Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Clin Cancer Res ; 25(8): 2610-2620, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30647079

ABSTRACT

PURPOSE: Inefficient homing of adoptively transferred cytotoxic T lymphocytes (CTLs) to tumors is a major limitation to the efficacy of adoptive cellular therapy (ACT) for cancer. However, through fucosylation, a process whereby fucosyltransferases (FT) add fucose groups to cell surface glycoproteins, this challenge may be overcome. Endogenously fucosylated CTLs and ex vivo fucosylated cord blood stem cells and regulatory T cells were shown to preferentially home to inflamed tissues and marrow. Here, we show a novel approach to enhance CTL homing to leukemic marrow and tumor tissue. EXPERIMENTAL DESIGN: Using the enzyme FT-VII, we fucosylated CTLs that target the HLA-A2-restricted leukemia antigens CG1 and PR1, the HER2-derived breast cancer antigen E75, and the melanoma antigen gp-100. We performed in vitro homing assays to study the effects of fucosylation on CTL homing and target killing. We used in vivo mouse models to demonstrate the effects of ex vivo fucosylation on CTL antitumor activities against leukemia, breast cancer, and melanoma. RESULTS: Our data show that fucosylation increases in vitro homing and cytotoxicity of antigen-specific CTLs. Furthermore, fucosylation enhances in vivo CTL homing to leukemic bone marrow, breast cancer, and melanoma tissue in NOD/SCID gamma (NSG) and immunocompetent mice, ultimately boosting the antitumor activity of the antigen-specific CTLs. Importantly, our work demonstrates that fucosylation does not interfere with CTL specificity. CONCLUSIONS: Together, our data establish ex vivo CTL fucosylation as a novel approach to improving the efficacy of ACT, which may be of great value for the future of ACT for cancer.


Subject(s)
Cytotoxicity, Immunologic , Epitopes, T-Lymphocyte/immunology , Immunotherapy, Adoptive , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Animals , Biomarkers , Cell Line, Tumor , Chemotaxis, Leukocyte/immunology , Gene Expression Regulation , Glycosylation , Humans , Immunophenotyping , Immunotherapy, Adoptive/methods , Lymphocyte Activation , Mice , Peptides/immunology , Transendothelial and Transepithelial Migration
2.
J Immunol ; 201(5): 1389-1399, 2018 09 01.
Article in English | MEDLINE | ID: mdl-30021768

ABSTRACT

Proteinase 3 (P3), a serine protease expressed by myeloid cells, localized within azurophil granules, and also expressed on the cellular membrane of polymorphonuclear neutrophils (PMN), is the target of autoimmunity in granulomatosis with polyangiitis. PR1, an HLA-A2 restricted nonameric peptide derived from P3, has been targeted effectively in myeloid leukemia. We previously showed (Molldrem et al. 2003. JClinInvest 111: 639-647) that overexpression of P3 in chronic myeloid leukemia induces apoptosis of high-affinity PR1-specific T cells, leading to deletional tolerance and leukemia outgrowth. In this study, we investigated the effect of membrane P3 (mP3)-expressing PMN and acute myeloid leukemia (AML) blasts on the proliferation of CD4 and CD8 T cells in vitro. We demonstrate that mP3-expressing PMN significantly inhibits autologous healthy donor T cell proliferation but does not affect cytokine production in activated T cells and that this effect requires cell proximity and was abrogated by P3 blockade. This inhibition required P3 enzyme activity. However, suppression was not reversed by either the addition of catalase or the inhibition of arginase I. In addition to P3 blockade, anti-low density lipoprotein receptor-related protein 1 (LRP1) Ab also restored T cells' capacity to proliferate. Last, we show dose-dependent inhibition of T cell proliferation by mP3-expressing AML blasts. Together, our findings demonstrate a novel mechanism whereby PMN- and AML-associated mP3 inhibits T cell proliferation via direct LRP1 and mP3 interaction, and we identify P3 as a novel target to modulate immunity in myeloid leukemia and autoimmune disease.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation , Leukemia, Myeloid, Acute/immunology , Myeloblastin/immunology , Neoplasm Proteins/immunology , Neutrophils/immunology , Adult , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , Female , Humans , Leukemia, Myeloid, Acute/pathology , Male , Neutrophils/pathology
3.
Clin Cancer Res ; 24(14): 3386-3396, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29661776

ABSTRACT

Purpose: PR1 is a human leukocyte antigen (HLA)-A2 nonameric peptide derived from neutrophil elastase (NE) and proteinase 3 (P3). We have previously shown that PR1 is cross-presented by solid tumors, leukemia, and antigen-presenting cells, including B cells. We have also shown that cross-presentation of PR1 by solid tumors renders them susceptible to killing by PR1-targeting immunotherapies. As multiple myeloma is derived from B cells, we investigated whether multiple myeloma is also capable of PR1 cross-presentation and subsequently capable of being targeted by using PR1 immunotherapies.Experimental Design: We tested whether multiple myeloma is capable of cross-presenting PR1 and subsequently becomes susceptible to PR1-targeting immunotherapies, using multiple myeloma cell lines, a xenograft mouse model, and primary multiple myeloma patient samples.Results: Here we show that multiple myeloma cells lack endogenous NE and P3, are able to take up exogenous NE and P3, and cross-present PR1 on HLA-A2. Cross-presentation by multiple myeloma utilizes the conventional antigen processing machinery, including the proteasome and Golgi, and is not affected by immunomodulating drugs (IMiD). Following PR1 cross-presentation, we are able to target multiple myeloma with PR1-CTL and anti-PR1/HLA-A2 antibody both in vitro and in vivoConclusions: Collectively, our data demonstrate that PR1 is a novel tumor-associated antigen target in multiple myeloma and that multiple myeloma is susceptible to immunotherapies that target cross-presented antigens. Clin Cancer Res; 24(14); 3386-96. ©2018 AACR.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , HLA-A2 Antigen/immunology , Multiple Myeloma/immunology , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/immunology , Animals , Antigen Presentation/drug effects , Antigen Presentation/immunology , Antigen-Presenting Cells/immunology , Biological Transport , Cell Line, Tumor , Complement Activation , Cross-Priming/drug effects , Cross-Priming/immunology , Cytotoxicity, Immunologic , Disease Models, Animal , HLA-A2 Antigen/chemistry , HLA-A2 Antigen/metabolism , Humans , Immunologic Factors/pharmacology , Immunomodulation/drug effects , Mice , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Proteasome Endopeptidase Complex/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Xenograft Model Antitumor Assays
4.
J Biol Chem ; 292(24): 10295-10305, 2017 06 16.
Article in English | MEDLINE | ID: mdl-28468826

ABSTRACT

Neutrophil elastase (NE) can be rapidly taken up by tumor cells that lack endogenous NE expression, including breast cancer, which results in cross-presentation of PR1, an NE-derived HLA-A2-restricted peptide that is an immunotherapy target in hematological and solid tumor malignancies. The mechanism of NE uptake, however, remains unknown. Using the mass spectrometry-based approach, we identify neuropilin-1 (NRP1) as a NE receptor that mediates uptake and PR1 cross-presentation in breast cancer cells. We demonstrated that soluble NE is a specific, high-affinity ligand for NRP1 with a calculated Kd of 38.7 nm Furthermore, we showed that NRP1 binds to the RRXR motif in NE. Notably, NRP1 knockdown with interfering RNA or CRISPR-cas9 system and blocking using anti-NRP1 antibody decreased NE uptake and, subsequently, susceptibility to lysis by PR1-specific cytotoxic T cells. Expression of NRP1 in NRP1-deficient cells was sufficient to induce NE uptake. Altogether, because NRP1 is broadly expressed in tumors, our findings suggest a role for this receptor in immunotherapy strategies that target cross-presented antigens.


Subject(s)
Absorption, Physiological , Breast Neoplasms/metabolism , Cross-Priming , Leukocyte Elastase/metabolism , Neoplasm Proteins/metabolism , Neuropilin-1/metabolism , Amino Acid Motifs , Antibodies, Blocking/metabolism , Breast Neoplasms/immunology , Breast Neoplasms/pathology , CRISPR-Cas Systems , Cell Line, Tumor , Female , Humans , Kinetics , Leukocyte Elastase/chemistry , Leukocyte Elastase/immunology , Ligands , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Neuropilin-1/antagonists & inhibitors , Neuropilin-1/chemistry , Neuropilin-1/genetics , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Interaction Domains and Motifs , RNA Interference , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Solubility , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism
5.
Cancer Immunol Res ; 5(4): 319-329, 2017 04.
Article in English | MEDLINE | ID: mdl-28254787

ABSTRACT

Immunotherapies targeting immune checkpoints have proven efficacious in reducing the burden of lung cancer in patients; however, the antigenic targets of these reinvigorated T cells remain poorly defined. Lung cancer tumors contain tumor-associated macrophages (TAM) and neutrophils, which release the serine proteases neutrophil elastase (NE) and proteinase 3 (P3) into the tumor microenvironment. NE and P3 shape the antitumor adaptive immune response in breast cancer and melanoma. In this report, we demonstrate that lung cancer cells cross-presented the tumor-associated antigen PR1, derived from NE and P3. Additionally, NE and P3 enhanced the expression of human leukocyte antigen (HLA) class I molecules on lung cancer cells and induced unique, endogenous peptides in the immunopeptidome, as detected with mass spectrometry sequencing. Lung cancer patient tissues with high intratumoral TAMs were enriched for MHC class I genes and T-cell markers, and patients with high TAM and cytotoxic T lymphocyte (CTL) infiltration had improved overall survival. We confirmed the immunogenicity of unique, endogenous peptides with cytotoxicity assays against lung cancer cell lines, using CTLs from healthy donors that had been expanded against select peptides. Finally, CTLs specific for serine proteases-induced endogenous peptides were detected in lung cancer patients using peptide/HLA-A2 tetramers and were elevated in tumor-infiltrating lymphocytes. Thus, serine proteases in the tumor microenvironment of lung cancers promote the presentation of HLA class I immunogenic peptides that are expressed by lung cancer cells, thereby increasing the antigen repertoire that can be targeted in lung cancer. Cancer Immunol Res; 5(4); 319-29. ©2017 AACR.


Subject(s)
Antigen Presentation/immunology , Antigens, Neoplasm/immunology , Immunomodulation , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Serine Proteases/metabolism , Amino Acid Sequence , Biomarkers , Cell Line, Tumor , Cytokines/metabolism , HLA-A2 Antigen/genetics , HLA-A2 Antigen/immunology , HLA-A2 Antigen/metabolism , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Humans , Immunophenotyping , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Lung Neoplasms/pathology , Lymphocyte Activation , Peptides/chemistry , Peptides/immunology , Peptides/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
6.
Cytotherapy ; 18(8): 995-1001, 2016 08.
Article in English | MEDLINE | ID: mdl-27378343

ABSTRACT

BACKGROUND AIMS: PR1 is an HLA-A2 restricted leukemia-associated antigen derived from neutrophil elastase and proteinase 3, both of which are normally stored in the azurophil granules of myeloid cells but overexpressed in myeloid leukemic cells. PR1-specific cytotoxic lymphocytes (PR1-CTLs) have activity against primary myeloid leukemia in vitro and in vivo and thus could have great potential in the setting of adoptive cellular therapy (ACT). Adult peripheral blood-derived PR1-CTLs are infrequent but preferentially lyse myeloid leukemia cells. We sought to examine PR1-CTLs in umbilical cord blood (UCB) because UCB units provide a rapidly available cell source and a lower risk of graft-versus-host disease, even in the setting of mismatched human leukocyte antigen (HLA) loci. METHODS: We first determined the frequency of PR1-CTLs in HLA-A2(+) UCB units and then successfully expanded them ex vivo using repeated stimulation with PR1 peptide-pulsed antigen-presenting cells (APCs). After expansion, we assessed the PR1-CTL phenotype (naive, effector, memory) and function against PR1-expressing target cells. RESULTS: PR1-CTLs are detected at an average frequency of 0.14% within the CD8(+) population of fresh UCB units, which is 45 times higher than in healthy adult peripheral blood. UCB PR1-CTLs are phenotypically naive, consistent with the UCB CD8(+) population as a whole. In addition, the cells can be expanded by stimulation with PR1 peptide-pulsed APCs. Expansion results in an increased frequency of PR1-CTLs, up to 4.56%, with an average 20-fold increase in total number. After expansion, UCB PR1-CTLs express markers consistent with effector memory T cells. Expanded UCB PR1-CTLs are functional in vitro as they are able to produce cytokines and lyse PR1-expressing leukemia cell lines. CONCLUSIONS: This study is the first report to show that T cells specific for a leukemia-associated antigen are found at a significantly higher frequency in UCB than adult blood. Our results also demonstrate specific cytotoxicity of expanded UCB-derived PR1-CTLs against PR1-expressing targets. Together, our data suggest that UCB PR1-CTLs could be useful to prevent or treat leukemia relapse in myeloid leukemia patients.


Subject(s)
Antibody-Dependent Cell Cytotoxicity , Fetal Blood/cytology , HLA-A2 Antigen/immunology , Immunotherapy, Adoptive , Leukemia, Myeloid/therapy , Myeloblastin/immunology , T-Lymphocytes, Cytotoxic/immunology , Adult , Cells, Cultured , Fetal Blood/immunology , HLA-A2 Antigen/chemistry , HLA-A2 Antigen/metabolism , Humans , K562 Cells , Leukemia, Myeloid/immunology , Lymphocyte Count , Myeloblastin/chemistry , Myeloblastin/metabolism , T-Lymphocytes, Cytotoxic/metabolism , U937 Cells
7.
Oncotarget ; 5(24): 12675-93, 2014 Dec 30.
Article in English | MEDLINE | ID: mdl-25544748

ABSTRACT

Mesoderm Inducer in Xenopus Like1 (MIXL1), a paired-type homeobox transcription factor induced by TGF-ß family of ligands is required for early embryonic specification of mesoderm and endoderm. Retrovirally transduced Mixl1 is reported to induce acute myelogenous leukemia (AML) with a high penetrance. But the mechanistic underpinnings of MIXL1 mediated leukemogenesis are unknown. Here, we establish the protooncogene c-REL to be a transcriptional target of MIXL1 by genome wide chromatin immune precipitation. Accordingly, expression of c-REL and its downstream targets BCL2L1 and BCL2A2 are elevated in MIXL1 expressing cells. Notably, MIXL1 regulates c-REL through a zinc finger binding motif, potentially by a MIXL1-Zinc finger protein transcriptional complex. Furthermore, MIXL1 expression is detected in the cancer genome atlas (TCGA) AML samples in a pattern mutually exclusive from that of HOXA9, CDX2 and HLX suggesting the existence of a core, yet distinct HOX transcriptional program. Finally, we demonstrate MIXL1 to be induced by BMP4 and not TGF-ß in primary human hematopoietic stem and progenitor cells. Consequently, MIXL1 expressing AML cells are preferentially sensitive to the BMPR1 kinase inhibitor LDN-193189. These findings support the existence of a novel MIXL1-c REL mediated survival axis in AML that can be targeted by BMPR1 inhibitors. (MIXL1- human gene, Mixl1- mouse ortholog, MIXL1- protein).


