scholarly journals Soluble MUC1 secreted by human epithelial cancer cells mediates immune suppression by blocking T-cell activation

Author(s):  
Allen K. Chan ◽  
Diane C. Lockhart ◽  
Wolfram von Bernstorff ◽  
Remco A. Spanjaard ◽  
Hong-Gu Joo ◽  
...  
2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A744-A744
Author(s):  
Tingting Zhong ◽  
Zhaoliang Huang ◽  
Xinghua Pang ◽  
Na Chen ◽  
Xiaoping Jin ◽  
...  

BackgroundCD73 (ecto-5’-nucleotidase) is an ecto-nucleotidase that dephosphorylate AMP to form adenosine. Activation of adenosine signaling pathway in immune cells leads to the suppression of effector functions, down-regulate macrophage phagocytosis, inhibit pro-inflammatory cytokine release, as well as yield aberrantly differentiated dendritic cells producing pro-tumorigenic molecules.1 In the tumor microenvironment, adenosinergic negative feedback signaling facilitated immune suppression is considered an important mechanism for immune evasion of cancer cells.2 3 Combination of CD73 and anti-PD-1 antibody has shown promising activity in suppressing tumor growth. Hence, we developed AK119, an anti- human CD73 monoclonal antibody, and AK123,a bi-specific antibody targeting both PD-1 and CD73 for immune therapy of cancer.MethodsAK119 is a humanized antibody against CD73 and AK123 is a tetrameric bi-specific antibody targeting PD-1 and CD73. Binding assays of AK119 and AK123 to antigens, and antigen expressing cells were performed by using ELISA, Fortebio, and FACS assays. In-vitro assays to investigate the activity of AK119 and AK123 to inhibit CD73 enzymatic activity in modified CellTiter-Glo assay, to induce endocytosis of CD73, and to activate B cells were performed. Assay to evaluate AK123 activity on T cell activation were additionally performed. Moreover, the activities of AK119 and AK123 to mediate ADCC, CDC in CD73 expressing cells were also evaluated.ResultsAK119 and AK123 could bind to its respective soluble or membrane antigens expressing on PBMCs, MDA-MB-231, and U87-MG cells with high affinity. Results from cell-based assays indicated that AK119 and AK123 effectively inhibited nucleotidase enzyme activity of CD73, mediated endocytosis of CD73, and induced B cell activation by upregulating CD69 and CD83 expression on B cells, and showed more robust CD73 blocking and B cell activation activities compared to leading clinical candidate targeting CD73. AK123 could also block PD-1/PD-L1 interaction and enhance T cell activation.ConclusionsIn summary, AK119 and AK123 represent good preclinical biological properties, which support its further development as an anti-cancer immunotherapy or treating other diseases.ReferencesDeaglio S, Dwyer KM, Gao W, Friedman D, Usheva A, Erat A, Chen JF, Enjyoji K, Linden J, Oukka M, et al. Adenosine generation catalyzed by CD39 and CD73 expressed on regulatory T cells mediates immune suppression. J Exp Med 2007; 204:1257–65.Huang S, Apasov S, Koshiba M, Sitkovsky M. Role of A2a extracellular adenosine receptor-mediated signaling in adenosine-mediated inhibition of T-cell activation and expansion. Blood. 1997; 90:1600–10.Novitskiy SV, Ryzhov S, Zaynagetdinov R, Goldstein AE, Huang Y, Tikhomirov OY, Blackburn MR, Biaggioni I,Carbone DP, Feoktistov I, et al. Adenosine receptors in regulation of dendritic cell differentiation and function. Blood 2008; 112:1822–31.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. SCI-46-SCI-46
Author(s):  
Melody A. Swartz

Tumor engagement or activation of surrounding lymphatic vessels is well-known to correlate with tumor progression and metastasis in melanoma and many other cancers. We and others have identified several mechanisms by which the lymphatic growth factor VEGF-C and lymphangiogenesis can promote metastasis, including (i) increasing immune suppressive cell types and factors in the tumor microenvironment both directly and indirectly, (ii) inhibiting maturation of antigen-presenting cells and T cell activation, and (iii) driving changes in the stromal microenvironment that promote both cancer invasion and immune suppression. However, lymphatic activation also enhances communication with cells in the draining lymph node by antigen and cell transport, which may trigger the initiation of adaptive immune responses against the tumor. Under normal conditions, the potential anti-tumor effects are rendered 'dormant' by the pro-tumor immune suppression, and the tumor progresses. However, we are now observing that lymphangiogenic tumors are exceptionally responsive to immunotherapy, implying that the anti-tumor aspects can be unleashed when the overall balance of pro- and anti-tumor immune aspects is tipped enough towards the latter (e.g., upon tumor cell killing). On the mechanistic side, we are finding that 'lymphangiogenic potentiation' depends on tumor cell infiltration of both CD103+ dendritic cells and naïve T cells, driving local T cell education post-immunotherapy and antigen spreading. On the translational side, we are developing novel strategies to exploit lymphangiogenesis for cancer immunotherapy. Understanding the yin and yang of lymphatic activation in the tumor microenvironment and how it affects immunity may lead to exciting new translational strategies for cancer immunotherapy. Disclosures No relevant conflicts of interest to declare.


