scholarly journals Lethal pulmonary infection with Francisella novicida causes depletion of αβ T cells from lungs

2009 ◽  
Vol 257 (1-2) ◽  
pp. 1-4 ◽  
Author(s):  
Jyotika Sharma ◽  
Qun Li ◽  
Bibhuti B. Mishra ◽  
Judy M. Teale
2001 ◽  
Vol 107 (2) ◽  
pp. 359-366 ◽  
Author(s):  
Amy L. Woodward ◽  
Jonathan M. Spergel ◽  
Harri Alenius ◽  
Emiko Mizoguchi ◽  
Atul K. Bhan ◽  
...  

1998 ◽  
Vol 63 (6) ◽  
pp. 707-714 ◽  
Author(s):  
Robert A. Collins ◽  
Dirk Werling ◽  
Sara E. Duggan ◽  
A. Patricia Bland ◽  
Keith R. Parsons ◽  
...  
Keyword(s):  
T Cells ◽  

2018 ◽  
Vol 37 (4) ◽  
pp. S221
Author(s):  
J. Hsu ◽  
A. Krishnan ◽  
S. Lee ◽  
J.M. Dodd-o ◽  
B. Kim ◽  
...  

2021 ◽  
Vol 9 (Suppl 1) ◽  
pp. A19.1-A19
Author(s):  
JGM Strijker ◽  
E Drent ◽  
JJF van der Hoek ◽  
R Pscheid ◽  
B Koopmans ◽  
...  

BackgroundCurrently ~50% of patients with the diagnosis of high-risk neuroblastoma will not survive due to relapsing or refractory disease. Recent innovations in immunotherapy for solid tumors are highly promising, but the low MHC-I expression of neuroblastoma represents a major challenge for T cell-mediated immunotherapy. Here, we propose a novel T cell-based immunotherapy approach for neuroblastoma, based on the use of TEG002, αβ-T cells engineered to express a defined γδ-T cell receptor, which are thought to recognize and kill target cells independent of MHC-I. In this pilot project we have tested the potential efficacy of TEG002 therapy as a novel treatment for neuroblastoma, with tumor organoids.Materials and MethodsEffector cells were created from healthy donor peripheral blood T cells. The TEG002 cells were engineered by transducing αβ-T cells with a defined Vγ9Vδ2-T cell receptor. Both the untransduced αβ-T cells and the endogenous Vγ9Vδ2-T cells from the same healthy donor were used as controls in all experiments. Activation and killing of TEG002 was tested in a co-culture setting with neuroblastoma organoids. Supernatant of the co-culture was collected at 24 hours for IFNγ ELISA to measure activation of TEG002. The dynamics of cytotoxicity were analyzed over time from 0 till 72 hours, using the live-cell imaging system IncuCyte from Sartorius®. Killing was quantified using a Caspase3/7 Green dye and the IncuCyte software. Transcriptional profiling of the neuroblastoma organoids was done by RNA sequencing and MHC-I expression of the neuroblastoma organoids was determined by flow cytometry.ResultsWe showed that 3 out of 6 neuroblastoma organoids could activate TEG002 as measured by IFNγ production. Transcriptional profiling of the neuroblastoma organoids showed that this effect correlates with an increased activity of processes involved in interferon signaling and extracellular matrix organization. Analysis of the dynamics of organoid killing by TEG002 over time confirmed that organoids which induced TEG002 activation were efficiently killed independently of their MHC-I expression. Of note, efficacy of TEG002 treatment was superior to donor-matched untransduced αβ-T cells or endogenous γδ-T cells.ConclusionsWe demonstrated that 50% of tested neuroblastoma organoids can effectively activate TEG002 and that killing of the organoids is independent of MHC-I expression. Hence, this pilot study identified TEG002 as a promising novel cellular product for immunotherapy for a subset of neuroblastoma tumors, warranting further investigations into its clinical application.Disclosure InformationJ.G.M. Strijker: None. E. Drent: A. Employment (full or part-time); Significant; Gadeta BV. J.J.F. van der Hoek: None. R. Pscheid: A. Employment (full or part-time); Significant; Gadeta BV. B. Koopmans: None. K. Ober: None. S.R. van Hooff: None. W.M. Kholosy: None. C. Coomans: A. Employment (full or part-time); Significant; Gadeta BV. A. Bisso: A. Employment (full or part-time); Significant; Gadeta BV. M. van Loenen: A. Employment (full or part-time); Significant; Gadeta BV. J.J. Molenaar: None. J. Wienke: None.


