You are on page 1of 11

NEW DRUG CLASSES

New drug classes

Gonadotropin-releasing-hormone-receptor antagonists

Judith A F Huirne, Cornelis B Lambalk

Pulsatile gonadotropin-releasing hormone (GnRH) stimulates the pituitary secretion of both luteinising hormone (LH)
and follicle-stimulating hormone (FSH) and thus controls the hormonal and reproductive function of the gonads.
Blockade of GnRH effects may be wanted for a variety of reasons—eg, to prevent untimely luteinisation during
assisted reproduction or in the treatment of sex-hormone-dependent disorders. Selective blockade of LH/FSH
secretion and subsequent chemical castration have previously been achieved by desensitising the pituitary to
continuously administered GnRH or by giving long-acting GnRH agonists. Only recently have GnRH-receptor
antagonists, that immediately block GnRH’s effects, been developed for clinical use with acceptable
pharmacokinetic, safety, and commercial profiles. In assisted reproduction, these compounds seem to be as effective
as established therapy but with shorter treatment times, less use of gonadotropic hormones, improved patient
acceptance, and fewer follicles and oocytes. All current indications for GnRH-agonist desensitisation may prove to be
indications for a GnRH antagonist, including endometriosis, leiomyoma, and breast cancer in women, benign prostatic
hypertrophy and prostatic carcinoma in men, and central precocious puberty in children. However, the best clinical
evidence so far has been in assisted reproduction and prostate cancer.

The hormone gonadorelin, also known as gonadotropin- brain and packaged into granules in the Golgi apparatus
releasing hormone or luteinising-hormone-releasing and then transported by the axons to the neuronal
hormone (GnRH, LHRH), induces both follicle- terminal, where it is released in a pulsatile fashion into the
stimulating hormone and LH secretion by the pituitary capillaries of the pituitary-portal circulation. GnRH
gonadotrope cells in a orderly way which is crucial for the mRNA has been found in the pituitary3 and in
control of gonadal function and normal ovarian cyclicity. extrapituitary tissue, including the placenta, ovary,
One logical consequence of the discovery of the aminoacid myometrium, endometrium, and prostate and blood
sequence of GnRH in 1967 was the development of mononuclear cells, indicating an autocrine/paracrine
synthetic agonists and antagonists. It was noted that role.4-6 The genes for human GnRH types I and II lie on
sustained stimulation of the pituitary with GnRH itself or chromosome 8 (8p11.2-p21)7 and chromosome 20
with a GnRH agonist caused desensitisation, by post- (20p13),8 respectively.
receptor mechanisms that are still not well understood.1
The result, not immediately but after some time, was a GnRH receptor
chemical hypophysectomy which was thought to be the The GnRH receptor (GnRH-r)9 (figure 1) is a member of a
indication for GnRH antagonists. At first it seemed that the family of receptors known as the rhodopsin-like G-protein-
development of clinically safe agents would be simple to coupled receptor family,10 which also includes the
achieve by changing just one or two aminoacids but it was thyrotropin-releasing hormone receptor. Most vertebrates
to take almost 30 years of trial and error with three or more have at least two types of GnRH receptor.9 The gene for
replacements, including sometimes the use of unnatural GnRH type I receptor lies on chromosome 4 (4q21.2),11
aminoacids, before antagonists with acceptable and the type II receptor is thought to be on chromosome
pharmacokinetic, safety, and commercial profiles were 1q.12 Cloning of a type II GnRH receptor in the marmoset
developed. Today these GnRH-antagonists are at an showed that only 41% is identical to the type I receptor and
advanced stage of clinical development. that, unlike the type I receptor, it has a carboxyl-terminal
In this review of the most commonly used GnRH tail, which is important for rapid desensitisation.12 In
antagonists we have made use of some information that has human beings, controversial data have been obtained for
so far been presented only in abstracts so some statements extrapituitary expression of mRNA for GnRH-r in ovary,
may need modification when the data are published in full. breast, endometrium, myometrium, placenta, prostate, and
testis and in the various cancers of these tissues and their
Physiological principles associated cell lines. Variable presence of the different
GnRH GnRH-r transcripts13 and different distribution of type I
GnRH is a decapeptide (panel 1) that was isolated and and type II receptors could have a role. The type II
characterised by the groups of A V Schally and receptor in mammals is more widely distributed than the
R C L Guillemin, the 1977 Nobel laureates. GnRH type I type I receptor. It is expressed throughout the brain,
is the classic reproductive neuroendocrine factor.2 It is including the pituitary and areas associated with sexual
synthesised in the cytoplasm of the diencephalon of the arousal, and also in diverse non-neural and reproductive
tissues, suggesting various functions.12
Lancet 2001; 358: 1793–803
GnRH signal transduction
Division of Reproductive Medicine, Department of Obstetrics and
GnRH binds to specific receptors on the gonadotrope
Gynaecology, Vrije Universiteit Medical Centre, PO Box 7057, cells in the anterior pituitary. In gonadotropes, GnRH
1007MB Amsterdam, Netherlands (J A F Huirne MD, C B Lambalk MD) receptor activation, after coupling to Gq s11 protein, leads
Correspondence to: Dr Cornelis B Lambalk to the generation of several second messengers, among
(e-mail cb.lambalk@azvu.nl) which are diacylglycerol and inositol-4,5-triphosphate.

THE LANCET • Vol 358 • November 24, 2001 1793

For personal use. Only reproduce with permission from The Lancet Publishing Group.
NEW DRUG CLASSES

Panel 1: Structural formulae of native GnRH and third-generation GnRH-antagonists


Name Aminoacid found at position:
1 2 3 4 5 6 7 8 9 10
GnRH-I pGlu His Trp Ser Tyr Gly Leu Arg Pro Gly-NH2
GnRH-II 1 2 3 4 His 6 Trp Tyr 9 10
Abarelix D-Ala D-Phe D-Ala 4 5 D-Asp 7 Lys(iPr) 9 D-Ala
Antarelix D-Nal D-Phe D-Pal 4 Phe D-Hcit 7 Lys(iPr) 9 D-Ala
Cetrorelix D-Nal D-Phe D-Pal 4 5 D-Cit 7 8 9 D-Ala
Ganirelix D-Nal D-Phe D-Pal 4 5 D-hArg 7 hArg 9 D-Ala
Iturelix D-Nal D-Phe D-Pal 4 NicLys D-NicLys 7 Lys(iPr) 9 D-Ala
Nal-Glu D-Nal D-Phe D-Pal 4 D-Glu D-Glu 7 8 9 D-Ala

The former leads to activation of protein kinase C and the post-receptor signalling is involved, true receptor loss
latter to the production of cyclic AMP and release of (down-regulation) having only an initial role.1
calcium ions from intracellular pools.14,15 Both events A clear dichotomy of GnRH and competitive antagonist
result in secretion and synthesis of LH and FSH. GnRH (in which antagonist blocks agonist-induced effects and
II selectively binds to the type II receptor and signalling is vice versa) has not been shown in any human extrapituitary
distinctly different from that in the type I receptor.12 The tissue that expresses the receptor.16 Why agonists and
FSH and LH releasing potency of mammalian GnRH I antagonists sometimes exert similar effects is not clear. If
and GnRH II, tested in Soay rams, showed that the FSH the effects seen with an antagonist result from direct
to LH ratio was higher after treatment with GnRH II than blockade of the receptor while a similar effect with the
with GnRH I.12 These two GnRHs and GnRH receptor agonist is achieved via desensitisation, one would expect
systems, along with different signalling pathways, provide antagonist effects to be mitigated by an agonist, but this
the potential for differential FSH and LH secretion.12 does not usually happen. On the other hand, more recent
The natural GnRH signal from the hypothalamus to data show that GnRH antagonist abolishes a dose-
the pituitary is episodic (every 1-4 h) and this ensures LH dependent antiproliferative effect of a GnRH agonist on
and FSH secretion. However, upon continuous human ovarian epithelial cells and in human granulosa-
stimulation with GnRH, via a long-acting agonist, there is luteal cells.4 The antagonist reverses GnRH induced
first a period of hypersecretion quickly followed by activation of mitogen activated protein (MAP) kinase
pituitary desensitisation and arrest of gonadotropin which is known to regulate cell growth and
secretion. This mechanism is still not clear except that differentiation.4,17

1·15 (15)

G Q M L P I S N N I A S C H N Q N Q E P S
N A S N A M NH2
L
P H L G F T H C S L N R
T A S V M L
I Q S F
Y A D V
L W G M S T Q C E S
Q E R K W Q S 7·32(302)
T F H W P D
2·65 V I
L T L L Y L Q A D F P V
S G (102) I C
N W K V Q Y N F Y W W H F N
K I R M G P G
2·51 SL A F F F I G F F
V
T (26) D L V L 3·32 V S T F L V L F
V T P K (121) S Y A
M V L F S C L Y P F L
F F I L L I W
L 2·50 S M 5·50 F I 6·50 T W N P
F T E
4·50 A
L (87) Y A (223) I P (282) C V C F
L S L N L
A F P (164) L G V L T D
A T A L M S F I F S P L
F M M Q G M L T A 7·50 I Y
N 1·50 T L
A S V V V I C F A (320) G Y
F (53) H I K S N V F
L L K L S L 3·50 A K T M S L
K L L L D R I I K L
(139) N
Q K S L F T T
W K A S L K
T Q I T T R L
K M K R
R
R P V A
K 3·58
L A L L
S (145) Q H R
E 3·58
D P
K L (147)
G K K P I
H
E L Q L N Q S K N N

Figure 1: Schematic representation of human GnRH-r


Receptor composed of single polypeptide chain, with seven hydrophobic transmembrane domains, extracellular amino terminus, and intracellular carboxy
terminus. Known glycosylation site is marked, and certain key functional residues are numbered.

