Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters











Publication year range
1.
J Thromb Haemost ; 14(2): 356-65, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26581031

ABSTRACT

UNLABELLED: ESSENTIALS: Antithrombin III (AT)ß binds heparin with higher affinity than ATα. A conformation-specific antibody against ATß, TPP2009, was made to investigate ATß in hemostasis. TPP2009 bound specifically to heparin-ATß and greatly reduced the anticoagulant effect of AT. This antibody was effective in elucidating the importance of ATß in hemostasis. BACKGROUND: Antithrombin III (AT)ß is an isoform of AT that lacks the post-translational carbohydrate modification at Asn135. This isoform binds heparin with greater affinity than ATα, and has been shown to target antithrombotic function to the extracellular vascular endothelial injury site. OBJECTIVES: To characterize a conformation-specific antibody against ATß and begin to investigate the role of ATß in maintaining hemostasis. METHODS: Surface plasmon resonance (SPR), antigen binding and functional assays were conducted to characterize the mode of action of antibodies generated against heparin-bound ATß (ATß*H) by the use of phage display. RESULTS: SPR and binding studies showed that one of the antibodies, TPP2009, bound specifically to ATß*H and glycosaminoglycan-associated ATß on endothelial cells. In diluted prothrombin and activated factor X (FXa)-induced clotting assays, TPP2009 dose-dependently reduced the anticoagulant effect of heparin in non-hemophilic and FVIII-deficient human plasma, with half-maximal effective concentrations (EC50 ) of 10.5 nm and 4.7 nm, respectively. In AT-deficient human plasma, TPP2009 dose-dependently inhibited the effects of exogenously added ATß and heparin. In purified systems with ATß and pentasaccharide, TPP2009 restored > 91% of FXa activity. TPP2009 dose-dependently reversed the effects of heparin in rabbit (EC50 , 25.7 nm) and cynomolgus monkey (EC50 , 21.5 nm) plasma, but not in mouse plasma. TPP2009 was also effective in partially restoring FXa activity in rabbit and cynomolgus monkey plasma treated with FVIII function-neutralizing antibodies. CONCLUSIONS: TPP2009 specifically targets a unique conformational epitope on ATß*H and blocks ATß-mediated anticoagulation. It effectively promotes coagulation in plasma, indicating the importance of ATß in hemostasis.


Subject(s)
Antibodies/pharmacology , Antithrombin III/metabolism , Blood Coagulation/drug effects , Coagulants/pharmacology , Animals , Antibodies/immunology , Antibodies/metabolism , Antibody Specificity , Antithrombin III/chemistry , Antithrombin III/immunology , Binding Sites, Antibody , Blood Coagulation Tests , Cell Line , Cell Surface Display Techniques , Coagulants/immunology , Coagulants/metabolism , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Epitope Mapping , Humans , Protein Binding , Protein Structure, Secondary , Rabbits , Structure-Activity Relationship , Surface Plasmon Resonance , Time Factors
2.
Gene Ther ; 20(1): 102-11, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22357510

ABSTRACT

Newcastle disease virus (NDV) is an oncolytic paramyxovirus with a nonsegmented single-stranded RNA genome. In this report, a recombinant oncolytic NDV was passaged in human tumor xenografts and reisolated and characterized after two rounds of bioselection. Several isolates could be recovered that differed from the parental virus with respect to virus spread in tumor cells and the ability to form syncytia in human tumor cells. Three isolates were identified that demonstrated superior oncolytic potency compared with the parental virus as measured by increased oncolytic potency in confluent tumor cell monolayers, in tumor cell spheroids and in a mouse xenograft tumor model. The surface proteins F and HN were sequence analyzed and characterized for fusogenicity. The present study demonstrates that in vivo NDV bioselection can enable the isolation of novel, oncolytic NDV and thus represents a powerful methodology for the development of highly potent oncolytic viruses.


