AN INTERLEUKIN-2-IgG-Fas LIGAND FUSION PROTEIN SUPPRESSES DELAYED-TYPE HYPERSENSITIVITY IN MICE BY TRIGGERING APOPTOSIS IN ACTIVATED T CELLS AS A NOVEL STRATEGY FOR IMMUNOSUPPRESSION1

2000 ◽  
Vol 69 (7) ◽  
pp. 1386-1391 ◽  
Author(s):  
Silvia Bulfone-Paus ◽  
Ren?? R??ckert ◽  
Hans Krause ◽  
Horst von Bernuth ◽  
Michael Notter ◽  
...  
1998 ◽  
Vol 66 (6) ◽  
pp. 2632-2639 ◽  
Author(s):  
Juneann W. Murphy ◽  
Fredda Schafer ◽  
Arturo Casadevall ◽  
Adekunle Adesina

ABSTRACT Mice immunized with two different cryptococcal antigen preparations, one a soluble culture filtrate antigen (CneF) in complete Freund’s adjuvant (CFA) and the other heat-killed Cryptococcus neoformans cells (HKC), develop two different profiles of activated T cells. CneF-CFA induces CD4+ T cells responsible for delayed-type hypersensitivity (DTH) reactivity and for amplification of the anticryptococcal DTH response, whereas HKC induce CD4+ and CD8+ T cells involved in anticryptococcal DTH reactivity and activated T cells which directly kill C. neoformans cells. The main purpose of this study was to assess the level of protection afforded by each of the two different T-cell profiles against challenge with viable C. neoformans cells, thereby identifying which activated T-cell profile provides better protection. CBA/J mice immunized with CneF-CFA had significantly better protective responses, based on better clearance of C. neoformans from tissues, on longer survival times, and on fewer and smaller lesions in the brain, than HKC-immunized mice or control mice similarly infected with C. neoformans. Both immunization protocols induced an anticryptococcal DTH response, but neither induced serum antibodies to glucuronoxylmannan, so the protection observed in the CneF-CFA immunized mice was due to the activated T-cell profile induced by that protocol. HKC-immunized mice, which displayed no greater protection than controls, did not have the amplifier cells. Based on our findings, we propose that the protective anticryptococcal T cells are the CD4+ T cells which have been shown to be responsible for DTH reactivity and/or the CD4+ T cells which amplify the DTH response and which have been previously shown to produce high levels of gamma interferon and interleukin 2. Our results imply that there are protective and nonprotective cell-mediated immune responses and highlight the complexity of the immune response to C. neoformans antigens.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1157-1157
Author(s):  
Mayur S Ramesh ◽  
Archana Thakur ◽  
Philip Pellett ◽  
Subhendu Das ◽  
Zaid S Al-Kadhimi ◽  
...  

