Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Mol Med Rep ; 24(5)2021 Nov.
Article in English | MEDLINE | ID: mdl-34490477

ABSTRACT

Phage display technology (PD) is a powerful technique for the generation of tumor­targeting antibodies. However, there are a number of different selection methods established in different laboratories around the world. Cell­based PD panning methods using primary tumor cells are particularly heterogeneous between laboratories, which can lead to inconsistent results. Therefore, the present study evaluated different cell­based PD selection methods regarding their potential to generate acute myeloid leukemia (AML) blast­binding antibodies. In addition to this evaluation, the present study improved the PD procedure by optimizing selection as well as depletion strategies. To the best of our knowledge, the current study demonstrated for the first time that antigen diversity during the depletion step is of importance for the enrichment of tumor­targeting phage antibodies. It is demonstrated that medium levels of depletion antigen diversity led to the most promising antibody candidates. In addition, it was determined that purification of blast cells from patients with AML by immunomagnetic separation ameliorated the selection of AML­binding phages during panning. Furthermore, suggesting a common design­related mechanism using a 'single­pot' PD library, such as the well­known Tomlinson single­chain fragment variable (scFv) library, the present study identified specific binding consensus phage particles in independent panning procedures. By means of these optimized strategies, four promising AML blast­binding phage particles were isolated and soluble scFv­Fc (scFv cloned to a fragment crystallizable of an IgG2a mouse antibody) fusion proteins were produced. These scFv­Fc antibodies bound the surface of AML blasts and were successfully internalized into their cytoplasm, indicating that they are potential immunoconjugate candidates for AML immunotherapy.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neoplasm/immunology , Cell Surface Display Techniques/methods , Immunotherapy/methods , Leukemia, Myeloid, Acute/therapy , Antibody Specificity , Bacteriophages/immunology , HEK293 Cells , Humans , Primary Cell Culture
2.
J Virol Methods ; 243: 172-176, 2017 05.
Article in English | MEDLINE | ID: mdl-28193493

ABSTRACT

Bluetongue is an infectious viral disease which can cause mortality in affected ruminants, and tremendous economic damage via impacts upon fertility, milk production and the quality of wool. The disease is caused by bluetongue virus (BTV) which is transmitted by species of Culicoides biting midge. Rapid detection of BTV is required to contain disease outbreaks and reduce economic losses. The purpose of this study was to develop a monoclonal sandwich ELISA for direct detection of BTV in infected animals. Phage display technology was used to isolate BTV specific antibody fragments by applying the human scFv Tomlinson antibody libraries directly on purified BTV-8 particles. Three unique BTV-8 specific human antibody fragments were isolated which were able to detect purified BTV particles and also BTV in serum of an infected sheep. A combination of a human/mouse scFv-Fc chimeric fusion protein and a human Fab fragment in a sandwich ELISA format was able to detect BTV specifically with a limit of detection (LOD) of 104 infectious virus particles, as determined by tissue culture titration. This approach provided pilot data towards the development of a novel diagnostic test that might be used for direct detection of BTV-8 particles.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Antigens, Viral/analysis , Bluetongue virus/isolation & purification , Bluetongue/diagnosis , Enzyme-Linked Immunosorbent Assay/methods , Serum/virology , Animals , Bluetongue/virology , Bluetongue virus/immunology , Mice , Serologic Tests/methods , Sheep
3.
Immunol Lett ; 166(2): 65-78, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26045318

ABSTRACT

Phage display is an effective method for the generation of target-specific human antibodies. Standard phage display panning use purified proteins, antigen-transfected cells or tumor cell lines as target structure to generate specific antibodies. However, recombinant proteins can be difficult to express and purify in their native conformation and suitable cell lines are not always available. Additionally the antigen expression profile may change during cultivation and thus differ from the malignant cells in patient. Here we describe a method for the selection of specific antibodies from phage display libraries by panning against formalin-fixed paraffin-embedded (FFPE) tissue biopsies immobilized on glass slides, using small cell lung cancer (SCLC) as a case study. The human Tomlinson single-chain variable fragment (scFv) phage libraries I and J were panned against SCLC FFPE tissue slides for positive selection and healthy lung tissue for subtraction. The specificity of the selected scFv antibodies was confirmed in vitro by ELISA on immobilized SCLC cell membranes, by flow cytometry using the SCLC cell lines NCI-H69, NCI-H82 and DMS 273, and ex vivo against tissue microarrays containing 35 different SCLC samples and 20 types of normal organs. We monitored the internalization of three selected scFv antibodies and fused them with Pseudomonas exotoxin A (ETA') to produce immunotoxins whose cytotoxicity was confirmed by cell viability and apoptosis assays on different SCLC cell lines, achieving IC50 values of up to 23nM. The selection of SCLC-specific scFv antibodies by panning against FFPE tissue slides circumvents the challenges of using purified antigens or cell lines for antibody selection.


Subject(s)
Antibodies, Neoplasm/immunology , Cell Surface Display Techniques , Immunohistochemistry , Neoplasms/diagnosis , Neoplasms/immunology , Antibodies, Monoclonal , Antibody Specificity/immunology , Apoptosis/drug effects , Apoptosis/immunology , Biopsy , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Humans , Immunoglobulin Fab Fragments , Immunoglobulin Fc Fragments , Immunohistochemistry/methods , Immunotoxins/immunology , Immunotoxins/metabolism , Immunotoxins/toxicity , Peptide Library , Protein Binding , Recombinant Fusion Proteins , Single-Chain Antibodies/immunology
4.
Cancer Immunol Immunother ; 63(10): 1047-60, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24973179

ABSTRACT

Disseminated or relapsed Ewing sarcoma (EwS) has remained fatal in the majority of patients. A promising approach to preventing relapse after conventional therapy is to establish tumor antigen-specific immune control. Efficient and specific T cell memory against the tumor depends on the expansion of rare T cells with native specificity against target antigens overexpressed by the tumor. Candidate antigens in EwS include six-transmembrane epithelial antigen of the prostate-1 (STEAP1), and the human cancer/testis antigens X-antigen family member 1 (XAGE1) and preferentially expressed antigen in melanoma (PRAME). Here, we screened normal donors and EwS patients for the presence of circulating T cells reactive with overlapping peptide libraries of these antigens by IFN-γ Elispot analysis. The majority of 22 healthy donors lacked detectable memory T cell responses against STEAP1, XAGE1 and PRAME. Moreover, ex vivo detection of T cells specific for these antigens in both blood and bone marrow were limited to a minority of EwS patients and required nonspecific T cell prestimulation. Cytotoxic T cells specific for the tumor-associated antigens were efficiently and reliably generated by in vitro priming using professional antigen-presenting cells and optimized cytokine stimulation; however, these T cells failed to interact with native antigen processed by target cells and with EwS cells expressing the antigen. We conclude that EwS-associated antigens fail to induce efficient T cell receptor (TCR)-mediated antitumor immune responses even under optimized conditions. Strategies based on TCR engineering could provide a more effective means to manipulating T cell immunity toward targeted elimination of tumor cells.


Subject(s)
Antigens, Neoplasm/immunology , Sarcoma, Ewing/immunology , T-Lymphocytes, Cytotoxic/immunology , Adolescent , Adult , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/immunology , Antigens, Neoplasm/biosynthesis , Antigens, Neoplasm/pharmacology , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Case-Control Studies , Cell Line, Tumor , Child , Child, Preschool , Epitopes, T-Lymphocyte/immunology , Female , Humans , K562 Cells , Male , Oxidoreductases/biosynthesis , Oxidoreductases/immunology , Oxidoreductases/pharmacology , Sarcoma, Ewing/blood , Sarcoma, Ewing/pathology , T-Lymphocytes, Cytotoxic/drug effects , Young Adult
5.
Cancer Immunol Immunother ; 61(3): 385-96, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21928126

