Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 245
Filter
Add more filters

Publication year range
1.
Vestn Ross Akad Med Nauk ; (12): 4-8, 2013.
Article in Russian | MEDLINE | ID: mdl-24741936

ABSTRACT

UNLABELLED: Currently one of the most promising approaches in development of cancer virotherapy is based on the ability of oncolytic viruses to selective infection and lysis of tumor cells. AIM: The goal of the study was to identify and evaluate perspective oncolytic viruses capable of selectively destroying human glioma cells. PATIENTS AND METHODS: Original GB2m, GA14m and GB22m glioma cell cultures derived from patients were used for evaluating in vitro oncolytic activity of some typical orthopoxviruses, adenoviruses and parvoviruses. RESULTS: The oncolytic activity in the human glioma cell models was confirmed for LIVP and WR strains of vaccinia virus, Adel2 and Ad2del strains with deletions within E1B/55K gene and derived from human adenoviruses type 2 and 5, respectively. CONCLUSIONS: We consider these oncolytic viruses as promising agents for the treatment of human malignant glioma.


Subject(s)
Glioma , Oncolytic Virotherapy/methods , Oncolytic Viruses/physiology , Adenoviridae/physiology , Cell Culture Techniques , Glioma/therapy , Glioma/virology , Humans , Orthopoxvirus/physiology , Parvovirus/physiology , Tumor Cells, Cultured/virology , Virus Physiological Phenomena
2.
Nat Med ; 6(10): 1128-33, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11017144

ABSTRACT

The adenovirus mutant dl1520 (ONYX-015) does not express the E1B-55K protein that binds and inactivates p53. This virus replicates in tumor cells with mutant p53, but not in normal cells with functional p53. Although intra-tumoral injection of dl1520 shows promising responses in patients with solid tumors, previous in vitro studies have not established a close correlation between p53 status and dl1520 replication. Here we identify loss of p14ARF as a mechanism that allows dl1520 replication in tumor cells retaining wild-type p53. We demonstrate that the re-introduction of p14ARF into tumor cells with wild-type p53 suppresses replication of dl1520 in a p53-dependent manner. Our study supports the therapeutic use of dl1520 in tumors with lesions within the p53 pathway other than mutation of p53.


Subject(s)
Adenoviridae/genetics , Mutation , Nuclear Proteins , Proteins/genetics , Virus Replication , Gene Expression Regulation, Neoplastic , Humans , Proteins/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-mdm2 , Tumor Cells, Cultured/virology , Tumor Suppressor Protein p14ARF , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
3.
Nat Med ; 6(7): 821-5, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10888934

ABSTRACT

Interferons are circulating factors that bind to cell surface receptors, activating a signaling cascade, ultimately leading to both an antiviral response and an induction of growth inhibitory and/or apoptotic signals in normal and tumor cells. Attempts to exploit the ability of interferons to limit the growth of tumors in patients has met with limited results because of cancer-specific mutations of gene products in the interferon pathway. Although interferon-non-responsive cancer cells may have acquired a growth/survival advantage over their normal counterparts, they may have simultaneously compromised their antiviral response. To test this, we used vesicular stomatitis virus (VSV), an enveloped, negative-sense RNA virus exquisitely sensitive to treatment with interferon. VSV rapidly replicated in and selectively killed a variety of human tumor cell lines even in the presence of doses of interferon that completely protected normal human primary cell cultures. A single intratumoral injection of VSV was effective in reducing the tumor burden of nude mice bearing subcutaneous human melanoma xenografts. Our results support the use of VSV as a replication-competent oncolytic virus and demonstrate a new strategy for the treatment of interferon non-responsive tumors.


Subject(s)
Cytopathogenic Effect, Viral , Neoplasms, Experimental/therapy , Neoplasms, Experimental/virology , Vesicular stomatitis Indiana virus/pathogenicity , Animals , Bone Marrow/virology , Humans , Leukemia, Myeloid, Acute/virology , Melanoma, Experimental/therapy , Melanoma, Experimental/virology , Mice , Mice, Nude , Neoplasm Transplantation , Tumor Cells, Cultured/virology , Vesicular stomatitis Indiana virus/drug effects
4.
J Exp Med ; 181(4): 1575-80, 1995 Apr 01.
Article in English | MEDLINE | ID: mdl-7699338

ABSTRACT

HTLV-1 is implicated in the development of diverse diseases. However, most HTLV-1-infected individuals remain asymptomatic. How HTLV-1 infection leads to disparate consequences remains a mystery, despite extensive investigation of HTLV-1 isolates from infected individuals. As in human infection, experimental HTLV-1 infection in rabbits is generally benign, although HTLV-1-infected rabbit T cell lines that mediate lethal leukemia-like disease have been reported. We report here that thymuses from mature outbred rabbits inoculated with a lethal leukemia-like disease have been reported. We report here that thymuses from mature outbred rabbits inoculated with a lethal HTLV-1 T cell line (RH/K34) showed morphological and biochemical evidence of apoptosis, whereas thymuses from rabbits inoculated with nonlethal HTLV-1 T cell lines showed no signs of apoptosis. Exposure of rabbit or human lymphocytes to purified virus from RH/K34 caused rapid induction of apoptosis, providing an in vitro correlate to the pathogenic effects. By contrast, virus isolated from a nonlethal cell line mediated dose-dependent lymphocyte proliferation. These data implicate lymphocyte apoptosis as a potential mechanism by which the lethal HTLV-1 cell line causes fulminant disease and provide a means to identify factors contributing to HTLV-1 disease. Results from this HTLV-1 infection model can provide insight into variations in HTLV-1 pathogenicity in human infection.


Subject(s)
Apoptosis , Human T-lymphotropic virus 1/pathogenicity , Leukemia-Lymphoma, Adult T-Cell/virology , Lymphocytes/virology , Thymus Gland/pathology , Animals , Disease Models, Animal , Human T-lymphotropic virus 1/isolation & purification , Humans , Leukemia-Lymphoma, Adult T-Cell/pathology , Lymphocyte Activation , Lymphocytes/pathology , Rabbits , Thymus Gland/virology , Tumor Cells, Cultured/virology , Virulence
5.
J Am Assoc Lab Anim Sci ; 59(3): 328-333, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32079556

ABSTRACT

Lactate dehydrogenase elevating virus (LDV) continues to be one of the most common contaminants of cells and cell byproducts. As such, many institutions require that tumor cell lines, blood products, and products derived or passaged in rodent tissues are free of LDV as well as other pathogens that are on institutional exclusion lists prior to their use in rodents. LDV is difficult to detect by using a live-animal sentinel health monitoring program because the virus does not reliably pass to sentinel animals. After switching to an exhaust air dust health monitoring system, our animal resources center was able to detect a presumably long-standing LDV infection in a mouse colony. This health monitoring system uses IVC rack exhaust air dust collection media in conjunction with PCR analysis. Ultimately, the source of the contamination was identified as multiple LDV-positive patient-derived xenografts and multiple LDV-positive breeding animals. This case study is the first to demonstrate the use of environmental PCR testing as a method for detecting LDV infection in a mouse vivarium.


Subject(s)
Arterivirus Infections/veterinary , Environmental Microbiology , Housing, Animal , Lactate dehydrogenase-elevating virus/isolation & purification , Mice , Rodent Diseases/virology , Animals , Arterivirus Infections/virology , Cell Line, Tumor/virology , Dust , Heterografts , Humans , Polymerase Chain Reaction , Tumor Cells, Cultured/virology
6.
Adv Biosyst ; 4(2): e1900143, 2020 02.
Article in English | MEDLINE | ID: mdl-32293137

ABSTRACT

Replicable oncolytic viruses (OVs) induce tumor cell lysis and release viral progeny. The released progeny virions and cell debris can spread within surrounding tumor cells or blood vessels. These released molecules may also induce bystander damage in additional tumor cells through spreading within surrounding tumor cells or blood vessels. However, this effect has not been clearly demonstrated due to the difficulty of direct observation. Here, the bystander infection of OVs by vessel delivery and selective infection in 3D multicellular tumoroids (MCTs) in an in vitro microphysiological system (MPS) with integrated medium flow is demonstrated. This study uses replicable vesicular stomatitis virus (VSV)-green fluorescence protein (GFP) to identify the location of infection in 3D MCTs. Using this MPS, the oncoselective infection by VSV-GFP and the spreading by delivery of OVs through flow via block-to-block linkage of the primary infected MPS with uninfected 3D MCTs in an integrated MPS is observed. This MPS enables real-time monitoring and various analysis for the bystander infection of OVs. It is expected that the 3D in vitro MPS can be suitable to investigate the oncoselective spreading and bystander infection of OVs.


