期刊论文详细信息
Journal of Translational Medicine
Clinical significance of plasmacytoid dendritic cells and myeloid-derived suppressor cells in melanoma
Lieve Brochez3  Nanja van Geel3  Mireille Van Gele3  Claus Bachert2  Olga Krysko2  Bart Neyns4  Max Schreuer1  Reinhart Speeckaert3  Ines Chevolet3 
[1] Department of Medical Oncology, Ghent University Hospital, Ghent, Belgium;Upper Airways Research Laboratory, Ghent University Hospital, Ghent, Belgium;Department of Dermatology, Ghent University Hospital, De Pintelaan 185, Ghent, 9000, Belgium;Department of Medical Oncology, UZ-Brussel, Brussels, Belgium
关键词: Immunoprofiling;    Prognosis;    Myeloid differentiation;    Myeloid-derived suppressor cell (MDSC);    Plasmacytoid dendritic cell (pDC);    Melanoma;   
Others  :  1146390
DOI  :  10.1186/s12967-014-0376-x
 received in 2014-09-22, accepted in 2014-12-26,  发布年份 2015
PDF
【 摘 要 】

Background

Immune markers in the peripheral blood of melanoma patients could provide prognostic information. However, there is currently no consensus on which circulating cell types have more clinical impact. We therefore evaluated myeloid-derived suppressor cells (MDSC), dendritic cells (DC), cytotoxic T-cells and regulatory T-cells (Treg) in a series of blood samples of melanoma patients in different stages of disease.

Methods

Flow cytometry was performed on peripheral blood mononuclear cells of 69 stage I to IV melanoma patients with a median follow-up of 39 months after diagnosis to measure the percentage of monocytic MDSCs (mMDSCs), polymorphonuclear MDSCs (pmnMDSCs), myeloid DCs (mDCs), plasmacytoid DCs (pDCs), cytotoxic T-cells and Tregs. We also assessed the expression of PD-L1 and CTLA-4 in cytotoxic T-cells and Tregs respectively. The impact of cell frequencies on prognosis was tested with multivariate Cox regression modelling.

Results

Circulating pDC levels were decreased in patients with advanced (P = 0.001) or active (P = 0.002) disease. Low pDC levels conferred an independent negative impact on overall (P = 0.025) and progression-free survival (P = 0.036). Even before relapse, a decrease in pDC levels was observed (P = 0.002, correlation coefficient 0.898). High levels of circulating MDSCs (>4.13%) have an independent negative prognostic impact on OS (P = 0.012). MDSC levels were associated with decreased CD3+ (P < 0.001) and CD3 + CD8+ (P = 0.017) T-cell levels. Conversely, patients with high MDSC levels had more PD-L1+ T-cells (P = 0.033) and more CTLA-4 expression by Tregs (P = 0.003). pDCs and MDSCs were inversely correlated (P = 0.004). The impact of pDC levels on prognosis and prediction of the presence of systemic disease was stronger than that of MDSC levels.

Conclusion

We demonstrated that circulating pDC and MDSC levels are inversely correlated but have an independent prognostic value in melanoma patients. These cell types represent a single immunologic system and should be evaluated together. Both are key players in the immunological climate in melanoma patients, as they are correlated with circulating cytotoxic and regulatory T-cells. Circulating pDC and MDSC levels should be considered in future immunoprofiling efforts as they could impact disease management.

【 授权许可】

   
2015 Chevolet et al.; licensee BioMed Central.

【 预 览 】
附件列表
Files Size Format View
20150403112420494.pdf 664KB PDF download
Figure 2. 49KB Image download
Figure 1. 33KB Image download
【 图 表 】

Figure 1.

Figure 2.

