Overcoming high level adenosine-mediated immunosuppression by DZD2269, a potent and selective A2aR antagonist

Yu Bai, Xin Zhang, Jie Zheng, Ziyi Liu, Zhenfan Yang, Xiaolin Zhang, Yu Bai, Xin Zhang, Jie Zheng, Ziyi Liu, Zhenfan Yang, Xiaolin Zhang

Abstract

Background: Adenosine is a potent immunosuppressant whose levels in the tumor microenvironment (TME) are often much higher than those in normal tissues. Binding of adenosine to its receptor A2aR activates a cascade of genes and leads to immunosuppression. In addition, immune checkpoint blockage markedly increases A2aR expression in T cells, which could dampen their anti-tumor response. Several A2aR antagonists are under clinical development, but with limited clinical benefit reported so far. These A2aR antagonists showed much diminished activity at high adenosine levels found in TME, which may explain their clinical underperformance. We report the discovery and early clinical development of DZD2269, a novel A2aR antagonist which can fully block A2aR mediated immunosuppression commonly found in TME. Adenosine stimulates phosphorylation of cyclic AMP response element binding protein (CREB) in T cells and inhibits anti-tumor cytokine secretion in PBMCs in a dose-dependent manner. DZD2269 was able to reverse the immunosuppression induced by high concentrations of adenosine, as demonstrated by inhibiting CREB phosphorylation in T cells, restoring Th1 cytokine secretion in PBMCs, and stimulating dendritic cells (DCs) maturation. As a single agent, DZD2269 showed anti-tumor growth in multiple syngeneic mouse tumor models, and more profound anti-tumor effects were observed when DZD2269 was in combination with immune checkpoint inhibitors, radiotherapy, or chemotherapy. A good PK/PD relationship was observed in these animal models. In the phase 1 clinical study, downregulation of pCREB was detected in human T cells, consistent with preclinical prediction. Our data support further clinical development of DZD2269 in patients with cancer.

Methods: The selectivity of DZD2269 for adenosine receptors was tested in engineered cell lines, and its efficacy in blocking A2aR signaling and reversing adenosine-mediated immunosuppression was assessed in human T cells and peripheral blood mononuclear cells (PBMCs). The anti-tumor effects of DZD2269 were evaluated in multiple syngeneic mouse models as a single agent as well as in combination with chemotherapy, radiotherapy, or immune checkpoint inhibitors. A phase 1 study in healthy volunteers (NCT04932005) has been initiated to assess safety, pharmacokinetics (PK) and pharmacodynamics (PD) of DZD2269.

Results: Adenosine stimulates phosphorylation of cyclic AMP response element binding protein (CREB) in T cells and inhibits anti-tumor cytokine secretion in PBMCs in a dose-dependent manner. DZD2269 was able to reverse the immunosuppression induced by high concentrations of adenosine, as demonstrated by inhibiting CREB phosphorylation in T cells, restoring Th1 cytokine secretion in PBMCs, and stimulating dendritic cells (DCs) maturation. As a single agent, DZD2269 showed anti-tumor growth in multiple syngeneic mouse tumor models, and more profound anti-tumor effects were observed when DZD2269 was in combination with immune checkpoint inhibitors, radiotherapy, or chemotherapy. A good PK/PD relationship was observed in these animal models. In the phase 1 clinical study, downregulation of pCREB was detected in human T cells, consistent with preclinical prediction.

Conclusion: DZD2269 is a novel A2aR antagonist which can fully block A2aR mediated immunosuppression commonly found in TME. Clinical development of DZD2269 in patients with cancer is warranted (NCT04634344).

Keywords: A2aR; Adenosine; DZD2269; Immunosuppression.

Conflict of interest statement

DZD2269 is an investigational drug of Dizal Pharmaceuticals, and currently in clinical evaluation. All authors are or have been employees and shareholders of Dizal Pharmaceuticals.

© 2022. The Author(s).

