Abstract

<div>Abstract<p>Cytokines often display substantial toxicities at low concentrations, preventing their escalation for therapeutic treatment of cancer. Fusion proteins comprising cytokines and recombinant antibodies may improve the anticancer activity of proinflammatory cytokines. Murine IFNγ was appended in the diabody format at the C-terminus of the F8 antibody, generating the F8–IFNγ fusion protein. The F8 antibody is specific for the extra-domain A (EDA) of fibronectin, a tumor-associated antigen that is expressed in the vasculature and stroma of almost all tumor types. Tumor-targeting properties were measured <i>in vivo</i> using a radioiodinated preparation of the fusion protein. Therapy experiments were performed in three syngeneic murine models of cancer [F9 teratocarcinoma, WEHI-164 fibrosarcoma, and Lewis lung carcinoma (LLC)]. F8–IFNγ retained the biologic activity of both the antibody and the cytokine moiety <i>in vitro</i>, but, unlike the parental F8 antibody, it did not preferentially localize to the tumors <i>in vivo</i>. However, when unlabeled F8–IFNγ was administered before radioiodinated F8–IFNγ, a selective accumulation at the tumor site was observed. F8–IFNγ showed dose-dependent anticancer activity with a clear superiority over untargeted recombinant IFNγ. The anticancer activity was potentiated by combining with F8–IL4 without additional toxicities, whereas combination of F8–IFNγ with F8–TNF was lethal in all mice. Unlike other antibody–cytokine fusions, the use of IFNγ as payload for anticancer therapy is associated with a receptor-trapping mechanism, which can be overcome by the administration of a sufficiently large amount of the fusion protein without any detectable toxicity at the doses used. <i>Cancer Immunol Res; 2(6); 559–67. ©2014 AACR</i>.</p></div>

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