Abstract

BackgroundThe unique responsiveness of Vγ9Vδ2 T-cells, the major γδ subset of human peripheral blood, to non-peptidic prenyl pyrophosphate antigens constitutes the basis of current γδ T-cell-based cancer immunotherapy strategies. However, the molecular mechanisms responsible for phosphoantigen-mediated activation of human γδ T-cells remain unclear. In particular, previous reports have described a very slow kinetics of activation of T-cell receptor (TCR)-associated signal transduction pathways by isopentenyl pyrophosphate and bromohydrin pyrophosphate, seemingly incompatible with direct binding of these antigens to the Vγ9Vδ2 TCR. Here we have studied the most potent natural phosphoantigen yet identified, (E)-4-hydroxy-3-methyl-but-2-enyl pyrophosphate (HMB-PP), produced by Eubacteria and Protozoa, and examined its γδ T-cell activation and anti-tumor properties.Methodology/Principal FindingsWe have performed a comparative study between HMB-PP and the anti-CD3ε monoclonal antibody OKT3, used as a reference inducer of bona fide TCR signaling, and followed multiple cellular and molecular γδ T-cell activation events. We show that HMB-PP activates MEK/Erk and PI-3K/Akt pathways as rapidly as OKT3, and induces an almost identical transcriptional profile in Vγ9+ T-cells. Moreover, MEK/Erk and PI-3K/Akt activities are indispensable for the cellular effects of HMB-PP, including γδ T-cell activation, proliferation and anti-tumor cytotoxicity, which are also abolished upon antibody blockade of the Vγ9+ TCR Surprisingly, HMB-PP treatment does not induce down-modulation of surface TCR levels, and thereby sustains γδ T-cell activation upon re-stimulation. This ultimately translates in potent human γδ T-cell anti-tumor function both in vitro and in vivo upon transplantation of human leukemia cells into lymphopenic mice,Conclusions/SignificanceThe development of efficient cancer immunotherapy strategies critically depends on our capacity to maximize anti-tumor effector T-cell responses. By characterizing the intracellular mechanisms of HMB-PP-mediated activation of the highly cytotoxic Vγ9+ T-cell subset, our data strongly support the usage of this microbial antigen in novel cancer clinical trials.

Highlights

  • The capacity to recognize and eliminate transformed cells is common to several lymphocyte subsets of both the adaptive and the innate immune systems that are being targeted in cancer immunotherapy [1,2]

  • Consistent with such potential, it has been recently shown that hydroxy-3-methyl-but-2-enyl pyrophosphate (HMB-PP) has the capacity to induce the formation of high-density T-cell receptor (TCR) nanoclusters on the surface of human cd T-cells [16], and a newly-developed tetramer reagent for the Vc9Vd2 TCR of rhesus macaques was reported to bind to HMB-PP loaded on the surface of human antigen presenting cells (APC) [17]

  • The stimulatory effect prenyl pyrophosphates have on Vc9Vd2 T-cells has been well documented and seems to require TCR expression, as indicated by antibody blocking and gene transfer experiments [26,27]

Read more

Summary

Introduction

The capacity to recognize and eliminate transformed cells is common to several lymphocyte subsets of both the adaptive and the innate immune systems that are being targeted in cancer immunotherapy [1,2]. HMB-PP is respectively 30,000 and 100 times more potent than IPP and bromohydrin pyrophosphate (BrH-PP, known as ‘‘Phosphostim’’), most of the studies on phosphoantigens have been performed with these compounds (already applied in the clinic) due to their historical precedence [3] Such studies revealed a very slow kinetics of activation of TCR-associated signal transduction pathways, and conflicting results regarding their potential interactions with the Vc9Vd2 TCR [12,13,14]. As HMB-PP is considered for cd T-cell-based cancer clinical trials, hoping to improve the performance of previous phosphoantigens [7,8], it is crucial to clarify its own molecular/cellular mechanisms of action, including its potential capacity to trigger bona fide Vc9Vd2 TCR signaling. We have studied the most potent natural phosphoantigen yet identified, (E)-4-hydroxy-3-methyl-but-2-enyl pyrophosphate (HMB-PP), produced by Eubacteria and Protozoa, and examined its cd T-cell activation and anti-tumor properties

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call