Next Article in Journal
Virus-like Particles Produced in the Baculovirus System Protect Hares from European Brown Hare Syndrome Virus (EBHSV) Infection
Next Article in Special Issue
Antibody Responses Following Primary Immunization with the Recombinant Herpes Zoster Vaccine (Shingrix®) in VZV Seronegative Immunocompromised Adults
Previous Article in Journal
An In Vitro Approach to Prime or Boost Human Antigen-Specific CD8+ T Cell Responses: Applications to Vaccine Studies
Previous Article in Special Issue
Human Alpha Herpesviruses Infections (HSV1, HSV2, and VZV), Alzheimer’s Disease, and the Potential Benefits of Targeted Treatment or Vaccination—A Virological Perspective
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Research Progress on Varicella-Zoster Virus Vaccines

1
State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, China
2
National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen 361102, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Vaccines 2025, 13(7), 730; https://doi.org/10.3390/vaccines13070730
Submission received: 30 April 2025 / Revised: 28 June 2025 / Accepted: 1 July 2025 / Published: 4 July 2025
(This article belongs to the Special Issue Varicella and Zoster Vaccination)

Abstract

Varicella-zoster virus (VZV) poses significant public health challenges as the etiological agent of varicella (chickenpox) and herpes zoster (HZ), given its high transmissibility and potential for severe complications. The introduction of VZV vaccines—particularly the vOka-based live attenuated and glycoprotein gE-based recombinant subunit vaccines—has substantially reduced the global incidence of these diseases. However, live attenuated vaccines raise concerns regarding safety and immunogenicity, especially in immunocompromised populations, while recombinant subunit vaccines, such as Shingrix, exhibit high efficacy but are associated with side effects and adjuvant limitations. Recent advancements in vaccine technology, including mRNA vaccines, viral vector vaccines, and virus-like particle (VLP) vaccines, offer promising alternatives with improved safety profiles and durable immunity. This review synthesizes current knowledge on VZV vaccine mechanisms, clinical applications, and immunization strategies, while also examining future directions in vaccine development. The findings underscore the pivotal role of VZV vaccines in disease prevention and highlight the need for continued research to enhance their public health impact.

1. Introduction

Varicella-zoster virus (VZV), also known as human herpesvirus 3 (HHV-3), belongs to the Herpesviridae family and the Alphaherpesvirinae subfamily. This nearly spherical, enveloped virus measures 150–200 nm in diameter and contains double-stranded DNA. Its structural components include nucleocapsid proteins and a lipoprotein envelope (Figure 1). The VZV genome consists of 124,884 base pairs (bp) and encodes at least 70 genes, organized into two covalently linked segments: a long (L) segment and a short (S) segment [1]. The nucleocapsid forms an icosahedral structure with 162 capsomeres, while the viral envelope is enriched with glycoproteins, including gB (ORF31), gC (ORF14), gE (ORF68), gH (ORF37), gI (ORF67), gK (ORF5), gL (ORF60), gM (ORF50), and gN (ORF50) [2]. These glycoproteins play vital roles in the viral life cycle and interactions with the immune system.
VZV is a highly contagious pathogen that spreads through aerosol particles or direct contact with vesicular lesions of infected individuals. Primary VZV infection causes varicella (chickenpox), which predominantly occurs in children and manifests as fever, fatigue, and an intensely pruritic vesicular rash, typically following seasonal patterns [3]. After primary infection, VZV becomes latent in dorsal root ganglia and can reactivate later in life due to immune decline or immunosenescence, leading to herpes zoster (HZ, shingles). Although HZ is less contagious than varicella, exposure to VZV from the vesicular lesions of HZ patients can still induce varicella in susceptible individuals [4].
While varicella and HZ are typically self-limiting, they may lead to severe or fatal complications. Varicella can induce secondary bacterial infections, encephalitis, pneumonia, hepatitis, hemorrhage, and other life-threatening conditions. Similarly, HZ is frequently complicated by postherpetic neuralgia (PHN), as well as meningitis, meningoencephalitis, meningoradiculitis, cerebellitis, myelopathy, vasculopathy, and ophthalmic complications [5], all of which seriously compromise of patients’ quality of life [6,7,8,9]. Among immunocompromised individuals, VZV infection may result in more severe complications while enhancing viral transmissibility, thereby exacerbating disease incidence and mortality rates [10,11]. Varicella, HZ and their complications cause significant productivity losses and impose a substantial burden on healthcare systems.
Currently, vaccination remains the primary strategy for preventing varicella and HZ. Before widespread varicella vaccination (1996–2011), approximately 4 million cases of varicella occurred annually in the United States, resulting in an estimated 10,000 hospitalizations and 100 deaths each year [12]. Studies indicate that implementation of a two-dose varicella vaccination program achieved a 95% reduction in varicella incidence in the United States and an 84% decrease in varicella-related hospitalizations among Australian children aged 18–59 months [13,14]. However, unvaccinated adults or those without prior varicella exposure remain susceptible to infection. Global epidemiological data indicate an annual HZ incidence of 3–5 cases per 1000 individuals [15]. The live zoster vaccine (Zostavax) and recombinant adjuvanted vaccine (Shingrix) were approved by the Food and Drug Administration (FDA). However, with global aging populations, HZ incidence is projected to increase, imposing growing economic burdens on healthcare systems. Additionally, immunocompromised populations, including cancer patients, organ transplant recipients, and human immunodeficiency virus (HIV)-positive individuals, face particular challenges with varicella and HZ vaccines due to impaired immune responses against VZV.
This review systematically outlines recent advancements in the development of VZV vaccines for both varicella and HZ. We conducted a comprehensive literature search using PubMed, supplemented by manual searches from Google Scholar. Vaccine-related data were sourced from official pharmaceutical company websites, vaccine package inserts, published literature, the Center for Drug Evaluation (CDE) of China’s National Medical Products Administration (NMPA) (https://www.cde.org.cn/), and ClinicalTrials.gov (https://clinicaltrials.gov/). Through a comprehensive analysis of both established and emerging vaccine strategies, this study aims to provide a robust scientific foundation and strategic guidance for future research in VZV vaccines.

2. Varicella Vaccines

Following VZV infection, the humoral immune system generates virus-specific antibodies capable of neutralizing the virus and blocking its entry into host cells, thereby limiting viral dissemination [16]. These antibodies exhibit long-term persistence, conferring durable immunity and substantially lowering the risk of reinfection. Additionally, VZV infection triggers a robust cellular immune response, mediated by CD4+ and CD8+ T-cells that recognize and eliminate infected cells, which plays a critical role in preventing viral reactivation [17]. The varicella vaccine effectively stimulates both humoral and cellular immunity against VZV, enabling control of primary infection while potentially reducing the incidence of HZ.
Varicella vaccines are formulated using live attenuated varicella virus strains and are available in two primary forms: a monovalent varicella vaccine and a measles–mumps–rubella–varicella (MMRV) combination vaccine (Table 1). MMRV confers protection against varicella along with three other common pediatric infectious diseases. Both vaccine formulations have demonstrated efficacy and safety in extensive global epidemiological studies and clinical trials.

2.1. Monovalent Varicella Vaccines

Currently, commercially available varicella vaccines are predominantly based on the vOka strain of live attenuated varicella virus. The vOka strain was derived from the parental Oka strain (pOka) of VZV, which was isolated in 1974 by Takahashi et al. from a child with varicella and subsequently attenuated through serial passaging in human embryonic lung (HEL) cells, guinea pig embryo (GPE) cells, and human diploid (WI-38) cells [31]. Experimental evidence shows that vOka maintains stable immunogenicity through at least 15 passages in WI-38 cells, supporting its suitability for large-scale vaccine production [32]. While the vOka vaccine contains a heterogeneous population of attenuated live viruses with minor inter-manufacturer and inter-batch variations, these differences are clinically insignificant [33]. Notably, the vOka strain demonstrates remarkable genetic stability, a critical attribute for vaccine safety. Compared to wild-type VZV, vOka induces a milder immune response while maintaining robust protective immunity and significantly lowering the incidence of HZ [34]. Large-scale clinical trials conducted in diverse age groups and populations with varying health statuses have confirmed the safety, efficacy, and long-term protective effects of vOka-based varicella vaccines [28,35,36,37].
Japan pioneered the use of the vOka varicella vaccine, granting approval to OkaVax (Biken) in 1984, marking the first national licensure of this vaccine. Subsequently, numerous countries have adopted varicella vaccination into their national immunization programs [18]. In 1995, the FDA approved Varivax (Oka/Merck), a live attenuated varicella vaccine derived from the vOka strain which later received European approval in 2003. Clinical data confirmed its favorable safety profile and tolerability with minimal vaccine-related adverse events [19,20]. Following this, VarilRix (Oka/GSK) gained FDA approval in 2006, demonstrating high clinical efficacy in pediatric populations during trials [21]. In 2008, BCHT’s live attenuated varicella vaccine was approved in China [22]. Phase III trials of SINOVAC’s live attenuated varicella vaccine achieved a 97.1% seroconversion rate in children aged 1–12 years, with protective efficacy rates of 87.1% against confirmed varicella, 89.2% against breakthrough cases, and 100% against moderate-to-severe disease, while maintaining a favorable safety profile [23]. This vaccine received Chinese approval and attained World Health Organization (WHO) prequalification (PQ) in 2019 [23]. Recent advancements include clinical studies by the Shanghai Institute of Biological Products in 2022, which established strong immunogenicity and safety in individuals ≥13 years using a two-dose regimen with a 4–8 week interval. Consequently, the CDE broadened the vaccine’s target population in 2024 to include all individuals ≥12 months [24].
Beyond the vOka strain, alternative vaccine strains have been developed. The MAV/06 strain, first developed in the 1990s, exhibits significant genetic similarity with the vOka strain. Since 1998, Suduvax® (GC Biopharma), a live attenuated vaccine utilizing the MAV/06 strain, has been commercially available worldwide [25]. More recently, in 2020, BARYCELA® (GC Biopharma), a novel MAV/06-based vaccine, received approval from the Korean Ministry of Food and Drug Safety (KMFDS) [26]. These MAV/06-based vaccines potently induce both humoral and cellular immunity through enhanced Th1-mediated immune responses [26,38]. They elicit robust antibody responses and cross-immunity against circulating Oka strains, demonstrating comparable efficacy to commercial vOka-based vaccines [26,38].
Recent years have witnessed significant advancements in the development of varicella vaccines. In 2022, Wang et al. engineered VZV-7D, a novel live attenuated varicella vaccine with dual attenuation properties targeting both skin and neural tissues [39]. The Phase IIa clinical trial results, published in 2024, revealed that VZV-7D induced humoral immunogenicity comparable to the vOka vaccine in children aged 3–12 years, while maintaining excellent tolerability, underscoring its potential as a safer next-generation live attenuated varicella vaccine [27]. Regulatory progress includes the CDE approval of BOAOVAX’s live attenuated varicella vaccine for Investigational New Drug (IND) status in January 2024 [28]. Furthermore, in December 2024, the CDE granted IND acceptance for Zhejiang Toyouvax Biopharming’s live attenuated varicella vaccine candidate [29].
The live attenuated varicella vaccine is routinely administered subcutaneously. Current vaccination guidelines recommend administering the first dose to children aged 12–18 months [40]. In China, while a single dose demonstrates 75% efficacy in children aged 1–12 years, its efficacy declines significantly in those aged ≥6 years [41]. In the United States, outbreaks continue to occur among highly vaccinated students following single-dose administration, with breakthrough cases occurring 3.4 times more frequently than with two-dose regimens [42]. Therefore, since 2006, the Advisory Committee on Immunization Practices (ACIP) has advocated for a second dose for children aged 4–6 years to establish long-term immunity [42]. Notably, two-dose vaccination achieves 94.9% protective efficacy, representing an approximate 20% increase over single-dose administration [43].

