Skip to main content
Endocrinology logoLink to Endocrinology
. 2024 May 8;165(6):bqae038. doi: 10.1210/endocr/bqae038

Interrogating Estrogen Signaling Pathways in Human ER-Positive Breast Cancer Cells Forming Bone Metastases in Mice

Julia N Cheng 1, Jennifer B Frye 2, Susan A Whitman 3, Sima Ehsani 4, Simak Ali 5, Janet L Funk 6,
PMCID: PMC11076418  PMID: 38715255

Abstract

Breast cancer bone metastases (BMET) are incurable, primarily osteolytic, and occur most commonly in estrogen receptor-α positive (ER+) breast cancer. ER+ human breast cancer BMET modeling in mice has demonstrated an estrogen (E2)-dependent increase in tumor-associated osteolysis and bone-resorbing osteoclasts, independent of estrogenic effects on tumor proliferation or bone turnover, suggesting a possible mechanistic link between tumoral ERα-driven osteolysis and ER+ bone progression. To explore this question, inducible secretion of the osteolytic factor, parathyroid hormone–related protein (PTHrP), was utilized as an in vitro screening bioassay to query the osteolytic potential of estrogen receptor- and signaling pathway–specific ligands in BMET-forming ER+ human breast cancer cells expressing ERα, ERß, and G protein–coupled ER. After identifying genomic ERα signaling, also responsibility for estrogen's proliferative effects, as necessary and sufficient for osteolytic PTHrP secretion, in vivo effects of a genomic-only ER agonist, estetrol (E4), on osteolytic ER+ BMET progression were examined. Surprisingly, while pharmacologic effects of E4 on estrogen-dependent tissues, including bone, were evident, E4 did not support osteolytic BMET progression (vs robust E2 effects), suggesting an important role for nongenomic ER signaling in ER+ metastatic progression at this site. Because bone effects of E4 did not completely recapitulate those of E2, the relative importance of nongenomic ER signaling in tumor vs bone cannot be ascertained here. Nonetheless, these intriguing findings suggest that targeted manipulation of estrogen signaling to mitigate ER+ metastatic progression in bone may require a nuanced approach, considering genomic and nongenomic effects of ER signaling on both sides of the tumor/bone interface.

Keywords: breast cancer, bone metastases, estrogen receptor-alpha, estradiol, estetrol, nongenomic


Breast cancer tumors expressing estrogen receptor-α (ER+) are the most commonly diagnosed subtype of breast cancer (1), and also account for more than 70% of clinically evident osteolytic breast cancer bone metastases (BMETs) (2-5) despite similar initial rates (prior to diagnosis) of clinically silent tumor cell dissemination to the bone by ER-negative tumors (6-9). Indeed, bone is the first site of metastatic disease in two-thirds of women who develop ER+ metastatic breast cancer (MBC) (10). These clinical observations, taken together with the high concordance of ERα expression between primary ER+ tumors and BMETs (11-13), suggest a possible role of bone-specific tumoral ERα signaling in driving the high propensity of ER+ breast cancers to form clinical BMET.

It is well understood that estrogen signaling promotes ER+ tumor proliferation (14), making adjuvant endocrine therapy, such as selective estrogen receptor modulators (SERMs), aromatase inhibitors (AIs), and gonadotropin-releasing hormone (GnRH) agonists a cornerstone of ER+ breast cancer treatment (1, 15). However, 30% of patients with ER+ breast cancer will relapse with advanced disease (16, 17), which remains incurable and, in more than 20% of cases, is associated with ERα gene (ESR1) activating mutations acquired during aromatase inhibitor therapy by tumor cells at metastatic sites (17-21). In addition, current bone-targeting agents mitigating BMET-induced fractures, bone pain, hypercalcemia, and other skeletal related events (SREs), while helpful, are not curative (22). Thus, there is a clinical need for more effective means of targeting bone-disseminated ER+ tumor cells. A better understanding of the biology of tumoral estrogen signaling within the estrogen-responsive bone microenvironment may assist with this goal.

To address this question, because most breast cancer bone metastases are osteolytic (23), and osteolysis not only allows for tumor expansion within bone, but is a bone-specific process, our laboratory previously postulated that tumoral ER signaling may drive ER+ breast cancer progression specific to bone by stimulating tumor-associated osteolysis (24). Using a mouse model of estrogen-dependent ER+ human breast cancer BMET, data supporting this hypothesis were obtained, demonstrating a 17ß-estradiol (E2) dose-dependent increase in tumor-associated osteolysis and osteoclasts at the bone/tumor interface occurring independently of estrogenic effects on tumor proliferation or bone turnover, which were constant across the E2 dosing range tested (24, 25). Estrogen-inducible parathyroid hormone–related protein (PTHrP), an osteolytic factor with higher expression prevalence reported in clinical breast cancer BMETs (>90%) vs primary tumors or metastases at other sites (26, 27), was also uniquely documented in ER+ breast cancer cells forming robust osteolytic BMET in the model, where in vivo osteolytic BMET progression was dependent on E2, as well as TGFß, which had additive effects to E2 (via Smad-mediated signaling) in stimulating ER+ tumor cell PTHrP secretion (24, 28).

In toto these prior findings support, but do not prove, the concept that tumoral ERα signaling, additive to effects of bone matrix-derived transforming growth factor β (TGFß) signaling (29-31), may contribute to the enhanced risk of osteolytic BMET progression seen clinically in ER+ (vs ER-negative) MBC. However, many questions remain. Among these is the lack of proof that tumoral ERα (vs other estrogen receptors) mediated in vivo and ex vivo pro-osteolytic effects of E2 documented in the prior studies. For example, additional estrogen receptors are co-expressed with ERα in ER+ tumors, including ERβ and the nonclassical G protein–coupled receptor estrogen receptor (GPER) (32-34), along with different ERα isoforms (eg, ERα-46) (34, 35). Thus, net effects of tumoral estrogen signaling in bone-disseminated ER+ breast cancer cells may reflect a complex interplay between multiple receptor types (eg, inhibitory GPER-mediated estrogenic effects on TGFβ signaling in breast cancer cells have been reported) (36).

Cellular ERα signaling is itself complex, involving both: (i) canonical genomic signaling by cytoplasmic ERα, which dimerizes upon ligand binding and travels to the nucleus as a transcription factor to initiate gene expression; and (ii) rapid nongenomic signaling initiated by a membrane-bound ERα fraction, also known as membrane-initiated steroid signaling (MISS) (37, 38), where mitogen-activated protein kinase (MAPK) and other signaling pathways are rapidly activated by membrane-bound ERα located in caveolin-enriched lipid rafts (39). The relative importance and differing roles of genomic vs nongenomic ERα signaling appear tissue-specific and are still being elucidated using pathways specific ligands (eg, estetrol [E4], a nuclear ER agonist and membrane ER antagonist) and mouse models (eg, mice lacking membrane ERα) (34, 40). Among relevant tissues studied: (i) in human ER+ breast cancer cells, nuclear ERα signaling alone (ie, E4-mediated) is sufficient to promote in vitro proliferation and in vivo estrogen-dependent orthotopic tumor progression in mice (41, 42); while (ii) in bone, both genomic and nongenomic ER pathways mediate effects of estrogen (43-45). In contrast to the importance of genomic ER signaling (+/− nongenomic) in ER+ breast tumors and bone, nongenomic ER signaling is a key in the vasculature, which is also of relevance to tumor progression (40).

Studies described here were therefore undertaken to better understand the complexities of estrogen signaling specific to ER+ tumor progression within the ER+ bone microenvironment using the previously studied human ER+ BMET model, which, like all ER+ breast cancer xenografts in mice, requires estrogen supplementation due to low endogenous E2 levels (24, 25). After first documenting expression of ERα, ERß, and GPER in ER+ breast cancer cells forming E2-dependent BMET in vivo, ex vivo effects of ER- and estrogen signaling pathway–specific ligands were queried using estrogen-inducible PTHrP secretion (+/− TGFß) as a clinically relevant screening bioassay for estrogen-dependent osteolytic potential.

Once genomic ERα signaling was identified as necessary and sufficient to induce osteolytic PTHrP in bone tropic ER+ human breast cancer cells (alone or in combination with TGFß), in vivo experiments were undertaken using the E2- (and TGFß-) dependent osteolytic ER+ BMET model, to examine the ability of a genomic-only ER ligand, estetrol (E4), a fetal estrogen capable of supporting orthotopic ER+ tumor progression in vivo (41), to support osteolytic ER+ BMET progression. By comparing in vivo effects of E2 vs E4, alone or in combination (since E4 can antagonize E2 stimulation of nongenomic MISS), the importance of genomic vs nongenomic ER signaling in osteolytic ER+ metastatic progression in bone, a tissue that is also estrogen-responsive, was queried.

Methods

Cell Lines

American Type Culture Collection (ATCC, Manassas, VA) human breast cancer cell lines (MCF-7, T47D, and ZR-75-1), were cultured in E2-replete Dulbecco's modified Eagle's medium (DMEM; Invitrogen, Carlsbad, CA) or RPMI-1640 (Invitrogen), as per ATCC's recommendation, containing 10% heat-inactivated fetal bovine serum (Atlanta Biologicals, Flowery Branch, GA), and 1% penicillin/streptomycin (Thermo Fisher, Waltham, MA) in 37 °C and 5% CO2 in a humidified atmosphere. Bone tropic ER-negative MDA-MB-231 (MDA-SA) cells were generously provided by Dr. Theresa Guise (24, 46-48). BMET-derived ER+ human tumor cell lines (38-2M and 43-4M) were isolated from in vivo BMET in E2-supplemented mice inoculated (intracardiac, IC) with ER+ MCF-7-derived tumor cells, as previously described (28). Two MCF-7-derived clonal lines encoding full length ERα proteins (and 46 kD variants) harboring 1 of 2 activating ESR1 mutations (MCF-7-Y537S CL3, MCF-7-D538G CL4) were generated from MCF-7luc cells (Cambridge Bioscience, Cambridge, UK) as previously described using CRISPR-Cas9 genome editing (49). Authentication of all human tumor cell lines were verified as previously described (47, 49).

Western Blot Analysis

Proteins were isolated from whole cell lysates of cells propagated in normal media, as described above; quantified by Bradford assay (Bio-Rad, Hercules, CA); and analyzed by Western blots with confirmation of even protein loading as previously described (28, 50). Blots were probed with primary rabbit antibodies against ERα (#8644, Cell Signaling Technology [CST], Danvers, MA; RRID:AB_2617128) followed by HRP-conjugated anti-rabbit secondary antibody (#7074, CST; RRID:AB_2099233) (28); or with primary mouse antibody against ERβ (#GTX70182, GeneTex, Irvine, CA; AB_370375) validated in doxycycline-inducible MDA-MB-231-ERβ cells (51), followed by HRP-conjugated anti-mouse secondary antibody (#7076, CST; RRID:AB_330924); or with primary goat antibody against GPER (#AF5534, R&D Systems, Minneapolis, MN; AB_2112482) followed by HRP-conjugated anti-goat secondary antibody (#R21459, Life Technologies, Carlsbad, CA; RRID:AB_2556529). Blots were visualized by chemiluminescence of SuperSignal West Femto ECL substrate (ThermoFisher). An antibody against β-actin (#4967, CST; RRID:AB_330288) was used as a loading control. Most blots are representative of ≥ 3 separate experiments.

Parathyroid Hormone–Related Protein Assay

For analysis of the tumor-secreted osteolytic factor PTHrP, cells were plated in 24-well tissue culture plates at a density of 1.3 × 105 cells/well, maintained in E2-deplete media (phenol red-free DMEM [Invitrogen], 10% charcoal-stripped fetal bovine serum [Valley Biomedical, Winchester, VA], 1% penicillin/streptomycin [Thermo Fisher], and 200 mM L-glutamine [Sigma-Aldrich, St. Louis, MO]) for 4 days (2 days for ER-negative MDA-SA cells) followed by treatments with TGFβ (5 ng/mL), E2 (10−7 or 10−8 M, as indicated) and/or selective estrogen receptor-specific agonists, dosed according to their relative binding affinity (Fig. 1), for 24, 52, or 72 hours, as indicated, to optimize detection, depending on cell line. Cell numbers remained statistically unchanged from day 0 for each cell line, and between cell lines, after 4 days in E2-deplete media. To assess the effects of various inhibitors on PTHrP secretion, cells were treated for 1 hour prior to addition of indicated ER agonists and/or TGFβ with nystatin (50 ug/mL; Alfa Aesar, Haverhill, MA) (52), rapamycin (1 nM; CST), or a p38 inhibitor SB202190 (10 μM; Selleckchem, Houston, TX) (52). Conditioned media, stored at −80 °C after addition of protease inhibitors (Sigma-Aldrich) was assayed for secreted PTHrP using a commercial immunoradiometric assay (DSL-8100, Beckman Coulter, Brea, CA: AB_3086791). A lack of treatment effect on cell number during the incubation periods was verified using a commercial MTT assay (ATCC).

Figure 1.

Figure 1.

Estrogen receptor ligand structures and relative binding affinities. ERα, ERß, and/or GPER receptor binding affinities for agonist/antagonists studied in vitro and/or in vivo are indicated relative to 17-ß estradiol, whose binding affinity for all 3 receptors is defined as 100%.

Animal Studies

All animal protocols were approved by the Institutional Animal Care and Use Committee at The University of Arizona in accordance with the National Institutes of Health Guide for the Care and Use of Laboratory Animals. Female Foxn1nu athymic nude outbred mice (Envigo, Indianapolis, IN) were housed in plastic cages in laminar flow isolated hoods with access to water and autoclaved mouse chow ad libitum (NIH-31 Modified diet, Envigo) (24, 25, 28, 47, 48). For one set of BMET experiments, 5-week-old mice (n = 10-13/group) were inoculated (1 × 105 cells) via the left cardiac ventricle (IC) as previously described (24, 28, 48) with either: (i) human ER-negative MDA-MB-231 cells without estrogen supplementation; or (ii) individual ER+ human breast cancer cell lines (ATCC MCF-7, T47D, or ZR-75-1; or BMET-derived MCF-7 cells [38-2M, 43-4M]), 3 days post placement of 60-day extended release 17β-estradiol (E2) pellets (0.05, 0.10, 0.36, or 0.72 mg, as indicated [Innovative Research of America, Sarasota, FL]) (24, 28).

In another set of experiments, 5-week-old mice were IC inoculated with MCF-7 BMET-derived ER+ 43-4M cells (1 × 105), with start of E2 and/or E4 treatments 4 days prior to inoculation (28). Treatment groups (n = 7-12/group) included: (i) E2 alone (0.72 mg 60-day extended release pellets [Innovative Research of America, Sarasota, FL]); (ii) E2 pellets plus vehicle (100 μL peanut oil/d; #P2144, Sigma-Aldrich); (iii) E4 (4 mg/kg/d) (#SML1523, Sigma-Aldrich; in peanut oil); or (iv) E2 pellet and E4 (4 mg/kg/d), with daily dosing of E4 or vehicle via gavage for the first 7 days, followed by a 5x/week regimen without consecutive nontreatment days until end of experiment. The daily oral 4 mg/kg E4 dosing strategy using peanut oil as a vehicle was the same as that used by Gerard et al to demonstrate in vivo stimulatory effects of 1 to 10 mg/kg/d E4 on orthotopic MCF-7 tumor growth (and on normal mammary epithelium) (53) when used in isolation, and partial inhibition of E2 driven growth when used in combination with E4 (41). E2 pellet dose used here (0.72 mg pellet), which yielded a maximal induction of ER + BMET in the E2 dose-response experiments, was similar to that used by Gerard et al to support in vivo MCF-7 orthotopic tumor growth (1.7 mg pellet) (41). Because all reported outcomes for E2 alone were unchanged by addition of vehicle, data for these groups were combined for analyses. In a third set of experiments, 4- to 5-week-old mice were IC inoculated (4 × 105 cells) with either: (i) a 1:1 mix of MCF-7 clones harboring clinically common activating ESR1 mutations (MCF-7-Y537S CL3 and MCF-7-D538G CL4) without estrogen supplementation; or (ii) “parental” ESR1-wild-type MCF-7luc cells with, or without, E2 treatment (0.72 mg 60-day pellets) beginning 4 days prior to inoculation, as indicated. At the end of all BMET experiments, mice were euthanized, hind limbs isolated for histologic analyses, and, in some cases (E2/E4 experiment), the fourth and ninth inguinal mammary glands were collected (54), and organ weights (eg, uteri) were recorded.

To assess bone dissemination, additional mice (vs untreated controls; n = 3-5/group) were either pelleted with 0.72 mg E2, or treated by daily oral administration with 4 mg/kg E4 for 3 days prior to IC inoculation with 8 × 105 43-4M cells freshly labeled with Vybrant DiD (Thermo Fisher), as per the manufacturer's instructions, with fluorescent staining remaining detectable for up to 7 days in culture, as previously described (24, 28). Daily E4 administration continued until 3 days postinoculation when mice were euthanized and cells were isolated from the proximal (25%) tibia and imaged for enumeration of DiD-positive cells using the Cy5 filter of a Keyence BZ-X700 fluorescent microscope (Keyence Corporation of America, Itasca, IL), as previously described (24, 28). Results are reported as DiD+ cells/106 bone marrow cells for each tibia. Similarly isolated bone marrow cells from tumor-naïve mice (n = 3) were included as negative controls and cultured 43-4M cells 3 days post DiD-labeling were used as positive controls.

