Autologous tumor vaccines processed to express alpha-gal epitopes: a practical approach to immunotherapy in cancer

Cancer Immunol Immunother. 2004 Nov;53(11):935-45. doi: 10.1007/s00262-004-0524-x. Epub 2004 Jun 16.

Abstract

This review describes a method by which the human natural anti-Gal antibody can be exploited as an endogenous adjuvant for targeting autologous tumor vaccines to antigen-presenting cells (APCs). Tumor cells remaining in the patient after completion of surgery, radiation, and chemotherapy are the cause of tumor relapse. These residual tumor cells can not be detected by imaging, but their destruction may be feasible by active immunotherapy. Since specific tumor-associated antigens (TAAs) have not been identified for the majority of cancers, irradiated autologous tumor vaccines have been considered as an immunotherapy treatment that may elicit an immune response against the residual tumor cells expressing TAAs. However, tumor cells evolve in cancer patients in a stealthy way, i.e., they are not detected by APCs, even in the form of vaccine. Effective targeting of tumor vaccines for uptake by APCs is a prerequisite for eliciting an effective immune response which requires transport of the vaccine by APCs from the vaccination site to the draining lymph nodes. In the lymph nodes, the APCs transporting the vaccine process and present peptides, including the autologous TAA peptides for activation of the tumor-specific T cells. The required targeting of vaccines to APCs is feasible in humans by the use of anti-Gal. This antibody interacts specifically with the alpha-gal epitope (Galalpha1-3Galbeta1-4GlcNAc-R) and is the only known natural IgG antibody to be present in large amounts in all humans who are not severely immunocompromised. The alpha-gal epitope can be synthesized on any type of human tumor cell by the use of recombinant alpha1,3galactosyltransferase (alpha1,3GT). Solid tumors obtained from surgery are homogenized and their membranes subjected to alpha-gal epitope synthesis. Similarly, alpha-gal epitopes can be synthesized on intact tumor cells from hematological malignancies. Administration of irradiated autologous tumor vaccines processed to express alpha-gal epitopes results in in situ opsonization of the vaccinating cells or cell membranes due to anti-Gal binding to these epitopes. The bound antibody serves to target the autologous tumor vaccine to APCs because the Fc portion of the antibody interacts with Fcgamma receptors on APCs. Since patients receive their own TAAs, the vaccine is customized for autologous TAAs in the individual patient. The repeated vaccination with such autologous tumor vaccines provides the immune system of each patient with an additional opportunity to be effectively activated by the autologous TAAs. In some of the immunized patients this activation may be potent enough to induce an immune-mediated eradication of the residual tumor cells expressing these TAAs.

Publication types

  • Review

MeSH terms

  • Animals
  • Antigen-Presenting Cells / chemistry
  • Cancer Vaccines*
  • Carbohydrate Sequence
  • Cell Line, Tumor
  • Disease Models, Animal
  • Epitopes
  • Galactosyltransferases / therapeutic use*
  • Humans
  • Immunoglobulin Fragments
  • Immunotherapy / methods*
  • Lymphatic Metastasis
  • Models, Biological
  • Molecular Sequence Data
  • Peptides / chemistry
  • Phagocytosis
  • Recurrence
  • T-Lymphocytes / immunology
  • alpha-Galactosidase / chemistry*

Substances

  • Cancer Vaccines
  • Epitopes
  • Immunoglobulin Fragments
  • Peptides
  • Galactosyltransferases
  • N-acetyllactosaminide alpha-1,3-galactosyltransferase
  • alpha-Galactosidase