Você está na página 1de 27

REVIEW

Gonadotropins and Their Analogs: Current and


Potential Clinical Applications

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/911/5047286 by Research 4 Life user on 22 November 2018


Ross C. Anderson,1,2 Claire L. Newton,1,3 Richard A. Anderson,4 and Robert P. Millar1,2,3,5,6

1
Centre for Neuroendocrinology, University of Pretoria, Pretoria 0001, South Africa; 2Department of
Physiology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; 3Department of
Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; 4MRC Centre for
Reproductive Health, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ,
United Kingdom; 5Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of
Cape Town, Cape Town 7700, South Africa; and 6Institute of Infectious Diseases and Molecular Medicine,
University of Cape Town, Cape Town 7700, South Africa

ABSTRACT The gonadotropin receptors LH receptor and FSH receptor play a central role in governing reproductive competency/fertility.
Gonadotropin hormone analogs have been used clinically for decades in assisted reproductive therapies and in the treatment of various
infertility disorders. Though these treatments are effective, the clinical protocols demand multiple injections, and the hormone preparations
can lack uniformity and stability. The past two decades have seen a drive to develop chimeric and modified peptide analogs with more
desirable pharmacokinetic profiles, with some displaying clinical efficacy, such as corifollitropin alfa, which is now in clinical use. More
recently, low-molecular-weight, orally active molecules with activity at gonadotropin receptors have been developed. Some have excellent
characteristics in animals and in human studies but have not reached the market—largely as a result of acquisitions by large pharma.
Nonetheless, such molecules have the potential to mitigate risks currently associated with gonadotropin-based fertility treatments, such as
ovarian hyperstimulation syndrome and the demands of injection-based therapies. There is also scope for novel use beyond the current
remit of gonadotropin analogs in fertility treatments, including application as novel contraceptives; in the treatment of polycystic ovary
syndrome; in the restoration of function to inactivating mutations of gonadotropin receptors; in the treatment of ovarian and prostate
cancers; and in the prevention of bone loss and weight gain in postmenopausal women. Here we review the properties and clinical
application of current gonadotropin preparations and their analogs, as well as the development of novel orally active, small-molecule
nonpeptide analogs. (Endocrine Reviews 39: 911 – 937, 2018)

G lobally, many couples desiring children expe-


rience problems with infertility (). With the
global demand for assisted reproductive technologies
provide an up-to-date review of the development,
properties, and activities of peptide and nonpeptide
gonadotropin analogs and their current and potential
(ARTs), there has been a drive toward the discovery clinical uses. The authors searched PubMed and
and development of effective therapeutic modula- Google Patents databases for original articles, appro-
tors of human reproduction. Central to this is the use priate reviews, and patent applications focused on
of gonadotropins [FSH, LH, and human chorionic gonadotropin receptors, gonadotropins, gonadotropin
gonadotropin (hCG)] and their analogs. Here, we analogs, and ARTs published until February .
ISSN Print: 0163-769X
ISSN Online: 1945-7189
Gonadotropins and Their Receptors The core events that define this system include the Printed: in USA
pulsatile release of GnRH from ~ neurons in the Copyright © 2018
hypothalamus () into the hypophyseal portal capillary Endocrine Society
The hypothalamic-pituitary-gonadal axis
Received: 14 February 2018
The hypothalamic-pituitary-gonadal (HPG) axis is an blood from where it is delivered to the anterior pituitary.
Accepted: 25 June 2018
endocrine signaling conduit that governs pubertal de- Binding and activation of its cognate receptor, the First Published Online:
velopment and reproductive competency in humans. GnRH receptor (GnRHR), on gonadotrope cells of the 2 July 2018

doi: 10.1210/er.2018-00052 https://academic.oup.com/edrv 911


REVIEW

ESSENTIAL POINTS
· Urinary gonadotropin analogs have been used clinically for decades in assisted reproductive therapies and in the
treatment of various infertility disorders
· Issues with the gonadotropin peptide hormones include clinical protocols requiring multiple injections, nonuniformity of
preparations, protein stability, and increased risk of ovarian hyperstimulation syndrome
· More recently, recombinant gonadotropin preparations have been favored as a result of their improved purity and batch-
to-batch consistency
· Gonadotropin hormones with differing half-lives and oral bioavailability may mitigate the need for repeated injections

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/911/5047286 by Research 4 Life user on 22 November 2018


and reduce side-effects associated with current treatment regimens
· Several peptide analogs have been developed, one of which, an FSH analog with an increased half-life (corifollitropin alfa),
has entered the clinic
· A spectrum of orally available, low-molecular-weight gonadotropin analogs (agonists and antagonists) has been in
development, but none is yet commercially available, and many development programs have been terminated as a result
of pharmaceutical company mergers or acquisitions
· In addition to use in assisted reproductive therapies, gonadotropin analogs may have potential application as
contraceptives and in the treatment of polycystic ovary syndrome and ovarian and prostate cancers and prevention of
bone loss and weight gain in postmenopausal women

anterior pituitary stimulate the production and secre- and females. Whereas LH pulse frequency remains
tion of the gonadotropin hormones LH and FSH, which constant in males, it varies over the female menstrual
are released into the general circulation to stimulate cycle with low-frequency pulses during the luteal
gonadal steroidogenesis, peptide hormone secretion phase of the cycle and higher-frequency pulses in the
(inhibin and activin), and gametogenesis. Gonadal follicular phase. FSH pulsatility is less discernible, as
steroids and peptide hormones then feed back in unlike LH, FSH is not stored in secretion granules and
negative and positive modalities at the hypothalamus has a longer half-life in the circulation. In females, FSH
and/or pituitary (, ). activity in the late-luteal/early-follicular phases of the
cycle is responsible for recruitment of five to seven
The actions of gonadotropins in the HPG axis Graafian follicles into the menstrual cycle and drives
The gonadotropin glycoproteins exert their effects on follicular development. Estradiol, produced by the
the HPG axis through interaction with and activation developing follicle, feeds back negatively to the hy-
of their cognate receptors, the LH receptor (LHCGR/ pothalamus, resulting in suppressed LH secretion.
LHR) and FSH receptor (FSHR). Whereas a number of Through poorly defined mechanisms, an estradiol
studies have documented the presence of LHR and threshold is reached, at which point estradiol switches
FSHR in extragonadal tissues (, ), physiologically, to positive feedback, resulting in a midcycle increase in
their primary site of activity is the gonads. FSHR LH pulse frequency and surge of LH, which are re-
activation stimulates follicular development in females sponsible for initiating ovulation and luteinization of
and the initiation of spermatogenesis in males whereas the ruptured follicle. The luteinized follicle, or corpus
LHR stimulation is responsible for steroidogenesis and luteum, subsequently produces progesterone that
follicular/gamete maturation, as well as the induction prepares the uterine endometrium for implantation.
of ovulation in females. LH regulates expression of Progesterone also feeds back negatively at the hypo-
steroidogenic enzymes and the subsequent production thalamus, suppressing LH secretion, resulting in at-
of androgens within the interstitial Leydig and thecal rophy of the corpus luteum should implantation not
cells of the testes and ovaries, respectively. Steroido- occur.
genesis is also dependent on the activity of FSH in The importance of both glycoprotein hormones
testicular Sertoli cells and the granulosa cells of the and their receptors in gonadal function is highlighted
ovaries, supporting gonadal development and facili- by the pathophysiologies attributed to inactivating (or
tating the conversion of androgen to estrogen via the constitutively activating) mutations that affect the LH/
enzyme aromatase. Activation of FSHR additionally LHR and FSH/FSHR signaling modalities. Loss of
results in the production of the TGF-b family functional LH and FSH in males and females can
members activin and inhibin, which positively and result in impaired fertility/infertility phenotypes and in
negatively feed back to the pituitary and hypothalamus the case of LH, a lack of pubertal development (, ).
to regulate FSH production/activity (). The severity of the phenotype attributed to muta-
The gonadotropin-secretion profiles are also sex- tions in the LHR correlates with the degree of receptor
ually dimorphic, differing dramatically between males inactivation and a resultant spectrum of phenotypes from

912 Anderson et al Peptide and Nonpeptide Gonadotropin Analogs Endocrine Reviews, December 2018, 39(6):911–937
REVIEW

pseudohermaphroditism and hypogonadism through and both activate the LHR with similar potencies in
to the less severe micropenis and oligomenorrhea (). vitro (). However, the CTP confers an increased half-
Interestingly, loss of FSH/FSHR signaling in females life to hCG. The a- and b-subunit heterodimers form
results in infertility, in contrast to males, where through noncovalent interactions, whereas the intra-
homozygous-inactivating FSH/FSHR mutations appear subunit tertiary structure is largely governed by
to result in a qualitative and quantitative reduction in disulfide bonds (), with the a- and b-subunits
spermatogenesis without rendering the patient com- containing  and  cysteine residues, respectively
pletely infertile, implying a level of redundancy in FSHR (). The resolution of the crystal structure of hCG
signaling with regard to male fertility (–). These demonstrated that the a- and b-subunit interaction is
findings appear to be supported by an FSH-deficient stabilized by a cysteine loop of the b-subunit that

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/911/5047286 by Research 4 Life user on 22 November 2018


mouse model in which female mice had folliculogenesis encircles loop two of the a-subunit in a manner
defects and were infertile, whereas males displayed im- resembling a seatbelt (). Two disulfide bonds,
paired testes development but remained fertile (). conserved in all of the human glycoprotein hormones,
are responsible for the “locking” of the seatbelt around
Regulation of gonadotropin secretion the a-subunit (). Whereas the a- and b-subunits
In both male and female systems, the processes reg- both contribute to receptor binding, molecular
ulating the production and secretion of the gonado- modeling and the use of chimeric proteins have
tropins are complex and represent an intricate identified a “determinant” loop in the b-subunit (aa 
relationship among hormonal and steroidal produc- to ) that confers receptor binding specificity
tion, secretion, and feedback (, , ). As men- (–).
tioned previously, the secretion of LH and FSH from The mature gonadotropin hormones are heavily
gonadotropes is sexually dimorphic, and in females, glycosylated. The common a-subunit has two N-
the circulating hormone levels fluctuate dramatically linked glycosylation sites (N and N), with the
throughout the menstrual cycle. This differential hormone-specific b-subunits containing either one
regulation is the result of a complex interplay of a (LH: N) or two (FSH: N and N and hCG: N
multitude of physiological inputs, including metabolic and N) N-linked sites (). The hCG CTP extension
status, stress, and inflammatory cues, in addition to also contains an additional four O-linked glycosylation
steroidal and peptide feedback mechanisms into the sites. Chemical and enzymatic removal of the oligo-
hypothalamus and pituitary (–). GnRH is released saccharide moieties or removal of a- and b-subunit
in a pulsatile manner (, ), and it has long been glycosylation sites via site-directed mutagenesis has
known that each pulse of GnRH results in a con- revealed a number of important roles for these resi-
comitant pulse of LH transcription and secretion (). dues. Whereas the function of O-linked glycosylation
Pulses of FSH can also be detected following GnRH of the CTP appears to be predominantly related to
stimulation of gonadotropes, but in contrast to LH, reduced clearance rates and increased half-life and
this regulation appears to be at the level of FSH biological activity (, ), N-linked glycosylation
b-subunit transcription, as FSH secretion appears to appears to be more complex. Evidence for roles in
be largely constitutive (–). The issue of how GnRH determining clearance rates has been reported, with a
differentially regulates the contrasting profiles of the number of studies suggesting that the number of
two gonadotropins across the menstrual cycle was terminal sialic acid or sulfonated N-acetylgalactos-
resolved following the discovery that LH and FSH amine residues present on the glycan determines the
transcription was differentially affected by GnRH pulse rate of hepatic clearance, with increased sialylation
frequency (, ). High-frequency pulses of GnRH reflecting a longer half-life and increased sulfonation
appear to favor LH, whereas low-frequency pulses resulting in faster clearance (–). Roles in receptor
promote FSH synthesis. activation have also been demonstrated, as deletion of
the N-linked glycosylation residues reduces biological
Gonadotropin hormone structure and function activity with only a marginal effect on receptor binding
The pituitary gonadotropin hormones LH and FSH, as (, –). Mutagenesis studies have demonstrated
well as the placentally derived LH paralog hCG, are all that removal of the a-subunit N site affects bio-
large, heterodimeric glycoproteins that consist of a- activity (, –), whereas loss of b-subunit gly-
and b-subunits. Along with the pituitary-derived TSH, cosylation sites may exacerbate the loss of bioactivity
this group of hormones is termed the glycoprotein induced by loss of N (). Roles of glycosylation in
hormones. The glycoprotein hormone heterodimers subunit synthesis and heterodimer secretion have also
are composed of a -amino acid (aa) a-subunit been indicated (, , , ).
encoded by a single gene common to all three hor- The nature, as well as the presence, of the glycosylation
mones () and a hormone-specific b-subunit (LH: also appears to affect gonadotropin bioactivity. Charge
 aa; hCG:  aa; FSH:  aa; and TSH:  aa). variants (due to differential glycosylation) and variants
LH and hCG differ only by a C-terminal, -aa peptide with the same net charge but different glycosylation status
[carboxy-terminal peptide (CTP)] extension in hCG, can affect biological activity. For example, basic isoforms

doi: 10.1210/er.2018-00052 https://academic.oup.com/edrv 913


REVIEW

(reduced sialylation) display higher binding affinities and glycoprotein hormones interact with this large N-
biopotencies in heterologous cell-assay systems than the terminal domain (). The LRRs have been demon-
acidic isoforms, and some of the most basic isoforms strated to form a slightly curved solenoid, with the
(pH . .) have antagonistic activity (–). In- hormone binding to the curved inner surface.
triguingly, antagonism of GnRHR has been shown to The receptor wraps around the central portion of the
result in elaboration of a greater proportion of these basic hormone through both hormone subunits and occurs
forms of FSH, which act as antagonists (). Differentially over a large interface, described as a “hand clasp.” This
glycosylated FSH isoforms appear to have physiological interaction is mediated via a number of charge in-
relevance, as during puberty and the transition from teractions, salt bridges, and aromatic ring-stacking
early-to-late follicular phase, there is a progressive increase interactions (, ). A region proximal to the ECL/

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/911/5047286 by Research 4 Life user on 22 November 2018


in the ratio of basic-to-acidic isoforms, and this appears to TM domains, which is designated the “hinge region,”
be tightly controlled by estradiol (). Moreover, different is known to play an important role in the activation of
FSH glycoforms are apparently capable of activating the receptor following hormone binding. The mech-
different FSHR signaling modalities, with a number of anism through which activation occurs is not fully
studies documenting differential recruitment of down- known, but resolution of the crystal structure of FSH,
stream signaling components (Gas, Gai, and b-arrestin) in complex with the entire ectodomain of FSHR
and differences in cAMP release, estradiol production, (including the hinge region), suggested a possible
tissue-type plasminogen activator, cytochrome P mechanism via a second hormone-interaction site at a
aromatase, and a-inhibin subunit expression (, , , sulfated tyrosine in the C-terminus of the hinge (Y)
, , ). (, ). This interaction is proposed to lift the hinge
region, leading to activation of the receptor through
Gonadotropin receptor structure and function the unmasking of an intramolecular agonist sequence
The gonadotropin receptors are members of the juxtaposed to the ECL/TM (Fig. ) (, ). This is in
glycoprotein-hormone receptor subfamily of the contrast to previous hypothesized mechanisms,
rhodopsin-like class of G protein–coupled receptors whereby the hormone itself was proposed to make
(GPCRs) and have the prototypical seven trans- direct contact with the ECLs and juxtamembrane
membrane (TM) domain architecture of GPCRs. The regions of the TM domains to induce receptor ac-
transmembrane (TM) domains are linked by three tivation ().
intracellular loops and three extracellular loops (ECLs)
with an intracellular C-terminus and an extracellular
N-terminal domain (ECD). Ligand activation of Gonadotropin Analogs in Clinical Use
GPCRs promotes association with intracellular GDP-
bound heterotrimeric G proteins. The activated re- The importance of the pituitary in reproductive
ceptor acts as a guanine-nucleotide exchange factor, physiology was determined in the early th century.
promoting GTP displacement of GDP on the G The pioneering work of Bernhard Zondek in the
protein Ga-subunit. This exchange promotes disso- s and s linked the secretion of gonado-
ciation of the heterotrimeric G protein into a tropins (then termed prolan A and B) from the
monomeric GTP-Ga-subunit and a heterodimeric pituitary to follicular growth and steroidogenesis
Gbg-subunit. The dissociated subunits then activate (), paving the way for the application of gonad-
intracellular signaling pathways. In the case of the otropins in clinical medicine. To this end, several
gonadotropin receptors, the activated Ga-subunits are gonadotropin analogs/preparations have been devel-
responsible for activation of the enzyme adenylyl oped for clinical use. Indeed, urinary hCG (pregnyl)
cyclase, resulting in the production of the second has been available for therapeutic application from as
messenger cAMP. Whereas cAMP is acknowledged as early as  ().
the “classical” intracellular signal, generated following
gonadotropin receptor activation, inositol phosphate/ Purified/recombinant gonadotropin preparations
Ca+, MAPK (ERK and p), and phosphatidylinositol Initial preparations of gonadotropins for clinical use
-kinase/AKT signaling modalities have also been were derived from pituitary extracts; however, this
reported (–). Moreover, the phenomena of “biased practice was discontinued, as the supply of human
agonism,” whereby specific ligands preferentially recruit pituitaries could not meet the demand and also, the
certain signaling pathways, which is well described for recognition of a possible link to the transmission of
other GPCRs, has only recently emerged for gonado- prion disease (). Endogenous gonadotropins are
tropin receptors, predominantly through the use of now extracted from the urine of pregnant or post-
small molecule agonists (discussed later). menopausal women, including chorionic gonadotro-
The glycoprotein hormone receptors are unique pin (hCG), urofollitropin (FSH), and menotropin/
among GPCRs in having very large ECDs (. aa), human menopausal gonadotropin (hMG; which
consisting of a cysteine-rich domain, followed by contains both FSH and LH). These preparations
several leucine-rich repeats (LRRs) (, ). The continue to be widely used, despite issues regarding a

914 Anderson et al Peptide and Nonpeptide Gonadotropin Analogs Endocrine Reviews, December 2018, 39(6):911–937
REVIEW

Figure 1. Two-step FSH/FSHR recognition and activation. The ECD LRRs of FSHR, in a putative orientation relative to the seven
transmembrane (7TM) domain, are shown as a magenta block with a hinge, and the 7TM domain is shown as a cylinder, with the
inactivated state colored gray and the activated state colored green. Sulfated Y335 is shown as a yellow ball, residue S273 is shown as
a green star, and disulfide bonds are shown as a brown stick. Heterotrimeric Gs or b-arrestin protein is shown as a green ellipsoid. FSH
heterodimer is represented in purple, with carbohydrates at N52a shown as a yellow, Y-shaped stick. (a) Inactive receptor is bound by
hormone, (b) resulting in Y335 in the receptor hinge lifting via interaction with the hormone, permitting (c) activation of the 7TM
domain. HBSD, hormone-binding subdomain; SSSD, signal-specificity subdomain. Reproduced with permission from Jiang X, Liu H,
Chen X, et al. Structure of follicle-stimulating hormone in complex with the entire ectodomain of its receptor. Proc Natl Acad Sci USA
2012; 109(31):12491–12496. [© 2018 Illustration Presentation ENDOCRINE SOCIETY]

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/911/5047286 by Research 4 Life user on 22 November 2018


lack of regulatory control, batch-to-batch variation, which the CTP of hCGb is fused to the C-terminus of
and limited source availability (Table ). However, the b-subunit of FSH to increase the biological half-
continued improvement of purification methodolo- life. The chimeric b-subunit (FSH-CTP) does not
gies has enabled the production of highly purified affect biosynthesis or secretion of the heterodimer in
preparations. cells used for production of recombinant protein.
The advent of the molecular era resulted in the Moreover, FSH binding to rat testes membrane
availability of recombinant gonadotropins [follitropin preparations and steroidogenic activity in granulosa
alfa/beta/delta (FSH), lutropin alfa (LH), and cho- cells were retained (). Furthermore, the in vivo
riogonadotropin alfa (hCG) (Table )], allowing for potency (measured as increased ovarian weight and
the production of highly pure preparations, batch-to- granulosa cell aromatase activity following adminis-
batch consistency, and flexible dosing regimens and tration of corifollitropin alfa to immature, estrogen-
protocols, resulting in more effective personalized- primed rats) and biological half-life of corifollitropin
treatment regimens. Responses to urinary and recombi- alfa were significantly increased compared with wild-
nant gonadotropin preparations are similar (), and type FSH, whereas no LHR binding was detected ().
because of the limited evidence of benefit in assisted A phase I nonblind multicenter study followed, in
reproduction and associated cost implications, recombi- which hypogonadotropic hypogonadal (HH) men
nant gonadotropins have only partly replaced urinary were injected subcutaneously (SC), four times with
hCG/hMG in fertility treatments.  mg corifollitropin alfa at -week intervals. No
Different sources and preparations of gonadotro- serious adverse effects or generation of antibodies
pins (both recombinant and human derived) can vary against the protein were reported, and the elimina-
in their receptor binding and signal-transduction tion half-life for corifollitropin alfa of  hours was
capabilities, and this is related to their glycosylation considerably longer than that of follitropin beta
status. For example, differences in the clinical activity (Puregon;  to  hours) (). In a subsequent study,
of different preparations have been attributed to in healthy females of reproductive age, a high dose of
carbohydrate content, which varies depending on the oral contraceptive (Lyndiol) was administered to
methods used for their production and/or purification suppress endogenous gonadotropin secretion, fol-
(). lowed by , , , or  mg SC injection of FSH-
CTP. As in the HH males, no antibodies against the
Gonadotropin chimeras (corifollitropin alfa) compound were detected, and increases in serum
In addition to enabling the production of large inhibin B concentration and follicular growth (both
quantities of native heterodimer, recombinant tech- number and size of follicles) were observed in a dose-
nologies have been used to generate chimeric go- dependent manner. The elimination half-life was
nadotropins, such as corifollitropin alfa (Org ; approximately twofold slower than follitropin alfa
Table  and Table ), an FSH heterodimeric protein in (Gonal-F). Interestingly, the time to reach maximum

doi: 10.1210/er.2018-00052 https://academic.oup.com/edrv 915


REVIEW

Table 1. Commercially Available Gonadotropin Preparations


Hormone Source Generic Name Example Brand Names (Manufacturer)

hMG Urinary Menotropin EEMA-HMG (Corona Remedies)

Eventin/Eventin HP (Maneesh Pharmaceuticals)

GMH/GMH HP (Sun Pharma)

Gynogen HP (Sanzyme)

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/911/5047286 by Research 4 Life user on 22 November 2018


Humegon (Organon)

Humog/Humog HP (Bharat Serums and Vaccines)

IVF-M/IVF-M HP (LG Lifesciences)

Materna HMG (Emcure)

Menopur (Ferring Pharmaceuticals)

Menogon (Ferring Pharmaceuticals)

Repronex (Ferring Pharmaceuticals)

Merional/Eigenorm/Meriofert (IBSA)

Normegon (Organon)

Ovulate-M (VHB Life Sciences)

Persinal (Serum Institute of India)

Puregraf (GUFIC)

ZyHMG/ZyHMG HP (Zydus Cadila)

Pergonal (Serono Laboratories)

FSH Urinary Urofollitropin Bravelle (Ferring Pharmaceuticals)

Endogen HP (Sanzyme)

EEMA-FSH (Corona Remedies)

Fertinex (Serono Laboratories)

Foliculin/Foliculin HP (Bharat Serums and Vaccines)

Foligem (VHB Life Sciences)

Follicare (GUFIC)

Follimon (LG Lifesciences)

Folliova (Sanzyme)

Fostimon/Fostipur/Altermon (IBSA)

Materna FSH (Emcure)

Metrodin (Serono Laboratories)

Metrodin HP/ Fertinorm HP (Serono Laboratories)

Neogentin (Maneesh Pharmaceuticals)

Orgafol (Organon)

Ovitrop (Sun Pharma)

Sitrodin (Serum Institute of India)

ZyFSH/ZyFSH HP (Zydus Cadila)


(Continued )

916 Anderson et al Peptide and Nonpeptide Gonadotropin Analogs Endocrine Reviews, December 2018, 39(6):911–937
REVIEW

Table 1. Continued
Hormone Source Generic Name Example Brand Names (Manufacturer)

Follitropin alfa Gonal-F (EMD Serono/Merck Serono)

Bemfola (Finox Biotech)

Cinnal-F (CinnaGen)

EEMA-rFSH (Corona Remedies)

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/911/5047286 by Research 4 Life user on 22 November 2018


Folligraf (Bharat Serums and Vaccines)

Ovaleap (Teva Pharma B.V.)

Recombinant Follitropin beta EEMA-rFSH (Corona Remedies)

Follistim/Puregon/Fertavid (Merck & Co./MSD)

Recagon (Organon India)

Follitropin delta Rekovelle (Ferring Pharmaceuticals)

FSH chimera Recombinant Corifollitropin alfa Elonva (MSD)

LH Recombinant Lutropin alfa Luveris (EMD Serono/Merck Serono)

FSH/LH mix (2:1 ratio) Recombinant Follitropin alfa:lutropin alfa Pergoveris (EMD Serono/Merck Serono)

hCG Urinary Chorionic gonadotropin Pregnyl (Merck & Co./MSD)

A.P.L. (Wyeth-Ayerst)

Brevactid (Ferring Pharmaceuticals)

Choragon (Ferring Pharmaceuticals)

Choriomon/Gonasi HP (IBSA)

Chorionic gonadotropin (Ferring Pharmaceuticals)

EEMA-HP (Corona Remedies)

Endocorion (ELEA)

Fertigyn (Sun Pharma)

Follutein (Bristol-Myers Squibb)

Hucog HP (Bharat Serums and Vaccines)

IVF-C (LG Lifesciences)

Materna-hCG (Emcure)

Novarel (Ferring Pharmaceuticals)

Ovipure HP (Bharat Serums and Vaccines)

Ovulate-C (VHB Life Sciences)

Ovutrig HP (VHB Life Sciences)

Profasi/Profasi HP (Serono Laboratories)

Proferti-C (CBC Pharma)

Provigil (Maneesh Pharmaceuticals)

Pubergen HP (Sanzyme)

Puretrig (GUFIC)

ZyhCG/ZyhCG HP (Zydus Cadila)


(Continued )

doi: 10.1210/er.2018-00052 https://academic.oup.com/edrv 917


REVIEW

Table 1. Continued
Hormone Source Generic Name Example Brand Names (Manufacturer)

Sifasi (Serum Institute of India)

Recombinant Choriogonadotropin alfa Ovidrel/Ovitrelle (EMD/Merck Serono)

Bold text, originator brands; italic text, marketing withdrawn.


Abbreviation: MSD, Merck Sharpe & Dohme.

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/911/5047286 by Research 4 Life user on 22 November 2018


serum concentration was also slower than that de- increased incidence of OHSS was reported in the
scribed for recombinant FSH, possibly as a result of Engage, Ensure, and Trust trials (, , ), women
slower absorption (, –). with polycystic ovarian syndrome (PCOS) or with a
In , pregnancy and live birth after use of history of OHSS had been excluded from these trial
corifollitropin alfa in an ovarian hyperstimulation cohorts. Therefore, the suitability of corifollitropin alfa
protocol were reported. In this phase II trial, a -year- for higher-risk patients remains to be determined.
old woman, who had primary infertility and two failed Following phase III trials, in early , corifolli-
in vitro fertilization cycles, was injected with  mg tropin alfa (under the trade name of Elonva; Table )
corifollitropin alfa SC. After  days, daily injections of was approved for use in the European Union in
follitropin beta (Puregon;  IU), in combination women undergoing controlled ovarian hyperstimu-
with a GnRH antagonist, were administered for  days, lation with a GnRH antagonist-assisted reproduction
after which final oocyte maturation and ovulation protocol who have no history of OHSS or PCOS.
were induced by administration of , IU hCG. Of
the  oocytes that were retrieved,  were fertilized via
intracytoplasmic sperm injection, and two embryos Therapeutic Applications of
were implanted, resulting in one live birth (). Gonadotropin Analogs
Subsequent studies showed that pregnancy rates in a
cohort of  patients undergoing ovarian stimula- Gonadotropin therapy in men
tion with a GnRH antagonist protocol were almost The differential sites of action of LH and FSH at Leydig
indistinguishable between women treated with one SC and Sertoli cells, respectively, underpin their clinical
injection of corifollitropin alfa ( mg) or  daily use in the support of steroidogenesis and of sper-
injections of recombinant FSH ( IU). Additional matogenesis in men suffering from HH. hCG
efficacy and dosing studies have demonstrated that provides a very convenient form of LH-like activity
corifollitropin alfa injections have favorable pharma- and is generally administered at doses of  to 
cokinetic profiles and comparable, or moderately IU, twice per week, with the opportunity to titrate the
improved, pharmacodynamic responses compared dose, depending on the serum testosterone level
with recombinant FSH. This suggests that a single achieved. These doses are, however, far higher than
weekly injection of corifollitropin alfa could replace those needed to normalize intratesticular testosterone
daily injections of recombinant FSH during ovarian concentrations in men with induced experimental
hyperstimulation protocols (–). gonadotropin deficiency. Thus, low doses of hCG (
As a result of the extended duration of action of IU) on alternate days can normalize serum testos-
corifollitropin alfa and the inability to control dose terone levels (). FSH (either urinary or recombinant)
after administration, there is a risk that ovarian hy- is conventionally administered in addition to hCG.
perstimulation syndrome (OHSS; the development of However, in adult men with previously normal
multiple luteinized ovarian cysts, leading to ovarian spermatogenesis who have subsequently become
enlargement and development of vascular hyper- gonadotropin deficient, for example, as a result of
permeability) may be exacerbated in patients treated pituitary surgery, hCG alone can often restore
with this long-acting gonadotropin analog. However, spermatogenesis. On the other hand, men who have
adverse events, including OHSS, in two additional never achieved spermatogenesis also require FSH.
double-blind randomized trials in healthy women Whereas overall, the prospects for spermatogenesis
(Engage) and women of lower body weight (Ensure). are high (~%) in men with either prepubertal- or
were of similar frequency to that observed in pa- postpubertal-onset HH, it is more successful in men
tients treated with recombinant FSH (, ). A with postpubertal onset (% vs %), with higher
further phase III trial (Trust) that studied the effects of sperm concentrations also being achieved (. vs .
repeated administration of  mg corifollitropin alfa million/ml). Prior androgen therapy was believed to be
in up to three controlled ovarian hyperstimulation associated with a slower response to gonadotropin
cycles concluded that there was no immunogenic- therapy (), but a meta-analysis of  studies revealed
ity or drug-related hypersensitivity (). Although no no association (). However, testicular volume is a

918 Anderson et al Peptide and Nonpeptide Gonadotropin Analogs Endocrine Reviews, December 2018, 39(6):911–937
REVIEW

positive prognostic indicator of response, as is previous with testosterone and with similar final testicular
treatment with gonadotropins, whereas cryptorchi- volumes (). Thus, in keeping with a meta-analysis in
dism is an important negative predictor. The long- adult men (), previous testosterone therapy was not
acting version of FSH, corifollitropin alfa, has also been found to be disadvantageous, although a possible
used in hypogonadotropic men, and in a small open advantage of gonadotropin-induced puberty would be
label trial, spermatogenesis was established in % of that spermatogenesis can subsequently be more rap-
men who remained azoospermic after treatment with idly induced in men previously treated with gonad-
hCG alone (). Administration of this long-acting otropins when fertility is desired.
analog, once every  weeks, greatly reduces the
treatment burden, and on the basis of this limited Gonadotropin therapy in women

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/911/5047286 by Research 4 Life user on 22 November 2018


evidence, it appears similarly efficacious to FSH. Gonadotropins can be used to induce ovulation, either
It is important to recognize that quantitatively, to stimulate mono-ovulation (for support of natural
normal spermatogenesis is often not achieved in these conception or intrauterine insemination) or in ARTs,
cases and additionally, that significant duration of where a multifollicular response is desired (conven-
treatment may be required before a detectable sperm tionally called “controlled superovulation,” albeit with
concentration is achieved. Thus, in one large study, the less control than that term indicates).
median time to achieve the first detectable sperm was
. months (% confidence interval, . to .), with Mono-ovulation
conception achieved after a median of . months The ability of exogenous gonadotropin preparations to
(. to .). The median sperm concentration at induce ovulation in both hypogonadotropic women
natural conception was . million/ml, with the lowest and in those with normogonadotropic anovulation
sperm concentration at conception at . million/ml (generally as a result of PCOS) has been recognized, as
(). Thus, men should be advised that treatment may such preparations became available in the late s
be required for at least  years. Moreover, the age of the (). This approach excludes the normal, negative-
female partner can be an important determinant of feedback pathways that fine tune LH and FSH se-
successful pregnancy. A commonly used protocol is cretion from the pituitary to ensure selection and
initiation of androgen production with the use of hCG ovulation of just one follicle from many antral follicles
alone for ~ months and addition of FSH at that stage present in the ovary at the start of the menstrual cycle.
unless sperm are already detectable in the ejaculate. To minimize multifollicular development and the
The main advantages of this approach are to save on resultant risk of a multiple pregnancy, the current
the cost of FSH and also, to reduce the frequency of regimen of choice is the low-dose step-up protocol,
injections, from the five required for administration of wherein low initial doses of gonadotropins (generally
both gonadotropins, to two per week for just hCG. It is  to  IU FSH) are increased only at infrequent
possible that administration of both gonadotropins intervals, initially after  weeks and thereafter, at
from the start may result in more rapid onset of -week intervals (). A step-down protocol, aiming to
spermatogenesis, but robust data supporting this are mimic more closely physiologic changes in FSH, has
absent. Additionally, pretreatment with FSH alone has also been developed with good success rates () but
been proposed, and in men with congenital HH has not been widely adopted.
without prior gonadotropin treatment, a small trial Recombinant/urinary FSH is generally used for
indicated that this approach could be successful (). women with PCOS (as they are not LH deficient) who
FSH treatment of  months before GnRH (thus, in- are resistant to antiestrogen therapy or who have
ducing both LH and FSH secretion) resulted in become ovulatory with antiestrogen but have not
normalization of serum inhibin B, increased testicular conceived. Recent meta-analyses and guidelines con-
volume, proliferation of Sertoli cells, and spermato- firm the value of gonadotropins as the appropriate
gonia (at biopsy) and the presence of sperm in the second-line therapy in women with PCOS wishing
ejaculate in all men. to become pregnant (–), and whereas it may
In adolescent boys with delayed puberty as a result offer a more rapid time to pregnancy than with clo-
of HH, the standard approach of using testosterone mifene (), the complexity (and expense) of treat-
administration results in growth, virilization, and ment generally precludes this. With this approach,
psychosexual maturation without testicular develop- mono-ovulation can be induced in over % of cycles,
ment or induction of fertility. Therefore, there is a with a live birth rate of ~% per cycle and a cu-
possibility that subsequent fertility may be improved if mulative live birth rate of ~%. This can be achieved
gonadotropins are instead used for this purpose. A with a very low multiple live-birth rate of .% but only
recent multicenter study of  patients addressed this. by application of rigorous cancellation criteria in the
The administration of hCG with recombinant FSH event of a multifollicular response ().
in boys and adolescents with HH resulted in the In severely hypogonadotropic women, prepara-
presence of sperm in the ejaculate in over % of tions containing LH activity must be used. In such
subjects, whether they had previously been treated patients, induction of ovulation is more difficult

doi: 10.1210/er.2018-00052 https://academic.oup.com/edrv 919


REVIEW

(with a higher cycle cancellation rate reflecting a higher The initial approach to more rapid-onset FSH
multifollicular response), and a lower pregnancy rate is administration involved luteolysis with the use of a
achieved. Therefore, such women are ideally treated GnRH antagonist (), but it subsequently became
with pulsatile GnRH (whereby the involvement of clear that FSH administration could be started at al-
physiological feedback pathways contribute to high most any stage of the cycle, other than in the im-
rates of mono-ovulation), but this is of very limited mediate periovulatory period. This approach gives
availability. comparable oocyte number and apparent de-
velopmental competence to those obtained after
Superovulation routine ovarian stimulation, and whereas data on
The use of high doses of FSH for controlled super- pregnancy rate remain limited as a result of the nature

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/911/5047286 by Research 4 Life user on 22 November 2018


ovulation before assisted reproduction has now of the circumstances of these patients, they appear
resulted in several million live births around the world. acceptable (). The addition of the antiestrogen
There are several protocols described and well tamoxifen and more recently, the aromatase inhibitor
established, and dosage is often guided by measure- letrozole to such regimens in women with hormone-
ment of antral follicle count or of serum anti- dependent cancer to reduce estradiol production has
Müllerian hormone, which can identify women at risk also been used as a further level of refinement under
for excessive or low response, with dose adaptation or those specific circumstances (–), although this
additional counseling as appropriate (, ). This use is “off label.”
continues to be an evolving field reflecting limited Double stimulation is an alternative approach,
evidence comparing key aspects of treatment, such as wherein gonadotropin administration is restarted a
the optimal dose of FSH to optimize the response (, few days after a GnRH agonist trigger (). This
). It does, however, carry some risk, particularly of approach can substantially increase the number of
OHSS and of multiple pregnancy if more than oocytes obtained, and indeed, it may be the case that
one embryo is subsequently transferred (, ). more oocytes are obtained during the second phase of
The risk of OHSS has been dramatically reduced in stimulation than in the first, thus more than doubling
recent years by using GnRH antagonist-based the oocyte yield (). This approach can also po-
protocols to suppress endogenous gonadotropin tentially reduce the cancellation rate, particularly in
secretion rather than GnRH agonists that cause an women with a poor ovarian response or who are older,
initial “flare” in GnRHR activity (, ). Whereas and the limited data that are available indicate that the
GnRH antagonist protocols may be associated with a developmental potential of embryos derived from each
slightly reduced live-birth rate, possibly as a result of the two stimulations is comparable (, ).
of as-yet underoptimized luteal replacement, this
seems not to be the case in women with PCOS, and
it has been suggested as standard therapy for Development of Gonadotropin Analogs
that group (). GnRH antagonist cycles do not,
however, completely prevent the risk of the devel- Polypeptide gonadotropin analogs
opment of OHSS, and other established thera-
pies include cycle cancellation, the use of a GnRH Single-chain and dual-activity gonadotropins
agonist to trigger oocyte maturation, and a freeze-all As discussed, gonadotropin preparations (both en-
strategy, with the latter often referred to as “cycle dogenous and recombinant) are used therapeutically
segmentation” (). in a number of clinical settings. The duration of
The risks of multiple pregnancies have been re- treatment (in some cases, months) has necessitated the
duced in recent years by using elective single-embryo development of long-acting polypeptide analogs to
transfer (). Although this results in reduced live- mitigate the need for multiple injections for improved
birth rates per cycle, cumulative rates over multiple patient convenience. Corifollitropin alfa is one ex-
cycles are maintained (). ample of a long-acting FSH analog, although a major
Novel approaches to assisted reproduction using drawback is that the corifollitropin alfa is a hetero-
gonadotropins have, in large part, been stimulated by dimer. The stability of corifollitropin alfa and heter-
the need for protocols that do not require any delay odimeric urinary gonadotropins is an issue during
before their commencement, generally known as purification, shipping, and storage. In an attempt
random start protocols, and the double-stimulation to overcome these issues, Sugahara et al. ()
approach, which allows two stimulation cycles to be synthesized a single-chain hCG analog in which the
administered with only a very brief interval in between. hCGb-subunit was fused to the N-terminus of the
Both of these approaches are of particular benefit in gonadotropin a-subunit (hCGba; Table ). The in
patients newly diagnosed with cancer, where time is of vivo biopotency of this single-chain analog was shown
the essence, and normal endometrial development is to be greater than the heterodimer, as measured in a
irrelevant, as all oocytes or resulting embryos will be rat ovulatory assay (). The enhanced biological
cryopreserved. activity was suggested to be a result of improved

920 Anderson et al Peptide and Nonpeptide Gonadotropin Analogs Endocrine Reviews, December 2018, 39(6):911–937
REVIEW

pharmacokinetics, as the single-chain analog is not FSHb-CTP-CGb-a and FSHb-CTP-LHb-CTP-a


subject to subunit dissociation (). Interestingly, the were administered to sheep using the same experi-
single-chain analog with the a-subunit oriented at the mental design to test the single-chain analogs. Both
N-terminus of the b-subunit was secreted but inac- dual-active constructs elicited increases in serum es-
tive, with no receptor binding or activity measured in tradiol concentration,  to  days after administration,
vitro, whereas orientation of the b-subunit at the N- similar to that stimulated by dual injection of the two
terminus of the a-subunit resulted in improved se- single-chain gonadotropin constructs. In addition, an
cretion rates and permitted intramolecular interactions increase in ovarian weight (attributed to increased
similar to the native heterodimer, in addition to com- numbers of follicles) and the presence of corpus lutea
parable in vitro IC and EC values (). were observed (, ). The clearance rate of the dual-

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/911/5047286 by Research 4 Life user on 22 November 2018


These findings with single-chain hCG led to the active construct FSHb-CTP-LHb-CTP-a was also
development of single-chain analogs of LH and FSH significantly improved compared with endogenous
[(LHba, LHbDa (in which the terminal  aa of LHb ovine gonadotropin ( to  hours) (, ).
were removed), LHbD-CTP-a, LHb-CTP-a, and Despite these promising indications, to date, single-
FSHb-CTP-a; Table ], which had comparable or chain or dual-active gonadotropin analogs remain
improved in vitro secretion and biological activity untested in humans.
comparable with the native heterodimers (, , ).
Again, these single-chain analogs were engineered Differentially glycosylated gonadotropin analogs
with the b-subunits oriented at the N-terminus of the Another strategy used to produce gonadotropins with
a-subunit, using the hCGb CTP sequence as a linker. altered signaling/pharmacokinetic properties was the
The CTP contains multiple proline and serine residues production of differentially glycosylated analogs. The
and is predicted to lack substantial secondary struc- addition of an N-terminal extension sequence to in-
ture, thereby facilitating folding and native inter- troduce two additional N-linked glycosylation sites
subunit heterodimer-like interactions. Additionally, (ANITVNITV) to the glycoprotein a-subunit of FSH
the O-linked glycosylation sites of the CTP reduced was found to increase its half-life, three- to fourfold
hepatic clearance rates, contributing to increased serum compared with FSH in a rat model (). This analog
half-life and increased biopotency of the single-chain (N-a FSH; Table ) exhibited lower in vitro–specific “To date, single-chain or dual-
analogs. The pharmacokinetic and pharmacodynamic activity than FSH but had higher in vivo potency active gonadotropin analogues
profiles of FSHb-CTP-a have been determined in (measured by ovarian weight and increased estrogen remain untested in humans.”
Rhesus monkeys by SC injection or IV bolus ( IU/kg). production in rats), likely as a result of its increased
The absorption half-life of FSHb-CTP-a was three- half-life. A single dose was as effective as daily doses of
fold slower than recombinant FSH following SC in- the same concentration of FSH ().
jection, and the elimination half-life was increased Single-chain analogs, which have substitution of
twofold or fourfold over recombinant FSH when ad- the CTP linker with a synthetic linker containing two
ministered SC or IV, respectively (). The same study N-linked carbohydrate signal sequences (N linker),
also demonstrated that a single SC dose of the single- have also been demonstrated to increase serum half-
chain analog could stimulate estrogen production for life in rats, with comparable in vitro and in vivo po-
 to  days (). tencies to those of analogs containing the CTP. A
In vivo biopotency of the single-chain analogs of single-chain FSH construct with this substitution
FSHb-CTP-a and hCGba were also tested in sheep (FSHb-N-a; Table ) had comparable activity in vitro
passively immunized against GnRH, to suppress en- to recombinant FSH. Its half-life was also similar to
dogenous gonadotropin secretion. Concurrent ad- FSHb-CTP-a and approximately twofold greater than
ministration of the single-chain analogs ( IU/kg IV), recombinant FSH in immature female rats after a
followed by  IU hCG IV after  days to induce single IV injection. Rats administered with FSHb-N-
ovulation, resulted in elevated serum progesterone and a also had higher mean ovarian weights,  days after
the presence of corpus lutea, highly suggestive of administration, compared with those administered
successful ovulation. Neither analog promoted estra- with recombinant FSH ().
diol production in isolation but dual administration A linker with N-linked glycosylation sites is ad-
resulted in a robust estradiol increase,  days post- vantageous, as in contrast to O-linked sites (such as
injection (), therefore confirming that the single- those of the CTP), there is a defined N-linked gly-
chain analogs have in vivo biological activity. This cosylation consensus sequence that can be exploited to
study, demonstrating that the analogs could be ad- generate differentially glycosylated hormone variants
ministered in combination to induce estradiol pro- (). Analogs with varying numbers of N-linked
duction and folliculogenesis, led to the development glycosylation sites (FSHb-N-a and FSHb-N-a)
of dual-active, single-chain gonadotropin constructs, between the subunits (Table ) have been pro-
including FSHb-CTP-CGb-a, FSHb-CTP-LHb- duced to determine effects on biological half-life and
CTP-a, and CGb-FSHb-CTP-a (Table ), with both biopotency. Whereas elimination half-life in female
FSHR and LHR activities (, , ). The constructs rats was comparable between analogs containing the

doi: 10.1210/er.2018-00052 https://academic.oup.com/edrv 921


REVIEW

N linker (FSHb-N-a) or CTP (FSHb-CTP-a) also had greater bioavailability compared with the
linkers, there was only a modest improvement in non-PEGylated control in rats ().
clearance rates for the FSHb-N-a analog (). In- Another strategy for increasing FSH half-life was
terestingly, despite a plateau in elimination half-life the creation of a fusion-protein consisting of FSH a-
and comparable augmentation of ovarian weight and b-subunits fused to a binding partner (e.g., im-
following injection of FSHb-CTP-a, FSHb-N-a, and munoglobulin Fc fragments) of the neonatal Fc
FSHb-N-a analogs, there were increases in the receptor (FcRn). FSH fusion proteins containing
number of antral follicles and substantial increases in immunoglobulin Fc fragments had increased stability
inhibin A following treatment with all the N-linked and serum half-life. In addition, these fusion proteins
analogs (). The in vivo bioactivities of FSHb-N-a are protected from endosomal degradation by binding

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/911/5047286 by Research 4 Life user on 22 November 2018


and FSHb-N-a have since been examined further, to the FcRn, facilitating recycling (–). When
with -week-old female mice given a single IP injection female rats were injected SC with  nmol/kg
of  IU of the N-linked analogs, recombinant FSH, or recombinant FSH or a heterodimeric FSH molecule
pregnant mare’s serum gonadotropin (PMSG; which conjugated to an immunoglobulin Fc fragment (FSH-
has activity at both FSHR and LHR in nonequine Fc; Table ), ovarian weight was increased twofold in
species). The bioactivities of FSHb-N-a and FSHb- the FSH-Fc-treated rats when compared with the
N-a, measured by ovarian and uterine weights, in- recombinant FSH-treated group ().
hibin A production, and a number of antral follicles, Modification of hormones by conjugation to en-
superseded the recombinant FSH responses. PMSG dogenous molecules, such as FcRn binding partners,
performed best in all measured parameters as a result may result in advantageous properties, in addition to
of its activities at both LHR and FSHR. However, the increased half-life, such as oral/pulmonary bio-
supplementation of the FSHb-N-a and FSHb-N-a availability. For example, fusion proteins comprising
injections with daily injections of . IU hCG resulted FcRn binding partners can be shuttled across epithelial
in responses comparable with that of PMSG (). In barriers, opening the intriguing possibility of creating a
another mouse study,  IU IP of FSHb-N-a, FSHb- gonadotropin analog that can be administered via
CTP-a, or PMSG was administered, followed by  IU nasal or pulmonary routes using aerosols. This has
hCG IP,  hours later. Increased ovulation and been demonstrated in the cynomolgus monkey, with a
embryo maturation were observed with FSHb-N-a single pulmonary dose of heterodimeric FSH-Fc (
when compared with FSHb-CTP-a (. 6 . mg/kg), eliciting a -fold increase in serum inhibin B
embryos vs  6  embryos, respectively) (). The levels (). Conjugation of peptidic hormones and
delivery rates and litter sizes were comparable between large macromolecules to vitamin B has also been
PMSG and FSHb-N-a and were higher or compa- shown to confer oral bioavailability. Vitamin B
rable with recombinant FSH treatment (). interacts with an intrinsic factor, which in turn, binds
Again, although promising in vivo results have to its cognate receptor located on the terminal ileum of
been obtained for these differentially glycosylated the small intestine and is internalized by endocytosis.
analogs, they have yet to be developed further for Vitamin B conjugation to macromolecules, such as
clinical use. gonadotropins with poor oral bioavailability, may
therefore improve uptake via oral administration.
Gonadotropin conjugates and suspensions with Whereas this method has not yet been applied to
increased half-life and bioavailability gonadotropins, a D-lys GnRH analog has been suc-
Alternative methods to try to improve gonadotropin/ cessfully crosslinked to monocarboxyl-vitamin B,
glycoprotein hormone pharmacokinetic properties and this conjugate has been shown to retain bioactivity
have included attachment of synthetic polymers to when administered orally (PO) in a mouse ovulation
increase half-life and bioavailability and therefore, assay ().
biological activity. Covalent addition of polyethylene Another approach to increase hormone half-life
glycol (PEG; PEGylation) results in many advanta- was the development of sustained-release formula-
geous properties, such as increased solubility of hy- tions. Single injections of porcine FSH dissolved in an
drophobic molecules, increased size (reduced renal aluminum hydroxide gel suspension were used to
clearance), reduced immunogenicity, and protection stimulate folliculogenesis in both rabbits and cows
from proteolytic degradation. Injection of Sprague- (, ). The number of oocytes recovered and the
Dawley rats with . mg/kg IM of a recombinant number fertilized following a single injection of the
PEGylated TSH analog resulted in a -fold increase in aluminum hydroxide adjuvant were not statistically
plasma half-life and elevated thyroxine (T) levels for different than those treated with daily injections of
up to  hours, with only moderate effects on receptor porcine FSH dissolved in saline, and measurement of
binding affinity (). FSH has also been PEGylated serum FSH concentrations indicated that the levels of
via covalent amide linkage (Table ), and this analog hormone were sustained throughout the treatment
retained FSH activity, as measured by progesterone period, confirming its sustained release (, ). In
secretion of cultured bovine cumulus cells (), and rabbits, the aluminum hydroxide gel preparation was

922 Anderson et al Peptide and Nonpeptide Gonadotropin Analogs Endocrine Reviews, December 2018, 39(6):911–937
REVIEW

compared with one of FSH dissolved in an alternative often result in incomplete deglycosylation, which may
adjuvant, polyvinylpyrrolidone, but superovulation confound the interpretation of data ().
was found to be much more effective when FSH was
administered in the aluminum hydroxide preparation
(). Nonpeptidic Low-Molecular-Weight
Whereas these modified analogs with improved Gonadotropin Receptor Agonists
bioactivity/bioavailability have not been tested clini- and Antagonists
cally, they do offer an exciting possibility for possible
future development of improved or less invasive go- Whereas the development of polypeptide gonado-
nadotropin therapeutics. tropin analogs with increased serum half-life/bioactivity

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/911/5047286 by Research 4 Life user on 22 November 2018


(described earlier) shows promise of single injections to
Polypeptide-gonadotropin antagonists replace the multiple injections of the natural hormones
In addition to the development of polypeptide-based used in ART, small-molecule/low-molecular-weight
gonadotropin preparations/analogs for activation of (LMW) orally active agonists would avoid trauma of
the gonadotropin receptors, a number of strategies injection and overcome problems of preparation uni-
have also been used in an attempt to generate formity (arising from posttranslational modification
polypeptide-based gonadotropin antagonists for po- of peptide-based analogs) and have greater com-
tential application in contraception. Fusion of two pound stability (). However, despite several drug-
hCG b-subunits as a single chain (hCGbb; Table ) development programs focused on identification of
resulted in an analog able to bind to the LHR (albeit LMW gonadotropin analogs, only three have entered
with a binding affinity lower than hCG;  and  nM, human trials, and none are commercially available.
respectively), which exhibited a dose-dependent in- Desirable properties of such LMW analogs would
hibition of hCG actions (measured by decreased se- include good oral bioavailability, high potency, high
rum testosterone concentration) in a rat model (). specificity, and high lipophilicity (which would enable
As a result of the importance of glycosylation for interaction with allosteric sites within the TM domains
gonadotropin function, it has also been proposed that of the target receptors; see later). “Small molecule/LMW orally
deglycosylated gonadotropins, which retain receptor In addition to application in ART, orally bioavailable, active agonists would avoid
binding activity but with reduced bioactivity (see the small-molecule antagonists at gonadotropin receptors trauma of injection and
“Gonadotropin hormone structure and function” may have clinical application as novel oral contraceptives overcome problems of
section), could also act as competitive antagonists with with potentially greater specificity and therefore, reduced preparation uniformity.”
therapeutic potential (, , , ). Deglycosyla- risk of side-effects, such as headaches, weight gain,
tion can disrupt dimerization of the gonadotropin nausea, and increased risk of serious complications, such
subunits, but this can be overcome by the addition of as venous thromboembolism, which occurs with current
specific dimerization sequences () or by use of steroidal contraceptive drugs ().
single-chain hormones in which the two subunits are LMW LHR agonists may also be used potentially
covalently joined (see the “Single-chain and dual- for women at risk for OHSS. This syndrome has been
activity gonadotropins” section). Indeed, treatment linked to fertility treatments using hCG injections to
with  IU of a single-chain version of partially achieve oocyte maturation and ovulation. hCG has a
deglycosylated hCG (Table ) has been shown to long serum half-life; therefore, LMW LHR agonists
reduce steroid hormone production and ovulation in with shorter half-lives may well circumvent/attenuate
rats pretreated with  IU PMSG when compared with the risk of OHSS. In addition, orally active LMW
stimulation with  IU hCG (). In addition, factors compounds are more amenable to titrating dosage.
associated with OHSS, such as vascular permeability Despite the theoretical, therapeutic potential of
and expression of vascular endothelial growth factor, LMW modulators of the gonadotropin receptors, a
were reduced in these animals, suggesting that this number of persistent problems have hindered their
analog could ameliorate the effects of prolonged ex- development into clinically applicable drugs. Firstly,
posure to hCG after ovulation has been triggered in the high degree of sequence conservation across the
patients with a high risk of OHSS (). However, there glycoprotein hormone receptor family and other re-
are conflicting opinions regarding the antagonistic lated receptors, such as the LRR containing GPCRs,
properties of the deglycosylated hormone, with one means that drug crossreactivity can be an obstacle.
study showing no decrease in progesterone levels when Indeed, this has been an issue with some LMW an-
the deglycosylated hormone was administered to alogs. However, as the natural glycoproteins are highly
healthy females in the midluteal phase, although the specific, there is no reason to believe that this cannot
authors acknowledged that this may be a result of the be achieved with the LMW molecules. Secondly, the
presence of “spare receptors” and the inability to glycoprotein hormones are large heterodimeric pro-
achieve sufficient serum concentrations of the an- teins that interact with their cognate receptors via
tagonist (). In addition, chemical methods of multiple structurally conserved contact sites and ad-
deglycosylation, such as hydrogen fluoride treatment, ditionally, have a two-step binding and activation

doi: 10.1210/er.2018-00052 https://academic.oup.com/edrv 923


REVIEW

mechanism (Fig. ). Therefore, it is difficult to imagine gonadotropin receptors, and a number of cell-based
orthosterically binding LMW molecules that could screening assays have been used over the last  years,
simultaneously occupy and induce the structural resulting in the identification of various lead
changes within the glycoprotein hormone receptor compounds/chemical scaffolds, with specific in vitro
ECDs to attain full efficacy of activation with low cellular and in vivo physiological activity profiles.
potency. However, this concern has been overcome The most frequently targeted of the glycoprotein
with allosterically binding LMW molecules (see later). hormone receptors for LMW drug discovery is the
Thirdly, promising lead compounds, identified through FSHR. This is perhaps unsurprising, given the multiple
in vitro screening, have been found to have little/no injections of hormone necessary for ovarian hyper-
in vivo efficacy (), as is a common feature of small- stimulation in assisted reproductive therapy protocols.

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/911/5047286 by Research 4 Life user on 22 November 2018


molecule development in the pharmaceutical industry. However, although a number of promising LMW
GPCRs are amphipathic molecules, complicating compounds have been developed for both gonado-
crystallization/structural analysis, but exciting ad- tropin receptors, none are yet in clinical use. The
vances in X-ray crystallography methodologies, in- following sections will describe the development of
cluding the use of thermodynamically stabilizing some of the most promising LMW compound classes
mutations and nanobody-based stabilization of GPCR that have been identified to date. For more detailed
active states, have revealed new opportunities for novel insight regarding the chemical properties of many of
drug design (, ). One such avenue is that of these different compound classes, the reader is referred
allosteric modulators of GPCR activity that interact to van Straten and Timmers ().
with the receptor at a site distinct from the orthosteric
ligand binding site (site of binding of the endogenous FSHR agonists
ligand). Allosteric GPCR modulators generally fall into
three classes: allosteric agonists and positive and Piperidine carboxyamides
negative allosteric modulators (PAMs and NAMs, In , Serono (now Merck Serono) used a luciferase
respectively). Allosteric agonists have agonist activity reporter gene high-throughput screen to identify pi-
in the absence of the orthosteric ligand, whereas PAMs peridine carboxyamide derivatives with activity in
and NAMs are defined as molecules that either Chinese hamster ovary (CHO) cells stably expressing
potentiate or attenuate agonist-mediated responses, FSHR. A lead compound with an EC of . nM was
respectively. This can be through modulation of identified and was subsequently shown to have poor
orthosteric agonist binding affinity or the ability of the stimulation of estradiol in rat granulosa cells (EC .
agonist-occupied receptor to interact with intracellu- mM). However, this initial finding was valuable in the
lar signal transducers. The targeting of allosteric sites discovery of a number of chemical scaffolds with
can be advantageous, as they are generally less evolu- improved FSHR agonistic activity ().
tionarily conserved and often contain unique structural
features, providing a platform for the development of Thiazolidinones
drugs with a greater degree of target specificity (). Thiazolidinone (TZD) scaffolds have been used pre-
Additionally, many allosteric modulators only have viously in a number of drug-discovery programs and
activity in the presence of the cobound endogenous have excellent drug-like properties (–). Indeed,
ligand, with the advantage that GPCR activation is low-potency FSHR agonists have been described that
limited to the spatiotemporal release of the endogenous contain thiazole core structures (). In , Affy-
ligand (). This is especially relevant in the context of max screened ., compounds from a large TZD
many neuroendocrine signaling axes, which have cy- library for agonist activity at the FSHR using a lu-
clical patterns of hormone release. ciferase reporter gene assay in CHO cells expressing
Allosteric binding sites for LMW compounds have FSHR. The design of this library was based on data that
now been described for a number of GPCRs. Through had identified a TZD compound with moderate ag-
the modeling of the gonadotropin hormone recep- onist activity at the FSHR ( to  mM). Several hits
tor TM domains, using adenosine A receptor and with increased potency were identified from this
rhodopsin receptor molecular models, alongside a screen, including “compound ,” which was a partial
number of biochemical assays, and studies using agonist (% relative to recombinant FSH stimu-
chimeric receptors, it has been proposed that the lation), with an EC of  nM (). Compound
gonadotropin receptors have two allosteric sites: one development addressed synthesis issues regarding
major and one minor, labeled P and P, respectively stereoselectivity on the heterocyclic ring (the less active
() (Fig. ). These sites are located toward the ECL trans isomer predominates over the desired cis isomer
half of the TM helices, close to the transition of TM- during synthesis) and resulted in stable TZD com-
to-ECL domains, with P located among TMs III, IV, pounds containing g-lactam congeners and -alkyl
V, and VI and P among TMs I, II, III, and VII (). substituents with comparable potencies and efficacies
A substantial number of structural scaffolds have to the original lead compound (, ). Another
been described that can allosterically bind to the screen by Affymax and Wyeth (since acquired by

924 Anderson et al Peptide and Nonpeptide Gonadotropin Analogs Endocrine Reviews, December 2018, 39(6):911–937
REVIEW

Pfizer) of a collection of combinatorial libraries con- Hexahydroquinolines


taining over , compounds and represent- Two patents filed in  by Organon detail the use
ing . core scaffolds identified two additional FSHR of hexahydroquinoline scaffolds (-phenyl--oxo-
agonists with TZD scaffolds (with potencies of ~ mM) l,,,,,-hexahydroquinolines) as FSHR activators
(). Parallel synthesis resulted in three compounds in vitro (, ). These patents describe several
with nanomolar potency. Subsequent studies using LMW compounds with potencies in the low nano-
chimeric receptors in which the N-terminal domain molar (, nM) range in an in vitro luciferase re-
and TM domains of the TSH receptor (TSHR) and porter assay. In , the same company published a
FSHR were interchanged indicated that these analogs description of a hexahydroquinoline LMW PAM of
act via an interaction with the TM domains of the the FSHR, Org - (Table ). This molecule had

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/911/5047286 by Research 4 Life user on 22 November 2018


FSHR (unlike the natural hormone that interacts with agonist activity in a luciferase reporter assay and was
the ECD). Further analysis using chimeric receptors demonstrated to bind via a site distinct from the en-
narrowed down the site of interaction to the area of the dogenous hormone. In addition to the allosteric agonist
receptor between TM I and ECL (). One of the activity, coincubation with FSH demonstrated Org
identified compounds, “compound ” (Table ), was - PAM activity, inducing a substantial (sixfold)
demonstrated to be a full agonist of the FSHR and increase in FSH affinity, which translated to a fourfold
could induce progesterone production in mouse ad- increase in FSH potency in the luciferase reporter assay
renal Y cells transfected with FSHR and estradiol (). Org - was also demonstrated to induce
production in primary rat granulosa cells (), had follicular growth in a rat ovulation assay when admin-
in vivo activity in a mouse cumulus expansion assay, istered PO and thus, represented an orally active FSHR
and demonstrated a dose-dependent increase in the LMW agonist with in vivo efficacy (). MSD has also
number of ovulated oocytes in a rat ovulation assay. investigated similar compound scaffolds, including MK-
Although this compound had promising FSHR ac-  (a dihydropyrroloisoquinoline) as LMW allosteric
tivity, unfortunately, it had poor oral bioavailability, FSHR agonists. However, when MK- was tested
and micronucleus screening assays identified geno- in a dose-escalation study in healthy human females,
toxic effects (–). it was unable to confirm effects on follicular develop-
Wyeth/Affymax further characterized the pharma- ment and found that it unexpectedly affected thyroid
cological properties of several TZD compounds and function (), suggestive of activity at the TSHR, which
demonstrated that small modifications to the TZD ring
can result in completely different pharmacologies,
resulting in anything from agonists through to negative
modulators able to inhibit estradiol production in rat
granulosa cells via activation of Gai () (Table ).
Thus, a single TZD core structure has the potential to be
used to develop a spectrum of LMW allosteric mod-
ulators targeting multiple FSHR signaling pathways.

Diketopiperazines
A high-throughput screen in  by Pharmacopeia
(since acquired by Ligand Pharmaceuticals) and Or-
ganon [since acquired by Schering-Plough and then
merged with Merck & Co./Merck Sharpe & Dohme
(MSD)] of approximately two million compounds,
identified a number of biaryl agonists of the
FSHR with different substituents on the phenyl rings.
The most potent compounds contained heterocyclic
diketopiperazine substituents (). Lead optimiza-
tion, aimed at modification of the diketopiperazine
core side-chains, increased their potency from the low
micromolar to the low nanomolar range in both lu- Figure 2. Top view of the LHCGR, with a partially clipped
ciferase reporter gene and cAMP assays (). How- backbone of the ECLs, visualizing the potential binding region
ever, these compounds do not appear to have been for small allosteric LMW ligands (magenta). This ligand-binding
developed further, and there is no information re- region can be subdivided further into a major and a minor
pocket. TMH, TM helix. Reproduced with permission from
garding specificity or in vivo activity of these hit
Heitman LH, Kleinau G, Brussee J, et al. Determination of different
compounds. At a similar time, Serono also filed a putative allosteric binding pockets at the lutropin receptor by
patent describing substituted piperazine compounds using diverse drug-like low molecular weight ligands. Mol Cell
with FSHR agonistic activity in the low micromolar to Endocrinol 2012;351(2):326–336. [© 2018 Illustration Presentation
low nanomolar range (). ENDOCRINE SOCIETY]

doi: 10.1210/er.2018-00052 https://academic.oup.com/edrv 925


REVIEW

Table 2. Chimeric and Single-Chain Gonadotropin Analogs

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/911/5047286 by Research 4 Life user on 22 November 2018


emphasizes the importance of good selectivity margins inhibition of ovulation resulted from the formation of
for these compounds. luteinized, unruptured follicles, and this effect was re-
versible, with spontaneous ovulation recovery following
Other agonists discontinuation of treatment (). These results could
MSD has also described two LMW FSHR agonists with not be fully replicated in mono-ovulatory species (cyn-
undisclosed scaffolds and interesting pharmacological omolgus monkey) (), but this still represents an
profiles. These agonists have nanomolar activity at the approach to the development of a reversible female
FSHR ( to  nM) and display similar responses to contraceptive.
recombinant FSH in a luciferase reporter assay ().
These LMW FSHR agonists were unique in that they had FSHR antagonists
particularly short-acting profiles (half-life . to . hours,
time to reach maximum serum concentration . to . Sulfonic acid-containing compounds
hour). Intriguingly these compounds resulted in % The sulfonic acid-containing compound, suramin, has
suppression of ovulation in cycling Orga rats at doses of been used as an IV antiparasitic for over  years
 mg/kg PO when administered twice daily. The (). In addition to its antiparasitic properties,

926 Anderson et al Peptide and Nonpeptide Gonadotropin Analogs Endocrine Reviews, December 2018, 39(6):911–937
Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/911/5047286 by Research 4 Life user on 22 November 2018
REVIEW

927
https://academic.oup.com/edrv
Table 3. Selected LMW Gonadotropin Analog Structures

doi: 10.1210/er.2018-00052
REVIEW

suramin has been demonstrated to inhibit the sig- Aminoalkylamides


naling activity of a number of peptide hormones, A substituted aminoalkylamide series of FSHR an-
including platelet-derived growth factor, epidermal tagonists has also been reported with a spectrum of
growth factor (), and FSH (, ). Subsequently, potencies. Two of the most potent compounds,
suramin has been shown to compete with FSH for identified in a cAMP screening assay by Ortho-McNeil
binding to the FSHR-ECD (). Interestingly, a Pharmaceutical, were tested for ovulation and
second mechanism of inhibition may involve dis- spermatogenic inhibitory effects in Wistar rats, but
ruption of certain GPCR-G protein interactions (, following oral dosing at  mg/kg, only very limited
). In a cohort of male patients receiving suramin effects were noted (). In the absence of any
as a therapy for refractory cancer, all showed a marked pharmacokinetic data, it is unclear whether the limited

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/911/5047286 by Research 4 Life user on 22 November 2018


decrease in plasma testosterone, and this effect effects are a result of restricted oral bioavailability/poor
was mirrored in a rat Leydig cell assay (). The intestinal absorption/rapid clearance, as the two
dramatic in vivo effect on testosterone secretion, compounds had activity in in vitro cAMP and gran-
combined with lack of specificity (including docu- ulosa cell assays.
mented LHR interaction) (, ), precipitated
the development of other LMW FSHR antagonists Tetrahydroquinolines
containing sulfonic acid moieties (, ). Com- Organon identified a tetrahydroquinoline (THQ)
pound screening by Wyeth identified three (bis) scaffold with FSHR agonistic activity in a luciferase
sulfonic acid, (bis)benzamide FSHR antagonists with assay screen, with a moderate potency of . mM and
moderate potencies of cAMP inhibition, estradiol an efficacy of % relative to FSH. This compound was
inhibition (granulosa cell assay), and progesterone selected for lead optimization, and surprisingly, it was
inhibition (in an adrenal Y cell assay) and IC discovered that introduction of an aromatic group at
values in the low micromolar range (). Unlike position  of the THQ scaffold resulted in a switch
suramin, these compounds displayed marked im- from full agonist to full antagonist, with nanomolar
provement in specificity, with no LHR binding ac- IC values (compound ; Table ) (). Biphenyl
tivity measured up to  mM and no inhibition of moieties were also tolerated, implying that the binding
TSHR cAMP response up to  mM (). Also in pocket was large and lipophilic, with competition
, a napthalene sulfonic acid compound (“com- binding assays determining that the pocket is likely to
pound ”; Table ) was identified by Wyeth through be located in the TM domain (). In a mouse ex
library screening for compounds that inhibited FSH vivo follicular growth assay, one THQ compound
binding to FSHR. The lead compound inhibited FSH with a biphenyl substituent was demonstrated to in-
binding with an IC of . mM, and interestingly, hibit follicular growth in the presence of FSH and
although it bound the FSHR ECD, the inhibition of inhibited ovulation in % of follicles ().
FSH binding was noncompetitive and led to a re-
duction in FSH binding sites but not affinity (). Benzamide derivatives
This compound was also able to inhibit the FSH An homogenous time-resolved fluorescence screening
cAMP response in a dose-dependent manner with assay was used by Addex Pharmaceuticals to identify
% efficacy, inhibited estradiol production in FSHR NAMs and identified a benzamide compound
granulosa cells, and when administered at  mg/kg (ADX; Table ) with NAM activity (IC . mM
IP to cycling female rats, inhibited ovulation in % with % inhibition of FSH EC). This NAM was
of treated animals (). Analogs were developed ascertained to be a biased antagonist of the FSHR, as in
to examine structure-activity relationships, and a the presence of FSH, it inhibited cAMP and pro-
number of substitutions were identified as critical for gesterone production in vitro in a dose-dependent
activity, including the sulfonic acid moiety (). manner but failed to inhibit estradiol production
Issues regarding off-target effects, safety, and oral and at doses . mM, stimulated estradiol production
bioavailability have hampered the development of (). This finding was surprising and challenges the
these compounds for clinical application. In the in vivo paradigm that estradiol production is entirely de-
rat ovulation model, napthalene sulfonic acid induced pendent on cAMP signaling. In an in vivo setting, SC
mild inflammation of the ovarian surface epithelium, administration of ADX at  mg/kg resulted in
and the rats displayed a substantial retardation in no differences in number of oocytes recovered and
growth rate (). It is unclear whether these effects ovarian weight of rats treated with  mg pituitary FSH
may have indirectly contributed to ovulation in- and ovulated with  IU hCG (). The authors of
hibition. Furthermore, the oral absorption of the this earlier article postulate that this may be a result of
(bis)sulfonic acid, (bis)benzamide compounds was the inability of ADX to suppress estradiol
predicted to be poor, and the conversion of the sul- production. However, women with PCOS have suc-
fonic acid moieties to carboxylic acid, although pre- cessful pregnancies despite suppression of estradiol
dicted to improve absorption, abolished FSHR binding with letrozole or tamoxifen, so suppression of estra-
activity (). diol may be necessary but not sufficient. Two additional

928 Anderson et al Peptide and Nonpeptide Gonadotropin Analogs Endocrine Reviews, December 2018, 39(6):911–937
REVIEW

benzamide analogs were subsequently identified by (Table ), which displayed nanomolar potency at the
Addex, corroborating the description of biased antag- LHR (EC of . nM; % efficacy compared with
onism identified for ADX and further sub- LH), no activity at the TSHR, and reduced activity at
stantiating the requirement for blockade of both arms of the FSHR (-fold reduced potency than at the LHR)
the FSHR steroidogenic signaling pathway to inhibit (). Interestingly, this study also highlighted biased
follicular maturation and ovulation in rats (). signaling properties of Org . Whereas LH ac-
tivation of the LHR stimulated both cAMP (EC 
Other FSHR antagonists pM) and phospholipase C (PLC; EC . nM), no
Pyrrolobenzodiazepine and acyltryptophanol LMW substantial activation of the PLC pathway by Org
FSHR antagonists were identified through luciferase  was measured, suggestive of biased agonism

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/911/5047286 by Research 4 Life user on 22 November 2018


and homogenous time-resolved fluorescence screen- (). In addition, whereas Org  did not antag-
ing assays, respectively, with IC concentrations in the onize LH-induced intracellular cAMP accumulation,
micro- and nanomolar ranges (, ). it did strongly antagonize (% maximal inhibition) the
LH-induced activation of PLC in a noncompetitive
LHR agonists manner, suggestive of an allosteric binding mechanism
A number of LHR agonists have been developed by (). In support of this, Org  was shown to
the pharmaceutical industry. reduce noncompetitively [I]hCG binding ().
Subsequent studies have both confirmed and re-
Thienopyrimidines pudiated the described increase in [I]hCG dis-
Organon reported an orally active, nonpeptidic LMW sociation (, ), but nonetheless, the observed
agonist for the LHR. In a high-throughput screen of a inability of the LMW compounds to compete with
compound library, using a luciferase reporter gene hormone binding supports the theory that these
assay, a thieno[,-d]pyrimidine was identified as compounds are acting in an allosteric manner.
having agonist activity at the LHR (). A series of The biological activity of Org  has since been
thienopyrimidine and thienopyridine analogs were investigated more thoroughly. In an ex vivo ovulation-
subsequently synthesized and their in vitro activity induction assay using cultured mouse follicles,  mM “Issues regarding off-target
determined. The most potent LHR agonist identified Org  was able to stimulate maximal levels of effects, safety, and oral
was the thienopyrimidine, Org  (EC of  nM) ovulation and progesterone production, similar to that bioavailability have hampered
(), which was found to stimulate testosterone seen using hCG, and was able to induce testosterone the development of these
production in cultured mouse Leydig cells (EC of production in primary Leydig cell cultures with high compounds.”
. mM) and was able to stimulate ovulation in % potency, although with lower efficacy than hCG ().
of urinary FSH-primed female mice, using a dose of In vivo, Org  has been shown to have good oral
 mg/kg PO (). Org  has since been shown availability (% in rat and % in dog) with a short
to have partial agonist activity at the TSHR but with half-life (. hours in rat and . hours in dog) ().
~-fold lower potency and no substantial activity at The occurrence of OHSS has been associated with the
the FSHR (, ). With the use of computer-based long half-life of hCG (half-life ~ to  hours
docking, a binding pocket was delineated among TMs depending on source and purification methodology)
III, IV, V, VI, and VII and ECL, with the cleft formed (); therefore, the short half-life of Org  may
in the LHR larger than that of TSHR (, ). This have appreciable advantages. Indeed, when Org 
pocket was verified through mutagenesis of receptor was administered to PMSG-primed female rats, even
residues predicted to be of importance for Org  at high concentrations ( mg/kg), there was no
interaction and by synthesis of analogs hypothesized to evidence of an increase in vascular permeability or
increase specificity or ablate binding. Of particular vascular endothelial growth factor levels, unlike that
importance is a glutamate residue in TMIII that is seen with hCG or LH ().
conserved between LHR and TSHR and is predicted to In GnRH antagonist-treated rats, a single -mg/kg
form a hydrogen bond with the amino group of Org dose of Org  PO resulted in ovulation of a
 (, ). Mutation of this residue completely normal number of ova, comparable with the hCG
ablated Org -mediated TSHR signaling. Fur- control ( IU/kg SC). Ova fertilization rate, im-
thermore, an analog of Org  with addition of plantation rate, and subsequent fetal development
“steric bulk” to the amide (an extra methyl group) was were all within normal parameters. In male rats at
sufficient to block Org -mediated TSHR sig- concentrations $ mg/kg PO, similar levels of tes-
naling but not LHR signaling, resulting in a substantial tosterone stimulation were measured to those pro-
improvement in specificity. This steric bulk was hy- duced after treatment with  IU hCG SC ().
pothesized to inhibit insertion into the smaller TSHR Perhaps most promisingly, Org  and another
binding cleft but still allow accommodation into the related thienopyrimidine, Org , have shown
slightly larger LHR pocket (, ). efficacy in humans. Both were shown to induce
Further lead optimization of Org  led to the ovulation when administered PO in a preclinical trial
development of another thienopyrimidine, Org  with healthy females. Org  was safe and well

doi: 10.1210/er.2018-00052 https://academic.oup.com/edrv 929


REVIEW

tolerated up to a maximum dose of  mg (). binding (, ). Several analogs were synthesized,
The GnRH analog, Ganirelix, was administered SC for including the potent allosteric inhibitor, compound 
pituitary suppression in conjunction with recombi- (LUF; Table ). In a luciferase reporter assay, this
nant FSH to stimulate follicular growth. Adminis- compound reduced the potency of LH or Org 
tration of  mg Org  resulted in ovulation stimulation of the receptor by approximately threefold
induction in .% of patients, as determined by ().
transvaginal ultrasound and a midluteal rise in pro-
gesterone (). Analogs with dual-receptor activity
Org  has activity at the FSHR, in addition to The high degree of sequence conservation among the
the LHR, and this lack of selectivity could be an issue glycoprotein hormone receptors frequently translates

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/911/5047286 by Research 4 Life user on 22 November 2018


for clinical use. However, it has been shown that ligand as a loss of selectivity during glycoprotein hormone
dimerization can aid in subtype selectivity for struc- receptor LMW compound development. One strategy
turally similar GPCRs. Dimeric compounds, in which used to circumvent this is the creation of bivalent
two Org  moieties were joined with flexible ligands combining combinations of LMW agonists
ethylene glycol linkers, or rigid benzene core spacers and antagonists via linkers to increase specificity of
have been synthesized and evaluated (). Both series responses (–). In a study that combined an Org
of compounds had increased LHR selectivity (two- to -based LMW LHR agonist (which also has FSHR
fivefold) compared with Org  alone in a lucif- activity) with a very selective LMW FSHR antagonist
erase reporter gene assay, either as a result of reduced based on the THQ compound  (Table ) using an
potency (rigid linkers) or reduced efficacy (flexible ethylene glycol spacer, a promising series of analogs
linkers) at the FSHR (). was developed, which exhibited activity at the LHR
(albeit with reduced potency) but no detectable FSHR
Pyrazoles activity (). The in vivo activities of these bivalent
In , screening of a compound library by a group at compounds have yet to be examined.
Merck Serono identified pyrazoles as another class of
LMW compounds with activity at the LHR (). From
the results of the initial screen, a focused pyrazole li- Conclusions and Future Perspectives
brary was developed, and the activity of compounds
within the library was assessed using a cAMP-specific Gonadotropins from natural and recombinant sources
scintillation proximity assay in CHO cells stably have been the mainstay of infertility treatment in men
expressing human LHR. A compound termed “com- and women for over eight decades and remain a
pound ” (Table ) displayed the best potency (EC crucial component of induction of ovulation for
of  nM) and efficacy (% of LH response) (). assisted reproduction, thus satisfying the needs of
Compound  was able to stimulate testosterone patients with infertility worldwide. Nevertheless, there
production in cultured rat Leydig cells and elicit a remain considerable challenges to develop less invasive
robust increase in serum testosterone in testosterone- approaches to minimize multiple injection protocols,
suppressed -month-old male rats treated with  mg/ reduce the risk of OHSS, and improve outcomes for
kg compound  IP (). No oral bioavailability or conditions, such as PCOS.
bioactivity of this compound has yet been reported. The development of an impressive suite of mol-
Interestingly, compound  was not able to compete ecules, including fused b-subunits, hybrid LH-FSH
with hCG in LHR radioligand binding assays, sug- molecules, differentially glycosylated gonadotropins,
gesting that it also interacts with the receptor at an polypeptide LH and FSH antagonists, longer-acting
allosteric site (). A structure-activity relationship analogs, and sustained release formulations has con-
study determined the allosteric pocket to be large and siderable potential, but their full development and use
lipophilic (similar to the THQ series), as polar groups have yet to be thoroughly explored. A contributing
were not tolerated as substituents of the pyrazole ring factor to this is the acquisition of the companies
nitrogen, providing further evidence of localization of developing these innovative drugs by large pharma
the allosteric site within the receptor TM domain (). and subsequent abandonment of the development
programs. Despite this huge endeavor and creativity
LHR antagonists in producing a wide range of novel polypeptide go-
nadotropin analogs, the only commercial products
Terphenyls arising from the gonadotropin analog developments
In an effort to identify other allosteric LHR modu- are recombinant gonadotropins and corifollitropin alfa.
lators, radiolabeled Org  was used to screen for Moreover, these recombinant FSH and LH prepara-
novel LMW ligands in a kinetic radioligand binding tions can still be subject to differential glycosylation with
assay. A number of hit compounds were identified that resulting variation in biological activity. Whereas the
increased the dissociation rate of [H]Org , recombinant gonadotropins have advantages of greater
indicative of allosteric modulation of [H]Org  batch-to-batch consistency and purity compared with

930 Anderson et al Peptide and Nonpeptide Gonadotropin Analogs Endocrine Reviews, December 2018, 39(6):911–937
REVIEW

hMG and hCG, the latter are still widely used, as they are compounds offer subtle advantages over traditional
less expensive and effective. agonist and antagonist ligands, such as improved
Good progress has been made in development of specificity and thus, reduced side-effects. Therefore,
LMW orally active LHR and FSHR agonists and development of PAMs/NAMs that target the go-
antagonists, and three have been evaluated in clinical nadotropin receptors would be an exciting develop-
studies. The allosteric LHR agonists Org  and ment in this field and is one currently being pursued
Org  displayed excellent bioavailability, safety, by companies, such as Addex Therapeutics.
and pharmacokinetic and pharmacodynamic prop- Another exciting development in the arena of
erties, and both demonstrated good efficacy in in- LMW gonadotropin analogs is their ability to restore
duction of ovulation protocols. Therefore, these function to inactivating mutations in human gonad-

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/911/5047286 by Research 4 Life user on 22 November 2018


compounds appear to fulfill the major criteria required otropin receptors. Not only have LMW LHR allosteric
for LMW gonadotropin analogs (e.g., specificity, oral agonists been shown to act as pharmacological chap-
bioavailability, good efficacy). However, evolving erones and restore cell-surface expression of intracellu-
priorities of the pharmaceutical companies running larly retained mutant LHRs () (an outcome of
these programs have meant that further clinical de- the majority of inactivating point mutations in the
velopment of these compounds has been halted. LHR) (), but also, function of binding-deficient and
As multiple injections of FSH are required for signaling-deficient mutants can be restored (). As
oocyte recruitment, LMW orally active FSH agonists there is no current treatment for these patients, the small
would be desirable substitutes. An LMW FSHR al- molecules potentially offer a breakthrough in personal-
losteric agonist (MK-; MSD), developed from a ized and precision medicine in reproduction, with
class of compounds that had promising activity in rat broader implications for treating a wide spectrum of
models, had no effect on follicular development or diseases arising from inactivating mutations in GPCRs.
estradiol production but an effect on thyroid function In addition to the application of gonadotropin
(). Thus, further research is required to produce analogs as regulators of reproduction, agonists and
efficacious orally active FSHR LMW agonists as antagonists of upstream regulators of GnRH (kiss-
substitutes for multiple injections of conventional peptin and neurokinin B) are emerging as subtle and
polypeptide FSH with high specificity. precise regulators of gonadotropin secretion (–). “Future roles and novel
applications of gonadotropins
The observation that MK- has thyroid activity These show promise of more closely simulating nor- targeting extragonadal tissues
serves to highlight the importance of good-selectivity mal physiology as a result of their more proximal are emerging.”
margins for these LMW compounds, which has been a position in the axis regulating GnRH. For example,
limiting factor for the development of many of these kisspeptin has been shown to be effective in a more
analogs. Although peptide analog preparations with gentle induction of ovulation, which may reduce the
mixed LHR/FSHR activity have been/are used clini- risk of OHSS (). Therefore, these upstream regu-
cally (e.g., hMG), “pure” analogs with activity at only lators may have substantial application as surrogates
one receptor are preferred, as this enables design of for gonadotropin therapies. The near-normal FSH and
personalized treatment regimens. In addition, differing low LH in patients with inactivating mutations of
potencies of “mixed activity” LMW analogs at the neurokinin B or its receptor () suggested that
different receptors may have implications for the neurokinin B antagonists may be able to reverse the
design/efficacy of clinical protocols. high LH-to-FSH ratio, characteristic of PCOS, and
As mentioned previously regarding polypeptide this has now been demonstrated (). As hot flashes
gonadotropin molecules, another major stumbling coincide with LH pulses, the slowing of LH pulses with
block to the realization of these exciting possibilities neurokinin B antagonists would potentially ame-
for novel therapeutics has been the acquisition of liorate the condition, which has has now been
reproductive health companies by big pharma and the shown (–).
termination of gonadotropin analog development in Future roles and novel applications of gonado-
view of different priorities of the acquiring companies. tropins targeting extragonadal tissues are emerging
The acquisitions of Organon and Schering halted the and may herald a new era of gonadotropin thera-
promising LHR allosteric agonist program. Likewise, peutics. Postmenopausal women have low estrogens,
the acquisition of Wyeth by Pfizer may have halted elevated FSH, concomitant bone loss, and increased
development of the FSHR agonist. Encouragingly, a body fat, suggesting that FSH, as well as low estrogens,
number of small biotech companies (e.g., Addex may be playing a role. In an ovariectomized mouse
Therapeutics and TocopheRx) have taken up the model, an antibody to FSHb was found to robustly
baton, and we may yet see LMW analogs enter the inhibit bone resorption and stimulate bone synthesis
clinic for a diversity of applications. (). In another study, the antibody was shown to
One exciting developing area of drug discovery is in induce thermogenic adipose tissue and reduce body fat
the arena of allosteric modulators, which can modu- (). There is now substantial literature on the
late the effects of exogenous or endogenous ligands, subject, which is beyond the scope of this review. The
either positively (PAMs) or negatively (NAMs). These reader is therefore referred to comprehensive reviews

doi: 10.1210/er.2018-00052 https://academic.oup.com/edrv 931


REVIEW

on this subject (, ). Despite an increasing body for FSH/FSHR in ovarian cancer (–) open up an
of evidence supporting a role for FSH in bone and fat even wider spectrum of potential novel applications for
regulation, there are contradictory findings in both LH and FSH agonists and antagonists.
human and rodent studies, and robust conclusions are Although these observations are compelling, the
difficult to tease out in view of the relation of estrogen extent of gonadotropin receptor expression in extra-
to FSH and the often-opposed effects of the two gonadal tissues and the effects of gonadotropins on
hormones, such as on osteoclasts (). diverse tissues remain to be fully explored, using a
Elevated LH may also exert neurodegenerative range of experimental approaches and advanced ep-
effects, such as those seen in Alzheimer’s disease idemiological studies. Therefore, it remains to be seen
(, ). Additionally, increased FSH and FSHR ex- whether gonadotropin analogs may have use in these

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/911/5047286 by Research 4 Life user on 22 November 2018


pression in prostatic cancers and a controversial role applications.

References
1. Boivin J, Bunting L, Collins JA, Nygren KG. In- gonadal function. Eur J Endocrinol. 1998;138(5): 26. Haisenleder DJ, Dalkin AC, Ortolano GA, Marshall
ternational estimates of infertility prevalence and 473–481. JC, Shupnik MA. A pulsatile gonadotropin-releasing
treatment-seeking: potential need and demand for 14. Kumar TR, Wang Y, Lu N, Matzuk MM. Follicle hormone stimulus is required to increase tran-
infertility medical care. Hum Reprod. 2007;22(6): stimulating hormone is required for ovarian follicle scription of the gonadotropin subunit genes: evi-
1506–1512. maturation but not male fertility. Nat Genet. 1997; dence for differential regulation of transcription by
2. Millar RP. GnRHs and GnRH receptors. Anim Reprod 15(2):201–204. pulse frequency in vivo. Endocrinology. 1991;128(1):
Sci. 2005;88(1-2):5–28. 15. Thackray VG, Mellon PL, Coss D. Hormones in 509–517.
3. Shaw ND, Histed SN, Srouji SS, Yang J, Lee H, Hall JE. synergy: regulation of the pituitary gonadotropin 27. Fiddes JC, Goodman HM. The gene encoding the
Estrogen negative feedback on gonadotropin se- genes. Mol Cell Endocrinol. 2010;314(2):192–203. common alpha subunit of the four human glyco-
cretion: evidence for a direct pituitary effect in 16. Coss D. Regulation of reproduction via tight control protein hormones. J Mol Appl Genet. 1981;1(1):3–18.
women. J Clin Endocrinol Metab. 2010;95(4): of gonadotropin hormone levels. Mol Cell Endo- 28. Grzesik P, Kreuchwig A, Rutz C, Furkert J, Wiesner B,
1955–1961. crinol. 2018;463:116–130. Schuelein R, Kleinau G, Gromoll J, Krause G. Dif-
4. Corradi PF, Corradi RB, Greene LW. Physiology of 17. Skorupskaite K, George JT, Anderson RA. The ferences in signal activation by LH and hCG are
the hypothalamic pituitary gonadal axis in the male. kisspeptin-GnRH pathway in human reproductive mediated by the LH/CG receptor’s extracellular
Urol Clin North Am. 2016;43(2):151–162. health and disease. Hum Reprod Update. 2014;20(4): hinge region. Front Endocrinol (Lausanne). 2015;6:
5. Rao CV, Lei ZM. The past, present and future of 485–500. 140.
nongonadal LH/hCG actions in reproductive bi- 18. Radovick S, Levine JE, Wolfe A. Estrogenic regulation 29. Lapthorn AJ, Harris DC, Littlejohn A, Lustbader JW,
ology and medicine. Mol Cell Endocrinol. 2007; of the GnRH neuron. Front Endocrinol (Lausanne). Canfield RE, Machin KJ, Morgan FJ, Isaacs NW.
269(1-2):2–8. 2012;3:52. Crystal structure of human chorionic gonadotropin.
6. Stilley JA, Christensen DE, Dahlem KB, Guan R, 19. Carmel PW, Araki S, Ferin M. Pituitary stalk portal Nature. 1994;369(6480):455–461.
Santillan DA, England SK, Al-Hendy A, Kirby PA, blood collection in rhesus monkeys: evidence for 30. Jiang X, Dias JA, He X. Structural biology of gly-
Segaloff DL. FSH receptor (FSHR) expression in pulsatile release of gonadotropin-releasing hormone coprotein hormones and their receptors: insights
human extragonadal reproductive tissues and the (GnRH). Endocrinology. 1976;99(1):243–248. to signaling. Mol Cell Endocrinol. 2014;382(1):
developing placenta, and the impact of its deletion 20. Antunes JL, Carmel PW, Housepian EM, Ferin M. 424–451.
on pregnancy in mice. Biol Reprod. 2014;91(3):74. Luteinizing hormone-releasing hormone in human 31. Ryan RJ, Charlesworth MC, McCormick DJ, Milius
7. Gregory SJ, Kaiser UB. Regulation of gonadotropins pituitary blood. J Neurosurg. 1978;49(3):382–386. RP, Keutmann HT. The glycoprotein hormones:
by inhibin and activin. Semin Reprod Med. 2004; 21. Levine JE, Pau KY, Ramirez VD, Jackson GL. Simul- recent studies of structure-function relationships.
22(3):253–267. taneous measurement of luteinizing hormone- FASEB J. 1988;2(11):2661–2669.
8. Huhtaniemi I. Mutations along the pituitary- releasing hormone and luteinizing hormone 32. Campbell RK, Dean-Emig DM, Moyle WR. Con-
gonadal axis affecting sexual maturation: novel release in unanesthetized, ovariectomized sheep. version of human choriogonadotropin into a fol-
information from transgenic and knockout mice. Endocrinology. 1982;111(5):1449–1455. litropin by protein engineering. Proc Natl Acad Sci
Mol Cell Endocrinol. 2006;254-255:84–90. 22. Levine JE, Duffy MT. Simultaneous measurement of USA. 1991;88(3):760–764.
9. Huhtaniemi I, Ahtiainen P, Pakarainen T, Rulli SB, luteinizing hormone (LH)-releasing hormone, LH, 33. Matzuk MM, Hsueh AJ, Lapolt P, Tsafriri A, Keene JL,
Zhang FP, Poutanen M. Genetically modified and follicle-stimulating hormone release in intact Boime I. The biological role of the carboxyl-terminal
mouse models in studies of luteinising hormone and short-term castrate rats. Endocrinology. 1988; extension of human chorionic gonadotropin [cor-
action. Mol Cell Endocrinol. 2006;252(1-2):126–135. 122(5):2211–2221. rected] beta-subunit. Endocrinology. 1990;126(1):
10. Newton CL, Anderson RC, Katz AA, Millar RP. Loss- 23. Ortolano GA, Haisenleder DJ, Dalkin AC, Iliff- 376–383.
of-function mutations in the human luteinizing Sizemore SA, Landefeld TD, Maurer RA, Marshall 34. Fares FA, Gruener N, Kraiem Z. The role of the
hormone receptor predominantly cause intra- JC. Follicle-stimulating hormone beta subunit asparagine-linked oligosaccharides of the alpha-
cellular retention. Endocrinology. 2016;157(11): messenger ribonucleic acid concentrations during subunit in human thyrotropin bioactivity. Endocri-
4364–4377. the rat estrous cycle. Endocrinology. 1988;123(6): nology. 1996;137(2):555–560.
11. Tapanainen JS, Aittomäki K, Min J, Vaskivuo T, 2946–2948. 35. Morell AG, Gregoriadis G, Scheinberg IH, Hickman J,
Huhtaniemi IT. Men homozygous for an inacti- 24. Padmanabhan V, McFadden K, Mauger DT, Karsch Ashwell G. The role of sialic acid in determining the
vating mutation of the follicle-stimulating hormone FJ, Midgley AR Jr. Neuroendocrine control of follicle- survival of glycoproteins in the circulation. J Biol
(FSH) receptor gene present variable suppression of stimulating hormone (FSH) secretion. I. Direct ev- Chem. 1971;246(5):1461–1467.
spermatogenesis and fertility. Nat Genet. 1997;15(2): idence for separate episodic and basal components 36. Wide L, Eriksson K, Sluss PM, Hall JE. Serum half-life
205–206. of FSH secretion. Endocrinology. 1997;138(1): of pituitary gonadotropins is decreased by sulfo-
12. Tapanainen JS, Vaskivuo T, Aittomäki K, Huhtaniemi 424–432. nation and increased by sialylation in women. J Clin
IT. Inactivating FSH receptor mutations and go- 25. Dalkin AC, Haisenleder DJ, Ortolano GA, Ellis TR, Endocrinol Metab. 2009;94(3):958–964.
nadal dysfunction. Mol Cell Endocrinol. 1998;145(1-2): Marshall JC. The frequency of gonadotropin- 37. LaPolt PS, Nishimori K, Fares FA, Perlas E, Boime I,
129–135. releasing-hormone stimulation differentially regu- Hsueh AJ. Enhanced stimulation of follicle matu-
13. Huhtaniemi IT, Aittomäki K. Mutations of follicle- lates gonadotropin subunit messenger ribonucleic ration and ovulatory potential by long acting
stimulating hormone and its receptor: effects on acid expression. Endocrinology. 1989;125(2):917–924. follicle-stimulating hormone agonists with extended

