13 research outputs found

    Transfer and loss of allergenā€specific responses via stem cell transplantation: A prospective observational study

    Full text link
    Background Currently, no estimates can be made on the impact of hematopoietic stem cell transplantation on allergy transfer or cure of the disease. By using componentā€resolved diagnosis, we prospectively investigated 50 donorā€recipient pairs undergoing allogeneic stem cell transplantation. This allowed calculating the rate of transfer or maintenance of allergenā€specific responses in the context of stem cell transplantation. Methods Allergenā€specific IgE and IgG to 156 allergens was measured pretransplantation in 50 donors and recipients and at 6, 12 and 24 months in recipients postā€transplantation by allergen microarray. Based on a mixed effects model, we determined risks of transfer of allergenā€specific IgE or IgG responses 24 months postā€transplantation. Results After undergoing stem cell transplantation, 94% of allergenā€specific IgE responses were lost. Two years postā€transplantation, recipients' allergenā€specific IgE was significantly linked to the pretransplantation donor or recipient status. The estimated risk to transfer and maintain individual IgE responses to allergens by stem cell transplantation was 1.7% and 2.3%, respectively. Allergenā€specific IgG, which served as a surrogate marker of maintaining protective IgG responses, was highly associated with the donor's (31.6%) or the recipient's (28%) pretransplantation response. Conclusion Hematopoietic stem cell transplantation profoundly reduces allergenā€specific IgE responses but also comes with a considerable risk to transfer allergenā€specific immune responses. These findings facilitate clinical decisionā€making regarding allergic diseases in the context of hematopoietic stem cell transplantation. In addition, it provides prospective data to estimate the risk of transmitting allergenā€specific responses via hematopoietic stem cell transplantation

    Human Adenovirus-Specific Ī³/Ī“ and CD8<sup>+</sup> T Cells Generated by T-Cell Receptor Transfection to Treat Adenovirus Infection after Allogeneic Stem Cell Transplantation

    No full text
    <div><p>Human adenovirus infection is life threatening after allogeneic haematopoietic stem cell transplantation (HSCT). Immunotherapy with donor-derived adenovirus-specific T cells is promising; however, 20% of all donors lack adenovirus-specific T cells. To overcome this, we transfected Ī±/Ī² T cells with mRNA encoding a T-cell receptor (TCR) specific for the HLA-A*0101-restricted peptide LTDLGQNLLY from the adenovirus hexon protein. Furthermore, since allo-reactive endogenous TCR of donor T lymphocytes would induce graft-versus-host disease (GvHD) in a mismatched patient, we transferred the TCR into Ī³/Ī“ T cells, which are not allo-reactive. TCR-transfected Ī³/Ī“ T cells secreted low quantities of cytokines after antigen-specific stimulation, which were increased dramatically after co-transfection of CD8Ī±-encoding mRNA. In direct comparison with TCR-transfected Ī±/Ī² T cells, TCR-CD8Ī±-co-transfected Ī³/Ī“ T cells produced more tumor necrosis factor (TNF), and lysed peptide-loaded target cells as efficiently. Most importantly, TCR-transfected Ī±/Ī² T cells and TCR-CD8Ī±-co-transfected Ī³/Ī“ T cells efficiently lysed adenovirus-infected target cells. We show here, for the first time, that not only Ī±/Ī² T cells but also Ī³/Ī“ T cells can be equipped with an adenovirus specificity by TCR-RNA electroporation. Thus, our strategy offers a new means for the immunotherapy of adenovirus infection after allogeneic HSCT.</p></div

    Direct comparison of cytokine production by HAdV/A1-TCR-transfected CD8<sup>+</sup> T cells and HAdV/A1-TCR-transfected Ī³/Ī“ T cells.

    No full text
    <p>CD8<sup>+</sup> T cells and Ī³/Ī“ T cells of the same donors were either mock electroporated, electroporated with HAdV/A1-TCR-RNA alone, or electroporated with HAdV/A1-TCR-RNA and CD8Ī±-RNA. These cells were stimulated with colo829 cells (<b>A</b>) or CCL cells (<b>B</b>), which were either loaded with a control peptide (contr. pept.) or the adenovirus peptide (Adeno pept.). Cytokine concentrations (IL-2, TNF, and IFNĪ³) in the supernatant after overnight co-incubation are depicted. Data of 3 individual experiments are shown. Bars indicate mean values. Raw data are summarized in <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0109944#pone.0109944.s001" target="_blank">Table S5A and B in File S1</a>.</p

    Cytolytic capacity of HAdV/A1-TCR-transfected CD8<sup>+</sup> T cells and Ī³/Ī“ T cells against adenovirus-infected target cells.

