<<

Endocrine-Related Cancer (2006) 13 689–706 REVIEW Pure oestrogen antagonists for the treatment of advanced

Anthony Howell

CRUK Department of Medical Oncology, University of Manchester, Christie Hospital NHS Trust, Manchester M20 4BX, UK (Requests for offprints should be addressed to A Howell; Email: [email protected])

Abstract For more than 30 years, has been the drug of choice in treating patients with oestrogen receptor (ER)-positive breast tumours. However, research has endeavoured to develop agents that match and improve the clinical efficacy of tamoxifen, but lack its partial agonist effects. The first ‘pure’ oestrogen antagonist was developed in 1987; from this, an even more potent derivative was developed for clinical use, known as (ICI 182,780, ‘Faslodex’). Mechanistic studies have shown that fulvestrant possesses high ER-binding affinity and has multiple effects on ER signalling: it blocks dimerisation and nuclear localisation of the ER, reduces cellular levels of ER and blocks ER-mediated gene transcription. Unlike anti-oestrogens chemically related to tamoxifen, fulvestrant also helps circumvent resistance to tamoxifen. There are extensive data to support the lack of partial agonist effects of fulvestrant and, importantly, its lack of cross-resistance with tamoxifen. In phase III studies in patients with locally advanced or metastatic breast cancer, fulvestrant was at least as effective as in patients with tamoxifen-resistant tumours, was effective in the first-line setting and was also well tolerated. These data are supported by experience from the compassionate use of fulvestrant in more heavily pretreated patients. Further studies are now underway to determine the best strategy for sequencing oestrogen endocrine therapies and to optimise dosing regimens offulvestrant. At present, and for the foreseeable future, fulvestrant is the only oestrogen antagonist with no agonistic effects licensed for the treatment of advanced breast cancer in postmenopausal women. Other similar oestrogen antagonists are undergoing research and development, with a few currently being evaluated in phase II trials. Endocrine-Related Cancer (2006) 13 689–706

Background use. As such, research has endeavoured to develop The critical role played by oestrogen in breast cancer agents that are at least as effective as tamoxifen but has driven a huge amount of research into the which lack its partial agonist effects. development of hormonal therapies that are able to First-generation selective ER modulators (SERMs, block oestrogen-mediated cell signalling. For more e.g. , and ) were based than 30 years, tamoxifen, a non-steroidal triphenyl- on the non-steroidal structure of ethylene derivative (Fig. 1), has been the drug of tamoxifen (Wakeling 2000). These agents have no choice in treating patients with oestrogen receptor efficacy benefits over tamoxifen and also show partial (ER)-positive and/or progesterone receptor (PgR)- agonist activity (Pyrhonen et al. 1994, Lee et al. 2000, positive tumours. Not only is tamoxifen effective in Buzdar et al. 2002), with some studies suggesting there patients with breast cancer, it also reduces the is cross-resistance ‘between’ first-generation SERMs incidence of disease in healthy women at high risk of and tamoxifen (Haarstad et al. 1992, Stenbygaard et al. developing breast cancer (Cuzick et al. 2003). The 1993, Pyrhonen et al. 1994, 1997, 1999, Hayes et al. success of tamoxifen has been achieved despite its 1995, Gershanovich et al. 1997, Lee et al. 2000, partial agonist activity, which is associated with Johnston et al. 2001, MillaSantos et al. 2001). increased risk of endometrial cancer, thromboembolic Disappointing efficacy data for both droloxifene and events and tumour flare, particularly with long-term idoxifene when compared with tamoxifen or when

Endocrine-Related Cancer (2006) 13 689–706 Downloaded from Bioscientifica.comDOI: 10.1677/erc.1.00846 at 09/27/2021 10:26:15PM 1351-0088/06/013–689 q 2006 Society for Endocrinology Printed in Great Britain Online version via http://www.endocrinology-journals.orgvia free access A Howell: Pure anti-oestrogens in advanced breast cancer

Figure 1 Chemical structures of oestradiol, tamoxifen, fulvestrant and . used in tamoxifen-resistant disease have led to their of steroidal 7a-alkylamide analogues of oestradiol. development being halted. Toremifene (chloro-tamox- Of these, ICI 164,384 was the first pure oestrogen ifen), which has similar efficacy but fewer agonist antagonist to be described, completely blocking the properties compared with tamoxifen (Marttunen et al. uterotrophic action of both oestradiol and tamoxifen in 1998, 2001), remains in clinical development in the rats (Wakeling & Bowler 1987). Following this, a far breast cancer prevention setting. more potent pure oestrogen antagonist was developed, Structurally distinct second- and third-generation ICI 182,780, which is now known as fulvestrant SERMs (‘fixed-ring’ benzothiophene derivatives (‘Faslodex’) (Fig. 1)(Wakeling et al. 1991). At present (Fig. 1)) such as raloxifene, and ERA- (and for the foreseeable future), fulvestrant, an 923 were then developed. Most of these have oestrogen antagonist with no agonist effects, is licensed demonstrated anti-tumour activity in breast cancer for the treatment of advanced breast cancer in but also have some partial agonist activity (Buzdar postmenopausal women. Therefore, this paper will et al. 1988, Gradishar et al. 2000, Munster et al. 2001, describe in detail the mode of action of fulvestrant, Buzdar et al. 2003). Results from the Study of and explore how this underlies its clinical efficacy, Tamoxifen and Raloxifene (STAR) trial, one of the tolerability and safety profile in the treatment of largest breast cancer prevention studies, which has advanced breast cancer. In considering the future recruited 19 000 postmenopausal women at risk of directions for oestrogen-antagonistic agents, ongoing breast cancer, are expected in 2006. Based on its clinical trials with fulvestrant will be reviewed. The partial oestrogen agonist effects (Delmas et al. 1997), current status of other ‘pure’ oestrogen antagonists in raloxifene is also currently being used for the preclinical and clinical development will also be prevention and treatment of and breast described. cancer. Arzoxifene showed promising initial results in a phase II study in advanced breast cancer, although its efficacy was greater in tamoxifen-sensitive than Mode of action tamoxifen-resistant disease (Buzdar et al. 2003). Results from a phase I study suggest that ERA-923 ER-mediated signalling has no trophic effects on the uterus (Cotreau et al. The biological actions of oestrogen are mediated by 2002), and its role in patients with breast cancer is two subtypes of ER, ERa and ERb. Both receptors currently being investigated. belong to a family of ligand-activated nuclear The search for novel agents that would completely transcription factors (Evans 1988) and share a high block all ER signalling led to the synthesis of a series degree of homology in their DNA binding domains

Downloaded from Bioscientifica.com at 09/27/2021 10:26:15PM via free access 690 www.endocrinology-journals.org Endocrine-Related Cancer (2006) 13 689–706

(Kuiper et al. 1996). However, they differ considerably to promoters of responsive genes. Genes that are in their N-terminal regions, which contain the ligand- regulated by oestrogen but do not have an ERE include independent transcription activation function, AF1 cyclin D1, p21 and the PgR (Shupnik 2004). Many (Kuiper et al. 1996). These differences suggest that such genes are regulated via ER interactions with ERa and ERb could have distinct functions in terms of stimulating protein 1 (Sp1), the signal transducers and gene regulation and may contribute to the selec- activators of transcription (STAT) family of transcrip- tive action of oestradiol in different target tissues tion factors and transcription factors associated with (Gustafsson & Warner 2000). There is evidence from activating protein 1 (AP1) sites. The AP1 transcription in vitro studies to suggest that imbalanced ERa/ERb factors are a family of homo- and heterodimers expression may be a feature of oestrogen-dependent composed of proteins that belong to the Jun, Fos, tumour progression and that ERb has a key role in Maf and ATF subfamilies that bind to 12-0-tetra- providing protection against ERa-induced hyperproli- decanoylphorbol-13-acetate (TPA) response elements feration (Bardin et al. 2004). It has also been suggested (TRE) or cAMP response elements (CRE) in target that ERb may be a dominant regulator of oestrogen genes (Chinenov & Kerppola 2001). The c-Jun signalling as it causes a dose-dependent reduction homodimer is the most potent transcriptional activator in ERa-mediated transcription when coexpressed with in the AP1 family (Matthews et al. 2006). ERa (Lindberg et al. 2003). While both ERa and ERb can bind oestradiol and activate transcription through an ERE, they have Classical pathway of ER signalling differential transcriptional activities at Sp1 and AP1 sites. Only ERa can stimulate genes via the Sp1 In classical ER signalling, unbound ER can be nuclear pathway and the two isoforms are differentially localised and bound loosely to the oestrogen response regulated by oestrogens and antioestrogens at AP1 elements (ERE) (Kumar & Chambon 1988, Reese & sites (Shupnik 2004). Results of a recent in vitro study Katzenellenbogen 1991). However, binding of oestra- suggested that ERb may preferentially antagonise the diol to the ER initiates a cascade of events leading to nonclassical AP1-mediated pathway regulated by ERa strong ER dimerisation, increased nuclear localisation (Matthews et al. 2006). Interestingly, another in vitro and binding to ERE in the regulatory regions of target study suggests that ER expression may positively genes, and gene transcription (mediated by AF1 and contribute to breast cancer growth in two ways. In AF2 of the ER) (Beato 1989, Tsai & O’Malley 1994, normal situations, ER is activated by oestrogen to Cheskis et al. 1997). AF1 (located in domains A and B promote breast cancer cell proliferation, primarily of the amino terminal of the ER) is activated by growth through ERE-mediated transcription. However, in factors acting through the mitogen-activated protein stressful conditions, such as during kinase (MAPK) pathway, while AF2 (located in treatment, ER may facilitate breast cancer growth by domain E of the carboxy terminal of the ER ligand- inhibiting cell death via binding c-Jun and regulating binding domain) is activated by oestradiol. In the AP1-mediated transcription (Qi et al. 2004). transcription complex, the activated AFs recruit coactivators and corepressors of oestrogen-regulated transcription (Horwitz et al. 1996, White & Parker Ligand-independent ER signalling pathways 1998), with full transcriptional activity requiring both During recent years, it has been documented that AFs to be active. Ligand-specific conformational crosstalk between oestrogen and growth factor-stimu- changes in the ER strongly influence the protein:pro- lated intracellular signalling pathways can directly tein interactions in the transcriptional complex, leading activate the ER and the transcription of ER-regulated to differences in gene transcription and activation for genes in the absence of oestrogen (Weigel & Zhang ER-binding ligands (Brzozowski et al. 1997, Shiau 1998). As AF1 is activated via factors involved in the et al. 1998). Indeed, ligand-dependent conformational MAPK pathway, crosstalk exists between the ER, changes in the ER show that there are distinct growth factor receptors (e.g. human epidermal growth differences between oestradiol–ER, tamoxifen–ER factor receptors 1 and 2 (EGFR and HER2)) and kinase and fulvestrant–ER complexes (Paige et al. 1999). pathways (Nicholson & Johnston 2005). ER–growth factor receptor cross-talk is thought to be involved in Nonclassical pathways of ER signalling the development of resistance to endocrine therapy by In addition to binding to classical ERE motifs, ERs can reducing the reliance on oestrogen-sensitive prolifer- stimulate gene transcription via a nonclassical pathway ation. As such, inhibition of growth factor receptor by interacting with other transcription factors bound pathways, particularly in conjunction with inhibition