932 Anderson et al Peptide and Nonpeptide Gonadotropin Analogs Endocrine Reviews, December 2018, 39(6):911–937
REVIEW

carboxyl-terminal peptides. Endocrinology. 1992; Casarini L. Heterogeneous hCG and hMG com- syndrome. Cochrane Database Syst Rev. 2015;(9):
131(6):2514–2520. mercial preparations result in different intracellular CD010290.
38. Lambert A, Rodgers M, Mitchell R, Wood AM, signalling but induce a similar long-term pro- 69. Orvieto R, Nahum R, Rabinson J, Ashkenazi J,
Wardle C, Hilton B, Robertson WR. In-vitro bio- gesterone response in vitro. Mol Hum Reprod. 2017; Anteby EY, Meltcer S. Follitropin-alpha (Gonal-F)
potency and glycoform distribution of recombi- 23(10):685–697. versus follitropin-beta (Puregon) in controlled
nant human follicle stimulating hormone (Org 53. Dahl KD, Bicsak TA, Hsueh AJ. Naturally occurring ovarian hyperstimulation for in vitro fertilization: is
32489), Metrodin and Metrodin-HP. Hum Reprod. antihormones: secretion of FSH antagonists by there any difference? Fertil Steril. 2009;91(4, Suppl):
1995;10(7):1928–1935. women treated with a GnRH analog. Science. 1988; 1522–1525.
39. Matzuk MM, Keene JL, Boime I. Site specificity of 239(4835):72–74. 70. Fares FA, Suganuma N, Nishimori K, LaPolt PS,
the chorionic gonadotropin N-linked oligosaccha- 54. Ulloa-Aguirre A, Timossi C, Barrios-de-Tomasi J, Hsueh AJ, Boime I. Design of a long-acting follitropin
rides in signal transduction. J Biol Chem. 1989; Maldonado A, Nayudu P. Impact of carbohydrate agonist by fusing the C-terminal sequence of the
264(5):2409–2414. heterogeneity in function of follicle-stimulating chorionic gonadotropin beta subunit to the folli-

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/911/5047286 by Research 4 Life user on 22 November 2018


40. Fares FA, Levi F, Reznick AZ, Kraiem Z. Engineering a hormone: studies derived from in vitro and tropin beta subunit. Proc Natl Acad Sci USA. 1992;
potential antagonist of human thyrotropin and in vivo models. Biol Reprod. 2003;69(2):379–389. 89(10):4304–4308.
thyroid-stimulating antibody. J Biol Chem. 2001; 55. Wehbi V, Tranchant T, Durand G, Musnier A, 71. Bouloux PM, Handelsman DJ, Jockenhövel F,
276(7):4543–4548. Decourtye J, Piketty V, Butnev VY, Bousfield GR, Nieschlag E, Rabinovici J, Frasa WL, de Bie JJ,
41. Azzam N, Bar-Shalom R, Kraiem Z, Fares F. Human Crépieux P, Maurel MC, Reiter E. Partially degly- Voortman G, Itskovitz-Eldor J; FSH-CTP study
thyrotropin (TSH) variants designed by site- cosylated equine LH preferentially activates beta- group. First human exposure to FSH-CTP in
directed mutagenesis block TSH activity in vitro arrestin-dependent signaling at the follicle-stimulating hypogonadotrophic hypogonadal males. Hum
and in vivo. Endocrinology. 2005;146(6):2845–2850. hormone receptor. Mol Endocrinol. 2010;24(3):561–573. Reprod. 2001;16(8):1592–1597.
42. Matzuk MM, Boime I. Mutagenesis and gene 56. Landomiel F, Gallay N, Jégot G, Tranchant T, 72. Duijkers IJ, Klipping C, Boerrigter PJ, Machielsen CS,
transfer define site-specific roles of the gonado- Durand G, Bourquard T, Crépieux P, Poupon A, De Bie JJ, Voortman G. Single dose pharmacoki-
tropin oligosaccharides. Biol Reprod. 1989;40(1): Reiter E. Biased signalling in follicle stimulating netics and effects on follicular growth and serum
48–53. hormone action. Mol Cell Endocrinol. 2014;382(1): hormones of a long-acting recombinant FSH
43. Matzuk MM, Boime I. Site-specific mutagenesis 452–459. preparation (FSH-CTP) in healthy pituitary-
defines the intracellular role of the asparagine- 57. Gloaguen P, Crépieux P, Heitzler D, Poupon A, suppressed females. Hum Reprod. 2002;17(8):
linked oligosaccharides of chorionic gonado- Reiter E. Mapping the follicle-stimulating hormone- 1987–1993.
tropin beta subunit. J Biol Chem. 1988;263(32): induced signaling networks. Front Endocrinol 73. Sugahara T, Pixley MR, Minami S, Perlas E, Ben-
17106–17111. (Lausanne). 2011;2:45. Menahem D, Hsueh AJ, Boime I. Biosynthesis of a
44. Matzuk MM, Boime I. The role of the asparagine- 58. Hunzicker-Dunn M, Maizels ET. FSH signaling biologically active single peptide chain containing
linked oligosaccharides of the alpha subunit in the pathways in immature granulosa cells that regulate the human common alpha and chorionic gonad-
secretion and assembly of human chorionic go- target gene expression: branching out from protein otropin beta subunits in tandem. Proc Natl Acad Sci
nadotrophin. J Cell Biol. 1988;106(4):1049–1059. kinase A. Cell Signal. 2006;18(9):1351–1359. USA. 1995;92(6):2041–2045.
45. Flack MR, Froehlich J, Bennet AP, Anasti J, Nisula BC. 59. Xie YB, Wang H, Segaloff DL. Extracellular domain 74. Garcia-Campayo V, Sato A, Hirsch B, Sugahara T,
Site-directed mutagenesis defines the individual of lutropin/choriogonadotropin receptor expressed Muyan M, Hsueh AJ, Boime I. Design of stable
roles of the glycosylation sites on follicle-stimulating in transfected cells binds choriogonadotropin biologically active recombinant lutropin analogs.
hormone. J Biol Chem. 1994;269(19):14015–14020. with high affinity. J Biol Chem. 1990;265(35): Nat Biotechnol. 1997;15(7):663–667.
46. Bishop LA, Robertson DM, Cahir N, Schofield PR. 21411–21414. 75. Sugahara T, Grootenhuis PD, Sato A, Kudo M, Ben-
Specific roles for the asparagine-linked carbohy- 60. Fan QR, Hendrickson WA. Structure of human Menahem D, Pixley MR, Hsueh AJ, Boime I. Ex-
drate residues of recombinant human follicle follicle-stimulating hormone in complex with its pression of biologically active fusion genes
stimulating hormone in receptor binding and signal receptor. Nature. 2005;433(7023):269–277. encoding the common alpha subunit and either
transduction. Mol Endocrinol. 1994;8(6):722–731. 61. Fan QR, Hendrickson WA. Structural biology of the CG beta or FSH beta subunits: role of a linker
47. Arey BJ, Stevis PE, Deecher DC, Shen ES, Frail DE, glycoprotein hormones and their receptors. Endo- sequence. Mol Cell Endocrinol. 1996;125(1-2):
Negro-Vilar A, Lopez FJ. Induction of promiscuous crine. 2005;26(3):179–188. 71–77.
G protein coupling of the follicle-stimulating hor- 62. Jiang X, Liu H, Chen X, Chen PH, Fischer D, Sriraman 76. Kanda M, Jablonka-Shariff A, Sato A, Pixley MR,
mone (FSH) receptor: a novel mechanism for V, Yu HN, Arkinstall S, He X. Structure of follicle- Bos E, Hiro’oka T, Ben-Menahem D, Boime I.
transducing pleiotropic actions of FSH isoforms. stimulating hormone in complex with the entire Genetic fusion of an alpha-subunit gene to
Mol Endocrinol. 1997;11(5):517–526. ectodomain of its receptor. Proc Natl Acad Sci USA. the follicle-stimulating hormone and chorionic
48. Timossi CM, Barrios de Tomasi J, Zambrano E, 2012;109(31):12491–12496. gonadotropin-beta subunit genes: production of a
González R, Ulloa-Aguirre A. A naturally occurring 63. Kreuchwig A, Kleinau G, Krause G. Research re- bifunctional protein. Mol Endocrinol. 1999;13(11):
basically charged human follicle-stimulating hor- source: novel structural insights bridge gaps in 1873–1881.
mone (FSH) variant inhibits FSH-induced androgen glycoprotein hormone receptor analyses. Mol 77. Rutigliano HM, Adams BM, Jablonka-Shariff A,
aromatization and tissue-type plasminogen acti- Endocrinol. 2013;27(8):1357–1363. Boime I, Adams TE. Effect of single-chain ovine
vator enzyme activity in vitro. Neuroendocrinology. 64. Krause G, Kreuchwig A, Kleinau G. Extended and gonadotropins with dual activity on ovarian
1998;67(3):153–163. structurally supported insights into extracellular function in sheep. Reproduction. 2014;148(2):
49. Timossi CM, Barrios-de-Tomasi J, González-Suárez hormone binding, signal transduction and organi- 129–136.
R, Arranz MC, Padmanabhan V, Conn PM, Ulloa- zation of the thyrotropin receptor. PLoS One. 2012; 78. Perlman S, van den Hazel B, Christiansen J, Gram-
Aguirre A. Differential effects of the charge variants 7(12):e52920. Nielsen S, Jeppesen CB, Andersen KV, Halkier T,
of human follicle-stimulating hormone. J Endocrinol. 65. Brüser A, Schulz A, Rothemund S, Ricken A, Okkels S, Schambye HT. Glycosylation of an
2000;165(2):193–205. Calebiro D, Kleinau G, Schöneberg T. The activation N-terminal extension prolongs the half-life and
50. Zambrano E, Zariñán T, Olivares A, Barrios-de- mechanism of glycoprotein hormone receptors increases the in vivo activity of follicle stimulating
Tomasi J, Ulloa-Aguirre A. Receptor binding ac- with implications in the cause and therapy of hormone. J Clin Endocrinol Metab. 2003;88(7):
tivity and in vitro biological activity of the human endocrine diseases. J Biol Chem. 2016;291(2): 3227–3235.
FSH charge isoforms as disclosed by heterologous 508–520. 79. Klein J, Lobel L, Pollak S, Lustbader B, Ogden RT,
and homologous assay systems: implications for the 66. Zondek B. Weitere untersuchungen zur dar- Sauer MV, Lustbader JW. Development and char-
structure-function relationship of the FSH variants. stellung. biologie und klinik des hypo- acterization of a long-acting recombinant hFSH
Endocrine. 1999;10(2):113–121. physenvorderlappenhormons (prolan). Zentralbl agonist. Hum Reprod. 2003;18(1):50–56.
51. Arey BJ, López FJ. Are circulating gonadotropin Gynakol. 1929;14:834–848. 80. Weenen C, Peña JE, Pollak SV, Klein J, Lobel L,
isoforms naturally occurring biased agonists? Basic 67. Lunenfeld B. Historical perspectives in gonadotro- Trousdale RK, Palmer S, Lustbader EG, Ogden RT,
and therapeutic implications. Rev Endocr Metab phin therapy. Hum Reprod Update. 2004;10(6): Lustbader JW. Long-acting follicle-stimulating
Disord. 2011;12(4):275–288. 453–467. hormone analogs containing N-linked glycosyla-
52. Riccetti L, Klett D, Ayoub MA, Boulo T, Pignatti E, 68. Weiss NS, Nahuis M, Bayram N, Mol BW, Van der tion exhibited increased bioactivity compared with
Tagliavini S, Varani M, Trenti T, Nicoli A, Veen F, van Wely M. Gonadotrophins for ovulation o-linked analogs in female rats. J Clin Endocrinol
Capodanno F, La Sala GB, Reiter E, Simoni M, induction in women with polycystic ovarian Metab. 2004;89(10):5204–5212.

doi: 10.1210/er.2018-00052 https://academic.oup.com/edrv 933


REVIEW

81. Low SC, inventor; Bioverativ Therapeutics Inc., as- efficacy and safety of corifollitropin alfa combined hormone and antral follicle count as biomarkers of
signee. Heterodimeric follicle stimulating hormone- with hCG in adult men with hypogonadotropic ovarian response. Hum Reprod Update. 2015;21(6):
Fc (FSH-Fc) fusion proteins for the treatment of hypogonadism. Reprod Biol Endocrinol. 2017;15(1): 698–710.
infertility. Patent US7601516B2. 2009. 17. 107. Polat M, Bozdag G, Yarali H. Best protocol for
82. Roy S, Setlur S, Gadkari RA, Krishnamurthy HN, 95. Dwyer AA, Sykiotis GP, Hayes FJ, Boepple PA, Lee H, controlled ovarian hyperstimulation in assisted
Dighe RR. Translational fusion of two beta-subunits Loughlin KR, Dym M, Sluss PM, Crowley WF Jr, reproductive technologies: fact or opinion? Semin
of human chorionic gonadotropin results in pro- Pitteloud N. Trial of recombinant follicle-stimulating Reprod Med. 2014;32(4):262–271.
duction of a novel antagonist of the hormone. hormone pretreatment for GnRH-induced fertility 108. Lensen SF, Wilkinson J, Leijdekkers JA, La Marca A,
Endocrinology. 2007;148(8):3977–3986. in patients with congenital hypogonadotropic Mol BWJ, Marjoribanks J, Torrance H, Broekmans FJ.
83. Vardhana PA, Julius MA, Pollak SV, Lustbader EG, hypogonadism. J Clin Endocrinol Metab. 2013;98(11): Individualised gonadotropin dose selection using
Trousdale RK, Lustbader JW. A unique human E1790–E1795. markers of ovarian reserve for women undergoing
chorionic gonadotropin antagonist suppresses 96. Rohayem J, Hauffa BP, Zacharin M, Kliesch S, in vitro fertilisation plus intracytoplasmic sperm

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/911/5047286 by Research 4 Life user on 22 November 2018


ovarian hyperstimulation syndrome in rats. Endo- Zitzmann M; “German Adolescent Hypogonado- injection (IVF/ICSI). Cochrane Database Syst Rev.
crinology. 2009;150(8):3807–3814. tropic Hypogonadism Study Group”. Testicular 2018;2:CD012693.
84. Uchiyama Y, Hosoe M, Sato T, Takahashi T. Bi- growth and spermatogenesis: new goals for pu- 109. Fauser BC, Devroey P, Macklon NS. Multiple birth
ological and immunological characteristics of bertal hormone replacement in boys with hypo- resulting from ovarian stimulation for subfertility
porcine follicle-stimulating hormone chemically gonadotropic hypogonadism? -a multicentre treatment. Lancet. 2005;365(9473):1807–1816.
modified with a polyethylene glycol derivative. Vet J. prospective study of hCG/rFSH treatment out- 110. Kulkarni AD, Jamieson DJ, Jones HW Jr, Kissin DM,
2010;184(2):208–211. comes during adolescence. Clin Endocrinol (Oxf). Gallo MF, Macaluso M, Adashi EY. Fertility treat-
85. Beckers NG, Macklon NS, Devroey P, Platteau P, 2017;86(1):75–87. ments and multiple births in the United States.
Boerrigter PJ, Fauser BC. First live birth after ovarian 97. Gemzell CA, Diczfalusy E, Tillinger G. Clinical effect N Engl J Med. 2013;369(23):2218–2225.
stimulation using a chimeric long-acting human of human pituitary follicle-stimulating hormone 111. Lambalk CB, Banga FR, Huirne JA, Toftager M,
recombinant follicle-stimulating hormone (FSH) (FSH). J Clin Endocrinol Metab. 1958;18(12): Pinborg A, Homburg R, van der Veen F, van Wely M.
agonist (recFSH-CTP) for in vitro fertilization. Fertil 1333–1348. GnRH antagonist versus long agonist protocols in
Steril. 2003;79(3):621–623. 98. White DM, Polson DW, Kiddy D, Sagle P, Watson H, IVF: a systematic review and meta-analysis ac-
86. Corifollitropin alfa Ensure Study Group. Corifolli- Gilling-Smith C, Hamilton-Fairley D, Franks S. In- counting for patient type. Hum Reprod Update.
tropin alfa for ovarian stimulation in IVF: a ran- duction of ovulation with low-dose gonadotropins 2017;23(5):560–579.
domized trial in lower-body-weight women in polycystic ovary syndrome: an analysis of 109 112. Al-Inany HG, Youssef MA, Ayeleke RO, Brown J, Lam
[published correction appears in Reprod Biomed pregnancies in 225 women. J Clin Endocrinol Metab. WS, Broekmans FJ. Gonadotrophin-releasing hor-
Online. 2014;29(3):395]. Reprod Biomed Online. 2010; 1996;81(11):3821–3824. mone antagonists for assisted reproductive tech-
21(1):66–76. 99. Fauser BC, Donderwinkel P, Schoot DC. The step- nology. Cochrane Database Syst Rev. 2016;4:
87. Fauser BC, Alper MM, Ledger W, Schoolcraft WB, down principle in gonadotrophin treatment and CD001750.
Zandvliet A, Mannaerts BM; Engage Investigators. the role of GnRH analogues. Baillieres Clin Obstet 113. Devroey P, Polyzos NP, Blockeel C. An OHSS-free
Pharmacokinetics and follicular dynamics of cor- Gynaecol. 1993;7(2):309–330. clinic by segmentation of IVF treatment. Hum
ifollitropin alfa versus recombinant FSH during 100. Balen AH, Morley LC, Misso M, Franks S, Legro RS, Reprod. 2011;26(10):2593–2597.
ovarian stimulation for IVF. Reprod Biomed Online. Wijeyaratne CN, Stener-Victorin E, Fauser BC, 114. Pandian Z, Marjoribanks J, Ozturk O, Serour G,
2010;21(5):593–601. Norman RJ, Teede H. The management of an- Bhattacharya S. Number of embryos for transfer
88. Fatemi HM, Oberyé J, Popovic-Todorovic B, Witjes ovulatory infertility in women with polycystic ovary following in vitro fertilisation or intra-cytoplasmic
H, Mannaerts B, Devroey P. Corifollitropin alfa in a syndrome: an analysis of the evidence to support sperm injection. Cochrane Database Syst Rev. 2013;
long GnRH agonist protocol: proof of concept trial. the development of global WHO guidance. Hum (7):CD003416.
Fertil Steril. 2010;94(5):1922–1924. Reprod Update. 2016;22(6):687–708. 115. Anderson RA, Kinniburgh D, Baird DT. Preliminary
89. Devroey P, Boostanfar R, Koper NP, Mannaerts BM, 101. Wang R, Kim BV, van Wely M, Johnson NP, Costello experience of the use of a gonadotrophin-releasing
Ijzerman-Boon PC, Fauser BC; ENGAGE In- MF, Zhang H, Ng EH, Legro RS, Bhattacharya S, hormone antagonist in ovulation induction/in-vitro
vestigators. A double-blind, non-inferiority RCT Norman RJ, Mol BW. Treatment strategies for fertilization prior to cancer treatment. Hum Reprod.
comparing corifollitropin alfa and recombinant FSH women with WHO group II anovulation: systematic 1999;14(10):2665–2668.
during the first seven days of ovarian stimulation review and network meta-analysis. BMJ. 2017;356: 116. Cakmak H, Katz A, Cedars MI, Rosen MP. Effective
using a GnRH antagonist protocol. Hum Reprod. j138. method for emergency fertility preservation: random-
2009;24(12):3063–3072. 102. Yu Y, Fang L, Zhang R, He J, Xiong Y, Guo X, Du Q, start controlled ovarian stimulation. Fertil Steril. 2013;
90. Norman RJ, Zegers-Hochschild F, Salle BS, Elbers J, Huang Y, Sun Y. Comparative effectiveness of 9 100(6):1673–1680.
Heijnen E, Marintcheva-Petrova M, Mannaerts B; ovulation-induction therapies in patients with 117. Oktay K, Buyuk E, Libertella N, Akar M, Rosenwaks
Trust Investigators. Repeated ovarian stimulation clomiphene citrate-resistant polycystic ovary syn- Z. Fertility preservation in breast cancer patients:
with corifollitropin alfa in patients in a GnRH an- drome: a network meta-analysis. Sci Rep. 2017;7(1): a prospective controlled comparison of ovarian
tagonist protocol: no concern for immunogenicity. 3812. stimulation with tamoxifen and letrozole for em-
Hum Reprod. 2011;26(8):2200–2208. 103. Homburg R, Hendriks ML, König TE, Anderson RA, bryo cryopreservation. J Clin Oncol. 2005;23(19):
91. Roth MY, Page ST, Lin K, Anawalt BD, Matsumoto Balen AH, Brincat M, Child T, Davies M, D’Hooghe 4347–4353.
AM, Snyder CN, Marck BT, Bremner WJ, Amory JK. T, Martinez A, Rajkhowa M, Rueda-Saenz R, 118. Oktay K, Hourvitz A, Sahin G, Oktem O, Safro B, Cil
Dose-dependent increase in intratesticular testos- Hompes P, Lambalk CB. Clomifene citrate or low- A, Bang H. Letrozole reduces estrogen and go-
terone by very low-dose human chorionic go- dose FSH for the first-line treatment of infertile nadotropin exposure in women with breast cancer
nadotropin in normal men with experimental women with anovulation associated with polycystic undergoing ovarian stimulation before chemo-
gonadotropin deficiency. J Clin Endocrinol Metab. ovary syndrome: a prospective randomized multi- therapy. J Clin Endocrinol Metab. 2006;91(10):
2010;95(8):3806–3813. national study. Hum Reprod. 2012;27(2):468–473. 3885–3890.
92. Liu PY, Baker HW, Jayadev V, Zacharin M, Conway 104. Li HW, Van Esch M, De Vries J, Duncan WC, 119. Oktay K, Turan V, Bedoschi G, Pacheco FS, Moy F.
AJ, Handelsman DJ. Induction of spermatogenesis Anderson RA. Gonadotrophin ovulation induction Fertility preservation success subsequent to con-
and fertility during gonadotropin treatment of is a successful treatment for World Health Orga- current aromatase inhibitor treatment and ovarian
gonadotropin-deficient infertile men: predictors of nisation Group II anovulatory subfertility in women stimulation in women with breast cancer. J Clin
fertility outcome. J Clin Endocrinol Metab. 2009; aged up to 40 and with body mass index up to 34. Oncol. 2015;33(22):2424–2429.
94(3):801–808. Hum Fertil (Camb). 2010;13(1):35–40. 120. Tsampras N, Gould D, Fitzgerald CT. Double ovarian
93. Rastrelli G, Corona G, Mannucci E, Maggi M. Factors 105. Nelson SM, Yates RW, Lyall H, Jamieson M, Traynor stimulation (DuoStim) protocol for fertility pres-
affecting spermatogenesis upon gonadotropin- I, Gaudoin M, Mitchell P, Ambrose P, Fleming R. ervation in female oncology patients. Hum Fertil
replacement therapy: a meta-analytic study. Androl- Anti-Müllerian hormone-based approach to con- (Camb). 2017;20(4):248–253.
ogy. 2014;2(6):794–808. trolled ovarian stimulation for assisted conception. 121. Liu C, Jiang H, Zhang W, Yin H. Double ovarian
94. Nieschlag E, Bouloux PG, Stegmann BJ, Shankar RR, Hum Reprod. 2009;24(4):867–875. stimulation during the follicular and luteal phase in
Guan Y, Tzontcheva A, McCrary Sisk C, Behre HM. 106. Iliodromiti S, Anderson RA, Nelson SM. Technical women $38 years: a retrospective case-control
An open-label clinical trial to investigate the and performance characteristics of anti-Müllerian study. Reprod Biomed Online. 2017;35(6):678–684.

934 Anderson et al Peptide and Nonpeptide Gonadotropin Analogs Endocrine Reviews, December 2018, 39(6):911–937
REVIEW

122. Ubaldi FM, Capalbo A, Vaiarelli A, Cimadomo D, 136. Rebois RV, Fishman PH. Deglycosylated human 154. Maclean D, Holden F, Davis AM, Scheuerman RA,
Colamaria S, Alviggi C, Trabucco E, Venturella R, chorionic gonadotropin. An antagonist to de- Yanofsky S, Holmes CP, Fitch WL, Tsutsui K, Barrett
Vajta G, Rienzi L. Follicular versus luteal phase sensitization and down-regulation of the gonado- RW, Gallop MA. Agonists of the follicle stimulating
ovarian stimulation during the same menstrual tropin receptor-adenylate cyclase system. J Biol hormone receptor from an encoded thiazolidinone
cycle (DuoStim) in a reduced ovarian reserve Chem. 1983;258(21):12775–12778. library. J Comb Chem. 2004;6(2):196–206.
population results in a similar euploid blastocyst 137. Fares F. The role of O-linked and N-linked oligo- 155. Wrobel J, Jetter J, Kao W, Rogers J, Di L, Chi J, Peréz
formation rate: new insight in ovarian reserve ex- saccharides on the structure-function of glyco- MC, Chen GC, Shen ES. 5-Alkylated thiazolidinones
ploitation. Fertil Steril. 2016;105(6):1488–1495.e1. protein hormones: development of agonists and as follicle-stimulating hormone (FSH) receptor ag-
123. Ben-Menahem D, Jablonka-Shariff A, Hyde RK, antagonists. Biochim Biophys Acta. 2006;1760(4): onists. Bioorg Med Chem. 2006;14(16):5729–5741.
Pixley MR, Srivastava S, Berger P, Boime I. The 560–567. 156. Pelletier JC, Rogers J, Wrobel J, Perez MC, Shen ES.
position of the alpha and beta subunits in a single 138. Moyle WR, inventor; University of Medicine and Preparation of highly substituted gamma-lactam
chain variant of human chorionic gonadotropin Dentistry of New Jersey, assignee. Method for follicle stimulating hormone receptor agonists.

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/911/5047286 by Research 4 Life user on 22 November 2018


affects the heterodimeric interaction of the sub- making hormone heterodimers. Patent US6486303B1. Bioorg Med Chem. 2005;13(21):5986–5995.
units and receptor-binding epitopes. J Biol Chem. 2002. 157. Yanofsky SD, Shen ES, Holden F, Whitehorn E,
2001;276(32):29871–29879. 139. Patton PE, Calvo FO, Fujimoto VY, Bergert ER, Aguilar B, Tate E, Holmes CP, Scheuerman R,
124. Lemke EP, Adams BM, Jablonka-Shariff A, Boime I, Kempers RD, Ryan RJ. The effect of deglycosylated MacLean D, Wu MM, Frail DE, López FJ, Winneker R,
Adams TE. Single-chain human gonadotropin an- human chorionic gonadotropin on corpora luteal Arey BJ, Barrett RW. Allosteric activation of the
alogs induce follicle development in sheep. function in healthy women. Fertil Steril. 1988;49(4): follicle-stimulating hormone (FSH) receptor by
J Endocrinol. 2008;196(3):593–600. 620–625. selective, nonpeptide agonists. J Biol Chem. 2006;
125. Klein J, Lobel L, Pollak S, Ferin M, Xiao E, Sauer M, 140. Keutmann HT, McIlroy PJ, Bergert ER, Ryan RJ. 281(19):13226–13233.
Lustbader JW. Pharmacokinetics and pharmaco- Chemically deglycosylated human chorionic go- 158. Sriraman V, Denis D, de Matos D, Yu H, Palmer S,
dynamics of single-chain recombinant human nadotropin subunits: characterization and bi- Nataraja S. Investigation of a thiazolidinone de-
follicle-stimulating hormone containing the human ological properties. Biochemistry. 1983;22(13): rivative as an allosteric modulator of follicle stim-
chorionic gonadotropin carboxyterminal peptide 3067–3072. ulating hormone receptor: evidence for its ability
in the rhesus monkey. Fertil Steril. 2002;77(6): 141. Nataraja SG, Yu HN, Palmer SS. Discovery and to support follicular development and ovulation.
1248–1255. development of small molecule allosteric modu- Biochem Pharmacol. 2014;89(2):266–275.
126. Ruman JI, Pollak S, Trousdale RK, Klein J, Lustbader lators of glycoprotein hormone receptors. Front 159. Arey BJ, Yanofsky SD, Claudia Pérez M, Holmes CP,
JW. Effects of long-acting recombinant human Endocrinol (Lausanne). 2015;6:142. Wrobel J, Gopalsamy A, Stevis PE, López FJ,
follicle-stimulating hormone analogs containing 142. Brynhildsen J. Combined hormonal contraceptives: Winneker RC. Differing pharmacological activities of
N-linked glycosylation on murine folliculogenesis. prescribing patterns, compliance, and benefits thiazolidinone analogs at the FSH receptor. Biochem
Fertil Steril. 2005;83(4, Suppl 1):1303–1309. versus risks. Ther Adv Drug Saf. 2014;5(5):201–213. Biophys Res Commun. 2008;368(3):723–728.
127. Trousdale RK, Yu B, Pollak SV, Husami N, Vidali A, 143. Wang CI, Lewis RJ. Emerging opportunities for al- 160. Dias JA, Bonnet B, Weaver BA, Watts J, Kluetzman K,
Lustbader JW. Efficacy of native and hyper- losteric modulation of G-protein coupled receptors. Thomas RM, Poli S, Mutel V, Campo B. A negative
glycosylated follicle-stimulating hormone analogs Biochem Pharmacol. 2013;85(2):153–162. allosteric modulator demonstrates biased antago-
for promoting fertility in female mice. Fertil Steril. 144. Manglik A, Kobilka BK, Steyaert J. Nanobodies to nism of the follicle stimulating hormone receptor.
2009;91(1):265–270. study G protein-coupled receptor structure and Mol Cell Endocrinol. 2011;333(2):143–150.
128. Park A, Honey DM, Hou L, Bird JJ, Zarazinski C, function. Annu Rev Pharmacol Toxicol. 2017;57(1): 161. Arey BJ, Deecher DC, Shen ES, Stevis PE, Meade EH
Searles M, Braithwaite C, Kingsbury JS, Kyazike J, 19–37. Jr, Wrobel J, Frail DE, López FJ. Identification and
Culm-Merdek K, Greene B, Stefano JE, Qiu H, 145. Gentry PR, Sexton PM, Christopoulos A. Novel characterization of a selective, nonpeptide follicle-
McPherson JM, Pan CQ. Carbohydrate-mediated allosteric modulators of G protein-coupled re- stimulating hormone receptor antagonist. Endo-
polyethylene glycol conjugation of TSH improves its ceptors. J Biol Chem. 2015;290(32):19478–19488. crinology. 2002;143(10):3822–3829.
pharmacological properties. Endocrinology. 2013; 146. Heitman LH, Kleinau G, Brussee J, Krause G, Ijzerman 162. van Straten NC, van Berkel TH, Demont DR, Karstens
154(3):1373–1383. AP. Determination of different putative allosteric WJ, Merkx R, Oosterom J, Schulz J, van Someren RG,
129. DeFrees S, Bayer RJ, Bowe C. inventors; Neose binding pockets at the lutropin receptor by using Timmers CM, van Zandvoort PM. Identification
Technologies Inc., assignee. Glycopegylated follicle diverse drug-like low molecular weight ligands. Mol of substituted 6-amino-4-phenyltetrahydroquinoline
stimulating hormone. Patent US 2008/0015142 A1. Cell Endocrinol. 2012;351(2):326–336. derivatives: potent antagonists for the follicle-
2008. 147. Arey BJ. Allosteric modulators of glycoprotein stimulating hormone receptor. J Med Chem. 2005;
130. Capon DJ, Chamow SM, Mordenti J, Marsters SA, hormone receptors: discovery and therapeutic 48(6):1697–1700.
Gregory T, Mitsuya H, Byrn RA, Lucas C, Wurm FM, potential. Endocrine. 2008;34(1-3):1–10. 163. van Koppen CJ, Verbost PM, van de Lagemaat R,
Groopman JE, Broder S, Smith DH. Designing CD4 148. van Straten N, Timmers M. Chapter 8 non- Karstens WJ, Loozen HJ, van Achterberg TA, van
immunoadhesins for AIDS therapy. Nature. 1989; peptide ligands for the gonadotropin receptors. Amstel MG, Brands JH, van Doornmalen EJ, Wat J,
337(6207):525–531. In: Macor JE, ed. Annual Reports in Medicinal Mulder SJ, Raafs BC, Verkaik S, Hanssen RG, Timmers
131. Strohl WR. Fusion proteins for half-life extension of Chemistry. Vol 44. Cambridge, MA: Academic CM. Signaling of an allosteric, nanomolar potent,
biologics as a strategy to make biobetters. BioDrugs. Press; 2009:171–188. low molecular weight agonist for the follicle-
2015;29(4):215–239. 149. El Tayer N, Reddy A, Buckler D, Magar S, inventors; stimulating hormone receptor. Biochem Pharma-
132. Levin D, Golding B, Strome SE, Sauna ZE. Fc fusion Applied Research Systems ARS Holding NV., as- col. 2013;85(8):1162–1170.
as a platform technology: potential for modulating signee. FSH mimetics for the treatment of infertility. 164. Heitman LH, Narlawar R, de Vries H, Willemsen MN,
immunogenicity. Trends Biotechnol. 2015;33(1): Patent US6235755B1. 2001. Wolfram D, Brussee J, Ijzerman AP. Substituted
27–34. 150. Look GC, Schullek JR, Holmes CP, Chinn JP, Gordon terphenyl compounds as the first class of low
133. Russell-Jones GJ, Howe P, de Aizpurua HJ, Burge GL, EM, Gallop M. The identification of cyclooxygenase- molecular weight allosteric inhibitors of the lutei-
inventors; Biotech Australia Pty Ltd., assignee. Oral 1 inhibitors from 4-thiazolidinone combinatorial nizing hormone receptor. J Med Chem. 2009;52(7):
delivery of biologically active substances bound to libraries. Bioorg Med Chem Lett. 1996;6(6):707–712. 2036–2042.
vitamin B12 or analogues thereof. Patent 151. Mutlib AE, Jurcak J, Hrib N. Metabolism of an atypical 165. van Koppen CJ, Zaman GJ, Timmers CM, Kelder J,
US5428023A. 1995. antipsychotic agent, 3-[4-[4-(6-fluorobenzo[b]thien-3-yl)- Mosselman S, van de Lagemaat R, Smit MJ,
134. Hashimoto S, Kimura K, Kuramochi T, Aoyagi K, 1-piperazinyl]butyl]-2, 5,5-trimethyl-4-thiazolidinone Hanssen RG. A signaling-selective, nanomolar
Hirako M, Kawaguchi M, Iwata H, Hirao M, Kitada K, (HP236). Drug Metab Dispos. 1996;24(10):1139–1150. potent allosteric low molecular weight agonist
Hirasawa K, Ueda M. Responsiveness of rabbits to 152. Tripathi M, Verma M, Gujrati VR, Palit G, Shanker K. for the human luteinizing hormone receptor.
superovulation treatment by a single injection of Thiazolidinone congeners as central nervous system Naunyn Schmiedebergs Arch Pharmacol. 2008;
follicle-stimulating hormone with aluminum hy- active agents. Arzneimittelforschung. 1993;43(6): 378(5):503–514.
droxide gel. Mol Reprod Dev. 2007;74(9):1208–1212. 632–635. 166. Jorand-Lebrun C, Brondyk B, Lin J, Magar S, Murray
135. Kimura K, Hirako M, Iwata H, Aoki M, Kawaguchi M, 153. Santora VJ, Covel JA, Hayashi R, Webb RR, inventors; R, Reddy A, Shroff H, Wands G, Weiser W, Xu Q,
Seki M. Successful superovulation of cattle by a Arena Pharmaceuticals Inc., assignee. Ligands of McKenna S, Brugger N. Identification, synthesis, and
single administration of FSH in aluminum hy- follicle stimulating hormone receptor and methods biological evaluation of novel pyrazoles as low
droxide gel. Theriogenology. 2007;68(4):633–639. of use thereof. Patent WO2005087765A1. 2004. molecular weight luteinizing hormone receptor

doi: 10.1210/er.2018-00052 https://academic.oup.com/edrv 935


REVIEW

agonists. Bioorg Med Chem Lett. 2007;17(7): 181. Coats SJ, Fitzpatrick LJ, Hlasta DJ, Lanter CL, reproductive age. J Clin Endocrinol Metab. 2013;
2080–2085. Macielag MJ, Pan K, Rivero RA, Palmer SS, inventors; 98(4):1558–1566.
167. Guo T, Adang AE, Dolle RE, Dong G, Fitzpatrick D, Ortho-McNeil Pharmaceutical Inc., assignee. 193. Bonger KM, van den Berg RJ, Knijnenburg AD,
Geng P, Ho KK, Kultgen SG, Liu R, McDonald E, Substituted aminoalkylamide derivatives as antag- Heitman LH, van Koppen CJ, Timmers CM,
McGuinness BF, Saionz KW, Valenzano KJ, van onists of follicle stimulating hormone. Patent Overkleeft HS, van der Marel GA. Discovery of
Straten NC, Xie D, Webb ML. Small molecule US6583179B2. 2003. selective luteinizing hormone receptor agonists
biaryl FSH receptor agonists. Part 1: Lead discovery 182. Dias JA, Campo B, Weaver BA, Watts J, Kluetzman K, using the bivalent ligand method. ChemMedChem.
via encoded combinatorial synthesis. Bioorg Med Thomas RM, Bonnet B, Mutel V, Poli SM. Inhibition 2009;4(7):1189–1195.
Chem Lett. 2004;14(7):1713–1716. of follicle-stimulating hormone-induced pre- 194. Bonger KM, Hoogendoorn S, van Koppen CJ,
168. Guo T, Adang AE, Dong G, Fitzpatrick D, Geng P, Ho ovulatory follicles in rats treated with a nonsteroidal Timmers CM, Overkleeft HS, van der Marel GA.
KK, Jibilian CH, Kultgen SG, Liu R, McDonald E, negative allosteric modulator of follicle-stimulating Synthesis and pharmacological evaluation of di-
Saionz KW, Valenzano KJ, van Straten NC, Xie D, hormone receptor. Biol Reprod. 2014;90(1):19. meric follicle-stimulating hormone receptor an-

