Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 19(4): e0301175, 2024.
Article in English | MEDLINE | ID: mdl-38574067

ABSTRACT

BACKGROUND: Canonical α/ß T-cell receptors (TCRs) bind to human leukocyte antigen (HLA) displaying antigenic peptides to elicit T cell-mediated cytotoxicity. TCR-engineered T-cell immunotherapies targeting cancer-specific peptide-HLA complexes (pHLA) are generating exciting clinical responses, but owing to HLA restriction they are only able to target a subset of antigen-positive patients. More recently, evidence has been published indicating that naturally occurring α/ß TCRs can target cell surface proteins other than pHLA, which would address the challenges of HLA restriction. In this proof-of-concept study, we sought to identify and engineer so-called HLA-independent TCRs (HiTs) against the tumor-associated antigen mesothelin. METHODS: Using phage display, we identified a HiT that bound well to mesothelin, which when expressed in primary T cells, caused activation and cytotoxicity. We subsequently engineered this HiT to modulate the T-cell response to varying levels of mesothelin on the cell surface. RESULTS: The isolated HiT shows cytotoxic activity and demonstrates killing of both mesothelin-expressing cell lines and patient-derived xenograft models. Additionally, we demonstrated that HiT-transduced T cells do not require CD4 or CD8 co-receptors and, unlike a TCR fusion construct, are not inhibited by soluble mesothelin. Finally, we showed that HiT-transduced T cells are highly efficacious in vivo, completely eradicating xenografted human solid tumors. CONCLUSION: HiTs can be isolated from fully human TCR-displaying phage libraries against cell surface-expressed antigens. HiTs are able to fully activate primary T cells both in vivo and in vitro. HiTs may enable the efficacy seen with pHLA-targeting TCRs in solid tumors to be translated to cell surface antigens.


Subject(s)
Mesothelin , Neoplasms , Humans , CD8-Positive T-Lymphocytes , Receptors, Antigen, T-Cell , Antigens, Neoplasm/metabolism , Neoplasms/metabolism , Receptors, Antigen, T-Cell, alpha-beta/metabolism , HLA Antigens/metabolism , Histocompatibility Antigens Class II/metabolism , Peptides/metabolism , Histocompatibility Antigens/metabolism
2.
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
3.
Oncoimmunology ; 9(1): 1682381, 2020.
Article in English | MEDLINE | ID: mdl-32002290

ABSTRACT

A substantial obstacle to the success of adoptive T cell-based cancer immunotherapy is the sub-optimal affinity of T-cell receptors (TCRs) for most tumor antigens. Genetically engineered TCRs that have enhanced affinity for specific tumor peptide-MHC complexes may overcome this barrier. However, this enhancement risks increasing weak TCR cross-reactivity to other antigens expressed by normal tissues, potentially leading to clinical toxicities. To reduce the risk of such adverse clinical outcomes, we have developed an extensive preclinical testing strategy, involving potency testing using 2D and 3D human cell cultures and primary tumor material, and safety testing using human primary cell and cell-line cross-reactivity screening and molecular analysis to predict peptides recognized by the affinity-enhanced TCR. Here, we describe this strategy using a developmental T-cell therapy, ADP-A2M4, which recognizes the HLA-A2-restricted MAGE-A4 peptide GVYDGREHTV. ADP-A2M4 demonstrated potent anti-tumor activity in the absence of major off-target cross-reactivity against a range of human primary cells and cell lines. Identification and characterization of peptides recognized by the affinity-enhanced TCR also revealed no cross-reactivity. These studies demonstrated that this TCR is highly potent and without major safety concerns, and as a result, this TCR is now being investigated in two clinical trials (NCT03132922, NCT04044768).


Subject(s)
Immunotherapy, Adoptive , Receptors, Antigen, T-Cell , Antigens, Neoplasm , Cell- and Tissue-Based Therapy , Humans , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes
SELECTION OF CITATIONS
SEARCH DETAIL
...