Skip to main content Accessibility help
×
Hostname: page-component-8448b6f56d-mp689 Total loading time: 0 Render date: 2024-04-23T07:12:26.569Z Has data issue: false hasContentIssue false

Section 2 - Ovarian Hyperstimulation for IVF

Published online by Cambridge University Press:  14 April 2022

Mohamed Aboulghar
Affiliation:
University of Cairo IVF Centre
Botros Rizk
Affiliation:
University of South Alabama
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
Ovarian Stimulation , pp. 55 - 108
Publisher: Cambridge University Press
Print publication year: 2022

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

Baird, DT. Endocrinology of female infertility. Br Med Bull 1979;35:193198.CrossRefGoogle ScholarPubMed
Kerin, JF, Edmonds, DK, Warnes, GM, et al. Morphological and functional relations of Graafian follicle growth to ovulation in women using ultrasonic, laparoscopic and biochemical measurements. Br J Obstet Gynaecol 1981;88:8190.Google Scholar
Edwards, RG, Steptoe, PC. Current status of in-vitro fertilisation and implantation of human embryos. Lancet 1983;2:12651269.Google Scholar
Hackelöer, BJ, Fleming, R, Robinson, HP, Adam, AH, Coutts, JR. Correlation of ultrasonic and endocrinologic assessment of human follicular development. Am J Obstet Gynecol 1979;135:122128.Google Scholar
Schally, AV, Arimura, A, Baba, Y, et al. Isolation and properties of the FSH and LH-releasing hormone. Biochem Biophys Res Commun 1971;43(2):393399.Google Scholar
Smitz, J, Devroey, P, Braeckmans, P, et al. Management of failed cycles in an IVF/GIFT programme with the combination of GnRH analogue and HMG. Hum Reprod 1987;2(4):309314.Google Scholar
Gemzell, CA, Kemman, E, Jones, JR. Premature ovulation during administration of human menopausal gonadotropins in non-ovulatory women. Infertility 1978;1:110.Google Scholar
Fraser, HM, Laird, NC, Blakeley, DM. Decreased pituitary responsiveness and inhibition of the luteinizing hormone surge and ovulation in the stumptailed monkey (Macaca arctoides) by chronic treatment with an agonist of luteinizing hormone-releasing hormone. Endocrinology 1980;106:452457.Google Scholar
Fleming, R, Adam, AH, Barlow, DH, et al. A new systematic treatment for infertile women with abnormal hormone profiles. Br J Obstet Gynaecol 1982;89:8083.CrossRefGoogle ScholarPubMed
Porter, RN, Smith, W, Craft, IL, Abdulwahid, NA, Jacobs, HS. Induction of ovulation for in-vitro fertilisation using buserelin and gonadotropins. Lancet 1984;2:12841285.Google Scholar
Fleming, R, Haxton, MJ, Hamilton, MP, et al. Combined gonadotropin-releasing hormone analog and exogenous gonadotropins for ovulation induction in infertile women: efficacy related to ovarian function assessment. Am J Obstet Gynecol 1988;159:376381.Google Scholar
Shalev, E, Leung, PC. Gonadotropin-releasing hormone and reproductive medicine. J Obstet Gynaecol Can 2003;25(2):98113.Google Scholar
Diedrich, K, Ludwig, M, Felberbaum, RE. The role of gonadotropin-releasing hormone antagonists in in vitro fertilization. Semin Reprod Med 2001;19(3):213220.Google Scholar
Conn, PM, Crowley, WF Jr. Gonadotropin-releasing hormone and its analogues. N Engl J Med 1991;324(2):93103.Google Scholar
Sandow, J, Engelbart, K, von Rechenberg, W. The different mechanisms for suppression of pituitary and testicular function. Med Biol 1985;63:192200.Google Scholar
Pelletier, G, Dubé, D, Guy, J, Séguin, C, Lefebvre, FA. Binding and internalization of a luteinizing hormone-releasing hormone agonist by rat gonadotrophic cells. A radio autographic study. Endocrinology 1982;111:10681076.Google Scholar
Filicori, M, Cognigni, GE, Arnone, R, et al. Role of different GnRH agonist regimens in pituitary suppression and the outcome of controlled ovarian hyperstimulation. Hum Reprod 1996;11 Suppl 3:123132.CrossRefGoogle ScholarPubMed
Janssens, RM, Lambalk, CB, Vermeiden, JP, et al. Dose-finding study of triptorelin acetate for prevention of a premature LH surge in IVF: a prospective, randomized, double-blind, placebo-controlled study. Hum Reprod 2000;15:23332340.CrossRefGoogle ScholarPubMed
Dada, T, Salha, O, Baillie, HS, Sharma, V. A comparison of three gonadotrophin-releasing hormone analogues in an in-vitro fertilization programme: a prospective randomized study. Hum Reprod 1999;14(2):288293.Google Scholar
El-Nemr, A, Bhide, M, Khalifa, Y, et al. Clinical evaluation of three different gonadotrophin-releasing hormone analogues in an IVF programme: a prospective study. Eur J Obstet Gynecol Reprod Biol 2002;103(2):140145.Google Scholar
Eftekhar, M, Rahmani, E, Mohammadian, F. Comparison of pregnancy outcome in half-dose Triptorelin and short-acting Decapeptyl in long protocol in ART cycles: a randomized clinical trial. Iran J Reprod Med 2013;11(2):133138.Google Scholar
Bastu, E, Buyru, F, Ozsurmeli, M, et al. A randomized, single-blind, prospective trial comparing three different gonadotropin doses with or without addition of letrozole during ovulation stimulation in patients with poor ovarian response. Eur J Obstet Gynecol Reprod Biol 2016;203:3034.Google Scholar
Albuquerque, LE, Tso, LO, Saconato, H, Albuquerque, MC, Macedo, CR. Depot versus daily administration of gonadotrophin‐releasing hormone agonist protocols for pituitary down regulation in assisted reproduction cycles. Cochrane Database Syst Rev 2013;1:CD002808.Google Scholar
Lockwood, GM, Pinkerton, SM, Barlow, DH. A prospective randomized single-blind comparative trial of nafarelin acetate with buserelin in long-protocol gonadotrophin-releasing hormone analogue controlled in-vitro fertilization cycles. Hum Reprod 1995;10:293298.Google Scholar
Meldrum, DR, Wisot, A, Hamilton, F, et al. Timing of initiation and dose schedule of leuprolide influence the time course of ovarian suppression. Fertil Steril 1988;50(3):400402.Google Scholar
Siristatidis, CS, Gibreel, A, Basios, G, Maheshwari, A, Bhattacharya, S. Gonadotrophin‐releasing hormone agonist protocols for pituitary suppression in assisted reproduction. Cochrane Database Syst Rev 2015;11:CD006919.Google Scholar
Berkkanoglu, M, Ozgur, K. What is the optimum maximal gonadotropin dosage used in microdose flare-up cycles in poor responders? Fertil Steril 2010;94(2):662665.Google Scholar
Lefebvre, J, Antaki, R, Kadoch, IJ, et al. 450 IU versus 600 IU gonadotropin for controlled ovarian stimulation in poor responders: a randomized controlled trial. Fertil Steril 2015;104(6):14191425.CrossRefGoogle ScholarPubMed
National Institute for Health and Care Excellence. Fertility problems: assessment and treatment (CG156). 2017.Google Scholar
van Wely, M, Westergaard, LG, Bossuyt, PM, van der Veen, F. Effectiveness of human menopausal gonadotropin versus recombinant follicle-stimulating hormone for controlled ovarian hyperstimulation in assisted reproductive cycles: a meta-analysis. Fertil Steril 2003;80(5):10861093.Google Scholar
Coomarasamy, A, Afnan, M, Cheema, D, et al. Urinary hMG versus recombinant FSH for controlled ovarian hyperstimulation following an agonist long down-regulation protocol in IVF or ICSI treatment: a systematic review and meta-analysis. Hum Reprod 2008;23(2):310315.Google Scholar
Tan, SL, Balen, A, El Hussein, E, et al. A prospective randomized study of the optimum timing of human chorionic gonadotropin administration after pituitary desensitization in in vitro fertilization. Fertil Steril 1992;57(6):12591264.Google Scholar
Youssef, MA, Abou‐Setta, AM, Lam, WS. Recombinant versus urinary human chorionic gonadotrophin for final oocyte maturation triggering in IVF and ICSI cycles. Cochrane Database Syst Rev 2016;4:CD003719.Google Scholar
Nelson, SM, Yates, RW, Lyall, H, et al. Anti-Müllerian hormone-based approach to controlled ovarian stimulation for assisted conception. Hum Reprod 2009;24:867875.Google Scholar
Frydman, R, Belaisch-Allart, J, Parneix, I, et al. Comparison between flare up and down regulation effects of luteinizing hormone-releasing hormone agonists in an in vitro fertilization program. Fertil Steril 1988;50:471475.Google Scholar
Cramer, DW, Powers, DR, Oskowitz, SP, et al. Gonadotropin-releasing hormone agonist use in assisted reproduction cycles: the influence of long and short regimens on pregnancy rates. Fertil Steril 1999;72:8389.Google Scholar
San Roman, GA, Surrey, ES, Judd, HL, Kerin, JF. A prospective randomized comparison of luteal phase versus concurrent follicular phase initiation of gonadotropin-releasing hormone agonist for in vitro fertilization. Fertil Steril 1992;58(4):744749.Google Scholar
Gelety, TJ, Pearlstone, AC, Surrey, ES. Short-term endocrine response to gonadotropin-releasing hormone agonist initiated in the early follicular, midluteal, or late luteal phase in normally cycling women. Fertil Steril 1995;64(6):10741080.CrossRefGoogle ScholarPubMed
Keltz, MD, Jones, EE, Duleba, AJ, et al. Baseline cyst formation after luteal phase gonadotropin-releasing hormone agonist administration is linked to poor in vitro fertilization outcome. Fertil Steril 1995;64(3):568572.Google Scholar
Feldberg, D, Ashkenazi, J, Dicker, D, et al. Ovarian cyst formation: a complication of gonadotropin-releasing hormone agonist therapy. Fertil Steril 1989;51(1):4245.CrossRefGoogle ScholarPubMed
Ron-El, R, Herman, A, Golan, A, et al. Follicle cyst formation following long-acting gonadotropin-releasing hormone analog administration. Fertil Steril 1989;52(6):10631066.CrossRefGoogle ScholarPubMed
Qublan, HS, Amarin, Z, Tahat, YA, Smadi, AZ, Kilani, M. Ovarian cyst formation following GnRH agonist administration in IVF cycles: incidence and impact. Hum Reprod 2006;21(3):640644.Google Scholar
Firouzabadi, RD, Sekhavat, L, Javedani, M. The effect of ovarian cyst aspiration on IVF treatment with GnRH. Arch Gynecol Obstet 2010;281(3):545549.Google Scholar
Segal, S, Shifren, JL, Isaacson, KB, et al. Effect of a baseline ovarian cyst on the outcome of in vitro fertilization–embryo transfer. Fertil Steril 1999;71(2):274277.Google Scholar
Rizk, B, Tan, SL, Kingsland, C, et al. Ovarian cyst aspiration and the outcome of in vitro fertilization. Fertil Steril 1990;54(4):661664.Google Scholar
Cédrin-Durnerin, I, Bulwa, S, Hervé, F, et al. The hormonal flare-up following gonadotrophin-releasing hormone agonist administration is influenced by a progestogen pretreatment. Hum Reprod 1996;11:18591863.Google Scholar
Sbracia, M, Farina, A, Poverini, R, et al. Short versus long gonadotropin-releasing hormone analogue suppression protocols for superovulation in patients > or = 40 years old undergoing intracytoplasmic sperm injection. Fertil Steril 2005;84:644648.CrossRefGoogle ScholarPubMed
Pandian, Z, McTavish, AR, Aucott, L, Hamilton, MP, Bhattacharya, S. Interventions for ‘poor responders’ to controlled ovarian hyper stimulation (COH) in in‐vitro fertilisation (IVF). Cochrane Database Syst Rev 2010;1:CD004379.Google Scholar
Berin, I, Stein, DE, Keltz, MD. A comparison of gonadotropin-releasing hormone (GnRH) antagonist and GnRH agonist flare protocols for poor responders undergoing in vitro fertilization. Fertil Steril 2010;93:360363.CrossRefGoogle ScholarPubMed
Ron-El, R, Golan, A, Herman, A, et al. Midluteal gonadotropin-releasing hormone analog administration in early pregnancy. Fertil Steril 1990;53(3):572574.Google Scholar
Isherwood, PJ, Ibrahim, ZH, Matson, PL, et al. Endocrine changes in women conceiving during treatment with an LHRH agonist. Hum Reprod 1990;5(4):409412.Google Scholar
Smitz, J, Camus, M, Devroey, P, Bollen, N, Tournaye, H, Van Steirteghem, AC. CLINICAL REPORT: The influence of inadvertent intranasal buserelin administration in early pregnancy. Hum Reprod 1991;6(2):290293.Google Scholar
Balasch, J, Martinez, F, Jové, I, et al. Inadvertent gonadotrophin-releasing hormone agonist (GnRHa) administration in the luteal phase may improve fecundity in in-vitro fertilization patients. Hum Reprod 1993;8(7):11481151.Google Scholar
Weissman, A, Shoham, Z. Favourable pregnancy outcome after administration of a long-acting gonadotrophin-releasing hormone agonist in the mid-luteal phase. Hum Reprod 1993;8(3):496497.Google Scholar
Janssens, RM, Brus, L, Cahill, DJ, et al. Direct ovarian effects and safety aspects of GnRH agonists and antagonists. Hum Reprod Update 2000;6:505518.Google Scholar
Delvigne, A, Rozenberg, S. Epidemiology and prevention of ovarian hyperstimulation syndrome (OHSS): a review. Hum Reprod Update 2002;8(6):559577.Google Scholar
Ragni, G, Vegetti, W, Riccaboni, A, et al. Comparison of GnRH agonists and antagonists in assisted reproduction cycles of patients at high risk of ovarian hyperstimulation syndrome. Hum Reprod 2005;20(9):24212425.Google Scholar
Fleming, R. Recruitment prior to ovarian stimulation: ways of improving follicular recruitment. RBM Online 2005;10 Suppl 3:5559.Google Scholar
Huirne, JA, Homburg, R, Lambalk, CB. Are GnRH antagonists comparable to agonists for use in IVF? Hum Reprod 2007;22:28052813.Google Scholar
Fleming, R, Deshpande, N, Traynor, I, Yates, RW. Dynamics of FSH-induced follicular growth in subfertile women: relationship with age, insulin resistance, oocyte yield and anti-Mullerian hormone. Hum Reprod 2006;21:14361441.Google Scholar
Devroey, P, Aboulghar, M, Garcia-Velasco, J, et al. Improving the patient’s experience of IVF/ICSI: a proposal for an ovarian stimulation protocol with GnRH antagonist co-treatment. Hum Reprod 2009;24:764774.CrossRefGoogle ScholarPubMed
Nelson, SM, Yates, RW, Fleming, R. Serum anti-Müllerian hormone and FSH: prediction of live birth and extremes of response in stimulated cycles-implications for individualization of therapy. Hum Reprod 2007;22:24142421.Google Scholar
Anckaert, E, Smitz, J, Schiettecatte, J, Klein, BM, Arce, JC. The value of anti-Müllerian hormone measurement in the long GnRH agonist protocol: association with ovarian response and gonadotrophin-dose adjustments. Hum Reprod 2012;27(6):18291839.Google Scholar
Fleming, R, Broekmans, F, Calhaz-Jorge, C, et al. Can anti-Müllerian hormone concentrations be used to determine gonadotrophin dose and treatment protocol for ovarian stimulation? Reprod Biomed Online 2013;26(5):431439.Google Scholar
Lambalk, CB, Banga, FR, Huirne, JA, et al. GnRH antagonist versus long agonist protocols in IVF: a systematic review and meta-analysis accounting for patient type. Hum Reprod Update 2017;23(5):560579.Google Scholar
The ESHRE Guideline Group on Ovarian Stimulation, Bosch, E, Broer, S, Griesinger, G, et al. ESHRE guideline: ovarian stimulation for IVF/ICSI. Hum Reprod Open 2020;2020(2):hoaa009.Google Scholar
Aboulghar, M. Luteal support in reproduction: when, what and how? Curr Opin Obstet Gynecol 2009;21:279284.CrossRefGoogle ScholarPubMed
Jee, BC, Suh, CS, Kim, SH, Kim, YB, Moon, SY. Effects of estradiol supplementation during the luteal phase of in vitro fertilization cycles: a meta-analysis. Fertil Steril 2010;93(2):428436.Google Scholar

References

Porter, RN, Smith, W, Craft, IL, Abdulwahid, NA, Jacobs, HS. Induction of ovulation for in-vitro fertilization using buserelin and gonadotrophins. Lancet 1984;2:12841285.Google Scholar
Diedrich, K, Diedrich, C, Santos, E, et al. Suppression of the endogenous luteinizing hormone surge by the gonadotrophin-releasing hormone antagonist cetrorelix during ovarian stimulation. Hum Reprod 1994;9(5):788791.CrossRefGoogle ScholarPubMed
Ron-El, R, Raziel, A, Schachter, M, et al. Induction of ovulation after GnRH antagonists. Hum Reprod Update 2000;6(4):318321.CrossRefGoogle ScholarPubMed
Gordon, K, Williams, RF, Danforth, DR, Hodgen, GD. The combined use of GnRH antagonists with gonadotrophins or pulsatile GnRH in ovulation induction. In: Bouchard, P, Caraty, A, Coelingh-Bennink, HJT, Pavlou, SN, eds. GnRH-analogs, Gonadotrophins and Gonadal Peptides. London, UK Parthenon Publishing Group; 1992:239 pp.Google Scholar
Hughes, EG, Fedorkow, DM, Daya, S, et al. The routine use of gonadotropin-releasing hormone agonists prior to in vitro fertilization and gamete intrafallopian transfer: a meta-analysis of randomized controlled trials. Fertil Steril 1992;58(5):888896.CrossRefGoogle ScholarPubMed
Clayton, RN, Catt, KJ. Gonadotrophin-releasing hormone receptors: characterization, physiological regulation, and relationship to reproductive function. Endocr Rev 1981;2:186209.Google Scholar
Clayton, RN, Catt, KJ. Receptor-binding affinity of gonadotropin-releasing hormone analogs: analysis by radioligand-receptor assay. Endocrinology 1980;106(4):11541159.Google Scholar
Coy, DH, Labrie, F, Savary, M, Coy, EJ, Schally, AV. LH-releasing activity of potent LH-RH analogs in vitro. Biochem Biophys Res Commun 1975;67(2):576582.Google Scholar
Ditkoff, EC, Cassidenti, DL, Paulson, RJ, et al. The gonadotropin-releasing hormone antagonist (Nal-Glu) acutely blocks the luteinizing hormone surge but allows for resumption of folliculogenesis in normal women. Am J Obstet Gynecol 1991;165:18111817Google Scholar
Kenigsberg, D, Hodgen, GD. Ovulation inhibition by administration of weekly gonadotropin-releasing hormone antagonist. J Clin Endocrinol Metab 1986;62(4):734738.Google Scholar
Matikainen, T, Ding, YQ, Vergara, M, et al. Differing responses of plasma bioactive and immunoreactive follicle-stimulating hormone and luteinizing hormone to gonadotropin-releasing hormone antagonist and agonist treatments in postmenopausal women. J Clin Endocrinol Metab 1992;75(3):820825.Google Scholar
Bouchard, PG, Charbonnel, B, Caraty, A, et al. The role of LHRH during the periovulatory period: a basis for the use of LHRH antagonists in ovulation induction. In: Filicori, M, Flamigni, C, eds. Ovulation: Basic Science and Clinical Advances. Elsevier Science BV International Congress series 1046. 1994.Google Scholar
The ganirelix dose-finding study group. A double-blind, randomized, dose-finding study to assess the efficacy of the gonadotrophin-releasing hormone antagonist ganirelix (Org 37462) to prevent premature luteinizing hormone surges in women undergoing ovarian stimulation with recombinant follicle stimulating hormone (Puregon). Hum Reprod 1998;13(11):30233031.Google Scholar
Olivennes, F, Alvarez, S, Bouchard, P, et al. The use of a GnRH antagonist (cetrorelix) in a single dose protocol in IVF-embryo transfer: a dose finding study of 3 versus 2 mg. Hum Reprod 1998;13(9):24112414.Google Scholar
Olivennes, F. Cunha-Filho, JS, Fanchin, R, Bouchard, P, Frydman, R. The use of GnRH antagonists in ovarian stimulation. Hum Reprod Update 2002;8:279290.Google Scholar
Al-Inany, H, Aboulghar, M. Gonadotrophin-releasing hormone antagonist for assisted conception. Cochrane Database Syst Rev 2001;4:CD001750.Google Scholar
Kolibianakis, EM, Collins, J, Tarlatzis, BC, et al. Among patients treated for IVF with gonadotrophins and GnRH analogues, is the probability of live birth dependent on the type of analogue used? A systematic review and meta-analysis. Hum Reprod Update 2006;12(6):651671.Google Scholar
Al-Inany, HG, Abou-Setta, AM, Aboulghar, M. Gonadotrophin-releasing hormone antagonists for assisted conception. Cochrane Database Syst Rev 2006;3:CD001750.Google Scholar
Al-Inany, HG, Youssef, MA, Ayeleke, RO, et al. Gonadotrophin-releasing hormone antagonists for assisted reproductive technology. Cochrane Database Syst Rev 2016;4:CD001750.Google Scholar
Lambalk, BC, Banga, FR, Huirne, JA, et al. GnRH antagonist versus long agonist protocols in IVF: a systematic review and meta-analysis accounting for patient type. Hum Reprod Update 2017;23(5):560579.Google Scholar
Sunkara, SK, Coomarasamy, A, Faris, R, Braude, P, Khalaf, Y. Long gonadotropin-releasing hormone agonist versus short agonist versus antagonist regimens in poor responders undergoing in vitro fertilization: a randomized controlled trial. Fertil Steril 2014;101(1):147153.CrossRefGoogle ScholarPubMed
Toftager, M, Bogstad, J, Løssl, K, et al. Cumulative live birth rates after one ART cycle including all subsequent frozen-thaw cycles in 1050 women: secondary outcome of an RCT comparing GnRH-antagonist and GnRH-agonist protocols. Hum Reprod 2017;32(3):556567.Google Scholar
Ludwig, M, Riethmüller-Winzen, H, Felberbaum, RE, et al. Health of 227 children born after controlled ovarian stimulation for in vitro fertilization using the luteinizing hormone-releasing hormone antagonist cetrorelix. Fertil Steril 2001;75(1):1822.Google Scholar
Ludwig, M, Katalinic, A, Banz, C, et al. Tailoring the GnRH antagonist cetrorelix acetate to individual patients’ needs in ovarian stimulation for IVF: results of a prospective, randomized study. Hum Reprod 2002;17(11):28422845.CrossRefGoogle ScholarPubMed
Mansour, RT, Aboulghar, MA, Serour, GI, et al. The use of gonadotropin-releasing hormone antagonist in a flexible protocol: a pilot study. Am J Obstet Gynecol 2003;189(2):444446.Google Scholar
Lainas, T, Zorzovilis, J, Petsas, G, et al. In a flexible antagonist protocol, earlier, criteria-based initiation of GnRH antagonist is associated with increased pregnancy rates in IVF. Hum Reprod 2005;20(9):24262433.CrossRefGoogle Scholar
Al-Inany, H, Aboulghar, MA, Mansour, RT, Serour, GI. Optimizing GnRH antagonist administration: meta-analysis of fixed versus flexible protocol. Reprod Biomed Online 2005;10(5):567570.Google Scholar
Messinis, IE, Loutradis, D, Domali, E, et al. Alternate day and daily administration of GnRH antagonist may prevent premature luteinization to a similar extent during FSH treatment. Hum Reprod 2005;20(11):31923197.Google Scholar
Aboulghar, MA, Mansour, RT, Serour, GI, et al. Increasing the dose of human menopausal gonadotrophins on day of GnRH antagonist administration: randomized controlled trial. Reprod Biomed Online 2004;8(5):524527.Google Scholar
Propst, AM, Bates, GW, Robinson, RD, et al. A randomized controlled trial of increasing recombinant follicle-stimulating hormone after initiating a gonadotropin-releasing hormone antagonist for in vitro fertilization-embryo transfer. Fertil Steril 2006;86(1):5863.Google Scholar
Engel, JB, Ludwig, M, Felberbaum, R, et al. Use of cetrorelix in combination with clomiphene citrate and gonadotrophins: a suitable approach to ‘friendly IVF’? Hum Reprod 2002;17(8):20222026.Google Scholar
Mansour, R, Aboulghar, M, Serour, GI, et al. The use of clomiphene citrate/human menopausal gonadotrophins in conjunction with GnRH antagonist in an IVF/ICSI program is not a cost effective protocol. Acta Obstet Gynecol Scand 2003;82(1):4852.CrossRefGoogle Scholar
Barmat, LI, Chantilis, SJ, Hurst, BS, Dickey, RP. A randomized prospective trial comparing gonadotropin-releasing hormone (GnRH) antagonist/recombinant follicle-stimulating hormone (rFSH) versus GnRH-agonist/rFSH in women pretreated with oral contraceptives before in vitro fertilization. Fertil Steril 2005;83(2):321330.Google Scholar
Bendikson, K, Milki, AA, Speck-Zulak, A, Westphal, LM. Comparison of GnRH antagonist cycles with and without oral contraceptive pretreatment in potential poor prognosis patients. Clin Exp Obstet Gynecol 2006;33(3):145147.Google Scholar
Pinkas, H, Sapir, O, Avrech, OM, et al. The effect of oral contraceptive pill for cycle scheduling prior to GnRH-antagonist protocol on IVF cycle parameters and pregnancy outcome. J Assist Reprod Genet 2008;25(1):2933.Google Scholar
Kim, CH, Jeon, GH, Cheon, YP, et al. Comparison of GnRH antagonist protocol with or without oral contraceptive pill pretreatment and GnRH agonist low-dose long protocol in low responders undergoing IVF/intracytoplasmic sperm injection. Fertil Steril 2009;92(5):17581760.CrossRefGoogle ScholarPubMed
Huirne, JA, van Loenen, AC, Donnez, J, et al. Effect of an oral contraceptive pill on follicular development in IVF/ICSI patients receiving a GnRH antagonist: a ranmdomized study. Reprod Biomed Online 2006;13:235245.Google Scholar
Kolibianakis, EM, Papanikolaou, EG, Camus, M, et al. Effect of oral contraceptive pill pretreatment on ongoing pregnancy rates in patients stimulated with GnRH antagonists and recombinant FSH for IVF. A randomized controlled trial. Hum Reprod 2006;21(2):352357.Google Scholar
Griesinger, G, Venetis, CA, Marx, T, et al. Oral contraceptive pill pretreatment in ovarian stimulation with GnRH antagonists for IVF: a systematic review and meta-analysis. Fertil Steril 2008;90(4):10551063.Google Scholar
Farquhar, C, Rombauts, L, Kremer, JA, Lethaby, A, Ayeleke, RO. Oral contraceptive pill, progestogen or oestrogen pretreatment for ovarian stimulation protocols for women undergoing assisted reproductive techniques. Cochrane Database Syst Rev 2017;5:CD006109.Google Scholar
Cavagna, M, Louzada Maldonado, LG, de Souza Bonetti, TC, et al. Supplementation with a recombinant human chorionic gonadotropin microdose leads to similar outcomes in ovarian stimulation with recombinant follicle-stimulating hormone for intracytoplasmic sperm injection cycles using either a gonadotropin-releasing hormone agonist or antagonist for pituitary suppression. Fertil Steril 2010;94(1):167172.Google Scholar
Griesinger, G, Schultze-Mosgau, A, Dafopoulos, K, et al. Recombinant luteinizing hormone supplementation to recombinant follicle-stimulating hormone induced ovarian hyperstimulation in the GnRH-antagonist multiple-dose protocol. Hum Reprod 2005;20(5):12001206.Google Scholar
Cédrin-Durnerin, I, Grange-Dujardin, D, Laffy, A, et al. Recombinant human LH supplementation during GnRH antagonist administration in IVF/ICSI cycles: a prospective randomized study. Hum Reprod 2004;19(9):19791984.Google Scholar
Rongières-Bertrand, C, Olivennes, F, Righini, C, et al. Revival of the natural cycles in in-vitro fertilization with the use of a new gonadotrophin-releasing hormone antagonist (cetrorelix): a pilot study with minimal stimulation. Hum Reprod 1999;14(3):683688.CrossRefGoogle ScholarPubMed
Pelinck, MJ, Vogel, NE, Arts, EG, et al. Cumulative pregnancy rates after a maximum of nine cycles of modified natural cycle IVF and analysis of patient drop-out: a cohort study. Hum Reprod 2007;22(9):24632470.Google Scholar
Humaidan, P, Kol, S, Papanikolaou, EG. GnRH agonist for triggering of final oocyte maturation: time for a change of practice? Hum Reprod Update 2011;17(4):510524.Google Scholar
Humaidan, P, Papanikolaou, EG, Kyrou, D, et al. The luteal phase after GnRH-agonist triggering of ovulation: present and future perspectives. Reprod Biomed Online 2012;24(2):134141.Google Scholar
Fauser, BC, de Jong, D, Olivennes, F, et al. Endocrine profiles after triggering of final oocyte maturation with GnRH agonist after cotreatment with the GnRH antagonist ganirelix during ovarian hyperstimulation for in vitro fertilization. J Clin Endocrinol Metab 2002;87(2):709715.Google Scholar
Griesinger, G, Diedrich, K, Devroey, P, Kolibianakis, EM. GnRH agonist for triggering final oocyte maturation in the GnRH antagonist ovarian hyperstimulation protocol: a systematic review and meta-analysis. Hum Reprod Update 2006;12(2):159168.Google Scholar
Humaidan, P, Bungum, L, Bungum, M, Yding Andersen, C. Rescue of corpus luteum function with peri-ovulatory HCG supplementation in IVF/ICSI GnRH antagonist cycles in which ovulation was triggered with a GnRH agonist: a pilot study. Reprod Biomed Online 2006;13(2):173178.Google Scholar
Youssef, MA, Van der Veen, F, Al-Inany, HG, et al. Gonadotropin-releasing hormone agonist versus HCG for oocyte triggering in antagonist-assisted reproductive technology. Cochrane Database Syst Rev 2014;10:CD008046.Google Scholar
Leth-Moller, K, Hammer Jagd, S, Humaidan, P. The luteal phase after GnRHa trigger-understanding an enigma. Int J Fertil Steril 2014;8(3):227234.Google Scholar
Elgindy, EA, Sibai, H, Mostafa, MI, et al. Towards an optimal luteal support modality in agonist triggered cycles: a randomized clinical trial. Hum Reprod 2018;33(6):10791086.Google Scholar
Franco, JG Jr., Baruffi, RL, Mauri, AL, et al. GnRH agonist versus GnRH antagonist in poor ovarian responders: a meta-analysis. Reprod Biomed Online 2006;13(5):618627.Google Scholar
Ozmen, B, Sönmezer, M, Atabekoglu, CS, Olmus, H. Use of aromatase inhibitors in poor-responder patients receiving GnRH antagonist protocols. Reprod Biomed Online 2009;19(4):478485.Google Scholar
Baruffi, RL, Mauri, AL, Petersen, CG, et al. Recombinant LH supplementation to recombinant FSH during induced ovarian stimulation in the GnRH-antagonist protocol: a meta-analysis. Reprod Biomed Online 2007;14(1):1425.Google Scholar
König, TE, van der Houwen, LE, Overbeek, A, et al. Recombinant LH supplementation to a standard GnRH antagonist protocol in women of 35 years or older undergoing IVF/ICSI: a randomized controlled multicentre study. Hum Reprod 2013;28(10):28042812.Google Scholar
Tavaniotou, A, Albano, C, Van Steirteghem, A, Devroey, P. The impact of LH serum concentration on the clinical outcome of IVF cycles in patients receiving two regimens of clomiphene citrate/gonadotrophin/0.25 mg cetrorelix. Reprod Biomed Online 2003;6(4):421426.Google Scholar
Lin, YH, Seow, KM, Chen, HJ, et al. Effect of cetrorelix dose on premature LH surge during ovarian stimulation. Reprod Biomed Online 2008;16(6):772777.Google Scholar
Messinis, IE, Templeton, A. Effect of high dose exogenous oestrogen on midcycle luteinizing hormone surge in human spontaneous cycles. Clin Endocrinol (Oxf) 1987;27(4):453459.Google Scholar
Glasier, A, Thatcher, SS, Wickings, EJ, Hillier, SG, Baird, DT. Superovulation with exogenous gonadotropins does not inhibit the luteinizing hormone surge. Fertil Steril 1988;49(1):8185.Google Scholar
Messinis, IE, Templeton, A, Baird, DT. Endogenous luteinizing hormone surge during superovulation induction with sequential use of clomiphene citrate and pulsatile human menopausal gonadotropin. J Clin Endocrinol Metab 1985;61(6):10761080.Google Scholar
Kolibianakis, EM, Bourgain, C, Papanikolaou, EG, et al. Prolongation of follicular phase by delaying hCG administration results in a higher incidence of endometrial advancement on the day of oocyte retrieval in GnRH antagonist cycles. Hum Reprod 2005;20(9):24532456.Google Scholar
Aboulghar, MA, Mansour, RT, Amin, YM, et al. A prospective randomized study comparing coasting with GnRH antagonist administration in patients at risk for severe OHSS. Reprod Biomed Online 2007;15(3):271279.Google Scholar
Lainas, TG, Sfontouris, IA, Zorzovilis, IZ, et al. Management of severe OHSS using GnRH antagonist and blastocyst cryopreservation in PCOS patients treated with long protocol. Reprod Biomed Online 2009;18(1):1520.Google Scholar
Bonilla-Musoles, FM, Raga, F, Castillo, JC, et al. High doses of GnRH antagonists are efficient in the management of severe ovarian hyperstimulation syndrome. Clin Exp Obstet Gynecol 2009;36(2):7881.Google Scholar
Wang, R, Lin, S, Wang, Y, Qian, W, Zhou, L. Comparisons of GnRH antagonist protocol versus GnRH agonist long protocol in patients with normal ovarian reserve: a systematic review and meta-analysis. PLoS One 2017;12(4):e0175985.CrossRefGoogle ScholarPubMed
Demirol, A, Gurgan, T. Comparison of microdose flare-up and antagonist multiple-dose protocols for poor-responder patients: a randomized study. Fertil Steril 2009;92(2):481485.Google Scholar
Toftager, M, Sylvest, R, Schmidt, L, et al. Quality of life and psychosocial and physical well-being among 1,023 women during their first assisted reproductive technology treatment: secondary outcome to a randomized controlled trial comparing gonadotropin-releasing hormone (GnRH) antagonist and GnRH agonist protocols. Fertil Steril 2018;109(1):154164.Google Scholar
Devroey, P, Aboulghar, M, Garcia-Velasco, J, et al. Improving the patient’s experience of IVF/ICSI: a proposal for an ovarian stimulation protocol with GnRH antagonist co-treatment. Hum Reprod 2009;24(4):764774.Google Scholar
Fauser, BC, Devroey, P, Yen, SS, et al. Minimal ovarian stimulation for IVF: appraisal of potential benefits and drawbacks. Hum Reprod 1999;14:26812686.Google Scholar
Ledger, WL. Favourable outcomes from “mild” in-vitro fertilization. Lancet 2007;369:717718.Google Scholar
Nargund, G, Fauser, BC, Macklon, NS, et al. The ISMAAR proposal on terminology for ovarian stimulation for IVF. Hum Reprod 2007;22:28012804.Google Scholar
Lambalk, CB, Leader, A, Olivennes, F, et al. Treatment with the GnRH antagonist ganirelix prevents premature LH rises and luteinization in stimulated intrauterine insemination: results of a double-blind, placebo-controlled, multicentre trial. Hum Reprod 2006;21(3):632639.CrossRefGoogle ScholarPubMed
Qublan, HS, Amarin, Z, Tahat, YA, Smadi, AZ, Kilani, M. Ovarian cyst formation following GnRH agonist administration in IVF cycles: incidence and impact. Hum Reprod 2006;21:640644.Google Scholar
Al-Inany, H, Aboulghar, M. GnRH antagonist in assisted reproduction: a Cochrane review. Hum Reprod 2002;17(4):874885.Google Scholar

References

Gemzell, CA, Diczfalusy, E, Tillinger, G. Clinical effect of human pituitary follicle-stimulating hormone (FSH). J Clin Endocrinol Metab 1958;18:13331348.Google Scholar
Bettendorf, S. Human hypophyseal gonadotropin in hypophysectomized women. Int J Fertil 1963;45:799809.Google Scholar
Cochius, JI, Mack, K, Burns, RJ. Creutzfeld–Jakob disease in a recipient human pituitary derived gonadotrophin. Aust N Z J Med 1990;20:592596.Google Scholar
Dumble, LD, Klein, RD. Creutzfeld-Jakob disease legacy for Australian women treated with human pituitary gonadotropins. Lancet 1992;330:848.Google Scholar
Lunenfeld, B, Menzi, A, Insler, V. Effeti clinicci della gonadotropina umana della post-menopausa. Rass Clin Terap 1960;59:213216.Google Scholar
Lunenfeld, B. Treatment of anovulation by human gonadotrophins. J Int Fedn Gynecol Obstet 1963;1:153.Google Scholar
Biffoni, M, Battaglia, A, Borrelli, F, et al. Allergenic potential of gonadotrophic preparations in experimental animals: relevance of purity. Hum Reprod 1994;9:18451848.Google Scholar
Biffoni, M, Marcucci, I, Ythier, A, Eshkol, A. Effects of urinary gonadotrophin preparations on human in-vitro immune function. Hum Reprod 1998;13:24302434.Google Scholar
Lunenfeld, B, Eshkol, A. Immunology of follicle stimulating hormone and luteinizing hormone. Vitam Horm 1970;27:131159.CrossRefGoogle Scholar
Balen, AH, Hayden, CJ, Rutherford, AJ. What are the clinical benefits of recombinant gonadotrophins? Clinical efficacy of recombinant gonadotrophins. Hum Reprod 1999;14:14111417.Google Scholar
World Health Organization. Agents stimulating gonadal function in human. Report of a WHO scientific group. World Health Organization Tech Rep Ser 514. Geneva: World Health Organization; 1973.Google Scholar
Reichl, H, Balen, A, Jansen, CA. Prion transmission in blood and urine: what are the implications for recombinant and urinary-derived gonadotrophins? Hum Reprod 2002;17:25012508.Google Scholar
Serban, A, Legname, G, Hansen, K, Kovaleva, N, Prusiner, SB. Immunoglobulins in urine of hamsters with scrapie. J Biol Chem 2004;279:4881748820.Google Scholar
Shaked, GM, Shaked, Y, Kariv-Inbal, Z, et al. A protease-resistant prion protein isoform is present in urine of animals and humans affected with prion diseases. J Biol Chem 2001;276:3147931482.Google Scholar
Hull, M, Corrigan, E, Piazzi, A, Loumaye, E. Recombinant human luteinising hormone: an effective new gonadotropin preparation. Lancet 1994;344:334335.Google Scholar
Abd-Elaziz, K, Duijkers, I, Stöckl, L, et al. A new fully human recombinant FSH (follitropin epsilon): two phase I randomized placebo and comparator-controlled pharmacokinetic and pharmacodynamic trials. Hum Reprod 2017;32(8):16391647.Google Scholar
Hård, K, Mekking, A, Damm, JB, et al. Isolation and structure determination of the intact sialylated N-linked carbohydrate chains of recombinant human follitropin expressed in Chinese hamster ovary cells. Eur J Biochem 1990;193:263271.CrossRefGoogle ScholarPubMed
Creus, S, Chaia, Z, Pellizzari, EH, et al. Human FSH isoforms: carbohydrate complexity as determinant of in-vitro bioactivity. Mol Cell Endocrinol 2001;174:4149.Google Scholar
Timossi, CM, Barrios-de-Tomasi, J, González-Suárez, R, et al. Differential effects of the charge variants of human follicle-stimulating hormone. J Endocrinol 2000;165:193205.Google Scholar
Ulloa-Aguirre, A, Timossi, C. Biochemical and functional aspects of gonadotrophin-releasing hormone and gonadotrophins. Reprod Biomed Online 2000;1:4862.Google Scholar
Ulloa-Aguirre, A, Timossi, C, Barrios-de-Tomasi, J, Maldonado, A, Nayudu, P. Impact of carbohydrate heterogeneity in function of follicle-stimulating hormone: studies derived from in vitro and in vivo models. Biol Reprod 2003;69:379389.CrossRefGoogle ScholarPubMed
Ulloa-Aguirre, A, Zambrano, E, Timossi, C, et al. On the nature of the follicle-stimulating signal delivered to the ovary during exogenously controlled follicular maturation. A search into the immunological and biological attributes and the molecular composition of two preparations of urofollitropin. Arch Med Res 1995;26 Spec No:S219S230.Google Scholar
Yding Andersen, C, Leonardsen, L, Ulloa-Aguirre, A, et al. FSH-induced resumption of meiosis in mouse oocytes: effect of different isoforms. Mol Hum Reprod 1999;5:726731.Google Scholar
Yding Andersen, CY, Leonardsen, L, Ulloa-Aguirre, A, et al. Effect of different FSH isoforms on cyclic-AMP production by mouse cumulus-oocyte-complexes: a time course study. Mol Hum Reprod 2001;7:129135.Google Scholar
Yding Andersen, C. Effect of FSH and its different isoforms on maturation of oocytes from pre-ovulatory follicles. Reprod Biomed Online 2002;5:232239.Google Scholar
Lambert, A, Rodgers, M, Mitchell, R, et al. In-vitro biopotency and glycoform distribution of recombinant human follicle stimulating hormone (Org 32489), Metrodin and Metrodin-HP Hum Reprod 1995;10:19281935.Google Scholar
Andersen, CY, Westergaard, LG, van Wely, M. FSH isoform composition of commercial gonadotrophin preparations: a neglected aspect? Reprod Biomed Online 2004;9:231236.Google Scholar
Dias, JA, Ulloa-Aguirre, A. New human follitropin preparations: how glycan structural differences may affect biochemical and biological function and clinical effect. Front Endocrinol (Lausanne) 2021;12:636038. doi: 10.3389/fendo.2021.636038.Google Scholar
Deeks, JJ, Higgins, JPT, Altman, DG (eds.). Chapter 10: Analysing data and undertaking meta-analyses. In: Higgins, JPT, Thomas, J, Chandler, J, et al., eds. Cochrane Handbook for Systematic Reviews of Interventions, version 6.2 (updated February 2021). Cochrane; 2021.Google Scholar
Daya, S, Gunby, J, Hughes, EG, Collins, JA, Sagle, MA. Follicle-stimulating hormone versus human menopausal gonadotropin for in vitro fertilization cycles: a meta-analysis. Fertil Steril 1995;64:347354.Google Scholar
Agrawal, R, Holmes, J, Jacobs, HS. Follicle-stimulating hormone or human menopausal gonadotropin for ovarian stimulation in in vitro fertilization cycles: a meta-analysis. Fertil Steril 2000;73:338343.Google Scholar
van Wely, M, Kwan, I, van der Veen, F, et al. Recombinant FSH versus urinary gonadotrophins for ovarian hyperstimulation in IVF or ICSI cycles. A systematic review and meta-analysis. Hum Reprod 2009;24:S1, i134.Google Scholar
Bordewijk, EM, Mol, F, van der Veen, F, van Wely, M. Required amount of rFSH, HP-hMG and HP-FSH to reach a live birth. A systematic review and meta-analysis. Hum Reprod Open 2019; 2019(3): hoz008. https://doi.org/10.1093/hropen/hoz008.Google Scholar
Mochtar, MH, Danhof, NA, Ayeleke, RO, Van der Veen, F, van Wely, M. Recombinant luteinizing hormone (rLH) and recombinant follicle stimulating hormone (rFSH) for ovarian stimulation in IVF/ ICSI cycles. Cochrane Database Syst Rev 2017;5: CD005070.Google Scholar
Lensen, SF, Wilkinson, J, Leijdekkers, JA, et al. Individualised gonadotropin dose selection using markers of ovarian reserve for women undergoing in vitro fertilisation plus intracytoplasmic sperm injection (IVF/ICSI). Cochrane Database Syst Rev 2018;2:CD012693.Google Scholar

References

Remohi, J, Gartner, B, Gallardo, E, et al. Pregnancy and birth rates after oocyte donation. Fertil Steril 1997;67:717723.Google Scholar
The European IVF-Monitoring Consortium (EIM), for the European Society of Human Reproduction and Embryology (ESHRE), Kupka, MS, D’Hooghe, T, Ferraretti, AP, et al. Assisted reproductive technology in Europe, 2011: results generated from European registers by ESHRE. Hum Reprod 2016;31:233248.Google Scholar
van Wely, M, Kwan, I, Burt, AL, et al. Recombinant versus urinary gonadotrophin for ovarian stimulation in assisted reproductive technology cycles. Cochrane Database Syst Rev 2011;2:CD005354.Google Scholar
Caligara, C, Navarro, J, Vargas, G, et al. The effect of repeated controlled ovarian stimulation in donors. Hum Reprod 2001;16:23202323.Google Scholar
Budak, E, Garrido, N, Soares, SR, et al. Improvements achieved in an oocyte donation program over a 10-year period: sequential increase in implantation and pregnancy rates and decrease in high-order multiple pregnancies. Fertil Steril 2007;88:342349.Google Scholar
Soares, SR, Troncoso, C, Bosch, E, et al. Age and uterine receptiveness: predicting the outcome of oocyte donation cycles. J Clin Endocrinol Metab 2005;90:43994404.Google Scholar
Kumar, P, Radhakrishnan, J, Chowdhary, MA, Giampietro, PF. Prevalence and patterns of presentation of genetic disorders in a pediatric emergency department. Mayo Clin Proc 2001;76:777783.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
Clua, E, Tur, R, Coroleu, B, et al. Elective single-embryo transfer in oocyte donation programmes: should it be the rule? Reprod Biomed Online 2012;25:642648.CrossRefGoogle ScholarPubMed
Pandian, Z, Marjoribanks, J, Ozturk, O, Serour, G, Bhattacharya, S. Number of embryos for transfer following in vitro fertilisation or intra-cytoplasmic sperm injection. Cochrane Database Syst Rev 2013;7:CD003416.Google Scholar
Cobo, A, Garrido, N, Pellicer, A, Remohí, J. Six years’ experience in ovum donation using vitrified oocytes: report of cumulative outcomes, impact of storage time, and development of a predictive model for oocyte survival rate. Fertil Steril 2015;104:14261434.Google Scholar
Baart, EB, Martini, E, Eijkemans, MJ, et al. Milder ovarian stimulation for in-vitro fertilization reduces aneuploidy in the human preimplantation embryo: a randomized controlled trial. Hum Reprod 2007;22:980988.Google Scholar
Arce, JC, Andersen, AN, Fernández-Sánchez, M, et al. Ovarian response to recombinant human follicle-stimulating hormone: a randomized, antimüllerian hormone-stratified, dose-response trial in women undergoing in vitro fertilization/intracytoplasmic sperm injection. Fertil Steril 2014;102:16331640.Google Scholar
Ata, B, Kaplan, B, Danzer, H, et al. Array CGH analysis shows that aneuploidy is not related to the number of embryos generated. Reprod Biomed Online 2012;24:614620.Google Scholar
Morin, S, Melzer-Ross, K, McCulloh, D, Grifo, J, Munne, S. A greater number of euploid blastocysts in a given cohort predicts excellent outcomes in single embryo transfer cycles. J Assist Reprod Genet 2014;31:667673.Google Scholar
Labarta, E, Bosch, E, Alamá, P, et al. Moderate ovarian stimulation does not increase the incidence of human embryo chromosomal abnormalities in in vitro fertilization cycles. J Clin Endocrinol Metab 2012;97:19871994.Google Scholar

References

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
Baerwald, AR, Adams, GP, Pierson, RA. Characterization of ovarian follicular wave dynamics in women. Biol Reprod 2003;69:10231031.Google Scholar
Baerwald, AR, Adams, GP, Pierson, RA. A new model for ovarian follicular development during the human menstrual cycle. Fertil Steril 2003;80:116122.CrossRefGoogle ScholarPubMed
McNatty, KP, Hillier, SG, van den Boogaard, AM, et al. Follicular development during the luteal phase of the human menstrual cycle. J Clin Endocrinol Metab 1983;56:10221031.Google Scholar
von Wolff, M, Thaler, CJ, Frambach, T, et al. Ovarian stimulation to cryopreserve fertilized oocytes in cancer patients can be started in the luteal phase. Fertil Steril 2009;92:13601365.Google Scholar
Bedoschi, GM, de Albuquerque, FO, Ferriani, RA, Navarro, PA. Ovarian stimulation during the luteal phase for fertility preservation of cancer patients: case reports and review of the literature. J Assist Reprod Genet 2010;27:491494.Google Scholar
Sonmezer, M, Turkcuoglu, I, Coskun, U, Oktay, K. Random-start controlled ovarian hyperstimulation for emergency fertility preservation in letrozole cycles. Fertil Steril 2011;95(2125):e9e11.Google Scholar
Kuang, YP, Chen, QJ, Hong, QQ, et al. Luteal-phase ovarian stimulation case report: three-year follow-up of a twin birth. J IVF Reprod Med Genet 2013;1(2):1000106.Google Scholar
Buendgen, NK, Schultze-Mosgau, A, Cordes, T, Diedrich, K, Griesinger, G. Initiation of ovarian stimulation independent of the menstrual cycle: a case-control study. Arch Gynecol Obstet 2013;288:901904.Google Scholar
Kuang, Y, Hong, Q, Chen, Q, et al. Luteal-phase ovarian stimulation is feasible for producing competent oocytes in women undergoing in vitro fertilization/intracytoplasmic sperm injection treatment, with optimal pregnancy outcomes in frozen-thawed embryo transfer cycles. Fertil Steril 2014;101:105111.Google Scholar
Wang, N, Wang, Y, Chen, Q, et al. Luteal-phase ovarian stimulation vs conventional ovarian stimulation in patients with normal ovarian reserve treated for IVF: a large retrospective cohort study. Clin Endocrinol 2016;84:720728.Google Scholar
Li, Y, Yang, W, Chen, X, et al. Comparison between follicular stimulation and luteal stimulation protocols with clomiphene and HMG in women with poor ovarian response. Gynecol Endocrinol 2016;32:7477.Google Scholar
Spies, HG, Niswender, GD. Blockade of the surge of preovulatory serum luteinizing hormone and ovulation with exogenous progesterone in cycling rhesus (Macaca mulatta) monkeys. J Clin Endocrinol Metab 1971;32:309316.Google Scholar
Dierschke, DJ, Yamaji, T, Karsch, FJ, et al. Blockade by progesterone of estrogen-induced LH and FSH release in the rhesus monkey. Endocrinology 1973;92:14961501.Google Scholar
Pohl, CR, Knobil, E. The role of the central nervous system in the control of ovarian function in higher primates. Annu Rev Physiol 1982;44:583593.Google Scholar
March, CM, Goebelsmann, U, Nakamura, RM, Mishell, DR, Jr. Roles of estradiol and progesterone in eliciting the midcycle luteinizing hormone and follicle-stimulating hormone surges. J Clin Endocrinol Metabol 1979;49:507513.Google Scholar
Wu, Y, Zhao, FC, Sun, Y, Liu, PS. Luteal-phase protocol in poor ovarian response: a comparative study with an antagonist protocol. J Int Med Res 2017;45:17311738.Google Scholar
Zhang, W, Wang, M, Wang, S, et al. Luteal phase ovarian stimulation for poor ovarian responders. JBRA Assist Reprod 2018;22:193198.Google Scholar
Ubaldi, FM, Capalbo, A, Vaiarelli, A, et al. Follicular versus luteal phase ovarian stimulation during the same menstrual cycle (DuoStim) in a reduced ovarian reserve population results in a similar euploid blastocyst formation rate: new insight in ovarian reserve exploitation. Fertil Steril 2016;105:1488.e1–1495.e1.Google Scholar
Chen, H, Wang, Y, Lyu, Q, et al. Comparison of live-birth defects after luteal-phase ovarian stimulation vs. conventional ovarian stimulation for in vitro fertilization and vitrified embryo transfer cycles. Fertil Steril 2015;103:1194.e2–1201.e2.Google Scholar
Cimadomo, D, Vaiarelli, A, Colamaria, S, et al. Luteal phase anovulatory follicles result in the production of competent oocytes: intra-patient paired case-control study comparing follicular versus luteal phase stimulations in the same ovarian cycle. Hum Reprod 2018;33:14421448.Google Scholar
Kuang, Y, Chen, Q, Fu, Y, et al. Medroxyprogesterone acetate is an effective oral alternative for preventing premature luteinizing hormone surges in women undergoing controlled ovarian hyperstimulation for in vitro fertilization. Fertil Steril 2015;104:62.e3–70.e3.Google Scholar
Lu, X, Hong, Q, Sun, L, et al. Dual trigger for final oocyte maturation improves the oocyte retrieval rate of suboptimal responders to gonadotropin-releasing hormone agonist. Fertil Steril 2016;106:13561362.Google Scholar
Dong, J, Wang, Y, Chai, WR, et al. The pregnancy outcome of progestin-primed ovarian stimulation using 4 versus 10 mg of medroxyprogesterone acetate per day in infertile women undergoing in vitro fertilisation: a randomised controlled trial. BJOG 2017;124:10481055.Google Scholar
Wang, Y, Chen, Q, Wang, N, et al. Controlled ovarian stimulation using medroxyprogesterone acetate and hMG in patients with polycystic ovary syndrome treated for IVF: a double-blind randomized crossover clinical trial. Medicine 2016;95:e2939.Google Scholar
Xiao, ZN, Peng, JL, Yang, J, Xu, WM. Flexible GnRH antagonist protocol versus progestin-primed ovarian stimulation (PPOS) protocol in patients with polycystic ovary syndrome: comparison of clinical outcomes and ovarian response. Curr Med Sci 2019;39:431436.Google Scholar
Chen, Q, Chai, W, Wang, Y, et al. Progestin vs. gonadotropin-releasing hormone antagonist for the prevention of premature luteinizing hormone surges in poor responders undergoing in vitro fertilization treatment: a randomized controlled trial. Front Endocrinol (Lausanne) 2019;10:796.Google Scholar
Yu, S, Long, H, Chang, HY, et al. New application of dydrogesterone as a part of a progestin-primed ovarian stimulation protocol for IVF: a randomized controlled trial including 516 first IVF/ICSI cycles. Hum Reprod 2018;33:229237.Google Scholar
Zhu, X, Zhang, X, Fu, Y. Utrogestan as an effective oral alternative for preventing premature luteinizing hormone surges in women undergoing controlled ovarian hyperstimulation for in vitro fertilization. Medicine 2015;94:e909.Google Scholar
Crha, I, Ventruba, P, Filipinska, E, et al. [Medroxyprogesterone acetate use to block LH surge in oocyte donor stimulation]. Ceska gynekologie 2018;83:1116.Google Scholar
diZerega, GS, Hodgen, GD. Cessation of folliculogenesis during the primate luteal phase. J Clin Endocrinol Metab 1980;51:158160.Google Scholar
Helmond, FA, Simons, PA, Hein, PR. Strength and duration characteristics of the facilitory and inhibitory effects of progesterone on the estrogen-induced gonadotropin surge in the female rhesus monkey. Endocrinology 1981;108:18371842.Google Scholar
Helmond, FA, Simons, PA, Hein, PR. The effects of progesterone on estrogen-induced luteinizing hormone and follicle-stimulating hormone release in the female rhesus monkey. Endocrinology 1980;107:478485.Google Scholar
Gougeon, A. Regulation of ovarian follicular development in primates: facts and hypotheses. Endocr Rev 1996;17:121155.Google Scholar
Richter, TA, Robinson, JE, Lozano, JM, Evans, NP. Progesterone can block the preovulatory gonadotropin-releasing hormone/luteinising hormone surge in the ewe by a direct inhibitory action on oestradiol-responsive cells within the hypothalamus. J Neuroendocrinol 2005;17:161169.Google Scholar
Yildiz, S, Turkgeldi, E, Angun, B, et al. Comparison of a novel flexible progestin primed ovarian stimulation protocol and the flexible gonadotropin-releasing hormone antagonist protocol for assisted reproductive technology. Fertil Steril 2019;112:677683.Google Scholar
Zhang, J, Mao, X, Wang, Y, et al. Neonatal outcomes and congenital malformations in children born after human menopausal gonadotropin and medroxyprogesterone acetate treatment cycles. Arch Gynecol Obstet 2017;296:1207–17.Google Scholar
Kuang, Y, Chen, Q, Hong, Q, et al. Double stimulations during the follicular and luteal phases of poor responders in IVF/ICSI programmes (Shanghai protocol). Reprod Biomed Online 2014;29:684–91.Google Scholar
Tsampras, N, Gould, D, Fitzgerald, CT. Double ovarian stimulation (DuoStim) protocol for fertility preservation in female oncology patients. Hum Fertil 2017;20:248253.Google Scholar
Smitz, J, Van Den Abbeel, E, Bollen, N, et al. The effect of gonadotrophin-releasing hormone (GnRH) agonist in the follicular phase on in-vitro fertilization outcome in normo-ovulatory women. Hum Reprod 1992;7:10981102.CrossRefGoogle ScholarPubMed
Niederberger, C, Pellicer, A, Cohen, J, et al. Forty years of IVF. Fertil Steril 2018;110:185.e5–324.e5.Google Scholar

References

Oudendijk, JF, Yarde, F, Eijkemans, MJ, Broekmans, FJ, Broer, SL. The poor responder in IVF: is the prognosis always poor? a systematic review. Hum Reprod Update 2012;18:111.Google Scholar
Bosdou, JK, Venetis, CA, Kolibianakis, EM, et al. The use of androgens or androgen modulating agents in poor responders undergoing in vitro fertilization: a systematic review and meta-analysis. Hum Reprod Update 2012;18:127145.Google Scholar
Papathanasiou, A, Searle, BJ, King, NM, Bhattacharya, S. Trends in ‘poor responder’ research: lessons learned from RCTs in assisted conception. Hum Reprod Update 2016;22:306319.Google Scholar
Surrey, ES, Schoolcraft, WB. Evaluating strategies for improving ovarian response of the poor responder undergoing assisted reproductive techniques. Fertil Steril 2000;73:667676.Google Scholar
Papathanasiou, A. Implementing the ESHRE “poor responder” criteria in research studies: methodological implications. Hum Reprod 2014;29:18351838.Google Scholar
Polyzos, NP, Devroey, P. A systematic review of randomized trials for the treatment of poor ovarian responders: is there any light at the end of the tunnel? Fertil Steril 2011;96:10581061.Google Scholar
Ferraretti, AP, La Marca, A, Fauser, BC, et al. ESHRE consensus on the definition of ‘poor response’ to ovarian stimulation for in vitro fertilization: the Bologna criteria. Hum Reprod 2011;26:16161624.Google Scholar
Ferraretti, AP, Gianaroli, L. The Bologna criteria for the definition of poor ovarian responders: is there a need for revision? Hum Reprod 2014;29:18421845.Google Scholar
Venetis, CA. The Bologna criteria for poor ovarian response: the good, the bad and the way forward. Hum Reprod 2014;29:18391841.Google Scholar
Younis, JS. The Bologna criteria for poor ovarian response; has the job been accomplished. Hum Reprod 2012;27:18741875.Google Scholar
Poseidon Group (Patient-Oriented Strategies Encompassing IndividualizeD Oocyte Number); Alviggi, C, Andersen, CY, Buehler, K, et al. A new more detailed stratification of low responders to ovarian stimulation: from a poor ovarian response to a low prognosis concept. Fertil Steril 2016;105:14521453.Google Scholar
Esteves, SC, Alviggi, C, Humaidan, P, et al. The POSEIDON criteria and its measure of success through the eyes of clinicians and embryologists. Front Endocrinol (Lausanne) 2019;10:814.Google Scholar
Humaidan, P, La Marca, A, Alviggi, C, Esteves, SC, Haahr, T. Future perspectives of POSEIDON stratification for clinical practice and research. Front Endocrinol (Lausanne) 2019;10:439.Google Scholar
Leijdekkers, JA, Eijkemans, MJC, van Tilborg, TC, et al. Cumulative live birth rates in low-prognosis women. Hum Reprod 2019;34:10301041.Google Scholar
Shi, W, Zhou, H, Tian, L, et al. Cumulative live birth rates of good and low prognosis patients according to POSEIDON criteria: a single center analysis of 18,455 treatment cycles. Front Endocrinol (Lausanne) 2019;10:409.Google Scholar
ESHRE Reproductive Endocrinology Guideline Group. Ovarian stimulation for IVF/ICSI: guideline of the European Society of Human Reproduction and Embryology. https://www.eshre.eu/Guidelines-and-Legal/Guidelines/Ovarian-Stimulation-in-IVF-ICSI.Google Scholar
Al-Inany, HG, Youssef, MA, Ayeleke, RO, et al. Gonadotrophin-releasing hormone antagonists for assisted reproductive technology. Cochrane Database Syst Rev 2016;4:CD001750.Google Scholar
Kuang, Y, Chen, Q, Fu, Y, et al. Medroxy progesterone acetate is an effective oral alternative for preventing premature luteinizing hormone surges in women undergoing controlled ovarian hyperstimulation for in vitro fertilization. Fertil Steril 2015;104:6270.Google Scholar
Zhang, J, Wang, Y, Mao, X, et al. Dual trigger of final oocyte maturation in poor ovarian responders undergoing IVF/ICSI cycles. Reprod Biomed Online 2017;35:701707.Google Scholar
Sunkara, SK, Ramaraju, GA, Kamath, MS. Management strategies for POSEIDON group 2. Front Endocrinol (Lausanne) 2020;11:105.Google Scholar
Gallot, V, Berwanger da Silva, AL, Genro, V, et al. Antral follicle responsiveness to follicle-stimulating hormone administration assessed by the follicular output RaTe (FORT) may predict in vitro fertilization-embryo transfer outcome. Hum Reprod 2012;27:10661072.Google Scholar
Alviggi, C, Conforti, A, Esteves, SC, et al. Understanding ovarian hypo-response to exogenous gonadotropin in ovarian stimulation and its new proposed marker-the follicle-to-oocyte (FOI) index. Front Endocrinol (Lausanne) 2018;9:589.Google Scholar
Benmachiche, A, Benbouhedja, S, Zoghmar, A, Humaidan, P. Low LH level on the day of GnRH agonist trigger is associated with reduced ongoing pregnancy and live birth rates and increased early miscarriage rates following IVF/ICSI treatment and fresh embryo transfer. Front Endocrinol (Lausanne) 2019;10:639.Google Scholar
Propst, AM, Hill, MJ, Bates, GW, et al. Low dose human chorionic gonadotropin may improve in vitro fertilization cycle outcomes in patients with low luteinizing hormone levels after gonadotropin-releasing hormone antagonist administration. Fertil Steril 2011;96:898904.Google Scholar
Revelli, A, Martiny, G, Delle Piane, L, et al. A critical review of bi-dimensional and three-dimensional ultrasound techniques to monitor follicle growth: do they help improving IVF outcome? Reprod Biol Endocrinol 2014;12:107.Google Scholar
Ectors, FJ, Vanderzwalmen, P, Van Hoeck, J, et al. Relationship of human follicular diameter with oocyte fertilization and development after in-vitro fertilization or intracytoplasmic sperm injection. Hum Reprod 1997;12:20022005.Google Scholar
Kolibianakis, EM, Albano, C, Camus, M, et al. Prolongation of the follicular phase in in vitro fertilization results in a lower ongoing pregnancy rate in cycles stimulated with recombinant follicle-stimulating hormone and gonadotropin-releasing hormone antagonists. Fertil Steril 2004;82:102107.Google Scholar
Abbara, A, Clarke, SA, Dhillo, WS. Novel concepts for inducing final oocyte maturation in in vitro fertilization treatment. Endocr Rev 2018;39:593628.Google Scholar
Meyer, L, Murphy, LA, Gumer, A, et al. Risk factors for a suboptimal response to gonadotropin-releasing hormone agonist trigger during in vitro fertilization cycles. Fertil Steril 2015;104:637642.Google Scholar
Griffin, D, Feinn, R, Engmann, L, et al. Dual trigger with gonadotropin releasing hormone agonist and standard dose human chorionic gonadotropin to improve oocyte maturity rates. Fertil Steril 2014;102:405409.Google Scholar
Perez Mayorga, M, Gromoll, J, Behre, HM, et al. Ovarian response to follicle-stimulating hormone (FSH) stimulation depends on the FSH receptor genotype. J Clin Endocrinol Metab 2000;85:33653369.Google Scholar
Simoni, M, Nieschlag, E, Gromoll, J. Isoforms and single nucleotide polymorphisms of the FSH receptor gene: implications for human reproduction. Hum Reprod Update 2002;8:413421.Google Scholar
Mohiyiddeen, L, Newman, WG, McBurney, H, et al. Follicle-stimulating hormone receptor gene polymorphisms are not associated with ovarian reserve markers. Fertil Steril 2012;97:677681.Google Scholar
Alviggi, C, Clarizia, R, Pettersson, K, et al. Suboptimal response to GnRHa long protocol is associated with a common LH polymorphism. Reprod Biomed Online 2009;18:914.Google Scholar
Alviggi, C, Pettersson, K, Longobardi, S, et al. A common polymorphic allele of the LH beta-subunit gene is associated with higher exogenous FSH consumption during controlled ovarian stimulation for assisted reproductive technology. Reprod Biol Endocrinol 2013;1:51.Google Scholar
Lu, X, Yan, Z, Cai, R, et al. Pregnancy and live birth in women with pathogenic LHCGR variants using their own oocytes. J Clin Endocrinol Metab 2019;104:58775892.Google Scholar
Chen, Q, Wang, Y, Sun, L, et al. Controlled ovulation of the dominant follicle using progestin in minimal stimulation in poor responders. Reprod Biol Endocrinol 2017;15:71.Google Scholar
Chen, Q, Chai, W, Wang, Y, et al. Progestin vs. gonadotropin-releasing hormone antagonist for the prevention of premature luteinizing hormone surges in poor responders undergoing in vitro fertilization treatment: a randomized controlled trial. Front Endocrinol (Lausanne) 2019;10:796.Google Scholar
Lainas, TG, Sfontouris, IA, Venetis, CA, et al. Live birth rates after modified natural cycle compared with high-dose FSH stimulation using GnRH antagonists in poor responders. Hum Reprod 2015;30:23212330.Google Scholar
Patrizio, P, Vaiarelli, A, Levi Setti, PE, et al. How to define, diagnose and treat poor responders? Responses from a worldwide survey of IVF clinics. Reprod Biomed Online 2015;30:581592.Google Scholar
Wu, YG, Barad, DH, Kushnir, VA, et al. Aging-related premature luteinization of granulosa cells is avoided by early oocyte retrieval. J Endocrinol 2015;226:167180.Google Scholar
Wu, YG, Barad, DH, Kushnir, VA, et al. With low ovarian reserve, highly individualized egg retrieval (HIER) improves IVF results by avoiding premature luteinization. J Ovarian Res 2018;11:23.Google Scholar
Souza, ALM, Sampaio, M, Noronha, GB, et al. Effect of follicular flushing on reproductive outcomes in patients with poor ovarian response undergoing assisted reproductive technology. J Assist Reprod Genet 2017;34:13531357.Google Scholar
Koning, CH, Popp-Snijders, C, Schoemaker, J, Lambalk, CB. Elevated FSH concentrations in imminent ovarian failure are associated with higher FSH and LH pulse amplitude and response to GnRH. Hum Reprod 2000;15:14521456.Google Scholar
Lukaszuk, K, Kunicki, M, Liss, J, Bednarowska, A, Jakiel, G. Probability of live birth in women with extremely low anti-Müllerian hormone concentrations. Reprod Biomed Online 2014;28:6469.Google Scholar
Zhang, Y, Zhang, C, Shu, J, et al. Adjuvant treatment strategies in ovarian stimulation for poor responders undergoing IVF: a systematic review and network meta-analysis. Hum Reprod Update 2020;26:247263.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
×