Skip to main content

REVIEW article

Front. Immunol., 29 September 2020
Sec. Cytokines and Soluble Mediators in Immunity
This article is part of the Research Topic Purinergic Signaling and Inflammation View all 10 articles

Targeting Hypoxia-A2A Adenosinergic Immunosuppression of Antitumor T Cells During Cancer Immunotherapy

  • Department of Pharmaceutical Sciences, New England Inflammation and Tissue Protection Institute, Bouvé College of Health Sciences, Northeastern University, Boston, MA, United States

The blockade of immunological negative regulators offered a novel therapeutic approach that revolutionized the immunotherapy of cancer. Still, a significant portion of patients fail to respond to anti-PD-1/PD-L1 and/or anti-CTLA-4 therapy or experience significant adverse effects. We propose that one of the major reasons that many patients do not respond to this form of therapy is due to the powerful physiological suppression mediated by hypoxia-adenosinergic signaling. Indeed, both inflamed and cancerous tissues are hypoxic and rich in extracellular adenosine, in part due to stabilization of the transcription factor hypoxia-inducible factor 1 alpha (HIF-1α). Adenosine signals through adenosine A2A receptors (A2AR) to suppress anti-tumor and anti-pathogen immune responses. Several classes of anti-hypoxia-A2AR therapeutics have been offered to refractory cancer patients, with A2AR blockers, inhibitors of adenosine-generating enzymes such as CD39 and CD73, and hypoxia-targeting drugs now reaching the clinical stage. Clinical results have confirmed preclinical observations that blockade of the hypoxia-adenosine-A2AR axis synergizes with inhibitors of immune checkpoints to induce tumor rejection. Thus, A2AR blockers provide a new hope for the majority of patients who are nonresponsive to current immunotherapeutic approaches including checkpoint blockade. Here, we discuss the discoveries that firmly implicate the A2AR as a critical and non-redundant biochemical negative regulator of the immune response and highlight the importance of targeting the hypoxia-adenosine-A2AR axis to manipulate anti-pathogen and anti-tumor immune responses.

Overview of the Hypoxia-Adenosine-A2AR Axis

While hypoxia-dependent generation of extracellular adenosine and subsequent immunosuppressive signaling through adenosine A2A receptors (A2AR) is deleterious in the tumor microenvironment (TME), this mechanism normally has an important tissue-protective function. The suppression of tumor-reactive T cells by hypoxia-adenosine-A2AR signaling in the TME is a commandeering of this evolutionarily conserved, non-redundant feedback mechanism to govern inflammation (13). Sitkovsky and colleagues were the first to confirm in vivo that this may explain the paradoxical peaceful coexistence of tumors and antitumor T cells in tumors (4, 5). These studies demonstrated that A2AR signaling inhibited important effector functions of T cells, such as secretion of pro-inflammatory cytokines (e.g., IFNγ) (6). However, the anti-inflammatory effects of the hypoxia-adenosine-A2AR axis have been confirmed and extended to include suppression of T cell proliferation, cytotoxicity, and induction of anti-inflammatory cytokine secretion (e.g., IL-10) (79).

The hypoxia-adenosine-A2AR axis of immunosuppression begins with hypoxia and the stabilization of hypoxia-inducible factor-1alpha (HIF-1α), which increases extracellular adenosine in part by upregulating adenosine-generating enzymes. Subsequent signaling through the Gs-coupled/cAMP-elevating A2ARs induces protein kinase A (PKA)-mediated inhibition of T-cell receptor signaling and immunosuppressive transcriptional changes (10). This includes the inhibition of pro-inflammatory cytokine secretion and an increase in the levels of anti-inflammatory cytokines that contain a cAMP response element (CRE) consensus sequence in their respective promoter regions. While adenosine can also activate cAMP-elevating adenosine A2B receptors (A2BRs), our research has focused on A2AR adenosinergic immunosuppression due to a higher affinity for adenosine and higher expression on T cells (1113). Importantly, A2AR expression seems to be the limiting factor in adenosine-mediated cAMP generation in T cells since there is no receptor reserve of A2AR (14). T cells can also possess a memory of A2AR signaling, allowing the effects of adenosine to persist long after exposure (15).

Adenosine also exerts immunosuppressive effects through A2BR, particularly on innate immune cells. Groundwork for this hypothesis can be found in studies demonstrating that adenosinergic immunosuppression of IL-12 and TNFα by macrophages is at least partially A2AR-independent (16). For example, in lipopolysaccharide-stimulated macrophages, A2BR activation increases anti-inflammatory IL-10 production by attenuating translational arrest of IL-10 mRNA (17). Conversely, A2BR signaling may enhance activation of alternative/Th2 cytokine-activated macrophages, which manifest several anti-inflammatory functions (18). In group 2 innate lymphoid cells (IL2C), adenosine has been demonstrated to decrease IL-5 and IL-13 production through A2BR, but increase IL-5 production through A2AR. Activation of both A2AR and A2BR in IL2C results in a net decrease in IL-5 production, indicating the importance of A2BR on this cell type (19). Interestingly, HIF-1α-dependent expression of A2BR has also been shown to induce the enrichment of breast cancer stem cells (20). Additional studies of preclinical models of acute lung injury have also demonstrated that an increase in HIF-1α levels in pulmonary epithelia subjected to cyclic mechanical stretch resulted in an increase in A2BR expression (21). A2BR-mediated immunosuppression of a variety of immune cells, including dendritic cells, has led to the development of dual A2AR/A2BR antagonists which may prevent adenosinergic immunosuppression of both innate and adaptive immune cells (22).

The main metabolic precursor to adenosine is ATP. Under homeostatic conditions, ATP is magnitudes higher intracellularly than in the extracellular space (23, 24). However, in inflamed and cancerous tissues, apoptotic and necrotic cells release ATP into the extracellular compartment, disrupting this gradient (25). Excess ATP is then degraded into adenosine by CD39/CD73 (2629), CD38/CD203a (3033) and other phosphatases in certain tissues (28). While the primary mechanism is thought to be mediated by CD39 and CD73 (34), alternative adenosine-generating pathways, such as CD38, are an important contributor to adenosine levels in the TME and inhibit antitumor T cells via A2AR. Indeed, recent studies have demonstrated that PD-1 blockade can increase CD38 expression, leading to resistance to αPD-1 therapy (35).

Consistent with findings regarding adenosine-A2AR immunosuppression, multiple studies from different teams have confirmed the tissue-protecting roles of CD39 and CD73. CD39, which converts ATP to AMP, also serves an anticoagulant function in vasculature (36). Indeed, CD39 has been demonstrated to attenuate both renal ischemia and acute lung injury (37, 38). CD73, which converts AMP to adenosine, has also been shown to have a role in the mediation of cell adhesion to endothelium (39). Moreover, some tumorigenic functions of CD73 have been shown to be independent of its enzymatic function, such as induction of angiogenesis (40). Interestingly, recent studies have also shown that A2AR signaling can promote angiogenesis, suggesting a role for the HIF-1α-CD73-adenosine-A2AR axis in tumor-associated lymphangiogenesis and metastasis (41).

The upstream portion of the hypoxia-adenosine-A2AR axis is mediated by hypoxia/HIF-1α. HIF-1α upregulates genes containing an hypoxia response element (HRE) consensus sequence that mediates cell survival in hypoxic conditions. The immunosuppressive role of HIF-1α was first implicated in studies of HIF-1α−/− Rag-2−/− mice with HIF-1α deletion in T cells and B cells. These experiments demonstrated that HIF-1α regulates lymphocyte development and prevents autoimmunity (42). Subsequent studies of mice with T cell-specific HIF-1α deletion confirmed an immunosuppressive role for HIF-1α. These mice exhibited an enhanced antibacterial response due to the lack of HIF-1α-mediated inhibition of T cells (43). Studies that prevent HIF-1α stabilization using supplemental oxygenation have also provided direct mechanistic evidence for HIF-1α-mediated upregulation of the hypoxia-adenosine-A2AR axis (44). It must be emphasized that upregulation of CRE-containing genes and HRE-containing genes may not be mutually exclusive. The gene encoding the characteristic regulatory T-cell transcription factor FoxP3, which upregulates HIF-1α, is induced by CRE activation (45, 46). Thus, it is suggested that crosstalk exists between CRE and HRE pathways and they may synergize to strengthen immunosuppression (47, 48). Physiologically, this is supported by the infectious tolerance mediated by regulatory T cells in inflamed and cancerous tissues (4951).

Pharmacological Targets in the Hypoxia-Adenosine-A2AR Axis for Cancer Immunotherapy

A2ARs

Inquiry into the immunosuppressive functions of adenosine was catalyzed by the established importance of cAMP as an immunosuppressive agent (52). cAMP has been demonstrated to inhibit many effector T cell functions via PKA activation (5359). Landmark studies by Sitkovsky provided the first genetic and pharmacological evidence that the cAMP-elevating A2AR has a critical and non-redundant immunosuppressive role in tissue protection during excessive inflammation (6). These studies also offered insights into why antitumor T cells often fail to mount an effective response against cancerous tissue. Indeed, tumors are rich in extracellular adenosine, in large part due to poor, irregular vasculature resulting in local hypoxia (6062). The tumor-protecting role of A2AR was conclusively established using mice with A2AR gene deletion (5). This study also complemented genetic evidence with pharmacological data, demonstrating that A2AR antagonism or silencing by siRNA enhanced the efficacy of adoptive cell transfer (ACT) (5). This was supported by follow-up studies demonstrating that A2AR antagonism during ACT or adoptive transfer of A2AR-deficient T cells were effective approaches for enhancing the efficacy of ACT in mice (63). The therapeutic benefit of A2AR antagonism was shown to be due in part by increased IFNγ secretion by tumor-infiltrating adoptively transferred T cells (63). Importantly, this study also demonstrated that A2AR antagonism improved anti-tumor immunity independent of the anatomical location of the tumor and provided long-term tumor-specific memory (63). Taken together, these studies provided proof of principal for the use of A2AR antagonists during cancer immunotherapies, particularly ACT.

The progress in methods of ACT and the studies reviewed above offered justification to test whether CAR-T cells might also be susceptible to hypoxia-adenosinergic immunosuppression. It has been hypothesized that A2AR blockade may improve efficacy of CAR-T therapies against cancers. This may prove essential for CAR-T that target solid tumors, which are known to be hypoxic and extracellular adenosine-rich. Indeed, early evidence was provided by Albelda's group demonstrating that genetic engineering to prevent PKA trafficking to the CAR-T cell membrane enhanced antitumor function in vivo and conferred resistance to adenosinergic immunosuppression in vitro (64). Critical studies by Darcy's Team demonstrated that both pharmacological and genetic inhibition of A2AR enhanced CAR-T efficacy in two distinct murine models of syngeneic breast cancer. Of clinical relevance, addition of αPD-1 to the CAR-T/A2AR blockade protocol further enhanced CAR-T efficacy, as indicated by increased IFNγ production by CAR-T (65). These findings confirm and extend the observations that A2AR antagonism enhances production of IFNγ by polyclonal adoptively transferred T cells in the TME to improve tumor regression (63).

