Molecular Cancer (Aug 2023)

Unleashing T cell anti-tumor immunity: new potential for 5-Nonloxytryptamine as an agent mediating MHC-I upregulation in tumors

  • Paweł Stachura,
  • Wei Liu,
  • Haifeng C. Xu,
  • Agnès Wlodarczyk,
  • Olivia Stencel,
  • Piyush Pandey,
  • Melina Vogt,
  • Sanil Bhatia,
  • Daniel Picard,
  • Marc Remke,
  • Karl S. Lang,
  • Dieter Häussinger,
  • Bernhard Homey,
  • Philipp A. Lang,
  • Arndt Borkhardt,
  • Aleksandra A. Pandyra

DOI
https://doi.org/10.1186/s12943-023-01833-8
Journal volume & issue
Vol. 22, no. 1
pp. 1 – 20

Abstract

Read online

Abstract Background New therapies are urgently needed in melanoma, particularly in late-stage patients not responsive to immunotherapies and kinase inhibitors. To uncover novel potentiators of T cell anti-tumor immunity, we carried out an ex vivo pharmacological screen and identified 5-Nonyloxytryptamine (5-NL), a serotonin agonist, as increasing the ability of T cells to target tumor cells. Methods The pharmacological screen utilized lymphocytic choriomeningitis virus (LCMV)-primed splenic T cells and melanoma B16.F10 cells expressing the LCMV gp33 CTL epitope. In vivo tumor growth in C57BL/6 J and NSG mice, in vivo antibody depletion, flow cytometry, immunoblot, CRISPR/Cas9 knockout, histological and RNA-Seq analyses were used to decipher 5-NL’s immunomodulatory effects in vitro and in vivo. Results 5-NL delayed tumor growth in vivo and the phenotype was dependent on the hosts’ immune system, specifically CD8+ T cells. 5-NL’s pro-immune effects were not directly consequential to T cells. Rather, 5-NL upregulated antigen presenting machinery in melanoma and other tumor cells in vitro and in vivo without increasing PD-L1 expression. Mechanistic studies indicated that 5-NL’s induced MHC-I expression was inhibited by pharmacologically preventing cAMP Response Element-Binding Protein (CREB) phosphorylation. Importantly, 5-NL combined with anti-PD1 therapy showed significant improvement when compared to single anti-PD-1 treatment. Conclusions This study demonstrates novel therapeutic opportunities for augmenting immune responses in poorly immunogenic tumors.

Keywords