Abstract
Endocrine therapy is an effective option for the treatment of estrogen receptor alpha (ERα)-positive breast cancers. Unfortunately, a large fraction of women relapse with endocrine-resistant tumors. The presence of constitutively active ERα mutants, found in a subset of relapse tumors, is thought to be an important endocrine resistance mechanism and has prompted the search for more effective anti-hormone drugs that can effectively inhibit these mutant versions of the receptor. The G protein-coupled estrogen receptor (GPER), is also thought to contribute to the development of endocrine resistance, in part, due to its activation by clinically used selective estrogen receptor modulators and downregulators (SERMs/SERDs). Therefore, next-generation drugs should be screened for potential activity towards GPER. Here we highlight the need for truly ERα-selective SERMs and SERDs that do not cross-react with GPER for the treatment of ERα-positive breast cancers.
Keywords: Breast Cancer, Endocrine Resistance, ERα, GPER, SERM, SERD
Introduction
Estrogen (17β-estradiol, E2) plays a role in various physiological processes, but is best known for its role in female reproduction and breast tissue development [1]. It exerts its effects through three known estrogen receptors: ERα, ERβ and GPER (formerly known as GPR30) [2–4]. ERα and ERβ, termed the classical estrogen receptors, are ligand-activated nuclear transcription factors that primarily exert estrogen-mediated genomic signaling inside the cell [5, 6]. Unlike the classical ERs, GPER is a G protein-coupled receptor that for the most part mediates estrogen-induced rapid, non-genomic signaling that results in the initiation of an array of downstream signals, including the activation of the mitogen-activated protein kinase (MAPK)/ERK and phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathways, calcium mobilization and cAMP production, via the transactivation of the epidermal growth factor receptor (EGFR) [4, 7]. However, GPER can also, to a certain extent, modulate transcription of genes, such as c-fos [8, 9]. Like GPER, the classical ERs also mediate E2-induced rapid, non-genomic signaling, which is thought to occur through a membrane-bound fraction of the receptors, resulting in rapid downstream signals similar to those activated by GPER [10]. Thus, overlapping signaling pathways between the classical ERs and GPER exist.
Aside from its physiological roles, ERα is also an important initiator and driver of breast cancer [11]. Due to its role and prevalence in the majority of breast cancers (~70%), ERα has been an important molecular target for the treatment of ERα-positive breast cancers [12]. ERβ, on the other hand, is generally downregulated in breast cancers, making it a potentially less desirable therapeutic target [13–15]. However, some groups have observed increased expression of ERβ in breast cancer samples and have reported that ERβ correlates with improved patient outcome [16, 17]. Due to conflicting reports on potential roles of ERβ in breast cancer, this review will focus on ERα [15]. Inhibition of ERα signaling by endocrine therapy, using selective estrogen receptor modulators (SERMs, such as tamoxifen), the selective estrogen receptor downregulator (SERD) fulvestrant, and aromatase inhibitors (AIs, e.g. letrozole and exemestane), has shown great clinical success in treating ERα-positive breast cancers [18, 19]. Unfortunately, a large fraction of patients relapse with endocrine therapy-resistant tumors that no longer respond to the primary therapy.
Endocrine resistance has become a major clinical hurdle in the treatment of ERα-positive breast cancers and approaches to overcome this resistance are an ongoing area of research. Several resistance mechanisms have been described, including the emergence of constitutively active mutant forms of ERα (e.g. Y537S and D538G), in relapse tumors that are resistant to current ERα-targeted endocrine therapies [20–22]. GPER has also been suggested to contribute to the development of endocrine resistance, possibly through its cross-reaction with tamoxifen and other clinically utilized SERMs and SERD, yet its role remains unclear [23].
In this review we highlight a) current endocrine therapies for the treatment of ERα-positive breast cancers, b) the role of mutant ERα as an endocrine resistance mechanism and c) the potential role of GPER in the development of endocrine resistance. We also discuss currently developing ERα-targeting compounds and why their potential cross-reactivity to GPER should be taken into consideration in the development of new therapies.
Breast cancer endocrine therapies
Endocrine therapy is effective at treating ERα-positive breast cancers and has prolonged the lives of millions of women [18, 19]. It focuses on inhibiting the growth of ERα-positive breast cancers by blocking the activation of the ERα, and thus its downstream proliferative and pro-survival signaling. Endocrine therapy achieves this either by directly blocking estrogen binding to ERα (using SERMs or SERDs) or by decreasing the plasma levels of circulating estrogen by inhibiting its production (using Als).
SERMs and SERDs
SERMs are a class of small molecules that act as antagonists of ERα by competing with estrogen for binding to the receptor, thereby blocking the recruitment of co-regulators to the receptor [24]. Their activity is tissue specific, acting as ERα antagonists in the breast, while functioning as ERα agonists in the bone and uterus [25, 26]. Currently FDA-approved SERMs include tamoxifen, raloxifene and toremifene (Figure 1).
Figure 1.
Structures of the SERMs tamoxifen, toremifene and raloxifene and the SERD fulvestrant.
Tamoxifen (Nolvadex®, Soltamox®), the most prescribed SERM, has shown great success in the clinic for both the treatment and prevention of ERα-positive breast cancers in pre- and post-menopausal women, showing a significant decrease in ERα-positive breast cancer-related mortality and recurrence rates when taken over a 5 (and up to 10) year period [27, 28]. Unfortunately, due to its agonist activity in the endometrium, women taking tamoxifen experience up to a 7.5-fold increased risk of developing endometrial cancer [25, 29, 30]. Patients taking tamoxifen also have an increased risk of developing thromboembolisms [31, 32]. However, these risks do not tend to outweigh the benefits of the drug. Compared to tamoxifen, raloxifene (Evista®) carries a lower risk of developing thromboembolisms, but is less effective at preventing invasive breast cancer [33]. Nevertheless, raloxifene significantly decreases the risk of developing invasive breast cancer in post-menopausal women with osteoporosis [34]. Raloxifene is currently approved for the prevention of invasive breast cancer in post-menopausal women with osteoporosis. The third FDA-approved SERM, toremifene (Fareston®), also increases the risk of developing thromboembolisms, but data regarding its causal role in developing endometrial cancer is limited [35]. Toremifene is approved for the treatment of advanced ERα-positive breast cancers in post-menopausal women. To date, tamoxifen is the only SERM approved for the treatment of ERα-positive breast cancers in pre-menopausal women.
SERDs are a class of small molecules that, like SERMs, act as antagonists of the ERα. However, unlike SERMs, SERDs not only antagonize the receptor, but also induce its degradation, resulting in a decrease in ERα protein levels [36]. Furthermore, SERDs are “pure” antagonists of the ERα, in that they do not show agonist properties in other tissues [37]. Currently, fulvestrant (Faslodex®, Figure 1) is the only FDA-approved SERD and is used for the treatment of metastatic ERα-positive breast cancers in post-menopausal women with disease progression following prior endocrine therapies.
Aromatase inhibitors
Aromatase inhibitors (Als) are a class of small molecules that block the production of estrogen by inhibiting the enzyme aromatase (CYP19A1) [38]. They have been highly effective in the clinic for the treatment of ERα-positive breast cancers in post-menopausal women [39]. In fact, they are more efficacious than tamoxifen in this setting [19, 40]. However, this class of drugs has significant side-effects, including muscle/joint pain, bone loss, hot flashes and insomnia, causing over 30% of patients to discontinue treatment [41–43]. Nevertheless, Als are highly effective in the treatment of ERα-positive breast cancers and are becoming a more popular standard first-line endocrine therapy option for post-menopausal women. Currently FDA-approved Als include the reversible Als anastrozole (Arimidex®) and letrozole (Femara®) and the irreversible Al, exemestane (Aromasin®) (Figure 2).
Figure 2.
Structures of the AIs exemestane, anastrozole and letrozole.
