1,571
Views
10
CrossRef citations to date
0
Altmetric
Editorial

Gynaecological uses of a new class of steroids: the selective progesterone receptor modulators

, &
Pages 67-73 | Received 01 Jun 2008, Accepted 01 Jul 2008, Published online: 07 Jul 2009

Abstract

Selective progesterone receptor modulators (SPRM) represent a new class of synthetic steroids, which can interact with the progesterone receptor (PR) and can exert agonist, antagonist or mixed effects on various progesterone target tissues in vivo. This review evaluates the actual and potential usefulness of SPRMs in gynaecology.

Introduction

The progesterone receptor (PR) belongs to the nuclear receptor super family and controls specific genes involved in female reproduction. The primary action of progesterone is to initiate and maintain pregnancy. Progesterone, the natural ligand of the PR, represents the pure agonist, onapristone, mifepristone show a pure antagonist activity and asoprisnil exhibits partial agonist/antagonist effects Citation[1].The synthesis of mifepristone, the first PR antagonist (PA) was a starting point of drug discovery and the research programme in the area of PAs Citation[2-4].

Progesterone is involved in the control of ovulation, facilitating the luteinising hormone (LH) surge.

It transforms the endometrium from a proliferative to a secretory state and, together with estradiol, maintains endometrial integrity Citation[5] preparing the endometrium for implantation. It inhibits uterine contractility Citation[6].

In uterus, progesterone controls the growth and differentiation of endometrial and myometrial cells. During the luteal phase, in the primate uterus, progesterone inhibits estrogen induced mitotic activity in the functional zones of the endometrial epithelium but shows some stimulatory effect on both the basalis and endometrial angiogenesis Citation[7]. Progesterone can also play a key role on growth of benign smooth muscles tumours from uterine myometrium Citation[8-10]. In normal breast epithelial cells, progesterone has important mitogenic properties with a peak of a mitotic activity during the luteal phase Citation[11].

Synthetic progestins administered with estrogen in post-menopausal women are involved in the moderately increased risk of breast cancer Citation[12],Citation[13]. In the mammary glands of nulliparous Brca1/p53 deficient mice, the PA mifepristone prevents mammary tumorigenesis Citation[14].

From the physiological properties of progesterone and the pharmacological profile of synthetic progestins, and their possible drawbacks, it appears that the potential advantages and the clinical applications of selective progesterone receptor modulators (SPRMs) are very promising in major public health areas.

Clinical application of the SPRMs

Medical abortion

Mifepristone, the first glucocorticoids, and PA Citation[2], a derivative of norethindrone, alters the endometrium and causes decidual necrosis and trophoblast to separate from decidua Citation[2],Citation[15-17]. Mifepristone sensitises the pregnant uterus and cervix to endogenous and exogenous prostaglandins, increasing uterine activity and inducing cervical softening. Mifepristone is well absorbed orally and reaches a peak serum concentration in pregnant woman within 2 hours. The pharmacokinetics is similar for any dose over 100 mg. The half-life of mifepristone is approximately 19 hours in pregnant women Citation[16].

Mifepristone used alone with oral doses ranging from 50 to 400 mg allows complete abortion in 60–80% up to 48 days gestation. The addition of small doses of a prostaglandin analogue (such as misoprostol) has a synergistic effect resulting in nearly 100% complete abortion Citation[18],Citation[19] with lower doses than those required when the prostaglandin analogue is used alone. Moreover, the addition of mifepristone to misoprostol increases complete abortion at a faster rate up to 63 days gestation Citation[16].

The most commonly used medical abortion regimen worldwide is presently mifepristone followed by a prostaglandin analogue. The FDA-approved regimen consists of 600 mg of oral mifepristone followed by a prostaglandin analogue, usually misoprostol, 36–48 hours later. This regimen has been reported to induce complete abortion in 92–99% women Citation[20-23]. As expected on the basis of its pharmacokinetics, lower doses of mifepristone are equally effective as the 600 mg dose when combined with a prostaglandin analogue Citation[24-26]. In the large trial performed by the World Health Organisation, pregnant women were included up to 63 days gestation and received either 200 mg or 600 mg mifepristone, followed 48 hours later by an oral administration of 400 μg misoprostol. Both groups showed similar complete abortion rates (89 and 88%, respectively) Citation[26]. This efficacy is similar to that of vacuum aspiration. In addition, studies using 200 mg mifepristone combined with 800 μg misoprostol administered vaginally to more than 4000 women also confirmed the efficacy of this low dose of mifepristone Citation[27-29]. The vaginal administration of misoprostol prolongs the efficacy of mifepristone regimens up to 63 days. A shorter interval between the administration of mifepristone and prostaglandin analogue does not impair the rate of abortion Citation[30].

