Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Arch Virol ; 165(3): 671-681, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31942645

ABSTRACT

Dengue virus (DENV) is the most common mosquito-borne viral disease. The World Health Organization estimates that 400 million new cases of dengue fever occur every year. Approximately 500,000 individuals develop severe and life-threatening complications from dengue fever, such as dengue shock syndrome (DSS) and dengue hemorrhagic fever (DHF), which cause 22,000 deaths yearly. Currently, there are no specific licensed therapeutics to treat DENV illness. We have previously shown that the MEK/ERK inhibitor U0126 inhibits the replication of the flavivirus yellow fever virus. In this study, we demonstrate that the MEK/ERK inhibitor AZD6244 has potent antiviral efficacy in vitro against DENV-2, DENV-3, and Saint Louis encephalitis virus (SLEV). We also show that it is able to protect AG129 mice from a lethal challenge with DENV-2 (D2S20). The molecule is currently undergoing phase III clinical trials for the treatment of non-small-cell lung cancer. The effect of AZD6244 on the DENV life cycle was attributed to a blockade of morphogenesis. Treatment of AG129 mice twice daily with oral doses of AZD6244 (100 mg/kg/day) prevented the animals from contracting dengue hemorrhagic fever (DHF)-like lethal disease upon intravenous infection with 1 × 105 PFU of D2S20. The effectiveness of AZD6244 was observed even when the treatment of infected animals was initiated 1-2 days postinfection. This was also followed by a reduction in viral copy number in both the serum and the spleen. There was also an increase in IL-1ß and TNF-α levels in mice that were infected with D2S20 and treated with AZD6244 in comparison to infected mice that were treated with the vehicle only. These data demonstrate the potential of AZD6244 as a new therapeutic agent to treat DENV infection and possibly other flavivirus diseases.


Subject(s)
Antiviral Agents/therapeutic use , Benzimidazoles/therapeutic use , Dengue Virus/growth & development , Severe Dengue/prevention & control , Animals , Cell Line , Cricetinae , Dengue Virus/drug effects , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Interleukin-1beta/blood , Mice , Severe Dengue/virology , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/blood
2.
PLoS Pathog ; 11(9): e1005148, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26325270

ABSTRACT

Vaccinia virus A33 is an extracellular enveloped virus (EEV)-specific type II membrane glycoprotein that is essential for efficient EEV formation and long-range viral spread within the host. A33 is a target for neutralizing antibody responses against EEV. In this study, we produced seven murine anti-A33 monoclonal antibodies (MAbs) by immunizing mice with live VACV, followed by boosting with the soluble A33 homodimeric ectodomain. Five A33 specific MAbs were capable of neutralizing EEV in the presence of complement. All MAbs bind to conformational epitopes on A33 but not to linear peptides. To identify the epitopes, we have adetermined the crystal structures of three representative neutralizing MAbs in complex with A33. We have further determined the binding kinetics for each of the three antibodies to wild-type A33, as well as to engineered A33 that contained single alanine substitutions within the epitopes of the three crystallized antibodies. While the Fab of both MAbs A2C7 and A20G2 binds to a single A33 subunit, the Fab from MAb A27D7 binds to both A33 subunits simultaneously. A27D7 binding is resistant to single alanine substitutions within the A33 epitope. A27D7 also demonstrated high-affinity binding with recombinant A33 protein that mimics other orthopoxvirus strains in the A27D7 epitope, such as ectromelia, monkeypox, and cowpox virus, suggesting that A27D7 is a potent cross-neutralizer. Finally, we confirmed that A27D7 protects mice against a lethal challenge with ectromelia virus.


Subject(s)
Antibodies, Neutralizing/metabolism , Membrane Glycoproteins/antagonists & inhibitors , Models, Molecular , Orthopoxvirus/physiology , Poxviridae Infections/virology , Viral Envelope Proteins/antagonists & inhibitors , Viral Tropism , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/genetics , Antibodies, Neutralizing/therapeutic use , Antibody Affinity , Antibody Specificity , Antigen-Antibody Complex/chemistry , Antigen-Antibody Complex/genetics , Antigen-Antibody Complex/metabolism , Chlorocebus aethiops , Female , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/genetics , Immunoglobulin Fab Fragments/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice, Inbred BALB C , Mutation , Orthopoxvirus/immunology , Poxviridae Infections/immunology , Poxviridae Infections/prevention & control , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Recombinant Proteins/therapeutic use , Vaccines, Synthetic/chemistry , Vaccines, Synthetic/genetics , Vaccines, Synthetic/metabolism , Vaccines, Synthetic/therapeutic use , Vero Cells , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Viral Vaccines/chemistry , Viral Vaccines/genetics , Viral Vaccines/metabolism , Viral Vaccines/therapeutic use
3.
PLoS Negl Trop Dis ; 9(6): e0003850, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26086739

