89
Views
0
CrossRef citations to date
0
Altmetric
Review

Investigational follicle-stimulating hormone receptor agonists for male infertility therapy

ORCID Icon, , ORCID Icon, ORCID Icon, ORCID Icon & ORCID Icon
Pages 813-824 | Received 07 Jun 2023, Accepted 21 Sep 2023, Published online: 29 Sep 2023

References

  • Zegers-Hochschild F, Adamson GD, Dyer S, et al. The international glossary on infertility and fertility care. Fertil Steril. 2017;108(3):393–406. doi: 10.1016/j.fertnstert.2017.06.005
  • [cited 2023 Aug 20]. https://www.who.int/news/item/04-04-2023-1-in-6-people-globally-affected-by-infertility
  • Thonneau P, Marchand S, Tallec A, et al. Incidence and main causes of infertility in a resident population (1,850,000) of three French regions (1988-1989). Hum Reprod. 1991;6(6):811–816. doi: 10.1093/oxfordjournals.humrep.a137433
  • Agarwal A, Mulgund A, Hamada A, et al. A unique view on male infertility around the globe. Reprod Biol Endocrinol. 2015;13(1):37. doi: 10.1186/s12958-015-0032-1
  • Sun H, Gong TT, Jiang YT, et al. Global, regional, and national prevalence and disability-adjusted life-years for infertility in 195 countries and territories, 1990–2017: results from a global burden of disease study, 2017. Aging. 2019;11(23):10952–10991. doi: 10.18632/aging.102497
  • Levine H, Jørgensen N, Martino-Andrade A, et al. Temporal trends in sperm count: a systematic review and meta-regression analysis of samples collected globally in the 20th and 21st centuries. Hum Reprod Update. 2022 Nov 15;29(2):157–176. Epub ahead of print. doi: 10.1093/humupd/dmac035
  • Jørgensen N, Lamb DJ, Levine H, et al. Are worldwide sperm counts declining? Fertil Sterility. 2021;116(6):1457–1463. doi: 10.1016/j.fertnstert.2021.10.020
  • Crafa A, Cannarella R, LA Vignera S, et al. Semen analysis: a workflow for an appropriate assessment of the male fertility status. Minerva Endocrinol (Torino). 2022;47(1):77–88. doi: 10.23736/S2724-6507.21.03650-2
  • Punab M, Poolamets O, Paju P, et al. Causes of male infertility: a 9-year prospective monocentre study on 1737 patients with reduced total sperm counts. Hum Reprod. 2017;32(1):18–31. doi: 10.1093/humrep/dew284
  • Tüttelmann F, Ruckert C, Röpke A. Disorders of spermatogenesis: perspectives for novel genetic diagnostics after 20 years of unchanged routine. Med Genet. 2018;30(1):12–20. doi: 10.1007/s11825-018-0181-7
  • Lopushnyan NA, Walsh TJ. Surgical techniques for the management of male infertility. Asian J Androl. 2012;14(1):94–102. doi: 10.1038/aja.2011.62
  • Schlegel PN, Sigman M, Collura B, et al. Diagnosis and treatment of infertility in men: AUA/ASRM guideline PART II. J Urol. 2021;205(1):44–51. doi: 10.1097/JU.0000000000001520
  • Calogero AE, Condorelli RA, Russo GI, et al. Conservative nonhormonal options for the treatment of male infertility: antibiotics, anti-inflammatory drugs, and antioxidants. Biomed Res Int . 2017;2017:1–17. (2017). doi: 10.1155/2017/4650182
  • La Vignera S, Condorelli RA, Cimino L, et al. Late-onset hypogonadism: the advantages of treatment with human chorionic gonadotropin rather than testosterone. Aging Male. 2016;19(1):34–39. doi: 10.3109/13685538.2015.1092021
  • Vicari E, Mongioì A, Calogero AE, et al. Therapy with human chorionic gonadotropin alone induces spermatogenesis in men with isolated hypogonadotrophic hypogonadism–long-term follow-up. Int J Androl. 1992;15(4):320–329. doi: 10.1111/j.1365-2605.1992.tb01131.x
