Early on-treatment transcriptional profiling as a tool for improving pathological response prediction in HER2-positive inflammatory breast cancer

Sonia Pernas, Jennifer L Guerriero, Sergey Naumenko, Shom Goel, Meredith M Regan, Jiani Hu, Beth T Harrison, Filipa Lynce, Nancy U Lin, Ann Partridge, Aki Morikawa, John Hutchinson, Elizabeth A Mittendorf, Artem Sokolov, Beth Overmoyer, Sonia Pernas, Jennifer L Guerriero, Sergey Naumenko, Shom Goel, Meredith M Regan, Jiani Hu, Beth T Harrison, Filipa Lynce, Nancy U Lin, Ann Partridge, Aki Morikawa, John Hutchinson, Elizabeth A Mittendorf, Artem Sokolov, Beth Overmoyer

Abstract

Background: Inflammatory breast cancer (IBC) is a rare and understudied disease, with 40% of cases presenting with human epidermal growth factor receptor 2 (HER2)-positive subtype. The goals of this study were to (i) assess the pathologic complete response (pCR) rate of short-term neoadjuvant dual-HER2-blockade and paclitaxel, (ii) contrast baseline and on-treatment transcriptional profiles of IBC tumor biopsies associated with pCR, and (iii) identify biological pathways that may explain the effect of neoadjuvant therapy on tumor response.

Patients and methods: A single-arm phase II trial of neoadjuvant trastuzumab (H), pertuzumab (P), and paclitaxel for 16 weeks was completed among patients with newly diagnosed HER2-positive IBC. Fresh-frozen tumor biopsies were obtained pretreatment (D1) and 8 days later (D8), following a single dose of HP, prior to adding paclitaxel. We performed RNA-sequencing on D1 and D8 tumor biopsies, identified genes associated with pCR using differential gene expression analysis, identified pathways associated with pCR using gene set enrichment and gene expression deconvolution methods, and compared the pCR predictive value of principal components derived from gene expression profiles by calculating and area under the curve for D1 and D8 subsets.

Results: Twenty-three participants were enrolled, of whom 21 completed surgery following neoadjuvant therapy. Paired longitudinal fresh-frozen tumor samples (D1 and D8) were obtained from all patients. Among the 21 patients who underwent surgery, the pCR and the 4-year disease-free survival were 48% (90% CI 0.29-0.67) and 90% (95% CI 66-97%), respectively. The transcriptional profile of D8 biopsies was found to be more predictive of pCR (AUC = 0.91, 95% CI: 0.7993-1) than the D1 biopsies (AUC = 0.79, 95% CI: 0.5905-0.9822).

Conclusions: In patients with HER2-positive IBC treated with neoadjuvant HP and paclitaxel for 16 weeks, gene expression patterns of tumor biopsies measured 1 week after treatment initiation not only offered different biological information but importantly served as a better predictor of pCR than baseline transcriptional analysis.

Trial registration: ClinicalTrials.gov identifier: NCT01796197 (https://ichgcp.net/clinical-trials-registry/NCT01796197); registered on February 21, 2013.

Keywords: HER2-positive; Inflammatory breast cancer; gene expression; immune response; on-treatment biopsy; treatment de-escalation.

Conflict of interest statement

Competing Interests: SP has served as an advisor/consultant for AstraZeneca, Daiichi Sankyo Eisai, Novartis, Polyphor, Roche, Pierre-Fabre, and SeattleGenetics. SG has received laboratory research funding from Eli Lilly and performs clinical research sponsored by Novartis and Eli Lilly. SG has served as a paid advisor to Eli Lilly, G1 therapeutics, and Novartis. JLG is a consultant for Glaxo-Smith Kline (GSK), Array BioPharma, Codagenix, Verseau Therapeutics, Kymera, Carisma, Kowa, Duke Street Bio and MPM Capital, and receives sponsored research support from GSK, Eli Lilly and Array BioPharma. NUL reports institutional research funding from Genentech, Merck, Pfizer, Seattle Genetics, AstraZeneca, Zion Pharmaceuticals, and Olema Pharmaceuticals; consultant/advisory board work for Pfizer, Puma, Seattle Genetics, Daiichi Sankyo, AstraZeneca, Prelude Therapeutics, Denali Therapeutics, Olema Pharmaceuticals, Aleta BioPharma, Affinia Therapeutics, Voyager Therapeutics, and Artera, Inc.; royalties from UpToDate; and stock or other ownership interests in Artera Inc. (a startup with no current value, but options only valued at <5% and <$50,000 will be provided at a later date). MR reports research funding (and/or provision of drug supply for clinical trials) from Novartis, Pfizer, Ipsen, TerSera, Merck, Ferring, Pierre Fabre, Roche, AstraZeneca, Bayer, Bristol-Myers Squibb; consulting or advisory role for Ipsen, Bristol-Myers Squibb, Tolmar Pharmaceuticals. BO has received clinical trial support from Genentech, Incyte, and Eisai. AM reports a consulting or advisory role for Lilly and Seagen, in-kind research support from Tempus, and institutional research funding from Eisai, Seagen, MTEM, Lilly, Seagen, Takeda Millennium, and Pfizer. EAM reports compensated service on scientific advisory boards for AstraZeneca, Exact Sciences, Merck, and Roche/Genentech; uncompensated service on steering committees for Bristol Myers Squibb, Lilly, and Roche/Genentech; honoraria from Physicians’ Education Resource; and institutional research support from Roche/Genentech (via SU2C grant) and Gilead. EAM reports the following nonfinancial interests, nonremunerated activities: Board of Directors for the American Society of Clinical Oncology and Scientific Advisor for Susan G. Komen for the Cure Foundation.

© The Author(s), 2022.

Figures

Figure 1.
Figure 1.
Scheme of prospective single-arm phase II clinical trial for newly diagnosed HER2-positive inflammatory breast cancer. AC, doxorubicin, cyclophosphamide; H, trastuzumab; pCR, pathologic complete response; RT, radiotherapy; wk, week.
Figure 2.
Figure 2.
Changes in TILs in tumor biopsy samples obtained pretreatment (D1) and post-dual HER2-blockade (D8). (a) Representative H&E images from frozen sections of selected cases with an increase, decrease, or no changes in TILs levels. (b) Percent of TILs by response at baseline (pretreatment, D1) and post-dual HER2 blockade (D8); *p = 0.0415. Unpaired one-tailed t-test. (c) Individual changes in TIL infiltration between baseline (pretreatment, D1) and post-dual HER2 blockade (D8) *p = 0.0333 and (d) according to hormonal receptor status and pathologic response by RCB (n = 21 independent paired patient samples). Two-way analysis of variance with Fisher’s Least Significant Difference (LSD) test. ER, estrogen receptor; pCR, pathologic complete response; TIL, tumor infiltrating lymphocyte.
Figure 3.
Figure 3.
The signal distinguishing responders and nonresponders is stronger in post-dual-HER2 blockade (D8) data. (a) Pairwise similarities between all 23 patients, computed using the top 1000 most variable genes. Individual cells of the heatmap show Pearson correlation values. Rows and columns are annotated with patient IDs. The columns are augmented with metadata describing the date of sequencing, tumor percentage, ER status, whether a patient achieved a pCR, and whether pretreatment (D1) or post-dual HER2 blockade (D8) data was used. (b) D1 and (c) D8 gene expression data projected onto the first two principal components, computed on the corresponding data slices. Individual points are colored by their response status [pCR versus residual disease (non-pCR)]. (d) ROC curves associated with the ability of the second principal component to distinguish pCR and non-pCR. ER, estrogen receptor; pCR, pathologic complete response.
Figure 4.
Figure 4.
Genes with the strongest signal distinguishing pCR from non-pCR. Differentially expressed genes in patients with pCR versus pCR in pretreatment (D1) data (a) and post-dual HER2 blockade (D8) data (b). Positive log2 fold change indicates higher expression in pCR samples. Genes corresponding to the False Discovery Rate below 0.01 are colored in cyan. pCR, pathologic complete response.
Figure 5.
Figure 5.
Evaluation of tumor cell intrinsic and immune related genes and signatures. (a) Hallmark gene sets enriched in tumors with pCR compared to non-pCR at D8 (post-dual HER2 blockade), with an adjusted p-value < 0.05. (b) Curated pathways and GO terms related to tumor cell intrinsic signaling, adaptative and innate immune response, and antigen presentation that are enriched in tumors with pCR compared to non-pCR at D8. All q values are below 0.05. (c–e) Raw expression of individual genes. Statistical analyses were performed using two-tailed unpaired t-test. Error bars represent ±SEM. *p < 0.05. **p < 0.01. ***p < 0.001. ****p < 0.0001. pCR, pathologic complete response.
Figure 6.
Figure 6.
Deconvolution of tumor infiltrating immune cells. FARDEEP was used to enumerate immune cell subsets from whole tumor tissue samples using the LM22 signature matrix. (a) Relative cell proportions and (b) absolute cell scores are shown. Distributions of several selected cell types are plotted by relative (c) and absolute (d) values. Statistical analyses were performed using two-tailed unpaired t-test. Error bars represent ±SEM. *p < 0.05. **p < 0.01. ****p < 0.0001.

References

    1. Dawood S, Lei X, Dent R, et al.. Survival of women with inflammatory breast cancer: a large population-based study. Ann Oncol 2014; 25: 1143–1151.
    1. Dawood S, Broglio K, Gong Y, et al.. Prognostic significance of HER-2 status in women with inflammatory breast cancer. Cancer 2008; 112: 1905–1911.
    1. Curigliano G. Inflammatory breast cancer and chest wall disease: the oncologist perspective. Eur J Surg Oncol 2018; 44: 1142–1147.
    1. Gianni L, Eiermann W, Semiglazov V, 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.
    1. Li J, Xia Y, Wu Q, et al.. Outcomes of patients with inflammatory breast cancer by hormone receptor- and HER2-defined molecular subtypes: a population-based study from the SEER program. Oncotarget 2017; 8: 49370–49379.
    1. Cortazar P, Zhang L, Untch M, et al.. Pathological complete response and long-term clinical benefit in breast cancer: the CTNeoBC pooled analysis. Lancet 2014; 384: 164–172.
    1. Gianni L, Pienkowski T, Im YH, et al.. 5-year analysis of neoadjuvant pertuzumab and trastuzumab in patients with locally advanced, inflammatory, or early-stage HER2-positive breast cancer (NeoSphere): a multicentre, open-label, phase 2 randomised trial. Lancet Oncol 2016; 17: 791–800.
    1. Pernas S, Tolaney SM. Management of early-stage human epidermal growth factor receptor 2-positive breast cancer. JCO Oncol Pract 2021; 17: 320–330.
    1. Byron SA, Van Keuren-Jensen KR, Engelthaler DM, et al.. Translating RNA sequencing into clinical diagnostics: opportunities and challenges. Nat Rev Genet 2016; 17: 257–271.
    1. Pernas S, Petit A, Climent F, et al.. PAM50 subtypes in baseline and residual tumors following neoadjuvant trastuzumab-based chemotherapy in HER2-positive breast cancer: a consecutive-series from a single institution. Front Oncol 2019; 9: 707.
    1. Fumagalli D, Venet D, Ignatiadis M, 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.
    1. Carey LA, Berry DA, Cirrincione CT, 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.
    1. Llombart-Cussac A, Cortes J, Pare L, 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.
    1. Wolff AC, Hammond MEH, Hicks DG, et al.. Recommendations for human epidermal growth factor receptor 2 testing in breast cancer: American Society of Clinical Oncology/College of American Pathologists Clinical Practice Guideline Update. J Clin Oncol 2013; 31: 3997–4013.
    1. Symmans WF, Peintinger F, Hatzis C, et al.. Measurement of residual breast cancer burden to predict survival after neoadjuvant chemotherapy. J Clin Oncol 2007; 25: 4414–4422.
    1. Salgado R, Denkert C, Demaria S, et al.. The evaluation of tumor-infiltrating lymphocytes (TILs) in breast cancer: recommendations by an International TILs Working Group 2014. Ann Oncol 2014; 26: 259–271.
    1. Nuciforo P, Pascual T, Cortes J, 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: 170–177.
    1. Chapman B, Kirchner R, Pantano L, et al.. Validated, scalable, community developed variant calling, RNA-seq and small RNA analysis, 10.5281/zenodo.3564938 (2021). (accessed July 10, 2022).