【 参考文献 】
  • [1]Chevolet I, Speeckaert R, Haspeslagh M, Neyns B, Kruse V, Schreuer M, et al.: Peri-tumoral indoleamine 2,3-dioxygenase expression in melanoma: an early marker of resistance to immune control? Br J Dermatol 2014, 171(5):987-995.
  • [2]Speeckaert R, Vermaelen K, van Geel N, Autier P, Lambert J, Haspeslagh M, van Gele M, Thielemans K, Neyns B, Roche N, et al.: Indoleamine 2,3-dioxygenase, a new prognostic marker in sentinel lymph nodes of melanoma patients. Eur J Cancer 2012, 48:2004-2011.
  • [3]Galon J, Pages F, Marincola FM, Angell HK, Thurin M, Lugli A, Zlobec I, Berger A, Bifulco C, Botti G, et al.: Cancer classification using the Immunoscore: a worldwide task force. J Transl Med 2012, 10:205. BioMed Central Full Text
  • [4]Ascierto PA, Capone M, Urba WJ, Bifulco CB, Botti G, Lugli A, Marincola FM, Ciliberto G, Galon J, Fox BA: The additional facet of immunoscore: immunoprofiling as a possible predictive tool for cancer treatment. J Transl Med 2013, 11:54. BioMed Central Full Text
  • [5]Gabrilovich DI, Nagaraj S: Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol 2009, 9:162-174.
  • [6]Khaled YS, Ammori BJ, Elkord E: Myeloid-derived suppressor cells in cancer: recent progress and prospects. Immunol Cell Biol 2013, 91:493-502.
  • [7]Diaz-Montero CM, Finke J, Montero AJ: Myeloid-derived suppressor cells in cancer: therapeutic, predictive, and prognostic implications. Semin Oncol 2014, 41:174-184.
  • [8]Lindenberg JJ, Fehres CM, van Cruijsen H, Oosterhoff D, de Gruijl TD: Cross-talk between tumor and myeloid cells: how to tip the balance in favor of antitumor immunity. Immunotherapy 2011, 3:77-96.
  • [9]Gabrilovich D: Mechanisms and functional significance of tumour-induced dendritic-cell defects. Nat Rev Immunol 2004, 4:941-952.
  • [10]Almand B, Resser JR, Lindman B, Nadaf S, Clark JI, Kwon ED, Carbone DP, Gabrilovich DI: Clinical significance of defective dendritic cell differentiation in cancer. Clin Cancer Res 2000, 6:1755-1766.
  • [11]Bakdash G, Schreurs I, Schreibelt G, Tel J: Crosstalk between dendritic cell subsets and implications for dendritic cell-based anticancer immunotherapy. Expert Rev Clin Immunol 2014, 10:915-926.
  • [12]Guilliams M, Ginhoux F, Jakubzick C, Naik SH, Onai N, Schraml BU, Segura E, Tussiwand R, Yona S: Dendritic cells, monocytes and macrophages: a unified nomenclature based on ontogeny. Nat Rev Immunol 2014, 14:571-578.
  • [13]Charles J, Di Domizio J, Salameire D, Bendriss-Vermare N, Aspord C, Muhammad R, Lefebvre C, Plumas J, Leccia MT, Chaperot L: Characterization of circulating dendritic cells in melanoma: role of CCR6 in plasmacytoid dendritic cell recruitment to the tumor. J Invest Dermatol 2010, 130:1646-1656.
  • [14]Failli A, Legitimo A, Orsini G, Romanini A, Consolini R: Numerical defect of circulating dendritic cell subsets and defective dendritic cell generation from monocytes of patients with advanced melanoma. Cancer Lett 2013, 337:184-192.
  • [15]Chavan R, Salvador D, Gustafson MP, Dietz AB, Nevala W, Markovic SN: Untreated stage IV melanoma patients exhibit abnormal monocyte phenotypes and decreased functional capacity. Cancer Immunol Res 2014, 2:241-248.
  • [16]Hasskamp J, Zapas J, Elias G: Dendritic cells in patients with melanoma. Ann Surg Oncol 2008, 15:1807.
  • [17]Pinzon-Charry A, Maxwell T, Lopez JA: Dendritic cell dysfunction in cancer: a mechanism for immunosuppression. Immunol Cell Biol 2005, 83:451-461.
  • [18]Hoffmann TK, Muller-Berghaus J, Ferris RL, Johnson JT, Storkus WJ, Whiteside TL: Alterations in the frequency of dendritic cell subsets in the peripheral circulation of patients with squamous cell carcinomas of the head and neck. Clin Cancer Res 2002, 8:1787-1793.
  • [19]Beckebaum S, Zhang X, Chen X, Yu Z, Frilling A, Dworacki G, Grosse-Wilde H, Broelsch CE, Gerken G, Cicinnati VR: Increased levels of interleukin-10 in serum from patients with hepatocellular carcinoma correlate with profound numerical deficiencies and immature phenotype of circulating dendritic cell subsets. Clin Cancer Res 2004, 10:7260-7269.
  • [20]Pico de Coana Y, Masucci G, Hansson J, Kiessling R. Myeloid-derived suppressor cells and their role in CTLA-4 blockade therapy. Cancer Immunol Immunother. 2014 Sep;63(9):977-83.
  • [21]Miracco C, Mourmouras V, Biagioli M, Rubegni P, Mannucci S, Monciatti I, Cosci E, Tosi P, Luzi P: Utility of tumour-infiltrating CD25 + FOXP3+ regulatory T cell evaluation in predicting local recurrence in vertical growth phase cutaneous melanoma. Oncol Rep 2007, 18:1115-1122.
  • [22]Budczies J, Klauschen F, Sinn BV, Gyorffy B, Schmitt WD, Darb-Esfahani S, Denkert C: Cutoff Finder: a comprehensive and straightforward Web application enabling rapid biomarker cutoff optimization. PLoS One 2012, 7:e51862.
  • [23]Filipazzi P, Valenti R, Huber V, Pilla L, Canese P, Iero M, Castelli C, Mariani L, Parmiani G, Rivoltini L: Identification of a new subset of myeloid suppressor cells in peripheral blood of melanoma patients with modulation by a granulocyte-macrophage colony-stimulation factor-based antitumor vaccine. J Clin Oncol 2007, 25:2546-2553.
  • [24]Poschke I, Mougiakakos D, Hansson J, Masucci GV, Kiessling R: Immature immunosuppressive CD14 + HLA-DR-/low cells in melanoma patients are Stat3hi and overexpress CD80, CD83, and DC-sign. Cancer Res 2010, 70:4335-4345.
  • [25]Jordan KR, Amaria RN, Ramirez O, Callihan EB, Gao D, Borakove M, Manthey E, Borges VF, McCarter MD: Myeloid-derived suppressor cells are associated with disease progression and decreased overall survival in advanced-stage melanoma patients. Cancer Immunol Immunother 2013, 62:1711-1722.
  • [26]Rudolph BM, Loquai C, Gerwe A, Bacher N, Steinbrink K, Grabbe S, Tuettenberg A: Increased frequencies of CD11b(+) CD33(+) CD14(+) HLA-DR(low) myeloid-derived suppressor cells are an early event in melanoma patients. Exp Dermatol 2014, 23:202-204.
  • [27]Gros A, Turcotte S, Wunderlich JR, Ahmadzadeh M, Dudley ME, Rosenberg SA: Myeloid cells obtained from the blood but not from the tumor can suppress T-cell proliferation in patients with melanoma. Clin Cancer Res 2012, 18:5212-5223.
  • [28]Mandruzzato S, Solito S, Falisi E, Francescato S, Chiarion-Sileni V, Mocellin S, Zanon A, Rossi CR, Nitti D, Bronte V, Zanovello P: IL4Ralpha + myeloid-derived suppressor cell expansion in cancer patients. J Immunol 2009, 182:6562-6568.
  • [29]Weide B, Martens A, Zelba H, Stutz C, Derhovanessian E, Di Giacomo AM, Maio M, Sucker A, Schilling B, Schadendorf D, et al.: Myeloid-derived suppressor cells predict survival of patients with advanced melanoma: comparison with regulatory T cells and NY-ESO-1- or melan-A-specific T cells. Clin Cancer Res 2014, 20:1601-1609.
  • [30]Gabrilovich DI, Ostrand-Rosenberg S, Bronte V: Coordinated regulation of myeloid cells by tumours. Nat Rev Immunol 2012, 12:253-268.
  • [31]Labidi-Galy SI, Treilleux I, Goddard-Leon S, Combes JD, Blay JY, Ray-Coquard I, Caux C, Bendriss-Vermare N: Plasmacytoid dendritic cells infiltrating ovarian cancer are associated with poor prognosis. Oncoimmunology 2012, 1:380-382.
  • [32]Jensen TO, Schmidt H, Moller HJ, Donskov F, Hoyer M, Sjoegren P, Christensen IJ, Steiniche T: Intratumoral neutrophils and plasmacytoid dendritic cells indicate poor prognosis and are associated with pSTAT3 expression in AJCC stage I/II melanoma. Cancer 2012, 118:2476-2485.
  • [33]Ascierto PA, Kalos M, Schaer DA, Callahan MK, Wolchok JD: Biomarkers for immunostimulatory monoclonal antibodies in combination strategies for melanoma and other tumor types. Clin Cancer Res 2013, 19:1009-1020.
  • [34]Meyer C, Cagnon L, Costa-Nunes CM, Baumgaertner P, Montandon N, Leyvraz L, Michielin O, Romano E, Speiser DE: Frequencies of circulating MDSC correlate with clinical outcome of melanoma patients treated with ipilimumab. Cancer Immunol Immunother 2014, 63:247-257.
  • [35]Kitano S, Postow MA, Cortez C, Rasalan T, Gallardo HF, Panageas K, Yuan JD, Wolchok JD, Lesokhin AM: Myeloid-derived suppressor cell quantity prior to treatment with ipilimumab at 10mg/kg to predict for overall survival in patients with metastatic melanoma. J Clin Oncol 2012, 30:2518.
  • [36]Kitano S, Postow MA, Ziegler CG, Kuk D, Panageas KS, Cortez C, Rasalan T, Adamow M, Yuan J, Wong P, et al.: Computational algorithm-driven evaluation of monocytic myeloid-derived suppressor cell frequency for prediction of clinical outcomes. Cancer Immunol Res 2014, 2:812-821.
  • [37]Schouppe E, Van Overmeire E, Laoui D, Keirsse J, Van Ginderachter JA: Modulation of CD8(+) T-cell activation events by monocytic and granulocytic myeloid-derived suppressor cells. Immunobiology 2013, 218:1385-1391.
  • [38]Schilling B, Sucker A, Griewank K, Zhao F, Weide B, Gorgens A, Giebel B, Schadendorf D, Paschen A: Vemurafenib reverses immunosuppression by myeloid derived suppressor cells. Int J Cancer 2013, 133:1653-1663.
  • [39]Finkelstein SE, Carey T, Fricke I, Yu D, Goetz D, Gratz M, Dunn M, Urbas P, Daud A, DeConti R, et al.: Changes in dendritic cell phenotype after a new high-dose weekly schedule of interleukin-2 therapy for kidney cancer and melanoma. J Immunother 2010, 33:817-827.
  • [40]Urosevic M, Oberholzer PA, Maier T, Hafner J, Laine E, Slade H, Benninghoff B, Burg G, Dummer R: Imiquimod treatment induces expression of opioid growth factor receptor: a novel tumor antigen induced by interferon-alpha? Clin Cancer Res 2004, 10:4959-4970.
  文献评价指标  
  下载次数:3次 浏览次数:9次