Downloaded from Bioscientifica.com at 09/27/2021 10:26:15PM via free access www.endocrinology-journals.org 691 A Howell: Pure anti-oestrogens in advanced breast cancer of ER signalling, may provide additional ways of activity, which itself is dependent upon cellular and circumventing endocrine resistance (Nicholson & promoter factors (Berry et al. 1990, Tzukerman et al. Johnston 2005). 1994). Other SERMs such as idoxifene and raloxifene also inhibit ER AF2 but show partial agonist activity ‘Non-genomic’ actions of oestradiol against AF1 in MCF-7 cells (Wijayaratne et al. 1999). However, not all of the biological effects of partial In addition to the classical action of ER as a ligand- oestrogen antagonists may be explained by their ability regulated transcription factor, oestradiol also exerts to differentially regulate AF1 and AF2, as other AF early signalling events within target cells minutes after domains may also exist that only function when AF1 stimulation, making it likely that these effects are and AF2 are inactive. Indeed, an autonomous independent of transcription. These processes may be activation domain, referred to as AF2a, has already mediated by membrane-associated ER (Hanstein et al. been described (Norris et al. 1997), although the 2004) or the G-coupled membrane receptor GPR30 impact of partial oestrogen antagonists on this domain (Filardo & Thomas 2005). Activation of membrane- remains unclear. Thus, the effects of these agents are associated ER has been shown to lead to activation of likely to be cell, promoter and effector sensitive, which MAPK signalling, phosphatidylinositol-3-kinase may explain their differential actions on bone, the (PI-3K) signalling and a rise in cytoplasmic calcium cardiovascular system and the uterus. levels. It can also activate release of epithelial growth Although ERa and ERb AF1 generally exhibit weak factor (EGF) and insulin-like growth factor (IGF), thus activity at genes with classical EREs, there are certain leading to stimulation of EGFR and IGFR, respectively cellular environments in which differential effects of (Hanstein et al. 2004). GPR30 can act independently of SERMs on AF1 activity at EREs are detectable. In such ERa and ERb and has been linked to oestrogen- situations, tamoxifen-bound ERa has similar activity mediated regulation of the EGFR to MAPK signalling to unbound AF1 and raloxifene-bound ERa has less axis via stimulation of adenylyl cyclase activity activity. When bound by the pure antioestrogen, ICI (Filardo et al. 2000, 2002, Filardo & Thomas 2005). 164,384, ERa has no activity at all. These differences appear to be due to differential recruitment of Effect of partial oestrogen antagonists upon corepressors (Webb et al. 2003). Other oestrogen oestrogen signalling agonist effects of SERMs stem from ER activity at Even though the affinity of tamoxifen for the ER is genes with alternate response elements. For example, 2.5% that of oestradiol (Wakeling & Bowler 1987, ERa enhances AP1 activity in the presence of Wakeling et al. 1991), tamoxifen has similar mole- tamoxifen and shows weaker activity in the presence cular effects to oestradiol once bound to the ER of raloxifene (Paech et al. 1997). SERMs can also (Katzenellenbogen et al. 1996) (i.e. increased nuclear enhance AP1 activity via an AF-independent mechan- translocation and ERE binding of the tamoxifen–ER ism; here, raloxifene strongly enhances ERa action complex). This may be due to the fact that the major whereas tamoxifen does not (Webb et al. 2003). mediator of tamoxifen action is 4-hydroxy-tamoxifen, Therefore, SERMs differentially regulate two mech- which has been shown to have a similar affinity for the anisms of ER action at AP1 sites. ER as oestradiol (Borgna & Rochefort 1980). Tamoxifen, idoxifene and raloxifene have all been shown to increase ERa protein and mRNA levels in Effect of pure oestrogen antagonists upon MCF-7 cells (Wijayaratne et al. 1999), possibly as a oestrogen signalling result of their oestrogen agonist activity. Fulvestrant is a 7a-alkylsulphinyl analogue of 17b- The gene-specific actions of partial oestrogen oestradiol that is structurally distinct from the non- antagonists such as tamoxifen, toremifene and raloxi- steroidal oestrogen antagonists. The C7 side chain of fene (all of which are non-steroidal in structure) can fulvestrant is crucial for its mode of action; it be accounted for by several factors, including ligand differentiates fulvestrant from oestradiol, and its affinity and differences in ER conformation, and position, length and flexibility determine the pure differences in the efficiency of coactivators and antagonistic activity of fulvestrant (Bowler et al. 1989). corepressors coupling to the transcriptional complex This chemical structure is also responsible for the Katzenellenbogen et al. 1996). While AF2 is inacti- important differences between fulvestrant and tamoxi- vated in the tamoxifen–ER complex, AF1-mediated fen. First, the binding affinity of fulvestrant to the ER is transcription is unaffected by tamoxifen. The partial 89% that of oestradiol (Wakeling & Bowler 1987). agonism observed with tamoxifen is attributed to AF1 Secondly, the conformational change induced by

Downloaded from Bioscientifica.com at 09/27/2021 10:26:15PM via free access 692 www.endocrinology-journals.org Endocrine-Related Cancer (2006) 13 689–706 fulvestrant binding means that ER homodimerisation Fulvestrant may help to circumvent hormone and energy-dependent nuclear-cytoplasmic shuttling is resistance via pathways of receptor cross-talk by prevented, blocking nuclear localisation of the ER reducing cellular ER levels (Hutcheson et al. 2003). (Fawell et al. 1990, Dauvois et al. 1993). In addition, Indeed, one major difference between fulvestrant and cellular levels of ER are decreased via several tamoxifen is the ability of fulvestrant to abrogate ER mechanisms: turnover is increased, half-life is activation mediated by other signalling factors, such as decreased and there is rapid ER degradation EGFR, IGFR and dopamine (Ignar-Trowbridge et al. (Fawell et al. 1990, Dauvois et al. 1992, Wijayaratne 1993, Smith et al. 1993, Newton et al. 1994). In et al. 1999). contrast, tamoxifen is unable to block the activation of Fulvestrant also disables both AF1 and AF2 on ERa; ER by the same mediators (Ignar-Trowbridge et al. thus, even if any fulvestrant–ER complexes are present 1993, Smith et al. 1993, Newton et al. 1994). in the nucleus, gene transcription is abrogated, despite the fact that the fulvestrant–ER complex binds DNA Biological effects of pure oestrogen antagonists (Wijayaratne et al. 1999). As mentioned previously, when ERa is bound to the pure antioestrogen, ICI There is extensive preclinical data to support the pure 164,384, no AF1 activity is detectable at classical EREs oestrogen antagonist effects of fulvestrant. In vitro, (Webb et al. 2003). Fulvestrant has been shown to fulvestrant significantly decreased ER protein inhibit ERa-mediated transcription from an ERE in levels and inhibited PgR expression in MCF-7 cells MCF-7 cells more effectively than tamoxifen, raloxifene (McClelland et al. 1996). In contrast, tamoxifen or idoxifene (Wijayaratne et al. 1999). In contrast to increased MCF-7 ER (oestrogen withdrawal has a tamoxifen, pure antioestrogens can strongly enhance similar effect) and PgR protein levels (Horowitz & ERa activity at AP1 sites via an AF-independent McGuire 1978, Nicholson et al. 1995, Early Breast mechanism (Webb et al. 2003). A recent cDNA Cancer Trialists’ Collaborative Group 1998, Hutcheson et al. 2003). In vivo, a single 5 mg dose of fulvestrant microarray study has also highlighted the differences (short-acting formulation) was as effective as tamoxifen in levels of ER antagonism between fulvestrant and the (10 mg/kg/day orally) in blocking the growth of MCF-7 SERMs (Frasor et al. 2004). Here, fulvestrant was xenografts in nude mice (Wakeling et al. 1991). In shown to antagonise the expression of 95% of addition, fulvestrant (5 mg weekly) suppressed the oestrogen-upregulated genes and 91% of oestrogen- growth of established MCF-7 xenografts in mice for downregulated genes. In contrast, tamoxifen twice as long as tamoxifen (500 mg/day) or oestrogen antagonised expression of only 47% of oestrogen- withdrawal, and was more effective than tamoxifen in upregulated genes and 26% of oestrogen-downregulated reducing the expression of oestrogen-regulated genes in genes. The antagonistic activity of raloxifene was MCF-7 xenograft tissue (Osborne et al. 1995). somewhere in between, being 67% for oestrogen- The effect of short-term administration of fulvestrant upregulated and 33% for oestrogen-downregulated on breast cancer was initially evaluated in 56 women genes. Conversely, only tamoxifen had full agonistic with primary breast cancer who were randomised to activity on a substantial proportion of genes; 23% of receive no preoperative treatment (nZ16) or daily i.m. those upregulated by oestrogen and 31% of those fulvestrant (nZ37 (6 mg, nZ21; 18 mg, nZ16)) for downregulated. In contrast, fulvestrant and raloxifene 7 days prior to primary breast surgery (DeFriend et al. % had full agonistic activity on 8% of oestrogen- 1994). There was no evidence of agonist activity using regulated genes overall (Frasor et al. 2004). fulvestrant, which significantly reduced tumour ER Fulvestrant has also been shown to affect rapid, (median ER index, 0.72 before vs 0.02 after treatment; ‘nongenomic’ oestrogen signalling via membrane- P!0.001), PgR (median PgR index, 0.50 before vs associated ER. Activation of the MAPK pathway via 0.01 after treatment; P!0.05) and Ki67 (median Ki67 oestrogen stimulation of membrane-associated ER is labelling index, 3.2 before vs 1.1 after treatment; abolished by fulvestrant in vitro; however, activation P!0.05) expression levels (DeFriend et al. 1994). of MAPK via EGF is unaffected (Improta-Brears et al. More recently in the preoperative setting, 201 1999). Interestingly, as seen with oestrogen, both postmenopausal women with stage T1-3 breast fulvestrant and tamoxifen have been shown to cancer were treated with fulvestrant (50 mg, 125 mg activate GPR30-mediated stimulation of adenylyl or 250 mg on day 0 as an i.m. injection), tamoxifen cyclase, leading to cAMP-mediated inhibition of (20 mg/day orally) or tamoxifen placebo for Erk-1 and Erk-2 and the attenuation of the EGFR- 14–21 days prior to surgery of curative intent MAPK signalling cascade (Filardo et al. 2000, 2002). (Robertson et al. 2001). Tumour biopsies (taken at

