Login / Signup

TREM2 deficiency reprograms intestinal macrophages and microbiota to enhance anti-PD-1 tumor immunotherapy.

Blanda Di LucciaMartina MolgoraDarya KhantakovaNatalia JaegerHao-Wei ChangRafael S CzepielewskiBeth A HelminkEmily J OnuferJosé Luís FachiBishan BhattaraiTihana TrsanPatrick Fernandes RodriguesJin-Chao HouJennifer K BandoCristiane Sécca da SilvaMarina CellaSusan GilfillanRobert D SchreiberJeffrey I GordonMarco Colonna
Published in: Science immunology (2024)
The gut microbiota and tumor-associated macrophages (TAMs) affect tumor responses to anti-programmed cell death protein 1 (PD-1) immune checkpoint blockade. Reprogramming TAM by either blocking or deleting the macrophage receptor triggering receptor on myeloid cells 2 (TREM2) attenuates tumor growth, and lack of functional TREM2 enhances tumor elimination by anti-PD-1. Here, we found that anti-PD-1 treatment combined with TREM2 deficiency in mice induces proinflammatory programs in intestinal macrophages and a concomitant expansion of Ruminococcus gnavus in the gut microbiota. Gavage of wild-type mice with R. gnavus enhanced anti-PD-1-mediated tumor elimination, recapitulating the effect occurring in the absence of TREM2. A proinflammatory intestinal environment coincided with expansion, increased circulation, and migration of TNF-producing CD4 + T cells to the tumor bed. Thus, TREM2 remotely controls anti-PD-1 immune checkpoint blockade through modulation of the intestinal immune environment and microbiota, with R. gnavus emerging as a potential probiotic agent for increasing responsiveness to anti-PD-1.
Keyphrases
  • wild type
  • rheumatoid arthritis
  • adipose tissue
  • induced apoptosis
  • oxidative stress
  • metabolic syndrome
  • small molecule
  • cell proliferation
  • combination therapy
  • cell cycle arrest
  • pi k akt
  • protein protein
  • human health