1794 THE LANCET • Vol 358 • November 24, 2001

For personal use. Only reproduce with permission from The Lancet Publishing Group.
NEW DRUG CLASSES

In in-vitro studies with cell lines of various tumour


types, antiproliferative activity was shown by both GnRH neuron
types of analogue. In a cell line of a human kidney Competitive receptor
embryo antiproliferative effects of agonist and blocking
antagonist occurred only when expression of a high GnRH
affinity GnRH-r was induced, which strongly suggests GnRH GnRH antagonist
direct, specific, receptor-mediated effects.16 GnRH-r
stimulation of MAP kinases (MAPKs) in granulosa-
luteal cells of human beings confirmed the receptor FSH No intrinsic
mediation; additional studies showed that cellular LH activity
responses (proliferation, hormone or peptide secretion)
to MAPK stimulation are tissue specific and duration
dependent.17 GnRH analogues may interfere with the
LH FSH
signal transduction of growth factor receptors and
related oncogene products associated with tyrosine- Figure 2: Working mechanism of GnRH antagonists
kinase activity.15 So far, most studies have been done
on the assumption that there was only the GnRH type I generation the undesirable risk of oedema was
receptor. Cloning of the type II GnRH receptor eliminated by replacing the D-Arg at position 6 by
provides the opportunity to study the different affinity neutral D-ureidoalkyl aminoacids, to produce
and potency of analogues to the different types of compounds such as cetrorelix, iturelix, azaline B,
receptor and their signalling pathways and cellular ganirelix, abarelix, and antarelix23–29 (panels 1 and 2).
reactions. Differential expression of the GnRH type I
and type II receptors could have a role in the paradox Pharmacokinetics
of similar effects of both GnRH agonists and The pharmacokinetic properties with regard to
antagonists on proliferation in specific tumour cell maximum antagonist administration (Cmax) are linear
lines. Certain mammalian GnRH type I antagonists following single or multiple doses.30–32 The predominant
behave as agonists on GnRH type II receptors.12 The clearance is metabolic and hepatic.33 Absorption is the
antiproliferative effects of GnRH analogues on cell time-limiting step for elimination. Plasma protein
lines of these tumours are consistent with the activation binding averages 80% and the plasma half-life of a non-
of MAPK p38␣ by the type II receptor, which is known depot antagonist varies from 5 to 30 h after single
to be antiproliferative.18 subcutaneous administration of clinical doses and is
increased after multiple dose administration (panel 3).
LHRH antagonists Abarelix for injectable suspension allows single
Unlike the agonists, GnRH antagonists do not induce intramuscular administration every 28 days in patients
an initial stimulation of gonadotropin release, but cause with prostatic cancer.34–36
an immediate and rapid, reversible suppression of
gonadotropin secretion. The principal mechanism of Pharmacodynamics
action of GnRH antagonists is competitive receptor 6 h (range 4–24 h) after administration of a GnRH-
occupancy of GnRH-r (figure 2). antagonist plasma LH concentrations will have fallen by
70% (range 52–91%) and plasma FSH by 30% (range
Structural modifications 23–61%). The size and duration of this suppression are
The effects of a single aminoacid substitution may alter dose dependent.37,38 Complete reversal is achieved in
affinity and agonist activity via modification of a side- 24–72 h. Comparable suppression is seen after
chain that interacts with the binding pocket and/or by subcutaneous and intramuscular administration39 but
altering the conformation of the peptide. Whether a the intranasal route requires higher dosages.40 In women
ligand functions as agonist or antagonist is species with regular menstrual cycles, the LH suppressive effect
dependent and is determined by the structure of the is greatest at the time of ovulation.41 Any residual
second extracellular loop of the GnRH-r.19 gonadotropin in the serum after administration of an
The structural features of GnRH antagonists are antagonist in maximally suppressive doses suggests
reviewed elsewhere.20,21 The weak first-generation drugs either incomplete blockade of pituitary receptors or, and
were hydrophilic and contained replacements for His at more probably, non-GnRH-dependent gonadotropin
position 2 and for Trp at position 3. Inhibitory activity secretion. The percentage decline in LH secretion
increased after incorporation of a D-aminoacid at achieved by GnRH-antagonists is consistently higher
position 6 but increased histamine-releasing activity than the decline in FSH secretion,33,42–45 indicating the
resulted in anaphylactic reactions to the second- existence of some additional, non-GnRH-dependent
generation antagonists such as detirelix.22 In the third control mechanism.

Panel 2: GnRH antagonists launched or under investigation


Name Manufacturer Route Status
Abarelix Praecis (Amgen) IM Phase III, prostate cancer
Sanofi-Synthelabo Phase II, endometriosis
Antarelix Asta Medica SC Phase I, prostate cancer
Cetrorelix (Cetrotide) Asta Medica/Serono SC Approved IVF. Phase II, benign prostatic hyperplasia,
uterine myoma, prostate and ovarian cancer
Ganirelix acetate
(Orgalutran, Antagon) Organon SC Approved, female infertility
Iturelix (Antide) Serono SC Phase II, IVF
SC=subcutaneous, IM=intramuscular.

THE LANCET • Vol 358 • November 24, 2001 1795

For personal use. Only reproduce with permission from The Lancet Publishing Group.
NEW DRUG CLASSES

Panel 3: Pharmacokinetic indices following single and multiple dose administrations of GnRH antagonists
in (pre)clinical studies*
Compound M/F Dose (mg) Tmax Cmax T1/2 Refs
(h) (ng/mL) (h)
Single dose
Abarelix-depot Both 30–120 NA 7·5–38 >240 Wong118
Cetrorelix M 10 NA NA 20 Behre119
F 0·25 1·0 5·0 5·0 Duijkers31
F 1·0 1·0 21·2 9·4 Duijkers31
F 3·0 1·5 28·5 62·8 Erb120
Ganirelix F 0·25 1·1 14·8 12·8 Oberye32
Multiple dose†
Cetrorelix F 0·25 1·0 6·4 20·6 Duijkers31
F 1·0 1·0 21·1 77·4 Duijkers31
Ganirelix F 0·25 1·1 11·2 16·2 Oberye32
*Results of phase I studies, which studied recommended dose as applied in phase III trials, and published in peer-reviewed journals. †After multiple dosing (daily): Tmax=time of
maximum plasma concentration at steady state, Cmax=maximum plasma concentration at steady state, T1/2=terminal half-life.

Safety and tolerability Hernandez57 has hypothesised that GnRH antagonists


The third-generation antagonists have low histamine- interact with the mitotic programming of cells involved
releasing potency, and iturelix and azaline-B have lower in folliculogenesis, blastomere formation, and
histamine-releasing potencies than GnRH itself though endometrium development. Moreover, there is
neither agent is currently under clinical evidence that GnRH (and agonists) increase the
development.23,26,46 Toxicological studies have confirmed cleavage rate of bovine oocytes, an effect that is
the safety of the third-generation drugs. There is no abolished by addition of a GnRH antagonist.62 Although
evidence for irreversible drug-related atrophy of the inhibition of sperm-binding to the zona pellucida was
target organs or for mutagenic/clastogenic potential. inhibited by GnRH antagonists in vitro,63 lower
Influence on the early embryonic development of rats fertilisation rates were not found in phase III studies of
could not be demonstrated.30,47,48 Over 2000 patients antagonists.50–53,64 The lower implantation rates seen with
have now been treated with ganirelix, cetrorelix, or higher doses of antagonists in in-vitro fertilisation
abarelix, without evidence of anaphylactic reactions.49–53 (IVF)65 are probably related to impaired endometrial
Adverse effects of antagonists are largely the same as receptiveness rather than a direct embryonic effect
those with agonists, and are attributable to hypo- because cryopreserved embryos from these cycles
oestrogenism in women and hypoandrogenism in males, yielded normal pregnancy rates.66 Follicular growth was
including hot flushes, mood changes, headache, and also not influenced by the daily dose of GnRH
decreased libido. Prolonged exposure could lead to loss antagonist.67
of bone-mineral density to the same extent as the 2-8% Despite the reassurance to date, the safety GnRH
after 6 months seen with agonists. One common side- analogues in respect of human embryo development
effect is a local reaction at the injection site (redness and remains a priority. So far no increased risk of birth
pain), which usually resolves within an hour.52,53 Local defects or pregnancy wastage in pregnancies exposed to
skin reactions seem to be less common with antagonists daily low-dose GnRH antagonist therapy in the first
than they are with agonists. No significant changes in weeks of gestation have been recorded.68
serum chemistry or haematological indices have been
noted.48,54 Clinical experience to date indicates that Therapeutic applications
GnRH antagonists are safe and well tolerated. On pharmacological grounds the primary indications for
GnRH antagonists will be in any situation in which
Extrapituitary effects chemical gonadotropic hypophysectomy is required
The safety of GnRH analogues in respect of structures (figure 3):
such as the ovary, oocyte, and granulosa cell and the (1) Immediate blockade of the effect of gonadotropic
embryo has become a matter of debate since the hormones –eg, in IVF to prevent the normal midcycle
discovery of extrapituitary human GnRH-receptors. The rise in LH.
many paradoxical results in studies of ovarian (2) Indirectly, to block gonadal sex-hormone secretion
steroidogenesis may be caused by factors such as the via blockade of pituitary gonadotropin secretion. An
type, dose, and regimen of the analogues used, the antagonist can, theoretically, be given to patients with a
patient’s ovarian status, the ovarian cell types used and sex-hormone-dependent disease such as benign
the hormonal pretreatment of those cells, and the type prostatic hypertrophy and prostate carcinoma,
of in-vitro stimulation, besides methodological leiomyoma and endometriosis, or precocious puberty.
differences in the experiments and physiological (3) The treatment of cancer, based on evidence that
variation in receptor abundance and type.12, 55–57 GnRH antagonists exert direct negative effects on
So far, intrinsic direct effects of GnRH antagonists on growth of certain malignant tumours.
human ovarian steroidogenesis or cAMP production in The only two registered GnRH antagonists for IVF
vitro have not been demonstrated.58–60 An important, are cetrorelix (Cetrotide; Serono International) and
recent finding is the expression of GnRH and GnRH-r ganirelix (Orgalutran, Antagon; Organon). Both are
in the mouse embryo.61 Incubation of the murine approved only for use during IVF.
embryos with a GnRH agonist enhanced preimplan-
tation embryonic development whereas an GnRH Prevention of premature LH surge in IVF
antagonist completely blocked this development.61 The Placebo-controlled studies of GnRH agonists revealed
blockade could be reversed by a GnRH agonist. that in about 20% of women there was a premature LH

1796 THE LANCET • Vol 358 • November 24, 2001

For personal use. Only reproduce with permission from The Lancet Publishing Group.
NEW DRUG CLASSES

Level Effects Agonist


hCG
Days 21–24 of the
GnRH neuron preceeding cycle
Hypothalamic FSH
desensitisation GnRH agonist
GnRH GnRH antagonist
Day 2 or 3 Day 6
Antagonist of menses of FSH hCG or GnRH
Selective chemical
Pituitary Pituitary
hypophysectomy
Multiple dose FSH
GnRH antagonist

LH and FSH LH and FSH Day 7 or 8


Chemical of FSH
Gonadal
castration Day 2 or 3 GnRH
ovary testis
of menses antagonist hCG or GnRH
Single dose
Oestrogens Androgens
FSH

Figure 3: Effects of GnRH antagonists Figure 4: GnRH analogue treatment regimens in in-vitro
Schematic representation of effects on different levels of hypothalamic- fertilisation
pituitary-gonadal axis. Schematic representation of most commonly used regimen in phase III
trials.

increase that led to cancellation of the IVF cycle.69,70 significantly lower rate of pregnancy of 5%. This
Preventing this untimely physiological change would meta-analysis unfortunately included the study that
clearly be beneficial. Moreover, the IVF harvest is bigger compared a single-dose analogue regimen with different
with more embryos, allowing better selection so that, on gonadotropin starting dose as an additional variable.75
average, the outcome in terms of pregnancy rates is There is some concern that this may be a consequence
better.71 It takes time for a state of desensitisation to be of the currently advised treatment regimen. It has been
reached and, to start with, LH secretion actually suggested that the larger numbers of oocytes and
increases (flare-up). In an IVF setting, treatment with a embryos with agonists allow better selection,71 although
GnRH agonist usually begins in the midluteal phase of the numbers of good quality embryos do not seem to be
the menstrual cycle preceding the IVF treatment cycle; different. A direct adverse effect on the embryo cannot
this is the so-called long protocol (figure 4). With an yet be ruled out but is not likely. There are many
antagonist, immediate blockade of pituitary gonadotropin reports of pregnancies inadvertently exposed to a GnRH
secretion when premature luteinisation during IVF agonist without any adverse effect.56 In IVF treatment
stimulation is imminent seemed an obvious approach. the risk of embryonic exposure to an antagonist is
Several studies of dose and treatment schedules have minimal. Cetrorelix plasma and follicular fluid levels fell
been done,65,72,73 and two general approaches have significantly after hCG administration, and at minimally
emerged. The first is a single subcutaneous injection of a effective doses cetrorelix was not detectable during
large dose on about the eighth day of stimulation with ovum retrieval and embryo transfer.76 Nevertheless, only
gonadotropins (figure 4). The alternative is five or six long-term follow-up of the children born after IVF
daily injections of a small dose from about day 6 of procedures in which these new drugs have been used
stimulation until the day that hCG for final oocyte can be conclusive. Follow-up of neonatal outcome after
maturation is given (figure 4, panel 4). pregnancies established in large phase II/III trials (474
Panel 4 summarises suggested daily dosages based on pregnancies after ganirelix, 134 after a long agonist
extensive dose-finding reports.65,72–74 The next step was to protocol,77 and 227 children after centrorelix68) did not
establish whether a GnRH antagonist is at least as show any negative effect.
effective as a GnRH agonist as the established reference No major side-effects have been reported. Minor side-
medication. So far, four such studies of repeated effects were limited to pain and redness at the injection
antagonist injections have been reported50–53 and one with site. This happened with both types of analogue but was
a single-dose regimen64 (panel 5). less common with the antagonist.52,53 The smaller number
With repeated injections (panel 6), a consistent finding of injections and the reduction in the duration of IVF
is that duration of treatment with gonadotropins is treatment are welcomed by patients and this could be the
shortened by 1-2 days. With an antagonist slightly fewer
follicles are seen at the time of hCG injection so the Panel 4: Prevention of premature LH surge; minimal
number of recovered oocytes tends to be lower. A likely effective dose of GnRH antagonists in patients
explanation is that an agonist suppresses the natural cycle undergoing in-vitro fertilisation
follicular recruitment initiated by the intercycle FSH
Compound Dose LH-surge Refs
rise50–53 so that longer treatment with gonadotropins is
required, which allows more follicles to enter the growing Cetrorelix 3 mg single dose 0/34 Olivennes72
phase. No significant difference was found with respect Cetrorelix 0·25 mg/day 0/30 Albano73
to percentages of metaphase II oocytes, fertilisation rates, Ganirelix 0·25 mg/day 1/69 Ganirelix group65
and number of good quality embryos.52,53 Iturelix 0·5mg/day 0/32 Serono74
Pregnancy rates were high in both groups in all Single subcutaneous injection on stimulation day (SD) 8 or until oestradiol level
four studies but in every one the rate was lower in 肁400 pg/mL.72 Multiple injections at one per day subcutaneous, from stimulation
day 6 or 7 up to hCG administration day.65,73,74 LH-surge defined as LH >10 IU/L
the antagonist group. A meta-analysis of the five after first antagonist injection.
randomised trials (panel 5), shows an overall

THE LANCET • Vol 358 • November 24, 2001 1797

For personal use. Only reproduce with permission from The Lancet Publishing Group.
NEW DRUG CLASSES

Panel 5: GnRH antagonist versus GnRH agonists: treatment schedules of phase III trials in in-vitro fertilisation
Antagonist group Agonist group
Drug dose FSH dose Drug Dose (mg) FSH dose No Refs
(mg) (units/day) (units/day)
Multiple dosing regimen
Cetrorelix 0·25 150 HMG Buserelin 0·6 150 HMG 293 Albano50
Ganirelix 0·25 150 rFSH Buserelin 0·6–1·2 150 rFSH 701 EUR Orgalutran study51
Ganirelix 0·25 150 rFSH Triptorelin 0·1 150 rFSH 337 EME Orgalutran study52
Ganirelix 0·25 225 rFSH Leuprolide 1–0·5 225 rFSH 297 Nam Ganirelix study53
Single dose regimen
Cetrorelix 3·0 150 HMG Triptorelin 3·75 225 HMG 154 Olivennes64
Multiple antagonist dosing one subcutaneous injection daily from stimulation day 6,7 or 8 up to HCG day; single dosing one subcutaneous injection on stimulation day 7 or if
oestradiol 500 pg/mL or follicle >14 mm. For agonists multiple dosing with daily subcutaneous or intranasal (buserelin) administration, starting midluteal phase of menstrual cycle
preceding ovarian stimulation cycle (long protocol). Single dosing regimen is one triptorelin depot injection during preceding luteal phase.

