Circulating myeloid-derived suppressor cells in patients with pancreatic cancer
 
Xiao-Dong Xu, Jun Hu, Min Wang, Feng Peng, Rui Tian, Xing-Jun Guo, Yu Xie and Ren-Yi Qin
Wuhan, China
 
 
Author Affiliations: Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China (Xu XD, Wang M, Peng F, Tian R, Guo XJ, Xie Y and Qin RY); Department of Colon Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China (Hu J)
Corresponding Author: Ren-Yi Qin, PhD, Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China (Tel/Fax: +86-27-83665294; Email: ryqin@tjh.tjmu.edu.cn)
 
© 2016, Hepatobiliary Pancreat Dis Int. All rights reserved.
doi: 10.1016/S1499-3872(15)60413-1
Published online September 9, 2015.
 
 
Acknowledgement: We thank Professor Jian-Xin Jiang for his help in preparation of this manuscript.
Contributors: WM and QRY proposed the study and revised the article. XXD and HJ performed the research and wrote the first draft. PF, TR, GXJ and XY collected and analyzed the data. All authors contributed to the design and interpretation of the study and to further drafts. QRY is the guarantor.
Funding: This study was supported by grants from the National Natural Science Foundation of China (81071775, 81272659, 81101621, 81160311, 81172064, 81001068, 81272425 and 81101870), National “Eleventh Five-Year” Scientific and Technological Support Projects (2006BAI02A13-402), Key Projects of Science Foundation of Hubei Province (2011CDA030), and Research Fund of Young Scholars for the Doctoral Program of Higher Education of China (20110142120014).
Ethical approval: These studies were approved by the Ethics Committee of Tongji Medical College, Huazhong University of Science and Technology.
Competing interest: No benefits in any form have been received or will be received from a commercial party related directly or indirectly to the subject of the article.
 
 
BACKGROUND: Myeloid-derived suppressor cells (MDSCs) are heterogeneous cell types that suppress T-cell responses in cancer patients and animal models, some MDSC subpopulations are increased in patients with pancreatic cancer. The present study was to investigate a specific subset of MDSCs in patients with pancreatic cancer and the mechanism of MDSCs increase in these patients.
 
METHODS: Myeloid cells from whole blood were collected from 37 patients with pancreatic cancer, 17 with cholangiocarcinoma, and 47 healthy controls. Four pancreatic cancer cell lines were co-cultured with normal peripheral blood mononuclear cells (PBMCs) to test the effect of tumor cells on the conversion of PBMCs to MDSCs. Levels of granulocyte-macrophage colony-stimulating factor (GM-CSF) and arginase activity in the plasma of cancer patients were analyzed by enzyme-linked immunosorbent assay.
 
RESULTS: CD14+/CD11b+/HLA-DR- MDSCs were increased in patients with pancreatic or bile duct cancer compared with those in healthy controls, and this increase was correlated with clinical cancer stage. Pancreatic cancer cell lines induced PBMCs to MDSCs in a dose-dependent manner. GM-CSF and arginase activity levels were significantly increased in the serum of patients with pancreatic cancer.
 
CONCLUSIONS: MDSCs were tumor related: tumor cells induced PBMCs to MDSCs in a dose-dependent manner and circulating CD14+/CD11b+/HLA-DR- MDSCs in pancreatic cancer patients were positively correlated with tumor burden. MDSCs might be useful markers for pancreatic cancer detection and progression.
 
(Hepatobiliary Pancreat Dis Int 2016;15:99-105)
 
KEY WORDS: pancreatic cancer; myeloid-derived suppressor cells; granulocyte-macrophage colony-stimulating factor; arginase
 
 
Introduction
Pancreatic cancer is a highly aggressive disease that is usually at an advanced stage at diagnosis in most patients and therefore, the prognosis is extremely poor. It is the sixth most common cause of cancer-related mortality in China. Novel therapies, such as immunotherapy, are therefore urgently needed for treatment of this disease. Pancreatic cancer-associated antigens have spurred the development of vaccination-based treatment strategies. Although animal models have offered promising results, most clinical studies found only limited success,[1] which may be due to immune suppression mechanisms.[2] Abnormal accumulation of myeloid-derived suppressor cells (MDSCs) is thought to play a critical immunosuppressive role in tumor immune evasion and promotion.[3]
 
MDSCs expand during cancer, inflammation and infection in both preclinical models and human patients. In mouse models, MDSCs are described as CD11b+/Gr-1+ cells. More recently, however, MDSCs have been divided into monocytic and granulocytic subsets, reflecting differential expression of Ly6C and Ly6G markers.[4, 5] Although CD15+ granulocytic MDSCs and CD33+ MDSCs have been widely studied,[6-11] the origin of monocytic MDSCs in pancreatic cancer is still not clear. It was demonstrated that in patients with hepatocellular carcinoma, CD14+ MDSCs induce T-cell anergy through multiple mechanisms.[12,13] Tumor-secreted proinflammatory molecules induce MDSCs, leading to the hypothesis that inflammation promotes accumulation of MDSCs, which down-regulates immune surveillance and anti-tumor immunity and therefore, facilitates tumor growth.[14] A recent study[15] showed that monocytic MDSC subset has the immunoregulatory properties in many solid tumors and in vitro assays. Aberrantly expressed granulocyte-macrophage colony stimulating factor (GM-CSF) represents a key inflammatory component that facilitates monocytic MDSC accumulation.[16] MDSCs have a remarkable ability to suppress T-cell reactivity by production of arginase, reactive oxygen species, inducible NO synthase, IL-10, IL-6, transforming growth factor-β, and sequestration of cysteine.[17-21] The present study focused on the accumulation of monocytic MDSCs, their relevance to tumor burden and the possible mechanisms.
 
 
Methods
Patients
Peripheral blood specimens were collected from 37 patients with pancreatic cancer (mean age: 57 years; range: 50-71) and 17 patients with cholangiocarcinoma (mean age: 55 years; range: 53-68) who were newly diagnosed at the Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, from May 2011 to October 2012 (Table). All cancer patients were grouped in accordance with the American Joint Committee on Cancer (AJCC) Cancer Staging Manual, 7th edition (2010). We collected 4 mL of venous blood from each patient in heparin-lithium-green top tubes (BD Biosciences, Bedford, MA, USA), before surgery, radiation, or any systemic chemotherapy. Forty-seven age-matched normal healthy volunteers served as controls. These studies were approved by the Ethics Committee of Tongji Medical College. Informed consent was obtained from all subjects.
 
Cell lines and cell culture
Pancreatic cancer cell lines were obtained from the Cell Repository, Chinese Academy of Sciences. BxPC-3 and SW-1990 were cultured in RPMI-1640 (Gibco, Life Technologies Corporation, Shanghai, China), Mia PaCa-2 and Panc-1 were in Dulbecco’s Modified Eagle’s Media (Gibco), supplemented with 10% FBS (Gibco), at 37 �� in a humidified 5% CO2 incubator, and passaged 2-3 times per week by brief trypsinization.
 
Antibodies and flow cytometry analysis
All monoclonal antibodies used in the study were purchased from BD Biosciences. Staining was performed on fresh venous blood collected in heparin-lithium-green tubes. Briefly 100 µL of blood was mixed with 10 µL of each antibody or isotype control. Tubes were incubated at 4 �� away from light for 30 minutes. After incubation, each sample was mixed with 2 mL of red blood cell lysing buffer and incubated for 10 minutes twice. Samples were washed with BD Pharmingen Stain Buffer. Pellets were resuspended in 200 µL of the buffer. Samples were acquired and analyzed by flow cytometry using LSR II (BD Biosciences). Fc receptors were blocked by pre-incubating cells with 10 µL of Fc receptor blocker (Miltenyi Biotec, Gladbach, Germany). Freshly drawn whole blood from patients was then labeled with FITC-conjugated CD14, PE-Cy7-conjugated CD11b, and APC-conjugated HLA-DR monoclonal antibodies. After lysing the red blood cells, flow cytometry samples were taken. Target cells were mainly found among peripheral blood mononuclear cells (PBMCs; P1), which were then gated based on their CD14+and CD11b+ expressions (P2); the HLA-DR- cell fraction was determined from this population. In our study, MDSCs were thus defined as CD14+/CD11b+/HLA-DR-, based on the study performed by Rodrigues et al.[15]
 
Cell isolation
PBMCs were isolated from the peripheral blood of healthy controls and cancer patients by density gradient centrifugation. In brief, blood was collected in heparin-treated tubes, diluted with RPMI 1640 medium, and carefully layered onto a density gradient Ficoll-Hypaque (Hao Yang, Tianjin, China). After centrifugation, the PBMC band was aspirated and washed three times with ice-cold phosphate buffered saline containing 1% human serum.
 
MDSC induction in vitro
In 12-well plates, 1×105 healthy control PBMCs were separately incubated with four pancreatic cancer cell lines for 48 hours at 37 �� in AIM V serum-free media (Gibco). Controls were cultured alone. The induced PBMCs were then harvested by scraping loosely attached cells. In co-culture experiments, isolated PBMCs were mixed with tumor cell lines.
 
Serum arginase assay
Part samples of pancreatic cancer patients (n=27) and controls (n=27) was analyzed for plasma arginase activity using a commercially available ELISA kit, specific for human arginase (Boster, Wuhan, China). Hemolyzed samples were excluded. Samples were assayed in triplicate. Mean absorbance was calculated from the standard curve. Arginase activity was expressed as units per liter.
 
Serum GM-CSF ELISA
Part samples of pancreatic cancer patients (n=29) and controls (n=25) were analyzed for plasma GM-CSF levels using a commercially available ELISA kit specific for human GM-CSF (Boster). Hemolyzed samples were excluded. Samples were assayed in triplicate. Mean absorbance was calculated from the standard curve.
 
Statistical analysis
Data were analyzed using GraphPad Prism version 5.0 (GraphPad Software Inc, La Jolla, CA, USA), and Student’s t test was performed using SPSS 17.0 statistical software when two conditions were compared. A P<0.05 was considered statistically significant. Graphical data were presented as means with standard errors.
 
 
Results
Elevation of CD14+/CD11b+/HLA-DR- MDSCs in patients with pancreatic cancer
Since MDSCs apparently increased in the peripheral blood of patients with pancreatic cancer,[6-11] we analyzed CD14+/CD11b+/HLA-DR- MDSCs in freshly drawn whole blood from patients with varying stages of pancreatic cancer (Table), and calculated them as percentages of PBMCs. Representative flow cytometry plots of a healthy control and two patients with stage I and III cancer respectively are shown in Fig. 1. More circulating MDSCs were seen in the patients with pancreatic cancer (3.05%±2.71%) than in normal controls (0.20%±0.21%; P<0.01; Fig. 2A). A similar expansion of CD14+/CD11b+/HLA-DR- MDSCs was observed in 17 patients with cholangiocarcinoma (P<0.05).
 
Correlation of frequency of MDSCs in peripheral blood with clinical cancer stage
We then grouped patients by clinical cancer stage. The differences between healthy controls (n=47; 0.20%±0.21%) and patients with stages I/II (n=8; 1.08%±0.34%; P=0.03), stage III (n=19; 2.25%±1.21%; P=0.009), and stage IV (n=10; 5.42%±2.76%; P=0.0007) disease were statistically significant (Fig. 2B). Moreover, the mean percentage of MDSC increased significantly at each advance in disease stage (P<0.05), and stage IV cancer patients had the greatest percentage of MDSC (Fig. 2C).
 
Induction of tumor-associated MDSCs in vitro
Patients with pancreatic cancer had increased MDSCs in PBMCs compared with their healthy counterparts. To see if tumor burden and tumor-derived factors are responsible for MDSC accumulation, four pancreatic cancer cell lines were co-cultured with PBMCs from four healthy controls to produce MDSCs. CD14+/CD11b+/HLA-DR- MDSCs were generated, and increased in number, in all four pancreatic cancer cell lines (Fig. 3A). We then compared the induction ability of the four cell lines with the same healthy PBMCs at the ratio of 10:1. The SW-1990 cells had the strongest MDSC-inducing capacity (Fig. 3B).
 
Elevated plasma GM-CSF and arginase activity in patients with pancreatic cancer
Multiple cytokines and growth factors, such as GM-CSF, VEGF, IL-6, and granulocyte-CSF, secreted in the tumor microenvironment promoted MDSC expansion and activation. However, serum GM-CSF levels in patients with pancreatic cancer were 54 pg/mL (n=29), not significantly different from those in healthy controls (62 pg/mL, n=25; Fig. 4A). L-arginine catabolismis was a primary mediator of MDSC immune suppression. Serum arginase activity was significantly elevated in patients with pancreatic cancer, compared with that in healthy controls (P=0.006; Fig. 4B).
 
 
Discussion
MDSCs possess strong immunosuppressive activities which limit immune-based cancer therapies, and identifying and characterizing these cells are of vital importance. Previous studies[6-11] confirmed the presence of CD33+ and CD15+ circulating MDSCs that suppress T-cell proliferation and cytokine production in patients with pancreatic cancer. Considering the heterogeneity of MDSC subgroups, we extended previous findings by analyzing the percentage, origin and immunosuppressive function of circulating CD14+ MDSCs in blood samples from patients with pancreatic cancer at various clinical stages.
 
Our results indicated that the percentage of monocytic MDSCs cells was significantly increased in patients with pancreatic cancer compared with that in healthy controls.[6-11] This increase is parallel to the cancer stage. The results were not consistent with those from another study[22] on colon cancer demonstrating that patients with stage I/II colon cancer had a moderate increase in CD33+ MDSCs.
 
Increased MDSCs have been reported in a variety of cancers and their accumulation appears to correlate with increased tumor burden in both animal models and cancer patients.[3, 7] We hypothesize that exposure of PBMCs to cancer cells results in the conversion from normal PBMCs to a MDSC-like phenotype. To test this hypothesis, four human pancreatic cancer cell lines were co-cultured with PBMCs from healthy controls. A previous study found that three of 10 pancreatic cancer cell lines induce PBMCs to CD33+ or CD11b+ MDSCs,[23] whereas all four pancreatic cancer cell lines in our study could induce CD14+ MDSCs. We found that CD14+ MDSCs were increased in parallel with disease stage in pancreatic cancer patients. Our data implied that CD14+ MDSCs were more specific for pancreatic cancer and might be a useful marker for pancreatic cancer detection and monitoring.
 
In both animal tumor models and cancer patients, tumor-derived factors, such as GM-CSF, granulocyte-CSF, IL-6, IL-13, prostaglandin E2 and VEGF, are responsible for the accumulation and activation of MDSC, which in turn contributed to tumor immune escape and the failure of anti-tumor immunotherapy.[24-30] GM-CSF is important in mobilization of hematopoietic stem cells, progenitors and mature cells. It also plays a role in regulating MDSC accumulation in both animal pancreatic cancer models and cancer patients.[10, 15] Furthermore, oncogenic K-ras-induced GM-CSF production may actively promote the development of MDSC.[24, 25] We therefore determined whether GM-CSF mediated induction of MDSC, and thus we examined whether patients with pancreatic cancer had increased serum GM-CSF levels. However, no difference was found between patients with pancreatic cancer and the healthy controls. Among studies of GM-CSF as a tumor marker in pancreatic cancer, one found that serum GM-CSF is increased in patients with pancreatic cancer,[31] whereas another study showed no difference.[32] Research showed that IL-6 had a similar effect on GM-CSF.[26] IL-6 derived from pancreatic cancer stroma was essential for MDSC generation, and IL-6 was critical for the development of pancreatic cancer in inducible K-ras driven mouse models.[27] Furthermore, one study showed a synergistic effect of inducing MDSC between GM-CSF and IL-6.[28] Therefore, multiple humoral factors produced by pancreatic cancer cells are likely responsible for MDSC proliferation. Two recent studies revealed that cytokines are diverse in patients with pancreatic cancer. Increased IL-6 other than granulocyte-CSF promoted systemic trafficking of immunosuppressive mesenchymal stem cells and other populations of bone marrow-derived cells.[33, 34]
 
Expression of high levels of arginase is the first major mechanism of MDSC immune suppression. Lack of L-arginine in the microenvironment significantly inhibits T-cell function.[35] Elevated arginase activity in serum and the tumor microenvironment has been reported in several cancers, including renal, melanoma and glioblastoma.[16, 36, 37] We proposed that the increased serum arginase in patients with pancreatic cancer is due to elevated MDSCs. Understanding these mechanisms should provide insights into novel therapeutic approaches to block immunosuppression and enhance the effect of immunotherapy.
 
In conclusion, pancreatic cancer cells induced the conversion from PBMCs to MDSCs which suppressed the immunoresponse via the catabolism of L-arginine. Circulating CD14+/CD11b+/HLA-DR- MDSCs correlated with clinical tumor stage. Pancreatic cancer burden was responsible for the abnormal accumulation of MDSC. MDSCs were a target in the treatment of patients with pancreatic cancer.
 
 
References
1 Beatty GL, Chiorean EG, Fishman MP, Saboury B, Teitelbaum UR, Sun W, et al. CD40 agonists alter tumor stroma and show efficacy against pancreatic carcinoma in mice and humans. Science 2011;331:1612-1616. PMID: 21436454
2 Almand B, Clark JI, Nikitina E, van Beynen J, English NR, Knight SC, et al. Increased production of immature myeloid cells in cancer patients: a mechanism of immunosuppression in cancer. J Immunol 2001;166:678-689. PMID: 11123353
3 Clark CE, Hingorani SR, Mick R, Combs C, Tuveson DA, Vonderheide RH. Dynamics of the immune reaction to pancreatic cancer from inception to invasion. Cancer Res 2007;67:9518-9527. PMID: 17909062
4 Bronte V, Chappell DB, Apolloni E, Cabrelle A, Wang M, Hwu P, et al. Unopposed production of granulocyte-macrophage colony-stimulating factor by tumors inhibits CD8+ T cell responses by dysregulating antigen-presenting cell maturation. J Immunol 1999;162:5728-5737. PMID: 10229805
5 Youn JI, Nagaraj S, Collazo M, Gabrilovich DI. Subsets of myeloid-derived suppressor cells in tumor-bearing mice. J Immunol 2008;181:5791-5802. PMID: 18832739
6 Schmielau J, Finn OJ. Activated granulocytes and granulocyte-derived hydrogen peroxide are the underlying mechanism of suppression of t-cell function in advanced cancer patients. Cancer Res 2001;61:4756-4760. PMID: 11406548
7 Diaz-Montero CM, Salem ML, Nishimura MI, Garrett-Mayer E, Cole DJ, Montero AJ. Increased circulating myeloid-derived suppressor cells correlate with clinical cancer stage, metastatic tumor burden, and doxorubicin-cyclophosphamide chemotherapy. Cancer Immunol Immunother 2009;58:49-59. PMID: 18446337
8 Mundy-Bosse BL, Young GS, Bauer T, Binkley E, Bloomston M, Bill MA, et al. Distinct myeloid suppressor cell subsets correlate with plasma IL-6 and IL-10 and reduced interferon-alpha signaling in CD4(+) T cells from patients with GI malignancy. Cancer Immunol Immunother 2011;60:1269-1279. PMID: 21604071
9 Gabitass RF, Annels NE, Stocken DD, Pandha HA, Middleton GW. Elevated myeloid-derived suppressor cells in pancreatic, esophageal and gastric cancer are an independent prognostic factor and are associated with significant elevation of the Th2 cytokine interleukin-13. Cancer Immunol Immunother 2011;60:1419-1430. PMID: 21644036
10 Porembka MR, Mitchem JB, Belt BA, Hsieh CS, Lee HM, Herndon J, et al. Pancreatic adenocarcinoma induces bone marrow mobilization of myeloid-derived suppressor cells which promote primary tumor growth. Cancer Immunol Immunother 2012;61:1373-1385. PMID: 22215137
11 Rey-Giraud F, Hafner M, Ries CH. In vitro generation of monocyte-derived macrophages under serum-free conditions improves their tumor promoting functions. PLoS One 2012;7:e42656. PMID: 22880072
12 Choi J, Suh B, Ahn YO, Kim TM, Lee JO, Lee SH, et al. CD15+/CD16low human granulocytes from terminal cancer patients: granulocytic myeloid-derived suppressor cells that have suppressive function. Tumour Biol 2012;33:121-129. PMID: 22081309
13 Hoechst B, Ormandy LA, Ballmaier M, Lehner F, Krüger C, Manns MP, et al. A new population of myeloid-derived suppressor cells in hepatocellular carcinoma patients induces CD4(+)CD25(+)Foxp3(+) T cells. Gastroenterology 2008;135: 234-243. PMID: 18485901
14 Hoechst B, Voigtlaender T, Ormandy L, Gamrekelashvili J, Zhao F, Wedemeyer H, et al. Myeloid derived suppressor cells inhibit natural killer cells in patients with hepatocellular carcinoma via the NKp30 receptor. Hepatology 2009;50:799-807. PMID: 19551844
15 Rodrigues JC, Gonzalez GC, Zhang L, Ibrahim G, Kelly JJ, Gustafson MP, et al. Normal human monocytes exposed to glioma cells acquire myeloid-derived suppressor cell-like properties. Neuro Oncol 2010;12:351-365. PMID: 20308313
16 Morales JK, Kmieciak M, Knutson KL, Bear HD, Manjili MH. GM-CSF is one of the main breast tumor-derived soluble factors involved in the differentiation of CD11b-Gr1- bone marrow progenitor cells into myeloid-derived suppressor cells. Breast Cancer Res Treat 2010;123:39-49. PMID: 19898981
17 Ochoa AC, Zea AH, Hernandez C, Rodriguez PC. Arginase, prostaglandins, and myeloid-derived suppressor cells in renal cell carcinoma. Clin Cancer Res 2007;13:721s-726s. PMID: 17255300
18 Kusmartsev S, Nefedova Y, Yoder D, Gabrilovich DI. Antigen-specific inhibition of CD8+ T cell response by immature myeloid cells in cancer is mediated by reactive oxygen species. J Immunol 2004;172:989-999. PMID: 14707072
19 Jia W, Jackson-Cook C, Graf MR. Tumor-infiltrating, myeloid-derived suppressor cells inhibit T cell activity by nitric oxide production in an intracranial rat glioma + vaccination model. J Neuroimmunol 2010;223:20-30. PMID: 20452681
20 Bellone G, Turletti A, Artusio E, Mareschi K, Carbone A, Tibaudi D, et al. Tumor-associated transforming growth factor-beta and interleukin-10 contribute to a systemic Th2 immune phenotype in pancreatic carcinoma patients. Am J Pathol 1999;155:537-547. PMID: 10433946
21 Srivastava MK, Sinha P, Clements VK, Rodriguez P, Ostrand-Rosenberg S. Myeloid-derived suppressor cells inhibit T-cell activation by depleting cystine and cysteine. Cancer Res 2010;70:68-77. PMID: 20028852
22 Zhang B, Wang Z, Wu L, Zhang M, Li W, Ding J, et al. Circulating and tumor-infiltrating myeloid-derived suppressor cells in patients with colorectal carcinoma. PLoS One 2013;8:e57114. PMID: 23437326
23 Lechner MG, Megiel C, Russell SM, Bingham B, Arger N, Woo T, et al. Functional characterization of human Cd33+ and Cd11b+ myeloid-derived suppressor cell subsets induced from peripheral blood mononuclear cells co-cultured with a diverse set of human tumor cell lines. J Transl Med 2011;9:90. PMID: 21658270
24 Bayne LJ, Beatty GL, Jhala N, Clark CE, Rhim AD, Stanger BZ, et al. Tumor-derived granulocyte-macrophage colony-stimulating factor regulates myeloid inflammation and T cell immunity in pancreatic cancer. Cancer Cell 2012;21:822-835. PMID: 22698406
25 Pylayeva-Gupta Y, Lee KE, Hajdu CH, Miller G, Bar-Sagi D. Oncogenic Kras-induced GM-CSF production promotes the development of pancreatic neoplasia. Cancer Cell 2012;21:836-847. PMID: 22698407
26 Zhang Y, Yan W, Collins MA, Bednar F, Rakshit S, Zetter BR, et al. Interleukin-6 is required for pancreatic cancer progression by promoting MAPK signaling activation and oxidative stress resistance. Cancer Res 2013;73:6359-6374. PMID: 24097820
27 Mace TA, Ameen Z, Collins A, Wojcik S, Mair M, Young GS, et al. Pancreatic cancer-associated stellate cells promote differentiation of myeloid-derived suppressor cells in a STAT3-dependent manner. Cancer Res 2013;73:3007-3018. PMID: 23514705
28 Marigo I, Bosio E, Solito S, Mesa C, Fernandez A, Dolcetti L, et al. Tumor-induced tolerance and immune suppression depend on the C/EBPbeta transcription factor. Immunity 2010;32:790-802. PMID: 20605485
29 Liu Y, Lai L, Chen Q, Song Y, Xu S, Ma F, et al. MicroRNA-494 is required for the accumulation and functions of tumor-expanded myeloid-derived suppressor cells via targeting of PTEN. J Immunol 2012;188:5500-5510. PMID: 22544933
30 Shen P, Wang A, He M, Wang Q, Zheng S. Increased circulating Lin(-/low) CD33(+) HLA-DR(-) myeloid-derived suppressor cells in hepatocellular carcinoma patients. Hepatol Res 2014;44:639-650. PMID: 23701406
31 Mroczko B, Szmitkowski M, Wereszczy��ska-Siemiatkowska U, Jurkowska G. Hematopoietic cytokines in the sera of patients with pancreatic cancer. Clin Chem Lab Med 2005;43:146-150. PMID: 15843207
32 Vasiliades G, Kopanakis N, Vasiloglou M, Zografos G, Margaris H, Masselou K, et al. Role of the hematopoietic cytokines SCF, IL-3, GM-CSF and M-CSF in the diagnosis of pancreatic and ampullary cancer. Int J Biol Markers 2012;27:e186-194. PMID: 22865301
33 Starzy��ska T, D?bkowski K, B?ogowski W, Zuba-Surma E, Budkowska M, Sa?ata D, et al. An intensified systemic trafficking of bone marrow-derived stem/progenitor cells in patients with pancreatic cancer. J Cell Mol Med 2013;17:792-799. PMID: 23672538
34 B?ogowski W, Deskur A, Budkowska M, Sa?ata D, Madej-Michniewicz A, D?bkowski K, et al. Selected cytokines in patients with pancreatic cancer: a preliminary report. PLoS One 2014;9:e97613. PMID: 24849506
35 Pauleau AL, Rutschman R, Lang R, Pernis A, Watowich SS, Murray PJ. Enhancer-mediated control of macrophage-specific arginase I expression. J Immunol 2004;172:7565-7573. PMID: 15187136
36 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. PMID: 20484028
37 Raychaudhuri B, Rayman P, Ireland J, Ko J, Rini B, Borden EC, et al. Myeloid-derived suppressor cell accumulation and function in patients with newly diagnosed glioblastoma. Neuro Oncol 2011;13:591-599. PMID: 21636707
 
Received October 5, 2014
Accepted after revision April 27, 2015