Open Access

Comparative study of dendritic cells matured by using IL‑1β, IL‑6, TNF‑α and prostaglandins E2 for different time span

  • Authors:
    • Xuejie Wu
    • Feng Xu
    • Jinliang Liu
    • Guiqiang Wang
  • View Affiliations

  • Published online on: June 22, 2017     https://doi.org/10.3892/etm.2017.4649
  • Pages: 1389-1394
  • Copyright: © Wu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Interleukin (IL)‑1β, IL‑6, tumor necrosis factor (TNF)‑α and prostaglandins E2 is considered as the standard cocktail for maturing dendritic cells (DCs). However, the appropriate time span for DC maturation with the standard cocktail remains unclear. The present study aimed to compare the differences between DCs matured with the standard cocktail for 24 and 48 h, respectively, and determine whether 24‑h stimulation was sufficient for DC maturation. The findings demonstrated that, compared with DCs matured for 48 h, the levels of cluster of differentiation (CD)80, CD83, CD86 and programmed death‑ligand 1 expression in DCs matured for 24 h were relatively lower. However, with the exception of CD80 whose mean fluorescence intensity (MFI) was higher in DCs matured for 48 h, the MFI values of other surface markers were comparable. Notably, the MFI of CD40 was higher in DCs matured for 24 h. In addition, the viability, T cell stimulatory capacity in allogeneic mixed lymphocyte reaction and cytokine production, including IL‑12p40, IL‑12p70 and IL‑10, were all comparable between DCs matured for 24 and 48 h, respectively. These results indicated that 24‑h stimulation may be sufficient for DC maturation when using the standard cocktail.

Introduction

Dendritic cells (DCs) are the most potent antigen-presenting cells and they have a critical role in innate and adaptive immunity (1,2). Notably, they have a unique ability to initiate naive T cells (3). Immature DCs (imDC), which are located in peripheral tissues (such as the skin, capture and process antigens), migrate to the draining lymphoid organs where they are able to prime cluster of differentiation (CD)4+ and CD8+ T cells (4). However, whether they induce T cell-mediated immune response or tolerance is determined by the status of DCs (5).

Over the past few years, great interest has been focused on the development of DC-based immunotherapy due to the unique capacity of DCs to initiate naive T cells. It is now straight-forward to generate monocyte-derived DCs (moDCs) in vitro using granulocyte-macrophage colony-stimulating factor (GM-CSF) and IL-4 (6,7). A number of protocols have been tested for their capacity to induce DC maturation (811) due to the fact that fully mature DCs are more powerful than imDC at inducing immune responses (5). Among these protocols, IL-1β, IL-6, TNF-α and prostaglandins E2 (PGE2), which was developed by Jonuleit et al (8), has become the gold standard cocktail for DC maturation. To date, DCs matured with this standard cocktail have been applied in the treatment of patients with different malignant tumors and promising results have been demonstrated in several clinical studies (1216).

Although this standard cocktail has been widely used, the appropriate time span for DC maturation has not been determined. It has been reported that DCs gradually lose their function over a few days after maturation (17). Therefore, shortening the time to mature DCs in vitro may be beneficial for the effectiveness of DC-based therapeutic vaccine in vivo. Therefore, the present study comprehensively compared DCs matured for 24 and 48 h using the standard cocktail to determine the appropriate time span for DC maturation.

Materials and methods

Isolation of PBMCs and positive selection of CD14+ monocytes

Peripheral blood mononuclear cells (PBMCs) were isolated by Ficoll-Paque Plus (GE Healthcare, Chicago, IL, USA) density gradient centrifugation from healthy human heparinized blood (Beijing 307 Hospital of Chinese People's Liberation Army, Beijing, China). All subjects were recruited in August 2015 (n=3; male: Female, 1:2; age 27.33±1.53 years). Inclusion criteria for subjects in this study were i) no fever; ii) normal renal and hepatic function; iii) no drug use within one month. Exclusion criteria were i) infection with hepatitis B virus, hepatitis C virus, hepatitis D virus or human immunodeficiency virus; ii) liver cirrhosis or hepatocellular carcinoma, fatty liver or alcoholic hepatitis. PBMCs were washed by PBS twice before CD14+ monocytes isolation using human CD14+ microbeads (Miltenyi Biotec GmbH, Bergisch Gladbach, Germany) according to the manufacturer's instructions. The purity of the isolated CD14+ monocytes was >90%. The Ethics Committee at the Second Affiliated Hospital, Zhejiang University School of Medicine (Hangzhou, China) approved this study. Informed consent was obtained from all participants.

