Sie sind auf Seite 1von 15

GYNECOLOGY AND MENOPAUSE

Selective progesterone receptor modulators


in reproductive medicine: pharmacology, clinical
efficacy and safety
Philippe Bouchard, M.D.,a,b Nathalie Chabbert-Buffet, M.D., Ph.D.,b,c and Bart C. J. M. Fauser, M.D., Ph.D.d
a
Endocrinology Unit, AP-HP Hospital Saint-Antoine, Paris, France; b University Pierre and Marie Curie, Paris, France;
c
Department of Obstetrics and Gynecology, AP-HP Hospital Tenon, Paris, France; d Department of Reproductive Medicine
and Gynecology, University Medical Center Utrecht, Utrecht, the Netherlands

Objective: To discuss the mechanism of action of selective progesterone receptor modulators (SPRMs) and sum-
marize the preclinical and clinical efficacy and safety data supporting the potential use of these compounds for
gynecologic indications.
Design: Relevant publications from 2005 onward were identified using a PubMed search. Additional relevant
articles were identified from citations within these publications.
Setting: None.
Patient(s): None.
Intervention(s): None.
Main Outcome Measure(s): None.
Result(s): Mifepristone was first developed as a progesterone receptor antagonist and licensed for pregnancy termi-
nation because of the unique property of this compound to terminate pregnancy when associated with prostaglandins.
Then SPRMs were developed, and among those ulipristal acetate, an efficient emergency contraceptive. Because
SPRMs effectively inhibit endometrial proliferation and reduce endometriotic lesions in animal models, this suggests
a possible role in the treatment of endometriosis in humans. Finally, a number of double-blind, randomized, placebo-
controlled trials have demonstrated the efficacy of asoprisnil, mifepristone, telapristone acetate, and ulipristal acetate
in reducing leiomyoma and uterine volume, and suppressing bleeding in women with uterine fibroids.
Conclusion(s): Mifepristone in combination with prostaglandins has been licensed for pregnancy termination
because of its unique ability is this area. Ulipristal acetate is available for emergency contraception. Several SPRMs
hold further promise as an effective medical therapy for patients suffering from endometriosis and leiomyoma. (Fer-
til Steril 2011;96:1175–89. 2011 by American Society for Reproductive Medicine.)
Key Words: Amenorrhea, contraception, leiomyoma, progesterone receptor, selective progesterone receptor
modulator, SPRM

Progesterone exerts biologic effects on a range of organ systems, in- tion of the female reproductive system, including during pregnancy
cluding the cardiovascular and central nervous systems, and bone. and mammary gland development (1). Progesterone mediates its
However, it is probably best known for its pivotal role in the regula- function by interacting with the progesterone receptor (PR), a mem-
ber of a superfamily of almost 50 ligand-activated nuclear transcrip-
Received March 21, 2011; revised August 8, 2011; accepted August 12, tion factors, which can be divided into six subfamilies (2). Other
2011; published online September 22, 2011. members of this large family of receptors include the glucocorticoid,
P.B. has received fees and grants from Ferring, HRA Pharma, Nordic androgen, estrogen, thyroid hormone, and retinoic acid receptors.
Pharma, Pierre Fabre, Schering Plough, Preglem, Pantarhei Bioscience, The profound importance of progesterone in the female reproduc-
Serono, and Wyeth, is a senior consultant at the Population Council
tive system has led to the development of synthetic PR ligands with
(New York), and is a member of the International Committee on Contra-
ceptive Research. N.C-B. has been investigator in studies funded by the
both agonistic and antagonistic properties. The first PR antagonist to
following companies: HRA Pharma, Organon, Theramex. B.C.J.M.F. be identified was RU486 (mifepristone), discovered in 1980 by
has received fees and grant support from the following companies: Teusch et al. at Roussel during their research on antiglucocorticoid
Andromed, Ardana, Ferring, Genovum, Glycotope, Merck, Merck agents (3). This discovery was followed by the development of other
Serono, Organon, Pantharei Bioscience, Philips, Preglem, Schering, steroidal progesterone ligands, while nonsteroidal PR ligands have
Schering Plough, Serono, and Wyeth. been identified more recently (4). In general, PR ligands with mixed
Editorial assistance was provided by Adelphi Communications, supported
activity act as agonists and/or antagonists in a tissue-specific man-
by PregLem SA.
Reprint requests: Philippe Bouchard, Service d’Endocrinologie, Ho ^ pital ner. These compounds therefore have great potential in a number
Saint Antoine 184 rue du Faubourg Saint Antoine 75012 Paris, France of gynecologic indications. This review will summarize what is cur-
(E-mail: philippe.bouchard@sat.aphp.fr). rently known about the mechanism of action of such molecules, their

0015-0282/$36.00 Fertility and Sterility Vol. 96, No. 5, November 2011 1175
doi:10.1016/j.fertnstert.2011.08.021 Copyright ª2011 American Society for Reproductive Medicine, Published by Elsevier Inc.
effects on tissues of the reproductive system, and the data demon- Selective Progesterone Receptor Modulators
strating their potential efficacy for gynecologic indications. The PR ligands can possess activity ranging from pure agonist activ-
ity through mixed agonist/antagonist activity to pure antagonist ac-
tivity. The McPhail test, in which the progestational activity of
MATERIALS AND METHODS
a substance is tested in estrogen-primed immature rabbits, remains
A PubMed search for relevant publications from 2005 onward was per-
the standard test for determining the PR agonist/antagonist activity
formed, using the following search strategy: ‘‘sprm OR progesterone receptor
modulator OR asoprisnil OR J867 OR mifepristone OR onapristone OR
of a compound (25, 26). However, while the McPhail test is
mixed profile progestagen OR ulipristal OR CDB-2914 OR VA2914 OR informative, it is apparently unable to distinguish between minor
CDB2914 OR CDB-4124 OR CDB4124.’’ The titles of the 1,548 articles differences in PR antagonistic activity. Using the McPhail test,
identified in this search were manually screened to identify those publica- mifepristone is characterized as a ‘‘pure’’ antagonist (26). Current
tions containing relevant information. Final searches using the following distinctions, based on traditional model systems, between molecules
search terms were performed without any date restrictions to ensure no rel- with apparently antagonistic functions and those with a mixed pro-
evant publications had been missed (n denotes number of publications iden- file may be somewhat imprecise. Indeed, all SPRMs potentially
tified): ‘‘CDB-2914 OR VA2914 OR CDB2914’’ (n ¼ 46); ‘‘CDB-4124 OR block ovulation and have similar effects on leiomyoma growth
CDB4124’’ (n ¼ 11); ‘‘asoprisnil OR J867’’ (n ¼ 39); ‘‘sprm OR progester- and endometrial changes. In addition, their effect on the uterus dif-
one receptor modulator’’ (n ¼ 298). Additional relevant articles were identi-
fers from those described in mice, where deletion of PR genes leads
fied from citations within all publications identified. In addition, information
about selective progesterone receptor modulators (SPRMs) in clinical or pre-
to pure endometrial hyperplasia (16). However, subtle differences
clinical development was obtained using Citeline Pipeline (Informa Health- have been described in terms of endometrial changes, which may re-
care). This review focuses on articles published since 2005 because trials flect the relative influence of the agonist and antagonist actions.
before 2005 were previously reviewed elsewhere (5–7). These more subtle differences make it difficult to compare dose re-
sponse effects between different agents. A new classification ap-
proach based on transcriptional activity in vitro has recently been
PROGESTERONE AND THE PROGESTERONE RECEPTOR suggested (27). This approach appears to refine the classification
Within the female reproductive system, the key function of proges- of PR ligands, demonstrating the unique activity of each SPRM.
terone is to establish and maintain pregnancy (8, 9). Progesterone In this review, the term selective progesterone receptor modulator
plays a crucial role in breast differentiation and antagonizes the (SPRM) will be used to describe all PR ligands that show some degree
proliferative effects of estradiol on the endometrium, while of cell and tissue context-dependent agonistic and antagonistic activ-
allowing the expression of genes involved in embryo implantation ity (28). Among all SPRMs studied, mifepristone, the pioneer drug,
(8, 10, 11). During pregnancy, progesterone secretion is associated somehow remains a separate entity because its properties as an antag-
with quiescence of the myometrium, and its decrease during labor onist are unique and because mifepristone is the only SPRM that is
is one of the signals for delivery (9). able to interrupt pregnancy in several species, including humans.
Progesterone has long been known to bind to the PR, inducing
conformational changes leading to binding of the progesterone–
PR protein complex to progesterone response elements in the pro- Molecular Mechanism of Action of SPRMs
moters of target genes. Once bound to the target genes, the proges- Like progesterone, SPRM molecules interact with the PR, allowing
terone–PR complex interacts with coactivators, leading to the binding of PR dimers to target gene promoters. However, the
transcription of the target genes (12, 13) (Fig. 1A). Confirmation conformation induced by each SPRM promotes interaction of the
that progesterone mainly mediates reproductive functions via inter- PR with not only coactivators but also corepressors, leading to
action with the PR has come from analysis of mice homozygous for mixed agonist/antagonist activity depending on their structure and
disruption of the PR gene (16). Adult female mice lacking the PR the relative tissue concentrations of these comodulators (29) (see
demonstrate a complete absence of ovulation, uterine hyperplasia Fig. 1B). Limited information is available regarding the specific af-
and inflammation, a lack of mammary gland development, and an finities of different SPRMs for the PR, and the differential interac-
inability to display appropriate sexual behavior (16). Two main tion of SPRMs with the PR-A and PR-B isoforms. However, it has
PR isoforms have been described, which have distinct biologic ac- been demonstrated that activation of transcription induced by
tivities and target genes. The PR-A isoform is similar to the PR-B a PR-B–bound SPRM may not involve binding to canonical hor-
isoform except that the PR-A form lacks the 164 N-terminal amino mone responsive elements and can be inhibited by PR-A in HeLa
acids found in PR-B (17, 18). PR-A appears to be a dominant inhib- cells (31). Moreover, the activities of PR isoforms are differentially
itor of PR-B (19). Specific deletion of PR isoforms has provided ad- modulated by SPRMs in breast cancer cell lines (32). In addition,
ditional information on their roles in mice. While PR-A controls coregulator shuttling and degradation is a crucial step in PR-
estrogen induced endometrial proliferation, PR-B mediates mam- mediated transcriptional activity; the PR agonist R5020 induces
mary gland epithelium proliferation and differentiation (20, 21). this recycling phenomenon, but mifepristone does not, and even fur-
Although it is clear that the tissue ratio of PR-A/PR-B is crucial ther impairs the agonist-induced SRC1 degradation (33). Finally, PR
for the SPRM effect, it should be noted that the relevance of these isoforms have also been shown to have distinct affinities for different
isoforms in terms of SPRM efficacy in vivo is still uncertain, partic- coregulators (19), a mechanism that may also be involved in the bi-
ularly as all SPRMs bind to both isoforms, which share identical ologic action. However, the physiologic relevance of these different
ligand-binding domains. Therefore, the physiologic relevance of regulatory mechanisms has not been confirmed in vivo thus far.
these in vitro findings remains hypothetical. However, unbalanced Several SPRMs developed to date can interact with steroid recep-
PR-A/PR-B ratios have been observed in breast and endometrial tors other than the PR to a greater or lesser extent (34). For example,
cancer (22, 23). Progesterone is also thought to play mifepristone binds to the glucocorticoid receptor (GR) with an affin-
a reproductive role as a neuroendocrine switch in the brain by ity three to four times higher than that of dexamethasone and conse-
attenuating the pulsatile release of gonadotropin-releasing hormone, quently may be of therapeutic value in the treatment of Cushing
which plays a part in regulating the luteinizing hormone surge (24). syndrome (35). In contrast, most other SPRMs bind weakly to the

