the molecular biology of ru486. is there a role for ... mole… · may, 1992 ru486 and breast...

18
0163-769X/92/1302-0146$03.00/0 Endocrine Reviews Copyright 0 1992 by The Endocrine Society Vol. 13, No. 2 Printed in U.S.A. The Molecular Biology of RU486. Is There a Role for Antiprogestins in the Treatment of Breast Cancer?* KATHRYN B. HORWITZ DeDartments of Medicine and Patholot!v. Universitv of Colorado Health Sciences Center, Division of Endocrinology,. Denuer, Colorado 8026~ I. Introduction II. Progesterone and the Normal Breast III. Progesterone and Breast Cancer A. Progestin agonists and tumor induction B. Progestin agonists and growth of established tumors IV. Molecular Mechanisms of Progesterone Antagonists A. Progesterone receptors B. Molecular mechanisms of progesterone antagonists 1. Affinity for inactive PR - 2. PR activation 3. DNA binding 4. Dimerization 5. Transcription 6. PR “processing” or down-regulation V. Progesterone Antagonists and the Treatment of Breast Cancer A. Human breast cancer cell lines B. Animal models of mammary cancer C. Human clinical trials VI. Progestin Resistance VII. Summary and Future Prospects I. Introduction E NDOCRINE therapy used either prophylactically or therapeutically for the treatment of locally ad- vanced or metastatic breast cancers offers many advan- tages to patients whose tumors contain functional estro- gen and progesterone receptors (ER and PR). The range of treatments defined as endocrine include surgical abla- tion of endocrine glands, administration of pharmacolog- ical doses of steroid hormones, chemical blockade of steroid hormone biosynthesis, and inhibition of endoge- nous steroid hormone action at the tumor with synthetic antagonists. The last of these approaches is the most widely used, making the antiestrogen tamoxifen the pre- ferred first-line therapeutic agent for treatment of hor- mone-dependent metastatic breast cancer. The wide- Address requests for reprints to: Kathryn B. Horwitz, Ph.D., Divi- sion of Endocrinology, Box B151, University of Colorado Health Sci- ences Center, 4200 East Ninth Avenue, Denver, Colorado 80262. *Supported in part by grants from the National Cancer Institute and the American Cancer Society. spread use of tamoxifen reflects its efficacy and low toxicity, and the fact that it makes good physiological sense to block the local proliferative effects of estrogens directly at the breast. But are estrogens the only hor- mones with a proliferative impact on the breast and on breast cancers? This review focuses on evidence that progesterone also has proliferative actions in the breast; on the role of synthetic progestins in breast cancer treatment; on the molecular biology of progesterone an- tagonists; and on the preliminary data showing that progesterone antagonists may be powerful new tools for the management of metastatic breast cancer, because they block the local effects of endogenous progesterone on breast cell proliferation. The reader is also referred to the excellent general review on progestin regulation of cell proliferation by Clarke and Sutherland (1). The structures of the agonists and antagonists discussed in this review are shown in Fig. 1. II. Progesterone and the Normal Breast Conventional wisdom holds that the mechanisms by which estradiol and progesterone regulate the prolifera- tion and differentiation of uterine epithelial cells apply equally to the breast. This is probably inaccurate (2-4). In the uterus, estrogens are clearly mitogenic, and addi- tion of progesterone to the estrogenized endometrium leads to the appearance of a secretory pattern character- ized by cells engaged in protein synthesis rather than cell division (5). That is, in the uterus, estradiol is a proliferative hormone; progesterone is a differentiating hormone. For this reason the unopposed actions of estra- diol are considered to be tumorigenic in the uterus, while the risk of endometrial hyperplasia and cancer is lowered when estrogens are combined with progestins. In fact, the combined regimen may even be protective since a decrease in endometrial cancers has been reported in women prescribed combined estrogens and progestins, compared to women receiving no treatment (Ref. 6 and references therein). 146 on February 27, 2005 edrv.endojournals.org Downloaded from

Upload: others

Post on 04-Oct-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: The Molecular Biology of RU486. Is There a Role for ... mole… · May, 1992 RU486 AND BREAST CANCER FIG. 1. The structure of progesterone, the synthetic progestin agonist R5020,

0163-769X/92/1302-0146$03.00/0 Endocrine Reviews Copyright 0 1992 by The Endocrine Society

Vol. 13, No. 2 Printed in U.S.A.

The Molecular Biology of RU486. Is There a Role for Antiprogestins in the Treatment of Breast Cancer?*

KATHRYN B. HORWITZ

DeDartments of Medicine and Patholot!v. Universitv of Colorado Health Sciences Center, Division of

Endocrinology,. Denuer, Colorado 8026~ ’

I. Introduction II. Progesterone and the Normal Breast

III. Progesterone and Breast Cancer A. Progestin agonists and tumor induction B. Progestin agonists and growth of established tumors

IV. Molecular Mechanisms of Progesterone Antagonists A. Progesterone receptors B. Molecular mechanisms of progesterone antagonists

1. Affinity for inactive PR - 2. PR activation 3. DNA binding 4. Dimerization 5. Transcription 6. PR “processing” or down-regulation

V. Progesterone Antagonists and the Treatment of Breast Cancer A. Human breast cancer cell lines B. Animal models of mammary cancer C. Human clinical trials

VI. Progestin Resistance VII. Summary and Future Prospects

I. Introduction

E NDOCRINE therapy used either prophylactically or therapeutically for the treatment of locally ad-

vanced or metastatic breast cancers offers many advan- tages to patients whose tumors contain functional estro- gen and progesterone receptors (ER and PR). The range of treatments defined as endocrine include surgical abla- tion of endocrine glands, administration of pharmacolog- ical doses of steroid hormones, chemical blockade of steroid hormone biosynthesis, and inhibition of endoge- nous steroid hormone action at the tumor with synthetic antagonists. The last of these approaches is the most widely used, making the antiestrogen tamoxifen the pre- ferred first-line therapeutic agent for treatment of hor- mone-dependent metastatic breast cancer. The wide-

Address requests for reprints to: Kathryn B. Horwitz, Ph.D., Divi- sion of Endocrinology, Box B151, University of Colorado Health Sci- ences Center, 4200 East Ninth Avenue, Denver, Colorado 80262.

*Supported in part by grants from the National Cancer Institute and the American Cancer Society.

spread use of tamoxifen reflects its efficacy and low toxicity, and the fact that it makes good physiological sense to block the local proliferative effects of estrogens directly at the breast. But are estrogens the only hor- mones with a proliferative impact on the breast and on breast cancers? This review focuses on evidence that progesterone also has proliferative actions in the breast; on the role of synthetic progestins in breast cancer treatment; on the molecular biology of progesterone an- tagonists; and on the preliminary data showing that progesterone antagonists may be powerful new tools for the management of metastatic breast cancer, because they block the local effects of endogenous progesterone on breast cell proliferation. The reader is also referred to the excellent general review on progestin regulation of cell proliferation by Clarke and Sutherland (1). The structures of the agonists and antagonists discussed in this review are shown in Fig. 1.

II. Progesterone and the Normal Breast

Conventional wisdom holds that the mechanisms by which estradiol and progesterone regulate the prolifera- tion and differentiation of uterine epithelial cells apply equally to the breast. This is probably inaccurate (2-4). In the uterus, estrogens are clearly mitogenic, and addi- tion of progesterone to the estrogenized endometrium leads to the appearance of a secretory pattern character- ized by cells engaged in protein synthesis rather than cell division (5). That is, in the uterus, estradiol is a proliferative hormone; progesterone is a differentiating hormone. For this reason the unopposed actions of estra- diol are considered to be tumorigenic in the uterus, while the risk of endometrial hyperplasia and cancer is lowered when estrogens are combined with progestins. In fact, the combined regimen may even be protective since a decrease in endometrial cancers has been reported in women prescribed combined estrogens and progestins, compared to women receiving no treatment (Ref. 6 and references therein).

146

on February 27, 2005 edrv.endojournals.orgDownloaded from

Page 2: The Molecular Biology of RU486. Is There a Role for ... mole… · May, 1992 RU486 AND BREAST CANCER FIG. 1. The structure of progesterone, the synthetic progestin agonist R5020,

May, 1992 RU486 AND BREAST CANCER

FIG. 1. The structure of progesterone,

the synthetic progestin agonist R5020, and four progesterone antagonists dis-

cussed in this review.

However, considerable evidence has now accrued to suggest that in the epithelium of the breast, progesterone has a different influence. That, like estradiol, progester- one in the breast has a strong proliferative effect. Studies in support of this come both from experimental models and from normal cycling women. Both the proliferation of normal mammary epithelium in virgin mice and the lobular-alveolar development of mammary tissues in pregnant mice require progesterone (7,8). A fundamental difference in the actions of estradiol and progesterone in the breast is that the latter stimulates DNA synthesis, not only in the epithelium of the terminal bud, but also in the ductal epithelium (9). The stimulating effects of progesterone on the development of mammary gland buds can be inhibited by progesterone antagonists (10).

Data from normal human mammary cells have been more difficult to obtain and are often equivocal. Com- pared to the increase caused by estradiol treatment (11.3- fold), progesterone treatment only marginally (2.0-fold) increases the mitotic index of normal human breast ductal epithelium maintained in intact athymic nude mice (11). In fact, Mauvais-Jarvis and colleagues (12, 13) concluded, using primary cultures of epithelial cells from normal human mammary glands, that while estradiol treatment stimulates growth, progestins inhibit growth. Their data are difficult to interpret, however, since the experiments using estradiol were done with cells growing in minimally supplemented medium, while the progestin treatment studies were done with cells in optimally sup- plemented medium, and any progestin growth-stimula- tory effect might have been masked. In contrast to the sparse and conflicting in. vitro data are studies of the mitotic rate in breast epithelial cells during the normal menstrual cycle and in women taking oral contraceptives. These data show that the highest thymidine labeling indices occur during the progestin-dominated, secretory phase of the menstrual cycle. Both the estrogen and the

progestin components of oral contraceptives increase the thymidine labeling index with progestin-only formula- tions exhibiting high activity (2-4). The investigators conclude that it is difficult to sustain the idea that progestins are protective in the breast (3). It would seem that more work must be done to understand the actions of progestins in the normal breast, but that clinical decisions based on an inappropriate uterine model sys- tem are unjustified (14).

III. Progesterone and Breast Cancer

A discussion of the role of progestins in breast cancer must distinguish between their effects on carcinogenesis and their role in regulating proliferation of established cancers.

A. Progestin agonists and tumor induction

Progestin agonists have been shown to be carcinogenic or to increase the incidence of spontaneous mammary tumors in dogs and mice (E-19). In mice, results vary with the strain tested, suggesting the contribution of a genetic component; however, tumorigenic effects of pro- gestins have been observed whether or not the strain harbors the mouse mammary tumor virus (MMTV). The importance of progesterone in carcinogen-induced rat mammary cancers is documented by the early reports of Huggins et al. (20-22), who showed that pregnancy pro- motes the growth of dimethylbenzanthracene (DMBA)- induced mammary tumors, and that administration of progesterone together with the carcinogen to intact rats accelerates the appearance of tumors, increases the num- ber of tumors, and augments the growth rate of estab- lished tumors. The relationship between progestins and carcinogenesis is temporally complex. In general, proges- terone administered simultaneously with, or after, the carcinogen enhances tumorigenesis, while progesterone

on February 27, 2005 edrv.endojournals.orgDownloaded from

Page 3: The Molecular Biology of RU486. Is There a Role for ... mole… · May, 1992 RU486 AND BREAST CANCER FIG. 1. The structure of progesterone, the synthetic progestin agonist R5020,

148 HORWITZ Vol. 13. No. 2

given before the carcinogen inhibits tumorigenesis (Ref. 23 and references therein). Thus, the high progesterone levels associated with pregnancy can be protective if the hormone precedes administration of the carcinogen (24). Extrapolation of these experimental models to human disease is unclear since the only data available for the latter are epidemiological in nature and relate hormone use, particularly oral contraceptive use, to the risk of breast cancer. The trend toward increased risk with increased duration of hormone use appears repeatedly (25, 26), and a possible adverse effect of progestins ap- pears to be likely (27, 28). This is discouraging when taken together with the likelihood that in the breast, unlike the uterus, progestins enhance proliferation dur- ing the menstrual cycle.

B. Progestin agonists and growth of established tumors

Carcinogen-induced rat mammary tumors are a major model for in uiuo studies of progestin-regulated growth (23). After ovariectomy, progesterone alone is usually unsuccessful in preventing regression of established tu- mors. The rapid decrease of PR levels due to estrogen withdrawal is probably a critical factor (29). In intact animals, which more closely mimic the clinical situation, progestin agonists at moderate doses have been reported to promote tumor growth and to reverse the antitumor effects of tamoxifen (30). Thus, it is possible that endog- enous circulating progesterone may enhance breast can- cer growth. Enigmatically, progestins at higher pharma- cological doses appear to be growth inhibitory (31). The molecular mechanisms responsible for the opposing ac- tions of physiological and high-dose progestins remain unclear and are discussed later in this review.

In vitro cell culture models designed to assess the role of progestin agonists in tumor cell proliferation have generated contradictory results. Experiments can be cited in support of any argument, that progestins stim- ulate (32-34), inhibit (35-39), or have no effect (40) on growth. Explanations for the lack of a consensus are as varied as the results. Responses of cells in culture are critically dependent on the conditions in which they are grown. In a rich medium, where growth is optimized, further growth enhancement is difficult to demonstrate, while inhibitory stimuli may be exaggerated. In a de- prived medium the reverse is true, although here, key cofactors may be lacking. There is no simple solution to these inherent problems. Couple this generic uncertainty with other variables including the use of different cell lines (35, 37, 40); heterogeneity and genetic instability even within the same cell lines (41,42); a burgeoning list of factors other than estradiol and progestins that di- rectly or indirectly modulate progestin sensitivity through regulation of PR levels (43-47); the possibility

that progestin-sensitive cells can generate resistant sub- populations (48); and it is possible that no physiological consensus is likely to be forthcoming using these in vitro models. These models remain invaluable, however, for the analysis of molecular mechanisms of progestin ac- tions, and to underscore the complexities inherent in tumor cell biology.

