1. Academic Validation
  2. Dual-aptamer-engineered M1 macrophage with enhanced specific targeting and checkpoint blocking for solid-tumor immunotherapy

Dual-aptamer-engineered M1 macrophage with enhanced specific targeting and checkpoint blocking for solid-tumor immunotherapy

  • Mol Ther. 2022 Aug 3;30(8):2817-2827. doi: 10.1016/j.ymthe.2022.04.015.
Husun Qian 1 Yixin Fu 1 Minkang Guo 2 Yu Chen 2 Dian Zhang 1 Yu Wei 1 Fangfang Jin 1 Qian Zeng 1 Yange Wang 1 Chengsen Chai 1 Shijia Ding 1 Wei Cheng 3 Tingmei Chen 4
Affiliations

Affiliations

  • 1 Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, P.R. China.
  • 2 Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
  • 3 The Center for Clinical Molecular Medical Detection, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China. Electronic address: [email protected].
  • 4 Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, P.R. China. Electronic address: [email protected].
Abstract

Chimeric antigen receptor T (CAR-T) cell therapy has faced a series of challenges and has shown very little efficacy in solid tumors to date. Although genetically engineered macrophages have achieved definite therapeutic effect in solid tumors, heterogeneous expression of engineered proteins and the potential for toxicity limit further applications. Herein, we propose a nongenetic and simple macrophage cell engineering strategy through glycan metabolic labeling and click reaction for the treatment of solid tumors. The aptamer-engineered M1 macrophage (ApEn-M1) showed enhanced active targeting ability for tumor cells in vitro and in vivo, resulting in significant cytotoxicity effects. Moreover, ApEn-M1 exhibited superior antitumor efficacy in a breast Cancer xenograft mouse model and a lung metastasis mouse model of breast Cancer. Interestingly, the ApEn-M1 could reprogram the immunity microenvironment by increasing T cell infiltration and enhancing T cell activity in the tumor region. Additionally, the administration of ApEn-M1 showed no obvious systemic side effects. With glycan metabolic labeling, the macrophages could be efficiently labeled with Aptamers on the cell surface via click reaction without genetic alteration or cell damage. Hence, this study serves as a proof of concept for cell-surface anchor engineering and expands the range of nongenetic macrophage cell engineering strategies.

Keywords

aptamer engineering; immunotherapy; macrophage cells; metabolic glycan labeling; solid-tumor treatment.

Figures
Products