Endocrine resistance
Endocrine therapy is the standard of care for the treatment of ERα-positive breast cancers. Unfortunately, some 30% of patients eventually develop acquired endocrine resistance to the initial therapy, with reports suggesting that this number may be as high as 40–50% [18, 44]. Several mechanisms have been proposed to account for the development of endocrine resistance, including alterations in growth factor signaling (e.g. overexpression of the EGFR or HER-2), changes in the expression of ERα and its co-activators/co-repressors, mutations in ERα, changes in the metabolism of tamoxifen and tamoxifen-induced GPER signaling (discussed later) [22, 45–47].
In the late 1990s, Zhang et al. reported the identification of three ESR1 (the gene encoding ERα) mutations in metastatic breast cancer samples [48]. One of the mutations, Y537N, was found to cause constitutive activation of the receptor, even in the absence of estrogen. More importantly, this mutation made the receptor less sensitive to inhibition by tamoxifen (the maximum tested dose was 100 nM). This led to the proposal that mutations of this site could contribute to breast cancer progression and endocrine resistance. Similar findings regarding mutations at the Y537 codon were previously reported by Weis et al. in a structure-activity relationship study [49]. More recently, studies identified several ESR1 mutations (e.g. E380Q, Y537S, Y537C and D538G) that were highly enriched in up to 55% of metastatic breast cancer samples of patients who relapsed while on endocrine therapy [20, 21, 50–52]. Importantly, these mutations were for the most part absent in matched primary breast tumors as well as primary breast tumors, in general, pointing to a possible role in endocrine resistance. However, several of these mutations have been identified at low frequencies in primary breast tumors (<1–3%), but at a far lower level than in relapse tumors [21]. The most commonly occurring mutations were Y537S and D538G, located in the ligand-binding domain (LBD) of ERα. These mutations result in constitutive activation of the receptor by inducing a ligand-bound confirmation of the receptor in the absence of estrogen. More specifically, they stabilize helix 12, an important structural component in the LBD of ERα, in a closed agonist-bound confirmation, thereby exposing a co-regulator binding site on the receptor that is normally only revealed following the binding of estrogen. This stabilized ligand-bound confirmation also makes the mutant receptor less susceptible to inhibition by tamoxifen and fulvestrant, by impairing access to the ligand binding pocket of the receptor [21, 50, 53]. These observations support the role of these mutations in endocrine resistance in response to not only AIs, but also SERM- and SERD-based therapies. Interestingly, the Y537S mutation has been reported to be more resistant than the D538G mutation, to in vivo inhibition by fulvestrant [54].
Clinical data of patients that have relapsed while on an AI therapy have shown that the Y537S and D538G mutants are associated with more aggressive relapse tumors [55]. This aggressive phenotype could be due to the unique transcriptome induced by the Y537S and D538G mutants (compared to that of wildtype ERα) that includes the modulation of pro-metastatic genes [56]. This merits further research into genes that are specific targets of the mutant forms of ERα, as potentially new therapeutic targets for the treatment of relapsed breast tumors expressing mutant ERα. Interestingly, in a retrospective analysis of the FERGI trial (a comparison of treatment with a PI3K inhibitor + fulvestrant versus placebo + fulvestrant, in ERα-positive breast cancer patients with locally advanced or metastatic disease), Spoerke et al. did not observe a difference in the control placebo + fulvestrant arm of the trial with respect to progression-free survival (PFS) between patients harboring wildtype ESR1 versus patients harboring mutated ESR1, as measured using circulating tumor DNA [57]. This underlines the complexity of tumor heterogeneity and how the presence of other mutations alongside ESR1 mutations could potentially influence ERα-targeted endocrine therapies. Furthermore, it could imply that ESR1 mutations are more important in endocrine resistance in a hormone-deprived setting (e.g. in patients on AI therapies) than in tumors that relapse on SERM/SERD therapies.
Nevertheless, the presence and activity of mutant forms of ERα in the metastatic relapse setting have become a highly studied resistance mechanism and are now strongly considered in the development of next-generation SERMs and SERDs (discussed in the next section). We acknowledge the existence of other resistance mechanisms that are important in treating ERα-positive breast cancers, but these will not be covered in this review. We direct the reader to comprehensive reviews on other mechanisms cited earlier in this section.
Current status of next-generation SERMs and SERDs
Acquired endocrine resistance is a major clinical problem in treating ERα-positive breast cancers. The presence of the Y537S and D538G ERα mutants in many relapse tumors highlights the need for the development of improved SERMs and SERDs that can effectively inhibit both the wildtype and clinically-observed mutant forms of ERα [21, 50, 51]. Such new drugs could be effective in treating relapse tumors harboring the mutant receptor and improve disease stabilization, thereby prolonging the lives of patients.
Currently, for women with advanced metastatic ERα-positive breast cancer that has relapsed while on endocrine therapy, therapeutic options are limited. In this setting, the SERD fulvestrant has shown the most promising results in extending PFS, either as a monotherapy or in combination with targeted therapies like cyclin-dependent kinase 4/6 (CDK4/6) inhibitors [58, 59]. With regard to the presence of ESR1 mutations, Fribbens et al. analyzed ESR1 mutations in baseline plasma from the SOFEA trial (comparing exemestane versus fulvestrant-containing therapies) and the PALOMA3 trial (comparing fulvestrant + placebo versus fulvestrant + the CDK4/6 inhibitor palbociclib) [60]. They found that fulvestrant provided a significant benefit over exemestane in patients with tumors harboring ESR1 mutations. However, treatment with fulvestrant had a modestly worse PFS outcome for patients with ESR1 mutations when compared to patients with wildtype ESR1. The latter result is not surprising given that mutant forms of ERα are less sensitive to fulvestrant and require higher drug concentrations to be fully inhibited [20, 21, 54]. The currently approved dosage of fulvestrant (500 mg) has been shown to be ineffective in fully blocking ERα in tumors, and in turn associated with early disease progression [61]. Taken together, the aforementioned observations imply that achieving higher plasma levels of fulvestrant could be of great therapeutic benefit, especially with regard to relapse tumors harboring ESR1 mutations. Unfortunately, fulvestrant has poor pharmacological properties, requiring it to be administered through painful, monthly intramuscular injections, thereby limiting the maximum achievable dose of the drug. As a consequence, the development of new, more bioavailable SERMs/SERDs that possess better pharmacological properties, while also efficaciously inhibiting ERα mutants, is the goal of much current research.
The small molecule ZB716 (Figure 3), a boronic acid-modified version of fulvestrant with high oral bioavailability, is an example of how improving the bioavailability of a drug can lead to higher plasma levels [62–64]. Liu et al. showed that ZB716 achieved over 10 times higher plasma levels than fulvestrant when orally administered to mice and that ZB716 has similar levels of efficacy (compared to fulvestrant) in vitro [62]. Similarly, Guo et al. showed that ZB716 achieved higher plasma levels in both the blood and tumors in xenograft models [64]. The latter study also demonstrated that ZB716 is more efficacious than fulvestrant in tumor growth inhibition in a cell line-derived xenograft model. Interestingly, ZB716 displayed no observed benefit (versus fulvestrant) in inhibiting tumor growth in a patient-derived xenograft (PDX) model.
Figure 3.
Structures of the currently in-development orally bioavailable SERDs AZD9496, LSZ102, GDC-0927, ZB716 and the orally bioavailable SERM bazedoxifene.
Several new SERDs are currently being developed and assessed in clinical trials to treat advanced endocrine-resistant breast cancers (Figure 3). AZD9496, an orally bioavailable SERD developed by AstraZeneca, has been shown to be highly effective at inhibiting ERα-positive breast cancer cells in vitro and in vivo, while also being highly effective at binding and degrading wildtype and mutant forms of ERα (the binding IC50 values for AZD9496 are >6-fold improved versus those of fulvestrant) [65, 66]. Furthermore, Weir et al. showed that AZD9496 inhibited the growth of a PDX tumor harboring the D538G ERα mutant [66]. A phase 1 clinical trial of AZD9496 was recently completed and indicated that the drug is tolerated at the maximum tested dose (600 mg), even showing some early signs of disease stabilization over a 12 month period [67].