Author(s):  
Christian R Schultze-Florey ◽  
Leonie Kuhlmann ◽  
Solaiman Raha ◽  
Joana Barros-Martins ◽  
Ivan Odak ◽  
...  

Donor lymphocyte infusion (DLI) is a standard of care for relapse of AML after allogeneic hematopoietic stem cell transplantation (aHSCT). Currently it is poorly understood how and when CD8+ αβ T cells exert graft-versus-leukemia (GvL) activity after DLI. Also, there is no reliable biomarker to monitor GvL activity of the infused CD8+ T cells. Therefore, we analyzed the dynamics of CD8+ αβ T cell clones in DLI-patients. In this prospective clinical study of 29 patients, we performed deep T cell receptor β (TRB) sequencing of sorted CD8+ αβ T cells to track patients' repertoire changes in response to DLI. Upon first occurrence of GvL, longitudinal analyses revealed a preferential expansion of distinct CD8+ TRB clones (n=14). This did not occur in samples of patients without signs of GvL (n=11). Importantly, early repertoire changes 15 days after DLI predicted durable remission for the 36 months study follow-up. Furthermore, absence of clonal outgrowth of the CD8+ TRB repertoire after DLI was an early biomarker that predicted relapse at a median time of 11.2 months ahead of actual diagnosis. Additionally, unbiased sample analysis regardless of the clinical outcome revealed that patients with decreasing CD8+ TRB diversity at day 15 after DLI (n=13) had a lower relapse incidence (P=0.0040) compared to patients without clonal expansion (n=6). In conclusion, CD8+ TRB analysis may provide a reliable tool for predicting the efficacy of DLI and holds the potential to identify patients at risk for progression and relapse after DLI.


2021 ◽  
Vol 11 (9) ◽  
pp. 923
Author(s):  
Josephine G. M. Strijker ◽  
Ronja Pscheid ◽  
Esther Drent ◽  
Jessica J. F. van der Hoek ◽  
Bianca Koopmans ◽  
...  

Currently ~50% of patients with a diagnosis of high-risk neuroblastoma will not survive due to relapsing or refractory disease. Recent innovations in immunotherapy for solid tumors are highly promising, but the low MHC-I expression of neuroblastoma represents a major challenge for T cell-mediated immunotherapy. Here, we propose a novel T cell-based immunotherapy approach for neuroblastoma, based on the use of TEG002, αβ-T cells engineered to express a defined γδ-T cell receptor, which can recognize and kill target cells independent of MHC-I. In a co-culture killing assay, we showed that 3 out of 6 neuroblastoma organoids could activate TEG002 as measured by IFNγ production. Transcriptional profiling showed this effect correlates with an increased activity of processes involved in interferon signaling and extracellular matrix organization. Analysis of the dynamics of organoid killing by TEG002 over time confirmed that organoids which induced TEG002 activation were efficiently killed independent of their MHC-I expression. Of note, efficacy of TEG002 treatment was superior to donor-matched untransduced αβ-T cells or endogenous γδ-T cells. Our data suggest that TEG002 may be a promising novel treatment option for a subset of neuroblastoma patients.


Immunology ◽  
1996 ◽  
Vol 88 (1) ◽  
pp. 82-89 ◽  
Author(s):  
R. ANZAI ◽  
S. SEKI ◽  
K. OGASAWARA ◽  
W. HASHIMOTO ◽  
K. SUGIURA ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document