Subject(s)
Newcastle disease virus/genetics , Oncolytic Viruses/genetics , Selection, Genetic , Animals , Cell Line, Tumor , Giant Cells , Hemagglutinins/genetics , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Newcastle disease virus/isolation & purification , Oncolytic Viruses/isolation & purification , Transplantation, Heterologous , Viral Fusion Proteins/genetics
3.
Gene Ther ; 16(9): 1169-74, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19458647

ABSTRACT

We have generated a novel oncolytic Adenovirus (Ad), ColoAd1, with significantly increased potency ( approximately 100-fold) relative to its parent viruses, Ad11p and Ad3, or to the clinically tested oncolytic Ad, ONYX-015. Although this agent has a significant increase in its therapeutic window relative to ONYX-015 or its parent viruses, its ability to intervene and control virotherapy in treated patient is an important safety consideration for a novel biological therapy, such as ColoAd1. As there are no approved treatments for Ad infections, we sought to define whether antivirals being used to experimentally treat Ad infections (cidofovir (CDV), ribavirin) had any activity against ColoAd1. In addition, we incorporated a well-described pro-drug converting enzyme, the herpes simplex virus-thymidine kinase (HSV-TK) gene, into the viral genome to test whether the expression of this enzyme directly from the virus could be exploited as a safety valve for arresting the viral infection in the presence of the pro-drug, ganciclovir. Both the antiviral drug, CDV, and the incorporation of the pro-drug-converting TK enzyme were validated as effective approaches to controlling ColoAd1 infection, and this represents an important advancement in the development of ColoAd1 as an anticancer treatment.


Subject(s)
Adenoviridae Infections/prevention & control , Adenoviridae/pathogenicity , Antiviral Agents/pharmacology , Cytosine/analogs & derivatives , Oncolytic Viruses/pathogenicity , Organophosphonates/pharmacology , Thymidine Kinase/genetics , Adenoviridae/drug effects , Adenoviridae/enzymology , Adenoviridae/genetics , Cell Survival/drug effects , Cells, Cultured , Cidofovir , Cytosine/pharmacology , Genetic Engineering/methods , Humans , Oncolytic Virotherapy/adverse effects , Oncolytic Virotherapy/methods , Oncolytic Viruses/drug effects , Oncolytic Viruses/genetics , Prodrugs/pharmacology , Ribavirin/pharmacology , Thymidine Kinase/metabolism , Tumor Cells, Cultured
4.
Cancer Gene Ther ; 16(2): 184-94, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18758433

ABSTRACT

Therapeutic monoclonal antibodies continue to achieve clinical success for the treatment of many different diseases, particularly cancer. However, the production and purification of antibodies continues to be a time and labor-intensive process with considerable technical challenges. Gene-based delivery of antibodies may address this, via direct production within the host that achieves therapeutic levels. In this report, we validate the feasibility that gene-based delivery is a viable approach for efficacious delivery of antibodies in the preclinical and, presumably, clinical setting. We demonstrate high and sustained in vivo expression of the murine antihuman epidermal growth factor receptor antibody 14E1 following intramuscular delivery by adeno-associated virus (AAV) 2/1. Incorporating the Furin/2A technology for monocistronic expression of both heavy and light chains, we achieved sustained serum levels of full-length 14E1 peaking over 1 mg ml(-1) in athymic nude mice. In the A431 xenograft tumor model, 14E1 was capable of significantly inhibiting tumor growth and prolonging survival when AAV was administered prior to tumor challenge. Furthermore, 14E1 demonstrated significant antitumor efficacy against well-established tumors (approximately 400 mm(3)) when AAV was administered up to 20 days after tumor challenge. Here we demonstrate for the first time growth inhibition of a well-established tumor by a full-length antibody following delivery by AAV.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Dependovirus/genetics , Dependovirus/metabolism , ErbB Receptors/antagonists & inhibitors , Neoplasms, Experimental/therapy , Neoplasms/therapy , Transplantation, Heterologous , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/metabolism , Antineoplastic Agents/therapeutic use , ErbB Receptors/metabolism , Gene Expression Regulation , Gene Transfer Techniques , Genetic Therapy , Humans , Injections, Intramuscular , Male , Mice , Mice, Nude , Neoplasms/metabolism , Neoplasms, Experimental/metabolism , Tumor Cells, Cultured
5.
Gene Ther ; 15(1): 1-11, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17637794

