Nonclinical Characterization of Bexmarilimab, a Clever-1-Targeting Antibody for Supporting Immune Defense Against Cancers

Maija Hollmén, Mikael Maksimow, Jenna H Rannikko, Matti K Karvonen, Marita Vainio, Sirpa Jalkanen, Markku Jalkanen, Jami Mandelin, Maija Hollmén, Mikael Maksimow, Jenna H Rannikko, Matti K Karvonen, Marita Vainio, Sirpa Jalkanen, Markku Jalkanen, Jami Mandelin

Abstract

Common lymphatic endothelial and vascular endothelial receptor-1 (Clever-1) is a multifunctional type-1 transmembrane protein that plays an important role in immunosuppression against tumors. Clever-1 is highly expressed in a subset of human tumor-associated macrophages and associated with poor survival. In mice, Clever-1 supports tumor growth and metastasis formation, and its deficiency or blockage induces T-cell-dependent killing of cancer cells. Therefore, targeting Clever-1 could lead to T-cell activation and restoration of immune response also in patients with cancer. This is studied in an on-going clinical trial [Macrophage Antibody To INhibit immune Suppression (MATINS); NCT03733990] in patients with advanced solid tumors where bexmarilimab, a humanized IgG4 antibody against human Clever-1, shows promising safety and efficacy. Here, we report the humanization and nonclinical characterization of physicochemical properties, biological potency, and safety profile of bexmarilimab. Bexmarilimab showed high affinity to Clever-1 on KG-1 cells and bound to Clever-1 on the surface of classical and intermediate monocytes derived from healthy human blood. Bexmarilimab inhibited the internalization of its natural ligand acetylated low-density lipoprotein into KG-1 cells and increased TNFα secretion from macrophages but did not impair phagocytic clearance. Bexmarilimab did not induce significant cytokine release in human whole-blood cultures, did not contain nonsafe immunogenic glycans, or show any significant binding to human Fcγ receptors or complement pathway component C1q. In vivo, bexmarilimab showed dose-dependent duration of monocyte Clever-1 receptor occupancy in cynomolgus monkeys but did not induce a cytokine storm up to a dose of 100 mg/kg. In conclusion, these data support the clinical development of bexmarilimab for the restoration of immune response in cancers.

©2022 The Authors; Published by the American Association for Cancer Research.

