SOLTI-1805 TOT-HER3 Study Concept: A Window-of-Opportunity Trial of Patritumab Deruxtecan, a HER3 Directed Antibody Drug Conjugate, in Patients With Early Breast Cancer

Tomás Pascual, Mafalda Oliveira, Eva Ciruelos, Meritxell Bellet Ezquerra, Cristina Saura, Joaquin Gavilá, Sonia Pernas, Montserrat Muñoz, Maria J Vidal, Mireia Margelí Vila, Juan M Cejalvo, Blanca González-Farré, Martin Espinosa-Bravo, Josefina Cruz, Francisco Javier Salvador-Bofill, Juan Antonio Guerra, Ana María Luna Barrera, Miriam Arumi de Dios, Stephen Esker, Pang-Dian Fan, Olga Martínez-Sáez, Guillermo Villacampa, Laia Paré, Juan M Ferrero-Cafiero, Patricia Villagrasa, Aleix Prat, Tomás Pascual, Mafalda Oliveira, Eva Ciruelos, Meritxell Bellet Ezquerra, Cristina Saura, Joaquin Gavilá, Sonia Pernas, Montserrat Muñoz, Maria J Vidal, Mireia Margelí Vila, Juan M Cejalvo, Blanca González-Farré, Martin Espinosa-Bravo, Josefina Cruz, Francisco Javier Salvador-Bofill, Juan Antonio Guerra, Ana María Luna Barrera, Miriam Arumi de Dios, Stephen Esker, Pang-Dian Fan, Olga Martínez-Sáez, Guillermo Villacampa, Laia Paré, Juan M Ferrero-Cafiero, Patricia Villagrasa, Aleix Prat

Abstract

Background: Preclinical data support a key role for the human epidermal growth factor receptor 3 (HER3) pathway in hormone receptor (HR)-positive breast cancer. Recently, new HER3 directed antibody drug conjugates have shown activity in breast cancer. Given the need to better understand the molecular biology, tumor microenvironment, and mechanisms of drug resistance in breast cancer, we designed this window-of-opportunity study with the HER3 directed antibody drug conjugate patritumab deruxtecan (HER3-DXd; U3-1402). Trial Design: Based on these data, a prospective, multicenter, single-arm, window-of-opportunity study was designed to evaluate the biological effect of patritumab deruxtecan in the treatment of naïve patients with HR-positive/HER2-negative early breast cancer whose primary tumors are ≥1 cm by ultrasound evaluation. Patients will be enrolled in four cohorts according to the mRNA-based ERBB3 expression by central assessment. The primary endpoint is a CelTIL score after one single dose. A translational research plan is also included to provide biological information and to evaluate secondary and exploratory objectives of the study. Trial Registration Number: EudraCT 2019-004964-23; NCT number: NCT04610528.

Keywords: Breast Cancer; CelTIL Score; ERBB3; HER3; HER3-DXd; U3-1402; patritumab deruxtecan.

Conflict of interest statement

SE and PF are employed by Daiichi Sankyo, Inc. AP has declared personal honoraria from Pfizer, Novartis, Roche, MSD Oncology, Lilly, and Daiichi Sankyo, travel, accommodations, and expenses paid by Daiichi Sankyo, research funding from Nanostring Technologies, Roche, and Novartis, and consulting/advisory role for Nanostring Technologies, Roche, Novartis, Pfizer, Oncolytics Biotech, Amgen, Lilly, MSD, PUMA, and Daiichi Sankyo, Inc. outside the submitted work. MO reports honoraria and consulting fees from Roche/Genentech, GSK, PUMA Biotechnology, AstraZeneca, Seattle Genetics, and Novartis; travel and accommodation paid by Roche, Pierre-Fabre, Novartis, GP Pharma, Grünenthal, and Eisai; and grant/Research Support (to the Institution) from AstraZeneca, Philips Healthcare, Genentech, Roche, Novartis, Immunomedics, Seattle Genetics, GSK, Boehringer-Ingelheim, PUMA Biotechnology, and Zenith Epigenetics outside the submitted work. EC reports personal fees from Roche, personal fees from Lilly, personal fees from Novartis, and personal fees from Pfizer, during the conduct of the study. SP reports an advisor/consultant role for AstraZeneca, Daiichi-Sankyo, Polyphor, and Roche, and travel and accommodation paid by Novartis. JC reports an advisor/consultant role for Roche, Novartis, Pfyzer, Pharmamar, Lilly, Eisai, and Amgen, and travel and accommodation by Novartis and Pharmamar. FS-B has declared personal honoraria from Pfizer, Novartis, Roche, and Daiichi Sankyo. PV has received honoraria as a Speaker from Nanostring. MM has declared an advisor role or consulting from Novartis, Pfizer, and Roche; research funding from Roche, Eisai, and AstraZeneca; and travel expenses from Roche. OM-S reports an advisor role from Roche; honoraria as a speaker from Eisai; and travel expenses from Novartis. GV reports receiving honoraria for speaker activities from MSD and an advisory role from AstraZeneca. CS has served as a consultant, participated in advisory boards, or received travel grants from AstraZeneca, Celgene, Daiichi Sankyo, Eisai, F. Hoffmann—La Roche Ltd., Genomic Health, Merck, Sharp and Dhome España S.A., Novartis, Odonate Therapeutics, Pfizer, Philips Healthwork, Pierre Fabre, prIME Oncology, Puma, Synthon, and Sanofi Aventis. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2021 Pascual, Oliveira, Ciruelos, Bellet Ezquerra, Saura, Gavilá, Pernas, Muñoz, Vidal, Margelí Vila, Cejalvo, González-Farré, Espinosa-Bravo, Cruz, Salvador-Bofill, Guerra, Luna Barrera, Arumi de Dios, Esker, Fan, Martínez-Sáez, Villacampa, Paré, Ferrero-Cafiero, Villagrasa and Prat.

