Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Vaccine ; 34(1): 101-9, 2016 Jan 02.
Article in English | MEDLINE | ID: mdl-26571309

ABSTRACT

BACKGROUND/OBJECTIVES: There is currently no licensed prophylactic or therapeutic vaccine for HSV-2 infection. METHODS: We developed a novel preclinical vaccine candidate, G103, consisting of three recombinantly expressed HSV-2 proteins (gD and the UL19 and UL25 gene products) adjuvanted with the potent synthetic TLR4 agonist glucopyranosyl lipid A (GLA) formulated in stable emulsion. The vaccine was tested for immunogenicity and efficacy in pre-clinical models for preventative and therapeutic vaccination. RESULTS: Vaccination of mice with G103 elicited antigen-specific binding and neutralizing antibody responses, as well as robust CD4 and CD8 effector and memory T cells. The T cell responses were further boosted by subsequent challenge with live virus. Prophylactic immunization completely protected against lethal intravaginal HSV-2 infection in mice, with only transient replication of virus in the genital mucosa and sterilizing immunity in dorsal root ganglia. Supporting the use of G103 therapeutically, the vaccine expanded both CD4 and CD8 T cells induced in mice by previous infection with HSV-2. In the guinea pig model of recurrent HSV-2 infection, therapeutic immunization with G103 was approximately 50% effective in reducing the number of lesions per animal as well as the overall lesions score. CONCLUSIONS: Taken together, the data show that G103 is a viable candidate for development of a novel prophylactic and therapeutic HSV-2 vaccine.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Glucosides/administration & dosage , Herpes Genitalis/prevention & control , Herpes Genitalis/therapy , Herpesvirus 2, Human/immunology , Herpesvirus Vaccines/immunology , Lipid A/administration & dosage , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies, Neutralizing/blood , Disease Models, Animal , Female , Guinea Pigs , Herpes Genitalis/immunology , Herpesvirus 2, Human/genetics , Herpesvirus Vaccines/administration & dosage , Herpesvirus Vaccines/isolation & purification , Immunologic Memory , Mice, Inbred C57BL , Treatment Outcome , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology , Vaccines, Subunit/isolation & purification , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology , Vaccines, Synthetic/isolation & purification
2.
Vaccine ; 33(44): 5897-904, 2015 Nov 04.
Article in English | MEDLINE | ID: mdl-26387436

ABSTRACT

Viral infections are associated with production losses in many animal production industries. Important examples of this are Marek's disease (MD) and bovine respiratory disease (BRD) which are significant issues in the chicken and cattle industries, respectively. Viruses play key roles in MD and BRD development and consequently have also been utilised in vaccination strategies to control these diseases. Despite the widespread availability and use of vaccines to control these diseases both are still major issues for their respective industries. Here the dual role of members of viruses from the family Herpesviridae in causation and control of MD and BRD will be discussed. The technologies that may lead to the development of improved vaccines to provide more sustainable control of MD and BRD will also be identified.


Subject(s)
Herpesviridae/immunology , Herpesviridae/pathogenicity , Herpesvirus Vaccines/immunology , Herpesvirus Vaccines/isolation & purification , Infectious Bovine Rhinotracheitis/virology , Marek Disease/virology , Veterinary Medicine/methods , Animals , Cattle , Chickens , Drug Discovery/methods , Herpesviridae/genetics , Herpesvirus Vaccines/genetics , Infectious Bovine Rhinotracheitis/prevention & control , Marek Disease/prevention & control
3.
Cell ; 162(5): 1090-100, 2015 Aug 27.
Article in English | MEDLINE | ID: mdl-26279189

ABSTRACT

Epstein-Barr virus (EBV) represents a major global health problem. Though it is associated with infectious mononucleosis and ∼200,000 cancers annually worldwide, a vaccine is not available. The major target of immunity is EBV glycoprotein 350/220 (gp350) that mediates attachment to B cells through complement receptor 2 (CR2/CD21). Here, we created self-assembling nanoparticles that displayed different domains of gp350 in a symmetric array. By focusing presentation of the CR2-binding domain on nanoparticles, potent neutralizing antibodies were elicited in mice and non-human primates. The structurally designed nanoparticle vaccine increased neutralization 10- to 100-fold compared to soluble gp350 by targeting a functionally conserved site of vulnerability, improving vaccine-induced protection in a mouse model. This rational approach to EBV vaccine design elicited potent neutralizing antibody responses by arrayed presentation of a conserved viral entry domain, a strategy that can be applied to other viruses.


Subject(s)
Herpesvirus Vaccines/chemistry , Herpesvirus Vaccines/immunology , Animals , Antibodies, Neutralizing/immunology , Crystallography, X-Ray , Drug Design , Female , Herpesvirus 4, Human , Herpesvirus Vaccines/genetics , Herpesvirus Vaccines/isolation & purification , Macaca fascicularis , Mice , Mice, Inbred BALB C , Nanoparticles/chemistry , Nanoparticles/ultrastructure , Receptors, Complement 3d/chemistry , Receptors, Complement 3d/immunology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification
4.
Curr Opin Virol ; 11: 89-97, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25829256

ABSTRACT

Although almost 15 years have passed since the birthdate of Reverse Vaccinology (RV), there are very limited applications of this approach to viral vaccines discovery. Undeniably, RV presents a series of advantages as it can virtually identify all potential antigens coded by a genome, irrespective of their abundance, phase of expression and immunogenicity. Additionally, it can be applied to all pathogens, including those that cannot be grown in vitro. In this review we summarize the few examples of RV application to viruses, in particular the Herpesviridae, and report the advantage and limitations of this approach. Next we focus on the novel approaches and additional technologies to vaccine development including structure based approach (Structural Vaccinology [SV]), synthetic biology and some examples of their application in the development of viral vaccines.


Subject(s)
Herpesviridae/genetics , Herpesviridae/immunology , Herpesvirus Vaccines/immunology , Herpesvirus Vaccines/isolation & purification , Reverse Genetics/methods , Drug Discovery/methods , Herpesvirus Vaccines/genetics , Humans
5.
Vaccine ; 32(50): 6733-45, 2014 Nov 28.
Article in English | MEDLINE | ID: mdl-25446827

ABSTRACT

Herpes simplex virus type 1 and type 2 (HSV-1 & HSV-2) infections have been prevalent since the ancient Greek times. To this day, they still affect a staggering number of over a billion individuals worldwide. HSV-1 infections are predominant than HSV-2 infections and cause potentially blinding ocular herpes, oro-facial herpes and encephalitis. HSV-2 infections cause painful genital herpes, encephalitis, and death in newborns. While prophylactic and therapeutic HSV vaccines remain urgently needed for centuries, their development has been difficult. During the most recent National Institute of Health (NIH) workshop titled "Next Generation Herpes Simplex Virus Vaccines: The Challenges and Opportunities", basic researchers, funding agencies, and pharmaceutical representatives gathered: (i) to assess the status of herpes vaccine research; and (ii) to identify the gaps and propose alternative approaches in developing a safe and efficient herpes vaccine. One "common denominator" among previously failed clinical herpes vaccine trials is that they either used a whole virus or a whole viral protein, which contain both "pathogenic symptomatic" and "protective asymptomatic" antigens and epitopes. In this report, we continue to advocate developing "asymptomatic" epitope-based sub-unit vaccine strategies that selectively incorporate "protective asymptomatic" epitopes which: (i) are exclusively recognized by effector memory CD4(+) and CD8(+) T cells (TEM cells) from "naturally" protected seropositive asymptomatic individuals; and (ii) protect human leukocyte antigen (HLA) transgenic animal models of ocular and genital herpes. We review the role of animal models in herpes vaccine development and discuss their current status, challenges, and prospects.


Subject(s)
Epitopes, T-Lymphocyte/immunology , Herpes Genitalis/prevention & control , Herpes Simplex/prevention & control , Herpesvirus Vaccines/immunology , Herpesvirus Vaccines/isolation & purification , Simplexvirus/immunology , Drug Discovery/methods , Herpes Genitalis/epidemiology , Herpes Simplex/epidemiology , Humans , Models, Animal
6.
Trans Am Clin Climatol Assoc ; 114: 103-12, 2003.
Article in English | MEDLINE | ID: mdl-12813914

ABSTRACT

Many viruses capable of persistent or recurrent infections have evolved strategies to evade host immunity. Viral evasion molecules target components of innate and acquired immunity, including complement proteins, natural killer cells, MHC Class I or Class II molecules and antibody. Our work focuses on HSV-1 glycoproteins gC and gE that impair antibody and complement responses. gC inhibits complement activation by binding C3b and blocking activities mediated by this pivotal complement protein, while gE binds the IgG Fc domain, blocking Fc-mediated activities, including complement activation and antibody-dependent cellular cytotoxicity. HSV-1 mutant viruses that lack the ability to bind C3b, IgG Fc, or both are much less virulent than wild-type virus in a murine model. These HSV-1 immunoevasins help explain the virus' ability to produce recurrent infections despite intact immunity. Strategies to prevent immune evasion may be required to develop successful HSV vaccines.


Subject(s)
Herpesvirus 1, Human/immunology , Herpesvirus 1, Human/pathogenicity , Animals , Antibodies, Viral , Female , Herpes Simplex/immunology , Herpes Simplex/prevention & control , Herpes Simplex/virology , Herpesvirus 1, Human/genetics , Herpesvirus Vaccines/isolation & purification , Immunization, Passive , Mice , Mice, Inbred BALB C , Mutation , Neutralization Tests , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Virulence/genetics , Virulence/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...