Comparative clinical outcome following individualized follitropin delta dosing in Chinese women undergoing ovarian stimulation for in vitro fertilization /intracytoplasmic sperm injection

Rui Yang, Yunshan Zhang, Xiaoyan Liang, Xueru Song, Zhaolian Wei, Jianqiao Liu, Yezhou Yang, Jichun Tan, Qingxue Zhang, Yingpu Sun, Wei Wang, Weiping Qian, Lei Jin, Shuyu Wang, Yang Xu, Jing Yang, Marie Goethberg, Bernadette Mannaerts, Wen Wu, Zugeng Zheng, Jie Qiao, Rui Yang, Yunshan Zhang, Xiaoyan Liang, Xueru Song, Zhaolian Wei, Jianqiao Liu, Yezhou Yang, Jichun Tan, Qingxue Zhang, Yingpu Sun, Wei Wang, Weiping Qian, Lei Jin, Shuyu Wang, Yang Xu, Jing Yang, Marie Goethberg, Bernadette Mannaerts, Wen Wu, Zugeng Zheng, Jie Qiao

Abstract

Background: To compare the efficacy and safety of follitropin delta in its individualized fixed-dose regimen with follitropin alfa in a conventional adjustable dosing regimen in Chinese women. METHODS: This was a subgroup analysis of the randomized, multi-center, assessor-blind, non-inferiority trial (GRAPE) including 759 Chinese women (aged 20-40 years) recruited in 16 reproductive medicine clinics in China. Women were randomized in a 1:1 ratio to be treated with either follitropin delta dose based on anti-Müllerian hormone (AMH) and body weight or conventional dosing with follitropin alfa following a gonadotropin-releasing hormone (GnRH) antagonist protocol. The primary outcome was ongoing pregnancy rate assessed 10-11 weeks after embryo transfer in the fresh cycle (non-inferiority margin -10.0%).

Results: 378 in the follitropin delta group and 381 in the follitropin alfa group were randomized and exposed. Non-inferiority was confirmed with respect to ongoing pregnancy with rates of 31.0% vs. 25.7% for follitropin delta compared to follitropin alfa, estimated mean difference of 5.1% (95% confidence interval (CI) -1.3% to 11.5%). The clinical pregnancy rate (35.4% vs. 31.5%, P = 0.239) and live birth rate (31.0% vs. 25.5%, P = 0.101) were comparable between the follitropin delta group and the follitropin alfa group. Overall, the individualized follitropin delta treatment resulted in fewer oocytes retrieved compared to follitropin alfa treatment (10.3 ± 6.2 vs. 12.5 ± 7.5, P < 0.001), which was mainly due to fewer oocytes (10.5 ± 6.4 vs. 13.9 ± 7.8) in women with AMH ≥ 15 pmol/L. Accordingly there was a lower incidence of early ovarian hyper-stimulation syndrome (OHSS) and/or preventive interventions (6.1% vs. 11.0%, P = 0.013). A daily follitropin delta dose of 10.2 µg (95% CI: 9.3-11.2 µg) was estimated to provide the same number of oocytes retrieved as a starting dose of 150 IU/d of follitropin alfa.

Conclusion: Follitropin delta in its individualized fixed-dose regimen showed similar efficacy and improved safety compared with follitropin alfa in a conventional adjustable dosing regimen in Chinese women.

Clinical trial registration number: NCT03296527.

Keywords: Algorithm; Dose equivalence; Follitropin delta; Individualized dosing; Ongoing pregnancy; Ovarian hyperstimulation syndrome.

Conflict of interest statement

Marie Goethberg and Bernadette Mannaerts are employees of Ferring Pharmaceuticals, Denmark. Wen Wu and Zugeng Zheng are employees of Ferring Pharmaceuticals, China. The other authors have stated explicitly that there are no conflicts of competing interests with the article.

© 2022. The Author(s).

Figures

Fig. 1
Fig. 1
Flow chart showing the number of women at each stage of the clinical trial. Note: AMH, anti-Müllerian hormone; GnRH, gonadotropin-releasing hormone; hCG: human chorionic gonadotrophin
Fig. 2
Fig. 2
Adjustments of treatment doses according to AMH and body weight. Note: AMH, anti-Müllerian hormone.aWomen randomized to follitropin delta were administered a fixed daily dose according to patients’ serum AMH concentration at screening and body weight at randomization (AMH < 15 pmol/L: 12 mg; AMH ≥ 15 pmol/L: 0.10–0.19 mg/kg, the maximum daily dose was 12 μg). bWomen randomized to follitropin alfa were administered a conventional daily dose of 150 IU (11 μg) for the first 5 days, thereafter the dose was adjusted up or down according to follicular response, the maximum daily dose was 450 IU
Fig. 3
Fig. 3
Pregnancy and live birth outcomes. Note: hCG: human chorionic gonadotrophin.aRates are calculated per treatment group as the percentage of women in the FAS. The retrieved rate was defined as women with at least one oocyte retrieved. bThe transfer rate was defined as percentage of women with embryo transfer. cβhCG rate was defined as percentage of women with a positive test of serum βhCG 2 weeks after transfer. dClinical pregnancy rate was defined as percentage of women with at least one gestational sac 5–6 weeks after transfer. eVital pregnancy rate was defined as percentage of women with at least one intrauterine gestational sac with fetal heart after transfer. fOngoing pregnancy rate was defined as percentage of women with at least one intrauterine viable fetus 10–11 weeks after transfer. gLive birth rate was defined as percentage of women with birth of at least one live-born neonate. hLive birth rate at 4 weeks after birth was defined as percentage of women with at least one live neonate 4 weeks after birth
Fig. 4
Fig. 4
Analysis of dose equivalence. Note: A Number of oocytes retrieved for follitropin delta and 150 IU/day follitropin alfa in the study (n = 759); B Number of follicles ≥ 12 mm at end of stimulation for follitropin delta and 150 IU/day follitropin alfa in the study (n = 759); C Log of serum oestradiol concentrations at the end of stimulation for follitropin delta and 150 IU/day follitropin alfa in the study (n = 759). Estimated means (circles) with 95% confidence intervals (95% CI) and number of patients for the subgroups based on the dose of follitropin delta corresponding to the patients’ AMH concentrations and body weight. The intersection of the blue and red regression lines indicates the dose of follitropin delta estimated to give the same response as 150 IU of follitropin alfa. The estimate dose equivalence factor and its 95% CI are indicated by the arrow and the solid horizontal black line on the x-axis

References

    1. Nyboe Andersen A, Nelson SM, Fauser BC, García-Velasco JA, Klein BM, Arce JC, et al. Individualized versus conventional ovarian stimulation for in vitro fertilization: a multicenter, randomized, controlled, assessor-blinded, phase 3 noninferiority trial. Fertil Steril. 2017;107(387–96):e4.
    1. Allegra A, Marino A, Volpes A, Coffaro F, Scaglione P, Gullo S, et al. A randomized controlled trial investigating the use of a predictive nomogram for the selection of the FSH starting dose in IVF/ICSI cycles. Reprod Biomed Online. 2017;34:429–438. doi: 10.1016/j.rbmo.2017.01.012.
    1. Stormlund S, Sopa N, Zedeler A, Bogstad J, Prætorius L, Nielsen HS, et al. Freeze-all versus fresh blastocyst transfer strategy during in vitro fertilisation in women with regular menstrual cycles: multicentre randomised controlled trial. BMJ. 2020;370:m2519. doi: 10.1136/bmj.m2519.
    1. Sun XY, Lan YZ, Liu S, Long XP, Mao XG, Liu L. Relationship Between Anti-Mullerian Hormone and In Vitro Fertilization-Embryo Transfer in Clinical Pregnancy. Front Endocrinol (Lausanne) 2020;11:595448. doi: 10.3389/fendo.2020.595448.
    1. Iliodromiti S, Anderson RA, Nelson SM. Technical and performance characteristics of anti-Mullerian hormone and antral follicle count as biomarkers of ovarian response. Hum Reprod Update. 2015;21:698–710. doi: 10.1093/humupd/dmu062.
    1. Gassner D, Jung R. First fully automated immunoassay for anti-Mullerian hormone. Clin Chem Lab Med. 2014;52:1143–1152. doi: 10.1515/cclm-2014-0022.
    1. Fleming R, Fairbairn C, Gaudoin M. Objective multicentre performance of the automated assays for AMH and estimation of established critical concentrations. Hum Fertil (Camb) 2018;21:269–274. doi: 10.1080/14647273.2017.1331298.
    1. Lu Liu YL. Correlation of geographical latitude with stature and body weight of urban Han people in China. Chinese Journal of Anatomy. 2017;40(307–10):29.
    1. Jonker D. Body weight is the main determinant of systemic follicle stimulating hormone (FSH) concentrations during controlled ovarian stimulation with follitropin delta. Hum Reprod. The 37th Virtual Annual Meeting of the European Society of Human Reproduction and Embryology, 26 June to 1 July 2021. Volume 36, Supp 1 2021 abstract book, P-650. ESHRE 2021. .
    1. Abbara A, Patel A, Hunjan T, Clarke SA, Chia G, Eng PC, et al. FSH Requirements for Follicle Growth During Controlled Ovarian Stimulation. Front Endocrinol (Lausanne) 2019;10:579. doi: 10.3389/fendo.2019.00579.
    1. Ledger WL, Fauser BC, Devroey P, Zandvliet AS, Mannaerts BM. Corifollitropin alfa doses based on body weight: clinical overview of drug exposure and ovarian response. Reprod Biomed Online. 2011;23:150–159. doi: 10.1016/j.rbmo.2011.04.002.
    1. Arce JC, Andersen AN, Fernández-Sánchez M, Višnová H, Bosch E, García-Velasco JA, et al. Ovarian response to recombinant human follicle-stimulating hormone: a randomized, antimullerian hormone-stratified, dose-response trial in women undergoing in vitro fertilization/intracytoplasmic sperm injection. Fertil Steril. 2014;102(1633–40):e5.
    1. Ishihara O, Arce JC, Japanese Follitropin Delta Phase 3 Trial G Individualized follitropin delta dosing reduces OHSS risk in Japanese IVF/ICSI patients: a randomized controlled trial. Reprod Biomed Online. 2021;42:909–18. doi: 10.1016/j.rbmo.2021.01.023.
    1. Qiao J, Zhang Y, Liang X, Ho T, Huang HY, Kim SH, et al. A randomised controlled trial to clinically validate follitropin delta in its individualised dosing regimen for ovarian stimulation in Asian IVF/ICSI patients. Hum Reprod. 2021;36:2452–2462. doi: 10.1093/humrep/deab155.
    1. Bosch E, Havelock J, Martin FS, Rasmussen BB, Klein BM, Mannaerts B, et al. Follitropin delta in repeated ovarian stimulation for IVF: a controlled, assessor-blind Phase 3 safety trial. Reprod Biomed Online. 2019;38:195–205. doi: 10.1016/j.rbmo.2018.10.012.
    1. Arce JC, Larsson P, García-Velasco JA. Establishing the follitropin delta dose that provides a comparable ovarian response to 150 IU/day follitropin alfa. Reprod Biomed Online. 2020;41:616–622. doi: 10.1016/j.rbmo.2020.07.006.
    1. Višnová HPE, Martin FS, Koziol K, Klein BM, Mannaerts B. Clinical outcomes of potential high responders after individualized FSH dosing based on anti-Müllerian hormone and body weight. Reprod Biomed Online. 2021;43:1019–1026. doi: 10.1016/j.rbmo.2021.08.024.
    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. doi: 10.1097/00006254-198906000-00004.
    1. Fernández-Sánchez M, Višnová H, Yuzpe A, Klein BM, Mannaerts B, Arce JC, et al. Individualization of the starting dose of follitropin delta reduces the overall OHSS risk and/or the need for additional preventive interventions: cumulative data over three stimulation cycles. Reprod Biomed Online. 2019;38:528–537. doi: 10.1016/j.rbmo.2018.12.032.
    1. Zhou Z, Zheng D, Wu H, Li R, Xu S, Kang Y, et al. Epidemiology of infertility in China: a population-based study. BJOG. 2018;125:432–441. doi: 10.1111/1471-0528.14966.
    1. Qiao JFH. Assisted reproductive technology in China: compliance and non-compliance. Transl Pediatr. 2014;3:91–97.
    1. Bai F, Wang DY, Fan YJ, Qiu J, Wang L, Dai Y, et al. Assisted reproductive technology service availability, efficacy and safety in mainland China: 2016. Hum Reprod. 2020;35:446–452. doi: 10.1093/humrep/dez245.

Source: PubMed

3
Abonnieren