DCs generation

Monocyte-derived DCs were generated as previously described with minor modifications (9). CD14+ monocytes were re-suspended in serum-free AIM-V medium (Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) supplemented with 100 U/ml penicillin and 100 µg/ml streptomycin and placed in 24-well plates (Corning, Inc., Corning, NY, USA) for incubation at 37°C in a humidified atmosphere containing 5% CO2 for 2 h. Following complete aspiration of the supernatant, fresh AIM-V medium supplemented with GM-CSF (1,000 IU/ml) and IL-4 (500 IU/ml; both PeproTech, Inc., Rocky Hill, NJ, USA) was added to the cells. The cells were supplied every 2 days with fresh medium. On day 5, imDC were harvested and cultured in the presence of IL-1β, IL-6, TNF-α (1,000 IU/ml; PeproTech, Inc.) and PGE2 (1 µg/ml; Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) for a further 24 or 48 h, respectively, to obtain mature DCs (mDC). Supernatants were collected and retained for cytokine analysis.

Flow cytometric analysis

Flow cytometry was performed using FACS Calibur (BD Biosciences). Cells were stained with the following monoclonal antibodies: Fluorescein isothiocyanate (FITC)-labeled antibodies against CD40 and CD86, phycoerythrin (PE)-labeled antibodies against CD80, CD83 and programmed death-ligand 1 (PD-L1), peridinin chlorophyll protein (PerCP)-labeled antibodies against human leukocyte antigen-D related (HLA-DR), allophycocyanin-labeled antibodies against CD14, CD11c, and isotype matched control antibodies (BD Biosciences). FACS data were analyzed using FlowJo software (version 5.7.2; Tree Star, Inc.).

Apoptosis assay

Freshly harvested mDC (1×105) were washed twice with cold PBS and incubated with Annexin-V-PE and 7-amino-actinomycin D (7-AAD) for 15 min before fluorescence-activated cell sorting (FACS) analysis. FACS data were analyzed using FlowJo software (version 5.7.2; Tree Star, Inc., Ashland, OR, USA).

Endocytic ability during the maturation of DCs

ImDC and mDC (1×105) cells were suspended in 100 µl of AIM-V and incubated with FITC-dextran (1 mg/ml) for 60 min either at 37°C or 4°C (negative control). Afterwards, cells were washed three times in cold PBS prior to FACS analysis. FACS data were analyzed using FlowJo software (version 5.7.2).

Allogeneic mixed lymphocyte reaction (MLR)

mDC matured for 24 or 48 h were treated with 50 µg/ml mitomycin-C at 37°C in a humidified 5% CO2 atmosphere for 45 min. Afterwards, DCs were washed three times and added to allogeneic CD14+ monocytes depleted PBMCs (105 cells) at a ratio 1:10 (DCs:PBMCs) in 96-well plates (Corning, Inc.) for 4 days, then 20 µl CellTiter 96 Aqueous non-radioactive reagent (Promega Corp., Madison, WI, USA) was added to each well and cultures were continued for another 4 h. Following this, absorbance at 490 nm was recorded using an ELISA plate reader.

Cytokines secretion analysis

Production of IL-12p40, IL-12p70 and IL-10 was assayed by ELISA kit (BioLegend, Inc., San Diego, CA, USA) according to the manufacturer's instructions.

Statistical analysis

Comparisons between groups of quantitative variables were performed using the Mann-Whitney U test. The test was two-sided and differences were considered significant if P<0.05. Data handling and analysis were performed with SPSS software for Windows, version 13.0 (SPSS Inc., Chicago, IL, USA).

Results

Purity of CD14+ monocytes and mature DCs

In order to improve the purity of monocyte-derived DCs, we first selected the CD14+ monocytes from PBMCs by using human CD14+ microbeads instead of the conventional cell adherent technique. Our data showed that the proportion of CD14+ monocytes in PBMCs was approximately 6.79% prior to selection. However, this number increased to 91.98% following selection (Fig. 1A). The purity of DCs matured for 24 and 48 h was 90.22 and 92.95%, respectively, of which the majority of mature DCs were CD11c+ DCs (Fig. 1B).

Phenotypic characteristics of mature DCs

In the present study, co-stimulatory and co-inhibitory surface markers, including CD40, CD80, CD83, CD86, HLA-DR and PD-L1, were compared between DCs matured via a standard cocktail for 24 and 48 h by flow cytometry. Compared to DCs matured for 24 h, DCs matured for 48 h expressed higher levels of CD80, both in frequency and mean fluorescence intensity (MFI). For CD83 and CD86 exhibited higher levels of frequency when matured for 48 h instead of 24 h, however no differences were found in MFI. Notably, similar expression levels of CD40 were found in frequency after 48 h, whereas MFI was higher in DCs matured for 24 h. No differences of HLA-DR were found in terms of frequency and MFI. However, for the inhibitory molecule, PD-L1, a higher frequency was also observed in DCs matured for 48 h. The frequencies of co-stimulatory molecules for both DCs all exceeded 90%, with the exception of CD83 (Table I).

Table I.

Phenotypic characteristics of mature DCs.

Table I.

Phenotypic characteristics of mature DCs.

Mature DCs

MarkersValue24 h48 h
CD40%   92.24   96.57
MFI   67.21   59.22a
CD80%   93.94   98.82a
MFI149.24247.51a
CD83%   83.55   94.98a
MFI   81.90   89.17
CD86%   94.47   99.52a
MFI198.56216.83
PD-L1%   95.03   98.01a
MFI105.18135.52
HLA-DR%   97.22   99.57
MFI629.59675.37

{ label (or @symbol) needed for fn[@id='tfn1-etm-0-0-4649'] } Phenotypes of mature DCs were analyzed by flow cytometry.

a P<0.05. MFI, mean fluorescence intensity; DCs, dendritic cells; CD, cluster of differentiation; PL-L1, programmed death-ligand 1; HLA-DR, human leukocyte antigen-D related.

Viability and endocytosis of mature DCs

High viability is important for the preparation of effective DC-based therapeutic vaccines. Therefore, we determined and compared the viability of DCs matured via the standard cocktail for different time spans. Our data showed that the viability of DCs matured for 24 and 48 h, respectively, were similarly high and exceeded 90% (Fig. 2A). It is known that the ability to take up antigens is one of the most important functions of immature DCs and this capacity decreases quickly upon maturation (18). Consistent with this, we found that imDCs showed high endocytosis while the endocytic capacity of DCs matured for 24 and 48 h both decreased rapidly to a similar extent (Fig. 2B).

T cell stimulatory capacity and cytokine productions of mature DCs

The T cell stimulatory capacity of DCs matured for different time spans was assessed via an allogeneic mixed lymphocyte reaction. Our data showed that the T cell stimulatory capacity of DCs matured for 24 and 48 h, respectively, was comparable (Fig. 3). We subsequently detected the cytokine production of mDCs from both time points and found that mDCs matured for 24 and 48 h, respectively, secreted comparable high levels of IL-12p40, which is a subunit of IL-12p70. Both groups of cells secreted comparable low levels of IL-10; however, mDCs matured for 24 h secreted relatively lower levels of bioactive IL-12p70 (Fig. 4).

Discussion

IL-1β, IL-6, TNF-α and PGE2 has been widely used as a standard cocktail for in vitro generation of mature DCs (8,1216,19,20). However, the optimal time for DC maturation using this standard cocktail has not been established. In the present study, we found that DCs matured for 24 h were, phenotypically speaking, also fully mature compared to DCs matured for 48 h. DCs matured for 24 h expressed even higher levels of the CD40 co-stimulatory molecule in terms of MFI, whereas lower levels of the co-inhibitory molecule, PD-L1, were detected in terms of frequency. Notably, the viability, endocytosis, T-cell stimulatory capacity and cytokine production were all comparable between DCs matured for 24 and 48 h, respectively.

Pioneering studies indicating the possibility of culturing murine DCs ex vivo from bone marrow precursors initiated DC vaccine development in the 1990s (21). Human applications followed soon thereafter and it was demonstrated that peripheral blood-derived monocytes and CD34+ hematopoietic progenitors are suitable for generating human DCs (22). In previous studies, DCs have been induced from adherent monocytes by washing out non-adherent cells, such as T and B cells. However, the purity of DCs obtained by this method is ~60%. In the present study, we induced DCs from CD14+ monocytes selected by using human CD14+ microbeads. The purity of DCs obtained from this method exceeds 90%, which is crucial for the improved effectiveness of DC-based immunotherapy (23).

The maturation state of DCs has been considered as a decisive factor in immune responses. Previous clinical studies have demonstrated that improved clinical outcomes were more frequently observed in trials using mature DCs in the therapeutic vaccination of patients with cancer, including prostate cancer, melanoma and glioblastoma (2426), although moderate clinical benefit was also reported in trials using IL-4 immature DCs (27). Due to their low co-stimulatory and MHC class I and II molecule expression, immature and semi-mature DCs are prone to inducing suboptimal T-cell priming and causing T-cell tolerance. Fully mature DCs (for example, matured with proinflammatory cytokines or TLR agonists) are able to prime CD4+ T and CD8+ T cells (5,28). It is well-known that co-stimulatory molecules, such as CD80 and CD86, have a key role in the induction of effective T cell responses (29). Our data showed that DCs matured for 24 and 48 h, respectively, expressed high levels of CD80 and CD86, which are important for the initiation of a robust immune response. Furthermore, CD40, which also has a critical role in T cell activation (30,31) expressed even higher levels in DCs matured for 24 h. Notably, PD-L1, which is well-known for its inhibitory role in T cell activation (3234), expressed relatively lower levels on DCs matured for 24 h and this may be beneficial for T cell priming.

It is increasingly recognized that abundant production of IL-12, particularly IL-12p70 during DC maturation has a crucial role in the differentiation and expansion of Th1 cell and Th1-polarized immunity (13,35,36). In clinical trials of melanoma (37) and glioblastoma (38), favorable outcomes were observed to be related to DC1-derived IL-12p70 production and Th1-polarized immunity. Similar to previous studies (39,40), the present study found that although DCs matured with a different time span secreted higher levels of IL-12p40, and secreted little bioactive IL-12p70. Therefore, studies to further improve the capacity of DCs to produce bioactive IL-12p70 are necessary. IL-10, known as an anti-inflammatory and immunosuppressive cytokine, was first described as a product of Th2 cells that inhibited cytokine synthesis in Th1 cells (41). It is now known that multiple immune cells, including macrophages, dendritic cells (DC), B cells, and various subsets of CD4+ and CD8+ T cells, are able to produce IL-10 (42). IL-10 inhibits the capacity of antigen-presenting cells, including DCs and macrophages, to present antigens to T cells in various ways to modulate immune responses (43). Recently, tumor cell-secreted IL-10 has been demonstrated to counteract the immunity of modified DCs in an established tumor model, which indicated that the high level of IL-10 within tumor microenvironment may impair DC vaccine functions (44). In the present study, DCs matured with the standard cocktail for different time spans (24 and 48 h) secreted minimal IL-10, which is a positive factor for DCs exerting immune responses.

High viability is another important factor in DC-based immunotherapy. In fact, DCs will gradually lose their function in a few days after maturation owing to apoptosis (17). In our present study, DCs matured for 24 and 48 h, respectively, exhibited high viability. The high viability may be due to the addition of PGE2 to the cocktail as previous studies have shown that PGE2 promotes apoptotic resistance and survival of DCs (45,46).

In conclusion, our preliminary results indicated that 24-h stimulation is sufficient for DC maturation when using IL-1β, IL-6, TNF-α and PGE2. Reducing the time to mature DCs in vitro from 48 to 28 h may be beneficial for the optimal preparation of tumor-pulsed DC therapeutic vaccine and improve its in vivo effectiveness.

