CD47xCD19 bispecific antibody triggers recruitment and activation of innate immune effector cells in a B-cell lymphoma xenograft model

Xavier Chauchet, Laura Cons, Laurence Chatel, Bruno Daubeuf, Gérard Didelot, Valéry Moine, Didier Chollet, Pauline Malinge, Guillemette Pontini, Krzysztof Masternak, Walter Ferlin, Vanessa Buatois, Limin Shang, Xavier Chauchet, Laura Cons, Laurence Chatel, Bruno Daubeuf, Gérard Didelot, Valéry Moine, Didier Chollet, Pauline Malinge, Guillemette Pontini, Krzysztof Masternak, Walter Ferlin, Vanessa Buatois, Limin Shang

Abstract

Background: CD47/SIRPα axis is recognized as an innate immune checkpoint and emerging clinical data validate the interest of interrupting this pathway in cancer, particularly in hematological malignancies. In preclinical models, CD47/SIRPα blocking agents have been shown to mobilize phagocytic cells and trigger adaptive immune responses to eliminate tumors. Here, we describe the mechanisms afforded by a CD47xCD19 bispecific antibody (NI-1701) at controlling tumor growth in a mouse xenograft B-cell lymphoma model.

Methods: The contribution of immune effector cell subsets behind the antitumor activity of NI-1701 was investigated using flow cytometry, transcriptomic analysis, and in vivo immune-cell depletion experiments.

Results: We showed that NI-1701 treatment transformed the tumor microenvironment (TME) into a more anti-tumorigenic state with increased NK cells, monocytes, dendritic cells (DC) and MHCIIhi tumor-associated macrophages (TAMs) and decreased granulocytic myeloid-derived suppressor cells. Notably, molecular analysis of isolated tumor-infiltrating leukocytes following NI-1701 administration revealed an upregulation of genes linked to immune activation, including IFNγ and IL-12b. Moreover, TAM-mediated phagocytosis of lymphoma tumor cells was enhanced in the TME in the presence of NI-1701, highlighting the role of macrophages in tumor control. In vivo cell depletion experiments demonstrated that both macrophages and NK cells contribute to the antitumor activity. In addition, NI-1701 enhanced dendritic cell-mediated phagocytosis of tumor cells in vitro, resulting in an increased cross-priming of tumor-specific CD8 T cells.

Conclusions: The study described the mechanisms afforded by the CD47xCD19 bispecific antibody, NI-1701, at controlling tumor growth in lymphoma mouse model. NI-1701 is currently being evaluated in a Phase I clinical trial for the treatment of refractory or relapsed B-cell lymphoma (NCT04806035).

Keywords: B-cell lymphoma; Bispecific antibody; CD47; Cancer immunotherapy; Dendritic cells; Macrophages; NK cells; Tumor microenvironment.

Conflict of interest statement

All authors are current or former employees of Light Chain Bioscience/Novimmune SA, excepted DC. GD is presently employee of Ichnos Sciences Biotherapeutics SA. All the authors declare that their employment does not alter their adherence to the policies of Cancer Immunology, Immunotherapy journal.

© 2022. The Author(s).

Figures

Fig. 1
Fig. 1
NI-1701 modulates the tumor microenvironment by promoting accumulation of immune cells associated with anti-tumorigenic functions and triggers enhanced engulfment of tumor cells by TAMs and monocytes. Immunodeficient NOD SCID mice were implanted with Raji GFPhi lymphoma tumor cells and treatment was initiated when tumor volume reached 100 mm3. Tumors were excised 14 and 25 days after treatment initiation to evaluate impact of NI-1701 on the tumor microenvironment. a Design of the experiment. b Mean tumor volume at days 14 and 25 after treatment with human IgG1 isotype control Ab or NI-1701. Data are presented as mean ± SD, with n = 7–9 mice per group. c Analysis by flow cytometry of tumor-infiltrating total mouse leukocytes, myeloid cell subsets (TAMs, monocytes, G-MDSCs, dendritic cells) and NK cells. Gating strategy to identify subpopulations in the TME is displayed in Additional file 1: Fig. S1. Data are presented as mean ± SD, with n = 4–9 mice per group. d Analysis by flow cytometry of MHCII+ TAMs. e Representative flow cytometry plots of phagocytic TAMs or monocytes, determined as F4/80+GFP+ or Ly6C+GFP+ respectively, from Raji GFPhi tumors dissected 14 days after treatment initiation. The threshold of GFP positivity and GFP internalization in TAMs and monocytes were determined based on methods and analysis depicted in supplementary experimental procedures. f The percentage of phagocytosis of GFPhi Raji tumor cells by TAMs and monocytes, assessed at day 14 and day 25 post-treatment initiation, is shown for each individual mouse. Data are presented as mean ± SD, with n = 4–9 mice per group. Significance was determined by unpaired t test. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001
Fig. 2
Fig. 2
Targeted transcriptomic analysis of tumor-infiltrating mouse leukocytes reveals upregulation of proinflammatory cytokine and chemokine genes upon NI-1701 treatment. Mouse CD45+ tumor-infiltrating leukocytes were sorted from tumors 14 days after treatment initiation with NI-1701 or hIgG1 control and submitted to Nanostring transcriptomic analysis using the mouse Myeloid Innate Immunity panel (n = 4 mice per group) a Volcano plot of differentially expressed genes in tumor-infiltrating leukocytes between hIgG1 versus NI-1701. Vertical grey lines correspond to absolute log2 fold change > 0.76 (i.e., fold-change > 1.7) and horizontal dot line correspond to a p value of p = 0.05. Grey zone corresponds to statistically non-significant changes and red and blue dots to significantly upregulated and downregulated genes, respectively. b Heatmap depicting row z scores of normalized mRNA count of significantly up-regulated and down-regulated genes with fold change > 1.7
Fig. 3
Fig. 3
Both macrophages and NK cells are important effector cells for NI-1701 antitumor efficacy in Raji lymphoma xenograft model. a Design of the macrophage depletion experiment. b Tumor volume follow-up of CB17-SCID mice carrying Raji tumors treated with i.p. injection of NI-1701 and i.v. injection of clodronate liposomes. n = 5 mice per group. c Design of the NK cell depletion experiment. d Tumor volume follow-up of NOD SCID mice carrying Raji tumors treated with i.v. injection of NI-1701 and i.p. injection of anti-Asialo GM1 antibody. n = 8 mice per group. For b and d mean ± SEM is shown. Significance was determined at endpoint by one-way ANOVA test. *p < 0.05, ***p < 0.001, ****p < 0.0001
Fig. 4
Fig. 4
NI-1701 induces tumor cell phagocytosis by bone-marrow-derived dendritic cells and promotes cross-priming of CD8+ T cells. Bone marrow-derived mouse dendritic cells (CD11c+) from BALB/c mice were cocultured for 2 h 30 (a) or 24 h (b) with Raji GFPhi tumor cells (1:1 ratio) in the presence of hIgG1 or NI-1701. Representative flow cytometry plots for hIgG1 and NI-1701 treated groups are depicted, and the percentage of phagocytosis is indicated (left panel). Phagocytic events were confirmed by imaging flow cytometry acquisition in the CD11c+GFP+ gate with tumor cells in green fluorescence and CD11c+ dendritic cells in red fluorescence (middle panel). The mean percentage of phagocytosis at 2 h 30 ± SD is shown (right panel), n = 5 independent experiments. b Percentage of residual Raji GFPhi tumor cells after 24 h of coculture with CD11c+ DCs was determined by the analysis of remaining GFP+ tumor cells within the total live cell gate. n = 5 experiments. c Bone-marrow derived dendritic cells from BALB/c mice were cocultured overnight with hemagglutinin-expressing Raji tumor cells (Raji HA-GFP) in the presence of human IgG1 or NI-1701 (ratio 1:1). The next day, HA-specific CD8+ T cells from CL-4 transgenic mice (bearing HA-specific TCR on CD8+ T cells) labelled with CellTrace violet were added (ratio 1:5). Analysis of CD8+ T cell proliferation was performed 3 days later. Plots represent an illustration of proliferating CD8+ T cells (left panel). Mean ± SD was calculated from 4 independent experiments (right panel). Significance was determined by unpaired t test. *p < 0.05, ****p < 0.0001

