Abstract
Several preclinical studies demonstrate that antitumor efficacy of programmed cell death-1 (PD-1)/programmed death-ligand 1 (PD-L1) blockade can be improved by combination with other checkpoint inhibitors. Lymphocyte-activation gene 3 (LAG-3) is an inhibitory checkpoint receptor involved in T cell exhaustion and tumor immune escape. Here, we describe ABL501, a bispecific antibody targeting LAG-3 and PD-L1 in modulating immune cell responses against tumors. ABL501 that efficiently inhibits both LAG-3 and PD-L1 pathways enhances the activation of effector CD4+ and CD8+ T cells with a higher degree than a combination of single anti-LAG-3 and anti-PD-L1. The augmented effector T cell responses by ABL501 resulted in mitigating regulatory-T-cell-mediated immunosuppression. Mechanistically, the simultaneous binding of ABL501 to LAG-3 and PD-L1 promotes dendritic cell (DC) activation and tumor cell conjugation with T cells that subsequently mounts effective CD8+ T cell responses. ABL501 demonstrates its potent in vivo antitumor efficacy in a humanized xenograft model and with knockin mice expressing human orthologs. The immune profiling analysis of peripheral blood reveals an increased abundance of LAG-3hiPD-1hi memory CD4+ T cell subset in relapsed cholangiocarcinoma patients after gemcitabine plus cisplatin therapy, which are more responsive to ABL501. This study supports the clinical evaluation of ABL501 as a novel cancer immunotherapeutic, and a first-in-human trial has started (NCT05101109).
Original language | English |
---|---|
Pages (from-to) | 2800-2816 |
Number of pages | 17 |
Journal | Molecular Therapy |
Volume | 30 |
Issue number | 8 |
DOIs | |
Publication status | Published - 2022 Aug 3 |
Bibliographical note
Funding Information:We thank the core facilities of Genetically Engineered Animal Core and flow cytometry at the Convergence Medicine Research Center (CREDIT), Asan Medical Center. This work was supported by an intramural grant of KIST, by the National Research Foundation of Korea grants (NRF-2021R1A2C1003551 to H.J. NRF-2020M3A9G7103935 to H.J. and NRF-2021R1A2C2006647 to Y.P.), and by a grant of the Korea Health Technology R&D Project through the Korea Health Industry Development Institute, funded by the Ministry of Health & Welfare of South Korea (HI20C0117 to J. Jung). Conception and design, E.S. E.P. Jonghwa Won, H.J. J. Jung, and Y.P.; development of methodology, E.S. M.K. H.J. J. Jung, and Y.P.; acquisition of data, Ju-young Won, Y.J. E.C. U.J. J. Jeon, Y.K. H.A. D.C. S.C. Y.H. H.P. H.L. Y.-G.S. and K.P.; technical and material support, S.J.O. S.L. K.K. C.Y. and H.K.S.; analysis and interpretation of data, E.S. E.P. H.K. H.P. H.L. Jonghwa Won, H.J. J. Jung, and Y.P.; writing the manuscript, E.S. H.J. J. Jung, and Y.P.; study supervision, H.J. J. Jung, and Y.P. E.S. E.P. H.K. U.J. J. Jeon, Y.K. Y.H. H.P. H.L. Y.-G.S. K.P. Jonghwa Won, and J. Jung are employees of ABL Bio Inc. The other authors declare no competing interests.
Funding Information:
We thank the core facilities of Genetically Engineered Animal Core and flow cytometry at the Convergence Medicine Research Center (CREDIT), Asan Medical Center. This work was supported by an intramural grant of KIST , by the National Research Foundation of Korea grants ( NRF-2021R1A2C1003551 to H.J., NRF-2020M3A9G7103935 to H.J., and NRF-2021R1A2C2006647 to Y.P.), and by a grant of the Korea Health Technology R&D Project through the Korea Health Industry Development Institute , funded by the Ministry of Health & Welfare of South Korea ( HI20C0117 to J. Jung).
Publisher Copyright:
© 2022 The Author(s)
Keywords
- LAG-3
- PD-L1
- bispecific antibody
- cancer immunotherapy
- cholangiocarcinoma
- immune checkpoint inhibitor
ASJC Scopus subject areas
- Molecular Medicine
- Molecular Biology
- Genetics
- Pharmacology
- Drug Discovery