Skip to main content
Journal cover image

CEA/CD3-bispecific T cell-engaging (BiTE) antibody-mediated T lymphocyte cytotoxicity maximized by inhibition of both PD1 and PD-L1.

Publication ,  Journal Article
Osada, T; Patel, SP; Hammond, SA; Osada, K; Morse, MA; Lyerly, HK
Published in: Cancer Immunol Immunother
June 2015

Bispecific T cell-engaging (BiTE) antibodies recruit polyclonal cytotoxic T cells (CTL) to tumors. One such antibody is carcinoembryonic antigen (CEA) BiTE that mediates T cell/tumor interaction by simultaneously binding CD3 expressed by T cells and CEA expressed by tumor cells. A widely operative mechanism for mitigating cytotoxic T cell-mediated killing is the interaction of tumor-expressed PD-L1 with T cell-expressed PD-1, which may be partly reversed by PD-1/PD-L1 blockade. We hypothesized that PD-1/PD-L1 blockade during BiTE-mediated T cell killing would enhance CTL function. Here, we determined the effects of PD-1 and PD-L1 blockade during initial T cell-mediated killing of CEA-expressing human tumor cell lines in vitro, as well as subsequent T cell-mediated killing by T lymphocytes that had participated in tumor cell killing. We observed a rapid upregulation of PD-1 expression and diminished cytolytic function of T cells after they had engaged in CEA BiTE-mediated killing of tumors. T cell cytolytic activity in vitro could be maximized by administration of anti-PD-1 or anti-PD-L1 antibodies alone or in combination if applied prior to a round of T cell killing, but T cell inhibition could not be fully reversed by this blockade once the T cells had killed tumor. In conclusion, our findings demonstrate that dual blockade of PD-1 and PD-L1 maximizes T cell killing of tumor directed by CEA BiTE in vitro, is more effective if applied early, and provides a rationale for clinical use.

Duke Scholars

Altmetric Attention Stats
Dimensions Citation Stats

Published In

Cancer Immunol Immunother

DOI

EISSN

1432-0851

Publication Date

June 2015

Volume

64

Issue

6

Start / End Page

677 / 688

Location

Germany

Related Subject Headings

  • T-Lymphocytes, Cytotoxic
  • Programmed Cell Death 1 Receptor
  • Pancreatic Neoplasms
  • Mice, SCID
  • Mice, Inbred NOD
  • Mice
  • Immunotherapy
  • Immunology
  • Humans
  • HT29 Cells
 

Citation

APA
Chicago
ICMJE
MLA
NLM
Osada, T., Patel, S. P., Hammond, S. A., Osada, K., Morse, M. A., & Lyerly, H. K. (2015). CEA/CD3-bispecific T cell-engaging (BiTE) antibody-mediated T lymphocyte cytotoxicity maximized by inhibition of both PD1 and PD-L1. Cancer Immunol Immunother, 64(6), 677–688. https://doi.org/10.1007/s00262-015-1671-y
Osada, Takuya, Sandip P. Patel, Scott A. Hammond, Koya Osada, Michael A. Morse, and H Kim Lyerly. “CEA/CD3-bispecific T cell-engaging (BiTE) antibody-mediated T lymphocyte cytotoxicity maximized by inhibition of both PD1 and PD-L1.Cancer Immunol Immunother 64, no. 6 (June 2015): 677–88. https://doi.org/10.1007/s00262-015-1671-y.
Osada T, Patel SP, Hammond SA, Osada K, Morse MA, Lyerly HK. CEA/CD3-bispecific T cell-engaging (BiTE) antibody-mediated T lymphocyte cytotoxicity maximized by inhibition of both PD1 and PD-L1. Cancer Immunol Immunother. 2015 Jun;64(6):677–88.
Osada, Takuya, et al. “CEA/CD3-bispecific T cell-engaging (BiTE) antibody-mediated T lymphocyte cytotoxicity maximized by inhibition of both PD1 and PD-L1.Cancer Immunol Immunother, vol. 64, no. 6, June 2015, pp. 677–88. Pubmed, doi:10.1007/s00262-015-1671-y.
Osada T, Patel SP, Hammond SA, Osada K, Morse MA, Lyerly HK. CEA/CD3-bispecific T cell-engaging (BiTE) antibody-mediated T lymphocyte cytotoxicity maximized by inhibition of both PD1 and PD-L1. Cancer Immunol Immunother. 2015 Jun;64(6):677–688.
Journal cover image

Published In

Cancer Immunol Immunother

DOI

EISSN

1432-0851

Publication Date

June 2015

Volume

64

Issue

6

Start / End Page

677 / 688

Location

Germany

Related Subject Headings

  • T-Lymphocytes, Cytotoxic
  • Programmed Cell Death 1 Receptor
  • Pancreatic Neoplasms
  • Mice, SCID
  • Mice, Inbred NOD
  • Mice
  • Immunotherapy
  • Immunology
  • Humans
  • HT29 Cells