Targeting CD73 in the tumor microenvironment with MEDI9447

Carl M Hay, Erin Sult, Qihui Huang, Kathy Mulgrew, Stacy R Fuhrmann, Kelly A McGlinchey, Scott A Hammond, Raymond Rothstein, Jonathan Rios-Doria, Edmund Poon, Nick Holoweckyj, Nicholas M Durham, Ching Ching Leow, Gundo Diedrich, Melissa Damschroder, Ronald Herbst, Robert E Hollingsworth, Kris F Sachsenmeier, Carl M Hay, Erin Sult, Qihui Huang, Kathy Mulgrew, Stacy R Fuhrmann, Kelly A McGlinchey, Scott A Hammond, Raymond Rothstein, Jonathan Rios-Doria, Edmund Poon, Nick Holoweckyj, Nicholas M Durham, Ching Ching Leow, Gundo Diedrich, Melissa Damschroder, Ronald Herbst, Robert E Hollingsworth, Kris F Sachsenmeier

Abstract

MEDI9447 is a human monoclonal antibody that is specific for the ectoenzyme CD73 and currently undergoing Phase I clinical trials. Here we show that MEDI9447 is a potent inhibitor of CD73 ectonucleotidase activity, with wide ranging immune regulatory consequences. MEDI9447 results in relief from adenosine monophosphate (AMP)-mediated lymphocyte suppression in vitro and inhibition of mouse syngeneic tumor growth in vivo. In contrast with other cancer immunotherapy agents such as checkpoint inhibitors or T-cell agonists, MEDI9447 drives changes in both myeloid and lymphoid infiltrating leukocyte populations within the tumor microenvironment of mouse models. Changes include significant alterations in a number of tumor micro-environmental subpopulations including increases in CD8(+) effector cells and activated macrophages. Furthermore, these changes correlate directly with responder and non-responder subpopulations within animal studies using syngeneic tumors. Combination data showing additive activity between MEDI9447 and anti-PD-1 antibodies using human cells in vitro and mouse tumor models further demonstrate the potential value of relieving adenosine-mediated immunosuppression. Based on these data, a Phase I study to test the safety, tolerability, and clinical activity of MEDI9447 in cancer patients was initiated (NCT02503774).

Keywords: Adenosine; CD73; MEDI9447; monoclonal antibody; syngeneic tumor model; tumor microenvironment.

Figures

Figure 1.
Figure 1.
CD73-expressing human NCI-H322 (Panel A) or mouse 4T1 (Panel B) cells were incubated with AMP and MEDI9447 or an isotype control antibody at the indicated concentrations. Following incubation, ATP and CellTiter-Glo® were added and light emission inhibition was measured by luminometer. The mean signal of duplicate samples is plotted with error bars representing the standard deviation of the mean. Data are representative of five independent experiments.
Figure 2.
Figure 2.
Internalization of CD73 upon binding by MEDI9447 was demonstrated. MDA-MB-231 human breast carcinoma cells were incubated with MEDI9447 and a fluorescently labeled anti-human IgG secondary antibody. Localization of CD73 was imaged after 2 h at 4°C (Panel A) or 37°C and again after 4 (Panel C) and 20 (Panel D) h at 37°C. Internalization of CD73 upon MEDI9447 binding was also demonstrated by measuring MDA-MB-231 (Panel E) and mouse 4T1 (Panel F) breast cancer cell viability following MEDI9447 binding in the presence of a toxin-conjugated secondary antibody. The indicated concentrations of MEDI9447 were incubated with a saporin-conjugated anti-human IgG antibody for 3 d and viability was estimated by measuring ATP levels using the Cell Titer-Glo assay kit. Data are representative of five independent experiments.
Figure 3.
Figure 3.
CFSE-labeled CD4+ T cells were pre-activated with anti-CD3 and anti-CD28 antibody-coated microbeads and recombinant human IL-2 and then transferred into sterile round-bottomed tissue culture 96 well plates at approximately 50,000 cells per well. T cell proliferation and division was suppressed by the addition of 100-µM AMP. Addition of MEDI9447 and Phen0203 human IgG1 overcame the inhibitory effect of AMP in a concentration-dependent manner. Isotype control antibody R3-47 had no effect. The average of triplicate samples is plotted and error bars represent the standard deviation. Data are representative of three independent experiment.
Figure 4.
Figure 4.
Equal proportions of peripheral blood mononuclear cells from two healthy donors were mixed and incubated in wells of a 96 well plate for 96 h. Panel (A) shows brightfield images were taken at 24 h intervals using a 2.5× objective. Cells were treated with 150 µg/mL of either MEDI9447 (top panel) or and isotype control antibody (bottom panel). Panels (B–D). Equal proportions of peripheral blood mononuclear cells from two healthy donors were mixed incubated in wells of a 96 well plate for 72 h. Cells were treated with the indicated concentrations of either MEDI9447 (circles) or an isotype control antibody (squares). The plate was centrifuged to pellet cells and interferon-γ (Panel B), interleukin-1 β (Panel C), and tumor necrosis factor-α (Panel D) levels in the supernatants were measured by ELISA. Data are representative of experiments involving over 50 different donor-pair combinations.
Figure 5.
Figure 5.
(A). Murine CT26 colon carcinoma tumor cells were implanted subcutaneously then treated at 10 mg/kg with MEDI9447, or an isotype control (or untreated), by intraperitoneal injection on days 3, 6, 10, and 13 post-tumor implantation. An untreated control group was also included. The mean tumor volumes of each 10 animal group are plotted with error bars representing the standard error of the mean. Asterisks indicate a statistically significant (*p < 0.05) reduction in tumor volume. (B). Tumor-infiltrating leukocytes were isolated at study day 16 and analyzed by flow cytometry. CD8+ T effector cells were significantly increased in the tumor following treatment with MEDI9447. Data are representative of three independent experiments.
Figure 6.
Figure 6.
CT26 tumor-infiltrating leukocyte analysis from individual responder (R) and non-responder (NR) mice following MEDI9447 treatment. Murine CT26 colon carcinoma tumors cells were implanted subcutaneously then treated at 10 mg/kg with MEDI9447 by intraperitoneal injection on days 3, 6, 10, and 13 post-tumor implantation. Panel (A). Tumor volumes for individual mice treated with MEDI9447. Whole tumors were removed, dissociated into single cell suspensions, and analyzed by flow cytometry. The fraction of the following subpopulations were determined: total leukocytes (CD45+, Panel B), CD4+ lymphocytes (Panel C), monocyte-derived suppressor cells (Panel D), major histocompatibility (MHCII, Panel E ) expressed on F480+ macrophages as well as the mean fluorescent intensity of MHC2 (Panel F). Each symbol represents the phenotype indicated in the y axis for individual mice. Asterisks indicate significant (p < 0.05) differences between treatment arms. Data are representative of two independent experiments.
Figure 7.
Figure 7.
Murine CT26 colon carcinoma tumor cells were implanted subcutaneously and then treated at 10 mg/kg, by intraperitoneal injection on days 3, 7, 10, and 14 post-tumor implantation, with isotype control antibody (Panel A, open circles), MEDI9447 (Panel A, closed circles), anti-PD-1 (Panel B, squares) antibodies or a combination of MEDI9447 and anti-PD-1 (Panel C, diamonds) antibodies. Tumor volumes for individual animals are plotted until study day 29. The proportion of surviving mice after 40 d (Panel E) and the number of tumor-free mice in each treatment arm are shown. A 72-h mixed leukocyte reaction was performed using human peripheral blood mononuclear cells in the presence of anti-PD-1, MEDI944 or a combination of each antibody. The mean levels of secreted IFNγ and TNFα from duplicate samples are plotted (Panel F). The asterisk indicates a significant (p < 0.05) increase in survival. Data are representative of four independent experiments.

References

    1. Linden J, Cekic C. Regulation of lymphocyte function by adenosine. Arterioscler Thromb Vasc Biol 2012; 32:2097-103; PMID:22772752;
    1. Saze Z, Schuler PJ, Hong CS, Cheng D, Jackson EK, Whiteside TL. Adenosine production by human B cells and B cell-mediated suppression of activated T cells. Blood 2013; 122:9-18; PMID:23678003;
    1. Jalkanen S, Salmi M. VAP-1 and CD73, endothelial cell surface enzymes in leukocyte extravasation. Arterioscler Thromb Vasc Biol 2008; 28:18-26; PMID:17962625;
    1. Antonioli L, Pacher P, Vizi ES, Hasko G. CD39 and CD73 in immunity and inflammation. Trends Mol Med 2013; 19:355-67; PMID:23601906;
    1. Ohta A, Gorelik E, Prasad SJ, Ronchese F, Lukashev D, Wong MK, Huang X, Caldwell S, Liu K, Smith P et al.. A2A adenosine receptor protects tumors from antitumor T cells. Proc Natl Acad Sci USA 2006; 103:13132-7; PMID:16916931;
    1. Waickman AT, Alme A, Senaldi L, Zarek PE, Horton M, Powell JD. Enhancement of tumor immunotherapy by deletion of the A2A adenosine receptor. Cancer Immunol Immunother 2012; 61:917-26; PMID:22116345;
    1. Stagg J, Beavis PA, Divisekera U, Liu MC, Moller A, Darcy PK, Smyth MJ. CD73-deficient mice are resistant to carcinogenesis. Cancer Res 2012; 72:2190-6; PMID:22396496;
    1. Antonioli L, Giron MC, Colucci R, Pellegrini C, Sacco D, Caputi V, Orso G, Tuccori M, Scarpignato C, Blandizzi C et al.. Involvement of the P2X7 purinergic receptor in colonic motor dysfunction associated with bowel inflammation in rats. PLoS One 2014; 9:e116253; PMID:25549098;
    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; 3:506-17; PMID:25672397;
    1. Young A, Mittal D, Stagg J, Smyth MJ. Targeting cancer-derived adenosine: New therapeutic approaches. Cancer Discov 2014; 4:879-88; PMID:25035124;
    1. Leone RD, Lo YC, Powell JD. A2aR antagonists: Next generation checkpoint blockade for cancer immunotherapy. Comput Struct Biotechnol J 2015; 13:265-72; PMID:25941561;
    1. Clancy-Thompson E, Perekslis TJ, Croteau W, Alexander MP, Chabanet TB, Turk MJ, Huang YH, Mullins DW. Melanoma induces, and adenosine suppresses, CXCR3-cognate chemokine production and T-cell infiltration of lungs bearing metastatic-like disease. Cancer Immunol Res 2015; 3(8):956-67; PMID:26048575;
    1. Oganesyan V, Gao C, Shirinian L, Wu H, Dall'Acqua WF. Structural characterization of a human FC fragment engineered for lack of effector functions. Acta Crystallogr D Biol Crystallogr 2008; 64:700-4; PMID:18560159;
    1. Rust S, Guillard S, Sachsenmeier K, Hay C, Davidson M, Karlsson A, Karlsson R, Brand E, Lowne D, Elvin J et al.. Combining phenotypic and proteomic approaches to identify membrane targets in a 'triple negative' breast cancer cell type. Mol Cancer 2013; 12:11-4598-12-11; PMID:23406016;
    1. Lesokhin AM, Callahan MK, Postow MA, Wolchok JD. On being less tolerant: Enhanced cancer immunosurveillance enabled by targeting checkpoints and agonists of T cell activation. Sci Transl Med 2015; 7:280sr1; PMID:25810313;
    1. Vonderheide RH, Glennie MJ. Agonistic CD40 antibodies and cancer therapy. Clin Cancer Res 2013; 19:1035-43; PMID:23460534;
    1. Platten M, von Knebel Doeberitz N, Oezen I, Wick W, Ochs K. Cancer immunotherapy by targeting IDO1/TDO and their downstream effectors. Front Immunol 2015; 5:673; PMID:25628622;
    1. Rolinski J, Hus I. Breaking immunotolerance of tumors: A new perspective for dendritic cell therapy. J Immunotoxicol 2014; 11:311-8; PMID:24495309;
    1. Moran AE, Kovacsovics-Bankowski M, Weinberg AD. The TNFRs OX40, 4-1BB, and CD40 as targets for cancer immunotherapy. Curr Opin Immunol 2013; 25:230-7; PMID:23414607;
    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; 19:5626-35; PMID:23983257;
    1. Resta R, Thompson LF. T cell signalling through CD73. Cell Signal 1997; 9:131-9; PMID:9113412;
    1. Mikhailov A, Sokolovskaya A, Yegutkin GG, Amdahl H, West A, Yagita H, Lahesmaa R, Thompson LF, Jalkanen S, Blokhin D et al.. CD73 participates in cellular multiresistance program and protects against TRAIL-induced apoptosis. J Immunol 2008; 181:464-75; PMID:18566412;
    1. 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; PMID:23982901;
    1. Terp MG, Olesen KA, Arnspang EC, Lund RR, Lagerholm BC, Ditzel HJ, Leth-Larsen R. Anti-human CD73 monoclonal antibody inhibits metastasis formation in human breast cancer by inducing clustering and internalization of CD73 expressed on the surface of cancer cells. J Immunol 2013; 191:4165-73; PMID:24043904;
    1. Cekic C, Linden J. Adenosine A2A receptors intrinsically regulate CD8+ T cells in the tumor microenvironment. Cancer Res 2014; 74:7239-49; PMID:25341542;
    1. Leone RD, Lo YC, Powell JD. A2aR antagonists: Next generation checkpoint blockade for cancer immunotherapy. Comput Struct Biotechnol J 2015; 13:265-72; PMID:25941561;
    1. Fedele G, Sanseverino I, D'Agostino K, Schiavoni I, Locht C, Horenstein AL, Malavasi F, Ausiello CM. Unconventional, adenosine-producing suppressor T cells induced by dendritic cells exposed to BPZE1 pertussis vaccine. J Leukoc Biol 2015; 98:631-9; PMID:26089537;
    1. Geoghegan JC, Diedrich G, Lu X, Rosenthal K, Sachsenmeier KF, Wu H, Dall'Acqua WF, Damschroder M. Inhibition of CD73 AMP hydrolysis by a therapeutic antibody with a dual, non-competitive mechanism of action. MAbs 2016; 8(3):454-67; PMID:26854859;
    1. Jin D, Fan J, Wang L, Thompson LF, Liu A, Daniel BJ, Shin T, Curiel TJ, Zhang B. CD73 on tumor cells impairs antitumor T-cell responses: A novel mechanism of tumor-induced immune suppression. Cancer Res 2010; 70:2245-55; PMID:20179192;
    1. Forte G, Sorrentino R, Montinaro A, Luciano A, Adcock IM, Maiolino P, Arra C, Cicala C, Pinto A, Morello S. Inhibition of CD73 improves B cell-mediated anti-tumor immunity in a mouse model of melanoma. J Immunol 2012; 189:2226-33; PMID:22826317;
    1. Ryzhov S, Novitskiy SV, Goldstein AE, Biktasova A, Blackburn MR, Biaggioni I, Dikov MM, Feoktistov I. Adenosinergic regulation of the expansion and immunosuppressive activity of CD11b+Gr1+ cells. J Immunol 2011; 187:6120-9; PMID:22039302;
    1. Muller-Haegele S, Muller L, Whiteside TL. Immunoregulatory activity of adenosine and its role in human cancer progression. Expert Rev Clin Immunol 2014; 10:897-914; PMID:24871693;
    1. Beavis PA, Slaney CY, Kershaw MH, Neeson PJ, Darcy PK. Enhancing the efficacy of adoptive cellular therapy by targeting tumor-induced immunosuppression. Immunotherapy 2015; 7:499-512; PMID:26065476;
    1. Teng MW, Ngiow SF, Ribas A, Smyth MJ. Classifying cancers based on T-cell infiltration and PD-L1. Cancer Res 2015; 75:2139-45; PMID:25977340;
    1. Thomson LF, Ruedi JM, Glass A, Moldenhauer G, Moller P, Low MG, Klemens MR, Massaia M, Lucas AH. Production and characterization of monoclonal antibodies to the glycosyl phosphatidylinositol-anchored lymphocyte differentiation antigen ecto-5'-nucleotidase (CD73). Tissue Antigens 1990; 35:9-19; PMID:2137649
    1. Airas L, Salmi M, Jalkanen S. Lymphocyte-vascular adhesion protein-2 is a novel 70-kDa molecule involved in lymphocyte adhesion to vascular endothelium. J Immunol 1993; 151:4228-38; PMID:8409398
    1. Airas L, Niemela J, Salmi M, Puurunen T, Smith DJ, Jalkanen S. Differential regulation and function of CD73, a glycosyl-phosphatidylinositol-linked 70-kD adhesion molecule, on lymphocytes and endothelial cells. J Cell Biol 1997; 136:421-31; PMID:9015312;
    1. Lloyd A, Vickery ON, Laugel B. Beyond the antigen receptor: Editing the genome of T-cells for cancer adoptive cellular therapies. Front Immunol 2013; 4:221; PMID:23935598;
    1. Groves M, Lane S, Douthwaite J, Lowne D, Rees DG, Edwards B, Jackson RH. Affinity maturation of phage display antibody populations using ribosome display. J Immunol Methods 2006; 313:129-39; PMID:16730741;
    1. Oganesyan V, Gao C, Shirinian L, Wu H, Dall'Acqua WF. Structural characterization of a human fc fragment engineered for lack of effector functions. Acta Crystallogr D Biol Crystallogr 2008; 64:700-4; PMID:18560159;
    1. Sachsenmeier KF, Hay C, Brand E, Clarke L, Rosenthal K, Guillard S, Rust S, Minter R, Hollingsworth R. Development of a novel ectonucleotidase assay suitable for high-throughput screening. J Biomol Screen 2012; 17:993-8; PMID:22522649;
    1. National Research Council (US) Committee for the Update of the Guide for the Care and Use of Laboratory Animals . 2011; PMID:21595115; [bookaccession]

Source: PubMed

3
구독하다