Antigen delivery to dendritic cells by poly(propylene sulfide) nanoparticles with disulfide conjugated peptides: Cross-presentation and T cell activation

Vaccine ◽  
2010 ◽  
Vol 28 (50) ◽  
pp. 7897-7906 ◽  
Author(s):  
Sachiko Hirosue ◽  
Iraklis C. Kourtis ◽  
André J. van der Vlies ◽  
Jeffrey A. Hubbell ◽  
Melody A. Swartz
2021 ◽  
Vol 478 (22) ◽  
pp. 3999-4004
Author(s):  
Lawrence P. Kane

Tim-3 is a transmembrane protein that is highly expressed on subsets of chronically stimulated CD4+ helper and CD8+ cytotoxic T cells, with more transient expression during acute activation and infection. Tim-3 is also constitutively expressed by multiple types of myeloid cells. Like other TIM family members, Tim-3 can bind to phosphatidylserine displayed by apoptotic cells, and this interaction has been shown to mediate uptake of such cells by dendritic cells and cross-presentation of antigens to CD8+ T cells. In contrast, how the recognition of PS by Tim-3 might regulate the function of Tim-3+ T cells is not known. In their recent paper, Lemmon and colleagues demonstrate for the first time that recognition of PS by Tim-3 leads to enhanced T cell activation.


2001 ◽  
Vol 194 (6) ◽  
pp. 769-780 ◽  
Author(s):  
Daniel Hawiger ◽  
Kayo Inaba ◽  
Yair Dorsett ◽  
Ming Guo ◽  
Karsten Mahnke ◽  
...  

Dendritic cells (DCs) have the capacity to initiate immune responses, but it has been postulated that they may also be involved in inducing peripheral tolerance. To examine the function of DCs in the steady state we devised an antigen delivery system targeting these specialized antigen presenting cells in vivo using a monoclonal antibody to a DC-restricted endocytic receptor, DEC-205. Our experiments show that this route of antigen delivery to DCs is several orders of magnitude more efficient than free peptide in complete Freund's adjuvant (CFA) in inducing T cell activation and cell division. However, T cells activated by antigen delivered to DCs are not polarized to produce T helper type 1 cytokine interferon γ and the activation response is not sustained. Within 7 d the number of antigen-specific T cells is severely reduced, and the residual T cells become unresponsive to systemic challenge with antigen in CFA. Coinjection of the DC-targeted antigen and anti-CD40 agonistic antibody changes the outcome from tolerance to prolonged T cell activation and immunity. We conclude that in the absence of additional stimuli DCs induce transient antigen-specific T cell activation followed by T cell deletion and unresponsiveness.


2020 ◽  
Vol 8 (Suppl 2) ◽  
pp. A38.1-A38
Author(s):  
S Schmitt ◽  
A Lohner ◽  
K Deiser ◽  
A Maiser ◽  
M Rothe ◽  
...  

BackgroundDendritic cells (DCs) are antigen-presenting cells that induce antigen-specific T-cell responses. Therefore, they are used as tools and targets for anti-tumor vaccination. In contrast to T-cell based immunotherapies, that are often limited to surface antigens, DC-based vaccination strategies open up new therapeutic options by utilizing highly abundant intracellular tumor antigens as a target source. Among those, recent interest has been focused on the identification of neoantigens derived from tumor-specific mutations. Especially mutated Nucleophosmin 1 (ΔNPM1) is a considered candidate for targeted therapy in acute myeloid leukemia (AML). We developed a multifunctional antibody construct consisting of a peptide domain including a variable T-cell epitope that is fused to an αCD40 single chain variable fragment (scFv) with agonistic function to target and activate dendritic cells in vivo. To potentiate therapeutic efficacy, toll-like receptor (TLR) agonists can be attached as co-stimulatory domains, thereby aiming to enhance cross-presentation of conjugated (neo)antigens to CD8+ T cells.Materials and MethodsFlow cytometry and microscopy-based binding and internalization experiments were performed using monocyte-derived dendritic cells (moDCs). Upregulation of surface markers (CD80, CD83, CD86, HLA-DR) as well as cytokine secretion (IL-6 and IL-12) indicated DC maturation. To validate peptide processing and presentation, moDCs were co-cultured with autologous as well as allogeneic T cells. IFN-γ and TNF-α secretion served as a readout for T-cell activation, peptide-MHC multimer staining for T-cell proliferation.ResultsFor proof-of-principle experiments, the multispecific antibody derivative was developed by fusing the αCD40 scFv to a cytomegalovirus (CMV)-specific peptide. The αCD40.CMV construct bound CD40 agonistically and showed efficient internalization into early endosomal compartments on immature moDCs. In co-cultures of immature and mature moDCs with autologous or allogeneic T cells, αCD40.CMV induced a significantly increased T-cell activation and proliferation compared to the control. The co-administration of αCD40.CMV with various TLR agonists as vaccine adjuvants resulted in a significant upregulation of DC maturation markers in comparison to αCD40.CMV only. Interestingly, not all adjuvants were able to enhance the T-cell response. To translate this principle to the AML setting, the CMV peptide sequence was replaced with the ΔNPM1-derived and HLA-A*02:01-binding neoantigen CLAVEEVSL. Cross-presentation to CD8+ T cells transduced with a ΔNPM1-specific T-cell receptor was proven by IFN-γ and TNF-α secretion in co-cultures with moDCs that have been pre-incubated with αCD40.ΔNPM1. The optimal vaccine adjuvant has yet to be identified.ConclusionsWe successfully demonstrated the development of a multifunctional antibody construct that specifically targets and stimulates DCs by an agonistic αCD40 scFv. It simultaneously delivers a T cell-specific peptide with a vaccine adjuvant to induce an efficient T-cell response. As neoantigens are promising targets and under intense investigaton, the αCD40.ΔNPM1 fusion protein is of high therapeutic interest. Thus, our approach displays a promising DC vaccination option for the treatment of AML.Disclosure InformationS. Schmitt: None. A. Lohner: None. K. Deiser: None. A. Maiser: None. M. Rothe: None. C. Augsberger: None. A. Moosmann: None. H. Leonhardt: None. N. Fenn: None. M. Griffioen: None. K. Hopfner: None. M. Subklewe: None.


Vaccines ◽  
2021 ◽  
Vol 9 (4) ◽  
pp. 409
Author(s):  
Enrique Gómez Alcaide ◽  
Sinduya Krishnarajah ◽  
Fabian Junker

Despite significant recent improvements in the field of immunotherapy, cancer remains a heavy burden on patients and healthcare systems. In recent years, immunotherapies have led to remarkable strides in treating certain cancers. However, despite the success of checkpoint inhibitors and the advent of cellular therapies, novel strategies need to be explored to (1) improve treatment in patients where these approaches fail and (2) make such treatments widely and financially accessible. Vaccines based on tumor antigens (Ag) have emerged as an innovative strategy with the potential to address these areas. Here, we review the fundamental aspects relevant for the development of cancer vaccines and the critical role of dendritic cells (DCs) in this process. We first offer a general overview of DC biology and routes of Ag presentation eliciting effective T cell-mediated immune responses. We then present new therapeutic avenues specifically targeting Fc gamma receptors (FcγR) as a means to deliver antigen selectively to DCs and its effects on T-cell activation. We present an overview of the mechanistic aspects of FcγR-mediated DC targeting, as well as potential tumor vaccination strategies based on preclinical and translational studies. In particular, we highlight recent developments in the field of recombinant immune complex-like large molecules and their potential for DC-mediated tumor vaccination in the clinic. These findings go beyond cancer research and may be of relevance for other disease areas that could benefit from FcγR-targeted antigen delivery, such as autoimmunity and infectious diseases.


Cancers ◽  
2021 ◽  
Vol 13 (15) ◽  
pp. 3818
Author(s):  
Maud Plantinga ◽  
Denise A. M. H. van den Beemt ◽  
Ester Dünnebach ◽  
Stefan Nierkens

Induction of long-lasting immunity by dendritic cells (DCs) makes them attractive candidates for anti-tumor vaccination. Although DC vaccinations are generally considered safe, clinical responses remain inconsistent in clinical trials. This initiated studies to identify subsets of DCs with superior capabilities to induce effective and memory anti-tumor responses. The use of primary DCs has been suggested to overcome the functional limitations of ex vivo monocyte-derived DCs (moDC). The ontogeny of primary DCs has recently been revised by the introduction of DC3, which phenotypically resembles conventional (c)DC2 as well as moDC. Previously, we developed a protocol to generate cDC2s from cord blood (CB)-derived stem cells via a CD115-expressing precursor. Here, we performed index sorting and single-cell RNA-sequencing to define the heterogeneity of in vitro developed DC precursors and identified CD14+CD115+ expressing cells that develop into CD1c++DCs and the remainder cells brought about CD123+DCs, as well as assessed their potency. The maturation status and T-cell activation potential were assessed using flow cytometry. CD123+DCs were specifically prone to take up antigens but only modestly activated T-cells. In contrast, CD1c++ are highly mature and specialized in both naïve as well as antigen-experienced T-cell activation. These findings show in vitro functional diversity between cord blood stem cell-derived CD123+DC and CD1c++DCs and may advance the efficiency of DC-based vaccines.


Oncogene ◽  
2021 ◽  
Author(s):  
Francesca Alfei ◽  
Ping-Chih Ho ◽  
Wan-Lin Lo

AbstractThe exploitation of T cell-based immunotherapies and immune checkpoint blockade for cancer treatment has dramatically shifted oncological treatment paradigms and broadened the horizons of cancer immunology. Dendritic cells have emerged as the critical tailors of T cell immune responses, which initiate and coordinate anti-tumor immunity. Importantly, genetic alterations in cancer cells, cytokines and chemokines produced by cancer and stromal cells, and the process of tumor microenvironmental regulation can compromise dendritic cell–T cell cross-talk, thereby disrupting anti-tumor T cell responses. This review summarizes how T cell activation is controlled by dendritic cells and how the tumor microenvironment alters dendritic cell properties in the context of the anti-tumor immune cycle. Furthermore, we will highlight therapeutic options for tailoring dendritic cell-mediated decision-making in T cells for cancer treatment.


Sign in / Sign up

Export Citation Format

Share Document