Sie sind auf Seite 1von 9

Human Reproduction Vol.20, No.8 pp. 2043–2051, 2005 doi:10.

1093/humrep/dei042
Advance Access publication May 5, 2005

Clomiphene citrate—end of an era? a mini-review

Roy Homburg
Division of Reproductive Medicine, Department of Obstetrics and Gynaecology, Vrije Universiteit Medical Centre, De Boelelaan
1117, PO Box 7057, 1007 MB Amsterdam, The Netherlands
Address for correspondence: E-mail: R.Homburg@vumc.nl

The purpose of this review is to examine whether the time has come to replace clomiphene citrate (CC) as the first
line therapy for WHO group II (eu-oestrogenic) infertility, the majority of which is associated with polycystic
ovary syndrome. CC has been the first line therapy for these cases for the last 40 years. It is a simple, cheap treat-
ment, almost devoid of side effects which yields a single live birth rate of ,25% of starters. Non-response to CC
and the gap between ovulation and pregnancy rates have variously been attributed to its anti-estrogen effects, and

Downloaded from humrep.oxfordjournals.org by guest on February 25, 2011


high LH and androgen concentrations. Three possible contenders for the replacement of CC as first-line treatment
are scrutinized: metformin, aromatase inhibitors and low-dose FSH. Each has their advantages and disadvantages,
but none of them, while showing much potential promise, has been proven, as yet, to be a feasible replacement for
CC in this role. For CC, it may not yet be the end of an era but it may be the beginning of the end.

Key words: aromatase inhibitors/clomiphene citrate/low-dose FSH/metformin/PCOS

Introduction Despite the use of CC in controlled ovarian stimulation, its


It is now . 40 years ago that Greenblatt first reported a new original indication of ovulation induction for WHO group II
compound, the anti-estrogen MRL-41, capable of inducing classification of oligo- or anovulation, particularly when
ovulation for anovulatory women (Greenblatt et al., 1961). associated with PCOS, remains its most frequent and most
Later to become known as clomiphene citrate (CC) this com- successfully treated indication. The fact that CC is an orally
pound has had a remarkably sustained career as the first-line administered, relatively cheap preparation, has proved an
treatment for women with absent or irregular ovulation due enormous advantage over its injected and expensive competi-
to hypothalamic –pituitary dysfunction associated with nor- tors. However, the field is becoming replete with alternatives
mal basal levels of endogenous estradiol (WHO group II). for the same indication and the time is ripe to re-evaluate the
The vast majority of these patients probably some 80% are place of CC in our armamentarium of ovulation-inducing
now known to be oligo- or anovulatory due to polycystic agents.
ovary syndrome (PCOS). Until the introduction of CC the
only plausible treatment for these patients who wished to
conceive was bilateral wedge resection of the ovaries. Mode of action
Although this abdominal surgical procedure had some con- Clomiphene contains an unequal mixture of two isomers as
siderable success in restoring ovulation and even inducing their citrate salts, enclomiphene and zuclomiphene. Zuclomi-
pregnancy it later fell from grace due to the generation of phene is much the more potent of the two for induction of
pelvic adhesions following the operation in a large number ovulation, accounts for 38% of the total drug content of one
of patients and the emergence of medical means to induce tablet and has a much longer half-life than enclomiphene,
ovulation. CC was the first medication capable of inducing being detectable in plasma 1 month following its adminis-
ovulation and as such created a welcome revolution in the tration. Rostami-Hodjegan et al. (2004) have suggested that
treatment of infertility associated with anovulation. wide variability in the metabolism of the zuclomiphene
Many years later, a further suggested indication was its component contributes to variability in response to the drug.
empirical use for ovarian stimulation in idiopathic (‘unex- CC is capable of inducing a discharge of FSH from the
plained’) infertility. Here its success was more limited but, anterior pituitary and this is often enough to reset the cycle of
with the introduction of assisted reproductive technology, it events leading to ovulation into motion. The release of even
became widely used as a ‘booster’ to ovulatory capacity in small amounts of FSH into the system will often induce ovu-
conjunction with intrauterine insemination (IUI) and, with lation and pregnancy in a proportion of eu-estrogenic anovula-
the rising costs of gonadotrophin preparations, as an adjuvant tory women. This is achieved indirectly, through the action of
to gonadotrophin stimulation of the ovaries in preparation for CC, a non-steroidal compound closely resembling an estro-
IVF. gen, in blocking hypothalamic estrogen receptors, signalling
q The Author 2005. Published by Oxford University Press on behalf of the European Society of Human Reproduction and Embryology. All rights reserved. 2043
For Permissions, please email: journals.permissions@oupjournals.org
R.Homburg

Table I. Results of treatment with clomiphene citrate: a collection of published data


No of patients Ovulation Pregnancy Abortion Live birth

McGregor et al. (1968) 4098 2869 1393 279 1114


Garcia et al. (1977) 159 130 64 16 48
Gysler et al. (1982) 428 364 184 24 160
Hammond (1984) 159 137 67 10 57
Kousta et al. (1997) 128 113 55 13 42
Messinis and Milingos (1998) 55 51 35 4 31
Imani et al. (2002) 259 194 111 11 98
Total (% of patients) 5268 (100) 3858 (73) 1909 (36) 357 1550 (29)

a lack of circulating estrogen to the hypothalamus and indu- of CC for the induction of ovulation, and is generally consist-
cing a change in the pattern of pulsatile release of GnRH. ent from series to series.
From these data, a theoretical projection of the results of
CC induction of ovulation in 100 women starting treatment
Dose has been made (Table III, column 2), concluding that 25 of
CC is given orally in a dose of 50– 250 mg per day for 5 these 100 will succeed in delivering a singleton, healthy baby.
days from day 2, 3, 4 or 5 of spontaneous or induced bleed- Although CC will restore ovulation in , 73% of patients,

Downloaded from humrep.oxfordjournals.org by guest on February 25, 2011


ing, starting with the lowest dose and increasing the dose in it will result in pregnancy in only , 36%. The 27% of anovu-
increments of 50 mg/day per cycle until an ovulatory cycle is latory women with normal FSH concentrations who do not
achieved. The starting day of treatment, whether on day 2 or respond at all are considered to be ‘CC resistant’.
through day 5 of the cycle, does not influence the results
(Wu and Winkel, 1989). Although 50 mg/day is the rec-
ommended dose in the first cycle, a meta-analysis of 13 Failure to ovulate
published reports (Rostami-Hodjegan et al., 2004) suggests Inability of CC to induce ovulation is more likely in patients
that only 46% will respond to this dose with ovulation, a who are obese, insulin resistant and hyperandrogenic com-
further 21% will respond to 100 mg and another 8% will ovu- pared with those who do respond (Imani et al., 1998). This
late with 150 mg/day. There is no apparent advantage of careful prospective study pinpointed a high free androgen
using a daily dose of . 150 mg which seems to significantly index as the best predictor of non-response to CC. Although
increase neither the ovulation rate nor follicular recruitment it is virtually impossible to predict who will respond to
(Dickey et al., 1997). Some practitioners often use a starting which dose of CC, if at all (Imani et al., 2002), body weight
dose of 100 mg/day from day 4 or 5, only resorting to has been found to be an impeding factor. Overweight women
50 mg/day in the case of exquisite sensitivity or persistent respond less well (Polson et al., 1989) and the dose of CC
cyst formation. The advantage of starting with a 100 mg needed to induce ovulation correlates with body weight
daily dose rather than 50 mg is that it will cut down the (Lobo et al., 1982).
number of ‘superfluous’ cycles of treatment until ovulation is
achieved and until those resistant to CC are identified. It is
difficult to state what effect, if any, this course of action has Failure to conceive
on the multiple pregnancy rate. It is frustrating that the restoration of ovulation by CC does
not produce a much higher pregnancy rate. This discrepancy
between ovulation and pregnancy rates (only 50% of those
Results who ovulate will conceive) may be partly explained by the
A compilation of published results regarding ovulation and peripheral anti-estrogenic effects of CC at the level of the
pregnancy rates following treatment with CC is shown in endometrium and cervical mucus or by hypersecretion of
Table I. It reveals an ovulation rate of 73% and a pregnancy LH. While the depression of the cervical mucus, occurring in
rate of 36% in data from 5268 patients. To the data on preg- , 15% of patients, may be overcome by performing IUI
nancy, abortion and live birth rates in Table I, I have added
similar data from four further studies dealing with the preg-
Table II. Outcome of pregnancy following treatment with clomiphene
nancies and their outcome. From a grand total of 4054 preg- citrate
nancies, , 20% terminated in a spontaneous abortion
Pregnancies Abortion Live births
and almost all the rest in a live birth (Table II). From this
collection of results, it was very difficult to estimate the Total data from Table 1 1909 357 1550
multiple pregnancy rate which was rarely addressed. How- Ahlgren et al. (1976) 159 18 141
Adashi et al. (1979) 86 23 62
ever, from the available numbers and from other review pub- Correy et al. (1982) 156 16 140
lications (Scialli et al., 1986), this is estimated to be 8– 13%, Dickey et al. (1996) 1744 413 1331
the vast majority being twin pregnancies. Obviously a collec- Total (% of pregnancies) 4054 (100) 827 (20.4) 3224 (79.5)
tion of results of this nature comprises very heterogeneous Results from four further publications dealing with the outcome of pregnancy
series but nevertheless gives a very good idea of the efficiency are added to data from Table I.

2044
Clomiphene citrate—end of an era?

Outcome of pregnancies
Table III. Projected results of treatment
CC blocks the negative feedback mechanism which the even-
Start with CC resistant on First-line treatment
CC (n ¼ 100) low-dose FSH with low-dose tually rising estradiol levels would normally invoke to reduce
(n ¼ 100) FSH (n ¼ 100) discharge of FSH. The continued flow of FSH encourages
multiple follicle development which is relatively common.
No response 27 15 8
Ovulation 73 85 92 The risk of multiple gestation is therefore increased and is
No conception 37 45 30 estimated at , 8 – 13% (Schenker et al., 1981; Scialli et al.,
Conception 36 40 62 1986; Kousta et al., 1997; Eijkmans et al., 2003). The vast
Miscarriages 8 8 14
Multiple 3 2 3 majority of these are twin pregnancies, but the risk may be
pregnancy reduced considerably by ultrasound monitoring and withhold-
Singleton live 25 30 45 ing HCG, IUI or intercourse if more than two follicles
birth
. 15 mm diameter are seen (Homburg and Insler, 2002).
Column 2 (based on actual results from Tables I and II) ¼ 100 anovulatory Although there are conflicting reports, the prevalence of
(WHO group II) women starting with CC; column 3 (based on data from spontaneous abortion of a clinical pregnancy following CC
Table IV) ¼ 100 CC-resistant women receiving low-dose FSH.
Column 4 are ‘virtual’ data of 100 anovulatory (WHO group II) women therapy has a reported mean of , 20% (McGregor et al.,
using low-dose FSH as first-line treatment. It is based on the assumptions 1968, Dickey et al., 1996; Kousta et al., 1997), slightly
that all those who would have conceived if starting with CC (n ¼ 36) plus higher than the 12– 15% of clinically recognized spontaneous
40% of CC failures (CC resistant þ ovulation but no conception (n ¼ 26)
would conceive if starting with low-dose FSH. miscarriages in the normal population (Warburton and Fraser,

