Anti-Gal-mediated targeting of human B lymphoma cells to antigen-presenting cells: a potential method for immunotherapy using autologous tumor cells

Haematologica. 2005 May;90(5):625-34.

Abstract

Background and objectives: The residual tumor cells remaining after completion of standard chemotherapy and radiation treatment in B lymphoma patients, may be eradicated by active immunotherapy that stimulates tumor-specific T lymphocytes. Irradiated autologous lymphoma cells expressing tumor-associated antigens (TAA) may serve as a potential tumor vaccine, provided that they are effectively targeted to the antigen-presenting cells (APC). We propose exploiting the natural anti-Gal antibody in order to target vaccinating tumor cells to APC. Anti-Gal constitutes 1% of IgG in human serum and interacts specifically with the alpha-gal epitope (Galalpha1-3Galphalbeta1-4GlcNAc-R).

Design and methods: Alpha-gal epitopes were synthesized in vitro on the membrane of primary lymphoma cells by using the recombinant glycosylation enzyme alpha1,3galactosyltransferase (alpha1,3GT). Processed tumor cells were opsonized by purified anti-Gal antibodies and studied for uptake (phagocytosis) by APC including monocyte-derived macrophages and dendritic cells. Cross-presentation of tumor antigens after phagocytosis of processed MHC-I negative lymphoma cells was measured by activation of a tumor-specific CD8+ T-cell line.

Results: We demonstrate synthesis of alpha-gal epitopes on freshly isolated B lymphoma cells of various types following the use of the recombinant enzyme alpha1,3GT. The subsequent binding of anti-Gal to the de novo synthesized alphagal epitopes opsonizes these tumor cells for effective uptake by macrophages and dendritic cells, through phagocytosis mediated by FcgammaR1 (CD64). Moreover, anti-Gal-mediated phagocytosis resulted in cross-presentation of TAA by dendritic cells.

Interpretation and conclusions: This study suggests that immunization with irradiated autologous lymphoma cells processed to express alpha-gal epitopes will result in anti-Gal-mediated, in vivo targeting of the autologous tumor vaccine to APC.

Publication types

  • Research Support, N.I.H., Extramural
  • Research Support, Non-U.S. Gov't
  • Research Support, U.S. Gov't, P.H.S.

MeSH terms

  • Amino Sugars / immunology
  • Animals
  • Antigen Presentation
  • Antigen-Presenting Cells / immunology*
  • Antigens, Neoplasm / immunology
  • Callithrix
  • Cancer Vaccines / therapeutic use*
  • Cells, Cultured / immunology
  • Galactosyltransferases / genetics
  • Galactosyltransferases / pharmacology
  • Glycosylation
  • Humans
  • Immunity, Innate
  • Immunization
  • Immunoglobulin G / immunology*
  • Immunotherapy / methods*
  • Lymphocyte Activation
  • Lymphoma, B-Cell / immunology
  • Lymphoma, B-Cell / pathology*
  • Lymphoma, B-Cell / therapy
  • Neoplastic Stem Cells / drug effects
  • Neoplastic Stem Cells / immunology*
  • Neoplastic Stem Cells / radiation effects
  • Neuraminidase / pharmacology
  • Phagocytosis
  • Receptors, IgG / immunology
  • Recombinant Fusion Proteins / pharmacology
  • T-Lymphocyte Subsets / immunology
  • Trisaccharides / immunology*

Substances

  • Amino Sugars
  • Antigens, Neoplasm
  • Cancer Vaccines
  • Immunoglobulin G
  • Receptors, IgG
  • Recombinant Fusion Proteins
  • Trisaccharides
  • alpha-galactosyl epitope
  • N-acetyllactosamine
  • Galactosyltransferases
  • N-acetyllactosaminide alpha-1,3-galactosyltransferase
  • Neuraminidase