Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 91
Filter
1.
Cancer Gene Ther ; 21(7): 264-74, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24903014

ABSTRACT

Pancreatic cancer is a significant clinical problem and novel therapeutic approaches are desperately needed. Recent advances in conditionally replicative adenovirus-based (CRAd) oncolytic virus design allow the application of CRAd vectors as a therapeutic strategy to efficiently target and eradicate chemoresistant pancreatic cancer cells, thereby improving the efficacy of pancreatic cancer treatment. The goal of this study was to construct and validate the efficacy of an infectivity-enhanced, liver-untargeted, tumor-specific CRAd vector. A panel of CRAds has been derived that embodies the C-X-C chemokine receptor type 4 promoter for conditional replication, two-fiber complex mosaicism for targeting expansion and hexon hypervariable region 7 (HVR7) modification for liver untargeting. We evaluated CRAds for cancer virotherapy using a human pancreatic tumor xenograft model. Employment of the fiber mosaic approach improved CRAd replication in pancreatic tumor xenografts. Substitution of the HVR7 of the Ad5 hexon for Ad serotype 3 hexon resulted in decreased liver tropism of systemically administrated CRAd. Obtained data demonstrated that employment of complex mosaicism increased efficacy of the combination of oncolytic virotherapy with chemotherapy in a human pancreatic tumor xenograft model.


Subject(s)
Adenoviridae/genetics , Oncolytic Virotherapy/methods , Pancreatic Neoplasms/therapy , Pancreatic Neoplasms/virology , Receptors, CXCR4/genetics , Animals , Female , Genetic Vectors/genetics , Humans , Mice , Mice, Nude , Random Allocation , Receptors, CXCR4/metabolism , Xenograft Model Antitumor Assays , Pancreatic Neoplasms
2.
Oncogene ; 33(20): 2620-8, 2014 May 15.
Article in English | MEDLINE | ID: mdl-23770854

ABSTRACT

Epithelial-mesenchymal transition is one of the critical cellular programs that facilitate the progression of breast cancer to an invasive disease. We have observed that the expression of N-myc interactor (NMI) decreases significantly during progression of breast cancer, specifically in invasive and metastatic stages. Recapitulation of this loss in breast cell lines with epithelial morphology (MCF10A (non-tumorigenic) and T47D (tumorigenic)) by silencing NMI expression causes mesenchymal-like morphological changes in 3D growth, accompanied by upregulation of SLUG and ZEB2 and increased invasive properties. Conversely, we found that restoring NMI expression attenuated the mesenchymal attributes of metastatic breast cancer cells, accompanied by distinctly circumscribed 3D growth with basement membrane deposition and decreased invasion. Further investigations into the downstream signaling modulated by NMI revealed that NMI expression negatively regulates SMAD signaling, which is a key regulator of cellular plasticity. We demonstrate that NMI blocks TGF-ß/SMAD signaling via upregulation of SMAD7, a negative feedback regulator of the pathway. We also provide evidence that NMI activates STAT signaling, which negatively modulates TGF-ß/SMAD signaling. Taken together, our findings suggest that loss of NMI during breast cancer progression could be one of the driving factors that enhance the invasive ability of breast cancer by aberrant activation of TGF-ß/SMAD signaling.


Subject(s)
Epithelial-Mesenchymal Transition , Proto-Oncogene Proteins c-myc/physiology , Signal Transduction , Smad Proteins/metabolism , Transforming Growth Factor beta/metabolism , Cell Line , Gene Silencing , Humans , Proto-Oncogene Proteins c-myc/genetics , Up-Regulation
3.
Curr Pharm Des ; 17(21): 2221-38, 2011.
Article in English | MEDLINE | ID: mdl-21777178

ABSTRACT

Human pancreatic cancer remains a highly malignant disease with almost similar incidence and mortality despite extensive research. Many targeted therapies are under development. However, clinical investigation showed that single targeted therapies and most combined therapies were not able to improve the prognosis of this disease, even though some of these therapies had excellent anti-tumor effects in pre-clinical models. Cross-talk between cell proliferation signaling pathways may be an important phenomenon in pancreatic cancer, which may result in cancer cell survival even though some pathways are blocked by targeted therapy. Pancreatic cancer may possess different characteristics and targets in different stages of pathogenesis, maintenance and metastasis. Sensitivity to therapy may also vary for cancer cells at different stages. The unique pancreatic cancer structure with abundant stroma creates a tumor microenvironment with hypoxia and low blood perfusion rate, which prevents drug delivery to cancer cells. In this review, the most commonly investigated targeted therapies in pancreatic cancer treatment are discussed. However, how to combine these targeted therapies and/or combine them with chemotherapy to improve the survival rate of pancreatic cancer is still a challenge. Genomic and proteomic studies using pancreatic cancer samples obtained from either biopsy or surgery are recommended to individualize tumor characters and to perform drug sensitivity study in order to design a tailored therapy with minimal side effects. These studies may help to further investigate tumor pathogenesis, maintenance and metastasis to create cellular expression profiles at different stages. Integration of the information obtained needs to be performed from multiple levels and dimensions in order to develop a successful targeted therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Pancreatic Neoplasms , Animals , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Clinical Trials as Topic , Drug Resistance, Neoplasm/drug effects , Genetic Therapy/methods , Humans , Immunotherapy/methods , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Treatment Outcome
4.
Cancer Chemother Pharmacol ; 67(4): 891-7, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20589377

ABSTRACT

PURPOSE: (1) To determine the safety of the epidermal growth factor receptor (EGFR) antibody cetuximab with concurrent gemcitabine and abdominal radiation in the treatment of patients with locally advanced adenocarcinoma of the pancreas. (2) To evaluate the feasibility of pancreatic cancer cell epithelial-mesenchymal transition (EMT) molecular profiling as a potential predictor of response to anti-EGFR treatment. METHODS: Patients with non-metastatic, locally advanced pancreatic cancer were treated in this dose escalation study with gemcitabine (0-300 mg/m(2)/week) given concurrently with cetuximab (400 mg/m(2) loading dose, 250 mg/m(2) weekly maintenance dose) and abdominal irradiation (50.4 Gy). Expression of E-cadherin and vimentin was assessed by immunohistochemistry in diagnostic endoscopic ultrasound fine-needle aspiration (EUS-FNA) specimens. RESULTS: Sixteen patients were enrolled in 4 treatment cohorts with escalating doses of gemcitabine. Incidence of grade 1-2 adverse events was 96%, and incidence of 3-4 adverse events was 9%. There were no treatment-related mortalities. Two patients who exhibited favorable treatment response underwent surgical exploration and were intraoperatively confirmed to have unresectable tumors. Median overall survival was 10.5 months. Pancreatic cancer cell expression of E-cadherin and vimentin was successfully determined in EUS-FNA specimens from 4 patients. CONCLUSIONS: Cetuximab can be safely administered with abdominal radiation and concurrent gemcitabine (up to 300 mg/m(2)/week) in patients with locally advanced adenocarcinoma of the pancreas. This combined therapy modality exhibited limited activity. Diagnostic EUS-FNA specimens could be analyzed for molecular markers of EMT in a minority of patients with pancreatic cancer.


Subject(s)
Adenocarcinoma/therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , ErbB Receptors/antagonists & inhibitors , Pancreatic Neoplasms/therapy , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Biopsy, Fine-Needle , Cadherins/genetics , Cetuximab , Cohort Studies , Combined Modality Therapy , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Dose-Response Relationship, Drug , Epithelial-Mesenchymal Transition , Feasibility Studies , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Pancreatic Neoplasms/pathology , Survival Rate , Vimentin/genetics , Gemcitabine
5.
Cancer Gene Ther ; 16(10): 794-805, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19363468

ABSTRACT

Malignant forms of glioma, the most common primary brain tumors, remain poorly responsive to multimodality therapeutic interventions, including chemotherapy. Suppressed apoptosis and extraordinary invasiveness are important distinctive features that contribute to the malignant phenotype of glioma. We have developed the vascular endothelial growth factor receptor 1 (VEGFR-1/flt-1) conditional replicating adenoviral vector (CRAdRGDflt-IL24) encoding the interleukin-24 (IL-24) gene. We investigated whether a combination of CRAdRGDflt-IL24-mediated oncolytic virotherapy and chemotherapy using temozolomide (TMZ) produces increased cytotoxicity against human glioma cells in comparison with these agents alone. Combination of CRAdRGDflt-IL24 and TMZ significantly enhanced cytotoxicity in vitro, inhibited D54MG tumor growth and prolonged survival of mice harboring intracranial human glioma xenografts in comparison with CRAdRGDflt-IL24 or TMZ alone. These data indicate that combined treatment with CRAdRGDflt-IL24-mediated oncolytic virotherapy and TMZ chemotherapy provides a promising approach for glioma therapy.


Subject(s)
Brain Neoplasms/therapy , Dacarbazine/analogs & derivatives , Glioma/therapy , Interleukins/genetics , Oncolytic Virotherapy/methods , Vascular Endothelial Growth Factor Receptor-1/genetics , Adenoviridae/genetics , Animals , Antineoplastic Agents, Alkylating/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/virology , Cell Growth Processes/genetics , Cell Line, Tumor , Combined Modality Therapy , Dacarbazine/pharmacology , Female , Genetic Therapy/methods , Genetic Vectors/genetics , Glioma/drug therapy , Glioma/genetics , Glioma/virology , Humans , Mice , Mice, Nude , Promoter Regions, Genetic , Recombinant Proteins/pharmacology , Temozolomide , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor Receptor-1/biosynthesis , Xenograft Model Antitumor Assays
6.
Cancer Gene Ther ; 16(3): 246-55, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18846112

ABSTRACT

Recent studies have demonstrated the efficacy of targeted therapy combined with radiotherapy in head and neck squamous cell carcinoma (HNSCC). We hypothesized that a combination treatment including a replicating adenovirus armed with tissue inhibitor of metalloproteinase-2 (TIMP-2), radiation and Cisplatin will augment treatment response and reduce tumor growth in vivo of HNSCC xenografts. Both single-agent (TIMP-2 virus, radiation and Cisplatin) and the combination therapies were evaluated in vitro and in vivo. The efficacy of both single-agent and combination therapies in vivo was determined by monitoring tumor growth and immunohistochemistry. Treatment with replicative Ad-TIMP-2 virus and radiation decreased cell viability in vitro and resulted in an additional antiangiogenic response in vivo. Tumor response rates to treatment with replicative Ad-TIMP-2, radiation, Cisplatin or combination therapies ranged from limited inhibition of tumor growth of the single-agent therapy to a statistically significant additive antitumor response with the combination therapies. Replicative Ad-TIMP-2+radiation+Cisplatin in the SCC1 nude mice demonstrated the greatest response rates in tumor growth and angiogenesis. Combination of Ad-TIMP-2 gene therapy with radiation and the triple treatment group resulted in an augmented therapeutic response. This is the first report of the potential benefits of combining radiation and MMP inhibitor treatment.


Subject(s)
Adenoviruses, Human/genetics , Antineoplastic Agents, Alkylating/therapeutic use , Carcinoma, Squamous Cell/therapy , Cisplatin/therapeutic use , Genetic Vectors/therapeutic use , Head and Neck Neoplasms/therapy , Oncolytic Virotherapy , Tissue Inhibitor of Metalloproteinase-2/physiology , Adenoviruses, Human/physiology , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Carcinoma, Squamous Cell/blood supply , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/radiotherapy , Cell Line, Tumor/transplantation , Cell Line, Tumor/virology , Combined Modality Therapy , Female , Genetic Vectors/genetics , Head and Neck Neoplasms/blood supply , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/radiotherapy , Humans , Mice , Mice, Nude , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/radiotherapy , Neovascularization, Pathologic/therapy , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/physiology , Tissue Inhibitor of Metalloproteinase-2/genetics , Virus Replication , Xenograft Model Antitumor Assays
7.
Gut ; 57(5): 634-41, 2008 May.
Article in English | MEDLINE | ID: mdl-18222985

ABSTRACT

OBJECTIVE: Investigators are currently interested in the epidermal growth factor receptor (EGFR) and interleukin 13 receptor (IL13R) as potential targets in the development of new biologicals for pancreatic cancer. Attempts to develop successful agents have met with difficulty. The novel approach used here was to target these receptors simultaneously with EGF and IL13 cloned on the same bispecific single-chain molecule with truncated diphtheria toxin (DT(390)) to determine if co-targeting with DTEGF13 had any advantages. DESIGN: Proliferation experiments were performed to measure the potency and selectivity of bispecific DTEGF13 and its monospecific counterparts against pancreatic cancer cell lines PANC-1 and MiaPaCa-2 in vitro. DTEGF13 was then administered intratumourally to nude mice with MiaPaCa-2 flank tumours to measure efficacy and toxicity (weight loss). RESULTS: In vitro, bispecific DTEGF13 was 2800-fold more toxic than monospecific DTEGF or DTIL13 against PANC-1. A similar enhancement was observed in vitro when MiaPaCa-2 pancreatic cancer cells or H2981-T3 lung adenocarcinoma cells were studied. DTEGF13 activity was blockable with recombinant EGF13. DTEGF13 was potent (IC(50) = 0.00017 nM) against MiaPaCa-2, receptor specific and significantly inhibited MiaPaCa-2 tumours in nude mice (p<0.008). CONCLUSIONS: In vitro studies show that the presence of both ligands on the same bispecific molecule is responsible for the superior activity of DTEGF13. Intratumoural administration showed that DTEGF13 was highly effective in checking aggressive tumour progression in mice. Lack of weight loss in these mice indicated that the drug was tolerated and a therapeutic index exists in an "on target" model in which DTEGF13 is cross-reactive with native mouse receptors.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/pharmacology , ErbB Receptors/antagonists & inhibitors , Pancreatic Neoplasms/drug therapy , Receptors, Interleukin-13/antagonists & inhibitors , Recombinant Fusion Proteins/pharmacology , Adenocarcinoma/pathology , Animals , Antineoplastic Agents/adverse effects , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Progression , Male , Mice , Mice, Nude , Neoplasm Transplantation/methods , Pancreatic Neoplasms/pathology , Random Allocation , Recombinant Fusion Proteins/adverse effects , Recombinant Fusion Proteins/chemistry , Transplantation, Heterologous , Treatment Outcome
8.
Gene Ther ; 14(14): 1111-9, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17495948

ABSTRACT

Combined treatment using adenoviral (Ad)-directed enzyme/prodrug therapy and radiation therapy has the potential to become a powerful method of cancer therapy. We have developed an Ad vector encoding a mutant bacterial cytosine deaminase (bCD) gene (AdbCD-D314A), which has a higher affinity for cytosine than wild-type bCD (bCDwt). The purpose of this study was to evaluate cytotoxicity in vitro and therapeutic efficacy in vivo of the combination of AdbCD-D314A with the prodrug 5-fluorocytosine (5-FC) and ionizing radiation against human glioma. The present study demonstrates that AdbCD-D314A infection resulted in increased 5-FC-mediated cell killing, compared with AdbCDwt. Furthermore, a significant increase in cytotoxicity following AdbCD-D314A and radiation treatment of glioma cells in vitro was demonstrated as compared to AdbCDwt. Animal studies showed significant inhibition of subcutaneous or intracranial tumor growth of D54MG glioma xenografts by the combination of AdbCD-D314A/5-FC with ionizing radiation as compared with either agent alone, and with AdbCDwt/5-FC plus radiation. The results suggest that the combination of AdbCD-D314A/5-FC with radiation produces markedly increased cytotoxic effects in cancer cells in vitro and in vivo. These data indicate that combined treatment with this novel mutant enzyme/prodrug therapy and radiotherapy provides a promising approach for cancer therapy.


Subject(s)
Adenoviridae/genetics , Brain Neoplasms/therapy , Cytosine Deaminase/genetics , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Glioma/therapy , Animals , Antimetabolites/therapeutic use , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/metabolism , Cell Line, Tumor , Combined Modality Therapy , Cytosine/metabolism , Cytosine Deaminase/metabolism , Escherichia coli/enzymology , Flucytosine/therapeutic use , Genes, Transgenic, Suicide , Genetic Vectors/genetics , Glioma/diagnostic imaging , Glioma/metabolism , Humans , Mice , Mice, Nude , Mutation , Neoplasm Transplantation , Prodrugs/therapeutic use , Radiography , Transplantation, Heterologous
9.
Cancer Gene Ther ; 13(2): 203-14, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16082379

ABSTRACT

Combined treatment using adenoviral-directed enzyme/prodrug therapy and immunotherapy has the potential to become a powerful alternative method of cancer therapy. We have developed adenoviral vectors encoding the cytosine deaminase gene (Ad-CD) and cytosine deaminase:uracil phosphoribosyltransferase fusion gene (Ad-CD:UPRT). A monoclonal antibody, TRA-8, specifically binds to death receptor 5, one of two death receptors bound by tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). The purpose of this study was to evaluate cytotoxicity in vitro and therapeutic efficacy in vivo of the combination of Ad-CD:UPRT and TRA-8 against human pancreatic cancer and glioma cell lines. The present study demonstrates that Ad-CD:UPRT infection resulted in increased 5-FC-mediated cell killing, compared with Ad-CD. Furthermore, a significant increase of cytotoxicity following Ad-CD:UPRT/5-FC and TRA-8 treatment of cancer cells in vitro was demonstrated. Animal studies showed significant inhibition of tumor growth of MIA PaCa-2 pancreatic and D54MG glioma xenografts by the combination of Ad-CD:UPRT/5-FC plus TRA-8 as compared with either agent alone or no treatment. The results suggest that the combination of Ad-CD:UPRT/5-FC with TRA-8 produces an additive cytotoxic effect in cancer cells in vitro and in vivo. These data indicate that combined treatment with enzyme/prodrug therapy and TRAIL immunotherapy provides a promising approach for cancer therapy.


Subject(s)
Adenoviridae/genetics , Antibodies, Monoclonal/therapeutic use , Cytosine Deaminase/genetics , Genes, Transgenic, Suicide/genetics , Genetic Therapy/methods , Glioma/therapy , Immunotherapy/methods , Pancreatic Neoplasms/therapy , Analysis of Variance , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Blotting, Western , Cell Line, Tumor , Cytotoxicity Tests, Immunologic , Female , Flow Cytometry , Glioma/immunology , Humans , Mice , Mice, Nude , Microscopy, Fluorescence , Pancreatic Neoplasms/immunology , Pentosyltransferases/genetics , Prodrugs/therapeutic use , Receptors, TNF-Related Apoptosis-Inducing Ligand , Receptors, Tumor Necrosis Factor/metabolism
10.
Gene Ther ; 12(5): 407-17, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15616600

ABSTRACT

Overexpression of vascular endothelial growth factor (VEGF) and its cognate receptor KDR has been linked to a more aggressive phenotype of human prostate carcinomas. The importance of signal transduction through the VEGF receptor 2 is illustrated by use of soluble KDR, which binds to VEGF and sequesters this ligand before its binding to cellular receptor. Treatment with recombinant adenovirus AdVEGF-sKDR, encoding sKDR under control of the human VEGF promoter, significantly inhibited the proliferation of human vascular endothelial cells and prostate cancer cells. AdVEGF-sKDR infection decreased migration of endothelial 1P-1B cells (61% reduction) and DU145 prostate carcinoma cells (47%) in comparison with AdCMV-Luc-infected control cells. Ionizing radiation upregulated VEGF promoter activity in prostate carcinoma and endothelial cells. AdVEGF-sKDR infection significantly reduced human vascular endothelial and prostate cancer cell proliferation and sensitized cancer cells to ionizing radiation. In vivo tumor therapy studies demonstrated significant inhibition of DU145 tumor growth in mice that received combined AdVEGF-sKDR infection and ionizing radiation versus AdVEGF-sKDR alone or radiation therapy alone. These results suggest that selective transcriptional targeting of sKDR gene expression employing a radiation inducible promoter can effectively inhibit tumor growth and demonstrate the advantage of combination radiotherapy and gene therapy for the treatment of prostate cancer.


Subject(s)
Adenoviridae/genetics , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Prostatic Neoplasms/therapy , Vascular Endothelial Growth Factor Receptor-2/genetics , Animals , Cell Proliferation , Combined Modality Therapy , Endothelium, Vascular/pathology , Gene Expression Regulation/radiation effects , Genetic Vectors/genetics , Humans , Male , Mice , Mice, Nude , Promoter Regions, Genetic , Prostatic Neoplasms/pathology , Prostatic Neoplasms/radiotherapy , Recombinant Proteins/metabolism , Transduction, Genetic/methods , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A/metabolism
11.
Gene Ther ; 11(8): 658-67, 2004 Apr.
Article in English | MEDLINE | ID: mdl-14973547

ABSTRACT

Specific activation of apoptosis in tumor cells offers a promising approach for cancer therapy. Induction of apoptosis leads to activation of specific proteases. Two major pathways for caspase activation in mammalian cells have been described. One apoptotic pathway involves members of the tumor necrosis factor family of cytokine receptors (eg death receptor 5 (DR5)). The other pathway is controlled by the Bcl-2 family of proteins. The purpose of this study was to investigate whether increased apoptosis occurs in human glioma cells following infection with a recombinant adenoviral vector encoding the human Bax gene under the control of human vascular endothelial growth factor (VEGF) promoter element (AdVEGFBax) in combination with an anti-human DR5 monoclonal antibody (TRA-8). Specific overexpression of exogenous Bax protein induced apoptosis and cell death in glioma cell lines, through activation of both caspase-8 and -9, leading to activation of downstream caspase-3. The relative sensitivity to AdVEGFBax for the glioma cell lines was U251MG>U373MG>U87MG>D54MG. The recently characterized TRA-8 monoclonal antibody induces apoptosis of most TRAIL-sensitive tumor cells by specific binding to DR5 receptors on the cellular membrane. TRA-8 induced rapid apoptosis and cell death in glioma cells, but did not demonstrate detectable cytotoxicity of primary normal human astrocytes. The efficiency of TRA-8-induced apoptosis was variable in different glioma cell lines. The relative sensitivity to TRA-8 was U373MG>U87MG>U251MG>D54MG. The combination of TRA-8 treatment and overexpression of Bax overcame TRA-8 resistance of glioma cells in vitro. Cell viability of U251MG cells was 71.1% for TRA-8 (100 ng/ml) alone, 75.9% for AdVEGFBax (5 MOI) alone and 41.1% for their combination as measured by MTS assay. Similar enhanced apoptosis results were obtained for the other glioma cell lines. In vivo studies demonstrated that the combined treatment significantly (P<0.05) suppressed the growth of U251MG xenografts and produced 60% complete tumor regressions without recurrence. These data suggest that the combination of TRA-8 treatment with specific overexpression of Bax using AdVEGFBax may be an effective approach for the treatment of human malignant gliomas.


Subject(s)
Adenoviridae/genetics , Brain Neoplasms/therapy , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Glioma/therapy , Proto-Oncogene Proteins c-bcl-2 , Proto-Oncogene Proteins/genetics , Animals , Antibodies, Monoclonal/pharmacology , Apoptosis , Blotting, Western/methods , Brain Neoplasms/metabolism , Caspases/metabolism , Cell Death , Cell Line, Tumor , Combined Modality Therapy , DNA Fragmentation , Female , Flow Cytometry , Gene Expression , Glioma/metabolism , Humans , Mice , Mice, Nude , Promoter Regions, Genetic , Receptors, TNF-Related Apoptosis-Inducing Ligand , Receptors, Tumor Necrosis Factor/analysis , Receptors, Tumor Necrosis Factor/immunology , Vascular Endothelial Growth Factor A/analysis , Vascular Endothelial Growth Factor A/genetics , bcl-2-Associated X Protein
12.
Gene Ther ; 9(4): 256-62, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11896464

ABSTRACT

Single chain antibodies (scFv) represent powerful interventional agents for the achievement of targeted therapeutics. The practical utility of these agents have been limited, however, by difficulties related to production of recombinant scFv and the achievement of effective and sustained levels of scFv in situ. To circumvent these limitations, we have developed an approach to express scFv in vivo. An anti-erbB2 scFv was engineered for secretion by eukaryotic cells. The secreted scFv could bind to its target and specifically suppress cell growth of erbB2-positive cells in vitro. Adenoviral vectors expressing the cDNA for the secretory scFv likewise could induce target cells to produce an anti-tumor anti-erbB2 scFv. In vivo gene transfer via the anti-erbB2 scFv encoding adenovirus also showed anti-tumor effects. Thus, by virtue of engineering a secreted version of the anti-tumor anti-erbB-2 scFv, and in vivo expression via adenoviral vector, effective concentrations of scFv were achieved. In vivo gene transfer clearly represents a powerful means to realize effective scFv-based approaches. This method will likely have applicability for a range of disorders amenable to targeted therapeutic approaches.


Subject(s)
Adenoviridae/genetics , Antibodies, Monoclonal/genetics , Genetic Therapy/methods , Genetic Vectors , Receptor, ErbB-2/immunology , Animals , Antibodies, Monoclonal/blood , Female , Gene Targeting/methods , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Ovarian Neoplasms/prevention & control , Transfection , Tumor Cells, Cultured
13.
Cancer Biother Radiopharm ; 16(4): 275-88, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11602998

ABSTRACT

The delineation of the molecular basis of neoplasia provides the possibility of specific intervention by gene therapy through the introduction of genetic material for therapeutic purposes. In this regard, several gene therapy approaches have been developed for the treatment of cancer: mutation compensation, genetic immunopotentiation, molecular chemotherapy, inhibition of angiogenesis, replicative vector oncolysis, and chemosensitization or radiosensitization. Clinical trials have been initiated to evaluate safety, toxicity, and efficacy of each of these approaches, based on promising preclinical results. Various limitations that have been identified include lack of in vivo selective tumor delivery of vectors, minimal expression of therapeutic genes, immune response against vectors, and normal tissue toxicity. Combined modality therapy with gene therapy and chemotherapy or radiation therapy has shown promising results. It is expected that as new therapeutic targets and approaches are identified, combined with advances in vector design, that gene therapy will play an increasing role in clinical cancer treatment.


Subject(s)
Genetic Therapy/methods , Neoplasms/therapy , Animals , Clinical Trials as Topic , Genetic Vectors , Humans
14.
Gynecol Oncol ; 83(2): 432-8, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11606113

ABSTRACT

OBJECTIVE: Gene therapy trials for ovarian cancer would benefit from a noninvasive imaging modality to detect the location and extent of gene transfer. The human type 2 somatostatin receptor gene (hSSTr2) was evaluated as a reporter gene for imaging adenoviral (Ad) gene transfer to ovarian cancer. METHODS: A replication-incompetent Ad vector encoding hSSTr2 (Ad-hSSTr2) was used to infect SKOV3.ip1 cells in vitro and tumors growing in nude mice. Gamma camera imaging detected uptake of 99m-Tc-P2045 (a somatostatin analogue) due to expressed hSSTr2. RESULTS: Specific uptake of 99m-Tc-P2045 was imaged in Ad-hSSTr2-infected cells in vitro. Noninvasive in vivo imaging detected gene transfer to intraperitoneal tumors. Uptake of 99m-Tc-P2045 (percentage dose per gram of tumor) averaged 2.2 and 0.18 for Ad-hSSTr2-injected mice and controls, respectively. CONCLUSION: This study reports the first noninvasive imaging method for imaging gene transfer to ovarian cancer. A human gene therapy trial is planned.


Subject(s)
Gene Transfer Techniques , Genetic Vectors/genetics , Ovarian Neoplasms/genetics , Receptors, Somatostatin/genetics , Adenoviridae/genetics , Animals , Female , Genes, Reporter , Genetic Vectors/biosynthesis , Humans , Mice , Mice, Nude , Organotechnetium Compounds/pharmacokinetics , Ovarian Neoplasms/diagnostic imaging , Ovarian Neoplasms/metabolism , Radionuclide Imaging , Radiopharmaceuticals/pharmacokinetics , Receptors, Somatostatin/biosynthesis , Xenograft Model Antitumor Assays
15.
Mol Ther ; 4(3): 223-31, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11545613

ABSTRACT

The adenovirus (Ad) is a useful vector for cancer gene therapy due to its unparalleled gene transfer efficiency to dividing and quiescent cells. Primary cancer cells, however, often have highly variable or low levels of the requisite coxsackie-adenovirus receptor (CAR). Also, assessment of gene transfer and vector persistence has been logistically difficult in human clinical trials. We describe here two novel bicistronic adenoviral (Ad) vectors, AdTKSSTR and RGDTKSSTR, which contain the herpes simplex virus thymidine kinase gene (TK) for molecular chemotherapy and bystander effect. In addition, the viruses contain the human somatostatin receptor subtype-2 gene (SSTR2), the expression of which can be noninvasively imaged. We enhanced the infectivity of RGDTKSSTR by genetically incorporating the RGD-4C motif into the HI-loop of the fiber. This allows the virus to circumvent CAR deficiency by binding to alpha(v)beta(3) and alpha(v)beta(5) integrins, which are highly expressed on most ovarian cancers. The expanded tropism of RGDTKSSTR results in increased infectivity of purified primary ovarian cancer cells and allows enhanced gene transfer in the presence of malignant ascites containing anti-Ad antibodies. RGDTKSSTR may be a useful agent for treating ovarian cancer in clinical trials.


Subject(s)
Adenoviridae/genetics , Adenoviridae/physiology , Diagnostic Imaging/methods , Gene Expression , Genetic Therapy/methods , Ovarian Neoplasms/genetics , Ovarian Neoplasms/therapy , Ascites/genetics , Ascites/metabolism , Ascites/pathology , Ascites/virology , Cell Survival/drug effects , Coxsackie and Adenovirus Receptor-Like Membrane Protein , DNA, Recombinant/genetics , Female , Ganciclovir/pharmacology , Genetic Vectors/genetics , Gentian Violet , HeLa Cells , Humans , Mutagenesis, Insertional , Ovarian Neoplasms/pathology , Ovarian Neoplasms/virology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Somatostatin/genetics , Receptors, Virus/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Thymidine Kinase/genetics , Tumor Cells, Cultured
16.
Gene Ther ; 8(4): 291-9, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11313803

ABSTRACT

The purpose of this work was to develop a high capacity method to image gene transfer to cancer cells growing as monolayers in cell culture plates. A sensitive and high capacity nuclear-imaging method for detection of gene transfer in vitro will allow rapid validation of vectors in different cell lines under various conditions. Human cancer cell lines (A-427 non-small cell lung, SKOV3.ip1 ovarian, MDA-MB-468 breast, and BxPC-3 pancreatic) were infected with a replication-incompetent adenoviral vector encoding the human type 2 somatostatin receptor (Ad-hSSTr2). Expression of the hSSTr2 reporter protein in cells was detected by imaging an internalized 99mTc-labeled, hSSTr2 binding peptide (P2045, Diatide, Inc.). Imaging provided an accurate measure of internally bound 99mTc as evidenced by equivalence of results for imaging region of interest (ROI) analyses and gamma counter measurements. Internally bound 99mTc-P2045 was linearly correlated (R2 = 0.98) with the percentage of hSSTr2-positive cells following gene transfer. Excess P2045 blocked binding and internalization of the 99mTc-P2045, indicating the specificity of the technique. Up to four 96-well plates could be imaged simultaneously, thereby demonstrating the high capacity of the system. This novel in vitro approach provides a new method to test enhanced gene transfer as new vectors are developed.


Subject(s)
Adenoviridae/genetics , Genetic Vectors/administration & dosage , Receptors, Somatostatin/genetics , Transfection/methods , Breast Neoplasms/therapy , Carcinoma, Non-Small-Cell Lung/therapy , Female , Genetic Therapy/methods , Humans , Lung Neoplasms/therapy , Ovarian Neoplasms/therapy , Pancreatic Neoplasms/therapy , Radionuclide Imaging , Technetium , Tumor Cells, Cultured/diagnostic imaging
17.
Nucl Med Biol ; 28(2): 135-44, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11295424

ABSTRACT

A gamma camera imaging method was developed to detect dual gene transfer to adherent cells growing as monolayers in cell culture plates. Human cancer cells were infected with replication-incompetent adenoviral vectors encoding the human type 2 somatostatin receptor (Ad-hSSTr2) and/or herpes virus thymidine kinase (Ad-TK). The hSSTr2 and TK reporter proteins were detected by imaging internally bound (99m)Tc-P2045 peptide (Diatide, Inc.) and radioiodinated 2'-deoxy-2'-fluoro-beta-D-arabinofuranosyl-5-iodouracil (FIAU), respectively. Following gene transfer, expression of hSSTr2 and TK were accurately imaged in vitro.


Subject(s)
Arabinofuranosyluracil/analogs & derivatives , Receptors, Somatostatin/genetics , Thymidine Kinase/genetics , Transfection/methods , Adenoviridae , Arabinofuranosyluracil/pharmacokinetics , Carcinoma, Non-Small-Cell Lung , Cell Adhesion , Cell Line , Gamma Cameras , Gamma Rays , Genetic Vectors , Herpesviridae/genetics , Humans , Iodine Radioisotopes/pharmacokinetics , Kidney , Lung Neoplasms , Radiopharmaceuticals , Technetium/pharmacokinetics , Tumor Cells, Cultured
18.
Clin Cancer Res ; 6(11): 4323-33, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11106250

ABSTRACT

Efficient gene transfer by recombinant adenovirus (Ad) vectors depends on expression of CAR and alpha(v) integrin on target cells. Because Ad may also infect nearby nontarget cells expressing these receptors, such as peritoneal mesothelial cells after i.p. injection, we hypothesized that targeting Ad gene delivery to a receptor overexpressed on most ovarian carcinoma cells, such as TAG-72, would enhance the selectivity of Ad gene transfer when used in this context. A monoclonal antibody that has been investigated clinically for immunotherapy and immunodetection of ovarian carcinomas, namely CC49, was used to construct a bispecific conjugate with the Fab fragment of a neutralizing anti-knob mAb to target Ad binding via TAG-72. This conjugate facilitated TAG-72-specific, CAR-independent Ad reporter gene transfer to both ovarian cancer cell lines and primary ovarian cancer cells cultured from malignant ascites fluid. Fab-CC49 was very selective for tumor cells, augmenting Ad gene transfer to primary ovarian cancer cells 2- to 28-fold relative to untargeted Ad, while also decreasing gene transfer to autologous cultured mesothelial cells 4- to 9-fold. These data suggest that targeting Ad via TAG-72 may improve the selectivity of Ad gene transfer for ovarian tumors 8- to 252-fold on i.p. vector injection. These results also define the requirements for a candidate target receptor in the rational design of a targeted Ad vector for ultimate clinical utility, one that selectively infects tumor cells and spares normal cells on i.p. injection. Such a vector may increase gene transfer and decrease the toxicity of Ad vectors, which would improve the therapeutic index of cytotoxic gene therapy for ovarian cancer in clinical trials.


Subject(s)
Adenoviridae/genetics , Antigens, Neoplasm/genetics , Genetic Therapy , Glycoproteins/genetics , Ovarian Neoplasms/therapy , Antigens, Neoplasm/biosynthesis , Cells, Cultured , Epithelial Cells/metabolism , Female , Gene Transfer, Horizontal , Glycoproteins/biosynthesis , Humans , Immunoglobulin Fab Fragments/administration & dosage , Integrins/analysis , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Receptors, Virus/analysis , Receptors, Vitronectin/analysis
19.
Q J Nucl Med ; 44(3): 208-23, 2000 Sep.
Article in English | MEDLINE | ID: mdl-11105586

ABSTRACT

Utilization of molecular biology techniques offers attractive options in nuclear medicine for improving cancer imaging and therapy with radiolabeled peptides. Two of these options include utilization of phage-panning to identify novel tumor-specific peptides or single chain antibodies and gene transfer techniques to increase the number of antigen/receptor sites expressed on malignant cells. Our group has focused on the latter approach for improving radiolabeled peptide imaging and therapy. The most widely used gene transfer vectors in clinical gene therapy trials include retrovirus, cationic lipids, and adenovirus. We have utilized adenovirus vectors for gene transfer because of their ability to accomplish efficient in vivo gene transfer. Adenovirus vectors encoding the genes for a variety of antigens/receptors (carcinoembryonic antigen, gastrin-releasing peptide receptor, somatostatin receptor subtype 2 (SSTr2)) have all shown that their expression is increased on cancer cells both in vitro and in vivo following adenovirus infection. Of particular interest has been the adenovirus encoding for SSTr2 (AdCMVSSTr2). Various radioisotopes have been attached to somatostatin analogues for imaging and therapy of SSTr2-positive tumors both clinically and in animal models. The use of these analogues in combination with AdCMVSSTr2 is a promising approach for improving the detection sensitivity and therapeutic efficacy of these radiolabeled peptides against solid tumors. In addition, we have proposed the use of SSTr2 as a marker for imaging the expression of another cancer therapeutic transgene (e.g. cytosine deaminase, thymidine kinase) encoded within the same vector. This would allow for non-invasive monitoring of gene delivery to tumor sites.


Subject(s)
Gene Transfer Techniques , Genetic Therapy , Peptides/therapeutic use , Radiopharmaceuticals/therapeutic use , Adenoviridae , Animals , Genetic Vectors , Humans , Neoplasms/diagnostic imaging , Neoplasms/genetics , Neoplasms/therapy , Radionuclide Imaging , Receptors, Somatostatin/genetics , Receptors, Somatostatin/metabolism , Somatostatin/analogs & derivatives
20.
Mol Ther ; 2(6): 562-78, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11124057

ABSTRACT

Adenoviral (Ad) vectors are promising gene therapy vehicles due to their in vivo stability and efficiency, but their potential utility is compromised by their restricted tropism. Targeting strategies have been devised to improve the efficacy of these agents, but specific targeting following in vivo systemic administration of vector has not previously been demonstrated. The distinct aim of the current study was to determine whether an Ad-targeting strategy could maintain fidelity upon systemic vascular administration. We used a bispecific antibody to target Ad infection specifically to angiotensin-converting enzyme (ACE), which is preferentially expressed on pulmonary capillary endothelium and which may thus enable gene therapy for pulmonary vascular disease. Cell-specific gene delivery to ACE-expressing cells was first confirmed in vitro. Administration of retargeted vector complex via tail vein injection into rats resulted in at least a 20-fold increase in both Ad DNA localization and luciferase transgene expression in the lungs, compared to the untargeted vector. Furthermore, targeting led to reduced transgene expression in nontarget organs, especially the liver, where the reduction was over 80%. Immunohistochemical and immunoelectron microscopy analysis confirmed that the pulmonary transgene expression was specifically localized to endothelial cells. Enhancement of transgene expression in the lungs as a result of the ACE-targeting strategy was also confirmed using a new noninvasive imaging technique. This study shows that a retargeting approach can indeed specifically modify the gene delivery properties of an Ad vector given systemically and thus has encouraging implications for the further development of targetable, injectable Ad vectors.


Subject(s)
Adenoviridae/genetics , Genetic Vectors , Lung/metabolism , 3T3 Cells , Animals , Base Sequence , CHO Cells , Cricetinae , DNA Primers , Endothelium/enzymology , Endothelium/metabolism , Endothelium/ultrastructure , Immunohistochemistry , Lung/enzymology , Lung/ultrastructure , Mice , Microscopy, Electron , Peptidyl-Dipeptidase A/genetics , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...