Skip to main content
Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
Respirology. 2018 Feb; 23(2): 130–137.
Published online 2017 Oct 20. doi: 10.1111/resp.13196
PMCID: PMC7169239
PMID: 29052924

MERS, SARS and other coronaviruses as causes of pneumonia

ABSTRACT

Human coronaviruses (HCoVs) have been considered to be relatively harmless respiratory pathogens in the past. However, after the outbreak of the severe acute respiratory syndrome (SARS) and emergence of the Middle East respiratory syndrome (MERS), HCoVs have received worldwide attention as important pathogens in respiratory tract infection. This review focuses on the epidemiology, pathogenesis and clinical characteristics among SARS‐coronaviruses (CoV), MERS‐CoV and other HCoV infections.

Keywords: human coronaviruses, Middle East respiratory syndrome, pneumonia, severe acute respiratory syndrome

Abbreviations

AKI
acute kidney injury
CAP
community‐acquired pneumonia
CK
creatinine kinase
CoV
coronavirus
DPP‐4
dipeptidyl peptidase 4
HCoV
human coronavirus
ICU
intensive care unit
IFN
interferon
ISG
IFN‐stimulated gene
MERS
Middle East respiratory syndrome
MPA
mycophenolic acid
RT‐PCR
reverse transcription polymerase chain reaction
SARS
severe acute respiratory syndrome
WHO
World Health Organization

INTRODUCTION

Coronaviruses (CoVs), a large family of single‐stranded RNA viruses, can infect a wide variety of animals, including humans, causing respiratory, enteric, hepatic and neurological diseases.1 As the largest known RNA viruses, CoVs are further divided into four genera: alpha‐, beta‐, gamma‐ and delta‐coronavirus. In humans, CoVs cause mainly respiratory tract infections. Currently, six human coronaviruses (HCoVs) have been identified. These include the alpha‐CoVs HCoV‐NL63 and HCoV‐229E and the beta‐CoVs HCoV‐OC43, HCoV‐HKU1, severe acute respiratory syndrome‐CoV (SARS‐CoV),2 and Middle East respiratory syndrome‐CoV (MERS‐CoV).3

Although HCoVs have been identified for decades, their clinical importance and epidemic possibility was not recognized until the outbreak of SARS and MERS.2, 3 In 2002, the SARS epidemic originated from an animal market in South China and then affected more than 8000 people, with 916 deaths in 29 countries.4 Subsequently, the World Health Organization (WHO) was notified of 2066 laboratory‐confirmed cases of MERS‐CoV infection, with at least 720 deaths between 2012 and 17 August 2017.5 While found in 27 countries, more that 80% of illnesses were reported from Saudi Arabia.

This article will review the epidemiology, pathogenesis, clinical characteristics and management of patients with HCoVs infection.

EPIDEMIOLOGY

Origin of HCOVs

Although CoVs are estimated to have circulated on earth for centuries,6, 7 the origin of CoVs remains obscure. At the beginning of the outbreak of SARS and MERS, palm civets8 and dromedary camels,9 respectively, were suggested to be the natural reservoir of these two HCoVs. But further virologic and genetic studies indicate that bats are reservoir hosts of both SARS‐CoV10 and MERS‐CoV,11 which then use palm civets and dromedary camels as intermediary host before dissemination to humans. Recent studies further propose that bat CoVs are the gene source of most alpha‐CoVs and beta‐CoVs, whereas avian CoVs are considered the gene source of most gamma‐ and delta‐CoVs.6, 12 Meanwhile, rodents are proposed to be the reservoir for ancestors of lineage A beta‐CoVs which include HCoV‐HKU1 and HCoV‐OC43.13

Transmission from animal to human

The mechanism and route of transmission of SARS‐CoV and MERS‐CoV remains elusive. Direct contact with intermediary host animals or consumption of milk, urine, or uncooked meat were hypothesized to be the main routes of SARS‐COV and MERS‐CoV transmission.

Transmission from human to human

Human‐to‐human transmission of SARS‐CoV and MERS‐CoV occurs mainly through nosocomial transmission. From 43.5–100% of MERS patients in individual outbreaks were linked to hospitals,14, 15 which was similar in SARS patients.16 A study from the Republic of Korea revealed that index patients who transmitted to others had more non‐isolated days in the hospital, body temperature of ≥38.5°C and pulmonary infiltration of ≥3 lung zones.17 Transmission between family members occurred in only 13–21% of MERS cases and 22–39% of SARS cases.17 Another Korean study suggested that transmission of MERS from an asymptomatic patient is rare.18 In contrast to SARS‐CoV and MERS‐CoV, direct human‐to‐human transmission was not reported for the other four HCoVs.19

Clinical epidemiology

The SARS epidemic originated from an animal market in Guangdong Province of China and subsequently spread to 29 countries. No large outbreaks have been reported in other areas after the initial epidemic. Nosocomial acquisition was very important for SARS as health care workers comprised 22% of reported cases in China and >40% in Canada.20

A large majority of MERS cases have occurred in the Arabian Peninsula.21, 22 A case–control study comparing 30 MERS patients to 116 controls in Saudi Arabia found direct contact with dromedaries in the 2 weeks before illness onset was associated with MERS‐HCoV illness.23 Outbreaks in other countries all resulted from index cases with travel history to the Middle East or North Africa.24, 25

The other HCoVs have a global distribution and are mainly transmitted in a seasonal endemic way,26 usually peaking in winter and spring, with a few cases occurred in early summer.27, 28 Epidemic studies of community‐acquired pneumonia (CAP) revealed that the four non‐SARS, non‐MERS HCoVs accounted for 0.6–2.5% of adult CAP patients.19, 29, 30, 31

PATHOGENESIS

Current understanding of the pathogenesis of HCoVs infection is still limited. However, several significant differences in the pathogenesis exist among SARS‐CoV, MERS‐CoV and the other HCoVs.

Cell entry and receptors

The critical first step for HCoV infection is entry into the susceptible host cells by combining with a specific receptor. Spike proteins (S proteins) of HCoVs are a surface‐located trimeric glycoprotein consisting of two subunits: the N‐terminal S1 subunit and the C‐terminal S2 subunit. The S1 subunit specializes in recognizing and binding to the host cell receptor while the S2 region is responsible for membrane fusion.32 To date, a wide range of diverse cellular receptors specifically recognized by the S1 domains have been identified for all HCoVs except HCoV‐HKU1 (Table (Table11).

Table 1

Biological characteristic of SARS‐COV, MERS‐CoV and other HCoVs

SARS‐CoVMERS‐CoVHCoV‐229EHCoV‐NL63HCoV‐OC43HCoV‐HKU1
GenusBeta‐CoVs lineage BBeta‐CoVs, lineage CAlpha‐CoVsAlpha‐CoVsBeta‐CoVs, lineage ABeta‐CoVs, lineage A
Intermediary hostPalm civetDromedary, camelNot definedNot definedNot definedNot defined
ReceptorACE2Dipeptidyl peptidase 4 (DPP4 or CD26)Human aminopeptidase N (CD13)ACE29‐O‐Acetyl‐ated sialic acidNot identified
Receptor distributionArterial and venous endothelium; arterial smooth muscle; small intestine, respiratory tract epithelium; alveolar monocytes and macrophagesRespiratory tract epithelium; kidney, small intestine; liver and prostate; activated leukocytesMonocytic and granulocytic lineage; synaptic membranes of the central nervous system; intestinal, lung and kidney epithelial cellsSame as SARS‐CoVSub‐maxillary mucin
Susceptibility in human cell lines in vitroRespiratory tract; kidney; liverRespiratory tract; intestinal tract; genitourinary tract; liver, kidney, neurons; monocyte; T lymphocyte; and histiocytic cell linesLiver, primary embryonic lung fibroblasts, neural tissue, monocytes, dendritic cells and macrophagesIntestinal tract; kidneyIntestinal tract; neural tissueCiliated airway epithelial

ACE2, angiotensin‐converting enzyme 2; CoV, coronavirus; HCoV, human coronavirus; MERS, Middle East respiratory syndrome; SARS, severe acute respiratory syndrome.

ACE2, the receptor for SARS‐CoV and HCoV‐NL63,1, 33, 34 is a surface molecule localized on arterial and venous endothelial cells, arterial smooth muscle cells, epithelia of the small intestine and the respiratory tract. In the respiratory tract, ACE2 is expressed on the epithelial cells of alveoli, trachea, and bronchi, bronchial serous glands, and alveolar monocytes and macrophages. ACE2 is a homologue of the ACE protein, and both are key enzymes of the renin–angiotensin system.35 ACE2 plays a protective role in lung failure and its counterpart ACE promoting lung oedema and impaired lung function.36 Downregulation of ACE2, as occurs during SARS‐CoV infection, is believed to contribute to pathological changes in the lung.35, 37 This form of lung damage can be attenuated by blocking the renin–angiotensin pathway.37 Interestingly, HCoV‐NL63 also employs the SARS receptor for cellular entry,34 despite their markedly different pathogenicity and disease courses. This finding suggests that receptor usage may not be the only factor that determines the severity of HCoV infection.

Dipeptidyl peptidase 4 (DPP4, also known as CD26), the receptor for MERS‐CoV,38 is a multifunctional cell‐surface protein widely expressed on epithelial cells in kidney, small intestine, liver and prostate and on activated leukocytes. DPP4 is expressed in the upper respiratory tract epithelium of camels.39 In the human respiratory tract, DPP4 is mainly expressed in alveoli rather than the nasal cavity or conducting airways.38 DPP4 is a key factor in the activation of T cells and immune response costimulatory signals in T cells, which could indicate a possible manipulation of the host immune system.40

Human aminopeptidase N (CD13), a cell‐surface metalloprotease on intestinal, lung and kidney epithelial cells, has been identified as the receptor for hCoV‐229E.41 The receptor for HCoV‐OC43 is 9‐O‐acetylated sialic acid. Currently, the receptor for HCoV‐HKU1 has not been identified.

Interferon and interferon‐stimulated genes

The interferon (IFN) family of cytokines, including IFN‐α, IFN‐β and IFN‐γ, provide the first line of defence against viral pathogens. They initiate transcription of hundreds of IFN‐stimulated genes (ISGs) that have antiviral, immune modulatory and cell regulatory functions.

Delayed recognition is critical for HCoVs to survive and replicate in the host. in vitro studies showed that both SARS‐CoV and MERS‐CoV have evolved genetic mechanisms to delay IFN induction and dysregulate ISG effector functions in primary human airway epithelial cells or in cultured cells.42, 43 Menachery et al. found SARS‐CoV infection could result in IFN‐α induction only after 12 h in cultured Calu3 cells, with IFN‐β5 and IFN‐γ1 induction even further delayed.42 Similar to SARS‐CoV, MERS‐CoV also fails to induce IFNs prior to 12 h, with the exception of IFN‐α5. Lau et al. serially measured mRNA levels of eight cytokine genes up to 30 h post‐infection in Calu‐3 cells infected with MERS‐CoV and SARS‐CoV.43 Calu‐3 cells infected by MERS‐CoV showed marked induction of the proinflammatory cytokines IL‐1β, IL‐6 and IL‐8 at 30 h but lack of production of the innate antiviral cytokines tumour necrosis factor (TNF)‐α, IFN‐β and IFN‐γ‐induced protein‐10, compared with SARS‐CoV. These data suggest that MERS‐CoV attenuates innate immunity and induces a delayed proinflammatory response in human lung epithelial cells, which correlates with disease severity and clinical course.

To date, no evidence exists that the other HCoVs have the ability to inhibit IFN production or regulate ISG expression. This decreased ability to escape from the innate immune responses of the host may explain the generally milder clinical disease associated with HCoV infection with these genera.

Cell line tropism

The differential cell line susceptibility, species tropism and viral replication efficiency of HCoVs correlate with clinical and epidemiologic characteristics. Compared with SARS‐CoV and other HCoVs, MERS‐CoV has a much broader cell line tropism (Table (Table1).1). Chan and colleagues tested cell line susceptibility of MERS‐CoV in 15 human cell lines and found significantly increased mean viral loads in 11 after infection, including lower airway (A549, Calu‐3 and HFL), intestinal tract (Caco‐2), liver (Huh‐7), kidney (HEK), neuronal (NT2), monocyte (THP‐1 and U937), T lymphocyte (H9) and histiocyte (His‐1) cell lines.44 Respiratory, intestinal, liver, kidney and histiocyte cell lines also showed viral nucleoprotein expression by immunofluorescence, in addition to a high viral load. MERS‐CoV could induce cytopathic effects as early as day 1 in the intestinal and liver cell lines and on day 3 in the lower respiratory tract cell lines, faster than those induced by SARS‐CoV.45, 46 These findings could partly explain the apparently more severe clinical presentations and higher fatality rate in MERS patients.

DIAGNOSIS

Diagnosis of SARS and MERS is based on a comprehensive contact and travel history and precise laboratory tests. Current diagnostic tools include molecular methods, serology and viral culture.27, 47 The most common diagnostic method is molecular detection such as RT (reverse transcription)‐PCR or real‐time RT‐PCR using RNA extracted from respiratory tract samples,27 such as nasopharyngeal swab, sputum, deep tracheal aspirate or bronchoalveolar lavage. Notably, lower respiratory tract samples usually yield significantly higher viral loads and genome fractions than upper respiratory tract samples,48 consistent with the tissue tropism.

Sensitivity of antibody detection is usually lower than molecular methods and mostly used in retrospective diagnosis. For antibody detection, an interval of 14–21 days between acute and convalescent serum samples is required in order to document seroconversion of at least a four‐fold rise of the antibody titres. If only a single sample can be collected, at least 14 days after the onset of symptoms is required for validity. Serology can be considered when virology testing by RT‐PCR is limited or the infection is considered late in the course of the illness (>14 days).47

Viral culture is relatively time and labour consuming. Culture is much more useful in the initial phase of emerging epidemics before other diagnostic assays are clinically available. Furthermore, viral culture can also be employed in in vitro and in vivo antiviral and vaccine evaluation studies.49 Antigen detection assay is another potential diagnostic tool to confirm SARS‐CoV and MERS‐CoV infection but is not recommended by current WHO guidelines.47

CLINICAL CHARACTERISTICS

Demographic and clinical features

Both SARS and MERS present with a spectrum of disease severity ranging from flu‐like symptoms to acute respiratory distress syndrome (ARDS). Clinical characteristics comparing SARS and MERS patients are seen in Table Table22.

Table 2

Demographic and clinical features of MERS‐CoV and SARS‐CoV infection

Clinical and epidemiologic aspectsSARS n = 357 (%)MERS n = 245 (%)
Health care workers142 (40%)42 (17%)
Male158 (44%)154 (63%)
Co‐morbidities
Diabetes21 (5.9%)75 (31%)
Malignancy9 (2.5%)27 (11%)
Chronic pulmonary diseases (including COPD and asthma)5 (1.4%)32 (13%)
Chronic renal failure2 (0.1%)37 (15%)
Chronic heart disease24 (6.7%)37 (15%)
Chronic liver diseases (including chronic hepatitis B)12 (3.4%)10 (4.1%)
HypertensionNot mentioned81 (33%)
Others6 (1.7%)13 (5.3%)
Symptoms on admission
Fever356 (99%)206 (84%)
Headache139 (39%)46 (19%)
Myalgia211 (59%)98 (40%)
Cough208 (58%)155 (63%)
Shortness of breath95 (27%)86 (35%)
Sore throat61 (17%)33 (13%)
Nausea/vomiting55 (15%)37 (15%)
Diarrhoea62 (17%)50 (20%)
Clinical outcome
Invasive mechanical ventilation 59 (17%) 91(37%)
Death 18 (5.0%) 71 (29%)

CoV, coronavirus; MERS, Middle East respiratory syndrome; SARS, severe acute respiratory syndrome.

Age and underlying disease are significant independent predictors of various adverse outcomes in SARS.50 SARS cases were mainly seen in young healthy individuals; whereas half of the cases of MERS‐CoV infection occurred in individuals older than 50 years.21 Compared with SARS patients, pre‐existing chronic illnesses, such as diabetes (31%), hypertension (33%), chronic renal failure (15%), chronic heart disease (15%) and chronic pulmonary disease (13%), were more frequent in MERS patients. Clinical symptoms on admission included fever, cough, myalgia and shortness of breath in both SARS and MERS patients, while symptoms of upper respiratory tract infection such as sore throat were also frequent. Atypical symptoms such as diarrhoea and vomiting developed in both SARS and MERS patients.

The other HCoVs infect people of all age groups seasonally and cause severe lower respiratory tract infection primarily in frail patients, such as neonates and the elderly.51 Chronic underlying disease, immunosuppression and extremes of age increase the risk of severe HCoV infections and associated death rate.19, 51, 52

LABORATORY FINDINGS

Kidney impairment

Acute kidney injury (AKI) is a significant characteristic of both SARS and MERS patients. One study reported that 6.7% of SARS patients had acute renal impairment and 84.6% had proteinuria.53 AKI is much more common in MERS patients, occurring in up to 43%.54

The mechanism of the high AKI incidence in both SARS and MERS patients is not well clarified. Pre‐existing co‐morbid conditions and direct viral involvement of the kidneys62, 63 may contribute to development of AKI.53, 54 Since ACE2 and DPP4, the receptors for SARS‐CoV and MERS‐CoV, are expressed at high levels in the kidney, functional impairment of these cell receptors by viral binding may contribute to the risk of AKI. Elevated creatinine kinase (CK) values (176–1466 U/L) observed in 36% of SARS patients suggests rhabdomyolysis may also contribute.55

Cardiovascular manifestations

A cardinal difference between MERS and SARS is the frequency of cardiovascular involvement. Despite the high lethality, shock was distinctly unusual in SARS until late stages when hypotension likely resulted from bacterial superinfections.56, 57, 58 In contrast, need for vasopressor therapy was much more common in MERS,50, 58 up to 81% in one series.58 Need for vasopressors was an independent risk factor for death in the intensive care unit (ICU) (odds ratio = 18.3, 95% confidence interval: 1.1–302.1, P = 0.04).58 Multi‐organ involvement was seldom reported with the endemic HCoV infections,19 despite occasional fatal pneumonia in highly immunocompromised patients.

Other manifestations

Haematological abnormalities such as thrombocytopenia and lymphopenia were common in both SARS55, 56 and MERS patients.21, 22 Thrombocytopenia and lymphopenia may be predictive of fatal outcome in MERS‐CoV patients.22 Other laboratory findings included elevated CK, lactate dehydrogenase, alanine aminotransferase and aspartate aminotransferase levels.

