Next Article in Journal
Co-Delivery Effect of CD24 on the Immunogenicity and Lethal Challenge Protection of a DNA Vector Expressing Nucleocapsid Protein of Crimean Congo Hemorrhagic Fever Virus
Next Article in Special Issue
Enhanced Ability of Oligomeric Nanobodies Targeting MERS Coronavirus Receptor-Binding Domain
Previous Article in Journal / Special Issue
Characterization of the Lipidomic Profile of Human Coronavirus-Infected Cells: Implications for Lipid Metabolism Remodeling upon Coronavirus Replication
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Middle East Respiratory Syndrome Vaccine Candidates: Cautious Optimism

Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, 77555 TX, USA
*
Author to whom correspondence should be addressed.
Submission received: 18 December 2018 / Revised: 10 January 2019 / Accepted: 12 January 2019 / Published: 17 January 2019
(This article belongs to the Special Issue MERS-CoV)

Abstract

:
Efforts towards developing a vaccine for Middle East respiratory syndrome coronavirus (MERS-CoV) have yielded promising results. Utilizing a variety of platforms, several vaccine approaches have shown efficacy in animal models and begun to enter clinical trials. In this review, we summarize the current progress towards a MERS-CoV vaccine and highlight potential roadblocks identified from previous attempts to generate coronavirus vaccines.

1. Introduction

In recent years, viral zoonotic diseases have caused outbreaks marked by rapid spread and high mortality, including the 2002 emergence of severe acute respiratory syndrome coronavirus (SARS-CoV) [1], the 2009 H1N1 swine flu pandemic [2], and the 2013 Ebola outbreak in West Africa [3]. Such outbreaks are difficult to predict as new strains emerge or reemerge from zoonotic reservoirs [4]. Coronaviruses (CoVs), large positive-stranded RNA viruses of the order Nidovirales [5], were considered minor human pathogens, causing cold-like symptoms and occasionally associated with pneumonia and more severe disease [6]. However, the emergence of SARS-CoV in 2002 and Middle East respiratory syndrome coronavirus (MERS-CoV) in 2012, members of βCoV lineages B and C, respectively, marked a shift in our understanding of the pathogenic potential of coronaviruses [7]. As these more virulent viruses are genetically similar to those currently circulating in bats [8,9], CoVs may pose a threat for future zoonoses [10].
Since the emergence of MERS-CoV in Saudi Arabia in 2012, over 2200 confirmed cases have been reported in at least 27 countries, with an overall mortality rate of 35% (https://www.who.int/emergencies/mers-cov). Additionally, more severe disease has been noted in the aged, immunocompromised, and those with chronic health conditions [11]. Camels, which show seropositivity to MERS-CoV in archived sera dating back to 1983 [12], serve as intermediate hosts and are able to spread the virus to humans [13], who may then spread the infection person-to-person [14]. While a range of therapeutics have been explored for CoV disease [15,16,17], a MERS-CoV vaccine remains the most scalable, cost-effective prophylactic measure. Currently, a vaccine for MERS-CoV is not available, although several candidates have been developed using a variety of approaches. Vaccine studies were initially hampered by a lack of small animal models of MERS-CoV disease [18]. While rodents possess homologues for dipeptidyl peptidase 4 (DPP4), the human receptor for MERS-CoV [19], rodent DPP4 homologues are incompatible with MERS-CoV infection [20,21,22]. However, several in vivo approaches have been developed to overcome these barriers and facilitate MERS-CoV vaccine testing in small animal models [23,24,25,26,27].
Preclinical vaccine development for both SARS-CoV and MERS-CoV has largely aimed to stimulate a robust immune response against the viral envelope-protruding spike (S) glycoprotein [28,29], a class I fusion protein, and/or the nucleocapsid (N) protein [30,31]. MERS-CoV S is proteolytically cleaved by host furin [32] during maturation into an S1 domain responsible for binding to DPP4 as well as an S2 domain containing two heptad-repeat regions that facilitate membrane fusion (Figure 1). The S1 domain can be further divided into the N-terminal domain (NTD), or S1A, associated with binding sialic acid [33], and the receptor binding domain (RBD), comprising the majority of the C-terminal domain of S1. Cryo-electron microscopy studies have shown that the RBD is flexible and opens upward or away from the viral envelope in order to establish contact with DPP4, which may expose S2’ [34], a second protease cleavage site within S2. Cleavage at S2’ is necessary for membrane fusion upon viral entry [32]. The centrality of S to viral entry helps explain why antibodies that target it are potently neutralizing [35]. On the other hand, while CoV N proteins are abundantly expressed during infection [36], immunization with SARS-CoV N did not induce strongly neutralizing antibodies [37], likely because N is not displayed on the viral surface. However, N is more conserved than S within CoV lineage [38], and vaccination with SARS-CoV N was shown to induce cytotoxic T cell responses in mice [39]. Therefore, N may help induce cell-mediated immunity to CoV infection [40], as may S [41]. CD4+ and CD8+ T cell responses from recovered MERS-CoV patients were particularly strong towards N peptides [42]. However, vaccination with N-based immunogens may carry risks associated with Th2-related eosinophilic immune enhancement [43], as may S-based vaccines [44]. Notwithstanding, because of the protection afforded by the robust immune response it generates, S has been the target of most vaccine candidates for MERS-CoV. In this review, we summarize the current state of MERS-CoV vaccine candidates and also describe potential barriers to MERS-CoV vaccine efficacy that first surfaced during research on developing a SARS-CoV vaccine.

2. Subunit Vaccines: Immunogenically Focused

Subunit vaccines comprise one or more immunogenic components derived from a pathogen [47]. They have gained popularity in recent decades due to the relative ease of their production and their reduced risks in vivo compared to vaccine types that involve live virus, namely live attenuated vaccines, viral vector vaccines, and even improperly prepared inactivated vaccines.

2.1. Receptor Binding Domain

It is known from studies of recovered SARS-CoV patients that antibodies generated against the receptor binding domain (RBD) are both long-lasting (>3 years) and neutralizing [48]. The RBD in the MERS-CoV S glycoprotein was initially mapped to a region spanning residues 358 to 662 [49,50]; antisera from RBD protein-immunized mice or rabbits protected against in vitro infection with MERS-CoV. In further studies exploring immune correlates of protection, intranasal administration of RBD protein induced S1-specific immunoglobulin (Ig) G1, IgG2a, IgG3, lung IgA, and neutralizing antibodies (NAb, that is, untyped antibodies shown to functionally inhibit free virus from infecting cells) [51], as well as cell-mediated responses as measured by IL-2 and IFN-γ production in antigen-stimulated CD4+ and CD8+ splenocytes [52]. In this and subsequent studies, the RBD protein was fused to a fragment crystallizable (Fc) region of human IgG1 to increase the in vivo half-life of the immunogen [53]. Profiling of the immunogenic region of the RBD indicated residues 377-588 bound with the highest affinity to soluble DPP4 and induced the highest NAb titers, when administered to both mice and rabbits [54]. This range-refined RBD protein vaccine has been stably expressed in a modified high-yield CHO cell line [55]. Purified and adjuvanted with AddaVaxTM (MF59-like), the RBD protein vaccine was protective when administered intramuscularly to transgenic mice expressing human DPP4 (hDPP4), with no evidence of immunological toxicity or eosinophilic immune enhancement.
Since the S glycoprotein exists in trimeric form on the virion, a quality lost in shortened forms of the protein, a trimer of this RBD protein vaccine has been generated containing a foldon trimerization motif [56]. This RBD protein trimer has been shown to elicit long-lasting NAb and be protective in challenged hDPP4-transgenic mice [57]. Independently, a monomeric RBD protein vaccine has been developed and tested in rhesus macaques, where it reduced MERS-associated lung pathology and reduced viral loads when adjuvanted with alum and administered intramuscularly in a three-dose regimen prior to challenge [58]. Finally, RBD proteins encoding sequences from different strains of MERS-CoV have been shown to induce cross-neutralizing antibodies against divergent human and camel MERS-CoV strains as well as monoclonal antibody (mAb) escape mutants, confirming the promise of the RBD as a valid vaccine target [59].

2.2. Full-Length S

Targeting the entire S glycoprotein has the advantage of including non-RBD neutralizing epitopes, including those in the more conserved domains. S protein “nanoparticles,” protein aggregates containing full-length S, have been proposed as a subunit vaccine, as nanoparticle vaccination adjuvanted with Matrix-M1TM elicited NAb in mice [60] protected adenovirally hDPP4-transduced mice [24] from MERS-CoV challenge [61]. Measurements of viral titer and viral RNA were near the limit of detection in these vaccinated mice. However, despite these promising results, antibody-dependent enhancement (ADE) of infection was previously noted in the context of vaccination with full-length SARS-CoV S protein vaccine [62]. While ADE has not been demonstrated with full length S from MERS-CoV, further studies must consider this as a potential issue.
A variation of the full-length S glycoprotein vaccine is a trimer of S ectodomain (all but the transmembrane domain) conformationally locked in the prefusion state by the substitution of two proline residues in the S2 domain [34]. Work on related fusion proteins such as the F glycoprotein of respiratory syncytial virus has shown that stabilizing the glycoprotein in its prefusion state helps elicit a stronger neutralizing antibody response [63]. This “prefusion” S administered to mice elicited sera with greater neutralization activity against a panel of pseudoviruses bearing strain-specific variants of MERS-CoV S as compared to wild-type S. Since the S2 domain of CoVs is more conserved than the S1 domain [64], targeting epitopes in S2 may provide broader protection against different MERS-CoV strains and other lineage C βCoVs.

2.3. N-Terminal Domain

RBD- and full-length S-based protein vaccines build upon prior vaccine efforts for SARS-CoV. In contrast, vaccines targeting the N-terminal domain of S1 (NTD) offer a novel target. The NTD of S1 does not contain the RBD for MERS-CoV; however, the NTD binds sialic acid and is key to infecting certain cell types [33]. Immunization with NTD protein protected against MERS-CoV challenge in adenovirally hDPP4-transduced mice, inducing cell-mediated responses in splenocytes (CD8+ IFN-γ production, CD4+ IL-2 production, and IL-17A production) as well as humoral responses (IgG and NAb), although NAb titer was lower compared to that of an RBD protein vaccine [65]. Overall, these results suggest that targeting S1 domains outside the RBD may be a viable strategy for MERS-CoV vaccines.

3. DNA Vaccines: Efficient Protection

DNA vaccines offer a rapid platform to design and deliver immunogenic proteins, typically encoded on plasmid vectors and injected into tissue with accompanying electroporation [66]. DNA vaccine administration via electroporation has been tested in clinical trials with immunogenicity comparable to other vaccine types and predominantly low-grade adverse events reported [67]. The in vivo expression of plasmid-encoded proteins recapitulates native post-translational modifications while maintaining the capacity to stimulate both humoral and cell-mediated immunity [68]. While concerns about the safety of DNA vaccines and their potential to integrate into host cell chromosomes were voiced early in their development [69], integration using various plasmids and inserts appears to be extremely rare [70]. Several DNA vaccines for MERS-CoV have been reported to date.

3.1. Full-Length S

pVax1TM is a proprietary, optimized plasmid vaccine vector that has been developed as a MERS-CoV vaccine by encoding a consensus MERS-CoV S glycoprotein containing codon and other proprietary optimizations, as well as an IgE leader sequence to promote expression and mRNA export [71]. Intramuscular administration of this construct with electroporation induced antibodies with cross-MERS-CoV-strain neutralization and antigen-specific, polyfunctional T cell responses in rhesus macaques. These humoral and cell-mediated immune responses correlated with minimal lung pathology and reduced lung viral loads upon MERS-CoV challenge. The same study reported NAb induction in dromedary camels, which indicates that the vaccine could be used in zoonotic reservoirs. Building upon these preclinical results, the pVax1TM vaccine (GLS-5300) has completed a Phase I clinical trial (clinicaltrials.gov/ct2/show/NCT02670187).
A second vaccine platform utilizes pVRC8400 [72,73], a plasmid vector engineered for high transgene expression and enhanced cell-mediated responses. A vaccine regimen consisting of intramuscular administration of MERS-CoV strain England1 full-length S encoded on pVRC8400, with electroporation, and an AlPO4-adjuvanted S1 protein booster, induced NAb in rhesus macaques up to 10 weeks following booster [74]. This vaccine resulted in lower lung pathology upon challenge with the MERS-CoV strain JordanN3. In the same study, the full-length S DNA/S1 protein vaccine induced higher NAb titer in mice than other prime/boost combinations involving constructs encoding either S1 or S with the transmembrane domain (TMD) deleted. Consistent with this finding, the study reported that mAbs induced against domains outside the RBD were able to neutralize MERS-CoV pseudovirus. Together, these results reiterated the immunogenic potential of non-RBD epitopes including those derived from S quaternary (trimer) structure, which could help generate immune responses able to minimize escape variants derived from immunization targeting either the RBD or the S1 domain alone.

3.2. S1 Domain

The S1 domain from MERS-CoV strain Al-Hasa_15_2013 encoded on pcDNATM3.1(+), a proprietary plasmid from which pVax1TM is derived, has also been tested as a vaccine platform. This vaccine induced NAb in mice when given intramuscularly and antigen-specific cytokine production including CD4+ and CD8+ production of both IL-4 and IFN-γ in murine splenocytes [75]. In addition, the vaccine protected adenovirally hDPP4-transduced mice against challenge with MERS-CoV strain EMC/2012. Moreover, this S1 vaccine elicited more NAb than did full-length S in the same vector. This result was attributed to increased secretion of the S1 protein, which lacked a TMD, and greater uptake by antigen-presenting cells [76]. A separate group’s study comparing full-length S- and S1-encoding pcDNATM3.1 vaccines found that the S1 vaccine elicited a more balanced IgG2a/IgG1 ratio in mice compared to that elicited by full-length S [77] suggesting a balanced Th1/Th2 response [78]. Together, these results show that multiple plasmid vaccine vectors encoding either full-length S or the S1 domain induce adaptive immunity and protect against MERS-CoV challenge.

4. Viral Vector Vaccines: Optimized Delivery

Viral vector vaccines contain one or more immunogenic proteins of the pathogen of interest in the context of an attenuated virus backbone. This approach takes advantage of cellular entry by the virus as well as adjuvantation from viral components, and induces both humoral and strong cell-mediated responses [79]. Early studies into viral vector vaccines for MERS-CoV built upon established platforms and have subsequently transitioned to newer viral vector approaches.
Venezuelan equine encephalitis (VEE) virus replicon particles (VRPs), an alphavirus-based platform that replaces the VEE structural genes with a foreign transgene, has been shown to induce strong humoral and cellular immune responses [80,81]. A VRP encoding MERS-CoV S elicited NAb in both young and aged mice [38]. Additional studies with this vector have shown that immunization with an N protein-expressing VRP protected adenovirally hDPP4-transduced mice from MERS-CoV challenge in a CD4+ T cell- and IFN-γ-dependent manner [82]. Moreover, a specific N protein epitope was stimulatory in mice transgenic for human leukocyte antigen DR2 and DR3, highlighting the relevance of this epitope to human antigen recognition and to promoting cell-mediated immunity in humans.
Modified vaccinia virus Ankara (MVA) [83,84], a well-established vaccine platform, has been developed to encode full-length MERS-CoV S. This vaccine induced NAb and CD8+ T cell responses in mice [85] and also protected against MERS-CoV-induced histopathology in adenovirally hDPP4-transduced mice before challenge [86]. Moreover, minimal inflammation and lymph node hyperplasia was observed at the site of injection [87]. This same MVA-MERS-CoV S vaccine injected intramuscularly into dromedary camels was shown to induce NAb and to limit excretion of infectious virus upon intranasal challenge with MERS-CoV [88]. A Phase I clinical trial is underway (clinicaltrials.gov/ct2/show/NCT03615911).
Adenoviruses compose a third platform of viral vectors for MERS-CoV vaccines. Adenovirus-vectored vaccines have been tested in clinical trials for a wide variety of diseases, notably HIV [89]. However, their efficacy may be hampered by pre-existing immunity to prevalent adenovirus serotypes [90,91]. For example, pre-existing immunity to human adenovirus serotype 5 (Ad5) was shown to result in reduced CD8+ T cell responses against an Ad5-vectored transgene [92]. To take advantage of the shared respiratory route of infection of both MERS-CoV and adenovirus, Ad5-vectored full-length S and S1 vaccines have been developed [93]. These elicited antigen-specific IgG and NAb when administered intramuscularly to mice with subsequent intranasal boosting. Importantly, this study did not detect immunity against the Ad5 vector in dromedary camels, the intended vaccination population. Moreover, camel peripheral blood mononuclear cells and a camel-derived fibroblast cell line were able to be infected with Ad5. Another Ad5-MERS-CoV S vaccine has been separately developed, as has a human adenovirus type 41 (Ad41)-MERS-CoV S vaccine [94]. Adenovirus type 41 (Ad41) is an enteric pathogen with potential use as an orally administered vaccine [95]. Both of these vaccines, Ad5-MERS-CoV S and Ad41-MERS-CoV S, were reported to induce humoral responses when administered intragastrically in mice. In addition to humoral responses, they also induced long-lasting cell-mediated responses in the lung and spleen when administered intramuscularly. One final Ad5-based MERS-CoV immunization regimen has been reported [96]. Immunizing with Ad5-vectored S followed by boosting with S nanoparticles induced S-specific IgG, NAb, and both Th1 and Th2 cell-mediated responses in mice, and also protected adenovirally hDPP4-transduced mice from MERS-CoV challenge.
To circumvent the seroprevalence of circulating human adenoviruses, chimpanzee adenoviruses have also been developed as viral vaccine vectors [97] and have entered clinical trials [98]. A MERS-CoV S-encoding vaccine based on a chimpanzee adenoviral vector (ChAdOx1) was shown to induce high levels of NAb and cell-mediated responses (CD8+ IFN-γ, TNFα, and IL-17 production) in mice 4 weeks post-immunization [99]. This vaccine was constructed with a codon-optimized S glycoprotein sequence and the tissue plasminogen activator (tPA) gene leader sequence to promote secretion [100,101]. The ChAdOx1-MERS-CoV S vaccine protected against lethal challenge in a transgenic hDPP4 mouse model [102]. Based on previous work with the ChAdOx1 vector demonstrating its safety in humans, the ChAdOx1-MERS-CoV S vaccine is undergoing a Phase I clinical trial (clinicaltrials.gov/ct2/show/NCT03399578).
Several additional viral vectors have been employed as MERS-CoV vaccines. Measles virus vector platforms have been developed over the past two decades [103]. A full-length or soluble form of S encoded in measles vaccine strain MVvac2 induced NAb, proliferation of T cells, S-specific IFN-γ production, and cytotoxic activity [104]. The vaccine also protected against MERS-CoV challenge in adenovirally hDPP4-transduced mice that were transgenic for a measles virus receptor. Further characterization of the T cell responses induced by this vaccine has been performed [105]. Of note, 5-fold higher numbers of reactive T cells were induced by vaccination with MVvac2-S than those induced by N protein using the same vector. Additionally, antigen-specific IFN-γ production by T cells could be induced in older mice (7 months old) at levels near those induced in younger mice (6–12 weeks old).
Newcastle disease virus (NDV) has been explored as a vaccine vector as it infects the respiratory tract and can induce systemic and mucosal immunity in non-human primates [106]. An NVD vector expressing MERS-CoV S was shown to induce long-lasting (up 14 weeks post-immunization) NAb titers in camels [107]. The research group behind this study also examined vesicular stomatitis virus (VSV) [108] as a viral vector. S expressed from a VSV reverse genetics system was shown to incorporate onto the surface of virions rescued in cell culture. Purified vaccine was able to infect cells in an hDPP4-dependent manner, induced S-specific IgG and NAb in mice, and stimulated humoral and cell-mediated (IFN-γ-production) responses in rhesus macaques [109].
Similar to the VSV platform, a rabies virus (RABV) vector has been explored. Inspired by studies combining rabies and Ebola vaccine platforms [110], a β-propiolactone-inactivated dual rabies/MERS vaccine has been proposed which incorporates the MERS-CoV S1 domain fused to rabies virus G protein on the RABV virion [111]. This vaccine elicited S-specific IgG and NAb and fully protected adenovirally hDPP4-transduced mice from MERS-CoV challenge. The VSV and RABV approaches described here are unique in that they encode S (or S1) in the vector genome and also display it on the virion surface.
Finally, virus-like particles (VLPs), which comprise self-assembling immunogenic proteins, but no genome [112], have also been used as viral vectors. A baculovirus VLP containing S as well as MERS-CoV envelope and matrix proteins elicited RBD-specific IgG and IFN-γ responses in rhesus macaques [113]. A subsequent baculovirus VLP vaccine was developed that focused only on a fusion of the RBD from S and the immunogenic VP2 protein of canine parvovirus. This vaccine induced RBD-specific IgG, NAb, and cell-mediated responses including IFN-γ, IL-2, and IL-4 production in mice, and also activated dendritic cells in inguinal lymph nodes [114]. In summary, a variety of viral vector vaccines for MERS-CoV induce promising immune responses in animal models and often demonstrate protection from challenge.

5. Live Attenuated and Inactivated Vaccines: Situationally Useful

A final approach to developing MERS-CoV vaccines delivers the whole virus, either inactivated or live but attenuated. Both of these vaccine types resemble the original virus, preserving structural features and a full or nearly-full repertoire of immunogenic components. Inactivated viruses may contain structural deformations introduced by inactivation, but, unlike attenuated viruses, they pose no risks, if properly inactivated, either of reversion to a virulent state or persistent infection in immunocompromised patients. Fewer examples of whole virus vaccines, compared to the other vaccine types, have been developed for MERS-CoV.

5.1. Inactivated

Development of inactivated vaccines for MERS-CoV has been stymied by prior concerns with SARS-CoV inactivated vaccines. Eosinophil-related lung pathology was observed for a SARS-CoV vaccine doubly inactivated with both formalin and UV irradiation [115]. This response was particularly notable in aged mice versus young mice, and following heterologous versus homologous challenge. Similarly, immunization with a gamma-irradiated MERS-CoV vaccine adjuvanted with either alum or MF59 elicited NAb and reduced viral titer upon challenge in hDPP4-transgenic mice, but induced eosinophil-related lung pathology in vaccinated mice after challenge [116].
A different inactivation method was tried for a second MERS-CoV inactivated vaccine. Formalin-inactivated MERS-CoV adjuvanted with alum and oligodeoxynucleotides containing unmethylated CpG motifs was shown to elicit levels of NAb on par with those elicited by an S glycoprotein-only vaccine [117]. Moreover, the vaccine offered better protection than S alone based on reduction of lung viral titer in adenovirally hDPP4-transduced mice after MERS-CoV challenge. Remarkably, eosinophil-mediated vaccine-related pathology was not observed in this animal model. Interestingly, it has also been shown that including Toll-like receptor agonists in a UV-inactivated SARS-CoV vaccine reduced Th2-associated pathology in lungs after challenge [118]. These results suggest that inactivated CoV vaccines may remain viable options for further development with the right inactivation method and adjuvants.

5.2. Live Attenuated

Live attenuated vaccines for MERS-CoV show efficacy in animal models, but so far have not been pursued in subsequent studies. While riskier than other vaccine types, live attenuated vaccines have historically offered protection against a variety of threatening illnesses [119] and may be reserved for outbreak scenarios where they offer an immunogenically robust solution. The CoV envelope (E) protein is important in virion assembly and egress, and has also been shown to inhibit the host cell stress response [120]. An E-deletion mutant of SARS-CoV was previously found to be protective in vivo against SARS-CoV challenge [121]. An initial study into a MERS-CoV reverse genetics system reported on a replication-competent, but propagation-defective mutant lacking the E protein that could be rescued in cell culture with E expressed in trans [122]. However, the E-deletion mutant was rescued at 100-fold lower titer compared to wild-type MERS-CoV, perhaps explaining why this mutant has not been further developed as a live attenuated vaccine candidate.
Other CoV components have been targeted in live attenuated vaccine development. Nonstructural protein 14 (nsp14) contains an exoribonuclease (ExoN) essential to replication fidelity that is found in all known nidoviruses with genome sizes greater than 20 kb [5]. A stable deficiency in nsp14 attenuated SARS-CoV in young, aged, and immunocompromised mice, and was able to induce protection following vaccination [123]. However, ExoN mutants for MERS-CoV have not been reported.
CoV gene nonstructural protein 16 (nsp16) is a 2′-O-methyl-transferase involved in viral mRNA capping [124] and was previously inactivated in a SARS-CoV live attenuated vaccine [125,126]. A live nsp16-deficient MERS-CoV vaccine was similarly attenuated in a type I interferon- and IFIT1-dependent manner. Immunization with the MERS-CoV nsp16 mutant induced NAb and protected CRISPR-engineered hDPP4-transgenic mice [23] from challenge with a mouse-adapted MERS-CoV strain [127].
Finally, live attenuated vaccines lacking CoV accessory proteins have also been considered. CoV accessory proteins are dispensable for viral replication but have been shown to modulate interferon signaling and pro-inflammatory cytokine production [128]. A MERS-CoV strain lacking accessory open reading frames (ORFs) 3, 4, and 5 was attenuated in vivo, induced NAb, and like the nsp16 mutant, protected CRISPR-engineered hDPP4-transgenic mice from challenge with a mouse-adapted MERS-CoV strain [129]. Overall, whole vaccine approaches to MERS-CoV vaccination appear both protective and safe in animal models.

6. Conclusions and Future Directions

Aided by knowledge gained from vaccine development against SARS-CoV and other contemporary viral diseases, MERS-CoV vaccine development efforts have multiplied since its emergence, yielding promising vaccine candidates spanning multiple platforms (Table 1). Nevertheless, key barriers to vaccine efficacy first noted for SARS-CoV may also hold true for MERS-CoV. As with SARS-CoV [130], mortality from MERS-CoV has disproportionately affected the aged. Additionally, immunocompromised individuals and those with chronic conditions are at greater risk of mortality from MERS-CoV infection [11]. A universal MERS-CoV vaccine must offer protection to these vulnerable classes of people.
More studies on the effectiveness of the proposed MERS-CoV vaccines in models of immunosenescence, immunocompromise, and chronic conditions are needed. In this regard, vaccination studies with SARS-CoV have indicated that vaccines may be capable of inducing protection in young animals while failing to protect aged animals [115]. In light of the threat of related coronavirus strains emerging, MERS-CoV vaccine studies must also consider heterologous challenge models to ensure safety from vaccine-induced immunopathology, especially in older individuals [115]. In short, vaccine-induced immunopathology, especially Th2-related eosinophilic immune enhancement [131], from both homologous and heterologous challenge should be specifically monitored in vulnerable populations. Interestingly, it was shown that pathology resulting from MERS-CoV challenge in the lungs of immunosuppressed rhesus macaques was lower compared to that of non-immunosuppressed macaques, underscoring the immunopathogenic component of respiratory disease caused by CoVs [132].
The different vaccine platforms described herein have unique advantages and disadvantages. Since severe CoV disease maintains an immunopathogenic component, a successful vaccine must strike a balance between protection and excessive immune activation. As seen with full-length S [44] as well as inactivated virus [116], vaccination may produce immunopathology under certain conditions. Alternatively, protection must be thorough enough to prevent NAb escape, a phenomenon inversely correlated with the number of immunogenic epitopes. While antibodies induced against S of either SARS-CoV or MERS-CoV poorly cross-neutralize across their respective lineages [38], a vaccine that contains multiple immunogenic epitopes would perhaps also offer greater cross-protection against heterologous strains within lineage as they emerge, especially if conserved epitopes are included in vaccine design. A greater understanding of MERS-CoV pathology will also help guide future vaccine development efforts by illuminating possibly critical differences in vaccine responses between MERS-CoV and SARS-CoV, the latter of which has largely influenced vaccine development against the former.
Overall, MERS-CoV vaccines have shown encouraging results in preclinical studies and we hope these vaccines stand up to safety considerations in order to proceed through clinical trials. While development of therapeutic treatment is critical, vaccination carries the promise of mitigating future outbreaks and alleviating disease burden from the most vulnerable populations including the aged, the immunosuppressed, healthcare workers, family members of infected patients, and those in endemic areas.

Funding

Research in this manuscript was supported by grants from the National Institute of Allergy & Infectious Disease and the National Institute of Aging of the NIH under awards U19AI100625 (VDM) and R00AG049092 (VDM). This work was also supported by the McLaughlin Endowment through the Institute of Human Infection and Immunity at the University of Texas Medical Branch. The content is solely the responsibility of the authors and does not necessarily represent the official views of the NIH.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Lau, S.K.; Woo, P.C.; Li, K.S.; Huang, Y.; Tsoi, H.W.; Wong, B.H.; Wong, S.S.; Leung, S.Y.; Chan, K.H.; Yuen, K.Y. Severe acute respiratory syndrome coronavirus-like virus in Chinese horseshoe bats. Proc. Natl. Acad. Sci. USA 2005, 102, 14040–14045. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Trifonov, V.; Khiabanian, H.; Rabadan, R. Geographic dependence, surveillance, and origins of the 2009 influenza A (H1N1) virus. N. Engl. J. Med. 2009, 361, 115–119. [Google Scholar] [CrossRef] [PubMed]
  3. Gire, S.K.; Goba, A.; Andersen, K.G.; Sealfon, R.S.; Park, D.J.; Kanneh, L.; Jalloh, S.; Momoh, M.; Fullah, M.; Dudas, G.; et al. Genomic surveillance elucidates Ebola virus origin and transmission during the 2014 outbreak. Science 2014, 345, 1369–1372. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Baize, S.; Pannetier, D.; Oestereich, L.; Rieger, T.; Koivogui, L.; Magassouba, N.; Soropogui, B.; Sow, M.S.; Keita, S.; De Clerck, H.; et al. Emergence of Zaire Ebola virus disease in Guinea. N. Engl. J. Med. 2014, 371, 1418–1425. [Google Scholar] [CrossRef] [PubMed]
  5. Snijder, E.J.; Decroly, E.; Ziebuhr, J. The Nonstructural Proteins Directing Coronavirus RNA Synthesis and Processing. Adv. Virus Res. 2016, 96, 59–126. [Google Scholar] [PubMed]
  6. Riski, H.; Hovi, T. Coronavirus infections of man associated with diseases other than the common cold. J. Med. Virol. 1980, 6, 259–265. [Google Scholar] [CrossRef] [PubMed]
  7. Chan, J.F.; Lau, S.K.; To, K.K.; Cheng, V.C.; Woo, P.C.; Yuen, K.Y. Middle East respiratory syndrome coronavirus: Another zoonotic betacoronavirus causing SARS-like disease. Clin. Microbiol. Rev. 2015, 28, 465–522. [Google Scholar] [CrossRef]
  8. Li, W.; Shi, Z.; Yu, M.; Ren, W.; Smith, C.; Epstein, J.H.; Wang, H.; Crameri, G.; Hu, Z.; Zhang, H.; et al. Bats are natural reservoirs of SARS-like coronaviruses. Science 2005, 310, 676–679. [Google Scholar] [CrossRef]
  9. Anthony, S.J.; Gilardi, K.; Menachery, V.D.; Goldstein, T.; Ssebide, B.; Mbabazi, R.; Navarrete-Macias, I.; Liang, E.; Wells, H.; Hicks, A.; et al. Further Evidence for Bats as the Evolutionary Source of Middle East Respiratory Syndrome Coronavirus. mBio 2017, 8. [Google Scholar] [CrossRef]
  10. Menachery, V.D.; Yount, B.L., Jr.; Debbink, K.; Agnihothram, S.; Gralinski, L.E.; Plante, J.A.; Graham, R.L.; Scobey, T.; Ge, X.Y.; Donaldson, E.F.; et al. A SARS-like cluster of circulating bat coronaviruses shows potential for human emergence. Nat. Med. 2015, 21, 1508–1513. [Google Scholar] [CrossRef] [Green Version]
  11. Zumla, A.; Hui, D.S.; Perlman, S. Middle East respiratory syndrome. Lancet 2015, 386, 995–1007. [Google Scholar] [CrossRef] [Green Version]
  12. Muller, M.A.; Corman, V.M.; Jores, J.; Meyer, B.; Younan, M.; Liljander, A.; Bosch, B.J.; Lattwein, E.; Hilali, M.; Musa, B.E.; et al. MERS coronavirus neutralizing antibodies in camels, Eastern Africa, 1983–1997. Emerg. Infect. Dis. 2014, 20, 2093–2095. [Google Scholar] [CrossRef] [PubMed]
  13. Azhar, E.I.; El-Kafrawy, S.A.; Farraj, S.A.; Hassan, A.M.; Al-Saeed, M.S.; Hashem, A.M.; Madani, T.A. Evidence for camel-to-human transmission of MERS coronavirus. N. Engl. J. Med. 2014, 370, 2499–2505. [Google Scholar] [CrossRef] [PubMed]
  14. The Health Protection Agency (HPA) UK Novel Coronavirus Investigation Team. Evidence of person-to-person transmission within a family cluster of novel coronavirus infections, United Kingdom, February 2013. Euro Surveill. 2013, 18, 20427. [Google Scholar]
  15. Zumla, A.; Chan, J.F.; Azhar, E.I.; Hui, D.S.; Yuen, K.Y. Coronaviruses-drug discovery and therapeutic options. Nat. Rev. Drug Discov. 2016, 15, 327–347. [Google Scholar] [CrossRef] [PubMed]
  16. Dyall, J.; Coleman, C.M.; Hart, B.J.; Venkataraman, T.; Holbrook, M.R.; Kindrachuk, J.; Johnson, R.F.; Olinger, G.G., Jr.; Jahrling, P.B.; Laidlaw, M.; et al. Repurposing of clinically developed drugs for treatment of Middle East respiratory syndrome coronavirus infection. Antimicrob. Agents Chemother. 2014, 58, 4885–4893. [Google Scholar] [CrossRef] [PubMed]
  17. Public Health England/ISARIC. Treatment of MERS-CoV: Information for Clinicians, Clinical Decision-Making Support for Treatment of MERS-CoV Patients; Public Health England: London, UK, 2015. [Google Scholar]
  18. Vergara-Alert, J.; Vidal, E.; Bensaid, A.; Segales, J. Searching for animal models and potential target species for emerging pathogens: Experience gained from Middle East respiratory syndrome (MERS) coronavirus. One Health 2017, 3, 34–40. [Google Scholar] [CrossRef]
  19. Raj, V.S.; Mou, H.; Smits, S.L.; Dekkers, D.H.; Muller, M.A.; Dijkman, R.; Muth, D.; Demmers, J.A.; Zaki, A.; Fouchier, R.A.; et al. Dipeptidyl peptidase 4 is a functional receptor for the emerging human coronavirus-EMC. Nature 2013, 495, 251–254. [Google Scholar] [CrossRef]
  20. Peck, K.M.; Cockrell, A.S.; Yount, B.L.; Scobey, T.; Baric, R.S.; Heise, M.T. Glycosylation of mouse DPP4 plays a role in inhibiting Middle East respiratory syndrome coronavirus infection. J. Virol. 2015, 89, 4696–4699. [Google Scholar] [CrossRef]
  21. Cockrell, A.S.; Peck, K.M.; Yount, B.L.; Agnihothram, S.S.; Scobey, T.; Curnes, N.R.; Baric, R.S.; Heise, M.T. Mouse dipeptidyl peptidase 4 is not a functional receptor for Middle East respiratory syndrome coronavirus infection. J. Virol. 2014, 88, 5195–5199. [Google Scholar] [CrossRef]
  22. Coleman, C.M.; Matthews, K.L.; Goicochea, L.; Frieman, M.B. Wild-type and innate immune-deficient mice are not susceptible to the Middle East respiratory syndrome coronavirus. J. Gen. Virol. 2014, 95 Pt 2, 408–412. [Google Scholar] [CrossRef]
  23. Cockrell, A.S.; Yount, B.L.; Scobey, T.; Jensen, K.; Douglas, M.; Beall, A.; Tang, X.C.; Marasco, W.A.; Heise, M.T.; Baric, R.S. A mouse model for MERS coronavirus-induced acute respiratory distress syndrome. Nat. Microbiol. 2016, 2, 16226. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Zhao, J.; Li, K.; Wohlford-Lenane, C.; Agnihothram, S.S.; Fett, C.; Gale, M.J., Jr.; Baric, R.S.; Enjuanes, L.; Gallagher, T.; McCray, P.B., Jr.; et al. Rapid generation of a mouse model for Middle East respiratory syndrome. Proc. Natl. Acad. Sci. USA 2014, 111, 4970–4975. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Pascal, K.E.; Coleman, C.M.; Mujica, A.O.; Kamat, V.; Badithe, A.; Fairhurst, J.; Hunt, C.; Strein, J.; Berrebi, A.; Sisk, J.M.; et al. Pre- and postexposure efficacy of fully human antibodies against Spike protein in a novel humanized mouse model of MERS-CoV infection. Proc. Natl. Acad. Sci. USA 2015, 112, 8738–8743. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Tao, X.; Garron, T.; Agrawal, A.S.; Algaissi, A.; Peng, B.H.; Wakamiya, M.; Chan, T.S.; Lu, L.; Du, L.; Jiang, S.; et al. Characterization and Demonstration of the Value of a Lethal Mouse Model of Middle East Respiratory Syndrome Coronavirus Infection and Disease. J. Virol. 2016, 90, 57–67. [Google Scholar] [CrossRef] [PubMed]
  27. Li, K.; Wohlford-Lenane, C.L.; Channappanavar, R.; Park, J.E.; Earnest, J.T.; Bair, T.B.; Bates, A.M.; Brogden, K.A.; Flaherty, H.A.; Gallagher, T.; et al. Mouse-adapted MERS coronavirus causes lethal lung disease in human DPP4 knockin mice. Proc. Natl. Acad. Sci. USA 2017, 114, E3119–E3128. [Google Scholar] [CrossRef] [PubMed]
  28. Du, L.; He, Y.; Zhou, Y.; Liu, S.; Zheng, B.J.; Jiang, S. The spike protein of SARS-CoV—A target for vaccine and therapeutic development. Nat. Rev. Microbiol. 2009, 7, 226–236. [Google Scholar] [CrossRef] [PubMed]
  29. Zhang, N.; Jiang, S.; Du, L. Current advancements and potential strategies in the development of MERS-CoV vaccines. Expert Rev. Vaccines 2014, 13, 761–774. [Google Scholar] [CrossRef]
  30. Surjit, M.; Lal, S.K. The SARS-CoV nucleocapsid protein: A protein with multifarious activities. Infect. Genet. Evolut. 2008, 8, 397–405. [Google Scholar] [CrossRef]
  31. Chang, C.K.; Lo, S.C.; Wang, Y.S.; Hou, M.H. Recent insights into the development of therapeutics against coronavirus diseases by targeting N protein. Drug Discov. Today 2016, 21, 562–572. [Google Scholar] [CrossRef]
  32. Millet, J.K.; Whittaker, G.R. Host cell entry of Middle East respiratory syndrome coronavirus after two-step, furin-mediated activation of the spike protein. Proc. Natl. Acad. Sci. USA 2014, 111, 15214–15219. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Li, W.; Hulswit, R.J.G.; Widjaja, I.; Raj, V.S.; McBride, R.; Peng, W.; Widagdo, W.; Tortorici, M.A.; van Dieren, B.; Lang, Y.; et al. Identification of sialic acid-binding function for the Middle East respiratory syndrome coronavirus spike glycoprotein. Proc. Natl. Acad. Sci. USA 2017, 114, E8508–E8517. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Pallesen, J.; Wang, N.; Corbett, K.S.; Wrapp, D.; Kirchdoerfer, R.N.; Turner, H.L.; Cottrell, C.A.; Becker, M.M.; Wang, L.; Shi, W.; et al. Immunogenicity and structures of a rationally designed prefusion MERS-CoV spike antigen. Proc. Natl. Acad. Sci. USA 2017, 114, E7348–E7357. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Jiang, L.; Wang, N.; Zuo, T.; Shi, X.; Poon, K.M.; Wu, Y.; Gao, F.; Li, D.; Wang, R.; Guo, J.; et al. Potent neutralization of MERS-CoV by human neutralizing monoclonal antibodies to the viral spike glycoprotein. Sci. Transl. Med. 2014, 6, 234ra59. [Google Scholar] [CrossRef] [PubMed]
  36. Irigoyen, N.; Firth, A.E.; Jones, J.D.; Chung, B.Y.; Siddell, S.G.; Brierley, I. High-Resolution Analysis of Coronavirus Gene Expression by RNA Sequencing and Ribosome Profiling. PLoS Pathog. 2016, 12, e1005473. [Google Scholar] [CrossRef] [PubMed]
  37. Buchholz, U.J.; Bukreyev, A.; Yang, L.; Lamirande, E.W.; Murphy, B.R.; Subbarao, K.; Collins, P.L. Contributions of the structural proteins of severe acute respiratory syndrome coronavirus to protective immunity. Proc. Natl. Acad. Sci. USA 2004, 101, 9804–9809. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Agnihothram, S.; Gopal, R.; Yount, B.L., Jr.; Donaldson, E.F.; Menachery, V.D.; Graham, R.L.; Scobey, T.D.; Gralinski, L.E.; Denison, M.R.; Zambon, M.; et al. Evaluation of serologic and antigenic relationships between middle eastern respiratory syndrome coronavirus and other coronaviruses to develop vaccine platforms for the rapid response to emerging coronaviruses. J. Infect. Dis. 2014, 209, 995–1006. [Google Scholar] [CrossRef] [PubMed]
  39. Zhu, M.S.; Pan, Y.; Chen, H.Q.; Shen, Y.; Wang, X.C.; Sun, Y.J.; Tao, K.H. Induction of SARS-nucleoprotein-specific immune response by use of DNA vaccine. Immunol. Lett. 2004, 92, 237–243. [Google Scholar] [CrossRef] [PubMed]
  40. Channappanavar, R.; Zhao, J.; Perlman, S. T cell-mediated immune response to respiratory coronaviruses. Immunol. Res. 2014, 59, 118–128. [Google Scholar] [CrossRef] [Green Version]
  41. Wang, Y.D.; Sin, W.Y.; Xu, G.B.; Yang, H.H.; Wong, T.Y.; Pang, X.W.; He, X.Y.; Zhang, H.G.; Ng, J.N.; Cheng, C.S.; et al. T-cell epitopes in severe acute respiratory syndrome (SARS) coronavirus spike protein elicit a specific T-cell immune response in patients who recover from SARS. J. Virol. 2004, 78, 5612–5618. [Google Scholar] [CrossRef]
  42. Zhao, J.; Alshukairi, A.N.; Baharoon, S.A.; Ahmed, W.A.; Bokhari, A.A.; Nehdi, A.M.; Layqah, L.A.; Alghamdi, M.G.; Al Gethamy, M.M.; Dada, A.M.; et al. Recovery from the Middle East respiratory syndrome is associated with antibody and T-cell responses. Sci. Immunol. 2017, 2. [Google Scholar] [CrossRef] [PubMed]
  43. Deming, D.; Sheahan, T.; Heise, M.; Yount, B.; Davis, N.; Sims, A.; Suthar, M.; Harkema, J.; Whitmore, A.; Pickles, R.; et al. Vaccine efficacy in senescent mice challenged with recombinant SARS-CoV bearing epidemic and zoonotic spike variants. PLoS Med. 2006, 3, e525. [Google Scholar] [CrossRef]
  44. Tseng, C.T.; Sbrana, E.; Iwata-Yoshikawa, N.; Newman, P.C.; Garron, T.; Atmar, R.L.; Peters, C.J.; Couch, R.B. Immunization with SARS coronavirus vaccines leads to pulmonary immunopathology on challenge with the SARS virus. PLoS ONE 2012, 7, e35421. [Google Scholar] [CrossRef]
  45. Weihofen, W.A.; Liu, J.; Reutter, W.; Saenger, W.; Fan, H. Crystal structure of CD26/dipeptidyl-peptidase IV in complex with adenosine deaminase reveals a highly amphiphilic interface. J. Biol. Chem. 2004, 279, 43330–43335. [Google Scholar] [CrossRef] [PubMed]
  46. Wang, Q.; Wong, G.; Lu, G.; Yan, J.; Gao, G.F. MERS-CoV spike protein: Targets for vaccines and therapeutics. Antivir. Res. 2016, 133, 165–177. [Google Scholar] [CrossRef] [PubMed]
  47. Hansson, M.; Nygren, P.A.; Stahl, S. Design and production of recombinant subunit vaccines. Biotechnol. Appl. Biochem. 2000, 32 (Pt 2), 95–107. [Google Scholar] [CrossRef] [PubMed]
  48. Cao, Z.; Liu, L.; Du, L.; Zhang, C.; Jiang, S.; Li, T.; He, Y. Potent and persistent antibody responses against the receptor-binding domain of SARS-CoV spike protein in recovered patients. Virol. J. 2010, 7, 299. [Google Scholar] [CrossRef] [Green Version]
  49. Du, L.; Zhao, G.; Kou, Z.; Ma, C.; Sun, S.; Poon, V.K.; Lu, L.; Wang, L.; Debnath, A.K.; Zheng, B.J.; et al. Identification of a receptor-binding domain in the S protein of the novel human coronavirus Middle East respiratory syndrome coronavirus as an essential target for vaccine development. J. Virol. 2013, 87, 9939–9942. [Google Scholar] [CrossRef]
  50. Mou, H.; Raj, V.S.; van Kuppeveld, F.J.; Rottier, P.J.; Haagmans, B.L.; Bosch, B.J. The receptor binding domain of the new Middle East respiratory syndrome coronavirus maps to a 231-residue region in the spike protein that efficiently elicits neutralizing antibodies. J. Virol. 2013, 87, 9379–9383. [Google Scholar] [CrossRef]
  51. Burton, D.R.; Williamson, R.A.; Parren, P.W. Antibody and virus: Binding and neutralization. Virology 2000, 270, 1–3. [Google Scholar] [CrossRef]
  52. Ma, C.; Li, Y.; Wang, L.; Zhao, G.; Tao, X.; Tseng, C.T.; Zhou, Y.; Du, L.; Jiang, S. Intranasal vaccination with recombinant receptor-binding domain of MERS-CoV spike protein induces much stronger local mucosal immune responses than subcutaneous immunization: Implication for designing novel mucosal MERS vaccines. Vaccine 2014, 32, 2100–2108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Zhang, M.Y.; Wang, Y.; Mankowski, M.K.; Ptak, R.G.; Dimitrov, D.S. Cross-reactive HIV-1-neutralizing activity of serum IgG from a rabbit immunized with gp41 fused to IgG1 Fc: Possible role of the prolonged half-life of the immunogen. Vaccine 2009, 27, 857–863. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Ma, C.; Wang, L.; Tao, X.; Zhang, N.; Yang, Y.; Tseng, C.K.; Li, F.; Zhou, Y.; Jiang, S.; Du, L. Searching for an ideal vaccine candidate among different MERS coronavirus receptor-binding fragments—The importance of immunofocusing in subunit vaccine design. Vaccine 2014, 32, 6170–6176. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Nyon, M.P.; Du, L.; Tseng, C.K.; Seid, C.A.; Pollet, J.; Naceanceno, K.S.; Agrawal, A.; Algaissi, A.; Peng, B.H.; Tai, W.; et al. Engineering a stable CHO cell line for the expression of a MERS-coronavirus vaccine antigen. Vaccine 2018, 36, 1853–1862. [Google Scholar] [CrossRef] [PubMed]
  56. Letarov, A.V.; Londer, Y.Y.; Boudko, S.P.; Mesyanzhinov, V.V. The carboxy-terminal domain initiates trimerization of bacteriophage T4 fibritin. Biochem. Biokhimiia 1999, 64, 817–823. [Google Scholar]
  57. Tai, W.; Zhao, G.; Sun, S.; Guo, Y.; Wang, Y.; Tao, X.; Tseng, C.K.; Li, F.; Jiang, S.; Du, L.; et al. A recombinant receptor-binding domain of MERS-CoV in trimeric form protects human dipeptidyl peptidase 4 (hDPP4) transgenic mice from MERS-CoV infection. Virology 2016, 499, 375–382. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Lan, J.; Yao, Y.; Deng, Y.; Chen, H.; Lu, G.; Wang, W.; Bao, L.; Deng, W.; Wei, Q.; Gao, G.F.; et al. Recombinant Receptor Binding Domain Protein Induces Partial Protective Immunity in Rhesus Macaques Against Middle East Respiratory Syndrome Coronavirus Challenge. EBioMedicine 2015, 2, 1438–1446. [Google Scholar] [CrossRef] [Green Version]
  59. Tai, W.; Wang, Y.; Fett, C.A.; Zhao, G.; Li, F.; Perlman, S.; Jiang, S.; Zhou, Y.; Du, L. Recombinant Receptor-Binding Domains of Multiple Middle East Respiratory Syndrome Coronaviruses (MERS-CoVs) Induce Cross-Neutralizing Antibodies against Divergent Human and Camel MERS-CoVs and Antibody Escape Mutants. J. Virol. 2017, 91, e01651-16. [Google Scholar] [CrossRef]
  60. Coleman, C.M.; Liu, Y.V.; Mu, H.; Taylor, J.K.; Massare, M.; Flyer, D.C.; Smith, G.E.; Frieman, M.B. Purified coronavirus spike protein nanoparticles induce coronavirus neutralizing antibodies in mice. Vaccine 2014, 32, 3169–3174. [Google Scholar] [CrossRef] [Green Version]
  61. Coleman, C.M.; Venkataraman, T.; Liu, Y.V.; Glenn, G.M.; Smith, G.E.; Flyer, D.C.; Frieman, M.B. MERS-CoV spike nanoparticles protect mice from MERS-CoV infection. Vaccine 2017, 35, 1586–1589. [Google Scholar] [CrossRef] [Green Version]
  62. Kam, Y.W.; Kien, F.; Roberts, A.; Cheung, Y.C.; Lamirande, E.W.; Vogel, L.; Chu, S.L.; Tse, J.; Guarner, J.; Zaki, S.R.; et al. Antibodies against trimeric S glycoprotein protect hamsters against SARS-CoV challenge despite their capacity to mediate FcgammaRII-dependent entry into B cells in vitro. Vaccine 2007, 25, 729–740. [Google Scholar] [CrossRef] [PubMed]
  63. McLellan, J.S.; Chen, M.; Joyce, M.G.; Sastry, M.; Stewart-Jones, G.B.; Yang, Y.; Zhang, B.; Chen, L.; Srivatsan, S.; Zheng, A.; et al. Structure-based design of a fusion glycoprotein vaccine for respiratory syncytial virus. Science 2013, 342, 592–598. [Google Scholar] [CrossRef] [PubMed]
  64. Rota, P.A.; Oberste, M.S.; Monroe, S.S.; Nix, W.A.; Campagnoli, R.; Icenogle, J.P.; Penaranda, S.; Bankamp, B.; Maher, K.; Chen, M.H.; et al. Characterization of a novel coronavirus associated with severe acute respiratory syndrome. Science 2003, 300, 1394–1399. [Google Scholar] [CrossRef] [PubMed]
  65. Jiaming, L.; Yanfeng, Y.; Yao, D.; Yawei, H.; Linlin, B.; Baoying, H.; Jinghua, Y.; Gao, G.F.; Chuan, Q.; Wenjie, T. The recombinant N-terminal domain of spike proteins is a potential vaccine against Middle East respiratory syndrome coronavirus (MERS-CoV) infection. Vaccine 2017, 35, 10–18. [Google Scholar] [CrossRef] [PubMed]
  66. Aihara, H.; Miyazaki, J. Gene transfer into muscle by electroporation in vivo. Nat. Biotechnol. 1998, 16, 867–870. [Google Scholar] [CrossRef] [PubMed]
  67. Sardesai, N.Y.; Weiner, D.B. Electroporation delivery of DNA vaccines: Prospects for success. Curr. Opin. Immunol. 2011, 23, 421–429. [Google Scholar] [CrossRef]
  68. Liu, M.A. DNA vaccines: An historical perspective and view to the future. Immunol. Rev. 2011, 239, 62–84. [Google Scholar] [CrossRef]
  69. Nichols, W.W.; Ledwith, B.J.; Manam, S.V.; Troilo, P.J. Potential DNA vaccine integration into host cell genome. Ann. N. Y. Acad. Sci. 1995, 772, 30–39. [Google Scholar] [CrossRef]
  70. Sheets, R.L.; Stein, J.; Manetz, T.S.; Duffy, C.; Nason, M.; Andrews, C.; Kong, W.P.; Nabel, G.J.; Gomez, P.L. Biodistribution of DNA plasmid vaccines against HIV-1, Ebola, Severe Acute Respiratory Syndrome, or West Nile virus is similar, without integration, despite differing plasmid backbones or gene inserts. Toxicol. Sci. 2006, 91, 610–619. [Google Scholar] [CrossRef]
  71. Muthumani, K.; Falzarano, D.; Reuschel, E.L.; Tingey, C.; Flingai, S.; Villarreal, D.O.; Wise, M.; Patel, A.; Izmirly, A.; Aljuaid, A.; et al. A synthetic consensus anti-spike protein DNA vaccine induces protective immunity against Middle East respiratory syndrome coronavirus in nonhuman primates. Sci. Transl. Med. 2015, 7, 301ra132. [Google Scholar] [CrossRef]
  72. Barouch, D.H.; Yang, Z.Y.; Kong, W.P.; Korioth-Schmitz, B.; Sumida, S.M.; Truitt, D.M.; Kishko, M.G.; Arthur, J.C.; Miura, A.; Mascola, J.R.; et al. A human T-cell leukemia virus type 1 regulatory element enhances the immunogenicity of human immunodeficiency virus type 1 DNA vaccines in mice and nonhuman primates. J. Virol. 2005, 79, 8828–8834. [Google Scholar] [CrossRef] [PubMed]
  73. Cayabyab, M.J.; Kashino, S.S.; Campos-Neto, A. Robust immune response elicited by a novel and unique Mycobacterium tuberculosis protein using an optimized DNA/protein heterologous prime/boost protocol. Immunology 2012, 135, 216–225. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Wang, L.; Shi, W.; Joyce, M.G.; Modjarrad, K.; Zhang, Y.; Leung, K.; Lees, C.R.; Zhou, T.; Yassine, H.M.; Kanekiyo, M.; et al. Evaluation of candidate vaccine approaches for MERS-CoV. Nat. Commun. 2015, 6, 7712. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Chi, H.; Zheng, X.; Wang, X.; Wang, C.; Wang, H.; Gai, W.; Perlman, S.; Yang, S.; Zhao, J.; Xia, X. DNA vaccine encoding Middle East respiratory syndrome coronavirus S1 protein induces protective immune responses in mice. Vaccine 2017, 35, 2069–2075. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Lanzavecchia, A. Mechanisms of antigen uptake for presentation. Curr. Opin. Immunol. 1996, 8, 348–354. [Google Scholar] [CrossRef]
  77. Al-Amri, S.S.; Abbas, A.T.; Siddiq, L.A.; Alghamdi, A.; Sanki, M.A.; Al-Muhanna, M.K.; Alhabbab, R.Y.; Azhar, E.I.; Li, X.; Hashem, A.M. Immunogenicity of Candidate MERS-CoV DNA Vaccines Based on the Spike Protein. Sci. Rep. 2017, 7, 44875. [Google Scholar] [CrossRef]
  78. Stevens, T.L.; Bossie, A.; Sanders, V.M.; Fernandez-Botran, R.; Coffman, R.L.; Mosmann, T.R.; Vitetta, E.S. Regulation of antibody isotype secretion by subsets of antigen-specific helper T cells. Nature 1988, 334, 255–258. [Google Scholar] [CrossRef]
  79. Rollier, C.S.; Reyes-Sandoval, A.; Cottingham, M.G.; Ewer, K.; Hill, A.V. Viral vectors as vaccine platforms: Deployment in sight. Curr. Opin. Immunol. 2011, 23, 377–382. [Google Scholar] [CrossRef]
  80. Pushko, P.; Parker, M.; Ludwig, G.V.; Davis, N.L.; Johnston, R.E.; Smith, J.F. Replicon-helper systems from attenuated Venezuelan equine encephalitis virus: Expression of heterologous genes in vitro and immunization against heterologous pathogens in vivo. Virology 1997, 239, 389–401. [Google Scholar] [CrossRef]
  81. Agnihothram, S.; Menachery, V.D.; Yount, B.L., Jr.; Lindesmith, L.C.; Scobey, T.; Whitmore, A.; Schafer, A.; Heise, M.T.; Baric, R.S. Development of a Broadly Accessible Venezuelan Equine Encephalitis Virus Replicon Particle Vaccine Platform. J. Virol. 2018, 92, e00027-18. [Google Scholar] [CrossRef]
  82. Zhao, J.; Mangalam, A.K.; Channappanavar, R.; Fett, C.; Meyerholz, D.K.; Agnihothram, S.; Baric, R.S.; David, C.S.; Perlman, S. Airway Memory CD4(+) T Cells Mediate Protective Immunity against Emerging Respiratory Coronaviruses. Immunity 2016, 44, 1379–1391. [Google Scholar] [CrossRef] [PubMed]
  83. Sutter, G.; Moss, B. Nonreplicating vaccinia vector efficiently expresses recombinant genes. Proc. Natl. Acad. Sci. USA 1992, 89, 10847–10851. [Google Scholar] [CrossRef] [PubMed]
  84. Stittelaar, K.J.; Kuiken, T.; de Swart, R.L.; van Amerongen, G.; Vos, H.W.; Niesters, H.G.; van Schalkwijk, P.; van der Kwast, T.; Wyatt, L.S.; Moss, B.; et al. Safety of modified vaccinia virus Ankara (MVA) in immune-suppressed macaques. Vaccine 2001, 19, 3700–3709. [Google Scholar] [CrossRef]
  85. Song, F.; Fux, R.; Provacia, L.B.; Volz, A.; Eickmann, M.; Becker, S.; Osterhaus, A.D.; Haagmans, B.L.; Sutter, G. Middle East respiratory syndrome coronavirus spike protein delivered by modified vaccinia virus Ankara efficiently induces virus-neutralizing antibodies. J. Virol. 2013, 87, 11950–11954. [Google Scholar] [CrossRef] [PubMed]
  86. Volz, A.; Kupke, A.; Song, F.; Jany, S.; Fux, R.; Shams-Eldin, H.; Schmidt, J.; Becker, C.; Eickmann, M.; Becker, S.; et al. Protective Efficacy of Recombinant Modified Vaccinia Virus Ankara Delivering Middle East Respiratory Syndrome Coronavirus Spike Glycoprotein. J. Virol. 2015, 89, 8651–8656. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Langenmayer, M.C.; Lulf-Averhoff, A.T.; Adam-Neumair, S.; Fux, R.; Sutter, G.; Volz, A. Distribution and absence of generalized lesions in mice following single dose intramuscular inoculation of the vaccine candidate MVA-MERS-S. Biologicals 2018, 54, 58–62. [Google Scholar] [CrossRef] [PubMed]
  88. Haagmans, B.L.; van den Brand, J.M.; Raj, V.S.; Volz, A.; Wohlsein, P.; Smits, S.L.; Schipper, D.; Bestebroer, T.M.; Okba, N.; Fux, R.; et al. An orthopoxvirus-based vaccine reduces virus excretion after MERS-CoV infection in dromedary camels. Science 2016, 351, 77–81. [Google Scholar] [CrossRef]
  89. Hammer, S.M.; Sobieszczyk, M.E.; Janes, H.; Karuna, S.T.; Mulligan, M.J.; Grove, D.; Koblin, B.A.; Buchbinder, S.P.; Keefer, M.C.; Tomaras, G.D.; et al. Efficacy trial of a DNA/rAd5 HIV-1 preventive vaccine. N. Engl. J. Med. 2013, 369, 2083–2092. [Google Scholar] [CrossRef]
  90. Chirmule, N.; Propert, K.; Magosin, S.; Qian, Y.; Qian, R.; Wilson, J. Immune responses to adenovirus and adeno-associated virus in humans. Gene Ther. 1999, 6, 1574–1583. [Google Scholar] [CrossRef] [Green Version]
  91. Mast, T.C.; Kierstead, L.; Gupta, S.B.; Nikas, A.A.; Kallas, E.G.; Novitsky, V.; Mbewe, B.; Pitisuttithum, P.; Schechter, M.; Vardas, E.; et al. International epidemiology of human pre-existing adenovirus (Ad) type-5, type-6, type-26 and type-36 neutralizing antibodies: Correlates of high Ad5 titers and implications for potential HIV vaccine trials. Vaccine 2010, 28, 950–957. [Google Scholar] [CrossRef]
  92. Mercier, S.; Rouard, H.; Delfau-Larue, M.H.; Eloit, M. Specific antibodies modulate the interactions of adenovirus type 5 with dendritic cells. Virology 2004, 322, 308–317. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Kim, E.; Okada, K.; Kenniston, T.; Raj, V.S.; AlHajri, M.M.; Farag, E.A.; AlHajri, F.; Osterhaus, A.D.; Haagmans, B.L.; Gambotto, A. Immunogenicity of an adenoviral-based Middle East Respiratory Syndrome coronavirus vaccine in BALB/c mice. Vaccine 2014, 32, 5975–5982. [Google Scholar] [CrossRef]
  94. Guo, X.; Deng, Y.; Chen, H.; Lan, J.; Wang, W.; Zou, X.; Hung, T.; Lu, Z.; Tan, W. Systemic and mucosal immunity in mice elicited by a single immunization with human adenovirus type 5 or 41 vector-based vaccines carrying the spike protein of Middle East respiratory syndrome coronavirus. Immunology 2015, 145, 476–484. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Lemiale, F.; Haddada, H.; Nabel, G.J.; Brough, D.E.; King, C.R.; Gall, J.G. Novel adenovirus vaccine vectors based on the enteric-tropic serotype 41. Vaccine 2007, 25, 2074–2084. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Jung, S.Y.; Kang, K.W.; Lee, E.Y.; Seo, D.W.; Kim, H.L.; Kim, H.; Kwon, T.; Park, H.L.; Lee, S.M.; Nam, J.H. Heterologous prime-boost vaccination with adenoviral vector and protein nanoparticles induces both Th1 and Th2 responses against Middle East respiratory syndrome coronavirus. Vaccine 2018, 36, 3468–3476. [Google Scholar] [CrossRef] [PubMed]
  97. Farina, S.F.; Gao, G.P.; Xiang, Z.Q.; Rux, J.J.; Burnett, R.M.; Alvira, M.R.; Marsh, J.; Ertl, H.C.; Wilson, J.M. Replication-defective vector based on a chimpanzee adenovirus. J. Virol. 2001, 75, 11603–11613. [Google Scholar] [CrossRef]
  98. Ledgerwood, J.E.; DeZure, A.D.; Stanley, D.A.; Coates, E.E.; Novik, L.; Enama, M.E.; Berkowitz, N.M.; Hu, Z.; Joshi, G.; Ploquin, A.; et al. Chimpanzee Adenovirus Vector Ebola Vaccine. N. Engl. J. Med. 2017, 376, 928–938. [Google Scholar] [CrossRef] [PubMed]
  99. Alharbi, N.K.; Padron-Regalado, E.; Thompson, C.P.; Kupke, A.; Wells, D.; Sloan, M.A.; Grehan, K.; Temperton, N.; Lambe, T.; Warimwe, G.; et al. ChAdOx1 and MVA based vaccine candidates against MERS-CoV elicit neutralising antibodies and cellular immune responses in mice. Vaccine 2017, 35, 3780–3788. [Google Scholar] [CrossRef]
  100. Li, Z.; Howard, A.; Kelley, C.; Delogu, G.; Collins, F.; Morris, S. Immunogenicity of DNA vaccines expressing tuberculosis proteins fused to tissue plasminogen activator signal sequences. Infect. Immunity 1999, 67, 4780–4786. [Google Scholar]
  101. Zhang, Y.; Feng, L.; Li, L.; Wang, D.; Li, C.; Sun, C.; Li, P.; Zheng, X.; Liu, Y.; Yang, W.; et al. Effects of the fusion design and immunization route on the immunogenicity of Ag85A-Mtb32 in adenoviral vectored tuberculosis vaccine. Hum. Vaccines Immunother. 2015, 11, 1803–1813. [Google Scholar] [CrossRef] [Green Version]
  102. Munster, V.J.; Wells, D.; Lambe, T.; Wright, D.; Fischer, R.J.; Bushmaker, T.; Saturday, G.; van Doremalen, N.; Gilbert, S.C.; de Wit, E.; et al. Protective efficacy of a novel simian adenovirus vaccine against lethal MERS-CoV challenge in a transgenic human DPP4 mouse model. NPJ Vaccines 2017, 2, 28. [Google Scholar] [CrossRef] [PubMed]
  103. Zuniga, A.; Wang, Z.; Liniger, M.; Hangartner, L.; Caballero, M.; Pavlovic, J.; Wild, P.; Viret, J.F.; Glueck, R.; Billeter, M.A.; et al. Attenuated measles virus as a vaccine vector. Vaccine 2007, 25, 2974–2983. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Malczyk, A.H.; Kupke, A.; Prufer, S.; Scheuplein, V.A.; Hutzler, S.; Kreuz, D.; Beissert, T.; Bauer, S.; Hubich-Rau, S.; Tondera, C.; et al. A Highly Immunogenic and Protective Middle East Respiratory Syndrome Coronavirus Vaccine Based on a Recombinant Measles Virus Vaccine Platform. J. Virol. 2015, 89, 11654–11667. [Google Scholar] [CrossRef] [Green Version]
  105. Bodmer, B.S.; Fiedler, A.H.; Hanauer, J.R.H.; Prufer, S.; Muhlebach, M.D. Live-attenuated bivalent measles virus-derived vaccines targeting Middle East respiratory syndrome coronavirus induce robust and multifunctional T cell responses against both viruses in an appropriate mouse model. Virology 2018, 521, 99–107. [Google Scholar] [CrossRef] [PubMed]
  106. Kim, S.H.; Samal, S.K. Newcastle Disease Virus as a Vaccine Vector for Development of Human and Veterinary Vaccines. Viruses 2016, 8, 183. [Google Scholar] [CrossRef] [PubMed]
  107. Liu, R.-Q.; Ge, J.-Y.; Wang, J.-L.; Yu, S.; Zhang, H.-L.; Wang, J.-L.; Wen, Z.-Y.; Bu, Z.-G. Newcastle disease virus-based MERS-CoV candidate vaccine elicits high-level and lasting neutralizing antibodies in Bactrian camels. J. Integr. Agric. 2017, 16, 2264–2273. [Google Scholar] [CrossRef]
  108. Lichty, B.D.; Power, A.T.; Stojdl, D.F.; Bell, J.C. Vesicular stomatitis virus: Re-inventing the bullet. Trends Mol. Med. 2004, 10, 210–216. [Google Scholar] [CrossRef]
  109. Liu, R.; Wang, J.; Shao, Y.; Wang, X.; Zhang, H.; Shuai, L.; Ge, J.; Wen, Z.; Bu, Z. A recombinant VSV-vectored MERS-CoV vaccine induces neutralizing antibody and T cell responses in rhesus monkeys after single dose immunization. Antivir. Res. 2018, 150, 30–38. [Google Scholar] [CrossRef]
  110. Willet, M.; Kurup, D.; Papaneri, A.; Wirblich, C.; Hooper, J.W.; Kwilas, S.A.; Keshwara, R.; Hudacek, A.; Beilfuss, S.; Rudolph, G.; et al. Preclinical Development of Inactivated Rabies Virus-Based Polyvalent Vaccine Against Rabies and Filoviruses. J. Infect. Dis. 2015, 212 (Suppl. 2), S414–S424. [Google Scholar] [CrossRef]
  111. Wirblich, C.; Coleman, C.M.; Kurup, D.; Abraham, T.S.; Bernbaum, J.G.; Jahrling, P.B.; Hensley, L.E.; Johnson, R.F.; Frieman, M.B.; Schnell, M.J. One-Health: A Safe, Efficient, Dual-Use Vaccine for Humans and Animals against Middle East Respiratory Syndrome Coronavirus and Rabies Virus. J. Virol. 2017, 91, e02040-16. [Google Scholar] [CrossRef]
  112. Zeltins, A. Construction and characterization of virus-like particles: A review. Mol. Biotechnol. 2013, 53, 92–107. [Google Scholar] [CrossRef]
  113. Wang, C.; Zheng, X.; Gai, W.; Zhao, Y.; Wang, H.; Feng, N.; Chi, H.; Qiu, B.; Li, N.; Wang, T.; et al. MERS-CoV virus-like particles produced in insect cells induce specific humoural and cellular imminity in rhesus macaques. Oncotarget 2017, 8, 12686–12694. [Google Scholar] [CrossRef] [PubMed]
  114. Wang, C.; Zheng, X.; Gai, W.; Wong, G.; Wang, H.; Jin, H.; Feng, N.; Zhao, Y.; Zhang, W.; Li, N.; et al. Novel chimeric virus-like particles vaccine displaying MERS-CoV receptor-binding domain induce specific humoral and cellular immune response in mice. Antivir. Res. 2017, 140, 55–61. [Google Scholar] [CrossRef] [PubMed]
  115. Bolles, M.; Deming, D.; Long, K.; Agnihothram, S.; Whitmore, A.; Ferris, M.; Funkhouser, W.; Gralinski, L.; Totura, A.; Heise, M.; et al. A double-inactivated severe acute respiratory syndrome coronavirus vaccine provides incomplete protection in mice and induces increased eosinophilic proinflammatory pulmonary response upon challenge. J. Virol. 2011, 85, 12201–12215. [Google Scholar] [CrossRef] [PubMed]
  116. Agrawal, A.S.; Tao, X.; Algaissi, A.; Garron, T.; Narayanan, K.; Peng, B.H.; Couch, R.B.; Tseng, C.T. Immunization with inactivated Middle East Respiratory Syndrome coronavirus vaccine leads to lung immunopathology on challenge with live virus. Hum. Vaccines Immunother. 2016, 12, 2351–2356. [Google Scholar] [CrossRef] [PubMed]
  117. Deng, Y.; Lan, J.; Bao, L.; Huang, B.; Ye, F.; Chen, Y.; Yao, Y.; Wang, W.; Qin, C.; Tan, W. Enhanced protection in mice induced by immunization with inactivated whole viruses compare to spike protein of middle east respiratory syndrome coronavirus. Emerg. Microbes Infect. 2018, 7, 60. [Google Scholar] [CrossRef] [Green Version]
  118. Iwata-Yoshikawa, N.; Uda, A.; Suzuki, T.; Tsunetsugu-Yokota, Y.; Sato, Y.; Morikawa, S.; Tashiro, M.; Sata, T.; Hasegawa, H.; Nagata, N. Effects of Toll-like receptor stimulation on eosinophilic infiltration in lungs of BALB/c mice immunized with UV-inactivated severe acute respiratory syndrome-related coronavirus vaccine. J. Virol. 2014, 88, 8597–8614. [Google Scholar] [CrossRef]
  119. Plotkin, S.A. Vaccines: Past, present and future. Nat. Med. 2005, 11 (Suppl. 4), S5–S11. [Google Scholar] [CrossRef]
  120. Ruch, T.R.; Machamer, C.E. The coronavirus E protein: Assembly and beyond. Viruses 2012, 4, 363–382. [Google Scholar] [CrossRef]
  121. Lamirande, E.W.; DeDiego, M.L.; Roberts, A.; Jackson, J.P.; Alvarez, E.; Sheahan, T.; Shieh, W.J.; Zaki, S.R.; Baric, R.; Enjuanes, L.; et al. A live attenuated severe acute respiratory syndrome coronavirus is immunogenic and efficacious in golden Syrian hamsters. J. Virol. 2008, 82, 7721–7724. [Google Scholar] [CrossRef]
  122. Almazan, F.; DeDiego, M.L.; Sola, I.; Zuniga, S.; Nieto-Torres, J.L.; Marquez-Jurado, S.; Andres, G.; Enjuanes, L. Engineering a replication-competent, propagation-defective Middle East respiratory syndrome coronavirus as a vaccine candidate. mBio 2013, 4, e00650-13. [Google Scholar] [CrossRef] [PubMed]
  123. Graham, R.L.; Becker, M.M.; Eckerle, L.D.; Bolles, M.; Denison, M.R.; Baric, R.S. A live, impaired-fidelity coronavirus vaccine protects in an aged, immunocompromised mouse model of lethal disease. Nat. Med. 2012, 18, 1820–1826. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Zust, R.; Cervantes-Barragan, L.; Habjan, M.; Maier, R.; Neuman, B.W.; Ziebuhr, J.; Szretter, K.J.; Baker, S.C.; Barchet, W.; Diamond, M.S.; et al. Ribose 2′-O-methylation provides a molecular signature for the distinction of self and non-self mRNA dependent on the RNA sensor Mda5. Nat. Immunol. 2011, 12, 137–143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Menachery, V.D.; Yount, B.L., Jr.; Josset, L.; Gralinski, L.E.; Scobey, T.; Agnihothram, S.; Katze, M.G.; Baric, R.S. Attenuation and restoration of severe acute respiratory syndrome coronavirus mutant lacking 2′-o-methyltransferase activity. J. Virol. 2014, 88, 4251–4264. [Google Scholar] [CrossRef] [PubMed]
  126. Menachery, V.D.; Gralinski, L.E.; Mitchell, H.D.; Dinnon, K.H., III; Leist, S.R.; Yount, B.L., Jr.; McAnarney, E.T.; Graham, R.L.; Waters, K.M.; Baric, R.S. Combination Attenuation Offers Strategy for Live Attenuated Coronavirus Vaccines. J. Virol. 2018, 92, e00710-18. [Google Scholar] [CrossRef] [PubMed]
  127. Menachery, V.D.; Gralinski, L.E.; Mitchell, H.D.; Dinnon, K.H., III; Leist, S.R.; Yount, B.L., Jr.; Graham, R.L.; McAnarney, E.T.; Stratton, K.G.; Cockrell, A.S.; et al. Middle East Respiratory Syndrome Coronavirus Nonstructural Protein 16 Is Necessary for Interferon Resistance and Viral Pathogenesis. mSphere 2017, 2, e00346-17. [Google Scholar] [CrossRef] [PubMed]
  128. Liu, D.X.; Fung, T.S.; Chong, K.K.; Shukla, A.; Hilgenfeld, R. Accessory proteins of SARS-CoV and other coronaviruses. Antivir. Res. 2014, 109, 97–109. [Google Scholar] [CrossRef] [PubMed]
  129. Menachery, V.D.; Mitchell, H.D.; Cockrell, A.S.; Gralinski, L.E.; Yount, B.L., Jr.; Graham, R.L.; McAnarney, E.T.; Douglas, M.G.; Scobey, T.; Beall, A.; et al. MERS-CoV Accessory ORFs Play Key Role for Infection and Pathogenesis. mBio 2017, 8. [Google Scholar] [CrossRef] [PubMed]
  130. World Health Organization. Consensus Document on the Epidemiology of Severe Acute Respiratory Syndrome (SARS); World Health Organization: Geneva, Switzerland, 2003. [Google Scholar]
  131. Hotez, P.J.; Bottazzi, M.E.; Tseng, C.T.; Zhan, B.; Lustigman, S.; Du, L.; Jiang, S. Calling for rapid development of a safe and effective MERS vaccine. Microbes Infect. 2014, 16, 529–531. [Google Scholar] [CrossRef]
  132. Prescott, J.; Falzarano, D.; de Wit, E.; Hardcastle, K.; Feldmann, F.; Haddock, E.; Scott, D.; Feldmann, H.; Munster, V.J. Pathogenicity and Viral Shedding of MERS-CoV in Immunocompromised Rhesus Macaques. Front. Immunol. 2018, 9, 205. [Google Scholar] [CrossRef]
Figure 1. The MERS-CoV spike (S) glycoprotein, a Class I fusion protein and the target of the majority of vaccine candidates, exists naturally in trimer form as shown in this simplified diagram. DPP4: dipeptidyl peptidase 4, the receptor for S. S1: S1 domain of S. S2: S2 domain of S. RBD: receptor binding domain. NTD: N-terminal domain of S1. TMD: transmembrane domain. Structural configurations adapted from [45,46,34].
Figure 1. The MERS-CoV spike (S) glycoprotein, a Class I fusion protein and the target of the majority of vaccine candidates, exists naturally in trimer form as shown in this simplified diagram. DPP4: dipeptidyl peptidase 4, the receptor for S. S1: S1 domain of S. S2: S2 domain of S. RBD: receptor binding domain. NTD: N-terminal domain of S1. TMD: transmembrane domain. Structural configurations adapted from [45,46,34].
Viruses 11 00074 g001
Table 1. MERS-CoV vaccine candidates grouped according to category.
Table 1. MERS-CoV vaccine candidates grouped according to category.
Vaccine TypeHumoral Response in:Cell-Mediated Response in:Protective in:Clinical TrialSource (s)
Subunit
RBDM, PM, PM, P [50,55,58]
S nanoparticlesM M [61]
Prefusion-locked SM [34]
NTDMMM [65]
DNA
pVax1-SM, P, CM, PPPhase I[71]
pVRC8400-S 1M, P P [74]
pcDNA3.1(+)-S1 or SMMM [75,77]
Viral Vector
VEEV-SM [38]
VEEV-N MM [82]
MVA-SM, CMM, CPhase I[88]
Ad5-S or S1MM [93,94]
Ad5-S 2MMM [96]
Ad41-SMM [94]
ChAdOx1-SMMMPhase I[102]
MVvac2-SMMM [104]
Newcastle-SM, C [107]
VSV-SM, PP [109]
Rabies-S1M M [111]
Bac-S,E,MPP [113]
Bac-RBD+VP2MM [114]
Whole
Formalin inactivatedM M [117]
MERS-ΔE [122]
MERS-dNSP16M M [127]
MERS-dORF3-5M M [129]
Humoral response denotes any antibody response generated, in most cases a NAb response. Cell-mediated responses denote T cell activation markers including IFN-γ. S: MERS-CoV spike protein. N: MERS-CoV nucleocapsid. RBD: receptor binding domain. NTD: N-terminal domain. S1: spike subdomain S1. Bac: baculovirus VLP. E: MERS-CoV envelope protein. M (under Vaccine Type): MERS-CoV membrane protein. VP2: canine parvovirus VP2 protein. M (under Protective in:): mouse. P: non-human primate. C: camel. 1 With S1 protein booster; 2 with S nanoparticles booster.

Share and Cite

MDPI and ACS Style

Schindewolf, C.; Menachery, V.D. Middle East Respiratory Syndrome Vaccine Candidates: Cautious Optimism. Viruses 2019, 11, 74. https://0-doi-org.brum.beds.ac.uk/10.3390/v11010074

AMA Style

Schindewolf C, Menachery VD. Middle East Respiratory Syndrome Vaccine Candidates: Cautious Optimism. Viruses. 2019; 11(1):74. https://0-doi-org.brum.beds.ac.uk/10.3390/v11010074

Chicago/Turabian Style

Schindewolf, Craig, and Vineet D. Menachery. 2019. "Middle East Respiratory Syndrome Vaccine Candidates: Cautious Optimism" Viruses 11, no. 1: 74. https://0-doi-org.brum.beds.ac.uk/10.3390/v11010074

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

Article Metrics

Back to TopTop