Your browser doesn't support javascript.
loading
Perfluorocarbon nanoparticle-mediated platelet inhibition promotes intratumoral infiltration of T cells and boosts immunotherapy.
Zhou, Zaigang; Zhang, Baoli; Zai, Wenjing; Kang, Lin; Yuan, Ahu; Hu, Yiqiao; Wu, Jinhui.
Afiliação
  • Zhou Z; State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University & School of Life Sciences, Nanjing University, 210093 Nanjing, China.
  • Zhang B; Institute of Drug R&D, Nanjing University, 210093 Nanjing, China.
  • Zai W; State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University & School of Life Sciences, Nanjing University, 210093 Nanjing, China.
  • Kang L; Institute of Drug R&D, Nanjing University, 210093 Nanjing, China.
  • Yuan A; State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University & School of Life Sciences, Nanjing University, 210093 Nanjing, China.
  • Hu Y; Institute of Drug R&D, Nanjing University, 210093 Nanjing, China.
  • Wu J; State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University & School of Life Sciences, Nanjing University, 210093 Nanjing, China.
Proc Natl Acad Sci U S A ; 116(24): 11972-11977, 2019 06 11.
Article em En | MEDLINE | ID: mdl-31142648
ABSTRACT
Cancer immunotherapy can stimulate and enhance the ability of the immune system to recognize, arrest, and eliminate tumor cells. Immune checkpoint therapies (e.g., PD-1/PD-L1) have shown an unprecedented and durable clinical response rate in patients among various cancer types. However, a large fraction of patients still does not respond to these checkpoint inhibitors. The main cause of this phenomenon is the limited T-cell infiltration in tumors. Therefore, additional strategies to enhance T-cell trafficking into tumors are urgently needed to improve patients' immune responses. In this study, we screened an array of perfluorocarbon compounds, reporting that albumin-based perfluorotributylamine nanoparticles (PFTBA@Alb) can effectively increase the permeability of tumor blood vessels, and no distinct side effects were found on normal blood vessels. After i.v. administration of PFTBA@Alb, the number of tumor-infiltrating CD8+ and CD4+ T cells showed an obvious rising trend. More important, a striking tumor inhibition rate, reaching nearly 90%, was observed when combining PFTBA@Alb with anti-PD-L1 antibody. These findings suggest that PFTBA@Alb can be regarded as an enhancer for anti-PD-L1 immunotherapy.
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Idioma: En Revista: Proc Natl Acad Sci U S A Ano de publicação: 2019 Tipo de documento: Article País de afiliação: China

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Idioma: En Revista: Proc Natl Acad Sci U S A Ano de publicação: 2019 Tipo de documento: Article País de afiliação: China