Figures

Figure 1
Figure 1
TOT-HER3 trial design.
Figure 2
Figure 2
Measurement of ERBB3 expression in breast cancer using the nCounter platform. (A) Box plots of ERBB3 gene expression in breast tumors as classified by hormone receptor and HER2 expression and intrinsic subtype. (B) Unsupervised hierarchical clustering using the 50 PAM50 genes and ERBB3 (rows) and 1,580 tumor samples (columns). Each colored square on the heatmap represents the relative median signature score for each sample with the highest expression being red, the lowest expression being green, and the average expression being black. (C) Pearson correlation between ERBB3, single genes, and PAM50 gene expression signatures evaluated in breast cancer samples.
Figure 3
Figure 3
Comparing ERBB3 expression across datasets (A) Evaluation of ERBB3 cutoff in breast cancer samples from patients with early breast cancer included in IN-HOUSE, METABRIC, and TCGA. (B) Proportion of samples in each immunohistochemistry subtype based on the ERBB3 cohort. Each bar is colored according to the ERBB3 distribution in each cohort. (C) Correlation coefficients of proportions of tumor samples within each quartile based on the IHC subtype between the three datasets. (D) Scatter plots of ERBB3 vs. ESR1 expression for samples from METABRIC, IN-HOUSE, and TCGA cohorts, colored by subtype. The three horizontal lines indicate the cutoffs of each cohort. Discontinued line in each figure represents the regression line. Pearson correlation coefficient (r) with significance (p-value) is presented in each figure.

References

    1. Ocana A, Vera-Badillo F, Seruga B, Templeton A, Pandiella A, Amir E. HER3 overexpression and survival in solid tumors: a meta-analysis. JNCI J Natl Cancer Inst. (2013) 105:266–73. 10.1093/jnci/djs501
    1. Campbell MR, Amin D, Moasser MM. HER3 comes of age: new insights into its functions and role in signaling, tumor biology, and cancer therapy. Clin Cancer Res. (2010) 16:1373–83. 10.1158/1078-0432.CCR-09-1218
    1. Thrane S, Lykkesfeldt AE, Larsen MS, Sorensen BS, Yde CW. Estrogen receptor α is the major driving factor for growth in tamoxifen-resistant breast cancer and supported by HER/ERK signaling. Breast Cancer Res Treat. (2013) 139:71–80. 10.1007/s10549-013-2485-2
    1. Collins D, Jacob W, Cejalvo JM, Ceppi M, James I, Hasmann M, et al. . Direct estrogen receptor (ER)/HER family crosstalk mediating sensitivity to lumretuzumab and pertuzumab in ER+ breast cancer. PLoS ONE. (2017) 12:e0177331. 10.1371/journal.pone.0177331
    1. Olayioye MA, Neve RM, Lane HA, Hynes NE. The ErbB signaling network: receptor heterodimerization in development and cancer. EMBO J. (2000) 19:3159–67. 10.1093/emboj/19.13.3159
    1. Hashimoto Y, Koyama K, Kamai Y, Hirotani K, Ogitani Y, Zembutsu A, et al. . A novel HER3-targeting antibody–drug conjugate, U3-1402, exhibits potent therapeutic efficacy through the delivery of cytotoxic payload by efficient internalization. Clin Cancer Res. (2019) 25:7151–61. 10.1158/1078-0432.CCR-19-1745
    1. Koyama K, Ishikawa H, Abe M, Shiose Y, Ueno S, Nakamaru K, et al. . Abstract 5201: U3-1402, a novel HER3-targeting antibody-drug conjugate, exhibits in vitro antitumor activity against breast cancer cells expressing HER3 mutations without dependence on HER2 overexpression. Cancer Res. (2020) 80(16 Supplement):5201. 10.1158/1538-7445.AM2020-5201
    1. Haratani K, Yonesaka K, Takamura S, Maenishi O, Kato R, Takegawa N, et al. . U3-1402 sensitizes HER3-expressing tumors to PD-1 blockade by immune activation. J Clin Invest. (2019) 130:374–8. 10.1172/JCI126598
    1. Yonemori K, Masuda N, Takahashi S, Kogawa T, Nakayama T, Yamamoto Y, et al. . 151O Single agent activity of U3-1402, a HER3-targeting antibody-drug conjugate, in HER3-overexpressing metastatic breast cancer: Updated results from a phase I/II trial. Ann Oncol. (2019). 30(Suppl. 3):mdz100. 002. 10.1093/annonc/mdz100.002
    1. Yu H, Baik C, Gold K, Hayashi H, Johnson M, Koczywas M, et al. . LBA62 Efficacy and safety of patritumab deruxtecan (U3-1402), a novel HER3 directed antibody drug conjugate, in patients (pts) with EGFR-mutated (EGFRm) NSCLC. Ann Oncol. (2020) 31:S1189–90. 10.1016/j.annonc.2020.08.2295
    1. Bae SY, La Choi Y, Kim S, Kim M, Kim J, Jung SP, et al. . HER3 status by immunohistochemistry is correlated with poor prognosis in hormone receptor-negative breast cancer patients. Breast Cancer Res Treat. (2013) 139:741–50. 10.1007/s10549-013-2570-6
    1. Luhtala S, Staff S, Kallioniemi A, Tanner M, Isola J. Clinicopathological and prognostic correlations of HER3 expression and its degradation regulators, NEDD4-1 and NRDP1, in primary breast cancer. BMC Cancer. (2018) 18:1045. 10.1186/s12885-018-4917-1
    1. Ogden A, Bhattarai S, Sahoo B, Mongan NP, Alsaleem M, Green AR, et al. . Combined HER3-EGFR score in triple-negative breast cancer provides prognostic and predictive significance superior to individual biomarkers. Sci Rep. (2020) 10:1–8. 10.1038/s41598-020-59514-1
    1. Pascual T, Oliveira M, Ciruelos E, Ezquerra MB, Saura C, Gregori JG, et al. . 2O ERBB3 mRNA expression in breast cancer (BC): A SOLTI biomarker discovery analysis. Ann Oncol. (2020) 31:S15–S16. 10.1016/j.annonc.2020.03.138
    1. Denkert C, von Minckwitz G, Darb-Esfahani S, Lederer B, Heppner BI, Weber KE, et al. . Tumour-infiltrating lymphocytes and prognosis in different subtypes of breast cancer: a pooled analysis of 3771 patients treated with neoadjuvant therapy. Lancet Oncol. (2018) 19:40–50. 10.1016/S1470-2045(17)30904-X
    1. Loi S, Giobbie-Hurder A, Gombos A, Bachelot T, Hui R, Curigliano G, et al. . Pembrolizumab plus trastuzumab in trastuzumab-resistant, advanced, HER2-positive breast cancer (PANACEA): a single-arm, multicentre, phase 1b-2 trial. Lancet Oncol. (2019) 20:371–82. 10.1016/S1470-2045(18)30812-X
