Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Leukemia ; 34(6): 1626-1636, 2020 06.
Article in English | MEDLINE | ID: mdl-31908357

ABSTRACT

Immunotherapy targeting leukemia-associated antigens has shown promising results. Because of the heterogeneity of leukemia, vaccines with a single peptide have elicited only a limited immune response. Targeting several peptides together elicited peptide-specific cytotoxic T lymphocytes (CTLs) in leukemia patients, and this was associated with clinical responses. Thus, the discovery of novel antigens is essential. In the current study, we investigated cyclin E as a novel target for immunotherapy. Cyclin E1 and cyclin E2 were found to be highly expressed in hematologic malignancies, according to reverse transcription polymerase chain reaction and western blot analysis. We identified two HLA-A*0201 binding nonameric peptides, CCNE1M from cyclin E1 and CCNE2L from cyclin E2, which both elicited the peptide-specific CTLs. The peptide-specific CTLs specifically kill leukemia cells. Furthermore, CCNE1M and CCNE2L CTLs were increased in leukemia patients who underwent allogeneic hematopoietic stem cell transplantation, and this was associated with desired clinical outcomes. Our findings suggest that cyclin E1 and cyclin E2 are potential targets for immunotherapy in leukemia.


Subject(s)
Antigens, Neoplasm/metabolism , Cancer Vaccines/immunology , Cyclin E/immunology , Cyclins/immunology , HLA-A2 Antigen/immunology , Leukemia/immunology , Oncogene Proteins/immunology , Adult , Aged , Antigens, Neoplasm/immunology , Cancer Vaccines/pharmacology , Epitopes, T-Lymphocyte/immunology , Female , Humans , Male , Middle Aged , T-Lymphocytes, Cytotoxic/immunology , Young Adult
2.
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
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.
Front Immunol ; 9: 3153, 2018.
Article in English | MEDLINE | ID: mdl-30713535

ABSTRACT

Despite substantial advances in the treatment of acute myeloid leukemia (AML), only 30% of patients survive more than 5 years. Therefore, new therapeutics are much needed. Here, we present a novel therapeutic strategy targeting PR1, an HLA-A2 restricted myeloid leukemia antigen. Previously, we have developed and characterized a novel T-cell receptor-like monoclonal antibody (8F4) that targets PR1/HLA-A2 and eliminates AML xenografts by antibody-dependent cellular cytotoxicity (ADCC). To improve the potency of 8F4, we adopted a strategy to link T-cell cytotoxicity with a bi-specific T-cell-engaging antibody that binds PR1/HLA-A2 on leukemia and CD3 on neighboring T-cells. The 8F4 bi-specific antibody maintained high affinity and specific binding to PR1/HLA-A2 comparable to parent 8F4 antibody, shown by flow cytometry and Bio-Layer Interferometry. In addition, 8F4 bi-specific antibody activated donor T-cells in the presence of HLA-A2+ primary AML blasts and cell lines in a dose dependent manner. Importantly, activated T-cells lysed HLA-A2+ primary AML blasts and cell lines after addition of 8F4 bi-specific antibody. In conclusion, our studies demonstrate the therapeutic potential of a novel bi-specific antibody targeting the PR1/HLA-A2 leukemia-associated antigen, justifying further clinical development of this strategy.


Subject(s)
Antibodies, Bispecific/immunology , Antigens, Neoplasm/immunology , HLA-A2 Antigen/immunology , Leukemia, Myeloid, Acute/immunology , T-Lymphocytes/immunology , Animals , Antibodies, Bispecific/pharmacology , Antibody Specificity/immunology , Antigens, Neoplasm/metabolism , CHO Cells , Cell Line , Cricetulus , Cytotoxicity, Immunologic , HLA-A2 Antigen/metabolism , Humans , Immunotherapy, Adoptive , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Leukemia, Myeloid, Acute/therapy , Lymphocyte Activation , Protein Binding , T-Lymphocytes/metabolism
5.
Cancer Res ; 77(19): 5374-5383, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28819024

ABSTRACT

Early-phase clinical trials evaluating CD8+ T cell-eliciting, HER2-derived peptide vaccines administered to HER2+ breast cancer patients in the adjuvant setting suggest synergy between the vaccines and trastuzumab, the mAb targeting the HER2 protein. Among 60 patients enrolled in clinical trials evaluating the E75 + GM-CSF and GP2 + GM-CSF vaccines, there have been no recurrences in patients vaccinated after receiving trastuzumab as part of standard therapy in the per treatment analyses conducted after a median follow-up of greater than 34 months. Here, we describe a mechanism by which this synergy may occur. Flow cytometry showed that trastuzumab facilitated uptake of HER2 by dendritic cells (DC), which was mediated by the Fc receptor and was specific to trastuzumab. In vitro, increased HER2 uptake by DC increased cross-presentation of E75, the immunodominant epitope derived from the HER2 protein, an observation confirmed in two in vivo mouse models. This increased E75 cross-presentation, mediated by trastuzumab treatment, enabled more efficient expansion of E75-specific cytotoxic T cells (E75-CTL). These results demonstrate a mechanism by which trastuzumab links innate and adaptive immunity by facilitating activation of antigen-specific T cells. On the basis of these data, we conclude that HER2-positive breast cancer patients that have been treated with trastuzumab may experience a more robust antitumor immune response by restimulation of T cells with the E75 peptide vaccine, thereby accounting for the improved disease-free survival observed with combination therapy. Cancer Res; 77(19); 5374-83. ©2017 AACR.


Subject(s)
Breast Neoplasms/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Ovarian Neoplasms/immunology , Receptor, ErbB-2/metabolism , T-Lymphocytes, Cytotoxic/immunology , Trastuzumab/therapeutic use , Animals , Apoptosis , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cancer Vaccines/therapeutic use , Cell Proliferation , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Transgenic , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Receptor, ErbB-2/immunology , Tumor Cells, Cultured , Vaccines, Subunit/therapeutic use
6.
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
7.
Front Immunol ; 8: 1975, 2017.
Article in English | MEDLINE | ID: mdl-29422892

ABSTRACT

Cathepsin G (CG) is a myeloid azurophil granule protease that is highly expressed by acute myeloid leukemia (AML) blasts and leukemia stem cells. We previously identified CG1 (FLLPTGAEA), a human leukocyte antigen-A2-restricted nonameric peptide derived from CG, as an immunogenic target in AML. In this report, we aimed to assess the level of CG expression in acute lymphoid leukemia (ALL) and its potential as an immunotherapeutic target in ALL. Using RT-PCR and western blots, we identified CG mRNA and protein, respectively, in B-ALL patient samples and cell lines. We also examined CG expression in a large cohort of 130 patients with ALL via reverse-phase protein array (RPPA). Our data show that CG is widely expressed by ALL and is a poor prognosticator. In addition to endogenous expression, we also provide evidence that CG can be taken up by ALL cells. Finally, we demonstrate that patient ALL can be lysed by CG1-specific cytotoxic T lymphocytes in vitro. Together, these data show high expression of CG by ALL and implicate CG as a target for immunotherapy in ALL.

9.
Cancer Immunol Immunother ; 65(6): 741-51, 2016 06.
Article in English | MEDLINE | ID: mdl-27129972

ABSTRACT

Neutrophil elastase (NE) is an innate immune cell-derived inflammatory mediator that we have shown increases the presentation of tumor-associated peptide antigens in breast cancer. In this study, we extend these observations to show that NE uptake has a broad effect on enhancing antigen presentation by breast cancer cells. We show that NE increases human leukocyte antigen (HLA) class I expression on the surface of breast cancer cells in a concentration and time-dependent manner. HLA class I upregulation requires internalization of enzymatically active NE. Western blots of NE-treated breast cancer cells confirm that the expression of total HLA class I as well as the antigen-processing machinery proteins TAP1, LMP2, and calnexin does not change following NE treatment. This suggests that NE does not increase the efficiency of antigen processing; rather, it mediates the upregulation of HLA class I by stabilizing and reducing membrane recycling of HLA class I molecules. Furthermore, the effects of NE extend beyond breast cancer since the uptake of NE by EBV-LCL increases the presentation of HLA class I-restricted viral peptides, as shown by their increased sensitivity to lysis by EBV-specific CD8+ T cells. Together, our results show that NE uptake increases the responsiveness of breast cancer cells to adaptive immunity by broad upregulation of membrane HLA class I and support the conclusion that the innate inflammatory mediator NE enhances tumor cell recognition and increases tumor sensitivity to the host adaptive immune response.


Subject(s)
Antigen Presentation/immunology , Gene Expression Regulation , Histocompatibility Antigens Class I/genetics , Leukocyte Elastase/metabolism , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/metabolism , Cell Line, Tumor , Cytotoxicity, Immunologic , Histocompatibility Antigens Class I/immunology , Humans , Peptides/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism
10.
PLoS One ; 8(4): e60031, 2013.
Article in English | MEDLINE | ID: mdl-23560068

ABSTRACT

Adoptive T-cell therapy (ACT) using tumor-infiltrating lymphocytes (TIL) can induce tumor regression in up to 50% or more of patients with unresectable metastatic melanoma. However, current methods to expand melanoma TIL, especially the "rapid expansion protocol" (REP) were not designed to enhance the generation of optimal effector-memory CD8(+) T cells for infusion. One approach to this problem is to manipulate specific co-stimulatory signaling pathways to enhance CD8(+) effector-memory T-cell expansion. In this study, we determined the effects of activating the TNF-R family member 4-1BB/CD137, specifically induced in activated CD8(+) T cells, on the yield, phenotype, and functional activity of expanded CD8(+) T cells during the REP. We found that CD8(+) TIL up-regulate 4-1BB expression early during the REP after initial TCR stimulation, but neither the PBMC feeder cells in the REP or the activated TIL expressed 4-1BB ligand. However, addition of an exogenous agonistic anti-4-1BB IgG4 (BMS 663513) to the REP significantly enhanced the frequency and total yield of CD8(+) T cells as well as their maintenance of CD28 and increased their anti-tumor CTL activity. Gene expression analysis found an increase in bcl-2 and survivin expression induced by 4-1BB that was associated with an enhanced survival capability of CD8(+) post-REP TIL when re-cultured in the absence or presence of cytokines. Our findings suggest that adding an agonistic anti-4-1BB antibody during the time of TIL REP initiation produces a CD8(+) T cell population capable of improved effector function and survival. This may greatly improve TIL persistence and anti-tumor activity in vivo after adoptive transfer into patients.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunoglobulin G/pharmacology , Immunotherapy, Adoptive , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma/therapy , Skin Neoplasms/therapy , Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics , CD28 Antigens/genetics , CD28 Antigens/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/transplantation , Cell Proliferation/drug effects , Cell Survival/drug effects , Gene Expression/drug effects , Humans , Immunologic Memory/drug effects , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/immunology , Lymphocyte Activation/drug effects , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/transplantation , Melanoma/genetics , Melanoma/immunology , Melanoma/pathology , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/immunology , Signal Transduction/drug effects , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Survivin , Tumor Cells, Cultured , Tumor Necrosis Factor Receptor Superfamily, Member 9/agonists , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology
11.
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
12.
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
13.
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
14.
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
15.
Clin Cancer Res ; 18(9): 2465-77, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22307139

ABSTRACT

PURPOSE: Tumor-specific T cells are frequently induced naturally in melanoma patients and infiltrate tumors. It is enigmatic why these patients fail to experience tumor regression. Given that CD8(+) T cells mediate antigen-specific killing of tumor cells, the focus of this study was to identify alterations in the differentiation of CD8(+) residing at the tumor site, with emphasis on a population expressing CD57, a marker for terminal differentiation. EXPERIMENTAL DESIGN: We conducted flow cytometric analysis of CD8(+) tumor-infiltrating lymphocytes (TIL) isolated from 44 resected melanoma metastases with known T-cell differentiation markers. For comparison, peripheral blood mononuclear cells were isolated from matched melanoma patients. We sorted different CD8(+) subsets found in TIL and determined their effector functions. In addition, we carried out Vß clonotype expression analysis of T-cell receptors to determine lineage relationship between the CD8(+) TIL subsets. RESULTS: The majority of CD8(+) TIL was in the early-effector memory stage of differentiation. A significant population consisted of an oligoclonal subset of cells coexpressing CD27, CD28, CD57, and Granzyme B, with little or no perforin. These cells could be induced to proliferate, produce a high level of IFN-γ, and differentiate into CD27(-)CD57(+), perforin(high) mature CTL in vitro. Addition of TGF-ß1 prevented further differentiation. CONCLUSIONS: Our studies identified a novel subset of incompletely differentiated CD8(+) CTL coexpressing early effector memory and late CTL markers. This population resembles that found in patients with uncontrolled chronic viral infections. TGF-ß1, frequently produced by melanoma tumors, may be a key cytokine inhibiting further maturation of this subset.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Differentiation , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma/immunology , Melanoma/secondary , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Cytotoxic/immunology , Adolescent , Adult , Aged , Aged, 80 and over , CD57 Antigens/metabolism , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Female , Flow Cytometry , Humans , Immunologic Memory , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Lymphocyte Activation , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Male , Melanoma/metabolism , Middle Aged , Neoplasm Staging , Phenotype , Receptors, Antigen, T-Cell/metabolism , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology , Transforming Growth Factor beta1/pharmacology , Tumor Cells, Cultured , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism , Young Adult
16.
J Immunol Methods ; 331(1-2): 13-26, 2008 Feb 29.
Article in English | MEDLINE | ID: mdl-17959194

ABSTRACT

Adoptive therapy with antigen-specific T cells is a promising approach for the treatment of infectious diseases and cancer. However, cloning of antigen-specific T cells by the traditional approach of limiting dilution is a time-consuming, laborious, and inefficient process. Here, we describe a novel flow cytometric strategy for rapid isolation of human tumor antigen-specific T-cell clones by using T-cell receptor (TCR) Vbeta antibodies in combination with carboxyfluorescein succinimidyl ester (CFSE)-based proliferation assay. The CFSE dilution following antigen stimulation identified proliferating antigen-specific T cells, and the TCRVbeta antibodies allowed distinguishing T cells at the clonal level from a heterogeneous T-cell population. This method of TCRVbeta/CFSE dilution was used for the isolation of four different human lymphoma and melanoma-specific CD4(+) and CD8(+) T-cell clones reactive against defined and undefined tumor antigens. Isolated tumor-specific T-cell clones could be expanded to large numbers ex vivo while maintaining phenotype, function, and tumor antigen specificity. The method was simple, efficient, and reproducible, and may have potential application for the development of adoptive immunotherapeutic strategies.


Subject(s)
Antigens, Neoplasm/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Separation/methods , Flow Cytometry/methods , Receptors, Antigen, T-Cell, alpha-beta/immunology , Cell Line , Clone Cells/immunology , Humans , Immunotherapy, Adoptive , Lymphoma/immunology , MART-1 Antigen , Melanoma/immunology , Neoplasm Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...