Immunization with aspartate-β-hydroxylase-loaded dendritic cells produces antitumor effects in a rat model of intrahepatic cholangiocarcinoma

Hepatology. 2012 Jan;55(1):86-97. doi: 10.1002/hep.24629.

Abstract

Dendritic cells (DCs) capture and process proteins and present peptides on the cell surface in the context of major histocompatibility complex I and II molecules to induce antigen-specific T cell immune responses. The aims of this study were to (1) employ an expanded and purified DC population and load them with aspartate-β-hydroxylase (ASPH), a highly expressed tumor-associated cell surface protein, and (2) to determine if immunization induced antitumor effects in an orthotopic rat model of intrahepatic cholangiocarcinoma. Splenocytes were incubated with ASPH-coated beads and passed through a magnetic field to yield an 80% pure DC OX62+ population. This DC subset was stimulated with granulocyte-macrophage colony-stimulating factor, interleukin-4, CD40L, and interferon-γ, resulting in a 40-fold increase in interleukin-12A messenger RNA expression to subsequently generate a T helper 1-type immune response. After incubation with the cytokine cocktail, DCs were found to have matured, as demonstrated by increased expression of CD40, CD80, and CD86 costimulatory molecules. Immunization with ASPH-loaded DCs induced antigen-specific immunity. A clone of the parental tumorigenic rat BDEneu cholangiocyte cell line, designated BDEneu-CL24, was found to have the highest number of cells expressing this surface protein (97%); it maintained the same phenotypic characteristics of the parental cell line and was used to produce intrahepatic tumors in immunocompetent syngeneic Fisher-344 rats. Immunization with ASPH-loaded DCs generated cytotoxicity against cholangiocarcinoma cells in vitro and significantly suppressed intrahepatic tumor growth and metastasis, and was associated with increased CD3+ lymphocyte infiltration into the tumors.

Conclusion: These findings suggest that immunization with ASPH-loaded DCs may constitute a novel therapeutic approach for intrahepatic cholangiocarcinoma, because this protein also appears to be highly conserved and expressed on human hepatobiliary tumors.

Publication types

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

MeSH terms

  • Adoptive Transfer / methods*
  • Animals
  • Asparagine / analogs & derivatives*
  • Asparagine / genetics
  • Asparagine / immunology
  • Bile Duct Neoplasms / immunology
  • Bile Duct Neoplasms / therapy*
  • Bile Ducts, Intrahepatic*
  • Cell Division / immunology
  • Cells, Cultured
  • Cholangiocarcinoma / immunology
  • Cholangiocarcinoma / therapy*
  • Dendritic Cells / cytology
  • Dendritic Cells / enzymology
  • Dendritic Cells / immunology*
  • Disease Models, Animal
  • Humans
  • Immunization / methods
  • Interleukin-12 / immunology
  • Interleukin-12 / metabolism
  • Male
  • Rats
  • Rats, Inbred F344
  • Recombinant Proteins / genetics
  • Recombinant Proteins / immunology
  • Spleen / cytology
  • Spleen / immunology

Substances

  • Recombinant Proteins
  • Interleukin-12
  • beta-aspartylhydroxamic acid
  • Asparagine