Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
2.
Nat Med ; 21(1): 81-5, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25531942

ABSTRACT

Tumor-specific neo-antigens that arise as a consequence of mutations are thought to be important for the therapeutic efficacy of cancer immunotherapies. Accumulating evidence suggests that neo-antigens may be commonly recognized by intratumoral CD8+ T cells, but it is unclear whether neo-antigen-specific CD4+ T cells also frequently reside within human tumors. In view of the accepted role of tumor-specific CD4+ T-cell responses in tumor control, we addressed whether neo-antigen-specific CD4+ T-cell reactivity is a common property in human melanoma.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Immunotherapy , Melanoma/immunology , Antigen Presentation/immunology , Antigens, Neoplasm/immunology , Cell Line, Tumor , DNA-Binding Proteins/genetics , Epitopes, T-Lymphocyte/genetics , Humans , Melanoma/genetics , Melanoma/pathology , Mutation , Proto-Oncogene Proteins c-bcl-6 , bcl-X Protein/genetics
3.
J Immunol ; 193(10): 4803-13, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25311806

ABSTRACT

Virus or tumor Ag-derived peptides that are displayed by MHC class I molecules are attractive starting points for vaccine development because they induce strong protective and therapeutic cytotoxic T cell responses. In thus study, we show that the MHC binding and consequent T cell reactivity against several HLA-A*02 restricted epitopes can be further improved through the incorporation of nonproteogenic amino acids at primary and secondary anchor positions. We screened more than 90 nonproteogenic, synthetic amino acids through a range of epitopes and tested more than 3000 chemically enhanced altered peptide ligands (CPLs) for binding affinity to HLA-A*0201. With this approach, we designed CPLs of viral epitopes, of melanoma-associated Ags, and of the minor histocompatibility Ag UTA2-1, which is currently being evaluated for its antileukemic activity in clinical dendritic cell vaccination trials. The crystal structure of one of the CPLs in complex with HLA-A*0201 revealed the molecular interactions likely responsible for improved binding. The best CPLs displayed enhanced affinity for MHC, increasing MHC stability and prolonging recognition by Ag-specific T cells and, most importantly, they induced accelerated expansion of antitumor T cell frequencies in vitro and in vivo as compared with the native epitope. Eventually, we were able to construct a toolbox of preferred nonproteogenic residues with which practically any given HLA-A*02 restricted epitope can be readily optimized. These CPLs could improve the therapeutic outcome of vaccination strategies or can be used for ex vivo enrichment and faster expansion of Ag-specific T cells for transfer into patients.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , HLA-A2 Antigen/immunology , Neoplasms/prevention & control , Peptides/immunology , Amino Acid Sequence , Animals , Antigens, Neoplasm/chemistry , Antigens, Neoplasm/genetics , B-Lymphocytes , Cancer Vaccines/administration & dosage , Cancer Vaccines/chemistry , Crystallography, X-Ray , Epitopes , Gene Expression , HLA-A2 Antigen/chemistry , HLA-A2 Antigen/genetics , Humans , Immunization , Mice , Mice, Transgenic , Minor Histocompatibility Antigens/chemistry , Minor Histocompatibility Antigens/genetics , Minor Histocompatibility Antigens/immunology , Models, Molecular , Molecular Sequence Data , Neoplasms/immunology , Peptides/administration & dosage , Peptides/chemistry , Peptides/genetics , Protein Binding , Recombinant Proteins/genetics , Recombinant Proteins/immunology , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism
4.
Mol Ther ; 22(11): 1983-91, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25048215

ABSTRACT

Genetically modified T cells that express a transduced T cell receptor (TCR) α/ß heterodimer in addition to their endogenous TCR are used in clinical studies to treat cancer. These cells express two TCR-α and two TCR-ß chains that do not only compete for CD3 proteins but also form potentially self-reactive mixed TCR dimers, composed of endogenous and transferred chains. To overcome these deficits, we developed an RNAi-TCR replacement vector that simultaneously silences the endogenous TCR and expresses an RNAi-resistant TCR. Transduction of the virus-specific P14 TCR without RNAi resulted in unequal P14 TCR-α and -ß chain surface levels, indicating heterodimerization with endogenous TCR chains. Such unequal expression was also observed following TCR gene optimization. Equal surface levels of the introduced TCR chains were however achieved by silencing the endogenous TCR. Importantly, all mice that received cells transduced with the native or optimized P14 TCR developed lethal TCR gene transfer-induced graft-versus-host-disease (TI-GVHD) due to formation of mixed TCR dimers. In contrast, TI-GVHD was almost completely prevented when using the RNAi-TCR replacement vector. Our data demonstrate that RNAi-assisted TCR replacement reduces the formation of mixed TCR dimers, and thereby significantly reduces the risk of TI-GVHD in TCR gene therapy.


Subject(s)
Graft vs Host Disease/prevention & control , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Melanoma, Experimental/therapy , Receptors, Antigen, T-Cell/antagonists & inhibitors , T-Lymphocytes/immunology , Animals , Autoimmunity , Cells, Cultured , Disease Models, Animal , Gene Knockdown Techniques , Genetic Vectors/administration & dosage , Lung Neoplasms/immunology , Melanoma, Experimental/immunology , Mice , RNA Interference , Receptors, Antigen, T-Cell/genetics , Transduction, Genetic
5.
J Immunol ; 191(6): 3232-9, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23940272

ABSTRACT

TCR gene therapy is a promising approach for the treatment of various human malignancies. However, the tumoricidal activity of TCR-modified T cells may be limited by local immunosuppressive mechanisms within the tumor environment. In particular, many malignancies induce T cell suppression in their microenvironment by TGF-ß secretion. In this study, we evaluate whether blockade of TGF-ß signaling in TCR-modified T cells enhances TCR gene therapy efficacy in an autochthonous mouse tumor model. Treatment of mice with advanced prostate cancer with T cells genetically engineered to express a tumor-reactive TCR and a dominant-negative TGF-ß receptor II induces complete and sustained tumor regression, enhances survival, and leads to restored differentiation of prostate epithelium. These data demonstrate the potential to tailor the activity of TCR-modified T cells by additional genetic modification and provide a strong rationale for the clinical testing of TGF-ß signaling blockade to enhance TCR gene therapy against advanced cancers.


Subject(s)
Genetic Therapy/methods , Immunotherapy, Adoptive/methods , Neoplasms, Experimental/therapy , Prostatic Neoplasms/immunology , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/immunology , Transforming Growth Factor beta/antagonists & inhibitors , Animals , Flow Cytometry , Immunohistochemistry , Male , Mice , Mice, Transgenic , Neoplasms, Experimental/genetics , Neoplasms, Experimental/immunology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/therapy , Receptors, Antigen, T-Cell/immunology , Signal Transduction/immunology , T-Lymphocytes/transplantation , Transduction, Genetic
6.
Nat Med ; 16(5): 565-70, 1p following 570, 2010 May.
Article in English | MEDLINE | ID: mdl-20400962

ABSTRACT

The transfer of T cell receptor (TCR) genes can be used to induce immune reactivity toward defined antigens to which endogenous T cells are insufficiently reactive. This approach, which is called TCR gene therapy, is being developed to target tumors and pathogens, and its clinical testing has commenced in patients with cancer. In this study we show that lethal cytokine-driven autoimmune pathology can occur in mouse models of TCR gene therapy under conditions that closely mimic the clinical setting. We show that the pairing of introduced and endogenous TCR chains in TCR gene-modified T cells leads to the formation of self-reactive TCRs that are responsible for the observed autoimmunity. Furthermore, we demonstrate that adjustments in the design of gene therapy vectors and target T cell populations can be used to reduce the risk of TCR gene therapy-induced autoimmune pathology.


Subject(s)
Genes, T-Cell Receptor , Genetic Therapy/methods , Graft vs Host Disease/pathology , Neoplasms/therapy , Receptors, Antigen, T-Cell/genetics , Animals , Graft vs Host Disease/immunology , Humans , Mice , Mice, Inbred C57BL , Models, Animal , Neoplasms/genetics , Neoplasms/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...