Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters











Publication year range
1.
Viruses ; 11(11)2019 10 26.
Article in English | MEDLINE | ID: mdl-31717800

ABSTRACT

Peptide-displayed phage libraries are billion-clone collections of diverse chimeric bacteriophage particles, decorated by genetically fused peptides built from a random combination of natural amino acids. Studying the molecular evolution of peptide-displayed libraries in mammalian model systems, using in vivo phage display techniques, can provide invaluable knowledge about the underlying physiology of the vasculature system, allow recognition of organ- and tissue-specific networks of protein-protein interactions, and provide ligands for targeted diagnostics and therapeutics. Recently, we discovered that landscape phage libraries, a specific type of multivalent peptide phage display library, expose on their surface comprehensive collections of elementary binding units (EBUs), which can form short linear motifs (SLiMs) that interact with functional domains of physiologically relevant proteins. Because of their unique structural and functional features, landscape phages can use an alternative mechanism of directed molecular evolution, i.e., combinatorial avidity selection. These discoveries fueled our interest in revisiting the in vivo evolution of phage displayed libraries using another format of display, i.e., landscape phages. In this study, we monitored the evolution of a landscape phage library in a mouse model with and without an implanted human breast cancer tumor xenograft. As expected, the multivalent architecture of landscape phage displayed proteins provided strong tissue selectivity and resulted in a huge diversity of tissue penetrating, chimeric phage particles. We identified several types of EBU interactions that evolved during the course of tissue distribution, which included interactions of EBUs with all tissue types, those EBUs that interacted selectively with specific organs or tissues with shared gene expression profiles or functionalities, and other EBUs that interacted in a tissue-selective manner. We demonstrated that landscape phage libraries are a rich collection of unique nanobioparticles that can be used to identify functional organ and tissue-binding elements after the evolution of a phage display library in vivo.


Subject(s)
Bacteriophages/genetics , Breast Neoplasms/genetics , Cell Surface Display Techniques/methods , Gene Library , Animals , Evolution, Molecular , Female , Heterografts , Humans , Mice , Mice, Nude
2.
Pharmaceuticals (Basel) ; 11(2)2018 04 23.
Article in English | MEDLINE | ID: mdl-29690576

ABSTRACT

5-aza-2',2'-difluorodeoxycytidine (NUC013) has been shown to be significantly safer and more effective than decitabine in xenograft models of human leukemia and colon cancer. However, it suffers from a similar short half-life as other DNA methyltransferase inhibitors with a 5-azacytosine base, which is problematic for nucleosides that primarily target tumor cells in S phase. Because of the relative instability of 5-azanucleosides, a prodrug approach was developed to improve the pharmacology of NUC013. NUC013 was conjugated with trimethylsilanol (TMS) at the 3' and 5' position of the sugar, rendering the molecule hydrophobic and producing 3',5'-di-trimethylsilyl-2',2'-difluoro-5-azadeoxycytidine (NUC041). NUC041 was designed to be formulated in a hydrophobic vehicle, protecting it from deamination and hydrolysis. In contact with blood, the TMS moieties are readily hydrolyzed to release NUC013. The half-life of NUC013 administered intravenously in mice is 20.1 min, while that of NUC013 derived from intramuscular NUC041 formulated in a pegylated-phospholipid depot is 3.4 h. In a NCI-H460 xenograft of non-small cell lung cancer, NUC013 was shown to significantly inhibit tumor growth and improve survival. Treatment with NUC041 also led to significant tumor growth inhibition. However, NUC041-treated mice had significantly more tumors ulcerate than either NUC013 treated mice or saline control mice, and such ulceration occurred at significantly lower tumor volumes. In these nude mice, tumor regression was likely mediated by the derepression of the tumor suppressor gene p53 and resultant activation of natural killer (NK) cells.