Downloaded from Bioscientifica.com at 09/27/2021 10:26:15PM via free access www.endocrinology-journals.org 693 A Howell: Pure anti-oestrogens in advanced breast cancer baseline and during surgery 15–22 days after dosing) point (Robertson et al. 2004). Thus, fulvestrant can were analysed for ER and PgR levels (H-score), maintain ER suppression for a sustained period proliferation (Ki67 levels) and apoptosis markers. (beyond 6 months), and downregulation may be higher ER levels were significantly decreased by tamoxifen in those experiencing CB than those with de novo when compared with placebo (PZ0.049) and by all progression. The presence of tumour ER in all patients doses of fulvestrant compared with placebo (P!0.026) at the time of progression supports the use of further (Fig. 2). In addition, ER levels were significantly endocrine therapy after fulvestrant. decreased by 250 mg fulvestrant compared with Moreover, ER expression in patients with PD might tamoxifen (PZ0.024). While tamoxifen significantly provide insights into mechanisms of resistance through increased PgR levels compared with placebo growth factor receptor cross-talk. The amount of cross- (PZ0.009), fulvestrant, 125 mg and 250 mg, signifi- resistance observed between tamoxifen and the newer cantly reduced PgR levels compared with either partial antagonists being developed was a major problem. placebo or tamoxifen (P%0.003). Although, fulves- Importantly, cross-resistance between fulvestrant and trant and tamoxifen significantly reduced proliferation tamoxifen has not been observed in vitro; fulvestrant is a compared with placebo (P!0.046 for both), none of far more potent inhibitor of tamoxifen-sensitive and w the treatment arms affected apoptotic indices tamoxifen-resistant MCF-7 proliferation ( 150 and (Robertson et al. 2001). The differential effects on 1540 times more potent than tamoxifen respectively) PgR levels observed highlight the ‘pure’ oestrogen (Hu et al. 1993), and tamoxifen inhibits both fulvestrant- antagonist effects of fulvestrant. A more recent study sensitive and fulvestrant-resistant MCF-7 cell lines has analysed the effect of long-term pre- and post- (Lykkesfeldt et al. 1995). In vivo, the growth of operative fulvestrant (250 mg/month) on ER tamoxifen-resistant MCF-7 xenografts in nude mice m expression in patients with locally advanced or (generated following treatment with 500 g/day tamoxifen for 6 months) was abrogated by fulvestrant metastatic breast cancer (Robertson et al. 2004). (5 mg/week) (Osborne et al. 1994). These data suggested Nineteen out of 24 patients experienced clinical benefit that fulvestrant had potential in the treatment of patients (CB), and this group had a significant reduction in with tamoxifen-resistant tumours. tumour ER levels after 4–6 weeks and 6 months of treatment compared with pretreatment levels (PZ0.029 and PZ0.011 respectively). In contrast, ER Lack of agonist effects levels were not significantly reduced in the five patients Despite the fact that tamoxifen is well tolerated, the with progressive disease (PD) at the 4–6 weeks time risks of thromboembolic disease and endometrial

Figure 2 Percentage change in tumour oestrogen receptor (ER) and progesterone receptor (PgR) levels using short-term fulvestrant in patients with ER-positive and PgR-positive tumours. Biopsies were taken prior to treatment and during surgical excision 15–22 days later. Based on data in Robertson et al. (2001).

Downloaded from Bioscientifica.com at 09/27/2021 10:26:15PM via free access 694 www.endocrinology-journals.org Endocrine-Related Cancer (2006) 13 689–706 cancer increase with long-term use due to partial suppressed tumour growth for longer than tamoxifen agonist activity (Fisher et al. 1994, 1996, Early Breast (Osborne et al. 1995). Thus, fulvestrant was shown not to Cancer Trialists’ Collaborative Group 1998). In be cross-resistant with tamoxifen in the clinical setting. contrast, a wealth of preclinical and clinical data These data were confirmed more recently in two phase show that the pure oestrogen antagonist fulvestrant III trials in postmenopausal women with advanced breast lacks these partial agonist effects. cancer who previously had progressed after receiving In vivo, fulvestrant (0.03–1.0 mg/kg/day for 14 days) anti-oestrogen treatment (Howell et al. 2002, Osborne inhibited the uterotrophic effect of oestradiol (0.5 mg) et al. 2002). Both studies had a similar design, in which or tamoxifen (1 mg/kg) in immature or ovariectomised fulvestrant (250 mg/month i.m., nZ428) was compared rodents in a dose-dependent manner (Wakeling et al. with the (AI) anastrozole (1 mg/day 1991). In contrast, tamoxifen only partially blocked the orally, nZ423). In one study, fulvestrant was adminis- uterotrophic activity of oestradiol (Wakeling et al. tered as a single 5 ml injection in an open-label 1991). Fulvestrant (4 mg short-acting formulation) comparison (International), whereas in the other study also inhibited oestradiol-induced increases in endo- fulvestrant was administered as two separate 2.5 ml metrial volume in ovariectomised primates to injections in a double-blind comparison (North Amer- below that present at the start of the menstrual cycle ican). The results of a prospectively designed, combined (Dukes et al. 1993). Moreover, the rate and extent of analysis of these studies are presented here. endometrial involution were similar to that observed Most patients in the fulvestrant and anastrozole following oestradiol withdrawal, demonstrating a lack groups had progressed following previous treatment of oestrogen-agonist effects (Dukes et al. 1993). In with tamoxifen (96 vs 97% respectively), and both both of these animal models, fulvestrant was not groups were generally well matched at baseline for associated with the uterine hyperplasia often observed age, weight, prior therapy, hormone receptor status and with tamoxifen treatment. extent of metastatic or recurrent disease (Robertson These preclinical data have been supported by data et al. 2003). At median follow-up of 15.1 months, the from a study in healthy volunteers. In a phase I trial, 30 median time to progression (TTP) (Fig. 3)was healthy postmenopausal women received a single dose comparable in both groups (5.5 vs 4.1 months for i.m. 125 mg or 250 mg fulvestrant or placebo, followed fulvestrant and anastrozole respectively; hazard ratio 2 weeks later by 20 mg/day ethinyloestradiol for 2 weeks (HR) 0.95; 95.14% confidence interval (CI) 0.82, 1.10; (Addo et al. 2002). No evidence of agonist activity in the PZ0.48). The objective response (OR; complete endometrium was observed. Moreover, 250 mg fulves- response (CR) C partial response (PR)) rate was not trant abrogated the mean change in oestrogen-stimu- significantly different between the two groups, being lated endometrial thickening after 21 days of treatment (1.5 mm vs 8.1 mm; P!0.001) (Addo et al. 2002). 19.2% in the fulvestrant group compared with 16.5% in Thus, fulvestrant lacks oestrogen agonist activity on the normal human postmenopausal endometrium.

Clinical efficacy of pure oestrogen antagonists In tamoxifen-resistant advanced breast cancer Preclinical data suggesting that fulvestrant may be beneficial in the treatment of tamoxifen-resistant tumours (Hu et al. 1993, Osborne et al. 1994)have been supported by the efficacy of fulvestrant in patients Figure 3 Kaplan–Meier plot for time to progression (TTP) in with this type of breast cancer. In a small, initial clinical patients who had previously progressed on endocrine anti- study (nZ19), over two-thirds (69%) of postmenopausal oestrogen therapy: fulvestrant vs anastrozole in advanced women with tamoxifen-resistant disease treated with breast cancer. Robertson JF, Osborne CK, Howell A, Jones SE, Mauriac L, Ellis M, Kleeberg UR, Come SE, Vergote I, Gertler S fulvestrant experienced CB (Howell et al. 1996). et al. 2003 Fulvestrant versus anastrozole for the treatment of Moreover, a long duration of response (DoR) was advanced breast carcinoma in postmenopausal women: a observed in these women (median duration 25 months) prospective combined analysis of two multicenter trials. Cancer 98 229–238. Copyright q 2003 American Cancer Society. (Howell & Robertson 1995, Howell et al. 1996), Reprinted by permission of Wiley-Liss, Inc., a subsidiary of John supporting preclinical evidence that fulvestrant Wiley & Sons, Inc.

Downloaded from Bioscientifica.com at 09/27/2021 10:26:15PM via free access www.endocrinology-journals.org 695 A Howell: Pure anti-oestrogens in advanced breast cancer the anastrozole group (PZ0.31). In addition, there As first-line therapy for advanced breast cancer were no differences in OR rates between fulvestrant The efficacy of first-line fulvestrant vs tamoxifen has and anastrozole in the subgroup of patients who had been investigated in postmenopausal women with any visceral metastases (15.7 vs 13.2% respectively; metastatic or locally advanced breast cancer. In a Z P 0.49) or those with visceral metastases only (18.8 phase III study, endocrine-naı¨ve patients or patients Z vs 14.0% respectively; P 0.43) (Mauriac et al. 2003). who had completed endocrine therapy R1year Furthermore, fulvestrant demonstrated similar efficacy previously were treated with either fulvestrant in patients with/without visceral metastases. (250 mg/month i.m. injection with placebo tamoxifen, In patients who had an OR, further follow-up was nZ313) or tamoxifen (20 mg/day orally with placebo performed at a median of 22.1 months (Robertson et al. fulvestrant, nZ274) (Howell et al. 2004). Approxi- 2003). The median DoR was 16.7 and 13.7 months in mately 20–25% of patients had received prior adjuvant Z those patients who responded to fulvestrant (n 84) and tamoxifen therapy for their primary breast cancer. Z anastrozole (n 73) respectively (Fig. 4); mean After a median follow-up of 14.5 months, there was duration of response was significantly greater for no significant difference between fulvestrant and fulvestrant than anastrozole (HR 1.30; 95% CI 1.13, tamoxifen for the primary endpoint TTP (median 6.8 ! 1.50; P 0.01) (Robertson et al. 2003). A similar vs 8.3 months respectively; HR 1.18; 95% CI 0.98, C proportion of patients experienced CB (OR stable 1.44; PZ0.088; Table 1). OR rates were similar R disease (SD) for 24 weeks; 43.5% patients receiving between the two arms (31.6% for fulvestrant vs 33.9% fulvestrant and 40.9% patients receiving anastrozole) for tamoxifen); however, CB rates were significantly and the median duration of CB was also similar between higher in the tamoxifen group (54.3% for fulvestrant vs the groups (11.8 vs 11.2 months respectively). 62.0% for tamoxifen; PZ0.026). In patients experienc- A survival analysis conducted at a median follow-up ing an OR, the median DoR was similar (17.3 vs 19.8 of 27.0 months showed that the overall survival was months with fulvestrant and tamoxifen respectively) very similar with fulvestrant and anastrozole (median (Howell et al. 2004). Thus, in the overall (intent-to- 27.4 vs 27.7 months respectively; HR 0.98; 95% CI treat (ITT)) population between-group differences in Z 0.84, 1.15; P 0.809), and that three-quarters of the efficacy favoured tamoxifen and the statistical non- patients had died in each group (74.5 vs 76.1% inferiority of fulvestrant could not be demonstrated. respectively) (Howell et al. 2005). Indeed, as the upper 95% CI was below the predefined limit of 1.25, fulvestrant may be considered non-inferior to anastro- Table 1 Impact of hormone receptor status on time to zole in terms of overall survival benefit. progression with fulvestrant vs tamoxifen in patients with advanced breast cancer (Howell A, Robertson JFR, Abram P, Lichinister MR, Elledge R, Bajetta E, Watanabe T, Morris C, Webster A, Dimery I et al. 2004 Comparison of fulvestrant vs tamoxifen for the treatment of advanced breast cancer in postmenopausal women previously untreated with endocrine therapy: a multi national, double blind, randomized trial. Journal of Clinical Oncology 22 1605–1613. Reprinted with permission from the American Society of Clinical Oncology.)