References

    1. Engblom C, Pfirschke C, Pittet MJ. The role of myeloid cells in cancer therapies. Nat Rev Cancer. 2016;16:447–462. doi: 10.1038/nrc.2016.54.
    1. Cassetta L, Pollard JW. Targeting macrophages: therapeutic approaches in cancer. Nat Rev Drug Discov. 2018 doi: 10.1038/nrd.2018.169.
    1. DeNardo DG, Ruffell B. Macrophages as regulators of tumour immunity and immunotherapy. Nat Rev Immunol. 2019 doi: 10.1038/s41577-019-0127-6.
    1. Beatty GL, Li Y, Long KB. Cancer immunotherapy: activating innate and adaptive immunity through CD40 agonists. Expert Rev Anticancer Ther. 2017;17:175–186. doi: 10.1080/14737140.2017.1270208.
    1. Georgoudaki A-M, Prokopec KE, Boura VF, et al. Reprogramming tumor-associated macrophages by antibody targeting inhibits cancer progression and metastasis. Cell Rep. 2016;15:2000–2011. doi: 10.1016/j.celrep.2016.04.084.
    1. Guerriero JL, Sotayo A, Ponichtera HE, et al. Class IIa HDAC inhibition reduces breast tumours and metastases through anti-tumour macrophages. Nature. 2017;543:428–432. doi: 10.1038/nature21409.
    1. Germano G, Frapolli R, Belgiovine C, et al. Role of macrophage targeting in the antitumor activity of trabectedin. Cancer Cell. 2013;23:249–262. doi: 10.1016/j.ccr.2013.01.008.
    1. Cannarile MA, Weisser M, Jacob W, et al. Colony-stimulating factor 1 receptor (CSF1R) inhibitors in cancer therapy. J Immunother Cancer. 2017;5:53. doi: 10.1186/s40425-017-0257-y.
    1. Logtenberg MEW, Scheeren FA, Schumacher TN. The CD47-SIRPα immune checkpoint. Immunity. 2020;52:742–752. doi: 10.1016/j.immuni.2020.04.011.
    1. Deuse T, Hu X, Agbor-Enoh S, et al. The SIRPα–CD47 immune checkpoint in NK cells. J Exp Med. 2021;218(3):e20200839. doi: 10.1084/jem.20200839.
    1. Oldenborg P-A. CD47: a cell surface glycoprotein which regulates multiple functions of hematopoietic cells in health and disease. ISRN Hematol. 2013;2013:1–19. doi: 10.1155/2013/614619.
    1. Chao MP, Alizadeh AA, Tang C, et al. Anti-CD47 antibody synergizes with rituximab to promote phagocytosis and eradicate non-hodgkin lymphoma. Cell. 2010;142:699–713. doi: 10.1016/j.cell.2010.07.044.
    1. Majeti R, Chao MP, Alizadeh AA, et al. CD47 is an adverse prognostic factor and therapeutic antibody target on human acute myeloid leukemia stem cells. Cell. 2009;138:286–299. doi: 10.1016/j.cell.2009.05.045.
    1. Willingham SB, Volkmer J-P, Gentles AJ, et al. The CD47-signal regulatory protein alpha (SIRPa) interaction is a therapeutic target for human solid tumors. Proc Natl Acad Sci. 2012;109:6662–6667. doi: 10.1073/pnas.1121623109.
    1. Jalil AR, Andrechak JC, Discher DE. Macrophage checkpoint blockade: results from initial clinical trials, binding analyses, and CD47-SIRPα structure–function. Antib Ther. 2020;3:80–94. doi: 10.1093/abt/tbaa006.
    1. Advani R, Flinn I, Popplewell L, et al. CD47 blockade by Hu5F9-G4 and rituximab in non-Hodgkin’s lymphoma. N Engl J Med. 2018;379:1711–1721. doi: 10.1056/NEJMoa1807315.
    1. Sallman DA, Donnellan WB, Asch AS, et al. The first-in-class anti-CD47 antibody Hu5F9-G4 is active and well tolerated alone or with azacitidine in AML and MDS patients: initial phase 1b results. J Clin Oncol. 2019;37:7009–7009. doi: 10.1200/JCO.2019.37.15_suppl.7009.
    1. Ansell SM, Maris MB, Lesokhin AM, et al. Phase I study of the CD47 blocker TTI-621 in patients with relapsed or refractory hematologic malignancies. Clin Cancer Res. 2021;27:2190–2199. doi: 10.1158/1078-0432.CCR-20-3706.
    1. Liu J, Wang L, Zhao F, et al. Pre-clinical development of a humanized anti-CD47 antibody with anti-cancer therapeutic potential. PLoS ONE. 2015;10:e0137345. doi: 10.1371/journal.pone.0137345.
    1. Kauder SE, Kuo TC, Harrabi O, et al. ALX148 blocks CD47 and enhances innate and adaptive antitumor immunity with a favorable safety profile. PLoS ONE. 2018;13:e0201832. doi: 10.1371/journal.pone.0201832.
    1. Peluso MO, Adam A, Armet CM, et al. The Fully human anti-CD47 antibody SRF231 exerts dual-mechanism antitumor activity via engagement of the activating receptor CD32a. J Immunother Cancer. 2020;8:e000413. doi: 10.1136/jitc-2019-000413.
    1. Sikic BI, Lakhani N, Patnaik A, et al. First-in-human, first-in-class phase i trial of the anti-CD47 antibody Hu5F9-G4 in patients with advanced cancers. J Clin Oncol. 2019 doi: 10.1200/JCO.18.02018.
    1. Dheilly E, Moine V, Broyer L, et al. Selective blockade of the ubiquitous checkpoint receptor CD47 is enabled by dual-targeting bispecific antibodies. Mol Ther J Am Soc Gene Ther. 2017;25:523–533. doi: 10.1016/j.ymthe.2016.11.006.
    1. Buatois V, Johnson Z, Salgado-Pires S, et al. Preclinical development of a bispecific antibody that safely and effectively targets CD19 and CD47 for the treatment of B-cell lymphoma and leukemia. Mol Cancer Ther. 2018;17:1739–1751. doi: 10.1158/1535-7163.MCT-17-1095.
    1. Morgan DJ, Liblau R, Scott B, et al. (1996) CD8(+) T cell-mediated spontaneous diabetes in neonatal mice. J Immunol Baltim Md. 1950;157:978–983.
    1. Shultz LD, Schweitzer PA, Christianson SW, et al. (1995) Multiple defects in innate and adaptive immunologic function in NOD/LtSz-scid mice. J Immunol Baltim Md. 1950;154:180–191.
    1. Takenaka K, Prasolava TK, Wang JCY, et al. Polymorphism in Sirpa modulates engraftment of human hematopoietic stem cells. Nat Immunol. 2007;8:1313–1323. doi: 10.1038/ni1527.
    1. Liu X, Pu Y, Cron K, et al. CD47 blockade triggers T cell-mediated destruction of immunogenic tumors. Nat Med. 2015;21:1209–1215. doi: 10.1038/nm.3931.
    1. Tokunaga R, Zhang W, Naseem M, et al. CXCL9, CXCL10, CXCL11/CXCR3 axis for immune activation—a target for novel cancer therapy. Cancer Treat Rev. 2018;63:40–47. doi: 10.1016/j.ctrv.2017.11.007.
    1. Yin B, Loike JD, Kako Y, et al. Lipoprotein lipase regulates Fc receptor-mediated phagocytosis by macrophages maintained in glucose-deficient medium. J Clin Invest. 1997;100:649–657. doi: 10.1172/JCI119576.
    1. Rabold K, Netea MG, Adema GJ, Netea-Maier RT. Cellular metabolism of tumor-associated macrophages—functional impact and consequences. FEBS Lett. 2017;591:3022–3041. doi: 10.1002/1873-3468.12771.
    1. Hu Z, Van Rooijen N, Yang Y-G. Macrophages prevent human red blood cell reconstitution in immunodeficient mice. Blood. 2011;118:5938–5946. doi: 10.1182/blood-2010-11-321414.
    1. Tseng D, Volkmer J-P, Willingham SB, et al. Anti-CD47 antibody-mediated phagocytosis of cancer by macrophages primes an effective antitumor T-cell response. Proc Natl Acad Sci. 2013;110:11103–11108. doi: 10.1073/pnas.1305569110.
    1. Sockolosky JT, Dougan M, Ingram JR, et al. Durable antitumor responses to CD47 blockade require adaptive immune stimulation. Proc Natl Acad Sci. 2016;113:E2646–E2654. doi: 10.1073/pnas.1604268113.
    1. Xu MM, Pu Y, Han D, et al. Dendritic cells but not macrophages sense tumor mitochondrial DNA for cross-priming through signal regulatory protein α signaling. Immunity. 2017;47:363–373.e5. doi: 10.1016/j.immuni.2017.07.016.
    1. Gauttier V, Pengam S, Durand J, et al. Selective SIRPα blockade reverses tumor T cell exclusion and overcomes cancer immunotherapy resistance. J Clin Invest. 2020 doi: 10.1172/JCI135528.
    1. Puro RJ, Bouchlaka MN, Hiebsch RR, et al. Development of AO-176, a next-generation humanized anti-CD47 antibody with novel anticancer properties and negligible red blood cell binding. Mol Cancer Ther. 2020;19:835–846. doi: 10.1158/1535-7163.MCT-19-1079.
    1. Meng Z, Wang Z, Guo B, et al. TJC4, a differentiated anti-CD47 antibody with novel epitope and RBC sparing properties. Blood. 2019;134:4063–4063. doi: 10.1182/blood-2019-122793.
