Faculty Opinions recommendation of Genomic definition of multiple ex vivo regulatory T cell subphenotypes.

Author(s):  
Laurence Turka ◽  
Scott Lieberman
2010 ◽  
Vol 107 (13) ◽  
pp. 5919-5924 ◽  
Author(s):  
M. Feuerer ◽  
J. A. Hill ◽  
K. Kretschmer ◽  
H. von Boehmer ◽  
D. Mathis ◽  
...  

2020 ◽  
Author(s):  
JL Reading ◽  
VD Roobrouck ◽  
CM Hull ◽  
PD Becker ◽  
J Beyens ◽  
...  

AbstractRecent clinical experience has demonstrated that adoptive regulatory T cell therapy is a safe and feasible strategy to suppress immunopathology via induction of host tolerance to allo- and autoantigens. However, clinical trials continue to be compromised due to an inability to manufacture a sufficient Treg cell dose. Multipotent adult progenitor cells (MAPCⓇ) promote regulatory T cell differentiation in vitro, suggesting they may be repurposed to enhance ex vivo expansion of Tregs for adoptive cellular therapy. Here, we use a GMP compatible Treg expansion platform to demonstrate that MAPC cell-co-cultured Tregs (MulTreg) exhibit a log-fold increase in yield across two independent cohorts, reducing time to target dose by an average of 30%. Enhanced expansion is linked with a distinct Treg cell-intrinsic transcriptional program, characterized by diminished levels of core exhaustion (BATF, ID2, PRDM1, LAYN, DUSP1), and quiescence (TOB1, TSC22D3) related genes, coupled to elevated expression of cell-cycle and proliferation loci (MKI67, CDK1, AURKA, AURKB). In addition, MulTreg display a unique gut homing (CCR7lo β7hi) phenotype and importantly, are more readily expanded from patients with autoimmune disease compared to matched Treg lines, suggesting clinical utility in gut and/or Th1-driven pathology associated with autoimmunity or transplantation. Relative to expanded Tregs, MulTreg retain equivalent and robust purity, FoxP3 TSDR demethylation, nominal effector cytokine production and potent suppression of Th1-driven antigen specific and polyclonal responses in vitro and xeno graft vs host disease (xGvHD) in vivo. These data support the use of MAPC cell co-culture in adoptive Treg therapy platforms as a means to rescue expansion failure and reduce the time required to manufacture a stable, potently suppressive product.


2020 ◽  
Vol 4 (8) ◽  
pp. 1594-1605 ◽  
Author(s):  
Scott N. Furlan ◽  
Karnail Singh ◽  
Christina Lopez ◽  
Victor Tkachev ◽  
Daniel Joel Hunt ◽  
...  

Abstract As regulatory T cell (Treg) adoptive therapy continues to develop clinically, there is a need to determine which immunomodulatory agents pair most compatibly with Tregs to enable persistence and stabilize suppressor function. Prior work has shown that mechanistic target of rapamycin inhibition can increase the stability of thymic Tregs. In this study, we investigated the transcriptomic signatures of ex vivo–expanded Tregs after adoptive transfer in the setting of clinically relevant immunosuppression using a nonhuman primate (NHP) model as a prelude to future transplant studies. Here, we found that adding interleukin-2 (IL-2) to rapamycin in vivo supported a logarithmic increase in the half-life of adoptively transferred carboxyfluorescein diacetate succinimidyl ester–labeled, autologous NHP Tregs, effectively doubling the number of cells in the peripheral blood Treg compartment compared with Treg infusion when rapamycin was given alone. Using single-cell transcriptomics, we found that transferred ex vivo–expanded Tregs initially exhibit a gene expression signature consistent with an activated state. Moreover, those cells with the highest levels of activation also expressed genes associated with p53-mediated apoptosis. In contrast, transferred Tregs interrogated at day +20 posttransfer demonstrated a gene signature more similar to published profiles of resting Tregs. Together, these preclinical data further support combining IL-2 and rapamycin in vivo as adjunctive therapy for ex vivo–expanded adoptively transferred Tregs and suggest that the activation status of ex vivo–expanded Tregs is critical to their persistence.


2017 ◽  
Vol 101 (10) ◽  
pp. e326-e327 ◽  
Author(s):  
Kraig V. Abrams ◽  
Billanna Hwang ◽  
Richard A. Nash ◽  
George E. Georges ◽  
Wayne Lamm ◽  
...  
Keyword(s):  
T Cell ◽  

PLoS ONE ◽  
2018 ◽  
Vol 13 (2) ◽  
pp. e0193382 ◽  
Author(s):  
Ricardo da Silva Antunes ◽  
Sinu Paul ◽  
John Sidney ◽  
Daniela Weiskopf ◽  
Jennifer M. Dan ◽  
...  

PLoS ONE ◽  
2017 ◽  
Vol 12 (1) ◽  
pp. e0169086 ◽  
Author(s):  
Ricardo da Silva Antunes ◽  
Sinu Paul ◽  
John Sidney ◽  
Daniela Weiskopf ◽  
Jennifer M. Dan ◽  
...  

Blood ◽  
2012 ◽  
Vol 119 (8) ◽  
pp. e57-e66 ◽  
Author(s):  
James B. Canavan ◽  
Behdad Afzali ◽  
Cristiano Scottà ◽  
Henrieta Fazekasova ◽  
Francis C. Edozie ◽  
...  

Abstract Regulatory T cells (CD4+CD25hiCD127loFOXP3+ T cells [Tregs]) are a population of lymphocytes involved in the maintenance of self-tolerance. Abnormalities in function or number of Tregs are a feature of autoimmune diseases in humans. The ability to expand functional Tregs ex vivo makes them ideal candidates for autologous cell therapy to treat human autoimmune diseases and to induce tolerance to transplants. Current tests of Treg function typically take up to 120 hours, a kinetic disadvantage as clinical trials of Tregs will be critically dependent on the availability of rapid diagnostic tests before infusion into humans. Here we evaluate a 7-hour flow cytometric assay for assessing Treg function, using suppression of the activation markers CD69 and CD154 on responder T cells (CD4+CD25− [Tresp]), compared with traditional assays involving inhibition of CFSE dilution and cytokine production. In both freshly isolated and ex vivo expanded Tregs, we describe excellent correlation with gold standard suppressor cell assays. We propose that the kinetic advantage of the new assay may place it as the preferred rapid diagnostic test for the evaluation of Treg function in forthcoming clinical trials of cell therapy, enabling the translation of the large body of preclinical data into potentially useful treatments for human diseases.


2013 ◽  
Vol 2013 ◽  
pp. 1-17 ◽  
Author(s):  
Leslie P. Cousens ◽  
Yan Su ◽  
Elizabeth McClaine ◽  
Xin Li ◽  
Frances Terry ◽  
...  

HLA class II-restricted regulatory T cell (Treg) epitopes in IgG (also called “Tregitopes”) have been reported to suppress immune responses to coadministered antigens by stimulating the expansion of natural Tregs (nTregs). Here we evaluate their impact on human immune responses to islet cell antigensex vivoand on the modulation of type 1 diabetes (T1D) in a murine modelin vivo. Co-administration of Tregitopes and T1D antigens delayed development of hyperglycemia and reduced the incidence of diabetes in NOD mice. Suppression of diabetes could be observed even following onset of disease. To measure the impact of Tregitope treatment on T cell responses, we evaluated the effect of Tregitope treatment in DO11.10 mice. Upregulation of FoxP3 in KJ1-26-stained OVA-specific CD4+T cells was observed following treatment of DO11.10 mice with Tregitopes, along with reductions in anti-OVA Ig and T effector responses. Inex vivostudies of human T cells, peripheral blood mononuclear cells’ (PBMC) responses to GAD65 epitopes in the presence and absence of Tregitope were variable. Suppression of immune responses to GAD65 epitopesex vivoby Tregitope appeared to be more effective in assays using PBMC from a newly diagnosed diabetic subject than for other more established diabetic subjects, and correlation of the degree of suppression with predicted HLA restriction of the Tregitopes was confirmed. Implementation of these defined regulatory T cell epitopes for therapy of T1D and other autoimmune diseases may lead to a paradigm shift in disease management.


2016 ◽  
Vol 44 (2) ◽  
pp. 342-348 ◽  
Author(s):  
Dominic Boardman ◽  
John Maher ◽  
Robert Lechler ◽  
Lesley Smyth ◽  
Giovanna Lombardi

Adoptive regulatory T-cell (Treg) therapy using autologous Tregs expanded ex vivo is a promising therapeutic approach which is currently being investigated clinically as a means of treating various autoimmune diseases and transplant rejection. Despite this, early results have highlighted the need for potent Tregs to yield a substantial clinical advantage. One way to achieve this is to create antigen-specific Tregs which have been shown in pre-clinical animal models to have an increased potency at suppressing undesired immune responses, compared to polyclonal Tregs. This mini review outlines where Treg therapy currently stands and discusses the approaches which may be taken to generate antigen-specific Tregs, including the potential use of chimeric antigen receptors (CARs), for future clinical trials.


Sign in / Sign up

Export Citation Format

Share Document