Patient population n (%) HR 95% CI P-value

ITT Fulvestrant 313 (100) 1.18 0.98, 1.44 0.088 Tamoxifen 274 (100) ERC and/or PgRC Fulvestrant 247 (78.9) 1.10 0.89, 1.36 0.39 Figure 4 Kaplan–Meier plot for duration of response (DoR) from Tamoxifen 212 (77.4) randomisation to disease progression in responding patients: ERC and PRC fulvestrant vs anastrozole in advanced breast cancer. Robertson Fulvestrant 131 (41.9) 0.85 0.63, 1.15* 0.31 JF, Osborne CK, Howell A, Jones SE, MauriacL, Ellis M, Kleeberg Tamoxifen 114 (41.6) UR, Come SE, Vergote I, Gertler S et al. 2003 Fulvestrant versus anastrozole for the treatment of advanced breast carcinoma in HR, hazard ratio; CI, confidence interval; ITT, intent-to-treat; postmenopausal women: a prospective combined analysis of two ERC, oestrogen receptor positive; PgRC, progesterone multicenter trials. Cancer 98 229–238. Copyright q 2003 receptor positive. American Cancer Society. Reprinted by permission of Wiley-Liss, *Non-inferiority of fulvestrant to tamoxifen is shown by an upper Inc., a subsidiary of John Wiley & Sons, Inc. 95% CI of %1.25.

Downloaded from Bioscientifica.com at 09/27/2021 10:26:15PM via free access 696 www.endocrinology-journals.org Endocrine-Related Cancer (2006) 13 689–706

The ITT population contained O20% of patients for have received multiple prior therapies (endocrine or whom the ER status was negative or unknown. Given chemotherapy). For patients who were not eligible the modes of action of tamoxifen and fulvestrant, to receive fulvestrant as part of a clinical trial and in patients with hormone receptor-positive breast tumours countries where fulvestrant was not yet approved, may be expected to gain the greatest benefit from these fulvestrant was available as part of a compassionate- drugs. In a prospectively planned analysis of those use programme (Franco et al. 2004, Petruzelka et al. patients with ER-positive and/or PgR-positive breast 2004, Steger et al. 2005). tumours (O75% of patients), median TTP was similar In the largest report from a single centre, data are between fulvestrant and tamoxifen (8.2 vs 8.3 months available from 126 patients (80% had received respectively; HR 1.10; 95% CI 0.89, 1.36; PZ0.39; fulvestrant as second- or third-line endocrine therapy Table 1, Fig. 5)(Howell et al. 2004). Furthermore, and 60% had received palliative chemotherapy). Of similar rates for CB (57.1 vs 62.7%; HR 0.79; 95% CI – these women, 12 (9.5%) had a PR and 43 (34%) had 15.01, 3.19; PZ0.22) and OR (33.2 vs 31.1%; HR 1.10; SD R6 months with fulvestrant treatment resulting in 95% CI 0.74, 1.63; PZ0.64) were observed in the a CB rate of 44% (Steger et al. 2005). Notably, 43/108 fulvestrant and tamoxifen groups respectively. There- patients (40%) who had received prior non-steroidal fore, in patients with hormone receptor-positive AI treatment gained CB with fulvestrant. Breast tumours, fulvestrant and tamoxifen had similar efficacy. tumours were ER-positive and PgR-positive in 9/12 In an additional post-hoc analysis of patients with patients experiencing a PR and 9/12 had previously breast tumours that were positive for both ER and PgR received an AI. Of the 43 patients with SD R6 (w42% of patients), the findings for median TTP were months in duration, 26 had ER-positive and PgR- again shown to be comparable in the two groups (11.4 vs positive tumours. Among 18 patients with HER2- 8.5 months for fulvestrant and tamoxifen respectively; HR positive tumours, nine patients experienced SD, 0.85; 95% CI 0.63, 1.15; PZ0.31; Table 1)(Buzdar 2004). supporting the preclinical observation that fulvestrant Further studies of first-line fulvestrant are warranted, can abrogate ER activation mediated by EGFR specifically in patients with confirmed hormone receptor- (Steger et al. 2005). positive breast tumours, as these patients represent Data on the compassionate use of fulvestrant are also the intended treatment population for the drug. available from a more heavily pretreated cohort of 42 patients (50% had received R4 prior endocrine R Experience from a compassionate-use therapies and 38% had received 4 chemotherapy programme regimens) (Franco et al. 2004). All patients from this centre had received prior AI therapy. Despite the failure Treatment options are severely limited in postmeno- of various previous treatment options, eight patients pausal women with advanced breast cancer who (19%) experienced SD for R24 weeks; five had ER-positive and PgR-positive tumours and three had ER-positive and PgR-negative tumours. Three patients (two with ER-positive and PgR-negative tumours) had experienced SD for 14, 20 and 23C months at the time data became available (Franco et al. 2004). A further report has described an additional cohort of 64 pretreated patients (78% had received fulvestrant as second- or third-line endocrine therapy and 64% had received prior chemotherapy) (Petruzelka et al. 2004). In total, 39 patients (61%) experienced CB (CR C PR C SD R24 weeks), including five patients (8%) with Figure 5 Kaplan–Meier plot for time to progression (TTP) in an OR. A durable and ongoing OR was observed in patients with ER-positive and/or PgR-positive tumours: fulves- 3/44 patients (R18.5 to R21.2 months in duration), trant vs tamoxifen in advanced breast cancer. Howell A, and 18/44 patients experienced ongoing SD of R24 Robertson JF, Abram P, Lichinitser MR, Elledge R, Bajetta R, R R Watanabe T, Morris C, Webster A, Dimery I & Osborne CK weeks ( 6.5 to 20.3 months). Of the five patients 2004 Comparison of fulvestrant versus tamoxifen for the with an OR, three had ER-positive and PgR-positive treatment of advanced breast cancer in postmenopausal tumours and all had received prior non-steroidal AI women previously untreated with endocrine therapy: a multi- national, double-blind, randomised trial. Journal of Clinical treatment (Petruzelka et al. 2004). Oncology 22 1605–1613. Reprinted with permission from the These data provide further evidence for the lack of American Society of Clinical Oncology. cross-resistance of fulvestrant with other endocrine

Downloaded from Bioscientifica.com at 09/27/2021 10:26:15PM via free access www.endocrinology-journals.org 697 A Howell: Pure anti-oestrogens in advanced breast cancer agents, including the AIs, and confirm that fulvestrant injection-site reactions occurred in 1.1% of courses in can provide CB even in heavily pretreated postmeno- patients given a single 5 ml injection, and in 4.6% and pausal women with advanced breast cancer. Moreover, 4.4% of courses in patients given two 2.5 ml there was evidence to suggest that the use of fulvestrant fulvestrant or placebo injections respectively. Only earlier in the treatment sequence led to better two patients (fulvestrant group) had injection-site responses. In all of these compassionate-use analyses, reactions leading to withdrawal. Among patients fulvestrant was very well tolerated. receiving either fulvestrant or anastrozole, few patients withdrew due to AEs (2.8 vs 1.9% respectively) or drug-related AEs (0.9 vs 1.2% respectively). At long- Tolerability of pure oestrogen antagonists term follow-up (median 27.0 months), no safety Owing to its pure oestrogen antagonistic effects, concerns had arisen in either treatment group (Howell fulvestrant has the potential to offer an improved et al. 2005). tolerability profile in the treatment of postmenopausal In the study comparing fulvestrant and tamoxifen, women with advanced breast cancer compared with the safety population comprised 310 and 271 patients, other endocrine agents that have less targeted efficacy. respectively, and the median follow-up was 14.5 The safety and tolerability findings of phase III studies months (Howell et al. 2004). The most commonly of fulvestrant, both as first-line therapy and in patients reported AEs in the fulvestrant and tamoxifen groups with tamoxifen-resistant disease, are described below. respectively, were nausea (20.3 vs 22.5%), asthenia In the combined analysis of the two phase III trials of (19.4 vs 20.3%), vasodilation (14.8 vs 21.4%), pain patients undergoing second-line treatment with fulves- (13.9 vs 19.2%) and bone pain (13.9 vs 17.0%). Four trant and anastrozole, the safety population comprised predefined AEs (gastrointestinal disturbances, hot 423 patients in each group (Robertson et al. 2003). flushes, thromboembolic disease and vaginitis) were Adverse events (AEs) were generally mild to moderate selected for statistical evaluation (Table 3). Of these, in intensity. The most commonly reported AEs with the incidence of hot flushes approached significance fulvestrant and anastrozole respectively were nausea between the two groups, being lower among patients (26.0 vs 25.3%), asthenia (22.7 vs 27.0%), pain (18.9 treated with fulvestrant (17.7 vs 24.7% with tamoxifen; vs 20.3%), vasodilation (17.7 vs 17.3%) and headache PZ0.05). There were also fewer gastrointestinal (15.4 vs 16.8%). Seven predefined AEs were selected disturbances in the fulvestrant-treated patients (37.1 for statistical evaluation: gastrointestinal disturbances, vs 43.2% in the tamoxifen group), but this difference hot flushes, joint disorders, thromboembolic disease, was not significant (PZ0.16). The incidence of urinary tract infection, vaginitis and weight gain thromboembolic events was higher in patients receiv- (Table 2). Of these, joint disorders was the only AE ing fulvestrant in this study (5.8 vs 3.3%), but not that was significantly different between the groups, significantly so (PZ0.22). This apparent increase in experienced by fewer patients receiving fulvestrant thromboembolic events was not noted in the other (5.4 vs 10.6% with anastrozole; PZ0.0036). Local phase III trials of fulvestrant (Howell et al. 2002,

Table 2 Incidence of prospectively defined adverse events in patients receiving fulvestrant or anastrozole as second-line therapy. Robertson JF, Osborne CK, Howell A, Jones SE, Mauriac L, Ellis M, Kleeberg UR, Come SE, Vergote I, Gertler S et al. 2003 Fulvestrant versus anastrozole for the treatment of advanced breast carcinoma in postmenopausal women: a prospective combined analysis of two multicenter trials. Cancer 98 229–238. Copyright q 2003 American Cancer Society. Reprinted by permission of Wiley-Liss, Inc., a subsidiary of John Wiley & Sons, Inc

Fulvestrant (nZ423) Anastrozole (nZ423)

Adverse event n % n % P-value

Gastrointestinal disturbances* 196 46.3 185 43.7 0.53 Hot flushes 89 21.0 87 20.6 0.91 Joint disorders† 23 5.4 45 10.6 0.036 Thromboembolic disease 15 3.5 17 4.0 0.68 Urinary tract infection 31 7.3 18 4.3 0.06 Vaginitis 11 2.6 8 1.9 0.51 Weight gain 4 0.9 7 1.7 0.35

*Includes patients with nausea, vomiting, constipation and haemorrhage. †Includes patients with arthralgia, arthrosis and arthritis.

