Your browser doesn't support javascript.
loading
Inhibition of glycolysis-driven immunosuppression with a nano-assembly enhances response to immune checkpoint blockade therapy in triple negative breast cancer.
Ren, Xijiao; Cheng, Zhuo; He, Jinming; Yao, Xuemei; Liu, Yingqi; Cai, Kaiyong; Li, Menghuan; Hu, Yan; Luo, Zhong.
Afiliação
  • Ren X; Key Laboratory of Biorheological Science and Technology, Ministry of Education, Chongqing University, Chongqing, 400044, PR China.
  • Cheng Z; School of Life Science, Chongqing University, Chongqing, 400044, PR China.
  • He J; School of Life Science, Chongqing University, Chongqing, 400044, PR China.
  • Yao X; School of Life Science, Chongqing University, Chongqing, 400044, PR China.
  • Liu Y; School of Life Science, Chongqing University, Chongqing, 400044, PR China.
  • Cai K; Key Laboratory of Biorheological Science and Technology, Ministry of Education, Chongqing University, Chongqing, 400044, PR China.
  • Li M; School of Life Science, Chongqing University, Chongqing, 400044, PR China. menghuanli@cqu.edu.cn.
  • Hu Y; Key Laboratory of Biorheological Science and Technology, Ministry of Education, Chongqing University, Chongqing, 400044, PR China. huyan303@cqu.edu.cn.
  • Luo Z; School of Life Science, Chongqing University, Chongqing, 400044, PR China. luozhong918@cqu.edu.cn.
Nat Commun ; 14(1): 7021, 2023 11 02.
Article em En | MEDLINE | ID: mdl-37919262
ABSTRACT
Immune-checkpoint inhibitors (ICI) are promising modalities for treating triple negative breast cancer (TNBC). However, hyperglycolysis, a hallmark of TNBC cells, may drive tumor-intrinsic PD-L1 glycosylation and boost regulatory T cell function to impair ICI efficacy. Herein, we report a tumor microenvironment-activatable nanoassembly based on self-assembled aptamer-polymer conjugates for the targeted delivery of glucose transporter 1 inhibitor BAY-876 (DNA-PAE@BAY-876), which remodels the immunosuppressive TME to enhance ICI response. Poly ß-amino ester (PAE)-modified PD-L1 and CTLA-4-antagonizing aptamers (aptPD-L1 and aptCTLA-4) are synthesized and co-assembled into supramolecular nanoassemblies for carrying BAY-876. The acidic tumor microenvironment causes PAE protonation and triggers nanoassembly dissociation to initiate BAY-876 and aptamer release. BAY-876 selectively inhibits TNBC glycolysis to deprive uridine diphosphate N-acetylglucosamine and downregulate PD-L1 N-linked glycosylation, thus facilitating PD-L1 recognition of aptPD-L1 to boost anti-PD-L1 therapy. Meanwhile, BAY-876 treatment also elevates glucose supply to tumor-residing regulatory T cells (Tregs) for metabolically rewiring them into an immunostimulatory state, thus cooperating with aptCTLA-4-mediated immune-checkpoint inhibition to abolish Treg-mediated immunosuppression. DNA-PAE@BAY-876 effectively reprograms the immunosuppressive microenvironment in preclinical models of TNBC in female mice and provides a distinct approach for TNBC immunotherapy in the clinics.
Assuntos

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Neoplasias de Mama Triplo Negativas Limite: Animals / Female / Humans Idioma: En Revista: Nat Commun Assunto da revista: BIOLOGIA / CIENCIA Ano de publicação: 2023 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Neoplasias de Mama Triplo Negativas Limite: Animals / Female / Humans Idioma: En Revista: Nat Commun Assunto da revista: BIOLOGIA / CIENCIA Ano de publicação: 2023 Tipo de documento: Article