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










Database
Language
Publication year range
1.
Cancer Res ; 73(17): 5569-79, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23867470

ABSTRACT

Acute myeloid leukemia (AML) is a malignancy of stem cells with an unlimited capacity for self-renewal. MUC1 is a secreted, oncogenic mucin that is expressed aberrantly in AML blasts, but its potential uses to target AML stem cells have not been explored. Here, we report that MUC1 is highly expressed on AML CD34(+)/lineage(-)/CD38(-) cells as compared with their normal stem cell counterparts. MUC1 expression was not restricted to AML CD34(+) populations as similar results were obtained with leukemic cells from patients with CD34(-) disease. Engraftment of AML stem cell populations that highly express MUC1 (MUC1(high)) led to development of leukemia in NOD-SCID IL2Rgamma(null) (NSG) immunodeficient mice. In contrast, MUC1(low) cell populations established normal hematopoiesis in the NSG model. Functional blockade of the oncogenic MUC1-C subunit with the peptide inhibitor GO-203 depleted established AML in vivo, but did not affect engraftment of normal hematopoietic cells. Our results establish that MUC1 is highly expressed in AML stem cells and they define the MUC1-C subunit as a valid target for their therapeutic eradication.


Subject(s)
Leukemia, Myeloid, Acute/prevention & control , Mucin-1/metabolism , Neoplastic Stem Cells/drug effects , Peptides/pharmacology , ADP-ribosyl Cyclase 1/genetics , ADP-ribosyl Cyclase 1/metabolism , Animals , Antigens, CD34/genetics , Antigens, CD34/metabolism , Apoptosis/drug effects , Blotting, Western , Cell Differentiation , Cell Proliferation/drug effects , Female , Flow Cytometry , Humans , Immunoenzyme Techniques , Immunophenotyping , In Situ Hybridization, Fluorescence , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Male , Mice , Mice, Inbred NOD , Mice, SCID , Mucin-1/chemistry , Mucin-1/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
2.
Clin Cancer Res ; 19(13): 3640-8, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23685836

ABSTRACT

PURPOSE: A multiple myeloma vaccine has been developed whereby patient-derived tumor cells are fused with autologous dendritic cells, creating a hybridoma that stimulates a broad antitumor response. We report on the results of a phase II trial in which patients underwent vaccination following autologous stem cell transplantation (ASCT) to target minimal residual disease. EXPERIMENTAL DESIGN: Twenty-four patients received serial vaccinations with dendritic cell/myeloma fusion cells following posttransplant hematopoietic recovery. A second cohort of 12 patients received a pretransplant vaccine followed by posttransplant vaccinations. Dendritic cells generated from adherent mononuclear cells cultured with granulocyte macrophage colony-stimulating factor, interleukin-4, and TNF-α were fused with autologous bone marrow-derived myeloma fusion cells using polyethylene glycol. Fusion cells were quantified by determining the percentage of cells that coexpress dendritic cell and myeloma fusion antigens. RESULTS: The posttransplant period was associated with reduction in general measures of cellular immunity; however, an increase in CD4 and CD8(+) myeloma-specific T cells was observed after ASCT that was significantly expanded following posttransplant vaccination. Seventy-eight percent of patients achieved a best response of complete response (CR)+very good partial response (VGPR) and 47% achieved a CR/near CR (nCR). Remarkably, 24% of patients who achieved a partial response following transplant were converted to CR/nCR after vaccination and at more than 3 months posttransplant, consistent with a vaccine-mediated effect on residual disease. CONCLUSIONS: The posttransplant period for patients with multiple myeloma provides a unique platform for cellular immunotherapy in which vaccination with dendritic cell/myeloma fusion fusions resulted in the marked expansion of myeloma-specific T cells and cytoreduction of minimal residual disease.


Subject(s)
Cancer Vaccines/immunology , Dendritic Cells/immunology , Hematopoietic Stem Cell Transplantation , Multiple Myeloma/immunology , Multiple Myeloma/therapy , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cancer Vaccines/administration & dosage , Cancer Vaccines/adverse effects , Female , Humans , Immunity, Cellular , Immunotherapy , Male , Middle Aged , Remission Induction , Transplantation, Autologous , Treatment Outcome , Vaccination
3.
Cancer Immunol Immunother ; 62(1): 39-49, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22733396

ABSTRACT

Lenalidomide is an effective therapeutic agent for multiple myeloma that exhibits immunomodulatory properties including the activation of T and NK cells. The use of lenalidomide to reverse tumor-mediated immune suppression and amplify myeloma-specific immunity is currently being explored. In the present study, we examined the effect of lenalidomide on T-cell activation and its ability to amplify responses to a dendritic cell-based myeloma vaccine. We demonstrate that exposure to lenalidomide in the context of T-cell expansion with direct ligation of CD3/CD28 complex results in polarization toward a Th1 phenotype characterized by increased IFN-γ, but not IL-10 expression. In vitro exposure to lenalidomide resulted in decreased levels of regulatory T cells and a decrease in T-cell expression of the inhibitory marker, PD-1. Lenalidomide also enhanced T-cell proliferative responses to allogeneic DCs. Most significantly, lenalidomide treatment potentiated responses to the dendritic cell/myeloma fusion vaccine, which were characterized by increased production of inflammatory cytokines and increased cytotoxic lymphocyte-mediated lysis of autologous myeloma targets. These findings indicate that lenalidomide enhances the immunologic milieu in patients with myeloma by promoting T-cell proliferation and suppressing inhibitory factors, and thereby augmenting responses to a myeloma-specific tumor vaccine.


Subject(s)
Antineoplastic Agents/pharmacology , Cancer Vaccines/immunology , Dendritic Cells/drug effects , Immunity, Cellular/drug effects , Multiple Myeloma/immunology , T-Lymphocytes/drug effects , Thalidomide/analogs & derivatives , Cell Proliferation/drug effects , Dendritic Cells/immunology , Humans , Immunoblotting , Lenalidomide , Lymphocyte Activation/drug effects , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocytes/immunology , Thalidomide/pharmacology
4.
J Immunother ; 35(7): 555-69, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22892452

ABSTRACT

The fowl pox vector expressing the tumor-associated antigens, mucin-1 and carcinoembryonic antigen in the context of costimulatory molecules (rF-PANVAC) has shown promise as a tumor vaccine. However, vaccine-mediated expansion of suppressor T-cell populations may blunt clinical efficacy. We characterized the cellular immune response induced by ex vivo dendritic cells (DCs) transduced with (rF)-PANVAC. Consistent with the functional characteristics of potent antigen-presenting cells, rF-PANVAC-DCs demonstrated strong expression of mucin-1 and carcinoembryonic antigen and costimulatory molecules, CD80, CD86, and CD83; decreased levels of phosphorylated STAT3, and increased levels of Tyk2, Janus kinase 2, and STAT1. rF-PANVAC-DCs stimulated expansion of tumor antigen-specific T cells with potent cytolytic capacity. However, rF-PANVAC-transduced DCs also induced the concurrent expansion of FOXP3 expressing CD4CD25 regulatory T cells (Tregs) that inhibited T-cell activation. Moreover, Tregs expressed high levels of Th2 cytokines [interleukin (IL)-10, IL-4, IL-5, and IL-13] together with phosphorylated STAT3 and STAT6. In contrast, the vaccine-expanded Treg population expressed high levels of Th1 cytokines IL-2 and interferon-γ and the proinflammatory receptor-related orphan receptor γt (RORγt) and IL-17A suggesting that these cells may share effector functions with conventional TH17 T cells. These data suggest that Tregs expanded by rF-PANVAC-DCs, exhibit immunosuppressive properties potentially mediated by Th2 cytokines, but simultaneous expression of Th1 and Th17-associated factors suggests a high degree of plasticity.


Subject(s)
Cancer Vaccines/metabolism , Carcinoembryonic Antigen/metabolism , Dendritic Cells/metabolism , Membrane Glycoproteins/metabolism , Mucin-1/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , CD4 Antigens/metabolism , Cancer Vaccines/genetics , Carcinoembryonic Antigen/genetics , Cells, Cultured , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/virology , Forkhead Transcription Factors/metabolism , Fowlpox virus , Genetic Vectors , Humans , Interleukin-2 Receptor alpha Subunit/metabolism , Lymphocyte Activation , Membrane Glycoproteins/genetics , Mucin-1/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , STAT Transcription Factors/genetics , STAT Transcription Factors/metabolism , Th1-Th2 Balance , Transduction, Genetic
5.
J Acquir Immune Defic Syndr ; 60(3): 244-8, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22549384

ABSTRACT

The cellullar immune response is important in the containment of progressive multifocal leukoencephalopathy (PML). We examined program cell death-1 (PD-1) expression, a marker of cellular immune exhaustion, on T lymphocytes in PML. PD-1 expression was elevated on total CD4(+) and CD8(+) T cells (medians 36% and 24%) in PML patients compared with healthy control subjects (medians 14% and 18%; P = 0.0015 and P = 0.033). In PML patients, JC virus (JCV)-specific CD8(+) cytotoxic T lymphocytes expressed PD-1 more frequently than total CD8 T lymphocytes (means 39% and 78%, P = 0.0004). Blocking the PD-1 receptor increased JCV-specific T-cell immune response in a subgroup of PML patients.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Leukoencephalopathy, Progressive Multifocal/immunology , Programmed Cell Death 1 Receptor/metabolism , AIDS-Related Opportunistic Infections/complications , AIDS-Related Opportunistic Infections/immunology , AIDS-Related Opportunistic Infections/virology , Adult , Aged , Aged, 80 and over , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Case-Control Studies , Female , HIV Infections/complications , HIV Infections/immunology , HIV Infections/virology , Humans , Immunity, Cellular , JC Virus/immunology , Leukoencephalopathy, Progressive Multifocal/complications , Leukoencephalopathy, Progressive Multifocal/virology , Male , Middle Aged , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Young Adult
6.
Carbohydr Res ; 350: 6-13, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22285512

ABSTRACT

The nitric acid oxidation of D-glucose was reinvestigated in an effort to better understand and improve the oxidation and subsequent work up steps. The oxidation was carried out using a computer controlled reactor employing a closed reaction flask under an atmosphere of oxygen which allowed for a catalytic oxidation process with oxygen as the terminal oxidant. Removal of nitric acid from product included the use of both diffusion dialysis and nanofiltration methodologies. Product analysis protocols were developed using ion chromatography.


Subject(s)
Glucose/chemistry , Nitric Acid/chemistry , Chromatography, High Pressure Liquid , Diffusion , Filtration , Glucaric Acid/chemistry , Nanotechnology , Nitric Acid/isolation & purification , Oxidation-Reduction
7.
J Immunother ; 34(5): 409-18, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21577144

ABSTRACT

We have developed a cancer vaccine in which autologous tumor is fused with dendritic cells (DCs) resulting in the presentation of tumor antigens in the context of DC-mediated costimulation. In clinical trials, immunologic responses have been observed, however responses may be muted by inhibitory pathways. The PD1/PDL1 pathway is an important element contributing to tumor-mediated immune suppression. In this study, we demonstrate that myeloma cells and DC/tumor fusions strongly express PD-L1. Compared with a control population of normal volunteers, increased PD-1 expression was observed on T cells isolated from patients with myeloma. It is interesting to note that after autologous transplantation, T-cell expression of PD-1 returned to levels seen in normal controls. We examined the effect of PD-1 blockade on T-cell response to DC/tumor fusions ex vivo. Presence of CT-011, an anti-PD1 antibody, promoted the vaccine-induced T-cell polarization towards an activated phenotype expressing Th1 compared with Th2 cytokines. A concomitant decrease in regulatory T cells and enhanced killing in a cytotoxicity assay was observed. In summary, we demonstrate that PD-1 expression is increased in T cells of patients with active myeloma, and that CT-011 enhances activated T-cell responses after DC/tumor fusion stimulation.


Subject(s)
Antigens, CD/immunology , Apoptosis Regulatory Proteins/immunology , Cancer Vaccines , Dendritic Cells/immunology , Leukocytes, Mononuclear/immunology , Multiple Myeloma/immunology , Multiple Myeloma/pathology , Antibodies, Anti-Idiotypic/immunology , Antigens, CD/genetics , Antigens, CD/metabolism , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , B7-H1 Antigen , Cancer Vaccines/immunology , Cancer Vaccines/pharmacology , Cell Fusion , Cell Proliferation/drug effects , Cytotoxicity, Immunologic , Dendritic Cells/chemistry , Dendritic Cells/metabolism , Flow Cytometry , Humans , In Vitro Techniques , Leukocytes, Mononuclear/chemistry , Leukocytes, Mononuclear/metabolism , Lymphocyte Activation/immunology , Multiple Myeloma/metabolism , Multiple Myeloma/therapy , Programmed Cell Death 1 Receptor , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Regulatory/immunology , Th1-Th2 Balance/drug effects , Transplantation, Autologous
8.
J Immunother ; 33(2): 155-66, 2010.
Article in English | MEDLINE | ID: mdl-20145548

ABSTRACT

Adoptive immunotherapy with tumor-specific T cells represents a promising treatment strategy for patients with malignancy. However, the efficacy of T-cell therapy has been limited by the ability to expand tumor-reactive cells with an activated phenotype that effectively target malignant cells. We have developed an anticancer vaccine in which patient-derived tumor cells are fused with autologous dendritic cells (DCs), such that a wide array of tumor antigens are presented in the context of DC-mediated costimulation. In this study, we demonstrate that DC/tumor fusions induce T cells that react with tumor and are dramatically expanded by subsequent ligation of the CD3/CD28 costimulatory complex. These T cells exhibit a predominantly activated phenotype as manifested by an increase in the percentage of cells expressing CD69 and interferon gamma. In addition, the T cells upregulate granzyme B expression and are highly effective in lysing autologous tumor targets. Targeting of tumor-specific antigen was demonstrated by the expansion of T cells with specificity for the MUC1 tetramer. Stimulation with anti-CD3/CD28 followed by DC/tumor fusions or either agent alone failed to result in a similar expansion of tumor-reactive T cells. Consistent with these findings, spectratyping analysis demonstrates selective expansion of T-cell clones as manifested by considerable skewing of the Vbeta repertoire following sequential stimulation with DC/tumor fusions and anti-CD3/CD28. Gene expression analysis was notable for the upregulation of inflammatory pathways. These findings indicate that stimulation with DC/tumor fusions provides a unique platform for subsequent expansion with anti-CD3/CD28 in adoptive T-cell therapy of cancer.


Subject(s)
Antigens, CD/biosynthesis , Antigens, Differentiation/biosynthesis , Dendritic Cells/metabolism , Granzymes/biosynthesis , T-Lymphocytes/metabolism , Antigen Presentation/immunology , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, Differentiation/genetics , Antigens, Differentiation/immunology , Cancer Vaccines , Cell Fusion , Cell Proliferation , Clone Cells , Cytotoxicity, Immunologic , Dendritic Cells/immunology , Dendritic Cells/pathology , Genes, T-Cell Receptor beta , Granzymes/genetics , Humans , Interferon-gamma/metabolism , Lymphocyte Activation , Mucin-1/immunology , Mucin-1/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...