ABSTRACT

Monkeypox virus (MPXV) is the etiological agent of human (MPX). It is an emerging orthopoxvirus zoonosis in the tropical rain forest of Africa and is endemic in the Congo-basin and sporadic in West Africa; it remains a tropical neglected disease of persons in impoverished rural areas. Interaction of the human population with wildlife increases human infection with MPX virus (MPXV), and infection from human to human is possible. Smallpox vaccination provides good cross-protection against MPX; however, the vaccination campaign ended in Africa in 1980, meaning that a large proportion of the population is currently unprotected against MPXV infection. Disease control hinges on deterring zoonotic exposure to the virus and, barring that, interrupting person-to-person spread. However, there are no FDA-approved therapies against MPX, and current vaccines are limited due to safety concerns. For this reason, new studies on pathogenesis, prophylaxis and therapeutics are still of great interest, not only for the scientific community but also for the governments concerned that MPXV could be used as a bioterror agent. In the present study, a new vaccination strategy approach based on three recombinant bovine herpesvirus 4 (BoHV-4) vectors, each expressing different MPXV glycoproteins, A29L, M1R and B6R were investigated in terms of protection from a lethal MPXV challenge in STAT1 knockout mice. BoHV-4-A-CMV-A29LgD106ΔTK, BoHV-4-A-EF1α-M1RgD106ΔTK and BoHV-4-A-EF1α-B6RgD106ΔTK were successfully constructed by recombineering, and their capacity to express their transgene was demonstrated. A small challenge study was performed, and all three recombinant BoHV-4 appeared safe (no weight-loss or obvious adverse events) following intraperitoneal administration. Further, BoHV-4-A-EF1α-M1RgD106ΔTK alone or in combination with BoHV-4-A-CMV-A29LgD106ΔTK and BoHV-4-A-EF1α-B6RgD106ΔTK, was shown to be able to protect, 100% alone and 80% in combination, STAT1(-/-) mice against mortality and morbidity. This work demonstrated the efficacy of BoHV-4 based vectors and the use of BoHV-4 as a vaccine-vector platform.


Subject(s)
Antigens, Viral/immunology , Herpesvirus 4, Bovine/physiology , Monkeypox virus/immunology , Mpox (monkeypox)/prevention & control , STAT1 Transcription Factor/metabolism , Viral Vaccines/immunology , Amino Acid Sequence , Animals , Base Sequence , Cattle , Cell Line , Gene Expression Regulation , Genetic Vectors , Herpesvirus 4, Bovine/immunology , Humans , Mice , Mice, Knockout , Molecular Sequence Data , STAT1 Transcription Factor/genetics , Transfection , Viral Vaccines/genetics
4.
J Virol ; 87(17): 9610-9, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23804638

ABSTRACT

Adenoviruses (Ads) are promising vectors for therapeutic interventions in humans. When injected into the bloodstream, Ad vectors can bind several vitamin K-dependent blood coagulation factors, which contributes to virus sequestration in the liver by facilitating transduction of hepatocytes. Although both coagulation factors FVII and FX bind the hexon protein of human Ad serotype 5 (HAdv5) with a very high affinity, only FX appears to play a role in mediating Ad-hepatocyte transduction in vivo. To understand the discrepancy between efficacy of FVII binding to hexon and its apparently poor capacity for supporting virus cell entry, we analyzed the HAdv5-FVII complex by using high-resolution cryo-electron microscopy (cryo-EM) followed by molecular dynamic flexible fitting (MDFF) simulations. The results indicate that although hexon amino acids T423, E424, and T425, identified earlier as critical for FX binding, are also involved in mediating binding of FVII, the FVII GLA domain sits within the surface-exposed hexon trimer depression in a different orientation from that found for FX. Furthermore, we found that when bound to hexon, two proximal FVII molecules interact via their serine protease (SP) domains and bury potential heparan sulfate proteoglycan (HSPG) receptor binding residues within the dimer interface. In contrast, earlier cryo-EM studies of the Ad-FX interaction showed no evidence of dimer formation. Dimerization of FVII bound to Ad may be a contributing mechanistic factor for the differential infectivity of Ad-FX and Ad-FVII complexes, despite high-affinity binding of both these coagulation factors to the virus.


