Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Mol Ther ; 26(11): 2553-2566, 2018 11 07.
Article in English | MEDLINE | ID: mdl-30217730

ABSTRACT

Checkpoint inhibitors and adoptive cell therapy provide promising options for treating solid cancers such as HBV-related HCC, but they have limitations. We tested the potential to combine advantages of each approach, genetically reprogramming T cells specific for viral tumor antigens to overcome exhaustion by down-modulating the co-inhibitory receptor PD-1. We developed a novel lentiviral transduction protocol to achieve preferential targeting of endogenous or TCR-redirected, antigen-specific CD8 T cells for shRNA knockdown of PD-1 and tested functional consequences for antitumor immunity. Antigen-specific and intrahepatic CD8 T cells transduced with lentiviral (LV)-shPD-1 consistently had a marked reduction in PD-1 compared to those transduced with a control lentiviral vector. PD-1 knockdown of human T cells rescued antitumor effector function and promoted killing of hepatoma cells in a 3D microdevice recapitulating the pro-inflammatory PD-L1hi liver microenvironment. However, upon repetitive stimulation, PD-1 knockdown drove T cell senescence and induction of other co-inhibitory pathways. We provide the proof of principle that T cells with endogenous or genetically engineered specificity for HBV-associated HCC viral antigens can be targeted for functional genetic editing. We show that PD-1 knockdown enhances immediate tumor killing but is limited by compensatory engagement of alternative co-inhibitory and senescence program upon repetitive stimulation.


Subject(s)
Carcinoma, Hepatocellular/therapy , Hepatitis B, Chronic/therapy , Liver Neoplasms/therapy , Programmed Cell Death 1 Receptor/immunology , Receptors, Antigen, T-Cell/therapeutic use , Antigens, Neoplasm/immunology , Antigens, Neoplasm/therapeutic use , Antigens, Viral/immunology , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/virology , Genetic Vectors/genetics , Hepatitis B virus/immunology , Hepatitis B virus/pathogenicity , Hepatitis B, Chronic/immunology , Hepatitis B, Chronic/pathology , Hepatitis B, Chronic/virology , Humans , Immunotherapy, Adoptive/methods , Lentivirus/genetics , Liver/immunology , Liver/metabolism , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Liver Neoplasms/virology , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/therapeutic use , Receptors, Antigen, T-Cell/immunology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
2.
J Immunol ; 194(3): 1080-9, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25539815

ABSTRACT

Ag receptors used for cancer immunotherapy are often directed against tumor-associated Ags also expressed in normal tissues. Targeting of such Ags can result in unwanted autoimmune attack of normal tissues or induction of tolerance in therapeutic T cells. We used a murine model to study the phenotype and function of T cells redirected against the murine double minute protein 2 (MDM2), a tumor-associated Ag that shows low expression in many normal tissues. Transfer of MDM2-TCR-engineered T cells into bone marrow chimeric mice revealed that Ag recognition in hematopoietic tissues maintained T cell function, whereas presentation of MDM2 in nonhematopoietic tissues caused reduced effector function. TCR-engineered CD8(+) T cells underwent rapid turnover, downmodulated CD8 expression, and lost cytotoxic function. We found that MDM2-TCR-engineered CD4(+) T cells provided help and restored cytotoxic function of CD8(+) T cells bearing the same TCR. Although the introduction of the CD8 coreceptor enhanced the ability of CD4(+) T cells to recognize MDM2 in vitro, the improved self-antigen recognition abolished their ability to provide helper function in vivo. The data indicate that the same class I-restricted TCR responsible for Ag recognition and tolerance induction in CD8(+) T cells can, in the absence of the CD8 coreceptor, elicit CD4 T cell help and partially reverse tolerance. Thus MHC class I-restricted CD4(+) T cells may enhance the efficacy of therapeutic TCR-engineered CD8(+) T cells and can be readily generated with the same TCR.


Subject(s)
Antigens, Neoplasm/immunology , Autoantigens/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Immune Tolerance , Receptors, Antigen, T-Cell/genetics , Adoptive Transfer , Animals , Cell Communication , Cytotoxicity, Immunologic , Gene Expression , Immunophenotyping , Mice , Mice, Transgenic , Phenotype , Proto-Oncogene Proteins c-mdm2/genetics , Receptors, Antigen, T-Cell/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transduction, Genetic
3.
Haematologica ; 101(4): 482-90, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26802053

ABSTRACT

Due to the lack of specificity for tumor antigens, allogeneic T-cell therapy is associated with graft-versus-host disease. Enhancing the anti-tumor specificity while reducing the graft-versus-host disease risk of allogeneic T cells has remained a research focus. In this study, we demonstrate that the introduction of 'dominant' T-cell receptors into primary murine T cells can suppress the expression of endogenous T-cell receptors in a large proportion of the gene-modified T cells. Adoptive transfer of allogeneic T cells expressing a 'dominant' T-cell receptor significantly reduced the graft-versus-host toxicity in recipient mice. Using two bone marrow transplant models, enhanced anti-tumor activity was observed in the presence of reduced graft-versus-host disease. However, although transfer of T-cell receptor gene-modified allogeneic T cells resulted in the elimination of antigen-positive tumor cells and improved the survival of treated mice, it was associated with accumulation of T cells expressing endogenous T-cell receptors and the development of delayed graft-versus-host disease. The in-vivo deletion of the engineered T cells, mediated by endogenous mouse mammary tumor virus MTV8 and MTV9, abolished graft-versus-host disease while retaining significant anti-tumor activity of adoptively transferred T cells. Together, this study shows that the in-vitro selection of allogeneic T cells expressing high levels of a 'dominant' T-cell receptor can lower acute graft-versus-host disease and enhance anti-tumor activity of adoptive cell therapy, while the in-vivo outgrowth of T cells expressing endogenous T-cell receptors remains a risk factor for the delayed onset of graft-versus-host disease.


Subject(s)
Graft vs Host Disease/prevention & control , Immunotherapy, Adoptive/methods , Mammary Tumor Virus, Mouse/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/transplantation , Animals , Bone Marrow Transplantation/methods , Cell Line, Tumor , Female , Gene Expression , Genes, Dominant , Genetic Vectors/immunology , Graft vs Host Disease/genetics , Graft vs Host Disease/immunology , Graft vs Host Disease/pathology , Humans , Lymphocyte Depletion/methods , Mammary Tumor Virus, Mouse/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Receptors, Antigen, T-Cell/genetics , Survival Analysis , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Transgenes , Transplantation, Homologous , Whole-Body Irradiation
4.
J Hepatol ; 62(2): 486-91, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25308176

ABSTRACT

HBV-DNA integration frequently occurs in HBV-related hepatocellular carcinoma (HCC), but whether HBV antigens are expressed in HCC cells and can be targeted by immune therapeutic strategies remains controversial. Here, we first characterized HBV antigen expression in HCC metastases, occurring in a patient who had undergone liver transplantation for HBV-related HCC. We then deployed for the first time in HCC autologous T cells, genetically modified to express an HBsAg specific T cell receptor, as therapy against chemoresistant extrahepatic metastases. We confirmed that HBV antigens were expressed in HCC metastases (but not in the donor liver) and demonstrated that tumour cells were recognized in vivo by lymphocytes, engineered to express an HBV-specific T cell receptor (TCR). Gene-modified T cells survived, expanded and mediated a reduction in HBsAg levels without exacerbation of liver inflammation or other toxicity. Whilst clinical efficacy was not established in this subject with end-stage metastatic disease, we confirm the feasibility of providing autologous TCR-redirected therapy against HCC and advocate this strategy as a novel therapeutic opportunity in hepatitis B-associated malignancies.


Subject(s)
Carcinoma, Hepatocellular/secondary , Hepatitis B Surface Antigens/metabolism , Immunotherapy/methods , Liver Neoplasms/pathology , Receptors, Antigen, T-Cell/therapeutic use , Tacrolimus/therapeutic use , Aged , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/therapy , Fatal Outcome , Follow-Up Studies , Humans , Immunosuppressive Agents/therapeutic use , Liver Neoplasms/immunology , Liver Neoplasms/therapy , Male , Neoplasm Metastasis
5.
Blood ; 118(2): 319-29, 2011 Jul 14.
Article in English | MEDLINE | ID: mdl-21606483

ABSTRACT

We have tested whether affinity-matured TCRs that retain peptide specificity improve the ability of primary human CD8(+) T cells to mount antigen-specific responses. We found that TCR affinity correlated with the speed of T-cell responses. High affinity TCR-antigen interactions rapidly initiated T-cell responses, but low affinity TCR/antigen interactions required longer time periods to elicit the same responses. Within the "natural" affinity range, increased TCR-to-antigen affinity correlated with improved ability of T cells to recognize low concentration of antigen. However, affinity-matured TCR with 700-fold enhanced affinity for MHC-to-antigen required 100-fold higher antigen-density to initiate T-cell responses than did wild-type TCR. Using modified peptides to reduce the affinity of TCR-to-antigen interaction, we demonstrate that affinity-matured TCRs are not defective, being superior to wild-type TCR in recognizing low concentration of modified peptides. These data indicate that enhancing TCR affinity can accelerate the speed of T-cell activation and reduce the ability to recognize low density of MHC-to-peptide antigen. We predict that future studies of the human T-cell repertoire will reveal 2 types of low avidity T cells: fast and slow responders, with high-affinity and low-affinity TCR, respectively.


Subject(s)
Histocompatibility Antigens/immunology , Major Histocompatibility Complex/immunology , Peptides/immunology , Receptors, Antigen, T-Cell/metabolism , T-Cell Antigen Receptor Specificity/physiology , T-Lymphocytes/metabolism , Cells, Cultured , HLA-A Antigens/metabolism , HLA-A2 Antigen , HeLa Cells , Histocompatibility Antigens/metabolism , Humans , Jurkat Cells , Kinetics , Peptides/metabolism , Protein Binding/physiology , Receptors, Antigen, T-Cell/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , Time Factors
6.
Blood ; 118(13): 3528-37, 2011 Sep 29.
Article in English | MEDLINE | ID: mdl-21750319

ABSTRACT

The function of T-cell receptor (TCR) gene modified T cells is dependent on efficient surface expression of the introduced TCR α/ß heterodimer. We tested whether endogenous CD3 chains are rate-limiting for TCR expression and antigen-specific T-cell function. We show that co-transfer of CD3 and TCR genes into primary murine T cells enhanced TCR expression and antigen-specific T-cell function in vitro. Peptide titration experiments showed that T cells expressing introduced CD3 and TCR genes recognized lower concentration of antigen than T cells expressing TCR only. In vivo imaging revealed that TCR+CD3 gene modified T cells infiltrated tumors faster and in larger numbers, which resulted in more rapid tumor elimination compared with T cells modified by TCR only. After tumor clearance, TCR+CD3 engineered T cells persisted in larger numbers than TCR-only T cells and mounted a more effective memory response when rechallenged with antigen. The data demonstrate that provision of additional CD3 molecules is an effective strategy to enhance the avidity, anti-tumor activity and functional memory formation of TCR gene modified T cells in vivo.


Subject(s)
CD3 Complex/physiology , Genes, T-Cell Receptor/genetics , Genetic Therapy , Animals , CD3 Complex/genetics , CD3 Complex/metabolism , Cells, Cultured , Down-Regulation , Female , Genetic Therapy/methods , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Lymphoma, T-Cell/genetics , Lymphoma, T-Cell/pathology , Lymphoma, T-Cell/therapy , Mice , Mice, Inbred C57BL , Mice, Transgenic , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Treatment Outcome
7.
Blood ; 117(25): 6813-24, 2011 Jun 23.
Article in English | MEDLINE | ID: mdl-21447831

