scholarly journals 2456 Cutaneous lupus erythematosus patients have increased circulating myeloid-derived suppressor cells with immunosuppressive properties

2018 ◽  
Vol 2 (S1) ◽  
pp. 7-8
Author(s):  
Stephanie Florez-Pollack ◽  
Lin-chiang Tseng ◽  
Masato Kobayashi ◽  
Benjamin F. Chong ◽  
Kiyoshi Ariizumi

OBJECTIVES/SPECIFIC AIMS: MDSCs are potent suppressors of T cell function, and have been recently found to be implicated in skin diseases driven by T cell dysregulation. However, the function of MDSCs in CLE is poorly understood. We sought to characterize the MDSC population in the peripheral blood of DLE patients and evaluate their ability to suppress autologous T cells. METHODS/STUDY POPULATION: All patients were recruited through the UT Southwestern Cutaneous Lupus Registry. PBMCs from 32 CLE patients and 16 age-matched and gender-matched controls were analyzed using flow cytometry. Monocytic MDSCs were identified by the phenotype of CD14+ HLA-DR neg/low. Furthermore, autologous MDSCs and T cells were purified from CLE PBMCs (n=4) and co-cultured at different ratios of these cells. T cell function was measured by secretion of IFN-γ by ELISA. RESULTS/ANTICIPATED RESULTS: Monocytic MDSCs in CLE PBMCs (median: 2.04%, IQR: 0.67%–5.07%) were significantly higher compared with healthy control PBMCs (median: 0.5%, IQR: 0.1%–1.07%, p=0.002). Although not significant on subset analysis, patients with CLE limited to the head and neck had the highest levels of MDSCs. CLE MDSCs (n=4) were found to suppress autologous activated T-cells in a dose-dependent manner. DISCUSSION/SIGNIFICANCE OF IMPACT: In this cross-sectional study of patients of the UT Southwestern Cutaneous Lupus Registry, we observed differences in the levels of MDSCs among PBMCs of CLE patients Versus healthy controls. CLE patients had significantly higher levels of MDSCs, which could be explained by the presence of an inflammatory state in this group. Furthermore, CLE MDSCs were able to suppress autologous T cells, showing that these cells are functionally patent in CLE blood. Their up-regulation in CLE blood may represent the body’s response to limiting disease severity, since most patients had mild disease activity.

Molecules ◽  
2015 ◽  
Vol 20 (10) ◽  
pp. 19014-19026 ◽  
Author(s):  
Masao Goto ◽  
Manabu Wakagi ◽  
Toshihiko Shoji ◽  
Yuko Takano-Ishikawa

2001 ◽  
Vol 193 (3) ◽  
pp. 317-328 ◽  
Author(s):  
Kanaga Sabapathy ◽  
Tuula Kallunki ◽  
Jean-Pierre David ◽  
Isabella Graef ◽  
Michael Karin ◽  
...  

Apoptotic and mitogenic stimuli activate c-Jun NH2-terminal kinases (JNKs) in T cells. Although T cells express both JNK1 and JNK2 isozymes, the absence of JNK2 alone can result in resistance to anti-CD3–induced thymocyte apoptosis and defective mature T cell proliferation. Similar defects in thymocyte apoptosis and mature T cell proliferation, the latter due to reduced interleukin 2 production, are also caused by JNK1 deficiency. Importantly, T cell function was compromised in Jnk1+/−Jnk2+/− double heterozygous mice, indicating that JNK1 and JNK2 play similar roles in regulating T cell function. The reduced JNK dose results in defective c-Jun NH2-terminal phosphorylation in thymocytes but not in peripheral T cells, in which nuclear factors of activated T cells (NK-ATs)–DNA binding activity is affected. Thus, JNK1 and JNK2 control similar functions during T cell maturation through differential targeting of distinct substrates.


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.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2567-2567
Author(s):  
Nicole Stephens ◽  
Sawa Ito ◽  
Stephen A. Strickland ◽  
Bipin N. Savani ◽  
Madan Jagasia ◽  
...  

Abstract Between presentation and remission of AML, loss of leukemia burden and the recovery of normal hematopoiesis are likely to be associated with major changes in cytokine profiles which could inform pathophysiology of hematopoiesis and immune recovery and may be predictive for outcome. However cytokine fluctuations in AML before and after induction chemotherapy are not well characterized. To profile the cytokine signatures of patients with AML, we analyzed 57 cytokines, chemokines, and growth factors in blood of 11 patients with AML (mean age 58 years; 31-69) undergoing conventional remission induction chemotherapy enrolled into an investigational study (VICCHEM 1073). Plasma was obtained from heparinized peripheral blood collected at onset of leukemia, 8-14 days, and 22-35 days after the initiation of induction chemotherapy and also from 12 healthy donors. Cytokine levels were measured in duplicate using magnetic beads based Luminex assay (Affymetrix, CA, USA). Compared with normal controls, 5 cytokine patterns were observed. i) levels significantly lower at onset of leukemia, and lowest 8-14 days after induction correlating with lymphocyte count: GM-CSF, M-CSF, PDGF-AA, EGF, FGF basic, IL1b, IL-2,IL4, IL10, IL12p40, IL12p70, IL13, IL15, IL17a, IL22, IL23p19, TNF beta, TNF alpha, IFN alpha, IFN gamma, TGF alpha, MCP3, LIF, Granzyme B, sFAS ligand, TRAIL, (p<0.05). Most of these cytokines are predominantly produced by T cells or other immune cells. ii) levels significantly higher in AML through chemotherapy induction and recovery: IL-27 (p=0.002), MPO, IL2Ra, IL-21, , IP-10, MIG, MIP1 alpha, SDF-1, MCP1(p<0.05), HGF (p=0.05), VEGF (p=0.07), IL1Ra (p=0.058). These cytokines are predominantly produced by stromal cells. iii) levels significantly higher at the onset of leukemia and correlating with lymphocyte count: CD40 ligand (p<0.05). iv) levels significantly lower at onset of leukemia but inversely correlated with lymphocyte count; Flt3-ligand, sFAS (p<0.05). v) No significant differences and fluctuation: NGF, GRO alpha, IL1a, IL3, IL5, IL6, IL7, IL8, IL9, MIP1b, SCF. Among the cytokines persistently elevated in AML, IL-27 was significantly higher in patients who did not achieve complete remission after induction chemotherapy (p=0.03). To investigate the biological consequences of elevated IL-27 in the AML microenvironment, we examined the effect of IL-27 on T-cell function. Previous studies in mice show that IL-27 rapidly induces PD-L1 expression on naïve CD4+ T-cells. Human CD4+ naïve and CD4+ cells were isolated from healthy volunteers (n=4) according to RoboSep protocols (Stemcell Technologies, Vancouver, Canada), then incubated with IL-27 or IL-6 for up to 72 hours. IL-27 was found to induce PD-L1 expression in a time and dose-dependent manner especially in CD4+ naïve and central memory populations. These findings support other findings that AML suppresses protective antileukemic immune responses and cause T cell exhaustion. IL-27 production induced by AML cells may explain exhaustion of CD4+ T-cells through increased PD-L1 expression. Targeting IL-27 may improve immune function in AML and lead to better survival. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 1039-1039
Author(s):  
Matthew B. Fletcher ◽  
Maria Elena Ramirez ◽  
Rosa Sierra ◽  
Patrick Raber ◽  
Paulo Rodriguez

Abstract Recent studies have suggested the relevance of different energy metabolic pathways in the balance between protective T cell immunity and T cell anergy in tumors. We and others have suggested the role of the depletion of the non-essential amino acid L-arginine as a mechanism for the induction of T cell suppression in tumors. Therefore, we hypothesize that it is possible to metabolically regulate T cell responses simply through the modulation of L-arginine. In this study, we aimed to determine the effect of a pegylated form of the human L-arginine-metabolizing enzyme arginase I (peg-Arg I) in T cell responses. Activation of naïve murine T cells and antigen-specific CD4+ or CD8+ T cells in the presence of peg-Arg I prevented cell proliferation and production of IFNγ in vitro and in vivo. Similarly, peg-Arg I impaired proliferation and IFNg production in T cells activated with PMA/Ionomycin, suggesting that the effect of peg-Arg I was independent of T cell receptor (TCR) signaling. In fact, the anti-proliferative effect induced by peg-Arg I correlated with an arrest of T cells in the G0-G1 phase of the cell cycle, a decreased expression of cyclin D3 and cdk4, and a major inhibition of de novo translation. Interestingly, treatment of T cells with peg-Arg I did not impair the expression of the activation marker CD69, which correlated with an intact mitochondrial biogenesis. As a result, peg-Arg I did not have an effect in oxygen consumption by mitochondrial respiration (OCR), but significantly blocked glycolytic pathways in activated T cells. Furthermore, peg-Arg I treated T cells increased the expression of genes associated with integrated stress responses (IRS) and arrest in translation including GCN2, Chop, and Atf4. We then tested the effect of peg-Arg I in mouse models of graft versus host disease (GVHD) and inflammatory bowel disease (IBD), both mediated through activated T cells. Peg-Arg I significantly extended the survival of mice in these 2 disease models, which was associated with a decreased production of IFNg. Altogether, these results suggest the potential effect of the modulation of the metabolism of L-arginine as a means to regulate T cell responses. Continuation of this study will advance in the understanding of the metabolic effects of L-arginine in T cell function, which could enable the development of therapies to modulate T cell responses in transplantation or autoimmunity. Disclosures: No relevant conflicts of interest to declare.


2021 ◽  
Vol 4 (1) ◽  
Author(s):  
Jeremy To ◽  
Doug Quackenbush ◽  
Emily Rowell ◽  
Lilin Li ◽  
Connor Reed ◽  
...  

AbstractOvercoming tumor-mediated immunosuppression and enhancing cytotoxic T-cell activity within the tumor microenvironment are two central goals of immuno-oncology (IO) drug discovery initiatives. However, exploratory assays involving immune components are often plagued by low-throughput and poor clinical relevance. Here we present an innovative ultra-high-content assay platform for interrogating T-cell-mediated killing of 3D multicellular tumor spheroids. Employing this assay platform in a chemical genomics screen of 1800 annotated compounds enabled identification of small molecule perturbagens capable of enhancing cytotoxic CD8+ T-cell activity in an antigen-dependent manner. Specifically, cyclin-dependent kinase (CDK) and bromodomain (BRD) protein inhibitors were shown to significantly augment anti-tumor T-cell function by increasing cytolytic granule and type II interferon secretion in T-cells in addition to upregulating major histocompatibility complex (MHC) expression and antigen presentation in tumor cells. The described biotechnology screening platform yields multi-parametric, clinically-relevant data and can be employed kinetically for the discovery of first-in-class IO therapeutic agents.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 3032-3032
Author(s):  
Arantxa Romero-Toledo ◽  
Robin Sanderson ◽  
John G. Gribben

The complex crosstalk between malignant chronic lymphocytic leukemia (CLL) cells and the tumor microenvironment (TME) is not fully understood. CLL is associated with an inflammatory TME and T cells exhibit exhaustion and multiple functional defects, fully recapitulated in Eµ-TCL1 (TCL1) mice and induced in healthy mice by adoptive transfer (AT) of murine CLL cells, making it an ideal model to test novel immunotherapies for this disease. Myeloid-derived suppressor cells (MDSCs), a non-leukemic cell type within the TME, are immature myeloid cells with the ability to suppress T cell function and promote Treg expansion. In humans, CLL cells can induce conversion of monocytes to MDSCs provoking their accumulation in peripheral blood (PB). MDSCs include two major subsets granulocytic (Gr) and monocytic (M)-MDSC. In mice, Gr-MDSCs are defined as CD11b+Ly6G+Ly6Clo and M-MDSC as CD11b+Ly6G-Ly6Chi. Both murine and human MDSCs express BTK. We observed that in CLL-bearing mice, MDSCs cells are lost in PB as disease progresses. Treatment with both BTK inhibitors (BTKi), ibrutinib (Ibr) and acalabrutinib (Acala), result in shift of T cell function from Th2 towards Th1 polarity and increase MDSC populations in vivo. We aimed to determine whether combination treatment with BTKi and chimeric antigen receptor (CAR) T cells renders recovery of the MDSC population in CLL-bearing mice. To address this question we designed a two-part experiment, aiming to mimic the clinically relevant scenario of pre-treatment of CLL with BTKi to improve CAR T cell function. Part 1 of our experiment consisted of 4 groups (n=12) of 2.5 month old C57/Bl6 mice. Three groups had AT with 30x106 TCL1 splenocytes. A fourth group of WT mice remained CLL-free as a positive control and donors for WT T cells. When PB CLL load reached >10% (day 14) animals were randomized to either Ibr or Acala at 0.15 mg/l in 2% HPBC or no treatment for 21 days. All animals from part 1 were culled at day 35 post-AT and splenic cells were isolated, analyzed and used to manufacture CAR T cells. WT, CLL, Ibr and Acala treated T cells were activated and transduced with a CD19-CD28 CAR to treat mice in part 2. Here, 50 WT mice were given AT with 20x106 TCL1 splenocytes for CLL engraftment. All mice were injected with lymphodepleting cyclophosphamide (100mg/kg IP) one day prior to IV CAR injection. At day 21 post-AT, mice were treated with WT CAR, CLL CAR, IbrCAR, AcalaCAR or untransduced T cells. MDSC sub-populations were monitored weekly in PB and SP were analysed by flow cytometry. As malignant CD19+CD5+ cells expands in PB, the overall myeloid (CD19-CD11b+) cell population was not affected, but MDSCs significantly decreased (p<0.0001). Treatment with Acala, but not Ibr restores total MDSCs. However, MDSC impairment occurs in the Gr- but not M- MDSC population and both Acala and Ibr restores this population (Figure 1a). When we examined the spleen, treatment with both Ibr (p<0.001) and Acala (p<0.001) reduced CD5+CD19+ cells, whereas neither BTKi affected the overall myeloid (CD19-CD11b+) cell population. Gr-MDSCs were restored by both treatments whilst M-MDSCs were only restored after Ibr treatment (p<0.001 in each case). In part 2 of this experiment we observed that treatment with all CAR-T cell groups provokes the clearance of all CD19+CD5+ cells. The overall CD19-CD11b+ population stays the same across all mice groups 35 days after treatment in PB with any group of CAR and untransduced T cells. Overall MDSC population is maintained following all CAR T cells compared to CLL-bearing mice (p<0.0001) and it is the Gr- but not the M- MDSC population which is recovered in PB (Figure 1b). These parts of the experiments can of course be influenced by treatment with cyclophosphamide. We conclude that novel therapies for CLL treatment have an effect not only in CLL cells but also in non-malignant cell components of the TME. In this animal model of CLL, the rapid expansion of CLL cells in PB and secondary lymphoid organs provokes loss of MDSC, particularly the Gr-MDSC subpopulation is affected. Treatment with BTKi and CAR T cells provokes clearance of CLL cells in PB and spleen allowing MDSC recovery; suggesting this may be BTK and ITK independent. We continue to explore secondary lymphoid organs to further characterize the shift of the CLL microenvironment from an immunosuppressive to an immune effective one and its impact on immune function in this model. Disclosures Sanderson: Kite/Gilead: Honoraria. Gribben:Celgene: Consultancy, Honoraria, Research Funding; Janssen: Consultancy, Honoraria, Research Funding; Abbvie: Consultancy, Honoraria, Research Funding; Acerta/Astra Zeneca: Consultancy, Honoraria, Research Funding.


Sign in / Sign up

Export Citation Format

Share Document