AMEERA-1 phase 1/2 study of amcenestrant, SAR439859, in postmenopausal women with ER-positive/HER2-negative advanced breast cancer

Aditya Bardia, Sarat Chandarlapaty, Hannah M Linden, Gary A Ulaner, Alice Gosselin, Sylvaine Cartot-Cotton, Patrick Cohen, Séverine Doroumian, Gautier Paux, Marina Celanovic, Vasiliki Pelekanou, Jeffrey E Ming, Nils Ternès, Monsif Bouaboula, Joon Sang Lee, Anne-Laure Bauchet, Mario Campone, Aditya Bardia, Sarat Chandarlapaty, Hannah M Linden, Gary A Ulaner, Alice Gosselin, Sylvaine Cartot-Cotton, Patrick Cohen, Séverine Doroumian, Gautier Paux, Marina Celanovic, Vasiliki Pelekanou, Jeffrey E Ming, Nils Ternès, Monsif Bouaboula, Joon Sang Lee, Anne-Laure Bauchet, Mario Campone

Abstract

AMEERA-1 is a Phase 1/2 open-label single-arm study evaluating once-daily (QD) amcenestrant, an orally bioavailable selective estrogen receptor (ER) degrader, in postmenopausal women with ER+/HER2- advanced breast cancer (NCT03284957), who were mostly heavily pretreated (including targeted therapies and fulvestrant). In the dose escalation phase (Part A: n = 16), patients received amcenestrant 20-600 mg QD. Based on absence of dose-limiting toxicities, paired functional 18F-fluoroestradiol positron emission tomography, and pharmacokinetics, 400 mg QD was selected as recommended Phase 2 dose (RP2D) for the dose expansion phase (Part B: n = 49). No Grade ≥3 treatment-related adverse events or clinically significant cardiac/eye toxicities were reported. The Part B primary endpoint, confirmed objective response rate (ORR) was 3/45 at the interim analysis and 5/46 (10.9%) at the final analysis. The overall clinical benefit rate (CBR) was 13/46 (28.3%). CBRs among patients with baseline wild-type and mutated ESR1 were 9/26 (34.6%) and 4/19 (21.1%), respectively. Paired tumor biopsy and cell-free DNA analyses revealed ER inhibition and degradation, and a reduction in detectable ESR1 mutations, including Y537S. In conclusion, amcenestrant at RP2D of 400 mg QD for monotherapy is well-tolerated with no dose-limiting toxicities, and demonstrates preliminary antitumor activity irrespective of baseline ESR1 mutation status.

Conflict of interest statement

A.B. has provided consulting or advisory roles for Astra Zeneca/Daiichi Sankyo, Lilly, Foundation Medicine, Gilead, Novartis, Genentech, Pfizer, Spectrum Pharmaceuticals, bioTheranostics, Merck, Radius Health, Immunomedics, Sanofi, and Puma Biotechnology, an institution consulting or advisory role for Immunomedics, Novartis, Genentech/Roche, Pfizer, Radius Health, Sanofi, and Innocrin Pharmaceuticals, and has received research funding from bioTheranostics. S.C. has received research support (to Memorial Sloan Kettering) from Daiichi Sankyo and Paige.ai, clinical trial funding (to Memorial Sloan Kettering) from Sanofi, Novartis, and Lilly, and consulting honoraria (to S.C.) from AstraZeneca, Inivata, Lilly, Novartis Sanofi, and Targeted Oncology. H.M.L. has provided consulting or advisory roles for Eisai and Pfizer, and her institution has received funding from Zionexa, Sanofi, Zentalis, Eisai, and Zymeworks. G.A.U. has provided consulting and/or has received research support from GE Healthcare, Genentech, Sanofi, Novartis, Puma Biotechnology, Siemens, Zionexa, and Lantheus. A.G., S.C-C., P.C., S.D., G.P., M. Celanovic, V.P., J.E.M., N.T., M.B., J.S.L., and A-L.B. are employees of Sanofi and may hold shares and/or stock options in the company. M. Campone has provided consulting, advisory, or speaker roles for AbbVie, Accord Healthcare, AstraZeneca, Daiichi Sankyo, G1 Therapeutics, Lilly, Novartis, Pfizer, Pierre Fabre Oncology, Sandoz, Sanofi, Seagene, and Servier; and has received honoraria for travel from AstraZeneca, Novartis, Pfizer, and Roche.