most important reason why GnRH antagonists will find a Other indications: female reproductive
therapeutic role in IVF clinics. So far, however, no large system
and moreover no blinded comparisons of GnRH agonists On theoretical grounds, many future indications are
and antagonists with pregnancy outcome as the endpoint feasible for GnRH antagonistic compounds (panel 7).
have been done. A further advantage of antagonists is
that less FSH is needed. Ovarian hyperstimulation syndrome
Antagonist blockade allows immediate reversal of Currently, ovarian hyperstimulation syndrome is
pituitary gonadotropin secretion. This means that in IVF managed by electrolyte and fluid administration, rest,
ovulatory ripening can be triggered via endogenous paracentesis, and, in severe forms, respiratory support.
gonadotropins by using a GnRH agonist.78,79 One The patient may benefit from immediate blockade of
advantage could be prevention of ovarian endogenous LH secretion. High doses of a GnRH
hyperstimulation syndrome, which is thought to result at antagonist may be used for this purpose.84
least in part from the prolonged LH effect of hCG.80
Finally, the use of GnRH antagonist blockade of Polycystic ovary syndrome
premature luteinisation can be used in IVF with very low Polycystic ovary syndrome (PCOS) is characterised by
or without any hormonal stimulation,81 lower risk of oligomenorrhoea, hyperandrogenism, and cystic
overstimulation, and simplification of the procedure.82 appearance of the ovaries. Raised LH levels are thought
A possible disadvantage of an antagonist in IVF is its to be responsible for the high androgen levels that
narrow therapeutic range with the currently advised adversely affect the development of follicles.
doses for repeated injections. Patient compliance needs Theoretically, blockade of endogenous LH secretion by
to be high because there is a risk of premature LH an antagonist combined with ovulation induction could
secretion if an injection is missed. result in improved follicular development. The abnormal
Another disadvantage is the unpredictable timing of LH secretion is thought to result from an abnormal
the start of the IVF procedure, which begins with the endogenous GnRH signal from the hypothalamus.85
administration of FSH on day 3 of the woman’s cycle Studies in monkeys showed that blockade of the
and so depends on how regular her menstruation is. This endogenous GnRH signal can be competed with by
problem may be solved by pretreatment with an oral exogenously administered pulsatile GnRH which restored
contraceptive.83 gonadotropin secretion and ovulation.86 So far the same

Panel 6: In-vitro fertilisation results in trials comparing repeated dose GnRH agonist and GnRH antagonist
Albano50 EUR Orgalutran Study51 EME Orgalutran Study52 NAM Ganirelix Study53
Cetrorelix Buserelin Ganirelix Buserelin Ganirelix Triptorelin Ganirelix Leuprolide
Patients (ITT) 188 85 463 237 226 109 198 99
LH surge (% ITT) 1·6 1·2 2·8 1·3 0·4 0 1·0 0
Days analogue (median, ITT) 5·7* 26·6* 5 26 5 26 4 22
Days FSH (median) 10·6* 11·4* 9 10 9 11 8 10
Total FSH (median, IU) 1770* 1920* 1500 1800 1350 1800 1800 2025
Follicles >10 mm hCG day (mean, ITT) 10·1* 12·3* 10·7 11·8 10·1 10·7 12·3 13·9
E2 on hCG day (median, pg/mL) 1625* 2082* 1190 1700 1090 1370 2001 2768
Oocytes (mean ITT) 7·4 9·6 8·7 9·7 7·9 9·6 11·6 14·1
Fertilisation rate (%) 53·6 52·9 62·1 62·1 64 64·9 62·4 61·9
Embryos (mean no, ITT) 4·0 4·1 6·0 7·1 4·0 4·7 6·9 8·2
Embryos transferred (mean no) 2·2 2·2 2·2 2·2 2·4 2·6 2·9 2·8
Implantation rate (IR) NA NA 15·7 21·8 22·9 22·9 21·1 26·1
Vital PR/ITT (%) 22·3 25·9 21·8 28·3 32·3 36·0 35·4 38·4
Ongoing PR/ITT (%) 18·1† 22·4† 20·3 25·7 31·0 33·9 30·8 36·4
OHSS (% ITT) 1·1 5·9 2·4 5·9 1·8 0·9 6·1 2·0
ITT=intent-to-treat group, is the number of patients who received at least one dose of LHRH analogue or FSH. E2=oestradiol. OHSS=ovarian hyperstimulation syndrome.
* Mean, calculated for the patients who reached the day of HCG. Fertilisation rate=% 2PN oocytes per number of oocytes incubated, IR=number of sacs/transferred embryo (patients
with embryo transfer). Vital pregnancy=intrauterine pregnancy with proof of at least one vital fetus as assessed by ultrasound scan 5–6 weeks after embryo transfer. Ongoing
pregnancy was defined as an intrauterine pregnancy with proof of at least one vital fetus as assessed by ultrasound at 12–16 weeks after transfer. † number of deliveries

1798 THE LANCET • Vol 358 • November 24, 2001

For personal use. Only reproduce with permission from The Lancet Publishing Group.
NEW DRUG CLASSES

Treatment with GnRH agonists of patients with


Panel 7: Other indications (being explored or advanced metastatic breast cancer has been shown to
future) for GnRH antagonists be effective and safe though in clinical practice
Female reproductive system antioestrogenic therapy by tamoxifen prevails. Human
Ovarian hyperstimulation syndrome breast cancer does express the GnRH receptor
Polycystic ovary syndrome with moderately high-affinity binding, inducing
Endometriosis inhibition of cell proliferation in cultured cell lines.96,97
Leiomyoma Although some studies with agonists show inhibition,
Ovarian cancer others found no effect.98 Unfortunately, at the moment
Endometrial cancer there are no reports or studies underway with regard
Breast cancer to the clinical application of GnRH antagonists in breast
Mammography cancer.
Male reproductive system
Benign prostate hypertrophy Mammography
Prostate cancer Injection of a GnRH antagonist before screening
Male contraception mammography, to achieve an antioestrogenic effect on
Paediatrics the breast, could permit better resolution.91
Precocious puberty
Delay of puberty in children with small stature Other indications: male reproductive system
Miscellaneous
Benign prostatic hypertrophy
Pituitary adenoma In one clinical study injections of cetrorelix (5 mg twice
Gonadal protection during radiation or chemotherapy daily for 2 days and then 1 mg twice daily for 2 months)
Paraphilias resulted in a rapid 27% reduction in prostate volume and
Direct antitumour effects a 53% reduction of symptoms with improvement in
Drug targeting quality of life.99 During follow-up urinary symptoms
usually improved and sexual function was enhanced.
However, phase III comparative studies have yet to
combination in PCOS patients normalised gonadotropin be done.
secretion but failed to induce ovulation.87
Prostate cancer
Endometriosis GnRH agonists have advantages over orchidectomy and
With endometriosis, an oestrogen-dependent disorder, oestrogens and were first-choice drugs, mostly initially
treatment with a GnRH antagonist is likely to be just as combined with antiandrogens, in the hormonal
effective as treatment with a GnRH agonist. Phase III treatment of prostate cancer.36 However, the initial
trials comparing abarelix with leuprolide are underway.88 androgen surge as result of the LH flare may cause
significant complications such as ureteric obstruction
Leiomyoma and severe pain from bone metastasis. This effect does
Treatment of leiomyoma with GnRH antagonists, by not occur with GnRH antagonists.36,100–104 With cetrorelix
daily injections or by depot injections, results in shrinkage 1–2 mg per day after an initial loading of 10 mg over 2–5
of the leiomyoma by 30–50% within 4–8 weeks.89,90 The days, castrate testosterone values are sustained with
avoidance of the flare-up in gonadotropin secretion seen reductions in prostate specific antigen (PSA), regression
with the GnRH agonist may explain the rapid reduction of metastases, and rapid improvement of disseminated
in fibroid size. Autocrine/paracrine effects mediated via prostate cancer.100 Immediate and sustained suppression
myometrial GnRH receptors may also be involved. of tumour is achieved, with a rapid decline in prostate
volume compared with conventional treatments.101
Endometrial ablation Abarelix-depot 100 mg by intramuscular injection every
Among other future benign gynaecological indications are 28 days with an additional injection on day 15 causes
prevention of endometrial proliferation – eg, with difficult rapid and profound reduction in androgen,
dysfunctional bleeding and as pretreatment before gonadotropin and PSA in men with various stages of
hysteroscopic surgery.91 prostate cancer, superior by comparison with the GnRH
agonists up to 29 days36,100–105 and comparable during 85
Gynaecological cancers to 169 days.104,105 An additional advantage of GnRH
The original rationale for a GnRH agonist in the antagonists by comparison with GnRH agonists is the
treatment of ovarian cancer was to block the endogenous long-term (up to 169 days) suppression of FSH,27,104
LH and FSH secretion which were thought to stimulate since FSH has been implicated as a potential growth
tumour growth. However, a role for gonadotropins in factor for prostate cancer.106 At 12 weeks the overall
ovarian epithelial cancer remains controversial. Clinical response to abarelix-depot is 74% (9% complete
studies have recorded objective responses (9% to 26%) response, 22% partial, 43% stable disease),102 median
with GnRH agonists, and there is evidence for a direct PSA and testosterone levels declined with 90% of
antiproliferative effects of GnRH analogues.92 A role for baseline, cancer pain was reduced and narcotic use
GnRH antagonists has yet to be established but studies declined with improvement of urinary complaints.107 A
are underway.93 New Drug Application has been submitted to the US
Favourable effects of GnRH agonists in patients with Food and Drug Administration, and has been filed for
recurrent endometrial cancer have been reported.94 These approval in Europe for its use in prostatic cancer.
favourable effects could be related to reduction in
oestrogen output but direct inhibitory effects are likely Male contraception
also. GnRH agonists and antagonists inhibit proliferation Weekly 200 mg testosterone injections cause reversible
in endometrial cancer cell lines,95 but no clinical data on oligozoospermia in 98% of men, and resulting pregnancy
the effects of antagonists are available. rates are lower than those for the oral contraceptive for

