Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Cancer Immunol Immunother ; 72(6): 1553-1565, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36526910

ABSTRACT

Immunotherapies targeting truly tumor-specific targets focus on the expansion and activation of T cells against neoantigens or oncogenic viruses. One target is the human papilloma virus type 16 (HPV16), responsible for several anogenital cancers and oropharyngeal carcinomas. Spontaneous and vaccine-induced HPV-specific T cells have been associated with better clinical outcome. However, the epitopes and restriction elements to which these T cells respond remained elusive. To identify CD8+ T cell epitopes in cultures of tumor infiltrating lymphocytes, we here used multimers and/or a functional screening platform exploiting single HLA class I allele-engineered antigen presenting cells. This resulted in the detection of 20 CD8+ T cell responses to 11 different endogenously processed HLA-peptide combinations within 12 HPV16-induced tumors. Specific HLA-peptide combinations dominated the response in patients expressing these HLA alleles. T cell receptors (TCRs) reactive to seven different HLA class I-restricted peptides could be isolated and analysis revealed tumor reactivity for five of the six TCRs analyzed. The tumor reactive TCRs to these dominant HLA class I peptide combinations can potentially be used to engineer tumor-specific T cells for adoptive cell transfer approaches to treat HPV16-induced cancers.


Subject(s)
Neoplasms , Papillomavirus Infections , Humans , Human papillomavirus 16 , Neoplasms/metabolism , CD8-Positive T-Lymphocytes , Receptors, Antigen, T-Cell , Histocompatibility Antigens Class I , Lymphocytes, Tumor-Infiltrating , Epitopes, T-Lymphocyte , Peptides
2.
Oncoimmunology ; 8(3): 1542917, 2019.
Article in English | MEDLINE | ID: mdl-30723575

ABSTRACT

We explored whether engineering of T cell specificity and effector function improves immunotherapy of solid tumors. Although IL-12 can enhance cancer immunity, a strategy of safe IL-12 delivery without toxicity is currently lacking. We engineered T cells to express IL-12 controlled by the NFAT promoter responsive to TCR stimulation, or by the Tet-On promoter responsive to doxycycline. In vivo, NFAT-engineered T cells caused lethal toxicity, while Tet-engineered T cells were safe in the absence of doxycycline. Combining gene transfer of the melanoma-specific TRP2-TCR with Tet-IL-12 engineering revealed that temporal induction of IL-12 was essential to inhibit the growth of B16F10 melanoma tumors. Induced IL-12 increased the number of tumor-infiltrating T cells and also prevented the down-modulation of the TRP2-TCR and the associated up-regulation of the PD1 marker that was observed in the absence of IL-12. In addition, temporal induction of IL-12 expression also increased the number of plasmacytoid DC in the tumor micro-environment. We show that repeated induction of IL-12 can be used to enhance control of tumor growth without encountering systemic toxicity. The observation that TCR engineering combined with Tet-regulated IL-12 expression can achieve tumor immunity without the side effects that are usually associated with the in vivo use of IL-12 warrants translation of this concept into the clinic.

3.
Nat Med ; 25(1): 89-94, 2019 01.
Article in English | MEDLINE | ID: mdl-30510250

ABSTRACT

Infiltration of human cancers by T cells is generally interpreted as a sign of immune recognition, and there is a growing effort to reactivate dysfunctional T cells at such tumor sites1. However, these efforts only have value if the intratumoral T cell receptor (TCR) repertoire of such cells is intrinsically tumor reactive, and this has not been established in an unbiased manner for most human cancers. To address this issue, we analyzed the intrinsic tumor reactivity of the intratumoral TCR repertoire of CD8+ T cells in ovarian and colorectal cancer-two tumor types for which T cell infiltrates form a positive prognostic marker2,3. Data obtained demonstrate that a capacity to recognize autologous tumor is limited to approximately 10% of intratumoral CD8+ T cells. Furthermore, in two of four patient samples tested, no tumor-reactive TCRs were identified, despite infiltration of their tumors by T cells. These data indicate that the intrinsic capacity of intratumoral T cells to recognize adjacent tumor tissue can be rare and variable, and suggest that clinical efforts to reactivate intratumoral T cells will benefit from approaches that simultaneously increase the quality of the intratumoral TCR repertoire.


