Abstract

Antiviral immune responses present a major hurdle to the efficacious use of oncolytic adenoviruses as cancer treatments. Despite the existence of a highly immunosuppressive tumor environment, adenovirus-infected cells can nonetheless be efficiently cleared by infiltrating cytotoxic T lymphocytes (CTL) without compromising tumor burden. In this study, we tested the hypothesis that tumor-infiltrating T cells could be more effectively activated and redirected by oncolytic adenoviruses that were armed with bispecific T-cell-engager (BiTE) antibodies. The oncolytic adenovirus ICOVIR-15K was engineered to express an EGFR-targeting BiTE (cBiTE) antibody under the control of the major late promoter, leading to generation of ICOVIR-15K-cBiTE, which retained its oncolytic properties in vitro cBiTE expression and secretion was detected in supernatants from ICOVIR-15K-cBiTE-infected cells, and the secreted BiTEs bound specifically to both CD3+ and EGFR+ cells. In cell coculture assays, ICOVIR-15K-cBiTE-mediated oncolysis resulted in robust T-cell activation, proliferation, and bystander cell-mediated cytotoxicity. Notably, intratumoral injection of this cBiTE-expressing adenovirus increased the persistence and accumulation of tumor-infiltrating T cells in vivo, compared with the parental virus lacking such effects. Moreover, in two distinct tumor xenograft models, combined delivery of ICOVIR-15K-cBiTE with peripheral blood mononuclear cells or T cells enhanced the antitumor efficacy achieved by the parental counterpart. Overall, our results show how arming oncolytic adenoviruses with BiTE can overcome key limitations in oncolytic virotherapy. Cancer Res; 77(8); 2052-63. ©2017 AACR.

Highlights

  • Oncolytic adenoviruses have gained considerable attention as anticancer agents

  • A panel of cancer cell lines with varying EGFR expression levels, and CD3þ Jurkat and human Peripheral blood mononuclear cells (PBMC) were used (Supplementary Fig. S1). cBiTE antibodies were detected only in the supernatants of ICO15K-cBiTE–infected cells and they bound to EGFRþ (A431, A549, HCT116, and FaDu) but not EGFRÀ (MDA-MB-453) cancer cells

  • We armed the oncolytic adenovirus ICO15K with an EGFR-targeting bispecific T-cell–engager (BiTE). cBiTEs secreted from infected cells retained key features of BiTEs including target cell–dependent T-cell activation and proliferation and redirected lysis of cancer cells [27, 28]

Read more

Summary

Introduction

Oncolytic adenoviruses have gained considerable attention as anticancer agents. Their attractiveness relies on a multimodal mechanism to kill cancer cells that include direct oncolysis, bystander effect when armed with therapeutic transgenes, and immunogenic cell death–mediated antitumor immune responses [1]. Clinical trials have demonstrated the safety and potential of oncolytic viruses [2, 3] and highlighted the importance of the immune system for their success in cancer patients. Tumors develop numerous mechanisms to evade antitumor immune responses [4], but oncolytic viruses can revert such immune suppression [5]. Oncolytic virus-infected cells are often cleared from the tumor microenvironment by infiltrating virus-specific cyto-

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