    1. Hendriks MAJM, Ploeg EM, Koopmans I, et al. Bispecific antibody approach for EGFR-directed blockade of the CD47-SIRPα “don’t eat me” immune checkpoint promotes neutrophil-mediated trogoptosis and enhances antigen cross-presentation. OncoImmunology. 2020;9:1824323. doi: 10.1080/2162402X.2020.1824323.
    1. Pahl J, Cerwenka A. Tricking the balance: NK cells in anti-cancer immunity. Immunobiology. 2017;222:11–20. doi: 10.1016/j.imbio.2015.07.012.
    1. Ma Y, Shurin GV, Peiyuan Z, Shurin MR. Dendritic cells in the cancer microenvironment. J Cancer. 2013;4:36–44. doi: 10.7150/jca.5046.
    1. Hoves S, Ooi C-H, Wolter C, et al. Rapid activation of tumor-associated macrophages boosts preexisting tumor immunity. J Exp Med. 2018;215:859–876. doi: 10.1084/jem.20171440.
    1. Ito M. NOD/SCID/gamma cnull mouse: an excellent recipient mouse model for engraftment of human cells. Blood. 2002;100:3175–3182. doi: 10.1182/blood-2001-12-0207.
    1. Veglia F, Sanseviero E, Gabrilovich DI. Myeloid-derived suppressor cells in the era of increasing myeloid cell diversity. Nat Rev Immunol. 2021;21:485–498. doi: 10.1038/s41577-020-00490-y.
    1. Boada-Romero E, Martinez J, Heckmann BL, Green DR. The clearance of dead cells by efferocytosis. Nat Rev Mol Cell Biol. 2020;21:398–414. doi: 10.1038/s41580-020-0232-1.
    1. Grugan KD, McCabe FL, Kinder M, et al. Tumor-associated macrophages promote invasion while retaining Fc-dependent anti-tumor function. J Immunol. 2012;189:5457–5466. doi: 10.4049/jimmunol.1201889.
    1. Leidi M, Gotti E, Bologna L, et al. M2 macrophages phagocytose rituximab-opsonized leukemic targets more efficiently than M1 cells in vitro. J Immunol. 2009;182:4415–4422. doi: 10.4049/jimmunol.0713732.
    1. Lin GHY, Chai V, Lee V, et al. TTI-621 (SIRPαFc), a CD47-blocking cancer immunotherapeutic, triggers phagocytosis of lymphoma cells by multiple polarized macrophage subsets. PLoS ONE. 2017;12:e0187262. doi: 10.1371/journal.pone.0187262.
    1. Zhang M, Hutter G, Kahn SA, et al. Anti-CD47 treatment stimulates phagocytosis of glioblastoma by M1 and M2 polarized macrophages and promotes M1 polarized macrophages in vivo. PLoS ONE. 2016;11:e0153550. doi: 10.1371/journal.pone.0153550.
    1. Hutter G, Theruvath J, Graef CM, et al. Microglia are effector cells of CD47-SIRPα antiphagocytic axis disruption against glioblastoma. Proc Natl Acad Sci. 2019 doi: 10.1073/pnas.1721434116.
    1. Chang CL, Garcia-Arcos I, Nyrén R, et al. Lipoprotein lipase deficiency impairs bone marrow myelopoiesis and reduces circulating monocyte levels. Arterioscler Thromb Vasc Biol. 2018;38:509–519. doi: 10.1161/ATVBAHA.117.310607.
    1. Ring NG, Herndler-Brandstetter D, Weiskopf K, et al. Anti-SIRPα antibody immunotherapy enhances neutrophil and macrophage antitumor activity. Proc Natl Acad Sci. 2017;114:E10578–E10585. doi: 10.1073/pnas.1710877114.
    1. Chowdhury S, Castro S, Coker C, et al. Programmable bacteria induce durable tumor regression and systemic antitumor immunity. Nat Med. 2019;25:1057–1063. doi: 10.1038/s41591-019-0498-z.
    1. Liu X, Liu L, Ren Z, et al. Dual targeting of innate and adaptive checkpoints on tumor cells limits immune evasion. Cell Rep. 2018;24:2101–2111. doi: 10.1016/j.celrep.2018.07.062.
    1. Hanke N, Alizadeh D, Katsanis E, Larmonier N. Dendritic cell tumor killing activity and its potential applications in cancer immunotherapy. Crit Rev Immunol. 2013;33:1–21. doi: 10.1615/CritRevImmunol.2013006679.
    1. Dheilly E, Majocchi S, Moine V, et al. Tumor-directed blockade of CD47 with bispecific antibodies induces adaptive antitumor immunity. Antibodies. 2018;7:3. doi: 10.3390/antib7010003.

Source: PubMed

3
Tilaa