THE LANCET • Vol 358 • November 24, 2001 1799

For personal use. Only reproduce with permission from The Lancet Publishing Group.
NEW DRUG CLASSES

women.108 However, reported side-effects are weight action of GnRH analogs on myometrial smooth muscle cells and
gain, oily skin, and acne and if the dose is lowered interaction with ovarian steroids in vitro. J Clin Endocrinol Metab
1996; 81: 3215–21.
contraceptive efficacy declines. Azoospermia can be
6 Cheng KW, Nathwani PS, Leung PC. Regulation of human
achieved with lower dosages (25 mg weekly) if combined gonadotropin-releasing hormone receptor gene expression in placental
with a GnRH antagonist.109 Low sperm counts can be cells. Endocrinology 2000; 141: 2340–49.
achieved with an initial combination of a daily sub- 7 Yang-Feng TL, Seeburg PH, Francke U. Human LHRH gene is
cutaneous antagonist (Nal Glu) and weekly low-dose located on short arm of chromosome 8 (region 8p 11.2-p21). Somat
testosterone followed by low-dose testosterone alone.108 Cell Molec Genet 1986; 12: 95–100.
8 White RB, Eisen JA, Kasten TL, Russel DF. Second gene for
The newly developed long-acting GnRH antagonist gonadotropin-releasing hormone in humans. Proc Natl Acad Sci USA
preparations could bring us one step closer to the goal of 1998; 95: 305–09.
a satisfactory male hormonal contraceptive. 9 Sealfon SC, Weinstein H, Millar RP. Molecular mechanisms of ligand
interaction with the gonadotropin- releasing hormone receptor. Endocr
Other indications Rev 1997; 18: 180–205.
10 Probst WC, Snyder LA, Schuster DI, Brosius J, Sealfon SC.
Puberty disorders Sequence alignment of the G-protein coupled receptor superfamily.
GnRH agonists are the established treatment of DNA Cell Biol 1992; 11: 1–20.
central precocious puberty. They act via desensitisation 11 Leung PC, Squire J, Peng C, Fan N, Hayden MR, Olofsson JI.
of the pituitary to prematurely released endogenous Mapping of the gonadotropin-releasing hormone (GnRH) receptor
GnRH. There are no clinical data on GnRH antagonists gene to human chromosome 4q21.2 by fluorescence in situ
hybridization. Mamm Genome 1995; 6: 309–10.
in this context, though animal experiments are en-
12 Millar R, Lowe S, Conklin D, et al. A novel mammalian receptor for
couraging.110Another future indication may be the the evalutionarily conserved type II GnRH. Proc Natl Acad Sci USA
delay of normal puberty in children with short stature 2001; 98: 9636–41.
but so far this has only been tested with a GnRH 13 Kottler ML, Lorenzo F, Bergametti F, Commercon P, Souchier C,
agonist.111 Counis R. Subregional mapping of the human gonadotropin-releasing
hormone receptor (GnRH-r) gene to 4q between the markers D45 and
D45S409. Hum Genet 1995; 96: 477–80.
Miscellaneous 14 Stojilkovic SS, Reinhart J, Catt KJ. Gonadotropin-releasing hormone
Among other possible indications are the use of GnRH receptors: structure and signal transduction pathways. Endocr Rev
antagonists in the hormonal treatment of pituitary 1994; 15: 462–99.
gonadotropin producing tumours,112,113 and the protection 15 Kaiser UB, Conn PM, Chin WW. Studies of gonadotropin-releasing
of the gonads in a similar way as agonists may act hormone (GnRH) action using GnRH receptor-expressing pituitary
cell lines. Endocr Rev 1997; 18: 46–70.
during radiation or chemotherapy.114,115 Finally, GnRH 16 Emons G, Muller V, Ortmann O, Schulz KD. Effects of LHRH-
antagonists may be useful as pharmacological agents in analogues on mitogenic signal transduction in cancer cells. J Steroid
treatment of hypersexuality and paraphilias.116 Biochem Mol Biol 1998; 65: 199–206.
17 Kang SK, Tai CJ, Nathwani PS, Choi KC, Leung PCK. Stimulation
Future prospects of mitogen-activated protein kinase by gonadotropin-releasing
hormone in human granulosa-luteal cells. Endocrinology 2001; 142:
Current GnRH antagonists seem to be safe and effective 671–79.
additions to the hormonal armamentarium for several 18 Neil JD, Duck LW, Wellers JC, Musgrove LC. A gonadotropin-
conditions though published clinical evidence so far is releasing hormone (GnRH) receptor specific for GnRH II in primates.
mainly in IVF and prostate cancer. The hope remains that Biochem Biophys Res Commun 2001; 282: 1012–18.
all current indications for GnRH agonist therapy will also 19 Sun YM, Flanagan CA, Illing N, Ott TR, Sellar R, Fromme BJ, et al.
A chicken gonadotropin-releasing hormone receptor that confers
become applications for GnRH antagonists. With respect agonist activity to mammalian antagonists: identification of D-Lys6 in
to direct anti-tumour effects we do not yet know whether the ligand and extracellular loop two of the receptor as determinants.
agonists or antagonists will prevail. More studies, looking J Biol Chem 2001; 276: 7754–61.
at tissue-specific distribution of GnRH receptor types, 20 Haviv F, Bush EN, Knittle J, Greer J. LHRH antagonists. Pharm
their analogue affinity, potency, and specific signalling Biotechnol 1998; 11: 131–49.
pathway activation are needed. In all situations sound 21 Schally AV. Lutenizing hormone-releasing hormone analogs:
their impact on the control of tumorigenesis. Peptides 1999; 20:
comparative studies, and publication of them, are 1247–62.
essential. Expected long-term developments include non- 22 Karten MJ. An overview of GnRH antagonist development: two
peptide orally active GnRH antagonists20,117 and targeted decades of progress. In: Crowley WF, Conn PM, eds. Modes of
drug administration for some cancers.21 Finally, licensed actions of GnRH and GnRH analogs. Amsterdam: Elsevier, 1992:
277–97.
GnRH antagonists are ideal research tools for further
23 Bajusz S, Kovacs M, Gazdag M, et al. Highly potent antagonists of
exploration of the pathophysiology of the human luteinizing hormone-releasing hormone free of edematogenic effects.
reproductive system. Proc Natl Acad Sci USA 1988; 85: 1637–41.
24 Ljungqvist A, Feng DM, Hook W, et al. Antide and related
antagonists of luteinizing hormone release with long action and oral
References activity. Proc Natl Acad Sci USA 1988; 85: 8236–40.
1 Conn PM, Crowley WF, Jr. Gonadotropin-releasing hormone and its 25 Rivier J, Porter J, Hoeger C, Theobald P, Craig AG, Dykert J, et al.
analogs. Annu Rev Med 1994; 45: 391–05. Gonadotropin-releasing hormone antagonists with N omega-
2 Carolsfeld J, Powell JF, Park M, Fischer WH, Craig AG, Chang JP, triazolylornithine, -lysine, or -p-aminophenylalanine residues at
et al. Primary structure and function of three gonadotropin-releasing positions 5 and 6. J Med Chem 1992; 35: 4270–78.
hormones, including a novel form, from an ancient teleost, herring. 26 Nestor JJ, Jr., Tahilramani R, Ho TL, Goodpasture JC, Vickery BH,
Endocrinology 2000; 141: 505–12. Ferrandon P. Potent gonadotropin releasing hormone antagonists
3 Krsmanovic LZ, Martinez-Fuentes AJ, Arora KK, Mores N, with low histamine- releasing activity. J Med Chem 1992; 35:
Tomic M, Stojilkovic SS, et al. Local regulation of gonadotroph 3942–48.
function by pituitary gonadotropin-releasing hormone. Endocrinology 27 Garnick MB, Campion M. Abarelix depot, a GnRH antagonist, v
2000; 141: 1187–95. LHRH superagonists in prostate cancer: differential effects on follicle-
4 Kang SK, Choi KC, Cheng KW, Nathwani PS, Auersperg N, stimulating hormone. Mol Urol 2000; 4: 275–77.
Leung PC. Role of gonadotropin-releasing hormone as an autocrine 28 Cook T, Sheridan WP. Development of GnRH antagonists for
growth factor in human ovarian surface epithelium. Endocrinology prostate cancer: new approaches to treatment. Oncologist 2000;
2000; 141: 72–80. 5: 162–8.
5 Chegini N, Rong H, Dou Q, Kipersztok S, Williams RS. 29 Deghenghi R, Boutignon F, Wuthrich P, Lenaerts V. Antarelix (EP
Gonadotropin-releasing hormone (GnRH) and GnRH receptor gene 24332) a novel water soluble LHRH antagonist. Biomed Pharmacother
expression in human myometrium and leiomyomata and the direct 1993; 47: 107–10.

1800 THE LANCET • Vol 358 • November 24, 2001

For personal use. Only reproduce with permission from The Lancet Publishing Group.
NEW DRUG CLASSES

30 Reissmann T, Felberbaum R, Diedrich K, Engel J, Comaru- 50 Albano C, Felberbaum RE, Smitz J, Riethmuller-Winzen H, Engel J,
Schally AM, Schally AV. Development and applications of luteinizing Diedrich K, et al. European Cetrorelix Study Group. Ovarian
hormone-releasing hormone antagonists in the treatment of infertility: stimulation with HMG: results of a prospective randomized phase III
an overview. Hum Reprod 1995; 10: 1974–81. European study comparing the luteinizing hormone-releasing
31 Duijkers IJ, Klipping C, Willemsen WN, Krone D, Schneider E, hormone (LHRH)-antagonist cetrorelix and the LHRH-agonist
Niebch G, et al. Single and multiple dose pharmacokinetics and buserelin. Hum Reprod 2000; 15: 526–31.
pharmacodynamics of the gonadotrophin-releasing hormone 51 Borm G, Mannaerts B. The European Orgalutran Study Group.
antagonist cetrorelix in healthy female volunteers. Hum Reprod 1998; Treatment with the gonadotrophin-releasing hormone antagonist
13: 2392–98. ganirelix in women undergoing ovarian stimulation with recombinant
32 Oberye JJ, Mannaerts BM, Kleijn HJ, Timmer CJ. Pharmacokinetic follicle stimulating hormone is effective, safe and convenient: results
and pharmacodynamic characteristics of ganirelix of a controlled, randomized, multicentre trial. Hum Reprod 2000; 15:
(Antagon/Orgalutran): parts I and II. Fertil Steril 1999; 72: 1001–05, 1490–98.
1006–12. 52 The European and Middle East Orgalutran Study Group.
33 Sommer L, Zanger K, Dyong T, Dorn C, Luckhaus J, Diedrich K, Comparable clinical outcome using the GnRH antagonist ganirelix or
et al. Seven-day administration of the gonadotropin-releasing a long protocol of the GnRH agonist triptorelin for the prevention of
hormone antagonist cetrorelix in normal cycling women. Eur J premature LH surges in women undergoing controlled ovarian
Endocrinol 1994; 131: 280–85. hyperstimulation. Hum Reprod 2001; 16: 644–51.
34 Garnick MB, Tomera K, Campion M, Kuca B, Gefter M. Abarelix- 53 Fluker M, Crifo J, Leader A, et al. The North American Ganirelix
depot (A-D), a potent GnRH pure antagonist in patients (pts) with Study Group. Efficacy and safety of ganirelix acetate (Antagon/
prostate cancer (PrCA): initial clinical results and endocrine Orgalutran) versus leuprolide acetate in women undergoing controlled
comparison with superagonists Lupron (L) and Zoladex (Z). Gynecol ovarian hyperstimulation. Fertil Steril 2001; 75: 38–45.
Endocrinol 1999; 13 (suppl 1): abstr. 54 Giuliani A, Schoell W, Auner J, Urdl W. Controlled ovarian
35 Garnick MB, Campion M, Abarelix Depot Study Group. Abarelix hyperstimulation in assisted reproduction: effect on the immune
depot, a GnRH antagonist, v LHRH superagonists in prostate cancer: system. Fertil Steril 1998; 70: 831–35.
differential effects on folicle-stimulating hormone. Mol Urol 2000; 55 Brus L, Lambalk CB, de Koning J, Helder MN, Janssens RM,
4: 275–77. Schoemaker J. Specific gonadotrophin-releasing hormone
36 Tomera K, Gleason D, Gittelman M, et al. The Abarelix Study analogue binding predominantly in human luteinized follicular
Group. The gonadotropin-releasing hormone antagonist abarelix aspirates and not in human pre-ovulatory follicles. Hum Reprod 1997;
depot versus luteinizing hormone releasing hormone agonists 12: 769–73.
leuprolide or goserelin: initial results of endocrinological and 56 Janssens RM, Brus L, Cahill DJ, Huirne JA, Schoemaker J,
biochemical efficacies in patients with prostate cancer. J Urol 2001; Lambalk CB. Direct ovarian effects and safety aspects of GnRH
165: 1585–89. agonists and antagonists. Hum Reprod Update 2000; 6: 505–18.
37 Sharpless JL, Supko JG, Martin KA, Hall JE. Disappearance of 57 Hernandez ER. Embryo implantation and GnRH antagonists: embryo
endogenous luteinizing hormone is prolonged in postmenopausal implantation: the Rubicon for GnRH antagonists. Hum Reprod 2000;
women. J Clin Endocrinol Metab 1999; 84: 688–94. 15: 1211–6.
38 Rabinovici J, Rothman P, Monroe SE, Nerenberg C, Jaffe RB. 58 Mannaerts B, Gordon K. Embryo implantation and GnRH
Endocrine effects and pharmacokinetic characteristics of a potent new antagonists: GnRH antagonists do not activate the GnRH receptor.
gonadotropin-releasing hormone antagonist (ganirelix) with minimal Hum Reprod 2000; 15: 1882–83.
histamine-releasing properties: studies in postmenopausal women. 59 Demirel LC, Weiss JM, Polack S, Unlu C, Diedrich K, Ortmann O.
J Clin Endocrinol Metab 1992; 75: 1220–25. Effect of the gonadotropin-releasing hormone antagonist ganirelix on
39 Gonzalez-Barcena D, Vadillo BM, Garcia PE, Guerra-Arguero L, cyclic adenosine monophosphate accumulation of human granulosa-
Cardenas CI, Comaru-Schally AM, et al. Inhibition of luteinizing lutein cells. Fertil Steril 2000; 74: 1001–07.
hormone, follicle-stimulating hormone and sex-steroid levels in men 60 Ortmann O, Weiss JM, Diedrich K. Embryo implantation and GnRH
and women with a potent antagonist analog of luteinizing hormone- antagonists: ovarian action of GnRH antagonists. Hum Reprod 2001;
releasing hormone, cetrorelix (SB-75). Eur J Endocrinol 1994; 16: 608–11.
131: 286–92. 61 Raga F, Casan EM, Kruessel J, Wen Y, Bonilla-Musoles F,
40 Fujimoto VY, Monroe SE, Nelson LR, Downey D, Jaffe RB. Dose- Polan ML. The role of gonadotropin-releasing hormone in murine
related suppression of serum luteinizing hormone in women by a preimplantation embryonic development. Endocrinology 1999;
potent new gonadotropin-releasing hormone antagonist (ganirelix) 140: 3705–12.
administered by intranasal spray. Fertil Steril 1997; 67: 469–73. 62 Funston RN, Seidel GE, Jr. Gonadotropin-releasing hormone
41 Hall JE, Taylor AE, Martin KA, Rivier J, Schoenfeld DA, increases cleavage rates of bovine oocytes fertilized in vitro. Biol
Crowley WF Jr. Decreased release of gonadotropin-releasing hormone Reprod 1995; 53: 541–55.
during the preovulatory midcycle luteinizing hormone surge in normal 63 Morales P, Kerr B, Oliva C, Pizarro E, Kong M. Gonadotrophin-
women. Proc Natl Acad Sci USA 1994; 91: 6894–98.
releasing hormone antagonists inhibit sperm binding to the human
42 Leroy I, d’Acremont M, Brailly-Tabard S, Frydman R, de Mouzon J, zona pellucida. Hum Reprod 1999; 14: 2069–74.
Bouchard P. A single injection of a gonadotropin-releasing hormone
64 Olivennes F, Belaisch-Allart J, Emperaire JC, Dechaud H, Alvarez S,
(GnRH) antagonist (cetrorelix) postpones the luteinizing hormone
Moreau L, et al. Prospective, randomized, controlled study of in vitro
(LH) surge: further evidence for the role of GnRH during the LH
surge. Fertil Steril 1994; 62: 461–67. fertilization- embryo transfer with a single dose of a luteinizing
hormone-releasing hormone (LH-RH) antagonist (cetrorelix) or a
43 Kettel LM, Roseff SJ, Chiu TC, Bangah ML, Vale W, Rivier J, et al.
depot formula of an LH-RH agonist (triptorelin). Fertil Steril 2000;
Follicular arrest during the midfollicular phase of the menstrual cycle:
73: 314–20.
a gonadotropin-releasing hormone antagonist imposed follicular-
follicular transition. J Clin Endocrinol Metab 1991; 73: 644–49. 65 The Ganirelix Dose-Finding Study Group. A double-blind,
44 Fluker MR, Marshall LA, Monroe SE, Jaffe RB. Variable ovarian randomized, dose-finding study to assess the efficacy of the
response to gonadotropin-releasing hormone antagonist- induced gonadotrophin-releasing hormone antagonist ganirelix (Org 37462) to
gonadotropin deprivation during different phases of the menstrual prevent premature luteinizing hormone surges in women undergoing
cycle. J Clin Endocrinol Metab 1991; 72: 912–19. ovarian stimulation with recombinant follicle stimulating hormone
(Puregon). Hum Reprod 1998; 13: 3023–31.
45 Couzinet B, Lahlou N, Thomas G, Thalabard JC, Bouchard P,
Roger M, et al. Effects of gonadotrophin releasing hormone 66 Kol S, Lightman A, Hillensjo T, et al. High doses of gonadotrophin-
antagonist and agonist on the pulsatile release of gonadotrophins and releasing hormone antagonist in in-vitro fertilization cycles do not
alpha-subunit in postmenopausal women. Clin Endocrinol (Oxf) 1991; adversely affect the outcome of subsequent freeze-thaw cycles. Hum
34: 477–83. Reprod 1999; 14: 2242–44.
46 Campen CA, Lai MT, Kraft P, Kirchner T, Phillips A, Hahn DW, 67 de Jong D, Macklon NS, Eijkemans MJ, et al. Dynamics of the
et al. Potent pituitary-gonadal axis suppression and extremely low development of multiple follicles during ovarian stimulation for in
anaphylactoid activity of a new gonadotropin releasing hormone vitro fertilization using recombinant follicle-stimulating hormone
(GnRH) receptor antagonist “azaline B”. Biochem Pharmacol 1995; (Puregon) and various doses of gonadotropin-releasing hormone
49: 1313–21. antagonist ganirelix (Orgalutran/Antagon). Fertil Steril 2001; 75:
47 Anon. Cetrorelix, LHRH antagonist. Drugs Future 1994; 19: 228–37. 688–93.
48 Anon. Ganirelix acetate, GnRH antagonist treatment of female 68 Ludwig M, Riethmüller-Winzen H, Felberbaum RE, et al. Health of
infertility. Drugs Future 2000; 24: 393–403. 227 children born after controlled ovarian stimulation for in vitro
49 Felberbaum R, Diedrich K. Ovarian stimulation for in-vitro fertilization using the luteinizing hormone releasing hormone
fertilization/intracytoplasmic sperm injection with gonadotrophins and antagonist cetrorelix. Fertil Steril 2001; 75: 18–22.
gonadotrophin-releasing hormone analogues: agonists and 69 Edwards RG, Lobo R, Bouchard P. Time to revolutionize ovarian
antagonists. Hum Reprod 1999; 14 (suppl 1): 207–21. stimulation. Hum Reprod 1996; 11: 917–19.

THE LANCET • Vol 358 • November 24, 2001 1801

For personal use. Only reproduce with permission from The Lancet Publishing Group.
NEW DRUG CLASSES

70 Janssens RM, Lambalk CB, Vermeiden JP, et al. Dose-finding study 89 Felberbaum RE, Ludwig M, Diedrich K. Medical treatment of
of triptorelin acetate for prevention of a premature LH surge in IVF: a uterine fibroids with the LHRH antagonist: cetrorelix. Contracept
prospective, randomized, double-blind, placebo-controlled study. Fertil Sex 1999; 27: 701–09.
Hum Reprod 2000; 15: 2333–40. 90 Gonzalez-Barcena D, Alvarez RB, Ochoa EP, Cornejo IC, Comaru-
71 Templeton A, Morris JK. Teducing the risk of multiple births by Schally AM, Schally AV, et al. Treatment of uterine leiomyomas with
transfer of two embryos after in vitro fertilization. N Engl J Med 1998; luteinizing hormone-releasing hormone antagonist cetrorelix. Hum
339: 573–77. Reprod 1997; 12: 2028–35.
72 Olivennes F, Alvarez S, Bouchard P, Fanchin R, Salat-Baroux J, 91 Fauser BC, Laven JS, de Jong D, Macklon NS. Gonadotrophin-
Frydman R. The use of a GnRH antagonist (cetrorelix) in a single releasing hormone antagonists: application in ovary-stimulating and
dose protocol in IVF-embryo transfer: a dose finding study of 3 versus sex-steroid dependent disorders. Ned Tijdschr Geneeskd 2000;
2 mg. Hum Reprod 1998; 13: 2411–14. 144: 370–74.
73 Albano C, Smitz J, Camus M, Riethmuller-Winzen H, Van 92 Emons G, Schulz KD. Primary and salvage therapy with LH-RH
Steirteghem A, Devroey P. Comparison of different doses of analogues in ovarian cancer. Recent Results Cancer Res 2000;
gonadotropin-releasing hormone antagonist cetrorelix during 153: 83–94.
controlled ovarian hyperstimulation. Fertil Steril 1997; 67: 917–22. 93 Emons G, Weiss S, Ortmann O, Grundker C, Schulz KD. LHRH
74 Serono. A double-blind, randomised, parallel group, dose-finding, might act as a negative autocrine regulator of proliferation of human
phase II study to determine the minimal effective dose of Antide for ovarian cancer. Eur J Endocrinol 2000; 142: 665–70.
the prevention of an untimely spontaneous LH surge in patients 94 Emons G, Heyl W. Hormonal treatment of endometrial cancer.
undergoing stimulation of multiple follicular development for assisted J Cancer Res Clin Oncol 2000; 126: 619–23.
reproductive technology (ART) with recombinant human follicle 95 Noci I, Coronnello M, Borri P, Borrani E, Giachi M, Chieffi O, et al.
stimulating hormone (Gonal-F). Data on file with Serono Inhibitory effect of luteinising hormone-releasing hormone analogues
International. on human endometrial cancer in vitro. Cancer Lett 2000; 150: 71–78.
75 Al-Inany H, Aboulghar M. GnRH antagonist in assisted reproduction. 96 Kakar SS, Grizzle WE, Neill JD. The nucleotide sequences of human
(Cochrane review). In: The Cochrane Library, issue 4. Oxford. Update GnRH receptors in breast and ovarian tumors are identical with that
Software, 2001. found in pituitary. Mol Cell Endocrinol 1994; 106: 145–94.
76 Ludwig M, Felberbaum R, Albano C, Olivennes F, Riethmiiller H, 97 Eidne KA, Flanagan CA, Harris NS, Millar RP. Gonadotropin-
Devroey P, Diedrich K. Cetrorelix levels in plasma and follicular releasing hormone (GnRH)-binding sites in human breast cancer cell
fluid. Gynecol Endocrinol 1999; 13 (suppl 1): 030 (abstr). lines and inhibitory effects of GnRH antagonists. J Clin Endocrinol
77 Bonduelle M. Neonatal outcome of pregnancies established after Metab 1987; 64: 425–32.
treatment with recombinant FSH and Ganirelix for ART. 6th 98 Miller WR, Scott WN, Morris R, Fraser HM, Sharpe RM. Growth of
International Symposium on GnRH Analogues in Cancer and Human human breast cancer cells inhibited by a luteinizing hormone-releasing
Reproduction (Geneva 2001): 0056 (abstr). hormone agonist. Nature 1985; 313: 231–33.
http:/www.kenes.com/gnrh/Abstracts/0056aBonduelle.htm 99 Comaru-Schally AM, Brannan W, Schally AV, Colcolough M,
78 Felberbaum RE, Reissmann T, Kupker W, et al. Preserved pituitary Monga M. Efficacy and safety of luteinizing hormone-releasing
response under ovarian stimulation with HMG and GnRH hormone antagonist cetrorelix in the treatment of symptomatic benign
antagonists (Cetrorelix) in women with tubal infertility. Eur J Obstet prostatic hyperplasia. J Clin Endocrinol Metab 1998; 83: 3826–31.
Gynecol Reprod Biol 1995; 61: 151–55. 100 Gonzales-Barcena D, Schally AV, Comaru-Schally AM, et al.
79 Christin-Maitre S, Olivennes F, Dubourdieu S, et al. Effect of Treatment of patients with advanced prostate cancer with LHRH
gonadotrophin-releasing hormone (GnRH) antagonist during the LH antagonist cetrorelix. In: Filicori M, Flamigni E, eds. Treatment
surge in normal women and during controlled ovarian with LHRH analogs: controversies and perspectives. London/New
hyperstimulation. Clin Endocrinol (Oxf) 2000; 52: 721–26. York: Parthenon Publishing, 1996: 139–45.
80 Itskovitz-Eldor J, Kol S, Mannaerts B. Use of a single bolus of GnRH 101 McLeod D, Zinner N, Gleason D, et al. Abarelix-depot (A-D)
agonist triptorelin to trigger ovulation after GnRH antagonist ganirelix versus leuprolide acetate (L) for prostate cancer: results of a multi-
treatment in women undergoing ovarian stimulation for assisted institutional, randomized, phase III study in 271 patients. Proc
reproduction, with special reference to the prevention of ovarian ASCO 2000; 19: 332a.
hyperstimulation syndrome: preliminary report: short communication. 102 Garnick MB, Tomera K, Campion M, Kuca B, Gefter M. Abarelix-
Hum Reprod 2000; 15: 1965–68. depot (A-D), a sustained-release (SR) formulation of a potent
81 de Jong D, Macklon NS, Fauser BC. A pilot study involving GnRH pure antagonist in patients with prostate cancer (PrCA):
minimal ovarian stimulation for in vitro fertilization: extending the phase II clinical results and endocrine comparison with superagonist
“follicle-stimulating hormone window” combined with the Lupron (L) and Zoladex (Z). Proc ASCO 1999; 18: 321a.
gonadotropin-releasing hormone antagonist cetrorelix. Fertil Steril 103 Stricker HJ. Luteinizing hormone-releasing hormone antagonist in
2000; 73: 1051–4. prostate cancer. Urology 2001; 58: 24–27.
82 Olivenne F, Ayoubi JM, Fanchin R, et al. GnRH antagonist in single- 104 Trachtenberg J, Fortheringham N, Campion M, Garnick M.
dose applications. Hum Reprod Update 2000; 6: 313–17. Avoidance of FSH surge and maintained suppression of follicle-
83 Van Loenen ACD, Huirne JAF, Schats R, Donnez J, Lambalk CB. stimulating-hormone (FSH) with Abarelix depot (A-D) compared to
An open label multicentre randomised parallel controlled phase II leuprolide (L) ± Bicalutamide in prostate cancer (PC) patients. Proc
study to assess the feasibility of a new programming regimen using an ASCO 2001; 20: 152.
oral contraceptive prior to the administration of recombinant FSH 105 Gittelman M, Gleave M, Pommerville P, et al. Greater and more
and an FNRH-antagonist in patients undergoing an ART(IVF/ICSI)- rapid decrease in prostate specific antigen (PSA) and testosterone
treatment. ESHRE 2001; 16: 144. (T) levels with abarelix depot (A-D) compared to leuprolide acetate
84 de Jong D, Macklon NS, Mannaerts BM, Coelingh Bennink HJ, (L): results of a multicenter 24-weeks safety study. Proc ASCO 2001;
Fauser BC. High dose gonadotrophin-releasing hormone antagonist 20: 154.
(ganirelix) may prevent ovarian hyperstimulation syndrome caused by 106 Ben-Josef E, Yang SY, JI TH, et al. Hormone refractory prostate
ovarian stimulation for in-vitro fertilization. Hum Reprod 1998; cancer cells express functional follicle-stimulating hormone receptor.
13: 573–75. J Urol 1999; 161: 970.
85 Hayes FJ, Taylor AE, Martin KA, Hall JE. Use of a gonadotropin- 107 Koch M, Steidle C, Brosman S, et al. Abarelix depot (A-D) a GnRH
releasing hormone antagonist as a physiologic probe in polycystic angagonist benefits highly symptomatic prostate cancer (PC)
ovary syndrome: assessment of neuroendocrine and androgen patients who are at risk for a clinical flare phenomenon with LHRH
dynamics. J Clin Endocrinol Metab 1998; 83: 2343–49. agonist treatment. 96th annual meeting of American Urologic
86 Gordon K, Danforth DR, Williams RF, Hodgen GD. New trends in Association, 2001; 753 (abstr).
combined use of gonadotropin-releasing hormone antagonists with 108 Swerdloff RS, Bagatell CJ, Wang C, et al. Suppression of
gonadotropins or pulsatile gonadotropin-releasing hormone in spermatogenesis in man induced by Nal-Glu gonadotropin releasing
ovulation induction and assisted reproductive technologies. Curr Opin hormone antagonist and testosterone enanthate (TE) is maintained
Obstet Gynecol 1992; 4: 690–96. by TE alone. J Clin Endocrinol Metab 1998; 83: 3527–33.
87 Dubourdieu S, Le Nestour E, Spitz IM, Charbonnel B, Bouchard P. 109 Pavlou SN, Brewer K, Farley MG, Lindner J, Bastias MC,
The combination of gonadotrophin-releasing hormone (GnRH) Rogers BJ, et al. Combined administration of a gonadotropin-
antagonist and pulsatile GnRH normalizes luteinizing hormone releasing hormone antagonist and testosterone in men induces
secretion in polycystic ovarian disease but fails to induce follicular reversible azoospermia without loss of libido. J Clin Endocrinol Metab
maturation. Hum Reprod 1993; 8: 2056–60. 1991; 73: 1360–69.
88 Martha PM, Gray ME, Campion M, Kuca B, Garnick MB. Initial 110 Roth C, Leonhardt S, Seidel C, Luft H, Wuttke W, Jarry H.
safety profile and hormonal dose-response characteristics of the pure Comparative analysis of different puberty inhibiting mechanisms of
GnRH antagonist, abarelix-depot, in women with endometriosis. two GnRH agonists and the GnRH antagonist cetrorelix using a
Gynecol Endocrinol 1999; 13 (suppl 1): 104 (abstr). female rat model. Pediatr Res 2000; 48: 468–74.

1802 THE LANCET • Vol 358 • November 24, 2001

For personal use. Only reproduce with permission from The Lancet Publishing Group.
NEW DRUG CLASSES

111 Kamp GA, Waelkens JJJ, Delemarre-van de Waal HA, et al. 116 Levitsky AM, Owens NJ. Pharmacologic treatment of hypersexuality
Randomized controlled trial of three years growth hormone and and paraphilias in nursing home residents. J Am Geriatr Soc 1999;
gonadotropin releasing hormone analog treatment in children with 47: 231-34.
idiopathic short stature and intra uterine growth retardation. J Clin 117 Ashton WT, Sisco RM, Kieczykowski GR, et al. Orally bioavailable,
Endocrinol Metab 2001; 86: 2969–75. indole-based nonpeptide GnRH receptor antagonists with high
112 Daneshdoost L, Pavlou SN, Molitch ME, et al. Inhibition of follicle- potency and functional activity. Bioorg Med Chem Lett 1999; 9:
stimulating hormone secretion from gonadotroph adenomas by 2597–602.
repetitive administration of a gonadotropin-releasing hormone
118 Wong SL, Dmowski WP, DePaoli A, Gray ME, Martha PM.
antagonist. J Clin Endocrinol Metab 1990; 71: 92–97.
Pharmacokinetics of abarelix depot-F by subcutaneous injection in
113 Chanson P, Lahlou N, Warnet A, et al. Responses to gonadotropin
women with endometriosis associated pain. Fertil Steril 2000;
releasing hormone agonist and antagonist administration in patients
74 (suppl 1): P–286(abstr)
with gonadotroph cell adenomas. J Endocrinol Invest 1994; 17:
91–98. 119 Behre HM, Bockers A, Schlingheider A, et al. Sustained suppression
114 Ataya K, Rao LV, Lawrence E, Kimmel R. Luteinizing hormone- of serum LH, FSH and testosterone and increase of high-density
releasing hormone agonist inhibits cyclophosphamide-induced lipoprotein cholesterol by daily injections of the GnRH antagonist
ovarian follicular depletion in rhesus monkeys. Biol Reprod 1995; 52: cetrorelix (SB-75) in normal men. J Clin Endocrinol Metab 1994;
365–72. 40: 241–248.
115 Ataya K, Pydyn E, Ramahi-Ataya A, Orton CG. Is radiation- 120 Erb K, Klipping C, Duijkers I, Pechstein B, Achueler A,
induced ovarian failute in rhesus monkeys preventable by luteinizing Hermann R. Pharmacodynamic effects and plasma
hormone-releasing hormone agonists?: Preliminary observations. pharmacokinetics of single doses of cetrorelix acetate in healthy
J Clin Endocrinol Metab 1995; 80: 790-95. premenopausal women. Fertil Steril 2001; 75: 316-23.

Futher reading available upon request from the authors

THE LANCET • Vol 358 • November 24, 2001 1803

For personal use. Only reproduce with permission from The Lancet Publishing Group.

You might also like