Bone Metastatic Breast Cancer Tumor and Mammary Gland Histology

Hind limbs were removed at end of experiments, fixed, decalcified, and paraffin-embedded for histologic analyses of midsagittal (depth of 400-500 μM) sections (5-6 μM thick), as previously described (24, 28). Sections from age-matched tumor-free mice were stained with hematoxylin and eosin (H&E) to assess effects of E4 vs E2 (vs control) on bone histology and osteoblast numbers (24, 25, 28). Hematoxylin-stained mononuclear cuboidal osteoblasts lining trabecular bone surfaces and tartrate-resistant acid phosphatase (TRAP)-stained multi-nucleated osteoclasts were identified in a blinded fashion 0.25 mm below the proximal tibial growth plate and reported as cell number per mm of bone surface (BS), as previously described (24, 25, 28, 55, 56). Epithelial ER+ breast cancer tumors were immunohistochemically identified at end of all experiments and their size measured in hind leg bones using primary antibodies to pan-cytokeratin (#Z0622, Agilent Dako, Santa Clara, CA; AB_2650434), as previously described (24, 28). Of note, prior immunohistochemical experiments have confirmed the continued expression of ERa in the bone metastatic cells (24, 28). The fourth and ninth inguinal mammary glands were fixed, defatted, and stained with carmine alum as previously described (54) for examination of ductal area and terminal end buds (TEB) in mice treated with E2 and/or E4 vs controls.

Bone Imaging

Radiographs of mouse hind limbs (Faxitron UltraFocus 1000, Faxitron Bioptics, Tucson, AZ) in ER+ or ER-negative breast cancer cell-inoculated mice were obtained weekly to assess osteolytic lesion formation, which was analyzed as previously described in a blinded fashion by 3 independent investigators using ImageJ software (NIH) (24, 28, 47), with osteolytic BMET incidence and/or total hind limb radiographic lesion area reported per mouse, including animals without osteolytic lesions. Of note, no osteoblastic lesions were detected. Because E2 can induce osteolytic osteosarcoma formation in nude mice (25), osteolytic breast cancer ER+ BMETs in each hind limb bone were verified by correlating radiographic lesions with cytokeratin-positive human tumors at end of experiments (24, 57). Areal bone mineral density (aBMD) of the proximal femur, a tumor-free area, was assessed by dual-energy x-ray absorptiometry (DXA; Faxitron UltraFocus 1000) in age-matched mouse hind limbs (with or without [naïve control] inoculated tumor cells) at end of experiments.

Statistical Analysis

Data are reported as mean ± SEM, with statistical significance of two-sided P values defined as P ≤ .05. Statistical differences were determined using Prism 8.0 software (Graphpad, San Diego, CA) for t tests, one- or two-way analyses of variance (ANOVA) with post hoc testing (as indicated), and tests for log-rank.

Results

Bone Tropic ER+ Human Breast Cancer Cells Express ERα, ERβ, and GPER

Western blot analysis was used to interrogate estrogen receptor expression in “bone tropic” human breast cancer cell lines that secrete E2 (if ER+)-inducible and/or TGFß-inducible osteolytic PTHrP and form osteolytic BMET in vivo that are TGFß- and estrogen-dependent (ER+ MCF-7, or MCF-7 BMET-derived 43-4M, 38-2M cells) or estrogen-independent (ER-MDA-MB-231), as compared to “non–bone tropic” ER+ cell lines that do neither (T47D, ZR-75-1) (24, 28) (and Supplementary Fig. S1) (58). GPER protein was detected in all ER + cell lines, independent of bone tropic behavior, but not in bone tropic ER-negative MDA-MB-231 cells (Fig. 2A). ERβ protein, using an antibody validated for Western blot analyses (51), was only readily detected in ER+ cell lines that were bone tropic (MCF-7, 38-2M, 43-4M), with questionable expression in bone tropic ER-negative MDA-MB-231 cells (Fig. 2B). As expected, ERα protein was identified by Western blot analysis in all ER+ breast cancer cell lines, but not in ER-negative MBC-MB-231 cells (Fig. 2B). However, ERα protein levels were much higher in bone tropic cells, where multiple ERα isoforms, including ERα-46, were also readily detected (Fig. 2B, left panel).

Figure 2.

Figure 2.

Estrogen receptor protein expression in human ER+ and ER-negative breast cancer cell lines as assessed by Western analyses. A, Expression of GPER protein in human breast cancer bone tropic cells ER-negative (MBD-MB-231), bone tropic ER+ human breast cancer cells (MCF-7, MCF-7 BMET-derived 38-2M, or 43-4M cells), or non–bone tropic ER+ cells (T47D, ZR-75-1). B, Comparison of ERα (including isoforms, left panel) and ERβ protein expression in ER+ human breast cancer cell lines that are bone tropic (MCF-7 and MCF-7 BMET-derived 38-2M, or 43-4M cells) or non–bone tropic (T47D, ZR-75-1) vs ER-negative bone tropic MBD-MB-231 cells. Note each image is from a single Western blot with intervening cell lines removed.

Nuclear ERα Signaling Is Sufficient to Induce Secretion of a Tumoral Mediator of Osteolysis From Bone Tropic ER+ Human Breast Cancer Cells

Having identified all 3 estrogen receptors in bone tropic human ER+ breast cancer cells where E2 (and TGFß) drive tumor-associated osteolysis in vivo and osteolytic PTHrP secretion in vitro (24, 28) (and Supplementary Fig S1) (58), PTHrP secretion (in control or TGFß stimulated ER+ bone tropic cells) was utilized as an in vitro screening bioassay for estrogen receptor-mediated osteolytic potential, querying a role for ERα vs other estrogen receptors by testing effects of receptor- and signaling pathway–specific ligands using affinity-normalized dosing (Fig. 1). Isolated treatment of 2 different BMET-derived MCF-7 cell lines (38-2M and 43-4M) with the ERα agonist propyl pyrazole triol (PPT), or nuclear ER agonist (and MISS antagonist) estetrol (E4), induced PTHrP secretion 2-fold over the media control, similar to levels induced by E2 (Fig. 3A). In contrast, neither an ERβ agonist (R-diarylpropionitrile, R-DPN), isolated nongenomic ER MISS agonist (pathway preferential estrogen, PaPE-1), nor GPER agonist (G1) significantly altered PTHrP relative to untreated controls (Fig. 3A). Analogous to E2, ERα agonist PPT and nuclear-only ER agonist E4 each also exhibited additive PTHrP secretory effects with TGFβ (Fig. 3B), while agonists for the other receptors (ERβ, ER MISS, and GPER) did not. When agonists lacking isolated effects were combined with agonists sufficient to induce PTHrP expression, no additional synergies were identified; combining either ERβ agonist R-DPN (Fig. 4A) or GPER agonist G1 (Fig. 4B) with ERα agonist PPT did not alter PPT-induced PTHrP secretion in control or TGFß-stimulated cells. Agonists for ERβ (R-DPN) or GPER (G1), which individually were without effect, also did not induce PTHrP secretion when combined in control or TGFβ-stimulated cells (data not shown). Stimulatory effects of the ER nuclear agonist/MISS antagonist E4 on PTHrP were not altered when combined with GPER agonist G1 (Fig. 4C), while in TGFβ-stimulated cells, PTHrP secretion was significantly reduced by G1 addition, as compared to E4 alone. In toto, these data suggest that in ER+ breast cancer cells forming E2 and TGFß-dependent osteolytic BMET in vivo (24, 28), nuclear effects of cytoplasmic ERα are sufficient to mediate both E2-inducible PTHrP secretion and additive effects with TGFβ, without involvement of other estrogen receptors or nongenomic ERα MISS.

Figure 3.

Figure 3.

Effects of estrogen receptor-specific agonists on constitutive or TGFβ-inducible secretion of osteolytic PTHrP from human ER+ breast cancer BMET-derived cells. Effect of estrogen receptor-specific agonists A, on constitutive secretion of PTHrP from MCF-7 BMET-derived 38-2M (48 hours) or 43-4M cells (48-52 hours) or B, in combination with TGFβ (5 ng/mL) treatment in 43-4M cells. Cells were maintained in E2-deplete media for 4 days prior to treatment with E2 at 10−8 M, or the indicated ER agonist: ERα agonist PPT (10−8 M), ERβ agonist R-DPN (10−8 M), nuclear ER agonist E2 (10−6 M), membrane ER agonist PaPE-1 (10−5 M), or GPER agonist G1 (10−8 M); or media control, for 48-52 hours, as indicated above. Secreted PTHrP levels are expressed as fold change from untreated cells (control). Cell number (by MTT assay) was not altered by agonist or TGFβ treatment under the conditions of the experiment (data not shown). *P ≤ .05 vs control, **P ≤ .001 E2 vs E2+ TGFβ, ***P ≤ .01 TGFβ vs E2+ TGFβ, not significant (n.s.) by one-way ANOVA with Holm-Sidak's post-test. Results are representative of up to 8 replicate experiments (n = 4 wells/condition), which in some cases were averaged (n = 4-32/group).

Figure 4.

Figure 4.

Combined effects of estrogen receptor-specific agonists on osteolytic PTHrP secretion from BMET-derived human ER+ breast cancer cells. Effect of PTHrP-inducing ERα agonist (PPT, 10−8 M) on constitutive or TGFβ-stimulated PTHrP secretion from MCF-7 BMET-derived 43-4M cells when combined with (A) ERβ agonist (R-DPN, 10−8 M) or (B) GPER agonist (G1, 10−8 M); or effect of PTHrP-inducing (C) nuclear ER agonist (E4, 10−6 M) when combined with GPER agonist (G1, 10−8 M) (n = 4/group); vs media control, for 52 hours. PTHrP levels are expressed as fold change of control. Cell number (by MTT assay) was not altered by treatments under the conditions of the experiment (data not shown). *P ≤ .01 vs control, **P ≤ .001 as indicated, not significant (n.s.), by one-way ANOVA with Holm-Sidak's post-test.

