key: cord-352527-eeyqh9nc authors: Zhou, Yusen; Yang, Yang; Huang, Jingwei; Jiang, Shibo; Du, Lanying title: Advances in MERS-CoV Vaccines and Therapeutics Based on the Receptor-Binding Domain date: 2019-01-14 journal: Viruses DOI: 10.3390/v11010060 sha: doc_id: 352527 cord_uid: eeyqh9nc Middle East respiratory syndrome (MERS) coronavirus (MERS-CoV) is an infectious virus that was first reported in 2012. The MERS-CoV genome encodes four major structural proteins, among which the spike (S) protein has a key role in viral infection and pathogenesis. The receptor-binding domain (RBD) of the S protein contains a critical neutralizing domain and is an important target for development of MERS vaccines and therapeutics. In this review, we describe the relevant features of the MERS-CoV S-protein RBD, summarize recent advances in the development of MERS-CoV RBD-based vaccines and therapeutic antibodies, and illustrate potential challenges and strategies to further improve their efficacy. Middle East respiratory syndrome (MERS) coronavirus (CoV) is an infectious virus that was first reported in June 2012 [1] . MERS-CoV may infect people of any age, but older age, underlying comorbidity (such as diabetes mellitus, renal disease, respiratory disease, heart disease, and hypertension), and delayed confirmation or late diagnosis are all factors that affect MERS disease outcomes and mortality [2] [3] [4] [5] [6] [7] . Sex could be a factor in MERS epidemiology, as more males seem to be affected than females [8] [9] [10] . MERS-CoV infection of women during pregnancy has adverse outcomes, with fetal mortality of~27%; however, only a limited number of pediatric MERS-CoV infections occur [11] [12] [13] [14] . At the end of December 2018, 2,279 laboratory-confirmed MERS infections were reported globally (in 27 countries), leading to 806 deaths, and a mortality of 35.3%. Among these infections, 1,901 (83.4%) were reported in Saudi Arabia, with mortality in 732 individuals (38.5%) (http://www.emro.who.int/health-topics/mers-cov/mers-outbreaks.html). The largest MERS outbreak outside Saudi Arabia occurred in South Korea in 2015, with 186 cases and 38 deaths [9, 15, 16] . The most recent MERS cases were reported in 2018 in South Korea, the United Kingdom, and Malaysia, in addition to Saudi Arabia, the United Arab Emirates, and Oman (http://www.who.int/emergencies/mers-cov/en/). MERS-CoV is thought to have originated in bats [17] [18] [19] [20] . MERS-like viruses have been isolated from bats that use (at lower efficiency) the same receptor for cell entry as the MERS-CoV isolated from humans [21] [22] [23] . Dromedary camels are potential intermediates for long-term evolution of MERS-CoV and seasonal zoonotic transfer of virus to humans [24] [25] [26] [27] . Antibodies specific to host cellular proteases for its activity in viral entry, but although evidence initially indicated that cellular furin activates S protein, subsequent results have demonstrated no evidence for the involvement of furin during viral entry [71, 78] . The DPP4 receptor varies among different host species, and MERS-CoV is thought to use multiple pathways to enable rapid adaptation to speciesspecific variations [79] [80] [81] . In addition to DPP4, MERS-CoV can bind to sialic acid via the S1 subunit of S protein, or utilize the membrane-associated 78 kDa glucose-regulated protein (GRP78) to attach to target cells, suggesting that these proteins may also have roles in virion attachment [82, 83] . The structures of MERS-CoV RBD alone and complexed with DPP4 have been determined ( Figure 2 ) [77, 84, 85] . The RBD has a fold-rich tertiary structure, which consists of a core and a receptor-binding motif (RBM), with stabilization provided by four disulfide bonds and two glycans [77] . A number of RBD residues are located at the DPP4-binding interface, and they have a critical role in RBD-DPP4 binding [77, 84, 85] . Structural analysis of MERS-CoV trimeric S protein has identified specific features of the RBD and its complex with DPP4. Notably, in the prefusion conformation of the S trimer, individual RBDs are either buried (lying state) or exposed (standing state), and this flexibility presumably facilitates recognition by DPP4 [86] . Other structural studies have revealed four S-trimer conformational states, in which each RBD is either tightly packed at the membrane-distal apex or rotated into a receptor-accessible conformation, suggesting fusion initiation through sequential RBD events [87] . In configurations with one, two, or three RBDs rotated out, RBD determinants are exposed at the apex of the RBD-DPP4 complex, and they are accessible for interaction with DPP4 ( Figure 3 ) [87] . MERS-CoV S protein has an important role in viral pathogenesis, determining host tropism and entry into host cells [58, 70, 71] . The S protein contains an S1 subunit at the N terminus and an S2 subunit at the C terminus. The S1 subunit is composed of the N-terminal domain (NTD) and RBD [58, 72, 73] . The RBD has a key role in the mediation of binding of MERS-CoV to cells expressing dipeptidyl peptidase 4 (DPP4) receptor, enabling the virus to enter into target cells by fusing with cell membranes through the formation of a fusion core ( Figure 1C ) [74] [75] [76] [77] . The S protein requires host cellular proteases for its activity in viral entry, but although evidence initially indicated that cellular furin activates S protein, subsequent results have demonstrated no evidence for the involvement of furin during viral entry [71, 78] . The DPP4 receptor varies among different host species, and MERS-CoV is thought to use multiple pathways to enable rapid adaptation to species-specific variations [79] [80] [81] . In addition to DPP4, MERS-CoV can bind to sialic acid via the S1 subunit of S protein, or utilize the membrane-associated 78 kDa glucose-regulated protein (GRP78) to attach to target cells, suggesting that these proteins may also have roles in virion attachment [82, 83] . The structures of MERS-CoV RBD alone and complexed with DPP4 have been determined ( Figure 2 ) [77, 84, 85] . The RBD has a fold-rich tertiary structure, which consists of a core and a receptor-binding motif (RBM), with stabilization provided by four disulfide bonds and two glycans [77] . A number of RBD residues are located at the DPP4-binding interface, and they have a critical role in RBD-DPP4 binding [77, 84, 85] . Structural analysis of MERS-CoV trimeric S protein has identified specific features of the RBD and its complex with DPP4. Notably, in the prefusion conformation of the S trimer, individual RBDs are either buried (lying state) or exposed (standing state), and this flexibility presumably facilitates recognition by DPP4 [86] . Other structural studies have revealed four S-trimer conformational states, in which each RBD is either tightly packed at the membrane-distal apex or rotated into a receptor-accessible conformation, suggesting fusion initiation through sequential RBD The MERS-CoV RBD core is colored in blue, the RBM is colored in red, and DPP4 is colored in green. The RBM residues directly involved in DPP4 binding are shown as sticks. DPP4, dipeptidyl peptidase 4; RBD, receptor-binding domain; RBM, receptor-binding motif; S, spike protein. The function and structure of the S-protein RBD demonstrate that it is an important target for development of vaccines and therapeutic agents against MERS-CoV. A number of MERS vaccines have been developed based on viral RBD, including nanoparticles, virus-like particles (VLPs), and recombinant proteins, and their protective efficacy has been evaluated in animal models, including mice with adenovirus 5 (Ad5)-directed expression of human DPP4 (Ad5/hDPP4), hDPP4-transgenic (hDPP4-Tg) mice, and non-human primates (NHPs) [88] [89] [90] [91] [92] [93] [94] . Features of these RBD-based vaccines, in terms of functionality, antigenicity, immunogenicity, and protective ability, are shown in Table 1 . The function and structure of the S-protein RBD demonstrate that it is an important target for development of vaccines and therapeutic agents against MERS-CoV. A number of MERS vaccines have been developed based on viral RBD, including nanoparticles, virus-like particles (VLPs), and recombinant proteins, and their protective efficacy has been evaluated in animal models, including mice with adenovirus 5 (Ad5)-directed expression of human DPP4 (Ad5/hDPP4), hDPP4-transgenic (hDPP4-Tg) mice, and non-human primates (NHPs) [88] [89] [90] [91] [92] [93] [94] . Features of these RBD-based vaccines, in terms of functionality, antigenicity, immunogenicity, and protective ability, are shown in Table 1 . A soluble nanoparticle vaccine formed in Escherichia coli by the RNA-mediated folding of a RBD-ferritin (FR) hybrid elicits robust RBD-specific antibody and cellular immune responses in mice, producing antisera that effectively block the binding of RBD to hDPP4 in vitro [89] . The adjuvants alum and the squalene-based MF59 significantly augment the antibody titers and T-cell responses induced by RBD-FR nanoparticle vaccines engineered with or without a SSG linker [89] . Similarly, a chimeric, spherical VLP (sVLP) vaccine expressing MERS-CoV RBD induces specific antibody and cellular immune responses in mice, preventing pseudotyped MERS-CoV entry into susceptible cells [90] . The protective efficacy of these two types of MERS vaccine does not yet seem to have been investigated in a viral-challenge animal model. Recombinant vaccines involving RBD subunits have been extensively studied for protection against MERS-CoV infection in MERS-CoV-susceptible animal models [93, [95] [96] [97] 100, 101] . A recombinant RBD (rRBD) fragment (residues 367-606) expressed in insect cells elicits an antibody response and the production of neutralizing antibodies in mice and NHPs [88, 91] . It gives incomplete protection in MERS-CoV-challenged NHPs, with the alleviation of pneumonia and clinical manifestations, as well as the reduction of viral load in lung, trachea, and oropharyngeal swabs [91] . A MERS-CoV S-protein RBD fragment containing residues 377-588 has been identified as a critical neutralizing domain [95] . A treatment regimen involving two doses of a fusion of this fragment and the Fc region of human IgG (S377-588-Fc) four weeks apart is able to induce strong, long-term antibody responses (including production of neutralizing antibodies) in mice [98] . These responses are significantly greater than those with a single dose or two doses at intervals of one, two, or three weeks [98] . rRBDs with single or multiple mutations corresponding to S-protein sequences of MERS-CoV strains isolated from humans or camels from 2012 to 2015 have also been studied [100] . All these rRBDs bind RBD-specific neutralizing monoclonal antibodies (mAbs) and DPP4, and are highly immunogenic, eliciting the production of S1-specific antibodies in mice, which cross-neutralizes multiple MERS pseudoviruses and live MERS-CoV [100] . A trimeric RBD-Fd protein formed by fusing a MERS-CoV RBD fragment (residues 377-588) to the foldon trimerization motif, binds strongly to DPP4, and elicits robust and long-term responses with the production of MERS-CoV S1-specific antibodies and neutralizing antibodies in mice, and protects hDPP4-Tg mice against MERS-CoV infection [94] . The protection provided by existing subunit vaccines based on wild-type MERS-CoV RBD is not complete, with survival rates in hDPP4-Tg mice after a MERS-CoV challenge of~67% for S377-588-Fc and 83% for RBD-Fd [94, 98] . However, a variant RBD (T579N) vaccine produced by masking a non-neutralizing epitope at residue 579 with a glycan probe has both functionality in binding DPP4, and antigenicity in binding four potent MERS-CoV RBD-specific neutralizing mAbs (hHS-1, m336, m337, and m338) [93] . The T579N vaccine has significantly greater efficacy than the wild-type RBD vaccine, and it fully protects against a lethal MERS-CoV challenge in immunized hDPP4-Tg mice [93] , demonstrating the possibility of developing RBD-based MERS-CoV vaccines with high efficacy. [102] [103] [104] [105] [106] [107] [108] [109] . These antibodies generally have greater neutralizing activity against MERS-CoV infection than non-RBD S1-based or S2-based antibodies [58, 103, 110, 111] . The prophylactic and therapeutic efficacies of RBD-targeting antibodies have been tested in Ad5/hDPP4 mice, hDPP4-Tg mice, and NHPs [102, 104, [112] [113] [114] . In an earlier review, we described the antiviral mechanisms, in vivo protection, and crystal structures of previously reported MERS-CoV RBD-specific mAbs, including mouse mAbs Mersmab1, 2E6, 4C2, F11, and D12, and human mAbs LCA60, MERS-4, MERS-27, REGN3048, REGN3051, 1E9, 1F8, 3A1, 3B11, 3B12, 3B11-N, 3C12, M14D3, m336, m337, m338, hMS-1, and 4C2h [58] . In this review, we focus on newly reported antibodies targeting MERS-CoV S-protein RBD, or on newly identified features of existing mAbs that were not described previously (Table 2 ) [102, [112] [113] [114] [115] . RBD-targeting human mAbs have been extensively reported. Most of these mAbs can neutralize pseudotyped or live MERS-CoV in vitro, and some have shown protection against MERS-CoV infection in animal models in vivo [102, [112] [113] [114] [115] . The structures of several of these mAbs with their antigen-binding fragments (Fabs) or single-chain variable fragments (scFvs) complexed with RBD are known ( Figure 4) [102, [112] [113] [114] [115] . Binding of these mAbs to RBD involves two major recognition modes, with binding to RBD residues contacted by or overlapping with DPP4 (as is the case for GD-27, MCA1, and CDC2-C2), or with binding to the RBD residues outside of the DPP4-binding interface (as seen with MERS-4) ( Table 2) . infection in animal models in vivo [102, [112] [113] [114] [115] . The structures of several of these mAbs with their antigen-binding fragments (Fabs) or single-chain variable fragments (scFvs) complexed with RBD are known ( Figure 4) [102, [112] [113] [114] [115] . Binding of these mAbs to RBD involves two major recognition modes, with binding to RBD residues contacted by or overlapping with DPP4 (as is the case for GD-27, MCA1, and CDC2-C2), or with binding to the RBD residues outside of the DPP4-binding interface (as seen with MERS-4) ( Table 2) . The human mAbs MERS-GD27 and MERS-GD33 each recognize distinct regions of the RBD [113] . These mAbs have a synergistic effect in the neutralization of pseudotyped MERS-CoV in vitro, with a much lower half-maximal inhibitory concentration (IC50) for their use in combination than separately [113] . An analysis of crystal structures has indicated that MERS-GD27 binds RBD at the DPP4-binding site, and that the neutralization and recognition epitopes almost completely overlap this site, as seen previously for MERS-CoV RBD-targeting neutralizing mAbs, such as m336 [106, 113] . The MERS-GD27 mAb protects hDPP4-Tg mice from MERS-CoV challenge, both preventively and therapeutically, with significantly lower lung virus titers and RNA copy numbers at day 5 postchallenge, and higher survival rates (60% for pre-challenge vaccination and 40% for post-challenge vaccination) relative to control mice treated with an irrelevant mAb [112] . The human mAb MCA1 was isolated from a MERS survivor via the construction of a phagedisplay antibody library from peripheral B cells [114] . Crystal structure analysis indicates that MCA1 binds MERS-CoV S-protein RBD at residues involved in receptor binding, thus interfering with RBD binding to hDPP4 ( Figure 4A ) [114] . This mAb prophylactically and therapeutically inhibits MERS-CoV replication in common marmosets, resulting in significantly improved outcomes and reduced The human mAbs MERS-GD27 and MERS-GD33 each recognize distinct regions of the RBD [113] . These mAbs have a synergistic effect in the neutralization of pseudotyped MERS-CoV in vitro, with a much lower half-maximal inhibitory concentration (IC 50 ) for their use in combination than separately [113] . An analysis of crystal structures has indicated that MERS-GD27 binds RBD at the DPP4-binding site, and that the neutralization and recognition epitopes almost completely overlap this site, as seen previously for MERS-CoV RBD-targeting neutralizing mAbs, such as m336 [106, 113] . The MERS-GD27 mAb protects hDPP4-Tg mice from MERS-CoV challenge, both preventively and therapeutically, with significantly lower lung virus titers and RNA copy numbers at day 5 post-challenge, and higher survival rates (60% for pre-challenge vaccination and 40% for post-challenge vaccination) relative to control mice treated with an irrelevant mAb [112] . The human mAb MCA1 was isolated from a MERS survivor via the construction of a phage-display antibody library from peripheral B cells [114] . Crystal structure analysis indicates that MCA1 binds MERS-CoV S-protein RBD at residues involved in receptor binding, thus interfering with RBD binding to hDPP4 ( Figure 4A ) [114] . This mAb prophylactically and therapeutically inhibits MERS-CoV replication in common marmosets, resulting in significantly improved outcomes and reduced lung disease, compared with unvaccinated controls, and undetectable virus titers 3 days post-challenge [114] . A probe-based single-B-cell cloning strategy has been used for the isolation of CDC2-C2 and CDC2-C5 mAbs from a patient convalescing from MERS, as well as for the isolation of JC57-11 and JC57-14 mAbs from NHPs immunized with MERS-CoV full-length S DNA and protein [102] . All these antibodies have neutralizing activities against both pseudotyped and live MERS-CoV. Among them, CDC2-C2 is the most potent against 10 pseudotyped MERS-CoV strains, with neutralization IC 50 values ranging from 0.002 µg/mL to 0.011 µg/mL [102] . Crystal-structure analysis of the CDC2-C2 and JC57-14 Fab-RBD complexes indicates that both mAbs bind RBD in the "out" (exposed) position, with the CDC2-C2 RBD binding overlapping with the DPP4-contacting residues ( Figure 4B ,C) [102] . In addition, CDC2-C2 prophylactically protects hDPP4-Tg mice from MERS-CoV infection, resulting in no detectable viral replication in the lungs three days post-challenge, and no fatalities over 28 days of observation [102] . The human mAb MERS-4 also neutralizes pseudotyped MERS-CoV and, notably, displays synergistic neutralization in combination with the MERS-CoV S-protein RBD-targeting MERS-27 and m336 mAbs [106, 118] , as well as the S-protein NTD-targeting 5F9 mAb, in each case with dramatic reduction of the IC 50 compared with individual mAbs [115] . Structural analysis of a MERS-4-Fab-RBD complex revealed that MERS-4 binds the RBD from outside the DPP4-binding interface, rather than competing with DPP4 ( Figure 4D ). Unlike MERS-27, which binds RBD regardless of its conformational state within the S trimer, MERS-4 binds RBD in the "standing" position where its epitopes are readily exposed and accessible [115] . Thus, MERS-4 displays unique epitope specificity, and an unusual mechanism of action involving indirect interference with DPP4 binding through conformational changes, which may explain the observation of synergistic neutralization in combination with other mAbs [115] . Single-domain antibody fragments (VHHs), or nanobodies, are the antigen-recognition regions of camelid heavy-chain-only antibodies (HcAbs), which do not contain light chains. VHHs are easily expressed with high yield, and they have intrinsic stability, strong binding affinity, and specificity to target antigens, and they have therefore been developed as important therapeutic tools against viral infection, including that of MERS-CoV [116, 117, [119] [120] [121] [122] [123] . Four VHHs (VHH-1, VHH-4, VHH-83, and VHH-101) have been identified from bone marrow cells of dromedary camels immunized with modified vaccinia virus (MVA) expressing MERS-CoV S protein, and challenged with MERS-CoV [116] . These VHHs bind MERS-CoV S protein with low K d values (0.1-1 nM), recognize an epitope at residue D539 of RBD, and neutralize MERS-CoV (PRNT 50 , 0.0014-0.012 µg/mL) [116] . These four monomeric VHHs have each been fused with a C-terminal human IgG2 tag to generate four HCAbs (HCAb-1, HCAb-4, HCAb-83, and HCAb-101), with a higher binding affinity and a longer half-life than the free VHHs [116] . Studies of protective efficacy show that hDPP4-Tg mice (K18) injected with monomeric VHH-83 (20 or 200 µg per mouse) lose weight, and die within seven days post-infection, possibly because of the short half-life of the VHH. However, when the mice are injected with HCAb-83 (200 µg per mouse), which has an extended half-life (~4.5 days), protection against MERS-CoV is complete, with no viral titers or pathological changes in the lungs of virally challenged mice [116] . By immunizing llamas with a recombinant RBD fragment (residues 377-588) fused to a C-terminal human IgG Fc tag (S377-588-Fc), we constructed a VHH library, and we used it to generate a monomeric VHH, NbMS10, and a human Fc-fused VHH, NbMS10-Fc [117] . Both VHHs can be expressed in a yeast expression system to high purity, and bind RBD with high affinity, recognizing a conformational epitope (residue 539) at the RBD-DPP4 interface, and blocking the binding of RBD to DPP4. These VHHs, particularly NbMS10-Fc, potently cross-neutralize pseudotyped MERS-CoV strains isolated from different countries, hosts, and time periods [117] . Importantly, the Fc-fused NbMS10-Fc significantly improves the serum half-life of NbMS10, and a single-dose treatment of hDPP4-Tg mice with this agent completely protects them against lethal MERS-CoV challenge [117] . These single-domain VHHs demonstrate the feasibility of developing cost-effective, potent, and broad-spectrum therapeutic antibodies against MERS-CoV infection. Compared with vaccines based on MERS-CoV full-length S protein, which have the potential to attenuate neutralizing activity or enhance immune pathology, vaccines developed from MERS-CoV S-protein RBD are safer, and they do not cause immunological toxicity or eosinophilic immune enhancement [55, 99, 110, 124] . Moreover, RBD-based therapeutic antibodies are generally more potent than non-RBD S1-based or S2-based antibodies [58, 104, 111] . Hence, RBD-based vaccines and therapeutic antibodies have the potential for further development as effective tools to prevent and treat MERS-CoV infection. Despite their acknowledged advantages, there are some issues associated with RBD-based interventions that need to be addressed. For example, RBD is under a high level of pressure of positive selection, and mutations occur in the RBD-DPP4 binding interface that might reduce the efficacy of these treatments [100, [125] [126] [127] . One possible way to avoid this effect, and to delay the emergence of escape mutants is to combine RBD-targeting therapeutics with those targeting other regions of the S protein, or to combine antibodies recognizing distinct epitopes within the RBD [102, 128] . Such combinatorial strategies could also dramatically reduce antibody neutralization doses, providing feasible means to combat the continual threat of MERS-CoV. Some recent advances have been made in the structure-guided design of anti-MERS-CoV interventions. Structurally designed inhibitors of the 3CL protease have demonstrated potency against MERS-CoV [129] . Also, a structurally designed S-protein trimer in the optimal prefusion conformation is shown to elicit production of high titers of anti-MERS-CoV neutralizing antibodies [87] . Indeed, based on the previous studies on the structural design of MERS-CoV RBD, non-neutralizing epitopes in the RBD can be masked, to refocus the immunogenicity of the RBD on the neutralizing epitopes, and thus to enhance its ability to confer immune protection [93] . Results from these structure-based studies will help to inform the design of innovative RBD-based anti-MERS-CoV vaccines and therapeutics with improved efficacy. Isolation of a novel coronavirus from a man with pneumonia in Saudi Arabia Fatality risks for nosocomial outbreaks of Middle East respiratory syndrome coronavirus in the Middle East and South Korea Risks of death and severe disease in patients with Middle East respiratory syndrome coronavirus Impact of comorbidity on fatality rate of patients with Middle East respiratory syndrome Clinical determinants of the severity of Middle East respiratory syndrome (MERS): A systematic review and meta-analysis Prevalence of comorbidities in the Middle East respiratory syndrome coronavirus (MERS-CoV): A systematic review and meta-analysis Epidemiological, demographic, and clinical characteristics of 47 cases of Middle East respiratory syndrome coronavirus disease from Saudi Arabia: A descriptive study Sex matters -a preliminary analysis of Middle East respiratory syndrome in the Republic of Korea Middle East respiratory syndrome coronavirus (MERS-CoV) outbreak in South Korea, 2015: Epidemiology, characteristics and public health implications Hospital outbreak of Middle East respiratory syndrome coronavirus Middle East respiratory syndrome coronavirus (MERS-CoV) infection during pregnancy: Report of two cases & review of the literature MERS-CoV infection in a pregnant woman in Korea Middle East respiratory syndrome coronavirus in pediatrics: A report of seven cases from Saudi Arabia Middle East respiratory syndrome coronavirus infection during pregnancy: A Report of 5 cases from Saudi Arabia An outbreak of Middle East respiratory syndrome coronavirus infection in South Korea Probable transmission chains of Middle East respiratory syndrome coronavirus and the multiple generations of secondary infection in South Korea Further evidence for bats as the evolutionary source of Middle East respiratory syndrome coronavirus Bat origins of MERS-CoV supported by bat coronavirus HKU4 usage of human receptor CD26 Receptor usage and cell entry of bat coronavirus HKU4 provide insight into bat-to-human transmission of MERS coronavirus Replication and shedding of MERS-CoV in Jamaican fruit bats (Artibeus jamaicensis) Discovery of novel bat coronaviruses in south China that use the same receptor as Middle East respiratory syndrome coronavirus Rapid detection of MERS coronavirus-like viruses in bats: Pote1ntial for tracking MERS coronavirus transmission and animal origin Receptor usage of a novel bat lineage C Betacoronavirus reveals evolution of Middle East respiratory syndrome-related coronavirus spike proteins for human dipeptidyl peptidase 4 binding MERS-CoV spillover at the camel-human interface Prevalence of Middle East respiratory syndrome coronavirus (MERS-CoV) in dromedary camels in Abu Dhabi Emirate Middle East respiratory syndrome coronavirus in dromedary camels: An outbreak investigation High prevalence of MERS-CoV infection in camel workers in Saudi Arabia The prevalence of Middle East respiratory syndrome coronavirus (MERS-CoV) antibodies in dromedary camels in Israel Serologic evidence for MERS-CoV infection in dromedary camels Sero-prevalence of Middle East respiratory syndrome coronavirus (MERS-CoV) specific antibodies in dromedary camels in Tabuk, Saudi Arabia Dromedary camels in northern Mali have high seropositivity to MERS-CoV Cross-sectional surveillance of Middle East respiratory syndrome coronavirus (MERS-CoV) in dromedary camels and other mammals in Egypt Serological evidence of MERS-CoV antibodies in dromedary camels (Camelus dromedaries) in Laikipia county Reported direct and indirect contact with dromedary camels among laboratory-confirmed MERS-CoV cases Middle East respiratory syndrome coronavirus: Risk factors and determinants of primary, household, and nosocomial transmission Unusual presentation of Middle East respiratory syndrome coronavirus leading to a large outbreak in Riyadh during 2017 Outbreaks of Middle East respiratory syndrome in two hospitals initiated by a single patient in Daejeon MERS-CoV outbreak following a single patient exposure in an emergency room in South Korea: An epidemiological outbreak study Outbreak of Middle East respiratory syndrome at tertiary care hospital Transmission of Middle East respiratory syndrome coronavirus infections in healthcare settings Hospital-associated Middle East respiratory syndrome coronavirus infections Hospital-associated Middle East respiratory syndrome coronavirus infections Family cluster of Middle East respiratory syndrome coronavirus infections Healthcare-associated infections: The hallmark of Middle East respiratory syndrome coronavirus with review of the literature Clinical features and viral diagnosis of two cases of infection with Middle East respiratory syndrome coronavirus: A report of nosocomial transmission Clinical course and outcomes of critically ill patients with Middle East respiratory syndrome coronavirus infection Human intestinal tract serves as an alternative infection route for Middle East respiratory syndrome coronavirus Persistence of antibodies against Middle East respiratory syndrome coronavirus Presence of Middle East respiratory syndrome coronavirus antibodies in Saudi Arabia: A nationwide, cross-sectional, serological study Feasibility of using convalescent plasma immunotherapy for MERS-CoV infection, Saudi Arabia Feasibility, safety, clinical, and laboratory effects of convalescent plasma therapy for patients with Middle East respiratory syndrome coronavirus infection: A study protocol Challenges of convalescent plasma infusion therapy in Middle East respiratory coronavirus infection: A single centre experience Safety and tolerability of a novel, polyclonal human anti-MERS coronavirus antibody produced from transchromosomic cattle: A phase 1 randomised, double-blind, single-dose-escalation study Prospects for a MERS-CoV spike vaccine Current advancements and potential strategies in the development of MERS-CoV vaccines Is the discovery of the novel human betacoronavirus 2c EMC/2012 (HCoV-EMC) the beginning of another SARS-like pandemic? CoV spike protein: A key target for antivirals Genomic characterization of a newly discovered coronavirus associated with acute respiratory distress syndrome in humans Engineering a replication-competent, propagation-defective Middle East respiratory syndrome coronavirus as a vaccine candidate Reverse genetics with a full-length infectious cDNA of the Middle East respiratory syndrome coronavirus The endonucleolytic RNA cleavage function of nsp1 of Middle East respiratory syndrome coronavirus promotes the production of infectious virus particles in specific human cell lines MERS coronavirus nsp1 participates in an efficient propagation through a specific interaction with viral RNA Middle East respiratory syndrome coronavirus nonstructural protein 16 Is necessary for interferon resistance and viral pathogenesis Structural and biochemical characterization of endoribonuclease Nsp15 encoded by Middle East respiratory syndrome coronavirus Structural insights into the Middle East respiratory syndrome coronavirus 4a protein and its dsRNA binding mechanism Middle East respiratory coronavirus accessory protein 4a inhibits PKR-mediated antiviral stress responses Inhibition of stress granule formation by Middle East respiratory syndrome coronavirus 4a accessory protein facilitates viral translation, leading to efficient virus replication Sola, I. MERS-CoV 4b protein interferes with the NF-kappaB-dependent innate immune response during infection Proteolytic processing of Middle East respiratory syndrome coronavirus spikes expands virus tropism Host cell entry of Middle East respiratory syndrome coronavirus after two-step, furin-mediated activation of the spike protein Structure, function, and evolution of coronavirus spike proteins MERS-CoV spike protein: Targets for vaccines and therapeutics Dipeptidyl peptidase 4 is a functional receptor for the emerging human coronavirus-EMC Structure of the fusion core and inhibition of fusion by a heptad repeat peptide derived from the S protein of Middle East respiratory syndrome coronavirus Structure-based discovery of Middle East respiratory syndrome coronavirus fusion inhibitor Crystal structure of the receptor-binding domain from newly emerged Middle East respiratory syndrome coronavirus Middle East respiratory syndrome coronavirus spike protein is not activated directly by cellular furin during viral entry into target cells Receptor variation and susceptibility to Middle East respiratory syndrome coronavirus infection Host species restriction of Middle East respiratory syndrome coronavirus through its receptor, dipeptidyl peptidase 4 Adaptive evolution of MERS-CoV to species variation in DPP4 Middle East respiratory syndrome coronavirus and bat coronavirus HKU9 both can utilize GRP78 for attachment onto host cells Identification of sialic acid-binding function for the Middle East respiratory syndrome coronavirus spike glycoprotein Structure of MERS-CoV spike receptor-binding domain complexed with human receptor DPP4 Molecular basis of binding between novel human coronavirus MERS-CoV and its receptor CD26 Cryo-EM structures of MERS-CoV and SARS-CoV spike glycoproteins reveal the dynamic receptor binding domains Immunogenicity and structures of a rationally designed prefusion MERS-CoV spike antigen Tailoring subunit vaccine immunity with adjuvant combinations and delivery routes using the Middle East respiratory coronavirus (MERS-CoV) receptor-binding domain as an antigen Chaperna-mediated assembly of ferritin-based Middle East respiratory syndrome-coronavirus nanoparticles Novel chimeric virus-like particles vaccine displaying MERS-CoV receptor-binding domain induce specific humoral and cellular immune response in mice Recombinant receptor binding domain protein induces partial protective immunity in Rhesus Macaques against Middle East respiratory syndrome coronavirus challenge Identification of an ideal adjuvant for receptor-binding domain-based subunit vaccines against Middle East respiratory syndrome coronavirus Introduction of neutralizing immunogenicity index to the rational design of MERS coronavirus subunit vaccines A recombinant receptor-binding domain of MERS-CoV in trimeric form protects human dipeptidyl peptidase 4 (hDPP4) transgenic mice from MERS-CoV infection Searching for an ideal vaccine candidate among different MERS coronavirus receptor-binding fragments-the importance of immunofocusing in subunit vaccine design Receptor-binding domain-based subunit vaccines against MERS-CoV Optimization of antigen dose for a receptor-binding domain-based subunit vaccine against MERS coronavirus Receptor-binding domain of MERS-CoV with optimal immunogen dosage and immunization interval protects human transgenic mice from MERS-CoV infection Engineering a stable CHO cell line for the expression of a MERS-coronavirus vaccine antigen Recombinant receptor-binding domains of multiple Middle East respiratory syndrome coronaviruses (MERS-CoVs) induce cross-neutralizing antibodies against divergent human and camel MERS-CoVs and antibody escape mutants 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 Importance of neutralizing monoclonal antibodies targeting multiple antigenic sites on MERS-CoV Spike to avoid neutralization escape A humanized neutralizing antibody against MERS-CoV targeting the receptor-binding domain of the spike protein Prophylactic and postexposure efficacy of a potent human monoclonal antibody against MERS coronavirus Pre-and postexposure efficacy of fully human antibodies against Spike protein in a novel humanized mouse model of MERS-CoV infection Junctional and allele-specific residues are critical for MERS-CoV neutralization by an exceptionally potent germline-like antibody Single-dose treatment with a humanized neutralizing antibody affords full protection of a human transgenic mouse model from lethal Middle East respiratory syndrome (MERS)-coronavirus infection A conformation-dependent neutralizing monoclonal antibody specifically targeting receptor-binding domain in Middle East respiratory syndrome coronavirus spike protein Exceptionally potent neutralization of middle East respiratory syndrome coronavirus by human monoclonal antibodies Middle East respiratory syndrome: Current status and future prospects for vaccine development Evaluation of candidate vaccine approaches for MERS-CoV A novel human mAb (MERS-GD27) provides prophylactic and postexposure efficacy in MERS-CoV susceptible mice Ultrapotent human neutralizing antibody repertoires against Middle East respiratory syndrome coronavirus from a recovered patient Human neutralizing monoclonal antibody inhibition of Middle East respiratory syndrome coronavirus replication in the common marmoset Structural definition of a unique neutralization epitope on the receptor-binding domain of MERS-CoV spike glycoprotein Chimeric camel/human heavy-chain antibodies protect against MERS-CoV infection A novel nanobody targeting Middle East respiratory syndrome coronavirus (MERS-CoV) receptor-binding domain has potent cross-neutralizing activity and protective efficacy against MERS-CoV Structural basis for the neutralization of MERS-CoV by a human monoclonal antibody MERS-27 Application of camelid heavy-chain variable domains (VHHs) in prevention and treatment of bacterial and viral infections Nanobodies(R) as inhaled biotherapeutics for lung diseases Generation and characterization of ALX-0171, a potent novel therapeutic nanobody for the treatment of respiratory syncytial virus infection Nanobodies as therapeutics: Big opportunities for small antibodies Nanobodies: Natural single-domain antibodies Vaccines for the prevention against the threat of MERS-CoV Evolutionary dynamics of MERS-CoV: Potential recombination, positive selection and transmission Spread of mutant Middle East respiratory syndrome coronavirus with reduced affinity to human CD26 during the South Korean outbreak Mutations in the spike protein of Middle East respiratory syndrome coronavirus transmitted in Korea increase resistance to antibody-mediated neutralization Combining a fusion inhibitory peptide targeting the MERS-CoV S2 protein HR1 domain and a neutralizing antibody specific for the S1 protein receptor-binding domain (RBD) showed potent synergism against pseudotyped MERS-CoV with or without mutations in RBD Structure-guided design of potent and permeable inhibitors of MERS coronavirus 3CL protease that utilize a piperidine moiety as a novel design element This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license Acknowledgments: This study was supported by the NSFC grant 81571983, and the NIH grants R21AI128311, R01AI137472, and R01AI139092. The funders had no role in study design, data collection and interpretation, or the decision to submit the work for publication. The authors declare no competing interests.