Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Haematologica ; 100(3): 336-44, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25480499

ABSTRACT

Current therapies for acute myeloid leukemia are associated with high failure and relapse rates. Adoptive immunotherapies, which have shown promise in the treatment of hematologic malignancies, have the potential to target acute myeloid leukemia through pathways that are distinct and complementary to current approaches. Here, we describe the development of a novel adoptive immunotherapy specific for this disease. We generated a second generation CD33-specific chimeric antigen receptor capable of redirecting cytolytic effector T cells against leukemic cells. CD33 is expressed in approximately 90% of acute myeloid leukemia cases and has demonstrated utility as a target of therapeutic antibodies. Chimeric antigen receptor-modified T cells efficiently killed leukemia cell lines and primary tumor cells in vitro. The anti-leukemia effect was CD33-specific, mediated through T-cell effector functions, and displayed tumor lysis at effector:target ratios as low as 1:20. Furthermore, the CD33-redirected T cells were effective in vivo, preventing the development of leukemia after prophylactic administration and delaying the progression of established disease in mice. These data provide pre-clinical validation of the effectiveness of a second-generation anti-CD33 chimeric antigen receptor therapy for acute myeloid leukemia, and support its continued development as a clinical therapeutic.


Subject(s)
Immunotherapy, Adoptive/methods , Leukemia, Myeloid, Acute/therapy , Mutant Chimeric Proteins/immunology , Receptors, Antigen, T-Cell/immunology , Sialic Acid Binding Ig-like Lectin 3/antagonists & inhibitors , T-Lymphocytes, Cytotoxic/immunology , Animals , Bone Marrow/immunology , Bone Marrow/pathology , Cell Line, Tumor , Coculture Techniques , Cytotoxicity, Immunologic , Gene Expression , Humans , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin Receptor Common gamma Subunit/genetics , Interleukin Receptor Common gamma Subunit/immunology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/pathology , Liver/immunology , Liver/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Mutant Chimeric Proteins/genetics , Plasmids/chemistry , Plasmids/genetics , Receptors, Antigen, T-Cell/genetics , Sialic Acid Binding Ig-like Lectin 3/genetics , Sialic Acid Binding Ig-like Lectin 3/immunology , Spleen/immunology , Spleen/pathology , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/transplantation , Transfection
2.
Head Neck ; 36(11): 1619-27, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24115092

ABSTRACT

BACKGROUND: Recombinant vesicular stomatitis virus expressing interferon-ß (VSV-IFN-ß) has demonstrated antitumor activity in vitro and in vivo. In preparation for clinical testing in human squamous cell carcinoma (SCC) of the head and neck, we conducted preclinical studies of VSV-IFN-ß in syngeneic SCC models. METHODS: In vitro, VSV-IFN-ß (expressing rat or mouse interferon [IFN]-ß)-induced cytotoxicity and propagated in rat (FAT-7) or mouse (SCC-VII) SCC cells during normoxia and hypoxia. In vivo, intratumoral administration of VSV-rat-IFN-ß or VSV-human-IFN-ß in FAT-7 bearing or non-tumor bearing immunocompetent rats did not result in acute organ toxicity or death. RESULTS: VSV-r-IFN-ß replicated predominantly in tumors and a dose dependent anti-VSV antibody response was observed. Intratumoral or intravenous administration of VSV-IFN-ß resulted in growth delay and improved survival compared with controls. CONCLUSION: The above data confirm safety and feasibility of VSV-IFN-ß administration in immunocompetent animals and support its clinical evaluation in advanced human head and neck cancer.


Subject(s)
Carcinoma, Squamous Cell/therapy , Head and Neck Neoplasms/therapy , Interferon-beta/pharmacology , Oncolytic Virotherapy/methods , Vesiculovirus/isolation & purification , Analysis of Variance , Animals , Blotting, Western , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/pathology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Feasibility Studies , Female , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/pathology , Humans , Immunocompetence , Kaplan-Meier Estimate , Mice , Random Allocation , Rats , Rats, Inbred F344 , Safety , Statistics, Nonparametric , Transplantation, Isogeneic , Treatment Outcome , Tumor Cells, Cultured , Vesicular Stomatitis/virology
3.
Cell Immunol ; 279(1): 60-5, 2012 Sep.
Article in English | MEDLINE | ID: mdl-23089195

ABSTRACT

Circulating Foxp3(+) regulatory T cells (Treg) may arise in the thymus (natural Treg, nTreg) or through the adaptive upregulation of Foxp3 after T cell activation (induced Treg, iTreg). In this brief review, we explore evidence for the formation and function of iTreg during pathologic conditions. Determining the ontogeny and function of Treg populations has relied on the use of manipulated systems in which either iTreg or nTreg are absent, or lineage tracing of T cell clones through repertoire analyses. iTreg appear particularly important at mucosal interfaces. iTreg can also ameliorate tissue-specific autoimmunity and are a prominent source of tumor-infiltrating Treg in some models. However, under many conditions, including in CNS autoimmunity, diabetes, and some tumor systems, iTreg formation appears limited. The immunological contribution of iTreg is thus highly context dependent. Deciphering immune parameters responsible for iTreg formation and their role in modulating pathologic immune responses will be important.


Subject(s)
Forkhead Transcription Factors/immunology , Immune Tolerance/immunology , Lymphocyte Activation/immunology , T-Lymphocytes, Regulatory/immunology , Autoimmune Diseases of the Nervous System/immunology , Autoimmune Diseases of the Nervous System/pathology , Diabetes Mellitus/immunology , Diabetes Mellitus/pathology , Forkhead Transcription Factors/metabolism , Gastrointestinal Tract/immunology , Gastrointestinal Tract/pathology , Humans , Models, Immunological , Neoplasms/immunology , Neoplasms/pathology , T-Lymphocytes, Regulatory/metabolism
4.
Methods Mol Biol ; 797: 217-38, 2012.
Article in English | MEDLINE | ID: mdl-21948479

ABSTRACT

Oncolytic viruses, the use of viruses to treat cancer, is emerging as a new option for cancer therapy. Oncolytic viruses, of both DNA and RNA origin, exhibit the ability to preferentially replicate in and kill cancer cells plausibly due to defects in innate immune signaling or translation regulation that are acquired during cellular transformation. Here, we review concepts and assays that describe how to analyze signaling pathways that govern the regulation of Type I IFN production as well as the induction of interferon-stimulated antiviral genes, events that are critical for mounting an effective antiviral response. The following procedures can be used to assess whether innate immune pathways that control antiviral host defense are defective in tumor cells - mechanisms that may help to explain viral oncolysis.


Subject(s)
Immunity, Innate , Interferon Type I/metabolism , Neoplasms/immunology , Oncolytic Viruses/immunology , Signal Transduction/immunology , Antiviral Agents/pharmacology , Cell Line, Transformed , Cell Line, Tumor , Gene Expression Regulation , Humans , Immunologic Techniques , Interferon Type I/genetics , Interferon Type I/immunology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Neoplasms/therapy , Oncolytic Virotherapy , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism
5.
Chin J Cancer ; 30(12): 805-14, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22059911

ABSTRACT

In the last decade, we have gained significant understanding of the mechanism by which vesicular stomatitis virus (VSV) specifically kills cancer cells. Dysregulation of translation and defective innate immunity are both thought to contribute to VSV oncolysis. Safety and efficacy are important objectives to consider in evaluating VSV as a therapy for malignant disease. Ongoing efforts may enable VSV virotherapy to be considered in the near future to treat drug-resistant ovarian cancer when other options have been exhausted. In this article, we review the development of VSV as a potential therapeutic approach for recurrent or drug-resistant ovarian cancer.


Subject(s)
Oncolytic Virotherapy/methods , Ovarian Neoplasms/therapy , Vesicular stomatitis Indiana virus/physiology , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Cell Proliferation , Drug Resistance, Neoplasm , Female , Humans , Neoplasm Recurrence, Local , Ovarian Neoplasms/pathology , Ovarian Neoplasms/virology , Virus Replication
6.
J Virol ; 85(20): 10440-50, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21813611

ABSTRACT

Vesicular stomatitis virus (VSV), a negative-strand RNA rhabdovirus, preferentially replicates in and eradicates transformed versus nontransformed cells and is thus being considered for use as a potential anticancer treatment. The genetic malleability of VSV also affords an opportunity to develop more potent agents that exhibit increased therapeutic activity. The tumor suppressor p53 has been shown to exert potent antitumor properties, which may in part involve stimulating host innate immune responses to malignancies. To evaluate whether VSV expressing p53 exhibited enhanced oncolytic action, the murine p53 (mp53) gene was incorporated into recombinant VSVs with or without a functional viral M gene-encoded protein that could either block (VSV-mp53) or enable [VSV-M(mut)-mp53] host mRNA export following infection of susceptible cells. Our results indicated that VSV-mp53 and VSV-M(mut)-mp53 expressed high levels of functional p53 and retained the ability to lyse transformed versus normal cells. In addition, we observed that VSV-ΔM-mp53 was extremely attenuated in vivo due to p53 activating innate immune genes, such as type I interferon (IFN). Significantly, immunocompetent animals with metastatic mammary adenocarcinoma exhibited increased survival following treatment with a single inoculation of VSV-ΔM-mp53, the mechanisms of which involved enhanced CD49b+ NK and tumor-specific CD8+ T cell responses. Our data indicate that VSV incorporating p53 could provide a safe, effective strategy for the design of VSV oncolytic therapeutics and VSV-based vaccines.


Subject(s)
Oncolytic Viruses/growth & development , Tumor Suppressor Protein p53/metabolism , Vesiculovirus/growth & development , Adenocarcinoma/drug therapy , Adenocarcinoma/secondary , Animals , Disease Models, Animal , Female , Mammary Neoplasms, Animal/secondary , Mammary Neoplasms, Animal/therapy , Mice , Mice, Inbred BALB C , Mice, Nude , Oncolytic Virotherapy/methods , Oncolytic Viruses/genetics , Oncolytic Viruses/pathogenicity , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Rodent Diseases/therapy , Treatment Outcome , Tumor Suppressor Protein p53/genetics , Vesiculovirus/genetics , Vesiculovirus/pathogenicity
7.
J Virol ; 85(19): 9716-25, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21795329

ABSTRACT

Eukaryotic translation initiation factor 2B (eIF2B) is a heteropentameric guanine nucleotide exchange factor that converts protein synthesis initiation factor 2 (eIF2) from a GDP-bound form to the active eIF2-GTP complex. Cellular stress can repress translation initiation by activating kinases capable of phosphorylating the alpha subunit of eIF2 (eIF2α), which sequesters eIF2B to prevent exchange activity. Previously, we demonstrated that tumor cells are sensitive to viral replication, possibly due to the occurrence of defects in eIF2B that overcome the inhibitory effects of eIF2α phosphorylation. To extend this analysis, we have investigated the importance of eIF2Bα function and report that this subunit can functionally substitute for its counterpart, GCN3, in yeast. In addition, a variant of mammalian eIF2Bα harboring a point mutation (T41A) was able overcome translational inhibition invoked by amino acid depravation, which activates Saccharomyces cerevisiae GCN2 to phosphorylate the yeast eIF2α homolog SUI2. Significantly, we also demonstrate that the loss of eIF2Bα, or the expression of the T41A variant in mammalian cells, is sufficient to neutralize the consequences of eIF2α phosphorylation and render normal cells susceptible to virus infection. Our data emphasize the importance of eIF2Bα in mediating the eIF2 kinase translation-inhibitory activity and may provide insight into the complex nature of viral oncolysis.


Subject(s)
Eukaryotic Initiation Factor-2B/metabolism , Vesiculovirus/growth & development , Virus Replication , Amino Acid Substitution/genetics , Animals , Cells, Cultured , Eukaryotic Initiation Factor-2B/deficiency , Eukaryotic Initiation Factor-2B/genetics , Genetic Complementation Test , Humans , Mice , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutation, Missense , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae Proteins
8.
Gynecol Oncol ; 116(2): 269-75, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19932656

ABSTRACT

OBJECTIVE: Current treatment options for epithelial ovarian cancer are limited and therapeutic development for recurrent and drug-resistant ovarian cancer is an urgent agenda. We investigated the potential use of genetically engineered Vesicular Stomatitis Virus (VSV) to treat ovarian cancer patients who fail to respond to available therapies. Specifically, we examined the toxicity to hosts and specificity of targeting ovarian tumors using a Wv ovarian tumor model. METHODS: We first tested recombinant VSV for oncolytic activity in a panel of human ovarian epithelial cancer, immortalized, and primary ovarian surface epithelial cells in culture. Then, we tested VSV oncolytic therapy using the immune competent Wv mice that develop tubular adenomas, benign tumor lesions derived from ovarian surface epithelial cells. RESULTS: The expression of GFP encoded by the recombinant VSV genome was detected in about 5% of primary ovarian surface epithelial cells (3 lines) up to 30 days without significantly altering the growth pattern of the cells, suggesting the lack of toxicity to the normal ovarian surface epithelial cells. However, VSV-GFP was detected in the majority (around 90%) of cells that are either "immortalized" by SV40 antigen expression or cancer lines. Some variation in killing time courses was observed, but all the transformed cell lines were killed within 3 days. We found that regardless of the inoculation route (intra bursal, IP, or IV), VSV specifically infected and replicated in the in situ ovarian tumors in the Wv mice without significant activity in any other organs and tissues, and showed no detectable toxicity. The epithelial tumor lesions were greatly reduced in VSV-targeted ovarian tumors in the Wv mice. CONCLUSIONS: VSV oncolytic activity depends on a cell autonomous property distinguishing primary and transformed cells. The efficient oncolytic activity of VSV for the "immortalized" non-tumorigenic ovarian surface epithelial cells suggests that the selective specificity extends from pre-neoplastic to overt cancer cells. The results demonstrated the explicit targeting of ovarian epithelial tumors by VSV in immune competent, ovarian tumor-bearing mouse models, and further support the utility of VSV as an effective and safe anti-cancer agent.


Subject(s)
Oncolytic Virotherapy/methods , Ovarian Neoplasms/therapy , Ovarian Neoplasms/virology , Vesicular stomatitis Indiana virus/physiology , Animals , Cell Line, Tumor , Cell Transformation, Viral , Female , Humans , Mice , Vesicular stomatitis Indiana virus/pathogenicity , Virus Replication , Xenograft Model Antitumor Assays
9.
EMBO Rep ; 10(10): 1154-60, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19745842

ABSTRACT

Vesicular stomatitis virus (VSV) infects and kills a wide range of cell types; however, the mechanisms involved in VSV-mediated cell death are not fully understood. Here we show that VSV infection interferes with mitotic progression, resulting in cell death. This effect requires the interaction of VSV matrix (M) protein with the Rae1-Nup98 complex in mitosis, which is associated with a subset of ribonucleoproteins (RNPs). VSV displaced Rae1 from spindle poles, caused spindle abnormalities and triggered substantial cell death during metaphase. These effects were attenuated in cells infected with VSV expressing a mutant M protein that does not bind efficiently to the Rae1-Nup98-RNP complex. In cells that progressed to late mitosis, M protein prevented proper nuclear formation and chromatin decondensation. VSV is an oncolytic (anti-tumour) agent as it preferentially replicates and kills tumour cells. As tumour cells have a high mitotic index, VSV-mediated mitotic cell death probably contributes to its oncolytic activity.


Subject(s)
Cell Death , Mitosis , Vesiculovirus/physiology , Animals , Cell Line , Cell Nucleus/metabolism , Humans , Nuclear Matrix-Associated Proteins/metabolism , Nuclear Pore Complex Proteins/metabolism , Nucleocytoplasmic Transport Proteins/metabolism , Oocytes/metabolism , Protein Binding , Rats , Ribonucleoproteins/metabolism , Viral Matrix Proteins/metabolism , Xenopus
SELECTION OF CITATIONS
SEARCH DETAIL
...