Skip to main content
Taylor & Francis - PMC COVID-19 Collection logoLink to Taylor & Francis - PMC COVID-19 Collection
. 2014 Jan 9;8(7):887–898. doi: 10.1586/erv.09.43

SARS vaccines: where are we?

Rachel L Roper 1,, Kristina E Rehm 2
PMCID: PMC7105754  PMID: 19538115

Abstract

In this review, the current state of vaccine development against human severe acute respiratory syndrome (SARS) coronavirus, focusing on recently published data is assessed. We discuss which strategies have been assessed immunologically and which have been evaluated in SARS coronavirus challenge models. We discuss inactivated vaccines, virally and bacterially vectored vaccines, recombinant protein and DNA vaccines, as well as the use of attenuated vaccines. Data regarding the correlates of protection, animal models and the available evidence regarding potential vaccine enhancement of SARS disease are discussed. While there is much evidence that various vaccine strategies against SARS are safe and immunogenic, vaccinated animals still display significant disease upon challenge. Current data suggest that intranasal vaccination may be crucial and that new or combination strategies may be required for good protective efficacy against SARS in humans.

Keywords: animal model, antibody, efficacy, SARS, T lymphocyte, vaccine


Severe acute respiratory syndrome (SARS) caused 8098 reported human infections and 774 deaths in 32 countries in a single fall-to-spring period (2002–2003), and also led to travel restrictions and significant effects on the global economy [201]. The etiologic agent of SARS was identified as a new human coronavirus (CoV), order Nidovirales, family Coronaviridae, by the sequencing of its genome [1,2] and by experimental infection of macaques to fulfill Koch’s postulates [3]. Serologic evidence suggests zoonotic transmission of SARS-associated CoV (SARS-CoV) into the human population for several years before the recognized outbreak [4], and transmission to humans has continued, resulting in at least four independent nonlaboratory-associated cases in 2004 [5–7,202]. The movement of SARS-CoV into the human population over several years suggests a need to prepare vaccines for protection from this potentially emerging agent. SARS-CoV is of particular concern as a zoonosis because it can replicate in a large number of animals including dogs, cats, pigs, mice, ferrets, foxes, monkeys and rats [8–10], in addition to Chinese palm civets, raccoon-dogs and bats, which appear to be the natural host [11,12].

SARS is primarily a respiratory disease, with the highest concentration of SARS-CoV found in the respiratory tract [13–15], although this virus is also detectable in other organs and tissues, as well as in stool [16–18]. The incubation period for the disease ranges from 2 to 10 days, and infectivity is maximal during the second week of disease [19,20]. The disease is characterized by fever, chills, malaise, dyspnea, cough, diarrhea and pneumonia [13–15]. Diffuse alveolar damage along with inflammatory cell infiltrate consisting particularly of macrophages are hallmarks found in SARS patients [21]. The fever of most patients abates within 2 weeks and is accompanied by resolution of chest symptoms and radiologic changes [3,13–15,22]. The major mode of transmission of SARS-CoV is believed to be through droplet spread [2,23], although SARS-CoV can remain viable when dried on surfaces for up to 6 days [24]. The majority of SARS patients are adults with only a few cases in children aged 15 years or younger [19,20,25]. The overall case–fatality rate is approximately 10% [19,203].

Currently, there are no approved antiviral drugs that effectively target SARS-CoV, hence vaccination is the most likely mode of preventing SARS in people, especially for those at highest risk (e.g., healthcare workers). A successful SARS vaccine could be used prophylactically to protect healthcare workers, laboratory personnel and other at-risk individuals. No vaccines are currently licensed for any of the human CoVs, but vaccines have been produced for a number of CoVs for use in chickens, cattle, dogs, cats and swine [26–28].

The positive-stranded RNA genome of SARS-CoV is 29.7 kb in length and contains approximately 14 open reading frames (ORFs), described in Table 1, with identification of each ORF by the four nomenclature systems [1,2]. These ORFs encode proteins that provide targets for vaccine and drug development. CoV enters target cells via receptor-mediated endocytosis driven by the spike (S) glycoprotein, which protrudes from the surface of the virion. The S protein serves as the major viral attachment protein, critical to virus binding and fusion of the viral envelope [29], and thus has been a major target antigen for vaccine development. The receptor–S protein interaction is a major determinant of species specificity and tissue tropism for CoV [30]. Angiotensin-converting enzyme 2 (ACE2) and CD209L were identified as functional receptors for SARS-CoV; however, entry through ACE2 is more efficient [31,32]. The receptor-binding domain of the S protein is a critical neutralization determinant.

Table 1. Severe acute respiratory virus-associated coronavirus open reading frames.

Protein Tor2 (GenBank) aa number (size) Gene nomenclature systems
    Qiu[139] Marra[1] Rota[2]
ORF 1a/b
S
1255
S
S
S
3a
274
PUP1
ORF3
X1
3b
154
PUP2
ORF4
X2
E
75
E
E
E
M
221
M
M
M
6
63
PUP3
ORF7
X3
7a
122
PUP4
ORF8
X4
7b
44
 
ORF9
 
8a
39
 
ORF10
 
8b
84
 
ORF11
X5
N
422
N
N
N
9b
98
PUP5
ORF13
 
Not designated 70   ORF14  

Bold ORFs have been experimentally deleted [103,106].

aa: Amino acid; E: Envelope; M: Matrix; ORF: Open reading frame.

Several strategies may be considered for vaccination against SARS-CoV, including an inactivated or whole-killed virus (WKV) vaccine, a live-attenuated SARS-CoV vaccine, a viral vector such as adenovirus (Ad) or vaccinia virus expressing SARS-CoV genes, bacterial vectors, recombinant SARS-CoV proteins or DNA vaccines. Live-attenuated CoV, killed CoV, DNA vaccines and viral vectored vaccines have all been used to successfully vaccinate against animal CoVs [28,33,34].

Inactivated virus vaccines

Inactivated, or WKV, vaccines are attractive because they are easily prepared (at least conceptually) and present an antigenic moiety similar to what the immune system will encounter in invading virus particles. In addition, these vaccines present multiple proteins on their surface for immune recognition (Figure 1). Antibodies were detected in patient sera to at least eight different proteins that may be in the viral particle membrane [35]. In addition to S, matrix (M) and envelope (E), four other ORFs (3a, 6, 7a and 7b) have been confirmed to encode additional structural proteins [36–40]. These data indicate that there are multiple epitopes and proteins that may be targets of protective antibodies. Mice vaccinated with inactivated SARS-CoV generated antibodies to a number of proteins including S, nucleocapsid (N), M and 3CL [41]. The main difficulties encountered with the production of inactivated vaccines are the biosafety level 3 growth of large amounts of pathogen and the difficulty ensuring that all virus has been successfully inactivated. For SARS-CoV, the large-scale production of UV-inactivated virus has been successfully described [42].

Figure 1. Severe acute respiratory syndrome-associated coronavirus virion.

Figure 1.

Known structural surface proteins including the viral attachment protein S, small E and M protein are shown in black text, as well as potential (gray text) predicted membrane proteins that may or may not be in the virion. Predicted orientations of the protein are also shown relative to their N, as well as the topology regarding the size of the protein mass relative to the membrane of the virus particle [1]. Nomenclature is shown as in Genbank, except for ORF14, which is designated as in the original severe acute respiratory syndrome (SARS) genome sequence manuscript [1]. For proteins that are primarily external, the number of predicted amino acids displayed on the surface of the virion is shown. Y indicates that antibodies to thiS protein have been detected in SARS patient sera [35].

E: Envelope; M: Membrane/matrix; N: Amino terminal; S: Spike.

A number of laboratories have pursued the development of inactivated whole SARS-CoV virus vaccines and demonstrated that they induce SARS-CoV neutralizing antibody [41,43–47]. However, demonstrations of efficacy against live SARS-CoV challenge are rare. In one study, WKV vaccine was shown to protect against pulmonary SARS-CoV replication in BALB/c mice, although characterization of the immune response was not reported [48]. Our consortium (The SARS Accelerated Vaccine Initiative) prepared a β-propiolactone-inactivated WKV SARS-CoV (Tor-2 strain) vaccine and compared its immunogenicity and efficacy to a combination of attenuated Ads expressing either S or N proteins for the ability to protect against live SARS-CoV challenge in a permissive mouse model [49]. Our results showed that the WKV vaccine, in the presence or absence of alum adjuvant, provided protection against live SARS-CoV challenge by the induction of high levels of neutralizing antibodies and the reduction of SARS-CoV load in the respiratory tract compared with mock-vaccinated mice [49]. CoV-like particles have also been developed by coexpression of SARS-CoV S protein with E, M and N proteins of mouse hepatitis virus, thus mimicking WKV. This preparation was shown to induce neutralizing antibodies and to protect mice against SARS-CoV replication in lungs [50], but there was no direct experimental comparison made with the WKV vaccine.

The lack of significant clinical disease in many mouse models, however, leads one to question whether efficacy would be maintained in a host where SARS is more virulent. WKV has also been tested in ferrets, a model that shows clinical signs and significant lung pathology [51,52]. Formalin-inactivated whole-virus Urbani strain of SARS-CoV without adjuvant induced some neutralizing antibodies, and led to earlier clearance of virus after challenge, but provided only mild protection in ferrets [53]. Lung tissues were analyzed 23 days postchallenge and did not show significant changes between mock and vaccinated animals, but this time point may have been too late to reveal vaccination-induced differences in disease. The authors commented, “the vaccine was not immunologically robust”. Our consortium also tested β-propiolactone-inactivated WKV and compared it with Ad-vectored S and N vaccines in ferrets [52], and found that both vaccines induced neutralizing antibody responses and reduced viral replication and shedding in the upper respiratory tract, and progression of virus to the lower respiratory tract. The vaccines also diminished hemorrhage in the thymus, and reduced the severity and extent of pneumonia and damage to lung epithelia. However, despite high neutralizing antibody titers, protection was incomplete for all vaccine preparations and administration routes tested.

Formaldehyde-inactivated SARS-CoV was used to vaccinate rhesus monkeys [54], and the vaccine was shown to be safe and immunogenic (it induced neutralizing antibodies). However, evidence of protection was lacking, as only one of the two PBS-vaccinated monkeys showed any clinical signs after challenge, and they were mild. Inactivated SARS-CoV vaccine has been administered to 36 human subjects and was found to be safe, well tolerated and able to elicit SARS-CoV-specific neutralizing antibodies; however, lacking a natural challenge, there is no data on efficacy [55]. While it is widely accepted that endogenous antigen production (via recombinant or attenuated bacterial or viral delivery) yields superior T-lymphocyte responses, we found that SARS-CoV-specific IFN-γ-secreting T-cell responses were similar in WKV and Ad vector S/N-vaccinated mice [49], suggesting that WKV may induce a T-cell response equal to vectored vaccines. Thus, the accumulated data indicate that WKV vaccines are safe and they induce SARS-CoV neutralizing antibodies and can even activate T lymphocytes; however, compelling evidence of protective efficacy is scant or absent.

Recombinant vector vaccines

Recombinant virus vaccines have several features that make them efficient in inducing B- and T-cell-mediated immune responses, including their ability to infect cells and persist in the body, their ability to infect antigen-presenting cells directly, and the fact that viral proteins and the infection itself can have strong adjuvant activity [56]. Recombinant viruses express the foreign target protein in the cytoplasm of the host cell, much like an intracellular pathogen. Thus, the endogenous antigen is available for processing by the cellular antigen-processing machinery for expression with MHC class I for presentation to CD8+ T lymphocytes and development of cytotoxic T cells. As a result, recombinant viruses result in activation of cellular immunity often necessary for elimination of infected cells. For SARS-CoV vaccines, several viruses have been used to express SARS-CoV proteins with the goal of inducing both strong cellular immunity and neutralizing antibodies.

Adenovirus vectored SARS-CoV vaccines

Some of the advantages of Ad vaccines include their lack of pathogenicity in humans, especially for replication-deficient mutants [57], oral or nasal administration, which promotes mucosal immunity, and the well-characterized genome of Ads [58]. Disadvantages of Ads compared with other viral delivery systems include their limited cloning capacity, the fact that human Ads have a restricted host range, often making animal testing difficult [59], and that a large percentage of the human population has pre-existing immunity against the vector due to natural infections [56]. The last difficulty may be circumvented by the use of prime–boost protocols where a different vector (e.g., DNA vaccine) is used to prime the immune response followed by a boost with a recombinant Ad [56].

First, it was demonstrated that Ad vectors encoding SARS proteins were immunogenic. Rhesus macaques immunized intramuscularly with a combination of three Ad5–SARS-CoV Ad-based vectors (N, S and M protein) all generated antibody responses against spike S1 fragment, SARS-CoV-neutralizing antibodies and T-cell responses against the N protein [60]. Vaccination of C57BL/6 mice with recombinant SARS N protein–Ad was able to induce SARS-CoV-specific IFN-γ secretion and T-cell proliferation, but not neutralizing antibodies [57].

Protection against SARS-CoV challenge by Ad-vectored vaccines was first tested in mice [49]. We developed recombinant Ad constructs expressing genes for either SARS-CoV S or N proteins, and immunized SARS-CoV-susceptible 129S6/SvEv mice [61] with both vectors combined, either intranasally or intramuscularly. The vaccine induced high levels of neutralizing antibody, anti-SARS-CoV N protein IFN-γ secretion, and significantly reduced viral titers and RNA in the lungs of challenged mice [49]. Interestingly, although the intramuscular route was more effective in inducing neutralizing SARS serum antibodies, the intranasal route of administration induced IgA and was more effective in blocking SARS-CoV replication in nose and lung tissue (1000-fold in nasal secretions). This finding suggests that the intranasal administration of recombinant Ad N and S proteins may induce crucial protective mucosal immunity. In the same study, we compared adeno-vectored S and N vaccines with a preparation of inactivated SARS-CoV. The combined adeno-S and -N vaccine induced significantly lower neutralizing antibodies and similar anti-N IFN-γ-secreting cells compared with inactivated SARS-CoV vaccine, but the inactivated vaccine provided superior protection measured as SARS-CoV lung titers and RNA [49]. We also compared these vaccines in a ferret model in which ferrets show clinical signs, including fever and lung damage [52]. Both the whole-killed SARS-CoV vaccine and the combination of Ads encoding N and S proteins induced neutralizing antibody responses, reduced viral replication in the respiratory tract, and decreased tissue damage in the thymus and lungs [52]. The adeno-S and -N vaccine delivered intranasally elicited a poor serum-neutralizing antibody response but provided the best protection from lung replication and lung damage [52], indicating that serum-neutralizing antibodies are not a sufficient measure of protective efficacy of a vaccine. In addition, despite high neutralizing antibody titers in some vaccines, protection was incomplete for all vaccine preparations (with one homologous boost) and administration routes, suggesting that combinations of vaccines may be necessary to provide adequate protection against SARS in susceptible animals and humans.

Adenovirus constructs expressing SARS-CoV S protein have also been evaluated in a ferret model using a heterologous prime–boost with human and chimpanzee Ads in order to avoid interference from the immune response to the first Ad vaccination during the boost. This vaccine regimen reduced viral load and the severity of pneumonia in ferrets, and it was also shown to be immunogenic in rhesus macaques [51].

Poxvirus vectors

Poxvirus recombinants are attractive as vaccine vectors owing to their ease of production, stability, capacity for encoding large genes, cytoplasmic gene expression, and ability to induce long-lasting cellular and humoral immune responses [56]. The replication-deficient poxvirus vector, modified vaccinia Ankara (MVA) strain, encoding SARS-CoV S protein delivered either intranasally or intramuscularly, induced neutralizing antibodies and reduced viral replication in the respiratory tract of challenged mice [62]. An MVA–S recombinant vaccine was also employed in one ferret study with apparent increased liver pathology in vaccinated groups after SARS-CoV challenge [63]. While these data suggest that liver pathology be evaluated in SARS-CoV vaccine studies, no other report has shown liver damage linked to vaccination [62].

Recombinant platform vaccines

Several other viral and bacterial vaccine platform technologies have been employed to encode S protein for SARS-CoV vaccine development. Monkeys vaccinated intranasally with parainfluenza encoding the SARS-CoV S protein produced neutralizing antibodies and had significantly reduced viral titers in the respiratory tract after challenge [64]. Parainfluenza virus encoding the S protein was also protective from SARS-CoV challenge in hamsters, and the inclusion of M and E proteins enhanced efficacy [65]. Recombinant adeno-associated virus encoding SARS-CoV S protein vaccine induced SARS-CoV neutralizing antibodies, T-cell responses, and decreased viral titers and lung damage in mice [66]. As with the Ad studies [49,52], intranasal administration led to IgA production and improved protection from SARS-CoV challenge. Newcastle disease virus, an avian-tropic virus that exhibits limited replication in primate respiratory tissues, was also used for SARS-CoV vaccination. Monkeys vaccinated with Newcastle disease virus expressing S protein had up to 1000-fold less virus in the lung tissue after SARS-CoV challenge [67]. A replication-defective vesicular stomatitis virus recombinant expressing the SARS-CoV S protein induced neutralizing antibodies and T-cell responses, and provided protection of immunized mice from SARS-CoV [68].

Several other recombinant strategies have been tested for immunogenicity but not yet for efficacy. For example, live-attenuated recombinant measles viruses expressing SARS-CoV S and N proteins both induced high antibody titers against their cognate antigen. Anti-S antibodies were SARS-CoV neutralizing and N protein induced specific cellular immune responses [69]. Rabies vector has been used to express S proteins and elicit neutralizing antibodies in mice [70]. Recombinant baculoviruses expressing N or S proteins induced both humoral and cellular immune responses in vaccinated mice [71]. Attenuated Salmonella expressing SARS-CoV N protein elicited cytotoxic T-lymphocyte activities and induced IFN-γ-producing T cells in mice. Interestingly, intranasal vaccination also showed advantages in this bacterial system, inducing the highest IgG and IgA levels [72].

Subunit vaccines: antigen targets & trials

Subunit vaccines comprised of purified antigen are advantageous owing to their safety and simplicity; however, protective efficacy is sometimes lacking. In particular, exogenously produced proteins are typically presented with MHC class II and thus often do not generate robust cytotoxic T-cell responses. CoV S proteins are the favorite targets in CoV subunit vaccine development since this viral protein contains determinants known to elicit protective immune responses [73,74]. Consequently, the SARS-CoV S glycoprotein, shown to be responsible for receptor binding to cellular ACE2, is an attractive target for the development of both vaccine and therapeutics [75,76]. This approach is strongly supported by the finding that a human monoclonal antibody that binds to the N-terminal of S protein potently neutralizes SARS-CoV infection and inhibits syncytia formation through blocking of receptor binding [77]. Moreover, the S protein has been shown to induce serum-neutralizing antibodies and confer protective immunity against SARS-CoV challenge in mice and African green monkeys [62,64,78]. Since several other proteins are also expressed on the surface of the virion (Figure 1) and elicit antibodies detectable in sera of convalescent SARS patients [35], other proteins may also be useful to augment protective immunity. For example, antibodies to M proteins have also been shown to have neutralizing activity [79]. In addition, it has also been shown that SARS-CoV S protein can generate CD4 and CD8 T-cell responses [37].

Studies from other animal CoV vaccines have also shown that the CoV N protein may represent another antigen candidate for vaccine development [27,80]. Although antibodies to CoV N proteins have no virus-neutralizing activity in vitro[79], there is evidence that the protein may provide in vivo protection by induction of cell-mediated immunity [73,81,82], as it has been shown to generate CoV-specific CD8+ T cells [82–85] and provide protection in animals following infection [84,86]. The expressed SARS-CoV N protein has been shown to be a vaccine candidate by inducing antigen-specific T-cell responses, but no in vivo protection experiments were performed with these vaccines [87,88]. A review was published recently addressing the progress in subunit vaccines [89].

DNA vaccines

For many pathogens, both antibody and T-cell-mediated immunity is a desirable outcome of vaccination, and generally only live-recombinant or -attenuated organism vaccines efficiently induce cellular immunity [90]. DNA vaccines, comprised of plasmid DNA encoding proteins from pathogens, have been demonstrated to induce both humoral and cellular immune responses, the latter due to the mimicking of the effects of live viruses, in that antigenic proteins are endogenously produced and efficiently presented by MHC class I, thus inducing CD8+ T-cell responses [90]. Furthermore, the stability, simplicity, safety and ease of manufacture make DNA vaccines an attractive alternative to the use of live vaccines [90]. Several DNA vaccine candidates have been reported for SARS-CoV proteins, including those for S [12,78,91–94], M [95] and N proteins [87,88], all of which can generate antibody and cellular immune responses [94].

A DNA vaccine expressing S protein induced both T-cell and neutralizing antibody responses, and reduced SARS-CoV replication in the lungs [78]. Furthermore, this study showed that protection was mediated by antibodies to the S gene, and was not T-cell dependent in mice [78]. Careful construction of the S plasmid (with splice sites and viral RNA export sequences) has now been shown to markedly increase efficacy of S-DNA vaccine in the mouse model [93], but these vaccines have not been tested in other animal models. A multiple-epitope DNA vaccine strategy elicited induction of antibody responses in mice to two epitopes, S (437–459) and M (1–20), which were able to neutralize SARS-CoV infectivity in vitro[95], but protection was not assessed.

Mice vaccinated with the N-DNA vaccine produced N-specific antibody and cytotoxic T-lymphocyte activity [87,96], although in one study this was also reported to induce a delayed-type hypersensitivity reaction, which might be problematic in a vaccine [96]. N-DNA vaccine, in which the N protein is expressed and linked to LAMP in order to enhance MHC class II presentation, increased memory responses [97]. DNA vaccination with SARS-CoV N protein linked to calreticulin to increase MHC class I presentation not only generated potent N-specific humoral and T-cell-mediated immune responses against N protein-expressing cells but also significantly reduced the titer of challenging vaccinia virus expressing the N protein [88]. These data suggest that such a response might also successfully target SARS-CoV-infected cells. A N-DNA vaccine candidate was also investigated in HLA-transgenic mice and elicited a specific CD8+ T-cell response in this model [98]. DNA vaccines expressing the M protein have also been shown to induce neutralizing antibody and cytotoxic T-lymphocyte activity in mice [99]. Interestingly, in a study comparing S-, M- and N-DNA vaccines, M gave the strongest T-cell response [94].

Although DNA vaccines show great promise in preclinical models, their efficacy in clinical studies has often been disappointing. Thus, various prime–boost strategies have been developed that increase efficacy. DNA vaccination may be performed in conjunction with a heterologous prime or boost with proteins, inactivated viral vaccine candidates or viral vectors [12,37,95,100,101]. These strategies often provide superior immune responses and can also determine the magnitude and type of immune response (e.g., Th1/Th2). While DNA vaccines for SARS may hold promise, evidence of protection in a good SARS animal model is needed.

Attenuated vaccines

The most long-lasting and protective vaccines are those comprised of an attenuated pathogen or a closely related but avirulent live virus, such as the use of the naturally occurring vaccinia virus, a low-virulence member of the same genus, to vaccinate against smallpox. These vaccines are more efficacious due to their persistence in the host, possession of pathogen-encoded immune-activating moieties, and their appropriate location both in the body and in the cell, yielding endogenouS protein production and efficient MHC class I presentation, generating a robust cytotoxic T-cell response. The difficulty with attenuated vaccines is that attenuating point mutations may revert causing virulence, and deletion-attenuated mutants may recombine with naturally occurring environmental wild viruses to regain virulence, as has been seen with the attenuated oral poliovirus. Given safety concerns, it is often difficult to gain regulatory approval of attenuated vaccines without strong proof that the threat of disease is sufficient to warrant the use of such a vaccine. For SARS, this threshold has not yet been met, but some interesting attenuated mutants have been developed. The immunogenicity and protective efficacy of a live-attenuated vaccine consisting of a recombinant SARS-CoV lacking the small E gene were studied. Deletion of E causes reduced viral morphogenesis and virus titers in vitro and in vivo, and thus attenuates the virus [102,103]. Hamsters immunized with this deletion mutant developed high levels of serum-neutralizing antibodies and were protected from clinical signs (decreased activity) and replication of homologous (SARS-CoV, Urbani) and heterologous (GD03) SARS-CoV in the upper and lower respiratory tract [104]. Thus, deletion of the structural E gene may be a first step toward development of a live-attenuated SARS-CoV vaccine. The deletion of the nsp-1 gene in the related CoV mouse hepatitis virus (MHV) has been shown to create a highly efficacious attenuated vaccine, suggesting that this approach may also be attempted for the development of a SARS-attenuated vaccine [105]. Two comprehensive studies on gene deletion and attenuation effects showed that deletion of ORFs 3a, 3b, 6, 7a, 7b, 8a, 8b or 9b (highlighted in Table 1) had little or no effect on viral replication both in vitro and in vivo[103,106]. It is not known if deletion of these genes might have an attenuating effect in higher mammals. However, given the disappointing protection afforded by most SARS-CoV vaccine strategies explored to date, it seems that further exploration of attenuated SARS-CoV vaccines is justified. In that vein, exploration has been undertaken on the effects of rearrangement of the SARS-CoV genome, which has been shown to be attenuating in MHV [107]. This system has the additional advantage of making recombination of the vaccine with wild CoVs (thus restoring virulence) less likely. Other strategies to protect from virulence-restoring recombination events include multiple attenuating gene knockouts, with or without growth of the virus in trans-complementing cells lines, and replacement of transcriptional regulatory sequences (analogous to promoters in most systems) with sequences incompatible for wild-type gene expression. These strategies have been fully and elegantly described in a recent review [108].

Animal models

Animal models developed for SARS include macaques [3,109], African green monkey [64,110], ferrets [9], mice [111,112] and hamsters [113], and the Chinese masked palm civet [114]. An excellent review of animal models was recently released [115], and we will focus here on some of the newer developments, especially in ferrets.

Mouse models are of questionable use for efficacy studies as they do not reproduce the clinical signs or severe disease of SARS in humans [111,112], unless immunodeficient or aged mice are used [116]. However, the model has been improved by the use of mouse-adapted SARS and human ACE2 transgenic mice, although both models still have significant caveats [115,117–119]. Hamsters also do not exhibit clinical signs of SARS-CoV infection. Ferrets have been used widely for the study of influenza and are susceptible to SARS-CoV infection, with lung pathology and virus shedding [9,120]. One ferret study indicated that upon intranasal administration of SARS-CoV Toronto 2 strain, no clinical signs were observed, although viral RNA could be detected in pharyngeal swabs [63,121]. However, several other studies showed the ferret to be one of the better models for the display of clinical signs, viral replication and lung pathology [51], reflecting SARS pathogenesis in humans [122].

In the most recently reported ferret study[52], SARS-CoV challenge resulted in ferrets with clinical signs of infection (elevated temperature, nasal discharge and sneezing). No other animal, including cynomolgus macaques, has been reported to regularly experience fever, which is the most common sign in human SARS-CoV infection, (>99%) [123,124]. Thus, ferrets are a good model for SARS-CoV because they support replication in the upper and lower respiratory tracts, develop clinical disease, shed virus from the upper airway and develop severe lung pathology. Ferrets are also outbred, allowing the assessment of a range of individual responses that are documented in human SARS. Finally, the ferret model is a nonrodent model and is significantly less expensive and difficult to study than nonhuman primates. The main disadvantages of this model are that the ferret immune system is not well defined, there is a dearth of reagents and, as they are outbred, larger numbers are needed to assess statistical significance.

Vaccine enhancement of disease

The greatest fear among vaccinologists is the creation of a vaccine that is not only ineffective, but which exacerbates disease. Unfortunately, CoV vaccines have a history of enhancing disease, notably with feline CoVs [80,125]. While several mechanisms may exist, the best understood is antibody-mediated entry of virus into cells via immunoglobulin Fc receptors. This has been demonstrated to occur for the SARS-CoV S protein in human B-cell lines [126], however, the same group showed that SARS-CoV S protein-vaccinated animals showed no signs of enhanced lung pathology or hepatitis, and indeed that the viral load was reduced following challenge with SARS-CoV; although hamsters may not respond in a way similar to humans immunologically [126]. Other groups have also shown that administration of anti-S antibody does not enhance disease upon SARS-CoV challenge in mice or ferrets [78], again suggesting that antibody is not enhancing disease in SARS-CoV infection.

There have only been two reports of possible vaccine-induced pathology in SARS vaccine trials to date. In one study, ferrets vaccinated with the poxvirus vector MVA expressing SARS-CoV S protein displayed increased liver pathology after challenge [63] compared to other groups, but liver pathology has not been increased with any other SARS-CoV vaccines. In our ferret vaccine trials using S and N proteins, we noted a delayed histopathology in vaccinated groups, but no increase in pathology compared to unvaccinated groups [52]. The lack of vaccine enhancement of disease is further supported by a recent study in WKV-vaccinated and challenged ferrets that were followed for 3 weeks [53]; however, the exact details and combination of vaccine vector and antigen may control this phenomenon. In the other study that raised vaccine safety concerns, vaccination with the N protein expressed in Venezuelan equine encephalitis virus replicon particles was reported to increase eosinophilic infiltration and damage in lungs of mice challenged with SARS-CoV [127]. This has not been reported in other studies, and in our studies with combination S and N protein expressed in Ad, no eosinophillic infiltration was noted in mice or ferrets [49,52]. While each vaccine and antigenic combination must be thoroughly evaluated for safety and efficacy, the overall picture for SARS-CoV vaccines shows no particular reason for concern with vaccine enhancement of disease. In the cases where it has been reported, it appears to be confined to a particular expression system rather than specifically related to any antigen. In the vast majority of studies, immunogenicity has been elicited without any negative impact on health after challenge with the virulent pathogen.

What mediates protection

Individuals convalescing from SARS develop high titers of neutralizing antibodies [128], and the appearance of antibodies coincides with the onset of resolution of SARS pneumonia [129,130]. In addition, antibodies to SARS S glycoprotein or whole SARS-CoV administered in several animal models have been shown to prevent or reduce SARS-CoV replication and disease [48,62,68,78,94,111,113,120,131–133]. However, we have shown that, while inactivated SARS with alum vaccine induced 15-fold higher serum-neutralizing antibody titers than the other vaccines (Ad-vectored), this vaccine did not universally protect ferrets better from SARS-CoV challenge [52]. These data provide a cautionary note about SARS rodent models, and indicate that the induction of strong neutralizing antibodies does not equate with protective efficacy in a relevant animal model where clinical signs are apparent and significant lung damage is seen. The ability of an antibody to neutralize virus infection is the easiest activity to measure, but it is not the only important function of antibody in antiviral defense. An important lesson may be drawn from the field of poxvirology where one of the most protective antigens (A33R) generates protective antibodies that are not neutralizing [134–136].

Several reports have indicated that intranasal vaccination may provide superior protection compared to other routes. A protollin-formulated SARS S protein delivered intranasally protected mice from SARS-CoV replication in the lung better than the same vaccine delivered intramuscularly, despite comparable serum levels of neutralizing anti-SARS IgG [137]. Presumably, this is due to the induction of IgA, which was detected only in the intranasally vaccinated animals. We found similar results in both mice and ferrets; the same vaccine given intranasally and intramuscularly gave stronger protection when delivered by the intranasal route, particularly in terms of viral load in the lung and shedding in nasal secretions [49,52]. Intranasal administration increases protection, despite greatly reduced serum antibody responses. These data underscore the importance of mucosal immunity.

Evaluation of T-lymphocyte responses in SARS protection has been problematic. Rodents may not provide an adequate disease model, and other models are difficult to evaluate because animals are outbread and there are inadequate reagents for measuring T-cell responses. Further studies are needed to determine the relative contributions of humoral and cell-mediated immunity in protection from SARS disease. Interestingly, in a study comparing S-, M- and N-DNA vaccines, M generated the strongest T-cell responses [94], and recovered SARS patients have long-lasting CD4 and CD8 memory to the M antigen [138]. These data suggest that further research should be directed toward evaluating the potential efficacy of the M antigen, as well as other viral proteins.

Conclusion

Much research to elucidate potential antigens, routes of vaccination, and methods for the design of SARS vaccines has been completed. Immunogenicity has been widely demonstrated, but identification of correlates of protection, and generation of immune responses that protect from clinical signs and lung damage remain elusive. Results suggest that a protective SARS vaccine should be possible; however, protection in mammals that are susceptible to severe disease (e.g., ferrets and humans) may be more difficult than the mouse models suggest. To date, the data indicate that the most efficacious vaccine strategy might be a heterologous combination of intranasal and systemic vaccination, since each delivers different aspects of protection. Given the incomplete protection of current vaccines, it seems unwise to discount T-cell responses, which have not been adequately evaluated, or the protection that might be afforded by the inclusion of additional viral proteins (especially those displayed on the virion and on the surface of infected cells) in SARS-CoV vaccine development.

Expert commentary & five-year view

Since protection from disease has not been demonstrated for any vaccine tested in an animal model that mimics human SARS disease, further vaccine development to improve vaccine efficacy is needed. While it is clear that antibodies to S protein offer some protection against SARS, it is equally apparent that high neutralizing antibody titers are not sufficient to protect animals from serious tissue damage after SARS-CoV challenge. Thus, it will likely be necessary to generate protective T-lymphocyte responses or antibodies to other SARS proteins, or to improve protection. Data collected thus far suggest that strategies including mucosal immunizations coupled with a heterologous systemic route of vaccination may improve efficacy. Alternatively, the development of safe attenuated SARS vaccines may be able to offer both the quality and quantity of immune response required to stop serious SARS-CoV-induced tissue damage. It will be necessary to conduct more trials with direct comparison of vaccines (and combinations of prime–boost) in appropriate animal models that more closely mimic human disease course. It will also be important to pursue an understanding of the role of T-cell immunity against SARS, since little is known at this time. Development of improved T-cell analysis reagents for ferrets will aid in this endeavor. The rate of SARS vaccine progress in the next 5 years likely depends on the perceived disease threat from SARS. If another epidemic or pandemic occurs, funding and research for vaccine development will be a priority. Likewise, efficacy in humans can only be demonstrated if there is another SARS outbreak among a population of vaccinated and control subjects.

Key issues

  • • Severe acute respiratory syndrome (SARS) caused thousands of human infections worldwide and hundreds of deaths in just a few months, and severely impacted the global economy. Evidence indicates that SARS-coronavirus (CoV) has continued to jump from animals to humans over several years, suggesting that another pandemic may occur.

  • • While SARS patients generated antibodies to multiple SARS proteins and antiviral T-lymphocyte responses, correlates of protective immunity are unknown.

  • • Inactivated virus, viral vector, bacterial vector, subunit and DNA vaccines encoding several antigens have been developed and shown to be immunogenic.

  • • Vaccine trials in animal models that mimic human disease indicate that no vaccine strategy tested to date would satisfactorily protect from disease, arguing for further vaccine development research, especially focusing on:
    • – Mucosal immunization, which appears to be important for protection
    • – Understanding T-cell immunity, which has been difficult to analyze
    • – Combinations of heterologous vaccines in prime–boost regimens or attenuated SARS-CoV vaccines.

Acknowledgements

The authors wish to thank Tom Voss (Tulane University, LA, USA) for helpful discussions.

Financial & competing interests disclosure

The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.

No writing assistance was utilized in the production of this manuscript.

References

Papers of special note have been highlighted as: • of interest •• of considerable interest

  • 1.Marra MA, Jones SJ, Astell CR et al. The genome sequence of the SARS-associated coronavirus. Science 300(5624), 1399–1404 (2003). [DOI] [PubMed] [Google Scholar]
  • 2.Rota PA, Oberste MS, Monroe SS et al. Characterization of a novel coronavirus associated with severe acute respiratory syndrome. Science 300(5624), 1394–1399 (2003). [DOI] [PubMed] [Google Scholar]
  • 3.Fouchier RA, Kuiken T, Schutten M et al. Aetiology: Koch’s postulates fulfilled for SARS virus. Nature 423(6937), 240 (2003). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Zheng BJ, Wong KH, Zhou J et al. SARS-related virus predating SARS outbreak, Hong Kong. Emerg. Infect. Dis. 10(2), 176–178 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Fleck F. SARS virus returns to China as scientists race to find effective vaccine. Bull. World Health Organ. 82(2), 152–153 (2004). [PMC free article] [PubMed] [Google Scholar]
  • 6.Guan YJ, Tang XP, Zhang FC et al. Study of laboratory characteristic of 4 patients with sporadic severe acute respiratory syndrome in 2004. Zhongguo Wei Zhong Bing Ji Jiu Yi Xue 17(6), 332–334 (2005). [PubMed] [Google Scholar]
  • 7.Che XY, Di B, Zhao GP et al. A patient with asymptomatic severe acute respiratory syndrome (SARS) and antigenemia from the 2003–2004 community outbreak of SARS in Guangzhou, China. Clin. Infect. Dis. 43(1), e1–e5 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Chen W, Yan M, Yang L et al. SARS-associated coronavirus transmitted from human to pig. Emerg. Infect. Dis. 11(3), 446–448 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Martina BE, Haagmans BL, Kuiken T et al. Virology: SARS virus infection of cats and ferrets. Nature 425(6961), 915 (2003). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Wang M, Jing HQ, Xu HF et al. Surveillance on severe acute respiratory syndrome associated coronavirus in animals at a live animal market of Guangzhou in 2004. Zhonghua Liu Xing Bing Xue Za Zhi 26(2), 84–87 (2005). [PubMed] [Google Scholar]
  • 11.Guan Y, Zheng BJ, He YQ et al. Isolation and characterization of viruses related to the SARS coronavirus from animals in southern China. Science 302(5643), 276–278 (2003). [DOI] [PubMed] [Google Scholar]
  • 12.Lau SK, Woo PC, Li KS et al. Severe acute respiratory syndrome coronavirus-like virus in Chinese horseshoe bats. Proc. Natl Acad. Sci. USA 102(39), 14040–14045 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Drosten C, Gunther S, Preiser W et al. Identification of a novel coronavirus in patients with severe acute respiratory syndrome. N. Engl. J. Med. 348(20), 1967–1976 (2003). [DOI] [PubMed] [Google Scholar]
  • 14.Ksiazek TG, Erdman D, Goldsmith CS et al. A novel coronavirus associated with severe acute respiratory syndrome. N. Engl. J. Med. 348(20), 1953–1966 (2003). [DOI] [PubMed] [Google Scholar]
  • 15.Peiris JS, Lai ST, Poon LL et al. Coronavirus as a possible cause of severe acute respiratory syndrome. Lancet 361(9366), 1319–1325 (2003). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Peiris JS, Chu CM, Cheng VC et al. Clinical progression and viral load in a community outbreak of coronavirus-associated SARS pneumonia: a prospective study. Lancet 361(9371), 1767–1772 (2003). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Yam WC, Chan KH, Poon LL et al. Evaluation of reverse transcription-PCR assays for rapid diagnosis of severe acute respiratory syndrome associated with a novel coronavirus. J. Clin. Microbiol. 41(10), 4521–4524 (2003). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Ding Y, He L, Zhang Q et al. Organ distribution of severe acute respiratory syndrome (SARS) associated coronavirus (SARS-CoV) in SARS patients: implications for pathogenesis and virus transmission pathways. J. Pathol. 203(2), 622–630 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Wang JT, Chang SC. Severe acute respiratory syndrome. Curr. Opin. Infect. Dis. 17(2), 143–148 (2004). [DOI] [PubMed] [Google Scholar]
  • 20.Knudsen TB, Kledal TN, Andersen O, Eugen-Olsen J, Kristiansen TB. Severe acute respiratory syndrome – a new coronavirus from the Chinese dragon’s lair. Scand. J. Immunol. 58(3), 277–284 (2003). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Nicholls J, Dong XP, Jiang G, Peiris M. SARS: clinical virology and pathogenesis. Respirology 8(Suppl.), S6–S8 (2003). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Lee N, Hui D, Wu A et al. A major outbreak of severe acute respiratory syndrome in Hong Kong. N. Engl. J. Med. 348(20), 1986–1994 (2003). [DOI] [PubMed] [Google Scholar]
  • 23.Pearson H, Clarke T, Abbott A, Knight J, Cyranoski D. SARS: what have we learned? Nature 424(6945), 121–126 (2003). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Rabenau HF, Cinatl J, Morgenstern B et al. Stability and inactivation of SARS coronavirus. Med. Microbiol. Immunol. (Berl.) 194(1–2), 1–6 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Hon KL, Leung CW, Cheng WT et al. Clinical presentations and outcome of severe acute respiratory syndrome in children. Lancet 361(9370), 1701–1703 (2003). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Olsen CW, Corapi WV, Jacobson RH et al. Identification of antigenic sites mediating antibody-dependent enhancement of feline infectious peritonitis virus infectivity. J. Gen. Virol. 74(Pt 4), 745–749 (1993). [DOI] [PubMed] [Google Scholar]
  • 27.Anton IM, Gonzalez S, Bullido MJ et al. Cooperation between transmissible gastroenteritis coronavirus (TGEV) structural proteins in the in vitro induction of virus-specific antibodies. Virus Res. 46(1–2), 111–124 (1996). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Cavanagh D. Severe acute respiratory syndrome vaccine development: experiences of vaccination against avian infectious bronchitis coronavirus. Avian Pathol. 32(6), 567–582 (2003). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Lai MM, Holmes KV. In: Fields Virology Knipe DM, Howley PM (Eds). Lippincott-Raven, PA, USA, 1163–1203 (2001). [Google Scholar]
  • 30.Lai MMC, Perlman S, Anderson LJ. Coronaviridae. In: Fields Virology Knipe DM, Howley PM (Eds). Lippincott Williams & Wilkins, PA, USA 1305–1335 (2007). [Google Scholar]
  • 31.Wong SK, Li W, Moore MJ, Choe H, Farzan M. A 193-amino acid fragment of the SARS coronavirus S protein efficiently binds angiotensin-converting enzyme 2. J. Biol. Chem. 279(5), 3197–3201 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Jeffers SA, Tusell SM, Gillim-Ross L et al. CD209L (L-SIGN) is a receptor for severe acute respiratory syndrome coronavirus. Proc. Natl Acad. Sci. USA, 101(44), 15748–15753 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Holmes KV. SARS coronavirus: a new challenge for prevention and therapy. J. Clin. Invest. 111(11), 1605–1609 (2003). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Navas-Martin SR, Weiss S. Coronavirus replication and pathogenesis: implications for the recent outbreak of severe acute respiratory syndrome (SARS), and the challenge for vaccine development. J. Neurovirol. 10(2), 75–85 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Guo JP, Petric M, Campbell W, McGeer PL. SARS corona virus peptides recognized by antibodies in the sera of convalescent cases. Virology 324(2), 251–256 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]; • Other potential targets for severe acute respiratory syndrome (SARS) vaccine development.
  • 36.Kamitani W, Narayanan K, Huang C et al. Severe acute respiratory syndrome coronavirus nsp1 protein suppresses host gene expression by promoting host mRNA degradation. Proc. Natl Acad. Sci. USA 103(34), 12885–12890 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Huang J, Cao Y, Du J et al. Priming with SARS CoV S DNA and boosting with SARS CoV S epitopes specific for CD4+ and CD8+ T cells promote cellular immune responses. Vaccine 25(39–40), 6981–6991 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Ito N, Mossel EC, Narayanan K et al. Severe acute respiratory syndrome coronavirus 3a protein is a viral structural protein. J. Virol. 79(5), 3182–3186 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Schaecher SR, Mackenzie JM, Pekosz A. The ORF7b protein of severe acute respiratory syndrome coronavirus (SARS-CoV) is expressed in virus-infected cells and incorporated into SARS-CoV particles. J. Virol. 81(2), 718–731 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Shen S, Lin PS, Chao YC et al. The severe acute respiratory syndrome coronavirus 3a is a novel structural protein. Biochem. Biophys. Res. Commun. 330(1), 286–292 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Xiong S, Wang YF, Zhang MY et al. Immunogenicity of SARS inactivated vaccine in BALB/c mice. Immunol. Lett. 95(2), 139–143 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Tsunetsugu-Yokota Y. Large-scale preparation of UV-inactivated SARS coronavirus virions for vaccine antigen. Methods Mol. Biol. 454, 1–8 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.He Y, Zhou Y, Siddiqui P, Jiang S. Inactivated SARS-CoV vaccine elicits high titers of spike protein-specific antibodies that block receptor binding and virus entry. Biochem. Biophys. Res. Commun. 325(2), 445–452 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Takasuka N, Fujii H, Takahashi Y et al. A subcutaneously injected UV-inactivated SARS coronavirus vaccine elicits systemic humoral immunity in mice. Int. Immunol. 16(10), 1423–1430 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Tang L, Zhu Q, Qin E et al. Inactivated SARS-CoV vaccine prepared from whole virus induces a high level of neutralizing antibodies in BALB/c mice. DNA Cell. Biol. 23(6), 391–394 (2004). [DOI] [PubMed] [Google Scholar]
  • 46.Qu D, Zheng B, Yao X et al. Intranasal immunization with inactivated SARS-CoV (SARS-associated coronavirus) induced local and serum antibodies in mice. Vaccine 23(7), 924–931 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Zhang CH, Lu JH, Wang YF et al. Immune responses in Balb/c mice induced by a candidate SARS-CoV inactivated vaccine prepared from F69 strain. Vaccine 23(24), 3196–3201 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Stadler K, Roberts A, Becker S et al. SARS vaccine protective in mice. Emerg. Infect. Dis. 11(8), 1312–1314 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.See RH, Zakhartchouk AN, Petric M et al. Comparative evaluation of two severe acute respiratory syndrome (SARS) vaccine candidates in mice challenged with SARS coronavirus. J. Gen. Virol. 87(Pt 3), 641–650 (2006). [DOI] [PubMed] [Google Scholar]
  • 50.Lokugamage KG, Yoshikawa-Iwata N, Ito N et al. Chimeric coronavirus-like particles carrying severe acute respiratory syndrome coronavirus (SCoV) S protein protect mice against challenge with SCoV. Vaccine 26(6), 797–808 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Kobinger GP, Figueredo JM, Rowe T et al. Adenovirus-based vaccine prevents pneumonia in ferrets challenged with the SARS coronavirus and stimulates robust immune responses in macaques. Vaccine 25(28), 5220–5231 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.See RH, Petric M, Lawrence DJ et al. Severe acute respiratory syndrome vaccine efficacy in ferrets: whole killed virus and adenovirus-vectored vaccines. J. Gen. Virol. 89(Pt 9), 2136–2146 (2008). [DOI] [PubMed] [Google Scholar]; • Recent data on ferrets as a model for SARS infection and vaccine testing, including data on the importance of mucosal immunity.
  • 53.Darnell ME, Plant EP, Watanabe H et al. Severe acute respiratory syndrome coronavirus infection in vaccinated ferrets. J. Infect. Dis. 196(9), 1329–1338 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Zhou J, Wang W, Zhong Q et al. Immunogenicity, safety, and protective efficacy of an inactivated SARS-associated coronavirus vaccine in rhesus monkeys. Vaccine 23(24), 3202–3209 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Lin JT, Zhang JS, Su N et al. Safety and immunogenicity from a Phase I trial of inactivated severe acute respiratory syndrome coronavirus vaccine. Antivir. Ther. 12(7), 1107–1113 (2007). [PubMed] [Google Scholar]
  • 56.Rocha CD, Caetano BC, Machado AV, Bruna-Romero O. Recombinant viruses as tools to induce protective cellular immunity against infectious diseases. Int. Microbiol. 7(2), 83–94 (2004). [PubMed] [Google Scholar]
  • 57.Zakhartchouk AN, Viswanathan S, Mahony JB, Gauldie J, Babiuk LA. Severe acute respiratory syndrome coronavirus nucleocapsid protein expressed by an adenovirus vector is phosphorylated and immunogenic in mice. J. Gen. Virol. 86(Pt 1), 211–215 (2005). [DOI] [PubMed] [Google Scholar]
  • 58.Rolph MS, Ramshaw IA. Recombinant viruses as vaccines and immunological tools. Curr. Opin. Immunol. 9(4), 517–524 (1997). [DOI] [PubMed] [Google Scholar]
  • 59.Imler JL. Adenovirus vectors as recombinant viral vaccines. Vaccine 13(13), 1143–1151 (1995). [DOI] [PubMed] [Google Scholar]
  • 60.Gao W, Tamin A, Soloff A et al. Effects of a SARS-associated coronavirus vaccine in monkeys. Lancet 362(9399), 1895–1896 (2003). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Hogan RJ, Gao G, Rowe T et al. Resolution of primary severe acute respiratory syndrome-associated coronavirus infection requires stat1. J. Virol. 78(20), 11416–11421 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Bisht H, Roberts A, Vogel L et al. Severe acute respiratory syndrome coronavirus spike protein expressed by attenuated vaccinia virus protectively immunizes mice. Proc. Natl Acad. Sci. USA 101(17), 6641–6646 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Czub M, Weingartl H, Czub S, He R, Cao J. Evaluation of modified vaccinia virus Ankara based recombinant SARS vaccine in ferrets. Vaccine 23(17–18), 2273–2279 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Bukreyev A, Lamirande EW, Buchholz UJ et al. Mucosal immunisation of African green monkeys (Cercopithecus aethiops) with an attenuated parainfluenza virus expressing the SARS coronavirus spike protein for the prevention of SARS. Lancet 363(9427), 2122–2127 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Buchholz UJ, Bukreyev A, Yang L et al. Contributions of the structural proteins of severe acute respiratory syndrome coronavirus to protective immunity. Proc. Natl Acad. Sci. USA 101(26), 9804–9809 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Du L, Zhao G, Lin Y et al. Intranasal vaccination of recombinant adeno-associated virus encoding receptor-binding domain of severe acute respiratory syndrome coronavirus (SARS-CoV) spike protein induces strong mucosal immune responses and provides long-term protection against SARS-CoV infection. J. Immunol. 180(2), 948–956 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.DiNapoli JM, Kotelkin A, Yang L et al. Newcastle disease virus, a host range-restricted virus, as a vaccine vector for intranasal immunization against emerging pathogens. Proc. Natl Acad. Sci. USA 104(23), 9788–9793 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Kapadia SU, Simon ID, Rose JK. SARS vaccine based on a replication-defective recombinant vesicular stomatitis virus is more potent than one based on a replication-competent vector. Virology 376(1), 165–172 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Liniger M, Zuniga A, Tamin A et al. Induction of neutralising antibodies and cellular immune responses against SARS coronavirus by recombinant measles viruses. Vaccine 26(17), 2164–2174 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Faber M, Lamirande EW, Roberts A et al. A single immunization with a rhabdovirus-based vector expressing severe acute respiratory syndrome coronavirus (SARS-CoV) S protein results in the production of high levels of SARS-CoV-neutralizing antibodies. J. Gen. Virol. 86(Pt 5), 1435–1440 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Bai B, Lu X, Meng J et al. Vaccination of mice with recombinant baculovirus expressing spike or nucleocapsid protein of SARS-like coronavirus generates humoral and cellular immune responses. Mol. Immunol. 45(4), 868–875 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Luo F, Feng Y, Liu M et al. Type IVB pilus operon promoter controlling expression of the severe acute respiratory syndrome-associated coronavirus nucleocapsid gene in Salmonella enterica serovar Typhi elicits full immune response by intranasal vaccination. Clin. Vaccine Immunol. 14(8), 990–997 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Enjuanes L, Smerdou C, Castilla J et al. Development of protection against coronavirus induced diseases. A review. Adv. Exp. Med. Biol. 380, 197–211 (1995). [DOI] [PubMed] [Google Scholar]
  • 74.Navas-Martin S, Weiss SR. SARS: lessons learned from other coronaviruses. Viral Immunol. 16(4), 461–474 (2003). [DOI] [PubMed] [Google Scholar]
  • 75.See R, Roper RL, Brunham RC, Finlay BB. Rapid response research – SARS coronavirus vaccines and application of processes to other emerging infectious diseases. Curr. Immunol. Rev. 1(2), 185–200 (2005). [Google Scholar]
  • 76.Li W, Moore MJ, Vasilieva N et al. Angiotensin-converting enzyme 2 is a functional receptor for the SARS coronavirus. Nature 426(6965), 450–454 (2003). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Berger A, Drosten C, Doerr HW, Sturmer M, Preiser W. Severe acute respiratory syndrome (SARS) – paradigm of an emerging viral infection. J. Clin. Virol. 29(1), 13–22 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Yang ZY, Kong WP, Huang Y et al. A DNA vaccine induces SARS coronavirus neutralization and protective immunity in mice. Nature 428(6982), 561–564 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Pang H, Liu Y, Han X et al. Protective humoral responses to severe acute respiratory syndrome-associated coronavirus: implications for the design of an effective protein-based vaccine. J. Gen. Virol. 85(Pt 10), 3109–3113 (2004). [DOI] [PubMed] [Google Scholar]
  • 80.Olsen CW. A review of feline infectious peritonitis virus: molecular biology, immunopathogenesis, clinical aspects, and vaccination. Vet. Microbiol. 36(1–2), 1–37 (1993). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Wesseling JG, Godeke GJ, Schijns VE et al. Mouse hepatitis virus spike and nucleocapsid proteins expressed by adenovirus vectors protect mice against a lethal infection. J. Gen. Virol. 74, 2061–2069 (1993). [DOI] [PubMed] [Google Scholar]
  • 82.Stohlman SA, Bergmann CC, van der Veen RC, Hinton DR. Mouse hepatitis virus-specific cytotoxic T lymphocytes protect from lethal infection without eliminating virus from the central nervous system. J. Virol. 69(2), 684–694 (1995). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Stohlman SA, Kyuwa S, Polo JM et al. Characterization of mouse hepatitis virus-specific cytotoxic T cells derived from the central nervous system of mice infected with the JHM strain. J. Virol. 67(12), 7050–7059 (1993). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Seo SH, Wang L, Smith R, Collisson EW. The carboxyl-terminal 120-residue polypeptide of infectious bronchitis virus nucleocapsid induces cytotoxic T lymphocytes and protects chickens from acute infection. J. Virol. 71(10), 7889–7894 (1997). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Boots AM, Kusters JG, van Noort JM et al. Localization of a T-cell epitope within the nucleocapsid protein of avian coronavirus. Immunology 74(1), 8–13 (1991). [PMC free article] [PubMed] [Google Scholar]
  • 86.Collisson EW, Pei J, Dzielawa J, Seo SH. Cytotoxic T lymphocytes are critical in the control of infectious bronchitis virus in poultry. Dev. Comp. Immunol. 24(2–3), 187–200 (2000). [DOI] [PubMed] [Google Scholar]
  • 87.Zhu MS, Pan Y, Chen HQ et al. Induction of SARS-nucleoprotein-specific immune response by use of DNA vaccine. Immunol. Lett. 92(3), 237–243 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Kim TW, Lee JH, Hung CF et al. Generation and characterization of DNA vaccines targeting the nucleocapsid protein of severe acute respiratory syndrome coronavirus. J. Virol. 78(9), 4638–4645 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Du L, He Y, Jiang S, Zheng BJ. Development of subunit vaccines against severe acute respiratory syndrome. Drugs Today (Barc.) 44(1), 63–73 (2008). [DOI] [PubMed] [Google Scholar]
  • 90.Gurunathan S, Klinman DM, Seder RA. DNA vaccines: immunology, application, and optimization. Annu. Rev. Immunol. 18, 927–974 (2000). [DOI] [PubMed] [Google Scholar]
  • 91.Zeng F, Chow KY, Hon CC et al. Characterization of humoral responses in mice immunized with plasmid DNAs encoding SARS-CoV spike gene fragments. Biochem. Biophys. Res. Commun. 315(4), 1134–1139 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Zakhartchouk AN, Viswanathan S, Moshynskyy I, Petric M, Babiuk LA. Optimization of a DNA vaccine against SARS. DNA Cell. Biol. 26(10), 721–726 (2007). [DOI] [PubMed] [Google Scholar]
  • 93.Callendret B, Lorin V, Charneau P et al. Heterologous viral RNA export elements improve expression of severe acute respiratory syndrome (SARS) coronavirus spike protein and protective efficacy of DNA vaccines against SARS. Virology 363(2), 288–302 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Wang Z, Yuan Z, Matsumoto M, Hengge UR, Chang YF. Immune responses with DNA vaccines encoded different gene fragments of severe acute respiratory syndrome coronavirus in BALB/c mice. Biochem. Biophys. Res. Commun. 327(1), 130–135 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Wang X, Xu W, Tong D et al. A chimeric multi-epitope DNA vaccine elicited specific antibody response against severe acute respiratory syndrome-associated coronavirus which attenuated the virulence of SARS-CoV in vitro Immunol. Lett. 119(1–2), 71–77 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Zhao P, Cao J, Zhao LJ et al. Immune responses against SARS-coronavirus nucleocapsid protein induced by DNA vaccine. Virology 331(1), 128–135 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Gupta V, Tabiin TM, Sun K et al. SARS coronavirus nucleocapsid immunodominant T-cell epitope cluster is common to both exogenous recombinant and endogenous DNA-encoded immunogens. Virology 347(1), 127–139 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Cheung YK, Cheng SC, Sin FW, Chan KT, Xie Y. Induction of T-cell response by a DNA vaccine encoding a novel HLA-A*0201 severe acute respiratory syndrome coronavirus epitope. Vaccine 25(32), 6070–6077 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Okada M, Okuno Y, Hashimoto S et al. Development of vaccines and passive immunotherapy against SARS corona virus using SCID-PBL/hu mouse models. Vaccine 25(16), 3038–3040 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Zakhartchouk AN, Liu Q, Petric M, Babiuk LA. Augmentation of immune responses to SARS coronavirus by a combination of DNA and whole killed virus vaccines. Vaccine 23(35), 4385–4391 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Ma C, Yao K, Zhou F, Zhu M. Comparative immunization in BALB/c mice with recombinant replication-defective adenovirus vector and DNA plasmid expressing a SARS-CoV nucleocapsid protein gene. Cell Mol. Immunol. 3(6), 459–465 (2006). [PubMed] [Google Scholar]
  • 102.DeDiego ML, Alvarez E, Almazan F et al. A severe acute respiratory syndrome coronavirus that lacks the E gene is attenuated in vitro and in vivo J. Virol. 81(4), 1701–1713 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Dediego ML, Pewe L, Alvarez E et al. Pathogenicity of severe acute respiratory coronavirus deletion mutants in hACE-2 transgenic mice. Virology 376(2), 379–389 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Lamirande EW, DeDiego ML, Roberts A et al. A live attenuated severe acute respiratory syndrome coronavirus is immunogenic and efficacious in golden Syrian hamsters. J. Virol. 82(15), 7721–7724 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Zust R, Cervantes-Barragan L, Kuri T et al. Coronavirus non-structural protein 1 is a major pathogenicity factor: implications for the rational design of coronavirus vaccines. PLoS Pathog. 3(8), e109 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Yount B, Roberts RS, Sims AC et al. Severe acute respiratory syndrome coronavirus group-specific open reading frames encode nonessential functions for replication in cell cultures and mice. J. Virol. 79(23), 14909–14922 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.de Haan CA, Masters PS, Shen X, Weiss S, Rottier PJ. The group-specific murine coronavirus genes are not essential, but their deletion, by reverse genetics, is attenuating in the natural host. Virology 296(1), 177–189 (2002). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Enjuanes L, Dediego ML, Alvarez E et al. Vaccines to prevent severe acute respiratory syndrome coronavirus-induced disease. Virus Res. 133(1), 45–62 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]; •• Excellent comprehensive review also covering serotypes and attenuated SARS vaccines.
  • 109.Kuiken T, Fouchier RA, Schutten M et al. Newly discovered coronavirus as the primary cause of severe acute respiratory syndrome. Lancet 362(9380), 263–270 (2003). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.McAuliffe J, Vogel L, Roberts A et al. Replication of SARS coronavirus administered into the respiratory tract of African Green, rhesus and cynomolgus monkeys. Virology 330(1), 8–15 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Subbarao K, McAuliffe J, Vogel L et al. Prior infection and passive transfer of neutralizing antibody prevent replication of severe acute respiratory syndrome coronavirus in the respiratory tract of mice. J. Virol. 78(7), 3572–3577 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Glass WG, Subbarao K, Murphy B, Murphy PM. Mechanisms of host defense following severe acute respiratory syndrome-coronavirus (SARS-CoV) pulmonary infection of mice. J. Immunol. 173(6), 4030–4039 (2004). [DOI] [PubMed] [Google Scholar]
  • 113.Roberts A, Vogel L, Guarner J et al. Severe acute respiratory syndrome coronavirus infection of golden Syrian hamsters. J. Virol. 79(1), 503–511 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Xiao Y, Meng Q, Yin X et al. Pathological changes in masked palm civets experimentally infected by severe acute respiratory syndrome (SARS) coronavirus. J. Comp. Pathol. 138(4), 171–179 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Roberts A, Lamirande EW, Vogel L et al. Animal models and vaccines for SARS-CoV infection. Virus Res. 133(1), 20–32 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]; •• Excellent review focusing on different animal models up to early 2008.
  • 116.Rockx B, Sheahan T, Donaldson E et al. Synthetic reconstruction of zoonotic and early human severe acute respiratory syndrome coronavirus isolates that produce fatal disease in aged mice. J. Virol. 81(14), 7410–7423 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.McCray PB Jr, Pewe L, Wohlford-Lenane C et al. Lethal infection of K18-hACE2 mice infected with severe acute respiratory syndrome coronavirus. J. Virol. 81(2), 813–821 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Tseng CT, Huang C, Newman P et al. Severe acute respiratory syndrome coronavirus infection of mice transgenic for the human angiotensin-converting enzyme 2 virus receptor. J. Virol. 81(3), 1162–1173 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Yang XH, Deng W, Tong Z et al. Mice transgenic for human angiotensin-converting enzyme 2 provide a model for SARS coronavirus infection. Comp. Med. 57(5), 450–459 (2007). [PubMed] [Google Scholar]
  • 120.ter Meulen J, Bakker AB, van den Brink EN et al. Human monoclonal antibody as prophylaxis for SARS coronavirus infection in ferrets. Lancet 363(9427), 2139–2141 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Weingartl H, Czub M, Czub S et al. Immunization with modified vaccinia virus Ankara-based recombinant vaccine against severe acute respiratory syndrome is associated with enhanced hepatitis in ferrets. J. Virol. 78(22), 12672–12676 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.To KF, Tong JH, Chan PK et al. Tissue and cellular tropism of the coronavirus associated with severe acute respiratory syndrome: an in-situ hybridization study of fatal cases. J. Pathol. 202(2), 157–163 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Skowronski DM, Astell C, Brunham RC et al. Severe acute respiratory syndrome (SARS): a year in review. Annu. Rev. Med. 56, 357–381 (2005). [DOI] [PubMed] [Google Scholar]
  • 124.Lawler JV, Endy TP, Hensley LE et al. Cynomolgus macaque as an animal model for severe acute respiratory syndrome. PLoS Med. 3(5), e149 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Weiss RC, Scott FW. Antibody-mediated enhancement of disease in feline infectious peritonitis: comparisons with dengue hemorrhagic fever. Comp. Immunol. Microbiol. Infect. Dis. 4(2), 175–189 (1981). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Kam YW, Kien F, Roberts A et al. Antibodies against trimeric S glycoprotein protect hamsters against SARS-CoV challenge despite their capacity to mediate FcγRII-dependent entry into B cells in vitro Vaccine 25(4), 729–740 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Deming D, Sheahan T, Heise M et al. Vaccine efficacy in senescent mice challenged with recombinant SARS-CoV bearing epidemic and zoonotic spike variants. PLoS Med. 3(12), 525 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Tan YJ, Goh PY, Fielding BC et al. Profiles of antibody responses against severe acute respiratory syndrome coronavirus recombinant proteins and their potential use as diagnostic markers. Clin. Diagn. Lab. Immunol. 11(2), 362–371 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Woo PC, Lau SK, Wong BH et al. Detection of specific antibodies to severe acute respiratory syndrome (SARS) coronavirus nucleocapsid protein for serodiagnosis of SARS coronavirus pneumonia. J. Clin. Microbiol. 42(5), 2306–2309 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Liu X, Shi Y, Li P et al. Profile of antibodies to the nucleocapsid protein of the severe acute respiratory syndrome (SARS)-associated coronavirus in probable SARS patients. Clin. Diagn. Lab. Immunol. 11(1), 227–228 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Greenough TC, Carville A, Coderre J et al. Pneumonitis and multi-organ system disease in common marmosets (Callithrix jacchus) infected with the severe acute respiratory syndrome-associated coronavirus. Am J. Pathol. 167(2), 455–463 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Sui J, Li W, Murakami A et al. Potent neutralization of severe acute respiratory syndrome (SARS) coronavirus by a human mAb to S1 protein that blocks receptor association. Proc. Natl Acad. Sci. USA 101(8), 2536–2541 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Traggiai E, Becker S, Subbarao K et al. An efficient method to make human monoclonal antibodies from memory B cells: potent neutralization of SARS coronavirus. Nat. Med. 10(8), 871–875 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Roper RL, Payne LG, Moss B. Extracellular vaccinia virus envelope glycoprotein encoded by the A33R gene. J. Virol. 70(6), 3753–3762 (1996). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Fogg C, Lustig S, Whitbeck JC et al. Protective immunity to vaccinia virus induced by vaccination with multiple recombinant outer membrane proteins of intracellular and extracellular virions. J. Virol. 78(19), 10230–10237 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Golden JW, Hooper JW. Heterogeneity in the A33 protein impacts the cross-protective efficacy of a candidate smallpox DNA vaccine. Virology 377(1), 19–29 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Hu MC, Jones T, Kenney RT et al. Intranasal protollin-formulated recombinant SARS S-protein elicits respiratory and serum neutralizing antibodies and protection in mice. Vaccine 25(34), 6334–6340 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]; • Shows the importance of mucosal immunity.
  • 138.Yang L, Peng H, Zhu Z et al. Persistent memory CD4+ and CD8+ T-cell responses in recovered severe acute respiratory syndrome (SARS) patients to SARS coronavirus M antigen. J. Gen.Virol. 88(Pt 10), 2740–2748 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Qiu M, Shi Y, Guo Z et al. Antibody responses to individual proteins of SARS coronavirus and their neutralization activities. Microbes Infect. 7(5–6), 882–889 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]

Websites


Articles from Expert Review of Vaccines are provided here courtesy of Taylor & Francis

RESOURCES