Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Neuromolecular Med ; 15(2): 420-34, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23575987

ABSTRACT

Glioblastoma represents one of the most challenging problems in neurooncology. Among key elements driving its behavior is the transmembrane epidermal growth factor receptor family, with the first member epidermal growth factor receptor (EGFR) centered in most studies. Engagement of the extracellular domain with a ligand activates the intracellular tyrosine kinase (TK) domain of EGFR, leading to autophosphorylation and signal transduction that controls proliferation, gene transcription, and apoptosis. Oncogenic missense mutations, deletions, and insertions in the EGFR gene are preferentially located in the extracellular domain in glioblastoma and cause constitutive activation of the receptor. The mutant EGFR may also transactivate other cell surface molecules, such as additional members of the EGFR family and the platelet-derived growth factor receptor, which ignite signaling cascades that synergize with the EGFR-initiated cascade. Because of the cell surface location and increased expression of the receptor along with its important biological function, EGFR has triggered much effort for designing targeted therapy. These approaches include TK inhibition, monoclonal antibody, vaccine, and RNA-based downregulation of the receptor. Treatment success requires that the drug penetrates the blood-brain barrier and has low systemic toxicity but high selectivity for the tumor. While the blockade of EGFR-dependent processes resulted in experimental and clinical treatment success, cells capable of using alternative signaling ultimately escape this strategy. A combination of interventions targeting tumor-specific cell surface regulators along with convergent downstream signaling pathways will likely enhance efficacy. Studies on EGFR in glioblastoma have revealed much information about the complexity of gliomagenesis and also facilitated the development of strategies for targeting drivers of tumor growth and combination therapies with increasing complexity.


Subject(s)
Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , ErbB Receptors/antagonists & inhibitors , Glioblastoma/drug therapy , Molecular Targeted Therapy , Neoplasm Proteins/antagonists & inhibitors , Nerve Tissue Proteins/antagonists & inhibitors , Protein Kinase Inhibitors/therapeutic use , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/pharmacology , Brain Neoplasms/enzymology , Brain Neoplasms/genetics , Brain Neoplasms/therapy , Cancer Vaccines , Cell Transformation, Neoplastic , Clinical Trials as Topic , Drug Screening Assays, Antitumor , ErbB Receptors/genetics , ErbB Receptors/immunology , Gene Expression Regulation, Neoplastic , Genetic Therapy , Glioblastoma/enzymology , Glioblastoma/genetics , Glioblastoma/therapy , Humans , Mutation , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/immunology , Oligonucleotides, Antisense/therapeutic use , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational/drug effects , Protein Structure, Tertiary , RNA Interference , RNA, Catalytic/therapeutic use , RNA, Small Interfering/therapeutic use , Signal Transduction/drug effects
2.
Cancer Gene Ther ; 18(2): 123-34, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20930860

ABSTRACT

We exploited the differential activation of hypoxia-inducible factor (HIF)-dependent gene expression in tumors versus normal tissue for the design of a targeted oncolytic herpes simplex virus type-1 (HSV-1). A gene that is essential for viral replication, infected cell polypeptide 4 (ICP4), was placed under the regulation of an HIF-responsive promoter and then introduced into the thymidine kinase locus (U(L)23) of HSV d120, which contains partial deletions in the two endogenous ICP4 genes. Recombinant HIF-HSV was isolated and their derivation from d120 was verified by expression of a truncated, non-functional form of ICP4 protein. Disruption of the U(L)23 locus was confirmed by loss of thymidine kinase expression and resistance to acyclovir. Unexpectedly, HIF-HSV expressed ICP4 and induced tumor cell lysis at similar levels under normoxia and hypoxia. The lack of HIF-dependent ICP4 transgene expression by HIF-HSV was due to two factors that have not previously been reported-reversion of the ICP4 gene region to its wild-type configuration and increased HIF-transcriptional activity under normoxia when cells were infected with any strain of HSV-1. The findings that an oncolytic HSV-1 is genetically unstable and can activate a tumor-related promoter in a non-specific manner have important implications for any proposed use of this virus in cancer therapy.


Subject(s)
Herpesvirus 1, Human/genetics , Hypoxia-Inducible Factor 1/genetics , Oncolytic Viruses/genetics , Animals , Cell Line, Tumor , Chlorocebus aethiops , Gene Deletion , Gene Expression Regulation, Viral , Glioma , Herpesvirus 1, Human/metabolism , Humans , Immediate-Early Proteins/biosynthesis , Immediate-Early Proteins/genetics , Oncolytic Viruses/metabolism , Promoter Regions, Genetic , Transcriptional Activation , Transfection , Vero Cells , Virus Replication/genetics
3.
Gene Ther ; 17(12): 1430-41, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20664541

ABSTRACT

There is a need to develop more potent oncolytic adenoviruses (Ads) that show increased antitumor activity in patients. The HYPR-Ads are targeted oncolytic Ads that specifically kill tumor cells, which express active hypoxia-inducible factor (HIF). While therapeutically efficacious, the HYPR-Ads showed attenuated replication and oncolytic activity. To overcome these deficiencies and improve antitumor efficacy, we created new HIF-activated oncolytic Ads, HIF-Ad and HIF-Ad-IL4, which have two key changes: (i) a modified HIF-responsive promoter to regulate the E1A replication gene and (ii) insertion of the E3 gene region. The HIF-Ads showed conditional activation of E1A expression under hypoxia. Importantly, the HIF-Ads show hypoxia-dependent replication, oncolytic and cellular release activities, and potent antitumor efficacy, all of which are significantly greater than that of the HYPR-Ads. Notably, HIF-Ad-IL4 treatment led to regressions in tumor size by 70% and extensive tumor infiltration by leukocytes resulting in an antitumor efficacy that is up to six-fold greater than that of the HYPR-Ads, HIF-Ad and wild-type Ad treatment. These studies show that treatment with an HIF-activated oncolytic Ad leads to a measurable therapeutic response. The novel design of the HIF-Ads represents a significant improvement compared with first-generation oncolytic Ads and has great potential to increase the efficacy of this cancer therapy.


Subject(s)
Adenoviridae/physiology , Antineoplastic Agents/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Gene Expression Regulation , Genetic Therapy/methods , Genetic Vectors/standards , Virus Replication , Adenoviridae/genetics , Adenovirus E1A Proteins/genetics , Adenovirus E1A Proteins/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Hypoxia/physiology , Genetic Therapy/standards , Genetic Vectors/genetics , HEK293 Cells , Humans , Interleukin-4/genetics , Interleukin-4/metabolism , Neoplasms/therapy , Oncolytic Viruses/genetics , Oncolytic Viruses/physiology , Tumor Cells, Cultured , Virus Release
4.
Gene Ther ; 11(15): 1215-23, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15141159

ABSTRACT

Increasing evidence indicates that survivin, an inhibitor of apoptosis protein (IAP), is expressed in human cancer cells but is absent from most normal adult tissues. Here, we examined the feasibility of using a survivin promoter (Sur-P) to direct therapeutic expression of a proapoptotic gene specifically in human tumor cells. First, we demonstrated that this promoter was highly active in human tumor cells but not in normal cells. Second, we found that Sur-P activity was upregulated by hypoxia in tumor cells. Third, to further enhance this promoter's activity under hypoxia, we added a hypoxia-responsive element (HRE) from the vascular endothelial growth factor gene promoter in its 5' region, and showed that this combination resulted in a further increase in the level of gene expression in hypoxic tumor cells. Finally, we demonstrated that expression of an autocatalytic reverse caspase-3 gene by this promoter specifically induced apoptotic cell death in human tumor cells but not in normal cells. These findings support the use of promoters Sur-P or chimeric HRE-Sur-P for generating novel vectors for cancer gene therapy.


Subject(s)
Genetic Therapy/methods , Hypoxia/metabolism , Microtubule-Associated Proteins/genetics , Neoplasms/therapy , Promoter Regions, Genetic , Vascular Endothelial Growth Factor A/genetics , Apoptosis , Blotting, Western/methods , Breast Neoplasms/therapy , Caspase 3 , Caspase Inhibitors , Cell Line , Cell Line, Tumor , Female , Gene Expression , Humans , Inhibitor of Apoptosis Proteins , Neoplasm Proteins , Neoplasms/pathology , Pancreatic Neoplasms/therapy , Recombinant Fusion Proteins/therapeutic use , Reverse Transcriptase Polymerase Chain Reaction , Survivin
5.
Gene Ther ; 8(23): 1801-7, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11803400

ABSTRACT

Hypoxia initiates an adaptive physiological response in all organisms and plays a role in the pathogenesis of several human diseases. The hypoxia/HIF-inducible factor-1 (HIF-1) transcription factor mediates transcriptional responses to hypoxia by binding to a cis-acting hypoxia-responsive element (HRE) present within target genes. The use of the HIF-1/HRE system of gene regulation can be utilized as a mechanism to target expression of therapeutic genes to hypoxic cells or cells that have a constitutively active HIF-1/HRE pathway due to cell transformation. Given the rapid resistance of tumors to single therapeutic strategies, new vector systems need to be developed that can deliver multimodal therapy. Here we show that HREs function as classical enhancer elements and function bidirectionally to co-regulate the expression of two genes. We designed a large series of novel bidirectional hypoxia/HIF-responsive expression vectors using HREs derived from the human vascular endothelial growth factor (VEGF) and erythropoietin (EPO) genes. We measured the ability of these constructs to express the luciferase and LacZ/beta-galactosidase (beta-gal) reporter genes bidirectionally under normoxic (21% O(2)) versus hypoxic (1, 3, 5, and 10% O(2)) conditions by transient transfection in three human glioma cell lines (LN229, U251MG and U138MG). Nine constructs were identified that exhibited moderate to high inducibility at 1% O(2) while maintaining tight regulation under normoxic conditions. Moreover, the level of activation was a function of O(2) concentration and was exponential at O(2) levels below 5%. These vectors will be valuable tools in a variety of gene therapy applications targeting pathological activation of the HIF-1/HRE pathway.


Subject(s)
Cell Hypoxia/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Genetic Vectors , Glioma/genetics , Nuclear Proteins/genetics , DNA-Binding Proteins/biosynthesis , Endothelial Growth Factors/genetics , Erythropoietin/genetics , Genes, Reporter , Glioma/metabolism , Helix-Loop-Helix Motifs/genetics , Humans , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Lymphokines/genetics , Nuclear Proteins/biosynthesis , Transcription Factors/genetics , Transfection , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
6.
Int J Food Microbiol ; 26(1): 43-76, 1995 Jun.
Article in English | MEDLINE | ID: mdl-7662519

ABSTRACT

The history of the development of selective media for isolation of campylobacters, including the rationale for choice of selective agents is described. Developments have included modifications to allow incubation at 37 degrees C instead of 42 or 43 degrees C and changes in the types and concentrations of antibiotics in order not to inhibit organisms such as Campylobacter upsaliensis, C. jejuni subsp. doylei and some strains of C. coli and C. lari. When examining foods, plating media originally developed for isolation from faeces are normally used, sometimes after liquid enrichment. Most of the media include ingredients intended to protect campylobacters from the toxic effect of oxygen derivatives. Most commonly used are lysed or defibrinated blood; charcoal; a combination of ferrous sulphate, sodium metabisulphite and sodium pyruvate (FBP); and haemin or haematin. To date no medium includes an indicator system--for instance a pH indicator to show whether colonies produce acid or alkali from particular substrates. The manner in which liquid enrichment media are used has been modified for food samples to avoid inhibitory effects on sublethally damaged cells by toxic components in the formula. This is done by a preliminary period of incubation at reduced temperature and sometimes by delayed addition of antibiotics. Expensive and time-consuming methods have been proposed to achieve a microaerobic atmosphere while using liquid enrichment media. To date there is no generally accepted 'standard' method of isolating campylobacters from food.


Subject(s)
Campylobacter/isolation & purification , Culture Media/chemistry , Food Microbiology , Anti-Bacterial Agents , Bacteriological Techniques , Campylobacter/growth & development , Temperature
SELECTION OF CITATIONS
SEARCH DETAIL
...