SARS and MERS: recent insights into emerging coronaviruses

Emmie de Wit, Neeltje van Doremalen, Darryl Falzarano, Vincent J Munster, Emmie de Wit, Neeltje van Doremalen, Darryl Falzarano, Vincent J Munster

Abstract

The emergence of Middle East respiratory syndrome coronavirus (MERS-CoV) in 2012 marked the second introduction of a highly pathogenic coronavirus into the human population in the twenty-first century. The continuing introductions of MERS-CoV from dromedary camels, the subsequent travel-related viral spread, the unprecedented nosocomial outbreaks and the high case-fatality rates highlight the need for prophylactic and therapeutic measures. Scientific advancements since the 2002-2003 severe acute respiratory syndrome coronavirus (SARS-CoV) pandemic allowed for rapid progress in our understanding of the epidemiology and pathogenesis of MERS-CoV and the development of therapeutics. In this Review, we detail our present understanding of the transmission and pathogenesis of SARS-CoV and MERS-CoV, and discuss the current state of development of measures to combat emerging coronaviruses.

Conflict of interest statement

The authors declare no competing financial interests.

Figures

Figure 1. SARS-CoV and MERS-CoV structure and…
Figure 1. SARS-CoV and MERS-CoV structure and replication.
a | The single-stranded RNA (ssRNA) genomes of severe acute respiratory syndrome coronavirus (SARS-CoV) and Middle East respiratory syndrome coronavirus (MERS-CoV) encode two large polyproteins, pp1a and pp1ab, which are proteolytically cleaved into 16 non-structural proteins (nsps), including papain-like protease (PLpro), 3C-like protease (3CLpro), RNA-dependent RNA polymerase (RdRp), helicase (Hel) and exonuclease (ExoN). An additional 9–12 ORFs are encoded through the transcription of a nested set of subgenomic RNAs. SARS-CoV and MERS-CoV form spherical particles that consist of four structural proteins. The envelope glycoprotein spike (S) forms a layer of glycoproteins that protrude from the envelope. Two additional transmembrane glycoproteins are incorporated in the virion: envelope (E) and membrane (M). Inside the viral envelope resides the helical nucleocapsid, which consists of the viral positive-sense RNA ((+)RNA) genome encapsidated by protein nucleocapsid (N). b | Following entry of the virus into the host cell, the viral RNA is uncoated in the cytoplasm. ORF1a and ORF1ab are translated to produce pp1a and pp1ab, which are cleaved by the proteases that are encoded by ORF1a to yield 16 nsps that form the RNA replicase–transcriptase complex. This complex localizes to modified intracellular membranes that are derived from the rough endoplasmic reticulum (ER) in the perinuclear region, and it drives the production of negative-sense RNAs ((−)RNAs) through both replication and transcription. During replication, full-length (−)RNA copies of the genome are produced and used as templates for full-length (+)RNA genomes. During transcription, a subset of 7–9 subgenomic RNAs, including those encoding all structural proteins, is produced through discontinuous transcription. In this process, subgenomic (−)RNAs are synthesized by combining varying lengths of the 3′ end of the genome with the 5′ leader sequence necessary for translation. These subgenomic (−)RNAs are then transcribed into subgenomic (+)mRNAs. Although the different subgenomic mRNAs may contain several ORFs, only the first ORF (that closest to the 5′ end) is translated. The resulting structural proteins are assembled into the nucleocapsid and viral envelope at the ER–Golgi intermediate compartment (ERGIC), followed by release of the nascent virion from the infected cell. PowerPoint slide
Figure 2. The emergence of SARS-CoV and…
Figure 2. The emergence of SARS-CoV and MERS-CoV.
Bats harbour a wide range of coronaviruses, including severe acute respiratory syndrome coronavirus (SARS-CoV)-like and Middle East respiratory syndrome coronavirus (MERS-CoV)-like viruses. SARS-CoV crossed the species barrier into masked palm civets and other animals in live-animal markets in China; genetic analysis suggests that this occurred in late 2002. Several people in close proximity to palm civets became infected with SARS-CoV. A MERS-CoV ancestral virus crossed the species barrier into dromedary camels; serological evidence suggests that this happened more than 30 years ago. Abundant circulation of MERS-CoV in dromedary camels results in frequent zoonotic transmission of this virus. SARS-CoV and MERS-CoV spread between humans mainly through nosocomial transmission, which results in the infection of health care workers and patients at a higher frequency than infection of their relatives. PowerPoint slide
Figure 3. Evasion of the innate immune…
Figure 3. Evasion of the innate immune response by SARS-CoV and MERS-CoV.
