum. Reprod. 2000; 6:510516.Google Scholar
Murase, Y, Araki, Y, Mizuno, S, et al. Pregnancy following chemical activation of oocytes in a couple with repeated failure of fertilization using ICSI: case report. Hum. Reprod. 2004; 19:16041607.Google Scholar
Borges, E Jr, de Almeida Ferreira Braga, DP, de Sousa Bonetti, TC, Iaconelli, A Jr, Franco, JG Jr. Artificial oocyte activation using calcium ionophore in ICSI cycles with spermatozoa from different sources. RBM Online 2009; 18:4552.Google Scholar
Schmiady, H, Schulze, W, Scheiber, I, Pfüller, B. High rate of premature chromosome condensation in human oocytes following microinjection with round-headed sperm: case report. Hum. Reprod. 2005; 20:13191323.Google Scholar
Wald, M, Ross, LS, Prins, GS, et al. Analysis of outcomes of cryopreserved surgically retrieved sperm for IVF/ICSI. J. Androl. 2006; 27:6065.Google Scholar
Chapter 5: Sperm Preparation Techniques. In: Cooper, TG, Aitken, J, Auger, J, et al., eds., World Health Organization Laboratory Manual for the Examination and Processing of Human Semen, 5th ed. Switzerland: WHO Press. 2010; 161168.Google Scholar
Brackett, NL, Kafetsoulis, A, Ibrahim, E, Aballa, TC, Lynne, CM. Application of 2 vibrators salvages ejaculatory failures to 1 vibrator during penile vibratory stimulation in men with spinal cord injuries. J. Urol. 2007; 177:660663.Google Scholar
Saito, K, Kinoshita, Y, Hosaka, M. Direct and indirect effects of electrical stimulation on the motility of human sperm. Int. J. Urol. 1999; 6:196199.Google Scholar
Brackett, NL, Padron, OF, Lynne, CM. Semen quality of spinal cord injured men is better when obtained by vibratory stimulation versus electroejaculation. J. Urol. 1997; 157:151157.Google Scholar
Hewitson, L, Simerly, C, Schatten, G. Cytoskeletal aspects of assisted fertilization. Semin. Reprod. Med. 2000;18:151159.Google Scholar
Hewitson, L, Dominko, T, Takahashi, D, et al. Unique checkpoints during the first cell cycle of fertilization after intracytoplasmic sperm injection in rhesus monkeys. Nat. Med. 1999; 5:431433.Google Scholar
Silber, SJ, Alagappan, R, Brown, LG, Page, DC. Y chromosome deletions in azoospermic and severely oligozoospermic men undergoing intracytoplasmic sperm injection after testicular sperm extraction. Hum. Reprod. 1998; 13:33323337.Google Scholar
Page, DC, Silber, S, Brown, LG. Men with infertility caused by AZFc deletion can produce sons by intracytoplasmic sperm injection, but are likely to transmit the deletion and infertility. Hum. Reprod. 1999; 14:17221726.Google Scholar
Tournaye, H, Devroey, P, Camus, M, et al. Comparision of in-vitro fertilization in male and tubal infertility: a 3 year survey. Hum. Reprod. 1992; 7:218222.Google Scholar
Devroey, P. Clinical application of new micromanipulative technologies to treat the male. Hum. Reprod. 1998; 13:112122.Google Scholar
Afzelius, BA, Eliasson, R. Male and female infertility problems in the immotile-cilia syndrome. Eur. J. Respir. Dis. Suppl. 1983; 127:144147.Google Scholar
Liu, J, Nagy, Z, Joris, H, et al. Analysis of 76 total fertilization failure cycles out of 2732 intracytoplasmic sperm injection cycles. Hum. Reprod. 1995; 10:26302636.Google Scholar
Gul, U, Turunc, T, Haydardedeoglu, B, et al. Sperm retrieval and live birth rates in presumed Sertoli-cell-only syndrome in testis biopsy: a single centre experience. Andrology 2013; 1:4751.Google Scholar
Oates, RD, Amos, JA. The genetic basis of congenital bilateral absence of the vas deferens and cystic fibrosis. J. Androl. 1994; 15:18.Google Scholar
Jarvi, K, Zielenski, J, Wilschanski, M, et al. Cystic fibrosis transmembrane conductance regulator and obstructive azoospermia. Lancet 1995; 345:1578.Google Scholar
Antinori, S, Versaci, C, Dani, G, et al. Fertilization with human testicular spermatids: four successful pregnancies. Hum. Reprod. 1997; 12:286291.Google Scholar
Araki, Y, Motoyama, M, Yoshida, A, et al. Intracytoplasmic injection with late spermatids: a successful procedure in achieving childbirth for couples in which the male partner suffers from azoospermia due to deficient spermatogenesis. Fertil. Steril. 1997; 67:559561.Google Scholar
Hansen, M, Kurinczuk, JJ, Bower, C, Webb, S. The risk of major birth defects after intracytoplasmic sperm injection and in vitro fertilization. N. Engl. J. Med. 2002; 10:725730.Google Scholar
Silber, S, Escudero, T, Lenahan, K, et al. Chromosomal abnormalities in embryos derived from testicular sperm extraction. Fertil. Steril. 2003; 79:3038.Google Scholar
Doornbos, ME, Maas, SM, McDonnell, J, Vermeiden, JP, Hennekam, RC. Infertility, assisted reproduction technologies and imprinting disturbances. Hum. Reprod. 2007; 22:24762480.Google Scholar
Van Steirteghem, A, Bonduelle, M, Devroey, P, Liebaers, I. Follow-up of children born after ICSI. Hum. Reprod. Update 2002; 8:111116.Google Scholar
Palermo, GD, Colombero, LT, Schattman, GL, Davis, OK, Rosenwaks, Z. Evolution of pregnancies and initial follow-up of newborns delivered after intracytoplasmic sperm injection. JAMA 1996; 276:19831987.Google Scholar
Bowen, JR, Gibson, FL, Leslie, GI, Saunders, DM. Medical and developmental outcome at 1 year for children conceived by intracytoplasmic sperm injection. Lancet 1998; 351:15291534.Google Scholar
Bondulle, M, Joris, H, Hofmans, K, Liebaers, I, Van Steirteghem, A. Mental development of 201 ICSI children at 2 years of age. Lancet 1998; 351:1553.Google Scholar
Sutcliffe, AG, Taylor, B, Li, J, et al. Children born after intracytoplasmic sperm injection: population control study. BMJ 1999; 318:704705.Google Scholar
Winter, C, Van Acker, F, Bonduelle, M, Desmyttere, S, Nekkebroeck, J. Psychosocial development of full term singletons, born after preimplantation genetic diagnosis (PGD) at preschool age and family functioning: a prospective case-controlled study and multi-informant approach. Hum. Reprod. 2015; 3:11221136.Google Scholar
DeBaun, MR, Niemitz, EL, Feinberg, AP. Association of in vitro fertilization with Beckwith–Wiedemann syndrome and epigenetic alterations of LIT1 and H19. Am. J. Hum. Genet. 2003; 72:156160.Google Scholar

References

Brinster, RL. A method for in vitro cultivation of mouse ova from two-cell to blastocyst. Exp. Cell Res. 1963; 32:205208.CrossRefGoogle ScholarPubMed
Gott, AL, Hardy, K, Winston, RM, Leese, HJ. Non-invasive measurement of pyruvate and glucose uptake and lactate production by single human preimplantation embryos. Hum. Reprod. 1990; 5:104108.Google Scholar
Hardarson, T, Bungum, M, Conaghan, J, et al. Noninferiority, randomized, controlled trial comparing embryo development using media developed for sequential or undisturbed culture in a time-lapse setup. Fertil. Steril. 2015; 104:14521459.Google Scholar
Machtinger, R, Racowsky, C. Culture systems: single step. Methods Mol. Biol. 2012; 912:199209.Google Scholar
McKiernan, SH, Bavister, BD. Environmental variables influencing in vitro development of hamster 2-cell embryos to the blastocyst stage. Biol. Reprod. 1990; 43:404413.Google Scholar
Gardner, DK, Lane, M. Culture and selection of viable blastocysts: a feasible proposition for human IVF? Hum. Reprod. Update 1997; 3:367382.Google Scholar
Borland, RM, Biggers, JD, Lechene, CP, Taymor, ML. Elemental composition of fluid in the human Fallopian tube. J. Reprod. Fertil. 1980; 58:479482.Google Scholar
Kleijkers, SH, van Montfoort, AP, Bekers, O, et al. Ammonium accumulation in commercially available embryo culture media and protein supplements during storage at 2–8°C and during incubation at 37°C. Hum. Reprod. 2016; 31:11921199.Google Scholar
Dunglison, GF, Barlow, DH, Sargent, IL. Leukaemia inhibitory factor significantly enhances the blastocyst formation rates of human embryos cultured in serum-free medium. Hum. Reprod. 1996; 11:191196.Google Scholar
Martin, KL, Barlow, DH, Sargent, IL. Heparin-binding epidermal growth factor significantly improves human blastocyst development and hatching in serum-free medium. Hum. Reprod. 1998; 13:16451652.Google Scholar
Lighten, AD, Moore, GE, Winston, RM, Hardy, K. Routine addition of human insulin-like growth factor-I ligand could benefit clinical in-vitro fertilization culture. Hum. Reprod. 1998; 13:31443150.Google Scholar
Sjöblom, C, Wikland, M, Robertson, SA. Granulocyte-macrophage colony-stimulating factor promotes human blastocyst development in vitro. Hum. Reprod. 1999; 14:30693076.Google Scholar
Squirrell, JM, Lane, M, Bavister, BD. Altering intracellular pH disrupts development and cellular organization in preimplantation hamster embryos. Biol. Reprod. 2001; 64:18451854.Google Scholar
Fawzy, M, Emad, M, Gad, MA, et al. Comparing 36.5°C with 37°C for human embryo culture: a prospective randomized controlled trial. RBM Online 2018; 36:620626.Google Scholar
Gardner, DK. Reduced oxygen tension increases blastocyst development, differentiation and viability. Fertil. Steril. 1999; 72:S30S31.Google Scholar
Shimada, M, Kawano, N, Terada, T. Delay of nuclear maturation and reduction in developmental competence of pig oocytes after mineral oil overlay of in vitro maturation media. Reproduction 2002; 124:557564.Google Scholar
Khamsi, F, Roberge, S, Lacanna, IC, Wong, J, Yavas, Y. Effects of granulosa cells, cumulus cells, and oocyte density on in vitro fertilization in women. Endocrine 1999; 10:161166.Google Scholar
Almagor, M, Bejar, C, Kafka, I, Yaffe, H. Pregnancy rates after communal growth of preimplantation human embryos in vitro. Fertil. Steril. 1996; 66:394397.Google Scholar
Rijnders, PM, Jansen, CA. Influence of group culture and culture volume on the formation of human blastocysts: a prospective randomized study. Hum. Reprod. 1999; 14:23332337.Google Scholar
Tao, T, Robichaud, A, Mercier, J, Ouellette, R. Influence of group embryo culture strategies on the blastocyst development and pregnancy outcome. J. Assist. Reprod. Genet. 2013; 30:6368.Google Scholar
Menezo, Y, Hazout, A, Dumont, M, Herbaut, N, Nicollet, B. Coculture of embryos on Vero cells and transfer of blastocysts in humans. Hum. Reprod. 1992; 7:101106.Google Scholar
Freeman, MR, Whitworth, CM, Hill, GA. Granulosa cell co-culture enhances human embryo development and pregnancy rate following in-vitro fertilization. Hum. Reprod. 1995; 10:408414.Google Scholar
Liu, LP, Chan, ST, Ho, PC, Yeung, WS. Partial purification of embryotrophic factors from human oviductal cells. Hum. Reprod. 1998; 13:16131619.Google Scholar
Sakkas, D, Shoukir, Y, Chardonnens, D, Bianchi, PG, Campana, A. Early cleavage of human embryos to the two-cell stage after intracytoplasmic sperm injection as an indicator of embryo viability. Hum. Reprod. 1998; 13:182187.Google Scholar
Sathananthan, AH, Kola, I, Osborne, J, et al. Centrioles in the beginning of human development. Proc. Natl. Acad. Sci. U. S. A. 1991; 88:48064810.Google Scholar
Papale, L, Fiorentino, A, Montag, M, Tomasi, G. The zygote. Hum. Reprod. 2012; 27(Suppl. 1):i22i49.Google Scholar
Gámiz, P, Rubio, C, de los Santos, MJ, et al. The effect of pronuclear morphology on early development and chromosomal abnormalities in cleavage-stage embryos. Hum. Reprod. 2003; 18:24132419.Google Scholar
Tesarik, J, Greco, E. The probability of abnormal preimplantation development can be predicted by a single static observation on pronuclear stage morphology. Hum. Reprod. 1999; 14:13181323.Google Scholar
Sadowy, S, Tomkin, G, Munné, S, Ferrara-Congedo, T, Cohen, J. Impaired development of zygotes with uneven pronuclear size. Zygote 1998; 6:137141.Google Scholar
Payne, D, Flaherty, SP, Barry, MF, Matthews, CD. Preliminary observations on polar body extrusion and pronuclear formation in human oocytes using time-lapse video cinematography. Hum. Reprod. 1997; 12:532541.Google Scholar
Kola, I, Trounson, A, Dawson, G, Rogers, P. Tripronuclear human oocytes: altered cleavage patterns and subsequent karyotypic analysis of embryos. Biol. Reprod. 1987; 37:395401.Google Scholar
Balakier, H, Bouman, D, Sojetcki, A, Librach, C, Squire, JA. Morphological and cytogenetic analysis of human giant oocytes and giant embryos. Hum. Reprod. 2002; 17:23942401.Google Scholar