2.2. Varicella Combination Vaccines

Advances in varicella combination vaccines have broadened immunization strategies. Typically, the live attenuated varicella vaccine is formulated with the measles, mumps, and rubella (MMR) vaccine to produce a quadrivalent MMRV vaccine. This combined approach simplifies vaccination schedules and enhances coverage rates. Studies have demonstrated that the MMRV vaccine induces immunogenicity comparable to separately administered MMR and varicella (MMR + V) vaccines [44].
The introduction of the MMRV vaccine has significantly influenced public health in the United States. Vaccination coverage rates among children aged 19–35 months have surpassed 90% for either the quadrivalent MMRV or the separate trivalent MMR + V vaccines [30]. Following widespread adoption of the vaccination program, the incidence rates for measles, mumps, rubella, and varicella have decreased substantially, falling to approximately 0.14%, 1.05%, 0.01%, and 10% of pre-vaccine levels, respectively [30].
Since the mid-2000s, two MMRV vaccines have been commercially available: ProQuad® (Merck) and Priorix-Tetra® (GSK) [30]. These formulations differ in their live virus strain compositions. ProQuad incorporates the Edmonston strain (measles), Jeryl Lynn™ strain (mumps), Wistar RA 27/3 strain (rubella), and Oka/Merck strain (varicella), whereas Priorix-Tetra utilizes the Schwarz strain (measles), RIT 4385 strain (a Jeryl Lynn™ derivative for mumps), Wistar RA 27/3 strain (rubella), and Oka/RIT strain (varicella). The recommended MMRV vaccination protocol follows a two-dose schedule: the first dose is administered at 12–15 months to establish immunity before preschool, and the second dose is given at 4–6 years, consistent with preschool vaccination requirements.
A ten-year follow-up study in Russia assessed the efficacy, antibody persistence, and safety of single-dose (Priorix, GSK + Varilrix, GSK) and two-dose (Priorix-Tetra, GSK) vaccination regimens in children. The two-dose regimen achieved 92.4% vaccine efficacy, significantly higher than the 74.7% efficacy observed with single-dose administration [45]. Moreover, anti-VZV seropositivity rates remained above 99.4% in two-dose recipients, compared to 89.7% in single-dose recipients, with comparable safety profiles between groups [45]. These data highlight the clinical benefits of multi-dose vaccination strategies in optimizing immune protection and safety.

3. HZ Vaccines

Accumulating clinical evidence underscores the pivotal role of T-cell immunity in preventing primary infection and viral reactivation [46]. Notably, CD4+ T-cell function exhibits a strong association with HZ pathogenesis, while CD8+ T-cells also contribute to viral control during active HZ by targeting VZV-infected cells [47]. Comparative studies reveal that HZ patients display significantly reduced CD4+ T-cells counts and a diminished CD4+/CD8+ T-cell ratio, with severe cases showing more pronounced CD4+ T-cell depletion relative to mild and moderate cases [46]. These findings collectively indicate that eliciting a robust CD4+ T-cell response is essential for enhancing HZ vaccines efficacy, and optimal protection may benefit from coordinated CD4+ and CD8+ T-cell activation.
In recent years, HZ vaccines have emerged as a crucial tool for preventing and managing HZ and its complications. The development of HZ vaccines employs multiple technological platforms, including traditional live attenuated and inactivated vaccines, as well as novel approaches such as subunit, mRNA, viral vector, and virus-like particle (VLP) vaccines (Table 2). These platforms vary significantly in efficacy, safety, immunogenicity, and production efficiency, reflecting their unique mechanisms of action and potential clinical applications. This technological diversity facilitates flexible vaccination strategies for different populations and informs future vaccine optimization and innovation.

3.1. Approved HZ Vaccines

3.1.1. Live Attenuated HZ Vaccine (ZVL)

Live attenuated HZ vaccines, such as Zostavax, differ from varicella vaccines in both immunological mechanisms and antigenic dosage, containing significantly higher concentrations of VZV. Specifically, Zostavax is formulated with a minimum of 19,400 PFU (plaque-forming units) per dose, compared to Varivax’s minimum of 1350 PFU [59,60]. This increased viral load is essential to stimulate a robust immune response sufficient to mitigate HZ risk.
Before the introduction of Shingrix (GSK), Zostavax (Merck), a live attenuated HZ vaccine (ZVL), served as the primary vaccine for individuals aged ≥60 years. Levin et al. reported that Zostavax significantly boosted VZV-specific cell-mediated immunity (CMI) and antibody responses in this population, with CMI enhancement lasting up to three years and being more pronounced among individuals aged 60–69 years [61]. However, its protective efficacy diminished progressively with increasing age [62], declining from 63.9% in 60–69-year-olds to 37.6% among those aged ≥70 after a single 0.65 mL subcutaneous injection [15]. Notably, Adriana et al. found that a second dose could enhance and prolong CMI responses [62]. In clinical trials, Zostavax exhibited a 63.6% incidence of injection-site reactions within 5 days post-vaccination [63]. Adverse reactions with an incidence of ≥1% included pain (53.9%), erythema (48.1%), swelling (40.4%), pruritis (11.3%), warmth (3.7%), hematoma (1.6%), and induration (1.1%) [63].
Two additional live attenuated HZ vaccines, SkyZoster (SK Bioscience) and Canvar (BCHT), have received regulatory approval. SkyZoster was first approved by the KMFDS in September 2017, followed by authorization from the Malaysian National Pharmaceutical Regulatory Agency (NPRA) on January 9, 2023. A Phase III trial demonstrated SkyZoster’s clinically acceptable immunogenicity and safety profiles [31], with a 2.75-fold increase in anti-VZV antibody titers observed among 824 vaccinated adults ≥50 years. Furthermore, SkyZoster demonstrated non-inferiority to Zostavax in eliciting CMI responses, while maintaining comparable adverse reaction rates within six weeks post-vaccination and no reported serious vaccine-related events through 26 weeks. Meanwhile, BCHT’s single-dose live attenuated HZ vaccine, Canvar®, received market approval on June 3, 2023. Canvar is intended for individuals aged ≥40 years, filling a critical gap in HZ vaccination for the 40–50 age group. Canvar achieved 57.6% overall efficacy against HZ in recipients ≥40 years [32], with age-stratified rates of 37.4% (40–49 years), 62.7% (50–59 years), 64.4% (60–69 years), and 18.6% (≥70 years). The vaccine’s safety profile was favorable, with common adverse reactions (e.g., fever) occurring in <10% of vaccinees [32,64].
While ZVL vaccination is generally considered safe, with most vaccine-associated HZ cases presenting as mild and uncomplicated, breakthrough infections remain possible. Tseng et al. reported the first case of a vaccinated 68-year-old immunocompetent adult with prior wild-type VZV infection who subsequently developed mild, uncomplicated HZ caused by the vaccine strain [65]. In immunocompromised patients, including those who received low-dose immunosuppressive therapy, systemic vOka strain infections may develop, potentially progressing to multi-organ failure [66]. An Australian surveillance study (2016–2020) of 1,089,966 ZVL doses reported 854 adverse events following immunization (AEFI) (78.4/100,000 doses), including 14 disseminated VZV infections (6 confirmed and 8 suspected), yielding a confirmed incidence rate of 0.55/100,000 doses. Notably, 5 of 6 confirmed cases occurred among immunocompromised individuals, with 3 resulting in fatalities. Moreover, studies have identified an association between ZVL vaccination and elevated giant cell arteritis incidence, particularly for clinically and pathologically confirmed cases [67]. These findings highlight the risk of life-threatening vaccine-strain infections in severely immunocompromised individuals, emphasizing the need for pre-vaccination immune evaluation and rigorous post-vaccination surveillance [68].

3.1.2. Recombinant Subunit HZ Vaccine (RZV)

Recombinant zoster vaccine (RZV) induces immunity through recombinant VZV antigenic proteins, thereby eliminating the requirement for intact viral particles. Current RZVs primarily target glycoprotein E, the most abundant VZV envelope protein [69,70]. Functionally, gE forms a heterodimer complex with gI, acting as an Fc receptor in infected cells, which facilitates viral recognition, dissemination, and immune evasion [71]. gE’s close association with host infection mechanisms renders it an optimal antigenic target for HZ vaccine design. Unlike live attenuated vaccines, RZV eliminates the potential for vaccine-strain breakthrough infections or reactivation, while effectively eliciting targeted antibody production and CMI responses.
Shingrix (GSK) is the most widely utilized recombinant zoster vaccine. The FDA granted approval to Shingrix (HZ/Su or GSK1437173A), a two-dose recombinant HZ vaccine, on 20 October 2017 [72]. The vaccination regimen consists of two 0.5 mL intramuscular doses administered 2–6 months apart [73]. This formulation combines recombinant gE with the proprietary AS01B adjuvant system. As the principal target of neutralizing antibodies, gE plays a pivotal role in vaccine efficacy. AS01B are composed of liposomes, monophosphoryl lipid A (MPL) and QS-21 (a saponin derived from Quillaja saponaria), enhancing immunogenicity by stimulating antigen-presenting cells, thereby promoting antibody production and T-cell responses.
On 25 October 2017, ACIP recommended Shingrix for use in immunocompetent adults aged ≥50 years. Although mild local and systemic adverse reactions are common, Shingrix maintains an overall favorable safety and tolerability profile [74]. In clinical studies, Shingrix-induced humoral and CMI responses persisted above pre-vaccination levels for at least 9 years in adults ≥60 years, with modeling data predicting protection lasting up to 15 years [75]. Post-marketing surveillance demonstrated vaccine efficacy rates of 70.1% among ≥65-year-olds receiving the two-dose regimen versus 56.9% for single-dose recipients [76]. Importantly, Shingrix has shown to be effective in immunocompromised populations [76].
The intensity of gE-specific immune responses varies with adjuvant dosage [77]. However, Shingrix’s adjuvant system is associated with side effects, and the scarcity of QS-21 constrains widespread use. Common adverse events comprise pain and fatigue, with 85% of recipients reporting local or systemic reactions in pre-licensure clinical trials [78].
Although rare, individual responses to Shingrix exhibit considerable variability. Schmidt and Maitland reported a 63-year-old male patient who developed acute immune thrombocytopenia (ITP) within two weeks post-vaccination, which subsequently resolved following dexamethasone and intravenous immunoglobulin therapy [79]. This case represents the first reported instance of Shingrix®-induced ITP. Despite its well-established efficacy, continued research remains essential to comprehensively characterize its side effects and interindividual response.
RZV is a safe and effective option for immunocompromised patients, offering the potential to reduce the health and socioeconomic burden of HZ [80]. Dagnew et al. reported 87.2% protective efficacy against HZ in immunocompromised patients with hematologic malignancies, with a favorable safety profile [81]. This robust protection highlights RZV’s potential as a preferred alternative to liveattenuated vaccines in immunocompromised populations. Although breakthrough infections following RZV vaccination are exceedingly rare, certain high-risk subgroups may remain vulnerable. For instance, Chen et al. reported the first case of VZV infection manifesting as acute retinal necrosis in an immunocompromised woman after a single Shingrix dose [82]. The cases highlight the importance of tailored vaccination strategies, considering individual treatment regimens to ensure safety and maximize protective efficacy in high-risk populations.

