Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Mol Ther ; 29(3): 1186-1198, 2021 03 03.
Article in English | MEDLINE | ID: mdl-33278563

ABSTRACT

Historically poor clinical results of tumor vaccines have been attributed to weakly immunogenic antigen targets, limited specificity, and vaccine platforms that fail to induce high-quality polyfunctional T cells, central to mediating cellular immunity. We show here that the combination of antigen selection, construct design, and a robust vaccine platform based on the Synthetically Modified Alpha Replicon RNA Technology (SMARRT), a self-replicating RNA, leads to control of tumor growth in mice. Therapeutic immunization with SMARRT replicon-based vaccines expressing tumor-specific neoantigens or tumor-associated antigen were able to generate polyfunctional CD4+ and CD8+ T cell responses in mice. Additionally, checkpoint inhibitors, or co-administration of cytokine also expressed from the SMARRT platform, synergized to enhance responses further. Lastly, SMARRT-based immunization of non-human primates was able to elicit high-quality T cell responses, demonstrating translatability and clinical feasibility of synthetic replicon technology for therapeutic oncology vaccines.


Subject(s)
Antigens, Neoplasm/immunology , CD4-Positive T-Lymphocytes/immunology , Cancer Vaccines/administration & dosage , Colonic Neoplasms/therapy , Immunity, Cellular/immunology , Replicon , Animals , Cancer Vaccines/immunology , Colonic Neoplasms/genetics , Colonic Neoplasms/immunology , Female , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Primates , Tumor Cells, Cultured , Vaccination
2.
Nanomedicine ; 24: 102154, 2020 02.
Article in English | MEDLINE | ID: mdl-31982617

ABSTRACT

In vivo delivery of large RNA molecules has significant implications for novel gene therapy, biologics delivery, and vaccine applications. We have developed cationic nanolipoprotein particles (NLPs) to enhance the complexation and delivery of large self-amplifying mRNAs (replicons) in vivo. NLPs are high-density lipoprotein (HDL) mimetics, comprised of a discoidal lipid bilayer stabilized by apolipoproteins that are readily functionalized to provide a versatile delivery platform. Herein, we systematically screened NLP assembly with a wide range of lipidic and apolipoprotein constituents, using biophysical metrics to identify lead candidates for in vivo RNA delivery. NLPs formulated with cationic lipids successfully complexed with RNA replicons encoding luciferase, provided measurable protection from RNase degradation, and promoted replicon in vivo expression. The NLP complexation of the replicon and in vivo transfection efficiency were further enhanced by modulating the type and percentage of cationic lipid, the ratio of cationic NLP to replicon, and by incorporating additive molecules.


Subject(s)
Lipoproteins, HDL/metabolism , RNA, Messenger/metabolism , Apolipoproteins/chemistry , Apolipoproteins/metabolism , Biomimetics , Lipid Bilayers/chemistry , Lipoproteins, HDL/chemistry , RNA, Messenger/chemistry , Replicon/genetics
3.
Nat Biotechnol ; 35(7): 672-675, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28553942

ABSTRACT

Manufacturing processes for biological molecules in the research laboratory have failed to keep pace with the rapid advances in automization and parellelization. We report the development of a digital-to-biological converter for fully automated, versatile and demand-based production of functional biologics starting from DNA sequence information. Specifically, DNA templates, RNA molecules, proteins and viral particles were produced in an automated fashion from digitally transmitted DNA sequences without human intervention.


