Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
2.
Cancer Res Commun ; 2(8): 827-841, 2022 08.
Article in English | MEDLINE | ID: mdl-36923303

ABSTRACT

Chimeric antigen receptor (CAR) T cells are efficacious in patients with B-cell malignancies, while their activity is limited in patients with solid tumors. We developed a novel heterodimeric TCR-like CAR (TCAR) designed to achieve optimal chain pairing and integration into the T-cell CD3 signaling complex. The TCAR mediated high antigen sensitivity and potent antigen-specific T-cell effector functions in short-term in vitro assays. Both persistence and functionality of TCAR T cells were augmented by provision of costimulatory signals, which improved proliferation in vitro and in vivo. Combination with a nanoparticulate RNA vaccine, developed for in vivo expansion of CAR T cells, promoted tightly controlled expansion, survival, and antitumor efficacy of TCAR T cells in vivo. Significance: A novel TCAR is tightly controlled by RNA vaccine-mediated costimulation and may provide an alternative to second-generation CARs for the treatment of solid tumors.


Subject(s)
Immunotherapy, Adoptive , Neoplasms , mRNA Vaccines , Humans , T-Lymphocytes , Receptors, Chimeric Antigen , CD3 Complex , Cell Proliferation , mRNA Vaccines/immunology , Neoplasms/therapy , Cancer Vaccines/therapeutic use , Animals , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Female , Cell Line, Tumor , Xenograft Model Antitumor Assays
3.
J Transl Med ; 19(1): 482, 2021 11 27.
Article in English | MEDLINE | ID: mdl-34838059

ABSTRACT

BACKGROUND: CAR T-cell therapy has been recently unveiled as one of the most promising cancer therapies in hematological malignancies. However, solid tumors mount a profound line of defense to escape immunosurveillance by CAR T-cells. Among them, cytokines with an inhibitory impact on the immune system such as IL-10 and TGFß are of great importance: TGFß is a pleiotropic cytokine, which potently suppresses the immune system and is secreted by a couple of TME resident and tumor cells. METHODS: In this study, we hypothesized that knocking out the TGFß receptor II gene, could improve CAR T-cell functions in vitro and in vivo. Hereby, we used the CRISPR/Cas9 system, to knockout the TGFßRII gene in T-cells and could monitor the efficient gene knock out by genome analysis techniques. Next, Mesothelin or Claudin 6 specific CAR constructs were overexpressed via IVT-RNA electroporation or retroviral transduction and the poly-functionality of these TGFßRII KO CAR T-cells in terms of proliferation, cytokine secretion and cytotoxicity were assessed and compared with parental CAR T-cells. RESULTS: Our experiments demonstrated that TGFßRII KO CAR T-cells fully retained their capabilities in killing tumor antigen positive target cells and more intriguingly, could resist the anti-proliferative effect of exogenous TGFß in vitro outperforming wild type CAR T-cells. Noteworthy, no antigen or growth factor-independent proliferation of these TGFßRII KO CAR T-cells has been recorded. TGFßRII KO CAR T-cells also resisted the suppressive effect of induced regulatory T-cells in vitro to a larger extent. Repetitive antigen stimulation demonstrated that these TGFßRII KO CAR T-cells will experience less activation induced exhaustion in comparison to the WT counterpart. CONCLUSION: The TGFßRII KO approach may become an indispensable tool in immunotherapy of solid tumors, as it may surmount one of the key negative regulatory signaling pathways in T-cells.


Subject(s)
Neoplasms , Receptors, Chimeric Antigen , CRISPR-Cas Systems/genetics , Humans , Immunotherapy, Adoptive , Mesothelin , Neoplasms/genetics , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism
5.
Mol Ther ; 27(1): 261-271, 2019 01 02.
Article in English | MEDLINE | ID: mdl-30528087

