Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 107
Filtrar
1.
Vaccine ; 42(19S1): S82-S100, 2024 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-39003018

RESUMEN

Herpes simplex virus type 1 (HSV-1) and type 2 (HSV-2) are chronic, highly prevalent viral infections that cause significant morbidity around the world. HSV-2 is sexually transmitted and is the leading cause of genital ulcer disease (GUD). It also increases the risk of HIV acquisition, fueling the HIV epidemic. HSV-1 is typically acquired in childhood through nonsexual contact and contributes to oral and ocular disease, but it can also be sexually transmitted to cause GUD. Both HSV-1 and HSV-2 cause neonatal herpes and neurologic disease. Given the ubiquitous nature of HSV-1 and HSV-2 infections and the limited existing prevention and control measures, vaccination would be the most efficient strategy to reduce the global burden of morbidity related to HSV infection. Vaccine strategies include prophylactic vaccination, which would prevent infection among susceptible persons and would likely be given to adolescents, and therapeutic vaccinations, which would be given to people with symptomatic genital HSV-2 infection. This document discusses the vaccine value profile of both types of vaccines. This 'Vaccine Value Profile' (VVP) for HSV is intended to provide a high-level, holistic assessment of the information and data that are currently available to inform the potential public health, economic and societal value of pipeline vaccines and vaccine-like products. This VVP was developed by subject matter experts from academia, non-profit organizations, government agencies and multi-lateral organizations. All contributors have extensive expertise on various elements of the HSV VVP and collectively aimed to identify current research and knowledge gaps. The VVP was developed using only existing and publicly available information.


Asunto(s)
Herpes Genital , Vacunas contra el Virus del Herpes Simple , Herpes Simple , Herpesvirus Humano 2 , Humanos , Herpesvirus Humano 2/inmunología , Vacunas contra el Virus del Herpes Simple/inmunología , Vacunas contra el Virus del Herpes Simple/administración & dosificación , Herpes Genital/prevención & control , Herpes Genital/inmunología , Herpes Simple/prevención & control , Herpes Simple/inmunología , Herpesvirus Humano 1/inmunología , Vacunación
2.
Front Immunol ; 12: 789454, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34868077

RESUMEN

Herpes simplex virus type-1 (HSV-1) ocular infection is one of the leading causes of infectious blindness in developed countries. The resultant herpetic keratitis (HK) is caused by an exacerbated reaction of the adaptive immune response that persists beyond virus clearance causing substantial damage to the cornea. Intramuscular immunization of mice with the HSV-1(VC2) live-attenuated vaccine strain has been shown to protect mice against lethal ocular challenge. Herein, we show that following ocular challenge, VC2 vaccinated animals control ocular immunopathogenesis in the absence of neutralizing antibodies on ocular surfaces. Ocular protection is associated with enhanced intracorneal infiltration of γδ T cells compared to mock-vaccinated animals. The observed γδ T cellular infiltration was inversely proportional to the infiltration of neutrophils, the latter associated with exacerbated tissue damage. Inhibition of T cell migration into ocular tissues by the S1P receptors agonist FTY720 produced significant ocular disease in vaccinated mice and marked increase in neutrophil infiltration. These results indicate that ocular challenge of mice immunized with the VC2 vaccine induce a unique ocular mucosal response that leads into the infiltration of γδ T cells resulting in the amelioration of infection-associated immunopathogenesis.


Asunto(s)
Quimiotaxis de Leucocito , Córnea/inmunología , Vacunas contra el Virus del Herpes Simple/administración & dosificación , Herpesvirus Humano 1/inmunología , Linfocitos Intraepiteliales/inmunología , Queratitis Herpética/prevención & control , Vacunación , Animales , Córnea/patología , Córnea/virología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Vacunas contra el Virus del Herpes Simple/inmunología , Herpesvirus Humano 1/patogenicidad , Interacciones Huésped-Patógeno , Inyecciones Intramusculares , Linfocitos Intraepiteliales/virología , Queratitis Herpética/inmunología , Queratitis Herpética/patología , Queratitis Herpética/virología , Linfangiogénesis , Ratones Endogámicos BALB C , Neovascularización Patológica , Infiltración Neutrófila , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/inmunología
3.
mSphere ; 6(2)2021 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-33910988

RESUMEN

Potent systemic immunity is important for recalled mucosal immune responses, but in the defense against mucosal viral infections, it usually remains low at mucosal sites. Based on our previous findings that enhanced immune responses can be achieved by immunization with an immunogen in combination with a molecular adjuvant, here we designed chemokine-antigen (Ag) fusion constructs (CCL19- or CCL28-herpes simplex virus 2 glycoprotein D [HSV-2 gD]). After intramuscular (i.m.) immunization with different DNA vaccines in a prime and boost strategy, BALB/c mice were challenged with a lethal dose of HSV-2 through the genital tract. Ag-specific immune responses and chemokine receptor-specific lymphocytes were analyzed to determine the effects of CCL19 and CCL28 in strengthening humoral and cellular immunity. Both CCL19 and CCL28 were efficient in inducing long-lasting HSV-2 gD-specific systemic immunity. Compared to CCL19, less CCL28 was required to elicit HSV-2 gD-specific serum IgA responses, Th1- and Th2-like responses of immunoglobulin (Ig) subclasses and cytokines, and CCR3+ T cell enrichment (>8.5-fold) in spleens. These findings together demonstrate that CCL28 tends to assist an immunogen to induce more potently protective immunity than CCL19. This work provides information for the application potential of a promising vaccination strategy against mucosal infections caused by HSV-2 and other sexually transmitted viruses.IMPORTANCE An effective HSV-2 vaccine should induce antigen (Ag)-specific immune responses against viral mucosal infection. This study reveals that chemokine CCL19 or CCL28 enhanced HSV-2 glycoprotein D ectodomain (gD-306aa)-induced immune responses against vaginal virus challenge. In addition to eliciting robust humoral immune responses, the chemokine-Ag fusion construct also induced Th1- and Th2-like immune responses characterized by the secretion of multiple Ig subclasses and cytokines that were able to be recalled after HSV-2 challenge, while CCL28 appeared to be more effective than CCL19 in promoting gD-elicited immune responses as well as the migration of T cells to secondary lymph tissues. Of importance, both CCL19 and CCL28 significantly facilitated gD to induce protective mucosal immune responses in the genital tract. The above-described findings together highlight the potential of CCL19 or CCL28 in combination with gD as a vaccination strategy to control HSV-2 infection.


Asunto(s)
Anticuerpos Antivirales/sangre , Quimiocina CCL19/inmunología , Quimiocinas CC/inmunología , Vacunas contra el Virus del Herpes Simple/inmunología , Herpesvirus Humano 2/genética , Herpesvirus Humano 2/inmunología , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Animales , Quimiocina CCL19/genética , Quimiocinas CC/genética , Femenino , Vacunas contra el Virus del Herpes Simple/administración & dosificación , Vacunas contra el Virus del Herpes Simple/clasificación , Inmunidad Mucosa , Memoria Inmunológica , Ratones , Ratones Endogámicos BALB C , Vacunación/métodos , Vagina/inmunología , Vagina/virología
4.
J Infect Dis ; 224(9): 1509-1519, 2021 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-33718970

RESUMEN

Previous herpes simplex virus type 2 (HSV-2) vaccines have not prevented genital herpes. Concerns have been raised about the choice of antigen, the type of antibody induced by the vaccine, and whether antibody is present in the genital tract where infection occurs. We reported results of a trial of an HSV-2 replication-defective vaccine, HSV529, that induced serum neutralizing antibody responses in 78% of HSV-1-/HSV-2- vaccine recipients. Here we show that HSV-1-/HSV-2- vaccine recipients developed antibodies to epitopes of several viral proteins; however, fewer antibody epitopes were detected in vaccine recipients compared with naturally infected persons. HSV529 induced antibodies that mediated HSV-2-specific natural killer (NK) cell activation. Depletion of glycoprotein D (gD)-binding antibody from sera reduced neutralizing titers by 62% and NK cell activation by 81%. HSV-2 gD antibody was detected in cervicovaginal fluid at about one-third the level of that in serum. A vaccine that induces potent serum antibodies transported to the genital tract might reduce HSV genital infection.


Asunto(s)
Anticuerpos Antivirales/sangre , Herpes Genital/prevención & control , Vacunas contra el Virus del Herpes Simple/administración & dosificación , Herpes Simple/prevención & control , Herpesvirus Humano 2/inmunología , Proteínas del Envoltorio Viral/inmunología , Vacunas Virales/administración & dosificación , Epítopos , Vacunas contra el Virus del Herpes Simple/inmunología , Herpesvirus Humano 1/inmunología , Humanos , Inmunización
5.
Immunohorizons ; 4(10): 608-626, 2020 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-33037098

RESUMEN

The protective efficacy of a live-attenuated HSV type 1 (HSV-1) vaccine, HSV-1 0∆ nuclear location signal (NLS), was evaluated in mice prophylactically in response to ocular HSV-1 challenge. Mice vaccinated with the HSV-1 0∆NLS were found to be more resistant to subsequent ocular virus challenge in terms of viral shedding, spread, the inflammatory response, and ocular pathology in a dose-dependent fashion. Specifically, a strong neutralizing Ab profile associated with low virus titers recovered from the cornea and trigeminal ganglia was observed in vaccinated mice in a dose-dependent fashion with doses ranging from 1 × 103 to 1 × 105 PFU HSV-1 0∆NLS. This correlation also existed in terms of viral latency in the trigeminal ganglia, corneal neovascularization, and leukocyte infiltration and expression of inflammatory cytokines and chemokines in infected tissue with the higher doses (1 × 104-1 × 105 PFU) of the HSV-1 0∆NLS-vaccinated mice, displaying reduced viral latency, ocular pathology, or inflammation in comparison with the lowest dose (1 × 103 PFU) or vehicle vaccine employed. Fifteen HSV-1-encoded proteins were uniquely recognized by antisera from high-dose (1 × 105 PFU)-vaccinated mice in comparison with low-dose (1 × 103 PFU)- or vehicle-vaccinated animals. Passive immunization using high-dose-vaccinated, but not low-dose-vaccinated, mouse sera showed significant efficacy against ocular pathology in HSV-1-challenged animals. In summary, we have identified the minimal protective dose of HSV-1 0∆NLS vaccine in mice to prevent HSV-mediated disease and identified candidate proteins that may be useful in the development of a noninfectious prophylactic vaccine against the insidious HSV-1 pathogen.


Asunto(s)
Córnea/patología , Vacunas contra el Virus del Herpes Simple/administración & dosificación , Vacunas contra el Virus del Herpes Simple/inmunología , Herpesvirus Humano 1/inmunología , Queratitis Herpética/inmunología , Queratitis Herpética/prevención & control , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Córnea/inmunología , Córnea/virología , Femenino , Herpesvirus Humano 1/patogenicidad , Inmunidad Humoral , Inmunización Pasiva , Queratitis Herpética/virología , Masculino , Ratones , Ratones Endogámicos C57BL , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/inmunología , Proteínas del Envoltorio Viral/administración & dosificación , Proteínas del Envoltorio Viral/inmunología , Esparcimiento de Virus
6.
J Virol ; 95(1)2020 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-33028712