1176 Bouchard et al. SPRMs in reproductive medicine Vol. 96, No. 5, November 2011
FIGURE 1
Mechanism of action of progesterone and selective progesterone receptor modulators (SPRMs). (A) After entering the cytoplasm of the target
cell, progesterone binds to the progesterone receptor (PR) which causes a conformational change in the PR, resulting in the dissociation of
chaperone proteins, PR dimerization, and binding of the progesterone–PR protein complex to progesterone response elements in the
promoters of target genes. Two major different isoforms of PR (PR-A and PR-B) exert different gene selective biological activities. This allows
the progesterone–PR protein complex to interact with coactivators, facilitating communication with the basal transcription apparatus and
ultimately leading to transcription of the target gene (12, 13). Steroid hormone receptors, including the PR, can also activate nongenomic
signaling pathways in the absence of steroid hormone (14, 15). (B) Interaction of SPRMs with the progesterone receptor isoforms (PR-A/B)
induces a conformational change in the PR, which allows a more potent recruitment of corepressors (such as nuclear receptor corepressor
(NCoR) and silencing mediator of retinoid and thyroid hormone receptor [SMRT]) to the PR than that induced by progesterone. However,
under these conditions the PR ligand-binding domain can also interact with the coactivators SRC-1 and AIB1 (29). As the precise
conformational change induced in the PR, and thus the balance of interaction with coactivators and corepressors depends upon the identity of
the individual SPRM molecule to which the PR is bound, each SPRM has a different and unique molecular signature (7, 28–30). Furthermore,
the precise activity of each SPRM will vary by tissue, depending on the relative levels of coactivators and corepressors in each cellular
environment.

Bouchard. SPRMs in reproductive medicine. Fertil Steril 2011.

GR (26, 36, 37). Nonsteroidal SPRMs, which may be more selective only a few are being pursued for clinical use at present, and only two
for the PR, are also in development (34, 38, 39). More recently, are currently licensed for gynecologic use. Mifepristone has been
a class of SPRMs, called mixed-profile progestogens, has been de- approved in more than 30 countries for the termination of pregnancy,
scribed. These compounds are reported to have more agonistic activ- cervical dilation, medical termination of pregnancy during the sec-
ity than the currently described SPRMs (40). ond trimester, and fetal death in utero (41, 42), and a single dose of
ulipristal acetate (30 mg) has been launched in Europe and the
Major SPRMs Currently Available and Currently in United States as an emergency contraceptive.
Development
The properties of selected SPRMs in clinical development and those SPRMS AS CONTRACEPTIVES
that have shown promise in preclinical or clinical research studies The critical role of progesterone in ovulation and preparing the en-
are summarized in Table 1. The chemical structures are shown in dometrium for implantation suggests that SPRMs may have poten-
Figure 2. Although a large number of SPRMs have been identified, tial for use as contraceptives.

Fertility and Sterility 1177


TABLE 1
Selected major selective progesterone receptor modulators (SPRMs) with current or past development noted in the literature.

Compound Therapeutic applicationa Current status

Mifepristone (RU-486) Termination of pregnancy Launched


Emergency contraception Phase III
Psychosisb Phase III
Cushing diseaseb Phase III
Long-term contraception Phase II
Uterine fibroids Phase II
Endometriosis Phase II
Alzheimer diseaseb Phase II
Endometrial cancer Phase I
Ulipristal acetate (CDB-2914; VA2914) Emergency contraception Launched
Uterine fibroids Phase III
Long-term contraceptionc Phase II
Asoprisnil (J-867) Uterine fibroids Withdrawn
Endometriosis Withdrawn
Long-term contraception Withdrawn
Telapristone acetate (CDB-4124) Uterine fibroids Phase III trials terminated; phase I/II trial initiated
Anemia Phase III trials terminated; phase I/II trial initiated
Endometriosis Phase II trials terminated; phase I/II trial initiated
Lonaprisan (ZK230211) Cancer Phase II
CP8816 and CP8863 Gynecologic disorders Preclinical
WAY-255348 Contraception Preclinical
Onapristone (ZK98299) Endometriosis No development since the 1990s
Cancer No development since the 1990s
ORG-31710 and ORG-31806 Contraception Preclinical; no development since the 1990s
Cancer Preclinical; no development since the 1990s
PF-2413873 Endometriosis Discontinued
ZK137316 Contraception Discontinued
a
Including licensed uses and those for which the drug has undergone trials or for which it has or had been marked for development.
b
Indications in development due to antiglucocorticoid activity of mifepristone.
c
Using a vaginal ring formulation.

Bouchard. SPRMs in reproductive medicine. Fertil Steril 2011.

Rationale interfere with blastocyst attachment (59). The short-term adminis-


Ovulation is blocked in women receiving mifepristone continu- tration of SPRMs for emergency contraception may induce a specific
ously at daily doses of 2 mg or more (43), and high rates of an- endometrial effect as compared with a progestin used for the same
ovulation are observed in women treated with daily ulipristal purpose (58). This effect has not been documented during long-
acetate, 5 mg or 10 mg (44). In contrast, although asoprisnil pro- term administration.
longs the menstrual cycle in a dose-dependent manner, its effect
on ovulation is inconsistent and does not show dose dependency Clinical Studies
(45). At high doses, continuous administration of mifepristone Emergency contraception Many clinical data exist to support the
can suppress follicular development in addition to preventing use of SPRMs as contraceptives. Key trials in this area from 2005
ovulation (46, 47). onward are summarized in Table 2; trials before 2005 have been
It is important, however, that despite their effects on ovula- reviewed previously elsewhere (5, 6). A number of large studies
tion, SPRMs do not result in the down-regulation of ovarian es- have demonstrated the efficacy of mifepristone for emergency
trogen levels, allowing estrogen concentrations to remain within contraception (see Table 2), and a recent meta-analysis encompass-
physiologic levels (43–51). Administration of a single dose of ing 45,842 women has confirmed that middle- or low-dose
mifepristone or ulipristal acetate also delays ovulation (52–56), mifepristone is more effective for emergency contraception than lev-
a property that allows the use of these compounds for onorgestrel (81). A single 30-mg dose of ulipristal acetate is also
emergency contraception. very effective as an emergency contraceptive, with a longer duration
Studies in baboons and humans have demonstrated that single of effect (120 hours) than levonorgestrel (61, 63). Ulipristal acetate
doses of CDB 2914 or ZK 137-316 also alter endometrial develop- has been approved for emergency contraception in 29 countries thus
ment (49, 57). Moreover, short treatment of the endometrium with far, and is commercially available in 25 of them.
mifepristone or ORG-31710 can inhibit blastocyst attachment
(58, 59) in ex vivo endometrial culture models. This may be Long-term contraception Development of SPRMs for long-term
related to the down-regulation of endometrial receptivity markers contraception is less advanced, but several trials have shown that
(60). Longer term treatment with ORG-31710 in the ex vivo baboon mifepristone administered either daily or weekly can be an effective
endometrial culture model did not induce modifications that could contraceptive, while ulipristal acetate (5 mg or 10 mg daily) induces

1178 Bouchard et al. SPRMs in reproductive medicine Vol. 96, No. 5, November 2011
These reports include a number of review articles (41, 82–84), which
FIGURE 2 will not be duplicated here. It should also be noted that mifepristone
Chemical structures of progesterone, norethindrone, and selected is not effective for the termination of pregnancy alone, but only in
selective progesterone receptor modulators. combination with a prostaglandin (82).

SPRMS FOR THE TREATMENT OF ENDOMETRIOSIS


Endometriosis—the growth of endometrial glands and stroma out-
side of the uterus (85, 86)—has been estimated to be present in up
to 10% of women of reproductive age and represents a significant
cause of pelvic pain and infertility (87, 88). The only definitive,
long-term treatment for endometriosis is abdominal hysterectomy
with removal of ovaries and all visible endometriosis (85). Such
radical therapy is clearly not appropriate for women who have
not completed their families. Medical treatments usually include
oral contraceptives, progestins, or gonadotropin-releasing hor-
mone (GnRH) agonists. However, these two latter therapies
suppress estrogen secretion and therefore induce a significant de-
crease in bone mass in addition to hot flushes and vaginal dryness,
which impair the quality of life of those women receiving the
treatments (88).

