Park et al. generated ex vivo IgG-[L]-scFv bispecific antibody (BsAb)-armed T cells (EATs) targeting GD2 or HER2. Both EATs induced durable antitumor responses and prolonged survival in osteosarcoma and neuroblastoma PDX mouse models, and autologous T cells were equally effective. Compared to separate BsAb and T cell mixturess, EATs demonstrated reduced cytokine release and more rapid T cell infiltration. The in vivo potency of EATs increased with higher BsAb dose for arming, and with higher infused EAT cell number and frequency. EATs retained their antitumor efficacy after cryopreservation. Allogeneic γδ cells were equally effective as αβ T cells.

Contributed by Shishir Pant

Background: T cell-based immunotherapies using chimeric antigen receptors (CAR) or bispecific antibodies (BsAb) have produced impressive responses in hematological malignancies. However, major hurdles remained, including cytokine release syndrome, neurotoxicity, on-target off-tumor effects, reliance on autologous T cells, and failure in most solid tumors. BsAb armed T cells offer a safe alternative.
Methods:
We generated ex vivo armed T cells (EATs) using IgG-[L]-scFv-platformed BsAb, where the anti-CD3 (huOKT3) scFv was attached to the light chain of a tumor-binding IgG. BsAb density on EAT, in vitro cytotoxicity, cytokine release, in vivo trafficking into tumors, and their antitumor activities were evaluated in multiple cancer cell lines and patient-derived xenograft mouse models. The efficacy of EATs after cryopreservation was studied, and gamma delta (γδ) T cells were investigated as unrelated alternative effector T cells.
Results:
The antitumor potency of BsAb armed T cells was substantially improved using the IgG-[L]-scFv BsAb platform. When compared with separate BsAb and T cell injection, EATs released less TNF-α, and infiltrated tumors faster, while achieving robust antitumor responses. The in vivo potency of EAT therapy depended on BsAb dose for arming, EAT cell number per injection, total number of EAT doses, and treatment schedule intensity. The antitumor efficacy of EATs was preserved following cryopreservation, and EATs using γδ T cells were safe and as effective as αβ T cell-EATs.
Conclusions:
EATs exerted potent antitumor activities against a broad spectrum of human cancer targets with remarkable safety. The antitumor potency of EATs depended on BsAb dose, cell number and total dose, and schedule. EATs were equally effective after cryopreservation, and the feasibility of third-party γδ-EATs offered an alternative for autologous T cell sources.

Author Info: (1) Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA. (2) Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA. (3) Pediatrics, Me

Author Info: (1) Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA. (2) Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA. (3) Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA. (4) Medicine, University of Virginia, Charlottesville, Virginia, USA. (5) Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA cheungn@mskcc.org.