Staessen, C, Van Steirteghem, AC. The chromosomal constitution of embryos developing from abnormally fertilized oocytes after intracytoplasmic sperm injection and conventional in-vitro fertilization. Hum. Reprod. 1997; 12:321327.Google Scholar
Gras, L, Trounson, AO. Pregnancy and birth resulting from transfer of a blastocyst observed to have one pronucleus at the time of examination for fertilization. Hum. Reprod. 1999; 14:18691871.Google Scholar
Cummins, JM, Breen, TM, Harrison, KL, et al. A formula for scoring human embryo growth rates in in vitro fertilization: its value in predicting pregnancy and in comparison with visual estimates of embryo quality. J. In Vitro Fert. Embryo Transf. 1986; 3:284295.Google Scholar
Braude, P, Bolton, V, Moore, S. Human gene expression first occurs between the four- and eight-cell stages of preimplantation development. Nature 1988; 332:459461.Google Scholar
Baczkowski, T, Kurzawa, R, Głabowski, W. Methods of embryo scoring in in vitro fertilization. Reprod. Biol. 2004; 4(1):522.Google Scholar
Moriwaki, T, Suganuma, N, Hayakawa, M, et al. Embryo evaluation by analysing blastomere nuclei. Hum. Reprod. 2004; 19:152156.Google Scholar
Seikkula, J, Oksjoki, S, Hurme, S, et al. Pregnancy and perinatal outcomes after transfer of binucleated or multinucleated frozen-thawed embryos: a case-control study. RBM Online 2018; 36:607613.Google Scholar
Tesarik, J. Is blastomere multinucleation a safeguard against embryo aneuploidy? Back to the future. RBM Online 2018; 37:506507.Google Scholar
Buster, JE, Bustillo, M, Rodi, IA, et al. Biologic and morphologic development of donated human ova recovered by nonsurgical uterine lavage. Am. J. Obstet. Gynecol. 1985; 153:211217.Google Scholar
Halvaei, I, Khalili, MA, Esfandiari, N, et al. Ultrastructure of cytoplasmic fragments in human cleavage stage embryos. J. Assist. Reprod. Genet. 2016; 33:16771684.Google Scholar
Racowsky, C, Combelles, CM, Nureddin, A, et al. Day 3 and day 5 morphological predictors of embryo viability. RBM Online 2003; 6:323331.Google Scholar
Almeida, PA, Bolton, VN. Cytogenetic analysis of human preimplantation embryos following developmental arrest in vitro. Reprod. Fertil. Dev. 1998; 10:505513.Google Scholar
Almeida, PA, Bolton, VN. The relationship between chromosomal abnormality in the human preimplantation embryos following developmental in vitro. Reprod. Fertil. Dev. 1996; 8:235241.Google Scholar
Magli, MC, Gianaroli, L, Munné, S, Ferraretti, AP. Incidence of chromosomal abnormalities from a morphologically normal cohort of embryos in poor-prognosis patients. J. Assist. Reprod. Genet. 1998; 15:297301.Google Scholar
Kligman, I, Benadiva, C, Alikani, M, Munne, S. The presence of multinucleated blastomeres in human embryos is correlated with chromosomal abnormalities. Hum. Reprod. 1996; 11:14921498.Google Scholar
Piotrowska-Nitsche, K, Perea-Gomez, A, Haraguchi, S, Zernicka-Goetz, M. Four-cell stage mouse blastomeres have different developmental properties. Development 2005; 132:479490.Google Scholar
Torres-Padilla, ME, Parfitt, DE, Kouzarides, T, Zernicka-Goetz, M. Histone arginine methylation regulates pluripotency in the early mouse embryo. Nature 2007; 445:214218.Google Scholar
Bischoff, M, Parfitt, DE, Zernicka-Goetz, M. Formation of the embryonic-abembryonic axis of the mouse blastocyst: relationships between orientation of early cleavage divisions and pattern of symmetric/asymmetric divisions. Development 2008; 135:953962.Google Scholar
Iwata, K, Yumoto, K, Sugishima, M, et al. Analysis of compaction initiation in human embryos by using time-lapse cinematography. J. Assist. Reprod. Genet. 2014; 31:421426.Google Scholar
Fierro-Gonzalez, JC, White, MD, Silva, JC, Plachta, N. Cadherin-dependent filopodia control preimplantation embryo compaction. Nat. Cell. Biol. 2013; 15:14241433.Google Scholar
Morris, SA, Teo, RT, Li, H, et al. Origin and formation of the first two distinct cell types of the inner cell mass in the mouse embryo. Proc. Natl. Acad. Sci. U. S. A. 2010; 107:63646369.Google Scholar
Watson, AJ, Natale, DR, Barcroft, LC. Molecular regulation of blastocyst formation. Anim. Reprod. Sci. 2004; 82–83:583592.Google Scholar
White, MD, Zenker, J, Bissiere, S, Plachta, N. Instructions for assembling the early mammalian embryo. Dev. Cell 2018; 45:667679.Google Scholar
Hardarson, T, Caisander, G, Sjögren, A, et al. A morphological and chromosomal study of blastocysts developing from morphologically suboptimal human pre-embryos compared with control blastocysts. Hum. Reprod. 2003; 18:399407.Google Scholar
Van Blerkom, J. Development of human embryos to the hatched blastocyst stage in the presence or absence of a monolayer of Vero cells. Hum. Reprod. 1993; 8:15251539.Google Scholar
Hardy, K, Handyside, AH, Winston, RM. The human blastocyst: cell number, death and allocation during late preimplantation development. Development 1989; 107:597604.Google Scholar
Woodward, BJ, Lenton, EA, Turner, K. Human chorionic gonadotrophin: embryonic secretion is a time-dependent phenomenon. Hum. Reprod. 1993; 8:14631468.Google Scholar
Woodward, BJ, Lenton, EA, Turner, K, Grace, WF. Embryonic human chorionic gonadotropin secretion and hatching: poor correlation with cleavage rate and morphological assessment during preimplantation development in vitro. Hum. Reprod. 1994; 9:19091914.Google Scholar
Munné, S, Alikani, M, Tomkin, G, Grifo, J, Cohen, J. Embryo morphology developmental rates, and maternal age are correlated with chromosome abnormalities. Fertil. Steril. 1995; 64:382391.Google Scholar
Rienzi, L, Capalbo, A, Stoppa, M, et al. No evidence of association between blastocyst aneuploidy and morphokinetic assessment in a selected population of poor-prognosis patients: a longitudinal cohort study. RBM Online 2015; 30:5766.Google Scholar

References

Barnes, FL. The effects of the early uterine environment on the subsequent development of embryo and fetus. Theriogenology 2000; 53:649658.Google Scholar
Gardner, DK, Lane, M, Calderon, I, Leeton, J. Environment of the preimplantation human embryo in vivo: metabolite analysis of oviduct and uterine fluids and metabolism of cumulus cells. Fertil. Steril. 1996; 65:349353.Google Scholar
Tay, JI, Rutherford, AJ, Killick, SR, et al. Human tubal fluid: production, nutrient composition and response to adrenergic agents. Hum. Reprod. 1997; 12:24512456.Google Scholar
Fischer, B, Bavister, BD. Oxygen tension in the oviduct and uterus of rhesus monkeys, hamsters and rabbits. J. Reprod. Fertil. 1993; 99:673679.Google Scholar
Kaufman, DL, Mitchell, JA. Intrauterine oxygen tension during the oestrous cycle in the hamster: patterns of change. Comp. Biochem. Physiol. Comp. Physiol. 1994; 107:673678.Google Scholar
Ottosen, LD, Hindkaer, J, Husth, M, et al. Observations on intrauterine oxygen tension measured by fibre-optic microsensors. RBM Online 2006; 13:380385.Google Scholar
Macdonald, RR, Lumley, IB. Endocervical pH measured in vivo through the normal menstrual cycle. Obstet. Gynecol. 1970; 35:202206.Google Scholar
Will, MA, Clark, NA, Swain, JE. Biological pH buffers in IVF: help or hindrance to success. J. Assist. Reprod. Genet. 2011; 28:711724.Google Scholar
Simic, N, Ravlic, A. Changes in basal body temperature and simple reaction times during the menstrual cycle. Arh. Hig. Rada Toksikol. 2013; 64:99106.Google Scholar
Zuspan, FP, Rao, P. Thermogenic alterations in the woman. I. Interaction of amines, ovulation, and basal body temperature. Am. J. Obstet. Gynecol. 1974; 118:671678.Google Scholar
Hunter, RH. Temperature gradients in female reproductive tissues. RBM Online 2012; 24:377380.Google Scholar
Wong, JJ, Au, AY, Gao, D, et al. RBM3 regulates temperature sensitive miR-142-5p and miR-143 (thermomiRs), which target immune genes and control fever. Nucleic Acids Res. 2016; 44:28882897.Google Scholar
Parsons, JH, Bolton, VN, Wilson, L, Campbell, S. Pregnancies following in vitro fertilization and ultrasound-directed surgical embryo transfer by perurethral and transvaginal techniques. Fertil. Steril. 1987; 48:691693.Google Scholar
Kato, O, Takatsuka, R, Asch, RH. Transvaginal-transmyometrial embryo transfer: the Towako method; experiences of 104 cases. Fertil. Steril. 1993; 59:5153.Google Scholar
Desai, NN, Goldstein, J, Rowland, DY, Goldfarb, JM. Morphological evaluation of human embryos and derivation of an embryo quality scoring system specific for day 3 embryos: a preliminary study. Hum. Reprod. 2000; 15:21902196.Google Scholar
Ziebe, S, Petersen, K, Lindenberg, S, et al. Embryo morphology or cleavage stage: how to select the best embryos for transfer after in-vitro fertilization. Hum. Reprod. 1997; 12:15451549.Google Scholar
Kahraman, S, Yakin, K, Dönmez, E, et al. Relationship between granular cytoplasm of oocytes and pregnancy outcome following intracytoplasmic sperm injection. Hum. Reprod. 2000; 15:23902393.Google Scholar
Von Royen, E, Mangelschots, K, De Neubourg, D, et al. Characterization of a top quality embryo, a step towards single-embryo transfer. Hum. Reprod. 1999; 14:23452349.Google Scholar
Giorgetti, C, Terriou, P, Auquier, P, et al. Embryo score to predict implantation after in-vitro fertilization: based on 957 single embryo transfers. Hum. Reprod. 1995; 10:24272431.Google Scholar
Antczak, M, Van Blerkom, J. Temporal and spatial aspects of fragmentation in early human embryos: possible effects on developmental competence and association with the differential elimination of regulatory proteins from polarized domains. Hum. Reprod. 1999; 14:429447.Google Scholar
Shoukir, Y, Campana, A, Farley, T, Sakkas, D. Early cleavage of in-vitro fertilized human embryos to the 2-cell stage: a novel indicator of embryo quality and viability. Hum. Reprod. 1997; 12:15311536.Google Scholar
Magli, MC, Gianaroli, L, Ferraretti, AP, et al. Embryo morphology and development are dependent on the chromosomal complement. Fertil. Steril. 2007; 87:534541.Google Scholar
Murakoshi, Y, Sueoka, K, Takahashi, K, et al. Embryo developmental capability and pregnancy outcome are related to the mitochondrial DNA copy number and ooplasmic volume. J. Assist. Reprod. Genet. 2013; 30:13671375.Google Scholar
Braude, P, Bolton, V, Moore, S. Human gene expression first occurs between the four- and eight-cell stages of preimplantation development. Nature 1988; 332:459461.Google Scholar
Gardner, DK, Vella, P, Lane, M, et al. Culture and transfer of human blastocysts to increase implantation rate and eliminate high order multiple gestations: a prospective randomised trial. Fertil. Steril. 1998; 69:8488.Google Scholar
Dokras, A, Sargent, IL, Barlow, DH. Human blastocyst grading: an indicator of developmental potential? Hum. Reprod. 1993; 8:21192127.Google Scholar
Gardner, DK, Lane, M, Stevens, J, Schlenker, T, Schoolcraft, WB. Blastocyst score affects implantation and pregnancy outcome: towards a single blastocyst transfer. Fertil. Steril. 2000; 73:11551158.Google Scholar
Shapiro, BS, Daneshmand, ST, Desai, J, et al. The risk of embryo-endometrium asynchrony increases with maternal age after ovarian stimulation and IVF. RBM Online 2016; 33:5055.Google Scholar
Cohen, J, Malter, H, Fehilly, C, et al. Implantation of embryos after partial opening of oocyte zona pellucida to facilitate sperm penetration. Lancet 1988; 2:162.Google Scholar
De Vos, A, Van Steirteghem, A. Zona hardening, zona drilling and assisted hatching: new achievements in assisted reproduction. Cell Tissues Organs 2000; 166:220227.Google Scholar
Kim, HJ, Kim, CH, Lee, SM, et al. Outcomes of preimplantation genetic diagnosis using either zona drilling with acidified Tyrode’s solution or partial zona dissection. Clin. Exp. Reprod. Med. 2012; 39:118124.Google Scholar
Obruca, A, Strohmer, H, Blaschitz, A, et al. Ultrastructural observations in human oocytes and preimplantation embryos after zona opening using an erbium–yttrium–aluminium–garnet (Er: YAG) laser. Hum Reprod. 1997; 12:22422245.Google Scholar
Mahadevan, MM, Miller, MM, Maris, MO, Moutos, D. Assisted hatching of embryos by micromanipulation for human in vitro fertilization, UAMS experience. J. Ark. Med. Soc. 1998; 94:529531.Google Scholar
Feng, HL, Hershlag, A, Scholl, GM, Cohen, MA. A retroprospective study comparing three different assisted hatching techniques. Fertil. Steril. 2009; 91:13231325.Google Scholar
