Optimizing parameters for clinical-scale production of high IL-12 secreting dendritic cells pulsed with oxidized whole tumor cell lysate

Cheryl L-L Chiang, Dawn A Maier, Lana E Kandalaft, Andrea L Brennan, Evripidis Lanitis, Qunrui Ye, Bruce L Levine, Brian J Czerniecki, Daniel J Powell Jr, George Coukos, Cheryl L-L Chiang, Dawn A Maier, Lana E Kandalaft, Andrea L Brennan, Evripidis Lanitis, Qunrui Ye, Bruce L Levine, Brian J Czerniecki, Daniel J Powell Jr, George Coukos

Abstract

Background: Dendritic cells (DCs) are the most potent antigen-presenting cell population for activating tumor-specific T cells. Due to the wide range of methods for generating DCs, there is no common protocol or defined set of criteria to validate the immunogenicity and function of DC vaccines.

Methods: Monocyte-derived DCs were generated during 4 days of culture with recombinant granulocyte-macrophage colony stimulating factor and interleukin-4, and pulsed with tumor lysate produced by hypochlorous acid oxidation of tumor cells. Different culture parameters for clinical-scale DC preparation were investigated, including: 1) culture media; 2) culture surface; 3) duration of activating DCs with lipopolysaccharide (LPS) and interferon (IFN)-gamma; 4) method of DC harvest; and 5) cryomedia and final DC product formulation.

Results: DCs cultured in CellGenix DC media containing 2% human AB serum expressed higher levels of maturation markers following lysate-loading and maturation compared to culturing with serum-free CellGenix DC media or AIM-V media, or 2% AB serum supplemented AIM-V media. Nunclon™Δ surface, but not Corning(®) tissue-culture treated surface and Corning(®) ultra-low attachment surface, were suitable for generating an optimal DC phenotype. Recombinant trypsin resulted in reduced major histocompatibility complex (MHC) Class I and II expression on mature lysate-loaded DCs, however presentation of MHC Class I peptides by DCs was not impaired and cell viability was higher compared to cell scraping. Preservation of DCs with an infusible cryomedia containing Plasma-Lyte A, dextrose, sodium chloride injection, human serum albumin, and DMSO yielded higher cell viability compared to using human AB serum containing 10% DMSO. Finally, activating DCs for 16 hours with LPS and IFN-γ stimulated robust mixed leukocyte reactions (MLRs), and high IL-12p70 production in vitro that continued for 24 hours after the cryopreserved DCs were thawed and replated in fresh media.

Conclusions: This study examined criteria including DC phenotype, viability, IL-12p70 production and the ability to stimulate MLR as metrics of whole oxidized tumor lysate-pulsed DC immunogenicity and functionality. Development and optimization of this unique method is now being tested in a clinical trial of autologous oxidized tumor lysate-pulsed DC in clinical-scale in recurrent ovarian, primary peritoneal or fallopian tube cancer (NCT01132014).

