A randomised controlled trial to clinically validate follitropin delta in its individualised dosing regimen for ovarian stimulation in Asian IVF/ICSI patients

Jie Qiao, Yunshan Zhang, Xiaoyan Liang, Tuong Ho, Hong-Yuan Huang, Sung-Hoon Kim, Marie Goethberg, Bernadette Mannaerts, Joan-Carles Arce, Jie Qiao, Yunshan Zhang, Xiaoyan Liang, Tuong Ho, Hong-Yuan Huang, Sung-Hoon Kim, Marie Goethberg, Bernadette Mannaerts, Joan-Carles Arce

Abstract

Study question: Is ovarian stimulation with follitropin delta in its individualised fixed-dose regimen at least as efficacious as follitropin alfa in a conventional dosing regimen in Asian population?

Summary answer: Ovarian stimulation with individualised follitropin delta dosing resulted in a non-inferior ongoing pregnancy rate, a significantly higher live birth rate and a significantly lower incidence of early ovarian hyperstimulation syndrome (OHSS) and/or preventive interventions compared to conventional follitropin alfa dosing.

What is known already: Previous randomised controlled trials conducted in Japan as well as in Europe, North- and South America have demonstrated that ovarian stimulation with the individualised follitropin delta dosing regimen based on serum anti-Müllerian hormone (AMH) level and body weight modulated the ovarian response and reduced the risk of OHSS without compromising pregnancy and live birth rates.

Study design, size, duration: Randomised, controlled, multi-centre, assessor-blind trial conducted in 1009 Asian patients from mainland China, South Korea, Vietnam and Taiwan, undergoing their first IVF/ICSI cycle. Randomisation was stratified by age (<35, 35-37, 38-40 years). The primary endpoint was ongoing pregnancy rate assessed 10-11 weeks after embryo transfer in the fresh cycle (non-inferiority limit -10.0%; analysis adjusted for age stratum).

Participants/materials, setting, methods: The follitropin delta treatment consisted of a fixed daily dose individualised according to each patient's initial AMH level and body weight (AMH <15 pmol/l: 12 μg; AMH ≥15 pmol/l: 0.19 to 0.10 μg/kg; min-max 6-12 μg). The follitropin alfa dose was 150 IU/day for the first 5 days with subsequent potential dose adjustments according to individual response. A GnRH antagonist protocol was applied. OHSS was classified based on Golan's system. Women with an ongoing pregnancy were followed until live birth and 4 weeks after.

Main results and the role of chance: The number of oocytes retrieved was significantly (P < 0.001) lower with individualised follitropin delta versus conventional follitropin alfa (10.0 ± 6.1 versus 12.4 ± 7.3). Nevertheless, compared to the conventional dosing approach, the individualised follitropin delta dosing regimen resulted in on average 2 more oocytes (9.6 ± 5.3 versus 7.6 ± 3.5) in potential low responders as indicated by AMH <15 pmol/l, and on average 3 fewer oocytes (10.1 ± 6.3 versus 13.8 ± 7.5) in potential high responders as indicated by AMH ≥15 pmol/l. Among women with AMH ≥15 pmol/l, excessive response occurred less frequently with individualised follitropin delta than with follitropin alfa (≥15 oocytes: 20.2% versus 39.1%; ≥20 oocytes: 6.7% versus 18.5%; both P < 0.001). The incidence of early OHSS and/or preventive interventions for early OHSS was significantly (P = 0.004) reduced from 9.6% with follitropin alfa to 5.0% with individualised follitropin delta. The total gonadotropin use was significantly (P < 0.001) reduced from an average of 109.9 ± 32.9 μg (1498 ± 448 IU) follitropin alfa to 77.5 ± 24.4 μg follitropin delta. Non-inferiority of follitropin delta in its individualised dosing regimen to conventional follitropin alfa was established with respect to the primary endpoint of ongoing pregnancy rate which was 31.3% with follitropin delta compared to 25.7% with follitropin alfa (estimated mean difference 5.4% [95% CI: -0.2%; 11.0%]). The live birth rate was significantly higher at 31.3% with individualised follitropin delta compared to 24.7% with follitropin alfa (estimated mean difference 6.4% [95% CI: 0.9%; 11.9%]; P = 0.023). The live birth rate for each stratum were as follows for follitropin delta and follitropin alfa, respectively; <35 years: 31.0% versus 25.0%, 35-37 years: 35.3% versus 26.7%, 38-40 years: 20.0% versus 14.3%.