Downloaded from Bioscientifica.com at 09/27/2021 10:26:15PM via free access 698 www.endocrinology-journals.org Endocrine-Related Cancer (2006) 13 689–706

Table 3 Incidence of prospectively defined adverse events in patients receiving fulvestrant or tamoxifen as first-line therapy. Howell A, Robertson JF, Abram P, Lichinitser MR, Elledge R, Bajetta R, Watanabe T, Morris C, Webster A, Dimery I & Osborne CK 2004 Comparison of fulvestrant versus tamoxifen for the treatment of advanced breast cancer in postmenopausal women previously untreated with endocrine therapy: a multinational, double-blind, randomised trial. Journal of Clinical Oncology 22 1605–1613. Reprinted with permission from the American Society of Clinical Oncology

Fulvestrant (nZ310) Tamoxifen (nZ271)

Adverse event n % n % P-value

Gastrointestinal disturbances* 115 37.1 117 432 0.16 Hot flushes 55 17.7 67 24.7 0.05 Vaginitis 12 3.9 17 6.3 0.26 Thromboembolic disease 18 5.8 9 3.3 0.22

*Includes patients with nausea, vomiting, constipation and haemorrhage.

Osborne et al. 2002) and no thromboembolic events turnover in this patient group (Donnez et al. 2003). were reported in two recent phase II studies (Ingle et al. However, the high circulating oestrogen levels in 2004, 2006, Perey et al. 2004). Three patients receiving premenopausal women may have negated any impact fulvestrant withdrew because of a drug-related AE of fulvestrant on bone turnover in this study. Further (cerebrovascular accident, deep vein thrombosis and studies are required to assess any impact on bone in pulmonary embolus (one patient each)). No AEs postmenopausal women. leading to withdrawal were considered to be drug related in the tamoxifen group (Howell et al. 2004). Future directions It is interesting to note that many so-called endocrine treatment-related AEs have also been noted in patients The sequential use of endocrine agents is a standard receiving placebo in breast cancer trials. For example, treatment strategy for advanced breast cancer. How- in the NCIC CTG MA.17 trial, postmenopausal ever, as non-steroidal AIs are now replacing tamoxifen patients receiving following 5 years of as the first-line choice, the optimal sequence of tamoxifen treatment experienced a similar frequency endocrine therapy needs to be re-evaluated in order of oedema (17.2 vs 15.6%), fatigue (29.9 vs 28.3%), to maximise the benefit that patients will gain from sweating (22.1 vs 20.7%), and headache (18.1 vs these treatments. While direct comparisons between endocrine therapies are not always available, indirect 18.6%) as placebo-treated patients (Goss et al. 2003). comparisons suggest that fulvestrant offers comparable Therefore, it is difficult to determine which AEs are efficacy and may also have tolerability advantages over truly fulvestrant-related without the use of placebo- some AIs in the second-line treatment of postmeno- controlled studies. pausal women with advanced breast cancer (Dodwell The effects of fulvestrant on bone mineral density & Vergote 2005). In the treatment of advanced breast and markers of bone turnover are not fully elucidated, cancer, third-generation AIs (e.g. anastrozole, letrozole and have not yet been reported in postmenopausal and ) have superior efficacy to tamoxifen women. Preclinical studies in rats suggest that and have become the first-line therapy of choice in this fulvestrant reduces cancellous bone volume, which disease setting (Nabholtz et al. 2000, Mouridsen et al. comprises only a small proportion of total bone 2003, Paridaens et al. 2003). Clearly, the most effective (Gallagher et al. 1993, Sibonga et al. 1998) and its endocrine therapy should always be used first and, in effects are consistent with blockade of ER-mediated cases where therapies are equally effective, the best- bone resorption and formation (Wakeling 1995). tolerated agent should be used first. A second key Despite the lack of uterotrophic activity with consideration is that cross-resistance is less likely to fulvestrant, some agonistic effects on bone were occur if endocrine therapies with different modes of apparent in the second study, which were highly action are used. Finally, tumour sensitivity to sub- dependent on circulating oestrogen levels (Sibonga sequent endocrine therapies should be maintained. et al. 1998). In a study involving 307 premenopausal Two large phase III trials have provided evidence that women with uterine fibroids awaiting hysterectomy, tamoxifen-resistant tumours remain sensitive to fulves- fulvestrant did not produce changes in markers of bone trant (Howell et al. 2002, Osborne et al. 2002). resorption (cross-linked N-telopeptides and free Furthermore, a phase II trial has shown that of 67 deoxypyridinoline), suggesting it did not affect bone patients who developed resistance initially to tamoxifen

Downloaded from Bioscientifica.com at 09/27/2021 10:26:15PM via free access www.endocrinology-journals.org 699 A Howell: Pure anti-oestrogens in advanced breast cancer and then to an AI, 28.4% subsequently gained CB (CR combination therapies, including those in which C PR C SD R24 weeks) with fulvestrant (median fulvestrant is co-administered with anastrozole (vs treatment duration 3.8 months) (Perey et al. 2004). anastrozole alone in SOFEA and FACT) and the novel Similarly, in another phase II trial of 77 patients with anti-epidermal growth factor receptor EGFR/HER2 advanced breast cancer who had progressed on prior AI agents gefitinib (ECOG 4101) and trastuzumab. therapy (21 had also received tamoxifen), 32.5% gained CB with fulvestrant (Ingle et al. 2004, 2006). Additionally, in the compassionate-use programme, Other pure oestrogen antagonists CB was achieved in patients who received fulvestrant in development after progression on prior endocrine therapies, includ- Although fulvestrant is the first pure oestrogen ing both AIs and SERMs (Petruzelka et al. 2004, Steger antagonist, several other pure oestrogen antagonists et al. 2005). Importantly, tumour sensitivity to are undergoing preclinical development. Two new subsequent endocrine therapies appears to be main- pure oestrogen antagonists, ZK-703 and ZK-253, tained after fulvestrant treatment (Vergote et al. 2003, which destabilise the ER, have been investigated Robertson et al. 2005). using the MCF-7 xenograft model. ZK-703 and ZK- Taken together, these data position fulvestrant as a 253 were not only more effective than either tamoxifen versatile agent that may be used in the second-line setting or fulvestrant at inhibiting the growth of ER-positive after tamoxifen, or potentially after AIs. However, xenografts, they also showed a highly potent activity initiating fulvestrant earlier in the treatment sequence in tamoxifen-resistant xenografts (Hoffmann et al. may lead to better and more durable responses. 2004). Clinical studies with both of these agents are Fulvestrant is now approved for the treatment of awaited with interest. (FC-1271a), a postmenopausal women with ER-positive, locally SERM being developed for the treatment of osteo- advanced or metastatic breast cancer who porosis, has been observed to inhibit MCF-7 tumour have either relapsed on prior adjuvant oestrogen growth in vivo when compared with control (P % 0.05; antagonist therapy or progressed on prior oestrogen Taras et al. 2001), and further studies are required to antagonist therapies. After progression on fulvestrant, evaluate its efficacy in the clinical setting. Another endocrine therapies such as or the steroidal oestrogen antagonist that has undergone steroidal AI exemestane may be the next appropriate preclinical investigation with promising results is RU drug. 58668, which has been shown to induce long-term Several new trials of fulvestrant are now under way regression of MCF-7 xenografts (van de Velde et al. to further validate its optimum position in the 1994). These data suggest that RU 58668 may have endocrine treatment sequence for advanced breast potential for the treatment of advanced breast cancer. cancer. The Evaluation of Faslodex and Exemestane To date, however, there do not appear to be any clinical Clinical Trial (EFECT) includes postmenopausal trials that are evaluating RU 58668 in the breast cancer patients with ER-positive and/or PgR-positive breast setting. tumours who have experienced recurrence or pro- Two non-steroidal fourth-generation SERMs that gression on a non-steroidal AI. Patients will be exert complete oestrogen antagonistic effects on the randomised for double-dummy treatment with fulves- breast and uterus have been developed– trant (loading dose: 500 mg i.m. on day 1 and 250 mg (EM-652) and its highly potent precursor EM-800 on day 14, plus a 250 mg on day 28 and every (Gauthier et al. 1997, Labrie et al. 1999). In a month thereafter) or exemestane (25 mg/day orally). preclinical xenograft (ZR-75-1) study, EM-652 caused The primary endpoint of this study is TTP. In a second the complete disappearance of 65% of tumours and no post-AI trial (Study Of Faslodex vs Exemestane tumours progressed (Roy et al. 2003). Moreover, 93% with/without Arimidex (SOFEA)), postmenopausal of tumours that had disappeared using EM-652 did not patients with locally advanced or metastatic breast reappear when exposed to oestrogen challenge for 12 cancer (ER positive) who have recurred/progressed on weeks (Roy et al. 2003). Development of EM-652 is treatment with non-steroidal AIs will be randomised to ongoing. The development of EM-800 for advanced treatment with fulvestrant, fulvestrant plus anastrozole, breast cancer has been halted, despite 35% of patients or exemestane. In this study, as well as in EFECT and (nZ43) with tamoxifen-resistant breast cancer who Faslodex and Arimidex Clinical Trial (FACT), the time had been treated with EM-800 experiencing CB in a to steady state of fulvestrant will be examined more phase II study (Labrie et al. 2004). In comparison, closely in order to optimise loading-dose regimens. O43.5% of tamoxifen-resistant patients treated with Ongoing and planned studies will also examine fulvestrant experience CB (Robertson et al. 2003).

Downloaded from Bioscientifica.com at 09/27/2021 10:26:15PM via free access 700 www.endocrinology-journals.org Endocrine-Related Cancer (2006) 13 689–706

TAS-108 is a synthetic ER ligand that blocks ER-a the receptor of [3H]4-hydroxytamoxifen, an transactivation but has an agonistic effect on ER-b active metabolite. Molecular and Cellular (Yamamoto et al. 2005). In a phase I study in 16 Endocrinology 20 71–85. patients who had received 6 to 7 prior therapies, Bowler J, Lilley TJ, Pittam JD & Wakeling AE 1989 Novel pharmacokinetic findings were favourable and there steroidal pure . 54 71–99. was evidence of biological anti-tumour activity Brzozowski AM, Pike AC, Dauter Z, Hubbard RE, Bonn T, Engstrom O, Ohman L, Greene GL, Gustafsson JA & (Blakely et al. 2004). Phase II studies involving Carlquist M 1997 Molecular basis of agonism and TAS-108 are planned in the USA. antagonism in the oestrogen receptor. Nature 389 753–758. Conclusions Buzdar AU 2004 Fulvestrant: a new type of antagonist for the treatment of advanced breast cancer. There is a clear need for the development of oestrogen Drugs Today (Barc) 40 751–764. antagonistic drugs that match the clinical efficacy of Buzdar AU, Marcus C & Holmes F 1988 Phase II evaluation tamoxifen but lack any partial agonist effects. of LY156758 in metastatic breast cancer. Oncology 45 Fulvestrant is the first ‘pure’ oestrogen antagonist to 344–345. be extensively characterised and to enter clinical use. Buzdar A, Hayes D, El-Khoudary A, Yan S, Lønning P, In postmenopausal women with advanced breast Lichinitser M, Gopal R, Falkson G, Pritchard K, Lipton A cancer, fulvestrant represents a valuable and versatile et al. 2002 Phase III randomized trial of droloxifene and new option for endocrine therapy. tamoxifen as first-line endocrine treatment of ER/PgR- positive advanced breast cancer. Breast Cancer Research and Treatment 73 161–175. Acknowledgements Buzdar A, O’Shaughnessy JA, Booser DJ, Pippen JE Jr, Jones SE, Munster PN, Peterson P, Melemed AS, Winer E & Hudis C The author would like to thank Carolyn Gray, PhD, for 2003 Phase II, randomized, double-blind study of two dose her editorial assistance with this manuscript. levels of arzoxifene in patients with locally advanced or metastatic breast cancer. Journal of Clinical Oncology 21 Funding 1007–1014. Cheskis BJ, Karathanasis S & Lyttle CR 1997 Estrogen The author declares that he has no conflicts of interest receptor ligands modulate its interaction with DNA. that would prejudice the impartiality of this article. Journal of Biological Chemistry 272 11384–11391. Chinenov Y & Kerppola TK 2001 Close encounters of the many kinds: Fos:Jun interactions that mediate References transcription regulatory specificity. Oncogene 20 Addo S, Yates RA & Laight A 2002 A phase I trial to assess 2438–2452. the pharmacology of the new oestrogen receptor Cotreau MM, Stonis L, Dykstra KH, Gandhi T, Gutierrez M, antagonist fulvestrant on the endometrium in healthy Xu J, Park Y, Burghart PH & Schwertschlag US 2002 postmenopausal volunteers. British Journal of Cancer 87 Multiple-dose, safety, , and pharmaco- 1354–1359. dynamics of a new selective estrogen receptor modulator, Bardin A, Boulle N, Lazennec G, Vignon F & Pujol P 2004 ERA-923, in healthy postmenopausal women. Journal of Loss of ERb expression as a common step in estrogen- Clinical Pharmacology 42 157–165. dependent tumor progression. Endocrine-Related Cancer Cuzick J, Powles T, Veronesi U, Forbes J, Edwards R, 11 537–551. Ashley S & Boyle P 2003 Overview of the main Beato M 1989 Gene regulation by hormones. Cell 56 outcomes in breast-cancer prevention trials. Lancet 335–344. 361 296–300. Berry M, Metzger D & Chambon P 1990 Role of the two Dauvois S, Danielian PS, White R & Parker MG 1992 activating domains of the oestrogen receptor in the Antiestrogen ICI 164,384 reduces cellular estrogen cell-type and promoter-context dependent agonistic receptor content by increasing its turnover. PNAS 89 activity of the anti-oestrogen 4-hydroxytamoxifen. 4037–4041. EMBO Journal 9 2811–2818. Dauvois S, White R & Parker MG 1993 The antiestrogen Blakely LJ, Buzdar A, Chang HY, Frye D, Theriault R, ICI 182780 disrupts estrogen receptor nucleocytoplas- Valero V, Rivera E, Booser D, Kuritani J & Tsuda M 2004 mic shuttling. Journal of Cell Science 106 1377–1388. A phase I and pharmacokinetic study of TAS-108 in DeFriend DJ, Howell A, Nicholson RI, Anderson E, Dowsett postmenopausal female patients with locally advanced, M, Mansel RE, Blamey RW, Bundred NJ, Robertson JF, locally recurrent inoperable, or progressive metastatic Saunders C et al. 1994 Investigation of a new pure breast cancer. Clinical Cancer Research 10 5425–5431. antiestrogen (ICI 182,780) in women with primary breast Borgna JL & Rochefort H 1980 High-affinity binding to cancer. Cancer Research 54 408–414.

Downloaded from Bioscientifica.com at 09/27/2021 10:26:15PM via free access www.endocrinology-journals.org 701 A Howell: Pure anti-oestrogens in advanced breast cancer

Delmas PD, Bjarnason NH, Mitlak BH, Ravoux AC, Shah negative lymph nodes and estrogen receptor-positive AS, Huster WJ, Draper M & Christiansen C 1997 Effects tumors. Journal of the National Cancer Institute 88 of raloxifene on bone mineral density, serum cholesterol 1529–1542. concentrations, and uterine endometrium in postmeno- Franco S, Perez A, Tan-Chiu E, Frankel C & Vogel C 2004 pausal women. New England Journal of Medicine 337 Response to fulvestrant in postmenopausal women 1641–1647. heavily pretreated with prior hormonal therapy: a single- Dodwell D & Vergote I 2005 A comparison of fulvestrant and center experience. Breast Cancer Research and Treat- third-generation aromatase inhibitors in the second-line ment 88 103–108. treatment of postmenopausal women with advanced Frasor J, Stossi F, Danes JM, Komm B, Lyttle R & breast cancer. Cancer Treatment Reviews 31 274–282. Katzenellenbogen BS 2004 Selective estrogen receptor Donnez J, Hervais Vivancos B, Kudela M, Audebert A & modulators: discrimination of agonistic versus antagon- Jadoul P 2003 A randomized, placebo-controlled, dose- istic activities by gene expression profiling in breast ranging trial comparing fulvestrant with in cancer cells. Cancer Research 64 1522–1533. premenopausal patients with uterine fibroids awaiting Gallagher A, Chambers TJ & Tobias JH 1993 The estrogen hysterectomy. Fertility and Sterility 79 1380–1389. antagonist ICI 182,780 reduces cancellous bone volume Dukes M, Waterton JC & Wakeling AE 1993 Antiutero- in female rats. Endocrinology 133 2787–2791. trophic effects of the pure antioestrogen ICI 182,780 in Gauthier S, Caron B, Cloutier J, Dory YL, Favre A, Larouche adult female monkeys (Macaca nemestrina): quantitative D, Mailhot J, Ouellet C, Schwerdtfeger A, Leblanc G et al. magnetic resonance imaging. Journal of Endocrinology 1997 (S)-(C)-4-[7-(2,2-dimethyl-1-oxopropoxy)-4- 138 203–210. methyl-2-[4-[2-(1-piperidinyl)- ethoxy]phenyl]-2H-1- Early Breast Cancer Trialists’ Collaborative Group 1998 benzopyran-3-yl]-phenyl 2,2-dimethylpropanoate Tamoxifen for early breast cancer: an overview of the (EM-800): a highly potent, specific, and orally active randomised trials. Lancet 351 1451–1467 antiestrogen. Journal of Medicinal Chemistry Evans RM 1988 The steroid and thyroid hormone receptor 40 2117–2122. superfamily. Science 240 889–895. Gershanovich M, Garin A, Baltina D, Kurvet A, Kangas L & Fawell SE, White R, Hoare S, Sydenham M, Page M & Ellmen J 1997 A phase III comparison of two toremifene Parker MG 1990 Inhibition of estrogen receptor-DNA doses to tamoxifen in postmenopausal women with binding by the “pure” antiestrogen ICI 164,384 appears to advanced breast cancer. Eastern European Study Group. be mediated by impaired receptor dimerization. Pro- Breast Cancer Research and Treatment 45 251–262. ceedings of the National Academy of Sciences USA 87 Goss PE, Ingle JN, Martino S, Robert NJ, Muss HB, Piccart 6883–6887. MJ, Castiglione M, Tu D, Shepherd LE, Pritchard KI et al. Filardo EJ & Thomas P 2005 GPR30: a seven-trans- 2003 A randomized trial of letrozole in postmenopausal membrane-spanning estrogen receptor that triggers EGF women after five years of tamoxifen therapy for early- release. Trends in Endocrinology and Metabolism 16 stage breast cancer. The New England Journal of 362–367. Medicine 349 1793–1802. Filardo EJ, Quinn JA, Bland KI & Frackelton AR Jr 2000 Gradishar WJ, Glusman J, Lu Y, Vogel C, Cohen FJ & Estrogen-induced activation of ERk-1 and ERk-2 requires Sledge GW Jr 2000 Effects of high dose raloxifene in the G protein-coupled receptor homolog, GPR30, and selected patients with advanced breast carcinoma. Cancer occurs via trans-activation of the epidermal growth factor 88 2047–2053. receptor through release of EGF. Molecular Endocrin- Gustafsson JA & Warner M 2000 Estrogen receptor b in the ology 14 1649–1660. breast: role in estrogen responsiveness and development Filardo EJ, Quinn JA, Frackelton AR Jr & Bland KI 2002 of breast cancer. Journal of Steroid Biochemistry and Estrogen action via the G protein-coupled receptor, Molecular Biology 74 245–248. GPR30: stimulation of adenylyl cyclase and cAMP- Haarstad H, Gundersen S, Wist E, Raabe N, Mella O & mediated attenuation of the epidermal growth factor Kvinnsland S 1992 Droloxifene – a new anti-estrogen. A receptor-to-MAPK signaling axis. Molecular Endocrin- phase II study in advanced breast cancer. Acta Oncologica ology 16 70–84. 31 425–428. Fisher B, Costantino JP, Redmond CK, Fisher ER, Hanstein B, Djahansouzi S, Dall P, Beckmann MW & Bender Wickerham DL & Cronin WM 1994 Endometrial cancer HG 2004 Insights into the molecular biology of the estrogen in tamoxifen-treated breast cancer patients: findings from receptor define novel therapeutic targets for breast cancer. the National Surgical Adjuvant Breast and Bowel Project European Journal of Endocrinology 150 243–255. (NSABP) B-14. Journal of the National Cancer Institute Hayes DF, Van Zyl JA, Hacking A, Goedhals L, Bezwoda 86 527–537. WR, Mailliard JA, Jones SE, Vogel CL, Berris RF, Fisher B, Dignam J, Bryant J, DeCillis A, Wickerham DL, Shemano I et al. 1995 Randomized comparison of Wolmark N, Costantino J, Redmond C, Fisher ER, tamoxifen and two separate doses of toremifene in Bowman DM et al. 1996 Five versus more than five years postmenopausal patients with metastatic breast cancer. of tamoxifen therapy for breast cancer patients with Journal of Clinical Oncology 13 2556–2566.

