Skip to main content
Log in

Follitropin Alpha in Infertility

A Review

  • Adis Drug Evaluation
  • Published:
BioDrugs Aims and scope Submit manuscript

Summary

Synopsis

Follitropin alpha (recombinant human follicle- stimulating hormone; follitropin alfa) is a recombinant form of follicle- stimulating hormone (FSH), an endogenous gonadotrophin. Unlike FSH products derived from urine [menotropins (human menopausal gonadotrophins), urofollitropin and highly purified urofollitropin], follitropin alpha is readily available and shows batch-to-batch consistency. As well, it is free of luteinising hormone (LH) activity and contaminant urinary proteins and can be self- administered subcutaneously.

In women undergoing in vitro fertilisation-embryo transfer (IVF- ET), follitropin alpha appears to have a greater stimulatory effect on follicular development than urine-derived FSH products as a group. In direct comparisons it had similar effects to urofollitropin but produced more oocytes per stimulated cycle than highly purified urofollitropin and menotropins. Preliminary results of 1 small trial indicate similar efficacy for follitropin alpha and follitropin beta. Rates of pregnancy, live births and multiple births have been similar among all treatment groups.

As ovulation induction in women with clomifene- resistant WHO group II anovulation, follitropin alpha produces rates of ovulation, follicular development and pregnancy resembling those seen with urofollitropin or highly purified urofollitropin. A long term low-dose regimen of follitropin alpha is associated with a lower number of follicles and a trend toward fewer multiple births compared with conventional follitropin alpha or urofollitropin regimens. Data from women with WHO group I anovulation and from infertile men are scant.

Tolerability has not differed between follitropin alpha and other FSH products. The incidence of general events (e.g. headache, nausea, ovarian cyst), local irritations at injection site and ovarian hyperstimulation syndrome resembled those for comparator FSH products. However, it appears that follitropin alpha can be tolerated in instances of severe allergic reaction to urine- derived products.

Conclusions. In women undergoing IVF- ET, follitropin alpha appears to have a greater stimulatory effect on follicle development than urine-derived FSH products as a group and is at least as well tolerated as these preparations; preliminary data indicate similar efficacy to follitropin beta. At present, its efficacy in women with WHO group II anovulation disorder has been shown to be similar to that of the older products.

Compared with urinary FSH products, the benefits of follitropin alpha lie in its reliable supply, consistency of production, lack of contaminant urinary proteins and ease of self-administration. Given these practical advantages, and the apparently greater effect onfollicular development overall in women undergoing IVF-ET, recombinant products such as follitropin alpha are expected to eventually replace older urine-derived FSH preparations and claim a prominent position in the treatment of infertility.

Role of FSH in Fertility

Follicle-stimulating hormone (FSH) and luteinising hormone (LH) are synthesised in the anterior pituitary gland. Their secretion is controlled by gonadorelin [gonadotrophin-releasing hormone (GnRH)].

In Women. During a typical ovarian/menstrual cycle, FSH levels are elevated in the first few days, decline around day 6, then rise sharply on days 12 to 14. FSH stimulates granulosa cell proliferation and differentiation and contributes to development of the mature follicle. Estrogen secreted by the follicle initiates endometrial proliferation and cervical changes favouring sperm entry. Feedback loops involving estrogen, progesterone and inhibin suppress additional FSH release, preventing development of other follicles.

In usual circumstances, a single dominant follicle develops while the others undergo atresia. Estrogen levels peak about a day before ovulation, prompting a surge in LH levels which triggers ovum release. The ruptured follicle becomes the corpus luteum and secretes progesterone to prepare the endometrium for implantation. After fertilisation, placental human chorionic gonadotrophin (hCG) maintains pregnancy.

In Men. Both FSH and LH are required for spermatogenesis. LH stimulates Leydig cells to produce testosterone which, together with FSH, stimulates seminiferous tubules and promotes spermatogenesis. FSH acts at the Sertoli cells in the seminiferous tubules and is regulated by inhibin within these cells, although testosterone and estradiol also inhibit FSH secretion.

Pharmacodynamic Properties

Follitropin alpha (recombinant human follicle-stimulating hormone; follitropin alfa) is a recombinant FSH product expressed in Chinese hamster ovary cells. It is structurally identical to endogenous FSH, a complex heterodimeric glycoprotein comprising an α and a β subunit. Follitropin alpha is free of potentially allergenic urinary proteins, has a higher specific activity (10 000 IU/mg) than urine-derived FSH preparations other than highly purified urofollitropin and shows similar biological activity to these products in standard animal assays.

