Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Mar 20.
Published in final edited form as: Vaccine. 2013 Sep 6;32(14):1553–1560. doi: 10.1016/j.vaccine.2013.08.066

Current status and prospects for development of an HSV vaccine

Christine Johnston 1,2, David M Koelle 1,2,3,5,6, Anna Wald 1,2,3,4
PMCID: PMC4106293  NIHMSID: NIHMS614175  PMID: 24016811

Abstract

Herpes simplex virus type 2 (HSV-2) infects 530 million people, is the leading cause of genital ulcer disease, and increases the risk of HIV-1 acquisition. Although several candidate vaccines have been promising in animal models, prophylactic and therapeutic vaccines have not been effective in clinical trials thus far. Negative results from the most recent prophylactic glycoprotein D2 subunit vaccine trial suggest that we must reevaluate our approach to HSV-2 vaccine development. We discuss HSV-2 pathogenesis, immunity, and vaccine efforts to date, as well as the current pipeline of candidate vaccines and design of trials to evaluate new vaccine constructs.

HSV Epidemiology: Global Burden of Disease

Herpes simplex virus type 2 (HSV-2) is an incurable sexually transmitted pathogen that infects over 500 million people worldwide and causes an estimated 23 million new infections annually [1]. In the United States, direct annual medical costs associated with HSV-2 are estimated to be $541 million dollars, making it the third most costly STI after HIV-1 and human papillomavirus (HPV) [2]. HSV-2 seroprevalence ranges from 16% among 14-49 year olds in the United States [3], to >80% in areas of sub-Saharan Africa [4]. HSV-2 infection rates in heavily exposed populations are nearly 100%, suggesting universal susceptibility [5]. Seroprevalence in women is up to twice as high as men, and increases with age [3, 6]. Although HSV-2 is the leading cause of genital ulcer disease (GUD) worldwide [7, 8], most people are unaware of having the infection [9].

HSV-2 transmission occurs through genital-genital contact during sexual activity. HSV-2 may be transmitted in the absence of signs or symptoms of infection in the infected partner, during episodes of subclinical shedding [10]. In addition, most people who acquire HSV-2 are asymptomatic at the time of acquisition [11]. Transmission of HSV from mother to infant during birth is a serious complication of genital herpes, and can result in long-term neurologic sequelae or mortality [12]. Women who acquire HSV during pregnancy are at the highest risk of transmitting the infection [13]. With an estimated incidence of 4-31/100,000 live births [14, 15], neonatal herpes is too rare to be used as an endpoint in a clinical trial. However, prevention of HSV acquisition during pregnancy is an important goal of developing an effective HSV vaccine.

The greatest public health impact of HSV-2 infection is its role in promulgating the HIV-1 epidemic. Persons with HSV-2 infection are 3-fold more likely to acquire HIV-1 infection [16]; this risk increases up to 8-fold if the exposure occurs soon after acquiring HSV-2 infection [17, 18]. In HIV-1 infected persons, HIV-1 is found in HSV-2 genital ulcers [19], and persons with genital ulcers are at increased risk of transmitting HIV-1 [20]. In regions with high HSV-2 seroprevalence (>80%), 25-50% of HIV-1 infections are attributable to HSV-2 [21]. Mathematical models suggest that even moderately effective prophylactic HSV-2 vaccines would lead to a marked decrease in HIV-1 incidence if given at high coverage [22]. The biologic basis for this predisposition is the persistent mucosal inflammatory response induced by HSV-2. Genital biopsy studies have revealed that HSV-2 ulcers are associated with an infiltrate of CD4+ T-cells bearing the HIV-1 co-receptors CCR5 or CXCR4, which persists during daily antiviral therapy for HSV [23]. Histopathologic studies of foreskins from HIV-1-seronegative men demonstrate that HSV-2 seropositive men have increased concentration of CD4+ and CD8+ T- cells as compared to HSV-2 seronegative men [24]. Similar findings have been found in cervical cytobrush samples from HIV-1 negative, HSV-2 seropositive women [25].

Currently available HSV-2 prevention strategies are inadequate; each reduces the risk of transmission by approximately 50%. Evidence-based methods include use of suppressive antiviral therapy [26], disclosure of serostatus to susceptible partners [27], and consistent condom use [28]. While male circumcision decreases the risk of HSV-2 acquisition by nearly 30% [29], there are conflicting data about the role of circumcision in transmission to women [30, 31]. These partly effective strategies may be useful for management of individual patients, but they are unlikely to be of public health benefit. Indeed, even with availability of suppressive antiherpes viral therapy in the USA, seroprevalence rates are similar to the pre-antiviral era [6].

Changing epidemiology of genital herpes: role of HSV-1

Over the past 2 decades, incident genital herpes in developed countries is increasingly caused by HSV type 1 (HSV-1), especially in persons <25 years of age [32]. This is likely due to declining seroprevalence of HSV-1 in adolescents [6], resulting in the first mucosal exposure to HSV-1 at initiation of sexual activity. As HSV-1 and HSV-2 have similar pathogenesis and host interactions, concepts for effective vaccine development may be relevant to both viruses. Infection with HSV-2 provides partial protection against HSV-1 [15], but the reverse is not true [33]. We need more information about HSV-1 genital infection, the risk of transmission to sex partners and neonates, and interactions between HIV-1 and HSV-1. Vaccines which provide protection against genital HSV-1 infection will be important to reduce genital herpes and its’ sequelae.

New insights into HSV pathogenesis: Frequent and dynamic reactivation

During primary infection, HSV infects epithelial cells at skin and mucosa surfaces and is transported along nerve axons to the dorsal root ganglia (DRG), where latency is established [34]. Neuronal cells are not destroyed during initial HSV infection and provide a reservoir for latent virus [35]. During reactivation the virus travels from the ganglia back to the skin and results in detection of virus (“viral shedding”) from epithelial surfaces. Viral reactivation is most often asymptomatic, but may be associated with genital symptoms or ulcers. Recent studies have demonstrated that episodes of genital HSV reactivation last a median of 13 hours and are likely rapidly cleared by host responses [36-38]. These may include tissue resident memory (TRM) T cells, discussed below, and suggest that frequent antigen exposure stimulates a chronic immune response in the mucosa.

The Immune Response to HSV-2

Murine HSV models are useful for basic HSV immunology [39], but mimic neither primary nor recurrent human infection. Guinea pigs experience recurrent infection [40], but tools for mechanistic studies are poor, and other models have practical problems or poor evidence for seroconversion [41, 42]. The host and viral determinants of the heterogeneous clinical and virological manifestations of genital HSV-2 in humans are poorly understood. Identification of the components of the host immune system that contain viral reactivation from neurons and promote viral clearance from the mucosa will be essential for development of a successful HSV- 2 vaccine. This information will be gained by detailed immunologic and genetic studies of persons with well-defined HSV-2 severity.

The importance of the innate immune system has been demonstrated by observations that human mutations in a TLR3-centric pathway are associated with severe primary HSV infection [43]. While TLR3 is required for priming CD8+ T-cell responses to HSV infection [44] and can be manipulated by adjuvants [45], data linking variation in TLR3 and recurrent HSV severity in humans are conflicting [46-48]. Variation in other host loci involved in immunity may be associated with HSV severity [49], but the ability manipulate these with vaccines is limited at this time. These findings suggest that adjuvant which promotes innate immune responses may be important for an HSV vaccine.

Antibody-driven vaccines remain of intense interest. The rationale for pursuing neutralizing antibodies is based on the biology of perinatal HSV transmission in the absence vs. presence of pre-existing maternal antibody [15], and animal passive transfer studies [50]. Neutralizing antibody titers correlate with protection to HPV infection, another epithelial STI [51]. The step- wise process of HSV entry, starting with glycoprotein (g)D binding to cell-type specific high affinity receptors and subsequent gB-mediated fusion with mandatory involvement by the gH-gL heterodimer, is becoming clear from structural biology and mutational work [52-55]. Recent advances in human B-cell cloning, high throughput antibody screening, sequencing and expression, and crystallization of complexes of antigens with highly favorable antibodies, as exemplified by HIV-1 and influenza [56, 57] could yield improved HSV immunogen designs.

Evidence is now emerging in both human and murine studies that local T-cells can serve as epithelial sentinels to provide a surveillance function to modulate primary and re-infection episodes. Using in situ methods, prolonged residence of HSV-2-specific CD8+ T-cells was documented at the dermo-epidermal junction (DEJ) in humans [58]. These cells have an activated phenotype and a unique expression pattern of homing-related molecules [59]. Elegant murine studies prove prolonged residence of HSV-specific CD8+ T-cells that is spatially limited to sites of previous infection and capable of mediating local protection to exogenous re- scarification, the best model of recurrence in this system [60]. Recently, systemic vaccination with replication-competent, attenuated HSV-2 was followed by a chemoattractant therapy given vaginally in mice [39]. This was found to “pull” vaccine-primed cells to the site of challenge, and to mediate long-lived functional protection [39], providing direct evidence of the importance of CD8 T cells. While vaginal administration of pro-inflammatory chemokines or upstream innate stimuli is challenging in humans, this is an important conceptual advance, establishing the ability to develop tissue resident-memory (TRM) cells without local infection. Mathematical models suggest that small fluctuations in TRM levels could tip the balance between subclinical and clinical reactivation [38]. Therefore, understanding protective T cell responses and stimulating such responses through a vaccine is an ongoing research priority.

