Influenza virus-specific TCR-transduced T cells as a model for adoptive immunotherapy

Standard

Influenza virus-specific TCR-transduced T cells as a model for adoptive immunotherapy. / Berdien, Belinda; Reinhard, Henrike; Meyer, Sabrina; Spöck, Stefanie; Kröger, Nicolaus-Martin; Atanackovic, Djordje; Fehse, Boris.

In: HUM VACC IMMUNOTHER, Vol. 9, No. 6, 01.06.2013, p. 1205-16.

Research output: SCORING: Contribution to journalSCORING: Journal articleResearchpeer-review

Harvard

APA

Vancouver

Bibtex

@article{a1367c6bcfd04b2088e52c75d3b9ecb3,
title = "Influenza virus-specific TCR-transduced T cells as a model for adoptive immunotherapy",
abstract = "Adoptive transfer of T lymphocytes equipped with tumor-antigen specific T-cell receptors (TCRs) represents a promising strategy in cancer immunotherapy, but the approach remains technically demanding. Using influenza virus (Flu)-specific T-cell responses as a model system we compared different methods for the generation of T-cell clones and isolation of antigen-specific TCRs. Altogether, we generated 12 CD8(+) T-cell clones reacting to the Flu matrix protein (Flu-M) and 6 CD4(+) T-cell clones reacting to the Flu nucleoprotein (Flu-NP) from 4 healthy donors. IFN-γ-secretion-based enrichment of antigen-specific cells, optionally combined with tetramer staining, was the most efficient way for generating T-cell clones. In contrast, the commonly used limiting dilution approach was least efficient. TCR genes were isolated from T-cell clones and cloned into both a previously used gammaretroviral LTR-vector, MP91 and the novel lentiviral self-inactivating vector LeGO-MP that contains MP91-derived promotor and regulatory elements. To directly compare their functional efficiencies, we in parallel transduced T-cell lines and primary T cells with the two vectors encoding identical TCRs. Transduction efficiencies were approximately twice higher with the gammaretroviral vector. Secretion of high amounts of IFN-γ, IL-2 and TNF-α by transduced cells after exposure to the respective influenza target epitope proved efficient specificity transfer of the isolated TCRs to primary T-cells for both vectors, at the same time indicating superior functionality of MP91-transduced cells. In conclusion, we have developed optimized strategies to obtain and transfer antigen-specific TCRs as well as designed a novel lentiviral vector for TCR-gene transfer. Our data may help to improve adoptive T-cell therapies.",
keywords = "CD4-Positive T-Lymphocytes, CD8-Positive T-Lymphocytes, Gene Expression, Genetic Vectors, Healthy Volunteers, Humans, Immunotherapy, Adoptive, Interferon-gamma, Interleukin-2, Lentivirus, RNA-Binding Proteins, Receptors, Antigen, T-Cell, Transduction, Genetic, Tumor Necrosis Factor-alpha, Viral Core Proteins, Viral Matrix Proteins",
author = "Belinda Berdien and Henrike Reinhard and Sabrina Meyer and Stefanie Sp{\"o}ck and Nicolaus-Martin Kr{\"o}ger and Djordje Atanackovic and Boris Fehse",
year = "2013",
month = jun,
day = "1",
doi = "10.4161/hv.24051",
language = "English",
volume = "9",
pages = "1205--16",
journal = "HUM VACC IMMUNOTHER",
issn = "2164-5515",
publisher = "LANDES BIOSCIENCE",
number = "6",

}

RIS

TY - JOUR

T1 - Influenza virus-specific TCR-transduced T cells as a model for adoptive immunotherapy

AU - Berdien, Belinda

AU - Reinhard, Henrike

AU - Meyer, Sabrina

AU - Spöck, Stefanie

AU - Kröger, Nicolaus-Martin

AU - Atanackovic, Djordje

AU - Fehse, Boris

PY - 2013/6/1

Y1 - 2013/6/1

N2 - Adoptive transfer of T lymphocytes equipped with tumor-antigen specific T-cell receptors (TCRs) represents a promising strategy in cancer immunotherapy, but the approach remains technically demanding. Using influenza virus (Flu)-specific T-cell responses as a model system we compared different methods for the generation of T-cell clones and isolation of antigen-specific TCRs. Altogether, we generated 12 CD8(+) T-cell clones reacting to the Flu matrix protein (Flu-M) and 6 CD4(+) T-cell clones reacting to the Flu nucleoprotein (Flu-NP) from 4 healthy donors. IFN-γ-secretion-based enrichment of antigen-specific cells, optionally combined with tetramer staining, was the most efficient way for generating T-cell clones. In contrast, the commonly used limiting dilution approach was least efficient. TCR genes were isolated from T-cell clones and cloned into both a previously used gammaretroviral LTR-vector, MP91 and the novel lentiviral self-inactivating vector LeGO-MP that contains MP91-derived promotor and regulatory elements. To directly compare their functional efficiencies, we in parallel transduced T-cell lines and primary T cells with the two vectors encoding identical TCRs. Transduction efficiencies were approximately twice higher with the gammaretroviral vector. Secretion of high amounts of IFN-γ, IL-2 and TNF-α by transduced cells after exposure to the respective influenza target epitope proved efficient specificity transfer of the isolated TCRs to primary T-cells for both vectors, at the same time indicating superior functionality of MP91-transduced cells. In conclusion, we have developed optimized strategies to obtain and transfer antigen-specific TCRs as well as designed a novel lentiviral vector for TCR-gene transfer. Our data may help to improve adoptive T-cell therapies.

AB - Adoptive transfer of T lymphocytes equipped with tumor-antigen specific T-cell receptors (TCRs) represents a promising strategy in cancer immunotherapy, but the approach remains technically demanding. Using influenza virus (Flu)-specific T-cell responses as a model system we compared different methods for the generation of T-cell clones and isolation of antigen-specific TCRs. Altogether, we generated 12 CD8(+) T-cell clones reacting to the Flu matrix protein (Flu-M) and 6 CD4(+) T-cell clones reacting to the Flu nucleoprotein (Flu-NP) from 4 healthy donors. IFN-γ-secretion-based enrichment of antigen-specific cells, optionally combined with tetramer staining, was the most efficient way for generating T-cell clones. In contrast, the commonly used limiting dilution approach was least efficient. TCR genes were isolated from T-cell clones and cloned into both a previously used gammaretroviral LTR-vector, MP91 and the novel lentiviral self-inactivating vector LeGO-MP that contains MP91-derived promotor and regulatory elements. To directly compare their functional efficiencies, we in parallel transduced T-cell lines and primary T cells with the two vectors encoding identical TCRs. Transduction efficiencies were approximately twice higher with the gammaretroviral vector. Secretion of high amounts of IFN-γ, IL-2 and TNF-α by transduced cells after exposure to the respective influenza target epitope proved efficient specificity transfer of the isolated TCRs to primary T-cells for both vectors, at the same time indicating superior functionality of MP91-transduced cells. In conclusion, we have developed optimized strategies to obtain and transfer antigen-specific TCRs as well as designed a novel lentiviral vector for TCR-gene transfer. Our data may help to improve adoptive T-cell therapies.

KW - CD4-Positive T-Lymphocytes

KW - CD8-Positive T-Lymphocytes

KW - Gene Expression

KW - Genetic Vectors

KW - Healthy Volunteers

KW - Humans

KW - Immunotherapy, Adoptive

KW - Interferon-gamma

KW - Interleukin-2

KW - Lentivirus

KW - RNA-Binding Proteins

KW - Receptors, Antigen, T-Cell

KW - Transduction, Genetic

KW - Tumor Necrosis Factor-alpha

KW - Viral Core Proteins

KW - Viral Matrix Proteins

U2 - 10.4161/hv.24051

DO - 10.4161/hv.24051

M3 - SCORING: Journal article

C2 - 23428899

VL - 9

SP - 1205

EP - 1216

JO - HUM VACC IMMUNOTHER

JF - HUM VACC IMMUNOTHER

SN - 2164-5515

IS - 6

ER -