Downloaded from humrep.oxfordjournals.org by guest on February 25, 2011


1964; Wentz and Cartwright, 1988). Although a very contro-
versial issue due to a lack of evidence for a direct causal
relationship, several investigators have found in retrospective
suppression of endometrial proliferation, unrelated to dose or studies that an increased prevalence of miscarriage is associ-
duration of treatment but apparently idiosyncratic, indicates a ated with high serum LH concentrations in the mid-follicular
poor prognosis for conception if the endometrial thickness on phase (Homburg et al., 1988b; Regan et al., 1990). The
ultrasound scanning does not reach 8 mm at ovulation. In our increased prevalence of miscarriage following CC therapy
experience, the prevalence of endometrial suppression is one may, in part, be due to the high LH values induced immedi-
in every 6 –7 patients and, if noted in the first cycle of treat- ately after this treatment. Kousta et al. (1997) found signifi-
ment with CC, it will almost certainly be seen in repeated cantly higher LH levels post-CC in those who miscarried
cycles in the same woman. There is little point in persisting compared with those who delivered, and just 37% of the
after even one cycle, and a step-up to other forms of ovu- ongoing pregnancy group had LH . 10 IU/l compared with
lation induction is recommended. 75% of those who aborted. We postulated that this is due to
The main action of CC, indirectly stimulating GnRH a premature maturation of the oocyte caused by high LH con-
secretion, not only increases the desired FSH release but also centrations and resulting in an ‘aged’ oocyte which is diffi-
produces an undesirable increase in LH concentrations. This cult to fertilize and, if fertilized, is more likely to abort
increase in LH, whose basal level is often already high in (Homburg et al., 1988a).
women with PCOS, may compromise pregnancy rates in The prevalence of congenital abnormalities following CC
those receiving CC (Homburg et al., 1988a; Shoham et al., treatment is no different from those seen in spontaneously
1990). We have demonstrated that pre-treatment with micro- conceived pregnancies (Correy et al., 1982).
nized progesterone is capable of modulating LH pulsatility,
reducing LH concentrations and inducing a more favourable
environment for ovulation induction with CC (Homburg Side effects
et al., 1988b). This treatment initiated a response to CC and Unpleasant side effects of CC are few and far between,
yielded consequent pregnancies in previous non-responders although some women will complain of hot flushes and some
to CC. of nausea. CC is, however, usually very well tolerated. While
A course of six ovulatory cycles is usually sufficient to mild ovarian enlargement is relatively common, in almost
know whether pregnancy will be achieved using CC before 40 years of practice, I have never seen a full blown ovarian
moving on to more complex treatment as it has been reported hyperstimulation syndrome (OHSS) as a result of CC
that 71 –87.5% of the pregnancies achieved with CC occur treatment. Occasional cyst formation can be treated
within the first three cycles of treatment (Gysler et al., 1982; conservatively.
Kousta et al., 1997; Imani et al., 1998). From our own very
large (unpublished) database, we noted that no further preg-
nancies at all were obtained with CC following seven ovula- Monitoring
tory cycles, although others have found that a further 5 – 6% CC is often given without any observation of the events of
do conceive between treatment cycle number 6 and 12 the cycle. However, monitoring of the CC-treated cycle by
(Kousta et al., 1997). In properly selected patients with no ultrasound evaluation of follicular growth and endometrial
other causes of infertility, the cumulative conception rate thickness on days 12– 14 of the cycle is justified by the
may reach as high as 60% after six cycles (Messinis and identification of those who are not responding or have
Milingos, 1998). depressed endometrial thickness, and is helpful in the timing
2045
R.Homburg

of natural intercourse or IUI. Confirmation, or otherwise, of anovulation and WHO group II infertility: (i) metformin; (ii)
ovulation can be obtained with estimation of the progesterone aromatase inhibitors; and (iii) recombinant FSH.
concentration in the assumed mid-luteal phase which is pre-
ferable to a basal body temperature chart. The added expense Metformin
of careful monitoring is neutralized by the prevention of pro- PCOS is associated with , 75% of all cases of anovulatory
tracted periods of possibly ineffective therapy and delay in infertility. Insulin is of prime importance in the pathophysio-
the inception of more efficient treatment. logy of PCOS. The rate of insulin resistance in women with
PCOS is 50 –80%, which means that a very large proportion
of cases of anovulation and infertility is associated with
Adjuvants hyperinsulinaemia and that the lowering of insulin concen-
In order to improve the outcome of treatment with CC, trations provides a new therapeutic pathway. Weight loss in
several adjuvants to CC treatment have been suggested. A the obese is a very effective way of achieving this, but
correctly timed ovulation-triggering dose of HCG (5000 – weight loss and lifestyle change often seem to be insurmoun-
10000 IU) is only theoretically warranted when the reason for table objects for the obese patient with PCOS and are not
a non-ovulatory response is that the LH surge is delayed or applicable for lean patients. The alternative possibility of
absent despite the presence of a well developed follicle. using insulin-lowering drugs (particularly metformin) is
Although the routine addition of HCG at mid-cycle seems to presently undergoing a thorough examination.
add little to the improvement of conception rates (Agarwal Metformin is an oral biguanide, well established for the

Downloaded from humrep.oxfordjournals.org by guest on February 25, 2011


and Buyalos, 1995), we have found it very useful, if given treatment of hyperglycaemia, that does not cause hypoglycae-
when an ultrasonically demonstrated leading follicle attains a mia in normoglycaemic patients. It is an insulin sensitizer
diameter of 19– 24 mm, for the timing of intercourse or IUI. which reduces insulin secretion and, consequently, lowers
The addition of dexamethazone as an adjunct to CC circulating total and free androgen levels with a resulting
therapy in a dose of 0.5 mg at bedtime is said to suppress improvement of the clinical sequelae of hyperandrogenism.
adrenal androgen secretion and induce responsiveness to CC Importantly, it also seems to have a direct action on ovarian
in previous non-responders, mostly hyperandrogenic women theca cells to decrease androgen production (Attia et al.,
with PCOS and elevated concentrations of dehydroepiandro- 2001; Mansfield et al., 2003).
sterone sulphate (DHEAS) (Daly et al., 1984). However, Metformin is taken orally in a dose of 1500 –2500 mg.
glucocorticoid steroid therapy often induces side effects About 15 –20% of patients may suffer from gastrointestinal
including increased appetite and weight gain, and should side effects. The indications for giving metformin to women
probably be reserved for women who have congenital adrenal with anovulatory PCOS have become progressively wider as
hyperplasia as a cause for their anovulation. it seems to be difficult to predict which individuals will
respond well with this medication (Fleming et al., 2002). The
difficulties of accurately measuring insulin sensitivity in all
Indications other than ovulation induction PCOS patients and the suggestion that metformin may also
CC has also been employed for ovarian stimulation in ovulat- be effective in non-hyperinsulinaemic subjects (Baillargeon
ing women, mainly for idiopathic (unexplained) infertility et al., 2004) has encouraged ‘blanket’ treatment with metfor-
and often combined with IUI. The rationale is presumably min of all PCOS patients in many centres. The wisdom of
that CC overcomes a subtle defect in ovulatory function or this strategy awaits ratification or, as noted by Harborne et al.
increases the number of mature follicles so increasing the (2003) in a critical review of the literature, clinical practice
likelihood of pregnancy (Guzick et al., 1998). Here the is ahead of the evidence.
success rate has been, understandably, notably less than in Here I will consider the best available evidence for the
anovulatory women. In a collection of data on the efficacy of treatment of anovulatory infertility with metformin, both as a
treatment for unexplained infertility, the use of CC alone pro- single agent and in combination with CC.
duced a pregnancy rate of 5.6% per cycle and CC combined Metformin alone. There are now a large number of studies
with IUI 8.3% per cycle (Guzick et al., 1998). While this is published on the effect of metformin in a dose of 1500 –
significantly superior to timed intercourse alone, it should be 2550 mg/day in women with PCOS. The majority of these
remembered that the baseline level from merely expectant studies have demonstrated a significant improvement in insu-
treatment in these cases ranges from 1.3 to 4.1%. lin concentrations, insulin sensitivity and serum androgen
In IVF, co-treatment with CC and gonadotrophins has concentrations, accompanied by decreased LH and increased
enjoyed brief periods of popularity as an attempt to decrease sex hormone-binding globulin (SHBG) concentrations
costs of medications. However, this co-treatment never (Nestler et al., 2002). The restoration of regular menstrual
seemed to achieve the same results as the more favoured cycles by metformin has been reported in a large majority of
protocols of today, and this combination is rarely used now. published series, and the reinstatement of ovulation occurred
in 78 –96% of patients (Velazquez et al., 1997; Nestler et al,
1998, Nestler et al., 2002; Moghetti et al., 2000; Ibanez et al.,
Possible alternatives to clomiphene 2001; Fleming et al., 2002; Harborne et al., 2003;
Three possible contenders have emerged as the replacement Baillargeon et al., 2004; Kashyap et al., 2004). Fleming et al.
of CC as primary, first-line treatment for oligo- or (2002), in the largest randomized placebo-controlled trial,
2046
Clomiphene citrate—end of an era?

