Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Immunol ; 208(1): 169-180, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34853077

ABSTRACT

Adoptive T cell therapy with T cells expressing affinity-enhanced TCRs has shown promising results in phase 1/2 clinical trials for solid and hematological tumors. However, depth and durability of responses to adoptive T cell therapy can suffer from an inhibitory tumor microenvironment. A common immune-suppressive agent is TGF-ß, which is secreted by tumor cells and cells recruited to the tumor. We investigated whether human T cells could be engineered to be resistant to inhibition by TGF-ß. Truncating the intracellular signaling domain from TGF-ß receptor (TGFßR) II produces a dominant-negative receptor (dnTGFßRII) that dimerizes with endogenous TGFßRI to form a receptor that can bind TGF-ß but cannot signal. We previously generated specific peptide enhanced affinity receptor TCRs recognizing the HLA-A*02-restricted peptides New York esophageal squamous cell carcinoma 1 (NY-ESO-1)157-165/l-Ag family member-1A (TCR: GSK3377794, formerly NY-ESO-1c259) and melanoma Ag gene A10254-262 (TCR: ADP-A2M10, formerly melanoma Ag gene A10c796). In this article, we show that exogenous TGF-ß inhibited in vitro proliferation and effector functions of human T cells expressing these first-generation high-affinity TCRs, whereas inhibition was reduced or abolished in the case of second-generation TCRs coexpressed with dnTGFßRII (e.g., GSK3845097). TGF-ß isoforms and a panel of TGF-ß-associated genes are overexpressed in a range of cancer indications in which NY-ESO-1 is commonly expressed, particularly in synovial sarcoma. As an example, immunohistochemistry/RNAscope identified TGF-ß-positive cells close to T cells in tumor nests and stroma, which had low frequencies of cells expressing IFN-γ in a non-small cell lung cancer setting. Coexpression of dnTGFßRII may therefore improve the efficacy of TCR-transduced T cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Carcinoma, Squamous Cell/therapy , Hematologic Neoplasms/therapy , Immunotherapy, Adoptive/methods , Melanoma/therapy , Receptor, Transforming Growth Factor-beta Type II/metabolism , Receptors, Antigen, T-Cell/metabolism , Receptors, Chimeric Antigen/metabolism , Sarcoma, Synovial/therapy , Transforming Growth Factor beta/metabolism , Antigens, Neoplasm/immunology , Carcinoma, Squamous Cell/immunology , Cell Line, Tumor , Genetic Engineering , HLA-A2 Antigen/metabolism , Hematologic Neoplasms/immunology , Humans , Immune Tolerance , Melanoma/immunology , Membrane Proteins/immunology , Neoplasm Proteins/immunology , Peptide Fragments/immunology , Receptor, Transforming Growth Factor-beta Type II/genetics , Receptors, Antigen, T-Cell/genetics , Receptors, Chimeric Antigen/genetics , Sarcoma, Synovial/immunology , T-Cell Antigen Receptor Specificity , Tumor Microenvironment
2.
PLoS One ; 13(10): e0205491, 2018.
Article in English | MEDLINE | ID: mdl-30321203

ABSTRACT

Robust preclinical testing is essential to predict clinical safety and efficacy and provide data to determine safe dose for first-in-man studies. There are a growing number of examples where the preclinical development of drugs failed to adequately predict clinical adverse events in part due to their assessment with inappropriate preclinical models. Preclinical investigations of T cell receptor (TCR)-based immunotherapies prove particularly challenging as these biologics are human-specific and thus the conventional testing in animal models is inadequate. As these molecules harness the full force of the immune system, and demonstrate tremendous potency, we set out to design a preclinical package that would ensure adequate evaluation of these therapeutics. Immune Mobilising Monoclonal TCR Against Cancer (ImmTAC) molecules are bi-specific biologics formed of an affinity-enhanced TCR fused to an anti-CD3 effector function. ImmTAC molecules are designed to activate human T lymphocytes and target peptides within the context of a human leukocyte antigen (HLA), thus require an intact human immune system and peptidome for suitable preclinical screening. Here we draw upon the preclinical testing of four ImmTAC molecules, including IMCgp100, the first ImmTAC molecule to reach the clinic, to present our comprehensive, informative and robust approach to in vitro preclinical efficacy and safety screening. This package comprises a broad range of cellular and molecular assays using human tissues and cultured cells to test efficacy, safety and specificity, and hence predict human responses in clinical trials. We propose that this entirely in vitro package offers a potential model to be applied to screening other TCR-based biologics.


Subject(s)
Antibodies, Bispecific/pharmacology , Drug Screening Assays, Antitumor/methods , Proteins/pharmacology , Single-Chain Antibodies/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Humans , In Vitro Techniques , Workflow
3.
Sci Transl Med ; 5(197): 197ra103, 2013 Aug 07.
Article in English | MEDLINE | ID: mdl-23926201

ABSTRACT

MAGE A3, which belongs to the family of cancer-testis antigens, is an attractive target for adoptive therapy given its reactivation in various tumors and limited expression in normal tissues. We developed an affinity-enhanced T cell receptor (TCR) directed to a human leukocyte antigen (HLA)-A*01-restricted MAGE A3 antigen (EVDPIGHLY) for use in adoptive therapy. Extensive preclinical investigations revealed no off-target antigen recognition concerns; nonetheless, administration to patients of T cells expressing the affinity-enhanced MAGE A3 TCR resulted in a serious adverse event (SAE) and fatal toxicity against cardiac tissue. We present a description of the preclinical in vitro functional analysis of the MAGE A3 TCR, which failed to reveal any evidence of off-target activity, and a full analysis of the post-SAE in vitro investigations, which reveal cross-recognition of an off-target peptide. Using an amino acid scanning approach, a peptide from the muscle protein Titin (ESDPIVAQY) was identified as an alternative target for the MAGE A3 TCR and the most likely cause of in vivo toxicity. These results demonstrate that affinity-enhanced TCRs have considerable effector functions in vivo and highlight the potential safety concerns for TCR-engineered T cells. Strategies such as peptide scanning and the use of more complex cell cultures are recommended in preclinical studies to mitigate the risk of off-target toxicity in future clinical investigations.


Subject(s)
Antigen Presentation/immunology , Antigens, Neoplasm/immunology , Connectin/chemistry , Cross Reactions/immunology , HLA-A1 Antigen/immunology , Neoplasm Proteins/immunology , Peptides/immunology , T-Lymphocytes/metabolism , Amino Acid Sequence , Antigens, Neoplasm/chemistry , Antineoplastic Agents/pharmacology , Cell Differentiation/drug effects , Cell Line, Tumor , Connectin/immunology , Cross Reactions/drug effects , HEK293 Cells , Humans , Lymphocyte Activation/drug effects , Molecular Sequence Data , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Neoplasm Proteins/chemistry , Peptides/chemistry , Protein Engineering , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/drug effects
4.
Cancer Immunol Immunother ; 62(4): 773-85, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23263452

ABSTRACT

NY-ESO-1 and LAGE-1 are cancer testis antigens with an ideal profile for tumor immunotherapy, combining up-regulation in many cancer types with highly restricted expression in normal tissues and sharing a common HLA-A*0201 epitope, 157-165. Here, we present data to describe the specificity and anti-tumor activity of a bifunctional ImmTAC, comprising a soluble, high-affinity T-cell receptor (TCR) specific for NY-ESO-1157-165 fused to an anti-CD3 scFv. This reagent, ImmTAC-NYE, is shown to kill HLA-A2, antigen-positive tumor cell lines, and freshly isolated HLA-A2- and LAGE-1-positive NSCLC cells. Employing time-domain optical imaging, we demonstrate in vivo targeting of fluorescently labelled high-affinity NYESO-specific TCRs to HLA-A2-, NY-ESO-1157-165-positive tumors in xenografted mice. In vivo ImmTAC-NYE efficacy was tested in a tumor model in which human lymphocytes were stably co-engrafted into NSG mice harboring tumor xenografts; efficacy was observed in both tumor prevention and established tumor models using a GFP fluorescence readout. Quantitative RT-PCR was used to analyze the expression of both NY-ESO-1 and LAGE-1 antigens in 15 normal tissues, 5 cancer cell lines, 10 NSCLC, and 10 ovarian cancer samples. Overall, LAGE-1 RNA was expressed at a greater frequency and at higher levels than NY-ESO-1 in the tumor samples. These data support the clinical utility of ImmTAC-NYE as an immunotherapeutic agent for a variety of cancers.


Subject(s)
Antigens, Neoplasm/immunology , Antigens, Surface/immunology , Membrane Proteins/immunology , Receptors, Antigen, T-Cell/immunology , Recombinant Fusion Proteins/pharmacology , T-Lymphocytes/immunology , Animals , Antibodies, Bispecific/immunology , Antibodies, Bispecific/pharmacology , Antigens, Neoplasm/biosynthesis , Antigens, Surface/biosynthesis , CD3 Complex/immunology , Cell Line, Tumor , Epitopes/immunology , Female , HLA-A2 Antigen/immunology , Humans , Immunoglobulin Fragments/immunology , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Melanoma/immunology , Melanoma/metabolism , Membrane Proteins/biosynthesis , Mice , Mice, Inbred NOD , Mice, SCID , Ovarian Neoplasms/immunology , Ovarian Neoplasms/metabolism , Random Allocation , Recombinant Fusion Proteins/immunology , Xenograft Model Antitumor Assays
5.
Nat Med ; 18(6): 980-7, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22561687

ABSTRACT

T cell immunity can potentially eradicate malignant cells and lead to clinical remission in a minority of patients with cancer. In the majority of these individuals, however, there is a failure of the specific T cell receptor (TCR)­mediated immune recognition and activation process. Here we describe the engineering and characterization of new reagents termed immune-mobilizing monoclonal TCRs against cancer (ImmTACs). Four such ImmTACs, each comprising a distinct tumor-associated epitope-specific monoclonal TCR with picomolar affinity fused to a humanized cluster of differentiation 3 (CD3)-specific single-chain antibody fragment (scFv), effectively redirected T cells to kill cancer cells expressing extremely low surface epitope densities. Furthermore, these reagents potently suppressed tumor growth in vivo. Thus, ImmTACs overcome immune tolerance to cancer and represent a new approach to tumor immunotherapy.


Subject(s)
Cytotoxicity, Immunologic , Neoplasms, Experimental/therapy , Receptors, Antigen, T-Cell/physiology , Animals , CD8-Positive T-Lymphocytes/immunology , Humans , Immunologic Memory , Immunotherapy , Interferon-gamma/biosynthesis , Lymphocyte Activation , Mice , Mice, SCID , Neoplasms, Experimental/immunology
6.
J Immunol ; 185(4): 2600-10, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20639478

ABSTRACT

Improving T cell Ags by altering MHC anchor residues is a common strategy used to enhance peptide vaccines, but there has been little assessment of how such modifications affect TCR binding and T cell recognition. In this study, we use surface plasmon resonance and peptide-MHC tetramer binding at the cell surface to demonstrate that changes in primary peptide anchor residues can substantially and unpredictably alter TCR binding. We also demonstrate that the ability of TCRs to differentiate between natural and anchor-modified heteroclitic peptides distinguishes T cells that exhibit a strong preference for either type of Ag. Furthermore, we show that anchor-modified heteroclitic peptides prime T cells with different TCRs compared with those primed with natural Ag. Thus, vaccination with heteroclitic peptides may elicit T cells that exhibit suboptimal recognition of the intended natural Ag and, consequently, impaired functional attributes in vivo. Heteroclitic peptide-based immune interventions therefore require careful evaluation to ensure efficacy in the clinic.


Subject(s)
HLA-A Antigens/immunology , Oligopeptides/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Amino Acid Sequence , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Cytokines/immunology , Cytokines/metabolism , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/metabolism , Flow Cytometry , HLA-A Antigens/genetics , HLA-A Antigens/metabolism , HLA-A2 Antigen , Humans , Mutation , Oligopeptides/genetics , Oligopeptides/metabolism , Peptide Library , Protein Binding/immunology , Receptors, Antigen, T-Cell/metabolism , Surface Plasmon Resonance , T-Lymphocytes/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism
7.
Protein Cell ; 1(12): 1118-27, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21213105

ABSTRACT

Using directed mutagenesis and phage display on a soluble fragment of the human immunoglobulin super-family receptor ILT2 (synonyms: LIR1, MIR7, CD85j), we have selected a range of mutants with binding affinities enhanced by up to 168,000-fold towards the conserved region of major histocompatibility complex (MHC) class I molecules. Produced in a dimeric form, either by chemical cross-linking with bivalent polyethylene glycol (PEG) derivatives or as a genetic fusion with human IgG Fc-fragment, the mutants exhibited a further increase in ligand-binding strength due to the avidity effect, with resident half-times (t(1/2)) on the surface of MHC I-positive cells of many hours. The novel compounds antagonized the interaction of CD8 co-receptor with MHC I in vitro without affecting the peptide-specific binding of T-cell receptors (TCRs). In both cytokine-release assays and cell-killing experiments the engineered receptors inhibited the activation of CD8(+) cytotoxic T lymphocytes (CTLs) in the presence of their target cells, with subnanomolar potency and in a dose-dependent manner. As a selective inhibitor of CD8(+) CTL responses, the engineered high affinity ILT2 receptor presents a new tool for studying the activation mechanism of different subsets of CTLs and could have potential for the development of novel autoimmunity therapies.


Subject(s)
Antigens, CD/genetics , Antigens, CD/pharmacology , Immunologic Factors/genetics , Immunologic Factors/pharmacology , Lymphocyte Activation/immunology , Receptors, Immunologic/genetics , Amino Acid Sequence , Antigens, CD/chemistry , Autoimmunity , Biological Assay , Cell Line , Cytotoxicity, Immunologic/genetics , Cytotoxicity, Immunologic/immunology , Dose-Response Relationship, Immunologic , Humans , Immunoglobulins/immunology , Immunoglobulins/metabolism , Immunologic Factors/chemistry , Kinetics , Leukocyte Immunoglobulin-like Receptor B1 , Lymphocyte Activation/genetics , Major Histocompatibility Complex/genetics , Major Histocompatibility Complex/immunology , Molecular Sequence Data , Molecular Targeted Therapy , Mutagenesis, Site-Directed , Peptide Library , Polyethylene Glycols , Protein Binding/genetics , Protein Binding/immunology , Receptors, Immunologic/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...