Acknowledgements

The present study was supported by grants from the National Natural Science Foundation of China (grant no. 81400621) and the Mega-projects of Science Research (grant no. 008ZX10002-008).

Glossary

Abbreviations

Abbreviations:

DCs

dendritic cells

PBMCs

peripheral blood mononuclear cells

GM-CSF

granulocyte-macrophage colony-stimulating factor

PGE2

prostaglandin E2

PD-L1

programmed cell death ligand 1

References

1 

Banchereau J and Steinman RM: Dendritic cells and the control of immunity. Nature. 392:245–252. 1998. View Article : Google Scholar : PubMed/NCBI

2 

Kushwah R and Hu J: Complexity of dendritic cell subsets and their function in the host immune system. Immunology. 133:409–419. 2011. View Article : Google Scholar : PubMed/NCBI

3 

Schraml BU and e Sousa C Reis: Defining dendritic cells. Curr Opin Immunol. 32:13–20. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Guermonprez P, Valladeau J, Zitvogel L, Théry C and Amigorena S: Antigen presentation and T cell stimulation by dendritic cells. Annu Rev Immunol. 20:621–667. 2002. View Article : Google Scholar : PubMed/NCBI

5 

Lutz MB and Schuler G: Immature, semi-mature and fully mature dendritic cells: Which signals induce tolerance or immunity? Trends Immunol. 23:445–449. 2002. View Article : Google Scholar : PubMed/NCBI

6 

Romani N, Gruner S, Brang D, Kampgen E, Lenz A, Trockenbacher B, Konwalinka G, Fritsch PO, Steinman RM and Schuler G: Proliferating dendritic cell progenitors in human blood. J Exp Med. 180:83–93. 1994. View Article : Google Scholar : PubMed/NCBI

7 

Sallusto F and Lanzavecchia A: Efficient presentation of soluble antigen by cultured human dendritic cells is maintained by granulocyte/macrophage colony-stimulating factor plus interleukin 4 and downregulated by tumor necrosis factor alpha. J Exp Med. 179:1109–1118. 1994. View Article : Google Scholar : PubMed/NCBI

8 

Jonuleit H, Kühn U, Müller G, Steinbrink K, Paragnik L, Schmitt E, Knop J and Enk AH: Pro-inflammatory cytokines and prostaglandins induce maturation of potent immunostimulatory dendritic cells under fetal calf serum-free conditions. Eur J Immunol. 27:3135–3142. 1997. View Article : Google Scholar : PubMed/NCBI

9 

Zheng Z, Takahashi M, Narita M, Toba K, Liu A, Furukawa T, Koike T and Aizawa Y: Generation of dendritic cells from adherent cells of cord blood by culture with granulocyte-macrophage colony-stimulating factor, interleukin-4, and tumor necrosis factor-alpha. J Hematother Stem Cell Res. 9:453–464. 2000. View Article : Google Scholar : PubMed/NCBI

10 

Boullart AC, Aarntzen EH, Verdijk P, Jacobs JF, Schuurhuis DH, Benitez-Ribas D, Schreibelt G, van de Rakt MW, Scharenborg NM, de Boer A, et al: Maturation of monocyte-derived dendritic cells with Toll-like receptor 3 and 7/8 ligands combined with prostaglandin E2 results in high interleukin-12 production and cell migration. Cancer Immunol Immunother. 57:1589–1597. 2008. View Article : Google Scholar : PubMed/NCBI

11 

Anguille S, Smits EL, Cools N, Goossens H, Berneman ZN and Van Tendeloo VF: Short-term cultured, interleukin-15 differentiated dendritic cells have potent immunostimulatory properties. J Transl Med. 7:1092009. View Article : Google Scholar : PubMed/NCBI