Subject(s)
Adenoviruses, Human/genetics , Adenoviruses, Human/physiology , Factor VII/chemistry , Factor VII/metabolism , Factor X/chemistry , Factor X/metabolism , Genetic Vectors , Animals , CHO Cells , Cricetinae , Cricetulus , HEK293 Cells , Hepatocytes/metabolism , Hepatocytes/virology , Humans , Models, Molecular , Molecular Dynamics Simulation , Protein Structure, Quaternary , Protein Structure, Tertiary , Virus Internalization
5.
Cell Rep ; 3(6): 1840-6, 2013 Jun 27.
Article in English | MEDLINE | ID: mdl-23770239

ABSTRACT

Although molecular components that execute noninflammatory apoptotic cell death are well defined, molecular pathways that trigger necrotic cell death remain poorly characterized. Here, we show that in response to infection with adenovirus or Listeria monocytogenes, macrophages in vivo undergo rapid proinflammatory necrotic death that is controlled by interferon-regulatory factor 3 (IRF3). The transcriptional activity of IRF3 is, surprisingly, not required for the induction of necrosis, and it proceeds normally in mice deficient in all known regulators of necrotic death or IRF3 activation, including RIPK3, caspases 1, 8, or 11, STING, and IPS1/MAVS. Although L. monocytogenes triggers necrosis to promote the infection, IRF3-dependent necrosis is required for reducing pathogen burden in the models of disseminated infection with adenovirus. Therefore, our studies implicate IRF3 as a principal and nonredundant component of a physiologically regulated necrotic cell-death pathway that operates as an effective innate immune mechanism of host protection against disseminated virus infection.


Subject(s)
Adenovirus Infections, Human/immunology , Adenoviruses, Human/immunology , Interferon Regulatory Factor-3/immunology , Listeria monocytogenes/immunology , Listeriosis/immunology , Macrophages/microbiology , Macrophages/pathology , Adenovirus Infections, Human/pathology , Animals , Caspases/metabolism , Immunity, Innate/immunology , Interferon Regulatory Factor-3/deficiency , Interferon Regulatory Factor-3/genetics , Listeriosis/pathology , Macrophages/immunology , Macrophages/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Necrosis/immunology , Necrosis/pathology , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Signal Transduction
6.
Science ; 338(6108): 795-8, 2012 Nov 09.
Article in English | MEDLINE | ID: mdl-23019612

ABSTRACT

Although coagulation factors play a role in host defense for "living fossils" such as horseshoe crabs, the role of the coagulation system in immunity in higher organisms remains unclear. We modeled the interface of human species C adenovirus (HAdv) interaction with coagulation factor X (FX) and introduced a mutation that abrogated formation of the HAdv-FX complex. In vivo genome-wide transcriptional profiling revealed that FX-binding-ablated virus failed to activate a distinct network of nuclear factor κB-dependent early-response genes that are activated by HAdv-FX complex downstream of TLR4/MyD88/TRIF/TRAF6 signaling. Our study implicates host factor "decoration" of the virus as a mechanism to trigger an innate immune sensor that responds to a misplacement of coagulation FX from the blood into intracellular macrophage compartments upon virus entry into the cell.


Subject(s)
Adenoviridae Infections/immunology , Adenoviruses, Human/immunology , Adenoviruses, Human/metabolism , Factor X/metabolism , Immunity, Innate , Adenoviridae Infections/metabolism , Adenoviridae Infections/virology , Adenoviruses, Human/genetics , Animals , CHO Cells , Capsid Proteins/chemistry , Capsid Proteins/genetics , Capsid Proteins/metabolism , Cell Line, Tumor , Cricetinae , Cricetulus , Cryoelectron Microscopy , Cytokines/metabolism , Factor X/chemistry , Gene Expression Profiling , Gene Expression Regulation , Hepatocytes/virology , Humans , Macrophages/metabolism , Macrophages/virology , Mice , Mice, Inbred C57BL , Molecular Dynamics Simulation , Mutation , NF-kappa B/metabolism , Signal Transduction , Virus Internalization
7.
Methods Mol Biol ; 797: 35-52, 2012.
Article in English | MEDLINE | ID: mdl-21948467

ABSTRACT

Oncolytic (replication-competent) adenoviruses (Ads) represent the most advanced platform for cancer gene therapy. These viral vectors ablate tumors by killing tumor cells in the process of virus replication. As progeny virions are released, they infect remaining cancer cells, generating a bystander effect. Ads engineered for increased cancer specificity produce less damage to normal tissues. First-generation oncolytic Ads have demonstrated acceptable levels of safety while the efficacy was observed only in combination with chemotherapy and/or radiation. Second-generation oncolytic Ads are armed with therapeutic transgenes to increase release, spread, and bystander effect for enhancing the efficacy. Third-generation oncolytic Ads are armed vectors with capsid modifications for transductional detargeting from normal tissues and targeting to cancer cells. Chemical modification of the capsid additionally improves therapeutic window. Here, we describe methods for generation and characterization of advanced-generation oncolytic Ads.


Subject(s)
Adenoviridae/chemistry , Adenoviridae/genetics , Genetic Therapy , Neoplasms/therapy , Oncolytic Virotherapy , Oncolytic Viruses/chemistry , Oncolytic Viruses/genetics , Adenoviridae/isolation & purification , Animals , Capsid/chemistry , Genetic Vectors , Humans , Mice , Mice, Nude , Oncolytic Viruses/isolation & purification
8.
Hum Gene Ther ; 21(2): 179-90, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19788385

ABSTRACT

Oncolytic virotherapy makes use of the natural ability of viruses to infect and kill cancer cells. Adenovirus serotype 5 (Ad5) has been approved for use in humans as a therapy for solid cancers. In this study, we have tested whether Ad5 and low-seroprevalence adenoviruses can be used as oncolytics for multiple myeloma (MM). We show that Ad5 productively infects most myeloma cell lines, replicates to various degrees, and mediates oncolytic cell killing in vitro and in vivo. Comparison of Ad5 with low-seroprevalence Ads on primary marrow samples from MM patients revealed striking differences in the abilities of different adenoviral serotypes to kill normal CD138(-) cells and CD138(+) MM cells. Ad5 and Ad6 from species C and Ad26 and Ad48 from species D all mediated killing of CD138(+) cells with low-level killing of CD138(-) cells. In contrast, Ad11, Ad35, Ad40, and Ad41 mediated weak oncolytic effects in all of the cells. Comparison of cell binding, cell entry, and replication revealed that Ad11 and Ad35 bound MM cells 10 to 100 times better than other serotypes. However, after this efficient interaction, Ad11 and Ad35 viral DNA was not replicated and cell killing did not occur. In contrast, Ad5, Ad6, Ad26, and Ad48 all replicated 10- to 100-fold in MM cells and this correlated with cell killing. These data suggest that Ad5 and other low-seroprevalence adenoviruses may have utility as oncolytic agents against MM and other hematologic malignancies.


Subject(s)
Adenoviridae/pathogenicity , Multiple Myeloma/therapy , Multiple Myeloma/virology , Oncolytic Virotherapy/methods , Oncolytic Viruses/physiology , Adenoviridae/classification , Adenoviridae/genetics , Adenoviridae/metabolism , Animals , Cell Line, Tumor , Cytopathogenic Effect, Viral , Disease Models, Animal , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Oncolytic Viruses/genetics , Oncolytic Viruses/metabolism , Treatment Outcome , Virus Replication
9.
Mol Ther ; 17(12): 2121-30, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19755961

ABSTRACT

One of the significant hurdles toward safe and efficacious systemic treatment of cancer with oncolytic adenoviruses (Ads) is dose-limiting hepatotoxicity that prevents the increase of a therapeutic dose. In this study, we expanded the therapeutic window of oncolytic serotype 5 Ad (Ad5) by a genetic modification of hypervariable loop 5 (HVR5) in the capsid protein hexon that prevented infection of hepatocytes due to ablation of binding to blood factors. This oncolytic virus, Ad-GL-HB, had significantly reduced levels of hepatocyte transduction in immunocompetent and immunodeficient mice as compared to parental virus Ad-GL. The hepatocyte detargeting decreased liver damage and increased the maximum tolerated dose of Ad-GL-HB tenfold relative to that of Ad-GL. Intravenous (i.v.) injection of Ad-GL or Ad-GL-HB into tumor-bearing mice produced equally increased survival rates demonstrating that while Ad-GL-HB detargeted hepatocytes, it sustained tumor cell infection after systemic administration. The significantly improved safety of the virus allowed it to be used at increased doses for improved systemic antitumor efficacy. Our results suggest that hexon modifications provide valuable strategies for systemic oncolytic Ad therapy.


Subject(s)
Adenoviridae/genetics , Capsid Proteins/genetics , Genetic Vectors/therapeutic use , Neoplasms, Experimental/therapy , Oncolytic Virotherapy , Oncolytic Viruses/genetics , Animals , Female , Hepatocytes/metabolism , Humans , Luminescence , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Nude , Neoplasms, Experimental/genetics , Neoplasms, Experimental/virology , Transduction, Genetic
10.
Hum Gene Ther ; 20(9): 975-88, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19469693

ABSTRACT

Oncolytic adenoviruses are anticancer agents that replicate within tumors and spread to uninfected tumor cells, amplifying the anticancer effect of initial transduction. We tested whether coating the viral particle with polyethylene glycol (PEG) could reduce transduction of hepatocytes and hepatotoxicity after systemic (intravenous) administration of oncolytic adenovirus serotype 5 (Ad5). Conjugating Ad5 with high molecular weight 20-kDa PEG but not with 5-kDa PEG reduced hepatocyte transduction and hepatotoxicity after intravenous injection. PEGylation with 20-kDa PEG was as efficient at detargeting adenovirus from Kupffer cells and hepatocytes as virus predosing and warfarin. Bioluminescence imaging of virus distribution in two xenograft tumor models in nude mice demonstrated that PEGylation with 20-kDa PEG reduced liver infection 19- to 90-fold. Tumor transduction levels were similar for vectors PEGylated with 20-kDa PEG and unPEGylated vectors. Anticancer efficacy after a single intravenous injection was retained at the level of unmodified vector in large established prostate carcinoma xenografts, resulting in complete elimination of tumors in all animals and long-term tumor-free survival. Anticancer efficacy after a single intravenous injection was increased in large established hepatocellular carcinoma xenografts, resulting in significant prolongation of survival as compared with unmodified vector. The increase in efficacy was comparable to that obtained with predosing and warfarin pretreatment, significantly extending the median of survival. Shielding adenovirus with 20-kDa PEG may be a useful approach to improve the therapeutic window of oncolytic adenovirus after systemic delivery to primary and metastatic tumor sites.