Another orally bioavailable SERD, G1T48, developed by G1 Therapeutics Inc., is effective at inhibiting ERα-positive tumor growth in in vivo models of tamoxifen and aromatase resistance [68]. Furthermore, G1T48 inhibits the growth of ERα-Y537S- and ERα-D538G-expressing breast cancer cells. G1T48 also showed synergistic inhibition in a tamoxifen-resistant xenograft model when used in combination with the novel CDK4/6 inhibitor, G1T38. G1T48 recently entered a Phase 1 clinical trial () for initial in-human safety testing in women with advanced ERα-positive breast cancer.
Other orally bioavailable SERDs, including LSZ102 (Novartis Pharmaceuticals, ), SAR439859 (Sanofi Inc., ) and GDC-0927 (Genentech Inc., ) are also currently being evaluated both in the lab and in clinical trials as potentially new therapeutic agents for the treatment of advanced ERα-positive breast cancers [69–73]. Newly emerging SERDs show promising results as potential ERα-targeting breast cancer therapies, but only time will tell if any of these compounds are efficacious in patients and gain clinical approval.
Aside from orally bioavailable SERDs, the already clinically-approved SERM/SERD hybrid, bazedoxifene (Figure 3), is also gaining more interest as a potential therapeutic agent for treating ERα-positive advanced breast cancers. Bazedoxifene, which is already used in hormone replacement therapy and for the prevention of post-menopausal osteoporosis, exhibits favorable efficacy in various endocrine-resistant breast cancer models [74–77]. Furthermore, Fanning et al. reported that bazedoxifene inhibits the activity of the Y537S and D538G mutant forms of ERα, offering structural insights into its inhibitory mechanism [78]. Its activity in models of endocrine resistance and its well tolerated drug profile, make bazedoxifene a candidate that could prove efficacious in treating ERα-positive breast cancers.
GPER: a culprit in endocrine resistance?
The non-classical estrogen receptor, GPER, has gained attention as a possible player in the development of endocrine resistance in breast cancer. This theory stems from clinical observations that found a significant increase in the expression of GPER in relapse tumors of patients that had undergone prior tamoxifen therapy [23, 79]. Furthermore, GPER expression has been shown to correlate negatively with relapse-free survival, further indicating an unfavorable role of the receptor in endocrine resistance [79]. Recently, Ignatov et al. reported the opposite, finding a positive correlation between GPER-expression and disease-free survival. However, this correlation was not statistically significant [80]. Interestingly, they did report that treatment with tamoxifen was less beneficial than an AI-based therapy in patients with GPER-positive breast tumors.
In vitro data also supports the clinical observations that point to a negative role for GPER in endocrine resistance. Ignatov et al. showed that tamoxifen-induced proliferation of tamoxifen-resistant MCF-7 cells could be abrogated by downregulation of GPER [47]. Similar findings were reported by Mo et al. who showed that GPER-inhibition, using the GPER-selective antagonist G15, could “re-sensitize” tamoxifen-resistant cells to tamoxifen [23, 81]. Tamoxifen-induced proliferation, via a GPER-mediated pathway, has also been observed in other cell types [8, 82].
Some elucidation for a role of GPER in endocrine resistance came from Catalano et al. who showed that tamoxifen-resistant MCF-7 cells expressed higher levels of aromatase [83]. Increased aromatase expression could potentially create a local increase in estrogen levels, thereby allowing tumors to overcome the inhibitory effects of tamoxifen and AI therapies. Importantly, the increased aromatase expression could be reverted back to baseline levels by reducing GPER expression or inhibiting its function using G15.
Zekas et al. also provided a potential mechanism for GPER in endocrine resistance [84]. They observed that tamoxifen and fulvestrant, through a GPER-mediated pathway, induce the translocation of the pro-apoptotic transcription factor Forkhead box protein O3a (FOXO3a) out of the nucleus. Nuclear expulsion of FOXO3a is a phenotype that is associated with its inactivation and promotes a pro-survival state [85–87]. Importantly, knockdown of GPER abrogated the observed ligand-induced translocation of FOXO3a. It was suggested that tamoxifen-induced inactivation of FOXO3a could provide a fraction of tumor cells with a survival advantage to overcome inhibitory effects of tamoxifen mediated through ERα, eventually leading to the acquisition of endocrine resistance through mutations.
An important observation linking GPER to endocrine resistance was provided by Mo et al. [23]. They reported that a tamoxifen-resistant MCF-7 tumor could be “re-sensitized” to tamoxifen treatment of mice with a combination of tamoxifen and G15. Importantly, mono-therapy with either tamoxifen or G15 had no effect on tumor growth. This implies that pro-survival signaling by GPER (through its downstream target Akt) is possibly overcoming the tamoxifen-induced apoptotic signaling (via ERα), a consequence of tamoxifen-mediated activation of GPER (discussed in the next section).
The body of scientific data pointing to a role for GPER in endocrine resistance has been met with some contradictory reports. A small number of publications have reported that activation of GPER, using the GPER-selective agonist G-1, induces cell death of breast cancer cells [88–90]. Whether this cell death is caused by hyperstimulation of GPER or is a possible off-target effect due to the high doses of G-1 tested, remains unclear and warrants further investigation. Nevertheless, these findings do not diminish the numerous observations implying a pro-survival role for GPER in endocrine resistance.
A proposed role for GPER in endocrine resistance and how to circumvent it
SERMs and SERDs are widely used to inhibit ERα signaling in breast cancers. However, contrary to their antagonism towards ERα, currently approved SERMs (tamoxifen and raloxifene) and SERD (fulvestrant) have been shown to act as agonists of GPER in breast cancer cell models, resulting in the activation of the ERK and PI3K pathway [4, 91, 92]. Similarly, many other estrogenic compounds (e.g. xenoestrogens and phytoestrogens) that act on the classical ERs have also been found to act as agonists of GPER [93].
This cross-activation of GPER, particularly by tamoxifen, is a common element seen in all of the observations discussed in the previous section and is a potential clue into the role of GPER in the development of endocrine resistance [23, 47, 83]. In the case of tamoxifen, patients are treated for a minimum of 5 years with cancer relapse often occurring after completion of this 5 year period [18]. It is possible that chronic activation of GPER by tamoxifen, promotes long-term survival of a fraction of primary tumor cells or alternatively breast cancer stem cells. This prolonged survival could grant this subset of tumor cells sufficient time to acquire additional mutations, resulting in resistance to the primary therapy (e.g. acquiring endocrine-resistant ESR1 mutations) and leading to the development of endocrine-resistant relapse tumors (Figure 4). This possible role highlights the potentially significant benefit of developing truly ERα-selective therapeutic antagonists that do not cross-activate GPER. A truly ERα-selective antagonist would lack cross-activation of GPER, and its resulting pro-survival signaling, thus potentially delaying or decreasing the development of endocrine-resistant relapse tumors.
Figure 4. Proposed role of GPER in the development of endocrine resistance.
Tamoxifen (as well as other SERMs and SERDs) inhibits its molecular target (ERα) (red line), but simultaneously cross-activates GPER (green arrow). This cross-activation induces the downstream activation of Akt and other survival signals. Chronic cross-activation of GPER by tamoxifen (over the course of the average 5 year tamoxifen regimen) provides a subset of primary tumor cells with prolonged survival signaling that opposes the inhibitory cell death induced through the tamoxifen-mediated inhibition of ERα. Prolonged survival thus provides this subset of surviving primary tumor cells sufficient time to acquire additional mutations that lead to resistance to the primary therapy, resulting in the development of endocrine-resistant relapse tumors.
At this time, no truly ERα-selective compounds (agonists or antagonists) have been identified in the literature. The current development of next-generation SERDs has focused on the compound’s growth inhibitory potential in models of endocrine resistance, yet none have been assessed for their potential cross-activity towards GPER. Although these new compounds look promising, harboring cross-activity towards GPER could limit their long-term efficacy in treating ERα-positive advanced breast cancers. This emphasizes the potential benefit of truly ERα-selective antagonists. It is worth noting that to date one compound, termed MIBE, has been reported to act as an antagonist towards both the ERα and GPER [94]. However, its efficacy has not yet been assessed in models of endocrine resistance.
Conclusions
Endocrine therapy is a mainstay in the treatment of ERα-positive breast cancers. Although highly effective, resistance to the therapy is common and many mechanisms have been described to account for the development of endocrine resistance, including the emergence of endocrine-resistant ERα mutants (e.g. Y537S and D538G). The constitutively active mutant forms of ERα are effective at overcoming therapeutic intervention with current SERM, SERD and AI-based endocrine therapies. Therefore, next-generation SERDs will need to be effective towards these mutant forms of the receptor. Moreover, new SERDs should be assessed for potential cross-activation of GPER, which is thought to be a causal role for the development of endocrine resistance to currently approved SERMs and SERD. We have proposed a model describing how GPER promotes the development of endocrine resistance through its cross-activation by tamoxifen and fulvestrant, highlighting the need for truly ERα-selective antagonists, and their possible long-term benefits for patients.
Acknowledgements
E.R.P. is supported by grants from the National Institutes of Health (NIH R01 CA163890 and CA194496) and Dialysis Clinic, Inc., the Autophagy, Inflammation and Metabolism (AIM) Center of Biomedical Research Excellence (NIH P20 GM121176) and by the UNM Comprehensive Cancer Center (NIH grant P30 CA118100).
Abbreviations
- AI
aromatase inhibitor
- cAMP
cyclic adenosine monophosphate
- CDK
cyclin-dependent kinase
- EGFR
epidermal growth factor receptor
- FOXO3a
forkhead box protein O3a
- GPER
G protein-coupled estrogen receptor
- HER-2
human epidermal growth factor receptor 2
- LBD
ligand-binding domain
- PDX
patient-derived xenograft
- PFS
progression-free survival
- SERM
selective estrogen receptor modulator
- SERD
selective estrogen receptor downregulator
Footnotes
Conflicts of interest
E.R.P. is an inventor on U.S. Patent Nos. 7,875,721 and 8,487,100 for GPER-selective ligands and imaging agents and U.S. Patent No. 10,251,870 and pending patents for applications of GPER-selective ligands.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- [1].Nilsson S, Makela S, Treuter E, Tujague M, Thomsen J, Andersson G, Enmark E, Pettersson K, Warner M, Gustafsson JA, Mechanisms of estrogen action, Physiological reviews 81(4) (2001) 1535–65. [DOI] [PubMed] [Google Scholar]
- [2].Jensen EV, DeSombre ER, Estrogen-receptor interaction, Science 182(4108) (1973) 126–34. [DOI] [PubMed] [Google Scholar]
- [3].Kuiper GG, Enmark E, Pelto-Huikko M, Nilsson S, Gustafsson JA, Cloning of a novel receptor expressed in rat prostate and ovary, Proceedings of the National Academy of Sciences of the United States of America 93(12) (1996) 5925–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [4].Revankar CM, Cimino DF, Sklar LA, Arterburn JB, Prossnitz ER, A transmembrane intracellular estrogen receptor mediates rapid cell signaling, Science 307(5715) (2005) 1625–30. [DOI] [PubMed] [Google Scholar]
- [5].Dahlman-Wright K, Cavailles V, Fuqua SA, Jordan VC, Katzenellenbogen JA, Korach KS, Maggi A, Muramatsu M, Parker MG, Gustafsson JA, International Union of Pharmacology. LXIV. Estrogen receptors, Pharmacological reviews 58(4) (2006) 773–81. [DOI] [PubMed] [Google Scholar]
- [6].Heldring N, Pike A, Andersson S, Matthews J, Cheng G, Hartman J, Tujague M, Strom A, Treuter E, Warner M, Gustafsson JA, Estrogen receptors: how do they signal and what are their targets, Physiological reviews 87(3) (2007) 905–31. [DOI] [PubMed] [Google Scholar]
- [7].Prossnitz ER, Arterburn JB, Smith HO, Oprea TI, Sklar LA, Hathaway HJ, Estrogen signaling through the transmembrane G protein-coupled receptor GPR30, Annual review of physiology 70 (2008) 165–90. [DOI] [PubMed] [Google Scholar]
- [8].Vivacqua A, Bonofiglio D, Recchia AG, Musti AM, Picard D, Ando S, Maggiolini M, The G protein-coupled receptor GPR30 mediates the proliferative effects induced by 17beta-estradiol and hydroxytamoxifen in endometrial cancer cells, Molecular endocrinology 20(3) (2006) 631–46. [DOI] [PubMed] [Google Scholar]
- [9].Madeo A, Maggiolini M, Nuclear alternate estrogen receptor GPR30 mediates 17beta-estradiol-induced gene expression and migration in breast cancer-associated fibroblasts, Cancer research 70(14) (2010) 6036–46. [DOI] [PubMed] [Google Scholar]
- [10].Levin ER, Extranuclear steroid receptors are essential for steroid hormone actions, Annual review of medicine 66 (2015) 271–80. [DOI] [PubMed] [Google Scholar]
- [11].Russo J, Russo IH, The role of estrogen in the initiation of breast cancer, The Journal of steroid biochemistry and molecular biology 102(1–5) (2006) 89–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [12].Ariazi EA, Ariazi JL, Cordera F, Jordan VC, Estrogen receptors as therapeutic targets in breast cancer, Current topics in medicinal chemistry 6(3) (2006) 181–202. [PubMed] [Google Scholar]
- [13].Shaaban AM, O’Neill PA, Davies MP, Sibson R, West CR, Smith PH, Foster CS, Declining estrogen receptor-beta expression defines malignant progression of human breast neoplasia, The American journal of surgical pathology 27(12) (2003) 1502–12. [DOI] [PubMed] [Google Scholar]
- [14].Leygue E, Dotzlaw H, Watson PH, Murphy LC, Altered estrogen receptor alpha and beta messenger RNA expression during human breast tumorigenesis, Cancer research 58(15) (1998) 3197–201. [PubMed] [Google Scholar]
- [15].Warner M, Huang B, Gustafsson JA, Estrogen Receptor beta as a Pharmaceutical Target, Trends in pharmacological sciences 38(1) (2017) 92–99. [DOI] [PubMed] [Google Scholar]
- [16].Speirs V, The evolving role of oestrogen receptor beta in clinical breast cancer, Breast cancer research : BCR 10(5) (2008) 111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [17].Hopp TA, Weiss HL, Parra IS, Cui Y, Osborne CK, Fuqua SA, Low levels of estrogen receptor beta protein predict resistance to tamoxifen therapy in breast cancer, Clinical cancer research : an official journal of the American Association for Cancer Research 10(22) (2004) 7490–9. [DOI] [PubMed] [Google Scholar]
- [18].Early G Breast Cancer Trialists’ Collaborative, Effects of chemotherapy and hormonal therapy for early breast cancer on recurrence and 15-year survival: an overview of the randomised trials, Lancet 365(9472) (2005) 1687–717. [DOI] [PubMed] [Google Scholar]
- [19].Early G Breast Cancer Trialists’ Collaborative, Aromatase inhibitors versus tamoxifen in early breast cancer: patient-level meta-analysis of the randomised trials, Lancet 386(10001) (2015) 1341–1352. [DOI] [PubMed] [Google Scholar]
- [20].Merenbakh-Lamin K, Ben-Baruch N, Yeheskel A, Dvir A, Soussan-Gutman L, Jeselsohn R, Yelensky R, Brown M, Miller VA, Sarid D, Rizel S, Klein B, Rubinek T, Wolf I, D538G mutation in estrogen receptor-alpha: A novel mechanism for acquired endocrine resistance in breast cancer, Cancer research 73(23) (2013) 6856–64. [DOI] [PubMed] [Google Scholar]
- [21].Toy W, Shen Y, Won H, Green B, Sakr RA, Will M, Li Z, Gala K, Fanning S, King TA, Hudis C, Chen D, Taran T, Hortobagyi G, Greene G, Berger M, Baselga J, Chandarlapaty S, ESR1 ligand-binding domain mutations in hormone-resistant breast cancer, Nature genetics 45(12) (2013) 1439–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [22].Musgrove EA, Sutherland RL, Biological determinants of endocrine resistance in breast cancer, Nature reviews. Cancer 9(9) (2009) 631–43. [DOI] [PubMed] [Google Scholar]
- [23].Mo Z, Liu M, Yang F, Luo H, Li Z, Tu G, Yang G, GPR30 as an initiator of tamoxifen resistance in hormone-dependent breast cancer, Breast cancer research : BCR 15(6) (2013) R114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [24].Osborne CK, Zhao H, Fuqua SA, Selective estrogen receptor modulators: structure, function, and clinical use, Journal of clinical oncology : official journal of the American Society of Clinical Oncology 18(17) (2000) 3172–86. [DOI] [PubMed] [Google Scholar]
- [25].Pinkerton JV, Goldstein SR, Endometrial safety: a key hurdle for selective estrogen receptor modulators in development, Menopause 17(3) (2010) 642–53. [DOI] [PubMed] [Google Scholar]
- [26].Martinkovich S, Shah D, Planey SL, Arnott JA, Selective estrogen receptor modulators: tissue specificity and clinical utility, Clinical interventions in aging 9 (2014) 1437–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [27].Early G Breast Cancer Trialists’ Collaborative, Davies C, Godwin J, Gray R, Clarke M, Cutter D, Darby S, McGale P, Pan HC, Taylor C, Wang YC, Dowsett M, Ingle J, Peto R, Relevance of breast cancer hormone receptors and other factors to the efficacy of adjuvant tamoxifen: patient-level meta-analysis of randomised trials, Lancet 378(9793) (2011) 771–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [28].Davies C, Pan H, Godwin J, Gray R, Arriagada R, Raina V, Abraham M, Medeiros Alencar VH, Badran A, Bonfill X, Bradbury J, Clarke M, Collins R, Davis SR, Delmestri A, Forbes JF, Haddad P, Hou MF, Inbar M, Khaled H, Kielanowska J, Kwan WH, Mathew BS, Mittra I, Muller B, Nicolucci A, Peralta O, Pernas F, Petruzelka L, Pienkowski T, Radhika R, Rajan B, Rubach MT, Tort S, Urrutia G, Valentini M, Wang Y, Peto R, Adjuvant Tamoxifen G: Longer Against Shorter Collaborative, Long-term effects of continuing adjuvant tamoxifen to 10 years versus stopping at 5 years after diagnosis of oestrogen receptor-positive breast cancer: ATLAS, a randomised trial, Lancet 381(9869) (2013) 805–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [29].van Leeuwen FE, Benraadt J, Coebergh JW, Kiemeney LA, Gimbrere CH, Otter R, Schouten LJ, Damhuis RA, Bontenbal M, Diepenhorst FW, et al. , Risk of endometrial cancer after tamoxifen treatment of breast cancer, Lancet 343(8895) (1994) 448–52. [DOI] [PubMed] [Google Scholar]
- [30].Bergman L, Beelen ML, Gallee MP, Hollema H, Benraadt J, van Leeuwen FE, Risk and prognosis of endometrial cancer after tamoxifen for breast cancer. Comprehensive Cancer Centres’ ALERT Group. Assessment of Liver and Endometrial cancer Risk following Tamoxifen, Lancet 356(9233) (2000) 881–7. [DOI] [PubMed] [Google Scholar]
- [31].Decensi A, Maisonneuve P, Rotmensz N, Bettega D, Costa A, Sacchini V, Salvioni A, Travaglini R, Oliviero P, D’Aiuto G, Gulisano M, Gucciardo G, del Turco MR, Pizzichetta MA, Conforti S, Bonanni B, Boyle P, Veronesi U, Italian Tamoxifen Study G, Effect of tamoxifen on venous thromboembolic events in a breast cancer prevention trial, Circulation 111(5) (2005) 650–6. [DOI] [PubMed] [Google Scholar]
- [32].Meier CR, Jick H, Tamoxifen and risk of idiopathic venous thromboembolism, British journal of clinical pharmacology 45(6) (1998) 608–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [33].Vogel VG, Costantino JP, Wickerham DL, Cronin WM, Cecchini RS, Atkins JN, Bevers TB, Fehrenbacher L, Pajon ER, Wade JL 3rd, Robidoux A, Margolese RG, James J, Runowicz CD, Ganz PA, Reis SE, McCaskill-Stevens W, Ford LG, Jordan VC, Wolmark N, Adjuvant B. National Surgical, Bowel P, Update of the National Surgical Adjuvant Breast and Bowel Project Study of Tamoxifen and Raloxifene (STAR) P-2 Trial: Preventing breast cancer, Cancer prevention research 3(6) (2010) 696–706. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [34].Cummings SR, Eckert S, Krueger KA, Grady D, Powles TJ, Cauley JA, Norton L, Nickelsen T, Bjarnason NH, Morrow M, Lippman ME, Black D, Glusman JE, Costa A, Jordan VC, The effect of raloxifene on risk of breast cancer in postmenopausal women: results from the MORE randomized trial. Multiple Outcomes of Raloxifene Evaluation, Jama 281(23) (1999) 2189–97. [DOI] [PubMed] [Google Scholar]
- [35].Vogel CL, Johnston MA, Capers C, Braccia D, Toremifene for breast cancer: a review of 20 years of data, Clinical breast cancer 14(1) (2014) 1–9. [DOI] [PubMed] [Google Scholar]
- [36].Wakeling AE, Similarities and distinctions in the mode of action of different classes of antioestrogens, Endocrine-related cancer 7(1) (2000) 17–28. [DOI] [PubMed] [Google Scholar]
- [37].Addo S, Yates RA, Laight A, A phase I trial to assess the pharmacology of the new oestrogen receptor antagonist fulvestrant on the endometrium in healthy postmenopausal volunteers, British journal of cancer 87(12) (2002) 1354–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [38].Smith IE, Dowsett M, Aromatase inhibitors in breast cancer, The New England journal of medicine 348(24) (2003) 2431–42. [DOI] [PubMed] [Google Scholar]
- [39].Baum M, Buzdar A, Cuzick J, Forbes J, Houghton J, Howell A, Sahmoud T, A.T. Group, Anastrozole alone or in combination with tamoxifen versus tamoxifen alone for adjuvant treatment of postmenopausal women with early-stage breast cancer: results of the ATAC (Arimidex, Tamoxifen Alone or in Combination) trial efficacy and safety update analyses, Cancer 98(9) (2003) 1802–10. [DOI] [PubMed] [Google Scholar]
- [40].Bonneterre J, Buzdar A, Nabholtz JM, Robertson JF, Thurlimann B, von Euler M, Sahmoud T, Webster A, Steinberg M, Writing C. Arimidex, Investigators Committee M, Anastrozole is superior to tamoxifen as first-line therapy in hormone receptor positive advanced breast carcinoma, Cancer 92(9) (2001) 2247–58. [DOI] [PubMed] [Google Scholar]
- [41].Kidwell KM, Harte SE, Hayes DF, Storniolo AM, Carpenter J, Flockhart DA, Stearns V, Clauw DJ, Williams DA, Henry NL, Patient-reported symptoms and discontinuation of adjuvant aromatase inhibitor therapy, Cancer 120(16) (2014) 2403–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [42].Zivian MT, and Salgado B,, Side Effects Revisited: Women’s Experiences With Aromatase Inhibitors. A Report from Breast Cancer Action, 2008. https://bcaction.org/site-content/uploads/2011/03/AI-Report-June-2008-Final-ONLINE.pdf. [Google Scholar]
- [43].Schover LR, Baum GP, Fuson LA, Brewster A, Melhem-Bertrandt A, Sexual problems during the first 2 years of adjuvant treatment with aromatase inhibitors, The journal of sexual medicine 11(12) (2014) 3102–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [44].Ma CX, Sanchez CG, Ellis MJ, Predicting endocrine therapy responsiveness in breast cancer, Oncology 23(2) (2009) 133–42. [PubMed] [Google Scholar]
- [45].Ma CX, Reinert T, Chmielewska I, Ellis MJ, Mechanisms of aromatase inhibitor resistance, Nature reviews. Cancer 15(5) (2015) 261–75. [DOI] [PubMed] [Google Scholar]
- [46].Jeselsohn R, Buchwalter G, De Angelis C, Brown M, Schiff R, ESR1 mutations-a mechanism for acquired endocrine resistance in breast cancer, Nature reviews. Clinical oncology 12(10) (2015) 573–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [47].Ignatov A, Ignatov T, Roessner A, Costa SD, Kalinski T, Role of GPR30 in the mechanisms of tamoxifen resistance in breast cancer MCF-7 cells, Breast cancer research and treatment 123(1) (2010) 87–96. [DOI] [PubMed] [Google Scholar]
- [48].Zhang QX, Borg A, Wolf DM, Oesterreich S, Fuqua SA, An estrogen receptor mutant with strong hormone-independent activity from a metastatic breast cancer, Cancer research 57(7) (1997) 1244–9. [PubMed] [Google Scholar]
- [49].Weis KE, Ekena K, Thomas JA, Lazennec G, Katzenellenbogen BS, Constitutively active human estrogen receptors containing amino acid substitutions for tyrosine 537 in the receptor protein, Molecular endocrinology 10(11) (1996) 1388–98. [DOI] [PubMed] [Google Scholar]
- [50].Robinson DR, Wu YM, Vats P, Su F, Lonigro RJ, Cao X, Kalyana-Sundaram S, Wang R, Ning Y, Hodges L, Gursky A, Siddiqui J, Tomlins SA, Roychowdhury S, Pienta KJ, Kim SY, Roberts JS, Rae JM, Van Poznak CH, Hayes DF, Chugh R, Kunju LP, Talpaz M, Schott AF, Chinnaiyan AM, Activating ESR1 mutations in hormone-resistant metastatic breast cancer, Nature genetics 45(12) (2013) 1446–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [51].Jeselsohn R, Yelensky R, Buchwalter G, Frampton G, Meric-Bernstam F, Gonzalez-Angulo AM, Ferrer-Lozano J, Perez-Fidalgo JA, Cristofanilli M, Gomez H, Arteaga CL, Giltnane J, Balko JM, Cronin MT, Jarosz M, Sun J, Hawryluk M, Lipson D, Otto G, Ross JS, Dvir A, Soussan-Gutman L, Wolf I, Rubinek T, Gilmore L, Schnitt S, Come SE, Pusztai L, Stephens P, Brown M, Miller VA, Emergence of constitutively active estrogen receptor-alpha mutations in pretreated advanced estrogen receptor-positive breast cancer, Clinical cancer research : an official journal of the American Association for Cancer Research 20(7) (2014) 1757–1767. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [52].Li S, Shen D, Shao J, Crowder R, Liu W, Prat A, He X, Liu S, Hoog J, Lu C, Ding L, Griffith OL, Miller C, Larson D, Fulton RS, Harrison M, Mooney T, McMichael JF, Luo J, Tao Y, Goncalves R, Schlosberg C, Hiken JF, Saied L, Sanchez C, Giuntoli T, Bumb C, Cooper C, Kitchens RT, Lin A, Phommaly C, Davies SR, Zhang J, Kavuri MS, McEachern D, Dong YY, Ma C, Pluard T, Naughton M, Bose R, Suresh R, McDowell R, Michel L, Aft R, Gillanders W, DeSchryver K, Wilson RK, Wang S, Mills GB, Gonzalez-Angulo A, Edwards JR, Maher C, Perou CM, Mardis ER, Ellis MJ, Endocrine-therapy-resistant ESR1 variants revealed by genomic characterization of breast-cancer-derived xenografts, Cell reports 4(6) (2013) 1116–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [53].Fanning SW, Mayne CG, Dharmarajan V, Carlson KE, Martin TA, Novick SJ, Toy W, Green B, Panchamukhi S, Katzenellenbogen BS, Tajkhorshid E, Griffin PR, Shen Y, Chandarlapaty S, Katzenellenbogen JA, Greene GL, Estrogen receptor alpha somatic mutations Y537S and D538G confer breast cancer endocrine resistance by stabilizing the activating function-2 binding conformation, eLife 5 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [54].Toy W, Weir H, Razavi P, Lawson M, Goeppert AU, Mazzola AM, Smith A, Wilson J, Morrow C, Wong WL, De Stanchina E, Carlson KE, Martin TS, Uddin S, Li Z, Fanning S, Katzenellenbogen JA, Greene G, Baselga J, Chandarlapaty S, Activating ESR1 Mutations Differentially Affect the Efficacy of ER Antagonists, Cancer discovery 7(3) (2017) 277–287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [55].Chandarlapaty S, Chen D, He W, Sung P, Samoila A, You D, Bhatt T, Patel P, Voi M, Gnant M, Hortobagyi G, Baselga J, Moynahan ME, Prevalence of ESR1 Mutations in Cell-Free DNA and Outcomes in Metastatic Breast Cancer: A Secondary Analysis of the BOLERO-2 Clinical Trial, JAMA oncology 2(10) (2016) 1310–1315. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [56].Jeselsohn R, Bergholz JS, Pun M, Cornwell M, Liu W, Nardone A, Xiao T, Li W, Qiu X, Buchwalter G, Feiglin A, Abell-Hart K, Fei T, Rao P, Long H, Kwiatkowski N, Zhang T, Gray N, Melchers D, Houtman R, Liu XS, Cohen O, Wagle N, Winer EP, Zhao J, Brown M, Allele-Specific Chromatin Recruitment and Therapeutic Vulnerabilities of ESR1 Activating Mutations, Cancer cell 33(2) (2018) 173–186 e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [57].Spoerke JM, Gendreau S, Walter K, Qiu J, Wilson TR, Savage H, Aimi J, Derynck MK, Chen M, Chan IT, Amler LC, Hampton GM, Johnston S, Krop I, Schmid P, Lackner MR, Heterogeneity and clinical significance of ESR1 mutations in ER-positive metastatic breast cancer patients receiving fulvestrant, Nature communications 7 (2016) 11579. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [58].Boer K, Fulvestrant in advanced breast cancer: evidence to date and place in therapy, Therapeutic advances in medical oncology 9(7) (2017) 465–479. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [59].Turner NC, Slamon DJ, Ro J, Bondarenko I, Im SA, Masuda N, Colleoni M, DeMichele A, Loi S, Verma S, Iwata H, Harbeck N, Loibl S, Andre F, Puyana Theall K, Huang X, Giorgetti C, Bartlett C. Huang, Cristofanilli M, Overall Survival with Palbociclib and Fulvestrant in Advanced Breast Cancer, The New England journal of medicine 379(20) (2018) 1926–1936. [DOI] [PubMed] [Google Scholar]
- [60].Fribbens C, O’Leary B, Kilburn L, Hrebien S, Garcia-Murillas I, Beaney M, Cristofanilli M, Andre F, Loi S, Loibl S, Jiang J, Bartlett CH, Koehler M, Dowsett M, Bliss JM, Johnston SR, Turner NC, Plasma ESR1 Mutations and the Treatment of Estrogen Receptor-Positive Advanced Breast Cancer, Journal of clinical oncology : official journal of the American Society of Clinical Oncology 34(25) (2016) 2961–8. [DOI] [PubMed] [Google Scholar]
- [61].van Kruchten M, de Vries EG, Glaudemans AW, van Lanschot MC, van Faassen M, Kema IP, Brown M, Schroder CP, de Vries EF, Hospers GA, Measuring residual estrogen receptor availability during fulvestrant therapy in patients with metastatic breast cancer, Cancer discovery 5(1) (2015) 72–81. [DOI] [PubMed] [Google Scholar]
- [62].Liu J, Zheng S, Akerstrom VL, Yuan C, Ma Y, Zhong Q, Zhang C, Zhang Q, Guo S, Ma P, Skripnikova EV, Bratton MR, Pannuti A, Miele L, Wiese TE, Wang G, Fulvestrant-3 Boronic Acid (ZB716): An Orally Bioavailable Selective Estrogen Receptor Downregulator (SERD), Journal of medicinal chemistry 59(17) (2016) 8134–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [63].Zhang C, Guo S, Yang L, Liu J, Zheng S, Zhong Q, Zhang Q, Wang G, Metabolism, pharmacokinetics, and bioavailability of ZB716, a Steroidal Selective Estrogen Receptor Downregulator (SERD), Oncotarget 8(61) (2017) 103874–103889. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [64].Guo S, Zhang C, Bratton M, Mottamal M, Liu J, Ma P, Zheng S, Zhong Q, Yang L, Wiese TE, Wu Y, Ellis MJ, Matossian M, Burow ME, Miele L, Houtman R, Wang G, ZB716, a steroidal selective estrogen receptor degrader (SERD), is orally efficacious in blocking tumor growth in mouse xenograft models, Oncotarget 9(6) (2018) 6924–6937. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [65].De Savi C, Bradbury RH, Rabow AA, Norman RA, de Almeida C, Andrews DM, Ballard P, Buttar D, Callis RJ, Currie GS, Curwen JO, Davies CD, Donald CS, Feron LJ, Gingell H, Glossop SC, Hayter BR, Hussain S, Karoutchi G, Lamont SG, MacFaul P, Moss TA, Pearson SE, Tonge M, Walker GE, Weir HM, Wilson Z, Optimization of a Novel Binding Motif to (E)-3-(3,5-Difluoro-4-((1R,3R)-2-(2-fluoro-2-methylpropyl)-3-methyl-2,3,4,9-tetra hydro-1H-pyrido[3,4-b]indol-1-yl)phenyl)acrylic Acid (AZD9496), a Potent and Orally Bioavailable Selective Estrogen Receptor Downregulator and Antagonist, Journal of medicinal chemistry 58(20) (2015) 8128–40. [DOI] [PubMed] [Google Scholar]
- [66].Weir HM, Bradbury RH, Lawson M, Rabow AA, Buttar D, Callis RJ, Curwen JO, de Almeida C, Ballard P, Hulse M, Donald CS, Feron LJ, Karoutchi G, MacFaul P, Moss T, Norman RA, Pearson SE, Tonge M, Davies G, Walker GE, Wilson Z, Rowlinson R, Powell S, Sadler C, Richmond G, Ladd B, Pazolli E, Mazzola AM, D’Cruz C, De Savi C, AZD9496: An Oral Estrogen Receptor Inhibitor That Blocks the Growth of ER-Positive and ESR1-Mutant Breast Tumors in Preclinical Models, Cancer research 76(11) (2016) 3307–18. [DOI] [PubMed] [Google Scholar]
- [67].Hamilton EP, Patel MR, Armstrong AC, Baird RD, Jhaveri K, Hoch M, Klinowska T, Lindemann JPO, Morgan SR, Schiavon G, Weir HM, Im SA, A First-in-Human Study of the New Oral Selective Estrogen Receptor Degrader AZD9496 for ER(+)/HER2(−) Advanced Breast Cancer, Clinical cancer research : an official journal of the American Association for Cancer Research 24(15) (2018) 3510–3518. [DOI] [PubMed] [Google Scholar]
- [68].Wardell SE, Yllanes AP, Baker JG, Baldi RM, Krebs TK, Sorrentino J, Bisi J, Strum J, Norris JD, Abstract 5641: Effects of G1T48, a novel orally bioavailable selective estrogen receptor degrader (SERD), and the CDK4/6 inhibitor, G1T38, on tumor growth in animal models of endocrine resistant breast cancer, Cancer research 77(13 Supplement) (2017) 5641–5641. [Google Scholar]
- [69].Tria GS, Abrams T, Baird J, Burks HE, Firestone B, Gaither LA, Hamann LG, He G, Kirby CA, Kim S, Lombardo F, Macchi KJ, McDonnell DP, Mishina Y, Norris JD, Nunez J, Springer C, Sun Y, Thomsen NM, Wang C, Wang J, Yu B, Tiong-Yip CL, Peukert S, Discovery of LSZ102, a Potent, Orally Bioavailable Selective Estrogen Receptor Degrader (SERD) for the Treatment of Estrogen Receptor Positive Breast Cancer, Journal of medicinal chemistry 61(7) (2018) 2837–2864. [DOI] [PubMed] [Google Scholar]
- [70].Dickler M, Villanueva R, Perez Fidalgo J, Mayer I, Boni V, Winer E, Hamilton E, Bellet M, Urruticoechea A, Gonzalez-Martin A, Cortes J, Martin M, Giltnane J, Gates M, Cheeti S, Fredrickson J, Wang X, Friedman L, Spoerke J, Metcalfe C, Liu L, Li R, Morley R, McCurry U, Chan I, Mueller L, Milan S, Lauchle J, Humke E, Bardia A, Abstract PD5–10: A first-in-human phase I study to evaluate the oral selective estrogen receptor degrader (SERD), GDC-0927, in postmenopausal women with estrogen receptor positive (ER+) HER2-negative metastatic breast cancer (BC), Cancer research 78(4 Supplement) (2018) PD5–10-PD5–10. [Google Scholar]
- [71].Shomali M, Cheng J, Koundinya M, Weinstein M, Malkova N, Sun F, Hebert A, Cindachao M, Hoffman D, McManus J, Levit M, Pollard J, Vincent S, Besret L, Adrian F, Winter C, El-Ahmad Y, Halley F, Hsu K, Lager J, Garcia-Echeverria C, Bouaboula M, Abstract P3–04-05: Identification of SAR439859, an orally bioavailable selective estrogen receptor degrader (SERD) that has strong antitumor activity in wild-type and mutant ER+ breast cancer models, Cancer research 77(4 Supplement) (2017) P3–04-05-P3–04-05. [Google Scholar]
- [72].Bouaboula M, Shomali M, Cheng J, Malkova N, Sun F, Koundinya M, Guo Z, Poirier S, Levit M, Hoffman D, Cao H, Bestret L, Adrian F, Winter C, El-Ahmad Y, Vincent S, Halley F, McCort G, Schio L, Richon V, Cheng H, Hsu K, Soria C, Cohen P, Lager J, Garcia-Echeverria C, Debussche L, Abstract 943: SAR439859, an orally bioavailable selective estrogen receptor degrader (SERD) that demonstrates robust antitumor efficacy and limited cross-resistance in ER+ breast cancer, Cancer research 78(13 Supplement) (2018) 943–943. [Google Scholar]
- [73].Juric D, Curigliano G, Cresta S, Yap Y-S, Terret C, Duhoux F, Takahashi S, Kundamal N, Bhansali S, Liao S, Crystal A, Jhaveri K, Abstract P5–21-04: Phase I/Ib study of the SERD LSZ102 alone or in combination with ribociclib in ER+ breast cancer, Cancer research 78(4 Supplement) (2018) P5–21-04-P5–21-04. [Google Scholar]
- [74].Lewis-Wambi JS, Kim H, Curpan R, Grigg R, Sarker MA, Jordan VC, The selective estrogen receptor modulator bazedoxifene inhibits hormone-independent breast cancer cell growth and down-regulates estrogen receptor alpha and cyclin D1, Molecular pharmacology 80(4) (2011) 610–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [75].Wardell SE, Nelson ER, ^Chao CA, McDonnell DP, Bazedoxifene exhibits antiestrogenic activity in animal models of tamoxifen-resistant breast cancer: implications for treatment of advanced disease, Clinical cancer research : an official journal of the American Association for Cancer Research 19(9) (2013) 2420–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [76].Wardell SE, Ellis MJ, Alley HM, Eisele K, VanArsdale T, Dann SG, Arndt KT, Primeau T, Griffin E, Shao J, Crowder R, Lai JP, Norris JD, McDonnell DP, Li S, Efficacy of SERD/SERM Hybrid-CDK4/6 Inhibitor Combinations in Models of Endocrine Therapy-Resistant Breast Cancer, Clinical cancer research : an official journal of the American Association for Cancer Research 21(22) (2015) 5121–5130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [77].Tikoo D, Gupta M, Duavee: a tissue-selective estrogen complex for menopausal symptoms and prevention of osteoporosis, 2017 4(2) (2017) 6. [Google Scholar]
- [78].Fanning SW, Jeselsohn R, Dharmarajan V, Mayne CG, Karimi M, Buchwalter G, Houtman R, Toy W, Fowler CE, Han R, Laine M, Carlson KE, Martin TA, Nowak J, Nwachukwu JC, Hosfield DJ, Chandarlapaty S, Tajkhorshid E, Nettles KW, Griffin PR, Shen Y, Katzenellenbogen JA, Brown M, Greene GL, The SERM/SERD bazedoxifene disrupts ESR1 helix 12 to overcome acquired hormone resistance in breast cancer cells, eLife 7 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [79].Ignatov A, Ignatov T, Weissenborn C, Eggemann H, Bischoff J, Semczuk A, Roessner A, Costa SD, Kalinski T, G-protein-coupled estrogen receptor GPR30 and tamoxifen resistance in breast cancer, Breast cancer research and treatment 128(2) (2011) 457–66. [DOI] [PubMed] [Google Scholar]
- [80].Ignatov T, Claus M, Nass N, Haybaeck J, Seifert B, Kalinski T, Ortmann O, Ignatov A, G-protein-coupled estrogen receptor GPER-1 expression in hormone receptor-positive breast cancer is associated with poor benefit of tamoxifen, Breast cancer research and treatment (2018). [DOI] [PubMed] [Google Scholar]
- [81].Dennis MK, Burai R, Ramesh C, Petrie WK, Alcon SN, Nayak TK, Bologa CG, Leitao A, Brailoiu E, Deliu E, Dun NJ, Sklar LA, Hathaway HJ, Arterburn JB, Oprea TI, Prossnitz ER, In vivo effects of a GPR30 antagonist, Nature chemical biology 5(6) (2009) 421–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [82].Vivacqua A, Bonofiglio D, Albanito L, Madeo A, Rago V, Carpino A, Musti AM, Picard D, Ando S, Maggiolini M, 17beta-estradiol, genistein, and 4-hydroxytamoxifen induce the proliferation of thyroid cancer cells through the g protein-coupled receptor GPR30, Molecular pharmacology 70(4) (2006) 1414–23. [DOI] [PubMed] [Google Scholar]
- [83].Catalano S, Giordano C, Panza S, Chemi F, Bonofiglio D, Lanzino M, Rizza P, Romeo F, Fuqua SA, Maggiolini M, Ando S, Barone I, Tamoxifen through GPER upregulates aromatase expression: a novel mechanism sustaining tamoxifen-resistant breast cancer cell growth, Breast cancer research and treatment 146(2) (2014) 273–85. [DOI] [PubMed] [Google Scholar]
- [84].Zekas E, Prossnitz ER, Estrogen-mediated inactivation of FOXO3a by the G protein-coupled estrogen receptor GPER, BMC cancer 15 (2015) 702. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [85].Calnan DR, Brunet A, The FoxO code, Oncogene 27(16) (2008) 2276–88. [DOI] [PubMed] [Google Scholar]
- [86].Brunet A, Bonni A, Zigmond MJ, Lin MZ, Juo P, Hu LS, Anderson MJ, Arden KC, Blenis J, Greenberg ME, Akt promotes cell survival by phosphorylating and inhibiting a Forkhead transcription factor, Cell 96(6) (1999) 857–68. [DOI] [PubMed] [Google Scholar]
- [87].Wen Q, Duan X, Liao R, Little P, Gao G, Jiang H, Lalit S, Quirion R, Zheng W, Characterization of intracellular translocation of Forkhead transcription factor O (FoxO) members induced by NGF in PC12 cells, Neuroscience letters 498(1) (2011) 31–6. [DOI] [PubMed] [Google Scholar]
- [88].Ariazi EA, Brailoiu E, Yerrum S, Shupp HA, Slifker MJ, Cunliffe HE, Black MA, Donato AL, Arterburn JB, Oprea TI, Prossnitz ER, Dun NJ, Jordan VC, The G protein-coupled receptor GPR30 inhibits proliferation of estrogen receptor-positive breast cancer cells, Cancer research 70(3) (2010) 1184–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [89].Weissenborn C, Ignatov T, Poehlmann A, Wege AK, Costa SD, Zenclussen AC, Ignatov A, GPER functions as a tumor suppressor in MCF-7 and SK-BR-3 breast cancer cells, Journal of cancer research and clinical oncology 140(4) (2014) 663–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [90].Bologa CG, Revankar CM, Young SM, Edwards BS, Arterburn JB, Kiselyov AS, Parker MA, Tkachenko SE, Savchuck NP, Sklar LA, Oprea TI, Prossnitz ER, Virtual and biomolecular screening converge on a selective agonist for GPR30, Nature chemical biology 2(4) (2006) 207–12. [DOI] [PubMed] [Google Scholar]
- [91].Petrie WK, Dennis MK, Hu C, Dai D, Arterburn JB, Smith HO, Hathaway HJ, Prossnitz ER, G protein-coupled estrogen receptor-selective ligands modulate endometrial tumor growth, Obstetrics and gynecology international 2013 (2013) 472720. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [92].Filardo EJ, Quinn JA, Bland KI, Frackelton AR Jr., Estrogen-induced activation of Erk-1 and Erk-2 requires the G protein-coupled receptor homolog, GPR30, and occurs via trans-activation of the epidermal growth factor receptor through release of HB-EGF, Molecular endocrinology 14(10) (2000) 1649–60. [DOI] [PubMed] [Google Scholar]
- [93].Prossnitz ER, Arterburn JB, International Union of Basic and Clinical Pharmacology. XCVII. G Protein-Coupled Estrogen Receptor and Its Pharmacologic Modulators, Pharmacological reviews 67(3) (2015) 505–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [94].Lappano R, Santolla MF, Pupo M, Sinicropi MS, Caruso A, Rosano C, Maggiolini M, MIBE acts as antagonist ligand of both estrogen receptor alpha and GPER in breast cancer cells, Breast cancer research : BCR 14(1) (2012) R12. [DOI] [PMC free article] [PubMed] [Google Scholar]




