Discovery of a new inhibitor targeting PD-L1 for cancer immunotherapy

Blockade of the PD-1/PD-L1 immunologic checkpoint using monoclonal antibodies has provided breakthrough therapies against cancer in the recent years. Nevertheless, intrinsic disadvantages of therapeutic antibodies may limit their applications. Thus, blocking of the PD-1/PD-L1 interaction by small mo...

Full description

Saved in:
Bibliographic Details
Published inNeoplasia (New York, N.Y.) Vol. 23; no. 3; pp. 281 - 293
Main Authors Wang, Fengling, Ye, Wenling, Wang, Shuang, He, Yongxing, Zhong, Haiyang, Wang, Yuwei, Zhu, Yongchang, Han, Jianting, Bing, Zhitong, Ji, Shaoping, Liu, Huanxiang, Yao, Xiaojun
Format Journal Article
LanguageEnglish
Published United States Elsevier Inc 01.03.2021
Neoplasia Press
Elsevier
Subjects
Online AccessGet full text

Cover

Loading…
More Information
Summary:Blockade of the PD-1/PD-L1 immunologic checkpoint using monoclonal antibodies has provided breakthrough therapies against cancer in the recent years. Nevertheless, intrinsic disadvantages of therapeutic antibodies may limit their applications. Thus, blocking of the PD-1/PD-L1 interaction by small molecules may be a promising alternative for cancer immunotherapy. We used a docking-based virtual screening strategy to rapidly identify new small molecular inhibitors targeting PD-L1. We demonstrated that a small molecule compound (N-[2-(aminocarbonyl)phenyl][1,1′-biphenyl]-4-carboxamide [APBC]) could effectively interrupt the PD-1/PD-L1 interaction by directly binding to PD-L1, presenting the KD and IC50 values at low-micromolar level. Molecular docking study revealed that APBC may have function through a PD-L1 dimer-locking mechanism, occluding the PD-1 interaction surface of PD-L1. We further confirmed the ligand blocking activity and T cell-reinvigoration potency of APBC using cell-based assays. APBC could dose-dependently elevate cytokine secretions of the primary T-lymphocytes that are cocultured with cancer cells. Importantly, APBC displayed superior antitumor efficacy in hPD-L1 knock-in B16F10-bearing mouse model without the induction of observable liver toxicity. Analyses on the APBC-treated mice further revealed drastically elevated levels of infiltrating CD4+ and CD8+ T cells, and inflammatory cytokines production in tumor microenvironment. The APBC compound could serve as a privileged scaffold in the design of improved PD pathway modulators, thus providing us promising drug candidates for tumor immunotherapy.
Bibliography:ObjectType-Article-1
SourceType-Scholarly Journals-1
ObjectType-Feature-2
content type line 23
These authors contributed equally to this article.
ISSN:1476-5586
1522-8002
1476-5586
DOI:10.1016/j.neo.2021.01.001