Grace, J, Bolton, V, Braude, P, Khalaf, Y. Assisted hatching is more effective when embryo quality was optimal in previous failed IVF/ICSI cycles. J. Obstet. Gynaecol. 2007; 27:5660.Google Scholar
Meldrum, DR, Wisot, A, Yee, B, et al. Assisted hatching reduces the age-related decline in IVF outcome in women younger than age 43 without increasing miscarriage or monozygotic twinning. J. Assist. Reprod. Genet. 1998; 15:418421.Google Scholar
Cohen, J, Alikani, M, Trowbridge, J, Rosenwaks, Z. Implantation enhancement by selective assisted hatching using zona drilling of human embryos with poor prognosis. Hum. Reprod. 1992; 7:685691.Google Scholar
Lanzendorf, SE, Nehchiri, F, Mayer, JF, Oehninger, S, Muasher, SJ. A prospective, randomized, double-blind study for the evaluation of assisted hatching in patients with advanced maternal age. Hum. Reprod. 1998; 13:409413.Google Scholar
Hellebaut, S, De Sutter, P, Dozortsev, D, et al. Does assisted hatching improve implantation rates after in vitro fertilization or intracytoplasmic sperm injection in all patients? A prospective randomized study. J. Assist. Reprod. Genet. 1996; 13:1922.Google Scholar
Tucker, MJ, Morton, PC, Wright, G, et al. Enhancement of outcome from intracytoplasmic sperm injection: does co-culture or assisted hatching improve implantation rates? Hum. Reprod. 1996; 11:24342437.Google Scholar
Agarwal, SK, Coe, S, Buyalos, RP. The influence of uterine position on pregnancy rates with in vitro fertilization-embryo transfer. J. Assist. Reprod. Genet. 1994; 11:323324.Google Scholar
Moini, A, Kiani, K, Bahmanabadi, A, Akhoond, M, Akhlaghi, A. Improvement in pregnancy rate by removal of cervical discharge prior to embryo transfer in ICSI cycles: a randomised clinical trial. Aust. N. Z. J. Obstet. Gynaecol. 2011; 51:315320.Google Scholar
Lewin, A, Schenker, JG, Avrech, O, et al. The role of uterine straightening by passive bladder distension before embryo transfer in IVF cycles. J. Assist. Reprod. Genet. 1997; 14:3234.Google Scholar
Bodri, D, Colodron, M, Garcia, D, et al. Transvaginal versus transabdominal ultrasound guidance for embryo transfer in donor oocyte recipients: a randomized clinical trial. Fertil. Steril. 2011; 95:22632268.Google Scholar
Mansour, R, Aboulghar, M, Serour, G. Dummy embryo transfer: a technique that minimizes the problems of embryo transfer and improves the pregnancy rate in human in vitro fertilization. Fertil. Steril. 1990; 54:678681.Google Scholar
Hearns-Stokes, RM, Miller, BT, Scott, L, et al. Pregnancy rates after embryo transfer depend on the provider at embryo transfer. Fertil. Steril. 2000; 74:8086.Google Scholar
Angelini, A, Brusco, GF, Barnocchi, N, et al. Impact of physician performing embryo transfer on pregnancy rates in an assisted reproductive program. J. Assist. Reprod. Genet. 2006; 23:329332.Google Scholar
Fanchin, R, Harmas, A, Benaoudia, F, et al. Microbial flora of the cervix assessed at the time of embryo transfer adversely affects in vitro fertilization outcome. Fertil. Steril. 1998; 70:866870.Google Scholar
Egbase, PE, Udo, EE. Al-Sharhan, M, Grudzinskas, JG. Prophylactic antibiotics and endocervical microbial inoculation of the endometrium at embryo transfer. Lancet 1999; 354:651652.Google Scholar
Buckett, W. A review and meta-analysis of prospective trials comparing different catheters used for embryo transfer. Fertil. Steril. 2006; 85:728734.Google Scholar
Abou-Setta, AM. Air fluid versus fluid-only models of embryo catheter loading: a systematic review and meta-analysis. RBM Online 2007; 14:8084.Google Scholar
Montag, M, Kupka, M, van der Ven, K, van der Ven, H. ET on day 3 using low versus high fluid volume. Eur. J. Obstet. Gynecol. Reprod. Biol. 2002; 102:5760.Google Scholar
Matorras, R, Mendoza, R, Exposito, A, Rodriguez-Escudero, FJ. Influence of the time interval between embryo catheter loading and discharging on the success of IVF. Hum. Reprod. 2004; 19:20272030.Google Scholar
Lesny, P, Killick, SR, Tetlow, RL, Robinson, J, Maguiness, SD. Uterine junctional zone contractions during assisted reproduction cycles. Hum. Reprod. Update 1998; 4:440445.Google Scholar
Waterstone, J, Curson, R, Parsons, J. Embryo transfer to low uterine cavity. Lancet 1991; 337:1413.Google Scholar
Fanchin, R, Righini, C, Olivennes, F, et al. Uterine contractions at the time of embryo transfer alter pregnancy rates after in-vitro fertilization. Hum. Reprod. 1998; 13:19681974.Google Scholar
Coroleu, B, Barri, PN, Carreras, O, et al. The influence of the depth of embryo replacement into the uterine cavity on implantation rates after IVF: a controlled, ultrasound-guided study. Hum. Reprod. 2002; 17:341346.Google Scholar
Franco, JG Jr, Martins, AM, Baruffi, RL, et al. Best site for embryo transfer: the upper or lower half of endometrial cavity? Hum. Reprod. 2004; 19:17851790.Google Scholar
Kwon, H, Choi, DH, Kim, EK. Absolute position versus relative position in embryo transfer: a randomized controlled trial. Reprod. Biol. Endocrinol. 2015; 13:78.Google Scholar
Sallam, HN, Sadek, SS. Ultrasound-guided embryo transfer: a meta-analysis of randomized controlled trials. Fertil. Steril. 2003; 80:10421046.Google Scholar
Wood, EG, Batzer, FR, Go, KJ, Gutmann, JN, Corson, SL. Ultrasound-guided soft catheter embryo transfers will improve pregnancy rates in in-vitro fertilization. Hum. Reprod. 2000; 15:107112.Google Scholar
Porat, N, Boehnlein, LM, Schouweiler, CM, Kang, J, Lindheim, SR. Interim analysis of a randomized clinical trial comparing abdominal versus transvaginal ultrasound-guided embryo transfer. J. Obstet. Gynaecol. Res. 2010; 36:384392.Google Scholar
Grygoruk, C, Pietrewicz, P, Modlinski, JA, et al. Influence of embryo transfer on embryo preimplantation development. Fertil. Steril. 2012; 97:14171421.Google Scholar
Groeneveld, E, de Leeuw, B, Vergouw, CG, et al. Standardization of catheter load speed during embryo transfer: comparison of manual and pump-regulated embryo transfer. RBM Online 2012; 24:163169.Google Scholar
Safari, S, Razi, MH, Razi, Y. Routine use of EmbryoGlue(®) as embryo transfer medium does not improve the ART outcomes. Arch. Gynecol. Obstet. 2015; 291:433437.Google Scholar
Singh, N, Gupta, M, Kriplani, A, Vanamail, P. Role of Embryo Glue as a transfer medium in the outcome of fresh non-donor in-vitro fertilization cycles. J. Hum. Reprod. Sci. 2015; 8:214217.Google Scholar
Friedler, S, Schachter, M, Strassburger, D, et al. A randomized clinical trial comparing recombinant hyaluronan/recombinant albumin versus human tubal fluid for cleavage stage embryo transfer in patients with multiple IVF-embryo transfer failure. Hum. Reprod. 2007; 22:24442448.Google Scholar
Bontekoe S, , Heineman, MJ, Johnson, N, Blake, D. Adherence compounds in embryo transfer media for assisted reproductive technologies. Cochrane Database Syst. Rev. 2014; 25:CD007421.Google Scholar
Uchiyama, T, Sakuta, T, Kanayama, T. Regulation of hyaluronan synthases in mouse uterine cervix. Biochem. Biophys. Res. Commun. 2005; 327:927932.Google Scholar
Furnus, CC, Valcarcel, A, Dulout, FN, Errecalde, AL. The hyaluronic acid receptor (CD44) is expressed in bovine oocytes and early stage embryos. Theriogenology 2003; 60:16331644.Google Scholar
Sroga, JM, Montville, CP, Aubuchon, M, Williams, DB, Thomas, MA. Effect of delayed versus immediate embryo transfer catheter removal on pregnancy outcomes during fresh cycles. Fertil. Steril. 2010; 93:20882090.Google Scholar
Tiras, B, Korucuoglu, U, Polat, M, et al. Effect of blood and mucus on the success rates of embryo transfers. Eur. J. Obstet. Gynecol. Reprod. Biol. 2012; 165:239242.Google Scholar
Plowden, TC, Hill, MJ, Miles, SM, et al. Does the presence of blood in the catheter or the degree of difficulty of embryo transfer affect live birth? Reprod. Sci. 2017; 24:726730.Google Scholar
Alvero, R, Hearns-Stokes, RM, Catherino, WH, Leondires, MP, Segars, JH. The presence of blood in the transfer catheter negatively influences outcome at embryo transfer. Hum. Reprod. 2003; 18:18481852.Google Scholar
Goudas, VT, Hammitt, DG, Damario, MA, et al. Blood on the embryo transfer catheter is associated with decreased rates of embryo implantation and clinical pregnancy with the use of in vitro fertilization-embryo transfer. Fertil. Steril. 1998; 70:878882.Google Scholar
Sharif, K, Afnan, M, Lashen, H, et al. Is bed rest following embryo transfer necessary? Fertil. Steril. 1998; 69:478481.Google Scholar
Bar-Hava, I, Kerner, R, Yoeli, R, et al. Immediate ambulation after embryo transfer: a prospective study. Fertil. Steril. 2005; 83:594597.Google Scholar
Gaikwad, S, Garrido, N, Cobo, A, Pellicer, A, Remohi, J. Bed rest after embryo transfer negatively affects in vitro fertilization: a randomized controlled clinical trial. Fertil. Steril. 2013; 100:729735.Google Scholar
Carrillo, AJ, Lane, B, Pridman, DD, et al. Improved clinical outcomes for in vitro fertilization with delay of embryo transfer from 48 to 72 hours after oocyte retrieval: use of glucose- and phosphate-free media. Fertil. Steril. 1998; 69:329334.Google Scholar
Ertzeid, G, Dale, PO, Tanbo, T, et al. Clinical outcome of day 2 versus day 3 embryo transfer using serum-free culture media: a prospective randomized study. J. Assist. Reprod. Genet. 1999; 16:529534.Google Scholar
Huisman, GJ, Alberda, AT, Leerentveld, RA, Verhoeff, A, Zeilmaker, GH. A comparison of in vitro fertilization results after embryo transfer after 2, 3, and 4 days of embryo culture. Fertil. Steril. 1994; 61:970971.Google Scholar
Scholtes, MC, Zeilmaker, GH. A prospective, randomized study of embryo transfer results after 3 or 5 days of embryo culture in in vitro fertilization. Fertil. Steril. 1996; 65:12451248.Google Scholar
Milki, AA, Hinckley, MD, Fisch, JD, Dasig, D, Behr, B. Comparison of blastocyst transfer with day 3 embryo transfer in similar patient populations. Fertil. Steril. 2000; 73:126129.Google Scholar
Stillman, RJ, Richter, KS, Banks, NK, Graham, JR. Elective single embryo transfer: a 6-year progressive implementation of 784 single blastocyst transfers and the influence of payment method on patient choice. Fertil. Steril. 2009; 92:18951906.Google Scholar
Shapiro, BS, Daneshmand, ST, Garner, FC, et al. Evidence of impaired endometrial receptivity after ovarian stimulation for in vitro fertilization: a prospective randomized trial comparing fresh and frozen-thawed embryo transfer in normal responders. Fertil. Steril. 2011; 96:344348.Google Scholar
Roque, M, Lattes, K, Serra, S, et al. Fresh embryo transfer versus frozen embryo transfer in in vitro fertilization cycles: a systematic review and meta-analysis. Fertil. Steril. 2013; 99:156162.Google Scholar
Evans, J, Hannan, NJ, Edgell, TA, et al. Fresh versus frozen embryo transfer: backing clinical decisions with scientific and clinical evidence. Hum. Reprod. Update 2014; 20:808821.Google Scholar
Ishihara, O, Araki, R, Kuwahara, A, et al. Impact of frozen-thawed single-blastocyst transfer on maternal and neonatal outcome: an analysis of 277,042 single-embryo transfer cycles from 2008 to 2010 in Japan. Fertil. Steril. 2014; 101:128133.Google Scholar
Maheshwari, A, Raja, EA, Bhattacharya, S. Obstetric and perinatal outcomes after either fresh or thawed frozen embryo transfer: an analysis of 112,432 singleton pregnancies recorded in the Human Fertilisation and Embryology Authority anonymized dataset. Fertil. Steril. 2016; 106:17031708.Google Scholar
Shih, W, Rushford, DD, Bourne, H, et al. Factors affecting low birth weight after assisted reproduction technology: difference between transfer of fresh and cryopreserved embryos suggests an adverse effect of oocyte collection. Hum. Reprod. 2008; 23:16441653.Google Scholar
Pinborg, A, Henningsen, AA, Loft, A, et al. Large baby syndrome in singletons born after frozen embryo transfer (FET): is it due to maternal factors or the cryotechnique? Hum. Reprod. 2014; 29:618627.Google Scholar
Shapiro, BS, Daneshmand, ST, Bedient, CE, Garner, FC. Comparison of birth weights in patients randomly assigned to fresh or frozen-thawed embryo transfer. Fertil. Steril. 2016; 106:317321.Google Scholar
Roque, M, Valle, M, Kostolias, A, Sampaio, M, Geber, S. Freeze-all cycle in reproductive medicine: current perspectives. JBRA Assist. Reprod. 2017; 21:4953.Google Scholar
Acharya, KS, Acharya, CR, Bishop, K, et al. Freezing of all embryos in in vitro fertilization is beneficial in high responders, but not intermediate and low responders: an analysis of 82,935 cycles from the Society for Assisted Reproductive Technology registry. Fertil. Steril. 2018; 110:880887.Google Scholar
Kogan, MD, Alexander, GR, Kotelchuck, M, et al. Trends in twin birth outcomes and prenatal care utilization in the United States, 1981–1997. JAMA 2000; 284:335341.Google Scholar
Scher, AI, Petterson, B, Blair, E, et al. The risk of mortality or cerebral palsy in twins: a collaborative population-based study. Pediatr. Res. 2002; 52:671681.Google Scholar
Guerif, F, Lemseffer, M, Bidault, R, et al.Single day 2 embryo versus blastocyst-stage transfer: a prospective study integrating fresh and frozen embryo transfers. Hum. Reprod. 2009; 24:10511058.Google Scholar
Yanaihara, A, Yorimitsu, T, Motoyama, H, Watanabe, H, Kawamura, T. Monozygotic multiple gestation following in vitro fertilization: analysis of seven cases from Japan. J. Exp. Clin. Assist. Reprod. 2007; 4:4.Google Scholar
Lee, SF, Chapman, M, Bowyer, L. Monozygotic triplets after single blastocyst transfer: case report and literature review. Aust. N. Z. J. Obstet. Gynaecol. 2008; 48:583586.Google Scholar
Thurin, A, Hausken, J, Hillensjö, T, et al. Elective single-embryo transfer versus double-embryo transfer in in-vitro fertilization. N. Engl. J. Med. 2004; 351:23922402.Google Scholar
Kjellberg, AT, Carlsson, P, Bergh, C. Randomized single versus double embryo transfer: obstetric and paediatric outcome and a cost-effectiveness analysis. Hum. Reprod. 2006; 21:210216.Google Scholar
Ikemoto, Y, Kuroda, K, Ochiai, A, et al. Prevalence and risk factors of zygotic splitting after 937 848 single embryo transfer cycles. Hum. Reprod. 2018; 33:19841991.Google Scholar
Tabibzadeh, S. Molecular control of the implantation window. Hum. Reprod. Update 1998; 4:465471.Google Scholar
Brosens, JJ, Salker, MS, Teklenburg, G, et al. Uterine selection of human embryos at implantation. Sci. Rep. 2014; 4:3894.Google Scholar
Achache, H, Revel, A. Endometrial receptivity markers, the journey to successful embryo implantation. Hum. Reprod. Update 2006; 12:731746.Google Scholar
Karizbodagh, MP, Rashidi, B, Sahebkar, A, Masoudifar, A, Mirzaei, H. Implantation window and angiogenesis. J. Cell Biochem. 2017; 118:41414151.Google Scholar
Shufaro, Y, Simon, A, Laufer, N, Fatum, M. Thin unresponsive endometrium – a possible complication of surgical curettage compromising ART outcome. J. Assist. Reprod. Genet. 2008; 25:421425.Google Scholar
El-Toukhy, T, Coomarasamy, A, Khairy, M, et al. The relationship between endometrial thickness and outcome of medicated frozen embryo replacement cycles. Fertil. Steril. 2008; 89:832839.Google Scholar
Gallos, ID, Khairy, M, Chu, J, et al. Optimal endometrial thickness to maximize live births and minimize pregnancy losses: analysis of 25,767 fresh embryo transfers. RBM Online 2018; 37:542548.Google Scholar
Liu, KE, Hartman, M, Hartman, A, Luo, ZC, Mahutte, N. The impact of a thin endometrial lining on fresh and frozen–thaw IVF outcomes: an analysis of over 40 000 embryo transfers. Hum. Reprod. 2018; 33:18831888.Google Scholar
Moran, NA, Sloan, DB. The hologenome concept: helpful or hollow? PLoS Biol. 2015; 13: e1002311.Google Scholar
Kroon, B, Hart, RJ, Wong, BM, Ford, E, Yazdani, A. Antibiotics prior to embryo transfer in ART. Cochrane Database Syst. Rev. 2012; 3:CD008995.Google Scholar
Moore, DE, Soules, MR, Klein, NA, Fujimoto, VY, Agnew, KJ, Eschenbach, DA. Bacteria in the transfer catheter tip influence the live-birth rate after in vitro fertilization. Fertil. Steril. 2000; 74:11181124.Google Scholar
Garsia-Velasco, JA, Menabrito, M, Catalan, IB. What fertility specialists should know about the vaginal microbiome: a review. RBM Online 2017; 35:103112.Google Scholar
Fox, CA, Wolff, HS, Baker, JA. Measurement of intra-vaginal and intra-uterine pressures during human coitus by radio-telemetry. J. Reprod. Fertil. 1970; 22:243251.Google Scholar
Franchin, R, Righini, C, Olivennes, F, et al. Uterine contractions at the time of embryo transfer alter pregnancy rates after in-vitro fertilization. Hum. Reprod. 1998; 13:19681974.Google Scholar
Tremellen, KP, Valbuena, D, Landeras, J, et al. The effect of intercourse on pregnancy rates during assisted human reproduction. Hum. Reprod. 2000; 15:26532658.Google Scholar
Salamonsen, LA. Tissue injury and repair in the female human reproductive tract. Reproduction 2003; 125:301311.Google Scholar
Gnainsky, Y, Aldo, PB, Barash, A, et al. Local injury of the endometrium induces an inflammatory response that promotes successful implantation. Fertil. Steril. 2010; 94:20302036.Google Scholar
Granot, I, Gnainsky, Y, Dekel, N. Endometrial inflammation and effect on implantation improvement and pregnancy outcome. Reproduction 2012; 144:661668.Google Scholar
Dunn, CL, Kelly, RW, Critchley, HO. Decidualization of the human endometrial stromal cell: an enigmatic transformation. RBM Online 2003; 7:151161.Google Scholar
Paria, BC, Reese, J, Das, SK, Dey, SK. Deciphering the cross-talk of implantation: advances and challenges. Science 2002; 296:21852188.Google Scholar
Siristatidis, C, Kreatsa, M, Koutlaki, N, et al. Endometrial injury for RIF patients undergoing IVF/ICSI: a prospective nonrandomized controlled trial. Gynecol. Endocrinol. 2017; 33:297300.Google Scholar
Gui, J, Xu, W, Yang, J, Feng, L, Jia, J. Impact of local endometrial injury on in vitro fertilization/intracytoplasmic sperm injection outcomes: a systematic review and meta-analysis. J. Obstet. Gynaecol. Res. 2018; 45:5768. doi:10.1111/jog.13854.Google Scholar

References

Zeilmaker, GH, Alberda, AT, van Gent, I, Rijkmans, CM, Drogendijk, AC. Two pregnancies following transfer of intact frozen-thawed embryos. Fertil. Steril. 1984; 42:293296.Google Scholar
Rall, WF, Fahy, GM. Ice-free cryopreservation of mouse embryos at -196°C by vitrification. Nature 1985; 313:573575.Google Scholar
Mazur, P. Kinetics of water loss from cells at subzero temperatures and the likelihood of intracellular freezing. J. Gen. Physiol. 1963; 47:347369.Google Scholar
Newton, HJ, Pegg, DE, Barrass, R, Gosden, RG. Osmotically inactive volume, hydraulic conductivity, and permeability to dimethyl sulphoxide of human mature oocytes. Reprod. Fertil. 1999; 117:2733.Google Scholar
Galvao, J, Davis, B, Tilley, M, et al. Unexpected low-dose toxicity of the universal solvent DMSO. FASEB J. 2014; 28:13171330.Google Scholar
Schneider, U, Mazur, P. Osmotic consequences of cryoprotectant permeability and its relation to the survival of frozen-thawed embryos. Theriogenology 1984; 21:6879.Google Scholar
Edashige, K, Tanaka, M, Ichimaru, N, et al. Channel dependent permeation of water and glycerol in mouse morulae. Biol. Reprod. 2006; 74:625632.Google Scholar
Kasai, M, Edashige, K. Movement of water and cryoprotectants in mouse oocytes and embryos at different stages: relevance to cryopreservation. In: Chian, RC, Quinn, P, eds., Fertility Cryopreservation. Cambridge: Cambridge University Press. 2010; 1623.Google Scholar
Maneiro, E, Ron-Corzo, A, Julve, J, Goyanes, VJ. Surface area/volume ratio and growth equation of the human early embryo. Int. J. Dev. Biol. 1991; 35:139143.Google Scholar
Senn, A, Vozzi, C, Chanson, A, De Grandi, P, Germond, M. Prospective randomized study of two cryopreservation policies avoiding embryo selection: the pronucleate stage leads to a higher cumulative delivery rate than the early cleavage stage. Fertil. Steril. 2000; 74:946952.Google Scholar
Al-Hasani, S, Ozmen, B, Koutlaki, N, et al. Three years of routine vitrification of human zygotes: is it still fair to advocate slow-rate freezing? RBM Online 2007; 14:288293.Google Scholar
Isachenko, V, Todorov, P, Dimitrov, Y, Isachenko, E. Integrity rate of pronuclei after cryopreservation of pronuclear-zygotes as a criteria for subsequent embryo development and pregnancy. Hum. Reprod. 2008; 23:819826.Google Scholar
Oehninger, S, Mayer, J, Muasher, S. Impact of different clinical variables on pregnancy outcome following embryo cryopreservation. Mol. Cell. Endocrinol. 2000; 169:7377.Google Scholar
Kuwayama, M, Vajta, G, Kato, O, Leibo, SP. Highly efficient vitrification method for cryopreservation of human oocytes. RBM Online 2005; 11:300308.Google Scholar
Hartshorne, GM, Wick, K, Elder, K, Dyson, H. Effect of cell number at freezing upon survival and viability of cleaving embryos generated from stimulated IVF cycles. Hum. Reprod. 1990; 5:857861.Google Scholar
Edgar, DH, Bourne, H, Spiers, AL, McBain, JC. A quantitative analysis of the impact of cryopreservation on the implantation potential of human early cleavage stage embryos. Hum. Reprod. 2000; 15:175179.Google Scholar
Zech, NH, Lejeune, B, Zech, H, Vanderzwalmen, P. Vitrification of hatching and hatched human blastocysts: effect of an opening in the zona pellucida before vitrification. RBM Online 2005; 11:355361.Google Scholar
Mukaida, T, Oka, C, Goto, T, Takahashi, K. Artificial shrinkage of blastocoeles using either a micro-needle or a laser pulse prior to the cooling steps of vitrification improves survival rate and pregnancy outcome of vitrified human blastocysts. Hum. Reprod. 2006; 21:32463252.Google Scholar
Richter, KS, Shipley, SK, McVearry, I, Tucker, MJ, Widra, EA. Cryopreserved embryo transfers suggest that endometrial receptivity may contribute to reduced success rates of later developing embryos. Fertil. Steril. 2006; 86:862866.Google Scholar
Gardner, DK, Schoolcraft, WB. In vitro culture of human blastocysts. In: Jansen, R, Mortimer, D, eds., Toward Reproductive Certainty: Fertility and Genetics Beyond. London: Parthenon Publishing. 1999; 378388.Google Scholar
Takahashi, K, Mukaida, T, Goto, T, Oka, C. Perinatal outcome of blastocyst transfer with vitrification using cryoloop: a 4-year follow-up study. Fertil. Steril. 2005; 84:8892.Google Scholar
Pinborg, A, Henningsen, AA, Loft, A, et al. Large baby syndrome in singletons born after frozen embryo transfer (FET): is it due to maternal factors or the cryotechnique? Hum. Reprod. 2014; 29:618627.Google Scholar
Chen, C. Pregnancy after human oocyte cryopreservation. Lancet 1986; 1:884886.Google Scholar
Alpha Scientists in Reproductive Medicine; ESHRE Special Interest Group Embryology. Istanbul consensus workshop on embryo assessment: proceedings of an expert meeting. RBM Online 2011; 22:632646.Google Scholar
Alpha Scientists in Reproductive Medicine and ESHRE Special Interest Group Embryology. Istanbul consensus workshop on embryo assessment: proceedings of an expert meeting. Hum. Reprod. 2011; 26:12701283.Google Scholar
Ebner, T, Yaman, C, Moser, M, et al. Prognostic value of first polar body morphology on fertilization rate and embryo quality in intracytoplasmic sperm injection. Hum. Reprod. 2000; 15:427430.Google Scholar
Hunter, JE, Bernard, A, Fuller, BJ, McGrath, JJ, Shaw, RW. Measurements of the membrane water permeability (Lp) and its temperature dependence (activation energy) in human fresh and failed-to-fertilize oocytes and mouse oocyte. Cryobiology 1992; 29:240249.Google Scholar
Pickering, SJ, Braude, PR, Johnson, MH, Cant, A, Currie, J. Transient cooling to room temperature can cause irreversible disruption of the meiotic spindle in the human oocyte. Fertil. Steril. 1990; 54:102108.Google Scholar
Zenzes, MT, Bielecki, R, Casper, RF, Leibo, SP. Effects of chilling to 0 degrees C on the morphology of meiotic spindles in human metaphase II oocytes. Fertil. Steril. 2001; 75:769777.Google Scholar
Cobo, A, Kuwayama, M, Pérez, S, et al. Comparison of concomitant outcome achieved with fresh and cryopreserved donor oocytes vitrified by the Cryotop method. Fertil. Steril. 2007; 89:16571664.Google Scholar
Doyle, JO, Richter, KS, Lim, J, et al. Successful elective and medically indicated oocyte vitrification and warming for autologous in vitro fertilization, with predicted birth probabilities for fertility preservation according to number of cryopreserved oocytes and age at retrieval. Fertil. Steril. 2016; 105:459466.Google Scholar
Ben Rafael, Z. The dilemma of social oocyte freezing: usage rate is too low to make it cost-effective. RBM Online 2018; 37:443448.Google Scholar
Leibo, SP, Mazur, P, Jackowski, SC. Factors affecting survival of mouse embryos during freezing and thawing. Exp. Cell Res. 1974; 89:7988.Google Scholar
Leibo, SP, Oda, K. High survival of mouse zygotes and embryos cooled rapidly or slowly in ethylene glycol plus polyvinylpyrrolidone. Cryo Letters 1993; 14:133134.Google Scholar
Van den Abbeel, E, Camus, M, Van Waesberghe, L, Devroey, P, Van Steirteghem, A. Viability of partially damaged human embryos after cryopreservation. Hum. Reprod. 1997; 12:20062010.Google Scholar
Rienzi, L, Nagy, ZP, Ubaldi, F, et al. Laser-assisted removal of necrotic blastomeres from cryopreserved embryos that were partially damaged. Fertil. Steril. 2002; 77:11961201.Google Scholar
Hardarson, T, Lofman, C, Coull, G, et al. Internalization of cellular fragments in a human embryo: time-lapse recordings. RBM Online 2002; 5:3638.Google Scholar
Rienzi, L, Ubaldi, F, Iacobelli, M, et al. Developmental potential of fully intact and partially damaged cryopreserved embryos after laser-assisted removal of necrotic blastomeres and post-thaw culture selection. Fertil. Steril. 2005; 84:888894.Google Scholar
Nagy, ZP, Taylor, T, Elliott, T, et al. Removal of lysed blastomeres from frozen-thawed embryos improves implantation and pregnancy rates in frozen embryo transfer cycles. Fertil. Steril. 2005; 84:16061612.Google Scholar
Leoni, GG, Berlinguer, F, Succu, S, et al. A new selection criterion to assess good quality ovine blastocysts after vitrification and to predict their transfer into recipients. Mol. Reprod. Dev. 2008; 75:373382.Google Scholar
Huang, JY, Tulandi, T, Holzer, H, et al. Cryopreservation of ovarian tissue and in vitro matured oocytes in a female with mosaic Turner syndrome: case report. Hum. Reprod. 2008; 23:336339.Google Scholar
De Roo, P, Lierman, S, Tilleman, K, et al. Ovarian tissue cryopreservation in female-to-male transgender people: insights into ovarian histology and physiology after prolonged androgen treatment. RBM Online 2017; 34:557566.Google Scholar
Westphal, JR, Gerritse, R, Braat, DDM, Beerendonk, CCM, Peek, RJ. Complete protection against cryodamage of cryopreserved whole bovine and human ovaries using DMSO as a cryoprotectant. Assist. Reprod. Genet. 2017; 34:12171229.Google Scholar
Haino, T, Tarumi, W, Kawamura, K, et al. Determination of follicular localization in human ovarian cortex for vitrification. J. Adolesc. Young Adult Oncol. 2018; 7:4653.Google Scholar
Newton, H, Fisher, J, Arnold, JR, et al. Permeation of human ovarian tissue with cryoprotective agents in preparation for cryopreservation. Hum. Reprod. 1998; 13:376380.Google Scholar
Kagawa, N, Kuwayama, M, Nakata, K, et al. Production of the first offspring from oocytes derived from fresh and cryopreserved preantral follicles of adult mice. RBM Online 2007; 14:693699.Google Scholar
Hasegawa, A, Mochida, N, Ogasawara, T, Koyama, K. Pup birth from mouse oocytes in preantral follicles derived from vitrified and warmed ovaries followed by in vitro growth, in vitro maturation, and in vitro fertilization. Fertil. Steril. 2006; 86:11821192.Google Scholar
Nakamura, Y, Obata, R, Okuyama, N, et al. Residual ethylene glycol and dimethyl sulphoxide concentration in human ovarian tissue during warming/thawing steps following cryopreservation. RBM Online 2017; 35:311313.Google Scholar
Bastings, L, Westphal, JR, Beerendonk, CCM, et al. Clinically applied procedures for human ovarian tissue cryopreservation result in different levels of efficacy and efficiency. J. Assist. Reprod. Genet. 2016; 33:16051614.Google Scholar
Demeestere, I, Simon, P, Emiliani, S, Delbaere, A, Englert, Y. Orthotopic and heterotopic ovarian tissue transplantation. Hum. Reprod. Update 2009; 15:649665.Google Scholar
Dalman, A, Deheshkar Gooneh Farahani, NS, Totonchi, M, et al. Slow freezing versus vitrification technique for human ovarian tissue cryopreservation: an evaluation of histological changes, WNT signaling pathway and apoptotic genes expression. Cryobiology 2017; 79:2936.Google Scholar
Meirow, D, Levron, J, Eldar-Geva, T, et al. Pregnancy after transplantation of cryopreserved ovarian tissue in a patient with ovarian failure after chemotherapy. N. Engl. J. Med. 2005; 353:318321.Google Scholar
Donnez, J, Dolmans, MM, Demylle, D, et al. Live birth after orthotopic transplantation of cryopreserved ovarian tissue. Lancet 2004; 364:14051410.Google Scholar
Shaw, J, Trounson, AO. Ovarian banking for cancer patients – oncological implications in the replacement of ovarian tissue. Hum. Reprod. 1997; 12:403405.Google Scholar
Polge, C, Smith, AU, Parkes, AS. Revival of spermatozoa after vitrification and dehydration at low temperatures. Nature 1949; 164:666.Google Scholar
Perloff, WH, Steinberger, E, Sherman, JK. Conception with human spermatozoa frozen by nitrogen vapor technic. Fertil. Steril. 1964; 15:501.Google Scholar
Serafini, P, Hauser, D, Moyer, D, Marrs, R. Cryopreservation of human spermatozoa: correlations of ultrastructural sperm head configuration with sperm motility and ability to penetrate zona-free hamster ova. Fertil. Steril. 1986; 46:691695.Google Scholar
Woolley, D, Richardson, D. Ultrastructural injury to human spermatozoa after freezing and thawing. J. Reprod. Fertil. 1978; 53:389394.Google Scholar
Emamverdi, M, Zhandi, M, Shahneh, AZ, et al. Flow cytometric and microscopic evaluation of post-thawed ram semen cryopreserved in chemically defined home-made or commercial extenders. Anim. Prod. Sci. 2015; 55:551558.Google Scholar
Cheung, RCF, Ng, TB, Wong, JH. Antifreeze proteins from diverse organisms and their applications: an overview. Curr. Protein Pept. Sci. 2017; 18:262283.Google Scholar
Kim, HJ, Lee, JH, Hur, YB, et al. Marine antifreeze proteins: structure, function, and application to cryopreservation as a potential cryoprotectant. Mar. Drugs 2017; 15:27.Google Scholar
Isachenko, E, Isachenko, V, Katkov, II, Dessole, S, Nawroth, F. Vitrification of mammalian spermatozoa in the absence of cryoprotectants: From past practical difficulties to present success. RBM Online 2003; 6:191200.Google Scholar
Centola, GM, Raubertas, RF, Mattox, JH. Cryopreservation of human semen. Comparision of cryopreservatives, sources of variability, and prediction of post-thaw survival. J. Andrology 1992; 13:283288.Google Scholar
Giraud, MN, Motta, C, Boucher, D, Grizard, G. Membrane fluidity predicts the outcome of cryopreservation of human spermatozoa. Hum. Reprod. 2000; 15:21602164.Google Scholar
James, PS, Wolfe, CA, Mackie, A, et al. Lipid dynamics in the plasma membrane of fresh and cryopreserved human spermatozoa. Hum. Reprod. 1999; 14:18271832.Google Scholar
Holt, WV. Basic aspects of frozen storage of semen. Anim. Reprod. Sci. 2000; 62:322.Google Scholar
Barthelemy, C, Royere, D, Hammahah, S, et al. Ultrastructural changes in membranes and acrosome of human sperm during cryopreservation. Syst. Biol. Reprod. Med. 1990; 25:2940.Google Scholar
Lalancette, C, Miller, D, Li, Y, Krawetz, SA. Paternal contributions: new functional insights for spermatozoal RNA. J. Cell. Biochem. 2008; 104:15701579.Google Scholar
Jodar, M, Selvaraju, S, Sendler, E, Diamond, MP, Krawetz, SA. The presence, role and clinical use of spermatozoal RNAs. Hum. Reprod. 2013; 19:604624.Google Scholar
Valcarce, DG, Carton-Garcia, F, Herraez, MP, Robles, V. Effect of cryopreservation on human sperm messenger RNAs crucial for fertilization and early embryo development. Cryobiology 2013; 67:8490.Google Scholar
Said, TM, Gaglani, A, Agarwal, A. Implication of apoptosis in sperm cryoinjury. RBM Online 2010; 21:456462.Google Scholar
Liu, T, Gao, J, Zhou, N, et al. The effect of two cryopreservation methods on human sperm DNA damage. Cryobiology 2016; 72:210215.Google Scholar
Sukcharoen, N, Sithipravej, T, Promviengchai, S, Chinpilas, V, Boonkasemsanti, W. Comparison of the outcome of intracytoplasmic sperm injection using fresh and frozen-thawed epididymal spermatozoa obtained by percutaneous epididymal sperm aspiration. J. Med. Assoc. Thai. 2001; 84(Suppl. 1):S331S337.Google Scholar
Hessel, M, Robben, JC, D’Hauwers, KW, Braat, DD, Ramos, L. The influence of sperm motility and cryopreservation on the treatment outcome after intracytoplasmic sperm injection following testicular sperm extraction. Acta Obstet. Gynecol. Scand. 2015; 94:13131321.Google Scholar
Devroey, P, Silber, S, Nagy, Z, et al. Ongoing pregnancies and birth after intracytoplasmic sperm injection with frozen-thawed epididymal spermatozoa. Hum. Reprod. 1995; 10:903906.Google Scholar
Tongdee, P, Sukprasert, M, Satirapod, C, Wongkularb, A, Choktanasiri, W. Comparison of cryopreserved human sperm between ultra rapid freezing and slow programmable freezing: effect on motility, morphology and DNA integrity. J. Med. Assoc. Thai. 2015; 98:S33S42.Google Scholar
Bordson, BL, Ricci, E, Dickey, RP, et al. Comparison of fecundability with fresh and frozen semen in therapeutic donor insemination. Fertil. Steril. 1986; 46:466469.Google Scholar
Cohen, J, Garrisi, GJ, Congedo-Ferrara, TA, et al. Cryopreservation of single human spermatozoa. Hum. Reprod. 1997; 12:9941001.Google Scholar
Liu, J, Zheng, XZ, Baramki, TA, et al. Cryopreservation of a small number of fresh human testicular spermatozoa and testicular spermatozoa cultured in vitro for 3 days in an empty zona pellucida. J. Androl. 2000; 21:409413.Google Scholar
Berkovitz, A, Miller, N, Silberman, M, Belenky, M, Itsykson, P. A novel solution for freezing small numbers of spermatozoa using a sperm vitrification device. Hum. Reprod. 2018; 33:19751983.Google Scholar
Smith, KD, Steinberger, E. Survival of spermatozoa in a human sperm bank. Effects of long-term storage in liquid nitrogen. J. Am. Med. Assoc. 1973; 223:774777.Google Scholar
Szell, AZ, Bierbaum, RC, Hazelrigg, WB, Chetkowski, RJ. Live births from frozen human semen stored for 40 years. J. Assist. Reprod. Genet. 2013; 36:743744.Google Scholar
Selvaggi, G, Ceulemans, P, De Cuypere, G, et al. Gender identity disorder: general overview and surgical treatment for vaginoplasty in male-to-female transsexuals. Plast. Reconstr. Surg. 2005; 116:135145.Google Scholar
De Sutter, P. Gender reassignment and assisted reproduction: present and future options for transsexual people. Hum. Reprod. 2001; 16:612614.Google Scholar
Onofre, J, Baert, Y, Faes, K, Goossens, E. Cryopreservation of testicular tissue or testicular cell suspensions: a pivotal step in fertility preservation. Hum. Reprod. Update 2016; 22:744761.Google Scholar
Curaba, M, Poels, J, van Langendonckt, A, Donnez, J, Wyns, C. Can prepubertal human testicular tissue be cryopreserved by vitrification? Fertil. Steril. 2011; 95:2123.e9–2123.e12.Google Scholar
Zarandi, NP, Galdon, G, Kogan, S, Atala, A, Sadri-Ardekani, H. Cryostorage of immature and mature human testis tissue to preserve spermatogonial stem cells (SSCs): a systematic review of current experiences toward clinical applications. Stem Cells Cloning 2018; 11:2338.Google Scholar

References

Eppig, JJ, Telfer, EE. Isolation and culture of oocytes. Methods Enzymol. 1993; 225:7784.Google Scholar
Eppig, JJ, O’Brien, MJ. Development in vitro of mouse oocytes from primordial follicles. Biol. Reprod. 1996; 54:197207.Google Scholar
Edwards, RG. Maturation in vitro of human ovarian oocytes. Lancet. 1965; 2:926929.Google Scholar
Cha, KY, Koo, JJ, Ko, JJ, et al. Pregnancy after in vitro fertilization of human follicular oocytes collected from nonstimulated cycles, their culture in vitro and their transfer in a donor oocyte program. Fertil. Steril. 1991; 55:109113.Google Scholar
Trounson, A, Wood, C, Kausche, A. In vitro maturation and the fertilization and developmental competence of oocytes recovered from untreated polycystic ovarian patients. Fertil. Steril. 1994; 62:353362.Google Scholar
Benkhalifa, M, Demirol, A, Ménézo, Y, et al. Natural cycle IVF and oocyte in-vitro maturation in polycystic ovary syndrome: a collaborative prospective study. RBM Online 2009; 18:2936.Google Scholar
