Comparison of Antigen-Loading Strategies for DC-Based Immunotherapy of B-CLL.

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 4821-4821
Author(s):  
Hakan Mellstedt ◽  
Parviz Kokhaei ◽  
Anders Osterborg ◽  
Aniruddha Choudhury

Abstract The slow indolent nature of B-CLL makes it eminently suitable for immune-based treatment strategies. Currently, there exist very few CLL-associated, defined antigen which can be used in vaccination approaches. The use of whole tumor preparations in conjunction with dendritic cells (DC) as cellular adjuvants presents an alternative method for generating therapeutic T-cell responses in CLL patients. We have examined various strategies of loading DC with whole tumor antigens; viz. tumor-DC hybrids, endocytosed apoptotic bodies, RNA or lysate from B-CLL cells. Dendritic cells were generated in vitro, using GM-CSF and IL-4 from immunomagnetically purified, CD14+ monocyte precursors obtained from the peripheral blood of B-CLL patients. The immature DC were loaded with whole tumor antigen using the above methods and matured using TNFα. The tumor antigen-loaded DC were compared for their ability to stimulate autologous T-cell responses. Among all the antigen-loading methods tested, DC that had endocytosed apoptotic bodies (Apo-DC) consistently generated the greatest numbers and magnitude of reactive T-cells as quantified in proliferation and ELISPOT assays. RT-PCR analysis for cytokine mRNA revealed that T-cells stimulated by Apo-DC resulted in an immune response that was almost entirely of the TH1 type as manifested by the production of mRNA for IFN-γ and TNFα. In contrast, other methods of loading antigen resulted in a mixed TH1-TH2 population with varying amounts of TH2 cytokines like IL-4 and IL-10. In a separate series of experiments we compared the T-cell stimulatory capacities of cryopreserved and thawed, with fresh antigen-loaded DC. The results of these studies are essential for the development of a protocol for clinical therapy of B-CLL patients using Apo-DC. Our results indicated that cryopreserved DC could be recovered with high viability. A comparison of functional abilities demonstrated that no significant differences in T-cell stimulatory capacity between cryopreserved and fresh Apo-DC could be noted over a wide variety of immunological assays. Cumulatively, our results suggest that Apo-DC may be a suitable vaccine candidate for immunotherapy of B-CLL patients.

2020 ◽  
Vol 219 ◽  
pp. 46-53
Author(s):  
Ondrej Palata ◽  
Nada Podzimkova Hradilova ◽  
Dagmar Mysiková ◽  
Beata Kutna ◽  
Hana Mrazkova ◽  
...  

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 1354-1354
Author(s):  
Annkristin Heine ◽  
Tobias Holderried ◽  
Frank Grünebach ◽  
Silke Appel ◽  
Markus M. Weck ◽  
...  

Abstract Transfection of dendritic cells (DC) with in vitro transcribed RNA was shown to be a highly efficient method to generate antigen specific T cells, probably due to the induction of a polyclonal T cell response directed against multiple antigens presented on different HLA allels. However, the experimental evidence of this assumption remains to be demonstrated. To accomplish this, we used monocyte derived DC that were electroporated with RNA coding for the CMV pp65 antigen. The induction and expansion of antigen specific CD8+ and CD4+ T cells was assessed using a pannel of peptides derived from this antigen and presented on HLA-A2, -A1, -A11, -A24, -B35 and -B7 in IFN-g ELISPOT, 51Cr-release and proliferation assays. Autologous DC generated from CMV positive healthy donors were pulsed with peptides or transfected with pp65 RNA and utilized as stimulators. Autologous purified CD8+ and CD4+ lymphocytes were used as effector cells. By applying this approach we found that transfection of DC with pp65 RNA induces an expansion of polyclonal CD8+ mediated T cell responses that recognized peptide antigens presented on different HLA molecules. These in vitro generated cytotoxic T cells were able to efficiently lyse DC pulsed with pp65 derived peptides or transfected with the cognate RNA in an antigen specific manner after several in vitro restimulations. Furthermore, this experimental approach allowed the identification of the immunodominace of T cell epitopes presented upon RNA transfection. The HLA-2 directed responses were more pronounced as compared to the HLA-A1, -A11, -A24 or -B35 allels. In contrast, in 7 out of 7 HLA-A2 and HLA-B7 positive donors B7-peptides elicited a stronger T cell response than the A2-peptide, indicating the immunodominance of HLA-B7 epitopes. Interestingly, transfection of DC with pp65 RNA resulted in the induction of CD4+ antigen specific T cells that produced IFN-g and proliferated upon stimulation with transfected DC. In the next set of experiments we analyzed the possible induction of CMV specific T cells that recognize epitopes deduced from different antigens. Co-transfection of DC with a mixture of RNAs coding for the CMV pp65 and IE1 antigens elicited polyclonal T lymphocytes specific for peptides derived from both antigens. More importantly, polyclonal cytotoxic T cells could be elicited in peripheral blood of 2 out of 3 CMV negative donors demonstrating the efficiency of this approach. Our results demonstrate that DC transfected with RNA can elicit polyclonal T cell responses and have implications for the development of immunotherapeutic strategies to target viral or tumor associated antigens.


