2,732
Views
13
CrossRef citations to date
0
Altmetric
Review

Development of novel vaccines against human cytomegalovirus

&
Pages 2673-2683 | Received 26 Dec 2018, Accepted 05 Mar 2019, Published online: 24 Apr 2019

ABSTRACT

Congenital human cytomegalovirus (HCMV) infection and HCMV infection of the immunosuppressed patients cause significant morbidity and mortality, and vaccine development against HCMV is a major public health priority. Efforts to develop HCMV vaccines have been ongoing for 50 y, though no HCMV vaccine has been licensed; encouraging and promising results have obtained from both preclinical and clinical trials. HCMV infection induces a wide range of humoral and T cell-mediated immune responses, and both branches of immunity are correlated with protection. In recent years, there have been novel approaches toward the development of HCMV vaccines and demonstrated that vaccine candidates could potentially provide superior protection over natural immunity acquired following HCMV infection. Further, rationally designed HCMV protein antigens that express native conformational epitopes could elicit optimal immune response. HCMV vaccine candidates, using a multi-antigen approach, to maximize the elicited protective immunity will most likely be successful in development of HCMV vaccine.

Human cytomegalovirus (HCMV) is an enveloped, double-stranded DNA β-herpesvirus of the Herpesviridae family and causes infection in 40–60% of the population in industrialized countries and 80–100% of the population in developing countries.Citation1, Citation2HCMV infection is correlated with older age, low household income, and poor hygiene standards.Citation3-Citation6 Although HCMV infection in immunocompetent individuals is generally asymptomatic, congenital infection of the neonates and infection of the immunosuppressed, including transplant recipients and patients with HIV/AIDS, cause significant morbidity and mortality.Citation1,Citation2,Citation7-Citation9 Congenital HCMV infection is the leading nongenetic cause of hearing loss in childhood, and additional congenital sequelae include microcephaly, seizures, intracranial calcifications, cerebral palsy, hepatitis, chorioretinitis resulting in vision loss, and neurodevelopmental delay including mental retardation.Citation1,Citation2,Citation10,Citation11 Congenital HCMV transmission to the fetus occurs in 0.5–0.7% of pregnancies in the United States and other developed countries, and in up to 2% of the pregnancies in developing countries.Citation7 Approximately 20–25% of infants who are congenitally infected will develop sensorineural hearing loss, and up to 35% will have other sequelae involving the central nervous system.Citation12 In developed countries, congenital cytomegalovirus (CMV) is the most common infectious cause of brain damage and sensorineural hearing loss and is an occasional cause of mortality.Citation13 In solid organ and hematopoietic stem cell transplant patients, HCMV infection causes viremia with attendant end-organ diseases such as hepatitis and pneumonitis and significantly increases the chance of graft rejection, graft failure, and in hematopoietic stem cell transplant patients, graft-versus-host disease.Citation14-Citation17 Despite active monitoring and management with antiviral drugs, the incidence of HCMV infection is still high, ranging from 20% to 70% in the first-year posttransplantation, and HCMV infection remains one of the most common complications affecting patient survival among solid organ and hematopoietic stem cell transplant recipients.Citation18-Citation22

HCMV is spread mainly via saliva and urine to seronegative children and adults, and transplacentally to the fetus.Citation23,Citation24 The target cells of HCMV include fibroblasts, epithelial cells, endothelial cells, monocyte–macrophages, hepatocytes, and neurons, and the mechanism of HCMV fusion and entry into mammalian cells is analogous to that employed by other members of the herpesvirus family.Citation25,Citation26 HCMV enters cells by fusing its envelope with either the plasma membrane or endosomal membrane.Citation27,Citation28 HCMV envelope proteins, glycoprotein B (gB), gH, gL, gO, and UL128/UL130/UL131A proteins have collectively been identified as the envelope proteins that play critical role in HCMV fusion and entry into host cells.Citation25,Citation27 The gB is the direct mediator of HCMV fusion with all host cell membranes.Citation29,Citation30 The activation of HCMV gB for fusogenic activity requires its association with the gH/gL/gO protein complex. However, the protein complex comprising five envelope proteins gH/gL/UL128/UL130/UL131A (pentameric complex) is further required for efficient targeting of HCMV to epithelial and endothelial cells.Citation25,Citation28,Citation31,Citation32

HCMV infection induces a wide range of humoral and T-cell-mediated immune responses, and both are correlated with protection. Potent-neutralizing antibody targeting the pentameric complex and phosphoprotein 65 (pp65)-specific CD4+ T cells have both been implicated with reduced risk of intrauterine HCMV transmission following primary maternal infection.Citation33-Citation36 In addition, hyperimmune globulin treatment during primary maternal HCMV infection has been shown to be beneficial to prevent or reduce congenital HCMV infection and disease, although this remains controversial.Citation37,Citation38 These clinical findings are further bolstered by studies using the guinea pig and rhesus macaque models of congenital HCMV infection, where hyperimmune globulin preparations, antibodies specific for gB or gH/gL, and CD4+ T cells were demonstrated to play a role in preventing intrauterine virus transmission and fetal demise.Citation39-Citation44

T-cell immunity is believed to play a pivotal role for HCMV containment in transplant organ recipients, and delayed recovery of HCMV-specific T cells in these patients was a significant risk factor for HCMV-related complications with higher rates of recurrent or persistent HCMV infection.Citation45 Further, delayed recovery of multicytokine producing T cells was associated with an increased antiviral drug usage.Citation46 More recently, the vaccine clinical trial using HCMV gB adjuvanted with microfluidized adjuvant 59 (MF59) demonstrated a significant reduction in viremia and the total number of days of antiviral drug treatment in solid-organ transplant recipients, with the best results observed in HCMV-seronegative recipients of transplants from HCMV-seropositive donors, suggesting a key role for humoral immunity against HCMV infection in transplantation setting.Citation50

For the past 50 y, a variety of experimental vaccine approaches to stimulate the host immune response to HCMV have been evaluated and many are in various stages of research, though no vaccine to prevent or treat HCMV infection or disease has yet been licensed.Citation47-Citation49 An efficient HCMV vaccine candidate will likely need to stimulate multifunctional immune responses that cover both arms of adaptive immunity.Citation48,Citation49 This review will be focused on HCMV vaccine development efforts taking novel approaches, with the potential to become licensed vaccines ().

Table 1. Novel HCMV vaccine candidates.

1. Recombinant trimeric HCMV gB and the pentameric complex

The anti-gB antibody in human sera was identified as the major neutralizing activity that prevents HCMV infection of fibroblasts. HCMV subunit vaccines incorporating soluble monomeric gB have been under development for years, and subunit approaches utilizing adjuvanted recombinant formulations of gB have advanced the furthest in clinical trials of HCMV vaccines to date.Citation65,Citation66 Several phase I and phase II clinical trials, utilizing a recombinant HCMV gB (Chiron gB) in MF59 adjuvant (MF59, Novartis), have been completed and demonstrated encouraging results.Citation24,Citation50,Citation51,Citation67,Citation68

In a phase II study in postpartum women, the gB/MF59 vaccine demonstrated 50% efficacy against primary HCMV infection in seronegative women vaccinated within 1 y of giving birth compared to women in the same cohort who received the placebo.Citation24 This landmark study was the first clinical trial demonstrating the efficacy of any vaccine for preventing primary HCMV infection, an important milestone in progress toward maternal immunization against congenital HCMV transmission. Another gB/MF59 vaccine multicenter study in healthy HCMV-seronegative adolescent women demonstrated 43% efficacy in preventing primary HCMV infection, though the difference was not statistically significant compared to placebo.Citation51 This was likely because the unexpectedly lower incidence of infection in controls than that had been previously observed in similar studies did not allow discernment of statistical significance.Citation47,Citation51 There is also the possibility that this multicenter trial in adolescent women was more objective than the single-center trial in postpartum women.Citation24,Citation51 Finally, solid-organ transplant recipients vaccinated with the gB/MF59 vaccine demonstrated both a reduction in viremia and in the total number of days requiring ganciclovir treatment compared to those who received placebo.Citation50 The benefit of vaccinating was most striking in HCMV-seronegative recipients of transplants from HCMV-seropositive donors, and the duration of viremia posttransplantation was inversely correlated with the magnitude of the gB antibody response.Citation50

The gB vaccine adjuvanted with MF59 used in these clinical trials was originally developed at Chiron Corporation (acquired by Novartis), expressed as a truncated, secreted polypeptide, and the protein was purified by chromatography from tissue culture supernatants in the Chinese hamster ovary (CHO) cells.Citation69 This Chiron gB did not recapitulate the conformation of gB antigen expressed on virions and/or the surface of HCMV-infected cells; therefore, recombinant gB proteins that allow expression of conformational epitopes may elicit more important protective vaccine responses.Citation69,Citation70

The natural conformation of HCMV gB within the viral envelope is a trimer, and thus, a trimeric gB is predicted to be a superior vaccine target, as trimeric HCMV gB likely expresses native conformational epitopes that will elicit higher titers of HCMV-neutralizing antibodies. The furin cleavage site within the HCMV gB protein is critical for mediating HCMV gB folding into its terminal trimeric form.Citation71-Citation73 However, in studies to express recombinant HCMV gB, the inclusion of the furin cleavage site led to low yields of monomeric gB, whereas the elimination of this site by mutation resulted in efficient production, but synthesis of mostly monomeric gB, with some higher MW forms, using a variety of mammalian and insect cells.Citation74-Citation77 Recently, mutations to the fusion loops of a HCMV gB consisting of amino acid residues 78–706 resulted in a trimeric gB produced in insect cells, with the structure subsequently analyzed by X-ray crystallography.Citation78 Another trimeric HCMV gB in a post-fusion conformation was produced and consisted of amino acid residues 86–698 bound to the Fab fragments of a neutralizing human anti-gB antibody, with the structure also analyzed by X-ray crystallography.Citation79 Of note, these trimeric gBs had mutations to their fusion loops that might have eliminated epitopes important for eliciting HCMV-neutralizing antibodies. Further, the trimeric gBs analyzed by X-ray crystallography had mutated furin cleavage sites that might have altered the native conformation of the protein. Therefore, these trimeric HCMV gB recombinant proteins may not be suitable for vaccine use.