Son, WY, Tan, SL. Laboratory and embryological aspects of hCG-primed in vitro maturation cycles for patients with polycystic ovaries. Hum. Reprod. Update 2010; 16:675689.Google Scholar
Hreinsson, J, Rosenlund, B, Fridén, B, et al. Recombinant LH is equally effective as recombinant hCG in promoting oocyte maturation in a clinical in-vitro maturation programme: a randomized study. Hum. Reprod. 2003; 18:21312136.Google Scholar
Söderström-Anttila, V, Mäkinen, S, Tuuri, T, Suikkari, AM. Favourable pregnancy results with insemination of in vitro matured oocytes from unstimulated patients. Hum. Reprod. 2005; 20:15341540.Google Scholar
Walls, ML, Junk, S, Ryan, JP, Hart, R. IVF versus ICSI for the fertilization of in-vitro matured human oocytes. RBM Online 2012; 25:603607.Google Scholar
Walls, ML, Douglas, K, Ryan, JP, Tan, J, Hart, R. In-vitro maturation and cryopreservation of oocytes at the time of oophorectomy. Gynecol. Oncol. Rep. 2015; 13:7981.Google Scholar
Sela-Abramovich, S, Edry, I, Galiani, D, Nevo, N, Dekel, N. Disruption of gap junctional communication within the ovarian follicle induces oocyte maturation. Endocrinology 2006;147:22802286.Google Scholar
Mehlmann, L. Signaling for meiotic resumption in granulosa cells, cumulus cells, and oocyte. In: Coticchio, G, Albertini, DF, De Santis, L, eds., Oogenesis. London: Springer. 2013; 171182.Google Scholar
Matzuk, M, Li, Q. How the oocyte influences follicular cell function and why. In: Coticchio, G, Albertini, DF, De Santis, L, eds., Oogenesis. London: Springer. 2013; 7592.Google Scholar
Wang, S, Ning, G, Chen, X, et al. PDE5 modulates oocyte spontaneous maturation via cGMP-cAMP but not cGMP-PKG signaling. Front. Biosci. 2008;13:70877095.Google Scholar
Assou, S, Anahory, T, Pantesco, V, et al. The human cumulus-oocyte complex gene-expression profile. Hum. Reprod. 2006; 21:17051719.Google Scholar
Conti, M, Hsieh, M, Zamah, AM, Oh, JS. Novel signaling mechanisms in the ovary during oocyte maturation and ovulation. Mol. Cell. Endocrinol. 2012; 356:6573.Google Scholar
Robinson, JW, Zhang, M, Shuhaibar, LC, et al. Luteinizing hormone reduces the activity of the NPR2 guanylyl cyclase in mouse ovarian follicles, contributing to the cyclic GMP decrease that promotes resumption of meiosis in oocytes. Dev. Biol. 2012; 366:308316.Google Scholar
Hyslop, LA, Nixon, VL, Levasseur, M, et al. Ca(2+)-promoted cyclin B1 degradation in mouse oocytes requires the establishment of a metaphase arrest. Dev. Biol. 2004; 269:206219.Google Scholar
Bar-Ami, S, Zlotkin, E, Brandes, JM, Itskovitz-Eldor, J. Failure of meiotic competence in human oocytes. Biol. Reprod. 1994; 50:11001107.Google Scholar
Lincoln, AJ, Wickramasinghe, D, Stein, P, et al. Cdc25b phosphatase is required for resumption of meiosis during oocyte maturation. Nat. Genet. 2002; 30:446449.Google Scholar
Masciarelli, S, Horner, K, Liu, C, et al. Cyclic nucleotide phosphodiesterase 3A-deficient mice as a model of female infertility. J. Clin. Invest. 2004; 114:196205.Google Scholar
Fan, HY, Sun, QY, Zou, H. Regulation of separase in meiosis: separase is activated at the metaphase I-II transition in Xenopus oocytes during meiosis. Cell Cycle 2006; 5:198204.Google Scholar
Winston, NJ. Stability of cyclin B protein during meiotic maturation and the first mitotic cell division in mouse oocytes. Biol. Cell 1997; 89:211219.Google Scholar
Furuya, M, Tanaka, M, Teranishi, T, et al. H1foo is indispensable for meiotic maturation of the mouse oocyte. J. Reprod. Dev. 2007; 53:895902.Google Scholar
Nasmyth, K. How do so few control so many? Cell 2005; 120:739746.Google Scholar
Libby, BJ, De La Fuente, R, O’Brien, MJ, et al. The mouse meiotic mutation mei1 disrupts chromosome synapsis with sexually dimorphic consequences for meiotic progression. Dev. Biol. 2002; 242:174187.Google Scholar
Madgwick, S, Jones, K. How eggs arrest at metaphase II: MPF stabilisation plus APC/C inhibition equals cytostatic factor. Cell Div. 2007; 2:4.Google Scholar
Shoji, S, Yoshida, N, Amanai, M, et al. Mammalian Emi2 mediates cytostatic arrest and transduces the signal for meiotic exit via Cdc20. EMBO J. 2006; 25:834845.Google Scholar
Araki, K, Naito, K, Haraguchi, S, et al. Meiotic abnormalities of c-mos knockout mouse oocytes: activation after first meiosis or entrance into third meiotic metaphase. Biol. Reprod. 1996; 55:13151324.Google Scholar
Saunders, CM, Larman, MG, Parrington, J, et al. PLC zeta: a sperm-specific trigger of Ca(2+) oscillations in eggs and embryo development. Development 2002; 129:35333544.Google Scholar
Madgwick, S, Nixon, VL, Chang, HY, et al. Maintenance of sister chromatid attachment in mouse eggs through maturation-promoting factor activity. Dev. Biol. 2004; 275:6881.Google Scholar
Madgwick, S, Levasseur, M, Jones, KT. Calmodulin-dependent protein kinase II, and not protein kinase C, is sufficient for triggering cell-cycle resumption in mammalian eggs. J. Cell Sci. 2005; 118:38493859.Google Scholar
Kuliev, A, Verlinsky, Y. Meiotic and mitotic nondisjunction: lessons from preimplantation genetic diagnosis. Hum. Reprod. Update 2004; 10:401407.Google Scholar
Gull, I, Geva, E, Lerner-Geva, L, et al. Anaerobic glycolysis. The metabolism of the preovulatory human oocyte. Eur. J. Obstet. Gynecol. Reprod. Biol. 1999; 85:225228.Google Scholar
Sutton-McDowall, ML, Gilchrist, RB, Thompson, JG. The pivotal role of glucose metabolism in determining oocyte developmental competence. Reproduction 2010; 139:685695.Google Scholar
Cetica, P, Pintos, L, Dalvit, G, Beconi, M. Activity of key enzymes involved in glucose and triglyceride catabolism during bovine oocyte maturation in vitro. Reproduction 2002; 124:675681.Google Scholar
Sugiura, K, Su, YQ, Diaz, FJ, et al. Oocyte-derived BMP15 and FGFs cooperate to promote glycolysis in cumulus cells. Development 2007; 134:25932603.Google Scholar
Paczkowski, M, Silva, E, Schoolcraft, W, Krisher, R. Comparative importance of fatty acid beta-oxidation to nuclear maturation, gene expression, and glucose metabolism in mouse, bovine, and porcine cumulus oocyte complexes. Biol. Reprod. 2013; 88:111.Google Scholar
Dunning, KR, Anastasi, MR, Zhang, VJ, Russell, DL, Robker, RL. Regulation of fatty acid oxidation in mouse cumulus-oocyte complexes during maturation and modulation by PPAR agonists. PLoS One 2014; 9:e87327.Google Scholar
Bedaiwy, MA, Elnashar, SA, Goldberg, JM, et al. Effect of follicular fluid oxidative stress parameters on intracytoplasmic sperm injection outcome. Gynecol. Endocrinol. 2012; 28:5155.Google Scholar
Palini, S, Benedetti, S, Tagliamonte, MC, et al. Influence of ovarian stimulation for IVF/ICSI on the antioxidant defence system and relationship to outcome. RBM Online 2014; 29:6571.Google Scholar
Ellenbogen, A, Shavit, T, Shalom-Paz, E. IVM results are comparable and may have advantages over standard IVF. Facts Views Vis. Obgyn. 2014; 6:7780.Google Scholar
Flageole, C, Toufaily, C, Bernard, DJ, et al. Successful in vitro maturation of oocytes in a woman with gonadotropin-resistant ovary syndrome associated with a novel combination of FSH receptor gene variants: a case report. J. Assist. Reprod. Genet. 2019; 36:425432. doi:10.1007/s10815-018-1394-z.Google Scholar
Gremeau, AS, Andreadis, N, Fatum, M, et al. In vitro maturation or in vitro fertilization for women with polycystic ovaries? A case-control study of 194 treatment cycles. Fertil. Steril. 2012; 98:355360.Google Scholar
Guzmán, L, Adriaenssens, T, Ortega-Hrepich, C, et al. Human antral follicles <6 mm: a comparison between in vivo maturation and in vitro maturation in non-hCG primed cycles using cumulus cell gene expression. Mol. Hum. Reprod. 2013; 19:716.Google Scholar
Junk, SM, Yeap, D. Improved implantation and ongoing pregnancy rates after single-embryo transfer with an optimized protocol for in vitro oocyte maturation in women with polycystic ovaries and polycystic ovary syndrome. Fertil. Steril. 2012; 98:888892.Google Scholar
Sánchez, F, Lolicato, F, Romero, S, et al. An improved IVM method for cumulus-oocyte complexes from small follicles in polycystic ovary syndrome patients enhances oocyte competence and embryo yield. Hum. Reprod. 2017; 32:20562068.Google Scholar
Fadini, R, Dal Canto, MB, Renzini, MM, et al. Effect of different gonadotrophin priming on IVM of oocytes from women with normal ovaries: a prospective randomized study. RBM Online 2009; 19:343351.Google Scholar
Ortega-Hrepich, C, Stoop, D, Guzmán, L, et al. A “freeze-all” embryo strategy after in vitro maturation: a novel approach in women with polycystic ovary syndrome? Fertil. Steril. 2013; 100:10021007.Google Scholar
Walls, ML, Hunter, T, Ryan, JP, et al. In vitro maturation as an alternative to standard in vitro fertilization for patients diagnosed with polycystic ovaries: a comparative analysis of fresh, frozen and cumulative cycle outcomes. Hum. Reprod. 2015; 30:8896.Google Scholar
Menezo, Y, Nicollet, B, Rollet, J, Hazout, A. Pregnancy and delivery after in vitro maturation of naked ICSI-GV oocytes with GH and transfer of a frozen thawed blastocyst: case report. J. Assist. Reprod. Genet. 2006; 23:4749.Google Scholar
Roberts, R, Iatropoulou, A, Ciantar, D, et al. Follicle-stimulating hormone affects metaphase I chromosome alignment and increases aneuploidy in mouse oocytes matured in vitro. Biol. Reprod. 2005; 72:107118.Google Scholar
Chen, J, Torcia, S, Xie, F, et al. Somatic cells regulate maternal mRNA translation and developmental competence of mouse oocytes. Nat. Cell Biol. 2013; 15:14151423.Google Scholar
Trounson, A, Anderiesz, C, Jones, G. Maturation of human oocytes in vitro and their developmental competence. Reproduction 2001; 121:5175.Google Scholar
Camargo, LSA, Munk, M, Sales, JN, et al. Differential gene expression between in vivo and in vitro maturation: a comparative study with bovine oocytes derived from the same donor pool. JBRA Assist. Reprod. 2019; 23:714. doi:10.5935/1518-0557.20180084.Google Scholar
Jones, GM, Cram, DS, Song, B, et al. Gene expression profiling of human oocytes following in vivo or in vitro maturation. Hum. Reprod. 2008; 23:11381144.CrossRefGoogle ScholarPubMed
Coticchio, G, Dal Canto, M, Fadini, R, et al. Ultrastructure of human oocytes after in vitro maturation. Mol. Hum. Reprod. 2016; 22:110118.Google Scholar
De Vincentiis, S, De Martino, E, Buffone, MG, Brugo-Olmedo, S. Use of metaphase I oocytes matured in vitro is associated with embryo multinucleation. Fertil. Steril. 2013; 99:414421.Google Scholar
Margalit, T, Ben-Haroush, A, Garor, R, et al. Morphokinetic characteristics of embryos derived from in-vitro-matured oocytes and their in-vivo-matured siblings after ovarian stimulation. RBM Online 2019; 38:711.Google ScholarPubMed
Lee, HJ, Barad, DH, Kushnir, VA, et al. Rescue in vitro maturation (IVM) of immature oocytes in stimulated cycles in women with low functional ovarian reserve (LFOR). Endocrine 2016; 52:165171.Google Scholar
Zhang, XY, Ata, B, Son, WY, et al. Chromosome abnormality rates in human embryos obtained from in-vitro maturation and IVF treatment cycles. RBM Online 2010; 21:552559.Google Scholar
Chian, RC, Xu, CL, Huang, JY, Ata, B. Obstetric outcomes and congenital abnormalities in infants conceived with oocytes matured in vitro. Facts Views Vis. Obgyn. 2014; 6:1518.Google Scholar
Roesner, S, von Wolff, M, Elsaesser, M, et al. Two-year development of children conceived by IVM: a prospective controlled single-blinded study. Hum. Reprod. 2017; 32:13411350.Google Scholar
O’Brien, MJ, Pendola, JK, Eppig, JJ. A revised protocol for in vitro development of mouse oocytes from primordial follicles dramatically improves their developmental competence. Biol. Reprod. 2003; 68:16821686.Google Scholar
Telfer, EE, McLaughlin, M, Ding, C, Thong, KJ. A two-step serum-free culture system supports development of human oocytes from primordial follicles in the presence of activin. Hum. Reprod. 2008; 23:11511158.Google Scholar
Xu, M, Barrett, SL, West-Farrell, E, et al. In vitro grown human ovarian follicles from cancer patients support oocyte growth. Hum. Reprod. 2009; 24:25312540.Google Scholar
Xiao, S, Zhang, J, Romero, MM, et al. In vitro follicle growth supports human oocyte meiotic maturation. Sci. Rep. 2015; 5:17323.CrossRefGoogle ScholarPubMed