The use of mifepristone between 9 and 13 weeks gestation is equally effective as a cervical primer Citation[31]. Animal studies showed that mifepristone induces collagen remodelling. It caused a decrease in collagen organisation with decreased fibril length and diameter Citation[32]. The prostaglandin analogue, misoprostol, is more commonly used in this indication because it is cheaper and largely available. Nevertheless, bleeding and abdominal pain are decreased with mifepristone compared with misoprostol Citation[31],Citation[33].

Mifepristone for second trimester abortion decreases the interval between induction to abortion and increases the success rate. It is administered in either 200 or 600 mg dosing and followed by prostaglandin analogue.

Management of miscarriage

A randomised placebo controlled trial showed that expulsion occurred in 82% of women with first trimester arrested pregnancy, within 5 days after administration of 600 mg mifepristone compared with only 8% of women given placebo but the success rate was comparable with that of a misoprostol used alone Citation[34].

Emergency contraception

Emergency contraception (EC) is a term used to describe a group of methods for preventing an unwanted pregnancy that are administered during the first few days after unprotected intercourse. It can decrease individual woman's pregnancy risks by as much as 89% after a single coitus. EC is an important action for woman giving them a chance to avoid the psychological and physical consequences of unwanted pregnancy including the need for legal or clandestine abortion Citation[35].

Several approaches to EC have been described, including high doses of estrogens, danazol, intra-uterine devices, oral contraceptive with estrogen and progestin Citation[36], a progestin alone (levonorgestrel) and SPRMs (mifepristone and VA 2914). The Yuzpe regimen involved the combined use of ethinyl estradiol 100 μg and 0.5 mg of levonorgestrel, repeated once 12 hours apart, with the first dose given within 72 hours of unprotected inter-course. This regimen, which was popular in the late seventies and early eighties, has now been supplanted by a more effective and better tolerated progestin only product, containing levonorgestrel. They were to be used within 72 hours of unprotected inter-course.

PR modulators offer another option for EC. They can maintain efficacy for periods of time, longer than 72 hours in a single dose regimen Citation[37],Citation[38]. A higher efficacy of mifepristone compared with the Yuzpe regimen was also observed. Side effects such as nausea, vomiting, headache, dizziness, fatigue, low abdominal pain and hot flushes were observed less frequently in women receiving mifepristone Citation[39]. Lowering the dose of mifepristone from 600 to 10 mg did not significantly impair its effectiveness as an emergency contraceptive Citation[40]. Low doses of mifepristone are associated with less disturbance of the menstrual cycle length. A dose as low as 10 mg seems preferable to the 600 mg dose Citation[38]. This trial also showed that unlike levonorgestrel or Yuzpe regimen, the efficacy of mifepristone does not appear to decline with increased interval up to 120 h.

Recently, a second generation PR modulator with lower anti-glucocorticoid activity than mifepristone was tested in EC. Participants were randomly assigned to receive a single dose of 50 μg of VA 2914 plus a placebo 12 hours later or two doses of 0.75 mg of levonorgestrel taken 12 hours apart. VA 2914 was at least as effective as levonorgestrel in preventing pregnancy after unprotected intercourse. Lower efficacy with increasing interval between intercourse and EC treatment above 48 hours was observed only with levonorgestrel. In contrast, the effectiveness of VA 2914 did not decline after 48 hours. Adverse effects were generally similar in both treatment groups, but more nausea was observed among the VA 2914 treated women. Nausea, as a side effect, has not been reported in other trials with VA 2914 at higher doses. Moreover, VA 2914 was also effective in preventing pregnancy when administered after ovulation Citation[36]. In both groups, women experienced considerable variation in menstrual cycle length when compared with their reported individual normal cycle length. On average, the onset of menses after EC use was 2, 1 day earlier than anticipated in levonorgestrel users and 2, 6 days later in VA 2914 users Citation[37].

Acceleration of tubal transport of fertilised eggs has been reported in rats exposed to anti-progestogen Citation[41]. No information is available in women. It is well documented, however, that mifepristone does not increase the risk of tubal pregnancy Citation[41],Citation[42].

Long-term contraception

PAs and SPRMs do have contraceptive potential possibly, by several mechanisms Citation[5],Citation[43]. They inhibit ovulation by blocking the LH surge and can induce endometrial desynchronisation, thereby interfering with implantation. Recently, mifepristone but not levonorgestrel was shown to inhibit human blastocyst attachment to an in vitro endometrial three-dimensional cell culture model Citation[44]. High doses of mifepristone may even induce follicular atresia Citation[45].

The threshold dose for ovulation inhibition is 2 mg/day. At a lower dosage, ovulation occurs and the administration of 0.5 mg mifepristone daily or 5 mg weekly is not effective in pregnancy prevention Citation[46],Citation[47]. Two-hundred milligrams of mifepristone, administered 48 hours after the LH surge, shows contraceptive efficacy. However, this option is not clinically relevant because the detection of an LH surge routinely is an expensive and an unreliable method Citation[5],Citation[47].

Administration of mifepristone, in the late luteal phase, is not effective in pregnancy prevention Citation[48],Citation[49].

VA 2914 is an orally active steroidal SPRM which demonstrates potent PA activity in vivo and in vitroCitation[50] with a reduced anti-glucocorticoid activity compared with mifepristone Citation[51]. This compound shows anti-ovulatory activity and post-coïtal anti-fertility activity in rats Citation[52].

We evaluated ovulation inhibition by VA 2914 in women, in a continuous regimen of administration of 2.5, 5 and 10 mg/day for 3 months. We also examined the endometrial impact in each group.

Anovulation (defined by absence of progesterone above 3 ng/mL) was obtained in nearly 80% women in the 5 and 10 mg/day groups with a high rate of amenorrhea (81.2 and 90%, respectively). Plasma estradiol levels remained in the physiological follicular phase range.

Endometrial histological analysis showed pre-dominantly a pattern of secretory phase. Some cystic glandular dilatations were observed in rare cases. No hyperplasia was detected Citation[53].

We quantified the effects of VA 2914 on endometrial vascularisation, fibrillar matrix and vascular endothelial growth factor (VEGF)-A expression in endometrial biopsies from 41 women before and after 12 weeks treatment. No changes were noted in structure, number and size of endometrial vessels. The collagen network and VEGF-A distribution remained comparable during the luteal phase at baseline and under VA 2914 treatment Citation[54]. From these observations, we conclude that long-term VA 2914 treatment does not result in an endometrial morphology comparable with that induced by a progestin. It thus acts on endometrial cells, in a specific way, which is clearly distinct from that of a progestin.

Treatment of uterine leiomyomata

Uterine leiomyomata, also named fibroids, are benign tumours that occur in up of 35% women above 35 years. They account for up to 40% of all hysterectomies Citation[55]. Non-surgical treatment options for symptomatic leiomyomata are limited because gonadotropin releasing hormone agonists (GnRHa) induce hypo estrogenism and side effects limiting the treatment duration Citation[56]. Even, when a decrease in leiomyomata size of 36% is observed after 12 weeks of GnRHa treatment, the uterus returns to pre-treatment size within 6 months after treatment completion. The indications of GnRHa are thus limited to short-term pre-surgical treatment Citation[57].

Progesterone and PR seem to play a key role in the control of uterine fibroid growth Citation[58]. Mitotic activity is maximal during the luteal phase Citation[59]. Several studies have shown an up-regulation of PR in uterine leiomyomata compared with normal adjacent myometrium at mRNA and protein levels Citation[60]. The expression of the proliferation marker Ki-67 is also increased in leiomyomata, when compared with that in normal myometrium, and up-regulated by progesterone Citation[60]. Expression of epidermal growth factor, a proliferative cytokine, and Bcl2, an apoptosis-inhibiting protein is also increased in fibroids relative to the adjacent myometrium, specifically during the secretory phase Citation[61]. Progesterone increases Bcl2 protein expression in primary leiomyomata cell cultures Citation[62]. Progestins finally attenuate or even reverse the inhibitory effects of GnRHa on leiomyomata size when used as add-back therapy Citation[63-65]. However, Mifepristone has opposite effects Citation[66] and may decrease proliferation of these smooth muscle cells, suggesting a clinical usefulness of SPRMs for the treatment of myomas. Asoprisnil, a mixed progesterone agonist/PA with no anti-glucocorticoid effect, inhibits proliferation and induces apoptosis in cultured uterine leiomyoma cells in the absence of comparable effects on cultured normal myometrial cells suggesting a cell type specific effect Citation[67].

Down regulation of VEGF, of adrenomedullin, a vasoactive hormone and of their receptors was shown by western blot analysis in cultured human uterine leiomyoma cells treated with VA 2914. This action of VA 2914 was not observed in the surrounding normal myometrial cells, suggesting a cell type specific action of this SPRM on leiomyoma cells Citation[68].

In clinical studies, daily treatment with 5–50 mg mifepristone for 3–6 months resulted in a reduction in uterus and leiomyoma volumes, ranging from 27 to 49% and 26 to 74%, respectively. Moreover, the prevalence and severity of dysmenorrhoea, menorrhagia and pelvis pressure were reduced. Treatment was well tolerated. Nevertheless, endometrial hyperplasia, a serious adverse effect of mifepristone was detected in 28% women Citation[65]. A 20% reduction of myoma volume was also observed in over 90% patients treated with 12.5 mg mifepristone or GnRHa Citation[69]. The recurrence rate after cessation of treatment was 40% after GnRHa and 17.8% after mifepristone Citation[64].

Asoprisnil (5, 10 or 25 mg) given orally, once daily for 12 weeks reduced the uterine volume as well as the volume of the largest leiomyoma in a dose-dependant manner. At 10 and 25 mg, it suppressed pelvis pressure after 12-weeks treatment in contrast to placebo, which was inactive. No decrease of plasma estradiol or increase of cortisol was observed. This treatment is well tolerated and reduces significantly both duration and intensity of uterine bleeding in a dose-dependant manner Citation[70].

Treatment of endometriosis

Endometriosis is an estrogen-dependent disease due to ectopic endometrium. It causes pelvic pain, dyspareunia, dysmenorrhoea and infertility Citation[71]. Pelvic pain is the result of a local inflammatory reaction and up-regulation of cyclooxygenase (COX)- 2. The use of COX-2 inhibitors is therefore an effective symptomatic option Citation[72]. Use of therapeutic agents, which hinder endometriosis, is limited by their side effects. GnRHa and GnRH antagonists use results in a hypo estrogenic state with hot flushes and bone loss, which limits the treatment duration. Progestins, the other therapeutic option, can induce bloating, breakthrough bleeding, mood changes, acne, hirsutism that can alter therapeutic compliance Citation[73].

Inhibiting ectopic endometrium proliferation without inducing estrogen deprivation would be an important goal for the treatment of endometriosis. In this indication, SPRMs may ultimately have an important place. Mifepristone inhibits endometrial cells proliferation by activating the nuclear factor-kappa B signalling pathway Citation[74]. Mifepristone also promotes apoptosis in human endometrial cells by over expression of Bax, the apoptosis promoting gene and by down-regulation of the anti-apoptosis gene Bcl2Citation[75].

Mifepristone (5 or 50 mg/day for 6 months or 100 mg/day for 3 months) improved the clinical symptoms associated to endometriosis Citation[75]. The 50 mg daily dose elicited a mean 55% regression of visible endometriosis after 6 months of treatment.

Asoprisnil, which inhibits endometrial proliferation and prostaglandin synthesis Citation[76], was studied in subjects with a laparoscopic diagnosis of endometriosis at 5, 10 and 25 mg (versus placebo) for 12 weeks. All doses were significantly effective on pain scores, at all treatment months compared with placebo Citation[70]. The effect on bleeding pattern was also dose-dependent. Asoprisnil was well tolerated in short-term studies, and no serious adverse event was reported during treatment period and follow-up Citation[76].

Conclusion

PAs and SPRMs are largely used for fertility control. They have proven efficacy for abortion and EC. In long-term contraception, they offer an estrogen-free contraception with a better bleeding pattern than that associated with progestins. Because they control the growth of leiomyoma and endometrial cells without inducing an hypo estrogenic state, their use in endometriosis and symptomatic leiomyomata could be promising. Large Phase III trials in patients with menorrhagia associated with uterine fibroids are in progress. Efficacy and safety of long-term administration for management of uterine bleeding, endometriosis and uterine fibroids are actively studied.

On the basis of their pharmacological and clinical features, SPRMs could have in the future important potential implications in women's health care. Some of them such as asoprisnil and mifepristone inhibit mammary epithelial cells proliferation, in animal models. They might have a key role in the prevention and treatment of benign and malignant breast pathologies.

Some concern appeared initially about endometrial safety because hyperplasia was described on mifepristone treatment. Endometrial biopsies from patients treated with different SPRMs were recently reviewed by a panel of experienced pathologists to develop consensus observations and recommendations Citation[77]. New terminology and diagnostic criteria were defined as PRM- associated endometrial changes. These observations are reassuring as no pre-malignant lesions were seen. However, long-term follow-up is necessary to better define the specific role of this new class of agents as well as of their regimen of administration.

Declaration of interest: The authors report no conflicts of interest. The authors alone are responsible for the content and writing of the paper.

References

  • Chwalisz K, Perez M C, DeManno D, Winkel C, Schubert G, Elger W. Selective progesterone receptor modulator development and use in the treatment of leiomyomata and endometriosis. Endocr Rev 2005; 26: 423–438
  • Philibert D. RU38486: an original multifacted antihormone in vivo. Adrenal Steroid Antagonism, M Agarwal. Walter de Gruyter and Co, Berlin 1984; 77–101
  • Neef G, Beier S, Elger W, Henderson D, Wiechert R. New steroids with anti-progestational and anti-glucocorticoid activities. Steroids 1984; 44: 349–372
  • Kloosterboer H J, Deckers G H, Van de Heuvel M J, Loozen H J. Screening of anti-progestagens by receptor studies and bioassays. J Steroid Biochem 1988; 31: 567–571
  • Chabbert-Buffet N, Meduri G, Bouchard P, Spitz E. Selective progesterone receptor modulator and progesterone antagonist: mechanism of action and clinical application. Hum Reprod Update 2005; 11: 293–307
  • Csapo A. Progesterone block. Am J Anat 1956; 98: 273–291
  • Padykula H A. Regeneration in the primate uterus: the role of stem cells. Ann NY Acad Sci 1991; 622: 47–56
  • Brandon D D, Bethea C L, Strawn E Y, Novy M J, Burry K A, Harrington M S, Erickson T E, Warner C, Keenan E J, Clinton G M. Progesterone receptor messenger ribonucleic acid and protein are over expressed in human uterine leiomyomas. Am J Obstet Gynecol 1993; 169: 78–85
  • Nisolle M, Gillerot S, Casanas-Roux F, Squifflet J, Berliere M, Donnez J. Immunohistochemical study of the proliferation index, oestrogen receptors and progesterone receptors A and B in leiomyomata and normal myometrium during the menstrual cycle and under gonadotrophin-releasing hormone agonist therapy. Hum Reprod 1999; 14: 2844–2850
  • Kawaguchi K, Fujii S, Konishi I, Nanbu Y, Nonogaki H, Mori T. Mitotic activity in uterine leiomyomas during the menstrual cycle. Am J Obstet Gynecol 1989; 160: 637–641
  • Lundstrom E, Wilczek B, von Palffy Z, Soderqvist G, von Schoultz B. Mammographic breast density during hormone replacement therapy: differences according to treatment. Am J Obstet Gynecol 1999; 181: 348–352
  • Rossouw J E, Anderson G L, Prentice R L, LaCroix A Z, Kooperberg C, Stefanick M L, Jackson R D, Beresford S A, Howard B V, Johnson K C, et al. Risks and benefits of estrogen plus progestin in healthy post-menopausal women: principal results from the Women's Health Initiative randomized controlled trial. JAMA 2002; 288: 321–333
  • Beral V. Breast cancer and hormone-replacement therapy in the million women study. Lancet 2003; 362: 419–427
  • Poole A J, Li Y, Kim Y, Lin S C, Lee W H, Lee E Y. Prevention of Brca1-mediated mammary tumorigenesis in mice by a progesterone antagonist. Science 2006; 314: 1467–1470
  • Mahajan D K, London S N. Mifepristone (RU 486): a review. Fertil Steril 1997; 86: 967–976
  • Hermann W L, Schlindler A M, Wyss R, Bischoff P. Effects of anti-progesterone RU 486 in early pregnancy and during the menstrual cycle. The Antiprogestin Steroid RU 486 and Human Fertility Control, E E Beaulieu, S Siegel. Plenum, New York 1985; 259–262
  • Schreiber C, Creinin M. Mifepristone in abortion care. Semin Reprod Med 2005; 23: 82–91
  • Tang O S, Ho P C. Clinical applications of mifepristone. Gynecol Endocrinol 2006; 22: 655–659
  • Kovacs L, Sas M, Resch B A, Ugocsai G, Swahn M L, Bygdeman M, Rowe P J. Termination of very early pregnancy by RU 486, an anti-progestational compound. Contraception 1984; 29: 399–410
  • Shoupe D, Mishell D R, Jr, Brenner P F, Spitz I M. Pregnancy termination with a high and medium dosage regimen of RU-486. Contraception 1986; 33: 455–461
  • Peyron R, Aubény E, Targosz V, Silvestre L, Renault M, Elkik F, Leclerc P, Ulmann A, Baulieu E E. Early termination on pregnancy with mifepristone (RU 486) and the orally active prostaglandin misoprostol. N Engl J Med 1993; 328: 1509–1513
  • Aubeny E, Peyron R, Turpin C L, Renault M, Targosz V, Silvestre L, Ulmann A, Baulieu E E. Termination of early pregnancy (up to 63 days of amenorrhea) with mifepristone (RU 486) and increasing doses of misoprostol. Int J Fertil Menopausal Stud 1995; 40(Suppl 2)85–91
  • Spitz I M, Bardin C W, Benton L, Robbins A. Early pregnancy termination with mifepristone and misoprostol in the United States. N Engl J Med 1998; 338: 1241–1247
  • Aubeny E. A two-stage increase in the dose of misoprostol improves the efficacy of medical abortion with mifepristone and prostaglandins (letter). Eur J Contracept Reprod Health Care 2001; 6: 54–55
  • World Health Organization Task Force on Post-ovulatory Methods for Fertility Regulation. Termination of pregnancy with reduced doses of mifepristone. BMJ 1993; 307: 532–537
  • McKinley C, Thong K J, Baird D T. The effect of dose of mifepristone and gestation on efficacy of medical abortion with mifepristone and misoprostol. Hum Reprod 1993; 8: 1502–1505
  • World Health Organization Task Force on Post-ovulatory Methods for Fertility Regulation. Comparison of two-doses of mifepristone in combination with misoprostol for early medical abortion: a randomised trial. BJOG 2000; 107: 524–530
  • Schaff E A, Eisinger S H, Stadalius L S, Franks P, Gore B Z, Poppema S. Low-dose mifepristone 200 mg and vaginal misoprostol for abortion. Contraception 1999; 59: 1–6
  • Ashok P, Penney G, Flett G, Templeton A. An effective regimen for early medical abortion: a report of 2000 consecutive cases. Hum Reprod 1998; 13: 2962–2965
  • Schaff E A, Fielding S L, Eisinger S H, Stadalius L S, Fuller L. Low-dose mifepristone followed by vaginal misoprostol at 48 hours for abortion up to 63 days. Contraception 2000; 61: 41–46
  • Schaff E A, Fielding S L, Westhoff C, Ellertson C, Eisinger S H, Stadalius L S, Fuller L. Vaginal misoprostol administrated 1, 2, or 3 days after mifepristone for early medical abortion: a randomized trial. JAMA 2000; 284: 1948–1953
  • Ashok P W, Gilian F, Templeton A. Mifepristone versus vaginally administered misoprostol for cervical priming before first-trimester termination of pregnancy: a randomized, controlled study. Am J Obstet Gynecol 2000; 183: 998–1002
  • Ngai S W, Yeung K C, Lao T, Ho P C. Oral misoprostol versus mifepristone for cervical dilatation before vacuum aspiration in first trimester nulliparous pregnancies: a double blind prospective randomized study. BJOG 1996; 103: 1120–1123
  • Tang O S, Ho P C. Clinical applications of mifepristone. Gynecol Endocrinol 2006; 22: 655–659
  • Westley E, von Hertzen H, Faundes A. Expanding access to emergency contraception. Int J Gynaecol Obstet 2007; 97: 235–237
  • Creinin M D, Schlaff W, Archer D F, Wan L, Frezieres R, Thomas M, Rosenberg M, Higgins J. Progesterone receptor modulator for emergency contraception: a randomized controlled trial. Obstet Gynecol 2006; 108: 1089–1097
  • von Hertzen H, Piaggio G, Ding J, Chen J, Song S, Bartfai G, Ng E, Gemzell-Sanielsson K, Oyunbileg A, Wu S. Low dose mifepristone and two regimens of levonorgestrel for emergency contraception: a WHO multicentre randomised trial. Lancet 2002; 360: 1803–1810
  • Comparison of three single doses of mifepristone as emergency contraception: a randomized trial. Task force on post-ovulatory methods of fertility regulation. Lancet 1999; 353: 697–702
  • Kulier R, Gulmezoglu A M, Hofmeyr G J, Cheng L N, Campana A. Medical methods for first trimester abortion (review). Cochrane Database Syst Rev 2004; 1, CD002855
  • Jin J, Weisberg E, Fraser I S. Comparison of three single doses of mifepristone as emergency contraception: a randomised controlled trial. Aust NZ J Obstet Gynaecol 2005; 45: 489–494
  • Roblero L S, Croxatto H B. Effects of RU 486 on development and implantation of rat embryos. Mol Reprod Dev 1991; 29: 342–346
  • Pisarska M D, Carson S A, Buster J E. Ectopic pregnancy. Lancet 1998; 351: 1115–1120
  • Spitz I M. Progesterone receptor antagonists. Curr Opin Investig Drugs 2006; 7: 882–890, (review)
  • Lalitkumar P GL, Lalitkumar S, Meng C X, Stavreus-Evers A, Hambiliki F, Bentin-Ley U, Gemzell-Danielsson K. Mifepristone, but not levonorgestrel, inhibits human blastocyst attachment to an in vitro endometrial three-dimensional cell culture model. Hum Reprod 2007; 22: 3031–3037
  • Croxatto H B, Salvatierra A M, Fuentealba B, Leiva L. Follicle stimulating hormone-granulosa cell axis involvement in the anti-folliculotrophic effect of low dose mifepristone (RU486). Hum Reprod 1995; 10: 1987–1991
  • Marions L, Viski S, Danielsson K G, Resch B A, Swahn M L, Bygdeman M, Kovâcs L. Contraceptive efficacy of daily administration of 0.5 mg mifepristone. Hum Reprod 1999; 14: 2788–2790
  • Bygdeman M, Danielsson K G, Marions L, Swahn M L. Contraceptive use of antiprogestin. Eur J Contracept Reprod Health Care 1999; 4: 103–107, (review)
  • Spitz I M, Chwalisz K. Progesterone receptor modulators and progesterone antagonists in women's health. Steroids 2000; 65: 807–815, (review)
  • Swahn M L, Bygdeman M, Chen J K, Gemzell-Danielsson K, Song S, Yang Q Y, Yang P J, Qian M L, Chang W F. Once-a-month treatment with a combination of mifepristone and the prostaglandin analogue misoprostol. Hum Reprod 1999; 14: 485–488
  • Gainer E E, Ulmann A. Pharmacologic properties of CDB(VA)- 2914. Steroids 2003; 68: 1005–1011
  • Attardi B J, Burgenson J, Hild S A, Reel J R. In vitro anti-progestational/anti-glucocorticoid activity and progestin and glucocorticoid receptor binding of the putative metabolites and synthetic derivatives of CDB-2914, CDB-4124, and mifepristone. J Steroid Biochem Mol Biol 2004; 88: 277–288
  • Reel J R, Hild-Petito S, Blye R P. Anti-ovulatory and post-coital antifertility activity of the antiprogestin CDB-2914 when administered as single, multiple, or continuous doses to rats. Contraception 1998; 58: 129–136
  • Chabbert-Buffet N, Pintiaux-Kairis A, Bouchard, VA2914 Study Group. Effects of the progesterone receptor modulator VA2914 in a continuous low dose on the hypothalamic-pituitary-ovarian axis and endometrium in normal women: a prospective, randomized, placebo-controlled trial. J Clin Endocrinol Metab 2007; 92: 3582–3589
  • Ravet S, Munaut C, Blacher S, Brichant G, Labied S, Beliard A, Chabbert-Buffet N, Bouchard P, Foidart J-M, Pintiaux A, Persistence of an intact endometrial matrix and vessels structure in women exposed to VA-2914, a selective progesterone receptor modulator. J Clin Endocrinol Metab 2008;93:4525–4531. Epub 2008 Aug
  • Whiteman M K, Hillis S D, Jamieson D J, Morrow B, Podgornik M N, Brett K M, Marchbanks P A. Inpatient hysterectomy surveillance in the United States, 2000–2004. AM J Obstet Gynecol 2008; 198: 34.e1–e7
  • Dodin S, Lemay A, Maheux R, Dumont M, Turcot-Lemay L. Bone mass in endometriosis patients treated with GnRH agonist implant or danazol. Obstet Gynecol 1991; 77: 410–415
  • Wang P H, Yang A H, Yuan C C, Lee W L, Chao H T. Uterine myoma after cessation of gonadotropin-releasing hormone agonist: ultrasound and histopathologic findings. Zhonghua Yi Xue Za Zhi (Taipei) 1998; 61: 625–629
  • Pavlovich S V, Volkov N I, Burlev V A. Proliferative activity and level of steroid hormone receptors in the myometrium and myoma nodes in different phases of menstrual cycle. Bull Exp Biol Med 2003; 136: 396–398
  • Nisolle M, Gillerot S, Casanas-Roux F, Squifflet J, Berliere M, Donnez J. Immunohistochemical study of the proliferation index, oestrogen receptors and progesterone receptors A and B in leiomyomata and normal myometrium during the menstrual cycle and under gonadotrophin-releasing hormone agonist therapy. Hum Reprod 1999; 14: 2844–2850
  • Brandon D D, Bethea C L, Strawn E Y, Novy M J, Burry K A, Harrington M S, Erickson T E, Warner C, Keenan E J, Clinton G M. Progesterone receptor messenger ribonucleic acid and protein are over expressed in human uterine leiomyomas. Am J Obstet Gynecol 1993; 169: 78–85
  • Harrison-Woolrych M L, Charnock-Jones D S, Smith S K. Quantification of messenger ribonucleic acid for epidermal growth factor in human myometrium and leiomyomata using reverse transcriptase polymerase chain reaction. J Clin Endocrinol Metab 1994; 78: 1179–1184
  • Matsuo H, Maruo T, Samoto T. Increased expression of Bcl-2 protein in human uterine leiomyoma and its up-regulation by progesterone. J Clin Endocrinol Metab 1997; 82: 293–299
  • Carr B R, Marshburn P B, Weatherall P T, Bradshaw K D, Breslau N A, Byrd W, Roark M, Steinkampf M P. An evaluation of the effect of gonadotropin-releasing hormone analogues and medroxyprogesterone acetate on uterine leiomyomata volume by magnetic resonance imaging: a prospective, randomized, double blind, placebo-controlled, crossover trial. J Clin Endocrinol Metab 1993; 76: 1217–1223
  • Zeng C, Gu M, Huang H, [A clinical control study on the treatment of uterine leiomyoma with gonadotrophin releasing hormone agonist or mifepristone] Zhonghua Fu Chan Ke Za Zhi 1998;33:490–492
  • Mizutani T, Sugihara A, Honma H, Komura H, Nakamuro K, Terada N. Effect of steroid add-back therapy on the proliferative activity of uterine leiomyoma cells under gonadotropin-releasing hormone agonist therapy. Gynecol Endocrinol 2005; 20: 80–83
  • Murphy A A, Morales A J, Kettel L M, Yen S S. Regression of uterine leiomyomata to the antiprogesterone RU486: dose-response effect. Fertil Steril 1995; 64: 187–190
  • Ohara N, Morikawa A, Chen W, Wang J, DeManno D A, Chwalisz K, Maruo T. Comparative effects of SPRM asoprisnil (J867) on proliferation, apoptosis, and the expression of growth factors in cultured uterine leiomyoma cells and normal myometrial cells. Reprod Sci 2007; 14(8 Suppl)20–27
  • Xu Q, Ohara N, Chen W, Liu J, Sasaki H, Morikawa A, Sitruk-Ware R, Johansson E D, Maruo T. Progesterone receptor modulator CDB-2914 down-regulates vascular endothelial growth factor, adrenomedullin and their receptors and modulates progesterone receptor content in cultured human uterine leiomyoma cells. Hum Reprod 2006; 21: 2408–2416
  • Steinauer J, Pritts E A, Jackson R, Jacoby A F. Systematic review of mifepristone for the treatment of uterine leiomyomata. Obstet Gynecol 2004; 103: 1331–1336, (review)
  • Chwalisz K, Perez M C, Demanno D, Winkel C, Schubert G, Elger W. Selective progesterone receptor modulator development and use in the treatment of leiomyomata and endometriosis. Endocr Rev 2005; 26: 423–438, (review). Erratum in: Endocr Rev 2005;26:703
  • Giudice L C, Kao L C. Endometriosis. Lancet 2004; 364: 1789–1799, (review)
  • Kyama C M, Mihalyi A, Simsa P, Mwenda J M, Tomassetti C, Meuleman C, D'Hooghe T M. Non-steroidal targets in the diagnosis and treatment of endometriosis. Curr Med Chem 2008; 15: 1006–1017, (review)
  • Rodgers A K, Falcone T. Treatment strategies for endometriosis. Expert Opin Pharmacother 2008; 9: 243–255, (review)
  • Han S, Sidell N. RU486-induced growth inhibition of human endometrial cells involves the nuclear factor-κ B signaling pathway. J Clin Endocrinol Metab 2003; 88: 713–719
  • Kettel L M, Murphy A A, Morales A J, Ulmann A, Baulieu E E, Yen S S. Treatment of endometriosis with the anti-progesterone mifepristone (RU486). Fertil Steril 1996; 65: 23–28
  • Elger W, Bartley J, Schneider B, Kaufmann G, Schubert G, Chwalisz K. Endocrine pharmacological characterization of progesterone antagonists and progesterone receptor modulators with respect to PR-agonistic and antagonistic activity. Steroids 2000; 65: 713–723, (review)
  • Mutter G L, Bergeron C, Deligdisch L, Ferenczy A, Glant M, Merino M, Williams A R, Blithe D L. The spectrum of endometrial pathology induced by progesterone receptor modulators. Mod Pathol 2008; 21: 591–598

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.