Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Front Immunol ; 13: 1039226, 2022.
Article in English | MEDLINE | ID: mdl-36569934

ABSTRACT

Background: Cancer neoantigens are important targets of cancer immunotherapy and neoantigen vaccines are currently in development in pancreatic ductal adenocarcinoma (PDAC) and other cancer types. Immune regulatory mechanisms in pancreatic cancer may limit the efficacy of neoantigen vaccines. Targeting immune checkpoint signaling pathways in PDAC may improve the efficacy of neoantigen vaccines. Methods: We used KPC4580P, an established model of PDAC, to test whether neoantigen vaccines can generate therapeutic efficacy against PDAC. We focused on two immunogenic neoantigens associated with genetic alterations in the CAR12 and CDK12 genes. We tested a neoantigen vaccine comprised of two 20-mer synthetic long peptides and poly IC, a Toll-like receptor (TLR) agonist. We investigated the ability of neoantigen vaccine alone, or in combination with PD-1 and TIGIT signaling blockade to impact tumor growth. We also assessed the impact of TIGIT signaling on T cell responses in human PDAC. Results: Neoantigen vaccines induce neoantigen-specific T cell responses in tumor-bearing mice and slow KPC4580P tumor growth. However, KPC4580P tumors express high levels of PD-L1 and the TIGIT ligand, CD155. A subset of neoantigen-specific T cells in KPC4580P tumors are dysfunctional, and express high levels of TIGIT. PD-1 and TIGIT signaling blockade in vivo reverses T cell dysfunction and enhances neoantigen vaccine-induced T cell responses and tumor regression. In human translational studies, TIGIT signaling blockade in vitro enhances neoantigen-specific T cell function following vaccination. Conclusions: Taken together, preclinical and human translational studies support testing neoantigen vaccines in combination with therapies targeting the PD-1 and TIGIT signaling pathways in patients with PDAC.


Subject(s)
Cancer Vaccines , Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Mice , Animals , Programmed Cell Death 1 Receptor , Antigens, Neoplasm , Pancreatic Neoplasms/genetics , Carcinoma, Pancreatic Ductal/genetics , Peptides/therapeutic use , Receptors, Immunologic/therapeutic use , Pancreatic Neoplasms
2.
Genome Med ; 13(1): 56, 2021 04 21.
Article in English | MEDLINE | ID: mdl-33879241

ABSTRACT

BACKGROUND: Preclinical studies and early clinical trials have shown that targeting cancer neoantigens is a promising approach towards the development of personalized cancer immunotherapies. DNA vaccines can be rapidly and efficiently manufactured and can integrate multiple neoantigens simultaneously. We therefore sought to optimize the design of polyepitope DNA vaccines and test optimized polyepitope neoantigen DNA vaccines in preclinical models and in clinical translation. METHODS: We developed and optimized a DNA vaccine platform to target multiple neoantigens. The polyepitope DNA vaccine platform was first optimized using model antigens in vitro and in vivo. We then identified neoantigens in preclinical breast cancer models through genome sequencing and in silico neoantigen prediction pipelines. Optimized polyepitope neoantigen DNA vaccines specific for the murine breast tumor E0771 and 4T1 were designed and their immunogenicity was tested in vivo. We also tested an optimized polyepitope neoantigen DNA vaccine in a patient with metastatic pancreatic neuroendocrine tumor. RESULTS: Our data support an optimized polyepitope neoantigen DNA vaccine design encoding long (≥20-mer) epitopes with a mutant form of ubiquitin (Ubmut) fused to the N-terminus for antigen processing and presentation. Optimized polyepitope neoantigen DNA vaccines were immunogenic and generated robust neoantigen-specific immune responses in mice. The magnitude of immune responses generated by optimized polyepitope neoantigen DNA vaccines was similar to that of synthetic long peptide vaccines specific for the same neoantigens. When combined with immune checkpoint blockade therapy, optimized polyepitope neoantigen DNA vaccines were capable of inducing antitumor immunity in preclinical models. Immune monitoring data suggest that optimized polyepitope neoantigen DNA vaccines are capable of inducing neoantigen-specific T cell responses in a patient with metastatic pancreatic neuroendocrine tumor. CONCLUSIONS: We have developed and optimized a novel polyepitope neoantigen DNA vaccine platform that can target multiple neoantigens and induce antitumor immune responses in preclinical models and neoantigen-specific responses in clinical translation.