Figures

Figure 1
Figure 1
Comparison of the phenotype and IL-12p70 production of mature HOCl-oxidized lysate-loaded DCs generated in human AB serum-free or serum-containing AIM-V and CellGenix DC media, and supplementation with different concentrations of GM-CSF and IL-4. (A-B) DCs cultured in CellGenix DC media containing 2% human AB serum displayed a desirable phenotype with the highest level of DC-LAMP (*P = 0.005; one-way ANOVA) when compared to DCs cultured in other media conditions, and significantly higher CD86 (*P = 0.05) and lower CD14 (*P = 0.04) when compared to DCs cultured in serum-free CellGenix DC media. (C) DCs generated in CellGenix DC media plus 2% human AB serum produced slightly, though not significantly, higher amount of IL-12p70 compared to DCs generated in other media conditions (P = 0.2; one-way ANOVA). (D) 500 IU/ml recombinant GM-CSF and 250 IU/ml recombinant IL-4 were sufficient concentrations for generating a favorable immature DC phenotype with low CD14, and intermediate HLA-DR and CD1c. All the data presented were the mean of 6 independent experiments (i.e. DCs from 6 different individuals) in research-scale cultures in Nunclon™Δ Surface T25 cm2 flasks ± standard error of the mean (SEM). P values were determined with unpaired Student's t test unless otherwise stated. * denoted that P values were significant.
Figure 2
Figure 2
Comparison of the effect of Nunclon™Δ Surface, Corning® cell-culture surface and Corning® ultra-low attachment surface on DC phenotype and differentiation. DC were cultured with CellGenix DC media containing 2% human AB serum in T25 cm2 flasks that had the same surface properties as one of the three commercially available cell factories - Nunclon™Δ Surface 1-tray Cell Factories, Corning® CellSTACK® culture chambers or Corning® cell culture ultra-low attachment CellSTACK® chamber. DC phenotype was evaluated following lysate-loading and maturation for 16 h with LPS and IFN-γ. (A) DCs cultured in Nunclon™Δ Surface flasks (surrogate of Nunclon™Δ Surface 1-tray Cell Factories) exhibited an overall favorable immunophenotype with significantly higher DC-LAMP (**P = 0.003) and lower CD14 (*P = 0.01) when compared to DCs cultured in Corning® cell-culture surface flasks (surrogate of Corning® CellSTACK® culture chambers). Data were the mean of 6 independent experiments (i.e. DCs from 6 different individuals) in research-scale cultures ± SEM. (B) DCs that were cultured in Corning® ultra-low attachment surface (surrogate of Corning® ultra-low attachment cell factory chambers) expressed significantly lower CD86 (**P = 0.011) and HLA-DR (***P = 0.0004) following lysate-loading and maturation when compared to DCs cultured in Nunclon™Δ Surface (surrogate of Nunclon™Δ Surface 1-tray Cell Factories) and treated the same way. Data were the mean of 3 independent experiments (i.e. DCs from 3 different individuals) in research-scale cultures ± SEM. P values were determined with unpaired Student's t test. * denoted that P values were highly significant.
Figure 3
Figure 3
Comparison of TrypLE™ Select animal-free tyrpsin immobilization and cold PBS with cell scraping for harvesting mature HOCl-oxidized lysate-loaded DCs. (A) DCs were matured with LPS and IFN-γ for 16 h following lysate-loading. It was observed that mature lysate-loaded DCs that were harvested using TrypLE™ Select animal-free tyrpsin replacement exhibited approximately 30-40% reduction in the levels of MHC Class I (P = 0.13; NS) and HLA-DR (P = 0.15; NS) compared to DCs that were harvested with cold DPBS and cell scraping. Data were the mean of 3 independent experiments (i.e. DCs from 3 different individuals) ± SEM in research-scale Nunclon™Δ Surface T25 cm2 flasks. P values were determined with unpaired Student's t test. NS indicated that P values were not significant. (B) Mature lysate-loaded DCs that were harvested with TrypLE™ Select animal-free tyrpsin replacement were as efficient as DCs that were harvested with cold DPBS and cell scraping in presenting MHC Class I-restricted HER-2/neu and MART-1 peptides and stimulating HER-2/neu and MART-1 CD8+ T cells in intracellular IFN-γ assessments.
Figure 4
Figure 4
Comparing the phenotype of DCs previously cryopreserved in infusible cryomedia or human AB serum containing 10% DMSO. (A) Middle panel, DCs that had been cyropreserved with infusible cryomedia showed similar expressions of CD86 and CD1c as fresh DCs (first panel) and higher CD80, CD40 and MHC Class I levels after being thawed and replated for 24 h in fresh human AB serum-supplemented CellGenix DC media. Third panel, DCs that were cryopreserved with 90% human AB serum plus 10% DMSO exhibited similar level of CD80, CD40, MHC Class I and CD1c as fresh DCs. DCs cryopreserved in infusible cryomedia expressed approximately 50% lower levels of HLA-DR (**P = 0.001) compared to fresh DCs. On the other hand, DCs cryopreserved in human AB serum plus 10% DMSO media expressed approximately 80% lower levels of HLA-DR (**P = 0.004) compared to fresh DCs. Data were from a clinical-scale Nunclon™Δ Surface 1-tray Cell Factory and was representative of 3 independent research-scales experiments (i.e. DCs from 3 different individuals) in Nunclon™Δ Surface T25 cm2 flasks. (B) Summary of flow cytometry results from the mean of 3 independent research-scales experiments in Nunclon™Δ Surface T25 cm2 flasks (MFI ± SEM). P values are determined with unpaired Student's t test. ** denotes P value highly significant.
Figure 5
Figure 5
Time-course of IL-12p70 production by DCs. IL-12p70 was detected from fresh DCs from 6 h post LPS and IFN-γ stimulation and peaked at 16 h. Fresh replated DCs and thawed replated DCs continued to produce IL-12p70 albeit at a lower level than fresh DCs. Data were the mean of 3 independent experiments (i.e. DCs from 3 different individuals) in duplicates in research-scale Nunclon™Δ Surface T25 cm2 flasks ± SEM. IL-12p70 concentration is expressed as pg/ml.
Figure 6
Figure 6
Schematic diagram of optimized DC preparation.
Figure 7
Figure 7
Mixed leukocyte reaction capability of optimized DCs. DCs generated from clinical-scale Nunclon™Δ Surface 1-tray Cell Factory and DCs generated from research-scale Nunclon™Δ Surface T25 cm2 flasks using the optimized protocol depicted in Figure 6 were equally potent in stimulating robust mixed leukocytes reactions from 3 different individuals' T cells.

References

    1. Thurner B, Röder C, Dieckmann D, Heuer M, Kruse M, Glaser A, Keikavoussi P, Kämpgen E, Bender A, Schuler G. Generation of large numbers of fully mature and stable dendritic cells from leukapheresis products for clinical application. J Immunol Methods. 1999;223:1–15. doi: 10.1016/S0022-1759(98)00208-7.
    1. Berger TG, Strasser E, Smith R, Carste C, Schuler-Thurner B, Kaempgen 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.
    1. Berger TG, Feuerstein B, Strasser E, Hirsch U, Schreiner D, Schuler G, Schuler-Thurner B. Large-scale generation of mature monocyte-derived dendritic cells for clinical application in cell factories™. J Immunol Methods. 2002;268:131–140. doi: 10.1016/S0022-1759(02)00189-8.
    1. Kim S, Kim HO, Baek EJ, Choi Y, Kim HS, Lee MG. Monocyte enrichment from leukapheresis products by using the Elutra cell separator. Transfusion. 2007;47:2290–2296. doi: 10.1111/j.1537-2995.2007.01470.x.
    1. Pickl W, Majdic O, Kohl P, Stockl J, Riedl E, Scheinecker C, Bello-Fernandez C, Knapp W. Molecular and functional characteristics of dendritic cells generated from highly purified CD14+ peripheral blood monocytes. J Immunol. 1996;157:3850–3859.
    1. Wong E, Lee S, Hines K, Lee J, Carter C, Kopp W, Bender J, Read E. Development of a closed-system process for clinical-scale generation of DCs: evaluation of two monocyte-enrichment methods and two culture containers. Cytotherapy. 2002;4:65–76. doi: 10.1080/146532402317251545.
    1. Bender A, Sapp M, Schuler G, Steinman RM, Bhardwaj N. Improved methods for the generation of dendritic cells from nonproliferating progenitors in human blood. J Immunol Methods. 1996;196:121–135. doi: 10.1016/0022-1759(96)00079-8.
    1. Nestle FO, Alijagic S, Gilliet M, Sun Y, Grabbe S, Dummer R, Burg G, Schadendorf D. Vaccination of melanoma patients with peptide- or tumorlysate-pulsed dendritic cells. Nat Med. 1998;4:328–332. doi: 10.1038/nm0398-328.
    1. Hernando JJPT, Kübler K, Offergeld R, Schlebusch H, Bauknecht T. Vaccination with autologous tumour antigen-pulsed dendritic cells in advanced gynaecological malignancies: clinical and immunological evaluation of a phase I trial. Cancer Immunol Immunother. 2002;51:45–52. doi: 10.1007/s00262-001-0255-1.
    1. Nagayama H, Sato K, Morishita M, Uchimaru K, Oyaizu N, Inazawa T, Yamasaki T, Enomoto M, Nakaoka T, Nakamura T. et al.Results of a phase I clinical study using autologous tumour lysate-pulsed monocyte-derived mature dendritic cell vaccinations for stage IV malignant melanoma patients combined with low dose interleukin-2. Melanoma Res. 2003;13:521–530. doi: 10.1097/00008390-200310000-00011.
    1. Märten A, Renoth S, Heinicke T, Albers P, Pauli A, Mey U, Caspari R, Flieger D, Hanfland P, von Ruecker A. et al.Allogeneic Dendritic Cells Fused with Tumor Cells: Preclinical Results and Outcome of a Clinical Phase I/II Trial in Patients with Metastatic Renal Cell Carcinoma. Hum Gene Ther. 2003;14:483–494. doi: 10.1089/104303403321467243.
    1. Maier T, Tun-Kyi A, Tassis A, Jungius K-P, Burg G, Dummer R, Nestle FO. Vaccination of patients with cutaneous T-cell lymphoma using intranodal injection of autologous tumor-lysate-pulsed dendritic cells. Blood. 2003;102:2338–2344. doi: 10.1182/blood-2002-08-2455.
    1. Gitlitz BJ, Belldegrun AS, Zisman A, Chao DH, Pantuck AJ, Hinkel A, Mulders P, Moldawer N, Tso C-L, Figlin RA. A Pilot Trial of Tumor Lysate-Loaded Dendritic Cells for the Treatment of Metastatic Renal Cell Carcinoma. J Immunother. 2003;26:412–419. doi: 10.1097/00002371-200309000-00004.
    1. Iwashita Y, Tahara K, Goto S, Sasaki A, Kai S, Seike M, Chen C-L, Kawano K, Kitano S. A phase I study of autologous dendritic cell-based immunotherapy for patients with unresectable primary liver cancer. Cancer Immunol Immunother. 2003;52:155–161.
    1. Pandha HS, John RJ, Hutchinson J, James N, Whelan M, Corbishley C, Dalgleish AG. Dendritic cell immunotherapy for urological cancers using cryopreserved allogeneic tumour lysate-pulsed cells: a phase I/II study. BJU Int. 2004;94:412–418. doi: 10.1111/j.1464-410X.2004.04922.x.
    1. Czerniecki BJ, Koski GK, Koldovsky U, Xu S, Cohen PA, Mick R, Nisenbaum H, Pasha T, Xu M, Fox KR. et al.Targeting HER-2/neu in Early Breast Cancer Development Using Dendritic Cells with Staged Interleukin-12 Burst Secretion. Cancer Res. 2007;67:1842–1852. doi: 10.1158/0008-5472.CAN-06-4038.
    1. Dauer M, Lam V, Arnold H, Junkmann J, Kiefl R, Bauer C, Schnurr M, Endres S, Eigler A. Combined use of toll-like receptor agonists and prostaglandin E2 in the FastDC model: Rapid generation of human monocyte-derived dendritic cells capable of migration and IL-12p70 production. J Immunol Methods. 2008;337:97–105. doi: 10.1016/j.jim.2008.07.003.
    1. Dauer M, Obermaier B, Herten J, Haerle C, Pohl K, Rothenfusser S, Schnurr M, Endres S, Eigler A. Mature Dendritic Cells Derived from Human Monocytes Within 48 Hours: A Novel Strategy for Dendritic Cell Differentiation from Blood Precursors. J Immunol. 2003;170:4069–4076.
    1. Chiang CL-L, Benencia F, Coukos G. Whole tumor antigen vaccines. Semin Immunol. 2010;22:132–143. doi: 10.1016/j.smim.2010.02.004.
    1. Janssen EM, Lemmens EE, Wolfe T, Christen U, von Herrath MG, Schoenberger SP. CD4+ T cells are required for secondary expansion and memory in CD8+ T lymphocytes. Nature. 2003;421:852–856. doi: 10.1038/nature01441.
    1. Shedlock DJ, Shen H. Requirement for CD4 T Cell Help in Generating Functional CD8 T Cell Memory. Science. 2003;300:337–339. doi: 10.1126/science.1082305.
    1. Sun JC, Williams MA, Bevan MJ. CD4+ T cells are required for the maintenance, not programming, of memory CD8+ T cells after acute infection. Nat Immunol. 2004;5:927–933. doi: 10.1038/ni1105.
    1. Chiang CL, Ledermann J, Rad AN, Katz D, Chain B. Hypochlorous acid enhances immunogenicity and uptake of allogeneic ovarian tumor cells by dendritic cells to cross-prime tumor-specific T cells. Cancer Immunol Immunother. 2006;55:1384–1395. doi: 10.1007/s00262-006-0127-9.
    1. Chiang CL, Ledermann JA, Aitkens E, Benjamin E, Katz DR, Chain BM. Oxidation of ovarian epithelial cancer cells by hypochlorous acid enhances immunogenicity and stimulates T cells that recognize autologous primary tumor. Clin Cancer Res. 2008;14:4898–4907. doi: 10.1158/1078-0432.CCR-07-4899.
    1. Prokopowicz ZM, Arce F, Biedron R, Chiang CL-L, Ciszek M, Katz DR, Nowakowska M, Zapotoczny S, Marcinkiewicz J, Chain BM. Hypochlorous Acid: A Natural Adjuvant That Facilitates Antigen Processing, Cross-Priming, and the Induction of Adaptive Immunity. J Immunol. 2010;184:824–835. doi: 10.4049/jimmunol.0902606.
    1. Fisk B, Belvins TL, Wharton JT, Ioannides CG. Identification of an immunodominant peptide of HER-2/neu protooncogene recognized by ovarian tumor-specific cytotoxic T lymphocyte lines. J Exp Med. 1995;181:2109–17. doi: 10.1084/jem.181.6.2109.
    1. Rivoltini L, Kawakami Y, Sakaguchi K, Southwood S, Sette A, Robbins P, Marincola F, Salgaller M, Yannelli J, Appella E. Induction of tumor-reactive CTL from peripheral blood and tumor- infiltrating lymphocytes of melanoma patients by in vitro stimulation with an immunodominant peptide of the human melanoma antigen MART-1. J Immunol. 1995;154:2257–2265.
    1. Romero P, Gervois N, Schneider J, Escobar P, Valmori D, Pannetier C, Steinle A, Wolfel T, Lienard D, Brichard V. et al.Cytolytic T lymphocyte recognition of the immunodominant HLA-A*0201- restricted Melan-A/MART-1 antigenic peptide in melanoma. J Immunol. 1997;159:2366–2374.
    1. Garderet L, Cao H, Salamero J, Vergé V, Tisserand E, Scholl S, Gorin N-C, Lopez M. In Vitro Production of Dendritic Cells from Human Blood Monocytes for Therapeutic Use. J Hematother Stem Cell Res. 2001;10:553–567. doi: 10.1089/15258160152509163.
    1. Erdmann M, Schuler-Thurner B. Towards a Standardized Protocol for the Generation of Monocyte-Derived Dendritic Cell Vaccines. Methods Mol Biol. 2010;595:149–63. doi: 10.1007/978-1-60761-421-0_9.
    1. Morse M, Clay T, Colling K, Lyerly H. Preparation of peptide-loaded dendritic cells for cancer immunotherapy. Mol Biotechnol. 2003;25:95–99. doi: 10.1385/MB:25:1:95.
    1. Loudovaris M, Hansen M, Suen Y, Lee SM, Casing P, Bender JG. Differential Effects of Autologous Serum on CD34+ or Monocyte-Derived Dendritic Cells. J Hematothera Stem Cell Res. 2001;10:569–578. doi: 10.1089/15258160152509172.
    1. Selvaggi TA, Walker RE, Fleisher TA. Development of Antibodies to Fetal Calf Serum With Arthus-Like Reactions in Human Immunodeficiency Virus-Infected Patients Given Syngeneic Lymphocyte Infusions. Blood. 1997;89:776–779.
    1. Mackensen A, Drager R, Schlesier M, Mertelsmann R, Lindemann A. Presence of IgE antibodies to bovine serum albumin in a patient developing anaphylaxis after vaccination with human peptide-pulsed dendritic cells. Cancer Immunol Immunother. 2000;49:152–156. doi: 10.1007/s002620050614.
    1. Tuschong L, Soenen SL, Blaese RM, Candotti F, Muul LM. Immune Response to Fetal Calf Serum by Two Adenosine Deaminase-Deficient Patients After T Cell Gene Therapy. Hum Gene Ther. 2002;13:1605–1610. doi: 10.1089/10430340260201699.
    1. Ratta M, Fagnoni F, Curti A, Vescovini R, Sansoni P, Oliviero B, Fogli M, Ferri E, Della Cuna GR, Tura S. et al.Dendritic cells are functionally defective in multiple myeloma: the role of interleukin-6. Blood. 2002;100:230–237. doi: 10.1182/blood.V100.1.230.
    1. Beckebaum S, Zhang X, Chen X, Yu Z, Frilling A, Dworacki G, Grosse-Wilde H, Erich Broelsch C, 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. doi: 10.1158/1078-0432.CCR-04-0872.