Subject(s)
Adenoviridae/chemistry , Adenoviridae/physiology , Carcinoma, Hepatocellular/therapy , Genetic Vectors/administration & dosage , Genetic Vectors/chemistry , Hepatocytes/virology , Liver Neoplasms/therapy , Oncolytic Virotherapy/methods , Polyethylene Glycols/chemistry , Transduction, Genetic , Adenoviridae/genetics , Animals , Carcinoma/therapy , Carcinoma, Hepatocellular/mortality , Cell Line, Tumor , Female , Genetic Vectors/adverse effects , Genetic Vectors/physiology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Injections, Intravenous , Liver Neoplasms/mortality , Luciferases/genetics , Luciferases/metabolism , Male , Mice , Mice, Nude , Polyethylene Glycols/pharmacology , Prostatic Neoplasms/therapy , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Treatment Outcome
11.
Cancer Res ; 68(14): 5896-904, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18632644

ABSTRACT

Liver tropism of systemically delivered adenoviruses (Ad) represents a considerable challenge for their use as anticancer therapeutics. More than 90% of i.v. injected Ad is rapidly taken up by the liver leading to hepatotoxicity, reduced virus uptake by target tumor tissue, and diminished therapeutic efficacy. The lack of clinical activity of systemically given oncolytic Ad demands for better understanding and improvement of virus pharmacokinetics. We studied the effects of Ad "detargeting" from liver macrophages (Kupffer cells) and hepatocytes on toxicity and anticancer efficacy using a nonattenuated oncolytic Ad expressing enhanced green fluorescent protein-firefly luciferase fusion protein (Ad-EGFPLuc). Kupffer cell depletion before i.v. injection of Ad-EGFPLuc increased transgene expression in the liver 40.7-fold on day 3 after the injection indicating compensatory enhancement of hepatocyte transduction due to increased bioavailability of the virus. Pretreatment of mice with the anticoagulant drug warfarin to block blood factor-dependent binding of the virus to hepatocytes markedly reduced luciferase expression in the liver and mediated the corresponding decrease of hepatotoxicity in mice with intact and depleted liver macrophages. Combined depletion of Kupffer cells and pretreatment with warfarin before a single i.v. injection of Ad-EGFPLuc significantly reduced tumor growth and prolonged survival of nude mice bearing subcutaneous xenografts of aggressive human hepatocellular carcinoma. The improved antitumor activity correlated with enhanced transgene expression and virus spread in the tumors. These data suggest that detargeting oncolytic Ad from liver macrophages and hepatocytes is an effective strategy to increase the therapeutic window for therapy against disseminated tumor sites.


Subject(s)
Anticoagulants/pharmacology , Gene Expression Regulation , Macrophages/metabolism , Oncolytic Virotherapy/methods , Animals , Carcinoma, Hepatocellular/drug therapy , Cell Line, Tumor , Female , Hepatocytes/metabolism , Humans , Kupffer Cells/metabolism , Liver Neoplasms/drug therapy , Luciferases/metabolism , Mice , Mice, Nude , Neoplasm Transplantation
12.
J Virol ; 81(23): 12918-26, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17881437

ABSTRACT

A short open reading frame named the "U exon," located on the adenovirus (Ad) l-strand (for leftward transcription) between the early E3 region and the fiber gene, is conserved in mastadenoviruses. We have observed that Ad5 mutants with large deletions in E3 that infringe on the U exon display a mild growth defect, as well as an aberrant Ad E2 DNA-binding protein (DBP) intranuclear localization pattern and an apparent failure to organize replication centers during late infection. Mutants in which the U exon DNA is reconstructed have a reversed phenotype. Chow et al. (L. T. Chow et al., J. Mol. Biol. 134:265-303, 1979) described mRNAs initiating in the region of the U exon and spliced to downstream sequences in the late DBP mRNA leader and the DBP-coding region. We have cloned this mRNA (as cDNA) from Ad5 late mRNA; the predicted protein is 217 amino acids, initiating in the U exon and continuing in frame in the DBP leader and in the DBP-coding region but in a different reading frame from DBP. Polyclonal and monoclonal antibodies generated against the predicted U exon protein (UXP) showed that UXP is approximately 24K in size by immunoblot and is a late protein. At 18 to 24 h postinfection, UXP is strongly associated with nucleoli and is found throughout the nucleus; later, UXP is associated with the periphery of replication centers, suggesting a function relevant to Ad DNA replication or RNA transcription. UXP is expressed by all four species C Ads. When expressed in transient transfections, UXP complements the aberrant DBP localization pattern of UXP-negative Ad5 mutants. Our data indicate that UXP is a previously unrecognized protein derived from a novel late l-strand transcription unit.


Subject(s)
Adenoviruses, Human/genetics , Adenoviruses, Human/physiology , Viral Proteins/genetics , Viral Proteins/physiology , Adenovirus E2 Proteins/analysis , Cell Nucleolus/chemistry , Cell Nucleus/chemistry , Cloning, Molecular , Humans , Immunoblotting , Molecular Sequence Data , Molecular Weight , Open Reading Frames , RNA, Messenger/genetics , RNA, Viral/genetics , Sequence Analysis, DNA , Sequence Deletion , Viral Proteins/biosynthesis , Viral Proteins/chemistry , Virus Replication/physiology
13.
Methods Mol Med ; 130: 223-35, 2007.
Article in English | MEDLINE | ID: mdl-17401177

ABSTRACT

Adenovirus research often requires purified high-titer virus stocks and accurate virus titers for use in experiments. Accurate titers are important for quantitative, interpretable, and reproducible results. This is especially true when there are comparisons of different mutant viruses following infection. This chapter details the large-scale preparation of adenovirus (either replication-competent or replication-defective) in spinner cultures (e.g., KB, HeLa, or 293 cells). Protocols for harvesting cells and isolation of adenovirus by CsCl banding are presented. Methods for titering adenovirus by plaque assay are presented along with a discussion of how plaque assays can be used to determine the kinetics of cell killing and cytolysis by adenoviruses.


Subject(s)
Adenoviridae/classification , Adenoviridae/isolation & purification , Cesium , Chlorides , Adenoviridae/genetics , Cell Line , Centrifugation, Density Gradient/methods , HeLa Cells , Humans , Indicators and Reagents , KB Cells , Kidney
14.
Mol Ther ; 15(3): 598-607, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17191072

ABSTRACT

Novel approaches are needed to improve the antitumor potency and to increase the cancer specificity of oncolytic adenoviruses (Ad). We hypothesized that the combination of interferon-alpha (IFN-alpha) expression with a specific mutation in the e1a gene of Ad could target vector replication to genetic defects in the IFN-alpha pathway resulting in both improved antitumor efficacy and reduced toxicity. The conditionally replicative Ad vector KD3-IFN carries the dl1101/1107 mutation in the e1a gene that eliminates binding of E1A proteins to p300/CBP and pRb. KD3-IFN expresses human IFN-alpha in concurrence with vector replication and overexpresses the adenovirus death protein (ADP; E3-11.6K). The antitumor activity of KD3-IFN was significantly higher than that of a control vector in established human hepatocellular carcinoma tumors in immunodeficient mice and in hamster kidney cancer tumors in immunocompetent Syrian hamsters. The dl1101/1107 mutation rendered Ad replication sensitive to the antiviral effect of IFN-alpha in normal as opposed to cancer cells. These results translated to reduced vector toxicity upon systemic administration to C57BL/6 mice. The combination of Ad oncolysis, ADP overexpression, and IFN-alpha-mediated immunotherapy represents a three-pronged approach for increasing the anticancer efficacy of replicative Ads. Exploiting the dl1101/1107 mutation provides a mechanism for additional selectivity of IFN-alpha-expressing replication-competent Ads.


Subject(s)
Adenoviridae/genetics , Adenovirus E1A Proteins/metabolism , Interferon-alpha/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Oncolytic Viruses/genetics , Adenovirus E1A Proteins/genetics , Animals , Cell Line , Cell Survival , Cricetinae , Female , Gene Expression , Genetic Therapy , Genetic Vectors/genetics , Genome, Viral/genetics , Interferon-alpha/genetics , Liver Neoplasms/genetics , Liver Neoplasms/therapy , Mice , Mice, Nude , Mutation/genetics , Survival Rate , Time Factors , Transgenes/genetics , Virus Replication , Xenograft Model Antitumor Assays
15.
Mol Ther ; 15(3): 598-607, 2007 Mar.
Article in English | MEDLINE | ID: mdl-28182898

ABSTRACT

Novel approaches are needed to improve the antitumor potency and to increase the cancer specificity of oncolytic adenoviruses (Ad). We hypothesized that the combination of interferon-alpha (IFN-α) expression with a specific mutation in the e1a gene of Ad could target vector replication to genetic defects in the IFN-α pathway resulting in both improved antitumor efficacy and reduced toxicity. The conditionally replicative Ad vector KD3-IFN carries the dl1101/1107 mutation in the e1a gene that eliminates binding of E1A proteins to p300/CBP and pRb. KD3-IFN expresses human IFN-α in concurrence with vector replication and overexpresses the adenovirus death protein (ADP; E3-11.6K). The antitumor activity of KD3-IFN was significantly higher than that of a control vector in established human hepatocellular carcinoma tumors in immunodeficient mice and in hamster kidney cancer tumors in immunocompetent Syrian hamsters. The dl1101/1107 mutation rendered Ad replication sensitive to the antiviral effect of IFN-α in normal as opposed to cancer cells. These results translated to reduced vector toxicity upon systemic administration to C57BL/6 mice. The combination of Ad oncolysis, ADP overexpression, and IFN-α-mediated immunotherapy represents a three-pronged approach for increasing the anticancer efficacy of replicative Ads. Exploiting the dl1101/1107 mutation provides a mechanism for additional selectivity of IFN-α-expressing replication-competent Ads.

16.
Cancer Gene Ther ; 12(7): 617-26, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15761475

ABSTRACT

Avian adenovirus CELO is a novel adenovirus vector system with the advantages of efficient production, high virion stability, and the absence of crossreactivity with Ad5-neutralizing antibodies. In this study, we evaluated the anticancer efficacy of a CELO vector encoding the herpes simplex virus type 1 thymidine kinase, a prodrug-activating therapeutic gene. Vectors carrying the gene for HSV-tk or EGFP under the control of the HCMV promoter in place of the "nonessential" region of the CELO genome were constructed. Anticancer activity of the CELO-TK vector was studied in vitro, in human and murine tumor cells in cell culture, and in vivo, in established subcutaneous murine B16 melanoma tumors in C57BL/6 mice. The CELO-TK vector mediated delivery of functional HSV-tk to tumor cell lines in cell culture. Comparison of the CELO-TK vector to a first-generation human adenovirus type 5 vector Ad5-TK in cultured H1299 cells showed equal levels of functional activity at increasing multiplicities of infection with CELO-based vector. CELO vectors allowed for transduction and expression of EGFP and HSV-tk genes in subcutaneous melanoma tumors in C57BL/6 mice. Intratumoral injections of CELO-TK followed by ganciclovir administration resulted in suppression of tumor growth and significantly increased the median of survival. The results of the study demonstrated the efficacy of CELO vector as a vehicle for the delivery of prodrug-activating genes such as HSV-tk to tumor cells in vitro and in vivo.


Subject(s)
Fowl adenovirus A/genetics , Genetic Therapy , Genetic Vectors , Melanoma, Experimental/therapy , Simplexvirus/enzymology , Thymidine Kinase/genetics , Animals , Cell Line, Tumor , Female , Green Fluorescent Proteins/genetics , Humans , Melanoma, Experimental/genetics , Melanoma, Experimental/virology , Mice , Mice, Inbred C57BL , Skin Neoplasms/genetics , Skin Neoplasms/therapy , Skin Neoplasms/virology , Transduction, Genetic , Xenograft Model Antitumor Assays
17.
Hum Gene Ther ; 16(1): 139-46, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15703497

ABSTRACT

Oncolytic human adenovirus (Ad) vectors exert their antitumor effect by replicating in and lysing tumor cells. These vectors are commonly evaluated in immunodeficient mice bearing human tumor xenografts. However, this model suffers because the mice are immunodeficient and are not permissive for human Ads. We have developed a cotton rat model to test the selectivity, immunogenicity, and efficacy of oncolytic Ad vectors. The cotton rat is a rodent species that is semipermissive for human Ads. We show that the cotton cancer rat cell line LCRT supports the replication of human Ad in tissue culture and that the cells are destroyed on virus replication. When injected subcutaneously, LCRT cells formed tumors in immunocompetent cotton rats, and the growth of these tumors was delayed by the injection of an oncolytic Ad vector. Replication of the Ad vector in the tumor was demonstrated by sampling tumor tissue and isolating infectious virus particles at various times after intratumoral injection of the virus. We propose that the cotton rat can be used as an animal model to evaluate oncolytic Ad vectors.


Subject(s)
Adenoviruses, Human/physiology , Disease Models, Animal , Genetic Vectors , Neoplasms/therapy , Adenovirus Infections, Human/therapy , Adenovirus Infections, Human/virology , Animals , Female , Genetic Therapy , Humans , Neoplasms/virology , Rats , Sigmodontinae , Virus Replication/physiology
18.
Virus Res ; 108(1-2): 149-59, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15681065

ABSTRACT

The majority of proteins encoded in the early 3 (E3) region of human subgroup C adenoviruses function to modulate the host immune response. For example, gp19K, one of these E3 proteins, prevents the major histocompatibility complex type I (MHC-I) from presenting viral antigens on the surface of the infected cell. Other E3 proteins, such as the RID and 14.7K proteins, counteract the effector phase of the cellular immune response. In order to study further the effects of these proteins, we constructed an E1-/E3- adenovirus vector, Ad/E3, that contains all the E3 genes with the exception of the cytolytic adp gene, inserted into the deleted E1 region. The transcription of the E3 genes in this vector is driven by a CMV promoter in place of the native E3 promoter. Ad/E3 expressed close to wild-type adenovirus levels of all E3 proteins, and these proteins appear to function normally in cell culture. For example, in Ad/E3-infected cells, surface expression of MHC-I was down-regulated, as was cell surface display of death receptors Fas and TRAIL Receptor 1. A human cell line of lung origin (A549), which was rapidly rejected after transplantation into C57BL/6 mice, was protected for an extended time from the host immune response after infection with an Ad/E3, and went through a number of divisions in immunocompetent mice. These latter results indicate that the E3 proteins protect cells from destruction by the immune system.


