GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    In: Blood, American Society of Hematology, Vol. 106, No. 11 ( 2005-11-16), p. 5537-5537
    Abstract: TCR transfer to engineer tumor specific T cells may be an alternative strategy for adoptive immunotherapy. We have previously shown that TCR-transduced T cells are capable of recognizing targets both via their endogenous TCR and the introduced TCR. Since the introduced TCR is regulated by a different promotor, we investigated whether triggering and modulation of the introduced TCR occur in a physiological manner compared to the endogenous TCR. Because introduction of a TCRα-chain and TCRβ-chain will lead to formation of chimeric TCR-complexes with the endogenous TCRα- and β-chain, we used models in which both specificities of the TCRs were known and tetramers were available to distinguish each TCR. CMV PP65 specific T cells were transduced with a retroviral construct encoding the hematopoietic minor histocompatibility antigen HA-2 specific TCR (HA-2-TCR). TCR-transduced T cells were antigen specifically triggered using EBV-LCLs that presented endogenously processed PP65- or HA-2-antigen. At various time points after stimulation cell surface expression of the TCRαβ-complexes and the BV-chain of the endogenous TCR was studied with monoclonal antibodies (mAbs). No mAb directed against the BV-chain of the introduced TCR was available. Tetramers specific for the endogenous or introduced TCR were used to distinguish the TCRs from chimeric TCRαβ-complexes. We observed that after stimulation of either the endogenous or the introduced TCR the total amount of TCRs decreased to 40% and 70% of unstimulated cells, respectively. When the endogenous TCR was triggered, HA2- and PP65-tetramer stainings as well as the endogenous BV chain were diminished after 1 day. Stimulation of the introduced TCR decreased the introduced TCR expression after 1 day, and the endogenous TCR expression measured by tetramers and BV specific mAb was marginally decreased. Two days after stimulation of the introduced TCR and 3 days after stimulation of the endogenous TCR, the total amount of TCRαβ-complexes was restored. Staining with specific BV-mAb and tetramers demonstrated that the endogenous TCR expression, both after triggering of the endogenous as well as the introduced TCR, was still decreased at day 3. These data indicate that TCRαβ-complexes on the surface at days 2 and 3 mainly consisted of the introduced HA-2-TCR. TCR expression of non-transduced T cells was still decreased at day 3 after specific stimulation. Functional analysis of the T cells in a chromium release assay after 1 day of TCR triggering demonstrated that the T cells exerted reduced cytolytic activity that correlated with the downmodulation of TCR expression of either the introduced and endogenous TCR. The lytic activity of the T cells was restored at day 2 or 3 and correlated with the tetramer stainings. In conclusion, we observed physiological downmodulation of TCRs which were regulated by a retroviral promotor after antigen specific triggering. However, the introduced TCR was more quickly re-expressed at the cell surface, probably due to the increased retroviral promotor activity. The downmodulation upon specific triggering of both introduced and endogenous TCRs implies that cell mechanisms other than promotor activity are also involved in regulation of TCR cell surface expression.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2005
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Journal of Immunotherapy, Ovid Technologies (Wolters Kluwer Health), Vol. 34, No. 2 ( 2011-03), p. 165-174
    Type of Medium: Online Resource
    ISSN: 1524-9557
    Language: English
    Publisher: Ovid Technologies (Wolters Kluwer Health)
    Publication Date: 2011
    detail.hit.zdb_id: 2048797-6
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 106, No. 11 ( 2005-11-16), p. 5529-5529
