Advanced Science (May 2022)

Tumor Derived Extracellular Vesicles Drive T Cell Exhaustion in Tumor Microenvironment through Sphingosine Mediated Signaling and Impacting Immunotherapy Outcomes in Ovarian Cancer

  • Prachi Gupta,
  • Ishaque Pulikkal Kadamberi,
  • Sonam Mittal,
  • Shirng‐Wern Tsaih,
  • Jasmine George,
  • Sudhir Kumar,
  • Dileep K. Vijayan,
  • Anjali Geethadevi,
  • Deepak Parashar,
  • Paytsar Topchyan,
  • Lindsey McAlarnen,
  • Brian F Volkman,
  • Weiguo Cui,
  • Kam Y. J. Zhang,
  • Dolores Di Vizio,
  • Pradeep Chaluvally‐Raghavan,
  • Sunila Pradeep

DOI
https://doi.org/10.1002/advs.202104452
Journal volume & issue
Vol. 9, no. 14
pp. n/a – n/a

Abstract

Read online

Abstract SPHK1 (sphingosine kinase‐1) catalyzes the phosphorylation of sphingosine to sphingosine‐1‐phosphate (S1P), is found to be highly expressed in solid tumors. Here, extracellular vesicles (EVs) are identified as the key transporters of SPHK1 to the tumor microenvironment. Consequently, SPHK1‐packaged EVs elevate S1P levels in the tumor microenvironment, where S1P appears as an immunosuppressive agent. However, the exact mechanism of how S1P mediates its immunosuppressive effects in cancer is not understood. It is investigated that S1P can induce T cell exhaustion. S1P can also upregulate programmed death ligand‐1 (PDL‐1) expression through E2F1‐mediated transcription. Notably, an SPHK1 inhibitor PF543 improves T cell‐mediated cytotoxicity. Furthermore, combining PF543 with an anti‐PD‐1 antibody reduces tumor burden and metastasis more effectively than PF543 alone in vivo. These data demonstrate a previously unrecognized mechanism of how SPHK1‐packaged EVs contribute to the progression of ovarian cancer and thus present the potential clinical application of inhibiting SPHK1/S1P signaling to improve immune checkpoint blockage (anti‐PD‐1 antibody) therapy in ovarian cancer.

Keywords