Activating Fc γ receptors contribute to the antitumor activities of immunoregulatory receptor-targeting antibodies

Yannick Bulliard, Rose Jolicoeur, Maurice Windman, Sarah M Rue, Seth Ettenberg, Deborah A Knee, Nicholas S Wilson, Glenn Dranoff, Jennifer L Brogdon, Yannick Bulliard, Rose Jolicoeur, Maurice Windman, Sarah M Rue, Seth Ettenberg, Deborah A Knee, Nicholas S Wilson, Glenn Dranoff, Jennifer L Brogdon

Abstract

Fc γ receptor (FcγR) coengagement can facilitate antibody-mediated receptor activation in target cells. In particular, agonistic antibodies that target tumor necrosis factor receptor (TNFR) family members have shown dependence on expression of the inhibitory FcγR, FcγRIIB. It remains unclear if engagement of FcγRIIB also extends to the activities of antibodies targeting immunoregulatory TNFRs expressed by T cells. We have explored the requirement for activating and inhibitory FcγRs for the antitumor effects of antibodies targeting the TNFR glucocorticoid-induced TNFR-related protein (GITR; TNFRSF18; CD357) expressed on activated and regulatory T cells (T reg cells). We found that although FcγRIIB was dispensable for the in vivo efficacy of anti-GITR antibodies, in contrast, activating FcγRs were essential. Surprisingly, the dependence on activating FcγRs extended to an antibody targeting the non-TNFR receptor CTLA-4 (CD152) that acts as a negative regulator of T cell immunity. We define a common mechanism that correlated with tumor efficacy, whereby antibodies that coengaged activating FcγRs expressed by tumor-associated leukocytes facilitated the selective elimination of intratumoral T cell populations, particularly T reg cells. These findings may have broad implications for antibody engineering efforts aimed at enhancing the therapeutic activity of immunomodulatory antibodies.

Figures

Figure 1.
Figure 1.
Activating, rather than inhibitory, FcγRs are necessary for the antitumor activity of an agonistic antibody to GITR. Efficacy study of anti-GITR antibody (DTA-1 rIgG2b; 5 mg/kg i.p.) in wild type (A), FcγRIIB−/− (B), and Fc common γ chain−/− (C) BALB/c mice bearing Colon26 tumors (n = 6–10 mice per treatment group). Day 0 refers to treatment day, 6–8 d after tumor inoculation. Data is a representative of two or more independent experiments.
Figure 2.
Figure 2.
Engagement of FcγRs by DTA-1 is required for antitumor activity. (A and B) GITR-binding assay. Primary splenocytes stimulated with CD3- and CD28-specific antibodies served as targets. DTA-1 variants were detected using rat IgG2b-specific (A) or murine IgG2a-specific (B) PE-conjugated antibodies. (C and D) In vitro activity of GITR-specific antibodies at various concentrations tested on GITR-expressing NF-kB 293 reporter cell line (C) and splenocytes incubated with suboptimal doses of anti-CD3 and anti-CD28 antibodies (D). The in vitro data are derived from triplicates and are a representative of two or more independent experiments. (E) Efficacy study of 5 mg/kg DTA-1 mIgG2a in wild-type, FcγRIIB−/−, and Fc common γ chain−/− mice bearing Colon26 tumors (n = 7–10 mice per treatment group). Mean and standard errors are based on triplicates, and the data is a representative of two or more independent experiments. (F) In vivo efficacy study after treatment with the DTA-1 variant antibodies (n = 7). The efficacy data are a representative of two or more independent experiments. (G) Saturation of GITR on T reg cells in the tumor and draining lymph node by the three versions of DTA-1.
Figure 3.
Figure 3.
Engagement of FcγRs by DTA-1 induces loss of intratumoral T reg cells early after treatment. (A) Cell surface expression of GITR on day 0 (n = 3). (B and C) Dot plots of CD11b+ myeloid and DX5+ NK cells, gated on live CD45+ leukocytes (B) and expression of FcγRII/III and IV by the two cell populations in the tumor (C). (D) Dot plots of T cells 3 d after treatment with the DTA-1 variant antibodies. Percentages of live CD45+ CD3+ T cells are indicated. The data are a representative of two or more independent experiments. (E and F) Density of T reg cells (E) and FoxP3-CD4+ T cells and CD8+ T cells (F) in the tumor after treatment with the 5 mg/kg DTA-1 variants. (G) Ratio between CD8+ T cells and T reg cells in the tumor. (H) Percentage of intratumoral CD45+ leukocytes. (I) T cell number in the draining lymph nodes after treatment with 5 mg/kg of the DTA-1 variants. Mean and standard errors are based on triplicates from two independent experiments. P-values were calculated using an unpaired Student’s t test (**, P < 0.001).
Figure 4.
Figure 4.
Activating, but not inhibitory, FcγRs are required for intratumoral T reg cell depletion by antibodies targeting GITR. Intratumoral T cell density and CD8+ T cells to T reg cells ratios 5 d after treatment with 5 mg/kg DTA-1-mIgG2a using wild-type (A), FcγRIIB−/− (B), or Fc common γ chain−/− (C) mice bearing Colon26 tumors. (D–F) Cell surface expression of GITR on T cells. Mean and standard errors are based on triplicates from two independent experiments. P-values were calculated using an unpaired Student’s t test (**, P < 0.001).
Figure 5.
Figure 5.
Engagement of activating FcγRs is also required for the antitumor activity of an antibody targeting the non-TNFR CTLA-4. (A) Efficacy study using 15 mg/kg CTLA-4–specific antibody (9D9) in wild-type (top) or Fc common γ chain−/− (middle) mice bearing Colon26 tumors (n = 8–10 mice per treatment group). (B) Intratumoral T cell density and CD8+ to T reg cells ratios 5 d after treatment. (C) Cell surface expression of CTLA-4 on T cells. The efficacy data are a representative of two or more independent experiments. Mean and standard errors are based on triplicates from two independent experiments. P-values were calculated using an unpaired Student’s t test (*, P < 0.01).

References

    1. Albanesi M., Mancardi D.A., Macdonald L.E., Iannascoli B., Zitvogel L., Murphy A.J., Daëron M., Leusen J.H., Bruhns P. 2012. Cutting edge: FcγRIII (CD16) and FcγRI (CD64) are responsible for anti-glycoprotein 75 monoclonal antibody TA99 therapy for experimental metastatic B16 melanoma. J. Immunol. 189:5513–5517 10.4049/jimmunol.1201511
    1. Chao D.T., Ma X., Li O., Park H., Law D. 2009. Functional characterization of N297A, a murine surrogate for low-Fc binding anti-human CD3 antibodies. Immunol. Invest. 38:76–92 10.1080/08820130802608238
    1. Chung K.Y., Gore I., Fong L., Venook A., Beck S.B., Dorazio P., Criscitiello P.J., Healey D.I., Huang B., Gomez-Navarro J., Saltz L.B. 2010. Phase II study of the anti-cytotoxic T-lymphocyte-associated antigen 4 monoclonal antibody, tremelimumab, in patients with refractory metastatic colorectal cancer. J. Clin. Oncol. 28:3485–3490 10.1200/JCO.2010.28.3994
    1. Clynes R.A., Towers T.L., Presta L.G., Ravetch J.V. 2000. Inhibitory Fc receptors modulate in vivo cytotoxicity against tumor targets. Nat. Med. 6:443–446 10.1038/74704
    1. Cohen A.D., Schaer D.A., Liu C., Li Y., Hirschhorn-Cymmerman D., Kim S.C., Diab A., Rizzuto G., Duan F., Perales M.A., et al. 2010. Agonist anti-GITR monoclonal antibody induces melanoma tumor immunity in mice by altering regulatory T cell stability and intra-tumor accumulation. PLoS ONE. 5:e10436 10.1371/journal.pone.0010436
    1. Curran M.A., Montalvo W., Yagita H., Allison J.P. 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. 107:4275–4280 10.1073/pnas.0915174107
    1. Hodi F.S., O’Day S.J., McDermott D.F., Weber R.W., Sosman J.A., Haanen J.B., Gonzalez R., Robert C., Schadendorf D., Hassel J.C., et al. 2010. Improved survival with ipilimumab in patients with metastatic melanoma. N. Engl. J. Med. 363:711–723 10.1056/NEJMoa1003466
    1. Hogarth P.M., Pietersz G.A. 2012. Fc receptor-targeted therapies for the treatment of inflammation, cancer and beyond. Nat. Rev. Drug Discov. 11:311–331 10.1038/nrd2909
    1. Junttila T.T., Parsons K., Olsson C., Lu Y., Xin Y., Theriault J., Crocker L., Pabonan O., Baginski T., Meng G., et al. 2010. Superior in vivo efficacy of afucosylated trastuzumab in the treatment of HER2-amplified breast cancer. Cancer Res. 70:4481–4489 10.1158/0008-5472.CAN-09-3704
    1. Kanamaru F., Youngnak P., Hashiguchi M., Nishioka T., Takahashi T., Sakaguchi S., Ishikawa I., Azuma M. 2004. Costimulation via glucocorticoid-induced TNF receptor in both conventional and CD25+ regulatory CD4+ T cells. J. Immunol. 172:7306–7314
    1. Ko K., Yamazaki S., Nakamura K., Nishioka T., Hirota K., Yamaguchi T., Shimizu J., Nomura T., Chiba T., Sakaguchi S. 2005. Treatment of advanced tumors with agonistic anti-GITR mAb and its effects on tumor-infiltrating Foxp3+CD25+CD4+ regulatory T cells. J. Exp. Med. 202:885–891 10.1084/jem.20050940
    1. Leach D.R., Krummel M.F., Allison J.P. 1996. Enhancement of antitumor immunity by CTLA-4 blockade. Science. 271:1734–1736 10.1126/science.271.5256.1734
    1. Lewis G.D., Figari I., Fendly B., Wong W.L., Carter P., Gorman C., Shepard H.M. 1993. Differential responses of human tumor cell lines to anti-p185HER2 monoclonal antibodies. Cancer Immunol. Immunother. 37:255–263 10.1007/BF01518520
    1. Li F., Ravetch J.V. 2012. A general requirement for FcγRIIB co-engagement of agonistic anti-TNFR antibodies. Cell Cycle. 11:3343–3344 10.4161/cc.21842
    1. Mellman I., Coukos G., Dranoff G. 2011. Cancer immunotherapy comes of age. Nature. 480:480–489 10.1038/nature10673
    1. Nimmerjahn F., Ravetch J.V. 2005. Divergent immunoglobulin g subclass activity through selective Fc receptor binding. Science. 310:1510–1512 10.1126/science.1118948
    1. Nimmerjahn F., Ravetch J.V. 2008. Fcgamma receptors as regulators of immune responses. Nat. Rev. Immunol. 8:34–47 10.1038/nri2206
    1. Nimmerjahn F., Ravetch J.V. 2012. Translating basic mechanisms of IgG effector activity into next generation cancer therapies. Cancer Immun. 12:13.
    1. Niwa R., Sakurada M., Kobayashi Y., Uehara A., Matsushima K., Ueda R., Nakamura K., Shitara K. 2005. Enhanced natural killer cell binding and activation by low-fucose IgG1 antibody results in potent antibody-dependent cellular cytotoxicity induction at lower antigen density. Clin. Cancer Res. 11:2327–2336 10.1158/1078-0432.CCR-04-2263
    1. Nordstrom J.L., Gorlatov S., Zhang W., Yang Y., Huang L., Burke S., Li H., Ciccarone V., Zhang T., Stavenhagen J., et al. 2011. Anti-tumor activity and toxicokinetics analysis of MGAH22, an anti-HER2 monoclonal antibody with enhanced Fcγ receptor binding properties. Breast Cancer Res. 13:R123 10.1186/bcr3069
    1. Robert C., Thomas L., Bondarenko I., O’Day S., M D J.W., Garbe C., Lebbe C., Baurain J.F., Testori A., Grob J.J., et al. 2011. Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. N. Engl. J. Med. 364:2517–2526 10.1056/NEJMoa1104621
    1. Ronchetti S., Nocentini G., Bianchini R., Krausz L.T., Migliorati G., Riccardi C. 2007. Glucocorticoid-induced TNFR-related protein lowers the threshold of CD28 costimulation in CD8+ T cells. J. Immunol. 179:5916–5926
    1. Ronchetti S., Nocentini G., Petrillo M.G., Riccardi C. 2012. CD8+ T cells: GITR matters. ScientificWorldJournal. 2012:308265 10.1100/2012/308265
    1. Schaer D.A., Murphy J.T., Wolchok J.D. 2012. Modulation of GITR for cancer immunotherapy. Curr. Opin. Immunol. 24:217–224 10.1016/j.coi.2011.12.011
    1. Sharma S., Dominguez A.L., Manrique S.Z., Cavallo F., Sakaguchi S., Lustgarten J. 2008. Systemic targeting of CpG-ODN to the tumor microenvironment with anti-neu-CpG hybrid molecule and T regulatory cell depletion induces memory responses in BALB-neuT tolerant mice. Cancer Res. 68:7530–7540 10.1158/0008-5472.CAN-08-1635
    1. Shields R.L., Namenuk A.K., Hong K., Meng Y.G., Rae J., Briggs J., Xie D., Lai J., Stadlen A., Li B., et al. 2001. High resolution mapping of the binding site on human IgG1 for Fc gamma RI, Fc gamma RII, Fc gamma RIII, and FcRn and design of IgG1 variants with improved binding to the Fc gamma R. J. Biol. Chem. 276:6591–6604 10.1074/jbc.M009483200
    1. Shimizu J., Yamazaki S., Takahashi T., Ishida Y., Sakaguchi S. 2002. Stimulation of CD25(+)CD4(+) regulatory T cells through GITR breaks immunological self-tolerance. Nat. Immunol. 3:135–142 10.1038/ni759
    1. Smith P., DiLillo D.J., Bournazos S., Li F., Ravetch J.V. 2012. Mouse model recapitulating human Fcγ receptor structural and functional diversity. Proc. Natl. Acad. Sci. USA. 109:6181–6186 10.1073/pnas.1203954109
    1. Tai Y.T., Horton H.M., Kong S.Y., Pong E., Chen H., Cemerski S., Bernett M.J., Nguyen D.H., Karki S., Chu S.Y., et al. 2012. Potent in vitro and in vivo activity of an Fc-engineered humanized anti-HM1.24 antibody against multiple myeloma via augmented effector function. Blood. 119:2074–2082 10.1182/blood-2011-06-364521
    1. Turk M.J., Guevara-Patiño J.A., Rizzuto G.A., Engelhorn M.E., Sakaguchi S., Houghton A.N. 2004. Concomitant tumor immunity to a poorly immunogenic melanoma is prevented by regulatory T cells. J. Exp. Med. 200:771–782 10.1084/jem.20041130
    1. Weng W.K., Levy R. 2003. Two immunoglobulin G fragment C receptor polymorphisms independently predict response to rituximab in patients with follicular lymphoma. J. Clin. Oncol. 21:3940–3947 10.1200/JCO.2003.05.013
    1. Wilson N.S., Yang B., Yang A., Loeser S., Marsters S., Lawrence D., Li Y., Pitti R., Totpal K., Yee S., et al. 2011. An Fcγ receptor-dependent mechanism drives antibody-mediated target-receptor signaling in cancer cells. Cancer Cell. 19:101–113 10.1016/j.ccr.2010.11.012
    1. Zhou P., L’italien L., Hodges D., Schebye X.M. 2007. Pivotal roles of CD4+ effector T cells in mediating agonistic anti-GITR mAb-induced-immune activation and tumor immunity in CT26 tumors. J. Immunol. 179:7365–7375

Source: PubMed

3
Abonner