Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Vaccine ; 29(15): 2803-9, 2011 Mar 24.
Article in English | MEDLINE | ID: mdl-21320541

ABSTRACT

Chikungunya virus, a mosquito-borne alphavirus, recently caused the largest epidemic ever seen for this virus. Chikungunya disease primarily manifests as a painful and debilitating arthralgia/arthritis, and no effective drug or vaccine is currently available. Here we describe a recombinant chikungunya virus vaccine comprising a non-replicating complex adenovirus vector encoding the structural polyprotein cassette of chikungunya virus. A single immunisation with this vaccine consistently induced high titres of anti-chikungunya virus antibodies that neutralised both an old Asian isolate and a Réunion Island isolate from the recent epidemic. The vaccine also completely protected mice against viraemia and arthritic disease caused by both virus isolates.


Subject(s)
Adenoviridae/genetics , Alphavirus Infections/prevention & control , Arthritis/prevention & control , Chikungunya virus/immunology , Drug Carriers , Viral Vaccines/immunology , Viremia/prevention & control , Alphavirus Infections/immunology , Animals , Antibodies, Viral/blood , Arthritis/immunology , Chikungunya virus/genetics , Female , Genetic Vectors , Mice , Mice, Inbred C57BL , Vaccination/methods , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology , Viral Vaccines/administration & dosage , Viremia/immunology
2.
Clin Vaccine Immunol ; 17(4): 572-81, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20181765

ABSTRACT

Ebola viruses are highly pathogenic viruses that cause outbreaks of hemorrhagic fever in humans and other primates. To meet the need for a vaccine against the several types of Ebola viruses that cause human diseases, we developed a multivalent vaccine candidate (EBO7) that expresses the glycoproteins of Zaire ebolavirus (ZEBOV) and Sudan ebolavirus (SEBOV) in a single complex adenovirus-based vector (CAdVax). We evaluated our vaccine in nonhuman primates against the parenteral and aerosol routes of lethal challenge. EBO7 vaccine provided protection against both Ebola viruses by either route of infection. Significantly, protection against SEBOV given as an aerosol challenge, which has not previously been shown, could be achieved with a boosting vaccination. These results demonstrate the feasibility of creating a robust, multivalent Ebola virus vaccine that would be effective in the event of a natural virus outbreak or biological threat.


Subject(s)
Adenoviridae/genetics , Ebola Vaccines/immunology , Ebolavirus/immunology , Genetic Vectors , Hemorrhagic Fever, Ebola/prevention & control , Animals , Disease Models, Animal , Ebola Vaccines/genetics , Ebolavirus/genetics , Hemorrhagic Fever, Ebola/immunology , Humans , Immunization, Secondary/methods , Macaca fascicularis , Macaca mulatta , Survival Analysis
3.
Clin Vaccine Immunol ; 16(11): 1624-32, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19776190

ABSTRACT

Rift Valley fever virus (RVFV) has been cited as a potential biological-weapon threat due to the serious and fatal disease it causes in humans and animals and the fact that this mosquito-borne virus can be lethal in an aerosolized form. Current human and veterinary vaccines against RVFV, however, are outdated, inefficient, and unsafe. We have incorporated the RVFV glycoprotein genes into a nonreplicating complex adenovirus (CAdVax) vector platform to develop a novel RVFV vaccine. Mice vaccinated with the CAdVax-based vaccine produced potent humoral immune responses and were protected against lethal RVFV infection. Additionally, protection was elicited in mice despite preexisting immunity to the adenovirus vector.


Subject(s)
Adenoviridae/genetics , Genetic Vectors , Rift Valley Fever/prevention & control , Rift Valley fever virus/immunology , Viral Vaccines/immunology , Animals , Antibodies, Viral/blood , Male , Mice , Rift Valley Fever/immunology , Rift Valley fever virus/genetics , Survival Analysis , Viral Structural Proteins/genetics , Viral Structural Proteins/immunology , Viral Vaccines/genetics
4.
J Virol ; 82(14): 6927-34, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18480438

ABSTRACT

Nearly a third of the human population is at risk of infection with the four serotypes of dengue viruses, and it is estimated that more than 100 million infections occur each year. A licensed vaccine for dengue viruses has become a global health priority. A major challenge to developing a dengue vaccine is the necessity to produce fairly uniform protective immune responses to all four dengue virus serotypes. We have developed two bivalent dengue virus vaccines, using a complex adenovirus vector, by incorporating the genes expressing premembrane (prM) and envelope (E) proteins of dengue virus types 1 and 2 (dengue-1 and -2, respectively) (CAdVax-Den12) or dengue-3 and -4 (CAdVax-Den34). Rhesus macaques were vaccinated by intramuscular inoculation of a tetravalent dengue vaccine formulated by combining the two bivalent vaccine constructs. Vaccinated animals produced high-titer antibodies that neutralized all four serotypes of dengue viruses in vitro. The ability of the vaccine to induce rapid, as well as sustained, protective immune responses was examined with two separate live-virus challenges administered at 4 and 24 weeks after the final vaccination. For both of these virus challenge studies, significant protection from viremia was demonstrated for all four dengue virus serotypes in vaccinated animals. Viremia from dengue-1 and dengue-3 challenges was completely blocked, whereas viremia from dengue-2 and dengue-4 was significantly reduced, as well as delayed, compared to that of control-vaccinated animals. These results demonstrate that the tetravalent dengue vaccine formulation provides significant protection in rhesus macaques against challenge with all four dengue virus serotypes.


Subject(s)
Adenoviridae/genetics , Dengue Vaccines/genetics , Dengue Vaccines/immunology , Dengue Virus/genetics , Dengue Virus/immunology , Dengue/prevention & control , Genetic Vectors , Animals , Antibodies, Viral/blood , Dengue/immunology , Injections, Intramuscular , Macaca mulatta , Neutralization Tests , Viral Structural Proteins/genetics , Viremia/prevention & control
5.
Vaccine ; 26(21): 2627-39, 2008 May 19.
Article in English | MEDLINE | ID: mdl-18395306

ABSTRACT

There are legitimate concerns that the highly pathogenic H5N1 avian influenza virus could adapt for human-to-human transmission and cause a pandemic similar to the 1918 "Spanish flu" that killed 50 million people worldwide. We have developed pandemic influenza vaccines by incorporating multiple antigens from both avian and Spanish influenza viruses into complex recombinant adenovirus vectors. In vaccinated mice, these vaccines induced strong humoral and cellular immune responses against pandemic influenza virus antigens, and protected vaccinated mice against lethal H5N1 virus challenge. These results indicate that this multi-antigen, broadly protective vaccine may serve as a safer and more effective approach than traditional methods for development of a pandemic influenza vaccine.


Subject(s)
Antigens, Viral/immunology , Genetic Vectors , Influenza A Virus, H5N1 Subtype/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/prevention & control , Adenoviridae/genetics , Animals , Antibodies, Viral/blood , Body Weight , Male , Mice , Survival Analysis , T-Lymphocytes/immunology
6.
Clin Vaccine Immunol ; 15(3): 460-7, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18216185

ABSTRACT

Filoviruses (Ebola and Marburg viruses) are among the deadliest viruses known to mankind, with mortality rates nearing 90%. These pathogens are highly infectious through contact with infected body fluids and can be easily aerosolized. Additionally, there are currently no licensed vaccines available to prevent filovirus outbreaks. Their high mortality rates and infectious capabilities when aerosolized and the lack of licensed vaccines available to prevent such infectious make Ebola and Marburg viruses serious bioterrorism threats, placing them both on the category A list of bioterrorism agents. Here we describe a panfilovirus vaccine based on a complex adenovirus (CAdVax) technology that expresses multiple antigens from five different filoviruses de novo. Vaccination of nonhuman primates demonstrated 100% protection against infection by two species of Ebola virus and three Marburg virus subtypes, each administered at 1,000 times the lethal dose. This study indicates the feasibility of vaccination against all current filovirus threats in the event of natural hemorrhagic fever outbreak or biological attack.


Subject(s)
Adenoviridae/genetics , Ebola Vaccines , Filoviridae , Genetic Vectors , Hemorrhagic Fever, Ebola/prevention & control , Marburg Virus Disease/prevention & control , Viral Vaccines , Adenoviridae/immunology , Animals , Antigens, Viral/genetics , Antigens, Viral/metabolism , Bioterrorism/prevention & control , Ebola Vaccines/administration & dosage , Ebola Vaccines/genetics , Ebola Vaccines/immunology , Ebolavirus/immunology , Ebolavirus/pathogenicity , Filoviridae/classification , Filoviridae/genetics , Filoviridae/immunology , Hemorrhagic Fever, Ebola/immunology , Humans , Macaca fascicularis , Marburg Virus Disease/immunology , Marburgvirus/classification , Marburgvirus/immunology , Marburgvirus/pathogenicity , Viral Vaccines/administration & dosage , Viral Vaccines/genetics , Viral Vaccines/immunology
7.
BMC Biotechnol ; 7: 9, 2007 Feb 12.
Article in English | MEDLINE | ID: mdl-17295927

ABSTRACT

BACKGROUND: Despite several effective treatment options available for prostate cancer, it remains the second leading cause of cancer death in American men. Thus, there is a great need for new treatments to improve outcomes. One such strategy is to eliminate cancer through the expression of cytotoxic genes specifically in prostate cells by gene therapy vectored delivery. To prevent systemic toxicity, tissue- and/or cancer-specific gene expression is required. However, the use of tissue- or cancer-specific promoters to target transgene expression has been hampered by their weak activity. RESULTS: To address this issue, we have developed a regulation strategy that includes feedback amplification of gene expression along with a differentially suppressible tetracycline regulated expression system (DiSTRES). By differentially suppressing expression of the tetracycline-regulated transcriptional activator (tTA) and silencer (tTS) genes based on the cell origin, this leads to the activation and silencing of the TRE promoter, respectively. In vitro transduction of LNCaP cells with Ad/GFPDiSTRES lead to GFP expression levels that were over 30-fold higher than Ad/CMV-GFP. Furthermore, Ad/FasL-GFPDiSTRES demonstrated cytotoxic effects in prostate cancer cells known to be resistant to Fas-mediated apoptosis. CONCLUSION: Prostate-specific regulation from the DiSTRES system, therefore, serves as a promising new regulation strategy for future applications in the field of cancer gene therapy and gene therapy as a whole.


Subject(s)
Androgen-Binding Protein/genetics , Gene Targeting/methods , Genetic Therapy/methods , Neoplasm Proteins/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Transcription Factors/genetics , Cell Line, Tumor , Feedback , Gene Expression Regulation, Neoplastic/genetics , Gene Silencing , Humans , Male , Neoplasm Proteins/genetics , Nucleic Acid Amplification Techniques , Promoter Regions, Genetic , Prostatic Neoplasms/therapy , Protein Engineering
8.
Virology ; 353(2): 324-32, 2006 Sep 30.
Article in English | MEDLINE | ID: mdl-16820184

ABSTRACT

The Marburg virus (MARV), an African filovirus closely related to the Ebola virus, causes a deadly hemorrhagic fever in humans, with up to 90% mortality. Currently, treatment of disease is only supportive, and no vaccines are available to prevent spread of MARV infections. In order to address this need, we have developed and characterized a novel recombinant vaccine that utilizes a single complex adenovirus-vectored vaccine (cAdVax) to overexpress a MARV glycoprotein (GP) fusion protein derived from the Musoke and Ci67 strains of MARV. Vaccination with the cAdVaxM(fus) vaccine led to efficient production of MARV-specific antibodies in both mice and guinea pigs. Significantly, guinea pigs vaccinated with at least 5 x 10(7) pfu of cAdVaxM(fus) vaccine were 100% protected against lethal challenges by the Musoke, Ci67 and Ravn strains of MARV, making it a vaccine with trivalent protective efficacy. Therefore, the cAdVaxM(fus) vaccine serves as a promising vaccine candidate to prevent and contain multi-strain infections by MARV.


Subject(s)
Adenoviridae/metabolism , Antigens, Viral/biosynthesis , Ebola Vaccines/administration & dosage , Genetic Vectors/metabolism , Marburg Virus Disease/prevention & control , Marburgvirus/immunology , Vaccination , Viral Envelope Proteins/biosynthesis , Viral Fusion Proteins/biosynthesis , Adenoviridae/genetics , Amino Acid Sequence , Animals , Antibodies, Viral/blood , Antigens, Viral/genetics , Cell Line , Chlorocebus aethiops , Dose-Response Relationship, Immunologic , Ebola Vaccines/genetics , Genetic Therapy/methods , Guinea Pigs , Humans , Injections, Intraperitoneal , Injections, Subcutaneous , Marburg Virus Disease/blood , Marburg Virus Disease/immunology , Mice , Molecular Sequence Data , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Sequence Alignment , Vaccines, Synthetic/administration & dosage , Viral Envelope Proteins/genetics , Viral Fusion Proteins/genetics
9.
Vaccine ; 24(15): 2975-86, 2006 Apr 05.
Article in English | MEDLINE | ID: mdl-16530297

ABSTRACT

Marburg virus (MARV) is an African filovirus that causes a deadly hemorrhagic fever in humans, with up to 90% mortality. Currently, there are no MARV vaccines or therapies approved for human use. We hypothesized that developing a vaccine that induces a de novo synthesis of MARV antigens in vivo will lead to strong induction of both a humoral and cell-mediated immune response against MARV. Here, we develop and characterize three novel gene-based vaccine candidates which express the viral glycoprotein (GP) from either the Ci67, Ravn or Musoke strain of MARV. Immunization of mice with complex adenovirus (Ad)-based vaccine candidates (cAdVax vaccines), led to efficient production of both antibodies and cytotoxic T lymphocytes (CTL) specific to Musoke strain GP and Ci67 strain GP, respectively. Antibody responses were also shown to be cross-reactive across the MARV strains, but not cross-reactive to Ebola virus, a related filovirus. Additionally, three 1 x 10(8)pfu doses of vaccine vector were demonstrated to be safe in mice, as this did not lead to any detectable toxicity in liver or spleen. These promising results indicate that a cAdVax-based vaccine could be effective for induction of both humoral and cell-mediated immune responses to multiple strains of the Marburg virus.


Subject(s)
Adenoviridae/genetics , Marburg Virus Disease/prevention & control , Marburgvirus/genetics , Viral Envelope Proteins/immunology , Viral Vaccines/immunology , Animals , Antibodies, Viral/blood , Antigens, Viral/biosynthesis , Antigens, Viral/genetics , Antigens, Viral/immunology , Cross Reactions , Cytotoxicity Tests, Immunologic , Enzyme-Linked Immunosorbent Assay , Genetic Vectors , Humans , Liver/pathology , Marburg Virus Disease/immunology , Marburgvirus/immunology , Mice , Mice, Inbred C57BL , Models, Animal , Spleen/pathology , T-Lymphocytes, Cytotoxic/immunology , Viral Envelope Proteins/biosynthesis , Viral Envelope Proteins/genetics , Viral Vaccines/administration & dosage , Viral Vaccines/adverse effects , Viral Vaccines/genetics
10.
J Virol ; 80(6): 2738-46, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16501083

ABSTRACT

Ebola virus (EBOV) causes a severe hemorrhagic fever for which there are currently no vaccines or effective treatments. While lethal human outbreaks have so far been restricted to sub-Saharan Africa, the potential exploitation of EBOV as a biological weapon cannot be ignored. Two species of EBOV, Sudan ebolavirus (SEBOV) and Zaire ebolavirus (ZEBOV), have been responsible for all of the deadly human outbreaks resulting from this virus. Therefore, it is important to develop a vaccine that can prevent infection by both lethal species. Here, we describe the bivalent cAdVaxE(GPs/z) vaccine, which includes the SEBOV glycoprotein (GP) and ZEBOV GP genes together in a single complex adenovirus-based vaccine (cAdVax) vector. Vaccination of mice with the bivalent cAdVaxE(GPs/z) vaccine led to efficient induction of EBOV-specific antibody and cell-mediated immune responses to both species of EBOV. In addition, the cAdVax technology demonstrated induction of a 100% protective immune response in mice, as all vaccinated C57BL/6 and BALB/c mice survived challenge with a lethal dose of ZEBOV (30,000 times the 50% lethal dose). This study demonstrates the potential efficacy of a bivalent EBOV vaccine based on a cAdVax vaccine vector design.


Subject(s)
Adenoviridae/genetics , Ebola Vaccines/administration & dosage , Hemorrhagic Fever, Ebola/immunology , Hemorrhagic Fever, Ebola/prevention & control , Viral Envelope Proteins/immunology , Adenoviridae/metabolism , Animals , Antibodies, Viral/blood , Cell Line , Ebola Vaccines/genetics , Ebola Vaccines/immunology , Ebolavirus/genetics , Ebolavirus/immunology , Ebolavirus/pathogenicity , HeLa Cells , Hemorrhagic Fever, Ebola/virology , Humans , Immunity, Cellular , Immunization , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Species Specificity , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism
11.
Future Oncol ; 1(3): 361-73, 2005 Jun.
Article in English | MEDLINE | ID: mdl-16556010

ABSTRACT

As gene therapy evolved over the years, it soon became evident that cancer would be a suitable target for this developing therapeutic area/field. Vector genomes can be manipulated to express therapeutic transgenes, which can lead to either a direct or indirect destruction of tumor cells. Gene therapy for cancer continues to evolve today. Current transgene strategies and their mechanisms of action will be reviewed here. In addition, approaches for targeting transgene expression using both transductional and transcriptional targeting strategies will be discussed. Finally, novel methods for improving tumor-targeted gene expression will be introduced. These new strategies incorporate an amplification feedback loop element to enhance tumor-restricted gene expression.


Subject(s)
Genetic Therapy , Genetic Vectors , Neoplasms/therapy , Animals , Gene Transfer Techniques , Humans , Transgenes/physiology
12.
Cancer Gene Ther ; 10(11): 814-22, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14605667

ABSTRACT

Brain tumors (BTs) are among the most malignant forms of human cancer. Unfortunately, current treatments are often ineffective and produce severe side effects. Cytotoxic gene therapy is an alternative treatment strategy, with the potential advantages of reduced toxicity to normal brain tissue. Apoptosis-inducing "death ligands" Fas ligand and TNF-related apoptosis-inducing ligand (TRAIL) are genes with substantial cytotoxic activity in susceptible tumor cells. Here, we compared the effectiveness of Ad vector-mediated delivery of Fas ligand-green fluorescent protein (FasL-GFP) fusion protein, human TRAIL, and both genes simultaneously. We examined a panel of 13 cell lines (eight derived from primary isolates) for susceptibility to Ad5-based vector infection and for sensitivity to FasL- and TRAIL-mediated apoptosis. All cell lines were efficiently transduced, but, as expected, varied in their sensitivity to ligand-induced apoptosis. Generally, sensitivity to FasL-GFP correlated with cell surface FasR levels, but no such correlation was seen for TRAIL and its functional receptors, DR4 and DR5. The vector expressing both FasL-GFP and TRAIL was more effective than either of the single-gene vectors at comparable transduction levels, and it was effective against a broader range of cell lines. In five cell lines, coexpression resulted in apoptosis levels greater than those predicted for strictly additive activity of the two death ligands. We believe that Ad vector-mediated delivery of multiple death ligands may be developed as a potential BT therapy, either alone or in conjunction with surgical resection of the primary tumor.


Subject(s)
Apoptosis/genetics , Brain Neoplasms/pathology , Genetic Therapy/methods , Glioma/pathology , Intracellular Signaling Peptides and Proteins , Membrane Glycoproteins/genetics , Tumor Necrosis Factor-alpha/genetics , Adenoviridae/genetics , Apoptosis Regulatory Proteins , Astrocytoma/genetics , Astrocytoma/pathology , Astrocytoma/therapy , Brain Neoplasms/genetics , Brain Neoplasms/therapy , CASP8 and FADD-Like Apoptosis Regulating Protein , Carrier Proteins/metabolism , Caspase 8 , Caspases/metabolism , Cell Line , Fas Ligand Protein , Genetic Vectors/genetics , Glioma/genetics , Glioma/therapy , Green Fluorescent Proteins , Humans , Luminescent Proteins/genetics , Protein Isoforms , Receptors, TNF-Related Apoptosis-Inducing Ligand , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , TNF-Related Apoptosis-Inducing Ligand , Transduction, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...