Subject(s)
Neoplasms/immunology , Neoplasms/pathology , Receptors, Antigen, T-Cell/metabolism , CD8-Positive T-Lymphocytes/immunology , Humans , Jurkat Cells , Lymphocytes, Tumor-Infiltrating/immunology , Phenotype , Reproducibility of Results
4.
J Immunother Cancer ; 6(1): 70, 2018 07 13.
Article in English | MEDLINE | ID: mdl-30001747

ABSTRACT

Mutation-derived neoantigens represent an important class of tumour-specific, tumour rejection antigens, and are attractive targets for TCR gene therapy of cancer. The majority of such mutations are patient-specific and targeting these requires a fully personalized approach. However, some mutations are found recurrently among cancer patients, and represent potential targets for neoantigen-specific TCR gene therapy that is more widely applicable. Therefore, we have investigated if some cancer mutations found recurrently in hematological malignancies encode immunogenic neoantigens presented by common European Caucasoid HLA class I alleles and can form targets for TCR gene therapy. We initially focused on identifying HLA class I neoepitopes derived from calreticulin (CALR) exon 9 mutations, found in ~ 80% of JAK2wt myeloproliferative neoplasms (MPN). Based on MHC class I peptide predictions, a number of peptides derived from mutant CALR (mCALR) were predicted to bind to HLA-A*03:01 and HLA-B*07:02. However, using mass spectrometry and ex vivo pMHC multimer staining of PBMC from MPN patients with CALR exon 9 mutations, we found no evidence that these peptides were naturally processed and presented on the surface of mCALR-expressing target cells. We next developed a protocol utilizing pMHC multimers to isolate CD8+ T cells from healthy human donor PBMC that are specific for mCALR and additional putative neoepitopes found recurrently in hematological malignancies. Using this approach, CD8+ T cells specific for HLA-A*03:01- and HLA-B*07:02-presented mCALR peptides and an HLA-A*11:01-presented mutant FBXW7 (mFBXW7) peptide were successfully isolated. TCRs isolated from mCALR-specific CD8+ T cell populations were not able to recognize target cells engineered to express mCALR. In contrast, mFBXW7-specific CD8+ T cells were able to recognize target cells engineered to express mFBXW7. In conclusion, while we found no evidence for mCALR derived neoepitope presentation in the context of the HLA class I alleles studied, our data suggests that the recurrent pR465H mutation in FBXW7 may encode an HLA-A*11:01 presented neoepitope, and warrants further investigation as a target for T cell based immunotherapy of cancer.


Subject(s)
Antigens, Neoplasm/immunology , Hematologic Neoplasms/immunology , Hematologic Neoplasms/metabolism , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Epitopes, T-Lymphocyte/immunology , HLA Antigens/genetics , HLA Antigens/immunology , Hematologic Neoplasms/pathology , Humans , Lymphocyte Activation/immunology , Mutation , Peptides/genetics , Peptides/immunology , Receptors, Antigen, T-Cell/genetics , Recurrence , T-Cell Antigen Receptor Specificity
5.
Cancer Res ; 75(15): 3043-53, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26054597

ABSTRACT

Neuroblastoma is the most common extracranial solid tumor of childhood, and survival remains poor for patients with advanced disease. Novel immune therapies are currently in development, but clinical outcomes have not matched preclinical results. Here, we describe key mechanisms in which neuroblastoma inhibits the immune response. We show that murine and human neuroblastoma tumor cells suppress T-cell proliferation through increased arginase activity. Arginase II is the predominant isoform expressed and creates an arginine-deplete local and systemic microenvironment. Neuroblastoma arginase activity results in inhibition of myeloid cell activation and suppression of bone marrow CD34(+) progenitor proliferation. Finally, we demonstrate that the arginase activity of neuroblastoma impairs NY-ESO-1-specific T-cell receptor and GD2-specific chimeric antigen receptor-engineered T-cell proliferation and cytotoxicity. High arginase II expression correlates with poor survival for patients with neuroblastoma. The results support the hypothesis that neuroblastoma creates an arginase-dependent immunosuppressive microenvironment in both the tumor and blood that leads to impaired immunosurveillance and suboptimal efficacy of immunotherapeutic approaches.