12 

Schuler-Thurner B, Schultz ES, Berger TG, Weinlich G, Ebner S, Woerl P, Bender A, Feuerstein B, Fritsch PO, Romani N and Schuler G: Rapid induction of tumor-specific type 1 T helper cells in metastatic melanoma patients by vaccination with mature, cryopreserved, peptide-loaded monocyte-derived dendritic cells. J Exp Med. 195:1279–1288. 2002. View Article : Google Scholar : PubMed/NCBI

13 

Xu S, Koski GK, Faries M, Bedrosian I, Mick R, Maeurer M, Cheever MA, Cohen PA and Czerniecki BJ: Rapid high efficiency sensitization of CD8+ T cells to tumor antigens by dendritic cells leads to enhanced functional avidity and direct tumor recognition through an IL-12-dependent mechanism. J Immunol. 171:2251–2261. 2003. View Article : Google Scholar : PubMed/NCBI

14 

Ellebaek E, Engell-Noerregaard L, Iversen TZ, Froesig TM, Munir S, Hadrup SR, Andersen MH and Svane IM: Metastatic melanoma patients treated with dendritic cell vaccination, Interleukin-2 and metronomic cyclophosphamide: Results from a phase II trial. Cancer Immunol Immunother. 61:1791–1804. 2012. View Article : Google Scholar : PubMed/NCBI

15 

Vik-Mo EO, Nyakas M, Mikkelsen BV, Moe MC, Due-Tønnesen P, Suso EM, Sæbøe-Larssen S, Sandberg C, Brinchmann JE, Helseth E, et al: Therapeutic vaccination against autologous cancer stem cells with mRNA-transfected dendritic cells in patients with glioblastoma. Cancer Immunol Immunother. 62:1499–1509. 2013. View Article : Google Scholar : PubMed/NCBI

16 

Berntsen A, Trepiakas R, Wenandy L, Geertsen PF, Straten P thor, Andersen MH, Pedersen AE, Claesson MH, Lorentzen T, Johansen JS and Svane IM: Therapeutic dendritic cell vaccination of patients with metastatic renal cell carcinoma: A clinical phase 1/2 trial. J Immunother. 31:771–780. 2008. View Article : Google Scholar : PubMed/NCBI

17 

Lanzavecchia A and Sallusto F: Regulation of T cell immunity by dendritic cells. Cell. 106:263–266. 2001. View Article : Google Scholar : PubMed/NCBI

18 

Mellman I: Dendritic cells: Master regulators of the immune response. Cancer Immunol Res. 1:145–149. 2013. View Article : Google Scholar : PubMed/NCBI

19 

Leverkus M, Walczak H, McLellan A, Fries HW, Terbeck G, Bröcker EB and Kämpgen E: Maturation of dendritic cells leads to up-regulation of cellular FLICE-inhibitory protein and concomitant down-regulation of death ligand-mediated apoptosis. Blood. 96:2628–2631. 2000.PubMed/NCBI

20 

Krause P, Singer E, Darley PI, Klebensberger J, Groettrup M and Legler DF: Prostaglandin E2 is a key factor for monocyte-derived dendritic cell maturation: Enhanced T cell stimulatory capacity despite IDO. J Leukoc Biol. 82:1106–1114. 2007. View Article : Google Scholar : PubMed/NCBI

21 

Young JW and Steinman RM: Accessory cell requirements for the mixed-leukocyte reaction and polyclonal mitogens, as studied with a new technique for enriching blood dendritic cells. Cell Immunol. 111:167–182. 1988. View Article : Google Scholar : PubMed/NCBI

22 

Markowicz S and Engleman EG: Granulocyte-macrophage colony-stimulating factor promotes differentiation and survival of human peripheral blood dendritic cells in vitro. J Clin Invest. 85:955–961. 1990. View Article : Google Scholar : PubMed/NCBI

23 

Wang W, Li J, Wu K, Azhati B and Rexiati M: Culture and identification of mouse bone marrow-derived dendritic cells and their capability to induce T lymphocyte proliferation. Med Sci Monit. 22:244–250. 2016. View Article : Google Scholar : PubMed/NCBI

24 

Draube A, Klein-González N, Mattheus S, Brillant C, Hellmich M, Engert A and von Bergwelt-Baildon M: Dendritic cell based tumor vaccination in prostate and renal cell cancer: A systematic review and meta-analysis. PLoS One. 6:e188012011. View Article : Google Scholar : PubMed/NCBI

25 

de Vries IJ, Lesterhuis WJ, Scharenborg NM, Engelen LP, Ruiter DJ, Gerritsen MJ, Croockewit S, Britten CM, Torensma R, Adema GJ, et al: Maturation of dendritic cells is a prerequisite for inducing immune responses in advanced melanoma patients. Clin Cancer Res. 9:5091–5100. 2003.PubMed/NCBI

26 

Yamanaka R, Homma J, Yajima N, Tsuchiya N, Sano M, Kobayashi T, Yoshida S, Abe T, Narita M, Takahashi M and Tanaka R: Clinical evaluation of dendritic cell vaccination for patients with recurrent glioma: Results of a clinical phase I/II trial. Clin Cancer Res. 11:4160–4167. 2005. View Article : Google Scholar : PubMed/NCBI

27 

Anguille S, Smits EL, Lion E, van Tendeloo VF and Berneman ZN: Clinical use of dendritic cells for cancer therapy. Lancet Oncol. 15:e257–e267. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Cintolo JA, Datta J, Mathew SJ and Czerniecki BJ: Dendritic cell-based vaccines: Barriers and opportunities. Future Oncol. 8:1273–1299. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Greenwald RJ, Freeman GJ and Sharpe AH: The B7 family revisited. Annu Rev Immunol. 23:515–548. 2005. View Article : Google Scholar : PubMed/NCBI

30 

Taraban VY, Rowley TF and Al-Shamkhani A: Cutting edge: A critical role for CD70 in CD8 T cell priming by CD40-licensed APCs. J Immunol. 173:6542–6546. 2004. View Article : Google Scholar : PubMed/NCBI

31 

Ma DY and Clark EA: The role of CD40 and CD154/CD40L in dendritic cells. Semin Immunol. 21:265–272. 2009. View Article : Google Scholar : PubMed/NCBI

32 

Freeman GJ, Long AJ, Iwai Y, Bourque K, Chernova T, Nishimura H, Fitz LJ, Malenkovich N, Okazaki T, Byrne MC, et al: Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J Exp Med. 192:1027–1034. 2000. View Article : Google Scholar : PubMed/NCBI

33 

Carter L, Fouser LA, Jussif J, Fitz L, Deng B, Wood CR, Collins M, Honjo T, Freeman GJ and Carreno BM: PD-1:PD-L inhibitory pathway affects both CD4(+) and CD8(+) T cells and is overcome by IL-2. Eur J Immunol. 32:634–643. 2002. View Article : Google Scholar : PubMed/NCBI

34 

Selenko-Gebauer N, Majdic O, Szekeres A, Höfler G, Guthann E, Korthäuer U, Zlabinger G, Steinberger P, Pickl WF, Stockinger H, et al: B7-H1 (programmed death-1 ligand) on dendritic cells is involved in the induction and maintenance of T cell anergy. J Immunol. 170:3637–3644. 2003. View Article : Google Scholar : PubMed/NCBI

35 

Trinchieri G: Interleukin-12 and the regulation of innate resistance and adaptive immunity. Nat Rev Immunol. 3:133–146. 2003. View Article : Google Scholar : PubMed/NCBI

36 

Strioga MM, Felzmann T, Powell DJ Jr, Ostapenko V, Dobrovolskiene NT, Matuskova M, Michalek J and Schijns VE: Therapeutic dendritic cell-based cancer vaccines: The state of the art. Crit Rev Immunol. 33:489–547. 2013. View Article : Google Scholar : PubMed/NCBI

37 

Carreno BM, Becker-Hapak M, Huang A, Chan M, Alyasiry A, Lie WR, Aft RL, Cornelius LA, Trinkaus KM and Linette GP: IL-12p70-producing patient DC vaccine elicits Tc1-polarized immunity. J Clin Invest. 123:3383–3394. 2013. View Article : Google Scholar : PubMed/NCBI

38 

Okada H, Kalinski P, Ueda R, Hoji A, Kohanbash G, Donegan TE, Mintz AH, Engh JA, Bartlett DL, Brown CK, et al: Induction of CD8+ T-cell responses against novel glioma-associated antigen peptides and clinical activity by vaccinations with {alpha}-type 1 polarized dendritic cells and polyinosinic-polycytidylic acid stabilized by lysine and carboxymethylcellulose in patients with recurrent malignant glioma. J Clin Oncol. 29:330–336. 2011. View Article : Google Scholar : PubMed/NCBI

39 

Lee AW, Truong T, Bickham K, Fonteneau JF, Larsson M, Da Silva I, Somersan S, Thomas EK and Bhardwaj N: A clinical grade cocktail of cytokines and PGE2 results in uniform maturation of human monocyte-derived dendritic cells: Implications for immunotherapy. Vaccine. 20 Suppl 4:A8–A22. 2002. View Article : Google Scholar : PubMed/NCBI

40 

Trepiakas R, Pedersen AE, Met O, Hansen MH, Berntsen A and Svane IM: Comparison of alpha-Type-1 polarizing and standard dendritic cell cytokine cocktail for maturation of therapeutic monocyte-derived dendritic cell preparations from cancer patients. Vaccine. 26:2824–2832. 2008. View Article : Google Scholar : PubMed/NCBI

41 

Fiorentino DF, Bond MW and Mosmann TR: Two types of mouse T helper cell. IV. Th2 clones secrete a factor that inhibits cytokine production by Th1 clones. J Exp Med. 170:2081–2095. 1989. View Article : Google Scholar : PubMed/NCBI

42 

Couper KN, Blount DG and Riley EM: IL-10: The master regulator of immunity to infection. J Immunol. 180:5771–5777. 2008. View Article : Google Scholar : PubMed/NCBI

43 

Mittal SK and Roche PA: Suppression of antigen presentation by IL-10. Curr Opin Immunol. 34:22–27. 2015. View Article : Google Scholar : PubMed/NCBI

44 

Song S, Wang Y, Wang J, Lian W, Liu S, Zhang Z, Liu F and Wei L: Tumour-derived IL-10 within tumour microenvironment represses the antitumour immunity of Socs1-silenced and sustained antigen expressing DCs. Eur J Cancer. 48:2252–2259. 2012. View Article : Google Scholar : PubMed/NCBI

45 

Vassiliou E, Sharma V, Jing H, Sheibanie F and Ganea D: Prostaglandin E2 promotes the survival of bone marrow-derived dendritic cells. J Immunol. 173:6955–6964. 2004. View Article : Google Scholar : PubMed/NCBI

46 

Baratelli F, Krysan K, Heuzé-Vourc'h N, Zhu L, Escuadro B, Sharma S, Reckamp K, Dohadwala M and Dubinett SM: PGE2 confers survivin-dependent apoptosis resistance in human monocyte-derived dendritic cells. J Leukoc Biol. 78:555–564. 2005. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2017
Volume 14 Issue 2

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wu X, Xu F, Liu J and Wang G: Comparative study of dendritic cells matured by using IL‑1β, IL‑6, TNF‑α and prostaglandins E2 for different time span. Exp Ther Med 14: 1389-1394, 2017
APA
Wu, X., Xu, F., Liu, J., & Wang, G. (2017). Comparative study of dendritic cells matured by using IL‑1β, IL‑6, TNF‑α and prostaglandins E2 for different time span. Experimental and Therapeutic Medicine, 14, 1389-1394. https://doi.org/10.3892/etm.2017.4649
MLA
Wu, X., Xu, F., Liu, J., Wang, G."Comparative study of dendritic cells matured by using IL‑1β, IL‑6, TNF‑α and prostaglandins E2 for different time span". Experimental and Therapeutic Medicine 14.2 (2017): 1389-1394.
Chicago
Wu, X., Xu, F., Liu, J., Wang, G."Comparative study of dendritic cells matured by using IL‑1β, IL‑6, TNF‑α and prostaglandins E2 for different time span". Experimental and Therapeutic Medicine 14, no. 2 (2017): 1389-1394. https://doi.org/10.3892/etm.2017.4649