scholarly journals MEK Inhibition Results In Selective Immunosuppression Of Human T Cells And Decreases Cytokine Production In A Memory Stage-Dependent Manner

2010 ◽  
Vol 16 (2) ◽  
pp. S302
Author(s):  
T.K. Kim ◽  
M. Valencia Gattas ◽  
C.L. Benjamin ◽  
R.B Levy ◽  
E.D. Wieder ◽  
...  
Blood ◽  
2013 ◽  
Vol 121 (23) ◽  
pp. 4617-4626 ◽  
Author(s):  
Takero Shindo ◽  
Tae Kon Kim ◽  
Cara L. Benjamin ◽  
Eric D. Wieder ◽  
Robert B. Levy ◽  
...  

Key Points RAS/MEK/ERK signaling is memory stage-dependent in human T cells, conferring susceptibility to alloreactive T-cell selective inhibition. MEK inhibitors selectively inhibit alloreactive but not herpesvirus-specific human T cells and inhibit murine GVHD.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 4014-4014
Author(s):  
Takero Shindo ◽  
Tae Kon Kim ◽  
Cara L. Benjamin ◽  
Robert B. Levy ◽  
Eric D. Wieder ◽  
...  

Abstract Abstract 4014 Background: In hematopoietic stem cell transplantation, most immunosuppressive strategies (e.g., using calcineurin inhibitors or corticosteroids) decrease graft-versus-host disease (GVHD) rates, but also impair pathogen- and cancer-specific T cells, increasing treatment-related mortality. Consequently, a key goal of transplant immunology is the identification of more selective inhibitors of alloreactivity. Recent murine studies have shown that alloreactive T cells mediating GVHD reside primarily in naïve and early memory T cell compartments (Anderson BE et al. J Clin Invest 2003;112:101–108, Chen BJ et al. Blood 2004;103:1534–1541, Zheng H et al. J Immunol 2009;182:5938–5948). In contrast, human virus-specific T cells are more differentiated (Gamadia LE et al. Blood 2001;98:754–761). We recently demonstrated that the RAS/MEK/ERK pathway is preferentially activated in naïve and early memory T cells, relative to late memory T cells (Kim TK, submitted). This pathway is activated in a murine experimental GVHD (Lu SX et al. Blood 2008; 112: 5254–5258). Therefore, we hypothesized that MEK inhibition would prove to be a more selective immunosuppressive strategy, suppressing alloreactive but not virus-specific T cells. Design: Using PBMC from healthy donors, we assessed the effects of several MEK inhibitors or the calcineurin inhibitor (CNI) tacrolimus on functional T cell responses using flow cytometry and/or western blotting. Using single-cell cytometric methods, we examined intracellular phosphorylation of ERK1/2 in response to PMA and Ionomycin. Alloreactivity induced by allogeneic monocyte-derived dendritic cell (DC) stimulation was also investigated. CMV-specific functional T cell responses following stimulation using pp65 pentadecapeptide pools were assessed using cytokine flow cytometry. To assess selectivity of immunosuppressive agents, we examined the effects of CNI and MEK inhibitors, independently and in combination, on alloreactivity and CMV-specific T cell responses. Results: We first confirmed that multiple RAS/MEK/ERK family proteins were differentially expressed with lymphocyte maturation, suggesting that MEK inhibitors would preferentially suppress naïve and early memory T cells. Consistent with this prediction, ERK1/2 was preferentially phosphorylated in naïve and early memory T cells stimulated with PMA/Ionomycin, relative to intermediate and late memory T cells. When we examined the effects of targeted T cell inhibition on alloreactive and virus-specific T cell responses, we confirmed that: 1) MEK inhibitors blocked phosphorylation of ERK1/2 in CD4 and CD8 T cells stimulated with PMA/Ionomycin at 100nM to 10μM; 2) Alloreactivity of T cells was suppressed as effectively by MEK inhibitors as by tacrolimus (see Figure 1, left panels). In addition, functional differentiation of naïve T cells was also suppressed. Furthermore, MEK inhibition and calcineurin inhibition worked synergistically and completely shut off alloreactivity. 3) MEK inhibition suppressed cytokine production in naïve and early memory T cells, but not in late memory T cells, while calcineurin inhibition suppressed all differentiation subsets. 4) At doses effectively suppressing DC-stimulated alloreactivity, MEK inhibition spared CMV-specific T cells, in contrast to tacrolimus (p <0.01) (see Figure 1, right panels). Conclusion: MEK inhibitors suppress T cells in a memory stage-dependent manner, suggesting a novel paradigm to achieve selective immunity in the transplant setting. MEK inhibitors abrogate alloreactivity while sparing the function of CMV-specific CD4 and CD8 T cells. These data suggest that MEK inhibitors may serve as a new class of agents with potential activity in GVHD, facilitating selective inhibition of alloreactivity by targeting T cells in a memory stage-dependent manner. Disclosures: No relevant conflicts of interest to declare.


2020 ◽  
Vol 38 (15_suppl) ◽  
pp. e18050-e18050
Author(s):  
Ben Buelow ◽  
Brian Avanzino ◽  
Aarti Balasubramani ◽  
Andrew Boudreau ◽  
Laura Davison ◽  
...  

e18050 Background: Ovarian Cancer (OvCa) is the leading cause of gynecologic cancer mortality in women. Since the introduction of platinum-based chemotherapy there has been little change in the prognosis of OvCa patients, with < 30% overall survival in advanced disease, creating an urgent medical need for novel therapies. Few ovarian epithelium-specific surface proteins are suited for Ab targeting. However, studies have shown folate receptor α (FRα) to be highly over-expressed in OvCa; expression level and stage correlate, and FRα is absent or minimally expressed in normal tissues. However, naked Ab therapy has shown limited efficacy while CAR-T therapy has been plagued by toxicity and limited efficacy. ADCs have demonstrated some activity but present the risk of toxin-mediated side effects. Using Teneobio’s unique antibody discovery platform, we have developed a CD3 x FRα T-BsAb that retains the potent cytotoxicity of other T-cell redirecting therapies but with significantly reduced cytokine release. Methods: Antibodies targeting CD3 and FRα were generated via immunization of our proprietary transgenic animals. Candidate antibodies were selected by repertoire deep sequencing of B-cells from draining lymph nodes, high-throughput gene assembly, recombinant expression, and functional screening. Bispecific antibodies targeting CD3 and FRα were assembled and evaluated for their ability to selectively activate primary human T-cells and mediate killing of FRα+ tumor cells in vitro and in vivo. T-cell activation surface markers, cytokine production and tumor cell cytotoxicity were measured. Results: Primary human T-cells were activated only in the presence of both the CD3 x FRα T-BsAb and FRα (either recombinant or cell-surface protein). Potent and selective cytotoxicity against FRα+ tumor cells was observed in co-cultures of primary human T-cells and OvCa tumor cell lines. Strikingly, our T-BsAb mediated comparable tumor cell cytotoxicity to CD3 x FRα T-BsAbs containing a high affinity anti-CD3 domain but with significantly reduced cytokine production. Our Ab showed preliminary evidence of tumor growth inhibition in xenograft models of OvCa in vivo. Conclusions: We have created a novel CD3 x FRα T-BsAb that mediates T-cell killing of FRα+ tumor cells with minimal production of cytokines. This molecule may improve safety, efficacy, and offer opportunities for combination therapy to treat OvCa.


Blood ◽  
2001 ◽  
Vol 98 (4) ◽  
pp. 1239-1245 ◽  
Author(s):  
Kuo-Jang Kao ◽  
Eileen S. Huang ◽  
Sandra Donahue

Transfusions of UV-B–irradiated peripheral blood mononuclear cells (UV-B–PBMCs) from BALB/c (H-2d) mice into CBA (H-2k) mice can induce humoral immune tolerance to H-2d antigens, and the induced tolerance is partially mediated by negative regulatory PBMCs. To further identify which subset of spleen mononuclear leukocytes (MNLs) in the tolerant CBA mice is responsible for the negative regulatory activity, adoptive transfer experiments were conducted using spleen MNLs from the tolerant CBA mice. Results showed that only CD4+ T cells could transfer the negative regulatory activity in a dose-dependent manner. This negative regulatory activity was significantly reduced when CD25+ helper T cells were removed. Further study suggested that inhibition of IL-12 production by UV-B–irradiated PBMCs played a role in the induction of immune tolerance. In vitro study of the cytokine production profile by CBA CD4+ T cells, after stimulation with gamma-irradiated BALB/c spleen cells, revealed an enhanced production of the type 2 T-cell cytokines after tolerance induction. Induction of tolerance also prevented the development of cytotoxic T cells in CBA mice against BALB/c MNLs. Adoptive transfer study suggested that the cellular immune tolerance was also mediated by CD4+ negative regulatory T cells. The induced immune tolerance was nullified after 400 cGy sublethal gamma irradiation. These results suggest that the ex vivo study of cytokine production by T cells may be used to monitor tolerance induction and the selection of gamma radiation dose is critical for potential clinical application of the tolerance induced by UV-B–PBMCs.


Blood ◽  
2010 ◽  
Vol 115 (11) ◽  
pp. 2203-2213 ◽  
Author(s):  
Dalya R. Soond ◽  
Elisa Bjørgo ◽  
Kristine Moltu ◽  
Verity Q. Dale ◽  
Daniel T. Patton ◽  
...  

Abstract We have previously described critical and nonredundant roles for the phosphoinositide 3-kinase p110δ during the activation and differentiation of naive T cells, and p110δ inhibitors are currently being developed for clinical use. However, to effectively treat established inflammatory or autoimmune diseases, it is important to be able to inhibit previously activated or memory T cells. In this study, using the isoform-selective inhibitor IC87114, we show that sustained p110δ activity is required for interferon-γ production. Moreover, acute inhibition of p110δ inhibits cytokine production and reduces hypersensitivity responses in mice. Whether p110δ played a similar role in human T cells was unknown. Here we show that IC87114 potently blocked T-cell receptor–induced phosphoinositide 3-kinase signaling by both naive and effector/memory human T cells. Importantly, IC87114 reduced cytokine production by memory T cells from healthy and allergic donors and from inflammatory arthritis patients. These studies establish that previously activated memory T cells are at least as sensitive to p110δ inhibition as naive T cells and show that mouse models accurately predict p110δ function in human T cells. There is therefore a strong rationale for p110δ inhibitors to be considered for therapeutic use in T-cell–mediated autoimmune and inflammatory diseases.


2010 ◽  
Vol 71 (1) ◽  
pp. 23-28 ◽  
Author(s):  
Tae Kon Kim ◽  
Matthew J. Billard ◽  
Eric D. Wieder ◽  
Bradley W. McIntyre ◽  
Krishna V. Komanduri

2019 ◽  
Author(s):  
Nicola Trendel ◽  
Philipp Kruger ◽  
Stephanie Gaglione ◽  
John Nguyen ◽  
Johannes Pettmann ◽  
...  

AbstractMaintaining and limiting T cell responses to constant antigen stimulation is critical to control pathogens and maintain self-tolerance, respectively. Antigen recognition by T cell receptors (TCRs) induces signalling that activates T cells to produce cytokines and also leads to the downregulation of surface TCRs. In other systems, receptor downregulation can induce perfect adaptation to constant stimulation by a mechanism known as state-dependent inactivation that requires complete downregulation of the receptor or the ligand. However, this is not the case for the TCR, and therefore, precisely how TCR downregulation maintains or limits T cell responses is controversial. Here, we observed that in vitro expanded primary human T cells exhibit perfect adaptation in cytokine production to constant antigen stimulation across a 100,000-fold variation in affinity with partial TCR downregulation. By directly fitting a mechanistic model to the data, we show that TCR downregulation produces imperfect adaptation, but when coupled to a switch produces perfect adaptation in cytokine production. A pre-diction of the model is that pMHC-induced TCR signalling continues after adaptation and this is confirmed by showing that, while costimulation cannot prevent adaptation, CD28 and 4-1BB signalling reactivated adapted T cells to produce cytokines in a pMHC-dependent manner. We show that adaptation also applied to 1st generation chimeric antigen receptor (CAR)-T cells but is partially avoided in 2nd generation CARs. These findings high-light that even partial TCR downregulation can limit T cell responses by producing perfect adaptation rendering T cells dependent on costimulation for sustained responses.


Sign in / Sign up

Export Citation Format

Share Document