ESR1 ligand-binding domain mutations in hormone-resistant breast cancer

Weiyi Toy, Yang Shen, Helen Won, Bradley Green, Rita A Sakr, Marie Will, Zhiqiang Li, Kinisha Gala, Sean Fanning, Tari A King, Clifford Hudis, David Chen, Tetiana Taran, Gabriel Hortobagyi, Geoffrey Greene, Michael Berger, José Baselga, Sarat Chandarlapaty, Weiyi Toy, Yang Shen, Helen Won, Bradley Green, Rita A Sakr, Marie Will, Zhiqiang Li, Kinisha Gala, Sean Fanning, Tari A King, Clifford Hudis, David Chen, Tetiana Taran, Gabriel Hortobagyi, Geoffrey Greene, Michael Berger, José Baselga, Sarat Chandarlapaty

Abstract

Seventy percent of breast cancers express estrogen receptor (ER), and most of these are sensitive to ER inhibition. However, many such tumors for unknown reasons become refractory to inhibition of estrogen action in the metastatic setting. We conducted a comprehensive genetic analysis of two independent cohorts of metastatic ER-positive breast tumors and identified mutations in ESR1 affecting the ligand-binding domain (LBD) in 14 of 80 cases. These included highly recurrent mutations encoding p.Tyr537Ser, p.Tyr537Asn and p.Asp538Gly alterations. Molecular dynamics simulations suggest that the structures of the Tyr537Ser and Asp538Gly mutants involve hydrogen bonding of the mutant amino acids with Asp351, thus favoring the agonist conformation of the receptor. Consistent with this model, mutant receptors drive ER-dependent transcription and proliferation in the absence of hormone and reduce the efficacy of ER antagonists. These data implicate LBD-mutant forms of ER in mediating clinical resistance to hormonal therapy and suggest that more potent ER antagonists may be of substantial therapeutic benefit.

Conflict of interest statement

Competing Financial interests

The authors declare no competing financial interests.

Figures

Figure 1. ESR1 mutations in ER+ metastatic…
Figure 1. ESR1 mutations in ER+ metastatic breast cancers
a. Percentage of matched cases with gene mutations detected in our patient samples (MSKCC) were compared to those of The Cancer Genome Atlas (TCGA) invasive breast cancer and luminal A/B cases. Only genes found mutated in 3 or more cases are shown. b. Top amplified genes detected in the patient samples. Genes were considered amplified when the ratio of their copy number in tumor to normal is greater than 2 c. Diagram of ER domains with the location of the identified mutations. AF-1: Activation Function-1; DBD: DNA binding domain; NLS: Nuclear localizing signal; LBD: Ligand binding domain; AF-2: Activation function-2.
Figure 2. ERα LBD mutants demonstrate elevated…
Figure 2. ERα LBD mutants demonstrate elevated activity in absence of hormone
a. SKBR3 and MCF7 cells were transfected with empty vector, HA-ERα wild type (WT) or mutants, ERE-luciferase and Renilla luciferase reporter constructs in hormone-depleted medium with 10 nM of E2 added for 24 hours where indicated. Relative light units (RLU) were calculated as the ratio of Firefly luciferase over Renilla luciferase activity. Graph was plotted with the mean ± SD of triplicate biological replicates. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001. b. MCF7 cells were transfected with HA-ERα wild type (WT) or mutants in hormone-depleted medium and harvested 48 hours post-transfection. The mRNA level of ERα target genes were quantified by qRT-PCR using gene-specific primers. Bars represent mean ± SD of triplicate technical replicates normalized to ESR1 expression. *, P < 0.05; ***, P < 0.001; ****, P < 0.0001. The data shown is a representative of two biological replicates. c. Gene expression microarray was performed using RNA derived from MCF7 cells transfected with empty vector, GFP-ERα WT or mutants in hormone-depleted medium for 48 hours. A total of 92 genes were commonly up or down-regulated by the ERα mutants and they were hierarchically clustered to generate a heat map. Selected genes previously well established as estrogen responsive genes were marked with *. d. Lysates of MCF7 cells transfected with empty vector, HA-ERα WT or mutants in hormone-depleted medium for 48 hours, were immunoblotted with phospho-Serine 118 of ERα, HA and actin antibodies. The immunoblots shown are a representative of experiments repeated at least three times. e. Lysates generated from MCF7 transfected with empty vector GFP-ERα WT, or D538G were immunoprecipitated with GFP-Trap beads and immunoblotted with AIB1 and GFP antibodies. The immunoblots shown are a representative of experiments repeated three times. f. MCF7 cells transfected with HA-ERα WT or mutants were treated with SNX2112 at various doses for 3 hours before harvest. Lysates were then immunoblotted with HA and actin antibodies. The immunoblots shown are a representative of experiments repeated three times. g. Mice bearing MCF7 inducible vector control (VC), HA-ER WT or Tyr537Ser or Asp538Gly tumors had their 17β-estradiol pellets removed when the tumors reached an approximate volume of 250 mm3 and then fed with water containing 0.2–0.5 mg/ml of doxycycline a week after for the induction of ER WT and mutant expression. The result was presented as percentage tumor growth by normalizing to the first mean tumor volume measured for each group ± SEM (n = 10 mice/group).
Figure 3. ER antagonists partially inhibit ER…
Figure 3. ER antagonists partially inhibit ER mutant transcriptional activity
SKBR3 cells were transfected with HA-ERα WT or mutants, ERE-luciferase and Renilla luciferase reporter constructs in hormone-depleted medium. The cells were then treated with either a. 1 nM E2 alone or in combination with 4-OHT (4-hydroxytamoxifen) and b. fulvestrant at the indicated doses for 24 hours before luciferase activity measurement. Relative light units (RLU) were calculated as the ratio of Firefly luciferase over Renilla luciferase activity. Graph was plotted with the mean ± SD of triplicate biological replicates. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001.
Figure 4. Molecular dynamics-modeled structures of WT…
Figure 4. Molecular dynamics-modeled structures of WT and mutant forms of ERα
a. WT (cyan), b. Tyr537Ser (green), and c. Asp538Gly (red) at 10 ns, overlaid with a co-crystallized structure (PDB accession code 1GWQ) of ERα WT in an agonist conformation (gray) and TIF2 (wheat). d. Hydrogen-to-acceptor distances between Asp538/Ser537/Gly538 and Asp351 in ERα WT/Tyr537Ser/Asp538Gly respectively, over the course of 10-ns molecular dynamics simulations.

References

    1. Effects of chemotherapy and hormonal therapy for early breast cancer on recurrence and 15-year survival: an overview of the randomised trials. Lancet. 2005;365:1687–717.
    1. Ariazi EA, Ariazi JL, Cordera F, Jordan VC. Estrogen receptors as therapeutic targets in breast cancer. Curr Top Med Chem. 2006;6:181–202.
    1. Strasser-Weippl K, Goss PE. Advances in adjuvant hormonal therapy for postmenopausal women. J Clin Oncol. 2005;23:1751–9.
    1. Forbes JF, et al. Effect of anastrozole and tamoxifen as adjuvant treatment for early-stage breast cancer: 100-month analysis of the ATAC trial. Lancet Oncol. 2008;9:45–53.
    1. Peng J, Sengupta S, Jordan VC. Potential of selective estrogen receptor modulators as treatments and preventives of breast cancer. Anticancer Agents Med Chem. 2009;9:481–99.
    1. Wagle N, et al. High-throughput detection of actionable genomic alterations in clinical tumor samples by targeted, massively parallel sequencing. Cancer Discov. 2012;2:82–93.
    1. Comprehensive molecular portraits of human breast tumours. Nature. 2012;490:61–70.
    1. Gutierrez MC, et al. Molecular changes in tamoxifen-resistant breast cancer: relationship between estrogen receptor, HER-2, and p38 mitogen-activated protein kinase. J Clin Oncol. 2005;23:2469–76.
    1. Lipton A, et al. Serum HER-2/neu conversion to positive at the time of disease progression in patients with breast carcinoma on hormone therapy. Cancer. 2005;104:257–63.
    1. Meng S, et al. HER-2 gene amplification can be acquired as breast cancer progresses. Proc Natl Acad Sci U S A. 2004;101:9393–8.
    1. Jirstrom K, et al. Adverse effect of adjuvant tamoxifen in premenopausal breast cancer with cyclin D1 gene amplification. Cancer Res. 2005;65:8009–16.
    1. Turner N, et al. FGFR1 amplification drives endocrine therapy resistance and is a therapeutic target in breast cancer. Cancer Res. 2010;70:2085–94.
    1. Baselga J, et al. Everolimus in postmenopausal hormone-receptor-positive advanced breast cancer. N Engl J Med. 2012;366:520–9.
    1. Nettles KW, et al. NFkappaB selectivity of estrogen receptor ligands revealed by comparative crystallographic analyses. Nat Chem Biol. 2008;4:241–7.
    1. 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 Res. 1997;57:1244–9.
    1. Kato S, et al. Activation of the estrogen receptor through phosphorylation by mitogen-activated protein kinase. Science. 1995;270:1491–4.
    1. Le Goff P, Montano MM, Schodin DJ, Katzenellenbogen BS. Phosphorylation of the human estrogen receptor. Identification of hormone-regulated sites and examination of their influence on transcriptional activity. J Biol Chem. 1994;269:4458–66.
    1. Anzick SL, et al. AIB1, a steroid receptor coactivator amplified in breast and ovarian cancer. Science. 1997;277:965–8.
    1. Azorsa DO, Cunliffe HE, Meltzer PS. Association of steroid receptor coactivator AIB1 with estrogen receptor-alpha in breast cancer cells. Breast Cancer Res Treat. 2001;70:89–101.
    1. Fan M, Park A, Nephew KP. CHIP (carboxyl terminus of Hsc70-interacting protein) promotes basal and geldanamycin-induced degradation of estrogen receptor-alpha. Mol Endocrinol. 2005;19:2901–14.
    1. Chandarlapaty S, et al. SNX2112, a synthetic heat shock protein 90 inhibitor, has potent antitumor activity against HER kinase-dependent cancers. Clin Cancer Res. 2008;14:240–8.
    1. Shang Y, Brown M. Molecular determinants for the tissue specificity of SERMs. Science. 2002;295:2465–8.
    1. 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. Mol Endocrinol. 1996;10:1388–98.
    1. Brzozowski AM, et al. Molecular basis of agonism and antagonism in the oestrogen receptor. Nature. 1997;389:753–8.
    1. Won H, SS Detecting Somatic Genetic Alterations in Tumor Specimens by Exon Capture and Massively Parallel Sequencing. Journal of Visualized Experiments. 2013
    1. Li H, et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics. 2009;25:2078–9.
    1. Li H, Durbin R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics. 2009;25:1754–60.
    1. DePristo MA, et al. A framework for variation discovery and genotyping using next-generation DNA sequencing data. Nat Genet. 2011;43:491–8.
    1. Robinson JT, et al. Integrative genomics viewer. Nat Biotechnol. 2011;29:24–6.
    1. Lipson D, et al. Identification of new ALK and RET gene fusions from colorectal and lung cancer biopsies. Nat Med. 2012;18:382–4.
    1. Chandarlapaty S, et al. AKT inhibition relieves feedback suppression of receptor tyrosine kinase expression and activity. Cancer Cell. 2011;19:58–71.
    1. MacKerell AD, Bashford JD, Dunbrack M, Evanseck RL, Jr, Field JD, Fischer MJ, Gao S, Guo J, Ha H, Joseph-McCarthy S, Kuchnir DL, Kuczera K, Lau FTK, Mattos C, Michnick S, Ngo T, Nguyen DT, Prodhom B, Reiher WE, III, Roux B, Schlenkrich M, Smith JC, Stote R, Straub J, Watanabe M, Wiorkiewicz-Kuczera J, Yin D, Karplus M. All-Atom Empirical Potential for Molecular Modeling and Dynamics Studies of Proteins. J Phys Chem B. 1998;102:3586–3616.
    1. Brooks BR, et al. CHARMM: the biomolecular simulation program. J Comput Chem. 2009;30:1545–614.
    1. Phillips JC, et al. Scalable molecular dynamics with NAMD. J Comput Chem. 2005;26:1781–802.

Source: PubMed

3
Subskrybuj