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










Publication year range
1.
PLoS One ; 9(8): e104015, 2014.
Article in English | MEDLINE | ID: mdl-25119572

ABSTRACT

Vascular endothelial growth factor-A (VEGF) is produced by most cancer cells as multiple isoforms, which display distinct biological activities. VEGF plays an undisputed role in tumour growth, vascularisation and metastasis; nevertheless the functions of individual isoforms in these processes remain poorly understood. We investigated the effects of three main murine isoforms (VEGF188, 164 and 120) on tumour cell behaviour, using a panel of fibrosarcoma cells we developed that express them individually under endogenous promoter control. Fibrosarcomas expressing only VEGF188 (fs188) or wild type controls (fswt) were typically mesenchymal, formed ruffles and displayed strong matrix-binding activity. VEGF164- and VEGF120-producing cells (fs164 and fs120 respectively) were less typically mesenchymal, lacked ruffles but formed abundant cell-cell contacts. On 3D collagen, fs188 cells remained mesenchymal while fs164 and fs120 cells adopted rounded/amoeboid and a mix of rounded and elongated morphologies respectively. Consistent with their mesenchymal characteristics, fs188 cells migrated significantly faster than fs164 or fs120 cells on 2D surfaces while contractility inhibitors accelerated fs164 and fs120 cell migration. VEGF164/VEGF120 expression correlated with faster proliferation rates and lower levels of spontaneous apoptosis than VEGF188 expression. Nevertheless, VEGF188 was associated with constitutively active/phosphorylated AKT, ERK1/2 and Stat3 proteins. Differences in proliferation rates and apoptosis could be explained by defective signalling downstream of pAKT to FOXO and GSK3 in fs188 and fswt cells, which also correlated with p27/p21 cyclin-dependent kinase inhibitor over-expression. All cells expressed tyrosine kinase VEGF receptors, but these were not active/activatable suggesting that inherent differences between the cell lines are governed by endogenous VEGF isoform expression through complex interactions that are independent of tyrosine kinase receptor activation. VEGF isoforms are emerging as potential biomarkers for anti-VEGF therapies. Our results reveal novel roles of individual isoforms associated with cancer growth and metastasis and highlight the importance of understanding their diverse actions.


Subject(s)
Cell Movement/genetics , Cell Proliferation/genetics , Fibrosarcoma/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Apoptosis , Carcinogenesis/genetics , Cell Adhesion , Cell Line, Tumor , Cell Survival/genetics , Fibrosarcoma/pathology , Gene Expression Regulation, Neoplastic , Mice , Neoplasm Metastasis/genetics , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Receptors, Vascular Endothelial Growth Factor/metabolism , Vascular Endothelial Growth Factor A/chemistry
2.
Cancer Res ; 68(7): 2301-11, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18381437

ABSTRACT

Tubulin-binding vascular-disrupting agents (VDA) are currently in clinical trials for cancer therapy but the factors that influence tumor susceptibility to these agents are poorly understood. We evaluated the consequences of modifying tumor vascular morphology and function on vascular and therapeutic response to combretastatin-A4 3-O-phosphate (CA-4-P), which was chosen as a model VDA. Mouse fibrosarcoma cell lines that are capable of expressing all vascular endothelial growth factor (VEGF) isoforms (control) or only single isoforms of VEGF (VEGF120, VEGF164, or VEGF188) were developed under endogenous VEGF promoter control. Once tumors were established, VEGF isoform expression did not affect growth or blood flow rate. However, VEGF188 was uniquely associated with tumor vascular maturity, resistance to hemorrhage, and resistance to CA-4-P. Pericyte staining was much greater in VEGF188 and control tumors than in VEGF120 and VEGF164 tumors. Vascular volume was highest in VEGF120 and control tumors (CD31 staining) but total vascular length was highest in VEGF188 tumors, reflecting very narrow vessels forming complex vascular networks. I.v. administered 40 kDa FITC-dextran leaked slowly from the vasculature of VEGF188 tumors compared with VEGF120 tumors. Intravital microscopy measurements of vascular length and RBC velocity showed that CA-4-P produced significantly more vascular damage in VEGF120 and VEGF164 tumors than in VEGF188 and control tumors. Importantly, this translated into a similar differential in therapeutic response, as determined by tumor growth delay. Results imply differences in signaling pathways between VEGF isoforms and suggest that VEGF isoforms might be useful in vascular-disrupting cancer therapy to predict tumor susceptibility to VDAs.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Fibrosarcoma/blood supply , Fibrosarcoma/drug therapy , Stilbenes/pharmacology , Vascular Endothelial Growth Factor A/biosynthesis , Animals , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Female , Fibrosarcoma/metabolism , Immunohistochemistry , Mice , Mice, SCID , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Protein Isoforms , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Vascular Endothelial Growth Factor A/genetics
3.
Int J Radiat Oncol Biol Phys ; 69(5): 1553-62, 2007 Dec 01.
Article in English | MEDLINE | ID: mdl-17920784

ABSTRACT

PURPOSE: To investigate the effects of radiation on the endothelial cytoskeleton and endothelial monolayer permeability and to evaluate associated signaling pathways, which could reveal potential mechanisms of known vascular effects of radiation. METHODS AND MATERIALS: Cultured endothelial cells were X-ray irradiated, and actin filaments, microtubules, intermediate filaments, and vascular endothelial (VE)-cadherin junctions were examined by immunofluorescence. Permeability was determined by the passage of fluorescent dextran through cell monolayers. Signal transduction pathways were analyzed using RhoA, Rho kinase, and stress-activated protein kinase-p38 (SAPK2/p38) inhibitors by guanosine triphosphate-RhoA activation assay and transfection with RhoAT19N. The levels of junction protein expression and phosphorylation of myosin light chain and SAPK2/p38 were assessed by Western blotting. The radiation effects on cell death were verified by clonogenic assays. RESULTS: Radiation induced rapid and persistent actin stress fiber formation and redistribution of VE-cadherin junctions in microvascular, but not umbilical vein endothelial cells, and microtubules and intermediate filaments remained unaffected. Radiation also caused a rapid and persistent increase in microvascular permeability. RhoA-guanosine triphosphatase and Rho kinase were activated by radiation and caused phosphorylation of downstream myosin light chain and the observed cytoskeletal and permeability changes. SAPK2/p38 was activated by radiation but did not influence either the cytoskeleton or permeability. CONCLUSION: This study is the first to show rapid activation of the RhoA/Rho kinase by radiation in endothelial cells and has demonstrated a link between this pathway and cytoskeletal remodeling and permeability. The results also suggest that the RhoA pathway might be a useful target for modulating the permeability and other effects of radiation for therapeutic gain.


Subject(s)
Cytoskeleton/radiation effects , Endothelial Cells/radiation effects , Actins/radiation effects , Cadherins/radiation effects , Cell Death/radiation effects , Cells, Cultured , Dextrans/pharmacokinetics , Endothelial Cells/ultrastructure , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Endothelium, Vascular/radiation effects , Humans , Intermediate Filaments/radiation effects , Microtubules/radiation effects , Mitogen-Activated Protein Kinase 11/physiology , Permeability , Radiation Tolerance , Signal Transduction/physiology , Signal Transduction/radiation effects , rhoA GTP-Binding Protein/physiology
4.
Int Rev Cytol ; 257: 181-212, 2007.
Article in English | MEDLINE | ID: mdl-17280898

ABSTRACT

Hypoxia is an integral characteristic of the tumor microenvironment, primarily due to the microvascular defects that accompany the accelerated neoplastic growth. The presence of tumor hypoxic areas correlates with negative outcome after radiotherapy, chemotherapy, and surgery, as hypoxia not only provides an environment directly facilitating chemo- and radio-resistance, but also encourages the evolution of phenotypic changes inducing permanent resistance to treatment and metastatic spread. Therefore, successful treatment of hypoxic cells has the potential to not only improve local control but also impact overall patient survival. Specific and selective targeting of hypoxic tumor areas can be achieved at all three steps of a gene therapy treatment: delivery of the therapeutic gene to the tumor, regulation of gene expression, and therapeutic efficacy. In this review the latest developments and innovations in hypoxia-targeted gene therapy are discussed. In particular, approaches such as hypoxia-conditionally replicating viruses, cellular vehicles, and gene therapy means to disrupt the hypoxia-inducible factor (HIF) signaling are outlined.


Subject(s)
Genetic Therapy , Neoplasms/pathology , Neoplasms/therapy , Animals , Cell Hypoxia , Combined Modality Therapy , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/chemistry , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neoplasms/genetics
5.
Virology ; 349(1): 121-33, 2006 May 25.
Article in English | MEDLINE | ID: mdl-16464484

ABSTRACT

To facilitate a more efficient radiation and chemotherapy of mammary tumours, synthetic enhancer elements responsive to hypoxia and ionizing radiation were coupled to the mammary-specific minimal promoter of the murine whey acidic protein (WAP) encoding gene. The modified WAP promoter was introduced into a retroviral promoter conversion (ProCon) vector. Expression of a transduced reporter gene in response to hypoxia and radiation was analysed in stably infected mammary cancer cell lines and an up to 9-fold increase in gene expression demonstrated in comparison to the respective basic vector. Expression analyses in vitro, moreover, demonstrated a widely preserved mammary cell-specific promoter activity. For in vivo analyses, xenograft tumours consisting of infected human mammary adenocarcinoma cells were established in SCID/beige mice. Immunohistochemical analyses demonstrated a hypoxia-specific, markedly increased WAP promoter-driven expression in these tumours. Thus, this retroviral vector will facilitate a targeted gene therapeutic approach exploiting the unique environmental condition in solid tumours.


Subject(s)
Breast Neoplasms/therapy , Cell Hypoxia , Genetic Therapy/methods , Genetic Vectors , Milk Proteins/genetics , Promoter Regions, Genetic , Retroviridae , Animals , Cell Line , Disease Models, Animal , Gene Expression Regulation , Genes, Reporter , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Humans , Immunohistochemistry , Mice , Mice, SCID , Microscopy, Fluorescence , Neoplasm Transplantation , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/radiation effects , Sensitivity and Specificity , Transplantation, Heterologous , X-Rays
6.
Cancer Gene Ther ; 12(7): 655-62, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15818381

ABSTRACT

Combining gene therapy with radiotherapy and chemotherapy holds potential to increase the efficacy of cancer treatment, while minimizing side effects. We tested the responsiveness of synthetic gene promoters containing CArG elements from the Early Growth Response 1 (Egr1) gene after neutron irradiation, doxorubicin and cisplatin. Human MCF-7 breast adenocarcinoma and U373-MG glioblastoma cells were transfected with plasmids containing CArG promoters controlling the expression of the green fluorescent protein (GFP). Exposing the cells to neutrons, doxorubicin or cisplatin resulted in a significant induction of transgene expression. Therapeutic advantage was demonstrated by replacing the reporter with the herpes simplex virus thymidine kinase (HSVtk), able to convert the prodrug ganciclovir (GCV) into a cytotoxin. A 1.3 Gy neutron dose caused 49% growth inhibition in MCF-7 cells, which increased to 63% in irradiated CArG-HSVtk-transfectants treated with GCV. Exposure to 0.5 microM cisplatin or 0.01 microM doxorubicin induced a growth inhibition of 25-30% in MCF-7 cells. In the presence of GCV, this value increased to 65-70% in cells transfected with the CArG promoter constructs driving the expression of HSVtk. These data indicate that combining CArG-mediated HSVtk/GCV suicide gene therapy with radio- and chemotherapy can enhance antitumor toxicity, and validates future in vivo investigations.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/therapy , Breast Neoplasms/therapy , DNA-Binding Proteins/genetics , Gene Expression Regulation/drug effects , Genetic Therapy/methods , Genetic Vectors , Immediate-Early Proteins/genetics , Transcription Factors/genetics , Adenocarcinoma/drug therapy , Adenocarcinoma/radiotherapy , Adenocarcinoma/therapy , Antiviral Agents/therapeutic use , Apoptosis , Brain Neoplasms/drug therapy , Brain Neoplasms/radiotherapy , Breast Neoplasms/drug therapy , Breast Neoplasms/radiotherapy , Cisplatin/pharmacology , Combined Modality Therapy , Doxorubicin/pharmacology , Early Growth Response Protein 1 , Ganciclovir/metabolism , Gene Transfer Techniques , Glioblastoma/drug therapy , Glioblastoma/radiotherapy , Glioblastoma/therapy , Green Fluorescent Proteins/metabolism , Humans , Neutrons , Promoter Regions, Genetic , Radiation-Sensitizing Agents/therapeutic use , Response Elements/genetics , Simplexvirus/enzymology , Simplexvirus/genetics , Thymidine Kinase/genetics , Thymidine Kinase/metabolism , Tumor Cells, Cultured , Zinc Fingers
7.
Int J Radiat Oncol Biol Phys ; 62(1): 213-22, 2005 May 01.
Article in English | MEDLINE | ID: mdl-15850924