Subject(s)
Adenovirus E3 Proteins/physiology , Graft Rejection , Immunosuppression Therapy , Transplants , Adenovirus E3 Proteins/genetics , Adenoviruses, Human/genetics , Adenoviruses, Human/physiology , Animals , Cell Line, Tumor , Cell Transformation, Viral , Female , Genetic Vectors , Humans , Immunocompetence , Mice , Mice, Inbred C57BL , Transplantation, Heterologous
19.
J Virol ; 78(22): 12297-307, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15507617

ABSTRACT

Adenoviruses (Ads) encode several proteins within the early region 3 (E3) transcription unit that help protect infected cells from elimination by the immune system. Among these immunomodulatory proteins, the receptor internalization and degradation (RID) protein complex, which is composed of the RIDalpha (formerly E3-10.4K) and RIDbeta (formerly E3-14.5K) subunits, stimulates the internalization and degradation of certain members of the tumor necrosis factor (TNF) receptor superfamily, thus blocking apoptosis initiated by Fas and TNF-related apoptosis-inducing ligand (TRAIL). The experiments reported here show that TRAIL receptor 2 (TR2) is cleared from the cell surface in Ad-infected cells. Virus mutants containing deletions that span E3 were used to show that the RID and E3-6.7K proteins are both necessary for the internalization and degradation of TR2, whereas only the RID protein is required for TRAIL receptor 1 downregulation. In addition, replication-defective Ad vectors that express individual E3 proteins were used to establish that the RID and E3-6.7K proteins are sufficient to clear TR2. These data demonstrate that E3-6.7K is an important component of the antiapoptosis arsenal encoded by the E3 transcription unit of subgroup C Ads.


Subject(s)
Adenovirus E3 Proteins/physiology , Proteins/physiology , Receptors, Tumor Necrosis Factor/metabolism , Apoptosis , Cell Line, Tumor , Down-Regulation , Humans , Receptor-Interacting Protein Serine-Threonine Kinases , Receptors, TNF-Related Apoptosis-Inducing Ligand
20.
Cancer Res ; 64(10): 3638-44, 2004 May 15.
Article in English | MEDLINE | ID: mdl-15150123

ABSTRACT

We have constructed a novel oncolytic adenovirus (Ad) vector named VRX-009 that combines enhanced cell spread with tumor-specific replication. Enhanced spread, which could significantly increase antitumor efficacy, is mediated by overexpression of the Ad cytolytic protein named ADP (also known as E3-11.6K). Replication of VRX-009 is restricted to cells with a deregulated wnt signal transduction pathway by replacement of the wild-type Ad E4 promoter with a synthetic promoter consisting of five consensus binding sites for the T-cell factor transcription factor. Tumor-selective replication is indicated by several lines of evidence. VRX-009 expresses E4ORF3, a representative Ad E4 protein, only in colon cancer cell lines. Furthermore, VRX-009 replicates preferentially in colon cancer cell lines as evidenced by virus productivity 2 orders of magnitude higher in SW480 colon cancer cells than in A549 lung cancer cells. Replication in primary human bronchial epithelial cells and human umbilical vein endothelial cells was also significantly lower than in SW480 cells. When tested in human tumor xenografts in nude mice, VRX-009 effectively suppressed the growth of SW480 colon tumors but not of A549 lung tumors. VRX-009 may provide greater level of antitumor efficacy than standard oncolytic Ad vectors in tumors in which a defect in wnt signaling increases the level of nuclear beta-catenin.


Subject(s)
Adenoviridae/physiology , Adenovirus E3 Proteins/physiology , Neoplasms/therapy , Neoplasms/virology , Proto-Oncogene Proteins/genetics , Adenoviridae/genetics , Adenovirus E3 Proteins/biosynthesis , Adenovirus E3 Proteins/genetics , Adenovirus E4 Proteins/biosynthesis , Adenovirus E4 Proteins/genetics , Animals , Cell Division/physiology , Cell Line, Tumor , Cytopathogenic Effect, Viral , Endothelium, Vascular/metabolism , Endothelium, Vascular/virology , Genetic Vectors/genetics , Humans , Mice , Neoplasms/genetics , Plasmids/genetics , Proto-Oncogene Proteins/physiology , Signal Transduction , Virus Replication , Wnt Proteins , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...