Where does this leave us on the critical issue of the use of progestin agonists in breast cancer treatment? Interestingly, here there is more agreement, but the data contradict the conclusion that physiological levels of progestins are growth stimulatory. Especially at high doses, progestins appear to be antiproliferative in breast cancers. A comprehensive review of the clinical literature (Ref. 49 and references therein and Refs. 50-55) shows that synthetic progestins, used at pharmacological doses for first- or second-line therapy, are as effective as ta- moxifen in the treatment of advanced breast cancer. That is, approximately 30% of patients whose tumors are not screened for steroid receptors have an objective, positive response. Since in addition, progestins are well- tolerated and have a relatively low toxicity (56), their use in the treatment of advanced breast cancer is expe- riencing a resurgence (57,58). However, the mechanisms underlying the actions of intermediate and high doses of progestin agonists in breast cancer regression remain unclear, when compared to their proliferative actions at physiological doses. While some studies suggest that PR- negative tumors respond just as well as do PR-positive tumors (implying that PR are not involved), others sug- gest that methodological problems produce false PR- negative values in responders (58), and that PR are indeed required to obtain a response to progestins. An interesting study, in which tamoxifen therapy was com- pared to therapy in which tamoxifen was alternated with medroxyprogesterone acetate (MPA) in ER-positive pa- tients, showed a 40% response to tamoxifen alone us. a 62% response with the alternating treatment (53). It is postulated that when tamoxifen is cycled, its agonist properties predominate, which increases PR levels thereby enhancing the efficacy of MPA. The same ar- gument is made for enhancing the therapeutic efficacy of antiprogestins (see below). In general, tamoxifen in- ducibility of PR is considered to be a good indicator for a positive response to hormone therapy (51). Thus, while definitive data are still lacking, it is likely that positive responses to therapy with progestins in breast cancer are mediated by the PR in the tumors.

In order to consider mechanisms by which progestin agonists and antagonists regulate tumor cell growth, and how the hormone dose influences cellular responses, the following section will review recent advances in the mo- lecular biology of PR especially as they apply to proges- terone antagonists.

on February 27, 2005 edrv.endojournals.orgDownloaded from

Page 4: The Molecular Biology of RU486. Is There a Role for ... mole… · May, 1992 RU486 AND BREAST CANCER FIG. 1. The structure of progesterone, the synthetic progestin agonist R5020,

May, 1992 RU486 AND BREAST CANCER 149

IV. Molecular Mechanisms of Progesterone Antagonists

Like all steroid hormones, progesterone and the syn- thetic progestins function by binding to intranuclear proteins, the PR, the presence of which specifies a pro- gesterone target tissue and target cell. PR, when bound to ligand, control the transcription of genes, which in the breast include, among others, the genes for ER (59), for insulin receptors (37, 60), and for epidermal growth factor (EGF) receptors (61), as well as the genes for EGF and tranforming growth factor-a (62, 63). Thus, the proliferative effects of progesterone may in part be a reflection of its ability to modulate the levels of growth factors and their cognate receptors. The mechanisms by which this regulation is achieved and the structure of the PR proteins are complex and are described here with specific reference to human PR. For additional infor- mation the reader is referred to several recent reviews of the steroid receptor family of proteins (64-67).

A. Progesterone receptors

Complementary DNAs for the chicken PR were cloned independently by Jeltsch et al. (68) and Conneely et al. (69) in 1986, and for the human PR by Misrahi et al. (70) in 1987. Structure/function analyses of the encoded proteins followed quickly. PR belong to the steroid/ thyroid hormone receptor superfamily of ligand-acti- vated DNA binding proteins which are composed of a number of independently functioning domains required for nuclear localization, ligand binding, DNA binding, dimerization, and transcriptional activation. Additional sites on the receptors are responsible for their interaction with one or more accessory proteins including heat shock proteins, and several of the serine residues are covalently modified by phosphorylation. Not all of these domains have been definitively mapped, and the functional re- gions exhibit some overlap, but in general the N terminus is involved in transactivation and contains several phos- phorylation sites; the DNA-binding domain is centrally positioned and may have a weak dimerization function; and ligand-binding activity is restricted to the C termi- nus, which also contains structural features required for protein-protein interactions plus a second transcrip- tional activation domain (64-67, 71).

The single-copy human PR gene encodes at least nine messenger RNA species ranging in size from 2.5-11.4 kilobases (72,73). The nine messages direct the synthesis of at least two (74-76), and possibly three (77), structur- ally related receptor proteins. The two major protein species, the B- and A receptors, were originally described by O’Malley and co-workers (78,79) in the chick oviduct. Subsequent studies using the breast cancer cell line T47D showed that human PR also exist as two isoforms, 116 kilodalton (kDa) B receptors and N-terminally truncated

94 kDa A receptors (74-76, 80). While A receptors were originally thought to be produced by a proteolytic artifact (81), it is now clear that the amino-truncated receptors, at least in chickens and humans, are a naturally synthe- sized form (76,82-85). In human endometrial carcinoma (86) and breast cancer cell lines (74,75), the two receptor isoforms are expressed in approximately equimolar amounts. It is not known whether this quantitative re- lationship between the two isoforms is maintained in all human target tissues and tumors, and the mechanisms for their differential regulation are not known, but at least two of the nine mRNA species lack the translation initiation site for B receptors and can therefore encode only A receptors (77, 84). These messages arise by tran- scription from an internal promoter in the human PR gene (84). Five other message species can potentially encode both receptor isoforms, synthesized by alternate translation initiation from two in-frame AUG codons. In theory, use of the upstream codon generates the B recep- tors and use of the downstream codon generates the A receptors, but it is not known whether initiation at the downstream site actually occurs in intact cells (85). In- terestingly, two additional message species have been mapped that lack both the A and B translation start sites and would encode neither of the two receptor iso- forms. These messages do, however, contain downstream translation start sites that if utilized would generate a protein with a partial DNA-binding domain, and com- plete hormone binding domain and dimerization se- quences (77). This “C-receptor,” when reconstructed by site-directed mutagenesis and expressed in PR-negative cells, binds the synthetic progestin R5020 with the same affinity as the A and B receptors (K. B. Horwitz, unpub- lished). Expression of such an N-terminally truncated , protein in breast cancer cells would lead to a functionally impaired but PR-positive phenotype. The existence of such variant forms of receptors, not only for progester- one, but also for estrogen, may in part explain the recur- rent descriptions of receptor-positive, but hormone-re- sistant breast cancers.

PR are unique among steroid receptors in having two naturally occurring hormone binding forms, and this structural feature may have important functional impli- cations with respect to antagonist action (discussed be- low). Originally, the unliganded A and B isoforms were thought to be subunits of a larger heteromeric 8S receptor (87). This is not the case; instead, each receptor species forms an independent 8s heteromeric complex on su- crose gradients (88-90) by binding to as many as five non-hormone-binding proteins including two molecules of the 90 kDa heat shock protein (hsp) and one molecule of hsp 70 (91-94). In this oligomeric state, the receptor proteins are basally phosphorylated and unable to bind DNA, but they are maintained in a stable conformation

on February 27, 2005 edrv.endojournals.orgDownloaded from

Page 5: The Molecular Biology of RU486. Is There a Role for ... mole… · May, 1992 RU486 AND BREAST CANCER FIG. 1. The structure of progesterone, the synthetic progestin agonist R5020,

150 HORWITZ Vol. 13, No. 2

that allows them to bind to progestins. Hormone binding to the SS receptor form activates a rapid series of changes, the exact sequential order of which is still unknown. Within minutes, if not seconds after hormone binding, the receptors become hyperphosphorylated (95- 97), and several of the associated proteins, but not hsp 70, dissociate, leading to generation of a faster sediment- ing 4s receptor form (93, 94, 98). Two 4S receptor molecules dimerize (98) and then bind to DNA at specific transcription enhancer sites called progesterone response elements (PREs) (Refs. 64-67 and references therein). Since both homo- and heterodimers can form between the A and B isoforms, three possible classes of receptor dimers (A:A, A:B, B:B) can bind to a PRE, each having a potentially different transcription regulatory capacity (71, 98). This diversity in the repertoire of responses to one hormone sets progesterone apart from the regulatory information carried by other sex steroid hormones stud- ied to date. However, no formal proof exists that both isoforms are in the same cell.

That this molecular heterogeneity is indeed translated into functional heterogeneity was first demonstrated by the study of Tora et al. (99) that assessed the cell-specific transcriptional activation of two different target genes by the chicken A and B receptors. When a vector con- taining the PRE from the MMTV promoter linked to a reporter gene was transfected into HeLa cells, transcrip- tional activation by form B was twice as efficient as that by form A; both receptor isoforms were similarly effective at stimulating the MMTV promoter when t,hey were transfected into chick embryo fibroblast cells. In con- trast, when the PRE from the ovalbumin promoter was tested, neither receptor form was functional in HeLa cells, and only the A receptor activated transcription in chick embryo fibroblast cells. Transcription from the ovalbumin promoter can also be stimulated through an estrogen response element activated by estradiol-bound ER. When ER and one of the two PRs were cotrans- fected, transcription from the ovalbumin promoter was additive with ER and the A receptor, but the B receptor negated the transcriptional activation by ER. It seems that the N-terminal region of PR, present in B but missing in A receptors, plays a specific role in transcrip- tion that differs depending both on the gene being mod- ulated and the cell being analyzed. Thus, the A receptor can be stimulatory in a setting where the B receptor is inactive or is inhibitory. The impact of this heterogeneity on antagonist action is discussed below, but it suggests that the N terminus of the B receptor confers specific transcription regulatory functions to PR. The data also suggest that as we learn more about the tissue-specific effects of the two PR isoforms, it may be important not only to measure PR levels but also to identify the partic- ular PR isoform present in a breast tumor. While equi-

molar amounts of A and B receptors have been demon- strated in breast cancer cell lines and endometrial can- cers, it is not known whether this stoichiometry applies to normal target tissues, and to PR-positive breast can- cers. A scenario can be envisioned in which a tumor is PR-positive by ligand or immunological assays, but con- tains only B receptors that either cannot activate a key proliferation regulatory gene, or that confer agonist prop- erties to an antagonist, providing molecular explanations for a PR-positive but hormone-resistant state.

The biological significance of two or more PR isoforms extends beyond breast cancer; there is evidence that, in the chicken, the two receptor isoforms are differentially expressed developmentally and differentially regulated by hormone treatment (100, 101). The two homodimers and the heterodimer may therefore differentially regulate transcription depending on the gene, the t,arget cell, the hormonal milieu, or the developmental program, leading to a complex combinatorial response that is unique to the progestins.

B. Molecular mechanisms of progesterone antagonists

Synthetic hormone antagonists are exceptionally use- ful as both clinical and experimental tools. Their efficacy and low toxicity have made antiestrogens the drugs of choice for the treatment of advanced hormone-depend- ent breast cancer. Experimentally, these compounds have been crucial for unraveling the mechanisms of ER action. Antiprogestins have even greater potential, not only as experimental tools and as anticancer agents, but as midcycle contraceptives and implantation inhibitors and as abortifacients. By the end of the 1970s more than a decade of computer-assisted design and testing of mol- ecules likely to have antiprogestational activity had not led to any clinically or experimentally useful products. In 1981, however, Philibert et al. (102) announced the synthesis of RU38486 [17/Y-hydroxy-lip-(4dimethylam- inophenyl)-17cY-(l-propynyl)estra-4,9-diene-3-one].

RU38486 was one of a large number of ll/%substituted steroid derivatives that were synthesized by scientists at Roussel-Uclaf exploiting their extensive experience in analyzing the structure/activity relationships of com- pounds that bind both glucocorticoid receptors (GR) and PR (103). The antiglucocorticoid activity of RU38486 was demonstrated first (102), and its antiprogestin and abortifacient properties in experimental animals were reported soon thereafter (104). Testing in humans fol- lowed rapidly, and it was shown that oral administration of RU38486 to women in preliminary clinical trials in- duced interruption of the luteal phase of the menstrual cycle and termination of early pregnancy (105). The mechanisms of action of RU38486 (renamed RU486 or mifepristone) have since been the subject of intense

on February 27, 2005 edrv.endojournals.orgDownloaded from

Page 6: The Molecular Biology of RU486. Is There a Role for ... mole… · May, 1992 RU486 AND BREAST CANCER FIG. 1. The structure of progesterone, the synthetic progestin agonist R5020,

May, 1992 RU486 AND BREAST CANCER 151

investigation (106). Using RU486 as a starting-point, scientists at Schering (107) reported the synthesis of a second generation of antagonists with potent antiproges- tational activity but weaker antiglucocorticoid activity than RU486 (108). These compounds hold much promise for long-term clinical use, as preliminary studies suggest.

In theory, an antagonist can act by blocking agonist activity at any of the multiple stages between its entry into the target cell and first interaction with the recep- tors, through its role in terminating the transcriptional signal. The effects of antiprogestins at each of these stages are reviewed below.