Subject(s)
Bone Morphogenetic Protein 4/genetics , Homeodomain Proteins/genetics , Leukemia, Myeloid, Acute/genetics , Animals , Bone Morphogenetic Protein Receptors, Type I/antagonists & inhibitors , Cell Differentiation/physiology , Cell Line, Tumor , Genes, Homeobox , Genes, rel , HEK293 Cells , HL-60 Cells , Homeodomain Proteins/biosynthesis , Humans , K562 Cells , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Mice , Molecular Targeted Therapy , U937 Cells
8.
Proc Natl Acad Sci U S A ; 110(51): 20717-22, 2013 Dec 17.
Article in English | MEDLINE | ID: mdl-24297924

ABSTRACT

Angiogenesis is fundamental to tumorigenesis and an attractive target for therapeutic intervention against cancer. We have recently demonstrated that CD13 (aminopeptidase N) expressed by nonmalignant host cells of unspecified types regulate tumor blood vessel development. Here, we compare CD13 wild-type and null bone marrow-transplanted tumor-bearing mice to show that host CD13(+) bone marrow-derived cells promote cancer progression via their effect on angiogenesis. Furthermore, we have identified CD11b(+)CD13(+) myeloid cells as the immune subpopulation directly regulating tumor blood vessel development. Finally, we show that these cells are specifically localized within the tumor microenvironment and produce proangiogenic soluble factors. Thus, CD11b(+)CD13(+) myeloid cells constitute a population of bone marrow-derived cells that promote tumor progression and metastasis and are potential candidates for the development of targeted antiangiogenic drugs.


Subject(s)
Angiogenesis Inducing Agents/metabolism , Bone Marrow Cells/metabolism , CD13 Antigens , Myeloid Cells/metabolism , Neoplasms, Experimental/metabolism , Neovascularization, Pathologic/metabolism , Animals , Bone Marrow Cells/pathology , CD11b Antigen , Cell Line, Tumor , Mice , Mice, Inbred BALB C , Mice, Knockout , Myeloid Cells/pathology , Neoplasm Metastasis , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , Neovascularization, Pathologic/pathology
9.
Clin Cancer Res ; 19(1): 247-57, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23147993

ABSTRACT

PURPOSE: Immunotherapy targeting aberrantly expressed leukemia-associated antigens has shown promise in the management of acute myeloid leukemia (AML). However, because of the heterogeneity and clonal evolution that is a feature of myeloid leukemia, targeting single peptide epitopes has had limited success, highlighting the need for novel antigen discovery. In this study, we characterize the role of the myeloid azurophil granule protease cathepsin G (CG) as a novel target for AML immunotherapy. EXPERIMENTAL DESIGN: We used Immune Epitope Database and in vitro binding assays to identify immunogenic epitopes derived from CG. Flow cytometry, immunoblotting, and confocal microscopy were used to characterize the expression and processing of CG in AML patient samples, leukemia stem cells, and normal neutrophils. Cytotoxicity assays determined the susceptibility of AML to CG-specific cytotoxic T lymphocytes (CTL). Dextramer staining and cytokine flow cytometry were conducted to characterize the immune response to CG in patients. RESULTS: CG was highly expressed and ubiquitinated in AML blasts, and was localized outside granules in compartments that facilitate antigen presentation. We identified five HLA-A*0201 binding nonameric peptides (CG1-CG5) derived from CG, and showed immunogenicity of the highest HLA-A*0201 binding peptide, CG1. We showed killing of primary AML by CG1-CTL, but not normal bone marrow. Blocking HLA-A*0201 abrogated CG1-CTL-mediated cytotoxicity, further confirming HLA-A*0201-dependent killing. Finally, we showed functional CG1-CTLs in peripheral blood from AML patients following allogeneic stem cell transplantation. CONCLUSION: CG is aberrantly expressed and processed in AML and is a novel immunotherapeutic target that warrants further development.


Subject(s)
Cathepsin G/immunology , HLA-A2 Antigen/immunology , Leukemia, Myeloid, Acute/immunology , Peptides/immunology , ADP-ribosyl Cyclase 1/metabolism , Antigens, CD34/metabolism , Cathepsin G/chemistry , Cathepsin G/metabolism , Cell Line, Tumor , Cytotoxicity, Immunologic , Epitopes/immunology , Epitopes/metabolism , HLA-A2 Antigen/metabolism , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , Humans , Immunotherapy , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/therapy , Peptides/metabolism , Protein Binding/immunology , Protein Transport , T-Lymphocytes, Cytotoxic/immunology , Transplantation, Homologous
10.
J Immunol ; 189(11): 5476-84, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-23105141

ABSTRACT

PR1 is a HLA-A2-restricted peptide that has been targeted successfully in myeloid leukemia with immunotherapy. PR1 is derived from the neutrophil granule proteases proteinase 3 (P3) and neutrophil elastase (NE), which are both found in the tumor microenvironment. We recently showed that P3 and NE are taken up and cross-presented by normal and leukemia-derived APCs, and that NE is taken up by breast cancer cells. We now extend our findings to show that P3 and NE are taken up and cross-presented by human solid tumors. We further show that PR1 cross-presentation renders human breast cancer and melanoma cells susceptible to killing by PR1-specific CTLs (PR1-CTL) and the anti-PR1/HLA-A2 Ab 8F4. We also show PR1-CTL in peripheral blood from patients with breast cancer and melanoma. Together, our data identify cross-presentation as a novel mechanism through which cells that lack endogenous expression of an Ag become susceptible to therapies that target cross-presented Ags and suggest PR1 as a broadly expressed tumor Ag.


Subject(s)
Antigens, Neoplasm/immunology , Breast Neoplasms/therapy , Immunotherapy , Leukocyte Elastase/immunology , Melanoma/therapy , Myeloblastin/immunology , Skin Neoplasms/therapy , Antibodies/pharmacology , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/immunology , Antigens, Neoplasm/metabolism , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cross-Priming , Female , HLA-A2 Antigen/immunology , Humans , Leukocyte Elastase/chemistry , Melanoma/immunology , Melanoma/pathology , Molecular Targeted Therapy , Myeloblastin/chemistry , Peptide Fragments/chemistry , Peptide Fragments/immunology , Skin Neoplasms/immunology , Skin Neoplasms/pathology , T-Lymphocytes, Cytotoxic/immunology , Tumor Cells, Cultured
11.
Cancer Res ; 72(13): 3153-62, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22564522

ABSTRACT

There is little understanding of the impact of tumor-associated neutrophils (TAN) on adaptive immunity to tumors. In this study, we report the results of an investigation of the pathobiologic basis for the prognostic significance of neutrophil elastase, a serine protease found in neutrophil granules, in a model of cyclin E (CCNE)-overexpressing breast cancer. We established that neutrophil elastase was expressed by TAN within breast cancer tissues but not by breast cancer cells. Neutrophil elastase modulated killing of breast cancer cells by CTLs specific for CCNE-derived HLA-A2-restricted peptide (ILLDWLMEV). Breast cancer cells exhibited striking antigen-specific uptake of neutrophil elastase from the microenvironment that was independent of neutrophil elastase enzymatic activity. Furthermore, neutrophil elastase uptake increased expression of low molecular weight forms of CCNE and enhanced susceptibility to peptide-specific CTL lysis, suggesting that CCNE peptides are naturally presented on breast cancer cells. Taken together, our findings reveal a previously unknown mechanism of antitumor adaptive immunity that links cancer cell uptake of an inflammatory mediator to an effective cytolytic response against an important breast cancer antigen.


Subject(s)
Breast Neoplasms/pathology , Inflammation Mediators/metabolism , Leukocyte Elastase/metabolism , Adaptation, Physiological , Amino Acid Sequence , Base Sequence , Breast Neoplasms/enzymology , Breast Neoplasms/immunology , Breast Neoplasms/metabolism , DNA Primers , Female , Humans , Immunohistochemistry , Molecular Sequence Data , Reverse Transcriptase Polymerase Chain Reaction
12.
Hum Immunol ; 73(7): 693-8, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22548720

ABSTRACT

Vitamin D deficiency is adversely associated with diseases characterized by inflammation. The combination of the high incidence of vitamin D deficiency in patients undergoing allogeneic stem cell transplants (SCT) and the potential role of vitamin D deficiency in influencing graft-versus-host disease led us to further characterize the expression of VDR on alloreactive T cells. We hypothesized that vitamin D receptor expression may directly regulate alloreactive T cell responses. To overcome existing limitations in measuring VDR in bulk cellular populations, we developed a flow cytometric assay to measure cytoplasmic VDR in human T cells. Upon stimulation, VDR was expressed extremely early and exhibited sustained upregulation with chronic stimulation. VDR expression was also coupled to cytokine production, proliferation, and ERK1/2 phosphorylation. In addition, VDR exhibited a maturation stage-specific pattern of expression, with greatest expression on cells known to mediate GVHD, naïve and early memory T cells. Alloreactive T cells upregulated VDR, whereas the nonreactive T cells did not. Finally, repletion of vitamin D in vitro was sufficient to significantly reduce alloreactive T cell responses. These data suggest that vitamin D effects on T cells may be important in reducing graft versus host disease (GVHD) in the allogeneic stem cell transplant setting.


Subject(s)
Graft vs Host Disease/immunology , Receptors, Calcitriol/metabolism , Stem Cell Transplantation , T-Lymphocyte Subsets/metabolism , T-Lymphocytes/metabolism , Cell Differentiation , Cell Separation , Cells, Cultured , Flow Cytometry , Humans , Immunologic Memory , Immunomodulation , Isoantigens/immunology , Receptors, Calcitriol/genetics , T-Lymphocyte Subsets/immunology , T-Lymphocytes/immunology , Up-Regulation , Vitamin D/metabolism
13.
J Immunother ; 35(4): 309-20, 2012 May.
Article in English | MEDLINE | ID: mdl-22495388

ABSTRACT

Cross-presentation is an important mechanism by which exogenous tumor antigens are presented to elicit immunity. Because neutrophil elastase (NE) and proteinase-3 (P3) expression is increased in myeloid leukemia, we investigated whether NE and P3 are cross-presented by dendritic cells (DC) and B cells, and whether the NE and P3 source determines immune outcomes. We show that NE and P3 are elevated in leukemia patient serum and that levels correlate with remission status. We demonstrate cellular uptake of NE and P3 into lysosomes, ubiquitination, and proteasome processing for cross-presentation. Using anti-PR1/human leukocyte antigen-A2 monoclonal antibody, we provide direct evidence that B-cells cross-present soluble and leukemia-associated NE and P3, whereas DCs cross-present only leukemia-associated NE and P3. Cross-presentation occurred at early time points but was not associated with DC or B-cell activation, suggesting that NE and P3 cross-presentation may favor tolerance. Furthermore, we show aberrant subcellular localization of NE and P3 in leukemia blasts to compartments that share common elements of the classic major histocompatibility class I antigen-presenting pathway, which may facilitate cross-presentation. Our data demonstrate distinct mechanisms for cross-presentation of soluble and cell-associated NE and P3, which may be valuable in understanding immunity to PR1 in leukemia.


Subject(s)
Antigens, Neoplasm/immunology , Cross-Priming/immunology , Leukemia/immunology , Leukocyte Elastase/immunology , Myeloblastin/immunology , Antigen-Presenting Cells/immunology , Antigens, Neoplasm/metabolism , B-Lymphocytes/immunology , Cell Line, Tumor , Dendritic Cells/immunology , Histocompatibility Antigens Class I/immunology , Humans , Leukocyte Elastase/metabolism , Lysosomes/metabolism , Myeloblastin/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Transport , Signal Transduction , Ubiquitination
14.
J Immunol ; 183(10): 6167-74, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19841187

ABSTRACT

Late memory T cell skewing is observed in the setting of immune recovery after cord blood transplantation, and may be associated with inferior control of viral reactivation and cancers. Therefore, we sought to understand how late memory cells differ functionally from earlier stage memory T cells, and whether surface phenotypes associated with differentiation stages were predictably associated with functional signatures. Higher order cytokine flow cytometry allows characterization of human T cells based on complex phenotypic markers and their differential capacity to simultaneously secrete effector proteins, including cytokines and chemokines. We used 8-color, 10-parameter cytokine flow cytometry to characterize the functional activation of human late memory CD8(+) T cells defined by CD45RA and CD27 expression (CD27(-)CD45RA(+)). We assessed the 15 possible functional signatures of cells defined by production of IL-2, IFN-gamma, TNF-alpha, and MIP-1beta alone or in combination, following activation with Ags stimulating bypassing surface proteins (PMA:ionomycin) or through the TCR (e.g., viral Ags). Late memory CD8(+) T cells produced abundant amounts of CC chemokines (MIP-1beta, MIP-1alpha, and RANTES) but not IL-2. IL-2/IFN-gamma coproduction, characteristic of protective immune responses to viral infections, was absent in late memory CD8(+) T cells. These data demonstrate that functional cytokine signatures are predictably associated with CD8(+) maturation stages, and that the polarization of late memory CD8(+) T cells toward CC chemokine production and away from IL-2 production suggests a unique functional role for this subset.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Chemokine CCL3/immunology , Chemokine CCL4/immunology , Chemokine CCL5/immunology , Immunologic Memory , Phosphoproteins/immunology , Viral Matrix Proteins/immunology , Adult , CD8-Positive T-Lymphocytes/metabolism , Chemokine CCL3/metabolism , Chemokine CCL4/metabolism , Chemokine CCL5/metabolism , Humans , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-2/immunology , Interleukin-2/metabolism , Leukocyte Common Antigens/immunology , Leukocyte Common Antigens/metabolism , Middle Aged , Phosphoproteins/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism , Viral Matrix Proteins/metabolism
15.
Blood ; 110(13): 4543-51, 2007 Dec 15.
Article in English | MEDLINE | ID: mdl-17671230

ABSTRACT

Advances in immune assessment, including the development of T-cell receptor excision circle (TREC) assays of thymopoiesis, cytokine-flow cytometry assays of T-cell function, and higher-order phenotyping of T-cell maturation subsets have improved our understanding of T-cell homeostasis. Limited data exist using these methods to characterize immune recovery in adult cord blood (CB) transplant recipients, in whom infection is a leading cause of mortality. We now report the results of a single-center prospective study of T-cell immune recovery after cord blood transplantation (CBT) in a predominantly adult population. Our primary findings include the following: (1) Prolonged T lymphopenia and compensatory expansion of B and natural killer (NK) cells was evident; (2) CB transplant recipients had impaired functional recovery, although we did observe posttransplantation de novo T-cell responses to cytomegalovirus (CMV) in a subset of patients; (3) Thymopoietic failure characterized post-CBT immune reconstitution, in marked contrast to results in other transplant recipients; and (4) Thymopoietic failure was associated with late memory T-cell skewing. Our data suggest that efforts to improve outcomes in adult CB transplant recipients should be aimed at optimizing T-cell immune recovery. Strategies that improve the engraftment of lymphoid precursors, protect the thymus during pretransplant conditioning, and/or augment the recovery of thymopoiesis may improve outcomes after CBT.


Subject(s)
Cord Blood Stem Cell Transplantation , Hematopoiesis , Immune System/cytology , Regeneration , T-Lymphocytes/immunology , Humans , Immune System/physiology , Immunologic Memory , Lymphocyte Count , Lymphocytes , Prospective Studies , Thymus Gland/cytology
16.
Blood ; 105(6): 2258-65, 2005 Mar 15.
Article in English | MEDLINE | ID: mdl-15546954

ABSTRACT

Aspergillus fumigatus (AF) is a ubiquitous mold and is the most common cause of invasive aspergillosis, an important source of morbidity and mortality in immunocompromised hosts. Using cytokine flow cytometry, we assessed the magnitude of functional CD4+ and CD8+ T-cell responses following stimulation with Aspergillus antigens. Relative to those seen with cytomegalovirus (CMV) or superantigen stimulation, responses to Aspergillus antigens were near background levels. Subsequently, we confirmed that gliotoxin, the most abundant mycotoxin produced by AF, was able to suppress functional T-cell responses following CMV or staphylococcal enterotoxin B (SEB) stimulation. Additional studies demonstrated that crude AF filtrates and purified gliotoxin inhibited antigen-presenting cell function and induced the preferential death of monocytes, leading to a marked decrease in the monocyte-lymphocyte ratio. Analysis of caspase-3 activation confirmed that gliotoxin preferentially induced apoptosis of monocytes; similar effects were observed in CD83+ monocyte-derived dendritic cells. Importantly, the physiologic effects of gliotoxin in vitro were observed below concentrations recently observed in the serum of patients with invasive aspergillosis. These studies suggest that the production of gliotoxin by AF may constitute an important immunoevasive mechanism that is mediated by direct effects on antigen-presenting cells and both direct and indirect effects on T cells.


Subject(s)
Antigens, Fungal/immunology , Apoptosis/immunology , Aspergillosis/immunology , Aspergillus fumigatus/immunology , Gliotoxin/immunology , Monocytes/immunology , Antigens, CD/immunology , Aspergillosis/mortality , Aspergillosis/pathology , Aspergillus fumigatus/chemistry , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Caspase 3 , Caspases/immunology , Cells, Cultured , Cytomegalovirus/immunology , Dendritic Cells/immunology , Dendritic Cells/pathology , Dose-Response Relationship, Immunologic , Enterotoxins/immunology , Gliotoxin/chemistry , Humans , Immunoglobulins/immunology , Lymphocyte Activation/immunology , Membrane Glycoproteins/immunology , Monocytes/pathology , CD83 Antigen
17.
Blood ; 104(12): 3429-36, 2004 Dec 01.
Article in English | MEDLINE | ID: mdl-15284108

ABSTRACT

Human T-cell alloreactivity plays an important role in many disease processes, including the rejection of solid organ grafts and graft-versus-host disease (GVHD) following allogeneic stem cell transplantation. To develop a better understanding of the T cells involved in alloreactivity in humans, we developed a cytokine flow cytometry (CFC) assay that enabled us to characterize the phenotypic and functional characteristic of T cells responding to allogeneic stimuli. Using this approach, we determined that most T-cell alloreactivity resided within the CD4(+) T-cell subset, as assessed by activation marker expression and the production of effector cytokines (eg, tumor necrosis factor alpha [TNF]alpha) implicated in human GVHD. Following prolonged stimulation in vitro using either allogeneic stimulator cells or viral antigens, we found that coexpression of activation markers within the CD4(+) T-cell subset occurred exclusively within a subpopulation of T cells that significantly increased their surface expression of CD4. We then developed a simple sorting strategy that exploited these phenotypic characteristics to specifically deplete alloreactive T cells while retaining broad specificity for other stimuli, including viral antigens and third-party alloantigens. This approach also was applied to specifically enrich or deplete human virus-specific T cells.


Subject(s)
Flow Cytometry , Immunity, Cellular , Lymphocyte Depletion/methods , T-Lymphocytes/immunology , Antigens, Differentiation, T-Lymphocyte/analysis , Blood Component Removal , CD4 Antigens/analysis , CD4-Positive T-Lymphocytes/immunology , Cytokines/biosynthesis , Humans , Kinetics , Lymphocyte Activation/immunology , Up-Regulation
18.
Blood ; 103(3): 1140-6, 2004 Feb 01.
Article in English | MEDLINE | ID: mdl-12907445

ABSTRACT

Graft-versus-host disease (GVHD) occurs in an unpredictable fashion after 30% to 50% of matched-related transplantations. The presence of increased frequencies of CD4(+)CD25(+) regulatory T cells in donor grafts has been shown to ameliorate GVHD after allogeneic transplantation in murine models. To determine whether a similar relationship exists in humans, we quantitated the coexpression of CD25 on CD4(+) and CD8(+) T cells within 60 donor grafts infused into matched siblings and examined GVHD incidence in the respective recipients. Recipients in whom GVHD developed received donor grafts containing significantly higher frequencies of CD4(+) T cells coexpressing CD25 than those who did not (median, 9.26% vs 2.22%; P =.004). Frequencies of donor graft CD8(+) T cells coexpressing CD25 were also higher (0.65% vs 0.14%; P =.002). Furthermore, transplant recipients who received grafts containing fewer CD4(+)CD25(+) and CD8(+)CD25(+) T cells were less likely to acquire acute GVHD, even though these donor-recipient pairs were similar to others with respect to relevant clinical variables. These data suggest that the coexpression of CD4 and CD25 may be insufficient to identify regulatory T cells in humans and that increased frequencies and numbers of CD25(+) T cells in donor grafts is associated with GVHD in transplant recipients.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Graft vs Host Disease/etiology , Graft vs Host Disease/immunology , Peripheral Blood Stem Cell Transplantation/adverse effects , Receptors, Interleukin-2/metabolism , Adolescent , Adult , Aged , Animals , CD4 Lymphocyte Count , Child , Female , Histocompatibility Testing , Humans , Lymphocyte Count , Male , Mice , Middle Aged , Risk Factors , Transplantation, Homologous
19.
Br J Haematol ; 119(3): 773-6, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12437658

ABSTRACT

The small number of progenitor cells is the major limitation to the use of umbilical cord blood (UCB) for the transplantation of adults. We tested the hypothesis that two units transplanted simultaneously could each contribute to haematopoietic reconstitution. A patient with advanced acute lymphocytic leukaemia received a mismatched, unrelated UCB transplant using units from two donors after conditioning. The recipient achieved a complete remission without graft-versus-host disease. Double chimaerism was documented in several leucocyte subpopulations; both units contributed to haematopoiesis until relapse. Triple chimaerism was present from relapse until death due to leukaemia. This approach may potentially improve UCB transplantation outcome for adults lacking a histocompatible donor.


Subject(s)
Cord Blood Stem Cell Transplantation/methods , HLA Antigens/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Adult , Humans , Leukocytes/immunology , Transplantation Chimera , Transplantation Conditioning/methods , Transplantation, Homologous
20.
Blood ; 100(10): 3690-7, 2002 Nov 15.
Article in English | MEDLINE | ID: mdl-12393402

ABSTRACT

Cytomegalovirus (CMV) infection causes significant morbidity and mortality in the setting of immunodeficiency, including the immune reconstitution phase following allogeneic stem cell transplantation (SCT). We assessed CMV-specific CD4(+) and CD8(+) T-cell responses in 87 HLA-A*0201-positive (A2+) and/or B*0702-positive (B7+) allogeneic stem cell transplant recipients using HLA-peptide tetramer staining and cytokine flow cytometry (CFC) to examine the association of CMV-specific immune reconstitution and CMV antigenemia following SCT. Strong CMV-specific T-cell responses recovered in most subjects (77 of 87, 88%) after SCT. Frequencies of CMV-specific CD8(+) T cells were significantly higher in those subjects who experienced early antigenemia relative to those who did not (2.2% vs 0.33%, P =.0002), as were frequencies of CMV-specific CD4(+) T cells (1.71% vs 0.75%, P =.002). Frequencies of CMV-specific CD8(+) T cells were also higher in subjects experiencing late antigenemia (2.4% vs 0.57%). When we combined tetramer staining and an assessment of cytokine production in a single assay, we found that individuals who experienced CMV antigenemia had lower tumor necrosis factor-alpha (TNF-alpha)-producing fractions of tetramer-staining CMV-specific CD8(+) T cells than subjects who did not (25% vs 65%, P =.015). Furthermore, individuals at high risk for CMV reactivation, including patients with acute graft-versus-host disease and those receiving steroids, had low fractions of cytokine-producing CMV-specific CD8(+) T cells (25% and 27%, respectively). These data suggest that the inability to control CMV reactivation following allogeneic SCT is due to the impaired function of antigen-specific CD8(+) T cells rather than an inability to recover sufficient numbers of CMV-specific T cells.


Subject(s)
Antigens, Viral/immunology , CD8-Positive T-Lymphocytes/pathology , Cytomegalovirus/immunology , Hematopoietic Stem Cell Transplantation/adverse effects , Virus Activation/immunology , Adult , Aged , Antigens, Viral/blood , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cross-Sectional Studies , Cytomegalovirus/growth & development , Cytomegalovirus/physiology , Female , Hematologic Neoplasms/immunology , Hematologic Neoplasms/therapy , Humans , Male , Middle Aged , Transplantation, Homologous/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL
...