We have produced, within CHO cells, a trimeric HCMV gB by insertion of a flexible 15 amino acid (Gly4Ser)3 linker at the furin cleavage site that allowed for terminal protein folding and efficient expression.Citation53 Trimeric HCMV gB induced 5- to 11-fold higher serum titers of gB-specific IgG relative to monomeric HCMV gB similar to the Chiron gB that was previously used in phase II clinical trials and elicited 50-fold higher complement-independent HCMV neutralization activity, suggesting that conformational epitopes of the trimeric HCMV gB played an important role in eliciting neutralization activity.Citation53 Soluble monomeric HCMV gB as well as different post-fusion HCMV trimeric gBs elicited mainly complement-dependent HCMV-neutralizing antibodies.Citation79-Citation81 In contrast, the trimeric HCMV gB produced in our laboratory elicited markedly higher serum HCMV-neutralizing antibodies that exhibited both complement-independent and complement-dependent activity.Citation53 These results may be due to the trimeric HCMV gB having a 15 amino acid flexible linker inserted into the furin cleavage site that allowed the two subdomains of HCMV gB to fold into their native conformation.Citation78,Citation79 The conformational epitopes expressed by the two subdomains of the trimeric gB might play key role in eliciting neutralizing anybody responses.Citation53,Citation78,Citation79

In addition, the trimeric gB made in our laboratory elicited markedly higher cross-strain neutralization activity against several clinical HCMV strains and an HCMV strain AD169 variant expressing a functional pentameric complex (AD169wtCitation131), compared to the monomeric gB that was similar to the gB protein made by Chiron.Citation53 In contrast, in phase II clinical trials, Chiron gB/MF59 vaccine elicited antibodies exhibited limited neutralization of the autologous virus and negligible neutralization of multiple heterologous strains.Citation24,Citation37,Citation52,Citation69,Citation82-Citation84 Though these data suggest that nonneutralizing antibody functions, including virion phagocytosis, antibody-dependent cell-mediated cytotoxicity. etc., likely played a role in the observed ~50% protection mediated by the Chiron gB/MF59 vaccine against HCMV acquisition.Citation52,Citation84 These data support that the trimeric HCMV gB produced in our laboratory is a promising vaccine candidate, and future studies of the trimeric HCMV gB should also take account of nonneutralizing antibody functions.

The pentameric complex has been extensively studied as a vaccine candidate in recent years. Analysis of sera from 365 HCMV seropositive women aged from 18 to 84 showed that the neutralizing activity against epithelial cells was 8–15-fold higher than that against fibroblast cells.Citation85 Further, the majority of the anti-cytomegalovirus neutralizing antibody in HCMV hyperimmune globulin was against the pentameric complex, and depletion with pentameric complex decreased 85% of the HCMV neutralizing activity against epithelial cells.Citation86 Immunization of mice with recombinant pentameric complex formulated with different adjuvants elicited long-term persistent HCMV neutralizing antibody titers that were a-100–1000-fold higher than those found in individuals that recovered from primary HCMV infection.Citation54 More importantly, sera from mice immunized with the pentameric complex neutralized the infection of both epithelial cells and fibroblasts and prevented cell-to-cell spread and viral dissemination from endothelial cells to leukocytes.Citation54

Pentameric complex elicited immune response is likely to provide protection against HCMV infection of epithelial cells, endothelial cells, and monocytes, but not fibroblasts or primary trophoblast progenitor cells.Citation54,Citation81,Citation87-Citation92 Since HCMV gB elicits relatively higher HCMV neutralization activity for fibroblasts than epithelial cells, whereas pentameric complex elicits high HCMV neutralization activity for epithelial cells, endothelial cells, and monocytes; but lower neutralization activity for fibroblasts, it suggests that an optimal prophylactic HCMV vaccine will consist of both trimeric gB and pentameric complex proteins.

2. Transgenic disabled infectious single-cycle HCMV vaccines

Earlier clinical trials using live attenuated Towne or AD169 HCMV viral vaccines, both of which lacked expression of the pentameric complex, proved to be ineffective in preventing HCMV infection in either healthy volunteers or renal transplant recipients, although some efficacy was demonstrated in overt HCMV disease in high risk recipient–donor+ renal transplant recipients.Citation23,Citation65 New HCMV viral strains engineered to express the pentameric complex are currently being evaluated, but safety concerns persist using this approach. A considerable barrier to the development of an attenuated HCMV vaccine is the concern that the vaccine strain could potentially establish viral latency, predisposing the recipient to reactivation and associated disease complications later in life.Citation47,Citation48

In light of the persistent and incompletely resolved concerns about the safety profile of live attenuated HCMV vaccines, the generation of transgenic disabled infectious single-cycle (DISC) vaccines has become an attractive alternative. DISC vaccines are replication defective but could elicit a full repertoire of antibody responses to envelope glycoproteins, including the pentameric complex, and could induce a broad range of T-cell responses to multiple viral proteins, providing a much greater breadth of responses than those induced by subunit vaccines.Citation47 V160 is one of the recently developed HCMV DISC vaccines currently undergoing phase I clinical trials in both seronegative and seropositive subjects.Citation56

This V160 vaccine, designed by Merck Vaccines, had a restored wild-type pentameric complex sequence in HCMV strain AD169 and was propagated in human retinal pigmented epithelial (ARPE-19) cells. V160 was further modified such that viral proteins immediate-early 1/2 (IE1/IE2) and UL51 were expressed as fusion proteins with FKBP12, a rapamycin-binding protein.Citation56,Citation65,Citation93-Citation95 As UL51 and IE1/2 are essential for replication competence, V160 is able to propagate in ARPE-19 cells only in the presence of a synthetic stabilizing ligand, Shield-1, whereas, in an immunized subject, the fusion protein is rapidly degraded and viral replication is inhibited, providing an excellent safety profile for the vaccine.Citation56,Citation93 V160 has recently completed phase I testing, and it was reported that after three doses immunization at 0, 1, and 6 months, V160 combined with Merck aluminum phosphate adjuvant-induced neutralizing antibody titers equal to or higher than those observed in naturally seropositive subjects measured in epithelial cells.Citation47 The vaccine also induced interferon gamma-producing T cells as measured by enzyme-linked immunosorbent spot (ELISPOT) assays at levels equal to or higher than those seen with natural seropositives. The vaccine was well tolerated in this phase I study, and there was no virus shedding in inoculated subjects. Merck plans to proceed to evaluate this candidate vaccine in a phase II study.Citation47,Citation56

3. Viral vector HCMV vaccines

The use of viral vectors to express HCMV-encoded proteins such as gB, pentameric complex, pp65, IE-1, and/or IE2 represents another promising approach to developing an HCMV vaccine. Several different viral vectors have been used for HCMV vaccine development, and modified vaccinia virus Ankara (MVA) vector vaccine candidates demonstrated the most promising results.Citation96-Citation105 MVA is one of the most advanced viral vectors for vaccine development and clinical investigation, because of its excellent safety profile and property of inducing potent immune responses against recombinant antigens.Citation106

MVA has been used to express a variety of HCMV antigens, including pp65, gB, IE1, IE2, and the pentameric complex proteins. In rodent and nonhuman primate model systems, MVA-vectored vaccines have demonstrated excellent immunogenicity in eliciting neutralizing antibody and T-cell immune response.Citation89,Citation107,Citation108 In the guinea pig CMV (Cytomegalovirus) congenital infection model, MVA-vectored gB/pp65 homolog (GP83)-based vaccines were immunogenic and protective against congenital transmission and disease.Citation109 Vaccination of mice or macaques with MVA-vectored pentameric complex vaccines elicited neutralizing antibody responses that reached serum peak levels comparable to neutralizing antibody titers found in HCMV hyperimmune globulins.Citation91 Moreover, a pp65/IE1 fusion protein has been expressed in MVA and has been shown to activate and expand the levels of pp65- and IE1-specific T cells derived from HCMV-seropositive donors following infection of CD40-activated B cells and to induce HCMV pp65- and IE1-epitope-specific T-cell responses in HLA transgenic mice.Citation57

A triplex MVA-vectored vaccine encoding pp65, IE1-exon4, and IE2-exon5 has been investigated in a phase Ib study and was found to induce robust and durable expansion of CD4 and CD8 T cells specific for each immuno-dominant HCMV protein both in HCMV seropositive and seronegative individuals.Citation58 This vaccine candidate is currently in a phase II trial in hematopoietic stem cell transplantation patients for the prevention of HCMV reactivation, HCMV disease, and use of antiviral therapy. More recently, MVA viral vector encoding a combination of the pentamer complex, gB, and pp65 has been conducted, and immunization in mice elicited potent complement-independent and complement-dependent HCMV-neutralizing antibodies as well as mouse and human Major histocompatibility complex (MHC)-restricted, polyfunctional T-cell responses by the individual antigens.Citation59 The major limitation for MVA vectored vaccines is the vector-specific immunity elicited after repeated immunization, which may prevent periodic booster immunizations for sustaining protection against congenital HCMV infection in women of reproductive age during serial pregnancies.Citation55,Citation110

4. Enveloped virus-like particle HCMV vaccines

Enveloped virus-like particles (eVLPs) are protein structures that mimic enveloped wild-type viruses but do not have a viral genome and create safer vaccine candidates in principle.Citation47 eVLPs could potentially elicit Immune responses comparable to or better than natural infection by closely mimicking structure of target virus.Citation48 An eVLP gB HCMV vaccine, manufactured by VBI laboratories, is currently in phase I studies in HCMV seronegative subjects. The eVLP gB was produced by co-transfection of the HCMV gB with the Moloney murine leukemia virus (MLV) gag protein in human embryonic kidney (HEK) cells. The expressed MLV gag protein is cleaved by cellular proteases to yield the viral matrix, capsid, and nucleocapsid proteins, and capsid proteins spontaneously assemble into VLPs which then acquire a lipid envelope as they are released from the cell.Citation60 Inclusion of HCMV gB allows this protein to be expressed in the envelope of eVLP, with an authentic glycosylation profile derived from posttranslational processing in HEK cells. Two gB-variant eVLPs were produced: one expressed the full-lehgth HCMV gB (gB eVLP) and the other expressed the extracellular portion of HCMV gB fused with the transmembrane domain and cytoplasmic domain of vesicular stomatitis virus G protein (gB-G eVLP).Citation60 Both vaccines were found to induce neutralizing antibody titers 10-fold higher than titers induced with the same dose of soluble recombinant gB after immunization in mice, with titer levels comparable to those observed with immunoglobulin (Cytogam) treatment.Citation60 Further, the gB-G eVLP was more immunogenic, which was proposed to be due to the gB-G assuming a “post-fusion” conformation in transfected cells.Citation60,Citation61

A phase I study of the gB-G eVLP (VBI-1501A) was initiated in 2016 where four dose formulations of the gB vaccine were administered with and without an alum adjuvant in a group of approximately 125 HCMV-seronegative volunteers.Citation61 An additional eVLP HCMV vaccine candidate, expressing both gB and pp65, has also been developed by VBI and a clinical trial has started for potential therapeutic benefit in patients with HCMV-associated glioblastoma multiforme.Citation47

Another candidate eVLP vaccine against HCMV was developed by Redvax GmbH, a derivative of Redbiotec AG. In contrast to the VBI approach, which uses mammalian (HEK) cells to produce the VLP, the Redbiotec expression platform is based on a baculovirus expression system.Citation61 The Redvax technology can potentially generate VLP vaccine candidates containing various combinations of HCMV gB, the pentameric complex, and glycoproteins gM and gN.Citation47 The potential pitfall is that the glycosylation pattern of the proteins produced in baculovirus is different from that in mammalian cells and may negatively impact on the quality of the elicited immune response. A study in rhesus macaques with the pentameric complex eVLP ± gB eVLP generated using the Redvax technology was recently reported by Pfizer. Despite the elicitation of high-titer neutralizing antibodies and good T-cell responses after immunization, no protection was demonstrated from viremia upon challenge.Citation62

5. RNA HCMV vaccines

Clinical trials of DNA-based HCMV vaccines encoding both gB and pp65 developed by Vical Corporation (ASP0113) have been conducted in the hematopoietic stem cell transplant and solid-organ transplant patient populations, with the goal of reducing HCMV disease in this uniquely vulnerable population.Citation111-Citation114 ASP0113 elicited pp65 and/or gB-specific T-cell responses and gB antibody responses in phase I and phase II clinical trials and demonstrated a statistically significant reduction of HCMV viremia following vaccination, as well as a trend toward reduced use of anti-HCMV antivirals in immunized subjects.Citation111,Citation113-Citation118 However, a recent communication from the randomized, double-blind, and placebo-controlled phase III study showed that it did not meet its primary or secondary endpointsCitation48 (https://www.astellas.com/en/search?keys=asp0113). The results did not demonstrate a significant improvement in overall survival and reduction in HCMV end-organ disease.Citation48 These disappointing results from the DNA-based HCMV vaccine candidate ASP0113 may be due to the poor immunogenicity of DNA vaccine technology, and safety concerns about DNA integration into the host genome post-transfection remain an additional barrier.Citation119

RNA-based nucleic acid vaccines against HCMV have also been developed and explored in preclinical studies. A self-amplifying mRNA vaccine platform encoding gB and pp65-IE1 developed by Novartis Vaccines was evaluated in rhesus macaques.Citation63,Citation64 Immunization of this vaccine formulated with a cationic nanoemulsion elicited antigen-specific immune responses, including both total anti-gB IgG and neutralizing antibody responses after a single immunization, and was boosted 3-fold after a second immunization.Citation64 Further, all animals also had measurable CD4+ and CD8+ T-cell responses after two immunizations.Citation64 Moderna Therapeutics has recently published preclinical development of a multiple-component HCMV mRNA vaccine consisting of the five constituents of pentameric complex, gB, and pp65.Citation120 Immunization of mice and nonhuman primates with lipid nanoparticles encapsulating modified mRNA encoding HCMV gBs and pentameric complex elicited potent and durable neutralizing antibody titers, and administration of pp65 vaccine with pentameric complex and gB elicited robust multi-antigenic T-cell responses in mice.Citation120

While preclinical studies have generated great optimism about the prospects and advantages of mRNA-based vaccines, two recent clinical trials with mRNA–lipid nanoparticle vaccines encoding influenza hemagglutinin and rabies virus glycoprotein have led to more tempered expectations.Citation121,Citation122 In both trials, immunogenicity was more modest in humans than was expected based on animal models, a phenomenon also observed with DNA-based vaccines.Citation123 To improve the efficacy of mRNA–lipid nanoparticle vaccines in clinical trials, it is expected that further research is required to determine how different animal species respond to mRNA vaccine components and inflammatory signals and which pathways of immune signaling are most effective in humans.Citation124

6. Concluding remarks

1. Preexisting HCMV immunity is protective. Natural immunity against HCMV infection is protective for congenital infection, though it is not complete. Prospective studies showed that maternal immunity is protective against congenital HCMV infection, with highly significantly reduced rates of vertical transmission in women with nonprimary compared to primary infections.Citation125,Citation126 Primary infections result in HCMV transmission in approximately 30% of affected pregnancies, whereas preexisting maternal immunity confers a 69% reduction of the risk of congenital HCMV in future pregnancies.Citation127,Citation128 Moreover, there is evidence that sequelae of congenital HCMV infection are reduced in the setting of preconception maternal immunity. This has been demonstrated for sensorineural hearing loss, where both the severity and risk of progression of hearing loss are more substantial in infected infants born to transmitting mothers with primary HCMV infections during pregnancy than in those infants acquiring congenital HCMV in the context of recurrent maternal infection.Citation129

2. Subunit and viral vector HCMV vaccine candidates could elicit distinctive and highly protective immune responses and could potentially provide superior protection over natural immunity. Subunit vaccine candidates based on purified HCMV proteins and viral vector HCMV vaccine candidates have the potential to elicit antigen-specific immune responses that are quantitatively or qualitatively different from those induced by HCMV during natural infection.Citation48 These vaccine candidates may potentially provide protection in HCMV seronegative and seropositive individuals that exceeds the protection level afforded by naturally acquired HCMV immunity and potentially provide superior protection than natural HCMV immunity.Citation48

3. HCMV protein antigens expressing native conformational epitopes could elicit optimal immune response. Immunogen conformation has been recognized as an extraordinarily important consideration for HCMV gB as well as the pentameric complex. Following natural infection, some gB-specific antibodies are neutralizing, though the majority are nonneutralizing.Citation130 HCMV gB is predicted to have pre-fusion form and post-fusion form, and it has been hypothesized that neutralizing antibodies preferentially target epitopes exposed on the pre-fusion form of the protein, and nonneutralizing antibodies those on the post-fusion form.Citation78 Immunization with soluble post-fusion gB elicited low-level binding responses against neutralizing gB epitopes in comparison with natural infection, suggesting that neutralizing epitopes are not adequately exposed to immune cells when gB is in the post-fusion form.Citation52 We have produced a trimeric HCMV gB by the insertion of a flexible 15 amino acid (Gly4Ser)3 linker at the furin cleavage site that allowed the two subdomains of HCMV gB to fold into their native conformation, with the expression of conformational epitopes.Citation53 Though the pre-fusion or post-fusion form of this trimeric gB has not been determined yet, it elicited markedly higher titers cross-reactive HCMV-neutralizing antibody in mice compared to a soluble HCMV gB.Citation53 The structural biology of the pentameric complex is also of interest in the HCMV vaccine field, and it is suggested that native folding and assembly of the full complex may be critical for optimal neutralizing antibody responses, and the elicitation of extremely potent epithelial cell-neutralizing antibodies.Citation49,Citation88,Citation131

4. A combination of antigens may be required for HCMV vaccine candidates to maximize protection. An efficient HCMV vaccine may require a multi-antigen approach, incorporating diverse epitopes to optimally engage both humoral and cellular immune factors, thus maximizing the protective immunity elicited.Citation48,Citation49 Multi-epitope immune responses can be achieved either through vaccination with a live-attenuated virus or through delivery and/or in vivo expression of a combination of antigens. Multi-antigen vaccine candidates for HCMV using the combination of gB and pp65, either as DNA or co-expressed in a viral vector, were highly immunogenic and demonstrated additive protection in a guinea pig congenital transmission model.Citation61,Citation105 Live attenuated vaccines are unlikely to provide protection that exceeds the level of natural immunity, and the efficacy of viral vector vaccines could be significantly reduced by vector-specific immune response elicited after repeated immunization. The use of a combination of HCMV recombinant proteins such as trimeric gB and the pentameric complex represents a safe and efficient approach that could potentially provide superior protection over natural immunity. Though recombinant proteins such as Chiron gB may elicit short-term protection, this could potentially be improved by using proteins expressing conformational epitopes and novel potent adjuvants.

Disclosure of potential conflicts of interest

Drs. Xinle Cui and Clifford M. Snapper are inventors of a patent for using trimeric herpesvirus gBs as vaccine candidates, and a pending patent for using combination of herpesvirus envelope proteins as vaccine candidates.

Mandatory disclaimer: The opinions expressed herein are those of the authors and are not necessarily representative of those of the Uniformed Services University of the Health Sciences (USUHS), the Department of Defense (DOD), or the United States Army, Navy or Air Force.

Additional information

Funding

Supported by USUHS Dean’s Research and Education Endowment Fund (CMS). USUHS had no involvement in study design, data collection, analysis or interpretation, nor writing report or decision for publication.