Figures

Fig. 1
Fig. 1
Ex vivo modulation of pCREB and IFN-γ in response to NECA. (A) NECA regulates CREB phosphorylation on CD3 + T cells in whole blood samples. Data from test samples from three different donors and plotted using GraphPad Prism. (B) NECA affects IFN-γ secretion in human PBMCs activated by anti-CD3/CD28 antibodies. The experiment was repeated at least 3 times, and the data shown here are from one representative experiment
Fig. 2
Fig. 2
In vitro selectivity of DZD2269 against adenosine receptors and its role in relieving NECA-induced immunosuppression. (A) Effects of DZD2269 on 10 µM NECA-induced cAMP accumulation in CHO-A2aR and CHO-A2bR cells and calcium increase in CHO-A1R and CHO-A3R cells. (B) The ability of A2aR antagonists to block CREB phosphorylation in CD4 + T cells was measured by flow cytometry after stimulation of whole blood with 10 µM NECA. (C-E) DZD2269 relieved the inhibition of 10 µM NECA on the secretion of IL-2 (C), IFN-γ (D) and TNF-α (E) by activated PBMCs. (F) DZD2269 relieved the inhibition of DC maturation by 10 µM NECA. Each data point represents the mean of at least two technical replicates, and error bars represent SD. Each experiment was repeated at least three times, and the data shown here are from one representative experiment
Fig. 3
Fig. 3
Anti-tumor efficacy of DZD2269 in animal models in the presence or absence of T cells (A) B16F10 cells were inoculated in the back of C57BL/6 mice, and the mice were treated with DZD2269. Statistically significant difference was calculated by using two-way ANOVA (**** P:

Fig. 4

DZD2269 enhances the efficacy of…

Fig. 4

DZD2269 enhances the efficacy of radiotherapy (RT) in syngeneic mouse in vivo models.…

Fig. 4
DZD2269 enhances the efficacy of radiotherapy (RT) in syngeneic mouse in vivo models. (A-B) RM-1 tumor cells were irradiated with 20 Gy radiation and were co-cultured anti-CD3/CD28 antibodies activated mouse splenocytes. (A) IFN-γ in supernatant was measured 72 h after co-culture. (B) IL-2 in supernatant was measured 24 h after co-culture. Significance Statistically significant difference was calculated by using one way ANOVA (*: p 

Fig. 5

DZD2269 synergizes with anti-PD-1 antibody…

Fig. 5

DZD2269 synergizes with anti-PD-1 antibody or oxaliplatin in syngeneic mouse in vivo model.…

Fig. 5
DZD2269 synergizes with anti-PD-1 antibody or oxaliplatin in syngeneic mouse in vivo model. (A) Prostate RM-1 tumor-bearing mice were treated with 10 mg/kg anti-PD-1 antibody, with or without DZD2269 (3 mg/kg). (B) Pancreatic Pan02 tumor-bearing mice were treated with 10 mg/kg oxaliplatin with or without DZD2269 (3 mg/kg). bid: twice daily, qw: once a week. tiw: three times a week. Statistically significant difference was calculated by using two-way ANOVA (**: p 

Fig. 6

Correlation of DZD2269 plasma concentration…

Fig. 6

Correlation of DZD2269 plasma concentration (PK) and pCREB inhibition (PD) on CD4 +…

Fig. 6
Correlation of DZD2269 plasma concentration (PK) and pCREB inhibition (PD) on CD4 + T Cell. Whole blood samples were collected at pre-dose, 2 h, and 24 h after single dose of DZD2269, samples were stimulated with or without 10 µM NECA. pCREB inhibition was calculated for each PD time point and plotted against PK at the corresponding time point. pCREB data from placebo-treated subjects (red dot) were also included in the analysis to enable assessment of biological and technical assay variation in PD assay. The dashed black line indicates the 90% pCREB inhibition
Similar articles
References
    1. Vaupel P, Mayer A. Hypoxia-Driven Adenosine Accumulation: A Crucial Microenvironmental Factor Promoting Tumor Progression. Adv Exp Med Biol. 2016;876:177–83. doi: 10.1007/978-1-4939-3023-4_22. - DOI - PubMed
    1. Di Virgilio F, Sarti AC, Coutinho-Silva R. Purinergic signaling, DAMPs, and inflammation. Am J Physiol Cell Physiol. 2020;318(5):C832-C5. doi: 10.1152/ajpcell.00053.2020. - DOI - PubMed
    1. Alvarez CL, Troncoso MF, Espelt MV. Extracellular ATP and adenosine in tumor microenvironment: Roles in epithelial-mesenchymal transition, cell migration, and invasion. J Cell Physiol. 2022;237(1):389–400. doi: 10.1002/jcp.30580. - DOI - PubMed
    1. Losenkova K, Zuccarini M, Karikoski M, Laurila J, Boison D, Jalkanen S, et al. Compartmentalization of adenosine metabolism in cancer cells and its modulation during acute hypoxia. J Cell Sci. 2020. - PubMed
    1. Martins I, Tesniere A, Kepp O, Michaud M, Schlemmer F, Senovilla L, et al. Chemotherapy induces ATP release from tumor cells. Cell Cycle. 2009;8(22):3723–8. doi: 10.4161/cc.8.22.10026. - DOI - PubMed
Show all 47 references
MeSH terms
Associated data
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Follow NCBI
Fig. 4
Fig. 4
DZD2269 enhances the efficacy of radiotherapy (RT) in syngeneic mouse in vivo models. (A-B) RM-1 tumor cells were irradiated with 20 Gy radiation and were co-cultured anti-CD3/CD28 antibodies activated mouse splenocytes. (A) IFN-γ in supernatant was measured 72 h after co-culture. (B) IL-2 in supernatant was measured 24 h after co-culture. Significance Statistically significant difference was calculated by using one way ANOVA (*: p 

Fig. 5

DZD2269 synergizes with anti-PD-1 antibody…

Fig. 5

DZD2269 synergizes with anti-PD-1 antibody or oxaliplatin in syngeneic mouse in vivo model.…

Fig. 5
DZD2269 synergizes with anti-PD-1 antibody or oxaliplatin in syngeneic mouse in vivo model. (A) Prostate RM-1 tumor-bearing mice were treated with 10 mg/kg anti-PD-1 antibody, with or without DZD2269 (3 mg/kg). (B) Pancreatic Pan02 tumor-bearing mice were treated with 10 mg/kg oxaliplatin with or without DZD2269 (3 mg/kg). bid: twice daily, qw: once a week. tiw: three times a week. Statistically significant difference was calculated by using two-way ANOVA (**: p 

Fig. 6

Correlation of DZD2269 plasma concentration…

Fig. 6

Correlation of DZD2269 plasma concentration (PK) and pCREB inhibition (PD) on CD4 +…

Fig. 6
Correlation of DZD2269 plasma concentration (PK) and pCREB inhibition (PD) on CD4 + T Cell. Whole blood samples were collected at pre-dose, 2 h, and 24 h after single dose of DZD2269, samples were stimulated with or without 10 µM NECA. pCREB inhibition was calculated for each PD time point and plotted against PK at the corresponding time point. pCREB data from placebo-treated subjects (red dot) were also included in the analysis to enable assessment of biological and technical assay variation in PD assay. The dashed black line indicates the 90% pCREB inhibition
Similar articles
References
    1. Vaupel P, Mayer A. Hypoxia-Driven Adenosine Accumulation: A Crucial Microenvironmental Factor Promoting Tumor Progression. Adv Exp Med Biol. 2016;876:177–83. doi: 10.1007/978-1-4939-3023-4_22. - DOI - PubMed
    1. Di Virgilio F, Sarti AC, Coutinho-Silva R. Purinergic signaling, DAMPs, and inflammation. Am J Physiol Cell Physiol. 2020;318(5):C832-C5. doi: 10.1152/ajpcell.00053.2020. - DOI - PubMed
    1. Alvarez CL, Troncoso MF, Espelt MV. Extracellular ATP and adenosine in tumor microenvironment: Roles in epithelial-mesenchymal transition, cell migration, and invasion. J Cell Physiol. 2022;237(1):389–400. doi: 10.1002/jcp.30580. - DOI - PubMed
    1. Losenkova K, Zuccarini M, Karikoski M, Laurila J, Boison D, Jalkanen S, et al. Compartmentalization of adenosine metabolism in cancer cells and its modulation during acute hypoxia. J Cell Sci. 2020. - PubMed
    1. Martins I, Tesniere A, Kepp O, Michaud M, Schlemmer F, Senovilla L, et al. Chemotherapy induces ATP release from tumor cells. Cell Cycle. 2009;8(22):3723–8. doi: 10.4161/cc.8.22.10026. - DOI - PubMed
Show all 47 references
MeSH terms
Associated data
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Fig. 5
Fig. 5
DZD2269 synergizes with anti-PD-1 antibody or oxaliplatin in syngeneic mouse in vivo model. (A) Prostate RM-1 tumor-bearing mice were treated with 10 mg/kg anti-PD-1 antibody, with or without DZD2269 (3 mg/kg). (B) Pancreatic Pan02 tumor-bearing mice were treated with 10 mg/kg oxaliplatin with or without DZD2269 (3 mg/kg). bid: twice daily, qw: once a week. tiw: three times a week. Statistically significant difference was calculated by using two-way ANOVA (**: p 

Fig. 6

Correlation of DZD2269 plasma concentration…

Fig. 6

Correlation of DZD2269 plasma concentration (PK) and pCREB inhibition (PD) on CD4 +…

Fig. 6
Correlation of DZD2269 plasma concentration (PK) and pCREB inhibition (PD) on CD4 + T Cell. Whole blood samples were collected at pre-dose, 2 h, and 24 h after single dose of DZD2269, samples were stimulated with or without 10 µM NECA. pCREB inhibition was calculated for each PD time point and plotted against PK at the corresponding time point. pCREB data from placebo-treated subjects (red dot) were also included in the analysis to enable assessment of biological and technical assay variation in PD assay. The dashed black line indicates the 90% pCREB inhibition
Fig. 6
Fig. 6
Correlation of DZD2269 plasma concentration (PK) and pCREB inhibition (PD) on CD4 + T Cell. Whole blood samples were collected at pre-dose, 2 h, and 24 h after single dose of DZD2269, samples were stimulated with or without 10 µM NECA. pCREB inhibition was calculated for each PD time point and plotted against PK at the corresponding time point. pCREB data from placebo-treated subjects (red dot) were also included in the analysis to enable assessment of biological and technical assay variation in PD assay. The dashed black line indicates the 90% pCREB inhibition

References

    1. Vaupel P, Mayer A. Hypoxia-Driven Adenosine Accumulation: A Crucial Microenvironmental Factor Promoting Tumor Progression. Adv Exp Med Biol. 2016;876:177–83. doi: 10.1007/978-1-4939-3023-4_22.
    1. Di Virgilio F, Sarti AC, Coutinho-Silva R. Purinergic signaling, DAMPs, and inflammation. Am J Physiol Cell Physiol. 2020;318(5):C832-C5. doi: 10.1152/ajpcell.00053.2020.
    1. Alvarez CL, Troncoso MF, Espelt MV. Extracellular ATP and adenosine in tumor microenvironment: Roles in epithelial-mesenchymal transition, cell migration, and invasion. J Cell Physiol. 2022;237(1):389–400. doi: 10.1002/jcp.30580.
    1. Losenkova K, Zuccarini M, Karikoski M, Laurila J, Boison D, Jalkanen S, et al. Compartmentalization of adenosine metabolism in cancer cells and its modulation during acute hypoxia. J Cell Sci. 2020.
    1. Martins I, Tesniere A, Kepp O, Michaud M, Schlemmer F, Senovilla L, et al. Chemotherapy induces ATP release from tumor cells. Cell Cycle. 2009;8(22):3723–8. doi: 10.4161/cc.8.22.10026.
    1. Feng LL, Cai YQ, Zhu MC, Xing LJ, Wang X. The yin and yang functions of extracellular ATP and adenosine in tumor immunity. Cancer Cell Int. 2020;20:110. doi: 10.1186/s12935-020-01195-x.
    1. Dubyak GR. Luciferase-assisted detection of extracellular ATP and ATP metabolites during immunogenic death of cancer cells. Methods Enzymol. 2019;629:81–102. doi: 10.1016/bs.mie.2019.10.006.
    1. Shevchenko I, Mathes A, Groth C, Karakhanova S, Muller V, Utikal J, et al. Enhanced expression of CD39 and CD73 on T cells in the regulation of anti-tumor immune responses. Oncoimmunology. 2020;9(1):1744946. doi: 10.1080/2162402X.2020.1744946.
    1. Baghbani E, Noorolyai S, Shanehbandi D, Mokhtarzadeh A, Aghebati-Maleki L, Shahgoli VK, et al. Regulation of immune responses through CD39 and CD73 in cancer: Novel checkpoints. Life Sci. 2021;282:119826. doi: 10.1016/j.lfs.2021.119826.
    1. Horenstein AL, Chillemi A, Zaccarello G, Bruzzone S, Quarona V, Zito A, et al. A CD38/CD203a/CD73 ectoenzymatic pathway independent of CD39 drives a novel adenosinergic loop in human T lymphocytes. Oncoimmunology. 2013;2(9):e26246. doi: 10.4161/onci.26246.
    1. Street SE, Kramer NJ, Walsh PL, Taylor-Blake B, Yadav MC, King IF, et al. Tissue-nonspecific alkaline phosphatase acts redundantly with PAP and NT5E to generate adenosine in the dorsal spinal cord. J Neurosci. 2013;33(27):11314–22. doi: 10.1523/JNEUROSCI.0133-13.2013.
    1. Morello S, Pinto A, Blandizzi C, Antonioli L. Myeloid cells in the tumor microenvironment: Role of adenosine. Oncoimmunology. 2016;5(3):e1108515. doi: 10.1080/2162402X.2015.1108515.
    1. Cekic C, Day YJ, Sag D, Linden J. Myeloid expression of adenosine A2A receptor suppresses T and NK cell responses in the solid tumor microenvironment. Cancer Res. 2014;74(24):7250–9. doi: 10.1158/0008-5472.CAN-13-3583.
    1. Kumar V. Adenosine as an endogenous immunoregulator in cancer pathogenesis: where to go? Purinergic Signal. 2013;9(2):145–65. doi: 10.1007/s11302-012-9349-9.
    1. Sun WC, Moore JN, Hurley DJ, Vandenplas ML, Murray TF. Effects of stimulation of adenosine A2A receptors on lipopolysaccharide-induced production of reactive oxygen species by equine neutrophils. Am J Vet Res. 2007;68(6):649–56. doi: 10.2460/ajvr.68.6.649.
    1. Stamatos NM, Carubelli I, van de Vlekkert D, Bonten EJ, Papini N, Feng C, et al. LPS-induced cytokine production in human dendritic cells is regulated by sialidase activity. J Leukoc Biol. 2010;88(6):1227–39. doi: 10.1189/jlb.1209776.
    1. Ma SR, Deng WW, Liu JF, Mao L, Yu GT, Bu LL, et al. Blockade of adenosine A2A receptor enhances CD8(+) T cells response and decreases regulatory T cells in head and neck squamous cell carcinoma. Mol Cancer. 2017;16(1):99. doi: 10.1186/s12943-017-0665-0.
    1. Sheth S, Brito R, Mukherjea D, Rybak LP, Ramkumar V. Adenosine receptors: expression, function and regulation. Int J Mol Sci. 2014;15(2):2024–52. doi: 10.3390/ijms15022024.
    1. Liu C, Shang Q, Bai Y, Guo C, Zhu F, Zhang L, et al. Adenosine A2A receptor, a potential valuable target for controlling reoxygenated DCs-triggered inflammation. Mol Immunol. 2015;63(2):559–65. doi: 10.1016/j.molimm.2014.10.012.
    1. Leone RD, Lo YC, Powell JD. A2aR antagonists: Next generation checkpoint blockade for cancer immunotherapy. Comput Struct Biotechnol J. 2015;13:265–72. doi: 10.1016/j.csbj.2015.03.008.
    1. Sun C, Wang B, Hao S. Adenosine-A2A Receptor Pathway in Cancer Immunotherapy. Front Immunol. 2022;13:837230. doi: 10.3389/fimmu.2022.837230.
    1. Beavis PA, Milenkovski N, Henderson MA, John LB, Allard B, Loi S, et al. Adenosine Receptor 2A Blockade Increases the Efficacy of Anti-PD-1 through Enhanced Antitumor T-cell Responses. Cancer Immunol Res. 2015;3(5):506–17. doi: 10.1158/2326-6066.CIR-14-0211.
    1. Fong L, Hotson A, Powderly JD, Sznol M, Heist RS, Choueiri TK, et al. Adenosine 2A Receptor Blockade as an Immunotherapy for Treatment-Refractory Renal Cell Cancer. Cancer Discov. 2020;10(1):40–53. doi: 10.1158/-19-0980.
    1. Mixed, Reviews for A2AR Inhibitor in NSCLC. Cancer Discov. 2019;9(1):OF2.
    1. Chiappori AA, Creelan B, Tanvetyanon T, Gray JE, Haura EB, Thapa R, et al. Phase I Study of Taminadenant (PBF509/NIR178), an Adenosine 2A Receptor Antagonist, with or without Spartalizumab (PDR001), in Patients with Advanced Non-Small Cell Lung Cancer. Clin Cancer Res. 2022;28(11):2313–20. doi: 10.1158/1078-0432.CCR-21-2742.
    1. Lim EA, Bendell JC, Falchook GS, Bauer TM, Drake CG, Choe JH, et al. Phase 1a/b, Open-label, Multicenter Study of AZD4635 (an Adenosine 2A Receptor Antagonist) as Monotherapy or Combined with Durvalumab, in Patients with Solid Tumors. Clin Cancer Res. 2022.
    1. Borodovsky A, Wang Y, Ye M, Shaw JC, Sachsenmeier K, Deng N, et al. Abstract 3751: Inhibition of A2AR by AZD4635 induces anti-tumor immunity alone and in combination with anti-PD-L1 in preclinical models. Cancer Res. 2018;78(13_Supplement):3751. doi: 10.1158/1538-7445.AM2018-3751.
    1. Willingham SB, Ho PY, Hotson A, Hill C, Piccione EC, Hsieh J, et al. A2AR Antagonism with CPI-444 Induces Antitumor Responses and Augments Efficacy to Anti-PD-(L)1 and Anti-CTLA-4 in Preclinical Models. Cancer Immunol Res. 2018;6(10):1136–49. doi: 10.1158/2326-6066.CIR-18-0056.
    1. Nunez F, Taura J, Camacho J, Lopez-Cano M, Fernandez-Duenas V, Castro N, et al. PBF509, an Adenosine A2A Receptor Antagonist With Efficacy in Rodent Models of Movement Disorders. Front Pharmacol. 2018;9:1200. doi: 10.3389/fphar.2018.01200.
    1. Huang J, Zhang D, Bai Y, Yang P, Xing L, Yu J. A2AR Antagonism with DZD2269 Augments Antitumor Efficacy of Irradiation in Murine Model. J Cancer. 2020;11(12):3685–92. doi: 10.7150/jca.43966.
    1. de Lera Ruiz M, Lim YH, Zheng J. Adenosine A2A receptor as a drug discovery target. J Med Chem. 2014;57(9):3623–50. doi: 10.1021/jm4011669.
    1. Jung U, Foley JE, Erdmann AA, Eckhaus MA, Fowler DH. CD3/CD28-costimulated T1 and T2 subsets: differential in vivo allosensitization generates distinct GVT and GVHD effects. Blood. 2003;102(9):3439–46. doi: 10.1182/blood-2002-12-3936.
    1. Pathology, Department, Animal and Plant Health Agency, Fixation, tissue processing, histology and immunohistochemistry procedures for diagnosis of animal TSE (BSE, Scrapie, Atypical Scrapie) 2016 [Available from: .
    1. Layland J, Carrick D, Lee M, Oldroyd K, Berry C. Adenosine: physiology, pharmacology, and clinical applications. JACC Cardiovasc Interv. 2014;7(6):581–91. doi: 10.1016/j.jcin.2014.02.009.
    1. Matthews GD, Grace AA. Unmasking Adenosine: The Purinergic Signalling Molecule Critical to Arrhythmia Pathophysiology and Management. Arrhythm Electrophysiol Rev. 2020;8(4):240–8. doi: 10.15420/aer.2019.05.
    1. Borodovsky A, Barbon CM, Wang Y, Ye M, Prickett L, Chandra D, et al. Small molecule AZD4635 inhibitor of A2AR signaling rescues immune cell function including CD103(+) dendritic cells enhancing anti-tumor immunity. J Immunother Cancer. 2020;8(2).
    1. Haddad M. Impact of Adenosine Analogue, Adenosine-5’-N-Ethyluronamide (NECA), on Insulin Signaling in Skeletal Muscle Cells. Biomed Res Int. 2021;2021:9979768. doi: 10.1155/2021/9979768.
    1. Gessi S, Merighi S, Varani K, Leung E, Mac Lennan S, Borea PA. The A3 adenosine receptor: an enigmatic player in cell biology. Pharmacol Ther. 2008;117(1):123–40. doi: 10.1016/j.pharmthera.2007.09.002.
    1. Fenton RA, Shea LG, Doddi C, Dobson JG. Jr. Myocardial adenosine A(1)-receptor-mediated adenoprotection involves phospholipase C, PKC-epsilon, and p38 MAPK, but not HSP27. Am J Physiol Heart Circ Physiol. 2010;298(6):H1671-8. doi: 10.1152/ajpheart.01028.2009.
    1. Prechtel AT, Steinkasserer A. CD83: an update on functions and prospects of the maturation marker of dendritic cells. Arch Dermatol Res. 2007;299(2):59–69. doi: 10.1007/s00403-007-0743-z.
    1. Li Z, Ju X, Silveira PA, Abadir E, Hsu WH, Hart DNJ, et al. CD83: Activation Marker for Antigen Presenting Cells and Its Therapeutic Potential. Front Immunol. 2019;10:1312. doi: 10.3389/fimmu.2019.01312.
    1. DiRenzo D, Piovesan D, Tan J, Miles DH, Leleti MR, Park T, et al. Abstract A162: AB928, a dual antagonist of the A2aR and A2bR adenosine receptors, relieves adenosine-mediated immune suppression. Cancer Immunol Res. 2019;7(2_Supplement):A162-A. doi: 10.1158/2326-6074.CRICIMTEATIAACR18-A162.
    1. Vaupel P, Multhoff G. Accomplices of the Hypoxic Tumor Microenvironment Compromising Antitumor Immunity: Adenosine, Lactate, Acidosis, Vascular Endothelial Growth Factor, Potassium Ions, and Phosphatidylserine. Front Immunol. 2017;8:1887. doi: 10.3389/fimmu.2017.01887.
    1. Eriksson D, Stigbrand T. Radiation-induced cell death mechanisms. Tumour Biol. 2010;31(4):363–72. doi: 10.1007/s13277-010-0042-8.
    1. Zhu M, Yang M, Zhang J, Yin Y, Fan X, Zhang Y, et al. Immunogenic Cell Death Induction by Ionizing Radiation Front Immunol. 2021;12:705361.
    1. Gallo PM, Gallucci S. The dendritic cell response to classic, emerging, and homeostatic danger signals. Implications for autoimmunity. Front Immunol. 2013;4:138. doi: 10.3389/fimmu.2013.00138.
    1. Gocher AM, Workman CJ, Vignali DAA. Interferon-gamma: teammate or opponent in the tumour microenvironment? Nat Rev Immunol. 2022;22(3):158–72. doi: 10.1038/s41577-021-00566-3.

Source: PubMed

3
订阅