    1. Dobin A, Davis CA, Schlesinger F, et al.. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 2013; 29: 15–21.
    1. Li H, Handsaker B, Wysoker A, et al.. The sequence alignment/map format and SAMtools. Bioinformatics 2009; 25: 2078–2079.
    1. Ewels P, Magnusson M, Lundin S, et al.. MultiQC: summarize analysis results for multiple tools and samples in a single report. Bioinformatics 2016; 32: 3047–3048.
    1. Patro R, Duggal G, Love MI, et al.. Salmon provides fast and bias-aware quantification of transcript expression. Nat Methods 2017; 14: 417–419.
    1. Soneson C, Love MI, Robinson MD. Differential analyses for RNA-seq: transcript-level estimates improve gene-level inferences. F1000Res 2015; 4: 1521.
    1. Wickham H, Averick M, Bryan J, et al.. Welcome to the Tidyverse. J Open Source Softw 2019; 4: 1686.
    1. Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol 2014; 15: 550.
    1. Robin X, Turck N, Hainard A, et al.. PROC: an open-source package for R and S+ to analyze and compare ROC curves. BMC Bioinformatics 2011; 12: 77.
    1. Wickham H. ggplot2: elegant graphics for data analysis. 2nd ed. Cham: Springer International Publishing, 2016.
    1. Subramanian A, Tamayo P, Mootha VK, 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.
    1. Mootha VK, Lindgren CM, Eriksson KF, et al.. PGC-1alpha-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nat Genet 2003; 34: 267–273.
    1. Hao Y, Yan M, Heath BR, et al.. Fast and robust deconvolution of tumor infiltrating lymphocyte from expression profiles using least trimmed squares. PLoS Comput Biol 2019; 15: e1006976.
    1. Yu G, Wang LG, Han Y, et al.. ClusterProfiler: an R package for comparing biological themes among gene clusters. OMICS 2012; 16: 284–287.
    1. Atkinson EN, Brown BW. Confidence limits for probability of response in multistage phase II clinical trials. Biometrics 1985; 41: 741–744.
    1. Hortobagyi G, Connolly JL, D’Orsi C, et al.. AJCC cancer staging manual. 8th ed. Cham: Springer, 2017.
    1. Bertucci F, Boudin L, Finetti P, et al.. Immune landscape of inflammatory breast cancer suggests vulnerability to immune checkpoint inhibitors. Oncoimmunology 2021; 10: 1929724.
    1. Reddy SM, Reuben A, Barua S, et al.. Poor response to neoadjuvant chemotherapy correlates with mast cell infiltration in inflammatory breast cancer. Cancer Immunol Res 2019; 7: 1025–1035.
    1. Bertucci F, Finetti P, Colpaert C, et al.. PDL1 expression in inflammatory breast cancer is frequent and predicts for the pathological response to chemotherapy. Oncotarget 2015; 6: 13506–13519.
    1. Avila Cobos F, Alquicira-Hernandez J, Powell JE, et al.. Benchmarking of cell type deconvolution pipelines for transcriptomics data. Nat Commun 2020; 11: 5650.
    1. Biswas T, Jindal C, Fitzgerald TL, et al.. Pathologic complete response (pCR) and survival of women with inflammatory breast cancer (IBC): an analysis based on biologic subtypes and demographic characteristics. Int J Environ Res Public Health 2019; 16: 124.
    1. Gianni L, Pienkowski T, Im YH, et al.. Efficacy and safety of neoadjuvant pertuzumab and trastuzumab in women with locally advanced, inflammatory, or early HER2-positive breast cancer (NeoSphere): a randomised multicentre, open-label, phase 2 trial. Lancet Oncol 2012; 13: 25–32.
    1. Schneeweiss A, Chia S, Hickish T, et al.. Long-term efficacy analysis of the randomised, phase II TRYPHAENA cardiac safety study: evaluating pertuzumab and trastuzumab plus standard neoadjuvant anthracycline-containing and anthracycline-free chemotherapy regimens in patients with HER2-positive early breast cancer. Eur J Cancer 2018; 89: 27–35.
    1. Gianni L, Eiermann W, Semiglazov V, et al.. Neoadjuvant and adjuvant trastuzumab in patients with HER2-positive locally advanced breast cancer (NOAH): follow-up of a randomised controlled superiority trial with a parallel HER2-negative cohort. Lancet Oncol 2014; 15: 640–647.
    1. Lim B, Woodward WA, Wang X, et al.. Inflammatory breast cancer biology: the tumour microenvironment is key. Nat Rev Cancer 2018; 18: 485–499.
    1. Dowsett M, Smith IE, Ebbs SR, et al.. Prognostic value of Ki67 expression after short-term presurgical endocrine therapy for primary breast cancer. J Natl Cancer Inst 2007; 99: 167–170.
    1. Ellis MJ, Suman VJ, Hoog J, et al.. Ki67 proliferation index as a tool for chemotherapy decisions during and after neoadjuvant aromatase inhibitor treatment of breast cancer: results from the American College of Surgeons Oncology Group Z1031 Trial (Alliance). J Clin Oncol 2017; 35: 1061–1069.
    1. Korde LA, Lusa L, McShane L, et al.. Gene expression pathway analysis to predict response to neoadjuvant docetaxel and capecitabine for breast cancer. Breast Cancer Res Treat 2010; 119: 685–699.
    1. Magbanua MJ, Wolf DM, Yau C, et al.. Serial expression analysis of breast tumors during neoadjuvant chemotherapy reveals changes in cell cycle and immune pathways associated with recurrence and response. Breast Cancer Res 2015; 17: 73.
    1. Bownes RJ, Turnbull AK, Martinez-Perez C, et al.. On-treatment biomarkers can improve prediction of response to neoadjuvant chemotherapy in breast cancer. Breast Cancer Res 2019; 21: 73.
    1. Bertucci F, Ueno NT, Finetti P, et al.. Gene expression profiles of inflammatory breast cancer: correlation with response to neoadjuvant chemotherapy and metastasis-free survival. Ann Oncol 2014; 25: 358–365.
    1. Bianchini G, Pusztai L, Pienkowski T, et al.. Immune modulation of pathologic complete response after neoadjuvant HER2-directed therapies in the NeoSphere trial. Ann Oncol 2015; 26: 2429–2436.
    1. Van Berckelaer C, Rypens C, van Dam P, et al.. Infiltrating stromal immune cells in inflammatory breast cancer are associated with an improved outcome and increased PD-L1 expression. Breast Cancer Res 2019; 21: 28.
    1. Salgado R, Denkert C, Campbell C, 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.
    1. Denkert C, von Minckwitz G, Darb-Esfahani S, 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.

Source: PubMed

Подписаться