    Abstract: Patients with hematological malignancies can be successfully treated by T cell-depleted allogeneic stem cell transplantation (alloSCT) followed by donor lymphocyte infusion (DLI). Failure of some relapsed hematological malignancies to respond to DLI is probably due to immune tolerance induced by regulatory and/or anergic T cells. We previously showed that functional T cells with redirected anti-leukemic reactivity can be generated by transfer of TCRs specific for minor histocompatibility antigens (mHags) to total peripheral blood mononuclear cells (PBMC) as well as CMV-specific T cells. By introducing TCRs into CMV or EBV specific T cells, T cells with proper memory/effector phenotypes are targeted, and due to virus persistence these T cells may show prolonged survival in vivo. The purpose of this study is to develop an efficient method for the isolation, retroviral transduction and expansion of TCR-transduced CMV- and EBV-specific T cells for cellular immunotherapy of patients with relapsed hematological malignancies after alloSCT. For clinical application, construction of single retroviral vectors coding for the α as well as β chains of mHag-specific TCRs is required. We used the MP71 retroviral vector for TCR gene transfer, since this vector contains Myeloproliferative Sarcoma Virus LTR sequences and a Mouse Embryonic Stem Cell Virus leader, which has been optimized for use in the clinic. The MP71 vector also contained a Woodchuck Hepatitis Response Element (WPRE). The WPRE, which is used as an element enhancing transgene expression at the post-transcriptional level, has recently been described to encode 60 amino acids of a protein with potential oncogenic activity. Therefore, we reconstructed the MP71 vector by introduction of a multiple cloning site (MCS) and, for safety issues, deleted the WPRE. The TCR α and β genes specific for the hematopoietic-restricted mHag HA-2 were linked by a 50-bp sequence encoding a “self-cleaving” 2A-like peptide and introduced into the MCS of the MP71 vector. Linkage of the TCR α and β genes by the 2A-like sequence allowed additional linkage of the low affinity nerve growth factor receptor (LNGFR) or human CD20 selection marker genes by an IRES sequence. The advantage of the human CD20 gene is that it can also function as suicide gene, allowing elimination of transduced cells in vivo if undesired side effects occur. Introduction of the single MP71 retroviral vector coding for the HA-2 TCR α and β chains as well as LNGFR into a TCR α- and β-deficient Jurkat T cell line led to high levels of TCR surface expression correlating with LNGFR marker gene expression. These data indicate proper cleavage and assembly of the transduced TCR α and β chains. Moreover, removal of the WPRE did not affect the surface expression level of the transduced TCR. Furthermore, CMV- and EBV-specific T cells were isolated from human individuals by the IFN-γ capture assay and subsequently transduced with a single retroviral vector coding for the HA-2-TCR α and β chains as well as LNGFR. CMV- and EBV-specific T cells from different human individuals could be successfully isolated to 60–90% purity and the TCR-transduced CMV- and EBV-specific T cells were shown to be fully functional, recognizing the viral peptides as well as the endogenously-processed HA-2 mHag.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2005
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: iScience, Elsevier BV, Vol. 27, No. 3 ( 2024-03), p. 109233-
    Type of Medium: Online Resource
    ISSN: 2589-0042
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2024
    detail.hit.zdb_id: 2927064-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 3832-3832
    Abstract: Therapeutic reactivity of CD20-specific monoclonal antibodies (mAb) or CD19-specific chimeric antigen receptor (CAR)-transduced T cells is exerted by targeting extracellular antigens. However, loss of CD20 and CD19 expression or absence of these molecules on other malignancies such as multiple myeloma restricts their application. Here, we identified the intracellular transcription factor Bob1 encoded by gene POU2AF1 as a suitable target for immunotherapy. Bob1 is highly expressed in CD19+ B cells, acute lymphoblastic leukemia (ALL), chronic lymphocytic leukemia (CLL), mantle cell lymphoma (MCL) and multiple myeloma (MM) and is absent in the non-B lineages including CD34+ hematopoietic progenitor cells (HPCs), T cells, fibroblasts, keratinocytes and gastrointestinal tract. Bob1 is localized intracellularly but HLA-presented Bob1-derived peptides are accessible on the cell surface to T cell receptors (TCRs) and can thus be recognized by T cells. From the HLA-presented ligandome (Mol Cell Proteomics, 2013;12:1829) we identified naturally processed Bob1-derived peptides displayed in HLA-A*0201 (HLA-A2) and in HLA-B*0702 (HLA-B7). Since auto-reactivity towards self-antigens such as Bob1 is prevented by depleting high-avidity T cells recognizing self-antigens