Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Antimicrob Agents Chemother ; 53(4): 1331-7, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19188381

ABSTRACT

Gallium (Ga) is a semimetallic element that has demonstrated therapeutic and diagnostic-imaging potential in a number of disease settings, including cancer and infectious diseases. Gallium's biological actions stem from its ionic radius being almost the same as that of ferric iron (Fe(3+)), whereby it can replace iron (Fe) in Fe(3+)-dependent biological systems, such as bacterial and mammalian Fe transporters and Fe(3+)-containing enzymes. Unlike Fe(3+), ionic gallium (Ga(3+)) cannot be reduced, and when incorporated, it inactivates Fe(3+)-dependent reduction and oxidation processes that are necessary for bacterial and mammalian cell proliferation. Most pathogenic bacteria require Fe for growth and function, and the availability of Fe in the host or environment can greatly enhance virulence. We examined whether gallium maltolate (GaM), a novel formulation of Ga, had antibacterial activity in a thermally injured acute infection mouse model. Dose-response studies indicated that a GaM dose as low as 25 mg/kg of body weight delivered subcutaneously was sufficient to provide 100% survival in a lethal P. aeruginosa-infected thermally injured mouse model. Mice treated with 100 mg/kg GaM had undetectable levels of Pseudomonas aeruginosa in their wounds, livers, and spleens, while the wounds of untreated mice were colonized with over 10(8) P. aeruginosa CFU/g of tissue and their livers and spleens were colonized with over 10(5) P. aeruginosa CFU/g of tissue. GaM also significantly reduced the colonization of Staphylococcus aureus and Acinetobacter baumannii in the wounds of thermally injured mice. Furthermore, GaM was also therapeutically effective in preventing preestablished P. aeruginosa infections at the site of the injury from spreading systemically. Taken together, our data suggest that GaM is potentially a novel antibacterial agent for the prevention and treatment of wound infections following thermal injury.


Subject(s)
Burns/complications , Organometallic Compounds/therapeutic use , Pseudomonas Infections/drug therapy , Pyrones/therapeutic use , Acinetobacter baumannii/drug effects , Animals , Burns/microbiology , Female , Gallium/therapeutic use , Mice , Staphylococcus aureus/drug effects
2.
Clin Cancer Res ; 14(11): 3514-9, 2008 Jun 01.
Article in English | MEDLINE | ID: mdl-18519784

ABSTRACT

PURPOSE: To assess whether adenovirus-mediated retinoblastoma 94 (Ad-RB94) transgene expression enhances efficacy of radiation therapy (XRT) of human head and neck squamous cell carcinoma (HNSCC). EXPERIMENTAL DESIGN: The HNSCC cell lines (JHU006 and JHU012) were treated in vitro and in a nude mouse xenograft model with Ad-RB94, Ad-DL312 control vector, or untreated as mock control. Cell viability and tumor growth were evaluated and combined RB94/XRT antitumor activity was analyzed by measuring DNA double-strand breaks, apoptosis-associated early DNA fragmentation, and levels of RB-regulated cell cycle progression E2F1 transcription factor. RESULTS: Ad-RB94/XRT resulted in significant HNSCC cell growth inhibition compared with XRT alone or Ad-RB94 alone in vitro and caused significant tumor regression compared with XRT alone and Ad-DL312/XRT in JHU006 and with XRT alone, Ad-DL312/XRT and Ad-RB94 alone in JHU012 in vivo. Neutral comet analysis revealed that DNA damage was significantly elevated in cells treated with Ad-RB94 alone and Ad-RB94/XRT. Tumors treated with Ad-RB94 alone showed a striking increase in early apoptosis DNA fragmentation, and DNA fragmentation was further enhanced with XRT. In addition, levels of E2F1 were up-regulated by Ad-RB94/XRT combination, whereas Ad-RB94 alone did not affect E2F1 levels and XRT alone led to down-regulation of E2F1. CONCLUSIONS: A potent antitumor effect has been observed after Ad-RB94/XRT combination treatment in HNSCC xenograft tumors. Enhanced tumor regression correlated with increased apoptosis. Ad-RB94 treatment enhances the efficacy of XRT through tumor cell sensitization by arresting the cells at the radiation-sensitive G(2)-M cell cycle and via E2F1 up-regulation.


Subject(s)
Carcinoma, Squamous Cell/radiotherapy , Genetic Therapy/methods , Head and Neck Neoplasms/radiotherapy , Retinoblastoma Protein/genetics , Adenoviridae , Animals , Cell Line, Tumor , Comet Assay , DNA Fragmentation , Female , Genetic Vectors , Humans , Immunohistochemistry , Mice , Mice, Nude , Radiotherapy , Transgenes , Xenograft Model Antitumor Assays
3.
Lung Cancer ; 45(3): 381-6, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15301879

ABSTRACT

This multicenter phase II trial evaluated the therapeutic activity and safety profile of pivaloyloxymethyl butyrate (Pivanex, AN-9) as a single agent in refractory non-small cell lung cancer (NSCLC). Pivanex (2.34 g/m2 per day) was administered as a 6-h continuous intravenous infusion, daily for 3 days, and repeated every 21 days until disease progression. Forty-seven patients were treated. More than 90% of patients had received both a platinum compound and a taxane and 32% had received three or more prior chemotherapy regimens. The most common toxicities were transient grade 1-2 fatigue (34%), nausea (17%), and dysgeusia (11%). Three patients had partial responses (6.4 and 95%; CI 1.4-18.7%) and 14 patients had stable disease for > or =12 weeks (30%). Median survival for all patients was 6.2 months with 1-year survival of 26%. For patients who received fewer than three prior chemotherapy regimens, median survival was 7.8 months and 1-year survival was 31%. Pivanex is well tolerated and appears to be active as a single agent in patients with advanced NSCLC refractory to previous chemotherapy. Based on its therapeutic activity and favorable safety profile, further studies of Pivanex in NSCLC, particularly in combination with current chemotherapeutic agents, are warranted.


Subject(s)
Butyrates/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Butyrates/administration & dosage , Butyrates/adverse effects , Carcinoma, Non-Small-Cell Lung/pathology , Drug Administration Schedule , Fatigue/chemically induced , Female , Humans , Infusions, Intravenous , Lung Neoplasms/pathology , Male , Middle Aged , Nausea/chemically induced , Survival Analysis
4.
Cancer Res ; 64(14): 4995-5003, 2004 Jul 15.
Article in English | MEDLINE | ID: mdl-15256474

ABSTRACT

In this report, we have studied the immunogenicity of the nominal antigen, carcinoembryonic antigen (CEA), and that of an anti-idiotype antibody, 3H1, which mimics CEA and can be used as a surrogate for CEA. We have demonstrated that immunization of CEA transgenic mice with bone marrow-derived mature dendritic cells (DC) loaded with anti-idiotype 3H1 or CEA could reverse CEA unresponsiveness and result in the induction of CEA-specific immune responses and the rejection of CEA-transfected MC-38 colon carcinoma cells, C15. Immunized mice splenocytes proliferated in an antigen-specific manner by a mechanism dependent on the functions of CD4, MHC II, B7-2, CD40, CD28, and CD25. However, immune splenic lymphocytes isolated from 3H1-DC-vaccinated mice when stimulated in vitro with 3H1 or CEA secreted significantly higher levels of Th1 cytokines than did CEA-DC vaccinated mice. DC vaccination also induced antigen-specific effector CD8+ T cells capable of expressing interleukin-2, IFN-gamma, and tumor necrosis factor (TNF)-alpha and displayed cytotoxic activity against C15 cells in an MHC class I-restricted manner. 3H1-DC vaccination resulted in augmented CTL responses and the elevated expression of CD69, CD25, and CD28 on CD8(+) CTLs. The immune responses developed in 3H1-DC-immunized mice resulted in rejection of C15 tumor cells in nearly 100% of experimental mice, whereas only 40% of experimental mice immunized with CEA-DC were protected from C15 tumor growth. These findings suggest that under the experimental conditions used, 3H1-DC vaccination was better than CEA-DC vaccination in breaking immune tolerance to CEA and inducing protective antitumor immune responses in this murine model transgenic for human CEA.


Subject(s)
Antibodies, Anti-Idiotypic/immunology , Carcinoembryonic Antigen/immunology , Colonic Neoplasms/immunology , Dendritic Cells/immunology , Immunotherapy, Adoptive/methods , Animals , Colonic Neoplasms/prevention & control , Colonic Neoplasms/therapy , Cytokines/biosynthesis , Cytokines/immunology , Cytotoxicity, Immunologic , Epitopes, T-Lymphocyte/immunology , Female , Humans , Immune Tolerance/immunology , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Up-Regulation
5.
Clin Cancer Res ; 10(4): 1454-62, 2004 Feb 15.
Article in English | MEDLINE | ID: mdl-14977849

ABSTRACT

PURPOSE: Gene transfer of a truncated variant of the retinoblastoma (RB) gene encoding a M(r) 94000 protein that lacks the NH(2)-terminal 112 amino acid residues, termed RB94, has been shown to inhibit proliferation of several human tumor cell types. We have assessed its therapeutic effectiveness on pancreatic cancer, one of the most aggressive and therapy-resistant types of cancer. For this purpose, preclinical studies aimed to evaluate the therapeutic potential of RB94 gene transfer in pancreatic cancer were carried out. EXPERIMENTAL DESIGN: We have compared the antiproliferative effects of adenovirus-mediated gene transfer of RBwt and RB94 at the in vitro and in vivo levels in three RB-positive human pancreatic tumor cell lines: (a). NP-9; (b). NP-18; and (c). NP-31. We have also examined their effects on cell cycle and their capacity to induce apoptosis. RESULTS: In vitro results indicate that RB94 gene transfer has stronger antiproliferative effects compared with RBwt. RB94 transduction correlated with accumulation at the S-G(2) phase of the cell cycle in the three cell lines tested and induction of apoptosis in two of them. In vivo studies show significant decreases in the growth rate of tumors treated with Ad-RB94 when compared with those treated with Ad-RBwt. Moreover, terminal deoxynucleotidyl transferase-mediated nick end labeling analyses of Ad-RB94-treated tumor sections revealed that only RB94 is able to significantly induce apoptosis. CONCLUSIONS: RB94 gene expression has antiproliferative effects also in human pancreatic tumor cells, being more effective than wild-type RB in preventing tumor growth.


Subject(s)
Adenoviridae/genetics , Gene Transfer Techniques , Pancreatic Neoplasms/therapy , Retinoblastoma Protein/genetics , Animals , Annexin A5/pharmacology , Apoptosis , Blotting, Western , Cell Cycle , Cell Division , Cell Line, Tumor , Coloring Agents/pharmacology , Dose-Response Relationship, Drug , Humans , In Situ Nick-End Labeling , Mice , Mice, Nude , Neoplasm Transplantation , Pancreatic Neoplasms/genetics , Protein Structure, Tertiary , Time Factors
6.
Cancer Res ; 62(16): 4637-44, 2002 Aug 15.
Article in English | MEDLINE | ID: mdl-12183420

ABSTRACT

A truncated retinoblastoma (RB) protein of approximately 94 kDa (RB94), lacking the NH2 -terminal 112 amino acid residues of the full-length RB, has been found to have great efficacy in tumor suppression. This study investigated the role of adenovirus-mediated RB94 (Ad-RB94) gene therapy for human head and neck squamous cell carcinoma (HNSCC) and explored the cellular and molecular mechanism of tumor inhibition after Ad-RB94 gene transfer. Randomized controlled studies in vitro and in vivo were performed to assess antitumor responses of Ad-RB94 gene transfer against human HNSCC. Human HNSCC cell lines, JHU006 and JHU012, were used in this study. Tumors originated from the HNSCC cell lines were propagated as xenografts in nude mice. Ad-RB94 gene transfer was performed both in vitro and in vivo with replication-defective virus (DL312) and no treatment as controls. Transgene expression, cell viability, and tumor growth were evaluated in transfected cells and tumor implants. To determine the mechanism behind the observed antitumor action, cell cycle analysis was performed, and telomerase activity was examined. Tumors were evaluated for RB94-induced apoptosis. Transgene expression of RB94 was detected by Western blot analysis, real-time quantification reverse transcription-PCR, and immunohistochemistry. RB94 expression led to flattening of cell growth curves and caused tumor regression. Animals treated with Ad-RB94 were seen to have a significant reduction in tumor size when compared with DL312 (P = 0.02, both cell lines) and to no treatment groups (P = 0.01, both cell lines). Cell cycle arrest in the G(2)-M phase and increased levels of apoptosis occurred in tumor cells treated with Ad-RB94. In addition, telomerase activity decreased significantly and specifically after Ad-RB94 treatment. This study demonstrates that Ad-RB94 gene transfer effectively inhibits HNSCC tumor cell growth in vitro and in vivo. The unique property of Ad-RB94 gene transfer to arrest HNSCC tumor cells in the G2-M phase of the cell cycle makes it a good candidate for adjuvant therapy with radiation or chemotherapy, as tumor cells are most sensitive to radiation or cytotoxic drug in this cell cycle phase.


Subject(s)
Carcinoma, Squamous Cell/therapy , Carrier Proteins/physiology , Genetic Therapy/methods , Head and Neck Neoplasms/therapy , Ubiquitin-Protein Ligases , Adenoviridae/genetics , Animals , Apoptosis/genetics , Apoptosis/physiology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Cell Division/genetics , Female , G2 Phase/genetics , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Immunohistochemistry , Mice , Mice, Inbred BALB C , Mice, Nude , Mitosis/genetics , Random Allocation , Reverse Transcriptase Polymerase Chain Reaction , Telomerase/antagonists & inhibitors , Telomerase/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...