Native FSH exists in many isoforms mainly distinguished by differences in the number of terminal sialic acid residues attached to the carbohydrate moiety. Follitropin alpha has a slightly narrower isoelectric point (pI) band than follitropin beta (mainly 4 to 5 vs 3.5 to 5.5) and contains fewer isoforms with a pI <4 (9 vs <24%). The clinical importance, if any, of differences in the FSH isoforms present in commercial preparations is undetermined.

In most patients, follitropin alpha is sufficient to stimulate follicular development without the addition of exogenous LH (see Clinical Efficacy summary).

There are few data on the influence of follitropin alpha on hormones other than estradiol. A comparative trial found insulin levels to be increased in follicular fluid in the follitropin alpha group and in serum in the follitropin beta group. Levels of human growth hormone in follicular fluid and serum were twice as high with follitropin alpha as with follitropin beta.

The effects of endogenous FSH on spermatogenesis in humans are well known; data on the effects of follitropin alpha are sparse at present. In rodents, follitropin alpha contributes to the maintenance of early spermatogenesis.

As shown in standard in vivo and in vitro models, follitropin alpha in doses up to 100 times greater than those used therapeutically exerts no important effects on the central or autonomic nervous system, cardiovascular or respiratory systems, gastrointestinal system or renal function.

Pharmacokinetic Properties

The pharmacokinetics of follitropin alpha resemble those of the endogenous hormone. After subcutaneous administration to healthy female volunteers given the GnRH agonist goserelin as pituitary down-regulation, follitropin alpha pharmacokinetics are described by a 2-compartment model with first-order absorption. Steady state is achieved within 4 days.

At steady state, mean peak plasma concentrations are 3-fold higher than after a single dose and the volume of distribution is similar to that of extracellular water. Serum FSH levels after follitropin alpha administration do not correlate with pharmacodynamic or clinical effects and are not useful predictors or monitors of treatment outcome.

Endogenous FSH is eliminated via renal and hepatic routes. Terminal elimination half-life (t½β) values for follitropin alpha vary by up to 70% among individuals: at steady state t½⨿ is about 24 hours. Renal clearance accounts for 10% of total clearance.

The pharmacokinetics of follitropin alpha and urofollitropin are similar after intravenous administration, except that renal urofollitropin clearance is faster (by about 30%).

Clinical Efficacy

Superovulation for Assisted Reproduction Techniques (ART). In women undergoing in vitro fertilisation-embryo transfer (IVF-ET), about 8 to 12 oocytes are collected per cycle stimulated with follitropin alpha, fertilisation rate is about 55 to 68% and the mean number of embryos obtained ranges from about 4 to 8. Clinical pregnancy rates are in the range of 10 to 45% per cycle and 14 to 48% per embryo transfer. The live birth rate in 1 trial was 34% per cycle and 36% per embryo transfer.

Follitropin alpha appears to be more beneficial than urine-derived FSH products as a group in stimulating follicle development. In direct comparisons it was similar in efficacy to urofollitropin, but in generally larger comparisons follitropin alpha yielded significantly more oocytes and embryos when given in a lower dosage for a shorter period than highly purified urofollitropin and more oocytes than menotropins. Rates of pregnancy, live births and multiple births did not differ among groups receiving any of these agents; however, these rates are difficult to interpret because of factors such as small sample size and differences in methods of reporting. Follitropin alpha had similar effects to follitropin beta in a small comparative study.

The type of follitropin alpha regimen used has not affected outcome, but the role of a GnRH agonist in ART protocols containing pure FSH is now recognised.

Ovulation Induction in Ovulatory Disorders. In women with WHO group II anovulation, who commonly have polycystic ovary syndrome, levels of FSH and other gonadotrophins are often asynchronous with estrogen production. In these patients, follitropin alpha produces high rates of ovulation (about 64 to 93%) and follicular development (about 60 to 88%) after the first cycle, while the mean number of developed follicles remains within the desired range of 1 to 3. Pregnancy rates per cycle are about 20 to 33%.

No significant differences in any of these outcomes have eventuated between patients receiving follitropin alpha, urofollitropin or highly purified urofollitropin. On the other hand, a long term low-dose regimen of follitropin alpha is associated with a reduced number of follicles and a trend toward fewer multiple births compared with conventional regimens of follitropin alpha or urofollitropin. In women with a history of severe ovarian hyperstimulation syndrome (OHSS) given follitropin alpha, fewer cycles were cancelled but pregnancy rates were similar to those in women given menotropins: no episodes of OHSS recurred.

Women with WHO group I anovulation, who have abnormally low gonado-trophin levels and negligible estrogen levels, require LH as well as FSH for conception. A protocol including follitropin alpha 150 IU/day and recombinant human LH in a dosage of 75 or 225 IU/day resulted in follicular development in 79% and pregnancy in 16% of 19 such women in the only clinical trial reported to date.

Male Infertility. One case report has described increased sperm density within 3 months of follitropin alpha therapy in a man with azoospermia and, subsequently, 2 successful pregnancies in his partner. A phase III trial of follitropin alpha in infertile men is ongoing.

Tolerability

Clinical trials have provided few specific details on adverse events during follitropin alpha therapy. However, the drug appears well tolerated. General events such as headache, nausea and ovarian cyst are most common but may be related to increased estrogen levels rather than to the drug per se.

Tolerability has not differed between follitropin alpha and other FSH products. Local irritations at the injection site were mild or infrequent in most trials regardless of which FSH formulation was used. The drug was successfully administered to a patient with a severe allergic reaction to urofollitropin. Subcutaneous formulation of follitropin alpha improves the ease of self-administration.

OHSS, the most serious complication, has occurred in 0 to 10% of cycles stimulated with follitropin alpha in clinical trials. OHSS was severe in 1.7% of 540 cycles, an incidence similar to that documented with other FSH products. Preliminary data indicate that implementation of a long term low-dose regimen may reduce the risk of OHSS.

Dosage and Administration

Follitropin alpha is administered subcutaneously. Patients may self-administer this agent after training and only if expert advice is easily available. Dosage is individualised to patient response as monitored by serum estradiol levels and transvaginal ultrasonography.

As ovulation induction in women with anovulation, follitropin alpha is started at a dosage of 75 IU/day. This may be increased by 37.5 IU/day after 14 days, then every 7 days to a maximum of 300 IU/day. Treatment should not exceed 35 days. One injection of hCG 5000IU is given intramuscularly or subcutaneously to induce ovulation after the last follitropin alpha injection.

If response is excessive in women with anovulation, treatment should be stopped, hCG withheld and treatment restarted at a lower dose in the next cycle.

For ovarian stimulation prior to IVF-ET, follitropin alpha should be started once down-regulation has been achieved with a GnRH agonist. The starting dose of 150 or 225 IU/day may be adjusted by 75 to 150 IU/day every 3 to 5 days. Subcutaneous or intramuscular hCG 5000 to 10 000IU is given once after the last day of follitropin alpha therapy.

Follitropin alpha is contraindicated in women with a variety of disorders, including primary ovarian failure, abnormal uterine haemorrhage of undetermined origin, and pituitary and sex hormone-dependent tumours.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Similar content being viewed by others

References

  1. Loumaye E, Martineau I, Piazzi A, et al. Clinical assessment of human gonadotrophins produced by recombinant DNA technology. Hum Reprod 1996 Sep; 11 Suppl. 1: 95–107

    Google Scholar 

  2. Strowitzki T. Biotechnological drugs for reproductive disorders. BioDrugs 1997 Nov; 8(5): 360–70

    Article  PubMed  CAS  Google Scholar 

  3. Harika G, Gabriel R, Quereux C, et al. Hypersensitization to human menopausal gonadotropins with anaphylactic shock syndrome during a fifth in vitro fertilization cycle. J Assist Reprod Genet 1994; 11(1): 51–3

    Article  PubMed  CAS  Google Scholar 

  4. Li TC, Hindle JE. Adverse local reaction to intramuscular injections of urinary-derived gonadotrophins. Hum Reprod 1993; 8(11): 1835–6

    PubMed  CAS  Google Scholar 

  5. Chappel SC, Howies C. Reevaluation of the roles of luteinizing hormone and follicle-stimulating hormone in the ovulatory process. Hum Reprod 1991 Oct; 6: 1206–12

    PubMed  CAS  Google Scholar 

  6. Giudice E, Crisci C, Eshkol A, et al. Composition of commercial gonadotrophin preparations extracted from human postmenopausal urine: characterization of non-gonadotrophin proteins. Hum Reprod 1994 Dec; 9: 2291–9

    PubMed  CAS  Google Scholar 

  7. le-Cotonnec JY, Porchet HC, Beltrami V, et al. Comparative pharmacokinetics of two urinary human follicle stimulating hormone preparations in healthy female and male volunteers. Hum Reprod 1993 Oct; 8: 1604–11

    PubMed  CAS  Google Scholar 

  8. O’Dea L, Banks K, Study 4550 Investigator Group. Metrodin(™) and Metrodin XP(™): comparative dynamics of folliculogenesis and the estrogen response [abstract]. Fertil Steril 1996 Nov 2 Program Suppl.: S190

  9. O’Dea L, Goodfellow JR. A phase III, open, comparative, randomized multicenter study to assess the safety and efficacy of highly-purified follicle-stimulating hormone (Metrodin XP) administered subcutaneously vs Metrodin administered intramuscularly in WHO group II infertile women. Serono Labs Inc. Study 4550. (Data on file)

  10. Jennings JC, Moreland K, Paterson CM. In vitro fertilisation: a review of drug therapy and clinical management. Drugs 1996 Sep; 52: 313–43

    Article  PubMed  CAS  Google Scholar 

  11. Chappel SC. Heterogeneity of follicle stimulating hormone: control and physiological function. Hum Reprod Update 1995; 1(5): 479–87

    Article  PubMed  CAS  Google Scholar 

  12. Howies CM. III. Expression of human FSH (Gonal-F™) by recombinant DNA technology. Hum Reprod Update 1996; 2 (2 Genetic engineering of human FSH (Gonal-F™)): 183-91

  13. Product news from Europe. First recombinant hFSH approved for infertility. Inpharma 1995 Jan 28; 971: 22

  14. Hillier SG. Cuurent concepts of the roles of follicle stimulating hormone and luteinizing hormone in folliculogenesis. Hum Reprod 1994; 9(2): 188–91

    PubMed  CAS  Google Scholar 

  15. Chapter 313. Neuroendocrine regulation and diseases of the anterior pituitary and hypothalamus. In: Wilson JD, Braunwald E, Isselbacher KJ, et al., editors. Harrison’s principles of internal medicine. 12th ed. Vol. 2. New York: McGraw-Hill Inc, 1991: 1665; 1778–9

    Google Scholar 

  16. Part Twelve. Endocrinology and metabolism. In: Wilson JD, Braunwald E, Isselbacher KJ, et al., editors. HaHrrison’s principles of internal medicine. 12th ed. Vol. 2. New York: McGraw-Hill Inc., 1991: 1766–7

    Google Scholar 

  17. Loumaye E, Eshkol A, Galazka A, et al. Recombinant follicle stimulating hormone: development of the first biotechnology product for treatment of infertility. Ares-Serono International S.A., Switzerland. (Data on file)

  18. Porchet HC, le-Cotonnec J-Y, Loumaye E. Clinical pharmacology of recombinant human follicle-stimulating hormone. III. Pharmacokinetic-pharmacodynamic modeling after repeated subcutaneous administration. Fertil Steril 1994 Apr; 61: 687–95

    PubMed  CAS  Google Scholar 

  19. McLachlan RI, Wreford NG, de -Kretser DM, et al. The effects of recombinant follicle-stimulating hormone on the restoration of spermatogenesis in the gonadotropin-releasing hormone-immunized adult rat. Endocrinology 1995 Sep; 136: 4035–43

    Article  PubMed  CAS  Google Scholar 

  20. Hikim AP, Swerdloff RS. Temporal and stage-specific effects of recombinant human follicle-stimulating hormone on the maintenance of spermatogenesis in gonadotropin-releasing hormone antagonist-treated rat. Endocrinology 1995 Jan; 136: 253–61

    Article  PubMed  CAS  Google Scholar 

  21. Russell LD, Corbin TJ, Borg KE, et al. Recombinant human follicle-stimulating hormone is capable of exerting a biological effect in the adult hypophysectomized rat by reducing the numbers of degenerating germ cells. Endocrinology 1993 Nov; 133: 2062–70

    Article  PubMed  CAS  Google Scholar 

  22. Vihko KK, LaPolt PS, Nishimori K, et al. Stimulatory effects of recombinant follicle-stimulating hormone on Leydig cell function and spermatogenesis in immature hypophysectomized rats. Endocrinology 1991 Oct; 129: 1926–32

    Article  PubMed  CAS  Google Scholar 

  23. Singh J, Handelsman DJ. The effects of recombinant FSH on testosterone-induced spermatogenesis in gonadotrophin-deficient (hpg) mice. J Androl 1996; 17(4): 382–93

    PubMed  CAS  Google Scholar 

  24. Biffoni M, Cantelmo A, Marcucci I, et al. General pharmacology studies on recombinant human follicle stimulating hormone. Arzneimittel Forschung 1994 Sep; 44: 1096–102

    PubMed  CAS  Google Scholar 

  25. Siebold B. Physicochemical characterization of recombinant human follicle stimulating hormone. Hum Reprod 1996 Sep; 11 Suppl. 1: 109–15

    Google Scholar 

  26. Wide L. Isoforms of human gonadotrophins under different physiological conditions. In: Kahn JA, editor. Gonadotrophin isoforms: facts and future. Copenhagen: Ciconia Foundation, 1997: 43–8

    Google Scholar 

  27. Bishop LA, Robertson DM, Cahir N, et al. Specific roles for the asparagine-linked carbohydrate residues of recombinant human follicle stimulating hormone in receptor binding and signal transduction. Mol Endocrinol 1994 Jun; 8: 722–31

    Article  PubMed  CAS  Google Scholar 

  28. Cerpa-Poljack A, Bishop LA, Hort YJ, et al. Isoelectric charge of recombinant human follicle-stimulating hormone isoforms determines receptor affinity and invitro bioactivity. Endocrinology 1993; 132(1): 351–6

    Article  Google Scholar 

  29. Galway AB, Hsueh AJ, Keene JL, et al. In. vitro and in vivo bioactivity of recombinant human follicle-stimulating hormone and partially deglycosylated variants secreted by transfected eukaryotic cell lines. Endocrinology 1990 Jul; 127: 93–100

    Article  PubMed  CAS  Google Scholar 

  30. Bishop LA, Nguyen TV, Schofield PR. Both of the β-subunit carbohydrate residues of follicle-stimulating hormone determine the metabolic clearance rate and in vivo potency. Endocrinology 1995; 136(6): 2635–40

    Article  PubMed  CAS  Google Scholar 

  31. Chappel S. The choice of gonadotrophin isoforms in present and future preparations: what can we expect? In: Kahn JA, editor. Gonadotrophin isoforms: facts and future. Copenhagen: Ciconia Foundaton, 1997: 61–9

    Google Scholar 

  32. Robertson WR. Gonadotrophin isoform pattern in different pharmaceutical preparations. In: Kahn JA, editor. Gonadotrophin isoforms: facts and future. Copenhagen: Ciconia Foundation, 1997: 53–60

    Google Scholar 

  33. Hillier SG, Smyth CD, Whitelaw PF, et al. Gonadotrophin control of follicular function. Horm Res 1995 May; 43: 216–23

    Article  PubMed  CAS  Google Scholar 

  34. Weston AM, Zelinski-Wooten MB, Hutchison JS, et al. Developmental potential of embryos produced by in-vitro fertilization from gonadotrophin-releasing hormone antagonist-treated macaques stimulated with recombinant human follicle stimulating hormone alone or in combination with luteinizing hormone. Hum Reprod 1996 Mar; 11: 608–13

    Article  PubMed  CAS  Google Scholar 

  35. Zelinski-Wooten MB, Hutchison JS, Hess DL, et al. Follicle stimulating hormone alone supports follicle growth and oocyte development in gonadotrophin-releasing hormone antagonist-treated monkeys. Hum Reprod 1995 Jul; 10: 1658–66

    PubMed  CAS  Google Scholar 

  36. Lockwood GM, Muttukrishna S, Groome NP, et al. Circulating inhibins and activin A during GnRH-analogue down-regulation and ovarian hyperStimulation with recombinant FSH for in-vitro fertilization-embryo transfer. Clin Endocrinol 1996; 45: 741–8

    Article  CAS  Google Scholar 

  37. Hoppe I, Starker W, Michels W, et al. Endocrinological pattern in ovarian follicles after controlled hyperStimulation for IVF — comparison between two recombinant FSH preparations [abstract no. PP-08-123]. J Assist Reprod Genet 1997; 14(5) Suppl.: 125S

    Google Scholar 

  38. Raivio T, Toppari J, Perheentupa A, et al. Treatment of prepubertal gonadotrophin-deficient boys with recombinant human follicle-stimulating hormone. Lancet 1997 Jul 26; 350: 263–4

    Article  PubMed  CAS  Google Scholar 

  39. Meachem SJ, McLachlan RI, De Kretser-DM, et al. Neonatal exposure of rats to recombinant follicle stimulating hormone increases adult Sertoli and spermatogenic cell numbers. Biol Reprod 1996; 54(1): 36–44

    Article  PubMed  CAS  Google Scholar 

  40. Ben-Rafael Z, Levy T, Schoemaker J. Pharmacokinetics of follicle-stimulating hormone: clinical significance. Fertil Steril 1995 Apr; 63(4): 689–700

    PubMed  CAS  Google Scholar 

  41. le -Cotonnec JY, Porchet HC, Beltrami V, et al. Clinical pharmacology of recombinant human follicle-stimulating hormone (FSH). I. Comparative pharmacokinetics with urinary human FSH. Fertil Steril 1994 Apr; 61: 669–78

    PubMed  Google Scholar 

  42. le Cotonnec J-Y, Porchet HC, Beltrami V, et al. Clinical pharmacology of recombinant human follicle-stimulating hormone. II. Single doses and steady state pharmacokinetics. Fertil Steril 1994 Apr; 61(4): 679–86

    Google Scholar 

  43. Karlsson MO, Wade JR, Loumaye E, et al. The population pharmacokinetics of recombinant and urinary-hormone follicle stimulating hormone in women. Br J Clin Pharmacol. In press

  44. Karlsson MO, Wade JR, Loumaye E, et al. A population model for the follicular growth in follicle stimulating hormone treated women,. Clin Pharmacol Ther 1997; 62: 665–74

    Article  PubMed  CAS  Google Scholar 

  45. Collins JA, Hughes EG. Pharmacological interventions for the induction of ovulation. Drags 1995 Sep; 50: 480–94

    Article  CAS  Google Scholar 

  46. Schlegel PN, Girardi SK. Clinical review 87. In vitro fertilisation for male factor infertility. J Clin Endocrinol Metab 1997; 82(3): 709–16

    Article  PubMed  CAS  Google Scholar 

  47. Hull M. Complications of pregnancy after infertility treatment: awareness and prevention. Br J Obstet Gynaecol 1995 Jul; 102: 520–4

    Article  PubMed  CAS  Google Scholar 

  48. Strowitzki T, Kentenich H, Kiesel L, et al. Ovarian stimulation in women undergoing in-vitro fertilization and embryo transfer using recombinant human follicle stimulating hormone (Gonal-F) in non-down-regulated cycles. Hum Reprod 1995 Dec; 10: 3097–101

    PubMed  CAS  Google Scholar 

  49. Fisch B, Avrech OM, Pinkas H, et al. Superovulation before IVF by recombinant versus urinary human FSH (combined with a long GnRH analog protocol): a comparative study. J Assist Reprod Genet 1995 Jan; 12: 26–31

    Article  PubMed  CAS  Google Scholar 

  50. Salat-Baroux J, Antoine JM, Loumaye E, et al. ’step-down’ versus ’step-up’ follicular stimulation in normo-ovulatory women undergoing in vitro fertilization: a prospective randomized study. Ares-Serono International SA, Switzerland. Study report no. 7324. (Data on file)

  51. Reddy R, Al-Oum M, Ledger W, et al. An alternate day step-down regimen using Gonal-F (rh-FSH) in IVF: a UK multi-centre study [abstract no. P076]. Abstracts of the 12th Annual Meeting of the European Society of Human Reproduction and Embryology (ESHRE); 1996 Jun 30–Jul 3; Maastricht: 130

  52. Recombinant Human FSH Study Group. Clinical assessment of recombinant human follicle-stimulating hormone in stimulating ovarian follicular development before in vitro fertilization. Fertil Steril 1995 Jan; 63(1): 77–86

    Google Scholar 

  53. Bergh C, Howies CM, Borg K, et al. Recombinant human follicle stimulating hormone (r-hFSH; Gonal-F) versus highly purified urinary FSH (Metrodin HP): results of a randomized comparative study in women undergoing assisted reproductive techniques. Hum Reprod 1997; 12(10): 2133–9

    Article  PubMed  CAS  Google Scholar 

  54. O’Dea L, Loumaye E, Liu H. A randomized, comparative, multicenter clinical trial of recombinant and urinary human FSH in in vitro fertilization and embryo transfer (IVF-ET) [abstract]. Fertil Steril 1993 Program Suppl.: S50-51

  55. Reyftmann L, Déchaud H, Arnal F, et al. Comparaison de l’efficacité de Gonal F versus Humegon sur les résultats de la stimulation ovarienne pour FIV [in French]. Presented at the 2nd Conference of the Fédération Française d’Etude de la Reproduction (FFER); 1997 Sep 24–26; Clermont-Ferrand.

  56. Khalaf Y, Taylor A, Pettigrew R, et al. The relative clinical efficacy of recombinant follicle stimulating hormone to the highly purified urinary preparation [abstract]. Presented at Treatment of Infertility: the New Frontiers; 1998 Jan 22–24; Boca Raton

  57. Ares-Serono International SA. Gonal F versus Puregon. Study no. GF 9180. (data on file)

  58. Ares-Serono International SA. Study no. GF 8407 (Data on file)

  59. Follitropin alfa (Gonal-F). The standard for a new era. Product information, Ares-Sereno International S.A

  60. Loumaye E, Engrand P, Howies CM, et al. Assessment of the role of serum luteinizing hormone and estradiol response to follicle-stimulating hormone on in vitro fertilisation treatment outcome. Fertil Steril 1997 May; 67(5): 889–99

    Article  PubMed  CAS  Google Scholar 

  61. Conway GS. Polycystic ovary syndrome: clinical aspects. Ballière’s Clin Endocrin Metab 1996 Apr; 10(2): 263–79

    Article  CAS  Google Scholar 

  62. Hedon B, Hugues JN, Emperaire JC, et al. A comparative prospective study of a chronic low dose versus a conventional ovulation stimulation regimen using r-hFSH in anovulatory infertile women. Report no. GF 8220, Ares-Serono International SA. (Data on file)

  63. Homburg R, Levy T, Ben-Rafael Z. A comparative prospective study of conventional regimen with chronic low-dose administration of follicle-stimulating hormone for anovulation associated with polycystic ovary syndrome. Fertil Steril 1995 Apr; 63: 729–33

    PubMed  CAS  Google Scholar 

  64. Yanaihara T, Satoh K, Taketani Y, et al. A phase III double blind, controlled study of r-hFSH to induce ovulation in WHO group II anovulatory infertile women [in Japanese]. Jpn Pharmacol Ther 1996; 24(1): 107–25

    Google Scholar 

  65. Aboulghar MA, Amin YM, Mansour RT, et al. Recombinant follicle-stimulating hormone in the treatment of patients with history of severe ovarian hyperStimulation syndrome. Fertil Steril 1996 Nov; 66: 757–60

    PubMed  CAS  Google Scholar 

  66. Hull M, Corrigan E, Piazzi A, et al. Recombinant human luteinising hormone: an effective new gonadotropin preparation. Lancet 1994; 344(8918): 334–5

    Article  PubMed  CAS  Google Scholar 

  67. Kousta E, White DM, Piazzi A, et al. Successful induction of ovulation and completed pregnancy using recombinant human luteinizing hormone and follicle stimulating hormone in a woman with Kallmann’s syndrome. Hum Reprod 1996 Jan; 11: 70–1

    Article  PubMed  CAS  Google Scholar 

  68. European Recombinant Human LH Study Group. Recombinant human luteinizing hormone (r-hLH) to support recombinant human follicle stimulating hormone (r-hFSH)-induced development in LH and FSH deficient anovulatory women: a dose- finding study. J Clin Endocrinol Metab. In press

  69. Quinton R, Matfin G, Bouloux P, et al. Successful induction of fertility in a hypogonadotropic male [letter]. Lancet 1994 Mar 19; 343: 733

    Article  PubMed  CAS  Google Scholar 

  70. Quinton R, Bouloux P-MG, Illingworth JM, et al. Efficacy of recombinant follicle-stimulating hormone [letter]. Fertil Steril 1996; 66(1): 164

    PubMed  CAS  Google Scholar 

  71. Gonal-F. Prescribing information for follitropin alpha. Serono Laboratories, Inc. Aubanne, Switzerland, September 1997

    Google Scholar 

  72. Phipps WR, Holden D, Sheehan RD. Use of recombinant human follicle-stimulating hormone for in vitro fertilization-embryo transfer after severe systemic immunoglobulin E-mediated reaction to urofollitropin. Fertil Steril 1996 Jul; 66: 148–50

    PubMed  CAS  Google Scholar 

  73. Golan A, Ron-El R, Herman A, et al. Ovarian hyperStimulation syndrome: an update review. Obstet Gynecol Surv 1989; 44(6): 430–40

    Article  PubMed  CAS  Google Scholar 

  74. Rizk B, Smitz J. Ovarian hyperStimulation syndrome after superovulation using GnRH agonists for IVF and related procedures. Hum Reprod 1992; 7(3): 320–7

    PubMed  CAS  Google Scholar 

  75. McDonough PG. The coming of wonders: editorial comment. Fertil Steril 1997 Feb; 67: 412–3

    Article  Google Scholar 

  76. Serono’s Gonal-F & SB’s Hycamtin launched in Canada. Scrip 1997 Jul 29 (2253): 20

  77. Recombinant FSH approved in the US. Scrip 1997 Oct 7; 2273: 21

  78. RPR’s Taxotere — 2nd EU approval. Scrip 1997 Dec 5; 2083: 18

    Google Scholar 

  79. Germond M, Dessole S, Senn A, et al. Successful in-vitro fertilisation and embryo transfer after treatment with recombinant human FSH. Lancet 1992 May 9; 339: 1170

    Article  PubMed  CAS  Google Scholar 

  80. Hornnes P, Giroud D, Howies C, et al. Recombinant human follicle-stimulating hormone treatment leads to normal follicular growth, estradiol secretion, and pregnancy in a World Health Organization group II anovulatory woman. Fertil Steril 1993 Oct; 60(4): 724–6

    PubMed  CAS  Google Scholar 

  81. 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 Nov; 58(5): 888–96

    PubMed  CAS  Google Scholar 

  82. Daya S, Gunby J, Hughes EG, et al. Follicle-stimulating hormone versus human menopausal gonadotropin for in vitro fertilization cycles: a meta-analysis. Fertil Steril 1995 Aug; 64(2): 347–54

    PubMed  CAS  Google Scholar 

  83. FIVNAT. Stimulation de l’ovulation. Contracept Fertil Sex 1996; 24(9): 710–2

    Google Scholar 

  84. Organon Inc. Follitropin beta. Prescribing information. West Orange, New Jersey, USA. Sep 1997

  85. Out HJ, Mannaerts BMJL, Driessen SGAJ, et al. A prospective, randomized, assessor-blind, multicentre study comparing recombinant and urinary follicle stimulating hormone (Puregon versus Metrodin) in in-vitro fertilization. Hum Reprod 1995 Oct; 10: 2534–40

    PubMed  CAS  Google Scholar 

  86. Out HJ, Mannaerts BMJL, Driessen SGAJ, et al. Recombinant follicle stimulating hormone (rFSH; Puregon) in assisted reproduction: more oocytes, more pregnancies. Results from five comparative studies. Hum Reprod Update 1996 Mar–Apr; 2: 162–71

    Article  PubMed  CAS  Google Scholar 

  87. Geurts TBP, Peters MJH, Van Bruggen-JGC, et al. Puregon(™) (ORG 32489) — recombinant human follicle-stimulating hormone. Drugs Today 1996; 32(3): 239–58

    CAS  Google Scholar 

  88. Out HJ, Olijve W, Coelingh Bennink HJT. The coming of wonders [letter]. Fertil Steril 1997 Feb; 67: 411

    Article  PubMed  CAS  Google Scholar 

  89. Agrawal R, West C, Conway GS, et al. Pregnancy after treatment with three recombinant gonadotropins. Lancet 1997; 349: 29–30

    Article  PubMed  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Karen L. Goa.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Goa, K.L., Wagstaff, A.J. Follitropin Alpha in Infertility. BioDrugs 9, 235–260 (1998). https://doi.org/10.2165/00063030-199809030-00006

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.2165/00063030-199809030-00006

Keywords

Navigation