in self-HLA, we exploited the immunogenicity of these peptides presented in allogeneic HLA. From a total of 3 x 109 peripheral blood mononuclear cells from 6 different HLA-A2/B7-negative healthy donors, we isolated and clonally expanded more than 1000 CD8+ T cells binding to peptide-MHC-tetramers composed of the Bob1-derived peptides bound to HLA-A2 or HLA-B7. The T cell clones were tested for stringent peptide-specificity by stimulation with Bob1-negative K562 cells expressing either HLA-A2 or B7 unloaded or pulsed with Bob1-derived peptides. This resulted in the selection of 15 T cell clones highly specific for Bob1. To identify the T cell clones of highest avidity, T cell clones were compared for peptide-sensitivity by testing the recognition of stimulator cells loaded with titrated amounts of Bob1-derived peptides and of Bob1-expressing HLA-A2/B7-positive EBV-transformed B cells. T cell clone 4G11 was selected because of high sensitivity and specificity for Bob1-derived peptide Bob144 presented in HLA-B7 and T cell clone 3C10 specifically recognized peptide Bob1245 bound to HLA-A2. Bob1-dependent recognition was demonstrated by transduction of Bob1 into cell lines that otherwise lack Bob1 expression. To investigate whether harmful toxicities could be caused by these T cell clones, we tested their reactivity against a wide panel of Bob1-negative stimulator cells demonstrating absence of recognition of HLA-B7-positive CD34+ HPCs, T cells, monocytes, immature and mature dendritic cells, and fibroblasts even under simulated inflamed conditions. Stringent HLA-B7-restricted recognition was observed for clone 4G11 when tested against a stimulator panel expressing a wide range of common and rare HLA class I and II molecules. These data illustrate a safe reactivity profile with little chance of off-target toxicity. To test their clinical applicability, clone 4G11 and 3C10 were tested for recognition of various primary B cell malignancies. Clone 4G11 efficiently recognized HLA-B7-positive primary ALL, CLL and mantle cell lymphoma while clone 3C10 recognized HLA-A2-positive primary B cell malignancies albeit to a lesser degree. Furthermore, reproducible strong recognition of purified primary HLA-B7-positive multiple myeloma could be demonstrated for clone 4G11. Therefore, T cell clone 4G11’s TCR may be used for immunotherapy by administering TCR-transduced T cells to multiple myeloma patients. To test whether introduction of 4G11’s TCR confers Bob1-reactivity onto recipient cells, the TCR was cloned into a retroviral vector. Highly specific reactivity against HLA-B7-positive Bob1-expressing target cells could be installed to TCR-transduced recipient T cells. In summary, we identified the intracellular transcription factor Bob1 encoded by gene POU2AF1 as a suitable target for TCR-based immunotherapies of B cell malignancies and multiple myeloma. Bob1-specific T cell clone 4G11 efficiently recognized primary B cell leukemia and multiple myeloma. TCR gene transfer approaches using Bob1-specific TCRs can bring novel treatment modalities for patients with B cell malignancies or multiple myeloma. Disclosures No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2014
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 128, No. 22 ( 2016-12-02), p. 914-914
    Abstract: T cells recognizing solid tumors and Hodgkin's lymphoma can be released from anergy by immune checkpoint inhibitors. Its therapeutic success seems to be associated with the abundance of neoepitopes within the tumor mutanome. In neoplastic B cells, VDJ recombination and somatic hypermutation (SHM) generate unique immunoglobulin (Ig) peptide sequences that predictably contribute to the lymphoma mutanome. Efficient presentation of a neoepitope by an individual's HLA complex is an essential requirement for neoepitope-directed T-cell immunity. Presentation of Ig-derived peptides in HLA class I has hitherto been demonstrated only indirectly, and there is as yet no direct evidence for presentation of Ig-derived neoepitopes of primary human lymphoma cells. We analyzed HLA-presented Ig-derived peptides in 9 clonal B-cell populations: 3 chronic lymphocytic leukemias (CLL), 1 hairy cell leukemia, 1 follicular lymphoma (FL), 3 EBV-transformed lymphoblastoid cell lines, and the U299 myeloma cell line. Full-length B-cell receptor (BCR) VDJ and VJ sequences were obtained by unbiased ARTISAN PCR and Pacific Biosciences next generation sequencing. 