Skip to main content
Erschienen in: Cancer Immunology, Immunotherapy 8/2018

19.06.2018 | Original Article

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

verfasst von: Robert D. Leone, Im-Meng Sun, Min-Hee Oh, Im-Hong Sun, Jiayu Wen, Judson Englert, Jonathan D. Powell

Erschienen in: Cancer Immunology, Immunotherapy | Ausgabe 8/2018

Einloggen, um Zugang zu erhalten

Abstract

Adenosine signaling via the A2a receptor (A2aR) is emerging as an important checkpoint of immune responses. The presence of adenosine in the inflammatory milieu or generated by the CD39/CD73 axis on tissues or T regulatory cells serves to regulate immune responses. By nature of the specialized metabolism of cancer cells, adenosine levels are increased in the tumor microenvironment and contribute to tumor immune evasion. To this end, small molecule inhibitors of the A2aR are being pursued clinically to enhance immunotherapy. Herein, we demonstrate the ability of the novel A2aR antagonist, CPI-444, to dramatically enhance immunologic responses in models of checkpoint therapy and ACT in cancer. Furthermore, we demonstrate that A2aR blockade with CPI-444 decreases expression of multiple checkpoint pathways, including PD-1 and LAG-3, on both CD8+ effector T cells (Teff) and FoxP3+ CD4+ regulatory T cells (Tregs). Interestingly, our studies demonstrate that A2aR blockade likely has its most profound effects during Teff cell activation, significantly decreasing PD-1 and LAG-3 expression at the draining lymph nodes of tumor bearing mice. In contrast to previous reports using A2aR knockout models, pharmacologic blockade with CPI-444 did not impede CD8 T cell persistence or memory recall. Overall these findings not only redefine our understanding of the mechanisms by which adenosine inhibits immunity but also have important implications for the design of novel immunotherapy regimens.
Anhänge
Nur mit Berechtigung zugänglich
Literatur
1.
Zurück zum Zitat Ohta A, Sitkovsky M (2001) Role of G-protein-coupled adenosine receptors in downregulation of inflammation and protection from tissue damage. Nature 414:916–920CrossRefPubMed Ohta A, Sitkovsky M (2001) Role of G-protein-coupled adenosine receptors in downregulation of inflammation and protection from tissue damage. Nature 414:916–920CrossRefPubMed
2.
Zurück zum Zitat Ohta A, Gorelik E, Prasad SJ, Ronchese F, Lukashev D, Wong MK, Huang X, Caldwell S, Liu K, Smith P, Chen JF, Jackson EK, Apasov S, Abrams S, Sitkovsky M (2006) A2A adenosine receptor protects tumors from antitumor T cells. Proc Natl Acad Sci USA 103:13132–13137CrossRefPubMed Ohta A, Gorelik E, Prasad SJ, Ronchese F, Lukashev D, Wong MK, Huang X, Caldwell S, Liu K, Smith P, Chen JF, Jackson EK, Apasov S, Abrams S, Sitkovsky M (2006) A2A adenosine receptor protects tumors from antitumor T cells. Proc Natl Acad Sci USA 103:13132–13137CrossRefPubMed
3.
Zurück zum Zitat Allard B, Pommey S, Smyth MJ, Stagg J (2013) Targeting CD73 enhances the antitumor activity of anti-PD-1 and anti-CTLA-4 mAbs. Clin Cancer Res 19:5626–5635CrossRefPubMed Allard B, Pommey S, Smyth MJ, Stagg J (2013) Targeting CD73 enhances the antitumor activity of anti-PD-1 and anti-CTLA-4 mAbs. Clin Cancer Res 19:5626–5635CrossRefPubMed
4.
Zurück zum Zitat Beavis PA, Milenkovski N, Henderson MA, John LB, Allard B, Loi S, Kershaw MH, Stagg J, Darcy PK (2015) Adenosine receptor 2A blockade increases the efficacy of anti-PD-1 through enhanced anti-tumor T cell responses. Cancer Immunol Res 3:506–517CrossRef Beavis PA, Milenkovski N, Henderson MA, John LB, Allard B, Loi S, Kershaw MH, Stagg J, Darcy PK (2015) Adenosine receptor 2A blockade increases the efficacy of anti-PD-1 through enhanced anti-tumor T cell responses. Cancer Immunol Res 3:506–517CrossRef
5.
Zurück zum Zitat Waickman AT, Alme A, Senaldi L, Zarek PE, Horton M, Powell JD (2012) Enhancement of tumor immunotherapy by deletion of the A2A adenosine receptor. Cancer Immunol Immunother 61:917–926CrossRefPubMed Waickman AT, Alme A, Senaldi L, Zarek PE, Horton M, Powell JD (2012) Enhancement of tumor immunotherapy by deletion of the A2A adenosine receptor. Cancer Immunol Immunother 61:917–926CrossRefPubMed
6.
Zurück zum Zitat Stagg J, Divisekera U, McLaughlin N, Sharkey J, Pommey S, Denoyer D, Dwyer KM, Smyth MJ (2010) Anti-CD73 antibody therapy inhibits breast tumor growth and metastasis. Proc Natl Acad Sci USA 107:1547–1552CrossRefPubMed Stagg J, Divisekera U, McLaughlin N, Sharkey J, Pommey S, Denoyer D, Dwyer KM, Smyth MJ (2010) Anti-CD73 antibody therapy inhibits breast tumor growth and metastasis. Proc Natl Acad Sci USA 107:1547–1552CrossRefPubMed
7.
Zurück zum Zitat Loi S, Pommey S, Haibe-Kains B, Beavis PA, Darcy PK, Smyth MJ, Stagg J (2013) CD73 promotes anthracycline resistance and poor prognosis in triple negative breast cancer. Proc Natl Acad Sci USA 110:11091–11096CrossRefPubMed Loi S, Pommey S, Haibe-Kains B, Beavis PA, Darcy PK, Smyth MJ, Stagg J (2013) CD73 promotes anthracycline resistance and poor prognosis in triple negative breast cancer. Proc Natl Acad Sci USA 110:11091–11096CrossRefPubMed
8.
Zurück zum Zitat Beavis PA, Divisekera U, Paget C, Chow MT, John LB, Devaud C, Dwyer K, Stagg J, Smyth MJ, Darcy PK (2013) Blockade of A2A receptors potently suppresses the metastasis of CD73 + tumors. Proc Natl Acad Sci USA 110:14711–14716CrossRefPubMed Beavis PA, Divisekera U, Paget C, Chow MT, John LB, Devaud C, Dwyer K, Stagg J, Smyth MJ, Darcy PK (2013) Blockade of A2A receptors potently suppresses the metastasis of CD73 + tumors. Proc Natl Acad Sci USA 110:14711–14716CrossRefPubMed
9.
Zurück zum Zitat Mittal D, Young A, Stannard K, Yong M, Teng MW, Allard B, Stagg J, Smyth MJ (2014) Antimetastatic effects of blocking PD-1 and the adenosine A2A receptor. Cancer Res 74:3652–3658CrossRefPubMed Mittal D, Young A, Stannard K, Yong M, Teng MW, Allard B, Stagg J, Smyth MJ (2014) Antimetastatic effects of blocking PD-1 and the adenosine A2A receptor. Cancer Res 74:3652–3658CrossRefPubMed
10.
Zurück zum Zitat Zarek PE, Huang CT, Lutz ER, Kowalski J, Horton MR, Linden J, Drake CG, Powell JD (2008) A2A receptor signaling promotes peripheral tolerance by inducing T-cell anergy and the generation of adaptive regulatory T cells. Blood 111:251–259CrossRefPubMedPubMedCentral Zarek PE, Huang CT, Lutz ER, Kowalski J, Horton MR, Linden J, Drake CG, Powell JD (2008) A2A receptor signaling promotes peripheral tolerance by inducing T-cell anergy and the generation of adaptive regulatory T cells. Blood 111:251–259CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Parks SK, Cormerais Y, Pouysségur J (2017) Hypoxia and cellular metabolism in tumour pathophysiology. J Physiol (Lond) 595:2439–2450CrossRef Parks SK, Cormerais Y, Pouysségur J (2017) Hypoxia and cellular metabolism in tumour pathophysiology. J Physiol (Lond) 595:2439–2450CrossRef
12.
13.
Zurück zum Zitat Blay J, White TD, Hoskin DW (1997) The extracellular fluid of solid carcinomas contains immunosuppressive concentrations of adenosine. Cancer Res 57:2602–2605PubMed Blay J, White TD, Hoskin DW (1997) The extracellular fluid of solid carcinomas contains immunosuppressive concentrations of adenosine. Cancer Res 57:2602–2605PubMed
14.
Zurück zum Zitat Dubyak GR, el-Moatassim C (1993) Signal transduction via P2-purinergic receptors for extracellular ATP and other nucleotides. Am J Physiol 265:C577–C606CrossRefPubMed Dubyak GR, el-Moatassim C (1993) Signal transduction via P2-purinergic receptors for extracellular ATP and other nucleotides. Am J Physiol 265:C577–C606CrossRefPubMed
15.
Zurück zum Zitat Robeva AS, Woodard RL, Jin X, Gao Z, Bhattacharya S, Taylor HE, Rosin DL, Linden J (1996) Molecular characterization of recombinant human adenosine receptors. Drug Dev Res 39:243–252CrossRef Robeva AS, Woodard RL, Jin X, Gao Z, Bhattacharya S, Taylor HE, Rosin DL, Linden J (1996) Molecular characterization of recombinant human adenosine receptors. Drug Dev Res 39:243–252CrossRef
16.
Zurück zum Zitat Apasov S, Koshiba M, Redegeld F, Sitkovsky MV (1995) Role of extracellular ATP and P1 and P2 classes of purinergic receptors in T-cell development and cytotoxic T lymphocyte effector functions. Immunol Rev 146:5–19CrossRefPubMed Apasov S, Koshiba M, Redegeld F, Sitkovsky MV (1995) Role of extracellular ATP and P1 and P2 classes of purinergic receptors in T-cell development and cytotoxic T lymphocyte effector functions. Immunol Rev 146:5–19CrossRefPubMed
17.
Zurück zum Zitat Apasov SG, Koshiba M, Chused TM, Sitkovsky MV (1997) Effects of extracellular ATP and adenosine on different thymocyte subsets: possible role of ATP-gated channels and G protein-coupled purinergic receptor. J Immunol 158:5095–5105PubMed Apasov SG, Koshiba M, Chused TM, Sitkovsky MV (1997) Effects of extracellular ATP and adenosine on different thymocyte subsets: possible role of ATP-gated channels and G protein-coupled purinergic receptor. J Immunol 158:5095–5105PubMed
18.
Zurück zum Zitat Filippini A, Taffs RE, Agui T, Sitkovsky MV (1990) Ecto-ATPase activity in cytolytic T-lymphocytes. Protection from the cytolytic effects of extracellular ATP. J Biol Chem 265:334–340PubMed Filippini A, Taffs RE, Agui T, Sitkovsky MV (1990) Ecto-ATPase activity in cytolytic T-lymphocytes. Protection from the cytolytic effects of extracellular ATP. J Biol Chem 265:334–340PubMed
19.
Zurück zum Zitat Resta R, Yamashita Y, Thompson LF (1998) Ecto-enzyme and signaling functions of lymphocyte CD73. Immunol Rev 161:95–109CrossRefPubMed Resta R, Yamashita Y, Thompson LF (1998) Ecto-enzyme and signaling functions of lymphocyte CD73. Immunol Rev 161:95–109CrossRefPubMed
20.
Zurück zum Zitat Emens L, Powderly J, Fong L, Brody J, Forde P, Hellmann M, Hughes B, Kummar S, Loi S, Luke J, Mahadevan D, Markman B, McCaffery I, Miller R, Laport G (2017) CPI-444, an oral adenosine A2a receptor (A2aR) antagonist, demonstrates clinical activity in patients with advanced solid tumors. AACR Annual Meeting 2017. Cancer Res 77:Abstract CT119CrossRef Emens L, Powderly J, Fong L, Brody J, Forde P, Hellmann M, Hughes B, Kummar S, Loi S, Luke J, Mahadevan D, Markman B, McCaffery I, Miller R, Laport G (2017) CPI-444, an oral adenosine A2a receptor (A2aR) antagonist, demonstrates clinical activity in patients with advanced solid tumors. AACR Annual Meeting 2017. Cancer Res 77:Abstract CT119CrossRef
21.
Zurück zum Zitat Willingham S, Ho P, Leone R, Piccione E, Choy C, Hotson A, Buggy J, Powell J, Miller R (2016) The adenosine A2A receptor antagonist CPI-444 blocks adenosine-mediated T-cell suppression and exhibits antitumor activity alone and in combination with anti-PD-1 and anti-PD-L1. AACR Annual Meeting 2017. Cancer Res 76:Abstract 2337CrossRef Willingham S, Ho P, Leone R, Piccione E, Choy C, Hotson A, Buggy J, Powell J, Miller R (2016) The adenosine A2A receptor antagonist CPI-444 blocks adenosine-mediated T-cell suppression and exhibits antitumor activity alone and in combination with anti-PD-1 and anti-PD-L1. AACR Annual Meeting 2017. Cancer Res 76:Abstract 2337CrossRef
22.
Zurück zum Zitat Cekic C, Sag D, Day YJ, Linden J (2013) Extracellular adenosine regulates naive T cell development and peripheral maintenance. J Exp Med 210:2693–2706CrossRefPubMedPubMedCentral Cekic C, Sag D, Day YJ, Linden J (2013) Extracellular adenosine regulates naive T cell development and peripheral maintenance. J Exp Med 210:2693–2706CrossRefPubMedPubMedCentral
23.
24.
Zurück zum Zitat Ngiow SF, Young A, Jacquelot N, Yamazaki T, Enot D, Zitvogel L, Smyth MJ (2015) A threshold level of intratumor CD8+ T-cell PD1 expression dictates therapeutic response to anti-PD1. Cancer Res 75:3800–3811CrossRefPubMed Ngiow SF, Young A, Jacquelot N, Yamazaki T, Enot D, Zitvogel L, Smyth MJ (2015) A threshold level of intratumor CD8+ T-cell PD1 expression dictates therapeutic response to anti-PD1. Cancer Res 75:3800–3811CrossRefPubMed
26.
Zurück zum Zitat Huang CT, Workman CJ, Flies D, Pan X, Marson AL, Zhou G, Hipkiss EL, Ravi S, Kowalski J, Levitsky HI, Powell JD, Pardoll DM, Drake CG, Vignali DA (2004) Role of LAG-3 in regulatory T cells. Immunity 21:503–513CrossRefPubMed Huang CT, Workman CJ, Flies D, Pan X, Marson AL, Zhou G, Hipkiss EL, Ravi S, Kowalski J, Levitsky HI, Powell JD, Pardoll DM, Drake CG, Vignali DA (2004) Role of LAG-3 in regulatory T cells. Immunity 21:503–513CrossRefPubMed
28.
Zurück zum Zitat Smyth MJ, Ngiow SF, Ribas A, Teng MWL (2016) Combination cancer immunotherapies tailored to the tumour microenvironment. Nat Rev Clin Oncol 13:143–158CrossRefPubMed Smyth MJ, Ngiow SF, Ribas A, Teng MWL (2016) Combination cancer immunotherapies tailored to the tumour microenvironment. Nat Rev Clin Oncol 13:143–158CrossRefPubMed
29.
30.
Zurück zum Zitat Mockler MB, Conroy MJ, Lysaght J (2014) Targeting T cell immunometabolism for cancer immunotherapy; understanding the impact of the tumor microenvironment. Front Oncol 4:107CrossRefPubMedPubMedCentral Mockler MB, Conroy MJ, Lysaght J (2014) Targeting T cell immunometabolism for cancer immunotherapy; understanding the impact of the tumor microenvironment. Front Oncol 4:107CrossRefPubMedPubMedCentral
32.
Zurück zum Zitat Curran MA, Montalvo W, Yagita H, Allison JP (2010) PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors. Proc Natl Acad Sci USA 107:4275–4280CrossRefPubMed Curran MA, Montalvo W, Yagita H, Allison JP (2010) PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors. Proc Natl Acad Sci USA 107:4275–4280CrossRefPubMed
33.
Zurück zum Zitat Koyama S, Akbay EA, Li YY, Herter-Sprie G, Buczkowski KA, Richards WG, Gandhi L, Redig AJ, Rodig SJ, Asahina H, Jones RE, Kulkarni MM, Kuraguchi M, Palakurthi S, Fecci PE, Johnson BE, Janne PA, Engelman JA, Gangadharan SP, Costa DB, Freeman GJ, Bueno R, Hodi FS, Dranoff G, Wong K, Hammerman PS (2016) Adaptive resistance to therapeutic PD-1 blockade is associated with upregulation of alternative immune checkpoints. Nat Commun 7:10501CrossRefPubMedPubMedCentral Koyama S, Akbay EA, Li YY, Herter-Sprie G, Buczkowski KA, Richards WG, Gandhi L, Redig AJ, Rodig SJ, Asahina H, Jones RE, Kulkarni MM, Kuraguchi M, Palakurthi S, Fecci PE, Johnson BE, Janne PA, Engelman JA, Gangadharan SP, Costa DB, Freeman GJ, Bueno R, Hodi FS, Dranoff G, Wong K, Hammerman PS (2016) Adaptive resistance to therapeutic PD-1 blockade is associated with upregulation of alternative immune checkpoints. Nat Commun 7:10501CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat Kamphorst AO, Wieland A, Nasti T, Yang S, Zhang R, Barber DL, Konieczny BT, Daugherty CZ, Koenig L, Yu K, Sica GL, Sharpe AH, Freeman GJ, Blazar BR, Turka LA, Owonikoko TK, Pillai RN, Ramalingam SS, Araki K, Ahmed R (2017) Rescue of exhausted CD8 T cells by PD-1-targeted therapies is CD28-dependent. Science 355:1423–1427CrossRefPubMedPubMedCentral Kamphorst AO, Wieland A, Nasti T, Yang S, Zhang R, Barber DL, Konieczny BT, Daugherty CZ, Koenig L, Yu K, Sica GL, Sharpe AH, Freeman GJ, Blazar BR, Turka LA, Owonikoko TK, Pillai RN, Ramalingam SS, Araki K, Ahmed R (2017) Rescue of exhausted CD8 T cells by PD-1-targeted therapies is CD28-dependent. Science 355:1423–1427CrossRefPubMedPubMedCentral
35.
Zurück zum Zitat Hui E, Cheung J, Zhu J, Su X, Taylor MJ, Wallweber HA, Sasmal DK, Huang J, Kim JM, Mellman I, Vale RD (2017) T cell costimulatory receptor CD28 is a primary target for PD-1-mediated inhibition. Science 355:1428–1433CrossRefPubMed Hui E, Cheung J, Zhu J, Su X, Taylor MJ, Wallweber HA, Sasmal DK, Huang J, Kim JM, Mellman I, Vale RD (2017) T cell costimulatory receptor CD28 is a primary target for PD-1-mediated inhibition. Science 355:1428–1433CrossRefPubMed
36.
Zurück zum Zitat Chamoto K, Chowdhury PS, Kumar A, Sonomura K, Matsuda F, Fagarasan S, Honjo T (2017) Mitochondrial activation chemicals synergize with surface receptor PD-1 blockade for T cell-dependent antitumor activity. Proc Natl Acad Sci USA 114:E770CrossRef Chamoto K, Chowdhury PS, Kumar A, Sonomura K, Matsuda F, Fagarasan S, Honjo T (2017) Mitochondrial activation chemicals synergize with surface receptor PD-1 blockade for T cell-dependent antitumor activity. Proc Natl Acad Sci USA 114:E770CrossRef
37.
Zurück zum Zitat Vijayan D, Young A, Teng MWL, Smyth MJ (2017) Targeting immunosuppressive adenosine in cancer. Nat Rev Cancer 17:709–724CrossRefPubMed Vijayan D, Young A, Teng MWL, Smyth MJ (2017) Targeting immunosuppressive adenosine in cancer. Nat Rev Cancer 17:709–724CrossRefPubMed
38.
Zurück zum Zitat Smyth LA, Ratnasothy K, Tsang JYS, Boardman D, Warley A, Lechler R, Lombardi G (2013) CD73 expression on extracellular vesicles derived from CD4+ CD25+ Foxp3+ T cells contributes to their regulatory function. Eur J Immunol 43:2430–2440CrossRefPubMed Smyth LA, Ratnasothy K, Tsang JYS, Boardman D, Warley A, Lechler R, Lombardi G (2013) CD73 expression on extracellular vesicles derived from CD4+ CD25+ Foxp3+ T cells contributes to their regulatory function. Eur J Immunol 43:2430–2440CrossRefPubMed
39.
Zurück zum Zitat Abbott RK, Thayer M, Labuda J, Silva M, Philbrook P, Cain DW, Kojima H, Hatfield S, Sethumadhavan S, Ohta A, Reinherz EL, Kelsoe G, Sitkovsky M (2016) Germinal center hypoxia potentiates immunoglobulin class switch recombination. J Immunol 197:4014–4020CrossRefPubMedPubMedCentral Abbott RK, Thayer M, Labuda J, Silva M, Philbrook P, Cain DW, Kojima H, Hatfield S, Sethumadhavan S, Ohta A, Reinherz EL, Kelsoe G, Sitkovsky M (2016) Germinal center hypoxia potentiates immunoglobulin class switch recombination. J Immunol 197:4014–4020CrossRefPubMedPubMedCentral
40.
Zurück zum Zitat Ohta A, Diwanji R, Kini R, Subramanian M, Ohta A, Sitkovsky M (2011) In vivo T cell activation in lymphoid tissues is inhibited in the oxygen-poor microenvironment. Front Immunol 2:27CrossRefPubMedPubMedCentral Ohta A, Diwanji R, Kini R, Subramanian M, Ohta A, Sitkovsky M (2011) In vivo T cell activation in lymphoid tissues is inhibited in the oxygen-poor microenvironment. Front Immunol 2:27CrossRefPubMedPubMedCentral
41.
Zurück zum Zitat Martin C, Leone M, Viviand X, Ayem ML, Guieu R (2000) High adenosine plasma concentration as a prognostic index for outcome in patients with septic shock. Crit Care Med 28:3198–3202CrossRefPubMed Martin C, Leone M, Viviand X, Ayem ML, Guieu R (2000) High adenosine plasma concentration as a prognostic index for outcome in patients with septic shock. Crit Care Med 28:3198–3202CrossRefPubMed
42.
Zurück zum Zitat Funaya H, Kitakaze M, Node K, Minamino T, Komamura K, Hori M (1997) Plasma adenosine levels increase in patients with chronic heart failure. Circulation 95:1363–1365CrossRefPubMed Funaya H, Kitakaze M, Node K, Minamino T, Komamura K, Hori M (1997) Plasma adenosine levels increase in patients with chronic heart failure. Circulation 95:1363–1365CrossRefPubMed
43.
Zurück zum Zitat Vallon V, Miracle C, Thomson S (2008) Adenosine and kidney function: potential implications in patients with heart failure. Eur J Heart Fail 10:176–187CrossRefPubMedPubMedCentral Vallon V, Miracle C, Thomson S (2008) Adenosine and kidney function: potential implications in patients with heart failure. Eur J Heart Fail 10:176–187CrossRefPubMedPubMedCentral
44.
Zurück zum Zitat Ramakers BPC, Riksen NP, Broek PHH, Franke B, Peters WHM, Hoeven JG, Smits P, Pickkers P (2011) Circulating adenosine increases during human experimental endotoxemia but blockade of its receptor does not influence the immune response and subsequent organ injury. Crit Care 15:R3CrossRefPubMedPubMedCentral Ramakers BPC, Riksen NP, Broek PHH, Franke B, Peters WHM, Hoeven JG, Smits P, Pickkers P (2011) Circulating adenosine increases during human experimental endotoxemia but blockade of its receptor does not influence the immune response and subsequent organ injury. Crit Care 15:R3CrossRefPubMedPubMedCentral
45.
Zurück zum Zitat Angelin A, Gil-de-Gómez L, Dahiya S, Jiao J, Guo L, Levine MH, Wang Z, Quinn WJ, Kopinski PK, Wang L, Akimova T, Liu Y, Bhatti TR, Han R, Laskin BL, Baur JA, Blair IA, Wallace DC, Hancock WW, Beier UH (2017) Foxp3 reprograms T cell metabolism to function in low-glucose, high-lactate environments. Cell Metab 25:1282–1293.e7CrossRefPubMedPubMedCentral Angelin A, Gil-de-Gómez L, Dahiya S, Jiao J, Guo L, Levine MH, Wang Z, Quinn WJ, Kopinski PK, Wang L, Akimova T, Liu Y, Bhatti TR, Han R, Laskin BL, Baur JA, Blair IA, Wallace DC, Hancock WW, Beier UH (2017) Foxp3 reprograms T cell metabolism to function in low-glucose, high-lactate environments. Cell Metab 25:1282–1293.e7CrossRefPubMedPubMedCentral
46.
Zurück zum Zitat Sitkovsky MV (2009) T regulatory cells: hypoxia-adenosinergic suppression and re-direction of the immune response. Trends Immunol 30:102–108CrossRefPubMed Sitkovsky MV (2009) T regulatory cells: hypoxia-adenosinergic suppression and re-direction of the immune response. Trends Immunol 30:102–108CrossRefPubMed
47.
Zurück zum Zitat Hatfield SM, Kjaergaard J, Lukashev D, Schreiber TH, Belikoff B, Abbott R, Sethumadhavan S, Philbrook P, Ko K, Cannici R, Thayer M, Rodig S, Kutok JL, Jackson EK, Karger B, Podack ER, Ohta A, Sitkovsky MV (2015) Immunological mechanisms of the antitumor effects of supplemental oxygenation. Sci Transl Med 7:277ra30CrossRefPubMedPubMedCentral Hatfield SM, Kjaergaard J, Lukashev D, Schreiber TH, Belikoff B, Abbott R, Sethumadhavan S, Philbrook P, Ko K, Cannici R, Thayer M, Rodig S, Kutok JL, Jackson EK, Karger B, Podack ER, Ohta A, Sitkovsky MV (2015) Immunological mechanisms of the antitumor effects of supplemental oxygenation. Sci Transl Med 7:277ra30CrossRefPubMedPubMedCentral
48.
49.
Zurück zum Zitat Beavis PA, Henderson MA, Giuffrida L, Mills JK, Sek K, Cross RS, Davenport AJ, John LB, Mardiana S, Slaney CY, Johnstone RW, Trapani JA, Stagg J, Loi S, Kats L, Gyorki D, Kershaw MH, Darcy PK (2017) Targeting the adenosine 2A receptor enhances chimeric antigen receptor T cell efficacy. J Clin Invest 127:929–941CrossRefPubMedPubMedCentral Beavis PA, Henderson MA, Giuffrida L, Mills JK, Sek K, Cross RS, Davenport AJ, John LB, Mardiana S, Slaney CY, Johnstone RW, Trapani JA, Stagg J, Loi S, Kats L, Gyorki D, Kershaw MH, Darcy PK (2017) Targeting the adenosine 2A receptor enhances chimeric antigen receptor T cell efficacy. J Clin Invest 127:929–941CrossRefPubMedPubMedCentral
50.
Zurück zum Zitat Sitkovsky MV, Ohta A (2005) The ‘danger’ sensors that STOP the immune response: the A2 adenosine receptors? Trends Immunol 26:299–304CrossRefPubMed Sitkovsky MV, Ohta A (2005) The ‘danger’ sensors that STOP the immune response: the A2 adenosine receptors? Trends Immunol 26:299–304CrossRefPubMed
Metadaten
Titel
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
verfasst von
Robert D. Leone
Im-Meng Sun
Min-Hee Oh
Im-Hong Sun
Jiayu Wen
Judson Englert
Jonathan D. Powell
Publikationsdatum
19.06.2018
Verlag
Springer Berlin Heidelberg
Erschienen in
Cancer Immunology, Immunotherapy / Ausgabe 8/2018
Print ISSN: 0340-7004
Elektronische ISSN: 1432-0851
DOI
https://doi.org/10.1007/s00262-018-2186-0

Weitere Artikel der Ausgabe 8/2018

Cancer Immunology, Immunotherapy 8/2018 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.