Blood ◽  
2005 ◽  
Vol 106 (1) ◽  
pp. 216-223 ◽  
Author(s):  
Elodie Segura ◽  
Carole Nicco ◽  
Bérangère Lombard ◽  
Philippe Véron ◽  
Graça Raposo ◽  
...  

Exosomes are secreted vesicles formed in late endocytic compartments. Immature dendritic cells (DCs) secrete exosomes, which transfer functional major histocompatibility complex (MHC)–peptide complexes to other DCs. Since immature and mature DCs induce different functional T-cell responses (ie, tolerance versus priming), we asked whether DC maturation also influenced the priming abilities of their exosomes. We show that exosomes secreted by lipopolysaccharide (LPS)–treated mature DCs are 50- to 100-fold more potent to induce antigen-specific T-cell activation in vitro than exosomes from immature DCs. In vitro, exosomes from mature DCs transfer to B lymphocytes the ability to prime naive T cells. In vivo, only mature exosomes trigger effector T-cell responses, leading to fast skin graft rejection. Proteomic and biochemical analyses revealed that mature exosomes are enriched in MHC class II, B7.2, intercellular adhesion molecule 1 (ICAM-1), and bear little milk-fat globule–epidermal growth factor–factor VIII (MFG-E8) as compared with immature exosomes. Functional analysis using DC-derived exosomes from knock-out mice showed that MHC class II and ICAM-1 are required for mature exosomes to prime naive T cells, whereas B7.2 and MFG-E8 are dispensable. Therefore, changes in protein composition and priming abilities of exosomes reflect the maturation signals received by DCs.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2025-2025
Author(s):  
Soley Thordardottir ◽  
Hangalapura Basav N. ◽  
Tim Hutten ◽  
Marta Cossu ◽  
Jan Spanholtz ◽  
...  

Abstract The prominent role of dendritic cells (DCs) in T cell activation is the rational for DC-based immunotherapy of cancer and infectious diseases. In cancer, DC therapy aims to induce tumor-specific effector T cell responses that can reduce or eliminate the tumor, and to develop immunological memory to control tumor relapse. So far, the vast majority of DC vaccination studies have been performed with DCs differentiated from monocytes (Mo-DCs) that are loaded with tumor-associated antigens (TAAs) or minor histocompatibility antigens (MiHA). This strategy has been reported to induce the expansion of antigen-specific CD4+ and/or CD8+ T cells in the majority of patients, however only a fraction of the patients develop clinical responses. Strategies to improve the potency of DC-based vaccines are to increase the stimulatory and migratory capacity of Mo-DCs, or to use alternative DC subtypes, such as naturally circulating plasmacytoid DCs (pDCs), BDCA1+ myeloid DCs (mDCs) or BDCA3+ mDCs. These DC subsets are potent inducers of antigen-specific T cell responses, and are therefore attractive cells to exploit for DC-based therapy. However, since their frequency in blood is very low, it is a challenge to obtain high enough numbers for immunotherapy. It would be advantageous if DCs, which are phenotypically and functionally similar to blood pDCs and mDCs, could be generated from CD34+ hematopoietic progenitor cells (HPCs). Interestingly, recent findings have indicated that the aryl hydrocarbon receptor (AhR) not only regulates toxic effects of environmental contaminants, but also plays a role in modulating hematopoiesis and the immune system. For instance, it has been reported that StemRegenin 1 (SR1), a small molecule inhibitor of AhR, promotes the ex vivo expansion of human CD34+ HPCs that are able to effectively engraft immunodeficient mice. Furthermore, differentiation of Langerhans cells and monocytes in vitro from HPCs can be inhibited by the addition of the AhR agonist VAF347. In light of these data, we investigated if we could generate DC subsets from CD34+ HPCs by supplementing SR1. Therefore, we cultured CD34+ HPCs in medium containing SCF, Flt3L, IL-6, TPO supplemented with 1 μM SR1 or DMSO as control. Interestingly, addition of SR1 explicitly promoted the emergence of pDCs (CD11c-HLA-DR+CD123hiBDCA2+BDCA4+ cells), BDCA1+ mDCs (Lin1-HLA-DR+BDCA1+BDCA3- cells) and BDCA3+ mDCs (Lin1-HLA-DR+BDCA1-BDCA3+ cells). After three weeks of culture, the frequency of these DC subsets was significantly higher in cultures with SR1 compared to control conditions; 2.9% vs. 0.04% for pDCs, 4.6% vs. 0.5% for BDCA1+ mDCs and 1.1% vs. 0.1% for BDCA3+ mDCs (n=3-5 donors). The average yield after three weeks of culture with SR1 starting from 105 CD34+ UCB cells was 3.8x106 pDCs, 5.3x106 BDCA1+ mDCs and 1.2x106 BDCA3+ mDCs (n=3-5 donors). Furthermore, SR1 also promoted the differentiation of DC subsets from CD34+ cells obtained from peripheral blood of G-CSF-mobilized donors. The average frequency of DCs in these SR1-cultures was 4.7%, 3.8% and 0.9% for pDCs, BDCA1+ and BDCA3+ mDCs, respectively (n=3 donors), which is comparable to the frequency obtained from UCB CD34+ cells. But the expansion potential of G-CSF-mobilized blood CD34+ HPCs was lower than that of UCB CD34+ cells, resulting in average DC yields of 0.6x106, 0.5x106 and 0.1x106 from 105 CD34+ cells (n=3). Flow cytometry analysis demonstrated that the SR1-induced pDCs and mDCs are phenotypically comparable to their naturally occurring counterpart in blood. Furthermore, the ex vivo-generated pDCs potently responded to stimulation with TLR7 and TLR9 ligands by secreting high amounts of IFN-α and upregulating CD83, CD80, CD86 and CCR7. The HPC-mDC subsets also upregulate CD80 and CD83 upon TLR3, TLR4 or TLR7/8 ligation. Finally, both the ex vivo-generated pDCs and mDCs induced potent allogeneic T cell responses and activated CD8+ effector T cells against hematopoietic-restricted MiHA. These findings demonstrate that our SR1 culture system not only allows detailed study of DC differentiation and molecular regulations in vitro, but it also offers the opportunity to evaluate the in vivo efficacy of cultured DC subsets upon vaccination into patients with cancer and viral infections. Disclosures: Spanholtz: Glycostem Therapeutics: Employment.


2008 ◽  
Vol 82 (7) ◽  
pp. 3561-3573 ◽  
Author(s):  
Ellen R. Van Gulck ◽  
Guido Vanham ◽  
Leo Heyndrickx ◽  
Sandra Coppens ◽  
Katleen Vereecken ◽  
...  

ABSTRACT Developing an immunotherapy to keep human immunodeficiency virus type 1 (HIV-1) replication suppressed while discontinuing highly active antiretroviral therapy (HAART) is an important challenge. In the present work, we evaluated in vitro whether dendritic cells (DC) electroporated with gag mRNA can induce HIV-specific responses in T cells from chronically infected subjects. Monocyte-derived DC, from therapy-naïve and HAART-treated HIV-1-seropositive subjects, that were electroporated with consensus codon-optimized HxB2 gag mRNA efficiently expanded T cells, secreting gamma interferon (IFN-γ) and interleukin 2 (IL-2), as well as other cytokines and perforin, upon restimulation with a pool of overlapping Gag peptides. The functional expansion levels after 1 week of stimulation were comparable in T cells from HAART-treated and treatment-naïve patients and involved both CD4+ and CD8+ T cells, with evidence of bifunctionality in T cells. Epitope mapping of p24 showed that stimulated T cells had a broadened response toward previously nondescribed epitopes. DC, from HAART-treated subjects, that were electroporated with autologous proviral gag mRNA equally efficiently expanded HIV-specific T cells. Regulatory T cells did not prevent the induction of effector T cells in this system, whereas the blocking of PD-L1 slightly increased the induction of T-cell responses. This paper shows that DC, loaded with consensus or autologous gag mRNA, expand HIV-specific T-cell responses in vitro.


1989 ◽  
Vol 169 (4) ◽  
pp. 1255-1264 ◽  
Author(s):  
S E Macatonia ◽  
P M Taylor ◽  
S C Knight ◽  
B A Askonas

We used well-gassed hanging drop (20 microliters) cultures with high concentrations of purified T cells from normal BALB/c mice to examine whether dendritic cells (DC) can induce primary antiviral proliferative T cell responses and generate virus-specific CTL. We found that DC exposed to infectious influenza virus in vitro or in vivo in small numbers (0.1-1%) resulted in strong proliferation of responder T cells within 3 d, and this was strongly inhibited by antibodies to class II MHC molecules. In addition, in 5-d cultures, the influenza-treated DC generated CTL specifically able to lyse influenza-infected syngeneic target cells bearing MHC class I antigens. The most potent nucleoprotein (NP) epitope recognized by BALB/c CTL is peptide 147-158 (Arg156-) and influenza-infected DC in vitro stimulated CTL recognizing this peptide, thus mimicking the response in mice primed by intranasal influenza infection. We also induced T cell proliferation and virus-specific CTL in cultures of normal T cells by stimulating with DC pulsed with the natural NP sequence 147-158 or the potent peptide 147-158 (Arg156-). Small numbers of peritoneal exudate cells, after activation with Con A to produce class II MHC expression and after removal of DC with a specific mAb (33DI), did not lead to primary CTL generation but initiated secondary stimulation in vitro. Our results using the hanging drop culture method and DC as APC have implications for studying the T cell repertoire for viral components in humans without the necessity of previous immunization.


Blood ◽  
2011 ◽  
Vol 117 (24) ◽  
pp. 6542-6551 ◽  
Author(s):  
Jeffrey S. Pufnock ◽  
Melinda Cigal ◽  
Lisa S. Rolczynski ◽  
Erica Andersen-Nissen ◽  
Mathias Wolfl ◽  
...  

Abstract TLRs expressed on dendritic cells (DCs) differentially activate DCs when activated alone or in combination, inducing distinct cytokines and costimulatory molecules that influence T-cell responses. Defining the requirements of DCs to program T cells during priming to become memory rather than effector cells could enhance vaccine development. We used an in vitro system to assess the influence of DC maturation signals on priming naive human CD8+ T cells. Maturation of DCs with lipopolysaccharide (LPS; TLR4) concurrently with R848 (TLR7/8) induced a heterogeneous population of DCs that produced high levels of IL12 p70. Compared with DCs matured with LPS or R848 alone, the DC population matured with both adjuvants primed CD8+ T-cell responses containing an increased proportion of antigen-specific T cells retaining CD28 expression. Priming with a homogenous subpopulation of LPS/R848–matured DCs that were CD83Hi/CD80+/CD86+ reduced this CD28+ subpopulation and induced T cells with an effector cytokine signature, whereas priming with the less mature subpopulations of DCs resulted in minimal T-cell expansion. These results suggest that TLR4 and TLR7/8 signals together induce DCs with fully mature and less mature phenotypes that are both required to more efficiently prime CD8+ T cells with qualities associated with memory T cells.


2021 ◽  
Vol 12 ◽  
Author(s):  
Sara Ness ◽  
Shiming Lin ◽  
John R. Gordon

Dendritic cells (DC) are antigen-presenting cells that can communicate with T cells both directly and indirectly, regulating our adaptive immune responses against environmental and self-antigens. Under some microenvironmental conditions DC develop into anti-inflammatory cells which can induce immunologic tolerance. A substantial body of literature has confirmed that in such settings regulatory DC (DCreg) induce T cell tolerance by suppression of effector T cells as well as by induction of regulatory T cells (Treg). Many in vitro studies have been undertaken with human DCreg which, as a surrogate marker of antigen-specific tolerogenic potential, only poorly activate allogeneic T cell responses. Fewer studies have addressed the abilities of, or mechanisms by which these human DCreg suppress autologous effector T cell responses and induce infectious tolerance-promoting Treg responses. Moreover, the agents and properties that render DC as tolerogenic are many and varied, as are the cells’ relative regulatory activities and mechanisms of action. Herein we review the most current human and, where gaps exist, murine DCreg literature that addresses the cellular and molecular biology of these cells. We also address the clinical relevance of human DCreg, highlighting the outcomes of pre-clinical mouse and non-human primate studies and early phase clinical trials that have been undertaken, as well as the impact of innate immune receptors and symbiotic microbial signaling on the immunobiology of DCreg.


2021 ◽  
Vol 11 ◽  
Author(s):  
Fu Long Nan ◽  
Wei Zheng ◽  
Wen Long Nan ◽  
Tong Yu ◽  
Chang Zhan Xie ◽  
...  

Newcastle disease virus (NDV) infects poultry and antagonizes host immunity via several mechanisms. Dendritic cells (DCs) are characterized as specialized antigen presenting cells, bridging innate and adaptive immunity and regulating host resistance to viral invasion. However, there is little specific knowledge of the role of DCs in NDV infection. In this study, the representative NDV lentogenic strain LaSota was used to explore whether murine bone marrow derived DCs mature following infection. We examined surface molecule expression and cytokine release from DCs as well as proliferation and activation of T cells in vivo and in vitro in the context of NDV. The results demonstrated that infection with lentogenic strain LaSota induced a phenotypic maturation of immature DCs (imDCs), which actually led to curtailed T cell responses. Upon infection, the phenotypic maturation of DCs was reflected by markedly enhanced MHC and costimulatory molecule expression and secretion of proinflammatory cytokines. Nevertheless, NDV-infected DCs produced the anti-inflammatory cytokine IL-10 and attenuated T cell proliferation, inducing Th2-biased responses. Therefore, our study reveals a novel understanding that DCs are phenotypically mature but dysfunctional in priming T cell responses during NDV infection.


Sign in / Sign up

Export Citation Format

Share Document