Abstract

BackgroundVirus-specific T-cells (VSTs) proliferate exponentially after adoptive transfer into hematopoietic stem cell transplant (HSCT) recipients, eliminate virus infections, then persist and provide long-term protection from viral disease. If VSTs behaved similarly when modified with tumor-specific chimeric antigen receptors (CARs), they should have potent anti-tumor activity. This theory was evaluated by Cruz et al. in a previous clinical trial with CD19.CAR-modified VSTs, but there was little apparent expansion of these cells in patients. In that study, VSTs were gene-modified on day 19 of culture and we hypothesized that by this time, sufficient T-cell differentiation may have occurred to limit the subsequent proliferative capacity of the transduced T-cells. To facilitate the clinical testing of this hypothesis in a project supported by the NHLBI-PACT mechanism, we developed and optimized a good manufacturing practices (GMP) compliant method for the early transduction of VSTs directed to Epstein-Barr virus (EBV), Adenovirus (AdV) and cytomegalovirus (CMV) using a CAR directed to the tumor-associated antigen disialoganglioside (GD2).ResultsAd-CMVpp65-transduced EBV-LCLs effectively stimulated VSTs directed to all three viruses (triVSTs). Transduction efficiency on day three was increased in the presence of cytokines and high-speed centrifugation of retroviral supernatant onto retronectin-coated plates, so that under optimal conditions up to 88% of tetramer-positive VSTs expressed the GD2.CAR. The average transduction efficiency of early-and late transduced VSTs was 55 ± 4% and 22 ± 5% respectively, and early-transduced VSTs maintained higher frequencies of T cells with central memory or intermediate memory phenotypes. Early-transduced VSTs also had higher proliferative capacity and produced higher levels of TH1 cytokines IL-2, TNF-α, IFN-γ, MIP-1α, MIP-1β and other cytokines in vitro.ConclusionsWe developed a rapid and GMP compliant method for the early transduction of multivirus-specific T-cells that allowed stable expression of high levels of a tumor directed CAR. Since a proportion of early-transduced CAR-VSTs had a central memory phenotype, they should expand and persist in vivo, simultaneously protecting against infection and targeting residual malignancy. This manufacturing strategy is currently under clinical investigation in patients receiving allogeneic HSCT for relapsed neuroblastoma and B-cell malignancies (NCT01460901 using a GD2.CAR and NCT00840853 using a CD19.CAR).Electronic supplementary materialThe online version of this article (doi:10.1186/s40425-015-0049-1) contains supplementary material, which is available to authorized users.

Highlights

  • Virus-specific T-cells (VSTs) proliferate exponentially after adoptive transfer into hematopoietic stem cell transplant (HSCT) recipients, eliminate virus infections, persist and provide long-term protection from viral disease

  • In a previous clinical trial we tested the hypothesis that extratumoral stimulation by an endogenous virus would ensure chimeric antigen receptor (CAR)-T-cell expansion in vivo in children with relapsed neuroblastoma infused with autologous Epstein-Barr virus (EBV)-specific T-cells (EBVSTs) genetically modified to express a CAR specific for GD2, a disialoganglioside that is highly expressed by this tumor [1,2]

  • EBV-specific T-cells (EBVSTs) can be transduced effectively on day 3 of culture To enhance the transduction efficiency of VSTs on day 2 or day 3 we attempted to increase the potency of the first Peripheral blood mononuclear cells (PBMC) stimulation without loss of virus specificity

Read more

Summary

Introduction

Virus-specific T-cells (VSTs) proliferate exponentially after adoptive transfer into hematopoietic stem cell transplant (HSCT) recipients, eliminate virus infections, persist and provide long-term protection from viral disease. We expected that endogenous EBV would provide in vivo stimulation of GD2.CAR-modified EBVSTs, increasing their expansion and anti-tumor function relative to -transduced CD3-activated T-cells (GD2.CAR-ATCs). In this original study, each T-cell component expressed a GD2.CAR that differed only in a few non-coding nucleotides that allowed us to compare the fate of infused GD2.CARATCs and GD2.CAR-EBVSTs in each patient treated. Transduced EBVSTs were detected at higher levels than transduced ATCs in the six weeks following infection, they did not apparently expand in numbers, at least as measured in the circulation, and tumor responses were associated with the long-term persistence of either population, albeit at low levels It was unclear which population was responsible for the clinical responses

Methods
Results
Discussion
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