RADIOLOGICAL

Air‐space opacities are the main radiographical feature in SARS patients.56, 59 In one retrospective study, initial chest radiographs were abnormal in 108 of 138 (78.3%) of SARS patients and all showed air‐space opacities.59 Of these 108 patients, 59 had unilateral focal involvement while 49 had either unilateral multifocal or bilateral involvement. Lower lung zone (64.8%) and right lung (75.9%) were more commonly involved. Four patterns of radiographical progression were recognized in those patients: type 1) initial radiographical deterioration to peak level followed by radiographical improvement occurred in in the majority (97 of 138 patients, 70.3%); type 2) fluctuating radiographical changes were seen in 24 patients (17.4%); type 3) static radiographical appearance in 10 patients (7.3%); and type 4) progressive radiographic deterioration in 7 patients (5.1%). In contrast, the most common radiographical features in MERS patients were ground–glass opacities and consolidation.60, 61 Das et al. reported that ground–glass opacity was the most common abnormality (66%) in 55 MERS patients, followed by consolidation (18%).61 Meanwhile, type 2 radiographical progression (20 patients) was most common in those MERS patients, followed by type 4 (14 patients) and type 3 (7 patients). Type 1 radiographical progression was observed only in four patients. Pleural effusion (P = 0.001), pneumothorax (P = 0.001) and type 4 radiographical progression (P = 0.001) were more frequent in MERS patients who died compared with recovered patients. Similar to the radiographical findings, computed tomography findings in MERS patients also included ground–glass opacity (53%), consolidation (20%) or a combination of both (33%).62 Pleural effusion was noted in 33% of cases and was associated with a poor prognosis for MERS‐CoV infection.61

OUTCOME

As shown in Table Table2,2, more MERS cases progressed to respiratory failure and received invasive mechanical ventilation therapy than SARS patients. The occurrence of AKI22, 54 and the usage of vasopressor therapy were also more frequent in MERS patients in comparison with SARS.53, 58 In a retrospective analysis, vasopressor therapy was proposed to be an independent risk factor for death in the ICU.58

MERS demonstrated a higher case fatality rate than SARS. Differences in host factors, such as age and underlying diseases,50, 60 may explain some differences. However, the differential cell line susceptibility, viral replication efficiency, ability to inhibit IFN production and receptor characteristics may also be responsible for the difference in the outcome of SARS‐CoV and MERS‐CoV infection.43, 44

Compared with SARS and MERS, other HCoVs‐associated pneumonia cases usually have relatively mild symptoms and recovered quickly.19 Fatal cases were reported mainly in frail patients, such as neonates, the elderly and immunocompromised patients.

TREATMENT

At the moment, no specific therapy for SARS‐CoV, MERS‐CoV and the other HCoVs infection is available. Symptomatic and supportive treatment is the mainstay of therapy for patients infected by HCoVs.

A number of agents show effectiveness in vitro and/or in animal models and may improve the outcome in patients (Table (Table3).3). Currently, the most commonly prescribed antiviral regimens in the clinical settings are ribavirin, IFNs and lopinavir/ritonavir.

Table 3

Comparison of the susceptibility of MERS‐CoV and SARS‐CoV with different antiviral agents

Antiviral agentsVirusesTested cell lineEC50 values
RibavirinSARS‐CoVCaco2 cells4.7 ± 2.6(0.3 ± 0.12 if ribavirin and IFN‐β combined)
Interferon‐βSARS‐CoVCaco2 cells28 ± 7 (0.6 ± 0.27 if ribavirin and IFN‐β combined)
RibavirinMERS‐CoVVero cells9.99 ± 2.97
Intron A (recombinant interferon‐α2b)MERS‐CoVVero cells6709.79 ± 1747.97
Avonex (recombinant interferon‐β1a)MERS‐CoVVero cells5073.33 ± 7333.86
Betaferon (recombinant interferon‐β1b)MERS‐CoVVero cells17.64 ± 1.09
Mycophenolic acidMERS‐CoVVero cells0.17 ± 0.03

CoV, coronavirus; EC50, 50% effective cytotoxic concentration; IFN, interferon; MERS, Middle East respiratory syndrome; SARS, severe acute respiratory syndrome.

To date, ribavirin and ribavirin plus various types of IFN have been the most common therapeutic interventions tried in patients with SARS and MERS.63, 64, 65 Ribavirin, a nucleoside analogue, has a wide spectrum of antiviral activity by inhibiting viral RNA synthesis and mRNA capping.66 When used alone for treatment of SARS, the clinical effect was inconsistent. Although in vitro studies show that combination with IFN‐β will give both these agents better antiviral activity, the clinical effect remains controversial.

IFNs are important for host defence against viruses. In in vitro experiments, IFN products were effective in inhibiting both SARS‐CoV and MRES‐CoV, with best antiviral activity seen with IFN‐β1b (Table (Table33).63, 67 Previous studies had shown a positive impact of various IFNs on aspects of treatment of SARS and MERS patients, such as a better oxygen saturation and rapid resolution of inflammation, but no effect on more significant outcomes like hospital stay and long‐term survival.64, 65, 68

Lopinavir and ritonavir are protease inhibitors that may inhibit the 3C‐like protease of MERS‐CoV and modulate apoptosis in human cells. Addition of lopinavir/ritonavir to ribavirin was associated with improved clinical outcome compared with ribavirin alone in SARS patients.69 Although lopinavir only showed suboptimal 50% effective cytotoxic concentration (EC50) against MERS‐CoV in vitro,67 lopinavir/ritonavir experimental therapy was proved to improve the outcome of MERS‐CoV infection in animal model.70

Mycophenolic acid (MPA) is another potential therapeutic choice. Frequently used as an immunosuppressant drug to prevent rejection in organ transplantation by inhibiting lymphocyte proliferation, MPA also prevents replication of viral RNA. in vitro studies showed that MPA had strong inhibition activity against MERS‐CoV.71 However, use in a non‐human primate model showed that all MPA‐treated animals developed severe and/or fatal disease with higher mean viral loads than the untreated animals.70

Passive immunotherapy using convalescent phase human plasma was also used in the treatment of SARS and MERS. An exploratory meta‐analysis found that convalescent plasma decreased mortality in SARS‐CoV patients only if administered within 14 days of illness.72 A network for the use of convalescent plasma in the treatment of MERS cases is currently being formed to test its safety, efficacy and feasibility.73

Corticosteroids were used extensively during the SARS outbreak, generally in combination with ribavirin. Lessons from SARS showed that corticosteroid treatment was associated with a higher subsequent plasma viral load74 with increased complications.

A variety of other agents, including antiviral peptides, monoclonal antibodies, cell or viral protease inhibitors antivirals, are shown to be effective in vitro and/or in animal models.75, 76, 77, 78 Clinical trials of these agents are awaited.

SUMMARY

The pandemic potential of HCoVs remains a threat for public health and active surveillance is prudent. As no specific treatment is currently available for HCoVs, further research into the pathogenesis of the HCoVs infection in order to find appropriate targets for treatment is needed. In immunosuppressed pneumonia patients, non‐SARS, non‐MERS HCoVs should be included in the differential diagnosis.

