Neoadjuvant Chemotherapy and Immunotherapy in Luminal B-like Breast Cancer: Results of the Phase II GIADA Trial

Maria Vittoria Dieci, Valentina Guarneri, Anna Tosi, Giancarlo Bisagni, Antonino Musolino, Simon Spazzapan, Gabriella Moretti, Grazia Maria Vernaci, Gaia Griguolo, Tommaso Giarratano, Loredana Urso, Francesca Schiavi, Claudia Pinato, Giovanna Magni, Marcello Lo Mele, Gian Luca De Salvo, Antonio Rosato, Pierfranco Conte, Maria Vittoria Dieci, Valentina Guarneri, Anna Tosi, Giancarlo Bisagni, Antonino Musolino, Simon Spazzapan, Gabriella Moretti, Grazia Maria Vernaci, Gaia Griguolo, Tommaso Giarratano, Loredana Urso, Francesca Schiavi, Claudia Pinato, Giovanna Magni, Marcello Lo Mele, Gian Luca De Salvo, Antonio Rosato, Pierfranco Conte

Abstract

Purpose: The role of immunotherapy in hormone receptor (HR)-positive, HER2-negative breast cancer is underexplored.

Patients and methods: The neoadjuvant phase II GIADA trial (NCT04659551, EUDRACT 2016-004665-10) enrolled stage II-IIIA premenopausal patients with Luminal B (LumB)-like breast cancer (HR-positive/HER2-negative, Ki67 ≥ 20%, and/or histologic grade 3). Patients received: three 21-day cycles of epirubicin/cyclophosphamide followed by eight 14-day cycles of nivolumab, triptorelin started concomitantly to chemotherapy, and exemestane started concomitantly to nivolumab. Primary endpoint was pathologic complete response (pCR; ypT0/is, ypN0).

Results: A pCR was achieved by 7/43 patients [16.3%; 95% confidence interval (CI), 7.4-34.9]; the rate of residual cancer burden class 0-I was 25.6%. pCR rate was significantly higher for patients with PAM50 Basal breast cancer (4/8, 50%) as compared with other subtypes (LumA 9.1%; LumB 8.3%; P = 0.017). Tumor-infiltrating lymphocytes (TIL), immune-related gene-expression signatures, and specific immune cell subpopulations by multiplex immunofluorescence were significantly associated with pCR. A combined score of Basal subtype and TILs had an AUC of 0.95 (95% CI, 0.89-1.00) for pCR prediction. According to multiplex immunofluorescence, a switch to a more immune-activated tumor microenvironment occurred following exposure to anthracyclines. Most common grade ≥3 treatment-related adverse events (AE) during nivolumab were γ-glutamyltransferase (16.7%), alanine aminotransferase (16.7%), and aspartate aminotransferase (9.5%) increase. Most common immune-related AEs were endocrinopathies (all grades 1-2; including adrenal insufficiency, n = 1).

Conclusions: Luminal B-like breast cancers with a Basal molecular subtype and/or a state of immune activation may respond to sequential anthracyclines and anti-PD-1. Our data generate hypotheses that, if validated, could guide immunotherapy development in this context.

©2021 The Authors; Published by the American Association for Cancer Research.

Figures

Figure 1.
Figure 1.
CONSORT diagram.
Figure 2.
Figure 2.
Association of gene-expression parameters and TILs with pCR. A, Rate of pCR within different molecular intrinsic subtypes. B, Volcano plot showing differential expression of baseline gene-expression signatures assessed with the NanoString Breast Cancer 360 Panel in tumors achieving pCR as compared with tumors not achieving pCR. Displayed is the log2 fold difference in each gene-expression signature score between pCR and non-pCR. The gray dashed lines indicate the thresholds for unadjusted statistical significance (P < 0.05 and P < 0.01). Every dot represents one gene-expression signature. Blue dots represent gene signatures showing an association with pCR at the level of adjusted P < 0.05. C, Boxplot showing TIL levels in pCR vs. non-pCR patients. D, ROC showing the performance of the combined Basal subtype and TILs to predict pCR.
Figure 3.
Figure 3.
A, Boxplots showing intratumoral immune cell populations by mIHC with significant different levels in pCR vs. non-pCR patients at t0. B, Representative pictures of mIHC staining at t0 in pCR and non pCR patients; original magnification 20×. C, Significant changes in TILs and intratumoral immune cell populations by mIHC from t0 to t1 in matched paired samples, with * indicating outlier values, red lines for patients with pCR, yellow lines for patients with RCB I, and gray lines for other patients. D, Representative pictures of a lymphoid mIHC panel at t0 and t1 from the same patient; original magnification 20×. Color code for mIHC pictures of checkpoint panel: CD3 in magenta, PD-1 in yellow, CD68 in white, CD163 in green, PD-L1 in red, pan-cytokeratin in cyan, nuclei in blue. Color code for mIHC pictures of lymphoid panel: CD8 in magenta, CD4 in yellow, granzyme B in green, FoxP3 in white, CD20 in orange, pan-cytokeratin in cyan, nuclei in blue.