ABSTRACT

A single plasmid regulated expression vector based upon a mifepristone-inducible two plasmid system, termed pBRES, has been constructed and tested in mice using murine interferon-b (mIFNb) as the transgene. The expression of mIFNb in the circulation was followed by measuring the systemic induction of IP-10, a validated biomarker for mIFNb in mice. Long-term, inducible expression of mIFNb was demonstrated following a single intramuscular (i.m.) injection of the pBRES mIFNb plasmid vector into the hind limb of mice. Induction of mIFNb expression was achieved by administration of the small molecule inducer, mifepristone (MFP). Plasmid DNA and mIFNb mRNA levels in the injected muscles correlated with mIFNb expression as monitored by IP-10 over a 3-month time period. Renewable transgene expression was achieved following repeat administration of the plasmid at 3 months following the first plasmid injection. A dose-dependent increase in expression was demonstrated by varying the amount of injected plasmid or the amount of the inducer administered to the mice. Finally, the pBRES plasmid expressing mIFNb under control of the inducer, MFP, was shown to be efficacious in a murine model of experimental allergic encephalomyelitis, supporting the feasibility of gene-based therapeutic approaches for treating diseases such as multiple sclerosis.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/therapy , Gene Expression Regulation , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Interferon-beta/genetics , Plasmids/administration & dosage , Animals , Biomarkers/blood , Chemokine CXCL10/analysis , Disease Progression , Female , Injections, Intramuscular , Interferon-beta/blood , Mice , Mice, Inbred Strains , Mifepristone/administration & dosage , Multiple Sclerosis/therapy , Plasmids/analysis , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Transgenes
6.
Gene Ther ; 15(1): 54-60, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17960164

ABSTRACT

Neutralizing antibodies (nAB) at the time of administration hamper the effectiveness of adeno-associated virus (AAV) as a clinical DNA delivery system. The present study was designed to investigate if AAV re-administration in muscle tissue is dependent on the nAB titer. Recombinant (r)AAV serotype 1, as a promising candidate for targeting skeletal muscle, was used for gene delivery. C57Bl/6 mice were infected intramuscularly with doses between 1 x 10(9) and 5 x 10(10) virus particles (vp) of AAV1-expressing luciferase (AAV1-luc) or human interferon-beta (AAV1-hIFNbeta). Increasing transgene expression was observed over the first 2 months and anti-AAV1 nAB titers peaked between weeks 4 and 8. Six months after the first administration, 5 x 10(10) vp of AAV1-IFNbeta were re-administered. Following re-administration, nAB titers increased but did not significantly affect transgene expression from the AAV vector that had been administered first. In contrast, hIFNbeta expression originating from the second vector administration was significantly diminished and reflected the nAB titer present at the day of re-administration. The present study extends earlier observations that preexisting nAB affects AAV1 re-administration. The level of nAB is proportional to the virus dose used for the first injection and transgene expression following re-administration is dependent on preexisting nAB titer.


Subject(s)
Dependovirus/genetics , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Transduction, Genetic/methods , Animals , Antibodies/analysis , Antibody Formation , Dependovirus/immunology , Gene Expression , Genetic Engineering , Genetic Vectors/immunology , Humans , Injections, Intramuscular , Interferon-beta/genetics , Interferon-beta/immunology , Luciferases/genetics , Male , Mice , Mice, Inbred C57BL , Time Factors , Transgenes , Viral Load
7.
Gene Ther ; 14(2): 180-4, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17024108

ABSTRACT

Accurate adenovirus (Ad) quantification requires labor- and time-intensive viral stock purification. While crude viral lysates can be titered by plaque assay, this cell-based assay is neither rapid nor accurate. Consequently, a method for quantification of crude, unpurified viral culture lysates is needed. Given growing interest in alternative Ad serotypes (different from well-studied and characterized serotype Ad5) for basic research and for therapeutic applications, such a method should also apply to alternative serotypes. Using a Q Sepharose XL (QSXL) column-based method, we describe a robust quantification method resulting in efficient retention of viral particles of all serotypes, while non-viral components of crude infected cultures remain largely in the flow-through. The high-performance liquid chromatography-QSXL method allows rapid, accurate adenoviral quantification in crude lysates as well as identification of the various serotypes present in mixed-serotype crude lysates. We also report on conditions that efficiently strip and regenerate the column, extending its functional life.


Subject(s)
Adenoviridae/isolation & purification , Chromatography, High Pressure Liquid/methods , Genetic Therapy/methods , Serotyping/methods , Virion
8.
Gene Ther ; 13(12): 986-90, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16525485

ABSTRACT

Replicating, tumor selective viruses are being tested as potential treatments for human cancers. Hypoxia is a pathophysiological cancer condition that alters the lytic potential of the replication-competent adenovirus serotype 5 (Ad5) virus by a mechanism independent of receptor levels or internalization rates. We extend these initial studies to examine the potential effects of hypoxia on the group B adenoviruses (Ads), adenovirus type 3 (Ad3) (group B1) and adenovirus type 11p (Ad11p) (group B2). Receptor expression (CD46) is not altered by hypoxia. However, the lytic potential is compromised in a cell-dependent fashion. Consequently, our study suggests that group B replicating Ad-based treatments, like the group C Ad-5-based viruses, will need to be modified in order to effectively treat hypoxic components of human tumors.


Subject(s)
Adenoviridae Infections/virology , Adenoviruses, Human/physiology , Genetic Therapy/methods , Hematologic Neoplasms/therapy , Hypoxia/virology , Oncolytic Virotherapy/methods , Adenoviruses, Human/genetics , Cytopathogenic Effect, Viral , Flow Cytometry , Genotype , Hematologic Neoplasms/metabolism , Hematologic Neoplasms/virology , Humans , Membrane Cofactor Protein/metabolism , Virus Replication
9.
Gene Ther ; 12(11): 902-10, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15690062

ABSTRACT

Oxygen deprivation (hypoxia) is a common feature of various human maladies, including cardiovascular diseases and cancer; however, the effect of hypoxia on Ad-based gene therapies has not been described. In this study, we evaluated how hypoxia (1% pO(2)) affects different aspects of Ad-based therapies, including attachment and uptake, transgene expression, and replication, in a series of cancer cell lines and primary normal cells. We found that hypoxia had no significant effect on the expression or function of the Ad5 attachment (Coxsackievirus and Adenovirus Receptor) and internalization (alpha(v) integrins) proteins, nor on the human cytomegalovirus-driven expression of an exogenous gene carried by a replication-incompetent Ad. Viral replication, however, was compromised by hypoxic conditions. Our studies revealed hypoxia-induced reductions in E1A levels that were mediated at the post-transcriptional level. E1A drives cells into the viral replication optimal S phase of the cell cycle; consequently, the combination of reduced E1A protein and hypoxia-induced G1 arrest of cells may be responsible for the lack of efficient viral replication under hypoxic conditions. Consequently, while traditional replication-incompetent Ad-based vectors appear to be viable delivery systems for hypoxia-associated disease indications, our studies suggest that Oncolytic Ads may need additional factors to efficiently treat hypoxic regions of human tumors.


Subject(s)
Adenovirus Infections, Human/physiopathology , Adenoviruses, Human/genetics , Transgenes , Virus Replication , Adenovirus E1A Proteins/metabolism , Adenovirus Infections, Human/virology , Adenoviruses, Human/physiology , Blotting, Western , Cell Hypoxia , Cell Line , G1 Phase , Gene Expression Regulation , Genetic Therapy/methods , Humans , Receptors, Virus/metabolism , Tumor Cells, Cultured
10.
Gene Ther ; 11(16): 1256-63, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15215884

ABSTRACT

Systemic delivery of adenoviral vectors is a major goal in cancer gene therapy, but is currently prohibited by rapid hepatic uptake of virus following intravenous injection with levels of viable virus in the murine plasma typically falling to less than 0.1% after 30 min. We have used a surface-masking technique based on multivalent copolymers of poly(N-(2-hydroxypropyl)methacrylamide) to ablate all pathways of receptor-mediated infection, combined with dose modulation to achieve partial saturation of nonspecific uptake pathways. Polymer coating gave at least 100-fold decreased hepatic transgene expression at all doses and even high doses of coated virus (pc-virus) showed no weight loss or stimulation of serum transaminases. Low doses of virus and pc-virus (10(9) viral particles (vp)/mouse) were mainly captured by the liver (assessed by quantitative PCR), although higher doses led to greater fractional persistence in the plasma (measured after 30 min). Coated virus at a dose of 6 x 10(11) vp/mouse showed nearly 50% plasma circulation, representing a 3.5-fold greater area under the concentration-time curve (0-30 min) compared to unmodified virus. Such an increase in the bioavailability of adenovirus, coupled with substantial decreases in toxicity and unwanted transgene expression is an important step towards producing systemically available tumour-targeted viruses.


Subject(s)
Adenoviridae , Genetic Vectors/genetics , Liver/virology , Plasma/virology , Polymers , Animals , Gene Expression , Gene Targeting/methods , Genetic Therapy/methods , Injections, Intravenous , Mice , Neoplasms/therapy , Phagocytosis/physiology , Receptors, Virus/genetics , Recombination, Genetic/genetics , Transduction, Genetic/methods , Transgenes/genetics , Tumor Cells, Cultured
11.
J Virol ; 75(19): 8875-87, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11533151

ABSTRACT

Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) induces apoptosis through two receptors, TRAIL-R1 (also known as death receptor 4) and TRAIL-R2 (also known as death receptor 5), that are members of the TNF receptor superfamily of death domain-containing receptors. We show that human adenovirus type 5 encodes three proteins, named RID (previously named E3-10.4K/14.5K), E3-14.7K, and E1B-19K, that independently inhibit TRAIL-induced apoptosis of infected human cells. This conclusion was derived from studies using wild-type adenovirus, adenovirus replication-competent mutants that lack one or more of the RID, E3-14.7K, and E1B-19K genes, and adenovirus E1-minus replication-defective vectors that express all E3 genes, RID plus E3-14.7K only, RID only, or E3-14.7K only. RID inhibits TRAIL-induced apoptosis when cells are sensitized to TRAIL either by adenovirus infection or treatment with cycloheximide. RID induces the internalization of TRAIL-R1 from the cell surface, as shown by flow cytometry and indirect immunofluorescence for TRAIL-R1. TRAIL-R1 was internalized in distinct vesicles which are very likely to be endosomes and lysosomes. TRAIL-R1 is degraded, as indicated by the disappearance of the TRAIL-R1 immunofluorescence signal. Degradation was inhibited by bafilomycin A1, a drug that prevents acidification of vesicles and the sorting of receptors from late endosomes to lysosomes, implying that degradation occurs in lysosomes. RID was also shown previously to internalize and degrade another death domain receptor, Fas, and to prevent apoptosis through Fas and the TNF receptor. RID was shown previously to force the internalization and degradation of the epidermal growth factor receptor. E1B-19K was shown previously to block apoptosis through Fas, and both E1B-19K and E3-14.7K were found to prevent apoptosis through the TNF receptor. These findings suggest that the receptors for TRAIL, Fas ligand, and TNF play a role in limiting virus infections. The ability of adenovirus to inhibit killing through these receptors may prolong acute and persistent infections.