ABSTRACT

Specific cellular immunotherapy of cancer requires efficient generation and expansion of cytotoxic T lymphocytes (CTLs) that recognize tumor-associated self-antigens. Here, we investigated the capacity of human γδ T cells to induce expansion of CD8+ T cells specific for peptides derived from the weakly immunogenic tumor-associated self-antigens PRAME and STEAP1. Coincubation of aminobisphosphonate-stimulated human peripheral blood-derived γδ T cells (Vγ9+Vδ2+), loaded with HLA-A*02-restricted epitopes of PRAME, with autologous peripheral blood CD8+ T cells stimulated the expansion of peptide-specific cytolytic effector memory T cells. Moreover, peptide-loaded γδ T cells efficiently primed antigen-naive CD45RA+ CD8+ T cells against PRAME peptides. Direct comparisons with mature DCs revealed equal potency of γδ T cells and DCs in inducing primary T-cell responses and peptide-specific T-cell activation and expansion. Antigen presentation by γδ T-APCs was not able to overcome the limited capacity of peptide-specific T cells to interact with targets expressing full-length antigen. Importantly, T cells with regulatory phenotype (CD4+ CD25hiFoxP3+) were lower in cocultures with γδ T cells compared to DCs. In summary, bisphosphonate-activated γδ T cells permit generation of CTLs specific for weakly immunogenic tumor-associated epitopes. Exploiting this strategy for effective immunotherapy of cancer requires strategies that enhance the avidity of CTL responses to allow for efficient targeting of cancer.


Subject(s)
Autoantigens/immunology , CD8-Positive T-Lymphocytes/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , T-Lymphocytes/immunology , Antigen Presentation/immunology , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Autoantigens/genetics , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Cell Proliferation , Cells, Cultured , Coculture Techniques , Dendritic Cells/immunology , Dendritic Cells/metabolism , Diphosphonates/pharmacology , Epitopes, T-Lymphocyte/immunology , Flow Cytometry , HLA-A2 Antigen/immunology , Humans , Imidazoles/pharmacology , K562 Cells , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , Oxidoreductases/genetics , Oxidoreductases/immunology , Peptides/genetics , Peptides/immunology , Receptors, Antigen, T-Cell, alpha-beta/metabolism , T-Lymphocytes/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Transfection , Zoledronic Acid
6.
Clin Cancer Res ; 15(15): 4857-66, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19638467

ABSTRACT

PURPOSE: Novel natural killer (NK) cell-directed strategies in cancer immunotherapy aim at specifically modulating the balance between NK cell receptor signals toward tumor-specific activation. The signaling lymphocyte activation molecule-related receptor 2B4 (CD244) is an important regulator of NK cell activation. We investigated whether 2B4-enhanced activation signals can redirect the cytolytic function of human NK cells to NK cell-resistant and autologous leukemia and tumor targets. EXPERIMENTAL DESIGN: In vitro-stimulated NK cells from healthy donors and pediatric leukemia patients were gene modified with CD19 or G(D2)-specific chimeric receptors containing either the T-cell receptor zeta or 2B4 endodomain alone or combined. RESULTS: Chimeric 2B4 signaling alone failed to induce interleukin-2 receptor up-regulation and cytokine secretion but triggered a specific degranulation response. Integration of the 2B4 endodomain into T-cell receptor zeta chimeric receptors significantly enhanced all aspects of the NK cell activation response to antigen-expressing leukemia or neuroblastoma cells, including CD25 up-regulation, secretion of IFN-gamma and tumor necrosis factor-alpha, release of cytolytic granules, and growth inhibition, and overcame NK cell resistance of autologous leukemia cells while maintaining antigen specificity. CONCLUSION: These data indicate that the 2B4 receptor has a potent costimulatory effect in NK cells. Antigen-specific 2B4zeta-expressing NK cells may be a powerful new tool for adoptive immunotherapy of leukemia and other malignancies.


Subject(s)
Antigens, CD/immunology , Killer Cells, Natural/immunology , Receptors, Antigen/immunology , Receptors, Immunologic/immunology , Recombinant Fusion Proteins/immunology , Antigens, CD19/immunology , Antigens, CD19/metabolism , Cell Line, Tumor , Cytotoxicity, Immunologic , Humans , Immunotherapy, Adoptive , Killer Cells, Natural/transplantation , Leukemia/immunology , Leukemia/therapy , Lymphocyte Activation/immunology , Lysosomal-Associated Membrane Protein 1/immunology , Lysosomal-Associated Membrane Protein 1/metabolism , Neoplasms, Neuroepithelial/immunology , Neoplasms, Neuroepithelial/therapy , Neuroblastoma/immunology , Neuroblastoma/therapy , Protein Engineering , Receptors, Antigen/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction/immunology , Signaling Lymphocytic Activation Molecule Family
7.
Int J Cancer ; 125(4): 879-86, 2009 Aug 15.
Article in English | MEDLINE | ID: mdl-19480009

ABSTRACT

Immunosuppressive CD4+CD25(hi)FoxP3+ T cells (T(reg) cells) have been found at increased densities within the tumor microenvironment in many malignancies and interfere with protective antitumor immune responses. Osseous Ewing sarcomas (ESs) are thought to derive from a bone marrow (BM) mesenchymal cell of origin, and microscopic marrow involvement defines a subpopulation of patients at a high risk of relapse. We hypothesized that BM-resident T cells may contribute to a permissive milieu for immune escape of ESs. Using 6-color-flow cytometry, we investigated the pattern of immune cell subset distribution including NK cells, gammadelta T cells, central and effector memory CD8+ and CD4+ T cells as well as T cells with regulatory phenotype (T(reg) cells) in BM obtained at diagnosis from 45 primary or relapsed ES patients treated within standardized protocols. Although patients at relapse had an inverted CD4:CD8 T-cell ratio, neither CD8+ effector/memory T-cell subsets nor T(reg) cells significantly differed from patients at diagnosis. No significant associations of innate and effector/memory T-cell subpopulations with known risk factors were found, including age, gender, tumor site, primary metastases and histological tumor response. By contrast, T(reg) cells were found at significantly higher frequencies in patients with primary metastatic disease compared with localized ESs (5.0 vs. 3.3%, p = 0.01). Thus, increased BM T(reg) cells in patients with metastasized ES may reflect an immune escape mechanism that contributes to the development of metastatic disease. Immunotherapeutic strategies will have to adequately consider the regulatory milieu within areas of Ewing tumor-immune interactions.


Subject(s)
Bone Marrow/immunology , Bone Neoplasms/immunology , CD4 Antigens/metabolism , Forkhead Transcription Factors/metabolism , Interleukin-2 Receptor alpha Subunit/metabolism , Sarcoma, Ewing/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes/immunology , Adolescent , Adult , Bone Marrow/pathology , Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Child , Child, Preschool , Female , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/pathology , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Lymphatic Metastasis , Male , Neoplasm Recurrence, Local/diagnosis , Neoplasm Recurrence, Local/prevention & control , Phenotype , Prognosis , Sarcoma, Ewing/metabolism , Sarcoma, Ewing/pathology , Survival Rate , T-Lymphocytes/pathology , T-Lymphocytes, Regulatory/pathology , Young Adult
8.
Cancer Immunol Immunother ; 58(12): 1991-2001, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19360406

ABSTRACT

Regulatory NK cell receptors can contribute to antigen-specific adaptive immune responses by modulating T cell receptor (TCR)-induced T cell activation. We investigated the potential of the NK cell receptor 2B4 (CD244) to enhance tumor antigen-induced activation of human T cells. 2B4 is a member of the CD2 receptor subfamily with both activating and inhibitory functions in NK cells. In T cells, its expression is positively associated with the acquisition of a cytolytic effector memory phenotype. Recombinant chimeric receptors that link extracellular single-chain Fv fragments specific for the tumor-associated surface antigens CD19 and G(D2) to the signaling domains of human 2B4 and/or TCRzeta were expressed in non-specifically activated peripheral blood T cells by retroviral gene transfer. While 2B4 signaling alone failed to induce T cell effector functions or proliferation, it significantly augmented the antigen-specific activation responses induced by TCRzeta. 2B4 costimulation did not affect the predominant effector memory phenotype of expanding T cells, nor did it increase the proportion of T cells with regulatory phenotype (CD4+CD25(hi)FoxP3+). These data support a costimulatory role for 2B4 in human T cell subpopulations. As an amplifier of TCR-mediated signals, 2B4 may provide a powerful new tool for immunotherapy of cancer, promoting sustained activation and proliferation of gene-modified antitumor T cells.


Subject(s)
Antigens, CD/immunology , Receptors, Antigen, T-Cell/immunology , Receptors, Immunologic/immunology , T-Lymphocytes/immunology , Antigen Presentation , Antigens, CD/metabolism , Cell Growth Processes/immunology , Cell Line, Tumor , Epitopes , Humans , Immunologic Memory , Interferon-gamma/immunology , Killer Cells, Natural/immunology , Lymphocyte Activation , Receptors, Antigen, T-Cell/genetics , Receptors, Immunologic/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Signal Transduction , Signaling Lymphocytic Activation Molecule Family
9.
J Immunother ; 32(3): 310-21, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19242369

ABSTRACT

The efficacy of current cancer vaccines is limited by the functional heterogeneity and poor availability and expansion of professional antigen-presenting cells (APCs). Besides their potent innate effector properties, gammadelta T cells have been suggested to be involved in the initiation and maintenance of adaptive immune responses. Here, we investigated the capacity of human gammadelta T cells to induce expansion of virus-specific T cells to Epstein Barr virus (EBV) antigens. Aminobisphosphonate-stimulated human peripheral blood-derived gammadelta T cells (Vgamma2+Vdelta2+) acquired a dual phenotype characteristic for both APCs and effector memory T cells. Coincubation of activated gammadelta T cells pulsed with human leukocyte antigen-restricted epitopes of either the highly stimulatory EBV lytic cycle antigen Bam H1 Z fragment leftward open reading frame or the tumor-associated latent EBV antigen latent membrane protein 2a (LMP2a) with autologous peripheral blood lymphocytes induced selective expansion of peptide-specific, fully functional CD3CD8 cytolytic effector memory T cells. Furthermore, gammadelta T APCs efficiently processed and presented endogenous antigen, as demonstrated by the capacity of LMP2a gene-transduced gammadelta T cells to induce expansion of T cells with broad specificity for various LMP2a peptides. The capacity of autologous gammadelta T cells to induce LMP2a-specific autologous cytotoxic T lymphocytes was confirmed in 2 patients with Hodgkin lymphoma. In summary, bisphosphonate-activated human gammadelta T cells stimulate expansion of cytotoxic effector T cells specific for both subdominant and dominant viral epitopes and thus show promise as a novel source of efficient APCs for immunotherapy of viral and malignant disease.


Subject(s)
Antigens, Viral/immunology , CD8-Positive T-Lymphocytes/immunology , Epitopes/immunology , Epstein-Barr Virus Infections/therapy , Herpesvirus 4, Human/immunology , Immunotherapy , Receptors, Antigen, T-Cell, gamma-delta/immunology , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/virology , Antigens, Viral/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Diphosphonates/pharmacology , Epitopes/metabolism , Humans , Imidazoles/pharmacology , Receptors, Antigen, T-Cell, gamma-delta/drug effects , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Transduction, Genetic , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/immunology , Zoledronic Acid
10.
Cancer Res ; 67(17): 8335-43, 2007 Sep 01.
Article in English | MEDLINE | ID: mdl-17804749