Abstract Abstract 1157 Poster Board I-179 Introduction CMV reactivation and infection can cause profound negative outcome post allogeneic SCT. Current management strategies against CMV are suboptimal and are associated with adverse effects. Induction of anti-CMV T cell responses by vaccination has not been helpful in immunocompromised hosts. Immunotherapy with CMV specific donor-derived cytotoxic T lymphocytes (CTL) is effective after allografting, but it is expensive, labor intensive, and diffcult to replicate in most centers. Non-toxic targeted therapy is needed to improve clinical outcomes. In earlier studies, we generated anti-CD3 chemically heteroconjugated with anti-Her2/neu or anti-CD20 bispecific antibody (BiAb). Ex vivo expanded anti-CD3 activated T cells (ATC) and armed with BiAb exhibit high levels of specific cytotoxicity directed at the respective tumor antigens. We propose a novel strategy of using ATC armed ex-vivo with engineered CMVBi to target CMV antigens. We tested the strategy initially in an in vitro cell culture model using CMV-infected fibroblasts and T cells from normal human donors. We hypothesize that donor anti-CD3 ATC armed with CMVBi will target and eliminate CMV-infected target cells. Materials and methods Normal donor peripheral blood mononuclear cells (PBMC) were used to generate ATC by activation with anti-CD3 (OKT3) and interleukin 2 (IL-2). CMVBi was created by chemical heteroconjugation of OKT3 (murine IgG2a) monoclonal antibody and polyclonal anti-CMV (Cytogam®). Specific cytotoxicity was tested by 51Cr release assay using CMV-infected or uninfected human fibroblasts as target cells. Effector populations tested included CMVBi armed ATC and ATC alone; additional controls included CMVBi alone, Cytogam® alone, CMVBi armed, and unarmed PBMC. Cytotoxicity was assessed for CMVBi and an irrelevant BiAb at arming doses ranging from 1 to 500 ng/106 ATC with effector to target ratios (E:T) ranging from 25:1 to 3.125:1. Interferon gamma (IFNγ) EliSpots were used to determine cytokine response after exposing CMV-infected and uninfected fibroblasts to unarmed and CMVBi-armed ATC. Results CMVBi arming with as little as 1 ng/106 ATC was significantly more cytotoxic for target cells than unarmed ATC. There was an incremental increase in cytotoxicity with CMVBi armed ATC as the mutiplicity of infection (MOI) was increased in target cells. At all E:T ratios (25:1, 12.5:1, 6.25:1, 3.125:1 ), ATC armed with a dose of 50 ng/106 CMVBi demonstrated markedly enhanced killing of CMV-infected targets (MOI 1) compared to unarmed ATC (see table). In the uninfected control cells, both unarmed and armed ATC lysis was barely detectable over spontaneous lysis. Immunofluorescent studies showed that CMVBi-armed ATC specifically aggregated around GFP fluorescence-marked CMV-infected fibroblasts, whereas unarmed ATC did not aggregate. Cytokine secretion analyzed by IFNγ EliSpot confirmed the superior cytotoxicity of CMVBi-armed ATC. Conclusion We used polyclonal Cytogam® to make CMVBi-armed ATC that were able to kill target cells expressing various CMV antigens with high specificity. CMVBi-armed PBMC was only minimally cytotoxic in comparision to CMVBi-armed ATC. Hence, infusion of CMVBi alone to patients is unlikely to be as effective as infusion of ATC armed ex-vivo with CMVBi. We demonstrated similar degree of cytotoxicity using different donor ATC including CMV sero-negative donors. This effect is independent of MHC mediated antigen presentation hence, overcomes CMV immune escape. This non-MHC restricted specific killing strategy could be easily adapted for the prevention and/or treatment of CMV infection/disease after allogeneic SCT using donor-derived ATC. Our current use of BiAb-armed ATC for cancer immunotherapy in humans illustrates the feasibility of adoptive immunotherapy with CMVBi-armed ATC in SCT. Disclosures Lum: Transtarget Corporation: Founder.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A189-A189
Author(s):  
Shannon Oda ◽  
Kristin Anderson ◽  
Philip Greenberg ◽  
Nicolas Garcia ◽  
Pranali Ravikumar ◽  
...  

BackgroundAdoptive cell therapy (ACT) with genetically-modified T cells has shown impressive results against some hematologic cancers, but limited efficacy against tumors with restrictive tumor microenvironments (TMEs). FasL is a particular obstacle for ACT;1 it is expressed in many tumors and TMEs,1 including AML,2 ovarian3 and pancreatic cancers,4 and upregulated on activated T cells, where it can mediate activation-induced cell death (AICD).5MethodsWe engineered T cells to boost function with novel immunomodulatory fusion proteins (IFPs) that combine an inhibitory ectodomain with a costimulatory endodomain. Like current checkpoint-blocking therapies, IFPs can abrogate an inhibitory signal, but also provide an often absent costimulatory signal. Additionally, IFP-driven signals are delivered only to the T cells concurrently engineered to be tumor-specific, thereby avoiding systemic T cell activation. For FasL-expressing TMEs, we developed an IFP that replaces the Fas intracellular tail with costimulatory 4-1BB. We tested the the Fas-4-1BB IFP in primary human T cells and in immunocompetent murine models of leukemia and pancreatic cancer.ResultsFas-4-1BB IFP expression enhanced primary human T cell function and enhanced lysis of Panc1 pancreatic tumor cells in vitro. Fas-4-1BB IFP-engineered murine T cells exhibited increased pro-survival signaling, proliferation, antitumor function and altered metabolism in vitro. Notably, the Fas ectodomain is trimeric5 and the 4-1BB intracellular domain requires trimerization to signal.6 In contrast, the CD28 domain is dimeric and did not enhance function when paired with 4-1BB.In vivo, Fas-4-1BB increased T cell persistence and function, and Fas-4-1BB T cell ACT significantly improved survival in a murine AML model. When delivered with a mesothelin-specific TCR, Fas-4-1BB T cells prolonged survival in the autochthonous KPC pancreatic cancer model, increasing median survival to 65 from 37 days (with TCR-only, **P=0.0042). Single-cell RNA sequencing revealed differences in the endogenous tumor-infiltrating immune cells, included changes in cell frequency and programming.ConclusionsWe developed an engineering approach to enhance the in vivo persistence and antitumor efficacy of transferred T cells. Our targeted, two-hit strategy uses a single fusion protein to overcome a death signal prevalent in the TME of many cancers and on activated T cells, and to provide a pro-survival costimulatory signal to T cells. Our results suggest that this fusion protein can increase T cell function when combined with murine or human TCRs, and can significantly improve therapeutic efficacy in liquid and solid tumors, supporting clinical translation.ReferencesYamamoto, T.N., et al., T cells genetically engineered to overcome death signaling enhance adoptive cancer immunotherapy. J Clin Invest 2019.Contini P, et al., In vivo apoptosis of CD8(+) lymphocytes in acute myeloid leukemia patients: involvement of soluble HLA-I and Fas ligand. Leukemia 2007;21(2):p. 253–60.Motz GT, et al., Tumor endothelium FasL establishes a selective immune barrier promoting tolerance in tumors. Nat Med 2014;20(6):p. 607–15.Kornmann M, et al., Fas and Fas-ligand expression in human pancreatic cancer. Ann Surg 2000. 231(3): p. 368–79.Villa-Morales M and J Fernandez-Piqueras, Targeting the Fas/FasL signaling pathway in cancer therapy. Expert Opin Ther Targets 2012;16(1):p. 85–101.Wyzgol, A., et al., Trimer stabilization, oligomerization, and antibody-mediated cell surface immobilization improve the activity of soluble trimers of CD27L, CD40L, 41BBL, and glucocorticoid-induced TNF receptor ligand. J Immunol 2009;183(3):p. 1851–61.


1999 ◽  
Vol 190 (11) ◽  
pp. 1561-1572 ◽  
Author(s):  
Haruhiko Suzuki ◽  
Yan Wen Zhou ◽  
Masashi Kato ◽  
Tak W. Mak ◽  
Izumi Nakashima

Although interleukin 2 (IL-2) has been thought to be the most important cytokine for T cell growth, animals lacking IL-2 or a component of its receptor molecules have more expanded T cells with activated memory phenotype, indicating an indispensable role for the IL-2/IL-2 receptor system in regulating the size and activity of the T cell population. In this study, we investigated the possible mechanism of abnormal expansion of activated T cells in IL-2 receptor β chain (IL-2Rβ)−/− mice using the systems of bone marrow transplantation and T cell transfer. Here, we show that IL-2Rβ2/− T cells in mice reconstituted with a mixture of IL-2Rβ2/− and IL-2Rβ1/+ bone marrow cells did not develop into an abnormally activated stage, and that already activated IL-2Rβ2/− T cells were effectively eliminated by IL-2Rβ1/+ T cells when both cells were cotransferred to T cell–deficient host mice. This regulation and/or elimination was dependent on T cells bearing α/β type T cell receptor, especially on CD8+ T cells and independent of the Fas–Fas ligand (FasL) system. IL-2Rβ1/+ T cells that eliminated activated IL-2Rβ2/− T cells expressed FasL, perforin, granzyme B, and tumor necrosis factor α/β. These results indicate a novel function of IL-2Rβ that is necessary for the induction of regulatory T cells acting to eliminate activated T cells.


1990 ◽  
Vol 49 (1) ◽  
pp. 198-200 ◽  
Author(s):  
GERD WALZ ◽  
BERND ZANKER ◽  
JOHN R. MURPHY ◽  
TERRY B. STROM

2000 ◽  
Vol 20 (2) ◽  
pp. 702-712 ◽  
Author(s):  
Chi-Wing Chow ◽  
Roger J. Davis

ABSTRACT Calcium-stimulated nuclear factor of activated T cells (NFAT) transcription activity at the interleukin-2 promoter is negatively regulated by cyclic AMP (cAMP). This effect of cAMP is mediated, in part, by protein kinase A phosphorylation of NFAT. The mechanism of regulation involves the creation of a phosphorylation-dependent binding site for 14-3-3. Decreased NFAT phosphorylation caused by the calcium-stimulated phosphatase calcineurin, or mutation of the PKA phosphorylation sites, disrupted 14-3-3 binding and increased NFAT transcription activity. In contrast, NFAT phosphorylation caused by cAMP increased 14-3-3 binding and reduced NFAT transcription activity. The regulated interaction between NFAT and 14-3-3 provides a mechanism for the integration of calcium and cAMP signaling pathways.


1998 ◽  
Vol 186 (2) ◽  
pp. 103-110 ◽  
Author(s):  
Yan-Wen Zhou ◽  
Yoshihiro Komada ◽  
Hiroto Inaba ◽  
Eiichi Azuma ◽  
Minoru Sakurai

Cartilage ◽  
2016 ◽  
Vol 8 (3) ◽  
pp. 300-306 ◽  
Author(s):  
Satomi Abe ◽  
Hitoshi Nochi ◽  
Hiroshi Ito

Introduction We previously showed that articular chondrocytes (ACs) have immune privilege and immunomodulatory functions like those of mesenchymal stem cells. To elucidate these mechanisms, we focused on interleukin-2 (IL-2), which plays critical roles in lymphocyte mitogenic activity. The purpose of this study was to explore whether ACs affect the role of IL-2 underlying immunomodulatory functions. Material and Methods Irradiated human ACs from osteoarthritis donors were used. Third-party ACs were added to the mixed lymphocyte reaction (MLR) with or without recombinant human IL-2 (rhIL-2), and the levels of IL-2 and the soluble form of the IL-2 receptor α (sIL-2Rα) protein in supernatant were measured by enzyme-linked immunosorbent assay. Recombinant human IL-2 (rhIL-2) was also added to the MLR. To detect the expression of IL-2 receptor α (CD25) on lymphocytes in the MLR, flow cytometric analysis was performed. Last, ACs and allogeneic activated CD4+ T cell were co-cultured, and the expression of CD25 on activated T cells was examined by flow cytometry. Results Third-party ACs significantly inhibited the MLR and reduced the level of sIL-2Rα in a dose-dependent manner, but did not affect the concentration of IL-2. Exogenous rhIL-2 accelerated MLR but did not rescue the inhibitory effect of ACs. ACs inhibited the expression of CD25 on activated CD4+ T cells. Discussion Our results showed that third-party ACs inhibited the proliferation of allogeneic activated lymphocytes, thereby inhibiting production sIL-2Rα, although ACs did not affect IL-2 secretion from lymphocytes. Also, ACs inhibited CD25 expression on activated CD4+ T cells. Thus, ACs inhibited the immune response of allogeneic lymphocytes by inducing IL-2 nonresponsiveness.


1995 ◽  
Vol 181 (1) ◽  
pp. 71-77 ◽  
Author(s):  
M R Alderson ◽  
T W Tough ◽  
T Davis-Smith ◽  
S Braddy ◽  
B Falk ◽  
...  

A significant proportion of previously activated human T cells undergo apoptosis when triggered through the CD3/T cell receptor complex, a process termed activation-induced cell death (AICD). Ligation of Fas on activated T cells by either Fas antibodies or recombinant human Fas-ligand (Fas-L) also results in cytolysis. We demonstrate that these two pathways of apoptosis are causally related. Stimulation of previously activated T cells resulted in the expression of Fas-L mRNA and lysis of Fas-positive target cells. Fas-L antagonists inhibited AICD of T cell clones and staphylococcus enterotoxin B (SEB)-specific T cell lines. The data indicate AICD in previously stimulated T cells is mediated by Fas/Fas-L interactions.


Sign in / Sign up

Export Citation Format

Share Document