Increased expression of adenosine 2A receptors in metastatic renal cell carcinoma is associated with poorer response to anti-vascular endothelial growth factor agents and anti-PD-1/Anti-CTLA4 antibodies and shorter survival

Takao Kamai, Toshiki Kijima, Toyonori Tsuzuki, Akinori Nukui, Hideyuki Abe, Kyoko Arai, Ken-Ichiro Yoshida, Takao Kamai, Toshiki Kijima, Toyonori Tsuzuki, Akinori Nukui, Hideyuki Abe, Kyoko Arai, Ken-Ichiro Yoshida

Abstract

Background: Adenosine and its adenosine 2A receptors (A2AR) mediate the immunosuppressive mechanism by which tumors escape immunosurveillance and impede anti-tumor immunity within the tumor microenvironment. However, we do not know whether the adenosine pathway (CD39/CD73/A2AR) plays a role in renal cell carcinoma (RCC). Therefore, we studied the role of immunosuppression in RCC by assessing the adenosine pathway in patients with RCC treated with anti-vascular endothelial growth factor (anti-VEGF) agents or immune checkpoints inhibitors (ICIs) or both.

Methods: In 60 patients with metastatic RCC, we examined the expression of CD39, CD73, A2AR, and programmed cell death 1 ligand 1 (PD-L1) immunohistochemically in surgically resected tumor tissues and studied the clinicopathological characteristics of these patients. Patients were treated by cytoreductive nephrectomy with systemic therapy with anti-VEGF agent or a combination of the ICIs anti-cytotoxic T-lymphocyte-associated antigen 4 (CTLA4) antibody and programmed cell death 1 (PD-1) antibody.

Results: Increased expression of A2AR in the primary tumors was associated with metastatic profiles. Patients treated with anti-PD-1 antibody in monotherapy, a combination of anti-PD-1 and anti-CTLA4 antibodies, or anti-VEGF agents showed better response and longer overall survival if the primary tumor had higher PD-L1 expression and lower A2AR expression. In Cox multivariate regression analysis, higher expression of A2AR was associated with shorter overall survival.

Conclusions: Our findings suggest that the expression of A2AR and PD-L1 in the primary tumors in RCC might predict the outcomes of treatment with anti-VEGF agents and ICIs and that the A2AR pathway might be a molecular target for immunotherapy.

Keywords: Adenosine 2A receptor (A2AR); CD39; CD73; PD-L1; Renal cell carcinoma.

Conflict of interest statement

The authors declare that they have no conflict of interest.

Figures

Fig. 1
Fig. 1
Immunohistochemistry of a case of better response to anti-VEGF therapies and longer survival. 73 y.o. male of left clear cell renal cell carcinoma with Fuhrman grade 2, pT2bN0M1 (PUL, LYM). CD8; high expression, PD-L1; low expression, CD39: high expression, CD73; low expression, A2AR; low expression. After cytoreductive nephrectomy, this patient had received sunitinib as first-line anti-VEGF therapy for 17 months with best response of partial response. After refractory to sunitinib, he has received axitinib as second-line anti-VEGF therapy with alive with disease for 29 months
Fig. 2
Fig. 2
Immunohistochemistry of a case of worse response to first-line anti-VEGF therapy and second-line nivolumab and shorter survival. 64 year old male of right clear cell renal cell carcinoma with Fuhrman grade 3, pT2aN0M1 (PUL, HEP). CD8; high expression, PD-L1; high expression, CD39: high expression, CD73; high expression, A2AR; high expression. After cytoreductive nephrectomy, this patient had received pazopanib as first-line anti-VEGF therapy for 3 months, while the metastatic disease progressed. Then the patient received nivolumab as second-line therapy for 3 months. The best response was partial response for pulmonary and liver lesions, however, the lesions progressed rapidly and the patient dead
Fig. 3
Fig. 3
Immunohistochemistry of a case of worse response to first-line combination of ipilimumab and nivolumab and shorter survival. 57 year old male of right collecting duct renal cell carcinoma with Fuhrman grade 3, pT2aN0M1 (PUL, HEP). CD8; low expression, PD-L1; low expression, CD39: high expression, CD73; high expression, A2AR; high expression. After cytoreductive nephrectomy, the patient had received combination of ipilimumab and nivolumab as first-line therapy. He showed no response to the therapy and dead after 4 months
Fig. 4
Fig. 4
Immunohistochemistry of clear cell RCC with muscle and pulmonary metastases. 71 y.o. male of left clear cell renal cell carcinoma with Fuhrman grade 3, pT1aN0M1 (PUL, OTH). Primary renal tumor showed high expression for A2AR and negative staining for PD-L1. Metastatic tumor in psoas major muscle showed high reaction for A2AR and negative reaction for PD-L1. After cytoreductive nephrectomy and resection of metastatic tumor in psoas major muscle, this patient had received sunitinib as first-line anti-VEGF therapy for 5 months, but pulmonary disease progressed slowly. Then the patient has received nivolumab as second-line therapy. He showed no response to the therapy and dead after 21 months
Fig. 5
Fig. 5
Overall survival curve. This overall survival curve is based on the expression status of PD-L1 (a), CD39 (b), CD73 (c), A2AR (d), combination of PD-L1 and A2AR in all patients (e) and in the patients treated with immune checkpoints inhibitors (ICIs) (f). In Figure E, in comparison to the patients in type I (PD-L1 low and A2AR high), the relative risk (RR) of the patients in type IV (PD-L1 high and A2AR low) was 0.107 (95% confidential intervals (CI); 0.030–0.380, P = 0.0005), RR in type III (PD-L1 low and A2AR low) was 0.134 (95%CI; 0.035–0.515, P = 0.0034), and RR in type II (PD-L1 high and A2AR high) was 0.468 (95%CI; 0.153–1.427, P = 0.1820). In Figure F, compared to the patients in low–high type-I, the relative risk (RR) of the patients in high-low type-IV was 0.060 (95% confidential intervals (CI); 0.006–0.628, P = 0.0108), RR in low-low type-III was 0.143 (95%CI; 0.014–1.512, P = 0.1060), and RR in high-high type-II was 0.595 (95%CI; 0.151–2.351, p = 0.4589)

References

    1. Bergers G, Hanahan D. Modes of resistance to anti-angiogenic therapy. Nat Rev Cancer. 2008;8:592–603. doi: 10.1038/nrc2442.
    1. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144:646–674. doi: 10.1016/j.cell.2011.02.013.
    1. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012;12:252–264. doi: 10.1038/nrc3239.
    1. Topalian SL, Drake CG, Pardoll DM. Immune checkpoint blockade: a common denominator approach to cancer therapy. Cancer Cell. 2015;27:450–461. doi: 10.1016/j.ccell.2015.03.001.
    1. Motzer RJ, Escudier B, McDermott DF, George S, Hammers HJ, Srinivas S, Tykodi SS, Sosman JA, Procopio G, Plimack ER, Castellano D, Choueiri TK, Gurney H, Donskov F, Bono P, Wagstaff J, Gauler TC, Ueda T, Tomita Y, Schutz FA, Kollmannsberger C, Larkin J, Ravaud A, Simon JS, Xu LA, Waxman IM, Sharma P; CheckMate 025 Investigators Nivolumab versus everolimus in advanced renal-cell carcinoma. N Engl J Med. 2015;373:1803–1813. doi: 10.1056/NEJMoa1510665.
    1. Motzer RJ, Rini BI, McDermott DF, Arén Frontera O, Hammers HJ, Carducci MA, Salman P, Escudier B, Beuselinck B, Amin A, Porta C, George S, Neiman V, Bracarda S, Tykodi SS, Barthélémy P, Leibowitz-Amit R, Plimack ER, Oosting SF, Redman B, Melichar B, Powles T, Nathan P, Oudard S, Pook D, Choueiri TK, Donskov F, Grimm MO, Gurney H, Heng DYC, Kollmannsberger CK, Harrison MR, Tomita Y, Duran I, Grünwald V, McHenry MB, Mekan S, Tannir NM; CheckMate 214 investigators Nivolumab plus ipilimumab versus sunitinib in first-line treatment for advanced renal cell carcinoma: extended follow-up of efficacy and safety results from a randomised, controlled, phase 3 trial. Lancet Oncol. 2019;20:1370–1385. doi: 10.1016/S1470-2045(19)30413-9.
    1. 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. A2A adenosine receptor protects tumors from antitumor T cells. Proc Natl Acad Sci U S A. 2006;103:13132–13137. doi: 10.1073/pnas.0605251103.
    1. 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.
    1. Young A, Mittal D, Stagg SMJ. Targeting cancer-derived adenosine: new therapeutic approaches. Cancer Discov. 2014;4:879–880. doi: 10.1158/-14-0341.
    1. Allard B, Beavis PA, Darcy PK, Stagg L. Immunosuppressive activities of adenosine in cancer. Curr Opin Pharmacol. 2016;29:7–16. doi: 10.1016/j.coph.2016.04.001.
    1. Vijayan D, Young A, Teng MWL, Smyth MJ. Targeting immunosuppressive adenosine in cancer. Nat Rev Cancer. 2017;7:709–724. doi: 10.1038/nrc.2017.86.
    1. Idzko M, Ferrari D, Eltzschig HK. Nucleotide signalling during inflammation. Nature. 2014;509:310–317. doi: 10.1038/nature13085.
    1. Junger WG. Immune cell regulation by autocrine purinergic signaling. Nat Rev Immunol. 2011;11:201–212. doi: 10.1038/nri2938.
    1. Antonioli L, Blandizzi C, Pacher P, Haskó G. Immunity, inflammation and cancer: a leading role for adenosine. Nat Rev Cancer. 2013;13:842–857. doi: 10.1038/nrc3613.
    1. 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;19:5626–5635. doi: 10.1158/1078-0432.CCR-13-0545.
    1. Beavis PA, Milenkovski N, Henderson MA, John LB, Allard B, Loi S, Kershaw MH, Stagg J, Darcy PK. Adenosine receptor 2A blockade increases the efficacy of anti-PD-1 through enhanced antitumor T-cell responses. Cancer Immunol Res. 2015;3:506–517. doi: 10.1158/2326-6066.CIR-14-0211.
    1. Yu Y, Wang W, Song L, Hu W, Dong C, Pei H, Zhou G, Yue Z. Ecto-5′-nucleotidase expression is associated with the progression of renal cell carcinoma. Oncol Lett. 2015;9:2485–2494. doi: 10.3892/ol.2015.3138.
    1. Fong L, Hotson A, Powderly JD, Sznol M, Heist RS, Choueiri TK, George S, Hughes BGM, Hellmann MD, Shepard DR, Rini BI, Kummar S, Weise AM, Riese MJ, Markman B, Emens LA, Mahadevan D, Luke JJ, Laport G, Brody JD, Hernandez-Aya L, Bonomi P, Goldman JW, Berim L, Renouf DJ, Goodwin RA, Munneke B, Ho PY, Hsieh J, McCaffery I, Kwei L, Willingham SB, Miller RA. Adenosine 2A receptor blockade as an immunotherapy for treatment-refractory renal cell cancer. Cancer Discov. 2020;10:40–53. doi: 10.1158/-19-0980.
    1. Nukui A, Kamai T, Arai K, Kijima T, Kobayashi M, Narimatsu T, Kambara T, Yuki H, Betsunoh H, Abe H, Fukabori Y, Yashi M, Yoshida KI. Association of cancer progression with elevated expression of programmed cell death protein 1 ligand 1 by upper tract urothelial carcinoma and increased tumor-infiltrating lymphocyte density. Cancer Immunol Immunother. 2020;69:689–702. doi: 10.1007/s00262-020-02499-7.
    1. Rittmeyer A, Barlesi F, Waterkamp D, Park K, Ciardiello F, von Pawel J, Gadgeel SM, Hida T, Kowalski DM, Dols MC, Cortinovis DL, Leach J, Polikoff J, Barrios C, Kabbinavar F, Frontera OA, De Marinis F, Turna H, Lee JS, Ballinger M, Kowanetz M, He P, Chen DS, Sandler A, Gandara DR; OAK Study Group Atezolizumab versus docetaxel in patients with previously treated non-small-cell lung cancer (OAK): a phase 3, open-label, multicentre randomised controlled trial. Lancet. 2017;389:255–265. doi: 10.1016/S0140-6736(16)32517-X.
    1. Gerlinger M, Rowan AJ, Horswell S, Larkin J, Endesfelder D, Gronroos E, Martinez P, Matthews N, Stewart A, Tarpey P, Varela I, Phillimore B, Begum S, McDonald NQ, Butler A, Jones D, Raine K, Latimer C, Santos CR, Nohadani M, Eklund AC, Spencer-Dene B, Clark G, Pickering L, Stamp G, Gore M, Szallasi Z, Downward J, Futreal PA, Swanton C. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. New Engl J Med. 2012;366:883–892. doi: 10.1056/NEJMoa1113205.
    1. Motzer RJ, Penkov K, Haanen J, Rini B, Albiges L, Campbell MT, Venugopal B, Kollmannsberger C, Negrier S, Uemura M, Lee JL, Vasiliev A, Miller WH, Jr, Gurney H, Schmidinger M, Larkin J, Atkins MB, Bedke J, Alekseev B, Wang J, Mariani M, Robbins PB, Chudnovsky A, Fowst C, Hariharan S, Huang B, di Pietro A, Choueiri TK. Avelumab plus axitinib versus sunitinib for advanced renal-cell carcinoma. N Engl J Med. 2019;380:1103–1115. doi: 10.1056/NEJMoa1816047.
    1. Rini BI, Plimack ER, Stus V, Gafanov R, Hawkins R, Nosov D, Pouliot F, Alekseev B, Soulières D, Melichar B, Vynnychenko I, Kryzhanivska A, Bondarenko I, Azevedo SJ, Borchiellini D, Szczylik C, Markus M, McDermott RS, Bedke J, Tartas S, Chang YH, Tamada S, Shou Q, Perini RF, Chen M, Atkins MB, Powles T; KEYNOTE-426 Investigators Pembrolizumab plus axitinib versus sunitinib for advanced renal-cell carcinoma. N Engl J Med. 2019;380:1116–1127. doi: 10.1056/NEJMoa1816714.
    1. Joyce JA, Fearon DT. T cell exclusion, immune privilege, and the tumor microenvironment. Science. 2015;348:74–80. doi: 10.1126/science.aaa6204.
    1. Sowell RT, Kaech SM. Probing the diversity of T cell dysfunction in cancer. Cell. 2016;166:1362–1364. doi: 10.1016/j.cell.2016.08.058.
    1. Sharma P, Hu-Lieskovan S, Wargo JA, Ribas A. Primary, adaptive, and acquired resistance to cancer immunotherapy. Cell. 2017;168:707–723. doi: 10.1016/j.cell.2017.01.017.
    1. Thommen DS, Schumacher TN. T cell dysfunction in cancer. Cancer Cell. 2018;33:547–562. doi: 10.1016/j.ccell.2018.03.012.
    1. Blank CU, Haining WN, Held W, Hogan PG, Kallies A, Lugli E, Lynn RC, Philip M, Rao A, Restifo NP, Schietinger A, Schumacher TN, Schwartzberg PL, Sharpe AH, Speiser DE, Wherry EJ, Youngblood BA, Zehn D. Defining T cell exhaustion. Nat Rev Immunol. 2019;19:665–674. doi: 10.1038/s41577-019-0221-9.
    1. Willingham SB, Ho PY, Hotson A, Hill C, Piccione EC, Hsieh J, Liu L, Buggy JJ, McCaffery I, Miller RA. 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;6:1136–1149. doi: 10.1158/2326-6066.CIR-18-0056.
    1. Deaglio S, Dwyer KM, Gao W, Friedman D, Usheva A, Erat A, Chen JF, Enjyoji K, Linden J, Oukka M, Kuchroo VK, Strom TB, Robson SC. Adenosine generation catalyzed by CD39 and CD73 expressed on regulatory T cells mediates immune suppression. J Exp Med. 2007;204:1257–1265. doi: 10.1084/jem.20062512.
    1. Voron T, Marcheteau E, Pernot S, Colussi O, Tartour E, Taieb J, Terme M. Control of the immune response by pro-angiogenic factors. Front Oncol. 2014;4:70. doi: 10.3389/fonc.2014.00070.
    1. Lanitis E, Irving M, Coukos G. Targeting the tumor vasculature to enhance T cell activity. Curr Opin Immunol. 2015;33:55–63. doi: 10.1016/j.coi.2015.01.011.
    1. Lapeyre-Prost A, Terme M, Pernot S, Pointet AL, Voron T, Tartour E, Taieb J. Immunomodulatory activity of VEGF in cancer. Int Rev Cell Mol Biol. 2017;330:295–342. doi: 10.1016/bs.ircmb.2016.09.007.
    1. Gabrilovich DI, Nagaraj S. Myeloid-derived suppressor cells as regulators of the immune system. Nature Rev Immunol. 2009;9:162–174. doi: 10.1038/nri2506.
    1. Diakos CI, Charles KA, McMillan DC, Clarke SJ. Cancer-related inflammation and treatment effectiveness. Lancet Oncol. 2014;15:e493–503. doi: 10.1016/S1470-2045(14)70263-3.
    1. Mantovani A, Allavena P, Sica A, Balkwill F. Cancer-related inflammation. Nature. 2008;454:436–444. doi: 10.1038/nature07205.

Source: PubMed

3
Tilaa