Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 294
Filter
1.
Vaccine ; 42(19S1): S82-S100, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39003018

ABSTRACT

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.


Subject(s)
Herpes Genitalis , Herpes Simplex Virus Vaccines , Herpes Simplex , Herpesvirus 2, Human , Humans , Herpesvirus 2, Human/immunology , Herpes Simplex Virus Vaccines/immunology , Herpes Simplex Virus Vaccines/administration & dosage , Herpes Genitalis/prevention & control , Herpes Genitalis/immunology , Herpes Simplex/prevention & control , Herpes Simplex/immunology , Herpesvirus 1, Human/immunology , Vaccination
2.
Viruses ; 15(1)2023 01 13.
Article in English | MEDLINE | ID: mdl-36680259

ABSTRACT

Genital herpes (GH) has become one of the most common sexually transmitted diseases worldwide, and it is spreading rapidly in developing countries. Approximately 90% of GH cases are caused by HSV-2. Therapeutic HSV-2 vaccines are intended for people already infected with HSV-2 with the goal of reducing clinical recurrences and recurrent virus shedding. In our previous work, we evaluated recombinant adenovirus-based vaccines, including rAd-gD2ΔUL25, rAd-ΔUL25, and rAd-gD2, for their potency as prophylactic vaccines. In this study, we evaluated these three vaccines as therapeutic vaccines against acute and recurrent diseases in intravaginal challenged guinea pigs. Compared with the control groups, the recombinant vaccine rAd-gD2ΔUL25 induced a higher titer of the binding antibody, and rAd-gD2 + rAd-ΔUL25 induced a higher titer of the neutralizing antibody. Both rAd-gD2ΔUL25 and rAd-gD2 + rAd-ΔUL25 vaccines significantly enhanced the survival rate by 50% compared to rAd-gD2 and reduced viral replication in the genital tract and recurrent genital skin disease. Our findings provide a new perspective for HSV-2 therapeutic vaccine research and provide a new technique to curtail the increasing spread of HSV-2.


Subject(s)
Adenoviridae Infections , Adenovirus Vaccines , Herpes Genitalis , Herpes Simplex Virus Vaccines , Guinea Pigs , Animals , Herpesvirus 2, Human/genetics , Adenoviridae/genetics , Viral Envelope Proteins/genetics , Herpes Genitalis/prevention & control , Vaccines, Synthetic/genetics , Antibodies, Viral
3.
Vaccine ; 40(42): 6093-6099, 2022 10 06.
Article in English | MEDLINE | ID: mdl-36114130

ABSTRACT

Herpes simplex virus type 1 and 2 (HSV-1 and HSV-2 respectively) cause life-long latent infections resulting in recurrent orofacial and genital blisters or sores. Ensued disease can be painful and may lead to significant mental anguish of infected individuals. Currently, there are no FDA-approved vaccines for either prophylactic or therapeutic use, and recent clinical trials of subunit vaccines failed to achieve endpoints goals. Development of a safe live-attenuated herpes simplex vaccine may provide the antigenic breadth to ultimately protect individuals from acquiring HSV disease. We have previously shown that prophylactic use of the non-neurotropic live attenuated HSV-1 vaccine, VC-2, provides potent and durable protection from genital HSV-2 disease in the guinea pig model. Here, we investigated the effects of intradermal administration as well as the deletion of the viral glycoprotein G (gG) on the efficacy of prophylactic vaccination. Vaccination with either VC-2, VC-2 gG null, or gD2 MPL/Alum offered robust protection from acute disease regardless of route of vaccination. However, both the VC-2 gG-null and the ID vaccination route were more effective compared to the parent VC2 administered by the IM route. Specifically, the VC-2 gG-null administered ID, reduced HSV-2 vaginal replication on day 2 and day 4 as well as mean recurrent lesion scores more effectively than VC2 administered IM. Most importantly, only VC-2 gG null IM and VC-2 ID significantly reduced the frequency of recurrent shedding, the most likely source for virus transmission. Similarly, while all vaccinated groups demonstrated a significant reduction in the number of animals testing PCR-positive for HSV-2 in their dorsal root ganglia following challenge only VC2 ID vaccinated animals demonstrated a significant reduction in DRG viral load. All vaccinations induced neutralizing antibodies to HSV-2 MS when compared to unvaccinated guinea pigs. Therefore, further investigation of VC-2 gG null delivered ID is warranted.


Subject(s)
Herpes Genitalis , Herpes Simplex Virus Vaccines , Herpes Simplex , Herpesvirus 1, Human , Animals , Antibodies, Neutralizing , Antibodies, Viral , Female , Glycoproteins , Guinea Pigs , Herpes Genitalis/prevention & control , Herpes Simplex/prevention & control , Herpesvirus 1, Human/genetics , Herpesvirus 2, Human/genetics , Vaccination , Vaccines, Attenuated , Vaccines, Subunit , Viral Envelope Proteins/genetics
4.
J Infect Dis ; 226(9): 1489-1498, 2022 11 01.
Article in English | MEDLINE | ID: mdl-35834278

ABSTRACT

BACKGROUND: The glycoprotein D (gD)/AS04 vaccine failed to prevent herpes simplex virus (HSV) 2 in clinical trials. Failure was recapitulated in mice, in which the vaccine elicited neutralizing antibody but not antibody-dependent cell-mediated cytotoxicity (ADCC) responses. Preclinical findings suggest that ADCC is important for protection, but the clinical data are limited. We hypothesized that gD/AS04 and acute HSV-2 infection elicit primarily neutralizing antibodies, whereas ADCC emerges over time. METHODS: HSV-specific immunoglobulin G, subclass, function (neutralization, C1q binding and ADCC), and antigenic targets were compared (paired t test or Mann-Whitney U test) at enrollment and after gD/AS04 vaccination, before and after HSV-2 acquisition in vaccine controls, and in an independent cohort with chronic HSV-2 infection. RESULTS: Vaccination elicited only a neutralizing antibody response, whereas acute infection elicited neutralizing and C1q-binding antibodies but not a significant ADCC response. Antibodies to gD were exclusively immunoglobulin G1 and only neutralizing. In contrast, women with chronic HSV-2 infection had significantly greater ADCC responses and targeted a broader range of viral antigens compared with acutely infected or gD/AS04 vaccine recipients (P < .001). CONCLUSIONS: Results from gD/AS04 vaccinated or acutely infected women recapitulate murine findings of limited functional antibody responses, supporting the speculation that vaccines that generate polyfunctional and specifically ADCC responses may be required to prevent HSV-2 acquisition and limit recurrences.


Subject(s)
Herpes Simplex Virus Vaccines , Herpes Simplex , Viral Vaccines , Female , Mice , Animals , Complement C1q , Antibodies, Viral , Herpesvirus 2, Human , Antibodies, Neutralizing , Antibody-Dependent Cell Cytotoxicity , Glycoproteins , Viral Envelope Proteins
5.
J Virol ; 96(6): e0172421, 2022 03 23.
Article in English | MEDLINE | ID: mdl-35045268

ABSTRACT

Previous studies by our group identified a highly efficacious vaccine 0ΔNLS (deficient in the nuclear localization signal of infected cell protein 0) against herpes simplex virus 1 (HSV-1) in an experimental ocular mouse model. However, details regarding fundamental differences in the initial innate and adaptive host immune response were not explored. Here, we present a side-by-side analysis of the primary infection characterizing differences of the host immune response in mice infected with 0ΔNLS versus the parental, GFP105. The results show that local viral infection and replication are controlled more efficiently in mice exposed to 0ΔNLS versus GFP105 but that the clearance of infectious virus is equivalent when the two groups are compared. Moreover, the 0ΔNLS-infected mice displayed enhanced effector CD8+ but not CD4+ T cell responses from the draining lymph nodes at day 7 postinfection measured by gamma interferon (IFN-γ) and tumor necrosis factor alpha production along with changes in cell metabolism. The increased effector function of CD8+ T cells from 0ΔNLS-infected mice was not driven by changes in antigen presentation but lost in the absence of a functional type I IFN pathway. These results are further supported by enhanced local expression of type I IFN and IFN-inducible genes along with increased IL-12 production by CD8α+ dendritic cells in the draining lymph nodes of 0ΔNLS-infected mice compared to the GFP105-infected animals. It was also noted the recall to HSV-1 antigen by CD8+ T cells was elevated in mice infected with HSV-1 0ΔNLS compared to GFP105. Collectively, the results underscore the favorable qualities of HSV-1 0ΔNLS as a candidate vaccine against HSV-1 infection. IMPORTANCE Cytotoxic T lymphocytes (CTLs) play a critical role in the clearance for many viral pathogens including herpes simplex virus 1 (HSV-1). Here, we compared the cellular innate and adaptive immune response in mice infected with an attenuated HSV-1 (0ΔNLS) found to be a highly successful experimental prophylactic vaccine to parental HSV-1 virus. We found that CD8+ T cell effector function is elevated in 0ΔNLS-infected mice through noncognate signals, including interleukin-12 and type I interferon pathways along with changes in CD8+ T cell metabolism, whereas other factors, including cell proliferation, costimulatory molecule expression, and antigen presentation, were dispensable. Thus, an increase in CTL activity established by exposure to HSV-1 0ΔNLS in comparison to parental HSV-1 likely contributes to the efficacy of the vaccine and underscores the nature of the attenuated virus as a vaccine candidate for HSV-1 infection.


Subject(s)
CD8-Positive T-Lymphocytes , Herpes Simplex Virus Vaccines , Herpesvirus 1, Human , Animals , CD8-Positive T-Lymphocytes/immunology , Herpes Simplex/immunology , Herpes Simplex Virus Vaccines/immunology , Interferon-gamma/immunology , Mice , Mice, Inbred C57BL , Receptor, Interferon alpha-beta/immunology
6.
Front Immunol ; 12: 789454, 2021.
Article in English | MEDLINE | ID: mdl-34868077

ABSTRACT

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.


Subject(s)
Chemotaxis, Leukocyte , Cornea/immunology , Herpes Simplex Virus Vaccines/administration & dosage , Herpesvirus 1, Human/immunology , Intraepithelial Lymphocytes/immunology , Keratitis, Herpetic/prevention & control , Vaccination , Animals , Cornea/pathology , Cornea/virology , Cytokines/metabolism , Disease Models, Animal , Female , Herpes Simplex Virus Vaccines/immunology , Herpesvirus 1, Human/pathogenicity , Host-Pathogen Interactions , Injections, Intramuscular , Intraepithelial Lymphocytes/virology , Keratitis, Herpetic/immunology , Keratitis, Herpetic/pathology , Keratitis, Herpetic/virology , Lymphangiogenesis , Mice, Inbred BALB C , Neovascularization, Pathologic , Neutrophil Infiltration , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology
7.
Sci Rep ; 11(1): 14494, 2021 07 14.
Article in English | MEDLINE | ID: mdl-34262096

ABSTRACT

A major challenge in developing an effective vaccine against HIV-1 is the genetic diversity of its viral envelope. Because of the broad range of sequences exhibited by HIV-1 strains, protective antibodies must be able to bind and neutralize a widely mutated viral envelope protein. No vaccine has yet been designed which induces broadly neutralizing or protective immune responses against HIV in humans. Nanomaterial-based vaccines have shown the ability to generate antibody and cellular immune responses of increased breadth and neutralization potency. Thus, we have developed supramolecular nanofiber-based immunogens bearing the HIV gp120 envelope glycoprotein. These immunogens generated antibody responses that had increased magnitude and binding breadth compared to soluble gp120. By varying gp120 density on nanofibers, we determined that increased antigen valency was associated with increased antibody magnitude and germinal center responses. This study presents a proof-of-concept for a nanofiber vaccine platform generating broad, high binding antibody responses against the HIV-1 envelope glycoprotein.


Subject(s)
HIV Antibodies/metabolism , HIV Antigens/immunology , HIV Envelope Protein gp120/chemistry , HIV Envelope Protein gp120/immunology , Nanofibers/chemistry , Animals , Female , Germinal Center/immunology , HIV Antibodies/immunology , HIV Envelope Protein gp120/metabolism , Herpes Simplex Virus Vaccines/immunology , Immunoglobulin G/blood , Mice, Inbred C57BL , T-Lymphocytes, Helper-Inducer/immunology
8.
Expert Rev Vaccines ; 20(9): 1077-1096, 2021 09.
Article in English | MEDLINE | ID: mdl-34296960

ABSTRACT

Introduction: Vaccine development for the disease caused by the herpes simplex virus (HSV) has been challenging over the years and is always in dire need of novel approaches for prevention and cure. To date, the HSV disease remains incurable and challenging to prevent. The disease is extremely widespread due to its high infection rate, resulting in millions of infection cases worldwide.Areas covered: This review first explains the diverse forms of HSV-related disease presentations and reports past vaccine history for the disease. Next, this review examines current and novel HSV vaccine approaches being studied and tested for efficacy and safety as well as vaccines in clinical trial phases I to III. Modern approaches to vaccine design using bioinformatics are described. Finally, we discuss measures to enhance new vaccine development pipelines for HSV.Expert opinion: Modernized approaches using in silico analysis and bioinformatics are emerging methods that exhibit potential for producing vaccines with enhanced targets and formulations. Although not yet fully established for HSV disease, we describe current studies using these approaches for HSV vaccine design to shed light on these methods. In addition, we provide up-to-date requirements of immunogenicity, adjuvant selection, and routes of administration.


Subject(s)
Herpes Genitalis , Herpes Simplex Virus Vaccines , Herpes Simplex , Viral Vaccines , Adjuvants, Immunologic , Herpes Genitalis/prevention & control , Herpes Simplex/prevention & control , Herpesvirus 2, Human , Humans
9.
Vaccine ; 39(18): 2526-2536, 2021 04 28.
Article in English | MEDLINE | ID: mdl-33814229

ABSTRACT

The neonatal Fc receptor (FcRn) is constitutively expressed in the cornea and is up-regulated in response to herpes simplex virus type 1 (HSV-1). Previously, we found targeting cornea FcRn expression by small interfering RNA-mediated knockdown reduced the local efficacy of HSV-1 0ΔNLS vaccinated C57BL/6 mice against ocular challenge with HSV-1. The current study was undertaken to evaluate the HSV-1 0ΔNLS vaccine efficacy in FcRn deficient (FcRn KO) mice challenged with HSV-1. Whereas there was little neutralizing antibody detected in the serum of HSV-1 0ΔNLS vaccinated FcRn KO mice, these mice exhibited the same degree of protection against ocular challenge with HSV-1 as wild type (WT) C57BL/6 mice as measured by cumulative survival, infectious virus shed or retained in tissue, and corneal pathology including opacity and neovascularization. Mock-vaccinated FcRn KO mice were found to be more sensitive to ocular HSV-1 infection compared to mock-vaccinated (WT) mice in terms of cumulative survival and virus shedding. In addition, the FcRn KO mice generated significantly fewer effector (CD3+CD44+CD62L-) and central (CD3+CD44+CD62L+) memory CD8+ T cells compared to the WT mice 7 days post infection. Collectively, mock-vaccinated FcRn KO mice are susceptible to ocular HSV-1 infection but HSV-1 0ΔNLS vaccinated FcRn KO mice are resistant suggesting that in addition to the FcRn, other pathways are involved in mediating the protective effect of the HSV-1 0ΔNLS vaccine against subsequent HSV-1 challenge.


Subject(s)
Eye Diseases/virology , Herpes Simplex Virus Vaccines , Herpes Simplex/prevention & control , Receptors, Fc/genetics , Animals , CD8-Positive T-Lymphocytes , Herpesvirus 1, Human , Histocompatibility Antigens Class I , Mice , Mice, Inbred C57BL
10.
mSphere ; 6(2)2021 04 28.
Article in English | MEDLINE | ID: mdl-33910988

ABSTRACT

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.


Subject(s)
Antibodies, Viral/blood , Chemokine CCL19/immunology , Chemokines, CC/immunology , Herpes Simplex Virus Vaccines/immunology , Herpesvirus 2, Human/genetics , Herpesvirus 2, Human/immunology , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Chemokine CCL19/genetics , Chemokines, CC/genetics , Female , Herpes Simplex Virus Vaccines/administration & dosage , Herpes Simplex Virus Vaccines/classification , Immunity, Mucosal , Immunologic Memory , Mice , Mice, Inbred BALB C , Vaccination/methods , Vagina/immunology , Vagina/virology
11.
J Infect Dis ; 224(9): 1509-1519, 2021 11 16.
Article in English | MEDLINE | ID: mdl-33718970

ABSTRACT

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.


Subject(s)
Antibodies, Viral/blood , Herpes Genitalis/prevention & control , Herpes Simplex Virus Vaccines/administration & dosage , Herpes Simplex/prevention & control , Herpesvirus 2, Human/immunology , Viral Envelope Proteins/immunology , Viral Vaccines/administration & dosage , Epitopes , Herpes Simplex Virus Vaccines/immunology , Herpesvirus 1, Human/immunology , Humans , Immunization
12.
J Virol ; 95(1)2020 12 09.
Article in English | MEDLINE | ID: mdl-33028712

ABSTRACT

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.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Estradiol/immunology , Herpes Simplex Virus Vaccines/immunology , Herpesvirus 2, Human/immunology , Immunologic Memory , Interleukin-17/immunology , Vaccination/methods , Administration, Intranasal , Animals , CD8-Positive T-Lymphocytes/immunology , Estradiol/administration & dosage , Female , Herpes Genitalis/prevention & control , Herpes Simplex Virus Vaccines/administration & dosage , Immunity, Mucosal , Mice , Respiratory System/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Vagina/immunology
13.
J Virol ; 94(24)2020 11 23.
Article in English | MEDLINE | ID: mdl-32999018

ABSTRACT

The contribution of T cell and antibody responses following vaccination in resistance to herpes simplex virus 1 (HSV-1) infection continues to be rigorously investigated. In the present article, we explore the contribution of CD8+ T cells specific for the major antigenic epitope for HSV-1 glycoprotein B (gB498-505, gB) in C57BL/6 mice using a transgenic mouse (gBT-I.1) model vaccinated with HSV-1 0ΔNLS. gBT-I.1-vaccinated mice did not generate a robust neutralization antibody titer in comparison to the HSV-1 0ΔNLS-vaccinated wild-type C57BL/6 counterpart. Nevertheless, the vaccinated gBT-I.1 mice were resistant to ocular challenge with HSV-1 compared to vehicle-vaccinated animals based on survival and reduced corneal neovascularization but displayed similar levels of corneal opacity. Whereas there was no difference in the virus titer recovered from the cornea comparing vaccinated mice, HSV-1 0ΔNLS-vaccinated animals possessed significantly less infectious virus during acute infection in the trigeminal ganglia (TG) and brain stem compared to the control-vaccinated group. These results correlated with a significant increase in gB-elicited interferon-γ (IFN-γ), granzyme B, and CD107a and a reduction in lymphocyte activation gene 3 (LAG-3), programmed cell death 1 (PD-1), and T cell immunoglobulin and mucin domain-containing protein 3 (TIM-3) expressed by TG infiltrating gB-specific CD8+ T cells from the HSV-1 0ΔNLS-vaccinated group. Antibody depletion of CD8+ T cells in HSV-1 0ΔNLS-vaccinated mice rendered animals highly susceptible to virus-mediated mortality similar to control-vaccinated mice. Collectively, the HSV-1 0ΔNLS vaccine is effective against ocular HSV-1 challenge, reducing ocular neovascularization and suppressing peripheral nerve virus replication in the near absence of neutralizing antibody in this unique mouse model.IMPORTANCE The role of CD8+ T cells in antiviral efficacy using a live-attenuated virus as the vaccine is complicated by the humoral immune response. In the case of the herpes simplex virus 1 (HSV-1) 0ΔNLS vaccine, the correlate of protection has been defined to be primarily antibody driven. The current study shows that in the near absence of anti-HSV-1 antibody, vaccinated mice are protected from subsequent challenge with wild-type HSV-1 as measured by survival. The efficacy is lost following depletion of CD8+ T cells. Whereas increased survival and reduction in virus replication were observed in vaccinated mice challenged with HSV-1, cornea pathology was mixed with a reduction in neovascularization but no change in opacity. Collectively, the study suggests CD8+ T cells significantly contribute to the host adaptive immune response to HSV-1 challenge following vaccination with an attenuated virus, but multiple factors are involved in cornea pathology in response to ocular virus challenge.


Subject(s)
Antibodies, Neutralizing/immunology , Herpes Simplex Virus Vaccines/immunology , Herpes Simplex/prevention & control , Herpesvirus 1, Human/immunology , Receptors, Antigen, T-Cell/metabolism , Vaccines, Attenuated/immunology , Viral Envelope Proteins/metabolism , Animals , Antibodies, Viral , CD8-Positive T-Lymphocytes/immunology , Cornea , Female , Herpes Simplex/immunology , Immunization/methods , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Trigeminal Ganglion/metabolism , Trigeminal Ganglion/virology , Vaccination , Viral Envelope Proteins/immunology
14.
Immunohorizons ; 4(10): 608-626, 2020 10 09.
Article in English | MEDLINE | ID: mdl-33037098

ABSTRACT

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.


Subject(s)
Cornea/pathology , Herpes Simplex Virus Vaccines/administration & dosage , Herpes Simplex Virus Vaccines/immunology , Herpesvirus 1, Human/immunology , Keratitis, Herpetic/immunology , Keratitis, Herpetic/prevention & control , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Cornea/immunology , Cornea/virology , Female , Herpesvirus 1, Human/pathogenicity , Immunity, Humoral , Immunization, Passive , Keratitis, Herpetic/virology , Male , Mice , Mice, Inbred C57BL , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology , Viral Envelope Proteins/administration & dosage , Viral Envelope Proteins/immunology , Virus Shedding
15.
Vaccine ; 38(47): 7409-7413, 2020 11 03.
Article in English | MEDLINE | ID: mdl-33041105

ABSTRACT

Neonatal herpes is a dreaded complication of genital herpes infection in pregnancy. We recently compared two vaccine platforms for preventing genital herpes in female mice and guinea pigs and determined that HSV-2 glycoproteins C, D and E expressed using nucleoside-modified mRNA in lipid nanoparticles provided better protection than the same antigens produced as baculovirus proteins and administered with CpG and alum. Here we evaluated mRNA and protein immunization for protection against neonatal herpes. Female mice were immunized prior to mating and newborns were infected intranasally with HSV-2. IgG binding and neutralizing antibody levels in mothers and newborns were comparable using the mRNA or protein vaccines. Both vaccines protected first and second litter newborns against disseminated infection based on virus titers in multiple organs. We conclude that both vaccines are efficacious at preventing neonatal herpes, which leaves the mRNA vaccine as our preferred candidate based on better protection against genital herpes.


Subject(s)
Herpes Genitalis , Herpes Simplex Virus Vaccines , Herpes Simplex , Nanoparticles , Vaccines , Animals , Antibodies, Viral , Disease Models, Animal , Female , Guinea Pigs , Herpes Genitalis/prevention & control , Herpesvirus 2, Human/genetics , Lipids , Mice , Nucleosides , Pregnancy , RNA, Messenger/genetics , Viral Envelope Proteins/genetics
16.
PLoS Pathog ; 16(8): e1008703, 2020 08.
Article in English | MEDLINE | ID: mdl-32776994

ABSTRACT

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.


Subject(s)
Herpes Simplex Virus Vaccines/immunology , Herpes Simplex/immunology , Herpesvirus 1, Human/immunology , Animals , Female , Herpes Simplex/prevention & control , Herpes Simplex/virology , Herpes Simplex Virus Vaccines/administration & dosage , Herpes Simplex Virus Vaccines/genetics , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/physiology , Humans , Mice , Mice, Inbred BALB C , Mutation , Phenotype , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Viral Proteins/administration & dosage , Viral Proteins/genetics , Viral Proteins/immunology
17.
Molecules ; 25(16)2020 Aug 11.
Article in English | MEDLINE | ID: mdl-32796656

ABSTRACT

Vaccines and immunotherapies depend on the ability of antibodies to sensitively and specifically recognize particular antigens and specific epitopes on those antigens. As such, detailed characterization of antibody-antigen binding provides important information to guide development. Due to the time and expense required, high-resolution structural characterization techniques are typically used sparingly and late in a development process. Here, we show that antibody-antigen binding can be characterized early in a process for whole panels of antibodies by combining experimental and computational analyses of competition between monoclonal antibodies for binding to an antigen. Experimental "epitope binning" of monoclonal antibodies uses high-throughput surface plasmon resonance to reveal which antibodies compete, while a new complementary computational analysis that we call "dock binning" evaluates antibody-antigen docking models to identify why and where they might compete, in terms of possible binding sites on the antigen. Experimental and computational characterization of the identified antigenic hotspots then enables the refinement of the competitors and their associated epitope binding regions on the antigen. While not performed at atomic resolution, this approach allows for the group-level identification of functionally related monoclonal antibodies (i.e., communities) and identification of their general binding regions on the antigen. By leveraging extensive epitope characterization data that can be readily generated both experimentally and computationally, researchers can gain broad insights into the basis for antibody-antigen recognition in wide-ranging vaccine and immunotherapy discovery and development programs.


Subject(s)
Antibodies, Monoclonal/immunology , Antigens, Viral/immunology , Epitope Mapping/methods , Epitopes/immunology , Herpes Simplex Virus Vaccines/immunology , Herpesvirus 1, Human/immunology , Viral Envelope Proteins/immunology , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/metabolism , Antigens, Viral/metabolism , Binding, Competitive , Herpes Simplex Virus Vaccines/metabolism , High-Throughput Screening Assays , Humans , Protein Conformation , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism
18.
Antiviral Res ; 180: 104821, 2020 08.
Article in English | MEDLINE | ID: mdl-32544409

ABSTRACT

Herpes simplex virus (HSV) infections type 1 (HSV-1) and type 2 (HSV-2) are common throughout the world. Infections are lifelong and may produce both acute and recurrent vesiculoulcerative disease as well as more severe diseases. Despite disappointing results from recent HSV vaccine trials new vaccines and more potent antiviral therapies continue to be developed. These newer approaches require initial evaluations in animal models. In this review I have briefly described some of the models available and then more thoroughly describe the guinea pig model of acute and recurrent genital herpes infections. As discussed, the guinea pig model most closely mimics human disease and provides several important endpoints for evaluating vaccines and antivirals.


Subject(s)
Antiviral Agents/therapeutic use , Herpes Genitalis , Herpes Simplex Virus Vaccines/therapeutic use , Animals , Antibodies, Viral/immunology , Disease Models, Animal , Female , Guinea Pigs , Herpes Genitalis/drug therapy , Herpes Genitalis/prevention & control , Herpes Genitalis/virology , Herpesvirus 1, Human/immunology , Herpesvirus 2, Human/immunology , Humans
20.
PLoS One ; 15(2): e0228252, 2020.
Article in English | MEDLINE | ID: mdl-32027675

ABSTRACT

Herpes simplex virus type-1 (HSV-1) can cause severe ocular infection and blindness. We have previously shown that the HSV-1 VC2 vaccine strain is protective in mice and guinea pigs against genital herpes infection following vaginal challenge with HSV-1 or HSV-2. In this study, we evaluated the efficacy of VC2 intramuscular vaccination in mice against herpetic keratitis following ocular challenge with lethal human clinical strain HSV-1(McKrae). VC2 vaccination in mice produced superior protection and morbidity control in comparison to its parental strain HSV-1(F). Specifically, after HSV-1(McKrae) ocular challenge, all VC2 vaccinated- mice survived, while 30% of the HSV-1(F)- vaccinated and 100% of the mock-vaccinated mice died post challenge. VC2-vaccinated mice did not exhibit any symptoms of ocular infection and completely recovered from initial conjunctivitis. In contrast, HSV-1(F)-vaccinated mice developed time-dependent progressive keratitis characterized by corneal opacification, while mock-vaccinated animals exhibited more severe stromal keratitis characterized by immune cell infiltration and neovascularization in corneal stroma with corneal opacification. Cornea in VC2-immunized mice exhibited significantly increased infiltration of CD3+ T lymphocytes and decreased infiltration of Iba1+ macrophages in comparison to mock- or HSV-1(F)-vaccinated groups. VC2 immunization produced higher virus neutralization titers than HSV-1(F) post challenge. Furthermore, VC-vaccination significantly increased the CD4 T central memory (TCM) subsets and CD8 T effector memory (TEM) subsets in the draining lymph nodes following ocular HSV-1 (McKrae) challenge, then mock- or HSV-1(F)-vaccination. These results indicate that VC2 vaccination produces a protective immune response at the site of challenge to protect against HSV-1-induced ocular pathogenesis.


Subject(s)
Herpes Simplex Virus Vaccines/immunology , Herpes Simplex/prevention & control , Herpesvirus 1, Human/pathogenicity , Animals , Antigens, Viral/immunology , Cornea/pathology , Cornea/virology , Female , Herpes Simplex/pathology , Herpes Simplex/veterinary , Herpesvirus 1, Human/metabolism , Humans , Injections, Intramuscular , Mice , Mice, SCID , Vaccination
SELECTION OF CITATIONS
SEARCH DETAIL
...