PLoS Pathogens (Jul 2021)

Interleukin-15 response signature predicts RhCMV/SIV vaccine efficacy.

  • Fredrik Barrenäs,
  • Scott G Hansen,
  • Lynn Law,
  • Connor Driscoll,
  • Richard R Green,
  • Elise Smith,
  • Jean Chang,
  • Inah Golez,
  • Taryn Urion,
  • Xinxia Peng,
  • Leanne Whitmore,
  • Daniel Newhouse,
  • Colette M Hughes,
  • David Morrow,
  • Kurt T Randall,
  • Andrea N Selseth,
  • Julia C Ford,
  • Roxanne M Gilbride,
  • Bryan E Randall,
  • Emily Ainslie,
  • Kelli Oswald,
  • Rebecca Shoemaker,
  • Randy Fast,
  • William J Bosche,
  • Michael K Axthelm,
  • Yoshinori Fukazawa,
  • George N Pavlakis,
  • Barbara K Felber,
  • Slim Fourati,
  • Rafick-Pierre Sekaly,
  • Jeffrey D Lifson,
  • Jan Komorowski,
  • Ewelina Kosmider,
  • Danica Shao,
  • Wenjun Song,
  • Paul T Edlefsen,
  • Louis J Picker,
  • Michael Gale

DOI
https://doi.org/10.1371/journal.ppat.1009278
Journal volume & issue
Vol. 17, no. 7
p. e1009278

Abstract

Read online

Simian immunodeficiency virus (SIV) challenge of rhesus macaques (RMs) vaccinated with strain 68-1 Rhesus Cytomegalovirus (RhCMV) vectors expressing SIV proteins (RhCMV/SIV) results in a binary outcome: stringent control and subsequent clearance of highly pathogenic SIV in ~55% of vaccinated RMs with no protection in the remaining 45%. Although previous work indicates that unconventionally restricted, SIV-specific, effector-memory (EM)-biased CD8+ T cell responses are necessary for efficacy, the magnitude of these responses does not predict efficacy, and the basis of protection vs. non-protection in 68-1 RhCMV/SIV vector-vaccinated RMs has not been elucidated. Here, we report that 68-1 RhCMV/SIV vector administration strikingly alters the whole blood transcriptome of vaccinated RMs, with the sustained induction of specific immune-related pathways, including immune cell, toll-like receptor (TLR), inflammasome/cell death, and interleukin-15 (IL-15) signaling, significantly correlating with subsequent vaccine efficacy. Treatment of a separate RM cohort with IL-15 confirmed the central involvement of this cytokine in the protection signature, linking the major innate and adaptive immune gene expression networks that correlate with RhCMV/SIV vaccine efficacy. This change-from-baseline IL-15 response signature was also demonstrated to significantly correlate with vaccine efficacy in an independent validation cohort of vaccinated and challenged RMs. The differential IL-15 gene set response to vaccination strongly correlated with the pre-vaccination activity of this pathway, with reduced baseline expression of IL-15 response genes significantly correlating with higher vaccine-induced induction of IL-15 signaling and subsequent vaccine protection, suggesting that a robust de novo vaccine-induced IL-15 signaling response is needed to program vaccine efficacy. Thus, the RhCMV/SIV vaccine imparts a coordinated and persistent induction of innate and adaptive immune pathways featuring IL-15, a known regulator of CD8+ T cell function, that support the ability of vaccine-elicited unconventionally restricted CD8+ T cells to mediate protection against SIV challenge.