3.2. Clinical HZ Vaccines

3.2.1. Recombinant Subunit HZ Vaccine (RZV)

Recent advances in the development of RZV have focused on optimizing immunogenicity and safety profiles beyond the benchmarks set by Shingrix.
Several companies are advancing antigen innovations to boost immune responses. Among these, LZ901, developed by Beijing Luzhu Biotechnology Co., Ltd. and GNW002, developed by Genavax, both utilize gE-Fc fusion proteins as antigens. LZ901 is the first global zoster vaccine featuring a tetrameric molecular structure in an Fc fusion protein format, adjuvanted with aluminum [33]. Extensive preclinical studies and Phase I/II clinical trials in China have demonstrated its high immunogenicity, efficacy, and safety [33]. The FDA granted IND approval for LZ901 in July 2022, and its Phase I trial in the United States commenced in February 2023. Meanwhile, GNW002, a Chinese hamster ovary (CHO) cell-based RZV, employs a dual-adjuvant platform and a gE dimer fusion protein, showing preclinical safety and immune responses comparable to Shingrix [52]. GNW002 received IND approvals in China and the United States in January 2024, with its Phase I trial in China initiating in September 2024. Another candidate, LYB004, a nanoparticle-based zoster vaccine developed by Patronus Biotech using its proprietary U-VLP™ platform, received approval for clinical trials from CDE in July 2024 [28]. Additionally, Gentize Biopharma (Nanjing) developed an RZV using DNA recombination technology to express recombinant gE in CHO cells, demonstrating significant protective efficacy against zoster. Its IND application was accepted by the CDE in September 2024 [28,56].
In addition to advancements in antigen innovation, several companies have developed novel adjuvant systems to enhance immune responses. Maxvax’s CHO cell-based RZV, which incorporates the novel composite adjuvant MA105, demonstrated favorable safety, tolerability, and immunogenicity in Phase I/II clinical trials, with a Phase III trial initiated in July 2024 [35]. Meanwhile, REC610, developed by Recbio and formulated with the novel adjuvant BFA01, showed acceptable safety and tolerability in a Phase I trial in the Philippines involving healthy adults aged ≥40 after two doses [34]. REC610 elicited robust gE-specific humoral and cellular immune responses, detectable immediately after the first dose and peaking 30 days after the second dose, with immune responses comparable to Shingrix [34]. REC610 entered Phase III trials in China in October 2024. Furthermore, Grand Theravac Life Science Nanjing Co. Ltd. developed an RZV incorporating the novel adjuvant system TVA01, which elicited superior cellular and humoral immune responses compared to Shingrix in preclinical studies, alongside favorable safety profiles [28]. This vaccine received approval from the Australian Human Research Ethics Committee in August 2023, leading to a Phase I trial in Australia, followed by a Phase I trial in China in December 2024.
Several additional RZVs are currently under development, though some details remain undisclosed. In June 2024, Dynavax commenced a Phase I/II randomized, observer-blind, active-controlled, dose-escalation, multicenter trial in Australia for its CpG 1018-adjuvanted zoster vaccine (z-1018) to assess safety, tolerability, and immunogenicity compared to Shingrix [48]. Separately, EuBiologics’s EuHZV demonstrated an efficacy rate exceeding 90% in its Phase I trial, which began in the United States in July 2024 [53].

3.2.2. mRNA Vaccines for Herpes Zoster

mRNA vaccines employ nucleic acid fragments encoding VZV antigens, which direct host cells to produce specific antigenic proteins. This mechanism induces robust and durable humoral and cellular immune responses. The distinct antigen expression pattern and generation sites in mRNA vaccines facilitate strong CMI, offering significant potential for HZ vaccines development [83]. Additionally, mRNA vaccines offer enhanced safety profiles by eliminating live viral components, thereby reducing infection risks and substantially accelerating vaccine development and production timelines.
Since 2020, the extensive clinical use of mRNA vaccines developed by Moderna and BioNTech for COVID-19 has demonstrated their safety profile and protective efficacy. This success has validated mRNA technology as a promising vaccine platform, stimulating worldwide research interest in developing mRNA-based vaccines for HZ.
Several investigational candidates are currently undergoing early-stage clinical trials to evaluate their safety, tolerability, and preliminary efficacy. Moderna’s HZ vaccine candidate, mRNA-1468, induced strong antigen-specific T-cell responses one month after a second dose in Phase I/II trials, with favorable tolerability supporting its continued clinical development for HZ prevention [49]. Separately, Pfizer and BioNTech initiated a Phase I/II trial for their mRNA-based HZ vaccine candidate in February 2023 [50,51]. Additionally, Innorna’s mRNA vaccine IN00 1receieved FDA approval for clinical trials in September 2023, followed by clearance from CDE in July 2024, enabling the commencement of its Phase I study [54].
Recent advancements in mRNA vaccine technology have highlighted the unique advantages of self-replicating RNA (srRNA), which enables substantial dose reduction while enhancing antigen expression levels [84]. Immorna’s mRNA-based HZ vaccine candidate, JCXH-105, utilizes the intrinsic adjuvant properties of srRNA to activate innate immune cells, thereby promoting CD4+ T-cell activation and memory cell generation [85]. This immunogenic mechanism may potentially prevent HZ recurrence. Following FDA approval, JCXH-105 entered Phase II trials on 16 October 2024 [86].
Several additional candidates have received IND approvals. In October 2024, Rhegen’s self-developed lyophilized mRNA vaccine for HZ received implicit approval from the CDE, becoming the world’s first lyophilized mRNA vaccine for HZ with independent intellectual property rights [57]. Subsequently, in November 2024, the CDE accepted the IND application for CSPC Pharmaceutical Group Limited’s mRNA vaccine, SYS6017 [28]. On 25 December 2024, the NMPA approved the clinical trial application for SINOVAC’s lyophilized mRNA vaccine against HZ, which is designed to induce immunity against VZV in individuals aged ≥40 [58].

3.2.3. Inactivated HZ Vaccines

Inactivated HZ vaccines are produced by chemically or physically inactivating VZV, preserving its immunogenicity while eliminating its pathogenicity. This process enables the vaccine to safely stimulate an immune response, providing protection against VZV without infection risk. Despite their potential, these vaccines exhibit relatively low immunogenicity. However, incorporating novel adjuvants and advanced delivery systems offers a promising approach to enhance their immune efficacy. To date, no inactivated HZ vaccine has obtained regulatory approval.
Immunocompromised individuals face an increased risk of VZV infection, making inactivated HZ vaccines a safe and practical preventive alternative for this population. Clinical studies have shown that inactivated VZV vaccines (ZVIN) induce strong VZV-specific immune responses in immunocompromised or immunodeficient adults, including those with autoimmune diseases, patients with solid tumors undergoing chemotherapy, and individuals receiving anti-CD20 monoclonal antibody therapy for hematologic malignancies [87,88,89]. These vaccines exhibit favorable safety, tolerability, and efficacy profiles. A pivotal Phase III trial demonstrated that V212 (Merck’s inactivated HZ vaccine) reduced the incidence of HZ by 64% in immunocompromised participants [36]. Further comprehensive clinical trials, combined with long-term studies, are needed to evaluate the clinical effectiveness of inactivated HZ vaccines and assess the influence of multi-dose vaccination strategies on the durability of protective immunity.

3.2.4. Herpes Zoster Viral Vector Vaccine

HZ viral vector vaccine incorporates specific gene fragments of VZV into a viral vector to elicit an immune response. This strategy leverages the vector virus’s natural infectivity for efficient delivery and presentation of VZV antigens (predominantly gE), thereby effectively stimulating the host immune system. The flexibility of viral vectors allows for genetic engineering to optimize immunogenicity and stability, further enhancing their therapeutic potential. Viral vector vaccines are designed to combine the immunogenicity of live attenuated vaccines with the safety profile of subunit vaccines, offering a safer and more effective immunization strategy for diverse populations.
Commonly used viral vectors, such as adenovirus or vaccinia virus, are genetically modified to maximize human safety while maintaining high-efficiency antigen expression and potent immune activation. Preclinical studies of the adenovirus-vectored HZ vaccine candidate CS-2032, co-developed by CanSinoBIO and Vaccitech, demonstrated its ability to induce both humoral and cellular immunity [55]. Notably, the resulting humoral immune response was comparable to that induced by Shingrix, while the cellular immune response was significantly enhanced [55]. A Phase I trial of CS-2032 commenced in Canada in November 2023.

3.3. Technical Approaches to Novel HZ Vaccines

Future research on ZVL should focus on exploring novel administration routes to enhance immunogenicity and safety. Studies have shown that intradermal (ID) administration significantly elevates VZV-specific antibody titers compared to subcutaneous (SC) delivery at equivalent doses [90]. Notably, even one-fifth of the intradermal dose can elicit a VZV-specific cellular immunity comparable to or exceeding that achieved via SC injection [91]. Additionally, advancements in vaccine preparation, such as the recombinant vOka bacterial artificial chromosome (rvOka-BAC) system for recombinant live vaccines, offer promising strategies to improve efficacy and safety profiles [92]. Notably, research by Gershon et al. revealed that low levels of constitutive interleukin-10 (IL-10) enhance both humoral immunity and VZV-specific T-cell responses induced by Zostavax, suggesting that pre-vaccination IL-10 modulation could enhance vaccine efficacy [93]. These findings provide valuable insights for optimizing live attenuated HZ vaccines.
Recent advancements in vaccine technology have accelerated the development of RZVs, driven by innovations in immunogen design, adjuvant systems and delivery platforms to enhance immunogenicity. For instance, a novel HZ vaccine incorporating a fusion antigen of glycoprotein E and I (gEgI) adjuvanted with XUA09C demonstrated superior early immune activation, B- and T-cell proliferation and memory T-cell augmentation compared to Shingrix, with stronger cellular immune responses observed in both adult and aged murine models [94]. Similarly, a VZV-gE vaccine formulated with an LNP-CpG ODN adjuvant (a lipid nanoparticle encapsulating VZV-gE and CpG oligodeoxynucleotide) demonstrated comparable immunogenicity and CMI to Shingrix [95]. Notably, all components of this vaccine are scalable for production, highlighting its potential as a cost-effective alternative for HZ prevention. Further optimization of vaccine efficacy relies on precise identification and targeting of immunodominant epitopes. Studies show a truncated gE protein (tgE, aa 31-358) expressed in Escherichia coli (E. coli) with AS01B adjuvant induced antibody and CMI responses comparable to Shingrix, especially for IFN-γ+ CD4+ T-cells [96]. This E. coli system provides a practical gE production method. Among the gE fragments tested, tgE350 showed particularly strong neutralization, informing truncated gE-based vaccine design [97]. Emerging protein engineering and nanoparticle technologies are expected to further improve the potency and stability of next-generation subunit vaccines.
The development of mRNA vaccines presents significant advantages, primarily due to their inherent flexibility, which enables rapid adaptation to genetic variations while enhancing efficacy. Optimizing the gE sequence can further enhance vaccine immunogenicity. Studies indicate that an mRNA-LNP encoding a C-terminal double mutant of gE—engineered to increase viral spread and titer—elicits humoral and cellular immune responses comparable to or stronger than those of Shingrix [98]. Moreover, an LNP-encapsulated mRNA vaccine, gE-M-P, developed by fusing the gE-M nucleic acid sequence with the untranslated region (UTR) of Pfizer/BioNTech’s BNT162b2, has induced robust humoral and cellular immune responses in mice [99]. Further optimization of a full-length gE antigen-encoding VZV mRNA vaccine through signal peptide replacement, C-terminal modification, and mRNA stabilization motifs has substantially enhanced immunogenicity, resulting in superior gE-specific antibody responses, memory B-cell activation and Th1-type T-cell responses across multiple mouse models [100].
In mRNA vaccine development, novel adjuvants and LNP delivery systems offer promising strategies for improving HZ prevention. Research has demonstrated that the adjuvant CIA09, which composed of 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP)-based cationic liposomes combined with the Toll-like receptor 4 agonist de-O-acylated lipooligosaccharide (dLOS), enhances antigen uptake, lymph node delivery, and antigen presentation, thereby strengthening the immune response against VZV gE and promoting a Th1-biased immune response [101]. Similarly, ionizable LNPs amplify the synergistic adjuvant effects of CpG-ODNs and QS21 on VZV gE, improving both humoral and cellular immunity [102]. The HZ vaccine combining VZV gE with the LNP-CpG-QS21 adjuvant system shows potential as a novel alternative to Shingrix, with high cholesterol content in LNPs mitigates the cytotoxicity of QS21 [102]. Both CIA09 and LNP-CpG-QS21 may serve as promising adjuvant candidates for augmenting mRNA vaccine efficacy.
Although HZ viral vector vaccines remain in experimental phases, preliminary data suggest their potential to elicit robust immune protection. A recombinant baculovirus vector vaccine, AcHERV gE-gB, induced total IgG, IgG2a, and neutralizing antibodies levels in mouse models that were comparable to or exceeded those produced by the vOka vaccine, while also stimulating strong VZV-specific CMI [103]. Future research should focus on developing safe and efficient viral vectors, optimizing antigen delivery and immune responses, and assessing their compatibility with existing vaccines.
HZ VLP vaccines, which display conserved VZV epitopes, represent a promising novel immunization strategy. Recombinant hybrid transposon yeast-VLPs (Ty-VLPs) expressing the immunodominant sequence of VZV gE elicit robust humoral and cellular immune responses, demonstrating neutralizing activity against VZV in small animal models [104]. Similarly, another study showed that Ty-VLPs presenting VZV gE or assembly protein fragments act as potent immunogens, stimulating strong immune responses, and were the first to induce specific lymphocyte responses to VZV assembly protein in varicella and HZ patients [85]. Furthermore, Zhu et al. utilized recombinant hepatitis B virus core (HBc)-VLPs to confirm the immunogenicity and neutralizing activity of a highly conserved gE epitope (residues 121–135), providing insights for epitope-based HZ vaccine design [105]. Additionally, a nanoparticle-based HZ vaccine, FR-gE, has also been shown to induce stronger neutralizing antibody responses than both live attenuated vaccines and Shingrix [106]. Future studies should focus on expanding the VLP platform, enhancing immunization strategies, and validating their efficacy to establish VLPs as a viable alternative in HZ vaccine development.

4. Discussion

VZV vaccines have significantly advanced the prevention of varicella (chickenpox) and HZ. Their global implementation has led to a substantial reduction in both the incidence and complications associated with these diseases.
Varicella vaccine development has primarily utilized live attenuated technology, which is now well-established. While this approach has demonstrated good tolerability and immunogenicity in healthy populations, attenuated VZV retains replicative capacity, and may cause breakthrough infections. Future research should investigate VZV virulence factors and attenuation mechanisms, with particularly emphasis on identifying key virulence determinants to enable safer vaccine design. Novel varicella vaccines or sequential immunization strategies should be explored to overcome diminished immune responses and breakthrough infections in immunocompromised individuals. Continued innovation, incorporating emerging technologies, will be crucial for the future development of varicella vaccines.
Similarly, both ZVLs and RZVs have reached advanced development stages. Shingrix has demonstrated high efficacy in high-risk populations and the elderly, with minimal breakthrough infection risk. Current research prioritizes novel vaccine platforms, with key focuses for RZVs including immunogen redesign [33], more potent adjuvant development [34], and alternative administration route exploration [91]. In mRNA vaccine development, efforts concentrate on sequence optimization [37] and LNP-based delivery systems advancement [54]. VLP vaccine efficacy depends on antigen structures optimization [106], while viral vector vaccine progress depends on novel vector identification [55]. These innovative platforms show potential for enhanced safety and durable immune protection, though large-scale clinical trials remain necessary to verify their efficacy and clinical applicability.
Despite its general safety and efficacy, the vOka vaccine strain maintains replicative capacity and can infect host cells, potentially establishing latency in dorsal root ganglia and transmitting to susceptible individuals. The administration of live attenuated varicella vaccines in immunocompromised individuals presents particular challenges due to elevated risks of vaccine-strain infection and transmission, which may result in severe or even fatal complications [107,108]. Documented cases include fatal infections in an immunodeficient child and a stem cell transplant recipient with non-Hodgkin lymphoma [109,110]. Among immunocompromised patients, including those receiving low-dose immunosuppressive therapy, ZVL vaccination may cause systemic infection by the vaccine strain of VZV, potentially leading to multi-organ failure [66]. These cases highlight the need for rigorous risk assessment and careful clinical evaluation when considering live attenuated vaccines for immunocompromised populations. Although live attenuated VZV vaccines carry inherent risks, their demonstrated protective benefits and manageable safety profile support their continued inclusion in routine immunization programs.
Unlike ZVL, breakthrough infections associated with RZV are exceedingly rare. RZV represents a safe and effective option for immunocompromised individuals, with potential to mitigate the health and socioeconomic burdens of HZ [80]. Studies have shown that RZV is well-tolerated, safe, and effective in immunocompromised populations such as autologous hematopoietic stem cell transplant recipients and patients with hematologic malignancies [81,111]. Furthermore, emerging next-generation vaccine technologies, particularly mRNA-based platforms, may offer improved safety and immunogenicity in future vaccine development.
The COVID-19 pandemic has introduced new complexities in vaccine management and efficacy, with reports of VZV reactivation following COVID-19 vaccination [112,113,114]. These observations highlight the need for further investigation into vaccine interactions and their immunological consequences. Anti-SARS-CoV-2 vaccines may modulate immune responses, potentially influencing VZV reactivation, particularly in individuals with age-related immune decline (immunosenescence). Consequently, vaccination strategies should account for these immunological considerations. Pre-vaccination immune assessments, particularly in high-risk populations, as well as post-vaccination surveillance, are essential to mitigate adverse effects and transmission risks.
Furthermore, the efficacy of these vaccines may be influenced by recipients’ baseline immune status before vaccination. Obesity has been linked to reduced seroconversion rates, potentially impairing vaccine-induced immune responses [115]. Likewise, untreated depression may increase the risk and severity of HZ while also reducing vaccine effectiveness. In contrast, antidepressant therapy can restore immune function and potentially improve vaccine efficacy [116]. These findings emphasize the critical role of maintaining overall health to support optimal antiviral immunity.
The success of VZV vaccines underscores their potential as platforms for broader infectious disease prevention. Given its ability to express foreign genes, VZV serves as a versatile viral vector in vaccine design, offering potential for a wide range of viral infections. VZV-based vaccines have been explored in HIV vaccine development, demonstrating potential to enhance both systemic and genital mucosal immunity [117]. Similarly, a VZV vector expressing simian immunodeficiency virus (SIV) antigens has demonstrated a 37.5% protection rate against pathogenic SIV challenges by eliciting mucosal memory, humoral, and cellular immune responses [118]. These findings underscore the potential of VZV as a robust platform for developing vaccines against immunodeficiency viruses. Furthermore, a recombinant VZV expressing Epstein–Barr virus (EBV) membrane glycoprotein (gp350/220) has emerged as a promising vector for active immunization against EBV and other pathogens [119].
In summary, vaccination against varicella and HZ remains a critical public health strategy, offering substantial benefits in preventing severe disease and reducing mortality. The benefits of VZV vaccination significantly outweigh the minimal risks of adverse effects. However, current research identifies key gaps in VZV vaccine development, particularly in enhancing vaccine diversity, efficacy, and safety. The continued development and refinement of safe and effective VZV vaccines, along with increased vaccination coverage, can substantially reduce varicella and HZ incidence, prevent severe complications, establish herd immunity, interrupt viral transmission, and mitigate healthcare costs. Continued scientific advancements and technological innovations show considerable promise for enhancing the public health benefits of VZV vaccines, ultimately strengthening global immunization efforts and contributing to long-term disease control.

Author Contributions

S.L. and T.L. designed and funded the study. H.L. (Hongjing Liu), L.C. (Lingyan Cui), and S.Z. wrote the original draft, reviewed, and edited the manuscript, H.W., W.X., H.L. (Hai Li), Y.Z., L.C. (Lin Chen), Y.G., N.X., T.L., and S.L. participated in discussion and interpretation of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by grants from the National Key Research and Development Program of China (grant no: 2024YFC2310401) and the Fundamental Research Funds for the Central Universities (20720220004, 20720220006).

Data Availability Statement

All relevant data are within the manuscript.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

ACIPAdvisory Committee on Immunization Practices
AEFIAdverse events following immunization
bpBase pair
CDECenter for Drug Evaluation
CHOChinese hamster ovary (cells)
CMICell-mediated immunity
CpGCytosine-phosphate-Guanine
dLOSde-O-acylated lipooligosaccharide
DOTAP1,2-dioleoyl-3-trimethylammonium-propane
E. coliEscherichia coli
EBVEpstein–Barr virus
FDAFood and Drug Administration
gEGlycoprotein E
gEgIGlycoprotein E and I
GPEGuinea pig embryo
HBcHepatitis B virus core
HELHuman embryonic lung
HHV-3Human herpesvirus 3
HIVHuman immunodeficiency virus
HZHerpes zoster
IDIntradermal
IL-10Interleukin-10
INDInvestigational New Drug
ITPImmune thrombocytopenia
KMFDSKorean Ministry of Food and Drug Safety
LNPLipid nanoparticle
LNP-CpG-ODNLipid nanoparticle encapsulating CpG-ODN
MMRMeasles–mumps–rubella
MMR + VMeasles–mumps–rubella and varicella
MMRVMeasles–mumps–rubella–varicella
MPLMonophosphoryl lipid A
NMPANational Medical Products Administration
NPRANational Pharmaceutical Regulatory Agency
ODNOligodeoxynucleotide
ORFOpen reading frame
PFUPlaque-forming units
PHNPostherpetic neuralgia
pOkaParental Oka strain
PQPrequalification
QS-21A saponin derived from Quillaja saponaria
rvOka-BACRecombinant vOka bacterial artificial chromosome
RZVRecombinant zoster vaccine
SCSubcutaneous
SIVSimian immunodeficiency virus
srRNASelf-replicating RNA
tgETruncated gE protein
Ty-VLPsHybrid transposon yeast-VLPs
UKUnited Kingdom
USUnited States
UTRUntranslated region
U-VLP™Patronus Biotech’s proprietary nanoparticle platform
VLPVirus-like particle
vOkaVaccine Oka strain
VZVVaricella-zoster virus
WHOWorld Health Organization
WI-38Human diploid cell line
ZVINInactivated VZV vaccine
ZVLLive attenuated HZ vaccine

References

  1. Davison, A.J.; Scott, J.E. The complete DNA sequence of varicella-zoster virus. J. Gen. Virol. 1986, 67 Pt 9, 1759–1816. [Google Scholar] [CrossRef]
  2. Storlie, J.; Maresova, L.; Jackson, W.; Grose, C. Comparative analyses of the 9 glycoprotein genes found in wild-type and vaccine strains of varicella-zoster virus. J. Infect. Dis. 2008, 197 (Suppl. 2), S49–S53. [Google Scholar] [CrossRef]
  3. de Martino Mota, A.; Carvalho-Costa, F.A. Varicella zoster virus related deaths and hospitalizations before the introduction of universal vaccination with the tetraviral vaccine. J. Pediatr. 2016, 92, 361–366. [Google Scholar] [CrossRef]
  4. Lachiewicz, A.M.; Srinivas, M.L. Varicella-zoster virus post-exposure management and prophylaxis: A review. Prev. Med. Rep. 2019, 16, 101016. [Google Scholar] [CrossRef]
  5. Galil, K.; Choo, P.W.; Donahue, J.G.; Platt, R. The sequelae of herpes zoster. Arch. Intern. Med. 1997, 157, 1209–1213. [Google Scholar] [CrossRef]
  6. Schmidt, M.; Kress, M.; Heinemann, S.; Fickenscher, H. Varicella-zoster virus isolates, but not the vaccine strain OKA, induce sensitivity to alpha-1 and beta-1 adrenergic stimulation of sensory neurones in culture. J. Med. Virol. 2003, 70 (Suppl. 1), S82–S89. [Google Scholar] [CrossRef]
  7. Sadaoka, T.; Mori, Y. Vaccine Development for Varicella-Zoster Virus. Adv. Exp. Med. Biol. 2018, 1045, 123–142. [Google Scholar] [CrossRef]
  8. Pepose, J.S. The potential impact of the varicella vaccine and new antivirals on ocular disease related to varicella-zoster virus. Am. J. Ophthalmol. 1997, 123, 243–251. [Google Scholar] [CrossRef]
  9. Tirat, W.R.; Schibler, M. Varicella-zoster virus (VZV) acute retinal necrosis and recombinant zoster vaccine. Rev. Med. Suisse 2022, 18, 714–717. [Google Scholar] [CrossRef]
  10. Newman, A.M.; Jhaveri, R. Myths and Misconceptions: Varicella-Zoster Virus Exposure, Infection Risks, Complications, and Treatments. Clin. Ther. 2019, 41, 1816–1822. [Google Scholar] [CrossRef]
  11. Feigin., R.D.; Cherry., J.D.; Demmler-Harrison., G.J.; Kaplan., S.L. Feigin and Cherry’s Textbook of Pediatric Infectious Diseases; Elsevier: Amsterdam, The Netherlands, 2009; ISBN 978-141-604-044-6. [Google Scholar]
  12. Bialek, S.R.; Perella, D.; Zhang, J.; Mascola, L.; Viner, K.; Jackson, C.; Lopez, A.S.; Watson, B.; Civen, R. Impact of a routine two-dose varicella vaccination program on varicella epidemiology. Pediatrics 2013, 132, e1134–e1140. [Google Scholar] [CrossRef] [PubMed]
  13. Shapiro, E.D.; Vazquez, M.; Esposito, D.; Holabird, N.; Steinberg, S.P.; Dziura, J.; LaRussa, P.S.; Gershon, A.A. Effectiveness of 2 doses of varicella vaccine in children. J. Infect. Dis. 2011, 203, 312–315. [Google Scholar] [CrossRef] [PubMed]
  14. Sheel, M.; Beard, F.; Quinn, H.; Dey, A.; Kirk, M.; Koehler, A.; Markey, P.; McIntyre, P.; Macartney, K. Australian vaccine preventable disease epidemiological review series: Varicella-zoster virus infections, 1998-2015. Commun. Dis. Intell. 2018, 2018, 42. [Google Scholar] [CrossRef]
  15. Bharucha, T.; Ming, D.; Breuer, J. A critical appraisal of ‘Shingrix’, a novel herpes zoster subunit vaccine (HZ/Su or GSK1437173A) for varicella zoster virus. Hum. Vaccin. Immunother. 2017, 13, 1789–1797. [Google Scholar] [CrossRef]
  16. Laing, K.J.; Ouwendijk, W.J.D.; Koelle, D.M.; Verjans, G. Immunobiology of Varicella-Zoster Virus Infection. J. Infect. Dis. 2018, 218, S68–S74. [Google Scholar] [CrossRef]
  17. Haberthur, K.; Engelmann, F.; Park, B.; Barron, A.; Legasse, A.; Dewane, J.; Fischer, M.; Kerns, A.; Brown, M.; Messaoudi, I. CD4 T cell immunity is critical for the control of simian varicella virus infection in a nonhuman primate model of VZV infection. PLoS Pathog. 2011, 7, e1002367. [Google Scholar] [CrossRef] [PubMed]
  18. Takahashi, M. 25 years’ experience with the Biken Oka strain varicella vaccine: A clinical overview. Paediatr. Drugs 2001, 3, 285–292. [Google Scholar] [CrossRef]
  19. Goulleret, N.; Mauvisseau, E.; Essevaz-Roulet, M.; Quinlivan, M.; Breuer, J. Safety profile of live varicella virus vaccine (Oka/Merck): Five-year results of the European Varicella Zoster Virus Identification Program (EU VZVIP). Vaccine 2010, 28, 5878–5882. [Google Scholar] [CrossRef]
  20. Paradis, E.M.; Tikhonov, O.; Cao, X.; Kharit, S.M.; Fokin, A.; Platt, H.L.; Wittke, F.; Jotterand, V. Phase 3, open-label, Russian, multicenter, single-arm trial to evaluate the immunogenicity of varicella vaccine (VARIVAX™) in healthy infants, children, and adolescents. Hum. Vaccines Immunother. 2021, 17, 4183–4189. [Google Scholar] [CrossRef]
  21. Sheffer, R.; Segal, D.; Rahamani, S.; Dalal, I.; Linhart, Y.; Stein, M.; Shohat, T.; Somekh, E. Effectiveness of the Oka/GSK attenuated varicella vaccine for the prevention of chickenpox in clinical practice in Israel. Pediatr. Infect. Dis. J. 2005, 24, 434–437. [Google Scholar] [CrossRef]
  22. BCHT. Live Attenuated Vaccine Against Varicella. Available online: http://www.bchtpharm.com/Home/Article/detail/id/804.html (accessed on 23 August 2021).
  23. SINOVAC. SINOVAC Varicella Attenuated Live Vaccine Has Been Pre Certified by the World Health Organization. Available online: http://www.sinovac.com.cn/zh-cn/news/id-3194 (accessed on 9 November 2022).
  24. SINOPHARM. The Vaccination Age for the Varicella Attenuated Live Vaccine of Shanghai Institute of Biological Products Co., Ltd. Has Been Extended to the Entire Population Aged 12 Months and Above. Available online: https://www.sinopharm.com/2024-08/27/c_19710.htm (accessed on 27 August 2024).
  25. Kim, J.I.; Jung, G.S.; Kim, Y.Y.; Ji, G.Y.; Kim, H.S.; Wang, W.D.; Park, H.S.; Park, S.Y.; Kim, G.H.; Kwon, S.N.; et al. Sequencing and characterization of Varicella-zoster virus vaccine strain SuduVax. Virol. J. 2011, 8, 547. [Google Scholar] [CrossRef] [PubMed]
  26. Shin, D.; Shin, Y.; Kim, E.; Nam, H.; Nan, H.; Lee, J. Immunological characteristics of MAV/06 strain of varicella-zoster virus vaccine in an animal model. BMC Immunol. 2022, 23, 27. [Google Scholar] [CrossRef] [PubMed]
  27. Pan, H.X.; Qiu, L.X.; Liang, Q.; Chen, Z.; Zhang, M.L.; Liu, S.; Zhong, G.H.; Zhu, K.X.; Liao, M.J.; Hu, J.L.; et al. Immunogenicity and safety of an ORF7-deficient skin-attenuated and neuro-attenuated live vaccine for varicella: A randomised, double-blind, controlled, phase 2a trial. Lancet Infect. Dis. 2024, 24, 922–934. [Google Scholar] [CrossRef] [PubMed]
  28. Center For Drug Evaluation, NMPA. Disclosure of Recipient Variety Information. Available online: https://www.cde.org.cn/main/xxgk/listpage/9f9c74c73e0f8f56a8bfbc646055026d (accessed on 19 January 2024).
  29. Pharmacy, B. Announcement of Shandong Stepwise Pharmaceutical Co., Ltd. on the Acceptance Notice of Clinical Trials of Its Controlled Subsidiary’s Drugs. Available online: https://www.cnpharm.com/upload/resources/file/2024/12/19/167923.pdf (accessed on 18 December 2024).
  30. Kowalzik, F.; Faber, J.; Knuf, M. MMR and MMRV vaccines. Vaccine 2018, 36, 5402–5407. [Google Scholar] [CrossRef]
  31. Newswire, P. SK Bioscience’s Zoster Vaccine Receives Biologics License Application Approval in Malaysia. Available online: https://en.prnasia.com/releases/global/sk-bioscience-s-zoster-vaccine-receives-biologics-license-application-approval-in-malaysia-389846.shtml (accessed on 9 January 2023).
  32. BCHT. Live Attenuated Herpes Zoster Vaccine: Canvar®. Available online: http://www.bchtpharm.com/Home/Article/detail/id/905.html (accessed on 13 April 2023).
  33. Biotech, L. Recombinant Herpes Zoster Vaccine (LZ901). Available online: http://www.luzhubiotech.com/techAndProduct/productInResearch (accessed on 7 January 2025).
  34. Zhang, Z.; Liu, X.; Suo, L.; Zhao, D.; Pan, J.; Lu, L. The incidence of herpes zoster in China: A meta-analysis and evidence quality assessment. Hum. Vaccin. Immunother. 2023, 19, 2228169. [Google Scholar] [CrossRef]
  35. Maxvax. Milestones|Phase III Clinical Enrollment of Maikokang Recombinant Herpes Zoster Vaccine (CHO Cells). Available online: http://www.maxvax.cn/info.aspx?id=119&t=9 (accessed on 13 July 2024).
  36. Merck. In First Phase 3 Trial, Merck’s Investigational Inactivated Varicella Zoster Virus Vaccine (V212) Reduced the Incidence of Confirmed Herpes Zoster Cases by an Estimated 64 Percent in Immunocompromised Subjects. Available online: https://www.merck.com/news/in-first-phase-3-trial-mercks-investigational-inactivated-varicella-zoster-virus-vaccine-v212-reduced-the-incidence-of-confirmed-herpes-zoster-cases-by-an-estimated-64-percent-in-immunocom/ (accessed on 24 February 2017).
  37. Immorna. JCXH-105, the World’s First RNA Shingles Vaccine, Has Been Approved for Phase I Registration Clinical Trials by the US Food and Drug Administration. Available online: https://www.immorna.com/cn/investors-detail-192.html (accessed on 7 January 2023).
  38. Choi, U.Y.; Huh, D.H.; Kim, J.H.; Kang, J.H. Seropositivity of Varicella zoster virus in vaccinated Korean children and MAV vaccine group. Hum. Vaccin. Immunother. 2016, 12, 2560–2564. [Google Scholar] [CrossRef] [PubMed]
  39. Wang, W.; Pan, D.; Fu, W.; Ye, X.; Han, J.; Yang, L.; Jia, J.; Liu, J.; Zhu, R.; Zhang, Y.; et al. Development of a skin- and neuro-attenuated live vaccine for varicella. Nat. Commun. 2022, 13, 824. [Google Scholar] [CrossRef]
  40. Kudesia, G.; Partridge, S.; Farrington, C.P.; Soltanpoor, N. Changes in age related seroprevalence of antibody to varicella zoster virus: Impact on vaccine strategy. J. Clin. Pathol. 2002, 55, 154–155. [Google Scholar] [CrossRef]
  41. Zhang, Z.J.Z.; Suo, L.D.; Zhao, D.; Pan, J.B.; Lu, L. Systematic reviews and evidence quality assessment on effectiveness of 1 dose varicella attenuated live vaccine for healthy children aged 1-12 years in China. Zhonghua Liu Xing Bing Xue Za Zhi 2020, 41, 1138–1144. [Google Scholar] [CrossRef]
  42. Marin, M.; Güris, D.; Chaves, S.S.; Schmid, S.; Seward, J.F. Prevention of varicella: Recommendations of the Advisory Committee on Immunization Practices (ACIP). MMWR Recomm. Rep. 2007, 56, 1–40. [Google Scholar]
  43. Prymula, R.; Bergsaker, M.R.; Esposito, S.; Gothefors, L.; Man, S.; Snegova, N.; Štefkovičova, M.; Usonis, V.; Wysocki, J.; Douha, M.; et al. Protection against varicella with two doses of combined measles-mumps-rubella-varicella vaccine versus one dose of monovalent varicella vaccine: A multicentre, observer-blind, randomised, controlled trial. Lancet 2014, 383, 1313–1324. [Google Scholar] [CrossRef] [PubMed]
  44. Leung, J.H.; Hirai, H.W.; Tsoi, K.K. Immunogenicity and reactogenicity of tetravalent vaccine for measles, mumps, rubella and varicella (MMRV) in healthy children: A meta-analysis of randomized controlled trials. Expert Rev. Vaccines 2015, 14, 1149–1157. [Google Scholar] [CrossRef]
  45. Namazova-Baranova, L.; Habib, M.A.; Povey, M.; Efendieva, K.; Fedorova, O.; Fedoseenko, M.; Ivleva, T.; Kovshirina, Y.; Levina, J.; Lyamin, A.; et al. A randomized trial assessing the efficacy, immunogenicity, and safety of vaccination with live attenuated varicella zoster virus-containing vaccines: Ten-year follow-up in Russian children. Hum. Vaccin. Immunother. 2022, 18, 1959148. [Google Scholar] [CrossRef]
  46. Xing, Q.; Hu, D.; Shi, F.; Chen, F. Role of regulatory T cells in patients with acute herpes zoster and relationship to postherpetic neuralgia. Arch. Dermatol. Res. 2013, 305, 715–722. [Google Scholar] [CrossRef]
  47. Steain, M.; Sutherland, J.P.; Rodriguez, M.; Cunningham, A.L.; Slobedman, B.; Abendroth, A. Analysis of T cell responses during active varicella-zoster virus reactivation in human ganglia. J. Virol. 2014, 88, 2704–2716. [Google Scholar] [CrossRef] [PubMed]
  48. Dynavax. Advancing a Broad Pipeline of Vaccines to Prevent Infectious Diseases. Available online: https://www.dynavax.com/pipeline/#trials (accessed on 1 October 2022).
  49. Moderna. Moderna Advances Multiple Vaccine Programs to Late-Stage Clinical Trials. Available online: https://news.modernatx.com/news/news-details/2024/Moderna-Advances-Multiple-Vaccine-Programs-to-Late-Stage-Clinical-Trials/default.aspx (accessed on 27 March 2024).
  50. Pfizer. Pfizer and BioNTech Initiate Phase 1/2 Study of First mRNA-Based Shingles Vaccine Program. Available online: https://www.pfizer.com/news/announcements/pfizer-and-biontech-initiate-phase-12-study-first-mrna-based-shingles-vaccine (accessed on 10 February 2023).
  51. National Library of Medicine. Study Details|A Study to Learn About a Modified RNA Vaccine Against Shingles in Healthy Adults|ClinicalTrials.Gov. Available online: https://clinicaltrials.gov/study/NCT05703607 (accessed on 30 December 2024).
  52. Genevax. Good News: Geneve Recombinant Herpes Zoster Vaccine Has Obtained FDA Clinical Trial Approval! Available online: https://www.genevax.com.cn/newsinfo/6818959.html (accessed on 27 January 2024).
  53. ClinicalTrials.Gov. Clinical Trial to Evaluate EuHZV in Healthy Adults Aged 50 to 69 Years. Available online: https://clinicaltrials.gov/study/NCT06409494?term=EuHZV&rank=1 (accessed on 16 July 2024).
  54. Innorna. Innorna Shingles mRNA Vaccine IN001 Has Been Approved for Clinical Trials in China. Available online: https://www.innorna.com/cn/news/314.html (accessed on 23 July 2024).
  55. CanSinoBio. CanSinoBio Recombinant Herpes Zoster Vaccine Launches Phase I Clinical Trial in Canada and Completes First Subject Enrollment. Available online: https://www.cansinotech.com.cn/detail-4188 (accessed on 9 November 2023).
  56. Biopharma, G. Recombinant Herpes Zoster Vaccine. Available online: http://www.gentize.com/product/9.html (accessed on 4 September 2024).
  57. RHEGEN. The Clinical Trial Application for the World’s First Freeze-Dried Herpes Zoster mRNA Vaccine Has Been Approved by CDE. Available online: https://www.rhegen.com/show-8-33-1.html (accessed on 23 October 2024).
  58. SINOVAC. SINOVAC Freeze-Dried Herpes Zoster Virus mRNA Vaccine Approved for Clinical Use. Available online: https://www.sinovac.com/zh-cn/news/id-3385 (accessed on 26 December 2024).
  59. Food and Drug Administration. ZOSTAVAX (Zoster Vaccine Live) Frozen Package Insert. Available online: https://www.fda.gov/media/119879/download?attachment (accessed on 29 March 2019).
  60. Food and Drug Administration. Clinical Review Memo—VARIVAX. Available online: https://www.fda.gov/media/166083/download (accessed on 27 February 2023).
  61. Levin, M.J.; Oxman, M.N.; Zhang, J.H.; Johnson, G.R.; Stanley, H.; Hayward, A.R.; Caulfield, M.J.; Irwin, M.R.; Smith, J.G.; Clair, J.; et al. Varicella-zoster virus-specific immune responses in elderly recipients of a herpes zoster vaccine. J. Infect. Dis. 2008, 197, 825–835. [Google Scholar] [CrossRef] [PubMed]
  62. Weinberg, A.; Popmihajlov, Z.; Schmader, K.E.; Johnson, M.J.; Caldas, Y.; Salazar, A.T.; Canniff, J.; McCarson, B.J.; Martin, J.; Pang, L.; et al. Persistence of Varicella-Zoster Virus Cell-Mediated Immunity After the Administration of a Second Dose of Live Herpes Zoster Vaccine. J. Infect. Dis. 2019, 219, 335–338. [Google Scholar] [CrossRef]
  63. RxList. Zostavax. Available online: https://www.rxlist.com/zostavax-drug.htm#side_effects (accessed on 26 September 2024).
  64. DXY. Canvar Product Manual. Available online: https://drugs.dxy.cn/pc/drug/eu8Irmepepm9Rhzuexg72hUofEA==?ky=%E5%B8%A6%E7%8A%B6%E7%96%B1%E7%96%B9%E7%96%AB%E8%8B%97 (accessed on 17 May 2023).
  65. Tseng, H.F.; Schmid, D.S.; Harpaz, R.; LaRussa, P.; Jensen, N.J.; Rivailler, P.; Radford, K.; Folster, J.; Jacobsen, S.J. Herpes zoster caused by vaccine-strain varicella zoster virus in an immunocompetent recipient of zoster vaccine. Clin. Infect. Dis. 2014, 58, 1125–1128. [Google Scholar] [CrossRef]
  66. Dubey, V.; MacFadden, D. Disseminated varicella zoster virus infection after vaccination with a live attenuated vaccine. CMAJ 2019, 191, E1025–E1027. [Google Scholar] [CrossRef]
  67. Agger, W.A.; Deviley, J.A.; Borgert, A.J.; Rasmussen, C.M. Increased Incidence of Giant Cell Arteritis After Introduction of a Live Varicella Zoster Virus Vaccine. Open Forum. Infect. Dis. 2021, 8, ofaa647. [Google Scholar] [CrossRef]
  68. Li-Kim-Moy, J.; Phillips, A.; Morgan, A.; Glover, C.; Jayasinghe, S.; Hull, B.P.; Dey, A.; Beard, F.H.; Hickie, M.; Macartney, K. Disseminated varicella zoster virus infection following live attenuated herpes zoster vaccine: Descriptive analysis of reports to Australia’s spontaneous vaccine pharmacovigilance system, 2016–2020. BMJ Open 2023, 13, e067287. [Google Scholar] [CrossRef] [PubMed]
  69. Cole, N.L.; Grose, C. Membrane fusion mediated by herpesvirus glycoproteins: The paradigm of varicella-zoster virus. Rev. Med. Virol. 2003, 13, 207–222. [Google Scholar] [CrossRef] [PubMed]
  70. Yao, Z.; Jackson, W.; Forghani, B.; Grose, C. Varicella-zoster virus glycoprotein gpI/gpIV receptor: Expression, complex formation, and antigenicity within the vaccinia virus-T7 RNA polymerase transfection system. J. Virol. 1993, 67, 305–314. [Google Scholar] [CrossRef] [PubMed]
  71. Yao, Z.; Grose, C. Unusual phosphorylation sequence in the gpIV (gI) component of the varicella-zoster virus gpI-gpIV glycoprotein complex (VZV gE-gI complex). J. Virol. 1994, 68, 4204–4211. [Google Scholar] [CrossRef]
  72. Dooling, K.L.; Guo, A.; Patel, M.; Lee, G.M.; Moore, K.; Belongia, E.A.; Harpaz, R. Recommendations of the Advisory Committee on Immunization Practices for Use of Herpes Zoster Vaccines. MMWR Morb. Mortal. Wkly. Rep. 2018, 67, 103–108. [Google Scholar] [CrossRef]
  73. Food and Drug Administration. Shingrix [Package Insert]. Available online: https://www.fda.gov/media/108597/download (accessed on 23 May 2025).
  74. Lal, H.; Zahaf, T.; Heineman, T.C. Safety and immunogenicity of an AS01-adjuvanted varicella zoster virus subunit candidate vaccine (HZ/su): A phase-I, open-label study in Japanese adults. Hum. Vaccin. Immunother. 2013, 9, 1425–1429. [Google Scholar] [CrossRef]
  75. Schwarz, T.F.; Volpe, S.; Catteau, G.; Chlibek, R.; David, M.P.; Richardus, J.H.; Lal, H.; Oostvogels, L.; Pauksens, K.; Ravault, S.; et al. Persistence of immune response to an adjuvanted varicella-zoster virus subunit vaccine for up to year nine in older adults. Hum. Vaccin. Immunother. 2018, 14, 1370–1377. [Google Scholar] [CrossRef]
  76. Izurieta, H.S.; Wu, X.; Forshee, R.; Lu, Y.; Sung, H.M.; Agger, P.E.; Chillarige, Y.; Link-Gelles, R.; Lufkin, B.; Wernecke, M.; et al. Recombinant Zoster Vaccine (Shingrix): Real-World Effectiveness in the First 2 Years Post-Licensure. Clin. Infect. Dis. 2021, 73, 941–948. [Google Scholar] [CrossRef]
  77. Chlibek, R.; Bayas, J.M.; Collins, H.; de la Pinta, M.L.; Ledent, E.; Mols, J.F.; Heineman, T.C. Safety and immunogenicity of an AS01-adjuvanted varicella-zoster virus subunit candidate vaccine against herpes zoster in adults >=50 years of age. J. Infect. Dis. 2013, 208, 1953–1961. [Google Scholar] [CrossRef]
  78. Hesse, E.M.; Shimabukuro, T.T.; Su, J.R.; Hibbs, B.F.; Dooling, K.L.; Goud, R.; Lewis, P.; Ng, C.S.; Cano, M.V. Postlicensure Safety Surveillance of Recombinant Zoster Vaccine (Shingrix)—United States, October 2017–June 2018. MMWR Morb. Mortal. Wkly. Rep. 2019, 68, 91–94. [Google Scholar] [CrossRef]
  79. Schmidt, N.; Maitland, H. Acute Immune Thrombocytopenia following administration of Shingrix recombinant zoster vaccine. Am. J. Hematol. 2021, 96, E136–E137. [Google Scholar] [CrossRef]
  80. Ishihara, R.; Watanabe, R.; Shiomi, M.; Katsushima, M.; Fukumoto, K.; Yamada, S.; Okano, T.; Hashimoto, M. Exploring the Link between Varicella-Zoster Virus, Autoimmune Diseases, and the Role of Recombinant Zoster Vaccine. Biomolecules 2024, 14, 739. [Google Scholar] [CrossRef] [PubMed]
  81. Ljungman, P. Varicella zoster virus vaccine in patients with haematological malignancies. Lancet Infect. Dis. 2019, 19, 921–922. [Google Scholar] [CrossRef] [PubMed]
  82. Chen, R.I.; Deaner, J.D.; Srivastava, S.K.; Lowder, C.Y. Acute retinal necrosis following recombinant subunit varicella-zoster virus vaccine. Am. J. Ophthalmol. Case Rep. 2020, 20, 100962. [Google Scholar] [CrossRef]
  83. Chaudhary, N.; Weissman, D.; Whitehead, K.A. mRNA vaccines for infectious diseases: Principles, delivery and clinical translation. Nat. Rev. Drug Discov. 2021, 20, 817–838. [Google Scholar] [CrossRef] [PubMed]
  84. Vogel, A.B.; Lambert, L.; Kinnear, E.; Busse, D.; Erbar, S.; Reuter, K.C.; Wicke, L.; Perkovic, M.; Beissert, T.; Haas, H.; et al. Self-Amplifying RNA Vaccines Give Equivalent Protection against Influenza to mRNA Vaccines but at Much Lower Doses. Mol. Ther. 2018, 26, 446–455. [Google Scholar] [CrossRef]
  85. Welsh, M.D.; Harper, D.R.; Garcia-Valcarcel, M.; Fowler, W.J.; Aitken, C.; Jeffries, D.J.; Layton, G.T. Ability of yeast Ty-VLPs (virus-like particles) containing varicella-zoster virus (VZV)gE and assembly protein fragments to induce in vitro proliferation of human lymphocytes from VZV immune patients. J. Med. Virol. 1999, 59, 78–83. [Google Scholar] [CrossRef]
  86. ClinicalTrials.Gov. A Phase 2 Study to Evaluate JCXH-105, an srRNA-Based Herpes Zoster Vaccine. Available online: https://clinicaltrials.gov/study/NCT06581575?cond=JCXH-105&rank=1 (accessed on 4 December 2024).
  87. Eberhardson, M.; Hall, S.; Papp, K.A.; Sterling, T.M.; Stek, J.E.; Pang, L.; Zhao, Y.; Parrino, J.; Popmihajlov, Z. Safety and Immunogenicity of Inactivated Varicella-Zoster Virus Vaccine in Adults With Autoimmune Disease: A Phase 2, Randomized, Double-Blind, Placebo-Controlled Clinical Trial. Clin. Infect. Dis. 2017, 65, 1174–1182. [Google Scholar] [CrossRef]
  88. Parrino, J.; McNeil, S.A.; Lawrence, S.J.; Kimby, E.; Pagnoni, M.F.; Stek, J.E.; Zhao, Y.; Chan, I.S.; Kaplan, S.S. Safety and immunogenicity of inactivated varicella-zoster virus vaccine in adults with hematologic malignancies receiving treatment with anti-CD20 monoclonal antibodies. Vaccine 2017, 35, 1764–1769. [Google Scholar] [CrossRef]
  89. Mullane, K.M.; Morrison, V.A.; Camacho, L.H.; Arvin, A.; McNeil, S.A.; Durrand, J.; Campbell, B.; Su, S.C.; Chan, I.S.F.; Parrino, J.; et al. Safety and efficacy of inactivated varicella zoster virus vaccine in immunocompromised patients with malignancies: A two-arm, randomised, double-blind, phase 3 trial. Lancet Infect. Dis. 2019, 19, 1001–1012. [Google Scholar] [CrossRef]
  90. Beals, C.R.; Railkar, R.A.; Schaeffer, A.K.; Levin, Y.; Kochba, E.; Meyer, B.K.; Evans, R.K.; Sheldon, E.A.; Lasseter, K.; Lang, N.; et al. Immune response and reactogenicity of intradermal administration versus subcutaneous administration of varicella-zoster virus vaccine: An exploratory, randomised, partly blinded trial. Lancet Infect. Dis. 2016, 16, 915–922. [Google Scholar] [CrossRef] [PubMed]
  91. Nakamura-Nishimura, Y.; Shinkuma, S.; Miyagawa, F.; Haredy, A.; Gomi, Y.; Yamanishi, K.; Asada, H. Immunogenicity of varicella-zoster virus vaccine by different routes of administration: Comparable vaccination efficacy of one-fifth dose intradermal vaccination to conventional subcutaneous vaccination. J. Dermatol. Sci. 2022, 106, 86–92. [Google Scholar] [CrossRef]
  92. Yoshii, H.; Somboonthum, P.; Takahashi, M.; Yamanishi, K.; Mori, Y. Cloning of full length genome of varicella-zoster virus vaccine strain into a bacterial artificial chromosome and reconstitution of infectious virus. Vaccine 2007, 25, 5006–5012. [Google Scholar] [CrossRef]
  93. Gershon, A.A.; Brooks, D.; Stevenson, D.D.; Chin, W.K.; Oldstone, M.B.A.; Gershon, M.D. High Constitutive Interleukin 10 Level Interferes With the Immune Response to Varicella-Zoster Virus in Elderly Recipients of Live Attenuated Zoster Vaccine. J. Infect. Dis. 2019, 219, 1338–1346. [Google Scholar] [CrossRef]
  94. Zhang, S.; Zeng, Y.; Cui, L.; Zhang, Y.; Chen, T.; Xue, W.; Wang, H.; Liu, H.; Zhang, Y.; Chen, L.; et al. Immunogenicity and cellular response of a herpes zoster virus gEgI fusion protein adjuvanted with CpG-emulsion in mice. J. Nanobiotechnol. 2025, 23, 395. [Google Scholar] [CrossRef]
  95. Luan, N.; Cao, H.; Wang, Y.; Lin, K.; Liu, C. LNP-CpG ODN-adjuvanted varicella-zoster virus glycoprotein E induced comparable levels of immunity with Shingrix™ in VZV-primed mice. Virol. Sin. 2022, 37, 731–739. [Google Scholar] [CrossRef]
  96. Chen, T.; Sun, J.; Zhang, S.; Li, T.; Liu, L.; Xue, W.; Zhou, L.; Liang, S.; Yu, Z.; Zheng, Q.; et al. Truncated glycoprotein E of varicella-zoster virus is an ideal immunogen for Escherichia coli-based vaccine design. Sci. China Life Sci. 2023, 66, 743–753. [Google Scholar] [CrossRef] [PubMed]
  97. Wu, J.; Li, H.; Yuan, Y.; Wang, R.; Shi, T.; Li, Z.; Cui, Q.; Fu, S.; Nie, K.; Li, F.; et al. Truncated VZV gE Induces High-Titer Neutralizing Antibodies in Mice. Vaccines 2024, 12, 1139. [Google Scholar] [CrossRef]
  98. Cao, H.; Wang, Y.; Luan, N.; Lin, K.; Liu, C. Effects of Varicella-Zoster Virus Glycoprotein E Carboxyl-Terminal Mutation on mRNA Vaccine Efficacy. Vaccines 2021, 9, 1440. [Google Scholar] [CrossRef] [PubMed]
  99. Wang, Y.; Cao, H.; Lin, K.; Hu, J.; Luan, N.; Liu, C. Evaluation of the Immunological Efficacy of an LNP-mRNA Vaccine Prepared from Varicella Zoster Virus Glycoprotein gE with a Double-Mutated Carboxyl Terminus in Different Untranslated Regions in Mice. Vaccines 2023, 11, 1475. [Google Scholar] [CrossRef]
  100. Huang, L.; Zhang, S.; Zhao, T.; Cai, T.; Bu, L.; Di, Z.; Zhang, Y.; Yang, C.; Yang, Y.; Lin, A. Rational optimization of glycoprotein E (gE)-encoding mRNA for improved Varicella-zoster virus mRNA vaccine development. Emerg. Microbes Infect. 2024, 13, 2392661. [Google Scholar] [CrossRef] [PubMed]
  101. Wui, S.R.; Ko, A.; Ryu, J.I.; Sim, E.; Lim, S.J.; Park, S.A.; Kim, K.S.; Kim, H.; Youn, H.; Lee, N.G. The Effect of a TLR4 Agonist/Cationic Liposome Adjuvant on Varicella-Zoster Virus Glycoprotein E Vaccine Efficacy: Antigen Presentation, Uptake, and Delivery to Lymph Nodes. Pharmaceutics 2021, 13, 390. [Google Scholar] [CrossRef]
  102. Luan, N.; Cao, H.; Wang, Y.; Lin, K.; Liu, C. Ionizable Lipid Nanoparticles Enhanced the Synergistic Adjuvant Effect of CpG ODNs and QS21 in a Varicella Zoster Virus Glycoprotein E Subunit Vaccine. Pharmaceutics 2022, 14, 973. [Google Scholar] [CrossRef] [PubMed]
  103. Lee, C.; Kim, M.; Chun, J.; Kim, S.; Yoon, D.; Lee, H.; Bang, H.; Lee, H.J.; Park, H.; Kim, Y.B. Baculovirus Vector-Based Varicella-Zoster Virus Vaccine as a Promising Alternative with Enhanced Safety and Therapeutic Functions. Vaccines 2024, 12, 333. [Google Scholar] [CrossRef] [PubMed]
  104. Garcia-Valcarcel, M.; Fowler, W.J.; Harper, D.R.; Jeffries, D.J.; Layton, G.T. Induction of neutralizing antibody and T-cell responses to varicella-zoster virus (VZV) using Ty-virus-like particles carrying fragments of glycoprotein E (gE). Vaccine 1997, 15, 709–719. [Google Scholar] [CrossRef]
  105. Zhu, R.; Liu, J.; Chen, C.; Ye, X.; Xu, L.; Wang, W.; Zhao, Q.; Zhu, H.; Cheng, T.; Xia, N. A highly conserved epitope-vaccine candidate against varicella-zoster virus induces neutralizing antibodies in mice. Vaccine 2016, 34, 1589–1596. [Google Scholar] [CrossRef]
  106. Wang, H.; Zhang, S.; Xue, W.; Zeng, Y.; Liu, L.; Cui, L.; Liu, H.; Zhang, Y.; Chen, L.; Nie, M.; et al. Glycoprotein E-Displaying Nanoparticles Induce Robust Neutralizing Antibodies and T-Cell Response against Varicella Zoster Virus. Int. J. Mol. Sci. 2024, 25, 9872. [Google Scholar] [CrossRef]
  107. Levin, M.J.; Dahl, K.M.; Weinberg, A.; Giller, R.; Patel, A.; Krause, P.R. Development of resistance to acyclovir during chronic infection with the Oka vaccine strain of varicella-zoster virus, in an immunosuppressed child. J. Infect. Dis. 2003, 188, 954–959. [Google Scholar] [CrossRef]
  108. Levy, O.; Orange, J.S.; Hibberd, P.; Steinberg, S.; LaRussa, P.; Weinberg, A.; Wilson, S.B.; Shaulov, A.; Fleisher, G.; Geha, R.S.; et al. Disseminated varicella infection due to the vaccine strain of varicella-zoster virus, in a patient with a novel deficiency in natural killer T cells. J. Infect. Dis. 2003, 188, 948–953. [Google Scholar] [CrossRef]
  109. Leung, J.; Siegel, S.; Jones, J.F.; Schulte, C.; Blog, D.; Schmid, D.S.; Bialek, S.R.; Marin, M. Fatal varicella due to the vaccine-strain varicella-zoster virus. Hum. Vaccin. Immunother. 2014, 10, 146–149. [Google Scholar] [CrossRef]
  110. Bhalla, P.; Forrest, G.N.; Gershon, M.; Zhou, Y.; Chen, J.; LaRussa, P.; Steinberg, S.; Gershon, A.A. Disseminated, persistent, and fatal infection due to the vaccine strain of varicella-zoster virus in an adult following stem cell transplantation. Clin. Infect. Dis. 2015, 60, 1068–1074. [Google Scholar] [CrossRef] [PubMed]
  111. Stadtmauer, E.A.; Sullivan, K.M.; Marty, F.M.; Dadwal, S.S.; Papanicolaou, G.A.; Shea, T.C.; Mossad, S.B.; Andreadis, C.; Young, J.A.; Buadi, F.K.; et al. A phase 1/2 study of an adjuvanted varicella-zoster virus subunit vaccine in autologous hematopoietic cell transplant recipients. Blood 2014, 124, 2921–2929. [Google Scholar] [CrossRef] [PubMed]
  112. Atiyat, R.; Elias, S.; Kiwan, C.; Shaaban, H.S.; Slim, J. Varicella-Zoster Virus Reactivation in AIDS Patient After Pfizer-BioNTech COVID-19 Vaccine. Cureus 2021, 13, e20145. [Google Scholar] [CrossRef]
  113. Santovito, L.S.; Pinna, G. A case of reactivation of varicella-zoster virus after BNT162b2 vaccine second dose? Inflamm. Res. 2021, 70, 935–937. [Google Scholar] [CrossRef]
  114. Maldonado, M.D.; Romero-Aibar, J. The Pfizer-BNT162b2 mRNA-based vaccine against SARS-CoV-2 may be responsible for awakening the latency of herpes varicella-zoster virus. Brain Behav. Immun. Health 2021, 18, 100381. [Google Scholar] [CrossRef]
  115. Chong, C.H.; Liu, C.E.; Leong, Y.Y.; Liao, S.Y.; Lai, H.W.; Lee, Y.L. Seroprevalence of varicella-zoster virus antibody and immunogenicity of live attenuated varicella vaccine in healthcare workers in Taiwan. J. Microbiol. Immunol. Infect. 2023, 56, 274–281. [Google Scholar] [CrossRef]
  116. Irwin, M.R.; Levin, M.J.; Laudenslager, M.L.; Olmstead, R.; Lucko, A.; Lang, N.; Carrillo, C.; Stanley, H.A.; Caulfield, M.J.; Weinberg, A.; et al. Varicella zoster virus-specific immune responses to a herpes zoster vaccine in elderly recipients with major depression and the impact of antidepressant medications. Clin. Infect. Dis. 2013, 56, 1085–1093. [Google Scholar] [CrossRef]
  117. Perciani, C.T.; Sekhon, M.; Hundal, S.; Farah, B.; Ostrowski, M.A.; Anzala, A.O.; McKinnon, L.R.; Jaoko, W.; MacDonald, K.S. Live Attenuated Zoster Vaccine Boosts Varicella Zoster Virus (VZV)-Specific Humoral Responses Systemically and at the Cervicovaginal Mucosa of Kenyan VZV-Seropositive Women. J. Infect. Dis. 2018, 218, 1210–1218. [Google Scholar] [CrossRef] [PubMed]
  118. Pahar, B.; Gray, W.; Fahlberg, M.; Grasperge, B.; Hunter, M.; Das, A.; Mabee, C.; Aye, P.P.; Schiro, F.; Hensley, K.; et al. Recombinant Simian Varicella Virus-Simian Immunodeficiency Virus Vaccine Induces T and B Cell Functions and Provides Partial Protection against Repeated Mucosal SIV Challenges in Rhesus Macaques. Viruses 2022, 14, 2819. [Google Scholar] [CrossRef]
  119. Lowe, R.S.; Keller, P.M.; Keech, B.J.; Davison, A.J.; Whang, Y.; Morgan, A.J.; Kieff, E.; Ellis, R.W. Varicella-zoster virus as a live vector for the expression of foreign genes. Proc. Natl. Acad. Sci. USA 1987, 84, 3896–3900. [Google Scholar] [CrossRef]
Figure 1. Structure of VZV. The varicella-zoster virus (VZV) virion is nearly spherical, with a diameter of approximately 150–200 nm. It consists of a double-stranded DNA genome, a nucleocapsid, and a lipoprotein envelope. The nucleocapsid is composed of 162 capsomeres arranged in an icosahedral structure, while the envelope is densely studded with surface glycoproteins, which play crucial roles in viral entry and immune evasion. [Source: https://www.figdraw.com/].
Figure 1. Structure of VZV. The varicella-zoster virus (VZV) virion is nearly spherical, with a diameter of approximately 150–200 nm. It consists of a double-stranded DNA genome, a nucleocapsid, and a lipoprotein envelope. The nucleocapsid is composed of 162 capsomeres arranged in an icosahedral structure, while the envelope is densely studded with surface glycoproteins, which play crucial roles in viral entry and immune evasion. [Source: https://www.figdraw.com/].
Vaccines 13 00730 g001
Table 1. Approved and clinical varicella vaccines: monovalent and combination formulations.
Table 1. Approved and clinical varicella vaccines: monovalent and combination formulations.
Vaccine
Type
Development
Progress
Manufacturer/CountryVaccine
Name
Main Vaccine Components
Monovalent vaccineApprovedBiken/Japan [18]OkaVaxOka strain
Merck/Germany [19,20]VarivaxOka strain
GlaxoSmithKline (GSK)/UK [21]VarilrixOka strain
BCHT/China [22]\Oka strain
SINOVAC/China [23]\Oka strain
Shanghai Institute of Biological Products Co., Ltd./China [24]\Oka strain
GC Biopharma/Republic of Korea [25]SuduvaxMAV/06 strain
GC Biopharma/Republic of Korea [26]BARYCELAMAV/06 strain
Phase IIXiamen University (China)/Rutgers New Jersey Medical School (US) [27]VZV-7DOka strain
INDBOAOVAX/China [28]\Oka strain
Monovalent vaccineINDZhejiang Toyouvax Biopharming/China [29]\Oka strain
Combination vaccineApprovedMerck/Germany [30]ProQuadEdmonston strain (measles)
Jeryl Lynn™ strain (mumps)
Wistar RA 27/3 strain (rubella)
Oka/Merck strain (varicella)
GlaxoSmithKline (GSK)/UK [30]Priorix-TetraSchwarz strain (measles)
RIT 4385 strain (a Jeryl Lynn™ derivative for mumps)
Wistar RA 27/3 strain (rubella)
Oka/RIT strain (varicella)
Table 2. Comparison of approved and clinical HZ vaccines by different technological platforms.
Table 2. Comparison of approved and clinical HZ vaccines by different technological platforms.
Development
Progress
Manufacturer/CountryVaccine
Name
Technical
Approach
Main Vaccine ComponentsAdjuvant
ApprovedMerck/GermanyZostavaxZVLOka strain\
SK Bioscience/Republic of Korea [31]SkyZosterZVLOka strain\
BCHT/China [32]CanvarZVLOka strain\
GlaxoSmithKline (GSK)
/UK
ShingrixRZVgE/CHOAS01B
Phase IIIBeijing Luzhu Biotechnology Co., Ltd./China [33]LZ901RZVgE-Fc fusion protein/CHOAlum
Recbio/China [34]REC610RZVgE/CHOBFA01
Maxvax/China [35]\RZVgE/CHOcombined adjuvant MA105
Phase IIIMerck/Germany [36]V212inactivated vaccineInactivated VZV
Phase IIImmorna/China [37]JCXH-105mRNA vaccinesrRNAthe adjuvant effect of srRNA itself
Phase I/IIDynavax/US [48]z-1018RZVgE/CHOCpG 1018
Moderna/US [49]mRNA-1468mRNA vaccinegE mRNA-LNPLNP
Pfizer (US)/BioNTech (Germany) [50,51]VZV modRNAmRNA vaccinegE mRNA-LNPLNP
Phase IGenevax/China [52]GNW002RZVgE-Fc fusion protein/CHOAlum+CpG
Patronus Biotech/China [28]LYB004RZVgE-VLP/CHOA01
Grand Theravac Life Science Nanjing Co. Ltd./China [28]\RZVgE/CHOTVA01
EuBiologics/Republic of Korea [53]EuHZVRZVgEEcML+CoPoP
Innorna/China [54]IN001mRNA vaccinegE mRNA-LNPLNP
CanSinoBIO (China)/Vaccitech (UK) [55]CS-2032adenovirus vector vaccineChOx1 replication-deficient chimpanzee adenovirus vector encoding VZV-gE
INDGentize Biopharma (Nanjing)/China [28,56]\RZVgE/CHO
Rhegen/China [57]Freeze-dried shingles mRNA vaccinemRNA vaccinegE mRNA-LNPLNP
CSPC Pharmaceutical
Group Limited/China [28]
SYS6017mRNA vaccinegE mRNA-LNPLNP
SINOVAC/China [58]Freeze-dried shingles mRNA vaccinemRNA vaccinegE mRNA-LNPLNP
ZVL: live attenuated HZ vaccine; RZV: recombinant subunit HZ vaccine.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Liu, H.; Cui, L.; Zhang, S.; Wang, H.; Xue, W.; Li, H.; Zhang, Y.; Chen, L.; Gu, Y.; Li, T.; et al. Research Progress on Varicella-Zoster Virus Vaccines. Vaccines 2025, 13, 730. https://doi.org/10.3390/vaccines13070730

AMA Style

Liu H, Cui L, Zhang S, Wang H, Xue W, Li H, Zhang Y, Chen L, Gu Y, Li T, et al. Research Progress on Varicella-Zoster Virus Vaccines. Vaccines. 2025; 13(7):730. https://doi.org/10.3390/vaccines13070730

Chicago/Turabian Style

Liu, Hongjing, Lingyan Cui, Sibo Zhang, Hong Wang, Wenhui Xue, Hai Li, Yuyun Zhang, Lin Chen, Ying Gu, Tingting Li, and et al. 2025. "Research Progress on Varicella-Zoster Virus Vaccines" Vaccines 13, no. 7: 730. https://doi.org/10.3390/vaccines13070730

APA Style

Liu, H., Cui, L., Zhang, S., Wang, H., Xue, W., Li, H., Zhang, Y., Chen, L., Gu, Y., Li, T., Xia, N., & Li, S. (2025). Research Progress on Varicella-Zoster Virus Vaccines. Vaccines, 13(7), 730. https://doi.org/10.3390/vaccines13070730

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop