Antibody-mediated phagocytosis contributes to the anti-tumor activity of the therapeutic antibody daratumumab in lymphoma and multiple myeloma

Marije B Overdijk, Sandra Verploegen, Marijn Bögels, Marjolein van Egmond, Jeroen J Lammerts van Bueren, Tuna Mutis, Richard W J Groen, Esther Breij, Anton C M Martens, Wim K Bleeker, Paul W H I Parren, Marije B Overdijk, Sandra Verploegen, Marijn Bögels, Marjolein van Egmond, Jeroen J Lammerts van Bueren, Tuna Mutis, Richard W J Groen, Esther Breij, Anton C M Martens, Wim K Bleeker, Paul W H I Parren

Abstract

Daratumumab (DARA) is a human CD38-specific IgG1 antibody that is in clinical development for the treatment of multiple myeloma (MM). The potential for IgG1 antibodies to induce macrophage-mediated phagocytosis, in combination with the known presence of macrophages in the tumor microenvironment in MM and other hematological tumors, led us to investigate the contribution of antibody-dependent, macrophage-mediated phagocytosis to DARA's mechanism of action. Live cell imaging revealed that DARA efficiently induced macrophage-mediated phagocytosis, in which individual macrophages rapidly and sequentially engulfed multiple tumor cells. DARA-dependent phagocytosis by mouse and human macrophages was also observed in an in vitro flow cytometry assay, using a range of MM and Burkitt's lymphoma cell lines. Phagocytosis contributed to DARA's anti-tumor activity in vivo, in both a subcutaneous and an intravenous leukemic xenograft mouse model. Finally, DARA was shown to induce macrophage-mediated phagocytosis of MM cells isolated from 11 of 12 MM patients that showed variable levels of CD38 expression. In summary, we demonstrate that phagocytosis is a fast, potent and clinically relevant mechanism of action that may contribute to the therapeutic activity of DARA in multiple myeloma and potentially other hematological tumors.

Keywords: ADCC, antibody-dependent cellular cytotoxicity; BL, Burkitt's lymphoma; BM, bone marrow; Burkitt's lymphoma; CCS, cosmic calf serum; CD38; CDC, complement-dependent cytotoxicity; DARA, daratumumab; DP, double positive; E:T, effector to target ratio; FcγR, Fc-gamma receptor; IMiD, immunomodulatory drug; MM, multiple myeloma; MNC, mononuclear cells; Mϕ, macrophage; PBMC, peripheral blood mononuclear cells; daratumumab; mAb, monoclonal antibody; macrophage; multiple myeloma; phagocytosis; therapeutic antibody.

Figures

Figure 1.
Figure 1.
Flow cytometry and live cell imaging reveals macrophage-mediated phagocytosis of CD38+ tumor cells in the presence of DARA. Co-cultures of mouse mφ and Daudi cells in the presence of 6.7 nM DARA or F(ab’)2 fragments thereof, E:T ratio of 1:1 (A, B) or 3:1 (C). (A) Double positive (DP) mφ were characterized as F4/80+calcein+CD19– and the percentage DP macrophages was calculated as described in Materials and Methods. (B) The percentage eliminated target cells was calculated from the number of remaining F4/80- cells as described in Materials & Methods. Each bar shows mean ± SEM, results from a representative experiment are shown (n = 3). (C) Time-lapse imaging microscopy, bright field images of a mouse mφ (arrow) that sequentially engulfed 5 individual Daudi cells (numbers) over a period of 800 s. The images are representative for observations in multiple independent phagocytosis experiments (n = 3) (****p < 0.0001 Bonferroni's multiple comparison test).
Figure 2.
Figure 2.
Induction of macrophage-mediated phagocytosis is strongly impaired for an IgG2 isotype variant of DARA. Phagocytosis of Daudi cells by mouse mφ in the presence of 6.7 nM mAb, E:T ratio of 1:1. (A) Double-positive (DP) mφ were characterized as F4/80+calcein+CD19– and the percentage DP macrophages was calculated as described in Materials and Methods. (B) Percentage eliminated target cells was calculated using the number of remaining F4/80- cells as described in Materials & Methods. Each bar shows mean ± SEM, results from a representative experiment (n = 3) (**p < 0.01, ****p < 0.0001 Bonferroni's multiple comparison test).
Figure 3.
Figure 3.
Phagocytosis contributes to the in vivo anti-tumor effect of DARA. (A) Kaplan-Meier plot showing time to tumor progression (cutoff set at a tumor volume > 800 mm3) for mice that had been inoculated s.c. with 20 × 106 Daudi-luc cells (8 mice per group). Subsequently, mice were treated i.p. with 250 μg mAb per mouse (∼12.5 mg/kg) at day 0. Tumor progression was significantly reduced in DARA-K322A-treated mice compared to DARA-IgG2-K322A treatment (p < 0.004 Mantle-Cox log-rank test at time to progression). (B) Kaplan-Meier plot showing time to tumor progression (cutoff set at bioluminescence > 50 000 cpm) for mice that had been inoculated i.v. with 2.5×106 Daudi-luc cells (10 mice per group). Subsequently, mice were treated i.p. with 10 μg mAb per mouse (∼0.5 mg/kg) at day 0. Tumor progression was significantly reduced in DARA-K322A-treated vs. DARA-IgG2-K322A-treated mice (p < 0.001 Mantle-Cox log-rank test at time to progression).
Figure 4.
Figure 4.
Human macrophages induce DARA-dependent phagocytosis of BL and MM cell lines. Phagocytosis of BL cells (left panel) or MM cells (right panel) by human mφ in the presence of 6.7 nM mAb, E:T ratio of 2:1. (A) Percentage of double positive (DP) mφ characterized as CD11b+calcein+target Ag–. (B) Percentage of eliminated target cells calculated from the number of remaining CD11b- cells as described in Materials & Methods. Each bar shows mean ± SEM, results from a representative experiment (n = 3) (**p < 0.01, ***p < 0.001, ****p < 0.0001 Bonferroni's multiple comparison test).
Figure 5.
Figure 5.
DARA-dependent phagocytosis of patient MM cells by human macrophages. Patient MM cells were incubated with human mφ, obtained from healthy donors (E:T ratio 2:1), in the presence of 6.7 nM mAb. To correct for differences in potency between batches of human donor mφ obtained from different donors, results were normalized by calculating the ratio of patient MM cells to an internal Daudi cell standard (observed in the same experiment). The 12 patient samples are ranked according to their CD38 expression level, with cells from patient 6 exhibiting lowest (10,000 molecules per cells) and patient 5 highest (550,000 molecules/cell) CD38 expression. (A) Normalized percentage of DARA-specific double positive (DP) mφ. (B) Normalized percentage of DARA-specific eliminated target cells. Each bar shows mean ± SEM of triplicates.

References

    1. Manches O, Lui G, Chaperot L, Gressin R, Molens JP, Jacob MC, Sotto JJ, Leroux D, Bensa JC, Plumas J. In vitro mechanisms of action of rituximab on primary non-Hodgkin lymphomas. Blood 2003; 101:949-54; PMID:12393572;
    1. Munn DH, McBride M, Cheung NK. Role of low-affinity Fc receptors in antibody-dependent tumor cell phagocytosis by human monocyte-derived macrophages. Cancer Res 1991; 51:1117-23; PMID:1825476
    1. Mantovani A, Schioppa T, Porta C, Allavena P, Sica A. Role of tumor-associated macrophages in tumor progression and invasion. Cancer Metastasis Rev 2006; 25:315-22; PMID:16967326;
    1. Ribatti D, Moschetta M, Vacca A. Macrophages in multiple myeloma. Immunol Lett 2014; 161:241–4; PMID:24370642.
    1. Zheng Y, Cai Z, Wang S, Zhang X, Qian J, Hong S, Li H, Wang M, Yang J, Yi Q. Macrophages are an abundant component of myeloma microenvironment and protect myeloma cells from chemotherapy drug-induced apoptosis. Blood 2009; 114:3625-8; PMID:19710503;
    1. Uchida J, Hamaguchi Y, Oliver JA, Ravetch JV, Poe JC, Haas KM, Tedder TF. The innate mononuclear phagocyte network depletes B lymphocytes through Fc receptor-dependent mechanisms during anti-CD20 antibody immunotherapy. J Exp Med 2004; 199:1659-69; PMID:15210744;
    1. Minard-Colin V, Xiu Y, Poe JC, Horikawa M, Magro CM, Hamaguchi Y, Haas KM, Tedder TF. Lymphoma depletion during CD20 immunotherapy in mice is mediated by macrophage FcgammaRI, FcgammaRIII, and FcgammaRIV. Blood 2008; 112:1205-13; PMID:18495955;
    1. McEarchern JA, Oflazoglu E, Francisco L, McDonagh CF, Gordon KA, Stone I, Klussman K, Turcott E, van Rooijen N, Carter P, Grewal IS, Wahl AF, Law CL. Engineered anti-CD70 antibody with multiple effector functions exhibits in vitro and in vivo antitumor activities. Blood 2007; 109:1185-92; PMID:17038522;
    1. Oflazoglu E, Stone IJ, Brown L, Gordon KA, van Rooijen N, Jonas M, Law CL, Grewal IS, Gerber HP. Macrophages and Fc-receptor interactions contribute to the antitumour activities of the anti-CD40 antibody SGN-40. Br J Cancer 2009; 100:113-7; PMID:19066610;
    1. Oflazoglu E, Stone IJ, Gordon KA, Grewal IS, van Rooijen N, Law CL, Gerber HP. Macrophages contribute to the antitumor activity of the anti-CD30 antibody SGN-30. Blood 2007; 110:4370-2; PMID:17909075;
    1. Lin P, Owens R, Tricot G, Wilson CS. Flow cytometric immunophenotypic analysis of 306 cases of multiple myeloma. Am J Clin Pathol 2004; 121:482-8; PMID:15080299;
    1. de Weers M, Tai YT, van der Veer MS, Bakker JM, Vink T, Jacobs DC, Oomen LA, Peipp M, Valerius T, Slootstra JW, et al. . Daratumumab, a novel therapeutic human CD38 monoclonal antibody, induces killing of multiple myeloma and other hematological tumors. J Immunol 2011; 186:1840-8; PMID:21187443;
    1. Deckert J, Wetzel MC, Bartle LM, Skaletskaya A, Goldmacher V, Vallee F, Zhou-Liu Q, Ferrari P, Pouzieux S, Lahoute C, et al. . SAR650984, a novel humanized CD38-targeting antibody, demonstrates potent anti-tumor activity in models of multiple myeloma and other CD38+ hematologic malignancies. Clin Cancer Res 2014; 20:4574-83; PMID:24987056;
    1. Tai YT, Horton HM, Kong SY, Pong E, Chen H, Cemerski S, Bernett MJ, Nguyen DH, Karki S, Chu SY, et al. . Potent in vitro and in vivo activity of an Fc-engineered humanized anti-HM1.24 antibody against multiple myeloma via augmented effector function. Blood 2012; 119:2074-82; PMID:22246035;
    1. Tai YT, Mayes PA, Acharya C, Zhong MY, Cea M, Cagnetta A, Craigen J, Yates J, Gliddon L, Fieles W, et al. . Novel anti-B-cell maturation antigen antibody-drug conjugate (GSK2857916) selectively induces killing of multiple myeloma. Blood 2014; 123:3128-38; PMID:24569262;
    1. Overdijk MB, Verploegen S, Ortiz Buijsse A, Vink T, Leusen JH, Bleeker WK, Parren PW. Crosstalk between human IgG isotypes and murine effector cells. J Immunol 2012; 189:3430-8; PMID:22956577;
    1. Duncan AR, Winter G. The binding site for C1q on IgG. Nature 1988; 332:738-40; PMID:3258649;
    1. Idusogie EE, Presta LG, Gazzano-Santoro H, Totpal K, Wong PY, Ultsch M, Meng YG, Mulkerrin MG. Mapping of the C1q binding site on rituxan, a chimeric antibody with a human IgG1 Fc. J Immunol 2000; 164:4178-84; PMID:10754313;
    1. Overdijk MB, Verploegen S, van den Brakel JH, Lammerts van Bueren JJ, Vink T, van de Winkel JG, Parren PW, Bleeker WK. Epidermal growth factor receptor (EGFR) antibody-induced antibody-dependent cellular cytotoxicity plays a prominent role in inhibiting tumorigenesis, even of tumor cells insensitive to EGFR signaling inhibition. J Immunol 2011; 187:3383-90; PMID:21832160;
    1. Leidi M, Gotti E, Bologna L, Miranda E, Rimoldi M, Sica A, Roncalli M, Palumbo GA, Introna M, Golay J. M2 macrophages phagocytose rituximab-opsonized leukemic targets more efficiently than m1 cells in vitro. J Immunol 2009; 182:4415-22; PMID:19299742;
    1. van Meerten T, van Rijn RS, Hol S, Hagenbeek A, Ebeling SB. Complement-induced cell death by rituximab depends on CD20 expression level and acts complementary to antibody-dependent cellular cytotoxicity. Clin Cancer Res 2006; 12:4027-35; PMID:16818702;
    1. Golay J, Lazzari M, Facchinetti V, Bernasconi S, Borleri G, Barbui T, Rambaldi A, Introna M. CD20 levels determine the in vitro susceptibility to rituximab and complement of B-cell chronic lymphocytic leukemia: further regulation by CD55 and CD59. Blood 2001; 98:3383-9; PMID:11719378;
    1. Swanson JA, Hoppe AD. The coordination of signaling during Fc receptor-mediated phagocytosis. J Leukoc Biol 2004; 76:1093-103; PMID:15466916;
    1. Paul D, Achouri S, Yoon YZ, Herre J, Bryant CE, Cicuta P. Phagocytosis dynamics depends on target shape. Biophys J 2013; 105:1143-50; PMID:24010657;
    1. Oldenborg PA, Gresham HD, Lindberg FP. CD47-signal regulatory protein alpha (SIRPalpha) regulates Fcgamma and complement receptor-mediated phagocytosis. J Exp Med 2001; 193:855-62; PMID:11283158;
    1. Jaiswal S, Jamieson CH, Pang WW, Park CY, Chao MP, Majeti R, Traver D, van Rooijen N, Weissman IL. CD47 is upregulated on circulating hematopoietic stem cells and leukemia cells to avoid phagocytosis. Cell 2009; 138:271-85; PMID:19632178;
    1. Chao MP, Jaiswal S, Weissman-Tsukamoto R, Alizadeh AA, Gentles AJ, Volkmer J, Weiskopf K, Willingham SB, Raveh T, Park CY, et al. . Calreticulin is the dominant pro-phagocytic signal on multiple human cancers and is counterbalanced by CD47. Sci Transl Med 2010; 2:63ra94; PMID:21178137
    1. Martins I, Kepp O, Galluzzi L, Senovilla L, Schlemmer F, Adjemian S, Menger L, Michaud M, Zitvogel L, Kroemer G. Surface-exposed calreticulin in the interaction between dying cells and phagocytes. Ann N Y Acad Sci 2010; 1209:77-82; PMID:20958319;
    1. Subramanian S, Parthasarathy R, Sen S, Boder ET, Discher DE. Species- and cell type-specific interactions between CD47 and human SIRPalpha. Blood 2006; 107:2548-56; PMID:16291597;
    1. Takenaka K, Prasolava TK, Wang JC, Mortin-Toth SM, Khalouei S, Gan OI, Dick JE, Danska JS. Polymorphism in Sirpa modulates engraftment of human hematopoietic stem cells. Nat Immunol 2007; 8:1313-23; PMID:17982459;
    1. Mehta K, Malavasi F. Human CD38 and related molecules. Basel: Karger, SAG; 2000.
    1. Schneider-Merck T, Lammerts van Bueren JJ, Berger S, Rossen K, van Berkel PH, Derer S, Beyer T, Lohse S, Bleeker WK, Peipp M, et al. . Human IgG2 antibodies against epidermal growth factor receptor effectively trigger antibody-dependent cellular cytotoxicity but, in contrast to IgG1, only by cells of myeloid lineage. J Immunol 2010; 184:512-20; PMID:19949082;
    1. Montalvao F, Garcia Z, Celli S, Breart B, Deguine J, Van Rooijen N, Bousso P. The mechanism of anti-CD20-mediated B cell depletion revealed by intravital imaging. J Clin Invest 2013; 123:5098-103; PMID:24177426;
    1. van der Veer MS, de Weers M, van Kessel B, Bakker JM, Wittebol S, Parren PW, Lokhorst HM, Mutis T. Towards effective immunotherapy of myeloma: enhanced elimination of myeloma cells by combination of lenalidomide with the human CD38 monoclonal antibody daratumumab. Haematologica 2011; 96:284-90; PMID:21109694;
    1. Gimsing P, Plesner T, Nahi H, Lokhorst H, Valentin M-L, Lisby S, Richardson PG. A phase I/II, dose-escalation study of daratumumab, a CD38 mab in patients with multiple myeloma - preliminary safety data. ASH Annu Meeting Abstr 2011; 118:1873.
    1. Plesner T, Lokhorst H, Gimsing P, Nahi H, Lisby S, Richardson PG. Daratumumab, a CD38 monoclonal antibody in patients with multiple myeloma - data from a dose-escalation phase I/II study. ASH Annu Meeting Abstr 2012; 120:73.
    1. van de Donk NW, Kamps S, Mutis T, Lokhorst HM. Monoclonal antibody-based therapy as a new treatment strategy in multiple myeloma. Leukemia 2012; 26:199-213; PMID:21852787;
    1. Ocio EM, Richardson PG, Rajkumar SV, Palumbo A, Mateos MV, Orlowski R, Kumar S, Usmani S, Roodman D, Niesvizky R, et al. . New drugs and novel mechanisms of action in multiple myeloma in 2013: a report from the International Myeloma Working Group (IMWG). Leukemia 2014; 28:525-42; PMID:24253022;
    1. Quach H, Ritchie D, Stewart AK, Neeson P, Harrison S, Smyth MJ, Prince HM. Mechanism of action of immunomodulatory drugs (IMiDS) in multiple myeloma. Leukemia 2010; 24:22-32; PMID:19907437;
    1. Grugan KD, McCabe FL, Kinder M, Greenplate AR, Harman BC, Ekert JE, van Rooijen N, Anderson GM, Nemeth JA, Strohl WR, et al. . Tumor-associated macrophages promote invasion while retaining Fc-dependent anti-tumor function. J Immunol 2012; 189:5457-66; PMID:23105143;
    1. Chao MP, Alizadeh AA, Tang C, Myklebust JH, Varghese B, Gill S, Jan M, Cha AC, Chan CK, Tan BT, et al. . Anti-CD47 antibody synergizes with rituximab to promote phagocytosis and eradicate non-Hodgkin lymphoma. Cell 2010; 142:699-713; PMID:20813259;
    1. Weiskopf K, Ring AM, Ho CC, Volkmer JP, Levin AM, Volkmer AK, Ozkan E, Fernhoff NB, van de Rijn M, Weissman IL, et al. . Engineered SIRPalpha variants as immunotherapeutic adjuvants to anticancer antibodies. Science 2013; 341:88-91; PMID:23722425;
    1. Zhao XW, van Beek EM, Schornagel K, Van der Maaden H, Van Houdt M, Otten MA, Finetti P, Van Egmond M, Matozaki T, Kraal G, et al. . CD47-signal regulatory protein-alpha (SIRPalpha) interactions form a barrier for antibody-mediated tumor cell destruction. Proc Natl Acad Sci U S A 2011; 108:18342-7; PMID:22042861;
    1. Kim D, Wang J, Willingham SB, Martin R, Wernig G, Weissman IL. Anti-CD47 antibodies promote phagocytosis and inhibit the growth of human myeloma cells. Leukemia 2012; 26:2538-45; PMID:22648449;
    1. Kuipers J, Vaandrager JW, Weghuis DO, Pearson PL, Scheres J, Lokhorst HM, Clevers H, Bast BJ. Fluorescence in situ hybridization analysis shows the frequent occurrence of 14q32.3 rearrangements with involvement of immunoglobulin switch regions in myeloma cell lines. Cancer Genet Cytogenet 1999; 109:99-107; PMID:10087940;
    1. Rozemuller H, van der Spek E, Bogers-Boer LH, Zwart MC, Verweij V, Emmelot M, Groen RW, Spaapen R, Bloem AC, Lokhorst HM, et al. . A bioluminescence imaging based in vivo model for preclinical testing of novel cellular immunotherapy strategies to improve the graft-versus-myeloma effect. Haematologica 2008; 93:1049-57; PMID:18492693;
    1. Burton DR, Pyati J, Koduri R, Sharp SJ, Thornton GB, Parren PW, Sawyer LS, Hendry RM, Dunlop N, Nara PL, et al. . Efficient neutralization of primary isolates of HIV-1 by a recombinant human monoclonal antibody. Science 1994; 266:1024-7; PMID:7973652;

Source: PubMed

3
Abonner