demonstrated a significantly increased frequency of ovulation Safety. The evidence is so far encouraging concerning the
with metformin (850 mg, twice a day) compared with placebo efficiency and safety of metformin as a single agent or in
in a group of 92 oligomenorrhoeic women with PCOS. combination with CC for induction of ovulation in women
Although significant, the increased frequency of ovulation with hyperinsulinaemic PCOS (Homburg, 2002). In addition,
was modest [relative risk (RR) 1.29, 95% confidence interval metformin seem to be safe when continued throughout preg-
(CI) 1.00 –1.66]. In a systematic review (Kashyap et al., nancy, having no increase in congenital abnormalities, terato-
2004), metformin was found to be 50% better than placebo genicity or adverse effect on infant development (Glueck
for ovulation induction in infertile PCOS patients (RR 1.5, et al., 2002). Preliminary data even suggest that this strategy
CI 1.13– 1.99). In a meta-analysis (Lord et al., 2003), the can significantly decrease the high miscarriage rate usually
superiority of metformin over placebo in inducing ovulation associated with PCOS and reduce the incidence of gestational
was emphatic [odds ratio (OR) 3.88, CI 2.25– 6.69]. diabetes, pre-eclampsia and fetal macrosomia (Glueck et al.,
Interpretation of studies evaluating the effect of metformin 2002; Jacubowicz et al., 2002). The apparent lack of terato-
as a sole agent for the induction of ovulation is made difficult genicity of metformin has earned it a B classification and,
by their heterogeneity as regards selection of patients whether hopefully, these apparently beneficial effects of metformin
hyperinsulinaemic, hyperandrogenaemic, diagnosis of PCOS, given throughout pregnancy will be confirmed by future
obese or slim, duration of metformin administration and dose, studies.
etc. Although it now seems obvious that metformin is superior The glitazones, notably rosiglitazone and pioglitazone,
to placebo in inducing ovulation, none of the studies was pow- which also have the property of lowering insulin concen-

Downloaded from humrep.oxfordjournals.org by guest on February 25, 2011


ered to assess pregnancy as an outcome. trations, are also under investigation for similar indications.
Comparisons of CC with metformin as single agents for It is too premature to judge their effect on ovulation induc-
the induction of ovulation and attainment of pregnancy must tion for WHO group II anovulation. A positive effect for
therefore await the results of a sufficiently powered random- rosiglitazone when used alone and more so when combined
ized controlled trial (RCT), directly comparing the two. with CC was demonstrated for ovulation induction in women
Metformin þ CC. The combination of CC and metformin, at with PCOS (Ghazeeri et al., 2003). In women with PCOS but
the present state of knowledge, seems to be more effective normal insulin sensitivity, metformin proved more efficient
than either CC or metformin given alone regarding both ovu- than rosiglitazone in restoring ovulation (Baillargeon et al.,
lation induction and pregnancy. This would seem to be the 2004).
case reflected by two meta-analyses (Lord et al., 2003;
Kashyap et al., 2004). In the first, metformin þ CC was Aromatase inhibitors
found to be more effective than CC alone in achieving ovu- A compound capable of inducing ovulation but devoid of the
lation (OR 4.41, CI 2.37 –8.22) and also demonstrated a sig- adverse anti-estrogen effects of CC could be a serious chal-
nificant effect for the combination on pregnancy rates (OR lenger for the role of first-line treatment for WHO group II
4.4, CI 1.96 – 9.85). In the second, metformin þ CC was 3 – oligo/anovulation.
4 times superior to CC alone for both ovulation induction Aromatase inhibitors are non-steroidal compounds that
and achievement of pregnancy. However, caution is advised suppress estrogen biosynthesis by blocking the action of the
as numbers are small in both these analyses, and some enzyme aromatase which converts androstenedione and tes-
recently conducted large RCTs comparing metformin þ CC tosterone to estrogens. Letrozole, the most widely used
versus CC alone are not demonstrating any significant differ- aromatase inhibitor, has mainly been employed for the treat-
ences. As these results have not yet been officially published, ment of post-menopausal women with advanced breast
judgment should be reserved, but initial enthusiasm will cancer. It is given orally in a dose of 2.5 –5 mg/day and is
probably be dampened. almost free of side effects.
It is, however, interesting to examine the individual pub- It has been hypothesized, in particular by Mitwally and
lished studies. In an RCT performed on CC-resistant infertile Casper (2001), that the efficient estrogen-lowering properties
patients with PCOS, compared with CC and placebo, metfor- of the aromatase inhibitors could be utilized to temporarily
min markedly improved ovulation and pregnancy rates when release the hypothalamus from the negative feedback effect
combined with CC (Vandermolen et al., 2001). In a large of estrogen so inducing an increased discharge of FSH.
study, 46 anovulatory obese women with PCOS who did not Although the final pathway, the sought-after discharge of
ovulate on metformin or placebo for 35 days were given FSH, is common to both aromatase inhibitors and CC, their
50 mg of CC daily for 5 days while continuing metformin or mechanism of action is obviously very different and this
placebo. Of those on metformin, 19 of 21 ovulated compared would seem to confer several advantages to aromatase inhibi-
with two of 25 on placebo (Nestler et al., 1998). In an inter- tors for the induction of ovulation.
esting RCT, CC-resistant women with PCOS received either Unlike CC, which blockades and depletes estrogen recep-
metformin for 6 months and then CC, or HMG alone for ovu- tors, aromatase inhibitors have no effect on estrogen recep-
lation induction (George et al., 2003). In this small study, as tors. Aromatase inhibitors should, therefore, not have any
metformin þ CC was equally as effective as HMG, less deleterious effect on cervical mucus or endometrium, quite
expensive and more convenient, it was suggested as an inter- frequently a side effect of CC which interferes with the
mediary step for CC-resistant patients, worth trying before attainment of a pregnancy during ovulation induction therapy
resorting to HMG. and is probably the main reason for the gap between
2047
R.Homburg