ABSTRACT

Recently, vaccines against the Wilms Tumor antigen 1 (WT1) have been tested in cancer patients. However, it is currently not known whether physiologic levels of WT1 expression in stem and progenitor cells of normal tissue result in the deletion or tolerance induction of WT1-specific T cells. Here, we used an human leukocyte antigen-transgenic murine model to study the fate of human leukocyte antigen class-I restricted, WT1-specific T cells in the thymus and in the periphery. Thymocytes expressing a WT1-specific T-cell receptor derived from high avidity human CD8 T cells were positively selected into the single-positive CD8 population. In the periphery, T cells specific for the WT1 antigen differentiated into CD44-high memory phenotype cells, whereas T cells specific for a non-self-viral antigen retained a CD44(low) naive phenotype. Only the WT1-specific T cells, but not the virus-specific T cells, displayed rapid antigen-specific effector function without prior vaccination. Despite long-term persistence of WT1-specific memory T cells, the animals did not develop autoimmunity, and the function of hematopoietic stem and progenitor cells was unimpaired. This is the first demonstration that specificity for a tumor-associated self-antigen may drive differentiation of functionally competent memory T cells.


Subject(s)
T-Lymphocytes/cytology , T-Lymphocytes/immunology , Thymus Gland/immunology , WT1 Proteins/immunology , Animals , Cell Line , Gene Expression , Gene Transfer Techniques , Humans , Immunologic Memory , Mice , Mice, Inbred C57BL , Mice, Transgenic , Stem Cells/immunology , Stem Cells/metabolism , Vaccination , WT1 Proteins/genetics , Wilms Tumor/immunology
8.
Proc Natl Acad Sci U S A ; 106(45): 19078-83, 2009 Nov 10.
Article in English | MEDLINE | ID: mdl-19884493

ABSTRACT

Regulatory T cells (Tregs) can suppress a wide range of immune cells, making them an ideal candidate for the treatment of autoimmunity. The potential clinical translation of targeted therapy with antigen-specific Tregs is hampered by the difficulties of isolating rare specificities from the natural polyclonal T cell repertoire. Moreover, the initiating antigen is often unknown in autoimmune disease. Here we tested the ability of antigen-specific Tregs generated by retroviral gene transfer to ameliorate arthritis through linked suppression and therefore without cognate recognition of the disease-initiating antigen. We explored two distinct strategies: T cell receptor (TCR) gene transfer into purified CD4+CD25+ T cells was used to redirect the specificity of naturally occurring Tregs; and co-transfer of FoxP3 and TCR genes served to convert conventional CD4(+) T cells into antigen-specific regulators. Following adoptive transfer into recipient mice, the gene-modified T cells engrafted efficiently and retained TCR and FoxP3 expression. Using an established arthritis model, we demonstrate antigen-driven accumulation of the gene modified T cells at the site of joint inflammation, which resulted in a local reduction in the number of inflammatory Th17 cells and a significant decrease in arthritic bone destruction. Together, we describe a robust strategy to rapidly generate antigen-specific regulatory T cells capable of highly targeted inhibition of tissue damage in the absence of systemic immune suppression. This opens the possibility to target Tregs to tissue-specific antigens for the treatment of autoimmune tissue damage without the knowledge of the disease-causing autoantigens recognized by pathogenic T cells.


Subject(s)
Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/therapy , Immunotherapy, Adoptive/methods , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , Autoantigens/immunology , Flow Cytometry , Gene Transfer Techniques , Genetic Vectors/genetics , Mice , Mice, Inbred C57BL , Retroviridae , T-Cell Antigen Receptor Specificity/genetics
9.
Front Immunol ; 13: 850358, 2022.
Article in English | MEDLINE | ID: mdl-35432319

ABSTRACT

Immunotherapy of cancer has made tremendous progress in recent years, as demonstrated by the remarkable clinical responses obtained from adoptive cell transfer (ACT) of patient-derived tumor infiltrating lymphocytes, chimeric antigen receptor (CAR)-modified T cells (CAR-T) and T cell receptor (TCR)-engineered T cells (TCR-T). TCR-T uses specific TCRS optimized for tumor engagement and can recognize epitopes derived from both cell-surface and intracellular targets, including tumor-associated antigens, cancer germline antigens, viral oncoproteins, and tumor-specific neoantigens (neoAgs) that are largely sequestered in the cytoplasm and nucleus of tumor cells. Moreover, as TCRS are naturally developed for sensitive antigen detection, they are able to recognize epitopes at far lower concentrations than required for CAR-T activation. Therefore, TCR-T holds great promise for the treatment of human cancers. In this focused review, we summarize basic, translational, and clinical insights into the challenges and opportunities of TCR-T. We review emerging strategies used in current ACT, point out limitations, and propose possible solutions. We highlight the importance of targeting tumor-specific neoAgs and outline a strategy of combining neoAg vaccines, checkpoint blockade therapy, and adoptive transfer of neoAg-specific TCR-T to produce a truly tumor-specific therapy, which is able to penetrate into solid tumors and resist the immunosuppressive tumor microenvironment. We believe such a combination approach should lead to a significant improvement in cancer immunotherapies, especially for solid tumors, and may provide a general strategy for the eradication of multiple cancers.


Subject(s)
Neoplasms , Receptors, Chimeric Antigen , Epitopes , Humans , Immunotherapy, Adoptive , Neoplasms/therapy , Receptors, Antigen, T-Cell/genetics , Receptors, Chimeric Antigen/genetics , T-Lymphocytes , Tumor Microenvironment
10.
J Hepatol ; 55(1): 103-10, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21145860

ABSTRACT

BACKGROUND & AIMS: Virus-specific T cells capable of controlling HBV and eliminating hepatocellular carcinoma (HCC) expressing HBV antigens are deleted or dysfunctional in patients with chronic HBV or HBV-related HCC. The goal of this study was to determine if T cell receptor (TCR) gene transfer can reconstitute HBV-specific T cell immunity in lymphocytes of chronic HBV patients and investigate whether HCC cells with natural HBV-DNA integration can be recognized by genetically modified T cells. METHODS: We used vector-mediated gene transfer to introduce HLA-A2-restricted, HBV-specific TCRs into T cells of chronic HBV as well as HBV-related HCC patients. RESULTS: The introduced TCRs were expressed on the cell surface, evidenced by Vß and pentamer staining. TCR transduced T cells produced IFN-γ, TNF-α, IL-2, and lysed HBV infected hepatocyte-like cell lines. Furthermore, HCC cell lines with natural HBV-DNA integration could be recognized by HBV-specific TCR-re-directed T cells. CONCLUSIONS: TCR re-directed HBV-specific T cells generated from PBMC of chronic HBV and HBV-related HCC patients were multifunctional and capable of recognizing HBV-infected cells and HCC tumor cells expressing viral antigens from naturally integrated HBV DNA. These genetically modified T cells could be used to reconstitute virus-specific T cell immunity in chronic HBV patients and target tumors in HBV-related HCC.


Subject(s)
Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/virology , Hepatitis B virus/immunology , Liver Neoplasms/immunology , Liver Neoplasms/virology , T-Lymphocytes/immunology , T-Lymphocytes/virology , Cell Line , Cell Line, Tumor , Gene Expression , Genetic Engineering , Genetic Vectors , HLA-A2 Antigen/metabolism , Hepatitis B Surface Antigens/genetics , Hepatitis B Surface Antigens/immunology , Hepatitis B, Chronic/immunology , Hepatitis B, Chronic/virology , Humans , Immunity, Cellular , Receptors, Antigen, T-Cell/genetics , Transduction, Genetic
11.
J Clin Invest ; 118(11): 3619-28, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18846251

ABSTRACT

T cell responses to MHC-mismatched transplants can be mediated via direct recognition of allogeneic MHC molecules on the cells of the transplant or via recognition of allogeneic peptides presented on the surface of recipient APCs in recipient MHC molecules - a process known as indirect recognition. As CD4(+)CD25(+) Tregs play an important role in regulating alloresponses, we investigated whether mouse Tregs specific for allogeneic MHC molecules could be generated in vitro and could promote transplantation tolerance in immunocompetent recipient mice. Tregs able to directly recognize allogeneic MHC class II molecules (dTregs) were obtained by stimulating CD4(+)CD25(+) cells from C57BL/6 mice (H-2(b)) with allogeneic DCs from BALB/c mice (H-2(d)). To generate Tregs that indirectly recognized allogeneic MHC class II molecules, dTregs were retrovirally transduced with TCR genes conferring specificity for H-2K(d) presented by H-2A(b) MHC class II molecules. The dual direct and indirect allospecificity of the TCR-transduced Tregs was confirmed in vitro. In mice, TCR-transduced Tregs, but not dTregs, induced long-term survival of partially MHC-mismatched heart grafts when combined with short-term adjunctive immunosuppression. Further, although dTregs were only slightly less effective than TCR-transduced Tregs at inducing long-term survival of fully MHC-mismatched heart grafts, histologic analysis of long-surviving hearts demonstrated marked superiority of the TCR-transduced Tregs. Thus, Tregs specific for allogeneic MHC class II molecules are effective in promoting transplantation tolerance in mice, which suggests that such cells have clinical potential.


Subject(s)
CD4 Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , Genes, T-Cell Receptor , T-Lymphocytes, Regulatory/immunology , Transplantation Tolerance/immunology , Animals , CD4 Antigens/genetics , CD4 Antigens/metabolism , CD4-Positive T-Lymphocytes/metabolism , Crosses, Genetic , Gene Transfer Techniques , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Retroviridae/genetics , Transduction, Genetic , Transplantation Tolerance/genetics , Transplantation, Homologous
12.
Cancer Immunol Immunother ; 60(9): 1243-55, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21553146

ABSTRACT

The cancer testis antigen Preferentially Expressed Antigen of Melanoma (PRAME) is overexpressed in many solid tumours and haematological malignancies whilst showing minimal expression in normal tissues and is therefore a promising target for immunotherapy. HLA-A0201-restricted peptide epitopes from PRAME have previously been identified as potential immunogens to drive antigen-specific autologous CTL responses, capable of lysing PRAME expressing tumour cells. CTL lines, from 13 normal donors and 10 melanoma patients, all of whom were HLA-A0201 positive, were generated against the PRAME peptide epitope PRA(100-108). Specific killing activity against PRA(100-108) peptide-pulsed targets was weak compared with CTL lines directed against known immunodominant peptides. Moreover, limiting dilution cloning from selected PRAME-specific CTL lines resulted in the generation of a clone of only low to intermediate avidity. Addition of the demethylating agent 5-aza-2'-Deoxycytidine (DAC) increased PRAME expression in 7 out of 11 malignant cell lines including several B lineage leukaemia lines and also increased class I expression. Pre-treatment of target cells was associated with increased sensitivity to antigen-specific killing by the low avidity CTL. When CTL, as well as of the target cells, were treated, the antigen-specific killing was further augmented. Interestingly, one HLA-A0201-negative DAC-treated line (RAJI) showed increased sensitivity to killing by clones despite a failure of expression of PRAME or HLA-A0201. Together these data point to a general increased augmentation of cancer immunogenocity by DAC involving both antigen-specific and non-specific mechanisms.


Subject(s)
Antigens, Neoplasm/immunology , Azacitidine/analogs & derivatives , Immunotherapy, Adoptive/methods , Neoplasms/immunology , Neoplasms/therapy , T-Lymphocytes, Cytotoxic/immunology , Antibody Affinity , Antigens, Neoplasm/metabolism , Antimetabolites, Antineoplastic/pharmacology , Azacitidine/pharmacology , Cell Line, Tumor , Cytotoxicity, Immunologic/immunology , Decitabine , HL-60 Cells , HLA-A2 Antigen/immunology , Humans , K562 Cells , Transfection
13.
Nat Med ; 9(8): 1009-14, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12847521

ABSTRACT

As a target for gene therapy, Duchenne muscular dystrophy (DMD) presents many obstacles but also an unparalleled prospect for correction by alternative splicing. The majority of mutations in the dystrophin gene occur in the region encoding the spectrin-like central rod domain, which is largely dispensable. Thus, splicing around mutations can generate a shortened but in-frame transcript, permitting translation of a partially functional dystrophin protein. We have tested this idea in vivo in the mdx dystrophic mouse (carrying a mutation in exon 23 of the dystrophin gene) by combining a potent transfection protocol with a 2-O-methylated phosphorothioated antisense oligoribonucleotide (2OMeAO) designed to promote skipping of the mutated exon*. The treated mice show persistent production of dystrophin at normal levels in large numbers of muscle fibers and show functional improvement of the treated muscle. Repeated administration enhances dystrophin expression without eliciting immune responses. Our data establishes the realistic practicality of an approach that is applicable, in principle, to a majority of cases of severe dystrophinopathy.


Subject(s)
Dystrophin/genetics , Exons , Genetic Therapy/methods , Muscular Dystrophy, Animal/therapy , Muscular Dystrophy, Duchenne/therapy , Mutation , Animals , Gene Expression Regulation , Humans , Mice , Mice, Inbred mdx , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Duchenne/genetics , Oligonucleotides, Antisense/metabolism , RNA Splicing
14.
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
15.
J Mol Med (Berl) ; 86(5): 573-83, 2008 May.
Article in English | MEDLINE | ID: mdl-18335188

ABSTRACT

The transfer of T cell receptor (TCR) genes allows to endow T cells with a new antigen specificity. For clinical applications of TCR-redirected T cells, efficient functional expression of the transgenic TCR is a key prerequisite. Here, we compared the influence of the transgene cassette on the expression and function of the murine TCR P14 (recognizing a LCMV gp33 epitope) and the human TCR WT-1 (recognizing an epitope of the tumor-associated antigen WT-1). We constructed different vectors, in which TCRalpha- and beta-chain genes were either (a) linked by an internal ribosomal entry site (IRES), (b) combined by a 2A peptide, or (c) introduced into two individual retroviral constructs. While in a TCR-deficient T cell line TCR P14 was expressed equally well by all constructs, we found that IRES- but not 2A-employing TCR expression is hampered in a TCR-bearing cell line and in primary murine T cells where the transgenic TCR has to compete with endogenous TCR chains. Similarly, 2A-linked TCR WT-1 genes yielded highest expression and function as measured by tetramer binding and peptide-specific IFN-gamma secretion. Differences in expression were independent of copy number integration as shown by real-time PCR. Thus, linking TCRalpha- and beta-chain genes by a 2A peptide is superior to an IRES for TCR expression and T cell function.


Subject(s)
Mutagenesis, Insertional , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/immunology , Transgenes/genetics , Animals , Cell Line , Cell Membrane/metabolism , Gene Dosage , Gene Expression Regulation , Genetic Vectors , Humans , Mice , Mice, Inbred C57BL , Peptides/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retroviridae
16.
Nucleic Acids Res ; 35(10): 3391-406, 2007.
Article in English | MEDLINE | ID: mdl-17478522

ABSTRACT

EBV has two lytic origins (oriLyt) of DNA replication lying at divergent sites on the viral genome within a duplicated sequence (DS). The latter contains potential hairpin loops, 'hinge' elements and the promoters for transcripts from viral genes BHLF1 and LF3. These genes themselves consist largely of 125 and 102 bp repetitive sequences, respectively, and encode basic proteins. We have examined these genomic regions in detail in attempts to understand why lytic replication--necessary for virus survival--is so inefficient, and to identify controlling elements. Our studies uncovered a diverse family of promoters (P) for BHLF1 and LF3, only one pair of which (P1) proved sensitive to chemical inducing agents. The others (P2-P3/4), abutting the replication 'core' origin elements in DS and extending into 5'-unique sequences, may play roles in the maintenance of viral latency. We further identified a family of overlapping small complementary-strand RNAs that transverse the replication 'core' origin elements in a manner suggesting a role for them as 'antisense' species and/or DNA replication primers. Our data are discussed in terms of alternative lytic replication models. We suggest our findings might prove useful in seeking better control over viral lytic replication and devising strategies for therapy.


Subject(s)
DNA Replication , DNA, Viral/biosynthesis , Herpesvirus 4, Human/genetics , Viral Proteins/genetics , Animals , Cell Line , Gene Expression Regulation, Viral , Herpesvirus 4, Human/metabolism , Herpesvirus 4, Human/physiology , Humans , Nuclease Protection Assays , Promoter Regions, Genetic , RNA, Antisense/analysis , RNA, Messenger/biosynthesis , RNA, Messenger/chemistry , RNA, Viral/biosynthesis , RNA, Viral/chemistry , Replication Origin , Reverse Transcriptase Polymerase Chain Reaction , Transcription Initiation Site , Transcription, Genetic , Viral Proteins/biosynthesis , Virus Replication
18.
Immunology ; 124(3): 315-21, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18217949

ABSTRACT

T-cell-based antigen-specific immunotherapy targeting tumour-associated antigens offers the potential for cancer immunotherapy. However, the majority of identified tumour-associated antigens are also expressed at low levels in normal tissues and mechanisms of tolerance induction are likely to affect the quality of T-cell responses to such antigens. In this study a T-cell receptor transgenic model was developed to determine the magnitude of T-cell tolerance to the tumour-associated antigen murine double minute-2 (MDM2), a widely expressed protein that is found at elevated levels in many tumours. The analysis of transgenic mice showed that thymic deletion was responsible for purging large numbers of MDM2-specific T cells from the repertoire. However, some T cells with specificity for MDM2 were able to escape thymic deletion and persisted in the peripheral T-cell pool. Functional analysis revealed that these T cells displayed defects in antigen-driven expansion. This functional impairment of the MDM2-specific T cells was maintained following adoptive transfer of the T cells into hosts that were unable to present the T-cell-receptor-recognized antigen. This study demonstrates that thymic deletion and the functional impairment of T cells present in the periphery both operate to establish T-cell tolerance to the tumour-associated antigen MDM2. Furthermore, the tolerant phenotype was stable and did not require continuous MDM2 peptide presentation in normal tissues.


Subject(s)
Proto-Oncogene Proteins c-mdm2/immunology , Receptors, Antigen, T-Cell/immunology , Animals , Antigen Presentation/immunology , Antigens, Neoplasm/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Clonal Deletion/immunology , Immune Tolerance , Mice , Mice, Transgenic , Spleen/immunology , Thymus Gland/immunology
20.
Mol Cell Biol ; 23(6): 2192-201, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12612089

ABSTRACT

Frameshift mutations provide recognized mechanisms for changing the coding potential of an organism. Here, multiple frameshifts are identified in repetitive sequences within an Epstein-Barr virus unspliced early gene, LF3, which is associated with the viral replicative cycle and also transcriptionally expressed in many virally associated tumors. On the DNA strand encoding LF3, there are three open reading frames, only one of which contains an initiation codon. Most (>95%) of the gene consists of numerous (>20, varying with cell source) GC-rich copies of a 102-bp direct repeat (called IR 4) flanked by small unique sequences. LF3 may express a protein if its initiation and termination codons reside in the same reading frame, but this is not always the case. Frameshifting events, occurring in short runs of pyrimidines (mainly C residues) in the repeats, give rise to mutations which may provide a mechanism for escape of an LF3 function from host surveillance. Sequence studies link these frameshifts to DNA replication errors. Notably, the number of sites in LF3 at which such mutations can occur permits a very large amount of diversity in this gene. Our data also suggest a second degeneracy mechanism within the protein itself, which influences its stability and may reflect a host defense mechanism. LF3 thus provides a potentially important model for studying the quest for supremacy between a virus and its host.


Subject(s)
Genes, Viral , Herpesvirus 4, Human/genetics , Repetitive Sequences, Nucleic Acid , Viral Proteins/genetics , Viral Structural Proteins/genetics , Amino Acid Sequence , Animals , Base Pairing , Base Sequence , Burkitt Lymphoma/pathology , Burkitt Lymphoma/virology , Carcinoma/pathology , Carcinoma/virology , Cell Line, Transformed , Codon/genetics , DNA Replication , DNA, Viral/genetics , Epstein-Barr Virus Infections/pathology , Epstein-Barr Virus Infections/virology , Frameshift Mutation , Genetic Variation , Humans , Infectious Mononucleosis/pathology , Infectious Mononucleosis/virology , Mice , Mice, Nude , Molecular Sequence Data , Nasopharyngeal Neoplasms/pathology , Nasopharyngeal Neoplasms/virology , Open Reading Frames , Selection, Genetic , Sequence Alignment , Tumor Escape , Viral Proteins/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL