scholarly journals Immunosuppressive Myeloid Cells Induce Nitric Oxide–Dependent DNA Damage and p53 Pathway Activation in CD8+ T Cells

Author(s):  
Adam N.R. Cartwright ◽  
Shengbao Suo ◽  
Soumya Badrinath ◽  
Sushil Kumar ◽  
Johannes Melms ◽  
...  
2021 ◽  
Vol 9 (Suppl 3) ◽  
pp. A802-A802
Author(s):  
Donghwan Jeon ◽  
Douglas McNeel

BackgroundT-cell checkpoint receptors are expressed when T-cell are activated, and activation of these receptors can impair the function of T-cells and their anti-tumor efficacy.1 We previously found that T-cells activated with cognate antigen increase the expression of PD-1, while this can be attenuated by the presence of specific Toll-like receptor (TLR) agonists.2 3 This effect was mediated by IL-12 secretion from professional antigen presenting cells and resulted in CD8+ T cells with greater anti-tumor activity. In the current report, we sought to determine whether combination of TLR agonists can further affect the expression of T-cell checkpoint receptors and improve T-cell anti-tumor immunity.MethodsOT-1 CD8+ T cells were stimulated with peptide (SIINFEKL) and dendritic cells (DC) in the presence of two different TLR agonists. The cells were collected and evaluated for the expression of T-cell checkpoint receptors (PD-1, CTLA-4, CD160, CD244, LAG-3, TIM-3, TIGIT and VISTA) by flow cytometry, and for transcriptional changes by RNA-seq. Purified DC were stimulated with TLR combinations and evaluated for cytokine release by ELISA. The anti-tumor efficacy of vaccination using peptide and TLR agonist combinations was evaluated in EG7-OVA tumor-bearing mice.ResultsActivation of CD8+ T cells in the presence of specific TLR ligands resulted in decreases in expression of PD-1 and/or CD160. These changes in T-cell checkpoint receptor expression were modestly affected when TLR ligands were used in combination, and notably with combinations of TLR1/2, TLR3, and TLR9 agonists. Immunization of tumor-bearing mice, co-administered with combinations of these agonists, showed greater anti-tumor effects. However, while the effect of TLR1/2 and/or TLR9 was abrogated in IL12KO mice, TLR3 demonstrated anti-tumor activity when co-administered with peptide vaccine. RNA sequencing of TLR-conditioned CD8+ T-cells revealed IL-12 pathway activation, and IFNß pathway activation following TLR3 stimulation. Stimulation of DC with TLR3 agonist, alone or in combination with other TLR agonists, resulted in increased IL-12 and IFNß secretion. Co-incubation of OT-1 splenocytes with rIL12 and/or rIFNß during peptide activation led to reduced expression of PD-1, and this could be reversed with antibodies blocking IL12R or IFNAR-1.ConclusionsMultiple TLR agonists can modulate the expression of T-cell checkpoint receptors, notably PD-1, by upregulating the secretion of IL-12 and IFNß. These data provide the mechanistic rationale for choosing optimal combinations of TLR ligands to use as adjuvants to improve the efficacy of anti-tumor vaccines.ReferencesJin H-T, et al. Cooperation of Tim-3 and PD-1 in CD8 T-cell exhaustion during chronic viral infection. Proceedings of the National Academy of Sciences 2010;107(33):14733–14738.Zahm CD, Colluru VT, McNeel DG. Vaccination with high-affinity epitopes impairs antitumor efficacy by increasing PD-1 expression on CD8+ T cells. Cancer Immunology Research 2017;5(8):630–641.Zahm CD, et al. TLR stimulation during T-cell activation lowers PD-1 expression on CD8+ T Cells. Cancer Immunology Research 2018;6(11):1364–1374.


1994 ◽  
Vol 24 (3) ◽  
pp. 746-752 ◽  
Author(s):  
Mariane M. A. Stefani ◽  
Ingrid Müller ◽  
Jacques A. Louis

PLoS ONE ◽  
2018 ◽  
Vol 13 (1) ◽  
pp. e0191188 ◽  
Author(s):  
David J. Barakat ◽  
Rahul Suresh ◽  
Theresa Barberi ◽  
Kenneth J. Pienta ◽  
Brian W. Simons ◽  
...  

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 3283-3283
Author(s):  
Ga Hye Lee ◽  
Kyung Taek Hong ◽  
Jung Yoon Choi ◽  
Hee Young Shin ◽  
Won-Woo Lee ◽  
...  