ovulation and pregnancy rates. This can theoretically be two studies. The first (Mitwally and Casper, 2002) is a report
avoided when aromatase inhibitors are used for the same in which a group of poor responders to FSH for IUI were
purpose. given co-treatment with letrozole, 2.5 mg/day from day 3 to
With aromatase inhibitors, estrogen production is even- day 7 of the cycle. A lower FSH dose and a significantly
tually advanced by the induced FSH discharge but, in con- higher number of mature follicles were achieved with the
trast to the use of CC, the hypothalamus is able to respond to combined treatment. These preliminary findings were con-
the estrogen feedback with a negative feedback mechanism. firmed in a large series, albeit retrospective and non-random-
This will modulate an overzealous discharge of FSH which ized, comparing stimulation with FSH alone (145 cycles) or
in turn is more likely to result in a monofollicular ovulation the combined therapy (60 cycles) (Healey et al., 2003). The
with moderate estrogen concentrations. This is all the more addition of letrozole to gonadotrophin treatment again
poignant as aromatase inhibitors have a much shorter half- decreased the dose of gonadotrophins and increased the num-
life (, 2 days) than CC. This confers an additional theoretical ber of pre-ovulatory follicles. Prospective, randomized trials
advantage to aromatase inhibitors as the prevalence of mul- are needed to verify these interesting findings.
tiple pregnancies could therefore be expected to be less than A subgroup of infertile women have been found to express
that witnessed with the use of CC for ovulation induction. high levels of aromatase P450 in the endometrium, and this
The solid, evidence-based data that are needed to convert was associated with poor IVF outcomes (Brosens et al.,
these hypothetical advantages into demonstrable practical 2004). This raises the interesting question of whether letro-
expression are, at the time of writing, still thin on the ground. zole or anastrozole could alleviate this situation and improve

Downloaded from humrep.oxfordjournals.org by guest on February 25, 2011


However, much groundwork to examine the use of the results.
aromatase inhibitor letrozole in reproductive medicine has Many other questions regarding the use of aromatase
come from the team of Casper and Mitwally. inhibitors in the treatment of infertility still remain (de
In a preliminary trial, 12 anovulatory PCOS patients and Ziegler, 2003). Trials with aromatase inhibitors have, reason-
10 ovulatory patients were given letrozole 2.5 mg/day on ably, mimicked treatment with CC, being administered on
days 3– 7 of the cycle. Endometrial thickness, which had days 3 –7 of the cycle. Would treatment beyond day 7 inter-
been severely depressed in previous CC-treated cycles, was fere with the estradiol rise induced by rising FSH concen-
markedly improved by letrozole which was as efficient as CC trations and have a deleterious effect on the endometrium
for ovulation induction. A direct comparison of letrozole and oocyte quality? In his commentary, de Ziegler (2003)
with 50 mg/day of CC in ovulatory women in an RCT also questions the timing of aromatase inhibitor adminis-
showed similar effects on stimulation of folliculogenesis, tration when the intention is to enhance the sensitivity to
whilst endometrial thickness was maintained by letrozole FSH receptors by increasing follicular androgen content.
despite much lower estradiol concentrations than with CC at Would it not be more logical to prime with aromatase inhibi-
mid-cycle (Fisher et al., 2002). tors before exposure to FSH? Further, although the dose of
A comparison of a very large number of cycles for timed 2.5 mg of letrozole is standard for the treatment of breast
intercourse or IUI included natural (423), CC- (994) and letro- cancer, should the same dose be used for the treatment of
zole- (167) treated cycles (Mitwally and Casper, 2003). The infertility? Biljan et al. (2002), for example, found that a
pregnancy rate with letrozole was more than twice that in daily dose of 5 mg/day produced more mature follicles appar-
natural or CC-stimulated cycles, while both multiple preg- ently by further extending the FSH window.
nancy and abortion rates were significantly lower in letrozole It is a little too early to enthuse optimistically about the
compared with CC cycles. A small pilot study comparing CC chances of letrozole and anastrozole conquering the fertility
(100 mg/day) with letrozole in IUI cycles demonstrated sig- market, but now the initial pilot studies have been completed,
nificantly less estradiol and fewer follicles developing in the I believe that there is enough there to encourage serious trials
letrozole cycles (Fatemi et al., 2003). for this potentially valuable, simple and inoffensive
The conclusion for the moment is that, although some treatment.
groundwork has been done, before aromatase inhibitors can
be regarded as possible replacements for CC for the first-line Low-dose FSH
treatment of anovulatory infertility, some solid, evidence- The third possible alternative to CC as first-line treatment for
based medicine is badly needed. WHO group II infertility is low-dose FSH. In contrast to the
The use of aromatase inhibitors should theoretically result conventional gonadotrophin regimen for treating this group
in an accumulation of androgens whose conversion to estro- of patients, in particular those with PCOS, the low-dose
gens is being blocked. This would, theoretically, be an protocol is designed to attain and maintain follicular
unwanted by-product, especially for women with PCOS who development, ideally one dominant follicle, without exceed-
already have an excessive production of androgens. However, ing the FSH threshold requirement of the ovary (Polson et al.,
paradoxically, this may be a further advantage as androgens 1987). Now in common use, the common complications of
may have a stimulatory role in early follicular growth by conventional gonadotrophin therapy have been severely
augmenting follicular FSH receptor expression and therefore reduced. With low-dose FSH, multiple pregnancy rates
amplifying FSH effects (Weil et al., 1999). This may explain should not exceed 6% and OHSS is almost completely elimi-
the relative success of combined letrozole and FSH for ovar- nated, while acceptable pregnancy rates are achieved
ian stimulation in improving the response to FSH, reported in (Table IV). A consistent rate of 70% of monofollicular
2048
Clomiphene citrate—end of an era?

women, results regarding pregnancy rates have been impeded


Table IV. Results of treatment of clomiphene-resistant patients with low
dose, step-up FSH by obesity, hyperandrogenaemia and a high miscarriage rate,
although multiple pregnancies could be largely avoided
No. of patients 841
No. of cycles 1556 (Homburg et al., 1989).
Pregnancies (% patients) 320 (38%) Laparoscopic ovarian drilling, although a very viable
Fecundity/cycle 20% second- or third-line therapy for anovulation associated with
Uni-ovulation 70%
OHSS 0.14% PCOS, especially in normal weight patients with a high LH
Multiple pregnancies 5.7% concentration, has not seriously been considered as an
alternative to CC as first-line treatment.
Updated from Homburg and Howles (1999).

ovulation in achieved in these cycles (Homburg and Howles, Conclusions


1999). The classical low-dose regimen is step-up, employing
The possibility of replacing CC as first-line treatment for
a low starting dose (usually 50– 75 IU of FSH) with small
anovulatory eu-estrogenic infertility with either metformin,
incremental dose rises (25 – 37.5 IU) on the starting dose if
aromatase inhibitors or low-dose FSH still needs to be sub-
necessary after 14 days and then at weekly intervals. Once
stantiated. RCTs are underway to examine all these possibili-
follicular development is initiated, that dose is maintained
ties, all of which are theoretically well based. At least until
until criteria for HCG administration are reached.
the publication of the results of these trials, CC will remain

Downloaded from humrep.oxfordjournals.org by guest on February 25, 2011


Until now, low-dose FSH has only been administered to
the first-line treatment for eu-estrogenic anovulatory inferti-
CC-resistant patients, and the results in Table IV only refer
lity although, in my humble opinion, aromatase inhibitors
to these patients. Based on this collection of published
and/or low-dose FSH therapy may well take its place in the
results, Table III, column 3 illustrates a projection of these
next few years.
results for 100 patients starting treatment, terminating in 30
singleton live births and two multiple pregnancies. If low-
dose FSH were to be used as first-line treatment, rather than
References
CC, based on the assumption that all those who would have
conceived on CC and 40% of those who would be CC resist- Adashi EY, Rock JA, Sapp KC, Martin EJ, Wentz AC and Jones GS (1979)
Gestational outcome of clomiphene-related conceptions. Fertil Steril
ant would also conceive on low-dose FSH, 45 of 100 starters 31,620–626.
could expect a singleton live birth and three a multiple preg- Agarwal SK and Buyalos RP (1995) Corpus luteum function and pregnancy
nancy (Table III, column 4). If this supposition is confirmed, rates with clomiphene citrate therapy: comparison of human chorionic
then the treatment – conception interval would be consider- gonadotrophin-induced versus spontaneous ovulation. Hum Reprod 10,
328 –331.
ably reduced and efficiency greatly increased by eliminating Ahlgren M, Kallen B and Rannevik G (1976) Outcome of pregnancy after
the time and the need for monitoring and treatment costs of clomiphene therapy. Acta Obstet Gynecol Scand 55,371– 375.
3 –12 cycles of CC therapy. Although costs of medication Attia GR, Rainey WE and Carr BR (2001) Metformin directly inhibits andro-
and patient comfort obviously differ, these may well be gen production in human thecal cells. Fertil Steril 76,517– 524.
Baillargeon JP, Jakubowicz DJ, Iuorno MJ, Jakubowicz S and Nestler JE
negated by a much more efficient first-line treatment.
(2004) Effects of metformin and rosiglitazone, alone and in combination,
One, single centre, randomized trial has tested this hypoth- in nonobese women with polycystic ovary syndrome and normal indices
esis (Lopez et al., 2004) but, unfortunately, the size of the of insulin sensitivity. Fertil Steril 82,893–902.
groups (38 patients in each arm) did not allow firm con- Biljan MM, Tan SL and Tulandi T (2002) Prospective randomized trial com-
paring the effects of 2.5 and 5.0 mg of letrozole (LE) on follicular devel-
clusions. Nevertheless, the RR and 95% confidence intervals opment, endometrial thickness and pregnancy rates in patients undergoing
were 1.78 (0.92 – 3.54) for pregnancy rate per woman and superovulation. Fertil Steril,S55.
1.83 (0.79 – 4.40) for live births per woman in favour of FSH. Brosens J, Verhoeven H, Campo R, Gianaroli L, Gordts S, Hazekamp J,
These are almost identical figures to those that had been pre- Hagglund L, Mardesic T, Varila E, Zech J and Brosens I (2004) High
endometrial aromatase P450 mRNA expression is associated with poor
dicted, prior to this study, in Table IV, based on assumptions IVF outcome. Hum Reprod 19,352–356.
and previous results (1.72 for pregnancy rates and 1.80 for Correy JF, Marsden DE and Schokman FC (1982) The outcome of preg-
live birth rates in favour of FSH). nancy resulting from clomiphene-induced ovulation. Aust N Z J Obstet
A sufficiently powered, multicentre, multinational trial is Gynaecol 22,18–21.
Daly DC, Walters CA, Soto-Albors, Tohan N and Riddick DH (1984) A ran-
now well under way to compare CC with low-dose FSH as domized study of dexamethasone in ovulation induction with clomiphene
first-line treatment for women with anovulatory infertility citrate. Fertil Steril 41,844–848.
associated with PCOS. Combined with a cost-efficiency de Ziegler D (2003) The dawning of the non-cancer uses of aromatase inhibi-
assessment, it will tell us whether replacing CC with tors in gynaecology. Hum Reprod 18,1598–1602.
Dickey RP, Taylor SN, Curole DN et al. (1996) Incidence of spontaneous
low-dose FSH is a feasible proposition.
abortion in clomiphene pregnancies. Hum Reprod 11,2623–2628.
Dickey RP, Taylor SN, Curole DN, Rye PH, Lu PY and Pyrzak R (1997)
Other possible first-line therapies Relationship of clomiphene dose and patient weight to successful treat-
The use of pulsatile GnRH for the treatment of anovulatory ment. Hum Reprod 12,449–453.
Eijkemans MJ, Habbema JD and Fauser BC (2003) Characteristics of the
(WHO group II) infertility has largely been abandoned.
best prognostic evidence: an example on prediction of outcome after
Although a normal pattern of pulsatile release of GnRH can clomiphene citrate induction of ovulation in normogonadotropic oligoame-
be superimposed on the abnormal pattern found in these norrheic infertility. Semin Reprod Med 21,39–47.

2049
R.Homburg

Fatemi HM, Kolibianakis E, Tournaye H, Camus M, Van Steirteghem AC Jakubowicz DJ, Iuorno MJ, Jakubowicz S, Roberts KA and Nestler JE
and Devroey P (2003) Clomiphene citrate versus letrozole for ovarian (2002) Effects of metformin on early pregnancy loss in the polycystic
stimulation: a pilot study. Reprod Biomed Online 7,543–546. ovary syndrome. J Clin Endocrinol Metab 87,524–529.
Fisher SA, Reid RL, Van Vugt DA and Casper RFA (2002) randomized Kashyap S, Wells GA and Rosenwaks Z (2004) Insulin-sensitizing agents as
double-blind comparison of the effects of clomiphene citrate and the primary therapy for patients with polycystic ovarian syndrome. Hum
aromatase inhibitor letrozole on ovulatory function in normal women. Reprod 19,2474–2483.
Fertil Steril 78,280–285. Kousta E, White DM and Frank S (1997) Modern use of clomiphene citrate
Fleming R, Hopkinson ZE, Wallace AM, Greer IA and Sattar N (2002) in induction of ovulation. Hum Reprod Update 3,359– 365.
Ovarian function and metabolic factors in women with oligomenorrhea Lobo RA, Gysler M, March CM, Goebelsmann U and Mishell DR Jr (1982)
treated with metformin in a randomized double blind placebo-controlled Clinical and laboratory predictors of clomiphene response. Fertil Steril 37,
trial. J Clin Endocrinol Metab 87,569–574. 168–174.
Garcia J, Jones GS and Wentz AC (1977) The use of clomiphene citrate. Lopez E, Gunby J, Daya S, Parrilla JJ, Abad L and Balasch J (2004) Ovu-
Fertil Steril 28,707–717. lation induction in women with polycystic ovary syndrome: randomized
George SS, George K, Irwin C, Job V, Selvakumar R, Jeyaseelan V and trial of clomiphene citrate versus low-dose recombinant FSH as first line
therapy. Reprod Biomed Online 9,382–390.
Seshadri MS (2003) Sequential treatment of metformin and clomiphene
citrate in clomiphene-resistant women with polycystic ovary syndrome: a Lord JM, Flight IH and Norman RJ (2003) Insulin-sensitising drugs (metfor-
randomized, controlled trial. Hum Reprod 18,299–304. min, troglitazone, rosiglitazone, pioglitazone, d-chiro-inositol) for polycys-
tic ovary syndrome. Cochrane Database Syst Rev,CD003053.
Ghazeeri G, Kutteh WH, Bryer-Ash M, Haas D and Ke RW (2003) Effect of
Macgregor AH, Johnson JE and Bunde CA (1968) Further clinical experi-
rosiglitazone on spontaneous and clomiphene citrate-induced ovulation in
ence with clomiphene citrate. Fertil Steril 19,616– 622.
women with polycystic ovary syndrome. Fertil Steril 79,562–566.
Mansfield R, Galea R, Brincat M, Hole D and Mason H (2003) Metformin
Glueck CJ, Wang P, Goldenberg N and Sieve-Smith L (2002) Pregnancy out-
has direct effects on human ovarian steroidogenesis. Fertil Steril 79,
comes among women with polycystic ovary syndrome treated with metfor- 956–962.
min. Hum Reprod 17,2858–2864.

Downloaded from humrep.oxfordjournals.org by guest on February 25, 2011


Messinis IE and Milingos SD (1998) Future use of clomiphene in ovarian
Greenblatt RB, Bafrield WE, Jungck EC and Ray AW (1961) Induction of stimulation. Clomiphene in the 21st century. Hum Reprod 13,2362– 2365.
ovulation with MRL/41. Preliminary report. J Am Med Assoc 178,
Mitwally MF and Casper RF (2001) Use of an aromatase inhibitor for induc-
101– 104. tion of ovulation in patients with an inadequate response to clomiphene
Guzick DS, Sullivan MW, Adamson GD, Cedars MI, Falk RJ, Peterson EP citrate. Fertil Steril 75,305–309.
and Steinkampf MP (1998) Efficacy of treatment for unexplained inferti- Mitwally MF and Casper RF (2002) Aromatase inhibition improves ovarian
lity. Fertil Steril 70,207–213. response to follicle-stimulating hormone in poor responders. Fertil Steril
Gysler M, March CM, Mishell DR Jr and Bailey EJ (1982) A decade’s 77,776–780.
experience with an individualized clomiphene treatment regimen including Mitwally MF and Casper RF (2003) Aromatase inhibitors for the treatment
its effect on the postcoital test. Fertil Steril 37,161–167. of infertility. Expert Opin Invest Drugs 12,353–371.
Hammond MG (1984) Monitoring techniques for improved pregnancy rates Moghetti P, Castello R, Negri C, Tos F, Perrone F, Caputo M, Zanolin E and
during clomiphene ovulation induction. Fertil Steril 42,499–509. Muggeo M (2000) Metformin effects on clinical features, endocrine and
Harborne L, Fleming R, Lyall H, Norman J, Sattar N et al. (2003) Descrip- metabolic profiles, and insulin sensitivity in polycystic ovary syndrome: a
tive review of the evidence for the use of metformin in polycystic ovary randomized, double-blind, placebo-controlled 6-month trial, followed by
syndrome. Lancet 361,1894–1901. open, long-term clinical evaluation. J Clin Endocrinol Metab 85,139– 146.
Healey S, Tan SL, Tulandi T and Biljan MM (2003) Effects of letrozole on Nestler JE, Jakubowicz DJ, Evans WS and Pasquali R (1998) Effects of met-
superovulation with gonadotropins in women undergoing intrauterine inse- formin on spontaneous and clomiphene-induced ovulation in the polycystic
mination. Fertil Steril 80,1325– 1329. ovary syndrome. N Engl J Med 338,1876–1880.
Homburg R (2002) Should patients with polycystic ovarian syndrome be Nestler JE, Stovall D, Akhter N, Iuorno MJ and Jakubowicz DJ (2002) Strat-
treated with metformin? A note of cautious optimism. Hum Reprod egies for the use of insulin-sensitizing drugs to treat infertility in women
17,853–856. with polycystic ovary syndrome. Fertil Steril 77,209–215.
Homburg R and Howles CM (1999) Low-dose FSH therapy for anovulatory Polson DW, Mason HD, Saldahna MB and Franks S (1987) Ovulation of a
infertility associated with polycystic ovary syndrome: rationale, results, single dominant follicle during treatment with low-dose pulsatile follicle
reflections and refinements. Hum Reprod Update 5,493–499. stimulating hormone in women with polycystic ovary syndrome. Clin
Endocrinol (Oxf) 26,205–212.
Homburg R and Insler V (2002) Ovulation induction in perspective. Hum
Reprod Update 8,449–462. Polson DW, Kiddy DS, Mason HD and Franks S (1989) Induction of ovu-
lation with clomiphene citrate in women with polycystic ovary syndrome:
Homburg R, Armar NA, Eshel A, Adams J and Jacobs HS (1988a) Influence
the difference between responders and nonresponders. Fertil Steril 51,
of serum luteinising hormone concentrations on ovulation, conception, and 30–34.
early pregnancy loss in polycystic ovary syndrome. Br Med J 297,
Regan L, Owen EJ and Jacobs HS (1990) Hypersecretion of luteinising hor-
1024– 1026.
mone, infertility, and miscarriage. Lancet 336,1141–1144.
Homburg R, Weissglas L and Goldman J (1988b) Improved treatment for Rostami-Hodjegan A, Lennard MS, Tucker GT and Ledger WL (2004)
anovulation in polycystic ovarian disease utilizing the effect of progester- Monitoring plasma concentrations to individualize treatment with clomi-
one on the inappropriate gonadotrophin release and clomiphene response. phene citrate. Fertil Steril 81,1187–1193.
Hum Reprod 3,285– 288.
Schenker JG, Yarkoni S and Granat M (1981) Multiple pregnancies follow-
Homburg R, Eshel A, Tucker M, Mason P, Adams J, Kilborn J, ing induction of ovulation. Fertil Steril 35,105–123.
Sutherland IA and Jacobs HS (1989) One hundred pregnancies following Scialli AR (1986) The reproductive toxicity of ovulation induction. Fertil
pulsatile luteinising hormone-releasing hormone therapy for ovulation Steril 45,315–323.
induction. Brit Med J 298,809–912. Shoham Z, Borenstein R, Lunenfeld B and Pariente C (1990) Hormonal pro-
Ibanez L, Valls C, Ferrer A, Marcos MV, Rodriguez-Hierro F and files following clomiphene citrate therapy in conception and nonconcep-
de Zegher F (2001) Sensitization to insulin induces ovulation in nonobese tion cycles. Clin Endocrinol (Oxf) 33,271–278.
adolescents with anovulatory hyperandrogenism. J Clin Endocrinol Metab Vandermolen DT, Ratts VS, Evans WS, Stovall DW, Kauma SW and Nestler
86,3595–3598. JE (2001) Metformin increases the ovulatory rate and pregnancy rate from
Imani B, Eijkemans MJ, te Velde ER, Habbema JD and Fauser BC (1998) clomiphene citrate in patients with polycystic ovary syndrome who are
Predictors of patients remaining anovulatory during clomiphene citrate resistant to clomiphene citrate alone. Fertil Steril 75,310–315.
induction of ovulation in normogonadotropic oligoamenorrheic infertility. Velazquez E, Acosta A and Mendoza SG (1997) Menstrual cyclicity after
J Clin Endocrinol Metab 83,2361–2365. metformin therapy in polycystic ovary syndrome. Obstet Gynecol 90,
Imani B, Eijkemans MJ, te Velde ER, Habbema JD and Fauser BC (2002) A 392–395.
nomogram to predict the probability of live birth after clomiphene citrate Warburton D and Fraser FC (1964) On the probability that a woman who
induction of ovulation in normogonadotropic oligoamenorrheic infertility. has had a spontaneous abortion will abort in subsequent pregnancies.
Fertil Steril 77,91–97. J Obstet Gynaecol Br Commonw 68,784–789.

2050
Clomiphene citrate—end of an era?

Weil S, Vendola K, Zhou J and Bondy CA (1999) Androgen and follicle- Wu CH and Winkel CA (1989) The effect of therapy initiation day on clomi-
stimulating hormone interactions in primate ovarian follicle development. phene citrate therapy. Fertil Steril 52,564–568.
J Clin Endocrinol Metab 84,2951–2956.
Wentz AC and Cartwright PS (1988) Recurrent and spontaneous abortions.
In Jones HW, Wentz AC, and Burnett LS (eds) Novak’s Textbook of Submitted on November 12, 2004; resubmitted on February 13, 2005;
Gynecology. Williams and Wilkins, Baltimore, MD, pp. 328 –350. accepted on April 3, 2005

Downloaded from humrep.oxfordjournals.org by guest on February 25, 2011

2051

Das könnte Ihnen auch gefallen