Generation of clinical grade dendritic cells with capacity to produce biologically active IL-12p70

Anke Zobywalski, Miran Javorovic, Bernhard Frankenberger, Heike Pohla, Elisabeth Kremmer, Iris Bigalke, Dolores J Schendel, Anke Zobywalski, Miran Javorovic, Bernhard Frankenberger, Heike Pohla, Elisabeth Kremmer, Iris Bigalke, Dolores J Schendel

Abstract

Background: For optimal T cell activation it is desirable that dendritic cells (DCs) display peptides within MHC molecules as signal 1, costimulatory molecules as signal 2 and, in addition, produce IL-12p70 as signal 3. IL-12p70 polarizes T cell responses towards CD4+ T helper 1 cells, which then support the development of CD8+ cytotoxic T lymphocytes. We therefore developed new maturation cocktails allowing DCs to produce biologically active IL-12p70 for large-scale cancer vaccine development.

Methods: After elutriation of leukapheresis products in a closed bag system, enriched monocytes were cultured with GM-CSF and IL-4 for six days to generate immature DCs that were then matured with cocktails, containing cytokines, interferon-gamma, prostaglandin E2, and a ligand for Toll-like receptor 8, with or without poly (I:C).

Results: Mature DCs expressed appropriate maturation markers and the lymph node homing chemokine receptor, CCR7. They retained full maturity after culture for two days without maturation cocktails and following cryopreservation. TLR ligand stimulation induced DCs capable of secreting IL-12p70 in primary cultures and after one day of coculture with CD40L-expressing fibroblasts, mimicking an encounter with T cells. DCs matured with our new cocktails containing TLR8 ligand, with or without poly (I:C), induced alloresponses and stimulated virus-specific T cells after peptide-pulsing. DCs matured in cocktails containing TLR8 ligand without poly (I:C) could also be loaded with RNA as a source of antigen, whereas DCs matured in cocktails containing poly (I:C) were unable to express proteins following RNA transfer by electroporation.

Conclusion: Our new maturation cocktails allowed easy DC harvesting, stable maturation and substantial recoveries of mature DCs after cryopreservation. Our procedure for generating DCs is easily adaptable for GMP-compliance and yields IL-12p70-secreting DCs suitable for development of cancer vaccines using peptides or RNA as sources of immunizing antigens.