Cadoret, V, Frapsauce, C, Jarrier, P, et al. Molecular evidence that follicle development is accelerated in vitro compared to in vivo. Reproduction 2017; 153:493508.Google Scholar
McLaughlin, M, Albertini, DF, Wallace, WHB, Anderson, RA, Telfer, EE. Metaphase II oocytes from human unilaminar follicles grown in a multi-step culture system. Mol. Hum. Reprod. 2018; 24:135142.Google Scholar
Barrett, SB, Albertini, DF. Cumulus cell contact during oocyte maturation in mice regulates meiotic spindle positioning and enhances developmental potential. J. Assist. Reprod. Genet. 2010; 27:2939.Google Scholar
Coticchio, G, Guglielmo, MC, Dal Canto, M, et al. Mechanistic foundations of the metaphase II spindle of human oocytes matured in vivo and in vitro. Hum. Reprod. 2013; 28:32713282.Google Scholar
Li, J, Kawamura, K, Cheng, Y, et al. Activation of dormant ovarian follicles to generate mature eggs. Proc. Natl.Acad. Sci. U. S. A. 2010, 107:1028010284.Google Scholar
Reddy, P, Liu, L, Adhikari, D, et al. Oocyte-specific deletion of Pten causes premature activation of the primordial follicle pool. Science 2008; 319:611613.Google Scholar
Kawamura, K, Kawamura, N, Hsueh, A. Activation of dormant follicles: a new treatment for premature ovarian failure? Curr. Opin. Obstet. Gynecol. 2016; 28:217222.Google Scholar
Kawamura, K, Ishizuka, B, Hsueh, A. Drug-free in-vitro activation of follicles for infertility treatment in poor ovarian response patients with decreased ovarian reserve. RBM Online 2020; 40:245253.Google ScholarPubMed
Kawashima, I, Kawamura, K. Regulation of follicle growth through hormonal factors and mechanical cues mediated by Hippo signaling pathway. Syst. Biol. Reprod. Med. 2018; 64:311.Google Scholar
Fabregues, F, Ferreri, J, Calafell, JM, et al. Pregnancy after drug-free in vitro activation of follicles and fresh tissue autotransplantation in primary ovarian insufficiency patient: a case report and literature review. J. Ovarian Res. 2018; 11:76.Google Scholar

References

Stephen, EH, Chandra, A. Declining estimates of infertility in the United States: 1982–2002. Fertil. Steril. 2006; 86:516523.Google Scholar
Jarow, JP, Espeland, MA, Lipshultz, LI. Evaluation of the azoospermic patient. J. Urol. 1989; 142:6265.Google Scholar
Dohle, GR, Halley, DJ, Van Hemel, JO et al. Genetic risk factors in infertile men with severe oligozoospermia and azoospermia. Hum. Reprod. 2002; 17:1316.Google Scholar
Desai, N, Gill, P, Tadros, NN, et al. Azoospermia and embryo morphokinetics: testicular sperm-derived embryos exhibit delays in early cell cycle events and increased arrest prior to compaction. J. Assist. Reprod. Genet. 2018; 35:13391348.Google Scholar
Carpi, A, Sabanegh, E, Mechanick, J. Controversies in the management of nonobstructive azoospermia. Fertil. Steril. 2009; 91:963970.Google Scholar
Singer, R, Sagiv, M, Barnet, M, Levinsky, H. Semen volume and fructose content of human semen. Survey of the years 1980–1989. Acta Eur. Fertil. 1990; 21:205206.Google Scholar
Paick, J, Kim, SH, Kim, SW. Ejaculatory duct obstruction in infertile men. BJU Int. 2000; 85:720724.Google Scholar
Roberts, M, Jarvi, K. Steps in the investigation and management of low semen volume in the infertile man. Can. Urol. Assoc. J. 2009; 3:479485.Google Scholar
Schlegel, PN, Shin, D, Goldstein, M. Urogenital anomalies in men with congenital absence of the vas deferens. J. Urol. 1996; 155:16441648.Google Scholar
Hall, S, Oates, RD. Unilateral absence of the scrotal vas deferens associated with contralateral mesonephric duct anomalies resulting in infertility: laboratory, physical and radiographic findings, and therapeutic alternatives. J. Urol. 1983; 50:11611164.Google Scholar
Chillon, M, Casals, T, Mercier, B, et al. Mutations in the cystic fibrosis gene in patients with congenital absence of the vas deferens. N. Engl. J. Med. 1995; 332:14751480.Google Scholar
Yu, J, Chen, Z, Ni, Y, Li, Z. CFTR mutations in men with congenital bilateral absence of the vas deferens (CBAVD): a systemic review and meta-analysis. Hum. Reprod. 2012; 27:2535.Google Scholar
McCallum, T, Milunsky, J, Munarriz, R, et al. Unilateral renal agenesis associated with congenital bilateral absence of the vas deferens: phenotypic findings and genetic considerations. Hum. Reprod. 2001; 16:282288.Google Scholar
Schwarzer, JU, Schwarz, M. Significance of CFTR gene mutations in patients with congenital aplasia of vas deferens with special regard to renal aplasia. Andrologia 2012; 44:305307.Google Scholar
Meng, MV, Black, LD, Cha, I, et al. Impaired spermatogenesis in men with congenital absence of the vas deferens. Hum. Reprod. 2001; 16:529533.Google Scholar
Friedler, S, Raziel, A, Schachter, M, et al. Outcome of first and repeated testicular sperm extraction and ICSI in patients with non-obstructive azoospermia. Hum. Reprod. 2002; 17:23562361.Google Scholar
Matsumiya, K, Namiki, M, Takahara, S, et al. Clinical study of azoospermia. Int. J. Androl. 1994; 17:140142.Google Scholar
Faes, K, Goossens, E. Short-term storage of human testicular tissue: effect of storage temperature and tissue size. RBM Online 2017; 35:180188.Google Scholar
Krege, S, Beyer, J, Souchon, R, et al. European consensus on diagnosis and treatment of germ cell cancer: a report of the European Germ Cell Cancer Consensus Group (EGCCCG). Ann. Oncol. 2004; 15:13771399.Google Scholar
Honig, SC, Lipshulz, LI, Jarow, J. Significant medical pathology uncovered by a comprehensive male infertility evaluation. Fertil. Steril. 1994; 62:10281034.Google Scholar
Shefi, S, Turek, PJ. Definition and current evaluation of subfertile men. Int. Braz. J. Urol. 2006; 32:385397.Google Scholar
Dohle, GR, Elzanaty, S, van Casteren, NJ. Testicular biopsy: clinical practice and interpretation. Asian J. Androl. 2012; 14:8893.Google Scholar
Donoso, P, Tournaye, H, Devroey, P. Which is the best sperm retrieval technique for nonobstructive azoospermia? A systematic review. Hum. Reprod. Update 2007; 13:539549.Google Scholar
Schlegel, PN, Su, LM. Physiological consequences of testicular sperm extraction. Hum. Reprod. 1997; 12:16881692.Google Scholar
Pilatz, A, Rusz, A, Wagenlehner, F, Weidner, W, Altinkilic, B. Reference values for testicular volume, epididymal head size and peak systolic velocity of the testicular artery in adult males measured by ultrasonography. Ultraschall Med. 2013; 34:349354.Google Scholar
Tsirigotis, M, Bennett, V, Nicholson, N, et al. Experience with subzonal insemination (SUZI) and intracytoplasmic sperm injection (ICSI) on unfertilized aged human oocytes. J. Assist. Reprod. Genet. 1994; 11:389394.Google Scholar
Henkel, RR, Schill, WB. Sperm preparation for ART. Reprod. Biol. Endocrinol. 2003; 1:108122.Google Scholar
Kovacic, B, Vlaisavljevic, V, Reljic, M. Clinical use of pentoxifylline for activation of immotile testicular sperm before ICSI in patients with azoospermia. J. Androl. 2006; 27:4552.Google Scholar
Caroppo, E, Colpi, EM, Gazzano, G, et al. Testicular histology may predict the successful sperm retrieval in patients with non-obstructive azoospermia undergoing conventional TESE: a diagnostic accuracy study. J. Assist. Reprod. Genet. 2017; 34:149154.Google Scholar
Selice, R, Di Mambro, A, Garolla, A, et al. Spermatogenesis in Klinefelter syndrome. J. Endocrinol. Invest. 2010; 33:789793.Google Scholar
Ostad, M, Liotta, D, Ye, Z, Schlegel, PN. Testicular sperm extraction for nonobstructive azoospermia: results of a multibiopsy approach with optimized tissue dispersion. Urology 1998; 52:692696.Google Scholar
Schlegel, PN, Li, PS. Microdissection TESE: sperm retrieval in non-obstructive azoospermia. Hum. Reprod. Update 1998; 4:439.Google Scholar
Schlegel, PN. Testicular sperm extraction: microdissection improves sperm yield with minimal tissue excision. Hum. Reprod. 1999; 14:131135.Google Scholar
Turunc, T, Gul, U, Haydardedeoglu, B, et al. Conventional testicular sperm extraction combined with the microdissection technique in nonobstructive azoospermic patients: a prospective comparative study. Fertil. Steril. 2010; 94:21572160.Google Scholar
Jensen, CFS, Ohl, DA, Hiner, MR, et al. Multiple needle-pass percutaneous testicular sperm aspiration as first line treatment in azoospermic men. Andrology 2016; 4:257262.Google Scholar
Eliveld, J, van Wely, M, Meibner, A, et al. The risk of TESE-induced hypogonadism: a systematic review and meta-analysis. Hum. Reprod. Update 2018; 24:442454.Google Scholar
Kalsi, J, Thum, MY, Muneer, A, Abdullah, H, Minhas, S. In the era of micro-dissection sperm retrieval (m-TESE) is an isolated testicular biopsy necessary in the management of men with non-obstructive azoospermia? BJU Int. 2012; 109:418424.Google Scholar
Bernie, AM, Mata, DA, Ramasamy, R, Schlegel, PN. Comparison of microdissection testicular sperm extraction, conventional testicular sperm extraction, and testicular sperm aspiration for nonobstructive azoospermia: a systematic review and meta-analysis. Fertil. Steril. 2015; 104:10991103.Google Scholar
Deruyver, Y, Vanderschueren, D, Van der Aa, F. Outcome of microdissection TESE compared with conventional TESE in non-obstructive azoospermia: a systematic review. Andrology 2014; 2:2024.Google Scholar
Caroppo, E, Colpi, EM, Gazzano, G, et al. The seminiferous tubule caliber pattern as evaluated at high magnification during microdissection testicular sperm extraction predicts sperm retrieval in patients with nonobstructive azoospermia. Andrology 2019; 7:814.Google Scholar
Amer, M, Zohdy, W, Abd El Naser, T, et al. Single tubule biopsy: a new objective microsurgical advancement for testicular sperm retrieval in patients with nonobstructive azoospermia. Fertil. Steril. 2008; 89:592596.Google Scholar
Hussein, A, Ozgok, Y, Ross, L, Niederberger, C. Clomiphene administration for cases of nonobstructive azoospermia: a multicenter study. J. Androl. 2005; 26:787791; discussion 792–793.Google Scholar
Shiraishi, K, Ohmi, C, Shimabukuro, T, Matsuyama, H. Human chorionic gonadotrophin treatment prior to microdissection testicular sperm extraction in non-obstructive azoospermia. Hum. Reprod. 2012; 27:331339.Google Scholar
Reifsnyder, JE, Ramasamy, R, Husseini, J, Schlegel, PN. Role of optimizing testosterone before microdissection testicular sperm extraction in men with nonobstructive azoospermia. J. Urol. 2012; 188:532536.Google Scholar
Esteves, SC, Miyaoka, R, Roque, M, Agarwal, A. Outcome of varicocele repair in men with nonobstructive azoospermia: systematic review and meta-analysis. Asian J. Androl. 2016; 18:246253.Google Scholar
Kirby, EW, Wiener, LE, Rajanahally, S, Crowell, K, Coward, RM. Undergoing varicocele repair before assisted reproduction improves pregnancy rate and live birth rate in azoospermic and oligospermic men with a varicocele: a systematic review and meta-analysis. Fertil. Steril. 2016; 106:13381343.Google Scholar
Ramasamy, R, Yagan, N, Schlegel, PN. Structural and functional changes to the testis after conventional versus microdissection testicular sperm extraction. Urology 2005; 65:11901194.Google Scholar
Tournaye, H, Verheyen, G, Nagy, P, et al. Are there any predictive factors for successful testicular sperm recovery in azoospermic patients? Hum. Reprod. 1997; 12:8086.Google Scholar
Verhoeven, G, Cailleau, J. Follicle-stimulating hormone and androgens increase the concentration of the androgen receptor in Sertoli cells. Endocrinology 1988; 122:15411550.Google Scholar
Kim, ED, Crosnoe, L, Bar-Chama, N, Khera, M, Lipshultz, L. The treatment of hypogonadism in men of reproductive age. Fertil. Steril. 2013; 99:718724.Google Scholar
Pitteloud, N, Hayes, FJ, Dwyer, A, et al. Predictors of outcome of long-term GnRH therapy in men with idiopathic hypogonadotropic hypogonadism. J. Clin. Endocrinol. Metab. 2002; 87:41284136.Google Scholar
Adamopoulos, DA, Pappa, A, Billa, E, et al. Effectiveness of combined tamoxifen citrate and testosterone undecanoate treatment in men with idiopathic oligozoospermia. Fertil. Steril. 2003; 80:914920.CrossRefGoogle ScholarPubMed
Rodrigo, L, Rubio, C, Peinado, V, et al. Testicular sperm from patients with obstructive and nonobstructive azoospermia: aneuploidy risk and reproductive prognosis using testicular sperm from fertile donors as control samples. Fertil. Steril. 2011; 95:10051012.Google Scholar
