scholarly journals 207 Enhanced antigen capture, antigen-presenting cell (APC)-like function, and cytotoxic responses with chimeric engulfment receptor (CER) T cells

2021 ◽  
Vol 9 (Suppl 3) ◽  
pp. A218-A218
Author(s):  
Daniel Corey ◽  
Sunil Thomas ◽  
Brandon Cieniewicz ◽  
Linh Nguyen ◽  
Jared Clever ◽  
...  

BackgroundActivated T cells have limited antigen presenting capability due to inefficient capture.1 This process can be enhanced through novel chimeric engulfment receptors (CERs) expressing a human Tim-4 phagocyte receptor that recognizes phosphatidylserine (Ptd-Ser)2 fused to T cell and macrophage/dendritic cell-derived signaling domains. CERs can facilitate antigen capture, processing, and presentation, and impart target-dependent cytotoxic function when expressed in T cells. This combined function is hypothesized to improve tumor clearance and durability of response, making CER T cell products ideal clinical candidates.MethodsWe generated Tim-4 receptors fused to toll-like receptor (TLR)-2 or -8, CD28 or CD3 zeta and tested phagocytic, antigen presentation and cytotoxic function in healthy donor T cells. To assess phagocytosis, target cells treated with a small molecule to induce Ptd-Ser externalization were labeled with pH-Rodo followed by co-culture with CER T cells. Activated CER T cells were evaluated by transmission electron microscopy (TEM) or flow cytometry (FC) for lysosomal uptake of cell fragments. Antigen capture and presentation were characterized by FC for the capacity of human papilloma virus 16 (HPV 16) E7 peptide-pulsed CER T cells to activate and induce proliferation of autologous HPV 16 E7-TCR transduced T cells. Cytotoxic function was evaluated in co-culture assays of CER T cells in the presence of subtherapeutic doses of BTKi (ibrutinib)-treated JeKo-1 lymphoma cells.ResultsTEM imaging demonstrated that CER T cells engulfed target cell fragments, illustrated by multi-vesicular bodies containing tumor fragments (some measuring >0.5 uM) and pseudo-pod like formations around apoptotic target cell blebs. RNA analysis revealed upregulation of TLR, myeloid differentiation, and antigen presentation pathways. In the HPV 16 E7 co-culture model, T-cell surface activation markers CD25 and CD69 were upregulated 41% and 23%, respectively, on E7-TCR-T cells relative to controls. In addition, the percentage of dividing E7-TCR-T cells was increased (44% vs 8%) after 6 days in co-culture. Addition of CER T cells to JeKo- 1 target cells in the presence of BTKi at low effector: target ratios enhanced cytotoxicity by over 99%, demonstrating synergy with a targeted small molecule to fully eliminate lymphoma cells.ConclusionsNovel Tim-4/TLR containing CERs can capture tumor cell fragments and present soluble antigen, a function previously demonstrated to be a barrier to effective antigen presentation in T cells. Enhanced T-cell antigen capture and presentation capability alongside inducible and target-specific cytotoxic function in single T cells represents a significant advancement in the potential for chimeric receptor-based therapies.ReferencesLanzavecchia A, Roosnek E, Gregory T, Berman P, Abrignani S. T cells can present antigens such as HIV gp120 targeted to their own surface molecules. Nature 1988 Aug 11;334(6182):530–2.Caronni N, Piperno GM, Simoncello F, Romano O, Vodret S, Yanagihashi Y, et al. TIM4 expression by dendritic cells mediates uptake of tumor-associated antigens and anti-tumor responses. Nat Commun 2021 Apr 14;12(1):2237.

Blood ◽  
2003 ◽  
Vol 101 (3) ◽  
pp. 983-991 ◽  
Author(s):  
Evangelos Andreakos ◽  
Clive Smith ◽  
Claudia Monaco ◽  
Fionula M. Brennan ◽  
Brian M. Foxwell ◽  
...  

AbstractAlthough dendritic cells (DCs) are the most potent antigen-presenting cells involved in numerous physiologic and pathologic processes, little is known about the signaling pathways that regulate DC activation and antigen-presenting function. Recently, we demonstrated that nuclear factor (NF)-κB activation is central to that process, as overexpression of IκBα blocks the allogeneic mixed lymphocyte reaction (MLR), an in vitro model of T-cell activation. In this study, we investigated the role of 2 putative NF-κB–inducing components, NF-κB–inducing kinase (NIK), and IκB kinase 2 (IKK2). Using an adenoviral gene transfer method to efficiently express dominant-negative (dn) forms of these molecules in monocyte-derived DCs, we found that IKK2dn but not NIKdn inhibited the allogeneic MLR. When DCs were fixed, this inhibitory effect of IKK2dn was lost, suggesting that IKK2 is involved in T-cell–derived signals that enhance DC antigen presentation during the allogeneic MLR period and does not have an effect on viability or differentiation state of DCs prior to coculture with T cells. One such signal is likely to be CD40 ligand (CD40L), as IKK2dn blocked CD40L but not lipopolysaccharide (LPS)–induced NF-κB activation, cytokine production, and up-regulation of costimulatory molecules and HLA-DR in DCs. In summary, our results demonstrate that IKK2 is essential for DC activation induced by CD40L or contact with allogeneic T cells, but not by LPS, whereas NIK is not required for any of these signals. In addition, our results support IKK2 as a potential therapeutic target for the down-regulation of unwanted immune responses that may occur during transplantation or autoimmunity.


1998 ◽  
Vol 187 (10) ◽  
pp. 1611-1621 ◽  
Author(s):  
Sarah E. Townsend ◽  
Christopher C. Goodnow

Antigen-specific B cells are implicated as antigen-presenting cells in memory and tolerance responses because they capture antigens efficiently and localize to T cell zones after antigen capture. It has not been possible, however, to visualize the effect of specific B cells on specific CD4+ helper T cells under physiological conditions. We demonstrate here that rare T cells are activated in vivo by minute quantities of antigen captured by antigen-specific B cells. Antigen-activated B cells are helped under these conditions, whereas antigen-tolerant B cells are killed. The T cells proliferate and then disappear regardless of whether the B cells are activated or tolerant. We show genetically that T cell activation, proliferation, and disappearance can be mediated either by transfer of antigen from antigen-specific B cells to endogenous antigen-presenting cells or by direct B–T cell interactions. These results identify a novel antigen presentation route, and demonstrate that B cell presentation of antigen has profound effects on T cell fate that could not be predicted from in vitro studies.


2006 ◽  
Vol 203 (2) ◽  
pp. 437-447 ◽  
Author(s):  
Ingo Klein ◽  
Ian Nicholas Crispe

The transplanted liver elicits systemic tolerance, and the underlying mechanism may also account for the persistence of liver infections, such as malaria and viral hepatitis. These phenomena have led to the hypothesis that antigen presentation within the liver is abortive, leading to T cell tolerance or apoptosis. Here we test this hypothesis in an optimized orthotopic liver transplantation model. In direct contradiction to this model, the liver itself induces full CD8+ T cell activation and differentiation. The effects of microchimerism were neutralized by bone marrow transplantation in the liver donor, and the lack of liver-derived antigen-presenting cells was documented by eight-color flow cytometry and by sensitive functional assays. We conclude that local antigen presentation cannot explain liver tolerance. On the contrary, the liver may be an excellent priming site for naive CD8+ T cells.


1984 ◽  
Vol 159 (3) ◽  
pp. 881-905 ◽  
Author(s):  
J D Ashwell ◽  
A L DeFranco ◽  
W E Paul ◽  
R H Schwartz

In this report we have examined the ability of small resting B cells to act as antigen-presenting cells (APC) to antigen-specific MHC-restricted T cells as assessed by either T cell proliferation or T cell-dependent B cell stimulation. We found that 10 of 14 in vitro antigen-specific MHC-restricted T cell clones and lines and three of four T cell hybridomas could be induced to either proliferate or secrete IL-2 in the presence of lightly irradiated (1,000 rads) purified B cells and the appropriate foreign antigen. All T cell lines and hybridomas were stimulated to proliferate or make IL-2 by macrophage- and dendritic cell-enriched populations and all T cells tested except one hybridoma caused B cell activation when stimulated with B cells as APC. Furthermore, lightly irradiated, highly purified syngeneic B cells were as potent a source of APC for inducing B cell activation as were low density dendritic and macrophage-enriched cells. Lymph node T cells freshly taken from antigen-primed animals were also found to proliferate when cultured with purified B cells and the appropriate antigen. Thus, small resting B cells can function as APC to a variety of T cells. This APC function was easily measured when the cells were irradiated with 1,000 rads, but was greatly diminished or absent when they were irradiated with 3,300 rads. Thus, the failure of some other laboratories to observe this phenomenon may be the result of the relative radiosensitivity of the antigen-presenting function of the B cells. In addition, this radiosensitivity allowed us to easily distinguish B cell antigen presentation from presentation by the dendritic cell and macrophage, as the latter was resistant to 3,300 rads. Finally, one T cell clone that failed to proliferate when B cells were used as APC was able to recruit allogeneic B cells to proliferate in the presence of syngeneic B cells and the appropriate antigen. This result suggests that there are at least two distinct pathways of activation in T cells, one that leads to T cell proliferation and one that leads to the secretion of B cell recruitment factor(s).


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 1411-1411 ◽  
Author(s):  
Anne Marijn Kramer ◽  
Mengyong Yan ◽  
Karl S Peggs ◽  
John Anderson ◽  
Kenth Gustafsson

Abstract Tumor-Associated Antigen Presentation by γδ T-Cells in Cancer Immunotherapy Human γδ T-cells are considered to represent a link between innate and adaptive immunity. Their innate killing properties display a potent cytotoxic activity against solid tumors as well as lymphoid and myeloid malignancies. Subsequently, by lysing affected target cells and liberating antigen for uptake, they can differentiate into professional antigen presenting cells (pAPCs) for induction of CD4+ and CD8+ T cell responses. The degree of antigen-specific stimulation of responder T cells is increased in the presence of antibody(Ab)-assisted opsonized target cells, involving the low-affinity receptor for IgG CD16 (Fc γRIII), equivalent to that seen with mature antigen-loaded DCs. To elaborate the implications of this combined killing and pAPC function we have studied how freshly isolated as well as expanded and cloned populations of γδ T-cell subsets kill a target tumor cell, and take up and cross-present tumor-associated antigens (TAA). We performed quantitative analysis on the cellular uptake of different sizes of microspheres, analyzing the correlation between opsonization and internalization. All γδ T-cell subtypes were expanded using artificial APC, engineered to express CD86, CD137L and IL-15, and anti- γδ TCR Ab (B1). Short (EAAGIGILTV) and long (GHSYTTAEEAAGIGILTVILGVLLL) MART-1 peptides were used as antigens for γδ T-cell presentation to MART-1 TCR-transduced cytotoxic T-cells. A CFSE assay was performed to assess cytotoxic T-cell proliferation. Target cells and polysterene microspheres were opsonized with human anti-CD20 IgG1, Rituximab (RTX). CD16 function was blocked with a mouse monoclonal IgG1 anti-CD16 blocking Ab (clone LNK16). Imaging flowcytometry allowed us to quantify internalization of FITC-labeled microspheres. The Internalization Score is defined as the ratio of intensity inside the cell to the intensity of the entire cell. Both γδ T-cell lines and expanded γδ T-cell clones cultured long-term, remarkably, retain both tumor cell killing and take up tumor cell lysates or long synthetic TAA peptides and cross-present these on MHC class I to CD8+ cytotoxic T-cells in a dynamic, controllable fashion, dependent on Ab-opsonization. (Figure 1). The Ab-opsonization of 1 µm microspheres correlates with a higher receptor-mediated phagocytic uptake, in a CD16 dependent manner (Figure 2). The opsonization of 0,5 µm microspheres led to clumping of the microspheres, accounting for the lower uptake in this particular subgroup. For a lack of better alternative, moDCs have been widely used in experimental immunotherapy settings. The ease of manipulation of human γδ T-cells, the ability to be expanded ex-vivo combined with antigen presentation makes them a great potential tool for immunotherapy as a complementary or integrative strategy. Ligation of the γδ T-cell receptor at the tumor site will activate their expansion and innate killing. Yet, antigen presentation will only occur after binding of an immunoglobulin to the tumor cell, thereby activating their dual role. Our goal is to define an effective adjuvant vaccine formulation for inducing leukemia-specific cytolytic effects. We are currently investigating whether γδ T-cells can directly present and/or cross-present to cytotoxic T-cells in-vivo in a humanized mouse model. We believe that the uptake of microspheres by γδ T-cells has an impact on the development of vaccination strategies for cancer immunotherapy, as the immunization of γδ T-cells is a powerful method for the induction or reactivation of cytotoxic T cell specific responses. FIGURE 1 CFSE assay of γδ T-cell lines cross-presenting short and long MART-1 peptides to MART-1 TCR-transduced cytotoxic T-cells in a dynamic, controllable fashion, dependent on Ab-opsonization FIGURE 1. CFSE assay of γδ T-cell lines cross-presenting short and long MART-1 peptides to MART-1 TCR-transduced cytotoxic T-cells in a dynamic, controllable fashion, dependent on Ab-opsonization FIGURE 2a FIGURE 2a. FIGURE 2b FIGURE 2b. Disclosures No relevant conflicts of interest to declare.


2021 ◽  
Author(s):  
◽  
Aras Toker

<p>Glatiramer acetate (GA) is approved for the treatment of relapsing-remitting multiple sclerosis (MS), and can suppress experimental autoimmune encephalomyelitis (EAE), a murine model of human MS. GA treatment is associated with the induction of anti-inflammatory TH2 responses and with the antigen specific expansion of regulatory T cells that counteract or inhibit pathogenic events in MS and EAE. These T cell mediated mechanisms of protection are considered to be a result of modulation of antigen presenting cells (APCs) by GA, rather than direct effects on T cells. However, it is unknown if GA preferentially targets a specific APC subset or can act through multiple APCs in vivo. In addition, GA-modulated innate cells may also exhibit direct antigen non-specific suppression of autoreactive cells. One objective of this study was to identify the in vivo target cell population of GA and to assess the potential of the target cells to antigen non-specifically suppress immune responses. Fluorophor-labelled GA bound to monocytes after intravenous injections, suggesting that monocytes may be the primary target of GA in vivo. In addition, intravenous GA treatment enhanced the intrinsic ability of monocytes to suppress T cell proliferation, both in vitro and in vivo. The findings of this study therefore suggest that GA-induced monocytes may contribute to GA therapy through direct mechanisms of antigen non-specific T cell immunosuppression. A further objective of this work was to investigate the potential of an in vivo drug targeting approach. This approach was hypothesised to increase the uptake of GA by the target cells and substantially improve GA treatment through antigen specific mechanisms such as induction of TH2 or regulatory T cells. Targeting antigens to professional APCs with an anti-MHC class II antibody resulted in significantly enhanced T cell proliferation in vitro. However, no EAE suppression occurred when GA was targeted to MHC class II in vivo. In addition, targeting GA specifically to monocytes also failed to suppress EAE. These findings suggest that GA treatment may selectively modulate monocytes to enhance their ability to inhibit autoreactive T cells, which could be part of the mechanism by which GA ameliorates MS. Targeting GA to a specific cell type may not be a powerful approach to improve treatment, because increased proliferation of GA specific T cells is not sufficient for disease suppression, and conjugation to antibodies may functionally reduce GA to a mere antigen devoid of immunomodulatory capacity.</p>


2019 ◽  
Vol 47 (5) ◽  
pp. 419-434
Author(s):  
A. V. Kil'dyushevskiy ◽  
V. A. Molochkov ◽  
T. A. Mitina ◽  
Ya. G. Moysyuk ◽  
A. V. Molochkov

Aim: To present well-known and disputable mechanisms of the effects of extracorporeal photopheresis (ECP) in heterogeneous clinical conditions, as well as to demonstrate its advantages over conventional hormonal, immunosuppressive and cytostatic treatments, with a recommendation to widely implement it into practical management of autoimmune disease and cutaneous T-cell lymphomas (CTCLs).Key points: Despite convincing evidence of the ECP efficacy in the treatment of T-cell mediated disorders, a unifying concept of its mechanism has not been established so far. In this review, we attempted to determine the value of multiple, sometimes contradictory and equivocal points of view to immunobiochemical processes underlying the restoration of mechanism of immune tolerance in some autoimmune diseases and CTCLs. We focused our attention on our own clinical and immunological data obtained during a 20-years' experience with the use of ECP in clinical departments of MONIKI (Russia). Based on this, we have shown that ECP is more effective in autoimmune diseases than conventional treatment approaches with hormones, immunosuppressants and cytostatics. Unlike them, ECP is selectively targeted to auto-aggressive T-cells without induction of systemic immunosuppression. The leading role is played by the transformation of activated (immunogenic) myeloid dendrite cells (DC) into tolerogenic cell associated with their synthesis of inhibitor cytokines. The interplay of the cytokines with an antigen results in polarization of CD4+ Т lymphocytes via the Th2 pathway with restoration of the Th1/Th2 balance and their cytokine production. ECP triggers regulatory anti-clonotypic effector memory cells at the end stage of CD3+/CD8+/CD27-/CD28-/CD62L+ differentiation, that provide and maintain the peripheral immune tolerance, by deletion of the clone of auto-reactive cytotoxic lymphocytes and inducing their apoptosis. In autoimmune disorders, ECP results in reduction of the expression of integrin adhesion molecules on auto-reactive cell membranes with subsequent loss of their ability to migrate through the endothelium to their target cells. In its turn, it leads to decreasing immunoinflammatory response in the lesion. Both clinical and experimental data indicate that the mechanism of ECP action against CTCLs is characterized by activation of tumor cell apoptosis, unblocking of co-activation receptors on the antigen-presenting DC providing the functioning of the second signaling pathway for T lymphocyte activation. This results in proliferation of anti-tumor effector cells pool, production of DC activating cytokines that participate in the CD4+ polarization via Th1 pathway. In addition, this review considers the mechanism of the immunomodulating effect of ECP in the context of its influence at the levels of transcription and translation of proteins contributing to the pathophysiology of the disorders, based on molecular immunogenetic studies. Thus, ECP is able to induce antigen-specific immunological tolerance through the transformation of antigen-presenting cells, modulation of cytokine profile, adhesion and activation molecules, as well as through formatting of the regulatory T cells (Tregs).Conclusion: Undoubtedly, the immunobiological ECP technique has significant advantages over well-known conventional hormonal, immunosuppressive, and cytostatic therapies of autoimmune diseases and CTCLs.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 3889-3889
Author(s):  
Klaus Brischwein ◽  
Scott A. Hammond ◽  
Larissa Parr ◽  
Schlereth Bernd ◽  
Mathias Locher ◽  
...  

Abstract Background: Bispecific antibodies have been extensively studied in vitro and in vivo for their use in redirected tumor cell lysis. A particular challenge of bispecific antibody constructs recognizing the CD3 signaling complex is to achieve a controlled polyclonal activation of T-cells that, ideally, is entirely dependent on the presence of target cells. If this is not the case, systemic production of inflammatory cytokines and secondary endothelial reactions may occur as side effects, as are observed with the murine anti-human CD3e antibody OKT-3 (muromab, Orthoclone®). Here we present evidence that MT103 (or MEDI-538), a bispecific single chain antibody of the BiTE class that targets CD19 and CD3, induces T-cell activation exclusively in the presence of target cells. Material and methods: Peripheral blood mononuclear cells from healthy donors were prepared by Ficoll density centrifugation. PBMC were incubated for 24 hours with MT103 in presence or absence of specific target cells. Target cell lysis was determined by measurement of adenylate kinase activity released from lysed cells. De novo expression of activation markers CD69 and CD25 on T-cells was assessed by flow cytometry using directly conjugated monoclonal antibodies, and the concentration of cytokines in the supernatant was determined by a commercial FACS-based bead array. Results: MT103 was analyzed for conditional T-cell activation. In the presence of target-expressing cell lines, low picomolar concentrations of MT103 were sufficient to stimulate a high percentage of peripheral human T-cells to express cytokines and surface activation markers, to enter into the cell cycle and to induce redirected lysis of target cells. However, in the absence of target cells, the BiTE molecules no longer detectably activated human T-cells even at concentrations exceeding the ED50 for redirected lysis and conditional T-cell activation by more than five orders of magnitude. Conclusion: Our data show that T-cell activation by MT103 is highly conditional in that it is strictly dependent on the presence.


Blood ◽  
2004 ◽  
Vol 103 (7) ◽  
pp. 2677-2682 ◽  
Author(s):  
Inge Jedema ◽  
Nicole M. van der Werff ◽  
Renée M. Y. Barge ◽  
Roel Willemze ◽  
J. H. Frederik Falkenburg

Abstract For the clinical evaluation of the efficacy of cellular immunotherapy it is necessary to analyze the effector functions of T cells against primary leukemic target cell populations which are usually considerably heterogeneous caused by differential maturation stages of the leukemic cells. An appropriate assay should not only allow the quantitative analysis of rapid cell death induction as measured by the conventional 51Cr release assay but also of the more slowly executing pathways of T-cell-induced apoptosis occurring within days instead of hours which cannot be measured using this method. Furthermore, it should dissect the differential susceptibility to T-cell-induced cell death of various target cell subpopulations and characterize the malignant precursor cells capable of producing malignant progeny. To fulfill these requirements we developed a new assay based on carboxyfluorescein diacetate succinimidyl ester (CFSE) labeling of the target cell population combined with antibody staining of specific cell populations and addition of fluorescent microbeads to quantitatively monitor target cell death occurring within a longer time frame up to at least 5 days. This new assay facilitates the analysis of differential recognition of distinct cell types within a heterogeneous target cell population and allows simultaneously evaluation of the proliferative status of surviving target cells in response to relevant cytokines. (Blood. 2004;103: 2677-2682)


Sign in / Sign up

Export Citation Format

Share Document