Improved Natural Killer cell activity and retained anti-tumor CD8(+) T cell responses contribute to the induction of a pathological complete response in HER2-positive breast cancer patients undergoing neoadjuvant chemotherapy

E Muraro, E Comaro, R Talamini, E Turchet, G Miolo, S Scalone, L Militello, D Lombardi, S Spazzapan, T Perin, S Massarut, D Crivellari, Riccardo Dolcetti, D Martorelli, E Muraro, E Comaro, R Talamini, E Turchet, G Miolo, S Scalone, L Militello, D Lombardi, S Spazzapan, T Perin, S Massarut, D Crivellari, Riccardo Dolcetti, D Martorelli

Abstract

Background: Locally advanced HER2-overexpressing breast cancer (BC) patients achieve a high rate of pathological complete responses (pCR) after neoadjuvant chemotherapy (NC). The apparently unaltered immune proficiency of these patients together with the immune-modulating activities of NC drugs suggest a potential contribution of host immunity in mediating clinical responses. We thus performed an extensive immunomonitoring in locally advanced BC patients undergoing NC to identify immunological correlates of pCR induction.

Methods: The immune profile of 40 HER2-positive and 38 HER2-negative BC patients was characterized at diagnosis and throughout NC (Paclitaxel and Trastuzumab, or Docetaxel and Epirubicin, respectively). The percentages of circulating immune cell subsets including T and B lymphocytes, Natural Killer (NK) cells, regulatory T cells, T helper 17 lymphocytes, were quantified by multiparametric flow cytometry. NK cells functional activity was evaluated through the analysis of NF-kB nuclear translocation by Multispectral flow cytometry, and with the in vitro monitoring of Trastuzumab-mediated antibody-dependent cell cytotoxicity (ADCC). CD8(+) T cell responses against six different tumor-associated antigens (TAA) were characterized by IFN-γ ELISPOT and IFN-γ/IL-2 DualSpot assays.

Results: After NC, HER2-positive patients showed a significant increase in the number of NK cells and regulatory T cells irrespective of the pathological response, whereas patients undergoing a pCR disclosed higher percentages of T helper 17 cells. Notably, a significant increase in the number of activated NK cells was observed only in HER2-positive patients achieving a pCR. Characterization of anti-tumor T cell responses highlighted sustained levels of CD8(+) T cells specific for survivin and mammaglobin-A throughout NC in patients undergoing a pCR in both arms. Moreover, HER2-positive patients achieving a pCR were characterized by a multi-epitopic and polyfunctional anti-tumor T cell response, markedly reduced in case of partial response.

Conclusions: These results indicate that maintenance of functional T cell responses against selected antigens and improvement of NK cell proficiency during NC are probably critical requirements for pCR induction, especially in HER2-positive BC patients. Trail registration:

Trial registration number: NCT02307227, registered on ClinicalTrials.gov ( http://www.clinicaltrials.gov , November 26, 2014).

Figures

Figure 1
Figure 1
Phenotyping of circulating immune cells in HER2-negative and HER2-positive BC patients during NC. a Comparison of the percentage of several immune cell subsets quantified in the peripheral blood of HER-negative (white plots; n = 33) and HER2-positive (black plots; n = 23) BC patients throughout NC. *p < 0.05 comparing HER2-positive with HER2-negative patients. b Analysis of immune cell percentages within HER2-negative patients comparing individuals achieving a pathological complete response (pCR; black plots; n = 4) with those characterized by a pathological partial response (pPR; white plots; n = 29). *p < 0.05 comparing pCR with pPR patients. c Immune cell percentages observed in HER2-positive patients undergoing a pCR (black plots; n = 11) and in HER2-positive patients showing a pPR (white plots; n = 12). *p < 0.05 comparing pCR with pPR patients. NK Natural Killer cells, Th17 T helper 17 cells, Treg regulatory T cells, W week, pCR pathological complete response, pPR pathological partial response; **p < 0.05 in respect to the corresponding diagnosis values.
Figure 2
Figure 2
NF-kB nuclear translocation in NK cells of HER2-positive and HER2-negative patients throughout NC. a Quantification of the nuclear translocation of NF-kB in NK cells of HER2-positive (HER2+; black dots; n = 12) and HER2-negative patients (HER2−; white dots; n = 10) at diagnosis and during NC, and in healthy donors (DONORS; gray dots; n = 10). b Analysis performed within HER2-positive patients comparing NF-kB nuclear translocation observed in NK cells of patients undergoing a pathological complete response (pCR, black dots; n = 5) with that measured in case of partial response (pPR, white dots; n = 7) throughout NC, and with levels found in healthy donors (gray dots; n = 10). *p < 0.05 comparing values observed in case of complete response with those measured in partial responders. c Comparison of NF-kB nuclear translocation observed in the NK cells of HER2-negative patients achieving a pathological Complete response (pCR, black dots; n = 3) with levels measured in NK cells of HER2-negative partial responders (pPR, white dots; n = 7) during NC, and with the amount noticed in NK cells from healthy donors (gray dots; n = 10). d Representative plots obtained by Multispectral Flow Cytometry showing typical histograms of NK cells without (upper plot) or with (lower plot) the nuclear translocation of NF-kB. Representative cell pictures are shown. W week, pCR pathological complete response, pPR pathological partial response; **p < 0.05 in respect to the corresponding diagnosis values.
Figure 3
Figure 3
CD8+ T cell responses to BC-associated epitopes assessed by IFN-γ-ELISPOT in BC patients throughout NC. a Quantification of CD8+ T cell responses against 13 HLA-A*0201-restricted epitopes derived from 6 different BC-associated antigens (or against the flu matrix protein1-derived epitope) at diagnosis and during NC (12° and 24° week) in HER2-negative patients achieving a complete response (pCR, black dots; n = 2) and in those characterized by a partial response (pPR, white dots; n = 5). PHA-treated T cells and empty monocytes (EMPTY MONO) were used as positive and negative controls, respectively. b Analysis of CD8+ T cell responses against TAA-derived epitopes in HER2-positive patients reaching a complete response (pCR, black dots; n = 4) and a partial response (pPR, white dots; n = 2). SFC Spot Forming Cells, pCR pathological complete response, pPR pathological partial response, PHA phytohemagglutinin, Flu influenza, Mam-A Mammaglobin-A, Sur survivin, Muc mucin-1, Trag3 taxol resistance associated gene 3, W week.
Figure 4
Figure 4
Polyfunctional CD8+ T cell responses to BC-associated antigens assessed by DualSpot in HER2-positive patients during NC. a Representative histograms of the quantification of IFN-γ (gray bars), IL-2 (black bars), and IFN-γ/IL-2 (white bars) releasing CD8+ T cells after stimulation with four epitopes derived from four different BC-associated antigens obtained at diagnosis and during NC in one HER2-positive patient achieving a pathological complete response (BC87, pCR) and in one HER2-positive patient characterized by a partial response (BC105, pPR). Medium and PBMCs alone were used as negative controls, CEF and α–CD3 as positive controls. b Slices of cakes represent the means of the percentages of CD8+ T cells releasing IFN-γ (gray), IL-2 (black), or IFN-γ/IL-2 (white) after stimulation with all the four epitopes derived from BC-associated antigens in case of complete response (left) or partial response (right). SFC spot forming cells, pCR pathological complete response, pPR pathological partial response, W week, Sur survivin, Mam mammaglobin-A, CEF mix of peptides derived from Cytomegalovirus, Epstein-Barr virus, and influenza.

References

    1. Houssami N, Macaskill P, von Minckwitz G, Marinovich ML, Mamounas E. Meta-analysis of the association of breast cancer subtype and pathologic complete response to neoadjuvant chemotherapy. Eur J Cancer. 2012;48(18):3342–3354. doi: 10.1016/j.ejca.2012.05.023.
    1. Bardia A, Baselga J. Neoadjuvant therapy as a platform for drug development and approval in breast cancer. Clin Cancer Res. 2013;19(23):6360–6370. doi: 10.1158/1078-0432.CCR-13-0916.
    1. Denkert C, Loibl S, Noske A, Roller M, Muller BM, Komor M, et al. Tumor-associated lymphocytes as an independent predictor of response to neoadjuvant chemotherapy in breast cancer. J Clin Oncol. 2010;28(1):105–113. doi: 10.1200/JCO.2009.23.7370.
    1. Issa-Nummer Y, Loibl S, von Minckwitz G, Denkert C. Tumor-infiltrating lymphocytes in breast cancer: a new predictor for responses to therapy. Oncoimmunology. 2014;3:e27926. doi: 10.4161/onci.27926.
    1. Bianchini G, Gianni L. The immune system and response to HER2-targeted treatment in breast cancer. Lancet Oncol. 2014;15(2):e58–e68. doi: 10.1016/S1470-2045(13)70477-7.
    1. Loi S. Tumor-infiltrating lymphocytes, breast cancer subtypes and therapeutic efficacy. Oncoimmunology. 2013;2(7):e24720. doi: 10.4161/onci.24720.
    1. Demaria S, Volm MD, Shapiro RL, Yee HT, Oratz R, Formenti SC, et al. Development of tumor-infiltrating lymphocytes in breast cancer after neoadjuvant paclitaxel chemotherapy. Clin Cancer Res. 2001;7(10):3025–3030.
    1. Ladoire S, Mignot G, Dabakuyo S, Arnould L, Apetoh L, Rebe C, et al. In situ immune response after neoadjuvant chemotherapy for breast cancer predicts survival. J Pathol. 2011;224(3):389–400. doi: 10.1002/path.2866.
    1. Ruffell B, Au A, Rugo HS, Esserman LJ, Hwang ES, Coussens LM. Leukocyte composition of human breast cancer. Proc Natl Acad Sci USA. 2012;109(8):2796–2801. doi: 10.1073/pnas.1104303108.
    1. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–674. doi: 10.1016/j.cell.2011.02.013.
    1. Verma C, Eremin JM, Robins A, Bennett AJ, Cowley GP, El-Sheemy MA, et al. Abnormal T regulatory cells (Tregs: FOXP3+, CTLA-4+), myeloid-derived suppressor cells (MDSCs: monocytic, granulocytic) and polarised T helper cell profiles (Th1, Th2, Th17) in women with large and locally advanced breast cancers undergoing neoadjuvant chemotherapy (NAC) and surgery: failure of abolition of abnormal treg profile with treatment and correlation of treg levels with pathological response to NAC. J Transl Med. 2013;11:16. doi: 10.1186/1479-5876-11-16.
    1. Stoll G, Enot D, Mlecnik B, Galon J, Zitvogel L, Kroemer G. Immune-related gene signatures predict the outcome of neoadjuvant chemotherapy. Oncoimmunology. 2014;3(1):e27884. doi: 10.4161/onci.27884.
    1. Denkert C, Darb-Esfahani S, Loibl S, Anagnostopoulos I, Johrens K. Anti-cancer immune response mechanisms in neoadjuvant and targeted therapy. Semin Immunopathol. 2011;33(4):341–351. doi: 10.1007/s00281-011-0261-0.
    1. Menard C, Martin F, Apetoh L, Bouyer F, Ghiringhelli F. Cancer chemotherapy: not only a direct cytotoxic effect, but also an adjuvant for antitumor immunity. Cancer Immunol Immunother. 2008;57(11):1579–1587. doi: 10.1007/s00262-008-0505-6.
    1. Varchetta S, Gibelli N, Oliviero B, Nardini E, Gennari R, Gatti G, et al. Elements related to heterogeneity of antibody-dependent cell cytotoxicity in patients under trastuzumab therapy for primary operable breast cancer overexpressing Her2. Cancer Res. 2007;67(24):11991–11999. doi: 10.1158/0008-5472.CAN-07-2068.
    1. Beano A, Signorino E, Evangelista A, Brusa D, Mistrangelo M, Polimeni MA, et al. Correlation between NK function and response to trastuzumab in metastatic breast cancer patients. J Transl Med. 2008;6:25. doi: 10.1186/1479-5876-6-25.
    1. Kim PS, Armstrong TD, Song H, Wolpoe ME, Weiss V, Manning EA, et al. Antibody association with HER-2/neu-targeted vaccine enhances CD8 T cell responses in mice through Fc-mediated activation of DCs. J Clin Invest. 2008;118(5):1700–1711. doi: 10.1172/JCI34333.
    1. Criscitiello C, Esposito A, Gelao L, Fumagalli L, Locatelli M, Minchella I, et al. Immune approaches to the treatment of breast cancer, around the corner? Breast Cancer Res. 2014;16:204. doi: 10.1186/bcr3620.
    1. Park S, Jiang Z, Mortenson ED, Deng L, Radkevich-Brown O, Yang X, et al. The therapeutic effect of anti-HER2/neu antibody depends on both innate and adaptive immunity. Cancer Cell. 2010;18(2):160–170. doi: 10.1016/j.ccr.2010.06.014.
    1. Taylor C, Hershman D, Shah N, Suciu-Foca N, Petrylak DP, Taub R, et al. Augmented HER-2 specific immunity during treatment with trastuzumab and chemotherapy. Clin Cancer Res. 2007;13(17):5133–5143. doi: 10.1158/1078-0432.CCR-07-0507.
    1. Perez SA, Karamouzis MV, Skarlos DV, Ardavanis A, Sotiriadou NN, Iliopoulou EG, et al. CD4+ CD25+ regulatory T-cell frequency in HER-2/neu (HER)-positive and HER-negative advanced-stage breast cancer patients. Clin Cancer Res. 2007;13(9):2714–2721. doi: 10.1158/1078-0432.CCR-06-2347.
    1. Horlock C, Stott B, Dyson PJ, Morishita M, Coombes RC, Savage P, et al. The effects of trastuzumab on the CD4+ CD25+ FoxP3+ and CD4+ IL17A+ T-cell axis in patients with breast cancer. Br J Cancer. 2009;100(7):1061–1067. doi: 10.1038/sj.bjc.6604963.
    1. Muraro E, Martorelli D, Turchet E, Miolo G, Scalone S, Comaro E, et al. A different immunologic profile characterizes patients with HER-2-overexpressing and HER-2-negative locally advanced breast cancer: implications for immune-based therapies. Breast Cancer Res. 2011;13(6):R117. doi: 10.1186/bcr3060.
    1. Rayson D, Richel D, Chia S, Jackisch C, van der V, Suter T. Anthracycline-trastuzumab regimens for HER2/neu-overexpressing breast cancer: current experience and future strategies. Ann Oncol. 2008;19(9):1530–1539. doi: 10.1093/annonc/mdn292.
    1. Liang B, Zhu L, Liang Z, Weng X, Lu X, Zhang C, et al. A simplified PCR-SSP method for HLA-A2 subtype in a population of Wuhan, China. Cell Mol Immunol. 2006;3(6):453–458.
    1. Staaf J, Ringner M, Vallon-Christersson J, Jonsson G, Bendahl PO, Holm K, et al. Identification of subtypes in human epidermal growth factor receptor 2–positive breast cancer reveals a gene signature prognostic of outcome. J Clin Oncol. 2010;28(11):1813–1820. doi: 10.1200/JCO.2009.22.8775.
    1. Tamura K, Shimizu C, Hojo T, Akashi-Tanaka S, Kinoshita T, Yonemori K, et al. FcgammaR2A and 3A polymorphisms predict clinical outcome of trastuzumab in both neoadjuvant and metastatic settings in patients with HER2-positive breast cancer. Ann Oncol. 2011;22(6):1302–1307. doi: 10.1093/annonc/mdq585.
    1. Andersen MH, Pedersen LO, Capeller B, Brocker EB, Becker JC, Thor Straten P. Spontaneous cytotoxic T-cell responses against survivin-derived MHC class I-restricted T-cell epitopes in situ as well as ex vivo in cancer patients. Cancer Res. 2001;61(16):5964–5968.
    1. Jaramillo A, Narayanan K, Campbell LG, Benshoff ND, Lybarger L, Hansen TH, et al. Recognition of HLA-A2-restricted mammaglobin-A-derived epitopes by CD8+ cytotoxic T lymphocytes from breast cancer patients. Breast Cancer Res Treat. 2004;88(1):29–41. doi: 10.1007/s10549-004-8918-1.
    1. Brossart P, Heinrich KS, Stuhler G, Behnke L, Reichardt VL, Stevanovic S, et al. Identification of HLA-A2-restricted T-cell epitopes derived from the MUC1 tumor antigen for broadly applicable vaccine therapies. Blood. 1999;93(12):4309–4317.
    1. Fisk B, Blevins TL, Wharton JT, Ioannides CG. Identification of an immunodominant peptide of HER-2/neu protooncogene recognized by ovarian tumor-specific cytotoxic T lymphocyte lines. J Exp Med. 1995;181(6):2109–2117. doi: 10.1084/jem.181.6.2109.
    1. Disis ML, Knutson KL, Schiffman K, Rinn K, McNeel DG. Pre-existent immunity to the HER-2/neu oncogenic protein in patients with HER-2/neu overexpressing breast and ovarian cancer. Breast Cancer Res Treat. 2000;62(3):245–252. doi: 10.1023/A:1006438507898.
    1. Andersen MH, Reker S, Kvistborg P, Becker JC, Thor Straten P. Spontaneous immunity against Bcl-xL in cancer patients. J Immunol. 2005;175(4):2709–2714. doi: 10.4049/jimmunol.175.4.2709.
    1. Meier A, Reker S, Svane IM, Holten-Andersen L, Becker JC, Sondergaard I, et al. Spontaneous T-cell responses against peptides derived from the Taxol resistance-associated gene-3 (TRAG-3) protein in cancer patients. Cancer Immunol Immunother. 2005;54(3):219–228. doi: 10.1007/s00262-004-0578-9.
    1. Miolo G, Muraro E, Martorelli D, Lombardi D, Scalone S, Spazzapan S, et al. Anthracycline-free neoadjuvant therapy induces pathological complete responses by exploiting immune proficiency in HER2+ breast cancer patients. BMC Cancer. 2014;14(1):954. doi: 10.1186/1471-2407-14-954.
    1. Zitvogel L, Apetoh L, Ghiringhelli F, Kroemer G. Immunological aspects of cancer chemotherapy. Nat Rev Immunol. 2008;8(1):59–73. doi: 10.1038/nri2216.
    1. Kono K, Sato E, Naganuma H, Takahashi A, Mimura K, Nukui H, et al. Trastuzumab (Herceptin) enhances class I-restricted antigen presentation recognized by HER-2/neu-specific T cytotoxic lymphocytes. Clin Cancer Res. 2004;10(7):2538–2544. doi: 10.1158/1078-0432.CCR-03-0424.
    1. Carson WE, III, Shapiro CL, Crespin TR, Thornton LM, Andersen BL. Cellular immunity in breast cancer patients completing taxane treatment. Clin Cancer Res. 2004;10(10):3401–3409. doi: 10.1158/1078-0432.CCR-1016-03.
    1. Kubo M, Morisaki T, Matsumoto K, Tasaki A, Yamanaka N, Nakashima H, et al. Paclitaxel probably enhances cytotoxicity of natural killer cells against breast carcinoma cells by increasing perforin production. Cancer Immunol Immunother. 2005;54(5):468–476. doi: 10.1007/s00262-004-0617-6.
    1. Kute T, Stehle JR, Jr, Ornelles D, Walker N, Delbono O, Vaughn JP. Understanding key assay parameters that affect measurements of trastuzumab-mediated ADCC against Her2 positive breast cancer cells. Oncoimmunology. 2012;1(6):810–821. doi: 10.4161/onci.20447.
    1. Casares N, Pequignot MO, Tesniere A, Ghiringhelli F, Roux S, Chaput N, et al. Caspase-dependent immunogenicity of doxorubicin-induced tumor cell death. J Exp Med. 2005;202(12):1691–1701. doi: 10.1084/jem.20050915.
    1. Oda N, Shimazu K, Naoi Y, Morimoto K, Shimomura A, Shimoda M, et al. Intratumoral regulatory T cells as an independent predictive factor for pathological complete response to neoadjuvant paclitaxel followed by 5-FU/epirubicin/cyclophosphamide in breast cancer patients. Breast Cancer Res Treat. 2012;136(1):107–116. doi: 10.1007/s10549-012-2245-8.
    1. Ghiringhelli F, Menard C, Puig PE, Ladoire S, Roux S, Martin F, et al. Metronomic cyclophosphamide regimen selectively depletes CD4+ CD25+ regulatory T cells and restores T and NK effector functions in end stage cancer patients. Cancer Immunol Immunother. 2007;56(5):641–648. doi: 10.1007/s00262-006-0225-8.
    1. Stagg J, Loi S, Divisekera U, Ngiow SF, Duret H, Yagita H, et al. Anti-ErbB-2 mAb therapy requires type I and II interferons and synergizes with anti-PD-1 or anti-CD137 mAb therapy. Proc Natl Acad Sci USA. 2011;108(17):7142–7147. doi: 10.1073/pnas.1016569108.
    1. Yang L, Qi Y, Hu J, Tang L, Zhao S, Shan B. Expression of Th17 cells in breast cancer tissue and its association with clinical parameters. Cell Biochem Biophys. 2012;62(1):153–159. doi: 10.1007/s12013-011-9276-3.
    1. Kryczek I, Zhao E, Liu Y, Wang Y, Vatan L, Szeliga W, et al. Human TH17 cells are long-lived effector memory cells. Sci Transl Med. 2011;3(104):104. doi: 10.1126/scitranslmed.3002949.

Source: PubMed

3
S'abonner