scholarly journals Tumor-Associated Antigen Presentation by γδ T-Cells in Cancer Immunotherapy

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 ◽  
Vol 11 (9) ◽  
pp. 923
Author(s):  
Josephine G. M. Strijker ◽  
Ronja Pscheid ◽  
Esther Drent ◽  
Jessica J. F. van der Hoek ◽  
Bianca Koopmans ◽  
...  

Currently ~50% of patients with a diagnosis of high-risk neuroblastoma will not survive due to relapsing or refractory disease. Recent innovations in immunotherapy for solid tumors are highly promising, but the low MHC-I expression of neuroblastoma represents a major challenge for T cell-mediated immunotherapy. Here, we propose a novel T cell-based immunotherapy approach for neuroblastoma, based on the use of TEG002, αβ-T cells engineered to express a defined γδ-T cell receptor, which can recognize and kill target cells independent of MHC-I. In a co-culture killing assay, we showed that 3 out of 6 neuroblastoma organoids could activate TEG002 as measured by IFNγ production. Transcriptional profiling showed this effect correlates with an increased activity of processes involved in interferon signaling and extracellular matrix organization. Analysis of the dynamics of organoid killing by TEG002 over time confirmed that organoids which induced TEG002 activation were efficiently killed independent of their MHC-I expression. Of note, efficacy of TEG002 treatment was superior to donor-matched untransduced αβ-T cells or endogenous γδ-T cells. Our data suggest that TEG002 may be a promising novel treatment option for a subset of neuroblastoma patients.


2021 ◽  
Vol 22 (16) ◽  
pp. 8910
Author(s):  
Masatsugu Miyashita ◽  
Teruki Shimizu ◽  
Eishi Ashihara ◽  
Osamu Ukimura

Human γδ T cells show potent cytotoxicity against various types of cancer cells in a major histocompatibility complex unrestricted manner. Phosphoantigens and nitrogen-containing bisphosphonates (N-bis) stimulate γδ T cells via interaction between the γδ T cell receptor (TCR) and butyrophilin subfamily 3 member A1 (BTN3A1) expressed on target cells. γδ T cell immunotherapy is classified as either in vivo or ex vivo according to the method of activation. Immunotherapy with activated γδ T cells is well tolerated; however, the clinical benefits are unsatisfactory. Therefore, the antitumor effects need to be increased. Administration of γδ T cells into local cavities might improve antitumor effects by increasing the effector-to-target cell ratio. Some anticancer and molecularly targeted agents increase the cytotoxicity of γδ T cells via mechanisms involving natural killer group 2 member D (NKG2D)-mediated recognition of target cells. Both the tumor microenvironment and cancer stem cells exert immunosuppressive effects via mechanisms that include inhibitory immune checkpoint molecules. Therefore, co-immunotherapy with γδ T cells plus immune checkpoint inhibitors is a strategy that may improve cytotoxicity. The use of a bispecific antibody and chimeric antigen receptor might be effective to overcome current therapeutic limitations. Such strategies should be tested in a clinical research setting.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A814-A814
Author(s):  
Vanessa Ott ◽  
Julia Gilden ◽  
Jamison Grailer ◽  
Michael Slater ◽  
Pete Stecha ◽  
...  

BackgroundTwo main approaches for T cell-based therapies involve molecular T cell redirection by CD3 bispecific molecules such as bispecific T-cell engagers (BiTE) and cellular T cell redirection by genetic modification of T cells with chimeric antigen receptors (CAR) or transgenic T cell receptors (TCR). BiTEs redirect the cytotoxic activity of endogenous polyclonal T cells by simultaneously engaging CD3 on T cells and tumor antigens on target cells. BiTE potency studies have relied on primary cells, which measure target cell killing through redirected T cell cytotoxicity (RTCC) or cytokine release. However, these primary cell-based assays suffer from high donor-to-donor variability, as well as lengthy and hard to implement protocolsMethodsWe have recently developed a new RTCC assay and cytokine immunoassays that are simple, sensitive and can quantitatively measure the potency of BiTEs and similar biologics. In this assay, preactivated cytotoxic T cells and target cells (both in cryopreserved thaw-and-use format) stably expressing a HaloTag-HiBiT fusion protein are co-incubated with a BiTE, which results in lysis of the target cells and subsequent release of the Halotag-HiBiT protein. These HiBiT proteins then bind to extracellular LgBiT provided in the detection reagent and form functional NanoLuc Luciferase to generate luminescence.ResultsThe assay is homogenous, highly sensitive, and has a robust assay window. Use of CAR-T has demonstrated promising results in treating leukemia, while the development of TCR-engineered T cells that can recognize intracellular tumor antigens, is still in early stages. To facilitate the screening and characterization of new transgenic TCRs, we used CRISPR/Cas9 to develop two TCRaß-null reporter T cell lines, which are CD4+ or CD8+. Reintroduction of peptide-specific TCR a and ß chains into TCRaß-null reporter T cell lines results in peptide-dependent TCR activation and luciferase reporter expression. The select expression of CD4 or CD8 in the TCRaß-null reporter T cell lines can enable the development of transgenic TCRs for both MHCI- and MHCII-restricted tumor antigen targets.ConclusionsTogether, these bioluminescent bioassays represent a new set of tools for the discovery and development of T cell-based immunotherapies.


Author(s):  
Rasika Kumarasingha ◽  
Lisa J. Ioannidis ◽  
Waruni Abeysekera ◽  
Stephanie Studniberg ◽  
Dinidu Wijesurendra ◽  
...  

Abstractγδ T cells play an essential role in the immune response to malaria infection. However, long-lasting effects of malaria infection on the γδ T cell population still remain inadequately understood. This study investigated transcriptional changes and memory-like functional capacity of malaria pre-exposed γδ T cells using a Plasmodium chabaudi infection model. We show that multiple genes associated with effector function (chemokines, cytokines and cytotoxicity) and antigen-presentation were upregulated in P. chabaudi-exposed γδ T cells compared to γδ T cells from naïve mice. This transcriptional profile was positively correlated with profiles observed in conventional memory CD8+ T cells and was accompanied by enhanced reactivation upon secondary encounter with Plasmodium-infected red blood cells in vitro. Collectively our data demonstrate that Plasmodium exposure result in “memory-like imprints” in the γδ T cell population and also promotes γδ T cells that can support antigen-presentation during subsequent infections.


2022 ◽  
Author(s):  
Linjie Yuan ◽  
Xianqiang Ma ◽  
Yunyun Yang ◽  
Xin Li ◽  
Weiwei Ma ◽  
...  

Tumor cells and pathogen-infected cells are presented to human γδ T cells based on "inside-out" signaling in which metabolites called phosphoantigens (pAgs) inside target cells are recognized by the intracellular domain of a butyrophilin protein (BTN3A1), leading to an extracellular conformational change. Here, we report that pAgs function as molecular "glues" that initiate a heteromeric association between the intracellular domains of BTN3A1 and the structurally similar BTN2A1. Working with both exogenous and endogenous pAgs, we used x-ray crystallography, mutational studies, cellular assays, synthetic probe as well as molecular dynamics investigations to determine how pAgs glue intracellular BTN3A1 and BTN2A1 together for the "inside-out" signaling that triggers γδ T cell activation. This γδ T cell-specific mode of antigen sensing creates opportunities for the development of alternative immunotherapies against cancer and infectious diseases that do not involve αβ T cells.


2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Ruben Serrano ◽  
Christoph Coch ◽  
Christian Peters ◽  
Gunther Hartmann ◽  
Daniela Wesch ◽  
...  

AbstractHuman Vγ9Vδ2 T cells recognize pyrophosphates produced by microbes and transformed cells and play a role in anti-infective immunity and tumor surveillance. Toll-like receptors (TLR) are pattern recognition receptors in innate immune cells which sense microbial structures including nucleic acids. Given that γδ T cells are in clinical development for application in cellular cancer immunotherapy and TLR ligands have potent adjuvant activity, we investigated the co-stimulatory role of selected TLR ligands in γδ T-cell activation. Here we have used recently described RNA ligands for TLR7 and TLR8 together with Vγ9Vδ2 T-cell specific pyrophosphate antigens to analyze the rapid cytokine induction in Vδ2 T cells as well as the accessory cell requirements. While TLR8- as well as TLR7/8-specific RNA did not induce IFN-γ in Vδ2 T cells on their own, they provided strong co-stimulation for Vδ2 T cells within peripheral blood mononuclear cells in the presence of additional T-cell receptor activation. In contrast, TLR7 ligands were ineffective. Purified γδ T cells did not directly respond to TLR8 co-stimulation but required the presence of monocytes. Further experiments revealed a critical role of IL-1β and IL-18, and to a slightly lesser extent of IL-12p70, in the co-stimulation of Vδ2 T cells by TLR8 and TLR7/8 RNA ligands. Results of intracellular cytokine expression were validated by ELISA analysis of cytokines in cell culture supernatants. The cell context-dependent adjuvant activity of TLR8 and TLR7/8 RNA ligands described here might be important for the future optimization of γδ T-cell based cancer immunotherapy.


1999 ◽  
Vol 67 (5) ◽  
pp. 2241-2249 ◽  
Author(s):  
Claudia A. Daubenberger ◽  
Evans L. N. Taracha ◽  
Laima Gaidulis ◽  
William C. Davis ◽  
Declan J. McKeever

ABSTRACT T cells bearing the γδ antigen receptor (γδ T cells) can constitute up to 50% of T cells in the peripheral blood and lymphoid organs of young cattle. We present data showing that γδ T cells are involved in immune responses against Theileria parva. γδ T cells isolated from peripheral blood mononuclear cells (PBMC) of T. parva-naive and -immune cattle proliferated in the presence of fixed or unfixed autologous T. parva-infected lymphoblasts (TpL) and heat-stressed concanavalin A (ConA)-induced blasts (ConA blasts) but not untreated ConA blasts. The specificity of response was further evaluated with a panel of γδ T-cell lines and clones. T-cell reactivity was blocked by GB21A, a monoclonal antibody (MAb) specific for the γδ T-cell receptor, but not by MAbs specific for class I and class II major histocompatibility complex (MHC) molecules. In addition, TpL but not ConA blasts from a variety of MHC-mismatched animals induced proliferation of the γδ T-cell lines and clones. These γδ T cells were found to respond to TpL infected with several different parasite stocks and failed to recognize TpL after elimination of the parasite by the theilericidal drug BW 720C. Assays for cytotoxic activity of γδ T cells sorted from bulk cultures of immune PBMC restimulated several times with autologous TpL demonstrated that effector cells whose specificity is similar to that of proliferating cells are generated. These results suggest that bovine γδ T cells are activated by and lyse T. parva-infected cells by recognizing conserved parasite-induced or parasite-derived antigens in an MHC-unrestricted fashion.


2021 ◽  
Vol 9 (4) ◽  
pp. e002051
Author(s):  
Ryan Michael Reyes ◽  
Yilun Deng ◽  
Deyi Zhang ◽  
Niannian Ji ◽  
Neelam Mukherjee ◽  
...  

BackgroundAnti-programmed death-ligand 1 (αPD-L1) immunotherapy is approved to treat bladder cancer (BC) but is effective in <30% of patients. Interleukin (IL)-2/αIL-2 complexes (IL-2c) that preferentially target IL-2 receptor β (CD122) augment CD8+ antitumor T cells known to improve αPD-L1 efficacy. We hypothesized that the tumor microenvironment, including local immune cells in primary versus metastatic BC, differentially affects immunotherapy responses and that IL-2c effects could differ from, and thus complement αPD-L1.MethodsWe studied mechanisms of IL-2c and αPD-L1 efficacy using PD-L1+ mouse BC cell lines MB49 and MBT-2 in orthotopic (bladder) and metastatic (lung) sites.ResultsIL-2c reduced orthotopic tumor burden and extended survival in MB49 and MBT-2 BC models, similar to αPD-L1. Using antibody-mediated cell depletions and genetically T cell-deficient mice, we unexpectedly found that CD8+ T cells were not necessary for IL-2c efficacy against tumors in bladder, whereas γδ T cells, not reported to contribute to αPD-L1 efficacy, were indispensable for IL-2c efficacy there. αPD-L1 responsiveness in bladder required conventional T cells as expected, but not γδ T cells, altogether defining distinct mechanisms for IL-2c and αPD-L1 efficacy. γδ T cells did not improve IL-2c treatment of subcutaneously challenged BC or orthotopic (peritoneal) ovarian cancer, consistent with tissue-specific and/or tumor-specific γδ T cell contributions to IL-2c efficacy. IL-2c significantly altered bladder intratumoral γδ T cell content, activation status, and specific γδ T cell subsets with antitumor or protumor effector functions. Neither IL-2c nor αPD-L1 alone treated lung metastatic MB49 or MBT-2 BC, but their combination improved survival in both models. Combination treatment efficacy in lungs required CD8+ T cells but not γδ T cells.ConclusionsMechanistic insights into differential IL-2c and αPD-L1 treatment and tissue-dependent effects could help develop rational combination treatment strategies to improve treatment efficacy in distinct cancers. These studies also provide insights into γδ T cell contributions to immunotherapy in bladder and engagement of adaptive immunity by IL-2c plus αPD-L1 to treat refractory lung metastases.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Xiaohui Wang ◽  
Xiang Lin ◽  
Zihan Zheng ◽  
Bingtai Lu ◽  
Jun Wang ◽  
...  

AbstractInnate immunity is important for host defense by eliciting rapid anti-viral responses and bridging adaptive immunity. Here, we show that endogenous lipids released from virus-infected host cells activate lung γδ T cells to produce interleukin 17 A (IL-17A) for early protection against H1N1 influenza infection. During infection, the lung γδ T cell pool is constantly supplemented by thymic output, with recent emigrants infiltrating into the lung parenchyma and airway to acquire tissue-resident feature. Single-cell studies identify IL-17A-producing γδ T (Tγδ17) cells with a phenotype of TCRγδhiCD3hiAQP3hiCXCR6hi in both infected mice and patients with pneumonia. Mechanistically, host cell-released lipids during viral infection are presented by lung infiltrating CD1d+ B-1a cells to activate IL-17A production in γδ T cells via γδTCR-mediated IRF4-dependent transcription. Reduced IL-17A production in γδ T cells is detected in mice either lacking B-1a cells or with ablated CD1d in B cells. Our findings identify a local host-immune crosstalk and define important cellular and molecular mediators for early innate defense against lung viral infection.


Sign in / Sign up

Export Citation Format

Share Document