Subject(s)
Biological Products/chemistry , Biopolymers/chemistry , Genetic Engineering/instrumentation , High-Throughput Nucleotide Sequencing/instrumentation , Robotics/instrumentation , Synthetic Biology/instrumentation , Equipment Design , Equipment Failure Analysis
4.
Expert Rev Vaccines ; 14(2): 283-312, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25382613

ABSTRACT

The advent of reverse genetic approaches to manipulate the genomes of both positive (+) and negative (-) sense RNA viruses allowed researchers to harness these genomes for basic research. Manipulation of positive sense RNA virus genomes occurred first largely because infectious RNA could be transcribed directly from cDNA versions of the RNA genomes. Manipulation of negative strand RNA virus genomes rapidly followed as more sophisticated approaches to provide RNA-dependent RNA polymerase complexes coupled with negative-strand RNA templates were developed. These advances have driven an explosion of RNA virus vaccine vector development. That is, development of approaches to exploit the basic replication and expression strategies of RNA viruses to produce vaccine antigens that have been engineered into their genomes. This study has led to significant preclinical testing of many RNA virus vectors against a wide range of pathogens as well as cancer targets. Multiple RNA virus vectors have advanced through preclinical testing to human clinical evaluation. This review will focus on RNA virus vectors designed to express heterologous genes that are packaged into viral particles and have progressed to clinical testing.


Subject(s)
Genetic Vectors/genetics , RNA Viruses/genetics , RNA, Viral/genetics , Genetic Therapy , Humans , Reverse Genetics , Virion/genetics
5.
J Virol ; 88(20): 12077-86, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25122801

ABSTRACT

Alphavirus replicons were evaluated as potential vaccine candidates for Venezuelan equine encephalitis virus (VEEV), western equine encephalitis virus (WEEV), or eastern equine encephalitis virus (EEEV) when given individually or in combination (V/W/E) to mice or cynomolgus macaques. Individual replicon vaccines or the combination V/W/E replicon vaccine elicited strong neutralizing antibodies in mice to their respective alphavirus. Protection from either subcutaneous or aerosol challenge with VEEV, WEEV, or EEEV was demonstrated out to 12 months after vaccination in mice. Individual replicon vaccines or the combination V/W/E replicon vaccine elicited strong neutralizing antibodies in macaques and demonstrated good protection against aerosol challenge with an epizootic VEEV-IAB virus, Trinidad donkey. Similarly, the EEEV replicon and V/W/E combination vaccine elicited neutralizing antibodies against EEEV and protected against aerosol exposure to a North American variety of EEEV. Both the WEEV replicon and combination V/W/E vaccination, however, elicited poor neutralizing antibodies to WEEV in macaques, and the protection conferred was not as strong. These results demonstrate that a combination V/W/E vaccine is possible for protection against aerosol challenge and that cross-interference between the vaccines is minimal. Importance: Three related viruses belonging to the genus Alphavirus cause severe encephalitis in humans: Venezuelan equine encephalitis virus (VEEV), western equine encephalitis virus (WEEV), and eastern equine encephalitis virus (EEEV). Normally transmitted by mosquitoes, these viruses can cause disease when inhaled, so there is concern that these viruses could be used as biological weapons. Prior reports have suggested that vaccines for these three viruses might interfere with one another. We have developed a combined vaccine for Venezuelan equine encephalitis, western equine encephalitis, and eastern equine encephalitis expressing the surface proteins of all three viruses. In this report we demonstrate in both mice and macaques that this combined vaccine is safe, generates a strong immune response, and protects against aerosol challenge with the viruses that cause Venezuelan equine encephalitis, western equine encephalitis, and eastern equine encephalitis.


Subject(s)
Alphavirus/immunology , Antibodies, Neutralizing/immunology , Encephalitis Virus, Eastern Equine/immunology , Replicon , Viral Vaccines/immunology , Alphavirus/classification , Animals , Blotting, Western , Chlorocebus aethiops , Cricetinae , Encephalitis Virus, Eastern Equine/classification , Enzyme-Linked Immunosorbent Assay , Female , Fluorescent Antibody Technique , Macaca fascicularis , Male , Mice , Vero Cells
6.
J Virol ; 87(9): 4952-64, 2013 May.
Article in English | MEDLINE | ID: mdl-23408633

ABSTRACT

There are no vaccines or therapeutics currently approved for the prevention or treatment of ebolavirus infection. Previously, a replicon vaccine based on Venezuelan equine encephalitis virus (VEEV) demonstrated protective efficacy against Marburg virus in nonhuman primates. Here, we report the protective efficacy of Sudan virus (SUDV)- and Ebola virus (EBOV)-specific VEEV replicon particle (VRP) vaccines in nonhuman primates. VRP vaccines were developed to express the glycoprotein (GP) of either SUDV or EBOV. A single intramuscular vaccination of cynomolgus macaques with VRP expressing SUDV GP provided complete protection against intramuscular challenge with SUDV. Vaccination against SUDV and subsequent survival of SUDV challenge did not fully protect cynomolgus macaques against intramuscular EBOV back-challenge. However, a single simultaneous intramuscular vaccination with VRP expressing SUDV GP combined with VRP expressing EBOV GP did provide complete protection against intramuscular challenge with either SUDV or EBOV in cynomolgus macaques. Finally, intramuscular vaccination with VRP expressing SUDV GP completely protected cynomolgus macaques when challenged with aerosolized SUDV, although complete protection against aerosol challenge required two vaccinations with this vaccine.


Subject(s)
Ebolavirus/immunology , Encephalitis Virus, Venezuelan Equine/genetics , Hemorrhagic Fever, Ebola/prevention & control , Replicon , Viral Vaccines/immunology , Animals , Antibodies, Viral/immunology , Ebolavirus/genetics , Encephalitis Virus, Venezuelan Equine/physiology , Genetic Vectors/genetics , Genetic Vectors/physiology , Hemorrhagic Fever, Ebola/immunology , Hemorrhagic Fever, Ebola/virology , Humans , Macaca fascicularis , Vaccination , Viral Envelope Proteins/administration & dosage , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/genetics
7.
Anim Health Res Rev ; 13(1): 1-9, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22436454

ABSTRACT

The alphavirus replicon technology has been utilized for many years to develop vaccines for both veterinary and human applications. Many developments have been made to the replicon platform recently, resulting in improved safety and efficacy of replicon particle (RP) vaccines. This review provides a broad overview of the replicon technology and safety features of the system and discusses the current literature on RP and replicon-based vaccines.


Subject(s)
Alphavirus Infections/prevention & control , Alphavirus/genetics , Alphavirus/immunology , Replicon , Viral Vaccines/genetics , Alphavirus/physiology , Animals , Dendritic Cells/immunology , Dendritic Cells/physiology , Dendritic Cells/virology , Gene Expression Regulation, Viral , Humans , Vaccines, DNA/genetics , Vaccines, DNA/immunology , Vaccines, DNA/standards , Viral Tropism , Viral Vaccines/immunology , Viral Vaccines/standards
8.
Vaccine ; 30(11): 1944-50, 2012 Mar 02.
Article in English | MEDLINE | ID: mdl-22269873

ABSTRACT

A single-cycle, propagation-defective replicon particle (RP) vaccine expressing a swine influenza virus hemagglutinin (HA) gene was constructed and evaluated in several different animal studies. Studies done in both the intended host (pigs) and non-host (mice) species demonstrated that the RP vaccine is not shed or spread by vaccinated animals to comingled cohorts, nor does it revert to virulence following vaccination. In addition, vaccinated pigs develop both specific humoral and IFN-γ immune responses, and young pigs are protected against homologous influenza virus challenge.


Subject(s)
Alphavirus/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/prevention & control , Replicon , Alphavirus/genetics , Animals , Antibodies, Viral/blood , Antibody Formation , Cytopathogenic Effect, Viral , Female , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Influenza Vaccines/genetics , Interferon-gamma/immunology , Male , Mice , Mice, Inbred BALB C , Orthomyxoviridae/immunology , Orthomyxoviridae/pathogenicity , Orthomyxoviridae Infections/immunology , Swine/immunology , Virulence , Virus Shedding
9.
J Virol ; 84(15): 7713-25, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20504925

ABSTRACT

Alphavirus-based replicon vector systems (family Togaviridae) have been developed as expression vectors with demonstrated potential in vaccine development against both infectious diseases and cancer. The single-cycle nature of virus-like replicon particles (VRP), generated by supplying the structural proteins from separate replicable helper RNAs, is an attractive safety component of these systems. MicroRNAs (miRNAs) have emerged as important cellular RNA regulation elements. Recently, miRNAs have been employed as a mechanism to attenuate or restrict cellular tropism of replication-competent viruses, such as oncolytic adenoviruses, vesicular stomatitis virus, and picornaviruses as well as nonreplicating lentiviral and adenoviral vectors. Here, we describe the incorporation of miRNA-specific target sequences into replicable alphavirus helper RNAs that are used in trans to provide the structural proteins required for VRP production. VRP were found to be efficiently produced using miRNA-targeted helper RNAs if miRNA-specific inhibitors were introduced into cells during VRP production. In the absence of such inhibitors, cellular miRNAs were capable of downregulating helper RNA replication in vitro. When miRNA targets were incorporated into a replicon RNA, cellular miRNAs were capable of downregulating replicon RNA replication upon delivery of VRP into animals, demonstrating activity in vivo. These data provide the first example of miRNA-specific repression of alphavirus replicon and helper RNA replication and demonstrate the feasibility of miRNA targeting of expression vector helper functions that are provided in trans.


Subject(s)
Alphavirus/growth & development , Alphavirus/genetics , Gene Targeting , Genetic Vectors , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Animals , Chlorocebus aethiops , Female , Mice , Mice, Inbred BALB C , RNA, Viral/genetics , RNA, Viral/metabolism , Vero Cells
10.
Vaccine ; 28(2): 494-511, 2009 Dec 11.
Article in English | MEDLINE | ID: mdl-19833247

ABSTRACT

Naturally occurring smallpox was eradicated as a result of successful vaccination campaigns during the 1960s and 1970s. Because of its highly contagious nature and high mortality rate, smallpox has significant potential as a biological weapon. Unfortunately, the current vaccine for orthopoxviruses is contraindicated for large portions of the population. Thus, there is a need for new, safe, and effective orthopoxvirus vaccines. Alphavirus replicon vectors, derived from strains of Venezuelan equine encephalitis virus, are being used to develop alternatives to the current smallpox vaccine. Here, we demonstrated that virus-like replicon particles (VRPs) expressing the vaccinia virus A33R, B5R, A27L, and L1R genes elicited protective immunity in mice comparable to vaccination with live-vaccinia virus. Furthermore, cynomolgus macaques vaccinated with a combination of the four poxvirus VRPs (4pox-VRP) developed antibody responses to each antigen. These antibody responses were able to neutralize and inhibit the spread of both vaccinia virus and monkeypox virus. Macaques vaccinated with 4pox-VRP, flu HA VRP (negative control), or live-vaccinia virus (positive control) were challenged intravenously with 5 x 10(6)pfu of monkeypox virus 1 month after the second VRP vaccination. Four of the six negative control animals succumbed to monkeypox and the remaining two animals demonstrated either severe or grave disease. Importantly, all 10 macaques vaccinated with the 4pox-VRP vaccine survived without developing severe disease. These findings revealed that a single-boost VRP smallpox vaccine shows promise as a safe alternative to the currently licensed live-vaccinia virus smallpox vaccine.


Subject(s)
Alphavirus/genetics , Alphavirus/immunology , Smallpox Vaccine/immunology , Animals , Antibodies, Neutralizing/immunology , Chlorocebus aethiops , Enzyme-Linked Immunosorbent Assay , Female , Macaca , Mice , Mice, Inbred BALB C , Polymerase Chain Reaction , Smallpox Vaccine/genetics , Vero Cells
11.
PLoS One ; 4(8): e6559, 2009 Aug 10.
Article in English | MEDLINE | ID: mdl-19668343

ABSTRACT

Using newer vaccine platforms which have been effective against malaria in rodent models, we tested five immunization regimens against Plasmodium knowlesi in rhesus monkeys. All vaccines included the same four P. knowlesi antigens: the pre-erythrocytic antigens CSP, SSP2, and erythrocytic antigens AMA1, MSP1. We used four vaccine platforms for prime or boost vaccinations: plasmids (DNA), alphavirus replicons (VRP), attenuated adenovirus serotype 5 (Ad), or attenuated poxvirus (Pox). These four platforms combined to produce five different prime/boost vaccine regimens: Pox alone, VRP/Pox, VRP/Ad, Ad/Pox, and DNA/Pox. Five rhesus monkeys were immunized with each regimen, and five Control monkeys received a mock vaccination. The time to complete vaccinations was 420 days. All monkeys were challenged twice with 100 P. knowlesi sporozoites given IV. The first challenge was given 12 days after the last vaccination, and the monkeys receiving the DNA/Pox vaccine were the best protected, with 3/5 monkeys sterilely protected and 1/5 monkeys that self-cured its parasitemia. There was no protection in monkeys that received Pox malaria vaccine alone without previous priming. The second sporozoite challenge was given 4 months after the first. All 4 monkeys that were protected in the first challenge developed malaria in the second challenge. DNA, VRP and Ad5 vaccines all primed monkeys for strong immune responses after the Pox boost. We discuss the high level but short duration of protection in this experiment and the possible benefits of the long interval between prime and boost.


Subject(s)
Macaca mulatta/immunology , Malaria Vaccines/immunology , Malaria/veterinary , Plasmodium knowlesi/immunology , Animals , Antibodies, Protozoan/biosynthesis , Antigens, Protozoan/genetics , Antigens, Protozoan/immunology , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Genetic Vectors , Malaria/parasitology , Malaria/prevention & control , Malaria Vaccines/administration & dosage , T-Lymphocytes/immunology , Viruses/genetics
12.
PLoS One ; 3(7): e2709, 2008 Jul 16.
Article in English | MEDLINE | ID: mdl-18628938

ABSTRACT

BACKGROUND: The Venezuelan equine encephalitis (VEE) virus replicon system was used to produce virus-like replicon particles (VRP) packaged with a number of different VEE-derived glycoprotein (GP) coats. The GP coat is believed to be responsible for the cellular tropism noted for VRP and it is possible that different VEE GP coats may have different affinities for cells. We examined VRP packaged in four different VEE GP coats for their ability to infect cells in vitro and to induce both humoral and cellular immune responses in vivo. METHODOLOGY/PRINCIPAL FINDINGS: The VRP preparations were characterized to determine both infectious units (IU) and genome equivalents (GE) prior to in vivo analysis. VRP packaged with different VEE GP coats demonstrated widely varying GE/IU ratios based on Vero cell infectivity. BALB/c mice were immunized with the different VRP based on equal GE titers and the humoral and cellular responses to the expressed HIV gag gene measured. The magnitude of the immune responses measured in mice revealed small but significant differences between different GP coats when immunization was based on GE titers. CONCLUSIONS/SIGNIFICANCE: We suggest that care should be taken when alternative coat proteins are used to package vector-based systems as the titers determined by cell culture infection may not represent accurate particle numbers and in turn may not accurately represent actual in vivo dose.


Subject(s)
Encephalitis Virus, Venezuelan Equine/metabolism , Replicon , Animals , Encephalomyelitis, Venezuelan Equine/virology , Enzyme-Linked Immunosorbent Assay , Female , Genetic Vectors , Genome , Glycoproteins/chemistry , Immune System , Mice , Mice, Inbred BALB C , Mutation , Reverse Transcriptase Polymerase Chain Reaction
13.
Vaccine ; 25(48): 8180-9, 2007 Nov 23.
Article in English | MEDLINE | ID: mdl-17961878

ABSTRACT

We used a propagation-defective, single-cycle, alphavirus replicon vector system to produce virus-like replicon particles (VRP) expressing the hemagglutinin (HA) and neuraminidase (NA) proteins from influenza A/Wyoming/03/2003 (H3N2). Efficient production methods were scaled to produce pilot lots of HA VRP and NA VRP and clinical lots of HA VRP. HA VRP-induced high-titered antibody responses in mice, rabbits and rhesus macaques, as measured by ELISA or hemagglutination inhibition (HI) assays, and robust cellular immune responses in mice and rhesus macaques, as measured by IFN-gamma ELISPOT. NA VRP also induced cellular immune responses in mice. A toxicology study with HA VRP and NA VRP in rabbits showed no adverse effects in any parameter. These studies support clinical testing of alphavirus replicon vaccines for influenza.


Subject(s)
Alphavirus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza Vaccines/immunology , Neuraminidase/immunology , Animals , Antibodies, Viral , Enzyme-Linked Immunosorbent Assay , Genetic Vectors/genetics , Immunity, Cellular , Influenza Vaccines/genetics , Macaca mulatta , Mice , Rabbits , Replicon
14.
Viral Immunol ; 20(1): 88-104, 2007.
Article in English | MEDLINE | ID: mdl-17425424

ABSTRACT

Dendritic cells (DCs) consist of heterogeneous phenotypic populations and have diverse immunostimulatory functions dependent on both lineage and functional phenotype, but as exceptionally potent antigen-presenting cells, they are targets for generating effective antigen-specific immune responses. A promising replicon particle vector derived from Venezuelan equine encephalitis virus (VEE) has been reported to transduce murine footpad DCs. However, the receptive DC subset, the degree of restriction for this tropism, and the extent of conservation between rodents and humans have not been well characterized. Using fresh peripheral blood DCs, mononuclear cells, monocyte-derived macrophages, and monocyte-derived DCs, our results demonstrate conservation of VEE replicon particle (VRP) tropism for DCs between humans and rodents. We observed that a subset of immature myeloid DCs is the target population, and that VRP-transduced immature DCs retain intact functional capacity, for example, the ability to resist the cytopathic effects of VRP transduction and the capacity to acquire the mature phenotype. These studies support the demonstration of selective VRP tropism for human DCs and provide further insight into the biology of the VRP vector, its parent virus, and human DCs.


Subject(s)
Dendritic Cells/virology , Encephalitis Virus, Venezuelan Equine/genetics , Genetic Vectors/genetics , Replicon , Dendritic Cells/physiology , Encephalitis Virus, Venezuelan Equine/immunology , Humans , Transduction, Genetic , Tropism
15.
Clin Vaccine Immunol ; 14(3): 331-3, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17229882

ABSTRACT

Recombinant severe acute respiratory syndrome (SARS) nucleocapsid and spike protein-based immunoglobulin G immunoassays were developed and evaluated. Our assays demonstrated high sensitivity and specificity to the SARS coronavirus in sera collected from patients as late as 2 years postonset of symptoms. These assays will be useful not only for routine SARS coronavirus diagnostics but also for epidemiological and antibody kinetic studies.


Subject(s)
Antibodies, Viral/blood , Membrane Glycoproteins/immunology , Nucleocapsid Proteins/immunology , Severe Acute Respiratory Syndrome/diagnosis , Severe acute respiratory syndrome-related coronavirus/immunology , Viral Envelope Proteins/immunology , Coronavirus Nucleocapsid Proteins , Enzyme-Linked Immunosorbent Assay , Humans , Recombinant Proteins/immunology , Sensitivity and Specificity , Serologic Tests , Spike Glycoprotein, Coronavirus
16.
Breast Cancer Res Treat ; 82(3): 169-83, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14703064

ABSTRACT

Many tumor-associated antigens (TAAs) represent 'self' antigens and as such, are subject to the constraints of immunologic tolerance. There are significant barriers to eliciting anti-tumor immune responses of sufficient magnitude. We have taken advantage of a Venezuelan equine encephalitis-derived alphavirus replicon vector system with documented in vivo tropism for immune system dendritic cells. We have overcome the intrinsic tolerance to the 'self' TAA rat neu and elicited an effective anti-tumor immune response using this alphavirus replicon vector system and a designed target antigen in a rigorous rat mammary tumor model. We have demonstrated the capacity to generate 50% protection in tumor challenge experiments (p = 0.004) and we have confirmed the establishment of immunologic memory by both second tumor challenge and Winn Assay (p = 0.009). Minor antibody responses were identified and supported the establishment of T helper type 1 (Th1) anti-tumor immune responses by isotype. Animals surviving in excess of 300 days with established effective anti-tumor immunity showed no signs of autoimmune phenomena. Together these experiments support the establishment of T lymphocyte dependent, Th1-biased anti-tumor immune responses to a non-mutated 'self' TAA in an aggressive tumor model. Importantly, this tumor model is subject to the constraints of immunologic tolerance present in animals with normal developmental, temporal, and anatomical expression of a non-mutated TAA. These data support the continued development and potential clinical application of this alphaviral replicon vector system and the use of appropriately designed target antigen sequences for anti-tumor immunotherapy.


Subject(s)
Antigens, Neoplasm/immunology , Disease Models, Animal , Encephalitis Virus, Venezuelan Equine/genetics , Mammary Neoplasms, Experimental/immunology , Replicon/immunology , Vaccines, Synthetic/immunology , Amino Acid Sequence , Animals , Female , Genetic Vectors/immunology , Genetic Vectors/therapeutic use , Humans , Immunization , Mammary Neoplasms, Experimental/therapy , Molecular Sequence Data , Neoplasm Proteins/immunology , Rats , Rats, Inbred F344 , Receptor, ErbB-2/chemistry , Receptor, ErbB-2/genetics , Receptor, ErbB-2/immunology , Self Tolerance
17.
Rev Med Virol ; 12(5): 279-96, 2002.
Article in English | MEDLINE | ID: mdl-12211042

ABSTRACT

Alphaviruses are positive-stranded RNA viruses that have a broad host range and therefore are capable of replicating in many vertebrate and invertebrate cells. The single-stranded alphavirus genome is divided into two ORFs. The first ORF encodes the nonstructural proteins that are translated upon entry of the virus into the cytoplasm and are responsible for transcription and replication of viral RNA. The second ORF is under the control of a subgenomic promoter and normally encodes the structural proteins, which are responsible for encapsidation of viral RNA and final assembly into enveloped particles. Expression vectors have been engineered from at least three alphaviruses in which the structural protein gene region has been replaced by heterologous genes and have been shown to express high levels of the heterologous protein in cultured cells. These RNA vectors, known as replicons, are capable of replicating on their own but are not packaged into virus-like particles unless the structural proteins are provided in trans. Thus, replicons are single cycle vectors incapable of spreading from infected to noninfected cells. Because of these features, alphavirus replicon vectors are being developed as a platform vaccine technology for numerous viral, bacterial, protozoan and tumour antigens where they have been shown to be efficient inducers of both humoral and T cell responses. In addition, as the alphavirus structural proteins are not expressed in vaccine recipients, antivector immune responses are generally minimal, allowing for multiple effective immunisations of the same individual.


Subject(s)
Alphavirus/genetics , Genetic Vectors , Vaccines, DNA , Vaccines, Synthetic , Animals , Communicable Disease Control , Humans , Replicon , Vaccination/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...