Subject(s)
Cytological Techniques , Models, Biological , Oncolytic Virotherapy/adverse effects , Oncolytic Viruses , A549 Cells , Cells, Cultured , Cytological Techniques/instrumentation , Cytological Techniques/methods , Equipment Design , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Oncolytic Viruses/genetics , Oncolytic Viruses/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Rhabdoviridae Infections/virology , Spheroids, Cellular/virology , Tumor Cells, Cultured/virology , Vesiculovirus/genetics
7.
Science ; 285(5424): 110-3, 1999 Jul 02.
Article in English | MEDLINE | ID: mdl-10390360

ABSTRACT

An estimated 170 million persons worldwide are infected with hepatitis C virus (HCV), a major cause of chronic liver disease. Despite increasing knowledge of genome structure and individual viral proteins, studies on virus replication and pathogenesis have been hampered by the lack of reliable and efficient cell culture systems. A full-length consensus genome was cloned from viral RNA isolated from an infected human liver and used to construct subgenomic selectable replicons. Upon transfection into a human hepatoma cell line, these RNAs were found to replicate to high levels, permitting metabolic radiolabeling of viral RNA and proteins. This work defines the structure of HCV replicons functional in cell culture and provides the basis for a long-sought cellular system that should allow detailed molecular studies of HCV and the development of antiviral drugs.


Subject(s)
Genome, Viral , Hepacivirus/physiology , RNA, Viral/biosynthesis , Replicon , Tumor Cells, Cultured/virology , Virus Replication , Carcinoma, Hepatocellular , Cloning, Molecular , Drug Resistance , Gentamicins/pharmacology , Hepacivirus/genetics , Hepatitis C/virology , Humans , Liver Neoplasms , RNA, Viral/genetics , Transfection , Viral Nonstructural Proteins/analysis , Viral Nonstructural Proteins/genetics , Virus Cultivation
8.
Clin Cancer Res ; 14(5): 1571-80, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-18316582

ABSTRACT

We have examined the spread and antitumor efficacy of an oncolytic herpes simplex virus-1-based vector (G207) in glioblastoma biopsy spheroids in vitro and in vivo after local delivery to corresponding intracranial xenografts. Spheroids from three patients were infected with increasing doses of G207 and transgene expression was quantified. Other infected spheroids were followed for 10 days to assess cytotoxic effects. For the in vivo study, spheroids were grafted intracerebrally into Rowett nude rats. The resulting highly infiltrative xenografts were injected with 3.4 x 10(6) plaque-forming units (penetration study) or 6.8 x 10(6) plaque-forming units (therapeutic study) of G207 using microprocessor-controlled stereotaxic delivery. Vector spread was tracked by histochemical staining. In the therapeutic study, tumor volumes were monitored weekly by magnetic resonance imaging, and survival data were collected. In vitro, lacZ expression was seen at the spheroid surfaces 24 h postinfection, whereas the spheroid cores were transgene positive after 96 h. Cytotoxic susceptibility varied between the patients, showing a 36% to 95% lysis 10 days postinfection. Local delivery of G207 into intracranial xenografts resulted in extensive vector spread throughout the lesions. In the therapeutic study, G207 application reduced tumor volumes compared with controls, but did not significantly improve survival of the animals. Histologic analysis revealed infection of host structures such as the ventricular and choroid plexus ependyma. In conclusion, G207 replicates in patient-derived glioblastoma multiforme xenografts and tumor volumes are reduced after intratumoral delivery; however, the survival data suggest that the therapeutic effect could be improved by repeated vector application or through combination with other treatment modalities.


Subject(s)
Disease Models, Animal , Glioblastoma/therapy , Herpesvirus 1, Human/physiology , Oncolytic Virotherapy , Virus Replication , Animals , Cell Survival , Cytopathogenic Effect, Viral , Genetic Vectors/administration & dosage , Glioblastoma/pathology , Glioblastoma/virology , Humans , Lac Operon , Magnetic Resonance Imaging , Rats , Rats, Nude , Spheroids, Cellular , Survival Rate , Tumor Cells, Cultured/virology
9.
Int J Cancer ; 122(1): 202-8, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-17764117

ABSTRACT

Oncolytic herpes simplex viruses (HSV) have demonstrated potent antitumoral effects against a variety of human malignancies in preclinical studies and are in early clinical trials. We explored the activity of an attenuated, replication-competent, oncolytic HSV (NV1023) for the treatment of human salivary gland carcinomas. NV1023 was able to successfully enter into 4 mucoepidermoid carcinoma (H292, H3118, HTB-41, UT-MUC-1) and 2 adenocarcinoma (HSY, HSG) cell lines, as measured by lacZ assays after exposure to 5 viral particles per cell (MOI 5). Viral plaque assays showed variation of viral replication within these cell lines, ranging from a 268-fold increase (H292) to a 3-fold increase (HSG) in viral titer. At MOI 5, all cell lines showed >95% cytotoxicity from NV1023 by Day 7, except for HSY (73%). At MOI 0.1, H3118 and UT-MUC-1 remained highly sensitive to NV1023, both showing >95% cytotoxicity by Day 7. The mucoepidermoid carcinomas were more sensitive to NV1023 at low viral concentrations compared with the adenocarcinomas. Flank tumors of H3118, HTB-41 and HSY in nude mice showed significant tumor volume reductions after a single intratumoral injection of NV1023 (2 x 10(7) plaque-forming units). These data suggest that oncolytic herpes viruses have significant efficacy entering, replicating within, and lysing human salivary gland carcinomas. These promising biologic agents should be further investigated as novel therapy for patients with salivary carcinomas failing conventional treatment.


Subject(s)
Adenocarcinoma, Follicular/therapy , Carcinoma, Mucoepidermoid/therapy , Herpesviridae Infections/therapy , Oncolytic Virotherapy , Salivary Gland Neoplasms/therapy , Simplexvirus/physiology , Adenocarcinoma, Follicular/pathology , Adenocarcinoma, Follicular/virology , Animals , Carcinoma, Mucoepidermoid/pathology , Carcinoma, Mucoepidermoid/virology , Cell Proliferation , Cell Survival/physiology , Herpesviridae Infections/pathology , Herpesviridae Infections/virology , Humans , Mice , Mice, Nude , Salivary Gland Neoplasms/pathology , Salivary Gland Neoplasms/virology , Tumor Cells, Cultured/metabolism , Tumor Cells, Cultured/virology , Virus Replication
10.
J Microbiol Biotechnol ; 28(1): 165-174, 2018 Jan 28.
Article in English | MEDLINE | ID: mdl-29032648

ABSTRACT

Glioblastoma multiforme is the most lethal malignant brain tumor. Despite many intensive studies, the prognosis of glioblastoma multiforme is currently very poor, with a median overall survival duration of 14 months and 2-year survival rates of less than 10%. Although viral infections have been emphasized as potential cofactors, their influences on pathways that support glioblastoma progression are not known. Some previous studies indicated that human Kaposi's sarcoma-associated herpesvirus (KSHV) was detected in healthy brains, and its microRNA was also detected in glioblastoma patients' plasma. However, a direct link between KSHV infection and glioblastoma is currently not known. In this study, we infected glioblastoma cells and glioma stem-like cells (GSCs) with KSHV to establish an in vitro cell model for KSHV-infected glioblastoma cells and glioma stem-like cells in order to identify virologic outcomes that overlap with markers of aggressive disease. Latently KSHV-infected glioblastoma cells and GSCs were successfully established. Additionally, using these cell models, we found that KSHV infection modulates the proliferation of glioma stem-like cells.


Subject(s)
Cell Proliferation , Glioma/virology , Herpesvirus 8, Human/growth & development , Stem Cells/physiology , Stem Cells/virology , Tumor Cells, Cultured/physiology , Tumor Cells, Cultured/virology , Cells, Cultured , Humans
11.
Mol Cell Biol ; 23(6): 2055-67, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12612078

ABSTRACT

An important step in the herpesvirus life cycle is the switch from latency to lytic reactivation. The RTA transcription activator of Kaposi's sarcoma-associated herpesvirus (KSHV) acts as a molecular switch for lytic reactivation. Here we demonstrate that KSHV RTA recruits CBP, the SWI/SNF chromatin remodeling complex, and the TRAP/Mediator coactivator into viral promoters through interactions with a short acidic sequence in the carboxyl region and that this recruitment is essential for RTA-dependent viral gene expression. The Brg1 subunit of SWI/SNF and the TRAP230 subunit of TRAP/Mediator were shown to interact directly with RTA. Consequently, genetic ablation of these interactions abolished KSHV lytic replication. These results demonstrate that the recruitment of CBP, SWI/SNF, and TRAP/Mediator complexes by RTA is the principal mechanism to direct well-controlled viral gene expression and thereby viral lytic reactivation.


Subject(s)
Gene Expression Regulation, Viral , Herpesvirus 8, Human/physiology , Immediate-Early Proteins/physiology , Nuclear Proteins/physiology , Receptors, Thyroid Hormone/physiology , Trans-Activators/physiology , Transcription Factors/physiology , Viral Proteins/physiology , Virus Activation , Amino Acid Sequence , B-Lymphocytes/virology , CREB-Binding Protein , Chromatin/metabolism , Chromatin/ultrastructure , DNA Helicases , Doxycycline/pharmacology , Humans , Macromolecular Substances , Mediator Complex , Molecular Sequence Data , Protein Interaction Mapping , Protein Subunits , Recombinant Fusion Proteins/physiology , Transcription Factors/metabolism , Transcription, Genetic/drug effects , Tumor Cells, Cultured/virology , Virus Replication
12.
Cancer Res ; 65(5): 2018-25, 2005 Mar 01.
Article in English | MEDLINE | ID: mdl-15753402

ABSTRACT

A major agenda for tumor immunology is the generation of specific immune responses leading to the destruction of incipient and frank neoplasia. In this report, we show that a novel HPV16 E7 fusion protein can produce objective therapeutic responses against incipient cervical cancer in genetically engineered mice that express in the cervix the HPV16 early region genes implicated as causative agents in human cervical cancer. Although nonresponsive toward the HPV16 E7 oncoprotein in the CD8+ T-cell compartment by virtue of MHC haplotype, the mice were capable of mounting an induced CD4+ T-cell response against E7, and in addition developed spontaneous anti-E7 antibodies. HPV16/CD4-/- mice showed increased tumor burden indicative of CD4-mediated immune surveillance. Seeking to enhance the CD4 response, we immunized mice bearing incipient cervical cancer with a recombinant protein fusing E7 with a mycobacterial heat shock protein. The incidences of cervical carcinoma and of high-grade dysplasia (CIN 3) were consequently reduced by comparison to control mice. Thus, an HPV16 E7 immunogen holds promise for noninvasive treatment and prevention of human cervical cancer.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Immunotherapy , Models, Animal , Oncogene Proteins, Viral/immunology , Uterine Cervical Neoplasms/therapy , Animals , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Bacterial Proteins/therapeutic use , CD4-Positive T-Lymphocytes/physiology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation , Chaperonin 60 , Chaperonins/genetics , Chaperonins/immunology , Chaperonins/therapeutic use , Female , Haplotypes/genetics , Homozygote , Humans , Immunization , Major Histocompatibility Complex/immunology , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Transgenic , Mycobacterium bovis/genetics , Oncogene Proteins, Viral/genetics , Ovary/immunology , Ovary/metabolism , Ovary/pathology , Papillomaviridae/genetics , Papillomaviridae/immunology , Papillomavirus E7 Proteins , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/therapeutic use , Tumor Cells, Cultured/transplantation , Tumor Cells, Cultured/virology , Uterine Cervical Neoplasms/immunology , Uterine Cervical Neoplasms/metabolism , Viral Vaccines/therapeutic use , Uterine Cervical Dysplasia/immunology , Uterine Cervical Dysplasia/metabolism , Uterine Cervical Dysplasia/therapy
13.
Free Radic Biol Med ; 41(9): 1488-98, 2006 Nov 01.
Article in English | MEDLINE | ID: mdl-17023276

ABSTRACT

Reactive species and perturbation of the redox balance have been implicated in the pathogenesis of many viral diseases, including hepatitis C. Previously, we made a surprising discovery that concentrations of H(2)O(2) that are nontoxic to host cells disrupted the hepatitis C virus (HCV) replication complex (RC) in Huh7 human hepatoma cells in a manner that suggested signaling. Here, we show that H(2)O(2) and interferon-gamma have comparable effects on the HCV subgenomic and genomic RNA replication in Huh7 cells. H(2)O(2) induced a gradual rise in the intracellular calcium concentration ([Ca(2+)](i)). Both rapid and sustained suppression of HCV RNA replication by H(2)O(2) depended on this calcium elevation. The peroxide-induced [Ca(2+)](i) elevation was independent of extracellular calcium and derived, at least in part, from the endoplasmic reticulum. Likewise, the suppression of the HCV RC by H(2)O(2) was independent of extracellular calcium but required an intracellular calcium source. Other agents that elevated [Ca(2+)](i) could also suppress the HCV RC, suggesting that calcium elevation might be sufficient to suppress HCV RNA replication. In conclusion, oxidants may modulate the HCV RC through calcium. Effects on the infectivity and the morphogenesis of HCV remain to be determined. These findings suggest possible regulatory roles for redox and calcium signaling during viral infections.


Subject(s)
Calcium/pharmacology , Carcinoma, Hepatocellular/virology , Hepacivirus/growth & development , Hepatitis C/virology , Reactive Oxygen Species/metabolism , Virus Replication , Adenosine Triphosphate/metabolism , Apoptosis , Blotting, Northern , Blotting, Western , Carcinoma, Hepatocellular/metabolism , Electroporation , Glucose Oxidase/pharmacology , Glutathione/metabolism , Hepacivirus/genetics , Hepatitis C/metabolism , Humans , Hydrogen Peroxide/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/virology , Oxidants/metabolism , Oxidation-Reduction , RNA, Viral/physiology , Tumor Cells, Cultured/virology
14.
Cancer Gene Ther ; 13(3): 273-80, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16151476

ABSTRACT

The Wnt signaling pathway is activated by mutations in the adenomatous polyposis coli (APC) or beta-catenin genes in most colon cancers, leading to the transactivation of promoters containing binding sites for the Tcf/LEF family of transcription factors. We have previously shown that it is possible to confer colon cancer specificity on autonomous parvoviruses by inserting Tcf sites into the viral P4 promoter. The mutant Tcf promoters were responsive to activation of the Wnt pathway but the viruses replicated poorly. We show here that reduction of the number of Tcf sites from four to two leads to an increase in the efficiency of replication and toxicity of the viruses in Co115 colon cancer cells, with only a small reduction in selectivity for cells with an active Wnt signaling pathway. Despite this improvement, virus production by most colon cancer cells remained low. Analysis of parental phH1 virus infection of SW480 colon cancer cells showed that the nonstructural and capsid proteins were expressed, but single stranded DNA and progeny virus were not produced. This defect reflects the dependence of autonomous parvoviruses on host functions for many steps in their replication cycle and represents a major limitation to the use of selectively replicating parvoviruses for colon cancer therapy.


Subject(s)
Colonic Neoplasms/virology , Gene Targeting , Parvovirus/physiology , Promoter Regions, Genetic , TCF Transcription Factors/genetics , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/virology , Binding Sites , Humans , Parvovirus/pathogenicity , Tumor Cells, Cultured/virology , Virus Replication
15.
Cancer Gene Ther ; 13(3): 326-34, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16138120

ABSTRACT

Replication-competent oncolytic herpes simplex viruses (HSV), modified by deletion of certain viral growth genes, can selectively target malignant cells. The viral growth gene gamma(1)34.5 has significant homology to GADD34 (growth arrest and DNA damage protein 34), which promotes cell cycle arrest and DNA repair in response to stressors such as radiation (XRT). By upregulating GADD34, XRT may result in greater oncolytic activity of HSV strains deficient in the gamma(1)34.5 gene. The human cholangiocarcinoma cell lines KMBC, SK-ChA-1 and YoMi were treated with NV1023, an oncolytic HSV lacking one copy of gamma(1)34.5. Viral proliferation assays were performed at a multiplicity of infection (MOI, number of viral particles per tumor cell) equal to 1, either alone or after XRT at 250 or 500 cGy. Viral replication was assessed by plaque assay. In vitro cytotoxicity assays were performed using virus at MOIs of 0.01 and 0.1, with or without XRT at 250 cGy and cell survival determined with lactate dehydrogenase assay. Established flank tumors in athymic mice were treated with a single intratumoral injection of virus (10(3) or 10(4) plaque forming units), either alone or after a single dose of XRT at 500 cGy, and tumor volumes measured. RT-PCR was used to measure GADD34 mRNA levels in all cell lines after a single dose of XRT at 250 or 500 cGy. NV1023 was tumoricidal in all three cell lines, but sensitivity to the virus varied. XRT enhanced viral replication in vitro in all cell lines. Combination treatment with low-dose XRT and virus was highly tumoricidal, both in vitro and in vivo. The greatest tumor volume reduction with combination therapy was seen with YoMi cells, the only cell line with increased GADD34 expression after XRT and the only cell line in which a synergistic treatment effect was suggested. In KMBC and SK-ChA-1 cells, neither of which showed increased GADD34 expression after XRT, tumor volume reduction was less pronounced and there was no suggestion of a synergistic effect in either case. Oncolytic HSV are effective in treating human cholangiocarcinoma cell lines, although sensitivity to virus varies. XRT-enhanced viral replication occurs through a mechanism that is not necessarily dependent on GADD34 upregulation. However, XRT-induced upregulation of GADD34 further promotes tumoricidal activity in viral strains deficient in the gamma(1)34.5 gene, resulting in treatment synergy; this effect is cell type dependent. Combined XRT and oncolytic viral therapy is a potentially important treatment strategy that may enhance the therapeutic ratios of both individual therapies.


Subject(s)
Cholangiocarcinoma/radiotherapy , Cholangiocarcinoma/therapy , Oncolytic Virotherapy , Simplexvirus/physiology , Virus Replication , Animals , Bile Duct Neoplasms/pathology , Bile Duct Neoplasms/radiotherapy , Bile Duct Neoplasms/therapy , Bile Ducts, Intrahepatic/radiation effects , Bile Ducts, Intrahepatic/virology , Cell Survival/physiology , Cell Survival/radiation effects , Cholangiocarcinoma/pathology , Combined Modality Therapy , Humans , L-Lactate Dehydrogenase/metabolism , Male , Mice , Mice, Nude , Tumor Cells, Cultured/metabolism , Tumor Cells, Cultured/radiation effects , Tumor Cells, Cultured/virology
16.
Cancer Res ; 57(8): 1447-51, 1997 Apr 15.
Article in English | MEDLINE | ID: mdl-9108444

ABSTRACT

Kaposi's sarcoma (KS) is a major AIDS-related malignancy associated with significant morbidity and mortality. Current chemotherapeutic regimens are associated with a dismal prognosis. In an effort to develop a new approach to KS treatment, we devised a gene therapy-based adenovirus retargeting schema that redirects the adenovirus to fibroblast growth factor receptors endogenously present on the cell surface of KS cells. By using a bifunctional conjugate consisting of a blocking antiadenoviral knob Fab linked to basic fibroblast growth factor, FGF2, the gene transduction of KS cells was enhanced 7.7-44 fold; recombinant adenoviruses encoding either the firefly luciferase reporter gene, or the herpes simplex thymidine kinase gene, demonstrated quantitative enhancement of expression in the KS cell lines. In this regard, two KS cell lines that were previously refractory to native adenovirus transduction could be successfully transduced by the addition of the conjugate. This study thus addresses the utility of adenoviral retargeting to the FGF receptor in KS cells that are ordinarily transduction refractory to standardized approaches and allows practical development of gene therapy approaches for the treatment of human KS.


Subject(s)
Acquired Immunodeficiency Syndrome/complications , Adenoviridae/genetics , Genetic Therapy , Genetic Vectors/therapeutic use , Immunoglobulin Fab Fragments/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Sarcoma, Kaposi/therapy , Transfection/methods , Humans , Immunoglobulin Fab Fragments/immunology , Luciferases/genetics , Luciferases/metabolism , Receptors, Fibroblast Growth Factor/immunology , Sarcoma, Kaposi/metabolism , Sarcoma, Kaposi/virology , Tumor Cells, Cultured/virology
17.
Cancer Res ; 58(9): 1893-900, 1998 May 01.
Article in English | MEDLINE | ID: mdl-9581830

ABSTRACT

Human papillomavirus type 16 (HPV16) E7 is a viral oncoprotein that is believed to play a major role in cervical neoplasia. Anti-HPV16 E7 intracellular single-chain antibodies (scFvs) were constructed to down-regulate HPV16 E7 oncoprotein in HPV DNA-containing cell lines. In these studies, we transfected anti-E7 scFvs into the HPV16-positive human cervical carcinoma cell lines CaSki and SiHa and tested them for their ability to inhibit cell proliferation and alter the level of HPV16 E7 oncoprotein. Our results showed that anti-HPV16 E7 scFvs inhibited cell proliferation by >85% in CaSki cells and by 95% in SiHa cells. E7 oncoprotein was down-regulated by anti-HPV16 E7 scFv, and its expression was inversely related to the amount of scFv transfected. However, there were no effects of transfecting scFvs alone in HPV-negative cell lines. These results imply that anti-HPV16 E7 scFvs only have specific anti-HPV16 E7 effects on cell proliferation and on the synthesis of virally encoded proteins in HPV-positive cell lines. Thus, transfection of HPV16 E7-positive tumors with antigen-specific scFvs may be a viable strategy for cervical cancer gene therapy.


Subject(s)
Antibodies, Viral/immunology , Gene Targeting , Genetic Therapy , Oncogene Proteins, Viral/antagonists & inhibitors , Papillomaviridae/immunology , Uterine Cervical Neoplasms/therapy , Animals , Antibodies, Viral/genetics , Cell Division , Down-Regulation , Female , Genes, Immunoglobulin/genetics , Humans , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/immunology , Papillomavirus E7 Proteins , Plasmids , Transfection , Tumor Cells, Cultured/virology , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/virology
18.
Cancer Res ; 63(12): 3073-8, 2003 Jun 15.
Article in English | MEDLINE | ID: mdl-12810631

ABSTRACT

The adenovirus E1A proteins are involved in the transcriptional activation of viral and cellular genes needed for controlling cell cycle and virus replication. Undifferentiated embryonic carcinoma cells have the ability to produce an E1A-like activity that can induce the expression of E1A-targeted adenoviral and cellular genes in the absence of the E1A products. Differentiated embryonic carcinoma cells lose the ability to produce the E1A-like activity. In this study, we investigated the E1A-like activity in cancer cells with an adenovirus having a mutated E1a gene. The mutation is generated by the insertion of a large DNA fragment in the E1a gene and interrupts the COOH-terminal region of both the E1A 12S and 13S proteins. The E1a-mutated virus can efficiently replicate in HepG2 and Hep3B liver cancer cells and produce high titers of virus. Replication of the E1a-mutated virus inhibits tumor formation and destroys tumors in vivo. The results obtained in this study imply that cancer cells may produce an E1A-like activity to support the selective replication of mutated virus in cancer cells. In addition, we found that although the E1a-mutated virus could not replicate in Huh1.cl2 liver cells, the viral DNA could amplify in the cells. This result suggests that replication of adenoviral DNA is necessary, but not sufficient, for generating infectious viral progeny and destroying tumor cells.


Subject(s)
Adenovirus E1A Proteins/genetics , Biological Therapy , Carcinoma, Hepatocellular/pathology , Defective Viruses/physiology , Liver Neoplasms/pathology , Mastadenovirus/physiology , Adenovirus E1A Proteins/deficiency , Adenovirus E1A Proteins/physiology , Animals , Carcinoma, Hepatocellular/therapy , Cytopathogenic Effect, Viral , DNA, Viral/genetics , Defective Viruses/genetics , Humans , Liver Neoplasms/therapy , Liver Neoplasms/virology , Mastadenovirus/genetics , Mice , Mice, Nude , Mutagenesis, Insertional , Tumor Cells, Cultured/virology , Virus Integration , Virus Replication , Xenograft Model Antitumor Assays
19.
Cancer Res ; 63(12): 3181-8, 2003 Jun 15.
Article in English | MEDLINE | ID: mdl-12810646

ABSTRACT

The catalytic component of human telomerase reverse transcriptase (hTERT) is not expressed in most primary somatic human cells, whereas the majority of cancer cells reactivate telomerase by transcriptional up-regulation of hTERT. Several studies demonstrated that the hTERT promoter can be used to restrict gene expression of E1-deleted replication defective adenoviral vectors to telomerase-positive cancer cells. In this study, a conditionally replicating adenovirus (hTERT-Ad) expressing E1A genes under control of a 255-bp hTERT-promoter was constructed. Additionally, an internal ribosomal entry site-enhanced green fluorescent protein cassette was inserted downstream of the E1B locus to monitor viral replication in vivo. Adenoviral replication of hTERT-Ad and enhancement of enhanced green fluorescent protein expression could be observed in all investigated telomerase-positive tumor cell lines. In contrast, hTERT-Ad infection of telomerase-negative primary human hepatocytes did not result in significant replication. The capability of hTERT-Ad to induce cytopathic effects in tumor cells was comparable with that of adenovirus wild type and significantly higher compared with ONYX-015, regardless of the p53 status of the tumor cells. Single application of low-dose hTERT-Ad to tumor xenografts led to significant inhibition of tumor growth, confirming the potential therapeutic value of conditionally replicative adenoviral vectors. These in vivo experiments also revealed that hTERT-Ad-mediated oncolysis was more efficient than ONYX-015 treatment. These results demonstrate that expression of E1A under transcriptional control of the hTERT promoter is sufficient for effective telomerase-dependent adenovirus replication as a promising perspective for the treatment of the majority of epithelial tumors.


Subject(s)
Adenoviridae , Adenoviruses, Human/physiology , Biological Therapy , Carcinoma, Hepatocellular/secondary , Liver Neoplasms/pathology , Telomerase/physiology , Virus Replication/physiology , Adenovirus E1A Proteins/genetics , Animals , DNA-Binding Proteins , Gene Expression Regulation, Neoplastic , Gene Expression Regulation, Viral , Genes, Reporter , Green Fluorescent Proteins , HeLa Cells/virology , Hepatocytes/virology , Humans , Immunotherapy, Active , Luminescent Proteins/genetics , Male , Mice , Mice, Nude , Neoplasm Proteins/physiology , Promoter Regions, Genetic , Recombinant Fusion Proteins/biosynthesis , Specific Pathogen-Free Organisms , TATA Box , Telomerase/genetics , Transcription, Genetic , Tumor Cells, Cultured/virology , Viral Vaccines/therapeutic use , Xenograft Model Antitumor Assays
20.
J Wildl Dis ; 52(1): 138-42, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26555105

ABSTRACT

Ranaviruses can be transmitted by contaminated water and sediment but must retain infectivity for a sufficient period to reach and infect a susceptible host. To determine the risk a virus represents once it enters the environment, its persistence in that environment must be determined. We evaluated the survival of frog virus 3 (FV3) in water and sediment from an English lake at temperatures of 4, 15, 20, and 30 C over time. The virus survived in both water and sediment; however, survival times were significantly lower in sediment. The virus lost infectivity in both matrices with a rise in temperature. In water, time required for a 90% reduction in virus titer decreased from 34 d at 4 C to 5 d at 30 C. In sediment, required time for a 90% reduction decreased from 10 d at 4 C to 1 d at 30 C. These results can be used to estimate the persistence of FV3 in the environment and indicate that the virus could remain infectious in temperate locations for extended periods during winter.


Subject(s)
Fresh Water/virology , Geologic Sediments/virology , Ranavirus/physiology , Amphibians , Animals , England , Temperature , Tumor Cells, Cultured/virology
SELECTION OF CITATIONS
SEARCH DETAIL