Increased p53 expression induced by APR-246 reprograms tumor-associated macrophages to augment immune checkpoint blockade. Academic Article uri icon

Overview

abstract

  • In addition to playing a major role in tumor cell biology, p53 generates a microenvironment that promotes antitumor immune surveillance via tumor-associated macrophages. We examined whether increasing p53 signaling in the tumor microenvironment influences antitumor T cell immunity. Our findings indicate that increased p53 signaling induced either pharmacologically with APR-246 (eprenetapopt) or in p53-overexpressing transgenic mice can disinhibit antitumor T cell immunity and augment the efficacy of immune checkpoint blockade. We demonstrated that increased p53 expression in tumor-associated macrophages induces canonical p53-associated functions such as senescence and activation of a p53-dependent senescence-associated secretory phenotype. This was linked with decreased expression of proteins associated with M2 polarization by tumor-associated macrophages. Our preclinical data led to the development of a clinical trial in patients with solid tumors combining APR-246 with pembrolizumab. Biospecimens from select patients participating in this ongoing trial showed that there was a suppression of M2-polarized myeloid cells and increase in T cell proliferation with therapy in those who responded to the therapy. Our findings, based on both genetic and a small molecule-based pharmacological approach, suggest that increasing p53 expression in tumor-associated macrophages reprograms the tumor microenvironment to augment the response to immune checkpoint blockade.

publication date

  • September 15, 2022

Research

keywords

  • Immune Checkpoint Inhibitors
  • Tumor-Associated Macrophages

Identity

PubMed Central ID

  • PMC9479603

Scopus Document Identifier

  • 85137930590

Digital Object Identifier (DOI)

  • 10.1172/JCI148141

PubMed ID

  • 36106631

Additional Document Info

volume

  • 132

issue

  • 18