You are on page 1of 12

Expert Review of Molecular Diagnostics

ISSN: (Print) (Online) Journal homepage: https://www.tandfonline.com/loi/iero20

Diagnosis of SARS-CoV-2 infection in the setting of


the cytokine release syndrome

Marwan M. Azar, Junghee J. Shin, Insoo Kang & Marie Landry

To cite this article: Marwan M. Azar, Junghee J. Shin, Insoo Kang & Marie Landry (2020)
Diagnosis of SARS-CoV-2 infection in the setting of the cytokine release syndrome, Expert Review
of Molecular Diagnostics, 20:11, 1087-1097, DOI: 10.1080/14737159.2020.1830760

To link to this article: https://doi.org/10.1080/14737159.2020.1830760

Published online: 12 Oct 2020.

Submit your article to this journal

Article views: 2030

View related articles

View Crossmark data

Citing articles: 5 View citing articles

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=iero20
EXPERT REVIEW OF MOLECULAR DIAGNOSTICS
2020, VOL. 20, NO. 11, 1087–1097
https://doi.org/10.1080/14737159.2020.1830760

SPECIAL REPORT

Diagnosis of SARS-CoV-2 infection in the setting of the cytokine release syndrome


Marwan M. Azara, Junghee J. Shinb, Insoo Kangb and Marie Landrya,c
a
Department of Internal Medicine, Section of Infectious Diseases, Yale School of Medicine, New Haven, CT, USA; bDepartment of Internal Medicine,
Section of Rheumatology, Allergy & Immunology, Yale School of Medicine, New Haven, CT, USA; cDepartment of Laboratory Medicine, Yale School
of Medicine, New Haven, CT, USA

ABSTRACT ARTICLE HISTORY


Introduction: Coronavirus disease (COVID-19) can trigger a cytokine response storm (CRS) that is Received 7 August 2020
associated with high mortality but for which the underlying pathophysiology and diagnostics are not Accepted 28 September
yet well characterized. This review provides an overview of the underlying immune profile of COVID-19- 2020
related CRS as well as laboratory markers for acute diagnosis and chronic follow-up of patients with KEYWORDS
SARS-CoV-2 and CRS. SARS-CoV-2; cytokine-
Areas covered: Innate and acquired immune profiles in COVID-19-CRS, RNA-detection methods for SARS- release syndrome; COVID-19;
CoV-2 in the setting of CRS including factors that affect assay performance, serology for SARS-CoV-2 in the cytokines; diagnosis; RNA;
setting of CRS, and other biomarkers for COVID-19 will be discussed. sensitivity
Expert opinion: Studies support the implication of CRS in the pathogenesis, clinical severity and
outcome of COVID-19 through the production of multiple inflammatory cytokines and chemokines
from activated innate and adaptive immune cells. Although these inflammatory molecules, including IL-
6, IL-2 R, IL-10, IP-10 and MCP-1, often correlate with disease severity as possible biomarkers, the
pathogenic contributions of individual molecules and the therapeutic benefits of targeting them are yet
to be demonstrated. Detection of SARS-CoV-2 RNA is the gold standard method for diagnosis of COVID-
19 in the context of CRS but assay performance varies and is susceptible to false-negative results even
as patients clinically deteriorate due to decreased viral shedding in the setting of CRS. Biomarkers
including CRP, ferritin, D-dimer and procalcitonin may provide early clues about progression to CRS and
help identify thrombotic and infectious complications of COVID-19.

1. Introduction severe COVID-19 disease including acute respiratory distress


syndrome and septic shock with multi-organ failure.
The novel Coronavirus, SARS-CoV-2, was first linked to an
Development of CRS is strongly linked to increased morbid­
acute respiratory syndrome in December of 2019 after cases
ity and mortality [3]. Though certain clinical findings includ­
of respiratory failure were reported in Wuhan, China. Since
ing anosmia and ageusia in the context of an interstitial
then, the world had witnessed a global pandemic not seen
pneumonia are suggestive of COVID-19, no signs or symp­
since the Spanish Influenza. As of this writing, more than
toms are pathognomonic of infection. Similarly, epidemiolo­
200,000 people have succumbed to SARS-CoV-2 in the
gic factors such as increased age, clinical signs like rising
United States, compared to an estimated 675,000 from the
oxygen supplementation requirement and radiologic fea­
Spanish Influenza. The predominant clinical manifestations
tures like worsening pulmonary infiltrates are associated
of coronavirus disease (COVID-19) are respiratory including
with a greater risk for severe disease but remain imperfect
upper and lower respiratory tract symptoms but other sys­
markers of progression to CRS in individual patients [4].
tems such as the gastrointestinal tract, coagulation cascade,
A combination of clinical, epidemiologic and laboratory
vascular and central nervous systems are commonly
markers are needed to discriminate between patients who
involved [1]. Clinical and physiologic studies of COVID-19
are at risk of or in the course of progressing to the inflam­
have suggested a biphasic pattern of illness with an initial
matory phase of COVID-19. Determining whether patients
viral response phase lasting 7 to 10 days after symptom-
have progressed to CRS has important therapeutic implica­
onset characterized by rapid viral replication followed by an
tions, since anti-inflammatory agents are likely to be more
inflammatory response phase driven by innate and adaptive
useful than anti-virals in this setting [5,6]. In this paper, we
immune responses to the virus [2]. In a subset of indivi­
review the underlying innate and acquired immune profile
duals, immune dysregulation leads to a catastrophic mala­
of COVID-19-related CRS and discuss laboratory markers for
daptive hyperimmune response. This cytokine-release
acute diagnosis and chronic follow-up of patients with
syndrome (CRS), which is well described in the setting of
SARS-CoV-2 and CRS.
cancer immunotherapy, is associated with progression to

CONTACT Marwan M. Azar marwan.azar@yale.edu Department of Internal Medicine, Section of Infectious Diseases, Yale School of Medicine, New Haven,
CT 06510, USA
This article has been republished with minor changes. These changes do not impact the academic content of the article.
© 2020 Informa UK Limited, trading as Taylor & Francis Group
1088 M. M. AZAR ET AL.

of endothelial cells with capillary leakage, hypotension, coa­


Article highlights gulopathy, and augmented inflammation [22,23]. Patients with
● CRS is a systemic inflammatory response with multi-organ failure that
CRS can develop a hemophagocytic lymphohistiocytosis (HLH)
can be triggered by a variety of factors including infections, rheu­ or macrophage activation syndrome (MAS)-like syndrome with
matic diseases, and malignancies, resulting in the overactivation of the rise of other cytokines and chemokines including IL-8, IL-
immune cells, largely innate immune cells and T cells.
● In CRS, activated immune cells produce high levels of pro-
18, IP-10, MCP-1, MIG and MIP-1β [24]. Patients with COVID-19
inflammatory cytokines such as IL-1β, IL-6, TNF-α and IFN-γ, leading was found to have increased levels of inflammatory cytokines
to the activation of endothelial cells with capillary leakage, hypoten­ and chemokines, encompassing IL-1β, IL-1 receptor antagonist
sion, coagulopathy, and augmented inflammation.
● Patients with COVID-19 can develop CRS with increased levels of
(IL-1RA), IL-7, IL-8, IL-9, IL-10, G-CSF, GM-CSF, IFN-γ, IP-10, MCP-
inflammatory cytokines and chemokines, encompassing IL-1β, IL-1 1, MIP-1a, and MIP-1b, with some of them correlating with
receptor antagonist (IL-1RA), IL-7, IL-8, IL-9, IL-10, G-CSF, GM-CSF, disease severity (Table 1) [25].
IFN-γ, IP-10, MCP-1, MIP-1a, and MIP-1b, with some of them correlat­
ing with disease severity.
● CRS is associated with progression to severe COVID-19 disease includ­
ing acute respiratory distress syndrome and septic shock with multi- 2.1. Innate immunity-related cytokines in COVID-19
organ failure, with increased morbidity and mortality
● The clinical sensitivity of SARS-CoV-2 RNA testing is dependent on Up to 20% of COVID-19 cases present with severe disease,
several factors including assay analytical sensitivity, specimen type, with ARDS and multiple-organ failure driven largely by CRS
specimen quality, severity of disease and the timing of specimen
[26]. Both SARS-CoV and SARS-CoV-2 use the angiotensin-
collection
● Specimens obtained ‘too early’ prior to the ramping up of viral converting enzyme-
replication or ‘too late’ after viral replication had declined below related carboxypeptidase (ACE2) to gain entry to cells [27].
the testing assay’s limit of detection may lead to false-negative
This molecule is widely expressed not only in cardiopulmonary
results.
● RNA testing during CRS may lead to false-negative results since and gastrointestinal tissues but also in some hematopoietic
clinical deterioration due to immune dysregulation can coincide cells including innate immune monocytes, macrophages, and
with a decrease of viral replication
dendritic cells. SARS-CoV-2 infection of monocytes, macro­
● Persistent low level positive nucleic acid testing for weeks after initial
diagnosis likely do not indicate ongoing active infection phages, and dendritic cells infected leads to activation of
● The analytical sensitivity of commercial NAAT assays for SARS-CoV-2 pattern recognition receptors (PRR) and its downstream tran­
varies but a more accurate assessment is a comparison of assays’
scription factors such as NF-kB, leading to the production of
performance using parallel testing low positive clinical samples.
● Though generally not recommended for diagnosis of acute SARS-CoV an array of proinflammatory cytokines and chemokines [3].
-2 infection, serology may be useful when clinical suspicion for Probably, the most potent pro-inflammatory cytokines of
COVID-19 is high despite negative-repeated RNA testing, a scenario
innate immunity are IL-1β, IL-6 and TNF-α which are produced
which may occur in late disease including in the CRS phase.
● Several biomarkers including CRP, ferritin, D-dimer and procalcitonin mainly by monocytes, macrophages, mast cells, endothelial
may provide early clues about progression to CRS and help identify
thrombotic and infectious complications of COVID-19.
Table 1. Changes of cytokines and chemokines in COVID-19.
Cytokines Changes in COVID-19
IL-6 Increased circulatory levels in association with disease severity.
[23,26,27]
Patients with SpO2 < 90% had higher levels of IL-6 than
patients with SpO2 ≥ 90% [29].
2. Cytokine-release syndrome and COVID-19 TNF-α Increased circulatory levels in association with disease severity.
[23,26,27]
CRS is a systemic inflammatory response with multi-organ IL-1β Increased circulatory levels in association with disease severity.
failure that can be triggered by a variety of factors such as [26,27]
infections, rheumatic diseases, malignancies, and certain drugs IL-10 Increased circulatory levels. [23,48–50]
Patients with SpO2 < 90% had higher levels of IL-6 than
[7]. The term ‘cytokine release syndrome’ was first described in patients with SpO2 ≥ 90% [29].
the early ‘90s, when the anti-T-cell antibody muromonab-CD3 IFN-γ Increased circulatory levels in association with disease severity
(OKT3) was introduced as an immunosuppressive treatment [44] and viral loads [45]
Decreased frequency of IFN-γ-secreting CD4 + T cells in
for solid organ transplantation [8]. Subsequently, CRS has severe cases than in moderate cases [26]
been reported after infusion of chimeric antigen receptor IL-7 Patients requiring ICU admission displayed higher levels of
(CAR) T cells, antibody-based therapies, and administration of circulatory IL-7 compared to patients not requiring ICU
admission [23]
non-protein-based cancer drugs as well as in association with IL-13 Increased circulatory levels [23,48–50]
rheumatic diseases and severe viral infections, especially those IL-4 Increased circulatory levels. [23,48–50]
with pathogenic coronaviruses (CoV) including Middle East IL-2 Patients requiring ICU admission showed higher IL-2 level
compared to patients not requiring ICU admission [23]
respiratory syndrome coronavirus (MERS-CoV), severe acute Chemokines
respiratory syndrome coronavirus (SARS-CoV) and SARS-CoV IP-10 Patients requiring ICU admission had higher levels of IP-10
-2 [9–1617––18–19]. Indeed, the morbidity and mortality of (CXCL10) compared to patients not requiring ICU admission [23] in
association with disease severity [44]
COVID-19 are due primarily to excessive immune activation, MCP-1 Patients requiring ICU admission had higher levels of MCP-1
inflammation, and sepsis [20], with acute respiratory distress (CCL2) compared to patients not requiring ICU admission [23]
syndrome (ARDS) the proximate cause of death [21]. In CRS, MCP-3 Increased in association with disease severity [44]
(CCL7)
the overactivated immune cells, largely innate immune cells MCP-1α Patients requiring ICU admission had higher levels of MCP-1 α
and T cells, produce high levels of pro-inflammatory cytokines (CCL3) compared to patients not requiring ICU admission [23] in
such as IL-1β, IL-6, TNF-α and IFN-γ, leading to the activation association with disease severity [44]
EXPERT REVIEW OF MOLECULAR DIAGNOSTICS 1089

and/or epithelial cells [28,29]. CRS likely results from a sudden macrophages, and NK cells while Th2 and Th17 cells can
increase in pro-inflammatory cytokines with the influx of activate eosinophils and neutrophils via producing IL-5 and
immune cells, including macrophages, neutrophils, NK cells IL-17, respectively [48,49]. Regulatory T cells such as FOXP3
and T cells, from the circulation into the infected site [29]. In + regulatory CD4 + T cells can regulate early innate immune
addition to pro-inflammatory cytokines, growth factors and responses by suppressing inflammatory cytokine production,
chemokines such as vascular endothelial growth factor providing checks and balances between host defense and
(VEGF), monocyte chemoattractant protein-1 (MCP-1), and IL- inflammatory damage. Patients with COVID-19 appear to
8 can contribute to the destabilization of endothelial cells in have alterations in adaptive immunity which includes reduced
CRS, leading to the development of vascular barrier impair­ numbers of circulatory T cell numbers but increased circula­
ment and capillary damage with multiorgan failure [29]. tory levels of T cell cytokines [28,50]. Indeed, patients with
Patients with COVID-19 have higher circulatory levels of IL- COVID-19 have increased circulatory levels of the T cell cyto­
1β, IL-6, IL-8, IL-10, TNF-α compared to healthy controls, cor­ kines IL-2, IFN-γ, IL-17, and the soluble IL-2 receptor alpha
relating with disease severity [25,30,31]. For instance, levels of chain (IL-2 R or CD25), which is cleaved from the surface of
IL-6 and IL-10 in peripheral blood were higher in patients with activated T cells [25,30–32]. It is noticeable that innate
severe COVID-19 than in patients with mild COVID-19 [32]. In immune cytokines IL-1β, IL-6, and IL-12 can promote the
fact, patients with SpO2 < 90% had higher levels of IL-6 and IL- activation, differentiation, and cytokine production of T cells
10 than patients with SpO2 ≥ 90% [33]. Although SARS-CoV-2 with the expression of the receptors for these cytokines. IL-1β
viral RNA was detected in only 15% of COVID-19 patients, can enhance IL-17 production from CD4 + T cells, namely
these patients had elevated levels of IL-6 with poor clinical T helper (Th) 17 cells, while IL-12 induces IFN-γ-producing
prognosis [34]. A transcriptional and serological analysis of Th1 cells [51,52]. IFN-γ can further activate innate immune
COVID-19 patients showed increased levels of pro- cells like monocytes, macrophages, and natural killer cells
inflammatory cytokines, including IL-6, along with low type with the expression of IFN-γ receptor, resulting in augmented
I and III IFNs and high chemokine signature [35], suggesting production of cytokines and chemokines as a feed-forward
reduced antiviral defense coupled with dysregulated inflam­ mechanism. Such interface of T cells and innate immune
matory responses. The inflammatory cytokines IL-1β, IL-6, and cells can be mediated by chemokines like monocyte chemoat­
TNF-α account largely for the clinical manifestations, including tractant protein 1 (MCP1 or CCL2), MCP3 (CCL7) and macro­
fever, hyperferritinemia, acute phase reactions, and coagulo­ phage inflammatory protein 1 alpha (MCP-1α or CCL3)
pathies in COVID-19 given the known proinflammatory prop­ released from monocytes and macrophages [53]. IFN-γ can
erties of these molecules [36–37–38]. These findings suggest also induce the chemokine IFN-γ–induced protein 10 (IP-10
a possible pathologic role of IL-6 in COVID-19-associated CRS, or CXCL10) from monocytes, endothelial cells and fibroblasts,
raising the consideration of IL-6 blockade as in CRS due to which can recruit T cells, NK cells, and monocytes. In COVID-
other conditions like CAR-T therapy [39,40]. Indeed, IL-6 inhi­ 19, the expression levels of IFN-γ, IP-10, MCP-3, and MIP-1α
biting therapy has been employed in COVID-19 using the anti- were shown to be increased and highly associated with dis­
IL-6R antibodies tocilizumab and sarilumab as well as the anti- ease severity during disease progression [25,54]. IFN-γ level
IL-6 antibody siltuximab, with mixed results. Some early was also found to correlate with viral load and disease severity
uncontrolled, open-labeled, and retrospective observational in patients with COVID-19 [55], suggesting the relationship of
studies reported the possible clinical benefits of tocilizumab the viral burden with T cell activation. Th2 cells produce high
with the improvement of fevers and inflammatory markers as levels of IL-4, IL-5 and IL-13 with the capacity to promote
well as with a reduced risk of invasive mechanical ventilation tissue fibrosis [56] whereas IL-10 from monocytes, Th2 cells
or death in severe COVID-19 [5,41,42]. However, a recent and regulatory T cells can inhibit pro-inflammatory cytokines
phase 3 clinical trial of sarilumab in patients with COVID-19 [57]. Increased levels of IL-4, IL-10 and IL-13 were reported in
was discontinued after finding no benefit for preventing patients with COVID-19 compared to healthy controls [25,58–
deaths or getting off ventilation [43]. IL-6 deficient mice had 59–60]. In persistent viral infections, CD4+ and CD8+ T cells
impaired immunity to viral and intracellular bacterial infec­ lose their effector function, become unable to control viral
tions [44]. Patients with rheumatoid arthritis treated with toci­ infection, and secrete high levels of TGF-β and IL-10 [61].
lizumab developed a higher rate of serious infections Thus, elevated IL-10 and IL-13 in COVID-19 could further dam­
compared to those who received alternative biologics pen the effector function of T cells and promote tissue fibrosis.
[45,46]. In a pre-printed platform, a study reported an IL-7 with the capacity to promote T cell survival increases in
increased risk of secondary infections with higher mortality COVID-19 [25] although patients with this disease have
in critically ill COVID-19 patients who received tocilizumab decreased frequencies of lymphocytes, including CD4+ and
compared to patients who did not receive this drug [47]. CD8 + T cells [32,62]. The increased IL-7 could be driven in
part by lymphopenia in COVID-19 as lymphopenia is known to
increase IL-7 [63].
2.2. T cell immunity-related cytokines in COVID-19
In addition to innate immune cells, analysis of immune mole­
3. SARS-CoV-2 RNA detection for diagnosis of COVID
cules in blood indicates the activation of T cells in COVID-19.
in the setting of CRS
Activated T cells can further stimulate innate immune
responses by producing cytokines. T helper 1 (Th1) cells that The primary diagnostic method for acute COVID-19 infection
dominantly produce IFN-γ can activate monocytes, is measurement of SARS-CoV-2 RNA via nucleic acid testing,
1090 M. M. AZAR ET AL.

most commonly polymerase chain reaction (PCR). Several prolonged low-level viral shedding may not reflect live infec­
assays are available including from commercial laboratories tious virus. Regardless of other factors, poor sampling espe­
and the CDC (Table 2), with varying analytical sensitivities cially of upper respiratory specimens can lead to false-
among other parameters. The clinical sensitivity of RNA detec­ negative results.
tion is dependent on the amount of viral shedding in clinical
specimens. In COVID-19, viral replication begins 2–5 days
before symptom onset and peaks between 0.6 days to 3.1. Factors that influence SARS-CoV-2 RNA detection
5 days after symptom onset when measured in upper respira­ assay performance
tory tract specimens [64,65]. Testing done ‘too early’ i.e.
before viral loads increase beyond the assay’s limit of detec­ 3.1.1. Specimen collection, storage and transport
tion will lead to false-negative results (Figure 1). However, The pre-analytical phase of laboratory testing is the most
viral replication may peak prior to development of symptoms common source of laboratory errors and testing for SARS-
and high viral loads in the pre-symptomatic stage likely CoV-2 is no exception [74,75]. Regardless of the sample type,
account for a significant portion of transmissions, up to 44% adequate specimen collection is fundamental to accurate
in one mathematical model [64]. In one study, the median nucleic acid testing. Proper specimen collection is critical in
time from detection of SARS-CoV-2 to symptom onset in pre- obtaining accurate results and poor specimen collection can
symptomatic patients was 15 days [66]. After 7 days of illness, lead false negative tests. The recommended collection techni­
viral loads drop rapidly but may remain above the threshold que varies across specimen types. For example, nasopharyn­
of nucleic acid detection for weeks post-symptom onset. geal (NP) specimens require insertion of the swab tip through
Though viral loads peak early in the course of COVID-19, the nostril to the pharynx until resistance is encountered, after
they are higher and persist longer in severe disease. In which the swab should be left in place for a few seconds to
a study comparing mild and severe cases, the median and absorb secretions. Deep nasal or mid-turbinate samples are
peak viral loads were 1 log10 higher in the latter group [67]. collected by inserting a swab one inch into the nose, to the
Similarly, patients with severe COVID-19 exhibited higher viral level of the turbinates, rotating several times, then repeated in
loads and more prolonged shedding than mild cases (21 vs. the other nostril using the same swab. For anterior nasal
14 days) [68] suggesting that clinical sensitivity of PCR testing specimens, the swab should be inserted 0.5 inches inside
is higher in more severe COVID19. Notably, among patients each nostril and rotated along the nasal membrane 5 times,
with mild disease, the median viral load of patients with then repeated in the other nostril using the same swab. After
symptomatic and asymptomatic disease are not significantly collection, specimens must be stored in appropriate condi­
different [66]. It is important, however, to understand the viral tions. Instructions vary based on the transport medium
kinetics of SARS-CoV-2 infection in the context of the COVID- employed by different assays, but CDC has a general recom­
19 disease process in which severity of disease, particularly in mendation for storage at 2–8°C for up to 72 hours after
the latter stages appears to be driven by a cytokine storm collection and at ≤70°C if transport is not possible within
rather than purely by virus-related cytotoxicity (Figure 1). In 72 hours. Improper storage can accelerate nucleic acid degra­
a subset of individuals who clinically deteriorate 1–2 weeks dation especially for RNA which is significantly less stable than
after initial symptom onset, reflecting inflection from a viral DNA even in optimal conditions, leading to false-negative
response phase to a cytokine response phase, nucleic acid results. In addition, each laboratory must use appropriate pre-
testing is likely to be less sensitive despite clinical decompen­ analytical processes to reduce contamination between clinical
sation since viral loads are significantly lower at the inflam­ specimens. Both mid-turbinate and anterior nasal swabs can
matory stage of disease. Falsely negative PCR tests may be self-collected with instructions under the observation of
theoretically occur in this period (‘too late’), especially when a healthcare worker.
sampling upper respiratory tract specimens, which are less
sensitive than lower respiratory tract specimens (such as spu­ 3.1.2. Considerations for home collection
tum or endotracheal fluid) at any one point after symptom Home collection has several advantages including increased
onset [67]. Conversely, PCR testing may remain positive for convenience, decreased risk of exposing other patients and
more than 6 weeks after initial diagnosis, with a median of healthcare workers and decreased need for personal protec­
24 days in the general population. Prolonged shedding is tive equipment [76]. However, from a testing standpoint, these
more common with advanced age, certain comorbidities like approaches present unique challenges including an increased
diabetes and hypertension and immunocompromising condi­ likelihood of suboptimal specimen collection and an increased
tions [69,70]. Though viral RNA can be detected for weeks likelihood of specimen degradation during transport. The FDA
after symptom onset including during the cytokine response recommends against self-collected NP swabs for safety.
phase of illness, it is unclear whether persistently low viral Therefore, in addition to standard validation studies, the FDA
loads represent infectious virus. In a studies of patients with requires additional stability and usability studies for assays
COVID, viral culture was successful only from specimens col­ that employ home collection. Self-collection kits that facilitate
lected within of symptom onset or with viral loads ≥6 log and optimize collection (such as for saliva), instructional
copies/mL [71,72]. In another study, virus was successfully pamphlets and videos that clarify testing steps for laypersons
cultured only from upper respiratory specimens when the and testing of less invasive specimen like nasal swabs can
PCR cycle threshold was below 34, correlating to a low viral improve specimen collection and diagnostic accuracy. Home
load [73]. Taken together, these studies suggest that collection must be approved by the FDA and tests must
Table 2. Characteristics of a selection of commercial assays for detection of SARS-CoV-2 RNA.
Developer/ Reported
Manufacturer Assay Method Gene Targets Ct values LOD* Specimen Sample volume Features Assay time
Abbott Abbott RealTime SARS CoV-2 Real time RT- N, RdRp Yes 100 copies/ NP, OP, Nasal 500 µL Automated 3.5 hr
on m2000 system PCR mL High throughput
Abbott ID NOW COVID-19 Isothermal RdRP No 125 copies/ NP, Nasal, OP Dry nasal swab placed Manual 5–13
nucleic mL directly into the Sample Low Throughput min
acid Receiver (1 sample per run)
amplification Low skill level. Can be used
(NEAR) at Point of Care.
Becton Dickinson BD BioGX SARS-CoV-2 for BD Real time RT- N1, N2 Yes 40 copies/ NP, OP, Nasal, MT 750 μL Automated 3.5 hr
MAX system PCR mL Two duplex reactions
based on CDC assay
24 samples per batch
Centers for Disease CDC 2019-Novel Coronavirus Real time RT- N1, N2 (original Yes 1–3 NP, OP, Nasal, MT, BAL, TA, 140–200 µL into separate Manual 3.5 hr
Control and real-time RT-PCR PCR assay copies/ Sputum extraction Low throughput (designed
Prevention diagnostic panel included N3) µL as 3 singleplex PCR
reactions)
96-well plate
High skill level
Cepheid Xpert Xpress SARS CoV-2 Real time RT- N2, E Yes 250 copies/ NP, OP, Nasal, MT 300 μL Automated 45 min
PCR mL Random access
Inoculate single sample
into cartridge and place on
instrument
Diasorin SimplexaCOVID-19 Direct Real time RT- ORF1ab, S Yes 242 copies/ NP, Nasal, BAL 50 μL Automated 1.5 hr
PCR mL Batched
Inoculate 1–8 wells in
wheel
Hologic SARS CoV-2 Assay (Panther Real time RT- ORF1ab Yes 0.01 TCID50 NP, OP, nasal, MT, BAL 500 µL into lysis tube Automated 2.5 hr
Fusion) PCR regions 1 /mL Random access
and 2
Hologic Aptima SARS CoV-2 Assay Transcription ORF1ab No 0.01 TCID50 NP, OP, nasal, MT 500 μL into lysis tube or Automated 3.5 hr
(Panther System) mediated regions 1 /mL direct testing of Aptima High Throughput
amplification and 2 Multitest swab
(TMA)
Quest SARS-CoV-2 RNA, Qualitative Real time RT- N1, N3 Yes 136 copies/ NP, OP, Nasal, MT, BAL, TA, 200 µL into separate Batched testing 3.5 hr
Real-time RT-PCR PCR µL Sputum, home collected extraction Multiplex RT-PCR based on
nasal swabs CDC assay
96 well plate
Can test unusual samples
types
RP control required for
home collection
Roche Cobas SARS CoV-2 for use on Real time RT- ORF1ab, E Yes 0.009 NP, OP, nasal, MT 600 µL Automated 3.5 hr first
6800/8800 Systems PCR TCID50/ High throughput 96 then
mL 96 tests per run 1.5 hr
Thermofisher TaqPath COVID-19 Combo Real time RT- ORF1ab, N, S Yes but not 10 copies NP, OP, Nasal, MT, BAL 200 or 400 µL into separate Manual or automated 3.5 hr
Kit PCR readily /reaction extraction High Throughput
obtained 96 or 384 well plates
EXPERT REVIEW OF MOLECULAR DIAGNOSTICS

High skill level


*LOD varies per specimen type. Lowest LOD is included here. LOD is an in vitro assessment that may not correlate when tests are compared in parallel using clinical specimens.
RT-PCR, reverse transcription polymerase chain reaction.
Gene targets: N, nucleoprotein; RdRp, RNA dependent RNA polymerase; E, envelope; ORF1ab, open reading frame 1ab; S, spike.
LOD, limit of detection or lowest concentration of analyte that can be detected 95% of the time.
1091

NP, nasopharyngeal; OP: oropharyngeal; MT, mid-turbinate; BAL, bronchoalveolar lavage; TA, tracheal aspirate.
1092 M. M. AZAR ET AL.

ensure human material has been collected, such as including and transport media, nucleic acid extraction reagents and
PCR for human RNase P gene. buffers, plasticware including pipet tips, test kits and reagents,
and instrumentation. These shortages have led to a constant
struggle to quickly validate alternative materials and methods,
3.1.3. Sample type
the need to maintain multiple test platforms, and to compro­
As a general rule, deeper specimens yield greater sensitivity.
mise and sometimes accept less than optimal results.
Nucleic acid testing on nasopharyngeal specimens is overall
more sensitive than for other URT specimens with mid-
turbinate, anterior nasal and oropharyngeal specimens being 4. SARS-CoV-2 serology for diagnosis of COVID in
incrementally less sensitive [77]. The data are mixed, however the setting of CRS
with some studies showing mid-turbinate swab specimens to
After COVID-19, most individuals develop a serologic response
be equally sensitive to nasopharyngeal specimens [78].
to SARS-CoV-2, with a median time to IgG positivity of
Similarly, saliva is less or equally sensitive to nasopharyngeal
11–14 days depending on several factors [83,84]. Patients
specimens in most studies though saliva collection methods
with very mild disease and those with underlying immuno­
are inconsistent, making an accurate analysis difficult [79,80].
compromising conditions may not mount an antibody
Lower respiratory track (LRT) specimens such as sputum,
response whereas those with critical illness may have higher
endotracheal fluid and bronchoalveolar lavage fluid are more
and more sustained antibody titers [85]. False-negative testing
sensitive than upper respiratory tract (URT) specimens and
may occur with poorly sensitive assays (particularly lateral flow
should be collected if initial URT testing is negative but suspi­
assays), if testing is done prior to 11 days after symptom onset
cion remains moderate to high [67]. Though certain conclu­
or in very mild cases. Serology is not recommended for clinical
sions can be made from available studies, variability in study
diagnosis of SARS-CoV-2 infection as it is less sensitive than
designs, collection and test methodologies, patient popula­
RNA detection for acute infection, and the implications of
tions including demographics and clinical severity inject sig­
positive serology for either immunity or infectivity to others
nificant heterogeneity into results.
have not been sufficiently characterized. Though not routinely
recommended, serology may have a role in the clinical diag­
3.1.4. Sensitivity and specificity nosis of COVID in certain circumstances. These include diag­
The clinical sensitivity of SARS-CoV-2 nucleic acid testing is nosis of suspected late COVID-19 complications such as
defined as the ability of the test to correctly identify patients Multisystem Inflammatory Syndrome in Children without
with COVID-19. Clinical sensitivity for SARS-CoV-2 testing is prior positive PCR/RNA and suspected recent COVID-19 with
dependent on several factors including severity of disease, negative-repeated RNA testing despite high clinical suspicion,
specimen type, specimen quality and analytical sensitivity. a scenario that may occur in the setting of CRS (Figure 1) [86].
Clinical sensitivity also encompasses test result interpretation SARS-CoV-2 IgM antibodies peak within 3 weeks of illness
in a clinical context in order to differentiate acute infection onset with most patients seroconverting to IgG between
from colonization (for example, with Pneumocystis jiroveci) or week 3 and week 7. Since development of SARS-CoV-2 IgM
prolonged shedding (for example, with rhinovirus). In the case and potentially IgG, coincide with the cytokine response phase
of COVID-19, SARS-CoV-2 RNA detection can be prolonged of illness, serology has diagnostic utility in the diagnosis of
with a subset of patients shedding virus beyond 6 weeks COVID when PCR testing is negative but suspicion for COVID
after initial infection [69]. Therefore, positive SARS-CoV-2 test remains high [87] In a study of patients with COVID-19, serol­
result may indicate acute recent COVID-19 infection but could ogy was positive in 28–100% of patients with negative upper
also indicate prolonged shedding weeks or months after acute respiratory PCR testing suggesting clinical value in this setting.
infection. Since prolonged positivity may not indicate infec­ Moreover, the sensitivity of IgG was substantially higher than
tiousness, making this distinction is important clinically and for PCR (80% vs.46%) in patients who were in a later phase (15–­
infection prevention purposes [71–73]. 39 days) from symptom onset [84]. If initial testing with serol­
The analytical sensitivity of a SARS-CoV-2 nucleic acid test is ogy is negative, a convalescent sample should be obtained
its ability to detect the target analyte (SARS-CoV-2 RNA in this 2–4 weeks later to exclude delayed seroconversion.
case) in a biologic sample with the limit of detection (LOD)
being the concentration SARS-CoV-2 RNA in a specimen that
5. Laboratory markers of severe COVID and CRS
can be consistently detected ≥ 95% of the time. These mea­
sures vary across various laboratory-developed and commer­ Laboratory testing is central to both the initial diagnosis and
cial SARS-CoV-2 nucleic acid tests with most available assays follow-up of SARS-CoV-2 infection and can moreover provide
reporting an LOD from 150 to 1,000 genome copies/mL (Table early clues of progression to CRS. Laboratory findings asso­
2). However, a more accurate assessment is a comparison of ciated with COVID-19 include decreased lymphocyte counts,
assays’ performance using parallel testing of low positive clin­ platelet counts and albumin levels as well as elevated CRP,
ical samples. Recently, falsely negative NAAT testing has been ferritin, lactate dehydrogenase (LDH), transaminases,
reported with mutated strains of SARS-CoV-2 highlighting the D-dimer, procalcitonin and interleukin-6 and 10 levels
need for epidemiologic monitoring of circulating SARS-CoV-2 among others. In retrospective case series of 393 patients
viruses [81,82]. with COVID-19 in New York, lymphopenia was present in
It should be noted that supply chain disruptions have 90% of cases, elevated ferritin in 66%, elevated CRP in
affected all aspects of laboratory testing, including swabs 43%, elevated D-dimer in 36%, elevated alanine
EXPERT REVIEW OF MOLECULAR DIAGNOSTICS 1093

Figure 1. Viral, serologic and diagnostic correlates of SARS-CoV-2 Infection.


After SARS-CoV-2 acquisition, a subset of individuals (40–45%) will become symptomatic after a median incubation period of 5 days. Viral replication accelerates rapidly in upper respiratory
tract cells (viral replication phase), reaching a peak viral load between 0.6 days to 5 days after symptom onset, before beginning to decrease gradually. Some patients may develop clinical
worsening during viral load decline as a result of an overactive inflammatory response (cytokine response phase). Testing done too early (>~5 days before symptom onset) or too late
(>~21 days after symptom onset) in the course of the infection may yield false-negative PCR results as viral load may be below the threshold of detection of the assay (purple-shaded
areas). SARS-CoV-2-specific serum IgG develops at a median of 11 days after infection.

aminotransferase in 32% and elevated procalcitonin in 17% procalcitonin may not be sufficiently sensitive to rule out
[88]. In addition to aiding in the initial diagnosis of COVID- bacterial pneumonia as a stand-alone marker [94].
19, several biomarkers may serve as indicators of greater As with secondary bacterial infection, the development of
disease severity at baseline as well as markers of disease a systemic coagulopathy is now a well-recognized complica­
progression, often a clinical manifestation of developing tion of severe COVID-19. Increased concentrations of inflam­
CRS. CRP is perhaps the best predictor of COVID-19 severity. matory cytokines induce a prothrombotic cascade. In
Elevated CRP levels have been associated with more severe particular, IL-6 induces tissue factor expression on mono­
disease at baseline, more extensive lung infiltrates on ima­ nuclear cells driving coagulation activation and thrombin
ging and increasing CRP levels are a risk factor for COVID-19 generation [95]. Complications which include deep vein
progression [89,90]. Decreased lymphocytes counts and thrombosis (DVT), pulmonary embolism (PE), ischemic
increased LDH levels have also been associated with severe stroke, myocardial infarction and systemic arterial events
COVID-19 both as individual biomarkers and when inte­ have been reported in up to approximately 30% of patients
grated into severity prediction indexes [91,92]. Immune dys­ with COVID-19 in the intensive care unit [96]. The most
regulation is also manifested by higher neutrophil- common findings of COVID-19-related coagulopathy are ele­
lymphocyte-ratio, and lower percentages of monocytes, vated D-dimer, mild thrombocytopenia and prothrombin
eosinophils, and basophils. Moreover, decreased T-cells sub­ time prolongation. D-dimer, a product of fibrin degradation,
sets, particularly helper T cells have been observed with is a sensitive marker of venous thromboembolism (VTE).
severe disease [31]. However, increased D-dimer levels are often driven by
Though progressive respiratory failure is the main driver of underlying inflammatory processes including sepsis, severe
morbidity and mortality in COVID-19, superimposed processes COVID-19 and CRS. Since elevated D-dimer levels may be
including bacterial superinfection and coagulopathy are a manifestation of both increased disease severity and an
known complications. When significantly elevated procalcito­ increased risk of thrombosis, the discriminatory power for
nin levels are strongly suggestive of a superimposed bacterial VTE is reduced [97]. Accordingly, rising D-dimer levels, var­
infection [93]. Indeed, while viral infections trigger production ious institutionally determined D-dimer thresholds and clin­
of interferon-γ which inhibits production of procalcitonin, ical signs of VTE including sharp rises in oxygen
bacterial infections stimulate production of IL 1β, IL-6 and requirements have been employed as indications for institu­
tumor necrosis factor α which enhance the production and tion of therapeutic anticoagulation in the setting of severe
release of procalcitonin from extrathyroidal tissues. However, COVID-19 and CRS.
mildly elevated procalcitonin levels have been observed in
patients with severe COVID-19 and no evidence of bacterial
infection. In these cases, it is possible that elevated inflamma­ 6. Conclusion
tory cytokines in the context CRS-related immune dysregula­ Studies support the implication of CRS in the pathogenesis,
tion lead to increases procalcitonin synthesis [31]. This is clinical severity and outcome of COVID-19 through the elevated
consistent with findings from severe influenza, in which production of multiple inflammatory cytokines and chemokines
1094 M. M. AZAR ET AL.

from activated innate and adaptive immune cells. Although these and affordable molecular tests that can be performed at the
inflammatory molecules, including IL-6, IL-2 R, IL-10, IP-10 and point of care or at home are expected soon. Less sensitive
MCP-1, often correlate with disease severity as possible biomar­ methods such as rapid antigen tests, with results in 15-30 min
kers, the pathogenic contributions of individual molecules and and minimal equipment requirements, may also help to fill the
the therapeutic benefits of targeting them are yet to be demon­ gap in some settings such as frequent re-testing schemes.
strated. Detection of SARS-CoV-2 RNA is the gold standard While concerns that the virus may mutate and elude detection
method for diagnosis of COVID-19 in the context of CRS but are valid, the use of multiple viral targets in most assays
assay performance varies according to analytical and clinical provide some assurance of continued efficacy. As these multi­
characteristics. Biomarkers including CRP, ferritin, D-dimer and ple strategies to improve access and shorten time to diagnosis
procalcitonin may provide early clues about progression to CRS come to fruition, SARS CoV-2 infections should be more
and help identify thrombotic and infectious complications of quickly detected and controlled, thus facilitating the safe
COVID-19. reopening of society.

7. Expert Opinion Funding


Studies clearly indicate the rise of multiple inflammatory cyto­ This paper was not funded.
kines and chemokines in patients with COVID-19, possibly
contributing to the pathogenesis of this disease accompanied
by systemic inflammation, especially in the form of CRS. As Declaration of interest
these cytokines and chemokines form a highly interactive The authors have no relevant affiliations or financial involvement with any
network, often serving as a feed-forward mechanism, it organization or entity with a financial interest in or financial conflict with
the subject matter or materials discussed in the manuscript. This includes
would be challenging to identify the most significant
employment, consultancies, honoraria, stock ownership or options, expert
upstream driver(s) dictating systemic inflammatory responses testimony, grants or patents received or pending, or royalties.
in COVID-19. Such molecules could be produced from the
innate and/or adaptive immune cells in response initially to
SARS-CoV-2 and later to molecules released from damaged Reviewer disclosures
cells as damage-associated molecular patterns. High-dimen­ Peer reviewers on this manuscript have no relevant financial or other
sional cytokine analysis done in a prospective manner, relationships to disclose.
together with careful clinical data collection and computa­
tional integration of both biological and clinical data, may be
able to reveal the possible upstream inflammatory cytokine(s) References
accountable for CRS with systemic inflammation and ARDS, Papers of special note have been highlighted as either of interest (•) or of
the major mortality driving manifestations, in COVID-19. Such considerable interest (••) to readers.
an investigation could be achieved using recently developed 1. Zhu N, Zhang D, Wang W, et al. A novel coronavirus from patients
high-dimensional single cell analytic tools like single cell RNA with pneumonia in China, 2019. N Engl J Med. 2020 Feb 20;382
(8):727–733.
sequencing and mass cytometry in addition to cytokine multi­
2. Lescure FX, Bouadma L, Nguyen D, et al. Clinical and virological
plex. The results of this study can lead to the development of data of the first cases of COVID-19 in Europe: a case series. Lancet
a new therapeutic approach with an appropriate selection of Infect Dis. 2020 June;20(6):697–706.
cytokine blockade(s) and its introductory time point, hopefully 3. Moore JB, June CH. Cytokine release syndrome in severe COVID-19.
before the progress to the full-blown CRS and ARDS. The same Science. 2020May1;368(6490):473–474.
• This is a viewpoint article describing the pathophysiology of
cytokine(s) also can serve as a biomarker guiding additional
cytokine release syndrome and the possible roles of IL-6
therapeutic strategies such as timing of initiating certain treat­ antagonists as the treatment of patients with severe COVID-
ment modalities and predicting prognosis. 19.
Many sensitive NAAT have been developed, yet the labora­ 4. Guan W-J, Ni Z-Y, Hu Y, et al. Clinical characteristics of coronavirus
tory diagnosis of SARS CoV-2 remains a challenge due to disease 2019 in China. N Engl J Med. 2020 Apr 30;382
escalating demands that outpace testing capacity. Detection (18):1708–1720.
5. Price CC, Altice FL, Shyr Y, et al. Tocilizumab treatment for cytokine
of viral RNA is preferred, but access has been limited by release syndrome in hospitalized COVID-19 patients: survival and
unreliable supply chains, and inability to obtain collection clinical outcomes. Chest. 2020 June 15. DOI:10.1016/j.
swabs, instruments, reagents and supplies. Test backlogs at chest.2020.06.006
reference laboratories lead to delays of a week or more before • In this study of 239 patients with COVID, a treatment algo­
rithm that included tocilizumab to target CRS was associated
results are received. Due to staffing shortages, risk to person­
with decreased rates of mechanical ventilation and survival at
nel, and need for scarce PPE, sample collection has been 21-days.
inadequate. These challenges have led to alternative strate­ 6. Zhang C, Wu Z, Li JW, et al. Cytokine release syndrome in severe
gies, such as testing saliva, self-collection of nasal swabs, and COVID-19: interleukin-6 receptor antagonist tocilizumab may be
home collection. High-throughput genetic testing laboratories the key to reduce mortality. Int J Antimicrob Agents. 2020 May;55
(5):105954.
have adapted their methods to SARS CoV-2 screening. Novel
7. Shimabukuro-Vornhagen A, Godel P, Subklewe M, et al. Cytokine
technologies such as CRISPR, as well as pooling of samples to release syndrome. J Immunother Cancer. 2018 June 15;6(1):56.
increase throughput and reduce cost, have been implemented 8. Chatenoud L, Ferran C, Reuter A, et al. Systemic reaction to the
for the first time for diagnostic testing. Rapid, sensitive, simple, anti-T-cell monoclonal antibody OKT3 in relation to serum levels of
EXPERT REVIEW OF MOLECULAR DIAGNOSTICS 1095

tumor necrosis factor and interferon-gamma [corrected]. N Engl This prospective multicenter study showed that patients who
J Med. 1989 May 25;320(21):1420–1421. were admitted to the intensive care units (ICU) compared with
9. Aue G, Njuguna N, Tian X, et al. Lenalidomide-induced upregula­ those who did not require ICU admission, presented with
tion of CD80 on tumor cells correlates with T-cell activation, the higher plasma levels of IL-2, IL-7, IL-10, G-SCF, IP-10, MCP-1,
rapid onset of a cytokine release syndrome and leukemic cell MIP-1α, and TNFα.
clearance in chronic lymphocytic leukemia. Haematologica. 2009 26. Ge H, Wang X, Yuan X, et al. The epidemiology and clinical infor­
Sept;94(9):1266–1273. mation about COVID-19. Eur J Clin Microbiol Infect Dis. 2020
10. Cho C, Perales MA. Rapid identification of cytokine release syn­ June;39(6):1011–1019.
drome after haploidentical PBSC transplantation and successful 27. Hoffmann M, Kleine-Weber H, Schroeder S, et al. SARS-CoV-2 cell
therapy with tocilizumab. Bone Marrow Transplant. 2016 Dec;51 entry depends on ACE2 and TMPRSS2 and is blocked by a clinically
(12):1620–1621. proven protease inhibitor. Cell. 2020 Apr 16;181(2):271–280e8.
11. de Jong MD, Simmons CP, Thanh TT, et al. Fatal outcome of human 28. Coperchini F, Chiovato L, Croce L, et al. The cytokine storm in
influenza A (H5N1) is associated with high viral load and COVID-19: an overview of the involvement of the chemokine/che­
hypercytokinemia. Nat Med. 2006 Oct;12(10):1203–1207. mokine-receptor system. Cytokine Growth Factor Rev. 2020
12. de Vos S, Forero-Torres A, Ansell SM, et al. A phase II study of June;53:25–32.
dacetuzumab (SGN-40) in patients with relapsed diffuse large B-cell 29. Ragab D, Salah Eldin H, Taeimah M, et al. The COVID-19 cytokine
lymphoma (DLBCL) and correlative analyses of patient-specific storm; what we know so far. Front Immunol. 2020;11:1446.
factors. J Hematol Oncol. 2014 June 12;7:44. 30. Chen G, Wu D, Guo W, et al. Clinical and immunological features of
13. Freeman CL, Morschhauser F, Sehn L, et al. Cytokine release in severe and moderate coronavirus disease 2019. J Clin Invest. 2020
patients with CLL treated with obinutuzumab and possible rela­ May 1;130(5):2620–2629.
tionship with infusion-related reactions. Blood. 2015 Dec 10;126 31. Qin C, Zhou L, Hu Z, et al. Dysregulation of immune response in
(24):2646–2649. patients with COVID-19 in Wuhan, China. Clin Infect Dis. 2020 Mar
14. Pihusch R, Holler E, Muhlbayer D, et al. The impact of antithymo­ 12;71(15):762–768.
cyte globulin on short-term toxicity after allogeneic stem cell 32. Liu J, Li S, Liu J, et al. Longitudinal characteristics of lymphocyte
transplantation. Bone Marrow Transplant. 2002 Sept;30(6):347–354. responses and cytokine profiles in the peripheral blood of
15. Rotz SJ, Leino D, Szabo S, et al. Severe cytokine release syndrome SARS-CoV-2 infected patients. EBioMedicine. 2020 May;55:102763.
in a patient receiving PD-1-directed therapy. Pediatr Blood Cancer. 33. Wang Z, Yang B, Li Q, et al. Clinical features of 69 cases with
2017 Dec;64(12). DOI:10.1002/pbc.26642 coronavirus disease 2019 in Wuhan, China. Clin Infect Dis. 2020
16. Suntharalingam G, Perry MR, Ward S, et al. Cytokine storm in Mar 16;71(15):769–777.
a phase 1 trial of the anti-CD28 monoclonal antibody TGN1412. 34. Chen X, Zhao B, Qu Y, et al. Detectable serum SARS-CoV-2 viral load
N Engl J Med. 2006 Sept 7;355(10):1018–1028. (RNAaemia) is closely correlated with drastically elevated interleu­
17. Wing MG, Moreau T, Greenwood J, et al. Mechanism of first-dose kin 6 (IL-6) level in critically ill COVID-19 patients. Clin Infect Dis.
cytokine-release syndrome by CAMPATH 1-H: involvement of CD16 2020 Apr 17.
(FcgammaRIII) and CD11a/CD18 (LFA-1) on NK cells. J Clin Invest. 35. Blanco-Melo D, Nilsson-Payant BE, Liu WC, et al. Imbalanced host
1996 Dec 15;98(12):2819–2826. response to SARS-CoV-2 drives development of COVID-19. Cell.
18. Winkler U, Jensen M, Manzke O, et al. Cytokine-release syndrome in 2020 May 28;181(5):1036–1045e9.
patients with B-cell chronic lymphocytic leukemia and high lym­ 36. Dinarello CA. Immunological and inflammatory functions of the
phocyte counts after treatment with an anti-CD20 monoclonal interleukin-1 family. Annu Rev Immunol. 2009;27:519–550.
antibody (rituximab, IDEC-C2B8). Blood. 1999 Oct 1;94 37. Kishimoto T. IL-6: from its discovery to clinical applications.
(7):2217–2224. Int Immunol. 2010 May;22(5):347–352.
19. Channappanavar R, Perlman S. Pathogenic human coronavirus 38. Sethi G, Sung B, Aggarwal BB. TNF: a master switch for inflamma­
infections: causes and consequences of cytokine storm and tion to cancer. Front Biosci. 2008;13:5094–5107.
immunopathology. Semin Immunopathol. 2017 July;39 39. Behrens EM, Koretzky GA. Review: cytokine storm syndrome: look­
(5):529–539. ing toward the precision medicine era. Arthritis Rheumatol. 2017
20. Winthrop KL. Who needs a Corona? Arthritis Rheumatol. June;69(6):1135–1143.
2020;72:874–876. In Press. 40. Murthy H, Iqbal M, Chavez JC, et al. Cytokine release syndrome:
21. Ruan Q, Yang K, Wang W, et al. Clinical predictors of mortality due current perspectives. Immunotargets Ther. 2019;8:43–52.
to COVID-19 based on an analysis of data of 150 patients from 41. Sciascia S, Apra F, Baffa A, et al. Pilot prospective open, single-arm
Wuhan, China. Intensive Care Med. 2020 Mar 3;46:846–848. multicentre study on off-label use of tocilizumab in patients with
• This retrospective multicenter study showed that predictors of severe COVID-19. Clin Exp Rheumatol. 2020 May-June;38
a fatal outcome of COVID-19 cases included significantly (3):529–532.
higher IL-6 and CRP levels in the blood, older age and the 42. Guaraldi G, Meschiari M, Cozzi-Lepri A, et al. Tocilizumab in patients
prsence of secondary infection. with severe COVID-19: a retrospective cohort study. Lancet
22. Hay KA, Hanafi LA, Li D, et al. Kinetics and biomarkers of severe Rheumatol. 2020;2:e474-e484.
cytokine release syndrome after CD19 chimeric antigen 43. Sanofi and Regeneron provide update on Kevzara (sarilumab)
receptor-modified T-cell therapy. Blood. 2017 Nov 23;130 Phase 3 U.S. trial in COVID-19 patients. SANOFI Press Release; 2020.
(21):2295–2306. 44. Kopf M, Baumann H, Freer G, et al. Impaired immune and
23. Matthys P, Dillen C, Proost P, et al. Modification of the anti-CD3- acute-phase responses in interleukin-6-deficient mice. Nature.
induced cytokine release syndrome by anti-interferon-gamma or 1994 Mar 24;368(6469):339–342.
anti-interleukin-6 antibody treatment: protective effects and bipha­ 45. Lang VR, Englbrecht M, Rech J, et al. Risk of infections in rheuma­
sic changes in blood cytokine levels. Eur J Immunol. 1993 Sept;23 toid arthritis patients treated with tocilizumab. Rheumatology
(9):2209–2216. (Oxford). 2012 May;51(5):852–857.
24. Teachey DT, Lacey SF, Shaw PA, et al. Identification of predictive 46. Pawar A, Desai RJ, Solomon DH, et al. Risk of serious infections in
biomarkers for cytokine release syndrome after chimeric antigen tocilizumab versus other biologic drugs in patients with rheuma­
receptor T-cell therapy for acute lymphoblastic leukemia. Cancer toid arthritis: a multidatabase cohort study. Ann Rheum Dis. 2019
Discov. 2016 June 6;6:664–679. Apr;78(4):456–464.
25. Huang C, Wang Y, Li X, et al. Clinical features of patients infected 47. Kimmig LM, Wu D, Gold M, et al. IL6 inhibition in critically ill
with 2019 novel coronavirus in Wuhan, China. Lancet. 2020 Feb COVID-19 patients is associated with increased secondary
15;395(10223):497–506. infections. medRxiv. 2020 July 6.
1096 M. M. AZAR ET AL.

48. Palm NW, Medzhitov R. Not so fast: adaptive suppression of innate confirmed cases in Wuhan, China. Eur Urol. 2020 June;77
immunity. Nat Med. 2007 Oct;13(10):1142–1144. (6):748–754.
49. Kim KD, Zhao J, Auh S, et al. Adaptive immune cells temper initial 71. Wolfel R, Corman VM, Guggemos W, et al. Virological assessment of
innate responses. Nat Med. 2007 Oct;13(10):1248–1252. hospitalized patients with COVID-2019. Nature. 2020 May;581
50. Chau AS, Weber AG, Maria NI, et al. The longitudinal immune (7809):465–469.
response to coronavirus disease 2019: chasing the cytokine • In this study of nine patients with COVID, infectious virus was
storm. Arthritis Rheumatol. 2020 Sept 15. DOI:10.1002/art.41526 isolated from respiratory samples but not from stool samples
51. Lee WW, Kang SW, Choi J, et al. Regulating human Th17 cells via only within 8 days of symptom onset, suggesting that pro­
differential expression of IL-1 receptor. Blood. 2010 Jan 21;115 longed PCR positivity may not reflect viable virus.
(3):530–540. 72. Bullard J, Dust K, Funk D, et al. Predicting infectious SARS-CoV-2 from
52. Lee N, Shin MS, Kang I. T-cell biology in aging, with a focus on lung diagnostic samples. Clin Infect Dis. 2020 May 22. DOI:10.1093/cid/
disease. J Gerontol A Biol Sci Med Sci. 2012 Mar;67(3):254–263. ciaa638
53. Griffith JW, Sokol CL, Luster AD. Chemokines and chemokine recep­ 73. La Scola B, Le Bideau M, Andreani J, et al. Viral RNA load as
tors: positioning cells for host defense and immunity. Annu Rev determined by cell culture as a management tool for discharge
Immunol. 2014;32:659–702. of SARS-CoV-2 patients from infectious disease wards. Eur J Clin
54. Yang Y, Shen C, Li J, et al. Plasma IP-10 and MCP-3 levels are highly Microbiol Infect Dis. 2020 June;39(6):1059–1061.
associated with disease severity and predict the progression of 74. Lippi G, Simundic AM, European Federation for Clinical C, et al. The
COVID-19. J Allergy Clin Immunol. 2020 Apr 29;146:119–127.e4. EFLM strategy for harmonization of the preanalytical phase. Clin
55. Wei LL, Wang WJ, Chen DX, et al. Dysregulation of the immune Chem Lab Med. 2018 Sept 25;56(10):1660–1666.
response affects the outcome of critical COVID-19 patients. J Med 75. Lippi G, Simundic AM, Plebani M. Potential preanalytical and ana­
Virol. 2020 June 16. DOI:10.1002/jmv.26181 lytical vulnerabilities in the laboratory diagnosis of coronavirus
56. Hashimoto S, Gon Y, Takeshita I, et al. IL-4 and IL-13 induce disease 2019 (COVID-19). Clin Chem Lab Med. 2020 June 25;58
myofibroblastic phenotype of human lung fibroblasts through (7):1070–1076.
c-Jun NH2-terminal kinase-dependent pathway. J Allergy Clin 76. McCulloch DJ, Kim AE, Wilcox NC, et al. Comparison of unsuper­
Immunol. 2001 June;107(6):1001–1008. vised home self-collected midnasal swabs with clinician-collected
57. Wang S, Gao X, Shen G, et al. Interleukin-10 deficiency impairs nasopharyngeal swabs for detection of SARS-CoV-2 infection. JAMA
regulatory T cell-derived neuropilin-1 functions and promotes Th1 Netw Open. 2020 July 1;3(7):e2016382.
and Th17 immunity. Sci Rep. 2016 Apr 14;6(1):24249. 77. Hanson KE, Caliendo AM, Arias CA, et al. Infectious diseases society
58. Wong CK, Lam CW, Wu AK, et al. Plasma inflammatory cytokines of america guidelines on the diagnosis of COVID-19. Clin Infect Dis.
and chemokines in severe acute respiratory syndrome. Clin Exp 2020 June 16. DOI:10.1093/cid/ciaa1343
Immunol. 2004 Apr;136(1):95–103. 78. Tu YP, Jennings R, Hart B, et al. Swabs collected by patients or
59. Han H, Ma Q, Li C, et al. Profiling serum cytokines in COVID-19 health care workers for SARS-CoV-2 testing. N Engl J Med. 2020
patients reveals IL-6 and IL-10 are disease severity predictors. June 3;383:494–496.
Emerg Microbes Infect. 2020 Dec 9;(1):1123–1130. DOI:10.1080/ 79. Pasomsub E, Watcharananan SP, Boonyawat K, et al. Saliva sample
22221751.2020.1770129 as a non-invasive specimen for the diagnosis of coronavirus disease
60. Alexandra N, Donlan MY, William A, et al. IL-13 predicts the need 2019: a cross-sectional study. Clin Microbiol Infect. 2020 May 15.
for mechanical ventilation in COVID-19 patients. medRxiv. 2020 80. Jamal AJ, Mozafarihashjin M, Coomes E, et al. Sensitivity of naso­
June 20. pharyngeal swabs and saliva for the detection of severe acute
61. Rojas JM, Avia M, Martin V, et al. IL-10: a multifunctional cytokine in respiratory syndrome coronavirus 2 (SARS-CoV-2). Clin Infect Dis.
viral infections. J Immunol Res. 2017;2017:6104054. 2020 June 25. DOI:10.1093/cid/ciaa848
62. Mathew D, Giles JR, Baxter AE, et al. Deep immune profiling of 81. Artesi M, Bontems S, Gobbels P, et al. A recurrent mutation at position
COVID-19 patients reveals distinct immunotypes with therapeutic 26,340 of SARS-CoV-2 is associated with failure of the E-gene qRT-PCR
implications. Science. 2020 July 15;369:eabc8511. utilized in a commercial dual-target diagnostic assay. J Clin Microbiol.
63. Fry TJ, Mackall CL. The many faces of IL-7: from lymphopoiesis to 2020 July 20. 58. DOI:10.1128/JCM.01598-20
peripheral T cell maintenance. J Immunol. 2005 June 1;174 • This study reports on 8 patients with COVID with failure of
(11):6571–6576. cobas® SARS-CoV-2 E-gene qRT-PCR which was linked to
64. He X, Lau EHY, Wu P, et al. Temporal dynamics in viral shedding a mutated strain of SARS-CoV-2 due to C-to-U transition at
and transmissibility of COVID-19. Nat Med. 2020 May;26 position 26,340.
(5):672–675. 82. Vogels CBF, Brito AF, Wyllie AL, et al. Analytical sensitivity and
65. Zou L, Ruan F, Huang M, et al. SARS-CoV-2 viral load in upper efficiency comparisons of SARS-CoV-2 RT-qPCR primer-probe sets.
respiratory specimens of infected patients. N Engl J Med. 2020 Nat Microbiol. 2020 July 10;5:1299–1305.
Mar 19;382(12):1177–1179. 83. Long QX, Liu BZ, Deng HJ, et al. Antibody responses to
66. Lee S, Kim T, Lee E, et al. Clinical course and molecular viral SARS-CoV-2 in patients with COVID-19. Nat Med. 2020 June;26
shedding among asymptomatic and symptomatic patients with (6):845–848.
SARS-CoV-2 infection in a community treatment center in the 84. Zhao J, Yuan Q, Wang H, et al. Antibody responses to SARS-CoV-2
Republic of Korea. JAMA Intern Med. 2020 Aug 6. DOI:10.1001/ in patients of novel coronavirus disease 2019. Clin Infect Dis. 2020
jamainternmed.2020.3862 Mar 28. DOI:10.1093/cid/ciaa344
67. To KK, Tsang OT, Leung WS, et al. Temporal profiles of viral load in • In this study of 173 PCR-confirmed COVID-19 inpatients in
posterior oropharyngeal saliva samples and serum antibody Shenzhen, China, SARS-CoV-2 serology was detectable in
responses during infection by SARS-CoV-2: an observational cohort 28–100% of patients with negative PCR testing and the sensi­
study. Lancet Infect Dis. 2020 May;20(5):565–574. tivity of total SARS-CoV-2 antibodies in the later phase of
68. Zheng S, Fan J, Yu F, et al. Viral load dynamics and disease severity disease (day 15–39 since onset), was 100% compared to
in patients infected with SARS-CoV-2 in Zhejiang province, China, 45.5% for PCR suggesting utility of serology for diagnosis of
January-March 2020: retrospective cohort study. BMJ. 2020 Apr suspected late cases of PCR-negative COVID.
21;369:m1443. 85. Goetz L, Yang J, Greene W, et al. A COVID-19 patient with repeat­
69. Xiao AT, Tong YX, Zhang S. Profile of RT-PCR for SARS-CoV-2: edly undetectable SARS-CoV-2 antibodies. J Appl Lab Med. 2020
a preliminary study from 56 COVID-19 patients. Clin Infect Dis. Aug 3. DOI:10.1093/jalm/jfaa137
2020 Apr 19. DOI:10.1093/cid/ciaa460 86. Feldstein LR, Rose EB, Horwitz SM, et al. Multisystem inflammatory
70. Zhu L, Gong N, Liu B, et al. Coronavirus disease 2019 pneumonia in syndrome in U.S. children and adolescents. N Engl J Med. 2020
immunosuppressed renal transplant recipients: a summary of 10 June 29;383:334–346.
EXPERT REVIEW OF MOLECULAR DIAGNOSTICS 1097

87. Xiao AT, Gao C, Zhang S. Profile of specific antibodies to SARS-CoV- 93. Lippi G, Plebani M. Procalcitonin in patients with severe corona­
2: the first report. J Infect. 2020 July;81(1):147–178. virus disease 2019 (COVID-19): a meta-analysis. Clin Chim Acta.
88. Goyal P, Choi JJ, Pinheiro LC, et al. Clinical characteristics of Covid-19 in 2020 June;505:190–191.
New York City. N Engl J Med. 2020 June 11;382(24):2372–2374. 94. Pfister R, Kochanek M, Leygeber T, et al. Procalcitonin for diagnosis of
89. Wang L. C-reactive protein levels in the early stage of COVID-19. bacterial pneumonia in critically ill patients during 2009 H1N1 influenza
Med Mal Infect. 2020 June;50(4):332–334. pandemic: a prospective cohort study, systematic review and individual
90. Tan C, Huang Y, Shi F, et al. C-reactive protein correlates with patient data meta-analysis. Crit Care. 2014 Mar 10;18(2):R44.
computed tomographic findings and predicts severe COVID-19 95. Levi M, Thachil J, Iba T, et al. Coagulation abnormalities and
early. J Med Virol. 2020 July;92(7):856–862. thrombosis in patients with COVID-19. Lancet Haematol. 2020
91. Hou W, Zhang W, Jin R, et al. Risk factors for disease progression in June 7;(6):e438–e440. DOI:10.1016/S2352-3026(20)30145-9
hospitalized patients with COVID-19: a retrospective cohort study. 96. Becker RC. COVID-19 update: Covid-19-associated coagulopathy.
Infect Dis (Lond). 2020 July;52(7):498–505. J Thromb Thrombolysis. 2020 July;50(1):54–67.
92. Ji D, Zhang D, Xu J, et al. Prediction for progression risk in patients 97. Zhang L, Yan X, Fan Q, et al. D-dimer levels on admission to predict
with COVID-19 pneumonia: the CALL score. Clin Infect Dis. 2020 in-hospital mortality in patients with Covid-19. J Thromb Haemost.
Apr 9;71:1393–1399. 2020 June;18(6):1324–1329.

You might also like