Figures

Figure 1
Figure 1
Time-line of experimental setup using monocytes derived from one representative donor. Monocytes were prepared from a leukapheresis by elutriation on day 0 and cultured for 6 days with GM-CSF and IL-4 to produce iDCs, which were then incubated with different maturation cocktails. After 24 h, mDCs were harvested and washed twice, phenotypes determined by FACS and aliquots were cryopreserved. The primary culture supernatants were collected to assess IL-12p70 and IL-10 by ELISA. Samples of the different DC populations were cocultured with fibroblast L-cells (signal-3 assay) for an additional 24 h and supernatants collected once again for IL-12p70 and IL-10 measurements. Mixed lymphocyte cultures were established using autologous and allogeneic lymphocytes (fraction 3 of elutriated leukaphesis cells, cryopreserved on day 0) as responding cells and DC1–DC5 cells as stimulating cells. Tritiated-thymidine incorporation into dividing cells was measured during the final 24 h of a 7-day coculture. Mature DC1–DC5 cells that were harvested and washed on day 7 were loaded with CEF peptides and used as stimulating cells for autologous lymphocytes (fraction 3 cells after elutriation, cryopreserved on day 0). Lymphocytes and the various DC populations were cocultured for 7 days, washed and restimulated with autologous monocytes (fraction 5 cells, cryopreserved on day 0), with or without CEF peptides. IFNγ secretion was assessed in a standard ELISPOT analysis 24 h later. DCs cryopreserved on day 7 were thawed and reassessed for phenotype after storage in liquid nitrogen. Cryopreserved monocytes (fraction 5 from day 0) were thawed and used to generate new mDCs which were loaded with EGFP RNA by electroporation on day 7. Flow cytometry to detect percentages of positive cells and intensity of fluorescence was performed at 24 h.
Figure 2
Figure 2
Generation of stable, mature DCs using different maturation cocktails. (A) Percentages of DCs harvested after primary cell culture (6 d differentiation + 24 h maturation) calculated on total seeded cells (mononuclear cells) or CD14-positive monocytes detected by FACS and manual counting. DC populations DC1–DC5 were matured in different cocktails as listed in Table 1. Viability was detected by 7AAD exclusion measured by flow cytometry in FL-3 of the FACS-Calibur and viable cells are expressed as percentages of total cells. Percentages of cells expressing various surface markers were determined by flow cytometry using the antibodies specified in "Methods", including CD14 (a monocyte marker), CD83 (a marker of mDCs), CD80 and CD86 (costimulatory molecules) and chemokine receptor 7 (CCR7 = CD197) as an indicator for DC migratory potential into lymph nodes. Data are expressed as percentages of total cells with acquisition of 1 × 104 events. CCR7 values presented here (Fig. 2A) represent the percentages in histograms overlayed by IgG2a isotype control, although generated from the same FACS stain they are slightly different to the CCR7 values shown as dot plots in Figure 2B. Broken lines indicate marker levels for DCs matured with DC1 (Jonuleit) cocktail. (B) Representative dot plots of DC1–DC5 populations showing percentages of cells positive for CD83 versus CCR7 and CD80 versus CD86. (C) DCs were washed free of maturation cytokines and cultured an additional 44 h in culture medium without cytokines. Viability was determined by 7AAD incorporation. Percentages of CD14, CD83, CD80, CD86 and CCR7 were determined as described above. Broken lines indicate marker levels for DCs matured with DC1 (Jonuleit) cocktail.
Figure 3
Figure 3
Production of IL-12p70 and IL-10 by DCs matured using different cocktails. Immature DCs were cultured with different maturation cocktails and the amounts of IL-12p70 and IL-10 were determined by standard ELISA. Filled bars indicate IL-12p70 and empty bars IL-10, respectively. Cytokine content was measured (A) in supernatant medium of primary maturation cultures after 7 d and (B) in supernatant medium of cultures containing washed mDCs and CD40L-transfected fibroblasts following coculture for 24 h, representing a signal-3 assay as described in "Methods". (C) The quotients of IL-12p70/IL-10 were determined for the DC populations matured in different cocktails, based on the pg/ml values of the signal-3 assay. For calculation of quotients, it was assumed that IL-12p70 and IL-10 have theoretically equal biological potential. Filled circles indicate quotients ranging from 0 (DC1 cells in Jonuleit cocktail) to 3.2 (DC5 cells in cocktail 5). The values of DCs matured in DC1 (Jonuleit) versus DC2 (Kalinski) cocktails are indicated by broken lines.
Figure 4
Figure 4
Analysis of allostimulatory capacity of different DCs in mixed lymphocyte reactions. (A) Negative controls of proliferation of irradiated (40 Gy) DCs alone. (B) Proliferation of autologous T cells stimulated by DC1–DC5 populations. (C) Proliferation of one representative allogeneic T cell responder stimulated by DC1–DC5 cells. Note that the y-axis is different for autologous and allogeneic T cell responses. (D) Summary of proliferation data of three independent allogeneic T cell responders in comparison to autologous T cells stimulated by DC1–DC5 cells.
Figure 5
Figure 5
Response of autologous lymphocytes from an HLA-A*0201-positive donor responding to virus-peptide pulsed DCs. T cell responses were assessed in an IFNγ-ELISPOT experiment using peripheral blood lymphocytes (PBLs: T cell enriched ELUTRA fraction 3 = 54.8% CD3 positive cells) that were first activated for 7 d with mature peptide-pulsed DCs and then restimulated for 24 h with monocytes plus CEF peptides. For the ELISPOT analyses, 4 × 103 autologous in vitro activated lymphocytes were stimulated with 2 × 103 monocytes together with the CEF peptide pool. The mean ± S.D. was calculated for triplicate wells. Note: Due to insufficient recoveries, lymphocytes activated by DC2 cells were not included in the assay. Because of limitations in HLA-A2 subtyped donors available for leukapheresis, this experiment was only included once in the full DC evaluation protocol.
Figure 6
Figure 6
Expression of EGFP in DCs transfected with EGFP-encoding in vitro transcribed RNA. Flow cytometry histogram overlays show EGFP expression following RNA transfer into mDCs on day 7 (filled curves) 24 h after electroporation and corresponding untransfected DCs (empty curves) as negative controls. DCs were matured in the four cocktails indicated, RNA was introduced by electroporation, the DCs were returned to their corresponding media containing maturation cocktails and harvested for flow cytometry 24 h later (day 8). Numbers indicate the percentages of EGFP-positive DCs and their mean fluorescence intensities. These data are representative of two independent experiments with measurements at 24 and 48 h.

References

    1. Banchereau J, Briere F, Caux C, Davoust J, Lebecque S, Liu YI, Pulandran B, Palucka K. Immunobiology of dendritic cells. Annu Rev Immunol. 2000;18:767–811. doi: 10.1146/annurev.immunol.18.1.767.
    1. Shortman K, Liu YJ. Mouse and human dendritic cell subtypes. Nat Rev Immunol. 2002;2:151–161. doi: 10.1038/nri746.
    1. Kuwajima S, Sato T, Ishida K, Tada H, Tezuka H, Ohteki T. Interleukin 15-dependent crosstalk between conventional and plasmacytoid dendritic cells is essential for CpG-induced immune activation. Nat Immunol. 2006;7:740–746. doi: 10.1038/ni1348.
    1. Pulendran B. Division of labor and cooperation between dendritic cells. Nat Immunol. 2006;7:699–700. doi: 10.1038/ni0706-699.
    1. Mellman I, Steinman RM. Dendritic cells: specialized and regulated antigen processing machines. Cell. 2001;106:255–258. doi: 10.1016/S0092-8674(01)00449-4.
    1. Lanzavecchia A, Sallusto F. Antigen decoding by T lymphocytes: from synapses to fate determination. Nat Immunol. 2001;2:487–492. doi: 10.1038/88678.
    1. Lanzavecchia A, Sallusto F. Regulation of T cell immunity by dendritic cells. Cell. 2001;106:263–266. doi: 10.1016/S0092-8674(01)00455-X.
    1. Nestle FO, Banchereau J, Hart D. Dendritic cells: On the move from bench to bedside. Nat Med. 2001;7:761–765. doi: 10.1038/89863.
    1. Steinman RM, Hawiger D, Nussenzweig MC. Tolerogenic dendritic cells. Annu Rev Immunol. 2003;21:685–711. doi: 10.1146/annurev.immunol.21.120601.141040.
    1. Fearnley DB, McLellan AD, Mannering SI, Hock BD, Hart DN. Isolation of human blood dendritic cells using the CMRF-44 monoclonal antibody: implications for studies on antigen-presenting cell function and immunotherapy. Blood. 1997;89:3708–3716.
    1. Bonifaz L, Bonnyay D, Mahnke K, Rivera M, Nussenzweig MC, Steinmann RM. Efficient targeting of protein antigen to the dendritic cell receptor DEC-205 in the steady state leads to antigen presentation on majorhistocompatibility complex class I products and peripheral CD8+ T cell tolerance. J Exp Med. 2002;196:1627–1638. doi: 10.1084/jem.20021598.
    1. Tacken PJ, de Vries IJ, Gijzen K, Joosten B, Wu D, Rother RP, Faas SJ, Punt CJ, Torensma R, Adema GJ, Figdor CG. Effective induction of naive and recall T-cell responses by targeting antigen to human dendritic cells via a humanized anti-DC-SIGN antibody. Blood. 2005;106:1278–1285. doi: 10.1182/blood-2005-01-0318.
    1. Markowicz S, Engleman EG. Granulocyte-macrophage colony-stimulating factor promotes differentiation and survival of human peripheral blood dendritic cells in vitro. J Clin Invest. 1990;85:955–961.
    1. Figdor CG, de Vries IJ, Lesterhuis WJ, Melief CJ. Dendritic cell immunotherapy: mapping the way. Nat Med. 2004;10:475–480. doi: 10.1038/nm1039.
    1. Ridgway D. The first 1000 dendritic cell vaccines. Cancer Invest. 2003;21:873–886. doi: 10.1081/CNV-120025091.