Limitations, reasons for caution: The trial only covered the clinical outcome of one treatment cycle with fresh cleavage-stage embryo transfers.

Wider implications of the findings: The present trial shows that in addition to reducing the early OHSS risk, follitropin delta in its individualised fixed-dose regimen has the potential to improve the success rate in fresh cycles across all ages and with a lower gonadotropin consumption compared to conventional follitropin alfa dosing.

Study funding/competing interest(s): This study was funded by Ferring Pharmaceuticals. J.Q., Y.Z., X.L., T.H., H.-Y.H. and S.-H.K. have received institutional (not personal) clinical trial fees from Ferring Pharmaceuticals. M.G., B.M. and J.-C.A. are employees of Ferring Pharmaceuticals. J.-C.A. has pending and issued patent applications in the WO 2013/020996 and WO 2019/043143 patent families that comprise allowed and granted patent rights related to follitropin delta.

Trial registration number: NCT03296527 (clinicaltrials.gov).

Trial registration date: 28 September 2017.

Date of first patient’s enrolment: 1 December 2017.

Keywords: anti-Müllerian hormone; follitropin delta; individualised dosing; live birth; ovarian stimulation.

© The Author(s) 2021. Published by Oxford University Press on behalf of European Society of Human Reproduction and Embryology.

Figures

Figure 1.
Figure 1.
Trial and participant flow. βhCG, beta unit of human chorionic gonadotropin; FAS, full analysis set (defined as all women randomised and exposed); PP, per-protocol (defined as all women randomised and exposed, except those excluded as a result of major protocol deviations).
Figure 2.
Figure 2.
Clinical efficacy outcomes. The estimated mean differences (follitropin delta-follitropin alfa) for all clinical efficacy outcomes are presented together with the corresponding 95% CIs for equal effect using the Mantel-Haenszel method to combine the risk differences across age strata. βhCG, beta unit of human chorionic gonadotropin.
Figure 3.
Figure 3.
Live birth rate for overall population and by age stratum. The blue (individualised follitropin delta) and red (conventional follitropin alfa) bars display the live birth rates for all patients who started stimulation and by each of the age randomisation strata (<35, 35–37 and 38–40 years). N, total number of patients.
Figure 4.
Figure 4.
Estimated risk of early OHSS (any grade) and/or preventive interventions for early OHSS by AMH. The solid blue (individualised follitropin delta) and red (conventional follitropin alfa) lines are based on a logistic regression model with treatment, log(AMH) and the treatment*log(AMH) interaction. The shadings represent 95% CIs for the estimated risks. The analysis of treatment difference indicates evidence of a benefit of follitropin delta over follitropin alfa (P < 0.01). AMH, anti-Müllerian hormone; OHSS, ovarian hyperstimulation syndrome.

