Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Mol Immunol ; 48(12-13): 1369-76, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21481940

ABSTRACT

Induction of the TCR signaling pathway terminates the expression of RAG genes, and a link between this pathway and their transcriptional control is evident from the recent demonstration of their re-expression if the TCR is subsequently lost or down-regulated. Since unstimulated T cells display a steady-state level of "tonic" TCR signaling, i.e. in the absence of any antigenic stimulus, it was uncertain whether this control was exerted through ligand-dependent or ligand-independent TCR signaling. Here we demonstrate for the first time that exogenous TCR α and ß chains transferred into the human immature RAG(+) T cell line Sup-T1 by lentiviral transduction inhibit RAG expression through tonic signaling, and that this inhibition could itself be reverted by pharmacological tonic pathway inhibitors. We also suggest that mature T cells already expressing an endogenous TCR on their surface maintain some levels of plasticity at the RAG locus when their basal TCR signaling is interfered with. Lastly, we show that the TCR constructs employed in TCR gene therapy do not possess the same basal signaling transduction capability, a feature that may have therapeutic implications.


Subject(s)
DNA-Binding Proteins/genetics , Genes, RAG-1 , Nuclear Proteins/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Benzamides , Cell Line , Cell Membrane/immunology , Cell Membrane/metabolism , Child , DNA-Binding Proteins/metabolism , Dimethyl Sulfoxide/pharmacology , Down-Regulation , Flow Cytometry , Fluorescent Antibody Technique , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Imatinib Mesylate , Immunoblotting , Jurkat Cells , Ligands , Nuclear Proteins/metabolism , Piperazines/pharmacology , Pyrimidines/pharmacology , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/immunology , Reverse Transcriptase Polymerase Chain Reaction , Tacrolimus/pharmacology , Transduction, Genetic
2.
Blood ; 117(1): 72-82, 2011 Jan 06.
Article in English | MEDLINE | ID: mdl-20889925

ABSTRACT

Adoptive transfer of immune effector cells that are gene modified by retroviral transduction to express tumor-specific receptors constitutes an attractive approach to treat cancer. In patients with metastatic renal cell carcinoma, we performed a study with autologous T cells genetically retargeted with a chimeric antibody receptor (CAR) directed toward carbonic anhydrase IX (CAIX), an antigen highly expressed in renal cell carcinoma. In the majority of patients, we observed distinct humoral and/or cellular anti-CAIX-CAR T-cell immune responses in combination with a limited peripheral persistence of transferred CAIX-CAR T cells in the majority of patients. Humoral immune responses were anti-idiotypic in nature and neutralized CAIX-CAR-mediated T-cell function. Cellular anti-CAIX-CAR immune responses were directed to the complementarity-determining and framework regions of the CAR variable domains. In addition, 2 patients developed immunity directed against presumed retroviral vector epitopes. Here, we document the novel feature that therapeutic cells, which were ex vivo engineered by means of transduction with a minimal γ-retroviral vector, do express immunogenic vector-encoded epitopes, which might compromise persistence of these cells. These observations may constitute a critical concern for clinical ex vivo γ-retroviral gene transduction in general and CAR-retargeted T-cell therapy in particular, and underscore the need to attenuate the immunogenicity of both transgene and vector.


Subject(s)
Antigens, Neoplasm/immunology , Carbonic Anhydrases/immunology , Carcinoma, Renal Cell/therapy , Genetic Engineering , Genetic Vectors/immunology , Kidney Neoplasms/therapy , T-Lymphocytes/immunology , Transgenes/immunology , Adoptive Transfer , Amino Acid Sequence , Antigens, Neoplasm/genetics , Carbonic Anhydrase IX , Carbonic Anhydrases/genetics , Carcinoma, Renal Cell/immunology , Cohort Studies , Humans , Kidney Neoplasms/immunology , Lymphocyte Activation , Molecular Sequence Data , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Retroviridae/genetics , Transgenes/physiology
3.
J Immunol Methods ; 359(1-2): 11-20, 2010 Jul 31.
Article in English | MEDLINE | ID: mdl-20460126

ABSTRACT

Gene-engineered T cell therapy represents a promising strategy to treat cancers. To enable pre-selection of patients sensitive to this type of treatment we have setup and validated a T cell activation assay to test antigen expression on patient-derived tumor tissues. Chimeric antibody-based receptor (CAR) directed against CAIX, currently used in a clinical trial to treat RCC patients, was used as a model receptor. Primary human T cells expressing CAIX CAR were able to respond to CAIX-positive but not CAIX-negative tumor tissue and showed an increased production of IFNgamma, TNFalpha, IL-10 and IL-4, but not IL-2 or IL-5. Tumor tissue driven responses of primary T cells were paralleled by NFAT activation measured in CAR-transduced Jurkat T cells, which was shown to be triggered in a CAR and antigen-specific manner. Next, the reporter gene assay was applied to two independent PSMA CARs, which both mediated NFAT activation in response to tumor tissue. Taken together, a sensitive and donor-independent assay was established to measure T cell activation upon exposure to patient-derived tumor tissue, which may facilitate pre-selection of patients for clinical adoptive T cell therapy.


Subject(s)
Adoptive Transfer/methods , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/therapy , Kidney Neoplasms/immunology , Kidney Neoplasms/therapy , Lymphocyte Activation/immunology , T-Lymphocytes/immunology , T-Lymphocytes/transplantation , Antigen-Antibody Reactions , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Carbonic Anhydrase IX , Carbonic Anhydrases/genetics , Carbonic Anhydrases/immunology , Carcinoma, Renal Cell/pathology , Genetic Engineering , Humans , Jurkat Cells , Kidney Neoplasms/pathology , Patient Selection , Receptors, Antigen, T-Cell/immunology , Tumor Cells, Cultured
4.
Pharm Res ; 27(11): 2274-82, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20300804

ABSTRACT

PURPOSE: A new universal tool for specific, non-covalent and non-destructive attachment of a recombinant antibody fragment to a polymer-modified adenovirus has been utilised to regulate the tropism of adenoviral gene delivery vector. METHODS: We have prepared a multivalent reactive N-(2-hydroxypropyl)methacrylamide-based copolymer (PHPMA) bearing an α-bungarotoxin-binding peptide (BTXbp). The copolymer was used for covalent surface modification of adenoviral vectors (Ad). The α-bungarotoxin protein (BTX) has a nanomolar binding affinity for BTXbp, allowing non-covalent linkage of BTX fusion proteins. A single chain variable fragment of anti-PSMA antibody bearing BTX (scFv-BTX) binding to the prostate-specific membrane antigen (PSMA) was conjugated with the copolymer-coated adenovirus to enable specific infection of prostate cancer cells via PSMA receptors. RESULTS: As shown by ELISA, the copolymer-coated virus exhibited much reduced binding to anti-Ad antibodies. Infection of PC-3 and LNCaP prostate cancer cells was ∼100-fold less efficient with copolymer-coated Ad than with un-modified Ad. Conjugation of scFv-BTX with Ad-PHPMA-BTXbp led to 5-10-fold restoration of infection in PSMA-positive LNCaP cells. In PSMA-negative PC-3 cells, the conjugation of scFv-BTX with Ad-PHPMA-BTXbp gave no enhancement of infection. CONCLUSIONS: We have shown that the presented Ad-PHPMA-BTXbp/scFv-BTX system can be used as a universal tool for a receptor-specific virotherapy.


Subject(s)
Bungarotoxins/chemistry , Gene Transfer Techniques , Polymers , Amino Acid Sequence , Cell Line, Tumor , Chromatography, High Pressure Liquid , Enzyme-Linked Immunosorbent Assay , Humans , Molecular Sequence Data
5.
Trends Mol Med ; 16(2): 77-87, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20122868

ABSTRACT

T cell receptor (TCR) gene therapy provides patients with autologous T cells that are genetically engineered with TCRalphabeta chains and constitutes a promising approach for the treatment of tumors and virus infections. Among the current challenges of TCR gene therapy is the optimization of TCRalpha and beta transgene pairing to enhance the functional avidity of therapeutic T cells. Recently, various genetically modified TCRs have been developed that enhance TCR pairing and minimize mispairing, i.e. pairing between transgenic and endogenous TCR chains. Here, we classify such receptors according to their CD3-dependence for surface expression and review their abilities to address functional T cell avidity. In addition, we discuss the anticipated clinical value of these and other strategies to generate high-avidity T cells.


Subject(s)
Genetic Therapy , Neoplasms/genetics , Neoplasms/therapy , Receptors, Antigen, T-Cell/therapeutic use , Virus Diseases/genetics , Virus Diseases/therapy , Animals , Humans , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Signal Transduction
6.
Hum Gene Ther ; 21(7): 795-805, 2010 Jul.
Article in English | MEDLINE | ID: mdl-19947826

ABSTRACT

Prostate cancer is a leading cause of death among men in Western countries. Whereas the survival rate approaches 100% for patients with localized cancer, the results of treatment in patients with metastasized prostate cancer at diagnosis are much less successful. The patients are usually presented with a variety of treatment options, but therapeutic interventions in prostate cancer are associated with frequent adverse side effects. Gene therapy and oncolytic virus therapy may constitute new strategies. Already a wide variety of preclinical studies has demonstrated the therapeutic potential of such approaches, with oncolytic prostate-specific adenoviruses as the most prominent vector. The state of the art and future prospects of gene therapy in prostate cancer are reviewed, with a focus on adenoviral vectors. We summarize advances in adenovirus technology for prostate cancer treatment and highlight areas where further developments are necessary.


Subject(s)
Adenoviridae/genetics , Genetic Therapy , Genetic Vectors/genetics , Prostatic Neoplasms/therapy , Genetic Therapy/methods , Genetic Therapy/trends , Humans , Male
7.
Hum Gene Ther ; 21(7): 807-13, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20001452

ABSTRACT

Prostate cancer is at present the most common malignancy in men in the Western world. When localized to the prostate, this disease can be treated by curative therapy such as surgery and radiotherapy. However, a substantial number of patients experience a recurrence, resulting in spreading of tumor cells to other parts of the body. In this advanced stage of the disease only palliative treatment is available. Therefore, there is a clear clinical need for new treatment modalities that can, on the one hand, enhance the cure rate of primary therapy for localized prostate cancer and, on the other hand, improve the treatment of metastasized disease. Gene therapy is now being explored in the clinic as a treatment option for the various stages of prostate cancer. Current clinical experiences are based predominantly on trials with adenoviral vectors. As the first of a trilogy of reviews on the state of the art and future prospects of gene therapy in prostate cancer, this review focuses on the clinical experiences and progress of adenovirus-mediated gene therapy for this disease.


Subject(s)
Adenoviridae/genetics , Genetic Therapy/methods , Prostatic Neoplasms/therapy , Genetic Therapy/trends , Humans , Male , Neoplasm Staging , Prostatic Neoplasms/radiotherapy , Prostatic Neoplasms/surgery , Treatment Outcome
8.
Cancer Res ; 69(24): 9385-94, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-19996290

ABSTRACT

T-cell receptor (TCR) gene transfer for cancer immunotherapy is limited by the availability of large numbers of tumor-specific T cells. TCR alpha and beta chains were isolated from a highly lytic HLA-A2-restricted cytotoxic T lymphocyte (CTL) clone recognizing the melanoma-associated Melan-A/MART-1 antigen and inserted into a lentiviral vector carrying a bidirectional promoter capable of robust and coordinated expression of the two transgenes. Lentiviral vector-based gene delivery systems have shown increased transfer efficiency and transgene expression compared with the widely used gamma-retroviral vectors. This vector performed more efficiently than a gamma-retrovirus-based vector containing the same expression cassette, resulting in a T-cell population with 60% to 80% of transgenic TCR expression with mainly CD8(+) intermediate effector phenotype. Transgenic T cells specifically produced cytokine in response to and killed antigen-expressing melanoma cells, retained an overlapping functional avidity in comparison with the TCR donor CTL clone, and exerted significant therapeutic effects in vivo upon adoptive transfer in melanoma-bearing severe combined immunodeficient mice. Optical imaging showed their accumulation in the tumor site. Overall, our results indicate that lentiviral vectors represent a valid tool for stable and high-intensity expression of transgenic TCR and support clinical exploitation of this approach for therapeutic application.


Subject(s)
Genes, T-Cell Receptor alpha , Genes, T-Cell Receptor beta , Immunotherapy, Adoptive/methods , Melanoma/therapy , T-Lymphocytes/physiology , Animals , Antigens, Neoplasm/immunology , Epitopes , Female , Genetic Vectors/genetics , HLA-A2 Antigen/immunology , Humans , Immunologic Memory , Jurkat Cells , Lentivirus/genetics , Leukocytes, Mononuclear/immunology , MART-1 Antigen , Melanoma/genetics , Melanoma/immunology , Mice , Mice, SCID , Neoplasm Proteins/immunology , Receptors, Antigen, T-Cell, alpha-beta/biosynthesis , Receptors, Antigen, T-Cell, alpha-beta/immunology , T-Lymphocytes/immunology , Transduction, Genetic
9.
J Immunother ; 32(3): 272-9, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19242373

ABSTRACT

The Food and Drug Administration/Center for Biologics Evaluation and Research has defined that for retroviral gene therapy, the vector-producing cell, the vector preparation, and the ex vivo gene-transduced cells have to be tested for absence of replication-competent retrovirus (RCR) if the transduced cells are cultured for >4 days. We assessed the sensitivity of the "extended PG4(S+L-) assay" to detect gibbon ape leukemia virus (GALV) RCR, and applied this assay to measure GALV RCR spread in retrovirally transduced T cells. To this end, T cells were expanded for 12 days after transduction with a GALV-envelope pseudotyped retroviral vector expressing single chain variable fragment (anticarbonic anhydrase IX) in presence or absence of GALV RCR. Results showed that: (1) the "extended PG4(S+L-) assay" detects 1 focus-forming unit (ffu) GALV RCR and thus is applicable and sufficiently sensitive to screen human T-cell cultures for absence of infectious GALV RCR; (2) although GALV RCR infect human T cells, it very poorly replicate in T cells; (3) GALV RCR, when present at low levels immediately upon gene transduction (ie, 100 ffu/20x10 T cells in 100 mL), did not spread during a 12-day T-cell culture at clinical scale. Our observation that GALV RCR poorly spreads in primary human T-cell cultures questions the relevance of testing T-cell transductants for RCR on top of testing the vector-producing cells and the clinical vector batch for RCR and warrants evaluation of the current policy for safety testing of ex vivo retrovirally transduced T lymphocytes for GALV RCR.


Subject(s)
Genetic Vectors/physiology , Leukemia Virus, Gibbon Ape/physiology , T-Lymphocytes/virology , Virus Replication , Animals , Biological Assay , Genetic Vectors/genetics , Humans , Leukemia Virus, Gibbon Ape/genetics , Sensitivity and Specificity , Transduction, Genetic
10.
Cytometry A ; 73(11): 1093-9, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18785268

ABSTRACT

Adoptive transfer of antigen-specific T-cells has shown therapeutic successes in the treatment of tumors in patients with metastatic melanoma. Tumor antigen-specific T-lymphocytes, however, occur only at low frequencies in a small proportion of patients. This low T-lymphocyte frequency together with the difficulties associated with in vitro generation of T-lymphocytes specific for cancers other than melanoma hampers adoptive T cell therapy. To make adoptive T-cell therapy more uniformly applicable, strategies were developed at transferring tumor-specificity to primary human T-lymphocytes via antibody (Ig) or T-cell receptor (TCR) molecules. We exploited the selection power of phage display that allows for the testing of tens of billions of individual clones with a high-throughput selection of Fabs with peptide/MHC complex binding capacity. Following in vitro selection, human "TCR-like" Fab fragments have been functionally expressed on human T-lymphocytes, resulting in MHC-restricted, tumor-specific lysis and cytokine production. Currently, we have extended our selections to a panel of class I and II MHC-restricted MAGE and other tumor-specific epitopes, and would like to propose that phage display represents a technology able to expand T-cell therapy to numerous tumor types.


Subject(s)
Epitopes, T-Lymphocyte/immunology , Immunoglobulin Fragments/immunology , Immunotherapy, Adoptive , Neoplasms/immunology , Antibodies, Neoplasm/immunology , Humans , Receptors, Antigen, T-Cell/immunology
11.
J Immunol ; 180(11): 7736-46, 2008 Jun 01.
Article in English | MEDLINE | ID: mdl-18490778

ABSTRACT

TCR gene therapy is adversely affected by newly formed TCRalphabeta heterodimers comprising exogenous and endogenous TCR chains that dilute expression of transgenic TCRalphabeta dimers and are potentially self-reactive. We have addressed TCR mispairing by using a modified two-chain TCR that encompasses total human CD3zeta with specificities for three different Ags. Transfer of either TCRalpha:CD3zeta or beta:CD3zeta genes alone does not result in surface expression, whereas transfer of both modified TCR chains results in high surface expression, binding of peptide-MHC complexes and Ag-specific T cell functions. Genetic introduction of TCRalphabeta:zeta does not compromise surface expression and functions of an endogenous TCRalphabeta. Flow cytometry fluorescence resonance energy transfer and biochemical analyses demonstrate that TCRalphabeta:CD3zeta is the first strategy that results in highly preferred pairing between CD3zeta-modified TCRalpha and beta chains as well as absence of TCR mispairing between TCR:CD3zeta and nonmodified TCR chains. Intracellular assembly and surface expression of TCR:CD3zeta chains is independent of endogenous CD3gamma, delta, and epsilon. Taken together, our data support the use of TCRalphabeta:CD3zeta to prevent TCR mispairing, which may provide an adequate strategy to enhance efficacy and safety of TCR gene transfer.


Subject(s)
CD3 Complex/genetics , Receptors, Antigen, T-Cell, alpha-beta/genetics , T-Lymphocytes/immunology , CD3 Complex/immunology , CD3 Complex/metabolism , Cell Line , Gene Transfer Techniques , Humans , Jurkat Cells , Receptors, Antigen, T-Cell, alpha-beta/immunology , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Signal Transduction , T-Lymphocytes/metabolism
12.
J Immunol ; 180(1): 391-401, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-18097040

ABSTRACT

A promising approach to adoptive transfer therapy of tumors is to reprogram autologous T lymphocytes by TCR gene transfer of defined Ag specificity. An obstacle, however, is the undesired pairing of introduced TCRalpha- and TCRbeta-chains with the endogenous TCR chains. These events vary depending on the individual endogenous TCR and they not only may reduce the levels of cell surface-introduced TCR but also may generate hybrid TCR with unknown Ag specificities. We show that such hybrid heterodimers can be generated even by the pairing of human and mouse TCRalpha- and TCRbeta-chains. To overcome this hurdle, we have identified a pair of amino acid residues in the crystal structure of a TCR that lie at the interface of associated TCR Calpha and Cbeta domains and are related to each other by both a complementary steric interaction analogous to a "knob-into-hole" configuration and the electrostatic environment. We mutated the two residues so as to invert the sense of this interaction analogous to a charged "hole-into-knob" configuration. We show that this inversion in the CalphaCbeta interface promotes selective assembly of the introduced TCR while preserving its specificity and avidity for Ag ligand. Noteworthily, this TCR modification was equally efficient on both a Mu and a Hu TCR. Our data suggest that this approach is generally applicable to TCR independently of their Ag specificity and affinity, subset distribution, and species of origin. Thus, this strategy may optimize TCR gene transfer to efficiently and safely reprogram random T cells into tumor-reactive T cells.


Subject(s)
Gene Transfer Techniques , Receptors, Antigen, T-Cell, alpha-beta/chemistry , Receptors, Antigen, T-Cell, alpha-beta/genetics , T-Lymphocytes/immunology , Animals , Cell Membrane/metabolism , Crystallography, X-Ray , Dimerization , Humans , Mice , Point Mutation , Protein Conformation , Protein Transport , Receptors, Antigen, T-Cell, alpha-beta/metabolism
13.
Cancer Res ; 67(23): 11309-16, 2007 Dec 01.
Article in English | MEDLINE | ID: mdl-18056457

ABSTRACT

To improve safety and specificity of oncolytic adenoviruses, we introduced T-cell receptors (TCR) specific for a unique class of truly tumor-specific antigens into the adenoviral fiber protein. The adenoviral fiber knob responsible for attachment to the coxsackie-adenoviral receptor (CAR) on target cells was replaced by a single-chain TCR (scTCR) molecule with specificity for the melanoma-associated cancer-testis antigen MAGE-A1, presented by HLA-A1, and an extrinsic trimerization motif in a replicating Ad5 vector (Ad5.R1-scTCR). The production of the recombinant virus was initiated in a novel producer cell line that expressed an antibody-based hexon-specific receptor (293T-AdR) in the cell membrane. This new production system allowed CAR-independent and target antigen-independent propagation of Ad5.R1-scTCR. Infection with adenovirus bearing the scTCR-based fiber resulted in an efficient killing of target tumor cells. The infection was cell type specific because only HLA-A1(+)/MAGE-A1(+) melanoma cells were killed, and thus, this retargeting strategy provides a versatile tool for future clinical application.


Subject(s)
Adenoviridae Infections/virology , Adenoviridae/physiology , Melanoma/virology , Oncolytic Virotherapy , Receptors, Antigen, T-Cell/immunology , Adenoviridae Infections/immunology , Adenoviridae Infections/therapy , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Blotting, Western , Coxsackie and Adenovirus Receptor-Like Membrane Protein , Flow Cytometry , Genetic Vectors , HLA-A1 Antigen/immunology , HLA-A1 Antigen/metabolism , Humans , Melanoma/immunology , Melanoma/therapy , Melanoma-Specific Antigens , Neoplasm Proteins/immunology , Neoplasm Proteins/metabolism , Receptors, Antigen, T-Cell/metabolism , Receptors, Virus/immunology , Receptors, Virus/metabolism , Tumor Cells, Cultured , Virus Replication
14.
J Gene Med ; 9(7): 561-70, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17471588

ABSTRACT

BACKGROUND: T cell receptor (TCR) gene therapy represents an attractive anti-cancer treatment but requires further optimization of its efficacy and safety in clinically relevant models, such as those using a tumor antigen and TCR of human origin. Currently, however, there is no consensus as to what protocol is most optimal for retroviral human TCR gene transfer into primary murine T cells, most notably with respect to virus pseudo-type. METHODS: Primary murine T cells were transduced, expanded and subsequently tested for transgene expression, proliferation and antigen-specific function. To this end, murine leukemia virus (MLV) retroviruses were produced upon transfection of various packaging cells with genes encoding either green fluorescent protein (GFP) or TCRalphabeta specific for human melanoma antigen gp100(280-288) and the helper elements GAG/POL and ENV. Next to viral pseudotyping, the following parameters were studied: T cell densities; T cell activation; the amounts of IL-2 and the source of serum used to supplement medium. RESULTS: The pseudo-type of virus produced by packaging cells critically determines T cell transduction efficiencies. In fact, MLV-A and MLV-E pseudo-typed viruses derived from a co-culture of Phoenix-A and 293T cells resulted in T cell transduction efficiencies that were two-fold higher than those based on retroviruses expressing either VSV-G, GALV, MLV-A or MLV-E envelopes. In addition, T cell densities during transduction were inversely related to transduction efficiencies. Further optimization resulted in transduction efficiencies of over 90% for GFP, and 68% for both a murine and a human (i.e. murinized) TCR. Importantly, TCR-transduced T cells proliferate (i.e. showing a log increase in cell number in a few days) and show antigen-specific function. CONCLUSIONS: We set up a quick and versatile method to genetically modify primary murine T cells based on transient production of TCR-positive retroviruses, and show that retroviral gene transfer of a human TCR into primary murine T cells is critically improved by viral pseudo-typing with both MLV-A and MLV-E envelopes.


Subject(s)
Membrane Glycoproteins/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Virus/metabolism , Sodium-Phosphate Cotransporter Proteins, Type III/metabolism , T-Lymphocytes/metabolism , T-Lymphocytes/virology , Transduction, Genetic/methods , Virus Assembly , Animals , Antibodies, Monoclonal , CD28 Antigens/immunology , CD3 Complex/immunology , Cell Count , Cell Proliferation/drug effects , Concanavalin A/pharmacology , Humans , Lymphocyte Activation/drug effects , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Receptors, Antigen, T-Cell/metabolism , Receptors, Virus/genetics , Retroviridae/genetics , Retroviridae/metabolism , Solubility/drug effects , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Virus Assembly/drug effects
15.
J Immunol ; 177(2): 991-8, 2006 Jul 15.
Article in English | MEDLINE | ID: mdl-16818755

ABSTRACT

Therapeutic success of TCR gene transfer to treat tumors depends on the ability of redirected T cells to become activated upon tumor recognition in vivo. Help provided by tumor-specific Th1 cells is reported to relieve T cells from an anergized state and to induce tumor regression. We recently demonstrated the ability to generate melanoma-specific Th1 cells by genetic introduction of both a CD8-dependent TCR and the CD8alpha coreceptor into CD4+ T cells. In this study, we analyzed a TCR that binds Ag independently of CD8, a property generally preferred to induce tumor-specific T cell responses, and addressed the contribution of CD8alpha following introduction into TCR-transduced CD4+ T cells. To this end, primary human CD4+ T cells were gene transferred with a high-avidity TCR, and were shown not only to bind peptide/MHC class I, but also to effectively kill Ag-positive tumor cells in the absence of CD8alpha. The introduction of CD8alpha up-regulates the tumor-specific production of TNF-alpha and IL-2 to some extent, but significantly down-regulates production of IL-4, IL-5, and IL-10 in CD4+ T cells. The introduction of a mutated cysteine motif in CD8alpha, which prevents its binding to LCK and linker for activation of T cells, did not adversely affect expression and T cell cytotoxicity, but counteracted the CD8alpha-mediated down-regulation of IL-4 and IL-5, but not IL-10. In conclusion, CD8alpha down-regulates the production of major Th2-type cytokines, in part mediated by LCK and/or linker for activation of T cells, and may induce differentiation of tumor-specific Th1 cells, which makes this coreceptor an interesting candidate to improve the clinical potential of TCR gene transfer to treat cancer.


Subject(s)
CD8 Antigens/genetics , Down-Regulation/immunology , Gene Transfer Techniques , Interleukin-10/antagonists & inhibitors , Interleukin-4/antagonists & inhibitors , Interleukin-5/antagonists & inhibitors , Melanoma/therapy , Receptors, Antigen, T-Cell, alpha-beta/genetics , Amino Acid Motifs/genetics , CD8 Antigens/physiology , CD8 Antigens/therapeutic use , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Line , Cell Line, Tumor , Cysteine/genetics , Cytotoxicity, Immunologic/genetics , Down-Regulation/genetics , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/metabolism , HLA-A2 Antigen/immunology , HLA-A2 Antigen/metabolism , Humans , Interleukin-10/biosynthesis , Interleukin-4/biosynthesis , Interleukin-5/biosynthesis , Melanoma/genetics , Melanoma/immunology , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , Peptide Fragments/immunology , Peptide Fragments/metabolism , Protein Binding/genetics , Protein Binding/immunology , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Receptors, Antigen, T-Cell, alpha-beta/therapeutic use , Th1 Cells/cytology , Th1 Cells/immunology , Th1 Cells/metabolism , Transduction, Genetic/methods , gp100 Melanoma Antigen
16.
Int Immunol ; 18(4): 591-601, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16507598

ABSTRACT

EBV is associated with a broad range of malignancies. Adoptive immunotherapy of these tumors with EBV-specific CTL proved useful. We generated a panel of primary human T cells specific to various EBV antigens (i.e. Epstein-Barr nuclear antigen 3A, 3B and BamHI-M leftward reading frame) via transfer of modified TCR genes that are either coupled to CD3zeta or Fc(epsilon)RIgamma. TCR-transduced T cells from 20-60% of donors (total number of 25) demonstrated specific lysis of EBV peptide-loaded target cells, whereas lymphoblastoid cell lines expressing native EBV antigens were not killed by any of the EBV-specific T cell populations. This non-responsiveness, confirmed at the level of nuclear factor of activated T cells activation, is not due to receptor configuration since identical receptor formats specific for melanoma antigens successfully re-targeted T cells to native melanoma cells. In an effort to generate a more potent receptor, we introduced a CD28 domain into one of the EBV-specific TCR. This TCR did not affect the cytotoxic response of re-targeted T cells, but dramatically enhanced antigen-specific IFNgamma production. We therefore conclude that these novel CD28-containing EBV-specific TCRs provide a basis for further development of TCR gene transfer to treat EBV-induced diseases.


Subject(s)
Epstein-Barr Virus Nuclear Antigens/immunology , Gene Transfer Techniques , Genes, T-Cell Receptor , Interferon-gamma/biosynthesis , T-Lymphocytes/immunology , CD28 Antigens/immunology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Genetic Therapy/methods , Humans , Jurkat Cells , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Transduction, Genetic
17.
J Immunol ; 174(12): 7853-8, 2005 Jun 15.
Article in English | MEDLINE | ID: mdl-15944290

ABSTRACT

T cells require both primary and costimulatory signals for optimal activation. The primary Ag-specific signal is delivered by engagement of the TCR. The second Ag-independent costimulatory signal is mediated by engagement of the T cell surface costimulatory molecule CD28 with its target cell ligand B7. However, many tumor cells do not express these costimulatory molecules. We previously constructed phage display derived F(AB), G8, and Hyb3, Ab-based receptors with identical specificity but distinct affinities for HLA-A1/MAGE-A1, i.e., "TCR-like" specificity. These chimeric receptors comprised the FcepsilonRI-gamma signaling element. We analyzed whether linking the CD28 costimulation structure to it (gamma + CD28) could affect the levels of MHC-restricted cytolysis and/or cytokine production. Human scFv-G8(POS) T lymphocytes comprising the gamma + CD28 vs the gamma signaling element alone produced substantially more IL-2, TNF-alpha, and IFN-gamma in response to HLA-A1/MAGE-A1(POS) melanoma cells. Also a drastic increase in cytolytic capacity of scFv-G8(POS) T cells, equipped with gamma + CD28 vs the gamma-chain alone was observed.


Subject(s)
Binding Sites, Antibody , CD28 Antigens/physiology , Cytokines/biosynthesis , Cytotoxicity, Immunologic , Epitopes, T-Lymphocyte/toxicity , HLA-A1 Antigen/immunology , Lymphocyte Activation , T-Lymphocytes, Cytotoxic/immunology , Adjuvants, Immunologic/genetics , Adjuvants, Immunologic/metabolism , Adjuvants, Immunologic/toxicity , Antigens, Neoplasm , Binding Sites, Antibody/genetics , CD28 Antigens/genetics , CD28 Antigens/immunology , Cell Line, Tumor , Cytotoxicity, Immunologic/genetics , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , HLA-A1 Antigen/genetics , HLA-A1 Antigen/metabolism , Humans , Immunoglobulin Fab Fragments/genetics , Immunoglobulin Fab Fragments/metabolism , Immunoglobulin Fab Fragments/toxicity , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/metabolism , K562 Cells , Lymphocyte Activation/genetics , Melanoma/immunology , Melanoma/pathology , Melanoma-Specific Antigens , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Neoplasm Proteins/metabolism , Protein Structure, Tertiary/genetics , Receptors, Antigen, T-Cell/biosynthesis , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Receptors, IgE/genetics , Receptors, IgE/metabolism , Receptors, IgE/physiology , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocytes, Cytotoxic/metabolism , Transduction, Genetic/methods
18.
J Gene Med ; 7(2): 158-70, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15538730

ABSTRACT

BACKGROUND: Novel antibody-based immunotherapeutic strategies exploit chimeric immune receptors (CIR), expressed on the surface of transduced human peripheral blood mononuclear cells (PBMC), to redirect potent non-MHC-dependent cytotoxicity to tumor cells expressing a tumor-associated antigen. However, clinical application of the strategy has been hampered by the potential side effects associated with immunogenicity and by low transduction efficiency. METHODS: A fully human CIR was constructed that triggers immune activation through the zeta chain of CD3 and contains a human single-chain antibody fragment specific for an extracellular epitope of HER2. PBMC were transduced with the CIR using gibbon-ape leukemia virus envelope pseudotyped retroviruses. In vitro cytotoxicity and inhibition assays were carried out using normal and tumor cell lines expressing different levels of HER2. RESULTS: Bulk populations of CIR-transduced PBMC could express high levels of the construct and subcloning ensured stable expression. CIR-mediated killing and growth inhibition of targets expressing high HER2 levels were very efficient at low effector-to-target ratios. Under the same experimental conditions, CIR-mediated activity against normal cells expressing low HER2 levels was marginal. The CIR-mediated recognition of target cells induced the release of soluble factors able to inhibit growth of both HER-positive and HER2-negative bystander tumor cells. CONCLUSIONS: Human CIR-transduced PBMC exert a potent and dose-dependent anti-tumor activity. Target antigen level appeared to be a critical determinant of specificity and delivery of signals leading to redirected effector functions. Soluble factors, released by redirected effectors at the site of antigen-driven activation, mediate potent bystander killing.


Subject(s)
Genetic Therapy/methods , Genetic Vectors/genetics , Immunotherapy/methods , Leukemia Virus, Gibbon Ape/genetics , Lymphocytes/metabolism , Neoplasms/therapy , Receptor, ErbB-2/metabolism , Antibodies, Monoclonal , Blotting, Western , CD3 Complex/genetics , CD3 Complex/immunology , Cell Line, Tumor , Cell Proliferation , Chromium Radioisotopes , DNA Primers , Flow Cytometry , Fluorescent Antibody Technique , Gene Transfer Techniques , Genetic Vectors/therapeutic use , Humans , Immunoglobulin Variable Region/immunology , Immunoglobulin Variable Region/metabolism , Lymphocytes/immunology , Neoplasms/genetics , Neoplasms/immunology , Receptor, ErbB-2/immunology , Tetrazolium Salts , Thiazoles , Transduction, Genetic/methods
19.
Urology ; 62(3): 559-65, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12946777

ABSTRACT

OBJECTIVES: The use of recombinant adenoviruses in cancer gene therapy is limited by the widespread expression of the coxsackievirus and adenovirus receptor on normal human cells. Targeting adenoviral vectors to renal cell carcinoma (RCC) cells may improve their potential in cancer gene therapy of patients with metastatic RCC. The G250 protein, also known as the carbonic anhydrase IX protein, is membranously expressed in all cases of clear cell RCC, and clinical studies have demonstrated exceptional tumor targeting with a G250 monoclonal antibody. METHODS: A recombinant bispecific single-chain antibody directed against the RCC-associated G250 protein and the adenovirus fiber knob domain was constructed and used to retarget recombinant adenovirus expressing the green fluorescent protein under control of the cytomegalovirus promoter. G250-specific adenoviral transduction of cells was examined by flow cytometric analysis of green fluorescent protein expression. RESULTS: G250-positive RCC cells displayed enhanced susceptibility for transduction by the green fluorescent protein recombinant adenovirus complexed with the G250-directed bispecific single-chain antibody when compared with native adenovirus. This enhanced transduction was restricted to G250-positive RCC cells and could be abolished completely in the presence of excess G250 protein. CONCLUSIONS: The results of this study demonstrate the feasibility of immunologic retargeting of adenovirus to RCC cells with the highly tumor-specific G250 protein as the target. This strategy may provide the possibility of improving cancer gene therapy for patients with RCC.


Subject(s)
Carcinoma, Renal Cell/therapy , Genetic Therapy/methods , Kidney Neoplasms/therapy , Adenoviridae , Antibodies, Monoclonal/therapeutic use , Antigens, Neoplasm , Antigens, Viral/therapeutic use , Carbonic Anhydrase IX , Carbonic Anhydrases , Carcinoma, Renal Cell/secondary , Carcinoma, Renal Cell/virology , Humans , Immunoglobulin Fragments/therapeutic use , Kidney Neoplasms/virology , Neoplasm Proteins , Recombinant Proteins , Transduction, Genetic , Tumor Cells, Cultured
20.
J Immunol ; 170(4): 2186-94, 2003 Feb 15.
Article in English | MEDLINE | ID: mdl-12574392

ABSTRACT

TCR with known antitumor reactivity can be genetically introduced into primary human T lymphocytes and provide promising tools for immunogene therapy of tumors. We molecularly characterized two distinct TCRs specific for the same HLA-A2-restricted peptide derived from the melanocyte differentiation Ag gp100, yet exhibiting different stringencies in peptide requirements. The existence of these two distinct gp100-specific TCRs allowed us to study the preservation of peptide fine specificity of native TCRalphabeta when engineered for TCR gene transfer into human T lymphocytes. Retroviral transduction of primary human T lymphocytes with either one of the two sets of TCRalphabeta constructs enabled T lymphocytes to specifically kill and produce TNF-alpha when triggered by native gp100(pos)/HLA-A2(pos) tumor target cells as well as gp100 peptide-loaded HLA-A2(pos) tumor cells. Peptide titration studies revealed that the cytolytic efficiencies of the T lymphocyte transductants were in the same range as those of the parental CTL clones. Moreover, primary human T lymphocytes expressing either one of the two engineered gp100-specific TCRs show cytolytic activities in response to a large panel of peptide mutants that are identical with those of the parental CTL. The finding that two gp100-specific TCR, derived from two different CTL, can be functionally introduced into primary human T lymphocytes without loss of the Ag reactivity and peptide fine specificity, holds great promise for the application of TCR gene transfer in cancer immunotherapy.


Subject(s)
Epitopes, T-Lymphocyte/immunology , Membrane Glycoproteins/immunology , Neoplasm Proteins/immunology , Peptide Fragments/immunology , Receptors, Antigen, T-Cell, alpha-beta/genetics , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Cytotoxic/immunology , Transduction, Genetic/methods , Amino Acid Sequence , Amino Acid Substitution/genetics , Amino Acid Substitution/immunology , Base Sequence , Cell Line , Cell Line, Transformed , Cells, Cultured , Clone Cells , Cytotoxicity Tests, Immunologic , Epitopes, T-Lymphocyte/genetics , HLA-A2 Antigen/immunology , Humans , K562 Cells , Melanoma/immunology , Melanoma/therapy , Membrane Glycoproteins/genetics , Molecular Sequence Data , Neoplasm Proteins/genetics , Peptide Fragments/genetics , Receptors, Antigen, T-Cell, alpha-beta/biosynthesis , Receptors, Antigen, T-Cell, alpha-beta/therapeutic use , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Cytotoxic/metabolism , Transfection , Tumor Cells, Cultured , gp100 Melanoma Antigen
SELECTION OF CITATIONS
SEARCH DETAIL
...