ArticlePDF Available

Identification and Development of Therapeutics for COVID-19

American Society for Microbiology
mSystems®
Authors:

Abstract

The COVID-19 pandemic is a rapidly evolving crisis. With the worldwide scientific community shifting focus onto the SARS-CoV-2 virus and COVID-19, a large number of possible pharmaceutical approaches for treatment and prevention have been proposed.
Identication and Development of Therapeutics for COVID-19
Halie M. Rando,
a
,
b
,
c
Nils Wellhausen,
a
Soumita Ghosh,
d
Alexandra J. Lee,
a
Anna Ada Dattoli,
e
Fengling Hu,
f
James Brian Byrd,
g
Diane N. Rafizadeh,
h
,
i
Ronan Lordan,
j
Yanjun Qi,
k
Yuchen Sun,
k
Christian Brueffer,
l
Jeffrey M. Field,
e
Marouen Ben Guebila,
m
Nafisa M. Jadavji,
n
,
o
Ashwin N. Skelly,
h
,
p
Bharath Ramsundar,
q
Jinhui Wang,
r
Rishi Raj Goel,
p
YoSon Park,
a
COVID-19 Review Consortium, Simina M. Boca,
s
,
t
Anthony Gitter,
u
,
v
Casey S. Greene
b
,
c
,
e
,
w
a
Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
b
Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado, USA
c
Center for Health AI, University of Colorado School of Medicine, Aurora, Colorado, USA
d
Institute of Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
e
Department of Systems Pharmacology & Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
f
Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
g
University of Michigan School of Medicine, Ann Arbor, Michigan, USA
h
Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
i
Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania, USA
j
Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
k
Department of Computer Science, University of Virginia, Charlottesville, Virginia, USA
l
Department of Clinical Sciences, Lund University, Lund, Sweden
m
Department of Biostatistics, Harvard School of Public Health, Boston, Massachusetts, USA
n
Biomedical Science, Midwestern University, Glendale, Arizona, USA
o
Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
p
Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
q
The DeepChem Project
r
Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
s
Innovation Center for Biomedical Informatics, Georgetown University Medical Center, Washington, DC, USA
t
Early Biometrics & Statistical Innovation, Data Science & Articial Intelligence, R & D, AstraZeneca, Gaithersburg,
Maryland, USA
u
Department of Biostatistics and Medical Informatics, University of WisconsinMadison, Madison, Wisconsin, USA
v
Morgridge Institute for Research, Madison, Wisconsin, USA
w
Childhood Cancer Data Lab, Alexs Lemonade Stand Foundation, Philadelphia, Pennsylvania, USA
ABSTRACT After emerging in China in late 2019, the novel coronavirus severe acute
respiratory syndrome coronavirus 2 (SARS-CoV-2) spread worldwide, and as of mid-
2021, it remains a signicant threat globally. Only a few coronaviruses are known to
infect humans, and only two cause infections similar in severity to SARS-CoV-2: Severe
acute respiratory syndrome-related coronavirus, a species closely related to SARS-CoV-2
that emerged in 2002, and Middle East respiratory syndrome-related coronavirus,which
emerged in 2012. Unlike the current pandemic, previous epidemics were controlled
rapidly through public health measures, but the body of research investigating severe
acute respiratory syndrome and Middle East respiratory syndrome has proven valuable
for identifying approaches to treating and preventing novel coronavirus disease 2019
(COVID-19). Building on this research, the medical and scientic communities have
responded rapidly to the COVID-19 crisis and identied many candidate therapeutics.
The approaches used to identify candidates fall into four main categories: adaptation
of clinical approaches to diseases with related pathologies, adaptation based on viro-
logical properties, adaptation based on host response, and data-driven identication
(ID) of candidates based on physical properties or on pharmacological compendia. To
date, a small number of therapeutics have already been authorized by regulatory
agencies such as the Food and Drug Administration (FDA), while most remain under
Editor Jack A. Gilbert, University of California
San Diego
Copyright © 2021 Rando et al. This is an open-
access article distributed under the terms of
the Creative Commons Attribution 4.0
International license.
Address correspondence to Casey S. Greene,
casey.s.greene@cuanschutz.edu.
This represents one section of a larger evolving
review on SARS-CoV-2 and COVID-19, which is
regularly updated and available at https://
greenelab.github.io/covid19-review/.
This is a review paper that is authored by
scientists for an audience of scientists to
discuss research that is in progress. If you are
interested in guidelines on testing, therapies,
or other issues related to your health, you
should not use this document. Instead, you
should collect information from your local
health department, the CDCs guidance, or
your own government.
Published
November/December 2021 Volume 6 Issue 6 e00233-21 msystems.asm.org 1
REVIEW
2 November 2021
Downloaded from https://journals.asm.org/journal/msystems on 03 November 2021 by 2607:f470:34:2303:5040:4a48:5de1:cdb5.
investigation. The scale of the COVID-19 crisis offers a rare opportunity to collect data
on the effects of candidate therapeutics. This information provides insight not only
into the management of coronavirus diseases but also into the relative success of dif-
ferent approaches to identifying candidate therapeutics against an emerging disease.
IMPORTANCE The COVID-19 pandemic is a rapidly evolving crisis. With the worldwide sci-
entic community shifting focus onto the SARS-CoV-2 virus and COVID-19, a large num-
ber of possible pharmaceutical approaches for treatment and prevention have been pro-
posed. What was known about each of these potential interventions evolved rapidly
throughout 2020 and 2021. This fast-paced area of research provides important insight
into how the ongoing pandemic can be managed and also demonstrates the power of
interdisciplinary collaboration to rapidly understand a virus and match its characteristics
with existing or novel pharmaceuticals. As illustrated by the continued threat of viral epi-
demics during the current millennium, a rapid and strategic response to emerging viral
threats can save lives. In this review, we explore how different modes of identifying can-
didate therapeutics have borne out during COVID-19.
KEYWORDS COVID-19, review, therapeutics
The novel coronavirus Severe acute respiratory syndrome-related coronavirus 2 (SARS-
CoV-2) emerged in late 2019 and quickly precipitated the worldwide spread of
novel coronavirus disease 2019 (COVID-19). COVID-19 is associated with symptoms
ranging from mild or even asymptomatic to severe, and up to 2% of patients diag-
nosed with COVID-19 die from COVID-19-related complications such as acute respira-
tory disease syndrome (ARDS) (1). As a result, public health efforts have been critical to
mitigating the spread of the virus. However, as of mid-2021, COVID-19 remains a signif-
icant worldwide concern (Fig. 1), with the cases in some regions in 2021 surging far
above the numbers reported during the initial outbreak in early 2020. While a number
of vaccines have been developed and approved in different countries starting in late
2020 (2), vaccination efforts have not proceeded at the same pace throughout the
world and are not yet close to ending the pandemic.
Due to the continued threat of the virus and the severity of the disease, the identi-
cation and development of therapeutic interventions have emerged as signicant
international priorities. Prior developments during other recent outbreaks of emerging
diseases, especially those caused by human coronaviruses (HCoVs), have guided bio-
medical research into the behavior and treatment of this novel coronavirus infection.
However, previous emerging HCoV-related disease threats were controlled much more
quickly than SARS-CoV-2 through public health efforts (Fig. 1). The scale of the COVID-
19 pandemic has made the repurposing and development of pharmaceuticals more
urgent than in previous coronavirus epidemics.
LESSONS FROM PRIOR HCoV OUTBREAKS
At rst, SARS-CoV-2s rapid shift from an unknown virus to a signicant worldwide
threat closely paralleled the emergence of Severe acute respiratory syndrome-related co-
ronavirus 1 (SARS-CoV-1), which was responsible for the 200222003 SARS epidemic.
The rst documented case of COVID-19 was reported in Wuhan, China, in November
2019, and the disease quickly spread worldwide in the early months of 2020. In com-
parison, the rst case of SARS was reported in November 2002 in the Guangdong
Province of China, and it spread within China and then into several countries across
continents during the rst half of 2003 (3, 8, 9). In fact, genome sequencing quickly
revealed the virus causing COVID-19 to be a novel betacoronavirus closely related to
SARS-CoV-1 (10).
While similarities between these two viruses are unsurprising given their close phy-
logenetic relationship, there are also some differences in how the viruses affect
humans. SARS-CoV-1 infection is severe, with an estimated case fatality rate (CFR) for
SARS of 9.5% (8), while estimates of the CFR associated with COVID-19 are much lower,
Rando et al.
November/December 2021 Volume 6 Issue 6 e00233-21 msystems.asm.org 2
Downloaded from https://journals.asm.org/journal/msystems on 03 November 2021 by 2607:f470:34:2303:5040:4a48:5de1:cdb5.
at up to 2% (1). SARS-CoV-1 is highly contagious and spread primarily by droplet trans-
mission, with a basic reproduction number (R
0
) of 4 (i.e., each person infected was esti-
mated to infect four other people) (8). There is still some controversy whether SARS-
CoV-2 is primarily spread by droplets or is primarily airborne (1114). Most estimates of
its R
0
fall between 2.5 and 3 (1). Therefore, SARS is thought to be a deadlier and more
transmissible disease than COVID-19.
With the 17-year difference between these two outbreaks, there were major differ-
ences in the tools available to efforts to organize international responses. At the time
that SARS-CoV-1 emerged, no new HCoV had been identied in almost 40 years (9).
The identity of the virus underlying the SARS disease remained unknown until April of
2003, when the SARS-CoV-1 virus was characterized through a worldwide scientic
effort spearheaded by the World Health Organization (WHO) (9). In contrast, the SARS-
CoV-2 genomic sequence was released on 3 January 2020 (10), only days after the
international community became aware of the novel pneumonia-like illness now
known as COVID-19. While SARS-CoV-1 belonged to a distinct lineage from the two
other HCoVs known at the time of its discovery (8), SARS-CoV-2 is closely related to
SARS-CoV-1 and is a more distant relative of another HCoV characterized in 2012,
Middle East respiratory syndrome-related coronavirus (MERS-CoV) (15, 16). Signicant
efforts had been dedicated toward understanding SARS-CoV-1 and MERS-CoV and
how they interact with human hosts. Therefore, SARS-CoV-2 emerged under very dif-
ferent circumstances than SARS-CoV-1 in terms of scientic knowledge about HCoVs
and the tools available to characterize them.
Despite the apparent advantages for responding to SARS-CoV-2 infections, COVID-
19 has caused many orders of magnitude more deaths than SARS did (Fig. 1). The SARS
FIG 1 Cumulative global incidence of COVID-19 and SARS. As of 8 September 2021, 222,559,803 COVID-19 cases and 4,596,394 COVID-19 deaths had been
reported worldwide since 22 January 2020. A total of 8,432 cases and 813 deaths were reported for SARS from 17 March 2003 to 11 July 2003. SARS-CoV-1
was ofcially contained on 5 July 2003, within 9 months of its appearance (3). In contrast, SARS-CoV-2 remains a signicant global threat nearly 2 years
after its emergence. COVID-19 data are from the COVID-19 Data Repository by the Center for Systems Science and Engineering at Johns Hopkins University
(4, 5). SARS data are from the WHO (6) and were obtained from a data set on GitHub (7). See https://greenelab.github.io/covid19-review/ for the most
recent version of this gure, which is updated daily.
ID and Development of Therapeutics for COVID-19
November/December 2021 Volume 6 Issue 6 e00233-21 msystems.asm.org 3
Downloaded from https://journals.asm.org/journal/msystems on 03 November 2021 by 2607:f470:34:2303:5040:4a48:5de1:cdb5.
outbreak was ofcially determined to be under control in July 2003, with the success
credited to infection management practices such as mask wearing (9). MERS-CoV is still
circulating and remains a concern; although the fatality rate is very high at almost
35%, the disease is much less easily transmitted, as its R
0
has been estimated to be 1
(8). The low R
0
in combination with public health practices allowed for its spread to be
contained (8). Neither of these trajectories are comparable to that of SARS-CoV-2,
which remains a serious threat worldwide over a year and a half after the rst cases of
COVID-19 emerged (Fig. 1).
POTENTIAL APPROACHES TO THE TREATMENT OF COVID-19
Therapeutic interventions can utilize two approaches: they can mitigate the effects
of an infection that harms an infected person, or they can hinder the spread of infec-
tion within a host by disrupting the viral life cycle. The goal of the former strategy is to
reduce the severity and risks of an active infection, while for the latter, it is to inhibit
the replication of a virus once an individual is infected, potentially freezing disease pro-
gression. Additionally, two major approaches can be used to identify interventions
that might be relevant to managing an emerging disease or a novel virus: drug repur-
posing and drug development. Drug repurposing involves identifying an existing com-
pound that may provide benets in the context of interest (17). This strategy can focus
on either approved or investigational drugs, for which there may be applicable preclin-
ical or safety information (17). Drug development, on the other hand, provides an op-
portunity to identify or develop a compound specically relevant to a particular need,
but it is often a lengthy and expensive process characterized by repeated failure (18).
Drug repurposing therefore tends to be emphasized in a situation like the COVID-19
pandemic due to the potential for a more rapid response.
Even from the early months of the pandemic, studies began releasing results from
analyses of approved and investigational drugs in the context of COVID-19. The rapid
timescale of this response meant that, initially, most evidence came from observational
studies, which compare groups of patients who did and did not receive a treatment to
determine whether it may have had an effect. This type of study can be conducted rap-
idly but is subject to confounding. In contrast, randomized controlled trials (RCTs) are
the gold standard method for assessing the effects of an intervention. Here, patients
are prospectively and randomly assigned to treatment or control conditions, allowing
for much stronger interpretations to be drawn; however, data from these trials take
much longer to collect. Both approaches have proven to be important sources of infor-
mation in the development of a rapid response to the COVID-19 crisis, but as the pan-
demic draws on and more results become available from RCTs, more denitive answers
are becoming available about proposed therapeutics. Interventional clinical trials are
currently investigating or have investigated a large number of possible therapeutics
and combinations of therapeutics for the treatment of COVID-19 (Fig. 2).
The purpose of this review is to provide an evolving resource tracking the status of
efforts to repurpose and develop drugs for the treatment of COVID-19. We highlight
four strategies that provide different paradigms for the identication of potential phar-
maceutical treatments. The WHO guidelines (20) and a systematic review (21) are com-
plementary living documents that summarize COVID-19 therapeutics.
REPURPOSING DRUGS FOR SYMPTOM MANAGEMENT
A variety of symptom proles with a range of severity are associated with COVID-19
(1). In many cases, COVID-19 is not life-threatening. A study of COVID-19 patients in a
hospital in Berlin, Germany, reported that the highest risk of death was associated with
infection-related symptoms, such as sepsis, respiratory symptoms such as ARDS, and
cardiovascular failure or pulmonary embolism (22). Similarly, an analysis in Wuhan,
China, reported that respiratory failure (associated with ARDS) and sepsis/multiorgan
failure accounted for 69.5% and 28.0% of deaths, respectively, among 82 deceased
patients (23). COVID-19 is characterized by two phases. The rst is the acute response,
Rando et al.
November/December 2021 Volume 6 Issue 6 e00233-21 msystems.asm.org 4
Downloaded from https://journals.asm.org/journal/msystems on 03 November 2021 by 2607:f470:34:2303:5040:4a48:5de1:cdb5.
where an adaptive immune response to the virus is established and in many cases can
mitigate viral damage to organs (24). The second phase characterizes more severe
cases of COVID-19. Here, patients experience a cytokine storm, whereby excessive pro-
duction of cytokines oods into circulation, leading to systemic inammation, immune
dysregulation, and multiorgan dysfunction that can cause multiorgan failure and death
if untreated (25). ARDS-associated respiratory failure can occur during this phase.
Cytokine dysregulation was also identied in patients with SARS (26, 27).
In the early days of the COVID-19 pandemic, physicians sought to identify potential
treatments that could benet patients, and in some cases shared their experiences and
advice with the medical community on social media sites such as Twitter (28). These on-
the-ground treatment strategies could later be analyzed retrospectively in observational
studies or investigated in an interventional paradigm through RCTs. Several notable cases
involved the use of small-molecule drugs, which are synthesized compounds of low molec-
ular weight, typically less than 1 kDa (29). Small-molecule pharmaceutical agents have been
a backbone of drug development since the discovery of penicillin in the early twentieth
century (30). It and other antibiotics have long been among the best-known applications of
small molecules to therapeutics, but biotechnological developments such as the prediction
of protein-protein interactions (PPIs) have facilitated advances in precise targeting of spe-
cic structures using small molecules (30). Small-molecule drugs today encompass a wide
range of therapeutics beyond antibiotics, including antivirals, protein inhibitors, and many
broad-spectrum pharmaceuticals.
FIG 2 COVID-19 clinical trials. Trial data are from the University of Oxford Evidence-Based Medicine Data Labs COVID-19 TrialsTracker (19). As of 31
December 2020, there were 6,987 COVID-19 clinical trials of which 3,962 were interventional. The study types include only types used in at least ve
trials. Only interventional trials are analyzed in the gures depicting status, phase, and intervention. Of the interventional trials, 98 trials had reported
results as of 31 December 31 2020. Recruitment status and trial phase are shown only for interventional trials in which the status or phase is recorded.
Common interventions refers to interventions used in at least 10 trials. Combinations of interventions, such as hydroxychloroquine with azithromycin,
are tallied separately from the individual interventions. See https://greenelab.github.io/covid19-review/ for the most recent version of this gure, which
is updated daily.
ID and Development of Therapeutics for COVID-19
November/December 2021 Volume 6 Issue 6 e00233-21 msystems.asm.org 5
Downloaded from https://journals.asm.org/journal/msystems on 03 November 2021 by 2607:f470:34:2303:5040:4a48:5de1:cdb5.
Many treatments considered for COVID-19 have relied on a broad-spectrum
approach. These treatments do not specically target a virus or particular host receptor
but rather induce broad shifts in host biology that are hypothesized to be potential
inhibitors of the virus. This approach relies on the fact that when a virus enters a host,
the host becomes the viruss environment. Therefore, the state of the host can also
inuence the viruss ability to replicate and spread. The administration and assessment
of broad-spectrum small-molecule drugs on a rapid time course were feasible because
they are often available in hospitals, or in some cases, they may also be prescribed to a
large number of outpatients. One of the other advantages is that these well-estab-
lished compounds, if found to be benecial, are often widely available, in contrast to
boutique experimental drugs.
In some cases, prior data were available from experiments examining the response
of other HCoVs or HCoV infections to a candidate drug. In addition to nonpharmaceuti-
cal interventions such as encouraging nonintubated patients to adopt a prone position
(31), knowledge about interactions between HCoVs and the human body, much of
which emerged from SARS and MERS research over the past 2 decades, led to the sug-
gestion that a number of common drugs might benet COVID-19 patients. However,
the short duration and low case numbers of prior outbreaks were less well suited to
the large-scale study of clinical applications than the COVID-19 pandemic is. As a result,
COVID-19 has presented the rst opportunity to robustly evaluate treatments that
were common during prior HCoV outbreaks to determine their clinical efcacy. The
rst year of the COVID-19 pandemic demonstrated that there are several different tra-
jectories that these clinically suggested, widely available candidates can follow when
assessed against a widespread, novel viral threat.
One approach to identifying candidate small-molecule drugs was to look at the
approaches used to treat SARS and MERS. Treatment of SARS and MERS patients priori-
tized supportive care and symptom management (8). Among the clinical treatments
for SARS and MERS that were explored, there was generally a lack of evidence indicat-
ing whether they were effective. Most of the supportive treatments for SARS were
found inconclusive in meta-analysis (32), and a 2004 review reported that not enough
evidence was available to make conclusions about most treatments (33). However, one
strategy adopted from prior HCoV outbreaks is currently the best-known treatment for
severe cases of COVID-19. Corticosteroids are broad-spectrum treatments and are a
well-known, widely available treatment for pneumonia (3439) that have also been
debated as a possible treatment for ARDS (4045). Corticosteroids were also used and
subsequently evaluated as possible supportive care for SARS and MERS. In general,
studies and meta-analyses did not identify support for corticosteroids to prevent mor-
tality in these HCoV infections (4648); however, one study found that the effects
might be masked by variability in treatment protocols, such as dosage and timing (33).
While the corticosteroids most often used to treat SARS were methylprednisolone and
hydrocortisone, availability issues for these drugs at the time led to dexamethasone
also being used in North America (49).
Dexamethasone (9
a
-uoro-16
a
-methylprednisolone) is a synthetic corticosteroid that
binds to glucocorticoid receptors (50, 51). It functions as an anti-inammatory agent by
binding to glucocorticoid receptors with higher afnity than endogenous cortisol (52).
Dexamethasone and other steroids are widely available and affordable, and they are often
used to treat community-acquired pneumonia (53) as well as chronic inammatory condi-
tions such as asthma, allergies, and rheumatoid arthritis (5456). Immunosuppressive
drugs such as steroids are typically contraindicated in the setting of infection (57), but
because COVID-19 results in hyperinammation that appears to contribute to mortality via
lung damage, immunosuppression may be a helpful approach to treatment (58). A clinical
trial that began in 2012 recently reported that dexamethasone may improve outcomes for
patients with ARDS (40), but a meta-analysis of a small amount of available data about
dexamethasone as a treatment for SARS suggested that it may, in fact, be associated with
patient harm (59). However, the ndings for SARS may have been biased by the fact that
Rando et al.
November/December 2021 Volume 6 Issue 6 e00233-21 msystems.asm.org 6
Downloaded from https://journals.asm.org/journal/msystems on 03 November 2021 by 2607:f470:34:2303:5040:4a48:5de1:cdb5.
allofthestudiesexaminedwereobservationalandalargenumberofinconclusivestudies
were not included (60). The questions of whether and when to counter hyperinammation
with immunosuppression in the setting of COVID-19 (as in SARS [27]) was an area of
intense debate, as the risks of inhibiting antiviral immunity needed to be weighed against
the benecial anti-inammatory effects (61). As a result, guidelines early in the pandemic
typically recommended avoiding treating COVID-19 patients with corticosteroids such as
dexamethasone (59).
Despite this initial concern, dexamethasone was evaluated as a potential treatment for
COVID-19 (Appendix). Dexamethasone treatment comprised one arm of the multisite
Randomized Evaluation of COVID-19 Therapy (RECOVERY) trial in the United Kingdom (62).
This study found that the 28-day mortality rate was lower in patients receiving dexametha-
sone than in those receiving the standard of care (SOC). However, this nding was driven
by differences in mortality among patients who were receiving mechanical ventilation or
supplementary oxygen at the start of the study. The report indicated that dexamethasone
reduced 28-day mortality relative to the SOC in patients who were ventilated (29.3% versus
41.4%) and among those who were receiving oxygen supplementation (23.3% versus
26.2%) at randomization, but not in patients who were breathing independently (17.8%
versus 14.0%). These ndings also suggested that dexamethasone may have reduced pro-
gression to mechanical ventilation, especially among patients who were receiving oxygen
support at randomization. Other analyses have supported the importance of disease course
in determining the efcacy of dexamethasone: additional results suggest greater potential
for patients who have experienced symptoms for at least 7 days and patients who were
not breathing independently (63). A meta-analysis that evaluated the results of the
RECOVERY trial alongside trials of other corticosteroids, such as hydrocortisone, similarly
concluded that corticosteroids may be benecial to patients with severe COVID-19 who are
receiving oxygen supplementation (64). Thus, it seems likely that dexamethasone is useful
for treating inammation associated with immunopathy or cytokine release syndrome
(CRS), which is a condition caused by detrimental overactivation of the immune system (1).
In fact, corticosteroids such as dexamethasone are sometimes used to treat CRS (65).
Guidelines were quickly updated to encourage the use of dexamethasone in severe cases
(66), and this affordable and widely available treatment rapidly became a valuable tool
against COVID-19 (67), with demand surging within days of the preprints release (68).
APPROACHES TARGETING THE VIRUS
Therapeutics that directly target the virus itself hold the potential to prevent people
infected with SARS-CoV-2 from developing potentially damaging symptoms (Fig. 3).
Such drugs typically fall into the broad category of antivirals. Antiviral therapies hinder
the spread of a virus within the host, rather than destroying existing copies of the virus,
and these drugs can vary in their specicity to a narrow or broad range of viral targets.
This process requires inhibiting the replication cycle of a virus by disrupting one of six
fundamental steps (69). In the rst of these steps, the virus attaches to and enters the
host cell through endocytosis. Then the virus undergoes uncoating, which is classically
dened as the release of viral contents into the host cell. Next, the viral genetic mate-
rial enters the nucleus where it is replicated during the biosynthesis stage. During
the assembly stage, viral proteins are translated, allowing new viral particles to be
assembled. In the nal step, new viruses are released into the extracellular environ-
ment. Although antivirals are designed to target a virus, they can also impact other
processes in the host and may have unintended effects. Therefore, these therapeutics
must be evaluated for both efcacy and safety. As the technology to respond to
emerging viral threats has also evolved over the past 2 decades, a number of candidate
treatments have been identied for prior viruses that may be relevant to the treatment
of COVID-19.
Many antiviral drugs are designed to inhibit the replication of viral genetic material
during the biosynthesis step. Unlike DNA viruses, which can use the host enzymes to
propagate themselves, RNA viruses like SARS-CoV-2 depend on their own polymerase,
ID and Development of Therapeutics for COVID-19
November/December 2021 Volume 6 Issue 6 e00233-21 msystems.asm.org 7
Downloaded from https://journals.asm.org/journal/msystems on 03 November 2021 by 2607:f470:34:2303:5040:4a48:5de1:cdb5.
the RNA-dependent RNA polymerase (RdRP), for replication (71, 72). RdRP is therefore a
potential target for antivirals against RNA viruses. Disruption of RdRP is the proposed
mechanism underlying the treatment of SARS and MERS with ribavirin (73). Ribavirin is
an antiviral drug effective against other viral infections that was often used in combina-
tion with corticosteroids and sometimes interferon (IFN) medications to treat SARS and
MERS (9). However, analyses of its effects in retrospective and in vitro analyses of SARS
and the SARS-CoV-1 virus, respectively, have been inconclusive (9). While IFNs and riba-
virin have shown promise in in vitro analyses of MERS, their clinical effectiveness remains
unknown (9). The current COVID-19 pandemic has provided an opportunity to assess the
clinical effects of these treatments. As one example, ribivarin was also used in the early
days of COVID-19, but a retrospective cohort study comparing patients who did and did
not receive ribivarin revealed no effect on the mortality rate (74).
Since nucleotides and nucleosides are the natural building blocks for RNA synthesis,
an alternative approach has been to explore nucleoside and nucleotide analogs for
their potential to inhibit viral replication. Analogs containing modications to nucleo-
tides or nucleosides can disrupt key processes, including replication (75). A single
incorporation does not inuence RNA transcription; however, multiple events of incor-
poration lead to the arrest of RNA synthesis (76). One candidate antiviral considered
for the treatment of COVID-19 is favipiravir (Avigan), also known as T-705, which was
discovered by Toyama Chemical Co., Ltd. (77). It was previously found to be effective
at blocking viral amplication in several inuenza virus subtypes as well as other RNA
FIG 3 Mechanisms of action for potential therapeutics. Potential therapeutics currently being studied can target the SARS-CoV-2 virus or modify the host
environment through many different mechanisms. Here, the relationships between the virus, host cells, and several therapeutics are visualized. Drug names
are color coded according to the grade assigned to them by the Center for Cytokine Storm Treatment & Laboratorys CORONA Project (70) (green for
grade A, lime for grade B, orange for grade C, and red for grade D).
Rando et al.
November/December 2021 Volume 6 Issue 6 e00233-21 msystems.asm.org 8
Downloaded from https://journals.asm.org/journal/msystems on 03 November 2021 by 2607:f470:34:2303:5040:4a48:5de1:cdb5.
viruses, such as Flaviviridae and Picornaviridae, through a reduction in plaque forma-
tion (78) and viral replication in Madin-Darby canine kidney cells (79). Favipiravir (6-u-
oro-3-hydroxy-2-pyrazinecarboxamide) acts as a purine and purine nucleoside analog
that inhibits viral RNA polymerase in a dose-dependent manner across a range of RNA
viruses, including inuenza viruses (8084). Biochemical experiments showed that favi-
piravir was recognized as a purine nucleoside analog and incorporated into the viral
RNA template. In 2014, the drug was approved in Japan for the treatment of inuenza
that was resistant to conventional treatments like neuraminidase inhibitors (85).
Though initial analyses of favipiravir in observational studies of its effects on COVID-19
patients were promising, recent results of two small RCTs suggest that it is unlikely to
affect COVID-19 outcomes (Appendix).
In contrast, another nucleoside analog, remdesivir, is one of the few treatments
against COVID-19 that has received FDA approval. Remdesivir (GS-5734) is an intrave-
nous antiviral that was proposed by Gilead Sciences as a possible treatment for Ebola
virus disease. It is metabolized to GS-441524, an adenosine analog that inhibits a broad
range of polymerases and then evades exonuclease repair, causing chain termination
(8688). Gilead received an emergency use authorization (EUA) for remdesivir from the
FDA early in the pandemic (May 2020) and was later found to reduce mortality and re-
covery time in a double-blind, placebo-controlled, phase 3 clinical trial performed at
60 trial sites, 45 of which were in the United States (8992). Subsequently, the WHO
Solidarity trial, a large-scale, open-label trial enrolling 11,330 adult inpatients at 405
hospitals in 30 countries around the world, reported no effect of remdesivir on in-hos-
pital mortality, duration of hospitalization, or progression to mechanical ventilation
(93). Therefore, additional clinical trials of remdesivir in different patient pools and in
combination with other therapies may be needed to rene its use in the clinic and
determine the forces driving these differing results. Remdesivir offers proof of principle
that SARS-CoV-2 can be targeted at the level of viral replication, since remdesivir tar-
gets the viral RNA polymerase at high potency. Identication of such candidates
depends on knowledge about the virological properties of a novel threat. However,
the success and relative lack of success, respectively, of remdesivir and favipiravir
underscore the fact that drugs with similar mechanisms will not always produce similar
results in clinical trials.
DISRUPTING HOST-VIRUS INTERACTIONS
Interrupting viral colonization of cells. Some of the most widely publicized exam-
ples of efforts to repurpose drugs for COVID-19 are broad-spectrum, small-molecule
drugs where the mechanism of action made it seem that the drug might disrupt inter-
actions between SARS-CoV-2 and human host cells (Fig. 3). However, the exact out-
comes of such treatments are difcult to predict a priori, and there are several exam-
ples where early enthusiasm was not borne out in subsequent trials. One of the most
famous examples of an analysis of whether a well-known medication could provide
benets to COVID-19 patients came from the assessment of chloroquine (CQ) and
hydroxychloroquine (HCQ), which are used for the treatment and prophylaxis of
malaria as well as the treatment of lupus erythematosus and rheumatoid arthritis in
adults (94). These drugs are lysosomotropic agents, meaning they are weak bases that
can pass through the plasma membrane. It was thought that they might provide bene-
ts against SARS-CoV-2 by interfering with the digestion of antigens within the lyso-
some and inhibiting CD4 T-cell stimulation while promoting the stimulation of CD8 T
cells (95). These compounds also have anti-inammatory properties (95) and can
decrease the production of certain key cytokines involved in the immune response,
including interleukin-6 (IL-6) and inhibit the stimulation of Toll-like receptors (TLRs)
and TLR signaling (95).
In vitro analyses reported that CQ inhibited cell entry of SARS-CoV-1 (96) and that
both CQ and HCQ inhibited viral replication within cultured cells (97), leading to early
hope that it might provide similar therapeutic or protective effects in patients.
ID and Development of Therapeutics for COVID-19
November/December 2021 Volume 6 Issue 6 e00233-21 msystems.asm.org 9
Downloaded from https://journals.asm.org/journal/msystems on 03 November 2021 by 2607:f470:34:2303:5040:4a48:5de1:cdb5.
However, while the rst publication on the clinical application of these compounds to
the inpatient treatment of COVID-19 was very positive (98), it was quickly discredited
(99). Over the following months, extensive evidence emerged demonstrating that CQ
and HCQ offered no benets for COVID-19 patients and, in fact, carried the risk of dan-
gerous side effects (Appendix). The nail in the cofn came when ndings from the
large-scale RECOVERY trial were released on 8 October 2020. This study enrolled
11,197 hospitalized patients whose physicians believed it would not harm them to par-
ticipate and used a randomized, open-label design to study the effects of HCQ com-
pared to the standard of care (SOC) at 176 hospitals in the United Kingdom (100).
Rates of COVID-19-related mortality did not differ between the control and HCQ arms,
but patients receiving HCQ were slightly more likely to die due to cardiac events.
Patients who received HCQ also had a longer duration of hospitalization than patients
receiving usual care and were more likely to progress to mechanical ventilation or
death (as a combined outcome). As a result, enrollment in the HCQ arm of the
RECOVERY trial was terminated early (101). The story of CQ/HCQ therefore illustrates
how initial promising in vitro analyses can fail to translate to clinical usefulness.
A similar story has arisen with the broad-spectrum, small-molecule anthelmintic
ivermectin, which is a synthetic analog of avermectin, a bioactive compound produced
by a microorganism known as Streptomyces avermectinius and Streptomyces avermitilis
(102, 103). Avermectin disrupts the ability of parasites to avoid the host immune
response by blocking glutamate-gated chloride ion channels in the peripheral nervous
system from closing, leading to hyperpolarization of neuronal membranes, disruption
of neural transmission, and paralysis (102, 104, 105). Ivermectin has been used since
the early 1980s to treat endo- and ectoparasitic infections by helminths, insects, and
arachnids in veterinary contexts (102, 106) and since the late 1980s to treat human par-
asitic infections as well (102, 104). More recent research has indicated that ivermectin
might function as a broad-spectrum antiviral by disrupting the trafcking of viral pro-
teins by both RNA and DNA viruses (105, 107, 108), although most of these studies
have demonstrated this effect in vitro (108). The potential for antiviral effects on SARS-
CoV-2 were investigated in vitro, and ivermectin was found to inhibit viral replication
in a cell line derived from Vero cells (Vero-hSLAM) (109). However, inhibition of viral
replication was achieved at concentrations that were much higher than that explored
by existing dosage guidelines (110, 111), which are likely to be associated with signi-
cant side effects due to the increased potential that the compound could cross the
mammalian blood-brain barrier (112, 113).
Retrospective studies and small RCTs began investigating the effects of standard
doses of this low-cost, widely available drug. One retrospective study reported that
ivermectin reduced all-cause mortality (114), while another reported no difference in
clinical outcomes or viral clearance (115). Small RCTs enrolling less than 50 patients
per arm have also reported a wide array of positive (116120) and negative results
(121, 122). A slightly larger RCT enrolling 115 patients in two arms reported inconclu-
sive results (123). Hope for the potential of ivermectin peaked with the release of a
preprint reporting results of a multicenter, double-blind RCT where a 4-day course of
ivermectin was associated with clinical improvement and earlier viral clearance in
400 symptomatic patients and 200 close contacts (124); however, concerns were
raised about both the integrity of the data and the paper itself (125, 126), and this
study was removed by the preprint server Research Square (127). A similarly sized
RCT suggested no effect on the duration of symptoms among 400 patients split
evenly across the intervention and control arms (128), and although meta-analyses
have reported both null (129, 130) and benecial (131138) effects of ivermectin on
COVID-19 outcomes, the certainty is likely to be low (132). These ndings are poten-
tially biased by a small number of low-quality studies, including the preprint that has
been taken down (139), and the authors of one (140) have issued a notice (131) that
they will revise their study with the withdrawn study removed. Thus, much like HCQ/
CQ, enthusiasm for research that either has not or should not have passed peer
Rando et al.
November/December 2021 Volume 6 Issue 6 e00233-21 msystems.asm.org 10
Downloaded from https://journals.asm.org/journal/msystems on 03 November 2021 by 2607:f470:34:2303:5040:4a48:5de1:cdb5.
review has led to large numbers of patients worldwide receiving treatments that
might not have any effect or could even be harmful. Additionally, comments on the
now-removed preprint include inquiries into how best to self-administer veterinary
ivermectin as a prophylactic (127), and the FDA has posted information explaining
why veterinary ivermectin should not be taken by humans concerned about COVID-
19 (141). Ivermectin is now one of several candidate therapeutics being investigated
in the large-scale TOGETHER (142) and PRINCIPLE (143) clinical trials. The TOGETHER
trial, which previously demonstrated no effect of HCQ and lopinavir-ritonavir (144),
released preliminary results in early August 2021 suggesting that ivermectin also has
no effect on COVID-19 outcomes (145).
While CQ/HCQ and ivermectin are well-known medications that have long been
prescribed in certain contexts, investigation of another well-established type of phar-
maceutical was facilitated by the fact that it was already being taken by a large number
of COVID-19 patients. Angiotensin-converting enzyme inhibitors (ACEIs) and angioten-
sin II receptor blockers (ARBs) are among todays most commonly prescribed medica-
tions, often being used to control blood pressure (146, 147). In the United States, for
example, they are prescribed well over 100,000,000 times annually (148). Prior to the
COVID-19 pandemic, the relationship between ACE2, ACEIs, and SARS had been con-
sidered as possible evidence that ACE2 could serve as a therapeutic target (149), and
the connection had been explored through in vitro and molecular docking analysis
(150) but ultimately was not pursued clinically (151). Data from some animal models
suggest that several, but not all, ACEIs and several ARBs increase ACE2 expression in
the cells of some organs (152), but clinical studies have not established whether
plasma ACE2 expression is increased in humans treated with these medications (153).
In this case, rather than introducing ARBs/ACEIs, a number of analyses have investi-
gated whether discontinuing use affects COVID-19 outcomes. An initial observational
study of the association of exposure to ACEIs or ARBs with outcomes in COVID-19 (154)
was retracted from the New England Journal of Medicine due to concerns related to
data availability (155). As RCTs have become available, they have demonstrated no
effect of continuing versus discontinuing ARBs/ACEIs on patient outcomes (156, 157)
(Appendix). Thus, once again, despite a potential mechanistic association with the pa-
thology of SARS-CoV-2 infection, these medications were not found to inuence the
trajectory of COVID-19 illness.
For medications that are widely known and common, clinical research into their ef-
cacy against a novel threat can be developed very quickly. This feasibility can present a
double-edged sword. For example, HCQ and CQ were incorporated into the SOC in
many countries early in the pandemic and had to be discontinued once their potential
to harm COVID-19 patients became apparent (158, 159). Dexamethasone remains the
major success story from this category of repurposed drugs and is likely to have saved
a large number of lives since summer 2020 (67).
Manipulating the host immune response. Treatments based on understanding a
virus and/or how a virus interacts with the human immune system can fall into two
categories: they can interact with the innate immune response, which is likely to be a
similar response across viruses, or they can be specically designed to imitate the
adaptive immune response to a particular virus. In the latter case, conservation of
structure or behavior across viruses determines interest in whether drugs developed
for one virus can treat another. During the COVID-19 pandemic, a number of candidate
therapeutics have been explored in these categories, with varied success.
Knowledge gained from characterizing SARS-CoV-1 and MERS-CoV from a funda-
mental biological perspective along with their interactions with the human immune
system provides a theoretical basis for identifying candidate therapies. Biologics are a
particularly important class of drugs for efforts to address HCoV through this paradigm.
They are produced from components of living organisms or viruses, historically primar-
ily from animal tissues (160). Biologics have become increasingly feasible to produce as
recombinant DNA technologies have advanced (160).
ID and Development of Therapeutics for COVID-19
November/December 2021 Volume 6 Issue 6 e00233-21 msystems.asm.org 11
Downloaded from https://journals.asm.org/journal/msystems on 03 November 2021 by 2607:f470:34:2303:5040:4a48:5de1:cdb5.
There are many differences on the development side between biologics and syn-
thesized pharmaceuticals, such as small-molecule drugs. Typically, biologics are orders
of magnitude larger than small-molecule drugs and are catabolized by the body to
their amino acid components (161). They are often heat sensitive, and their toxicity can
vary, as it is not directly associated with the primary effects of the drug; in general,
their physiochemical properties are much less understood compared to small mole-
cules (161). Biologics include signicant medical breakthroughs such as insulin for the
management of diabetes and vaccines and monoclonal antibodies (MAbs) and interfer-
ons (IFNs), which can be used to target the host immune response after infection.
MAbs have revolutionized the way we treat human diseases and have become some
of the best-selling drugs in the pharmaceutical market in recent years (162). There are
currently 79 FDA approved MAbs on the market, including antibodies for viral infections
(e.g., ibalizumab for human immunodeciency virus and palivizumab for respiratory syn-
cytial virus) (162, 163). Virus-specic neutralizing antibodies commonly target viral sur-
face glycoproteins or host structures, thereby inhibiting viral entry through receptor
binding interference (164, 165). This interference is predicted to reduce the viral load,
mitigate disease, and reduce overall hospitalization. MAbs can be designed for a particu-
lar virus, and signicant advances have been made in the speed at which new MAbs can
be identied and produced. At the time of the SARS and MERS epidemics, interest in
MAbs to reduce infection was never realized (166, 167), but this allowed for MAbs to
quickly be considered among the top candidates against COVID-19.
(i) Biologics and the innate immune response. Deaths from COVID-19 often occur
when inammation becomes dysregulated following an immune response to the SARS-
CoV-2 virus. Therefore, one potential approach to reducing COVID-19 mortality rates is to
manage the inammatory response in severely ill patients. One candidate therapeutic
identied that uses this mechanism is tocilizumab (TCZ). TCZ is a MAb that was developed
to manage chronic inammation caused by the continuous synthesis of the cytokine IL-6
(168). IL-6 is a proinammatory cytokine belonging to the interleukin family, which is com-
prised by immune system regulators that are primarily responsible for immune cell differ-
entiation. Often used to treat chronic inammatory conditions such as rheumatoid arthritis
(168), TCZ has become a pharmaceutical of interest for the treatment of COVID-19 because
of the role IL-6 plays in this disease. It has also been approved to treat CRS caused by chi-
meric antigen receptor T-cell therapy (CAR-T) treatments (169). While the secretion of IL-6
can be associated with chronic conditions, IL-6 is a key player in the innate immune
response and is secreted by macrophages in response to the detection of pathogen-asso-
ciated molecular patterns and damage-associated molecular patterns (168). An analysis of
191 inpatients at two Wuhan hospitals revealed that blood concentrations of IL-6 differed
between patients who did and did not recover from COVID-19. Patients who ultimately
died had higher IL-6 levels at admission than those who recovered (170). Additionally, IL-6
levels remained higher throughout the course of hospitalization in the patients who ulti-
mately died (170).
Currently, TCZ is being administered either as a monotherapy or in combination
with other treatments in 73 interventional COVID-19 clinical trials (Fig. 2). A number of
retrospective studies have been conducted in several countries (171176). In general,
these studies have reported a positive effect of TCZ on reducing mortality in COVID-19
patients, although due to their retrospective designs, signicant limitations are present
in all of them (Appendix). It was not until 11 February 2021 that a preprint describing
preliminary results of the rst RCT of TCZ was released as part of the RECOVERY trial
(177). TCZ was found to reduce 28-day mortality from 33% in patients receiving the
SOC alone to 29% in those receiving TCZ. Combined analysis of the RECOVERY trial
data with data from smaller RCTs suggested a 13% reduction in 28-day mortality (177).
While this initial report did not include the full results expected from the RECOVERY
trial, this large-scale RCT provides strong evidence that TCZ may offer benets for
COVID-19 patients. The RECOVERY trial along with results from several other RCTs
(178182) were cited as support for the EUA issued for TCZ in June 2021 (183).
Rando et al.
November/December 2021 Volume 6 Issue 6 e00233-21 msystems.asm.org 12
Downloaded from https://journals.asm.org/journal/msystems on 03 November 2021 by 2607:f470:34:2303:5040:4a48:5de1:cdb5.
However, the fact that TCZ suppresses the immune response means that it does carry
risks for patients, especially a potential risk of secondary infection (Appendix).
TCZ is just one example of a candidate drug targeting the host immune response
and specically excessive inammation. For example, interferons (IFNs) have also been
investigated; these are a family of cytokines critical to activating the innate immune
response against viral infections. Synairgen has been investigating a candidate drug,
SNG001, which is an IFN-
b
-1a formulation to be delivered to the lungs via inhalation
(184) that they reported reduced progression to ventilation in a double-blind, placebo-
controlled, multicenter study of 101 patients with an average age in the late 50s (185,
186). However, these ndings were not supported by the large-scale WHO Solidarity
trial, which reported no signicant effect of IFN-
b
-1a on patient survival during hospi-
talization (93), although differences in the designs of the two studies, and specically
the severity of illness among enrolled patients, may have inuenced their divergent
outcomes (Appendix). Other biologics inuencing inammation are also being
explored (Appendix). It is also important that studies focused on inammation as a
possible therapeutic target consider the potential differences in baseline inammation
among patients from different backgrounds, which may be caused by differing life
experiences (see reference 187).
(ii) Biologics and the adaptive immune response. While TCZ is an example of an
MAb focused on managing the innate immune response, other treatments are more
specic, targeting the adaptive immune response after an infection. In some cases,
treatments can utilize biologics obtained directly from recovered individuals. From
the very early days of the COVID-19 pandemic, polyclonal antibodies from convales-
cent plasma were investigated as a potential treatment for COVID-19 (188, 189).
Convalescent plasma was used in prior epidemics, including SARS, Ebola virus disease,
and even the 1918 Spanish inuenza (188, 190). Use of convalescent plasma transfu-
sion (CPT) over more than a century has aimed to reduce symptoms and improve
mortality in infected people (190), possibly by accelerating viral clearance (188).
However, it seems unlikely that this classic treatment confers any benet for COVID-
19 patients. Several systematic reviews have investigated whether CPT reduced mor-
tality in COVID-19 patients, and although ndings from early in the pandemic (up to
19 April 2020) did support the use of CPT (190), the tide has shifted as the body of
available literature has grown (191). While titer levels were suggested as a possible
determining factor in the success of CPT against COVID-19 (192), the large-scale
RECOVERY trial evaluated the effect of administering high-titer plasma specically and
found no effect on mortality or hospital discharge over a 28-day period (193). These
results thus suggest that, despite initial optimism and an EUA from the FDA, CPT is
unlikely to be an effective therapeutic for COVID-19.
A different narrative is shaping up around the use of MAbs specically targeting
SARS-CoV-2. During the rst SARS epidemic in 2002, neutralizing antibodies (nAbs)
were found in SARS-CoV-1-infected patients (194, 195). Several studies following up on
these ndings identied various S-glycoprotein epitopes as the major targets of nAbs
against SARS-CoV-1 (196). Coronaviruses use trimeric spike (S) glycoproteins on their
surface to bind to the host cell, allowing for cell entry (197, 198). Each S-glycoprotein
protomer is comprised of an S1 domain, also called the receptor binding domain
(RBD), and an S2 domain. The S1 domain binds to the host cell, while the S2 domain
facilitates the fusion between the viral envelope and host cell membranes (196). The
genomic identity between the RBD of SARS-CoV-1 and SARS-CoV-2 is around 74%
(199). Due to this high degree of similarity, preexisting antibodies against SARS-CoV-1
were initially considered candidates for neutralizing activity against SARS-CoV-2. While
some antibodies developed against the SARS-CoV-1 spike protein showed cross-neu-
tralization activity with SARS-CoV-2 (200, 201), others failed to bind to SARS-CoV-2
spike protein at relevant concentrations (202). Cross-neutralizing activities were de-
pendent on whether the epitope recognized by the antibodies were conserved
between SARS-CoV-1 and SARS-CoV-2 (200).
ID and Development of Therapeutics for COVID-19
November/December 2021 Volume 6 Issue 6 e00233-21 msystems.asm.org 13
Downloaded from https://journals.asm.org/journal/msystems on 03 November 2021 by 2607:f470:34:2303:5040:4a48:5de1:cdb5.
Technological advances in antibody drug design as well as in structural biology mas-
sively accelerated the discovery of novel antibody candidates and the mechanisms by
which they interact with the target structure. Within just a year of the structure of the
SARS-CoV-2 spike protein being published, an impressive pipeline of monoclonal anti-
bodies targeting SARS-CoV-2 entered clinical trials, with hundreds more candidates in
preclinical stages. The rst human monoclonal neutralizing antibody specically against
the SARS-CoV-2 S glycoprotein was developed using hybridoma technology (203), where
antibody-producing B cells developed by mice are inserted into myeloma cells to pro-
duce a hybrid cell line (the hybridoma) that is grown in culture. The 47D11 antibody
clone was able to cross-neutralize SARS-CoV-1 and SARS-CoV-2. This antibody (now ABVV-
47D11) has recently entered clinical trials in collaboration with AbbVie. Additionally, an
extensive monoclonal neutralizing antibody pipeline has been developed to combat the
ongoing pandemic, with over 50 different antibodies in clinical trials (204). Thus far, the
monotherapy sotrovimab and two antibody cocktails (bamlanivimab/estesevimab and
casirivimab/imdevimab) have been granted EUAs by the FDA.
One of the studied antibody cocktails consists of bamlanivimab and estesevimab.
Bamlanivimab (Ly-CoV555) is a human MAb that was derived from convalescent
plasma donated by a recovered COVID-19 patient, evaluated in research by the
National Institute of Allergy and Infectious Diseases (NIAID), and subsequently devel-
oped by AbCellera and Eli Lilly. The neutralizing activity of bamlanivimab was initially
demonstrated in vivo using a nonhuman primate model (205). On the basis of these
positive preclinical data, Eli Lilly initiated the rst human clinical trial for a monoclonal
antibody against SARS-CoV-2. The phase 1 trial, which was conducted in hospitalized
COVID-19 patients, was completed in August 2020 (206). Estesevimab (LY-CoV016 or
JS-016) is also a monoclonal neutralizing antibody against the spike protein of SARS-
CoV-2. It was initially developed by Junshi Biosciences and later licensed and devel-
oped through Eli Lilly. A phase 1 clinical trial to assess the safety of etesevimab was
completed in October 2020 (207). Etesevimab was shown to bind an epitope on the
spike protein different from that of bamlanivimab, suggesting that the two antibodies
used as a combination therapy would further enhance their clinical use compared to a
monotherapy (208). To assess the efcacy and safety of bamlanivimab alone or in com-
bination with etesevimab for the treatment of COVID-19, a phase 2/3 trial (BLAZE-1)
(209) was initiated. The interim analysis of the phase 2 portion suggested that bamlani-
vimab alone was able to accelerate the reduction in viral load (210). However, more
recent data suggest that only the bamlanivimab/etesevimab combination therapy is
able to reduce viral load in COVID-19 patients (208). Based on these data, the combina-
tion therapy received an EUA for COVID-19 from the FDA in February 2021 (211).
A second therapy is comprised of casirivimab and imdevimab (REGN-COV2).
Casirivimab (REGN10933) and imdevimab (REGN10987) are two monoclonal antibodies
against the SARS-CoV-2 spike protein. They were both developed by Regeneron in a
parallel high-throughput screening (HTS) to identify neutralizing antibodies from either
humanized mice or patient-derived convalescent plasma (212). In these efforts, multi-
ple antibodies were characterized for their ability to bind and neutralize the SARS-CoV-
2 spike protein. The investigators hypothesized that an antibody cocktail, rather than
each individual antibody, could increase the therapeutic efcacy while minimizing the
risk for virus escape. Therefore, the authors tested pairs of individual antibodies for
their ability to simultaneously bind the RBD of the spike protein. Based on these data,
casirivimab and imdevimab were identied as the lead antibody pair, resulting in the
initiation of two clinical trials (213, 214). Data from this phase 1 to 3 trial published in
the New England Journal of Medicine shows that the REGN-COV2 antibody cocktail
reduced viral load, particularly in patients with high viral load or whose endogenous
immune response had not yet been initiated (215). However, in patients who already
initiated an immune response, exogenous addition of REGN-COV2 did not improve the
endogenous immune response. Both doses were well tolerated with no serious events
related to the antibody cocktail. Based on these data, the FDA granted an EUA for
Rando et al.
November/December 2021 Volume 6 Issue 6 e00233-21 msystems.asm.org 14
Downloaded from https://journals.asm.org/journal/msystems on 03 November 2021 by 2607:f470:34:2303:5040:4a48:5de1:cdb5.
REGN-COV2 in patients with mild to moderate COVID-19 who are at risk of developing
severe disease (216). Ongoing efforts are trying to evaluate the efcacy of REGN-COV2
to improve clinical outcomes in hospitalized patients (213).
Sotrovimab is the most recent MAb to receive an EUA. It was identied in the mem-
ory B cells of a 2003 survivor of SARS (217) and was found to be cross-reactive with
SARS-CoV-2 (201). This cross-reactivity is likely attributable to conservation within the
epitope, with 17 out of 22 residues conserved between the two viruses, four conserva-
tively substituted, and one semiconservatively substituted (201). In fact, these residues
are highly conserved among sarbecoviruses, a clade that includes SARS-CoV-1 and
SARS-CoV-2 (201). This versatility has led to it being characterized as a super-antibody
(218), a potent, broadly neutralizing antibody (219). Interim analysis of data from a clin-
ical trial (220) reported high safety and efcacy of this MAb in 583 COVID-19 patients
(221). Compared to placebo, sotrovimab was found to be 85% more effective in reduc-
ing progression to the primary endpoint, which was the proportion of patients who,
within 29 days, were either hospitalized for more than 24 h or died. Additionally, rates
of adverse events were comparable, and in some cases lower, among patients receiv-
ing sotrovimab compared to patients receiving a placebo. Sotrovimab therefore repre-
sents a MAb therapeutic that is effective against SARS-CoV-2 and may also be effective
against other sarbecoviruses.
Several potential limitations remain in the application of MAbs to the treatment of
COVID-19. One of the biggest challenges is identifying antibodies that not only bind to
their target but also prove to be benecial for disease management. Currently, use of
MAbs is limited to people with mild to moderate disease that are not hospitalized, and
it has yet to be determined whether they can be used as a successful treatment option
for severe COVID-19 patients. While preventing people from developing severe illness
provides signicant benets, patients with severe illness are at the greatest risk of
death, and therefore therapeutics that provide benets against severe illness are par-
ticularly desirable. It remains to be seen whether MAbs confer any benets for patients
in this category.
Another concern about therapeutics designed to amplify the response to a specic
viral target is that they may need to be modied as the virus evolves. With the ongoing
global spread of new SARS-CoV-2 variants, there is a growing concern that mutations
in the SARS-CoV-2 spike protein could escape antibody neutralization, thereby reduc-
ing the efcacy of monoclonal antibody therapeutics and vaccines. A comprehensive
mutagenesis screen recently identied several amino acid substitutions in the SARS-
CoV-2 spike protein that can prevent antibody neutralization (222). While some muta-
tions result in resistance to only one antibody, others confer broad resistance to multi-
ple MAbs as well as polyclonal human sera, suggesting that some amino acids are hot
spotsfor antibody resistance. However, it was not investigated whether the resistance
mutations identied result in a tness advantage. Accordingly, an impact on neutraliz-
ing efciency has been reported for the B.1.1.7 (Alpha) variant rst identied in the
United Kingdom and the B.1.351 (Beta) variant rst identied in in South Africa (223
225). As of 25 June 2021, the CDC recommended a pause in the use of bamlanivimab
and etesevimab due to decreased efcacy against the P.1 (Gamma) and B.1.351 (Beta)
variants of SARS-CoV-2 (226). While the reported impact on antibody neutralization
needs to be conrmed in vivo, it suggests that some adjustments to therapeutic anti-
body treatments may be necessary to maintain the efcacy that was reported in previ-
ous clinical trials.
Several strategies have been employed to try to mitigate the risk of diminished anti-
body neutralization. Antibody cocktails such as those already holding an EUA may help
overcome the risk for attenuation of the neutralizing activity of a single monoclonal
antibody. These cocktails consist of antibodies that recognize different epitopes on the
spike protein, decreasing the likelihood that a single amino acid change can cause re-
sistance to all antibodies in the cocktail. However, neutralizing resistance can emerge
even against an antibody cocktail if the individual antibodies target subdominant
ID and Development of Therapeutics for COVID-19
November/December 2021 Volume 6 Issue 6 e00233-21 msystems.asm.org 15
Downloaded from https://journals.asm.org/journal/msystems on 03 November 2021 by 2607:f470:34:2303:5040:4a48:5de1:cdb5.
epitopes (224). Another strategy is to develop broadly neutralizing antibodies that tar-
get structures that are highly conserved, as these are less likely to mutate (227, 228) or
to target epitopes that are insensitive to mutations (229). Sotrovimab, one such
super-antibody,is thought to be somewhat robust to neutralization escape (230) and
has been found to be effective against all variants assessed as of 12 August 2021 (231).
Another antibody (ADG-2) targets a highly conserved epitope that overlaps the human
angiotensin-converting enzyme 2 (hACE2) binding site of all clade 1 sarbecoviruses
(232). Prophylactic administration of ADG-2 in an immunocompetent mouse model of
COVID-19 resulted in protection against viral replication in the lungs and respiratory
burden. Since the epitope targeted by ADG-2 represents an Achillesheel for clade 1
sarbecoviruses, this antibody, like sotrovimab, might be a promising candidate against
all circulating variants as well as emerging SARS-related coronaviruses. To date, it has
fared well against the Alpha, Beta, Gamma, and Delta variants (231).
The development of MAbs against SARS-CoV-2 has made it clear that this technol-
ogy is rapidly adaptable and offers great potential for the response to emerging viral
threats. However, additional investigation may be needed to adapt MAb treatments to
SARS-CoV-2 as it evolves and potentially to pursue designs that confer benets for
patients at the greatest risk of death. While polyclonal antibodies from convalescent
plasma have been evaluated as a treatment for COVID-19, these studies have sug-
gested fewer potential benets against SARS-CoV-2 than MAbs; convalescent plasma
therapy has been thoroughly reviewed elsewhere (188, 189). Thus, advances in biolog-
ics for COVID-19 illustrate that an understanding of how the host and virus interact can
guide therapeutic approaches. The FDA authorization of two combination MAb thera-
pies, in particular, underscores the potential for this strategy to allow for a rapid
response to a novel pathogen. Additionally, while TCZ is not yet as established, this
therapy suggests that the strategy of using biologics to counteract the cytokine storm
response may provide therapies for the highest-risk patients.
HIGH-THROUGHPUT SCREENING FOR DRUG REPURPOSING
The drug development process is slow and costly, and developing compounds specif-
ically targeted to an emerging viral threat is not a practical short-term solution.
Screening existing drug compounds for alternative indications is a popular alternative
(233236). HTS has been a goal of pharmaceutical development since at least the mid-
1980s (237). Traditionally, phenotypic screens were used to test which compounds
would induce a desired change in in vitro or in vivo models, focusing on empirical, func-
tion-oriented exploration naive to molecular mechanism (238240). In many cases, these
screens utilize large libraries that encompass a diverse set of agents varying in many
pharmacologically relevant properties (e.g., reference 241). The compounds inducing a
desired effect could then be followed up on. Around the turn of the millennium, advan-
ces in molecular biology allowed for HTS to shift toward screening for compounds inter-
acting with a specic molecular target under the hypothesis that modulating that target
would have a desired effect. These approaches both offer pros and cons, and today a
popular view is that they are most effective in combination (238, 240, 242).
Today, some efforts to screen compounds for potential repurposing opportunities are
experimental, but others use computational HTS approaches (233, 243). Computational
drug repurposing screens can take advantage of big data in biology (17) and as a result
aremuchmorefeasibletodaythanduringtheheightoftheSARSandMERSoutbreaksin
the early 2000s and early 2010s, respectively. Advancements in robotics also facilitate the
experimental component of HTS (235). For viral diseases, the goal of drug repurposing is
typically to identify existing drugs that have an antiviral effect likely to impede the virus of
interest. While both small molecules and biologics can be candidates for repurposing, the
signicantly lower price of many small-molecule drugs means that they are typically more
appealing candidates (244).
Depending on the study design, screens vary in how closely they are tied to a hy-
pothesis. As with the candidate therapeutics described above, high-throughput
Rando et al.
November/December 2021 Volume 6 Issue 6 e00233-21 msystems.asm.org 16
Downloaded from https://journals.asm.org/journal/msystems on 03 November 2021 by 2607:f470:34:2303:5040:4a48:5de1:cdb5.
experimental or computational screens can proceed based on a hypothesis. Just as
remdesivir was selected as a candidate antiviral because it is a nucleoside analog (245),
so too can high-throughput screens select libraries of compounds based on a molecu-
lar hypothesis. Likewise, when the library of drugs is selected without basis in a poten-
tial mechanism, a screen can be considered hypothesis free (245). Today, both types of
analyses are common both experimentally and computationally. Both strategies have
been applied to identifying candidate therapeutics against SARS-CoV-2.
Hypothesis-driven screening. Hypothesis-driven screens often select drugs likely
to interact with specic viral or host targets or drugs with desired clinical effects, such
as immunosuppressants. There are several properties that might identify a compound
as a candidate for an emerging viral disease. Drugs that interact with a target that is
shared between pathogens (i.e., a viral protease or a polymerase) or between a viral
pathogen and another illness (i.e., a cancer drug with antiviral potential) are potential
candidates, as are drugs that are thought to interact with additional molecular targets
beyond those they were developed for (243). Such research can be driven by in vitro or
in silico experimentation. Computational analyses depend on identifying compounds
that modulate preselected proteins in the virus or host. As a result, they build on ex-
perimental research characterizing the molecular features of the virus, host, and candi-
date compounds (236).
One example of the application of this approach to COVID-19 research comes from
work on protease inhibitors. Studies have shown that viral proteases play an important
role in the life cycle of viruses, including coronaviruses, by modulating the cleavage of vi-
ral polyprotein precursors (246). Several FDA-approved drugs target proteases, such as
lopinavir and ritonavir for human immunodeciency virus (HIV) infection and simeprevir
for hepatitis C virus infection. Serine protease inhibitors were previously suggested as
possible treatments for SARS and MERS (247). One early study (197) suggested that
camostat mesylate, a protease inhibitor, could block the entry of SARS-CoV-2 into lung
cells in vitro. Two polyproteins encoded by the SARS-CoV-2 replicase gene, pp1a and
pp1ab, are critical for viral replication and transcription (248). These polyproteins must
undergo proteolytic processing, which is usually conducted by the main protease (M
Pro
),
a 33.8-kDa SARS-CoV-2 protease that is therefore fundamental to viral replication and
transcription. Therefore, it was hypothesized that compounds targeting M
Pro
could be
used to prevent or slow the replication of the SARS-CoV-2 virus.
Both computational and experimental approaches facilitated the identication of
compounds that might inhibit SARS-CoV-2 M
Pro
. In 2005, computer-aided design facili-
tated the development of a Michael acceptor inhibitor, now known as N3, to target M
Pro
of SARS-like coronaviruses (249). N3 binds in the substrate binding pocket of M
Pro
in sev-
eral human CoVs (HCoVs) (249252). The structure of N3-bound SARS-CoV-2 M
Pro
has
been solved, conrming the computational prediction that N3 would similarly bind in
the substrate binding pocket of SARS-CoV-2 (248). N3 was tested in vitro on SARS-CoV-2-
infected Vero cells, which belong to a line of cells established from the kidney epithelial
cells of an African green monkey, and was found to inhibit SARS-CoV-2 (248). A library of
approximately 10,000 compounds was screened in a uorescence resonance energy
transfer assay constructed using SARS-CoV-2 M
Pro
expressed in Escherichia coli (248).
Six leads were identied in this hypothesis-driven screen. In vitro analysis revealed
that ebselen had the strongest potency in reducing the viral load in SARS-CoV-2-
infected Vero cells (248). Ebselen is an organoselenium compound with anti-inamma-
tory and antioxidant properties (253). Molecular dynamics analysis further demon-
strated the potential for ebselen to bind to M
Pro
and disrupt the proteases enzymatic
functions (254). However, ebselen is likely to be a promiscuous binder, which could di-
minish its therapeutic potential (248, 255), and compounds with higher specicity may
be needed to translate this mechanism effectively to clinical trials. In July 2020, phase
2 clinical trials commenced to assess the effects of SPI-1005, an investigational drug
from Sound Pharmaceuticals that contains ebselen (256), on 60 adults presenting with
each of moderate (257) and severe (258) COVID-19. Other M
Pro
inhibitors are also being
ID and Development of Therapeutics for COVID-19
November/December 2021 Volume 6 Issue 6 e00233-21 msystems.asm.org 17
Downloaded from https://journals.asm.org/journal/msystems on 03 November 2021 by 2607:f470:34:2303:5040:4a48:5de1:cdb5.
evaluated in clinical trials (259, 260). Pending the results of clinical trials, N3 remains a
computationally interesting compound based on both computational and experimen-
tal data, but whether these potential effects will translate to the clinic remains
unknown.
Hypothesis-free screening. Hypothesis-free screens use a discovery-driven
approach, where screens are not targeted to specic viral proteins, host proteins, or
desired clinical modulation. Hypothesis-free drug screening began 20 years ago with
the testing of libraries of drugs experimentally. Today, like many other areas of biology,
in silico analyses have become increasingly popular and feasible through advances in
biological big data (245, 261). Many efforts have collected data about interactions
between drugs and SARS-CoV-2 and about the host genomic response to SARS-CoV-2
exposure, allowing for hypothesis-free computational screens that seek to identify new
candidate therapeutics. Thus, they utilize a systems biology paradigm to extrapolate
the effect of a drug against a virus based on the host interactions with both the virus
and the drug (236).
Resources such as the COVID-19 Drug and Gene Set Library, which at the time of its
publication contained 1,620 drugs sourced from 173 experimental and computational
drug sets and 18,676 human genes sourced from 444 gene sets (262), facilitate such
discovery-driven approaches. Analysis of these databases indicated that some drugs
had been identied as candidates across multiple independent analyses, including
high-prole candidates such as CQ/HCQ and remdesivir (262). Computational screen-
ing efforts can then mine databases and other resources to identify potential PPIs
among the host, virus, and established and/or experimental drugs (263). Subject mat-
ter expertise from human users may be integrated to various extents depending on
the platform (e.g., references 263 and 264). These resources have allowed studies to
identify potential therapeutics for COVID-19 without an a priori reason for selecting
them.
One example of a hypothesis-free screen for COVID-19 drugs comes from a PPI net-
work-based analysis that was published early in the pandemic (265). Here, researchers
cloned the proteins expressed by SARS-CoV-2 in vitro and quantied 332 virus-host PPIs
using afnity purication mass spectrometry (265). They identied two SARS-CoV-2 pro-
teins (Nsp6 and Orf9c) that interacted with host Sigma-1 and Sigma-2 receptors. Sigma
receptors are located in the endoplasmic reticulum of many cell types, and type 1 and 2
Sigma receptors have overlapping but distinct afnities for a variety of ligands (266).
Molecules interacting with the Sigma receptors were then analyzed and found to have
an effect on viral infectivity in vitro (265). A follow-up study evaluated the effect of per-
turbing these 332 proteins in two cell lines, A549 and Caco-2, using knockdown and
knockout methods, respectively, and found that the replication of SARS-CoV-2 in cells
from both lines was dependent on the expression of SIGMAR1, which is the gene that
encodes the Sigma-1 receptor (267). Following these results, drugs interacting with
Sigma receptors were suggested as candidates for repurposing for COVID-19 (e.g., refer-
ence 268). Because many well-known and affordable drugs interact with the Sigma
receptors (265, 269), they became a major focus of drug repurposing efforts. Some of
the drugs suggested by the apparent success of Sigma receptor-targeting drugs were al-
ready being investigated at the time. HCQ, for example, forms ligands with both Sigma-1
and Sigma-2 receptors and was already being explored as a candidate therapeutic for
COVID-19 (265). Thus, this computational approach yielded interest in drugs whose anti-
viral activity was supported by initial in vitro analyses.
Follow-up research, however, called into question whether the emphasis on drugs
interacting with Sigma receptors might be based on a spurious association (270). This
study built on the prior work by examining whether antiviral activity among com-
pounds correlated with their afnity for the Sigma receptors and found that it did not.
The study further demonstrated that cationic amphiphilicity was a shared property
among many of the candidate drugs identied through both computational and phe-
notypic screens and that it was likely to be the source of many compoundsproposed
Rando et al.
November/December 2021 Volume 6 Issue 6 e00233-21 msystems.asm.org 18
Downloaded from https://journals.asm.org/journal/msystems on 03 November 2021 by 2607:f470:34:2303:5040:4a48:5de1:cdb5.
antiviral activity (270). Cationic amphiphilicity is associated with the induction of phos-
pholipidosis, which is when phospholipids accumulate in the lysosome (271).
Phospholipidosis can disrupt viral replication by inhibiting lipid processing (272) (see
the discussion of HCQ in the Appendix). However, phospholipidosis is known to trans-
late poorly from in vitro models to in vivo models or clinical applications. Thus, this
nding suggested that these screens were identifying compounds that shared a physi-
ochemical property rather than a specic target (270). The authors further demon-
strated that antiviral activity against SARS-CoV-2 in vitro was correlated with the induc-
tion of phospholipidosis for drugs both with and without cationic amphiphilicity (270).
This nding supports the idea that the property of cationic amphicility was being
detected as a proxy for the shared effect of phospholipidosis (270). They demonstrated
that phospholipidosis-inducing drugs were not effective at preventing viral propaga-
tion in vivo in a murine model of COVID-19 (270). Therefore, removing hits that induce
phospholipidosis from computational and in vitro experimental repurposing screens
(e.g., reference 273) may help emphasize those that are more likely to provide clinical
benets. This work illustrates the importance of considering confounding variables in
computational screens, a principle that has been incorporated into more traditional
approaches to drug development (274).
One drug that acts on Sigma receptors does, however, remain a candidate for the
treatment of COVID-19. Several psychotropic drugs target Sigma receptors in the central
nervous system and thus attracted interest as potential COVID-19 therapeutics following
the ndings of two host-virus PPI studies (275). For several of these drugs, the in vitro
antiviral activity (267) was not correlated with their afnity for the Sigma-1 receptor (270,
275) but was correlated with phospholipidosis (270). However, uvoxamine, a selective
serotonin reuptake inhibitor that is a particularly potent Sigma-1 receptor agonist (275),
has shown promise as a preventative of severe COVID-19 in a preliminary analysis of
data from the large-scale TOGETHER trial (145). As of 6 August 2021, this trial had col-
lected data from over 1,400 patients in the uvoxamine arm of their study, half of whom
received a placebo (145). Only 74 patients in the uvoxamine group had progressed to
hospitalization for COVID-19 compared to 107 in the placebo group, corresponding to a
relative risk of 0.69; additionally, the relative risk of mortality between the two groups
was calculated to be 0.71. These ndings support the results of small clinical trials that
have found uvoxamine to reduce clinical deterioration relative to a placebo (276, 277).
However, the ongoing therapeutic potential of uvoxamine does not contradict the nd-
ing that hypothesis-free screening hits can be driven by confounding factors. The
authors point out that its relevance would not just be antiviral as it has a potential
immunomodulatory mechanism (276). It has been found to be protective against septic
shock in an in vivo mouse model (278). It is possible that uvoxamine also exerts an anti-
viral effect (279). Thus, Sigma-1 receptor activity may contribute to uvoxamines poten-
tial effects in treating COVID-19, but it is not the only mechanism by which this drug can
interfere with disease progression.
Potential and limitations of high-throughput analyses. Computational screening
allows for a large number of compounds to be evaluated to identify those most likely
to display a desired behavior or function. This approach can be guided by a hypothesis
or can aim to discover underlying characteristics that produce new hypotheses about
the relationship between a host, a virus, and candidate pharmaceuticals. The examples
outlined above illustrate that HTS-based evaluations of drug repurposing can potentially
provide valuable insights. Computational techniques were used to design compounds
targeting M
Pro
based on an understanding of how this protease aids viral replication, and
M
Pro
inhibitors remain promising candidates (235), although the clinical trial data are not
yet available. Similarly, computational analysis correctly identied the Sigma-1 receptor
as a protein of interest. Although the process of identifying which drugs might modulate
the interaction led to an emphasis on candidates that ultimately have not been sup-
ported, uvoxamine remains an appealing candidate. The difference between the pre-
liminary evidence for uvoxamine compared to other drugs that interact with Sigma
ID and Development of Therapeutics for COVID-19
November/December 2021 Volume 6 Issue 6 e00233-21 msystems.asm.org 19
Downloaded from https://journals.asm.org/journal/msystems on 03 November 2021 by 2607:f470:34:2303:5040:4a48:5de1:cdb5.
receptors underscores a major critique of hypothesis-free HTS in particular: while these
approaches allow for brute force comparison of a large number of compounds against a
virus of interest, they lose the element of expertise that is associated with most successes
in drug repurposing (245).
There are also practical limitations to these methods. One concern is that computa-
tional analyses inherently depend on the quality of the data being evaluated. The ur-
gency of the COVID-19 pandemic led many research groups to pivot toward computa-
tional HTS research without familiarity with best practices in this area (235). As a result,
there is an excessive amount of information available from computational studies
(280), but not all of it is high quality. Additionally, the literature used to identify and
validate targets can be difcult to reproduce (281), which may pose challenges to tar-
get-based experimental screening and to in silico screens. Some efforts to repurpose
antivirals have focused on host, rather than viral, proteins (236), which might be
expected to translate poorly in vivo if the targeted proteins serve essential functions in
the host. Concerns about the practicality of hypothesis-free screens to gain novel
insights are underscored by the fact that very few or possibly no success stories have
emerged from hypothesis-free screens over the past 20 years (245). These ndings sug-
gest that data-driven research can be an important component of the drug repurpos-
ing ecosystem, but that drug repurposing efforts that proceed without a hypothesis,
an emphasis on biological mechanisms, or an understanding of confounding effects
may not produce viable candidates.
CONSIDERATIONS IN BALANCING DIFFERENT APPROACHES
The approaches described here offer a variety of advantages and limitations in
responding to a novel viral threat and building on existing bodies of knowledge in dif-
ferent ways. Medicine, pharmacology, basic science (especially virology and immunol-
ogy), and biological data science can all provide different insights and perspectives for
addressing the challenging question of which existing drugs might provide benets
against an emerging viral threat. A symptom management-driven approach allows
clinicians to apply experience with related diseases or related symptoms to organize a
rapid response aimed at saving the lives of patients already infected with a new dis-
ease. Oftentimes, the pharmaceutical agents that are applied are small-molecule,
broad-spectrum pharmaceuticals that are widely available and affordable to produce,
and they may already be available for other purposes, allowing clinicians to administer
them to patients quickly either with an EUA or off-label. In this vein, dexamethasone
has emerged as the strongest treatment against severe COVID-19 (Table 1).
Alternatively, many efforts to repurpose drugs for COVID-19 have built on information
gained through basic scientic research of HCoV. Understanding how related viruses
function has allowed researchers to identify possible pharmacological strategies to dis-
rupt pathogenesis (Fig. 3). Some of the compounds identied through these methods
include small-molecule antivirals, which can be boutique and experimental medications
like remdesivir (Table 1). Other candidate drugs that intercept host-pathogen interac-
tions include biologics, which imitate the function of endogenous host compounds.
Most notably, several MAbs that have been developed (casirivimab, imdevimab, bamla-
nivimab, and etesevimab) or repurposed (sotrovimab and tocilizumab) have now been
granted EUAs (Table 1). Although not discussed here, several vaccine development pro-
grams have also met huge success using a range of strategies (2).
All of the small-molecule drugs evaluated and most of the biologics are repurposed,
and thus hinge on a theoretical understanding of how the virus interacts with a human
host and how pharmaceuticals can be used to modify those interactions rather than
being designed specically against SARS-CoV-2 or COVID-19. As a result, signicant
attention has been paid to computational approaches that automate the identication
of potentially desirable interactions. However, work in COVID-19 has made it clear that
relevant compounds can also be masked by confounding factors, and spurious associa-
tions can drive investment in candidate therapeutics that are unlikely to translate to
Rando et al.
November/December 2021 Volume 6 Issue 6 e00233-21 msystems.asm.org 20
Downloaded from https://journals.asm.org/journal/msystems on 03 November 2021 by 2607:f470:34:2303:5040:4a48:5de1:cdb5.
TABLE 1 Summary table of candidate therapeutics examined in this paper
Treatment Grade
a
Category FDA status
b
Evidence available
c
Suggested effectiveness
d
Dexamethasone A Small molecule, broad
spectrum
Used off-label RCT Supported: RCT shows improved outcomes over the SOC,
especially in severe cases such as CRS
Remdesivir A Small molecule, antiviral,
adenosine analog
Approved for
COVID-19 (and
EUA for
combination with
baricitinib)
RCT Mixed: Conicting evidence from large WHO-led Solidarity
trial vs U.S.-focused RCT and other studies
Tocilizumab A Biologic, monoclonal
antibody
EUA RCT Mixed: It appears that TCZ may work well in combination
with dexamethasone in severe cases, but not as
monotherapy
Sotrovimab NA Biologic, monoclonal
antibody
EUA RCT Supported: Phase 2/3 clinical trial showed reduced
hospitalization/death
Bamlanivimab and etesevimab B and NA Biologic, monoclonal
antibodies
EUA RCT Supported: Phase 2 clinical trial showed reduction in viral
load, but FDA pause recommended because it may be
less effective against the Delta variant
Casirivimab and imdevimab NA Biologic, monoclonal
antibodies
EUA RCT Supported: Reduced viral load at interim analysis
Fluvoxamine B Small-molecule, Sigma-1
receptor agonist
NA RCT Supported: Support from two small RCTs and preliminary
support from interim analysis of TOGETHER trial
SNG001 B Biologic, interferon None RCT Mixed: Support from initial RCT but no effect found in
WHOs Solidarity trial
M
Pro
protease inhibitors NA Small molecule,
protease inhibitor
None Computational prediction,
in vitro studies
Unknown
ARBs and ACEIs C Small molecule, broad
spectrum
None Observational studies and
some RCTs
Not supported: Observational study retracted, RCTs suggest
no association
Favipiravir D Small molecule, antiviral,
nucleoside analog
None RCT Not supported: RCTs do not show signicant improvements
for individuals taking this treatment, good safety prole
HCQ/CQ D Small molecule, broad
spectrum
None RCT Not supported, possibly harmful: Nonblinded RCTs showed
no improvement over the SOC, safety prole may be
problematic
Convalescent plasma transfusion D Biologic, polyclonal
antibodies
EUA RCT Mixed: Supported in small trials but not in large-scale
RECOVERY trial
Ivermectin D Small molecule, broad
spectrum
None RCT Mixed: Mixed results from small RCTs, major supporting RCT
now withdrawn, Preliminary results of large RCT
(TOGETHER trial) suggest no effect on emergency room
visits or hospitalization for COVID-19
a
Gradeis the rating given to each treatment by the Systematic Tracker of Off-label/Repurposed Medicines Grades (STORM) maintained by the Center for Cytokine Storm Treatment & Laboratory (CSTL) at the University of
Pennsylvania (70). A grade of A indicates that a treatment is considered effective, a grade of B indicates that all or most RCTs have shown positive results, a grade of C indicates that RCT data are not yet available, and a grade of D
indicates that multiple RCTs have produced negative results. Treatments not in the STORM database are indicated as NA for not available.
b
FDA status is also provided where available.
c
The evidence available is based on the progression of the therapeutic through the pharmaceutical development pipeline, with RCTs as the most informative source of evidence.
d
The effectiveness is summarized based on the current available evidence; large trials such as RECOVERY and Solidarity trial are weighted heavily in this summary. This table was last updated on 20 August 2021. This table was last
updated on 20 August 2021.
ID and Development of Therapeutics for COVID-19
November/December 2021 Volume 6 Issue 6 e00233-21 msystems.asm.org 21
Downloaded from https://journals.asm.org/journal/msystems on 03 November 2021 by 2607:f470:34:2303:5040:4a48:5de1:cdb5.
the clinic. Such spurious hits are especially likely to impact hypothesis-free screens.
However, hypothesis-free screens may still be able to contribute to the drug discovery
or repurposing ecosystem, assuming the computational arm of HTS follows the same
trends seen in its experimental arm. In 2011, a landmark study in drug discovery dem-
onstrated that although more new drugs were discovered using target-based rather
than phenotypic approaches, the majority of drugs with a novel molecular mechanism
of action (MMOA) were identied in phenotypic screens (282). This pattern applied
only to rst-in-class drugs, with most follower drugs produced by target-based screen-
ing (239). These ndings suggest that target-based drug discovery is more successful
when building on a known MMOA and that modulating a target is most valuable when
the target is part of a valuable MMOA (240). Building on this, many within the eld sug-
gested that mechanism-informed phenotypic investigations may be the most useful
approach to drug discovery (238, 240, 242). As it stands, data-driven efforts to identify
patterns in the results of computational screens allowed researchers to notice the
shared property of cationic amphicility among many of the hits from computational
screening analyses (270). While easier said than done, efforts to ll in the black box
underlying computational HTS and recognize patterns among the identied com-
pounds aid in moving data-oriented drug repurposing efforts in this direction.
The unpredictable nature of success and failure in drug repurposing for COVID-19 thus
highlights one of the tenets of phenotypic screening: there are a lot of unknown
unknowns,and a promising mechanism at the level of an MMOA will not necessarily prop-
agate up to the pathway, cellular, or organismal level (238). Despite the fact that apparently
mechanistically relevant drugs may exist, identifying effective treatments for a new viral dis-
ease is extremely challenging. Targets of repurposed drugs are often nonspecic, meaning
that the MMOA can appear to be relevant to COVID-19 without a therapeutic or prophylac-
tic effect being observed in clinical trials. The difference in the current status of remdesivir
and favipiravir as treatments for COVID-19 (Table 1) underscores how difcult it is to predict
whether a specic compound will produce a desired effect, even when the mechanisms are
similar. Furthermore, the fact that many candidate COVID-19 therapeutics were ultimately
identied because of their shared propensity to induce phospholipidosis underscores how
challenging it can be to identify a mechanism in silico or in vitro that will translate to a suc-
cessful treatment. While signicant progress has been made thus far in the pandemic, the
therapeutic landscape is likely to continue to evolve as more results become available from
clinical trials and as efforts to develop novel therapeutics for COVID-19 progress.
TOWARDS THE NEXT HCOV THREAT
Only very limited testing of candidate therapies was feasible during the SARS and
MERS epidemics, and as a result, few treatments were available at the outset of the
COVID-19 pandemic. Even corticosteroids, which were used to treat SARS patients,
were a controversial therapeutic prior to the release of the results of the large
RECOVERY trial. The scale and duration of the COVID-19 pandemic have made it possi-
ble to conduct large, rigorous RCTs such as RECOVERY, Solidarity, TOGETHER, and
others. As results from these trials have continued to emerge, it has become clear that
small clinical trials often produce spurious results. In the case of HCQ/CQ, the therapeu-
tic had already attracted so much attention based on small, preliminary (and in some
cases, methodologically concerning) studies that it took the results of multiple large
studies before attention began to be redirected to more promising candidates (283). In
fact, most COVID-19 clinical trials lack the statistical power to reliably test their hypoth-
eses (284, 285). In the face of an urgent crisis like COVID-19, the desire to act quickly is
understandable, but it is imperative that studies maintain strict standards of scientic
rigor (235, 274), especially given the potential dangers of politicization, as illustrated
by HCQ/CQ (286). Potential innovations in clinical trial structure, such as adaptable clin-
ical trials with master protocols (287) or the sharing of data among small clinical trials
(285) may help to address future crises and to bolster the results from smaller studies,
respectively.
Rando et al.
November/December 2021 Volume 6 Issue 6 e00233-21 msystems.asm.org 22
Downloaded from https://journals.asm.org/journal/msystems on 03 November 2021 by 2607:f470:34:2303:5040:4a48:5de1:cdb5.
In the long term, new drugs specic for treatment of COVID-19 may also enter de-
velopment. Development of novel drugs is likely to be guided by what is known about
the pathogenesis and molecular structure of SARS-CoV-2. For example, understanding
the various structural components of SARS-CoV-2 may allow for the development of
small-molecule inhibitors of those components. Crystal structures of the SARS-CoV-2
main protease have been resolved (248, 288). Much work remains to be done to deter-
mine further crystal structures of other viral components, understand the relative utility
of targeting different viral components, perform additional small-molecule inhibitor
screens, and determine the safety and efcacy of the potential inhibitors. While still
nascent, work in this area is promising. Over the longer term, this approach and others
may lead to the development of novel therapeutics specically for COVID-19 and
SARS-CoV-2. Such efforts are likely to prove valuable in managing future emergent
HCoV, just as research from the SARS and MERS pandemic has provided a basis for the
COVID-19 response.
APPENDIX
Dexamethasone. In order to understand how dexamethasone reduces inammation,
it is necessary to consider the stress response broadly. In response to stress, corticotropin-
releasing hormone stimulates the release of neurotransmitters known as catecholamines,
such as epinephrine, and steroid hormones known as glucocorticoids, such as cortisol
(289, 290). While catecholamines are often associated with the ght-or-ight response,
the specic role that glucocorticoids play is less clear, although they are thought to be
important to restoring homeostasis (291). Immune challenge is a stressor that is known to
interact closely with the stress response. The immune system can therefore interact with
the central nervous system; for example, macrophages can both respond to and produce
catecholamines (289). Additionally, the production of both catecholamines and
glucocorticoids is associated with inhibition of proinammatory cytokines such as IL-6, IL-
12, and tumor necrosis factor alpha (TNF-
a
) and the stimulation of anti-inammatory
cytokines such as IL-10, meaning that the stress response can regulate inammatory
immune activity (290). Administration of dexamethasone has been found to correspond
to dose-dependent inhibition of IL-12 production, but not to affect IL-10 (292); the fact
that this relationship could be disrupted by administration of a glucocorticoid receptor
antagonist suggests that it is regulated by the receptor itself (292). Thus, the
administration of dexamethasone for COVID-19 is likely to simulate the release of
glucocorticoids endogenously during stress, resulting in binding of the synthetic steroid
to the glucocorticoid receptor and the associated inhibition of the production of
proinammatory cytokines. In this model, dexamethasone reduces inammation by
stimulating the biological mechanism that reduces inammation following a threat such
as immune challenge.
Initial support for dexamethasone as a treatment for COVID-19 came from the United
Kingdoms RECOVERY trial (62), which assigned over 6,000 hospitalized COVID-19
patients to the standard of care (SOC) or treatment (dexamethasone) arms of the trial at a
2:1 ratio. At the time of randomization, some patients were ventilated (16%), others were
on noninvasive oxygen (60%), and others were breathing independently (24%). Patients
in the treatment arm were administered dexamethasone either orally or intravenously at
6 mg per day for up to 10 days. The primary endpoint was the patientsstatusat28days
postrandomization (mortality, discharge, or continued hospitalization), and secondary
outcomes analyzed included the progression to invasive mechanical ventilation over the
same period. The 28-day mortality rate was found to be lower in the treatment group
than in the SOC group (21.6% versus 24.6%; P,0.001). However, the effect was driven
by improvements in patients receiving mechanical ventilation or supplementary oxygen.
One possible confounding factor is that patients receiving mechanical ventilation tended
to be younger than patients who were not receiving respiratory support (by 10 years on
average) and to have had symptoms for a longer period. However, adjusting for age did
not change the conclusions, although the duration of symptoms was found to be
ID and Development of Therapeutics for COVID-19
November/December 2021 Volume 6 Issue 6 e00233-21 msystems.asm.org 23
Downloaded from https://journals.asm.org/journal/msystems on 03 November 2021 by 2607:f470:34:2303:5040:4a48:5de1:cdb5.
signicantly associated with the effect of dexamethasone administration. Thus, the
results of this large, randomized, and multisite, albeit not placebo-controlled, study
suggests that administration of dexamethasone to patients who are unable to breathe
independently may signicantly improve survival outcomes. Additionally, dexamethasone
is a widely available and affordable medication, raising the hope that it could be made
available to COVID-19 patients globally.
It is not surprising that administration of an immunosuppressant would be most
benecial in severe cases where the immune system was dysregulated toward
inammation. However, it is also unsurprising that care must be taken in administering
an immunosuppressant to patients ghting a viral infection. In particular, the concern
has been raised that treatment with dexamethasone might increase patient susceptibility
to concurrent (e.g., nosocomial) infections (293). Additionally, the drug could potentially
slow viral clearance and inhibit patientsability to develop antibodies to SARS-CoV-2 (59,
293), with the lack of data about viral clearance being put forward as a major limitation of
the RECOVERY trial (294). Furthermore, dexamethasone has been associated with side
effects that include psychosis, glucocorticoid-induced diabetes, and avascular necrosis
(59), and the RECOVERY trial did not report outcomes with enough detail to be able to
determine whether they observed similar complications. The effects of dexamethasone
have also been found to differ among populations, especially in high-income versus
middle- or low-income countries (295). However, since the RECOVERY trials results were
released, strategies have been proposed for administering dexamethasone alongside
more targeted treatments to minimize the likelihood of negative side effects (293). Given
the available evidence, dexamethasone is currently the most promising treatment for
severe COVID-19.
Favipiravir. The effectiveness of favipiravir for treating patients with COVID-19 is
under investigation. Evidence for the drug inhibiting viral RNA polymerase is based on
time-of-drug addition studies that found that viral loads were reduced with the addition
of favipiravir in early times postinfection (80, 83, 84). An open-label, nonrandomized,
before-after controlled study for COVID-19 was recently conducted (296). The study
included 80 COVID-19 patients (35 treated with favipiravir, 45 control) from the isolation
ward of the National Clinical Research Center for Infectious Diseases (The Third Peoples
Hospital of Shenzhen), Shenzhen, China. The patients in the control group were treated
with other antivirals, such as lopinavir and ritonavir. It should be noted that although the
control patients received antivirals, two subsequent large-scale analyses, the WHO
Solidarity trial and the Randomized Evaluation of COVID-19 Therapy (RECOVERY) trial,
identied no effect of lopinavir or of a lopinavir-ritonavir combination, respectively, on
the metrics of COVID-19-related mortality that each assessed (93, 297, 298). Treatment
was applied on days 2 to 14; treatment stopped either when viral clearance was
conrmed or on day 14. The efcacy of the treatment was measured rst by the time
until viral clearance using Kaplan-Meier survival curves and second by the improvement
rate of chest computed tomography (CT) scans on day 14 after treatment. The study
found that favipiravir increased the speed of recovery, measured as viral clearance from
the patient by reverse transcription-PCR (RT-PCR), with patients receiving favipiravir
recovering in 4 days compared to 11 days for patients receiving antivirals such as
lopinavir and ritonavir. Additionally, the lung CT scans of patients treated with favipiravir
showed signicantly higher improvement rates (91%) on day 14 compared to control
patients (62%) (P= 0.004). However, there were adverse side effects in 4 (11%) favipiravir-
treated patients and 25(56%) control patients. The adverse side effects included diarrhea,
vomiting, nausea, rash, and liver and kidney injury. Despite the study reporting clinical
improvement in favipiravir-treated patients, several study design issues are problematic
and lower condence in the overall conclusions. For example, the study was neither
randomized nor carried out in a blind manner. Moreover, the selection of patients did not
take into consideration important factors such as previous clinical conditions or sex, and
there was no age categorization. Additionally, it should be noted that this study was
temporarily retracted and then restored without explanation (299).
Rando et al.
November/December 2021 Volume 6 Issue 6 e00233-21 msystems.asm.org 24
Downloaded from https://journals.asm.org/journal/msystems on 03 November 2021 by 2607:f470:34:2303:5040:4a48:5de1:cdb5.
In late 2020 and early 2021, the rst randomized controlled trials of favipiravir for the
treatment of COVID-19 released results (300302). The rst (300) used a randomized,
controlled, open-label design to compare two drugs, favipiravir and blaver marboxil, to
the SOC alone. Here, the SOC included antivirals such as lopinavir/ritonavir and was
administered to all patients. The primary endpoint analyzed was viral clearance at day
14. The sample size for this study was very small, with 29 total patients enrolled, and no
signicant effect of the treatments was found for the primary outcome or any of the
secondary outcomes analyzed, which included mortality. The second study (301) was
larger, with 96 patients enrolled, and included only individuals with mild to moderate
symptoms who were randomized into two groups: one receiving chloroquine (CQ) in
addition to the SOC, and the other receiving favipiravir in addition to the SOC. This
study reported a nonsignicant trend for patients receiving favipiravir to have a shorter
hospital stay (13.29 days compared to 15.89 for CQ; P= 0.06) and less likelihood of
progressing to mechanical ventilation (P= 0.118) or to an oxygen saturation of ,90%
(P= 0.129). These results, combined with the fact that favipiravir was being compared
to CQ, which is now widely understood to be ineffective for treating COVID-19, thus do
not suggest that favipiravir was likely to have had a strong effect on these outcomes.
On the other hand, another trial of 60 patients reported a signicant effect of favipiravir
on viral clearance at 4 days (a secondary endpoint), but not at 10 days (the primary
endpoint) (302). This study, as well as a prior study of favipiravir (303), also reported that
the drug was generally well tolerated. Thus, in combination, these small studies suggest
that the effects of favipiravir as a treatment for COVID-19 cannot be determined based
on the available evidence, but additionally, none raise major concerns about the safety
prole of the drug.
Remdesivir. At the outset of the COVID-19 pandemic, remdesivir did not have any
have any FDA-approved use. A clinical trial in the Democratic Republic of Congo found
some evidence of effectiveness against Ebola virus disease (EVD), but two antibody
preparations were found to be more effective, and remdesivir was not pursued (304).
Remdesivir also inhibits polymerase and replication of the coronaviruses MERS-CoV and
SARS-CoV-1 in cell culture assays with submicromolar 50% inhibitory concentrations (IC
50
s)
(305). It has also been found to inhibit SARS-CoV-2, showing synergy with CQ in vitro (88).
Remdesivir was rst used on some COVID-19 patients under compassionate use
guidelines (306308). All were in late stages of COVID-19 infection, and initial reports
were inconclusive about the drugsefcacy. Gilead Sciences, the maker of remdesivir,
led a recent study that reported outcomes for compassionate use of the drug in 61
patients hospitalized with conrmed COVID-19. Here, 200 mg of remdesivir was
administered intravenously on day 1, followed by a further 100 mg/day for 9 days (92).
There were signicant issues with the study design, or lack thereof. There was no
randomized control group. The inclusion criteria were variable: some patients required
only low doses of oxygen, while others required ventilation. The study included many
sites, potentially with variable inclusion criteria and treatment protocols. The patients
analyzed had mixed demographics. There was a short follow-up period of investigation.
Eight patients were excluded from the analysis mainly due to missing postbaseline
information; thus, their health was unaccounted for. Therefore, even though the study
reported clinical improvement in 68% of the 53 patients ultimately evaluated, due to
the signicant issues with study design, it could not be determined whether treatment
with remdesivir had an effect or whether these patients would have recovered
regardless of treatment. Another study comparing 5- and 10-day treatment regimens
reported similar results but was also limited because of the lack of a placebo control
(309). These studies did not alter the understanding of the efcacy of remdesivir in
treating COVID-19, but the encouraging results provided motivation for placebo-
controlled studies.
The double-blind placebo-controlled ACTT-1 trial (89, 90) recruited 1,062 patients
and randomly assigned them to placebo treatment or treatment with remdesivir.
Patients were stratied for randomization based on site and the severity of disease
ID and Development of Therapeutics for COVID-19
November/December 2021 Volume 6 Issue 6 e00233-21 msystems.asm.org 25
Downloaded from https://journals.asm.org/journal/msystems on 03 November 2021 by 2607:f470:34:2303:5040:4a48:5de1:cdb5.
presentation at baseline (89). The treatment was 200 mg on day 1, followed by 100 mg
on days 2 through 10. Data were analyzed from a total of 1,059 patients who completed
the 29-day course of the trial, with 517 assigned to remdesivir and 508 to placebo (89).
The two groups were well matched demographically and clinically at baseline. Those
who received remdesivir had a median recovery time of 10 days, compared with
15 days in those who received placebo (rate ratio for recovery, 1.29; 95% condence
interval [95% CI], 1.12 to 1.49; P,0.001). The Kaplan-Meier estimates of mortality by
14 days were 6.7% with remdesivir and 11.9% with placebo, with a hazard ratio (HR) for
death of 0.55 and a 95% CI of 0.36 to 0.83, and at day 29, remdesivir corresponded to
11.4% and the placebo to 15.2% (HR, 0.73; 95% CI, 0.52 to 1.03). Serious adverse events
were reported in 131 of the 532 patients who received remdesivir (24.6%) and in 163 of
the 516 patients in the placebo group (31.6%). This study also reported an association
between remdesivir administration and both clinical improvement and a lack of
progression to more invasive respiratory intervention in patients receiving noninvasive
and invasive ventilation at randomization (89). Largely on the results of this trial, the
FDA reissued and expanded the EUA for remdesivir for the treatment of hospitalized
COVID-19 patients ages 12 and older (310). Additional clinical trials (88, 311314) are
under way to evaluate the use of remdesivir to treat COVID-19 patients at both early
and late stages of infection and in combination with other drugs (Fig. 2). As of 22
October 2020, remdesivir received FDA approval based on three clinical trials (315).
However, results suggesting no effect of remdesivir on survival were reported by the
WHO Solidarity trial (93). Patients were randomized in equal proportions into four
experimental conditions and a control condition, corresponding to four candidate
treatments for COVID-19 and the SOC, respectively; no placebo was administered. The
2,750 patients in the remdesivir group were administered 200 mg intravenously on the
rst day and 100 mg on each subsequent day until day 10 and assessed for in-hospital
death (primary endpoint), duration of hospitalization, and progression to mechanical
ventilation. There were also 2,708 control patients who would have been eligible and
able to receive remdesivir were they not assigned to the control group. A total of 604
patients among these two cohorts died during initial hospitalization, with 301 in the
remdesivir group and 303 in the control group. The rate ratio of death between these
two groups was therefore not signicant (0.95; P= 0.50), suggesting that the
administration of remdesivir did not affect survival. The two secondary analyses similarly
did not nd any effect of remdesivir. Additionally, the authors compared data from their
study with data from three other studies of remdesivir (including reference 89) stratied
by supplemental oxygen status. A meta-analysis of the four studies yielded an overall rate
ratio for death of 0.91 (P= 0.20). These results thus do not support the previous ndings
that remdesivir reduced median recovery time and mortality risk in COVID-19 patients.