    1. Schadendorf D, Ugurel S, Schuler-Thurner B, Nestle FO, Enk A, Brocker EB, Grabbe S, Rittgen W, Edler L, Sucker A, Zimpfer-Rechner C, Berger T, Kamarashev J, Burg G, Jonuleit H, Tuttenberg A, Becker CJ, Keikavoussi P, Kaempken E, Schuler G. Dacarbazine (DTIC) versus vaccination with autologous peptide-pulsed dendritic cells (DC) in first-line treatment of patients with metastatic melanoma: a randomized phase III trial of the DC study group of the DeCOG. Ann Oncol. 2006;17:563–570. doi: 10.1093/annonc/mdj138.
    1. Trinchieri G. Interleukin-12 and the regulation of innate resistance and adaptive immunity. Nat Rev Immunol. 2003;3:133–146. doi: 10.1038/nri1001.
    1. Sporri R, Reise Sousa C. Inflammatory mediators are insufficient for full dendritic cell activation and promote expansion of CD4+ T cell populations lacking helper function. Nat Immunol. 2005;6:163–170. doi: 10.1038/ni1162.
    1. Jonuleit H, Kuhn U, Muller G, Steinbrink K, Paragnik L, Schmitt E, Knop J, Enk AH. Pro-inflammatory cytokines and prostaglandins induce maturation of potent immunostimulatory dendritic cells under fetal calf serum-free conditions. Eur J Immunol. 1997;27:3135–3142. doi: 10.1002/eji.1830271209.
    1. Mailliard RB, Wankowicz-Kalinska A, Cai Q, Wesa A, Hilkens CM, Kapsenberg ML, Kirkwood JM, Storkus WJ, Kalinski P. alpha-type-1 polarized dendritic cells: a novel immunization tool with optimized CTL-inducing activity. Cancer Res. 2004;64:5934–5937. doi: 10.1158/0008-5472.CAN-04-1261.
    1. Frankenberger B, Regn S, Geiger C, Noessner E, Falk CS, Pohla H, Javorovic M, Silberzahn T, Wilde S, Buchner A, Siebles M, Oberneder R, Willimsky G, Pezzutto A, Blankenstein T, Schendel DJ. Cell-based vaccines for metastatic renal cell carcinoma: genetically-engineered tumor cells and monocyte-derived dendritic cells. World J Urol. 2005;3:166–174. doi: 10.1007/s00345-005-0505-5.
    1. Feuerstein B, Berger TG, Maczek C, Roder C, Schreiner D, Hirsch U, Haendle I, Leisgang W, Glaser A, Kuss O, Diepgen TL, Schuler G, Schuler-Thurner B. A method for the production of cryopreserved aliquots of antigen-preloaded, mature dendritic cells ready for clinical use. J Immunol Methods. 2000;245:15–29. doi: 10.1016/S0022-1759(00)00269-6.
    1. Kalinski P, Vieira P, Schuitemaker JH, Cai Q, Kapsenberg M. Generation of human type 1- and type 2-polarized dendritic cells from peripheral blood. Methods Mol Biol. 2003;215:427–436.
    1. Garrone P, Neidhardt EM, Garcia E, Galibert L, van Kooten C, Banchereau J. Fas ligation induces apoptosis of CD40 activated human B lymphocytes. J Exp Med. 1995;182:1265–1273. doi: 10.1084/jem.182.5.1265.
    1. Becker C, Pohla H, Frankenberger B, Schuler T, Assenmacher M, Schendel DJ, Blankenstein T. Adoptive tumor therapy with T lymphocytes enriched through an IFNγ capture assay. Nat Med. 2001;7:1159–1162. doi: 10.1038/nm1001-1159.