Figures

Figure 1.
Figure 1.
Characterization of bexmarilimab binding in vitro. A, Binding of biotinylated parental anti–Clever-1 antibody 3–372 to human Clever-1 in the presence of nonbiotinylated 3–372, chimeric anti–Clever-1 antibody and three composite antibodies (VH2/VK5, VH3/VK5, VH4/VK5) as determined by a competitive ELISA. IC50 values were calculated by three-parameter nonlinear curve fitting with 95% confidence intervals (CI). B, Bexmarilimab-recognized region of Clever-1. The orange boxes and blue closed circles indicate fasciclin domains and EGF-like domains, respectively. The arrowheads indicate relative positions of identified binding motifs, the core epitopes are shown in red. C, Bexmarilimab immunoreactivity (magenta) in human lymph node with CD31 (green) and Hoechst (blue) co-staining. Scale bar 40 μm. Arrowheads point to Clever-1 expression on lymphatic endothelial cells and the arrow points to a single CD31-negative cell. Binding of bexmarilimab to human CD14+ cells (D) and human KG-1 acute myelogenous leukemia cells (E) as determined by flow cytometry. F, Binding and quantitation of bexmarilimab and the isotype control to KG-1 cells transfected with a Clever-1–targeting shRNA as determined by flow cytometry. The wild-type histogram is shown in black, the Clever-1 knockdown histogram in red, and the isotype control binding in solid gray. APC, allophycocyanin. Receptor occupancy of bexmarilimab in classical (G) and intermediate monocytes (H), shown as change in fluorescence intensity of CD14+ cells that bind to labeled bexmarilimab or mAb 9–11. Representative data are shown for 1 of 3 donors. MFI, mean fluorescence intensity.
Figure 2.
Figure 2.
Bexmarilimab binds to Clever-1 on monocytes and inhibits internalization of acLDL while inducing TNFα secretion. A, Internalized and surface-bound S. aureus particles in monocytes from healthy human donors after treatment with bexmarilimab, mAb 9–11, or their respective isotype controls. B, Internalization of acLDL into KG-1 cells via Clever-1 receptor in the presence of two batches of bexmarilimab, mAb 9–11, parental mouse antibody 3–372, and an isotype control as determined by a competition assay and flow cytometry. The dotted line indicates the level of internalization without a competitor compound present. C, Mean fold changes in TNFα secretion by primary human macrophages upon treatment with 0.1–50 μg/mL bexmarilimab compared with an isotype control. The macrophages were induced in vitro from CD14+ monocytes obtained from seven healthy donors. **, P < 0.01; NS, not significant compared with isotype control. The statistical analysis was performed using one-way repeated measures ANOVA followed by Dunnett test. D, Absolute levels of TNFα secretion by primary human macrophages (C) upon treatment with 0.1 to 50 μg/mL bexmarilimab or an isotype control. Each donor is shown separately. E, Whole blood from 3 healthy donors was incubated with bexmarilimab or Ig-control for 24 hours at concentrations of 1.33, 13.3, or 133 μg/mL. These correspond to the initial estimated concentrations after injection of the antibodies into a blood volume of 4.5 L at doses of 0.1, 1, or 10 mg/kg, respectively, in a 60-kg person. Receptor occupancy of bexmarilimab on human CD14+ cells was measured by flow cytometry. The binding of fluorescently labeled bexmarilimab on CD14+ cells after administration of nonlabeled bexmarilimab is shown in blue and the binding of fluorescently labeled 9–11 antibody is shown in grey. Data normalized to Ig-treated cells. F, Flow cytometry analysis of acLDL internalization in CD14+ cells after bexmarilimab treatment (E). The blood was spiked with 1 μg/mL of AF488-acLDL during bexmarilimab incubation. Data normalized to Ig-treated cells.
Figure 3.
Figure 3.
Bexmarilimab does not contain immunogenic glycans or bind to human FcγRI or complement 1q. A, N-glycoform distribution on bexmarilimab as determined by quantitative N-glycan profiling. The analysis was performed in triplicate and glycans were assigned according to retention time. B, Binding of bexmarilimab and the positive control IgG rituximab to FcγRI as determined by SPR analysis. Colored lines correspond raw data, black lines correspond fitted data. C, Binding of bexmarilimab and rituximab to component C1q of the complement pathway as determined by ELISA; a representative graph of three experiments. s, seconds.
Figure 4.
Figure 4.
Bexmarilimab does not induce cytokine release in whole blood. Release of cytokines (A) IL6 (A), IL10 (B), IFNγ (C), TNFα (D), and IL8 (E) upon bexmarilimab treatment of human whole blood as determined by Cytokine ScreenTM assay. Alemtuzumab and cetuximab were used as reference compounds. Each data point represents one donor (n = 21) and the graphs show the median, minimum, and maximum values. F, Cytokines induced by positive control [pokeweed mitogen (PWM)] compared with PBS. Statistical analysis was performed using one-way ANOVA followed by paired Student t test. ***, P < 0.001; ****, P < 0.0001 as compared with cetuximab.
Figure 5.
Figure 5.
Bexmarilimab shows dose-dependent duration of receptor occupancy in vivo and only moderate increase in ALT levels in cynomolgus monkeys. Receptor occupancy of bexmarilimab in cynomolgus monkey monocytes after administration of a single i.v. dose of bexmarilimab at 3 mg/kg (A), 30 mg/kg (B), or 100 mg/kg (C; n  =  6 animals/dosing group). Change in MFI of CD14+ cells binding to fluorescently labeled bexmarilimab after administration of nonlabeled bexmarilimab is shown in blue and binding to fluorescently labeled 9–11 antibody is shown in orange. Concentration of the bexmarilimab in circulation (grey) was measured at the same time points as the receptor occupancy. D, Effect of bexmarilimab on ALT levels in cynomolgus monkeys on days 7 and 20 compared with predose levels. h, hours.

References

    1. Goerdt S, Walsh LJ, Murphy GF, Pober JS. Identification of a novel high molecular weight protein preferentially expressed by sinusoidal endothelial cells in normal human tissues. J Cell Biol 1991;113:1425–37.
    1. Irjala H, Elima K, Johansson EL, Merinen M, Kontula K, Alanen K, et al. . The same endothelial receptor controls lymphocyte traffic both in vascular and lymphatic vessels. Eur J Immunol 2003;33:815–24.
    1. Palani S, Elima K, Ekholm E, Jalkanen S, Salmi M. Monocyte stabilin-1 suppresses the activation of Th1 lymphocytes. J Immunol 2016;196:115–23.
    1. Palani S, Maksimow M, Miiluniemi M, Auvinen K, Jalkanen S, Salmi M. Stabilin-1/CLEVER-1, a type 2 macrophage marker, is an adhesion and scavenging molecule on human placental macrophages. Eur J Immunol 2011;41:2052–63.
    1. Kzhyshkowska J. Multifunctional receptor stabilin-1 in homeostasis and disease. ScientificWorldJournal 2010;10:2039–53.
    1. Shetty S, Weston CJ, Oo YH, Westerlund N, Stamataki Z, Youster J, et al. . Common lymphatic endothelial and vascular endothelial receptor-1 mediates the transmigration of regulatory T cells across human hepatic sinusoidal endothelium. J Immunol 2011;186:4147–55.
    1. Virtakoivu R, Rannikko JH, Viitala M, Vaura F, Takeda A, Lönnberg T, et al. . Systemic blockade of Clever-1 elicits lymphocyte activation alongside checkpoint molecule downregulation in patients with solid tumors: Results from a phase I/II clinical trial. Clin Cancer Res 2021;27:4205–20.
    1. Dunkel J, Viitala M, Karikoski M, Rantakari P, Virtakoivu R, Elima K, et al. . Enhanced antibody production in clever-1/stabilin-1-deficient mice. Front Immunol 2018;9:2257.
    1. Yin SP, Gao Y, Xie XS, Xu DD, Riabov V, Du WD. Accumulation of stabilin-1 positive macrophages in the early stage of gastric cancer is associated with short cumulative survival. Oncol Lett 2020;19:2404–12.
    1. Algars A, Irjala H, Vaittinen S, Huhtinen H, Sundström J, Salmi M, et al. . Type and location of tumor-infiltrating macrophages and lymphatic vessels predict survival of colorectal cancer patients. Int J Cancer 2012;131:864–73.
    1. Riabov V, Yin S, Song B, Avdic A, Schledzewski K, Ovsiy I, et al. . Stabilin-1 is expressed in human breast cancer and supports tumor growth in mammary adenocarcinoma mouse model. Oncotarget 2016;7:31097–110.
    1. Karikoski M, Marttila-Ichihara F, Elima K, Rantakari P, Hollmén M, Kelkka T, et al. . Clever-1/stabilin-1 controls cancer growth and metastasis. Clin Cancer Res 2014;20:6452–64.
    1. Viitala M, Virtakoivu R, Tadayon S, Rannikko J, Jalkanen S, Hollmén M. Immunotherapeutic blockade of macrophage clever-1 reactivates the CD8+ T-cell response against immunosuppressive tumors. Clin Cancer Res 2019;25:3289–303.
    1. Angal S, King DJ, Bodmer MW, Turner A, Lawson AD, Roberts G, et al. . A single amino acid substitution abolishes the heterogeneity of chimeric mouse/human (IgG4) antibody. Mol Immunol 1993;30:105–8.
    1. Reddy MP, Kinney CA, Chaikin MA, Payne A, Fishman-Lobell J, Tsui P, et al. . Elimination of Fc receptor-dependent effector functions of a modified IgG4 monoclonal antibody to human CD4. J Immunol 2000;164:1925–33.
    1. Rantakari P, Patten DA, Valtonen J, Karikoski M, Gerke H, Dawes H, et al. . Stabilin-1 expression defines a subset of macrophages that mediate tissue homeostasis and prevent fibrosis in chronic liver injury. Proc Natl Acad Sci U S A 2016;113:9298–303.
    1. Idusogie EE, Presta LG, Gazzano-Santoro H, Totpal K, Wong PY, Ultsch M, et al. . Mapping of the C1q binding site on rituxan, a chimeric antibody with a human IgG1 Fc. J Immunol 2000;164:4178–84.
    1. Bugelski PJ, Achuthanandam R, Capocasale RJ, Treacy G, Bouman-Thio E. Monoclonal antibody-induced cytokine-release syndrome. Expert Rev Clin Immunol 2009;5:499–521.
    1. Roopenian DC, Akilesh S. FcRn: the neonatal Fc receptor comes of age. Nat Rev Immunol 2007;7:715–25.
    1. George TJ, Laplant KD, Walden EO, Davis AB, Riggs CE, Close JL, et al. . Managing cetuximab hypersensitivity-infusion reactions: incidence, risk factors, prevention, and retreatment. J Support Oncol 2010;8:72–7.
    1. Patel DD, Goldberg RM. Cetuximab-associated infusion reactions: pathology and management. Oncology (Williston Park) 2006;20:1373–82.
    1. Stančič B, Qvarfordt B, Berglund MM, Brenden N, Sydow Bäckman M, Fransson M, et al. . The blood endothelial cell chamber - An innovative system to study immune responses in drug development. Int Immunopharmacol 2021;90:107237.
    1. Thomas K, Eisele J, Rodriguez-Leal FA, Hainke U, Ziemssen T. Acute effects of alemtuzumab infusion in patients with active relapsing-remitting MS. Neurol Neuroimmunol Neuroinflamm 2016;3:e228.
    1. Chanmee T, Ontong P, Konno K, Itano N. Tumor-associated macrophages as major players in the tumor microenvironment. Cancers (Basel) 2014;6:1670–90.
    1. Noy R, Pollard JW. Tumor-associated macrophages: from mechanisms to therapy. Immunity 2014;41:49–61.
    1. Fritz JM, Tennis MA, Orlicky DJ, Lin H, Ju C, Redente EF, et al. . Depletion of tumor-associated macrophages slows the growth of chemically induced mouse lung adenocarcinomas. Front Immunol 2014;5:587.
    1. Wu X, Schulte BC, Zhou Y, Haribhai D, Mackinnon AC, Plaza JA, et al. . Depletion of M2-like tumor-associated macrophages delays cutaneous T-cell lymphoma development in vivo. J Invest Dermatol 2014;134:2814–22.
    1. O'Brien SA, Orf J, Skrzypczynska KM, Tan H, Kim J, DeVoss J, et al. . Activity of tumor-associated macrophage depletion by CSF1R blockade is highly dependent on the tumor model and timing of treatment. Cancer Immunol Immunother 2021;70:2401–10.
    1. Karikoski M, Irjala H, Maksimow M, Miiluniemi M, Granfors K, Hernesniemi S, et al. . Clever-1/Stabilin-1 regulates lymphocyte migration within lymphatics and leukocyte entrance to sites of inflammation. Eur J Immunol 2009;39:3477–87.
    1. Bono P, Virtakoivu R, Vaura F, Jaakkola P, Shetty S, Thibault A, et al. . Immune activation in first-in-human anti-macrophage antibody (anti-Clever-1 mAb; FP-1305) phase I/II MATINS trial: Part 1 dose-escalation, safety and efficacy results. J Clin Oncol 38:15s, 2020. (suppl; abstr 3097).
    1. Kleinerman ES, Kurzrock R, Wyatt D, Quesada JR, Gutterman JU, Fidler IJ. Activation or suppression of the tumoricidal properties of monocytes from cancer patients following treatment with human recombinant gamma-interferon. Cancer Res 1986;46:5401–5.
    1. Seeley JJ, Ghosh S. Molecular mechanisms of innate memory and tolerance to LPS. J Leukoc Biol 2017;101:107–19.
    1. Stewart R, Hammond S, Oberst MW, Wilkinson R. The role of Fc gamma receptors in the activity of immunomodulatory antibodies for cancer. J Immunother Cancer 2014;2:29.
    1. Meyer S, Leusen JH, Boross P. Regulation of complement and modulation of its activity in monoclonal antibody therapy of cancer. MAbs 2014;6:1133–44.
    1. Fajgenbaum DC, June CH. Cytokine storm. N Engl J Med 2020;383:2255–73.
    1. Schledzewski K, Géraud C, Arnold B, Wang S, Gröne HJ, Kempf T, et al. . Deficiency of liver sinusoidal scavenger receptors stabilin-1 and -2 in mice causes glomerulofibrotic nephropathy via impaired hepatic clearance of noxious blood factors. J Clin Invest 2011;121:703–14.
    1. Politz O, Gratchev A, McCourt PA, Schledzewski K, Guillot P, Johansson S, et al. . Stabilin-1 and -2 constitute a novel family of fasciclin-like hyaluronan receptor homologues. Biochem J 2002;362:155–64.
    1. Miller CM, Donner AJ, Blank EE, Egger AW, Kellar BM, Østergaard ME, et al. . Stabilin-1 and Stabilin-2 are specific receptors for the cellular internalization of phosphorothioate-modified antisense oligonucleotides (ASOs) in the liver. Nucleic Acids Res 2016;44:2782–94.
    1. Zhou B, Weigel JA, Fauss L, Weigel PH. Identification of the hyaluronan receptor for endocytosis (HARE). J Biol Chem 2000;275:37733–41.
    1. Tamura Y, Adachi H, Osuga J, Ohashi K, Yahagi N, Sekiya M, et al. . FEEL-1 and FEEL-2 are endocytic receptors for advanced glycation end products. J Biol Chem 2003;278:12613–7.
    1. Jung MY, Park SY, Kim IS. Stabilin-2 is involved in lymphocyte adhesion to the hepatic sinusoidal endothelium via the interaction with alphaMbeta2 integrin. J Leukoc Biol 2007;82:1156–65.
    1. Bono P, Minchom A, Shetty S, Ma Y, Cruz R, de Jonge M, et al. . LBA38 - Bexmarilimab, a novel macrophage re-programmer shows promising anti-tumour activity in phase I/II trial in several last line solid tumour types. Ann Oncol 2021:32:S1283–S346.

Source: PubMed

3
Tilaa