Other ERα Cell Signaling Pathways Contribute to Inducible PTHrP Secretion From Bone Tropic ER+ Human Breast Cancer Cells

While nuclear/genomic ERα signaling appeared sufficient for PTHrP induction, additional studies were conducted to query possible interactions between genomic vs nongenomic ERα signaling pathways, and involvement of additional intracellular signaling pathways known to alter ERα signal transduction. As E4 has been reported to antagonize E2 stimulated ERα MISS in MCF-7 cells (40), effects of E4 (10−6 M) and E2 (10−8 M) in combination vs isolation were assessed using comparable maximal doses (10−6 M vs 10−8 M, respectively), consistent with the 100-fold lower affinity of E4 (vs E2) for ERα (Fig. 1) (40), and their differential dose-dependent effects in stimulating PTHrP secretion from 43-4M cells (Fig. 5A). Each induced constitutive and TGFβ-stimulated PTHrP secretion to a similar degree without any evidence of an inhibitory effect when used in combination (Fig. 5B), which would have been suggestive of E4 antagonism of an E2-mediated stimulatory ERα MISS effect on PTHrP secretion. Instead, PTHrP levels were higher with combined E2 and E4 treatment, reaching statistical significance in TGFβ-stimulated cells (Fig. 5B), suggestive of possible additive effects of E2/E4 nuclear ERα signaling (potentially less likely given maximal doses used) and/or E4 blockade of an E2-mediated inhibitory ERα MISS effect when combined with TGFß. Pretreatment with rapamycin, an inhibitor of E2-inducible mTOR signaling in 43-4M cells (28) that is reported to facilitate both genomic and nongenomic ERα signaling (59, 60), completely blocked inducible PTHrP secretion in response to E4 or E2 alone, and significantly inhibited their combined effects (Fig. 5C). Inhibition of p38 MAPK, an E2-activated signaling pathway in 43-4M cells (28, 61, 62) that is possibly ER MISS-regulated (59, 63) and can also enhance nuclear ER transcriptional activity (via ER phosphorylation and/or stimulation of nuclear ER coactivators) (64, 65), partially blocked PTHrP induction by both E2 and by E4 (Fig. 5D). Because membrane ERα signaling involves receptor localization in lipid rafts (61, 62), as does TGFß-induced MAPK signaling, which is also induced in 43-4M cells (28), the lipid-raft disruptor nystatin was also examined (52), causing a partial (∼50%) reduction in E2- and TGFβ-induced PTHrP secretion, alone or in combination (Fig. 5E). In toto, PTHrP screening bioassay data were thus consistent with a necessary and sufficient role for nuclear ERα signaling, possibly involving mTOR and p38 MAPK activation, in stimulating osteolytic PTHrP secretion from bone tropic ER+ breast cancer cells in response to E2 or E4. However, the ability of ERα MISS to alter stimulatory ERα nuclear effects in complex ways could not be ruled out, as evidenced by: 1) the partial suppressive effects of lipid-raft disruption on E2 signaling; 2) the stimulatory effects of an ER MISS antagonist, E4, when combined with E2; and 3) the inhibitory effects of an agonist (G1) for GPER (which is reported to interact with membrane ERα) (41) only when combined with E4, with all of the above being most notable in the context of TGFβ signaling, which also likely involves caveolin-enriched lipid rafts (66).

Figure 5.

Figure 5.

ERα signaling pathways mediating osteolytic PTHrP secretion from BMET-derived human ER+ breast cancer cells. A, E2 and E4 dose-dependency of PTHrP secretion in MCF-7 BMET-derived 43-4M cells maintained in E2-deplete media for 4 days prior to treatment with various concentrations of E2 or E4 (M), as indicated, for 52 hours (n = 3-4/group). Effect of E2 (10−8 M), E4 (10−6 M) and/or their combination on PTHrP secretion B, in control or TGFβ (5 mg/mL)-stimulated cells; C, in the presence of an mTOR inhibitor (1 hour pre-treatment with rapamycin [1 nM]); D, in the presence of a p38 inhibitor (1 hour pretreatment with SB202190 [10 μM]); or E, in the presence of a lipid raft disrupting agent (1 hour pretreatment with nystatin [50 μg/mL)]; or media control, for 48-52 hours. BMET-derived 43-4M cells were maintained for 4 days in E2-deplete media prior to addition of treatments. Secreted PTHrP levels are reported as fold change of untreated controls, with results representative of replicate experiments (n = 4 wells/condition), which in some cases were averaged (n = 4-20/group). *P ≤ 0.05 vs respective vehicle control (eg, no inhibitor); #P ≤ .001 vs TGFβ only; $P ≤ .05 vs vehicle only (no TGFß); ^P ≤ .001 as indicated; or not significant (n.s.), by two-way ANOVA with Holm-Sidak's post-test.

ER MISS Is Required to Support Estrogen-dependent ER+ BMET Progression In Vivo

Comparable effects of E4 (to E2) in supporting MCF-7 orthotopic tumor growth in vivo, as reported by Gerard et al (41), and stimulating osteolytic PTHrP secretion (alone or in combination with TGFß), as demonstrated here, suggested that pharmacologic activation of genomic ER signaling alone by E4 could also be sufficient to support estrogen-dependent osteolytic human ER+ tumoral progression within bone. To test this hypothesis, effects of E2 vs E4 were compared in the ER+ BMET model. As previously described (24, 28), osteolytic BMET formed robustly in 43-4M cell-inoculated mice treated with E2 in terms of both osteolytic lesion incidence (Fig. 6A) and size (Fig. 6B), forming bone-destructive tumors with TRAP+ osteoclasts at leading edges (Fig. 7A-7C). However, most notably and contrary to anticipated outcomes, in mice treated with an E4 dose (4 mg/kg) within the range (1-10 mg/kg) previously reported to support orthotopic MCF-7 tumor progression in vivo (41, 42), there was no radiological evidence of osteolytic BMET formation (Fig. 6A-6B). Histologically, while cytokeratin-positive tumor cells were detected in the hind limbs of 43% (3/7) of E4-treated animals (Fig. 6C), histologic lesions were quite small (Fig. 6D) and, while bone-adjacent (Fig. 7D-7F), were without evidence of associated osteolytic activity. A difference in bone dissemination as a potential driver of reduced osteolytic BMET in E4-treated mice was ruled out by detection of similar number of bone-disseminated DiD-labeled 43-4M cells in E2- vs E4-treated (or untreated) mice within 3 days of tumor cell inoculation (Supplementary Fig. S2) (58).

Figure 6.

Figure 6.

Querying roles of genomic and nongenomic ER signaling for estrogen-dependent osteolytic ER+ human BMET progression in mice treated with E4 and/or E2. A, Osteolytic BMET lesion incidence and B, area in hind limbs of 5-week-old mice IC inoculated with 43-4M cells with E2 and/or E4 treatments starting 4 days prior to inoculation. Treatment groups (n = 7-12/group) included: 1) E2 pellets alone (0.72 mg 60-day pellets); 2) E2 pellets plus vehicle (100 μL peanut oil/d); 3) E4 4 mg/kg/d (in peanut oil); or 4) E2 pellet and E4 (4 mg/kg/d), dosed daily with E4 or vehicle via gavage for the first 7 days, followed by a 5x/week regimen without consecutive nontreatment days until end of experiment. Data from groups 1 and 2 were not different and therefore combined. Osteolytic lesion incidence and area in E2 vs E2+ E4 treated groups were not significantly different (n.s.) by log-rank (incidence) or two-way ANOVA with Holm-Sidak's post-test (area). E4-only mice did not form any radiographic osteolytic lesions (open circles C) Histologic incidence of cytokeratin-positive breast cancer tumors and D, cytokeratin-positive tumor area in mid-sections of hind limbs 6 weeks post-43-4M tumor cell inoculation in mice, with treatments as indicated. *P ≤ .05 as indicated, not significant (n.s), by one-way ANOVA with Holm-Sidak's post-test (n = 7-14/group).

Figure 7.

Figure 7.

Bone-disseminated human ER+ cells do not form osteolytic BMET in the absence of ER MISS signaling. Representative image of a single osteolytic ER+ BMET in an E2-treated mouse demonstrated by: (A) H&E staining of breast cancer tumor cells (white asterisk) in bone, and (B) Immunohistochemical (brown) cytokeratin staining of tumor cells, with (C) TRAP+ (fuchsia) staining demonstrating osteoclasts (black arrows) at the tumor/bone interface on a sequential section. In mice treated with pharmacologic doses of E4, which only activates genomic ER signaling, cytokeratin-positive tumors in bone were infrequent (white asterisk), small and usually located adjacent to cortical (D) or trabecular bone (E), without evidence of associated osteoclastogenesis (F, TRAP stain of section sequential to section in E). Abbreviations: cort, cortical bone; gp, growth plate; trab, trabecular bone.

Reduced effects of E4, a nuclear ER-specific agonist (vs E2), suggested nongenomic ER MISS was important for estrogen-dependent ER+ BMET progression; thus, combined effects of E2 with E4, which can antagonize E2-stimulated ER MISS, were examined. Consistent with a possible stimulatory role of ER MISS in E2-treated mice, the incidence (Fig. 6A) and size (Fig. 6B) of osteolytic BMET lesions developing in 43-4M cell-inoculated mice treated with E2 in combination with E4 were lower than in E2-only, as were the incidence (Fig. 6C) and size (Fig. 6D) of cytokeratin-positive histologic tumors in bone; however, these trends did not reach statistical significance.

Sufficiency of In Vivo E4 Dosing for Mediating Pharmacologic Effects

To determine whether the minimal effect of E4 on human ER+ osteolytic BMET progression was due to insufficient steroid bioavailability or dosing, pharmacologic effects of E4 (+/− E2) on estrogen-dependent tissues were examined in the tumor-inoculated mice. Consistent with anabolic bone effects of E2 in the ER+ BMET model that have been well documented by our laboratory and are identical even with > 10-fold lower E2 dosing (24, 25), BMD in tumor-free proximal femurs of E2-treated mice was significantly increased (vs age-matched control mice, Fig. 8A). In E4-treated mice, BMD also significantly increased, but to a lesser degree (+8% [E4] vs +18% [E2], P < .01) (Fig. 8A). In mice treated with E2 in combination with E4, increased BMD in tumor-free proximal femurs was not statistically different than E2 alone (Fig. 8A). Differential pharmacologic effects of E4 vs E2 were also seen in uterine weights, which tended to increase slightly in E2-treated mice (+/− E4), while being significantly reduced by E4 treatment alone in these ovary-intact mice (Fig. 8B). Mammary gland ductal branching and alveoli development in also appeared qualitatively reduced in the presence of E4 (Supplementary Fig. S3) (58), alone or in combination with E2 treatment, as compared to naïve or E2-treated mice, consistent with prior reports (41).

Figure 8.

Figure 8.

E4 dosing was sufficient to induce pharmacologic effects in estrogen-responsive tissues. A, Areal bone mineral density (aBMD; left panel) of BMET-free proximal femurs as measured by DXA 42 days post-tumor-inoculation of 5-week-old mice treated with 0.72 mg E2, 4 mg/kg E4, or a combination of E2 and E4, as compared to naïve controls (n = 14-30/group). BMD are reported as % of control (n = 14-30/group). B, Uterine weights (normalized to body weight) 42 days post-tumor-inoculation of 5-week-old mice treated with 0.72 mg E2, 4 mg/kg E4, or a combination of E2 and E4, as compared to naïve controls (n = 4-21/group). *P ≤ .05 vs control, not significant (n.s.), by one-way ANOVA with Dunnett's post-test.

Anabolic Effects of ER Signaling in the Tumor Microenvironment Are Not Required for ER+ Human Breast Cancer Progression in Bone

In normal age-matched mice treated for 6 weeks with the same E2 dose as in the BMET model, histologic assessment of the proximal tibiae, a common site of human ER-negative or ER+ breast cancer BMET formation in mice (14, 24, 28, 47), reflected a marked increase in bone with (vs age-matched control mice), as has been previously described (25), that was not recapitulated in mice treated with 4 mg/kg/d E4 (Fig. 9A), where changes were modest (Fig. 9A). Osteoblast (Ob) numbers in the tibial metaphysis doubled in response to E2 treatment but remained unchanged in E4-treated mice (Fig. 9B), consistent with prior reports of a possible role for ER MISS in mediating estrogen effects on bone-forming osteoblasts (44). Osteoclast (Oc) numbers on trabecular bone surfaces were unchanged by either treatment (Fig. 9C) in these ovary-intact mice, suggesting that ER MISS effects on bone anabolism could be key drivers of the more marked E2 (vs E4) effects on the tumor microenvironment.

Figure 9.

Figure 9.

Nongenomic ER signaling contributes to estrogenic effects on bone microenvironment. Comparison of E4 vs E2 effects on normal bone as demonstrated by (A) representative histology of proximal tibiae (mid-sagittal H&E-stained sections), and number of (B) osteoblasts (Ob), or (C) osteoclasts (Oc) per bone surface (N./BS [mm]) in tibial metaphyses after 6 weeks of treating 5-week-old mice with 0.72 mg E2 or 4 mg/kg/d E2, as compared to age-matched controls (n = 4-10/group). *P ≤ .05, **P ≤ .01, or ****P ≤ .0001 vs control or indicated comparison by one-way ANOVA with Holm-Sidak's post-test.

Bone effects of E2 across a range of doses supporting ER+ human breast cancer BMET progression in mice are uniform (not dose-dependent) (24, 25) and can increase ER-negative BMET size (but not incidence) (24), suggesting that bone effects of estrogen are tumor-promoting. Given the less marked bone effects of E4 (vs E2), an experiment was undertaken to determine whether isolated ERα signaling in tumor (and not bone) could still support ER+ BMET progression. BMET formation in mice inoculated with ER+ MCF-7 cells with ESR1 activating mutations vs ESR1 wild-type cells was examined in the absence of E2 supplementation. When E2 supplementation was not given (and associated bone effects were absent), MCF-7 cells harboring activating ESR1 mutations formed BMET (with an incidence similar to ESR1 wild-type cells in mice supplemented with E2), while ESR1 wild-type MCF-7 cells did not (Supplementary Fig. S4) (58).

Discussion

The role of estrogen signaling in ER+ breast cancer, the most common subtype, is both simple and complex. In breast cancer cells expressing ERα (which increases with menopause in normal breast epithelium) (67), nuclear ER signaling clearly drives breast cancer cell proliferation, with adjuvant endocrine therapy markedly reducing progression (68). And yet, in advanced ER+ MBC, where most women have BMET, anti-estrogen treatments are not curative, with tumor cell acquisition of activating ESR1 mutations contributing to treatment resistance in some cases (18). At the same time, and somewhat paradoxically, indefinite use of presumably cytostatic endocrine treatments in early breast cancer is not required to yield benefit. However, unique to ER+ breast cancer, rates of metastatic progression after cessation of endocrine treatment for women with early-stage disease remains constant for decades (17), with bone often the first clinically evident site of metastatic spread (10). The experimental results reported here are intended to shed additional light on the pathogenic role of genomic vs nongenomic ERα on both sides of the ER+ tumor/ER+ bone interface in driving breast cancer progression at this site.

Prior documentation of estrogen-dependent in vivo osteolysis in an ER+ BMET model and in vitro secretion from these ER+ BMET-forming cells of osteolytic PTHrP, a peptide expressed in most clinical bone metastases (26, 27), were suggestive of ERα-mediated osteolytic effects by bone-disseminated ER+ tumor cells. In vitro screening bioassay results proved this postulate for estrogen-regulated PTHrP secretion, which was ERα-mediated in BMET-forming ER+ cell lines that were also unique among the cell lines studied (bone tropic ER-negative or ER+ cells, and non–bone tropic ER+ cells) for their expression of all 3 estrogen receptors (ERα, ERß, GPER), a common feature in clinical breast cancer (32-35). Moreover, genomic (nuclear) ERα signaling specifically, which also drives ER+ breast cancer cell proliferation and in vivo orthotopic tumor progression (41), was necessary and sufficient for osteolytic PTHrP secretion, including additive effects with TGFß, a signaling pathway on which these ER+ breast cancer cells also depend for in vivo BMET progression (28). Inhibitor experiments suggesting involvement of both mTOR, which is constitutively active in these ER+ bone tropic cells (28) and translocates with ER to the nucleus in MCF-7 cells (60), and p38 MAPK, which can enhance nuclear ER signaling (transcription) (64, 65), were also consistent with nuclear ER signaling, while involvement of these pathways with ER MISS is also possible (59, 63). In further support of genomic ER signaling driving PTHrP secretion, stimulatory effects of E2/E4 combined with TGFß were also consistent with putative nuclear interactions between ERα and TGFβ-regulated Smad transcription factors, including direct ER binding (69) and/or colocalization with pioneer factor, FOXA1 (70-73). While nuclear ERα signaling appeared necessary and sufficient for the induction of osteolytic PTHrP secretion from bone tropic ER+ cells, a possible secondary role for ERα MISS could not be excluded (eg, a lipid-raft disrupting agent partially blocked E2 stimulated PTHrP alone or in combination with TGFß, with both membrane ER (34) and TGFß receptors (52) reported to be located in caveolin-enriched lipids rafts).

The clear sufficiency of nuclear ERα-only (ie, E4) signaling in driving osteolytic PTHrP secretion from BMET-forming MCF-7 ER+ cells, and, perhaps even more importantly, previous evidence that E4 supports MCF-7 orthotopic tumor growth in vivo (41, 42), led to in vivo BMET experiments using E4 to test the postulate that genomic ER signaling was also sufficient to promote ER+ breast cancer progression in bone. Our contrary finding of osteolytic BMET supported by isolated E2 but not by isolated E4 treatment when dosed well within the range supporting orthotopic tumor growth, was therefore quite intriguing, as were trends, albeit without statistical significance, for lower BMET progression when E2 was combined with E4, consistent with E4 antagonism of tumor-promoting pharmacologic effects of E2-stimulated ER MISS. In toto, these in vivo results suggested, contrary to expectation, that pharmacologic stimulation of genomic ER was insufficient to support ER+ BMET progression in the model and that nongenomic ER signaling, in combination with genomic, may be required.

One limitation of these experiments was the use of single doses of E4 and E2. However, documentation of pharmacologic effects of the E4 dose on estrogen-responsive tissues (bone, uterus, mammary gland) in tumor-bearing mice, while in some cases (eg, uterus) likely attributable to antagonism of endogenous E2 MISS effects in these ovary-intact mice (40, 53), suggested that the inability of E4 to support ER+ BMET progression was not due to inadequate dosing, but rather was attributable to a lack of ER MISS stimulation by E4 (in comparison to robust effects of E2). Future experiments using a higher E4:E2 dosing ratio to probe the ability of E4 to reduce E2-mediated BMET progression via antagonism of pharmacologic E2 stimulation of ER MISS would lend credence to the conclusion that both genomic and nongenomic ER signaling are required for ER+ BMET progression. Manipulation of estrogen receptor expression on both sides of the tumor/bone interface in future experiments, in addition to the pharmacologic approach used here, will also help to clarify genomic vs nongenomic ER signaling effects in ER+ BMET. Nonetheless, the totality of current in vivo BMET findings presented here are consistent with a necessary stimulatory role for ER MISS, in combination with nuclear ER signaling, in driving in vivo ER+ tumor progression specific to bone, a site where metastases occur with twice the prevalence in ER+ (vs ER-negative) MBC and almost 100% prevalence in women with acquired activating ER mutations (4, 18).

Still to be determined are the relative contributions of bone vs tumor ER MISS in driving ER+ metastases at this metastatic site. Clearly, the reduced bone anabolic effect of E4 (vs E2) documented here (eg, unchanged osteoblast numbers in E4-treated mice vs 2-fold increases with E2-treatment) could be of mechanistic relevance for BMET progression, while also explaining the much smaller increase in BMD in E4 vs E2 treated mice. These findings, likely attributable to absent ER MISS signaling with E4, are consistent with prior evidence that ERα MISS mediates estrogen's anti-apoptotic effect in osteoblasts, including: (i) significantly lower osteoblast numbers in E2-treated OVX mice lacking ERα MISS receptors (vs ERα wild-type mice) (44); and (ii) dose-dependent decreases in osteoblast-derived osteocalcin levels in postmenopausal women treated with E4 (vs unchanged levels with E2) (45). While increased osteolytic BMET size in E2-treated mice inoculated with ER-negative cells (24) suggests estrogen-driven bone anabolism can indeed impact BMET progression, reduced bone anabolism with E4 appeared unlikely to be the sole cause of absent BMET progression since MCF-7 cells with activating ESR1 mutations formed BMET in mice in the absence of E2 supplementation, with an incidence analogous to “parental” ESR1 wild-type cells supplementation with E2. Moreover, the activating ESR1 mutation findings point to ERα signaling, in particular, as being sufficient for driving ER+ BMET progression.

It should be noted that there are several limitations to the experiments described here, including their primary reliance on multiple variants of a single human ER+ cell line (MCF-7), due in part to low in vivo ER+ PDX take rates and rare BMET formation (74-76). At the same time, comparison of isogenic lines (ie, ESR1 wild-type vs mutant) provides benefits, as does use of MCF-7 cells, a human ER+ breast cancer cell that is not only highly studied (including effects of genomic vs nongenomic ERα signaling, unrelated to BMET) (34, 40-42, 59) but has also been described as highly representative of clinical ER+ breast cancer tumors (40, 77). Secondly, estrogen-stimulated PTHrP secretion, while well-reasoned as a screening bioassay, clearly did not recapitulate all factors of relevance to in vivo estrogen-dependent osteolytic progression. At the same time, this dichotomy of findings may also provide mechanistic insights, as for example, osteoblasts, which were only increased in number in E2-treated mice, are an important mediator of osteolytic PTHrP effects in vivo (78).

In summary, data presented here suggest a pathogenic role for ERα MISS in promoting ER+ breast cancer progression that could be bone-specific, and may therefore be of therapeutic importance, meriting further exploration on both sides of the tumor/bone interface. For example, truncated ERα isoforms may contribute to ERα MISS effects in bone-disseminated breast cancer cells in unique ways, and involvement of additional microenvironmental targets not examined here may also be of importance (eg, ER MISS effects in bone's unique vasculature) (40). While much remains to be explored, these results do suggest that better harnessing of the complex interplay of genomic and nongenomic ERα signaling in both tumor cells and their bone microenvironment could improve targeting of bone-disseminated ER+ breast cancer cells for both treatment and prevention of ER+ BMET, which remain incurable.

Acknowledgments

We would like to acknowledge University of Arizona undergraduate researchers Albiya Thomas, Geethika Ameneni, and Alyssa S. Magee for their contributions to the data analyses.

Abbreviations

ANOVA

analysis of variance

ATCC

American Type Culture Collection

BMD

bone mineral density

BMET

bone metastasis

DXA

dual-energy x-ray absorptiometry

E2

estrogen (17ß-estradiol)

E4

estetrol

ER

estrogen receptor

ER+

expressing estrogen receptor-α

GPER

G protein–coupled ER

H&E

hematoxylin and eosin

MAPK

mitogen-activated protein kinase

IC

intracardiac

MBC

metastatic breast cancer

MISS

membrane-initiated steroid signaling

PaPE-1

pathway preferential estrogen 1

PPT

propyl pyrazole triol

PTHrP

parathyroid hormone–related protein

R-DPN

R-diarylpropionitrile

TGFβ

transforming growth factor β

TRAP

tartrate-resistant acid phosphatase

Contributor Information

Julia N Cheng, Cancer Biology Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ 85724, USA.

Jennifer B Frye, Department of Medicine, University of Arizona, Tucson, AZ 86724, USA.

Susan A Whitman, Department of Medicine, University of Arizona, Tucson, AZ 86724, USA.

Sima Ehsani, Department of Medicine, University of Arizona, Tucson, AZ 86724, USA.

Simak Ali, Department of Surgery & Cancer, Imperial College London, London W12 0NN, UK.

Janet L Funk, Department of Medicine, University of Arizona, Tucson, AZ 86724, USA.

Funding

This work was supported by the National Cancer Institute (NCI) of the National Institutes of Health (NIH) (R03CA181893 and R01CA174926 to J.L.F., T32CA009213 to J.N.C., P30CA023074); METAvivor (Translational Research Award, J.L.F.); the Phoenix Chapter of ARCS Foundation (J.N.C.); the Louise Foucar Marshall Foundation Dissertation Fellowship (J.N.C.).

Disclosures

All authors have no conflicts of interest and nothing to disclose.

Data Availability

Some or all datasets generated during and/or analyzed during the current study are not publicly available but are available from the corresponding author on reasonable request.

References

  • 1. Harbeck  N, Penault-Llorca  F, Cortes  J, et al.  Breast cancer. Nat Rev Dis Primers. 2019;5(1):66. [DOI] [PubMed] [Google Scholar]
  • 2. Colzani  E, Johansson  ALV, Liljegren  A, et al.  Time-dependent risk of developing distant metastasis in breast cancer patients according to treatment, age and tumour characteristics. Br J Cancer. 2014;110(5):1378‐1384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Haque  R, Ahmed  SA, Inzhakova  G, et al.  Impact of breast cancer subtypes and treatment on survival: an analysis spanning two decades. Cancer Epidemiol Biomarkers Prev. 2012;21(10):1848‐1855. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Soni  A, Ren  Z, Hameed  O, et al.  Breast cancer subtypes predispose the site of distant metastases. Am J Clin Pathol. 2015;143(4):471‐478. [DOI] [PubMed] [Google Scholar]
  • 5. Turner  NC, Neven  P, Loibl  S, Andre  F. Advances in the treatment of advanced oestrogen-receptor-positive breast cancer. Lancet. 2017;389(10087):2403‐2414. [DOI] [PubMed] [Google Scholar]
  • 6. Braun  S, Auer  D, Marth  C. The prognostic impact of bone marrow micrometastases in women with breast cancer. Cancer Invest. 2009;27(6):598‐603. [DOI] [PubMed] [Google Scholar]
  • 7. Braun  S, Vogl  FD, Naume  B, et al.  A pooled analysis of bone marrow micrometastasis in breast cancer. N Engl J Med. 2005;353(8):793‐802. [DOI] [PubMed] [Google Scholar]
  • 8. Hussein  O, Komarova  SV. Breast cancer at bone metastatic sites: recent discoveries and treatment targets. J Cell Commun Signal. 2011;5(2):85‐99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Falck  A-K, Bendahl  P-O, Ingvar  C, et al.  Analysis of and prognostic information from disseminated tumour cells in bone marrow in primary breast cancer: a prospective observational study. BMC Cancer. 2012;12(1):403‐403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. D'Oronzo  S, Gregory  W, Nicholson  S, Chong  YK, Brown  J, Coleman  R. Natural history of stage II/III breast cancer, bone metastasis and the impact of adjuvant zoledronate on distribution of recurrences. J Bone Oncol. 2021;28:100367. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Aurilio  G, Monfardini  L, Rizzo  S, et al.  Discordant hormone receptor and human epidermal growth factor receptor 2 status in bone metastases compared to primary breast cancer. Acta Oncol. 2013;52(8):1649‐1656. [DOI] [PubMed] [Google Scholar]
  • 12. Hilton  JF, Amir  E, Hopkins  S, et al.  Acquisition of metastatic tissue from patients with bone metastases from breast cancer. Breast Cancer Res Treat. 2010;129(3):761‐765. [DOI] [PubMed] [Google Scholar]
  • 13. Kamby  C, Rasmussen  BB, Kristensen  B. Oestrogen receptor status of primary breast carcinomas and their metastases. Relation to pattern of spread and survival after recurrence. Br J Cancer. 1989;60(2):252‐257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Manavathi  B, Dey  O, Gajulapalli  VNR, Bhatia  RS, Bugide  S, Kumar  R. Derailed estrogen signaling and breast cancer: an authentic couple. Endocr Rev. 2013;34(1):1‐32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Lumachi  F, Brunello  A, Maruzzo  M, Basso  U, Basso  SMM. Treatment of estrogen receptor-positive breast cancer. Curr Med Chem. 2013;20(5):596‐604. [DOI] [PubMed] [Google Scholar]
  • 16. Kennecke  H, Yerushalmi  R, Woods  R, et al.  Metastatic behavior of breast cancer subtypes. J Clin Oncol. 2010;28(20):3271‐3277. [DOI] [PubMed] [Google Scholar]
  • 17. Pan  H, Gray  R, Braybrooke  J, et al.  20-year risks of breast-cancer recurrence after stopping endocrine therapy at 5 years. N Engl J Med. 2017;377(19):1836‐1846. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Kuang  Y, Siddiqui  B, Hu  J, et al.  Unraveling the clinicopathological features driving the emergence of ESR1 mutations in metastatic breast cancer. NPJ Breast Cancer. 2018;4:22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Pedersen  RN, Esen  B, Mellemkjær  L, et al.  The incidence of breast cancer recurrence 10-32 years after primary diagnosis. J Natl Cancer Inst. 2022;114(3):391‐399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Reinert  T, Barrios  CH. Optimal management of hormone receptor positive metastatic breast cancer in 2016. Ther Adv Med Oncol. 2015;7(6):304‐320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Fribbens  C, Garcia Murillas  I, Beaney  M, et al.  Tracking evolution of aromatase inhibitor resistance with circulating tumour DNA analysis in metastatic breast cancer. Ann Oncol. 2018;29(1):145‐153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. D’Oronzo  S, Coleman  R, Brown  J, Silvestris  F. Metastatic bone disease: pathogenesis and therapeutic options: up-date on bone metastasis management. J Bone Oncol. 2019;15:004‐004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Macedo  F, Ladeira  K, Pinho  F, et al.  Bone metastases: an overview. Oncol Rev. 2017;11(1):321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Cheng  JN, Frye  JB, Whitman  SA, Kunihiro  AG, Brickey  JA, Funk  JL. Osteolytic effects of tumoral estrogen signaling in estrogen receptor-positive breast cancer bone metastasis progression in mice. J Cancer Metastasis Treat.  2021;7(17). 10.20517/2394-4722.2021.27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Cheng  JN, Frye  JB, Whitman  SA, Funk  JL. Skeletal impact of 17β-estradiol in T cell-deficient mice: age-dependent bone effects and osteosarcoma formation. Clin Exp Metastasis. 2020;37(2):269‐281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Powell  GJ, Southby  J, Danks  JA, et al.  Localization of parathyroid hormone-related protein in breast cancer metastases: increased incidence in bone compared with other sites. Cancer Res. 1991;51(11):3059‐3061. [PubMed] [Google Scholar]
  • 27. Southby  J, Kissin  MW, Danks  JA, et al.  Immunohistochemical localization of parathyroid hormone-related protein in human breast cancer. Cancer Res. 1990;50(23):7710‐7716. [PubMed] [Google Scholar]
  • 28. Cheng  JN, Frye  JB, Whitman  SA, Kunihiro  AG, Pandey  R, Funk  JL. A role for TGFβ signaling in preclinical osteolytic estrogen receptor-positive breast cancer bone metastases progression. Int J Mol Sci. 2021;22(9):4463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Kakonen  S-M, Selander  KS, Chirgwin  JM, et al.  Transforming growth factor-beta stimulates parathyroid hormone-related protein and osteolytic metastases via Smad and mitogen-activated protein kinase signaling pathways. J Biol Chem. 2002;277(27):24571‐24578. [DOI] [PubMed] [Google Scholar]
  • 30. Lindemann  RK, Ballschmieter  P, Nordheim  A, Dittmer  J. Transforming growth factor beta regulates parathyroid hormone-related protein expression in MDA-MB-231 breast cancer cells through a novel Smad/Ets synergism. J Biol Chem. 2001;276(49):46661‐46670. [DOI] [PubMed] [Google Scholar]
  • 31. Chiechi  A, Waning  DL, Stayrook  KR, Buijs  JT, Guise  TA, Mohammad  KS. Role of TGF- β in breast cancer bone metastases. Adv Biosci Biotechnol. 2014;4(10C):15‐30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Leygue  E, Murphy  LC. A bi-faceted role of estrogen receptor β in breast cancer. Endocr Relat Cancer. 2013;20(3):R127‐R139. [DOI] [PubMed] [Google Scholar]
  • 33. Ariazi  EA, Brailoiu  E, Yerrum  S, et al.  The G protein-coupled receptor GPR30 inhibits proliferation of estrogen receptor-positive breast cancer cells. Cancer Res. 2010;70(3):1184‐1194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Arnal  JF, Lenfant  F, Metivier  R, et al.  Membrane and nuclear estrogen receptor alpha actions: from tissue specificity to medical implications. Am Physiol Soc. 2017;97:1045‐1087. [DOI] [PubMed] [Google Scholar]
  • 35. Flouriot  G, Brand  H, Denger  S, et al.  Identification of a new isoform of the human estrogen receptor-alpha (hER-alpha) that is encoded by distinct transcripts and that is able to repress hER-alpha activation function 1. EMBO J. 2000;19(17):4688‐4700. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Kleuser  B, Malek  D, Gust  R, Pertz  HH, Potteck  H. 17-Beta-estradiol inhibits transforming growth factor-beta signaling and function in breast cancer cells via activation of extracellular signal-regulated kinase through the G protein-coupled receptor 30. Mol Pharmacol. 2008;74(6):1533‐1543. [DOI] [PubMed] [Google Scholar]
  • 37. Fuentes  N, Silveyra  P. Estrogen Receptor Signaling Mechanisms. Vol 116. Academic Press Inc.; 2019. p. 135‐170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Mauvais-Jarvis  F, Lange  CA, Levin  ER. Membrane-initiated estrogen, androgen, and progesterone receptor signaling in health and disease. Endocr Rev. 2022;43(4):720‐742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Driggers  PH, Segars  JH. Estrogen action and cytoplasmic signaling pathways. Part II: the role of growth factors and phosphorylation in estrogen signaling. Trends Endocrinol Metab. 2002;13(10):422‐427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Abot  A, Fontaine  C, Buscato  M, et al.  The uterine and vascular actions of estetrol delineate a distinctive profile of estrogen receptor modulation, uncoupling nuclear and membrane activation. EMBO Mol Med. 2014;6(10):1328‐1346. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Gérard  C, Mestdagt  M, Tskitishvili  E, et al.  Combined estrogenic and anti-estrogenic properties of estetrol on breast cancer may provide a safe therapeutic window for the treatment of menopausal symptoms. Oncotarget. 2015;6(19):17621‐17636. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Gallez  A, Blacher  S, Maquoi  E, et al.  Estetrol combined to progestogen for menopause or contraception indication is neutral on breast cancer. Cancers (Basel). 2021;13(10):2486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Gustafsson  KL, Farman  H, Henning  P, et al.  The role of membrane ERα signaling in bone and other major estrogen responsive tissues. Sci Rep. 2016;6(1):29473‐29473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Vinel  A, Hay  E, Valera  M, et al.  Role of ERαMISS in the effect of estradiol on cancellous and cortical femoral bone in growing female mice. Endocrinology. 2016;157(6):2533‐2544. [DOI] [PubMed] [Google Scholar]
  • 45. Visser  M, Coelingh Bennink  HJT. Clinical applications for estetrol. J Steroid Biochem Mol Biol. 2009;114(1-2):85‐89. [DOI] [PubMed] [Google Scholar]
  • 46. Guise  TA, Yin  JJ, Taylor  SD, et al.  Evidence for a causal role of parathyroid hormone-related protein in the pathogenesis of human breast cancer-mediated osteolysis. J Clin Invest. 1996;98(7):1544‐1549. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Wright  LE, Frye  JB, Lukefahr  AL, et al.  Curcuminoids block TGF-β signaling in human breast cancer cells and limit osteolysis in a murine model of breast cancer bone metastasis. J Nat Prod. 2013;76(3):316‐321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Waning  DL, Mohammad  KS, Reiken  S, et al.  Excess TGF-β mediates muscle weakness associated with bone metastases in mice. Nat Med. 2015;21(11):1262‐1271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Harrod  A, Lai  C-F, Goldsbrough  I, et al.  Genome engineering for estrogen receptor mutations reveals differential responses to anti-estrogens and new prognostic gene signatures for breast cancer. Oncogene. 2022;41:4905‐4915. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Kunihiro  AG, Brickey  JA, Frye  JB, Luis  PB, Schneider  C, Funk  JL. Curcumin, but not curcumin-glucuronide, inhibits Smad signaling in TGFβ-dependent bone metastatic breast cancer cells and is enriched in bone compared to other tissues. J Nutr Biochem. 2019;63:150‐156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Nelson  AW, Groen  AJ, Miller  JL, et al.  Comprehensive assessment of estrogen receptor beta antibodies in cancer cell line models and tissue reveals critical limitations in reagent specificity. Mol Cell Endocrinol. 2017;440:138‐150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Kunihiro  AG, Brickey  JA, Frye  JB, et al.  Curcumin inhibition of TGFβ signaling in bone metastatic cancer cells and the possible role of oxidative metabolites. J Nutr Biochem. 2022;99:108842. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Gérard  C, Blacher  S, Communal  L, et al.  Estetrol is a weak estrogen antagonizing estradiol-dependent mammary gland proliferation. J Endocrinol. 2015;224(1):85‐95. [DOI] [PubMed] [Google Scholar]
  • 54. Wright  LE, Frye  JB, Lukefahr  AL, et al.  4-Vinylcyclohexene diepoxide (VCD) inhibits mammary epithelial differentiation and induces fibroadenoma formation in female Sprague Dawley rats. Reprod Toxicol. 2011;32(1):26‐32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Dempster  DW, Compston  JE, Drezner  MK, et al.  Standardized nomenclature, symbols, and units for bone histomorphometry: a 2012 update of the report of the ASBMR Histomorphometry Nomenclature Committee. J Bone Miner Res. 2013;28(1):2‐17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Streicher  C, Heyny  A, Andrukhova  O, et al.  Estrogen regulates bone turnover by targeting RANKL expression in bone lining cells. Sci Rep. 2017;7(1):6460‐6460. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Sowder  ME, Johnson  RW. Enrichment and detection of bone disseminated tumor cells in models of low tumor burden. Sci Rep. 2018;8(1):14299‐14299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Cheng  JN, Frye  JB, Whitman  SA, Ehsani  S, Ali  S, Funk  JL. Supplementary materials for: interrogating estrogen signaling pathways in human ER-positive breast cancer cells forming bone metastases in mice. University of Arizona Research Data Repository. 2024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Madak-Erdogan  Z, Kim  SH, Gong  P, et al.  Design of pathway preferential estrogens that provide beneficial metabolic and vascular effects without stimulating reproductive tissues. Sci Signal. 2016;9(429):ra53‐ra53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Alayev  A, Salamon  RS, Berger  SM, et al.  MTORC1 directly phosphorylates and activates ERα upon estrogen stimulation. Oncogene. 2016;35(27):3535‐3543. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Zhang  YE. Non-Smad pathways in TGF-beta signaling. Cell Res. 2009;19(1):128‐139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Levin  ER. Membrane oestrogen receptor alpha signalling to cell functions. J Physiol. 2009;587(Pt 21):5019‐5023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Razandi  M, Oh  P, Pedram  A, Schnitzer  J, Levin  ER. ERs associate with and regulate the production of caveolin: implications for signaling and cellular actions. Mol Endocrinol. 2002;16(1):100‐115. [DOI] [PubMed] [Google Scholar]
  • 64. Lee  H, Bai  W. Regulation of estrogen receptor nuclear export by ligand-induced and p38-mediated receptor phosphorylation. Mol Cell Biol. 2002;22(16):5835‐5845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Antoon  JW, Bratton  MR, Guillot  LM, Wadsworth  S, Salvo  VA, Burow  ME. Inhibition of p38-MAPK alters SRC coactivation and estrogen receptor phosphorylation. Cancer Biol Ther. 2012;13(11):1026‐1033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Zuo  W, Chen  YG. Specific activation of mitogen-activated protein kinase by transforming growth factor-beta receptors in lipid rafts is required for epithelial cell plasticity. Mol Biol Cell. 2009;20(3):1020‐1029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Shoker  BS, Jarvis  C, Clarke  RB, et al.  Estrogen receptor-positive proliferating cells in the normal and precancerous breast. Am J Pathol. 1999;155(6):1811‐1815. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Burstein  HJ, Lacchetti  C, Anderson  H, et al.  Adjuvant endocrine therapy for women with hormone receptor-positive breast cancer: ASCO clinical practice guideline focused update. J Clin Oncol. 2019;37(5):423‐438. [DOI] [PubMed] [Google Scholar]
  • 69. Matsuda  T, Yamamoto  T, Muraguchi  A, Saatcioglu  F. Cross-talk between transforming growth factor-beta and estrogen receptor signaling through Smad3. J Biol Chem. 2001;276(46):42908‐42914. [DOI] [PubMed] [Google Scholar]
  • 70. Baxter  E, Windloch  K, Kelly  G, Lee  JS, Gannon  F, Brennan  DJ. Molecular basis of distinct oestrogen responses in endometrial and breast cancer. Endocr Relat Cancer. 2019;26(1):31‐46. [DOI] [PubMed] [Google Scholar]
  • 71. Bochkis  IM, Schug  J, Ye  DZ, et al.  Genome-wide location analysis reveals distinct transcriptional circuitry by paralogous regulators Foxa1 and Foxa2. PLoS Genet. 2012;8(6):e1002770. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Carroll  JS, Liu  XS, Brodsky  AS, et al.  Chromosome-wide mapping of estrogen receptor binding reveals long-range regulation requiring the forkhead protein FoxA1. Cell. 2005;122(1):33‐43. [DOI] [PubMed] [Google Scholar]
  • 73. Taube  JH, Allton  K, Duncan  SA, Shen  L, Barton  MC. Foxa1 functions as a pioneer transcription factor at transposable elements to activate Afp during differentiation of embryonic stem cells. J Biol Chem. 2010;285(21):16135‐16144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Holen  I, Speirs  V, Morrissey  B, Blyth  K. In vivo models in breast cancer research: progress, challenges and future directions. Dis Model Mech. 2017;10(4):359‐371. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Murayama  T, Gotoh  N. Patient-Derived Xenograft models of breast cancer and their application. Cells. 2019;8(6):621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Wright  LE, Ottewell  PD, Rucci  N, et al.  Murine models of breast cancer bone metastasis. Bonekey Rep. 2016;5:804‐804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Jiang  G, Zhang  S, Yazdanparast  A, et al.  Comprehensive comparison of molecular portraits between cell lines and tumors in breast cancer. BMC Genomics. 2016;17(Suppl 7):525. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Thomas  RJ, Guise  TA, Yin  JJ, et al.  Breast cancer cells interact with osteoblasts to support osteoclast formation. Endocrinology. 1999;140(10):4451‐4458. [DOI] [PubMed] [Google Scholar]
  • 79. Jiang  Y, Gong  P, Madak-Erdogan  Z, et al.  Mechanisms enforcing the estrogen receptor β selectivity of botanical estrogens. FASEB J. 2013;27(11):4406‐4418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Kraichely  DM, Sun  J, Katzenellenbogen  JA, Katzenellenbogen  BS. Conformational changes and coactivator recruitment by novel ligands for estrogen receptor-alpha and estrogen receptor-beta: correlations with biological character and distinct differences among SRC coactivator family members. Endocrinology. 2000;141(10):3534‐3545. [DOI] [PubMed] [Google Scholar]
  • 81. Stauffer  SR, Coletta  CJ, Tedesco  R, et al.  Pyrazole ligands: structure—affinity/activity relationships and estrogen receptor-α-selective agonists. J Med Chem. 2000;43(26):4934‐4947. [DOI] [PubMed] [Google Scholar]
  • 82. Carroll  VM, Jeyakumar  M, Carlson  KE, Katzenellenbogen  JA. Diarylpropionitrile (DPN) enantiomers: synthesis and evaluation of estrogen receptor β-selective ligands. J Med Chem. 2012;55(1):528‐537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Cao  L-Y, Ren  X-M, Yang  Y, et al.  Hydroxylated polybrominated biphenyl ethers exert estrogenic effects via non-genomic G protein–coupled estrogen receptor mediated pathways. Environ Health Perspect. 2018;126(5):057005‐057005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Bologa  CG, Revankar  CM, Young  SM, et al.  Virtual and biomolecular screening converge on a selective agonist for GPR30. Nat Chem Biol. 2006;2(4):207‐212. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

Some or all datasets generated during and/or analyzed during the current study are not publicly available but are available from the corresponding author on reasonable request.


Articles from Endocrinology are provided here courtesy of The Endocrine Society

RESOURCES