The Authors

Yu‐dong Yin, MD is an Infectious Diseases and Clinical Microbiology physician from Beijing Chao‐Yang Hospital, affiliated with Capital Medical University in Beijing, China. He has been actively involved in research on aetiology of community‐acquired pneumonia. He has a particular interest in atypical pathogens, especially Mycoplasma. Richard G. Wunderink MD is a Pulmonary and Critical Care physician from Northwestern University Feinberg School of Medicine in Chicago IL. He has a longstanding interest in diagnosis of pneumonia, particularly severe community‐acquired pneumonia. He was the site principal investigator for the recently completed US Centers for Disease Control and Prevention‐sponsored Epidemiology of Pneumonia in the Community (EPIC) study.

Notes

Yin, Y. and Wunderink, R.G. (2017) MERS, SARS and other coronaviruses as causes of pneumonia. Respirology, 23: 130–137., doi: 10.1111/resp.13196. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

Series Editor: Grant Waterer

REFERENCES

1. Weiss SR, Leibowitz JL. Coronavirus pathogenesis. Adv. Virus Res. 2011; 81: 85–164. [PMC free article] [PubMed] [Google Scholar]
2. Drosten C, Gunther 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: 1967–76. [PubMed] [Google Scholar]
3. 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: 1814–20. [PubMed] [Google Scholar]
4. World Health Organization . WHO Guidelines for the Global Surveillance of Severe Acute Respiratory Syndrome (SARS) Updated recommendations, October 2004. [Accessed 28 Aug 2017] Available from URL: http://www.who.int/csr/resources/publications/WHO_CDS_CSR_ARO_2004_1/en/.
5. World Health Organization . Middle East Respiratory Syndrome Coronavirus (MERS‐CoV) Fact Sheet [Accessed 28 Aug 2017.] Available from URL: http://www.who.int/mediacentre/factsheets/mers-cov/en/.
6. Woo PC, Lau SK, Lam CS, Lau CC, Tsang AK, Lau JH, Bai R, Teng JL, Tsang CC, Wang M et al. Discovery of seven novel Mammalian and avian coronaviruses in the genus deltacoronavirus supports bat coronaviruses as the gene source of alphacoronavirus and betacoronavirus and avian coronaviruses as the gene source of gammacoronavirus and deltacoronavirus. J. Virol. 2012; 86: 3995–4008. [PMC free article] [PubMed] [Google Scholar]
7. Huynh J, Li S, Yount B, Smith A, Sturges L, Olsen JC, Nagel J, Johnson JB, Agnihothram S, Gates JE et al. Evidence supporting a zoonotic origin of human coronavirus strain NL63. J. Virol. 2012; 86: 12816–25. [PMC free article] [PubMed] [Google Scholar]
8. Guan Y, Zheng BJ, He YQ, Liu XL, Zhuang ZX, Cheung CL, Luo SW, Li PH, Zhang LJ, Guan YJ et al. Isolation and characterization of viruses related to the SARS coronavirus from animals in southern China. Science 2003; 302: 276–8. [PubMed] [Google Scholar]
9. Azhar EI, El‐Kafrawy SA, Farraj SA, Hassan AM, Al‐Saeed MS, Hashem AM, Madani TA. Evidence for camel‐to‐human transmission of MERS coronavirus. N. Engl. J. Med. 2014; 370: 2499–505. [PubMed] [Google Scholar]
10. Li W, Shi Z, Yu M, Ren W, Smith C, Epstein JH, Wang H, Crameri G, Hu Z, Zhang H et al. Bats are natural reservoirs of SARS‐like coronaviruses. Science 2005; 310: 676–9. [PubMed] [Google Scholar]
11. Ithete NL, Stoffberg S, Corman VM, Cottontail VM, Richards LR, Schoeman MC, Drosten C, Drexler JF, Preiser W. Close relative of human Middle East respiratory syndrome coronavirus in bat, South Africa. Emerg. Infect. Dis. 2013; 19: 1697–9. [PMC free article] [PubMed] [Google Scholar]
12. Woo PC, Lau SK, Li KS, Poon RW, Wong BH, Tsoi HW, Yip BC, Huang Y, Chan KH, Yuen KY. Molecular diversity of coronaviruses in bats. Virology 2006; 351: 180–7. [PMC free article] [PubMed] [Google Scholar]
13. Lau SK, Woo PC, Li KS, Tsang AK, Fan RY, Luk HK, Cai JP, Chan KH, Zheng BJ, Wang M et al. Discovery of a novel coronavirus, China Rattus coronavirus HKU24, from Norway rats supports the murine origin of Betacoronavirus 1 and has implications for the ancestor of Betacoronavirus lineage A. J. Virol. 2015; 89: 3076–92. [PMC free article] [PubMed] [Google Scholar]
14. Hunter JC, Nguyen D, Aden B, Al Bandar Z, Al Dhaheri W, Abu Elkheir K, Khudair A, Al Mulla M, El Saleh F, Imambaccus H et al. Transmission of Middle East respiratory syndrome coronavirus infections in healthcare settings, Abu Dhabi. Emerg. Infect. Dis. 2016; 22: 647–56. [PMC free article] [PubMed] [Google Scholar]
15. Korea Centers for Disease Control and Prevention . Middle East Respiratory syndrome coronavirus outbreak in the Republic of Korea, 2015. Osong Public Health Res. Perspect. 2015; 6: 269–78. [PMC free article] [PubMed] [Google Scholar]
16. Anderson RM, Fraser C, Ghani AC, Donnelly CA, Riley S, Ferguson NM, Leung GM, Lam TH, Hedley AJ. Epidemiology, transmission dynamics and control of SARS: the 2002–2003 epidemic. Philos. Trans. R. Soc. Lond. B Biol. Sci. 2004; 359: 1091–105. [PMC free article] [PubMed] [Google Scholar]
17. Kang CK, Song KH, Choe PG, Park WB, Bang JH, Kim ES, Park SW, Kim HB, Kim NJ, Cho SI et al. Clinical and epidemiologic characteristics of spreaders of Middle East respiratory syndrome coronavirus during the 2015 Outbreak in Korea. J. Korean Med. Sci. 2017; 32: 744–9. [PMC free article] [PubMed] [Google Scholar]
18. Moon SY, Son JS. Infectivity of an asymptomatic patient with Middle East respiratory syndrome coronavirus infection. Clin. Infect. Dis. 2017; 64: 1457–8. [PMC free article] [PubMed] [Google Scholar]
19. Woo PC, Lau SK, Yuen KY. Clinical features and molecular epidemiology of coronavirus‐HKU1‐associated community‐acquired pneumonia. Hong Kong Med. J. 2009; 15 (Suppl. 9): 46–7. [PubMed] [Google Scholar]
20. Skowronski DM, Astell C, Brunham RC, Low DE, Petric M, Roper RL, Talbot PJ, Tam T, Babiuk L. Severe acute respiratory syndrome (SARS): a year in review. Annu. Rev. Med. 2005; 56: 357–81. [PubMed] [Google Scholar]
21. Assiri A, Al‐Tawfiq JA, Al‐Rabeeah AA, Al‐Rabiah FA, Al‐Hajjar S, Al‐Barrak A, Flemban H, Al‐Nassir WN, Balkhy HH, Al‐Hakeem RF 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: 752–61. [PMC free article] [PubMed] [Google Scholar]
22. 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: 389–97. [PubMed] [Google Scholar]
23. Alraddadi BM, Watson JT, Almarashi A, Abedi GR, Turkistani A, Sadran M, Housa A, Almazroa MA, Alraihan N, Banjar A et al. Risk Factors for Primary Middle East Respiratory Syndrome Coronavirus Illness in Humans, Saudi Arabia, 2014. Emerg. Infect. Dis. 2016; 22: 49–55. [PMC free article] [PubMed] [Google Scholar]
24. Park SH, Kim YS, Jung Y, Choi SY, Cho NH, Jeong HW, Heo JY, Yoon JH, Lee J, Cheon S et al. Outbreaks of Middle East respiratory syndrome in two hospitals initiated by a single patient in Daejeon, South Korea. Infect. Chemother. 2016; 48: 99–107. [PMC free article] [PubMed] [Google Scholar]
25. Mailles A, Blanckaert K, Chaud P, van der Werf S, Lina B, Caro V, Campese C, Guery B, Prouvost H, Lemaire X et al. Investigation Team . First cases of Middle East Respiratory Syndrome Coronavirus (MERS‐CoV) infections in France, investigations and implications for the prevention of human‐to‐human transmission, France, May 2013. Euro Surveill. 2013; 18: pii 20502. [PubMed] [Google Scholar]
26. Lau SK, Woo PC, Yip CC, Tse H, Tsoi HW, Cheng VC, Lee P, Tang BS, Cheung CH, Lee RA et al. Coronavirus HKU1 and other coronavirus infections in Hong Kong. J. Clin. Microbiol. 2006; 44: 2063–71. [PMC free article] [PubMed] [Google Scholar]
27. Woo PC, Yuen KY, Lau SK. Epidemiology of coronavirus‐associated respiratory tract infections and the role of rapid diagnostic tests: a prospective study. Hong Kong Med. J. 2012; 18 (Suppl. 2): 22–4. [PubMed] [Google Scholar]
28. Lee WJ, Chung YS, Yoon HS, Kang C, Kim K. Prevalence and molecular epidemiology of human coronavirus HKU1 in patients with acute respiratory illness. J. Med. Virol. 2013; 85: 309–14. [PMC free article] [PubMed] [Google Scholar]
29. Johnstone J, Majumdar SR, Fox JD, Marrie TJ. Viral infection in adults hospitalized with community‐acquired pneumonia: prevalence, pathogens, and presentation. Chest 2008; 134: 1141–8. [PMC free article] [PubMed] [Google Scholar]
30. Jain S, Self WH, Wunderink RG, Fakhran S, Balk R, Bramley AM, Reed C, Grijalva CG, Anderson EJ, Courtney DM et al. Community‐acquired pneumonia requiring hospitalization among U.S. adults. N. Engl. J. Med. 2015; 373: 415–27. [PMC free article] [PubMed] [Google Scholar]
31. Cao B, Ren LL, Zhao F, Gonzalez R, Song SF, Bai L, Yin YD, Zhang YY, Liu YM, Guo P et al. Viral and Mycoplasma pneumoniae community‐acquired pneumonia and novel clinical outcome evaluation in ambulatory adult patients in China. Eur. J. Clin. Microbiol. Infect. Dis. 2010; 29: 1443–8. [PMC free article] [PubMed] [Google Scholar]
32. Gallagher TM, Buchmeier MJ. Coronavirus spike proteins in viral entry and pathogenesis. Virology 2001; 279: 371–4. [PMC free article] [PubMed] [Google Scholar]
33. 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: 450–4. [PMC free article] [PubMed] [Google Scholar]
34. Hofmann H, Pyrc K, van der Hoek L, Geier M, Berkhout B, Pohlmann S. Human coronavirus NL63 employs the severe acute respiratory syndrome coronavirus receptor for cellular entry. Proc. Natl. Acad. Sci. U. S. A. 2005; 102: 7988–93. [PMC free article] [PubMed] [Google Scholar]
35. Imai Y, Kuba K, Ohto‐Nakanishi T, Penninger JM. Angiotensin‐converting enzyme 2 (ACE2) in disease pathogenesis. Circ. J. 2010; 74: 405–10. [PubMed] [Google Scholar]
36. Imai Y, Kuba K, Rao S, Huan Y, Guo F, Guan B, Yang P, Sarao R, Wada T, Leong‐Poi H et al. Angiotensin‐converting enzyme 2 protects from severe acute lung failure. Nature 2005; 436: 112–6. [PMC free article] [PubMed] [Google Scholar]
37. Kuba K, Imai Y, Rao S, Gao H, Guo F, Guan B, Huan Y, Yang P, Zhang Y, Deng W et al. A crucial role of angiotensin converting enzyme 2 (ACE2) in SARS coronavirus‐induced lung injury. Nat. Med. 2005; 11: 875–9. [PMC free article] [PubMed] [Google Scholar]
38. Meyerholz DK, Lambertz AM, PB MC Jr. Dipeptidyl peptidase 4 distribution in the human respiratory tract: implications for the Middle East respiratory syndrome. Am J Pathol. 2016; 186: 78–86. [PMC free article] [PubMed] [Google Scholar]
39. Widagdo W, Raj VS, Schipper D, Kolijn K, van Leenders GJ, Bosch BJ, Bensaid A, Segales J, Baumgartner W, Osterhaus AD et al. Differential expression of the Middle East respiratory syndrome coronavirus receptor in the upper respiratory tracts of humans and dromedary camels. J. Virol. 2016; 90: 4838–42. [PMC free article] [PubMed] [Google Scholar]