    1. Pohla H, Frankenberger B, Stadlbauer B, Oberneder R, Hofstetter A, Willimsky G, Perzutto A, Dorken B, Blankenstein T, Schendel DJ. Allogeneic vaccination for renal cell carcinoma: Development and monitoring. Bone Marrow Transplant. 2000;25:83–87. doi: 10.1038/sj.bmt.1702362.
    1. Nair SK, Boczkowski D, Morse M, Morse M, Cumming RI, Lyerly HK, Gilboa E. Induction of primary carcinoembryonic antigen (CEA)-specific cytotoxic T lymphocytes in vitro using human dendritic cells transfected with RNA. Nat Biotechnol. 1998;16:364–369. doi: 10.1038/nbt0498-364.
    1. Javorovic M, Pohla H, Frankenberger B, Wolfel T, Schendel DJ. RNA transfer by electroporation into mature dendritic cells leading to reactivation of effector-memory cytotoxic T lymphocytes: a quantitative analysis. Mol Ther. 2005;12:734–743. doi: 10.1016/j.ymthe.2005.03.034.
    1. Banchereau J, Palucka AK, Dhodapkar M, Burkeholder S, Taquet N, Rolland A, Taquet S, Coquery S, Wittkowski KM, Bhardwai N, Pineiro L, Steinmann RM, Fay J. Immune and clinical responses in patientswith metastatic melanoma to CD34(+) progenitor-derived dendritic cell vaccine. Cancer Res. 2001;61:6451–6458.
    1. Jonuleit H, Giesecke-Tuettenberg A, Tuting T, Thurner-Schuler B, Stuge TB, Paragnik L, Kandemir A, Lee PP, Schuler G, Knop J, Enk AH. A comparison of two types of dendritic cell as adjuvants for the induction of melanoma-specific T-cell responses in humans following intranodal injection. Int J Cancer. 2001;93:243–251. doi: 10.1002/ijc.1323.
    1. Schuler-Thurner B, Schultz ES, Berger TG, Weinlich G, Ebner S, Woerl P, Bender A, Feuerstein B, Fritsch PO, Romani N, 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. 2002;195:1279–1288. doi: 10.1084/jem.20012100. Erratum in: J Exp Med 2003, 197:395.
    1. deVries IJ, Lesterhuis WJ, Scharenborg NM, Engelen LP, Ruiter DJ, Gerritsen MJ, Crookewit S, Britten CM, Torensma R, Adema GJ, Figdor CG, Punt CJ. Maturation of dendritic cells is a prerequisite for inducing immune responses in advanced melanoma patients. Clin Cancer Res. 2003;9:5091–5100.
    1. Kalinski P, Hilkens C, Wierenga E, Kapsenberg M. T cell priming by type-1 and type-2 polarized dendritic cell: the concept of a third signal. Immunol Today. 1999;20:561–567. doi: 10.1016/S0167-5699(99)01547-9.
    1. Vieira PL, de Jong EC. Transcriptional regulation of Th1/Th2 polarization. Immunol Today. 2000;21:479–483. doi: 10.1016/S0167-5699(00)01712-6.
    1. Wierenga EA, Kapsenberg ML, Kalinski P. Development of Th1-inducing capacity in myeloid dendritic cells requires environmental instruction. J Immunol. 2000;164:4507–4512.
    1. O'Gara A. Cytokines induce the development of functionally heterogeneous T helper cell subsets. Immunity. 1998;8:275–283. doi: 10.1016/S1074-7613(00)80533-6.
    1. Rengarajan J, Szabo SJ, Glimcher LH. Transcriptionalregulation of Th1/Th2 polarization. Immunol Today. 2000;21:479–483. doi: 10.1016/S0167-5699(00)01712-6.
    1. Berger TG, Strasser E, Smith R, Carste C, Schuler-Thurner B, Kaempken E, Schuler G. Efficient elutriation of monocytes within a closed system (Elutra) for clinical-scale generation of dendritic cells. J Immunol Methods. 2005;298:61–72. doi: 10.1016/j.jim.2005.01.005. Erratum in: J Immunol Methods 2005, 303:152.
    1. Napolitani G, Rinaldi A, Bertoni F, Sallusto F, Lanzavecchia A. Selected Toll-like receptor agonist combinations synergistically trigger a T helper type 1-polarizing program in dendritic cells. Nat Immunol. 2005;6:769–776. doi: 10.1038/ni1223.
    1. Prins RM, Craft N, Bruhn KW, Khan-Faroogi H, Koya RC, Stripecke R, Miller JF, Liau LM. The TLR-7 agonist, imiquimod, enhances dendritic cell survival and promotes tumor antigen-specific T cell priming: relation to central nervous system antitumor immunity. J Immunol. 2006;176:157–164.
    1. Heil F, Ahmad-Nejad P, Hemmi H, Hochrein H, Ampenberger F, Gellert T, Dietrich , Lipford G, Takeda K, Akira S, Wagner H, Bauer S. The Toll-like receptor 7 (TLR7)-specific stimulus loxoribine uncovers a strong relationship within the TLR7, 8 and 9 subfamily. Eur J Immunol. 2003;33:2987–2997. doi: 10.1002/eji.200324238.
    1. Reis e Sousa C. Dendritic cells in a mature age. Nat Rev Immunol. 2006;6:476–483. doi: 10.1038/nri1845.
    1. Langenkamp A, Messi M, Lanzavecchia A, Sallusto F. Kinetics of dendritic cell activation: impact on priming of TH1, TH2 and nonpolarized T cells. Nat Immunol. 2000;1:311–316. doi: 10.1038/79758.
    1. Gautier G, Humbert M, Deauvieau F, Scuiller M, Hiscott J, Bates EE, Trinchieri G, Caux C, Garrone P. A type I interferon autocrine-paracrine loop is involved in Toll-like receptor-induced interleukin-12p70 secretion by dendritic cells. J Exp Med. 2005;201:1435–1446. doi: 10.1084/jem.20041964.
    1. Gitlin L, Barchet W, Gilfillan S, Cella M, Beutler B, Flavell RA, Diamond MS, Colonna M. Essential role of mda-5 in type I IFN responses to polyriboinosinic: polyribocytidylic acid and encephalomyocarditis picornavirus. Proc Nat Acad Sci USA. 2006;103:8459–8464. doi: 10.1073/pnas.0603082103.
    1. Kato H, Takeuchi O, Sato S, Yoneyama M, Yamamoto M, Matsui K, Uematsu S, Jung A, Kawai T, Ishii KJ, Yamaguchi O, Otsu K, Tsujimaura T, Koh CS, Reis e Sousa C, Matsuura Y, Fujita T, Akira S. Differential roles of MDA5 and RIG-I helicases in the recognition of RNA viruses. Nature. 2006;441:101–105. doi: 10.1038/nature04734.
    1. Kadowaki N, Ho S, Antonenko S, Malefyt RW, Kastelein RA, Bazan F, Liu YJ. Subsets of human dendritic cell precursors express different toll-like receptors and respond to different microbial antigens. J Exp Med. 2001;194:863–869. doi: 10.1084/jem.194.6.863.

Source: PubMed

3
Abonner