ABSTRACT

T cells with grafted specificities for surface antigens provide an avenue for rapidly producing immune effector cells with tumor specificity. However, the function of chimeric receptor (chRec) gene-modified T cells is limited by lack of T-cell expansion and persistence. We propose to use varicella zoster virus (VZV)-reactive T cells as host for the chRec because these cells can be expanded both in vitro and in vivo by stimulation of their native receptor during endogenous reexposure to the virus or by administration of VZV vaccine. We obtained human T cells reactive with VZV from the peripheral blood of seropositive donors by stimulation with VZV lysate and evaluated their characteristics after genetic modification with two tumor-specific model chRecs. Cultures dominated by cytolytic CD4(+) T cells (VZV-CTL) could be expanded and maintained in vitro. Gene-modified VZV-CTL recognized and lysed tumor targets in a MHC-independent manner while maintaining functional, MHC-restricted interaction with VZV antigen through their native receptor. Thus, chRec-transduced VZV-CTL may provide a source of potent tumor-reactive cells for adoptive immunotherapy of cancer. The availability of a safe and effective VZV vaccine provides the option of repeated in vivo stimulation to maintain high T-cell numbers until the tumor is eliminated.


Subject(s)
CD4-Positive T-Lymphocytes/physiology , Herpesvirus 3, Human/immunology , Immunotherapy/methods , Neoplasms/immunology , Neoplasms/therapy , Organisms, Genetically Modified/physiology , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Cytotoxicity Tests, Immunologic , Humans , Lymphocyte Activation/immunology , Neoplasms/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Retroviridae/genetics , Substrate Specificity , T-Lymphocytes, Cytotoxic/metabolism , Transduction, Genetic
11.
Cancer Res ; 66(1): 24-8, 2006 Jan 01.
Article in English | MEDLINE | ID: mdl-16397210

ABSTRACT

Rhabdomyosarcomas are the most frequent malignant soft tissue tumors of childhood; however, because current multimodality treatments fail to improve the poor survival rate of children with metastatic rhabdomyosarcoma, new treatments are required. We previously identified the gamma-subunit of the fetal acetylcholine receptor (fAChR) as a specific cell surface target in rhabdomyosarcoma. Here, we engineered human T lymphocytes to express chimeric receptors composed of the antigen-binding domain of a human anti-fAChR antibody joined to the signaling domain of the human T-cell receptor zeta-chain. The interaction of fAChRzeta-transduced T cells with fAChR-positive rhabdomyosarcoma cell lines, but not with fAChR-negative control cells, induced T-cell activation characterized by strong secretion of IFN-gamma and delayed lysis of tumor cells. Importantly, we found that in six of six rhabdomyosarcoma patients, chemotherapy increased fAChR expression on residual tumor cells in vivo. Our observations suggest that these fully human chimeric fAChRzeta-transduced T cells, which should be well tolerated by the patient, have potential use in vivo both as a primary treatment for rhabdomyosarcoma and as a complementary approach to eradicate residual tumor cells after chemotherapy.


Subject(s)
Autoantibodies/immunology , Receptors, Cholinergic/immunology , Rhabdomyosarcoma/immunology , T-Lymphocytes/immunology , Autoantibodies/biosynthesis , Humans , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Interferon-gamma/metabolism , Lymphocyte Activation/immunology , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Membrane Proteins/immunology , Receptors, Antigen, T-Cell/biosynthesis , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Cholinergic/biosynthesis , Receptors, Cholinergic/genetics , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Rhabdomyosarcoma/drug therapy , Rhabdomyosarcoma/genetics , Rhabdomyosarcoma/metabolism , T-Cell Antigen Receptor Specificity/immunology , Transduction, Genetic
12.
J Immunother ; 29(1): 21-31, 2006.
Article in English | MEDLINE | ID: mdl-16365597

ABSTRACT

Human T cells expressing tumor antigen-specific chimeric receptors fail to sustain their growth and activation in vivo, which greatly reduces their therapeutic value. The defective proliferative response to tumor cells in vitro can partly be overcome by concomitant CD28 costimulatory signaling. We investigated whether T-cell activation via chimeric receptors (chRec) can be further improved by ligand expression on antigen-presenting cells of B-cell origin. We generated Epstein-Barr virus (EBV)-specific cytotoxic T lymphocytes (CTLs) expressing a CD19-specific chRec. These CTLs are provided with native receptor stimulation by autologous EBV-transformed B-lymphoblastoid cell lines (LCLs) but exclusively with chRec (CD19-specific) stimulation by allogeneic, human leukocyte antigen (HLA)-mismatched CD19+ LCLs. CD19zeta-transduced EBV-specific CTLs specifically lysed both allogeneic EBV targets and CD19+ tumor cells through the chRec in a major histocompatibility complex-independent manner, while maintaining their ability to recognize autologous EBV targets through the native T-cell receptor. The transduced CTLs failed to proliferate in response to CD19+ tumor targets even in the presence of CD28 costimulatory signaling. By contrast, CD19 expressed on HLA-mismatched LCL-induced T-cell activation and long-term proliferation that essentially duplicated the result from native receptor stimulation with autologous LCLs, suggesting that a deficit of costimulatory molecules on target cells in addition to CD28 is indeed responsible for inadequate chRec-mediated T-cell function. Hence, effective tumor immunotherapy may be favored if engagement of the chRec on modified T cells is complemented by interaction with multiple costimulator molecules. The use of T cells with native specificity for EBV may be one means of attaining this objective.


Subject(s)
Antigen-Presenting Cells/immunology , Leukemia, B-Cell/immunology , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes, Cytotoxic/immunology , Animals , Antigen Presentation/immunology , Antigens, CD19/immunology , Cell Line, Tumor , Cell Proliferation/drug effects , Genetic Engineering/methods , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/immunology , Humans , Immunotherapy, Adoptive/methods , Leukemia, B-Cell/drug therapy , Lymphocyte Activation/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Cytotoxic/virology , Transduction, Genetic
13.
Br J Haematol ; 126(4): 583-92, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15287953

ABSTRACT

Human peripheral blood gammadelta T cells (Vgamma9(+) Vdelta2(+)) can be selectively expanded in vivo by the systemic administration of aminobisphosphonates without prior antigen priming. To assess the potential of human gammadelta T cells to serve as effector cells of specific anti-tumour immunity, we expanded peripheral blood-derived gammadelta T cells and transduced them with recombinant retrovirus encoding G(D2)- or CD19-specific chimaeric receptors. Flow cytometric analysis of T cells from four individual donors cultured in the presence of zoledronate at day 14 of culture showed selective enrichment of the gammadelta T cell population (Vgamma9(+) Vdelta2(+) CD3(+) CD4(-) CD8(-)) to 73-96% of total CD3(+) T cells. Retroviral gene transfer resulted in chimaeric receptor surface expression in 73 +/- 12% of the population. Transduced gammadelta T cells efficiently recognized antigen-expressing tumour cell targets, as demonstrated by target-specific upregulation of CD69 and secretion of interferon-alpha. Moreover, transduced gammadelta T cells efficiently and specifically lysed the antigen-expressing tumour targets. They could be efficiently expanded in vitro and maintained in culture for prolonged periods. Zoledronate-activated human gammadelta T cells expressing chimaeric receptors may thus serve as potent and specific anti-tumour effector cells. Their responsiveness to stimulation with aminobisphosphonates may enable the selective re-expansion of adoptively transferred T cells in vivo, permitting long lasting anti-tumour immune control.


Subject(s)
Immunotherapy, Adoptive/methods , Neoplasms/immunology , Receptors, Antigen, T-Cell, gamma-delta/blood , T-Lymphocyte Subsets/immunology , Antigens, Neoplasm/immunology , Cell Division/drug effects , Cell Division/immunology , Cytotoxicity, Immunologic , Diphosphonates/pharmacology , Epitopes, T-Lymphocyte/immunology , Humans , Imidazoles/pharmacology , Immunophenotyping , Interferon-gamma/biosynthesis , Lymphocyte Activation/immunology , Retroviridae/genetics , T-Lymphocyte Subsets/drug effects , Transduction, Genetic , Tumor Cells, Cultured , Zoledronic Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...