1932

Abstract

Cancer immunotherapy has revolutionized the way that we think about treating cancer. Although checkpoint blockade therapy, including anti-PD-1/PD-L1 and anti-CTLA-4, has shown remarkable success, the responses are limited to only a subset of patients. This discrepancy highlights the many overlapping avenues for immune evasion or suppression that can be employed by a tumor. One such mechanism of immunosuppression is adenosinergic signaling within the tumor microenvironment. We provide an overview of the current status of clinical trials targeting the adenosine pathway, including CD73, CD39, and adenosine receptors. Additionally, we highlight several avenues that may be explored to further potentiate responses in the clinic by combining adenosine-targeting agents to target multiple arms of the pathway or by using conventional immunotherapy agents.

Loading

Article metrics loading...

/content/journals/10.1146/annurev-med-060619-023155
2021-01-27
2024-04-19
Loading full text...

Full text loading...

/deliver/fulltext/med/72/1/annurev-med-060619-023155.html?itemId=/content/journals/10.1146/annurev-med-060619-023155&mimeType=html&fmt=ahah

Literature Cited

  1. 1. 
    Di Virgilio F, Sarti AC, Falzoni S et al. 2018. Extracellular ATP and P2 purinergic signalling in the tumour microenvironment. Nat. Rev. Cancer 18:601–18
    [Google Scholar]
  2. 2. 
    Vigano S, Alatzoglou D, Irving M et al. 2019. Targeting adenosine in cancer immunotherapy to enhance T-cell function. Front. Immunol. 10:925
    [Google Scholar]
  3. 3. 
    Jacobson KA, Gao ZG. 2006. Adenosine receptors as therapeutic targets. Nat. Rev. Drug Discov. 5:247–64
    [Google Scholar]
  4. 4. 
    Ohta A, Sitkovsky M. 2001. Role of G-protein-coupled adenosine receptors in downregulation of inflammation and protection from tissue damage. Nature 414:916–20
    [Google Scholar]
  5. 5. 
    Borsellino G, Kleinewietfeld M, Di Mitri D et al. 2007. Expression of ectonucleotidase CD39 by Foxp3+ Treg cells: hydrolysis of extracellular ATP and immune suppression. Blood 110:1225–32
    [Google Scholar]
  6. 6. 
    Mizumoto N, Kumamoto T, Robson SC et al. 2002. CD39 is the dominant Langerhans cell-associated ecto-NTPDase: modulatory roles in inflammation and immune responsiveness. Nat. Med. 8:358–65
    [Google Scholar]
  7. 7. 
    Koziak K, Sevigny J, Robson SC et al. 1999. Analysis of CD39/ATP diphosphohydrolase (ATPDase) expression in endothelial cells, platelets and leukocytes. Thromb. Haemost. 82:1538–44
    [Google Scholar]
  8. 8. 
    Jiang T, Xu X, Qiao M et al. 2018. Comprehensive evaluation of NT5E/CD73 expression and its prognostic significance in distinct types of cancers. BMC Cancer 18:267
    [Google Scholar]
  9. 9. 
    Leone RD, Emens LA. 2018. Targeting adenosine for cancer immunotherapy. J. Immunother. Cancer 6:57
    [Google Scholar]
  10. 10. 
    Horenstein AL, Chillemi A, Zaccarello G et al. 2013. A CD38/CD203a/CD73 ectoenzymatic pathway independent of CD39 drives a novel adenosinergic loop in human T lymphocytes. Oncoimmunology 2:e26246
    [Google Scholar]
  11. 11. 
    Zimmermann H. 2009. Prostatic acid phosphatase, a neglected ectonucleotidase. Purinerg. Signal. 5:273–75
    [Google Scholar]
  12. 12. 
    Zimmermann H, Zebisch M, Sträter N 2012. Cellular function and molecular structure of ecto-nucleotidases. Purinerg. Signal. 8:437–502
    [Google Scholar]
  13. 13. 
    Chiappori A, Williams CC, Creelan BC et al. 2018. Phase I/II study of the A2AR antagonist NIR178 (PBF-509), an oral immunotherapy, in patients (pts) with advanced NSCLC. J. Clin. Oncol. 36:9089
    [Google Scholar]
  14. 14. 
    Bendell J, Bauer T, Patel M et al. 2019. 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 79:CT026
    [Google Scholar]
  15. 15. 
    Powderly JD, de Souza PL, Gutierrez R et al. 2019. AB928, a novel dual adenosine receptor antagonist, combined with chemotherapy or AB122 (anti-PD-1) in patients (pts) with advanced tumors: preliminary results from ongoing phase I studies. J. Clin. Oncol. 37:2604
    [Google Scholar]
  16. 16. 
    Fong L, Powderly J II, Luke J et al. 2018. Refractory renal cell cancer (RCC) exhibits high adenosine A2A receptor (A2AR) expression and prolonged survival following treatment with A2AR antagonist CPI-444 Paper presented at Society for Immunotherapy of Cancer (SITC) 33rd Annual Meeting Nov 7–11 Washington, DC:
  17. 17. 
    Willingham SB. 2017. Inhibition of A2AR induces anti-tumor immunity alone and in combination with anti-PD-L1 in preclinical and clinical studies Paper presented at American Association for Cancer Research (AACR) 2017 Annual Meeting Dec. 5–9 San Antonio, TX:
  18. 18. 
    Emens L, Powderly J, Fong L et al. 2017. Abstract CT119: CPI-444, an oral adenosine A2a receptor (A2AR) antagonist, demonstrates clinical activity in patients with advanced solid tumors. Cancer Res 77:CT119
    [Google Scholar]
  19. 19. 
    Fong LCM, George S, Hughes B et al. 2020. Adenosine receptor blockade with ciforadenant + atezolizumab in advanced metastatic castration resistant prostate cancer (mCRPC). J. Clin. Oncol. 38:129
    [Google Scholar]
  20. 20. 
    Geoghegan JC, Diedrich G, Lu X et al. 2016. Inhibition of CD73 AMP hydrolysis by a therapeutic antibody with a dual, non-competitive mechanism of action. MAbs 8:454–67
    [Google Scholar]
  21. 21. 
    Hay CM, Sult E, Huang Q et al. 2016. Targeting CD73 in the tumor microenvironment with MEDI9447. Oncoimmunology 5:e1208875
    [Google Scholar]
  22. 22. 
    Overman MJ, LoRusso P, Strickler JH et al. 2018. 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. 36:4123
    [Google Scholar]
  23. 23. 
    Siu LL, Burris H, Le DT et al. 2018. 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 78:CT180
    [Google Scholar]
  24. 24. 
    Luke JJ, Powderly JD, Merchan JR et al. 2019. Immunobiology, preliminary safety, and efficacy of CPI-006, an anti-CD73 antibody with immune modulating activity, in a phase 1 trial in advanced cancers. J. Clin. Oncol. 37:2505
    [Google Scholar]
  25. 25. 
    Ohta A, Gorelik E, Prasad SJ et al. 2006. A2A adenosine receptor protects tumors from antitumor T cells. PNAS 103:13132–37
    [Google Scholar]
  26. 26. 
    Leone RD, Sun IM, Oh MH et al. 2018. 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. 67:1271–84
    [Google Scholar]
  27. 27. 
    Mittal D, Young A, Stannard K et al. 2014. Antimetastatic effects of blocking PD-1 and the adenosine A2A receptor. Cancer Res 74:3652–58
    [Google Scholar]
  28. 28. 
    Beavis PA, Divisekera U, Paget C et al. 2013. Blockade of A2A receptors potently suppresses the metastasis of CD73+ tumors. PNAS 110:14711–16
    [Google Scholar]
  29. 29. 
    Waickman AT, Alme A, Senaldi L et al. 2012. Enhancement of tumor immunotherapy by deletion of the A2A adenosine receptor. Cancer Immunol. Immunother. 61:917–26
    [Google Scholar]
  30. 30. 
    Fong L, Hotson A, Powderly JD et al. 2019. Adenosine 2A receptor blockade as an immunotherapy for treatment-refractory renal cell cancer. Cancer Discov 10:40–53
    [Google Scholar]
  31. 31. 
    Sitkovsky MV. 2020. Lessons from the A2A adenosine receptor antagonist-enabled tumor regression and survival in patients with treatment-refractory renal cell cancer. Cancer Discov 10:16–19
    [Google Scholar]
  32. 32. 
    Seitz L, Jin L, Leleti M et al. 2018. Safety, tolerability, and pharmacology of AB928, a novel dual adenosine receptor antagonist, in a randomized, phase 1 study in healthy volunteers. Investig. New Drug. 37:711–21
    [Google Scholar]
  33. 33. 
    Morandi F, Horenstein AL, Costa F et al. 2018. CD38: a target for immunotherapeutic approaches in multiple myeloma. Front. Immunol. 9:2722
    [Google Scholar]
  34. 34. 
    Feng X, Zhang L, Acharya C et al. 2017. Targeting CD38 suppresses induction and function of T regulatory cells to mitigate immunosuppression in multiple myeloma. Clin. Cancer Res. 23:4290–300
    [Google Scholar]
  35. 35. 
    Hausler SF, Del Barrio IM, Diessner J et al. 2014. Anti-CD39 and anti-CD73 antibodies A1 and 7G2 improve targeted therapy in ovarian cancer by blocking adenosine-dependent immune evasion. Am. J. Transl. Res. 6:129–39
    [Google Scholar]
  36. 36. 
    Hausler SF, Montalban del Barrio I, Strohschein J et al. 2011. Ectonucleotidases CD39 and CD73 on OvCA cells are potent adenosine-generating enzymes responsible for adenosine receptor 2A-dependent suppression of T cell function and NK cell cytotoxicity. Cancer Immunol. Immunother. 60:1405–18
    [Google Scholar]
  37. 37. 
    Saif MW, Alexander D, Wicox CM 2005. Serum alkaline phosphatase level as a prognostic tool in colorectal cancer: a study of 105 patients. J. Appl. Res. 5:88–95
    [Google Scholar]
  38. 38. 
    Ren HY, Sun LL, Li HY, Ye ZM 2015. Prognostic significance of serum alkaline phosphatase level in osteosarcoma: a meta-analysis of published data. Biomed. Res. Int. 2015:160835
    [Google Scholar]
  39. 39. 
    Rao SR, Snaith AE, Marino D et al. 2017. Tumour-derived alkaline phosphatase regulates tumour growth, epithelial plasticity and disease-free survival in metastatic prostate cancer. Br. J. Cancer 116:227–36
    [Google Scholar]
  40. 40. 
    Graddis TJ, McMahan CJ, Tamman J et al. 2011. Prostatic acid phosphatase expression in human tissues. Int. J. Clin. Exp. Pathol. 4:295–306
    [Google Scholar]
  41. 41. 
    Torres Á, Erices JI, Sanchez F et al. 2019. Extracellular adenosine promotes cell migration/invasion of glioblastoma stem-like cells through A3 adenosine receptor activation under hypoxia. Cancer Lett 446:112–22
    [Google Scholar]
  42. 42. 
    Liu T-Z, Wang X, Bai Y-F et al. 2014. The HIF-2alpha dependent induction of PAP and adenosine synthesis regulates glioblastoma stem cell function through the A2B adenosine receptor. Int. J. Biochem. Cell Biol. 49:8–16
    [Google Scholar]
  43. 43. 
    Spychala J. 2000. Tumor-promoting functions of adenosine. Pharmacol. Ther. 87:161–73
    [Google Scholar]
  44. 44. 
    Young A, Ngiow SF, Barkauskas DS et al. 2016. Co-inhibition of CD73 and A2AR adenosine signaling improves anti-tumor immune responses. Cancer Cell 30:391–403
    [Google Scholar]
  45. 45. 
    Sitkovsky M, Lukashev D, Deaglio S et al. 2008. Adenosine A2A receptor antagonists: blockade of adenosinergic effects and T regulatory cells. Br. J. Pharmacol. 153:Suppl. 1S457–64
    [Google Scholar]
  46. 46. 
    Allard D, Turcotte M, Stagg J 2017. Targeting A2 adenosine receptors in cancer. Immunol. Cell Biol. 95:333–39
    [Google Scholar]
  47. 47. 
    Leone RD, Lo YC, Powell JD 2015. A2aR antagonists: next generation checkpoint blockade for cancer immunotherapy. Comput. Struct. Biotechnol. J. 13:265–72
    [Google Scholar]
  48. 48. 
    Addi AB, Lefort A, Hua X et al. 2008. Modulation of murine dendritic cell function by adenine nucleotides and adenosine: involvement of the A2B receptor. Eur. J. Immunol. 38:1610–20
    [Google Scholar]
  49. 49. 
    Wilson JM, Ross WG, Agbai ON et al. 2009. The A2B adenosine receptor impairs the maturation and immunogenicity of dendritic cells. J. Immunol. 182:4616–23
    [Google Scholar]
  50. 50. 
    Chen S, Akdemir I, Fan J et al. 2020. The expression of adenosine A2B receptor on antigen-presenting cells suppresses CD8+ T-cell responses and promotes tumor growth. Cancer Immunol. Res. 8:81064–74
    [Google Scholar]
  51. 51. 
    Sciaraffia E, Riccomi A, Lindstedt R et al. 2014. Human monocytes respond to extracellular cAMP through A2A and A2B adenosine receptors. J. Leukoc. Biol. 96:113–22
    [Google Scholar]
  52. 52. 
    Novitskiy SV, Ryzhov S, Zaynagetdinov R et al. 2008. Adenosine receptors in regulation of dendritic cell differentiation and function. Blood 112:1822–31
    [Google Scholar]
  53. 53. 
    Nemeth ZH, Lutz CS, Csoka B et al. 2005. Adenosine augments IL-10 production by macrophages through an A2B receptor-mediated posttranscriptional mechanism. J. Immunol. 175:8260–70
    [Google Scholar]
  54. 54. 
    Gabrilovich DI, Bronte V, Chen SH et al. 2007. The terminology issue for myeloid-derived suppressor cells. Cancer Res 67:425
    [Google Scholar]
  55. 55. 
    Talmadge JE, Gabrilovich DI. 2013. History of myeloid-derived suppressor cells. Nat. Rev. Cancer 13:739–52
    [Google Scholar]
  56. 56. 
    Ryzhov S, Novitskiy SV, Goldstein AE et al. 2011. Adenosinergic regulation of the expansion and immunosuppressive activity of CD11b + Gr1 + cells. J. Immunol. 187:6120–29
    [Google Scholar]
  57. 57. 
    Sorrentino C, Miele L, Porta A et al. 2015. Myeloid-derived suppressor cells contribute to A2B adenosine receptor-induced VEGF production and angiogenesis in a mouse melanoma model. Oncotarget 6:27478–89
    [Google Scholar]
  58. 58. 
    Yu M, Guo G, Huang L et al. 2020. CD73 on cancer-associated fibroblasts enhanced by the A2B-mediated feedforward circuit enforces an immune checkpoint. Nat. Commun. 11:515
    [Google Scholar]
  59. 59. 
    Topalian SL, Drake CG, Pardoll DM 2015. Immune checkpoint blockade: a common denominator approach to cancer therapy. Cancer Cell 27:450–61
    [Google Scholar]
  60. 60. 
    Zappasodi R, Merghoub T, Wolchok JD 2018. Emerging concepts for immune checkpoint blockade-based combination therapies. Cancer Cell 33:581–98
    [Google Scholar]
  61. 61. 
    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. 19:5626–35
    [Google Scholar]
  62. 62. 
    Beavis PA, Milenkovski N, Henderson MA et al. 2015. Adenosine receptor 2A blockade increases the efficacy of anti-PD-1 through enhanced antitumor T-cell responses. Cancer Immunol. Res. 3:506–17
    [Google Scholar]
  63. 63. 
    Mittal D, Sinha D, Barkauskas D et al. 2016. Adenosine 2B receptor expression on cancer cells promotes metastasis. Cancer Res 76:4372–82
    [Google Scholar]
  64. 64. 
    Beavis PA, Henderson MA, Giuffrida L et al. 2017. Targeting the adenosine 2A receptor enhances chimeric antigen receptor T cell efficacy. J. Clin. Investig. 127:929–41
    [Google Scholar]
  65. 65. 
    Jin D, Fan J, Wang L et al. 2010. CD73 on tumor cells impairs antitumor T-cell responses: a novel mechanism of tumor-induced immune suppression. Cancer Res 70:2245–55
    [Google Scholar]
  66. 66. 
    Wang L, Fan J, Thompson LF et al. 2011. CD73 has distinct roles in nonhematopoietic and hematopoietic cells to promote tumor growth in mice. J. Clin. Investig. 121:2371–82
    [Google Scholar]
  67. 67. 
    Cekic C, Linden J. 2014. Adenosine A2A receptors intrinsically regulate CD8+ T cells in the tumor microenvironment. Cancer Res 74:7239–49
    [Google Scholar]
  68. 68. 
    Kjaergaard J, Hatfield S, Jones G et al. 2018. A2A adenosine receptor gene deletion or synthetic A2A antagonist liberate tumor-reactive CD8+ T cells from tumor-induced immunosuppression. J. Immunol. 201:782–91
    [Google Scholar]
  69. 69. 
    Thompson EA, Ols S, Miura K et al. 2018. TLR-adjuvanted nanoparticle vaccines differentially influence the quality and longevity of responses to malaria antigen Pfs25. JCI Insight 3:e120692
    [Google Scholar]
  70. 70. 
    Lynn GM, Sedlik C, Baharom F et al. 2020. Peptide-TLR-7/8a conjugate vaccines chemically programmed for nanoparticle self-assembly enhance CD8 T-cell immunity to tumor antigens. Nat. Biotechnol. 38:320–32
    [Google Scholar]
  71. 71. 
    Moyer TJ, Zmolek AC, Irvine DJ 2016. Beyond antigens and adjuvants: formulating future vaccines. J. Clin. Investig. 126:799–808
    [Google Scholar]
  72. 72. 
    Dahan R, Ravetch JV. 2016. Co-targeting of adenosine signaling pathways for immunotherapy: potentiation by Fc receptor engagement. Cancer Cell 30:369–71
    [Google Scholar]
  73. 73. 
    Dahan R, Barnhart BC, Li F et al. 2016. Therapeutic activity of agonistic, human anti-CD40 monoclonal antibodies requires selective FcγR engagement. Cancer Cell 29:820–31
    [Google Scholar]
  74. 74. 
    Graziano RF, Engelhardt JJ. 2019. Role of FcγRs in antibody-based cancer therapy. Curr. Top. Microbiol. Immunol 423:13–34
    [Google Scholar]
  75. 75. 
    Moser GH, Schrader J, Deussen A 1989. Turnover of adenosine in plasma of human and dog blood. Am. J. Physiol. 256:C799–806
    [Google Scholar]
  76. 76. 
    DiRenzo D, Ashok D, Anderson AE et al. 2019. Abstract 3168: Methods for assessment of the “adenosine fingerprint” in clinical trials of AB928. Cancer Res 79:3168
    [Google Scholar]
/content/journals/10.1146/annurev-med-060619-023155
Loading
/content/journals/10.1146/annurev-med-060619-023155
Loading

Data & Media loading...

  • Article Type: Review Article
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error