Cancers ◽  
2019 ◽  
Vol 11 (11) ◽  
pp. 1797 ◽  
Author(s):  
Sarah Di Somma ◽  
Jussara Amato ◽  
Nunzia Iaccarino ◽  
Bruno Pagano ◽  
Antonio Randazzo ◽  
...  

Background: DNA G-quadruplex (G4) structures represent potential anti-cancer targets. In this study, we compared the effect of two G4-targeting compounds, C066-3108 and the gold standard BRACO-19. Methods: In breast and prostate cancer cells, cytotoxicity induced by both molecules was measured by a sulforhodamine B assay. In breast cancer cells, cycle, apoptosis, the formation of G4 structures, calreticulin and high mobility group box 1 (HMGB1), as well as T cell activation, were analyzed by flow cytometry and adenosine triphosphate (ATP) by luminescence. Results: Both ligands inhibited cell survival and induced DNA damage. In MCF-7 cells, G4 ligands increased the subG0/G1 phase of the cell cycle inducing apoptosis and reduced intracellular ATP. In untreated MCF-7 cells, we observed a slight presence of G4 structures associated with the G2/M phase. In MDA-MB231 cells, G4 ligands decreased the G1 and enhanced the G2/M phase. We observed a decrease of intracellular ATP, calreticulin cell surface exposure and an increase of HMGB1, accompanied by T cell activation. Both compounds induced G4 structure formation in the subG0/G1 phase. Conclusions: Our data report similar effects for both compounds and the first evidence that G4 ligands induce the release of danger signals associated with immunogenic cell death and induction of T cell activation.


2009 ◽  
Vol 32 (6) ◽  
pp. 565-573 ◽  
Author(s):  
Patrick Bühler ◽  
Eszter Molnar ◽  
Elaine P. Dopfer ◽  
Philipp Wolf ◽  
Dorothee Gierschner ◽  
...  

2013 ◽  
Vol 191 (5) ◽  
pp. 2829-2836 ◽  
Author(s):  
Samuel T. Haile ◽  
Sonia P. Dalal ◽  
Virginia Clements ◽  
Koji Tamada ◽  
Suzanne Ostrand-Rosenberg

2020 ◽  
Vol 38 (15_suppl) ◽  
pp. TPS9080-TPS9080
Author(s):  
Taofeek Kunle Owonikoko ◽  
Hossein Borghaei ◽  
Stéphane Champiat ◽  
Luis G. Paz-Ares ◽  
Ramaswamy Govindan ◽  
...  

TPS9080 Background: SCLC is an aggressive neuroendocrine tumor with poor prognosis and few treatment options. Delta-like ligand 3 (DLL3) is an inhibitory Notch ligand that is highly expressed on the surface of most SCLC tumors but minimally expressed in normal tissues. As such, DLL3 may be a promising therapeutic target. AMG 757 is an HLE BiTE immune therapy designed to redirect cytotoxic T cells to cancer cells by binding to DLL3 on cancer cells and CD3 on T cells, resulting in T cell activation and expansion and T cell-dependent killing of tumor cells. In addition to its direct antitumor effect, BiTE immune therapy can inflame the tumor microenvironment. Combining AMG 757 with a PD-1 pathway inhibitor may lead to increased antitumor activity by enabling sustained T cell-dependent killing of tumor cells. Methods: NCT03319940 is an open-label, ascending, multiple-dose, phase 1 study evaluating AMG 757 as monotherapy; the protocol was recently amended to also evaluate AMG 757 in combination with pembrolizumab. The study will include a dose exploration (monotherapy and combination) followed by a dose expansion (monotherapy). Key eligibility criteria: adult patients with relapsed/refractory SCLC whose disease progressed or recurred after at least 1 platinum-based chemotherapy regimen, ECOG performance status 0–2, at least 2 measurable lesions per modified RECIST 1.1, no untreated or symptomatic brain metastases, and adequate organ function. Primary objectives are to evaluate safety/tolerability and determine the maximum tolerated dose or recommended phase 2 dose of AMG 757 as monotherapy and in combination with pembrolizumab. Secondary objectives are to characterize pharmacokinetics and evaluate preliminary antitumor activity; exploratory objectives are to assess immunogenicity and changes in biomarkers in blood and tumor tissue. In the dose exploration phase, dose escalation/de-escalation decisions will be guided by a Bayesian logistic regression model; backfill enrollment at dose levels deemed safe and tolerable will be allowed. The study is open and recruiting patients. Clinical trial information: NCT03319940.


2005 ◽  
Vol 54 (11) ◽  
pp. 1137-1142 ◽  
Author(s):  
Pratima Sinha ◽  
Virginia K. Clements ◽  
Seth Miller ◽  
Suzanne Ostrand-Rosenberg

2020 ◽  
Vol 165 ◽  
pp. 112389 ◽  
Author(s):  
Seungman Park ◽  
Yu Shi ◽  
Byoung Choul Kim ◽  
Myung Hyun Jo ◽  
Leilani O. Cruz ◽  
...  

2019 ◽  
Vol 7 (1) ◽  
pp. 45-61
Author(s):  
Pedro J. Llanos ◽  
Kristina Andrijauskaite

AbstractResearch indicates that exposure to microgravity leads to immune system dysregulation. However, there is a lack of clear evidence on the specific reasons and precise mechanisms accounting for these immune system changes. Past studies investigating space travel-induced alterations in immunological parameters report many conflicting results, explained by the role of certain confounders, such as cosmic radiation, individual body environment, or differences in experimental design. To minimize the variability in results and to eliminate some technical challenges, we advocate conducting thorough feasibility studies prior to actual suborbital or orbital space experiments. We show how exposure to suborbital flight stressors and the use of a two-dimensional slow rotating device affect T-cells and cancer cells survivability. To enhance T-cell activation and viability, we primed them alone or in combination with IL-2 and IL-12 cytokines. Viability of T-cells was assessed before, during the experiment, and at the end of the experiment for which T-cells were counted every day for the last 4 days to allow the cells to form clear structures and do not disturb their evolution into various geometries. The slow rotating device could be considered a good system to perform T-cell activation studies and develop cell aggregates for various types of cells that react differently to thermal stressors.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A593-A593
Author(s):  
Remya Raja ◽  
Christopher Wu ◽  
Kristina Butler ◽  
Marion Curtis

BackgroundIncreased immune infiltration in ovarian tumors has been linked to improved patient outcome. Nonetheless, responses to checkpoint blockade therapies have been disappointing in ovarian cancer patients. This has been attributed to the low mutational burden present in ovarian tumors. However, many tumor antigens have been identified in ovarian cancer, which underscores the critical need to identify new treatment strategies that will trigger anti-tumor immunity in ovarian cancer. Recent studies have revealed that defects in DNA damage repair (DDR) pathways can contribute to improved responses to immune-directed therapies.1 2 We previously discovered that CT45 expression sensitizes ovarian cancer cells to chemotherapy via its interaction with the protein phosphatase 4 (PP4) complex.3 PP4 is known to play a key role in DDR pathways; however, its potential effects on anti-tumor immunity remain unknown.MethodsUsing fostriecin, a commercially available inhibitor of PP4, we studied the effect of fostriecin on chemosensitivity using cell cycle analysis and cell viability assays. To study the effect of fostriecin on DNA damage, we performed comet assays and measured micronuclei along with FANCD2 foci formation. Furthermore, using western blot, qPCR, and T cell activation assays, we assessed the role of fostriecin in promoting an inflammatory response. We tested the efficacy of combining fostriecin with carboplatin and PD-1 inhibition in a syngeneic mouse model of ovarian cancer.ResultsOur results show that fostriecin treatment combined with carboplatin leads to increased carboplatin sensitivity, DNA damage, and micronuclei formation. Using a panel of ovarian cancer cells, we show that fostriecin treatment triggers an anti-tumor immune response via STAT1 activation resulting in increased expression of pro-inflammatory cytokines. Furthermore, in a syngeneic mouse ID8 ovarian cancer cell line, we demonstrate that combination treatment of fostriecin and carboplatin significantly increased CD8 T cell activation over carboplatin treatment alone.ConclusionsOur work has identified a role for PP4 inhibition in promoting anti-tumor immunity. These findings form the groundwork for the rationale design of a clinical trial combining PP4 inhibitors with chemo-immunotherapy as a new approach in ovarian cancer treatment.ReferencesRodier F, Coppé J-P, Patil CK, Hoeijmakers WAM, Muñoz DP, Raza SR, et al. Persistent DNA damage signalling triggers senescence-associated inflammatory cytokine secretion. Nature Cell Biology 2009; 11: 973–979.Zhang H, Christensen CL, Dries R, Oser MG, Deng J, Diskin B, et al. CDK7 Inhibition potentiates genome instability triggering anti-tumor immunity in small cell lung cancer. Cancer cell 2020;37:37–54.e39.Coscia F, Lengyel E, Duraiswamy J, Ashcroft B, Bassani-Sternberg M, Wierer M, et al. Multi-level proteomics identifies CT45 as a chemosensitivity mediator and immunotherapy target in ovarian cancer. Cell 2018;175:159–170.e116.


Sign in / Sign up

Export Citation Format

Share Document