Abstract

The response to cancer immune therapy is dependent on endogenous tumor-reactive T cells. To bypass this requirement, CD3-bispecific antibodies have been developed to induce a polyclonal T-cell response against the tumor. Anti-HER2/CD3 T-cell-dependent bispecific (TDB) antibody is highly efficacious in the treatment of HER2-overexpressing tumors in mice. Efficacy and immunologic effects of anti-HER2/CD3 TDB were investigated in mammary tumor model with very few T cells prior treatment. We further describe the mechanism for TDB-induced T-cell recruitment to tumors. The immunologic effects and the mechanism of CD3-bispecific antibody-induced T-cell recruitment into spontaneous HER2-overexpressing mammary tumors was studied using human HER2 transgenic, immunocompetent mouse models. Anti-HER2/CD3 TDB treatment induced an inflammatory response in tumors converting them from poorly infiltrated to an inflamed, T-cell abundant, phenotype. Multiple mechanisms accounted for the TDB-induced increase in T cells within tumors. TDB treatment induced CD8+ T-cell proliferation. T cells were also actively recruited post-TDB treatment by IFNγ-dependent T-cell chemokines mediated via CXCR3. This active T-cell recruitment by TDB-induced chemokine signaling was the dominant mechanism and necessary for the therapeutic activity of anti-HER2/CD3 TDB. In summary, we demonstrate that the activity of anti-HER2/CD3 TDB was not dependent on high-level baseline T-cell infiltration. Our results suggest that anti-HER2/CD3 TDB may be efficacious in patients and indications that respond poorly to checkpoint inhibitors. An active T-cell recruitment mediated by TDB-induced chemokine signaling was the major mechanism for T-cell recruitment.

Highlights

  • Therapies that direct T cells to tumors, including adoptive transfer of genetically engineered T cells and T-cell–dependent bispecific (TDB) antibodies, have been clinically validated for the treatment of B-cell leukemias as well as lymphomas, [1, 2] and have demonstrated promising activity for the treatment of multiple myeloma [3, 4]

  • T cells were actively recruited post-TDB treatment by IFNg-dependent T-cell chemokines mediated via CXCR3

  • In summary, we demonstrate that the activity of anti-HER2/CD3 TDB was not dependent on high-level baseline T-cell infiltration

Read more

Summary

Introduction

Therapies that direct T cells to tumors, including adoptive transfer of genetically engineered T cells and T-cell–dependent bispecific (TDB) antibodies, have been clinically validated for the treatment of B-cell leukemias as well as lymphomas, [1, 2] and have demonstrated promising activity for the treatment of multiple myeloma [3, 4]. Simultaneous binding of the bispecific antibody to T cells and tumor targets leads to rapid formation of an immunologic synapse by induction of TCR clustering and exclusion of a key negative regulator of TCR activity (CD45) from the synapse [6]. Because these molecules mediate T-cell activation via binding to CD3 rather than TCR and peptide/MHC interactions, CD3-bispecific antibodies are able to redirect a polyclonal T-cell response to tumor cells independent from tumor-antigen specificity of the T cell. The mechanism(s) underlying increased T-cell accumulation has yet to be described

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call