References

    1. Viale G, Hanlon Newell AE, Walker E, Harlow G, Bai I, Russo L, et al. . Ki-67 (30-9) scoring and differentiation of Luminal A- and Luminal B-like breast cancer subtypes. Breast Cancer Res Treat 2019;178:451–8.
    1. Cardoso F, Kyriakides S, Ohno S, Penault-Llorca F, Poortmans P, Rubio IT, et al. . Early breast cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 2019;30:1194–220.
    1. Cortazar P, Zhang L, Untch M, Mehta K, Costantino JP, Wolmark N, et al. . Pathological complete response and long-term clinical benefit in breast cancer: the CTNeoBC pooled analysis. Lancet 2014;384:164–72.
    1. von Minckwitz G, Untch M, Blohmer J-U, Costa SD, Eidtmann H, Fasching PA, et al. . Definition and impact of pathologic complete response on prognosis after neoadjuvant chemotherapy in various intrinsic breast cancer subtypes. J Clin Oncol 2012;30:1796–804.
    1. Gianni L, Mansutti M, Anton A, Calvo L, Bisagni G, Bermejo B, et al. . Comparing neoadjuvant nab-paclitaxel vs paclitaxel both followed by anthracycline regimens in women with ERBB2/HER2-negative breast cancer—the evaluating treatment with neoadjuvant abraxane (ETNA) trial: a randomized phase 3 clinical trial. JAMA Oncol 2018;4:302–8.
    1. Schmid P, Rugo HS, Adams S, Schneeweiss A, Barrios CH, Iwata H, et al. . Atezolizumab plus nab-paclitaxel as first-line treatment for unresectable, locally advanced or metastatic triple-negative breast cancer (IMpassion130): updated efficacy results from a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol 2020;21:44–59.
    1. Schmid P, Cortes J, Pusztai L, McArthur H, Kümmel S, Bergh J, et al. . Pembrolizumab for early triple-negative breast cancer. N Engl J Med 2020;382:810–21.
    1. Mittendorf EA, Zhang H, Barrios CH, Saji S, Jung KH, Hegg R, et al. . Neoadjuvant atezolizumab in combination with sequential nab-paclitaxel and anthracycline-based chemotherapy versus placebo and chemotherapy in patients with early- stage triple-negative breast cancer (IMpassion031): a randomised, double-blind, phase 3 trial. Lancet 2020;396:1090–100.
    1. Dieci MV, Griguolo G, Miglietta F, Guarneri V. The immune system and hormone-receptor positive breast cancer: is it really a dead end? Cancer Treat Rev 2016;46:9–19.
    1. Criscitiello C, Vingiani A, Maisonneuve P, Viale G, Viale G, Curigliano G. Tumor-infiltrating lymphocytes (TILs) in ER+/HER2− breast cancer. Breast Cancer Res Treat 2020;183:347–54.
    1. Luen S, Virassamy B, Savas P, Salgado R, Loi S. The genomic landscape of breast cancer and its interaction with host immunity. Breast 2016;29:241–50.
    1. Anurag M, Zhu M, Huang C, Vasaikar S, Wang J, Hoog J, et al. . Immune checkpoint profiles in luminal B breast cancer (alliance). J Natl Cancer Inst 2020;112:737–46.
    1. Voorwerk L, Slagter M, Horlings HM, Sikorska K, van de Vijver KK, de Maaker M, et al. . Immune induction strategies in metastatic triple-negative breast cancer to enhance the sensitivity to PD-1 blockade: the TONIC trial. Nat Med 2019;25:920–8.
    1. Apetoh L, Ghiringhelli F, Tesniere A, Obeid M, Ortiz C, Criollo A, et al. . Toll-like receptor 4-dependent contribution of the immune system to anticancer chemotherapy and radiotherapy. Nat Med 2007;13:1050–9.
    1. Generali D, Bates G, Berruti A, Brizzi MP, Campo L, Bonardi S, et al. . Immunomodulation of FOXP3+ regulatory T cells by the aromatase inhibitor letrozole in breast cancer patients. Clin Cancer Res 2009;15:1046–51.
    1. Huang H, Zhou J, Chen H, Li J, Zhang C, Jiang X, et al. . The immunomodulatory effects of endocrine therapy in breast cancer. J Exp Clin Cancer Res 2021;40:19.
    1. Parker JS, Mullins M, Cheang MCU, Leung S, Voduc D, Vickery T, et al. . Supervised risk predictor of breast cancer based on intrinsic subtypes. J Clin Oncol 2009;27:1160–7.
    1. Salgado R, Denkert C, Demaria S, Sirtaine N, Klauschen F, Pruneri G, et al. . The evaluation of tumor-infiltrating lymphocytes (TILs) in breast cancer: recommendations by an International TILs Working Group 2014. Ann Oncol 2015;26:259–71.
    1. Bender R, Lange S. Adjusting for multiple testing—when and how? J Clin Epidemiol 2001;54:343–9.
    1. Core Team R. R: a language and environment for statistical computing. 2017[cited 2020 Oct 19]. Available at: .
    1. Symmans WF, Wei C, Gould R, Yu X, Zhang Y, Liu M, et al. . Long-term prognostic risk after neoadjuvant chemotherapy associated with residual cancer burden and breast cancer subtype. J Clin Oncol 2017;35:1049–60.
    1. Nanda R, Liu MC, Yau C, Shatsky R, Pusztai L, Wallace A, et al. . Effect of pembrolizumab plus neoadjuvant chemotherapy on pathologic complete response in women with early-stage breast cancer: an analysis of the ongoing phase 2 adaptively randomized I-SPY2 trial. JAMA Oncol 2020;6:676–84.
    1. Pusztai L, Yau C, Wolf DM, Han HS, Du L, Wallace AM, et al. . Durvalumab with olaparib and paclitaxel for high-risk HER2-negative stage II/III breast cancer: results from the adaptively randomized I-SPY2 trial. Cancer Cell 2021;39:989–98.
    1. Prat A, Saura C, Pascual T, Hernando C, Muñoz M, Paré L, et al. . Ribociclib plus letrozole versus chemotherapy for postmenopausal women with hormone receptor-positive, HER2-negative, luminal B breast cancer (CORALLEEN): an open-label, multicentre, randomised, phase 2 trial. Lancet Oncol 2020;21:33–43.
    1. Cottu P, D'Hondt V, Dureau S, Lerebours F, Desmoulins I, Heudel P-E, et al. . Letrozole and palbociclib versus chemotherapy as neoadjuvant therapy of high-risk luminal breast cancer. Ann Oncol 2018;29:2334–40.
    1. Yau C, Van Der Noordaa M, Wei J, Osdoit M, Reyal F, Hamy AS, et al. . Abstract GS5–01: residual cancer burden after neoadjuvant therapy and long-term survival outcomes in breast cancer: a multi-center pooled analysis. Cancer Res 2020;80:GS5–01.
    1. Esserman LJ, Berry DA, DeMichele A, Carey L, Davis SE, Buxton M, et al. . Pathologic complete response predicts recurrence-free survival more effectively by cancer subset: results from the I-SPY 1 TRIAL–CALGB 150007/150012, ACRIN 6657. J Clin Oncol 2012;30:3242–9.
    1. Francis PA, Pagani O, Fleming GF, Walley BA, Colleoni M, Láng I, et al. . Tailoring adjuvant endocrine therapy for premenopausal breast cancer. N Engl J Med 2018;379:122–37.
    1. Cejalvo JM, Pascual T, Fernández-Martínez A, Brasó-Maristany F, Gomis RR, Perou CM, et al. . Clinical implications of the non-luminal intrinsic subtypes in hormone receptor-positive breast cancer. Cancer Treat Rev 2018;67:63–70.
    1. Prat A, Fan C, Fernández A, Hoadley KA, Martinello R, Vidal M, et al. . Response and survival of breast cancer intrinsic subtypes following multi-agent neoadjuvant chemotherapy. BMC Med 2015;13:303.
    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.

Source: PubMed

3
Prenumerera