In order to eliminate the immunosuppressive tumor stroma, which also prevents oncolytic adenoviruses (OAds) from entering the tumor, de Sostoa et al. designed a bispecific T-cell engager (BiTE) targeting the cancer-associated fibroblast (CAF) activation protein-α (FAP) and transduced it into an OAd. Cells infected with FBiTE-OAd secreted BiTE, which induced activation and proliferation of CD4+ and CD8+ T cells in the presence of FAP-expressing cells in vitro. When injected intratumorally, FBiTE-OAd increased accumulation and retention of T cells, killed FAP+ CAFs, reduced tumor growth, and increased survival in two mouse tumor models.

BACKGROUND: Oncolytic virus (OV)-based therapies have an emerging role in the treatment of solid tumors, involving both direct cell lysis and immunogenic cell death. Nonetheless, tumor-associated stroma limits the efficacy of oncolytic viruses by forming a barrier that blocks efficient viral penetration and spread. The stroma also plays a critical role in progression, immunosuppression and invasiveness of cancer. Fibroblast activation protein-alpha (FAP) is highly overexpressed in cancer-associated fibroblasts (CAFs), the main cellular component of tumor stroma, and in this study we assessed whether arming oncolytic adenovirus (OAd) with a FAP-targeting Bispecific T-cell Engager (FBiTE) could retarget infiltrated lymphocytes towards CAFs, enhancing viral spread and T cell-mediated cytotoxicity against the tumor stroma to improve therapeutic activity. METHODS: The bispecific T-cell Engager against FAP was constructed using an anti-human CD3 single-chain variable fragment (scFv) linked to an anti-murine and human FAP scFv. This FBiTE was inserted in the oncolytic adenovirus ICOVIR15K under the control of the major late promoter, generating the ICO15K-FBiTE. ICO15K-FBiTE replication and potency were assessed in HT1080 and A549 tumor cell lines. The expression of the FBiTE and the activation and proliferation of T cells that induced along with the T cell-mediated cytotoxicity of CAFs were evaluated by flow cytometry in vitro. In vivo, T-cell biodistribution and antitumor efficacy studies were conducted in NOD/scid/IL2rg(-)/(-) (NSG) mice. RESULTS: FBiTE expression did not decrease the infectivity and replication potency of the armed virus. FBiTE-mediated binding of CD3(+) effector T cells and FAP(+) target cells led to T-cell activation, proliferation, and cytotoxicity of FAP-positive cells in vitro. In vivo, FBiTE expression increased intratumoral accumulation of T cells and decreased the level of FAP, a marker of CAFs, in tumors. The antitumor activity of the FBiTE-armed adenovirus was superior to the parental virus. CONCLUSIONS: Combination of viral oncolysis of cancer cells and FBiTE-mediated cytotoxicity of FAP-expressing CAFs might be an effective strategy to overcome a key limitation of oncolytic virotherapy, encouraging its further clinical development.

Author Info: (1) ProCure Program, IDIBELL-Institut Catala d'Oncologia, l'Hospitalet de Llobregat, El Prat de Llobregat, Spain. (2) ProCure Program, IDIBELL-Institut Catala d'Oncologia, l'Hospit

Author Info: (1) ProCure Program, IDIBELL-Institut Catala d'Oncologia, l'Hospitalet de Llobregat, El Prat de Llobregat, Spain. (2) ProCure Program, IDIBELL-Institut Catala d'Oncologia, l'Hospitalet de Llobregat, El Prat de Llobregat, Spain. (3) ProCure Program, IDIBELL-Institut Catala d'Oncologia, l'Hospitalet de Llobregat, El Prat de Llobregat, Spain. (4) ProCure Program, IDIBELL-Institut Catala d'Oncologia, l'Hospitalet de Llobregat, El Prat de Llobregat, Spain. (5) ProCure Program, IDIBELL-Institut Catala d'Oncologia, l'Hospitalet de Llobregat, El Prat de Llobregat, Spain. VCN Biosciences S.L., Grifols Corporate Offices, Sant Cugat del Valles, Spain. (6) ProCure Program, IDIBELL-Institut Catala d'Oncologia, l'Hospitalet de Llobregat, El Prat de Llobregat, Spain. ralemany@iconcologia.net.