40. Boonacker E, Van Noorden CJ. The multifunctional or moonlighting protein CD26/DPPIV. Eur. J. Cell Biol. 2003; 82: 53–73. [PubMed] [Google Scholar]
41. Yeager CL, Ashmun RA, Williams RK, Cardellichio CB, Shapiro LH, Look AT, Holmes KV. Human aminopeptidase N is a receptor for human coronavirus 229E. Nature 1992; 357: 420–2. [PMC free article] [PubMed] [Google Scholar]
42. Menachery VD, Eisfeld AJ, Schafer A, Josset L, Sims AC, Proll S, Fan S, Li C, Neumann G, Tilton SC et al. Pathogenic influenza viruses and coronaviruses utilize similar and contrasting approaches to control interferon‐stimulated gene responses. MBio 2014; 5: e01174–14. [PMC free article] [PubMed] [Google Scholar]
43. 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: 2679–90. [PubMed] [Google Scholar]
44. Chan JF, Chan KH, Choi GK, To KK, Tse H, Cai JP, Yeung ML, Cheng VC, Chen H, Che XY et al. Differential cell line susceptibility to the emerging novel human betacoronavirus 2c EMC/2012: implications for disease pathogenesis and clinical manifestation. J. Infect. Dis. 2013; 207: 1743–52. [PMC free article] [PubMed] [Google Scholar]
45. Yoshikawa T, Hill TE, Yoshikawa N, Popov VL, Galindo CL, Garner HR, Peters CJ, Tseng CT. Dynamic innate immune responses of human bronchial epithelial cells to severe acute respiratory syndrome‐associated coronavirus infection. PLoS One 2010; 5: e8729. [PMC free article] [PubMed] [Google Scholar]
46. Kaye M. SARS‐associated coronavirus replication in cell lines. Emerg. Infect. Dis. 2006; 12: 128–33. [PMC free article] [PubMed] [Google Scholar]
47. World Health Organization . Clinical Management of Severe Acute Respiratory Infection When Middle East Respiratory Syndrome Coronavirus (MERS‐CoV) Infection Is Suspected [Accessed 28 Aug 2017.] Available from URl: http://www.who.int/csr/disease/coronavirus_infections/case-management-ipc/en.
48. Memish ZA, Al‐Tawfiq JA, Makhdoom HQ, Assiri A, Alhakeem RF, Albarrak A, Alsubaie S, Al‐Rabeeah AA, Hajomar WH, Hussain R et al. Respiratory tract samples, viral load, and genome fraction yield in patients with Middle East respiratory syndrome. J. Infect. Dis. 2014; 210: 1590–4. [PMC free article] [PubMed] [Google Scholar]
49. Chan JF, Sridhar S, Yip CC, Lau SK, Woo PC. The role of laboratory diagnostics in emerging viral infections: the example of the Middle East respiratory syndrome epidemic. J. Microbiol. 2017; 55: 172–82. [PMC free article] [PubMed] [Google Scholar]
50. Chan KS, Zheng JP, Mok YW, Li YM, Liu YN, Chu CM, Ip MS. SARS: prognosis, outcome and sequelae. Respirology 2003; 8(Suppl): S36–40. [PMC free article] [PubMed] [Google Scholar]
51. Nunes MC, Kuschner Z, Rabede Z, Madimabe R, Van Niekerk N, Moloi J, Kuwanda L, Rossen JW, Klugman KP, Adrian PV et al. Clinical epidemiology of bocavirus, rhinovirus, two polyomaviruses and four coronaviruses in HIV‐infected and HIV‐uninfected South African children. PLoS One 2014; 9: e86448. [PMC free article] [PubMed] [Google Scholar]
52. Garbino J, Crespo S, Aubert JD, Rochat T, Ninet B, Deffernez C, Wunderli W, Pache JC, Soccal PM, Kaiser L. A prospective hospital‐based study of the clinical impact of non‐severe acute respiratory syndrome (Non‐SARS)‐related human coronavirus infection. Clin. Infect. Dis. 2006; 43: 1009–15. [PMC free article] [PubMed] [Google Scholar]
53. Chu KH, Tsang WK, Tang CS, Lam MF, Lai FM, To KF, Fung KS, Tang HL, Yan WW, Chan HW et al. Acute renal impairment in coronavirus‐associated severe acute respiratory syndrome. Kidney Int. 2005; 67: 698–705. [PMC free article] [PubMed] [Google Scholar]
54. Saad M, Omrani AS, Baig K, Bahloul A, Elzein F, Matin MA, Selim MA, Al Mutairi M, Al Nakhli D, Al Aidaroos AY 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: 301–6. [PMC free article] [PubMed] [Google Scholar]
55. 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: 1767–72. [PMC free article] [PubMed] [Google Scholar]
56. Booth CM, Matukas LM, Tomlinson GA, Rachlis AR, Rose DB, Dwosh HA, Walmsley SL, Mazzulli T, Avendano M, Derkach P et al. Clinical features and short‐term outcomes of 144 patients with SARS in the greater Toronto area. JAMA 2003; 289: 2801–9. [PubMed] [Google Scholar]
57. Lee N, Hui D, Wu A, Chan P, Cameron P, Joynt GM, Ahuja A, Yung MY, Leung CB, To KF et al. A major outbreak of severe acute respiratory syndrome in Hong Kong. N. Engl. J. Med. 2003; 348: 1986–94. [PubMed] [Google Scholar]
58. Almekhlafi GA, Albarrak MM, Mandourah Y, Hassan S, Alwan A, Abudayah A, Altayyar S, Mustafa M, Aldaghestani T, Alghamedi A et al. Presentation and outcome of Middle East respiratory syndrome in Saudi intensive care unit patients. Crit. Care 2016; 20: 123. [PMC free article] [PubMed] [Google Scholar]
59. Wong KT, Antonio GE, Hui DS, Lee N, Yuen EH, Wu A, Leung CB, Rainer TH, Cameron P, Chung SS et al. Severe acute respiratory syndrome: radiographic appearances and pattern of progression in 138 patients. Radiology 2003; 228: 401–6. [PubMed] [Google Scholar]
60. Choi WS, Kang CI, Kim Y, Choi JP, Joh JS, Shin HS, Kim G, Peck KR, Chung DR, Kim HO et al.; Korean Society of Infectious Diseases . Clinical presentation and outcomes of Middle East respiratory syndrome in the Republic of Korea. Infect. Chemother. 2016; 48: 118–26. [PMC free article] [PubMed] [Google Scholar]
61. Das KM, Lee EY, Al Jawder SE, Enani MA, Singh R, Skakni L, Al‐Nakshabandi N, AlDossari K, Larsson SG. Acute Middle East respiratory syndrome coronavirus: temporal lung changes observed on the chest radiographs of 55 patients. Am. J. Roentgenol. 2015; 205: W267–74. [PubMed] [Google Scholar]
62. Das KM, Lee EY, Enani MA, AlJawder SE, Singh R, Bashir S, Al‐Nakshbandi N, AlDossari K, Larsson SG. CT correlation with outcomes in 15 patients with acute Middle East respiratory syndrome coronavirus. Am. J. Roentgenol. 2015; 204: 736–42. [PubMed] [Google Scholar]
63. Morgenstern B, Michaelis M, Baer PC, Doerr HW, Cinatl J Jr. Ribavirin and interferon‐beta synergistically inhibit SARS‐associated coronavirus replication in animal and human cell lines. Biochem. Biophys. Res. Commun. 2005; 326: 905–8. [PMC free article] [PubMed] [Google Scholar]
64. 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: 42–6. [PMC free article] [PubMed] [Google Scholar]
65. Omrani AS, Saad MM, Baig K, Bahloul A, Abdul‐Matin M, Alaidaroos AY, Almakhlafi GA, Albarrak MM, Memish ZA, Albarrak AM. Ribavirin and interferon alfa‐2a for severe Middle East respiratory syndrome coronavirus infection: a retrospective cohort study. Lancet Infect. Dis. 2014; 14: 1090–5. [PMC free article] [PubMed] [Google Scholar]
66. von Grotthuss M, Wyrwicz LS, Rychlewski L. mRNA cap‐1 methyltransferase in the SARS genome. Cell 2003; 113: 701–2. [PMC free article] [PubMed] [Google Scholar]
67. Chan JF, Chan KH, Kao RY, To KK, Zheng BJ, Li CP, Li PT, Dai J, Mok FK, Chen H, Hayden FG, Yuen KY. Broad‐spectrum antivirals for the emerging Middle East respiratory syndrome coronavirus. J. Infect. 2013; 67: 606–16. [PMC free article] [PubMed] [Google Scholar]
68. Loutfy MR, Blatt LM, Siminovitch KA, Ward S, Wolff B, Lho H, Pham DH, Deif H, LaMere EA, Chang M et al. Interferon alfacon‐1 plus corticosteroids in severe acute respiratory syndrome: a preliminary study. JAMA 2003; 290: 3222–8. [PubMed] [Google Scholar]
69. Chu CM, Cheng VC, Hung IF, Wong MM, Chan KH, Chan KS, Kao RY, Poon LL, Wong CL, Guan Y et al. HKU/UCH SARS Study Group . Role of lopinavir/ritonavir in the treatment of SARS: initial virological and clinical findings. Thorax 2004; 59: 252–6. [PMC free article] [PubMed] [Google Scholar]
70. Chan JF, Yao Y, Yeung ML, Deng W, Bao L, Jia L, Li F, Xiao C, Gao H, Yu P et al. Treatment with lopinavir/ritonavir or interferon‐beta1b improves outcome of MERS‐CoV infection in a nonhuman primate model of common marmoset. J. Infect. Dis. 2015; 212: 1904–13. [PMC free article] [PubMed] [Google Scholar]
71. Hart BJ, Dyall J, Postnikova E, Zhou H, Kindrachuk J, Johnson RF, Olinger GG Jr, Frieman MB, Holbrook MR, Jahrling PB et al. Interferon‐beta and mycophenolic acid are potent inhibitors of Middle East respiratory syndrome coronavirus in cell‐based assays. J. Gen. Virol. 2014; 95: 571–7. [PMC free article] [PubMed] [Google Scholar]
72. Mair‐Jenkins J, Saavedra‐Campos M, Baillie JK, Cleary P, Khaw FM, Lim WS, Makki S, Rooney KD, Nguyen‐Van‐Tam JS, Beck CR 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. [PMC free article] [PubMed] [Google Scholar]
73. Arabi Y, Balkhy H, Hajeer AH, Bouchama A, Hayden FG, Al‐Omari A, Al‐Hameed FM, Taha Y, Shindo N, Whitehead J et al. Feasibility, safety, clinical, and laboratory effects of convalescent plasma therapy for patients with Middle East respiratory syndrome coronavirus infection: a study protocol. Springerplus 2015; 4: 709. [PMC free article] [PubMed] [Google Scholar]
74. Lee N, Allen Chan KC, Hui DS, Ng EK, Wu A, Chiu RW, Wong VW, Chan PK, Wong KT, Wong E et al. Effects of early corticosteroid treatment on plasma SARS‐associated Coronavirus RNA concentrations in adult patients. J. Clin. Virol. 2004; 31: 304–9. [PMC free article] [PubMed] [Google Scholar]
75. Zumla A, Chan JF, Azhar EI, Hui DS, Yuen KY. Coronaviruses ‐ drug discovery and therapeutic options. Nat. Rev. Drug Discov. 2016; 15: 327–47. [PMC free article] [PubMed] [Google Scholar]
76. Agrawal AS, Ying T, Tao X, Garron T, Algaissi A, Wang Y, Wang L, Peng BH, Jiang S, Dimitrov DS et al. Passive transfer of a germline‐like neutralizing human monoclonal antibody protects transgenic mice against lethal Middle East respiratory syndrome coronavirus infection. Sci. Rep. 2016; 6: 31629. [PMC free article] [PubMed] [Google Scholar]
77. Tao X, Mei F, Agrawal A, Peters CJ, Ksiazek TG, Cheng X, Tseng CT. Blocking of exchange proteins directly activated by cAMP leads to reduced replication of Middle East respiratory syndrome coronavirus. J. Virol. 2014; 88: 3902–10. [PMC free article] [PubMed] [Google Scholar]
78. Ohnuma K, Haagmans BL, Hatano R, Raj VS, Mou H, Iwata S, Dang NH, Bosch BJ, Morimoto C. Inhibition of Middle East respiratory syndrome coronavirus infection by anti‐CD26 monoclonal antibody. J. Virol. 2013; 87: 13892–9. [PMC free article] [PubMed] [Google Scholar]

Articles from Respirology (Carlton, Vic.) are provided here courtesy of Wiley

-