Repertoire enhancement with adoptively transferred female lymphocytes controls the growth of pre-implanted murine prostate cancer

PLoS One. 2012;7(4):e35222. doi: 10.1371/journal.pone.0035222. Epub 2012 Apr 6.

Abstract

Background: In prostate cancer, genes encoding androgen-regulated, Y-chromosome-encoded, and tissue-specific antigens may all be overexpressed. In the adult male host, however, most high affinity T cells targeting these potential tumor rejection antigens will be removed during negative selection. In contrast, the female mature T-cell repertoire should contain abundant precursors capable of recognizing these classes of prostate cancer antigens and mediating effective anti-tumor immune responses.

Methodology/principal findings: We find that syngeneic TRAMP-C2 prostatic adenocarcinoma cells are spontaneously rejected in female hosts. Adoptive transfer of naïve female lymphocytes to irradiated male hosts bearing pre-implanted TRAMP-C2 tumor cells slows tumor growth and mediates tumor rejection in some animals. The success of this adoptive transfer was dependent on the transfer of female CD4 T cells and independent of the presence of CD25-expressing regulatory T cells in the transferred lymphocytes. We identify in female CD4 T cells stimulated with TRAMP-C2 a dominant MHC II-restricted response to the Y-chromosome antigen DBY. Furthermore, CD8 T cell responses in female lymphocytes to the immunodominant MHC I-restricted antigen SPAS-1 are markedly increased compared to male mice. Finally, we find no exacerbation of graft-versus-host disease in either syngeneic or minor-antigen mismatched allogeneic lymphocyte adoptive transfer models by using female into male versus male into male cells.

Conclusions/significance: This study shows that adoptively transferred female lymphocytes, particularly CD4 T cells, can control the outgrowth of pre-implanted prostatic adenocarcinoma cells. This approach does not significantly worsen graft-versus-host responses suggesting it may be viable in the clinic. Further, enhancing the available immune repertoire with female-derived T cells may provide an excellent pool of prostate cancer reactive T cells for further augmentation by combination with either vaccination or immune regulatory blockade strategies.

Publication types

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

MeSH terms

  • Adenocarcinoma / immunology
  • Adenocarcinoma / therapy*
  • Adult
  • Animals
  • Antigens, Neoplasm / immunology
  • CD4-Positive T-Lymphocytes / immunology
  • CD4-Positive T-Lymphocytes / transplantation*
  • CD8-Positive T-Lymphocytes / immunology
  • CD8-Positive T-Lymphocytes / transplantation*
  • DEAD-box RNA Helicases / immunology
  • Female
  • Graft vs Host Disease / prevention & control
  • Humans
  • Immunotherapy, Adoptive / methods*
  • Lymphocyte Count
  • Male
  • Mice
  • Minor Histocompatibility Antigens
  • Neoplasm Transplantation
  • Prostatic Neoplasms / immunology
  • Prostatic Neoplasms / therapy*
  • Sex Factors
  • Whole-Body Irradiation

Substances

  • Antigens, Neoplasm
  • Minor Histocompatibility Antigens
  • Ddx3y protein, mouse
  • DEAD-box RNA Helicases