Skip to main content

REVIEW article

Front. Endocrinol., 18 February 2019
Sec. Reproduction
This article is part of the Research Topic Follicle-Stimulating Hormone: Fertility and Beyond View all 22 articles

Modulation of Gonadotropins Activity by Antibodies

\r\nElodie Kara*Elodie Kara1*Laurence DupuyLaurence Dupuy1Cline Bouillon,,,,,Céline Bouillon1,2,3,4,5,6Sophie CasteretSophie Casteret1Marie-Christine MaurelMarie-Christine Maurel1
  • 1Igyxos SA, Nouzilly, France
  • 2Service de Médecine et Biologie de la Reproduction, CHRU de Tours, Tours, France
  • 3Biologie Intégrative de l'Ovaire, INRA, UMR85, Physiologie de la Reproduction et des Comportements, Nouzilly, France
  • 4CNRS, UMR7247, Nouzilly, France
  • 5Université François Rabelais, Tours, France
  • 6IFCE, Nouzilly, France

Gonadotropins are essential for reproduction control in humans as well as in animals. They are widely used all over the world for ovarian stimulation in women, spermatogenesis stimulation in men, and ovulation induction and superovulation in animals. Despite the availability of many different preparations, all are made of the native hormones. Having different ligands with a wide activity range for a given receptor helps better understand its molecular and cellular signaling mechanisms as well as its physiological functions, and thus helps the development of more specific and adapted medicines. One way to control the gonadotropins' activity could be the use of modulating antibodies. Antibodies are powerful tools that were largely used to decipher gonadotropins' actions and they have shown their utility as therapeutics in several other indications such as cancer. In this review, we summarize the inhibitory and potentiating antibodies to gonadotropins, and their potential therapeutic applications.

Introduction

Gonadotropins, namely follicle-stimulating hormone (FSH), luteinizing hormone (LH), and chorionic gonadotropin (CG) are heterodimeric glycoproteins, constituted by an alpha- and a beta- subunit. The alpha subunit is common to all glycoprotein hormones, including thyroid stimulating hormone (TSH) (1). FSH and LH/CG receptors are G-protein coupled receptors (GPCR), mainly expressed in granulosa cells in female ovaries and in Sertoli cells in male testis for FSH receptor (FSH-R) (2), and in granulosa and theca cells in female and Leydig cells in male for LH receptor (LH-R) (3).

Because of their role in reproduction, gonadotropins are routinely used in fertility treatments in men and women for assisted reproductive technologies (ART) (4, 5). In women, it consists on daily injections of FSH or a mixture of FSH/LH, for 8–12 days, to grow and mature follicles. The final maturation is then completed with an injection of human CG (hCG) 36 h after the last injection of FSH. In men, FSH and hCG injections 2–3 times a week for several months are used to treat hypogonadotropic hypogonadism and induce spermatogenesis (69). Currently, the preparations used are either endogenous FSH extracted from post-menopausal women urine (human menopausal gonadotropins, hMG), highly purified urinary FSH, or recombinant FSH. The first recombinant versions of FSH (follitropin alpha and beta, corifollitropin) and all their biosimilars were produced in Chinese hamster ovary (CHO) cells. The follitropin delta approved in Europe in 2015 (Rekovelle™) and follitropin epsilon still under development are produced in human cell lines: PER.C6 for follitropin delta (10) and GlycoExpress for follitropin epsilon (11). For LH and hCG, recombinant versions produced in CHO cells are also available. In animals, equine CG (eCG), formerly named pregnant-mare serum gonadotropin (PMSG), is widely used to induce ovulation in small ruminants (12). Porcine pituitary extracts are used for current superovulation treatments in cattle (13), whereas ovulation in swine herds is induced with a mixture of hCG and eCG (14).

Despite all the preparations that are on the market, the only ligands available for gonadotropin receptors as therapeutics are native hormones. New biased ligands or ligands with different potencies and efficacies on gonadotropin receptors can help better understand receptor signaling, decipher the implication of the different signaling pathways in physiological and pathophysiological mechanisms, and finally bring to the market new molecules to improve ART treatments. In 2015 in USA, 182,111 ART procedures were performed leading to 59,334 live-birth deliveries (15). Among other strategies like small molecule ligands, one way to modulate the gonadotropins' activity could be the use of antibodies, targeting either directly the receptor itself, or its ligand to modulate hormone's activity.

Antibodies are useful tools that help to better understand gonadotropins' structure by epitope mapping (1618) and their function by neutralizing the effect of endogenous gonadotropins (1922). They also allow their quantification by the development of radioimmunoassays (RIA) and enzyme-linked immunosorbent assays (ELISA) (2327). Finally, antibodies permitted the development of specific purification methods for gonadotropins, making them safer to use as therapeutic agents (2830).

The effect of antibodies, if any, is expected to be inhibitory on antigen activity by impairing its interaction with its receptor. Surprisingly, some of them were described as being able to increase the activity of their antigenic protein. Potentiating polyclonal antibodies directed against epidermal growth factor (EGF) and insulin were first described by Shechter et al. in 1979 (31, 32). A few years later, potentiating monoclonal antibodies (mAbs) were described for ovine (o) and human (h) growth hormones (GH) (3335). When hypopituitary Snell dwarf mice were treated with hGH in complex with a mAb, the actions of hGH on growth and body composition were enhanced compared to animals treated with GH alone (34, 35). The same kind of antibodies were described for TSH: mAbs directed against TSH were able to enhance its biological activity in vivo in Snell dwarf mice (36), suggesting that glycoprotein hormones' activity may be modulated by mAbs. Anti-receptor antibodies with stimulating activities were also described for TSH receptor (37, 38).

In this review, we give a brief overview of antibodies modulating gonadotropins' activity, either positively or negatively.

Structure and Function of Gonadotropins

Alpha- and beta-subunits of gonadotropins are non-covalently linked. The alpha-subunit is common to all glycoprotein hormones in a given species, and presents two major sites of N-glycosylation (1). The specificity of each hormone is conferred by the beta-subunit, that contains 2 N-glycosylation sites. hCG, eLH and eCG present a longer beta-subunit with an additional carboxy-terminal peptide (CTP) that is ~30 amino-acids long and contains multiple O-glycosylation sites.

eCG originates from uterine endometrial cups and is extracted from pregnant mare serum (39, 40). eLH and eCG beta-subunits are encoded by a single gene (41) but they differ in glycosylation. They both exhibit N-glycans on alpha- and beta-subunits, and O-glycans on the carboxy-terminal peptide (CTP) constituted of the last 29 amino-acids of the beta-subunit (beta 121–149) (42). With a carbohydrate content higher than 40% (43) and N-glycan chains terminated by sialic acids, eCG is the most heavily glycosylated glycoprotein hormone and has a longer in vivo half-life than other gonadotropins (~60 h) (44, 45). eCG binds to LH receptors in equine, but exerts FSH and LH actions in non-equine species by stimulating FSH and LH receptors respectively (4651). Its dual FSH/LH activity and its longer half-life were the reasons why eCG was widely used since decades to induce ovulation in breeding animals, especially in goats and ewes for out-of-season breeding to allow artificial insemination.

hCG is mainly produced during pregnancy, by syncytiotrophoblast cells and a hyperglycosylated isoform is produced by cytotrophoblast cells. These two isoforms are implicated in implantation and early embryo development. Regular hCG for example promotes progesterone secretion by corpus lutea, angiogenesis of uterine vasculature, or growth, and differentiation of fetal organs, whereas hyperglycosylated hCG stimulates implantation by invasion of cytotrophoblast cells or stimulates growth of placenta (52). However, hCG can also be produced in non-pregnant women: it is produced at low levels by gonadotrope cells of the anterior pituitary, and seems to have an LH-like activity during menstrual cycle. Free beta-subunit of hCG is produced by multiple non-trophoblastic cancers. It is elevated in most cancers such as bladder, renal, prostate, gastrointestinal, lung, breast, neuroendocrine, gynecological, head and neck, and hematological cancers (53) and promotes their growth and malignancy by blocking apoptosis in cancer cells (52). hCG beta-subunit is thus used as a tumor biomarker usually associated with poor prognosis (53).

hCG, unlike eCG, has an LH like activity only and does not bind FSH-R (1, 54). CG and LH exert their effects via the same receptor, LH/CG receptor (LHCGR), that is coupled to Gs and Gq in granulosa cells and theca cells (55). However, the receptor is able to differentiate the binding and activity of these two hormones (56), and hLH and hCG differentially regulate signaling pathways (57, 58).

FSH plays an important role in reproduction. In females, it is implicated in follicular growth. FSH-R is expressed in granulosa cells and it is mainly coupled to Gs, which activates adenylyl cyclase and induces the secretion of cyclic adenosine monophosphate (cAMP), but it is also known to be coupled to Gq. In males, FSH regulates spermatogenesis. FSH-R is expressed in Sertoli cells and signals via Gs and Gi (59). In HEK 293 cells expressing FSH-R, FSH stimulates Gs/cAMP/PKA signaling pathway as in granulosa or Sertoli cells, but also signals via a beta-arrestin-dependent pathway, leading in both case to extracellular signal-regulated kinases (ERK) 1/2 phosphorylation but with different kinetics (60).

FSH-R is also expressed in other tissues than reproductive organs, such as osteoclasts (61) or adipose tissue (62), suggesting that FSH may play other physiological roles. In adipose tissue, FSH-R is coupled to Gi. An increase in Ca2+ influx induces the phosphorylation of cAMP-response-element-binding protein, which in turn activates an array of genes involved in fatty acids and triglycerides biosynthesis (63). FSH is thus implicated in lipid biosynthesis and its storage in adipocytes, which may contribute to age-related obesity and diseases due to high FSH levels in aging populations (62, 63). The first paper mentioning the role of FSH in bone mass regulation was published in 2006 (61). The authors proposed a mechanism where FSH was able to increase osteoclasts formation and function via a Gi2a-coupled FSH-R expressed in these cells and their precursors (64), suggesting that high circulating FSH levels were responsible for post-menopausal osteoporosis.

