scholarly journals Liposomal Nanovaccine Containing α-Galactosylceramide and Ganglioside GM3 Stimulates Robust CD8+ T Cell Responses via CD169+ Macrophages and cDC1

Vaccines ◽  
2021 ◽  
Vol 9 (1) ◽  
pp. 56
Author(s):  
Joanna Grabowska ◽  
Dorian A. Stolk ◽  
Maarten K. Nijen Twilhaar ◽  
Martino Ambrosini ◽  
Gert Storm ◽  
...  

Successful anti-cancer vaccines aim to prime and reinvigorate cytotoxic T cells and should therefore comprise a potent antigen and adjuvant. Antigen targeting to splenic CD169+ macrophages was shown to induce robust CD8+ T cell responses via antigen transfer to cDC1. Interestingly, CD169+ macrophages can also activate type I natural killer T-cells (NKT). NKT activation via ligands such as α-galactosylceramide (αGC) serve as natural adjuvants through dendritic cell activation. Here, we incorporated ganglioside GM3 and αGC in ovalbumin (OVA) protein-containing liposomes to achieve both CD169+ targeting and superior DC activation. The systemic delivery of GM3-αGC-OVA liposomes resulted in specific uptake by splenic CD169+ macrophages, stimulated strong IFNγ production by NKT and NK cells and coincided with the maturation of cDC1 and significant IL-12 production. Strikingly, superior induction of OVA-specific CD8+ T cells was detected after immunization with GM3-αGC-OVA liposomes. CD8+ T cell activation, but not B cell activation, was dependent on CD169+ macrophages and cDC1, while activation of NKT and NK cells were partially mediated by cDC1. In summary, GM3-αGC antigen-containing liposomes are a potent vaccination platform that promotes the interaction between different immune cell populations, resulting in strong adaptive immunity and therefore emerge as a promising anti-cancer vaccination strategy.

2021 ◽  
Author(s):  
◽  
Taryn Louise Osmond

<p>Splenic CD8α⁺ dendritic cells (DCs) have been described as key antigen presenting cells for the induction of CD8⁺ T cell responses to circulating antigen. This is through a heightened capacity to acquire and present the antigens via the process of cross-presentation, expression of high levels of the co-stimulatory and adhesion molecules required to stimulate CD8⁺ T cells, and the capacity to release high levels of the cytokines required to drive differentiation of CD8⁺ T cells into cytotoxic T lymphocytes (CTLs). However, recent research has indicated that the splenic CD8α⁺ DC population is more heterogeneous than originally thought. A previous study from my own laboratory suggested that a population of CD8α⁺ DCs that express the c-type lectin langerin primarily possess the heightened functions previously attributed to the total CD8α⁺ population. Therefore, the aim of this thesis research was to explore this subset of DCs in more detail, with specific emphasis on gaining mechanistic insight into their ability to elicit CD8⁺ T cell responses to circulating proteins. In the first section of this thesis, the hypothesis that the splenic langerin⁺ CD8α⁺ DCs were the critical subset involved in the induction of strong systemic CD8⁺ T cell responses to circulating antigen was tested in detail. This was examined using a genetically modified mouse model in which langerin-expressing cells could be easily identified and/or specifically depleted. It was first shown that the induction of CD8⁺ T cell responses to the model antigen ovalbumin was dependent on entry into the spleen in the presence of appropriate stimulation, which in these studies was provided by agonists for the toll-like receptors (TLRs) and/or signals from innate-like lymphocytes called natural killer T (NKT) cells. The primary targets for these signals were shown to be splenic langerin⁺ CD8α⁺ DCs, as CD8⁺ T cell responses were significantly reduced in hosts depleted of these cells within the spleen. Furthermore, agonists for TLRs that were not expressed by langerin⁺ CD8α⁺ DCs failed to enhance T cell responses. The langerin⁺ CD8α⁺ DCs were shown to be located in the marginal zone of the spleen, where they could readily screen the blood for antigens, and their function was critical to the induction of CD8⁺ T cell responses within six hours of antigen delivery. Interestingly, other local langerin-negative antigen presenting cells (APCs) were shown to be capable of cross-presentation, but with significantly reduced capacity to prime CD8⁺ T cell responses. Therefore, in the second section of this thesis the hypothesis that the langerin-negative APCs were capable of contributing to CD8⁺ T cell responses with appropriately timed stimuli was investigated. One of the downstream effects of inducing NKT cell activation at the time of priming was shown to be the “pre-conditioning” of langerin-negative DCs, allowing them to respond strongly to subsequent TLR ligation. Using SiglecH-DTR mice, it was shown that plasmacytoid DCs (which are langerin-negative) were pre-conditioned by NKT cell activation, allowing them to respond more actively to the delayed TLR stimulation by producing significantly enhanced levels of IFN-α. This factor was also potentially responsible for “feeding back” to the CD8α⁺ DCs (including langerin-expressing CD8α⁺ DCs), to enhance their function, as indicated by increases in cytokine production. Significantly, the major langerin-negative DC populations, defined as CD8α⁻ DCs, were pre-conditioned to have an enhanced cytokine release response to subsequent stimulation through TLR7, a receptor not expressed by langerin-positive DCs. This enhanced ability to respond to TLR7 ligation permitted these langerin-negative APCs to contribute to increased CD8⁺ T cell accumulation, with enhanced functional activity. Importantly, the CD8⁺ T cell response induced remained significantly dependent on initial cross-priming by langerin⁺ CD8α⁺ DCs, and it was only through pre-conditioning that langerinnegative APCs could contribute to enhancing the T cell response. In the third section of this thesis, the hypothesis that the CD8⁺ T cell responses generated in the presence of langerin⁺ CD8α⁺ DCs were phenotypically and functionally distinct from those responses generated in their absence was tested. No obvious differences were seen in CD8⁺ T cell homing, memory phenotype, restimulatory capacity, and expression of key molecules involved in metabolic function, survival and cytolytic function. However, in vivo cytotoxic function several weeks after priming was comparable, suggesting that this function was not related to initial burst size, providing some evidence of difference in function between CD8⁺ T cells primed in the presence or absence of langerin⁺ CD8α⁺ DCs. In summary, the splenic langerin⁺ CD8α⁺ DCs are the major subset responsible for cross-priming CD8⁺ T cell responses to circulating antigen, and for interpreting multiple stimulatory signals for enhancing the response. However, effective CD8⁺ T cell responses can be generated in their absence, particularly when antigens are provided in the context of appropriately temporally phased stimuli.</p>


2020 ◽  
Vol 3 (Supplement_1) ◽  
pp. 139-140
Author(s):  
B K Hardman ◽  
L C Osborne

Abstract Background Human Norovirus infection is the most common viral cause of gastroenteritis globally and the second most reported viral infection in Canada after the common cold. Most infections are acute, symptomatic, and rapidly cleared but some cases persist asymptomatically or induce post-infectious irritable bowel syndrome. Despite the global burden of these infections, no vaccine to prevent disease exists nor is the mechanism for persistence understood. MNV-CW3 and MNV-CR6 are murine noroviruses which demonstrate distinct biological behaviors that correlate with differential quantity and quality of antiviral CD8+ T cell responses. MNV-CW3 causes acute systemic infections initiated in the small intestine and cleared by day 8 due to a robust antiviral CD8+ T cell response. In contrast, MNV-CR6 causes chronic infections localized to the colonic intestinal epithelium and induces fewer antiviral CD8+ T cells with reduced effector molecule expression. Aims This research interrogates the mechanisms underlying strain-specific differential antiviral CD8+ T cell responses. Methods At days 3, 4, 5 and 8 post-infection, the phenotype and quantity of adoptively transferred MNV specific CD8+ T cells in the spleen, mesenteric lymph node (MLN), and the small and large intestine are analyzed by flow cytometry. Concurrently, immunofluorescent microscopy is used to determine whether CD8+ T cells are broadly disseminated throughout the intestines or localize in acute clusters of antiviral response. Combining these complementary techniques provides novel insight into mechanisms governing intestinal antiviral T cell responses. Results Activated MNV-specific CD8 T cells first accumulate in the MLN following oral infection with both MNV-CW3 and CR6, suggesting this is the site of immune activation. Supporting this hypothesis, preliminary data indicates that preventing T cell egress from activation sites by treatment with the S1PR1 agonist FTY720 leads to an enrichment of activated CD8+ T cells in the MLN following CW3 infection. Notably, the earliest stages of CD8+ T cell activation to CR6 infection is delayed compared to that elicited by CW3. Furthermore, at the peak of CD8+ T cell expansion (day 8 post-infection), CR6-elicited CD8+ T cells preferentially develop into short-lived effector populations rather than memory precursor populations. Conclusions These data reveal previously unknown differences in early events in CD8+ T cell activation following infection with two highly related viral strains that correlate with long-lasting effects on T cell differentiation and function. We are currently investigating the hypothesis that MNV-CW3 and CR6 may induce activation of distinct populations of, or pathways in, APC populations that would drive these differences. These results may have broad impacts on our understanding of how non-latent, chronic viral infections persist within a host. Funding Agencies CIHR


2021 ◽  
Author(s):  
◽  
Taryn Louise Osmond

<p>Splenic CD8α⁺ dendritic cells (DCs) have been described as key antigen presenting cells for the induction of CD8⁺ T cell responses to circulating antigen. This is through a heightened capacity to acquire and present the antigens via the process of cross-presentation, expression of high levels of the co-stimulatory and adhesion molecules required to stimulate CD8⁺ T cells, and the capacity to release high levels of the cytokines required to drive differentiation of CD8⁺ T cells into cytotoxic T lymphocytes (CTLs). However, recent research has indicated that the splenic CD8α⁺ DC population is more heterogeneous than originally thought. A previous study from my own laboratory suggested that a population of CD8α⁺ DCs that express the c-type lectin langerin primarily possess the heightened functions previously attributed to the total CD8α⁺ population. Therefore, the aim of this thesis research was to explore this subset of DCs in more detail, with specific emphasis on gaining mechanistic insight into their ability to elicit CD8⁺ T cell responses to circulating proteins. In the first section of this thesis, the hypothesis that the splenic langerin⁺ CD8α⁺ DCs were the critical subset involved in the induction of strong systemic CD8⁺ T cell responses to circulating antigen was tested in detail. This was examined using a genetically modified mouse model in which langerin-expressing cells could be easily identified and/or specifically depleted. It was first shown that the induction of CD8⁺ T cell responses to the model antigen ovalbumin was dependent on entry into the spleen in the presence of appropriate stimulation, which in these studies was provided by agonists for the toll-like receptors (TLRs) and/or signals from innate-like lymphocytes called natural killer T (NKT) cells. The primary targets for these signals were shown to be splenic langerin⁺ CD8α⁺ DCs, as CD8⁺ T cell responses were significantly reduced in hosts depleted of these cells within the spleen. Furthermore, agonists for TLRs that were not expressed by langerin⁺ CD8α⁺ DCs failed to enhance T cell responses. The langerin⁺ CD8α⁺ DCs were shown to be located in the marginal zone of the spleen, where they could readily screen the blood for antigens, and their function was critical to the induction of CD8⁺ T cell responses within six hours of antigen delivery. Interestingly, other local langerin-negative antigen presenting cells (APCs) were shown to be capable of cross-presentation, but with significantly reduced capacity to prime CD8⁺ T cell responses. Therefore, in the second section of this thesis the hypothesis that the langerin-negative APCs were capable of contributing to CD8⁺ T cell responses with appropriately timed stimuli was investigated. One of the downstream effects of inducing NKT cell activation at the time of priming was shown to be the “pre-conditioning” of langerin-negative DCs, allowing them to respond strongly to subsequent TLR ligation. Using SiglecH-DTR mice, it was shown that plasmacytoid DCs (which are langerin-negative) were pre-conditioned by NKT cell activation, allowing them to respond more actively to the delayed TLR stimulation by producing significantly enhanced levels of IFN-α. This factor was also potentially responsible for “feeding back” to the CD8α⁺ DCs (including langerin-expressing CD8α⁺ DCs), to enhance their function, as indicated by increases in cytokine production. Significantly, the major langerin-negative DC populations, defined as CD8α⁻ DCs, were pre-conditioned to have an enhanced cytokine release response to subsequent stimulation through TLR7, a receptor not expressed by langerin-positive DCs. This enhanced ability to respond to TLR7 ligation permitted these langerin-negative APCs to contribute to increased CD8⁺ T cell accumulation, with enhanced functional activity. Importantly, the CD8⁺ T cell response induced remained significantly dependent on initial cross-priming by langerin⁺ CD8α⁺ DCs, and it was only through pre-conditioning that langerinnegative APCs could contribute to enhancing the T cell response. In the third section of this thesis, the hypothesis that the CD8⁺ T cell responses generated in the presence of langerin⁺ CD8α⁺ DCs were phenotypically and functionally distinct from those responses generated in their absence was tested. No obvious differences were seen in CD8⁺ T cell homing, memory phenotype, restimulatory capacity, and expression of key molecules involved in metabolic function, survival and cytolytic function. However, in vivo cytotoxic function several weeks after priming was comparable, suggesting that this function was not related to initial burst size, providing some evidence of difference in function between CD8⁺ T cells primed in the presence or absence of langerin⁺ CD8α⁺ DCs. In summary, the splenic langerin⁺ CD8α⁺ DCs are the major subset responsible for cross-priming CD8⁺ T cell responses to circulating antigen, and for interpreting multiple stimulatory signals for enhancing the response. However, effective CD8⁺ T cell responses can be generated in their absence, particularly when antigens are provided in the context of appropriately temporally phased stimuli.</p>


2014 ◽  
Vol 211 (2) ◽  
pp. 297-311 ◽  
Author(s):  
Danya Liu ◽  
Scott M. Krummey ◽  
I. Raul Badell ◽  
Maylene Wagener ◽  
Lumelle A. Schneeweis ◽  
...  

Mounting evidence in models of both autoimmunity and chronic viral infection suggests that the outcome of T cell activation is critically impacted by the constellation of co-stimulatory and co-inhibitory receptors expressed on the cell surface. Here, we identified a critical role for the co-inhibitory SLAM family member 2B4 (CD244) in attenuating primary antigen-specific CD8+ T cell responses in the presence of immune modulation with selective CD28 blockade. Our results reveal a specific up-regulation of 2B4 on antigen-specific CD8+ T cells in animals in which CD28 signaling was blocked. However, 2B4 up-regulation was not observed in animals treated with CTLA-4 Ig (abatacept) or CD28 blockade in the presence of anti–CTLA-4 mAb. 2B4 up-regulation after CD28 blockade was functionally significant, as the inhibitory impact of CD28 blockade was diminished when antigen-specific CD8+ T cells were deficient in 2B4. In contrast, 2B4 deficiency had no effect on CD8+ T cell responses during unmodified rejection or in the presence of CTLA-4 Ig. We conclude that blockade of CD28 signals in the presence of preserved CTLA-4 signals results in the unique up-regulation of 2B4 on primary CD8+ effectors, and that this 2B4 expression plays a critical functional role in controlling antigen-specific CD8+ T cell responses.


2021 ◽  
Vol 9 (Suppl 3) ◽  
pp. A814-A814
Author(s):  
Zachary Morrow ◽  
John-Demian Sauer

BackgroundThe aspiration of cancer immunotherapy is to generate large numbers of highly functional anti-tumor CD8+ T-cells. We and others have optimized Listeria monocytogenes as a powerful anti-cancer vaccine platform to drive such T-cell responses. Early clinical trial data suggest the number of T-cells generated correlates with efficacy, demanding an understanding of the factors that dictate vaccine-induced T-cell responses. The CD8+ T-cell response is intimately linked to magnitude and quality of the innate immune response triggered by vaccines. Listeria-based vaccines activate numerous innate pathways and can be engineered to hyper- or hypo-induce these pathways. We sought to understand how modulating innate immunity would impact vaccine efficacy.MethodsTo dissect the impact of type I interferon signaling and the inflammasomes on L. monocytogenes induced T-cell responses, we immunized IFNAR-/-, Caspase1/11-/-, and novel IFNAR-/-Caspase1/11-/- double knockouts mice we generated for this study. CD8+ T-cell responses were assessed at the peak T-cell response, after contraction and memory formation, and after rechallenge. The phenotype and magnitude of CD8+ T-cells was assessed at each stage, and functional outcomes were assessed by measuring protection from reinfection by wild-type Listeria.ResultsIFNAR-/- mice developed the largest number of CD8+ T-cells during the peak primary response contradicting the dogma that Type-I Interferon promotes robust CD8+ T-cell responses. Caspase1/11-/- mice were not significantly different from wild-type mice. The frequency of short-lived effector cells (assessed by expression of CD127 and KLRG1) was no different between wild-type and IFNAR-/- mice, however we observed more than twice as many memory precursor cells at the peak CD8+ T-cell response. These findings extend to the memory and recall stage with more antigen-specific T-cells observed after contraction and upon rechallenge. Finally, IFNAR-/- mice are remarkably more protected from wild-type Listeria rechallenge than their counterparts after immunization demonstrating the efficacy of the increased memory T-cell pool. Data are representative of at least two independent replicates with at least 5 mice per group and significance was assessed by one-way ANOVA with *p<0.05.ConclusionsWe demonstrated that type-I interferon signaling deficiency leads to enhanced prophylactic vaccine efficacy through increased memory T-cell formation. Ultimately, for patients with slow growing tumors or with high-risk mutations, prophylactic tumor vaccines could elicit life-long protection from disease. Importantly, increased memory precursor T-cell abundance did not come at the expense of short-lived effectors leaving open the possibility that blocking Type-I IFN could potentiate lasting immunological memory in both the therapeutic and prophylactic setting.


2021 ◽  
pp. annrheumdis-2021-220435
Author(s):  
Theresa Graalmann ◽  
Katharina Borst ◽  
Himanshu Manchanda ◽  
Lea Vaas ◽  
Matthias Bruhn ◽  
...  

ObjectivesThe monoclonal anti-CD20 antibody rituximab is frequently applied in the treatment of lymphoma as well as autoimmune diseases and confers efficient depletion of recirculating B cells. Correspondingly, B cell-depleted patients barely mount de novo antibody responses during infections or vaccinations. Therefore, efficient immune responses of B cell-depleted patients largely depend on protective T cell responses.MethodsCD8+ T cell expansion was studied in rituximab-treated rheumatoid arthritis (RA) patients and B cell-deficient mice on vaccination/infection with different vaccines/pathogens.ResultsRituximab-treated RA patients vaccinated with Influvac showed reduced expansion of influenza-specific CD8+ T cells when compared with healthy controls. Moreover, B cell-deficient JHT mice infected with mouse-adapted Influenza or modified vaccinia virus Ankara showed less vigorous expansion of virus-specific CD8+ T cells than wild type mice. Of note, JHT mice do not have an intrinsic impairment of CD8+ T cell expansion, since infection with vaccinia virus induced similar T cell expansion in JHT and wild type mice. Direct type I interferon receptor signalling of B cells was necessary to induce several chemokines in B cells and to support T cell help by enhancing the expression of MHC-I.ConclusionsDepending on the stimulus, B cells can modulate CD8+ T cell responses. Thus, B cell depletion causes a deficiency of de novo antibody responses and affects the efficacy of cellular response including cytotoxic T cells. The choice of the appropriate vaccine to vaccinate B cell-depleted patients has to be re-evaluated in order to efficiently induce protective CD8+ T cell responses.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A469-A469
Author(s):  
Bernard Fox ◽  
Tarsem Moudgil ◽  
Traci Hilton ◽  
Noriko Iwamoto ◽  
Christopher Paustian ◽  
...  

BackgroundOutcomes for recurrent or metastatic (R/M) head and neck squamous cell carcinoma (HNSCC) are dismal and responses to anti-PD-1 appear best in tumors with PD-1+ T cells in proximity to PD-L1+ cells, arguing that improved outcome is associated with a pre-existing anti-cancer immune response. Based on this, we hypothesize that vaccines which prime and/or expand T cells to a spectrum of antigens overexpressed by HNSCC combined with T cell agonists, like anti-GITR, that provide costimulatory signals will improve the anti-PD-1 response rates. We have developed a cancer vaccine, DPV-001, that contains more than 300 proteins for genes overexpressed by HNSCC, encapsulated in a CLEC9A-targeted microvesicle and containing TLR/NOD agonists and DAMPs. Recently, we reported that combining anti-GITR + vaccine + anti-PD-1 augmented therapeutic efficacy in a preclinical model and now plan a phase 1b trial of this combination in patients with advanced HNSCC.MethodsSera from patients receiving DPV-001 as adjuvant therapy for definitively treated NSCLC, were analyzed for IgG responses to human proteins by MAP bead arrays and results compared to TCGA gene expression data sets for HNSCC. HNSCC cell lines were evaluated by RNASeq and peptides were eluted from HLA, analyzed by mass spectroscopy and correlated against MAP bead arrays and TCGA data sets. Tumor-reactive T cells from a vaccinated patient were enriched and expanded, and used in cytokine release assay (CRA) against autologous NSCLC and partially HLA matched allogeneic HNSCC cell lines.ResultsPatients receiving DPV-001 (N=13) made 147 IgG responses to at least 70 proteins for genes overexpressed by HNSCC. Preliminary evaluation of the HNSCC peptidome against the results of MAP bead array identify antigens that are target of a humoral immune response. Additionally, tumor-reactive T cells from DPV-001 vaccinated patient recognize two partially HLA-matched HNSCC targets, but not a mis-matched target.ConclusionsRecent observations from our lab and others have correlated IgG Ab responses with T cell responses to epitopes of the same protein. Based on the data summarized above, we hypothesize that we have induced T cell responses against a broad spectrum of shared cancer antigens that are common among adenocarcinomas and squamous cell cancers. Our planned clinical trial will vaccinate and boost the induced responses by costimulation with anti-GITR and then sequence in delayed anti-PD-1 to relieve checkpoint inhibition. MAP bead arrays and the peptidome library generated above will be used to assess anti-cancer B and T cell responses.Trial RegistrationNCT04470024Ethics ApprovalThe original clinical trial was approved by the Providence Portland Medical Center IRB, approval # 13-046. The proposed clinical trial has not yet been reviewed by the IRB.


2005 ◽  
Vol 79 (15) ◽  
pp. 9419-9429 ◽  
Author(s):  
Nicole E. Miller ◽  
Jennifer R. Bonczyk ◽  
Yumi Nakayama ◽  
M. Suresh

ABSTRACT Although it is well documented that CD8 T cells play a critical role in controlling chronic viral infections, the mechanisms underlying the regulation of CD8 T-cell responses are not well understood. Using the mouse model of an acute and chronic lymphocytic choriomeningitis virus (LCMV) infection, we have examined the relative importance of peripheral T cells and thymic emigrants in the elicitation and maintenance of CD8 T-cell responses. Virus-specific CD8 T-cell responses were compared between mice that were either sham thymectomized or thymectomized (Thx) at ∼6 weeks of age. In an acute LCMV infection, thymic deficiency did not affect either the primary expansion of CD8 T cells or the proliferative renewal and maintenance of virus-specific lymphoid and nonlymphoid memory CD8 T cells. Following a chronic LCMV infection, in Thx mice, although the initial expansion of CD8 T cells was normal, the contraction phase of the CD8 T-cell response was exaggerated, which led to a transient but striking CD8 T-cell deficit on day 30 postinfection. However, the virus-specific CD8 T-cell response in Thx mice rebounded quickly and was maintained at normal levels thereafter, which indicated that the peripheral T-cell repertoire is quite robust and capable of sustaining an effective CD8 T-cell response in the absence of thymic output during a chronic LCMV infection. Taken together, these findings should further our understanding of the regulation of CD8 T-cell homeostasis in acute and chronic viral infections and might have implications in the development of immunotherapy.


Sign in / Sign up

Export Citation Format

Share Document