a | The innate immune response is activated by the detection of viral pathogen-associated molecular patterns (PAMPs), such as double-stranded RNA (dsRNA) or uncapped mRNA. This occurs via host pattern recognition receptors (PRRs), such as retinoic acid-inducible gene I protein (RIG-I) and melanoma differentiation-associated protein 5 (MDA5), potentially via dsRNA-binding partners such as IFN-inducible dsRNA-dependent protein kinase activator A (PRKRA). Following PRR-mediated detection of a PAMP, the resulting interaction of PRRs with mitochondrial antiviral-signalling protein (MAVS) activates nuclear factor-κB (NF-κB) through a signalling cascade involving several kinases. Activated NF-κB translocates to the nucleus, where it induces the transcription of pro-inflammatory cytokines. The kinases also phosphorylate (P) IFN-regulatory factor 3 (IRF3) and IRF7, which form homodimers and heterodimers and enter the nucleus to initiate the transcription of type I interferons (type I IFNs). Both severe acute respiratory syndrome coronavirus (SARS-CoV) and Middle East respiratory syndrome coronavirus (MERS-CoV) have developed mechanisms to interfere with these signalling pathways, as shown; these subversion strategies involve both structural proteins (membrane (M) and nucleocapsid (N)) and non-structural proteins (nsp1, nsp3b, nsp4a, nsp4b, nsp5, nsp6 and papain-like protease (PLpro); indicated in the figure by just their nsp numbers and letters). b | Binding of type I IFNs to their dimeric receptor, IFNα/β receptor (IFNAR), activates the Janus kinase (JAK)–signal transducer and activator of transcription (STAT) signalling pathway, in which JAK1 and TYK2 kinases phosphorylate STAT1 and STAT2, which form complexes with IRF9. These complexes move into the nucleus to initiate the transcription of IFN-stimulated genes (ISGs) under the control of promoters that contain an IFN-stimulated response element (ISRE). Collectively, the expression of cytokines, IFNs and ISGs establishes an antiviral innate immune response that limits viral replication in infected and in neighbouring cells. Again, viral proteins have been shown to inhibit these host signalling pathways to evade this immune response. IκBα, NF-κB inhibitor-α. PowerPoint slide

References

    1. Zhong NS, et al. Epidemiology and cause of severe acute respiratory syndrome (SARS) in Guangdong, People's Republic of China, in February, 2003. Lancet. 2003;362:1353–1358.
    1. Lee N, et al. A major outbreak of severe acute respiratory syndrome in Hong Kong. N. Engl. J. Med. 2003;348:1986–1994.
    1. Guan Y, et al. Molecular epidemiology of the novel coronavirus that causes severe acute respiratory syndrome. Lancet. 2004;363:99–104.
    1. Drosten C, et al. Identification of a novel coronavirus in patients with severe acute respiratory syndrome. N. Engl. J. Med. 2003;348:1967–1976.
    1. Ksiazek TG, et al. A novel coronavirus associated with severe acute respiratory syndrome. N. Engl. J. Med. 2003;348:1953–1966.
    1. Peiris JS, et al. Coronavirus as a possible cause of severe acute respiratory syndrome. Lancet. 2003;361:1319–1325.
    1. WHO. Summary of probably SARS cases with onset of illness from 1 November 2002 to 31 July 2003. WHO, (2004).
    1. Wang M, et al. SARS-CoV infection in a restaurant from palm civet. Emerg. Infect. Dis. 2005;11:1860–1865.
    1. Ge XY, et al. Isolation and characterization of a bat SARS-like coronavirus that uses the ACE2 receptor. Nature. 2013;503:535–538.
    1. Menachery VD, et al. A SARS-like cluster of circulating bat coronaviruses shows potential for human emergence. Nat. Med. 2015;21:1508–1513.
    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. Hijawi B, et al. Novel coronavirus infections in Jordan, April 2012: epidemiological findings from a retrospective investigation. East. Mediterr. Health J. 2013;19(Suppl. 1):S12–S18.
    1. Wise J. Patient with new strain of coronavirus is treated in intensive care at London hospital. BMJ. 2012;345:e6455.
    1. Korea Centers for Disease Control and Prevention. Middle East respiratory syndrome coronavirus outbreak in the Republic of Korea, 2015. Osong Public Health Res. Perspect.6, 269–278 (2015).
    1. WHO. Coronavirus infections: disease outbreak news. WHO, (2016).
    1. Pasternak AO, Spaan WJ, Snijder EJ. Nidovirus transcription: how to make sense...? J. Gen. Virol. 2006;87:1403–1421.
    1. Perlman S, Netland J. Coronaviruses post-SARS: update on replication and pathogenesis. Nat. Rev. Microbiol. 2009;7:439–450.
