Pathologic and molecular responses to neoadjuvant trastuzumab and/or lapatinib from a phase II randomized trial in HER2-positive breast cancer (TRIO-US B07)

Sara A Hurvitz, Jennifer L Caswell-Jin, Katherine L McNamara, Jason J Zoeller, Gregory R Bean, Robert Dichmann, Alejandra Perez, Ravindranath Patel, Lee Zehngebot, Heather Allen, Linda Bosserman, Brian DiCarlo, April Kennedy, Armando Giuliano, Carmen Calfa, David Molthrop, Aruna Mani, Hsiao-Wang Chen, Judy Dering, Brad Adams, Eran Kotler, Michael F Press, Joan S Brugge, Christina Curtis, Dennis J Slamon, Sara A Hurvitz, Jennifer L Caswell-Jin, Katherine L McNamara, Jason J Zoeller, Gregory R Bean, Robert Dichmann, Alejandra Perez, Ravindranath Patel, Lee Zehngebot, Heather Allen, Linda Bosserman, Brian DiCarlo, April Kennedy, Armando Giuliano, Carmen Calfa, David Molthrop, Aruna Mani, Hsiao-Wang Chen, Judy Dering, Brad Adams, Eran Kotler, Michael F Press, Joan S Brugge, Christina Curtis, Dennis J Slamon

Abstract

In this multicenter, open-label, randomized phase II investigator-sponsored neoadjuvant trial with funding provided by Sanofi and GlaxoSmithKline (TRIO-US B07, Clinical Trials NCT00769470), participants with early-stage HER2-positive breast cancer (N = 128) were recruited from 13 United States oncology centers throughout the Translational Research in Oncology network. Participants were randomized to receive trastuzumab (T; N = 34), lapatinib (L; N = 36), or both (TL; N = 58) as HER2-targeted therapy, with each participant given one cycle of this designated anti-HER2 therapy alone followed by six cycles of standard combination chemotherapy with the same anti-HER2 therapy. The primary objective was to estimate the rate of pathologic complete response (pCR) at the time of surgery in each of the three arms. In the intent-to-treat population, we observed similar pCR rates between T (47%, 95% confidence interval [CI] 30-65%) and TL (52%, 95% CI 38-65%), and a lower pCR rate with L (25%, 95% CI 13-43%). In the T arm, 100% of participants completed all protocol-specified treatment prior to surgery, as compared to 69% in the L arm and 74% in the TL arm. Tumor or tumor bed tissue was collected whenever possible pre-treatment (N = 110), after one cycle of HER2-targeted therapy alone (N = 89), and at time of surgery (N = 59). Higher-level amplification of HER2 and hormone receptor (HR)-negative status were associated with a higher pCR rate. Large shifts in the tumor, immune, and stromal gene expression occurred after one cycle of HER2-targeted therapy. In contrast to pCR rates, the L-containing arms exhibited greater proliferation reduction than T at this timepoint. Immune expression signatures increased in all arms after one cycle of HER2-targeted therapy, decreasing again by the time of surgery. Our results inform approaches to early assessment of sensitivity to anti-HER2 therapy and shed light on the role of the immune microenvironment in response to HER2-targeted agents.

Conflict of interest statement

S.H. received contracted research and medical writing assistance from Ambrx, Amgen, Arvinas, Bayer, Daiichi-Sankyo, Genentech/Roche, GSK, Immunomedics, Lilly, Macrogenics, Novartis, Pfizer, OBI Pharma, Pieris, Puma, Radius, Sanofi, Seattle Genetics, and Dignitana. A.P. received institutional research support from Genentech, AstraZeneca, Immunomedics, Nektar, and Macrogenics. L.B. received honoraria from and/or was on the speaker bureau for AstraZeneca, Pfizer, Merck, Puma, Novartis, GlaxoSmithKline, Amgen, Johnson & Johnson, Genentech/Roche, Sandoz, Dendreon, and Genomic Health, and performed consulting work for Integra Connect and Anthem Blue Cross. A.M. is employed by Genentech/Roche and owns stock in Roche. M.P. received research support from Cepheid, was on the Scientific Advisory Board and received research support from Eli Lilly, Zymeworks, and Novartis, was a consultant for and received research support from Puma, and was on the Scientific Advisory Board for Biocartis. C.C. is a scientific advisor to GRAIL and reports stock options as well as consulting for GRAIL and Genentech. D.S. received research funding from Pfizer, Novartis, Syndax, Millenium Pharmaceuticals, Aileron Therapeutics, Bayer, and Genentech, owned stock in Biomarin, Amgen, Seattle Genetics, and Pfizer, served on the Board of Directors for BioMarin, and performed consulting/advisory board work for Eli Lilly, Novartis, Bayer, and Pfizer. The remaining authors declare no competing interests.

Figures

Fig. 1. TRIO-US B07 clinical trial participants.
Fig. 1. TRIO-US B07 clinical trial participants.
130 participants were enrolled across three treatment arms. All participants received docetaxel plus carboplatin (TC) every 3 weeks. In addition, participants in Arm 1 received trastuzumab (TCH), Arm 2 received lapatinib (TCL), and Arm 3 received both trastuzumab and lapatinib (TCHL). Two participants withdrew from the study prior to starting any treatment, leaving 128 participants remaining in the intent to treat (ITT) population. Of 128 participants, 25 came off study treatment prior to surgery (10 in Arm 2, 15 Arm 3), leaving 103 participants included in the evaluable analysis.
Fig. 2. Characteristics of the cohort prior…
Fig. 2. Characteristics of the cohort prior to treatment.
a Selected clinical and expression characteristics and tumor subtypes across the cohort. White squares reflect missing data. b Expression values of selected genes within the HER2 amplicon. c Pearson correlation coefficient matrix of key gene expression signatures. *P = 0.010 and **P = 0.012 for correlation with pCR (two-sided Wald test). d Distribution of ESR1 pathway gene expression scores (from ref. ) by HR subtype (top) and their correlation with immune scores (from ref. ) (bottom). FISH: fluorescent in situ hybridization; IHC: immunohistochemistry; TILs: tumor-infiltrating lymphocytes; HR: hormone receptor; pCR: pathologic complete response; IC; integrative subtype/integrative cluster.
Fig. 3. Tumor and microenvironmental changes on…
Fig. 3. Tumor and microenvironmental changes on short-term HER2-targeted therapy.
a Subtype classifications (intrinsic and integrative) pre-treatment and after 14–21 days of HER2-targeted therapy. b Normalized enrichment scores from gene set enrichment analysis (GSEA) representing the degree of change of each gene set after 14–21 days of HER2-targeted therapy. Dotted lines separate those with FDR < 0.1. Black gene sets are Hallmark Molecular Signatures and blue gene sets were curated. c Pearson correlation coefficient matrix of single-sample GSEA (ssGSEA) scores. Ordering of gene sets is based on hierarchical clustering. Gene sets without labeled source in parentheses are Hallmark Molecular Signatures. d ER IHC H-score pre-treatment and after 14–21 days of HER2-targeted therapy correlate (r = 0.83) (left), and few tumors shift their ER H-score substantially (right) with treatment. e Change in proliferation ssGSEA scores after 14–21 days of HER2-targeted therapy by treatment arm. The mean drop in proliferation was highest with combination therapy, followed by lapatinib therapy (two-sided t test p = 0.28 compared to combination therapy), followed by trastuzumab therapy (p = 0.0087 compared to combination therapy; p = 0.13 compared to lapatinib). Centerline is median, box limits are upper and lower quartiles, whiskers are 1.5x the interquartile range, and empty points are outliers. f Change in percentage of cells positive for Ki67 by immunohistochemistry, pre-treatment to after 14–21 days of HER2-targeted therapy, stratified by treatment arm. P-values are from two-sided paired t-tests of the log-transformed Ki67 values. HR: hormone receptor; ER: estrogen receptor; H-score: histochemical score.
Fig. 4. Microenvironment changes across HER2-targeted therapy…
Fig. 4. Microenvironment changes across HER2-targeted therapy and chemotherapy.
a Normalized enrichment scores from gene set enrichment analysis (GSEA) representing the degree of change of each gene set at the time of surgery compared to pre-treatment. Dotted lines separate those with FDR < 0.1. b Normalized enrichment scores at surgery compared to pre-treatment (y-axis) vs after 14–21 days of HER2-targeted therapy compared to pre-treatment (x-axis). Dotted lines separate those with FDR < 0.1. Immune and some stromal sets increase after 14–21 days of HER2-targeted therapy but are reduced at the time of surgery. c Change in stromal TILs and inflammatory cell percentage across treatment. P-values are from two-sided paired t-tests, comparing on-treatment to pre-treatment (N = 55 for stromal TIL and N = 94 for inflammatory cells) and surgery to pre-treatment (N = 14 for stromal TIL and N = 78 for inflammatory cells). d Proportion of immune infiltrate represented by each immune cell subtype, according to CIBERSORT (top) and immunoStates (bottom). Pre-treatment (N = 89) is matched to on-treatment (N = 89); surgery (N = 59) is a subset. TIL: tumor-infiltrating lymphocytes.

References

    1. Slamon DJ, et al. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N. Engl. J. Med. 2001;344:783–792. doi: 10.1056/NEJM200103153441101.
    1. Marty M, et al. Randomized phase II trial of the efficacy and safety of trastuzumab combined with docetaxel in patients with human epidermal growth factor receptor 2-positive metastatic breast cancer administered as first-line treatment: the M77001 study group. J. Clin. Oncol. 2005;23:4265–4274. doi: 10.1200/JCO.2005.04.173.
    1. Romond EH, et al. Trastuzumab plus adjuvant chemotherapy for operable HER2-positive breast cancer. N. Engl. J. Med. 2005;353:1673–1684. doi: 10.1056/NEJMoa052122.
    1. Piccart-Gebhart MJ, et al. Trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer. N. Engl. J. Med. 2005;353:1659–1672. doi: 10.1056/NEJMoa052306.
    1. Slamon D, et al. Adjuvant trastuzumab in HER2-positive breast cancer. N. Engl. J. Med. 2011;365:1273–1283. doi: 10.1056/NEJMoa0910383.
    1. Perez EA, et al. Trastuzumab plus adjuvant chemotherapy for human epidermal growth factor receptor 2-positive breast cancer: planned joint analysis of overall survival from NSABP B-31 and NCCTG N9831. J. Clin. Oncol.: Off. J. Am. Soc. Clin. Oncol. 2014;32:3744–3752. doi: 10.1200/JCO.2014.55.5730.
    1. Goldhirsch A, et al. 2 years versus 1 year of adjuvant trastuzumab for HER2-positive breast cancer (HERA): an open-label, randomised controlled trial. Lancet. 2013;382:1021–1028. doi: 10.1016/S0140-6736(13)61094-6.
    1. Piccart-Gebhart M, et al. The association between event-free survival and pathological complete response to neoadjuvant lapatinib, trastuzumab, or their combination in HER2-positive breast cancer. Survival follow-up analysis of the NeoALTTO study. Cancer Res. 2013;73:S1–S01.
    1. Cortazar P, et al. Pathological complete response and long-term clinical benefit in breast cancer: the CTNeoBC pooled analysis. Lancet. 2014;384:164–172. doi: 10.1016/S0140-6736(13)62422-8.
    1. Bardia A, Baselga J. Neoadjuvant therapy as a platform for drug development and approval in breast cancer. Clin. Cancer Res.: Off. J. Am. Assoc. Cancer Res. 2013;19:6360–6370. doi: 10.1158/1078-0432.CCR-13-0916.
    1. Buzdar AU, et al. Significantly higher pathologic complete remission rate after neoadjuvant therapy with trastuzumab, paclitaxel, and epirubicin chemotherapy: results of a randomized trial in human epidermal growth factor receptor 2-positive operable breast cancer. J. Clin. Oncol. 2005;23:3676–3685. doi: 10.1200/JCO.2005.07.032.
    1. Gianni L, et al. Neoadjuvant chemotherapy with trastuzumab followed by adjuvant trastuzumab versus neoadjuvant chemotherapy alone, in patients with HER2-positive locally advanced breast cancer (the NOAH trial): a randomised controlled superiority trial with a parallel HER2-negative cohort. Lancet. 2010;375:377–384. doi: 10.1016/S0140-6736(09)61964-4.
    1. Pierga JY, et al. A multicenter randomized phase II study of sequential epirubicin/cyclophosphamide followed by docetaxel with or without celecoxib or trastuzumab according to HER2 status, as primary chemotherapy for localized invasive breast cancer patients. Breast Cancer Res. Treat. 2010;122:429–437. doi: 10.1007/s10549-010-0939-3.
    1. Steger GG, et al. Epirubicin and docetaxel with or without capecitabine as neoadjuvant treatment for early breast cancer: final results of a randomized phase III study (ABCSG-24) Ann. Oncol. 2014;25:366–371. doi: 10.1093/annonc/mdt508.
    1. Konecny GE, et al. Activity of the dual kinase inhibitor lapatinib (GW572016) against HER-2-overexpressing and trastuzumab-treated breast cancer cells. Cancer Res. 2006;66:1630–1639. doi: 10.1158/0008-5472.CAN-05-1182.
    1. Xia W, et al. Combining lapatinib (GW572016), a small molecule inhibitor of ErbB1 and ErbB2 tyrosine kinases, with therapeutic anti-ErbB2 antibodies enhances apoptosis of ErbB2-overexpressing breast cancer cells. Oncogene. 2005;24:6213–6221. doi: 10.1038/sj.onc.1208774.
    1. Scaltriti M, et al. Lapatinib, a HER2 tyrosine kinase inhibitor, induces stabilization and accumulation of HER2 and potentiates trastuzumab-dependent cell cytotoxicity. Oncogene. 2009;28:803–814. doi: 10.1038/onc.2008.432.
    1. Blackwell KL, et al. Randomized study of Lapatinib alone or in combination with trastuzumab in women with ErbB2-positive, trastuzumab-refractory metastatic breast cancer. J. Clin. Oncol. 2010;28:1124–1130. doi: 10.1200/JCO.2008.21.4437.
    1. Blackwell KL, et al. Overall survival benefit with lapatinib in combination with trastuzumab for patients with human epidermal growth factor receptor 2-positive metastatic breast cancer: final results from the EGF104900 Study. J. Clin. Oncol.: Off. J. Am. Soc. Clin. Oncol. 2012;30:2585–2592. doi: 10.1200/JCO.2011.35.6725.
    1. Baselga J, et al. Lapatinib with trastuzumab for HER2-positive early breast cancer (NeoALTTO): a randomised, open-label, multicentre, phase 3 trial. Lancet. 2012;379:633–640. doi: 10.1016/S0140-6736(11)61847-3.
    1. Robidoux A, et al. Lapatinib as a component of neoadjuvant therapy for HER2-positive operable breast cancer (NSABP protocol B-41): an open-label, randomised phase 3 trial. Lancet Oncol. 2013;14:1183–1192. doi: 10.1016/S1470-2045(13)70411-X.
    1. Guarneri V, et al. Preoperative chemotherapy plus trastuzumab, lapatinib, or both in human epidermal growth factor receptor 2-positive operable breast cancer: results of the randomized phase II CHER-LOB study. J. Clin. Oncol.: Off. J. Am. Soc. Clin. Oncol. 2012;30:1989–1995. doi: 10.1200/JCO.2011.39.0823.
    1. Carey, L. A. et al. Clinical and translational results of CALGB 40601. J. Clin. Oncol.31, 500 (2013).
    1. Bonnefoi H, et al. Neoadjuvant treatment with docetaxel plus lapatinib, trastuzumab, or both followed by an anthracycline-based chemotherapy in HER2-positive breast cancer: results of the randomised phase II EORTC 10054 study. Ann. Oncol.: Off. J. Eur. Soc. Med. Oncol. 2015;26:325–332. doi: 10.1093/annonc/mdu551.
    1. Holmes FA, et al. Pathologic complete response after preoperative anti-HER2 therapy correlates with alterations in PTEN, FOXO, phosphorylated Stat5, and autophagy protein signaling. BMC Res Notes. 2013;6:507. doi: 10.1186/1756-0500-6-507.
    1. Fumagalli D, et al. RNA sequencing to predict response to neoadjuvant Anti-HER2 therapy: a secondary analysis of the NeoALTTO randomized clinical trial. JAMA Oncol. 2017;3:227–234. doi: 10.1001/jamaoncol.2016.3824.
    1. Carey LA, et al. Molecular heterogeneity and response to neoadjuvant human epidermal growth factor receptor 2 targeting in CALGB 40601, a randomized phase III trial of paclitaxel plus trastuzumab With Or without lapatinib. J. Clin. Oncol. 2016;34:542–549. doi: 10.1200/JCO.2015.62.1268.
    1. Veeraraghavan, J. et al. A combinatorial biomarker predicts pathologic complete response to neoadjuvant lapatinib and trastuzumab without chemotherapy in patients with HER2+ breast cancer. Ann. Oncol.30, 927–933 (2019).
    1. Dieci MV, et al. Integrated evaluation of PAM50 subtypes and immune modulation of pCR in HER2-positive breast cancer patients treated with chemotherapy and HER2-targeted agents in the CherLOB trial. Ann. Oncol.: Off. J. Eur. Soc. Med. Oncol. 2016;27:1867–1873. doi: 10.1093/annonc/mdw262.
    1. Llombart-Cussac A, 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;18:545–554. doi: 10.1016/S1470-2045(17)30021-9.
    1. Lesurf R, et al. Genomic characterization of HER2-positive breast cancer and response to neoadjuvant trastuzumab and chemotherapy-results from the ACOSOG Z1041 (Alliance) trial. Ann. Oncol.: Off. J. Eur. Soc. Med. Oncol. 2017;28:1070–1077. doi: 10.1093/annonc/mdx048.
    1. Salgado R, et al. Tumor-infiltrating lymphocytes and associations with pathological complete response and event-free survival in HER2-positive early-stage breast cancer treated with lapatinib and trastuzumab: a secondary analysis of the NeoALTTO trial. JAMA Oncol. 2015;1:448–454. doi: 10.1001/jamaoncol.2015.0830.
    1. Denkert C, et al. Tumor-infiltrating lymphocytes and response to neoadjuvant chemotherapy with or without carboplatin in human epidermal growth factor receptor 2-positive and triple-negative primary breast cancers. J. Clin. Oncol. 2015;33:983–991. doi: 10.1200/JCO.2014.58.1967.
    1. Tanioka M, et al. Integrated analysis of RNA and DNA from the phase III trial CALGB 40601 identifies predictors of response to trastuzumab-based neoadjuvant chemotherapy in HER2-positive breast cancer. Clin. Cancer Res. 2018;24:5292–5304. doi: 10.1158/1078-0432.CCR-17-3431.
    1. Braso-Maristany F, et al. Phenotypic changes of HER2-positive breast cancer during and after dual HER2 blockade. Nat. Commun. 2020;11:385. doi: 10.1038/s41467-019-14111-3.
    1. Curtis C, et al. The genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroups. Nature. 2012;486:346–352. doi: 10.1038/nature10983.
    1. Ali, H. R. et al. Genome-driven integrated classification of breast cancer validated in over 7,500 samples. Genome Biol.15, 431 (2014).
    1. Elloumi F, et al. Systematic bias in genomic classification due to contaminating non-neoplastic tissue in breast tumor samples. BMC Med. Genomics. 2011;4:54. doi: 10.1186/1755-8794-4-54.
    1. Barbie DA, et al. Systematic RNA interference reveals that oncogenic KRAS-driven cancers require TBK1. Nature. 2009;462:108–112. doi: 10.1038/nature08460.
    1. Desmedt C, et al. Biological processes associated with breast cancer clinical outcome depend on the molecular subtypes. Clin. Cancer Res. 2008;14:5158–5165. doi: 10.1158/1078-0432.CCR-07-4756.
    1. Nielsen TO, et al. A comparison of PAM50 intrinsic subtyping with immunohistochemistry and clinical prognostic factors in tamoxifen-treated estrogen receptor-positive breast cancer. Clin. Cancer Res. 2010;16:5222–5232. doi: 10.1158/1078-0432.CCR-10-1282.
    1. Yoshihara K, et al. Inferring tumour purity and stromal and immune cell admixture from expression data. Nat. Commun. 2013;4:2612. doi: 10.1038/ncomms3612.
    1. Socinski MA, et al. Atezolizumab for first-line treatment of metastatic nonsquamous NSCLC. N. Engl. J. Med. 2018;378:2288–2301. doi: 10.1056/NEJMoa1716948.
    1. Krop IE, et al. Genomic correlates of response to adjuvant trastuzumab (H) and pertuzumab (P) in HER2+ breast cancer (BC): Biomarker analysis of the APHINITY trial. J. Clin. Oncol. 2019;37:1012–1012. doi: 10.1200/JCO.2019.37.15_suppl.1012.
    1. Perez EA, et al. Genomic analysis reveals that immune function genes are strongly linked to clinical outcome in the North Central Cancer Treatment Group n9831 Adjuvant Trastuzumab Trial. J. Clin. Oncol. 2015;33:701–708. doi: 10.1200/JCO.2014.57.6298.
    1. Farmer P, et al. A stroma-related gene signature predicts resistance to neoadjuvant chemotherapy in breast cancer. Nat. Med. 2009;15:68–74. doi: 10.1038/nm.1908.
    1. Liberzon A, et al. The Molecular Signatures Database (MSigDB) hallmark gene set collection. Cell Syst. 2015;1:417–425. doi: 10.1016/j.cels.2015.12.004.
    1. Bianchini G, Gianni L. The immune system and response to HER2-targeted treatment in breast cancer. Lancet Oncol. 2014;15:e58–e68. doi: 10.1016/S1470-2045(13)70477-7.
    1. Newman AM, et al. Robust enumeration of cell subsets from tissue expression profiles. Nat. methods. 2015;12:453–457. doi: 10.1038/nmeth.3337.
    1. Vallania F, et al. Leveraging heterogeneity across multiple datasets increases cell-mixture deconvolution accuracy and reduces biological and technical biases. Nat. Commun. 2018;9:4735. doi: 10.1038/s41467-018-07242-6.
    1. Subramanian A, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA. 2005;102:15545–15550. doi: 10.1073/pnas.0506580102.
    1. McNamara, K. L. et al. Spatial proteomic characterization of HER2-positive breast tumors through neoadjuvant therapy predicts response. Preprint available at 10.1101/2020.09.23.20199091 (2020).
    1. Varadan V, et al. Immune signatures following single dose trastuzumab predict pathologic response to preoperative trastuzumab and chemotherapy in HER2-positive early breast cancer. Clin. Cancer Res. 2016;22:3249–3259. doi: 10.1158/1078-0432.CCR-15-2021.
    1. Nuciforo P, 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.: Off. J. Eur. Soc. Med. Oncol. 2018;29:170–177. doi: 10.1093/annonc/mdx647.
    1. Gelmon KA, et al. Lapatinib or trastuzumab plus taxane therapy for human epidermal growth factor receptor 2-positive advanced breast cancer: final results of NCIC CTG MA.31. J. Clin. Oncol. 2015;33:1574–1583. doi: 10.1200/JCO.2014.56.9590.
    1. Untch M, et al. Lapatinib versus trastuzumab in combination with neoadjuvant anthracycline-taxane-based chemotherapy (GeparQuinto, GBG 44): a randomised phase 3 trial. Lancet Oncol. 2012;13:135–144. doi: 10.1016/S1470-2045(11)70397-7.
    1. Dowsett M, et al. Disease-free survival according to degree of HER2 amplification for patients treated with adjuvant chemotherapy with or without 1 year of trastuzumab: the HERA Trial. J. Clin. Oncol. 2009;27:2962–2969. doi: 10.1200/JCO.2008.19.7939.
    1. Perez EA, et al. HER2 and chromosome 17 effect on patient outcome in the N9831 adjuvant trastuzumab trial. J. Clin. Oncol. 2010;28:4307–4315. doi: 10.1200/JCO.2009.26.2154.
    1. Leary A, et al. Antiproliferative effect of lapatinib in HER2-positive and HER2-negative/HER3-high breast cancer: results of the presurgical randomized MAPLE trial (CRUK E/06/039) Clin. Cancer Res. 2015;21:2932–2940. doi: 10.1158/1078-0432.CCR-14-1428.
    1. Schmid P, et al. Phase II randomized preoperative window-of-opportunity study of the PI3K inhibitor pictilisib plus anastrozole compared with anastrozole alone in patients with estrogen receptor-positive breast cancer. J. Clin. Oncol. 2016;34:1987–1994. doi: 10.1200/JCO.2015.63.9179.
    1. Baselga J, et al. Phase II randomized study of neoadjuvant everolimus plus letrozole compared with placebo plus letrozole in patients with estrogen receptor-positive breast cancer. J. Clin. Oncol. 2009;27:2630–2637. doi: 10.1200/JCO.2008.18.8391.
    1. Johnston S, et al. Randomized phase II study evaluating palbociclib in addition to letrozole as neoadjuvant therapy in estrogen receptor-positive early breast cancer: PALLET trial. J. Clin. Oncol. 2019;37:178–189. doi: 10.1200/JCO.18.01624.
    1. Zervantonakis IK, et al. Fibroblast-tumor cell signaling limits HER2 kinase therapy response via activation of MTOR and antiapoptotic pathways. Proc. Natl Acad. Sci. USA. 2020;117:16500–16508. doi: 10.1073/pnas.2000648117.
    1. Jeselsohn RM, et al. Digital quantification of gene expression in sequential breast cancer biopsies reveals activation of an immune response. PLoS ONE. 2013;8:e64225. doi: 10.1371/journal.pone.0064225.
    1. Aran D, et al. Widespread parainflammation in human cancer. Genome Biol. 2016;17:145. doi: 10.1186/s13059-016-0995-z.
    1. Waks AG, et al. The immune microenvironment in hormone receptor-positive breast cancer before and after preoperative chemotherapy. Clin. Cancer Res. 2019;25:4644–4655. doi: 10.1158/1078-0432.CCR-19-0173.
    1. Coudert BP, et al. Pre-operative systemic (neo-adjuvant) therapy with trastuzumab and docetaxel for HER2-overexpressing stage II or III breast cancer: results of a multicenter phase II trial. Ann. Oncol. 2006;17:409–414. doi: 10.1093/annonc/mdj096.
    1. Chang HR. A phase II study of neoadjuvant docetaxel/carboplatin with or without trastuzumab in locally advanced breast cancer: response and cardiotoxicity. J. Clin. Oncol. 2006;24:10515. doi: 10.1200/jco.2006.24.18_suppl.10515.
    1. Coudert BP, et al. Multicenter phase II trial of neoadjuvant therapy with trastuzumab, docetaxel, and carboplatin for human epidermal growth factor receptor-2-overexpressing stage II or III breast cancer: results of the GETN(A)-1 trial. J. Clin. Oncol.: Off. J. Am. Soc. Clin. Oncol. 2007;25:2678–2684. doi: 10.1200/JCO.2006.09.9994.
    1. Chow, S.-C. C., et al. Sample Size Calculations in Clinical Research. 2nd edn (Chapman & Hall/CRC, Boca Raton, 2008).
    1. Fleiss, J. L. (ed.) in Statistical Methods for Rates and Proportions 2nd edn 24–26 (Wiley, New Jersey, 1981).
    1. Salgado R, et al. The evaluation of tumor-infiltrating lymphocytes (TILs) in breast cancer: recommendations by an International TILs Working Group 2014. Ann. Oncol.: Off. J. Eur. Soc. Med. Oncol. 2015;26:259–271. doi: 10.1093/annonc/mdu450.
    1. Zoeller JJ, Bronson RT, Selfors LM, Mills GB, Brugge JS. Niche-localized tumor cells are protected from HER2-targeted therapy via upregulation of an anti-apoptotic program in vivo. NPJ Breast Cancer. 2017;3:18. doi: 10.1038/s41523-017-0020-z.
    1. Ritchie ME, et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015;43:e47. doi: 10.1093/nar/gkv007.
    1. Ritchie ME, et al. A comparison of background correction methods for two-colour microarrays. Bioinformatics. 2007;23:2700–2707. doi: 10.1093/bioinformatics/btm412.
    1. Johnson WE, Li C, Rabinovic A. Adjusting batch effects in microarray expression data using empirical Bayes methods. Biostatistics. 2007;8:118–127. doi: 10.1093/biostatistics/kxj037.
    1. Paquet ER, Hallett MT. Absolute assignment of breast cancer intrinsic molecular subtype. J. Natl. Cancer Inst. 2015;107:357. doi: 10.1093/jnci/dju357.

Source: PubMed

3
S'abonner