Subject(s)
Arginase/metabolism , Neuroblastoma/immunology , Tumor Microenvironment/immunology , Animals , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Arginase/immunology , Arginine/metabolism , Cell Proliferation , Gangliosides/metabolism , Humans , Lymphocyte Activation/immunology , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice , Neoplasms, Experimental/immunology , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Neuroblastoma/metabolism , Neuroblastoma/mortality , Neuroblastoma/pathology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism
6.
Hum Gene Ther ; 26(5): 276-85, 2015 May.
Article in English | MEDLINE | ID: mdl-25860661

ABSTRACT

Adoptive T-cell therapy is delivering objective clinical responses across a number of cancer indications in the early phase clinical setting. Much of this clinical activity is taking place at major clinical academic centers across the United States. This review focuses upon cancer-focused cell therapy activity within the United Kingdom as a contribution to the 2015 British Society of Gene and Cell Therapy annual general meeting. This overview reflects the diversity and expansion of clinical and preclinical studies within the United Kingdom while considering the background context of this work against new infrastructural developments and the requirements of nationalized healthcare delivery within the UK National Health Service.


Subject(s)
Immunotherapy, Adoptive , Neoplasms/immunology , Neoplasms/therapy , T-Lymphocytes/immunology , Animals , Biotechnology , Cell- and Tissue-Based Therapy , Clinical Trials as Topic , Drug Evaluation, Preclinical , Humans , Immunotherapy, Adoptive/methods , T-Lymphocytes/metabolism , United Kingdom
7.
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
8.
Nat Med ; 19(11): 1534-41, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24121928

ABSTRACT

The transfer of T cell receptor (TCR) genes into patient T cells is a promising approach for the treatment of both viral infections and cancer. Although efficient methods exist to identify antibodies for the treatment of these diseases, comparable strategies to identify TCRs have been lacking. We have developed a high-throughput DNA-based strategy to identify TCR sequences by the capture and sequencing of genomic DNA fragments encoding the TCR genes. We establish the value of this approach by assembling a large library of cancer germline tumor antigen-reactive TCRs. Furthermore, by exploiting the quantitative nature of TCR gene capture, we show the feasibility of identifying antigen-specific TCRs in oligoclonal T cell populations from either human material or TCR-humanized mice. Finally, we demonstrate the ability to identify tumor-reactive TCRs within intratumoral T cell subsets without knowledge of antigen specificities, which may be the first step toward the development of autologous TCR gene therapy to target patient-specific neoantigens in human cancer.


Subject(s)
Genes, T-Cell Receptor , High-Throughput Nucleotide Sequencing/methods , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Animals , Antigens, Neoplasm/metabolism , Gene Library , Genetic Therapy , Humans , Mice , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/therapy , T-Lymphocytes/immunology
9.
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
10.
J Invest Dermatol ; 131(9): 1806-16, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21677669

ABSTRACT

T-cell receptor (TCR) gene therapy aims to induce immune reactivity against tumors by introducing genes encoding a tumor-reactive TCR into patient T cells. This approach has been extensively tested in preclinical mouse models, and initial clinical trials have demonstrated the feasibility and potential of TCR gene therapy as a cancer treatment. However, data obtained from preclinical and clinical studies suggest that both the therapeutic efficacy and the safety of TCR gene therapy can be and needs to be further enhanced. This review highlights those strategies that can be followed to develop TCR gene therapy into a clinically relevant treatment option for cancer patients.


Subject(s)
Genetic Therapy/methods , Genetic Therapy/trends , Neoplasms/genetics , Neoplasms/therapy , Receptors, Antigen, T-Cell/genetics , Animals , Humans , Neoplasms/immunology , Receptors, Antigen, T-Cell/immunology , Treatment Outcome
11.
J Biomed Opt ; 16(1): 016004, 2011.
Article in English | MEDLINE | ID: mdl-21280910

ABSTRACT

To improve surgical guidance toward prostate draining lymph nodes, we investigate the potential of intraoperative fluorescence imaging and combined pre- and intraoperative multimodality imaging approaches. Transgenic adenocarcinoma mouse prostate mice with spontaneous prostate tumors are injected intratumorally with: 1. a cocktail of patent blue (Pb) and indocyanine green (ICG); 2. a cocktail of albumin radiocolloids (99mTc-NanoColl), Pb, and ICG; or 3. a cocktail of radiolabeled albumin (99mTc-Vasculosis), Pb, and ICG. The distribution of these imaging agents over the lymph nodes (LNs) are studied at different time points after injection. We find that at 60-min postinjection, ICG significantly improves the detection of the LNs compared to Pb, 53 versus 7%, respectively. Moreover, a cocktail of ICG and 99mTc-NanoColl improves the fluorescent detection rate to 86%, equalling that of the clinically applied 99mTc-NanoColl. A similar overlap is observed in our initial clinical pilot data. Fluorescent detection of the LNs using a ICG with 99mTc-Vasculosis gives similar results as "free" ICG (58%; 60 min). A 99mTc-NanoColl, Pb, and cocktail ICG enriches the standard 99mTc-NanoColl approach by adding optical detection of the sentinel lymph nodes. Furthermore, this approach improves fluorescent-based guidance and enables both accurate surgical planning and intraoperative detection, based on a single injection.


Subject(s)
Adenocarcinoma/diagnosis , Adenocarcinoma/secondary , Lymph Nodes/diagnostic imaging , Lymph Nodes/pathology , Microscopy, Fluorescence/methods , Prostatic Neoplasms/diagnosis , Radioisotopes , Animals , Contrast Media/administration & dosage , Injections , Male , Mice , Radioisotopes/administration & dosage , Radionuclide Imaging/methods , Radiopharmaceuticals/administration & dosage , Surgery, Computer-Assisted/methods
12.
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
13.
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
14.
Curr Opin Immunol ; 21(2): 209-14, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19321326

ABSTRACT

The adoptive transfer of TCR gene-modified T cells has been developed with the aim to induce immune reactivity toward defined tumor-associated antigens to which the endogenous T cell repertoire is non-responsive. Here we discuss in which areas preclinical studies in mouse models can or cannot be expected to be of value to guide clinical trial design, and how the available data from preclinical studies should influence forthcoming clinical trials.


Subject(s)
Genetic Therapy/methods , Immunotherapy, Adoptive/methods , Neoplasms/therapy , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/metabolism , Animals , Antigens, Neoplasm/immunology , Disease Models, Animal , Humans , Mice , Neoplasms/genetics , Neoplasms/metabolism , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , T-Lymphocytes/transplantation
15.
J Immunol ; 181(7): 5128-36, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18802117

ABSTRACT

Adoptive transfer of TCR gene-modified T cells has been proposed as an attractive approach to target tumors for which it is difficult or impossible to induce strong tumor-specific T cell responses by vaccination. Whereas the feasibility of generating tumor Ag-specific T cells by gene transfer has been demonstrated, the factors that determine the in vivo effectiveness of TCR-modified T cells are largely unknown. We have analyzed the value of a number of clinically feasible strategies to enhance the antitumor potential of TCR modified T cells. These experiments reveal three factors that contribute greatly to the in vivo potency of TCR-modified T cells. First, irradiation-induced host conditioning is superior to vaccine-induced activation of genetically modified T cells. Second, increasing TCR expression through genetic optimization of TCR sequences has a profound effect on in vivo antitumor activity. Third, a high precursor frequency of TCR modified T cells within the graft is essential. Tumors that ultimately progress in animals treated with this optimized regimen for TCR-based adoptive cell transfer invariably display a reduced expression of the target Ag. This suggests TCR gene therapy can achieve a sufficiently strong selective pressure to warrant the simultaneous targeting of multiple Ags. The strategies outlined in this study should be of value to enhance the antitumor activity of TCR-modified T cells in clinical trials.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/transplantation , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Receptors, Antigen, T-Cell/administration & dosage , Receptors, Antigen, T-Cell/genetics , Transduction, Genetic , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/radiation effects , Cancer Vaccines/administration & dosage , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Cell Line, Tumor , Gamma Rays , Genetic Vectors/radiation effects , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Transgenic , Ovalbumin/genetics , Receptors, Antigen, T-Cell/radiation effects , Receptors, Antigen, T-Cell/therapeutic use , Retroviridae/genetics , Retroviridae/immunology , Transplantation Conditioning , Viral Vaccines/administration & dosage , Viral Vaccines/genetics , Viral Vaccines/immunology , Whole-Body Irradiation
16.
J Immunol ; 181(4): 2563-71, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18684947

ABSTRACT

Analogous to the clinical use of recombinant high-affinity Abs, transfer of TCR genes may be used to create a T cell compartment specific for self-Ags to which the endogenous T cell repertoire is immune tolerant. In this study, we show in a spontaneous prostate carcinoma model that the combination of vaccination with adoptive transfer of small numbers of T cells that are genetically modified with a tumor-specific TCR results in a marked suppression of tumor development, even though both treatments are by themselves without effect. These results demonstrate the value of TCR gene transfer to target otherwise nonimmunogenic tumor-associated self-Ags provided that adoptive transfer occurs under conditions that allow in vivo expansion of the TCR-modified T cells.


Subject(s)
Immunotherapy, Adoptive , Lymphocyte Activation/immunology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/therapy , Receptors, Antigen, T-Cell/therapeutic use , T-Lymphocytes/immunology , Transduction, Genetic , Adenocarcinoma/genetics , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Adenocarcinoma/therapy , Animals , Antigens, Viral, Tumor/biosynthesis , Antigens, Viral, Tumor/genetics , Clone Cells , Immunotherapy, Adoptive/methods , Influenza A virus/immunology , Lymphocyte Activation/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology , Receptors, Antigen, T-Cell/administration & dosage , Simian virus 40/immunology , T-Lymphocytes/virology , Transduction, Genetic/methods , Vaccinia/immunology
17.
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
18.
Eur J Immunol ; 37(6): 1634-41, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17506031

ABSTRACT

In this study, we have started to dissect the molecular basis of CD8 dependence of a high and low avidity CTL clone specific for the same peptide epitope. Using anti-CD8alpha and anti-CD8beta antibodies, we found that cytotoxicity and IFN-gamma production by high but not by low avidity CTL was strongly CD8 dependent. We isolated the TCR genes of both types of CTL clones and used retroviral gene transfer to analyse the function of these TCR in primary T cells of wild-type and CD8beta-deficient mice. Both TCR triggered antigen-specific killing in wild-type T cells, and blocking experiments showed that CD8 dependence/independence co-transferred with the TCR into primary T cells, indicating that it was dictated by the TCR itself. Gene transfer experiments into CD8beta-deficient T cells revealed that only the TCR derived from the CD8-independent CTL clone elicited antigen-specific cytotoxicity, while the CD8-dependent TCR was non-functional in the absence of the CD8beta-chain. These data indicate a striking difference between CD8alpha/beta heterodimers and CD8alpha/alpha homodimers as only the former were able to provide co-receptor function for the CD8-dependent TCR.


Subject(s)
CD8 Antigens/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , CD3 Complex/immunology , CD8 Antigens/genetics , Cell Line, Tumor , Cytotoxicity, Immunologic/drug effects , Cytotoxicity, Immunologic/immunology , Interferon-gamma/metabolism , Interleukin-4/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Peptides/immunology , Protein Subunits/immunology , Protein Subunits/metabolism , Proto-Oncogene Proteins c-mdm2/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell, alpha-beta/immunology , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Spleen/cytology , T-Lymphocytes, Cytotoxic/metabolism , Transfection
19.
PLoS One ; 2(4): e353, 2007 Apr 04.
Article in English | MEDLINE | ID: mdl-17406677

ABSTRACT

BACKGROUND: Most tumor-associated antigens (TAA) currently used for immunotherapy of cancer are also expressed in normal tissues, which may induce tolerance and impair T cell-mediated immunity. However, there is limited information about how physiological expression in normal tissues alters the function of TAA-specific T cells. METHODOLOGY/PRINCIPAL FINDINGS: We used a T cell receptor transgenic model to study how MDM2 expression in normal tissues affects the function of T cells specific for this TAA that is found at high levels in many different types of tumors. We found that some MDM2-specific T cells escaped thymic deletion and persisted in the peripheral T cell pool. When stimulated with antigen, these T cells readily initiated cell division but failed to proliferate and expand, which was associated with a high rate of apoptosis. Both IL-2 and IL-15 efficiently rescued T cell survival and antigen-specific T cell proliferation, while IL-7 and IL-21 were ineffective. Antigen-stimulated T cells showed impaired expression of the effector molecules CD43, granzyme-B and IFN-gamma, a defect that was completely restored when T cells were stimulated in the presence of IL-2. In contrast, IL-15 and IL-21 only restored the expression of CD43 and granzyme-B, but not IFN-gamma production. Finally, peptide titration experiments with IL-2 rescued T cells indicated that they were of lower avidity than non-tolerant control T cells expressing the same TCR. CONCLUSIONS/SIGNIFICANCE: These data indicate that cytokines can rescue the antigen-specific proliferation and effector function of MDM2-specific T cells, although this does not lead to the recovery of high avidity T cell function. This study sheds light on possible limitations of immunotherapy approaches that target widely expressed TAA, such as MDM2.


Subject(s)
Antibody Affinity , Cytokines/physiology , Immune Tolerance , Proto-Oncogene Proteins c-mdm2/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Base Sequence , Cell Differentiation , Cell Proliferation , DNA Primers , Flow Cytometry , Mice , Mice, Transgenic , Polymerase Chain Reaction , T-Lymphocytes, Cytotoxic/cytology
20.
Expert Opin Biol Ther ; 5(9): 1183-92, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16120049

ABSTRACT

T cell-based antigen-specific immunotherapy targeting self-proteins aberrantly expressed in many tumours offers the potential for widely applicable cancer immunotherapy, but carries the risk of autoimmunity. Immunological tolerance represents an inherent limitation of cancer vaccines targeting such broadly expressed tumour-associated proteins. Therefore, strategies to circumvent T cell tolerance have been developed and, when combined with T cell receptor (TCR) gene transfer technology, can generate highly avid tumour-reactive patient cytotoxic T lymphocytes (CTLs) specific for peptide epitopes of tumour-associated proteins. This review analyses the level of tolerance to broadly expressed tumour-associated proteins in the autologous T cell repertoire, assesses strategies that have been developed to circumvent T cell tolerance to such antigens, and evaluates the prospects for effective immunotherapy targeting broadly expressed tumour-associated proteins.


Subject(s)
Adoptive Transfer , Antigens, Neoplasm/immunology , Cancer Vaccines/therapeutic use , Immunotherapy , Neoplasms/therapy , Animals , Antigens, Neoplasm/metabolism , Cancer Vaccines/immunology , Clinical Trials as Topic , Genetic Therapy , Humans , Immune Tolerance , Immunotherapy/methods , Models, Animal , Neoplasms/immunology , Neoplasms/metabolism , Proto-Oncogene Proteins c-mdm2/immunology , Proto-Oncogene Proteins c-mdm2/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Cytotoxic/transplantation
SELECTION OF CITATIONS
SEARCH DETAIL
...