Polymorphonuclear myeloid-derived suppressor cells impair the anti-tumor efficacy of GD2.CAR T-cells in patients with neuroblastoma

Nicola Tumino, Gerrit Weber, Francesca Besi, Francesca Del Bufalo, Valentina Bertaina, Paola Paci, Linda Quatrini, Laura Antonucci, Matilde Sinibaldi, Concetta Quintarelli, Enrico Maggi, Biagio De Angelis, Franco Locatelli, Lorenzo Moretta, Paola Vacca, Ignazio Caruana, Nicola Tumino, Gerrit Weber, Francesca Besi, Francesca Del Bufalo, Valentina Bertaina, Paola Paci, Linda Quatrini, Laura Antonucci, Matilde Sinibaldi, Concetta Quintarelli, Enrico Maggi, Biagio De Angelis, Franco Locatelli, Lorenzo Moretta, Paola Vacca, Ignazio Caruana

Abstract

The outcome of patients affected by high-risk or metastatic neuroblastoma (NB) remains grim, with ≥ 50% of the children experiencing relapse or progression of the disease despite multimodal, intensive treatment. In order to identify new strategies to improve the overall survival and the quality of life of these children, we recently developed and optimized a third-generation GD2-specific chimeric antigen receptor (CAR) construct, which is currently under evaluation in our Institution in a phase I/II clinical trial (NCT03373097) enrolling patients with relapsed/refractory NB. We observed that our CAR T-cells are able to induce marked tumor reduction and even achieve complete remission with a higher efficiency than that of other CAR T-cells reported in previous studies. However, often responses are not sustained and relapses occur. Here, we demonstrate for the first time a mechanism of resistance to GD2.CAR T-cell treatment, showing how polymorphonuclear myeloid-derived suppressor cells (PMN-MDSC) increase in the peripheral blood (PB) of NB patients after GD2.CAR T-cell treatment in case of relapse and loss of response. In vitro, isolated PMN-MDSC demonstrate to inhibit the anti-tumor cytotoxicity of different generations of GD2.CAR T-cells. Gene-expression profiling of GD2.CAR T-cells "conditioned" with PMN-MDSC shows downregulation of genes involved in cell activation, signal transduction, inflammation and cytokine/chemokine secretion. Analysis of NB gene-expression dataset confirms a correlation between expression of these genes and patient outcome. Moreover, in patients treated with GD2.CAR T-cells, the frequency of circulating PMN-MDSC inversely correlates with the levels of GD2.CAR T-cells, resulting more elevated in patients who did not respond or lost response to the treatment. The presence and the frequency of PMN-MDSC in PB of high-risk and metastatic NB represents a useful prognostic marker to predict the response to GD2.CAR T-cells and other adoptive immunotherapy. This study underlines the importance of further optimization of both CAR T-cells and clinical trial in order to target elements of the tumor microenvironment.

Keywords: Clinical response; GD2.CAR T-cells; Long-term response; Neuroblastoma; Polymorphonuclear myeloid-derived suppressor cells; T-cell functionality.

Conflict of interest statement

The authors declare that they have no competing interests.

© 2021. The Author(s).

Figures

Fig. 1
Fig. 1
Presence of PMN-MDSC in PB of NB patients and their effect on GD2.CAR T-cells. A, B Mononuclear cells isolated from the PB of NB patients were analyzed by flow-cytometry for the expression of specific markers allowing the identification of PMN-MDSC subsets. A Percentages of PMN-MDSC on CD45+cells (CD66b+CD14−cells) in healthy donors (HD, n = 10) and in NB patients (NB pts, n = 9). B A representative gating strategy for PMN-MDSC identification by flow-cytometry is shown. C, D Second- and third-generation GD2.CAR T-cells were cultured either in the absence (w/o, blue bars) or in the presence (cond, red bars) of PMN-MDSC collected from stem cell donors given G-CSF and undergoing to leukapheresis. After 48h, GD2.CAR T-cells and non-transduced (NT) cells (used as control) were collected, purified and co-cultured at the effector:target ratio 1:1 with the SH-SY5Y-eGFP+ NB cell line. Percentages of SH-SY5Y-eGFP+ NB residual live cells at day 5 of co-culture with (C) second- (n = 10) and D third- (n = 10) generation GD2.CAR T-cells are reported. Patient derived GD2.CAR T-cells expressing CD28.4-1BBζ (the same used in our clinical trial) were cultured in the presence of PB-derived (n = 8) or BM-derived (n = 3) PMN-MDSC or PB-derived neutrophils (n = 3) collected from NB patients. After 48h, GD2.CAR T-cells were collected, purified from PMN-MDSC/neutrophils as described in supplementary data (Additional file 6), and co-cultured at an Effector:Target (E/T) ratio 1:1 with SH-SY5Y-eGFP+ NB cell line for 3 days. Percentages of SH-SY5Y-eGFP+ NB residual live cells were analyzed. E One representative co-culture experiment of GD2.CAR T-cells and PMN-MDSC is shown. F Percentages of residual live NB tumor cells in different culture conditions is reported. G Correlation between PMN-MDSC and GD2.CAR T-cells in CD45+ cells in NB patients (n = 55, different time point after GD2.CAR T-cell infusion), r2 = 0.07 and p value is indicated. Mononuclear cells present in the PB of NB patients were analyzed by flow-cytometry. Graphs represent percentages of PMN-MDSC and the corresponding absolute number/µl of GD2.CAR T-cells in NB patients. H Graphs represent GD2.CAR T-cells/µl (blue squares) and the percentages of PMN-MDSC (red dots) in CD45+ cells in responder (n = 5) and non-responder (n = 6) patients (multiple comparison with mixed-effect analysis). *p < 0.05, **p < 0.01, ***p < 0.005. Where not indicated, data were not statistically significant. A Mann–Whitney and C, D, F Wilcoxon Student’s t test. Data are shown as mean ± standard error of the mean (SEM)
Fig. 2
Fig. 2
PMN-MDSC induce modulation of the gene-expression profile and inhibit cytokine secretion of GD2.CAR T-cells. AC NT and GD2.CAR T-cells were cultured for 48h either in the absence or in the presence (cond.) of PMN-MDSC (NT n = 3; GD2.CAR T-cells n = 3). A Gene expression level (Rq) represented as mean plot with the 95% of confidence interval for the total gene analyzed in NT and GD2.CAR T-cells conditioned or not with PMN-MDSC. B Heat-maps representing the unsupervised hierarchical clustering of samples analyzed for inflammation, kinome and signal transduction gene-expression profile. Columns represent samples (NT and GD2.CAR T-cells), rows represent genes. C Volcano plots show inflammation, kinome and signal transduction gene expression data. The red or green dots indicate genes-of-interest that display highly positive or negative fold-change (x axis) and high statistical significance (− log10 of p value, y axis). The gene name related to a red and green dot is reported in the table (right panel). D Levels (pg/ml) of the indicated cytokines/chemokines in the supernatant of NT (n = 6) and GD2.CAR T-cells (n = 12) conditioned (red) or not (blue) with PMN-MDSC are shown. E Kaplan–Meier curves estimate of overall survival of HR-NB patients according to the level of S100A8 or S100A9 or TNFAIP6 gene expression. Red (low) and blue (high) lines indicate the level of gene expression using median cut-off modus. Dataset used for the analysis is indicated. One-way ANOVA was used for statistical analysis. *p < 0.05, **p < 0.01, ***p < 0.005

References

    1. Park JH, Riviere I, Gonen M, Wang X, Senechal B, Curran KJ, et al. Long-term follow-up of CD19 CAR therapy in acute lymphoblastic leukemia. N Engl J Med. 2018;378(5):449–459. doi: 10.1056/NEJMoa1709919.
    1. Louis CU, Savoldo B, Dotti G, Pule M, Yvon E, Myers GD, et al. Antitumor activity and long-term fate of chimeric antigen receptor-positive T cells in patients with neuroblastoma. Blood. 2011;118(23):6050–6056. doi: 10.1182/blood-2011-05-354449.
    1. Thistlethwaite FC, Gilham DE, Guest RD, Rothwell DG, Pillai M, Burt DJ, et al. The clinical efficacy of first-generation carcinoembryonic antigen (CEACAM5)-specific CAR T cells is limited by poor persistence and transient pre-conditioning-dependent respiratory toxicity. Cancer Immunol Immunother. 2017;66(11):1425–1436. doi: 10.1007/s00262-017-2034-7.
    1. Heczey A, Louis CU, Savoldo B, Dakhova O, Durett A, Grilley B, et al. CAR T cells administered in combination with lymphodepletion and PD-1 inhibition to patients with neuroblastoma. Mol Ther. 2017;25(9):2214–2224. doi: 10.1016/j.ymthe.2017.05.012.
    1. Long AH, Highfill SL, Cui Y, Smith JP, Walker AJ, Ramakrishna S, et al. Reduction of MDSCs with all-trans retinoic acid improves CAR therapy efficacy for sarcomas. Cancer Immunol Res. 2016;4(10):869–880. doi: 10.1158/2326-6066.CIR-15-0230.
    1. Ruella M, Klichinsky M, Kenderian SS, Shestova O, Ziober A, Kraft DO, et al. Overcoming the immunosuppressive tumor microenvironment of hodgkin lymphoma using chimeric antigen receptor T cells. Cancer Discov. 2017;7(10):1154–1167. doi: 10.1158/-16-0850.
    1. Kloss CC, Lee J, Zhang A, Chen F, Melenhorst JJ, Lacey SF, et al. Dominant-negative TGF-beta receptor enhances PSMA-targeted human CAR T cell proliferation and augments prostate cancer eradication. Mol Ther. 2018;26(7):1855–1866. doi: 10.1016/j.ymthe.2018.05.003.
    1. Caruana I, Savoldo B, Hoyos V, Weber G, Liu H, Kim ES, et al. Heparanase promotes tumor infiltration and antitumor activity of CAR-redirected T lymphocytes. Nat Med. 2015;21(5):524–529. doi: 10.1038/nm.3833.
    1. Tumino N, Besi F, Di Pace AL, Mariotti FR, Merli P, Li Pira G, et al. PMN-MDSC are a new target to rescue graft-versus-leukemia activity of NK cells in haplo-HSC transplantation. Leukemia. 2020;34(3):932–937. doi: 10.1038/s41375-019-0585-7.
    1. Kumar V, Patel S, Tcyganov E, Gabrilovich DI. The nature of myeloid-derived suppressor cells in the tumor microenvironment. Trends Immunol. 2016;37(3):208–220. doi: 10.1016/j.it.2016.01.004.
    1. Knier B, Hiltensperger M, Sie C, Aly L, Lepennetier G, Engleitner T, et al. Myeloid-derived suppressor cells control B cell accumulation in the central nervous system during autoimmunity. Nat Immunol. 2018;19(12):1341–1351. doi: 10.1038/s41590-018-0237-5.
    1. Zoller M, Zhao K, Kutlu N, Bauer N, Provaznik J, Hackert T, et al. Immunoregulatory effects of myeloid-derived suppressor cell exosomes in mouse model of autoimmune alopecia areata. Front Immunol. 2018;9:1279. doi: 10.3389/fimmu.2018.01279.
    1. Veglia F, Perego M, Gabrilovich D. Myeloid-derived suppressor cells coming of age. Nat Immunol. 2018;19(2):108–119. doi: 10.1038/s41590-017-0022-x.
    1. Tumino N, Bilotta MT, Pinnetti C, Ammassari A, Antinori A, Turchi F, et al. Granulocytic myeloid-derived suppressor cells increased in early phases of primary HIV infection depending on TRAIL plasma level. J Acquir Immune Defic Syndr. 2017;74(5):575–582. doi: 10.1097/QAI.0000000000001283.
    1. Marvel D, Gabrilovich DI. Myeloid-derived suppressor cells in the tumor microenvironment: expect the unexpected. J Clin Invest. 2015;125(9):3356–3364. doi: 10.1172/JCI80005.
    1. Zhang S, Ma X, Zhu C, Liu L, Wang G, Yuan X. The role of myeloid-derived suppressor cells in patients with solid tumors: a meta-analysis. PLoS ONE. 2016;11(10):e0164514. doi: 10.1371/journal.pone.0164514.
    1. Burga RA, Thorn M, Point GR, Guha P, Nguyen CT, Licata LA, et al. Liver myeloid-derived suppressor cells expand in response to liver metastases in mice and inhibit the anti-tumor efficacy of anti-CEA CAR-T. Cancer Immunol Immunother. 2015;64(7):817–829. doi: 10.1007/s00262-015-1692-6.
    1. Quintarelli C, Orlando D, Boffa I, Guercio M, Polito VA, Petretto A, et al. Choice of costimulatory domains and of cytokines determines CAR T-cell activity in neuroblastoma. Oncoimmunology. 2018;7(6):e1433518. doi: 10.1080/2162402X.2018.1433518.
    1. Sinha P, Okoro C, Foell D, Freeze HH, Ostrand-Rosenberg S, Srikrishna G. Proinflammatory S100 proteins regulate the accumulation of myeloid-derived suppressor cells. J Immunol. 2008;181(7):4666–4675. doi: 10.4049/jimmunol.181.7.4666.
    1. Ghavami S, Eshragi M, Ande SR, Chazin WJ, Klonisch T, Halayko AJ, et al. S100A8/A9 induces autophagy and apoptosis via ROS-mediated cross-talk between mitochondria and lysosomes that involves BNIP3. Cell Res. 2010;20(3):314–331. doi: 10.1038/cr.2009.129.
    1. Wang S, Song R, Wang Z, Jing Z, Wang S, Ma J. S100A8/A9 in Inflammation. Front Immunol. 2018;9:1298. doi: 10.3389/fimmu.2018.01298.
    1. Sandu I, Cerletti D, Claassen M, Oxenius A. Exhausted CD8(+) T cells exhibit low and strongly inhibited TCR signaling during chronic LCMV infection. Nat Commun. 2020;11(1):4454. doi: 10.1038/s41467-020-18256-4.
    1. Lan G, Yu X, Sun X, Li W, Zhao Y, Lan J, et al. Comprehensive analysis of the expression and prognosis for TNFAIPs in head and neck cancer. Sci Rep. 2021;11(1):15696. doi: 10.1038/s41598-021-95160-x.
    1. Sacchi A, Tumino N, Sabatini A, Cimini E, Casetti R, Bordoni V, et al. Myeloid-derived suppressor cells specifically suppress IFN-gamma production and antitumor cytotoxic activity of Vdelta2 T cells. Front Immunol. 2018;9:1271. doi: 10.3389/fimmu.2018.01271.
    1. Tumino N, Di Pace AL, Besi F, Quatrini L, Vacca P, Moretta L. Interaction between MDSC and NK cells in solid and hematological malignancies: impact on HSCT. Front Immunol. 2021;12:638841. doi: 10.3389/fimmu.2021.638841.
    1. Veglia F, Sanseviero E, Gabrilovich DI. Myeloid-derived suppressor cells in the era of increasing myeloid cell diversity. Nat Rev Immunol. 2021;21(8):485–498. doi: 10.1038/s41577-020-00490-y.
    1. Ugel S, Delpozzo F, Desantis G, Papalini F, Simonato F, Sonda N, et al. Therapeutic targeting of myeloid-derived suppressor cells. Curr Opin Pharmacol. 2009;9(4):470–481. doi: 10.1016/j.coph.2009.06.014.
    1. Mirza N, Fishman M, Fricke I, Dunn M, Neuger AM, Frost TJ, et al. All-trans-retinoic acid improves differentiation of myeloid cells and immune response in cancer patients. Can Res. 2006;66(18):9299–9307. doi: 10.1158/0008-5472.CAN-06-1690.
    1. Friedman AD. Transcriptional control of granulocyte and monocyte development. Oncogene. 2007;26(47):6816–6828. doi: 10.1038/sj.onc.1210764.
    1. Sevko A, Michels T, Vrohlings M, Umansky L, Beckhove P, Kato M, et al. Antitumor effect of paclitaxel is mediated by inhibition of myeloid-derived suppressor cells and chronic inflammation in the spontaneous melanoma model. J Immunol. 2013;190(5):2464–2471. doi: 10.4049/jimmunol.1202781.
    1. Vincent J, Mignot G, Chalmin F, Ladoire S, Bruchard M, Chevriaux A, et al. 5-Fluorouracil selectively kills tumor-associated myeloid-derived suppressor cells resulting in enhanced T cell-dependent antitumor immunity. Can Res. 2010;70(8):3052–3061. doi: 10.1158/0008-5472.CAN-09-3690.
    1. Suzuki E, Kapoor V, Jassar AS, Kaiser LR, Albelda SM. Gemcitabine selectively eliminates splenic Gr-1+/CD11b+ myeloid suppressor cells in tumor-bearing animals and enhances antitumor immune activity. Clin Cancer Res. 2005;11(18):6713–6721. doi: 10.1158/1078-0432.CCR-05-0883.

Source: PubMed

3
Subskrybuj