    1. 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 α-Type 1 Polarized Dendritic Cells and Polyinosinic-Polycytidylic Acid Stabilized by Lysine and Carboxymethylcellulose in Patients With Recurrent Malignant Glioma. J Clin Oncol. 2011;29:330–336. doi: 10.1200/JCO.2010.30.7744.
    1. DeBenedette MA, Calderhead DM, Tcherepanova IY, Nicolette CA, Healey DG. Potency of Mature CD40L RNA Electroporated Dendritic Cells Correlates With IL-12 Secretion by Tracking Multifunctional CD8+/CD28+ Cytotoxic T-cell Responses In Vitro. J Immunother. 2011;34:45–57. doi: 10.1097/CJI.0b013e3181fb651a.
    1. Watchmaker PB, Berk E, Muthuswamy R, Mailliard RB, Urban JA, Kirkwood JM, Kalinski P. Independent Regulation of Chemokine Responsiveness and Cytolytic Function versus CD8+ T Cell Expansion by Dendritic Cells. J Immunol. 2010;184:591–597. doi: 10.4049/jimmunol.0902062.
    1. Butterfield LH, Gooding W, Whiteside TL. Development of a Potency Assay for Human Dendritic Cells: IL-12p70 Production. J Immunother. 2008;31:89–100. doi: 10.1097/CJI.0b013e318158fce0.
    1. Lopez MN, Pereda C, Segal G, Munoz L, Aguilera R, Gonzalez FE, Escobar A, Ginesta A, Reyes D, Gonzalez R. et al.Prolonged Survival of Dendritic Cell-Vaccinated Melanoma Patients Correlates With Tumor-Specific Delayed Type IV Hypersensitivity Response and Reduction of Tumor Growth Factor (beta)-Expressing T Cells. J Clin Oncol. 2009;27:945–952. doi: 10.1200/JCO.2008.18.0794.
    1. Lau R, Wang F, Jeffery G, Marty V, Kuniyoshi J, Bade E, Ryback ME, Weber J. Phase I Trial Of Intravenous Peptide-Pulsed Dendritic Cells in Patients With Metastatic Melanoma. J Immunother. 2001;24:66–78. doi: 10.1097/00002371-200101000-00008.
    1. Escobar A, López M, Serrano A, Ramirez M, Pérez C, Aguirre A, González R, Alfaro J, Larrondo M, Fodor M. et al.Dendritic cell immunizations alone or combined with low doses of interleukin-2 induce specific immune responses in melanoma patients. Clin Exp Immunol. 2005;142:555–568.
    1. von Euw E, Barrio M, Furman D, Levy E, Bianchini M, Peguillet I, Lantz O, Vellice A, Kohan A, Chacon M. et al.A phase I clinical study of vaccination of melanoma patients with dendritic cells loaded with allogeneic apoptotic/necrotic melanoma cells. Analysis of toxicity and immune response to the vaccine and of IL-10 -1082 promoter genotype as predictor of disease progression. J Transl Med. 2008;6:6–19. doi: 10.1186/1479-5876-6-6.
    1. Ridolfi R, Petrini M, Fiammenghi L, Stefanelli M, Ridolfi L, Ballardini M, Migliori G, Riccobon A. Improved overall survival in dendritic cell vaccination-induced immunoreactive subgroup of advanced melanoma patients. J Transl Med. 2006;4:36–46. doi: 10.1186/1479-5876-4-36.
    1. Rouas R, Akl H, Fayyad-Kazan H, El Zein N, Badran B, Nowak B, Duvillier H, Martiat P, Lewalle P. Dendritic Cells Generated in Clinical Grade Bags Strongly Differ in Immune Functionality When Compared With Classical DCs Generated in Plates. J Immunother. 2010;33:352–363. doi: 10.1097/CJI.0b013e3181cc266b.
    1. Rapoport AP, Stadtmauer EA, Aqui N, Vogl D, Chew A, Fang H-B, Janofsky S, Yager K, Veloso E, Zheng Z. et al.Rapid Immune Recovery and Graft-versus-Host Disease-like Engraftment Syndrome following Adoptive Transfer of Costimulated Autologous T Cells. Clin Cancer Res. 2009;15:4499–4507. doi: 10.1158/1078-0432.CCR-09-0418.

Source: PubMed

3
Tilaa