Pioneering studies by Powell's Team established that A2AR agonism can upregulate negative regulators of the immune response such as LAG-3 (8). Subsequent studies using the A2AR antagonist CPI-444 have also provided strong justification for A2AR blockade during cancer immunotherapies. These studies confirmed and extended observations of improved antitumor efficacy of ACT in combination with A2AR blockade. Additional mechanistic evidence justifying A2AR blockade was provided by demonstrations that A2AR blockade reduced PD-1 and LAG-3 expression on effector and regulatory T cells, as well as reduced expression of these immune checkpoint molecules in tumor-draining lymph nodes (66). Taken together, these findings indicate that A2AR blockade can prevent inhibition of already active antitumor T cells, and also prevent inhibition during initial activation (66). Consistent with this finding, it has also been demonstrated that A2AR deletion increases terminally mature natural killer cells in the TME, implicating adenosine as a negative regulator of innate immune cell maturation as well (67). Important studies by Miller and Willingham in multiple preclinical cancer models confirmed that combining A2AR antagonism with checkpoint blockade improved tumor regression, strengthening mechanistic evidence to justify clinical testing of this approach (68). In vitro assays also demonstrated that CPI-444 prevented adenosinergic inhibition of IL-2 and IFNγ production by T cells (68). Through analysis of gene expression, these studies were also able to identify a Th1 expression signature that was associated with positive responses to dual blockade of A2AR/PD-L1 (68).

These preclinical studies have led to the clinical testing of A2AR antagonists as a cancer therapy and have yielded promising results. Against renal cell cancer, A2AR antagonism using CPI-444 induced durable responses both as a monotherapy and when combined with the PD-L1 inhibitor atezolizumab. Patients experiencing positive responses included individuals who had previously shown resistance to αPD-L1 therapy. Consistent with preclinical data, alleviation of adenosinergic immunosuppression resulted in higher cytotoxic T cell tumor infiltration. This study also elucidated a gene-expression signature that was associated with positive response (69). In another clinical study, the A2AR antagonist NIR178 administered both as a monotherapy and in combination with the PD-1 inhibitor spartalizumab to 24 non-small lung cancer patients resulted in stable disease in fifteen patients in addition to one partial response and one complete response (70). Furthermore, the A2AR antagonist AZD4635 used as a monotherapy and in combination with the PD-L1 inhibitor durvalumab induced strong responses in three of eight metastatic castration-resistant prostate cancer patients (71). These tumors may be naturally adenosine-rich due to prostatic acid phosphatase activity and therefore a good candidate for A2AR blockade (71).

CD39/CD73

It has been established that CD39/CD73 also have a major role in facilitating immune escape by tumors. Indeed, Robson's Team established the field of CD39 and were the first to demonstrate that CD39 deletion alleviated tumor burden in a preclinical model of hepatic metastatic cancer (72). Parallel studies by Smyth's Team also demonstrated that administration of a CD73 monoclonal antibody (mAb) decreased tumor burden in two distinct murine tumor models. This approach also suggested that not only did CD73 inhibit antitumor leukocytes via adenosine generation, but affected tumor metastasis as well (73). Moreover, Stagg's Team demonstrated that CD73 overexpression in human triple-negative breast cancer correlated with poor prognosis and resistance to chemotherapy in a preclinical model of breast cancer (74). Important studies by Smyth's Team also demonstrated improved anti-tumor efficacy using an A2AR antagonist in combination with a CD73 inhibitor to alleviate tumor burden (75). These findings also highlight the importance of targeting multiple components of the hypoxia-adenosine-A2AR axis. Indeed, small molecule inhibitors or monoclonal antibodies against CD39 and CD73 are emerging as potent anti-cancer therapies (49, 74, 7682). Furthermore, αCD73 therapy has been demonstrated to improve the therapeutic benefit of αPD-1/αCTLA-4 therapy in multiple preclinical cancer models (80).

Several mAb CD73 inhibitors have exhibited strong antitumor efficacy in clinical trials with findings consistent with preclinical data. In 66 pancreatic or colorectal cancer patients, the αCD73 mAb MEDI9447 as monotherapy and in combination with durvalumab decreased CD73 expression on peripheral T cells. In addition, MEDI9447 decreased CD73 expression in five out of nine tumors, which correlated with increased cytotoxic T cell infiltration (83). The αCD73 mAb BMS986179 as a monotherapy and in combination with the PD-1 inhibitor nivolumab also induced partial responses or stable disease in 17 of 59 patients with various malignancies (84).

HIF-1α

Given the hypoxia-HIF-1α-mediated upregulation of adenosine-generating enzymes, Sitkovsky's Team established in decades-long studies that hypoxia-HIF-1α inhibits T cells (10). It was then hypothesized and confirmed that the reversal of hypoxia could prevent the inhibition of antitumor T cells by hypoxia-adenosine-A2AR-mediated immunosuppression. Indeed, preclinical studies demonstrated that supplemental oxygen (60% O2) decreased levels of hypoxia, HIF-1α, and extracellular adenosine in the TME (44). This was supported by data demonstrating oxygenation-mediated reduction in CD39, CD73, A2AR, A2BR, and COX-2 expression (44). Importantly, supplemental oxygen was also shown to upregulate MHC class I expression by tumor cells, allowing for increased recognition and subsequent elimination by antitumor T cells (44). Parallel studies demonstrated the immunological effects of supplemental oxygen by showing that oxygenation converts an immunosuppressive TME to an immunopermissive TME. This resulted in an increase in many pro-inflammatory cytokines as well as recruitment of endogenous and adoptively transferred antitumor T cells into the TME. This was also accompanied by a reduction in many anti-inflammatory molecules such as TGFβ, CTLA-4, and FoxP3, as well as an overall reduction in regulatory T cells in the TME (85). This resulted in significant tumor regression and long-term survival in preclinical tumor models. Importantly, these studies also established that the reversal of hypoxia improved the efficacy of immune checkpoint blockade with αCTLA-4/αPD-1 (85).

HIF-1α can also be pharmacologically targeted using small molecule drugs such as digoxin, acriflavine, and ganetespib. Indeed, these drugs have shown efficacy in preclinical tumor models (8688). While the immunosuppressive effects of HIF-1α have been shown to be mediated in part by hypoxia-adenosinergic signaling, HIF-1α also has other non-adenosinergic immunosuppressive effects (89). Additionally, immunosuppression via adenosine-A2AR signaling may not be completely reversed by only targeting hypoxia/HIF-1α. Therefore, an ideal approach for completely abrogating the immunosuppressive effects of the hypoxia-adenosine-A2AR axis might be the co-administration of both anti-hypoxia-HIF-1α therapies and A2AR antagonists during cancer immunotherapy (90).

Conclusion

The hypoxia-adenosine-A2AR axis is a potent inhibitor of antitumor T cells. This pathway presents multiple pharmacological targets. Of particular importance and translational value are A2ARs, CD39/CD73, and HIF-1α. Inhibition of this pathway has been shown to enhance the efficacy of current cancer immunotherapy approaches, including αCTLA-4/αPD-1. Multiple studies have reported synergism between checkpoint inhibitors and several classes of anti-hypoxia-adenosine-A2AR therapeutics. Our preclinical studies provided the rationale and justification for combining A2AR blockade and supplemental oxygen/oxygenation agents during cancer immunotherapies. We postulate that this approach will maximize the efficacy of the antitumor immune response in clinical studies.

Author Contributions

All authors listed have made a substantial, direct and intellectual contribution to the work, and approved it for publication.

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

1. Sitkovsky MV, Lukashev D, Apasov S, Kojima H, Koshiba M, Caldwell C, et al. Physiological control of immune response and inflammatory tissue damage by hypoxia-inducible factors and adenosine A2A receptors. Annu Rev Immunol. (2004) 22:657–82. doi: 10.1146/annurev.immunol.22.012703.104731

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Sitkovsky M, Lukashev D. Regulation of immune cells by local-tissue oxygen tension: HIF1 alpha and adenosine receptors. Nat Rev Immunol. (2005) 5:712–21. doi: 10.1038/nri1685

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Sitkovsky MV, Ohta A. The 'danger' sensors that STOP the immune response: the A2 adenosine receptors? Trends Immunol. (2005) 26:299–304. doi: 10.1016/j.it.2005.04.004

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Hellström I, Hellström KE, Pierce GE, Yang JP. Cellular and humoral immunity to different types of human neoplasms. Nature. (1968) 220:1352–4. doi: 10.1038/2201352a0

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Ohta A, Gorelik E, Prasad SJ, Ronchese F, Lukashev D, Wong MK, et al. A2A adenosine receptor protects tumors from antitumor T cells. Proc Natl Acad Sci USA. (2006) 103:13132–7. doi: 10.1073/pnas.0605251103

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Ohta A, Sitkovsky M. Role of G-protein-coupled adenosine receptors in downregulation of inflammation and protection from tissue damage. Nature. (2001) 414:916–20. doi: 10.1038/414916a

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Haskó G, Szabó C, Németh ZH, Kvetan V, Pastores SM, Vizi ES. Adenosine receptor agonists differentially regulate IL-10, TNF-alpha and nitric oxide production in RAW 264.7 macrophages and in endotoxemic mice. J Immunol. (1996) 157:4634–40.

PubMed Abstract | Google Scholar

8. Zarek PE, Huang CT, Lutz ER, Kowalski J, Horton MR, Linden J, et al. A2A receptor signaling promotes peripheral tolerance by inducing T-cell anergy and the generation of adaptive regulatory T cells. Blood. (2008) 111:251–9. doi: 10.1182/blood-2007-03-081646

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Ohta A, Ohta A, Madasu M, Kini R, Subramanian M, Goel N, et al. A2A adenosine receptor may allow expansion of T cells lacking effector functions in extracellular adenosine-rich microenvironments. J Immunol. (2009) 183:5487–93. doi: 10.4049/jimmunol.0901247

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Sitkovsky MV, Hatfield S, Abbott R, Belikoff B, Lukashev D, Ohta A. Hostile, hypoxia-A2-adenosinergic tumor biology as the next barrier to overcome for tumor immunologists. Cancer Immunol Res. (2014) 2:598–605. doi: 10.1158/2326-6066.CIR-14-0075

PubMed Abstract | CrossRef Full Text | Google Scholar

11. 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. doi: 10.1182/blood.V90.4.1600.1600_1600_1610

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Apasov SG, Koshiba M, Chused TM, Sitkovsky MV. Effects of extracellular ATP and adenosine on different thymocyte subsets: possible role of ATP-gated channels and G protein-coupled purinergic receptor. J Immunol. (1997) 158:5095–105.

PubMed Abstract | Google Scholar

13. Koshiba M, Rosin DL, Hayashi N, Linden J, Sitkovsky MV. Patterns of A2A extracellular adenosine receptor expression in different functional subsets of human peripheral T cells. Flow cytometry studies with anti-A2A receptor monoclonal antibodies. Mol Pharmacol. (1999) 55:614–24.

PubMed Abstract | Google Scholar

14. Armstrong JM, Chen JF, Schwarzschild MA, Apasov S, Smith PT, Caldwell C, et al. Gene dose effect reveals no Gs-coupled A2A adenosine receptor reserve in murine T-lymphocytes: studies of cells from A2A-receptor-gene-deficient mice. Biochem J. (2001) 354(Pt 1):123–30. doi: 10.1042/bj3540123

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Koshiba M, Kojima H, Huang S, Apasov S, Sitkovsky MV. Memory of extracellular adenosine A2A purinergic receptor-mediated signaling in murine T cells. J Biol Chem. (1997) 272:25881–9. doi: 10.1074/jbc.272.41.25881

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Haskó G, Kuhel DG, Chen JF, Schwarzschild MA, Deitch EA, Mabley JG, et al. Adenosine inhibits IL-12 and TNF-[alpha] production via adenosine A2a receptor-dependent and independent mechanisms. FASEB J. (2000) 14:2065–74. doi: 10.1096/fj.99-0508com

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Németh ZH, Lutz CS, Csóka B, Deitch EA, Leibovich SJ, Gause WC, et al. Adenosine augments IL-10 production by macrophages through an A2B receptor-mediated posttranscriptional mechanism. J Immunol. (2005) 175:8260–70. doi: 10.4049/jimmunol.175.12.8260

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Csóka B, Selmeczy Z, Koscsó B, Németh ZH, Pacher P, Murray PJ, et al. Adenosine promotes alternative macrophage activation via A2A and A2B receptors. FASEB J. (2012) 26:376–86. doi: 10.1096/fj.11-190934

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Csoka B, Nemeth ZH, Duerr CU, Fritz JH, Pacher P, Hasko G. Adenosine receptors differentially regulate type 2 cytokine production by IL-33-activated bone marrow cells, ILC2s, and macrophages. FASEB J. (2018) 32:829–37. doi: 10.1096/fj.201700770R

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Lan J, Lu H, Samanta D, Salman S, Lu Y, Semenza GL. Hypoxia-inducible factor 1-dependent expression of adenosine receptor 2B promotes breast cancer stem cell enrichment. Proc Natl Acad Sci USA. (2018) 115:E9640–8. doi: 10.1073/pnas.1809695115

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Eckle T, Kewley EM, Brodsky KS, Tak E, Bonney S, Gobel M, et al. Identification of hypoxia-inducible factor HIF-1A as transcriptional regulator of the A2B adenosine receptor during acute lung injury. J Immunol. (2014) 192:1249–56. doi: 10.4049/jimmunol.1100593

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Galezowski M, Wȩgrzyn P, Bobowska A, Dziedzic K, Szeremeta-Spisak J, Nowogrodzki M, et al. Abstract 4135: Novel dual A2A/A2B adenosine receptor antagonists for cancer immunotherapy: in vitro and in vivo characterization. Cancer Res. (2019) 79:4135. doi: 10.1158/1538-7445.SABCS18-4135

CrossRef Full Text | Google Scholar

23. Milo R, Jorgensen P, Moran U, Weber G, Springer M. BioNumbers–the database of key numbers in molecular and cell biology. Nucleic Acids Res. (2010) 38(Database issue):D750–3. doi: 10.1093/nar/gkp889

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Falzoni S, Donvito G, Di Virgilio F. Detecting adenosine triphosphate in the pericellular space. Interface Focus. (2013) 3:20120101. doi: 10.1098/rsfs.2012.0101

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Di Virgilio F, Sarti AC, Falzoni S, De Marchi E, Adinolfi E. Extracellular ATP and P2 purinergic signalling in the tumour microenvironment. Nat Rev Cancer. (2018) 18:601–18. doi: 10.1038/s41568-018-0037-0

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Zimmermann H. Extracellular metabolism of ATP and other nucleotides. Naunyn Schmiedebergs Arch Pharmacol. (2000) 362:299–309. doi: 10.1007/s002100000309

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Yegutkin GG. Nucleotide- and nucleoside-converting ectoenzymes: important modulators of purinergic signalling cascade. Biochim Biophys Acta. (2008) 1783:673–94. doi: 10.1016/j.bbamcr.2008.01.024

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Zimmermann H, Zebisch M, Sträter N. Cellular function and molecular structure of ecto-nucleotidases. Purinergic Signal. (2012) 8:437–502. doi: 10.1007/s11302-012-9309-4

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Allard B, Longhi MS, Robson SC, Stagg J. The ectonucleotidases CD39 and CD73: Novel checkpoint inhibitor targets. Immunol Rev. (2017) 276:121–44. doi: 10.1111/imr.12528

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Adriouch S, Haag F, Boyer O, Seman M, Koch-Nolte F. Extracellular NAD(+): a danger signal hindering regulatory T cells. Microbes Infect. (2012) 14:1284–92. doi: 10.1016/j.micinf.2012.05.011

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Haag F, Adriouch S, Brass A, Jung C, Moller S, Scheuplein F, et al. Extracellular NAD and ATP: Partners in immune cell modulation. Purinergic Signal. (2007) 3:71–81. doi: 10.1007/s11302-006-9038-7

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Chini EN, Chini CCS, Espindola Netto JM, de Oliveira GC, van Schooten W. The pharmacology of CD38/NADase: an emerging target in cancer and diseases of aging. Trends Pharmacol Sci. (2018) 39:424–36. doi: 10.1016/j.tips.2018.02.001

PubMed Abstract | CrossRef Full Text | Google Scholar

33. 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:e26246. doi: 10.4161/onci.26246

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Vigano S, Alatzoglou D, Irving M, Menetrier-Caux C, Caux C, Romero P, et al. Targeting adenosine in cancer immunotherapy to enhance T-cell function. Front Immunol. (2019) 10:925. doi: 10.3389/fimmu.2019.00925

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Chen L, Diao L, Yang Y, Yi X, Rodriguez BL, Li Y, et al. CD38-mediated immunosuppression as a mechanism of tumor cell escape from PD-1/PD-L1 blockade. Cancer Discov. (2018) 8:1156–75. doi: 10.1158/2159-8290.CD-17-1033

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Kaczmarek E, Koziak K, Sévigny J, Siegel JB, Anrather J, Beaudoin AR, et al. Identification and characterization of CD39/vascular ATP diphosphohydrolase. J Biol Chem. (1996) 271:33116–22. doi: 10.1074/jbc.271.51.33116

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Grenz A, Zhang H, Hermes M, Eckle T, Klingel K, Huang DY, et al. Contribution of E-NTPDase1 (CD39) to renal protection from ischemia-reperfusion injury. FASEB J. (2007) 21:2863–73. doi: 10.1096/fj.06-7947com

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Eckle T, Füllbier L, Wehrmann M, Khoury J, Mittelbronn M, Ibla J, et al. Identification of ectonucleotidases CD39 and CD73 in innate protection during acute lung injury. J Immunol. (2007) 178:8127–37. doi: 10.4049/jimmunol.178.12.8127

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Airas L, Hellman J, Salmi M, Bono P, Puurunen T, Smith DJ, et al. CD73 is involved in lymphocyte binding to the endothelium: characterization of lymphocyte-vascular adhesion protein 2 identifies it as CD73. J Exp Med. (1995) 182:1603–8. doi: 10.1084/jem.182.5.1603

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Allard B, Turcotte M, Spring K, Pommey S, Royal I, Stagg J. Anti-CD73 therapy impairs tumor angiogenesis. Int J Cancer. (2014) 134:1466–73. doi: 10.1002/ijc.28456

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Allard B, Cousineau I, Allard D, Buisseret L, Pommey S, Chrobak P, et al. Adenosine A2a receptor promotes lymphangiogenesis and lymph node metastasis. Oncoimmunology. (2019) 8:1601481. doi: 10.1080/2162402X.2019.1601481

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Kojima H, Gu H, Nomura S, Caldwell CC, Kobata T, Carmeliet P, et al. Abnormal B lymphocyte development and autoimmunity in hypoxia-inducible factor 1alpha -deficient chimeric mice. Proc Natl Acad Sci USA. (2002) 99:2170–4. doi: 10.1073/pnas.052706699

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Thiel M, Caldwell CC, Kreth S, Kuboki S, Chen P, Smith P, et al. Targeted deletion of HIF-1alpha gene in T cells prevents their inhibition in hypoxic inflamed tissues and improves septic mice survival. PLoS ONE. (2007) 2:e853. doi: 10.1371/journal.pone.0000853

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Hatfield SM, Kjaergaard J, Lukashev D, Belikoff B, Schreiber TH, Sethumadhavan S, et al. Systemic oxygenation weakens the hypoxia and hypoxia inducible factor 1alpha-dependent and extracellular adenosine-mediated tumor protection. J Mol Med. (2014) 92:1283–92. doi: 10.1007/s00109-014-1189-3

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Kim H.P., Leonard WJ. CREB/ATF-dependent T cell receptor-induced FoxP3 gene expression: a role for DNA methylation. J Exp Med. (2007) 204:1543–51. doi: 10.1084/jem.20070109

CrossRef Full Text | Google Scholar

46. Zheng Y, Josefowicz SZ, Kas A, Chu TT, Gavin MA, Rudensky AY. Genome-wide analysis of Foxp3 target genes in developing and mature regulatory T cells. Nature. (2007) 445:936–40. doi: 10.1038/nature05563

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Ramanathan M, Pinhal-Enfield G, Hao I, Leibovich SJ. Synergistic up-regulation of vascular endothelial growth factor (VEGF) expression in macrophages by adenosine A2A receptor agonists and endotoxin involves transcriptional regulation via the hypoxia response element in the VEGF promoter. Mol Biol Cell. (2007) 18:14–23. doi: 10.1091/mbc.e06-07-0596

PubMed Abstract | CrossRef Full Text | Google Scholar

48. De Ponti C, Carini R, Alchera E, Nitti MP, Locati M, Albano E, et al. Adenosine A2a receptor-mediated, normoxic induction of HIF-1 through PKC and PI-3K-dependent pathways in macrophages. J Leukoc Biol. (2007) 82:392–402. doi: 10.1189/jlb.0107060

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Deaglio S, Dwyer KM, Gao W, Friedman D, Usheva A, Erat A, et al. Adenosine generation catalyzed by CD39 and CD73 expressed on regulatory T cells mediates immune suppression. J Exp Med. (2007) 204:1257–65. doi: 10.1084/jem.20062512

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Sitkovsky MV. T regulatory cells: hypoxia-adenosinergic suppression and re-direction of the immune response. Trends Immunol. (2009). 30:102–8. doi: 10.1016/j.it.2008.12.002

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Ohta A, Kini R, Ohta A, Subramanian M, Madasu M, Sitkovsky M. The development and immunosuppressive functions of CD4(+) CD25(+) FoxP3(+) regulatory T cells are under influence of the adenosine-A2A adenosine receptor pathway. Front Immunol. (2012) 3:190. doi: 10.3389/fimmu.2012.00190

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Cronstein BN, Levin RI, Belanoff J, Weissmann G, Hirschhorn R. Adenosine: an endogenous inhibitor of neutrophil-mediated injury to endothelial cells. J Clin Invest. (1986) 78:760–70. doi: 10.1172/JCI112638

PubMed Abstract | CrossRef Full Text | Google Scholar

53. Takayama H, Sitkovsky MV. Antigen receptor-regulated exocytosis in cytotoxic T lymphocytes. J Exp Med. (1987) 166:725–43. doi: 10.1084/jem.166.3.725

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Sitkovsky MV. Mechanistic, functional and immunopharmacological implications of biochemical studies of antigen receptor-triggered cytolytic T-lymphocyte activation. Immunol Rev. (1988) 103:127–60. doi: 10.1111/j.1600-065X.1988.tb00754.x

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Sitkovsky MV, Trenn G, Takayama H. Cyclic AMP-dependent protein kinase as a part of the possible down-regulating pathway in the antigen receptor-regulated cytotoxic T lymphocyte conjugate formation and granule exocytosis. Ann N Y Acad Sci. (1988) 532:350–8. doi: 10.1111/j.1749-6632.1988.tb36352.x

PubMed Abstract | CrossRef Full Text | Google Scholar

56. Takayama H, Sitkovsky MV. Potential use of an antagonist of cAMP-dependent protein kinase to block inhibition and modulate T-cell receptor-triggered activation of cytotoxic T-lymphocytes. J Pharm Sci. (1989) 78:8–10. doi: 10.1002/jps.2600780104

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Bjørgo E, Moltu K, Tasken K. Phosphodiesterases as targets for modulating T-cell responses. Handb Exp Pharmacol. (2011) 2011:345-63. doi: 10.1007/978-3-642-17969-3_15

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Trenn G, Takayama H, Sitkovsky MV. Antigen-receptor regulated exocytosis of cytolytic granules may not be required for target cell lysis by cytotoxic T lymphocytes. Nature. (1987) 330:72–4. doi: 10.1038/330072a0

PubMed Abstract | CrossRef Full Text | Google Scholar

59. Takayama H, Trenn G, Sitkovsky MV. Locus of inhibitory action of cAMP-dependent protein kinase in the antigen receptor-triggered cytotoxic t lymphocyte activation pathway. J Biol Chem. (1988) 263:2330–6.

PubMed Abstract | Google Scholar

60. Dewhirst MW, Cao Y, Moeller B. Cycling hypoxia and free radicals regulate angiogenesis and radiotherapy response. Nat Rev Cancer. (2008) 8:425–37. doi: 10.1038/nrc2397

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Sitkovsky MV. Damage control by hypoxia-inhibited. A Blood K. (2008) 111:5424–5. doi: 10.1182/blood-2008-03-143990

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Vijayan D, Young A, Teng MWL, Smyth MJ. Targeting immunosuppressive adenosine in cancer. Nat Rev Cancer. (2017) 17:709–24. doi: 10.1038/nrc.2017.86

CrossRef Full Text | Google Scholar

63. Kjaergaard J, Hatfield S, Jones G, Ohta A, Sitkovsky M. A2A adenosine receptor gene deletion or synthetic A2A antagonist liberate tumor-reactive CD8(+) T cells from tumor-induced immunosuppression. J Immunol. (2018) 201:782–91. doi: 10.4049/jimmunol.1700850

PubMed Abstract | CrossRef Full Text | Google Scholar

64. Newick K, O'Brien S, Sun J, Kapoor V, Maceyko S, Lo A, et al. Augmentation of CAR t-cell trafficking and antitumor efficacy by blocking protein kinase a localization. Cancer Immunol Res. (2016) 4:541–51. doi: 10.1158/2326-6066.CIR-15-0263

PubMed Abstract | CrossRef Full Text | Google Scholar

65. Beavis PA, Henderson MA, Giuffrida L, Mills JK, Sek K, Cross RS, et al. Targeting the adenosine 2A receptor enhances chimeric antigen receptor T cell efficacy. J Clin Invest. (2017) 127:929–41. doi: 10.1172/JCI89455

PubMed Abstract | CrossRef Full Text | Google Scholar

66. Leone RD, Sun IM, Oh MH, Sun IH, Wen J, Englert J, et al. Inhibition of the adenosine A2a receptor modulates expression of T cell coinhibitory receptors and improves effector function for enhanced checkpoint blockade and ACT in murine cancer models. Cancer Immunol Immunother. (2018) 67:1271–84. doi: 10.1007/s00262-018-2186-0

PubMed Abstract | CrossRef Full Text | Google Scholar

67. Young A, Ngiow SF, Gao Y, Patch AM, Barkauskas DS, Messaoudene M, et al. A2AR adenosine signaling suppresses natural killer cell maturation in the tumor microenvironment. Cancer Res. (2018) 78:1003–16. doi: 10.1158/0008-5472.CAN-17-2826

PubMed Abstract | CrossRef Full Text | Google Scholar

68. 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) 6:1136–49. doi: 10.1158/2326-6066.CIR-18-0056

PubMed Abstract | CrossRef Full Text | Google Scholar

69. 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) 10:40–53. doi: 10.1158/2159-8290.CD-19-0980

PubMed Abstract | CrossRef Full Text | Google Scholar

70. Chiappori A, Williams CC, Creelan BC, Tanvetyanon T, Gray JE, Haura EB, et al. Phase I/II study of the A2AR antagonist NIR178 (PBF-509), an oral immunotherapy, in patients (pts) with NSCLC. J Clin Oncol. (2018) 36:9089. doi: 10.1200/JCO.2018.36.15_suppl.9089

CrossRef Full Text | Google Scholar

71. Bendell J, Bauer T, Patel M, Falchook G, Karlix JL, Lim E, et al. Abstract CT026: Evidence of immune activation in the first-in-human Phase Ia dose escalation study of the adenosine 2a receptor antagonist, AZD4635, in patients with advanced solid tumors. Cancer Res. (2019) 79(13 Supplement):CT026. doi: 10.1158/1538-7445.SABCS18-CT026

CrossRef Full Text | Google Scholar

72. Sun X, Wu Y, Gao W, Enjyoji K, Csizmadia E, Muller CE, et al. CD39/ENTPD1 expression by CD4+Foxp3+ regulatory T cells promotes hepatic metastatic tumor growth in mice. Gastroenterology. (2010) 139:1030–40. doi: 10.1053/j.gastro.2010.05.007

PubMed Abstract | CrossRef Full Text | Google Scholar

73. Stagg J, Divisekera U, McLaughlin N, Sharkey J, Pommey S, Denoyer D, et al. Anti-CD73 antibody therapy inhibits breast tumor growth and metastasis. Proc Natl Acad Sci US A. (2010) 107:1547–52. doi: 10.1073/pnas.0908801107

PubMed Abstract | CrossRef Full Text | Google Scholar

74. Loi S, Pommey S, Haibe-Kains B, Beavis PA, Darcy PK, Smyth MJ, et al. CD73 promotes anthracycline resistance and poor prognosis in triple negative breast cancer. Proc Natl Acad Sci USA. (2013) 110:11091–6. doi: 10.1073/pnas.1222251110

PubMed Abstract | CrossRef Full Text | Google Scholar

75. Young A, Ngiow SF, Barkauskas DS, Sult E, Hay C, Blake SJ, et al. Co-inhibition of CD73 and A2AR adenosine signaling improves anti-tumor immune responses. Cancer Cell. (2016) 30:391–403. doi: 10.1016/j.ccell.2016.06.025

PubMed Abstract | CrossRef Full Text | Google Scholar

76. Feng L, Sun X, Csizmadia E, Han L, Bian S, Murakami T, et al. Vascular CD39/ENTPD1 directly promotes tumor cell growth by scavenging extracellular adenosine triphosphate. Neoplasia. (2011) 13:206–16. doi: 10.1593/neo.101332

PubMed Abstract | CrossRef Full Text | Google Scholar

77. Künzli BM, Bernlochner MI, Rath S, Käser S, Csizmadia E, Enjyoji K, et al. Impact of CD39 and purinergic signalling on the growth and metastasis of colorectal cancer. Purinergic Signal. (2011) 7:231–41. doi: 10.1007/s11302-011-9228-9

PubMed Abstract | CrossRef Full Text | Google Scholar

78. Stagg J, Divisekera U, Duret H, Sparwasser T, Teng MW, Darcy PK, et al. CD73-deficient mice have increased antitumor immunity and are resistant to experimental metastasis. Cancer Res. (2011) 71:2892–900. doi: 10.1158/0008-5472.CAN-10-4246

PubMed Abstract | CrossRef Full Text | Google Scholar

79. Stagg J, Beavis PA, Divisekera U, Liu MC, Moller A, Darcy PK, et al. CD73-deficient mice are resistant to carcinogenesis. Cancer Res. (2012) 72:2190–6. doi: 10.1158/0008-5472.CAN-12-0420

PubMed Abstract | CrossRef Full Text | Google Scholar

80. Allard B, Pommey S, Smyth MJ, Stagg J. Targeting CD73 enhances the antitumor activity of anti-PD-1 and anti-CTLA-4 mAbs. Clin Cancer Res. (2013) 19:5626–35. doi: 10.1158/1078-0432.CCR-13-0545

PubMed Abstract | CrossRef Full Text | Google Scholar

81. Zhang B. CD73: a novel target for cancer immunotherapy. Cancer Res. (2010). 70:6407-11. doi: 10.1158/0008-5472.CAN-10-1544

CrossRef Full Text | Google Scholar

82. Jin D, Fan J, Wang L, Thompson LF, Liu A, Daniel BJ, et al. CD73 on tumor cells impairs antitumor T-cell responses: a novel mechanism of tumor-induced immune suppression. Cancer Res. (2010) 70:2245–55. doi: 10.1158/0008-5472.CAN-09-3109

PubMed Abstract | CrossRef Full Text | Google Scholar

83. Overman MJ, LoRusso P, Strickler JH, Patel SP, Clarke SJ, Noonan AM, et al. Safety, efficacy, and pharmacodynamics (PD) of MEDI9447 (oleclumab) alone or in combination with durvalumab in advanced colorectal cancer (CRC) or pancreatic cancer (panc). J Clin Oncol. (2018) 36:4123–23. doi: 10.1200/JCO.2018.36.15_suppl.4123

CrossRef Full Text | Google Scholar

84. Siu LL, Burris H, Le DT, Hollebecque A, Steeghs N, Delord J-P, et al. Abstract CT180: Preliminary phase 1 profile of BMS-986179, an anti-CD73 antibody, in combination with nivolumab in patients with advanced solid tumors. Cancer Res. (2018) 78:CT180. doi: 10.1158/1538-7445.AM2018-CT180

CrossRef Full Text | Google Scholar

85. Hatfield SM, Kjaergaard J, Lukashev D, Schreiber TH, Belikoff B, Abbott R, et al. Immunological mechanisms of the antitumor effects of supplemental oxygenation. Sci Transl Med. (2015) 7:277ra30. doi: 10.1126/scitranslmed.aaa1260

PubMed Abstract | CrossRef Full Text | Google Scholar

86. Zhang H, Qian DZ, Tan YS, Lee K, Gao P, Ren YR, et al. Digoxin and other cardiac glycosides inhibit HIF-1alpha synthesis and block tumor growth. Proc Natl Acad Sci USA. (2008) 105:19579–86. doi: 10.1073/pnas.0809763105

PubMed Abstract | CrossRef Full Text | Google Scholar

87. Wong CC, Zhang H, Gilkes DM, Chen J, Wei H, Chaturvedi P, et al. Inhibitors of hypoxia-inducible factor 1 block breast cancer metastatic niche formation and lung metastasis. J Mol Med (Berl). (2012) 90:803–15. doi: 10.1007/s00109-011-0855-y

PubMed Abstract | CrossRef Full Text | Google Scholar

88. Xiang L, Gilkes DM, Chaturvedi P, Luo W, Hu H, Takano N, et al. Ganetespib blocks HIF-1 activity and inhibits tumor growth, vascularization, stem cell maintenance, invasion, and metastasis in orthotopic mouse models of triple-negative breast cancer. J Mol Med (Berl). (2014) 92:151–64. doi: 10.1007/s00109-013-1102-5

PubMed Abstract | CrossRef Full Text | Google Scholar

89. Madasu MM, Subramanian M, Kini R, Ohta A, Sitkovsky M, Ohta A. Hypoxia can suppress T cell activation in a mechanism independent of A2A adenosine receptor (46.27). J Immunol. (2012) 188(Supplement 1):46.27.

Google Scholar

90. Sitkovsky MV. Lessons from the A2A adenosine receptor antagonist-enabled tumor regression and survival in patients with treatment-refractory renal cell cancer. Cancer Discov. (2020). 10:16–9. doi: 10.1158/2159-8290.CD-19-1280

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: adenosine, hypoxia, cancer immunotherapies, T cell, HIF−1α, immune checkpoint, immunology

Citation: Steingold JM and Hatfield SM (2020) Targeting Hypoxia-A2A Adenosinergic Immunosuppression of Antitumor T Cells During Cancer Immunotherapy. Front. Immunol. 11:570041. doi: 10.3389/fimmu.2020.570041

Received: 05 June 2020; Accepted: 26 August 2020;
Published: 29 September 2020.

Edited by:

Holger Klaus Eltzschig, The University of Texas Health Science Center at Houston, United States

Reviewed by:

George Hasko, Columbia University, United States
Bertrand Allard, University of Montreal Hospital Center (CRCHUM), Canada

Copyright © 2020 Steingold and Hatfield. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Stephen M. Hatfield, s.hatfield@neu.edu

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.