1,751
Views
10
CrossRef citations to date
0
Altmetric
Review

Human cytomegalovirus vaccine development: Immune responses to look into vaccine strategy

, , , &
Pages 292-303 | Received 07 Jun 2017, Accepted 08 Oct 2017, Published online: 01 Dec 2017

ABSTRACT

Human cytomegalovirus (HCMV) causes considerable morbidity and disability in high risk, immunocompromised populations including recipients of solid organ transplants, and fetuses whose immune systems are not yet mature. Vaccines aimed at ameliorating the severity of disease and preventing HCMV infection can be categorized into two main approaches of vaccine design, with one focusing on virus modification and the other on individual antigens. However, no candidates in either class have been successful in achieving durable and protective immunity. Recent studies on the natural immune response provide new insight into HCMV vaccine strategy. In particular, studies have demonstrated that the incorporation of a pentameric complex is necessary for a vaccine to generate the potent neutralizing antibodies often seen in seropositive individuals. This review summarizes recent findings in the development of HCMV vaccines and key considerations that should be taken into vaccine design based on improved understanding of natural HCMV immunity.

Introduction

Human cytomegalovirus (HCMV) is a prototypical β-herpesvirus that commonly infects humans but rarely causes disease in healthy subjects. HCMV infection can have serious consequences in fetuses and immunocompromised individualsCitation1–4 such as recipients of solid organ transplants (SOT) or hematopoietic stem cell transplant (HSCT) with immune suppression. Consequences of HCMV infection in transplant recipients can include pneumonitis, encephalitis, and hepatitis. HCMV infection remains a risk factor for transplantation-related mortality and graft failure.Citation5–7 Despite antiviral therapy, the incidence of HCMV infection is still high, ranging from 20–70% in the first year post transplantation.Citation4,8,9 In addition, pregnant women are susceptible to HCMV infection through contact with infected body fluids such as urine or saliva.Citation2,10,11 It is estimated that there are 20,000 to 40,000 children are born with congenital HCMV infection in the United States each year. Of these cases, there are about 100–200 deaths as a consequence of symptomatic infections, and about 4000–8000 children suffer permanent auditory, cognitive, and neurologic disabilities each year. These sequelae bring high economic burden on healthcare system.Citation12–14 Thus, an effective HCMV vaccine would address an unmet medical need. Efforts to develop a vaccine for the prevention of HCMV congenital infection has been assigned to the category of the highest priority by the Institute of Medicine since 1999.Citation15,16

Natural history and experimental intervention studies support the hypothesis that natural immunity acquired through HCMV infection in healthy subjects is protective against secondary infection or congenital transmission. From analysis of women with infants shedding virus, Adler and coworkers showed that over 47% HCMV seronegative mothers (9 out of 19 subjects) were infected with HCMV, while only 7% seropositive mothers (3 out 42) acquired new infection from their children.Citation17 Serological responses in this study were measured by enzyme immunoassay (EIA). A titer was defined as the highest dilution of serum that produced an OD of ≧ 0.1 (OD = 0.1 is lower limit of detection, LLOD). In addition, Fowler and coworkers showed that maternal seropositive status before conception was associated with reduced rate of congenital transmission.Citation18,19 To prevent congenital transmission in women with primary HCMV infection during pregnancy, therapy includes administration of high-titer anti-HCMV hyperimmune globulin (CMVIG), usually prepared from the plasma of seropositive donors with high titers of antibody against HCMV. Administration of CMVIG providing passive immunity has been evaluated as an intervention. In a seminal study by Negro et al, 6/37 women who received hyperimmune globulin during pregnancy had infants with congenital HCMV infection, compared with 19/47 women who did not receive the high-titer HCMV globulin.Citation20 CMVIG has also been studied in post-transplantation patients; prophylactic administration of CMVIG in kidney transplant patients showed that CMVIG has a beneficial effect on total survival, reduced HCMV disease, and HCMV-associated deaths.Citation7,21 HCMV natural immunity was determined by measuring HCMV-specific IgG antibodies in serological EIA assay. Besides CMVIG, early reconstitution of HCMV natural immunity was also significantly associated with a lower risk of HCMV infection in transplant patients. The strongest immune correlates with protection against HCMV viremia in HSCT recipients are reconstitution of HCMV-specific T cell memory responses, measured by lymphoproliferative assays.Citation22

Natural immunity demonstrates efficacy against HCMV infection and congenital transmission. The rational approach for HCMV vaccine design is to focus on the immune response seen in natural immunity, including potent neutralizing antibodies and HCMV-specific cellular responses such as HCMV-specific T cell memory responses. A number of experimental vaccines have been evaluated in clinical trials.Citation15 This review summarizes the current state of HCMV vaccine development () and discusses related quality and quantity of the immune responses by induced by vaccination.

Table 1. Summary of HCMV vaccine candidates.

HCMV viral particles and cell tropism in HCMV neutralization

HCMV is one of the largest and most complex enveloped viruses. It has a 230-kb double stranded linear DNA genome surrounded by a cellular lipid layer and a number of viral glycoproteins.Citation23,24 It is capable of encoding over 165 open reading frames (ORFs).Citation25–30 The roles of these components in infection and replication were not completely understood, due to the difficulty of viral gene expression and the lack of HCMV animal model.Citation31 HCMV can infect a broad range of cell types. Epithelial cells, endothelial cells, smooth muscle cells, and fibroblast cells are the cell types most commonly infected for virus replication.Citation23,32 The mature virions are ∼200 nm in diameter and contain over 50 viral proteins, including viral capsid proteins, tegument proteins, and envelope glycoproteins.Citation24 Many viral structural proteins, such as tegument proteins pp65, pp150, and pp50/52, are targets of host cellular immune responses.Citation33 The phospholipid envelope contains 6 encoded glycoproteins: gpUL55 (gB), gpUL73 (gN), gpUL74 (gO), gpUL75 (gH), UL100 (gM), and gpUL115 (gL). These glycoproteins are essential in virus entry, replication, and spreading.Citation24,34 The protein products of these six major glycoproteins form three associate complexes: gCI (gB), gCII (gM/gN), and gCIII (gH/gL/gO).Citation35 These complexes are highly conserved in the herpesvirus family. They are assembled for a variety of experimental vaccines being evaluated in clinical trials (). All major viral glycoprotein complexes are capable of eliciting neutralizing antibodies in naturally infected individuals or in vaccinated experimental animals. However, the majority of neutralizing antibody activity in naturally acquired immunity is directed at the pentameric gH complex consisting of gH, gL, pUL128, pUL130, and pUL131Citation36,37 (). Sha Ha and coworkers expressed and purified soluble gH/gL/pUL128–131 pentameric complex and gH/gL to better understand the neutralizing mechanism. They proved that the pentameric gH complex, but not gH/gL, presents dominant native neutralizing epitopes. Antibodies that bind only to the pentameric complex are expected to be more potent neutralizers than those that bind both the pentameric complex and gH/gL.Citation38

Table 2. Potential CMV proteins in vaccine design and their roles in immune response.

HCMV was isolated for the first time in 1956 in human fibroblasts.Citation39 In early vaccine development, researchers established two clinical isolates AD16940 and TowneCitation41 from fibroblasts. Since then, neutralizing antibodies have been evaluated in this cell system. These laboratory strains were developed as attenuated vaccines by serial passages cultured exclusively in fibroblasts. Scientists found that these vaccines failed to infect epithelial and endothelial cells, which are permissive for virus replication.Citation42,43 In a panel of antibodies from human sera with high neutralizing titers against the HCMV clinical isolates, a striking difference was observed. The neutralizing antibodies showed higher blocking ability in endothelial or epithelial cells than was measured in fibroblast cells. Hence, conventional determination of neutralizing activity in fibroblasts is misleading.Citation44,45 More recently, the UL131–128 locus of the HCMV genome has been shown to be indispensable for the infection of endothelial cells and epithelial cells; its mutation will cause loss of the pentameric gH complex.Citation46–48 This complex, which is a determinant for viral tropism to endothelial and epithelial cells, comprises the gH/gL scaffold and UL131, UL130, UL128 proteins.Citation49 When the UL131 mutation was restored in the AD169 parental virus, the revertant virus induced 10-fold higher neutralizing antibody titers than an attenuated AD169 virus. Further, the peak neutralizing titers post vaccination in rabbits and monkeys were 2-4-fold higher than the levels determined in HCMV seropositive subjects.Citation50 These results reveal the importance of the pentameric gH complex as a target of potent neutralizing antibodies in vaccinated animals or seropositive humans.Citation37

Defining the efficacy for HCMV vaccines in target populations

Despite significant progress in HCMV vaccine development, no product is yet under consideration for licensing. One challenge is how to define vaccine effects in clinical trials. Recently, a multidisciplinary meeting addressed priorities on the relevant and practical endpoints for assessing vaccine efficacy for prevention of HCMV disease in target populations.Citation51 The first endpoint is that a universal childhood HCMV vaccine should rapidly reduce congenital HCMV disease, as infected children are sources of viral transmission to seronegative and seropositive mothers.Citation51 The second is that clinical trials of HCMV vaccines in women should be evaluated for an endpoint of prevention of congenital HCMV infection. This is a more practical and acceptable endpoint for assessing vaccine effects on maternal-fetal transmission.Citation51 Recipients of allogeneic HSCT and SOT are at high risk for HCMV disease, pneumonitis, enteritis, retinitis, and viremia. HCMV serological status is associated with clinical outcomes including transplant-related mortality, re-infections, and failure of graft survival.Citation6 The third endpoint is that employment of vaccines should be of value in decreasing incidence and severity of HCMV disease, as well as the other complications of transplantation.Citation51 Such vaccines would be expected to be capable of eliciting humoral and cellular response after administration to either the transplant recipients or to the HSCT or solid organ donor prior to transplantation.