ABSTRACT

Genetic engineering of T cells with a T cell receptor (TCR) targeting tumor antigen is a promising strategy for cancer immunotherapy. Inefficient expression of the introduced TCR due to TCR mispairing may limit the efficacy and adversely affect the safety of TCR gene therapy. Here, we evaluated the safety and therapeutic efficiency of an optimized single-chain TCR (scTCR) specific for an HLA-A2.1-restricted (non-mutated) p53(264-272) peptide in adoptive T cell transfer (ACT) models using our unique transgenic mice expressing human p53 and HLA-A2.1 that closely mimic the human setting. Specifically, we showed that adoptive transfer of optimized scTCR-redirected T cells does not induce on-target and off-target autoimmunity. Furthermore, ACT resulted in full tumor protection and led to a long-lived effective, antigen-specific memory T cell response in syngeneic and xenograft models. Taken together, the study demonstrated that our scTCR specific for the broadly expressed tumor-associated antigen p53(264-272) can eradicate p53+A2.1+ tumor cells without inducing off-target or self-directed toxicities in mouse models of ACT. These data strongly support the improved safety and therapeutic efficacy of high-affinity p53scTCR for TCR-based immunotherapy of p53-associated malignancies.


Subject(s)
Autoimmunity/physiology , Receptors, Antigen, T-Cell/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Genetic Therapy , HLA-A2 Antigen/genetics , HLA-A2 Antigen/metabolism , Humans , Mice , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Suppressor Protein p53/genetics
7.
Oncotarget ; 7(16): 21199-221, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-27028870

ABSTRACT

Immunotherapy of cancer envisions the adoptive transfer of T-cells genetically engineered with tumor-specific heterodimeric α/ß T-cell receptors (TCRα/ß). However, potential mispairing of introduced TCRα/ß-chains with endogenous ß/α-ones may evoke unpredictable autoimmune reactivities. A novel single chain (sc)TCR format relies on the fusion of the Vα-Linker-Vß-fragment to the TCR Cß-domain and coexpression of the TCR Cα-domain capable of recruiting the natural CD3-complex for full and hence, native T-cell signaling. Here, we tested whether such a gp100(280-288)- or p53(264-272) tumor antigen-specific scTCR is still prone to mispairing with TCRα. In a human Jurkat-76 T-cell line lacking endogenous TCRs, surface expression and function of a scTCR could be reconstituted by any cointroduced TCRα-chain indicating mispairing to take place on a molecular basis. In contrast, transduction into human TCRα/ß-positive T-cells revealed that mispairing is largely reduced. Competition experiments in Jurkat-76 confirmed the preference of dcTCR to selfpair and to spare scTCR. This also allowed for the generation of dc/scTCR-modified cytomegalovirus/tumor antigen-bispecific T-cells to augment T-cell activation in CMV-infected tumor patients. Residual mispairing was prevented by strenghtening the Vα-Li-Vß-fragment through the design of a novel disulfide bond between a Vα- and a linker-resident residue close to Vß. Multimer-stainings, and cytotoxicity-, IFNγ-secretion-, and CFSE-proliferation-assays, the latter towards dendritic cells endogenously processing RNA-electroporated gp100 antigen proved the absence of hybrid scTCR/TCRα-formation without impairing avidity of scTCR/Cα in T-cells. Moreover, a fragile cytomegalovirus pp65(495-503)-specific scTCR modified this way acquired enhanced cytotoxicity. Thus, optimized scTCR/Cα inhibits residual TCR mispairing to accomplish safe adoptive immunotherapy for bulk endogenous TCRα/ß-positive T-cells.


Subject(s)
CD3 Complex/immunology , Leukemia, T-Cell/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , T-Lymphocytes/immunology , Adoptive Transfer , Animals , Biomarkers, Tumor , CD3 Complex/genetics , Cell Membrane , Cell Proliferation , Humans , Immunotherapy, Adoptive , Leukemia, T-Cell/genetics , Leukemia, T-Cell/pathology , Mice , Receptors, Antigen, T-Cell, alpha-beta/genetics , Signal Transduction , T-Lymphocytes/metabolism , Tumor Cells, Cultured
8.
Cancer Cell ; 24(5): 589-602, 2013 Nov 11.
Article in English | MEDLINE | ID: mdl-24209604

