Your browser doesn't support javascript.
loading
A polymeric nanoplatform enhances the cGAS-STING pathway in macrophages to potentiate phagocytosis for cancer immunotherapy.
Li, Yongjuan; Yi, Jinmeng; Ma, Rong; Wang, Yayun; Lou, Xiaohan; Dong, Ya; Cao, Yongjian; Li, Xinyan; Wang, Ming; Dang, Xiaowei; Li, Rui; Lei, Ningjing; Song, Haiwei; Qin, Zhihai; Yang, Weijing.
Afiliação
  • Li Y; Medical Research Center, The First Affiliated Hospital of Zhengzhou University, The center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; School of basic medical sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
  • Yi J; Medical Research Center, The First Affiliated Hospital of Zhengzhou University, The center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; School of basic medical sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
  • Ma R; School of Pharmaceutical Sciences, Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, Zhengzhou University, Zhengzhou 450001, China.
  • Wang Y; School of Pharmaceutical Sciences, Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, Zhengzhou University, Zhengzhou 450001, China.
  • Lou X; Medical Research Center, The First Affiliated Hospital of Zhengzhou University, The center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
  • Dong Y; Medical Research Center, The First Affiliated Hospital of Zhengzhou University, The center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
  • Cao Y; School of Pharmaceutical Sciences, Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, Zhengzhou University, Zhengzhou 450001, China.
  • Li X; Medical Research Center, The First Affiliated Hospital of Zhengzhou University, The center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
  • Wang M; Medical Research Center, The First Affiliated Hospital of Zhengzhou University, The center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
  • Dang X; Medical Research Center, The First Affiliated Hospital of Zhengzhou University, The center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
  • Li R; Medical Research Center, The First Affiliated Hospital of Zhengzhou University, The center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
  • Lei N; School of basic medical sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
  • Song H; Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), Singapore, 138673, Singapore. Electronic address: haiwei@imcb.a-star.edu.sg.
  • Qin Z; Medical Research Center, The First Affiliated Hospital of Zhengzhou University, The center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China. Electronic address: zhihai@ibp.ac.cn.
  • Yang W; Medical Research Center, The First Affiliated Hospital of Zhengzhou University, The center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; School of Pharmaceutical Sciences, Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and
J Control Release ; 373: 447-462, 2024 Jul 24.
Article em En | MEDLINE | ID: mdl-39038546
ABSTRACT
Immunosuppressive tumor-associated macrophages (TAMs) account for a high proportion of the tumor tissue and significantly impede immunoefficacy. Furthermore, the signal regulatory protein α (SIRPα) expressed in TAMs adversely correlates with macrophage activation and phagocytosis, resulting in immunosurveillance escape. To address these difficulties, a mannose-modified, pH-responsive nanoplatform with resiquimod (R848) and 2', 3'-cyclic GMP-AMP (cGAMP) co-encapsulation (named M-PNP@R@C) is designed to polarize TAMs and lower SIRPα expression. The co-delivery of R848 and cGAMP synergistically facilitates the polarization of TAMs from the anti-inflammatory M2 phenotype into the pro-inflammatory M1 phenotype, thereby enhancing antitumor immunotherapy. Remarkably, activation of the cGAMP-mediated stimulator of interferon genes (STING) in TAMs significantly downregulates the expression of SIRPα, which synergizes with the cluster of differentiation 47 (CD47) antibody for the dual blockade of the CD47-SIRPα axis. Further analysis of single-cell RNA sequencing indicates that STING activation downregulates SIRPα by regulating intracellular fatty acid oxidation metabolism. In vivo studies indicate that M-PNP@R@C significantly inhibits tumor growth with a potent antitumor immune response in melanoma graft tumor models. After synergy with anti-CD47, the double blockade strategies of the SIRPα/CD47 axis result in a notable inhibition of lung metastasis. A prolonged survival rate is observed after combination treatment with CD47 and programmed death ligand-1 antibodies for the triple immune checkpoint blockade. In summary, our study provides original insights into the potential role of the STING pathway in macrophage-based immunotherapy, thus offering a potential combinatorial strategy for cancer therapy.
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Idioma: En Revista: J Control Release Assunto da revista: FARMACOLOGIA Ano de publicação: 2024 Tipo de documento: Article País de afiliação: China

Texto completo: 1 Base de dados: MEDLINE Idioma: En Revista: J Control Release Assunto da revista: FARMACOLOGIA Ano de publicação: 2024 Tipo de documento: Article País de afiliação: China