    1. Fehr AR, Perlman S. Coronaviruses: an overview of their replication and pathogenesis. Methods Mol. Biol. 2015;1282:1–23.
    1. Knoops K, et al. SARS-coronavirus replication is supported by a reticulovesicular network of modified endoplasmic reticulum. PLoS Biol. 2008;6:e226.
    1. Snijder EJ, et al. Unique and conserved features of genome and proteome of SARS-coronavirus, an early split-off from the coronavirus group 2 lineage. J. Mol. Biol. 2003;331:991–1004.
    1. Eckerle LD, et al. Infidelity of SARS-CoV nsp14-exonuclease mutant virus replication is revealed by complete genome sequencing. PLoS Pathog. 2010;6:e1000896.
    1. Sevajol M, Subissi L, Decroly E, Canard B, Imbert I. Insights into RNA synthesis, capping, and proofreading mechanisms of SARS-coronavirus. Virus Res. 2014;194:90–99.
    1. Raj VS, et al. Dipeptidyl peptidase 4 is a functional receptor for the emerging human coronavirus-EMC. Nature. 2013;495:251–254.
    1. Masters PS, Perlman S. Fields Virology. 2013. pp. 825–858.
    1. Guan Y, et al. Isolation and characterization of viruses related to the SARS coronavirus from animals in southern China. Science. 2003;302:276–278.
    1. Wang LF, et al. Review of bats and SARS. Emerg. Infect. Dis. 2006;12:1834–1840.
    1. Drexler JF, Corman VM, Drosten C. Ecology, evolution and classification of bat coronaviruses in the aftermath of SARS. Antiviral Res. 2014;101:45–56.
    1. Reusken CB, et al. Middle East respiratory syndrome coronavirus neutralizing serum antibodies in dromedary camels: a comparative serological study. Lancet Infect. Dis. 2013;13:859–866.
    1. Haagmans BL, et al. Middle East respiratory syndrome coronavirus in dromedary camels: an outbreak investigation. Lancet Infect. Dis. 2014;14:140–145.
    1. Azhar EI, et al. Evidence for camel-to-human transmission of MERS coronavirus. N. Engl. J. Med. 2014;370:2499–2505.
    1. Hemida MG, et al. MERS coronavirus in dromedary camel herd, Saudi Arabia. Emerg. Infect. Dis. 2014;20:1231–1234.
    1. Raj VS, et al. Isolation of MERS coronavirus from a dromedary camel, Qatar, 2014. Emerg. Infect. Dis. 2014;20:1339–1342.
    1. Muller MA, et al. MERS coronavirus neutralizing antibodies in camels, Eastern Africa, 1983–1997. Emerg. Infect. Dis. 2014;20:2093–2095.
    1. Sabir JS, et al. Co-circulation of three camel coronavirus species and recombination of MERS-CoVs in Saudi Arabia. Science. 2016;351:81–84.
    1. Chowell G, et al. Transmission characteristics of MERS and SARS in the healthcare setting: a comparative study. BMC Med. 2015;13:210.
    1. Hunter JC, et al. Transmission of Middle East respiratory syndrome coronavirus infections in healthcare settings, Abu Dhabi. Emerg. Infect. Dis. 2016;22:647–656.
    1. Anderson RM, et al. Epidemiology, transmission dynamics and control of SARS: the 2002–2003 epidemic. Philos. Trans. R. Soc. Lond. B. Biol. Sci. 2004;359:1091–1105.
    1. Cowling BJ, et al. Preliminary epidemiological assessment of MERS-CoV outbreak in South Korea, May to June 2015. Euro Surveill. 2015;20:7–13.
    1. Peiris JS, 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. Bin SY, et al. Environmental contamination and viral shedding in MERS patients during MERS-CoV outbreak in South Korea. Clin. Infect. Dis. 2015;62:755–760.
    1. Kucharski AJ, Althaus CL. The role of superspreading in Middle East respiratory syndrome coronavirus (MERS-CoV) transmission. Euro Surveill. 2015;20:14–18.
    1. Oh MD, et al. Middle East respiratory syndrome coronavirus superspreading event involving 81 persons, Korea 2015. J. Korean Med. Sci. 2015;30:1701–1705.
    1. Wong G, et al. MERS, SARS, and Ebola: the role of super-spreaders in infectious disease. Cell Host Microbe. 2015;18:398–401.
    1. Ng DL, 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. Kuba K, et al. A crucial role of angiotensin converting enzyme 2 (ACE2) in SARS coronavirus-induced lung injury. Nat. Med. 2005;11:875–879.
    1. Imai Y, et al. Angiotensin-converting enzyme 2 protects from severe acute lung failure. Nature. 2005;436:112–116.
    1. Wang WK, et al. Temporal relationship of viral load, ribavirin, interleukin (IL)-6, IL-8, and clinical progression in patients with severe acute respiratory syndrome. Clin. Infect. Dis. 2004;39:1071–1075.
    1. Drosten C, et al. Clinical features and virological analysis of a case of Middle East respiratory syndrome coronavirus infection. Lancet Infect. Dis. 2013;13:745–751.
    1. Poissy J, et al. Kinetics and pattern of viral excretion in biological specimens of two MERS-CoV cases. J. Clin. Virol. 2014;61:275–278.
    1. Binnie A, Tsang JL, dos Santos CC. Biomarkers in acute respiratory distress syndrome. Curr. Opin. Crit. Care. 2014;20:47–55.
    1. Williams AE, Chambers RC. The mercurial nature of neutrophils: still an enigma in ARDS? Am. J. Physiol. Lung Cell. Mol. Physiol. 2014;306:L217–L230.
    1. Baas T, Taubenberger JK, Chong PY, Chui P, Katze MG. SARS-CoV virus–host interactions and comparative etiologies of acute respiratory distress syndrome as determined by transcriptional and cytokine profiling of formalin-fixed paraffin-embedded tissues. J. Interferon Cytokine Res. 2006;26:309–317.
    1. Faure E, et al. Distinct immune response in two MERS-CoV-infected patients: can we go from bench to bedside? PLoS ONE. 2014;9:e88716.
    1. Kong SL, Chui P, Lim B, Salto-Tellez M. Elucidating the molecular physiopathology of acute respiratory distress syndrome in severe acute respiratory syndrome patients. Virus Res. 2009;145:260–269.
    1. Tang NL, et al. Early enhanced expression of interferon-inducible protein-10 (CXCL-10) and other chemokines predicts adverse outcome in severe acute respiratory syndrome. Clin. Chem. 2005;51:2333–2340.
    1. Cameron MJ, et al. Interferon-mediated immunopathological events are associated with atypical innate and adaptive immune responses in patients with severe acute respiratory syndrome. J. Virol. 2007;81:8692–8706.
    1. Gralinski LE, et al. Genome wide identification of SARS-CoV susceptibility loci using the Collaborative Cross. PLoS Genet. 2015;11:e1005504.
    1. Jensen S, Thomsen AR. Sensing of RNA viruses: a review of innate immune receptors involved in recognizing RNA virus invasion. J. Virol. 2012;86:2900–2910.
    1. Frieman MB, 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, et al. MyD88 is required for protection from lethal infection with a mouse-adapted SARS-CoV. PLoS Pathog. 2008;4:e1000240.
    1. Zhao J, et al. Rapid generation of a mouse model for Middle East respiratory syndrome. Proc. Natl Acad. Sci. USA. 2014;111:4970–4975.
    1. de Wilde AH, et al. MERS-coronavirus replication induces severe in vitro cytopathology and is strongly inhibited by cyclosporin A or interferon-α treatment. J. Gen. Virol. 2013;94:1749–1760.
    1. Snijder EJ, et al. Ultrastructure and origin of membrane vesicles associated with the severe acute respiratory syndrome coronavirus replication complex. J. Virol. 2006;80:5927–5940.
    1. Bouvet M, et al. In vitro reconstitution of SARS-coronavirus mRNA cap methylation. PLoS Pathog. 2010;6:e1000863.
    1. Menachery VD, et al. Attenuation and restoration of severe acute respiratory syndrome coronavirus mutant lacking 2′-O-methyltransferase activity. J. Virol. 2014;88:4251–4264.
    1. Menachery VD, Debbink K, Baric RS. Coronavirus non-structural protein 16: evasion, attenuation, and possible treatments. Virus Res. 2014;194:191–199.
    1. Cui L, et al. The nucleocapsid protein of coronaviruses acts as a viral suppressor of RNA silencing in mammalian cells. J. Virol. 2015;89:9029–9043.
    1. Lu X, Pan J, Tao J, Guo D. SARS-CoV nucleocapsid protein antagonizes IFN-β response by targeting initial step of IFN-β induction pathway, and its C-terminal region is critical for the antagonism. Virus Genes. 2011;42:37–45.
    1. Niemeyer D, et al. Middle East respiratory syndrome coronavirus accessory protein 4a is a type I interferon antagonist. J. Virol. 2013;87:12489–12495.
    1. Siu KL, et al. Middle east respiratory syndrome coronavirus 4a protein is a double-stranded RNA-binding protein that suppresses PACT-induced activation of RIG-I and MDA5 in the innate antiviral response. J. Virol. 2014;88:4866–4876.
    1. Yang Y, et al. The structural and accessory proteins M, ORF 4a, ORF 4b, and ORF 5 of Middle East respiratory syndrome coronavirus (MERS-CoV) are potent interferon antagonists. Protein Cell. 2013;4:951–961.
    1. Yang Y, et al. Middle East respiratory syndrome coronavirus ORF4b protein inhibits type I interferon production through both cytoplasmic and nuclear targets. Sci. Rep. 2015;5:17554.
    1. Frieman M, Ratia K, Johnston RE, Mesecar AD, Baric RS. Severe acute respiratory syndrome coronavirus papain-like protease ubiquitin-like domain and catalytic domain regulate antagonism of IRF3 and NF-κB signaling. J. Virol. 2009;83:6689–6705.
    1. Devaraj SG, et al. Regulation of IRF-3-dependent innate immunity by the papain-like protease domain of the severe acute respiratory syndrome coronavirus. J. Biol. Chem. 2007;282:32208–32221.
    1. Matthews K, Schafer A, Pham A, Frieman M. The SARS coronavirus papain like protease can inhibit IRF3 at a post activation step that requires deubiquitination activity. Virol. J. 2014;11:209.
    1. Bailey-Elkin BA, et al. Crystal structure of the Middle East respiratory syndrome coronavirus (MERS-CoV) papain-like protease bound to ubiquitin facilitates targeted disruption of deubiquitinating activity to demonstrate its role in innate immune suppression. J. Biol. Chem. 2014;289:34667–34682.
    1. Huang C, et al. SARS coronavirus nsp1 protein induces template-dependent endonucleolytic cleavage of mRNAs: viral mRNAs are resistant to nsp1-induced RNA cleavage. PLoS Pathog. 2011;7:e1002433.
    1. Kamitani W, Huang C, Narayanan K, Lokugamage KG, Makino S. A two-pronged strategy to suppress host protein synthesis by SARS coronavirus nsp1 protein. Nat. Struct. Mol. Biol. 2009;16:1134–1140.
    1. Tanaka T, Kamitani W, DeDiego ML, Enjuanes L, Matsuura Y. Severe acute respiratory syndrome coronavirus nsp1 facilitates efficient propagation in cells through a specific translational shutoff of host mRNA. J. Virol. 2012;86:11128–11137.
    1. Wathelet MG, Orr M, Frieman MB, Baric RS. Severe acute respiratory syndrome coronavirus evades antiviral signaling: role of nsp1 and rational design of an attenuated strain. J. Virol. 2007;81:11620–11633.
    1. Lokugamage KG, et al. Middle East respiratory syndrome coronavirus nsp1 inhibits host gene expression by selectively targeting mRNAs transcribed in the nucleus while sparing mRNAs of cytoplasmic origin. J. Virol. 2015;89:10970–10981.
    1. Freundt EC, Yu L, Park E, Lenardo MJ, Xu XN. Molecular determinants for subcellular localization of the severe acute respiratory syndrome coronavirus open reading frame 3b protein. J. Virol. 2009;83:6631–6640.
    1. Kopecky-Bromberg SA, Martinez-Sobrido L, Frieman M, Baric RA, Palese P. Severe acute respiratory syndrome coronavirus open reading frame (ORF) 3b, ORF 6, and nucleocapsid proteins function as interferon antagonists. J. Virol. 2007;81:548–557.
    1. Lei Y, et al. MAVS-mediated apoptosis and its inhibition by viral proteins. PLoS ONE. 2009;4:e5466.
    1. Menachery VD, et al. Pathogenic influenza viruses and coronaviruses utilize similar and contrasting approaches to control interferon-stimulated gene responses. mBio. 2014;5:e01174-14.
    1. Graci JD, Cameron CE. Mechanisms of action of ribavirin against distinct viruses. Rev. Med. Virol. 2006;16:37–48.
    1. Al-Tawfiq JA, Momattin H, Dib J, Memish ZA. Ribavirin and interferon therapy in patients infected with the Middle East respiratory syndrome coronavirus: an observational study. Int. J. Infect. Dis. 2014;20:42–46.
    1. Ling Y, Qu R, Luo Y. Clinical analysis of the first patient with imported Middle East respiratory syndrome in China. Zhonghua Wei Zhong Bing Ji Jiu Yi Xue. 2015;27:630–634.
    1. Booth CM, et al. Clinical features and short-term outcomes of 144 patients with SARS in the greater Toronto area. JAMA. 2003;289:2801–2809.
    1. Poutanen SM, et al. Identification of severe acute respiratory syndrome in Canada. N. Engl. J. Med. 2003;348:1995–2005.
    1. So LK, et al. Development of a standard treatment protocol for severe acute respiratory syndrome. Lancet. 2003;361:1615–1617.
    1. Tsang KW, et al. A cluster of cases of severe acute respiratory syndrome in Hong Kong. N. Engl. J. Med. 2003;348:1977–1985.
    1. Loutfy MR, et al. Interferon alfacon-1 plus corticosteroids in severe acute respiratory syndrome: a preliminary study. JAMA. 2003;290:3222–3228.
    1. Zhao Z, et al. Description and clinical treatment of an early outbreak of severe acute respiratory syndrome (SARS) in Guangzhou, PR China. J. Med. Microbiol. 2003;52:715–720.
    1. Hsu LY, et al. Severe acute respiratory syndrome (SARS) in Singapore: clinical features of index patient and initial contacts. Emerg. Infect. Dis. 2003;9:713–717.
    1. Smith EC, Blanc H, Surdel MC, Vignuzzi M, Denison MR. Coronaviruses lacking exoribonuclease activity are susceptible to lethal mutagenesis: evidence for proofreading and potential therapeutics. PLoS Pathog. 2013;9:e1003565.
    1. Chan JF, et al. Broad-spectrum antivirals for the emerging Middle East respiratory syndrome coronavirus. J. Infect. 2013;67:606–616.
    1. Falzarano D, et al. Inhibition of novel β coronavirus replication by a combination of interferon-α2b and ribavirin. Sci. Rep. 2013;3:1686.
    1. Hart BJ, et al. Interferon-β and mycophenolic acid are potent inhibitors of Middle East respiratory syndrome coronavirus in cell-based assays. J. Gen. Virol. 2014;95:571–577.
    1. Morgenstern B, Michaelis M, Baer PC, Doerr HW, Cinatl J. Ribavirin and interferon-β synergistically inhibit SARS-associated coronavirus replication in animal and human cell lines. Biochem. Biophys. Res. Commun. 2005;326:905–908.
    1. Falzarano D, et al. Treatment with interferon-α2b and ribavirin improves outcome in MERS-CoV-infected rhesus macaques. Nat. Med. 2013;19:1313–1317.
    1. Omrani AS, et al. Ribavirin and interferon alfa-2a for severe Middle East respiratory syndrome coronavirus infection: a retrospective cohort study. Lancet Infect. Dis. 2014;14:1090–1095.
    1. Shalhoub S, et al. IFN-α2a or IFN-β1a in combination with ribavirin to treat Middle East respiratory syndrome coronavirus pneumonia: a retrospective study. J. Antimicrob. Chemother. 2015;70:2129–2132.
    1. Khalid M, et al. Ribavirin and interferon-α2b as primary and preventive treatment for Middle East respiratory syndrome coronavirus: a preliminary report of two cases. Antivir. Ther. 2015;20:87–91.
    1. Chan JF, et al. Treatment with lopinavir/ritonavir or interferon-β1b improves outcome of MERS-CoV infection in a non-human primate model of common marmoset. J. Infect. Dis. 2015;212:1904–1913.
    1. Chan KS, et al. Treatment of severe acute respiratory syndrome with lopinavir/ritonavir: a multicentre retrospective matched cohort study. Hong Kong Med. J. 2003;9:399–406.
    1. Chu CM, et al. Role of lopinavir/ritonavir in the treatment of SARS: initial virological and clinical findings. Thorax. 2004;59:252–256.
    1. Spanakis N, et al. Virological and serological analysis of a recent Middle East respiratory syndrome coronavirus infection case on a triple combination antiviral regimen. Int. J. Antimicrob. Agents. 2014;44:528–532.
    1. Choi WJ, Lee KN, Kang EJ, Lee H. Middle East respiratory syndrome-coronavirus infection: a case report of serial computed tomographic findings in a young male patient. Korean J. Radiol. 2016;17:166–170.
    1. Kim, U. J., Won, E. J., Kee, S. J., Jung, S. I. & Jang, H. C. Combination therapy with lopinavir/ritonavir, ribavirin and interferon-α for Middle East respiratory syndrome: a case report. Antivir. Ther.10.3851/IMP3002 (2015).
    1. Rhee, J. Y., Hong, G. & Ryu, K. M. Clinical implications of five cases of Middle East respiratory syndrome coronavirus infection in South Korea Outbreak. Jpn J. Infect. Dis.10.7883/yoken.JJID.2015.445 (2016).
    1. Hilgenfeld R. From SARS to MERS: crystallographic studies on coronaviral proteases enable antiviral drug design. FEBS J. 2014;281:4085–4096.
    1. Cheng KW, et al. Thiopurine analogs and mycophenolic acid synergistically inhibit the papain-like protease of Middle East respiratory syndrome coronavirus. Antiviral Res. 2015;115:9–16.
    1. Tomar S, et al. Ligand-induced dimerization of Middle East respiratory syndrome (MERS) coronavirus nsp5 protease (3CLpro): implications for nsp5 regulation and the development of antivirals. J. Biol. Chem. 2015;290:19403–19422.
    1. de Wilde AH, et al. Screening of an FDA-approved compound library identifies four small-molecule inhibitors of Middle East respiratory syndrome coronavirus replication in cell culture. Antimicrob. Agents Chemother. 2014;58:4875–4884.
    1. International Severe Acute Respiratory & Emerging Infection Consortium. Treatment of MERS-CoV: decision support tool. International Severe Acute Respiratory & Emerging Infection Consortium, (updated 29 July 2013).
    1. Mair-Jenkins J, et al. The effectiveness of convalescent plasma and hyperimmune immunoglobulin for the treatment of severe acute respiratory infections of viral etiology: a systematic review and exploratory meta-analysis. J. Infect. Dis. 2015;211:80–90.
    1. Du L, et al. A conformation-dependent neutralizing monoclonal antibody specifically targeting receptor-binding domain in Middle East respiratory syndrome coronavirus spike protein. J. Virol. 2014;88:7045–7053.
    1. Jiang L, et al. Potent neutralization of MERS-CoV by human neutralizing monoclonal antibodies to the viral spike glycoprotein. Sci. Transl Med. 2014;6:234ra59.
    1. Tang XC, et al. Identification of human neutralizing antibodies against MERS-CoV and their role in virus adaptive evolution. Proc. Natl Acad. Sci. USA. 2014;111:E2018–E2026.
    1. Ying T, et al. Exceptionally potent neutralization of Middle East respiratory syndrome coronavirus by human monoclonal antibodies. J. Virol. 2014;88:7796–7805.
    1. Zhao J, et al. Passive immunotherapy with dromedary immune serum in an experimental animal model for Middle East respiratory syndrome coronavirus infection. J. Virol. 2015;89:6117–6120.
    1. Luke T, et al. Human polyclonal immunoglobulin G from transchromosomic bovines inhibits MERS-CoV in vivo. Sci. Transl Med. 2016;8:326ra21.
    1. Li Y, et al. A humanized neutralizing antibody against MERS-CoV targeting the receptor-binding domain of the spike protein. Cell Res. 2015;25:1237–1249.
    1. Corti D, et al. Prophylactic and postexposure efficacy of a potent human monoclonal antibody against MERS coronavirus. Proc. Natl Acad. Sci. USA. 2015;112:10473–10478.
    1. Pascal KE, et al. Pre- and postexposure efficacy of fully human antibodies against Spike protein in a novel humanized mouse model of MERS-CoV infection. Proc. Natl Acad. Sci. USA. 2015;112:8738–8743.
    1. Houser KV, et al. Prophylaxis with a MERS-CoV-specific human monoclonal antibody protects rabbits from MERS-CoV infection. J. Infect. Dis. 2016;213:1557–1561.
    1. Johnson RF, et al. 3B11-N, a monoclonal antibody against MERS-CoV, reduces lung pathology in rhesus monkeys following intratracheal inoculation of MERS-CoV Jordan-n3/2012. Virology. 2016;490:49–58.
    1. Ohnuma K, et al. Inhibition of Middle East respiratory syndrome coronavirus infection by anti-CD26 monoclonal antibody. J. Virol. 2013;87:13892–13899.
    1. Elshabrawy HA, et al. Identification of a broad-spectrum antiviral small molecule against severe acute respiratory syndrome coronavirus and Ebola, Hendra, and Nipah viruses by using a novel high-throughput screening assay. J. Virol. 2014;88:4353–4365.
    1. Glowacka I, et al. Evidence that TMPRSS2 activates the severe acute respiratory syndrome coronavirus spike protein for membrane fusion and reduces viral control by the humoral immune response. J. Virol. 2011;85:4122–4134.
    1. Zhou Y, et al. Protease inhibitors targeting coronavirus and filovirus entry. Antiviral Res. 2015;116:76–84.
    1. Wang R, Xiao H, Guo R, Li Y, Shen B. The role of C5a in acute lung injury induced by highly pathogenic viral infections. Emerg. Microbes Infect. 2015;4:e28.
    1. Josset L, et al. Cell host response to infection with novel human coronavirus EMC predicts potential antivirals and important differences with SARS coronavirus. mBio. 2013;4:e00165–13.
    1. Graham RL, Donaldson EF, Baric RS. A decade after SARS: strategies for controlling emerging coronaviruses. Nat. Rev. Microbiol. 2013;11:836–848.
    1. Roper RL, Rehm KE. SARS vaccines: where are we? Expert Rev. Vaccines. 2009;8:887–898.
    1. Du L, Jiang S. Middle East respiratory syndrome: current status and future prospects for vaccine development. Expert Opin. Biol. Ther. 2015;15:1647–1651.
    1. Wang L, et al. Evaluation of candidate vaccine approaches for MERS-CoV. Nat. Commun. 2015;6:7712.
    1. Lan J, 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. Muthumani K, 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. Mastalerz-Migas A, Bujnowska-Fedak M, Brydak LB. Immune efficacy of first and repeat trivalent influenza vaccine in healthy subjects and hemodialysis patients. Adv. Exp. Med. Biol. 2015;836:47–54.
    1. Muller MA, et al. Presence of Middle East respiratory syndrome coronavirus antibodies in Saudi Arabia: a nationwide, cross-sectional, serological study. Lancet Infect. Dis. 2015;15:629.
    1. Haagmans BL, et al. An orthopoxvirus-based vaccine reduces virus excretion after MERS-CoV infection in dromedary camels. Science. 2016;351:77–81.
    1. Assiri A, et al. Epidemiological, demographic, and clinical characteristics of 47 cases of Middle East respiratory syndrome coronavirus disease from Saudi Arabia: a descriptive study. Lancet Infect. Dis. 2013;13:752–761.
    1. Leung GM, et al. The epidemiology of severe acute respiratory syndrome in the 2003 Hong Kong epidemic: an analysis of all 1755 patients. Ann. Intern. Med. 2004;141:662–673.
    1. Zumla A, Hui DS, Perlman S. Middle East respiratory syndrome. Lancet. 2015;386:995–1007.
    1. Al-Abdallat MM, et al. Hospital-associated outbreak of Middle East respiratory syndrome coronavirus: a serologic, epidemiologic, and clinical description. Clin. Infect. Dis. 2014;59:1225–1233.
    1. Saad M, et al. Clinical aspects and outcomes of 70 patients with Middle East respiratory syndrome coronavirus infection: a single-center experience in Saudi Arabia. Int. J. Infect. Dis. 2014;29:301–306.
    1. Memish ZA, et al. Respiratory tract samples, viral load, and genome fraction yield in patients with Middle East respiratory syndrome. J. Infect. Dis. 2014;210:1590–1594.
    1. Feikin DR, et al. Association of higher MERS-CoV virus load with severe disease and death, Saudi Arabia, 2014. Emerg. Infect. Dis. 2015;21:2029–2035.
    1. Majumder MS, Kluberg SA, Mekaru SR, Brownstein JS. Mortality risk factors for Middle East respiratory syndrome outbreak, South Korea, 2015. Emerg. Infect. Dis. 2015;21:2088–2090.
    1. Gretebeck LM, Subbarao K. Animal models for SARS and MERS coronaviruses. Curr. Opin. Virol. 2015;13:123–129.
    1. Munster VJ, et al. Replication and shedding of MERS-CoV in Jamaican fruit bats (Artibeus jamaicensis) Sci. Rep. 2016;6:21878.
    1. Lau SK, et al. Severe acute respiratory syndrome coronavirus-like virus in Chinese horseshoe bats. Proc. Natl Acad. Sci. USA. 2005;102:14040–14045.
    1. Li W, et al. Bats are natural reservoirs of SARS-like coronaviruses. Science. 2005;310:676–679.
    1. Wu D, et al. Civets are equally susceptible to experimental infection by two different severe acute respiratory syndrome coronavirus isolates. J. Virol. 2005;79:2620–2625.
    1. Adney DR, et al. Replication and shedding of MERS-CoV in upper respiratory tract of inoculated dromedary camels. Emerg. Infect. Dis. 2014;20:1999–2005.
    1. Becker MM, et al. Synthetic recombinant bat SARS-like coronavirus is infectious in cultured cells and in mice. Proc. Natl Acad. Sci. USA. 2008;105:19944–19949.
    1. Menachery VD, et al. SARS-like WIV1-CoV poised for human emergence. Proc. Natl Acad. Sci. USA. 2016;113:3048–3053.

Source: PubMed

3
Subscribe