Introduction: Pediatric and adolescent patients in need of allogeneic hematopoietic stem cell transplantation generally receive stem cells from older, unrelated or parental donors when a sibling donor is not available. Despite encouraging clinical outcomes, it has been suggested that immune reconstitution accompanied by increased replicative stress and a large difference between donor and recipient age may worsen immunosenescence in pediatric recipients. Therefore, in this study paired samples were collected at the same time from donors and recipients of haploidentical hematopoietic stem cell transplantation (HaploSCT). Methods: We conducted flow cytometry-based phenotypic and functional analyses and telomere length measurements of 21 paired T-cell sets from parental donors and children who received T cell-replete HaploSCT with post-transplant cyclophosphamide (PTCy) at Seoul National University Children's Hospital between February 2014 and January 2017. The conditioning regimen was comprised of targeted busulfan (total target area under the curve, 75,000 mg•h/L) with intensive pharmacokinetic monitoring, fludarabine and cyclophosphamide. Results: Fourteen pediatric, adolescent, and young adult patients with malignant disease and seven with nonmalignant disease were included with a median post-transplantation period of 16.9 months (range, 12.4-38.8). Senescent T cells, CD28- or CD57+ subsets of both CD4+ and CD8+ T cells, were significantly expanded in patients compared with parental donors. Further, not only CD4+CD28- T cells, but also CD4+CD28+ T cells showed reduced cytokine production capacity and impaired polyfunctionality compared with parental donors, whereas their TCR mediated proliferation capacity was comparable. Of note, the telomere length in patient T cells was preserved, or even slightly longer, in senescent T cells compared with donor cells. We also found that the patients had a higher level of γ-H2AX-expressing CD28- senescent T cells compared with the donors, which is used as a DNA damage marker. Regression analysis showed that senescent features of CD4+ and CD8+ T cells in patients were influenced by donor age and the frequency of CD28- cells, respectively. Conclusions: Our data suggest that T cells undergo premature immunosenescent changes and exhibit functional defects in pediatric HaploSCT recipients. Further, there is an increased level of DNA damage in patient CD4+ T cells compared to those of parental donors. Therefore, long-term, comprehensive immune monitoring of these patients is necessary. Disclosures No relevant conflicts of interest to declare.


2020 ◽  
Vol 38 (15_suppl) ◽  
pp. e15644-e15644
Author(s):  
Yuhong Xu ◽  
Anjie Zheng ◽  
Shanshan Jin ◽  
Xiaolong Chen

e15644 Background: All-Trans-Retinoid-Acid (ATRA) is a naturally occurring vitamin A metabolite that participates in many biological processes. Beside its highly potent effect of promoting terminal differentiation of acute promyelocytic leukemia blasts into mature granulocytes, there have been many other studies suggesting its activity on the myeloid derived suppressor cells (MDSCs) and tumor specific CD8+ T cells in animal models as well as using clinical samples. But the use of ATRA as an immune-oncological agent in solid tumor therapy has been limited by the very poor solubility of the compound, its fast metabolism, and very limited exposure achieved after oral administration. Methods: We prepared a new dosage form by encapsulating ATRA inside PEGylated liposomes. The liposomes were shown to accumulate inside solid tumor tissues and deliver more ATRA with longer duration. Results: The effect and dose response of the liposomal ATRA on CT26 murine tumor growth were examined, as well as specific molecular signatures concerning tumor infiltrating myeloid cells. Notably, there was significant higher expression of CD86 and lower expression of PD-L1. These myeloid cells had very low inhibitory effect on ex vivo activated T cells, while on the other hand could promote specific antigen presentation to amplify CD8+ T cells. Furthermore, the liposomal ATRA was also shown to synergize with anti-PD-1 treatment to result in more CD8 T cell distribution in the tumor tissues. Conclusions: These data may suggest an exciting opportunity for targeting MDSCs using liposomal ATRA for combination with T cell based therapeutics in cancer immunotherapy.


AIDS ◽  
2018 ◽  
Vol 32 (18) ◽  
pp. 2669-2677 ◽  
Author(s):  
Fatema Z. Chowdhury ◽  
Zhengyu Ouyang ◽  
Maria Buzon ◽  
Bruce D. Walker ◽  
Mathias Lichterfeld ◽  
...  

2013 ◽  
Vol 7 (8) ◽  
pp. e2325 ◽  
Author(s):  
Simone Bernardino ◽  
Adriana Pina ◽  
Maíra Felonato ◽  
Tânia A. Costa ◽  
Eliseu Frank de Araújo ◽  
...  

2008 ◽  
Vol 27 (12) ◽  
pp. 1333-1339 ◽  
Author(s):  
Jonathan C. Choy ◽  
Tai Yi ◽  
Deepak A. Rao ◽  
George Tellides ◽  
Karen Fox-Talbot ◽  
...  

Immunity ◽  
2018 ◽  
Vol 48 (4) ◽  
pp. 773-786.e5 ◽  
Author(s):  
Jing Li ◽  
Younghee Lee ◽  
Yanjian Li ◽  
Yu Jiang ◽  
Huiping Lu ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document