You are on page 1of 24

Analytic Review

Journal of Intensive Care Medicine


2021, Vol. 36(1) 18-41
Evidence-Based Management of the Critically ª The Author(s) 2020
Article reuse guidelines:
sagepub.com/journals-permissions
Ill Adult With SARS-CoV-2 Infection DOI: 10.1177/0885066620969132
journals.sagepub.com/home/jic

Raghu R. Chivukula, MD, PhD1,2, Jason H. Maley, MD1,


David M. Dudzinski, MD3,4 , Kathryn Hibbert, MD1,
and C. Corey Hardin, MD, PhD1

Abstract
Human infection by the novel viral pathogen SARS-CoV-2 results in a clinical syndrome termed Coronavirus Disease 2019
(COVID-19). Although the majority of COVID-19 cases are self-limiting, a substantial minority of patients develop disease severe
enough to require intensive care. Features of critical illness associated with COVID-19 include hypoxemic respiratory failure,
acute respiratory distress syndrome (ARDS), shock, and multiple organ dysfunction syndrome (MODS). In most (but not all)
respects critically ill patients with COVID-19 resemble critically ill patients with ARDS due to other causes and are optimally
managed with standard, evidence-based critical care protocols. However, there is naturally an intense interest in developing
specific therapies for severe COVID-19. Here we synthesize the rapidly expanding literature around the pathophysiology, clinical
presentation, and management of COVID-19 with a focus on those points most relevant for intensivists tasked with caring for
these patients. We specifically highlight evidence-based approaches that we believe should guide the identification, triage,
respiratory support, and general ICU care of critically ill patients infected with SARS-CoV-2. In addition, in light of the pressing
need and growing enthusiasm for targeted COVID-19 therapies, we review the biological basis, plausibility, and clinical evidence
underlying these novel treatment approaches.

Keywords
coronavirus, SARS-CoV-2, COVID-19, pandemic, critical care, acute respiratory failure, acute respiratory distress syndrome
(ARDS), hypoxemia

Since the first reports of a novel viral respiratory illness Bronchoalveolar lavage samples from 3 affected patients with
emerged in December 2019 in Hubei Province, China,1 the pneumonia revealed the presence of a previously unidentified
illness has spread to pandemic proportions—infecting tens of viral sequence, “2019-nCoV,” aligning to lineage B of the
millions of individuals on all populated continents and respon- betacoronavirus (Beta-CoV) family and highly homologous
sible for over a million deaths.2 Both professional society to a bat coronavirus, CoV ZXC21.9 Existing Beta-CoV family
guidelines3 and individual approaches4-7 for the care of patients members, namely SARS-CoV and MERS-CoV, had previously
with what became known as Coronavirus Disease 2019 been linked to epidemic respiratory viral infections (SARS and
(COVID-19) were rapidly promulgated. These early efforts MERS, respectively), strongly suggesting a causal role for this
naturally preceded the wealth of clinical and basic science novel agent in disease pathophysiology. Further sequence
data8 which have since become available regarding the patho-
physiology and clinical management of COVID-19. In this
1
review, we synthesize what is now known about COVID-19 Division of Pulmonary and Critical Care Medicine, Department of Medicine,
critical illness into a consensus, evidence-based approach Massachusetts General Hospital, Boston, MA, USA
2
Whitehead Institute for Biomedical Research, Cambridge, MA, USA
informed by the pre-existing critical care literature, emerging 3
Corrigan Minehan Heart Center, Division of Cardiology, Department
basic biology,9-11 multiple observational reports,12-18 and of Medicine, Massachusetts General Hospital, Boston, MA, USA
recently published randomized controlled trial data.19,20 4
Cardiac Intensive Care Unit, Division of Cardiology, Department of Medicine,
Massachusetts General, Hospital, Boston, MA, USA

Epidemiology, and Pathophysiology Received April 10, 2020. Received revised September 14, 2020. Accepted
of SARS-CoV-2 October 07, 2020.

Epidemiology and Transmission Corresponding Author:


C. Corey Hardin, MD, PhD, Massachusetts General Hospital, Division of Pul-
Reports of a respiratory illness among individuals linked to an monary and Critical Care Medicine, 55 Fruit Street, Boston, MA 02114, USA.
indoor seafood market in Wuhan first arose in late 2019.1 Email: charles.hardin@mgh.harvard.edu
Chivukula et al 19

analysis established 2019-nCoV as most similar to SARS-CoV among care providers: droplet-transmitted pathogens spread
among previously identified coronaviruses; it was therefore via close, relatively extended contact with an infected person
renamed “SARS-CoV-2” and the disease that it causes in whereas airborne-transmitted pathogens can remain in the
humans was termed Coronavirus Disease 2019, abbreviated environment for hours36 and are far more easily transmissible.
COVID-19.1 Although potent, specific therapies against SARS-CoV-2
Early analyses of disease transmission estimated the remain elusive to date, a large number of candidate drug thera-
SARS-CoV-2 basic reproductive number (R0) between 2.0 and pies targeting the viral life cycle are under investigation in
2.5, indicating that an average infected individual would infect pre-clinical and clinical studies and one agent (remdesivir) has
*2 others and thereby propagate disease.21,22 Given a modest been approved for clinical use.41 Here we briefly review key
case fatality rate under 5%23 (versus, for example, MERS’ of steps in viral pathophysiology (Figure 1) in order to place these
>25%24) these data raised concern for the possibility of wide- therapies (addressed later) in context.
spread dissemination within and outside of China. In the Whether encountered directly in respiratory secretions or
approximately 7 months following the initial report and char- via an intermediate fomite, SARS-CoV-2 enters cells of the
acterization of COVID-19, the illness has indeed spread to host respiratory tract by way of a viral envelope protein (spike
pandemic proportions.25 Given the challenges in case identifi- protein, “S”) that engages cell-surface receptors. 1 The
cation these data are almost certainly substantial underesti- receptor-binding region of SARS-CoV-2 S protein is nearly
mates, highlighting the pressing need for physical distancing identical to SARS-CoV,10 which has previously been estab-
and other public health responses in curbing COVID-19 lished to engage at least 2 cell surface transmembrane pro-
spread.26 Even if such efforts are successful, however, the bur- teins—angiotensin-converting enzyme 2 (ACE2) 42 and
den of disease already present worldwide guarantees a surge in CD209 L (L-SIGN).43 Recent work has definitively established
intensive care utilization—a phenomenon already experienced that ACE2 can engage the SARS-CoV-2 S protein as well11 but
in areas with high infection burden27 and which demands whether this is the only relevant receptor in vivo remains
familiarity among intensivists with identifying and managing unclear and the observed pattern of affected tissues44 is not
this infection and its sequelae. entirely consistent with the known ACE2 expression domain.45
Following receptor binding, coronaviruses may enter host
cells through either of 2 mechanisms: (1) direct fusion or
Virology and Pathophysiology (2) receptor-mediated endocytosis46 (Figure 1). In the former
An appreciation of SARS-CoV-2 life cycle and pathobiology is case, one or more host cell surface proteases cleave the S protein,
worthwhile for the practicing clinician, particularly in under- driving fusion of the viral envelope with host cell membrane and
standing the basis for therapies currently in development and delivering the naked virus to the cytoplasm.11 In the latter case,
trials. Like other betacoronaviruses, SARS-CoV-2 is an envel- the entire virion is internalized into an “endosome” from which
oped, positive-strand RNA virus28,29 and, as for SARS-CoV it must escape in a lysosome-dependent process.47
and MERS-CoV, is transmitted primarily through respiratory Once in the cell, SARS-CoV-2 replicates its genome using a
droplets harboring intact virions.30 Unlike SARS, however, virally-encoded RNA-dependent RNA polymerase (RdRp) and
multiple reports indicate the potential for viral transmission hijacks host cell translation machinery to produce viral poly-
from currently asymptomatic individuals and even those in proteins required for virion assembly.48 These polyproteins are
whom symptoms never develop.31,32 In addition to respiratory cleaved by a virally encoded protease (CoV Mpro),49 after
droplets, SARS-CoV-2 exhibits laboratory stability in aerosols which viral assembly can occur. Infected cells transport assem-
(half-life *1 hour) and on a wide variety of inorganic sur- bled virions to the cell surface, where they are released through
faces.33,34 Epidemiologic investigations to date indicate that exocytosis to infect other host cells. As for SARS-CoV before
droplet and contact transmission are the major routes of trans- it, the specific mechanisms that account for the ability of
mission of SARS-CoV-2,30,35 in line with the longstanding SARS-CoV-2 to cause severe disease in humans remain poorly
recognition that persistence in aerosols is but one parameter understood. Early lung pathology reports50-53 from biopsy and
required for airborne transmission.36 Accordingly, true air- autopsy specimens indicate (as for SARS54 and MERS55) dif-
borne transmission is rare, having been firmly established for fuse alveolar damage (DAD) with alveolar desquamation, hya-
only a very small number of pathogens.36 Nevertheless, it line membranes, and variable degrees of viral cytopathic
remains possible that special circumstances may allow changes—overall consistent with viral-induced acute respira-
droplet-transmitted pathogens such as SARS-CoV-2 to traverse tory distress syndrome (ARDS). In patients who recover, a
larger times and distances as in airborne transmission.37 Some polyclonal humoral (antibody) and cellular immune response
reports during the SARS outbreak indicated that such permis- typically develops with evidence of neutralizing antibodies.9,56-58
sive airborne transmission may have occurred,38 while others The duration and quality of immunity provided by those antibo-
suggested transmission was exclusively by the droplet route.39 dies against re-infection remains to be comprehensively charac-
As discussed in following sections, the precise contribution of terized, though analogous responses in nonhuman primates
droplets, direct contact, aerosols, and fomites40 in driving confer protection against SARS-CoV-2 re-challenge.59 As dis-
SARS-CoV-2 spread remains a topic of intense debate, with cussed in detail below, specific targeted therapies intended to
attendant implications for contact and respiratory precautions block S protein binding, S protein cleavage, endo-lysosomal
20 Journal of Intensive Care Medicine 36(1)

Figure 1. Life cycle of SARS-CoV-2 and targets of investigational therapies. SARS-CoV-2 enters cells by fusion (1a) or by endocytosis (1b), the
latter of which requires escape from acidified endolysosomal compartments. Following entry, viral RNA (2) and protein synthesis (3) occur in
order to produce new virions which are assembled and packaged (4) prior to release from the cell (5). Therapies targeting the viral life cycle are
depicted according to their proposed mechanism of action as detailed in the text. Figure created with BioRender.com.

escape, RdRp activity, and Mpro activity are in active develop- Data from Wuhan, China63 and Lombardy, Italy27 indicate
ment and testing, as are approaches aimed at vaccination that a minority (7% and *16%, respectively) of patients prog-
and adoptive transfer of neutralizing antibodies from convales- ress to require ICU admission, almost universally for hypoxe-
cent sera. mic respiratory failure. While initial reports indicated that ICU
mortality was quite high,63 subsequent cohorts 12-15 have
reported substantially lower mortality rates in line with
Clinical Presentation, Diagnostic Testing, pre-COVID-19 mortalities in moderate to severe ARDS.64
and Triage Patient characteristics associated with need for critical care
include age >60 years, male sex, active or historical smoking,
Clinical Features and presence of comorbidities including cardiac disease, dia-
The initial presentation of COVID-19 is non-specific and may betes, and chronic pulmonary disease.65 Laboratory abnormal-
include fever, malaise, sore throat, cough, and myalgias— ities significantly associated with risk of severe disease include
though no single symptom is present in a majority of cases. lymphopenia, elevated troponin, elevated creatine phosphoki-
Fever is the most common presenting symptom but is present nase (CPK), elevated creatinine, elevated lactate dehydrogen-
on presentation in less than half of cases.60 “Atypical” respira- ase (LDH), elevated d-dimer, prolonged international
tory viral symptoms including anosmia61 and gastrointestinal normalized ratio of prothrombin time (PT/INR), and increased
complaints62 have also been reported, sometimes as the domi- C-reactive protein (CRP); procalcitonin and total leukocyte
nant or sole early symptoms. In published case series, the med- count are often normal.66-69 The vast majority of patients exhi-
ian incubation period is approximately 4 days and the median bit abnormalities on chest imaging—typically bilateral patchy
time to ICU transfer from symptom onset is approximately opacities and/or consolidation.12-16,63 CT imaging does not
10 days.63 reliably distinguish COVID-19 from other causes of infectious
Chivukula et al 21

Table 1. Clinical Characteristics and Preliminary Outcomes of COVID-19 Respiratory Failure.a

Respiratory system
Cohort Location Number of patients Patient population P:F ratio (median) compliance (median) Mortality

Schenck et al15 New York City, NY, USA 267 Mechanical 101 28 18.3%
Ventilation
Ziehr et al12 Boston, MA, USA 66 Mechanical 182 35 16.7%
Ventilation
Auld et al13 Atlanta, GA, USA 217 ICU 132 34 25.8%
Mitra et al14 Vancouver, BC, Canada 117 ICU 180 35 15.4%
Bhatraju et al16 Seattle, WA, USA 24 ICU 142 37 50%
Cummings et al17 New York City, NY, USA 257 ICU 129 27 39%
a
The table summarizes physiologic variables and outcomes of patients with severe (requiring ICU admission) COVID-19 in North America. Mortality figures
include outcomes for all patients at the time of data censoring, including those still hospitalized, in the ICU or one mechanical ventilation. P: F refers to the ration
of partial pressure of arterial oxygen to fraction of inspired oxygen. Reported values are medians. Respiratory system compliance values are also reported as
medians.

pneumonitis, but SARS-CoV-2 has been associated with a A second controversial topic surrounds the possibility of a
preferentially peripheral distribution of opacities, absence of pathologically hyperinflammatory state in severe COVID-19,77
pleural effusions, and absence of lymphadenopathy.70 The colloquially described as “cytokine storm.” Similar pathophysiol-
most common severe manifestation of COVID-19 in the ICU ogy has been proposed in SARS and influenza as well,78,79 though
is ARDS, defined clinically by acute onset of hypoxemia, bilat- definitive evidence of a causative role for immune dysfunction in
eral opacities not clearly caused by heart failure, and hypox- driving mortality remains lacking. The majority of data support-
emia that persists on at least 5 cm H 2 O of positive ing a role for “cytokine storm” in SARS-CoV-2 infection have
end-expiratory pressure (PEEP).71 focused on interleukin-6 (IL-6) and ferritin, secreted factors
previously associated with immunotherapy-related cytokine
release syndrome (CRS) and hemophagocytic lymphohistiocyto-
Controversies Regarding Clinical Features sis (HLH). For example, Ruan and colleagues65 reported *2-fold
of Severe COVID-19 higher IL-6, ferritin, and CRP levels in COVID-19 non-survivors
Some controversy as to the nature of COVID-19 ARDS has than survivors while Zhu et al. identified IL-6 elevation as highly
arisen following early anecdotal reports suggesting that, as predictive of outcome.80 However, the mere presence of particu-
compared with patients suffering from comparable degrees of lar laboratory abnormalities in a heterogeneous disease (or in
hypoxemia from alternate causes, COVID-19 patients exhibit severe cases of the disease) does not imply an etiologic role for
lower driving pressures (plateau pressure—PEEP), lower the analyte in question. Accordingly, hemophagocytosis and
“recruitability,” and higher respiratory system compliance.5 It hypercytokinemia have long been recognized as relatively com-
was suggested from such reports that COVID-19 uniquely pre- mon findings in patients who die with ARDS or sepsis.81,82 On
sents with a novel phenotype characterized by preserved com- average, levels of pro-inflammatory cytokines such as IL-6, IL-8,
pliance in the face of severely impaired oxygenation. Some and TNFa do not differ between patients with severe COVID-19,
went further to propose that COVID-19 respiratory failure rep- non-COVID-19 ARDS, and sepsis.83 There has, however, been a
resents a state analogous to high-altitude pulmonary edema longstanding interest in defining subtypes or endotypes84 of
(HAPE)72,73 and should be treated accordingly. It was not ARDS which are characterized by hyperinflammation. Hyperin-
immediately clear from these reports why (or even how) flammatory sub-types of ARDS have previously been linked with
organ-level mechanical consequences of biopsy-proven DAD poor outcome85 and differential treatment responses86 but were
might systematically differ by inciting pathogen, and such associated with significantly higher levels of cytokines than have
speculation has not been supported by subsequent investiga- been reported in COVID-19. Indeed, IL-6 levels reported in
tions. Instead, multiple reports12-17 demonstrate that critically severe COVID-1968 would actually have been classified as
ill patients with COVID-19 have, on average, both low respira- hypo-inflammatory by previous subtyping schemes.87 In 2 large
tory system compliance and arterial oxygenation (as measured (>200 patient) independent cohorts of severe COVID-19, for
by the ratio of partial pressure of arterial oxygen to fraction of example, median IL-6 levels of 25pg/ml88 and 26pg/ml17 were
inspired oxygen, P: F, Table 1). Indeed, reported data on gas reported; by contrast, Calfee and colleagues have previously
exchange and pulmonary mechanics in COVID-19 are entirely described hyper- and hypo-inflammatory subtypes of ARDS85
in line with published series on ARDS in the pre-COVID-19 wherein the hypo-inflammatory subtype was characterized by
era64,74,75 and are inconsistent with HAPE.76 These findings mean IL-6 levels of 282 pg/ml and the hyper-inflammatory by
have important implications for therapy and strongly support mean levels of 1618 pg/ml.87,89 Another study examined levels of
the application of evidence-based ARDS therapeutic strategies TNFa, IL-6, and IL-8 in COVID-19 ARDS patients and again
in this disease, as discussed below. found lower concentrations of these cytokines in critically-ill
22 Journal of Intensive Care Medicine 36(1)

COVID-19 patients than in matched cases of ARDS due to bac- SARS-CoV-2 testing but may not have had the full comple-
terial septic shock.90 Thus, while a fascinating hypothesis, it is far ment of ancillary laboratory evaluation useful for risk stratifi-
from clear whether immune activation in COVID-19 represents a cation and triage. As lymphopenia and elevations in CPK,
causal event in disease progression (ie, “hyperinflammation”) or d-dimer and LDH have been associated with poor prognosis
instead identifies critically ill patients with robust and possibly in COVID-19,66 we suggest obtaining CBC with differential,
appropriate immune response. As discussed below, these data complete metabolic panel, CPK, d-dimer, and LDH. ARDS is
have important implications for the use of targeted immunomo- associated with an increase in the risk of renal failure98 and
dulators in the treatment of COVID-19 patients admitted to the elevated indices of liver function have been associated with
ICU. poor outcome.99 For these reasons we recommend a compre-
hensive metabolic panel including liver function tests. Routine
echocardiography is discouraged by the American Society for
Initial Diagnostic Testing, ICU Triage, and Care
Echocardiography100 but may be of use for patients in whom
Delivery Considerations initial cardiac biomarkers are markedly elevated or if there
A detailed technical discussion of various SARS-CoV-2 diag- exists suspicion for a component of cardiogenic shock. The
nostic assays is well beyond the scope of this critical greatest utility of these data may be in adding to clinical para-
care-focused article, but treating physicians should be familiar meters for risk stratification and ICU triage decisions which
with the strengths and limitations of current tests. The most should, as always, be guided by clinical assessment, trajectory,
sensitive assay for lower respiratory tract infection is likely laboratory evaluation, and resource availability.6 There are few
bronchoalveolar fluid (BAL) reverse transcription polymerase evidence-based criteria for triggering ICU transfer in critical
chain reaction (RT-PCR) but is rarely performed owing to inva- illness generally, and certainly not in COVID-19 specifically,
siveness and aerosol exposure risk to healthcare providers.91 By so the decision to escalate care setting remains a function of
far the most commonly used diagnostic test for SARS-CoV-2 physician and nursing assessment, institutional resources, and
infection is RT-PCR from nasal or nasopharyngeal swab sam- clinical trajectory.
ples, which recent data suggest perform comparably.92 Notably, In addition to level of care, it is important to conduct triage
and in contrast with SARS-CoV, SARS-CoV-2 RNA is detect- in such a way as to minimize risk to staff and the possibility of
able early in the course of (if not prior to) symptoms.93 It has nosocomial transmission. Considerable debate continues to
been suggested, based in large part on a study indicating high exist regarding the most appropriate forms of personal protec-
sensitivity of imaging compared to RT-PCR, that computed tive equipment (PPE) for healthcare workers. The U.S. Centers
tomography (CT) of the chest may be used to diagnose for Disease Control101 suggests a number of measures includ-
COVID-19.94 This study, however, has been criticized for the ing strict contact and droplet precautions and, where possible,
quality of the PCR assay used and the American College of cohorting COVID-19 patients to dedicated units with staff
Radiology95 recommends against the routine use of CT in the trained in donning and doffing PPE. Current CDC guidelines
diagnosis of COVID-19, concluding that imaging findings are recommend routine use of N95 respirators for healthcare work-
neither adequately sensitive nor specific for ruling in or ruling ers caring for COVID-19 patients (when possible), based
out the disease. As such, imaging does not significantly add to largely upon the theoretical possibility of airborne transmission
the diagnostic yield in an RT-PCRþ COVID-19 patient and as discussed above. World Health Organization (WHO) guide-
should be reserved for situations where alternate diagnoses need lines instead specify standard surgical masks outside of specif-
to be evaluated (ex. pulmonary embolism, pleural effusion) or to ically aerosol-generating procedures (AGPs), in line with
identify high risk PCR-negative patients for further work-up. In multiple studies which have found such standard droplet pre-
at least a subset of such patients, a definitive diagnosis could cautions effective in protecting hospital staff.30,102 Further-
change management, particularly as targeted SARS-CoV-2 more, a meta-analysis examining RCTs which compared the
therapies become more available. Some evidence does indicate efficacy of standard masks to N95 respirators in preventing
that lower respiratory tract sampling is likely to enhance diag- transmission of viral pathogens (including coronaviruses) like-
nostic sensitivity for SARS-CoV-291,96 and a recent ACCP/ wise found no clear benefit associated with N95 use during
AABIP consensus statement suggests non-bronchoscopic endo- standard care.103 Our conclusion from the available evidence
tracheal sampling as a reasonable next diagnostic step.97 is that N95 respirators may be safely reserved for situations
While no direct clinical data can yet support this practice with high risk of aerosol exposure, but in resource-rich envir-
as changing outcomes, endotracheal sampling would likely onments may theoretically provide some small additional mar-
be performed regardless to evaluate for other infectious causes gin of protection in routine use. For patients undergoing
of respiratory failure in an intubated patient and is therefore a aerosol-generating procedures, staff use of N95 or more strin-
reasonable strategy. Importantly, any sampling associated with gent particle-restricting respirators should be compulsory.
opening the ventilator circuit to room air should be considered an Recent evidence also suggests that masks worn by patients may
aerosol-generating procedure (implications discussed further further reduce the risk of droplet and aerosol-mediated infec-
below). tion of others.104
The vast majority of patients in whom a critical care con- Beyond personal protective equipment for care providers,
sultation is sought will have already undergone targeted efforts should be made to minimize the number of staff in and
Chivukula et al 23

out of all patient rooms, and examinations limited to the min- need for endotracheal intubation, though without altering mor-
imum necessary for care provision. In the intensive care unit, tality risk.110 In a setting where ICU and ventilator resources
one practical change in physician practice to facilitate this is may be limited, this finding provides a potentially compelling
the “bundling” of procedures such as central venous catheter rationale for HFNC use. Potential benefits to HFNC must,
placement, arterial line placement, and orogastric tube place- however, be weighed against potential harms both to the patient
ment to limit staff exposure and PPE use. Nursing interventions and to caregivers. To the extent that hypoxemia in COVID-19
such as patient assessment, medication administration, lab represents ARDS, HFNC presents a theoretical (but yet unpro-
draws, and turns may likewise be combined to reduce don- ven111) risk of propagating lung injury by allowing uncon-
ning/doffing and exposure. Finally, technological and practice trolled tidal volumes and large pleural pressure
changes may allow adequately equipped ICUs to place infusion swings. 108,112-115 While the importance of such patient
pumps and ventilator control panels outside of patient rooms, self-induced lung injury112 remains highly controversial116 and
thereby allowing titration of continuous infusions and respira- supported primarily by animal data(108,112-114) and limited clin-
tory support without direct contact. ical studies115 it is also true that there is currently no direct trial
A further challenge in caring for critically ill COVID-19 evidence demonstrating the superiority of HFNC to invasive
patients surrounds the difficulties in adequately communicating ventilation with regard to patient centered outcomes such as
with patients and families while maintaining the aforemen- mortality. Furthermore, HFNC is unlikely to provide any
tioned infection controls. As has been documented in prior epi- meaningful alveolar recruitment and attendant mitigation of
demic infectious disease outbreaks requiring limitation of shunt fraction.117,118 For caregivers, HFNC has been suggested
physical visitation, such communication challenges can, in turn, to increase the risk of aerosol generation. While plausible, the
erode the therapeutic alliance as well as staff morale if unad- risk of increased droplet dispersion or aerosol generation with
dressed.105-107 Few data can inform a specific set of best prac- HFNC has been called into question by extensive experimental
tices to facilitate communication, but many ICUs have taken data indicating the risk is similar to other forms of oxygen
advantage of the near universal access to smartphones capable support.119-121 Modeling studies, in addition, suggest that what-
of video calling to supplement or supplant traditional visitation ever droplet dispersion exists can be substantially mitigated by
and communication. No matter an institution’s individual tech- placing a surgical mask on the patient while employing
nical and logistical chosen solutions, we would stress the impor- HFNC.122 Non-invasive positive pressure ventilation suffers
tance of retaining principles of family-centered care in the ICU. from many of the same disadvantages as HFNC (potential for
lung injury and potential for aerosol generation) and has, in
previously published studies, underperformed HFNC.123,124
Management of Respiratory Failure Furthermore, it is less comfortable for most patients and does
in SARS-CoV-2 Infection not represent a practical strategy for extended periods of sup-
As discussed above, hypoxemic respiratory failure caused by port123 as is typically required in ARDS. “Helmet” based
SARS-CoV-2 represents a form of ARDS75 and should there- NIPPV has recently been associated with reduction in need for
fore be managed as such, drawing upon the decades of avail- intubation and reduced mortality in hypoxemic respiratory fail-
able basic, translational, and clinical evidence in this area. ure,125 but this modality has yet to be established in large ARDS
Anecdotal reports4,5,7,108 from early in the COVID-19 pan- cohorts. Published guidelines126 on the care of COVID-19
demic suggesting an atypical or unique combination of high patients generally recommend a trial of HFNC (over NIPPV)
compliance and severely impaired gas exchange which might prior to intubation but the level of evidence for these recommen-
merit non-standard treatment have been conclusively refuted dations is graded as low. NIPPV, with appropriate aerosol safe-
by more comprehensive subsequent analyses.12,75,109 Since guards, should still be offered to patients with primarily
evidence to date supporting significant deviation from hypercarbic respiratory failure (ex. known COPD), an indica-
guideline-based care of ARDS in this illness is lacking, treat- tion for which there exist high quality data for benefit.127 The
ment algorithms should codify the primacy of established, evi- available evidence does suggest that a trial of HFNC may reduce
dence based principles (Figure 2). That said, there are some intubations in COVID-19 associated hypoxemic respiratory
specific ways in which care of COVID-19 does differ from failure, but is unlikely to affect mortality.
usual care. These differences stem from (1) infection control
issues associated with SARS-CoV-2, (2) the expectation that
ICU resources may be limited in the course of a pandemic, and
Decision to Intubate
(3) the use of therapies specific to SARS-CoV-2. Specific triggers for intubation in hypoxemic respiratory fail-
ure remain controversial and largely in the realm of clinician
judgment; indications include increased work of breathing
Use of Non-Invasive Support Strategies:
(accessory muscle use, tachypnea), persistent hypoxemia
HFNC and NIPPV despite supplemental oxygen, agitation/altered mental status
Oxygen delivery by humidified high-flow nasal cannula precluding care, and trajectory of worsening hypoxe-
(HFNC) circuits has become increasingly utilized in hypoxe- mia.6,108,111,112 In the presence of bilateral pneumonia (and
mic inpatients. Meta-analyses suggest HFNC may reduce the thus heterogeneous inflation), mechanical ventilation with low
24 Journal of Intensive Care Medicine 36(1)

Figure 2. Evaluation and management of critically ill adults with SARS-CoV-2. This single-page guideline outlines consensus recommendations
for the evaluation and the management of critically ill patients with COVID-19. The left panels describe clinical and laboratory diagnostics, the
middle panel provides a stepwise algorithm for respiratory failure management, and the right panel highlights COVID-19 specific aspects of
general critical care. URI, upper respiratory infection; HTN, hypertension; CBC, complete blood count; CMP, comprehensive metabolic panel;
PT, prothrombin time; PTT, partial thromboplastin time; EKG, electrocardiogram; LFTs, liver function tests; CPK, creatine phosphokinase; CRP,
C-reactive protein; FiO2, fraction of inspired oxygen; SaO2, arterial hemoglobin oxygen saturation; Vt, tidal volume; PEEP, positive end-
expiratory pressure; POCUS, point-of-care ultrasound; MDI, metered dose inhaler; CVO2, central venous hemoglobin oxygen saturation; SAT,
spontaneous awakening trial; SBT, spontaneous breathing trial; ABCDEF bundle, SCCM ICU liberation bundle; VTE, venous thromboembolism,
NMB, Neuromuscular blockade.

tidal volumes may theoretically be less injurious than contin- outcomes in COVID-19 from centers employing an “early
ued vigorous spontaneous breathing with or without intubation” (ie. minimal use of HFNC/NIPPV) strategy sug-
non-invasive support112,128 but this theoretical benefit is gests favorable outcomes.12 Prior analyses of intubation com-
balanced by the risks of sedation and invasive procedures asso- plications (particularly cardiac arrest) have also identified
ciated with mechanical ventilation, particularly as sedation has pre-intubation hypoxemia and lack of pre-oxygenation as
been independently associated with increased mortality129 in major risk factors,131,132 arguing against delaying intubation
ICU patients. In the particular case of COVID-19, mechanical to the point of truly refractory hypoxemia. However, as for
ventilation results in the patient breathing in a closed, filtered other forms of respiratory failure, no specific objective cutoffs
circuit that may also reduce the risk of viral transmission. to guide this decision are likely forthcoming and the decision to
Reports on the use of non-invasive ventilation in prior out- intubate must be made based on individual provider clinical
breaks of MERS have indicated a high rate of failure and pro- judgment. In practice, one argument for erring on the side of
gression to mechanical ventilation130 and initial reports of “early” intubation in COVID-19 may be logistical:
Chivukula et al 25

non-emergent intubation allows staff adequate time to don moderate hypoxemia without evidence of end organ
appropriate PPE and prepare for the procedure.3 hypoxia.150 While no single algorithmic approach to optimiz-
ing ventilator settings can be succinctly summarized, below we
outline a reasonable approach which incorporates physiologi-
Initial Ventilator Settings cal and clinical trial evidence:
Management of ARDS in the setting of COVID-19 does not If Pplat exceeds 30 cmH2O and/or driving pressure exceeds
meaningfully differ from standard ARDS management.3 Based 15 cmH2O, patients may be at risk of barotrauma despite low
on extensive clinical trial data133,134 demonstrating mortality tidal volume ventilation. One common situation in which high
benefits of a low-tidal volume strategy, patients should be ini- plateau pressures do not necessarily indicate high transpulmon-
tially placed on assist/control ventilation with tidal volume of ary pressures (and thus risk for barotrauma) is in the obese, in
approximately 4-8cc/kg predicted body weight (PBW, calcu- whom elevated plateau pressure may nevertheless be associ-
lated from height) and a set rate less than or equal to 35 breaths ated with low transpulmonary pressures (Ptp) due to elevated
per minute.133 A number of published approaches to setting pleural pressure (Ppl; Ptp ¼ Pairway opening—Ppl).151 In circum-
positive end-expiratory pressure (PEEP) level are available and stances where persistent high plateau pressures and concern for
are discussed below. Median PEEP levels in most available barotrauma persist, it may be beneficial to further reduce tidal
series of COVID-19 patients are in the range of 8-12 cm volume below 6cc/kg to as low as 4cc/kg PBW.133,142 The
H2O135,12-15 and moderate (8-10 cmH2O) initial PEEP is there- lower limit on the ability to decrease tidal volume is deter-
fore suggested. Strategies employing higher or individualized mined by the associated decrease in minute ventilation and
PEEP, despite strong physiologic rationale136,137 and promis- resultant hypercarbia.133,142 Respiratory rate can be increased
ing results in retrospectively identified subsets of patients,138 as needed to compensate minute ventilation, as long this does
have not been shown to be beneficial in prospective rando- not result in significant auto-PEEP—a phenomenon in which
mized, controlled trials.139-141 Plateau airway pressure (Pplat, expiratory time is insufficient for complete exhalation.152
airway pressure measured during an end-inspiratory pause) Patients with pre-existing obstructive lung disease (COPD,
should be maintained below 30 cmH2O, though more recent asthma, or bronchiectasis) and subsequent flow limitation153
evidence indicates that driving pressure (P plat —PEEP) are at highest risk of auto-PEEP. Auto-PEEP can be recognized
<15cmH2O better predicts mortality than either set tidal vol- by an expiratory flow curve that does not return to zero prior to
ume or plateau pressure.142 Hypercarbia is acceptable (permis- the initiation of the next inspiration.154 Auto-PEEP can be
sive hypercarbia) if there is no evidence of increased mitigated or eliminated most effectively155 by decreasing
intracranial pressure and arterial pH remains greater than respiratory rate but also, potentially, by increasing inspiratory
7.25. Initial tidal volume and PEEP should be adjusted (see flow rate (thereby decreasing I: E ratio) and by treating rever-
below) to maintain targeted driving pressures, oxygen saturation, sible airflow obstruction with bronchodilators.
and plateau pressure. A discussion of alternative modes of venti- In the presence of persistent hypoxemia (SaO2 < 90%)
lation is beyond the scope of this article, but pressure-cycled requiring high FiO2, attempts should be made to formally opti-
modes143 such as pressure-assist control (PC), volume support mize the PEEP.156 The physiological consequences of varying
ventilation, 144 and airway pressure-release ventilation PEEP are well-reviewed elsewhere157 but increased PEEP can
(APRV)145 should be avoided insofar as these modes are incon- maintain in the open state alveoli at risk for collapse. This
sistent with the maintenance of low tidal volumes,146 may results in decreased intrapulmonary shunt and augmented oxy-
respond inappropriately to increases in patients respiratory genation. However, higher PEEP may also impair respiratory
demand, and have not conclusively been shown to improve out- system mechanics, paradoxically increase intrapulmonary
comes. In this context it is worth reiterating that the benefit of low shunt, and precipitate hemodynamic instability (particularly
tidal volume ventilation is among the most robust results in the in the setting of hypovolemia or limited right ventricular
critical literature. In a recent re-analysis147 of trials reporting an reserve). A physiological approach to PEEP titration attempts
effect on mortality in critical care, low tidal volume ventilation to identify the end-expiratory pressure that maximizes the ben-
and prone ventilation represented 2 of only 4 (out of 862 studied) efits of PEEP and minimizes its potential harms. Despite
interventions with robust fragility index and positive effect on numerous clinical trials, however, no method of PEEP optimi-
mortality. zation in ARDS has been demonstrated as clearly superior to
any other and some titration protocols can cause harm.139-141 In
addition, physiological PEEP titration may be laborious and
Adjustment of Ventilator Settings in COVID-19 ARDS require substantial time investments on the part of intensivists
Patients with ARDS may fail to respond to initial ventilator and respiratory therapists—both likely in short supply in the
settings, either through persistent high airway pressures setting of a pandemic. Thus, it is our opinion that a reasonable
(Pplat > 30cmH20 and/or driving pressure > 15cmH2O) or per- initial approach to persistent hypoxemia is to implement the
sistent hypoxemia (SaO2 <90% and/or persistently low PaO2 to ARDSnet “low PEEP” algorithm140 in most circumstances.
FiO2 (P: F) ratio. Although ideally oxygen saturation should be In unusual circumstances (refractory hypoxemia, extreme
maintained between 90 and 96%,148,149 trial data suggest opti- body habitus, unacceptable hemodynamic or respiratory
mization of airway pressures is to be prioritized over correcting mechanical parameters) and if time and staffing allow, PEEP
26 Journal of Intensive Care Medicine 36(1)

may alternatively be set by best tidal compliance156 using a Anecdotally, several centers have established hospital-wide
decremental “best PEEP” trial.158 In a decremental best PEEP “proning teams” to ease these resource needs and ensure expe-
trial, the optimal PEEP is the one that maximizes the compli- rienced operators are available for positioning critically ill
ance (Crs ¼ Vt/driving pressure).159 When setting PEEP by best patients. Contraindications to prone ventilation include an
tidal compliance, a sustained application of high airway pres- inability to turn the neck (e.g. fixed or unstable c-spine) and
sure (“recruitment maneuver”) is often performed first. How- unstable sternum. Vascular access lines, thoracostomy tubes,
ever, the protocol of a recruitment maneuver followed by and hemodialysis lines are not absolute contraindications to
decremental PEEP was specifically associated with harm in the prone ventilation162 The patient should be maintained in the
recent Alveolar Recruitment Trial (ART).141 This trial has been prone position for at least 12169 hours, and potentially longer
criticized for applying recruitment maneuvers in an unselected (24þ hours)170 as patient care needs allow. One approach
population and, in that context, it is notable that few of the which may also mitigate the additional needs for caregiver PPE
patients in ART demonstrated evidence of recruitment. This and exposure in COVID-19 patients is to supinate patients for
leaves open the possibility that it may yet be possible to iden- daily care needs in the morning, allowing an extended period
tify a subgroup of patients, such as those described above, in for prone positioning thereafter. The decision to discontinue
whom recruitment is beneficial but it remains unknown prone ventilation is driven by physiological parameters: if
whether COVID-19 ARDS might be enriched or depleted for P:F remains >150-200 and driving pressure <15 cmH2O at the
such patients. Therefore, while there may be a role for such end of a 2 hour period of supine ventilation on PEEP
maneuvers as a rescue for refractory hypoxemia, our opinion <10 cmH2O, we discontinue prone ventilation.162
based on the available data is that they are best used rarely.
A gentler approach, which attempts to reconcile the findings of
ART and other PEEP titration trials with physiologic reason-
Management of Ventilator Asynchrony
ing, may be to simply increment PEEP in a stepwise fashion, Patients with ARDS may have a high respiratory drive, so
starting from current PEEP, with careful attention to driving attempts to reduce tidal volumes and airway pressures can result
pressure to ensure it is not increasing at each step. This man- in dyspnea and ventilator asynchrony.171 Asynchrony com-
euver can be followed by a decremental trial from some more monly manifests as “double triggering,” during which ongoing
modestly increased PEEP than the 25-30 cmH2O traditionally inspiratory effort triggers a second ventilator-delivered breath
used in recruitment maneuvers. The best PEEP is again chosen prior to expiration. Double triggering results in high tidal
by best compliance as long as this coincides with hemody- volumes and airway pressures that can be injurious.172 Increas-
namic stability and acceptable oxygenation (arterial oxygen ing inspiratory flow may decrease dyspnea, but if asynchrony is
saturation 90-96%). persistent then consider augmenting sedation and/or neuromus-
cular blockade (NMB). Effective sedation is a prerequisite for
neuromuscular blockade and may obviate its need altogether;
Prone Ventilation narcotics are particularly effective suppressors of respiratory
Prone ventilation for ARDS is strongly recommended in cur- drive. The initiation of neuromuscular blockade should be only
rent clinical practice guidelines160 and should be implemented undertaken in response to asynchrony or persistent high airway
in intubated COVID-19 patients with persistent moderate to pressures as recent high-quality RCTs do not indicate a benefit to
severe ARDS. Physiological benefits associated with prone routine NMB173 in ARDS. Double triggering which increases
positioning include improved recruitment, decreased inhomo- with deep sedation may indicate “reverse triggering” or entrain-
geneity of ventilatory units, improved V/Q matching, and ment,174 a reflex breathing pattern which may abate with reduc-
decreased pulmonary vascular resistance.161 Evidence-based ing sedation.
indications162,163 for prone ventilation include a P:F ratio
<100-150 and FiO2 >0.6 following initial stabilization. How-
ever, since prone ventilation results in a host of improvements Pulmonary Vasodilators
to lung mechanics and is associated with low risk and low cost, In case of persistent hypoxemia despite optimization of venti-
we feel it is reasonable to employ a relatively low threshold lator settings, patients may be treated with inhaled pulmonary
(P:F *150) for considering prone positioning. Furthermore, as vasodilators as temporizing or salvage therapy.175 These agents
many of the physiological benefits associated with prone posi- work to match ventilation and perfusion by specifically redir-
tioning do not depend on positive pressure ventilation, it is not ecting pulmonary arterial blood flow to those regions that are
unreasonable to consider employing this strategy even in spon- best ventilated. The net effect is to decrease V/Q mismatch and
taneously breathing COVID-19 patients (“self-proning”). thereby augment oxygenation.176 Importantly, while inhaled
Mindful of the potential need to conserve ventilators, many pulmonary vasodilators typically improve P:F ratios for
centers have employed such a strategy in the current outbreak approximately 24 hours in ARDS, their use has never been
with evident benefit.164-168 linked to reduced mortality or even liberation from mechanical
Prone ventilation can be carried out in the patient’s existing ventilation—underscoring the pathophysiology of ARDS as a
bed and requires minimal additional equipment, though it does multi-organ system disorder of more than hypoxemia.177 While
require additional staff for physically positioning the patient. physiologically equivalent, the use of inhaled nitric oxide
Chivukula et al 27

(iNO) over inhaled prostacyclin analogs in COVID-19 may be COVID-19 but some early reports183 indicate survival in line
preferred in practice, if available, as inhaled prostacyclin with pre-COVID-19 ECMO data.
requires the use of a filter in the ventilator circuit which has
to be changed periodically, thus creating an additional expo-
sure for staff. In addition, NO exerts anti-viral effects in vitro, General Care of the Critically Ill
theoretically a useful if clinically unproven adjunctive property COVID-19 Patient
in viral respiratory illness.178,179 A successful trial of iNO may
In addition to lung protective ventilation, ICU management of
be defined by a 20% increase in PaO2 with administration of
severe COVID-19 should focus on support of organ function
40 ppm iNO; if a patient responds to iNO, its use should be
while minimizing risk of transmission with effective isolation
maintained and ventilator settings re-titrated to maximize lung
practices and avoidance of low-value diagnostics. 3
protection. There is, however, likely little benefit to prolonged
Evidence-based management of common problems in the ICU
use of iNO.
should be employed as per usual care of ARDS and sepsis187
and a comprehensive review of topics such as transfusion
Extracorporeal Membrane Oxygenation (ECMO) thresholds, indications for renal replacement therapy, glucose
control, stress ulcer prophylaxis, nutrition, etc. are well beyond
Patients with persistent hypoxemia or unacceptable airway
the scope of this work. Instead, below we discuss some of the
pressures despite the optimization of ventilator settings, neuro-
ways in which care for critically ill COVID-19 patients may
muscular blockade, prone positioning, and inhaled pulmonary
differ from standard practice, but stress again that these meth-
vasodilators are deemed to have “refractory” ARDS. In such
ods are more similar than not to those provided to patients with
cases, consideration should be given to initiation of venove-
ARDS and/or sepsis generally (Figure 2).
nous extracorporeal membrane oxygenation (VV-ECMO) or
transfer to a center where this therapy is available.180,181 Trial
data are mixed and complex on the question of timing of Antimicrobial Therapy
ECMO initiation, but point to improved outcomes in refractory Bacterial superinfection has been reported in ICU patients with
ARDS.180-182 Society guidelines generally recommend initia- COVID-19 but is not a frequent finding on initial presenta-
tion of ECMO in COVID-19 patients only after the failure of tion.16 If initial clinical or laboratory features raise concern for
less invasive therapies including mechanical ventilation and coincident bacterial infection (neutrophilia, elevated serum
prone positioning.183 As ECMO therapy is associated with very procalcitonin, lobar consolidation, purulent sputum, etc.), con-
high resource utilization184 and resources may be limited in the sideration should be given to the early initiation of empiric
setting of a SARS-CoV-2 surge, the criteria for initiation of antibiotics with rapid de-escalation, as outlined in the ATS/
ECMO in the setting of COVID-19 may differ from criteria for IDSA guidelines.188 Intubated and critically ill patients should
non-COVID-19 ARDS. The Extracorporeal Life Support Orga- receive ventilator-associated pneumonia (VAP) therapy guided
nization (ELSO) has published guidelines185 which stress the by cultures or, if an organism is not identified, empirically per
dynamic nature of ECMO triage and emphasize the guiding usual hospital protocol. Invasive diagnostic techniques such as
principle that resource intensive therapies should be directed bronchoscopy and mini-BAL offer little benefit over endotra-
towards patients who are most likely to benefit. For that reason, cheal aspiration (using an in-line catheter and semiquantitative
guidelines suggest usual criteria for the initiation of ECMO culture),188,189 increase transmission risk to staff, and should
while hospitals are functioning within normal capacity and therefore generally be avoided. COVID-19 may alter the ease
more stringent criteria (prioritizing younger patients, those in of providing evidence-based bundled care to reduce VAP
single organ failure and those with non-COVID-19 indications risk.190 Some interventions, such as head of bed elevation and
which have been proven to benefit from ECMO) when capacity avoidance of protocolized ventilator circuit changes, should be
is stretched. Absolute contraindications to ECMO in largely unaffected. Others, including subglottic suctioning and
COVID-19, according to ELSO guidelines, include limited life targeted sedation may be impractical or unsafe given aerosol
expectancy, clinical frailty, mechanical ventilation >10 days, risks and inability for staff to rapidly enter rooms.
inability to tolerate anticoagulation, severe neurologic injury,
or severe multi-organ failure.186 Relative contraindications
include body mass index (BMI) >40, immunocompromise, and
Imaging
multipressor shock not being considered for veno-arterial sup- Radiographic findings consistent with a diagnosis of
port.185 The decision to offer ECMO support should be multi- COVID-19 are discussed above (See Clinical Features). Rou-
disciplinary and take into consideration patient prognosis, tine daily chest radiographs do not alter ICU outcomes191 and
resource availability, and comorbidities.186 Patients with should certainly be avoided in the care of COVID-19 patients
co-existing cardiogenic shock may be candidates for venoar- in light of staff exposure risks. Possible indications for chest
terial ECMO (VA-ECMO), though the precise etiology and radiography subsequent to ICU admission include hemoptysis,
potential reversibility of myocardial dysfunction in suspected lung volume loss consistent with mucus plugging,
COVID-19 remains poorly defined (discussed further below). rapidly progressive hypoxemia and/or hypotension suggestive
Little outcome data is available for ECMO specifically in of pneumothorax, or concern for VAP. Various patterns
28 Journal of Intensive Care Medicine 36(1)

(primarily bilateral ground glass opacity and/or consolidation) invasive tests have not yielded sufficient diagnostic informa-
on chest CT have been associated with COVID-19 but these are tion both due to aerosol risk and limited evidence for additional
non-specific and are not likely to inform daily management nor diagnostic yield. Respiratory samples for diagnosis of bacterial
to improve during a typical ICU admission.192 Transport of superinfection may be obtained by close-loop endotracheal
patients to the CT scanner presents risks to staff and other aspirate in intubated patients. Inhaled medications should be
patients (viral transmission), as well as to the critically ill given by metered dose inhaler instead of nebulizer whenever
patient undergoing transport (potential for derecruitment and possible to decrease the risk of viral transmission and ventila-
instability). For these reasons, chest CT should be reserved for tors should be set up with adaptors in the dry arm of the circuit
situations in which an alternative and actionable diagnosis to facilitate subsequent use of inhalers without opening the
(i.e. pulmonary embolus) is suspected and the risks of empiric circuit.
therapy (ex. anticoagulation) are deemed significant.
Hemodynamics and Fluid Management
Laboratory Investigation Case series from China and Italy18,67 indicated a predominance
Recommended laboratory testing includes CBC with differen- of isolated respiratory failure associated with COVID-19 with
tial (in order to trend total lymphocyte count and surveil for lower than expected rates of secondary organ failure, shock,
superimposed infection) and complete metabolic panel and renal failure than unselected ARDS patients. Nevertheless,
(in order to surveil for renal and hepatic injury). Elevations shock occurs in approximately a third of critically ill
in CRP, CPK, LDH, and D-dimer193 have been associated with COVID-19 patients.16,18,67 Importantly, while distributive
poor outcome and are indicated for prognosis and triage; their shock due to viral sepsis likely underlies the majority of such
role beyond initial risk-stratification remains ill-defined. Bac- cases, it remains important for clinicians to entertain a broader
terial and fungal superinfection have been reported in a minor- differential diagnosis. Multiple case reports have described the
ity of cases, so routine sputum and blood cultures on ICU occurrence of acute heart failure198 and cardiogenic shock199
admission are reasonable. As discussed previously, the role associated with severe COVID-19. ACE2 is expressed in the
of measuring inflammatory markers such as ferritin, IL-6, solu- heart and some reports have described the isolation of viral
ble IL-2 receptor, and CRP remains unclear outside of clinical RNA in post-mortem samples of cardiac tissue.200 One small
trial settings in which immunomodulatory therapy may be con- Chinese series reported an isolated cardiac cause of death in 7%
sidered, but may be reasonable to consider in patients with of patients and an additional third of whom died of combined
refractory shock, sustained fever, MODS, or other signs sug- cardiac and respiratory failure.65 The extent to which these
gestive of “cytokine storm.” The role for biomarkers such as reports constitute evidence of a true viral myocarditis201
procalcitonin remains unvalidated, though low procalcitonin remains controversial, however, as endomyocardial biopsies
levels have previously predicted absence of bacterial infection in the setting of COVID-19 related heart failure showed no
in critically ill patients with influenza.194 evidence of direct viral infection of cardiac myocytes.202 The
diagnosis of a genuine myocarditis is also made challenging by
the high prevalence of cardiac biomarker elevation in the cri-
Aerosol Generating Procedures tically ill, with COVID-19 patients being no exception.69 Thus,
It was has been postulated195 that procedures such as broncho- while the true incidence of viral myocarditis accompanying
scopy, endotracheal intubation, extubation, nebulizer adminis- SARS-CoV-2 infection remains unclear, acute heart failure is
tration, and tracheostomy placement/manipulation may well-appreciated in COVID-19 critical illness and requires
generate smaller, more persistent particles and thereby facili- exoneration in the decompensating patient. Beyond cardio-
tate airborne transmission of agents more typically spread by genic shock, multiple case reports have also suggested the
respiratory droplets. That said, there is no experimentally vali- presence of an elevated rate of acute mesenteric ischemia and
dated reference list of such procedures101 and it is likely such accompanying shock in COVID-19. Kaafarani et al. reported
activities represent a spectrum of risk rather than a binary dis- acute mesenteric ischemia in 4/141 critically ill patients with
tinction. While the risk of most such aerosol generating proce- COVID-19203 with similar findings independently reported
dures (AGPs) remains somewhat controversial, both the elsewhere.204 Although ACE2 expression in the gut has been
CDC101 and WHO196 recommend conducting potentially aero- reported205 and SARS-CoV-2 can infect enterocytes in vitro,205
sol generating procedures with all staff wearing N95 respirators whether these observations are attributable to GI infection,
and other appropriate PPE. The CDC further recommends such microvascular thrombosis (see below), or some other patho-
procedures ideally be performed in airborne infection isolation physiological process remains unclear. Computed tomography
rooms (AIIRs). Intubation, in particular, seems to be high risk angiography (CTA) is the preferred imaging technique for the
for aerosol generation197 and the decision to intubate should diagnosis of acute mesenteric ischemia.206 As abdominal CTA
therefore be made early in the course of anticipated clinical is not routinely performed in respiratory failure patients, the
deterioration to reduce the need for urgent or uncontrolled diagnosis of acute mesenteric ischemia requires a high index of
procedures which may carry higher risks of transmission. As suspicion in the face of evolving hemodynamic instability with
outlined above, bronchoscopy is not indicated unless less urgent involvement of surgical consultants if identified.
Chivukula et al 29

In the majority of COVID-19 patients experiencing shock recommendations193 largely advise prophylactic anticoagula-
related to progressive viral sepsis, it is appropriate to treat per tion for all COVID-19 patients and reserve therapeutic antic-
usual protocols for distributive shock with particular care taken oagulation for those with confirmed thrombosis or pre-existing
to avoid fluid overload. Patients with hypoxemic respiratory indications. Whether there is a role for augmented intensity
failure should generally be managed with a conservative fluid VTE prophylaxis remains to be studied in controlled trials.
strategy207 which includes (1) limiting fluid boluses to patients
in shock who have indications of volume responsiveness,208,209
and (2) early initiation of diuresis in those with resolved shock. Role for Targeted Therapies and
Positive fluid balance should be avoided in patients who are not Immunomodulation
volume responsive, a practice supported by high-quality ran- The pathophysiology of SARS-CoV-2 remains poorly defined
domized trial data.210 In addition, positive fluid balance over and the precise mechanistic basis underlying severe,
the course of ICU stay is associated with increased mortal- life-threatening COVID-19 is unknown. To date, 2 therapies
ity.211,212 As for most critically ill patients, the use of balanced have been shown in randomized controlled trials to improve
crystalloids (ex. lactated Ringer’s solution) is preferred over outcomes in COVID-19: the antiviral nucleoside analog remde-
normal saline, albumin, or alternative colloid prepara- sivir20 and the steroid dexamethasone.19 However, dozens of
tions.213,214 Current guidelines for treatment of distributive additional trials are ongoing and can be broadly divided into
shock3 suggest titrating therapy to end-organ perfusion as indi- 2 classes: (1) those that aim to directly target viral infectivity or
cated by renal function, mental status, serum lactate, and (if replication and (2) those that aim to alter or modulate the host
available) central venous oxygen saturation.187 Adjunctive response to infection.
therapies for septic shock such as glucocorticoids are the sub-
ject of conflicting trial data215-217 with evidence of minimal
benefit or harm in prior respiratory viral illnesses.218-221 Nev-
Experimental Therapies Targeting SARS-CoV-2 Life Cycle
ertheless, steroids should be considered in refractory distribu- As outlined in the “Virology and pathophysiology” section
tive shock associated with COVID-19, particularly in light of above, SARS-CoV-2 employs a well-characterized series of
strong evidence for glucocorticoid benefit in COVID-19 ARDS steps to enter host cells and replicate (Figure 1). A number of
(discussed below). efforts aim to block viral engagement of the cell surface ACE2
receptor. One strategy centers on anti-spike protein neutraliz-
ing antibodies. Analogous blocking antibodies have been iden-
Coagulation Abnormalities and Thrombotic Risk tified against SARS-CoV and there is intense interest in
While microvascular and macrovascular pulmonary thrombo- characterizing the structural basis for antibody recognition.236
sis are long-recognized features of ARDS,222,223 efforts at One such antibody has been reported to efficiently block
blunting coagulation in the setting of ARDS have largely failed SARS-CoV-2 infection in vitro237 and it (among others) are
to show benefit in clinical trials.224-226 Early observational moving toward clinical trials. However, it remains unclear
reports from China67 indicated that COVID-19 was associated whether such strategies will be effective in vivo or even
with abnormal indices of coagulation, a finding subsequently whether they are necessarily safe, particularly in light of a
confirmed in larger independent cohorts which highlighted ele- recent report of anti-S protein immunoglobulin induced
vations in d-dimer and fibrinogen as consistent features of exacerbation of acute lung injury in SARS-CoV-infected
COVID-19.227 These findings have led multiple investigators non-human primates.238 The use of “decoy” ACE2 receptor
to hypothesize that the tropism of SARS-CoV-2 for ACE2 and to inhibit viral infection in human tissue organoids was also
the expression of that receptor on endothelial cells228 might recently reported,239 but whether such a strategy is viable in
result in a systematic endothelialitis and resultant widespread patients remains unclear.
coagulation abnormalities. However these reports remain con- On August 23, 2020 the U.S. Food and Drug Administration
troversial, both because the extent of genuine endothelial cell (FDA) issued an emergency use authorization (EUA) for con-
involvement has come under scrutiny229 and because abnorm- valescent plasma therapy in COVID-19. Convalescent plasma
alities in such parameters are common in the critically ill.193 involves the transfusion of plasma from recovered patients in
Klok et al reported230 venous or arterial thrombosis in 31/184 order to achieve a passive immunization. To date, there are no
(16.8%) critically ill patients with COVID-19, consistent with randomized clinical trials on the use of convalescent plasma.
other case series from Italy, 231 the United States, 12 and The administration of convalescent plasma resulted in
France.232 By comparison, a large cohort of ICU patients with decreased viral load and clinical improvement in 5 mechani-
severe H1N1 influenza233 pneumonia was reported to have an cally ventilated patients in an early report from China,240 The
incidence of venous thromboembolism of 10%. Complicating FDA based its decision, in part, on analysis of data from an
matters, however, the rates of bleeding complications in expanded use program run by the Mayo Clinic. These data
COVID-19 have not been fully characterized and there is some have, at the time of this writing, been released only as a
evidence that bleeding risk is elevated as well.234 Due to the non-peer reviewed pre-print(241) and describe the transfusion
uncertainties as to the magnitude thrombotic risk and possibly of plasma in a total of 35,322 patients (52% requiring critical
offsetting risks of bleeding, guidelines 235 and published care). Mortality was 8.7% in patients who were transfused
30 Journal of Intensive Care Medicine 36(1)

within 3 days of diagnosis and 11.9% in patients transfused CQ/HCQ is lacking and that these drugs should therefore not
after 4 days. A potential issue with convalescent plasma ther- be used for the treatment of SARS-CoV-2 infection.
apy is that the repertoire and titer of antibodies may vary by As for HIV, HCV, and other clinically relevant viral patho-
donor. Indeed, reported mortality was lower in patients trans- gens, specific small molecule inhibitors of viral enzymes have
fused with high-titer plasma than those transfused with proven more promising. Remdesivir (GS-5734), a nucleoside
low-titer plasma. While the Mayo Clinic experience suggests analog of adenosine, is an inhibitor of viral RNA-dependent
that convalescent plasma therapy is likely to be safe, guidelines RNA polymerases with broad antiviral activity.255 Notably,
continue to recommend against consideration of convalescent remdesivir exhibits potent inhibitory activity against
plasma as standard of care242 until controlled trials are per- SARS-CoV and MERS-CoV in vitro and in animal models256
formed. It is thus too soon to know if convalescent plasma and is not associated with development of fitness-maintaining
therapy is truly effective. resistance mutations. In non-human primates, remdesivir
As outlined previously, following receptor engagement reduced lung injury after infection with SARS-CoV-2,257
SARS-CoV-2 can enter cells by fusion or by endocytosis. The though this model is notable for not inducing ARDS as
fusion-dependent pathway depends on host cell surface pro- observed in human patients. Remdesivir received an emer-
tease activity, a process recently found to be inhibited by the gency use authorization from the FDA on the basis of the
pancreatitis drug camostat mesylate.11 A placebo-controlled preliminary results from the Adaptive COVID-19 Treatment
phase IIa trial of camostat is now underway (trial Trial-1 (ACTT-1). In ACTT-1, 1063 participants were rando-
NCT04321096) following promising results in mouse models. mized to receive up to 10 days of remdesivir versus placebo;
As the endocytic pathway depends on endosome/lysosome patients in the remdesivir arm had a shorter time to recovery
escape, inhibitors of this process (particularly chloroquine deri- than those in the placebo group (11 versus 15 days).20 A sub-
vatives) have also received intense interest in the scientific and group analysis revealed this benefit was driven by the group
lay communities. Chloroquine (CQ) belongs to a class of requiring oxygen, rather than those requiring HFNC, NIPPV,
agents known as cationic amphiphilic drugs (CADs), which mechanical ventilation, or ECMO. A subsequent trial 258
become trapped and highly concentrated in acidified subcellu- showed comparable outcomes in oxygen-requiring
non-critically ill patients treated with 5 and 10 days of remde-
lar compartments such as endosomes and lysosomes. By virtue
sivir. Based on these data, current guidelines suggest remdesi-
of this trapping, CADs can increase pH and inhibit enzyme
vir for hospitalized COVID-19 patients requiring oxygen,
function within these organelles. CQ’s interference with
suggesting 5 (non-intubated patients) or 10 days (intubated
endo-lysosomal processing of antigens likely accounts for its
patients) of therapy.253 Guidelines for the use of remdesivir are
immunosuppressant activities, and a similar mechanism in
rapidly evolving, however, in part driven by drug shortages.259
virally infected cells provides an opportunity to block viral
Like RdRp, the viral Mpro protease represents an attractive
endosome escape and replication.243 Indeed, it has been appre-
target for therapy as several existing antiviral agents may effec-
ciated for decades that various CADs can interfere with the
tively inhibit its function. To date, attempts to repurpose HIV
replication of a diverse array of enveloped viruses including
protease inhibitors have not borne fruit260 and more recent
influenza, Ebola virus, HIV, dengue, Zika and hepatitis C in large clinical trials have been halted at interim analyses due
vitro . Building on these data, Wang and colleagues published to lack of evidence of benefit. Other trials remain ongoing261
2 papers47,244 examining the effects of CQ and hydroxychlor- but as of this writing off-label use of such agents outside of
oquine (HCQ, more widely available and less toxic) on the clinical trials is not supported by the evidence.
novel SARS-CoV-2 in vitro, uncovering that CQ dramatically
inhibits SARS-CoV-2 replication at low micromolar concen-
trations, while HCQ inhibits replication at *10 mM. However, Experimental Immunomodulators
previous experience with CQ/HCQ suggested skepticism was As introduced above, it has been suggested that some patients
warranted; chloroquine failed to prevent influenza infection in with SARS-CoV-2 infection may exhibit a hyperinflammatory
a clinical trial, 245 increased viral load in HIV-infected phenotype or “cytokine storm”9,77,88 meriting therapy with
patients, 246 and paradoxically increased viremia in immunomodulatory agents. Interest in such syndromes has bur-
non-human primates infected with Chikungunya virus.247 geoned in recent years as targeted therapies have become avail-
Indeed, more systematic investigations of the efficacy of CQ/ able for the treatment of cytokine release syndrome (CRS)
HCQ in COVID-19 have largely disproved its suggested utility. triggered by immune-activating oncologic therapies. However,
Large observational series248 showed no evidence of benefit it remains unclear whether such putative “hyperactivation” is
and large NIH- and WHO-sponsored trials of HCQ were necessarily a rational therapeutic target in COVID-19 given the
stopped for futility249,250 (final data pending). Published clin- challenges in distinguishing immune activation as a cause
ical trials have also failed to demonstrate benefit251 and rather than a consequence of critical illness. Indeed, prior
recently published data failed to identify any treatment effect attempts at targeted immunomodulation262,263 in critical illness
to HCQ in non-human primates infected with SARS-CoV-2.252 have largely proven ineffective. IL-6 antagonists such as toci-
Taken together, in line with the IDSA, 253 FDA, 254 and lizumab have seen widespread recent use in the management of
CDC101), we believe evidence for a therapeutic benefit for immunotherapy side effects, a context in which they mitigate
Chivukula et al 31

CRS and reduce need for steroids. It is less clear whether IL-6 is formulations and dosing strategies. A recent meta-analysis281
causally related to the pathogenesis of ARDS, though elevated of data from 7 of these trials demonstrated lower mortality in
cytokine levels have been associated retrospectively with steroid treated patients—increasing confidence in the RECOV-
improved response to steroids in septic shock.264 Numerous clin- ERY results. Accordingly, steroid therapy should be consid-
ical trials are evaluating the use of IL-6 blockade (NCT04315480, ered standard of care in hospitalized COVID-19 patients
NCT04320615, NCT04322773, NCT04330638, NCT04332913, requiring supplemental oxygen, mechanical ventilation, or
NCT04335071), though at least one has already been stopped for ECMO support.
futility and potential harm.265 In line with IDSA guidelines,253 Statins, inhibitors of HMG CoA-reductase, mediate wide-
therefore, we suggest limiting cytokine-targeting therapy to the spread effects on cellular metabolism beyond their roles in reg-
clinical trial setting. ulating serum LDL cholesterol levels. Arguments for the use of
Beyond such targeted therapies, a role for corticosteroids statins in severe COVID-19 infection arise from 2 independent
(which have a long and complex history of use in ARDS) is rationales. The first is that patients with pre-existing cardiovas-
emerging in the management of severe COVID-19. The theo- cular disease appear to be at high risk for severe disease and
retical rationale for steroid therapy in ARDS generally death after SARS-CoV-2 infection; many of these patients are
(reviewed extensively elsewhere266,267) stems from the obser- likely to meet existing guideline-based indications for statin
vation that this disease is characterized by exuberant lung therapy as primary or secondary prevention. A second, much
inflammation and that glucocorticoids provide benefit in a vari- more speculative, proposed indication for statin use is as mod-
ety of inflammatory human disease states via pleiotropic ulators of the innate immune system. Because they inhibit sig-
effects.268 That said, glucocorticoids exert myriad complex naling through the innate immune adaptor MYD88, statins have
effects in the critically ill and the precise mechanistic basis for been posited to act as beneficial “anti-inflammatory” agents in
any steroid treatment effects in ARDS remains to be clarified. viral respiratory infections,282 though it should be noted that
The history of steroid trials in ARDS spans some 3 decades269 such signaling is complex and exquisitely tightly controlled in
and cannot be reviewed here comprehensively but, in brief, vivo. Highlighting this point, genetic knockout of Myd88 has
early data did not support the use of high-dose “pulse” ther- proven fatal in mouse models of SARS.256 As for ARDS more
apy.270-273 Subsequent studies of lower-dose regimens indi- generally,283 there may exist a subgroup of hyperinflammatory
cated a benefit as evidenced by more rapid liberation from COVID-19 patients in whom statins mediate protective effects,
mechanical ventilation263,274 but without mortality benefit. but there is no validated way to identify them prospectively at
A recent multicenter RCT275 reported a 60 day mortality ben- this time. In light of the risks of transaminitis and rhabdomyo-
efit in moderate to severe ARDS patients treated with dexa- lysis with these agents, we therefore do not recommend their
methasone, but was hampered by low enrollment rates and universal use in SARS-CoV-2 infection absent an
open label design. These potential benefits of steroids in ARDS evidence-based primary indication.
are tempered by signals for harm. Steinberg and colleagues A final approach for immunomodulation in severe
identified an increase in mortality and rates of re-intubation COVID-19 aims to augment, rather than inhibit, host response
associated with steroid therapy in late (>14d) ARDS.263 to SARS-CoV-2 in order to potentiate viral clearance.
Furthermore, observational data from epidemics of influ- Granulocyte-monocyte colony stimulating factor (GM-CSF)
enza,276-278 SARS,279 and MERS220 suggested the immunosup- is a growth factor with important functions in the lung—sup-
pressive properties of glucocorticoids might actually porting the maturation of alveolar macrophages284 and promot-
exacerbate virally-induced acute lung injury. Accordingly, ing surfactant turnover and recycling. GM-CSF levels are
early expert opinion argued against the routine use of corticos- elevated in the blood of COVID-19 patients,285 an observation
teroids in COVID-19.280 This was the setting in which initial that has triggered attempts both to augment and to inhibit sig-
results from the RECOVERY trial were recently published.19 naling through this pathway—highlighting the urgent need for
RECOVERY is a multi-arm, open-label platform trial testing a deeper pathophysiological understanding. Although it failed to
number of specific therapies for COVID-19 in hospitalized show benefit in an RCT in ARDS,286 recombinant GM-CSF
patients. Patients received dexamethasone (6 mg daily for up has shown preclinical promise in ameliorating influenza acute
to 10 days, n ¼ 2104) or usual care (n ¼ 4321) with primary lung injury287 and is being studied in an active COVID-19 trial
outcome of all-cause 28 day mortality. The trial revealed a 3% (trial NCT04326920). As for many of the experimental thera-
absolute risk reduction in favor of the steroid group, a benefit pies discussed here, our conclusion is that there is an inade-
which varied by degree of respiratory support with greater quate understanding of viral pathogenesis and host response to
benefit in the mechanically ventilated (absolute risk reduction justify use of such agents outside of monitored clinical trial
12%) and a trend towards harm in non-oxygen-requiring settings.
patients. With the caveats that long-term mortality data remain
to be seen and that control arm mortality was at the upper end
of reported ranges, these findings support a therapeutic effect
Conclusion
for glucocorticoids in severe COVID-19. At the time of publi- SARS-CoV-2 is a novel coronavirus of zoonotic origin that has
cation of RECOVERY a number of other trials of steroid ther- infected millions of people worldwide and is placing severe
apy in COVID-19 were underway—using various steroid demands on intensive care resource utilization across the globe.
32 Journal of Intensive Care Medicine 36(1)

The biology and natural history of COVID-19 is becoming 6. Tobin MJ. Basing respiratory management of COVID-19 on phy-
increasingly well-understood and should inform the approach siological principles. Am J Respir Crit Care Med. 2020;201(11):
to diagnostic testing, healthcare worker precautions, and ICU 1319-1320.
triage. COVID-19 associated acute hypoxemic respiratory fail- 7. Gattinoni L, Chiumello D, Caironi P, et al. COVID-19 pneumo-
ure develops in a substantial fraction of affected patients and nia: different respiratory treatments for different phenotypes?
represents a form of ARDS, a finding consistent with what is Intensive Care Med. 2020;46(6):1099-1102. doi:10.1007/
now understood regarding the virology and pathophysiology of s00134-020-06033-2
SARS-CoV-2. The keys to evidence-based management of 8. Chen Q, Allot A, Lu Z. Keep up with the latest coronavirus
COVID-19 therefore include lung-protective low-tidal volume research. Nature. 2020;579(7798):193.
ventilation, avoidance of barotrauma, a conservative fluid strat- 9. Zhou P, Yang XL, Wang XG, et al. A pneumonia outbreak asso-
egy, and prone ventilation, with utilization of inhaled pulmon- ciated with a new coronavirus of probable bat origin. Nature.
ary vasodilators, neuromuscular blockade, and ECMO when 2020;579(7798):270-273.
necessary. The lifecycle of SARS-CoV-2 and its effects on the 10. Shang J, Ye G, Shi K, et al. Structural basis of receptor recogni-
host immune system suggest possible targets for therapy and, to tion by SARS-CoV-2. Nature. 2020;581(7807), 221-224.
date, 2 such drugs are indicated in hospitalized COVID-19 11. Hoffmann M, Kleine-Weber H, Schroeder S. SARS-CoV-2 cell
patients. In time, we anticipate the continued development and entry depends on ACE2 and TMPRSS2 and is blocked by a
adoption of additional targeted therapies which will help bring clinically proven protease inhibitor. Cell. 2020;181(2):
the pandemic under control. 271-280. e8.
12. Ziehr DR, Alladina J, Petri CR, et al. Respiratory pathophysiology
of mechanically ventilated patients with COVID-19: a cohort
Declaration of Conflicting Interests
study. Am J Respir Crit Care Med. 2020;201(12):1560-1564.
The author(s) declared no potential conflicts of interest with respect to
doi:10.1164/rccm.202004-1163LE
the research, authorship, and/or publication of this article.
13. Auld SC, Caridi-Scheible M, Blum JM. ICU and ventilator mor-
tality among critically ill adults with coronavirus disease 2019.
Funding Cri Care Med. 2020;48(9):e799-e804. doi:10.1097/CCM.
The author(s) disclosed receipt of the following financial support for 0000000000004457
the research, authorship, and/or publication of this article: RRC 14. Mitra AR, Fergusson NA, Smith EL, et al. Baseline characteristics
received support from NIH T32HL116275 and is a fellow of the and outcomes of patients with COVID-19 admitted to intensive
Parker B. Francis Family Foundation. care units in Vancouver, Canada: a case series. CMAJ. 2020;
192(26):E694-E701. doi:10.1503/cmaj.200794
ORCID iDs 15. Schenck EJ, Hoffman K, Goyal P, et al. Respiratory mechanics
and gas exchange in COVID-19 associated respiratory failure.
David M. Dudzinski, MD https://orcid.org/0000-0001-9363-9345
Ann Am Thorac Soc. 2020;17(9):1158-1161. doi:10.1513/Annal-
C. Corey Hardin, MD, PhD https://orcid.org/0000-0002-4074-0420
sATS.202005-427RL
16. Bhatraju PK, Ghassemieh BJ, Nichols M, et al. Covid-19 in cri-
References tically ill patients in the Seattle region—case series. N Engl J
1. Wu F, Zhao S, Yu B, et al. A new coronavirus associated with Med. 2020;382(21):2012-2022.
human respiratory disease in China. Nature. 2020;579(7798): 17. Cummings MJ, Baldwin MR, Abrams D, et al. Epidemiology,
265-269. clinical course, and outcomes of critically ill adults with
2. WHO Coronavirus Disease (COVID-19) Dashboard. 2020. COVID-19 in New York city: a prospective cohort study. Lancet.
https://covid19.who.int/?gclid¼CjwKCAjw5cL2BRASEiw 2020;395(10239):1763-1770. doi:10.1016/S0140-6736(20)
AENqAPt0H4sec7Rz3KXpBfYUGg6qbsVfpBD4b2u8nHGZ- 31189-2
mxXraqSarZOLSxoCwgEQAvD_BwE. Accessed October 19, 18. Grasselli G, Zangrillo A, Zanella A, et al. Baseline characteristics
2020. and outcomes of 1591 patients infected with SARS-CoV-2 admit-
3. Alhazzani W, Møller MH, Arabi YM, et al. Surviving sepsis ted to ICUs of the Lombardy region, Italy. JAMA. 2020;323(16):
campaign: guidelines on the management of critically ill adults 1574-1581. doi:10.1001/jama.2020.5394
with coronavirus disease 2019 (COVID-19). Intensive Care Med. 19. Recovery Collaborative Group. Dexamethasone in hospitalized
2020;46(5):854-887. patients with Covid-19—preliminary report. N Engl J Med.
4. Marini JJ, Gattinoni L. Management of COVID-19 respiratory 2020;NEJMoa2021436. doi:10.1056/NEJMoa2021436
distress. JAMA. 2020;323(22):2329-2330. doi:10.1001/jama. 20. Beigel JH, Tomashek KM, Dodd LE. Remdesivir for the treat-
2020.6825 ment of Covid-19—preliminary report. N Engl J Med. 2020;
5. Gattinoni L., Coppola S, Cressoni M, Busana M, Rossi S, 383(10):994. doi:10.1056/NEJMoa2007764
Chiumello D. COVID-19 does not lead to a ‘typical’ acute 21. Li Q, Guan X, Wu P, et al. Early transmission dynamics in
respiratory distress syndrome. Am J Respir Crit Care Med. Wuhan, China, of novel coronavirus-infected pneumonia. N Engl
2020;201(10):1299-1300. J Med. 2020;382:1199-1207.
Chivukula et al 33

22. Zhang S, Diao M, Yu W, Pei L, Lin Z, Chen D. Estimation of the 38. Yu IT, Li Y, Wong TW, et al. Evidence of airborne transmission
reproductive number of novel coronavirus (COVID-19) and the of the severe acute respiratory syndrome virus. N Engl J Med.
probable outbreak size on the diamond princess cruise ship: a 350(17):1731-1739.
data-driven analysis. Int J Infect Dis. 2020;93:201-204. 39. Seto WH, Tsang D, Yung RW, et al. Effectiveness of precautions
23. Mortality Analyses. Johns Hopkins Coronavirus Resource Center. against droplets and contact in prevention of nosocomial trans-
2020. https://coronavirus.jhu.edu/data/mortality. Accessed Octo- mission of severe acute respiratory syndrome (SARS). Lancet.
ber 19, 2020. 2003;361(9368):1519-1520.
24. Arabi YM, Arifi AA, Balkhy HH, et al. Clinical course and out- 40. Ong SWX, Tan YK, Chia PY, et al. Air, surface environmental,
comes of critically ill patients with Middle East respiratory syn- and personal protective equipment contamination by severe acute
drome coronavirus infection. Ann Intern Med. 2014;160:389-397. respiratory syndrome coronavirus 2 (SARS-CoV-2) from a symp-
25. Cumulative Cases. Johns Hopkins Coronavirus Resource Center. tomatic patient. JAMA. 2020;323(16):1610-1612. doi:10.1001/
https://coronavirus.jhu.edu/data/cumulative-cases. Accessed jama.2020.3227
October 19, 2020. 41. Beigel JH, Omashek KM, Dodd LE, et al. Remdesivir for the
26. Tuite AR, Fisman DN, Greer AL. Mathematical modelling of treatment of covid-19—preliminary report. N Engl J Med.
COVID-19 transmission and mitigation strategies in the popula- 2020;383(10):994.
tion of Ontario, Canada. CMAJ. 2020;192(19):E497-E505. doi: 42. Li W, Moore MJ, Vasilieva N, et al. Angiotensin-converting
10.1503/cmaj.200476 enzyme 2 is a functional receptor for the SARS coronavirus.
27. Grasselli G, Pesenti A, Cecconi M. Critical care utilization for the Nature. 2003;426(6965):450-454.
COVID-19 outbreak in Lombardy, Italy: early experience and 43. Jeffers SA, Tusell SM, Gillim-Ross L, et al. CD209 L (L-SIGN) is
forecast during an emergency response. JAMA. 2020. 323(16): a receptor for severe acute respiratory syndrome coronavirus.
Proc Natl Acad Sci USA. 2004;101(44):15748-15753.
1545-1546. doi:10.1001/jama.2020.4031
44. Ziegler CGK, Allon SJ, Nyquist SK, et al. SARS-CoV-2 receptor
28. Zhong NS, Zheng BJ, Li YM, et al. Epidemiology and cause of
ACE2 is an interferon-stimulated gene in human airway epithelial
severe acute respiratory syndrome (SARS) in Guangdong, Peo-
cells and is detected in specific cell subsets across tissues. Cell.
ple’s Republic of China, in February, 2003. Lancet. 2003;
2020;181(5):1016-1035. e19.
362(9393):1353-1358.
45. Hamming I, Timens W, Bulthuis ML, Lely AT, Navis GV, van
29. Viruses CS. G. of T. I. C. on T. of & Coronaviridae Study Group
Goor H. A first step in understanding SARS pathogenesis.
of the International Committee on Taxonomy of Viruses. The
J Pathol. 2004;203(2):631-637.
species severe acute respiratory syndrome-related coronavirus:
46. Wang X, Xu W, Hu G, et al. SARS-CoV-2 infects T lymphocytes
classifying 2019-nCoV and naming it SARS-CoV-2. Nat Micro-
through its spike protein-mediated membrane fusion. Cell Mol
biol. 2020;5:536-544.
Immunol. 2020;1-3. doi:10.1038/s41423-020-0424-9
30. Wong SY, Kwong RS, Wu TC, et al. Risk of nosocomial trans-
47. Liu J, Cao R, Xu M, et al. Hydroxychloroquine, a less toxic deri-
mission of coronavirus disease 2019: an experience in a general
vative of chloroquine, is effective in inhibiting SARS-CoV-2
ward setting in Hong Kong. J Hosp Infect. 2020;105(2):119-127.
infection in vitro. Cell Discov. 2020;6:16.
31. Bai Y, Yao L, Wei T, et al. Presumed asymptomatic carrier trans-
48. Snijder EJ, Decroly E, Ziebuhr J. The nonstructural proteins
mission of COVID-19. JAMA. 2020;323(14):1406-1407. doi:10. directing coronavirus RNA synthesis and processing. Adv Virus
1001/jama.2020.2565 Res. 2016;96:59-126.
32. Rothe C, Schunk M, Sothmann P, et al. Transmission of 2019- 49. Anand K, Ziebuhr J, Wadhwani P, Mesters JR, Hilgenfeld R.
nCoV infection from an asymptomatic contact in Germany. Coronavirus main proteinase (3CLpro) structure: basis for design
N Engl J Med. 2020;382(10):970-971. of anti-SARS drugs. Science. 2003;300:1763-1767.
33. van Doremalen N, Bushmaker T, Morris DH, et al. Aerosol 50. Xu Z, Shi L, Wang Y, et al. Pathological findings of COVID-19
and surface stability of SARS-CoV-2 as compared with associated with acute respiratory distress syndrome. Lancet
SARS-CoV-1. N Engl J Med. 2020;382(16):1564-1567. Respir Med. 2020;8(4):420-422.
34. Wei J, Li Y. Airborne spread of infectious agents in the indoor 51. Schaller T, Hirschbühl K, Burkhardt K, et al. Postmortem exam-
environment. Am J Infect Control. 2016;44(9):S102-S108. ination of patients with COVID-19. JAMA. 2020;323(24):
35. Ran L, Chen X, Wang Y, Wu W, Zhang L, Tan X. Risk factors of 2518-2520. doi:10.1001/jama.2020.8907
healthcare workers with corona virus disease 2019: a retrospec- 52. Zhang H, Zhou P, Wei Y, et al. Histopathologic changes and
tive cohort study in a designated hospital of Wuhan in China. Clin SARS-CoV-2 immunostaining in the lung of a patient with
Infect Dis. 2020;ciaa287. doi:10.1093/cid/ciaa287 COVID-19. Ann Intern Med. 2020;172(9):629-632.
36. Roy CJ, Milton DK. Airborne transmission of communicable 53. Ackermann M, Verleden SE, Kuehnel M, et al. Pulmonary vas-
infection—the elusive pathway. N Engl J Med. 2004;350(17): cular endothelialitis, thrombosis, and angiogenesis in Covid-19.
1710-1712. N Engl J Med. 2020;383(2):120-128. doi:10.1056/NEJMoa
37. Bourouiba L. Turbulent gas clouds and respiratory pathogen 2015432
emissions: potential implications for reducing transmission of 54. Ding Y, Wang H, Shen H, et al. The clinical pathology of severe
COVID-19. JAMA. 2020;323(18):1837-1838. doi:10.1001/jama. acute respiratory syndrome (SARS): a report from China.
2020.4756 J Pathol. 2003;200(3):282-289.
34 Journal of Intensive Care Medicine 36(1)

55. Ng DL, Hosani F, Keating MK, et al. Clinicopathologic, immu- China. JAMA Cardiol. 2020;5(7):802-810. doi:10.1001/jamacar-
nohistochemical, and ultrastructural findings of a fatal case of dio.2020.0950
Middle East respiratory syndrome coronavirus infection in the 70. Bai HX, Hsieh B, Xiong Z, et al. Performance of radiologists in
United Arab Emirates, April 2014. Am J Pathol. 2016;186(3): differentiating COVID-19 from viral pneumonia on chest CT.
652-658. Radiology. 2020;296(2):E46-E54.
56. Ni L, Ye F, Cheng ML, et al. Detection of SARS-CoV-2-specific 71. Ferguson ND, Fan E, Camporota L, et al. The Berlin definition of
humoral and cellular immunity in COVID-19 convalescent indi- ARDS: an expanded rationale, justification, and supplementary
viduals. Immunity. 2020;52(6):971-977. doi:10.1016/j.immuni. material. Intensive Care Med. 2012;38(10):1573-1582.
2020.04.023 72. Archer SL, Sharp WW, Weir EK. Differentiating COVID-19
57. Xiang F, Ye F, Cheng ML, et al. Antibody detection and dynamic pneumonia from acute respiratory distress syndrome and high
characteristics in patients with COVID-19. Clin Infect Dis. 2020; altitude pulmonary edema. Circulation. 2020;142(2):101-104.
ciaa461. doi:10.1093/cid/ciaa461 73. Solaimanzadeh I. Acetazolamide, nifedipine and phosphodiester-
58. Rokni M, Ghasemi V, Tavakoli Z. Immune responses and patho- ase inhibitors: rationale for their utilization as adjunctive counter
genesis of SARS-CoV-2 during an outbreak in Iran: comparison measures in the treatment of coronavirus disease 2019
with SARS and MERS. Rev Med Virol. 2020;30:e2107. (COVID-19). Cureus. 2020;12(3):e7343. doi:10.7759/cureus.
59. Chandrashekar A, Liu J, Martinot AJ, et al. SARS-CoV-2 infec- 7343
tion protects against rechallenge in rhesus macaques. Science. 74. Beitler JR, Thompson BT, Matthay MA, et al. Estimating dead-
2020;369(6505):812-817. doi:10.1126/science.abc4776 space fraction for secondary analyses of acute respiratory distress
60. Guan WJ, Ni ZY, Hu Y, et al. Clinical characteristics of corona- syndrome clinical trials. Crit Care Med. 2015;43(5):1026-1035.
virus disease 2019 in China. N Engl J Med. 2020;382(18): 75. Haudebourg AF, Perier F, Tuffet S, et al. Respiratory mechanics
1708-1720. of COVID-19 vs. non-COVID-19 associated acute respiratory
61. Sedaghat AR, Gengler I, Speth MM. Olfactory dysfunction: a distress syndrome. Am J Respir Crit Care Med. 2020;202(2):
highly prevalent symptom of COVID-19 with public health sig- 287-290 doi:10.1164/rccm.202004-1226LE
nificance. Otolaryngol Head Neck Surg. 2020;163(1):12-15. 76. Luks AM, Swenson ER. COVID-19 lung injury and high altitude
62. Cholankeril G, Podboy A, Aivaliotis VI, et al. High prevalence of pulmonary edema: a false equation with dangerous implications
concurrent gastrointestinal manifestations in patients with severe [published online April 24, 2020]. Ann Am Thorac Soc. 2020. doi:
acute respiratory syndrome coronavirus 2: early experience from 10.1513/AnnalsATS.202004-327FR
California. Gastroenterology. 2020;159(2):775-777. doi:10.1053/ 77. Mehta P, McAuley DF, Brown M, et al. COVID-19: consider
j.gastro.2020.04.008 cytokine storm syndromes and immunosuppression. Lancet.
63. Yang X, Yu Y, Xu J, et al. Clinical course and outcomes of 2020;395(10229);1033-1034.
critically ill patients with SARS-CoV-2 pneumonia in Wuhan, 78. Wong CK, Lam CWK, Wu AKL, et al. Plasma inflammatory
China: a single-centered, retrospective, observational study. Lan- cytokines and chemokines in severe acute respiratory syndrome.
cet Respir Med. 2020;8(5):475-481. Clin Exp Immunol. 2004;136(1):95-103.
64. Bellani G, Laffey JG, Pham T, et al. Epidemiology, patterns of 79. de Jong MD, Simmons CP, Thanh TT, et al. Fatal outcome of
care, and mortality for patients with acute respiratory distress human influenza A (H5N1) is associated with high viral load and
syndrome in intensive care units in 50 countries. JAMA. 2016; hypercytokinemia. Nat Med. 2006;12(10):1203-1207.
315:788-800. 80. Zhu Z, Cai T, Fan T, et al. Clinical value of immune-
65. Ruan Q, Yang K, Wang W, Jiang L, Song J. Correction to: Clin- inflammatory parameters to assess the severity of coronavirus
ical predictors of mortality due to COVID-19 based on an analysis disease 2019. Int J Infect Dis.2020;95:332-339.
of data of 150 patients from Wuhan, China [published online 81. Strauss R, Neureiter D, Westenburger B, Wehler M, Kirchner T,
April 6, 2020]. Intensive Care Med. 2020. doi:10.1007/s00134- Hahn EG. Multifactorial risk analysis of bone marrow histiocytic
020-06028-z hyperplasia with hemophagocytosis in critically ill medical
66. Wu C, Chen X, Cai Y, et al. Risk factors associated with acute patients—a postmortem clinicopathologic analysis. Crit Care
respiratory distress syndrome and death in patients with corona- Med. 2004;32(6):1316.
virus disease 2019 pneumonia in Wuhan, China. JAMA Intern 82. Inai K, Noriki S, Iwasaki H, Naiki H. Risk factor analysis for bone
Med. 2020;180(7):934-943. doi:10.1001/jamainternmed.2020. marrow histiocytic hyperplasia with hemophagocytosis: an
0994 autopsy study. Virchows Arch. 2014;465(1):109-118.
67. Wang D, Hu B, Hu C, et al. Clinical characteristics of 138 hospi- 83. Wilson JG, Simpson LJ, Ferreira AM, et al. Cytokine profile in
talized patients with 2019 novel coronavirus-infected pneumonia plasma of severe COVID-19 does not differ from ARDS and
in Wuhan, China. JAMA. 2020;323(11):1061-1069. sepsis. JCI Insight. 2020;5(17):e140289. doi:10.1172/jci.insight.
68. Zhou F, Yu T, Du R., et al. Clinical course and risk factors for 140289
mortality of adult inpatients with COVID-19 in Wuhan, China: a 84. Prescott HC, Calfee CS, Thompson BT, Angus DC, Liu VX.
retrospective cohort study. Lancet. 2020;395(11):1054-1062. Toward smarter lumping and smarter splitting: rethinking strate-
69. Shi S, Qin M, Shen B, et al. Association of cardiac injury with gies for sepsis and acute respiratory distress syndrome clinical
mortality in hospitalized patients with COVID-19 in Wuhan, trial design. Am J Respir Crit Care Med. 2016;194(2):147-155.
Chivukula et al 35

85. Calfee CS, Delucchi K, Parsons PE, et al. Subphenotypes in acute 100. Kirkpatrick JN, Mitchell C, Taub C, Kort S, Hung J, Swami-
respiratory distress syndrome: latent class analysis of data from nathan M. ASE statement on protection of patients and echocar-
two randomised controlled trials. Lancet Respir Med. 2014;2(8): diography service providers during the 2019 novel coronavirus
611-620. outbreak. J Am Coll Cardiol. 2020;33(6):648-653.
86. Famous KR, Delucchi K, Ware LB, et al. Acute respiratory dis- 101. Centers for Disease Control and Prevention. Coronavirus
tress syndrome subphenotypes respond differently to randomized Disease 2019 (COVID-19). Centers for Disease Control and
fluid management strategy. Am J Respir Crit Care Med. 2017; Prevention. 2020. https://www.cdc.gov/coronavirus/2019-
195(3):331-338. ncov/hcp/infection-control-recommendations.html. Accessed
87. Sinha P, Delucchi KL, McAuley DF, O’Kane CM, Matthay MA, October 19, 2020.
Calfee CS. Development and validation of parsimonious algo- 102. Durante-Mangoni E, Andini R, Bertolino L, et al. Low rate of
rithms to classify acute respiratory distress syndrome phenotypes: severe acute respiratory syndrome coronavirus 2 spread
a secondary analysis of randomised controlled trials. Lancet among health-care personnel using ordinary personal
Respir Med. 2020;8(3):247-257. protection equipment in a medium-incidence setting. Clin
88. Qin C, Zhou L, Hu Z, et al. Dysregulation of immune response in Microbiol Infect. 2020;26(9):1269-1270. doi:10.1016/j.cmi.
patients with COVID-19 in Wuhan, China. Clin Infect Dis. 2020; 2020.04.042
71(15):762-768. doi:10.1093/cid/ciaa248 103. Bartoszko JJ, Farooqi MAM, Alhazzani W, Loeb M.Medical
89. Sinha P, Matthay MA, Calfee CS. Is a ‘cytokine storm’ relevant to masks vs N95 respirators for preventing COVID-19 in health-
COVID-19? JAMA Intern Med. 2020;180(9):1152-1154. doi:10. care workers: a systematic review and meta-analysis of rando-
1001/jamainternmed.2020.3313 mized trials. Influenza Other Respir Viruses. 2020;14(4):
90. Kox M, Waalders NJB, Kooistra EJ, Gerretsen J, Pickkers P. 365-373.
Cytokine levels in critically ill patients with COVID-19 and other 104. Leung NHL, Chu DKW, Shiu EYC, et al. Respiratory virus
shedding in exhaled breath and efficacy of face masks. Nat Med.
conditions. JAMA. 2020:e2017052. Advance online September
2020;26(5):676-680. doi:10.21203/rs.3.rs-16836/v1
03, 2020. doi:10.1001/jama.2020.17052
105. Davidson JE, Aslakson RA, Long AC, et al. Guidelines for
91. Wang W, Xu Y, Gao R, et al. Detection of SARS-CoV-2 in
family-centered care in the neonatal, pediatric, and adult ICU.
different types of clinical specimens. JAMA. 2020;323(18):
Crit Care Med. 2017;45(1):103-128.
1843-1844. doi:10.1001/jama.2020.3786
106. Mehrotra P, Shane AL, Milstone AM. Family-centered care and
92. Tu Y.-P, Jennings R, Hart B, et al. Swabs collected by patients or
high-consequence pathogens: thinking outside the room. JAMA
health care workers for SARS-CoV-2 testing. N Engl J Med.
Pediatr. 2015;169(11):985-986.
2020;383(5):494-496. doi:10.1056/NEJMc2016321
107. Maunder RG, Lancee WJ, Balderson KE, et al. Long-term psy-
93. Zou L, Ruan F, Huang M, et al. SARS-CoV-2 viral load in upper
chological and occupational effects of providing hospital health-
respiratory specimens of infected patients. N Engl J Med. 2020;
care during SARS outbreak. Emerg Infect Dis. 2006;12(12):
382(12):1177-1179.
1924-1932.
94. Ai T, Yang Z, Hou H, et al. Correlation of chest CT and RT-PCR
108. Gattinoni L, Chiumello D, Rossi S. COVID-19 pneumonia:
testing in coronavirus disease 2019 (COVID-19) in China: a
ARDS or not? Crit Care. 2020;24(1):154.
report of 1014 cases. Radiology. 2020;296(2):E32-E40. doi:10. 109. Bos LDJ, Paulus F, Vlaar APJ, Beenen LFM, Schultz MJ. Sub-
1148/radiol.2020200642 phenotyping ARDS in COVID-19 patients: consequences for
95. ACR Recommendations for the Use of Chest Radiography and ventilator management. Ann Am Thoracic Soc. 2020. doi:10.
Computed Tomography (CT) for Suspected COVID-19 Infection. 1513/annalsats.202004-376rl
2020. https://www.acr.org/Advocacy-and-Economics/ACR-Posi 110. Rochwerg B, Granton D, Wang DX, et al. High flow nasal
tion-Statements/Recommendations-for-Chest-Radiography-and- cannula compared with conventional oxygen therapy for acute
CT-for-Suspected-COVID19-Infection. Accessed October 19, hypoxemic respiratory failure: a systematic review and meta-
2020. analysis. Intensive Care Med. 2019;45(5):563-572. doi:10.
96. Yu F, Yan L, Wang N, et al. Quantitative detection and viral load 1007/s00134-019-05590-5
analysis of SARS-CoV-2 in infected patients. Clin Infect Dis. 111. Patel BK, Wolfe KS, Hall JB, Kress JP. A word of caution
2020;71(15):793-798. doi:10.1093/cid/ciaa345 regarding patient self-inflicted lung injury and prophylactic intu-
97. Wahidi MM, Shojaee S, Lamb CR, et al. The use of bronchoscopy bation. Am J Respir Crit Care Med. 2017;196(7):936.
during the COVID-19 pandemic: CHEST/AABIP guideline and 112. Brochard L, Slutsky A, Pesenti A. Mechanical ventilation to
expert panel report. Chest. 2020;158(3):1268-1281. doi:10.1016/ minimize progression of lung injury in acute respiratory failure.
j.chest.2020.04.036 Am J Respir Crit Care Med. 2017;195(4):438-442.
98. Darmon M, Clec’h C, Adrie C, et al. Acute respiratory distress 113. Mascheroni D, Kolobow T, Fumagalli R, Moretti MP, Chen V,
syndrome and risk of AKI among critically ill patients. Clin J Am Buckhold D. Acute respiratory failure following pharmacologi-
Soc Nephrol. 2014;9(8):1347-1353. cally induced hyperventilation: an experimental animal study.
99. Fan Z, Chen L, Li J, et al. Clinical features of COVID-19-related Intensive Care Med. 1988;15(1):8-14.
liver functional abnormality. Clin Gastroenterol Hepatol. 2020; 114. Yoshida T, Uchiyama A, Matsuura N, Mashimo T, Fujino Y.
18(7):1561-1566. Spontaneous breathing during lung-protective ventilation in an
36 Journal of Intensive Care Medicine 36(1)

experimental acute lung injury model. Crit Care Med. 2012; 130. Alraddadi BM, Qushmaq I, Al-Hameed FM, et al. Noninvasive
40(5):1578-1585. ventilation in critically ill patients with the Middle East respira-
115. Tonelli R, Fantini R, Tabbı̀ L, et al. Early inspiratory effort tory syndrome. Influenza Other Respir Viruses. 2019;13(4):
assessment by esophageal manometry predicts noninvasive ven- 382-390.
tilation outcome in de novo respiratory failure. A Pilot Study. 131. Kaiser SK. Cardiac arrest and mortality related to intubation
Am J Respir Crit Care Med. 2020;202(4):558-567. procedure in critically ill adult patients: a multicenter cohort
116. Tobin MJ, Laghi F, Jubran A. Caution about early intubation and study. J Emerg Med. 2018;54:903-904.
mechanical ventilation in COVID-19. Ann Intensive Care. 2020; 132. Mort TC. The incidence and risk factors for cardiac arrest during
10(1):78. emergency tracheal intubation: a justification for incorporating
117. Parke RL, McGuinness SP. Pressures delivered by nasal high the ASA Guidelines in the remote location. J Clin Anesth. 2004;
flow oxygen during all phases of the respiratory cycle. Respir 16(7):508-516.
Care. 2013;58(10):1621-1624. 133. Acute Respiratory Distress Syndrome Network; Brower RG,
118. Karbing DS, Panigada M, Bottino N, et al. Changes in shunt, Matthay MA, et al. Ventilation with lower tidal volumes as
ventilation/perfusion mismatch, and lung aeration with PEEP in compared with traditional tidal volumes for acute lung injury
patients with ARDS: a prospective single-arm interventional and the acute respiratory distress syndrome. N Engl J Med. 2000;
study. Crit Care. 2020;24(1):111. 342(18):1301-1308.
119. Kotoda M, Hishiyama S, Mitsui K, et al. Assessment of the 134. Walkey AJ, Goligher EC, Sorbo LD, et al. Low tidal volume
potential for pathogen dispersal during high-flow nasal therapy. versus non-volume-limited strategies for patients with acute
J Hosp Infect. 2020;104(4):534-537. respiratory distress syndrome. A systematic review and meta-
120. Ip M, Tang JW, Hui DSC, et al. Airflow and droplet spreading analysis. Ann Am Thorac Soc. 2017;14(supplement_4):
around oxygen masks: a simulation model for infection control
S271-S279.
research. Am J Infect Contr. 2007;35(10):684-689.
135. Zangrillo A, Beretta L, Scandroglio AM, et al. Characteristics,
121. Hui DS, Chow BK, Lo T, et al. Exhaled air dispersion during
treatment, outcomes and cause of death of invasively ventilated
high-flow nasal cannula therapy versus CPAP via different
patients with COVID-19 ARDS in Milan, Italy. Crit Care
masks. Eur Respir J. 2019;53(4):1802339.
Resusc. 2020. Advance online April 23, 2020.
122. Leonard S, Atwood CW Jr, Walsh BK, et al. Preliminary find-
136. Harris RS. Pressure-volume curves of the respiratory system.
ings on control of dispersion of aerosols and droplets during
Respir Care. 2005;50(1):78-98; discussion 98-99.
high-velocity nasal insufflation therapy using a simple surgical
137. Hickling KG. Best compliance during a decremental, but not
mask: implications for the high-flow nasal cannula. Chest. 2020;
incremental, positive end-expiratory pressure trial is related to
158(3):1046-1049. doi:10.1016/j.chest.2020.03.043
open-lung positive end-expiratory pressure. Am J Respir Crit
123. Frat J-P, Thille AW, Mercat A, et al. High-flow oxygen through
Care Med. 2001;163(1):69-78.
nasal cannula in acute hypoxemic respiratory failure. N Engl J
138. Briel M, Meade M, Mercat A, et al. Higher vs lower positive
Med. 2015;372(23):2185-2196.
end-expiratory pressure in patients with acute lung injury and
124. Bellani G, Laffey JG, Pham T, et al. Noninvasive ventilation of
acute respiratory distress syndrome: systematic review and
patients with acute respiratory distress syndrome. Insights from
meta-analysis. JAMA. 2010;303(9):865-873.
the LUNG SAFE study. Am J Respir Crit Care Med. 2017;
139. Beitler JR, Sarge T, Banner-Goodspeed VM, et al. Effect of
195(1):67-77.
125. Ferreyro BL, Angriman F, Munshi L, et al. Association of non- titrating positive end-expiratory pressure (PEEP) with an eso-
invasive oxygenation strategies with all-cause mortality in adults phageal pressure-guided strategy vs an empirical high
with acute hypoxemic respiratory failure: a systematic review PEEP-FiO2 strategy on death and days free from mechanical
and meta-analysis. JAMA. 2020;324(1):57-67. doi:10.1001/ ventilation among patients with acute respiratory distress syn-
jama.2020.9524. drome: a randomized clinical trial. JAMA. 2019;321(9):846-857.
126. Poston JT, Patel BK, Davis AM. Management of critically ill 140. Brower RG, Lanken PN, MacIntyre N, et al. Higher versus lower
adults with COVID-19. JAMA. 2020;323(18):1839-1841. doi: positive end-expiratory pressures in patients with the acute
10.1001/jama.2020.4914 respiratory distress syndrome. N Engl J Med. 2004;351(4):
127. Osadnik CR, Tee VS, Carson-Chahhoud KS, Picot J, Wedzicha 327-336.
JA, Smith BJ. Non-invasive ventilation for the management of 141. Writing Group for the Alveolar Recruitment for Acute Respira-
acute hypercapnic respiratory failure due to exacerbation of tory Distress Syndrome Trial (ART) Investigators; Cavalcanti
chronic obstructive pulmonary disease. Cochrane Database Syst AB, Suzumura EA, Laranjeira LN, et al. Effect of lung recruit-
Rev. 2017;7(7):CD004104. ment and titrated positive end-expiratory pressure (PEEP) vs low
128. Mead J, Takishima T, Leith D. Stress distribution in lungs: a PEEP on mortality in patients with acute respiratory distress
model of pulmonary elasticity. J Appl Physiol. 1970;28(5): syndrome: a randomized clinical trial. JAMA. 2017;318(14):
596-608. 1335-1345.
129. Aragón RE, Proaño A, Mongilardi N, et al. Sedation practices 142. Amato MBP, Meade MO, Slutsky AS, et al. Driving pressure
and clinical outcomes in mechanically ventilated patients in a and survival in the acute respiratory distress syndrome. N Engl J
prospective multicenter cohort. Crit Care. 2019;23(1):130. Med. 2015;372(8):747-755.
Chivukula et al 37

143. Figueroa-Casas JB, Montoya R. Effect of tidal volume size and 160. Fan E, Sorbo LD, Goligher EC, et al. An official American
its delivery mode on patient-ventilator dyssynchrony. Ann Am Thoracic Society/European Society of Intensive Care Medi-
Thorac Soc. 2016;13(12):2207-2214. cine/Society of Critical Care Medicine Clinical Practice Guide-
144. Jaber S, Delay J-M, Matecki S, Sebbane M, Eledjam J-J, line: mechanical ventilation in adult patients with acute
Brochard L. Volume-guaranteed pressure-support ventilation respiratory distress syndrome. Am J Respir Crit Care Med.
facing acute changes in ventilatory demand. Intensive Care 2017;195(9):1253-1263.
Med. 2005;31(9):1181-1188. 161. Scholten EL, Beitler JR, Prisk GK, Malhotra A. Treatment of
145. Maxwell RA, Green JM, Waldrop J, et al. A randomized pro- ARDS with prone positioning. Chest. 2017;151(1):215-224.
spective trial of airway pressure release ventilation and low tidal 162. Guérin C, Reignier J, Richard J-C, et al. Prone positioning in
volume ventilation in adult trauma patients with acute respira- severe acute respiratory distress syndrome. N Engl J Med. 2013;
tory failure. J Trauma. 2010;69(3):501-510; discussion 511. 368(23):2159-2168.
146. Branson RD, Johannigman JA. The role of ventilator graphics 163. Sud S, Friedrich JO, Taccone P, et al. Prone ventilation reduces
when setting dual-control modes. Respir Care. 2005;50(2): mortality in patients with acute respiratory failure and severe
187-201. hypoxemia: systematic review and meta-analysis. Intensive
147. Ridgeon EE, Young PJ, Bellomo R, Mucchetti M, Lembo R, Care Med. 2010;36(4):585-599.
Landoni G. The Fragility index in multicenter randomized con- 164. Ding L, Wang L, Ma W, He H. Efficacy and safety of early
trolled critical care trials. Crit Care Med. 2016;44(7):1278-1284. prone positioning combined with HFNC or NIV in moderate
148. ICU-ROX Investigators and the Australian and New Zealand to severe ARDS: a multi-center prospective cohort study. Crit
Intensive Care Society Clinical Trials Group; Mackle D, Care. 2020;24(1):28.
Bellomo R, Bailey M, et al. Conservative oxygen therapy during 165. Elharrar X, Trigui Y, Dols A-M, et al. Use of prone positioning
mechanical ventilation in the ICU. N Engl J Med. 2020;382(11):
in nonintubated patients with COVID-19 and hypoxemic acute
989-998.
respiratory failure. JAMA. 2020;323(22):2336-2338. doi:10.
149. Barrot L, Asfar P, Mauny F, et al. Liberal or conservative oxy-
1001/jama.2020.8255
gen therapy for acute respiratory distress syndrome. N Engl J
166. Sun Q, Qiu H, Huang M, Yang Y. Lower mortality of
Med. 2020;382(11):999-1008.
COVID-19 by early recognition and intervention: experience
150. Henderson WR, Chen L, Amato MBP, Brochard LJ. Fifty years of
from Jiangsu Province. Ann Intensive Care. 2020;10(1):33.
research in ARDS. Respiratory mechanics in acute respiratory dis-
167. Thompson AE, Ranard BL, Wei Y, Jelic S. Prone positioning in
tress syndrome. Am J Respir Crit Care Med. 2017;196(7):822-833.
awake, nonintubated patients with COVID-19 hypoxemic
151. Behazin N, Jones SB, Cohen RI, Loring SH. Respiratory restric-
respiratory failure. JAMA Intern Med. 2020. doi:10.1001/
tion and elevated pleural and esophageal pressures in morbid
jamainternmed.2020.3030
obesity. J Appl Physiol. 2010;108(1):212-218.
168. Ng Z, Tay WC, Ho CHB. Awake prone positioning for non-
152. Armaganidis A, Stavrakaki-Kallergi K, Koutsoukou A,
intubated oxygen dependent COVID-19 pneumonia patients.
Lymberis A, Milic-Emili J, Roussos C. Intrinsic positive end-
Eur Respir J. 2020;56(1):2001198.
expiratory pressure in mechanically ventilated patients with and
169. Munshi L, Sorbo LD, Adhikari NKJ, et al. Prone position for
without tidal expiratory flow limitation. Crit Care Med. 2000;
28(12):3837-3842. acute respiratory distress syndrome. A systematic review and
153. Junhasavasdikul D, Telias I, Grieco DL, et al. Expiratory flow meta-analysis. Ann Am Thorac Soc. 2017;14(Supplement_4):
limitation during mechanical ventilation. Chest. 2018;154(4): S280-S288.
948-962. 170. Romero CM, Cornejo RA, Gálvez LR, et al. Extended prone
154. Dhand R. Ventilator graphics and respiratory mechanics in the position ventilation in severe acute respiratory distress syn-
patient with obstructive lung disease. Respir Care. 2005;50(2): drome: a pilot feasibility study. J Crit Care. 2009;24(1):81-88.
246-261; discussion 259-261. 171. Mauri T, Cambiaghi B, Spinelli E, Langer T, Grasselli G. Spon-
155. Hess D, MacIntyre NR, Galvin WF. Respiratory Care: Princi- taneous breathing: a double-edged sword to handle with care.
ples and Practice. Jones & Bartlett Learning; 2020. Ann Transl Med. 2017;5(14):292.
156. Suter PM, Fairley B, Isenberg MD. Optimum end-expiratory 172. Hess DR, Thompson BT. Patient-ventilator dyssynchrony during
airway pressure in patients with acute pulmonary failure. N Engl lung protective ventilation: what’s a clinician to do?* Crit Care
J Med. 1975;292(6):284-289. Med. 2006;34(1):231-233.
157. Wayne KS.Positive end-expiratory pressure (PEEP) ventilation. 173. Heart N, Moss M, Huang DT, et al. Early neuromuscular block-
A review of mechanisms and actions. JAMA. 1976;236(12): ade in the acute respiratory distress syndrome. N Engl J Med.
1394-1396. 2019;380(21):1997-2008.
158. Chiumello D, Coppola S, Froio S, et al. Time to reach a new 174. Blokpoel RG, Wolthuis DW, Koopman AA, Kneyber MC.
steady state after changes of positive end expiratory pressure. Reverse triggering: a novel type of patient-ventilator asynchrony
Intensive Care Med. 2013;39(8):1377-1385. in mechanically ventilated children. Am J Respir Crit Care Med.
159. Suter PM, Barrie Fairley H, Isenberg MD. Optimum end- 2019;200(2):e4-e5.
expiratory airway pressure in patients with acute pulmonary 175. Gebistorf F, Karam O, Wetterslev J, Afshari A. Inhaled nitric
failure. N Engl J Med. 1975;292(6):284-289. oxide for acute respiratory distress syndrome (ARDS) in
38 Journal of Intensive Care Medicine 36(1)

children and adults. Cochrane Database Syst Rev. 2016(6): 192. Simpson S, Kay FU, Abbara S, et al. Radiological Society of
CD002787. doi:10.1002/14651858.cd002787.pub3 North America Expert consensus statement on reporting chest
176. Griffiths MJ, Evans TW. Inhaled nitric oxide therapy in adults. CT findings related to COVID-19. Endorsed by the Society of
New Engl J Med. 2005;353(25):2683-2695. Thoracic Radiology, the American College of Radiology, and
177. Afshari A, Bille AB, Allingstrup M. Aerosolized prostacyclins RSNA. Radiology. 2020;2(2):e200152.
for acute respiratory distress syndrome (ARDS). Cochrane 193. Connors JM, Levy JH. COVID-19 and its implications for
Database Syst Rev. 2017;7(7):CD007733. doi:10.1002/ thrombosis and anticoagulation. Blood. 2020;135(23):
14651858.cd007733.pub3 2033-2040.
178. Croen KD. Evidence for antiviral effect of nitric oxide. Inhibi- 194. Rodrı́guez AH, Avilés-Jurado FX, Dı́az E, et al. Procalcitonin
tion of herpes simplex virus type 1 replication. J Clin Invest. (PCT) levels for ruling-out bacterial coinfection in ICU patients
1993;91(6):2446-2452. with influenza: a CHAID decision-tree analysis. J Infect. 2016;
179. Rimmelzwaan GF, Baars MM, de Lijster P, Fouchier RA, 72(2):143-151.
Osterhaus AD. Inhibition of influenza virus replication by nitric 195. Scales DC, Green K, Chan AK, et al. Illness in intensive care
oxide. Int Congr Ser. 2001;73(10):8880-8883. staff after brief exposure to severe acute respiratory syndrome.
180. Combes A, Hajage D, Capellier G, et al. Extracorporeal mem- Emerg Infect Dis. 2003;9(10):1205-1210.
brane oxygenation for severe acute respiratory distress syn- 196. Technical Guidance Publications: Transmission of SARS-CoV-
drome. N Engl J Med. 2018;378(21):1965-1975. 2: implications for infection prevention precautions. 2020.
181. Hardin CC, Hibbert K. ECMO for severe ARDS. New Engl J https://www.who.int/emergencies/diseases/novel-coronavirus-
Med. 2018:378(21):2032-2034. 2019/technical-guidance/infection-prevention-and-control.
182. Peek GJ. Efficacy and economic assessment of conventional Accessed October 19, 2020.
ventilatory support versus extracorporeal membrane oxygena- 197. Tran K, Cimon K, Severn M, Pessoa-Silva CL, Conly J. Aerosol
tion for severe adult respiratory failure (CESAR): a multicentre generating procedures and risk of transmission of acute respira-
tory infections to healthcare workers: a systematic review. PLoS
randomised controlled trial. Lancet. 2009;374(9698):1351-1363.
One. 2012;7(4):e35797.
183. Rajagopal K, Keller SP, Akkanti B, et al. Advanced pulmonary
198. Inciardi RM, Lupi L, Zaccone G, et al. Cardiac involvement in a
and cardiac support of COVID-19 patients: emerging recom-
patient with coronavirus disease 2019 (COVID-19). JAMA Car-
mendations from ASAIO-A ‘living working document’. ASAIO
diol. 2020;5(7):819-824. doi:10.1001/jamacardio.2020.1096
J. 2020;66(6):588-598.
199. Hu H, Ma F, Wei X, Fang Y. Coronavirus fulminant myocarditis
184. Ñamendys-Silva SA. ECMO for ARDS due to COVID-19.
saved with glucocorticoid and human immunoglobulin. Eur
Heart Lung. 2020;49(4):348-349.
Heart J. 2020;ehaa190. doi:10.1093/eurheartj/ehaa190
185. Bartlett RH, Ogino MT, Brodie D, et al. Initial ELSO guidance
200. Puelles VG, Lütgehetmann M, Lindenmeyer MT, et al. Multi-
document: ECMO for COVID-19 patients with severe cardio-
organ and renal tropism of SARS-CoV-2. N Engl J Med. 2020;
pulmonary failure. ASAIO J. 2020;66(5):472-474.
383(6):590-592. doi:10.1056/nejmc2011400
186. Shekar K, Badulak J, Peek G, et al. Extracorporeal life support
201. Caforio AL, Pankuweit S, Arbustini E, et al. Current state of
organization coronavirus disease 2019 interim guidelines: a con-
knowledge on aetiology, diagnosis, management, and therapy
sensus document from an international group of interdisciplinary
of myocarditis: a position statement of the European Society
extracorporeal membrane oxygenation providers. ASAIO J. of Cardiology Working Group on Myocardial and Pericardial
2020;66(7):707-721. Diseases. Eur Heart J. 2013;34(33):2636-2648.
187. Dellinger RP, Carlet JM, Masur H, et al. Surviving sepsis cam- 202. Sala S, Peretto G, Gramegna M, et al. Acute myocarditis pre-
paign guidelines for management of severe sepsis and septic senting as a reverse Tako-Tsubo syndrome in a patient with
shock. Crit Care Med. 2004;32(3):858-873. SARS-CoV-2 respiratory infection. Eur Heart J 2020;41(19):
188. Kalil AC, Metersky ML, Klompas M, et al. Management of 1861-1862.
adults with hospital-acquired and ventilator-associated pneumo- 203. Kaafarani HM, El Moheb M, Hwabejire JO, et al. Gastrointest-
nia: 2016 clinical practice guidelines by the Infectious Diseases inal complications in critically ill patients with COVID-19. Ann
Society of America and the American Thoracic Society. Clin Surg. 2020;272(2):e61-e62. doi:10.1097/SLA.00000000000
Infect Dis. 2016;63(5):e61-e111. 04004
189. Canadian Critical Care Trials Group. A randomized trial of diag- 204. Bhayana R, Som A, Li MD, et al. Abdominal imaging findings
nostic techniques for ventilator-associated pneumonia. N Engl J in COVID-19: preliminary observations. Radiology. 2020;
Med. 2006;355(25):2619-2630. 297(1):E207-E215.
190. Hellyer TP, Ewan V, Wilson P, Simpson AJ. The Intensive Care 205. Lamers MM, Beumer J, Van der Vaart J, et al. SARS-CoV-2
Society recommended bundle of interventions for the prevention productively infects human gut enterocytes. Science. 2020;
of ventilator-associated pneumonia. Pediatr Crit Care Med. 369(6499):50-54. doi:10.1126/science.abc1669
2016;17:238-243. 206. Parry AH, Wani AH, Yaseen M.Acute mesenteric ischemia in
191. Oba Y, Zaza T. Abandoning daily routine chest radiography in severe coronavirus-19 (COVID-19): possible mechanisms and
the intensive care unit: meta-analysis. Radiology. 2010;255(2): diagnostic pathway. Acad Radiol. 2020;369(6499):50-54. doi:
386-395. 10.1016/j.acra.2020.05.016
Chivukula et al 39

207. Grissom CK, Hirshberg EL, Dickerson JB, et al. Fluid manage- 225. Cornet AD, Groeneveld AJ, Hofstra JJ, et al. Recombinant
ment with a simplified conservative protocol for the acute human activated protein C in the treatment of acute respiratory
respiratory distress syndrome. Crit Care Med. 2015;43(2): distress syndrome: a randomized clinical trial. PLoS One. 2014;
288-295. 9(3):e90983.
208. Michard F, Teboul JL. Predicting fluid responsiveness in ICU 226. Hofstra JJ, Vlaar AP, Prins DJ, et al. Early intravenous unfrac-
patients: a critical analysis of the evidence. Chest. 2002;121(6): tionated heparin and outcome in acute lung injury and acute
2000-2008. respiratory distress syndrome—a retrospective propensity
209. Magder S. Fluid status and fluid responsiveness. Curr Opin Crit matched cohort study. BMC Pulm Med. 2012;12(1):43.
Care. 2010;16(4):289-296. 227. Spiezia L, Boscolo A, Poletto F, et al. COVID-19-related severe
210. National Heart, Lung, and Blood Institute Acute Respiratory hypercoagulability in patients admitted to intensive care unit for
Distress Syndrome (ARDS) Clinical Trials Network. Compari- acute respiratory failure. Thromb Haemost. 2020;120(6):
son of two fluid-management strategies in acute lung injury. 998-1000.
N Engl J Med. 2006;354(24):2564-2575. 228. Varga Z, Flammer AJ, Steiger P, et al. Endothelial cell infection
211. Lee J, De Louw E, Niemi M, et al. Association between fluid and endotheliitis in COVID-19. Lancet. 2020;395(10234):
balance and survival in critically ill patients. J Intern Med. 2015; 1417-1418.
277(4):468-477. 229. Goldsmith CS, Miller SE, Martines RB, Bullock HA, Zaki SR.
212. Boyd JH, Forbes J, Nakada TA, Walley KR, Russell JA. Fluid Electron microscopy of SARS-CoV-2: a challenging task. Lan-
resuscitation in septic shock: a positive fluid balance and ele- cet. 2020;395:e99.
vated central venous pressure are associated with increased mor- 230. Klok FA, Kruip MJ, Van der Meer NJ, et al. Incidence of throm-
tality. Crit Care Med. 2011;39(2):259-265. botic complications in critically ill ICU patients with
213. Finfer S, Bellomo R, Myburgh J, Norton R. A comparison of
COVID-19. Thromb Res. 2020;191:145-147.
albumin and saline for fluid resuscitation in the intensive care
231. Lodigiani C, Iapichino G, Carenzo L, et al. Venous and arterial
unit. N Engl J Med. 2004;350(22):2247-2256.
thromboembolic complications in COVID-19 patients admitted
214. Semler MW, Self WH, Wanderer JP, et al. Balanced crystalloids
to an academic hospital in Milan, Italy. Thromb Res. 2020;191:
versus saline in critically ill adults. N Engl J Med. 2018;378(9):
9-14.
829-839.
232. Poissy J, Goutay J, Caplan M, et al. Pulmonary embolism in
215. Annane D, Renault A, Brun-Buisson C, et al. Hydrocortisone
COVID-19 patients: awareness of an increased prevalence. Cir-
plus fludrocortisone for adults with septic shock. N Engl J Med.
culation. 2020;142(2):184-186. doi:10.1161/CIRCULATIO-
2018;378(9):809-818.
NAHA.120.047430
216. Venkatesh B, Finfer S, Cohen J, et al. Adjunctive glucocorticoid
233. Rice TW, Rubinson L, Uyeki TM, et al. Critical illness from
therapy in patients with septic shock. N Engl J Med. 2018;
2009 pandemic influenza A virus and bacterial coinfection in the
378(9):797-808.
United States. Crit Care Med. 2012;40(5):1487-1498.
217. Rhodes A, Evans LE, Evans LE, et al. Surviving sepsis cam-
234. Conti CB, Henchi S, Coppeta GP, Testa S, Grassia R. Bleeding
paign: international guidelines for management of sepsis and
in COVID-19 severe pneumonia: the other side of abnormal
septic shock. Crit Care Med. 2016;45(3):486-552.
218. Ni YN, Chen G, Sun J, Liang BM, Liang ZA. The effect of coagulation pattern? Eur J Intern Med. 2020;77:147-149. doi:
corticosteroids on mortality of patients with influenza pneumo- 10.1016/j.ejim.2020.05.002
nia: a systematic review and meta-analysis. Crit Car. 2019; 235. Doerschug K, Holley AB, Jimenez D, LeGal G, Rali P, Wells P.
23(1):99. Prevention, diagnosis and treatment of venous thromboembo-
219. Tsai MJ, Yang KY, Chan MC, et al. Impact of corticosteroid lism in patients with COVID-19: CHEST Guideline and Expert
treatment on clinical outcomes of influenza-associated ARDS: a Panel Report. Chest. 2020;158(3):1143-1163. doi:10.1016/j.
nationwide multicenter study. Ann Intensive Care. 2010;10(1):1. chest.2020.05.559
220. Arabi YM, Mandourah Y, Al-Hameed F, et al. Corticosteroid 236. Yuan M, Wu NC, Zhu X, et al. A highly conserved cryptic
therapy for critically ill patients with middle east respiratory epitope in the receptor binding domains of SARS-CoV-2 and
syndrome. Am J Respir Crit Care Med. 2018;197(6):757-767. SARS-CoV. Science. 2020;368(6491):630-633.
221. Stockman LJ, Bellamy R, Garner P. SARS: systematic review of 237. Wang C, Li W, Drabek D, et al. A human monoclonal antibody
treatment effects. PLoS Med. 2006;3(9):e343. blocking SARS-CoV-2 infection. Nat Commun. 2020;11(1):1-6.
222. Tomashefski JF Jr, Davies P, Boggis C, Greene R, Zapol WM, 238. Liu L, Wei Q, Lin Q, et al. Anti-spike IgG causes severe acute
Reid L. The pulmonary vascular lesions of the adult respiratory lung injury by skewing macrophage responses during acute
distress syndrome. Am J Pathol. 1983;112(1):112-126. SARS-CoV infection. JCI Insight. 2019;4(4):e123158.
223. Greene R, Lind S, Jantsch H, et al. Pulmonary vascular obstruc- 239. Monteil V, Kwon H, Prado P, et al. Inhibition of SARS-CoV-2
tion in severe ARDS: angiographic alterations after i.v. fibrino- infections in engineered human tissues using clinical-grade solu-
lytic therapy. AJR Am J Roentgenol. 1987;148(3):501-508. ble human ACE2. Cell. 2020;181(4):905-913. e7.
224. Glas GJ, Neto AS, Horn J, et al. Nebulized heparin for patients 240. Shen C, Wang Z, Zhao F, et al. Treatment of 5 critically ill
under mechanical ventilation: an individual patient data meta- patients with COVID-19 with convalescent plasma. JAMA.
analysis. Ann Intensive Care. 2016;6(1):33. 2020;323(16):1582-1589. doi:10.1001/jama.2020.4783
40 Journal of Intensive Care Medicine 36(1)

241. Joyner MJ, Senefeld JW, Klassen SA, et al. Effect of convales- 257. Williamson BN, Feldmann F, Schwarz B, et al. Clinical benefit
cent plasma on mortality among hospitalized patients with of remdesivir in rhesus macaques infected with SARS-CoV-2.
COVID-19: initial three-month experience. Medrxiv. 2020. Nature. 2020;585(7824):273-276. doi:10.1038/s41586-020-
doi:10.1101/2020.08.12.20169359 2423-5
242. Statement on Convalescent Plasma EUA. 2020. https://www. 258. Goldman JD, Lye DC, Hui DS, et al. Remdesivir for 5 or 10 days
covid19treatmentguidelines.nih.gov/statement-on-convales in patients with severe Covid-19. N Engl J Med. 2020. doi:10.
cent-plasma-eua/. Accessed October 19, 2020. 1056/NEJMoa2015301
243. Salata C, Calistri A, Parolin C, Baritussio A, Palù G. Antiviral 259. Remdesivir. Coronavirus Disease COVID-19. COVID-19 Treat-
activity of cationic amphiphilic drugs. Expert Rev Anti Infect ment Guidelines. 2020. https://www.covid19treatmentguide
Ther. 2017;15(5):483-492. lines.nih.gov/antiviral-therapy/remdesivir/. Accessed October
244. Wang M, Cao R, Zhang L, et al. Remdesivir and chloroquine 19, 2020.
effectively inhibit the recently emerged novel coronavirus 260. Cao B, Wang Y, Wen D, et al. A trial of lopinavir-ritonavir in
(2019-nCoV) in vitro. Cell Res. 2020;30(3):269-271. adults hospitalized with severe covid-19. N Engl J Med. 2020;
245. Paton NI, Lee L, Xu Y, et al. Chloroquine for influenza preven- 382(19):1787-1799.
tion: a randomised, double-blind, Placebo controlled trial. Lan- 261. Zhang L, Lin D, Sun X, et al. Crystal structure of SARS-CoV-2
cet Infect Dis. 2011;11(9):677-683. main protease provides a basis for design of improved a-ketoa-
246. Paton NI, Goodall RL, Dunn DT, et al. Effects of hydroxychlor- mide inhibitors. Science. 2020;368(6489):409-412. doi:10.1126/
oquine on immune activation and disease progression among science.abb3405
HIV-infected patients not receiving antiretroviral therapy. 262. Fisher CJ, Dhainaut JF, Opal SM, et al. Recombinant human
JAMA. 2012;308(4):353-361. interleukin 1 receptor antagonist in the treatment of patients with
247. Roques P, Thiberville SD, Dupuis-Maguiraga L, et al. Paradox-
sepsis syndrome. JAMA. 1994;271(23):1836-1843.
ical effect of chloroquine treatment in enhancing chikungunya
263. Steinberg KP. Efficacy and safety of corticosteroids for persis-
virus infection. Viruses. 2018;10(5):268.
tent acute respiratory distress syndrome. N Engl J Med. 2006;
248. Geleris J, Sun Y, Platt J, et al. Observational study of hydroxy-
354(16):1671-1684.
chloroquine in hospitalized patients with Covid-19. N Engl J
264. Bentzer P, Fjell C, Walley KR, Boyd J, Russell JA. Plasma
Med. 2020;382(25):2411-2418.
cytokine levels predict response to corticosteroids in septic
249. NIH Halts Clinical Trial of Hydroxychloroquine. National Insti-
shock. Intens Care Med. 2016;42(12):1970-1979.
tutes of Health (NIH). 2020 https://www.nih.gov/news-events/
265. Tirumalaraju D. Sanofi and regeneron’s Kevzara fails in phase
news-releases/nih-halts-clinical-trial-hydroxychloroquine.
III Covid-19 trial. Clinical Trials Arena. 2020. https://www.clin
Accessed October 19, 2020.
icaltrialsarena.com/news/kevzara-us-covid19-trial-data/.
250. Q&A: Hydroxychloroquine and COVID-19. 2020. https://www.
Accessed October 19, 2020.
who.int/news-room/q-a-detail/q-a-hydroxychloroquine-and-
266. Ware LB, Matthay MA. The acute respiratory distress syndrome.
covid-19. Accessed October 19, 2020.
New Eng J Med. 2000;342(18):1334-1349.
251. Cavalcanti AB, Zampieri FG, Rosa RG, et al. Hydroxychloro-
267. Hough CL. Steroids for acute respiratory distress syndrome?
quine with or without azithromycin in mild-to-moderate covid-
19 [published online July 23, 2020]. N Engl J Med. 2020. doi:10. Clin Chest Med. 2014;35(4):781-795.
1056/NEJMoa2019014 268. Rhen T, Cidlowski JA. Antiinflammatory action of glucocorti-
252. Maisonnasse P, Guedj J, Contreras V, et al. Hydroxychloroquine coids—new mechanisms for old drugs. N Eng J Med. 2005;
use against SARS-CoV-2 infection in non-human primates. 353(16):1711-1723.
Nature. 2020;585(7826):584-587. doi:10.1038/s41586-020- 269. Lewis SR, Pritchard MW, Thomas CM, Smith AF. Pharmaco-
2558-4 logical agents for adults with acute respiratory distress syn-
253. Bhimraj A, Morgan R, Shumaker AH, et al. COVID-19 guide- drome. Cochrane Database Syst Rev. 2019;7(7):CD004477.
line, Part 1: treatment and management. 2020. https://www.idso 270. Bone RC, Fisher CJ Jr, Clemmer TP, Slotman GJ, Metz CA.
ciety.org/practice-guideline/covid-19-guideline-treatment-and- Early methylprednisolone treatment for septic syndrome and the
management/. Accessed October 19, 2020. adult respiratory distress syndrome. Chest. 1987;92(6):
254. Center for Drug Evaluation & Research. FDA Cautions Use of 1032-1036.
Hydroxychloroquine/Chloroquine for COVID-19. U.S. Food 271. Luce JM, Montgomery AB, Marks JD, et al. Ineffectiveness of
Drug Administration. 2020. https://www.fda.gov/drugs/drug- high-dose methylprednisolone in preventing parenchymal lung
safety-and-availability/fda-cautions-against-use-hydroxychloro injury and improving mortality in patients with septic shock. Am
quine-or-chloroquine-covid-19-outside-hospital-setting-or Rev Respir Dis. 1988;138(1):62-68.
255. Ko WC, Rolain JM, Lee NY, et al. Arguments in favour of 272. Bernard GR, Luce JM, Sprung CL, et al. High-dose corticoster-
remdesivir for treating SARS-CoV-2 infections. Int J Antimi- oids in patients with the adult respiratory distress syndrome. New
crob Agents. 2020;55(4):105933. Engl J Med. 1987;317(25):1565-1570.
256. Sheahan T, Morrison TE, Funkhouser W, et al. MyD88 is 273. Weigelt JA, Norcross JF, Borman KR, Snyder WH. Early steroid
required for protection from lethal infection with a mouse- therapy for respiratory failure. Survey Anesthesiol. 1986;120(5):
adapted SARS-CoV. PLoS Pathog. 2008; 4(12):e1000240. 536-540.
Chivukula et al 41

274. Meduri GU, Golden E, Freire AX, et al. Methylprednisolone 281. WHO Rapid Evidence Appraisal for COVID-19 Therapies
infusion in early severe ARDS: results of a randomized con- (REACT) Working Group; Murthy S, Diaz JV. Association
trolled trial. Chest. 2007;131(4):954-963. between administration of systemic corticosteroids and mortal-
275. Villar J, Ferrando C, Martı́nez D, et al. Dexamethasone treat- ity among critically ill patients with COVID-19: a meta-analy-
ment for the acute respiratory distress syndrome: a multicentre, sis. JAMA. 2020;324(13):1-13. doi:10.1001/jama.2020.17023
randomised controlled trial. Lancet Respir Med. 2020;8(3): 282. Vandermeer ML, Thomas AR, Kamimoto L, et al. Association
267-276. between use of statins and mortality among patients hospitalized
276. Zhang Y, Sun W, Svendsen ER, et al. Do corticosteroids reduce with laboratory-confirmed influenza virus infections: a multi-
the mortality of influenza a (H1N1) infection? A meta-analysis. state study. J Infect Dis. 2012;205(1):13-19.
Crit Care. 2015;19(1):46. 283. Calfee CS, Delucchi KL, Sinha P, et al. Acute respiratory
277. Brun-Buisson C, Richard JC, Mercat A, Thiébaut AC, Brochard distress syndrome subphenotypes and differential response to
L. Early corticosteroids in severe influenza A/H1N1 pneumonia simvastatin: secondary analysis of a randomised controlled trial.
and acute respiratory distress syndrome. Am J Respir Crit Care Lancet Respir Med. 2018;6(9):691-698.
Med. 2011;183(9):1200-1206. 284. Trapnell BC, Whitsett JA. Gm-CSF regulates pulmonary surfac-
278. Moreno G, Rodrı́guez A, Reyes LF, et al. Corticosteroid treat- tant homeostasis and alveolar macrophage-mediated innate host
ment in critically ill patients with severe influenza pneumonia: a defense. Annu Rev Physiol. 2002;64(1):775-802.
propensity score matching study. Intens Care Med. 2018;44(9): 285. Huang C, Wang Y, Li X, et al. Clinical features of patients
1470-1482. infected with 2019 novel coronavirus in Wuhan, China. Lancet.
279. Lee N, Chan KA, Hui DS, et al. Effects of early corticosteroid 2020;395(10223):497-506.
treatment on plasma SARS-associated coronavirus RNA 286. Mathru M. A randomized trial of recombinant human
concentrations in adult patients. J Clin Virol. 2004;31(4): granulocyte-macrophage colony stimulating factor for patients
304-309. with acute lung injury. Reg Anesth Pain Med. 2012;2012:184-185.
280. Russell CD, Millar JE, Baillie JK. Clinical evidence does not 287. Huang FF, Barnes PF, Feng Y, et al. GM-CSF in the lung pro-
support corticosteroid treatment for 2019-nCoV lung injury. tects against lethal influenza infection. Am J Respir Crit Care
Lancet. 2020;395(10223):473-475. Med. 2011;184(2):259-268.

You might also like