ABSTRACT

Inefficient T cell migration is a major limitation of cancer immunotherapy. Targeted activation of the tumor microenvironment may overcome this barrier. We demonstrate that neoadjuvant local low-dose gamma irradiation (LDI) causes normalization of aberrant vasculature and efficient recruitment of tumor-specific T cells in human pancreatic carcinomas and T-cell-mediated tumor rejection and prolonged survival in otherwise immune refractory spontaneous and xenotransplant mouse tumor models. LDI (local or pre-adoptive-transfer) programs the differentiation of iNOS⁺ M1 macrophages that orchestrate CTL recruitment into and killing within solid tumors through iNOS by inducing endothelial activation and the expression of TH1 chemokines and by suppressing the production of angiogenic, immunosuppressive, and tumor growth factors.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Insulinoma/therapy , Macrophages/physiology , Nitric Oxide Synthase Type II/metabolism , Pancreatic Neoplasms/therapy , Animals , CD4-Positive T-Lymphocytes/transplantation , CD8-Positive T-Lymphocytes/transplantation , Cell Differentiation/radiation effects , Cells, Cultured , Female , Humans , Immunotherapy, Adoptive , Inflammation Mediators/metabolism , Insulinoma/blood supply , Insulinoma/immunology , Macrophages/radiation effects , Melanoma/immunology , Melanoma/therapy , Mice , Mice, Inbred C3H , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Neoplasm Transplantation , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/immunology , Phenotype , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Radiotherapy Dosage , Radiotherapy, Adjuvant , Tumor Escape , Vaccination
9.
PLoS One ; 8(5): e63521, 2013.
Article in English | MEDLINE | ID: mdl-23717444

ABSTRACT

Tumor-growth is often associated with the expansion of myeloid derived suppressor cells that lead to local or systemic arginine depletion via the enzyme arginase. It is generally assumed that this arginine deficiency induces a global shut-down of T cell activation with ensuing tumor immune escape. While the impact of arginine depletion on polyclonal T cell proliferation and cytokine secretion is well documented, its influence on chemotaxis, cytotoxicity and antigen specific activation of human T cells has not been demonstrated so far. We show here that chemotaxis and early calcium signaling of human T cells are unimpaired in the absence of arginine. We then analyzed CD8(+) T cell activation in a tumor peptide as well as a viral peptide antigen specific system: (i) CD8(+) T cells with specificity against the MART-1aa26-35*A27L tumor antigen expanded with in vitro generated dendritic cells, and (ii) clonal CMV pp65aa495-503 specific T cells and T cells retrovirally transduced with a CMV pp65aa495-503 specific T cell receptor were analyzed. Our data demonstrate that human CD8(+) T cell antigen specific cytotoxicity and perforin secretion are completely preserved in the absence of arginine, while antigen specific proliferation as well as IFN-γ and granzyme B secretion are severely compromised. These novel results highlight the complexity of antigen specific T cell activation and demonstrate that human T cells can preserve important activation-induced effector functions in the context of arginine deficiency.


Subject(s)
Arginine/deficiency , CD8-Positive T-Lymphocytes/immunology , MART-1 Antigen/immunology , T-Lymphocytes, Cytotoxic/immunology , CD8-Positive T-Lymphocytes/metabolism , Calcium Signaling , Cell Proliferation , Cells, Cultured , Chemotaxis , Cytotoxicity, Immunologic , Granzymes/metabolism , Humans , Interferon-gamma/metabolism , Lymphocyte Activation , Perforin/metabolism , Tumor Escape
10.
Blood ; 115(25): 5154-63, 2010 Jun 24.
Article in English | MEDLINE | ID: mdl-20378753

ABSTRACT

Transfer of tumor antigen-specific T-cell receptors (TCRs) into human T cells aims at redirecting their cytotoxicity toward tumors. Efficacy and safety may be affected by pairing of natural and introduced TCRalpha/beta chains potentially leading to autoimmunity. We hypothesized that a novel single-chain (sc)TCR framework relying on the coexpression of the TCRalpha constant alpha (Calpha) domain would prevent undesired pairing while preserving structural and functional similarity to a fully assembled double-chain (dc)TCR/CD3 complex. We confirmed this hypothesis for a murine p53-specific scTCR. Substantial effector function was observed only in the presence of a murine Calpha domain preceded by a TCRalpha signal peptide for shuttling to the cell membrane. The generalization to a human gp100-specific TCR required the murinization of both C domains. Structural and functional T-cell avidities of an accessory disulfide-linked scTCR gp100/Calpha were higher than those of a dcTCR. Antigen-dependent phosphorylation of the proximal effector zeta-chain-associated protein kinase 70 at tyrosine 319 was not impaired, reflecting its molecular integrity in signaling. In melanoma-engrafted nonobese diabetic/severe combined immunodeficient mice, adoptive transfer of scTCR gp100/Calpha transduced T cells conferred superior delay in tumor growth among primary and long-term secondary tumor challenges. We conclude that the novel scTCR constitutes a reliable means to immunotherapeutically target hematologic malignancies.


Subject(s)
Adoptive Transfer , Antigens, Neoplasm/immunology , Melanoma/therapy , Receptors, Antigen, T-Cell, alpha-beta/immunology , T-Lymphocytes/immunology , Tumor Suppressor Protein p53/immunology , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Cell Line, Tumor , Humans , Immunity, Cellular , Melanoma/genetics , Melanoma/immunology , Melanoma/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Phosphorylation/genetics , Protein Structure, Tertiary , Receptors, Antigen, T-Cell, alpha-beta/biosynthesis , Receptors, Antigen, T-Cell, alpha-beta/genetics , T-Lymphocytes/metabolism , T-Lymphocytes/transplantation , Transduction, Genetic , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
11.
Haematologica ; 95(1): 126-34, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19679884

ABSTRACT

BACKGROUND: The Wilms' tumor antigen (WT1) is an attractive target for immunotherapy of leukemia. In the past, we isolated and characterized the specificity and function of a WT1-specific T-cell receptor. The goal of this translational study was to develop a safe and efficient WT1-T-cell receptor retroviral vector for an adoptive immunotherapy trial with engineered T cells. DESIGN AND METHODS: We generated a panel of retroviral constructs containing unmodified or codon-optimized WT1-T-cell receptor alpha and beta genes, linked via internal ribosome entry sites or 2A sequences, with or without an additional inter-chain disulfide bond in the T-cell receptor constant domains. These constructs were functionally analyzed in vitro, and the best one was tested in an autologous primary leukemia model in vivo. RESULTS: We identified a WT1-T-cell receptor construct that showed optimal tetramer staining, antigen-specific cytokine production and killing activity when introduced into primary human T cells. Fresh CD34(+) cells purified from a patient with leukemia were engrafted into NOD/SCID mice, followed by adoptive immunotherapy with patient's autologous T cells transduced with the WT1-T-cell receptor. This therapeutic treatment evidently decreased leukemia engraftment in mice and resulted in a substantial improvement of leukemia-free survival. CONCLUSIONS: This is the first report that patient's T cells, engineered to express the WT1-T-cell receptor, can eliminate autologous leukemia progenitor cells in an in vivo model. This study provides a firm basis for the planned WT1-T-cell receptor gene therapy trial in leukemia patients.


Subject(s)
Antigens, Neoplasm/immunology , Blast Crisis/immunology , Genetic Engineering/methods , Leukemia/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/therapeutic use , T-Lymphocytes/pathology , Wilms Tumor/pathology , Adult , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Blast Crisis/genetics , Blast Crisis/therapy , Genetic Therapy/methods , Genetic Vectors/biosynthesis , Genetic Vectors/chemistry , Hepatitis B Virus, Woodchuck/genetics , Humans , Jurkat Cells , Leukemia/genetics , Leukemia/therapy , Mice , Mice, Inbred NOD , Mice, SCID , Receptors, Antigen, T-Cell/physiology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transplantation, Autologous/immunology , Wilms Tumor/immunology , Wilms Tumor/therapy
12.
J Exp Med ; 206(2): 463-75, 2009 Feb 16.
Article in English | MEDLINE | ID: mdl-19171765

ABSTRACT

Adoptive transfer of T lymphocytes transduced with a T cell receptor (TCR) to impart tumor reactivity has been reported as a potential strategy to redirect immune responses to target cancer cells (Schumacher, T.N. 2002. Nat. Rev. Immunol. 2:512-519). However, the affinity of most TCRs specific for shared tumor antigens that can be isolated is usually low. Thus, strategies to increase the affinity of TCRs or the functional avidity of TCR-transduced T cells might be therapeutically beneficial. Because glycosylation affects the flexibility, movement, and interactions of surface molecules, we tested if selectively removing conserved N-glycoslyation sites in the constant regions of TCR alpha or beta chains could increase the functional avidity of T cells transduced with such modified TCRs. We observed enhanced functional avidity and improved recognition of tumor cells by T cells harboring TCR chains with reduced N-glycosylation (DeltaTCR) as compared with T cells with wild-type (WT) TCR chains. T cells transduced with WT or DeltaTCR chains bound tetramer equivalently at 4 degrees C, but tetramer binding was enhanced at 37 degrees C, predominantly as a result of reduced tetramer dissociation. This suggested a temperature-dependent mechanism such as TCR movement in the cell surface or structural changes of the TCR allowing improved multimerization. This strategy was effective with mouse and human TCRs specific for different antigens and, thus, should be readily translated to TCRs with any specificity.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Models, Molecular , Neoplasms/immunology , Receptors, Antigen, T-Cell/metabolism , Adoptive Transfer , Animals , CD4-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Glycosylation , Humans , Mice , Protein Binding/immunology , Receptors, Antigen, T-Cell/genetics , Reverse Transcriptase Polymerase Chain Reaction , Temperature
13.
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
14.
Mol Med ; 13(3-4): 216-26, 2007.
Article in English | MEDLINE | ID: mdl-17592557

ABSTRACT

Many strategies for redirecting the tropism of murine Moloney leukemia virus (MMLV) have been described. Preformed virion-liposome complexes, termed virosomes, have been reported to be relatively stable. Virosomes mediate envelope-independent transduction that allows efficient superinfection of resistant cell lines; however, virosome-mediated transduction behaves in a non-target-specific manner. We developed a novel method using antibodies to direct MMLV to vascular endothelium. We have given the term immunovirosomes to the complexes formed between viruses, liposomes, and antibodies. These immunovirosomes improve the transduction efficiency of the viruses and alter their tropism. We have shown improved transduction when immunovirosomes were targeted at the endocytic receptors CD71 and CD62E/P and rather less good delivery when targeted at CD106. The enhancement of the transduction efficiency was transient, however, suggesting that rerouting the entry pathway of viruses alters the expression properties of the viruses.


Subject(s)
Gene Expression , Gene Transfer Techniques , Liposomes , Tropism , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Antigens, CD/immunology , Antigens, CD/metabolism , E-Selectin/immunology , E-Selectin/metabolism , Genes, Viral , Humans , Hybridomas , Moloney murine leukemia virus/genetics , Moloney murine leukemia virus/metabolism , Receptors, Transferrin/immunology , Receptors, Transferrin/metabolism , Retroviridae/genetics , Transduction, Genetic , Vascular Cell Adhesion Molecule-1/immunology , Vascular Cell Adhesion Molecule-1/metabolism , Virosomes/metabolism
15.
Blood ; 109(6): 2331-8, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-17082316

ABSTRACT

Adoptive transfer of T lymphocytes is a promising treatment for a variety of malignancies but often not feasible due to difficulties generating T cells that are reactive with the targeted antigen from patients. To facilitate rapid generation of cells for therapy, T cells can be programmed with genes encoding the alpha and beta chains of an antigen-specific T-cell receptor (TCR). However, such exogenous alpha and beta chains can potentially assemble as pairs not only with each other but also with endogenous TCR alpha and beta chains, thereby generating alphabetaTCR pairs of unknown specificity as well as reducing the number of exogenous matched alphabetaTCR pairs at the cell surface. We demonstrate that introducing cysteines into the constant region of the alpha and beta chains can promote preferential pairing with each other, increase total surface expression of the introduced TCR chains, and reduce mismatching with endogenous TCR chains. This approach should improve both the efficacy and safety of ongoing efforts to use TCR transfer as a strategy to generate tumor-reactive T cells.


Subject(s)
Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/metabolism , Cell Line , Cysteine/genetics , Cysteine/metabolism , Humans , Protein Biosynthesis/genetics , Transcription, Genetic/genetics
16.
Immunol Res ; 34(1): 67-87, 2006.
Article in English | MEDLINE | ID: mdl-16720899

ABSTRACT

Retroviral transfer of T cell antigen receptor (TCR) genes selected by circumventing tolerance to broad tumor- and leukemia-associated antigens in human leukocyte antigen (HLA)-A*0201 (A2.1) transgenic (Tg) mice allows the therapeutic reprogramming of human T lymphocytes. Using a human CD8 x A2.1/Kb mouse derived TCR specific for natural peptide-A2.1 (pA2.1) complexes comprising residues 81-88 of the human homolog of the murine double-minute 2 oncoprotein, MDM2(81-88), we found that the heterodimeric CD8 alpha beta coreceptor, but not normally expressed homodimeric CD8 alpha alpha, is required for tetramer binding and functional redirection of TCR- transduced human T cells. CD8+T cells that received a humanized derivative of the MDM2 TCR bound pA2.1 tetramers only in the presence of an anti-human-CD8 anti-body and required more peptide than wild-type (WT) MDM2 TCR+T cells to mount equivalent cytotoxicity. They were, however, sufficiently effective in recognizing malignant targets including fresh leukemia cells. Most efficient expression of transduced TCR in human T lymphocytes was governed by mouse as compared to human constant (C) alphabeta domains, as demonstrated with partially humanized and murinized TCR of primary mouse and human origin, respectively. We further observed a reciprocal relationship between the level of Tg WT mouse relative to natural human TCR expression, resulting in T cells with decreased normal human cell surface TCR. In contrast, natural human TCR display remained unaffected after delivery of the humanized MDM2 TCR. These results provide important insights into the molecular basis of TCR gene therapy of malignant disease.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Proto-Oncogene Proteins c-mdm2/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , Animals , Cell Line, Tumor , Flow Cytometry , HLA-A2 Antigen/immunology , Humans , Mice , Mice, Transgenic , Proto-Oncogene Proteins c-mdm2/genetics , Reverse Transcriptase Polymerase Chain Reaction , Self Tolerance/immunology
17.
Immunity ; 22(1): 117-29, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15664164

ABSTRACT

Efficient immune attack of malignant disease requires the concerted action of both CD8+ CTL and CD4+ Th cells. We used human leukocyte antigen (HLA)-A*0201 (A2.1) transgenic mice, in which the mouse CD8 molecule cannot efficiently interact with the alpha3 domain of A2.1, to generate a high-affinity, CD8-independent T cell receptor (TCR) specific for a commonly expressed, tumor-associated cytotoxic T lymphocyte (CTL) epitope derived from the human p53 tumor suppressor protein. Retroviral expression of this CD8-independent, p53-specific TCR into human T cells imparted the CD8+ T lymphocytes with broad tumor-specific CTL activity and turned CD4+ T cells into potent tumor-reactive, p53A2.1-specific Th cells. Both T cell subsets were cooperative and interacted synergistically with dendritic cell intermediates and tumor targets. The intentional redirection of both CD4+ Th cells and CD8+ CTL by the same high-affinity, CD8-independent, tumor-specific TCR could provide the basis for novel broad-spectrum cancer immunotherapeutics.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Receptors, Antigen, T-Cell/metabolism , T-Cell Antigen Receptor Specificity , Animals , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cloning, Molecular , Flow Cytometry , Humans , Mice , Mice, Transgenic , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Cytotoxic/immunology , Transduction, Genetic , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/immunology , Tumor Suppressor Protein p53/metabolism
18.
Methods Mol Med ; 109: 229-56, 2005.
Article in English | MEDLINE | ID: mdl-15585925

ABSTRACT

Reprogramming T-cell populations by T-cell receptor (TCR) gene transfer is a new therapeutic tool for adoptive tumor immunotherapy. Gene transfer of human leukocyte antigen (HLA)-transgenic mice-derived TCR into human T-cells allows the circumvention of tolerance to tumor-associated (self) antigens (TAA). This chapter reports on the identification of the alpha and beta chains of the heterodimeric TCR derived from a mouse T-cell clone. The related DNA fragments are inserted into a retroviral vector for heterologous expression of the TAA-specific TCR in human T-cells. Polymerase chain reaction (PCR)-based cloning protocols are provided for the tailor-made customization of murine TCR. We describe the humanization and chimerization of such TCR as well as their expression in human T-cells.


Subject(s)
Immunotherapy/methods , Neoplasms/immunology , Neoplasms/therapy , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cells, Cultured , Cloning, Molecular , Gene Expression Regulation , Humans , Mice , Mice, Transgenic , Molecular Sequence Data , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/genetics , Reverse Transcriptase Polymerase Chain Reaction , Substrate Specificity , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...