    1. Loi S, Sirtaine N, Piette F, Salgado R, Viale G, Van Eenoo F, et al. . Prognostic and predictive value of tumor-infiltrating lymphocytes in a phase III randomized adjuvant breast cancer trial in node-positive breast cancer comparing the addition of docetaxel to doxorubicin with doxorubicin-based chemotherapy: BIG 02-98. J Clin Oncol. (2013) 31:860–7. 10.1200/JCO.2011.41.0902
    1. Adams S, Gray RJ, Demaria S, Goldstein L, Perez EA, Shulman LN, et al. . Prognostic value of tumor-infiltrating lymphocytes in triple-negative breast cancers from two phase III randomized adjuvant breast cancer trials: ECOG 2197 and ECOG 1199. J Clin Oncol. (2014) 32:2959–66. 10.1200/JCO.2013.55.0491
    1. Luen SJ, Salgado R, Fox S, Savas P, Eng-Wong J, Clark E, et al. . Tumour-infiltrating lymphocytes in advanced HER2-positive breast cancer treated with pertuzumab or placebo in addition to trastuzumab and docetaxel: a retrospective analysis of the CLEOPATRA study. Lancet Oncol. (2017) 18:52–62. 10.1016/S1470-2045(16)30631-3
    1. Asano Y, Kashiwagi S, Goto W, Takada K, Takahashi K, Hatano T, et al. . Prediction of survival after neoadjuvant chemotherapy for breast cancer by evaluation of tumor-infiltrating lymphocytes and residual cancer burden. BMC Cancer. (2017) 17:888. 10.1186/s12885-017-3927-8
    1. Harbeck N, Gluz O, Christgen M, Kates RE, Braun M, Küemmel S, et al. . De-Escalation Strategies in Human Epidermal Growth Factor Receptor 2 (HER2)–positive early breast cancer (BC): final analysis of the West German study group adjuvant dynamic marker-adjusted personalized therapy trial optimizing risk assessment and therapy response prediction in early BC HER2- and hormone receptor–positive phase ii randomized trial—efficacy, safety, and predictive markers for 12 weeks of neoadjuvant trastuzumab emtansine with or without endocrine therapy (ET) versus trastuzumab plus ET. J Clin Oncol. (2017) 35:3046–54. 10.1200/JCO.2016.71.9815
    1. Nuciforo P, Pascual T, Cortés J, Llombart-Cussac A, Fasani R, Paré L, et al. . A predictive model of pathologic response based on tumor cellularity and tumor-infiltrating lymphocytes (CelTIL) in HER2-positive breast cancer treated with chemo-free dual HER2 blockade. Ann Oncol. (2018) 29:170–7. 10.1093/annonc/mdx647
    1. Colomer R Saura C Sánchez-Rovira P Pascual T Rubio IT Burgués O . Neoadjuvant management of early breast cancer: a clinical and investigational position statement. Oncologist. (2019) 24:603–11. 10.1634/theoncologist.2018-0228
    1. Pascual T, Cejalvo JM, Oliveira M, Vidal M, Vega E, Ganau S, et al. . SOLTI-1503 PROMETEO TRIAL: combination of talimogene laherparepvec with atezolizumab in early breast cancer. Future Oncol. (2020) 16:1801–13. 10.2217/fon-2020-0246
    1. Salgado R, Moore H, Martens JW, Lively T, Malik S, McDermott U, et al. . Steps forward for cancer precision medicine. Nat Rev Drug Discov. (2018) 17:1–2. 10.1038/nrd.2017.218
    1. Llombart-Cussac A, Cortés J, Paré L, Galván P, Bermejo B, Martínez N, et al. . HER2-enriched subtype as a predictor of pathological complete response following trastuzumab and lapatinib without chemotherapy in early-stage HER2-positive breast cancer (PAMELA): an open-label, single-group, multicentre, phase 2 trial. Lancet Oncol. (2017) 18:545–54. 10.1016/S1470-2045(17)30021-9
    1. Holmes FA, Nagarwala YM, Espina VA, Liotta LA, Danso MA, Gallagher RI, et al. . Correlation of molecular effects and pathologic complete response to preoperative lapatinib and trastuzumab, separately and combined prior to neoadjuvant breast cancer chemotherapy. J Clin Oncol. (2011) 29(Suppl. 15):506. 10.1200/jco.2011.29.15_suppl.506
    1. Baselga J, Bradbury I, Eidtmann H, Di Cosimo S, De Azambuja E, Aura C, et al. . Lapatinib with trastuzumab for HER2-positive early breast cancer (NeoALTTO): a randomised, open-label, multicentre, phase 3 trial. Lancet. (2012) 379:633–40. 10.1016/S0140-6736(11)61847-3
    1. Chic N, Luen S, Nuciforo P, et al. . Abstract PS5-03: Celtil score and long-term survival outcome in early stage HER2-positive (HER2+) breast cancer treated with anti-HER2-based chemotherapy: A correlative analysis of neoALTTO trial. Cancer Res. (2021) 81(4 Supplement):PS5–03.
    1. González-Farré B, Nuciforo P, Brunet LP, Cortés J, Cussac AL, Gregori JG, et al. . 15P The CelTIL score as an early predictor of anti-tumour response following neoadjuvant therapy (NAT): A SOLTI biomarker analysis. Ann Oncol. (2020) 31:S15–88. 10.1016/j.annonc.2020.03.151

Source: PubMed

3
Prenumerera