In Vitro Induction and Expression of T-Cell Immunity to Tumor-Associated Antigens

Author(s):  
Robert C. Burton ◽  
Stanley E. Chism ◽  
Noel L. Warner
Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 3891-3891
Author(s):  
Zwi N. Berneman ◽  
Nathalie Cools ◽  
Viggo F.I. Van Tendeloo ◽  
Marc Lenjou ◽  
Griet Nijs ◽  
...  

Abstract Dendritic cells (DC), the professional antigen presenting cells of the immune system, exert important functions both in induction of T cell immunity as well as of tolerance. Previously, it was accepted that the main function of immature DC (iDC) in their in vivo steady state condition is to maintain peripheral tolerance to self-antigens and that these iDC mature upon encounter of so-called danger signals and subsequently promote T cell immunity. However, a growing body of experimental evidence now indicates that traditional DC maturation can no longer be used to distinguish between tolerogenic and immunogenic properties of DC. In this study, we compared the in vitro stimulatory capacity of immature DC (iDC), cytokine cocktail-matured DC (CC-mDC) and poly I:C-matured DC (pIC-mDC) in the absence and presence of antigen. All investigated DC types could induce at least 2 subsets of regulatory T cells. We observed a significant increase in both the number of functionally suppressive transforming growth factor (TGF)-beta+ interleukin (IL)-10+ T cells as well as of CD4+CD25+FOXP3+ T cells within DC/T cell co-cultures as compared to T cell cultures without DC. The induction of these regulatory T cells correlates with in vitro T cell non-responsiveness after co-culture with iDC and CC-mDC, while stimulation with pIC-mDC resulted in reproducible cytomegalovirus pp65 or influenza M1 matrix peptide-specific T cell activation as compared to control cultures in the absence of DC. In addition, the T cell non-responsiveness after stimulation with iDC was shown to be mediated by TGF-beta and IL-10. Moreover, the suppressive capacity of CD4+ T cells activated by iDC and CC-mDC was shown to be transferable when these CD4+ T cells were added to an established T cell response. In contrast, addition of CD4+ T cells stimulated by pIC-mDC made responder T cells refractory to their suppressive activity. In conclusion, we hypothesize that DC have a complementary role in inducing both regulatory T cells and effector T cells, where the final result of antigen-specific T cell activation will depend on the activation state of the DC. This emphasizes the need for proper DC activation when T cell immunity is the desired effect, especially when used in clinical trials.


2000 ◽  
Vol 74 (21) ◽  
pp. 9987-9993 ◽  
Author(s):  
Rachel S. Friedman ◽  
Fred R. Frankel ◽  
Zhan Xu ◽  
Judy Lieberman

ABSTRACT Induction of cell-mediated immunity may be essential for an effective AIDS vaccine. Listeria monocytogenes is an attractive bacterial vector to elicit T-cell immunity to human immunodeficiency virus (HIV) because it specifically infects monocytes, key antigen-presenting cells, and because natural infection originates at the mucosa. Immunization with recombinant L. monocytogenes has been shown to protect mice from lymphocytic choriomeningitis virus, influenza virus, and tumor inoculation.L. monocytogenes expressing HIV gag elicits sustained high levels of Gag-specific cytotoxic T lymphocytes (CTLs) in mice. We have examined the ability of Listeria to infect human monocytes and present HIV antigens to CD8 T lymphocytes of HIV-infected donors to induce a secondary T-cell immune response. Using this in vitro vaccination protocol, we show that L. monocytogenes expressing the HIV-1 gag gene efficiently provides a strong stimulus for Gag-specific CTLs in HIV-infected donor peripheral blood mononuclear cells.Listeria expressing Nef also elicits a secondary in vitro anti-Nef CTL response. Since L. monocytogenes is a pathogen, before it can be seriously considered as a human vaccine vector, safety concerns must be addressed. We therefore have produced a highly attenuated strain of L. monocytogenes that requiresd-alanine for viability. The recombinant bacteria are attenuated at least 105-fold. We show that when these hyperattenuated bacteria are engineered to express HIV-1 Gag, they are at least as efficient at stimulating Gag-specific human CTLs in vitro as wild-type recombinants. These results suggest that attenuatedListeria is an attractive candidate vaccine vector to induce T-cell immunity to HIV in humans.


2012 ◽  
Vol 86 (16) ◽  
pp. 8713-8719 ◽  
Author(s):  
Lars T. Joeckel ◽  
Reinhard Wallich ◽  
Sunil S. Metkar ◽  
Christopher J. Froelich ◽  
Markus M. Simon ◽  
...  

The T cell granule exocytosis pathway is essential to control hepatotropic lymphocytic choriomeningitis virus strain WE (LCMV-WE) but also contributes to the observed pathology in mice. Although effective antiviral T cell immunity and development of viral hepatitis are strictly dependent on perforin and granzymes, the molecular basis underlying induction of functionally competent virus-immune T cells, including participation of the innate immune system, is far from being resolved. We demonstrate here that LCMV-immune T cells of interleukin-1 receptor (IL-1R)-deficient mice readily express transcripts for perforin and granzymes but only translate perforin, resulting in the lack of proapoptotic potentialin vitro. LCMV is not cleared in IL-1R-deficient mice, and yet the infected mice develop neither splenomegaly nor hepatitis. These results demonstrate that IL-1R signaling is central to the induction of proapoptotic CD8 T cell immunity, including viral clearance and associated tissue injuries in LCMV infection.


2005 ◽  
Vol 201 (4) ◽  
pp. 567-577 ◽  
Author(s):  
Jianuo Liu ◽  
Takashi Miwa ◽  
Brendan Hilliard ◽  
Youhai Chen ◽  
John D. Lambris ◽  
...  

Decay-accelerating factor ([DAF] CD55) is a glycosylphosphatidylinositol-anchored membrane inhibitor of complement with broad clinical relevance. Here, we establish an additional and unexpected role for DAF in the suppression of adaptive immune responses in vivo. In both C57BL/6 and BALB/c mice, deficiency of the Daf1 gene, which encodes the murine homologue of human DAF, significantly enhanced T cell responses to active immunization. This phenotype was characterized by hypersecretion of interferon (IFN)-γ and interleukin (IL)-2, as well as down-regulation of the inhibitory cytokine IL-10 during antigen restimulation of lymphocytes in vitro. Compared with wild-type mice, Daf1−/− mice also displayed markedly exacerbated disease progression and pathology in a T cell–dependent experimental autoimmune encephalomyelitis (EAE) model. However, disabling the complement system in Daf1−/− mice normalized T cell secretion of IFN-γ and IL-2 and attenuated disease severity in the EAE model. These findings establish a critical link between complement and T cell immunity and have implications for the role of DAF and complement in organ transplantation, tumor evasion, and vaccine development.


2021 ◽  
Vol 21 (3) ◽  
pp. 178-192
Author(s):  
D. A. Poteryaev ◽  
S. G. Abbasova ◽  
P. E. Ignatyeva ◽  
O. M. Strizhakova ◽  
S. V. Kolesnik ◽  
...  

With the onset of the COVID-19 pandemic, a number of molecular-based tests have been developed to diagnose SARS-CoV-2 infection. However, numerous available serological tests lack sufficient sensitivity or specificity. They do not detect specific antibodies in a significant proportion of patients with PCR-confirmed COVID-19. There is evidence that some convalescents have a relatively short-lived humoral immunity. In contrast, a number of publications have shown that T-cell response to human coronaviruses, including SARS-CoV-1, MERS, and SARS-CoV-2, can be strong and long-term. Assessment of T-cell immunity to SARS-CoV-2 is important not only for stratification of risks and identification of potentially protected populations with immunity acquired as a result of previous infection, but also for determining immunogenicity and potential efficacy of vaccines under development. The existing methods of quantitative or semi-quantitative assessment of specific T-cell response are mainly used in scientific research and are not standardised. The aim of the study was to develop and verify experimentally a test kit to be used in a standardised procedure for in vitro determination of T-cells specific to SARS-CoV-2 antigens, in human peripheral blood. Materials and methods: the TigraTest® SARS-CoV-2 kit developed by GENERIUM, which determines the number of T-cells secreting interferon gamma in vitro, was tested in the study. Samples of venous blood of volunteers from three different groups were analysed in the study: presumably healthy volunteers; COVID-19 convalescents; individuals vaccinated against SARS-CoV-2. Results: the authors developed the TigraTest® SARS-CoV-2 kit for in vitro determination of T-cells specific to SARS-CoV-2 antigens in human peripheral blood, demonstrated its specificity and performed preliminary assessment of its sensitivity. The study analysed the range and magnitude of the T-cell response in convalescent and vaccinated individuals. A pronounced T-cell response was also shown in some individuals with no symptoms or with unconfirmed diagnosis. It was discovered that the mean T-cell response to peptides of the spike protein (S-protein) was higher in the vaccinated individuals than in the convalescent patients. A correlation was determined between the severity of the disease and the level of T-cell response. Specific contributions of various groups of antigens to the T-cell response after COVID-19 infection were also determined. Conclusions: the TigraTest® SARS-CoV-2 kit is a specific and sensitive tool for the assessment of T-cell immunity to the SARS-CoV-2 virus, which can also be used for vaccinated individuals. The kit may be used in clinical practice for comprehensive assessment of immunity to SARS-CoV-2.


2013 ◽  
Vol 191 (12) ◽  
pp. 6178-6190 ◽  
Author(s):  
Jia Liu ◽  
Min Jiang ◽  
Zhiyong Ma ◽  
Kirsten K. Dietze ◽  
Gennadiy Zelinskyy ◽  
...  

2003 ◽  
Vol 105 (2) ◽  
pp. 221-225 ◽  
Author(s):  
Dirk Nagorsen ◽  
Carmen Scheibenbogen ◽  
Gerhard Schaller ◽  
Binta Leigh ◽  
Alexander Schmittel ◽  
...  

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 3240-3240
Author(s):  
Hannah Cullup ◽  
John Wilson ◽  
Alison M. Rice ◽  
Anne M. Dickinson ◽  
David J. Munster ◽  
...  

Abstract AGVHD is a life threatening complication of allogeneic HSCT, initiated by host and donor DC stimulation of donor T lymphocytes. Current AGVHD prophylaxis targets T lymphocytes, compromising anti-viral and therapeutic anti-leukemic responses. The presence of activated blood DC predicts for clinical AGVHD and we predict that a new strategy targeting therapy to activated DC should prevent alloimmune induction of AGVHD but preserve protective T cell immune responses. CD83 is a cell surface molecule expressed by activated DC. Having shown that a rabbit polyclonal antibody to human CD83 (RA83) depletes activated DC and suppresses alloimmune reactions in vitro, we tested the effect of RA83 treatment on T cell immunity in vitro and its ability to prevent human PBMC induced xenogeneic AGVHD in vivo. The effect of RA83 treatment on T cell numbers, proliferation and cytokine secretion in allogeneic MLR was compared with appropriate controls, including Campath-1H. Allogeneic responses in vitro were mirrored in a DC dependent in vivo model of xenogeneic AGVHD, in which 50×106 human PBMC were injected into an irradiated SCID mouse. Human cytokine levels were measured in MLR tissue culture supernatant (TCSN) and mouse serum at the time of sacrifice. Cytotoxic T cell responses to viral antigens (CMV and FMP) were analysed by specific pentamer analysis and Cr51 release assays, prior to and following HLA restricted peptide antigen specific expansion. RA83 was shown to have NK-mediated ADCC capacity. Cellular proliferation in the allogeneic MLR was reduced by both RA83 and Campath-1H treatment (p= 0.004 and 0.01 respectively vs controls) and both antibodies improved mouse survival in the human xenogenic AGVHD model (RA83: 93%, Campath-1H: 100%, p<0.0001). IFN-g was significantly reduced in the TCSN from MLR treated with RA83 (p=0.0391) and in sera taken from RA83 (p=0.0002) and Campath-1H (p=0.0051) treated mice. Serum IL-4 levels were maintained in RA83 and Campath-1H treated mice. The serum levels of IL-5, IL-8 and TNF in mice treated with RA83 were markedly reduced compared to controls (p=0.0256, 0.0025, 0.025, respectively), and the reductions were similar to those seen in Campath-1H treated mice. Similar numbers of T cells were recovered from RA83 treated and control MLR, and both CMV and FMP specific CD8+ T cells were retained. These cells were readily expanded by peptide pulsing and autologous restimulation and had specific cytotoxic activity comparable to control cultures (see figure). In contrast, Campath-1H treatment removed specific anti-viral responses (vs controls: CMV: p<0.00001 and FMP: p=0.0051). Specific antibody to CD83 depletes activated DC in vitro and prevents xenogeneic human DC dependent AGVHD in vivo. This was accompanied by a Th1 to Th2 skewing of the cytokine response. RA83, but not Campath-1H treatment retained normal numbers of T cells and maintained normal cytotoxic responses to common post-transplant viral infections. Depletion of activated DC may be an effective means of AGVHD control, which maintains T cell immunity to life threatening infections and potentially anti-leukaemia responses. RA83 treatment of allo-MLR preserves T cell anti-CMV immunity RA83 treatment of allo-MLR preserves T cell anti-CMV immunity


Sign in / Sign up

Export Citation Format

Share Document