6,200
Views
6
CrossRef citations to date
0
Altmetric
Original Research

A novel bispecific antibody for EGFR-directed blockade of the PD-1/PD-L1 immune checkpoint

, , , , , , & show all
Article: e1466016 | Received 24 Jan 2018, Accepted 11 Apr 2018, Published online: 31 May 2018
 

ABSTRACT

PD-L1-blocking antibodies produce significant clinical benefit in selected cancer patients by reactivating functionally-impaired antigen-experienced anticancer T cells. However, the efficacy of current PD-L1-blocking antibodies is potentially reduced by ‘on-target/off-tumor’ binding to PD-L1 widely expressed on normal cells. This lack of tumor selectivity may induce a generalized activation of all antigen-experienced T cells which may explain the frequent occurrence of autoimmune-related adverse events during and after treatment.

To address these issues, we constructed a bispecific antibody (bsAb), designated PD-L1xEGFR, to direct PD-L1-blockade to EGFR-expressing cancer cells and to more selectively reactivate anticancer T cells. Indeed, the IC50 of PD-L1xEGFR for blocking PD-L1 on EGFR+ cancer cells was ∼140 fold lower compared to that of the analogous PD-L1-blocking bsAb PD-L1xMock with irrelevant target antigen specificity. Importantly, activation status, IFN-γ production, and oncolytic activity of anti-CD3xanti-EpCAM-redirected T cells was enhanced when cocultured with EGFR-expressing carcinoma cells. Similarly, the capacity of PD-L1xEGFR to promote proliferation and IFN-γ production by CMVpp65-directed CD8+ effector T cells was enhanced when cocultured with EGFR-expressing CMVpp65-transfected cancer cells. In contrast, the clinically-used PD-L1-blocking antibody MEDI4736 (durvalumab) promoted T cell activation indiscriminate of EGFR expression on cancer cells. Additionally, in mice xenografted with EGFR-expressing cancer cells 111In-PD-L1xEGFR showed a significantly higher tumor uptake compared to 111In-PD-L1xMock. In conclusion, PD-L1xEGFR blocks the PD-1/PD-L1 immune checkpoint in an EGFR-directed manner, thereby promoting the selective reactivation of anticancer T cells. This novel targeted approach may be useful to enhance efficacy and safety of PD-1/PD-L1 checkpoint blockade in EGFR-overexpressing malignancies.

Conflicts of interest

No potential conflicts of interest were disclosed.

Author contributions

IK, DH, DFS performed experiments, analyzed data, and wrote the manuscript. RJG, SH and JLW analyzed data. E.B and W.H conceived and designed the study and wrote the manuscript.

Additional information

Funding

This work was supported by the Dutch Cancer Society, (RUG2014-6986), (RUG2013-6209), (RUG2012-5541), Netherlands Organisation for scientific research (NWO 91617039) and the UMCG Cancer Foundation.