Rationale
An ideal medical therapy for endometriosis should be able to ame-
liorate pain and suppress endometrial proliferation while avoiding
a hypoestrogenic state. A wealth of data has demonstrated that
SPRMs inhibit endometrial proliferation. In nonhuman primates,
a number of SPRMs, including ulipristal acetate, asoprisnil, PRA-
910, and onapristone suppress endometrial proliferation, resulting
in endometrial atrophy (48, 89–91). Similar suppression has been
described in human cell lines after treatment with ulipristal
acetate or mifepristone (92, 93), and in subjects treated with
SPRMs including asoprisnil, lonaprisan, mifepristone, or
telapristone acetate (50, 94, 95, 96–98). Further evidence for the
potential of SPRMs in endometriosis comes from the suppression
of endometriotic lesions in animal models by mifepristone,
onapristone, and ZK136799 (99, 100). Moreover, some SPRMs
can suppress endometrial prostaglandin production in mammalian
model systems (26, 101, 102), potentially reducing endometrial-
associated pain. The relevance of these nonhuman models remains
uncertain because SPRMs do not behave similarly in terms of endo-
metrial effects in nonhuman primates and in women (7).

Clinical Studies
A small number of clinical studies have demonstrated that SPRMs
Bouchard. SPRMs in reproductive medicine. Fertil Steril 2011. and PR antagonists have potential for the treatment of endometri-
osis. Mifepristone at 50 mg daily has been shown to improve pain
and cause regression of endometriosis (103), although a lower
anovulation in most women. The potential of asoprisnil for dose of 5 mg daily is unable to control the growth of endometriotic
long-term contraception is less clear (see Table 2). The use of lesions (104). Mifepristone-loaded subcutaneous implants may also
SPRMs for long-term contraception is of great interest as it would be an effective treatment for endometriosis (105). Asoprisnil and te-
provide an estrogen-free method. However, the use of SPRMs for lapristone acetate have also been reported to relieve pain associated
this indication cannot progress until the long-term effects of SPRMs with endometriosis (7, 106, 107). These beneficial effects of SPRM
on the endometrium have been clarified. treatment may reflect changes in the endometrial morphology and/or
the absence of bleeding. However, the potential consequences of
Mifepristone for termination of pregnancy Mifepristone is the only progesterone receptor modulator-associated endometrial change
SPRM used for pregnancy termination because of its unique ability (PAEC) formation on endometriotic lesions remain unknown, and
to terminate pregnancy in women. Other SPRMs have not been stud- long-term treatment of endometriosis with SPRMs will have to
ied for this indication (26). A wealth of published data describes the wait until the long-term effect of SPRMs on the endometrium are de-
efficacy of mifepristone at doses between 200 mg and 600 mg, fol- termined. In the future, intermittent treatment regimens with SPRMs
lowed by a prostaglandin, for the medical termination of pregnancy. might be appropriate (107).

Fertility and Sterility 1179


1180
TABLE 2
Key clinical trials since 2005 describing the use of selective progesterone receptor modulators (SPRMs) for contraception and uterine leiomyomata.
Bouchard et al.

Treatment Treatment
Study Design and dose n duration/timing Key results

Contraception
Ulipristal acetate
Emergency contraception
Creinin et al., 2006 (61) R, DB %72 h after unprotected Pregnancy rate, %
intercourse
Ulipristal acetate (1  50 mg) 775 0.9
SPRMs in reproductive medicine

Levonorgestrel (2  0.75 mg) 774 1.7


Arms had similar side
effect profiles
Fine et al., 2010 (62) OL Ulipristal acetate (1  30 mg) 1,241 48–120 h after unprotected 2.1% pregnancy rate (satisfies
intercourse the protocol-defined statistical
criteria for success)
Mainly mild or moderate AEs
Glasier et al., 2010 (63) R, SB %120 h after unprotected Pregnancy rate, %
intercourse
Ulipristal acetate (1  30 mg) 1,104 1.8
Levonorgestrel (1  1.5 mg) 1,117 2.6
Arms had similar AE frequency
Two serious AEs were judged
possibly treatment-related;
dizziness (ulipristal acetate)
and a molar pregnancy
(levonorgestrel)
Long-term contraception (ovulation)
Chabbert-Buffet R, PC, DB 84 d Anovulation rate, % (P value
et al., 2007 (44) vs. placebo)
Placebo 11 0
Ulipristal acetate (2.5 mg daily) 11 9.1 (P¼NS)
Ulipristal acetate (5 mg daily) 11 81.8 (P< .001)
Ulipristal acetate (10 mg daily) 10 80.0 (P< .001)
General and gynecologic
safety data were
satisfactory
Vol. 96, No. 5, November 2011

Mifepristone
Emergency contraception
Jin et al., 2005 (64) R, DB %120 h after unprotected Pregnancy rate, %
intercourse
Mifepristone (1  10 mg) 49 2.0
Mifepristone (1  50 mg) 47 2.1
Mifepristone (1  600 mg) 48 2.1

Bouchard. SPRMs in reproductive medicine. Fertil Steril 2011.


Fertility and Sterility

TABLE 2
Continued.

Treatment Treatment
Study Design and dose n duration/timing Key results

Esteve et al., 2007 (65) OL Mifepristone (1  10 mg) 635 %144 h after unprotected 1.1% pregnancy rate
intercourse Acceptable profile of side effects
Taneepanichskul, 2009 (66) OL Mifepristone (1  10 mg) 120 %120 h after unprotected 0 pregnancy rate
intercourse Few side effects
Wu et al., 2010 (67) R, DB %72 h after unprotected Pregnancy rate, %
intercourse
Mifepristone (1  10 mg) 499 1.8
Gestrinone (1  10 mg) 499 2.4
No difference in side effects
between arms
Long-term contraception
Lakha et al., 2007 (68) R, DB 24 wk Pregnancy rate, %
Mifepristone (5 mg daily) 73 0.6
Levonorgestrel (0.03 mg daily) 23 1.2
There were no major AEs
in either group
Pei et al., 2007 (69) R, DB 24 wk Pregnancy rate, %
Mifepristone (25 mg weekly) 39 0
Mifepristone (50 mg weekly) 37 0
83 Side effects were uncommon
and mild
Agarwal et al., 2009 (70) OL 2–12 mo Pregnancy rate, %
Mifepristone (200 mg monthly) 80 0.2
Combined oral contraceptive 83 1.0
(COC) daily
Significantly fewer side
effects in the mifepristone
vs COC group (P¼ .001)

Bouchard. SPRMs in reproductive medicine. Fertil Steril 2011.


1181
1182
TABLE 2
Continued.
Bouchard et al.

Treatment Treatment
Study Design and dose n duration/timing Key results

Asoprisnil
Long-term contraception
Chwalisz K et al., 2005 (45) DB, PC 28 d Anovulation rate, % (measured by
progesterone levels <3.5 ng/mL
during treatment)
Placebo 12 8.3
SPRMs in reproductive medicine

Asoprisnil (5 mg daily) 8 12.5


Asoprisnil (5 mg twice daily) 8 62.5
Asoprisnil (10 mg daily) 8 50.0
Asoprisnil (25 mg daily) 8 37.5
Asoprisnil (25 mg twice daily) 8 37.5
Asoprisnil (50 mg daily) 8 87.5
Asoprisnil was well tolerated

Treatment Treatment
Study Design and dose n duration/timing Key results

Uterine leiomyomata
Ulipristal acetate
Levens et al., 2008 (51) R, DB, PC 3 cycles or Change in leiomyoma Amenorrhea during cycle
90–102 d volume (%) 3 (% women)
Placebo 8 þ6 0
Ulipristal acetate (10 mg daily) 8 36 87.5
Ulipristal acetate (20 mg daily) 6 21 100.0
Ulipristal acetate was well tolerated
Nieman et al., 2011 (71) R, DB, PC 3 cycles or Change in leiomyoma Amenorrhea during
90–102 d volume (%) treatment (% women)
Placebo 12 þ7 0
Ulipristal acetate (10 mg daily) 13 17 61.5
Ulipristal acetate (20 mg daily) 13 24 92.0
Ulipristal acetate was well tolerated
Asoprisnil
Chwalisz et al., 2007 (50) R, DB, PC 12 wk Median change in uterine Amenorrhea (% women)
volume (%)
Vol. 96, No. 5, November 2011

Placebo 31 þ1 0
Asoprisnil (5 mg daily) 33 14 16
Asoprisnil (10 mg daily) 29 9 36
Asoprisnil (25 mg daily) 36 17 70
Asoprisnil was well tolerated

Bouchard. SPRMs in reproductive medicine. Fertil Steril 2011.


Fertility and Sterility

TABLE 2
Continued.

Treatment Treatment
Study Design and dose n duration/timing Key results

Wilkens et al., 2008 (72) R, DB, PC 12 wk Median change in largest Average number of
leiomyoma volume, % bleeding days
in cycle 3
Placebo 10 þ4.9 7.3
Asoprisnil (10 mg daily) 12 0.4 1.2
Asoprisnil (25 mg daily) 11 25.8 0.2
Asoprisnil was well tolerated
Telapristone acetate
Wiehle et al., 2008 (73) R, DB, PC 3 mo Mean change in leiomyoma volume, %
Placebo 10.6
Telapristone acetate (12.5 mg daily) 17.9
Telapristone acetate (25 mg daily) 40.3
Telapristone acetate (50 mg daily) 40.3
Lupron (3.75 mg monthly) 32.6
Mifepristone
Eisinger et al., 2005 (74) R, OL 1y Change in mean uterine Amenorrhea at 1 y
volume at 1 y, % (% women)
Mifepristone (5 mg daily) 52 75
Mifepristone (10 mg daily) 53 40
1 patient in the mifepristone 10 mg group showed simple
hyperplasia at 1 y
Fiscella et al., 2006 (75) R, PC 26 wk Mean change in uterine volume, % Amenorrhea at 26 wk
(% women)
Placebo 20 þ10 0
Mifepristone (5 mg daily) 22 47 41
No statistically significant differences in adverse events
between treatment groups
Carbonell Esteve et al., R 3 mo Change in leiomyoma volume, % Amenorrhea during
2008 (76) cycle 3 (% women)
Mifepristone (5 mg daily) 50 57 90.0
Mifepristone (10 mg daily) 49 45 89.8
1 patient in the mifepristone 10 mg group showed simple
hyperplasia
Bagaria et al., 2009 (77) R, DB, PC 3 mo Median change in leiomyoma Amenorrhea during cycle
volume, % 3 (% women)
Placebo 20 þ0.5 0
Mifepristone (10 mg daily) 20 30.2 84.2
63.1% of mifepristone-treated patients with endometrial
hyperplasia without atypia diagnosis

Bouchard. SPRMs in reproductive medicine. Fertil Steril 2011.


1183
SPRMS FOR THE TREATMENT OF DYSFUNCTIONAL
UTERINE BLEEDING

Amenorrhea during months

Change in health-related
Some studies have suggested that SPRMs may be able to control

Cystic glandular dilation, but no endometrial hyperplasia or atypia


3 and 6, respectively
uterine bleeding via a direct effect on endometrial blood vessels

65 and 32

quality of life, %
(45, 72, 108, 109), as amenorrhea in patients receiving SPRMs

þ40.9
þ123.4
12
34
does not appear to depend on inhibition of ovulation in all cases

(% women)
(45, 72). The mechanism of the amenorrhea observed after SPRM
treatment is still poorly understood (110). Suppression of bleeding

No premalignant changes
in women with uterine fibroids treated with SPRMs has been shown
to be associated with a moderate reduction in uterine artery blood
flow, without major changes in angiogenic factors and extracellular
Key results

Mean change in leiomyoma volume, %


matrix composition (72). This is a clear difference to the modifica-
tions observed after progestin administration (72, 110, 111).
Furthermore, intrauterine administration of the SPRM ZK230211
Change in uterine volume, %
in women scheduled for hysterectomy due to menorrhagia or

Note: AE ¼ adverse event; DB ¼ double-blind; NS ¼ not statistically significant; OL ¼ open label; PC ¼ placebo-controlled; R ¼ randomized; SB ¼ single-blind.
dysmenorrhea resulted in a reduced number of days with bleeding
Mean change in uterine

þ17.7
17.6
and spotting compared with intrauterine administration of
volume (%) at 6 mo

11

28
þ6

levonorgestrel (96). Finally, women experiencing breakthrough


bleeding during progestin treatment show a clear improvement in
bleeding pattern following SPRM administration (112, 113).

SPRMS FOR THE TREATMENT OF UTERINE FIBROIDS


Uterine leiomyomata arise from the smooth muscle cells of the
myometrium (114, 115). Despite their benign status, uterine
leiomyomata can cause debilitating symptoms, including
duration/timing
Treatment

menorrhagia, abdominal pain, and infertility (114–116). There is


6 mo

3 mo

6 mo

still much debate regarding the etiology of uterine leiomyomata,


with genetics, hormones, and growth factors all thought to play
a part (114). It is, however, likely that progesterone and the PR
have key roles in their development (117–125), and it has been
shown that PR–ligand complexes reduce apoptosis and increase
23

16
14

19
43
n

proliferation of leiomyoma cells (116). Traditionally, the favored


treatment for uterine leiomyoma has been hysterectomy or myomec-
tomy, with more modern, less invasive therapies including uterine
Mifepristone (2.5 or 5 mg daily)

artery embolization, high-intensity focused ultrasound, and uterine


artery ligation. Available pharmacologic therapies are limited but in-
Mifepristone (2.5 mg daily)

clude GnRH agonists and antagonists, oral contraceptives, and pro-


Treatment
and dose

gestins (115, 126).


Mifepristone (50 mg
every other day)

Rationale
The apparent importance of progesterone for the growth and devel-
Placebo

Placebo

opment of uterine leiomyoma suggests that SPRMs may be an effec-


tive treatment. Accordingly, treatment of cultured leiomyoma cells
with ulipristal acetate, telapristone acetate, or asoprisnil results in
Bouchard. SPRMs in reproductive medicine. Fertil Steril 2011.

inhibition of proliferation (120, 127–129) and induction of the


Partially R, PC

endoplasmic reticulum stress-induced (130), intrinsic (120, 127,


Design

129), or extrinsic (131) apoptosis pathways. In contrast, these agents


have no effect on the proliferation or apoptosis of normal myome-
R, PC

trial cells (120, 127–129, 131). Asoprisnil and ulipristal acetate


OL

have also been shown to down-regulate a number of growth factors


and their receptors in cultured leiomyoma cells (132, 133). In
Engman et al., 2009 (79)
Eisinger et al., 2009 (78)

addition, both asoprisnil and ulipristal acetate can reduce collagen


Feng et al., 2010 (80)

synthesis in cultured leiomyoma cells through up-regulation of the


extracellular matrix metalloproteinase inducer (134, 135).

Clinical Studies
Continued.

A number of clinical trials have investigated the efficacy and safety of


TABLE 2

SPRMs as a treatment for uterine leiomyoma. Trials before 2005 have


Study

been reviewed previously elsewhere (6, 7, 107), and key trials from
2005 onward are summarized in Table 2. Studies have demonstrated
that mifepristone, ulipristal acetate, asoprisnil, and telapristone acetate

1184 Bouchard et al. SPRMs in reproductive medicine Vol. 96, No. 5, November 2011
are all effective at reducing leiomyoma and uterine volume. Leio- of this drug in patients with uterine fibroids (148, 149). Clinical trials
myoma volumes are reduced by 17% to 57% and uterine volume by with this drug have restarted employing lower doses (150). The de-
9% to 53% during treatment with SPRMs, compared with general in- velopment of another SPRM, onapristone, was also halted due to
creases in volume in patients receiving placebo (see Table 2). In liver toxicity (151). For other SPRMs, only mild and transient eleva-
contrast to GnRH agonist therapy, these SPRMS are also able to sup- tion of transaminases has been reported (50, 51, 71, 76, 79). Because
press bleeding rapidly in women with uterine leiomyomata, without liver toxicity has not been reported as a feature of mifepristone,
suppressing estrogen secretion, with the majority of women experi- asoprisnil, or ulipristal acetate, it may be that the liver toxicity of
encing amenorrhea during treatment with ulipristal acetate, asoprisnil, telapristone acetate and onapristone is specific to their structure,
or mifepristone (see Table 2). Moreover, a number of trials reported an and/or the pathways by which telapristone acetate and onapristone
improvement in quality of life measures for women receiving are metabolized, rather than being a class effect. Possibly, the
ulipristal acetate, asoprisnil, or mifepristone (51, 71, 72, 75, 78, 80). ethanol-like 1-hydroxyalkyl moiety present in onapristone—and
likely to be formed during metabolic degradation of telapristone—
EFFECT OF SPRMS ON BREAST TISSUE will be further transformed into hydroxyl alkyl radical-forming
It is thought that progesterone drives mitotic activity—which peaks adducts (152).
during the luteal phase—in normal, adult breast tissue, with proges-
tins able to stimulate proliferation of breast cancer cells and enhance Ovarian Cysts
their invasiveness (136, 137). Importantly, treatment of Brca1/p53 –/– There have been a number of reports suggesting that ovarian cysts
deficient mice with mifepristone prevents mammary tumorigenesis are more common in women treated with SPRMs. However, the
(138). The potential role that SPRMs may play in preventing or cysts—which probably arise from abnormal ovulation—are gener-
treating breast cancer has been reviewed previously elsewhere ally small, asymptomatic, and resolve without treatment (44, 45,
(139) and lies outside the scope of this article. However, it is also im- 49, 50, 68, 153).
portant to understand the effect that SPRMs may have on normal
breast tissue.
Prolactin Levels
Although limited data describing the activity of SPRMs in normal
Two studies of ulipristal acetate in women with uterine fibroids have
breast tissue are available, it appears that SPRMs have an antiproli-
shown prolactin levels to be mildly elevated during treatment in
ferative effect in a number of systems. Exposure of mammary glands
some women; elevations were transient in most cases, and none
from progesterone-primed virgin mice to ZK114043 led to a reduc-
was considered a serious adverse event (51, 71). Another study of
tion in epithelial cell proliferation and differentiation of alveolar
ulipristal acetate (44) and studies of mifepristone and asoprisnil
cells (140). In primates, too, treatment with asoprisnil or mifepris-
(45, 79) reported no effect of SPRM treatment on prolactin levels.
tone results in antiproliferative effects in breast tissue (48, 141),
and the antiproliferative effects of mifepristone have been
described in the normal human breast (141). These data suggest Bone Mineral Density
that treatment with SPRMs may lower the risk for breast cancer, al- Other hormonal therapies, such as GnRH agonists, cause loss of
though further studies are necessary to investigate this potentially bone mineral density due to hypoestrogenic effects. In contrast,
beneficial effect. SPRMs maintain physiologic levels of estrogen (43–51) and thus
maintain bone mineral density (99, 103).
PROGESTERONE RECEPTOR MODULATOR-ASSOCIATED
ENDOMETRIAL CHANGES CONCLUSION
Previously, there was some concern regarding the endometrial The SPRMs are PR ligands that display molecule- and tissue-
changes induced by medium- to long-term (3 to 6 months) continuous specific interactions with coactivators and corepressors, leading to
daily dosing of mifepristone (2 to 200 mg daily) and asoprisnil mixed agonist and antagonist activity. Although each SPRM has
(142–144). These unusual changes are characterized by dilated, a different molecular signature, SPRMs all have very similar effects
weakly secretory endometrial glands with few mitotic figures, and on the reproductive system, blocking ovulation, inducing nonphy-
stromal effects ranging from compaction to nonuniform edema (45, siologic endometrial changes, suppressing bleeding, and reducing
50, 74, 78). However, a panel of expert pathologists—convened the size of uterine leiomyoma. Nonetheless, differences are also ap-
to examine these novel changes—concluded that the changes parent, with mifepristone being the only SPRM to carry significant
revealed nothing that should be considered a safety concern (145). abortifacient activity and a far higher affinity for the GR than other
It is clear that modifications to the diagnostic criteria are required SPRM molecules.
to describe these unusual changes, which have been termed proges-
terone receptor modulator-associated endometrial changes (PAECs) Expectations for the Future
(146). Short-term studies of asoprisnil, ulipristal acetate, and telapri- The SPRMs are presently approved for emergency contraception
stone acetate have confirmed the absence of endometrial hyperplasia (ulipristal acetate) and for termination of pregnancy (mifepristone,
in all subjects when pathologic review encompasses the spectrum of in association with prostaglandins). They are currently in develop-
PAECs (72, 94, 95, 147). However, long-term studies are needed, ment for a number of different gynecologic applications, including
especially if SPRMs are to be used for more than 3 months. estrogen-free contraception, uterine leiomyoma, and eventually
treatment of endometriosis. There is also the prospect that SPRMs
SAFETY OF LONG-TERM TREATMENT WITH SPRMS could be used before surgery to facilitate leiomyoma surgery and im-
Liver Toxicity prove surgical outcomes. Currently, GnRH agonists are often used in
The most concerning side effect reported in patients treated with this capacity and have been shown to improve preoperative and post-
SPRMs is the elevation of liver enzymes in patients receiving telap- operative hemoglobin levels and hematocrit, reduce the proportion
ristone acetate (50 mg), which led to suspension of the phase 3 trials of hysterectomies with difficult surgery, reduce the invasiveness of

Fertility and Sterility 1185


the surgical technique used, and reduce the duration of hospital stay strate the efficacy and safety of SPRMs both for use presurgically
(154, 155). However, this treatment takes several weeks to become and for long-term, chronic use. For long-term use, intermittent
effective and induces low estrogen levels and hot flushes. treatment regimens may reduce or avoid the need to monitor
The next step on the path to enabling SPRMs to achieve their for PAECs by ensuring regular and predictable withdrawal
full potential as a novel and much-needed therapy is to demon- bleeding (107).

REFERENCES
1. Rocha A, Soares R. Unraveling progesterone- sponsible for the differential activity of the A and binding to progesterone response elements and are
induced molecular mechanisms in physiological B isoforms of the human progesterone receptor. J dominantly inhibited by A-receptors. Mol Endocri-
and pathological conditions. Curr Clin Pharmacol Biol Chem 1997;272:32889–900. nol 1993;7:1256–65.
2009;4:148–53. 18. Giangrande PH, McDonnell DP. The A and B iso- 32. Leo JC, Lin VC. The activities of progesterone re-
2. McEwan IJ. Nuclear receptors: one big family. forms of the human progesterone receptor: two ceptor isoform A and B are differentially modulated
Methods Mol Biol 2009;505:3–18. functionally different transcription factors encoded by their ligands in a gene-selective manner. Int J
3. Philibert D. RU 38486: an original multifaceted by a single gene. Recent Prog Horm Res 1999;54: Cancer 2008;122:230–43.
antihormone in vivo. In: Agarwal M, ed. Adrenal 291–314. 33. Amazit L, Roseau A, Khan JA, Chauchereau A,
steroid antagonism. Berlin: Walter de Gruyter; 19. Giangrande PH, Kimbrel EA, Edwards DP, Tyagi RK, Loosfelt H, et al. Ligand-dependent deg-
1984;77–101. McDonnell DP. The opposing transcriptional activ- radation of SRC-1 is pivotal for progesterone recep-
4. Allan GF, Sui Z. Non-steroidal progesterone recep- ities of the two isoforms of the human progesterone tor transcriptional activity. Mol Endocrinol
tor specific ligands. Mini Rev Med Chem 2005;5: receptor are due to differential cofactor binding. 2011;25:394–408.
701–8. Mol Cell Biol 2000;20:3102–15. 34. Madauss KP, Stewart EL, Williams SP. The evolu-
5. Chabbert-Buffet N, Ouzounian S, Kairis AP, 20. Conneely OM, Mulac-Jericevic B, Lydon JP, De tion of progesterone receptor ligands. Med Res
Bouchard P. Contraceptive applications of proges- Mayo FJ. Reproductive functions of the progester- Rev 2007;27:374–400.
terone receptor modulators. Eur J Contracept Re- one receptor isoforms: lessons from knock-out 35. Johanssen S, Allolio B. Mifepristone (RU 486) in
prod Health Care 2008;13:222–30. mice. Mol Cell Endocrinol 2001;179:97–103. Cushing’s syndrome. Eur J Endocrinol 2007;157:
6. Chabbert-Buffet N, Meduri G, Bouchard P, 21. Mulac-Jericevic B, Lydon JP, DeMayo FJ, 561–9.
Spitz IM. Selective progesterone receptor modula- Conneely OM. Defective mammary gland 36. Schubert G, Elger W, Kaufmann G, Schneider B,
tors and progesterone antagonists: mechanisms of morphogenesis in mice lacking the progesterone Reddersen G, Chwalisz K. Discovery, chemistry,
action and clinical applications. Hum Reprod Up- receptor B isoform. Proc Natl Acad Sci USA and reproductive pharmacology of asoprisnil and re-
date 2005;11:293–307. 2003;100:9744–9. lated 11b-benzaldoxime substituted selective pro-
7. Chwalisz K, Perez MC, Demanno D, Winkel C, 22. Arnett-Mansfield RL, DeFazio A, Mote PA, gesterone receptor modulators (SPRMs). Semin
Schubert G, Elger W. Selective progesterone recep- Clarke CL. Subnuclear distribution of progester- Reprod Med 2005;23:58–73.
tor modulator development and use in the treatment one receptors A and B in normal and malignant 37. Attardi BJ, Burgenson J, Hild SA, Reel JR. In vitro
of leiomyomata and endometriosis. Endocr Rev endometrium. J Clin Endocrinol Metab antiprogestational/antiglucocorticoid activity and
2005;26:423–38. 2004;89:1429–42. progestin and glucocorticoid receptor binding of
8. Conneely OM, Mulac-Jericevic B, DeMayo F, 23. Mote PA, Bartow S, Tran N, Clarke CL. Loss of co- the putative metabolites and synthetic derivatives
Lydon JP, O’Malley BW. Reproductive functions ordinate expression ofprogesterone receptors A and of CDB-2914, CDB-4124, and mifepristone. J Ste-
of progesterone receptors. Recent Prog Horm Res B is an early event in breast carcinogenesis. Breast roid Biochem Mol Biol 2004;88:277–88.
2002;57:339–55. Cancer Res Treat 2002;72:163–72. 38. Winneker RC, Fensome A, Zhang P, Yudt MR,
9. Mesiano S, Welsh TN. Steroid hormone control of 24. Chabbert-Buffet N, Skinner DC, Caraty A, McComas CC, Unwalla RJ. A new generation of
myometrial contractility and parturition. Semin Bouchard P. Neuroendocrine effects of progester- progesterone receptor modulators. Steroids
Cell Dev Biol 2007;18:321–31. one. Steroids 2000;65:613–20. 2008;73:689–701.
10. Conneely OM, Jericevic BM, Lydon JP. Progester- 25. McPhail MK. The assay of progestin. J Physiol 39. Zhi L. Discovery of structurally diverse nonsteroidal
one receptors in mammary gland development and 1934;83:145–56. SPRMs based on a screening hit, 1,2-dihydro-
tumorigenesis. J Mammary Gland Biol Neoplasia 26. Elger W, Bartley J, Schneider B, Kaufmann G, 2,2,4-trimethyl-6-phenylquinolinone. Curr Top
2003;8:205–14. Schubert G, Chwalisz K. Endocrine pharmacologi- Med Chem 2008;8:766–80.
11. Li Q, Kannan A, DeMayo FJ, Lydon JP, Cooke PS, cal characterization of progesterone antagonists 40. Rewinkel J, Enthoven M, Golstein I, van der RM,
Yamagishi H, et al. The antiproliferative action of and progesterone receptor modulators with respect Scholten A, van Tilborg M, et al. 11-(pyridinyl-
progesterone in uterine epithelium is mediated by to PR-agonistic and antagonistic activity. Steroids phenyl)steroids—a new class of mixed-profile pro-
Hand2. Science 2011;331:912–6. 2000;65:713–23. gesterone agonists/antagonists. Bioorg Med Chem
12. Scarpin KM, Graham JD, Mote PA, Clarke CL. Pro- 27. Afh€uppe W, Sommer A, M€uller J, Schwede W, 2008;16:2753–63.
gesterone action in human tissues: regulation by Fuhrmann U, M€oller C. Global gene expression pro- 41. Schaff EA. Mifepristone: ten years later. Contracep-
progesterone receptor (PR) isoform expression, nu- filing of progesterone receptor modulators in T47D tion 2010;81:1–7.
clear positioning and coregulator expression. Nucl cells provides a new classification system. J Steroid 42. Im A, Appleman LJ. Mifepristone: pharmacology
Recept Signal 2009;7:e009. Biochem Mol Biol 2009;113:105–15. and clinical impact in reproductive medicine, endo-
13. Leonhardt SA, Edwards DP. Mechanism of action of 28. Smith CL, O’Malley BW. Coregulator function: crinology and oncology. Expert Opin Pharmacother
progesterone antagonists. Exp Biol Med (May- a key to understanding tissue specificity of selec- 2010;11:481–8.
wood) 2002;227:969–80. tive receptor modulators. Endocr Rev 2004;25: 43. Baird DT, Thong KJ, Hall C, Cameron ST. Failure of
14. Boonyaratanakornkit V, Edwards DP. Receptor 45–71. oestrogen induced luteinizing hormone surge in
mechanisms mediating non-genomic actions of 29. Madauss KP, Grygielko ET, Deng SJ, Sulpizio AC, women treated with mifepristone (RU 486) every
sex steroids. Semin Reprod Med 2007;25: Stanley TB, Wu C, et al. A structural and in vitro day for 30 days. Hum Reprod 1995;10:2270–6.
139–53. characterization of asoprisnil: a selective progester- 44. Chabbert-Buffet N, Pintiaux-Kairis A, Bouchard P.
15. Blaustein JD. Minireview: neuronal steroid hor- one receptor modulator. Mol Endocrinol 2007;21: Effects of the progesterone receptor modulator
mone receptors: they’re not just for hormones any- 1066–81. VA2914 in a continuous low dose on the
more. Endocrinology 2004;145:1075–81. 30. Wardell SE, Edwards DP. Mechanisms controlling hypothalamic-pituitary-ovarian axis and endome-
16. Lydon JP, DeMayo FJ, Funk CR, Mani SK, agonist and antagonist potential of selective proges- trium in normal women: a prospective, randomized,
Hughes AR, Montgomery CA Jr, et al. Mice lacking terone receptor modulators (SPRMs). Semin Reprod placebo-controlled trial. J Clin Endocrinol Metab
progesterone receptor exhibit pleiotropic reproduc- Med 2005;23:9–21. 2007;92:3582–9.
tive abnormalities. Genes Dev 1995;9:2266–78. 31. Tung L, Mohamed MK, Hoeffler JP, Takimoto GS, 45. Chwalisz K, Elger W, Stickler T,
17. Giangrande PH, Pollio G, McDonnell DP. Mapping Horwitz KB. Antagonist-occupied human proges- Mattia-Goldberg C, Larsen L. The effects of
and characterization of the functional domains re- terone B-receptors activate transcription without 1-month administration of asoprisnil (J867), a selec-

1186 Bouchard et al. SPRMs in reproductive medicine Vol. 96, No. 5, November 2011
tive progesterone receptor modulator, in healthy 60. Meng CX, Andersson KL, Bentin-Ley U, 76. Carbonell Esteve JL, Acosta R, Heredia B, Perez Y,
premenopausal women. Hum Reprod 2005;20: Gemzell-Danielsson K, Lalitkumar PG. Effect of Castaneda MC, Hernandez AV. Mifepristone for the
1090–9. levonorgestrel and mifepristone on endometrial re- treatment of uterine leiomyomas: a randomized con-
46. Croxatto HB, Salvatierra AM, Croxatto HD, ceptivity markers in a three-dimensional human trolled trial. Obstet Gynecol 2008;112:1029–36.
Fuentealba B. Effects of continuous treatment with endometrial cell culture model. Fertil Steril 2009; 77. Bagaria M, Suneja A, Vaid NB, Guleria K,
low dose mifepristone throughout one menstrual cy- 91:256–64. Mishra K. Low-dose mifepristone in treatment of
cle. Hum Reprod 1993;8:201–7. 61. Creinin MD, Schlaff W, Archer DF, Wan L, uterine leiomyoma: a randomised double-blind pla-
47. Liu JH, Garzo G, Morris S, Stuenkel C, Ulmann A, Frezieres R, Thomas M, et al. Progesterone receptor cebo-controlled clinical trial. Aust NZ J Obstet Gy-
Yen SS. Disruption of follicular maturation and de- modulator for emergency contraception: a random- naecol 2009;49:77–83.
lay of ovulation after administration of the antipro- ized controlled trial. Obstet Gynecol 2006;108: 78. Eisinger SH, Fiscella J, Bonfiglio T, Meldrum S,
gesterone RU486. J Clin Endocrinol Metab 1089–97. Fiscella K. Open-label study of ultra low-dose mif-
1987;65:1135–40. 62. Fine P, Mathe H, Ginde S, Cullins V, Morfesis J, epristone for the treatment of uterine leiomyomata.
48. Chwalisz K, Garg R, Brenner R, Slayden O, Gainer E. Ulipristal acetate taken 48–120 hours after Eur J Obstet Gynecol Reprod Biol 2009;146:215–8.
Winkel C, Elger W. Role of nonhuman primate intercourse for emergency contraception. Obstet 79. Engman M, Granberg S, Williams AR, Meng CX,
models in the discovery and clinical development of Gynecol 2010;115:257–63. Lalitkumar PG, Gemzell-Danielsson K. Mifepris-
selective progesterone receptor modulators (SPRMs). 63. Glasier AF, Cameron ST, Fine PM, Logan SJ, tone for treatment of uterine leiomyoma: a prospec-
Reprod Biol Endocrinol 2006;4(Suppl 1):S8. Casale W, Van Horn J, et al. Ulipristal acetate versus tive randomized placebo controlled trial. Hum
49. Stratton P, Levens ED, Hartog B, Piquion J, Wei Q, levonorgestrel for emergency contraception: a rand- Reprod 2009;24:1870–9.
Merino M, et al. Endometrial effects of a single omised non-inferiority trial and meta-analysis. Lan- 80. Feng C, Meldrum S, Fiscella K. Improved quality of
early luteal dose of the selective progesterone recep- cet 2010;375:555–62. life is partly explained by fewer symptoms after
tor modulator CDB-2914. Fertil Steril 2009;93: 64. Jin J, Weisberg E, Fraser IS. Comparison of three treatment of fibroids with mifepristone. Int J Gynae-
2035–41. single doses of mifepristone as emergency contra- col Obstet 2010;109:121–4.
50. Chwalisz K, Larsen L, Mattia-Goldberg C, ception: a randomised controlled trial. Aust NZ 81. Cheng L, Gulmezoglu AM, Piaggio G, Ezcurra E,
Edmonds A, Elger W, Winkel CA. A randomized, J Obstet Gynaecol 2005;45:489–94. Van Look PF. Interventions for emergency contra-
controlled trial of asoprisnil, a novel selective pro- 65. Esteve JL, Garcıa R, Breto A, Llorente M. Emer- ception. Cochrane Database Syst Rev 2008;2:
gesterone receptor modulator, in women with uter- gency contraception in Cuba with 10 mg of mife- CD001324.
ine leiomyomata. Fertil Steril 2007;87:1399–412. pristone. Eur J Contracept Reprod Health Care 82. Fiala C, Gemzel-Danielsson K. Review of medical
51. Levens ED, Potlog-Nahari C, Armstrong AY, 2007;12:162–7. abortion using mifepristone in combination with
Wesley R, Premkumar A, Blithe DL, et al. 66. Taneepanichskul S. Emergency contraception with a prostaglandin analogue. Contraception 2006;74:
CDB-2914 for uterine leiomyomata treatment: a ran- mifepristone 10 mg in Thai women. J Med Assoc 66–86.
domized controlled trial. Obstet Gynecol 2008;111: Thai 2009;92:999–1002. 83. Marions L. Mifepristone dose in the regimen with
1129–36. 67. Wu S, Dong J, Cong J, Wang C, VonHertzen H, misoprostol for medical abortion. Contraception
52. van der Stege JG, Pahl-van Beest EH, Godfrey EM. Gestrinone compared with mifepris- 2006;74:21–5.
Beerthuizen RJ, van Lunsen RH, Scholten PC, tone for emergency contraception: a randomized 84. Schreiber C, Creinin M. Mifepristone in abortion
Bogchelman DH. Effects of a preovulatory single controlled trial. Obstet Gynecol 2010;115:740–4. care. Semin Reprod Med 2005;23:82–91.
low dose of mifepristone on ovarian function. Eur 68. Lakha F, Ho PC, Van der Spuy ZM, Dada K, 85. Bedaiwy MA, Abdel-Aleem MA, Miketa A,
J Contracept Reprod Health Care 2006;11:104–8. Elton R, Glasier AF, et al. A novel estrogen-free Falcone T. Endometriosis: a critical appraisal of
53. Leminen R, Raivio T, Ranta S, Oehler J, von oral contraceptive pill for women: multicentre, the advances and the controversies of a challenging
Hertzen H, J€anne OA, et al. Late follicular phase ad- double-blind, randomized controlled trial of mife- health problem. Minerva Ginecol 2009;61:285–98.
ministration of mifepristone suppresses circulating pristone and progestogen-only pill (levonorgestrel). 86. Signorile PG, Baldi A. Endometriosis: new concepts
leptin and FSH—mechanism(s) of action in emer- Hum Reprod 2007;22:2428–36. in the pathogenesis. Int J Biochem Cell Biol
gency contraception? Eur J Endocrinol 2005;152: 69. Pei K, Xiao B, Jing X, Lu S, Wei L, Zhao H. Weekly 2010;42:778–80.
411–8. contraception with mifepristone. Contraception 87. Eskenazi B, Warner ML. Epidemiology of endome-
54. Sengupta J, Dhawan L, Lalitkumar PG, Ghosh D. A 2007;75:40–4. triosis. Obstet Gynecol Clin North Am 1997;24:
multiparametric study of the action of mifepristone 70. Agarwal M, Das V, Agarwal A, Pandey A, 235–58.
used in emergency contraception using the Rhesus Srivastava D. Evaluation of mifepristone as a once 88. Crosignani P, Olive D, Bergqvist A, Luciano A. Ad-
monkey as a primate model. Contraception a month contraceptive pill. Am J Obstet Gynecol vances in the management of endometriosis: an up-
2003;68:453–69. 2009;200:e27–9. date for clinicians. Hum Reprod Update 2006;12:
55. Stratton P, Hartog B, Hajizadeh N, Piquion J, 71. Nieman LK, Blocker W, Nansel T, Mahoney S, 179–89.
Sutherland D, Merino M, et al. A single mid- Reynolds J, Blithe D, et al. Efficacy and tolerability 89. Brenner RM, Slayden OD, Nath A, Tsong YY, Si-
follicular dose of CDB-2914, a new antiprogestin, of CDB-2914 treatment for symptomatic uterine truk-Ware R. Intrauterine administration of CDB-
inhibits folliculogenesis and endometrial differenti- fibroids: a randomized, double-blind, placebo- 2914 (Ulipristal) suppresses the endometrium of
ation in normally cycling women. Hum Reprod controlled, phase IIb study. Fertil Steril rhesus macaques. Contraception 2010;81:336–42.
2000;15:1092–9. 2011;95:767–72.e1–2. 90. Gopalkrishnan K, Katkam RR, Sachdeva G,
56. Reel JR, Hild-Petito S, Blye RP. Antiovulatory and 72. Wilkens J, Chwalisz K, Han C, Walker J, Kholkute SD, Padwal V, Puri CP. Effects of an anti-
postcoital antifertility activity of the antiprogestin Cameron IT, Ingamells S, et al. Effects of the selec- progestin onapristone on the endometrium of bonnet
CDB-2914 when administered as single, multiple, tive progesterone receptor modulator asoprisnil on monkeys: morphometric and ultrastructural studies.
or continuous doses to rats. Contraception uterine artery blood flow, ovarian activity, and clin- Biol Reprod 2003;68:1959–67.
1998;58:129–36. ical symptoms in patients with uterine leiomyomata 91. Zhang Z, Lundeen SG, Slayden O, Zhu Y, Cohen J,
57. Banaszak S, Brudney A, Donnelly K, Chai D, scheduled for hysterectomy. J Clin Endocrinol Berrodin TJ, et al. In vitro and in vivo characteriza-
Chwalisz K, Fazleabas AT. Modulation of the action Metab 2008;93:4664–71. tion of a novel nonsteroidal, species-specific proges-
of chorionic gonadotropin in the baboon (Papio anu- 73. Wiehle R, Goldberg J, Brodniewicz T, Jarus- terone receptor modulator, PRA-910. Ernst
bis) uterus by a progesterone receptor antagonist Dziedzic K, Jabiry-Zieniewicz Z. Effects of a new Schering Found Symp Proc 2007:171–97.
(ZK 137.316). Biol Reprod 2000;63:820–5. progesterone receptor modulator, CDB-4124, on fi- 92. Wu Y, Guo SW. Inhibition of proliferation of endo-
58. Lalitkumar PG, Lalitkumar S, Meng CX, Stavreus- broid size and uterine bleeding. US Obstetr Gynae- metrial stromal cells by trichostatin A, RU486,
Evers A, Hambiliki F, Bentin-Ley U, et al. Mifepris- col 2008;3:17–20. CDB-2914, N-acetylcysteine, and ICI 182780. Gy-
tone, but not levonorgestrel, inhibits human 74. Eisinger SH, Bonfiglio T, Fiscella K, Meldrum S, necol Obstet Invest 2006;62:193–205.
blastocyst attachment to an in vitro endometrial Guzick DS. Twelve-month safety and efficacy of 93. Moe BG, Vereide AB, Orbo A, Sager G. High con-
three-dimensional cell culture model. Hum Reprod low-dose mifepristone for uterine myomas. J Minim centrations of progesterone and mifepristone mutu-
2007;22:3031–7. Invasive Gynecol 2005;12:227–33. ally reinforce cell cycle retardation and induction of
59. Petersen A, Bentin-Ley U, Ravn V, Qvortrup K, 75. Fiscella K, Eisinger SH, Meldrum S, Feng C, apoptosis. Anticancer Res 2009;29:1053–8.
Sorensen S, Islin H, et al. The antiprogesterone Fisher SG, Guzick DS. Effect of mifepristone for 94. Ioffe OB, Zaino RJ, Mutter GL. Endometrial
Org 31710 inhibits human blastocyst-endometrial symptomatic leiomyomata on quality of life and changes from short-term therapy with CDB-4124,
interactions in vitro. Fertil Steril 2005;83(Suppl 1): uterine size: a randomized controlled trial. Obstet a selective progesterone receptor modulator. Mod
1255–63. Gynecol 2006;108:1381–7. Pathol 2009;22:450–9.

Fertility and Sterility 1187


95. Wilkens J, Williams AR, Chwalisz K, Han C, 111. Stephanie R, Labied S, Blacher S, Frankenne F, ulator CDB4124 inhibits proliferation and induces
Cameron IT, Critchley HO. Effect of asoprisnil on Munaut C, Fridman V, et al. Endometrial vessel mat- apoptosis in uterine leiomyoma cells. Fertil Steril
uterine proliferation markers and endometrial ex- uration in women exposed to levonorgestrel-releasing 2010;93:2668–73.
pression of the tumour suppressor gene, PTEN. intrauterine system for a short or prolonged period of- 128. Maruo T, Ohara N, Matsuo H, Xu Q, Chen W, Sitruk-
Hum Reprod 2009;24:1036–44. time. Hum Reprod 2007;22:3084–91. Ware R, et al. Effects of levonorgestrel-releasing IUS
96. Heikinheimo O, Vani S, Carpen O, Tapper A, 112. Gemzell-Danielsson K, van Heusden AM, and progesterone receptor modulator PRM CDB-
Harkki P, Rutanen EM, et al. Intrauterine release Killick SR, Croxatto HB, Bouchard P, Cameron S, 2914 on uterine leiomyomas. Contraception
of progesterone antagonist ZK230211 is feasible et al. Improving cycle control in progestogen-only 2007;75(Suppl):S99–103.
and results in novel endometrial effects: a pilot contraceptive pill users by intermittent treatment 129. Xu Q, Takekida S, Ohara N, Chen W, Sitruk-Ware R,
study. Hum Reprod 2007;22:2515–22. with a new anti-progestogen. Hum Reprod 2002;17: Johansson ED, et al. Progesterone receptor modula-
97. Williams AR, Critchley HO, Osei J, Ingamells S, 2588–93. tor CDB-2914 down-regulates proliferative cell
Cameron IT, Han C, et al. The effects of the selective 113. Massai MR, Pavez M, Fuentealba B, Croxatto HB, nuclear antigen and Bcl-2 protein expression and
progesterone receptor modulator asoprisnil on the d’Arcangues C. Effect of intermittent treatment up-regulates caspase-3 and poly(adenosine 5’-di-
morphology of uterine tissues after 3 months treat- with mifepristone on bleeding patterns in Norplant phosphate-ribose) polymerase expression in
ment in patients with symptomatic uterine leiomyo- implant users. Contraception 2004;70:47–54. cultured human uterine leiomyoma cells. J Clin En-
mata. Hum Reprod 2007;22:1696–704. 114. Parker WH. Etiology, symptomatology, and diagno- docrinol Metab 2005;90:953–61.
98. Baird DT, Brown A, Critchley HO, Williams AR, sis of uterine myomas. Fertil Steril 2007;87:725–36. 130. Xu Q, Ohara N, Liu J, Nakabayashi K, Demanno D,
Lin S, Cheng L. Effect of long-term treatment 115. Miller CE. Unmet therapeutic needs for uterine my- Chwalisz K, et al. Selective progesterone receptor
with low-dose mifepristone on the endometrium. omas. J Minim Invasive Gynecol 2009;16:11–21. modulator asoprisnil induces endoplasmic reticulum
Hum Reprod 2003;18:61–8. 116. Catherino WH, Parrott E, Segars J. Proceedings stress in cultured human uterine leiomyoma cells.
99. Grow DR, Williams RF, Hsiu JG, Hodgen GD. Anti- from the National Institute of Child Health and Hu- Am J Physiol Endocrinol Metab 2007;293:
progestin and/or gonadotropin-releasing hormone man Development conference on the Uterine Fi- E1002–11.
agonist for endometriosis treatment and bone main- broid Research Update Workshop. Fertil Steril 131. Sasaki H, Ohara N, Xu Q, Wang J, DeManno DA,
tenance: a 1-year primate study. J Clin Endocrinol 2011;95:9–12. Chwalisz K, et al. A novel selective progesterone re-
Metab 1996;81:1933–9. 117. Nisolle M, Gillerot S, Casanas-Roux F, Squifflet J, ceptor modulator asoprisnil activates tumor necrosis
100. Stoeckemann K, Hegele-Hartung C, Chwalisz K. Berliere M, Donnez J. Immunohistochemical study factor-related apoptosis-inducing ligand (TRAIL)-
Effects of the progesterone antagonists onapristone of the proliferation index, oestrogen receptors and mediated signaling pathway in cultured human uter-
(ZK 98 299) and ZK 136 799 on surgically induced progesterone receptors A and B in leiomyomata ine leiomyoma cells in the absence of comparable
endometriosis in intact rats. Hum Reprod 1995;10: and normal myometrium during the menstrual cycle effects on myometrial cells. J Clin Endocrinol
3264–71. and under gonadotrophin-releasing hormone agonist Metab 2007;92:616–23.
101. Elger W, Ivell R, Nandy A, Rasch A, Triller A, therapy. Hum Reprod 1999;14:2844–50. 132. Wang J, Ohara N, Wang Z, Chen W, Morikawa A,
Chwalisz K. Modulation of uterine prostaglandin 118. Brandon DD, Bethea CL, Strawn EY, Novy MJ, Sasaki H, et al. A novel selective progesterone re-
secretion by the selective progesterone receptor Burry KA, Harrington MS, et al. Progesterone re- ceptor modulator asoprisnil (J867) down-regulates
modulator (SPRM) asoprisnil, progestins, and anti- ceptor messenger ribonucleic acid and protein are the expression of EGF, IGF-I, TGFb3 and their re-
progestins in cycling and ovariectomized guinea overexpressed in human uterine leiomyomas. Am J ceptors in cultured uterine leiomyoma cells. Hum
pigs. Fertil Steril 2004;82(Suppl):S316. Obstet Gynecol 1993;169:78–85. Reprod 2006;21:1869–77.
102. Gemzell-Danielsson K, Hamberg M. The effect of 119. Englund K, Blanck A, Gustavsson I, Lundkvist U, 133. Xu Q, Ohara N, Chen W, Liu J, Sasaki H,
antiprogestin (RU 486) and prostaglandin biosyn- Sj€oblom P, Norgren A, et al. Sex steroid receptors Morikawa A, et al. Progesterone receptor modulator
thesis inhibitor (naproxen) on uterine fluid prosta- in human myometrium and fibroids: changes during CDB-2914 down-regulates vascular endothelial
glandin F2 alpha concentrations. Hum Reprod the menstrual cycle and gonadotropin-releasing hor- growth factor, adrenomedullin and their receptors
1994;9:1626–30. mone treatment. J Clin Endocrinol Metab 1998;83: and modulates progesterone receptor content in cul-
103. Kettel LM, Murphy AA, Morales AJ, Ulmann A, 4092–6. tured human uterine leiomyoma cells. Hum Reprod
Baulieu EE, Yen SS. Treatment of endometriosis 120. Chen W, Ohara N, Wang J, Xu Q, Liu J, 2006;21:2408–16.
with the antiprogesterone mifepristone (RU486). Morikawa A, et al. A novel selective progesterone 134. Xu Q, Ohara N, Liu J, Amano M, Sitruk-Ware R,
Fertil Steril 1996;65:23–8. receptor modulator asoprisnil (J867) inhibits prolif- Yoshida S, et al. Progesterone receptor modulator
104. Kettel LM, Murphy AA, Morales AJ, Yen SS. Pre- eration and induces apoptosis in cultured human CDB-2914 induces extracellular matrix metallopro-
liminary report on the treatment of endometriosis uterine leiomyoma cells in the absence of compara- teinase inducer in cultured human uterine leio-
with low-dose mifepristone (RU 486). Am J Obstet ble effects on myometrial cells. J Clin Endocrinol myoma cells. Mol Hum Reprod 2008;14:181–91.
Gynecol 1998;178:1151–6. Metab 2006;91:1296–304. 135. Morikawa A, Ohara N, Xu Q, Nakabayashi K,
105. Mei L, Bao J, Tang L, Zhang C, Wang H, Sun L, 121. Shimomura Y, Matsuo H, Samoto T, Maruo T. Up- DeManno DA, Chwalisz K, et al. Selective progester-
et al. A novel mifepristone-loaded implant for regulation by progesterone of proliferating cell one receptor modulator asoprisnil down-regulates col-
long-term treatment of endometriosis: in vitro and nuclear antigen and epidermal growth factor expres- lagen synthesis in cultured human uterine leiomyoma
in vivo studies. Eur J Pharm Sci 2010;39:421–7. sion in human uterine leiomyoma. J Clin Endocrinol cells through up-regulating extracellular matrix met-
106. Chwalisz K, Mattia-Goldberg C, Elger W, Metab 1998;83:2192–8. alloproteinase inducer. Hum Reprod 2008;23:944–51.
Edmonds A. Treatment of endometriosis with the 122. Matsuo H, Kurachi O, Shimomura Y, Samoto T, 136. Gellersen B, Fernandes MS, Brosens JJ. Non-geno-
novel selective progesterone receptor modulator Maruo T. Molecular bases for the actions of ovarian mic progesterone actions in female reproduction.
(SPRM) asoprisnil. Fertil Steril 2004;82:S83–4. sex steroids in the regulation of proliferation and ap- Hum Reprod Update 2009;15:119–38.
107. Spitz IM. Clinical utility of progesterone receptor optosis of human uterine leiomyoma. Oncology 137. Ismail PM, Amato P, Soyal SM, DeMayo FJ,
modulators and their effect on the endometrium. 1999;57(Suppl 2):49–58. Conneely OM, O’Malley BW, et al. Progesterone in-
Curr Opin Obstet Gynecol 2009;21:318–24. 123. Maruo T, Matsuo H, Samoto T, Shimomura Y, volvement in breast development and tumorigene-
108. Chwalisz K, Brenner RM, Fuhrmann UU, Kurachi O, Gao Z, et al. Effects of progesterone sis—as revealed by progesterone receptor
Hess-Stumpp H, Elger W. Antiproliferative effects on uterine leiomyoma growth and apoptosis. ‘‘knockout’’ and ‘‘knockin’’ mouse models. Steroids
of progesterone antagonists and progesterone recep- Steroids 2000;65:585–92. 2003;68:779–87.
tor modulators on the endometrium. Steroids 124. Matsuo H, Maruo T, Samoto T. Increased expression 138. Poole AJ, Li Y, Kim Y, Lin SC, Lee WH, Lee EY.
2000;65:741–51. of Bcl-2 protein in human uterine leiomyoma and its Prevention of Brca1-mediated mammary tumori-
109. Nayak NR, Slayden OD, Mah K, Chwalisz K, up-regulation by progesterone. J Clin Endocrinol genesis in mice by a progesterone antagonist. Sci-
Brenner RM. Antiprogestin-releasing intrauterine Metab 1997;82:293–9. ence 2006;314:1467–70.
devices: a novel approach to endometrial contracep- 125. Ishikawa H, Ishi K, Serna VA, Kakazu R, Bulun SE, 139. Benagiano G, Bastianelli C, Farris M. Selective pro-
tion. Contraception 2007;75:S104–11. Kurita T. Progesterone is essential for maintenance gesterone receptor modulators 3: use in oncology,
110. Ravet S, Munaut C, Blacher S, Brichant G, and growth of uterine leiomyoma. Endocrinology endocrinology and psychiatry. Expert Opin Phar-
Labied S, Beliard A, et al. Persistence of an intact 2010;151:2433–42. macother 2008;9:2487–96.
endometrial matrix and vessels structure in women 126. Lumsden MA. Modern management of fibroids. Ob- 140. Li M, Spitzer E, Zschiesche W, Binas B, Parczyk K,
exposed to VA-2914, a selective progesterone re- stet Gynecol Reprod Med 2010;20:82–6. Grosse R. Antiprogestins inhibit growth and stimu-
ceptor modulator. J Clin Endocrinol Metab 127. Luo X, Yin P, Coon VJ, Cheng YH, Wiehle RD, late differentiation in the normal mammary gland.
2008;93:4525–31. Bulun SE. The selective progesterone receptor mod- J Cell Physiol 1995;164:1–8.

1188 Bouchard et al. SPRMs in reproductive medicine Vol. 96, No. 5, November 2011
141. Engman M, Skoog L, S€oderqvist G, Gemzell- associated endometrial changes (PAEC): a pilot his- 151. Klijn JG, Setyono-Han B, Foekens JA. Progesterone
Danielsson K. The effect of mifepristone on breast tological, dose-escalation study of ulipristal acetate. antagonists and progesterone receptor modulators in
cell proliferation in premenopausal women evalu- Fact View Vision Obgyn 2010:2 [special issue, the treatment of breast cancer. Steroids 2000;65:
ated through fine needle aspiration cytology. Hum May]. 825–30.
Reprod 2008;23:2072–9. 148. Repros Therapeutics. Repros Therapeutics Inc. pro- 152. Repros Therapeutics. Repros requests lift of clinical
142. Safety of treatment of uterine fibroids with asopris- vides clarification on increased liver enzymes at hold on Proellex. April 05, 2010. Available at: http://
nil. http://clinicaltrials gov/ct2/show/NCT00156208 highest dose in Proellex clinical program. July 23, www.businesswire.com/news/home/201004050050
2010. 2009. News Blaze.com. Available at: http://newsbla 89/en/Repros-Requests-Lift-Clinical-Hold-Proellex.
143. Abbott CM-G. Study of asoprisnil in the treatment ze.com/story/2009072303060800003.bw/topstory. Accessed August 20, 2011.
of uterine fibroid. Available at: http://clinicaltrials. html. Accessed August 20, 2011. 153. Cameron ST, Thong KJ, Baird DT. Effect of daily
gov/ct2/show/NCT00156156. Last accessed Sep- 149. BioMedReports.com/ Repros Therapeutics Inc. low dose mifepristone on the ovarian cycle and on
tember 14, 2011. suspends dosing of Proellex and provides update dynamics of follicle growth. Clin Endocrinol (Oxf)
144. Eisinger SH, Meldrum S, Fiscella K, le Roux HD, on financial status. BioMedRports, August 3, 2009. 1995;43:407–14.
Guzick DS. Low-dose mifepristone for uterine leio- Available at: http://biomedreports.com/200908033 154. Lethaby A, Vollenhoven B, Sowter M. Pre-operative
myomata. Obstet Gynecol 2003;101:243–50. 992/repros-therapeutics-inc-suspends-dosing-of- GnRH analogue therapy before hysterectomy or my-
145. Horne FM, Blithe DL. Progesterone receptor modu- proellexr-and-provides-update-on-financial-status.html. omectomy for uterine fibroids. Cochrane Database
lators and the endometrium: changes and conse- Accessed August 20, 2011. Syst Rev 2001;2:CD000547.
quences. Hum Reprod Update 2007;13:567–80. 150. Repros Therapeutics. Determination of the lowest, 155. Lethaby A, Vollenhoven B, Sowter M. Efficacy of
146. Mutter GL, Bergeron C, Deligdisch L, Ferenczy A, safe and effective dose of the anti-progestin, Proel- pre-operative gonadotrophin hormone releasing an-
Glant M, Merino M, et al. The spectrum of endome- lex, in healthy women. Last updated June 28, alogues for women with uterine fibroids undergoing
trial pathology induced by progesterone receptor 2011. Available at: http://clinicaltrials.gov/ct2/ hysterectomy or myomectomy: a systematic review.
modulators. Mod Pathol 2008;21:591–8. show/NCT01187043. Accessed August 20, 2011. BJOG 2002;109:1097–108.
147. Williams A, Bergeron C, Chabbert-Buffet N,
Ferenczy A. Progesterone receptor modulator-

Fertility and Sterility 1189

Das könnte Ihnen auch gefallen