  • Tadros NN, Sabanegh ES. Empiric medical therapy with hormonal agents for idiopathic male infertility. Indian J Urol. 2017;33(3):194–198. doi: 10.4103/iju.IJU_368_16
  • Duca Y, Calogero AE, Cannarella R, et al. Current and emerging medical therapeutic agents for idiopathic male infertility. Expert Opin Pharmacother. 2019;20(1):55–67. doi: 10.1080/14656566.2018.1543405
  • Cannarella R, La Vignera S, Condorelli RA, et al. FSH dosage effect on conventional sperm parameters: a meta-analysis of randomized controlled studies. Asian J Androl. 2020;22(3):309–316. doi: 10.4103/aja.aja_42_19
  • Ko EY, Siddiqi K, Brannigan RE, et al. Empirical medical therapy for idiopathic male infertility: a survey of the American Urological Association. J Urol. 2012;187(3):973–978. doi: 10.1016/j.juro.2011.10.137
  • Kaiser UB, Conn PM, Chin WW. Studies of gonadotropin-releasing hormone (GnRH) action using GnRH receptor-expressing pituitary cell lines. Endocr Rev. 1997;18(1):46–70. doi: 10.1210/edrv.18.1.0289
  • Das N, Kumar TR. Molecular regulation of follicle-stimulating hormone synthesis, secretion and action. J Mol Endocrinol. 2018;60(3):R131–55. doi: 10.1530/JME-17-0308
  • O’Donnell L, Smith LB, Rebourcet D. Sertoli cells as key drivers of testis function. Semin Cell Dev Biol. 2022;121:2–9. doi: 10.1016/j.semcdb.2021.06.016
  • Gondos B, Berndtson WE. Postnatal and pubertal development. In: Russell LD Griswold MD, editors The sertoli cell. Cache River Press, Clearwater: USA; 1993. p. 115–154.
  • Valeri C, Schteingart HF, Rey RA. The prepubertal testis: biomarkers and functions. Curr Opin Endocrinol Diabetes Obes. 2013;20(3):224–233. doi: 10.1097/MED.0b013e328360be2c
  • Condorelli RA, Cannarella R, Calogero AE, et al. Evaluation of testicular function in prepubertal children. Endocrine. 2018;62(2):274–280. doi: 10.1007/s12020-018-1670-9
  • Adan L, Lechevalier P, Couto-Silva AC, et al. Plasma inhibin B and antimüllerian hormone concentrations in boys: discriminating between congenital hypogonadotropic hypogonadism and constitutional pubertal delay. Med Sci Monit. 2010;16(11):CR511–7.
  • Sá R, Neves R, Fernandes S, et al. Cytological and expression studies and quantitative analysis of the temporal and stage-specific effects of follicle-stimulating hormone and testosterone during cocultures of the normal human seminiferous epithelium. Biol Reprod. 2008;79(5):962–975. doi: 10.1095/biolreprod.107.067546
  • Wang JM, Li ZF, Yang WX, et al. Follicle-stimulating hormone signaling in Sertoli cells: a licence to the early stages of spermatogenesis. Reprod Biol Endocrinol. 2022;20(1):97. doi: 10.1186/s12958-022-00971-w
  • Casarini L, Crépieux P. Molecular mechanisms of action of FSH. Front Endocrinol. 2019;10:305. doi: 10.3389/fendo.2019.00305
  • Gromoll J, Pekel E, Nieschlag E. The structure and organization of the human follicle-stimulating hormone receptor (FSHR) gene. Genomics. 1996;35(2):308–311. doi: 10.1006/geno.1996.0361
  • Sun L, Peng Y, Sharrow AC, et al. FSH directly regulates bone mass. Cell. 2006;125(2):247–260. doi: 10.1016/j.cell.2006.01.051
  • Robinson LJ, Tourkova I, Wang Y, et al. FSH-receptor isoforms and FSH-dependent gene transcription in human monocytes and osteoclasts. Biochem Biophys Res Commun. 2010;394(1):12–17. doi: 10.1016/j.bbrc.2010.02.112
  • Song Y, Wang ES, Xing LL, et al. Follicle-stimulating hormone induces postmenopausal dyslipidemia through inhibiting hepatic cholesterol metabolism. J Clin Endocrinol Metab. 2016;101(1):254–263. doi: 10.1210/jc.2015-2724
  • Stilley JA, Christensen DE, Dahlem KB, et al. FSH receptor (FSHR) expression in human extragonadal reproductive tissues and the developing placenta, and the impact of its deletion on pregnancy in mice. Biol Reprod. 2014;91(3):74. doi: 10.1095/biolreprod.114.118562
  • Siraj A, Desestret V, Antoine M, et al. Expression of follicle-stimulating hormone receptor by the vascular endothelium in tumor metastases. BMC Cancer. 2013;13(1):246. doi: 10.1186/1471-2407-13-246
  • Song GJ, Park YS, Lee YS, et al. Alternatively spliced variants of the follicle-stimulating hormone receptor gene in the testis of infertile men. Fertil Steril. 2002;77(3):499–504. doi: 10.1016/S0015-0282(01)03221-6
  • Sairam MR, Babu PS. The tale of follitropin receptor diversity: a recipe for fine tuning gonadal responses? Mol Cell Endocrinol. 2007;260-2:163–171. doi: 10.1016/j.mce.2005.11.052
  • Panza S, Giordano F, De Rose D, et al. FSH-R human early male genital tract, testicular tumors and sperm: its involvement in testicular disorders. Life. 2020;10(12):336. doi: 10.3390/life10120336
  • Cannarella R, Mancuso F, Barone N, et al. Effects of follicle-stimulating hormone on human sperm motility in vitro. Int J Mol Sci. 2023;24(7):6536. doi: 10.3390/ijms24076536
  • Choenfeld C, Amelar RD, Dubin L, et al. Follicle-stimulating hormone, luteinizing hormone, and testosterone levels found in human seminal plasma. Fertil Steril. 1978;29(1):69–71. doi: 10.1016/S0015-0282(16)43040-2
  • Shirai M, Matsuda S, Mitsukawa S, et al. FSH, LH and testosterone levels in human seminal plasma. Tohoku J Exp Med. 1975;116(2):201–202. doi: 10.1620/tjem.116.201
  • Wu S, Yan M, Ge R, et al. Crosstalk between Sertoli and germ cells in male fertility. Trends Mol Med. 2020;26(2):215–231. doi: 10.1016/j.molmed.2019.09.006
  • Riera MF, Regueira M, Galardo MN, et al. Signal transduction pathways in FSH regulation of rat Sertoli cell proliferation. Am J Physiol Endocrinol Metab. 2012;302(8):E914–23. doi: 10.1152/ajpendo.00477.2011
  • Cannarella R, Mancuso F, Condorelli RA, et al. Effects of GH and IGF1 on basal and FSH-Modulated porcine Sertoli cells in-vitro. J Clin Med. 2019;8(6):811. doi: 10.3390/jcm8060811
  • Cannarella R, Condorelli RA, La Vignera S, et al. Effects of the insulin-like growth factor system on testicular differentiation and function: a review of the literature. Andrology. 2018;6(1):3–9. doi: 10.1111/andr.12444
  • Pitetti JL, Calvel P, Zimmermann C, et al. An essential role for insulin and IGF1 receptors in regulating sertoli cell proliferation, testis size, and FSH action in mice. Mol Endocrinol. 2013;27(5):814–827. doi: 10.1210/me.2012-1258
  • Cannarella R, Arato I, Condorelli RA, et al. The IGF1 receptor is involved in follicle-stimulating hormone signaling in porcine neonatal Sertoli cells. J Clin Med. 2019;8(5):577. doi: 10.3390/jcm8050577
  • La Vignera S, Condorelli RA, Duca Y, et al. FSH treatment for normogonadotropic male infertility: a synergistic role for metformin? Eur Rev Med Pharmacol Sci. 2019;23(13):5994–5998. doi: 10.1080/13685538.2019.1590696
  • Nordenström A, Ahmed SF, van den Akker E, et al. Pubertal induction and transition to adult sex hormone replacement in patients with congenital pituitary or gonadal reproductive hormone deficiency: an endo-ERN clinical practice guideline. Eur J Endocrinol. 2022;186(6):G9–G49. doi: 10.1530/EJE-22-0073
  • Salonia A, Bettocchi C, Boeri L, et al. European Association of urology guidelines on sexual and reproductive Health-2021 update: male sexual dysfunction. Eur Urol. 2021Sep;80(3):333–357. doi: 10.1016/j.eururo.2021.06.007
  • Chemes HE, Dym M, Raj HG. Hormonal regulation of Sertoli cell differentiation. Biol Reprod. 1979;21(1):251–262. doi: 10.1095/biolreprod21.1.251
  • Dwyer AA, Sykiotis GP, Hayes FJ, et al. Trial of recombinant follicle-stimulating hormone pretreatment for GnRH-induced fertility in patients with congenital hypogonadotropic hypogonadism. J Clin Endocrinol Metab. 2013;98(11):E1790–E5. doi: 10.1210/jc.2013-2518
  • Acosta AA, Khalifa E, Oehninger S. Pure human follicle stimulating hormone has a role in the treatment of severe male infertility by assisted reproduction: Norfolk’s total experience. Hum Reprod. 1992;7(8):1067–1072. doi: 10.1093/oxfordjournals.humrep.a137794
  • Paradisi R, Natali F, Fabbri R, et al. Evidence for a stimulatory role of high doses of recombinant human follicle-stimulating hormone in the treatment of male-factor infertility. Andrologia. 2014;46(9):1067–1072. doi: 10.1111/and.12194
  • Paradisi R, Busacchi P, Seracchioli R, et al. Effects of high doses of recombinant human follicle-stimulating hormone in the treatment of male factor infertility: results of a pilot study. Fertil Steril. 2006;86(3):728–731. doi: 10.1016/j.fertnstert.2006.02.087
  • Kamischke A, Behre HM, Bergmann M, et al. Recombinant human follicle stimulating hormone for treatment of male idiopathic infertility: a randomized, double-blind, placebo-controlled, clinical trial. Hum Reprod. 1998;13(3):596–603. doi: 10.1093/humrep/13.3.596
  • Ding YM, Zhang XJ, Li JP. Treatment of idiopathic oligozoospermia with recombinant human follicle-stimulating hormone: a prospective, randomized, double-blind, placebo-controlled clinical study in Chinese population. Clin Endocrinol (Oxf). 2015;83(6):866–871. doi: 10.1111/cen.12770
  • Efesoy O, Cayan S, Akbay E. The efficacy of recombinant human follicle-stimulating hormone in the treatment of various types of male-factor infertility at a single university hospital. J Androl. 2009;30(6):679–684. doi: 10.2164/jandrol.108.007278
  • Colacurci N, Monti MG, Fornaro F, et al. Recombinant human FSH reduces sperm DNA fragmentation in men with idiopathic oligoasthenoteratozoospermia. J Androl. 2012;33(4):588–593. doi: 10.2164/jandrol.111.013326
  • Condorelli RA, Calogero AE, Vicari E, et al. Reduced seminal concentration of CD45pos cells after follicle-stimulating hormone treatment in selected patients with idiopathic oligoasthenoteratozoospermia. Int J Endocrinol. 2014;2014(2014):1–8. doi: 10.1155/2014/372060
  • Foresta C, Bettella A, Merico M, et al. Use of recombinant human follicle-stimulating hormone in the treatment of male factor infertility. Fertil Steril. 2002;77(2):238–244. doi: 10.1016/S0015-0282(01)02966-1
  • Casamonti E, Vinci S, Serra E, et al. Short-term FSH treatment and sperm maturation: a prospective study in idiopathic infertile men. Andrology. 2017;5(3):414–422. doi: 10.1111/andr.12333
  • Garolla A, Ghezzi M, Cosci I, et al. FSH treatment in infertile males candidate to assisted reproduction improved sperm DNA fragmentation and pregnancy rate. Endocrine. 2017;56(2):416–425. doi: 10.1007/s12020-016-1037-z
  • Baccetti B, Strehler E, Capitani S, et al. The effect of follicle-stimulating hormone therapy on human sperm structure (notulae seminologicae). Hum Reprod. 1997;12(9):1955–1968. doi: 10.1093/humrep/12.9.1955
  • Strehler E, Sterzik K, De Santo M, et al. The effect of follicle-stimulating hormone therapy on sperm quality: an ultrastructural mathematical evaluation. J Androl. 1997;18(4):439–447.
  • Arnaldi G, Balercia G, Barbatelli G, et al. Effects of long-term treatment with human pure follicle-stimulating hormone on semen parameters and sperm-cell ultrastructure in idiopathic oligoteratoasthenozoospermia. Andrologia. 2000;32(3):155–161. doi: 10.1046/j.1439-0272.2000.00358.x
  • Bergmann M, Behre HM, Nieschlag E. Serum FSH and testicular morphology in male infertility. Clin Endocrinol (Oxf). 1994;40(1):133–136. doi: 10.1111/j.1365-2265.1994.tb02455.x
  • Foresta C, Bettella A, Ferlin A, et al. Evidence for a stimulatory role of follicle-stimulating hormone on the spermatogonial population in adult males. Fertil Steril. 1998;69(4):636–642. doi: 10.1016/S0015-0282(98)00008-9
  • Foresta C, Bettella A, Garolla A, et al. Treatment of male idiopathic infertility with recombinant human follicle-stimulating hormone: a prospective, controlled, randomized clinical study. Fertil Steril. 2005;84(3):654–661. doi: 10.1016/j.fertnstert.2005.03.055
  • Garolla A, Selice R, Engl B, et al. Spermatid count as a predictor of response to FSH therapy. Reprod Biomed Online. 2014;29(1):102–112. doi: 10.1016/j.rbmo.2014.02.014
  • Ferlin A, Calogero AE, Krausz C, et al. Management of male factor infertility: position statement from the Italian Society of Andrology and sexual Medicine (SIAMS): endorsing organization: Italian Society of Embryology reproduction, and research (SIERR). J Endocrinol Invest. 2022;45(5):1085–1113. doi: 10.1007/s40618-022-01741-6
  • Peng T, Liao C, Ye X, et al. Gonadotropins treatment prior to microdissection testicular sperm extraction in non-obstructive azoospermia: a single-center cohort study. Reprod Biol Endocrinol. 2022;20(1):61. doi: 10.1186/s12958-022-00934-1
  • Cocci A, Cito G, Russo GI, et al. Effectiveness of highly purified urofollitropin treatment in patients with idiopathic azoospermia before testicular sperm extraction. Urologia. 2018;85(1):19–21. doi: 10.5301/uj.5000253
  • Zhang X, Chen J, Cui Y, et al. FSH can improve semen parameters in patients with idiopathic oligoasthenoteratozoospermia: a systematic review and meta-analysis. Andrologia. 2022;54(11):e14596. doi: 10.1111/and.14596
  • Santi D, Granata AR, Simoni M. FSH treatment of male idiopathic infertility improves pregnancy rate: a meta-analysis. Endocr Connect. 2015;4(3):R46–58. doi: 10.1530/EC-15-0050
  • Olijve W, de Boer W, Mulders JW, et al. Molecular biology and biochemistry of human recombinant follicle stimulating hormone (puregon). Mol Hum Reprod. 1996;2(5):371–382. doi: 10.1093/molehr/2.5.371
  • Zwart-van Rijkom JE, Broekmans FJ, Leufkens HG. From HMG through purified urinary FSH preparations to recombinant FSH: a substitution study. Human Reproduction. 2002;17(4):857–865. doi: 10.1093/humrep/17.4.857
  • Kliesch S, Behre HM, Nieschlag E. Recombinant human follicle-stimulating hormone and human chorionic gonadotropin for induction of spermatogenesis in a hypogonadotropic male. Fertil Steril. 1995;63(6):1326–1328. doi: 10.1016/S0015-0282(16)57619-5
  • Foresta C, Bettella A, Merico M, et al. FSH in the treatment of oligozoospermia. Mol Cell Endocrinol. 2000;161(1–2):89–97. doi: 10.1016/S0303-7207(99)00228-2
  • Palomba S, Falbo A, Espinola S, et al. Effects of highly purified follicle-stimulating hormone on sperm DNA damage in men with male idiopathic subfertility: a pilot study. J Endocrinol Invest. 2011;34(10):747–752. doi: 10.3275/7745
  • Baccetti B, Piomboni P, Bruni E, et al. Effect of follicle-stimulating hormone on sperm quality and pregnancy rate. Asian J Androl. 2004;6(2):133–137.
  • Caroppo E, Niederberger C, Vizziello GM, et al. Recombinant human follicle-stimulating hormone as a pretreatment for idiopathic oligoasthenoteratozoospermic patients undergoing intracytoplasmic sperm injection. Fertil Steril. 2003;80(6):1398–1403. doi: 10.1016/S0015-0282(03)02202-7
  • Barbonetti A, Calogero AE, Balercia G, et al. The use of follicle stimulating hormone (FSH) for the treatment of the infertile man: position statement from the Italian Society of Andrology and sexual Medicine (SIAMS). J Endocrinol Invest. 2018;41(9):1107–1122. doi: 10.1007/s40618-018-0843-y
  • Colpi GM, Francavilla S, Haidl G, et al. European Academy of Andrology guideline management of oligo-astheno-teratozoospermia. Andrology. 2018;6(4):513–524. doi: 10.1111/andr.12502
  • Valenti D, La Vignera S, Condorelli RA, et al. Follicle-stimulating hormone treatment in normogonadotropic infertile men. Nat Rev Urol. 2013;10(1):55–62. doi: 10.1038/nrurol.2012.234
  • Ferlin A, Vinanzi C, Selice R, et al. Toward a pharmacogenetic approach to male infertility: polymorphism of follicle-stimulating hormone beta-subunit promoter. Fertil Steril. 2011;96(6):1344–1349.e2. doi: 10.1016/j.fertnstert.2011.09.034
  • Mongioì LM, Condorelli RA, Alamo A, et al. Follicle-stimulating hormone treatment and male idiopathic infertility: effects on sperm parameters and oxidative stress indices according to FSHR c. 2039 A/G and c. -29 G/A genotypes. J Clin Med. 2020;9(6):1690. doi: 10.3390/jcm9061690
  • Casarini L, Moriondo V, Marino M, et al. FSHR polymorphism p.N680S mediates different responses to FSH in vitro. Mol Cell Endocrinol. 2014;393(1–2):83–91. doi: 10.1016/j.mce.2014.06.013
  • Simoni M, Santi D, Negri L, et al. Treatment with human, recombinant FSH improves sperm DNA fragmentation in idiopathic infertile men depending on the FSH receptor polymorphism p.N680S: a pharmacogenetic study. Hum Reprod. 2016;31(9):1960–1969. doi: 10.1093/humrep/dew167
  • Fernández-Arjona M, Díaz J, Cortes I, et al. Relationship between gonadotrophin secretion, inhibin B and spermatogenesis in oligozoospermic men treated with highly purified urinary follicle-stimulating hormone (uFSH-HP): a preliminary report. Eur J Obstet Gynecol Reprod Biol. 2003;107(1):47–51. doi: 10.1016/S0301-2115(02)00313-5
  • Bartlett JM, Weinbauer GF, Nieschlag E. Differential effects of FSH and testosterone on the maintenance of spermatogenesis in the adult hypophysectomized rat. J Endocrinol. 1989;121(1):49–58. doi: 10.1677/joe.0.1210049
  • Amory JK, Coviello AD, Page ST, et al. Serum 17-hydroxyprogesterone strongly correlates with intratesticular testosterone in gonadotropin-suppressed normal men receiving various dosages of human chorionic gonadotropin. Fertil Steril. 2008;89(2):380–386. doi: 10.1016/j.fertnstert.2007.02.059
  • Lima TFN, Rakitina E, Blachman-Braun R, et al. Evaluation of a serum 17-hydroxyprogesterone as a predictor of semen parameter improvement in men undergoing medical treatment for infertility. Can Urol Assoc J. 2021;15(7):E340–E5. doi: 10.5489/cuaj.6846
  • Mouzannar A, Narasimman M, Patel P, et al. Using 17-OHP as serum biomarker to monitor therapy in patients with hypogonadotropic hypogonadism. Rev Urol. 2019;21(4):180–182.
  • Foresta C, Selice R, Moretti A, et al. Gonadotropin administration after gonadotropin-releasing-hormone agonist: a therapeutic option in severe testiculopathies. Fertil Steril. 2009;92(4):1326–1332. doi: 10.1016/j.fertnstert.2008.07.1766

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.