ABSTRACT

PURPOSE: Tumor hypoxia is unequivocally linked to poor radiotherapy outcome. This study aimed to identify enhancer sequences that respond maximally to a combination of radiation and hypoxia for use in genetic radiotherapy approaches. METHODS AND MATERIALS: The influence of radiation (5 Gy) and hypoxia (1% O2) on reporter-gene expression driven by hypoxia (HRE) and radiation (Egr-1) responsive elements was evaluated in tumor cells grown as monolayers or multicellular spheroids. Hypoxia-inducible factor-1alpha (HIF-1alpha) and HIF-2alpha protein expression was monitored in parallel. RESULTS: Of the sequences tested, an HRE from the phosphoglycerate kinase-1 gene (PGK-18[5+]) was maximally induced in response to hypoxia plus radiation in all 5 cell lines tested. The additional radiation treatment afforded a significant increase in the induction of PGK-18[5+] compared with hypoxia alone in 3 cell lines. HIF-1alpha/2alpha were induced by radiation but combined hypoxia/radiation treatment did not yield a further increase. The dual responsive nature of HREs was maintained when spheroids were irradiated after delivery of HRE constructs in a replication-deficient adenovirus. CONCLUSIONS: Hypoxia-responsive enhancer element sequences are dually responsive to combined radiation and hypoxic treatment. Their use in genetic radiotherapy in vivo could maximize expression in the most radio-resistant population at the time of radiation and also exploit microenvironmental changes after radiotherapy to yield additional switch-on.


Subject(s)
Cell Hypoxia/genetics , Gene Expression Regulation , Phosphoglycerate Kinase/genetics , Radiation Tolerance/genetics , Basic Helix-Loop-Helix Transcription Factors , Hypoxia-Inducible Factor 1, alpha Subunit , Phosphoglycerate Kinase/metabolism , Promoter Regions, Genetic , Radiation Tolerance/physiology , Spheroids, Cellular , Transcription Factors/genetics , Transcription Factors/metabolism
8.
Am J Pathol ; 165(4): 1401-11, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15466404

ABSTRACT

The tubulin-binding agent combretastatin A-4-phosphate (CA-4-P), rapidly disrupts the vascular network of tumors leading to secondary tumor cell death. In vitro, CA-4-P destabilizes microtubules and causes endothelial cell death. In this study we analyze the mechanisms by which CA-4-P induces the death of proliferating endothelial cells. We demonstrate that at >/=7.5 nmol/L, CA-4-P damages mitotic spindles, arrests cells at metaphase, and leads to the death of mitotic cells with characteristic G(2)/M DNA content. Mitotic arrest was associated with elevated levels of cyclin B1 protein and p34(cdc2) activity. Inhibition of p34(cdc2) activity by purvalanol A caused mitotic-arrested cells to rapidly exit mitosis, suggesting that sustained p34(cdc2) activity was responsible for metaphase arrest. Pharmacological prevention of entry into mitosis protected cells from undergoing cell death, further establishing the link between mitosis and cell death induction by CA-4-P. CA-4-P-mediated cell death shared characteristics of apoptosis but was independent of caspase activation suggesting the involvement of a non-caspase pathway(s). These data suggest that induction of apoptosis in endothelial cells by CA-4-P is associated with prolonged mitotic arrest. Therefore, by activating cell death pathways, CA-4-P, in addition to being an effective anti-vascular agent, may also interfere with regrowth of blood vessels in the tumor.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/pathology , Mitosis/drug effects , Stilbenes/pharmacology , Apoptosis/physiology , Blotting, Western , CDC2 Protein Kinase/drug effects , CDC2 Protein Kinase/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Fluorescent Antibody Technique , Humans
9.
Cancer Metastasis Rev ; 23(3-4): 269-76, 2004.
Article in English | MEDLINE | ID: mdl-15197328

ABSTRACT

Radiotherapy remains one of the primary treatment modalities for most malignancies. Biologically based improvements in the scheduling of conventional radiotherapy and treatment planning, innovations like conformal radiotherapy and intensity-modulated radiation therapy have considerably improved the targeting and effectiveness of radiation for treatment of solid tumors. These new radiotherapy technologies are also promising means of focusing the activation of anti-tumor gene therapy systems, as an approach to further improve radiotherapeutic treatment, particularly for tumors refractive to current therapies. Gene therapy vectors that express therapeutic genes following irradiation have been produced. Delivery of such vectors to the tumor allows temporal and spatial expression of the transgenes within the radiation field. Hypoxia is a physiological characteristic of solid tumors and an independent prognostic marker for poor radiation treatment outcome. Nevertheless, hypoxia has been exploited to drive therapeutic gene expression from gene therapy vectors delivered to solid tumors exhibiting significant areas of low oxygen tension. Radiation and hypoxia inducible gene therapy systems rely on the activation of gene promoters containing specific responsive elements. Recent studies have shown the potential to combine these elements, permitting either or both stimuli to drive therapeutic gene expression. Furthermore, transgene expression can be amplified and sustained using novel 'signal feedback' or recombination systems. Such innovations allow promising new strategies to improve radiation treatment outcome, particularly where tumor hypoxia is a predominant issue.


Subject(s)
Cell Hypoxia/genetics , Genetic Therapy/methods , Neoplasms/genetics , Neoplasms/radiotherapy , Animals , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/metabolism , Neoplasms/therapy , Oxygen/metabolism
10.
Cancer Gene Ther ; 11(7): 508-13, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15143345

ABSTRACT

Horseradish peroxidase has previously been shown to catalyze the conversion of indole-3-acetic acid (IAA) to a potent cytotoxin in a gene therapy setting. A three-dimensional spheroid model composed of a human head and neck carcinoma cell line, has been used to mimic the tumor microenvironment, such as regions of hypoxia. Exposure of intact spheroids to 0.05-5 mM concentrations of IAA and the halogenated indole, 5-bromoindole-3-acetic acid (5Br-IAA), resulted in decreased cell survival, and demonstrates that this combination is effective under tumor-simulated conditions. In addition, 5Br-IAA, displayed selectivity for spheroids with a large hypoxic fraction following short exposure times.


Subject(s)
Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Horseradish Peroxidase/metabolism , Indoleacetic Acids/metabolism , Indoleacetic Acids/pharmacology , Models, Biological , Cell Culture Techniques , Cell Hypoxia/physiology , Cell Survival/drug effects , Head and Neck Neoplasms/drug therapy , Horseradish Peroxidase/genetics , Humans , Indoleacetic Acids/therapeutic use , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Time Factors , Transfection , Tumor Cells, Cultured , Tumor Stem Cell Assay
12.
Curr Pharm Des ; 9(26): 2131-54, 2003.
Article in English | MEDLINE | ID: mdl-14529410

ABSTRACT

Improvements in the radiotherapeutic management of solid tumors through the concurrent use of gene therapy is a realistic possibility. Of the broad array of candidate genes that have been evaluated, those encoding prodrug-activating enzymes are particularly appealing since they directly complement ongoing clinical chemoradiation regimes. Gene-Directed Enzyme-Prodrug Therapy (GDEPT) only requires a fraction of the target cells to be genetically modified, providing that the resultant cytotoxic prodrug metabolites redistribute efficiently (the bystander effect). This transfer of cytotoxicity to neighboring non-targeted cancer cells is central to the success of any gene therapy strategy, irrespective of the therapeutic gene employed. In the context of genetic chemoradiotherapy, efficient prodrug metabolite diffusion will be a prerequisite for efficient radiosensitization. Some, but not all GDEPT approaches have been analysed in combination with radiotherapy. Examples of prodrugs of clinically established chemotherapeutic agents currently used in conjunction with radiotherapy include: 5-fluorocytosine (5FC), cyclophosphamide (CPA), irinotecan (CPT-11), gemcitabine (dFdC), capecitabine, mitomycin C (MMC) and AQ4N. Other GDEPT paradigms, such as ganciclovir (GCV) and Herpes Simplex thymidine kinase (HSV-tk), dinitrobenzamide (DNB) mustard or aziridinyl analogs and the E. coli nitroreductase (NTR), CMDA or ZP2767P with Pseudomonas aeruginosa carboxypeptidase G2 (CPG2), and indole-3-acetic acid (IAA) activated by horseradish peroxidase (HRP) have no clinically established chemotherapeutic counterpart. Each prodrug is discussed in this review in the context of GDEPT, with a particular attention to translational research and clinical utility in combination with radiotherapy.


Subject(s)
Genetic Therapy/methods , Neoplasms/therapy , Prodrugs/therapeutic use , Animals , Combined Modality Therapy , Enzymes/genetics , Enzymes/metabolism , Humans , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/radiotherapy , Prodrugs/chemistry , Radiation-Sensitizing Agents , Radiotherapy, Adjuvant
13.
J Cell Physiol ; 197(3): 312-25, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14566961

ABSTRACT

Tumor hypoxia has long been recognized as a critical issue in oncology. Resistance of hypoxic areas has been shown to affect treatment outcome after radiation, chemotherapy, and surgery in a number of tumor sites. Two main strategies to overcome tumor hypoxia are to increase the delivery of oxygen (or oxygen-mimetic drugs), and exploiting this unique environmental condition of solid tumors for targeted therapy. The first strategy includes hyperbaric oxygen breathing, the administration of carbogen and nicotinamide, and the delivery of chemical radiosensitizers. In contrast, bioreductive drugs and hypoxia-targeted suicide gene therapy aim at activating cytotoxic agents at the tumor site, while sparing normal tissue from damage. The cellular machinery responds to hypoxia by activating the expression of genes involved in angiogenesis, anaerobic metabolism, vascular permeability, and inflammation. In most cases, transcription is initiated by the binding of the transcription factor hypoxia-inducible factor (HIF) to hypoxia responsive elements (HREs). Hypoxia-targeting for gene therapy has been achieved by utilizing promoters containing HREs, to induce selective and efficient transgene activation at the tumor site. Hypoxia-targeted delivery and prodrug activation may add additional levels of selectivity to the treatment. In this article, the latest developments of cancer gene therapy of the hypoxic environment are discussed, with particular attention to combined protocols with ionizing radiation. Ultimately, it is proposed that by adopting specific transgene activation and molecular amplification systems, resistant hypoxic tumor tissues may be effectively targeted with gene therapy.


Subject(s)
Genetic Therapy/methods , Hypoxia/metabolism , Neoplasms/metabolism , Neoplasms/therapy , Transcription Factors , Animals , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/radiation effects , Genetic Therapy/trends , Humans , Hypoxia/drug therapy , Hypoxia/genetics , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Neoplasms/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Radiotherapy, Adjuvant/methods , Radiotherapy, Adjuvant/trends , Transcriptional Activation/drug effects , Transcriptional Activation/genetics , Transcriptional Activation/radiation effects
14.
J Cell Biochem ; 87(2): 221-32, 2002.
Article in English | MEDLINE | ID: mdl-12244574

ABSTRACT

We have previously proposed the horseradish peroxidase (HRP) and the non-toxic plant hormone indole-3-acetic acid (IAA) as a novel system for gene-directed enzyme/prodrug therapy (GDEPT). The cytotoxic potential of HRP/IAA GDEPT and the induction of a bystander effect were demonstrated in vitro under normoxic as well as hypoxic tumour conditions. To date, the chemical agents and the cellular targets involved in HRP/IAA-mediated toxicity have not been identified. In the present work, some of the molecular and morphological features of the cells treated with HRP/IAA gene therapy were analysed. Human T24 bladder carcinoma cells transiently transfected with the HRP cDNA and exposed to the prodrug IAA showed chromatin condensation, formation of apoptotic bodies, DNA fragmentation, and Annexin V binding. Similar effects were observed when the cells were incubated with the apoptotic agent cisplatin. Caspases appeared to be involved as effectors in HRP/IAA-mediated apoptosis, since treatment with a general caspase inhibitor decreased the fraction of cells with micronuclei (MN) by 30%, with fragmented DNA by 50%, and with condensed chromatin by 60%. However, very little degradation of one of the downstream targets of caspase-3, PARP, could be detected, and apoptosis alone did not appear to account for the killing levels measured with a clonogenic assay. The effect of HRP/IAA treatment on cell cycle progression was also investigated, and a rapid cytostatic effect, equally affecting all phases of the division cycle, was observed.


Subject(s)
Genetic Therapy/adverse effects , Horseradish Peroxidase/genetics , Indoleacetic Acids/pharmacology , Prodrugs/pharmacology , Annexin A5/metabolism , Cell Nucleus/drug effects , Cell Nucleus/ultrastructure , Chromatin/metabolism , Combined Modality Therapy , DNA Fragmentation/drug effects , DNA, Complementary/metabolism , G1 Phase/drug effects , Genetic Therapy/methods , Horseradish Peroxidase/metabolism , Humans , Indoleacetic Acids/pharmacokinetics , Poly(ADP-ribose) Polymerases/metabolism , Prodrugs/pharmacokinetics , Time Factors , Transfection , Tumor Cells, Cultured/metabolism , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/therapy
15.
Front Biosci ; 7: d1516-24, 2002 Jun 01.
Article in English | MEDLINE | ID: mdl-12045003

ABSTRACT

Gene therapy for cancer treatment represents a promising approach that has shown selectivity and efficacy in experimental systems as well as clinical trials. Some major problems remain to be solved before this strategy becomes routinely adopted in the clinic, one of the main challenges being the improvement of gene delivery. Namely, the development of DNA vectors characterized by maximum efficiency and minimal toxicity will define the success of gene therapy and its chances of being accepted by public and clinicians. A number of issues need to be considered. The "magic" vector should be targeted, protected from degradation and immune attack, and safe for the recipient and the environment. Moreover, it should express the therapeutic gene for as long as required, in an appropriately regulated fashion. Vehicles such as retroviruses, adenoviruses and liposomes have been adopted in clinical studies, with varying results. New therapeutic modalities are also being explored in order to overcome the limitation of poor gene transfer and patient toxicity, including bacteria, adeno-associated and herpes simplex viruses, lentiviruses, cationic polymer-DNA complexes and electroporation. Some of the delivery systems tested in preclinical and clinical models are reviewed in this article, with particular attention to the targeting of the tumor environment.


Subject(s)
Gene Transfer Techniques , Genetic Therapy/methods , Neoplasms/therapy , Animals , Gene Transfer Techniques/adverse effects , Genetic Therapy/adverse effects , Genetic Vectors/adverse effects , Genetic Vectors/therapeutic use , Humans
16.
Biochem Pharmacol ; 63(2): 265-72, 2002 Jan 15.
Article in English | MEDLINE | ID: mdl-11841802

ABSTRACT

Indole-3-acetic acid and some derivatives are oxidized by horseradish peroxidase, forming a radical-cation that rapidly fragments (eliminating CO(2)) to form cytotoxic products. No toxicity is seen when either indole-3-acetic acid or horseradish peroxidase is incubated alone at concentrations that together form potent cytotoxins. Unexpectedly, 5-fluoroindole-3-acetic acid, which is oxidized by horseradish peroxidase compound I 10-fold more slowly than indole-3-acetic acid, is much more cytotoxic towards V79 hamster fibroblasts in the presence of peroxidase than the unsubstituted indole. The fluorinated prodrug/peroxidase combination also shows potent cytotoxic activity in human and rodent tumor cell lines. Cytotoxicity is thought to arise in part from the formation of 3-methylene-2-oxindole (or analogues) that can conjugate with thiols and probably DNA or other biological nucleophiles. Levels of the fluorinated prodrug in the murine carcinoma NT after intraperitoneal administration of 50 mg/kg were about 200 microM. Although these were 4-5-fold lower than plasma levels (which reached 1mM), the integrated area under the concentration/time curve in tumors over 2 hr was approximately 20 mM min, almost double the exposure needed to achieve approximately 90-99% cell kill in human MCF7 breast or HT29 colon tumor cell lines and CaNT murine cells in vitro, although the human bladder T24 carcinoma cell line was more resistant. The high cytotoxicity of 5-fluoroindole-3-acetic acid after oxidative activation suggests its further evaluation as a prodrug for targeted cancer therapy involving antibody-, polymer-, or gene-directed delivery of horseradish peroxidase or similar activating enzymes.


Subject(s)
Antineoplastic Agents/pharmacology , Indoleacetic Acids/pharmacology , Peroxidases/metabolism , Prodrugs/pharmacology , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacokinetics , Cell Survival/drug effects , Disease Models, Animal , Drug Delivery Systems , Female , Fluorine/metabolism , Glutathione/metabolism , HT29 Cells , Humans , Indoleacetic Acids/metabolism , Indoleacetic Acids/pharmacokinetics , Mice , Mice, Inbred CBA , Neoplasms, Experimental/metabolism , Oxidation-Reduction , Prodrugs/metabolism , Prodrugs/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...