A unique anti-CD115 monoclonal antibody which inhibits osteolysis and skews human monocyte differentiation from M2-polarized macrophages toward dendritic cells

MAbs. 2013 Sep-Oct;5(5):736-47. doi: 10.4161/mabs.25743. Epub 2013 Jul 15.

Abstract

Cancer progression has been associated with the presence of tumor-associated M2-macrophages (M2-TAMs) able to inhibit anti-tumor immune responses. It is also often associated with metastasis-induced bone destruction mediated by osteoclasts. Both cell types are controlled by the CD115 (CSF-1R)/colony-stimulating factor-1 (CSF-1, M-CSF) pathway, making CD115 a promising target for cancer therapy. Anti-human CD115 monoclonal antibodies (mAbs) that inhibit the receptor function have been generated in a number of laboratories. These mAbs compete with CSF-1 binding to CD115, dramatically affecting monocyte survival and preventing osteoclast and macrophage differentiation, but they also block CD115/CSF-1 internalization and degradation, which could lead to potent rebound CSF-1 effects in patients after mAb treatment has ended. We thus generated and selected a non-ligand competitive anti-CD115 mAb that exerts only partial inhibitory effects on CD115 signaling without blocking the internalization or the degradation of the CD115/CSF-1 complex. This mAb, H27K15, affects monocyte survival only minimally, but downregulates osteoclast differentiation and activity. Importantly, it inhibits monocyte differentiation to CD163(+)CD64(+) M2-polarized suppressor macrophages, skewing their differentiation toward CD14(-)CD1a(+) dendritic cells (DCs). In line with this observation, H27K15 also drastically inhibits monocyte chemotactic protein-1 secretion and reduces interleukin-6 production; these two molecules are known to be involved in M2-macrophage recruitment. Thus, the non-depleting mAb H27K15 is a promising anti-tumor candidate, able to inhibit osteoclast differentiation, likely decreasing metastasis-induced osteolysis, and able to prevent M2 polarization of TAMs while inducing DCs, hence contributing to the creation of more efficient anti-tumor immune responses.

Keywords: CD115; CSF-1R; FcγR; M-CSFR; M2-macrophages; cancer immunotherapy; dendritic cells; immunomodulation; osteolysis; tumor microenvironment.

MeSH terms

  • Animals
  • Antibodies, Monoclonal / immunology
  • Antibodies, Monoclonal / pharmacology*
  • Cell Differentiation / drug effects*
  • Cell Differentiation / immunology
  • Cell Line, Tumor
  • Cell Survival / drug effects
  • Cell Survival / immunology
  • Cells, Cultured
  • Chemokine CCL2 / immunology
  • Chemokine CCL2 / metabolism
  • Dendritic Cells / drug effects*
  • Dendritic Cells / immunology
  • Dendritic Cells / metabolism
  • Flow Cytometry
  • HL-60 Cells
  • Humans
  • Interleukin-6 / immunology
  • Interleukin-6 / metabolism
  • Macrophage Colony-Stimulating Factor / immunology
  • Macrophage Colony-Stimulating Factor / metabolism
  • Macrophages / drug effects*
  • Macrophages / immunology
  • Macrophages / metabolism
  • Mice
  • Mice, Inbred BALB C
  • Monocytes / drug effects*
  • Monocytes / immunology
  • Monocytes / metabolism
  • NIH 3T3 Cells
  • Osteoclasts / drug effects
  • Osteoclasts / immunology
  • Osteoclasts / metabolism
  • Osteolysis / immunology
  • Osteolysis / prevention & control*
  • Phosphorylation / drug effects
  • Protein Binding / drug effects
  • Protein Binding / immunology
  • Receptor, Macrophage Colony-Stimulating Factor / immunology
  • Receptor, Macrophage Colony-Stimulating Factor / metabolism
  • Signal Transduction / drug effects
  • Signal Transduction / immunology

Substances

  • Antibodies, Monoclonal
  • Chemokine CCL2
  • Interleukin-6
  • Macrophage Colony-Stimulating Factor
  • Receptor, Macrophage Colony-Stimulating Factor