The oral selective estrogen receptor degrader GDC-0810 (ARN-810) in postmenopausal women with hormone receptor-positive HER2-negative (HR + /HER2 -) advanced/metastatic breast cancer

Aditya Bardia, Ingrid Mayer, Eric Winer, Hannah M Linden, Cynthia X Ma, Barbara A Parker, Meritxell Bellet, Carlos L Arteaga, Sravanthi Cheeti, Mary Gates, Ching-Wei Chang, Jill Fredrickson, Jill M Spoerke, Heather M Moore, Jennifer Giltnane, Lori S Friedman, Edna Chow Maneval, Iris Chan, Komal Jhaveri, Aditya Bardia, Ingrid Mayer, Eric Winer, Hannah M Linden, Cynthia X Ma, Barbara A Parker, Meritxell Bellet, Carlos L Arteaga, Sravanthi Cheeti, Mary Gates, Ching-Wei Chang, Jill Fredrickson, Jill M Spoerke, Heather M Moore, Jennifer Giltnane, Lori S Friedman, Edna Chow Maneval, Iris Chan, Komal Jhaveri

Abstract

Purpose: GDC-0810 (ARN-810) is a novel, non-steroidal, orally bioavailable, selective estrogen receptor degrader (SERD) that potentially inhibits ligand-dependent and ligand-independent estrogen receptor (ER)-mediated signaling.

Methods: A phase Ia/Ib/IIa dose escalation, combination treatment with palbociclib or a luteinizing hormone-releasing hormone, and expansion study determined the safety, pharmacokinetics, and recommended phase 2 dose (RP2D) of GDC-0810 in postmenopausal women with ER + (HER2 -) locally advanced or metastatic breast cancer (MBC). Baseline plasma ctDNA samples were analyzed to determine the ESR1 mutation status.

Results: Patients (N = 152) received GDC-0810 100-800 mg once daily (QD) or 300-400 mg twice daily, in dose escalation, expansion, as single agent or combination treatment. Common adverse events regardless of attribution to study drug were diarrhea, nausea, fatigue, vomiting, and constipation. There was one dose-limiting toxicity during dose escalation. The maximum tolerated dose was not reached. GDC-0810 600 mg QD taken with food was the RP2D. Pharmacokinetics were predictable. FES reduction (> 90%) highlighting pharmacodynamic engagement of ER was observed. Outcomes for the overall population and for patients with tumors harboring ESR1 mutations included partial responses (4% overall; 4% ESR1), stable disease (39% overall; 42% ESR1), non-complete response/non-progressive disease (13% overall; 12% ESR1), progressive disease (40% overall; 38% ESR1), and missing/unevaluable (5% overall; 5% ESR1). Clinical benefit (responses or SD, lasting ≥ 24 weeks) was observed in patients in dose escalation (n = 16, 39%) and expansion (n = 24, 22%).

Conclusion: GDC-0810 was safe and tolerable with preliminary anti-tumor activity in heavily pretreated patients with ER + advanced/MBC, with/without ESR1 mutations, highlighting the potential for oral SERDs. Clinical Trial and registration date April 4, 2013. NCT01823835 .

Keywords: GDC-0810; HR +; HER2 −; Metastatic breast cancer; Phase 1; Postmenopausal; Selective estrogen receptor degrader.

Conflict of interest statement

A.B. held consulting/advisory board positions at Pfizer, Novartis, Genentech, Merck, Radius Health, Immunomedics/Gilead, Sanofi, Daiichi Pharma/AstraZeneca, Phillips, Eli Lilly, Foundation Medicine, and received contracted research/grant to institution from Genentech, Novartis, Pfizer, Merck, Sanofi, Radius Health, Immunomedics/Gilead, Daiichi Pharma/AstraZeneca, and Eli Lilly. I.M. received consulting fees/ research support from Genentech and is an employee of AstraZeneca. E.W. and H.M.L. received consulting fees and research support from Genentech. C.X.M. held consulting/advisory board positions at Gilead, AstraZeneca, Sanofi-Genzyme, Jacobio, Natera, Novartis, Inivata, Biovica, Athenex, Bayer, Esai, and OncoSignal and received funding from Pfizer and Puma. B.A.P. received contracted research support to the institution from Pfizer, Novartis, Glaxo Smith Kline, Genentech/Roche, and Oncternal Therapeutics Inc., consulting fees from Dare Bioscience, Bioatla Inc. (spouse), EMD Serona (spouse), and Samumed LLC (spouse), and has stock ownership in Merck. M.B. held advisory board positions at Pfizer, Novartis, and Lilly and received speakers bureau fees from Pfizer, Novartis, and Lilly and travel expenses from Pfizer and Roche. C.L.A. held advisory board positions at Novartis, Lilly, Merck, TAIHO Oncology, Immunomedics, Daiichi Sankyo, AstraZeneca, Sanofi, OrigiMed, and Susan G. Komen Foundation, received research grant support from Pfizer, Lilly, and Takeda, and has stock ownership in Provista. S.C., M.G., C.-W.C., J.F., H.M.M., J.G., and I.C. are employees and stockholders of Roche/Genentech. J.M.S. and L.S.F. are former employees and stockholders of Roche/Genentech. E.C.M. is a former employee of Seragon. K.J. received consulting fees from AbbVie, AstraZeneca, Blueprint Medicines, Biotheranostics, BMS, Genentech, Jounce Therapeutics, Lilly Pharmaceuticals, Novartis, Pfizer, Seattle Genetics, SunPharma Pvt Ltd, and Taiho Oncology and contracted research grants to institution from ADC Therapeutics, AstraZeneca, Clovis Oncology, Debio Pharmaceuticals, Genentech, Immunomedics, Novartis, Lilly Pharmaceuticals, Merck/VelosBio, Novartis, Novita Pharmaceuticals, Pfizer, Puma Biotechnology, and Zymeworks.

© 2022. The Author(s).

Figures

Fig. 1
Fig. 1
Pharmacodynamics of GDC-0810. A Functional imaging with 18F-FES-PET for patient on GDC-0810 600-mg QD (fasting regimen) at baseline and cycle 2 day 3 with best response of stable disease. B ER protein levels and tumor cell proliferation (Ki-67) at baseline and treatment-related reductions after 3 cycles of treatment for patient on GDC-0810 600-mg QD (non-fasting regimen) with best response of stable disease. C Waterfall 18F-FES-PET response for patients in six dose cohorts in phase Ia and cohort A1 in phase IIa
Fig. 2
Fig. 2
Patient time on study. Two cohorts in phase Ia, and all cohorts in phase Ib and IIa were dosed under non-fasting conditions
Fig. 3
Fig. 3
Kaplan–Meier plot of progression-free survival for patients in phase Ia (top) and phases 1b/IIa (bottom)

References

    1. Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global cancer statistics. CA Cancer J Clin. 2011;61(2):69–90. doi: 10.3322/caac.20107[published.
    1. Najim O, Seghers S, Sergoynne L, et al. 2019 The association between type of endocrine therapy and development of estrogen receptor-1 mutation(s) in patients with hormone-sensitive advanced breast cancer: a systematic review and meta-analysis of randomized and non-randomized trials. Biochim Biophys Acta Rev Cancer. 1872;2:188315. doi: 10.1016/j.bbcan.2019.188315[published.
    1. Leal MF, Haynes BP, Schuster EF, et al. Early enrichment of ESR1 mutations and the impact on gene expression in pre-surgical primary breast cancer treated with aromatase inhibitors. Clin Cancer Res. 2019 doi: 10.1158/1078-0432.ccr-19-1129[published.
    1. Lopez-Knowles E, Pearson A, Schuster G, et al. Molecular characterisation of aromatase inhibitor-resistant advanced breast cancer: the phenotypic effect of ESR1 mutations. Br J Cancer. 2019;120(2):247–255. doi: 10.1038/s41416-018-0345-x[published.
    1. Sammons S, Shastry M, Dent S, Anders C, Hamilton E. Practical treatment strategies and future directions after progression while receiving CDK4/6 inhibition and endocrine therapy in advanced HR(+)/HER2(-) breast cancer. Clin Breast Cancer. 2020;20(1):1–11. doi: 10.1016/j.clbc.2019.06.017[published.
    1. Joseph JD, Darimont B, Zhou W, et al. The selective estrogen receptor downregulator GDC-0810 is efficacious in diverse models of ER+ breast cancer. Elife. 2016 doi: 10.7554/eLife.15828[published.
    1. Lai A, Kahraman M, Govek S, et al. Identification of GDC-0810 (ARN-810), an orally bioavailable selective estrogen receptor degrader (SERD) that demonstrates robust activity in tamoxifen-resistant breast cancer xenografts. J Med Chem. 2015;58(12):4888–4904. doi: 10.1021/acs.jmedchem.5b00054[published.
    1. Eisenhauer EA, Therasse P, Bogaerts J, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1) Eur J Cancer. 2009;45(2):228–247. doi: 10.1016/j.ejca.2008.10.026[published.
    1. Cheung KWK, Yoshida K, Cheeti S, et al. GDC-0810 pharmacokinetics and transporter-mediated drug interaction evaluation with an endogenous biomarker in the first-in-human Dose Escalation Study. Drug Metab Dispos. 2019;47(9):966–973. doi: 10.1124/dmd.119.087924.
    1. Wang Y, Ayres KL, Goldman DA, et al. (18)F-fluoroestradiol PET/CT measurement of estrogen receptor suppression during a phase I trial of the novel estrogen receptor-targeted therapeutic GDC-0810: using an imaging biomarker to guide drug dosage in subsequent trials. Clin Cancer Res. 2017;23(12):3053–3060. doi: 10.1158/1078-0432.ccr-16-2197[published.
    1. Turner NC, Kingston B, Kilburn LS, et al. Circulating tumour DNA analysis to direct therapy in advanced breast cancer (plasmaMATCH): a multicentre, multicohort, phase 2a, platform trial. Lancet Oncol. 2020;21(10):1296–1308. doi: 10.1016/s1470-2045(20)30444-7[published.
    1. News in brief, no authors listed (2022) Novel SERD Has PFS Edge against Breast Cancer. Cancer Discov 12(2):281. 10.1158/-NB2021-406
    1. Bardia A, Neven P, Streich G. Elacestrant. 2021 an oral selective estrogen receptor degrader vs investigator’s choice of endocrine monotherapy for ER+/HER2–advanced/metastatic breast cancer following progression on prior endocrine and CDK4/6 inhibitor therapy: Results of EMERALD phase 3 trial. Proceedings of the 2021 San Antonio Breast Cancer Symposium, San Antonio, TX, USA;8
    1. Pagani O, Regan MM, Walley BA, et al. Adjuvant exemestane with ovarian suppression in premenopausal breast cancer. N Engl J Med. 2014;371(2):107–118. doi: 10.1056/NEJMoa1404037[published.
    1. Turner NC, Ro J, André F, et al. Palbociclib in hormone-receptor-positive advanced breast cancer. N Engl J Med. 2015;373(3):209–219. doi: 10.1056/NEJMoa1505270[published.
    1. van Kruchten M, de Vries EG, Glaudemans AW, et al. Measuring residual estrogen receptor availability during fulvestrant therapy in patients with metastatic breast cancer. Cancer Discov. 2015;5(1):72–81. doi: 10.1158/-14-0697[published.
    1. Dehdashti F, Mortimer JE, Trinkaus K, et al. PET-based estradiol challenge as a predictive biomarker of response to endocrine therapy in women with estrogen-receptor-positive breast cancer. Breast Cancer Res Treat. 2009;113(3):509–517. doi: 10.1007/s10549-008-9953-0.
    1. Kurland BF, Peterson LM, Lee JH, et al. Estrogen receptor binding (18F-FES PET) and glycolytic activity (18F-FDG PET) Predict progression-free survival on endocrine therapy in patients with ER+ breast cancer. Clin Cancer Res. 2017;23(2):407–415. doi: 10.1158/1078-0432.ccr-16-0362.
    1. Guan J, Zhou W, Hafner M, et al. Therapeutic ligands antagonize estrogen receptor function by impairing its mobility. Cell. 2019;178(4):949–63.e18. doi: 10.1016/j.cell.2019.06.026.
    1. Pinkerton JV, Goldstein SR. Endometrial safety: a key hurdle for selective estrogen receptor modulators in development. Menopause. 2010;17(3):642–653. doi: 10.1097/gme.0b013e3181c4f1d6.
    1. Garuti G, Cellani F, Centinaio G, Montanari G, Nalli G, Luerti M. Prospective endometrial assessment of breast cancer patients treated with third generation aromatase inhibitors. Gynecol Oncol. 2006;103(2):599–603. doi: 10.1016/j.ygyno.2006.04.004.
    1. Kahraman M, Govek SP, Nagasawa JY, et al. Maximizing ER-alpha degradation maximizes activity in a tamoxifen-resistant breast cancer model: identification of GDC-0927. ACS Med Chem Lett. 2019;10(1):50–55. doi: 10.1021/acsmedchemlett.8b00414.
    1. Liang J, Zbieg JR, Blake RA, et al. GDC-9545 (Giredestrant): a potent and orally bioavailable selective estrogen receptor antagonist and degrader with an exceptional preclinical profile for ER+ breast cancer. J Med Chem. 2021;64(16):11841–11856. doi: 10.1021/acs.jmedchem.1c00847.
    1. Kahraman M, Govek SP, Nagasawa JY, et al. Abstract 1648: discovery and evolution of orally bioavailable selective estrogen receptor degraders for ER+ breast cancer: from GDC-0810 to GDC-0927. Cancer Res. 2018;78(13 Supplement):1648–1748. doi: 10.1158/1538-7445.am2018-1648.
    1. Dickler M, Villanueva R, Perez Fidalgo J, et al. 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. Cancer Res. 2018 doi: 10.1158/1538-7445.SABCS17-PD5-10.
    1. Jhaveri K, Winer EP, Lim E, et al. Abstract PD7–05: a first-in-human phase I study to evaluate the oral selective estrogen receptor degrader (SERD), GDC-9545, in postmenopausal women with estrogen receptor-positive (ER+) HER2-negative (HER2-) metastatic breast cancer. Cancer Res. 2020;80(4 Supplement):PD7-05–PD7-05. doi: 10.1158/1538-7445.sabcs19-pd7-05.

Source: PubMed

3
Abonneren