Subject(s)
Adenoviridae Infections/virology , Adenoviridae/physiology , Adenovirus E3 Proteins/physiology , Apoptosis/physiology , Membrane Glycoproteins/physiology , Receptors, Tumor Necrosis Factor/physiology , Tumor Necrosis Factor-alpha/physiology , Adenoviridae Infections/metabolism , Adenoviridae Infections/pathology , Apoptosis Regulatory Proteins , Cell Line , Humans , Ligands , Receptors, TNF-Related Apoptosis-Inducing Ligand , TNF-Related Apoptosis-Inducing Ligand , Virus Replication
12.
Gene Ther ; 8(15): 1123-31, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11509942

ABSTRACT

The use of genetically engineered, replication-selective viruses to treat cancer is being realized with viruses such as ONYX-015, a human adenovirus that selectively destroys p53 mutant cancer cells. To enhance further the clinical efficacy of ONYX-015 and viruses like it, we have developed a novel gene delivery system for replicating adenoviruses. This system has two unique features. First, it uses the endogenous adenoviral gene expression machinery (promoter, splicing, polyadenylation) to drive transgene expression. Second, a single region or gene in the multi-gene E3 transcription unit is selectively substituted for by the therapeutic transgene(s). Analyzing various transgene substitutions for the 6.7 K/gp19 K region of E3, we demonstrate the following: (1) transgene expression in this system is predictable and mimics the substituted endogenous gene expression pattern, (2) expression of surrounding E3 genes can be retained, (3) the insertion site choice can effect both the transgene expression level and the viral life cycle, and, (4) expression levels from this system are superior to those generated from a replication-defective virus using the HCMV enhancer-promoter and this is dependent on viral DNA replication. This unique methodology has broad application to the rapidly evolving field of replicating virus-based therapies.


Subject(s)
Adenovirus E3 Proteins/genetics , Genetic Therapy/methods , Genetic Vectors/genetics , Mutagenesis, Insertional/methods , Nucleoside Deaminases/genetics , Tumor Necrosis Factor-alpha/genetics , Adenoviruses, Human/genetics , Blotting, Western , Cell Line , Cytosine Deaminase , Gene Expression , Genes, p16 , Humans , Nucleoside Deaminases/analysis , Transfection/methods , Transgenes , Tumor Necrosis Factor-alpha/analysis
13.
Gene Ther ; 8(15): 1132-41, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11509943

ABSTRACT

Genetically modified replication-selective human adenoviruses are currently undergoing testing in the clinical setting as anticancer agents. Coupling the lytic function of these viruses with virus-mediated transgene delivery represents a powerful extension of this treatment. We have designed a unique system for gene delivery from the replicating virus. It takes advantage of the endogenous gene expression control sequences (promoter, splicing, polyadenylation signals) to efficiently and predictably deliver transgenes from the non-essential E3 transcription unit while still maintaining the expression of the remaining E3 genes in the multi-gene transcription unit. In this article, we engineered restriction enzyme sites into the virus genome selectively to delete the ADP gene and replace it with the therapeutic transgenes CD and TNFalpha. We demonstrate that: (1) transgene expression from this region mirrors the substituted ADP gene; (2) the loss of ADP in these viruses results in infected cells with extended viability and protein synthesis when compared with a wild-type Ad5 infected cell; and (3) expression of surrounding E3 genes can be maintained in such a system. The potential advantages of delivering transgenes from the ADP region of the replicating adenovirus are discussed.


