Monitoring the responsiveness of T and antigen presenting cell compartments in breast cancer patients is useful to predict clinical tumor response to neoadjuvant chemotherapy

David A Bernal-Estévez, Oscar García, Ramiro Sánchez, Carlos A Parra-López, David A Bernal-Estévez, Oscar García, Ramiro Sánchez, Carlos A Parra-López

Abstract

Background: Vaccination of mice with tumors treated with Doxorubicin promotes a T cell immunity that relies on dendritic cell (DC) activation and is responsible for tumor control in vaccinated animals. Despite Doxorubicin in combination with Cyclophosphamide (A/C) is widely used to treat breast cancer patients, the stimulating effect of A/C on T and APC compartments and its correlation with patient's clinical response remains to be proved.

Methods: In this prospective study, we designed an in vitro system to monitor various immunological readouts in PBMCs obtained from a total of 17 breast cancer patients before, and after neoadjuvant anti-tumor therapy with A/C.

Results: The results show that before treatment, T cells and DCs, exhibit a marked unresponsiveness to in vitro stimulus: whereas T cells exhibit poor TCR internalization and limited expression of CD154 in response to anti-CD3/CD28/CD2 stimulation, DCs secrete low levels of IL-12p70 and limited CD83 expression in response to pro-inflammatory cytokines. Notably, after treatment the responsiveness of T and APC compartments was recovered, and furthermore, this recovery correlated with patients' residual cancer burden stage.

Conclusions: Our results let us to argue that the model used here to monitor the T and APC compartments is suitable to survey the recovery of immune surveillance and to predict tumor response during A/C chemotherapy.

Keywords: Breast cancer; Chemotherapy; Dendritic cells; Doxorubicin; Immune-monitoring; Neoadjuvant; T cells.

Conflict of interest statement

Ethical approval and consent to participate

This study was approved by the ethics committee of the Instituto Nacional de Cancerología (Bogotá-Colombia) by the reference number ACT-018 May 2012. The healthy donors and breast cancer patients sign the informed consent before the blood sample was drawn.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Figures

Fig. 1
Fig. 1
Assessing different cell populations ex vivo in PBMCs from healthy donors and BC patients before and after chemotherapy. a Paired analysis of tumor size (area in cm2) of the patients before therapy and after three cycles of A/C chemotherapy (n = 17). b Working strategy for multi-parametric cell analysis using flow cytometry. Monocytes and lymphocytes were defined by contour plots using SSC-A vs. FSC-A. Myeloid and plasmacytoid dendritic cell (DCs) (cells HLA-DR+ Lin1/CD15- CD11c + or HLA-DR+ Lin1/CD15- CD123+ respectively) and myeloid-derived suppressor cells (MDSCs) (cells HLA-DR- Lin1/CD15- CD13+ CD33+ ± Arginase 1+), were analyzed within the monocytic cell region. Finally, the percentage of CD4+ and regulatory T cells CD4+ CD25+ CD127- FoxP3+ (Tregs) was estimated within the lymphoid cell region. c Percentage of different sub-populations ex vivo in PBMCs from healthy donors (white box n = 10) and BC patients before (gray box n = 12) and after chemotherapy (dashed box n = 12). Panels summarize the percentages of DCs populations (top panel), MDSCs (middle panel) and CD4+ and CD4+ Tregs: CD4+ CD25+ CD127- and CD4+ CD25+ CD127- FoxP3+ (middle and right panels at the bottom). Paired analysis by Wilcoxon test, *** p < 0.001. Box and whiskers graph with 10–90% of data
Fig. 2
Fig. 2
DC maturation and IL-12p70 production are hampered in cancer patients before treatment. a The analysis by contour plots of a representative sample of myeloid (HLA-DR+ Lin1- CD11c+) and plasmacytoid (HLA-DR+ Lin1- CD123+) DCs is shown. b Representative histograms comparing the phenotype (CD83, CCR7, and CD86) of immature (empty histogram) and mature (gray histogram) in myeloid DCs (HLA-DR+ Lin1- CD11c+) derived from HD. c Quantification of CD83 expression in response to maturation stimulus (delta of the percentage of CD83 expression between mature and immature DCs) in DCs derived in PBMCs from HD (white box), and breast cancer patients before and after chemotherapy (grey and dashed box, respectively) in monocytic cells (defined by FSC-A vs SSC-A (left)), myeloid (HLA-DR+ Lin1- CD11c + (middle)) or plasmacytoid DCs (HLA-DR+ Lin1- CD123+ (right)). d Delta of concentration in pg/mL of IL-12p70 secreted in culture supernatants (difference in concentration secreted by mature and immature) DCs from HD (white box, n = 10) and patients before (gray box, n = 17) and after chemotherapy (dashed box, n = 17). Boxes and whiskers 10–90%, two-way ANOVA analysis, with Turkey’s multiple comparison tests, * p < 0.05, ** p < 0.01
Fig. 3
Fig. 3
Suppressed T cell responsiveness in BC patients before chemotherapy. a Representative contour plots of T cells (SSC-A vs. CD3+) of HD unstimulated or stimulated 24 h with anti-CD3/CD28/CD2 beads (numbers represent MFI of CD3). b Quantification of CD3 MFI in PBMCs of HD in response to in vitro stimulation (left panel), and delta of CD3 MFI from HD (white box), and BC patients before (gray box) and after chemotherapy (dashed box – right panel). c Representative contour plots of T cell (gated on CD3+) activation phenotype (CD154 vs. CD69) of cells obtained from HD in response to in vitro stimulation with anti-CD3/CD28/CD2 beads, numbers represent the percentage of each population. d Quantification of MFI of each activation marker (delta of stimulated minus unstimulated cells) of CD25 (left panel), CD69 (middle panel) and CD154 (right panel) of HD (white box), and BC patients before (gray box) and after chemotherapy (dashed box). Box and whiskers 10–90%. HD (n = 12), patients before (n = 17) and after chemotherapy (n = 17). Non-parametric t-test (panel B – left panel) and Two-way ANOVA analysis, with Turkey’s multiple comparison tests, * p < 0.05, ** p < 0.01
Fig. 4
Fig. 4
The predictive capacity of immune readouts for clinical response to chemotherapy. a Scatter plot of percentage of CD3 internalization vs. residual cancer burden (RCB) index in BC patients after chemotherapy (Pearson correlation = −0.583, p < 0.05). b Predictive value of TCR (CD3) internalization (left panel) and the delta percentage of CD83 expression in plasmacytoid DCs evaluated before therapy and compared in patients with or without better clinical response (pCR/RCB-I vs RCB-II respectively). c ROC curves of TCR internalization (AUC = 0.816, p = 0.0452), and delta percentage of CD83 expression in plasmacytoid DCs to predict tumor response (AUC = 0.825, p = 0.039). Box and whiskers 10–90%, Pearson correlation test, Mann-Whitney test, * p < 0.05, ** p < 0.01

References

    1. Kepp O, et al. Consensus guidelines for the detection of immunogenic cell death. Oncoimmunology. 2014;3(9):e955691. doi: 10.4161/21624011.2014.955691.
    1. Vacchelli E, et al. Loss-of-function alleles of P2RX7 and TLR4 fail to affect the response to chemotherapy in non-small cell lung cancer. Oncoimmunology. 2012;1(3):271–278. doi: 10.4161/onci.18684.
    1. Ladoire S, 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. Angell H, Galon J. From the immune contexture to the Immunoscore: the role of prognostic and predictive immune markers in cancer. Curr Opin Immunol. 2013;25(2):261–267. doi: 10.1016/j.coi.2013.03.004.
    1. Galon J, et al. Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science. 2006;313(5795):1960–1964. doi: 10.1126/science.1129139.
    1. Peng RQ, et al. Expression of calreticulin is associated with infiltration of T-cells in stage IIIB colon cancer. World J Gastroenterol. 2010;16(19):2428–2434. doi: 10.3748/wjg.v16.i19.2428.
    1. Stoll G, et al. Immune-related gene signatures predict the outcome of neoadjuvant chemotherapy. Oncoimmunology. 2014;3(1):e27884. doi: 10.4161/onci.27884.
    1. Rabinovich GA, Gabrilovich D, Sotomayor EM. Immunosuppressive strategies that are mediated by tumor cells. Annu Rev Immunol. 2007;25:267–296. doi: 10.1146/annurev.immunol.25.022106.141609.
    1. Bernal-Estevez D, et al. Chemotherapy and radiation therapy elicits tumor specific T cell responses in a breast cancer patient. BMC Cancer. 2016;16:591. doi: 10.1186/s12885-016-2625-2.
    1. Symmans WF, et al. Measurement of residual breast cancer burden to predict survival after neoadjuvant chemotherapy. J Clin Oncol. 2007;25(28):4414–4422. doi: 10.1200/JCO.2007.10.6823.
    1. Martinuzzi E, et al. Accelerated co-cultured dendritic cells (acDCs) enhance human antigen-specific T-cell responses. Blood. 2011;
    1. Mailliard RB, et al. Alpha-type-1 polarized dendritic cells: a novel immunization tool with optimized CTL-inducing activity. Cancer Res. 2004;64(17):5934–5937. doi: 10.1158/0008-5472.CAN-04-1261.
    1. Jonuleit H, et al. Pro-inflammatory cytokines and prostaglandins induce maturation of potent immunostimulatory dendritic cells under fetal calf serum-free conditions. Eur J Immunol. 1997;27(12):3135–3142. doi: 10.1002/eji.1830271209.
    1. Lipsitz SR, Kim K, Zhao L. Analysis of repeated categorical data using generalized estimating equations. Stat Med. 1994;13(11):1149–1163. doi: 10.1002/sim.4780131106.
    1. Whiteside TL. Induced regulatory T cells in inhibitory microenvironments created by cancer. Expert Opin Biol Ther. 2014;14(10):1411–1425. doi: 10.1517/14712598.2014.927432.
    1. Hamilton MJ, et al. Macrophages are more potent immune suppressors ex vivo than immature myeloid-derived suppressor cells induced by metastatic murine mammary carcinomas. J Immunol. 2014;192(1):512–522. doi: 10.4049/jimmunol.1300096.
    1. Markowitz J, et al. Myeloid-derived suppressor cells in breast cancer. Breast Cancer Res Treat. 2013;140(1):13–21. doi: 10.1007/s10549-013-2618-7.
    1. Wolf AM, et al. Increase of regulatory T cells in the peripheral blood of cancer patients. Clin Cancer Res. 2003;9(2):606–612.
    1. Marigo I, et al. Tumor-induced tolerance and immune suppression by myeloid derived suppressor cells. Immunol Rev. 2008;222:162–179. doi: 10.1111/j.1600-065X.2008.00602.x.
    1. Della Bella S, et al. Altered maturation of peripheral blood dendritic cells in patients with breast cancer. Br J Cancer. 2003;89(8):1463–1472. doi: 10.1038/sj.bjc.6601243.
    1. Limagne E, et al. Accumulation of MDSC and Th17 cells in patients with metastatic colorectal cancer predicts the efficacy of a FOLFOX-bevacizumab drug treatment regimen. Cancer Res. 2016;76(18):5241–5252. doi: 10.1158/0008-5472.CAN-15-3164.
    1. Gottfried E, Kreutz M, Mackensen A. Tumor-induced modulation of dendritic cell function. Cytokine Growth Factor Rev. 2008;19(1):65–77. doi: 10.1016/j.cytogfr.2007.10.008.
    1. Finn OJ. Cancer immunology. N Engl J Med. 2008;358(25):2704–2715. doi: 10.1056/NEJMra072739.
    1. Krowka JF, et al. Expression of CD69 after in vitro stimulation: a rapid method for quantitating impaired lymphocyte responses in HIV-infected individuals. J Acquir Immune Defic Syndr Hum Retrovirol. 1996;11(1):95–104. doi: 10.1097/00042560-199601010-00013.
    1. Wieland E, Shipkova M. Lymphocyte surface molecules as immune activation biomarkers. Clin Biochem. 2016;49(4–5):347–354. doi: 10.1016/j.clinbiochem.2015.07.099.
    1. Cochran JR, et al. Receptor clustering and transmembrane signaling in T cells. Trends Biochem Sci. 2001;26(5):304–310. doi: 10.1016/S0968-0004(01)01815-1.
    1. Mackey MF, Barth RJ, Jr, Noelle RJ. The role of CD40/CD154 interactions in the priming, differentiation, and effector function of helper and cytotoxic T cells. J Leukoc Biol. 1998;63(4):418–428. doi: 10.1002/jlb.63.4.418.
    1. Van Gool SW, et al. CD80, CD86 and CD40 provide accessory signals in a multiple-step T-cell activation model. Immunol Rev. 1996;153:47–83. doi: 10.1111/j.1600-065X.1996.tb00920.x.
    1. Mittal D, et al. New insights into cancer immunoediting and its three component phases--elimination, equilibrium and escape. Curr Opin Immunol. 2014;27:16–25. doi: 10.1016/j.coi.2014.01.004.
    1. Desmedt C, et al. Multifactorial approach to predicting resistance to anthracyclines. J Clin Oncol. 2011;29(12):1578–1586. doi: 10.1200/JCO.2010.31.2231.
    1. Zitvogel L, Tesniere A, Kroemer G. Cancer despite immunosurveillance: immunoselection and immunosubversion. Nat Rev Immunol. 2006;6(10):715–727. doi: 10.1038/nri1936.
    1. Datta J, et al. Progressive loss of anti-HER2 CD4+ T-helper type 1 response in breast tumorigenesis and the potential for immune restoration. Oncoimmunology. 2015;4(10):e1022301. doi: 10.1080/2162402X.2015.1022301.
    1. Ge Y, et al. Metronomic cyclophosphamide treatment in metastasized breast cancer patients: immunological effects and clinical outcome. Cancer Immunol Immunother. 2012;61(3):353–362. doi: 10.1007/s00262-011-1106-3.
    1. Verma C, 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. Yu J, et al. Myeloid-derived suppressor cells suppress antitumor immune responses through IDO expression and correlate with lymph node metastasis in patients with breast cancer. J Immunol. 2013;190(7):3783–3797. doi: 10.4049/jimmunol.1201449.
    1. Diaz-Montero CM, et al. Increased circulating myeloid-derived suppressor cells correlate with clinical cancer stage, metastatic tumor burden, and doxorubicin-cyclophosphamide chemotherapy. Cancer Immunol Immunother. 2009;58(1):49–59. doi: 10.1007/s00262-008-0523-4.
    1. Lee JJ, et al. Type 1-polarized dendritic cells loaded with autologous tumor are a potent immunogen against chronic lymphocytic leukemia. J Leukoc Biol. 2008;84(1):319–325. doi: 10.1189/jlb.1107737.
    1. Verronese E, et al. Immune cell dysfunctions in breast cancer patients detected through whole blood multi-parametric flow cytometry assay. Oncoimmunology. 2016;5(3):e1100791. doi: 10.1080/2162402X.2015.1100791.
    1. Shurin GV, et al. Chemotherapeutic agents in noncytotoxic concentrations increase antigen presentation by dendritic cells via an IL-12-dependent mechanism. J Immunol. 2009;183(1):137–144. doi: 10.4049/jimmunol.0900734.
    1. Shu U, et al. Activated T cells induce interleukin-12 production by monocytes via CD40-CD40 ligand interaction. Eur J Immunol. 1995;25(4):1125–1128. doi: 10.1002/eji.1830250442.
    1. Stuber E, Strober W, Neurath M. Blocking the CD40L-CD40 interaction in vivo specifically prevents the priming of T helper 1 cells through the inhibition of interleukin 12 secretion. J Exp Med. 1996;183(2):693–698. doi: 10.1084/jem.183.2.693.
    1. Alderson MR, et al. CD40 expression by human monocytes: regulation by cytokines and activation of monocytes by the ligand for CD40. J Exp Med. 1993;178(2):669–674. doi: 10.1084/jem.178.2.669.
    1. Campbell KA, et al. CD40 ligand is required for protective cell-mediated immunity to Leishmania major. Immunity. 1996;4(3):283–289. doi: 10.1016/S1074-7613(00)80436-7.
    1. Caux C, et al. Activation of human dendritic cells through CD40 cross-linking. J Exp Med. 1994;180(4):1263–1272. doi: 10.1084/jem.180.4.1263.
    1. Cella M, et al. Ligation of CD40 on dendritic cells triggers production of high levels of interleukin-12 and enhances T cell stimulatory capacity: T-T help via APC activation. J Exp Med. 1996;184(2):747–752. doi: 10.1084/jem.184.2.747.
    1. Kelsall BL, et al. Interleukin-12 production by dendritic cells. The role of CD40-CD40L interactions in Th1 T-cell responses. Ann N Y Acad Sci. 1996;795:116–126. doi: 10.1111/j.1749-6632.1996.tb52660.x.
    1. Kiener PA, et al. Stimulation of CD40 with purified soluble gp39 induces proinflammatory responses in human monocytes. J Immunol. 1995;155(10):4917–4925.
    1. Koch F, et al. High level IL-12 production by murine dendritic cells: upregulation via MHC class II and CD40 molecules and downregulation by IL-4 and IL-10. J Exp Med. 1996;184(2):741–746. doi: 10.1084/jem.184.2.741.
    1. Gu-Trantien C, et al. CD4(+) follicular helper T cell infiltration predicts breast cancer survival. J Clin Invest. 2013;123(7):2873–2892. doi: 10.1172/JCI67428.
    1. Ladoire S, et al. Immunogenic cell death-related biomarkers: impact on the survival of breast cancer patients after adjuvant chemotherapy. Oncoimmunology. 2016;5(2):e1082706. doi: 10.1080/2162402X.2015.1082706.
    1. Sistigu A, et al. Cancer cell-autonomous contribution of type I interferon signaling to the efficacy of chemotherapy. Nat Med. 2014;20(11):1301–1309. doi: 10.1038/nm.3708.

Source: PubMed

3
구독하다