Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Ther Oncolytics ; 5: 87-96, 2017 Jun 16.
Article in English | MEDLINE | ID: mdl-28812060

ABSTRACT

Oncolytic viruses (OVs) are emerging as powerful anti-cancer agents and are currently being tested for their safety and efficacy in patients. Reovirus (Reolysin), a naturally occurring non-pathogenic, double-stranded RNA virus, has natural oncolytic activity and is being tested in phase I-III clinical trials in a variety of tumor types. With its recent US Food and Drug Administration (FDA) orphan drug designation for several tumor types, Reolysin is a potential therapeutic agent for various cancers, including head and neck squamous cell carcinomas (HNSCCs), which have a 5-year survival of ∼55%. Histone deacetylase inhibitors (HDACis) comprise a structurally diverse class of compounds with targeted anti-cancer effects. The first FDA-approved HDACi, vorinostat (suberoylanilide hydroxamic acid [SAHA]), is currently being tested in patients with head and neck cancer. Recent findings indicate that HDAC inhibition in myeloma cells results in the upregulation of the Reolysin entry receptor, junctional adhesion molecule 1 (JAM-1), facilitating reovirus infection and tumor cell killing both in vitro and in vivo. In this study, we tested the anti-tumor efficacy of HDAC inhibitors AR-42 or SAHA in conjunction with Reolysin in HNSCCs. While HDAC inhibition increased JAM-1 and reovirus entry, the impact of this combination therapy was tested on the development of anti-tumor immune responses.

2.
Melanoma Res ; 26(4): 329-37, 2016 08.
Article in English | MEDLINE | ID: mdl-27035691

ABSTRACT

The folate receptor (FR) is overexpressed on the vascular side of cancerous cells including those of the breast, ovaries, testes, and cervix. We hypothesized that a folate-conjugated immunoglobulin (F-IgG) would bind to the FR that is overexpressed on melanoma tumor cells to target these cells for lysis by natural killer (NK) cells. Folate receptor expression was confirmed in the Mel-39 (human melanoma) cell line by flow cytometry and immunoblot analysis using KB (human oral epithelial) and F01 (human melanoma) as a positive and a negative control, respectively. FR-positive and FR-negative cell lines were treated with F-IgG or control immunoglobulin G in the presence or absence of cytokines to determine NK cell ability to lyse FR-positive cell lines. NK cell activation was significantly upregulated and lysis of Mel 39 tumor cells increased following treatment with F-IgG compared with control immunoglobulin G at all effector : target (E : T) ratios (P<0.01). This trend further increased by NK cell stimulation with the activating cytokine interleukin-12. NK cell production of cytokines such as interferon-gamma, macrophage inflammatory protein 1α, and regulated on activation normal T-cell expressed and secreted (RANTES) was also significantly increased in response to costimulation with interleukin-12 stimulation and F-IgG-coated Mel 39 target cells compared with controls (P<0.01). In contrast, F-IgG did not bind to the FR-negative cell line F01 and had no significant effect on NK cell lysis or cytokine production. This research indicates the potential use of F-IgG for its ability to induce an immune response from NK cells against FR-positive melanoma tumor cells, which can be further increased by the addition of cytokines.


Subject(s)
Folic Acid/administration & dosage , Immunoconjugates/administration & dosage , Killer Cells, Natural/immunology , Melanoma/drug therapy , Skin Neoplasms/drug therapy , Antibody-Dependent Cell Cytotoxicity , Folate Receptor 1/biosynthesis , Folate Receptor 1/immunology , Humans , Immunoconjugates/immunology , Immunoglobulin G/immunology , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Interleukin-12/immunology , KB Cells , Melanoma/immunology , Skin Neoplasms/immunology
3.
Cancer Immunol Res ; 4(4): 323-336, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26865456

ABSTRACT

Optimally effective antitumor therapies would not only activate immune effector cells but also engage them at the tumor. Folate conjugated to immunoglobulin (F-IgG) could direct innate immune cells with Fc receptors to folate receptor-expressing cancer cells. F-IgG bound to human KB and HeLa cells, as well as murine L1210JF, a folate receptor (FR)-overexpressing cancer cell line, as determined by flow cytometry. Recognition of F-IgG by natural killer (NK) cell Fc receptors led to phosphorylation of the ERK transcription factor and increased NK cell expression of CD69. Lysis of KB tumor cells by NK cells increased by about 5-fold after treatment with F-IgG, an effect synergistically enhanced by treatment with IL2, IL12, IL15, or IL21 (P< 0.001). F-IgG also enhanced the lysis of chronic lymphocytic leukemia cells by autologous NK cells. NK cells significantly increased production of IFNγ, MIP-1α, and RANTES in response to F-IgG-coated KB target cells in the presence of the NK cell-activating cytokine IL12, and these coculture supernatants induced significant T-cell chemotaxis (P< 0.001). F-IgG-coated targets also stimulated FcR-mediated monocyte effector functions. Studies in a murine leukemia model confirmed the intratumoral localization and antitumor activity of F-IgG, as well as enhancement of its effects by IL12 (P =0.05). The antitumor effect of this combination was dependent on NK cells and led to decreased tumor cell proliferation in vivo Thus, F-IgG can induce an immune response against FR-positive tumor cells that is mediated by NK cells and can be augmented by cytokine therapy.


Subject(s)
Cytokines/metabolism , Cytotoxicity, Immunologic , Folic Acid/administration & dosage , Immunoconjugates/immunology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Neoplasms/immunology , Neoplasms/metabolism , Animals , Antibody-Dependent Cell Cytotoxicity/immunology , Cell Line, Tumor , Cytotoxicity, Immunologic/drug effects , Disease Models, Animal , Female , Folate Receptor 1/genetics , Folate Receptor 1/metabolism , Gene Expression , Humans , Immunoglobulin G/immunology , Immunomodulation , Interleukin-12/biosynthesis , Lymphocyte Activation/immunology , Mice , Monocytes/immunology , Monocytes/metabolism , Neoplasms/genetics , Neoplasms/pathology , Tumor Burden/immunology , Xenograft Model Antitumor Assays
4.
Cancer Res ; 71(15): 5101-10, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21680779

ABSTRACT

Our group and others have determined that immune effector cells from patients with advanced cancers exhibit reduced activation of IFN signaling pathways. We hypothesized that increases in immune regulatory cells termed myeloid-derived suppressor cells (MDSC) could interfere with the host immune response to tumors by inhibiting immune cell responsiveness to IFNs. The C26 murine adenocarcinoma model was employed to study immune function in advanced malignancy. C26-bearing mice had significantly elevated levels of GR1(+)CD11b(+) MDSC as compared with control mice, and splenocytes from tumor-bearing mice exhibited reduced phosphorylation of STAT1 (P-STAT1) on Tyr(701) in response to IFN-α or IFN-γ. This inhibition was seen in splenic CD4(+) and CD8(+) T cells as well as natural killer cells. In vitro coculture experiments revealed that MDSC inhibited the IFN responsiveness of splenocytes from normal mice. Treatment of C26-bearing mice with gemcitabine or an anti-GR1 antibody led to depletion of MDSC and restored splenocyte IFN responsiveness. Spleens from C26-bearing animals displayed elevated levels of iNOS protein and nitric oxide. In vitro treatment of splenocytes with a nitric oxide donor led to a decreased STAT1 IFN response. The elevation in nitric oxide in C26-bearing mice was associated with increased levels of nitration on STAT1. Finally, splenocytes from iNOS knockout mice bearing C26 tumors exhibited a significantly elevated IFN response as compared with control C26 tumor-bearing mice. These data suggest that nitric oxide produced by MDSC can lead to reduced IFN responsiveness in immune cells.


Subject(s)
Adenocarcinoma/immunology , Colonic Neoplasms/immunology , Interferons/antagonists & inhibitors , Myeloid Cells/physiology , Tumor Escape/immunology , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Animals , CD11b Antigen/analysis , Coculture Techniques , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Female , Interferon Type I/pharmacology , Interferon-gamma/pharmacology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Mice, Knockout , Neoplasm Proteins/metabolism , Nitric Oxide/metabolism , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase Type II/analysis , Nitric Oxide Synthase Type II/deficiency , Phosphorylation , Protein Processing, Post-Translational , Receptors, Cell Surface/analysis , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Cell Surface/immunology , Receptors, Interferon , Recombinant Proteins , STAT1 Transcription Factor/metabolism , Spleen/enzymology , Spleen/pathology , Gemcitabine , Interferon gamma Receptor
SELECTION OF CITATIONS
SEARCH DETAIL
...