Research paper
Structure-activity relationship study of amidobenzimidazole derivatives as stimulator of interferon genes (STING) agonists

https://doi.org/10.1016/j.ejmech.2022.114943Get rights and content

Highlights

  • 88 compounds were designed and synthesized as amidobenzimidazole monomer STING agonists.

  • Compound 72 potently activated the STING Signaling in PBMCs.

  • Compound 72 showed high selectivity for human STING.

Abstract

Stimulator of interferon genes (STING) is a crucial adaptor protein that can regulate the innate immune response by inducing the secretion of type Ι interferons and other cytokines after recognizing endogenous or exogenous DNA. Due to the key role of STING in the innate immune system, the activation of STING pathway is expected to be an efficacious immunotherapeutic tactic to treat cancer. In this study, we performed a structure-activity relationship study of amidobenzimidazole monomer, led to a series of ABZI STING agonist derivatives with potent STING-activating effects. Among them, compound 72, as a representative compound, markedly activated the STING-TBK1-IRF3 signaling pathway and significantly increased the mRNA and protein levels of IFN-β, CXCL10 and IL-6 in both WT THP-1 cells and human peripheral blood mononuclear cells (hPBMCs). In addition, it was confirmed that compound 72 was highly selective for human STING, specifically targeting human STING signaling and showing no activation of m-STING.

Introduction

Stimulator of interferon gene (STING, also known as MITA, ERIS, or MPYS) is a stimulatory molecule residing on the endoplasmic reticulum (ER) [1]. As an adaptor protein, STING can regulate the innate immune response by linking the upstream DNA sensor cyclic GMP-AMP synthase (cGAS) with downstream recruitment of tank-binding kinase 1 (TBK1), leading to the activation of the IRF and NF-κB pathways and the expression of type I interferons (IFNs) and other inflammatory cytokines [[2], [3], [4]], such as CXCL10, TNF-α and IL-6. The research value of STING inhibitors in immunotherapy was presented [5]. In addition, activation of the STING pathway was found to be crucial for priming tumor-specific CD8+ T cells to eliminate tumor cells, and the release of IFN-β can promote dendritic cell (DC) maturation, making tumor antigen presentation for the cross-priming of CD8+ T cells possible [[6], [7]]. Therefore, STING agonists have received increasing attention as antitumor agents.

First-generation STING agonists are cyclic dinucleotide (CDN) derivatives, mimetics of the endogenous STING activator 2′,3′-cGAMP (1, Fig. 1A), and several have already entered clinical trials [[8], [9], [10], [11], [12], [13], [14], [15], [16], [17], [18], [19]]. However, because of the poor biochemical properties of CDN derivatives, most are limited to intratumoral administration. Some groups are working on developing non-CDN small-molecule agonists. DMXAA (2, Fig. 1B) [20], one of the first reported non-CDN small-molecule STING agonists, has high efficacy. However, it is a mouse-specific STING agonist that cannot bind to human STING [[21], [22], [23]]. More recently, other non-CDN STING agonists have been reported by several groups, including GSK (3, Fig. 1C) [[24], [25], [26]], Scripps (4, Fig. 1D) [27], Merck (5, Fig. 1E) [[28], [29], [30]], and others [31].

Our previous study reported a series of amidobenzimidazole monomer STING agonists and identified lead compound 6 with improved biochemical and cellular potency (with EC50 = 0.287 μM in the THP1-Dual cell line), but the in vivo antitumor potency was modest [32]. Additionally, compound 6 had poor stability and was easily metabolized (with a residual of 36.4% of the initial concentration after incubation with rat plasma for 1 h). Because of the significant antitumor efficacy of compound 6 in vitro and the limited structure-activity relationship (SAR) study reported, compound 6 was considered amenable to structural modifications, which we believe to be potentially capable of improving its biological activity in vivo. In this regard, structural optimization was performed to discover more potent amidobenzimidazole monomer STING agonists. Herein, we describe the design, synthesis, and biological evaluation of these new STING agonists.

Section snippets

Chemistry

The synthetic pathway of compounds 7–15 is shown in Scheme 1. Intermediate 1a was obtained by amination of methyl 4-chloro-3-methoxy-5-nitrobenzoate with an ammonia solution. Then, 1a was subjected to a substitution reaction with tert-butyl (E)-(4-aminobut-2-en-1-yl) carbamate-delivered intermediate 1b. Subsequent reduction of 1b was performed with sodium dithionite to afford intermediate 1c. Intermediate 1c was then reacted with 1-ethyl-3-methyl-1H-pyrazole-5-carbonyl isothiocyanate to merge

Chemical reagents and general method

Unless otherwise specified, all commercially available starting materials and solvents are reagent grade and used without further purification. All air-sensitive reactions were carried out under an atmosphere of argon with magnetic stirring. Melting points were determined on Yanaco MP-J3 microscope melting point apparatus. 1H NMR and 13C NMR spectra were recorded on Mercury-400, Mercury-500, and Mercury-700 spectrometers at room temperature. Chemical shifts (δ) were reported in ppm downfifield

Author contributions

Xiaojian Wang, Jing Jin, and Xiaoguang Chen designed this project. Xue Liu and Mingjin Wang assisted in designing this project. Xue Liu performed the chemical synthesis. Minjian Yang and Hanyu Sun assisted in chemical synthesis. Mingjin Wang performed in vitro assays of STING. Xue Liu, and Xiaojian Wang contributed to the writing, review and editing of the manuscript. All authors have given approval to the final version of the manuscript.

Declaration of competing interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Acknowledgments

This work was financially supported by the National Natural Science Foundation of China (NSFC no. 82073692), CAMS Innovation Fund for Medical Sciences (CIFMS 2021-1-I2M − 028).

References (34)

  • Safety and efficacy study of IMSA101 in refractory malignancies. Identifier: NCT04020185; Retrieved from:...
  • An investigational immunotherapy study of BMS-986301 alone or in combination with nivolumab, and ipilimumab in participants with advanced solid cancers. Identifier: NCT03956680

  • Efficacy and safety trial of ADU-S100 and pembrolizumab in head and neck cancer

  • Phase 1 first time in humans (FTIH), open label study of GSK3745417 administered to subjects with advanced solid tumors. 3537Identififier: NCT03843359

  • M.K.- Study of

    2118 administered as intratumoral injection as monotherapy and in combination with pembrolizumab (MK-3475) or by subcutaneous injection in combination with pembrolizumab in the treatment of adults with advanced/metastatic solid tumors or lymphomas (MK-2118-001)

  • Study of the safety and efficacy of MIW815 with PDR001 in patients with advanced/metastatic solid tumors or lymphomas. Identifier: NCT03172936

  • M.K.- Study of

    1454 alone or in combination with pembrolizumab (MK-3475) in participants with advanced/metastatic solid tumors or lymphomas (MK-1454-001)

  • Cited by (5)

    1

    These authors made equal contributions to this work.

    View full text