At the whole pathogen level, the integrated CD4 and CD8 response in chronically infected persons occupies about 0.1 to 3% of the PBMC compartment [61, 62]. We found no correlation between the magnitude or functionality (IFN-γ, IL-2, TNF-α) of the integrated CD4 responses to whole HSV-2 and shedding or clinical severity in a cross-section survey [61]. Thus, target CD4 levels for preventative vaccines are hard to define, and simply boosting pre-existing CD4 responses may not be rational for immunotherapy. Because HSV-1 and HSV-2 have immune evasive mechanisms and are directly cytotoxic to activated lymphocytes, measuring the size or phenotype of the integrated CD8 response to the whole virus has been challenging. Whether a critical level or phenotype of circulating CD8 responses will correlate with vaccine success is unknown.

Recently developed tools which contain every HSV-1 and HSV-2 open reading frame allow examination of responses at antigen-and epitope-specific levels [62, 63]. Using this unbiased proteomic approach, we found that CD4+ and CD8+ T-cells in HSV-1 infected humans recognize an average of 17 and 22 ORFs, respectively, with a high population prevalence of both CD8 and CD4 responses to UL39, encoding an enzyme, and UL46, encoding a tegument protein [62]. These inherently immunogenic proteins are thus potential candidates for a multivalent subunit approach. Responses to individual epitopes and proteins have been correlated with symptom status [64, 65]. A cross-sectional HSV-2 proteome approach in cohorts with clinically defined severity was used to select partial-length HSV-2 ORFs for an adjuvanted, multivalent subunit candidate [66]. These diversity data argue that vaccine candidates using whole viruses are more likely to mimic natural infection with regards to antigenic complexity, albeit whether this is desirable or required is unknown.

Within these poly-specific responses, a pattern of immunodominance is perceptible for both CD8+ and CD4+ T-cell responses. Cells specific for some CD8+ T-cell epitopes are detectable directly ex vivo by tetramers or other methods [67], while responder cells specific for most CD8 epitopes are below the limit of detection for most sensitive ex vivo method [62]. This implies a steep immunodominance curve, as noted in mice [68]. The dominant epitopes tend to be in tegument and capsid proteins [69]. Dominant CD4 epitope recognition included glycoprotein and regulatory immediate early proteins [70]. Further studies of correlates of immunity using the proteome may identify potentialvaccine candidates.

Predictably, HSV-specific CD8+ and CD4+ T-cells are found at sites of clinically evident recurrent infection [71], because responder cells must physically contact antigen presenting cells (APC). Infiltration of antigen-specific cytotoxic cells correlates with resolution of recurrent genital herpes, and priming or augmenting such cells makes sense for vaccines. The molecular mechanism for homing includes CLA on T-cells and endothelial E-selectin in inflamed tissues [72]. HSV-specific T-cells also localize to the cervix during recurrences in women, to infected ocular tissues, and to the human trigeminal ganglia (for HSV-1) [73-75], suggesting that HSV proteins are expressed at immunologically perceptible sites within the peripheral nervous system throughout the life of the infected host. Mathematical models based on shedding data mirror these findings, and support the view that HSV reactivation is a frequent process with a slow “drip” of virions that are released into the axons [76].

Clinical trials of established HSV vaccine platforms

Prophylactic Vaccines

Several platforms have been tested for prophylactic HSV-2 vaccines; these have been recently reviewed [77]. The most promising and advanced have been recombinant glycoprotein vaccines, with more than 20,000 human volunteers studied in clinical trials. Four envelope glycoproteins elicit neutralizing antibodies to HSV: gD, gB, gH, and gL. The first two are particularly attractive as they bind to high affinity receptors or are involved in membrane fusion, respectively, and are sequence-conserved between strains and relatively conserved between HSV-2 and HSV-1. A recombinant bivalent gB2 and gD2 subunit vaccine formulated with an oil/water emulsion adjuvant was safe and induced strong neutralizing antibody and CD4+ T-cell responses [78, 79] in humans. However, this vaccine did not prevent HSV-2 infection in at-risk members of discordant heterosexual couples or STD clinic enrollees [78]. Two parallel studies showed that a recombinant secreted gD2 subunit vaccine with an adjuvant containing alum and a biologically-derived TLR4 agonist, 3-O-deacylated monophosphoryl lipid A (MPL) induced both neutralizing antibody and CD4+ immune responses [80] in HSV-2 seronegative persons in an HSV-2 discordant sexual relationship. Although the vaccine did not prevent HSV-2 in men or HSV-1 seropositive women, HSV-2 disease was reduced by 70% and HSV-2 infection by 40% in a subgroup analysis of HSV-1 and HSV-2 seronegative women who received vaccine [81]. In a follow-up trial, 8323 sexually active HSV-1/HSV-2 seronegative women in North America received three doses of the gD2 vaccine or control [82]. Unfortunately, the gD2 vaccine failed to prevent HSV-2 infection or disease. However, gD2 vaccine was associated with significant decrease in HSV-1 infection (35% efficacy) and genital disease (58% efficacy). Lower gD2 antibody titers were associated with acquisition of HSV-1 but not HSV-2, suggesting a potential correlate of protection [82]. The magnitude of CD4+ T-cell responses to gD2 was not associated with prevention of HSV infection; CD8+ T-cell responses were not detected. This finding provides proof of concept that an HSV-2 vaccine may also target HSV-1, suggesting potential for cross-reactive immunity [83]. It is unclear why this vaccine showed partial efficacy in HSV-1/2 seronegative women, but not men; differences in the anatomic site at risk of infection (stratum corneum for men, mucosal surfaces, which may contain more vaccine induced immune responses for women), or differences in the immune response to vaccination have been hypothesized to be possible explanations [81].

Therapeutic Vaccines

While an early study of a recombinant gD2 vaccine adjuvanted with alum reduced the rate of virologically confirmed recurrences one year post vaccination [84], later studies of glycoprotein vaccines were not effective [85]. Participants with frequent genital HSV-2 recurrences who received a live, attenuated growth compromised strain of HSV-2 with a deletion in UL39 (ICP10ΔPK) had decreased self-reported recurrences as compared to placebo [86]. Importantly, this construct was safe, providing proof-of-concept for replication competent vaccine constructs. A replication defective HSV-2 strain with a gH deletion which was able to undergo a single cycle of replication (disabled infectious single cycle, DISC) had similar time to first recurrence, lesion healing rates, and genital shedding rates in HSV-2 seropositive persons with recurrent genital herpes as placebo [87].

Future Clinical Trials: Population, Design & Endpoints

Prophylactic Vaccination

Safe and effective prevention of genital HSV infection is the ultimate goal HSV vaccine research. Because the correlate of protective immunity is unknown, testing the efficacy of prophylactic HSV vaccines requires prospective follow up of persons at risk for genital HSV acquisition. Prior prophylactic vaccine trials have been performed almost exclusively in North America, where the HSV-2 acquisition rate is low. In the per-protocol analysis of the recent gD2 subunit vaccine study, only 1.6% of participants acquired HSV-2 infection, and 1.0% had genital ulcer disease due to HSV-1 or HSV-2, the primary endpoint [82]. In contrast, HSV-2 is rapidly acquired among men and women initiating sexual activity in sub-Saharan Africa, with incidence up to 23 per 100 person years [88]. Prophylactic HSV-2 vaccine studies should be performed in international settings, where the greatest burden of disease exists. Multi-national trials are also important since there may be geographical strain differences which affect HSV-2 pathogenicity and immunogenicity [89]. It will be important to understand genotypic and phenotypic variation in HSV-2 strains from around the world prior to performing these trials, as these differences may affect vaccine efficacy [89]. Synergy with established networks, such as the HIV Vaccine Trials Network (HVTN), should be explored. Young women are at highest risk for acquiring HSV-2, and serve as an ideal population for prophylactic vaccine trials. Given the sex differences in vaccine efficacy from the gD2 vaccines, it may be important to power trials to stratify vaccine efficacy by sex.

As the efficacy of a vaccine may be different in persons who are HSV-1 seropositive and seronegative, both populations should be evaluated. Importantly, HSV-1 is often acquired early in childhood, especially in resource-limited settings, which may shift the optimal time for vaccination to infancy/early childhood. A vaccine targeting both HSV-1 and HSV-2 could be tested in parallel in HSV-1/HSV-2 seronegative children for prevention of HSV-1 infection.

Acquisition of HSV-1 and HSV-2, measured by seroconversion, should be the primary endpoints for a prophylactic vaccine study. As HSV-2 infection is often subclinical measurement of clinical disease as a primary endpoint is problematic. An important feature of candidate vaccines will be modification of the construct so that an antibody assay can distinguish between vaccinated and infected persons. Secondary endpoints should include frequency of clinically apparent HSV genital disease, and in those who seroconvert, frequency of genital viral shedding. Mathematical modeling suggests that even low efficacy preventative vaccine could impact the HSV-2 epidemic by decreasing shedding and reducing viral transmission [90]. Such a vaccine would have the highest impact in high-prevalence populations [91]; for instance, a vaccine which marginally decreases HSV-2 susceptibility but reduces shedding frequency by 75% could reduce HSV-2 incidence by 30% over a 10 year period [92]. Thus, it is important to study both acquisition, and in those who acquire, frequency of viral shedding.

Therapeutic vaccines

An effective therapeutic HSV-2 vaccine could both improve the clinical course in individual patients, and decrease HSV transmission through reduction in shedding, for a public health benefit. The approach to efficiently evaluate such vaccines relies on evaluation of viral shedding in a cohort of highly adherent persons with clinically apparent genital HSV-2; we have found that this population is highly motivated to participate in daily genital shedding studies [93]. The participants obtain genital swabs for detection of viral shedding before and after vaccination in a one-way crossover study design. These studies are ideal for proof-of-concept, as they can rapidly provide an answer to whether the vaccine has efficacy and can be efficiently performed with fewer than 100 persons [94]. Reduction in viral shedding is the more sensitive primary endpoint for therapeutic vaccine trials, and serves as a useful surrogate endpoint for recurrence rate and transmission likelihood. As initial therapeutic vaccine trials should target persons with symptomatic infection, important secondary endpoints include frequency of genital lesions and prodromal symptoms. These are the clinical endpoints that have been requested in the past by FDA for licensure studies. In addition, the density of HIV receptor-positive cells in the genital mucosal following therapeutic immunization will need to be evaluated.

The Pipeline

Although prior vaccines that have made it to human clinical trials have almost exclusively targeted glycoproteins, the HSV vaccine pipeline is rich with novel platforms that have shown efficacy in animal models (Table 1). The challenge will be quickly moving these candidate vaccines into human clinical trials.

Table 1.

HSV Vaccine Pipeline.

Vaccine Name Construct Stage of
development
HSV-2 functionally mutated
for ICP0 [1]
0ΔNLS Replication-competent whole virus Preclinical
HSV-1 functionally mutated
for UL43, UL49.5, UL55,
UL56, and LAT expression [2]
HF10 Replication-competent whole virus Preclinical
HSV2 glycoprotein E deletion
mutant [3]
HSV2 gE mutant Replication-competent whole virus Preclinical
HSV-2 functionally mutated
for γ34.5, UL43.5, UL55-56
US10, US11, US12 [4]
AD472 Replication-competent whole virus Preclinical
HSV-2 functionally mutated
for UL5/UL29 [5-7]
ACAM-529 Replication-incompetent whole virus Preclinical
HSV-2 glycoprotein D
dominant negative,
functionally mutated for
ICP0/UL9 [8]
CJ2-gD2 Replication-incompetent whole virus Preclinical
HSV-2 functionally mutated
for thymidine kinase (TK) [9]
Replication-competent whole virus,
followed by topical vaginal
application of CXCL9 and CXCL10
Preclinical
HSV-2 functionally mutated
for ICP10 [10, 11]
ICP10APK Replication-competent whole virus. No active program,
has been in phase
I/II, therapeutic
HSV-2 functionally mutated
for ICP47, vhs, γ34.5, US5,
UL43 [12]
ImmunoVEXHSV2 Replication-competent whole virus Phase I
Inactivated HSV-2 in
MPL/alum [13, 14]
Formalin-inactivated virus
administered after DNA boost
Preclinical
HSV-1 glycoprotein B [15] Lentiviral vector Preclinical
Recombinant secreted HSV-1
glycoprotein B [16]
Intranasal immunization with
immunogen in non-ionic surfactant
vesicles
Preclinical
Recombinant HSV-2 gD [17] Recombinant gD2 with IC31 adjuvant Preclinical
Recombinant HSV-2 gD [18] gD2 DNA prime followed by
intranasal protein boost (liposomal
encapsulation)
Preclinical
gD2/UL46/UL47 DNA [19] gD2-Vaxfectin Plasmid gD2/UL46/UL47 polyvalent
DNA with cationic lipid adjuvant
Phase I therapeutic
announced
Modified gD2 DNA [20] Plasmid gD2 fused to ubiquitin mixed
with codon-optimized non-linked gD2
Pre-clinical
32 unique 35-mer HSV-2
peptides [21]
HerpV Synthetic peptides, multivalent with
heat shock protein adjuvant
Phase II,
therapeutic
gD2 and ICP4 [22] GEN-003/MM2 Recombinant bivalent proteins with
Iscomatrix adjuvant
Phase 1 b/ II,
therapeutic

UL=unique long genes, ICP=infected cell polypeptide. vhs=virion host shutoff protein

1

Halford WP, Puschel R, Gershburg E, Wilber A, Gershburg S, Rakowski B. A Live-Attenuated HSV-2 ICP0(−) Virus Elicits 10 to 100 Times Greater Protection against Genital Herpes than a Glycoprotein D Subunit Vaccine. PLoS One. 2011;6:e17748.

2

Luo C, Goshima F, Kamakura M, Mutoh Y, Iwata S, Kimura H, et al. Immunization with a highly attenuated replication-competent herpes simplex virus type 1 mutant, HF10, protects mice from genital disease caused by herpes simplex virus type 2. Frontiers in Microbiology. 2012;3.

3

Awasthi S, Zumbrun EE, Si H, Wang F, Shaw CE, Cai M, et al. Live Attenuated Herpes Simplex Virus Type 2 Glycoprotein E Deletion Mutant as a Vaccine Candidate Defective in Neuronal Spread. Journal of Virology. 2012.

4

Prichard MN, Kaiwar R, Jackman WT, Quenelle DC, Collins DJ, Kern ER, et al. Evaluation of AD472, a live attenuated recombinant herpes simplex virus type 2 vaccine in guinea pigs. Vaccine. 2005;23:5424-31.

5

Hoshino Y, Pesnicak L, Dowdell KC, Lacayo J, Dudek T, Knipe DM, et al. Comparison of immunogenicity and protective efficacy of genital herpes vaccine candidates herpes simplex virus 2 dl5-29 and dl5-29-41L in mice and guinea pigs. Vaccine. 2008;26:4034-40.

6

Hoshino Y, Pesnicak L, Dowdell KC, Burbelo PD, Knipe DM, Straus SE, et al. Protection from Herpes Simplex Virus (HSV)-2 Infection with Replication-Defective HSV-2 or Glycoprotein D2 Vaccines in HSV-1-Seropositive and HSV-1-Seronegative Guinea Pigs. Journal of Infectious Diseases. 2009;200:1088-95.

7

Da Costa X, Kramer MF, Zhu J, Brockman MA, Knipe DM. Construction, phenotypic analysis, and immunogenicity of a UL5/UL29 double deletion mutant of herpes simplex virus 2. J Virol. 2000;74:7963-71.

8

Akhrameyeva NV, Zhang P, Sugiyama N, Behar SM, Yao F. Development of a Glycoprotein DExpressing Dominant-Negative and Replication-Defective Herpes Simplex Virus 2 (HSV-2) Recombinant Viral Vaccine against HSV-2 Infection in Mice. Journal of Virology. 2011;85:5036-47.

9

Shin H, Iwasaki A. A vaccine strategy that protects against genital herpes by establishing local memory T cells. Nature. 2012;491:463-7.

10

Casanova G, Cancela R, Alonzo L, Benuto R, Magana Mdel C, Hurley DR, et al. A double-blind study of the efficacy and safety of the ICP10deltaPK vaccine against recurrent genital HSV-2 infections. Cutis. 2002;70:235-9.

11

Aurelian L, Kokuba H, Smith CC. Vaccine potential of a herpes simplex virus type 2 mutant deleted in the PK domain of the large subunit of ribonucleotide reductase (ICP10). Vaccine. 1999;17:1951-63.

12

Thomas S, Barton S, Reay P, Marshall T, Love C, Goldsweig H, et al. Clinical development of an immune evasion gene-deleted live attenuated vaccine for HSV-2 (ImmunoVEX HSV2). International Herpesvirus Workshop. Salt Lake City, UT2010.

13

Morello CS, Levinson MS, Kraynyak KA, Spector DH. Immunization with herpes simplex virus 2 (HSV-2) genes plus inactivated HSV-2 is highly protective against acute and recurrent HSV-2 disease. J Virol. 2011;85:3461-72.

14

Morello CS, Kraynyak KA, Levinson MS, Chen Z, Lee K-F, Spector DH. Inactivated HSV-2 in MPL/alum adjuvant provides nearly complete protection against genital infection and shedding following long term challenge and rechallenge. Vaccine. 2012;30:6541-50.

15

Chiuppesi F, Vannucci L, De Luca A, Lai M, Matteoli B, Freer G, et al. A Lentiviral Vector-Based, Herpes Simplex Virus 1 (HSV-1) Glycoprotein B Vaccine Affords Cross-Protection against HSV-1 and HSV-2 Genital Infections. Journal of Virology. 2012;86:6563-74.

16

Cortesi R, Ravani L, Rinaldi F, Marconi P, Drechsler M, Manservigi M, et al. Intranasal immunization in mice with non-ionic surfactants vesicles containing HSV immunogens: A preliminary study as possible vaccine against genital herpes. International Journal of Pharmaceutics. 2013;440:229-37.

17

Wizel B, Persson J, Thörn K, Nagy E, Harandi AM. Nasal and skin delivery of IC31®-adjuvanted recombinant HSV-2 gD protein confers protection against genital herpes. Vaccine. 2012;30:4361-8.

18

Tirabassi RS, Ace CI, Levchenko T, Torchilin VP, Selin LK, Nie S, et al. A mucosal vaccination approach for herpes simplex virus type 2. Vaccine. 2011;29:1090-8.

19

Veselenak RL, Shlapobersky M, Pyles RB, Wei Q, Sullivan SM, Bourne N. A Vaxfectin®-adjuvanted HSV-2 plasmid DNA vaccine is effective for prophylactic and therapeutic use in the guinea pig model of genital herpes. Vaccine. 2012;30:7046-51.

20

Dutton J, Li B, Woo WP, Marshak J, Xu Y, Huang M, et al. Protection against viral challenge in a murine model of HSV-2 infection conferred by mixed DNA vaccines, Abstract 6.52. 37th Annual Herpesvirus Workshop. Calgary, Alberta, Canada 2012.

21

Wald A, Koelle DM, Fife K, Warren T, LeClair K, Chicz RM, et al. Safety and immunogenicity of long HSV-2 peptides complexed with rhHsc70 in HSV-2 seropositive persons. Vaccine. 2011;29:8520-9.

22

Skoberne M, Cardin R, Lee A, Kazimirova A, Zielinski V, Garvie D, et al. An Adjuvanted Herpes Simplex Virus Type 2 (HSV-2) Subunit Vaccine Elicits a T Cell Response In Mice and Is an Effective Therapeutic Vaccine In Guinea Pigs. Journal of Virology. 2013.

Whole virus approaches: Replication-competent

There has been concern about safety of replication-competent vaccines due to possibility of recombination with clinical strains or the establishment of latency. However, given the success of the varicella zoster virus (VZV) vaccine, a related alphaherpesvirus, in preventing chicken pox in children and shingles in adults, this is an approach that may reduce the requirement for knowledge of correlates of immunity. Since seroconversion is an appropriate primary outcome in prophylactic vaccine studies, constructs based on whole virus will need to include a serologic marker that identifies the immune response as vaccine- rather than natural infection-specific. Several candidates have yielded promising results in animal models. An HSV-2 ICP0 deletion mutant protected mice from infection, and was more potent than a gD2 subunit approach [95]. HF10 is a highly attenuated naturally occurring HSV-1 mutant that does not express latency associated transcripts and other important viral proteins such as the UL49.5 product and which prevented genital symptoms, systemic disease, and death after intravaginal HSV-2 challenge in mice [96]. Another attractive replication-competent candidate is an HSV-2 glycoprotein E mutant, which is unable to spread from epithelial cells to neuronal cells [97]. In the guinea pig model, the HSV-2 glycoprotein E mutant has shown potential both as a prophylactic and therapeutic vaccine, although it was unable to completely prevent challenge virus infection or recurrent vaginal shedding [98]. Importantly, infectious glycoprotein E mutant virus was not recovered from dorsal root ganglia or spinal cord in mouse models, although vector DNA was found in the DRG in a minority of animals [98]. AD472, a vaccine strain with deletions in γ34.5 and several other genes designed to improve genetic stability of the virus also protected guinea pigs, but similar to the glycoprotein E mutant, was not able to prevent wild-type infection [99].

Replication-incompetent

These candidates cannot replicate in normal human cells and therefore, cannot establish latency. This inherent safety advantage may be counterweighed by weaker immunogenicity, possibly requiring higher doses and/or repeated dosing. dl5-29 is a double mutant with deletions in UL5 and UL29, two genes which are essential for viral replication [100]. This construct protected against infection and recurrences in the guinea pig model [101]. In both HSV-1 seropositive and HSV-1 seronegative animals, vaccination with dl5- 29 resulted in decreased vaginal shedding after challenge compared with gD2 subunit vaccines [102]. Recently described improvements in production and purification of this construct may make it scalable for clinical testing [103]. Another novel replication-incompetent mutant is CJ- gD2, in which both copies the ICP0 gene are replaced by gD2 controlled by HSV-1 ICP4 promoter, resulting in gD2 expression at wild type levels and protection from HSV-2 in the murine model [104].

Other approaches

Other novel platforms include formalin inactivated HSV-2 formulated with MPL/alum adjuvant, which protected guinea pigs from primary infection and recurrent disease when given with a DNA prime [105], and induced neutralizing antibody and HSV-2 specific CD4+ T-cells [106]. In a lentiviral vector delivery system, HSV-1 glycoprotein B expressed in feline immunodeficiency virus vector showed cross-protection against both HSV-1 and HSV-2 vaginal challenge in mice [107]. A plasmid based vaccine which includes gD2, UL46 and UL47 formulated with a novel cationic lipid-based adjuvant was effective as a prophylactic and therapeutic vaccine in guinea pigs [108].

Novel routes of delivery are also being evaluated. With increasing evidence for importance of TRM T-cells, there is growing interest in stimulation of genital mucosal immunity through mucosal delivery methods. For instance, intranasal delivery of gB1 packaged in non-ionic surfactant vesicles protected mice from HSV-2 vaginal challenge [109]. Mucosal immunization with gD2 adjuvanted with IC31 [45] or given in a DNA prime followed by a protein boost delivered through liposomal encapsulation [110], both of which stimulate a Th1 response, protected mice from HSV-2 vaginal challenge. Combining the DNA approach with trans-dermal microneedle delivery was found to have a dose-sparing effect in mice; localization of the effector cells is undefined[111]. The “prime-pull” approach in which mice were immunized followed by application of chemokine to genital area is another novel approach that will require further study [39].

There are two ongoing Phase I/II trials of therapeutic vaccines which use novel antigens and adjuvants. One vaccine design consists of 32 35-mer HSV-2 peptides directed against 22 HSV- 2 proteins complexed with human heat shock protein 70 and saponin adjuvant. This vaccine increased detection of HSV-2 specific CD4+ and CD8+ T-cell responses in HSV-2 seropositive persons and was safe in a Phase I trial [112], and is being tested in a Phase II trial for prevention of shedding and lesions (NCT01687595). A subunit vaccine containing secreted gD2, and truncated ICP4, which was identified as a CD8+ T-cell antigen through a high- throughput proteomic screening method, formulated with an adjuvant to stimulate humoral and cellular immunity, showed efficacy against infection and recurrent disease in the guinea pig model [66], and is being tested in a Phase I/II trial as a therapeutic vaccine (NCT01667341).

Conclusions

The field of HSV vaccines is rapidly evolving. Although the results of the prophylactic glycoprotein D2 vaccine were disappointing, the field has been reenergized by improved understanding of the frequency of viral shedding, the importance of the mucosal immune response, availability of novel adjuvants and delivery mechanisms, identification of T cell epitopes via proteomic screening and advancement in replication competent and replication- incompetent candidates. In addition, we have learned from past vaccine studies; we need to depend on objective evidence of seroconversion rather than the variable phenotype of clinical disease in preventative vaccine studies. Similarly, viral shedding should be measured rather than recurrence rate as the most sensitive tool to evaluate both prophylactic and therapeutic vaccines, to provide data on potential impact of vaccination on HSV transmission. Given the increasing incidence of genital HSV-1, we must consider a vaccination strategy that will provide cross-protection against both HSV-1 and HSV-2, which may ultimately shift the optimal timing of vaccination from adolescence to childhood. Finally, prophylactic vaccines must be tested in populations with high prevalence and incidence of genital HSV-2, as this will provide the benefit of rapid evaluation of candidate vaccine in the populations where it is most desperately needed.

Highlights.

  • HSV-2 is a highly prevalent infection that triples the risk of HIV-1 acquisition

  • Prophylactic glycoprotein subunit vaccines have failed to decrease HSV-2 acquisition

  • Identification of T-cell epitopes and new understanding of mucosal immunity have advanced vaccine development

  • Re-evaluation of study population and endpoints for HSV vaccine trials is needed

  • The HSV vaccine pipeline is rich with promising novel strategies in preclinical phase

Acknowledgments

Funding: National Institutes of Health K24 AI071113 (AW), PO1 AI30731 (CJ, AW, DMK), R01 AI094019 (DMK)

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Conflicts of Interest: CJ, DMK, and AW receive research funding from NIH. CJ has received research funding from AiCuris. DMK is listed as a co-inventor on patents describing T-cell responses to HSV-2, receives funding from Immune Design Corporation, and is a consultant to Agenus Inc and EISAI. AW has received research funding from Gilead, Agenus, Genentech and Genocea. She has been a consultant for Aicuris. CJ and AW receive royalties from UpToDate.

REFERENCES

  • [1].Looker KJ, Garnett GP, Schmid GP. An estimate of the global prevalence and incidence of herpes simplex virus type 2 infection. Bull World Health Organ. 2008;86:805–12. doi: 10.2471/BLT.07.046128. A. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [2].Owusu-Edusei KJ, Chesson HW, Gift TL, Tao G, Mahajan R, Ocfemia MCB, et al. The Estimated Direct Medical Cost of Selected Sexually Transmitted Infections in the United States, 2008. Sexually Transmitted Diseases. 2013;40:197–201. doi: 10.1097/OLQ.0b013e318285c6d2. 10.1097/OLQ.0b013e318285c6d2. [DOI] [PubMed] [Google Scholar]
  • [3].Center for Disease Control and Prevention Seroprevalence of herpes simplex virus type 2 among persons aged 14-49 years--United States, 2005-2008. MMWR Morb Mortal Wkly Rep. 2010;59:456–9. [PubMed] [Google Scholar]
  • [4].Paz-Bailey G, Ramaswamy M, Hawkes SJ, Geretti AM. Herpes simplex virus type 2: epidemiology and management options in developing countries. Sexually Transmitted Infections. 2007;83:16–22. doi: 10.1136/sti.2006.020966. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [5].Watson-Jones D, Weiss HA, Rusizoka M, Baisley K, Mugeye K, Changalucha J, et al. Risk factors for herpes simplex virus type 2 and HIV among women at high risk in northwestern Tanzania: preparing for an HSV-2 intervention trial. J Acquir Immune Defic Syndr. 2007;46:631–42. doi: 10.1097/QAI.0b013e31815b2d9c. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [6].Xu F, Sternberg MR, Kottiri BJ, McQuillan GM, Lee FK, Nahmias AJ, et al. Trends in herpes simplex virus type 1 and type 2 seroprevalence in the United States. JAMA. 2006;296:964–73. doi: 10.1001/jama.296.8.964. [DOI] [PubMed] [Google Scholar]
  • [7].Mertz KJ, Trees D, Levine WC, Lewis JS, Litchfield B, Pettus KS, et al. The Genital Ulcer Disease Surveillance Group Etiology of genital ulcers and prevalence of human immunodeficiency virus coinfection in 10 US cities. J Infect Dis. 1998;178:1795–8. doi: 10.1086/314502. [DOI] [PubMed] [Google Scholar]
  • [8].Paz-Bailey G, Rahman M, Chen C, Ballard R, Moffat HJ, Kenyon T, et al. Changes in the etiology of sexually transmitted diseases in Botswana between 1993 and 2002: implications for the clinical management of genital ulcer disease. Clin Infect Dis. 2005;41:1304–12. doi: 10.1086/496979. [DOI] [PubMed] [Google Scholar]
  • [9].Xu F, Schillinger JA, Sternberg MR, Johnson RE, Lee FK, Nahmias AJ, et al. Seroprevalence and coinfection with herpes simplex virus type 1 and type 2 in the United States, 1988-1994. J Infect Dis. 2002;185:1019–24. doi: 10.1086/340041. [DOI] [PubMed] [Google Scholar]
  • [10].Mertz GJ, Benedetti J, Ashley R, Selke SA, Corey L. Risk factors for the sexual transmission of genital herpes. Ann Intern Med. 1992;116:197–202. doi: 10.7326/0003-4819-116-3-197. [DOI] [PubMed] [Google Scholar]
  • [11].Langenberg AG, Corey L, Ashley RL, Leong WP, Straus SE, Chiron HSV Vaccine Study Group A prospective study of new infections with herpes simplex virus type 1 and type 2. N Engl J Med. 1999;341:1432–8. doi: 10.1056/NEJM199911043411904. [DOI] [PubMed] [Google Scholar]
  • [12].Kimberlin DW, Lin C-Y, Jacobs RF, Powell DA, Frenkel LM, Gruber WC, et al. Natural History of Neonatal Herpes Simplex Virus Infections in the Acyclovir Era. Pediatrics. 2001;108:223–9. doi: 10.1542/peds.108.2.223. [DOI] [PubMed] [Google Scholar]
  • [13].Brown ZA, Selke S, Zeh J, Kopelman J, Maslow A, Ashley RL, et al. The acquisition of herpes simplex virus during pregnancy. N Engl J Med. 1997;337:509–15. doi: 10.1056/NEJM199708213370801. [DOI] [PubMed] [Google Scholar]
  • [14].Dinh T-H, Dunne EF, Markowitz LE, Weinstock H, Berman S. Assessing Neonatal Herpes Reporting in the United States, 2000-2005. Sexually Transmitted Diseases. 2008;35:19–21. doi: 10.1097/OLQ.0b013e318162c4c6. 10.1097/OLQ.0b013e318162c4c6. [DOI] [PubMed] [Google Scholar]
  • [15].Brown ZA, Wald A, Morrow RA, Selke S, Zeh J, Corey L. Effect of serologic status and cesarean delivery on transmission rates of herpes simplex virus from mother to infant. JAMA. 2003;289:203–9. doi: 10.1001/jama.289.2.203. [DOI] [PubMed] [Google Scholar]
  • [16].Freeman EE, Weiss HA, Glynn JR, Cross PL, Whitworth JA, Hayes RJ. Herpes simplex virus 2 infection increases HIV acquisition in men and women: systematic review and metaanalysis of longitudinal studies. AIDS. 2006;20:73–83. doi: 10.1097/01.aids.0000198081.09337.a7. [DOI] [PubMed] [Google Scholar]
  • [17].Reynolds SJ, Risbud AR, Shepherd ME, Zenilman JM, Brookmeyer RS, Paranjape RS, et al. Recent herpes simplex virus type 2 infection and the risk of human immunodeficiency virus type 1 acquisition in India.[see comment] Journal of Infectious Diseases. 2003;187:1513–21. doi: 10.1086/368357. [DOI] [PubMed] [Google Scholar]
  • [18].Tobian A, Charvat B, Ssempijja V, Kigozi G, Serwadda D, Makumbi F, et al. Factors Associated with the Prevalence and Incidence of Herpes Simplex Virus Type 2 Infection among Men in Rakai, Uganda. J Infect Dis. 2009;199:945–9. doi: 10.1086/597074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [19].Schacker T, Ryncarz AJ, Goddard J, Diem K, Shaughnessy M, Corey L. Frequent recovery of HIV-1 from genital herpes simplex virus lesions in HIV-1-infected men. Jama. 1998;280:61–6. doi: 10.1001/jama.280.1.61. [DOI] [PubMed] [Google Scholar]
  • [20].Gray RH, Wawer MJ, Brookmeyer R, Sewankambo NK, Serwadda D, Wabwire-Mangen F, et al. Probability of HIV-1 transmission per coital act in monogamous, heterosexual, HIV-1-discordant couples in Rakai, Uganda. The Lancet. 2001;357:1149–53. doi: 10.1016/S0140-6736(00)04331-2. [DOI] [PubMed] [Google Scholar]
  • [21].Wald A, Link K. Risk of human immunodeficiency virus infection in herpes simplex virus type 2-seropositive persons: a meta-analysis. J Infect Dis. 2002;185:45–52. doi: 10.1086/338231. [DOI] [PubMed] [Google Scholar]
  • [22].Freeman EE, White RG, Bakker R, Orroth KK, Weiss HA, Buvé A, et al. Population-level effect of potential HSV2 prophylactic vaccines on HIV incidence in sub-Saharan Africa. Vaccine. 2009;27:940–6. doi: 10.1016/j.vaccine.2008.11.074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [23].Zhu J, Hladik F, Woodward A, Klock A, Peng T, Johnston C, et al. Persistence of HIV-1 receptor-positive cells after HSV-2 reactivation is a potential mechanism for increased HIV-1 acquisition. Nature Medicine. 2009;15:886–U83. doi: 10.1038/nm.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [24].Johnson K, Sherman M, Ssempiija V, Tobian A, Zenilman J, Duggan M, et al. Foreskin inflammation is associated with HIV and herpes simplex virus type-2 infections in Rakai, Uganda. AIDS. 2009;23:1807–15. doi: 10.1097/QAD.0b013e32832efdf1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [25].Rebbapragada A, Wachihi C, Pettengell C, Sunderji S, Huibner S, Jaoko W, et al. Negative mucosal synergy between Herpes simplex type 2 and HIV in the female genital tract. AIDS. 2007;21:589–98. doi: 10.1097/QAD.0b013e328012b896. [DOI] [PubMed] [Google Scholar]
  • [26].Corey L, Wald A, Patel R, Sacks SL, Tyring SK, Warren T, et al. Once-daily valacyclovir to reduce the risk of transmission of genital herpes. N Engl J Med. 2004;350:11–20. doi: 10.1056/NEJMoa035144. [DOI] [PubMed] [Google Scholar]
  • [27].Wald A, Krantz E, Selke S, Lairson E, Morrow RA, Zeh J. Knowledge of partners' genital herpes protects against herpes simplex virus type 2 acquisition. J Infect Dis. 2006;194:42–52. doi: 10.1086/504717. [DOI] [PubMed] [Google Scholar]
  • [28].Martin ET, Krantz E, Gottlieb SL, Magaret AS, Langenberg A, Stanberry L, et al. A pooled analysis of the effect of condoms in preventing HSV-2 acquisition. Arch Intern Med. 2009;169:1233–40. doi: 10.1001/archinternmed.2009.177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [29].Tobian AAR, Serwadda D, Quinn TC, Kigozi G, Gravitt PE, Laeyendecker O, et al. Male Circumcision for the Prevention of HSV-2 and HPV Infections and Syphilis. N Engl J Med. 2009;360:1298–309. doi: 10.1056/NEJMoa0802556. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [30].Tobian AAR, Kigozi G, Redd AD, Serwadda D, Kong X, Oliver A, et al. Male Circumcision and Herpes Simplex Virus Type 2 Infection in Female Partners: A Randomized Trial in Rakai, Uganda. Journal of Infectious Diseases. 2012;205:486–90. doi: 10.1093/infdis/jir767. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [31].Mujugira A, Magaret AS, Celum C, Baeten JM, Lingappa JR, Morrow RA, et al. Daily Acyclovir To Decrease HSV-2 Transmission From HSV-2/HIV-1 Co-Infected Persons: A Randomized Controlled Trial. J Infect Dis. doi: 10.1093/infdis/jit333. submitted. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [32].Ryder N, Jin F, McNulty AM, Grulich AE, Donovan B. Increasing role of herpes simplex virus type 1 in first-episode anogenital herpes in heterosexual women and younger men who have sex with men, 1992-2006. Sexually Transmitted Infections. 2009;85:416–9. doi: 10.1136/sti.2008.033902. [DOI] [PubMed] [Google Scholar]
  • [33].Looker KJ, Garnett GP. A systematic review of the epidemiology and interaction of herpes simplex virus types 1 and 2. Sex Transm Infect. 2005;81:103–7. doi: 10.1136/sti.2004.012039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [34].Cunningham AL, Diefenbach RJ, Miranda-Saksena M, Bosnjak L, Kim M, Jones C, et al. The cycle of human herpes simplex virus infection: virus transport and immune control. J Infect Dis. 2006;194(Suppl 1):S11–8. doi: 10.1086/505359. [DOI] [PubMed] [Google Scholar]
  • [35].Oxman MN. Zoster vaccine: current status and future prospects. Clin Infect Dis. 2010;51:197–213. doi: 10.1086/653605. [DOI] [PubMed] [Google Scholar]
  • [36].Mark KE, Wald A, Magaret AS, Selke S, Kuntz S, Huang ML, et al. Rapidly cleared episodes of oral and anogenital herpes simplex virus shedding in HIV-infected adults. J Acquir Immune Defic Syndr. 2010;54:482–8. doi: 10.1097/QAI.0b013e3181d91322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [37].Mark KE, Wald A, Magaret AS, Selke S, Olin L, Huang ML, et al. Rapidly cleared episodes of herpes simplex virus reactivation in immunocompetent adults. J Infect Dis. 2008;198:1141–9. doi: 10.1086/591913. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [38].Schiffer JT, Abu-Raddad L, Mark KE, Zhu J, Selke S, Koelle DM, et al. Mucosal host immune response predicts the severity and duration of herpes simplex virus-2 genital tract shedding episodes. Proc Natl Acad Sci U S A. 2010;107:18973–8. doi: 10.1073/pnas.1006614107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [39].Shin H, Iwasaki A. A vaccine strategy that protects against genital herpes by establishing local memory T cells. Nature. 2012;491:463–7. doi: 10.1038/nature11522. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [40].Stanberry LR, Kern ER, Richards JT, Abbott TM, Overall JC., Jr. Genital herpes in guinea pigs: pathogenesis of the primary infection and description of recurrent disease. J Infect Dis. 1982;146:397–404. doi: 10.1093/infdis/146.3.397. [DOI] [PubMed] [Google Scholar]
  • [41].Yim KC, Carroll CJ, Tuyama A, Cheshenko N, Carlucci MJ, Porter DD, et al. The cotton rat provides a novel model to study genital herpes infection and to evaluate preventive strategies. J Virol. 2005;79:14632–9. doi: 10.1128/JVI.79.23.14632-14639.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [42].Crostarosa F, Aravantinou M, Akpogheneta OJ, Jasny E, Shaw A, Kenney J, et al. A Macaque Model to Study Vaginal HSV-2/Immunodeficiency Virus Co-Infection and the Impact of HSV-2 on Microbicide Efficacy. PLoS One. 2009;4:e8060. doi: 10.1371/journal.pone.0008060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [43].Herman M, Ciancanelli M, Ou Y-H, Lorenzo L, Klaudel-Dreszler M, Pauwels E, et al. Heterozygous TBK1 mutations impair TLR3 immunity and underlie herpes simplex encephalitis of childhood. The Journal of Experimental Medicine. 2012;209:1567–82. doi: 10.1084/jem.20111316. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [44].Davey GM, Wojtasiak M, Proietto AI, Carbone FR, Heath WR, Bedoui S. Cutting Edge: Priming of CD8 T Cell Immunity to Herpes Simplex Virus Type 1 Requires Cognate TLR3 Expression In Vivo. The Journal of Immunology. 2010;184:2243–6. doi: 10.4049/jimmunol.0903013. [DOI] [PubMed] [Google Scholar]
  • [45].Wizel B, Persson J, Thorn K, Nagy E, Harandi AM. Nasal and skin delivery of IC31®-adjuvanted recombinant HSV-2 gD protein confers protection against genital herpes. Vaccine. 2012;30:4361–8. doi: 10.1016/j.vaccine.2012.02.019. [DOI] [PubMed] [Google Scholar]
  • [46].Svensson A, Tunback P, Nordstrom I, Padyukov L, Eriksson K. Polymorphisms in Toll-like receptor 3 confer natural resistance to human herpes simplex virus type 2 infection. Journal of General Virology. 2012;93:1717–24. doi: 10.1099/vir.0.042572-0. [DOI] [PubMed] [Google Scholar]
  • [47].Bochud PY, Magaret AS, Koelle DM, Aderem A, Wald A. Polymorphisms in TLR2 are associated with increased viral shedding and lesional rate in patients with genital herpes simplex virus Type 2 infection. J Infect Dis. 2007;196:505–9. doi: 10.1086/519693. [DOI] [PubMed] [Google Scholar]
  • [48].Yang C-A, Raftery MJ, Hamann L, Guerreiro M, Grutz G, Haase D, et al. Association of TLR3-hyporesponsiveness and functional TLR3 L412F polymorphism with recurrent herpes labialis. Human Immunology. 2012;73:844–51. doi: 10.1016/j.humimm.2012.04.008. [DOI] [PubMed] [Google Scholar]
  • [49].Moraru M, Cisneros E, Gomez-Lozano N, de Pablo R, Portero F, Canizares M, et al. Host Genetic Factors in Susceptibility to Herpes Simplex Type 1 Virus Infection: Contribution of Polymorphic Genes at the Interface of Innate and Adaptive Immunity. The Journal of Immunology. 2012;188:4412–20. doi: 10.4049/jimmunol.1103434. [DOI] [PubMed] [Google Scholar]
  • [50].Balachandran N, Bacchetti S, Rawls WE. Protection against lethal challenge of BALB/c mice by passive transfer of monoclonal antibodies to five glycoproteins of herpes simplex virus type 2. Infect Immun. 1982;37:1132–7. doi: 10.1128/iai.37.3.1132-1137.1982. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [51].Schiller JT, Castellsague X, Garland SM. A Review of Clinical Trials of Human Papillomavirus Prophylactic Vaccines. Vaccine. 2012;30(Supplement 5):F123–F38. doi: 10.1016/j.vaccine.2012.04.108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [52].Kopp SJ, Karaba AH, Cohen LK, Banisadr G, Miller RJ, Muller WJ. Pathogenesis of Neonatal Herpes Simplex 2 Disease in a Mouse Model Is Dependent on Entry Receptor Expression and Route of Inoculation. Journal of Virology. 2013;87:474–81. doi: 10.1128/JVI.01849-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [53].Wang K, Kappel JD, Canders C, Davila WF, Sayre D, Chavez M, et al. A Herpes Simplex Virus 2 Glycoprotein D Mutant Generated by Bacterial Artificial Chromosome Mutagenesis Is Severely Impaired for Infecting Neuronal Cells and Infects Only Vero Cells Expressing Exogenous HVEM. Journal of Virology. 2012;86:12891–902. doi: 10.1128/JVI.01055-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [54].Eisenberg RJ, Atanasiu D, Cairns TM, Gallagher JR, Krummenacher C, Cohen GH. Herpes Virus Fusion and Entry: A Story with Many Characters. Viruses. 2012;4:800–32. doi: 10.3390/v4050800. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [55].Taylor JM, Lin E, Susmarski N, Yoon M, Zago A, Ware CF, et al. Alternative Entry Receptors for Herpes Simplex Virus and Their Roles in Disease. Cell Host & Microbe. 2007;2:19–28. doi: 10.1016/j.chom.2007.06.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [56].Liao H-X, Bonsignori M, Alam SM, McLellan Jason S, Tomaras Georgia D, Moody MA, et al. Vaccine Induction of Antibodies against a Structurally Heterogeneous Site of Immune Pressure within HIV-1 Envelope Protein Variable Regions 1 and 2. Immunity. 2013;38:176–86. doi: 10.1016/j.immuni.2012.11.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [57].Krause JC, Tsibane T, Tumpey TM, Huffman CJ, Albrecht R, Blum DL, et al. Human Monoclonal Antibodies to Pandemic 1957 H2N2 and Pandemic 1968 H3N2 Influenza Viruses. Journal of Virology. 2012;86:6334–40. doi: 10.1128/JVI.07158-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [58].Zhu J, Koelle DM, Cao J, Vazquez J, Huang ML, Hladik F, et al. Virus-specific CD8+ T cells accumulate near sensory nerve endings in genital skin during subclinical HSV-2 reactivation. J Exp Med. 2007;204:595–603. doi: 10.1084/jem.20061792. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [59].Peng T, Zhu J, Phasouk K, Koelle DM, Wald A, Corey L. An effector phenotype of CD8+ T cells at the junction epithelium during clinical quiescence of HSV-2 infection. Journal of Virology. 2012;86:10587–96. doi: 10.1128/JVI.01237-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [60].Mackay LK, Stock AT, Ma JZ, Jones CM, Kent SJ, Mueller SN, et al. Long-lived epithelial immunity by tissue-resident memory T (TRM) cells in the absence of persisting local antigen presentation. Proceedings of the National Academy of Sciences. 2012;109:7037–42. doi: 10.1073/pnas.1202288109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [61].Moss NJ, Magaret A, Laing KJ, Kask AS, Wang M, Mark KE, et al. Peripheral Blood CD4 T-Cell and Plasmacytoid Dendritic Cell (pDC) Reactivity to Herpes Simplex Virus 2 and pDC Number Do Not Correlate with the Clinical or Virologic Severity of Recurrent Genital Herpes. Journal of Virology. 2012;86:9952–63. doi: 10.1128/JVI.00829-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [62].Jing L, Haas J, Chong TM, Bruckner JJ, Dann GC, Dong L, et al. Cross-presentation and genome-wide screening reveal candidate T cells antigens for a herpes simplex virus type 1 vaccine. The Journal of Clinical Investigation. 2012;122:654–73. doi: 10.1172/JCI60556. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [63].Kalantari-Dehaghi M, Chun S, Chentoufi A, Pablo J, Liang L, Dasgupta G, et al. Discovery of potential diagnostic and vaccine antigens in herpes simplex virus-1 and -2 by proteome-wide antibody profiling. Journal of Virology. 2012;86:4328–39. doi: 10.1128/JVI.05194-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [64].Dasgupta G, Chentoufi AA, Kalantari M, Falatoonzadeh P, Chun S, Lim CH, et al. Immunodominant "Asymptomatic" Herpes Simplex Virus 1 and 2 Protein Antigens Identified by Probing Whole-ORFome Microarrays with Serum Antibodies from Seropositive Asymptomatic versus Symptomatic Individuals. Journal of Virology. 2012;86:4358–69. doi: 10.1128/JVI.07107-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [65].Chentoufi AA, Binder NR, Berka N, Durand G, Nguyen A, Bettahi I, et al. Asymptomatic human CD4+ cytotoxic T-cell epitopes identified from herpes simplex virus glycoprotein B. J Virol. 2008;82:11792–802. doi: 10.1128/JVI.00692-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [66].Skoberne M, Cardin R, Lee A, Kazimirova A, Zielinski V, Garvie D, et al. An Adjuvanted Herpes Simplex Virus Type 2 (HSV-2) Subunit Vaccine Elicits a T Cell Response In Mice and Is an Effective Therapeutic Vaccine In Guinea Pigs. Journal of Virology. 2013;87:3930–42. doi: 10.1128/JVI.02745-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [67].Laing K, Magaret A, Mueller D, Zhao L, Johnston C, De Rosa S, et al. Diversity in CD8(+) T Cell Function and Epitope Breadth Among Persons with Genital Herpes. Journal of Clinical Immunology. 2010;30:703–22. doi: 10.1007/s10875-010-9441-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [68].St. Leger AJ, Peters B, Sidney J, Sette A, Hendricks RL. Defining the Herpes Simplex Virus-Specific CD8+ T Cell Repertoire in C57BL/6 Mice. The Journal of Immunology. 2011;186:3927–33. doi: 10.4049/jimmunol.1003735. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [69].Koelle DM, Liu Z, McClurkan CL, Cevallos RC, Vieira J, Hosken NA, et al. Immunodominance among herpes simplex virus-specific CD8 T cells expressing a tissue-specific homing receptor. Proc Natl Acad Sci U S A. 2003;100:12899–904. doi: 10.1073/pnas.2131705100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [70].Jing L, Schiffer JT, Chong TM, Bruckner JJ, Davies DH, Felgner PL, et al. CD4 T-Cell Memory Responses to Viral Infections of Humans Show Pronounced Immunodominance Independent of Duration or Viral Persistence. Journal of Virology. 2013;87:2617–27. doi: 10.1128/JVI.03047-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [71].Koelle DM, Posavad CM, Barnum GR, Johnson ML, Frank JM, Corey L. Clearance of HSV-2 from recurrent genital lesions correlates with infiltration of HSV-specific cytotoxic T lymphocytes. J Clin Invest. 1998;101:1500–8. doi: 10.1172/JCI1758. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [72].Koelle DM, Liu Z, McClurkan CM, Topp MS, Riddell SR, Pamer EG, et al. Expression of cutaneous lymphocyte-associated antigen by CD8(+) T cells specific for a skin-tropic virus. J Clin Invest. 2002;110:537–48. doi: 10.1172/JCI15537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [73].Verjans GMGM, Hintzen RQ, van Dun JM, Poot A, Milikan JC, Laman JD, et al. Selective retention of herpes simplex virus-specific T cells in latently infected human trigeminal ganglia. Proc Natl Acad Sci U S A. 2007;104:3496–501. doi: 10.1073/pnas.0610847104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [74].Verjans GM, Dings ME, McLauchlan J, van Der Kooi A, Hoogerhout P, Brugghe HF, et al. Intraocular T cells of patients with herpes simplex virus (HSV)-induced acute retinal necrosis recognize HSV tegument proteins VP11/12 and VP13/14. J Infect Dis. 2000;182:923–7. doi: 10.1086/315759. [DOI] [PubMed] [Google Scholar]
  • [75].Koelle DM, Schomogyi M, Corey L. Antigen-specific T cells localize to the uterine cervix in women with genital herpes simplex virus type 2 infection. J Infect Dis. 2000;182:662–70. doi: 10.1086/315749. [DOI] [PubMed] [Google Scholar]
  • [76].Schiffer JT, Abu-Raddad L, Mark KE, Zhu J, Selke S, Magaret A, et al. Frequent release of low amounts of herpes simplex virus from neurons: results of a mathematical model. Sci Transl Med. 2009;1:7–16. doi: 10.1126/scitranslmed.3000193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [77].Johnston C, Koelle DM, Wald A. HSV-2: in pursuit of a vaccine. The Journal of Clinical Investigation. 2011;121:4600–9. doi: 10.1172/JCI57148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [78].Corey L, Langenberg AG, Ashley R, Sekulovich RE, Izu AE, Douglas JM, Jr., et al. Chiron HSV Vaccine Study Group Recombinant glycoprotein vaccine for the prevention of genital HSV-2 infection: two randomized controlled trials. JAMA. 1999;282:331–40. doi: 10.1001/jama.282.4.331. [DOI] [PubMed] [Google Scholar]
  • [79].Langenberg AG, Burke RL, Adair SF, Sekulovich R, Tigges M, Dekker CL, et al. A recombinant glycoprotein vaccine for herpes simplex virus type 2: safety and immunogenicity [corrected] Ann Intern Med. 1995;122:889–98. doi: 10.7326/0003-4819-122-12-199506150-00001. [DOI] [PubMed] [Google Scholar]
  • [80].Bernstein DI, Aoki FY, Tyring SK, Stanberry LR, St-Pierre C, Shafran SD, et al. Safety and immunogenicity of glycoprotein D-adjuvant genital herpes vaccine. Clin Infect Dis. 2005;40:1271–81. doi: 10.1086/429240. [DOI] [PubMed] [Google Scholar]
  • [81].Stanberry LR, Spruance SL, Cunningham AL, Bernstein DI, Mindel A, Sacks S, et al. Glycoprotein-D-adjuvant vaccine to prevent genital herpes. N Engl J Med. 2002;347:1652–61. doi: 10.1056/NEJMoa011915. [DOI] [PubMed] [Google Scholar]
  • [82].Belshe RB, Leone PA, Bernstein DI, Wald A, Levin MJ, Stapleton JT, et al. Efficacy Results of a Trial of a Herpes Simplex Vaccine. New England Journal of Medicine. 2012;366:34–43. doi: 10.1056/NEJMoa1103151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [83].Balachandran N, Oba DE, Hutt-Fletcher LM. Antigenic cross-reactions among herpes simplex virus types 1 and 2, Epstein-Barr virus, and cytomegalovirus. J Virol. 1987;61:1125–35. doi: 10.1128/jvi.61.4.1125-1135.1987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [84].Straus SE, Corey L, Burke RL, Savarese B, Barnum G, Krause PR, et al. Placebo-controlled trial of vaccination with recombinant glycoprotein D of herpes simplex virus type 2 for immunotherapy of genital herpes. Lancet. 1994;343:1460–3. doi: 10.1016/s0140-6736(94)92581-x. [DOI] [PubMed] [Google Scholar]
  • [85].Straus SE, Wald A, Kost RG, McKenzie R, Langenberg AG, Hohman P, et al. Immunotherapy of recurrent genital herpes with recombinant herpes simplex virus type 2 glycoproteins D and B: results of a placebo-controlled vaccine trial. J Infect Dis. 1997;176:1129–34. doi: 10.1086/514103. [DOI] [PubMed] [Google Scholar]
  • [86].Casanova G, Cancela R, Alonzo L, Benuto R, Magana Mdel C, Hurley DR, et al. A double-blind study of the efficacy and safety of the ICP10deltaPK vaccine against recurrent genital HSV-2 infections. Cutis. 2002;70:235–9. [PubMed] [Google Scholar]
  • [87].de Bruyn G, Vargas-Cortez M, Warren T, Tyring SK, Fife KH, Lalezari J, et al. A randomized controlled trial of a replication defective (gH deletion) herpes simplex virus vaccine for the treatment of recurrent genital herpes among immunocompetent subjects. Vaccine. 2006;24:914–20. doi: 10.1016/j.vaccine.2005.08.088. [DOI] [PubMed] [Google Scholar]
  • [88].Okuku HS, Sanders EJ, Nyiro J, Ngetsa C, Ohuma E, McClelland RS, et al. Factors Associated With Herpes Simplex Virus Type 2 Incidence in a Cohort of Human Immunodeficiency Virus Type 1-Seronegative Kenyan Men and Women Reporting High-Risk Sexual Behavior. Sexually Transmitted Diseases. 2011;38:837–44. doi: 10.1097/OLQ.0b013e31821a6225. 10.1097/OLQ.0b013e31821a6225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [89].Dudek TE, Torres-Lopez E, Crumpacker C, Knipe DM. Evidence for differences in immunologic and pathogenesis properties of herpes simplex virus 2 strains from the United States and South Africa. J Infect Dis. 2011;203:1434–41. doi: 10.1093/infdis/jir047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [90].Garnett GP, Dubin G, Slaoui M, Darcis T. The potential epidemiological impact of a genital herpes vaccine for women. Sex Transm Infect. 2004;80:24–9. doi: 10.1136/sti.2002.003848. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [91].Schwartz EJ, Blower S. Predicting the potential individual- and population-level effects of imperfect herpes simplex virus type 2 vaccines. J Infect Dis. 2005;191:1734–46. doi: 10.1086/429299. [DOI] [PubMed] [Google Scholar]
  • [92].Alsallaq RA, Schiffer JT, Longini IM, Jr., Wald A, Corey L, Abu-Raddad LJ. Population Level Impact of an Imperfect Prophylactic Vaccine for Herpes Simplex Virus-2. Sexually Transmitted Diseases. 2010;37:290–7. doi: 10.1097/OLQ.0b013e3181d3d023. 10.1097/OLQ.0b013e3181d3d023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [93].Johnston C, Saracino M, Kuntz S, Magaret A, Selke S, Huang M-l, et al. Standard-dose and high-dose daily antiviral therapy for short episodes of genital HSV-2 reactivation: three randomised, open-label, cross-over trials. The Lancet. 2012;379:641–7. doi: 10.1016/S0140-6736(11)61750-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [94].Tronstein E, Johnston C, Huang ML, Selke S, Magaret A, Warren T, et al. Genital shedding of herpes simplex virus among symptomatic and asymptomatic persons with HSV-2 infection. JAMA. 2011;305:1441–9. doi: 10.1001/jama.2011.420. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [95].Halford WP, Puschel R, Gershburg E, Wilber A, Gershburg S, Rakowski B. A Live-Attenuated HSV-2 ICP0(−) Virus Elicits 10 to 100 Times Greater Protection against Genital Herpes than a Glycoprotein D Subunit Vaccine. PLoS One. 2011;6:e17748. doi: 10.1371/journal.pone.0017748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [96].Luo C, Goshima F, Kamakura M, Mutoh Y, Iwata S, Kimura H, et al. Immunization with a highly attenuated replication-competent herpes simplex virus type 1 mutant, HF10, protects mice from genital disease caused by herpes simplex virus type 2. Frontiers in Microbiology. 2012:3. doi: 10.3389/fmicb.2012.00158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [97].Wang F, Zumbrun EE, Huang J, Si H, Makaroun L, Friedman HM. Herpes simplex virus type 2 glycoprotein E is required for efficient virus spread from epithelial cells to neurons and for targeting viral proteins from the neuron cell body into axons. Virology. 2010;405:269–79. doi: 10.1016/j.virol.2010.06.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [98].Awasthi S, Zumbrun EE, Si H, Wang F, Shaw CE, Cai M, et al. Live attenuated herpes simplex virus 2 glycoprotein E deletion mutant as a vaccine candidate defective in neuronal spread. J Virol. 2012;86:4586–98. doi: 10.1128/JVI.07203-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [99].Prichard MN, Kaiwar R, Jackman WT, Quenelle DC, Collins DJ, Kern ER, et al. Evaluation of AD472, a live attenuated recombinant herpes simplex virus type 2 vaccine in guinea pigs. Vaccine. 2005;23:5424–31. doi: 10.1016/j.vaccine.2005.02.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [100].Da Costa X, Kramer MF, Zhu J, Brockman MA, Knipe DM. Construction, phenotypic analysis, and immunogenicity of a UL5/UL29 double deletion mutant of herpes simplex virus 2. J Virol. 2000;74:7963–71. doi: 10.1128/jvi.74.17.7963-7971.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [101].Hoshino Y, Dalai SK, Wang K, Pesnicak L, Lau TY, Knipe DM, et al. Comparative Efficacy and Immunogenicity of Replication-Defective, Recombinant Glycoprotein, and DNA Vaccines for Herpes Simplex Virus 2 Infections in Mice and Guinea Pigs. J Virol. 2005;79:410–8. doi: 10.1128/JVI.79.1.410-418.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [102].Hoshino Y, Pesnicak L, Dowdell KC, Burbelo PD, Knipe DM, Straus SE, et al. Protection from Herpes Simplex Virus (HSV)-2 Infection with Replication-Defective HSV-2 or Glycoprotein D2 Vaccines in HSV-1-Seropositive and HSV-1-Seronegative Guinea Pigs. Journal of Infectious Diseases. 2009;200:1088–95. doi: 10.1086/605645. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [103].Mundle ST, Hernandez H, Hamberger J, Catalan J, Zhou C, Stegalkina S, et al. High-Purity Preparation of HSV-2 Vaccine Candidate ACAM529 Is Immunogenic and Efficacious In Vivo. PLoS One. 2013;8:e57224. doi: 10.1371/journal.pone.0057224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [104].Akhrameyeva NV, Zhang P, Sugiyama N, Behar SM, Yao F. Development of a Glycoprotein D-Expressing Dominant-Negative and Replication-Defective Herpes Simplex Virus 2 (HSV-2) Recombinant Viral Vaccine against HSV-2 Infection in Mice. Journal of Virology. 2011;85:5036–47. doi: 10.1128/JVI.02548-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [105].Morello CS, Levinson MS, Kraynyak KA, Spector DH. Immunization with herpes simplex virus 2 (HSV-2) genes plus inactivated HSV-2 is highly protective against acute and recurrent HSV-2 disease. J Virol. 2011;85:3461–72. doi: 10.1128/JVI.02521-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [106].Morello CS, Kraynyak KA, Levinson MS, Chen Z, Lee K-F, Spector DH. Inactivated HSV-2 in MPL/alum adjuvant provides nearly complete protection against genital infection and shedding following long term challenge and rechallenge. Vaccine. 2012;30:6541–50. doi: 10.1016/j.vaccine.2012.08.049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [107].Chiuppesi F, Vannucci L, De Luca A, Lai M, Matteoli B, Freer G, et al. A Lentiviral Vector-Based, Herpes Simplex Virus 1 (HSV-1) Glycoprotein B Vaccine Affords Cross-Protection against HSV-1 and HSV-2 Genital Infections. Journal of Virology. 2012;86:6563–74. doi: 10.1128/JVI.00302-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [108].Veselenak RL, Shlapobersky M, Pyles RB, Wei Q, Sullivan SM, Bourne N. A Vaxfectin®-adjuvanted HSV-2 plasmid DNA vaccine is effective for prophylactic and therapeutic use in the guinea pig model of genital herpes. Vaccine. 2012;30:7046–51. doi: 10.1016/j.vaccine.2012.09.057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [109].Cortesi R, Ravani L, Rinaldi F, Marconi P, Drechsler M, Manservigi M, et al. Intranasal immunization in mice with non-ionic surfactants vesicles containing HSV immunogens: A preliminary study as possible vaccine against genital herpes. International Journal of Pharmaceutics. 2013;440:229–37. doi: 10.1016/j.ijpharm.2012.06.042. [DOI] [PubMed] [Google Scholar]
  • [110].Tirabassi RS, Ace CI, Levchenko T, Torchilin VP, Selin LK, Nie S, et al. A mucosal vaccination approach for herpes simplex virus type 2. Vaccine. 2011;29:1090–8. doi: 10.1016/j.vaccine.2010.11.076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [111].Kask AS, Chen X, Marshak JO, Dong L, Saracino M, Chen D, et al. DNA vaccine delivery by densely-packed and short microprojection arrays to skin protects against vaginal HSV-2 challenge. Vaccine. 2010;28:7483–91. doi: 10.1016/j.vaccine.2010.09.014. [DOI] [PubMed] [Google Scholar]
  • [112].Wald A, Koelle DM, Fife K, Warren T, LeClair K, Chicz RM, et al. Safety and immunogenicity of long HSV-2 peptides complexed with rhHsc70 in HSV-2 seropositive persons. Vaccine. 2011;29:8520–9. doi: 10.1016/j.vaccine.2011.09.046. [DOI] [PubMed] [Google Scholar]

RESOURCES