RESUMEN

Estradiol (E2) is a sex hormone which has been shown to be protective against sexually transmitted infections such as herpes simplex virus 2 (HSV-2). However, few studies have examined the underlying mechanisms by which this occurs. Here, we investigated the effect of E2 on the establishment of memory T cells post-intranasal immunization with HSV-2. CD4+ T cell responses first appeared in the upper respiratory tract (URT) within 3 days postimmunization before being detected in the female reproductive tract (FRT) at 7 days. E2 treatment resulted in greater and earlier Th17 responses, which preceded augmented Th1 responses at these sites. The CD4+ T cells persisted in the URT for up to 28 days, and E2 treatment resulted in higher frequencies of memory T cells. Intranasal immunization also led to the establishment of CD4+ tissue-resident memory T cells (TRM cells) in the FRT, and E2 treatment resulted in increased Th1 and Th17 TRM cells. When the migration of circulating T cells into the FRT was blocked by FTY720, immunized E2-treated mice remained completely protected against subsequent genital HSV-2 challenge compared to non-E2 controls, confirming that TRM cells alone are adequate for protection in these mice. Finally, the enhanced vaginal Th1 TRM cells present in E2-treated mice were found to be modulated through an interleukin 17 (IL-17)-mediated pathway, as E2-treated IL-17A-deficient mice had impaired establishment of Th1 TRM cells. This study describes a novel role for E2 in enhancing CD4+ memory T cells and provides insight on potential strategies for generating optimal immunity during vaccination.IMPORTANCE Herpes simplex virus 2 (HSV-2) is a highly prevalent sexually transmitted infection for which there is currently no vaccine available. Interestingly, the female sex hormone estradiol has been shown to be protective against HSV-2. However, the underlying mechanisms by which this occurs remains relatively unknown. Our study demonstrates that under the influence of estradiol treatment, intranasal immunization with an attenuated strain of HSV-2 leads to enhanced establishment of antiviral memory T cell responses in the upper respiratory tract and female reproductive tract. In these sites, estradiol treatment leads to greater Th17 memory cells, which precede enhanced Th1 memory responses. Consequently, the T cell responses mounted by tissue-resident memory cells in the female reproductive tract of estradiol-treated mice are sufficient to protect mice against vaginal HSV-2 challenge. This study offers important insights regarding the regulation of mucosal immunity by hormones and on potential strategies for generating optimal immunity during vaccination.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Estradiol/inmunología , Vacunas contra el Virus del Herpes Simple/inmunología , Herpesvirus Humano 2/inmunología , Memoria Inmunológica , Interleucina-17/inmunología , Vacunación/métodos , Administración Intranasal , Animales , Linfocitos T CD8-positivos/inmunología , Estradiol/administración & dosificación , Femenino , Herpes Genital/prevención & control , Vacunas contra el Virus del Herpes Simple/administración & dosificación , Inmunidad Mucosa , Ratones , Sistema Respiratorio/inmunología , Células TH1/inmunología , Células Th17/inmunología , Vagina/inmunología
7.
PLoS Pathog ; 16(8): e1008703, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32776994

RESUMEN

Herpes simplex virus type 1 (HSV1) is a complicated structural agent with a sophisticated transcription process and a high infection rate. A vaccine against HSV1 is urgently needed. As multiple viral-encoded proteins, including structural and nonstructural proteins, contribute to immune response stimulation, an attenuated or deficient HSV1 vaccine may be relatively reliable. Advances in genomic modification technologies provide reliable means of constructing various HSV vaccine candidates. Based on our previous work, an M6 mutant with mutations in the UL7, UL41, LAT, Us3, Us11 and Us12 genes was established. The mutant exhibited low proliferation in cells and an attenuated phenotype in an animal model. Furthermore, in mice and rhesus monkeys, the mutant can induce remarkable serum neutralizing antibody titers and T cell activation and protect against HSV1 challenge by impeding viral replication, dissemination and pathogenesis.


Asunto(s)
Vacunas contra el Virus del Herpes Simple/inmunología , Herpes Simple/inmunología , Herpesvirus Humano 1/inmunología , Animales , Femenino , Herpes Simple/prevención & control , Herpes Simple/virología , Vacunas contra el Virus del Herpes Simple/administración & dosificación , Vacunas contra el Virus del Herpes Simple/genética , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/fisiología , Humanos , Ratones , Ratones Endogámicos BALB C , Mutación , Fenotipo , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Proteínas Virales/administración & dosificación , Proteínas Virales/genética , Proteínas Virales/inmunología
8.
Vaccine ; 38(1): 79-89, 2020 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-31611098

RESUMEN

Infection with Herpes Simplex Viruses (HSVs) represents a significant health burden worldwide with HSV-1 and HSV-2 causing genital disease and HSV-2 contributing to human immunodeficiency virus acquisition. Despite great need, there is currently no licensed vaccine against HSV. In this report, we evaluated the protective efficacy of a vaccine containing highly purified, inactivated HSV-2 particles (with and without additional recombinant glycoprotein D) formulated with a monophosphoryl lipid A/Alhydrogel adjuvant in a guinea pig HSV genital model. The key results from 3 independent studies were: (1) vaccination consistently provided significant 3-3.5 Log10 reductions in vaginal HSV-2 titers on day 2 postchallenge; (2) following homologous or heterologous challenge with two U.S. isolates, all vaccine groups showed complete protection against lesion formation, significant 3 Log10 reductions in day 2 virus shedding, enhanced virus clearance, significant reductions in HSV-2 DNA within ganglia, and no detectable shedding (<2 PFU) or latent viral DNA in some immunized animals; (3) following challenge with a third heterologous strain, vaccination provided complete protection against primary and recurrent lesions, significant reductions in primary virus shedding, a 50% reduction in recurrent shedding days, and undetectable latent virus in the ganglia and spinal cords of most animals; and (4) adding glycoprotein D provided no enhanced protection relative to that elicited by the inactivated HSV-2 particles alone. Together, these data provide strong support for further development of this exceedingly protective and highly feasible vaccine candidate for human trials.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Herpes Genital/prevención & control , Vacunas contra el Virus del Herpes Simple/administración & dosificación , Herpesvirus Humano 2/efectos de los fármacos , Virión , Administración Intravaginal , Animales , Chlorocebus aethiops , Femenino , Cobayas , Herpes Genital/inmunología , Vacunas contra el Virus del Herpes Simple/inmunología , Herpesvirus Humano 2/inmunología , Inmunidad Humoral/efectos de los fármacos , Inmunidad Humoral/inmunología , Células Vero , Virión/inmunología , Esparcimiento de Virus/efectos de los fármacos , Esparcimiento de Virus/inmunología
9.
J Virol ; 94(1)2019 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-31597775

RESUMEN

Demyelinating central nervous system (CNS) disorders like multiple sclerosis (MS) and acute disseminated encephalomyelitis (ADEM) have been difficult to study and treat due to the lack of understanding of their etiology. Numerous cases point to the link between herpes simplex virus (HSV) infection and multifocal CNS demyelination in humans; however, convincing evidence from animal models has been missing. In this work, we found that HSV-1 infection of the cotton rat Sigmodon hispidus via a common route (lip abrasion) can cause multifocal CNS demyelination and inflammation. Remyelination occurred shortly after demyelination in HSV-1-infected cotton rats but could be incomplete, resulting in "scars," further supporting an association between HSV-1 infection and multifocal demyelinating disorders. Virus was detected sequentially in the lip, trigeminal ganglia, and brain of infected animals. Brain pathology developed primarily on the ipsilateral side of the brain stem, in the cerebellum, and contralateral side of the forebrain/midbrain, suggesting that the changes may ascend along the trigeminal lemniscus pathway. Neurologic defects occasionally detected in infected animals (e.g., defective whisker touch and blink responses and compromised balance) could be representative of the brain stem/cerebellum dysfunction. Immunization of cotton rats with a split HSV-1 vaccine protected animals against viral replication and brain pathology, suggesting that vaccination against HSV-1 may protect against demyelinating disorders.IMPORTANCE Our work demonstrates for the first time a direct association between infection with herpes simplex virus 1, a ubiquitous human pathogen generally associated with facial cold sores, and multifocal brain demyelination in an otherwise normal host, the cotton rat Sigmodon hispidus For a long time, demyelinating diseases were considered to be autoimmune in nature and were studied by indirect methods, such as immunizing animals with myelin components or feeding them toxic substances that induce demyelination. Treatment against demyelinating diseases has been elusive, partially because of their unknown etiology. This work provides the first experimental evidence for the role of HSV-1 as the etiologic agent of multifocal brain demyelination in a normal host and suggests that vaccination against HSV-1 can help to combat demyelinating disorders.


Asunto(s)
Enfermedades Desmielinizantes/prevención & control , Encefalitis/prevención & control , Vacunas contra el Virus del Herpes Simple/administración & dosificación , Herpes Simple/prevención & control , Herpesvirus Humano 1/efectos de los fármacos , Animales , Tronco Encefálico/efectos de los fármacos , Tronco Encefálico/inmunología , Tronco Encefálico/patología , Tronco Encefálico/virología , Cerebelo/efectos de los fármacos , Cerebelo/inmunología , Cerebelo/patología , Cerebelo/virología , Enfermedades Desmielinizantes/inmunología , Enfermedades Desmielinizantes/patología , Enfermedades Desmielinizantes/virología , Modelos Animales de Enfermedad , Encefalitis/inmunología , Encefalitis/patología , Encefalitis/virología , Femenino , Herpes Simple/inmunología , Herpes Simple/patología , Herpes Simple/virología , Herpesvirus Humano 1/inmunología , Herpesvirus Humano 1/patogenicidad , Humanos , Masculino , Prosencéfalo/efectos de los fármacos , Prosencéfalo/inmunología , Prosencéfalo/patología , Prosencéfalo/virología , Sigmodontinae , Ganglio del Trigémino/efectos de los fármacos , Ganglio del Trigémino/inmunología , Ganglio del Trigémino/patología , Ganglio del Trigémino/virología , Vacunación , Carga Viral/efectos de los fármacos
10.
Vaccine ; 37(43): 6470-6477, 2019 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-31515143

RESUMEN

Genital herpes is a sexually transmitted disease representing a major global health concern. Currently, there is no approved vaccine and existing antiviral therapies exhibit limited efficacy. Herein, we describe an intranasal (IN) vaccine comprised of HSV-2 surface glycoproteins gD2 and gB2 formulated in a nanoemulsion adjuvant (NE01-gD2/gB2). Using the HSV-2 genital herpes guinea pig model, we demonstrate that IN NE01-gD2/gB2 induces higher levels of neutralizing antibody compared to a monovalent IN NE01-gD2 vaccine, but less than an intramuscular (IM) Alum/MPL-gD2 vaccine. Following intravaginal (IVag) challenge with HSV-2, the group immunized with IN NE01-gD2/gB2 exhibited significantly reduced acute and recurrent disease scores compared to placebo recipients. Significantly, latent virus was only detected in the dorsal root ganglia of 1 of 12 IN NE01-gD2/gB2-vaccinated animals compared to 11 of 12 placebo recipient. In the therapeutic model, IN NE01-gD2/gB2 immunized guinea pigs exhibited a significant reduction in the recurrent lesions scores (64%, p < 0.01), number of animal days with disease (64%, p < 0.01), number of animals with viral shedding (50%, p < 0.04) and reduction in virus positive vaginal swabs (56%, p < 0.04), These data suggests that the treatment may be effective in treating chronic disease and minimizing virus transmission. These results warrant advancing the development of IN NE01-gD2/gB2 as both a prophylactic and therapeutic vaccine against HSV-2.


Asunto(s)
Anticuerpos Antivirales/sangre , Herpes Genital/prevención & control , Herpes Genital/terapia , Vacunas contra el Virus del Herpes Simple/administración & dosificación , Vacunas contra el Virus del Herpes Simple/uso terapéutico , Adyuvantes Inmunológicos/administración & dosificación , Administración Intranasal , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Modelos Animales de Enfermedad , Emulsiones/administración & dosificación , Emulsiones/química , Femenino , Cobayas , Herpes Genital/inmunología , Herpesvirus Humano 2 , Nanopartículas/administración & dosificación , Nanopartículas/química , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/uso terapéutico , Esparcimiento de Virus
11.
PLoS One ; 14(3): e0213401, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30917165

RESUMEN

BACKGROUND: Although herpes simplex viruses (HSV) are a major target for vaccine development no vaccine is currently licensed. METHODS: A live attenuated HSV virus vaccine, VC2 was compared to a subunit HSV vaccine, glycoprotein D (gD2) administered with the adjuvant, MPL/Alum using the guinea pig model of genital herpes. Three doses of intramuscular (IM) vaccine were provided followed by intravaginal challenge with HSV-2 at either 3 weeks or six months after the last vaccination. RESULTS: Both VC2 and gD2 vaccines reduced acute genital disease. VC2 was somewhat more effective in reducing acute vaginal replication, the amount of virus in neural tissue, subsequent recurrent disease and recurrent virus shedding following challenge at 3 weeks post vaccination. Both vaccines continued to provide protection at 6 months after vaccination but the differences between the vaccines became more pronounced in favor of the live attenuated vaccine, VC2. Significant differences in acute disease, acute vaginal virus replication, recurrent disease and recurrent virus shedding (P<0.05 for each) was observed comparing the vaccines. Re-examination of protection for this study using criteria similar to those used in recent clinical trials (inclusion of recurrent disease) showed that efficacy may not be as high in this model as previously thought prompting a need to assess the best predictive outcomes for protection in humans. CONCLUSION: While both the live attenuated vaccine, VC2, and the gD2 subunit vaccine provided protection, the duration of protection appeared to be greater for VC2. Using the same evaluation criteria as used in human trials provided unique insights into the utility of the guinea pig model.


Asunto(s)
Herpes Genital/prevención & control , Vacunas contra el Virus del Herpes Simple/farmacología , Herpesvirus Humano 2/inmunología , Animales , Ensayos Clínicos como Asunto , Modelos Animales de Enfermedad , Femenino , Cobayas , Herpes Genital/inmunología , Herpes Genital/virología , Vacunas contra el Virus del Herpes Simple/administración & dosificación , Humanos , Esquemas de Inmunización , Factores de Tiempo , Resultado del Tratamiento , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/farmacología , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/farmacología , Vagina/inmunología , Vagina/virología , Proteínas del Envoltorio Viral/inmunología
12.
Vaccine ; 37(50): 7396-7407, 2019 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-29625767

RESUMEN

Development of a vaccine against herpes simplex virus type 2 (HSV-2), a life-long sexually-transmitted infection (STI), would be a major step forward in improving global sexual and reproductive health. In this review, we identified published literature of dynamic mathematical models assessing the impact of either prophylactic or therapeutic HSV-2 vaccination at the population level. We compared each study's model structure and assumptions as well as predicted vaccination impact. We examined possible causes of heterogeneity across model predictions, key gaps, and the implications of these findings for future modelling efforts. Only eight modelling studies have assessed the potential public health impact of HSV-2 vaccination, with the majority focusing on impact of prophylactic vaccines. The studies showed that even an imperfect prophylactic HSV-2 vaccine could have an important public health impact on HSV-2 incidence, and could also impact HIV indirectly in high HIV prevalence settings. Therapeutic vaccines also may provide public health benefits, though they have been explored less extensively. However, there was substantial variation in predicted population-level impact for both types of vaccine, reflecting differences in assumptions between model scenarios. Importantly, many models did not account for heterogeneity in infection rates such as by age, sex and sexual activity. Future modelling work to inform decisions on HSV vaccine development and implementation should consider cost-effectiveness, account for additional HSV-2 sequelae such as neonatal transmission, and model greater heterogeneity in infection rates between individuals, more realistic vaccine deployment, and more thorough sensitivity and uncertainty analyses.


Asunto(s)
Infecciones por VIH/prevención & control , Herpes Genital/prevención & control , Vacunas contra el Virus del Herpes Simple/administración & dosificación , Herpesvirus Humano 2/efectos de los fármacos , Modelos Estadísticos , Vacunación/métodos , Factores de Edad , Coinfección , Femenino , VIH/inmunología , VIH/patogenicidad , Infecciones por VIH/epidemiología , Infecciones por VIH/inmunología , Infecciones por VIH/virología , Herpes Genital/epidemiología , Herpes Genital/inmunología , Herpes Genital/virología , Herpesvirus Humano 2/inmunología , Herpesvirus Humano 2/patogenicidad , Humanos , Incidencia , Masculino , Profilaxis Posexposición/métodos , Profilaxis Pre-Exposición/métodos , Salud Pública/estadística & datos numéricos , Factores Sexuales , Conducta Sexual/fisiología , Conducta Sexual/estadística & datos numéricos
13.
Acta Virol ; 62(2): 164-171, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29895157

RESUMEN

HSV-1 is a mucosal and nerve pathogen, whose morbidity shows an increasing tendency. Although several antiviral drugs exist, there is no cure for viral latency for virtually all carriers. There is an urgent need for an HSV-1 vaccine to control infection and limit its spread and recurrence. The UL18 gene, encoding a vital component of capsids, is one of the essential genes of HSV-1. Deletion of UL18 from HSV-1 may be exploited as a new approach to develop an attenuated vaccine. The purpose of this study was to construct a DNA vaccine with a full-length UL18 gene deletion of the HSV-1 genome that can induce an effective immune response. A UL18-knockdown plasmid (BAC-HSV-1ΔUL18) was constructed using the bacterial markerless gene knockout system, consisting of the functional pREDI plasmid and BAC-HSV-1 plasmid. Mice were immunized weekly for 3 weeks, and at 1 week post immunization, blood and splenocyte samples of vaccinated and control groups of mice were prepared for immunogenicity assessment. The level of immune response was evaluated using a DTH assay, cytokine determination, and splenocyte proliferation assay. Combination of the pREDI plasmid and BAC-HSV-1 plasmid provides an effective bacterial markerless gene knockout system. Using two-step homologous recombination with the UL18 homologous recombination fragment constructed by multistep PCR amplification, BAC-HSV-1ΔUL18 plasmid vaccine was successfully constructed and was found to significantly enhance cellular immune responses.


Asunto(s)
Proteínas de la Cápside/genética , Vacunas contra el Virus del Herpes Simple/inmunología , Herpes Simple/inmunología , Herpesvirus Humano 1/inmunología , Animales , Anticuerpos Antivirales/inmunología , Proteínas de la Cápside/administración & dosificación , Proteínas de la Cápside/inmunología , Eliminación de Gen , Herpes Simple/prevención & control , Herpes Simple/virología , Vacunas contra el Virus del Herpes Simple/administración & dosificación , Vacunas contra el Virus del Herpes Simple/genética , Herpesvirus Humano 1/genética , Humanos , Inmunización , Masculino , Ratones , Eliminación de Secuencia , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología
14.
J Virol ; 92(16)2018 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-29899087

RESUMEN

Herpes simplex virus 1 (HSV-1) is a prevalent human pathogen that infects the cornea, causing potentially blinding herpetic disease. A clinical herpes vaccine is still lacking. In the present study, a novel prime/pull vaccine was tested in a human leukocyte antigen (HLA) transgenic rabbit model of ocular herpes (HLA Tg rabbits). Three peptide epitopes were selected, from the HSV-1 membrane glycoprotein C (UL44400-408), the DNA replication binding helicase (UL9196-204), and the tegument protein (UL25572-580), all preferentially recognized by CD8+ T cells from "naturally protected" HSV-1-seropositive healthy asymptomatic (ASYMP) individuals (who never had recurrent corneal herpetic disease). HLA Tg rabbits were immunized with a mixture of these three ASYMP CD8+ T cell peptide epitopes (UL44400-408, UL9196-204, and UL25572-580), which were delivered subcutaneously with CpG2007 adjuvant (prime). Fifteen days later, half of the rabbits received a topical ocular treatment with a recombinant neurotropic adeno-associated virus type 8 (AAV8) vector expressing the T cell-attracting CXCL10 chemokine (pull). The frequency and function of HSV-specific CD8+ T cells induced by the prime/pull vaccine were assessed in the peripheral blood, cornea, and trigeminal ganglion (TG). Compared to the cells generated in response to peptide immunization alone, the peptide/CXCL10 prime/pull vaccine generated frequent polyfunctional gamma interferon-positive (IFN-γ+) CD107+ CD8+ T cells that infiltrated both the cornea and TG. CD8+ T cell mobilization into the cornea and TG of prime/pull-vaccinated rabbits was associated with a significant reduction in corneal herpesvirus infection and disease following an ocular HSV-1 (strain McKrae) challenge. These findings draw attention to the novel prime/pull vaccine strategy for mobilizing antiviral CD8+ T cells into tissues to protect against herpesvirus infection and disease.IMPORTANCE There is an urgent need for a vaccine against widespread herpes simplex virus infections. The present study demonstrates that immunization of HLA transgenic rabbits with a peptide/CXCL10 prime/pull vaccine triggered mobilization of HSV-specific CD8+ T cells locally into the cornea and TG, the sites of acute and latent herpesvirus infections, respectively. Mobilization of antiviral CD8+ T cells into the cornea and TG of rabbits that received the prime/pull vaccine was associated with protection against ocular herpesvirus infection and disease following an ocular HSV-1 challenge. These results highlight the importance of the prime/pull vaccine strategy to bolster the number and function of protective CD8+ T cells within infected tissues.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Quimiocina CXCL10/metabolismo , Córnea/inmunología , Vacunas contra el Virus del Herpes Simple/inmunología , Queratitis Herpética/prevención & control , Subgrupos de Linfocitos T/inmunología , Ganglio del Trigémino/inmunología , Animales , Animales Modificados Genéticamente , Quimiocina CXCL10/administración & dosificación , Modelos Animales de Enfermedad , Epítopos/inmunología , Antígenos HLA/genética , Antígenos HLA/metabolismo , Vacunas contra el Virus del Herpes Simple/administración & dosificación , Humanos , Interferón gamma/análisis , Queratitis Herpética/patología , Queratitis Herpética/virología , Proteína 1 de la Membrana Asociada a los Lisosomas/análisis , Conejos , Simplexvirus/inmunología , Simplexvirus/aislamiento & purificación , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/inmunología , Carga Viral
15.
Curr Opin Ophthalmol ; 29(4): 340-346, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29846207

RESUMEN

PURPOSE OF REVIEW: Ophthalmic herpes simplex virus (HSV) of the anterior segment is responsible for a range of corneal complications such as scarring, thinning, neovascularization, and severe loss of vision. This review provides current guidelines for treating anterior segment disease related to HSV. RECENT FINDINGS: We first review findings from the Herpetic Eye Disease Study (HEDS) clinical trials, and then review new topical and antiviral therapies developed since the HEDS studies. The development of vaccines to prevent recurrent episodes of herpetic infection is briefly reviewed. New corneal surgical procedures, developed since HEDS, may put patients at risk for ocular HSV disease: cross-linking and excimer refractive surgery. SUMMARY: HEDS established the standard of HSV ocular therapy and is still valid today. However, newer antivirals may provide easier compliance with improved bioavailability, efficacy, dosage, and tolerability. Further research is needed to prevent latency of HSV, decrease recurrences, and more effectively treat necrotizing keratitis associated with HSV.


Asunto(s)
Antivirales/uso terapéutico , Infecciones Virales del Ojo/tratamiento farmacológico , Queratitis Herpética/tratamiento farmacológico , Infecciones Virales del Ojo/prevención & control , Vacunas contra el Virus del Herpes Simple/administración & dosificación , Humanos , Queratitis Herpética/prevención & control , Recurrencia , Simplexvirus/patogenicidad
16.
Expert Rev Vaccines ; 17(3): 239-248, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29313728

RESUMEN

INTRODUCTION: Despite overwhelming experimental work, there are no licensed vaccines against the most frequent Alphaherpesviruses, namely herpes simplex virus 1 and 2 (HSV1 and 2) nor against the Epstein-Barr virus (EBV), a member of the subfamily Gammaherpesvirus. AREAS COVERED: Since the DNAs of both HSVs reside in the regional sensory ganglia in a latent state (i.e. as circularized episomal molecules), a corresponding vaccine might be useful for immunotherapy rather than for prevention of primary infection. Here we describe the design of a purified subunit vaccine as well as the preparation and efficacy of a recombinant fusion protein consisting of the gD ectodomain from our domestic attenuated HSV1 strain HSZP. The EBV vaccines considered so far, were destined for prevention of infectious mononucleosis (IM) or to prevent formation of EBV related tumors. To design the EBV peptide vaccine, at least 15 carefully selected immunogenic epitopes coming from 12 virus coded proteins were bound to synthetic micro-particle carriers along with a non-specific pathogen recognizing receptor (PRR) stimulating both the T as well as B lymphocytes. EXPERT COMMENTARY: The efficacy of a novel EBV peptide in the rabbit model was based on criteria such as antibody formation (EA-D detected by ELISA, early and capsid proteins tested by immunoblot), presence of LMP1 antigen and of viral DNA in peripheral white blood cells. Out of 19 peptide combinations used for vaccination, at least 6 showed a satisfactory protective effect.


Asunto(s)
Infecciones por Virus de Epstein-Barr/prevención & control , Herpes Simple/prevención & control , Vacunas contra Herpesvirus/administración & dosificación , Animales , Infecciones por Virus de Epstein-Barr/inmunología , Herpes Simple/inmunología , Vacunas contra el Virus del Herpes Simple/administración & dosificación , Vacunas contra el Virus del Herpes Simple/inmunología , Herpesvirus Humano 1/inmunología , Herpesvirus Humano 2/inmunología , Herpesvirus Humano 4/inmunología , Vacunas contra Herpesvirus/inmunología , Humanos , Conejos
17.
J Virol ; 91(7)2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-28122977

RESUMEN

Viral fitness dictates virulence and capacity to evade host immune defenses. Understanding the biological underpinnings of such features is essential for rational vaccine development. We have previously shown that the live-attenuated herpes simplex virus 1 (HSV-1) mutant lacking the nuclear localization signal (NLS) on the ICP0 gene (0ΔNLS) is sensitive to inhibition by interferon beta (IFN-ß) in vitro and functions as a highly efficacious experimental vaccine. Here, we characterize the host immune response and in vivo pathogenesis of HSV-1 0ΔNLS relative to its fully virulent parental strain in C57BL/6 mice. Additionally, we explore the role of type 1 interferon (IFN-α/ß) signaling on virulence and immunogenicity of HSV-1 0ΔNLS and uncover a probable sex bias in the induction of IFN-α/ß in the cornea during HSV-1 infection. Our data show that HSV-1 0ΔNLS lacks neurovirulence even in highly immunocompromised mice lacking the IFN-α/ß receptor. These studies support the translational viability of the HSV-1 0ΔNLS vaccine strain by demonstrating that, while it is comparable to a virulent parental strain in terms of immunogenicity, HSV-1 0ΔNLS does not induce significant tissue pathology.IMPORTANCE HSV-1 is a common human pathogen associated with a variety of clinical presentations ranging in severity from periodic "cold sores" to lethal encephalitis. Despite the consistent failures of HSV subunit vaccines in clinical trials spanning the past 28 years, opposition to live-attenuated HSV vaccines predicated on unfounded safety concerns currently limits their widespread acceptance. Here, we demonstrate that a live-attenuated HSV-1 vaccine has great translational potential.


Asunto(s)
Córnea/metabolismo , Vacunas contra el Virus del Herpes Simple/inmunología , Herpes Simple/prevención & control , Herpesvirus Humano 1/inmunología , Interferón Tipo I/fisiología , Inmunidad Adaptativa , Animales , Córnea/inmunología , Córnea/virología , Vacunas contra el Virus del Herpes Simple/administración & dosificación , Ratones Endogámicos C57BL , Ratones Noqueados , Vacunación , Vacunas Atenuadas/inmunología
18.
Vaccine ; 35(4): 536-543, 2017 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-28017425

RESUMEN

We have shown that the live-attenuated HSV-1 VC2 vaccine strain with mutations in glycoprotein K (gK) and the membrane protein UL20 is unable to establish latency in vaccinated animals and produces a robust immune response capable of completely protecting mice against lethal vaginal HSV-1 or HSV-2 infections. To better understand the immune response generated by vaccination with VC2, we tested its ability to elicit immune responses in rhesus macaques. Vaccinated animals showed no signs of disease and developed increasing HSV-1 and HSV-2 reactive IgG1 after two booster vaccinations, while IgG subtypes IgG2 and IgG3 remained at low to undetectable levels. All vaccinated animals produced high levels of cross protective neutralizing antibodies. Flow cytometry analysis of cells isolated from draining lymph nodes showed that VC2 vaccination stimulated significant increases in plasmablast (CD27highCD38high) and mature memory (CD21-IgM-) B cells. T cell analysis on cells isolated from draining lymph node biopsies demonstrated a statistically significant increase in proliferating (Ki67+) follicular T helper cells and regulatory CXCR5+ CD8+ cytotoxic T cells. Analysis of plasma isolated two weeks post vaccination showed significant increases in circulating CXCL13 indicating increased germinal center activity. Cells isolated from vaginal biopsy samples collected over the course of the study exhibited vaccination-dependent increases in proliferating (Ki67+) CD4+ and CD8+ T cell populations. These results suggest that intramuscular vaccination with the live-attenuated HSV-1 VC2 vaccine strain can stimulate robust IgG1 antibody responses that persist for >250days post vaccination. In addition, vaccination lead to the maturation of B cells into plasmablast and mature memory B cells, the expansion of follicular T helper cells, and affects in the mucosal immune responses. These data suggest that the HSV VC2 vaccine induces potent immune responses that could help define correlates of protection towards developing an efficacious HSV-1/HSV-2 vaccine in humans.


Asunto(s)
Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Centro Germinal/inmunología , Vacunas contra el Virus del Herpes Simple/inmunología , Herpesvirus Humano 1/inmunología , Linfocitos Intraepiteliales/inmunología , Animales , Diferenciación Celular , Protección Cruzada , Femenino , Vacunas contra el Virus del Herpes Simple/administración & dosificación , Inmunoglobulina G/sangre , Memoria Inmunológica , Inyecciones Intramusculares , Subgrupos Linfocitarios/inmunología , Macaca mulatta , Ratones , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/inmunología
19.
Hum Vaccin Immunother ; 12(12): 3079-3088, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27580249

RESUMEN

This paper describes a single site, open-label Phase I clinical trial evaluating the safety, tolerability and immunogenicity in healthy volunteers of a herpes simplex polynucleotide vaccine that has previously been shown to enhance immunogenicity and protect against lethal herpes simplex virus type 2 (HSV-2) challenge in mice. Five escalating doses of the vaccine, COR-1, were given by intradermal injection to HSV-1 and 2 seronegative healthy individuals. COR-1 was found to be safe and well-tolerated; the only vaccine-related adverse events were mild. While vaccine-induced antibody responses were not detectable, cell-mediated immune responses to HSV-specific peptide groups were identified in 19 of the 20 subjects who completed the study, and local inflammation at the immunisation site was observed. This study indicates COR-1 has potential to be used as a therapeutic vaccine for HSV-2 infection.


Asunto(s)
Vacunas contra el Virus del Herpes Simple/efectos adversos , Vacunas contra el Virus del Herpes Simple/inmunología , Herpes Simple/prevención & control , Herpesvirus Humano 2/inmunología , Vacunas de ADN/efectos adversos , Vacunas de ADN/inmunología , Adulto , Anticuerpos Antivirales/sangre , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/epidemiología , Femenino , Vacunas contra el Virus del Herpes Simple/administración & dosificación , Humanos , Inyecciones Intradérmicas , Leucocitos Mononucleares/inmunología , Masculino , Vacunas de ADN/administración & dosificación , Adulto Joven
20.
Vaccine ; 34(44): 5314-5320, 2016 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-27642130

RESUMEN

PURPOSE: GEN-003 is a candidate therapeutic HSV-2 vaccine containing a fragment of infected cell protein 4 (ICP4.2), a deletion mutant of glycoprotein D2 (gD2ΔTMR), and Matrix-M2 adjuvant. In a dose-ranging phase 1/2a clinical trial, immunization with GEN-003 reduced viral shedding and the percentage of reported herpetic lesion days. Here we examine the immune responses in the same trial, to characterize vaccine-related changes in antibody and cell-mediated immunity. METHODS: Participants with genital HSV-2 infection were randomized to 1 of 3 doses of GEN-003, antigens without adjuvant, or placebo. Subjects received 3 intramuscular doses, three weeks apart, and were monitored for viral shedding, lesions and immunogenicity. Antibody titers were measured by ELISA and neutralization assay in serum samples collected at baseline and 3weeks post each dose. T cell responses were assessed pre-immunization and 1week post each dose by IFN-γ ELISpot and intracellular cytokine staining. Blood was also collected at 6 and 12months to monitor durability of immune responses. RESULTS: Antibody and T cell responses increased with vaccination and were potentiated by adjuvant. Among the doses tested, the rank order of reduction in viral shedding follows the ranking of fold change from baseline in T cell responses. Some immune responses persisted up to 12months. CONCLUSION: All measures of immunity are increased by vaccination with GEN-003; however, a correlate of protection is yet to be defined.


Asunto(s)
Herpes Genital/inmunología , Herpes Genital/terapia , Vacunas contra el Virus del Herpes Simple/inmunología , Vacunas contra el Virus del Herpes Simple/uso terapéutico , Herpesvirus Humano 2/inmunología , Adyuvantes Inmunológicos , Adolescente , Adulto , Anticuerpos Antivirales/sangre , Relación Dosis-Respuesta Inmunológica , Ensayo de Immunospot Ligado a Enzimas , Femenino , Vacunas contra el Virus del Herpes Simple/administración & dosificación , Humanos , Inmunidad Celular , Inmunoterapia , Interferón gamma/biosíntesis , Masculino , Glicoproteínas de Membrana/inmunología , Persona de Mediana Edad , Linfocitos T/inmunología , Proteínas de la Matriz Viral/administración & dosificación , Proteínas de la Matriz Viral/inmunología , Esparcimiento de Virus , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA