Abstract

The adoptive transfer of ex vivo-expanded T cells is a promising approach to treat several malignancies. Several lines of evidence support that the infusion of T cells with early memory features, capable of expanding and persisting after transfer, are associated with better outcomes. We report herein that exposure to exogenous TGFβ during human T-cell stimulation ex vivo leads to the accumulation of early/central memory (Tcm) cells. Exposure to TGFβ suppressed the expression of BLIMP-1, a key orchestrator of effector T-cell differentiation, and led to the upregulation of the memory-associated transcription factor ID3. Accordingly, this was associated with an early memory transcriptional signature in both CD4+ and CD8+ T-cell subsets. The T cells stimulated in the presence of TGFβ expanded normally, and displayed polyfunctional features and no suppressive activity. The adoptive transfer of ex vivo-stimulated T cells into immunodeficient mice confirmed that TGFβ-conditioned cells had an enhanced capacity to persist and mediate xenogeneic graft-versus-host disease, as predicted by their early T-cell memory phenotype. Chimeric antigen receptor-expressing T cells generated in the presence of exogenous TGFβ were cytotoxic and more effective at controlling tumor growth in immunodeficient animals. This work unveils a new role for TGFβ in memory T-cell differentiation and indicates that TGFβ signaling may be harnessed to program Tcm differentiation in the context of ex vivo T-cell stimulation for adoptive immunotherapy in humans.

Highlights

  • The integration of stimulatory signals during T-cell activation programs the differentiation of effector and memory T cells

  • According to the progressive differentiation model, T cells differentiate depending on the nature and strength of activation signals following a one-way linear path from na€ve to early memory, effector memory (Tem), and terminally differentiated effector T cells

  • the accumulation of early/central memory (Tcm) accumulation Total T cells from healthy donor volunteers were stimulated with plate-bound anti-CD3 and soluble anti-CD28 in the presence or absence of exogenous TGFb

Read more

Summary

Introduction

The integration of stimulatory signals during T-cell activation programs the differentiation of effector and memory T cells. Primarily known for its immunoregulatory and antiproliferative properties, TGFb orchestrates both regulatory T-cell (Treg) and inflammatory subset differentiation depending on the presence of additional signals (11--14). This pleiotropy is further exemplified by the contrasting prosurvival effects of TGFb on na€ve and memory T cells, and the proapoptotic and functional inhibitory effects on differentiated Teff (15--17). Memory T-cell Differentiation by TGFb transfer of activated T cells in immunodeficient mice revealed that TGFb exposure in culture conferred an enhanced capacity to expand, persist, and mediate xenogeneic graft-versus-host disease (GVHD), as previously reported for cells with Tscm features [3]. The TGFb pathway can be used to program early memory differentiation in human T cells and has, immediate relevance for the field of adoptive immunotherapy

Materials and Methods
Results
Discussion
Disclosure of Potential Conflicts of Interest
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