References

  • Demmler-Harrison GJ. Congenital cytomegalovirus: public health action towards awareness, prevention, and treatment. J Clin Virol. 2009;46 Suppl 4:S1–5. Epub 2009/ 11/03. doi: 10.1016/j.jcv.2009.10.007. PubMed PMID: 19879187. doi:. .
  • Jeon J, Victor M, Adler SP, Arwady A, Demmler G, Fowler K, Goldfarb J, Keyserling H, Massoudi M, Richards K, et al. Knowledge and awareness of congenital cytomegalovirus among women. Infect Dis Obstet Gynecol. 2006;2006:80383. doi:10.1155/IDOG/2006/80383. PubMed PMID: 17485810; PubMed Central PMCID: PMCPMC1779612.
  • Staras SAS, Dollard SC, Radford KW, Flanders WD, Pass RF, Cannon MJ. Seroprevalence of cytomegalovirus infection in the United States, 1988–1994. Clin Infect Dis. 2006;43(9):1143–51. doi:10.1086/508173. PubMed PMID: 17029132.
  • Staras SAS, Flanders WD, Dollard SC, Pass RF, McGowan JE Jr., Cannon MJ. Cytomegalovirus seroprevalence and childhood sources of infection: A population-based study among pre-adolescents in the United States. J Clin Virol. 2008;43(3):266–71. doi:10.1016/j.jcv.2008.07.012. PubMed PMID: 18778968.
  • Bate SL, Dollard SC, Cannon MJ. Cytomegalovirus seroprevalence in the United States: the national health and nutrition examination surveys, 1988–2004. Clin Infect Dis. 2010;50(11):1439–47. doi:10.1086/652438. PubMed PMID: 20426575.
  • Colugnati FA, Staras SA, Dollard SC, Cannon MJ. Incidence of cytomegalovirus infection among the general population and pregnant women in the United States. BMC Infect Dis. 2007;7:71. doi:10.1186/1471-2334-7-71. PubMed PMID: 17605813; PubMed Central PMCID: PMCPMC1925089.
  • Kenneson A, Cannon MJ. Review and meta-analysis of the epidemiology of congenital cytomegalovirus (CMV) infection. Rev Med Virol. 2007;17(4):253–76. doi:10.1002/rmv.535. PubMed PMID: 17579921.
  • 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(1):89–97. doi:10.1111/j.1399-0012.2007.00750.x. PubMed PMID: 18217909.
  • Steininger C, Puchhammer-Stöckl E, Popow-Kraupp T. Cytomegalovirus disease in the era of highly active antiretroviral therapy (HAART). J Clin Virol. 2006;37(1):1–9. doi:10.1016/j.jcv.2006.03.005. PubMed PMID: 16675299.
  • Morton CC, Nance WE. Newborn hearing screening–a silent revolution. N Engl J Med. 2006;354(20):2151–64. doi:10.1056/NEJMra050700. PubMed PMID: 16707752.
  • Manicklal S, Emery VC, Lazzarotto T, Boppana SB, Gupta RK. The “silent” global burden of congenital cytomegalovirus. Clin Microbiol Rev. 2013;26(1):86–102. doi:10.1128/CMR.00062-12. PubMed PMID: 23297260; PubMed Central PMCID: PMCPMC3553672.
  • Pass RF, Fowler KB, Boppana SB, Britt WJ, Stagno S. Congenital cytomegalovirus infection following first trimester maternal infection: symptoms at birth and outcome. J Clin Virol. 2006;35(2):216–20. doi:10.1016/j.jcv.2005.09.015. PubMed PMID: 16368262.
  • Bristow BN, O’Keefe KA, Shafir SC, Sorvillo FJ. Congenital cytomegalovirus mortality in the United States, 1990–2006. PLoS Negl Trop Dis. 2011;5(4):e1140. doi:10.1371/journal.pntd.0001140. PubMed PMID: 21541359; PubMed Central PMCID: PMCPMC3082510.
  • Ramanan P, Razonable RR. Cytomegalovirus infections in solid organ transplantation: a review. Infect Chemother. 2013;45(3):260–71. doi:10.3947/ic.2013.45.3.260. PubMed PMID: 24396627; PubMed Central PMCID: PMCPMC3848521.
  • McIntosh M, Hauschild B, Miller V. Human cytomegalovirus and transplantation: drug development and regulatory issues. J Virus Erad. 2016;2(3):143–48. PubMed PMID: 27482453; PubMed Central PMCID: PMCPMC4967965
  • Reddehase MJ. Mutual interference between cytomegalovirus and reconstitution of protective immunity after hematopoietic cell transplantation. Front Immunol. 2016;7:294. doi:10.3389/fimmu.2016.00294. PubMed PMID: 27540380; PubMed Central PMCID: PMCPMC4972816.
  • Rubin RH. The indirect effects of cytomegalovirus infection on the outcome of organ transplantation. JAMA. 1989;261(24):3607–09. PubMed PMID: 2542634
  • Kotton CN. Management of cytomegalovirus infection in solid organ transplantation. Nat Rev Nephrol. 2010;6(12):711–21. doi:10.1038/nrneph.2010.141. PubMed PMID: 20978468.
  • Beam E, Razonable RR. Cytomegalovirus in solid organ transplantation: epidemiology, prevention, and treatment. Curr Infect Dis Rep. 2012;14(6):633–41. doi:10.1007/s11908-012-0292-2. PubMed PMID: 22992839.
  • Ariza-Heredia EJ, Nesher L, Chemaly RF. Cytomegalovirus diseases after hematopoietic stem cell transplantation: a mini-review. Cancer Lett. 2014;342(1):1–8. doi:10.1016/j.canlet.2013.09.004. PubMed PMID: 24041869.
  • Marty FM, Ljungman P, Chemaly RF, Maertens J, Dadwal SS, Duarte RF, Haider S, Ullmann AJ, Katayama Y, Brown J, et al. Letermovir prophylaxis for cytomegalovirus in hematopoietic-cell transplantation. N Engl J Med. 2017;377(25):2433–44. doi:10.1056/NEJMoa1706640. PubMed PMID: 29211658.
  • Razonable RR. Management strategies for cytomegalovirus infection and disease in solid organ transplant recipients. Infect Dis Clin North Am. 2013;27(2):317–42. doi:10.1016/j.idc.2013.02.005. PubMed PMID: 23714343.
  • 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(1):4–10. doi:10.1016/j.vaccine.2013.09.042. PubMed PMID: 24129123; PubMed Central PMCID: PMCPMC4623576.
  • Pass RF, Zhang C, Evans A, Simpson T, Andrews W, Huang M-L, Corey L, Hill J, Davis E, Flanigan C, et al. Vaccine prevention of maternal cytomegalovirus infection. N Engl J Med. 2009;360(12):1191–99. doi:10.1056/NEJMoa0804749. PubMed PMID: 19297572; PubMed Central PMCID: PMCPMC2753425.
  • Heldwein EE, Krummenacher C. Entry of herpesviruses into mammalian cells. Cell Mol Life Sci. 2008;65(11):1653–68. doi:10.1007/s00018-008-7570-z. PubMed PMID: 18351291.
  • White JM, Delos SE, Brecher M, Schornberg K. Structures and mechanisms of viral membrane fusion proteins: multiple variations on a common theme. Crit Rev Biochem Mol Biol. 2008;43(3):189–219. doi:10.1080/10409230802058320. PubMed PMID: 18568847; PubMed Central PMCID: PMCPMC2649671.
  • Compton T, Nepomuceno RR, Nowlin DM. Human cytomegalovirus penetrates host cells by pH-independent fusion at the cell surface. Virology. 1992;191(1):387–95. doi:10.1016/0042-6822(92)90200-9. PubMed PMID: 1329327.
  • Ryckman BJ, Jarvis MA, Drummond DD, Nelson JA, Johnson DC. Human cytomegalovirus entry into epithelial and endothelial cells depends on genes UL128 to UL150 and occurs by endocytosis and low-pH fusion. J Virol. 2006;80(2):710–22. doi:10.1128/JVI.80.2.710-722.2006. PubMed PMID: 16378974; PubMed Central PMCID: PMCPMC1346879.
  • Backovic M, Longnecker R, Jardetzky TS. Structure of a trimeric variant of the Epstein-Barr virus glycoprotein B. Proc Natl Acad Sci U S A. 2009;106(8):2880–85. doi:10.1073/pnas.0810530106. PubMed PMID: 19196955; PubMed Central PMCID: PMCPMC2650359.
  • Heldwein EE, Lou H, Bender FC, Cohen GH, Eisenberg RJ, Harrison SC. Crystal structure of glycoprotein B from herpes simplex virus 1. Science. 2006;313(5784):217–20. doi:10.1126/science.1126548. PubMed PMID: 16840698.
  • 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(18):10023–33. doi:10.1128/JVI.78.18.10023-10033.2004. PubMed PMID: 15331735; PubMed Central PMCID: PMCPMC515016.
  • Wang D, Shenk T. Human cytomegalovirus UL131 open reading frame is required for epithelial cell tropism. J Virol. 2005;79(16):10330–38. doi:10.1128/JVI.79.16.10330-10338.2005. PubMed PMID: 16051825; PubMed Central PMCID: PMCPMC1182637.
  • Lilleri D, Kabanova A, Revello MG, Percivalle E, Sarasini A, Genini E, Sallusto F, Lanzavecchia A, Corti D, Gerna G, et al. Fetal human cytomegalovirus transmission correlates with delayed maternal antibodies to gH/gL/pUL128-130-131 complex during primary infection. PLoS One. 2013;8(3):e59863. doi:10.1371/journal.pone.0059863. PubMed PMID: 23555812; PubMed Central PMCID: PMCPMC3612069.
  • Boppana SB, Britt WJ. Antiviral antibody responses and intrauterine transmission after primary maternal cytomegalovirus infection. J Infect Dis. 1995;171(5):1115–21. PubMed PMID: 7751685
  • Lilleri D, Kabanova A, Lanzavecchia A, Gerna G. Antibodies against neutralization epitopes of human cytomegalovirus gH/gL/pUL128-130-131 complex and virus spreading may correlate with virus control in vivo. J Clin Immunol. 2012;32(6):1324–31. doi:10.1007/s10875-012-9739-3. PubMed PMID: 22836657.
  • Fornara C, Furione M, Arossa A, Gerna G, Lilleri D. Comparative magnitude and kinetics of human cytomegalovirus-specific CD4+ and CD8+ T-cell responses in pregnant women with primary versus remote infection and in transmitting versus non-transmitting mothers: its utility for dating primary infection in pregnancy. J Med Virol. 2016;88(7):1238–46. doi:10.1002/jmv.24449. PubMed PMID: 26680747.
  • Nigro G, Adler SP, La Torre R, Best AM, Congenital Cytomegalovirus Collaborating G. Passive immunization during pregnancy for congenital cytomegalovirus infection. N Engl J Med. 2005;353(13):1350–62. doi:10.1056/NEJMoa043337. PubMed PMID: 16192480.
  • Revello MG, Lazzarotto T, Guerra B, Spinillo A, Ferrazzi E, Kustermann A, Guaschino S, Vergani P, Todros T, Frusca T, et al. A randomized trial of hyperimmune globulin to prevent congenital cytomegalovirus. N Engl J Med. 2014;370(14):1316–26. doi:10.1056/NEJMoa1310214. PubMed PMID: 24693891.
  • Bialas KM, Tanaka T, Tran D, Varner V, Cisneros De La Rosa E, Chiuppesi F, Wussow F, Kattenhorn L, Macri S, Kunz EL, et al. Maternal CD4+ T cells protect against severe congenital cytomegalovirus disease in a novel nonhuman primate model of placental cytomegalovirus transmission. Proc Natl Acad Sci USA. 2015;112(44):13645–50. doi:10.1073/pnas.1511526112. PubMed PMID: 26483473; PubMed Central PMCID: PMCPMC4640765.
  • Nelson CS, Cruz DV, Tran D, Bialas KM, Stamper L, Wu H, Gilbert M, Blair R, Alvarez X, Itell H, et al. Preexisting antibodies can protect against congenital cytomegalovirus infection in monkeys. JCI Insight. 2017;2(13). doi:10.1172/jci.insight.94002. PubMed PMID: 28679960; PubMed Central PMCID: PMCPMC5499366.
  • Schleiss MR, McVoy MA. Guinea Pig Cytomegalovirus (GPCMV): a model for the study of the prevention and treatment of maternal-fetal transmission. Future Virol. 2010;5(2):207–17. doi:10.2217/fvl.10.8. PubMed PMID: 23308078; PubMed Central PMCID: PMCPMC3539792.
  • Auerbach MR, Yan D, Vij R, Hongo J-A, Nakamura G, Vernes J-M, Meng YG, Lein S, Chan P, Ross J, et al. A neutralizing anti-gH/gL monoclonal antibody is protective in the guinea pig model of congenital CMV infection. PLoS Pathog. 2014;10(4):e1004060. doi:10.1371/journal.ppat.1004060. PubMed PMID: 24722349; PubMed Central PMCID: PMCPMC3983071.
  • Schleiss MR, Choi KY, Anderson J, Mash JG, Wettendorff M, Mossman S, Van Damme M. Glycoprotein B (gB) vaccines adjuvanted with AS01 or AS02 protect female guinea pigs against cytomegalovirus (CMV) viremia and offspring mortality in a CMV-challenge model. Vaccine. 2014;32(23):2756–62. doi:10.1016/j.vaccine.2013.07.010. PubMed PMID: 23867012; PubMed Central PMCID: PMCPMC3894257.
  • Chatterjee A, Harrison CJ, Britt WJ, Bewtra C. Modification of maternal and congenital cytomegalovirus infection by anti-glycoprotein b antibody transfer in guinea pigs. J Infect Dis. 2001;183(11):1547–53. doi:10.1086/320714. PubMed PMID: 11343203.
  • Gratama JW, Boeckh M, Nakamura R, Cornelissen JJ, Brooimans RA, Zaia JA, Forman SJ, Gaal K, Bray KR, Gasior GH, et al. Immune monitoring with iTAg MHC Tetramers for prediction of recurrent or persistent cytomegalovirus infection or disease in allogeneic hematopoietic stem cell transplant recipients: a prospective multicenter study. Blood. 2010;116(10):1655–62. doi:10.1182/blood-2010-03-273508. PubMed PMID: 20508161.
  • Zhou W, Longmate J, Lacey SF, Palmer JM, Gallez-Hawkins G, Thao L, Spielberger R, Nakamura R, Forman SJ, Zaia JA, et al. Impact of donor CMV status on viral infection and reconstitution of multifunction CMV-specific T cells in CMV-positive transplant recipients. Blood. 2009;113(25):6465–76. doi:10.1182/blood-2009-02-203307. PubMed PMID: 19369230; PubMed Central PMCID: PMCPMC2710937.
  • Schleiss MR, Permar SR, Plotkin SA, Papasian CJ. Progress toward Development of a Vaccine against Congenital Cytomegalovirus Infection. Clin Vaccine Immunol. 2017;24(12). doi:10.1128/CVI.00268-17. PubMed PMID: 29046308; PubMed Central PMCID: PMCPMC5717185.
  • Diamond DJ, La Rosa C, Chiuppesi F, Contreras H, Dadwal S, Wussow F, Bautista S, Nakamura R, Zaia JA. A fifty-year odyssey: prospects for a cytomegalovirus vaccine in transplant and congenital infection. Expert Rev Vaccines. 2018;17(10):889–911. doi:10.1080/14760584.2018.1526085. PubMed PMID: 30246580.
  • Nelson CS, Herold BC, Permar SR. A new era in cytomegalovirus vaccinology: considerations for rational design of next-generation vaccines to prevent congenital cytomegalovirus infection. NPJ Vaccines. 2018;3:38. doi:10.1038/s41541-018-0074-4. PubMed PMID: 30275984; PubMed Central PMCID: PMCPMC6148244 preclinical HCMV vaccine programs. B.C.H. is an inventor on a pending patent application for a delta gD-2 vaccine. The other authors have no conflicts of interest to declare.
  • 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(9773):1256–63. doi:10.1016/S0140-6736(11)60136-0. PubMed PMID: 21481708; PubMed Central PMCID: PMCPMC3075549.
  • Bernstein DI, Munoz FM, Callahan ST, Rupp R, Wootton SH, Edwards KM, Turley CB, Stanberry LR, Patel SM, Mcneal MM, et al. Safety and efficacy of a cytomegalovirus glycoprotein B (gB) vaccine in adolescent girls: A randomized clinical trial. Vaccine. 2016;34(3):313–19. doi:10.1016/j.vaccine.2015.11.056. PubMed PMID: 26657184; PubMed Central PMCID: PMCPMC4701617.
  • Nelson CS, Huffman T, Jenks JA, Cisneros de la Rosa E, Xie G, Vandergrift N, Pass RF, Pollara J, Permar SR. HCMV glycoprotein B subunit vaccine efficacy mediated by nonneutralizing antibody effector functions. Proc Natl Acad Sci U S A. 2018;115(24):6267–72. doi:10.1073/pnas.1800177115. PubMed PMID: 29712861; PubMed Central PMCID: PMCPMC6004431.
  • Cui X, Cao Z, Wang S, Lee RB, Wang X, Murata H, Adler SP, McVoy MA, Snapper CM. Novel trimeric human cytomegalovirus glycoprotein B elicits a high-titer neutralizing antibody response. Vaccine. 2018;36(37):5580–90. doi:10.1016/j.vaccine.2018.07.056. PubMed PMID: 30082162.
  • Kabanova A, Perez L, Lilleri D, Marcandalli J, Agatic G, Becattini S, Preite S, Fuschillo D, Percivalle E, Sallusto F, et al. Antibody-driven design of a human cytomegalovirus gHgLpUL128L subunit vaccine that selectively elicits potent neutralizing antibodies. Proc Natl Acad Sci U S A. 2014;111(50):17965–70. doi:10.1073/pnas.1415310111. PubMed PMID: 25453106; PubMed Central PMCID: PMCPMC4273412.
  • Chiuppesi F, Wussow F, Scharf L, Contreras H, Gao H, Meng Z, Nguyen J, Barry PA, Bjorkman PJ, Diamond DJ, et al. Comparison of homologous and heterologous prime-boost vaccine approaches using Modified Vaccinia Ankara and soluble protein to induce neutralizing antibodies by the human cytomegalovirus pentamer complex in mice. PLoS One. 2017;12(8):e0183377. doi:10.1371/journal.pone.0183377. PubMed PMID: 28813507; PubMed Central PMCID: PMCPMC5558987.
  • 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(362):362ra145. doi:10.1126/scitranslmed.aaf9387. PubMed PMID: 27797961.
  • Link EK, Brandmüller C, Suezer Y, Ameres S, Volz A, Moosmann A, Sutter G, Lehmann MH. A synthetic human cytomegalovirus pp65-IE1 fusion antigen efficiently induces and expands virus specific T cells. Vaccine. 2017;35(38):5131–39. doi:10.1016/j.vaccine.2017.08.019. PubMed PMID: 28818566.
  • La Rosa C, Longmate J, Martinez J, Zhou Q, Kaltcheva TI, Tsai W, Drake J, Carroll M, Wussow F, Chiuppesi F, et al. MVA vaccine encoding CMV antigens safely induces durable expansion of CMV-specific T cells in healthy adults. Blood. 2017;129(1):114–25. doi:10.1182/blood-2016-07-729756. PubMed PMID: 27760761; PubMed Central PMCID: PMCPMC5216266.
  • Chiuppesi F, Nguyen J, Park S, Contreras H, Kha M, Meng Z, Yekwa E, Barrot L, Barron S, Vallve A, et al. Multiantigenic modified vaccinia virus ankara vaccine vectors to elicit potent humoral and cellular immune reponses against human cytomegalovirus in mice. J Virol. 2018;92(19). doi:10.1128/JVI.01012-18. PubMed PMID: 30045984; PubMed Central PMCID: PMCPMC6146800.
  • Kirchmeier M, Fluckiger A-C, Soare C, Bozic J, Ontsouka B, Ahmed T, Diress A, Pereira L, Schödel F, Plotkin S, et al. Enveloped virus-like particle expression of human cytomegalovirus glycoprotein B antigen induces antibodies with potent and broad neutralizing activity. Clin Vaccine Immunol. 2014;21(2):174–80. doi:10.1128/CVI.00662-13. PubMed PMID: 24334684; PubMed Central PMCID: PMCPMC3910943.
  • Schleiss MR. Cytomegalovirus vaccines under clinical development. J Virus Erad. 2016;2(4):198–207. PubMed PMID: 27781101; PubMed Central PMCID: PMCPMC5075346
  • Cytomegalovirus infection: advancing strategies for prevention and treatment [Internet]. NIAID/NICHD/FDA meeting, Bethesda (MD); 2018 Sep 4–6.
  • Geall AJ, Verma A, Otten GR, Shaw CA, Hekele A, Banerjee K, Cu Y, Beard CW, Brito LA, Krucker T, et al. Nonviral delivery of self-amplifying RNA vaccines. Proc Natl Acad Sci USA. 2012;109(36):14604–09. doi:10.1073/pnas.1209367109. PubMed PMID: 22908294; PubMed Central PMCID: PMCPMC3437863.
  • Brito LA, Chan M, Shaw CA, Hekele A, Carsillo T, Schaefer M, Archer J, Seubert A, Otten GR, Beard CW, et al. A cationic nanoemulsion for the delivery of next-generation RNA vaccines. Mol Ther. 2014;22(12):2118–29. doi:10.1038/mt.2014.133. PubMed PMID: 25027661; PubMed Central PMCID: PMCPMC4429691.
  • Fu T-M, An Z, Wang D. Progress on pursuit of human cytomegalovirus vaccines for prevention of congenital infection and disease. Vaccine. 2014;32(22):2525–33. doi:10.1016/j.vaccine.2014.03.057. PubMed PMID: 24681264.
  • Rieder F, Steininger C. Cytomegalovirus vaccine: phase II clinical trial results. Clin Microbiol Infect. 2014;20 Suppl 5:95–102. doi:10.1111/1469-0691.12449. PubMed PMID: 24283990; PubMed Central PMCID: PMCPMC5716458.
  • 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(4):970–75. doi:10.1086/315022. PubMed PMID: 10479120.
  • 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(11):1534–41. doi:10.1093/infdis/jir138. PubMed PMID: 21592981; PubMed Central PMCID: PMCPMC3096785.
  • Spaete RR. A recombinant subunit vaccine approach to HCMV vaccine development. Transplant Proc. 1991;23(3 Suppl 3):90–96. PubMed PMID: 1648843
  • Sharma S, Wisner TW, Johnson DC, Heldwein EE. HCMV gB shares structural and functional properties with gB proteins from other herpesviruses. Virology. 2013;435(2):239–49. doi:10.1016/j.virol.2012.09.024. PubMed PMID: 23089254; PubMed Central PMCID: PMCPMC3534942.
  • Singh J, Compton T. Characterization of a panel of insertion mutants in human cytomegalovirus glycoprotein B. J Virol. 2000;74(3):1383–92. PubMed PMID: 10627549; PubMed Central PMCID: PMCPMC111473
  • Britt WJ, Vugler LG. Processing of the gp55-116 envelope glycoprotein complex (gB) of human cytomegalovirus. J Virol. 1989;63(1):403–10. PubMed PMID: 2535741; PubMed Central PMCID: PMCPMC247697
  • Britt WJ, Auger D. Synthesis and processing of the envelope gp55-116 complex of human cytomegalovirus. J Virol. 1986;58(1):185–91. PubMed PMID: 3005648; PubMed Central PMCID: PMCPMC252892
  • Britt WJ, Vugler LG. Oligomerization of the human cytomegalovirus major envelope glycoprotein complex gB (gp55–116). J Virol. 1992;66(11):6747–54. PubMed PMID: 1328688; PubMed Central PMCID: PMCPMC240171
  • Billstrom MA, Britt WJ. Postoligomerization folding of human cytomegalovirus glycoprotein B: identification of folding intermediates and importance of disulfide bonding. J Virol. 1995;69(11):7015–22. PubMed PMID: 7474121; PubMed Central PMCID: PMCPMC189621
  • Lopper M, Compton T. Disulfide bond configuration of human cytomegalovirus glycoprotein B. J Virol. 2002;76(12):6073–82. PubMed PMID: 12021340; PubMed Central PMCID: PMCPMC136243
  • Lopper M, Compton T. Coiled-coil domains in glycoproteins B and H are involved in human cytomegalovirus membrane fusion. J Virol. 2004;78(15):8333–41. doi:10.1128/JVI.78.15.8333-8341.2004. PubMed PMID: 15254205; PubMed Central PMCID: PMCPMC446119.
  • Burke HG, Heldwein EE, Rey FA. Crystal Structure of the Human Cytomegalovirus Glycoprotein B. PLoS Pathog. 2015;11(10):e1005227. doi:10.1371/journal.ppat.1005227. PubMed PMID: 26484870; PubMed Central PMCID: PMCPMC4617298.
  • Chandramouli S, Ciferri C, Nikitin PA, Caló S, Gerrein R, Balabanis K, Monroe J, Hebner C, Lilja AE, Settembre EC, et al. Structure of HCMV glycoprotein B in the postfusion conformation bound to a neutralizing human antibody. Nat Commun. 2015;6:8176. doi:10.1038/ncomms9176. PubMed PMID: 26365435; PubMed Central PMCID: PMCPMC4579600.
  • Britt WJ, Vugler L, Stephens EB. Induction of complement-dependent and -independent neutralizing antibodies by recombinant-derived human cytomegalovirus gp55–116 (gB). J Virol. 1988;62(9):3309–18. PubMed PMID: 2841483; PubMed Central PMCID: PMCPMC253452
  • Wen Y, Monroe J, Linton C, Archer J, Beard CW, Barnett SW, Palladino G, Mason PW, Carfi A, Lilja AE. Human cytomegalovirus gH/gL/UL128/UL130/UL131A complex elicits potently neutralizing antibodies in mice. Vaccine. 2014;32(30):3796–804. doi:10.1016/j.vaccine.2014.05.004. PubMed PMID: 24837507.
  • Pass RF. Development and evidence for efficacy of CMV glycoprotein B vaccine with MF59 adjuvant. J Clin Virol. 2009;46 Suppl 4:S73–6. doi:10.1016/j.jcv.2009.07.002. PubMed PMID: 19647480; PubMed Central PMCID: PMCPMC2805195.
  • Li F, Freed DC, Tang A, Rustandi RR, Troutman MC, Espeseth AS, Zhang N, An Z, McVoy M, Zhu H, et al. Complement enhances in vitro neutralizing potency of antibodies to human cytomegalovirus glycoprotein B (gB) and immune sera induced by gB/MF59 vaccination. NPJ Vaccines. 2017;2:36. doi:10.1038/s41541-017-0038-0. PubMed PMID: 29263890; PubMed Central PMCID: PMCPMC5730571 of Merck & Co., Inc. and as such receiving salaries and benefits from the company.
  • Baraniak I, Kropff B, Ambrose L, McIntosh M, McLean GR, Pichon S, Atkinson C, Milne RSB, Mach M, Griffiths PD, et al. Protection from cytomegalovirus viremia following glycoprotein B vaccination is not dependent on neutralizing antibodies. Proc Natl Acad Sci U S A. 2018;115(24):6273–78. doi:10.1073/pnas.1800224115. PubMed PMID: 29686064; PubMed Central PMCID: PMCPMC6004462.
  • Wang D, Li F, Freed DC, Finnefrock AC, Tang A, Grimes SN, Casimiro DR, Fu T-M. Quantitative analysis of neutralizing antibody response to human cytomegalovirus in natural infection. Vaccine. 2011;29(48):9075–80. doi:10.1016/j.vaccine.2011.09.056. PubMed PMID: 21945962.
  • Fouts AE, Chan P, Stephan J-P, 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(13):7444–47. doi:10.1128/JVI.00467-12. PubMed PMID: 22532696; PubMed Central PMCID: PMCPMC3416310.
  • 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(51):E4997–5005. doi:10.1073/pnas.1316517110. PubMed PMID: 24297878; PubMed Central PMCID: PMCPMC3870741.
  • 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(2):1005–13. doi:10.1128/JVI.01809-09. PubMed PMID: 19889756; PubMed Central PMCID: PMCPMC2798344.
  • Wussow F, Chiuppesi F, Martinez J, Campo J, Johnson E, Flechsig C, Newell M, Tran E, Ortiz J, La Rosa C, et al. Human cytomegalovirus vaccine based on the envelope gH/gL pentamer complex. PLoS Pathog. 2014;10(11):e1004524. doi:10.1371/journal.ppat.1004524. PubMed PMID: 25412505; PubMed Central PMCID: PMCPMC4239111.
  • Zydek M, Petitt M, Fang-Hoover J, Adler B, Kauvar LM, Pereira L, Tabata T. HCMV infection of human trophoblast progenitor cells of the placenta is neutralized by a human monoclonal antibody to glycoprotein B and not by antibodies to the pentamer complex. Viruses. 2014;6(3):1346–64. doi:10.3390/v6031346. PubMed PMID: 24651029; PubMed Central PMCID: PMCPMC3970154.
  • Chiuppesi F, Wussow F, Johnson E, Bian C, Zhuo M, Rajakumar A, Barry PA, Britt WJ, Chakraborty R, Diamond DJ. Vaccine-Derived Neutralizing Antibodies to the Human Cytomegalovirus gH/gL Pentamer Potently Block Primary Cytotrophoblast Infection. J Virol. 2015;89(23):11884–98. doi:10.1128/JVI.01701-15. PubMed PMID: 26378171; PubMed Central PMCID: PMCPMC4645301.
  • Bootz A, Karbach A, Spindler J, Kropff B, Reuter N, Sticht H, Winkler TH, Britt WJ, Mach M, Permar SR. Protective capacity of neutralizing and non-neutralizing antibodies against glycoprotein B of cytomegalovirus. PLoS Pathog. 2017;13(8):e1006601. doi:10.1371/journal.ppat.1006601. PubMed PMID: 28854233; PubMed Central PMCID: PMCPMC5595347.
  • Banaszynski LA, Chen L-C, Maynard-Smith LA, Ooi AG, Wandless TJ. A rapid, reversible, and tunable method to regulate protein function in living cells using synthetic small molecules. Cell. 2006;126(5):995–1004. doi:10.1016/j.cell.2006.07.025. PubMed PMID: 16959577; PubMed Central PMCID: PMCPMC3290523.
  • Glass M, Busche A, Wagner K, Messerle M, Borst EM. Conditional and reversible disruption of essential herpesvirus proteins. Nat Methods. 2009;6(8):577–79. doi:10.1038/nmeth.1346. PubMed PMID: 19578384.
  • Borst EM, Kleine-Albers J, Gabaev I, Babic M, Wagner K, Binz A, Degenhardt I, Kalesse M, Jonjic S, Bauerfeind R, et al. The human cytomegalovirus UL51 protein is essential for viral genome cleavage-packaging and interacts with the terminase subunits pUL56 and pUL89. J Virol. 2013;87(3):1720–32. doi:10.1128/JVI.01955-12. PubMed PMID: 23175377; PubMed Central PMCID: PMCPMC3554196.
  • 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(3):843–46. doi:10.1086/314951. PubMed PMID: 10438376.
  • 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(8):1171–79. doi:10.1086/319680. PubMed PMID: 11262198.
  • Bernstein DI, Schleiss MR, Berencsi K, Gonczol E, Dickey M, Khoury P, Cadoz M, Meric C, Zahradnik J, Duliege A-M, et al. 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(5):686–90. doi:10.1086/339003. PubMed PMID: 11865427.
  • 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(42):7441–49. doi:10.1016/j.vaccine.2007.08.016. PubMed PMID: 17870214; PubMed Central PMCID: PMCPMC2744093.
  • Reap EA, Dryga SA, Morris J, Rivers B, Norberg PK, Olmsted RA, Chulay JD. Cellular and humoral immune responses to alphavirus replicon vaccines expressing cytomegalovirus pp65, IE1, and gB proteins. Clin Vaccine Immunol. 2007;14(6):748–55. doi:10.1128/CVI.00037-07. PubMed PMID: 17442845; PubMed Central PMCID: PMCPMC1951075.
  • Bernstein DI, Reap EA, Katen K, Watson A, Smith K, Norberg P, Olmsted RA, Hoeper A, Morris J, Negri S, et al. Randomized, double-blind, Phase 1 trial of an alphavirus replicon vaccine for cytomegalovirus in CMV seronegative adult volunteers. Vaccine. 2009;28(2):484–93. doi:10.1016/j.vaccine.2009.09.135. PubMed PMID: 19857446.
  • 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(6):919–26. doi:10.1016/j.vaccine.2012.12.009. PubMed PMID: 23246547.
  • Flatz L, Hegazy AN, Bergthaler A, Verschoor A, Claus C, Fernandez M, Gattinoni L, Johnson S, Kreppel F, Kochanek S, et al. Development of replication-defective lymphocytic choriomeningitis virus vectors for the induction of potent CD8+ T cell immunity. Nat Med. 2010;16(3):339–45. doi:10.1038/nm.2104. PubMed PMID: 20139992; PubMed Central PMCID: PMCPMC3247638.
  • Cardin RD, Bravo FJ, Pullum DA, Orlinger K, Watson EM, Aspoeck A, Fuhrmann G, Guirakhoo F, Monath T, Bernstein DI. Replication-defective lymphocytic choriomeningitis virus vectors expressing guinea pig cytomegalovirus gB and pp65 homologs are protective against congenital guinea pig cytomegalovirus infection. Vaccine. 2016;34(17):1993–99. doi:10.1016/j.vaccine.2016.03.005. PubMed PMID: 26973071.
  • Schleiss MR, Berka U, Watson E, Aistleithner M, Kiefmann B, Mangeat B, Swanson EC, Gillis PA, Hernandez-Alvarado N, Fernández-Alarcón C, et al. Additive Protection against Congenital Cytomegalovirus Conferred by Combined Glycoprotein B/pp65 Vaccination Using a Lymphocytic Choriomeningitis Virus Vector. Clin Vaccine Immunol. 2017;24(1). doi:10.1128/CVI.00300-16. PubMed PMID: 27795301; PubMed Central PMCID: PMCPMC5216435.
  • Cottingham MG, Carroll MW. Recombinant MVA vaccines: dispelling the myths. Vaccine. 2013;31(39):4247–51. doi:10.1016/j.vaccine.2013.03.021. PubMed PMID: 23523407.
  • Gillis PA, Hernandez-Alvarado N, Gnanandarajah JS, Wussow F, Diamond DJ, Schleiss MR. Development of a novel, guinea pig-specific IFN-gamma ELISPOT assay and characterization of guinea pig cytomegalovirus GP83-specific cellular immune responses following immunization with a modified vaccinia virus Ankara (MVA)-vectored GP83 vaccine. Vaccine. 2014;32(31):3963–70. doi:10.1016/j.vaccine.2014.05.011. PubMed PMID: 24856783; PubMed Central PMCID: PMCPMC4279957.
  • Wussow F, Yue Y, Martinez J, Deere JD, Longmate J, Herrmann A, Barry PA, Diamond DJ. A vaccine based on the rhesus cytomegalovirus UL128 complex induces broadly neutralizing antibodies in rhesus macaques. J Virol. 2013;87(3):1322–32. doi:10.1128/JVI.01669-12. PubMed PMID: 23152525; PubMed Central PMCID: PMCPMC3554163.
  • Swanson EC, Gillis P, Hernandez-Alvarado N, Fernández-Alarcón C, Schmit M, Zabeli JC, Wussow F, Diamond DJ, Schleiss MR. Comparison of monovalent glycoprotein B with bivalent gB/pp65 (GP83) vaccine for congenital cytomegalovirus infection in a guinea pig model: inclusion of GP83 reduces gB antibody response but both vaccine approaches provide equivalent protection against pup mortality. Vaccine. 2015;33(32):4013–18. doi:10.1016/j.vaccine.2015.06.019. PubMed PMID: 26079615; PubMed Central PMCID: PMCPMC4772145.
  • Ura T, Okuda K, Shimada M. Developments in Viral Vector-Based Vaccines. Vaccines (Basel). 2014;2(3):624–41. doi:10.3390/vaccines2030624. PubMed PMID: 26344749; PubMed Central PMCID: PMCPMC4494222.
  • 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(4):290–99. doi:10.1016/S1473-3099(11)70344-9. PubMed PMID: 22237175.
  • Selinsky C, Luke C, Wloch M, Geall A, Hermanson G, Kaslow D, Evans T. A DNA-based vaccine for the prevention of human cytomegalovirus-associated diseases. Hum Vaccin. 2005;1(1):16–23. PubMed PMID: 17038834
  • Wloch MK, Smith LR, Boutsaboualoy S, Reyes L, Han C, Kehler J, Smith HD, Selk L, Nakamura R, Brown JM, et al. Safety and immunogenicity of a bivalent cytomegalovirus DNA vaccine in healthy adult subjects. J Infect Dis. 2008;197(12):1634–42. doi:10.1086/588385. PubMed PMID: 18444883; PubMed Central PMCID: PMCPMC2956065.
  • Mori T, Kanda Y, Takenaka K, Okamoto S, Kato J, Kanda J, Yoshimoto G, Gondo H, Doi S, Inaba M, et al. Safety of ASP0113, a cytomegalovirus DNA vaccine, in recipients undergoing allogeneic hematopoietic cell transplantation: an open-label phase 2 trial. Int J Hematol. 2017;105(2):206–12. doi:10.1007/s12185-016-2110-3. 10.1007/s12185-016-2110-3. PubMed PMID: 27796740.
  • 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(10):1540–48. doi:10.1016/j.vaccine.2009.01.006. PubMed PMID: 19168107.
  • Sullivan SM, Doukas J, Hartikka J, Smith L, Rolland A. Vaxfectin: a versatile adjuvant for plasmid DNA- and protein-based vaccines. Expert Opin Drug Deliv. 2010;7(12):1433–46. doi:10.1517/17425247.2010.538047. PubMed PMID: 21118032.
  • McVoy MA, Lee R, Saccoccio FM, Hartikka J, Smith LR, Mahajan R, Wang JB, Cui X, Adler SP. A cytomegalovirus DNA vaccine induces antibodies that block viral entry into fibroblasts and epithelial cells. Vaccine. 2015;33(51):7328–36. doi:10.1016/j.vaccine.2015.10.078. PubMed PMID: 26597035; PubMed Central PMCID: PMCPMC4684450.
  • Vincenti F, Budde K, Merville P, Shihab F, Ram Peddi V, Shah M, Wyburn K, Cassuto-Viguier E, Weidemann A, Lee M, et al. A randomized, phase 2 study of ASP0113, a DNA-based vaccine, for the prevention of CMV in CMV-seronegative kidney transplant recipients receiving a kidney from a CMV-seropositive donor. Am J Transplant. 2018;18(12):2945–54. doi:10.1111/ajt.14925. PubMed PMID: 29745007.
  • Li L, Petrovsky N. Molecular mechanisms for enhanced DNA vaccine immunogenicity. Expert Rev Vaccines. 2016;15(3):313–29. doi:10.1586/14760584.2016.1124762. PubMed PMID: 26707950; PubMed Central PMCID: PMCPMC4955855.
  • John S, Yuzhakov O, Woods A, Deterling J, Hassett K, Shaw CA, Ciaramella G. Multi-antigenic human cytomegalovirus mRNA vaccines that elicit potent humoral and cell-mediated immunity. Vaccine. 2018;36(12):1689–99. doi:10.1016/j.vaccine.2018.01.029. PubMed PMID: 29456015.
  • Bahl K, Senn JJ, Yuzhakov O, Bulychev A, Brito LA, Hassett KJ, Laska ME, Smith M, Almarsson Ö, Thompson J, et al. Preclinical and Clinical Demonstration of Immunogenicity by mRNA Vaccines against H10N8 and H7N9 Influenza Viruses. Mol Ther. 2017;25(6):1316–27. doi:10.1016/j.ymthe.2017.03.035. PubMed PMID: 28457665; PubMed Central PMCID: PMCPMC5475249.
  • Alberer M, Gnad-Vogt U, Hong HS, Mehr KT, Backert L, Finak G, Gottardo R, Bica MA, Garofano A, Koch SD, et al. Safety and immunogenicity of a mRNA rabies vaccine in healthy adults: an open-label, non-randomised, prospective, first-in-human phase 1 clinical trial. Lancet. 2017;390(10101):1511–20. doi:10.1016/S0140-6736(17)31665-3. PubMed PMID: 28754494.
  • Liu MA, Ulmer JB. Human clinical trials of plasmid DNA vaccines. Adv Genet. 2005;55:25–40. doi:10.1016/S0065-2660(05)55002-8. PubMed PMID: 16291211.
  • Pardi N, Hogan MJ, Porter FW, Weissman D. mRNA vaccines - a new era in vaccinology. Nat Rev Drug Discov. 2018;17(4):261–79. doi:10.1038/nrd.2017.243. PubMed PMID: 29326426; PubMed Central PMCID: PMCPMC5906799.
  • Leruez-Ville M, Magny J-F, Couderc S, Pichon C, Parodi M, Bussières L, Guilleminot T, Ghout I, Ville Y. Risk factors for congenital cytomegalovirus infection following primary and nonprimary maternal infection: a prospective neonatal screening study using polymerase chain reaction in saliva. Clin Infect Dis. 2017;65(3):398–404. doi:10.1093/cid/cix337. PubMed PMID: 28419213.
  • Simonazzi G, Curti A, Cervi F, Gabrielli L, Contoli M, Capretti MG, Rizzo N, Guerra B, Farina A, Lazzarotto T. Perinatal outcomes of non-primary maternal cytomegalovirus infection: A 15-year experience. Fetal Diagn Ther. 2018;43(2):138–42. doi:10.1159/000477168. PubMed PMID: 28697499.
  • Picone O, Vauloup-Fellous C, Cordier AG, Guitton S, Senat MV, Fuchs F, Ayoubi JM, Grangeot Keros L, Benachi A. A series of 238 cytomegalovirus primary infections during pregnancy: description and outcome. Prenat Diagn. 2013;33(8):751–58. doi:10.1002/pd.4118. PubMed PMID: 23553686.
  • Enders G, Daiminger A, Bäder U, Exler S, Enders M. Intrauterine transmission and clinical outcome of 248 pregnancies with primary cytomegalovirus infection in relation to gestational age. J Clin Virol. 2011;52(3):244–46. doi:10.1016/j.jcv.2011.07.005. PubMed PMID: 21820954.
  • 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(3):332–36. doi:10.1016/j.jpeds.2005.09.003. PubMed PMID: 16615962.
  • Pötzsch S, Spindler N, Wiegers A-K, Fisch T, Rücker 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(8):e1002172. doi:10.1371/journal.ppat.1002172. PubMed PMID: 21852946; PubMed Central PMCID: PMCPMC3154849.
  • Saccoccio FM, Sauer AL, Cui X, Armstrong AE, Habib El E-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(15):2705–11. doi:10.1016/j.vaccine.2011.01.079. PubMed PMID: 21310190; PubMed Central PMCID: PMCPMC3084484.