Combination of two novel blocking antibodies, anti-PD-1 antibody ezabenlimab (BI 754091) and anti-LAG-3 antibody BI 754111, leads to increased immune cell responses

Markus Zettl, Melanie Wurm, Otmar Schaaf, Sven Mostböck, Iñigo Tirapu, Ilse Apfler, Ivo C Lorenz, Lee Frego, Cynthia Kenny, Michael Thibodeau, Elisa Oquendo Cifuentes, Markus Reschke, Jürgen Moll, Norbert Kraut, Anne Vogt, Jonathon D Sedgwick, Irene C Waizenegger, Markus Zettl, Melanie Wurm, Otmar Schaaf, Sven Mostböck, Iñigo Tirapu, Ilse Apfler, Ivo C Lorenz, Lee Frego, Cynthia Kenny, Michael Thibodeau, Elisa Oquendo Cifuentes, Markus Reschke, Jürgen Moll, Norbert Kraut, Anne Vogt, Jonathon D Sedgwick, Irene C Waizenegger

Abstract

Upregulation of inhibitory receptors, such as lymphocyte activation gene-3 (LAG-3), may limit the antitumor activity of therapeutic antibodies targeting the programmed cell death protein-1 (PD-1) pathway. We describe the binding properties of ezabenlimab, an anti-human PD-1 antibody, and BI 754111, an anti-human LAG-3 antibody, and assess their activity alone and in combination. Ezabenlimab bound with high affinity to human PD-1 (KD = 6 nM) and blocked the interaction of PD-1 with PD-L1 and PD-L2. Ezabenlimab dose-dependently increased interferon-γ secretion in human T cells expressing PD-1 in co-culture with PD-L1-expressing dendritic cells. Administration of ezabenlimab to human PD-1 knock-in mice dose-dependently inhibited growth of MC38 tumors. To reduce immunogenicity, ezabenlimab was reformatted from a human IgG4 to a chimeric variant with a mouse IgG1 backbone (BI 905725) for further in vivo studies. Combining BI 905725 with anti-mouse LAG-3 antibodies improved antitumor activity versus BI 905725 monotherapy in the MC38 tumor model. We generated BI 754111, which bound with high affinity to human LAG-3 and prevented LAG-3 interaction with its ligand, major histocompatibility complex class II. In an in vitro model of antigen-experienced memory T cells expressing PD-1 and LAG-3, interferon-γ secretion increased by an average 1.8-fold versus isotype control (p = 0.027) with BI 754111 monotherapy, 6.9-fold (p < 0.0001) with ezabenlimab monotherapy and 13.2-fold (p < 0.0001) with BI 754111 plus ezabenlimab. Overall, ezabenlimab and BI 754111 bound to their respective targets with high affinity and prevented ligand binding. Combining ezabenlimab with BI 754111 enhanced in vitro activity versus monotherapy, supporting clinical investigation of this combination (NCT03156114; NCT03433898).

Keywords: BI 754091; BI 754111; LAG-3; PD-1; ezabenlimab.

Conflict of interest statement

No potential conflict of interest was reported by the authors.

© 2022 Boehringer Ingelheim RCV GmbH & Co KG. Published with license by Taylor & Francis Group, LLC.