Subject(s)
Adenosine Diphosphate/genetics , Adenovirus E3 Proteins/genetics , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Mutagenesis, Insertional/methods , Transfection/methods , Adenoviruses, Human , Cytosine Deaminase , Enzyme-Linked Immunosorbent Assay/methods , Gene Deletion , Gene Expression , Humans , Nucleoside Deaminases/genetics , Tumor Necrosis Factor-alpha/analysis , Tumor Necrosis Factor-alpha/genetics
14.
Gene Ther ; 8(15): 1142-8, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11509944

ABSTRACT

Successful therapies for cancer need to deal with the complexity associated with the human tumor. Studies of tumor and viral biology have progressed to a point where replicating viruses are now being engineered as potential treatments for human cancers. The complex nature of human cancers dictates that successful treatments will require combination therapies. To this end, we have focused on developing the gene delivery capacity of the replicating adenovirus, using the non-essential E3 region transcription unit as a target site for therapeutic transgene insertions. Utilizing the endogenous expression machinery of the E3 region (promoter, splicing, polyA) we show that a therapeutic transgene, TNF, is efficiently expressed from the E3B region and with exclusive late gene expression kinetics. Potential clinical applications are discussed.


Subject(s)
Adenovirus E3 Proteins/genetics , Genetic Therapy/methods , Genetic Vectors/genetics , Mutagenesis, Insertional/methods , Neoplasms/therapy , Transfection/methods , Base Sequence , Blotting, Western/methods , Cell Line , Gene Expression , Humans , Molecular Sequence Data , Tumor Necrosis Factor-alpha/genetics , Virus Replication
15.
J Virol ; 75(9): 4297-307, 2001 May.
Article in English | MEDLINE | ID: mdl-11287579

ABSTRACT

The E1B-55K protein plays an important role during human adenovirus type 5 productive infection. In the early phase of the viral infection, E1B-55K binds to and inactivates the tumor suppressor protein p53, allowing efficient replication of the virus. During the late phase of infection, E1B-55K is required for efficient nucleocytoplasmic transport and translation of late viral mRNAs, as well as for host cell shutoff. In an effort to separate the p53 binding and inactivation function and the late functions of the E1B-55K protein, we have generated 26 single-amino-acid mutations in the E1B-55K protein. These mutants were characterized for their ability to modulate the p53 level, interact with the E4orf6 protein, mediate viral late-gene expression, and support virus replication in human cancer cells. Of the 26 mutants, 24 can mediate p53 degradation as efficiently as the wild-type protein. Two mutants, R240A (ONYX-051) and H260A (ONYX-053), failed to degrade p53 in the infected cells. In vitro binding assays indicated that R240A and H260A bound p53 poorly compared to the wild-type protein. When interaction with another viral protein, E4orf6, was examined, H260A significantly lost its ability to bind E4orf6, while R240A was fully functional in this interaction. Another mutant, T255A, lost the ability to bind E4orf6, but unexpectedly, viral late-gene expression was not affected. This raised the possibility that the interaction between E1B-55K and E4orf6 was not required for efficient viral mRNA transport. Both R240A and H260A have retained, at least partially, the late functions of wild-type E1B-55K, as determined by the expression of viral late proteins, host cell shutoff, and lack of a cold-sensitive phenotype. Virus expressing R240A (ONYX-051) replicated very efficiently in human cancer cells, while virus expressing H260A (ONYX-053) was attenuated compared to wild-type virus dl309 but was more active than ONYX-015. The ability to separate the p53-inactivation activity and the late functions of E1B-55K raises the possibility of generating adenovirus variants that retain the tumor selectivity of ONYX-015 but can replicate more efficiently than ONYX-015 in a broad spectrum of cell types.


Subject(s)
Adenovirus E1B Proteins/metabolism , Adenovirus E4 Proteins/metabolism , Adenoviruses, Human/metabolism , Tumor Suppressor Protein p53/metabolism , Viral Proteins/metabolism , Adenovirus E1B Proteins/genetics , Adenoviruses, Human/genetics , Amino Acid Sequence , Amino Acid Substitution , Base Sequence , DNA, Viral , Fluorescent Antibody Technique, Indirect , Humans , Molecular Sequence Data , Staining and Labeling/methods , Tumor Cells, Cultured , Viral Proteins/genetics
16.
Nat Med ; 6(10): 1134-9, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11017145

ABSTRACT

Replication-selective oncolytic viruses constitute a rapidly evolving and new treatment platform for cancer. Gene-deleted viruses have been engineered for tumor selectivity, but these gene deletions also reduce the anti-cancer potency of the viruses. We have identified an E1A mutant adenovirus, dl922-947, that replicates in and lyses a broad range of cancer cells with abnormalities in cell-cycle checkpoints. This mutant demonstrated reduced S-phase induction and replication in non-proliferating normal cells, and superior in vivo potency relative to other gene-deleted adenoviruses. In some cancers, its potency was superior to even wild-type adenovirus. Intravenous administration reduced the incidence of metastases in a breast tumor xenograft model. dl922-947 holds promise as a potent, replication-selective virus for the local and systemic treatment of cancer.


Subject(s)
Adenoviridae/genetics , Adenovirus E1A Proteins/genetics , Antineoplastic Agents/pharmacology , Genetic Vectors/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Drug Screening Assays, Antitumor , Female , Genetic Vectors/administration & dosage , Humans , Injections, Intralesional , Injections, Intravenous , Mice , Mice, Nude , Neoplasm Metastasis/drug therapy , Neoplasms/drug therapy , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
20.
Hum Gene Ther ; 9(13): 1885-98, 1998 Sep 01.
Article in English | MEDLINE | ID: mdl-9741427

ABSTRACT

Herein, we report that the adenovirus E3-14.7K protein inhibits the inflammatory response to adenovirus in transgenic mice in which the E3-14.7K gene was selectively expressed in the respiratory epithelium, using the human surfactant protein C (SP-C) promoter. E3-14.7K mRNA and protein were detected specifically in the lungs of SPC/E3-14.7K transgenic mice. Responses of the transgenic mice to Av1Luc1, an E1-E3-deleted Ad vector encoding the luciferase reporter gene, were examined, including vector transgene expression and lung inflammation. In wild-type mice, luciferase activity declined rapidly and was lost 14 days following Av1Luc1 administration. The loss of luciferase activity was associated with pulmonary infiltration by macrophages and lymphocytes. In heterozygous SPC/E3-14.7K mice, luciferase activity was increased by 7 days compared with control littermates, and pulmonary infiltration by macrophages was decreased. In homozygous (+/+) SPC/E3-14.7K mice, luciferase activity was increased 7, 14, and 21 days following administration compared with wild-type mice, and lung inflammation was markedly reduced. After Av1Luc1 administration, PCNA staining of bronchiolar and alveolar respiratory epithelial cells was decreased in SPC/E3-14.7K transgenic mice, indicating decreased epithelial cell proliferation, a finding consistent with the observed reduction in inflammation. CD4 and CD8 lymphocyte populations were only mildly altered, while humoral responses to adenoviral vectors were unchanged in the SPC/E3-14.7K mice. The E3-14.7K protein expressed selectively in respiratory epithelial cells suppresses Ad-induced pulmonary epithelial cell cytotoxicity and lung inflammation in vivo and prolongs reporter gene expression.


Subject(s)
Adenoviridae/genetics , Adenovirus E3 Proteins/genetics , Genetic Vectors/genetics , Lung/immunology , Adenoviridae/immunology , Adenovirus E3 Proteins/metabolism , Animals , Antibodies, Viral/blood , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Epithelium/metabolism , Gene Expression Regulation/genetics , Genetic Vectors/immunology , Humans , Luciferases/metabolism , Lung/metabolism , Lung/pathology , Macrophages, Alveolar , Mice , Mice, Transgenic , Organ Specificity , Promoter Regions, Genetic/genetics , Proteolipids/genetics , Pulmonary Surfactants/genetics , RNA, Messenger/analysis , Recombinant Fusion Proteins , Transgenes
SELECTION OF CITATIONS
SEARCH DETAIL