    No full text
    <p>CD8<sup>+</sup> T cells (<b>A</b> and <b>C</b>) and Ī³/Ī“ T cells (<b>B</b> and <b>D</b>) of the same donors were either electroporated with mRNA encoding the MAGE-1/A1-specific TCR (open symbols) or electroporated with mRNA encoding the HAdV/A1-specific TCR (closed symbols) either in combination with CD8Ī± mRNA (Ī³/Ī“ T cells) or not (CD8<sup>+</sup> T cells). These cells were used as effector cells in standard 4ā€“6 h cytotoxicity assays. Autologous (<b>A</b> and <b>B</b>) and allogenic (<b>C</b> and <b>D</b>) DC either untreated (non-loaded), loaded with the adenovirus peptide (Adeno pept.), or infected with adenovirus (adenovirus) were used as target cells and the percentage of lysed cells was calculated. The target to effector cell ratios were 1āˆ¶60, 1āˆ¶20, 1āˆ¶6, and 1āˆ¶2. Average values of 6 (<b>A</b> and <b>B</b>) or 4 (<b>C</b> and <b>D</b>) individual experiments (each performed in triplicates) +/āˆ’ SEM are shown. P-values were calculated by the Mann-Whitney U test. For statistics, conditions were compared to the non-loaded DC condition (* and **) and to T cells transfected with the M1/A1 control TCR (<sup>ā€”</sup> and <sup>ā€ </sup>). (** or <sup>ā€”</sup> pā‰¤0.01; *or <sup>ā€ </sup> pā‰¤0.05). Raw data are summarized in <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0109944#pone.0109944.s001" target="_blank">Table S7A to D in File S1</a>.</p

    Antigen-specific cytokine production by HAdV/A1-TCR-transfected CD8<sup>+</sup> and Ī³/Ī“ T cells in response to peptide-loaded target cells.

    No full text
    <p>CD8<sup>+</sup> T cells and Ī³/Ī“ T cells were either electroporated with M1/A1-TCR RNA (+CD8Ī± RNA in case of Ī³/Ī“ T cells) or with HAdV/A1-TCR-RNA (+CD8Ī± RNA in case of Ī³/Ī“ T cells) and were cryoconserved 4 h after electroporation. After thawing, these cells were stimulated with CCL cells, which were either left unloaded (w/o pept.), or were loaded with the adenovirus peptide (w/pept.). Intracellular cytokine stainings for IL-2 (white bars), TNF (grey bars), and IFNĪ³ (black bars) were performed and analyzed by flow-cytometry. The percentages of cytokine containing cells are depicted. Average values of 4 (CD8<sup>+</sup> T cells) and 3 (Ī³/Ī“ T cells) individual experiments Ā± SEM are shown. Raw data are summarized in <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0109944#pone.0109944.s001" target="_blank">Table S6 in File S1</a>.</p

    The newly cloned HAdV/HLA-A1-specific TCR is functional in Jurkat T cells.

    No full text
    <p>Parental CD4<sup>+</sup> and transgenic CD8<sup>+</sup> Jurkat T cells were co-electroporated with RNA encoding the HAdV/A1-specific TCR or MAGE-A3/A1 (M3/A1)-specific TCR and an NFAT-inducible luciferase reporter plasmid. These Jurkat T cells were stimulated with DC either non-loaded (w/o pept.) or loaded with the adenovirus peptide (Adeno pept.) or the MAGE-A3 peptide (MAGE-A3 pept.) (as indicated). The luciferase activity was measured, and the specific activation of the Jurkat T cells was calculated as fold induction by dividing the luciferase activity induced by peptide-loaded DC by that of similarly electroporated Jurkat T cells stimulated with non-loaded DC. Data of 4 (CD4<sup>+</sup> Jurkat T cells) and 5 (CD8<sup>+</sup> Jurkat T cells) individual experiments are shown. Bars indicate mean values. P-values were calculated by the Mann-Whitney U test. ns ā€Š=ā€Š not significant; ** pā‰¤0.01; * pā‰¤0.05. Raw data are summarized in <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0109944#pone.0109944.s001" target="_blank">Table S1 in File S1</a>.</p

    Antigen-specific cytokine production by HAdV/A1-TCR-transfected CD8<sup>+</sup> T cells in response to peptide-loaded and adenovirus-infected targets.

    No full text
    <p>CD8<sup>+</sup> T cells were either mock electroporated or electroporated with HAdV/A1-TCR-RNA and were stimulated with DC, which were either left unloaded (<b>A</b>: w/o pept., <b>B</b>: w/o virus), or were loaded with the adenovirus peptide (Adeno pept.) (<b>A</b>), or were infected with adenovirus (w/virus) (<b>B</b>). Cytokine concentrations (IL-2, TNF, and IFNĪ³) in the supernatant after over-night co-incubation are depicted. Data of 5 (<b>A</b>) and 7 (<b>B</b>) individual experiments are shown. Bars indicate mean values. P-values were calculated by the Mann-Whitney U test. ns ā€Š=ā€Š not significant, *** pā‰¤0.001; ** pā‰¤0.01; * pā‰¤0.05. Raw data are summarized in <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0109944#pone.0109944.s001" target="_blank">Table S2A and B in File S1</a>.</p

    Surface marker and TCR expression on HAdV/A1-TCR-transfected CD8<sup>+</sup> and Ī³/Ī“ T cells.

    No full text
    <p>CD8<sup>+</sup> T cells (A and B; upper panels) and Ī³/Ī“ T cells (A and B; lower panels) were either electroporated with M1/A1-TCR RNA (+CD8Ī± RNA in case of Ī³/Ī“ T cells) or with HAdV/A1-TCR-RNA (+CD8Ī± RNA in case of Ī³/Ī“ T cells) and were cryoconserved 4 h after electroporation. Surface marker and TCR expression on thawed T cells was determined by staining with anti-CD4, anti-CD8, anti-CD16, anti-CD19, anti-CD14, and anti-pan Ī³/Ī“ TCR antibodies, and the expression of the HAdV/A1-specific TCR was determined by staining with HAdV/A1 streptamer. Expression levels were analyzed by flow-cytometry and shown as % positive cells; isotype control stainings were subtracted (<b>A</b>; average values of 4 experiments Ā± SEM) or as histograms (<b>B</b>; grey histogram: T cells electroporated with M1/A1-TCR RNA, black line: T cells electroporated with HAdV/A1-TCR-RNA). Raw data are summarized in <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0109944#pone.0109944.s001" target="_blank">Table S4A and B in File S1</a>.</p

    Antigen-specific cytokine production by HAdV/A1-TCR-transfected Ī³/Ī“ T cells in response to peptide-loaded target cells.

    No full text
    <p>Ī³/Ī“ T cells were either mock electroporated, electroporated with HAdV/A1-TCR-RNA alone, or electroporated with HAdV/A1-TCR-RNA and CD8Ī±-RNA. These cells were stimulated with DC (<b>A</b>) or colo829 cells (<b>B</b>), which were either left unloaded (w/o pept.), or were loaded with the adenovirus peptide (Adeno pept.). Cytokine concentrations (IL-2, TNF, and IFNĪ³) in the supernatant after overnight co-incubation are depicted. Data of 3 individual experiments are shown. Bars indicate mean values. Raw data are summarized in <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0109944#pone.0109944.s001" target="_blank">Table S3A and B in in File S1</a>.</p

    Risk assessment of relapse by lineage-specific monitoring of chimerism in children undergoing allogeneic stem cell transplantation for acute lymphoblastic leukemia

    Get PDF
    Allogeneic hematopoietic stem cell transplantation is required as rescue therapy in about 20% of pediatric patients with acute lymphoblastic leukemia. However, the relapse rates are considerable, and relapse confers a poor outcome. Early assessment of the risk of relapse is therefore of paramount importance for the development of appropriate measures. We used the EuroChimerism approach to investigate the potential impact of lineage-specific chimerism testing for relapse-risk analysis in 162 pediatric patients with acute lymphoblastic leukemia after allogeneic stem cell transplantation in a multicenter study based on standardized transplantation protocols. Within a median observation time of 4.5 years, relapses have occurred in 41/162 patients at a median of 0.6 years after transplantation (range, 0.13-5.7 years). Prospective screening at defined consecutive time points revealed that reappearance of recipient-derived cells within the CD34(+) and CD8(+) cell subsets display the most significant association with the occurrence of relapses with hazard ratios of 5.2 (P=0.003) and 2.8 (P=0.008), respectively. The appearance of recipient cells after a period of pure donor chimerism in the CD34(+) and CD8(+) leukocyte subsets revealed dynamics indicative of a significantly elevated risk of relapse or imminent disease recurrence. Assessment of chimerism within these lineages can therefore provide complementary information for further diagnostic and, potentially, therapeutic purposes aiming at the prevention of overt relapse. This study was registered at clinical. TRIALS: gov with the number NC01423747
    corecore