Figures

Figure 1.
Figure 1.
Blockade of inhibitory PD-1/PD-L1 signaling by ezabenlimab leads to T-cell activation. Beads coated with PD-L1 protein and agonistic CD3 and CD28 antibodies were used to stimulate and simultaneously block activation of Jurkat cells expressing human PD-1. Dose titration with ezabenlimab blocked the inhibitory interaction leading to NFAT-mediated luciferase activity. Left panel (a) shows graphical description of the assay. Right panel (b) shows the assay results from one study (EC90 was calculated from three studies). Luciferase activity was calculated: 0% activation represents the luminescence value of cells incubated in the presence of anti-CD3/anti-CD28/PD-L1 beads without antibodies, and 100% activation represents the signal obtained with maximum luciferase levels in the presence of anti-CD3/anti-CD28/mouse IgG1 loaded control beads without antibodies present. In red, ezabenlimab mediated a dose-dependent increase in luciferase activity. In gray, titration of control IgG4 antibody did not affect luciferase activity.
Figure 2.
Figure 2.
Ezabenlimab treatment leads to tumor regression in the MC38 mouse tumor model. MC38 colon adenocarcinoma were subcutaneously grown in hPD-1 knock-in mice. Isotype control (a) or ezabenlimab (b–f) were administered by intraperitoneal injection once every 3 weeks (starting on d 6 after tumor cell injection). Tumor diameters were measured three times per week. TGI was calculated on d 25 (as indicated by the vertical red line) and the responding tumors were counted on d 36. Response was defined by a tumor size that was the same or smaller than the measurement at the start of treatment. Graphs show individual tumor volumes over time per treatment group and the table provides a summary of TGI and response for each dosage schedule.TGI, tumor growth inhibition; q21d, every 21 days.
Figure 3.
Figure 3.
Murine LAG-3 (mLAG-3) tool antibodies potently block the interaction of mLAG-3 and murine MHC-II. (a) 3A9 mouse T cell line expressing mLAG-3 (3A9-mLAG-3) was incubated with 20 µg/mL isotype control or 20 µg/mL C9B7W rat IgG1 anti-mLAG-3 antibody. Bound antibodies were detected by PE-labeled secondary antibody and the signal was detected by flow cytometry. The graph illustrates that a specific signal was seen only when cells were incubated with C9B7W confirming that it binds to mLAG-3 in this particular test. (b) 3A9-mLAG-3 expressing cells were incubated with 20 µg/mL isotype control or 20 µg/mL C9B7W in the presence of PE-labeled OVA323-339 MHC-II-IAb tetramer for 60 min, washed and fluorescent signals were detected by flow cytometry. Bound tetramers were observed only in the isotype treated control (left graph); presence of C9B7W blocked the interaction of these mouse Class II tetramers to mLAG-3 (right graph). (c) C9B7W (rat IgG1) and EX 80658 (C9B7W variant on mouse IgG1 D265A backbone) were tested for their binding properties to mouse LAG-3 expressed on a 3A9 mouse T cell line. Bound primary antibody was detected using either anti-mouse or anti-rat fluorescently labeled secondary antibody. After washing, the remaining signal was detected by flow cytometry and EC50 values were calculated. Left graph shows dose-dependent binding of antibodies to mLAG-3 as indicated by increasing MFI. The table provides an overview of the calculated EC50 values including the 95% CI for the two antibodies. CI, confidence interval; MHC-II, major histocompatibility complex class II; MFI, median fluorescent intensity.
Figure 4.
Figure 4.
