4.7 Article

Multifaceted modes of action of the anticancer probiotic Enterococcus hirae

Journal

CELL DEATH AND DIFFERENTIATION
Volume 28, Issue 7, Pages 2276-2295

Publisher

SPRINGERNATURE
DOI: 10.1038/s41418-021-00753-8

Keywords

-

Funding

  1. Fondation pour la Recherche Medicale (FRM)
  2. Ligue contre le Cancer (equipes labellisees)
  3. Agence National de la Recherche (ANR) [ANR-10-COHO-0004]
  4. Chancelerie des universites de Paris (Legs Poix), FRM
  5. European Research Area Network on Cardiovascular Diseases (ERA-CVD, MINOTAUR)
  6. Gustave Roussy Odyssea
  7. European Union Horizon 2020 Project Oncobiome
  8. Fondation Carrefour
  9. High-end Foreign Expert Program in China [GDW20171100085, GDW20181100051]
  10. Institut National du Cancer (INCa)
  11. Inserm (HTE)
  12. Institut Universitaire de France
  13. LeDucq Foundation
  14. LabEx Immuno-Oncology [ANR-18-IDEX0001]
  15. RHU Torino Lumiere [ANR-16-RHUS-0008]
  16. Seerave Foundation
  17. SIRIC Stratified Oncology Cell DNA Repair and Tumor Immune Elimination (SOCRATE)
  18. SIRIC Cancer Research and Personalized Medicine (CARPEM)
  19. Association pour la recherche sur le cancer (ARC)
  20. Canceropole Ile-de-France

Ask authors/readers for more resources

The study demonstrates that the gut microbiome's deviation is associated with resistance to cancer immunotherapy, and Enterococcus hirae can compensate for tumor-associated dysbiosis in various cancer models by stimulating T cell responses, activating autophagy, and altering the host microbiome.
A deviated repertoire of the gut microbiome predicts resistance to cancer immunotherapy. Enterococcus hirae compensated cancer-associated dysbiosis in various tumor models. However, the mechanisms by which E. hirae restored the efficacy of cyclophosphamide administered with concomitant antibiotics remain ill defined. Here, we analyzed the multifaceted modes of action of this anticancer probiotic. Firstly, E. hirae elicited emigration of thymocytes and triggered systemic and intratumoral IFN gamma-producing and CD137-expressing effector memory T cell responses. Secondly, E. hirae activated the autophagy machinery in enterocytes and mediated ATG4B-dependent anticancer effects, likely as a consequence of its ability to increase local delivery of polyamines. Thirdly, E. hirae shifted the host microbiome toward a Bifidobacteria-enriched ecosystem. In contrast to the live bacterium, its pasteurized cells or membrane vesicles were devoid of anticancer properties. These pleiotropic functions allow the design of optimal immunotherapies combining E. hirae with CD137 agonistic antibodies, spermidine, or Bifidobacterium animalis. We surmise that immunological, metabolic, epithelial, and microbial modes of action of the live E. hirae cooperate to circumvent primary resistance to therapy.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available