Subject(s)
Antigens, Neoplasm/immunology , Epitopes/immunology , Immunity , Translational Research, Biomedical , Vaccines, DNA/immunology , Adult , Animals , Antigen Presentation/immunology , Cell Proliferation , Disease Models, Animal , Female , HeLa Cells , Humans , Immune Checkpoint Inhibitors , Immunotherapy , Male , Mammary Neoplasms, Animal/pathology , Mice, Inbred C57BL , Neoplasm Metastasis , Neuroendocrine Tumors/immunology , Neuroendocrine Tumors/pathology , Peptides/immunology , T-Lymphocytes/immunology
3.
Eur J Immunol ; 44(7): 1936-46, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24723377

ABSTRACT

We used a newly generated T-cell receptor mimic monoclonal antibody (TCRm MAb) that recognizes a known nonself immunodominant peptide epitope from West Nile virus (WNV) NS4B protein to investigate epitope presentation after virus infection in C57BL/6 mice. Previous studies suggested that peptides of different length, either SSVWNATTAI (10-mer) or SSVWNATTA (9-mer) in complex with class I MHC antigen H-2D(b) , were immunodominant after WNV infection. Our data establish that both peptides are presented on the cell surface after WNV infection and that CD8(+) T cells can detect 10- and 9-mer length variants similarly. This result varies from the idea that a given T-cell receptor (TCR) prefers a single peptide length bound to its cognate class I MHC. In separate WNV infection studies with the TCRm MAb, we show that in vivo the 10-mer was presented on the surface of uninfected and infected CD8α(+) CD11c(+) dendritic cells, which suggests the use of direct and cross-presentation pathways. In contrast, CD11b(+) CD11c(-) cells bound the TCRm MAb only when they were infected. Our study demonstrates that TCR recognition of peptides is not limited to certain peptide lengths and that TCRm MAbs can be used to dissect the cell-type specific mechanisms of antigen presentation in vivo.


Subject(s)
Dendritic Cells/immunology , Immunodominant Epitopes , Receptors, Antigen, T-Cell/physiology , West Nile virus/immunology , Animals , CD11b Antigen/analysis , CD11c Antigen/analysis , CD8-Positive T-Lymphocytes/immunology , Mice , Mice, Inbred C57BL , Viral Nonstructural Proteins/immunology
4.
Mol Immunol ; 55(2): 123-5, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23200143

ABSTRACT

The mature conformation of major histocompatibility complex class I (MHC-I) proteins depends on the presence of bound peptides, permitting recognition at the cell surface by CD8(+) T lymphocytes. Newly synthesized MHC-I molecules in the endoplasmic reticulum are maintained in a peptide-receptive (PR) transition state by several chaperones until they are released concomitant with the loading of peptides. By determining the crystallographic structure of a region of an MHC-I molecule that is recognized by a unique monoclonal antibody and comparing this with docking and molecular dynamics simulations with the whole molecule, we demonstrate the movement of a hinged unit supporting the part of the binding groove that interacts with the amino terminal residues of the bound peptide. This unit contains a conserved 310 helix that flips from an exposed "open" position in the PR form to a "closed" position in the peptide-loaded (PL) mature molecule. These analyses indicate how this segment of the MHC-I molecule moves to help establish the A and B pockets critical for tight peptide binding and the stable structure required for antigen presentation and T cell recognition at the cell surface.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Histocompatibility Antigens Class I/immunology , Molecular Dynamics Simulation , Receptors, Pattern Recognition/immunology , Receptors, Pattern Recognition/ultrastructure , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Antigen Presentation , Crystallography, X-Ray , Histocompatibility Antigens Class I/metabolism , Humans , Protein Binding , Protein Structure, Tertiary
5.
J Immunol ; 189(3): 1391-9, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22753930

ABSTRACT

MHC class I (MHC-I) proteins of the adaptive immune system require antigenic peptides for maintenance of mature conformation and immune function via specific recognition by MHC-I-restricted CD8(+) T lymphocytes. New MHC-I molecules in the endoplasmic reticulum are held by chaperones in a peptide-receptive (PR) transition state pending release by tightly binding peptides. In this study, we show, by crystallographic, docking, and molecular dynamics methods, dramatic movement of a hinged unit containing a conserved 3(10) helix that flips from an exposed "open" position in the PR transition state to a "closed" position with buried hydrophobic side chains in the peptide-loaded mature molecule. Crystallography of hinged unit residues 46-53 of murine H-2L(d) MHC-I H chain, complexed with mAb 64-3-7, demonstrates solvent exposure of these residues in the PR conformation. Docking and molecular dynamics predict how this segment moves to help form the A and B pockets crucial for the tight peptide binding needed for stability of the mature peptide-loaded conformation, chaperone dissociation, and Ag presentation.


Subject(s)
H-2 Antigens/metabolism , Molecular Dynamics Simulation , Peptide Fragments/metabolism , Amino Acid Sequence , Animals , Crystallography, X-Ray , H-2 Antigens/chemistry , Histocompatibility Antigen H-2D , Humans , Ligands , Mice , Molecular Sequence Data , Peptide Fragments/chemistry , Structure-Activity Relationship , beta 2-Microglobulin/chemistry , beta 2-Microglobulin/metabolism
6.
Immunol Res ; 32(1-3): 109-21, 2005.
Article in English | MEDLINE | ID: mdl-16106063

ABSTRACT

Generation of CD8 T-cell responses to pathogens and tumors requires optimal expression of class I major histocompatibility complex/peptide complexes, which, in turn, is dependent on host cellular processing events and subject to interference by pathogens. To create a stable structure that is more immunogenic and resistant to immune evasion pathways, we have engineered class I molecules as single-chain trimers (SCTs), with flexible linkers connecting peptide, beta2m, and heavy chain. Herein we extend our earlier studies with SCTs to the K(b) ligand derived from vesicular stomatitis virus (VSV) to characterize further SCTs as probes of immune function as well as their potential in immunotherapy. The VSVp-beta2m-K(b) SCTs were remarkably stable at the cell surface, and immunization with DNA encoding SCTs elicited complex-specific antibody. In addition, SCTs were detected by cytotoxic T-lymphocytes specific for the native molecule, and the covalently bound peptide was highly resistant to displacement by exogenous peptide. SCTs can also prime CD8 T-cells in vivo that recognize the native molecule. Furthermore, SCTs were resistant to downregulation by the immune evasion protein mK3 of gamma herpesvirus 68. Moreover, owing to their preassembled nature, SCTs should be resistant to other immune evasion proteins that restrict peptide supply. Thus, SCTs possess therapeutic potential both for prophylactic treatment and for the treatment of ongoing infection.


Subject(s)
Histocompatibility Antigens Class I/chemistry , Animals , Antibody Specificity , CD8-Positive T-Lymphocytes/immunology , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Humans , In Vitro Techniques , Ligands , Mice , Models, Molecular , Protein Engineering , Protein Structure, Quaternary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Vaccines, DNA/genetics , Vaccines, DNA/immunology
7.
J Immunol ; 169(6): 3105-11, 2002 Sep 15.
Article in English | MEDLINE | ID: mdl-12218127

ABSTRACT

The two mouse MHC class I alleles, L(d) and L(q), share complete amino acid sequence identity except in the alpha2 domain, where they differ at six positions. Despite their similarity, L(q) has a stronger association with beta2-microglobulin (beta2m), is expressed at higher levels on the cell surface, demonstrates an increased cell surface half-life, and has fewer open forms on the cell surface than L(d). To determine the basis for their phenotypic differences, L(d) molecules containing chimeric L(d)-L(q) alpha2 domains were characterized, and these analyses implicated residue 97 (L(d)Trp and L(q)Arg) as the polymorphic site responsible for the disparity in beta2m association between the two alleles. Single substitution analysis at this site (L(d)W97R and L(q)R97W) confirmed this. Furthermore, the L(d)W97R mutant molecule has a longer cell surface half-life than either L(q) or L(d), and fewer open forms of L(d)W97R are observed on the cell surface. In addition, both L(d)W97R and L(q) possess decreased binding affinity for the L(d)-restricted tum(-) P91A(14-22) peptide compared with L(d). Collectively, these results and the known location of Trp(97) in the peptide binding cleft of L(d) strongly suggest that the substitution of Arg for Trp(97) in L(d) alters the peptide binding cleft, increasing its affinity for endogenous peptides, which results in greater cell surface stability and better retention of beta2m. Furthermore, these results imply that Trp(97) plays an important role in the ability of L(d) to efficiently participate in alternative MHC class I Ag presentation pathways.


Subject(s)
Amino Acid Substitution/immunology , Epitopes/genetics , Epitopes/immunology , H-2 Antigens/genetics , Peptides/genetics , Peptides/immunology , Polymorphism, Genetic/immunology , beta 2-Microglobulin/metabolism , Alleles , Amino Acid Substitution/genetics , Animals , Arginine/genetics , Cell Line , Cell Membrane/genetics , Cell Membrane/immunology , Cell Membrane/metabolism , Epitopes/metabolism , H-2 Antigens/metabolism , Half-Life , Histocompatibility Antigen H-2D , Isoantigens/genetics , Isoantigens/metabolism , Mice , Peptides/metabolism , Protein Binding/genetics , Protein Binding/immunology , Transfection , Tryptophan/genetics
8.
J Virol ; 76(6): 2796-803, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11861847

ABSTRACT

To persist in the presence of an active immune system, viruses encode proteins that decrease expression of major histocompatibility complex class I molecules by using a variety of mechanisms. For example, murine gamma-2 herpesvirus 68 expresses the K3 protein, which causes the rapid turnover of nascent class I molecules. In this report we show that certain mouse class I alleles are more susceptible than others to K3-mediated down regulation. Prior to their rapid degradation, class I molecules in K3-expressing cells exhibit impaired assembly with beta(2)-microglobulin. Furthermore, K3 is detected predominantly in association with class I molecules lacking assembly with high-affinity peptides, including class I molecules associated with the peptide loading complex TAP/tapasin/calreticulin. The detection of K3 with class I assembly intermediates raises the possibility that molecular chaperones involved in class I assembly are involved in K3-mediated class I regulation.


Subject(s)
Down-Regulation , Gammaherpesvirinae/physiology , Histocompatibility Antigens Class I/physiology , Peptides/metabolism , Viral Proteins/physiology , beta 2-Microglobulin/metabolism , Animals , Gammaherpesvirinae/pathogenicity , Herpesviridae Infections/virology , Histocompatibility Antigens Class I/chemistry , Histocompatibility Antigens Class I/genetics , L Cells , Mice , Protein Folding
SELECTION OF CITATIONS
SEARCH DETAIL
...