Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Clin Invest ; 131(16)2021 08 16.
Article in English | MEDLINE | ID: mdl-34396985

ABSTRACT

Although immune-checkpoint inhibitors (ICIs) have been a remarkable advancement in bladder cancer treatment, the response rate to single-agent ICIs remains suboptimal. There has been substantial interest in the use of epigenetic agents to enhance ICI efficacy, although precisely how these agents potentiate ICI response has not been fully elucidated. We identified entinostat, a selective HDAC1/3 inhibitor, as a potent antitumor agent in our immune-competent bladder cancer mouse models (BBN963 and BBN966). We demonstrate that entinostat selectively promoted immune editing of tumor neoantigens, effectively remodeling the tumor immune microenvironment, resulting in a robust antitumor response that was cell autonomous, dependent upon antigen presentation, and associated with increased numbers of neoantigen-specific T cells. Finally, combination treatment with anti-PD-1 and entinostat led to complete responses and conferred long-term immunologic memory. Our work defines a tumor cell-autonomous mechanism of action for entinostat and a strong preclinical rationale for the combined use of entinostat and PD-1 blockade in bladder cancer.


Subject(s)
Antigens, Neoplasm/drug effects , Benzamides/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Pyridines/pharmacology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Animals , Antigens, Neoplasm/immunology , Antineoplastic Agents, Immunological/pharmacology , Cell Line, Tumor , Humans , Immunity/drug effects , Immunocompetence/drug effects , Immunotherapy/methods , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , T-Lymphocytes/drug effects , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/immunology
2.
Cancer Immunol Res ; 7(10): 1591-1604, 2019 10.
Article in English | MEDLINE | ID: mdl-31515258

ABSTRACT

Current tumor neoantigen calling algorithms primarily rely on epitope/major histocompatibility complex (MHC) binding affinity predictions to rank and select for potential epitope targets. These algorithms do not predict for epitope immunogenicity using approaches modeled from tumor-specific antigen data. Here, we describe peptide-intrinsic biochemical features associated with neoantigen and minor histocompatibility mismatch antigen immunogenicity and present a gradient boosting algorithm for predicting tumor antigen immunogenicity. This algorithm was validated in two murine tumor models and demonstrated the capacity to select for therapeutically active antigens. Immune correlates of neoantigen immunogenicity were studied in a pan-cancer data set from The Cancer Genome Atlas and demonstrated an association between expression of immunogenic neoantigens and immunity in colon and lung adenocarcinomas. Lastly, we present evidence for expression of an out-of-frame neoantigen that was capable of driving antitumor cytotoxic T-cell responses. With the growing clinical importance of tumor vaccine therapies, our approach may allow for better selection of therapeutically relevant tumor-specific antigens, including nonclassic out-of-frame antigens capable of driving antitumor immunity.


Subject(s)
Algorithms , Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Epitopes, T-Lymphocyte/immunology , Machine Learning , Neoplasms/immunology , Peptide Fragments/immunology , Animals , Computational Biology/methods , Female , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class II/immunology , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasms/drug therapy , Neoplasms/pathology , T-Lymphocytes, Cytotoxic/immunology
3.
J Clin Invest ; 128(11): 4804-4820, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30137025

ABSTRACT

Human endogenous retroviruses (hERVs) are remnants of exogenous retroviruses that have integrated into the genome throughout evolution. We developed a computational workflow, hervQuant, which identified more than 3,000 transcriptionally active hERVs within The Cancer Genome Atlas (TCGA) pan-cancer RNA-Seq database. hERV expression was associated with clinical prognosis in several tumor types, most significantly clear cell renal cell carcinoma (ccRCC). We explored two mechanisms by which hERV expression may influence the tumor immune microenvironment in ccRCC: (i) RIG-I-like signaling and (ii) retroviral antigen activation of adaptive immunity. We demonstrated the ability of hERV signatures associated with these immune mechanisms to predict patient survival in ccRCC, independent of clinical staging and molecular subtyping. We identified potential tumor-specific hERV epitopes with evidence of translational activity through the use of a ccRCC ribosome profiling (Ribo-Seq) dataset, validated their ability to bind HLA in vitro, and identified the presence of MHC tetramer-positive T cells against predicted epitopes. hERV sequences identified through this screening approach were significantly more highly expressed in ccRCC tumors responsive to treatment with programmed death receptor 1 (PD-1) inhibition. hervQuant provides insights into the role of hERVs within the tumor immune microenvironment, as well as evidence that hERV expression could serve as a biomarker for patient prognosis and response to immunotherapy.


Subject(s)
Carcinoma, Renal Cell , Endogenous Retroviruses , Immunotherapy , Kidney Neoplasms , Tumor Microenvironment , Carcinoma, Renal Cell/diagnosis , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/therapy , Endogenous Retroviruses/genetics , Endogenous Retroviruses/immunology , Humans , Kidney Neoplasms/diagnosis , Kidney Neoplasms/genetics , Kidney Neoplasms/immunology , Kidney Neoplasms/therapy , Prognosis , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
4.
Cancer Res ; 78(14): 3954-3968, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29784854

ABSTRACT

High-grade urothelial cancer contains intrinsic molecular subtypes that exhibit differences in underlying tumor biology and can be divided into luminal-like and basal-like subtypes. We describe here the first subtype-specific murine models of bladder cancer and show that Upk3a-CreERT2; Trp53L/L; PtenL/L; Rosa26LSL-Luc (UPPL, luminal-like) and BBN (basal-like) tumors are more faithful to human bladder cancer than the widely used MB49 cells. Following engraftment into immunocompetent C57BL/6 mice, BBN tumors were more responsive to PD-1 inhibition than UPPL tumors. Responding tumors within the BBN model showed differences in immune microenvironment composition, including increased ratios of CD8+:CD4+ and memory:regulatory T cells. Finally, we predicted and confirmed immunogenicity of tumor neoantigens in each model. These UPPL and BBN models will be a valuable resource for future studies examining bladder cancer biology and immunotherapy.Significance: This work establishes human-relevant mouse models of bladder cancer. Cancer Res; 78(14); 3954-68. ©2018 AACR.


Subject(s)
Antigens, Neoplasm/immunology , Carcinoma/immunology , Immunocompetence/immunology , Urologic Neoplasms/immunology , Urothelium/immunology , Animals , Disease Models, Animal , Humans , Immunotherapy/methods , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/immunology , T-Lymphocytes/immunology , Tumor Microenvironment/immunology , Urinary Bladder Neoplasms/immunology
5.
PLoS One ; 13(3): e0193832, 2018.
Article in English | MEDLINE | ID: mdl-29513764

ABSTRACT

The ability to non-invasively monitor tumor-infiltrating T cells in vivo could provide a powerful tool to visualize and quantify tumor immune infiltrates. For non-invasive evaluations in vivo, an anti-CD3 mAb was modified with desferrioxamine (DFO) and radiolabeled with zirconium-89 (Zr-89 or 89Zr). Radiolabeled 89Zr-DFO-anti-CD3 was tested for T cell detection using positron emission tomography (PET) in both healthy mice and mice bearing syngeneic bladder cancer BBN975. In vivo PET/CT and ex vivo biodistribution demonstrated preferential accumulation and visualization of tracer in the spleen, thymus, lymph nodes, and bone marrow. In tumor bearing mice, 89Zr-DFO-anti-CD3 demonstrated an 11.5-fold increase in tumor-to-blood signal compared to isotype control. Immunological profiling demonstrated no significant change to total T cell count, but observed CD4+ T cell depletion and CD8+ T cell expansion to the central and effector memory. This was very encouraging since a high CD8+ to CD4+ T cell ratio has already been associated with better patient prognosis. Ultimately, this anti-CD3 mAb allowed for in vivo imaging of homeostatic T cell distribution, and more specifically tumor-infiltrating T cells. Future applications of this radiolabeled mAb against CD3 could include prediction and monitoring of patient response to immunotherapy.


Subject(s)
Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/pathology , Positron Emission Tomography Computed Tomography/methods , Urinary Bladder Neoplasms/diagnostic imaging , Urinary Bladder Neoplasms/immunology , Animals , Antibodies , Butylhydroxybutylnitrosamine , CD3 Complex/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cell Line, Tumor , Deferoxamine , Flow Cytometry , Mice, Inbred C57BL , Neoplasm Transplantation , Radioisotopes , Radiopharmaceuticals , Tissue Distribution , Urinary Bladder Neoplasms/pathology , Zirconium
6.
Blood ; 130(3): 348-359, 2017 07 20.
Article in English | MEDLINE | ID: mdl-28550042

ABSTRACT

The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that affects the function and development of immune cells. Here, we show that recipient mice receiving AhR-/- T cells have improved survival and decreased acute graft-versus-host disease (aGVHD) in 2 different murine allogeneic bone marrow transplant (BMT) models. We also show that CD4+ T cells lacking AhR demonstrate reduced accumulation in secondary lymphoid tissue because of low levels of proliferation 4 days after BMT. Additionally, we found a significant increase in the quantity of peripherally induced regulatory donor T (pTreg) cells in the colon of recipients transplanted with AhR-/- T cells 14 days after transplant. Blockade of AhR using a clinically available AhR antagonist greatly enhanced the in vitro generation of inducible Treg (iTreg) cells from naïve CD4+ human T cells. We have identified AhR as a novel target on donor T cells that is critical to the pathogenesis of aGVHD.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/immunology , Bone Marrow Transplantation , Colon/immunology , Graft vs Host Disease/prevention & control , Receptors, Aryl Hydrocarbon/immunology , T-Lymphocytes, Regulatory/immunology , Acute Disease , Animals , Basic Helix-Loop-Helix Transcription Factors/deficiency , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Proliferation , Colon/pathology , Gene Expression Regulation , Graft vs Host Disease/immunology , Graft vs Host Disease/mortality , Graft vs Host Disease/pathology , Humans , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Purines/pharmacology , Receptors, Aryl Hydrocarbon/deficiency , Receptors, Aryl Hydrocarbon/genetics , Sirolimus/pharmacology , Survival Analysis , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/transplantation , Transplantation, Heterologous
7.
Bioinformatics ; 32(24): 3729-3734, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27559159

ABSTRACT

MOTIVATION: B-cell receptor (BCR) repertoire profiling is an important tool for understanding the biology of diverse immunologic processes. Current methods for analyzing adaptive immune receptor repertoires depend upon PCR amplification of VDJ rearrangements followed by long read amplicon sequencing spanning the VDJ junctions. While this approach has proven to be effective, it is frequently not feasible due to cost or limited sample material. Additionally, there are many existing datasets where short-read RNA sequencing data are available but PCR amplified BCR data are not. RESULTS: We present here V'DJer, an assembly-based method that reconstructs adaptive immune receptor repertoires from short-read RNA sequencing data. This method captures expressed BCR loci from a standard RNA-seq assay. We applied this method to 473 Melanoma samples from The Cancer Genome Atlas and demonstrate V'DJer's ability to accurately reconstruct BCR repertoires from short read mRNA-seq data. AVAILABILITY AND IMPLEMENTATION: V'DJer is implemented in C/C ++, freely available for academic use and can be downloaded from Github: https://github.com/mozack/vdjer CONTACT: benjamin_vincent@med.unc.edu or parkerjs@email.unc.eduSupplementary information: Supplementary data are available at Bioinformatics online.


Subject(s)
Computational Biology/methods , Receptors, Antigen, B-Cell/genetics , Sequence Analysis, RNA/methods , Software , B-Lymphocytes , Humans
8.
Cancer Immunol Res ; 3(3): 228-35, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25576336

ABSTRACT

Testing of T cell-based cancer therapeutics often involves measuring cancer antigen-specific T-cell populations with the assumption that they arise from in vivo clonal expansion. This analysis, using peptide/MHC tetramers, is often ambiguous. From a leukemia cell line, we identified a CDK4-derived peptide epitope, UNC-CDK4-1 (ALTPVVVTL), that bound HLA-A*02:01 with high affinity and could induce CD8⁺ T-cell responses in vitro. We identified UNC-CDK4-1/HLA-A*02:01 tetramer⁺ populations in 3 of 6 patients with acute myeloid leukemia who had undergone allogeneic stem cell transplantation. Using tetramer-based, single-cell sorting and T-cell receptor ß (TCRß) sequencing, we identified recurrent UNC-CDK4-1 tetramer-associated TCRß clonotypes in a patient with a UNC-CDK4-1 tetramer⁺ population, suggesting in vivo T-cell expansion to UNC-CDK4-1. In parallel, we measured the patient's TCRß repertoire and found it to be highly restricted/oligoclonal. The UNC-CDK4-1 tetramer-associated TCRß clonotypes represented >17% of the entire TCRß repertoire-far in excess of the UNC-CDK4-1 tetramer⁺ frequency-indicating that the recurrent TCRß clonotypes identified from UNC-CDK-4-1 tetramer⁺ cells were likely a consequence of the extremely constrained T-cell repertoire in the patient and not in vivo UNC-CDK4-1-driven clonal T-cell expansion. Mapping recurrent TCRß clonotype sequences onto TCRß repertoires can help confirm or refute antigen-specific T-cell expansion in vivo.


Subject(s)
Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , T-Lymphocyte Subsets/immunology , Female , HLA-A2 Antigen/immunology , Humans , Leukemia/immunology , Male , Middle Aged , Peptides/immunology , U937 Cells
9.
J Immunol ; 181(4): 2644-50, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18684955

ABSTRACT

CD8(+) T cell responses to persistent infections caused by intracellular pathogens are dominated by resting T effectors and T effector memory cells, with little evidence suggesting that a T central memory (T(CM)) population is generated. Using a model of Trypanosoma cruzi infection, we demonstrate that in contrast to the T effector/T effector memory phenotype of the majority of T. cruzi-specific CD8(+) T cells, a population of cells displaying hallmark characteristics of T(CM) cells is also present during long-term persistent infection. This population expressed the T(CM) marker CD127 and a subset expressed one or more of three other T(CM) markers: CD62L, CCR7, and CD122. Additionally, the majority of CD127(high) cells were KLRG1(low), indicating that they have not been repetitively activated through TCR stimulation. These CD127(high) cells were better maintained than their CD127(low) counterparts following transfer into naive mice, consistent with their observed surface expression of CD127 and CD122, which confer the ability to self-renew in response to IL-7 and IL-15. CD127(high) cells were capable of IFN-gamma production upon peptide restimulation and expanded in response to challenge infection, indicating that these cells are functionally responsive upon Ag re-encounter. These results are in contrast to what is typically observed during many persistent infections and indicate that a stable population of parasite-specific CD8(+) T cells capable of Ag-independent survival is maintained in mice despite the presence of persistent Ag.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/parasitology , Chagas Disease/immunology , Immunologic Memory , Trypanosoma cruzi/immunology , Animals , CD8-Positive T-Lymphocytes/pathology , Cell Proliferation , Cell Survival/immunology , Chagas Disease/parasitology , Chronic Disease , Epitopes, T-Lymphocyte/immunology , Interleukin-7 Receptor alpha Subunit/biosynthesis , Mice , Mice, Congenic , Mice, Inbred C57BL , Mice, Transgenic , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/parasitology , T-Lymphocyte Subsets/pathology , Time Factors
10.
Nat Med ; 14(5): 542-50, 2008 May.
Article in English | MEDLINE | ID: mdl-18425131

ABSTRACT

In this study, we document the development of stable, antigen-independent CD8+ T cell memory after drug-induced cure of a chronic infection. By establishing a system for drug cure of chronic Trypanosoma cruzi infection, we present the first extensively documented case of total parasite clearance after drug treatment of this infection. Cure resulted in the emergence of a stable, parasite-specific CD8+ T cell population with the characteristics of central memory cells, based upon expression of CD62L, CCR7, CD127, CD122, Bcl-2 and a reduced immediate in vivo CTL function. CD8+ T cells from treated and cured mice also expanded more rapidly and provided greater protection following challenge than those from chronically infected mice. These results show that complete pathogen clearance results in stable, antigen-independent and protective T cell memory, despite the potentially exhausting effects of prior long-term exposure to antigen in this chronic infection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Chagas Disease/drug therapy , Chagas Disease/immunology , Immunologic Memory/immunology , Nitroimidazoles/therapeutic use , Trypanosoma cruzi/immunology , Animals , Bromodeoxyuridine , Immunophenotyping , Mice , Nitroimidazoles/pharmacology , Receptors, Chemokine/immunology , Treatment Outcome , Trypanosoma cruzi/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL