Targeting PARP11 to avert immunosuppression and improve CAR T therapy in solid tumors

Nat Cancer. 2022 Jul;3(7):808-820. doi: 10.1038/s43018-022-00383-0. Epub 2022 May 30.

Abstract

Evasion of antitumor immunity and resistance to therapies in solid tumors are aided by an immunosuppressive tumor microenvironment (TME). We found that TME factors, such as regulatory T cells and adenosine, downregulated type I interferon receptor IFNAR1 on CD8+ cytotoxic T lymphocytes (CTLs). These events relied upon poly-ADP ribose polymerase-11 (PARP11), which was induced in intratumoral CTLs and acted as a key regulator of the immunosuppressive TME. Ablation of PARP11 prevented loss of IFNAR1, increased CTL tumoricidal activity and inhibited tumor growth in an IFNAR1-dependent manner. Accordingly, genetic or pharmacologic inactivation of PARP11 augmented the therapeutic benefits of chimeric antigen receptor T cells. Chimeric antigen receptor CTLs engineered to inactivate PARP11 demonstrated a superior efficacy against solid tumors. These findings highlight the role of PARP11 in the immunosuppressive TME and provide a proof of principle for targeting this pathway to optimize immune therapies.

Publication types

  • Research Support, N.I.H., Extramural
  • Research Support, Non-U.S. Gov't

MeSH terms

  • Humans
  • Immunosuppression Therapy
  • Immunotherapy, Adoptive
  • Neoplasms* / drug therapy
  • Poly(ADP-ribose) Polymerases / metabolism*
  • Receptors, Chimeric Antigen* / genetics
  • Tumor Microenvironment

Substances

  • Receptors, Chimeric Antigen
  • Poly(ADP-ribose) Polymerases