ABSTRACT
There are two licensed herpes zoster vaccines. One is a live vaccine (ZVL) based on an attenuated varicella-zoster virus (VZV). The other is a recombinant vaccine (RZV) based on the VZV glycoprotein E (gE) combined with AS01B, a multicomponent adjuvant system. RZV is superior to ZVL in efficacy, and differs from ZVL in that protection is not diminished by the age of the vaccinee and has not waned significantly during 4 years of follow-up. Immunologic studies demonstrated higher peak memory and persistence of T cell responses in RZV compared with ZVL recipients. RZV recipients also showed development and persistence of polyfunctional T cell responses. Taken together, we conclude that the immunologic data parallel and support the higher efficacy over time of RZV compared with ZVL.
Keywords: herpes zoster, older adults, immune responses, zoster vaccine live, recombinant zoster vaccine, adjuvants, vaccinology, viral
Pathogenesis of herpes zoster
Herpes zoster (HZ) has its origin during primary infection (varicella) with the varicella-zoster virus (VZV). VZV enters sensory and enteric neurons during varicella, either by retrograde movement in sensory nerves from skin and mucosal lesions or during the prolonged VZV viremia that is integral to varicella; or both.1 Since most adults in the world (>95%) have developed varicella, they have the complete VZV genome latent in 5% of their neurons (~5–10 copies/latently infected neuron).2,3 Latency is defined by the inability to recover the virus in tissue culture or visualize it by electron microscopy. A limited number of early transcripts and the viral proteins they encode have been previously detected in sensory neurons.2 The details about latent gene replication are being re-evaluated, with some data suggesting that a single latency-associated transcript is the essential feature of VZV latency.4
Of equal importance for HZ pathogenesis is that varicella results in the appearance of VZV-specific humoral and T cell-mediated immunity (CMI). These are readily detected shortly before the rash and peak in the month after rash healing.5 The VZV-CMI is essential for terminating varicella, and also for preventing HZ as described below.
Latent VZV reactivates intermittently to form infectious virions, as indicated by a variety of clinical and laboratory observations.6-10 The frequency and magnitude of reactivation events are unknown, but the reactivations typically remain subclinical because they are controlled by the VZV-specific immune responses that previously developed with varicella. However, while post-varicella antibody responses remain relatively unchanged lifelong, VZV-CMI responses decline with age.11 The correspondence of the age-related decline of these responses with the age-related increase in HZ frequency and severity, observed world-wide, is suggestive evidence for the essential role of VZV-CMI in preventing HZ.12 Additional evidence includes the following observations: neither varicella nor HZ frequency and severity are increased in disease states defined solely by defects in antibody synthesis;13,14 HZ frequency in immune compromised individuals correlates with VZV-CMI, but not with VZV antibody;15 protection of immune compromised patients with an investigational VZV-based vaccine correlated with VZV-CMI and not antibody;16 HZ continues to occur with high frequency after hematopoietic stem cell transplantation in spite of routinely providing passive immunization with γ-globulin products containing high titers of VZV antibodies;17 the severity of HZ is prevented by pre-existing robust memory CMI, but not by high titers of VZV antibodies.18
These protective VZV-CMI responses wane with increasing age.11 As a result, when latent VZV reactivates in sensory ganglia, and the local immune responses have become inadequate to prevent propagation of the infection, VZV infection will spread within the ganglia (which often causes neuropathic pain) and will spread antegrade to the skin to cause the characteristic painful HZ rash (called nociceptive pain) in the dermatome innervated by that ganglion. It then follows that the goal of a successful HZ vaccine is to restore VZV-specific CMI responses that decline during the aging process (or as a result of iatrogenic or disease-related immune compromise).
Live attenuated HZ vaccine (ZVL)
Clinical
The first licensed HZ vaccine consisted of the attenuated Oka (Merck) strain of VZV that also comprises the varicella vaccine. The dose administered was 14-fold greater than is used to prevent varicella. The utility of this HZ vaccine was determined in participants 50 to >80 years old and was characteristic of many vaccines for older people, namely its efficacy was progressively lower as the age of the vaccinee increased (70% in age 50–59; 64% in age 60–69; 38% age 70–79; 18% age >80 years)19-21 (Table 1). However, vaccine efficacy was greater against severity (67% average) of HZ and this protection against severe pain did not vary as much with age. ZVL efficacy against HZ waned significantly at 5–8 years after vaccination, but it had better persistence against severity of HZ.22 Nevertheless, ZVL was an important advance that annually prevented >100,000 severe cases of HZ during recent years. Effectiveness studies suggested that the age effect on efficacy was less and the persistence of efficacy against post-herpetic neuralgia was preserved longer than in the placebo-controlled pivotal trial.23,24 ZVL was safe, well tolerated, and required a single dose. However, it was contraindicated for immune compromised patients, who make up about 10% of HZ cases annually in the US.25
Table 1.
Characteristic | Zoster Vaccine Live | Recombinant Zoster Vaccine |
---|---|---|
Antigen | Live attenuated VZV (vOka) | Recombinant viral glycoprotein (gE) |
Doses Delivered | ~36 million | >3 million |
Adjuvant | None | AS01B |
Overall Efficacy | 51% | 91% |
Age Effect | Pronounced | Minimal |
Reactogenicity | Low | High |
Persistence of protection | 5 to 8 years | ≥4 years* |
Doses | One | Two (separated by 2 to 6 months) |
Protection with 1 dose | Yes | Limited – need 2 doses |
* Studies done up to 4 years.
Immunologic
In a substudy of the pivotal ZVL trial, we showed that VZV-specific antibody and two measures of T cell immunity (CMI) were stimulated by the vaccine.26 The kinetics of the immune responses were similar to those of efficacy of the vaccine – immunogenicity was greater in the first 6 months after administration and waned significantly within 3 years. Furthermore, the extent of the immune enhancement from vaccination was inversely related to the age of the vaccinee. This substudy also indicated that protection from HZ correlated with the magnitude of the VZV-specific immune responses that were present at the time of vaccination; at 6 weeks after vaccination; and in the last measurement available prior to the diagnosis of HZ. This was most evident with VZV-specific CMI, but was also observed with antibody responses. The additional large clinical trial of people 50–59 years of age showed that the 6-week antibody response was a correlate of protection.20
Subsequent studies showed that ZVL not only increased the magnitude, but also the breadth and polyfunctionality of VZV-specific CD4+ and CD8+ T cell responses.26-29 Epitopes recognized by CD8+ T cells that were increased by vaccination predominantly concentrated on ORF9 immediate early gene products, whereas CD4+ T cell responses targeted epitopes on multiple gene products including IE63, IE62, gB, ORF9 and gE in hierarchical order.27,28 New CD4+ and CD8+ T cell responses were at least partially maintained for up to 6 months27.
To identify the defects that might account for reduced efficacy as a function of increasing age, we compared T cell responses to ZVL in young and older adults and found both phenotypic and functional differences. Older adults displayed robust increases in VZV-specific senescent CD8+ CD57+ T cells after vaccination and lower numbers of polyfunctional CD4+ and CD8+ Th1 responses (IL2, IFNg and CD107a markers) compared with young adults.30 Our findings were in accordance with another study showing that older vaccinees lost T cell responses acquired after vaccination more rapidly than younger vaccinees.31 The T cell attrition was associated with a specific gene expression signature in cell cycle, cell division, DNA repair and mismatch repair modules. Additional transcriptomic and metabolomic signatures also differentiated young and older adults after vaccination, but these correlated best with peak antibody responses to ZVL.32 We and others also showed that older adults with high proportions of regulatory T cells had low Th1 responses to ZVL.33,34 Collectively, these studies show that immunologic changes characteristic of immune senescence profoundly affect T cell responses to ZVL.
Given the age effect on immunity induced by ZVL and waning of the initial response over time, attempts were made to enhance or restore VZV-specific immunity. Intradermal administration was dose-sparing (3-fold less required vs subcutaneous administration) and was more effective in stimulating VZV-specific CD4+ central and effector memory responses.35 A strategy to restore waning VZV-specific immunity utilized a second dose of ZVL, administered 10 years after an initial dose.36 This resulted in a significant increase in VZV-CMI measured 1 year later, achieving levels higher than those of age-matched controls immunized for the first time. However, at 3 years the boosted group maintained only a marginal advantage in IFNγ+IL2+ effector memory T cells over the first-time immunized individuals.37
Recombinant glycoprotein E (gE) adjuvanted HZ vaccine (RZV)
This vaccine is based on a single VZV glycoprotein which is abundantly expressed by VZV-infected cells and is the largest component in the viral envelope.38 gE is also a major target of antibodies and CD4+ T cell responses to VZV. This antigen is combined with an adjuvant system (AS01B) that contains 3-O-desacyl-4-monophosphoryl lipid A (MPL) and a triterpene plant product (Quillaja saponaria Molina, fraction 21 [QS21]). The adjuvant components are packaged in liposomes.
Clinical
Two placebo-controlled trials were completed, including >30,000 participants ≥50 years old, among which 16,500 were ≥70 years old. The trials documented an efficacy unique for older vaccinees, of 97% overall and 91% for those ≥70 and ≥80 years old39,40 (Table 1). This protection persisted at 85–89% for at least 4 years after vaccination in participants >70 years old. Follow-up is proceeding for an additional 6 years, although there will be no placebo comparator group. The strong adjuvant contained in RZV is associated with significant reactogenicity. Grade III (limits normal daily activity) injection site reactions occurred in 8.5% of vaccinees vs 0.2% of placebo recipients. Grade III systemic reactions occurred in 6–11% of vaccinees vs ~2% of placebo recipients. Older individuals were less likely to have grade III reactions; severity of reactions did not differ appreciably between the two doses. There was no safety signal for serious adverse events or possible immune mediated diseases. A trial in autologous hematopoietic stem cell transplant recipients demonstrated 68% efficacy against HZ and 89% efficacy against PHN.41
Immunologic
The remarkable protection offered to individuals of advanced age is largely due to the adjuvant system. Pre-clinical studies (using gE and hepatitis B antigens) and Phase I and II studies established that optimal immune responses, both antibody and CMI, required that all components of the adjuvant system and gE be co-localized without any significant interval between administration of the components.42-44 A synergistic effect was demonstrated between MPL and QS21, and two doses of RZV were required for optimal responses. The absence of an age effect on response to RZV was apparent from these early experiments.
The subsequent two pivotal clinical trials included an immunology substudy. This showed that gE antibody titers, measured by ELISA at one month after the second dose of RZV, increased in 98% of vaccinees.45 The mean increment in antibody titer was 39-fold, persisting as 8.3-fold higher than baseline at 3 years after vaccination. The decline in antibody titers was slightly greater in the oldest individuals. VZV-specific CMI was measured by flow cytometry to detect Th1 biomarker expression after ex vivo stimulation with gE overlapping peptide pools. Responses defined by CD4+ T cells expressing 2 or more markers among CD40L, IFNg, TNFa and IL-2 occurred in 93% of vaccinees. These declined to 57% at 3 years, with levels lower at all time points in people > 79 years old. The mean increase in VZV-CMI, which was 25-fold shortly after vaccination, fell to 7.9-fold at 3 years. As the interval after vaccination increased, the proportion of polyfunctional cells increased, such that at least 50% of VZV-specific CD4+ T cells had 3 or 4 biomarkers at 3 years after immunization. This was similar for all age groups vaccinated. Since 43% of vaccinees lost the RZV-induced CMI boost at 3 years after immunization, while they remained protected against HZ, this suggests that the measures of immunity in these trials were likely not correlates of protection.
The substudy confirmed the importance of the second dose of vaccine, which was administered 60 days after the first. A small clinical trial indicated that a 6 month interval between doses resulted in non-inferior immune responses. The kinetics of gE-specific immune responses were determined at 6 and 9 years after vaccination.46,47 Antibodies declined for 2 years and plateaued subsequently; CMI declined for 4 years before stabilizing. Both types of responses remained above pre-vaccination levels and there was little age effect observed. Because the substudy had a limited sample size (2900 for antibody assessment; 430 for CMI) and because of the paucity of HZ cases in the RZV recipients, it was not possible to define an immune surrogate of protection.
Immune compromised patients are an important target population for RZV. In addition to the efficacy mentioned above for autologous stem cell recipients, RZV induced gE antibodies in 67–72% and gE-specific CMI in 50–80% of patients with hematologic and solid malignancies receiving chemotherapy, and of renal transplant recipients. Immunogenicity was limited in allogeneic transplant recipients and in any vaccinees who received RZV during courses of chemotherapy.48-50
To understand the basis for the different immunogenicity and efficacy of the two zoster vaccines, we compared immune responses to the ZVL and RZV in adults 50 to 85 years old.51 gE-specific T cells were very low or undetectable before vaccination when analyzed by FluoroSpot and flow cytometry. Both ZVL and RZV increased gE-specific responses, but at 30 days after the last dose of each vaccine, corresponding to the peak memory response to vaccination, gE-specific CD4+ and CD8+ effector and memory T-cell responses were ≥10-fold higher in RZV compared with ZVL recipients. In addition, VZV-specific T cell memory responses were higher in RZV recipients, whereas CD8+ cytotoxic and effector T cell responses were higher in ZVL recipients. VZV- and gE-specific regulatory T cells expressing FOXP3 or immunologic checkpoints were also increased in RZV compared with ZVL recipients. At 1 year after vaccination, all gE-Th1 and VZV-memory responses remained higher in RZV compared to ZVL recipients. Mediation analyses showed that peak memory responses to gE or VZV were necessary for the persistence of Th1 responses to either vaccine. For example, the VZV-specific peak memory response in RZV recipients explained 73% of the total effect of RZV on persistence of its immunogenicity. Among effector responses, polyfunctional responses including IFNg, IL2 and TNFa were more common among RZV compared with ZVL recipients. The difference in the responses to RZV and ZVL is somewhat reminiscent of the difference between immune responses in older adults and young adults, with respect to memory, persistence and polyfunctionality, suggesting that RZV is able to neutralize the effects of immune senescence that are quite prominent on the responses to ZVL.
Studies of the AS01B Adjuvant System in several non-primate animal models showed that this adjuvant enhanced immune responses by increasing the number of activated antigen presenting cells (APC).52-54 While these studies did not utilize gE antigen, they are likely informative for understanding the RZV results, and some conclusions were confirmed with gE in non-human primates. Shortly after immunization the local innate response resulted in an influx to the draining lymph node (dLN) of conventional dendritic cells (cDC), as well as neutrophils and monocytes that carried most of the gE antigen. Likely, because of QS21 transported from the injection site, subcapsular sinus macrophages produced IL12 and IL18, which signaled resident NK and CD8+ unconventional virtual memory T cells to produce large amount of IFNg. This also activated CD11c+ cDC. Within 24 hours after vaccination there was a 200-fold increase in monocytes in the dLN and an 8.6-fold increase in cDC. A correlate in the blood compartment of these events in the dLN was an increase of IFNg and circulating polyfunctional CD4+ T cells.
Gene transcription profiling of the dLN indicated enrichment of cytokine transcription pathways, especially those involved in interferon-signaling, within 4–6 hours of vaccination. Numerous genes were transcriptionally active, including emergent genes that were represented only when both components of the adjuvant system were present. This confirmed the synergistic interaction of both components of AS01B.
Inactivated (non-live) zoster vaccine (ZVI)
Since ZVL, a live VZV vaccine, was contraindicated for immune compromised people, development of a non-live vaccine was undertaken before RZV was in clinical trials. ZVI was prepared by inactivation (heat or irradiation) of the attenuated VZV (Oka/Merck) used in the varicella vaccine and ZVL. Efficacy was demonstrated in autologous hematopoietic stem cell transplant recipients utilizing a dose prior to transplantation and 3 additional monthly doses after transplantation. At one year after vaccination 7/53 (13%) of vaccinees developed HZ versus 19/58 (33%) of placebo-recipients. Protection correlated with VZV-CMI, but not antibody.16,55 ZVI was not immunogenic in allogeneic stem cell transplant recipients.56,57 A phase III trial in autologous stem cell transplant recipients using the 4-dose schedule and γ-irradiated VZV, demonstrated an efficacy against HZ of 64%; against complications of HZ of 74%; and against PHN of 84%58 at a mean 2.3 year follow-up. In our opinion, because of the success of RZV, this potential competitor is unlikely to be further investigated.
Concluding remarks
The striking efficacy of RZV has brightened the horizon for vaccines for elderly individuals. RZV demonstrates that a single viral glycoprotein can stimulate robust and lasting protective responses, providing that an appropriate adjuvant shapes that response. That AS01B was able to overcome the limitations resulting from immune senescence suggests that appropriate adjuvantation might improve other vaccines needed for this population. It is important to note that although RZV is more reactogenic than other vaccines, especially with respect to systemic adverse events, over 3 million people have successfully taken both doses of the vaccine to date. Nevertheless, equally effective vaccines with less side effects, which could also be administered as a single dose are desirable.
Current and proposed studies of RZV should provide mechanistic insight that will be useful in designing future vaccines. The knowledge that can be gathered from peripheral blood studies is limited. Rather, a better understanding of the mechanism of action of RZV may be obtained from studying human draining lymph nodes and other tissues, as suggested by the animal models.
This new vaccine is likely to fill the unmet need of protecting immune compromised individuals from HZ. Many clinical trials to demonstrate safety and efficacy in such patients are in progress. These trials may also provide an immunologic correlate of protection. At least four studies of RZV in immune compromised individuals have been undertaken (renal transplant, hematologic malignancies, solid tumor with chemotherapy and autologous hematopoietic stem cell transplant). HZ break through will likely be more common in these immune compromised populations and, in conjunction with the immunologic studies performed in these trials, it may be possible to identify an immunologic correlate of protection.
Funding Statement
MJL and AW receive research grants from Merck, Sharp & Dohme and GlaxoSmithKline (all monies to University of Colorado Denver). MJL is a consultant for Merck, Sharp & Dohme and GlaxoSmithKline and holds the patent for ZVL together with Merck, Sharp & Dohme and University of Colorado Denver.
Disclosure of potential conflicts of interest
No potential conflicts of interest were disclosed.
References
- 1.Gershon AA, Gershon MD.. Pathogenesis and current approaches to control of varicella-zoster virus infections. Clin Microbiol Rev. 2013;26:728–43. doi: 10.1128/CMR.00052-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Mitchell BM, Bloom DC, Cohrs RJ, Gilden DH, Kennedy PG. Herpes simplex virus-1 and varicella-zoster virus latency in ganglia. J Neurovirol. 2003;9:194–204. doi: 10.1080/13550280390194000. [DOI] [PubMed] [Google Scholar]
- 3.Levin MJ, Cai GY, Manchak MD, Pizer LI. Varicella-zoster virus DNA in cells isolated from human trigeminal ganglia. J Virol. 2003;77:6979–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Depledge DP, Ouwendijk WJD, Sadaoka T, Braspenning SE, Mori Y, Cohrs RJ, Verjans GMGM, Breuer J. A spliced latency-associated VZV transcript maps antisense to the viral transactivator gene 61. Nat Commun. 2018;9:1167. doi: 10.1038/s41467-018-03569-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Weinberg A, Levin MJ. VZV T cell-mediated immunity. Curr Top Microbiol Immunol. 2010;342:341–57. doi: 10.1007/82_2010_31. [DOI] [PubMed] [Google Scholar]
- 6.Mehta SK, Cohrs RJ, Forghani B, Zerbe G, Gilden DH, Pierson DL. Stress-induced subclinical reactivation of varicella zoster virus in astronauts. J Med Virol. 2004;72:174–79. doi: 10.1002/jmv.10555. [DOI] [PubMed] [Google Scholar]
- 7.Ljungman P, Lonnqvist B, Gahrton G, Ringden O, Sundqvist VA, Wahren B. Clinical and subclinical reactivations of varicella-zoster virus in immunocompromised patients. J Infect Dis. 1986;153:840–47. [DOI] [PubMed] [Google Scholar]
- 8.Cinque P, Bossolasco S, Vago L, Fornara C, Lipari S, Racca S, Lazzarin A, Linde A. Varicella-zoster virus (VZV) DNA in cerebrospinal fluid of patients infected with human immunodeficiency virus: VZV disease of the central nervous system or subclinical reactivation of VZV infection? Clin Infect Dis. 1997;25:634–39. [DOI] [PubMed] [Google Scholar]
- 9.Schunemann S, Mainka C, Wolff MH. Subclinical reactivation of varicella-zoster virus in immunocompromised and immunocompetent individuals. Intervirology. 1998;41:98–102. doi: 10.1159/000024920. [DOI] [PubMed] [Google Scholar]
- 10.Wilson A, Sharp M, Koropchak CM, Ting SF, Arvin AM. Subclinical varicella-zoster virus viremia, herpes zoster, and T lymphocyte immunity to varicella-zoster viral antigens after bone marrow transplantation. J Infect Dis. 1992;165:119–26. [DOI] [PubMed] [Google Scholar]
- 11.Weinberg A, Lazar AA, Zerbe GO, Hayward AR, Chan ISF, Vessey R, Silber JL, MacGregor RR, Chan K, Gershon AA, et al. Influence of age and nature of primary infection on varicella-zoster virus-specific cell-mediated immune responses. J Infect Dis. 2010;201:1024–30. doi: 10.1086/651199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Kawai K, Gebremeskel BG, Acosta CJ. Systematic review of incidence and complications of herpes zoster: towards a global perspective. BMJ Open. 2014;4:e004833. doi: 10.1136/bmjopen-2014-004833. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Good RA, Zak SJ. Disturbances in gamma globulin synthesis as experiments of nature. Pediatrics. 1956;18:109–49. [PubMed] [Google Scholar]
- 14.Rosen FS, Janeway CA. The gamma globulins. 3. The antibody deficiency syndromes. N Engl J Med. 1966;275:709–15. doi: 10.1056/NEJM196609292751307. [DOI] [PubMed] [Google Scholar]
- 15.Arvin AM, Pollard RB, Rasmussen LE, Merigan TC. Cellular and humoral immunity in the pathogenesis of recurrent herpes viral infections in patients with lymphoma. J Clin Invest. 1980;65:869–78. doi: 10.1172/JCI109739. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Hata A, Asanuma H, Rinki M, Sharp M, Wong RM, Blume K, Arvin AM. Use of an inactivated varicella vaccine in recipients of hematopoietic-cell transplants. N Engl J Med. 2002;347:26–34. doi: 10.1056/NEJMoa013441. [DOI] [PubMed] [Google Scholar]
- 17.Boeckh M, Kim HW, Flowers ME, Meyers JD, Bowden RA. Long-term acyclovir for prevention of varicella zoster virus disease after allogeneic hematopoietic cell transplantation–a randomized double-blind placebo-controlled study. Blood. 2006;107:1800–05. doi: 10.1182/blood-2005-09-3624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Weinberg A, Zhang JH, Oxman MN, Johnson GR, Hayward AR, Caulfield MJ, Irwin MR, Clair J, Smith JG, Stanley H, et al. Varicella-zoster virus-specific immune responses to herpes zoster in elderly participants in a trial of a clinically effective zoster vaccine. J Infect Dis. 2009;200:1068–77. doi: 10.1086/605611. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Oxman MN, Levin MJ, Johnson GR, Schmader KE, Straus SE, Gelb LD, Arbeit RD, Simberkoff MS, Gershon AA, Davis LE, et al. A vaccine to prevent herpes zoster and postherpetic neuralgia in older adults. N Engl J Med. 2005;352:2271–84. doi: 10.1056/NEJMoa051016. [DOI] [PubMed] [Google Scholar]
- 20.Schmader KE, Levin MJ, Gnann JW Jr., McNeil SA, Vesikari T, Betts RF, Keay S, Stek JE, Bundick ND, Su S-C, et al. Efficacy, safety, and tolerability of herpes zoster vaccine in persons aged 50–59 years. Clin Infect Dis. 2012;54:922–28. doi: 10.1093/cid/cir970. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Merck & Co I Zostavax (zoster vaccine live) product insert. 2014:1–12. www.merck.com. online. Accessed 2018 Oct 30.
- 22.Morrison VA, Johnson GR, Schmader KE, Levin MJ, Zhang JH, Looney DJ, Betts R, Gelb L, Guatelli JC, Harbecke R, et al. Long-term persistence of zoster vaccine efficacy. Clin Infect Dis. 2015;60:900–09. doi: 10.1093/cid/ciu918. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Tseng HF, Harpaz R, Luo Y, Hales CM, Sy LS, Tartof SY, Bialek S, Hechter RC, Jacobsen SJ. Declining effectiveness of herpes zoster vaccine in adults aged ≥60 years. J Infect Dis. 2016;213:1872–75. doi: 10.1093/infdis/jiw047. [DOI] [PubMed] [Google Scholar]
- 24.Baxter R, Bartlett J, Fireman B, Marks M, Hansen J, Lewis E, Aukes L, Chen Y, Klein NP, Saddier P. Long-term effectiveness of the live zoster vaccine in preventing shingles: a Cohort study. Am J Epidemiol. 2018;187:161–69. doi: 10.1093/aje/kwx245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Yawn BP, Saddier P, Wollan PC, St Sauver JL, Kurland MJ, Sy LS. A population-based study of the incidence and complication rates of herpes zoster before zoster vaccine introduction. Mayo Clin Proc. 2007;82:1341–49. [DOI] [PubMed] [Google Scholar]
- 26.Levin MJ, Oxman MN, Zhang JH, Johnson GR, Stanley H, Hayward AR, Caulfield MJ, Irwin MR, Smith JG, Clair J, et al. Varicella-zoster virus-specific immune responses in elderly recipients of a herpes zoster vaccine. J Infect Dis. 2008;197:825–35. doi: 10.1086/528696. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Laing KJ, Russell RM, Dong L, Schmid DS, Stern M, Magaret A, Haas JG, Johnston C, Wald A, Koelle DM. Zoster vaccination increases the breadth of CD4+ T cells responsive to varicella zoster virus. J Infect Dis. 2015;212:1022–31. doi: 10.1093/infdis/jiv164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Sei JJ, Cox KS, Dubey SA, Antonello JM, Krah DL, Casimiro DR, Vora KA. Effector and central memory poly-functional CD4+ and CD8+ T cells are boosted upon ZOSTAVAX® vaccination. Front Immunol. 2015;6:553. doi: 10.3389/fimmu.2015.00553. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Qi Q, Cavanagh MM, Le Saux S, NamKoong H, Kim C, Turgano E, Helman LJ, Kastan MB, Knapp DW, Levin WJ, et al. Diversification of the antigen-specific T cell receptor repertoire after varicella zoster vaccination. Sci Transl Med. 2016;8:332ra46. doi: 10.1126/scitranslmed.aaf0746. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Weinberg A, Canniff J, Rouphael N, Mehta A, Mulligan M, Whattaker J, Levin MJ. VZV-specific cellular immune responses to the live attenuated zoster vaccine in young and older adults. J Immunol. 2017;199:604–12. doi: 10.4049/jimmunol.1700290. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Qi Q, Cavanagh MM, Le Saux S, Wagar LE, Mackey S, Hu J, Maecker H, Swan GE, Davis MM, Dekker CL, et al. Defective T memory cell differentiation after varicella zoster vaccination in older individuals. PLoS Pathog. 2016;12:e1005892. doi: 10.1371/journal.ppat.1005892. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Li S, Sullivan NL, Rouphael N, Yu T, Banton S, Maddur MS, McCausland M, Chiu C, Canniff J, Dubey S, et al. Metabolic phenotypes of response to vaccination in humans. Cell. 2017;169:862–77.e17. doi: 10.1016/j.cell.2017.04.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Lelic A, Verschoor CP, Lau VW, Parsons R, Evelegh C, Bowdish DM, Bramson JL, Loeb MB. Immunogenicity of varicella vaccine and immunologic predictors of response in a Cohort of elderly nursing home residents. J Infect Dis. 2016;214:1905–10. doi: 10.1093/infdis/jiw462. [DOI] [PubMed] [Google Scholar]
- 34.Weinberg A, Schmader K, Johnson M, Popmihajlov Z, Tovar-Salazar A, Caldas Y, Pang L, Cho A, Levin M. Immune senescence factors associated with the immunogenicity of a live attenuated zoster vaccine (ZV) in older adults. Open Forum Infect Dis. 2017;4:S413–S4. doi: 10.1093/ofid/ofx163.1036. [DOI] [Google Scholar]
- 35.Beals CR, Railkar RA, Schaeffer AK, Levin Y, Kochba E, Meyer BK, Evans RK, Sheldon EA, 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;16:915–22. doi: 10.1016/S1473-3099(16)00133-X. [DOI] [PubMed] [Google Scholar]
- 36.Levin MJ, Schmader KE, Pang L, Williams-Diaz A, Zerbe G, Canniff J, Johnson MJ, Caldas Y, Cho A, Lang N, et al. Cellular and humoral responses to a second dose of herpes zoster vaccine administered 10 years after the first dose among older adults. J Infect Dis. 2016;213:14–22. doi: 10.1093/infdis/jiv480. [DOI] [PubMed] [Google Scholar]
- 37.Weinberg A, Popmihajlov Z, Schmader KE, Johnson MJ, Caldas Y, Salazar AT, Canniff J, McCarson BJ, 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. 2018. doi: 10.1093/infdis/jiy514. [DOI] [PubMed] [Google Scholar]
- 38.Cunningham AL. The herpes zoster subunit vaccine. Expert Opin Biol Ther. 2016;16:265–71. doi: 10.1517/14712598.2016.1134481. [DOI] [PubMed] [Google Scholar]
- 39.Lal H, Cunningham AL, Godeaux O, Chlibek R, Diez-Domingo J, Hwang SJ, Levin MJ, McElhaney JE, Poder A, Puig-Barberà J, et al. Efficacy of an adjuvanted herpes zoster subunit vaccine in older adults. N Engl J Med. 2015;372:2087–96. doi: 10.1056/NEJMoa1501184. [DOI] [PubMed] [Google Scholar]
- 40.Cunningham AL, Lal H, Kovac M, Chlibek R, Hwang SJ, Diez-Domingo J, Godeaux O, Levin MJ, McElhaney JE, Puig-Barberà J, et al. Efficacy of the herpes zoster subunit vaccine in adults 70 years of age or older. N Engl J Med. 2016;375:1019–32. doi: 10.1056/NEJMoa1603800. [DOI] [PubMed] [Google Scholar]
- 41.de la Serna J, Campora L, Chandrasekar P, El Idrissi M, Gaidano G, Faqued ML, Oostvolgels L, Schwartz S, Sullivan K, Szer J, et al. LBA2: efficacy and safety of an adjuvanted herpes zoster subunit vaccine in autologous hematopoietic stem cell transplant recipients 18 years of age or older: first results of the phase 3 randomized, placebo-controlled ZOE-HSCT clinical trial. BMT Tandem Meetings; 2018; Salt Lake City, Utah. [Google Scholar]
- 42.Dendouga N, Fochesato M, Lockman L, Mossman S, Giannini SL. Cell-mediated immune responses to a varicella-zoster virus glycoprotein E vaccine using both a TLR agonist and QS21 in mice. Vaccine. 2012;30:3126–35. doi: 10.1016/j.vaccine.2012.01.088. [DOI] [PubMed] [Google Scholar]
- 43.Leroux-Roels G, Marchant A, Levy J, Van Damme P, Schwarz TF, Horsmans Y, Jilg W, Kremsner PG, Haelterman E, Clément F, et al. Impact of adjuvants on CD4(+) T cell and B cell responses to a protein antigen vaccine: results from a phase II, randomized, multicenter trial. Clin Immunol. 2016;169:16–27. doi: 10.1016/j.clim.2016.05.007. [DOI] [PubMed] [Google Scholar]
- 44.Chlibek R, Smetana J, Pauksens K, Rombo L, Van Den Hoek JA, Richardus JH, Plassmann G, Schwarz TF, Ledent E, Heineman TC. Safety and immunogenicity of three different formulations of an adjuvanted varicella-zoster virus subunit candidate vaccine in older adults: a phase II, randomized, controlled study. Vaccine. 2014;32:1745–53. doi: 10.1016/j.vaccine.2014.01.019. [DOI] [PubMed] [Google Scholar]
- 45.Cunningham AL, Levin MJ. Herpes Zoster Vaccines. J Infect Dis. 2018;218:S127–s33. doi: 10.1093/infdis/jiy382. [DOI] [PubMed] [Google Scholar]
- 46.Chlibek R, Pauksens K, Rombo L, van Rijckevorsel G, Richardus JH, Plassmann G, Schwarz TF, Catteau G, Lal H, Heineman TC. Long-term immunogenicity and safety of an investigational herpes zoster subunit vaccine in older adults. Vaccine. 2016;34:863–68. doi: 10.1016/j.vaccine.2015.09.073. [DOI] [PubMed] [Google Scholar]
- 47.Schwarz TF, Volpe S, Catteau G, Chlibek R, David MP, Richardus JH, 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;14:1370–77. doi: 10.1080/21645515.2018.1442162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Vink P. for the Zoster-028 Study Group Immumnogenicity and safety of a candidate subunit adjuvanted herpes zoster vaccine in adults with solid tumors vaccinated before or after immunosuppressive chemotherapy treatment: A phase II/III, randomized clinical trial. Poster 1349. ID Week. San Diego (CA), 2017. [Google Scholar]
- 49.Vink P. for the Zoster-041 Study Group Immumnogenicity and safety of a candidate subunit adjuvanted herpes zoster vaccine in adults post renal transplant: A phase III randomized clinical trial. Poster 1348. ID Week 2017. San Diego (CA), 2017. [Google Scholar]
- 50.Oostvogels L. Immunogenicity and safety of an adjuvanted herpes zoster subunit candidate vaccine in adults with hematologic malignancies: A phase III, randomized clinical trial. Abstract 1344. ID Week 2017. San Diego (CA), 2017. [Google Scholar]
- 51.Levin MJ, Kroehl ME, Johnson MJ, Hammes A, Reinhold D, Lang N, Weinberg A. Th1 memory differentiates recombinant from live herpes zoster vaccines. J Clin Invest. 2018;128:4429–40. doi: 10.1172/JCI121484. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Didierlaurent AM, Collignon C, Bourguignon P, Wouters S, Fierens K, Fochesato M, Dendouga N, Langlet C, Malissen B, Lambrecht BN, et al. Enhancement of adaptive immunity by the human vaccine adjuvant AS01 depends on activated dendritic cells. J Immunol. 2014;193:1920–30. doi: 10.4049/jimmunol.1400948. [DOI] [PubMed] [Google Scholar]
- 53.Neeland MR, Shi W, Collignon C, Taubenheim N, Meeusen EN, Didierlaurent AM, de Veer MJ. The lymphatic immune response induced by the adjuvant AS01: a comparison of intramuscular and subcutaneous immunization routes. J Immunol. 2016;197:2704–14. doi: 10.4049/jimmunol.1600817. [DOI] [PubMed] [Google Scholar]
- 54.Coccia M, Collignon C, Herve C, Chalon A, Welsby I, Detienne S, van Helden MJ, Dutta S, Genito CJ, Waters NC, et al. Cellular and molecular synergy in AS01-adjuvanted vaccines results in an early IFNgamma response promoting vaccine immunogenicity. NPJ vaccines. 2017;2:25. doi: 10.1038/s41541-017-0027-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Redman RL, Nader S, Zerboni L, Liu C, Wong RM, Brown BW, Arvin AM. Early reconstitution of immunity and decreased severity of herpes zoster in bone marrow transplant recipients immunized with inactivated varicella vaccine. J Infect Dis. 1997;176:578–85. [DOI] [PubMed] [Google Scholar]
- 56.Eberhardson M, Hall S, Papp KA, Sterling TM, Stek JE, 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;65:1174–82. doi: 10.1093/cid/cix484. [DOI] [PubMed] [Google Scholar]
- 57.Mullane KM, Winston DJ, Wertheim MS, Betts RF, Poretz DM, Camacho LH, Pergam SA, Mullane MR, Stek JE, Sterling TM, et al. Safety and immunogenicity of heat-treated zoster vaccine (ZVHT) in immunocompromised adults. J Infect Dis. 2013;208:1375–85. doi: 10.1093/infdis/jit344. [DOI] [PubMed] [Google Scholar]
- 58.Winston DJ, Mullane KM, Boeckh MJ, Cornerly OA, Hurtado K, Su S-C, Pang L, Zhao Y, Chan ISF, Stek JE, et al. A Phase III, double-blind, randomized, placebo-controlled, multicenter clinical trial to study the safety, tolerability, efficacy, and immunogenicity of inactivated Vzv vaccine (ZVIN) in recipients of autologous hematopoietic cell transplants (Auto-HCTs). Abstract #6 BMT Tandem Meetings Orlando (FL), 2017. [Google Scholar]