In vivo combination of BI 905725 with anti-mLAG-3 treatment is more efficacious than BI 905725 therapy only. MC38 colon adenocarcinoma were subcutaneously grown in hPD-1 knock-in mice. Starting on d 6, animals were treated by intraperitoneal injection with isotype control (a), C9B7W (rat IgG1 anti-mLAG-3) (b), EX 80658 (mouse IgG1 D265A anti-mLAG-3) (c), BI 905725 (mouse IgG1 D265A anti-hPD-1) (d) or the combination of BI 905725 with either C9B7W (e) or EX 80658 (f). All antibodies were administered at a dose of 10 mg/kg twice weekly. Tumor diameters were measured three times per week. Responding tumors were counted on d 47. Graphs show individual tumor volumes over time per treatment group and the table provides a summary of response per treated group. Response was defined by a tumor size that was the same or smaller than the measurement at the start of treatment. TGI values were determined on d 21 because control tumors are required for calculation; long-term efficacy results are best described in this study by response and the individual tumor growth curves. hPD-1, human programmed cell death protein-1; mLAG-3, mouse lymphocyte activation gene-3, q3or4d, every 3 or 4 d.
Figure 5.
Figure 5.
BI 754111 potently blocks binding of soluble hLAG-3 to MHC class II expressing Raji cells. Recombinant human LAG-3 Fc chimera was incubated with serially diluted BI 754111 or isotype control. Raji cells expressing MHC class II were added. After incubation and washing, bound human LAG-3 was detected using fluorescently labeled anti-human IgG antibody using flow cytometry. For dose response curves and IC90 calculation, data were normalized to 0% inhibition (defined as the fluorescence value of cells incubated with the soluble hLAG-3 and no antibodies present) and 100% inhibition (defined as the signal obtained with cells without soluble hLAG-3 or antibodies). The graph shows BI 754111 dose-dependent inhibition of binding of soluble LAG-3 to Raji cells expressing MHC class II (one example from four donors). hLAG-3, human lymphocyte activation gene-3; Ig, immunoglobulin; MHC, major histocompatibility complex.
Figure 6.
Figure 6.
Blockade of inhibitory LAG-3/MHC class II signaling by BI 754111 leads to T-cell activation. (a) Detached LK35.2 cells were pre-incubated with HEL48-62 peptide. In parallel, a 3A9 mouse T cell line expressing human LAG-3 was incubated with serially diluted BI 754111 or isotype control. A co-culture of 3A9-hLAG-3: LK35.2 cells (ratio of 2:1) was set up and incubated for 24 hours. Thereafter the supernatant was harvested and mouse IL-2 concentrations were analyzed by ELISA. EC50 was calculated by data transformation; 0% activation was defined as IL-2 secretion of cells incubated with assay medium only and 100% activation was defined as IL-2 secretion of cells incubated with the highest concentration of the reference tool anti-human LAG-3 antibody. (b) PBMCs from healthy human donors who had recently been vaccinated against tetanus were cultivated in the presence of the antigen and T cells were expanded for 13 d. In parallel, monocyte derived DCs were generated and loaded with tetanus antigen. T cell culture and antigen-loaded DCs were co-cultured for one day and on the next day freshly antigen-loaded DCs were added a second time together with antibodies. After 4 d of treatment, supernatants were analyzed for IFN-γ secretion by ELISA (see the scheme in Supplementary Fig. S1). Left graph shows results from one donor: a dose-dependent increase of IFN-γ secretion following treatment with ezabenlimab (blue curve), BI 754111 (orange curve) or ezabenlimab in combination with BI 754111 (green curve). Right graph shows the data from four donors treated with ezabenlimab, BI 754111 or their combination. APC, antigen-presenting cell; DCs, dendritic cells; IFN, interferon; IL, interleukin; MHC-II, major histocompatibility complex class II; LAG-3, lymphocyte activation gene-3; PBMCs, peripheral blood mononuclear cells; SD, standard deviation.

References

    1. Nishimura H, Nose M, Hiai H, Minato N, Honjo T.. Development of lupus-like autoimmune diseases by disruption of the PD-1 gene encoding an ITIM motif-carrying immunoreceptor. Immunity. 1999;11(2):141–13. doi:10.1016/s1074-7613(00)80089-8.
    1. Freeman GJ, Long AJ, Iwai Y, Bourque K, Chernova T, Nishimura H, Fitz LJ, Malenkovich N, Okazaki T, Byrne MC, et al. Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J Exp Med. 2000;192(7):1027–1034. doi:10.1084/jem.192.7.1027.
    1. Wei SC, Duffy CR, Allison JP. Fundamental mechanisms of immune checkpoint blockade therapy. Cancer Discov. 2018;8(9):1069–1086. doi:10.1158/-18-0367.
    1. McDermott DF, Atkins MB. PD −1 as a potential target in cancer therapy. Cancer Med. 2013;2(5):662–673. doi:10.1002/cam4.106.
    1. Iwai Y, Ishida M, Tanaka Y, Okazaki T, Honjo T, Minato N. Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade. Proc Natl Acad Sci U S A. 2002;99(19):12293–12297. doi:10.1073/pnas.192461099.
    1. Iwai Y. PD-1 blockade inhibits hematogenous spread of poorly immunogenic tumor cells by enhanced recruitment of effector T cells. Int Immunol. 2005;17(2):133–144. doi:10.1093/intimm/dxh194.
    1. Strome SE, Dong H, Tamura H, Voss SG, Flies DB, Tamada K, Salomao D, Cheville J, Hirano F, Lin W, et al. B7-H1 blockade augments adoptive T-cell immunotherapy for squamous cell carcinoma. Cancer Res. 2003;63(19):6501–6505.
    1. Hirano F, Kaneko K, Tamura H, Dong H, Wang S, Ichikawa M, Rietz C, Flies DB, Lau JS, Zhu G, et al. Blockade of B7-H1 and PD-1 by monoclonal antibodies potentiates cancer therapeutic immunity. Cancer Res. 2005;65(3):1089–1096.
    1. Ribas A, Wolchok JD. Cancer immunotherapy using checkpoint blockade. Science. 2018;359(6382):1350–1355. doi:10.1126/science.aar4060.
    1. Sade-Feldman M, Jiao YJ, Chen JH, Rooney MS, Barzily-Rokni M, Eliane J-P, Bjorgaard SL, Hammond MR, Vitzthum H, Blackmon SM, et al. Resistance to checkpoint blockade therapy through inactivation of antigen presentation. Nature Commun. 2017;8(1):1136. doi:10.1038/s41467-017-01062-w.
    1. Shin DS, Zaretsky JM, Escuin-Ordinas H, Garcia-Diaz A, Hu-Lieskovan S, Kalbasi A, Grasso CS, Hugo W, Sandoval S, Torrejon DY, et al. Primary resistance to PD-1 blockade mediated by JAK1/2 mutations. Cancer Discov. 2017;7(2):188–201. doi:10.1158/-16-1223%JCancerDiscovery.
    1. Zaretsky JM, Garcia-Diaz A, Shin DS, Escuin-Ordinas H, Hugo W, Hu-Lieskovan S, Torrejon DY, Abril-Rodriguez G, Sandoval S, Barthly L, et al. Mutations associated with acquired resistance to PD-1 blockade in melanoma. New Engl J Med. 2016;375(9):819–829. doi:10.1056/nejmoa1604958.
    1. Qin S, Xu L, Yi M, Yu S, Wu K, Luo S. Novel immune checkpoint targets: moving beyond PD-1 and CTLA-4. Mol Cancer. 2019;18(1):155. doi:10.1186/s12943-019-1091-2.
    1. Matsuzaki J, Gnjatic S, Mhawech-Fauceglia P, Beck A, Miller A, Tsuji T, Eppolito C, Qian F, Lele S, Shrikant P, et al. Tumor-infiltrating NY-ESO-1–specific CD8 + T cells are negatively regulated by LAG-3 and PD-1 in human ovarian cancer. Proc Natl Acad Sci USA. 2010;107(17):7875–7880. doi:10.1073/pnas.1003345107.
    1. Woo SR, Turnis ME, Goldberg MV, Bankoti J, Selby M, Nirschl CJ, Bettini ML, Gravano DM, Vogel P, Liu CL, et al. Immune inhibitory molecules LAG-3 and PD-1 synergistically regulate T-cell function to promote tumoral immune escape. Cancer Res. 2012;72(4):917–927. doi:10.1158/0008-5472.Can-11-1620.
    1. Maruhashi T, Sugiura D, Okazaki IM, Okazaki T. LAG-3: from molecular functions to clinical applications. J Immunother Cancer. 2020;8(2):e001014. doi:10.1136/jitc-2020-001014.
    1. Gestermann N, Saugy D, Martignier C, Tillé L, Fuertes Marraco SA, Zettl M, Tirapu I, Speiser DE, Verdeil G. LAG-3 and PD-1+LAG-3 inhibition promote anti-tumor immune responses in human autologous melanoma/T cell co-cultures. Oncoimmunology. 2020;9(1):1736792. doi:10.1080/2162402x.2020.1736792.
    1. Workman CJ, Dugger KJ, Vignali DA. Cutting edge: molecular analysis of the negative regulatory function of lymphocyte activation gene-3. J Immunol. 2002;169(10):5392–5395. doi:10.4049/jimmunol.169.10.5392.
    1. Silva JP, Vetterlein O, Jose J, Peters S, Kirby H. The S228P mutation prevents in vivo and in vitro IgG4 Fab-arm exchange as demonstrated using a combination of novel quantitative immunoassays and physiological matrix preparation. J Biol Chem. 2015;290(9):5462–5469. doi:10.1074/jbc.M114.600973.
    1. Wang F, Tsai JC, Davis JH, Chau B, Dong J, West SM, Hogan JM, Wheeler ML, Bee C, Morishige W, et al. Design and characterization of mouse IgG1 and IgG2a bispecific antibodies for use in syngeneic models. MAbs. 2020;12(1):1685350. doi:10.1080/19420862.2019.1685350.
    1. Dohlsten M, Hedlund G, Segren S, Lando PA, Herrmann T, Kelly AP, Kalland T. Human major histocompatibility complex class II-negative colon carcinoma cells present staphylococcal superantigens to cytotoxic T lymphocytes: evidence for a novel enterotoxin receptor. Eur J Immunol. 1991;21(5):1229–1233. doi:10.1002/eji.1830210520.
    1. Niehrs A, Garcia-Beltran WF, Norman PJ, Watson GM, Holzemer A, Chapel A, Richert L, Pommerening-Roser A, Korner C, Ozawa M, et al. A subset of HLA-DP molecules serve as ligands for the natural cytotoxicity receptor NKp44. Nat Immunol. 2019;20(9):1129–1137. doi:10.1038/s41590-019-0448-4.
    1. Huang RY, Francois A, McGray AR, Miliotto A, Odunsi K. Compensatory upregulation of PD-1, LAG-3, and CTLA-4 limits the efficacy of single-agent checkpoint blockade in metastatic ovarian cancer. Oncoimmunology. 2017;6(1):e1249561. doi:10.1080/2162402x.2016.1249561.
    1. Workman CJ, Rice DS, Dugger KJ, Kurschner C, Vignali DA. Phenotypic analysis of the murine CD4-related glycoprotein, CD223 (LAG-3). Eur J Immunol. 2002;32(8):2255–2263. doi:10.1002/1521-4141(200208)32:8<2255::AID-IMMU2255>;2-A.
    1. Haines BB, Javaid S, Cui L, Hirsch H, Cemerski S, McClanahan T, Sathe M, Zhang S, Rosenzweig M, et al. Blockade of LAG-3 amplifies immune activation signatures and augments curative antitumor responses to anti-PD-1 therapy in immune competent mouse models of cancer. Cancer Res. 2017;77:Abstr4714. doi:10.1158/1538-7445.AM2017-4714.
    1. Burova E, Hermann A, Dai J, Ullman E, Halasz G, Potocky T, Hong S, Liu M, Allbritton O, Woodruff A, et al. Preclinical development of the anti-LAG-3 antibody REGN3767: characterization and activity in combination with the anti-PD-1 antibody cemiplimab in human PD-1xLAG-3 –Knockin Mice. Mol Cancer Ther. 2019;18(11):2051–2062. doi:10.1158/1535-7163.MCT-18-1376.
    1. Ghosh S, Sharma G, Travers J, Kumar S, Choi J, Jun HT, Kehry M, Ramaswamy S, Jenkins D. TSR-033, a novel therapeutic antibody targeting LAG-3, enhances T-cell function and the activity of PD-1 blockade In Vitro and In Vivo. Mol Cancer Ther. 2019;18(3):632–641. doi:10.1158/1535-7163.Mct-18-0836.
    1. Kouo T, Huang L, Pucsek AB, Cao M, Solt S, Armstrong T, Jaffee E. Galectin-3 shapes antitumor immune responses by suppressing CD8 + T cells via LAG-3 and inhibiting expansion of plasmacytoid dendritic cells. Cancer Immunol Res. 2015;3(4):412–423. doi:10.1158/2326-6066.Cir-14-0150.
    1. Xu F, Liu J, Liu D, Liu B, Wang M, Hu Z, Du X, Tang L, He F. LSECtin expressed on melanoma cells promotes tumor progression by inhibiting antitumor T-cell responses. Cancer Res. 2014;74(13):3418–3428. doi:10.1158/0008-5472.Can-13-2690.
    1. Wang J, Sanmamed MF, Datar I, Su TT, Ji L, Sun J, Chen L, Chen Y, Zhu G, Yin W, et al. Fibrinogen-like protein 1 is a major immune inhibitory ligand of LAG-3. Cell. 2019;176(1–2):334–347.e312. doi:10.1016/j.cell.2018.11.010.
    1. Bendell J, Ulahannan SV, Chu Q, Patel M, George B, Landsberg R, Elgadi M, Duffy C, Graeser R, et al. A phase I, dose finding study of BI 754111, an anti-LAG-3 antibody, in combination with BI 754091, an anti-PD-1 antibody, in patients with advanced solid tumors: preliminary results from the microsatellite stable (MSS) metastatic colorectal cancer (mCRC) cohort. Mol Cancer Ther. 2019;18:AbstrC027. doi:10.1158/1535-7163.TARG-19-C027.
    1. Yamaguchi K, Kang Y-K, D-Y O, Kondo S, Rha S, Kuboki Y, Morimoto M, Hara H, Lin -C-C, Tachibana Y, et al. Phase I study of BI 754091 plus BI 754111 in Asian patients with gastric/gastroesophageal junction or esophageal cancer. J Clin Oncol. 2021;39(3_suppl):Abstr212. doi:10.1200/JCO.2021.39.3_suppl.212.
    1. Hong DS, Patrick S, Aitana C, John S, María ODO, Carvajal RD, Amy P, Chrisann K, Taito E, et al. Phase I/II study of LAG525 ± spartalizumab (PDR001) in patients (pts) with advanced malignancies. J Clin Oncol. 2018;36(15_suppl):3012. doi:10.1200/JCO.2018.36.15_suppl.3012.
    1. Ascierto PA, Melero I, Bhatia S, Bono P, Sanborn RE, Lipson EJ, Callahan MK, Gajewski T, Gomez-Roca CA, Hodi FS, et al. Initial efficacy of anti-lymphocyte activation gene-3 (anti–LAG-3; BMS-986016) in combination with nivolumab (nivo) in pts with melanoma (MEL) previously treated with anti–PD-1/PD-L1 therapy. J Clin Oncol. 2017;35(15_suppl):Abstr9520. doi:10.1200/JCO.2017.35.15_suppl.9520.
    1. Lipson EJ, Tawbi HAH, Schadendorf D, Ascierto PA, Matamala LA, Gutiérrez EC, Rutkowski P, Gogas H, Lao CD, Janoski de Menezes J, et al. Relatlimab (RELA) plus nivolumab (NIVO) versus NIVO in first-line advanced melanoma: primary phase III results from RELATIVITY-047 (CA224-047). J Clin Oncol. 2021;39(Suppl 15_suppl):Abstr9503. doi:10.1200/JCO.2021.39.15_suppl.9503.

Source: PubMed

3
Se inscrever