References

    1. Arce J-C, Klein BM, Erichsen L.. Using AMH for determining a stratified gonadotropin dosing regimen for IVF/ICSI and optimizing outcomes. In: Seifer DB, Tal R (eds). Anti-Müllerian Hormone: Biology, Role in Ovarian Function and Clinical Significance. Hauppauge, NY: Nova Science Publishers, Inc, 2016, 83–102.
    1. Bleil ME, Gregorich SE, Adler NE, Sternfeld B, Rosen MP, Cedars MI.. Race/ethnic disparities in reproductive age: an examination of ovarian reserve estimates across four race/ethnic groups of healthy, regularly cycling women. Fertil Steril 2014;101:199–207.
    1. Bosch E, Havelock J, Martin FS, Rasmussen BB, Klein BM, Mannaerts B, Arce J-C; ESTHER-2 Study Group. Follitropin delta in repeated ovarian stimulation for IVF: a controlled, assessor-blind Phase 3 safety trial. Reprod Biomed Online 2019;38:195–205.
    1. Bosch E, Labarta E, Crespo J, Simón C, Remohí J, Jenkins J, Pellicer A.. Circulating progesterone levels and ongoing pregnancy rates in controlled ovarian stimulation cycles for in vitro fertilization: analysis of over 4000 cycles. Hum Reprod 2010;25:2092–2100.
    1. Broer SL, Broekmans FJM, Laven JSE, Fauser BCJM.. Anti-Müllerian hormone: ovarian reserve testing and its potential clinical implications. Hum Reprod Update 2014;20:688–701.
    1. Deurenberg P, Deurenberg-Yap M, Guricci S.. Asians are different from Caucasians and from each other in their body mass index/body fat per cent relationship. Obes Rev 2002;3:141–146.
    1. European Medicines Agency. Gonal-f. Summary of Product Characteristics. 2021. (15 March 2021, date last accessed).
    1. Fauser BCJM, Diedrich K, Devroey P; Evian Annual Reproduction Workshop Group 2007. Predictors of ovarian response: progress towards individualized treatment in ovulation induction and ovarian stimulation. Hum Reprod Update 2008;14:1–14.
    1. Fleming R, Broekmans F, Calhaz-Jorge C, Dracea L, Alexander H, Nyboe Andersen A, Blockeel C, Jenkins J, Lunenfeld B, Platteau P. et al.Can anti-Müllerian hormone concentrations be used to determine gonadotrophin dose and treatment protocol for ovarian stimulation? Reprod Biomed Online 2013;26:431–439.
    1. Golan A, Ron-el R, Herman A, Soffer Y, Weinraub Z, Caspi E.. Ovarian hyperstimulation syndrome: an update review. Obstet Gynecol Surv 1989;44:430–440.
    1. Ishihara O, Arce J-C; Follitropin Delta Phase 3 Trial (STORK) Group. Individualized follitropin delta dosing reduces OHSS risk in Japanese IVF/ICSI patients: a randomized controlled trial. Reprod Biomed Online 2021;42:909–914.
    1. La Marca A, Sunkara SK.. Individualization of controlled ovarian stimulation in IVF using ovarian reserve markers: from theory to practice. Hum Reprod Update 2014;20:124–140.
    1. Nelson SM, Aijun S, Ling Q, Tengda X, Wei X, Yan D, Yanfang W, Zenghui T, Xinqi C, Fraser A. et al.Ethnic discordance in serum anti-Müllerian hormone in healthy women: a population study from China and Europe. Reprod Biomed Online 2020;40:461–467.
    1. Nelson SM, Yates RW, Lyall H, Jamieson M, Traynor I, Gaudoin M, Mitchell P, Ambrose P, Fleming R.. Anti-Müllerian hormone-based approach to controlled ovarian stimulation for assisted conception. Hum Reprod 2009;24:867–875.
    1. Nelson SM.Biomarkers of ovarian response: current and future applications. Fertil Steril 2013;99:963–969.
    1. Nyboe Andersen A, Nelson SM, Fauser BCJM, García-Velasco JA, Klein BM, Arce J-C; ESTHER-1 Study Group. Individualized versus conventional ovarian stimulation for in vitro fertilization: a multicenter, randomized, controlled, assessor-blinded, phase 3 noninferiority trial. Fertil Steril 2017;107:387–396.
    1. Sterrenburg MD, Veltman-Verhulst SM, Eijkemans MJC, Hughes EG, Macklon NS, Broekmans FJ, Fauser BCJM.. Clinical outcomes in relation to the daily dose of recombinant follicle-stimulating hormone for ovarian stimulation in in vitro fertilization in presumed normal responders younger than 39 years: a meta-analysis. Hum Reprod Update 2011;17:184–196.
    1. WHO Expert Consultation. Appropriate body-mass index for Asian populations and its implications for policy and intervention strategies. Lancet 2004;363:157–163.

Source: PubMed

3
Abonnieren