Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 118
Filter
1.
Cancer Gene Ther ; 23(9): 303-14, 2016 09.
Article in English | MEDLINE | ID: mdl-27492853

ABSTRACT

The combination of radiation with radiosensitizing gene delivery or oncolytic viruses promises to provide an advantage that could improve the therapeutic results for glioblastoma. X-rays can induce significant molecular changes in cancer cells. We isolated the GIRLRG peptide that binds to radiation-inducible 78 kDa glucose-regulated protein (GRP78), which is overexpressed on the plasma membranes of irradiated cancer cells and tumor-associated microvascular endothelial cells. The goal of our study was to improve tumor-specific adenovirus-mediated gene delivery by selectively targeting the adenovirus binding to this radiation-inducible protein. We employed an adenoviral fiber replacement approach to conduct a study of the targeting utility of GRP78-binding peptide. We have developed fiber-modified adenoviruses encoding the GRP78-binding peptide inserted into the fiber-fibritin. We have evaluated the reporter gene expression of fiber-modified adenoviruses in vitro using a panel of glioma cells and a human D54MG tumor xenograft model. The obtained results demonstrated that employment of the GRP78-binding peptide resulted in increased gene expression in irradiated tumors following infection with fiber-modified adenoviruses, compared with untreated tumor cells. These studies demonstrate the feasibility of adenoviral retargeting using the GRP78-binding peptide that selectively recognizes tumor cells responding to radiation treatment.


Subject(s)
Adenoviridae/genetics , Gene Transfer Techniques , Genetic Vectors/genetics , Radiation, Ionizing , Adenoviridae/metabolism , Animals , Antigens, Neoplasm/immunology , Capsid Proteins/genetics , Capsid Proteins/metabolism , Cell Line, Tumor , Disease Models, Animal , Endoplasmic Reticulum Chaperone BiP , Female , Gene Expression , Genes, Reporter , Genetic Therapy , Genetic Vectors/administration & dosage , Glioma/genetics , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Integrin alphaVbeta3/metabolism , Mice , Oncolytic Virotherapy , Oncolytic Viruses/genetics , Receptors, Vitronectin/metabolism , Transduction, Genetic , Transgenes , X-Rays , Xenograft Model Antitumor Assays
2.
Oncogene ; 33(12): 1558-69, 2014 Mar 20.
Article in English | MEDLINE | ID: mdl-23563176

ABSTRACT

PDZ domains represent one group of the major structural units that mediate protein interactions in intercellular contact, signal transduction and assembly of biological machineries. Tax-interacting protein (TIP)-1 protein is composed of a single PDZ domain that distinguishes TIP-1 from other PDZ domain proteins that more often contain multiple protein domains and function as scaffolds for protein complex assembly. However, the biological functions of TIP-1, especially in cell transformation and tumor progression, are still controversial as observed in a variety of cell types. In this study, we have identified ARHGEF7, a guanine nucleotide exchange factor for Rho GTPases, as one novel TIP-1-interacting protein in human glioblastoma cells. We found that the presence of TIP-1 protein is essential to the intracellular redistribution of ARHGEF7 and rhotekin, one Rho effector and the spatiotemporally coordinated activation of Rho GTPases (RhoA, Cdc42 and Rac1) in migrating glioblastoma cells. TIP-1 knockdown resulted in both aberrant localization of ARHGEF7 and rhotekin, as well as abnormal activation of Rho GTPases that was accompanied with impaired motility of glioblastoma cells. Furthermore, TIP-1 knockdown suppressed tumor cell dispersal in orthotopic glioblastoma murine models. We also observed high levels of TIP-1 expression in human glioblastoma specimens, and the elevated TIP-1 levels are associated with advanced staging and poor prognosis in glioma patients. Although more studies are needed to further dissect the mechanism(s) by which TIP-1 modulates the intracellular redistribution and activation of Rho GTPases, this study suggests that TIP-1 holds potential as both a prognostic biomarker and a therapeutic target of malignant gliomas.


