Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 115
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 17(1): 87-94, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26523866

ABSTRACT

The T cell antigen receptor (TCR)-peptide-major histocompatibility complex (MHC) interface is composed of conserved and diverse regions, yet the relative contribution of each in shaping recognition by T cells remains unclear. Here we isolated cross-reactive peptides with limited homology, which allowed us to compare the structural properties of nine peptides for a single TCR-MHC pair. The TCR's cross-reactivity was rooted in highly similar recognition of an apical 'hot-spot' position in the peptide with tolerance of sequence variation at ancillary positions. Furthermore, we found a striking structural convergence onto a germline-mediated interaction between the TCR CDR1α region and the MHC α2 helix in twelve TCR-peptide-MHC complexes. Our studies suggest that TCR-MHC germline-mediated constraints, together with a focus on a small peptide hot spot, might place limits on peptide antigen cross-reactivity.


Subject(s)
Antigens/immunology , Cross Reactions/immunology , Lymphocyte Activation/immunology , Major Histocompatibility Complex/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , Amino Acid Sequence , Animals , Antigens/chemistry , Crystallography, X-Ray , Humans , Models, Molecular , Molecular Sequence Data , Peptides/immunology , Protein Binding/immunology , Protein Conformation , Receptors, Antigen, T-Cell, alpha-beta/chemistry
2.
Immunity ; 44(6): 1444-54, 2016 06 21.
Article in English | MEDLINE | ID: mdl-27332733

ABSTRACT

Genetically modified T cells expressing chimeric antigen receptors (CARs) demonstrate robust responses against lineage restricted, non-essential targets in hematologic cancers. However, in solid tumors, the full potential of CAR T cell therapy is limited by the availability of cell surface antigens with sufficient cancer-specific expression. The majority of CAR targets have been normal self-antigens on dispensable hematopoietic tissues or overexpressed shared antigens. Here, we established that abnormal self-antigens can serve as targets for tumor rejection. We developed a CAR that recognized cancer-associated Tn glycoform of MUC1, a neoantigen expressed in a variety of cancers. Anti-Tn-MUC1 CAR T cells demonstrated target-specific cytotoxicity and successfully controlled tumor growth in xenograft models of T cell leukemia and pancreatic cancer. These findings demonstrate the therapeutic efficacy of CAR T cells directed against Tn-MUC1 and present aberrantly glycosylated antigens as a novel class of targets for tumor therapy with engineered T cells.


Subject(s)
Adenocarcinoma/therapy , Epitopes, T-Lymphocyte/immunology , Immunotherapy/methods , Mucin-1/immunology , T-Lymphocytes/physiology , Adenocarcinoma/immunology , Animals , Cell Line, Tumor , Cytotoxicity, Immunologic , Genetic Engineering , Glycosylation , Humans , Jurkat Cells , Mice , Mice, Inbred Strains , Mucin-1/chemistry , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Xenograft Model Antitumor Assays
3.
Proc Natl Acad Sci U S A ; 117(26): 15148-15159, 2020 06 30.
Article in English | MEDLINE | ID: mdl-32541028

ABSTRACT

The potency of adoptive T cell therapies targeting the cell surface antigen CD19 has been demonstrated in hematopoietic cancers. It has been difficult to identify appropriate targets in nonhematopoietic tumors, but one class of antigens that have shown promise is aberrant O-glycoprotein epitopes. It has long been known that dysregulated synthesis of O-linked (threonine or serine) sugars occurs in many cancers, and that this can lead to the expression of cell surface proteins containing O-glycans comprised of a single N-acetylgalactosamine (GalNAc, known as Tn antigen) rather than the normally extended carbohydrate. Previously, we used the scFv fragment of antibody 237 as a chimeric antigen receptor (CAR) to mediate recognition of mouse tumor cells that bear its cognate Tn-glycopeptide epitope in podoplanin, also called OTS8. Guided by the structure of the 237 Fab:Tn-OTS8-glycopeptide complex, here we conducted a deep mutational scan showing that residues flanking the Tn-glycan contributed significant binding energy to the interaction. Design of 237-scFv libraries in the yeast display system allowed us to isolate scFv variants with higher affinity for Tn-OTS8. Selection with a noncognate human antigen, Tn-MUC1, yielded scFv variants that were broadly reactive with multiple Tn-glycoproteins. When configured as CARs, engineered T cells expressing these scFv variants showed improved activity against mouse and human cancer cell lines defective in O-linked glycosylation. This strategy provides CARs with Tn-peptide specificities, all based on a single scFv scaffold, that allows the same CAR to be tested for toxicity in mice and efficacy against mouse and human tumors.


Subject(s)
Antigens, Tumor-Associated, Carbohydrate/immunology , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/physiology , Amino Acid Sequence , Animals , Antibodies , Cell Line, Tumor , Directed Molecular Evolution , Epitopes/genetics , Humans , Mice , Models, Molecular , Mutation , Protein Conformation , Receptors, Chimeric Antigen/genetics
4.
Proc Natl Acad Sci U S A ; 116(8): 3136-3145, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30728302

ABSTRACT

Successful efforts to activate T cells capable of recognizing weak cancer-associated self-antigens have employed altered peptide antigens to activate T cell responses capable of cross-reacting on native tumor-associated self. A limitation of this approach is the requirement for detailed knowledge about the altered self-peptide ligands used in these vaccines. In the current study we considered allorecognition as an approach for activating CTL capable of recognizing weak or self-antigens in the context of self-MHC. Nonself antigen-presenting molecules typically contain polymorphisms that influence interactions with the bound peptide and TCR interface. Recognition of these nonself structures results in peptide-dependent alloimmunity. Alloreactive T cells target their inducing alloantigens as well as third-party alloantigens but generally fail to target self-antigens. Certain residues located on the alpha-1/2 domains of class I antigen-presenting molecules primarily interface with TCR. These residues are more conserved within and across species than are residues that determine peptide antigen binding properties. Class I variants designed with amino acid substitutions at key positions within the conserved helical structures are shown to provide strong activating signals to alloreactive CD8 T cells while avoiding changes in naturally bound peptide ligands. Importantly, CTL activated in this manner can break self-tolerance by reacting to self-peptides presented by native MHC. The ability to activate self-tolerant T cells capable of cross-reacting on self-peptide-MHC in vivo represents an approach for inducing autoimmunity, with possible application in cancer vaccines.


Subject(s)
Antigen Presentation/immunology , Cytotoxicity, Immunologic , Histocompatibility Antigens Class I/immunology , T-Lymphocytes, Cytotoxic/immunology , Amino Acid Sequence/genetics , Animals , CD8-Positive T-Lymphocytes/immunology , Humans , Immune Tolerance , Ligands , Lymphocyte Activation/immunology , Mice , Peptides/genetics , Peptides/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology
5.
Biochemistry ; 59(43): 4163-4175, 2020 11 03.
Article in English | MEDLINE | ID: mdl-33074657

ABSTRACT

T cell receptors (TCRs) orchestrate cellular immunity by recognizing peptides presented by a range of major histocompatibility complex (MHC) proteins. Naturally occurring TCRs bind the composite peptide/MHC surface, recognizing peptides that are structurally and chemically compatible with the TCR binding site. Here we describe a molecularly evolved TCR variant that binds the human class I MHC protein HLA-A2 independent of the bound peptide, achieved by a drastic perturbation of the TCR binding geometry that places the molecule far from the peptide binding groove. This unique geometry is unsupportive of normal T cell signaling. A substantial divergence between affinity measurements in solution and in two dimensions between proximal cell membranes leads us to attribute the lack of signaling to steric hindrance that limits binding in the confines of a cell-cell interface. Our results provide an example of how receptor binding geometry can impact T cell function and provide further support for the view that germline-encoded residues in TCR binding loops evolved to drive productive TCR recognition and signaling.


Subject(s)
Receptors, Antigen, T-Cell/metabolism , Binding Sites , HLA-A Antigens/metabolism , Humans , Major Histocompatibility Complex/genetics , Major Histocompatibility Complex/physiology , Protein Binding , Protein Conformation
6.
Immunity ; 35(5): 681-93, 2011 Nov 23.
Article in English | MEDLINE | ID: mdl-22101157

ABSTRACT

T cell receptor (TCR) engagement of peptide-major histocompatibility complex (pMHC) is essential to adaptive immunity, but it is unknown whether TCR signaling responses are influenced by the binding topology of the TCR-peptide-MHC complex. We developed yeast-displayed pMHC libraries that enabled us to identify new peptide sequences reactive with a single TCR. Structural analysis showed that four peptides bound to the TCR with distinct 3D and 2D affinities using entirely different binding chemistries. Three of the peptides that shared a common docking mode, where key TCR-MHC germline interactions are preserved, induced TCR signaling. The fourth peptide failed to induce signaling and was recognized in a substantially different TCR-MHC binding mode that apparently exceeded geometric tolerances compatible with signaling. We suggest that the stereotypical TCR-MHC docking paradigm evolved from productive signaling geometries and that TCR signaling can be modulated by peptides that are recognized in alternative TCR-pMHC binding orientations.


Subject(s)
Histocompatibility Antigens Class I/chemistry , Peptides/chemistry , Peptides/immunology , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/immunology , Signal Transduction , Amino Acid Motifs/immunology , Amino Acid Sequence , Animals , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/metabolism , Humans , Lymphocyte Activation/immunology , Mice , Models, Molecular , Peptide Library , Peptides/metabolism , Protein Binding/immunology , Protein Conformation , Receptors, Antigen, T-Cell/metabolism , Reproducibility of Results , Sequence Alignment , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
7.
Crit Rev Immunol ; 39(2): 105-122, 2019.
Article in English | MEDLINE | ID: mdl-31679251

ABSTRACT

The past decade has seen enormous progress in cancer immunotherapy. Checkpoint inhibitors are a class of immunotherapy that act to recruit endogenous T cells of a patient's immune system against cancer-associated peptide- MHC antigens. In this process, mutated antigenic peptides referred to as neoantigens often serve as the target on cancer cells that are recognized by the T cell receptor (TCR) on endogenous T cells. Another successful immunotherapy has involved adoptive T cell therapy, where therapeutic doses of T cells expressing a gene for an anti-cancer receptor are delivered to a patient. This approach has been used primarily against hematopoietic cancers using synthetic receptors called chimeric antigen receptors (CARs). CARs typically contain an antibody fragment (single-chain Fv, scFv) against a cancer cell surface antigen such as the B cell molecule CD19. While therapeutic CARs (and full antibodies) target antigens expressed on cell surfaces, TCRs can target a much larger array of intracellular proteins by binding to any cellular peptide associated with an MHC product. These cancer targets include self-peptides from aberrantly expressed/overexpressed proteins or neoantigens. In this review, we discuss the use of TCRs in adoptive T cell therapy and their target antigens. We focus on two properties that impact sensitivity, potency, and possible toxic cross-reactivity of TCR-mediated therapy: (1) the affinity of the TCR for the target antigen, and (2) the density of the target antigen. Finally, we provide a comprehensive listing of the current clinical trials that involve TCRs in adoptive T cell cancer therapy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunotherapy, Adoptive/methods , Neoplasms/therapy , Receptors, Antigen, T-Cell/genetics , Receptors, Chimeric Antigen/genetics , Animals , Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/transplantation , Cytotoxicity, Immunologic , Genetic Engineering , Humans , Neoplasms/immunology
8.
J Immunol ; 200(3): 1088-1100, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29288199

ABSTRACT

Adoptive T cell therapies have achieved significant clinical responses, especially in hematopoietic cancers. Two types of receptor systems have been used to redirect the activity of T cells, normal heterodimeric TCRs or synthetic chimeric Ag receptors (CARs). TCRs recognize peptide-HLA complexes whereas CARs typically use an Ab-derived single-chain fragments variable that recognizes cancer-associated cell-surface Ags. Although both receptors mediate diverse effector functions, a quantitative comparison of the sensitivity and signaling capacity of TCRs and CARs has been limited due to their differences in affinities and ligands. In this study we describe their direct comparison by using TCRs that could be formatted either as conventional αß heterodimers, or as single-chain fragments variable constructs linked to CD3ζ and CD28 signaling domains or to CD3ζ alone. Two high-affinity TCRs (KD values of ∼50 and 250 nM) against MART1/HLA-A2 or WT1/HLA-A2 were used, allowing MART1 or WT1 peptide titrations to easily assess the impact of Ag density. Although CARs were expressed at higher surface levels than TCRs, they were 10-100-fold less sensitive, even in the absence of the CD8 coreceptor. Mathematical modeling demonstrated that lower CAR sensitivity could be attributed to less efficient signaling kinetics. Furthermore, reduced cytokine secretion observed at high Ag density for both TCRs and CARs suggested a role for negative regulators in both systems. Interestingly, at high Ag density, CARs also mediated greater maximal release of some cytokines, such as IL-2 and IL-6. These results have implications for the next-generation design of receptors used in adoptive T cell therapies.


Subject(s)
Antibody Affinity/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , MART-1 Antigen/immunology , Receptors, Antigen, T-Cell/immunology , WT1 Proteins/immunology , Antigens, Tumor-Associated, Carbohydrate/immunology , HLA Antigens/immunology , Humans , Lymphocyte Activation/immunology , Mutant Chimeric Proteins/immunology
9.
J Biol Chem ; 293(5): 1820-1834, 2018 02 02.
Article in English | MEDLINE | ID: mdl-29229779

ABSTRACT

Most affinity-maturation campaigns for antibodies and T-cell receptors (TCRs) operate on the residues at the binding site, located within the loops known as complementarity-determining regions (CDRs). Accordingly, mutations in contact residues, or so-called "second shell" residues, that increase affinity are typically identified by directed evolution involving combinatorial libraries. To determine the impact of residues located at a distance from the binding site, here we used single-codon libraries of both CDR and non-CDR residues to generate a deep mutational scan of a human TCR against the cancer antigen MART-1·HLA-A2. Non-CDR residues included those at the interface of the TCR variable domains (Vα and Vß) and surface-exposed framework residues. Mutational analyses showed that both Vα/Vß interface and CDR residues were important in maintaining binding to MART-1·HLA-A2, probably due to either structural requirements for proper Vα/Vß association or direct contact with the ligand. More surprisingly, many Vα/Vß interface substitutions yielded improved binding to MART-1·HLA-A2. To further explore this finding, we constructed interface libraries and selected them for improved stability or affinity. Among the variants identified, one conservative substitution (F45ßY) was most prevalent. Further analysis of F45ßY showed that it enhanced thermostability and increased affinity by 60-fold. Thus, introducing a single hydroxyl group at the Vα/Vß interface, at a significant distance from the TCR·peptide·MHC-binding site, remarkably affected ligand binding. The variant retained a high degree of specificity for MART-1·HLA-A2, indicating that our approach provides a general strategy for engineering improvements in either soluble or cell-based TCRs for therapeutic purposes.


Subject(s)
Complementarity Determining Regions/chemistry , HLA-A2 Antigen/chemistry , MART-1 Antigen/chemistry , Binding Sites , Complementarity Determining Regions/genetics , Complementarity Determining Regions/immunology , HLA-A2 Antigen/genetics , HLA-A2 Antigen/immunology , Humans , MART-1 Antigen/genetics , MART-1 Antigen/immunology , Protein Binding , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Saccharomyces cerevisiae
SELECTION OF CITATIONS
SEARCH DETAIL