1067 unique Ig-derived nonamers were predicted to bind the HLA class I alleles expressed by the respective B-cell clone by the NetCTLpan bioinformatics tool. 650 candidate epitopes (60.9%) were derived from constant (C) regions; 417 (39.1%) from variable (V) regions. 146 predicted peptides from V regions (35.0%) contained at least one amino acid change due to SHM or at least one amino acid from CDR3 and were therefore considered potential neoepitopes. To investigate experimentally which epitopes are processed intracellularly and presented by HLA class I, HLA class I-peptide complexes were immunoaffinity purified by the monoclonal antibody W6.23 and analyzed by liquid chromatography and tandem mass spectrometry. In total, 53,663 unique peptides (8-15mers) were identified by Uniprot matching with a Mascot Ion Score of 〉 20. HLA ligandome sizes of individual B-cell populations ranged from 600 to 14,091 unique peptides per case. Within any HLA ligandome, 6 to 81 peptides were annotated as BCR-derived epitopes if they matched the individual BCR sequences. Of the total of 276 eluted BCR peptides, 203 (73.5%) where derived from C regions and 73 (26.5%) from V regions. 25 eluted peptides (range 0-10 per case) were derived from CDR3 regions or contained SHM-induced amino acid changes, thus fulfilling the definition of neoepitopes. Neoepitopes were detected in all cases with more than 109 cells as input material. Up to date, 4 neoepitopes have been synthesized and their mass spectra confirmed. Confirmation of all remaining neoepitopes is ongoing. No BCR-derived neoepitopes could be detected in an IGHV-unmutated CLL and the FL. These two cases had the lowest number of input cells and small overall ligandome sizes of only 600 and 1015 unique UniProt-matched peptides, respectively. We demonstrate for the first time that primary neoplastic B cells process and present BCR-derived neoepitopes in the context of HLA class I. Despite their origin from only two polypeptides, BCR epitopes and neoepitopes represent app. 0.5% and 0.05% of the total HLA class I ligandome, respectively. The challenging identification of BCR neoepitopes was possible by unbiased identification of BCR transcripts combined with next generation sequencing and targeted search for HLA class I-bound peptides. Matching fragmentation patterns of native and synthetic peptides suggest high specificity of this strategy. With respect to sensitivity, the ability to identify low frequency peptides appears to be strongly dependent on the amount of input cells. Our data close a gap in the mechanism underlying the evidence that highly immunogenic formulations of an idiotype vaccine are able to induce MHC class I-restricted cytotoxic T-cell responses. While phase III trials of idiotype vaccination aiming to induce anti-Ig antibodies failed to demonstrate convincing prolongation of clinical remissions achieved by chemotherapy, our data lend support for exploring idiotype-specific T-cell immunity against B-cell lymphomas. With recent advances in peptide synthesis, adjuvant formulations, and the availability of check point inhibitors to surpass regulatory activity, active immunotherapy targeting the lymphoma idiotype may regain appeal as truly personalized immune therapy. Disclosures No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2016
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Blood, American Society of Hematology, Vol. 108, No. 11 ( 2006-11-16), p. 5164-5164
    Abstract: HLA mismatched Stem Cell Transplantation (SCT) can be performed in patients with leukemia if no HLA identical donor can be found. Although HLA class I is expressed by almost all recipient cells and the frequency of allo-HLA reactive T-cells is high in all normal donors, the risk of GVHD after single locus mismatched SCT is comparable to that after HLA identical SCT. Thus, the occurrence of GVHD may not simply be explained by recognition of the mismatched HLA by allo-HLA reactive T-cells. Therefore, we characterized in detail the nature of the allo-immune response in an HLA mismatched setting. A patient with acute myeloid leukemia was treated with T-cell depleted SCT from a sibling donor who was HLA identical except for an HLA-A2 crossover. 6 Months after SCT, Donor Lymphocyte Infusion (DLI) of 2.5*10e6 CD3+ T-cells/kg was given for mixed chimerism caused by persistence of patient T-cells. No clinical response and no GVHD developed. 