© 2022. The Author(s).

Figures

Fig. 1. Study flow diagram for Parts…
Fig. 1. Study flow diagram for Parts A and B.
During Part A, patients were enrolled into a dose escalation phase of amcenestrant monotherapy from 20-600 mg once daily. During Part B, patients were enrolled into a dose-expansion phase at the RP2D determined in Part A. aThe first patient treated at each new DL will be followed for a minimum of 1 week prior to enrolling and treating two additional patients. If none of the three patients experience a DLT, the next cohort starts one DL higher. If one of the three patients experiences a DLT, up to three additional patients are treated at this DL. If two or more patients experience a DLT, the maximum administered dose is reached. bDL2bis is investigated if at least one patient at DL1 shows ≤ 30% inhibition of the target by 18F-FES PET or all patients treated at DL2 have >85% inhibition of the target by 18F-FES PET. DL dose level, DLT dose-limiting toxicity, ER+ estrogen receptor positive, 18F-FES PET 18F-fluoroestradiol positron emission tomography, HER2− human epidermal growth factor receptor 2-negative, MTD maximum tolerated dose, RP2D recommended Phase 2 dose, PK pharmacokinetics, QD once daily.
Fig. 2. Part A dose escalation: amcenestrant…
Fig. 2. Part A dose escalation: amcenestrant plasma concentration-time profiles, pharmacokinetic/pharmacodynamic relationships, and 18F-FES PET/CT images.
a Mean amcenestrant plasma concentration-time profiles observed following repeated oral administration (cycle 1, day 22) under fasting conditions; b pharmacokinetic/pharmacodynamic relationship between plasma concentrations of amcenestrant measured just before 18F-FES radioisotope administration and individual mean percent occupancy of estrogen receptors (mean percent reduction in 18F-FES SUVmax(cor)); c representative 18F-FES PET; and d coronal CT images at screening and on-treatment with amcenestrant 150 mg. 18F-FES PET/CT scans were performed 16–24 h after the previous amcenestrant dose, except for two patients whose scans occurred 8 h after the previous dosea. Of 16 patients, 14 patients had scans at baseline and between 11–15 days after first amcenestrant administration (as per protocol) and two patients had post-baseline scans on days 10 and 28, respectively. The patient with a post-baseline scan on day 10 was included in the pharmacodynamic analysis because the scan occurred after ≥8 days of continuous treatment, but the patient receiving amcenestrant 200 mg with a post-baseline scan on day 28 was excluded from pharmacodynamic analysis and dose escalation decisionsb. cAnnotated example of the reduction in SUVmax(cor) for the first index lesion in the 5th lumbar vertebra; physiologic 18F-FES avidity is noted in the liver, intestines, and bladder. dAnnotated example of the reduction in tumor diameter of a right pelvic lymph node in the associated CT scan. CT computerized tomography, ER estrogen receptor 18F-FES PET 18F-fluoroestradiol positron emission tomography, LLOQ lower limit of quantification, SUVmax(cor) maximum standardized uptake value, standardized by body weight and corrected for background.
Fig. 3. Waterfall and swimmer plots.
Fig. 3. Waterfall and swimmer plots.
a Waterfall plot of best relative change from baseline in the sum of diameters of target lesions in the response-evaluable populations of Part A by local investigators/radiologists review (n = 16) and Part B by independent central review (n = 41; three patients were missing relative change data and two patients had a non-evaluable best overall response) and b Swimmer plot of duration of treatment in the safety populations of Part A (n = 16) and Part B (n = 49) with overall responses assessed by local investigators/radiologists review. CDK4/6i cyclin-dependent kinase 4/6 inhibitor, chemo chemotherapy, ICR independent central review, Local local investigators/radiologists review, mTORi mammalian target of rapamycin inhibitor, SERD selective estrogen receptor degrader. Checkboxes correspond to baseline characteristics.
Fig. 4. Post hoc exploratory biomarker analyses…
Fig. 4. Post hoc exploratory biomarker analyses of on-target ER degradation/pathway inhibition during amcenestrant therapy.
Changes from screening to cycle 2, day 28 in a ER nucleus H-score by IHC; b PgR nucleus H-score by IHC; c percent of positive cells showing Ki67 protein expression by IHC; and d ER activation scores by GSVA; and changes from cycle 1, day 1 to cycle 2, day 28 in e allele frequency of ESR1 mutations in cfDNA by ddPCR; and f numbers of ESR1 mutations in cfDNA by ddPCR. *Each line refers to an ESR1 mutation, but several lines may refer to a single patient. C1D1, cycle 1, day 1; C1D15, cycle 1, day 15; C2D28, cycle 2, day 28; CB clinical benefit (complete response + partial response + stable disease ≥ 24 weeks), cfDNA circulating free DNA, ddPC, droplet digital polymerase chain reaction, ER estrogen receptor, ESR1 estrogen receptor 1, GSVA gene set variation analysis, IHC immunohistochemistry, PgR progesterone receptor, PR partial response.

References

    1. Bray F, et al. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018;68:394–424. doi: 10.3322/caac.21492.
    1. American Cancer Society. Breast cancer facts & figures 2019−2020. (2019).
    1. National Cancer Institute Surveillance Epidemiology and End Results Program. Cancer stat facts: female breast cancer subtypes. (2013–2017).
    1. Waks AG, Winer EP. Breast cancer treatment: a review. JAMA. 2019;321:288–300. doi: 10.1001/jama.2018.19323.
    1. National Cancer Institute Surveillance Epidemiology and End Results Program. SEER*Explorer. (2020).
    1. Patel HK, Bihani T. Selective estrogen receptor modulators (SERMs) and selective estrogen receptor degraders (SERDs) in cancer treatment. Pharm. Ther. 2018;186:1–24. doi: 10.1016/j.pharmthera.2017.12.012.
    1. Chandarlapaty S, et al. Prevalence of ESR1 mutations in cell-free DNA and outcomes in metastatic breast cancer: a secondary analysis of the BOLERO-2 clinical trial. JAMA Oncol. 2016;2:1310–1315. doi: 10.1001/jamaoncol.2016.1279.
    1. Fribbens C, et al. Plasma ESR1 mutations and the treatment of estrogen receptor-positive advanced breast cancer. J. Clin. Oncol. 2016;34:2961–2968. doi: 10.1200/JCO.2016.67.3061.
    1. Jeselsohn R, et al. Emergence of constitutively active estrogen receptor-alpha mutations in pretreated advanced estrogen receptor-positive breast cancer. Clin. Cancer Res. 2014;20:1757–1767. doi: 10.1158/1078-0432.CCR-13-2332.
    1. Schiavon G, et al. Analysis of ESR1 mutation in circulating tumor DNA demonstrates evolution during therapy for metastatic breast cancer. Sci. Transl. Med. 2015;7:313ra182. doi: 10.1126/scitranslmed.aac7551.
    1. O’Leary B, et al. The genetic landscape and clonal evolution of breast cancer resistance to palbociclib plus fulvestrant in the PALOMA-3 trial. Cancer Discov. 2018;8:1390–1403. doi: 10.1158/-18-0264.
    1. Griffith OL, et al. The prognostic effects of somatic mutations in ER-positive breast cancer. Nat. Commun. 2018;9:3476. doi: 10.1038/s41467-018-05914-x.
    1. Razavi P, et al. The genomic landscape of endocrine-resistant advanced breast cancers. Cancer Cell. 2018;34:427–438.e6. doi: 10.1016/j.ccell.2018.08.008.
    1. Bertucci F, et al. Genomic characterization of metastatic breast cancers. Nature. 2019;569:560–564. doi: 10.1038/s41586-019-1056-z.
    1. Nagarajan S, et al. ARID1A influences HDAC1/BRD4 activity, intrinsic proliferative capacity and breast cancer treatment response. Nat. Genet. 2020;52:187–197. doi: 10.1038/s41588-019-0541-5.
    1. Xu G, et al. ARID1A determines luminal identity and therapeutic response in estrogen-receptor-positive breast cancer. Nat. Genet. 2020;52:198–207. doi: 10.1038/s41588-019-0554-0.
    1. Osborne CK, Schiff R. Mechanisms of endocrine resistance in breast cancer. Annu. Rev. Med. 2011;62:233–247. doi: 10.1146/annurev-med-070909-182917.
    1. Niu Y, Xu J, Sun T. Cyclin-dependent kinases 4/6 inhibitors in breast cancer: current status, resistance, and combination strategies. J. Cancer. 2019;10:5504–5517. doi: 10.7150/jca.32628.
    1. Jeselsohn, R., Buchwalter, G., De Angelis, C., Brown, M. & Schiff, R. ESR1 mutations-a mechanism for acquired endocrine resistance in breast cancer. Nat. Rev. Clin. Oncol. 12, 573–583 (2015).
    1. Toy W, et al. Activating ESR1 mutations differentially affect the efficacy of ER antagonists. Cancer Discov. 2017;7:277–287. doi: 10.1158/-15-1523.
    1. Traboulsi T, El Ezzy M, Gleason JL, Mader S. Antiestrogens: structure-activity relationships and use in breast cancer treatment. J. Mol. Endocrinol. 2017;58:R15–R31. doi: 10.1530/JME-16-0024.
    1. Mortimer, J. E. & Urban, J. H. Long-term toxicities of selective estrogen-receptor modulators and antiaromatase agents. Oncology17, 652–659, discussion 659, 662, 666 passim (2003).
    1. Hamilton EP, et al. A phase I dose escalation and expansion study of the next generation oral SERD AZD9833 in women with ER-positive, HER2-negative advanced breast cancer. J. Clin. Oncol. 2020;38:1024. doi: 10.1200/JCO.2020.38.15_suppl.1024.
    1. Lim E, et al. A phase Ib study to evaluate the oral selective estrogen receptor degrader GDC-9545 alone or combined with palbociclib in metastatic ER-positive HER2-negative breast cancer. J. Clin. Oncol. 2020;38:1023. doi: 10.1200/JCO.2020.38.15_suppl.1023.
    1. van Kruchten M, et al. Measuring residual estrogen receptor availability during fulvestrant therapy in patients with metastatic breast cancer. Cancer Discov. 2015;5:72–81. doi: 10.1158/-14-0697.
    1. AstraZeneca. FASLODEX® (fulvestrant) injection, for intramuscular use. (2020).
    1. Hopkins, U. & Arias, C.Y. Large-volume IM injections: a review of best practices. Oncol. Nurse Advis. February 22, 32–37 (2013).
    1. Ohno S, et al. Three dose regimens of fulvestrant in postmenopausal Japanese women with advanced breast cancer: results from a double-blind, phase II comparative study (FINDER1) Ann. Oncol. 2010;21:2342–2347. doi: 10.1093/annonc/mdq249.
    1. Pritchard KI, et al. Results of a phase II study comparing three dosing regimens of fulvestrant in postmenopausal women with advanced breast cancer (FINDER2) Breast Cancer Res. Treat. 2010;123:453–461. doi: 10.1007/s10549-010-1022-9.
    1. He M, et al. The predictive value of early changes in (18) F-fluoroestradiol positron emission tomography/computed tomography during fulvestrant 500 mg therapy in patients with estrogen receptor-positive metastatic breast cancer. Oncologist. 2020;25:927–936. doi: 10.1634/theoncologist.2019-0561.
    1. Shomali M, et al. SAR439859, a novel selective estrogen receptor degrader (SERD), demonstrates effective and broad antitumor activity in wild-type and mutant ER-positive breast cancer models. Mol. Cancer Ther. 2021;20:250–262. doi: 10.1158/1535-7163.MCT-20-0390.
    1. Bouaboula M, et al. Abstract 943: SAR439859, an orally bioavailable selective estrogen receptor degrader (SERD) that demonstrates robust antitumor efficacy and limited cross-resistance in ER+ breast cancer. Cancer Res. 2018;78:943. doi: 10.1158/1538-7445.AM2018-943.
    1. El-Ahmad, Y. et al. Discovery of 6-(2,4-Dichlorophenyl)−5-[4-[(3S)−1-(3-fluoropropyl)pyrrolidin-3-yl]oxyphenyl]−8, 9-dihydro-7H-benzo[7]annulene-2-carboxylic acid (SAR439859), a potent and selective estrogen receptor degrader (SERD) for the treatment of estrogen-receptor-positive breast cancer. J. Med. Chem. 63, 512–528 (2020).
    1. Besret L, et al. Translational strategy using multiple nuclear imaging biomarkers to evaluate target engagement and early therapeutic efficacy of SAR439859, a novel selective estrogen receptor degrader. EJNMMI Res. 2020;10:70. doi: 10.1186/s13550-020-00646-w.
    1. Brett JO, Spring LM, Bardia A, Wander SA. ESR1 mutation as an emerging clinical biomarker in metastatic hormone receptor-positive breast cancer. Breast Cancer Res. 2021;23:85. doi: 10.1186/s13058-021-01462-3.
    1. Bardia, A. et al. GS2-02. Elacestrant, an oral selective estrogen receptor degrader (SERD), vs investigator’s choice of endocrine monotherapy for ER + /HER2- advanced/metastatic breast cancer (mBC) following progression on prior endocrine and CDK4/6 inhibitor therapy: results of EMERALD phase 3 trial. in San Antonio Breast Cancer Symposium December 7–10, 2021 (San Antonio, Texas, 2021).
    1. Kalinsky, K., Albramson, V., Chalasani, P. & Linden, H.M. Zn-C5. in San Antonio Breast Cancer Symposium December 7–10, 2021 (San Antonio, Texas, 2021).
    1. Turner, N.C. et al. PD13-07. Activity and biomarker analyses from a phase Ia/b study of giredestrant (GDC-9545; G) with or without palbociclib (palbo) in patients with estrogen receptor-positive, HER2-negative locally advanced/metastatic breast cancer (ER + /HER2- LA/mBC). in San Antonio Breast Cancer Symposium December 7–10, 2021 (San Antonio, Texas, 2021).
    1. Pritchard KI, et al. Results of a phase II study comparing three dosing regimens of fulvestrant in postmenopausal women with advanced breast cancer (FINDER2) Breast Cancer Res. Treat. 2010;123:453–461. doi: 10.1007/s10549-010-1022-9.
    1. Cristofanilli M, et al. Fulvestrant plus palbociclib versus fulvestrant plus placebo for treatment of hormone-receptor-positive, HER2-negative metastatic breast cancer that progressed on previous endocrine therapy (PALOMA-3): final analysis of the multicentre, double-blind, phase 3 randomised controlled trial. Lancet Oncol. 2016;17:425–439. doi: 10.1016/S1470-2045(15)00613-0.
    1. Jones RH, et al. Fulvestrant plus capivasertib versus placebo after relapse or progression on an aromatase inhibitor in metastatic, oestrogen receptor-positive breast cancer (FAKTION): a multicentre, randomised, controlled, phase 2 trial. Lancet Oncol. 2020;21:345–357. doi: 10.1016/S1470-2045(19)30817-4.
    1. Andre F, et al. Alpelisib for PIK3CA-mutated, hormone receptor-positive advanced breast cancer. N. Engl. J. Med. 2019;380:1929–1940. doi: 10.1056/NEJMoa1813904.
    1. Dent S, et al. Phase III randomized study of taselisib or placebo with fulvestrant in estrogen receptor-positive, PIK3CA-mutant, HER2-negative, advanced breast cancer: the SANDPIPER trial. Ann. Oncol. 2021;32:197–207. doi: 10.1016/j.annonc.2020.10.596.
    1. Moy B, et al. Chemotherapy and targeted therapy for patients with human epidermal growth factor receptor 2-negative metastatic breast cancer that is either endocrine-pretreated or hormone receptor-negative: ASCO guideline update. J. Clin. Oncol. 2021;39:3938–3958. doi: 10.1200/JCO.21.01374.
    1. Wang Y, 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;23:3053–3060. doi: 10.1158/1078-0432.CCR-16-2197.
    1. Besret L, et al. Translational strategy using multiple nuclear imaging biomarkers to evaluate target engagement and early therapeutic efficacy of SAR439859, a novel selective estrogen receptor degrader. EJNMMI Res. 2020;10:70. doi: 10.1186/s13550-020-00646-w.
    1. Dobin A, et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. 2013;29:15–21. doi: 10.1093/bioinformatics/bts635.
    1. Trapnell C, et al. Transcript assembly and quantification by RNA-Seq reveals unannotated transcripts and isoform switching during cell differentiation. Nat. Biotechnol. 2010;28:511–515. doi: 10.1038/nbt.1621.
    1. Chandarlapaty S, et al. 277MO SAR439859, an oral selective estrogen receptor (ER) degrader (SERD), in ER+/ HER2- metastatic breast cancer (mBC): biomarker analyses from a phase I/II study. Ann. Oncol. 2020;31:277MO. doi: 10.1016/j.annonc.2020.08.379.

Source: PubMed

Подписаться