Kraman, Faroudi, and Allen et al. carried out studies with FS118, a human, tetravalent, high-affinity bispecific antibody targeting LAG3 and PD-L1, and an equivalent murine bispecific surrogate (mLAG3/PD-L1mAb2), and compared them to combination therapy with single PD-L1 and LAG3 mAbs. FS118 had potent activity in LAG3/PD-L1-dependent T cell assays, reversed immune suppression, and enhanced IFNγ levels. In syngeneic tumor models, compared to the antibody combination, mLAG-3/PD-L1mAb2 comparably suppressed tumor growth, but decreased, rather than increased, surface LAG3, suggesting a differentiated mechanism.

Contributed by Katherine Turner

PURPOSE: Although programmed death-(ligand) 1 (PD-(L)1) antibody-based therapy has improved the outcome of cancer patients, acquired resistance to these treatments limits their clinical efficacy. FS118 is a novel bispecific, tetravalent antibody (mAb(2)) against human Lymphocyte Activation Gene 3 (LAG-3) and PD-L1 with the potential to reinvigorate exhausted immune cells and overcome resistance mechanisms to PD-(L)1 blockade. Here, using FS118 and a murine surrogate, we characterised the activity and report a novel mechanism of action of this bispecific antibody. EXPERIMENTAL DESIGN: This study characterises the binding activity and immune function of FS118 in cell lines and human peripheral blood mononuclear cells and further investigates its anti-tumour activity and mechanism of action using a surrogate murine bispecific antibody (mLAG-3/PD-L1 mAb(2)). RESULTS: FS118 demonstrated simultaneous binding to LAG-3 and PD-L1 with high affinity and comparable or better activity than the combination of the single component parts of the mAb(2) in blocking LAG-3 and PD-L1-mediated immune suppression and enhancing T cell activity. In syngeneic tumour mouse models, mLAG-3/PD-L1 mAb(2) significantly suppressed tumour growth. Mechanistic studies revealed decreased LAG-3 expression on T cells following treatment with the mouse surrogate mLAG-3/PD-L1 mAb(2), whereas LAG-3 expression increased upon treatment with the combination of monoclonal antibodies targeting LAG-3 and PD-L1. Moreover, following binding of mLAG-3/PD-L1 mAb(2) to target expressing cells, mouse LAG-3 is rapidly shed into the blood. CONCLUSIONS: This study demonstrates a novel benefit of the bispecific approach over a combination of monoclonal antibodies and supports the further development of FS118 for the treatment of cancer patients.

Author Info: (1) Research, F-star Biotechnology Ltd. (2) Research, F-star Biotechnology Ltd. (3) Research, F-star Biotechnology Ltd. (4) Research, F-star Biotechnology Ltd. (5) Research, F-star

Author Info: (1) Research, F-star Biotechnology Ltd. (2) Research, F-star Biotechnology Ltd. (3) Research, F-star Biotechnology Ltd. (4) Research, F-star Biotechnology Ltd. (5) Research, F-star Biotechnology Ltd. (6) Research, F-star Biotechnology Ltd. (7) Preclinical Translational Pharmacology, F-star Biotechnology Ltd. (8) Research, F-star Biotechnology Ltd. (9) Research, F-star Biotechnology Ltd. (10) Research, F-star Therapeutics Ltd. (11) DEvelopment, F-star Biotechnology Ltd. (12) Research, Adaptate Biotherapeutics. (13) Research, F-star Biotechnology Ltd. (14) Research, F-star Therapeutics Ltd michelle.morrow@f-star.com. (15) Research, F-star Biotechnology.