Subject(s)
Glioblastoma/pathology , Intracellular Signaling Peptides and Proteins/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Apoptosis Regulatory Proteins , Brain Neoplasms/diagnosis , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Transformation, Neoplastic , Enzyme Activation , Female , GTP-Binding Proteins , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Glioblastoma/diagnosis , Glioblastoma/drug therapy , Glioblastoma/metabolism , Humans , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Male , Mice , Molecular Targeted Therapy , Neoplasm Invasiveness , Neoplasm Staging , Prognosis , Protein Transport , Rho Guanine Nucleotide Exchange Factors/metabolism , Spatio-Temporal Analysis , beta Catenin/metabolism
3.
Cell Death Differ ; 19(3): 387-96, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21738215

ABSTRACT

Exposure of the brain to ionizing radiation can cause neurocognitive deficiencies. The pathophysiology of these neurological changes is complex and includes radiation-induced apoptosis in the subgranular zone of the hippocampus. We have recently found that inhibition of glycogen synthase kinase 3ß (GSK-3ß) resulted in significant protection from radiation-induced apoptosis in hippocampal neurons. The molecular mechanisms of this cytoprotection include abrogation of radiation-induced accumulation of p53. Here we show that pretreatment of irradiated HT-22 hippocampal-derived neurons with small molecule inhibitors of GSK-3ß SB216763 or SB415286, or with GSK-3ß-specific shRNA resulted in accumulation of the p53-specific E3 ubiquitin ligase MDM2. Knockdown of MDM2 using specific shRNA or chemical inhibition of MDM2-p53 interaction prevented the protective changes triggered by GSK-3ß inhibition in irradiated HT-22 neurons and restored radiation cytotoxicity. We found that this could be due to regulation of apoptosis by subcellular localization and interaction of GSK-3ß, p53 and MDM2. These data suggest that the mechanisms of radioprotection by GSK-3ß inhibitors in hippocampal neurons involve regulation of MDM2-dependent p53 accumulation and interactions between GSK-3ß, MDM2 and p53.


Subject(s)
Apoptosis , Enzyme Inhibitors/pharmacology , Gamma Rays/adverse effects , Glycogen Synthase Kinase 3/antagonists & inhibitors , Hippocampus/metabolism , Neurons/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Line, Transformed , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Mice , Protein Transport/drug effects , Protein Transport/radiation effects , Radiation Tolerance/drug effects , Radiation Tolerance/radiation effects
4.
Cell Death Differ ; 15(10): 1641-53, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18566601

ABSTRACT

Radiosensitivity of various normal tissues is largely dependent on radiation-triggered signal transduction pathways. Radiation simultaneously initiates distinct signaling from both DNA damage and cell membrane. Specifically, DNA strand breaks initiate cell-cycle delay, strand-break repair or programmed cell death, whereas membrane-derived signaling through phosphatidylinositol 3-kinase/Akt and mitogen-activated protein kinase/extracellular signal-regulated kinase (ERK) enhances cell viability. Here, activation of cytosolic phospholipase A(2) (cPLA(2)) and production of the lipid second-messenger lysophosphatidylcholine were identified as initial events (within 2 min) required for radiation-induced activation of Akt and ERK1/2 in vascular endothelial cells. Inhibition of cPLA(2) significantly enhanced radiation-induced cytotoxicity due to an increased number of multinucleated giant cells and cell cycle-independent accumulation of cyclin B1 within 24-48 h of irradiation. Delayed programmed cell death was detected at 72-96 h after treatment. Endothelial functions were also affected by inhibition of cPLA(2) during irradiation resulting in attenuated cell migration and tubule formation. The role of cPLA(2) in the regulation of radiation-induced activation of Akt and ERK1/2 and cell viability was confirmed using human umbilical vein endothelial cells transfected with shRNA for cPLA(2)alpha and cultured embryonic fibroblasts from cPLA(2)alpha(-/-) mice. In summary, an immediate radiation-induced cPLA(2)-dependent signaling was identified that regulates cell viability and, therefore, represents one of the key regulators of radioresistance of vascular endothelial cells.


Subject(s)
Cell Survival/physiology , Cytosol/enzymology , Endothelial Cells/physiology , Endothelial Cells/radiation effects , Group IV Phospholipases A2/metabolism , Signal Transduction/radiation effects , Animals , Cells, Cultured , Endothelial Cells/cytology , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Group IV Phospholipases A2/genetics , Humans , Lysophosphatidylcholines/metabolism , Mice , Mice, Knockout , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction/physiology , Vascular Endothelial Growth Factor Receptor-2/metabolism
5.
Ann Biomed Eng ; 36(5): 821-30, 2008 May.
Article in English | MEDLINE | ID: mdl-18273706

ABSTRACT

PURPOSE: P-selectin expression is significantly increased in tumor microvasculature following exposure to ionizing radiation. The purpose of this study was to image radiation-induced P-selectin expression in vivo using optical imaging and gamma camera imaging in a heterotopic lung cancer model by using ScFv antibodies to P-selectin. PROCEDURES: In vitro studies using endothelial cells were done using 3 Gy radiation and selected ScFv antibodies to P-selectin. In vivo studies were performed using Lewis lung carcinoma cells subcutaneously injected into the hind limbs of nude mice. Mice were treated with 6 Gy radiation and sham radiation 10 days post-inoculation. P-selectin expression was assessed with near-infrared imaging using Cy7 labeled antibody, and gamma camera imaging using( 111)In-DTPA labeled antibody. RESULTS: In vitro studies showed antibody binding to P-selectin in radiation treated endothelial cells. In vivo optical imaging and gamma camera imaging studies showed significant tumor-specific binding to P-selectin in irradiated tumors compared to unirradiated tumors. CONCLUSIONS: Optical imaging and gamma camera imaging are effective methods for visualizing in vivo targeting of radiation-induced P-selectin in lung tumors. This study suggests that fluorescent-labeled and radiolabeled ScFv antibodies can be used to target radiation-induced P-selectin for the tumor-specific delivery of therapeutic drugs and radionuclides in vivo.


Subject(s)
Antibodies, Neoplasm/metabolism , Lung Neoplasms , P-Selectin/metabolism , Radiotherapy/methods , Animals , Cell Line, Tumor , Dose-Response Relationship, Radiation , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/metabolism , Lung Neoplasms/radiotherapy , Male , Mice , Mice, Nude , Radiation Dosage , Radionuclide Imaging
6.
J R Soc Interface ; 3(8): 441-51, 2006 Jun 22.
Article in English | MEDLINE | ID: mdl-16849272

ABSTRACT

The ability of certain reptiles to adhere to vertical (and hang from horizontal) surfaces has been attributed to the presence of specialized adhesive setae on their feet. Structural and compositional studies of such adhesive setae will contribute significantly towards the design of biomimetic fibrillar adhesive materials. The results of electron microscopy analyses of the structure of such setae are presented, indicating their formation from aggregates of proteinaceous fibrils held together by a matrix and potentially surrounded by a limiting proteinaceous sheath. Microbeam X-ray diffraction analysis has shown conclusively that the only ordered protein constituent in these structures exhibits a diffraction pattern characteristic of beta-keratin. Raman microscopy of individual setae, however, clearly shows the presence of additional protein constituents, some of which may be identified as alpha-keratins. Electrophoretic analysis of solubilized setal proteins supports these conclusions, indicating the presence of a group of low-molecular-weight beta-keratins (14-20 kDa), together with alpha-keratins, and this interpretation is supported by immunological analyses.


Subject(s)
Lizards/metabolism , Reptilian Proteins/ultrastructure , Adhesiveness , Animals , Blotting, Western , Extremities/anatomy & histology , Keratins/metabolism , Keratins/ultrastructure , Lizards/anatomy & histology , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Reptilian Proteins/classification , Reptilian Proteins/metabolism , Spectrum Analysis, Raman , X-Ray Diffraction , beta-Keratins/metabolism , beta-Keratins/ultrastructure
7.
Technol Cancer Res Treat ; 3(5): 479-90, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15453813

ABSTRACT

Gene therapy involves identifying a gene of interest and then manipulating the expression of this gene through a variety of techniques. Here we specifically address gene therapy's role in cancer research. This paper will encompass thoroughly investigated techniques such as cancer vaccines and suicide gene therapy and the latest advancements in and applications of these techniques. It will also cover newer techniques such as Antisense Oligonucleotides and small interfering RNAs and how these technologies are being developed and used. The use of gene therapy continues to expand in cancer research and has an integral role in the advancement of cancer treatment.


Subject(s)
Genetic Therapy/methods , Neoplasms/genetics , Neoplasms/therapy , Research Design , Gene Expression Regulation, Neoplastic/radiation effects , Genes, Tumor Suppressor , Humans
8.
Br J Cancer ; 91(3): 537-40, 2004 Aug 02.
Article in English | MEDLINE | ID: mdl-15266313

