American Association for Cancer Research
Browse
15357163mct200563-sup-246667_2_supp_6665938_qgkjkv.xlsx (1.15 MB)

Supplementary Data from Rapid Induction of the Unfolded Protein Response and Apoptosis by Estrogen Mimic TTC-352 for the Treatment of Endocrine-Resistant Breast Cancer

Download (1.15 MB)
dataset
posted on 2023-04-03, 18:23 authored by Balkees Abderrahman, Philipp Y. Maximov, Ramona F. Curpan, Sean W. Fanning, Jay S. Hanspal, Ping Fan, Charles E. Foulds, Yue Chen, Anna Malovannaya, Antrix Jain, Rui Xiong, Geoffrey L. Greene, Debra A. Tonetti, Gregory R.J. Thatcher, V. Craig Jordan

Table S5

Funding

National Institutes of Health MD Anderson Cancer Center Support

George and Barbara Bush Foundation for Innovative Cancer Research

Cancer Prevention Research Institute of Texas

Baylor College of Medicine Mass Spectrometry Proteomics Core

CPRIT Proteomics and Metabolomics Core Facility Award

Office of Biological and Environmental Research

History

ARTICLE ABSTRACT

Patients with long-term estrogen-deprived breast cancer, after resistance to tamoxifen or aromatase inhibitors develops, can experience tumor regression when treated with estrogens. Estrogen's antitumor effect is attributed to apoptosis via the estrogen receptor (ER). Estrogen treatment can have unpleasant gynecologic and nongynecologic adverse events; thus, the development of safer estrogenic agents remains a clinical priority. Here, we study synthetic selective estrogen mimics (SEM) BMI-135 and TTC-352, and the naturally occurring estrogen estetrol (E4), which are proposed as safer estrogenic agents compared with 17β-estradiol (E2), for the treatment of endocrine-resistant breast cancer. TTC-352 and E4 are being evaluated in breast cancer clinical trials. Cell viability assays, real-time PCR, immunoblotting, ERE DNA pulldowns, mass spectrometry, X-ray crystallography, docking and molecular dynamic simulations, live cell imaging, and Annexin V staining were conducted in 11 biologically different breast cancer models. Results were compared with the potent full agonist E2, less potent full agonist E4, the benchmark partial agonist triphenylethylene bisphenol (BPTPE), and antagonists 4-hydroxytamoxifen and endoxifen. We report ERα's regulation and coregulators’ binding profiles with SEMs and E4. We describe TTC-352′s pharmacology as a weak full agonist and antitumor molecular mechanisms. This study highlights TTC-352′s benzothiophene scaffold that yields an H-bond with Glu353, which allows Asp351-to-helix 12 (H12) interaction, sealing ERα's ligand-binding domain, recruiting E2-enriched coactivators, and triggering rapid ERα-induced unfolded protein response (UPR) and apoptosis, as the basis of its anticancer properties. BPTPE's phenolic OH yields an H-Bond with Thr347, which disrupts Asp351-to-H12 interaction, delaying UPR and apoptosis and increasing clonal evolution risk.