Downloaded from Bioscientifica.com at 09/27/2021 10:26:15PM via free access 702 www.endocrinology-journals.org Endocrine-Related Cancer (2006) 13 689–706

Hoffmann J, Bohlmann R, Heinrich N, Hofmeister H, Kroll J, Improta-Brears T, Whorton AR, Codazzi F, York JD, Meyer Kunzer H, Lichtner RB, Nishino Y, Parczyk K, Sauer G T & McDonnell DP 1999 Estrogen-induced activation of et al. 2004 Characterization of new estrogen receptor mitogen-activated protein kinase requires mobilization of destabilizing compounds: effects on estrogen-sensitive intracellular calcium. PNAS 96 4686–4691. and tamoxifen-resistant breast cancer. Journal of the Ingle JN, Rowland KM, Suman VJ, Mirchandani D, Bernath National Cancer Institute 96 210–218. AM, Camoriano JK & Perez EA 2004 Evaluation of Horwitz KB & McGuire WL 1978 Estrogen control of fulvestrant in women with advanced breast cancer and progesterone receptor and estrogen receptor in human progression on prior aromatase inhibitor therapy: a phase breast cancer. Journal of Biological Chemistry 253 II trial of the North Central Cancer Treatment Group. 2223–2238. Breast Cancer Research and Treatment 88 (Suppl 1) S38, Horwitz KB, Jackson TA, Bain DL, Richer JK, Takimoto GS abstract 409. & Tung L 1996 Nuclear receptor coactivators and Ingle JN, Suman VJ, Rowland KM, Mirchandani D, Bernath corepressors. Molecular Endocrinology 10 1167–1177. AM, Camoriano JK, Fishkin PA, Nikcevich DA & Perez Howell A & Robertson J 1995 Response to a specific EA 2006 Fulvestrant in women with advanced breast antioestrogen (ICI 182780) in tamoxifen-resistant breast cancer after progression on prior aromatase inhibitor cancer. Lancet 345 989–990. therapy. North American Cancer Treatment Group Trial Howell A, DeFriend DJ, Robertson JF, Blamey RW, No. 32. Journal of Clinical Oncology 24 1052–1056. Anderson L, Anderson E, Sutcliffe FA & Walton P 1996 Johnston SR, Gorbunova V, Lichinitser M, Manikas G, Pharmacokinetics, pharmacological and anti-tumour Koralewski P, Pluzanska A, Garin A, Harvey E & effects of the specific anti-oestrogen ICI 182780 in International Idoxifene Study Group 2001 A multicentre women with advanced breast cancer. British Journal of double-blind randomised phase III trial of idoxifene Cancer 74 300–308. versus tamoxifen as first-line endocrine therapy Howell A, Robertson JFR, Quaresma Albano J, Ascherman- for metastatic breast cancer. Proceedings of the nova A, Mauriac L, Kleeberg UR, Vergote I, Erikstein B, American Society of Clinical Oncology 20 29a, Webster A & Morris C 2002 Fulvestrant, formerly ICI abstract 113. 182,780, is as effective as anastrozole in postmenopausal Katzenellenbogen JA, O’Malley BW & Katzenellenbogen women with advanced breast cancer progressing after prior BS 1996 Tripartite steroid hormone receptor pharma- endocrine treatment. Journal of Clinical Oncology 20 cology: interaction with multiple effector sites as a basis 3396–3403. for the cell- and promoter-specific action of these Howell A, Robertson JFR, Abram P, Lichinitser MR, Elledge hormones. Molecular Endocrinology 10 119–131. R, Bajetta E, Watanabe T, Morris C, Webster A, Dimery I Kuiper G, Enmark E, Pelto-Huikko M, Nilsson MS & et al. 2004 Comparison of fulvestrant v tamoxifen for the Gustafsson JA 1996 Cloning of a novel estrogen receptor treatment of advanced breast cancer in postmenopausal expressed in rat prostate and ovary. PNAS 93 5925–5930. women previously untreated with endocrine therapy: a Kumar V & Chambon P 1988 The estrogen receptor binds multinational, double-blind, randomized trial. Journal of tightly to its responsive element as a ligand-induced Clinical Oncology 22 1605–1613. Cell 55 Howell A, Pippen J, Elledge RM, Mauriac L, Vergote I, Jones homodimer. 145–156. SE, Come SE, Osborne CK & Robertson JF 2005 Labrie F, Labrie C, Belanger A, Simard J, Gauthier S, Luu- Fulvestrant versus anastrozole for the treatment of The V, Merand Y, Giguere V, Candas B, Luo S et al. 1999 advanced breast carcinoma: a prospectively planned EM-652 (SCH 57068), a third generation SERM acting as combined survival analysis of two multicenter trials. pure antiestrogen in the mammary gland and endome- Cancer 104 236–239. trium. Journal of Steroid Biochemistry and Molecular Hu XF, Veroni M, De Luise M, Wakeling A, Sutherland Biology 69 51–84. R, Watts CK & Zalcberg JR 1993 Circumvention Labrie F, Champagne P, Labrie C, Roy J, Laverdiere J, of tamoxifen resistance by the pure anti-estrogen Provencher L, Potvin M, Drolet Y, Pollak M, Panasci L ICI 182,780. International Journal of Cancer 55 et al. 2004 Activity and safety of the antiestrogen 873–876. EM-800, the orally active precursor of acolbifene, in Hutcheson IR, Knowlden JM, Madden T-A, Barrow D, tamoxifen-resistant breast cancer. Journal of Clinical Gee JM, Wakeling AE & Nicholson RI 2003 Oestrogen Oncology 22 864–871. receptor-mediated modulation of the EGFR/MAPK Lee ES, Schafer JM, Yao K, England G, O’Regan RM, pathway in tamoxifen-resistant MCF-7 cells. Breast De Los Reyes A & Jordan VC 2000 Cross resistance of Cancer Research and Treatment 81 81–93. triphenylethylene-type antiestrogens but not ICI 182,780 Ignar-Trowbridge DM, Teng CT, Ross KA, Parker MG, in tamoxifen-stimulated breast tumors grown in athymic Korach KS & McLachlan JA 1993 Peptide growth factors mice. Clinical Cancer Research 6 4893–4899. elicit estrogen receptor-dependent transcriptional acti- Lindberg MK, Moverare S, Skrtic S, Gao H, Dahlman- vation of an estrogen-responsive element. Molecular Wright K, Gustafsson JA & Ohlsson C 2003 Estrogen Endocrinology 7 992–998. receptor (ER)-beta reduces ERalpha-regulated gene

Downloaded from Bioscientifica.com at 09/27/2021 10:26:15PM via free access www.endocrinology-journals.org 703 A Howell: Pure anti-oestrogens in advanced breast cancer

transcription, supporting a “Ying Yang” relationship women: results of a North American multicenter between ERalpha and ERbeta in mice. Molecular randomized trial. Arimidex Study Group. Journal of Endocrinology 17 203–208. Clinical Oncology 18 3758–3767. Lykkesfeldt AE, Larsen SS & Briand P 1995 Human breast Newton CJ, Buric R, Trapp T, Brockmeier S, Pagotto U & cancer cell lines resistant to pure anti- are Stalla GK 1994 The unliganded estrogen receptor (ER) sensitive to tamoxifen treatment. International Journal of transduces growth factor signals. Journal of Steroid Cancer 61 529–534. Biochemistry and Molecular Biology 48 481–486. Marttunen MB, Hietanen P, Tiitinen A & Ylikorkala O 1998 Nicholson RI, Gee JM, Manning DL, Wakeling AE, Montano Comparison of effects of tamoxifen and toremifene on MM & Katzenellenbogen BS 1995 Responses to pure bone biochemistry and bone mineral density in post- antiestrogens (ICI 164,384, ICI 182,780) in estrogen- menopausal breast cancer patients. Journal of Clinical sensitive and -resistant experimental and clinical breast Endocrinology and Metabolism 83 1158–1162. cancer. Annals of the New York Academy of Sciences 761 Marttunen MB, Cacciatore B, Hietanen P, Pyrhonen S, 148–163. Tiitinen A, Wahlstrom T & Ylikorkala O 2001 Nicholson RI & Johnston SR 2005 Endocrine therapy – Prospective study on gynaecological effects of two current benefits and limitations. Breast Cancer Research antioestrogens tamoxifen and toremifene in postmeno- and Treatment 93 (Suppl 1) S3–S10. pausal women. British Journal of Cancer 84 897–902. Norris JD, Fan D, Kerner SA & McDonnell DP 1997 Matthews J, Wihle´n B, Tujague M, Wan J, Stro¨mA& Identification of a third autonomous activation domain Gustafsson JA 2006 ERb modulates ERa-mediated within the human estrogen receptor. Molecular Endo- transcriptional activation by altering the recruitment of crinology 11 747–754. c-Fos and c-Jun to estrogen responsive promoters. Osborne CK, Jarman M, McCague R, Coronado EB, Molecular Endocrinology 20 534–543. Hilsenbeck SG & Wakeling AE 1994 The importance of Mauriac L, Pippen JE, Quaresma Albano J, Gertler SZ & tamoxifen metabolism in tamoxifen-stimulated breast Osborne CK 2003 Fulvestrant (Faslodex) versus tumor growth. Cancer Chemotherapy and Pharmacology anastrozole for the second-line treatment of subgroups 34 89–95. of postmenopausal women with visceral and non- Osborne CK, Coronado-Heinsohn EB, Hilsenbeck SG, visceral metastases: combined results from two McCue BL, Wakeling AE, McClelland RA, Manning DL multicentre trials. European Journal of Cancer 39 & Nicholson RI 1995 Comparison of the effects of a pure 1228–1233. steroidal antiestrogen with those of tamoxifen in a model McClelland RA, Manning DL, Gee JM, Anderson E, Clarke of human breast cancer. Journal of the National Cancer R, Howell A, Dowsett M, Robertson JF, Blamey RW, Institute 87 746–750. Wakeling AE et al. 1996 Effects of short-term antiestro- Osborne CK, Pippen J, Jones SE, Parker LM, Ellis M, Come gen treatment of primary breast cancer on estrogen S, Gertler SZ, May JT, Burton G, Dimery I et al. 2002 receptor mRNA and protein expression and on estrogen- Double-blind, randomized trial comparing the efficacy regulated genes. Breast Cancer Research and Treatment and tolerability of fulvestrant versus anastrozole in 41 31–41. postmenopausal women with advanced breast cancer Milla-Santos A, Milla L, Rallo L & Solano V 2001 Phase III progressing on prior endocrine therapy: results of a randomized trial of toremifene vs tamoxifen in hormo- North American trial. Journal of Clinical Oncology 20 nodependent advanced breast cancer. Breast Cancer 3386–3395. Research and Treatment 65 119–124. Paech K, Webb P, Kuiper GG, Nilsson S, Gustafsson J, Mouridsen H, Gershanovich M, Sun Y, Perez-Carrion R, Kushner PJ & Scanlan TS 1997 Differential ligand Boni C, Monnier A, Apffelstaedt J, Smith R, Sleeboom activation of estrogen receptors ERalpha and ERbeta at HP, Jaenicke F et al. 2003 Phase III study of letrozole AP1 sites. Science 277 1508–1510. versus tamoxifen as first-line therapy of advanced breast Paige LA, Christensen DJ, Gron H, Norris JD, Gottlin EB, cancer in postmenopausal women: analysis of survival Padilla KM, Chang CY, Ballas LM, Hamilton PT, and update of efficacy from the International Letrozole McDonnell DP et al. 1999 Estrogen receptor (ER) Breast Cancer Group. Journal of Clinical Oncology 21 modulators each induce distinct conformational changes 2101–2109. in ER alpha and ER beta. PNAS 96 3999–4004. Munster PN, Buzdar A, Dhingra K, Enas N, Ni L, Major M, Paridaens R, Dirix L, Lohrisch C, Beex L, Nooij M, Cameron Melemed A, Seidman A, Booser D, Theriault R et al. D, Biganzoli L, Cufer T, Duchateau L, Hamilton A et al. 2001 Phase I study of a third-generation selective estrogen 2003 Mature results of a randomized phase II multicenter receptor modulator, LY353381.HCL, in metastatic breast study of exemestane versus tamoxifen as first-line cancer. Journal of Clinical Oncology 19 2002–2009. for postmenopausal women with Nabholtz JM, Buzdar A, Pollak M, Harwin W, Burton G, metastatic breast cancer. Annals of Oncology 14 Mangalik A, Steinberg M, Webster A & von Euler M 1391–1398. 2000 Anastrozole is superior to tamoxifen as first-line Perey L, Paridaens R, Nole´ F, Bonnefoi H, Aebi S, therapy for advanced breast cancer in postmenopausal Goldhirsch A, Dietrich D & Thu¨rlimann B for the Swiss

Downloaded from Bioscientifica.com at 09/27/2021 10:26:15PM via free access 704 www.endocrinology-journals.org Endocrine-Related Cancer (2006) 13 689–706

Group for Clinical Cancer Research (SAKK) 2004 human breast cancer during long-term fulvestrant treat- Fulvestrant (Faslodexe) as hormonal treatment in ment. Proceedings of the American Society of Clinical postmenopausal patients with advanced breast cancer Oncology 11, abstract 536. (ABC) progressing after treatment with tamoxifen and Robertson JF, Howell A, Gorbunova VA, Watanabe T, aromatase inhibitors: update of a phase II SAKK trial. Pienkowski T & Lichinitser MR 2005 Sensitivity to Breast Cancer Research and Treatment 88 (Suppl 1) further endocrine therapy is retained following pro- S236, abstract 6048. gression on first-line fulvestrant. Breast Cancer Research Petruzelka L, Zimovjanova M, Konopasek B, Mares P & and Treatment 92 169–174. Dlouha Z 2004 Fulvestrant in postmenopausal women Roy J, Couillard S, Gutman M & Labrie F 2003 A novel pure with metastatic breast cancer progressing on prior SERM achieves complete regression of the majority of endocrine therapy – updated results from an expanded human breast cancer tumors in nude mice. Breast Cancer access programme. Annals of Oncology 15 (Suppl 3) iii36, Research and Treatment 81 223–229. abstract 136P. Shiau AK, Barstad D, Loria PM, Cheng L, Kushner PJ, Agard Pyrhonen S, Valavaara R, Vuorinen J & Hajba A 1994 High DA & Greene GL 1998 The structural basis of estrogen dose toremifene in advanced breast cancer resistant to or receptor/coactivator recognition and the antagonism of relapsed during tamoxifen treatment. Breast Cancer this interaction by tamoxifen. Cell 95 927–937. Research and Treatment 29 223–228. Shupnik MA 2004 Crosstalk between steroid receptors and Pyrhonen S, Valavaara R, Modig H, Pawlicki M, the c-src-receptor tyrosine kinase pathways: implications Pienkowski T, Gundersen S, Bauer J, Westman G, for cell proliferation. Oncogene 23 7979–7989. Lundgren S, Blanco G et al. 1997 Comparison of Sibonga JD, Dobnig H, Harden RM & Turner RT 1998 Effect toremifene and tamoxifen in post-menopausal patients of the high-affinity estrogen receptor ligand ICI 182,780 with advanced breast cancer: a randomized double- on the rat tibia. Endocrinology 139 3736–3742. blind, the ‘nordic’ phase III study. British Journal of Smith CL, Conneely OM & O’Malley BW 1993 Modulation Cancer 76 270–277. of the ligand-independent activation of the human estrogen Pyrhonen S, Ellmen J, Vuorinen J, Gershanovich M, receptor by hormone and antihormone. PNAS 90 Tominaga T, Kaufmann M & Hayes DF 1999 Meta- 6120–6124. analysis of trials comparing toremifene with tamoxifen Steger GG, Bartsch R, Wenzel C, Pluschnig U, Hussain D, and factors predicting outcome of antiestrogen therapy in Sevelda U, Locker GJ, Gnant MF, Jakesz R & Zielinski CC postmenopausal women with breast cancer. Breast 2005 (‘Faslodex’) in pre-treated patients with advanced Cancer Research and Treatment 56 133–143. breast cancer. European Journal of Cancer 41 2655–2661. Qi X, Stanley B, Pramanik R, Schultz RM, Han J & Chen G Stenbygaard LE, Herrstedt J, Thomsen JF, Svendsen KR, 2004 Estrogen receptor inhibits c-Jun-dependent stress- Engelholm SA & Dombernowsky P 1993 Toremifene and induced cell death by binding and modifying c-Jun tamoxifen in advanced breast cancer – a double-blind activity in human breast cancer cells. Journal of cross-over trial. Breast Cancer Research and Treatment Biological Chemistry 279 6769–6777. 25 57–63. Reese JC & Katzenellenbogen BS 1991 Differential DNA- Taras TL, Wurz GT & DeGregorio MW 2001 In vitro and binding abilities of estrogen receptor occupied with two in vivo biologic effects of Ospemifene (FC-1271a) in classes of antiestrogens: studies using human estrogen breast cancer. Journal of Steroid Biochemistry and receptor overexpressed in mammalian cells. Nucleic Molecular Biology 77 271–279. Acids Research 19 6595–6602. Tsai MJ & O’Malley BW 1994 Molecular mechanisms of Robertson JF, Nicholson RI, Bundred NJ, Anderson E, action of steroid/thyroid receptor superfamily members. Rayter Z, Dowsett M, Fox JN, Gee JM, Webster A, Annual Review of Biochemistry 63 451–486. Wakeling AE et al. 2001 Comparison of the short-term Tzukerman MT, Esty A, Santiso-Mere D, Danielian P, Parker biological effects of 7alpha-[9-(4,4,5,5,5-pentafluoro- MG, Stein RB, Pike JW & McDonnell DP 1994 Human pentylsulfinyl)-nonyl]estra-1,3,5, (10)-triene-3,17beta- estrogen receptor transactivational capacity is determined diol (Faslodex) versus tamoxifen in postmenopausal by both cellular and promoter context and mediated women with primary breast cancer. Cancer Research 61 by two functionally distinct intramolecular regions. 6739–6746. Molecular Endocrinology 8 21–30. Robertson JF, Osborne CK, Howell A, Jones SE, Mauriac L, van de Velde CJ, Nique F, Bouchoux F, Bremaud J, Ellis M, Kleeberg UR, Come SE, Vergote I, Gertler S Hameau MC, Lucas D, Moratille C, Viet S, Philibert D et al. 2003 Fulvestrant versus anastrozole for the & Teutsch G 1994 RU 58,668, a new pure antiestrogen treatment of advanced breast carcinoma in postmeno- inducing a regression of human mammary carcinoma pausal women – a prospective combined analysis of two implanted in nude mice. Journal of Steroid Biochem- multicenter trials. Cancer 98 229–238. istry and Molecular Biology 48 187–196. Robertson JFR, Gutteridge E, Cheung KL, Pinder S & Vergote I, Robertson JFR, Kleeberg U, Burton G, Osborne Wakeling A 2004 Oestrogen receptor expression in CK & Mauriac L 2003 Postmenopausal women who

Downloaded from Bioscientifica.com at 09/27/2021 10:26:15PM via free access www.endocrinology-journals.org 705 A Howell: Pure anti-oestrogens in advanced breast cancer

progress on fulvestrant (’Faslodex’) remain sensitive to Weigel NL & Zhang Y 1998 Ligand-independent activation further endocrine therapy. Breast Cancer Research and of steroid hormone receptors. Journal of Molecular Treatment 79 207–211. Medicine 76 469–479. Wakeling AE 1995 Use of pure antioestrogens to elucidate White R & Parker MG 1998 Molecular mechanisms of the mode of action of oestrogens. Biochemical Pharma- steroid hormone action. Endocrine-Related Cancer 5 cology 49 1545–1549. 1–14. Wakeling AE 2000 Similarities and distinctions in the mode Wijayaratne AL, Nagel SC, Paige LA, Christensen DJ, Norris of action of different classes of antioestrogens. Endocrine- JD, Fowlkes DM & McDonnell DP 1999 Comparative Related Cancer 7 17–28. analyses of mechanistic differences among antioestrogens. Wakeling AE & Bowler J 1987 Steroidal pure antioestrogens. Endocrinology 140 5828–5840. Journal of Endocrinology 112 R7–R10. Yamamoto Y, Shibata J, Yonekura K, Sato K, Hashimoto A, Wakeling AE, Dukes M & Bowler J 1991 A potent specific Aoyagi Y, Wierzba K, Yano S, Asao T, Buzdar AU & pure antiestrogen with clinical potential. Cancer Research Terada T 2005 TAS-108, a novel oral steroidal 51 3867–3873. antiestrogenic agent, is a pure antagonist on estrogen Webb P, Nguyen P & Kushner PJ 2003 Differential SERM receptor alpha and a partial agonist on estrogen receptor effects on corepressor binding dictate ERa activity beta with low uterotrophic effect. Clinical Cancer in vivo. Journal of Biological Chemistry 278 6912–6920. Research 11 315–322.

Downloaded from Bioscientifica.com at 09/27/2021 10:26:15PM via free access 706 www.endocrinology-journals.org