Vozdova, M, Heracek, J, Sobotka, V, Rubes, J. Testicular sperm aneuploidy in non-obstructive azoospermic patients. Hum. Reprod. 2012; 27:22332239.Google Scholar
Schlegel, PN. Male infertility: evaluation and sperm retrieval. Clin. Obstet. Gynecol. 2006; 49:5572.Google Scholar
Samli, H, Samli, MM, Solak, M, Imirzalioglu, N. Genetic anomalies detected in patients with non-obstructive azoospermia and oligozoospermia. Arch. Androl. 2006; 52:263267.Google Scholar
Pryor, JL, Kent-First, M, Muallem, A, et al. Microdeletions in the Y chromosome of infertile men. N. Engl. J. Med. 1997; 336:534539.Google Scholar
Oates, RD, Silber, S, Brown, LG, Page, DC. Clinical characterization of 42 oligospermic or azoospermic men with microdeletion of the AZFc region of the Y chromosome, and of 18 children conceived via ICSI. Hum. Reprod. 2002; 17:28132824.Google Scholar
Hopps, CV, Mielnik, A, Goldstein, M, et al. Detection of sperm in men with Y chromosome microdeletions of the AZFa, AZFb and AZFc regions. Hum. Reprod. 2003; 18:16601665.CrossRefGoogle ScholarPubMed
Lee, SH, Ahn, SY, Lee, KW, et al. Intracytoplasmic sperm injection may lead to vertical transmission, expansion, and de novo occurrence of Y-chromosome microdeletions in male fetuses. Fertil. Steril. 2006; 85:15121515.Google Scholar
Practice Committee for the ASRM. Evaluation of the azoospermic male: a committee opinion. Fertil. Steril. 2018; 109:777782.Google Scholar

References

Maung, HH. Ethical problems with ethnic matching in gamete donation. J. Med. Ethics 2019; 45:112116.Google Scholar
Sauer, M, Paulson, R, Lobo, R. Pregnancy in women over 50 or more years: outcome of 22 consecutive established pregnancies from oocyte donation. Fertil. Steril. 1995; 64:111115.CrossRefGoogle ScholarPubMed
Isley, L, Falk, RE, Shamonki, J, Sims, CA, Callum, P. Management of the risks for inherited disease in donor-conceived offspring. Fertil. Steril. 2016;106:14791484.Google Scholar
Henneman, L, Borry, P, Chokoshvili, D, et al. Responsible implementation of expanded carrier screening. Eur. J. Hum. Genet. 2016; 24:e1e12.Google Scholar
Martin, J, Asan, , Yi, Y, et al. Comprehensive carrier genetic test using next-generation deoxyribonucleic acid sequencing in infertile couples wishing to conceive through assisted reproductive technology. Fertil. Steril. 2015; 104:12861293.Google Scholar
Amor, DJ, Kerr, A, Somanathan, N, et al. Attitudes of sperm, egg and embryo donors and recipients towards genetic information and screening of donors. Reprod. Health 2018; 15:26.Google Scholar
Kenney, NJ, McGowan, ML. Looking back: egg donors’ retrospective evaluations of their motivations, expectations, and experiences during their first donation cycle. Fertil. Steril. 2008; 93:455466.Google Scholar
Subak, LL, Adamson, GD, Boltz, NL. Therapeutic donor insemination: a prospective randomized trial of fresh versus frozen sperm. Am. J. Obstet. Gynecol. 1992; 166:15971604.Google Scholar
Amuzu, B, Laxova, R, Shapiro, SS. Pregnancy outcome, health of children, and family adjustment after donor insemination. Obstet. Gynecol. 1990; 75:899905.Google Scholar
Sauer, M. Reproductive prohibition: restricting donor payment will lead to medical tourism. Hum. Reprod. 1997; 12:18441845.Google Scholar
Williams, RA, Machin, LL. Rethinking gamete donor care: a satisfaction survey of egg and sperm donors in the UK. Plos One 2018; 13:e0199971. https://doi.org/10.1371/journal.pone.0199971.Google Scholar
American Society for Reproductive Medicine.Guidelines for gamete and embryo donation. Fertil. Steril. 1998; 70:1S4S.Google Scholar
Prateek, S, Sindhu, SG. Ethical and legal aspects in ART. In: Talwar, P, Sindhu, SG, eds., Step by Step Protocols in Clinical Embryology and ART. New Dehli: Jaypee Brothers Medical Publishers. 1995; 441461.Google Scholar
Linden, JV, Centola, G. New American Association of Tissue Banks standards for semen banking. Fertil. Steril. 1997; 68:597600.Google Scholar
Trounson, A, Leeton, J, Besanko, M, Wood, C, Conti, A. Pregnancy established in an infertile patient after transfer of a donated embryo fertilised in vitro. Br. Med. J. 1983; 286:835838.Google Scholar
Rosenwaks, Z. Donor eggs: their application in modern reproductive technologies. Fertil. Steril. 1987; 47:895909.Google Scholar
Sauer, MV, Paulson, RJ, Lobo, RA. A preliminary report on oocyte donation extending reproductive potential to women over 40. N. Engl. J. Med. 1990; 232:11571160.Google Scholar
Domingues, TS, Aquino, AP, Barros, B, et al. Egg donation of vitrified oocytes bank produces similar pregnancy rates by blastocyst transfer when compared to fresh cycle. J. Assist. Reprod. Genet. 2017; 34:15531557.Google Scholar
Van Katwijk, C, Peeters, LL. Clinical aspects of pregnancy after age of 35 years: a review of the literature. Hum. Reprod. Update 1998; 4:185194.Google Scholar
Sauer, M, Paulson, RJ, Lobo, R. Oocyte donation to women of advanced age: pregnancy results and obstetrical outcomes in patients 45 years and older. Hum. Reprod. 1996; 11:25402543.Google Scholar
Jacobsson, B, Ladfors, L, Milsom, I. Advanced maternal age and adverse perinatal outcome. Obstet. Gynecol. 2004; 104:727733.Google Scholar
Simchen, MJ, Yinon, Y, Moran, O, Schiff, E, Sivan, E. Pregnancy outcome after age 50. Obstet. Gynecol. 2006; 108:10841088.Google Scholar
Cleary-Goldman, J, Malone, FD, Vidaver, J, et al. Impact of maternal age on obstetric outcome. Obstet. Gynecol. 2005; 105:983990.Google Scholar
Joseph, KS, Allen, AC, Dodds, L, et al. The perinatal effects of delayed childbearing. Obstet. Gynecol. 2005; 105:14101418.Google Scholar
Schutte, JM, Schuitemaker, NW, Steegers, EA, van Roosmalen, J; Dutch Maternal Mortality Committee. Maternal death after oocyte donation at high maternal age: case report. Reprod. Health 2008; 5:12.Google Scholar
Cohen, J, Scott, R, Schimmel, T, Levron, J, Willadsen, S. Birth of infant donor after transfer of anucleate donor oocyte cytoplasm into recipient eggs. Lancet 1997; 350:186187.Google Scholar
Cohen, J, Scott, R, Alikani, M, et al. Ooplasmic transfer in mature human oocytes. Mol. Hum. Reprod. 1998; 4:269280.Google Scholar
Dale, B, Wilding, M, Botta, G, et al. Pregnancy after cytoplasmic transfer in a couple suffering from idiopathic infertility. Hum. Reprod. 2001; 16:14691472.Google Scholar
Soini, S, Ibarreta, D, Anastasiadou, V, et al. The interface between assisted reproductive technologies and genetics: technical, social, ethical and legal issues. Eur. J. Hum. Genet. 2006; 14:588645.Google Scholar
St John, J. The control of mtDNA replication during differentiation and development. Biochim. Biophys. Acta 2014; 1840:13451354.Google Scholar
Otten, AB, Smeets, HJ. Evolutionary defined role of the mitochondrial DNA in fertility, disease and ageing. Hum. Reprod. Update 2015; 21:671689.Google Scholar
Scheibye-Knudsen, M, Fang, EF, Croteau, DL, Wilson, DM III, Bohr, VA. Protecting the mitochondrial powerhouse. Trends Cell Biol. 2015; 25:158170.Google Scholar
Elliott, HR, Samuels, DC, Eden, JA, Relton, CL, Chinnery, PF. Pathogenic mitochondrial DNA mutations are common in the general population. Am. J. Hum. Genet. 2008; 83:254260.Google Scholar
Schaefer, AM, McFarland, R, Blakely, EL, et al. Prevalence of mitochondrial DNA disease in adults. Ann. Neurol. 2008; 63:3539.Google Scholar
Trushina, E, McMurray, CT. Oxidative stress and mitochondrial dysfunction in neurodegenerative diseases. Neuroscience 2007; 145:12331248.Google Scholar
Reeve, AK, Krishnan, KJ, Turnbull, D. Mitochondrial DNA mutations in disease, aging, and neurodegeneration. Ann. N. Y. Acad. Sci. 2008; 1147:2129.Google Scholar
Keating, DJ. Mitochondrial dysfunction, oxidative stress, regulation of exocytosis and their relevance to neurodegenerative diseases. J. Neurochem. 2008; 104:298305.CrossRefGoogle ScholarPubMed
Isasi, R, Kleiderman, E, Knoppers, BM. Genetic technology regulation. Editing policy to fit the genome? Science 2016; 351:337339.Google Scholar
Tachibana, M, Sparman, M, Sritanaudomchai, H, et al. Mitochondrial gene replacement in primate offspring and embryonic stem cells. Nature 2009; 461:367372.Google Scholar
Liu, CS, Chang, JC, Kuo, SJ, et al. Delivering healthy mitochondria for the therapy of mitochondrial diseases and beyond. Int. J. Biochem. Cell Biol. 2014; 53:141146.Google Scholar
Koopman, WJ, Willems, PH, Smeitink, JA. Monogenic mitochondrial disorders. N. Engl. J. Med. 2012; 366:11321141.Google Scholar
Wolf, DP, Mitalipov, N, Mitalipov, S. Mitochondrial replacement therapy in reproductive medicine. Trends Mol. Med. 2015; 21:6876.Google Scholar
Darbandi, S, Darbandi, M, Khorshid, HRK, et al. Experimental strategies towards increasing intracellular mitochondrial activity in oocytes: a systematic review. Mitochondrion 2016; 30:817.Google Scholar
Woods, DC, Tilly, JL. Autologous germline mitochondrial energy transfer (AUGMENT) in human assisted reproduction. Semin. Reprod. Med. 2015; 33:410421.Google Scholar
Fakih, MHSM, Szeptycki, J, dela Cruz, DB, et al. The AUGMENT treatment: physician reported outcomes of the initial global patient experience. JFIV Reprod. Med. Genet. 2015; 3:154.CrossRefGoogle Scholar
Boucret, L, Bris, C, Seegers, V, et al. Deep sequencing shows that oocytes are not prone to accumulate mtDNA heteroplasmic mutations during ovarian ageing. Hum. Reprod. 2017; 32:21012109.Google Scholar
Liu, S, Li, Y, Gao, X, Yan, JH, Chen, ZJ. Changes in the distribution of mitochondria before and after in vitro maturation of human oocytes and the effect of in vitro maturation on mitochondria distribution. Fertil. Steril. 2010; 93:15501555.Google Scholar
Wilding, M, Dale, B, Marino, M, et al. Mitochondrial aggregation patterns and activity in human oocytes and preimplantation embryos. Hum. Reprod. 2001; 16:909917.Google Scholar
Van Blerkom, J, Davis, P, Alexander, S. Differential mitochondrial distribution in human pronuclear embryos leads to disproportionate inheritance between blastomeres: relationship to microtubular organization, ATP content and competence. Hum. Reprod. 2000; 15:26212633.Google Scholar
Paull, D, Emmanuele, V, Weiss, KA, et al. Nuclear genome transfer in human oocytes eliminates mitochondrial DNA variants. Nature 2013; 493:632637.Google Scholar
Craven, L, Tuppen, HA, Greggains, GD, et al. Pronuclear transfer in human embryos to prevent transmission of mitochondrial DNA disease. Nature 2010; 465:8285.Google Scholar
Shenfield, F, Pennings, G, Cohen, J, et al. ESHRE Task Force on ethics and law 10: surrogacy. Hum. Reprod. 2005; 20:27052707.Google Scholar
Blake, L, Carone, N, Raffanello, E, et al. Gay fathers’ motivations for and feelings about surrogacy as a path to parenthood. Hum. Reprod. 2017; 32:860867.Google Scholar
Murphy, DA. The desire for parenthood: gay men choosing to become parents through surrogacy. J. Fam. Issues 2013; 34:11041124.Google Scholar
Lindenman, E, Shepard, MK, Pescovitz, OH. Müllerian agenesis: an update. Obstet. Gynecol. 1997; 90:307312.CrossRefGoogle ScholarPubMed
Beale, JM, Creighton, SM. Long-term health issues related to disorders or differences in sex development/intersex. Maturitas 2016; 94:143148.Google Scholar
Aflatoonian, N, Eftekhar, M, Aflatoonian, B, Rahmani, E, Aflatoonian, A. Surrogacy as a good option for treatment of repeated implantation failure: a case series. Iran. J. Reprod. Med. 2013; 11:7780.Google Scholar
Wang, AY, Dill, SK, Bowman, M, Sullivan, EA. Gestational surrogacy in Australia 2004–2011: treatment, pregnancy and birth outcomes. Aust. N. Z. J. Obstet. Gynaecol. 2016; 56:255259.Google Scholar
Blyth, E, Landau, R. (Eds.) Third Party Assisted Conception Across Cultures: Social, Legal and Ethical Perspectives. London; New York: Jessica Kingsley Publishers. 2004.Google Scholar
Thompson, C. Making Parents: The Ontological Choreography of Reproductive Technologies. Cambridge; London: MIT Press. 2005.Google Scholar
Jennings, S, Mellish, L, Tasker, F, Lamb, M, Golombok, S. Why adoption? Gay, lesbian, and heterosexual adoptive parents’ reproductive experiences and reasons for adoption. Adopt. Q 2014; 17:205226.Google Scholar

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×