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/911/5047286 by Research 4 Life user on 22 November 2018


Webb ML. Small molecule biaryl FSH receptor 183. Wortmann L, Cleve A, Muhn HP, Langer G, Schrey tagonists. ChemMedChem. 2009;4(12):2098–2102.
agonists. Part 2: Lead optimization via parallel A, Kuhne R, Menzenbach B, Koppitz M, Kosemund 195. Bonger KM, Hoogendoorn S, van Koppen CJ,
synthesis. Bioorg Med Chem Lett. 2004;14(7): D, inventors; Bayer Schering Pharma Aktiengesell- Timmers CM, van der Marel GA, Overkleeft HS.
1717–1720. schaft, assignee. Acyltryptophanols for fertility Development of selective LH receptor agonists by
169. Magar S, Goutopoulos A, Liao Y, Schwarz M, Russell control. Patent WO2007017289A2. 2005. heterodimerization with a FSH receptor antagonist.
TJ, inventors; Applied Research Systems Ars Holding 184. Failli A, Heffernan GD, Santilli AA, Quagliato DA, ACS Med Chem Lett. 2010;2(1):85–89.
NV., assignee. Piperazine derivatives and methods of Coghlan RD, Andrae PM, Croce SC, Shen ES, Trybulski 196. Newton CL, Katz AA, Millar RP. Fundamental
use. Patent WO2004031182A1. 2002. EJ, inventors; Wyeth, assignee. Pyrrolobenzodiaze- pharmacological mechanisms. Basic Clin Pharmacol
170. Timmers CM, Karstens WF, Grima PPM, inventors; pine pyridine carboxamides and derivatives as Toxicol. 2014;115:250–284.
NV Organon, assignee. 4-Phenyl-5-oxo-l,4,5,6,7,8- follicle-stimulating hormone receptor antagonists. 197. Pinilla L, Aguilar E, Dieguez C, Millar RP, Tena-
hexahydroquinoline derivatives as medicaments for Patent WO2006135687A1. 2005. Sempere M. Kisspeptins and reproduction: physi-
the treatment of infertility. Patent WO2006117370A1. 185. van Straten NC, Schoonus-Gerritsma GG, van ological roles and regulatory mechanisms. Physiol
2006. Someren RG, Draaijer J, Adang AE, Timmers CM, Rev. 2012;92(3):1235–1316.
171. Poveda PMG, Karstens WFJ, Timmers CM, in- Hanssen RG, van Boeckel CA. The first orally active 198. Roseweir AK, Millar RP. Kisspeptin antagonists. Adv
ventors; Organon NV, assignee. 4-Phenyl-5-oxo- low molecular weight agonists for the LH receptor: Exp Med Biol. 2013;784:159–186.
1,4,5,6,7,8-hexahydroquinoline derivatives for the thienopyr(im)idines with therapeutic potential for 199. Newton CL, Anderson RC, Millar RP. Therapeutic
treatment of infertility. Patent US8022218B2. 2011. ovulation induction. ChemBioChem. 2002;3(10): neuroendocrine agonist and antagonist analogs of
172. Gerrits MG, Kramer H, el Galta R, van Beerendonk 1023–1026. hypothalamic neuropeptides as modulators of the
G, Hanssen R, Abd-Elaziz K, Klipping C, Duijkers I, 186. Moore S, Jaeschke H, Kleinau G, Neumann S, hypothalamic-pituitary-gonadal axis. Endocr Dev.
Stoch SA. Oral follicle-stimulating hormone agonist Costanzi S, Jiang JK, Childress J, Raaka BM, Colson A, 2016;30:106–129.
tested in healthy young women of reproductive age Paschke R, Krause G, Thomas CJ, Gershengorn MC. 200. Abbara A, Jayasena CN, Christopoulos G,
failed to demonstrate effect on follicular develop- Evaluation of small-molecule modulators of the Narayanaswamy S, Izzi-Engbeaya C, Nijher GM,
ment but affected thyroid function. Fertil Steril. luteinizing hormone/choriogonadotropin and Comninos AN, Peters D, Buckley A, Ratnasabapathy
2016;105(4):1056–1062.e4. thyroid stimulating hormone receptors: structure- R, Prague JK, Salim R, Lavery SA, Bloom SR, Szigeti M,
173. van de Lagemaat R, van Koppen CJ, Krajnc-Franken activity relationships and selective binding patterns. Ashby DA, Trew GH, Dhillo WS. Efficacy of
MA, Folmer BJ, van Diepen HA, Mulders SM, J Med Chem. 2006;49(13):3888–3896. kisspeptin-54 to trigger oocyte maturation in
Timmers CM. Contraception by induction of 187. Jäschke H, Neumann S, Moore S, Thomas CJ, Colson women at high risk of ovarian hyperstimulation
luteinized unruptured follicles with short-acting low AO, Costanzi S, Kleinau G, Jiang JK, Paschke R, Raaka syndrome (OHSS) during in vitro fertilization (IVF)
molecular weight FSH receptor agonists in female BM, Krause G, Gershengorn MC. A low molecular therapy. J Clin Endocrinol Metab. 2015;100(9):
animal models. Reproduction. 2011;142(6):893–905. weight agonist signals by binding to the trans- 3322–3331.
174. Gartrell BA, Tsao CK, Galsky MD. The follicle- membrane domain of thyroid-stimulating hor- 201. Young J, George JT, Tello JA, Francou B, Bouligand J,
stimulating hormone receptor: a novel target in mone receptor (TSHR) and luteinizing hormone/ Guiochon-Mantel A, Brailly-Tabard S, Anderson RA,
genitourinary malignancies. Urol Oncol. 2013;31(8): chorionic gonadotropin receptor (LHCGR). J Biol Millar RP. Kisspeptin restores pulsatile LH secretion
1403–1407. Chem. 2006;281(15):9841–9844. in patients with neurokinin B signaling deficiencies:
175. Danesi R, La Rocca RV, Cooper MR, Ricciardi MP, 188. Heitman LH, Oosterom J, Bonger KM, Timmers CM, physiological, pathophysiological and therapeutic
Pellegrini A, Soldani P, Kragel PJ, Paparelli A, Del Wiegerinck PH, Ijzerman AP. [3H]Org 43553, the implications. Neuroendocrinology. 2013;97(2):193–202.
Tacca M, Myers CE. Clinical and experimental ev- first low-molecular-weight agonistic and allosteric 202. George JT, Kakkar R, Marshall J, Scott ML, Finkelman
idence of inhibition of testosterone production by radioligand for the human luteinizing hormone RD, Ho TW, Veldhuis J, Skorupskaite K, Anderson
suramin. J Clin Endocrinol Metab. 1996;81(6): receptor. Mol Pharmacol. 2008;73(2):518–524. RA, McIntosh S, Webber L. Neurokinin B receptor
2238–2246. 189. Newton CL, Whay AM, McArdle CA, Zhang M, van antagonism in women with polycystic ovary syn-
176. Daugherty RL, Cockett AT, Schoen SR, Sluss PM. Koppen CJ, van de Lagemaat R, Segaloff DL, Millar drome: a randomized, placebo-controlled trial. J Clin
Suramin inhibits gonadotropin action in rat testis: RP. Rescue of expression and signaling of human Endocrinol Metab. 2016;101(11):4313–4321.
implications for treatment of advanced prostate luteinizing hormone G protein-coupled receptor 203. Prague JK, Roberts RE, Comninos AN, Clarke S,
cancer. J Urol. 1992;147(3):727–732. mutants with an allosterically binding small- Jayasena CN, Nash Z, Doyle C, Papadopoulou DA,
177. Stevis PE, Deecher DC, Lopez FJ, Frail DE. Phar- molecule agonist. Proc Natl Acad Sci USA. 2011; Bloom SR, Mohideen P, Panay N, Hunter MS,
macological characterization of soluble human FSH 108(17):7172–7176. Veldhuis JD, Webber LC, Huson L, Dhillo WS.
receptor extracellular domain: facilitated secretion 190. van de Lagemaat R, Timmers CM, Kelder J, van Neurokinin 3 receptor antagonism as a novel
by coexpression with FSH. Endocrine. 1999;10(2): Koppen C, Mosselman S, Hanssen RG. Induction of treatment for menopausal hot flushes: a phase 2,
153–160. ovulation by a potent, orally active, low molecular randomised, double-blind, placebo-controlled trial.
178. Beindl W, Mitterauer T, Hohenegger M, Ijzerman weight agonist (Org 43553) of the luteinizing Lancet. 2017;389(10081):1809–1820.
AP, Nanoff C, Freissmuth M. Inhibition of receptor/ hormone receptor. Hum Reprod. 2009;24(3): 204. Jayasena CN, Comninos AN, Stefanopoulou E,
G protein coupling by suramin analogues. Mol 640–648. Buckley A, Narayanaswamy S, Izzi-Engbeaya C,
Pharmacol. 1996;50(2):415–423. 191. van de Lagemaat R, Raafs BC, van Koppen C, Abbara A, Ratnasabapathy R, Mogford J, Ng N,
179. Freissmuth M, Boehm S, Beindl W, Nickel P, Timmers CM, Mulders SM, Hanssen RG. Prevention Sarang Z, Ghatei MA, Bloom SR, Hunter MS, Dhillo
Ijzerman AP, Hohenegger M, Nanoff C. Suramin of the onset of ovarian hyperstimulation syndrome WS. Neurokinin B administration induces hot
analogues as subtype-selective G protein inhibitors. (OHSS) in the rat after ovulation induction with a flushes in women. Sci Rep. 2015;5(1):8466.
Mol Pharmacol. 1996;49(4):602–611. low molecular weight agonist of the LH receptor 205. Prague JK, Dhillo WS. Treating hot flushes with a
180. Wrobel J, Green D, Jetter J, Kao W, Rogers J, Pérez compared with hCG and rec-LH. Endocrinology. neurokinin 3 receptor antagonist. Oncotarget. 2017;
MC, Hardenburg J, Deecher DC, López FJ, Arey BJ, 2011;152(11):4350–4357. 8(63):106153–106154.
Shen ES. Synthesis of (bis)sulfonic acid, (bis)ben- 192. Gerrits M, Mannaerts B, Kramer H, Addo S, Hanssen 206. Skorupskaite K, George JT, Veldhuis JD, Millar RP,
zamides as follicle-stimulating hormone (FSH) an- R. First evidence of ovulation induced by oral Anderson RA. Neurokinin 3 receptor antagonism
tagonists. Bioorg Med Chem. 2002;10(3):639–656. LH agonists in healthy female volunteers of reveals roles for neurokinin B in the regulation of

936 Anderson et al Peptide and Nonpeptide Gonadotropin Analogs Endocrine Reviews, December 2018, 39(6):911–937
REVIEW

gonadotropin secretion and hot flashes in post- 213. Arslan AA, Zeleniuch-Jacquotte A, Lundin E, Micheli Current Affiliation: R. P. Millar and C. L. Newton’s
menopausal women. Neuroendocrinology. 2018; A, Lukanova A, Afanasyeva Y, Lenner P, Krogh V, current affiliation is Biomedical Sciences, College of Medicine
106(2):148–157. Muti P, Rinaldi S, Kaaks R, Berrino F, Hallmans G, and Veterinary Medicine, University of Edinburgh, Edinburgh
207. Zhu LL, Blair H, Cao J, Yuen T, Latif R, Guo L, Toniolo P. Serum follicle-stimulating hormone and EH8 9XD, United Kingdom.
Tourkova IL, Li J, Davies TF, Sun L, Bian Z, Rosen C, risk of epithelial ovarian cancer in postmenopausal
Correspondence and Reprint Requests: Robert P.
Zallone A, New MI, Zaidi M. Blocking antibody to Millar, PhD, Centre for Neuroendocrinology and Department
women. Cancer Epidemiol Biomarkers Prev. 2003;
the b-subunit of FSH prevents bone loss by of Physiology, Faculty of Health Sciences, University of Pre-
inhibiting bone resorption and stimulating bone 12(12):1531–1535. toria, P.O. Box 2034, Pretoria 0001, South Africa. E-mail: bob.
synthesis. Proc Natl Acad Sci USA. 2012;109(36): 214. Zheng W, Lu JJ, Luo F, Zheng Y, Feng Y, Felix JC, millar@up.ac.za.
14574–14579. Lauchlan SC, Pike MC. Ovarian epithelial tumor Disclosure Summary: R.A.A. and R.P.M. undertake
208. Liu P, Ji Y, Yuen T, Rendina-Ruedy E, DeMambro VE, growth promotion by follicle-stimulating hormone consultancy work for KaNDy Therapeutics Ltd. The
Dhawan S, Abu-Amer W, Izadmehr S, Zhou B, Shin and inhibition of the effect by luteinizing hormone. remaining authors have nothing to disclose.

Downloaded from https://academic.oup.com/edrv/article-abstract/39/6/911/5047286 by Research 4 Life user on 22 November 2018


AC, Latif R, Thangeswaran P, Gupta A, Li J, Shnayder Gynecol Oncol. 2000;76(1):80–88.
V, Robinson ST, Yu YE, Zhang X, Yang F, Lu P, Zhou 215. Mariani S, Salvatori L, Basciani S, Arizzi M, Franco G,
Y, Zhu LL, Oberlin DJ, Davies TF, Reagan MR, Brown Petrangeli E, Spera G, Gnessi L. Expression and Abbreviations
A, Kumar TR, Epstein S, Iqbal J, Avadhani NG, New cellular localization of follicle-stimulating hormone 7TM, seven transmembrane; aa, amino acid; ART, assisted
MI, Molina H, van Klinken JB, Guo EX, Buettner C, receptor in normal human prostate, benign pros- reproductive technology; CHO, Chinese hamster ovary; CTP,
Haider S, Bian Z, Sun L, Rosen CJ, Zaidi M. Blocking carboxy-terminal peptide; ECD, extracellular N-terminal
tatic hyperplasia and prostate cancer. J Urol. 2006;
FSH induces thermogenic adipose tissue and re- domain; ECL, extracellular loop; FcRn, neonatal Fc receptor;
175(6):2072–2077, discussion 2077.
duces body fat. Nature. 2017;546(7656):107–112. FSHR, FSH receptor; GnRHR, gonadotropin-releasing hor-
216. Choi JH, Wong AS, Huang HF, Leung PC. Gonad-
209. Zaidi M, New MI, Blair HC, Zallone A, Baliram R, mone receptor; GPCR, G protein–coupled receptor; hCG,
Davies TF, Cardozo C, Iqbal J, Sun L, Rosen CJ, Yuen otropins and ovarian cancer. Endocr Rev. 2007;28(4): human chorionic gonadotropin; hCGba, human chorionic
T. Actions of pituitary hormones beyond traditional 440–461. gonadotropin b-subunit fused to the N-terminus of the
targets. J Endocrinol. 2018;237(3):R83–R98. gonadotropin a-subunit; HH, hypogonadotropic hypo-
210. Kumar TR. Extragonadal actions of FSH: a critical gonadal; hMG, human menopausal gonadotropin; HPG,
need for novel genetic models. Endocrinology. 2018; Acknowledgments hypothalamic-pituitary-gonadal; LHCGR/LHR, LH receptor;
159(1):2–8. Financial Support: The research of R.C.A., C.L.N., and LMW, low molecular weight; LRR, leucine-rich repeat; MSD,
211. Meethal SV, Smith MA, Bowen RL, Atwood CS. The R.P.M. from the Centre for Neuroendocrinology is supported Merck Sharpe & Dohme; NAM, negative allosteric modu-
gonadotropin connection in Alzheimer’s disease. by grants from the University of Pretoria, South Africa; lator; OHSS, ovarian hyperstimulation syndrome; PAM,
Endocrine. 2005;26(3):317–326. National Research Foundation (NRF), South Africa; Tech- positive allosteric modulator; PCOS, polycystic ovarian syn-
212. Barron AM, Verdile G, Martins RN. The role of nology Innovation Agency (TIA), South Africa; and National drome; PEG, polyethylene glycol; PLC, phospholipase C;
gonadotropins in Alzheimer’s disease: potential Health Laboratory Services (NHLS) Trust, South Africa. The PMSG, pregnant mare’s serum gonadotropin; PO, orally; SC,
neurodegenerative mechanisms. Endocrine. 2006; research of R.A.A. is supported by a grant from the Medical subcutaneous(ly); THQ, tetrahydroquinoline; TM, trans-
29(2):257–269. Research Council (MRC), United Kingdom. membrane; TSHR, TSH receptor; TZD, thiazolidinone.

doi: 10.1210/er.2018-00052 https://academic.oup.com/edrv 937

Você também pode gostar