Immune Netw. 2020 Aug;20(4):e28. English.
Published online Jul 22, 2020.
Copyright © 2020. The Korean Association of Immunologists
Review

Newly Emerging Human Coronaviruses: Animal Models and Vaccine Research for SARS, MERS, and COVID-19

Pureum Lee,1,2 and Doo-Jin Kim1,3
    • 1Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea.
    • 2University of Science and Technology (UST), Daejeon 34113, Korea.
    • 3Department of Biochemistry, Chungnam National University, Daejeon 34134, Korea.
Received May 28, 2020; Revised June 26, 2020; Accepted June 29, 2020.

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (https://creativecommons.org/licenses/by-nc/4.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

The recent emergence of the novel coronavirus (CoV) or severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) poses a global threat to human health and economy. As of June 26, 2020, over 9.4 million cases of infection, including 482,730 deaths, had been confirmed across 216 countries. To combat a devastating virus pandemic, numerous studies on vaccine development are urgently being accelerated. In this review article, we take a brief look at the characteristics of SARS-CoV-2 in comparison to SARS and Middle East respiratory syndrome (MERS)-CoVs and discuss recent approaches to coronavirus disease-2019 (COVID-19) vaccine development.

Keywords
Coronavirus; Vaccines; SARS; MERS; COVID-19; SARS-CoV-2

INTRODUCTION

Coronaviruses are positive-sense RNA viruses belonging to the family Coronaviridae. They are divided into 4 genera: alpha (α), beta (β), gamma (γ), and delta (δ) coronaviruses, based on their phylogenetic relationships and genomic structures. α- and β-coronaviruses infect only mammals whereas the γ- and δ-coronaviruses mainly infect birds (1). Typically, coronaviruses are known to cause only mild illnesses, like the common cold in humans, but the outbreak of severe acute respiratory syndrome (SARS) in 2002 (2) demonstrated that coronaviruses originating from other animal species may cross the species barrier and could become life-threatening pathogens in humans. A decade after the SARS outbreak, Middle East respiratory syndrome coronavirus (MERS-CoV), another pathogenic coronavirus, emerged in Saudi Arabia (3). Most recently, another β-coronavirus—severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2)—has been newly identified from a cluster of patients with severe pneumonia (4, 5). To date, 2 α-coronaviruses (human coronavirus [HCoV]-229E and HCoV-NL63) and 5 β-coronaviruses (HCoV-OC43, HKU1, SARS-CoV, MERS-CoV, and SARS-CoV-2) have been identified that infect humans (1).

GENOME, VIRION, AND LIFE CYCLE

Coronaviruses are enveloped positive single-stranded RNA viruses. They have 26–32 kb genomic RNA, which is the largest among the genomes of RNA viruses (6, 7). The genomic structures of all coronaviruses are similarly arranged: replicase genes encoded within two-thirds of the 5′ end and genes encoding structural proteins in the other one-third of the 3′ end. Replicase genes, which occupy 20–22 kb of the entire coronavirus genome, is composed of open reading frame (ORF) 1a and ORF1b. ORF1a includes papain-like proteases (PLpro or PLP) and picornavirus 3C-like protease (3Clpro) genes, and ORF1b features viral RNA-dependent RNA polymerase (RdRp), helicase, and exoribonuclease (ExoN) genes (8). Upon viral infection, translation first begins from the 5′ end of ORF1 to create a huge complex of ORF1a polyprotein. At a lesser frequency, the −1 ribosomal frameshift occurs at the pseudoknot immediately before the termination codon of ORF1a, and translation resumes to constitute the ORF1ab polyprotein. Subsequently, the polyprotein is cleaved by the viral proteases into 16 mature nonstructural proteins (nsp1–16). As the frequency of frameshift is roughly 25%–30%, proteins encoded by ORF1b are produced in relatively smaller amounts compared to those by ORF1a. Nevertheless, ORF1b is the most conserved gene within the coronavirus genome, suggesting that ORF1b plays a crucial role in viral replication.

The latter one-third of the 3′ end of genomic RNA encodes the four major coronavirus structural proteins, including spike (S), envelope (E), membrane (M), and nucleocapsid (N) proteins, along with accessory proteins that are virus species-specific with functions that are not fully understood (Fig. 1) (6, 7). S, E, and M proteins are displayed on the coronavirus virion surface. Certain coronaviruses have hemagglutinin esterase (HE) on their surface, but SARS- and MERS-CoVs do not. The S protein is presented on the viral surface as trimers and protrude, forming corona-like structure on the envelope. S consists of two functional subunits—S1 is responsible for the binding to host receptors and S2 for the fusion of viral and cellular membranes. The E protein, formerly called sM, plays a major role in viral assembly and release by interacting with M protein via its cytoplasmic tails. In addition, E is closely associated with viral pathogenesis by interfering with the formation and maintenance of tight junctions in the lung mucosal epithelium, leading to acute alveolar damage (9). M protein is the most abundant protein in the virus envelope. It consists of a short N-terminal glycosylated domain and three transmembrane domains followed by a long C-terminal tail. Moreover, M is localized within the intracellular membrane in the endoplasmic reticulum-Golgi intermediate compartment (ERGIC), where virion assembly and budding out occur, thereby controlling viral assembly by interacting with S and N proteins. N protein is a basic RNA-binding protein composed of three highly conserved domains: N-terminal, C-terminal, and RNA-binding domains. N binds to viral genomic RNA to form a helical capsid structure called the ribonucleoprotein (RNP) complex. The interaction of N, E, and M proteins drive the incorporation of the RNP into the assembling virions. In addition to RNP formation, N proteins play a critical role in enhancing the replication and synthesis of genomic RNA by interacting with nsp3.

Figure 1
Genome structure of human coronaviruses (SARS-CoV, MERS-CoV, and SARS-CoV-2) and an overview of coronavirus vaccine platforms.

NEWLY EMERGING HUMAN CORONAVIRUSES

SARS-CoV

SARS was first reported in Guangdong province, China in November 2002 and promptly spread worldwide, resulting in 8,096 cases, including 774 deaths in 29 countries (10). The clinical symptoms of SARS are similar to those of other respiratory infections, like influenza. During the initial phase of infection, patients with SARS exhibited fever, cough, sore throat, and other mild symptoms, and some subsequently progressed to severe pneumonia (11). High levels of pro-inflammatory cytokines and chemokines were detected in the sera of SARS patients with severe disease (12). They also displayed low levels of anti-inflammatory cytokines, such as IL-10, compared to those of patients with mild symptoms.

The receptor binding domain (RBD) of S protein binds to host angiotensin-converting enzyme 2 (ACE2) for their entry into cells (13). ACE2 is expressed on a wide variety of body tissues and cells, including small intestine epithelium, arterial and venous endothelium, arterial smooth muscle, respiratory tract epithelium, alveolar monocytes, and alveolar macrophages (14). Owing to the widespread expression of ACE2 throughout the body, SARS-CoV infects various tissues and causes lesions. Specifically, SARS-CoV primarily infects airway epithelial cells, resulting in the induction of large amounts of chemokines, such as CCL2, CCL3, CCL5, and CXCL10. The virus also infects hematopoietic cells, like dendritic cells (DCs) and macrophages. In such cases, DCs and macrophages exhibit the upregulated expression of pro-inflammatory cytokines and chemokines, such as TNF-α, IL-6, and CXCL10, but downregulated or delayed type I IFN (IFN-α/β) response (15). Consequently, the excess concentrations of pro-inflammatory molecules recruit various inflammatory immune cells into the lungs, leading to consolidation, hemorrhages, edema, and diffuse alveolar damage (DAD) (16).

MERS-CoV

Ten years after the SARS outbreak, MERS emerged in the Kingdom of Saudi Arabia in 2012. To date, 2,519 MERS cases with 866 deaths have been confirmed in 27 countries across the Middle East, Asia, and Europe (17). In 2015, 186 cases of infection, including 38 deaths, were reported in South Korea (18), which was the most unique and largest MERS outbreak outside the Arabian Peninsula.

Major clinical symptoms of MERS are fever, non-productive cough, dyspnea, myalgia, and sore throat (19). Unlike patients with influenza and SARS, certain patients with MERS distinctively presented gastrointestinal symptoms, including diarrhea and vomiting (20). The majority of MERS patients progressed to severe pneumonia; particularly, immunocompromised individuals and patients with comorbidities exhibited high incidences of acute respiratory distress syndrome (ARDS) and renal dysfunction (21). Similar to SARS, in MERS patients, a dysregulated immune response is thought to be the cause of pathological changes, such as extensive infiltration of immune cells into the lungs (22).

In contrast to the case of SARS, MERS-CoV particles or MERS-CoV-specific Abs were detected in a large number of dromedary camels in the Middle East and North Africa (23). This strongly indicates that the virus, which is thought to originate from bats (24, 25), has been circulating for more than several decades in dromedary camels in those areas, which may be a reason for continuing zoonotic transmission of MERS-CoV. Human-to-human transmission of MERS-CoV mainly occurs through the nosocomial route, especially between hospitalized patients (26), probably because virus shedding takes place efficiently after the onset of disease symptoms.

MERS-CoV infects host cells by interacting with dipeptidyl peptidase 4 (DPP4) or CD26 (27). DPP4 is expressed in the respiratory tract epithelium, kidney, small intestine, liver, prostate, and also on activated leukocytes (28). The virus primarily infects airway epithelial cells, resulting in delayed IFN responses and upregulated pro-inflammatory cytokines, such as IL-6, IL-1β, and IL-8 (29). MERS-CoV-infected macrophages and DCs also produce high levels of pro-inflammatory cytokines and chemokines, such as CCL2, CCL3, CCL5, and IL-2 (30, 31), and the concentration of these soluble factors closely correlates with disease severity (32). Increased numbers of neutrophils and monocytes were observed in the lungs of these types of patients (22), indicating that these cells are responsible for lung immunopathology.

SARS-CoV-2

In December 2019, severe cases of pneumonia with unknown etiology were reported in Wuhan, China (4). As the causative agent of the disease was identified as a coronavirus, the disease and virus were named as coronavirus disease-2019 (COVID-19) and SARS-CoV-2, respectively (5, 33). As the virus drastically disseminates on a global scale, the World Health Organization (WHO) declared a global pandemic on 11th March 2020. To date, over 9.4 million cases of SARS-CoV-2 infection, including 482,730 deaths, have been reported, and the infection curve is still rising at a steep angle (34).

SARS-CoV-2 was determined to be a lineage B β-coronavirus, sharing 79% of genome sequence identity with SARS-CoV (35). It is thought to originate from bats, like other human β-coronaviruses, and to be transmitted to humans through probable, but unproven, intermediate hosts, such as snakes or pangolins (36, 37). However, despite ongoing studies, the hosts have not been specified to date, suggesting transmission took place incidentally, like in the case of SARS-CoV.

Similar to SARS-CoV, the S protein of SARS-CoV-2 binds to ACE2 as its receptor (38, 39), and subsequently trigger fusion of viral and cellular membranes, thereafter entering host cells. As such, amino acid sequence and distribution of ACE2 is the major determinant of host and cell tropism (40). In the human body, ACE2 is expressed at a high level in the small intestine, testis, and kidneys, and at a relatively low level in the lungs and heart in healthy individuals. However, in the case of smokers and patients with heart conditions, ACE2 levels are elevated compared to that in the healthy (41, 42), partially accounting for the high pathogenicity in those populations.

Most patients with COVID-19 exhibit mild to moderate clinical symptoms, such as fever and dry cough, and sometimes muscle and/or joint pain (43). However, the elderly or individuals with underlying diseases, such as asthma, heart conditions, and diabetes, are more vulnerable to SARS-CoV-2, leading to severe pneumonia or ARDS, the main cause of COVID-19-related death (43). Other less common symptoms have also been reported, such as gastrointestinal symptoms (44) and loss of taste or smell (45).

Upon infection, SARS-CoV-2 activates the innate immune system and induces pro-inflammatory cytokines and chemokines in the lungs along with recruitment of monocytes and T cells (43). In most healthy individuals, this local immune response contributes to the clearance of viral infection, but in patients with preconditions, dysregulated immune response results in massive infiltration of immune cells, respiratory failure, or systemic inflammation. In particular, unlike other respiratory viruses causing mild symptoms, SARS-CoV-2 is unique, as it drives low type I and III interferon levels and a moderate IFN-stimulated gene (ISG) response (46, 47). Consistent with these observations, SARS-CoV-2 ORF3b, a 22-amino acid-long nonstructural protein, exhibited a significantly stronger antagonistic activity against type I IFN induction than that displayed by SARS-CoV ORF3b ortholog (48). Contrary to this weak antiviral response, inflammatory response represented by the production of proinflammatory cytokines and chemokines, such as IL-6, CCL8, and CXCL9, was markedly elevated under SARS-CoV-2 infection both in vitro and in vivo (46). Patients with severe COVID-19 also exhibited abundant distribution of proinflammatory monocyte-derived macrophages in the bronchoalveolar lavage fluid (BALF) (49) and high serum levels of proinflammatory cytokines and chemokines, such as IL-1β, IL-2, IL-6, IL-8, IL-17, IFN-γ-induced protein (IP)-10, MCP-1, TNF-α, G-CSF, and GM-CSF (43, 50, 51). This imbalanced—low antiviral but high inflammatory—host response is believed to be a major factor affecting disease outcome.

In addition to uncontrolled innate immune responses, impaired adaptive immune responses can affect disease severity. The number of lymphocytes including T, B, or NK cells was significantly reduced in patients with severe COVID-19 requiring intensive care unit (ICU) care (50, 51, 52). Decreased T cell number has also been observed in patients with SARS (53), and MERS-CoV has been reported to infect human T lymphocytes and activate apoptotic pathways in the infected cells (54). Further investigation is required to elucidate the reason underlying the decrease in the number of lymphocytes in patients with severe COVID-19. It is intriguing that T cells from patients with COVID-19 highly express PD-1 and T-cell immunoglobulin mucin-3 (TIM-3), which are exhaustion markers (52). Additionally, the frequency of T and NK cells producing CD107a, IFN-γ, IL-2, and granzyme B was significantly reduced in patients with severe infection, compared to those from healthy controls (55). However, despite the increased exhaustion marker levels and decreased cellular function, it should be carefully defined whether the T cells are really “exhausted” by continuous antigenic stimulation through Ag-specific TCRs.

ANIMAL MODELS

To elucidate the mechanisms of viral pathogenesis and develop optimal prophylactic and therapeutic strategies for newly emerging human coronaviruses, several animal models have been developed and evaluated (Table 1).

Table 1
Epidemiology, biological characteristics, and vaccine studies of SARS-CoV, MERS-CoV, and SARS-CoV-2

SARS-CoV and SARS-CoV-2

As mentioned earlier, SARS-CoV and SARS-CoV-2 enter target cells by binding to ACE2 as their receptor (38, 39, 40). In mice, SARS-CoV is able to interact with murine ACE2 and replicate in mouse tissues, including the lungs and small intestine (57), but disease symptoms are limited to mild respiratory disease and minimal bodyweight loss that is less than 5%. Aged mice present a relatively larger number of severe symptoms than young mice (59, 60, 61). To improve the availability of a murine SARS model, mouse-adapted SARS-CoV strains (149, 150) and transgenic mice expressing human ACE2 (hACE2) were developed (62, 63). Myeloid differentiation primary response 88 (MyD88) as well as STAT1 knock-out mice presented severe respiratory diseases, like pneumonitis and bronchiolitis, along with reduced survival compared to wild-type mice (58, 64, 65), suggesting that innate immunity involved with these molecules plays an important role in the clearance of SARS-CoV.

In addition to mice, various other animal models are available for SARS-CoV studies. Golden Syrian and Chinese hamsters (66) and ferrets (67, 68) are susceptible to SARS-CoV infection and display moderate to severe respiratory symptoms. SARS-CoV infects non-human primates (NHPs), including rhesus macaques, cynomolgus macaques, African green monkeys, common marmosets, squirrel monkeys, and mustached tamarins because these NHP species express a form of ACE2 closely related to that of humans (69, 70, 71). More importantly, the virus successfully replicates in these NHPs and causes severe symptoms, like fever, pneumonitis, diarrhea, and hepatitis (72).

As SARS-CoV-2 was revealed to also utilize ACE2 for viral entry (38, 39, 40), SARS animal models were promptly tested in SARS-CoV-2 studies. hACE2 transgenic mice exhibited moderate interstitial pneumonia (84), and Golden Syrian hamsters presented clinical symptoms and histopathological findings closely resembling what is observed in humans (85). In ferrets, SARS-CoV-2 viral RNA was detected in the nasal turbinate, soft palate, and tonsil, but the viral infection induced only mild clinical symptoms (86). In NHP models, the virus was excreted from the respiratory tract and detected in multiple organs in virus-infected cynomolgus macaques, but they did not develop any clinical signs (88). Aged rhesus macaques exhibited more severe and diffuse pneumonia along with serious inflammatory responses versus young monkeys (89), suggesting the age is a decisive factor in both NHP models and humans. SARS-CoV-2 susceptibility was also investigated in domesticated animals—SARS-CoV-2 can efficiently replicate in cats but does so poorly in dogs, pigs, chickens, and ducks (87). Most recently, a new mouse model using recombinant adenovirus 5 expressing hACE2 (Ad5-hACE2) was reported (90), which is similar to the model developed for a MERS study by the same group (73). Upon intranasal transduction with Ad5-hACE2, mice transiently expressed hACE2 in their respiratory tract and exhibited significant viral replication and lung inflammation upon subsequent SARS-CoV-2 infection.

MERS-CoV

MERS-CoV employs host cellular DPP4 as its receptor for entry (27). Whereas humans and NHPs are susceptible to MERS-CoV infection, hamsters, ferrets, and mice are not because of differences in major amino acid sequences of DPP4 (151, 152, 153). The first model for MERS-CoV study was based on mice transduced with recombinant adenovirus expressing human DPP4 (Ad5-hDPP4) (73). The Ad5-hDPP4-transduced mice developed clinical symptoms including pneumonitis and lung edema upon MERS-CoV infection. Global or tissue-specific hDPP4 transgenic mice were successfully infected with MERS-CoV and displayed respiratory symptoms and weight loss (74, 75). hDPP4 knock-in mice have also been developed within which murine DPP4 is replaced by hDPP4 using CRISPR/Cas9 (77, 78).

In addition to these mouse models, dromedary camels, an intermediate animal in MERS-CoV transmission, and alpacas were tested for a MERS study. Although they were susceptible to the virus, they were asymptomatic or exhibited, if any, only mild respiratory symptoms (79, 80). In NHPs, MERS-CoV effectively infected the host cells and replicated within their lungs (81, 82, 83), but disease severity was higher in the common marmosets than that in rhesus macaques. Furthermore, symptoms observed in severe patients, such as lung consolidation and liver or kidney failure, were reproduced only in common marmosets (76), indicating that they are a more reliable animal model for MERS study.

VACCINE RESEARCH

Although there are still no approved vaccines for SARS and MERS, studies on the two previous coronaviruses provided important information about a strategy and considerations for COVID-19 vaccine development. First, as S protein, especially RBD, was known to be responsible for binding to host receptors, it has been extensively evaluated as a primary target Ag for vaccine development. Second, the majority of vaccine studies have utilized various recombinant vaccine platforms, including recombinant proteins, nucleic acids, and virus-vectored vaccines, rather than conventional live-attenuated or inactivated virus platforms (Fig. 1 and Table 1).

Live-attenuated virus vaccines

As the live-attenuated virus vaccine is composed of almost all proteins of the virus, immune responses induced by attenuated virus vaccination are most similar to those by real viral infection (154). In a conventional method, live-attenuated virus has been made by serial culture of the virus, which leads to a spontaneous deletion of or mutation within a pathogenic gene. However, in recent studies, recombination technology-based modification of the target gene has been more widely studied in coronavirus vaccines. Among the coronavirus proteins, E and nsp16 have been thought of as the most potential targets because of their potential association with the virulence in vivo (9). Immunization of engineered mutant SARS-CoV lacking the E protein (rSARS-CoV-ΔE) provided protective immunity in hACE2 transgenic mice and golden Syrian hamsters against viral challenge (91, 92). E gene-deleted MERS-CoV (rMERS-CoV-ΔE) was able to replicate only by providing E protein in trans, but unable to propagate in vivo (94). Recombinant MERS-CoV lacking the accessory genes 3, 4a, and 5, was also replication-competent in vitro but propagation-defective in vivo, indicating that recombinant MERS-CoV could be a safe and promising vaccine candidate (94). SARS- and MERS-CoVs harboring a D130A mutation in each nsp16 gene (SARS-CoV [D130A] and MERS-CoV [D130A]) induced a neutralizing Ab response and protected against lethal virus challenge in young animals without any pathologic symptoms (93, 95). Currently, 2 live-attenuated COVID-19 vaccines based on codon-deoptimization are under preclinical development (148).

Inactivated whole-virus vaccines

Inactivated whole-virus vaccines are prepared by inactivation of the cultured virus by heat, ultraviolet (UV), or chemicals, such as formalin. When compared to a live-attenuated virus vaccine, they can be produced relatively quickly and easily upon the emergence of a new or variant virus. In coronavirus vaccine studies, inactivated virus vaccine induced high-titer neutralizing Abs and cell-mediated immune responses. Immunization with SARS-CoV inactivated with UV light (96, 97), formaldehyde (97), and β-propiolactone (98) induced potent neutralizing Abs and CD4+ and CD8+ T cell responses in mice and rabbits. In addition to SARS-CoV, MERS-CoV treated with γ-irradiation or formaldehyde also offered protection against MERS-CoV infection (99, 100). Some studies have shown that inactivated SARS- and MERS-CoV vaccines were efficacious in protection from challenge in mice and NHPs (155, 156, 157, 158). The efficacy of an inactivated virus COVID-19 vaccine has been tested in mice, rats, and rhesus macaques (101). Currently, at least nine research groups are developing COVID-19 vaccines using an inactivated virus platform. Among them, four institutions in China (Sinovac, Wuhan Institute of Biological Products, Beijing Institute of Biological Products, and Chinese Academy of Medical Sciences) have started clinical studies (148).

Recombinant protein vaccines

Recombinant protein vaccines have long been studied and assessed in terms of efficacy and safety. Many researchers have also evaluated human coronavirus vaccines, mainly focusing on S and RBD proteins of SARS- and MERS-CoVs. Immunization with the full-length, extracellular domain of S proteins or Fc-fused RBD proteins of SARS-CoV induced potent neutralizing Abs in mice and/or rabbits (102, 103, 104, 105). Trimeric S or RBD proteins also induced humoral and cellular immune responses and provided protection against SARS-CoV infection in hamsters (106). Similar to the results from SARS vaccine studies, recombinant S, RBD, Fc-fused RBD, and trimeric RBD proteins of MERS-CoV also elicited neutralizing Abs in various animal models, including mice and monkeys, and exhibited protective effects upon viral challenge (107, 108, 109, 110, 111, 112, 113, 114, 116). Immunization of the N-terminal domain (NTD) of the MERS-CoV S protein also led to protection against MERS-CoV challenge in a transient hDPP4-expressing mice model (Ad5-hDPP4 mice) (115). In most of these experiments, recombinant protein Ags were used with an adjuvant, such as alum and MF59, to increase Ab or cell-mediated immune responses. Intriguingly, MERS-CoV S proteins alone self-assemble into nanoparticles of a size of approximately 25 nm. With this, the S nanoparticles effectively induce neutralizing Abs and Th1-type cellular immune responses in mice and NHPs (116, 123), providing protection against MERS-CoV infection in mice (117). In addition, heterologous prime-boost vaccination with adenoviral vector-expressing S and S nanoparticles led to balanced Th1/Th2 responses and safeguarded mice from MERS-CoV challenge (118).

In terms of SARS-CoV-2, Novavax (Gaithersburg, MD, USA) is developing a spike nanoparticle vaccine at phase 1/2 clinical stage, and another 50 institutions are working on recombinant protein vaccines focusing on S or RBD proteins at a pre-clinical stage (148). One of the most outstanding advances in the recent study of recombinant protein vaccines is the design of a prefusion form of viral surface Ags based on structural biology. Target Ags expressed as a stable prefusion form induced more potent and high-affinity neutralizing Ab responses than wild-type proteins in various infectious disease models (159, 160, 161, 162). Several institutions, including Queensland University (Brisbane, Australia) and Clover Biopharmaceuticals (Chengdu, China), are applying this technology to COVID-19 vaccine development.

Meanwhile, for the optimal efficacy and dose sparing of recombinant protein Ags, it is essential to develop a vaccine in combination with an appropriate adjuvant. In previous SARS and MERS vaccine studies, the effects of diverse adjuvants such as alum (106, 108, 109, 115, 116, 118, 123), MF59® (109, 110, 112), Matrix™ M (116, 117), Montanide ISA™ 51 (109, 111, 113), and monophosphoryl-lipid A plus trehalose dicorynomycolate (MPL® + TDM) (102, 104, 114), have been widely tested. During the development of a recombinant protein-based COVID-19 vaccine, the choice of an adjuvant would be a considerable factor affecting the quality of the immune response, the efficacy and safety of the vaccine, and the economic feasibility of the developer.

Virus-like particle (VLP) vaccines

VLPs are nano-sized particles composed of viral proteins with self-assembly properties. They mimic the morphology of a real virus particle but do not replicate owing to the lack of genomic material. As VLPs maintain the ideal conformation of native Ags, they can elicit an appropriate and strong immune response (163).

VLPs of SARS- and MERS-CoVs were produced by coexpressing the S, E, and M proteins from insect or mammalian cells (119, 123), and these VLPs induced potent neutralizing Abs and Th1-biased cellular immune responses in mice and NHPs (120, 121, 123). Interestingly, SARS-CoV S and influenza virus matrix 1 (M1) coexpression efficiently formed chimeric VLP and induced protective immunity against SARS-CoV (122). At present, ten COVID-19 vaccine candidates are under pre-clinical investigation on the basis of VLP technology (148).

DNA and RNA vaccines

Since it was first reported that immunization of naked plasmid DNA encoding foreign protein induces an Ag-specific immune response in mammals in the early 1990s (164), DNA vaccines have been widely tested in various pathogen models. Although DNA vaccines have not been approved for humans to date, their immunogenicity and therapeutic effects have long been tested across various clinical trials for infectious diseases as well as human papillomavirus-mediated cervical intraepithelial neoplasia (165, 166). DNA vaccines expressing full-length or truncated forms of SARS-CoV S protein induced humoral and cellular immunity, supplying protection against SARS-CoV infection in murine models (98, 124, 125). Further, heterologous prime-boost immunization with DNA vaccines and inactivated SARS-CoV induced strong CD4+ T cell and Ab responses (126). Meanwhile, DNA vaccines encoding SARS-CoV N proteins induced Abs and T cell responses in mice, but their protective efficacy has not been fully investigated (127, 128). In MERS vaccine studies, immunization of full-length or S1 subunit-expressing plasmids also induced neutralizing Abs and T cell responses in mice, camels, and NHPs (114, 129, 130), while also alleviating clinical symptoms upon MERS-CoV infection in a rhesus macaque model (129).

mRNA vaccines are the most recent vaccine technology characterized by rapid development and production along with high potency. Recently, encapsulation and delivery methods have been substantially improved—the use of carrier molecules, such as liposomes, cationic polymers, and polysaccharide particles, significantly increases delivery efficacy, allowing for rapid uptake and high expression of target Ags. Owing to the safety, potent efficacy, as well as mass and prompt producibility, mRNA vaccines are being extensively evaluated in various infectious diseases and cancers (167). Yet, no striking results have been reported in SARS and MERS vaccine studies.

Currently, 12 DNA-based and 19 RNA-based COVID-19 vaccine candidates are under investigation and development (102). Moderna (Cambridge, MA, USA), together with the Vaccine Research Center at National Institutes of Health (NIH) of the USA, has promptly begun a phase 1 clinical trial, the first clinical study in COVID-19 vaccines, using mRNA vaccine, mRNA-1273. The immunogenicity and/or protective efficacy of COVID-19 DNA vaccine candidates has been evaluated in mice and rhesus macaques (131, 132), and INOVIO Pharmaceuticals (Plymouth Meeting, PA, USA) and Genexine (Seongnam, South Korea) are undergoing phase 1 to 2a clinical trials (ClinicalTrials.gov: NCT04445389) (148).

Viral vector-based vaccines

As vesicular stomatitis virus (VSV)-based Ebola vaccine (ERVEBO®) has been approved for human use by the Food and Drug Administration (FDA) of the USA in 2019 (168), the use of viral vectors for vaccines against infectious diseases appears to be more flexible than how it was before. Viral vector-based vaccines are able to induce strong and rapid Ab and cell-mediated immune responses, and several viral vectors have been developed to date, including VSV, modified vaccinia Ankara (MVA), adenovirus (Ad), and adenovirus-associated virus (AAV) (169). In coronavirus vaccine studies, Ad and MVA are the most frequently employed. Replication-defective adenoviral vector expressing S and N proteins of SARS-CoV elicited humoral and cellular immune responses in mice (98, 170). MVA expressing SARS-CoV S protein induced neutralizing Ab responses in mice, ferrets, and NHPs (133, 134, 135), and reduced lung viral titer in SARS-CoV-challenged mice (135). Immunization with adenoviral vector encoding S protein of MERS-CoV induced systemic neutralizing Abs and T cell responses (136, 137). To avoid pre-existing immunity against human adenovirus, chimpanzee adenovirus (ChAdOx1) was utilized in recent vaccine development (138). The ChAdOx1 vector encoding MERS-CoV S induced neutralizing Abs and T cell responses in hDPP4 transgenic mice (139, 140), and reduced virus shedding and nasal discharge in dromedary camels upon MERS-CoV infection (141). MVA encoding the S protein of MERS-CoV also induced neutralizing Abs and T cell responses, protecting Ad-hDPP4-transduced mice and camels from challenge with MERS-CoV (139, 142, 143). MVA expressing N protein of MERS-CoV elicited CD8+ T cell responses in mice, but its protective efficacy was not determined (144). In addition to adenovirus and MVA, several viruses, such as Newcastle disease virus, live-attenuated measles virus (MV), rabies virus, Venezuelan equine encephalitis virus, and VSV have also been investigated in the context of MERS vaccine studies ((73, 145, 146, 147, 169).

A total of 37 vaccine institutions are developing viral vector-based COVID-19 vaccines, and among them, University of Oxford (Oxford, UK) and CanSino Biological (Tianjin, China) are performing phase 1 to 2b/3 clinical trials using a replication-deficient chimpanzee adenovirus (ChAdOx1) and adenovirus type 5 (Ad5), respectively (148).

Lessons and remaining questions in SARS and MERS studies

Based on the reports introduced above, the Ab response inhibiting the interaction between S or RBD and the corresponding receptors is sufficient for the prevention of SARS- and MERS-CoV infection. Passive transfer of human monoclonal Abs also provided considerable protection against subsequent viral challenge in mice (171, 172, 173). Taken together, these results suggest that S is a promising target Ag for coronavirus vaccines. Meanwhile, the contribution of T cell immunity for the prevention and clearance of the virus has been widely advocated. CD8+ T cells play a crucial role in viral clearance by secreting effector molecules directly to infected cells (73, 174, 175). Airway CD4+ T cells also mediate protective immunity against SARS- and MERS-CoV infection through rapid production of IFN-γ (176). However, the long-term efficacy and safety of SARS or MERS vaccines in humans has not been tested to date. Moreover, in some animal studies, vaccine-induced or monoclonal S-specific neutralizing Abs markedly enhanced the infectivity of SARS- and MERS-CoVs (177, 178), necessitating further dedicated investigation.

FACTORS TO BE CONSIDERED

As discussed previously, vaccine technology has significantly advanced over the last several decades. We also have gained useful information and materials for the study of vaccines for the novel coronavirus—how the virus enters the host cells, which Ag we should target, and what kind of animal models we can use. From the aspect of a pre-clinical study, some scientists appear to already have several successful vaccine candidates. However, in terms of a COVID-19 vaccine that is applicable to humans, several factors remain to be considered and intensively investigated.

First, safety issues must be initially evaluated. Certain vaccine formulations have induced sub-optimal Abs and inappropriate Th2-mediated immune responses, leading to Ab dependent enhancement (ADE) and/or vaccine-associated enhanced respiratory disease (VAERD) (99, 179, 180, 181, 182, 183). Additionally, each candidate should be also tested for toxicity in rats or rabbits. Although nucleic acid vaccines are regarded as safe in the aspect of nonclinical toxicology, long-term safety of an RNA vaccine in humans should be carefully investigated.

The next factor to consider during COVID-19 vaccine development is efficacy, particularly in the elderly and immunocompromised. The mortality of the disease manifests a close correlation with age: less than 1.0% under the age 50, but significantly increasing up to 1.25, 3.99, and 8.61% in the 50s, 60s, and 70s, respectively, and surpassing 13% over 80 (184). This strongly suggests that the primary target population for COVID-19 vaccine should be the elderly. In the case of conventional vaccines, high-dose or adjuvanted vaccines are recommended to enhance weak immune responses in those populations (185, 186, 187). Taking this into account, the efficacy of COVID-19 vaccines in the elderly or immunocompromised must be carefully assessed. Nevertheless, even if a COVID-19 vaccine is unsatisfactory in those populations, it might still be beneficial because of indirect protection by establishing “herd immunity”.

Other important questions to be addressed are how each immune response contributes to the protection against or clearance of the virus and for how long this effect can last following vaccination or natural infection and recovery. In most pre-clinical vaccine studies, immunization of an S Ag or a part of it efficiently induced a potent neutralizing Ab response. Transfer of a SARS-CoV-2-specific monoclonal Ab and convalescent plasma also provided significant protection against the disease in an animal model and alleviated disease severity in humans (188, 189). These reports suggest that neutralizing Abs play a key role in the protection or clearance, although partial, of the virus. However, the precise mechanism and extent of contribution of virus-specific T cells to the quality, quantity, and duration of the Ab response have not been addressed, and whether T cells per se provide sufficient protection or exert a therapeutic effect remains unknown (190). This knowledge is particularly important because it can provide critical information for designing a COVID-19 vaccine and developing a quarantine policy. Currently, the therapeutic effect of adoptively-transferred SARS-CoV-2-specific T cells is being tested in a clinical trial (ClinicalTrials.gov: NCT04351659).

CONCLUSIONS

This review has presented a brief introduction to three human coronaviruses, SARS-CoV, MERS-CoV, and SARS-CoV-2, and summarized previous and current coronavirus vaccine studies. In addition to the scientific issues discussed herein, there also remain several problems to be resolved in the effort to produce an “available” COVID-19 vaccine—the arrangement of existing infrastructure or build-up of new facilities for mass production, distribution of final goods, and vaccination of large proportions of the population, and so on. Facing a novel coronavirus pandemic, we are engaging in desperate efforts for the development of a safe and effective vaccine. Ultimately, the information in this review will be beneficial and valuable for a better understanding of human coronaviruses and COVID-19 vaccine development.

Notes

Conflict of Interest:The authors declare no potential conflicts of interest.

Author Contributions:

  • Conceptualization: Kim DJ.

  • Supervision: Kim DJ.

  • Visualization: Kim DJ.

  • Writing - original draft: Lee P.

  • Writing - review & editing: Lee P, Kim DJ.

Abbreviations

3Clpro picornavirus 3C-like protease
AAV adeno-associated virus
ACE angiotensin converting enzyme
Ad adenovirus
ADE Ab-dependent enhancement
ARDS acute respiratory distress syndrome
BALF bronchoalveolar lavage fluid
COVID-19 coronavirus disease-2019
CRISPR/Cas9 clustered regularly interspaced short palindromic repeats-associated protein-9 nuclease
DAD diffuse alveolar damage
DPP4 dipeptidyl peptidase-4
E envelope
ERGIC endoplasmic reticulum-Golgi intermediate compartment
ExoN exoribonuclease
FDA Food and Drug Administration
hACE2 human ACE2
HCoV human coronavirus
HE hemagglutinin esterase
ICU intensive care unit
IP IFN-γ-induced protein
ISG interferon-stimulated gene
M membrane
M1 matrix 1
MERS Middle East respiratory syndrome
MV measles virus
MVA modified vaccinia virus Ankara
MyD88 myeloid differentiation primary response 88
N nucleocapsid
NHP non-human primate
NIH National Institutes of Health
nsp nonstructural protein
NTD N-terminal domain
ORF open reading frame
PLpro or PLP papain-like proteases
RBD receptor-binding domain
RdRp RNA-dependent RNA polymerase
RNP ribonucleoprotein
S spike
SARS severe acute respiratory syndrome
TIM-3 T-cell immunoglobulin mucin-3
VAERD vaccine-associated enhanced respiratory disease
VLP virus-like particle
VSV vesicular stomatitis virus
WHO World Health Organization

ACKNOWLEDGEMENTS

This work was supported by grants of the Bio & Medical Technology Development Program (2016M3A9B6918675 and 2018M3A9H4077992) of the National Research Foundation of Korea (NRF) and the KRIBB Initiative program (KGM9942011), funded by the Korean government (Ministry of Science & ICT). We would like to thank Editage (www.editage.co.kr) for English language editing.

References

    1. Amanat F, Krammer F. SARS-CoV-2 vaccines: status report. Immunity 2020;52:583–589.
    1. Drosten C, Günther S, Preiser W, van der Werf S, Brodt HR, Becker S, Rabenau H, Panning M, Kolesnikova L, Fouchier RA, et al. Identification of a novel coronavirus in patients with severe acute respiratory syndrome. N Engl J Med 2003;348:1967–1976.
    1. Zaki AM, van Boheemen S, Bestebroer TM, Osterhaus AD, Fouchier RA. Isolation of a novel coronavirus from a man with pneumonia in Saudi Arabia. N Engl J Med 2012;367:1814–1820.
    1. Zhu N, Zhang D, Wang W, Li X, Yang B, Song J, Zhao X, Huang B, Shi W, Lu R, et al. A novel coronavirus from patients with pneumonia in China, 2019. N Engl J Med 2020;382:727–733.
    1. Coronaviridae Study Group of the International Committee on Taxonomy of Viruses. The species severe acute respiratory syndrome-related coronavirus: classifying 2019-nCoV and naming it SARS-CoV-2. Nat Microbiol 2020;5:536–544.
    1. Masters PS. The molecular biology of coronaviruses. Adv Virus Res 2006;66:193–292.
    1. Cui J, Li F, Shi ZL. Origin and evolution of pathogenic coronaviruses. Nat Rev Microbiol 2019;17:181–192.
    1. Hofer U. Viral evolution: fooling the coronavirus proofreading machinery. Nat Rev Microbiol 2013;11:662–663.
    1. DeDiego ML, Nieto-Torres JL, Jiménez-Guardeño JM, Regla-Nava JA, Alvarez E, Oliveros JC, Zhao J, Fett C, Perlman S, Enjuanes L. Severe acute respiratory syndrome coronavirus envelope protein regulates cell stress response and apoptosis. PLoS Pathog 2011;7:e1002315
    1. World Health Organization. Summary of probable SARS cases with onset of illness from 1 November 2002 to 1 July 2003 [Internet]. [accessed on 25 June 2020].
    1. Peiris JS, Chu CM, Cheng VC, Chan KS, Hung IF, Poon LL, Law KI, Tang BS, Hon TY, Chan CS, et al. Clinical progression and viral load in a community outbreak of coronavirus-associated SARS pneumonia: a prospective study. Lancet 2003;361:1767–1772.
    1. Wong CK, Lam CW, Wu AK, Ip WK, Lee NL, Chan IH, Lit LC, Hui DS, Chan MH, Chung SS, et al. Plasma inflammatory cytokines and chemokines in severe acute respiratory syndrome. Clin Exp Immunol 2004;136:95–103.
    1. Li W, Moore MJ, Vasilieva N, Sui J, Wong SK, Berne MA, Somasundaran M, Sullivan JL, Luzuriaga K, Greenough TC, et al. Angiotensin-converting enzyme 2 is a functional receptor for the SARS coronavirus. Nature 2003;426:450–454.
    1. Hamming I, Timens W, Bulthuis ML, Lely AT, Navis G, van Goor H. Tissue distribution of ACE2 protein, the functional receptor for SARS coronavirus. A first step in understanding SARS pathogenesis. J Pathol 2004;203:631–637.
    1. Law HK, Cheung CY, Ng HY, Sia SF, Chan YO, Luk W, Nicholls JM, Peiris JS, Lau YL. Chemokine up-regulation in SARS-coronavirus-infected, monocyte-derived human dendritic cells. Blood 2005;106:2366–2374.
    1. Nicholls JM, Poon LL, Lee KC, Ng WF, Lai ST, Leung CY, Chu CM, Hui PK, Mak KL, Lim W, et al. Lung pathology of fatal severe acute respiratory syndrome. Lancet 2003;361:1773–1778.
    1. World Health Organization. MERS situation update, January 2020 [Internet]. [accessed on 25 June 2020].
    1. Ki M. 2015 MERS outbreak in Korea: hospital-to-hospital transmission. Epidemiol Health 2015;37:e2015033
    1. Arabi YM, Arifi AA, Balkhy HH, Najm H, Aldawood AS, Ghabashi A, Hawa H, Alothman A, Khaldi A, Al Raiy B. Clinical course and outcomes of critically ill patients with Middle East respiratory syndrome coronavirus infection. Ann Intern Med 2014;160:389–397.
    1. Zhou J, Li C, Zhao G, Chu H, Wang D, Yan HH, Poon VK, Wen L, Wong BH, Zhao X, et al. Human intestinal tract serves as an alternative infection route for Middle East respiratory syndrome coronavirus. Sci Adv 2017;3:eaao4966
    1. Zumla A, Hui DS, Perlman S. Middle East respiratory syndrome. Lancet 2015;386:995–1007.
    1. Ng DL, Al Hosani F, Keating MK, Gerber SI, Jones TL, Metcalfe MG, Tong S, Tao Y, Alami NN, Haynes LM, et al. Clinicopathologic, immunohistochemical, and ultrastructural findings of a fatal case of Middle East respiratory syndrome coronavirus infection in the united Arab Emirates, April 2014. Am J Pathol 2016;186:652–658.
    1. Alagaili AN, Briese T, Mishra N, Kapoor V, Sameroff SC, Burbelo PD, de Wit E, Munster VJ, Hensley LE, Zalmout IS, et al. Middle East respiratory syndrome coronavirus infection in dromedary camels in Saudi Arabia. MBio 2014;5:e00884-14
    1. De Benedictis P, Marciano S, Scaravelli D, Priori P, Zecchin B, Capua I, Monne I, Cattoli G. Alpha and lineage C betaCoV infections in Italian bats. Virus Genes 2014;48:366–371.
    1. Wacharapluesadee S, Sintunawa C, Kaewpom T, Khongnomnan K, Olival KJ, Epstein JH, Rodpan A, Sangsri P, Intarut N, Chindamporn A, et al. Group C betacoronavirus in bat guano fertilizer, Thailand. Emerg Infect Dis 2013;19:1349–1351.
    1. Chowell G, Abdirizak F, Lee S, Lee J, Jung E, Nishiura H, Viboud C. Transmission characteristics of MERS and SARS in the healthcare setting: a comparative study. BMC Med 2015;13:210.
    1. Raj VS, Mou H, Smits SL, Dekkers DH, Müller MA, Dijkman R, Muth D, Demmers JA, Zaki A, Fouchier RA, et al. Dipeptidyl peptidase 4 is a functional receptor for the emerging human coronavirus-EMC. Nature 2013;495:251–254.
    1. Meyerholz DK, Lambertz AM, McCray PB Jr. Dipeptidyl peptidase 4 distribution in the human respiratory tract: implications for the Middle East respiratory syndrome. Am J Pathol 2016;186:78–86.
    1. Lau SK, Lau CC, Chan KH, Li CP, Chen H, Jin DY, Chan JF, Woo PC, Yuen KY. Delayed induction of proinflammatory cytokines and suppression of innate antiviral response by the novel Middle East respiratory syndrome coronavirus: implications for pathogenesis and treatment. J Gen Virol 2013;94:2679–2690.
    1. Tynell J, Westenius V, Rönkkö E, Munster VJ, Melén K, Österlund P, Julkunen I. Middle East respiratory syndrome coronavirus shows poor replication but significant induction of antiviral responses in human monocyte-derived macrophages and dendritic cells. J Gen Virol 2016;97:344–355.
    1. Zhou J, Chu H, Li C, Wong BH, Cheng ZS, Poon VK, Sun T, Lau CC, Wong KK, Chan JY, et al. Active replication of Middle East respiratory syndrome coronavirus and aberrant induction of inflammatory cytokines and chemokines in human macrophages: implications for pathogenesis. J Infect Dis 2014;209:1331–1342.
    1. Min CK, Cheon S, Ha NY, Sohn KM, Kim Y, Aigerim A, Shin HM, Choi JY, Inn KS, Kim JH, et al. Comparative and kinetic analysis of viral shedding and immunological responses in MERS patients representing a broad spectrum of disease severity. Sci Rep 2016;6:25359.
    1. Wu JT, Leung K, Leung GM. Nowcasting and forecasting the potential domestic and international spread of the 2019-nCoV outbreak originating in Wuhan, China: a modelling study. Lancet 2020;395:689–697.
    1. World Health Organization. coronavirus disease (covid-19) pandemic [internet]. [accessed on 26 June 2020].
    1. Xu X, Chen P, Wang J, Feng J, Zhou H, Li X, Zhong W, Hao P. Evolution of the novel coronavirus from the ongoing Wuhan outbreak and modeling of its spike protein for risk of human transmission. Sci China Life Sci 2020;63:457–460.
    1. Ji W, Wang W, Zhao X, Zai J, Li X. Cross-species transmission of the newly identified coronavirus 2019-nCoV. J Med Virol 2020;92:433–440.
    1. Lam TT, Jia N, Zhang YW, Shum MH, Jiang JF, Zhu HC, Tong YG, Shi YX, Ni XB, Liao YS, et al. Identifying SARS-CoV-2-related coronaviruses in Malayan pangolins. Nature. 2020 [doi: 10.1038/s41586-020-2169-0]
    1. Walls AC, Park YJ, Tortorici MA, Wall A, McGuire AT, Veesler D. Structure, function, and antigenicity of the SARS-CoV-2 spike glycoprotein. Cell 2020;181:281–292.e6.
    1. Yan R, Zhang Y, Li Y, Xia L, Guo Y, Zhou Q. Structural basis for the recognition of SARS-CoV-2 by full-length human ACE2. Science 2020;367:1444–1448.
    1. Letko M, Marzi A, Munster V. Functional assessment of cell entry and receptor usage for SARS-CoV-2 and other lineage B betacoronaviruses. Nat Microbiol 2020;5:562–569.
    1. Leung JM, Yang CX, Tam A, Shaipanich T, Hackett TL, Singhera GK, Dorscheid DR, Sin DD. ACE-2 expression in the small airway epithelia of smokers and COPD patients: implications for COVID-19. Eur Respir J 2020;55:2000688
    1. Chen L, Li X, Chen M, Feng Y, Xiong C. The ACE2 expression in human heart indicates new potential mechanism of heart injury among patients infected with SARS-CoV-2. Cardiovasc Res 2020;116:1097–1100.
    1. Huang C, Wang Y, Li X, Ren L, Zhao J, Hu Y, Zhang L, Fan G, Xu J, Gu X, et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 2020;395:497–506.
    1. Lin L, Jiang X, Zhang Z, Huang S, Zhang Z, Fang Z, Gu Z, Gao L, Shi H, Mai L, et al. Gastrointestinal symptoms of 95 cases with SARS-CoV-2 infection. Gut 2020;69:997–1001.
    1. Spinato G, Fabbris C, Polesel J, Cazzador D, Borsetto D, Hopkins C, Boscolo-Rizzo P. Alterations in smell or taste in mildly symptomatic outpatients with SARS-CoV-2 infection. JAMA 2020;323:2089.
    1. Blanco-Melo D, Nilsson-Payant BE, Liu WC, Uhl S, Hoagland D, Møller R, Jordan TX, Oishi K, Panis M, Sachs D, et al. Imbalanced host response to SARS-CoV-2 drives development of COVID-19. Cell 2020;181:1036–1045.e9.
    1. Vanderheiden A, Ralfs P, Chirkova T, Upadhyay AA, Zimmerman MG, Bedoya S, Aoued H, Tharpb GM, Pellegrini KL, Lowen AC, et al. Type I and type III IFN restrict SARS-CoV-2 infection of human airway epithelial cultures. bioRxiv. 2020 [doi: 10.1101/2020.05.19.105437]
    1. Konno Y, Kimura I, Uriu K, Fukushi M, Irie T, Koyanagi Y, Nakagawa S, Sato K. SARS-CoV-2 ORF3b is a potent interferon antagonist whose activity is further increased by a naturally occurring elongation variant. bioRxiv. 2020 [doi: 10.1101/2020.05.11.088179]
    1. Liao M, Liu Y, Yuan J, Wen Y, Xu G, Zhao J, Cheng L, Li J, Wang X, Wang F, et al. Single-cell landscape of bronchoalveolar immune cells in patients with COVID-19. Nat Med 2020;26:842–844.
    1. Qin C, Zhou L, Hu Z, Zhang S, Yang S, Tao Y, Xie C, Ma K, Shang K, Wang W, et al. Dysregulation of immune response in patients with COVID-19 in Wuhan, China. Clin Infect Dis. 2020 [doi: 10.1093/cid/ciaa248]
    1. Zhang X, Tan Y, Ling Y, Lu G, Liu F, Yi Z, Jia X, Wu M, Shi B, Xu S, et al. Viral and host factors related to the clinical outcome of COVID-19. Nature. 2020 [doi: 10.1038/s41586-020-2355-0]
    1. Diao B, Wang C, Tan Y, Chen X, Liu Y, Ning L, Chen L, Li M, Liu Y, Wang G, et al. Reduction and functional exhaustion of t cells in patients with coronavirus disease 2019 (COVID-19). Front Immunol 2020;11:827.
    1. Li T, Qiu Z, Zhang L, Han Y, He W, Liu Z, Ma X, Fan H, Lu W, Xie J, et al. Significant changes of peripheral T lymphocyte subsets in patients with severe acute respiratory syndrome. J Infect Dis 2004;189:648–651.
    1. Chu H, Zhou J, Wong BH, Li C, Chan JF, Cheng ZS, Yang D, Wang D, Lee AC, Li C, et al. Middle East respiratory syndrome coronavirus efficiently infects human primary t lymphocytes and activates the extrinsic and intrinsic apoptosis pathways. J Infect Dis 2016;213:904–914.
    1. Zheng M, Gao Y, Wang G, Song G, Liu S, Sun D, Xu Y, Tian Z. Functional exhaustion of antiviral lymphocytes in COVID-19 patients. Cell Mol Immunol 2020;17:533–535.
    1. Wang LF, Shi Z, Zhang S, Field H, Daszak P, Eaton BT. Review of bats and SARS. Emerg Infect Dis 2006;12:1834–1840.
    1. 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 2004;173:4030–4039.
    1. Hogan RJ, Gao G, Rowe T, Bell P, Flieder D, Paragas J, Kobinger GP, Wivel NA, Crystal RG, Boyer J, et al. Resolution of primary severe acute respiratory syndrome-associated coronavirus infection requires Stat1. J Virol 2004;78:11416–11421.
    1. Frieman M, Yount B, Agnihothram S, Page C, Donaldson E, Roberts A, Vogel L, Woodruff B, Scorpio D, Subbarao K, et al. Molecular determinants of severe acute respiratory syndrome coronavirus pathogenesis and virulence in young and aged mouse models of human disease. J Virol 2012;86:884–897.
    1. Roberts A, Paddock C, Vogel L, Butler E, Zaki S, Subbarao K. Aged BALB/c mice as a model for increased severity of severe acute respiratory syndrome in elderly humans. J Virol 2005;79:5833–5838.
    1. Zhao J, Zhao J, Legge K, Perlman S. Age-related increases in PGD2 expression impair respiratory DC migration, resulting in diminished T cell responses upon respiratory virus infection in mice. J Clin Invest 2011;121:4921–4930.
    1. Yang XH, Deng W, Tong Z, Liu YX, Zhang LF, Zhu H, Gao H, Huang L, Liu YL, Ma CM, et al. Mice transgenic for human angiotensin-converting enzyme 2 provide a model for SARS coronavirus infection. Comp Med 2007;57:450–459.
    1. McCray PB Jr, Pewe L, Wohlford-Lenane C, Hickey M, Manzel L, Shi L, Netland J, Jia HP, Halabi C, Sigmund CD, et al. Lethal infection of K18-hACE2 mice infected with severe acute respiratory syndrome coronavirus. J Virol 2007;81:813–821.
    1. Frieman MB, Chen J, Morrison TE, Whitmore A, Funkhouser W, Ward JM, Lamirande EW, Roberts A, Heise M, Subbarao K, et al. SARS-CoV pathogenesis is regulated by a STAT1 dependent but a type I, II and III interferon receptor independent mechanism. PLoS Pathog 2010;6:e1000849
    1. Sheahan T, Morrison TE, Funkhouser W, Uematsu S, Akira S, Baric RS, Heise MT. MyD88 is required for protection from lethal infection with a mouse-adapted SARS-CoV. PLoS Pathog 2008;4:e1000240
    1. Roberts A, Vogel L, Guarner J, Hayes N, Murphy B, Zaki S, Subbarao K. Severe acute respiratory syndrome coronavirus infection of golden Syrian hamsters. J Virol 2005;79:503–511.
    1. Chu YK, Ali GD, Jia F, Li Q, Kelvin D, Couch RC, Harrod KS, Hutt JA, Cameron C, Weiss SR, et al. The SARS-CoV ferret model in an infection-challenge study. Virology 2008;374:151–163.
    1. Martina BE, Haagmans BL, Kuiken T, Fouchier RA, Rimmelzwaan GF, Van Amerongen G, Peiris JS, Lim W, Osterhaus AD. Virology: SARS virus infection of cats and ferrets. Nature 2003;425:915.
    1. Rowe T, Gao G, Hogan RJ, Crystal RG, Voss TG, Grant RL, Bell P, Kobinger GP, Wivel NA, Wilson JM. Macaque model for severe acute respiratory syndrome. J Virol 2004;78:11401–11404.
    1. McAuliffe J, Vogel L, Roberts A, Fahle G, Fischer S, Shieh WJ, Butler E, Zaki S, St Claire M, Murphy B, et al. Replication of SARS coronavirus administered into the respiratory tract of African Green, rhesus and cynomolgus monkeys. Virology 2004;330:8–15.
    1. Lawler JV, Endy TP, Hensley LE, Garrison A, Fritz EA, Lesar M, Baric RS, Kulesh DA, Norwood DA, Wasieloski LP, et al. Cynomolgus macaque as an animal model for severe acute respiratory syndrome. PLoS Med 2006;3:e149
    1. Greenough TC, Carville A, Coderre J, Somasundaran M, Sullivan JL, Luzuriaga K, Mansfield K. Pneumonitis and multi-organ system disease in common marmosets (Callithrix jacchus) infected with the severe acute respiratory syndrome-associated coronavirus. Am J Pathol 2005;167:455–463.
    1. Zhao J, Li K, Wohlford-Lenane C, Agnihothram SS, Fett C, Zhao J, Gale MJ Jr, Baric RS, Enjuanes L, Gallagher T, et al. Rapid generation of a mouse model for Middle East respiratory syndrome. Proc Natl Acad Sci U S A 2014;111:4970–4975.
    1. Agrawal AS, Garron T, Tao X, Peng BH, Wakamiya M, Chan TS, Couch RB, Tseng CT. Generation of a transgenic mouse model of Middle East respiratory syndrome coronavirus infection and disease. J Virol 2015;89:3659–3670.
    1. Li K, Wohlford-Lenane C, Perlman S, Zhao J, Jewell AK, Reznikov LR, Gibson-Corley KN, Meyerholz DK, McCray PB Jr. Middle East respiratory syndrome coronavirus causes multiple organ damage and lethal disease in mice transgenic for human dipeptidyl peptidase 4. J Infect Dis 2016;213:712–722.
    1. van Doremalen N, Munster VJ. Animal models of Middle East respiratory syndrome coronavirus infection. Antiviral Res 2015;122:28–38.
    1. Li K, Wohlford-Lenane CL, Channappanavar R, Park JE, Earnest JT, Bair TB, Bates AM, Brogden KA, Flaherty HA, Gallagher T, et al. Mouse-adapted MERS coronavirus causes lethal lung disease in human DPP4 knockin mice. Proc Natl Acad Sci U S A 2017;114:E3119–E3128.
    1. Cockrell AS, Yount BL, Scobey T, Jensen K, Douglas M, Beall A, Tang XC, Marasco WA, Heise MT, Baric RS. A mouse model for MERS coronavirus-induced acute respiratory distress syndrome. Nat Microbiol 2016;2:16226.
    1. Adney DR, Bielefeldt-Ohmann H, Hartwig AE, Bowen RA. Infection, replication, and transmission of Middle East respiratory syndrome coronavirus in alpacas. Emerg Infect Dis 2016;22:1031–1037.
    1. Adney DR, van Doremalen N, Brown VR, Bushmaker T, Scott D, de Wit E, Bowen RA, Munster VJ. Replication and shedding of MERS-CoV in upper respiratory tract of inoculated dromedary camels. Emerg Infect Dis 2014;20:1999–2005.
    1. Munster VJ, de Wit E, Feldmann H. Pneumonia from human coronavirus in a macaque model. N Engl J Med 2013;368:1560–1562.
    1. Falzarano D, de Wit E, Feldmann F, Rasmussen AL, Okumura A, Peng X, Thomas MJ, van Doremalen N, Haddock E, Nagy L, et al. Infection with MERS-CoV causes lethal pneumonia in the common marmoset. PLoS Pathog 2014;10:e1004250
    1. Yao Y, Bao L, Deng W, Xu L, Li F, Lv Q, Yu P, Chen T, Xu Y, Zhu H, et al. An animal model of MERS produced by infection of rhesus macaques with MERS coronavirus. J Infect Dis 2014;209:236–242.
    1. Bao L, Deng W, Huang B, Gao H, Liu J, Ren L, Wei Q, Yu P, Xu Y, Qi F, et al. The pathogenicity of SARS-CoV-2 in hACE2 transgenic mice. Nature. 2020 [doi: 10.1038/s41586-020-2312-y]
    1. Chan JF, Zhang AJ, Yuan S, Poon VK, Chan CC, Lee AC, Chan WM, Fan Z, Tsoi HW, Wen L, et al. Simulation of the clinical and pathological manifestations of coronavirus disease 2019 (COVID-19) in golden Syrian hamster model: implications for disease pathogenesis and transmissibility. Clin Infect Dis. 2020 [doi: 10.1093/cid/ciaa325]
    1. Kim YI, Kim SG, Kim SM, Kim EH, Park SJ, Yu KM, Chang JH, Kim EJ, Lee S, Casel MA, et al. Infection and rapid transmission of SARS-CoV-2 in ferrets. Cell Host Microbe 2020;27:704–709.e2.
    1. Shi J, Wen Z, Zhong G, Yang H, Wang C, Huang B, Liu R, He X, Shuai L, Sun Z, et al. Susceptibility of ferrets, cats, dogs, and other domesticated animals to SARS-coronavirus 2. Science 2020;368:1016–1020.
    1. Rockx B, Kuiken T, Herfst S, Bestebroer T, Lamers MM, Oude Munnink BB, de Meulder D, van Amerongen G, van den Brand J, Okba NM, et al. Comparative pathogenesis of COVID-19, MERS, and SARS in a nonhuman primate model. Science 2020;368:1012–1015.
    1. Yu P, Qi F, Xu Y, Li F, Liu P, Liu J, Bao L, Deng W, Gao H, Xiang Z, et al. Age-related rhesus macaque models of COVID-19. Animal Model Exp Med 2020;3:93–97.
    1. Sun J, Zhuang Z, Zheng J, Li K, Wong RL, Liu D, Huang J, He J, Zhu A, Zhao J, et al. Generation of a broadly useful model for covid-19 pathogenesis, vaccination, and treatment. Cell. 2020 [doi: 10.1016/j.cell.2020.06.010]
    1. Netland J, DeDiego ML, Zhao J, Fett C, Álvarez E, Nieto-Torres JL, Enjuanes L, Perlman S. Immunization with an attenuated severe acute respiratory syndrome coronavirus deleted in E protein protects against lethal respiratory disease. Virology 2010;399:120–128.
    1. Lamirande EW, DeDiego ML, Roberts A, Jackson JP, Alvarez E, Sheahan T, Shieh WJ, Zaki SR, 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.
    1. Menachery VD, Yount BL Jr, Josset L, Gralinski LE, Scobey T, Agnihothram S, Katze MG, Baric RS. Attenuation and restoration of severe acute respiratory syndrome coronavirus mutant lacking 2′-O-methyltransferase activity. J Virol 2014;88:4251–4264.
    1. Almazán F, DeDiego ML, Sola I, Zuñiga S, Nieto-Torres JL, Marquez-Jurado S, Andrés G, Enjuanes L. Engineering a replication-competent, propagation-defective Middle East respiratory syndrome coronavirus as a vaccine candidate. MBio 2013;4:e00650-13
    1. Menachery VD, Gralinski LE, Mitchell HD, Dinnon KH 3rd, Leist SR, Yount BL Jr, Graham RL, McAnarney ET, Stratton KG, Cockrell AS, et al. Middle East respiratory syndrome coronavirus nonstructural protein 16 is necessary for interferon resistance and viral pathogenesis. MSphere 2017;2:e00346-17
    1. Takasuka N, Fujii H, Takahashi Y, Kasai M, Morikawa S, Itamura S, Ishii K, Sakaguchi M, Ohnishi K, Ohshima M, et al. A subcutaneously injected UV-inactivated SARS coronavirus vaccine elicits systemic humoral immunity in mice. Int Immunol 2004;16:1423–1430.
    1. Spruth M, Kistner O, Savidis-Dacho H, Hitter E, Crowe B, Gerencer M, Brühl P, Grillberger L, Reiter M, Tauer C, et al. A double-inactivated whole virus candidate SARS coronavirus vaccine stimulates neutralising and protective antibody responses. Vaccine 2006;24:652–661.
    1. Kong WP, Xu L, Stadler K, Ulmer JB, Abrignani S, Rappuoli R, Nabel GJ. Modulation of the immune response to the severe acute respiratory syndrome spike glycoprotein by gene-based and inactivated virus immunization. J Virol 2005;79:13915–13923.
    1. Agrawal AS, Tao X, Algaissi A, Garron T, Narayanan K, Peng BH, Couch RB, Tseng CT. Immunization with inactivated Middle East respiratory syndrome coronavirus vaccine leads to lung immunopathology on challenge with live virus. Hum Vaccin Immunother 2016;12:2351–2356.
    1. 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.
    1. Gao Q, Bao L, Mao H, Wang L, Xu K, Yang M, Li Y, Zhu L, Wang N, Lv Z, et al. Development of an inactivated vaccine candidate for SARS-CoV-2. Science. 2020 [doi: 10.1126/science.abc1932]
    1. He Y, Lu H, Siddiqui P, Zhou Y, Jiang S. Receptor-binding domain of severe acute respiratory syndrome coronavirus spike protein contains multiple conformation-dependent epitopes that induce highly potent neutralizing antibodies. J Immunol 2005;174:4908–4915.
    1. He Y, Zhou Y, Liu S, Kou Z, Li W, Farzan M, Jiang S. Receptor-binding domain of SARS-CoV spike protein induces highly potent neutralizing antibodies: implication for developing subunit vaccine. Biochem Biophys Res Commun 2004;324:773–781.
    1. Bisht H, Roberts A, Vogel L, Subbarao K, Moss B. Neutralizing antibody and protective immunity to SARS coronavirus infection of mice induced by a soluble recombinant polypeptide containing an N-terminal segment of the spike glycoprotein. Virology 2005;334:160–165.
    1. He Y, Li J, Heck S, Lustigman S, Jiang S. Antigenic and immunogenic characterization of recombinant baculovirus-expressed severe acute respiratory syndrome coronavirus spike protein: implication for vaccine design. J Virol 2006;80:5757–5767.
    1. Kam YW, Kien F, Roberts A, Cheung YC, Lamirande EW, Vogel L, Chu SL, Tse J, Guarner J, Zaki SR, 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 2007;25:729–740.
    1. Mou H, Raj VS, van Kuppeveld FJ, Rottier PJ, Haagmans BL, Bosch BJ. 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.
    1. Lan J, Yao Y, Deng Y, Chen H, Lu G, Wang W, Bao L, Deng W, Wei Q, Gao GF, 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.
    1. Zhang N, Channappanavar R, Ma C, Wang L, Tang J, Garron T, Tao X, Tasneem S, Lu L, Tseng CT, et al. Identification of an ideal adjuvant for receptor-binding domain-based subunit vaccines against Middle East respiratory syndrome coronavirus. Cell Mol Immunol 2016;13:180–190.
    1. Tang J, Zhang N, Tao X, Zhao G, Guo Y, Tseng CT, Jiang S, Du L, Zhou Y. Optimization of antigen dose for a receptor-binding domain-based subunit vaccine against MERS coronavirus. Hum Vaccin Immunother 2015;11:1244–1250.
    1. Du L, Kou Z, Ma C, Tao X, Wang L, Zhao G, Chen Y, Yu F, Tseng CT, Zhou Y, et al. A truncated receptor-binding domain of MERS-CoV spike protein potently inhibits MERS-CoV infection and induces strong neutralizing antibody responses: implication for developing therapeutics and vaccines. PLoS One 2013;8:e81587
    1. Tai W, Zhao G, Sun S, Guo Y, Wang Y, Tao X, Tseng CK, 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.
    1. Ma C, Li Y, Wang L, Zhao G, Tao X, Tseng CT, 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.
    1. Wang L, Shi W, Joyce MG, Modjarrad K, Zhang Y, Leung K, Lees CR, Zhou T, Yassine HM, Kanekiyo M, et al. Evaluation of candidate vaccine approaches for MERS-CoV. Nat Commun 2015;6:7712.
    1. Jiaming L, Yanfeng Y, Yao D, Yawei H, Linlin B, Baoying H, Jinghua Y, Gao GF, 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.
    1. Coleman CM, Liu YV, Mu H, Taylor JK, Massare M, Flyer DC, Smith GE, Frieman MB. Purified coronavirus spike protein nanoparticles induce coronavirus neutralizing antibodies in mice. Vaccine 2014;32:3169–3174.
    1. Coleman CM, Venkataraman T, Liu YV, Glenn GM, Smith GE, Flyer DC, Frieman MB. MERS-CoV spike nanoparticles protect mice from MERS-CoV infection. Vaccine 2017;35:1586–1589.
    1. Jung SY, Kang KW, Lee EY, Seo DW, Kim HL, Kim H, Kwon T, Park HL, Kim H, Lee SM, et al. 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.
    1. Mortola E, Roy P. Efficient assembly and release of SARS coronavirus-like particles by a heterologous expression system. FEBS Lett 2004;576:174–178.
    1. Lu B, Huang Y, Huang L, Li B, Zheng Z, Chen Z, Chen J, Hu Q, Wang H. Effect of mucosal and systemic immunization with virus-like particles of severe acute respiratory syndrome coronavirus in mice. Immunology 2010;130:254–261.
    1. Lu X, Chen Y, Bai B, Hu H, Tao L, Yang J, Chen J, Chen Z, Hu Z, Wang H. Immune responses against severe acute respiratory syndrome coronavirus induced by virus-like particles in mice. Immunology 2007;122:496–502.
    1. Liu YV, Massare MJ, Barnard DL, Kort T, Nathan M, Wang L, Smith G. Chimeric severe acute respiratory syndrome coronavirus (SARS-CoV) S glycoprotein and influenza matrix 1 efficiently form virus-like particles (VLPs) that protect mice against challenge with SARS-CoV. Vaccine 2011;29:6606–6613.
    1. Wang C, Zheng X, Gai W, Zhao Y, Wang H, Wang H, Feng N, Chi H, Qiu B, Li N, 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.
    1. Yang ZY, Kong WP, Huang Y, Roberts A, Murphy BR, Subbarao K, Nabel GJ. A DNA vaccine induces SARS coronavirus neutralization and protective immunity in mice. Nature 2004;428:561–564.
    1. Hu H, Lu X, Tao L, Bai B, Zhang Z, Chen Y, Zheng F, Chen J, Chen Z, Wang H. Induction of specific immune responses by severe acute respiratory syndrome coronavirus spike DNA vaccine with or without interleukin-2 immunization using different vaccination routes in mice. Clin Vaccine Immunol 2007;14:894–901.
    1. 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 2005;23:4385–4391.
    1. Chunling M, Kun Y, Jian X, Jian Q, Hua S, Minsheng Z. Enhanced induction of SARS-CoV nucleocapsid protein-specific immune response using DNA vaccination followed by adenovirus boosting in BALB/c mice. Intervirology 2006;49:307–318.
    1. Hu H, Tao L, Wang Y, Chen L, Yang J, Wang H. Enhancing immune responses against SARS-CoV nucleocapsid DNA vaccine by co-inoculating interleukin-2 expressing vector in mice. Biotechnol Lett 2009;31:1685–1693.
    1. Muthumani K, Falzarano D, Reuschel EL, Tingey C, Flingai S, Villarreal DO, 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
    1. Al-Amri SS, Abbas AT, Siddiq LA, Alghamdi A, Sanki MA, Al-Muhanna MK, Alhabbab RY, Azhar EI, Li X, Hashem AM. Immunogenicity of candidate MERS-CoV DNA vaccines based on the spike protein. Sci Rep 2017;7:44875.
    1. Smith TR, Patel A, Ramos S, Elwood D, Zhu X, Yan J, Gary EN, Walker SN, Schultheis K, Purwar M, et al. Immunogenicity of a DNA vaccine candidate for COVID-19. Nat Commun 2020;11:2601.
    1. Yu J, Tostanoski LH, Peter L, Mercado NB, McMahan K, Mahrokhian SH, Nkolola JP, Liu J, Li Z, Chandrashekar A, et al. DNA vaccine protection against SARS-CoV-2 in rhesus macaques. Science. 2020 [doi: 10.1126/science.abc6284]
    1. Czub M, Weingartl H, Czub S, He R, Cao J. Evaluation of modified vaccinia virus Ankara based recombinant SARS vaccine in ferrets. Vaccine 2005;23:2273–2279.
    1. Chen Z, Zhang L, Qin C, Ba L, Yi CE, Zhang F, Wei Q, He T, Yu W, Yu J, et al. Recombinant modified vaccinia virus Ankara expressing the spike glycoprotein of severe acute respiratory syndrome coronavirus induces protective neutralizing antibodies primarily targeting the receptor binding region. J Virol 2005;79:2678–2688.
    1. Bisht H, Roberts A, Vogel L, Bukreyev A, Collins PL, Murphy BR, Subbarao K, Moss B. Severe acute respiratory syndrome coronavirus spike protein expressed by attenuated vaccinia virus protectively immunizes mice. Proc Natl Acad Sci U S A 2004;101:6641–6646.
    1. 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.
    1. Kim E, Okada K, Kenniston T, Raj VS, AlHajri MM, Farag EA, AlHajri F, Osterhaus AD, Haagmans BL, Gambotto A. Immunogenicity of an adenoviral-based Middle East respiratory syndrome coronavirus vaccine in BALB/c mice. Vaccine 2014;32:5975–5982.
    1. Dicks MD, Spencer AJ, Edwards NJ, Wadell G, Bojang K, Gilbert SC, Hill AV, Cottingham MG. A novel chimpanzee adenovirus vector with low human seroprevalence: improved systems for vector derivation and comparative immunogenicity. PLoS One 2012;7:e40385
    1. Alharbi NK, Padron-Regalado E, Thompson CP, Kupke A, Wells D, Sloan MA, 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.
    1. Munster VJ, Wells D, Lambe T, Wright D, Fischer RJ, Bushmaker T, Saturday G, van Doremalen N, Gilbert SC, 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.
    1. Alharbi NK, Qasim I, Almasoud A, Aljami HA, Alenazi MW, Alhafufi A, Aldibasi OS, Hashem AM, Kasem S, Albrahim R, et al. Humoral immunogenicity and efficacy of a single dose of ChAdOx1 MERS vaccine candidate in dromedary camels. Sci Rep 2019;9:16292.
    1. 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.
    1. Haagmans BL, van den Brand JM, Raj VS, Volz A, Wohlsein P, Smits SL, Schipper D, Bestebroer TM, 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.
    1. Veit S, Jany S, Fux R, Sutter G, Volz A. CD8+ T cells responding to the Middle East respiratory syndrome coronavirus nucleocapsid protein delivered by vaccinia virus MVA in mice. Viruses 2018;10:718.
    1. Bodmer BS, Fiedler AH, Hanauer JR, Prüfer S, Mühlebach MD. 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.
    1. Malczyk AH, Kupke A, Prüfer S, Scheuplein VA, 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.
    1. Wirblich C, Coleman CM, Kurup D, Abraham TS, Bernbaum JG, Jahrling PB, Hensley LE, Johnson RF, Frieman MB, Schnell MJ. 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
    1. World Health Organization. Draft landscape of COVID-19 candidate vaccines [Internet]. [accessed on 22 June 2020].
    1. Roberts A, Deming D, Paddock CD, Cheng A, Yount B, Vogel L, Herman BD, Sheahan T, Heise M, Genrich GL, et al. A mouse-adapted SARS-coronavirus causes disease and mortality in BALB/c mice. PLoS Pathog 2007;3:e5
    1. Day CW, Baric R, Cai SX, Frieman M, Kumaki Y, Morrey JD, Smee DF, Barnard DL. A new mouse-adapted strain of SARS-CoV as a lethal model for evaluating antiviral agents in vitro and in vivo . Virology 2009;395:210–222.
    1. van Doremalen N, Miazgowicz KL, Milne-Price S, Bushmaker T, Robertson S, Scott D, Kinne J, McLellan JS, Zhu J, Munster VJ. Host species restriction of Middle East respiratory syndrome coronavirus through its receptor, dipeptidyl peptidase 4. J Virol 2014;88:9220–9232.
    1. de Wit E, Prescott J, Baseler L, Bushmaker T, Thomas T, Lackemeyer MG, Martellaro C, Milne-Price S, Haddock E, Haagmans BL, et al. The Middle East respiratory syndrome coronavirus (MERS-CoV) does not replicate in Syrian hamsters. PLoS One 2013;8:e69127
    1. Cockrell AS, Peck KM, Yount BL, Agnihothram SS, Scobey T, Curnes NR, Baric RS, Heise MT. Mouse dipeptidyl peptidase 4 is not a functional receptor for Middle East respiratory syndrome coronavirus infection. J Virol 2014;88:5195–5199.
    1. Lauring AS, Jones JO, Andino R. Rationalizing the development of live attenuated virus vaccines. Nat Biotechnol 2010;28:573–579.
    1. See RH, Zakhartchouk AN, Petric M, Lawrence DJ, Mok CP, Hogan RJ, Rowe T, Zitzow LA, Karunakaran KP, Hitt MM, et al. Comparative evaluation of two severe acute respiratory syndrome (SARS) vaccine candidates in mice challenged with SARS coronavirus. J Gen Virol 2006;87:641–650.
    1. Stadler K, Roberts A, Becker S, Vogel L, Eickmann M, Kolesnikova L, Klenk HD, Murphy B, Rappuoli R, Abrignani S, et al. SARS vaccine protective in mice. Emerg Infect Dis 2005;11:1312–1314.
    1. Qin E, Shi H, Tang L, Wang C, Chang G, Ding Z, Zhao K, Wang J, Chen Z, Yu M, et al. Immunogenicity and protective efficacy in monkeys of purified inactivated Vero-cell SARS vaccine. Vaccine 2006;24:1028–1034.
    1. Zhou J, Wang W, Zhong Q, Hou W, Yang Z, Xiao SY, Zhu R, Tang Z, Wang Y, Xian Q, et al. Immunogenicity, safety, and protective efficacy of an inactivated SARS-associated coronavirus vaccine in rhesus monkeys. Vaccine 2005;23:3202–3209.
    1. Crank MC, Ruckwardt TJ, Chen M, Morabito KM, Phung E, Costner PJ, Holman LA, Hickman SP, Berkowitz NM, Gordon IJ, et al. A proof of concept for structure-based vaccine design targeting RSV in humans. Science 2019;365:505–509.
    1. Valkenburg SA, Mallajosyula VV, Li OT, Chin AW, Carnell G, Temperton N, Varadarajan R, Poon LL. Stalking influenza by vaccination with pre-fusion headless HA mini-stem. Sci Rep 2016;6:22666.
    1. McLellan JS, Chen M, Joyce MG, Sastry M, Stewart-Jones GB, 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.
    1. Cheng C, Pancera M, Bossert A, Schmidt SD, Chen RE, Chen X, Druz A, Narpala S, Doria-Rose NA, McDermott AB, et al. Immunogenicity of a prefusion HIV-1 envelope trimer in complex with a quaternary-structure-specific antibody. J Virol 2015;90:2740–2755.
    1. Kushnir N, Streatfield SJ, Yusibov V. Virus-like particles as a highly efficient vaccine platform: diversity of targets and production systems and advances in clinical development. Vaccine 2012;31:58–83.
    1. Tang DC, DeVit M, Johnston SA. Genetic immunization is a simple method for eliciting an immune response. Nature 1992;356:152–154.
    1. Choi YJ, Hur SY, Kim TJ, Hong SR, Lee JK, Cho CH, Park KS, Woo JW, Sung YC, Suh YS, et al. A phase II, prospective, randomized, multicenter, open-label study of GX-188E, an HPV DNA vaccine, in patients with cervical intraepithelial neoplasia 3. Clin Cancer Res 2020;26:1616–1623.
    1. Gary EN, Weiner DB. DNA vaccines: prime time is now. Curr Opin Immunol 2020;65:21–27.
    1. Pardi N, Hogan MJ, Porter FW, Weissman D. mRNA vaccines - a new era in vaccinology. Nat Rev Drug Discov 2018;17:261–279.
    1. Food and Drug Administration. Approval letter - Ervebo (Ebloa Zaire Vaccine, Live) [Internet]. [accessed on 19 December 2019].
    1. Ura T, Okuda K, Shimada M. Developments in viral vector-based vaccines. Vaccines (Basel) 2014;2:624–641.
    1. Du L, Zhao G, Lin Y, Sui H, Chan C, Ma S, He Y, Jiang S, Wu C, Yuen KY, 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 2008;180:948–956.
    1. Choi JH, Woo HM, Lee TY, Lee SY, Shim SM, Park WJ, Yang JS, Kim JA, Yun MR, Kim DW, et al. Characterization of a human monoclonal antibody generated from a B-cell specific for a prefusion-stabilized spike protein of Middle East respiratory syndrome coronavirus. PLoS One 2020;15:e0232757
    1. Widjaja I, Wang C, van Haperen R, Gutiérrez-Álvarez J, van Dieren B, Okba NM, Raj VS, Li W, Fernandez-Delgado R, Grosveld F, et al. Towards a solution to MERS: protective human monoclonal antibodies targeting different domains and functions of the MERS-coronavirus spike glycoprotein. Emerg Microbes Infect 2019;8:516–530.
    1. Coughlin MM, Babcook J, Prabhakar BS. Human monoclonal antibodies to SARS-coronavirus inhibit infection by different mechanisms. Virology 2009;394:39–46.
    1. Channappanavar R, Fett C, Zhao J, Meyerholz DK, Perlman S. Virus-specific memory CD8 T cells provide substantial protection from lethal severe acute respiratory syndrome coronavirus infection. J Virol 2014;88:11034–11044.
    1. Zhao J, Zhao J, Perlman S. T cell responses are required for protection from clinical disease and for virus clearance in severe acute respiratory syndrome coronavirus-infected mice. J Virol 2010;84:9318–9325.
    1. Zhao J, Zhao J, Mangalam AK, Channappanavar R, Fett C, Meyerholz DK, Agnihothram S, Baric RS, David CS, Perlman S. Airway memory CD4+ T cells mediate protective immunity against emerging respiratory coronaviruses. Immunity 2016;44:1379–1391.
    1. Tirado SM, Yoon KJ. Antibody-dependent enhancement of virus infection and disease. Viral Immunol 2003;16:69–86.
    1. Wan Y, Shang J, Sun S, Tai W, Chen J, Geng Q, He L, Chen Y, Wu J, Shi Z, et al. Molecular mechanism for antibody-dependent enhancement of coronavirus entry. J Virol 2020;94:e02015-19
    1. Polack FP, Hoffman SJ, Crujeiras G, Griffin DE. A role for nonprotective complement-fixing antibodies with low avidity for measles virus in atypical measles. Nat Med 2003;9:1209–1213.
    1. Polack FP, Teng MN, Collins PL, Prince GA, Exner M, Regele H, Lirman DD, Rabold R, Hoffman SJ, Karp CL, et al. A role for immune complexes in enhanced respiratory syncytial virus disease. J Exp Med 2002;196:859–865.
    1. Kim HW, Canchola JG, Brandt CD, Pyles G, Chanock RM, Jensen K, Parrott RH. Respiratory syncytial virus disease in infants despite prior administration of antigenic inactivated vaccine. Am J Epidemiol 1969;89:422–434.
    1. Luo F, Liao FL, Wang H, Tang HB, Yang ZQ, Hou W. Evaluation of antibody-dependent enhancement of SARS-CoV infection in rhesus macaques immunized with an inactivated SARS-CoV vaccine. Virol Sin 2018;33:201–204.
    1. Houser KV, Broadbent AJ, Gretebeck L, Vogel L, Lamirande EW, Sutton T, Bock KW, Minai M, Orandle M, Moore IN, et al. Enhanced inflammation in New Zealand white rabbits when MERS-CoV reinfection occurs in the absence of neutralizing antibody. PLoS Pathog 2017;13:e1006565
    1. Verity R, Okell LC, Dorigatti I, Winskill P, Whittaker C, Imai N, Cuomo-Dannenburg G, Thompson H, Walker PG, Fu H, et al. Estimates of the severity of coronavirus disease 2019: a model-based analysis. Lancet Infect Dis 2020;20:669–677.
    1. Cruz-Valdez A, Valdez-Zapata G, Patel SS, Castelli FV, Garcia MG, Jansen WT, Arora AK, Heijnen E. MF59-adjuvanted influenza vaccine (FLUAD®) elicits higher immune responses than a non-adjuvanted influenza vaccine (Fluzone®): a randomized, multicenter, Phase III pediatric trial in Mexico. Hum Vaccin Immunother 2018;14:386–395.
    1. Robertson CA, DiazGranados CA, Decker MD, Chit A, Mercer M, Greenberg DP. Fluzone® high-dose influenza vaccine. Expert Rev Vaccines 2016;15:1495–1505.
    1. Camilloni B, Basileo M, Valente S, Nunzi E, Iorio AM. Immunogenicity of intramuscular MF59-adjuvanted and intradermal administered influenza enhanced vaccines in subjects aged over 60: a literature review. Hum Vaccin Immunother 2015;11:553–563.
    1. Rogers TF, Zhao F, Huang D, Beutler N, Burns A, He WT, Limbo O, Smith C, Song G, Woehl J, et al. Isolation of potent SARS-CoV-2 neutralizing antibodies and protection from disease in a small animal model. Science. 2020 [doi: 10.1126/science.abc7520]
    1. Duan K, Liu B, Li C, Zhang H, Yu T, Qu J, Zhou M, Chen L, Meng S, Hu Y, et al. Effectiveness of convalescent plasma therapy in severe COVID-19 patients. Proc Natl Acad Sci U S A 2020;117:9490–9496.
    1. Chandrashekar A, Liu J, Martinot AJ, McMahan K, Mercado NB, Peter L, Tostanoski LH, Yu J, Maliga Z, Nekorchuk M, et al. SARS-CoV-2 infection protects against rechallenge in rhesus macaques. Science. 2020 [doi: 10.1126/science.abc4776]

Metrics
Share
Figures

1 / 1

Tables

1 / 1

Funding Information
PERMALINK