3.
Pharmaceuticals (Basel) ; 11(1)2018 Feb 06.
Article in English | MEDLINE | ID: mdl-29415423

ABSTRACT

Vitamin E phosphate (VEP) nucleoside prodrugs are designed to bypass two mechanisms of tumor resistance to therapeutic nucleosides: nucleoside transport and kinase downregulation. Certain isoforms of vitamin E (VE) have shown activity against solid and hematologic tumors and result in chemosensitization. Because gemcitabine is one of the most common chemotherapeutics for the treatment of cancer, it was used to demonstrate the constructs utility. Four different VE isoforms were conjugated with gemcitabine at the 5' position. Two of these were δ-tocopherol-monophosphate (MP) gemcitabine (NUC050) and δ-tocotrienol-MP gemcitabine (NUC052). NUC050 was shown to be able to deliver gemcitabine-MP intracellularly by a nucleoside transport independent mechanism. Its half-life administered IV in mice was 3.9 h. In a mouse xenograft model of non-small cell lung cancer (NSCLC) NCI-H460, NUC050 at a dose of 40 mg/kg IV qwk × 4 resulted in significant inhibition to tumor growth on days 11-31 (p < 0.05) compared to saline control (SC). Median survival was 33 days (NUC050) vs. 25.5 days (SC) ((hazard ratio) HR = 0.24, p = 0.017). Further, NUC050 significantly inhibited tumor growth compared to historic data with gemcitabine at 135 mg/kg IV q5d × 3 on days 14-41 (p < 0.05). NUC052 was administered at a dose of 40 mg/kg IV qwk × 2 followed by 50 mg/kg qwk × 2. NUC052 resulted in inhibition to tumor growth on days 14-27 (p < 0.05) and median survival was 34 days (HR = 0.27, p = 0.033). NUC050 and NUC052 have been shown to be safe and effective in a mouse xenograft of NSCLC.

4.
ACS Med Chem Lett ; 7(2): 177-81, 2016 Feb 11.
Article in English | MEDLINE | ID: mdl-26985294

ABSTRACT

In this letter we report first nonpeptide inhibitors of hepatocyte growth factor (HGF) activation. These compounds inhibit the three proteases (matriptase, hepsin, and HGF activator) required for HGF maturation. We show that 6, 8a, 8b, and 8d block activation of fibroblast-derived pro-HGF, thus preventing fibroblast-induced scattering of DU145 prostate cancer cells. Compound 6 (SRI 31215) is very soluble (91 µM) and has excellent microsome stability (human t 1/2 = 162 min; mouse t 1/2 = 296 min). In mouse 6 has an in vivo t 1/2 = 5.8 h following IV administration. The high solubility of 6 and IV t 1/2 make this compound a suitable prototype "triplex inhibitor" for the study of the inhibition of HGF activation in vivo.

5.
Radiother Oncol ; 108(3): 403-8, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23891091

ABSTRACT

PURPOSE: Activation of the DNA damage responsive protein kinase ATM is a critical step for cellular survival in response to ionizing irradiation (IR). Direct targets of ATM regulating radiosensitivity remain to be fully investigated. We have recently reported that ATM phosphorylates the transcriptional repressor Snail on Serine 100. We aimed to further study the functional significance of ATM-mediated Snail phosphorylation in response to IR. MATERIAL AND METHODS: We transfected vector-only, wild-type, the Serine 100 to alanine (S100A) or to glutamic acid (S100E) substitution of Snail into various cell lines. We assessed colony formation, γ-H2AX focus formation and the invasion index in the cells treated with or without IR. RESULTS: We found that over-expression of the S100A mutant Snail in HeLa cells significantly increased radiosensitivity. Meanwhile the expression of S100E, a phospho-mimicking mutation, resulted in enhanced radio-resistance. Interestingly, S100E could rescue the radiosensitive phenotype in ATM-deficient cells. We also found that expression of S100E increased γ-H2AX focus formation and compromised inhibition of invasion in response to IR independent of cell survival. CONCLUSION: ATM-mediated Snail Serine 100 phosphorylation in response to IR plays an important part in the regulation of radiosensitivity.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/physiology , Neoplasms/radiotherapy , Radiation Tolerance , Transcription Factors/metabolism , Histones/analysis , Humans , Neoplasm Invasiveness , Phosphorylation , Serine , Snail Family Transcription Factors
6.
Article in English | MEDLINE | ID: mdl-23004932

ABSTRACT

Clofarabine, an approved anticancer drug, was evaluated in combination with radiation in six subcutaneously implanted human tumor xenograft models. Clofarabine had no effect on the growth of SF-295 glioblastoma, which was not enhanced by radiation. There was no difference between clofarabine with and without radiation in the DU-145 prostate model. The combined effect on NCI-H460 lung tumors appeared to be additive. SR475 head and neck, PANC-1 pancreatic, and HCT-116 colon tumors were radiomodified by clofarabine. The radiomodifying capacity of clofarabine was superior to that for gemcitabine in two models (PANC-1 and HCT-116) and was comparable in the other four models.


Subject(s)
Adenine Nucleotides/therapeutic use , Antineoplastic Agents/therapeutic use , Arabinonucleosides/therapeutic use , Neoplasms, Experimental/therapy , Adenine Nucleotides/administration & dosage , Animals , Antineoplastic Agents/administration & dosage , Arabinonucleosides/administration & dosage , Cell Line, Tumor , Clofarabine , Combined Modality Therapy , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Drug Evaluation, Preclinical , Humans , Mice , Mice, Nude , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/radiotherapy , Tumor Burden/drug effects , Xenograft Model Antitumor Assays , Gemcitabine
7.
Int J Radiat Oncol Biol Phys ; 70(1): 213-20, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-18037589

ABSTRACT

PURPOSE: Combination treatment with radiotherapy and chemotherapy has emerged as the dominant form of cancer adjuvant regimens in recent years. Clofarabine, a newly approved drug for pediatric leukemia, is a second-generation purine nucleoside analogue that can block DNA synthesis and inhibit DNA repair. Therefore, we hypothesized that clofarabine could work synergistically with radiotherapy to increase the tumor cell response. METHODS AND MATERIALS: The effects of clofarabine on radiosensitivity have been established in several tumor cell lines in vitro and in vivo using colony-forming assays and tumor xenografts. The effect of clofarabine on the DNA damage response was also studied in vitro by measuring gamma-H2AX focus formation. RESULTS: Clonogenic survival was significantly reduced in irradiated cells treated with clofarabine, demonstrating the strong radiosensitizing effect of clofarabine. Furthermore, clofarabine displayed a radiosensitizing effect that was greater than gemcitabine or 5-fluorouracil. We also found that low doses of clofarabine can prolong the presence of radiation-induced gamma-H2AX nuclear focus formation, and high doses of clofarabine can induce DNA double-strand breaks, suggesting that clofarabine can interfere with DNA damage response pathways. In addition, clofarabine-induced radiosensitization was also established in vivo using a colorectal cancer model, DLD-1, in athymic nude mice. When combined with fractionated radiotherapy, a moderate dose of clofarabine led to a significant increase in tumor growth inhibition. CONCLUSION: Clofarabine acts as a powerful radiosensitizer both in vitro and in vivo by interfering with the DNA damage response.


Subject(s)
Adenine Nucleotides/pharmacology , Arabinonucleosides/pharmacology , DNA Damage , DNA Repair/drug effects , Radiation Tolerance/drug effects , Radiation-Sensitizing Agents/pharmacology , Animals , Cell Line, Tumor , Clofarabine , DNA, Neoplasm/drug effects , DNA, Neoplasm/radiation effects , HeLa Cells , Histones/metabolism , Humans , Male , Mice , Mice, Nude , Radiation Dosage , Xenograft Model Antitumor Assays
8.
Int J Radiat Oncol Biol Phys ; 55(4): 1037-50, 2003 Mar 15.
Article in English | MEDLINE | ID: mdl-12605984

ABSTRACT

PURPOSE: We explore the utility of the adenovirus-mediated delivery of proapoptotic Bax for enhancing the cytotoxicity of radiotherapy (RT) in RT-refractory glioma cells. MATERIALS AND METHODS: Cell lines D54 MG and U87 MG (p53 wild-type), and U251 MG and U373 MG (p53 mutant), and patient-derived astrocytes were evaluated. Cells were irradiated and infected with an inducible adenovirus encoding Bax. Cell proliferation, colony formation assay, quantification of early apoptotic alteration in the plasma membrane by fluorescence-activated cell sorter using annexin V, and nuclear staining with H33258 were used to evaluate apoptosis. The capacity of the combined treatment to induce regression of subcutaneous D54 MG tumors was tested in nude mice. A dose of 5 Gy was administered every other day, four times, for a total dose of 20 Gy. One day after each irradiation, tumors were injected with 1 x 10(9) plaque-forming units (PFU). RESULTS: Apoptotic death was enhanced by the combination of Ad/Bax and RT. In D54 MG, levels of apoptosis after RT alone, Ad/Bax alone, or the combination were, respectively, 12.3%, 32.1%, and 78.5%. In contrast, treatment of astrocytes did not significantly induce apoptosis. A colony-formation assay showed a 2-log inhibition with respect to controls after combined treatment, irrespective of the endogenous levels of p53. The other apoptosis assays also showed the defining characteristics of apoptosis in the combination group. Remarkably, combined treatment induced regression of tumors in mice. CONCLUSIONS: Ad/Bax synergistically radiosensitizes glioma, with a seemingly favorable therapeutic index.


Subject(s)
Adenoviridae/genetics , Apoptosis/physiology , Brain Neoplasms/therapy , Genetic Therapy/methods , Genetic Vectors/therapeutic use , Glioma/therapy , Proto-Oncogene Proteins c-bcl-2 , Proto-Oncogene Proteins/genetics , Analysis of Variance , Animals , Apoptosis/genetics , Astrocytes/radiation effects , Astrocytes/virology , Brain Neoplasms/radiotherapy , Cell Survival/physiology , Combined Modality Therapy , Dose-Response Relationship, Radiation , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor/physiology , Genes, p53/physiology , Glioma/radiotherapy , Humans , Mice , Mice, Nude , Tumor Cells, Cultured , Tumor Stem Cell Assay , bcl-2-Associated X Protein
9.
Int J Radiat Oncol Biol Phys ; 54(4): 1180-93, 2002 Nov 15.
Article in English | MEDLINE | ID: mdl-12419447

ABSTRACT

PURPOSE: To investigate treatment of human pancreatic cancer cell lines and xenografts with combinations of Erbitux (IMC-C225) anti-epidermal growth factor receptor (EGFR) antibody, gemcitabine, and radiation. METHODS AND MATERIALS: BxPC-3 and MiaPaCa-2 human pancreatic carcinoma cells were treated in vitro for 24 h with IMC-C225 (5 microg/mL), then exposed to epidermal growth factor (EGF) (10 mM) for 5 min. Immunoblots were screened for EGFR expression and the ability of IMC-C225 to block EGF-induced tyrosine phosphorylation of EGFR. Cells were treated with IMC-C225 (5 microg/mL) on Day 0, the IC(50) dose of gemcitabine on Day 1 for 24 h, followed by 3 Gy 60Co irradiation on Day 2, or the combination of each agent. For cell proliferation, cells were counted on Day 4, and for apoptosis, cells were stained with annexin V-FITC and propidium iodide, then analyzed by FACS. Cells were treated with the same single or multiple treatments and analyzed in a clonogenic cell survival assay. The effect of IMC-C225, gemcitabine, and radiation on the growth of BxPC-3 and MiaPaCa-2 tumor xenografts was determined. Athymic nude mice bearing established s.c. tumor xenografts of 6-8 mm diameter received 6 weeks of treatment with IMC-C225 (1 mg every 3 days x 6) alone or in combination with gemcitabine (120 mg/kg i.v. every 6 days x 6), and 6 weekly fractions of 3 Gy radiation on the days after gemcitabine administration. Tumor growth was measured with Vernier calipers. RESULTS: BxPC-3 and MiaPaCa-2 cell lines expressed low levels of EGFR. IMC-C225 inhibited EGF-induced tyrosine phosphorylation of the EGF receptor on both cell lines. Treatment of cells with a combination of IMC-C225 + gemcitabine + radiation produced the highest induction of apoptosis and inhibition of proliferation in vitro. Combination treatment with IMC-C225, gemcitabine, and radiation produced 100% complete regression of MiaPaCa-2 tumors for more than 250 days, and the greatest growth inhibition of BxPC-3 tumors compared to any single or dual treatments. CONCLUSIONS: The IMC-C225 therapy in combination with gemcitabine chemotherapy and radiation therapy demonstrated statistically significantly greater efficacy over the single and double combination therapies. This form of multimodality treatment shows potential clinical application in the treatment of pancreatic cancer in humans.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , ErbB Receptors/antagonists & inhibitors , Pancreatic Neoplasms/therapy , Animals , Antibodies, Monoclonal, Humanized , Apoptosis , Cell Cycle/drug effects , Cell Cycle/radiation effects , Cell Division , Cell Survival/drug effects , Cell Survival/radiation effects , Cetuximab , Combined Modality Therapy , ErbB Receptors/analysis , ErbB Receptors/metabolism , Female , Humans , Immunohistochemistry , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Phosphorylation , Transplantation, Heterologous , Tumor Cells, Cultured , Gemcitabine
10.
Mol Ther ; 6(2): 190-8, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12161185

ABSTRACT

Apoptosis induction is a promising approach for cancer gene therapy. Bax is a death-promoting member of the Bcl2 family of genes that are intimately involved in apoptosis. Overexpression of BAX protein can accelerate cell death by homodimers that promote apoptosis in a variety of cancer cell lines. The cytotoxic effect of BAX was evaluated in vitro by a recombinant adenovirus system expressing the human BAX gene under control of human vascular endothelial growth factor (VEGF) promoter element (AdVEGFBAX). Overexpression of BAX in human lung carcinoma cells resulted in apoptosis induction, caspase activation, and cell growth suppression, none of which were observed in BEAS-2B normal human bronchial epithelial cells that do not overexpress VEGF under normoxic conditions. To examine the hypoxia responsiveness of the VEGF promoter, lung cancer cells were transiently exposed to hypoxia; this treatment increased enhanced green fluorescent protein (EGFP) expression after AdVEGFEGFP infection in both normal and cancer cell lines, and enhanced apoptosis and decreased the number of surviving cancer cells compared with the Ad/BAX plus Ad/Cre binary adenoviral system. These results suggest a possible therapeutic application of cancer-specific expression of the pro-apoptotic Bax gene driven by the VEGF promoter.


Subject(s)
Adenoviridae/genetics , Genetic Therapy/methods , Lung Neoplasms/therapy , Proto-Oncogene Proteins c-bcl-2 , Proto-Oncogene Proteins/genetics , Apoptosis , Cell Hypoxia , Endothelial Growth Factors/genetics , Gene Expression , Gene Transfer Techniques , Green Fluorescent Proteins , Humans , Luminescent Proteins/genetics , Lung Neoplasms/pathology , Lymphokines/genetics , Promoter Regions, Genetic , Recombinant Proteins/genetics , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , bcl-2-Associated X Protein
SELECTION OF CITATIONS
SEARCH DETAIL