Antibodies Modulating the Activity of Chorionic Gonadotropins

Antibodies Modulating the Activity of eCG

Most of the antibodies against eCG were developed for structural analysis purposes. Maurel et al. identified an antibody able to inhibit eCG binding to LH and FSH receptors (65). Chopineau et al. analyzed the affinity and the specificity of 14 mAbs directed against eCG (66). The aim of this study was to analyze the epitopic sites of eCG and permitted to draw a topographic map of antigenic and functional sites of this hormone. The affinity of antibodies for eCG was ranging between 10−7 and 10−11 M. Ten of them were alpha-subunit specific because they recognized both native eCG and free alpha-subunit, but not free beta-subunit. One antibody exhibited a higher affinity for alpha-subunit than for the native eCG, and 13 mAbs exhibited a better affinity for the dimer than for the free subunits. The effect of these mAbs was then tested on FSH and LH bioactivities of eCG in in vitro bioassays. One beta-subunit specific, one alpha-subunit specific and one native alpha/beta dimer specific antibodies did not show any effect on FSH and LH bioactivities. Nine alpha-subunit specific antibodies either weakly or strongly inhibited both bioactivities. Finally, two mAbs were potentiating FSH bioactivity of eCG: one was beta-subunit specific and the other was native dimeric eCG specific. They did not inhibit eCG binding to LH-R. The degree of inhibition of inhibitory antibodies was correlated with their affinity for eCG, but it wasn't for the two antibodies potentiating FSH bioactivity of eCG. These data suggest that the inhibitory or potentiating effects of mAbs on eCG bioactivities neither depend on their specificity nor their affinity. Moreover, the two antibodies potentiating the FSH bioactivity of eCG were either not affecting or inhibiting weakly the LH bioactivity of eCG, demonstrating that the effect on both bioactivities can be opposite (inhibitory or potentiating), with different degrees of activity (none or weak) on hormone bioactivities, highlighting the potential multiple mode of action of antibodies.

The high carbohydrate content makes also eCG highly immunogenic. Repeated injections of eCG for ovulation induction decrease the fertility of goats from 60 to 40% (12, 67). Roy et al. detected an immune response in animals treated with eCG for ovulation induction (68, 69), and demonstrated that the secreted antibodies from a previous treatment were inhibiting the action of eCG injected for the following treatment. The goats with high antibody levels at the time of eCG administration did have a much lower kidding rate (41%) than the other females (66%). This immune response was thus altering the fertility of these animals by delaying both the onset of estrus and the preovulatory LH surge. However, a deeper analysis revealed that some of the goats secreting high levels of anti-eCG antibodies did have a fertility beyond expected, ovulating and getting pregnant after each treatment, even after four treatments. The antibodies from the plasma of hypo-fertile or hyper-fertile goats secreting high levels of antibodies were purified and the IgG fractions were analyzed for their effect on FSH bioactivity in Y1 cell line derived from a mouse adrenal cortex tumor stably expressing human FSH-R, and for their effect on LH bioactivity in rat Leydig cells. The plasmas and the corresponding IgG fractions from different eCG treated goats exhibited either inhibitory, enhancing or no effects on FSH activity of eCG by modulating progesterone secretion by Y1 cells, and on LH activity of eCG by modulating testosterone secretion in Leydig cells (70). As expected, antibodies were mainly recognizing carbohydrate chains of eCG. Twenty-one either inhibitory, potentiating or neutral antibodies for LH and/or FSH bioactivities of eCG were analyzed. None of the antibodies cross-reacted with totally deglycosylated eCG or alpha-subunit of eCG. Interestingly, the inhibitory or stimulatory effects of these antibodies were not correlated with the affinity of the tested antibody for eCG (70). All together, these data demonstrated that gonadotropins' in vivo bioactivity and animals' fertility can be modulated with antibodies, especially since these antibodies are naturally occurring.

Later, Wehbi et al. (71) analyzed the effect of eCG in complex with three of these antibodies on FSH receptor signaling pathways. The eCG/antibody complexes effect was tested on HEK 293 cells expressing mouse FSH receptor and on granulosa cells punctured from slaughterhouse cows for their effect on cAMP production. The tested antibodies were differently modulating cAMP production: two of them were potentiating and one was slightly inhibiting eCG effect. In contrast, all three antibodies were enhancing ERK1/2 phosphorylation in HEK 293 cells expressing mouse FSH-R. Deeper analysis revealed that the antibodies were potentiating eCG signaling preferentially via beta-arrestin pathway, via cAMP/PKA pathway or via both. An antibody complexed to eCG was thus able to change the full agonist effect of eCG into a biased agonist effect, modulating differentially the balance between the signaling pathways induced by this hormone. This paper was also the demonstration that these antibodies were achieving the same in vivo effect in goat (i.e., high prolificity) via different signaling pathways. That was the first report of biased agonism at FSH-R and the authors suggested that such antibodies could help optimize glycoprotein hormones' bioactivities and thus the development of new therapies.

At the very beginning of infertility treatments, eCG was also used to treat women. The first successful treatment with eCG was described in 1945. Although its use lasted more than 30 years, scientists realized very early that women treated with eCG extracts, like animals, did produce “antigonadotrophic substances” which neutralize hormone's effect over time and after repeated injections. The immune response induced by eCG and the arrival of less immunogenic pituitary gonadotropin extracts led to the market withdrawal of eCG [reviewed in (4)].

Antibodies Modulating the Activity of Human Chorionic Gonadotropin (hCG)

As for other gonadotropins, anti-hCG antibodies were essentially developed for epitope mapping and variant specific mAbs permitted the development of immunoassays, leading in fine to pregnancy tests (7274).

Naturally occurring endogenous antibodies were also reported: patients treated with exogenous gonadotropins can develop anti-hCG antibodies that impair fertility. They were detected in young men with hypogonadotropic hypogonadism treated with hCG (75, 76). These antibodies, detected in a 15 year-old patient following a secondary resistance during a third treatment to hCG, were low affinity but high binding capacity antibodies (76). A few years later, seven additional young men with hypogonadotropic hypogonadism, aged between 11 and 18 years, and resistant to classical hCG regimen were tested for the presence of anti-hCG antibodies. Four of them showed antibodies, but the neutralizing effect of hCG could be counter passed by increasing the doses of hCG used (75). The same kind of antibodies were described in women (77, 78). Immune response against hCG impairs fertility of women and induces pregnancy loss within the first trimester of pregnancy. To thwart this negative effect, Muller and collaborators described a treatment that was successful in three women positive for hCG antibodies. This treatment combined membrane plasmapheresis, prednisolone, and intravenous immunoglobulin therapy (78).

Anti-hCG auto-antibodies were also detected in sera of men and women that never received any injection of exogenous hormone (79). These antibodies were low affinity and did not interfere with hormone activity. However, few years later, antibodies with high affinity and the capacity to neutralize hCG and LH activities were detected in a patient with a history of spontaneous abortion, that was never exposed to exogenous hormone therapy (80).

Antibodies Modulating the Activity of FSH or LH

FSH and/or LH Neutralization With Antibodies

Several inhibiting antibodies were described for FSH and most of them were used to better understand its physiological functions in vivo. Antibodies permit to block reversibly the action of one or several hormones, at a precise time of the lifespan of the studied model, rather than suppressing a whole organ like in hypophysectomy or a gene like in transgenic animals. For example, for female studies, in 1969, Goldman and Mahesh used an anti-sera obtained by rabbit immunization with ovine LH, that neutralized FSH as well as LH, to study the role of these hormones in ovulation (81). In 1970, the same group published data on the effect of the same anti-sera on neonatal rats (82). In 1971, Eshkol and Lunenfeld used the strategy of neutralization with antibodies to demonstrate the crucial role of FSH and LH in ovarian development during the first 2 weeks of life in rodents. FSH was found to be responsible for the stimulation of granulosa cell proliferation, organization and structure. FSH plus LH initiated secretory activity of granulosa cells, increased intrafollicular spaces, antrum formation, enrichment and maintenance of the theca layer, and development of the vascular system (19). At the same time, it was shown that LH anti-sera could block ovulation in rat, but not FSH anti-sera (8386). Schwartz et al. suggested that FSH neutralization during estrus cycle could have a deleterious effect on follicles destined to grow and ovulate in following cycles (86). Several other studies have confirmed the role of LH as the indispensable trigger of ovulation, whereas FSH was required for the recruitment of antral follicles at the start of a new cycle in rat and hamster (8791). The neutralization of FSH or LH with antibodies also permitted to study the role of these hormones in the synthesis of estrogen (92), on ornithine decarboxylase activity (93) and on gonadotropin surge-inhibiting factor/attenuating factor bioactivity (20) in rat and/or hamster. In monkey, the antibody neutralization of FSH highlighted the importance of FSH during follicular growth and showed that the mature follicle becomes less sensitive to FSH about 48 h before ovulation (94).

For male studies, Wickings and Nieschlag actively immunized Macaca mulatta against ovine FSH and observed a spermatogenesis suppression over a period of 2 years, confirming the importance of FSH for spermatogenesis (21). The importance of FSH in spermatogenesis was further confirmed by active immunization of Macaca radiata (95). In rat, even if the first studies obtained with immunoneutralization of FSH were controversial on the role of FSH in spermatogenesis (96, 97), later works with either passive (98) or active (22) immunization against FSH confirmed its crucial role in the maintenance of spermatogenesis. For active immunization, peptides from region 19–36 of rat FSH beta-subunit were used (22). Altogether, these results suggest that immunoneutralization of FSH could be used as a contraception in men by suppressing spermatogenesis. However, Nieschlag recommended to abandon the approach of immunization as a contraception because a complete suppression of spermatogenesis was not achieved even after 4.5 years of immunization (99). Nevertheless, Moudgal and collaborators carried out a pilot study in 1997 where five male volunteers were immunized with ovine FSH isolated from sheep pituitaries (100). The subjects only responded to the first two immunizations (day 1 and 20), and did not respond to the boosters given at day 40 and 70. Ovine FSH vaccination generated antibodies against human FSH, but only 25–45% of the antibodies generated against ovine FSH were able to bind human FSH and the sperm count reduction was around 30–64%, which is not enough to consider this method as a contraception.

Anti-FSH antibodies were also detected in women. Two types of antibodies have been identified: naturally occurring anti-FSH antibodies (101, 102) and anti-FSH antibodies resulting from exogenous gonadotropins (103106). First, Haller et al. found naturally occurring anti-FSH antibodies in patients with endometriosis or polycystic ovary syndrome (PCOS) and none of these patients had undergone ovarian stimulation for IVF. They also detected anti-FSH antibodies in healthy non-pregnant women but at lower rates than for patients with endometriosis or PCOS (101). Likewise, Shatavi et al. found spontaneous anti-gonadotropin antibodies in 27% of patients with unexplained infertility and never treated with gonadotropins, but only in 8% of women in the general population (102). To explain the presence of such spontaneous antibodies, it was supposed that an alteration of the immune system might be necessary and that the antigen responsible for their production could be either the circulating FSH from the female organism or the FSH in seminal fluid that may upregulate the anti-FSH immune response in females (101). In in vitro fertilization (IVF) patients, Haller et al. demonstrated that anti-FSH antibodies increase in infertile women with common autoantibodies (against nuclear antigens, smooth muscle, gastric parietal cells, b2-glycoprotein I, cardiolipin, and thyroid peroxidase) and with a history of IVF stimulation (103, 104). Shatavi et al. also found that anti-FSH antibodies were more recurrent in infertile patients with history of gonadotropin treatment than in infertile patients never treated with exogenous FSH or in women in the general population (102). Anti-FSH antibodies could also be associated with anti-ovarian antibodies (AOA) in patients with history of gonadotropin treatment (102, 107). The association of anti-FSH antibodies and AOA was also detected in infertile patients never treated by FSH and in women in the general population but at a lower frequency (102). Other studies have investigated the consequences of the presence of anti-FSH antibodies on the results of controlled ovarian stimulation (COS). Some studies found that anti-FSH antibodies were associated with poor ovarian response to IVF stimulation (104, 105). Thus, anti-FSH antibodies might have an inhibitory effect on FSH by preventing the binding of the hormone to its receptor or by trapping FSH in immune complexes (101, 107). On the contrary, Reznik et al. identified a higher proportion of anti-FSH antibodies in patients with a good response to COS compared to patients with a poor response (106). Antibodies produced in patients with a good response might have either no effect or a potentiating effect on the action of FSH. However, in humans, no in vitro study of the inhibitory or potentiating action of anti-FSH antibodies on FSH receptor signaling has been published yet. In human FSH, one of the major epitopes seems to be the 78–93 amino acid sequence of the β-chain (101, 107). This region contains a loop called cysteine noose which plays a role in the specificity of FSH receptor binding (101). Therefore, it was supposed that the binding of the hormone to its receptor could be modulated by antibodies directed against this region (101). To explain why some infertile patients develop anti-gonadotropin antibodies, some studies focused on the Major Histocompatibility Complex (MHC) Class II (103). The role of the MHC Class II is to present exogenous proteins to immune cells, which leads to a humoral immune response. In IVF patients, only anti-FSH IgA were associated with HLA-DQB1*03 (103). However, the development of anti-FSH antibody response to exogenous FSH treatment remains controversial. Indeed, a recent study conducted in healthy oocyte donors and infertile women has concluded that repeated gonadotropin treatments for IVF do not induce an immune response to FSH (108).

Antibodies Potentiating the Activity of FSH

The first anti-FSH mAbs described were directed against human FSH (16). Their binding specificities were well-characterized, but their effect on FSH activity was not investigated. The second anti-FSH mAb described in the literature was directed against bovine FSH (29) and was beta-subunit specific. It did not cross react with ovine or porcine FSH, indicating that it recognizes an area of bovine FSH not homologous to ovine or porcine hormones (29). Glencross et al. tested this antibody later on for its effect on bovine FSH bioactivity in hypopituitary Snell dwarf mice (109). The mAb injected in complex with FSH increased uterine weight whereas FSH alone or the mAb alone at the same concentrations did not have any effect, showing for the first time a potentiating effect with an anti-FSH mAb.

Holder's group described anti-sera directed against peptides derived from the beta-subunit of bovine FSH (110). When injected to hypopituitary Snell dwarf mice concomitantly with ovine FSH, these anti-sera produced by sheep immunized with peptides corresponding to 31–45 and 38–49 amino-acid regions of bovine FSH beta-subunit were able to enhance FSH activity, as measured by an increase in uterine and ovarian weight, and an increase in the percentage of keratinized cells in vaginal smears. The authors hypothesized that the administration of anti-peptide anti-sera precomplexed with FSH or active immunization of breeding animals with these peptides should result in a superovulatory response, and that the potentiating anti-sera strategy, in the case of FSH, could be used for several clinical situations, such as treatment of ovarian disorders related to low FSH secretion, induction of estrus and treatment leading to increase spermatogenesis.

Anti-Gonadotropin Antibodies as Therapeutic Agents

Therapeutic Antibodies in General

MAbs, initially developed for scientific purposes, now take part of the human therapeutic arsenal. Over the last three decades, they have grown to become more than 55% of the overall biotherapeutic market of the drug industry sales (111).

The regulatory story of therapeutic mAbs started in 1986 with the first FDA-approved therapeutic mAb, the murine mAb Orthoclone OKT3® (Muromonab CD3) indicated for the prevention of kidney transplant rejection. Unfortunately, the development of murine mAbs has been hindered because of the risk of immunogenicity in humans due to their murine elements. Replacing the constant region of murine mAb by human sequences resulted in the generation of the chimeric antibodies (~30% murine content). The first-approved one, Rituxan® (Rituximab) in 1997, was used for the treatment of low grade B cell lymphoma. To overcome immunogenicity risk even further, new technologies for the generation of predominately or entirely human origin mAbs were developed. The humanized mAbs (5–10% murine content) are tailored by replacing all sequences by human except antigen binding complementary determining regions, which were derived from the mouse. The humanization technology developed by Sir Winter lead to the first humanized therapeutic antibody CAMPATH-1H® (Alemtuzumab, approved in 2001). Sir Winter was awarded the 2018 Nobel Prize in Chemistry along with George Smith for this technology and the fully humanization using phage display. Fully human mAbs (last generation) were developed by replacing the whole of the rodent sequences by human sequences. Humira® (Adalimumab) is the first fully human antibody approved in 2004, for the treatment of rheumatoid arthritis. Thanks to these new technologies, the rate of approval and mAbs available on the market for the treatment of various diseases has increased dramatically. In 2017, the FDA and the European Medicines Agency broke the record and approved 10 new therapeutic antibodies (112). Currently, more than 70 mAb products are available on the market, most of them being humanized (32%) or fully human (54%) (113).

To date, approved mAbs are from different isotypes, but the preferred mAbs in clinical use are of the IgG1 isotype (80%) (114). Additionally, there are five monovalent antibody-fragments on the market, four antigen-binding fragments (Fab) and 1 single chain variable fragments (scFv) (114). More sophisticated forms have been engineered, such as Fc-modification, IgG2/IgG4 hybrid Fc, glyco-engineered mAbs, bispecifics, or antibody-drug conjugates. These types of sophisticated mAbs reach more and more clinical trial studies (114, 115). According to Zhou and Mark (116), common mechanism of action proposed for mAb drugs include: (i) disruption of ligand–receptor interaction; (ii) target cell elimination via antibody-dependent cellular cytotoxicity (ADCC), complement-dependent cytotoxicity (CDC), and antibody-dependent cellular phagocytosis (ADCP); (iii) engagement of cytotoxic T cell by bispecific Abs; (iv) receptor downregulation by enhanced internalization and degradation; and (v) targeted drug delivery.

The indication of treatment for ~80% of the therapeutic mAb drugs could be classified into oncology and immune diseases. The last ~20% are used for the treatment of infection and cardiovascular diseases, orthopedic, eye and rare diseases (113). Despite the high treatment cost, the success of therapeutic mAb has recently reached the veterinary health with the launch in the European Union, in 2017, of Cytopoint® (Lokivetmab), a treatment for atopic dermatitis in dogs. Notwithstanding all the therapeutic mAbs developed to date, so far, none of them have succeeded in the field of fertility, and none of them are potentiating antibodies.

Therapeutics Involving Anti-gonadotropin Antibodies

Neutra-PMSG®, an Antibody Against eCG/PMSG

The unique antibody commercialized until now in the field of animal fertility is Neutra-PMSG®. This anti-eCG mAb is alpha-subunit specific and inhibits both LH bioactivity on small bovine luteal cells and FSH bioactivity on granulosa cells of bovine follicles (66). It was developed and marketed for cattle to limit the adverse effects of PMSG to improve the embryo production after superovulation treatment (117, 118). Due to a long half-life, PMSG had the disadvantage to cause a prolonged stimulation of follicular growth after preovulatory LH peak, inducing a poor response to superovulation treatment for cattle (117, 119, 120), sheep (121), and goat (122). Neutra-PMSG® injected 1–2 days after PMSG in superovulation treatment neutralized the adverse effects of PMSG by reducing its half-life in systemic circulation, improving embryo production. This mAb did not recognize endogenous gonadotropins in treated animals. Therefore, Neutra-PMSG® was highly specific for PMSG (117). Currently, most cows are superovulated using pituitary extracts containing FSH and LH (13), mainly because the pharmaceutical company (Intervet, The Netherlands) that developed the Neutra-PMSG® mAb stopped production in the 1990s.

Active Immunization Against hCG

A vaccination against hCG was also considered in the 1970s as a contraception method to avoid pregnancy in women (123, 124). The aim is to induce the secretion of antibodies that will bind hCG and block its activity, thus impeding pregnancy. Because an immunization with the whole dimeric hCG (alpha+beta-subunits) was raising antibodies not only against hCG but also against human LH (125), a special preparation of beta-hCG was made by processing it against heterologous anti-LH immunosorbents (124) and conjugating it to purified tetanus toxoid as an immunogenic carrier (123). This processed molecule was able to induce an immune response with antibodies specific to hCG. The antibodies produced were able to abrogate the binding of hCG to its receptor and its biological effects in in vivo bioassays. Moreover, the antibody titer declines over time, indicating that the vaccination is reversible (126). This vaccination system went through phase 1 clinical trials in several countries (India, Finland, Sweden, Chile and Brazil) under the International Committee on Contraception Research of Population Council. A slightly different preparation consisting of a dimer of hCG beta-subunit non-covalently associated with ovine LH alpha-subunit conjugated to tetanus and diphtheria toxoids (127) underwent phase 1 and phase 2 clinical trials in several centers in India. Eighty percent of treated patients generated sufficient antibody titer (>50 ng/ml) to be protected against pregnancy, and only one pregnancy was recorded over 1,124 cycles in fertile and sexually active women with an antibody titer higher than 50 ng/ml. After 12 years of inactivity on this project, Talwar and his collaborators are now working on a genetically engineered recombinant vaccine that is expected to go through clinical development in the next few years (128).

A similar approach was also tested for colorectal and pancreatic cancer treatments. In 2000, AVI BioPharma collaborated with SuperGen for the clinical development and marketing of Avicine, a synthetic vaccine constituted of the C-terminal peptide of hCG (CTP-37) conjugated to diphtheria toxoid. The vaccine went through several clinical trials until phase 3 pivotal study. In phase 2 efficacy study for colorectal cancer, 73% of treated patients developed an immune response against hCG and this response was associated with an improved median survival time (129, 130). As far as we know, the product has not reached the market yet. Recently, another group proposed another vaccine, where one residue in the amino-acid sequence of hCG beta-subunit is substituted (hCGβR68E) to eliminate cross reactivity with LH and conjugated to heat shock protein (Hsp70) as carrier to increase its immunogenicity (131). This vaccine has not been tested in humans yet.

Other Potential Therapeutic Antibodies

Antibodies Targeting FSH

To consolidate the assumption that high circulating FSH levels were responsible for post-menopausal osteoporosis, Zhu et al. showed that blocking FSH action attenuates bone loss in ovariectomized mice via two mechanisms: by inhibiting bone resorption and by stimulating bone formation (132). To do so, they used a mouse polyclonal antibody targeting a 13-amino acid sequence (LVYKDPARPNTQK) of mouse FSH beta-subunit that is a receptor-binding domain.

Liu et al. hypothesized that a pharmacological blockade of FSH action could also reduce body fat mass. In fact, they have shown that the same polyclonal antibody targeting the receptor-binding domain of FSH beta-subunit, injected daily for 8 weeks in mice, prevented the gain of body fat induced by the diet in male and female mice (133). This decrease of body fat was associated with an increase of fat thermogenesis (133, 134). For the purpose of potential therapeutic application in human, this team has developed a mAb targeting the same epitope in human FSH beta-subunit (LVYKDPARPKIQK) that had the same effects on body fat and thermogenesis on the mouse as the mouse polyclonal antibody directed against a sequence that is 2 amino-acids different (LVYKDPARPNTQK) (135, 136). The modulation of FSH activity by anti-FSH antibodies may be considered as therapeutic means to reduce the risk of obesity in elderly people with high levels of FSH (133). It was thus proposed that the same antibody could both inhibit bone loss and body fat gain (133, 136). However, while some of the other studies published on the subject supported the role of FSH in bone mass regulation (137141), some others were contradictory (142, 143). Moreover, clinical studies on human subjects reported that FSH suppression with GnRH agonist had no effect on bone resorption in women (144). In men, this suppression either increased bone loss (145), or had no effect when a testosterone supplementation was given (146). Furthermore, body fat mass is also increased in men treated with GnRH analog (147). Altogether, these data suggest that further investigations are needed to better understand the mechanisms underlying the role of FSH in bone mass and body fat regulation before a therapeutic approach can be envisaged (148). A therapeutic antibody to prevent osteoporosis is on the market since 2010. Receptor activator of nuclear factor-kB (RANK) ligand (RANKL) is a cytokine necessary for the development and the activity of osteoclasts. A fully human antibody, denosumab (Prolia®, Amgen) prevents RANKL binding to its receptor RANK. Denosumab, when given subcutaneously twice yearly for 36 months, reduces the risk of vertebral, non-vertebral, and hip fractures in women with osteoporosis (149), demonstrating that the strategy of therapeutic antibody can be used in this indication.

Antibodies Targeting hCG

Antibodies able to inhibit hCG activity were first described in 1980 (150). They were specific of the beta-subunit of hCG and did not cross react at all with other gonadotropins. Recently, one of these antibodies (mAb PIPP) was expressed recombinantly in tobacco leaves in different formats (scFv, diabody and entire antibody) and tested, after their extraction and purification, for their efficacy to neutralize hCG. The three formats of the same antibody were able to inhibit in vitro testosterone production induced by hCG in Leydig cells. In vivo, the entire mAb was able to block uterine weight gain in mouse model (151). These antibodies were envisaged as a contraception method by passive immunization in women, and were considered as a better method than active immunization where the response may be variable between patients, and a sufficient titer determined as the protective level of antibody was observed in 80% of the patients only. These anti-hCG antibodies have not entered a clinical development so far.

The same antibody was used for the development of an immunotoxin targeting hCG-expressing cancer cells (152). The VH and VL domains of the full antibody were linked together to form the scFv fragment (scFv PIPP). This scFv's gene was then fused with a gene expressing Pseudomonas exotoxin (PE38) and expressed in Escherichia coli as a recombinant protein (scPiPP-PE38). Once purified and tested on cancer cells, the immunotoxin showed 90% killing of hCG beta expressing histiocytic lymphoma, T-lymphoblastic leukemia, and lung carcinoma cells in vitro. However, further studies are needed to evaluate the potential of scPiPP-PE38 as a therapeutic agent for management of cancer cells expressing hCG or its subunits.

Conclusion

Many different antibodies against gonadotropins were developed and have proven to be very useful tools for many applications. They can also be naturally secreted due to a humoral immune response to endogenous or exogenous gonadotropins. With the same structure, immunoglobulins can have inhibitory or potentiating effects depending on their paratope defined by CDRs and their epitope (binding site) on the antigen. In the case of eCG, its naturally occurring potentiating antibodies have demonstrated that a differential activation of signaling pathways of FSH-R could lead to the same in vivo effect, i.e., high prolificity in goats (71). The development of antibodies with a range of modulating effects on the potency and the efficacy of FSH on its signaling pathways could help deciphering the importance of each pathway for FSH roles in reproduction, bone mass and body fat regulation. Moreover, these antibodies can represent potential therapeutics, targeting one pathophysiological or physiological condition in particular. Several applications for anti-gonadotropin antibodies have already been proposed and are under exploration, like osteoporosis, obesity, contraception, or cancer. All of these indications require inhibition of gonadotropins' action. On the other hand, in small ruminants, the naturally occurring anti-eCG potentiating antibodies induced a better fertility and prolificity demonstrating that it is possible to improve fertility by potentiating gonadotropins' activity during several estrus cycles, without any side effects. All these studies demonstrated that it is possible to target each gonadotropin very specifically despite their similarities.

To conclude, the development of antibodies modulating gonadotropins' activity could not only provide new tools to better understand their roles in different physiological processes, but could also bring to the market innovative drugs. Taking into account the state of the art and the clinical development time, there is a long way to go until a therapeutic antibody targeting a gonadotropin can reach the market.

Author Contributions

All authors listed have made a substantial, direct and intellectual contribution to the work, and approved it for publication.

Conflict of Interest Statement

EK, LD, SC, and M-CM were employed by Igyxos SA.

The remaining author declares that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

1. Pierce JG, Parsons TF. Glycoprotein hormones: structure and function. Annu Rev Biochem. (1981) 50:465–95. doi: 10.1146/annurev.bi.50.070181.002341

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Simoni M, Gromoll J, Nieschlag E. The follicle-stimulating hormone receptor: biochemistry, molecular biology, physiology, and pathophysiology. Endocr Rev. (1997) 18:739–73. doi: 10.1210/er.18.6.739

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Huhtaniemi I, Zhang FP, Kero J, Hamalainen T, Poutanen M. Transgenic and knockout mouse models for the study of luteinizing hormone and luteinizing hormone receptor function. Mol Cell Endocrinol. (2002) 187:49–56. doi: 10.1016/S0303-7207(01)00698-0

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Lunenfeld B. Development of gonadotrophins for clinical use. Reprod Biomed Online (2002) 4 (Suppl. 1):11–7. doi: 10.1016/S1472-6483(12)60006-6

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Lunenfeld B. Gonadotropin stimulation: past, present and future. Reprod Med Biol. (2012) 11:11–25. doi: 10.1007/s12522-011-0097-2

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Boehm U, Bouloux PM, Dattani MT, de Roux N, Dode C, Dunkel L, et al. Expert consensus document: European Consensus Statement on congenital hypogonadotropic hypogonadism–pathogenesis, diagnosis and treatment. Nat Rev Endocrinol. (2015) 11:547–64. doi: 10.1038/nrendo.2015.112

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Dwyer AA, Raivio T, Pitteloud N. Gonadotrophin replacement for induction of fertility in hypogonadal men. Best Pract Res Clin Endocrinol Metab. (2015) 29:91–103. doi: 10.1016/j.beem.2014.10.005

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Han TS, Bouloux PM. What is the optimal therapy for young males with hypogonadotropic hypogonadism? Clin Endocrinol (Oxf). (2010) 72:731–7. doi: 10.1111/j.1365-2265.2009.03746.x

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Zitzmann M, Nieschlag E. Hormone substitution in male hypogonadism. Mol Cell Endocrinol. (2000) 161:73–88. doi: 10.1016/S0303-7207(99)00227-0

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Koechling W, Plaksin D, Croston GE, Jeppesen JV, Macklon KT, Andersen CY. Comparative pharmacology of a new recombinant FSH expressed by a human cell line. Endocr Connect. (2017) 6:297–305. doi: 10.1530/EC-17-0067

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Abd-Elaziz K, Duijkers I, Stockl L, Dietrich B, Klipping C, Eckert K, 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) 32:1639–47. doi: 10.1093/humrep/dex220

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Baril G, Remy B, Vallet JC, Beckers JF. Effect of repeated use of progestagen-PMSG treatment for estrus control in dairy goats out of breeding season. Reprod Dom Anim. (1992) 27:161–8. doi: 10.1111/j.1439-0531.1992.tb01135.x

CrossRef Full Text | Google Scholar

13. Bo GA, Mapletoft RJ. Historical perspectives and recent research on superovulation in cattle. Theriogenology (2014) 81:38–48. doi: 10.1016/j.theriogenology.2013.09.020

PubMed Abstract | CrossRef Full Text | Google Scholar

14. De Rensis F, Kirkwood RN. Control of estrus and ovulation: Fertility to timed insemination of gilts and sows. Theriogenology (2016) 86:1460–6. doi: 10.1016/j.theriogenology.2016.04.089

CrossRef Full Text | Google Scholar

15. Sunderam S, Kissin DM, Crawford SB, Folger SG, Boulet SL, Warner L, et al. Assisted reproductive technology surveillance - United States, 2015. MMWR Surveill Summ. (2018) 67:1–28. doi: 10.15585/mmwr.ss6703a1

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Hojo H, Ryan RJ. Monoclonal antibodies against human follicle-stimulating hormone. Endocrinology (1985) 117:2428–34. doi: 10.1210/endo-117-6-2428

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Lunenfeld B, Isersky C, Shelesnyakmc. Immunologic studies on gonadotropins. I. Immunogenic properties and immunologic characterization of human chorionic gonadotropin preparations (HCG) and their homologous antisera. J Clin Endocrinol Metab. (1962) 22:555–63. doi: 10.1210/jcem-22-6-555

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Weiner RS, Dias JA, Andersen TT. Epitope mapping of human follicle stimulating hormone-alpha using monoclonal antibody 3A identifies a potential receptor binding sequence. Endocrinology (1991) 128:1485–95. doi: 10.1210/endo-128-3-1485

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Eshkol A, Lunenfeld B. Biological effects of antibodies to gonadotropins. Gynecol Invest. (1971) 2:23–56. doi: 10.1159/000301850

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Tio S, van Dieten JA, de Koning J. Immunoneutralization of follicle stimulating hormone does not affect gonadotrophin surge-inhibiting factor/attenuating factor bioactivity during the rat ovarian cycle. Hum Reprod. (1998) 13:2731–7. doi: 10.1093/humrep/13.10.2731

CrossRef Full Text | Google Scholar

21. Wickings EJ, Nieschlag E. Suppression of spermatogenesis over two years in rhesus monkeys actively immunized with follicle-stimulating hormone. Fertil Steril. (1980) 34:269–74. doi: 10.1016/S0015-0282(16)44961-7

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Yao B, Yi N, Zhou S, OuYang W, Xu H, Ge Y, et al. The effect of induced anti-follicle-stimulating hormone autoantibody on serum hormone level and apoptosis in rat testis. Life Sci. (2012) 91:83–8. doi: 10.1016/j.lfs.2012.04.026

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Aono T, Taymor ML. Radioimmunoassay for follicle-stimulating hormone (FSH) with 125-I-labeled FSH. Am J Obstet Gynecol. (1968) 100:110–7. doi: 10.1016/S0002-9378(15)33647-4

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Check JH, Nazari A, Kuhn R, Lauer C. Relationship of early follicular phase sera follicle stimulating hormone and luteinizing hormone levels as measured by a radioimmunoassay and an enzyme-linked immunosorbent assay to number of oocytes retrieved. Clin Exp Obstet Gynecol. (1996) 23:83–6.

PubMed Abstract | Google Scholar

25. L'Hermite M, Niswender GD, Reichert LE Jr, Midgley AR Jr. Serum follicle-stimulating hormone in sheep as measured by radioimmunoassay. Biol Reprod. (1972) 6:325–32. doi: 10.1093/biolreprod/6.2.325

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Midgley AR. Radioimmunoassay for human follicle-stimulating hormone. J Clin Endocrinol Metab. (1967) 27:295–9. doi: 10.1210/jcem-27-2-295

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Odell WD, Parlow AF, Cargille CM, Ross GT. Radioimmunoassay for human follicle-stimulating hormone: physiological studies. J Clin Invest. (1968) 47:2551–62. doi: 10.1172/JCI105937

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Donini P, Puzzuoli D, D'Alessio I, Lunenfeld B, Eshkol A, Parlow AF. Purification and separation of follicle stimulating hormone (FSH) and luteinizing hormone (LH) from human postmenopausal gonadotrophin (HMG). II. Preparation of biological apparently pure FSH by selective binding of the LH with an anti-HGG serum and subsequent chromatography. Acta Endocrinol (Copenh). (1966) 52:186–98. doi: 10.1530/acta.0.0520186

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Miller KF, Goldsby RA, Bolt DJ. Immunoaffinity chromatography of bovine FSH using monoclonal antibodies. J Endocrinol. (1987) 115:283–8. doi: 10.1677/joe.0.1150283

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Zandian M, Jungbauer A. An immunoaffinity column with a monoclonal antibody as ligand for human follicle stimulating hormone. J Sep Sci. (2009) 32:1585–91. doi: 10.1002/jssc.200900103

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Schechter Y, Hernaez L, Schlessinger J, Cuatrecasas P. Local aggregation of hormone-receptor complexes is required for activation by epidermal growth factor. Nature (1979) 278:835–8. doi: 10.1038/278835a0

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Shechter Y, Chang KJ, Jacobs S, Cuatrecasas P. Modulation of binding and bioactivity of insulin by anti-insulin antibody: relation to possible role of receptor self-aggregation in hormone action. Proc Natl Acad Sci USA. (1979) 76:2720–4. doi: 10.1073/pnas.76.6.2720

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Aston R, Holder AT, Ivanyi J, Bomford R. Enhancement of bovine growth hormone activity in vivo by monoclonal antibodies. Mol Immunol. (1987) 24:143–50. doi: 10.1016/0161-5890(87)90086-1

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Holder AT, Aston R, Preece MA, Ivanyi J. Monoclonal antibody-mediated enhancement of growth hormone activity in vivo. J Endocrinol. (1985) 107:R9–12. doi: 10.1677/joe.0.107R009

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Holder AT, Blows JA, Aston R, Bates PC. Monoclonal antibody enhancement of the effects of human growth hormone on growth and body composition in mice. J Endocrinol. (1988) 117:85–90. doi: 10.1677/joe.0.1170085

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Holder AT, Aston R, Rest JR, Hill DJ, Patel N, Ivanyi J. Monoclonal antibodies can enhance the biological activity of thyrotropin. Endocrinology (1987) 120:567–73. doi: 10.1210/endo-120-2-567

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Costagliola S, Bonomi M, Morgenthaler NG, Van Durme J, Panneels V, Refetoff S, et al. Delineation of the discontinuous-conformational epitope of a monoclonal antibody displaying full in vitro and in vivo thyrotropin activity. Mol Endocrinol. (2004) 18:3020–34. doi: 10.1210/me.2004-0231

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Costagliola S, Franssen JD, Bonomi M, Urizar E, Willnich M, Bergmann A, et al. Generation of a mouse monoclonal TSH receptor antibody with stimulating activity. Biochem Biophys Res Commun. (2002) 299:891–6. doi: 10.1016/S0006-291X(02)02762-6

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Allen WR, Moor RM. The origin of the equine endometrial cups. I. Production of PMSG by fetal trophoblast cells. J Reprod Fertil. (1972) 29:313–6. doi: 10.1530/jrf.0.0290313

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Papkoff H. Chemical and biological properties of the subunits of pregnant mare serum gonadotropin. Biochem Biophys Res Commun. (1974) 58:397–404. doi: 10.1016/0006-291X(74)90378-7

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Sherman GB, Wolfe MW, Farmerie TA, Clay CM, Threadgill DS, Sharp DC, et al. A single gene encodes the beta-subunits of equine luteinizing hormone and chorionic gonadotropin. Mol Endocrinol. (1992) 6:951–9.

PubMed Abstract | Google Scholar

42. Bousfield GR, Butnev VY, Butnev VY. Identification of twelve O-glycosylation sites in equine chorionic gonadotropin beta and equine luteinizing hormone ss by solid-phase Edman degradation. Biol Reprod. (2001) 64:136–47. doi: 10.1095/biolreprod64.1.136

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Christakos S, Bahl OP. Pregnant mare serum gonadotropin. Purification and physicochemical, biological, and immunological characterization. J Biol Chem. (1979) 254:4253–61.

PubMed Abstract | Google Scholar

44. Matzuk MM, Hsueh AJ, Lapolt P, Tsafriri A, Keene JL, Boime I. The biological role of the carboxyl-terminal extension of human chorionic gonadotropin [corrected] beta-subunit. Endocrinology (1990) 126:376–83. doi: 10.1210/endo-126-1-376

PubMed Abstract | CrossRef Full Text | Google Scholar

45. McIntosh JE, Moor RM, Allen WR. Pregnant mare serum gonadotrophin: rate of clearance from the circulation of sheep. J Reprod Fertil. (1975) 44:95–100. doi: 10.1530/jrf.0.0440095

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Combarnous Y, Guillou F, Martinat N. Comparison of in vitro follicle-stimulating hormone (FSH) activity of equine gonadotropins (luteinizing hormone, FSH, and chorionic gonadotropin) in male and female rats. Endocrinology (1984) 115:1821–7. doi: 10.1210/endo-115-5-1821

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Combarnous Y, Hennen G, Ketelslegers JM. Pregnant mare serum gonadotropin exhibits higher affinity for lutropin than for follitropin receptors of porcine testis. FEBS Lett. (1978) 90:65–8. doi: 10.1016/0014-5793(78)80299-3

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Guillou F, Combarnous Y. Purification of equine gonadotropins and comparative study of their acid-dissociation and receptor-binding specificity. Biochim Biophys Acta (1983) 755:229–36. doi: 10.1016/0304-4165(83)90208-8

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Licht P, Gallo AB, Aggarwal BB, Farmer SW, Castelino JB, Papkoff H. Biological and binding activities of equine pituitary gonadotrophins and pregnant mare serum gonadotrophin. J Endocrinol. (1979) 83:311–22. doi: 10.1677/joe.0.0830311

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Matsui T, Mizuochi T, Titani K, Okinaga T, Hoshi M, Bousfield GR, et al. Structural analysis of N-linked oligosaccharides of equine chorionic gonadotropin and lutropin beta-subunits. Biochemistry (1994) 33:14039–48. doi: 10.1021/bi00251a012

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Smith PL, Bousfield GR, Kumar S, Fiete D, Baenziger JU. Equine lutropin and chorionic gonadotropin bear oligosaccharides terminating with SO4-4-GalNAc and Sia alpha 2,3Gal, respectively. J Biol Chem. (1993) 268:795–802.

PubMed Abstract | Google Scholar

52. Cole LA. Biological functions of hCG and hCG-related molecules. Reprod Biol Endocrinol. (2010) 8:102. doi: 10.1186/1477-7827-8-102

PubMed Abstract | CrossRef Full Text | Google Scholar

53. Stenman UH, Alfthan H, Hotakainen K. Human chorionic gonadotropin in cancer. Clin Biochem. (2004) 37:549–61. doi: 10.1016/j.clinbiochem.2004.05.008

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Moyle WR, Campbell RK, Myers RV, Bernard MP, Han Y, Wang X. Co-evolution of ligand-receptor pairs. Nature (1994) 368:251–5. doi: 10.1038/368251a0

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Casarini L, Santi D, Brigante G, Simoni M. Two hormones for one receptor: evolution, biochemistry, actions, and pathophysiology of LH and hCG. Endocr Rev. (2018) 39:549–92. doi: 10.1210/er.2018-00065

PubMed Abstract | CrossRef Full Text | Google Scholar

56. Galet C, Ascoli M. The differential binding affinities of the luteinizing hormone (LH)/choriogonadotropin receptor for LH and choriogonadotropin are dictated by different extracellular domain residues. Mol Endocrinol. (2005) 19:1263–76. doi: 10.1210/me.2004-0410

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Casarini L, Lispi M, Longobardi S, Milosa F, La Marca A, Tagliasacchi D, et al. LH and hCG action on the same receptor results in quantitatively and qualitatively different intracellular signalling. PLoS ONE (2012) 7:e46682. doi: 10.1371/journal.pone.0046682

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Riccetti L, De Pascali F, Gilioli L, Poti F, Giva LB, Marino M, et al. Human LH and hCG stimulate differently the early signalling pathways but result in equal testosterone synthesis in mouse Leydig cells in vitro. Reprod Biol Endocrinol. (2017) 15:2. doi: 10.1186/s12958-016-0224-3

PubMed Abstract | CrossRef Full Text | Google Scholar

59. De Pascali F, Reiter E. beta-arrestins and biased signaling in gonadotropin receptors. Minerva Ginecol. (2018) 70:525–38. doi: 10.23736/S0026-4784.18.04272-7

CrossRef Full Text | Google Scholar

60. Kara E, Crepieux P, Gauthier C, Martinat N, Piketty V, Guillou F, et al. A phosphorylation cluster of five serine and threonine residues in the C-terminus of the follicle-stimulating hormone receptor is important for desensitization but not for beta-arrestin-mediated ERK activation. Mol Endocrinol. (2006) 20:3014–26. doi: 10.1210/me.2006-0098

CrossRef Full Text | Google Scholar

61. Sun L, Peng Y, Sharrow AC, Iqbal J, Zhang Z, Papachristou DJ, et al. FSH directly regulates bone mass. Cell (2006) 125:247–60. doi: 10.1016/j.cell.2006.01.051

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Cui H, Zhao G, Liu R, Zheng M, Chen J, Wen J. FSH stimulates lipid biosynthesis in chicken adipose tissue by upregulating the expression of its receptor FSHR. J Lipid Res. (2012) 53:909–17. doi: 10.1194/jlr.M025403

PubMed Abstract | CrossRef Full Text | Google Scholar

63. Liu XM, Chan HC, Ding GL, Cai J, Song Y, Wang TT, et al. FSH regulates fat accumulation and redistribution in aging through the Galphai/Ca(2+)/CREB pathway. Aging Cell (2015) 14:409–20. doi: 10.1111/acel.12331

PubMed Abstract | CrossRef Full Text | Google Scholar

64. Zhu LL, Tourkova I, Yuen T, Robinson LJ, Bian Z, Zaidi M, et al. Blocking FSH action attenuates osteoclastogenesis. Biochem Biophys Res Commun. (2012) 422:54–8. doi: 10.1016/j.bbrc.2012.04.104

PubMed Abstract | CrossRef Full Text | Google Scholar

65. Maurel MC, Ban E, Bidart JM, Combarnous Y. Immunochemical study of equine chorionic gonadotropin (eCG/PMSG): antigenic determinants on alpha- and beta-subunits. Biochim Biophys Acta (1992) 1159:74–80. doi: 10.1016/0167-4838(92)90077-Q

PubMed Abstract | CrossRef Full Text | Google Scholar

66. Chopineau M, Maurel MC, Combarnous Y, Durand P. Topography of equine chorionic gonadotropin epitopes relative to the luteinizing hormone and follicle-stimulating hormone receptor interaction sites. Mol Cell Endocrinol. (1993) 92:229–39. doi: 10.1016/0303-7207(93)90013-A

PubMed Abstract | CrossRef Full Text | Google Scholar

67. Baril G, Leboeuf B, Saumande J. Synchronization of estrus in goats: the relationship between time of occurrence of estrus and fertility following artificial insemination. Theriogenology (1993) 40:621–8. doi: 10.1016/0093-691X(93)90414-Z

PubMed Abstract | CrossRef Full Text | Google Scholar

68. Roy F, Combes B, Vaiman D, Cribiu EP, Pobel T, Deletang F, et al. Humoral immune response to equine chorionic gonadotropin in ewes: association with major histocompatibility complex and interference with subsequent fertility. Biol Reprod. (1999) 61:209–18. doi: 10.1095/biolreprod61.1.209

PubMed Abstract | CrossRef Full Text | Google Scholar

69. Roy F, Maurel MC, Combes B, Vaiman D, Cribiu EP, Lantier I, et al. The negative effect of repeated equine chorionic gonadotropin treatment on subsequent fertility in Alpine goats is due to a humoral immune response involving the major histocompatibility complex. Biol Reprod. (1999) 60:805–13. doi: 10.1095/biolreprod60.4.805

PubMed Abstract | CrossRef Full Text | Google Scholar

70. Herve V, Roy F, Bertin J, Guillou F, Maurel MC. Antiequine chorionic gonadotropin (eCG) antibodies generated in goats treated with eCG for the induction of ovulation modulate the luteinizing hormone and follicle-stimulating hormone bioactivities of eCG differently. Endocrinology (2004) 145:294–303. doi: 10.1210/en.2003-0595

PubMed Abstract | CrossRef Full Text | Google Scholar

71. Wehbi V, Decourtye J, Piketty V, Durand G, Reiter E, Maurel MC. Selective modulation of follicle-stimulating hormone signaling pathways with enhancing equine chorionic gonadotropin/antibody immune complexes. Endocrinology (2010) 151:2788–99. doi: 10.1210/en.2009-0892

PubMed Abstract | CrossRef Full Text | Google Scholar

72. Berger P, Bidart JM, Delves PS, Dirnhofer S, Hoermann R, Isaacs N, et al. Immunochemical mapping of gonadotropins. Mol Cell Endocrinol (1996) 125:33–43. doi: 10.1016/S0303-7207(96)03943-3

PubMed Abstract | CrossRef Full Text | Google Scholar

73. Berger P, Paus E, Hemken PM, Sturgeon C, Stewart WW, Skinner JP, et al. Candidate epitopes for measurement of hCG and related molecules: the second ISOBM TD-7 workshop. Tumour Biol. (2013) 34:4033–57. doi: 10.1007/s13277-013-0994-6

PubMed Abstract | CrossRef Full Text | Google Scholar

74. Bidart JM, Bellet D. Human chorionic gonadotropin Molecular forms, detection, and clinical implications. Trends Endocrinol Metab. (1993) 4:285–91. doi: 10.1016/1043-2760(93)90047-I

PubMed Abstract | CrossRef Full Text | Google Scholar

75. Claustrat B, David L, Faure A, Francois R. Development of anti-human chorionic gonadotropin antibodies in patients with hypogonadotropic hypogonadism. A study of four patients. J Clin Endocrinol Metab. (1983) 57:1041–7. doi: 10.1210/jcem-57-5-1041

PubMed Abstract | CrossRef Full Text | Google Scholar

76. Sokol RZ, McClure RD, Peterson M, Swerdloff RS. Gonadotropin therapy failure secondary to human chorionic gonadotropin-induced antibodies. J Clin Endocrinol Metab. (1981) 52:929–32. doi: 10.1210/jcem-52-5-929

PubMed Abstract | CrossRef Full Text | Google Scholar

77. Amato F, Warnes GM, Kirby CA, Norman RJ. Infertility caused by HCG autoantibody. J Clin Endocrinol Metab. (2002) 87:993–7. doi: 10.1210/jcem.87.3.8334

PubMed Abstract | CrossRef Full Text | Google Scholar

78. Muller V, Ob'edkova K, Krikheli I, Kogan I, Fedorova I, Lesik E, et al. Successful pregnancy outcome in women with recurrent IVF failure and Anti-hCG autoimmunity: a report of three cases. Case Rep Immunol. (2016) 2016:4391537. doi: 10.1155/2016/4391537

PubMed Abstract | CrossRef Full Text | Google Scholar

79. Wass M, McCann K, Bagshawe KD. Isolation of antibodies to HCG/LH from human sera. Nature (1978) 274:369–70. doi: 10.1038/274368a0

PubMed Abstract | CrossRef Full Text | Google Scholar

80. Pala A, Coghi I, Spampinato G, Di Gregorio R, Strom R, Carenza L. Immunochemical and biological characteristics of a human autoantibody to human chorionic gonadotropin and luteinizing hormone. J Clin Endocrinol Metab. (1988) 67:1317–21. doi: 10.1210/jcem-67-6-1317

PubMed Abstract | CrossRef Full Text | Google Scholar

81. Goldman BD, Mahesh VB. A possible role of acute FSH-release in ovulation in the hamster, as demonstrated by utilization of antibodies to LH and FSH. Endocrinology (1969) 84:236–43. doi: 10.1210/endo-84-2-236

PubMed Abstract | CrossRef Full Text | Google Scholar

82. Goldman BD, Mahesh VB. Induction of infertility in male rats by treatment with gonadotropin antiserum during neonatal life. Biol Reprod. (1970) 2:444–51. doi: 10.1095/biolreprod2.3.444

PubMed Abstract | CrossRef Full Text | Google Scholar

83. Ely CA, Schwartz NB. Elucidation of the role of the luteinizing hormone in estrogen secretion and ovulation by use of antigonadotropic sera. Endocrinology (1971) 89:1103–8. doi: 10.1210/endo-89-4-1103

PubMed Abstract | CrossRef Full Text | Google Scholar

84. Schwartz NB. The role of FSH and LH and of their antibodies on follicle growth and on ovulation. Biol Reprod. (1974) 10:236–72. doi: 10.1095/biolreprod10.2.236

PubMed Abstract | CrossRef Full Text | Google Scholar

85. Schwartz NB, Ely CA. Comparison of effects of hypophysectomy, antiserum to ovine LH, and ovariectomy on estrogen secretion during the rat estrous cycle. Endocrinology (1970) 86:1420–35. doi: 10.1210/endo-86-6-1420

PubMed Abstract | CrossRef Full Text | Google Scholar

86. Schwartz NB, Krone K, Talley WL, Ely CA. Administration of antiserum to ovine FSH in the female rat: failure to influence immediate events of cycle. Endocrinology (1973) 92:1165–74. doi: 10.1210/endo-92-4-1165

PubMed Abstract | CrossRef Full Text | Google Scholar

87. Rani CS, Moudgal NR. Examination of the role of FSH in periovulatory events in the hamster. J Reprod Fertil. (1977) 50:37–45. doi: 10.1530/jrf.0.0500037

PubMed Abstract | CrossRef Full Text | Google Scholar

88. Rao AJ, Moudgal NR, Raj HG, Lipner H, Greep RO. The role of FSH and LH in the initiation of ovulation in rats and hamsters: a study using rabbit antisera to ovine FSH and LH. J Reprod Fertil. (1974) 37:323–30. doi: 10.1530/jrf.0.0370323

PubMed Abstract | CrossRef Full Text | Google Scholar

89. Schwartz NB, Cobbs SB, Talley WL, Ely CA. Induction of ovulation by LH and FSH in the presence of antigonadotrophic sera. Endocrinology (1975) 96:1171–8. doi: 10.1210/endo-96-5-1171

PubMed Abstract | CrossRef Full Text | Google Scholar

90. Sheela Rani CS, Moudgal NR. Role of the proestrous surge of gonadotropins in the initiation of follicular maturation in the cyclic hamster: a study using antisera to follicle stimulating hormone and luteinizing hormone. Endocrinology (1977) 101:1484–94. doi: 10.1210/endo-101-5-1484

CrossRef Full Text | Google Scholar

91. Welschen R, Dullaart J. Administration of antiserum against ovine follicle-stimulating hormone or ovine luteinizing hormone at pro-poestrus in the rat: effects on follicular development during the oncoming cycle. J Endocrinol. (1976) 70:301–6. doi: 10.1677/joe.0.0700301

PubMed Abstract | CrossRef Full Text | Google Scholar

92. Rani CS, Moudgal NR. Examination of the role of follicle stimulating hormone in estrogen biosynthesis in vivo and in vitro in the ovary of the cyclic hamster. Steroids (1978) 32:435–51. doi: 10.1016/0039-128X(78)90057-0

PubMed Abstract | CrossRef Full Text | Google Scholar

93. Sheela Rani CS, Moudgal NR. Effect of follicle-stimulating hormone and its antiserum on the activity of ornithine decarboxylase in the ovary of rat and hamster. Endocrinology (1979) 104:1480–3. doi: 10.1210/endo-104-5-1480

CrossRef Full Text | Google Scholar

94. Ravindranath N, Sheela Rani CS, Martin F, Moudgal NR. Effect of FSH deprivation at specific times on follicular maturation in the bonnet monkey (Macaca radiata). J Reprod Fertil. (1989) 87:231–41. doi: 10.1530/jrf.0.0870231

PubMed Abstract | CrossRef Full Text | Google Scholar

95. Aravindan GR, Gopalakrishnan K, Ravindranath N, Moudgal NR. Effect of altering endogenous gonadotrophin concentrations on the kinetics of testicular germ cell turnover in the bonnet monkey (Macaca radiata). J Endocrinol. (1993) 137:485–95. doi: 10.1677/joe.0.1370485

PubMed Abstract | CrossRef Full Text | Google Scholar

96. Dym M, Raj HG, Lin YC, Chemes HE, Kotite NJ, Nayfeh SN, et al. Is FSH required for maintenance of spermatogenesis in adult rats? J Reprod Fertil Suppl. (1979) 26:175–81.

Google Scholar

97. Madhwa Raj HG, Dym M. The effects of selective withdrawal of FSH or LH on spermatogenesis in the immature rat. Biol Reprod. (1976) 14:489–94. doi: 10.1093/biolreprod/14.4.489

PubMed Abstract | CrossRef Full Text | Google Scholar

98. Shetty J, Marathe GK, Dighe RR. Specific immunoneutralization of FSH leads to apoptotic cell death of the pachytene spermatocytes and spermatogonial cells in the rat. Endocrinology (1996) 137:2179–82. doi: 10.1210/endo.137.5.8612566

PubMed Abstract | CrossRef Full Text | Google Scholar

99. Nieschlag E. Reasons for abandoning immunization against FSH as an approach to male fertility regulation. In: Zatuchni GI, Goldsmith A, Spieler JM, Sciarra JJ, editors. Male contraception: advances and future prospects. Philadelphia: Harper and Row (1986). p. 395–400.

Google Scholar

100. Moudgal NR, Murthy GS, Prasanna Kumar KM, Martin F, Suresh R, Medhamurthy R, et al. Responsiveness of human male volunteers to immunization with ovine follicle stimulating hormone vaccine: results of a pilot study. Hum Reprod. (1997) 12:457–63. doi: 10.1093/humrep/12.3.457

PubMed Abstract | CrossRef Full Text | Google Scholar

101. Haller K, Mathieu C, Rull K, Matt K, Bene MC, Uibo R. IgG, IgA and IgM antibodies against FSH: serological markers of pathogenic autoimmunity or of normal immunoregulation? Am J Reprod Immunol. (2005) 54:262–9. doi: 10.1111/j.1600-0897.2005.00306.x

CrossRef Full Text | Google Scholar

102. Shatavi SV, Llanes B, Luborsky JL. Association of unexplained infertility with gonadotropin and ovarian antibodies. Am J Reprod Immunol. (2006) 56:286–91. doi: 10.1111/j.1600-0897.2006.00428.x

PubMed Abstract | CrossRef Full Text | Google Scholar

103. Haller K, Salumets A, Grigorova M, Talja I, Salur L, Bene MC, et al. Putative predictors of antibodies against follicle-stimulating hormone in female infertility: a study based on in vitro fertilization patients. Am J Reprod Immunol. (2007) 57:193–200. doi: 10.1111/j.1600-0897.2006.00462.x

PubMed Abstract | CrossRef Full Text | Google Scholar

104. Haller K, Salumets A, Uibo R. Anti-FSH antibodies associate with poor outcome of ovarian stimulation in IVF. Reprod Biomed Online (2008) 16:350–5. doi: 10.1016/S1472-6483(10)60595-0

PubMed Abstract | CrossRef Full Text | Google Scholar

105. Meyer WR, Lavy G, DeCherney AH, Visintin I, Economy K, Luborsky JL. Evidence of gonadal and gonadotropin antibodies in women with a suboptimal ovarian response to exogenous gonadotropin. Obstet Gynecol. (1990) 75:795–9.

PubMed Abstract | Google Scholar

106. Reznik Y, Benhaim A, Morello R, Herlicoviez M, Ballet JJ, Mahoudeau J. High frequency of IgG antagonizing follicle-stimulating hormone-stimulated steroidogenesis in infertile women with a good response to exogenous gonadotropins. Fertil Steril. (1998) 69:46–52. doi: 10.1016/S0015-0282(97)00430-5

PubMed Abstract | CrossRef Full Text | Google Scholar

107. Gobert B, Jolivet-Reynaud C, Dalbon P, Barbarino-Monnier P, Faure GC, Jolivet M, et al. An immunoreactive peptide of the FSH involved in autoimmune infertility. Biochem Biophys Res Commun. (2001) 289:819–24. doi: 10.1006/bbrc.2001.6059

PubMed Abstract | CrossRef Full Text | Google Scholar

108. Morte C, Celma C, De Geyter C, Urbancsek J, Coroleu Lletget B, Cometti B. Assessment of the immunogenicity of gonadotrophins during controlled ovarian stimulation. Am J Reprod Immunol. (2017) 78:e12675. doi: 10.1111/aji.12675

PubMed Abstract | CrossRef Full Text | Google Scholar

109. Glencross RG, Lovell RD, Holder AT. Monoclonal antibody enhancement of FSH-induced uterine growth in snell dwarf mice. J Endocrinol. (1993) 136:R5–7. doi: 10.1677/joe.0.136R005

PubMed Abstract | CrossRef Full Text | Google Scholar

110. Ferasin L, Gabai G, Beattie J, Bono G, Holder AT. Enhancement of FSH bioactivity in vivo using site-specific antisera. J Endocrinol. (1997) 152:355–63. doi: 10.1677/joe.0.1520355

PubMed Abstract | CrossRef Full Text | Google Scholar

111. Levine HL, Cooney BR. Monoclonal antibodies – the development of therapeutic monoclonal antibody products: a comprehensive guide to CMC activities from clone to clinic. Drug Dev Deliv. (2018) 18:32–5.

Google Scholar

112. Kaplon H, Reichert JM. Antibodies to watch in 2018. MAbs (2018) 10:183–203. doi: 10.1080/19420862.2018.1415671

PubMed Abstract | CrossRef Full Text | Google Scholar

113. Grilo AL, Mantalaris A. The increasingly human and profitable monoclonal antibody market. Trends Biotechnol. (2018) 37:9–16. doi: 10.1016/j.tibtech.2018.05.014

PubMed Abstract | CrossRef Full Text | Google Scholar

114. Strohl WR. Current progress in innovative engineered antibodies. Protein Cell (2018) 9:86–120. doi: 10.1007/s13238-017-0457-8

PubMed Abstract | CrossRef Full Text | Google Scholar

115. Lopes Dos Santos M, Quintilio W, Manieri TM, Tsuruta LR, Moro AM. Advances and challenges in therapeutic monoclonal antibodies drug development. Braz J Pharm Sci. (2018) 54:e01007. doi: 10.1590/s2175-97902018000001007

CrossRef Full Text | Google Scholar

116. Zhou Y, Marks JD. Mechanism of Action for Therapeutic Antibodies. In: Liu C, Morrow KJ, editors. Biosimilars of Monoclonal Antibodies: A Practical Guide to Manufacturing, Preclinical, and Clinical Development. Hoboken, NJ: Wiley (2016). p. 85–111. doi: 10.1002/9781118940648.ch3

CrossRef Full Text | Google Scholar

117. Nell T, Gielen J. The development of a monoclonal antibody against PMSG for a veterinary application. Livestock Prod Sci. (1995) 42:223–8. doi: 10.1016/0301-6226(95)00024-F

CrossRef Full Text | Google Scholar

118. Van Der Lende T. Generation and applications of monoclonal antibodies for livestock production. Biotechnol Adv. (1994) 12:71–87. doi: 10.1016/0734-9750(94)90291-7

CrossRef Full Text | Google Scholar

119. Dieleman SJ, Bevers MM, Wurth YA, Gielen JT, Willemse AH. Improved embryo yield and condition of donor ovaries in cows after PMSG superovulation with monoclonal anti-PMSG administered shortly after the preovulatory LH peak. Theriogenology (1989) 31:473–87. doi: 10.1016/0093-691X(89)90552-9

PubMed Abstract | CrossRef Full Text | Google Scholar

120. Dielman SJ, Bevers MM, Vos PLAM, De Loos FAM. PMSG/anti-PMSG in cattle: a simple and efficient superovulatory treatment? Theriogenology (1993) 39:25–41. doi: 10.1016/0093-691X(93)90022-W

CrossRef Full Text | Google Scholar

121. Martemucci G, D'Alessandro A, Toteda F, Facciolongo AM, Gambacorta M. Embryo production and endocrine response in ewes superovulated with PMSG, with or without monoclonal anti-PMSG administered at different times. Theriogenology (1995) 44:691–703. doi: 10.1016/0093-691X(95)00249-8

PubMed Abstract | CrossRef Full Text | Google Scholar

122. Pintado B, Gutierrez-Adan A, Perez Llano B. Superovulatory response of Murciana goats to treatments based on PMSG/Anti-PMSG or combined FSH/PMSG administration. Theriogenology (1998) 50:357–64. doi: 10.1016/S0093-691X(98)00145-9

PubMed Abstract | CrossRef Full Text | Google Scholar

123. Talwar GP, Sharma NC, Dubey SK, Salahuddin M, Das C, Ramakrishnan S, et al. Isoimmunization against human chorionic gonadotropin with conjugates of processed beta-subunit of the hormone and tetanus toxoid. Proc Natl Acad Sci USA. (1976) 73:218–22. doi: 10.1073/pnas.73.1.218

PubMed Abstract | CrossRef Full Text | Google Scholar

124. Talwar GP, Sharma NC, Dubey SK, Salahuddin M, Shastri N, Ramakrishnan S. Processing of the preparations of beta-subunit of human chorionic gonadotropin for minimization of cross-reactivity with human luteinizing hormone. Contraception (1976) 13:131–9. doi: 10.1016/0010-7824(76)90025-1

PubMed Abstract | CrossRef Full Text | Google Scholar

125. Stevens VC, Crystle CD. Effects of immunization with hapten-coupled HCG on the human menstrual cycle. Obstet Gynecol. (1973) 42:485–95. doi: 10.1097/00006250-197310000-00001

PubMed Abstract | CrossRef Full Text | Google Scholar

126. Talwar GP, Dubey SK, Salahuddin M, Shastri N. Kinetics of antibody response in animals injected with processed beta-HCG conjugated to tetanus toxoid (Pr- beta-HCG-TT). Contraception (1976) 13:153–61. doi: 10.1016/0010-7824(76)90027-5

PubMed Abstract | CrossRef Full Text | Google Scholar

127. Talwar GP, Singh O, Pal R, Chatterjee N, Sahai P, Dhall K, et al. A vaccine that prevents pregnancy in women. Proc Natl Acad Sci USA. (1994) 91:8532–6. doi: 10.1073/pnas.91.18.8532

PubMed Abstract | CrossRef Full Text | Google Scholar

128. Talwar GP, Nand KN, Gupta JC, Bandivdekar AH, Sharma RS, Lohiya NK. Current status of a unique vaccine preventing pregnancy. Front Biosci (Elite Ed). (2017) 9:321–32. doi: 10.2741/e805

PubMed Abstract | CrossRef Full Text | Google Scholar

129. Ferro VA, Mordini E. Peptide vaccines in immunocontraception. Curr Opin Mol Ther. (2004) 6:83–9.

PubMed Abstract | Google Scholar

130. Moulton HM, Yoshihara PH, Mason DH, Iversen PL, Triozzi PL. Active specific immunotherapy with a beta-human chorionic gonadotropin peptide vaccine in patients with metastatic colorectal cancer: antibody response is associated with improved survival. Clin Cancer Res. (2002) 8:2044–51.

PubMed Abstract | Google Scholar

131. Kvirkvelia N, Chikadze N, Makinde J, McBride JD, Porakishvili N, Hills FA, et al. Investigation of factors influencing the immunogenicity of hCG as a potential cancer vaccine. Clin Exp Immunol. (2018) 193:73–83. doi: 10.1111/cei.13131

PubMed Abstract | CrossRef Full Text | Google Scholar

132. Zhu LL, Blair H, Cao J, Yuen T, Latif R, Guo L, et al. Blocking antibody to the beta-subunit of FSH prevents bone loss by inhibiting bone resorption and stimulating bone synthesis. Proc Natl Acad Sci USA. (2012) 109:14574–9. doi: 10.1073/pnas.1212806109

PubMed Abstract | CrossRef Full Text | Google Scholar

133. Liu P, Ji Y, Yuen T, Rendina-Ruedy E, DeMambro VE, Dhawan S, et al. Blocking FSH induces thermogenic adipose tissue and reduces body fat. Nature (2017) 546:107–12. doi: 10.1038/nature22342

PubMed Abstract | CrossRef Full Text | Google Scholar

134. Sponton CH, Kajimura S. Burning fat and building bone by FSH blockade. Cell Metab. (2017) 26:285–7. doi: 10.1016/j.cmet.2017.07.018

PubMed Abstract | CrossRef Full Text | Google Scholar

135. Ji Y, Liu P, Yuen T, Haider S, He J, Romero R, et al. Epitope-specific monoclonal antibodies to FSHbeta increase bone mass. Proc Natl Acad Sci USA. (2018) 115:2192–7. doi: 10.1073/pnas.1718144115

PubMed Abstract | CrossRef Full Text | Google Scholar

136. Zaidi M, Lizneva D, Kim SM, Sun L, Iqbal J, New MI, et al. FSH, bone mass, body fat, and biological aging. Endocrinology (2018) 159:3503–14. doi: 10.1210/en.2018-00601

PubMed Abstract | CrossRef Full Text | Google Scholar

137. Geng W, Yan X, Du H, Cui J, Li L, Chen F. Immunization with FSHbeta fusion protein antigen prevents bone loss in a rat ovariectomy-induced osteoporosis model. Biochem Biophys Res Commun. (2013) 434:280–6. doi: 10.1016/j.bbrc.2013.02.116

PubMed Abstract | CrossRef Full Text | Google Scholar

138. Liu S, Cheng Y, Fan M, Chen D, Bian Z. FSH aggravates periodontitis-related bone loss in ovariectomized rats. J Dent Res. (2010) 89:366–71. doi: 10.1177/0022034509358822

PubMed Abstract | CrossRef Full Text | Google Scholar

139. Qian H, Guan X, Bian Z. FSH aggravates bone loss in ovariectomised rats with experimental periapical periodontitis. Mol Med Rep. (2016) 14:2997–3006. doi: 10.3892/mmr.2016.5613

PubMed Abstract | CrossRef Full Text | Google Scholar

140. Tabatabai LS, Bloom J, Stewart S, Sellmeyer DE. FSH levels predict bone loss in premenopausal women treated for breast cancer more than one year after treatment. J Clin Endocrinol Metab. (2016) 101:1257–62. doi: 10.1210/jc.2015-3149

PubMed Abstract | CrossRef Full Text | Google Scholar

141. Zhu C, Ji Y, Liu S, Bian Z. Follicle-stimulating hormone enhances alveolar bone resorption via upregulation of cyclooxygenase-2. Am J Transl Res. (2016) 8:3861–71.

PubMed Abstract | Google Scholar

142. Allan CM, Kalak R, Dunstan CR, McTavish KJ, Zhou H, Handelsman DJ, et al. Follicle-stimulating hormone increases bone mass in female mice. Proc Natl Acad Sci USA. (2010) 107:22629–34. doi: 10.1073/pnas.1012141108

PubMed Abstract | CrossRef Full Text | Google Scholar

143. Rouach V, Katzburg S, Koch Y, Stern N, Somjen D. Bone loss in ovariectomized rats: dominant role for estrogen but apparently not for FSH. J Cell Biochem. (2011) 112:128–37. doi: 10.1002/jcb.22908

CrossRef Full Text | Google Scholar

144. Drake MT, McCready LK, Hoey KA, Atkinson EJ, Khosla S. Effects of suppression of follicle-stimulating hormone secretion on bone resorption markers in postmenopausal women. J Clin Endocrinol Metab. (2010) 95:5063–8. doi: 10.1210/jc.2010-1103

PubMed Abstract | CrossRef Full Text | Google Scholar

145. Crawford ED, Schally AV, Pinthus JH, Block NL, Rick FG, Garnick MB, et al. The potential role of follicle-stimulating hormone in the cardiovascular, metabolic, skeletal, and cognitive effects associated with androgen deprivation therapy. Urol Oncol. (2017) 35:183–91. doi: 10.1016/j.urolonc.2017.01.025

PubMed Abstract | CrossRef Full Text | Google Scholar

146. Uihlein AV, Finkelstein JS, Lee H, Leder BZ. FSH suppression does not affect bone turnover in eugonadal men. J Clin Endocrinol Metab. (2014) 99:2510–5. doi: 10.1210/jc.2013-3246

PubMed Abstract | CrossRef Full Text | Google Scholar

147. Finkelstein JS, Lee H, Burnett-Bowie SA, Pallais JC, Yu EW, Borges LF, et al. Gonadal steroids and body composition, strength, and sexual function in men. N Engl J Med. (2013) 369:1011–22. doi: 10.1056/NEJMoa1206168

CrossRef Full Text | Google Scholar

148. Kumar TR. Extragonadal actions of FSH: a critical need for novel genetic models. Endocrinology (2018) 159:2–8. doi: 10.1210/en.2017-03118

PubMed Abstract | CrossRef Full Text | Google Scholar

149. Cummings SR, San Martin J, McClung MR, Siris ES, Eastell R, Reid IR, et al. Denosumab for prevention of fractures in postmenopausal women with osteoporosis. N Engl J Med. (2009) 361:756–65. doi: 10.1056/NEJMoa0809493

PubMed Abstract | CrossRef Full Text | Google Scholar

150. Gupta SK, Talwar GP. Development of hybridomas secreting anti-human chorionic gonadotropin antibodies. Indian J Exp Biol. (1980) 18:1361–5.

PubMed Abstract | Google Scholar

151. Kathuria S, Sriraman R, Nath R, Sack M, Pal R, Artsaenko O, et al. Efficacy of plant-produced recombinant antibodies against HCG. Hum Reprod. (2002) 17:2054–61. doi: 10.1093/humrep/17.8.2054

PubMed Abstract | CrossRef Full Text | Google Scholar

152. Nand KN, Gupta JC, Panda AK, Jain SK. Development of a recombinant hCG-specific single chain immunotoxin cytotoxic to hCG expressing cancer cells. Protein Expr Purif. (2015) 106:10–7. doi: 10.1016/j.pep.2014.10.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: potentiating antibodies, inhibitory antibodies, follicle-stimulating hormone, luteinizing hormone, chorionic gonadotropin, signaling

Citation: Kara E, Dupuy L, Bouillon C, Casteret S and Maurel M-C (2019) Modulation of Gonadotropins Activity by Antibodies. Front. Endocrinol. 10:15. doi: 10.3389/fendo.2019.00015

Received: 30 November 2018; Accepted: 10 January 2019;
Published: 18 February 2019.

Edited by:

Livio Casarini, Università Degli Studi di Modena e Reggio Emilia, Italy

Reviewed by:

Marco Bonomi, Istituto Auxologico Italiano (IRCCS), Italy
Luca De Toni, University of Padova, Italy

Copyright © 2019 Kara, Dupuy, Bouillon, Casteret and Maurel. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Elodie Kara, elodie.kara@igyxos.com

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.