ABSTRACT

Survivin inhibits apoptosis and promotes mitosis. We determined whether nuclear or cytoplasmic localisation of survivin predicts survival of 48 patients with resected non-small-cell lung cancer (NSCLC). Patients with nuclear staining of survivin had significantly worse survival (relative risk: 3.9, P=0.02). Therefore, survivin may be a biomarker for NSCLC.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma, Non-Small-Cell Lung/pathology , Cell Nucleus/chemistry , Cysteine Proteinase Inhibitors/analysis , Lung Neoplasms/pathology , Microtubule-Associated Proteins/analysis , Aged , Apoptosis , Cytoplasm/chemistry , Female , Humans , Immunohistochemistry , Inhibitor of Apoptosis Proteins , Male , Middle Aged , Neoplasm Proteins , Prognosis , Retrospective Studies , Survival Analysis , Survivin
9.
Am J Clin Oncol ; 24(5): 473-80, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11586099

ABSTRACT

Clinical trials of radiotherapy to control drug delivery were initiated in 1999 at Vanderbilt University. The initial studies exploited the findings that platelets are activated in tumor blood vessels after high-dose irradiation as used in radiosurgery and high-dose-rate brachytherapy. Platelets labeled with 111In showed binding in tumor blood vessels. However, the platelet labeling process caused platelets to also accumulate in the spleen. That clinical trial was closed, and subsequent clinical trials targeted protein activation in irradiated tumor blood vessels. Preclinical studies showed that peptide libraries that bind within irradiated tumor blood vessels contained the peptide sequence Arg-Gln-Asp (RGD). RGD binds to integrin receptors (e.g., receptors for fibrinogen, fibronectin, and vitronectin). We found that the fibrinogen receptor (GPIIb/IIIa, alpha2bbeta3) is activated within irradiated tumor blood vessels. RGD peptidemimetics currently in clinical trials include GPIIb/IIIa antagonists and the platelet-imaging agent biapcitide. Biapcitide is an RGD mimetic that is labeled with 99Tc to allow gamma camera imaging of the biodistribution of the GPIIb/IIIa receptor in neoplasms of patients treated with radiosurgery. This study has shown that the schedule of administration of the RGD mimetic is crucial. The peptide mimetic must be administered immediately before irradiation, whereas the natural ligands to the receptor compete for biapcitide binding if biapcitide is administered after irradiation. The authors currently are conducting a dose deescalation study to determine the threshold dosage required for RGD mimetic binding to radiation activated receptor. Radiation-guided clinical trials have been initiated by use of high-dose-rate brachytherapy. In a separate trial, the pharmacokinetics of radiation-inducible gene therapy are being investigated. In this trial, the radiation-activated promoter Egr-1 regulates expression of the tumor necrosis factor alpha gene, which is administered by use of the attenuated adenovirus vector. The Ad.Egr-TNF (ADGV) gene is administered by intratumoral injection of vector followed by irradiation in patients with soft-tissue sarcomas. This review highlights recent findings in these phase I pharmacokinetic studies of radiation-controlled drug delivery systems.


Subject(s)
Antineoplastic Agents/administration & dosage , Bacterial Proteins , Drug Delivery Systems , Immunoconjugates/administration & dosage , Radiation, Ionizing , Radiation-Sensitizing Agents/administration & dosage , Animals , Antineoplastic Agents/pharmacokinetics , Clinical Trials, Phase I as Topic , Drug Evaluation, Preclinical , Electromagnetic Phenomena , Fibrinogen , Genetic Therapy/methods , Humans , Immunoconjugates/pharmacokinetics , Ligands , Liposomes , Microspheres , Neoplasms/drug therapy , Neoplasms/pathology , Neoplasms/radiotherapy , Oligopeptides , Peptide Library , Radiation-Sensitizing Agents/pharmacokinetics , Radioimmunotherapy , Receptors, Immunologic , Receptors, Peptide , Repressor Proteins
10.
J Control Release ; 74(1-3): 183-91, 2001 Jul 06.
Article in English | MEDLINE | ID: mdl-11489494

ABSTRACT

Radiation can be used to guide drugs to specific sites such as neoplasms or aberrant blood vessels. When blood vessels are treated with ionizing radiation, they respond by expressing a number of cell adhesion molecules and receptors that participate in homeostasis. Examples of radiation-induced molecules in blood vessels include ICAM-1, E-selectin, P-selectin and the beta(3) integrin. We have observed that the endothelium and blood components respond to oxidative stress in a similar, if not identical manner in all tumor models. Although we have identified several other radiation-induced molecules within tumor blood vessels, the beta(3) target for drug delivery achieves the greatest site-specific peptide binding within irradiated tumor blood vessels. We have focused on peptides and antibodies that bind to integrin beta(3). beta(3)-binding proteins have been conjugated to fluorochromes and radionuclides to study the site specificity and microscopic distribution. We have found immunofluorescent and immunohistochemical staining of beta(3) within the lumen of blood vessels immediately following irradiation. To determine whether it is feasible to guide drug delivery to irradiated tumors, we studied ligands to alpha(2b)beta(3) (fibrinogen). Peptides within fibrinogen that bind to alpha(2b)beta(3) includes the dodecapeptide, HHLGGAKQAGDV and the RGD peptide. We utilized 131I conjugation to these ligands to study the biodistribution in tumor bearing mice. Our clinical trial consists of the RGD peptidomimetic, biapcitide, labeled with 99mTc. This study shows that it is feasible to guide drugs to human neoplasms by use of radiation-guided peptides. These studies have shown that peptides that bind to these integrins bind to tumors following exposure to ionizing radiation.


Subject(s)
Antigens, Neoplasm/immunology , Drug Delivery Systems , Neoplasms/immunology , Neoplasms/radiotherapy , Radioimmunotherapy/methods , Amino Acid Sequence , Humans , Image Processing, Computer-Assisted , Luminescent Measurements , Microcirculation , Molecular Sequence Data , Neoplasms/blood supply , Platelet Glycoprotein GPIIb-IIIa Complex/drug effects , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Regional Blood Flow/physiology , Technetium
11.
Free Radic Biol Med ; 30(12): 1426-39, 2001 Jun 15.
Article in English | MEDLINE | ID: mdl-11390188

ABSTRACT

The effect of nonprotein thiol (NPT) free radical scavengers WR-1065 (SH) and WR-33278 (SS), the active thiol and disulfide metabolites of amifostine, N-acetylcysteine (NAC; both L- and D- isomers), mesna, captopril, and dithiothreitol (DTT) on NFkappaB activation in human microvascular endothelial cells (HMEC) was investigated and contrasted to TNFalpha. The use of each of these NPTs at millimolar concentrations independent of oxidative damage-inducing agents resulted in a marked activation of NFkappaB, with the maximum effect observed between 30 min and 1 h after treatment. Only the SH and SS forms of amifostine, however, were effective in activating NFkappaB when administered at micromolar levels. Using a supershift assay, SH and SS equally affected the p50-p65 heterodimer, but not homodimers or heterodimers containing p52 or c-Rel subunits of NFkappaB. Neither catalase nor pyruvate when added to the culture medium to minimize hydrogen peroxide production had an effect on NFkappaB activation by SH. Thus, while oxidative damage is known to activate NFkappaB, the intracellular redox environment may also be affected by the addition of free radical scavenging agents such as NPT, and these in turn are capable of activating the redox sensitive transcription factor NFkappaB. There does not appear to be a significant role, if any, for the production of H(2)O(2) as an intermediate step in the activation of NFkappaB by either the SH or the SS form of amifostine. Rather, the underlying mechanism of action, especially for the SS form, may be related to the close structural and functional similarities of these agents to polyamines, which have been reported to be capable of activating NFkappaB. In contrast to TNFalpha, exposure of cells to either 40 microM or 4 mM of SH for 30 min did not induce intercellular adhesion molecule-1 (ICAM-1) gene expression, but did increase manganese superoxide dismutase (MnSOD) gene expression. MnSOD expression rose by 2-fold and remained elevated from 4 to 22 h following SH exposure.


Subject(s)
Acetylcysteine/analogs & derivatives , Endothelium, Vascular/drug effects , Free Radical Scavengers/pharmacology , Gene Expression Regulation/drug effects , NF-kappa B/biosynthesis , Superoxide Dismutase/biosynthesis , Acetylcysteine/pharmacology , Blotting, Northern , Catalase/pharmacology , Cell Line, Transformed/drug effects , Dimerization , Endothelium, Vascular/metabolism , Enzyme Induction/drug effects , Humans , Hydrogen Peroxide/metabolism , Intercellular Adhesion Molecule-1/biosynthesis , Intercellular Adhesion Molecule-1/genetics , Mercaptoethylamines/pharmacology , NF-kappa B/antagonists & inhibitors , NF-kappa B/chemistry , NF-kappa B/genetics , Oxidation-Reduction , Oxidative Stress , Phosphorylation , Prodrugs/metabolism , Pyruvates/pharmacology , RNA, Messenger/biosynthesis , Radiation-Protective Agents/pharmacology , Skin/blood supply , Superoxide Dismutase/genetics , Tumor Necrosis Factor-alpha/pharmacology
12.
Cancer Res ; 61(6): 2413-9, 2001 Mar 15.
Article in English | MEDLINE | ID: mdl-11289107

ABSTRACT

Certain refractory neoplasms, such as glioblastoma multiforme (GBM) and melanoma, demonstrate a resistant tumor phenotype in vivo. We observed that these refractory tumor models (GBM and melanoma) contain blood vessels that are relatively resistant to radiotherapy. To determine whether the vascular endothelial growth factor receptor-2 (Flk-1/KDR) may be a therapeutic target to improve the effects of radiotherapy, we used the soluble extracellular component of Flk-1 (ExFlk), which blocks vascular endothelial growth factor binding to Flk-1 receptor expressed on the tumor endothelium. Both sFlk-1 and the Flk-1-specifc inhibitor SU5416 eliminated the resistance phenotype in GBM and melanoma microvasculature as determined by both the vascular window and Doppler blood flow methods. Human microendothelial cells and human umbilical vein endothelial cells showed minimal radiation-induced apoptosis. The Flk-1 antagonists sFlk-1 and SU5416 reverted these cell models to apoptosis-prone phenotype. Flk-1 antagonists also reverted GBM and melanoma tumor models to radiation-sensitive phenotype after treatment with 3 Gy. These findings demonstrate that the tumor microenvironment including the survival of tumor-associated endothelial cells contributes to tumor blood vessel resistance to therapy.


Subject(s)
Enzyme Inhibitors/pharmacology , Glioblastoma/radiotherapy , Melanoma, Experimental/radiotherapy , Radiation Tolerance/physiology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptors, Growth Factor/antagonists & inhibitors , Signal Transduction/drug effects , Angiogenesis Inhibitors/pharmacology , Animals , Cell Survival/drug effects , Cell Survival/physiology , Cell Survival/radiation effects , Dose-Response Relationship, Radiation , Endothelial Growth Factors/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/radiation effects , Glioblastoma/blood supply , Indoles/pharmacology , Lymphokines/metabolism , Melanoma, Experimental/blood supply , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/radiotherapy , Pyrroles/pharmacology , Radiation Tolerance/drug effects , Receptor Protein-Tyrosine Kinases/metabolism , Receptor Protein-Tyrosine Kinases/pharmacology , Receptor Protein-Tyrosine Kinases/physiology , Receptors, Growth Factor/metabolism , Receptors, Growth Factor/physiology , Receptors, Vascular Endothelial Growth Factor , Signal Transduction/physiology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
14.
Radiology ; 219(1): 166-70, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11274552

ABSTRACT

PURPOSE: To evaluate the ability of a quantified power Doppler ultrasonography (US) system to help quantitate differences in tumor vascularity after radiation therapy and administration of tumor necrosis factor (TNF). MATERIALS AND METHODS: Murine glioblastoma tumors were grown in the thighs of two sets of 25 mice each. Each mouse was assigned to one of four treatment groups: control (no treatment), radiation therapy, TNF therapy, or combination therapy (both radiation and TNF therapies). Mice were then evaluated with quantified power Doppler US, and a vascularity index (color area) was calculated for different tumor regions in each group. The tumors were then excised, and histologic evaluation was performed by using an immunofluorescence-tagged monoclonal antibody against blood vessel endothelium. The number of stained blood vessels per high-power field was correlated with the sonographically determined vascularity index. RESULTS: The color area of the total tumor decreased to 37% of that in the control group in mice treated with radiation therapy alone (P: =.02), 26% of that in the control group in mice treated with TNF alone (P: =.05), and 8% of that in the control group in those treated with both TNF and radiation (P: =.006). These results correlated well with the quantified results from immunofluorescent staining (r = 0.98). CONCLUSION: Quantified power Doppler US is a noninvasive method for the evaluation of tumor vascularity and blood flow.


Subject(s)
Glioblastoma/blood supply , Neoplasms, Experimental/blood supply , Ultrasonography, Doppler, Color , Animals , Dose Fractionation, Radiation , Glioblastoma/diagnostic imaging , Glioblastoma/pathology , Glioblastoma/radiotherapy , Mice , Mice, Inbred C57BL , Microcirculation , Microscopy, Fluorescence , Neoplasm Transplantation , Neoplasms, Experimental/diagnostic imaging , Neoplasms, Experimental/pathology , Regional Blood Flow/physiology , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/pharmacology
16.
Bone Marrow Transplant ; 25(8): 807-13, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10808200

ABSTRACT

Since approximately 30% of leukemia patients relapse after allogeneic BMT using total body irradiation (TBI)-based preparative regimens, treatment intensity may be suboptimal. The killing of leukemia cells is proportional to the radiation absorbed dose. We studied the feasibility and toxicity of escalating the doses of fractionated TBI above our previous prescription of 13.5 Gy. Sixteen evaluable patients with advanced hematologic malignancies were treated with twice daily TBI using a high-energy source (18-24 MV). The first patient cohort (n = 11) received a total dose of 14.4 Gy in nine fractions, and the second cohort (n = 5) received doses escalated to 15.3 Gy. All patients received high-dose etoposide (60 mg/kg) and allogeneic stem cell transplantation following the TBI. All patients had HLA-identical sibling donors. The median times for neutrophil and platelet engraftment were 13.5 and 12 days, respectively, and did not differ between the two cohorts. All but one patient developed treatment-related grade 3 or 4 mucositis. There were three cases of grade 4 pulmonary toxicity and three cases of grade 4 hepatic toxicity among the 14.4 Gy cohort, and one case each of grade 4 pulmonary and hepatic toxicities among the 15.3 Gy cohort. In most cases comorbid conditions contributed to these toxicities. Two patients had significant GVHD of the GI tract. Six relapses occurred, five (45%) in the 14.4 Gy cohort and one (20%) in the 15.3 Gy cohort. The 100-day treatment-related mortality rates were 9% and 20% for the 14.4 Gy and 15.3 Gy cohorts, respectively, and the median survivals were 226 and 201 days, respectively. We conclude that TBI dose escalation above the previously used 13.5 Gy dose is feasible using a high-energy source and high-dose etoposide. Acute and chronic toxicities were primarily related to GVHD, infection and relapse rather than to TBI.


Subject(s)
Etoposide/therapeutic use , Hematologic Neoplasms/therapy , Hematopoietic Stem Cell Transplantation , Whole-Body Irradiation/methods , Adult , Blood Platelets/cytology , Cohort Studies , Combined Modality Therapy , Etoposide/toxicity , Female , Graft Survival , Graft vs Host Disease/etiology , Heart Failure/etiology , Heart Failure/therapy , Hematologic Neoplasms/complications , Hematologic Neoplasms/radiotherapy , Humans , Liver/radiation effects , Lung/radiation effects , Lung Diseases/etiology , Male , Middle Aged , Mouth Mucosa/radiation effects , Neutrophils/cytology , Recurrence , Stomatitis/drug therapy , Stomatitis/etiology , Survival , Transplantation, Homologous , Treatment Outcome , Whole-Body Irradiation/adverse effects , Whole-Body Irradiation/standards
17.
Cancer Gene Ther ; 7(1): 13-9, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10678351

ABSTRACT

Recombinant adenovirus (Ad) vectors provide a means of local, therapeutic gene delivery to a wide range of neoplasms. Ad-mediated gene therapy trials in malignant glioma models have been limited by the need for high viral titers and multiple dosages. In an attempt to improve Ad vector gene transfer, we studied human (U87, D54) and rodent (GL261, C6) malignant glioma cell lines transfected with various doses of unmodified Ad vectors (AdZ), Ad vectors that contain an alteration of the fiber-coat protein and that direct virus binding to heparan sulfate receptors (AdZ.F(pK7)), and Ad vectors with modifications of the fiber-coat protein that direct virus binding to alpha1, integrin cellular receptors (AdZ.F(RGD)). AdZ.F(pK7) increased the frequency of cells expressing the reporter gene, beta-galactosidase, and improved transduction by 2- to 20-fold compared with AdZ in U87, D54, and GL261 cells. In U87, D54, GL261, and C6 tumors, AdZ.F(pK7) increased gene transfer by 10- to 100-fold compared with AdZ. AdZ.F(RGD) increased gene expression in C6 xenografts compared with AdZ, but had reduced transduction compared with the C6 xenografts of AdZ in all other glioma tumors. These findings suggest that the increased tropisms resulting from alterations of the Ad vector fiber-coat protein as in AdZ.F(pK7) and AdZ.F(RGD) offer a feasible approach to improving in vitro and in vivo transduction efficiencies in certain malignant glioma cell lines.


Subject(s)
Adenoviridae/genetics , Capsid Proteins , Capsid/genetics , Gene Expression Regulation, Neoplastic , Genetic Therapy/methods , Genetic Vectors/genetics , Glioma/therapy , Adenoviridae/metabolism , Gene Transfer Techniques , Genetic Vectors/metabolism , Glioma/genetics , Heparan Sulfate Proteoglycans/metabolism , Humans , Receptors, Virus/metabolism , Tropism , Tumor Cells, Cultured , beta-Galactosidase/metabolism
19.
Oncology (Williston Park) ; 13(10 Suppl 5): 71-7, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10550829

ABSTRACT

Obliteration of the tumor vasculature is an effective means of achieving tumor regression. Antiangiogenic agents have begun to enter cancer clinical trials. Ionizing radiation activates the inflammatory cascade and increases the procoagulative state within blood vessels of both tumors and normal tissues. These responses are mediated through oxidative injury to the endothelium, leading to induction of cell-adhesion molecules and exocytosis of stored proteins from the endothelial cytoplasm. Agents that activate homeostatic responses in the endothelium can enhance thrombosis and vasculitis of irradiated tumor blood vessels. Proinflammatory and prothrombotic biological response modifiers given concurrently with ionizing radiation are known to induce vascular obliteration and necrosis of tumors. Other mechanisms of interaction between antiangiogenic agents and ionizing radiation include the direct cytotoxic effects of these agents. Interactions between drugs and radiation therapy might therefore occur at the level of the vascular endothelium. The importance of this paradigm is that the endothelium might not develop resistance to drugs or radiation because of lessened potential for mutagenesis and clonogenesis. The future design of clinical trials must consider the effects of radiation therapy on the vascular endothelium.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Immunologic Factors/pharmacology , Neoplasms/blood supply , Neoplasms/therapy , Angiogenesis Inhibitors/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Blood Vessels/drug effects , Blood Vessels/radiation effects , Cell Adhesion Molecules/biosynthesis , Combined Modality Therapy , Endothelium, Vascular/drug effects , Endothelium, Vascular/radiation effects , Genetic Therapy , Humans , Immunologic Factors/therapeutic use , Neoplasms/drug therapy , Neoplasms/radiotherapy , P-Selectin/biosynthesis , Radiation, Ionizing , Tumor Necrosis Factor-alpha/pharmacology , Tumor Necrosis Factor-alpha/therapeutic use
20.
Radiat Res ; 152(1): 6-13, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10381836

ABSTRACT

Ionizing radiation induces the inflammatory response in part through leukocyte binding to cell adhesion molecules that are expressed on the vascular endothelium. We studied the effects of X radiation on the pattern of immunohistochemical staining of CD62P (P-selectin). P-selectin was localized within cytoplasmic granules in the untreated vascular endothelium. Immunohistochemical staining of P-selectin was observed at the luminal surface of vascular endothelium within 1 h of irradiation. Increased P-selectin staining at the blood-tissue interface occurred primarily in pulmonary and intestinal blood vessels. To determine whether localization of P-selectin at the vascular lumen occurs through exocytosis of endothelial cell stores in addition to platelet aggregation, we removed the vascular endothelium from the circulation and irradiated endothelial cells in vitro. In this system, we studied the mechanisms by which ionizing radiation induced translocation of P-selectin by using immunofluorescence of human umbilical vein endothelial cells (HUVEC) and confocal microscopy. Prior to irradiation, P-selectin is localized in cytoplasmic reservoirs of HUVEC. After irradiation of HUVEC, P-selectin was translocated to the cell membrane, where it remained tethered. The lowest dose at which we could expect translocation of P-selectin to the cell membrane was 2 Gy. To determine whether P-selectin in Weibel-Palade bodies requires microtubule-dependent membrane transport, we added two microtubule-depolymerizing agents, Colcemid and nocodazole. Microtubule-depolymerizing agents prevented radiation-induced trans- location of P-selectin to the cell membrane. Thus P-selectin accumulates in irradiated blood vessels through both platelet aggregation and microtubule-dependent exocytosis of storage reservoirs within the vascular endothelium.


Subject(s)
Endothelium, Vascular/radiation effects , Lung/radiation effects , Microcirculation/radiation effects , P-Selectin/metabolism , Animals , Cell Membrane/physiology , Cell Membrane/radiation effects , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Exocytosis/radiation effects , Gamma Rays , Humans , Immunohistochemistry , Intestine, Small/blood supply , Intestine, Small/radiation effects , Lung/blood supply , Lung/cytology , Lung/physiology , Mice , Mice, Inbred C57BL , Microcirculation/physiology , P-Selectin/analysis , Umbilical Veins , X-Rays
SELECTION OF CITATIONS
SEARCH DETAIL
...