12 months after SCT the leukemia relapsed with 9% blasts in bone marrow, and a second DLI of 7.5*10e6 CD3+ T-cells/kg was given. The patient died of grade IV GVHD 5 weeks after DLI. durign the GVHD flow cytometry of PBMC’s showed conversion of patient to donor type T-cells. 80% Of the CD8 and 40% of the CD4 T-cells were activated, as determined by co-expression of HLA-DR. These activated T-cells were single cell sorted, non-specifically expanded, and tested for alloreactivity using cytotoxicity and cytokine production assays. 46 Out of 56 isolated CD8 clones and 7 out of 88 CD4 clones recognized patient but not donor target cells, indicating that at the time of the GVHD almost 70% of circulating T-cells were alloreactive. The response was highly polyclonal as shown by usage of at least 13 different TCR Vβs by the CD8 clones, and 6 by the CD4 clones. HLA restriction of the clones was tested with HLA blocking antibodies, a panel of HLA-typed target cells and donor EBV-LCL transduced with HLA-A2. All alloreactive CD8 clones were HLA-A2 specific. To further characterize the specificity, CD8 clones were tested against T2 cells loaded with HPLC fractions of peptides eluted from HLA-A2. Some CD8 clones recognized HLA-A2 with all different HPLC fractions, indicating peptide-independent recognition. Other clones recognized one fraction indicating peptide specificity, or several fractions indicating “promiscuous” peptide recognition. The CD4 clones were HLA-DR1 restricted and recognized donor EBV-LCL transduced with HLA-A2, indicating that the peptide recognized in HLA-DR1 was derived from the mismatched HLA-A2 molecule. Therefore, CD4 clones were tested against different peptides covering the whole HLA-A2 sequence. All clones recognized epitope 101–122 derived from a hyper variable region of HLA-A2. These results indicate that the GVHD in this HLA-A2 mismatched transplantation was caused by a combined highly polyclonal CD8 response directed against the HLA-A2 molecule and a CD4 response recognizing an HLA-A2 derived peptide presented by HLA class II. We speculate that the absence of an immune response observed after the first DLI despite high frequency of allo-HLA reactive T-cells, indicates that CD8 anti-HLA-A2 T-cells are insufficient to cause severe GVHD. We hypothesize that the rise of HLA-DR expressing leukemic blasts presenting HLA-A2 derived peptide in HLA class II triggered the CD4 response which was necessary to initiate the CD8 alloresponse resulting in this clinical outcome.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2006
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 1886-1886
    Abstract: Introduction: Use of T cells engineered to express antigen-specific T cell receptors (TCRs) has shown promise as a cancer immunotherapy treatment; however, durable responses have been limited by poor T cell persistence and expansion in vivo. Additionally, MHC class I downregulation on tumor cells weakens T cell recognition, further reducing therapeutic efficacy. To address these deficiencies, we co-expressed in human T cells a novel, small molecule (rimiducid)-dependent T cell activation switch, inducible MyD88/CD40 (iMC), along with PRAME-specific TCR to allow control of T cell expansion and activation, while upregulating MHC class I expression on tumor cells. Methods: Human T cells were activated with anti-CD3/CD28 and transduced with retrovirus encoding TCR α and β chains recognizing PRAME-derived, HLA-A*201-restricted peptide SLLQHLIGL (SFG-PRAME) or a polycistronic vector encoding the PRAME-specific TCR along with tandem rimiducid (AP1903)-binding domains (FKBP12v36) cloned in-frame with MyD88 and CD40 signaling domains (SFG-iMC-PRAME). Proliferation, cytokine production and cytotoxicity of modified T cells was assessed using peptide-pulsed T2 cells or against PRAME-expressing, HLA-A2+ U266 myeloma tumor cells with or without rimiducid (10 nM) stimulation. MHC class I expression on tumor cells was measured by flow cytometry using a transwell assay. In vitro tumor killing was analyzed using T cell and tumor coculture assays with various effector to target ratios over a 7-day period. In vivo efficacy was determined using immune-deficient NSG mice engrafted s.c. with U266 cells and treated i.v. with 1x107 transduced T cells. iMC was activated in vivo by weekly i.p. injections of 5 mg/kg rimiducid. Tumor size and T cell expansion was measured using in vivo luciferase bioluminescence imaging and flow cytometric phenotyping. Results: Both PRAME and iMC-PRAME retroviral vectors efficiently transduced activated human T cells (81±2.1% and 89±2.8%, respectively) and showed antigen-specific IFN-g production and cytolytic function against peptide-pulsed T2 cells and PRAME+ U266 myeloma cells. However, both TCR ligation and rimiducid-dependent costimulation were required for IL-2 production (PRAME, 217±256 pg/ml; iMC-PRAME, 23±56 pg/ml; iMC-PRAME + rimiducid, 5417±2599 pg/ml) against peptide-pulsed T2 cells. Coculture assays against PRAME-expressing U266 myeloma cells showed that tumor elimination was optimized with concurrent rimiducid-driven iMC activation, and this effect was accompanied by increased IL-2 secretion and robust T cell proliferation (PRAME, 0.18-fold; iMC-PRAME, 0.28-fold; iMC-PRAME + rimiducid, 7.7-fold). Further, iMC activation produced IFN-g independently of TCR ligation, which significantly increased MHC class I expression on tumor cells (no T cells, 61±3 MFI; PRAME, 1256±493 MFI; iMC-PRAME, 6747±656 MFI; iMC-PRAME + rimiducid, 9096±1583 MFI). In NSG mice engrafted with PRAME+ U266 myeloma tumors, PRAME TCR-modified T cells showed significant tumor control compared to non-transduced control T cells (p-value = 0.01, 0.01 and 0.0001 for PRAME, iMC-PRAME and iMC-PRAME + rimiducid, respectively) and rimiducid activation of iMC-PRAME-modified T cells showed significant tumor control compared to T cells transduced with only the PRAME TCR (p = 0.005). Importantly, weekly injections of rimiducid dramatically expanded PRAME TCR-expressing T cell numbers by 473-fold 4 weeks post-injection compared to T cells expressing the PRAME TCR only (p = 0.02). Summary: iMC is a novel "Go" switch that utilizes rimiducid, a small molecule dimerizer, to drive activation and expansion of PRAME-specific TCR-engineered T cells while sensitizing tumor to TCR-mediated recognition by upregulating MHC class I via IFN-g, thereby increasing antitumor efficacy and durability. Thus, iMC-PRAME is the prototype of a class of novel "Go-TCR" engineered T cell therapies that may increase efficacy, safety and durability of adoptive T cell therapies. Disclosures Hoang: Bellicum Pharmaceuticals: Employment. Foster:Bellicum Pharmaceuticals: Employment. Crisostomo:Bellicum Pharmaceuticals: Employment. Lu:Bellicum Pharmaceuticals: Employment. Moseley:Bellicum Pharmaceuticals: Employment, Equity Ownership. Slawin:Bellicum Pharmaceuticals: Employment, Equity Ownership. Spencer:Bellicum Pharmaceuticals: Employment, Equity Ownership.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2015
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 3008-3008
    Abstract: Abstract 3008 Invasive aspergillosis is a common and life-threatening complication in recipients of allogeneic stem cell transplantation (SCT). Patients are most severely at risk in the neutropenic phase, but there is increasing evidence that impaired T cell mediated immunity also increases the risk of invasive aspergillosis. In healthy individuals we identified Aspergillus -specific T cells by measuring IFN γ production in response to stimulation with overlapping peptides of the A. fumigatus proteins Crf1 and Catalase1. Antigen specific CD4+ T cells were single cell sorted to identify the recognized epitope and Aspergillus specificity was confirmed, based on reactivity to dendritic cells loaded with Aspergillus crude extract. Further characterization of the T cell mediated immune response against A. fumigatus is crucial for the development of new therapeutic strategies including adoptive transfer of antigen specific T cells. To investigate the role of Aspergillus -specific T cells in the clearance of aspergillus infection, we analyzed the T cell mediated immune response against A. fumigatus in patients who recovered from invasive aspergillosis. All patients had received an allogeneic SCT because of a hematological malignancy and aspergillus infection was diagnosed 4 to 5 months after SCT. At 4 time points after the diagnosis of invasive aspergillosis the T cell mediated immune response was analyzed. Peripheral blood mononuclear cells (PBMC) of patients who recovered from invasive aspergillosis were stimulated with the overlapping peptides of Crf1 and Catalase1. Directly ex vivo no Aspergillus -specific T cells could be detected by intracellular staining for IFN γ and CD154. However, when PBMC were stimulated with the peptide mixtures of Crf1 and Catalase1, cultured for 7 days in the presence of IL-2 and restimulated with Crf1 and Catalase1 we detected clear populations of Aspergillus -specific T cells in 3 of the 4 analyzed patients. In one patient 3% Crf1-specific and 1.7% Catalase1-specific CD154-expressing CD4+ T cells were shown 1 week after aspergillosis was diagnosed, coinciding with a rapid improvement of the infection with a clear regression of the pulmonary lesions. Up to 45% of the activated T cells produced IFN γ. After resolution of the infection, a decline in the percentage of activated and IFN γ producing T cells was observed, with a low percentage of 0.2% Aspergillus -specific CD4+ T cells persisting 3 years after the clearance of invasive aspergillosis. Two other patients were treated for aspergillosis for a long time, because of a prolonged period of leucopenia. Both were treated with prednisolone, one for GvHD, the other for auto-immune hemolytic anemia and granulocytopenia. The first patient had a mixed radiological response with a decline in the number of peribronchial infiltrates and a newly arisen nodular lesion 3 months after diagnosis, which was stable in size in the following 6 months. During this period no Aspergillus -specific T cells could be detected. After 1 year, 3 months after recovery of the leukocyte count, low numbers of Catalase1-specific CD4+ T cells were present coinciding with regression of the nodular lesion. Both Catalase1-specific (1%) and Crf1-specific (0.3%) CD4+ T cells could be shown 21 months after initial diagnosis, when the nodular lesion was still present, but declining in size. In the other patient 0.1 % Crf1-specific CD4+ T cells were detected during the whole period of Voriconazole treatment. One year after restoration of normal leukocyte counts and clearance of the aspergillus infection 0.65% Crf1-specific CD4+ T cells were present, but no Catalase1-specific T cells could be shown. In conclusion, using only 2 A. fumigatus proteins as target antigens, we have demonstrated the development of Aspergillus -specific T cells in 3 out of 4 patients, coinciding with the decline of aspergillus lesions. These data indicate that an immune response directed against A. fumigatus proteins helps to clear an aspergillus infection. Therefore, Aspergillus -specific T cells generated in vitro by stimulating with A. fumigatus proteins like Crf1 and Catalase1, may be used for adoptive T cell therapy for invasive aspergillosis. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2011
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Blood, American Society of Hematology, Vol. 94, No. 8 ( 1999-10-15), p. 2637-2646
    Abstract: Transgenic and gene targeted mice have contributed greatly to our understanding of the mechanisms underlying B-cell development. We describe here a model system that allows us to apply molecular genetic techniques to the analysis of human B-cell development. We constructed a retroviral vector with a multiple cloning site connected to a gene encoding green fluorescent protein by an internal ribosomal entry site. Human CD34+CD38− fetal liver cells, cultured overnight in a combination of stem cell factor and interleukin-7 (IL-7), could be transduced with 30% efficiency. We ligated the gene encoding the dominant negative helix loop helix (HLH) factor Id3 that inhibits many enhancing basic HLH transcription factors into this vector. CD34+CD38− FL cells were transduced with Id3-IRES-GFP and cultured with the murine stromal cell line S17. In addition, we cultured the transduced cells in a reaggregate culture system with an SV-transformed human fibroblast cell line (SV19). It was observed that overexpression of Id3 inhibited development of B cells in both culture systems. B-cell development was arrested at a stage before expression of the IL-7R. The development of CD34+CD38− cells into CD14+ myeloid cells in the S17 system was not inhibited by overexpression of Id3. Moreover, Id3+ cells, although inhibited in their B-cell development, were still able to develop into natural killer (NK) cells when cultured in a combination of Flt-3L, IL-7, and IL-15. These findings confirm the essential role of bHLH factors in B-cell development and demonstrate the feasibility of retrovirus-mediated gene transfer as a tool to genetically modify human B-cell development.
    Type of Medium: Online Resource
    ISSN: 1528-0020 , 0006-4971
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 1999
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...