Natural immunity provides new insights for HCMV vaccine efficiency

HCMV natural immunity provides protection against the most devastating forms of congenital infection. Studies from Fowler and coworkers showed the presence of maternal antibody before conception provides substantial protection against congenital HCMV infection in newborns. Serologic assays were conducted in EIA using HCMV-specific IgG antibodies.Citation18,19 Following 197 newborns with congenital infection for 4.7 years, the study showed that 13% of infants whose mothers had primary infection during pregnancy had mental impairment, as compared with no symptoms for those whose mothers had recurrent HCMV infections.Citation19 There was a marked difference between the infants who acquired immunity vertically transmitted from their mothers and those who did not. Another line of evidence indicated that naturally acquired immunity resulted in a 69% reduction in the risk of congenital HCMV infection in future pregnancies.Citation18 Fowler and coworkers also concluded that protection against congenital HCMV infection was enhanced by longer time intervals between pregnancies.Citation52 An emphasis should be placed on HCMV vaccine design for which adaptive immune responses after vaccination mimic those components observed in the natural human immune response. The immune response may be imitated in both humoral and cellular immunity, including functional antibodies and T cells that have a significant impact on preventing HCMV infections.

Neutralizing antibodies in humoral response

Before clinical trials, HCMV vaccine strategies must be evaluated in animal models. One strategy that has been explored in the murine cytomegalovirus (MCMV) disease model is that of live attenuated vaccination. Studies of this strategy revealed that the attenuated mutants used as candidate vaccines were capable of inducing both humoral and cellular immune responses.Citation53 In addition, several investigations have examined the efficacy of subunit vaccinesCitation54,55 and DNA vaccinesCitation56 in MCMV models. A prime-boost immunization strategy used in the MCMV model supported further study of prime-boost approaches for CMV vaccination in other animal models, potentially, for human clinical trials.Citation57 The MCMV is similar to HCMV in virion structure, genome organization, gene expression, tissue tropism and latency. MCMV M123 (IE-1) is the homologous gene of HCMV UL123 (IE-1), and is expressed at the IE phase of MCMV replication as a non-structural protein pp89. M84 is expressed in E phase of replication as a non-structural protein of 65 kD (p65), and its homologous protein pp65 in HCMV is the main antigen for cytotoxic T lymphocyte (CTL) response against HCMV infection in humans. The m04 gene is only present in MCMV, and is expressed in E phase of virus replication as gp34 protein and plays an important role in immune escape of MCMV. M105 is expressed as DNA helicase of MCMV, and its HCMV homolog UL105 is highly conserved. M55 gene and its HCMV counterpart UL55 gene both encode the envelope glycoprotein B, the target antigen recognized by HCMV specific antibodies. The five DNA vaccines were constructed by Ze Chen's group. They found that the DNA vaccines, especially m04, M84 and IE-1, could effectively reduce the virus loads in salivary glands and spleens of mice, but they couldn't completely clear the residual virus. Immunization with M55 or M105 DNA at four doses offered mice only 62.5% survival rate after the lethal challenge.Citation58 Evidence from several animal models of CMV infection indicates that a variety of vaccine strategies are capable of inducing immune responses sufficient to protect against CMV-associated illness following viral challenge.Citation59 However, we still cannot study the virus–host interactions in any animal model for HCMV. HCMV infection is restricted to its natural host, and human pathologies caused by HCMV cannot be easily emulated in animal models.

Therefore, studies of naturally infected humans are the best basis for us to interpret and define the effectiveness of HCMV vaccine. But the hurdle is that we are limited to characterize the immune responses accurately by in vitro methods, for example investigation of epithelial or endothelial tropism deficiency of laboratory strains. To address this issue, a panel of antibodies from human sera exhibiting ≥ 128-fold higher neutralizing potency were measured with respect to their ability to block virus infection in endothelial and epithelial cells, but not tested in fibroblasts during early primary infection.Citation45 Results of this study suggest that induction of robust epithelial/endothelial specific neutralizing activities would be necessary for an effective HCMV vaccine.Citation60,61 In humans, the primary HCMV infection route is infection first of epithelial cells. Then endothelial cells and leukocytes disseminate virus into blood stream, resulting in viral infection of organ and tissue-specific cells.Citation62,63 As most HCMV infections are transmitted orally, epithelial specific neutralizing antibodies have the potential to block viral transmission by preventing virus entry into mucosal epithelial cells.Citation64 This may explain why Towne and gB/MF59 vaccines failed to induce high levels of neutralizing titers against virus epithelia entry to sustain durable protection in congenitally infected women.Citation65 On the other hand, virus or immunity vertical transmission between maternal and fetal is known to spread via placenta.Citation66,67 Studies showed that pregnant women developing antibodies with high avidity early after the onset of infection appeared to be at a lower risk of vertical transmission.Citation68 Furthermore, the children born to HCMV seropositive mothers were less likely to develop congenital HCMV disease than those born to mothers with primary HCMV infection.Citation69,70 To dissect the antibody response to HCMV glycoproteins in transmitter and non-transmitter pregnant women, 23 pregnant women were analyzed for the presence of neutralizing antibodies against different glycoproteins and glycoprotein complexes. The neutralizing antibodies were detected using ARPE-19 cells (human retinal pigment epithelial cells) and HELF cells (human lungs fibroblast cells) in a neutralization assay. This study demonstrated that neutralizing antibodies targeting the pentamer gH/gL/pUL128-131 complex were predominant, and that the early presence of neutralizing antibodies directed to multiple sites on the pentamer was associated with a reduced risk of HCMV vertical transmission.Citation71

Functional T cells in cellular response

HCMV infections are characterized by a dynamic, life-long interaction in which host immune responses, particularly of T cells, restrain viral replication and prevent disease but do not eliminate the virus or preclude transmission. From a study of cytokine flow cytometry screening, scientists found that 151 HCMV ORFs were immunogenic for CD4+ or CD8+ T cells, and that ORF immunogenicity was only modestly influenced by ORF expression, kinetics and function. They also reported that total HCMV-specific T cell responses in seropositive subjects were enormous, comprising on average 10% of both the CD4+ and CD8+ memory compartments in blood.Citation33 HCMV antigen-specific T cell responses involving both CD4+ and CD8+ T cells were further confirmed in HCMV seronegative vaccine recipients for clinical protective efficacy.Citation72 Studies in MCMV model revealed that the adoptive transfer of murine CMV specific CD8+ cytotoxic T cells to immunodeficient mice conferred protection from MCMV disease.Citation73,74 Further research supported this approach, showing that the recovery of CD4+ and CD8+ HCMV specific T cell responses in BMT (bone marrow transplant) patients who were HCMV seropositive was strongly correlated with protection from HCMV disease.Citation75–77 To investigate the therapeutic application of HCMV specific T cell lines, Hermann and colleagues adoptively transferred donor-derived HCMV-specific T cell lines into 8 stem cell transplant recipients lacking HCMV-specific T cell proliferation. They found that at a median of 11 days after transfer, T cells proliferation were detected in 6 of them, a significant increase of HCMV-specific CD4+ T cells in 5 patients. At a median of 13 days, 1.12 to 41 HCMV specific CD8+ T cells/μL blood were detected after transfer. In conclusion, their results demonstrated that anti-HCMV cellular therapy represents a therapeutic option in viremic patients after stem cell transplantation.Citation78 Together, HCMV-specific CD4+ and CD8+ T cells are the dominant compartments for HCMV natural infected response or adoptive derived response, also the golden measurement for vaccine efficiency.Citation79

Experiences in HMCV vaccine development and related immune responses

There is no experimental vaccine approach with imminent licensure in the pharmaceutical market. There are main reasons proposed for the failure to achieve the goal. First, the immune correlation for HCMV vaccine is not yet established due to deficiency of animal models. The ideal target protein capable of eliciting durable immune responses that closely mimic those seen in HCMV seropositive subjects is not fully characterized. This section will provide the strategy for development of vaccines in preclinical and clinical trials, and immune response induced by these vaccines (). In summary, there are two categories of vaccine candidates, one involves modification of attenuated or replication-defective virus to maintain safe efficiency in humans. This category is referred as modified virus vaccine (MVV). The second approach is to present targeted viral particles in the form of recombinant protein antigen through a DNA vector. This approach is referred as individual antigen vaccine (IAV).

Live attenuated HCMV vaccines

Early efforts to produce a HCMV vaccine were directed at developing a live, attenuated vaccine that presents all relevant antigens to the immune system in a conformation that is the same as what would be presented during natural HCMV infection. Some examples are the Towne and AD169 viruses, Towne/Toledo chimeric viruses, and dense body (DB) vaccines. The first candidate tested in human was developed from the AD169 laboratory strain by serial passages in fibroblasts, which were isolated from the adenoids of a child. This vaccine was found to be safe and well tolerated when administered in HCMV seronegative adults.Citation40,80,81 Subsequently, another Towne strain was established, derived from the virus isolated from the urine of an infant with congenital HCMV infection.Citation82,83 In the initial human trial, 11 HCMV seronegative adults were inoculated with Towne vaccine. None of them developed an augmented antibody response. In another human test, those inoculated developed only mild local reactions two weeks after vaccination. AD169 yielded similar results to those observed in the Towne trials.Citation84 The efficacy of Towne vaccine was evaluated in a series of studies in recipients of renal transplants.Citation85 Each study indicated that although Towne failed to prevent HCMV infection,Citation86,87 it did provide a degree of protection comparable with that conferred by natural infection.Citation88–90 Towne was also assessed in a placebo controlled study involving seronegative mothers who had children in daycare. It failed to protect women from HCMV infection transmitted from their children. In contrast, women with preexisting naturally infected immunity were protected from re-infection with a new strain being transmitted in the daycare.Citation17 Furthermore, it was shown that Towne was not capable of establishing persistent or latent infection in humans.Citation91 The inability of Towne vaccine to convey protection in high risk settings might be due to over-attenuation of non-invasive vaccines. To test this possibility, chimeric viruses between Towne and Toledo strains were constructed in the late 1990s and tested for safety in HCMV seropositive volunteers.Citation92,93 Researchers realized that the more important explanation is its genomic difference from clinical isolates. The failure of Towne to elicit neutralizing antibodies comparable to those induced by natural infection,Citation94–96 the fact that Towne is insufficiently immunogenic, is due to the lack of pentameric gH complex in its composition and the related to the loss of ability to infect endothelial or epithelial cells.

Subunit vaccines

Subunit vaccines are usually combined with an adjuvant to present defined viral antigens in the form of recombinant protein or delivered as a DNA vaccine or through a viral vector, then attempt to engender immune response recognized by specific immunogenic viral proteins to protect against infection or disease.Citation97 In clinical application, subunit vaccines focus primarily on gB, pp65, and IE1 as individual antigen vaccines.

Subunit glycoprotein B vaccine

The most fully characterized HCMV protein is the glycoprotein complex I (gCI) consisting of gB (UL55), which is based on Towne strain sequence and expressed in Chinese hamster ovary (CHO) cell as secreted protein. Reports assert that this protein is the dominant target of virus neutralizing antibody response in natural infection.Citation98–100 Formulated with an oil-in-water adjuvant, MF59, the vaccine was well tolerated and capable of inducing gB-specific antibodies in HCMV seronegative subjects.Citation101 After administration of the third dose of gB/MF59 vaccine, the levels of gB-specific antibodies and neutralizing activity exceeded those seen in HCMV seropositive controls.Citation102 The immunogenicity and safety of gB/MF59 vaccine has also been studied in a limited number of toddlers.Citation103 A Phase II trial was implemented with gB/MF59 vaccine in cohort of HCMV seronegative, postpartum women, who are at high risk of primary HCMV infection following delivery.Citation104 Unfortunately, these trials finally failed; the antibody response and neutralizing titers were transient, and returned to baseline within several months of vaccine immunization.Citation79 In contrast to Towne vaccine, gB/MF59 vaccine could boost gB-specific antibodies and T cells in HCMV seropositive volunteers,Citation105 but immune sera of the gB vaccine did not show any level of neutralizing titers against viral infection in epithelial cells.Citation65 To identify whether gB is the dominant antigen in the natural immunity to HCMV, German scientists isolated antibody repertoire against gB from different seropositive individuals in unbiased fashion. Their study revealed that most of the anti-gB antibodies produced during infection failed to neutralize cell-free virus, and most neutralizing antibodies were found to bind to epitopes that were not located within the previously characterized antigenic domains (AD) of gB.Citation106 This study will be useful tool to provide an immunogenic map of important proteins, showing that gB is not the prominent antigenic particle for neutralizing antibodies. Depletion of gB-specific antibodies in human polyclonal IgG would not affect its neutralizing activity in epithelial cells.Citation37 However, it is an essential viral protein that is involved in the early events of infection.

Subunit vectored vaccine

The purpose of the subunit vectored vaccine approach is to induce cellular and humoral response in a safe way. Protein pp65 (UL83) was defined as a significant target antigen for CD8+ class I major histocompatibility complex (MHC)-restricted HCMV-specific cytotoxic T lymphocytes (CTL). Protein pp65 was employed as a subunit vectored vaccine to develop HCMV-specific T cell response.Citation107,108 The TransVax vaccine consisted of plasmid DNAs encoding pp65 and gB formulated with poloxamer adjuvant. Clinical studies showed that TransVax was well-tolerated and significantly reduced viremia and reoccurrence in the HSCT setting. The reported safety and efficacy outcomes supported further development in a phase III trial, notwithstanding a lack of significant reduction in the use of HCMV-specific antiviral therapy compared with placebo in this phase II trial.Citation109 HCMV IE1 gene is also an important target of the CD8+ T cell response to HCMV infection.Citation110 A trivalent DNA vaccine targeting gB, pp65, and IE1 was under evaluation in a Phase I trial involving a total of forty healthy adult subjects.Citation111,112

Poxvirus vectors and alphavirus vectors have both been studied for their potential as HCMV vaccine candidates to stimulate CTL responses. Another formulation of gB vaccine evaluated in clinical trial is based on canarypox vector ALVAC, an attenuated poxvirus that replicates abortively in mammalian cells. ALVAC-gB vaccine was modestly immunogenic. The vaccine was usually boosted with a live, attenuated Towne vaccine or gB/MF59, being otherwise unable to develop binding and neutralizing antibody titers comparable to those observed in naturally seropositive subjects.Citation113,114 Although these concomitant vaccine approaches induced high titer antibodies and lymphoproliferative response, it appears to be more like an augmented gB-specific response, with no benefit detected for prime-boost strategy.

Novel strategy facing the immunological challenge

Advances in studies of natural immunity inform development of strategy and definition for future vaccine candidates in the clinic. An effective HCMV vaccine should be designed to induce broad neutralizing antibodies capable of protecting a variety of cell types and balanced CD4+ and CD8+ T cell responses, ideally comparable to those seen in naturally infected individuals. There are two main points that should be taken into consideration. First, the immunogenic target for both arms of adaptive immune responses should be incorporated into vaccine formation. Second, we should choose an appropriate way to establish and express vaccine candidates challenging modalities for development.

Primary target in HCMV vaccine design

Prior knowledge of neutralizing antibodies of natural immunity to HCMV are limited to those targeting gB, gM/gN, and gH/gL/gO complexes, which are essential for virus entry in fibroblast cells.Citation49 More recent studies revealed that gB and the gH/gL dimer are necessary for fibroblast entry. In contrast, entry into endothelial or epithelial cells requires three additional viral proteins, UL128, UL130, and UL131, which interact with gH/gL to form a pentameric complex.Citation43,47,48,60,115 Researchers measured the neutralizing activities of HCMV seropositive sera during pregnancy on fibroblasts and epithelial/endothelial cells. Results showed that the neutralizing antibody response measured in epithelial or endothelial cells was potent, occurred very early, and was directed mostly against combinations of two or three gene products of the UL131-128 locus. On the contrary, neutralizing antibody measured in fibroblasts appeared late, was relatively weak, and was directed against gH and gB.Citation116

To identify the neutralizing component of CMVIG, serial depletions of CMVIG on cell-surface-expressed HCMV antigens as well as purified antigens was studied. The results indicated that major neutralizing antibodies for epithelial cell entry in CMVIG were directed against the gH/gL/pUL128/pUL130/pUL131 complex. The gH/gL antibodies in CMVIG were shown to have a dominant role in inhibition of viral entry into fibroblasts; anti-gB antibodies played little role in CMVIG neutralization.Citation37 This conclusion was also supported by the clinical data associated with the Towne vaccine and the gB/MF59subunit vaccine, which induced epithelial entry-specific neutralizing activities that were on average 28-fold (Towne) and 15-fold (gB/MF59) lower than those observed in natural infection. These results suggest that HCMV vaccine efficacy may be enhanced by induction of epithelial entry-specific neutralizing antibodies.Citation65

Subsequent studies further proved that the most potent neutralizing epitopes were target the pentameric gH complex. Macagno and coworkers isolated 27 human monoclonal antibodies with extraordinary high potency in neutralization from immortalized memory B cells of HCMV-immune donors. Only one of them recognized conserved epitope of UL128, all others recognized conformational epitopes that required expression of two or more proteins of the gH/gL/pUL128-131 complex.Citation117 In another report, Potzcsch and coworkers isolated over 600 gB-specific antibodies from six human donors and found the vast majority of these antibodies had no neutralizing activity.Citation106 Tong-ming Fu and his group isolated a panel of 45 monoclonal antibodies from a rabbit immunized with an experimental vaccine virus in which the expression of the pentameric gH complex was restored. Their studies revealed that over one-half (25 of 45) of the antibodies had high titer neutralizing activity. These results clearly suggest that the pentameric gH complex is the primary target for potent neutralization in humoral immunity to natural infection.Citation118 Recently, this group developed a whole-virus vaccine candidate from the live attenuated AD169 strain, with genetic modifications to improve its immunogenicity and attenuation. This vaccine restored the expression of the pentameric gH/gL/pUL128-131 protein complex, a major target for neutralizing antibodies in natural immunity.Citation119

Alternative expression options in vaccine design

HCMV vaccine candidates are generally grouped into two categories. In one category are modified virus vaccines, which were designed as live attenuated viruses, include Towne and AD169 viruses and Towne/Toledo chimeric viruses. In the other category are dense body (DB) vaccines consisting of defined quantities of noninfectious virus particles. Individual antigen vaccines are designed to deliver immunogenic antigen to induce durable immune responses. Delivery is achieved through a viral or DNA vector such as is used for subunit vaccines gB/MF59, pp65, and IE1. Modified virus vaccines must be safe for human use,Citation60 and a unique manufacturing process would have to be developed. In contrast to modified virus vaccines, individual antigen vaccines are developed based on optimized antigens which potentially combine the immunogenicity of a killed vaccine with safety or replication-defective viral vectors (e.g., pox, adenovirus, alphavirus, and others) expressing subunits or multi-subunit complexes. Because of the defined target, the dose potency and quality could be detected in a straightforward manner. The correlation between immunogenicity, protection, and vaccine would be easier to define and standardize. The key remaining question is whether the antigen composition in individual antigen vaccines is sufficient for protection and prevention.

As previously described, pentameric gH complex is unique for epithelial or endothelial cell entry. It also has a potent ability to generate neutralizing antibodies either in the form of virus vaccine or in live virus during natural infection. Glycoprotein gB is required for virus entry, cell-cell spread, and driving fusion of the viral envelope with the target cell membrane.Citation120 The answer to whether more immunogenic forms of virus are effective for vaccine development remains elusive. Considering this, the modified virus vaccine approach is preferred, as it targets both arms of the adaptive immune response. The incorporation of the pentameric gH complex into vaccines will improve the vaccine efficacy, while other forms of the virus could be retained to maintain virus integrity and to be able to mimic the immune response observed in naturally infected individuals. This approach was achieved using an experimental vaccine, an epithelial tropism revertant virus of AD169 origin in which the pentameric gH complex was restored.Citation50,119 Use of this experimental vaccine in an animal model elicited high levels of neutralizing antibodies titers, significantly better than those achieved by attenuated AD169/Towne vaccine and recombinant gB vaccines. The neutralizing titers after revertant virus vaccine immunization were comparable to those seen in HCMV-seropositive human sera.Citation118

Lessons from herpesvirus family vaccines

HCMV is a member of the herpesvirus family. The development of other herpesvirus family vaccines provides us clues suggesting that the vaccine design should be based on improved understanding of natural immunity. We may focus determination of whether a candidate vaccine is capable of eliciting neutralizing antibodies for disease prevention and treatment and what antigens are important for eliciting immune responses from protective antibodies and diverse and polyfunctional T cells. Based on this strategy, a promising herpes zoster subunit vaccine, Shingrix, was developed, with vaccine efficacy against Herpes Zoster (HZ) 97.2% for all age groups,Citation121 which consists of varicella zoster virus (VZV) glycoprotein E and AS01B adjuvant was assessed in Phase III. Of noted, the combination of Shingrix antigen containing T and B cell epitopes and an adjuvant was able to stimulate both VZV-specific antibody and T cells and restore age-related decline in T cell immunity. Besides Shingrix, the most studied subunit herpesvirus vaccines are herpes simplex virus type 2 (HSV-2) vaccines. However, to date, no HSV-2 vaccines have conferred protective immune responses against infections in clinical trials.Citation122 The combination of HSV-2 gD and gB with MF59, Chiron vaccine, was shown to be immunogenic in humans, but it was ineffective in the treatment of recurrent genital herpes.Citation123 Both antibodies and T cells can contribute to control of viral replication in the genital tract. This study suggests that rather than focusing on induction of only antibodies or T cell responses, vaccines that lead to combined immune responses against HSV-2 may have increased efficacy. Hence, live attenuated vaccines may have a distinct advantage over subunit and inactivated vaccines, primarily because replication of the pathogen allows for the entire repertoire of pathogen-specific antigen expression. The licensed vaccines VarivaxCitation124 and ZostavaxCitation125 are classic examples. Varivax is a vaccine for varicella and Zostavax for zoster. These live attenuated vaccines have been used extensively in prophylactic and therapeutic approaches to combat primary and recurrent zoster infection. For HCMV vaccine, the whole-virus vaccine candidate from the live attenuated AD169 strain, with genetic modifications to improve its immunogenicity and attenuation, might also be a more practical vaccine design approach.Citation119

Conclusions

Although several experimental HCMV vaccines have advanced to clinical evaluation (), a major barrier remains in the progress of vaccine development: the optimal HCMV vaccine strategy must be suitable for the population of potential patients. However, better understanding of the correlates of protective immunity to natural HCMV infection has been achieved recently. In particular, the correlates of protective immunity involved in epithelial/endothelial cell entry have been elucidated as areas for future vaccine strategy. The subunit expression of the gH/gL/pUL128-131 proteins would be exploited in vaccine design, which also merits consideration for vaccine efficiency evaluation. The studies on preclinical and clinical analysis of vaccine candidates will substantially accelerate the pace of successful vaccine emergence and solve this public health urgency.

Abbreviations

AD=

antigenic domains

ARPE-19 cells=

human retinal pigment epithelial cells

BMT=

bone marrow transplant

CHO=

Chinese hamster ovary

CMVIG=

HCMV hyperimmune globulin

CTL=

cytotoxic T lymphocytes

DB=

dense body

EIA=

enzyme immunoassay

HCMV=

human cytomegalovirus

HELF=

cellshuman lungs fibroblast cells

HSCT=

hematopoietic stem cell transplant

HSV-2=

herpes simplex virus type 2

HZ=

herpes zoster

IAV=

individual antigen vaccine

LLOD=

lower limit of detection

MCMV=

murine cytomegalovirus

MHC=

major histocompatibility complex

MVV=

modified virus vaccine

ORFs=

open reading frames

R/D+=

a HCMV seronegative recipient with SOT from a seropositive donor

SOT=

solid organ transplants

VEE=

Venezuelan equine encephalitis virus

VRP=

alphavirus VEE replicon particles

VZV=

varicella zoster virus

Disclosure of potential conflicts of interest

No potential conflicts of interest were disclosed.

Acknowledgments

We thank Dr. Georgina Salazar at the University of Texas Health Science Center at Houston for her careful and critical reading of the manuscript.

Funding

This work was supported in part by the Welch Foundation (Grant AU-0042-20030616 to Z.A.) and National Natural Science Foundation of China Grant 31670927 (W.L.).

References

  • Boeckh M, Geballe AP. Cytomegalovirus: pathogen, paradigm, and puzzle. J Clin Invest. 2011;121:1673–80. doi:10.1172/JCI45449. PMID:21659716.
  • Kenneson A, Cannon MJ. Review and meta-analysis of the epidemiology of congenital cytomegalovirus (CMV) infection. Rev Med Virol. 2007;17:253–76. doi:10.1002/rmv.535. PMID:17579921.
  • La Torre R, Nigro G, Mazzocco M, Best AM, Adler SP. Placental Enlargement in Women with Primary Maternal Cytomegalovirus Infection Is Associated with Fetal and Neonatal Disease. Clin Infect Dis. 2006;43:994–1000. doi:10.1086/507634. PMID:16983610.
  • Beam E, Razonable R. Cytomegalovirus in Solid Organ Transplantation: Epidemiology, Prevention, and Treatment. Curr Infect Dis Rep. 2012;14:633–41. doi:10.1007/s11908-012-0292-2. PMID:22992839.
  • Kotton CN. Management of cytomegalovirus infection in solid organ transplantation. Nat Rev Nephrol. 2010;6:711–21. doi:10.1038/nrneph.2010.141. PMID:20978468.
  • Boeckh M, Ljungman P. How we treat cytomegalovirus in hematopoietic cell transplant recipients. Blood. 2009;113:5711–9. doi:10.1182/blood-2008-10-143560. PMID:19299333.
  • Opelz G, Dohler B, Ruhenstroth A. Cytomegalovirus prophylaxis and graft outcome in solid organ transplantation: a collaborative transplant study report. Am J Transplant. 2004;4:928–36. doi:10.1111/j.1600-6143.2004.00451.x. PMID:15147427.
  • Kotton CN. Management of cytomegalovirus infection in solid organ transplantation. Nat Rev Nephrol. 2010;6:711–21. doi:10.1038/nrneph.2010.141. PMID:20978468.
  • Ariza-Heredia EJ, Nesher L, Chemaly RF. Cytomegalovirus diseases after hematopoietic stem cell transplantation: A mini-review. Cancer Lett. 2014;342:1–8. doi:10.1016/j.canlet.2013.09.004. PMID:24041869.
  • Revello MG, Gerna G. Diagnosis and Management of Human Cytomegalovirus Infection in the Mother, Fetus, and Newborn Infant. Clin Microbiol Rev. 2002;15:680–715. doi:10.1128/CMR.15.4.680-715.2002. PMID:12364375.
  • Bodeus M, Kabamba-Mukadi B, Zech F, Hubinont C, Bernard P, Goubau P. Human cytomegalovirus in utero transmission: follow-up of 524 maternal seroconversions. J Clin Virol. 2010;47:201–2. doi:10.1016/j.jcv.2009.11.009. PMID:20006542.
  • Pass RF, Zhang C, Evans A, Simpson T, Andrews W, Huang ML, Corey L, Hill J, Davis E, Flanigan C, et al. Vaccine prevention of maternal cytomegalovirus infection. N Engl J Med. 2009;360:1191–9. doi:10.1056/NEJMoa0804749. PMID:19297572.
  • Dollard SC, Grosse SD, Ross DS. New estimates of the prevalence of neurological and sensory sequelae and mortality associated with congenital cytomegalovirus infection. Rev Med Virol. 2007;17:355–63. doi:10.1002/rmv.544. PMID:17542052.
  • Ross SA, Boppana SB. Congenital cytomegalovirus infection: outcome and diagnosis. Semin Pediatr Infect Dis. 2005;16:44–9. doi:10.1053/j.spid.2004.09.011. PMID:15685149.
  • Schleiss MR. Cytomegalovirus Vaccine Development. Curr Top Microbiol Immunol. 2008;325:361–82. PMID:18637516.
  • Stratton KR, Durch JS, Lawrence RS. Vaccines for the 21st Century: A Tool for Decisionmaking. Inst Med. 2000. Washington (DC): National Academies Press (US). doi:10.17226/5501.
  • Adler SP, Starr SE, Plotkin SA, Hempfling SH, Buis J, Manning ML, Manning ML, Best AM. Immunity induced by primary human cytomegalovirus infection protects against secondary infection among women of childbearing age. J Infect Dis. 1995;171:26–32. doi:10.1093/infdis/171.1.26. PMID:7798679.
  • Fowler KB, Stagno S, Pass RF. Maternal immunity and prevention of congenital cytomegalovirus infection. JAMA. 2003;289:1008–11. doi:10.1001/jama.289.8.1008. PMID:12597753.
  • Fowler KB, Stagno S, Pass RF, Britt WJ, Boll TJ, Alford CA. The outcome of congenital cytomegalovirus infection in relation to maternal antibody status. N Engl J Med. 1992;326:663–7. doi:10.1056/NEJM199203053261003. PMID:1310525.
  • Nigro G, Adler SP, La Torre R, Best AM. Congenital Cytomegalovirus Collaborating Group. Passive immunization during pregnancy for congenital cytomegalovirus infection. N Engl J Med. 2005;353:1350–62. doi:10.1056/NEJMoa043337. PMID:16192480.
  • Bonaros N, Mayer B, Schachner T, Laufer G, Kocher A. CMV-hyperimmune globulin for preventing cytomegalovirus infection and disease in solid organ transplant recipients: a meta-analysis. Clin Transplant. 2008;22:89–97. PMID:18217909.
  • Barron MA, Gao D, Springer KL, Patterson JA, Brunvand MW, McSweeney PA, Zeng C, Barón AE, Weinberg A. Relationship of reconstituted adaptive and innate cytomegalovirus (CMV)-specific immune responses with CMV viremia in hematopoietic stem cell transplant recipients. Clin Infect Dis. 2009;49:1777–83. doi:10.1086/648423. PMID:19911937.
  • Landolfo S, Gariglio M, Gribaudo G, Lembo D. The human cytomegalovirus. Pharmacol Therapeutics. 2003;98:269–97. doi:10.1016/S0163-7258(03)00034-2.
  • Varnum SM, Streblow DN, Monroe ME, Smith P, Auberry KJ, Pasa-Tolic L, Wang D, Camp DG 2nd, Rodland K, Wiley S, et al. Identification of proteins in human cytomegalovirus (HCMV) particles: the HCMV proteome. J Virol. 2004;78:10960–6. doi:10.1128/JVI.78.20.10960-10966.2004. PMID:15452216.
  • Bankier AT, Beck S, Bohni R, Brown CM, Cerny R, Chee MS, Hutchison CA 3rd, Kouzarides T, Martignetti JA, Preddie E, et al. The DNA sequence of the human cytomegalovirus genome. Mitochondrial DNA. 1991;2:1–11.
  • Cha TA, Tom E, Kemble GW, Duke GM, Mocarski ES, Spaete RR. Human cytomegalovirus clinical isolates carry at least 19 genes not found in laboratory strains. J Virol. 1996;70:78–83. PMID:8523595.
  • Chee MS, Bankier AT, Beck S, Bohni R, Brown CM, Cerny R, Horsnell T, Hutchison CA 3rd, Kouzarides T, Martignetti JA, et al. Analysis of the protein-coding content of the sequence of human cytomegalovirus strain AD169. Curr Top Microbiol Immunol. 1990;154:125–69. PMID:2161319.
  • Dolan A, Cunningham C, Hector RD, Hassan-Walker AF, Lee L, Addison C, Dargan DJ, McGeoch DJ, Gatherer D, Emery VC, et al. Genetic content of wild-type human cytomegalovirus. J Gen Virol. 2004;85:1301–12. doi:10.1099/vir.0.79888-0. PMID:15105547.
  • Gatherer D, Seirafian S, Cunningham C, Holton M, Dargan DJ, Baluchova K, Hector RD, Galbraith J, Herzyk P, Wilkinson GW, et al. High-resolution human cytomegalovirus transcriptome. Proc Natl Acad Sci U S A. 2011;108:19755–60. doi:10.1073/pnas.1115861108. PMID:22109557.
  • Stern-Ginossar N, Weisburd B, Michalski A, Le VT, Hein MY, Huang SX, Ma M, Shen B, Qian SB, Hengel H, et al. Decoding human cytomegalovirus. Science. 2012;338:1088–93. doi:10.1126/science.1227919. PMID:23180859.
  • Dunn W, Chou C, Li H, Hai R, Patterson D, Stolc V, Zhu H, Liu F. Functional profiling of a human cytomegalovirus genome. Proc Natl Acad Sci U S A. 2003;100:14223–8. doi:10.1073/pnas.2334032100. PMID:14623981.
  • Sinzger C, Digel M, Jahn G. Cytomegalovirus cell tropism. Curr Top Microbiol Immun. 2008;325:63–83.
  • Sylwester AW, Mitchell BL, Edgar JB, Taormina C, Pelte C, Ruchti F, Sleath PR, Grabstein KH, Hosken NA, Kern F, et al. Broadly targeted human cytomegalovirus-specific CD4+ and CD8+ T cells dominate the memory compartments of exposed subjects. J Exp Med. 2005;202:673–85. doi:10.1084/jem.20050882. PMID:16147978.
  • Britt WJ, Mach M. Human cytomegalovirus glycoproteins. Intervirology. 1996;39:401–12. doi:10.1159/000150510. PMID:9130049.
  • Gretch DR, Kari B, Rasmussen L, Gehrz RC, Stinski MF. Identification and characterization of three distinct families of glycoprotein complexes in the envelopes of human cytomegalovirus. J Virol. 1988;62:875–81. PMID:2828682.
  • Xia L, Tang A, Meng W, Freed DC, He L, Wang D, Li F, Li L, Xiong W, Gui X, et al. Active evolution of memory B-cells specifc to viral gHgL pentameric complex in healthy subjects with silent human cytomegalovirus infection. Oncotarget. 2017;8:73654–69. doi:10.18632/oncotarget.18359.
  • Fouts AE, Chan P, Stephan JP, Vandlen R, Feierbach B. Antibodies against the gH/gL/UL128/UL130/UL131 complex comprise the majority of the anti-cytomegalovirus (anti-CMV) neutralizing antibody response in CMV hyperimmune globulin. J Virol. 2012;86:7444–7. doi:10.1128/JVI.00467-12. PMID:22532696.
  • Loughney JW, Rustandi RR, Wang D, Troutman MC, Dick LW Jr, Li G, Sallusto F, Lanzavecchia A, Corti D, Gerna G. Soluble Human Cytomegalovirus gH/gL/pUL128-131 Pentameric Complex, but Not gH/gL, Inhibits Viral Entry to Epithelial Cells and Presents Dominant Native Neutralizing Epitopes. J Biol Chem. 2015;290:15985–95. doi:10.1074/jbc.M115.652230. PMID:25947373.
  • Smith MG. Propagation in Tissue Cultures of a Cytopathogenic Virus from Human Salivary Gland Virus (SGV) Disease. Exp Biol Med. 1956;92:424–30. doi:10.3181/00379727-92-22498.
  • Elek SD, Stern H. Development of a vaccine agaisnt mental retardation caused by cytomegalovirus infeciton in utero. Lancet. 1974;303:1–5. doi:10.1016/S0140-6736(74)92997-3.
  • Plotkin SA, Furukawa T, Zygraich N, Huygelen C. Candidate HCMV strain for human vaccination. Infect Immun. 1975;12:521–7. PMID:170203.
  • Murphy E, Yu D, Grimwood J, Schmutz J, Dickson M, Jarvis MA, Hahn G, Nelson JA, Myers RM, Shenk TE. Coding potential of laboratory and clinical strains of human cytomegalovirus. Proc Natl Acad Sci U S A. 2003;100:14976–81. doi:10.1073/pnas.2136652100. PMID:14657367.
  • Wang D, Shenk T. Human cytomegalovirus virion protein complex required for epithelial and endothelial cell tropism. Proc Natl Acad Sci U S A. 2005;102:18153–8. doi:10.1073/pnas.0509201102. PMID:16319222.
  • Murphy E, Shenk T. Human Cytomegalovirus Genome. Curr Top Microbiol Immunol. 2008;325:1–19.
  • Gerna G, Sarasini A, Patrone M, Percivalle E, Fiorina L, Campanini G, Gallina A, Baldanti F, Revello MG. Human cytomegalovirus serum neutralizing antibodies block virus infection of endothelial/epithelial cells, but not fibroblasts, early during primary infection. J Gen Virol. 2008;89:853–65. doi:10.1099/vir.0.83523-0. PMID:18343825.
  • Yu D, Silva MC, Shenk T. Functional map of human cytomegalovirus AD169 defined by global mutational analysis. Proc Natl Acad Sci U S A. 2003;100:12396–401. doi:10.1073/pnas.1635160100. PMID:14519856.
  • Hahn G, Revello MG, Patrone M, Percivalle E, Campanini G, Sarasini A, Wagner M, Gallina A, Milanesi G, Koszinowski U, et al. Human cytomegalovirus UL131-128 genes are indispensable for virus growth in endothelial cells and virus transfer to leukocytes. J Virol. 2004;78:10023–33. doi:10.1128/JVI.78.18.10023-10033.2004. PMID:15331735.
  • Wang D, Shenk T. Human cytomegalovirus UL131 open reading frame is required for epithelial cell tropism. J Virol. 2005;79:10330–8. doi:10.1128/JVI.79.16.10330-10338.2005. PMID:16051825.
  • Compton T. Receptors and immune sensors: the complex entry path of human cytomegalovirus. Trends Cell Biol. 2004;14:5–8. doi:10.1016/j.tcb.2003.10.009. PMID:14729174.
  • Fu TM, Wang D, Freed DC, Tang A, Li F, He X, Cole S, Dubey S, Finnefrock AC, ter Meulen J, et al. Restoration of viral epithelial tropism improves immunogenicity in rabbits and rhesus macaques for a whole virion vaccine of human cytomegalovirus. Vaccine. 2012;30:7469–74. doi:10.1016/j.vaccine.2012.10.053. PMID:23107592.
  • Krause PR, Bialek SR, Boppana SB, Griffiths PD, Laughlin CA, Ljungman P, Mocarski ES, Pass RF, Read JS, Schleiss MR, et al. Priorities for CMV vaccine development. Vaccine. 2013;32:4–10. doi:10.1016/j.vaccine.2013.09.042. PMID:24129123.
  • Fowler KB, Stagno S, Pass RF. Interval between Births and Risk of congeital CMV infection. Clin Infect Dis. 2004;38:1035–7. doi:10.1086/382533. PMID:15034839.
  • Morley PJ, Ertl P, Sweet C. Immunisation of Balb/c mice with severely attenuated murine cytomegalovirus mutants induces protective cellular and humoral immunity. J Med Virol. 2002;67:187–99. doi:10.1002/jmv.2207. PMID:11992579.
  • Shanley JD, Wu CA. Mucosal immunization with a replication-deficient adenovirus vector expressing murine cytomegalovirus glycoprotein B induces mucosal and systemic immunity. Vaccine. 2003;21:2632–42. doi:10.1016/S0264-410X(03)00037-9. PMID:12744900.
  • Shanley JD, Wu CA. Intranasal immunization with a replication-deficient adenovirus vector expressing glycoprotein H of murine cytomegalovirus induces mucosal and systemic immunity. Vaccine. 2005;23:996–1003. doi:10.1016/j.vaccine.2004.07.041. PMID:15620472.
  • González Armas JC, Morello CS, Cranmer LD, Spector DH. DNA immunization confers protection against murine cytomegalovirus infection. J Virol. 1996;70:7921–8. PMID:8892915.
  • Morello CS, Ye M, Hung S, Kelley LA, Spector DH. Systemic Priming-Boosting Immunization with a Trivalent Plasmid DNA and Inactivated Murine Cytomegalovirus (MCMV) Vaccine Provides Long-Term Protection against Viral Replication following Systemic or Mucosal MCMV Challenge. J Virol. 2005;79:159–75. doi:10.1128/JVI.79.1.159-175.2005. PMID:15596812.
  • Huang C, Wang H, Wu S, Chang H, Liu L, Peng B, Fang F, Chen Z. Comparison of multiple DNA vaccines for protection against cytomegalovirus infection in BALB/c mice. Virol J. 2014;11:104. doi:10.1186/1743-422X-11-104. PMID:24898886.
  • Schleiss MR, Heineman TC. Progress toward an elusive goal: current status of cytomegalovirus vaccines. Expert Rev Vaccines. 2005;4:381–406. doi:10.1586/14760584.4.3.381. PMID:16026251.
  • Mcvoy MA. Cytomegalovirus Vaccines. Clin Infect Dis. 2013;57:S196–9. doi:10.1093/cid/cit587. PMID:24257427.
  • Wang D, Li F, Freed DC, Finnefrock AC, Tang A, Grimes SN, Casimiro DR, Fu TM. Quantitative analysis of neutralizing antibody response to human cytomegalovirus in natural infection. Vaccine. 2011;29:9075–80. doi:10.1016/j.vaccine.2011.09.056. PMID:21945962.
  • Gerna G. Endothelial cells and CMV dissemination. Future Microbiol. 2012;7:441–4. doi:10.2217/fmb.12.12. PMID:22439720.
  • Smith MS, Bentz GL, Alexander JS, Yurochko AD. Human cytomegalovirus induces monocyte differentiation and migration as a strategy for dissemination and persistence. J Virol. 2004;78:4444–53. doi:10.1128/JVI.78.9.4444-4453.2004. PMID:15078925.
  • Saccoccio FM, Gallagher MK, Adler SP, McVoy MA. Neutralizing activity of saliva against cytomegalovirus. Clin Vaccine Immunol. 2011;18:1536–42. doi:10.1128/CVI.05128-11. PMID:21795465.
  • Cui X, Meza BP, Adler SP, McVoy MA. Cytomegalovirus vaccines fail to induce epithelial entry neutralizing antibodies comparable to natural infection. Vaccine. 2008;26:5760–6. doi:10.1016/j.vaccine.2008.07.092. PMID:18718497.
  • Pereira L, Maidji E. Cytomegalovirus infection in the human placenta: maternal immunity and developmentally regulated receptors on trophoblasts converge. Curr Top Microbiol Immunol. 2008;325:383–95. PMID:18637517.
  • Lilleri D, Kabanova A, Revello MG, Percivalle E, Sarasini A, Genini E, Sallusto F, Lanzavecchia A, Corti D, Gerna G. Human Cytomegalovirus Transmission Correlates with Delayed Maternal Antibodies to gH/gL/pUL128-130-131 Complex during Primary Infection. PloS One. 2013;8:e59863. doi:10.1371/journal.pone.0059863. PMID:23555812.
  • Furione M, Rognoni V, Sarasini A, Zavattoni M, Lilleri D, Gerna G, Revello MG. Slow increase in IgG avidity correlates with prevention of human cytomegalovirus transmission to the fetus. J Med Virol. 2013;85:1960–7. doi:10.1002/jmv.23691. PMID:23893782.
  • Ross SA, Fowler KB, Ashrith G, Stagno S, Britt WJ, Pass RF, Boppana SB. Hearing loss in children with congenital cytomegalovirus infection born to mothers with preexisting immunity. J Pediatr. 2006;148:332–6. doi:10.1016/j.jpeds.2005.09.003. PMID:16615962.
  • Yamamoto AY, Mussi-Pinhata MM, Isaac Mde L, Amaral FR, Carvalheiro CG, Aragon DC, Manfredi AK, Boppana SB, Britt WJ. Congenital cytomegalovirus infection as a cause of sensorineural hearing loss in a highly immune population. Pediatr Infect Dis J. 2011;30:1043–6. doi:10.1097/INF.0b013e31822d9640. PMID:21814153.
  • Lilleri D, Kabanova A, Revello MG, Percivalle E, Sarasini A, Genini E, Sallusto F, Lanzavecchia A, Corti D, Gerna G. Fetal human cytomegalovirus transmission correlates with delayed maternal antibodies to gH/gL/pUL128-130-131 complex during primary infection. PLoS One. 2013;8:e59863. doi:10.1371/journal.pone.0059863. PMID:23555812.
  • Jacobson MA, Sinclair E, Bredt B, Agrillo L, Black D, Epling CL, Carvidi A, Ho T, Bains R, Adler SP. Antigen-specific T cell responses induced by Towne cytomegalovirus (CMV) vaccine in CMV-seronegative vaccine recipients. J Clin Virol. 2006;35:332–7. doi:10.1016/j.jcv.2005.09.019. PMID:16387547.
  • Riddell SR, Greenberg PD. T-cell therapy of cytomegalovirus and human immunodeficiency virus infection. J Antimicrob Chemother. 2000;45:35–43. doi:10.1093/jac/45.suppl_4.35. PMID:10855770.
  • Reddehase MJ, Mutter W, Münch K, Bühring HJ, Koszinowski UH. CD8-positive T lymphocytes specific for murine cytomegalovirus immediate-early antigens mediate protective immunity. J Virol. 1987;61:3102–8. PMID:3041033.
  • Quinnan GV, Kirmani N, Rook AH, Manischewitz JF, Jackson L, Moreschi G, Santos GW, Saral R, Burns WH. Cytotoxic T Cells in Cytomegalovirus Infection: HLA-restricted T-lymphocyte and non-T-lymphocyte cytotoxic responses correlate with recovery from cytomegalovirus infection in bone-marrow-transplant recipients. N Engl J Med. 1982;307:7–13. doi:10.1056/NEJM198207013070102. PMID:6281647.
  • Reusser P, Riddell SR, Meyers JD, Greenberg PD. Cytotoxic T-lymphocyte response to cytomegalovirus after human allogeneic bone marrow transplantation: pattern of recovery and correlation with cytomegalovirus infection and disease. Blood. 1991;78:1373–80. PMID:1652311.
  • Li CR, Greenberg PD, Gilbert MJ, Goodrich JM, Riddell SR. Recovery of HLA-restricted cytomegalovirus (CMV)-specific T-cell responses after allogeneic bone marrow transplant: correlation with CMV disease and effect of ganciclovir prophylaxis. Blood. 1994 83:1971–9. PMID:8142663.
  • Einsele H. Infusion of cytomegalovirus (CMV)-specific T cells for the treatment of CMV infection not responding to antiviral chemotherapy. Blood. 2002;99:3916–22. doi:10.1182/blood.V99.11.3916. PMID:12010789.
  • Khanna R, Diamond DJ. Human cytomegalovirus vaccine: time to look for alternative options. Trends Mol Med. 2006;12:26–33. doi:10.1016/j.molmed.2005.11.006. PMID:16337831.
  • Neff BJ, Weibel RE, Buynak EB, McLean AA, Hilleman MR. Clinical and laboratory studies of live cytomegalovirus vaccine Ad-169. Proc Soc Exp Biol Med. 1979;160:32–7. doi:10.3181/00379727-160-40382.
  • Stern H. Live cytomegalovirus vaccination of healthy volunteers: eight year follow-up studies. Birth Defects Orig Artic Ser. 1984;20:263–9. PMID:6329366.
  • Gehrz RC, Christianson WR, Linner KM, Groth KE, Balfour HH Jr. CYtomegalovirus vaccine. Arch Intern Med. 1980;140:936–9. doi:10.1001/archinte.1980.00040020936013. PMID:6247988.
  • Plotkin SA, Farquhar J, Horberger E. Clinical trials of immunization with the Towne 125 strain of human cytomegalovirus. J Infect Dis. 1976;134:470–5. doi:10.1093/infdis/134.5.470. PMID:186541.
  • Schleiss M. Progress in Cytomegalovirus vaccine development. Herpes. 2005;12:66–75. PMID:16393522.
  • Plotkin SA, Higgins R, Kurtz JB, Morris PJ, Campbell DA Jr, Shope TC, Spector SA, Dankner WM. Multicenter trial of Towne strain attenuated virus vaccine in seronegative renal transplant recipients. Transplantation. 1994 58:1176–8. PMID:7992358.
  • Glazer JP, Friedman HM, Grossman RA, Starr SE, Barker CF, Perloff LJ, Huang ES, Plotkin SA. Live cytomegalovirus vaccination of renal transplant candidates. Ann Intern Med. 1979;91:676–83. doi:10.7326/0003-4819-91-5-676.
  • Marker SC, Simmons RL, Balfour HH Jr. Cytomegalovirus vaccine in renal allograft recipients. Transplant Proc. 1981;13:117–9. PMID:6267748.
  • Plotkin SA, Smiley ML, Friedman HM, Starr SE, Fleisher GR, Wlodaver C, Dafoe DC, Friedman AD, Grossman RA, Barker CF. Towne-vaccine-induced prevention of cytomegalovirus disease after renal transplants. Lancet. 1984;1:528–30. doi:10.1016/S0140-6736(84)90930-9. PMID:6142252.
  • Brayman KL, Dafoe DC, Smythe WR, Barker CF, Perloff LJ, Naji A, Fox IJ, Grossman RA, Jorkasky DK, Starr SE, et al. Prophylaxis of serious cytomegalovirus infection in renal transplant candidates using live human cytomegalovirus vaccine. Arch Surg. 1988;123:1502–8. doi:10.1001/archsurg.1988.01400360072012. PMID:2847687.
  • Plotkin SA, Starr SE, Friedman HM, Brayman K, Harris S, Jackson S, Tustin NB, Grossman R, Dafoe D, Barker C. Effect of Towne live virus vaccine on cytomegalovirus disease after renal transplant.A controlled trial. Ann Intern Med. 1991;114:525–31. doi:10.7326/0003-4819-114-7-525. PMID:1848053.
  • Plotkin SA, Huang ES. Cytomegalovirus vaccine virus (Towne strain) does not induce latency. J Infect Dis. 1985;152:395–7. doi:10.1093/infdis/152.2.395. PMID:2993436.
  • Kemble G, Duke G, Winter R, Spaete R. Defined large-scale alterations of the human cytomegalovirus genome constructed by cotransfection of overlapping cosmids. J Virol. 1996;70:2044–8. PMID:8627734.
  • Heineman TC, Schleiss M, Bernstein DI, Spaete RR, Yan L, Duke G, Prichard M, Wang Z, Yan Q, Sharp MA, et al. A phase 1 study of 4 live, recombinant human cytomegalovirus Towne/Toledo chimeric vaccines. J Infect Dis. 2006;193:1350–60. doi:10.1086/503365. PMID:16619181.
  • Bradley AJ, Lurain NS, Ghazal P, Trivedi U, Cunningham C, Baluchova K, Gatherer D, Wilkinson GW, Dargan DJ, Davison AJ. High-throughput sequence analysis of variants of human cytomegalovirus strains Towne and AD169. J Gen Virol. 2009;90:2375–80. doi:10.1099/vir.0.013250-0. PMID:19553388.
  • Gerna G, Percivalle E, Sarasini A, Baldanti F, Campanini G, Revello MG. Rescue of human cytomegalovirus strain AD169 tropism for both leukocytes and human endothelial cells. J Gen Virol. 2003;84:1431–6. doi:10.1099/vir.0.19055-0. PMID:12771411.
  • Gerna G, Percivalle E, Sarasini A, Baldanti F, Revello MG. The attenuated Towne strain of human cytomegalovirus may revert to both endothelial cell tropism and leuko- (neutrophil- and monocyte-) tropism in vitro. J Gen Virol. 2002;83:1993–2000. doi:10.1099/0022-1317-83-8-1993. PMID:12124463.
  • McVoy MA. Cytomegalovirus vaccines. Clin Infect Dis. 2013;57(Suppl 4):S196–9. doi:10.1093/cid/cit587. PMID:24257427.
  • Britt WJ, Vugler L, Butfiloski EJ, Stephens EB. Cell surface expression of human cytomegalovirus (HCMV) gp55-116 (gB): use of HCMV recombinant vaccinia virus infected cells in analysis of the human neutralizing antibody response. J Virol. 1990;64:1079–85. PMID:2154594.
  • Marshall GS, Rabalais GP, Stout GG, Waldeyer SL. Antibodies to recombinant-derived glycoprotein B after natural human cytomegalovirus infection correlate with neutralizing activity. J Infect Dis. 1992;165:381–4. doi:10.1093/infdis/165.2.381. PMID:1309850.
  • Spaete RR. A recombinant subunit vaccine approach to HCMV vaccine development. Transplant Proc. 1991;23:90–6. PMID:1648843.
  • Frey SE, Harrison C, Pass RF, Yang E, Boken D, Sekulovich RE, Percell S, Izu AE, Hirabayashi S, Burke RL, et al. Effects of antigen dose and immunization regimens on antibody responses to a cytomegalovirus glycoprotein B subunit vaccine. J Infect Dis. 1999;180:1700–3. doi:10.1086/315060. PMID:10515836.
  • Pass RF, Duliegè AM, Boppana S, Sekulovich R, Percell S, Britt W, Burke RL. A subunit cytomegalovirus vaccine based on recombinant envelope glycoprotein B and a new adjuvant. J Infect Dis. 1999;180:970–5. doi:10.1086/315022. PMID:10479120.
  • Mitchell DK, Holmes SJ, Burke RL, Duliege AM, Adler SP Immunogenicity of a recombinant human cytomegalovirus gB vaccine in seronegative toddlers. Pediatr Infect Dis J. 2002;21:133–8 doi:10.1097/00006454-200202000-00009. PMID:11840080.
  • Zhang C, Pass RF. Detection of cytomegalovirus infection during clinical trials of glycoprotein B vaccine. Vaccine. 2004;23:507–10. doi:10.1016/j.vaccine.2004.06.027. PMID:15530699.
  • Sabbaj S, Pass RF, Goepfert PA, Pichon S. Glycoprotein B vaccine is capable of boosting both antibody and CD4 T-cell responses to cytomegalovirus in chronically infected women. J Infect Dis. 2011;203:1534–4. doi:10.1093/infdis/jir138. PMID:21592981.
  • Potzsch S, Spindler N, Wiegers AK, Fisch T, Rucker P, Sticht H, Grieb N, Baroti T, Weisel F, Stamminger T, et al. B cell repertoire analysis identifies new antigenic domains on glycoprotein B of human cytomegalovirus which are target of neutralizing antibodies. PLoS Pathog. 2011;7:e1002172. doi:10.1371/journal.ppat.1002172. PMID:21852946.
  • McLaughlin-Taylor E, Pande H, Forman SJ, Tanamachi B, Li CR, Zaia JA, Greenberg PD, Riddell SR. Identification of the major late human cytomegalovirus matrix protein pp65 as a target antigen for CD8+ virus-specific cytotoxic T lymphocytes. J Med Virol. 1994;43:103–10. doi:10.1002/jmv.1890430119. PMID:8083644.
  • Wills MR, Carmichael AJ, Mynard K, Jin X, Weekes MP, Plachter B, Sissons JG. The human cytotoxic T-lymphocyte (CTL) response to cytomegalovirus is dominated by structural protein pp65: frequency, specificity, and T-cell receptor usage of pp65-specific CTL. J Virol. 1996;70:7569–79. PMID:8892876.
  • Kharfan-Dabaja MA, Boeckh M, Wilck MB, Langston AA, Chu AH, Wloch MK, Guterwill DF, Smith LR, Rolland AP, Kenney RT. A novel therapeutic cytomegalovirus DNA vaccine in allogeneic haemopoietic stem-cell transplantation: a randomised, double-blind, placebo-controlled, phase 2 trial. Lancet Infect Dis. 2012;12:290–99. doi:10.1016/S1473-3099(11)70344-9. PMID:22237175.
  • Slezak SL, Bettinotti M, Selleri S, Adams S, Marincola FM, Stroncek DF. CMV pp65 and IE-1 T cell epitopes recognized by healthy subjects. J Transl Med. 2007;5:17. doi:10.1186/1479-5876-5-17. PMID:17391521.
  • Vilalta A, Mahajan RK, Hartikka J, Rusalov D, Martin T, Bozoukova V, Leamy V, Hall K, Lalor P, Rolland A, Kaslow DC. I. Poloxamer-formulated plasmid DNA-based human cytomegalovirus vaccine: evaluation of plasmid DNA biodistribution/persistence and integration. Hum Gene Ther. 2005;16:1143–50. doi:10.1089/hum.2005.16.1143. PMID:16218775.
  • Jacobson MA, Adler SP, Sinclair E, Black D, Smith A, Chu A, Moss RB, Wloch MK. A CMV DNA vaccine primes for memory immune responses to live-attenuated CMV (Towne strain). Vaccine. 2009;27:1540–8. doi:10.1016/j.vaccine.2009.01.006. PMID:19168107.
  • Adler SP, Plotkin SA, Gonczol E, Cadoz M, Meric C, Wang JB, Dellamonica P, Best AM, Zahradnik J, Pincus S, et al. A canarypox vector expressing cytomegalovirus (CMV) glycoprotein B primes for antibody responses to a live attenuated CMV vaccine (Towne). J Infect Dis. 1999;180:843–6 doi:10.1086/314951. PMID:10438376.
  • Bernstein DI, Schleiss MR, Berencsi K, Gonczol E, Dickey M, Khoury P, Cadoz M, Meric C, Zahradnik J, Duliege AM, Plotkin S. Effect of previous or simultaneous immunization with canarypox expressing cytomegalovirus (CMV) glycoprotein B (gB) on response to subunit gB vaccine plus MF59 in healthy CMV-seronegative adults. J Infect Dis. 2002;185:686–90. doi:10.1086/339003. PMID:11865427.
  • Wang D, Shenk T. Human cytomegalovirus virion protein complex required for epithelial and endothelial cell tropism. Proc Natl Acad Sci USA. 2005;102:18153–8. doi:10.1073/pnas.0509201102. PMID:16319222.
  • Revello MG, Gerna G. Human cytomegalovirus tropism for endothelial/epithelial cells: scientific background and clinical implications. Rev Med Virol. 2010;20:136–55. doi:10.1002/rmv.645. PMID:20084641.
  • Macagno A, Bernasconi NL, Vanzetta F, Dander E, Sarasini A, Revello MG, Gerna G, Sallusto F, Lanzavecchia A. Isolation of human monoclonal antibodies that potently neutralize human cytomegalovirus infection by targeting different epitopes on the gH/gL/UL128-131A complex. J Virol. 2010;84:1005–13. doi:10.1128/JVI.01809-09. PMID:19889756.
  • Freed DC, Tang Q, Tang A, Li F, He X, Huang Z, Meng W, Xia L, Finnefrock AC, Durr E, et al. Pentameric complex of viral glycoprotein H is the primary target for potent neutralization by a human cytomegalovirus vaccine. Proc Natl Acad Sci U S A. 2013;110:E4997–5005. doi:10.1073/pnas.1316517110. PMID:24297878.
  • Wang D, Freed DC, He X, Li F, Tang A, Cox KS, Dubey SA, Cole S, Medi MB, Liu Y, et al. A replication-defective human cytomegalovirus vaccine for prevention of congenital infection. Sci Transl Med. 2016;8:362ra145. doi:10.1126/scitranslmed.aaf9387. PMID:27797961.
  • Wille PT, Wisner TW, Ryckman B, Johnson DC. Human cytomegalovirus (HCMV) glycoprotein gB promotes virus entry in trans acting as the viral fusion protein rather than as a receptor-binding protein. MBio. 2013;4:1–9. doi:10.1128/mBio.00332-13.
  • Cunningham AL. The herpes zoster subunit vaccine. Expert Opin Biol Ther. 2016;16:265–71. doi:10.1517/14712598.2016.1134481. PMID:26865048.
  • Roth K, Ferreira VH, Kaushic C. HSV-2 vaccine: current state and insights into development of a vaccine that targets genital mucosal protection. Microb Pathog. 2013;58:45–54. doi:10.1016/j.micpath.2012.11.001. PMID:23159485.
  • Stanberry LR. HERPES. Dermatol Clin. 1998;16:811–6. doi:10.1016/S0733-8635(05)70052-8. PMID:9891686.
  • White CJ, Kuter BJ, Hildebrand CS, Isganitis KL, Matthews H, Calandra GB, Provost PJ, Ellis RW, Gerety RJ, Calandra GB. Varicella Vaccine (VARIVAX) in Healthy Children and Adolescents: Results From Clinical Trials, 1987 to 1989. Pediatrics. 1991;87:604–10. PMID:1850506.
  • Keating GM. Shingles (Herpes Zoster) Vaccine (Zostavax): A Review in the Prevention of Herpes Zoster and Postherpetic Neuralgia. BioDrugs. 2016;30:243–54. doi:10.1007/s40259-016-0180-7. PMID:27189459.
  • Pepperl S, Münster J, Mach M, Harris JR, Plachter B. Dense Bodies of Human Cytomegalovirus Induce both Humoral and Cellular Immune Responses in the Absence of Viral Gene Expression. J Virol. 2000;74:6132–46. doi:10.1128/JVI.74.13.6132-6146.2000. PMID:10846097.
  • Griffiths PD, Stanton A, McCarrell E, Smith C, Osman M, Harber M, Davenport A, Jones G, Wheeler DC, O'Beirne J, et al. Cytomegalovirus glycoprotein-B vaccine with MF59 adjuvant in transplant recipients: a phase 2 randomised placebo-controlled trial. Lancet. 2011;377:1256–63. doi:10.1016/S0140-6736(11)60136-0. PMID:21481708.
  • Reap EA, Morris J, Dryga SA, Maughan M, Talarico T, Esch RE, Negri S, Burnett B, Graham A, Olmsted RA, et al. Development and preclinical evaluation of an alphavirus replicon particle vaccine for cytomegalovirus. Vaccine. 2007;25:7441–9. doi:10.1016/j.vaccine.2007.08.016. PMID:17870214.
  • Loomis RJ, Lilja AE, Monroe J, Balabanis KA, Brito LA, Palladino G, Franti M, Mandl CW, Barnett SW, Mason PW. Vectored co-delivery of human cytomegalovirus gH and gL proteins elicits potent complement-independent neutralizing antibodies. Vaccine. 2013;31:919–26. doi:10.1016/j.vaccine.2012.12.009. PMID:23246547.
  • Berencsi K, Gyulai Z, Gönczöl E, Pincus S, Cox WI, Michelson S, Kari L, Meric C, Cadoz M, Zahradnik J, et al. A Canarypox Vector–Expressing Cytomegalovirus (CMV) Phosphoprotein 65 Induces Long-Lasting Cytotoxic T Cell Responses in Human CMV-Seronegative Subjects. J Infect Dis. 2001;183:1171–9. doi:10.1086/319680. PMID:11262198.
  • Wang Z, La Rosa C, Lacey SF, Maas R, Mekhoubad S, Britt WJ, Diamond DJ. Attenuated poxvirus expressing three immunodominant CMV antigens as a vaccine strategy for CMV infection. J Clin Virol. 2006;35:324–31. doi:10.1016/j.jcv.2005.09.018. PMID:16388983.
  • Saccoccio FM, Sauer AL, Cui X, Armstrong AE, Habib el-SE, Johnson DC, Ryckman BJ, Klingelhutz AJ, Adler SP, McVoy MA. Peptides from cytomegalovirus UL130 and UL131 proteins induce high titer antibodies that block viral entry into mucosal epithelial cells. Vaccine. 2011;29:2705–11. doi:10.1016/j.vaccine.2011.01.079. PMID:21310190.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.