1. Affinity for inactive PR. RU486 binds to the PR of rodents and humans with a dissociation constant (Kd) of approximately 2 x lo-’ M, roughly equivalent to the affinity of the synthetic progestin R5020 for PR (109, llO), but tighter than the affinity of the natural hormone, progesterone. Interestingly, unlike progesterone, RU486 does not bind to chicken or hamster PR (106), which suggests that there are subtle differences in the contact sites of agonists and antagonists at the hormone binding domain of the receptors. As in R5020-bound PR, unac- tivated RU486-bound PR complexes sediment at 8s on sucrose density gradients (111, 112). RU486 absorbs UV at approximately 300 nm, and this wavelength can be used to covalently cross-link the drug to PR in intact cells. This method was used to show that, like R5020, low concentrations of [3H]RU486 bind to both the A receptors and the B receptors of human PR (110). In intact breast cancer cells, R5020 and RU486 have iden- tical affinity for PR, and low concentrations (6 to 8 IIM)

of RU486 rapidly deplete more than 95% of hormone- unoccupied PR and activate them to a state in which they are tightly bound to DNA and require high concen- trations of salt for their extraction (110). Thus, it was clear soon after its development that RU486 promotes binding of PR to DNA in intact cells. Like other syn- thetic progestins, RU486 is not metabolized in intact breast cancer cells (113), allowing its activity to be pro- longed. Consequently, it chronically suppresses receptor replenishment and lowers the levels of inactive 8s PR available for ligand binding (but see Section IV.B.6 be- low). In this fashion RU486 resembles long-acting syn- thetic agonists, so that this property is unlikely to be involved in its antagonist actions.

2. PR actiuation. Initial studies by Rauch et al. (114) suggested that while RU486-bound PR complexes were activation-deficient and had lower affinity for DNA than agonist-bound complexes, these differences were modest and could not fully explain its antagonist properties. These investigators suggested that the major defect was at the transcriptional step. The receptor activation step that follows hormone binding is unclearly defined; it

appears to be related to rapid dissociation of the multiple accessory proteins, including hsp 90, from PR (92-94, 98), and to the equally rapid hormone-dependent phos- phorylation of PR (95-97). The two reactions occur in a still undetermined order. The dissociation of accessory proteins is readily measured on sucrose density gradients as a decrease in the sedimentation coefficient. Mullick and Katzenellenbogen (115) first showed that while ac- tivated R5020-receptor complexes sediment at approxi- mately 4S, the RU486-bound receptors sediment as a prominent 6s species as well as a 4s species; t.he heavier species is likely to be due to persistent receptor oligo- merization with other proteins (116,117). The suggestion that RU486 traps hetero-oligomeric 8s PR in an inactive state, thereby preventing their interaction with DNA (112), is not supported by in viuo data (71).

In addition to the loss of accessory proteins, PR acti- vation may be regulated by phosphorylation (95-97). Human PR are phosphorylated at serine residues at the time of their synthesis, and in the absence of progestins this basal phosphorylation continues for several hours. However, progestin binding to the receptors rapidly pro- motes additional serine phosphorylation concomitant with receptor activation and is accompanied by struc- tural changes that alter the mobility of human PR on electrophoretic gels (118, 119). The function of this hor- mone-dependent PR phosphorylation is still unknown, but it may play a role in the release of hsp proteins, in receptor dimerization, in DNA binding, and in regulating the functional state of PR while they are bound to the transcription complex. Since a subset of the hormone- dependent phosphorylation precedes DNA binding, and the remainder is DNA-dependent (K. B. Horwitz, un- published), it is likely that phosphorylation of human PR subserves more than one function. Of interest is the fact that compared to agonists like R5020, binding of the antagonist RU486 results in hyperphosphorylation of the PR (118, 119), so that this stage of receptor activation is, if anything, enhanced by the antagonist. It remains to be determined whether appropriate or inappropriate serine residues are the targets of this intensive RU486- dependent phosphorylation.

3. DNA binding. Whether RU486-bound PR complexes have altered DNA binding capacity when compared to agonist-bound PR complexes remains a matter of con- tention, partially due to the lack of consensus for any hormone requirement! This issue has been addressed by in vivo studies, by in vitro DNA binding studies, and most recently by cell-free transcription studies, with often contradictory conclusions. The following example illustrates the methodological conundrum for glucocor- ticoids: In consecutive articles published in Nature, Becker et al. (120) concluded that, based on in uiuo

on February 27, 2005 edrv.endojournals.orgDownloaded from

Page 7: The Molecular Biology of RU486. Is There a Role for ... mole… · May, 1992 RU486 AND BREAST CANCER FIG. 1. The structure of progesterone, the synthetic progestin agonist R5020,

152 HORWITZ Vol. 13, No. 2

genomic footprinting analysis, GR interactions with a glucocorticoid response element (GRE) require the pres- ence of hormone, and Willmann and Beato (121) con- cluded that based on in vitro DNase I footprinting, hormone-free GR bind specifically to a GRE.

There is little question that progesterone is required to activate PR in intact cells, and that this is accom- panied by increased affinity of receptors for chromatin/ DNA as assessed by the concentration of salt required to extract the receptors from cells or nuclei [for a caveat see recent studies by O’Malley and collaborators (122)]. Hormone-free receptors can be extracted with little or no salt in buffers; after hormone treatment at least 0.3 M salt is required. When PR are bound by RU486 in viuo, the receptors also require high salt concentrations for their extraction, suggesting that the antagonist in- deed promotes PR binding to DNA (110). That both B and A receptors can bind DNA when occupied by RU486 was first demonstrated by their high salt requirement for extraction after in situ photoaffinity labeling (110). These in vivo assays cannot, however, distinguish be- tween PR that are bound to nonspecific DNA sequences and PR bound to specific PREs. Becker et al. (120) used in vivo protein DNA footprinting to show that GR bind- ing to the GRE of the tyrosine amino transferase gene could be detected only after glucocorticoid treatment of hepatoma cells. Similar studies have not been done to analyze PR binding. Instead, in vitro kinetic methods have been used in an attempt to measure the affinity of receptors for DNA under varying conditions of ligand occupancy. Using crude receptors, Milgrom and his col- laborators (114) showed that RU486-PR complexes bind to nonspecific DNA with a slightly lower affinity than do R5020-PR complexes, but that a ligand was required for any PR/DNA interactions. The affinity differences between R5020- and RU486-bound PR were insufficient to explain antagonist action, but it appeared that hor- mone occupancy can indeed modulate binding, even to nonspecific DNA. It was therefore surprising that in a later study (123), this same group found that PR, whether hormone-free or bound to an agonist or antagonist, in- teracted with a similar affinity and protected the same DNA regions on three fragments of the uteroglobin gene. A major difference between the two studies was that instead of the crude PR used in the earlier study, the later study used immunopurified PR, which may have lost a component necessary to demonstrate the hormone effect. It has been suggested that this repressor is hsp 90. Alternatively, it is possible that extensive purification irreversibly modifies the structure of receptors in a man- ner that mimics the structural changes associated with hormone binding. Thus, paradoxically, excessive purifi- cation may hinder in vitro study of the. relationship’ between hormone occupancy and DNA binding,-unless

lost components are restored. This is clearly evident in more recent studies using in vitro transcription methods. O’Malley and his colleagues have developed such an assay using purified chick PR or crude human PR. The purified chick PR (124) lacks a hormonal requirement for transcription that the cruder human PR (125) retains.

In addition to issues of receptor purification, accurate kinetic data obtained from in vitro DNA binding studies require that the reactants be at equilibrium conditions, and that many factors including pH, incubation times, salt concentrations, DNA and receptor concentrations, etc., be controlled. Differences among these parameters, plus differences among the hormones and DNAs tested, may account for the lack of consensus about hormone requirements, or about the kinetics of reactions. The greatest kinetic difference between agonist and antago- nist regulation of PR binding to DNA was reported by Schauer et al. (126) who showed that in the presence of agonist and under equilibrium conditions, the on-rate of receptor binding to DNA was accelerated 2- to 5-fold, and the off-rate was accelerated lo- to 20-fold compared to ligand-free receptors. It is thus surprising that the actual DNA occupancy time of receptors was shortened by the agonist. The investigators postulated that this reduced occupancy time accelerated the DNA scanning potential of PR, but it was apparent that these results were unexpected. In the same study, the kinetic values observed with RU486 occupancy were closer to those observed with ligand-free receptors. That is, both hor- mone-free receptors and RU486-occupied receptors bind tighter to DNA than do agonist-bound receptors. This apparently tighter DNA binding of RU486-bound com- plexes compared to agonist-bound complexes (due to the slowed off-rate with RU486) had previously been ob- served by Bagchi et al. (127) using gel mobility shift assays. In contrast, ligand-free receptors fail to bind PREs when gel mobility shift methods are used (71,127), results that differ from those observed using the kinetic equilibrium assays. Finally, while confirming that ligand is required for PR binding to DNA by gel mobility shift assay, Meyer et al. (71) found no differences with respect to specificity, affinity, or stability when comparing R5020 to RU486.

Newly developed cell-free assays reaffirm the hormone and DNA binding requirements for receptor-mediated transcription (124, 125, 128). Of interest is the report by Bagchi et al. (125) that nuclear PR, salt-extracted from T47D breast cancer cells grown in the absence of hor- mone, are in a 4s form and dissociated from hsp 90. While these criteria characterize “activated” receptors, these 4s PR complexes nevertheless require hormone for DNA binding and transcriptional activation, suggesting that hsp 90 dissociation is necessary but not sufficient to generate receptors capable of binding DNA. Other

on February 27, 2005 edrv.endojournals.orgDownloaded from

Page 8: The Molecular Biology of RU486. Is There a Role for ... mole… · May, 1992 RU486 AND BREAST CANCER FIG. 1. The structure of progesterone, the synthetic progestin agonist R5020,

May, 1992 RU486 AND BREAST CANCER 153

nuclear factors or receptor modifications are likely to regulate DNA binding of the 4s complexes, with phos- phorylation being a leading candidate.

Finally, it is important to recall the danger of deriving generalized conclusions about the effects of antagonists on PR/DNA binding when only one antagonist is tested. Recently, Klein-Hitpass et al. (129) compared RU486 and three newer Schering antiprogestins with regard to the DNA binding properties of PR by gel mobility shift assay. They found that while three of the antiprogestins promoted DNA binding, one, ZK98299, did not induce binding of PR to PREs, and in competition assays, blocked the DNA binding of RU486-bound PR com- plexes. The investigators postulate that there are at least two types of antiprogestins: type I that block PR binding to PREs; and type II that promote PR binding to PREs but impede transcription, echoing a general mechanism of antagonist action proposed by Guiochon-Mantel et al. (130) based on studies of antiglucocorticoids and anti- estrogens.

To summarize, in vitro analysis of PR binding to DNA for the purposes of assessing the function of agonists and antagonists remains complex, and the data obtained may depend on variables in assay conditions that are difficult to control. Critical among them is the structural status of the PR. Warming and salt exposure are among the factors known to artifactually activate receptors in uitro; the former is often included in binding reactions, and the latter is commonly used to extract or elute receptors. When artifactual activation is minimized, it appears certain that hormone is required to generate the DNA binding form of PR. It is also clear that the most widely tested antagonist, RU486, also promotes PR binding to DNA, but that not all antiprogestins do so. However, because equally convincing studies have yielded contra- dictory conclusions, it is still not clear whether antago- nist-bound receptors bind to DNA with kinetics that differ from those of agonist-bound receptors.

4. Dimerization. The palindromic structure of PREs, the dyad symmetry shown by receptor DNA contact sites, the failure of PR to bind to half-palindromic PREs in uitro, and the leucine zipper/helix-loop-helix motif found in PR all suggest that receptors bind to DNA as dimers (64-67,71,131-133). Since human PR exist naturally as two size classes, the A and B receptors, homo- and heterodimers formed between them can be distinguished by differences in their mobility on nondenaturing elec- trophoretic gels. This interaction between monomers is activated by ligand binding and takes place in solution independent of DNA binding (98, 133). Similar to the agonists, RU486 promotes dimerization as shown by its ability to induce nuclear transfer of a nuclear localiza- tion-deficient mutant, when the mutant dimerizes to

wild-type RU486-occupied PR (133). As with R5020, RU486 and the Schering antiprogestins, ZK98434 and ZK112993, promote both homo- and heterodimerization between forms A and B, excluding the possibility that antiprogestins induce only one dimeric form of PR as a mechanism for their action (129). Complexes formed between the PRE and the PR dimers migrate faster on nondenaturing gels when the receptors are occupied by RU486 (71, 117); these differences are likely to be due to altered conformation of the receptors at the hormone binding domain generated by the antagonist, or to differ- ences in DNA bending produced by occupancy of the receptors. How this structural change is translated into an antagonist effect is still unknown. This finding, how- ever, raises an interesting question: can dimerization occur if one PR monomer is bound by agonist and another by antagonist? Since the receptor dimerization site is probably located in the hormone-binding domain, conformational differences at the dimerization domain interface of the two monomers may be incompatible. Indeed, Meyer et al. (71) found that R5020 and RU486- liganded receptors are unable to bind to PREs as hetero- dimers, thereby introducing a novel mechanism for an- tagonist action. This is confirmed by in vitro kinetic studies showing that the slope of the competition curve for RU486 is different than the slopes for progesterone and R5020 at receptor concentrations that favor dimer- ization. This is consistent with a preference of the recep- tor to have RU486 occupy both sites of a dimer, rather than having progesterone and RU486 each occupy one of the two monomers (134). On the other hand, failure to promote formation of stable receptor dimers is the postulated mechanism for antagonism of the non-DNA binding antiprogestin ZK98299 (129).

5. Transcription. The ultimate effect of antiprogestins is to modify transcription of genes regulated by progester- one through PR. While it is usually assumed that antag- onists inhibit the effects of agonists, this is not always the case since antagonists often have agonist actions as well. The paradoxical effects of steroid hormone antag- onists are well-known (see below), but the mechanisms that underlie them are poorly understood. That antago- nists regulate the transcription of specific genes, rather than having global, nonspecific effects, is certain (114), and it is also likely that whether agonist or antagonist effects predominate depends on complex interacting fac- tors that include the antagonist concentration, the spe- cific target gene, other transcriptional regulators of that gene, and the cellular environment in which that gene is active. Studies to unravel this complexity are still in the preliminary stages. While it is clear that progestins and antiprogestins regulate the expression of a variety of proteins in intact breast cells, transcriptional regulation

on February 27, 2005 edrv.endojournals.orgDownloaded from

Page 9: The Molecular Biology of RU486. Is There a Role for ... mole… · May, 1992 RU486 AND BREAST CANCER FIG. 1. The structure of progesterone, the synthetic progestin agonist R5020,

154 HORWITZ Vol. 13, No. 2

of PRE-containing genes by PR has not been analyzed in situ. Instead, two structurally different model systems have been used: the palindromic PRE of the tyrosine amino transferase (TAT) promoter (137) and the com- plex promoter of the MMTV long terminal repeat which has, in addition to one distal palindromic PRE, a cluster of three tandemly arrayed (138-141) proximal PRE half- sites.

The first model, the PRE of the TAT gene, consists of two symmetrical half-sites, each of which may be occu- pied by one molecule of the PR dimer (131, 135). This type of dimeric binding has been characterized as coop- erative for GR, i.e. that (using a DNA-binding domain fragment) occupancy of one half-site by one monomer enhances binding to the second half-site by another monomer (131). This DNA binding cooperativity is dif- ficult to reconcile with the fact that receptor monomers dimerize in solution. Assuming that only the preformed dimer binds DNA, one would have to postulate that when one monomer of the pair encounters the first PRE half- site, the other monomer is in close physical proximity and binds more efficiently to the second half-site. On the other hand, since dimers and monomers are likely to be in equilibrium in solution (141)) it may be the monomeric form that accounts for the binding cooperativity seen in the in vitro assays. The receptor concentration used in these assays controls the state of the reactants at equi- librium with more dilute receptor concentrations favor- ing a monomeric state (134); these concentrations are rarely carefully controlled. When two PREs (PREP) are tandemly linked, occupancy of one PRE by receptor dimers increases the binding affinity of additional recep- tor dimers to the second PRE by as much as loo-fold depending on the distance between the PREs (136,137). This probably involves protein-protein contacts between dimers, with looping of the intervening DNA (142). Co- operative binding of PR to PREs leads to synergistic induction of a coupled reporter gene (136, 137).

Transcriptional studies with the TAT PRE use either single (71, 135) or tandem copies (124). Assuming that R5020/RU486 PR heterodimers do not form (71), a single PRE should give clearly interpretable results when RU486 function is analyzed since both monomers would be occupied by the same hormone. But what happens in the tandem PRE, system when one of the dyads is occupied by a progestin-bound homodimer, and the other dyad is bound by an RU486-bound homodimer? The answer to this is unknown. Two studies using the TAT PREP linked to the thymidine kinase and/or ovalbumin promoter, and using human PR in cell-free transcription assays, suggest that like the agonist R5020, the antago- nist RU486 stimulates transcription (124, 125); that is, RU486 acts like a weak agonist having 25-30% the activity of R5020. The effects of combining R5020 and

RU486 were not tested in these studies, but Strahle et al. (135) showed that RU486 was able to inhibit an R5020 stimulatory effect using the same PRE-promoter con- structs in transient transfection experiments. Unfortu- nately, in this study the agonist effects of RU486 alone were not tested. Clearly, even in this simple system, a considerable amount of work remains to be done in order to understand the different agonist and antagonist ac- tions of RU486. Another observation that requires expla- nation is that the agonist actions of RU486 using the TAT PRE, can be blocked by the type I antagonist ZK98299 (129).

The second model used for transcription studies is the MMTV-long terminal repeat which may behave primar- ily as a PRE in mammary cells (143). By analogy with calculations of GR binding to the MMTV promoter (141), it is likely that one PR dimer interacts with the distal palindromic PRE. Additionally, four to six PR monomers bind the three proximal half-sites so that at full occupancy six to eight PR molecules are associated with the MMTV promoter. The distal and proximal PREs act synergistically to stimulate transcription (144, 145), and mutations in any of the four PREs reduce significantly the overall hormone inducibility of the MMTV promoter (144, 145). The complexity is obvious. How many RU486 bound PR molecules would it take to modify progesterone-stimulated transcription from this promoter? What is the functional difference between PR monomer and dimer binding sites?

Several studies using both stable and transient trans- fection models show that, when acting at the MMTV promoter, RU486 is a pure antagonist with no apparent agonist activity. In kinetic analyses Guiochon-Mantel et al. (130) calculate that when 6% of PR molecules are occupied by RU486, they can inhibit 50% of stimulatory PR activity (in this case the constitutive activity from a mutant PR). They propose that since all four PREs of the MMTV promoter are necessary for optimal tran- scriptional activity, partial occupancy of one PRE by a PR-RU486 complex is inhibitory, even if the other sites are occupied by stimulatory PR complexes. These con- clusions are consistent with other studies using MMTV (117, 143), which show that lower concentrations of RU486 than R5020 are needed to shut down the agonist effects of R5020, despite the fact that both hormones bind to human PR with similar affinities.

Finally, by comparing both the TAT PREz and the MMTV model systems in one study, Meyer et al. (71) lend support to the findings that RU486 can be a partial agonist or a pure antagonist, depending on the context of the promoter and the simplicity or complexity of the PREs. They confirmed the agonist activity of RU486- occupied human PR when they are bound to PRE, in the context of a thymidine kinase promoter and their

on February 27, 2005 edrv.endojournals.orgDownloaded from

Page 10: The Molecular Biology of RU486. Is There a Role for ... mole… · May, 1992 RU486 AND BREAST CANCER FIG. 1. The structure of progesterone, the synthetic progestin agonist R5020,

May, 1992 RU486 AND BREAST CANCER 155

lack of agonist activity when bound to the MMTV pro- moter. In addition, these investigators tested the func- tional capacity of the B and A isoforms of human PR separately with totally unexpected findings: only the B form of the receptors exhibited the agonist properties when bound by RU486. Since the A and B isoforms also had quite different inductive capacities when bound to R5020 (the A receptors had only 10% the agonist activity of the B receptors on the MMTV promoter but 60% of B on the TAT PREJ, the combinatorial possibilities for modulating transcriptional responsiveness (by type of ligand, by receptor isoform, by promoter, or by cell type) are staggering. Tora et al. (99) showed that the cell type into which receptors are transfected and tested strongly influences transcriptional capacity. Note also that the transcriptional actions of an AB heterodimer, likely to be the most abundant naturally occurring human PR form, remain unknown.

6. PR “processing” or down-regulation. A persistent tran- scriptional signal by hormone-receptor complexes cannot be tolerated by the cell, so mechanisms must exist to terminate it. Little is known about this final stage in receptor action. Receptor recycling and covalent modi- fications are among the mechanisms that have been proposed, but down-regulation may be the most common feature (146-153) associated with receptor inactivation. After agonist treatment, both DNA-bound ER and PR are “processed” or down-regulated, a stage that begins approximately 1 h after DNA binding and is complete 6- 12 h later. Since this loss of DNA-bound receptors is not immediately accompanied by replenishment of unacti- vated receptors, total cellular receptor levels fall by 80- 90%. The down-regulated receptors are restored only many hours (>24) later, presumably after synthesis of new receptors. The receptor replenishment time differs depending on the persistence of agonists within the cell (147-153).

While down-regulation has been documented for ER and PR bound to agonists, the scenario is entirely differ- ent for receptors bound to antagonists. Antagonist- bound receptor/DNA complexes down-regulate only par- tially or not at all; instead nuclear receptor levels remain elevated for hours, even days, after hormone treatment. In the specific case of RU486 in breast cancer cells, PR remain complexed to DNA for at least 48 h (72,97,118). The sustained DNA occupancy of RU486-bound PR may, at least in part, explain its antagonist actions. Is it possible that agonist effects of RU486 result from the initial contact between RU486-bound PR and DNA, and that antagonist effects follow from the subsequent pro- longed DNA occupancy? Recall also that RU486-PR complexes bound to DNA are hyperphosphorylated com- pared to their R5020 counterparts (97, 118). Nothing is

known about the role of PR phosphorylation in initiating or sustaining the transcriptional signal, or in regulating receptor down-regulation. It is possible that PR struc- tural changes associated with hyperphosphorylation pre- vent the down-regulatory machinery from being acti- vated or from functioning.

In the case of ER, it is known that this structural block to down-regulation imparted by antagonist occu- pancy of the receptors is rapidly reversible. In intact breast cancer cells, ER occupied by the antiestrogen nafoxidine bind persistently to DNA and are not down- regulated for at least 24 h. If, at 24 h, cells are switched to medium containing estradiol, the excess agonist ex- changes with the antagonist on the DNA-bound recep- tors, and down-regulation is initiated and proceeds with a normal time course. The estradiol-substituted ER, which were transcriptionally silent when bound to an- tagonist, are rapidly reactivated; in fact, a superinduction of the transcriptional signal is seen (154). This is an interesting observation for which no molecular explana- tions are presently forthcoming. These studies also ad- dress persistent questions regarding the length of time that a ligand must occupy receptors after they bind to DNA. Is the function of the ligand then terminated and is it dispensible? The antagonist to agonist switching study suggests either that ligand occupancy persists or that conformational changes are permanently imprinted on the receptor by ligand binding and are preserved when the ligand dissociates. The imprints imparted by agonists and antagonists differ but are exchangeable.

V. Progesterone Antagonists and the Treatment of Breast Cancer

Because progestin antagonists are relatively new com- pounds, and because of the political controversy that surrounds them, their promise and use in the treatment of breast cancer are just beginning to be evaluated. What is the rationale for their use? What is the explanation for the paradox that both antiprogestins and high-dose progestin agonists inhibit growth of breast cancers?

A. Human breast cancer cell lines

Two PR-positive human breast cancer cell lines that are phenotypically different have served as the major models for studies of growth regulation by antiprogestins. The MCF-7 cell line is classically estrogen-responsive cells; the cells are ER-positive but have only low PR levels unless these are induced by estradiol (153). The T47D cell line (155) is more complex; it is genetically unstable (41, 42) and differs phenotypically among (34, 35, 37) and within laboratories, which leads to reported differences in response to hormone treatment. One major T47D subline, clone 11, is ER-positive and PR-positive,

on February 27, 2005 edrv.endojournals.orgDownloaded from

Page 11: The Molecular Biology of RU486. Is There a Role for ... mole… · May, 1992 RU486 AND BREAST CANCER FIG. 1. The structure of progesterone, the synthetic progestin agonist R5020,

156 HORWITZ Vol. 13, No. 2

and the cells respond to estradiol treatment by prolifer- ating, a response that is tamoxifen-inhibitable (35). An- other subline, T47D,,, is estrogen-resistant, and cell growth is neither accelerated by estradiol nor inhibited by tamoxifen (147). The response to antiprogestins is generally similar among these cell lines, but interpreta- tion of the results differs. In general, RU486 inhibits the growth of both T47D cells and MCF-7 cells (110, 156- 161). The antiproliferative effects are evident at low doses, and their magnitude correlates loosely with PR levels; T47D > estrogen primed MCF-7 > unprimed MCF-7 (156). That the antiproliferative effects are me- diated by PR is also shown by the fact that in T47D cells growth inhibition is confined to progestins; other steroid hormones are ineffective (156, 158). Moreover, the fact that RU486 is not antiproliferative in PR-negative breast cancer cell lines also argues for a receptor-mediated mechanism of action (156).

The antiproliferative actions of RU486 in these models of breast cancer would support a logical treatment strat- egy if it were not for one disturbing fact: progestin agonists, including R5020, also inhibit their growth. This effect of R5020 is seen even at low doses in the T47D,, cells (110) but requires higher doses in the clone 11 cells, in which, interestingly, low doses actually protect the cells from the antiproliferative effects of RU486 (156). Furthermore, in the estrogen-responsive clone 11 cells, R5020 inhibits only the estradiol-stimulated growth frac- tion where it is cytostatic, while RU486 reduces cell numbers below the estrogen-untreated baselines, sug- gesting that the latter has a more profound cytotoxic effect (160, 161). Thus the antiproliferative mechanisms for progestin agonists like R5020 and antagonists like RU486 may be fundamentally different. R5020 appears to have dual proliferative/antiproliferative effects de- pending on the dose that is tested, and the antiprolifer- ative doses produce growth stasis. By contrast, RU486 is more purely antiproliferative even at low doses, and it can produce growth regression. Indeed, ultrastructural studies show effects of RU486 on cell and chromatin condensation and pyknosis consistent with the induction of cell death by apoptosis (161). This cytotoxic effect is prevented by low doses of R5020. To explain these re- sults, Bardon et al. (161) propose that there are three different mechanisms by which progestins inhibit growth of breast tumor cells: 1) “PR-mediated cytotoxic” mech- anisms that apply to antagonists like RU486. These are observed only in PR-positive cells using “physiological” hormone doses and are preventable by receptor occu- pancy with an agonist. This cytotoxicity is characterized by ultrastructural evidence of cell death. 2) “PR-me- diated cytostatic” effects are produced by physiological doses of antagonists or agonists and are characterized by inhibition of the growth-stimulatory actions of unrelated

(nonprogestin) growth factors including estradiol. 3) “Nonspecific cytotoxic” effects are not receptor-me- diated and are seen with most steroid hormones at high doses. The molecular explanations that underlie these three mechanisms are unknown, but they serve as an important departure for further research. For example, the prolonged DNA occupancy time of RU486-bound PR compared to R5020-bound PR may account for cytotoxic vs. cytostatic effects (97). As described above, variables like the gene or cell being tested (99) and regulation by either the A or B receptor (71) may dictate an agonist or antagonist response. Alternatively, it has been suggested that R5020 is inhibitory because it is “antiestrogenic” (35) while RU486 is inhibitory through a direct antipro- liferative effect involving PR (156). While these expla- nations may begin to address the paradox that allows both R5020 and RU486 to be growth inhibitory in the appropriate physiological setting, it is clear that a con- siderable amount of research remains to be done. Prelim- inary analyses of cell cycle parameters show no definitive differences between agonists and antagonists. Both ap- pear to inhibit growth by significantly decreasing the proportion of cells in the S-phase of the cell cycle; cells accumulate in Go/G1 possibly due to an increase in the G1 transit time (158, 162).

While the majority of studies using cell culture models ascribe growth- inhibitory properties to progestins and to antiprogestins through direct effects involving PR, contradictory results have also been reported. Given the fact that, at physiological levels, progesterone is believed to be mitogenic in the normal breast (see above), it is not entirely surprising that Hissom and Moore (33) consistently report proliferative effects of R5020 at all doses in T47D cells. It is surprising, however, that RU486 also stimulates growth in these cells (163). The expla- nation for this discrepancy is unknown. However, it is now clear that T47D cells are exceptionally unstable; during prolonged time in culture, subpopulations can develop that are phenotypically different from the paren- tal stocks, and some of these subpopulations may respond to hormones in a manner that differs markedly from the expected response. For example, some sublines or sub- populations of T47D cells respond to high doses of ta- moxifen by growth stimulation (48, 164). In the case of tamoxifen, these aberrant responses may be mediated by the presence of mutant or variant ER (164). By analogy, it is possible that the T47D cells of Moore and his colleagues have arisen from a subpopulation harboring a mutant PR. This scenario would be very interesting, but the PR of these cells have not been analyzed in detail. Less interesting trivial explanations for discrepancies among laboratories studying growth regulation by pro- gestins in cell culture were discussed above (see also Ref. 1).

on February 27, 2005 edrv.endojournals.orgDownloaded from

Page 12: The Molecular Biology of RU486. Is There a Role for ... mole… · May, 1992 RU486 AND BREAST CANCER FIG. 1. The structure of progesterone, the synthetic progestin agonist R5020,

May, 1992 RU486 AND BREAST CANCER 157

B. Animal models of mammary cancer

The antiproliferative properties of progesterone antag- onists are well documented in animal models of hormone- dependent mammary cancer. These include rats bearing DMBA-induced or nitrosomethylurea (NMU)-induced tumors, and mice bearing the transplantable MXT tumor line. Growth of these tumors is inhibited by ovariectomy and maintained by physiological doses of estrogens (165 170). While treatment of rats with progestins at the time of DMBA administration accelerates tumor formation (21-23), prophylactic treatment of rats with RU486 at the time of DMBA administration delays the initial appearance of tumors from an average of 39 days, to an average of 81 days (157). The reversal by progesterone of the inhibition of tumor induction produced by tamox- ifen can in turn be blocked by RU486 (30). These impli- cate a PR-mediated mechanism. Treatment of estab- lished tumors with RU486 for 3 weeks prevents their further enlargement, but tumor remission is not observed (165). In contrast to the stasis seen with RU486, ovar- iectomy leads to a decrease in tumor size (169), which in MXT tumors is accompanied by necrosis and cytolysis of the tumor cells (168). While ovariectomy-induced tumor regression is known to be accompanied by exten- sive loss of PR (29), loss of tumor PR was also seen with RU486 treatment (165). Since in the latter, the PR assay was not performed under exchange conditions or by immunological methods, the validity of this decrease requires reexamination in light of other studies that show persistently high levels of PR in RU486-treated human breast cancer cells (97). Since adrenal weights are un- changed by RU486, participation of the antiglucocorti- coid effects in the antitumor activity is considered to be unlikely (169). This is supported by studies in human breast cancer cell lines, where the inhibitory effects of RU486 cannot be rescued by dexamethasone (156). An- tigonadotrophic effects have also been excluded as a mechanism (169).

In established DMBA tumors inhibition of growth by tamoxifen resembles the inhibition seen with RU486. The two hormones have equal growth-inhibitory effects when each is used alone (165). When the two drugs are combined, however, the inhibitory effects are additive, and tumor remission similar to that induced by ovariec- tomy is observed (166). This effect of combined treat- ment with an antiprogestin and antiestrogen is extremely exciting and has considerable therapeutic promise. The mechanisms underlying these effects remain unclear, but several proposals have surfaced. First, tamoxifen can have agonist actions among which is the induction of PR (153). A tumor with increased, or restored, PR may have greater or more sustained sensitivity to RU486. This hypothesis could be tested by the use of an antiestrogen

having no agonist activity. Second, among the physiolog- ical effects seen in RU486-treated intact female rats are increased plasma levels of LH, PRL, estradiol, and pro- gesterone, as well as the persistence of numerous and actively secretory corpora lutea associated with hyper- trophic pituitaries (165, 166, 168, 169). It has therefore been proposed that the efficacy of simultaneous tamox- ifen results from its ability to counteract the proliferative effects of the high estrogen levels.

Several newer antiprogestins, ORG31710 and ORG31806 (166), and ZK98299 and ZK112993, have equal or greater antiproliferative actions than RU486 (166,168). In the hormone-dependent MXT-transplant- able tumor model, treatment with ZK98299 or RU486 starting 1 day after transplantation led to an almost complete inhibition of tumor growth. Their effect on established tumors was equivalent to that of ovariectomy (168, 169). In this model, the potent antiproliferative actions of the antiprogestins completely counteracted the growth-stimulatory actions of estradiol, or of approx- imately equimolar doses of MPA, but at higher MPA doses, the agonist actions of the progestin prevailed (170). It appears that antiprogestins inhibit growth by direct antagonism of progesterone action at the tumor, probably mediated by PR. This conclusion is bolstered by the fact that the hormone-independent MXT tumor is antiprogestin-resistant (168).

In DMBA-induced tumors, ZK98299 was more potent than an equal concentration of RU486 (168). It produced tumor regression analogous to that of ovariectomy, rather than the tumor stasis observed with RU486. A similar trend was observed with NMU-induced rat mam- mary tumors (168-170). However, lack of comparative metabolic and pharmacokinetic data on the two antipro- gestins in rats and mice makes these quantitative differ- ences uninterpretable at present. Also of interest is the finding that strong antitumor activity was noted at 20% of the doses needed to obtain abortifacient actions in these rodent systems. This is important because by use of lower doses of antiprogestins, their antiglucocorticoid effects may be mitigated. After treatment with the anti- progestins, the morphology of the hormone-dependent MXT and DMBA tumors showed signs of differentiation of the mitotically active polygonal epithelial tumor cells toward the nonproliferating glandular secretory pattern with formation of acini and evidence of secretory activity (168). Based on this it is suggested that the antiprolif- erative efficacy of the antiprogestins is related to their ability to induce terminal differentiation. Recall that they block tumor cells in Go/G1 (158, 162). Note that an antiproliferative mechanism based on induction of ter- minal differentiation is fundamentally different from a mechanism involving tumor cell death. Tumor cell deg- radation and cytolysis were features of ovariectomy-

on February 27, 2005 edrv.endojournals.orgDownloaded from

Page 13: The Molecular Biology of RU486. Is There a Role for ... mole… · May, 1992 RU486 AND BREAST CANCER FIG. 1. The structure of progesterone, the synthetic progestin agonist R5020,

158 HORWITZ Vol. 13. No. 2

induced regression (168). The mechanisms underlying the antitumor effects of antiprogestational agents require further study, especially in human tissues and cells.

C. Human clinical trials

The enormous promise of progestin antagonists in the treatment of breast cancer remains largely unexplored in clinical practice. Only two small clinical trials using RU486 have been reported, both from European labora- tories. The first involved a series from France (171) of 22 oophorectomized or postmenopausal patients in whom chemotherapy, radiotherapy, or tamoxifen and other hor- monal therapy had already been used. RU486 at 200 mg/ day led to partial regression or stabilization of lesions in 12 of 22 (53%) women after 4 to 6 weeks of treatment. The response rate at 3 months had dropped to 18%. It is important to note that for ethical reasons, this untried therapy was used only in patients with advanced breast cancers in whom other treatment modalities had already failed. PR levels were not measured in all patients, but of the responders, four of four were PR-positive, while of the nonresponders zero of four were PR-positive. In general, RU486 was well-tolerated in long-term treat- ment with few symptoms of adrenal dysfunction, but plasma cortisol levels were elevated. Of interest was the fact that a strong analgesic effect was observed in most of the patients with bone metastases.

The second trial, from the Netherlands (166, 170), involved 11 postmenopausal patients with metastatic breast cancer who were treated with 200-400 mg RU486 for 3-34 weeks as second-line therapy after first-line treatment with tamoxifen, irrespective of the response to tamoxifen. Six of 11 patients had a short-term (3 to 8 months) stabilization of disease, and one had an objective response lasting 5 months after RU486 treatment. Again, response was associated with presence of PR in the tumors. In this study, which involved prolonged use of RU486, two patients had undesirable side-effects asso- ciated with the antiglucocorticoid actions of the drug. Three days of treatment with dexamethasone reversed these symptoms after RU486 was stopped. As in the animal studies, plasma estradiol levels increased despite the fact that these women were postmenopausal, and it is suggested that the simultaneous administration of tamoxifen might be beneficial because of its ability to blockade tumor ER. Alternatively, symptoms of adrenal hypersecretion and elevated plasma estradiol levels might be reduced with concurrent aminoglutethimide or aromatase inhibitors. By modification of the dose and time of RU486 administration, its antiglucocorticoid ef- fects might be further minimized (172), although main- tenance of high sustained blood levels of the drug is likely to be important.

VI. Progestin Resistance

The emergence of hormone-resistant cells eventually reduces the effectiveness of all therapies in advanced breast cancer, and progestin agonists or antagonists are unlikely to be exceptions. This is essentially an unex- plored field. Unlike the case for other members of the steroid receptor family, no examples of natural PR mu- tants have yet been reported. It is possible that unlike mutations in androgen receptors, systemic mutations in PR are incompatible with life. It is likely, however, that acquired mutations can develop in tumors as one mech- anism for the development of resistance, and that a systematic search would demonstrate them. In a similar manner, resistance to tamoxifen appears to be due in part to the development of mutant ER (164). To address other possible mechanisms of progestin resistance Mur- phy et al. (173) generated a subline of T47D cells that are resistant to the growth-inhibitory effects of proges- tins. This was done by sequential selection in medium containing 1 pM MPA. The cells remained PR-positive, but receptor levels were halved. Transforming growth factor-a and EGF receptor mRNA levels were both in- creased. The investigators suggest that increased growth factor expression and action and decreased PR levels may be involved in the development of progestin resist- ance. Finally, it is likely that extensive heterogeneity in PR content exists within cell subpopulations of tumors that are PR-positive based on analyses of solid tumors (174) and of human breast cancer cell lines (48). Factors or treatments that lead to the selection and expansion of PR-poor or PR-negative populations would, in the long run, produce progestin resistance.

VII. Summary and Future Prospects

The foregoing suggests that progesterone antagonists could have an important place in the routine manage- ment of hormone-dependent breast cancers. Our knowl- edge of the actions of these compounds is rudimentary, however. The following points provide an outline for future directions:

1. If endogenous progesterone has the mitogenic ac- tions in normal breast epithelia that the current data would indicate, then it is likely that physiological pro- gesterone is also a mitogen in breast cancers. Blockade of endogenous progesterone with antiprogestins, espe- cially in premenopausal women, would seem to be an important therapeutic goal. However, nothing is known about the pattern of mitosis in breast cancer cells during the normal menstrual cycle. Obtaining such data is im- portant, and it should be possible to analyze the mitotic patterns of tumors taken from cycling patients. The problem, of course, is the difference in mitotic indices among tumors. Ideally, each tumor should serve as its

on February 27, 2005 edrv.endojournals.orgDownloaded from

Page 14: The Molecular Biology of RU486. Is There a Role for ... mole… · May, 1992 RU486 AND BREAST CANCER FIG. 1. The structure of progesterone, the synthetic progestin agonist R5020,

May, 1992 RU486 AND BREAST CANCER 159

own control, with multiple samples analyzed for prolif- erative activity at different times of the cycle. This approach is fraught with ethical problems. However, it might be possible to obtain a fine needle aspirate of a tumor for initial mitotic analysis, then to acutely treat the patient with RU486 before the tumor is removed 24 h later for reanalysis. Additionally, it is important to know whether RU486 is cytostatic or cytolytic in human breast cancers. Electron microscopy of tumors taken from patients entered into trials may provide answers to this. However, while it is always preferable to ask biolog- ical questions using clinical tissues, much of the work on the mechanisms of the mitogenic actions of progestins and progestin antagonists will require well-controlled studies using organ-cultured human breast tumors, hu- man breast cancer cell lines, and human tumors im- planted into nude mice.

2. There are sufficient theoretical and preclinical data to justify large-scale clinical trials. Trials must include accurate measurements of ER and PR in tumors, and analysis of pre- and posttreatment levels of endogenous hormones to monitor the status of the pituitary-adrenal- ovarian axis. Progestin antagonists may be useful both for adjuvant endocrine therapy when used either alone or in combination with tamoxifen, and for therapy of locally advanced or metastatic cancers again used either alone or in combination with tamoxifen. The usual issues of drug doses and of metabolism and other pharmacoki- netic parameters must be addressed. Schedules in which the antiprogestins and antiestrogens are combined or alternated must be tested. Is an antiestrogen with agonist properties preferable because it induces PR or is a pure antiestrogen preferable because it blocks the actions of elevated circulating estrogens? Is the answer different in premenopausal vs. postmenopausal women?

3. The antiglucocorticoid side effects of RU486 remain a major impediment to its long-term use. However, as the newer Schering antiprogestins show, it appears to be possible to design molecules with maximal antiprogestin activity and minimal antiglucocorticoid activity. Further, even RU486 may be used for long-term treatment if it is combined with drugs that block adrenal steroidogenesis or prevent peripheral aromatization of adrenal steroids to estrogens. Additionally, since the antiglucocorticoid effects of RU486 are apparently tolerable in the short- term (i.e. 2 to 3 months), perhaps it is reasonable to ask whether alternating RU486 with tamoxifen maximizes its antiprogestin effects while it minimizes its antiglu- cocorticoid ones.

4. Basic tumor biological and molecular research must continue in order for us to understand the precise molec- ular targets and mechanisms of antagonist action. If different antagonists target different molecular sites, which antagonist would be best for clinical use? What

are the genes in breast cells that are regulated by proges- tins, whose inhibition is associated with lowered cell proliferation? What is the underlying cause of the tissue- specific differences in progestin action at the breast and uterus? What is the molecular explanation for the dual agonist/antagonist effects seen on only some promoters, with only one or the other PR isoform, and in only some cells when using RU486? Can pure progesterone antag- onists devoid of antiglucocorticoid activity be synthe- sized? Are there mutant PR in breast cancers?

Where do we begin? Ensuring that scientists and cli- nicians have access to antiprogestins, unencumbered by the Byzantine bureaucratic obstacles and the “antago- nistic” political climate currently encountered in the United States, is a good place to start.

Acknowledgments

Dedicated to the memory of William L. McGuire.

I thank the past and present members of my laboratory who gener- ated the data cited here, Linda Trefry for her expert secretarial work,

and my colleague, William Wood, for reviewing the manuscript.

1.

2.

3.

4.

5.

6.

7.

8.

9.

10.

11.

12.

13.

14.

References

Clarke CL, Sutherland RL 1990 Progestin regulation of cellular proliferation. Endocr Rev 11:266 Going JJ, Anderson TJ, Battersby S, MacIntyre CCA 1988 Pro- liferative and secretory activity in human breast during natural and artificial menstrual cycles. Am J Path01 130:193 Anderson TJ, Battersby S, King RJB, McPherson K, Going JJ 1989 Oral contraceptive use influences resting breast prolifera- tion. Hum Path01 20:1139 Anderson TJ, Howell A, King RJB 1987 Comment on progester- one effects in breast tissue. Breast Cancer Res Treat 10:65 Berliner JA, Gerschenson LE 1976 Sex steroid induced morpho- logical changes in primary uterine cell cultures. J Steroid Biochem 7:153 Henderson BE, Ross R, Bernstein L 1988 Estrogens as a cause of human cancer. The Richard and Hinda Rosenthal Foundation Award Lecture. Cancer Res 48246 Haslam SZ 1988 Progesterone effects on deoxyribonucleic acid synthesis in normal mouse mammary glands. Endocrinology 122:464 Imagawa W, Tomooka Y, Hamamoto S, Nandi S 1985 Stimulation of mammary epithelial cell growth in uitro: interaction of epider- ma1 growth factor and mammogenic hormones. Endocrinology 116:1514 Bresciani F 1971 Ovarian steroid control of cell proliferation in the mammary gland and cancer. In: Basic Actions of Sex Steroids on Target Organs. Karger Publishing Company, Basel, p 130 Michna H, Nishino Y, Schneider MR, Louton T, El Etreby MF 1991 A bioassay for the evaluation of antiproliferative potencies of progesterone antagonists. J Steroid Biochem Mol Biol38:359 McManus MJ, Welsch CW 1984 The effect of estrogen, proges- terone, thyroxine, and human placental lactogen on DNA synthe- sis of human breast ductal epithelium maintained in athymic nude mice. Cancer 54:1920 Mauvais-Jarvis P, Kuttenn F, Gompel A 1986 Antiestrogen action of progesterone in breast tissue. Breast Cancer Res Treat 8179 Gompel A, Malet C, Spritzer P, Lalardrie J-P, Kuttenn F, Mau- vais-Jarvis P 1986 Progestin effect on cell proliferation and 17/3- hydroxysteroid dehydrogenase activity in normal human breast cells in culture. J Clin Endocrinol Metab 63:1174 McCarty Jr KS 1989 Proliferative stimuli in the normal breast: estrogens or progestins? Human Path01 20:1137

on February 27, 2005 edrv.endojournals.orgDownloaded from

Page 15: The Molecular Biology of RU486. Is There a Role for ... mole… · May, 1992 RU486 AND BREAST CANCER FIG. 1. The structure of progesterone, the synthetic progestin agonist R5020,

160 HORWITZ Vol. 13, No. 2

15. Frank DW, Kirton KT, Murchism TE, Quinlan WJ, Coleman TJ, Gilbertson TJ, Feenstra ES, Kimball FA 1979 Mammary tumors and serum hormones in the bitch treated with medroxy- progesterone acetate and progesterone for four years. Fertil Steril 31:340

16. Lanari C, Molinolo AA, Dosne PC 1986 Induction of mammary adenocarcinomas by medroxyprogesterone acetate in BALB/c female mice. Cancer Lett 33:215

17. Nagasawa H, Aoki M, Sakagami N, Ishida M 1988 Medroxy- progesterone acetate enhances spontaneous mammary tumorigen- esis and uterine adenomyosis in mice. Breast Cancer Res Treat 12:59

18. Lanari C, Kordon E, Molinolo A, Dosne PC, Charreau EH 1989 Mammary adenocarcinomas induced by medroxyprogesterone acetate: hormone dependence and EGF receptors of BALB/c in uiuo sublines. Int J Cancer 43:345

19. Kordon E, Lanari C, Meiss R, Elizalde P, Charreau E, Pasqualini CD 1990 Hormone dependence of a mouse mammary tumor line induced in uiuo by medroxyprogesterone acetate. Breast Cancer Res Treat 17:33

20. Huggins C, Moon RC, Morii S 1962 Extinction of experimental mammary cancer. I. Estradiol-17P and progesterone. Proc Nat1 Acad Sci USA 48:379

21. Huggins C, Yang NC 1962 Induction and extinction of mammary cancer. Science 137:257

22. Huggins C 1965 Two principles in endocrine therapy of cancers: hormone deprival and-hormone interference. Cancer-Res 25:1163

23. Welsch CW 1985 Host factors affecting the growth of carcinoeen- induced rat mammary carcinomas: a revview and tribute to Charles Brenton Huggins. Cancer Res 45:3415

24. Russo IH, Gimotty P, Dupuis M, Russo J 1989 Effect of medrox- yprogesterone acetate on the response of the rat mammary gland to carcinogenesis. Br J Cancer 59:210

25. Meirik 0, Lund E, Hans-Olov A, Bergstrom R, Christoffersen T, Bergsjo P 1986 Oral contraceptive use and breast cancer in young women. Lancet 2:650

26. Hulka BS 1990 Hormone-replacement therapy and the risk of breast cancer. CA 40:289

27. Bergkvist L, Adami HO, Persson I, Hoover R, Schairer C 1989 The risk of breast cancer after estrogen and estrogen-progestin replacement. N Engl J Med 321:293

28. Ewertz M 1988 Influence of non-contraceptive exogenous and endogenous sex hormones on breast cancer risk in Denmark. Int J Cancer 42:832

29. Horwitz KB, McGuire WL 1977 Progesterone and progesterone receptors in experimental breast cancer. Cancer Res 37:1733

30. Robinson SP, Jordan VC 1987 Reversal of the antitumor effects of tamoxifen by progesterone in the 7,12-dimethylbenzanthra- cene-induced rat mammary carcinoma model. Cancer Res 47:5386

31. Danguy A, Legros N, Develeeschouwer N, Heuson-Stennon JA, Heuson JC 1980 Effects of medroxyprogesterone acetate (MPA) on growth of DMBA-induced rat mammary tumors: histopatho- logical and endocrine studies. In: Iacobelli S, Di Marco A (eds) Role of Medroxyprogesterone in Endocrine-Related Tumors. Raven Press, New York, p 21

32. Dao TL, Sinha DK, Nemoto T, Pate1 J 1982 Effect of estrogen and progesterone on cellular replication of human breast tumors. Cancer Res 42:359

33. Hissom JR, Moore MR 1987 Progestin effects on growth in the human breast cancer cell line T47D-possible therapeutic impli- cations. Biochem Biophys Res Commun 145:706

34. Hissom JR, Bowden RT, Moore MR 1989 Effects of progestins, estrogens, and antihormones on growth and lactate dehydroge- nase in the human breast cancer cell line T47D. Endocrinology 125:418

35. Vignon F, Bardon S, Chalbos D, Rochefort H 1983 Antiestrogenic effect of R5020, a synthetic progestin in human breast cancer cells in culture. J Clin Endocrinol Metab 56:1124

36. Chalbos D, Rochefort H 1984 Dual effects of the progestin R5020 on proteins released by the T47D human breast cancer cells. J Biol Chem 259:1231

37. Horwitz KB, Freidenberg GR 1985 Growth inhibition and increase

38.

39.

40.

41.

42.

43.

44.

45.

46.

47.

48.

49.

50.

51.

52.

53.

54.

55.

of insulin receptors in antiestrogen-resistant T47D,, human breast cancer cells by progestins: implications for endocrine ther- apies. Cancer Res 45:167 Purohit A, Lai LC, Singh A, Coldham NG, Ghilchik MW, Reed MJ, James VHT 1989 The effect of medroxyprogesterone acetate on aromatase and DNA polymerase (Y activities in breast tumours. J Steroid Biochem 34:443 Poulin R, Baker D, Poirier D, Labrie F 1990 Multiple actions of synthetic ‘progestins’ on the growth of ZR-75-1 human breast cancer cells: an in vitro model for the simultaneous assay of androgen, progestin, estrogen, and glucocorticoid agonistic and antagonistic activities of steroids. Breast Cancer Res Treat 17:197 Lippman M, Bolan G, Huff K 1976 The effects of glucocorticoids and progesterone on hormone-responsive human breast cancer in long-term tissue culture. Cancer Res 364602 Reddel RR, Alexander IE, Koga M, Shine J, Sutherland RL 1988 Genetic instability and the development of steroid hormone in- sensitivity in cultured T47D human breast cancer cells. Cancer Res 48:4340 Graham II ML, Dalquist KE, Horwitz KB 1989 Simultaneous measurement of progesterone receptors and DNA indices bv flow cytometry: analysis of breast cancer cell mixtures and genetic instabilitv of the T47D line. Cancer Res 49:3943 Sarup JC; Rao KVS, Fox CF 1988 Decreased progesterone binding and attenuated progesterone action in cultured human breast carcinoma cells treated with epidermal growth factor. Cancer Res 485071 Katzenellenbogen BS, Norman MJ 1990 Multihormonal regula- tion of the progesterone receptor in MCF-7 human breast cancer cells: interrelationships among insulin, insulin-like growth factor- I, serum, and estrogen. Endocrinology 126:891 Clarke CL, Graham J, Roman SD, Sutherland RL 1991 Direct transcriptional regulation of the progesterone receptor by retinoic acid diminishes progestin responsiveness in the breast cancer cell line T-47D. J Biol Chem 266:18969 Aronica SM, Katzenellenbogen BS 1991 Progesterone receptor regulation in uterine cells: stimulation by estrogen, cyclic adeno- sine 3’,5’-monophosphate, and insulin-like growth factor I and suppression by antiestrogens and protein kinase inhibitors. En- docrinology 128:2045 Eckert RL, Katzenellenbogen BS 1983 Modulation of progestin binding activity in cultured human breast carcinoma cells: the effect of serum type and concentration. J Recept Res 3599 Graham II ML, Smith JA, Jewett PB, Horwitz KB 1992 Hetero- geneity of progesterone receptor content and remodeling by ta- moxifen characterize subpopulations of cultured human breast cancer cells: analysis by quantitative dual parameter flow cytom- etry. Cancer Res, 52:593 Sedlacek SM, Horwitz KB 1984 The role of progestins and pro- gesterone receptors in the treatment of breast cancer. Steroids 44:467 Canobbio L, Galligioni E, Gasparini G, Fassio T, Crivellari D, Villalta D, Santini G, Monfardini S, Boccardo F 1987 Alternatine tamoxifen and medroxyprogesterone acetate in postmenopausal advanced breast cancer patients short and long term endocrine effects. Breast Cancer Res Treat lo:201 Howell A, Harland RNL, Barnes DM, Baildam AD, Wilkinson MJS, Hayward E, Swindell R, Sellwood RA 1987 Endocrine therapy for advanced carcinoma of the breast: relationship be- tween the effect of tamoxifen upon concentrations of progesterone receptor and subsequent response to treatment. Cancer Res 47:300 Lundgren S, Gundersen S, Klepp R, Lonning PE, Lund E, Kvinnsland S 1989 Megestrol acetate uersus aminoelutethimide for metastatic breast cancer. Breast Cancer Res Treat 14:201 Gundersen S, Kvinnsland S, Lundgren S, Klepp 0, Lund E, Bormer 0, Host H 1990 Cyclical use of tamoxifen and high-dose medroxyprogesterone acetate in advanced estrogen receptor pos- itive breast cancer. Breast Cancer Res Treat 17:45 Parnes HL. Abrams JS. Tchekmedvian NS. Tait N. Aisner J 1991 A phase I/II study of high-dose megestrol acetate in the treatment of metastatic breast cancer. Breast Cancer Res Treat 18:171 Pronzato P, Brema F, Amoroso D, Bertelli G, Conte PF, Martini

on February 27, 2005 edrv.endojournals.orgDownloaded from

Page 16: The Molecular Biology of RU486. Is There a Role for ... mole… · May, 1992 RU486 AND BREAST CANCER FIG. 1. The structure of progesterone, the synthetic progestin agonist R5020,

May, 1992 RU486 AND BREAST CANCER 161

56.

57.

58.

59.

60.

61.

62.

63.

64.

65.

66. 67.

68.

69.

70.

71.

72.

73.

74.

75.

76.

MC, Pastorino G, Rosso R 1990 Megestrol acetate: phase II study of a single daily administration in advanced breast cancer. Breast Cancer Res Treat 17:51 Henderson IC, Harris JR, Kinne DW, Hellman S 1989 Cancer of the breast. In: DeVita VT, Hellman S, Rosenberg SA (eds) Cancer: Principles & Practice of Oncology. JB Lippincott Company, Phil- adelphia, p 1197 McGuire WL, Cavalli F, Bonomi P, Alexieva-Figusch J 1985 Pregestin therapy for breast cancer. Breast Cancer Res Treat 6:213 McGuire WL, Johnson PA, Muss HB, Osborne CK 1989 Meges- trol acetate in breast cancer-a panel discussion. Breast Cancer Res Treat 14:33 Alexander IE, Shine J, Sutherland RL 1990 Progestin regulation of estrogen receptor messenger RNA in human breast cancer cells. Mol Endocrinol 4:821 Papa V, Reese CC, Brunetti A, Vigneri R, Siiteri PK, Goldfine ID 1990 Progestins increase insulin receptor content and insulin stimulation of growth in human breast carcinoma cells. Cancer Res 50:7858 Murphy LC, Murphy LJ, Shiu RPC 1988 Progestin regulation of EGF-receptor mRNA accumulation in T-47D human breast can- cer cells. Biochem Biophys Res Commun 150:192 Murphy LC, Dotzlaw H 1989 Endogenous growth factor expres- sion in T47D human breast cancer cells associated with reduced sensitivity to antiproliferative effects of progestins and antiestro- gens. Cancer Res 49:599 Murphy LC, Murphy LJ, Dubik D, Bell GI, Shiu RPC 1988 Epidermal growth factor gene expression in human breast cancer cells: regulation of expression by progestins. Cancer Res 484555 Green S, Chambon P 1986 A superfamily of potentially oncogenic hormone receptors. Nature 324:615 Evans RM 1988 The steroid and thyroid hormone receptor super- family. Science 240889 Beato M 1989 Gene regulation by steroid hormones. Cell 56335 Carson-Jurica MA, Schrader WT, O’Malley BW 1990 Steroid receptor family: structure and functions. Endocr Rev 11:201 Jeltsch JM, Krozowski Z, Quirin-Stricker C, Gronemeyer H, Simpson RJ, Garnier JM, Krust A, Jacob F, Chambon P 1986 Cloning of the chicken progesterone receptor. Proc Nat1 Acad Sci USA 83:5424 Conneely OM, Sullivan WP, Toft DO, Birnbaumer M, Cook RG, Maxwell BL. Zarucki-Schulz T. Greene GL. Schrader WT. O’Mal- ley BW 1986 Molecular cloning of the’chicken progesterone receptor. Science 233:767 Misrahi M, Atger M, d’Aurio1 L, Loosfelt H, Meriel C, Fridlansky F, Guiochon-Mantel A, Galibert F, Milgrom E 1987 Complete amino acid sequence of the human progesterone receptor deduced from cloned cDNA. Biochem Biophys Res Commun 143:740 Meyer M-E, Pornon A, Ji J, Bocquel M-T, Chambon P, Grone- meyer H 1990 Agonistic and antagonistic activities of RU486 on the functions of the human progesterone receptor. EMBO J 9:3923 Wei LL, Krett NL, Francis MD, Gordon DF, Wood WM, O’Mal- ley BW, Horwitz KB 1988 Multiple human progesterone receptor messenger ribonucleic acids and their autoregulation by progestin agonists and antagonists in breast cancer cells. Mol Endocrinol 2:62 Read LD, Snider CE, Miller JS, Greene GL, Katzenellenbogen BS 1988 Ligand-modulated regulation of progesterone receptor messenger ribonucleic acid and protein in human breast cancer cell lines. Mol Endocrinol 2:263 Horwitz KB, Alexander PS 1983 In situ photolinked nuclear progesterone receptors of human breast cancer cells: subunit molecular weights after transformation and translocation. Endo- crinology 113:2195 Lessey BA, Alexander PS, Horwitz KB 1983 The subunit struc- ture of human breast cancer progesterone receptors: characteriza- tion by chromatography and photoaffinity labeling. Endocrinol- ogy 112:1267 Krett NL, Wei LL, Francis MD, Nordeen SK, Gordon DF, Wood WM, Horwitz KB 1988 Human progesterone A-receptors can be

77.

78.

79.

synthesized intracellularly and are biologically functional. Biochem Bionhvs Res Commun 157:278 Wei LL, Gonzalez-Aller C, Wood WM, Miller LA, Horwitz KB 1990 5’-heterogeneity in human progesterone receptor transcripts predicts a new amino-terminal truncated “C”-receptor and unique A-receptor messages. Mol Endocrinol4:1833 Sherman MR, Corvol PL, O’Malley BW 1970 Progesterone-bind- ing components of chick oviduct. J Biol Chem 245:6085 Schrader WT, O’Malley BW 1972 Progesterone-binding compo- nents of chick oviduct. Characterization of purified subunits. J Biol Chem 247:51

80.

81.

82.

83.

Horwitz KB, Francis MD, Wei LL 1985 Hormone-dependent covalent modification and processing of human progesterone re- ceptors in the nucleus. DNA 4:451 Logeat F, Pamphile R, Loosfelt H, Jolivet A, Fournier A, Milgrom E 1985 One-step immunoaffinity purification of active progester- one receptor: further evidence in favor of the existence of a single steroid binding subunit. Biochemistry 24:1029 Conneely OM, Maxwell BL, Toft DO, Schrader WT, O’Malley BW 1987 The A and B forms of the chicken progesterone receptor arise by alternate initiation of translation of a unique mRNA. Biochem Biophys Res Commun 149:493 Conneely OM, Kettelberger DM, Tsai M-J, Schrader WT, O’Mal- ley BW 1989 The chicken progesterone receptor A and B isoforms are products of an alternate translation initiation event. J Biol Chem 264:14062

84.

85.

86.

Kastner P, Krust A, Turcotte B, Stropp U, Tora L, Gonemeyer H, Chambon P 1990 Two distinct estrogen-regulated promoters generate transcripts encoding the two functionally different hu- man progesterone receptor forms A and B. EMBO J 9:1603 Kastner P, Bocquel M-T, Turcotte B, Garnier J-M, Horwitz KB, Chambon P, Gronemeyer H 1990 Transient expression of human and chicken progesterone receptors does not support alternative translational initiation from a single mRNA as the mechanism generating two receptor isoforms. J Biol Chem 265:12163 Feil PD, Clarke CL, Satyaswaroop PG 1988 Progesterone receptor structure and protease activity in primary human endometrial carcinoma. Cancer Res 481143

87. Birnbaumer ME, Schrader WL, O’Malley BW 1979 Chemical cross-iinking of chick oviduct progesterone-receptor subunits by using a reversible bifunctional cross-linking agent. Biochem J 181:201

88.

89.

90.

91.

92.

93.

94.

95.

Dougherty JJ, Toft DO 1982 Characterization of two 8 S forms of chick oviduct progesterone receptor. J Biol Chem 257:3113 Puri RK, Grandics P, Dougherty JJ, Toft DO 1982 Purification of “nontransformed” avian progesterone receptor and preliminary characterization. J Biol Chem 257:10831 Wei LL, Sheridan PL, Krett NL, Francis MD, Toft DO, Edwards DP, Horwitz KB 1987 Immunologic analysis of human breast cancer progesterone receptors. 2. Structure, phosphorylation and processing. Biochemistry 26:6262 Schuh S, Yonemoto W, Brugge J, Bauer VJ, Riehl RM, Sullivan WP, Toft DO 1985 A 90,000-dalton binding protein common to both steroid receptors and the Rous Sarcoma virus transforming nrotein. DD~O’.““. J Biol Chem 260:14292 Catelli ‘MG, Binart N, Jung-Testas I, Renoir JM, Baulieu EE, Feramisco JR, Welch WJ 1985 The common 90-kd protein com- ponent of non-transformed ‘8s’ steroid receptors is a heat-shock protein. EMBO J 4:3131 Pratt WB 1990 Interaction of hsp90 with steroid receptors: orga- nizing some diverse observations and presenting the newest con- cepts. Mol Cell Endocrinol 74:C69 Smith DF, Faber LE, Toft DO 1990 Purification of unactivated progesterone receptor and identification of novel receptor-asso- ciated proteins. J Biol Chem 265:3996 Loaeat F. LeCunff M. Pamnhile R. Milerom E 1985 The nuclear bo;nd form of the progesterone receptor is generated through a hormone-dependent phosphorylation. Biochem Biophys Res Commun 131:421

96. Dougherty JJ, Puri RK, Toft DO 1982 Phosphorylation in uiuo of chicken oviduct nroaesterone recentor. J Biol Chem 257:14226 .

97. Sheridan PL, Krett-NL, Gordon-JA, Horwitz KB 1988 Human

on February 27, 2005 edrv.endojournals.orgDownloaded from

Page 17: The Molecular Biology of RU486. Is There a Role for ... mole… · May, 1992 RU486 AND BREAST CANCER FIG. 1. The structure of progesterone, the synthetic progestin agonist R5020,

162 HORWITZ Vol. 13, No. 2

progesterone receptor transformation and nuclear down-regula- tionare independent of phosphorylation. Mol Endocrinol 2:i329

98. DeMarzo AM. Beck CA. Onate SA. Edwards DP 1991 Dimeriza- tion of mammalian progesterone receptors occurs in the absence of DNA and is related to the release of the 90-kDa heat shock protein. Proc Nat1 Acad Sci USA 88:72

99. Tora L, Gronemeyer H, Turcotte B, Gaub M-P, Chambon P 1988 The N-terminal region of the chicken progesterone receptor spec- ifies target gene activation. Nature 333:185

_ _

100. Soelsbere TC. Halbere F 1980 Circannual rhvthms in steroid

101.

102.

103.

104.

105.

106.

107.

108.

109.

110.

111.

112.

113.

114.

115.

116.

117.

receptor concentration and nuclear binding in the chick oviduct. Endocrinology 107:1234 Boyd-Leinen PA, Fournier D, Spelsberg TC 1982 Nonfunctioning progesterone receptors in the developed oviducts from estrogen- withdrawn immature chicks and in aged nonlaying hens. Endo- crinology 111:30 Philibert D, Deraedt R, Teutsch G, RU38486, a potent antiglu- cocorticoid in uiuo. In Abstracts, VIIIth International Congress of Pharmacology, Tokyo, Japan, 1981 (No. 1463) Ravnaud J-P. Oiasoo T 1986 The design and use of sex-steroid antagonists. J Steroid Biochem 25:811 - Philibert D, Deraedt R, Teutsch G, Tournemine C, Sakiz E, RU 38486 -a new lead for steroidal anti-hormones. Program of the 64th Annual Meeting of The Endocrine Society, San Francisco, California, 1982, p 246 (Abstract 668) Hermann DW, Wyss R, Riondel A, Philibert D, Teutsch G, Sakiz E, Baulieu E-E 1982 The effects of an antiprogesterone steroid in women: interruption of the menstrual cycle and of early preg- nancy. C R Acad Sci Paris 294:933 Baulieu E-E 1989 Contragestion and other clinical applications of RU 486, an antiprogesterone at the receptor. Science 245:1351 Neef G, Beier S, Elger W, Henderson D, Wiechert R 1984 New steroids with antiprogestational and antiglucocorticoid activities. Steroids 44:349 Elger W, Beier S, Chwalisz K, Fahnrich M, Hasan SH, Henderson D, Neef G, Rohde R 1986 Studies on the mechanisms of action of progesterone antagonists. J Steroid Biochem 25:835 Ojasoo T, Raynaud J-P, Mornon J-P 1989 Steroid hormone receptors. In: Sammes PG, Taylor JB (eds) Comprehensive Me- dicinal Chemistry, Volume 3: Membranes and Receptors. Perga- mon Press, New York, p 1175 Horwitz KB 1985 The antiprogestin RU38 486: receptor-mediated progestin uersu.s antiprogestin actions screened in estrogen-insen- sitive T47D,, human breast cancer cells. Endocrinology 116:2236 Renoir J-M, Radanyi C, Baulieu E-E 1989 The antiprogesterone RU486 stabilizes the heterooligomeric, non-DNA-binding, 8S- form of the rabbit uterus cytosol progesterone receptor. Steroids 53:l Renoir J-M, Radanyi C, Jung-Testas I, Faber LE, Baulieu E-E 1990 The nonactivated progesterone receptor is a nuclear heter- ooligomer. J Biol Chem 265:14402 Horwitz KB, Pike AW, Gonzalez-Aller C, Fennessey PV 1986 Progesterone metabolism in T47D,, human breast cancer cells. II. Intracellular metabolic path of progesterone and synthetic progestins. J Steroid Biochem 25:911 Rauch M, Loosfelt H, Philibert D, Milgrom E 1985 Mechanism of action of an antiprogesterone, RU486, in the rabbit endome- trium. Effects of RU486 on the progesterone receptor and on the expression of the uteroglobin gene. Eur J Biochem 148:213 Mullick A, Katzenellenbogen BS 1986 Antiprogestin-receptor complexes: differences in the interaction of the antiprogestin RU38,486 and the progestin R5020 with the progesterone receptor of human breast cancer cells. Biochem Biophys Res Commun 135:90 Groyer A, Schweizer-Groyer G, Cadepond F, Mariller M, Baulieu E-E 1987 Antiglucocorticosteroid effects suggest why steroid hor- mone is required for receptors to bind DNA in uiuo but not in uitro. Nature 328624 El-Ashry D, Oxiate SA, Nordeen SK, Edwards DP 1989 Human progesterone receptor complexed with the antagonist RU486 binds to hormone response elements in a structurally altered form. Mol Endocrinol 3:1545

118. Sheridan PL, Evans RM, Horwitz KB 1989 Phosphotryptic pep- tide analysis of human progesterone receptors. New phosphory- lated sites formed in nuclei after hormone treatment. J Biol Chem 264:6520

119. Sheridan PL, Francis MD, Horwitz KB 1989 Synthesis of human progesterone receptors in T47D cells. Nascent A- and B-receptors are active without a phosphorylation dependent posttranslational maturation step. J Biol Chem 264:7054

120. Becker PB, Gloss B, Schmid W, Strahle U, Schutz G 1986 In uiuo protein-DNA interactions in a glucocorticoid response element require the presence of the hormone. Nature 324:686

121. Willmann T, Beato M 1986 Steroid-free glucocorticoid receptor binds specifically to mouse mammary tumour virus DNA. Nature 324:688

122. Denner LA, Weigel NL, Maxwell BL, Schrader WT, O’Malley BW 1990 Regulation of progesterone receptor-mediated transcrip- tion by phosphorylation. Science 250:1740

123. Bailly A, Le Page C, Rauch M, Milgrom E 1986 Sequence-specific DNA binding of the progesterone receptor to the uteroglobin gene: effects of hormone, antihormone and receptor phosphorylation. EMBO J 5:3235.

124. Klein-Hitpass L, Tsai SY, Weigel NL, Allan GF, Riley D, Rodri- guez R, Schrader WT, Tsai M-J, O’Malley BW 1990 The proges- terone receptor stimulates cell-free transcription by enhancing the formation of a stable preinitiation complex. Cell 60~247

125. Bagchi MK, Tsai SY, Tsai M-J, O’Malley BW 1990 Identification of a functional intermediate in receptor activation in progester- one-dependent cell-free transcription. Nature 345:547

126. Schauer M, Chalepakis G, Willmann T, Beato M 1989 Binding of hormone accelerates the kinetics of glucocorticoid and progesier- one recentor binding to DNA. Proc Nat1 Acad Sci USA 86:1123

127. Bagchi MK, Elliston JF, Tsai SY, Edwards DP, Tsai M-J, O’Mal- ley BW 1988 Steroid hormone-dependent interaction of human progesterone receptor with its target enhancer element. Mol En- docrinol2:1221

128. Kalff M, Gross B, Beato M 1990 Progesterone receptor stimulates transcription of mouse mammary tumour virus in a cell-free system. Nature 344:360

129. Klein-Hitpass L, Cato ACB, Henderson D, Ryffel GU 1991 Two types of antiprogestins identified by their differential action in transcriptionally active extracts from T47D cells. Nucleic Acids Res 19:1227

130. Guiochon-Mantel A, Loosfelt H, Ragot T, Bailly A, Atger M, Misrahi M. Perricaudet M. Milerom E 1988 Recenters bound to I

antiprogestin form abortive complexes with hormone response elements. Nature 336:695

131. Tsai SY, Carlstedt-Duke J, Weigel NL, Dahlman K, Gustafsson J-A, Tsai M-J, O’Malley BW 1988 Molecular interactions of steroid hormone receptor with its enhancer element: evidence for receptor dimer formation. Cell 55:361

132. Fawell SE, Lees JA, White R, Parker MG 1990 Characterization and colocalization of steroid and dimerization activities in the mouse estrogen receptor. Cell 60:953

133. Guiochon-Mantel A, Loosfelt H, Lescop P, Sar S, Atger M, Perrot-Applanat M, Milgrom E 1989 Mechanisms of nuclear localization of the progesterone receptor: evidence for interaction between monomers. Cell 57:1147

134. Skafar D 1991 Differences in the binding mechanism of RU486 and progesterone to the progesterone receptors. Biochemistry 30:10829

135. Strahle U, Klock G, Schutz G 1987 A DNA sequence of 15 base pairs is sufficient to mediate both glucocorticoid and progesterone induction of gene expression. Proc Nat1 Acad Sci USA 84:7871

136. Jantzen H-M. Strahle U. Gloss B. Stewart F. Schmid W. Boshart M, Miksicek ‘R, Schutz’G 1987 ‘Cooperativity of glucdcorticoid response elements located far upstream of the tyrosine amino transferase gene. Cell 49:29

137. Tsai SY, Tsai M-J, O’Malley BW 1989 Cooperative binding of steroid hormone receptors contributes to transcriptional syner- gism at target enhancer elements. Cell 57:443

138. Majors J, Varmus HE 1983 A small region of the mouse mammary tumor virus long terminal repeat confers glucocorticoid hormone

on February 27, 2005 edrv.endojournals.orgDownloaded from

Page 18: The Molecular Biology of RU486. Is There a Role for ... mole… · May, 1992 RU486 AND BREAST CANCER FIG. 1. The structure of progesterone, the synthetic progestin agonist R5020,

May, 1992 RU486 AND BREAST CANCER 163

139.

140.

141.

142.

143.

144.

145.

146.

147.

148

149.

150.

151.

152.

153.

154.

155.

156.

157.

regulation of a linked heterologous gene. Proc Nat1 Acad Sci USA 80:5866 Buetti E, Diggelmann H 1983 Glucocorticoid regulation of mouse mammary tumour virus: identification of a short essential DNA region. EMBO J 2:1423 Nordeen SK, Suh BJ, Kuhnel B, Hutchinson CA 1990 Structural determinants of a glucocorticoid receptor recognition element. Mol Endocrinol 4:1866 Perlmann T, Eriksson P, Wrange 0 1990 Quantitative analysis of the glucocorticoid receptor-DNA interaction at the mouse mammary tumor virus glucocorticoid response element. J Biol Chem 265:17222 Theveny B, Bailly A, Rauch C, Rauch M, Delain E, Milgrom E 1987 Association of DNA-bound receptors. Nature 329:79 Cato ACB, Miksicek R, Schutz G, Arnemann J, Beato M 1986 The hormone regulatory element of mouse mammary tumour virus mediates progesterone induction. EMBO J 5:2237 Cato ACB, Skroch P, Weinmann J, Butkeraitis P, Ponta H 1988 DNA sequences outside the receptor-binding sites differentially modulate the responsiveness of the mouse mammary tumour virus promoter to various steroid hormones. EMBO J 7:1403 Buetti E, Kuhnel B 1986 Distinct sequence elements involved in the glucocorticoid regulation of the mouse mammary tumor virus promoter identified by linker scanning mutagenesis. J Mol Biol 190:379 Burnstein KL, Bellingham DL, Jewel1 CM, Powell-Oliver FE, Cidlowski JA 1991 Autoregulation of glucocorticoid receptor gene expression. Steroids 56:52 Horwitz KB, Mockus MB, Lessey BA 1982 Variant T47D human breast cancer cells with high progesterone-receptor levels despite estrogen and antiestrogen resistance. Cell 28633 Mockus MB, Horwitz KB 1983 Progesterone receptors in human breast cancer. Stoichiometric translocation and nuclear process- ing. J Biol Chem 258:4778 Horwitz KB. Mockus MB. Pike AW. Fennessev PV. Sheridan RL 1983 Progesterone receptor replenishment in T47Dhuman breast cancer cells. Roles of protein synthesis and hormone metabolism. J Biol Chem 2587603 Mullick A, Katzenellenbogen BS 1986 Progesterone receptor syn- thesis and degradation in MCF-7 human breast cancer cells as studied by dense amino acid incorporation. Evidence for a non- hormone binding receptor precursor. J Biol Chem 261:13236 Nardulli AM, Katzenellenbogen BS 1988 Progesterone receptor regulation in T47D human breast cancer cells: analysis by density labeling of progesterone receptor synthesis and degradation and their modulation by progestin. Endocrinology 122:1532 Horwitz KB, McGuire WL 1978 Nuclear mechanisms of estrogen action. Effects of estradiol and antiestrogens on estrogen receptors and nuclear receptor processing. J Biol Chem 253:8185 Horwitz KB, McGuire WL 1978 Estrogen control of progesterone receptor in human breast cancer. Correlation with nuclear proc- essinn of estrogen recenters. J Biol Chem 253:2223 Hor&z KB, &ginger P, Kuttenn F, McGuire WL 1981 Nuclear estrogen receptor release from antiestrogen suppression: amplified induction of progesterone receptor in MCF-7 human breast cancer cells. Endocrinology 108:1703 Keydar I, Chen L, Karby S, Weiss FR, Delarea J, Radu M, Chaitcik S, Brenner HJ 1979 Establishment and characterization of a cell line of human carcinoma origin. Eur J Cancer 15:659 Bardon S, Vignon F, Chalbos D, Rochefort H 1985 RU486, a progestin and glucocorticoid antagonist, inhibits the growth of breast cancer cells via the progesterone receptor. J Clin Endocri- no1 Metab 60:692 Bakker GH, Setyono-Han B, Henkelman MS, De Jong FH, Lam- berts SWJ. van der Schoot P. Kliin JGM 1987 Comnarison of the actions of the antiprogestin mifepristone (RU486), the progestin megestrol acetate, the LHRH analog buserelin, and ovariectomy in treatment of rat mammary tumors. Cancer Treatment Rep 71:1021

158.

159.

160.

161.

162.

163.

164.

165.

166.

167.

168.

169.

170.

171.

172.

Sutherland RL, Hall RE, Pang GYN, Musgrove EA, Clarke CL 1988 Effect of medroxyprogesterone acetate on proliferation and cell cycle kinetics of human mammary carcinoma cells. Cancer Res 485084 Rochefort H, Chalbos 1985 RU 486: A full progestin antagonist in human breast cancer cell lines. In: Baulieu E-E, Segal SJ (eds) The Antiprogestin Steroid RU 486 and Human Fertility Control. Plenum Publishing Corporation, New York, p 307 Gill PG, Vignon F, Bardon S, Derocq D, Rochefort H 1987 Difference between R5020 and the antiprogestin RU486 in anti- proliferative effects on human breast cancer cells. Breast Cancer Res Treat l&37 Bardon S, Vignon F, Montcourrier P, Rochefort H 1987 Steroid receptor-mediated cytotoxicity of an antiestrogen and an antipro- gestin in breast cancer cells. Cancer Res 47:1441 Michna H, Schneider M, Nishino Y, El Etreby MF, McGuire WL 1990 Progesterone antagonists block the growth of experimental mammary tumors in Go/G1. Breast Cancer Res Treat i7:155 Bowden RT. Hissom JR. Moore MR 1989 Growth stimulation of T47D human breast cancer cells by the anti-progestin RU486. Endocrinology 124:2642

_ -

Graham ML. Krett NL. Miller LA. Leslie KK. Gordon DF. Wood WM, Wei LL, Horwitz KB 1990 T47D,, cells, genetically unstable and containing estrogen receptor mutations, are a model for the progression of breast cancers to hormone resistance. Cancer Res 50:6208 Bakker GH, Setyono-Han B, Portengen H, De Jong FH, Foekens JA, Klijn JGM 1989 Endocrine and antitumor effects of combined treatment with an antiprogestin and antiestrogen or luteinizing hormone-releasing hormone agonist in female rats bearing mam- mary tumors. Endocrinology 125:1593 Bakker GH, Setyono-Han B, Portengen H, De Jong FH, Foekens JA, Klijn JGM 1990 Treatment of breast cancer with different antiprogestins: preclinical and clinical studies. J Steroid Biochem 37:789 Klijn JGM, De Jong FH, Bakker GH, Lamberts SWJ, Rodenburg CJ, Alexieva-Figusch J 1989 Antiprogestins, a new form of endo- crine therapy for human breast cancer. Cancer Res 49:2851 Michna H, Schneider MR, Nishino Y, El Etreby FM 1989 Anti- tumor activity of the antiprogestins ZK98.299 and RU38.486 in hormone dependent rat and mouse mammary tumors: mechanistic studies. Breast Cancer Res Treat 14:275 Schneider MR, Michna H, Nishino Y, El Etreby FM 1989 Anti- tumor activity of the progesterone antagonists ZK 98.299 and RU 38.486 in the hormone-dependent MXT mammary tumor model of the mouse and the DMBA- and the MNU-induced mammary tumor models of the rat. Eur J Cancer Clin Oncol25:691 Michna H. Schneider MR. Nishino Y. El Etrebv MF 1989 The antitumor mechanism of progesterone antagonists is a receptor mediated antiproliferative effect by induction of terminal cell death. J Steroid Biochem 34:447 Maudelonde T, Romieu G, Ulmann A, Pujol H, Grenier J, Khalaf S, Cavalie G, Rochefort H 1987 First clinical trial on the use of the antiprogestin RU486 in advanced breast cancer. In: Klijn JGM. Paridaens R. Foekens JA (eds) Hormonal Maninulation of Cancer: Peptides, Growth Factors’ and New (AntL)Steroidal Agents. Raven Press, New York, p 55 Gaillard RC, Riondel A, Muller AF, Herrmann W, Baulieu E-E 1984 RU486: a steroid with antiglucocorticosteroid activity that only disinhibits the human pituitary-adrenal system at a specific time of day. Proc Nat1 Acad Sci USA 81:3879

173. Murphy LC, Dotzlaw H, Johnson Wong MS, Miller T, Murphy LJ 1991 Mechanisms involved in the evolution of progestin re- sistance in human breast cancer cells. Cancer Res 51:2051

174. Howell A, Harland RNL, Barnes DM, Hayward E, Redford J, Swindell R, Sellwood RA 1987 Endocrine therapy for advanced carcinoma of the breast: effect of tumor heterogeneity and site of biopsy on the predictive value of progesterone receptor estima- tions. Cancer Res 47:296

on February 27, 2005 edrv.endojournals.orgDownloaded from