In response to the results of the Solidaritytrial, Gilead, which manufactures remdesivir,
released a statement pointing to the fact that the Solidarity trial was not placebo
controlled or double blind and at the time of release, the statement had not been peer
reviewed (316); these sentiments have been echoed elsewhere (317). Other critiques of
this study have noted that antivirals are not typically targeted at patients with severe
illness, and therefore, remdesivir could be more benecial for patients with mild cases
rather than severe cases (298, 318). However, the publication associated with the trial
sponsored by Gilead did purport an effect of remdesivir on patients with severe disease,
identifying an 11- versus 18-day recovery period (rate ratio for recovery, 1.31; 95% CI, 1.12
to 1.52) (89). Additionally, a smaller analysis of 598 patients, of whom two-thirds were
randomized to receive remdesivir for either 5 or 10 days, reported a small effect of
treatment with remdesivir for 5 days relative to the standard of care in patients with
moderate COVID-19 (319). These results suggest that remdesivir could improve outcomes
for patients with moderate COVID-19 but that additional information would be needed
to understand the effects of different durations of treatment. Therefore, the Solidarity trial
may point to limitations in the generalizability of other research on remdesivir, especially
since the broad international nature of the Solidarity clinical trial, which included
Rando et al.
November/December 2021 Volume 6 Issue 6 e00233-21 msystems.asm.org 26
Downloaded from https://journals.asm.org/journal/msystems on 03 November 2021 by 2607:f470:34:2303:5040:4a48:5de1:cdb5.
countries with a wide range of economic proles and a variety of health care systems,
provides a much-needed global perspective in a pandemic (298). On the other hand, only
62% of patients in the Solidarity trial were randomized on the day of admission or 1 day
afterwards (93), and concerns have been raised that differences in disease progression
could inuence the effectiveness of remdesivir (298). Despite the ndings of the
Solidarity trial, remdesivir remains available for the treatment of COVID-19 in many
places. Remdesivir has also been investigated in combination with other drugs, such as
baricitinib, which is an inhibitor of Janus kinase 1 and 2 (320); the FDA has issued an EUA
for the combination of remdesivir and baricitinib in adult and pediatric patients (321).
Follow-up studies are needed and, in many cases, are under way to further investigate
remdesivir-related outcomes.
Similarly, the extent to which the remdesivir dosing regimen could inuence outcomes
continues to be under consideration. A randomized, open-label trial compared the effect of
remdesivir on 397 patients with severe COVID-19 over 5 versus 10 days (91, 309),
complementing the study that found that a 5-day course of remdesivir improved outcomes
for patients with moderate COVID-19 but a 10-day course did not (319). Patients in the two
groups were administered 200 mg of remdesivir intravenously on the rst day, followed by
100 mg on the subsequent 4 or 9 days, respectively. The two groups differed signicantly
in their clinical status, with patients assigned to the 10-day group having more severe
illness. This study also differed from most because it included not only adults, but also
pediatric patients as young as 12 years old. It reported no signicant differences across
several outcomes for patients receiving a 5-day or 10-day course, when correcting for
baseline clinical status. The data did suggest that the 10-day course might reduce mortality
in the most severe patients at day 14, but the representation of this group in the study
population was too low to justify any conclusions (309). Thus, additional research is also
required to determine whether the dosage and duration of remdesivir administration
inuences outcomes.
In summary, remdesivir is the rst FDA-approved antiviral against SARS-CoV-2 as
well as the rst FDA-approved COVID-19 treatment. Early investigations of this drug
established proof of principle that drugs targeting the virus can benet COVID-19
patients. Moreover, one of the most successful strategies for developing therapeutics
for viral diseases is to target the viral replication machinery, which are typically virally
encoded polymerases. Small-molecule drugs targeting viral polymerases are the
backbones of treatments for other viral diseases, including human immunodeciency
virus (HIV) and herpes. Notably, the HIV and herpesvirus polymerases are a reverse
transcriptase and a DNA polymerase, respectively, whereas SARS-CoV-2 encodes an
RdRP, so most of the commonly used polymerase inhibitors are not likely to be active
against SARS-CoV-2. In clinical use, polymerase inhibitors show short-term benets for
HIV patients, but for long-term benets, they must be part of combination regimens.
They are typically combined with protease inhibitors, integrase inhibitors, and even
other polymerase inhibitors. Remdesivir provides evidence that a related approach may
be benecial for the treatment of COVID-19.
Hydroxychloroquine and chloroquine. Chloroquine (CQ) and hydroxychloroquine
(HCQ) increase cellular pH by accumulating in their protonated form inside lysosomes
(95, 322). This shift in pH inhibits the breakdown of proteins and peptides by the
lysosomes during the process of proteolysis (95). Interest in CQ and HCQ for treating
COVID-19 was catalyzed by a mechanism observed in in vitro studies of both SARS-CoV-
1 and SARS-CoV-2. In one study, CQ inhibited viral entry of SARS-CoV-1 into Vero E6
cells, a cell line that was derived from Vero cells in 1968, through the elevation of
endosomal pH and the terminal glycosylation of ACE2 (96). Increased pH within the cell,
as discussed above, inhibits proteolysis, and terminal glycosylation of ACE2 is thought
to interfere with virus receptor binding. An in vitro study of SARS-CoV-2 infection of
Vero cells found both HCQ and CQ to be effective in inhibiting viral replication, with
HCQ being more potent (97). Additionally, an early case study of three COVID-19
patients reported the presence of antiphospholipid antibodies in all three patients
ID and Development of Therapeutics for COVID-19
November/December 2021 Volume 6 Issue 6 e00233-21 msystems.asm.org 27
Downloaded from https://journals.asm.org/journal/msystems on 03 November 2021 by 2607:f470:34:2303:5040:4a48:5de1:cdb5.
(323). Antiphospholipid antibodies are central to the diagnosis of the antiphospholipid
syndrome, a disorder that HCQ has often been used to treat (324326). Because the
90% effective concentration (EC
90
) of CQ in Vero E6 cells (6.90
m
M) can be achieved in
and tolerated by rheumatoid arthritis (RA) patients, it was hypothesized that it might
also be possible to achieve the effective concentration in COVID-19 patients (327).
Additionally, clinical trials have reported HCQ to be effective in treating HIV (328) and
chronic hepatitis C (329). Together, these studies triggered initial enthusiasm about the
therapeutic potential for HCQ and CQ against COVID-19. HCQ/CQ has been proposed
both as a treatment for COVID-19 and a prophylaxis against SARS-CoV-2 exposure, and
trials often investigated these drugs in combination with azithromycin (AZ) and/or zinc
supplementation. However, as more evidence has emerged, it has become clear that
HCQ/CQ offer no benets against SARS-CoV-2 or COVID-19.
(i) Trials assessing therapeutic administration of HCQ/CQ. The initial study
evaluating HCQ as a treatment for COVID-19 patients was published on 20 March 2020
by Gautret et al. (98). This nonrandomized, nonblinded, nonplacebo clinical trial
compared HCQ to the SOC in 42 hospitalized patients in southern France. It reported
that patients who received HCQ showed higher rates of virological clearance by
nasopharyngeal swab on days 3 to 6 compared to the SOC. This study also treated six
patients with both HCQ plus AZ and found this combination therapy to be more
effective than HCQ alone. However, the design and analyses used showed weaknesses
that severely limit interpretability of results, including the small sample size and the
lack of the following: randomization, blinding, placebo (no placebo pillgiven to the
SOC group), intention-to-Treat analysis, correction for sequential multiple comparisons,
and trial preregistration. Furthermore, the trial arms were entirely confounded by the
hospital, and there were false-negative outcome measurements (see reference 330).
Two of these weaknesses are due to inappropriate data analysis and can therefore be
corrected post hoc by recalculating the Pvalues (lack of intention-to-treat analysis and
multiple comparisons). However, all other weaknesses are fundamental design aws
and cannot be corrected for. Thus, the conclusions cannot be generalized outside the
study. The International Society of Antimicrobial Chemotherapy, the scientic
organization that publishes the journal where the article appeared, subsequently
announced that the article did not meet its expected standard for publications (99),
although it has not been ofcially retracted.
Because of the preliminary data presented in this study, HCQ treatment was
subsequently explored by other researchers. About 1 week later, a follow-up case study
reported that 11 consecutive patients were treated with HCQ plus AZ using the same
dosing regimen (331). One patient died, two were transferred to the intensive care unit
(ICU), and one developed a prolonged QT interval, leading to discontinuation of HCQ-
plus-AZ administration. As in the Gautret et al. study (98), the outcome assessed was
virological clearance at day 6 posttreatment, as measured from nasopharyngeal swabs.
Of the 10 living patients on day 6, 8 remained positive for SARS-CoV-2 RNA. As in the
original study, interpretability was severely limited by the lack of a comparison group
and the small sample size. However, these results stand in contrast to the claims by
Gautret et al. that all six patients treated with HCQ plus AZ tested negative for SARS-
CoV-2 RNA by day 6 posttreatment. This case study illustrated the need for further
investigation using robust study design to evaluate the efcacy of HCQ and/or CQ.
On 10 April 2020, the results of a randomized, nonplacebo trial of 62 COVID-19
patients at the Renmin Hospital of Wuhan University were released (332). This study
investigated whether HCQ decreased time to fever break or time to cough relief
compared to the SOC (332). This trial found HCQ decreased both average time to fever
break and average time to cough relief, dened as mild or no cough. While this study
improved on some of the methodological aws in the study of Gautret et al. (98) by
randomizing patients, it also had several aws in trial design and data analysis that
prevent generalization of the results. These weaknesses include the lack of placebo, lack
of correction for multiple primary outcomes, inappropriate choice of outcomes, lack of
Rando et al.
November/December 2021 Volume 6 Issue 6 e00233-21 msystems.asm.org 28
Downloaded from https://journals.asm.org/journal/msystems on 03 November 2021 by 2607:f470:34:2303:5040:4a48:5de1:cdb5.
sufcient detail to understand analysis, drastic disparities between preregistration (333)
and published protocol (including differences in the inclusion and exclusion criteria,
number of experimental groups, number of patients enrolled, and outcome analyzed),
and small sample size. The choice of outcomes may be inappropriate as both fevers and
cough may break periodically without resolution of illness. Additionally, for these
outcomes, the authors reported that 23 of 62 patients did not have a fever and 25 of 62
patients did not have a cough at the start of the study, but the authors failed to describe
how these patients were included in a study assessing time to fever break and time to
cough relief. It is important to note here that the authors claimed neither the research
performers nor the patients were aware of the treatment assignments.This blinding
seems impossible in a nonplacebo trial because at the very least, providers would know
whether they were administering a medication or not, and this knowledge could lead to
systematic differences in the administration of care. Correction for multiple primary
outcomes can be adjusted post hoc by recalculating Pvalues, but all of the other issues
were design and statistical weaknesses that cannot be corrected for. Additionally,
disparities between the preregistered and published protocols raise concerns about
experimental design. The design limitations mean that the conclusions cannot be
generalized outside the study.
A second randomized trial, conducted by the Shanghai Public Health Clinical Center,
analyzed whether HCQ increased rates of virological clearance at day 7 in respiratory
pharyngeal swabs compared to the SOC (334). This trial was published in Chinese along
with an abstract in English, and only the English abstract was read and interpreted for this
review. The trial found comparable outcomes in virological clearance rate, time to
virological clearance, and time to body temperature normalization between the
treatment and control groups. The small sample size is one weakness, with only 30
patients enrolled and 15 in each arm. This problem suggests the study is underpowered
to detect potentially useful differences and precludes interpretation of results.
Additionally, because only the abstract could be read, other design and analysis issues
could be present. Thus, though these studies added randomization to their assessment of
HCQ, their conclusions should be interpreted very cautiously. These two studies assessed
different outcomes and reached differing conclusions about the efcacy of HCQ for
treating COVID-19; the designs of both studies, especially with respect to sample size,
meant that no general conclusions can be made about the efcacy of the drug.
Several widely reported studies on HCQ also have issues with data integrity and/or
provenance. A Letter to the Editor published in BioScience Trends on 16 March 2020
claimed that numerous clinical trials have shown that HCQ is superior to control
treatment in inhibiting the exacerbation of COVID-19 pneumonia (335). This letter has
been cited by numerous papers in the primary literature, review articles, and media
alike (336, 337). However, the letter referred to 15 preregistration identiers from the
Chinese Clinical Trial Registry. When these identiers are followed back to the registry,
most trials claim they are not yet recruiting patients or are currently recruiting patients.
For all of these 15 identiers, no data uploads or links to publications could be located
on the preregistrations. At the very least, the lack of availability of the primary data
means the claim that HCQ is efcacious against COVID-19 pneumonia cannot be
veried. Similarly, a recent multinational registry analysis (338) analyzed the efcacy of
CQ and HCQ with and without a macrolide, which is a class of antibiotics that includes
azithromycin, for the treatment of COVID-19. The study observed 96,032 patients split
into a control and four treatment conditions (CQ with and without a macrolide and
HCQ with and without a macrolide). They concluded that treatment with CQ or HCQ was
associated with increased risk of de novo ventricular arrhythmia during hospitalization.
However, this study has since been retracted by The Lancet due to an inability to validate
the data used (339). These studies demonstrate that increased skepticism in evaluation of
the HCQ/CQ and COVID-19 literature may be warranted, possibly because of the
signicant attention HCQ and CQ have received as possible treatments for COVID-19 and
the politicization of these drugs.
ID and Development of Therapeutics for COVID-19
November/December 2021 Volume 6 Issue 6 e00233-21 msystems.asm.org 29
Downloaded from https://journals.asm.org/journal/msystems on 03 November 2021 by 2607:f470:34:2303:5040:4a48:5de1:cdb5.
Despite the fact that the study suggesting that CQ/HCQ increased risk of ventricular
arrhythmia in COVID-19 patients has now been retracted, previous studies have
identied risks associated with HCQ/CQ. A patient with systemic lupus erythematosus
developed a prolonged QT interval that was likely exacerbated by use of HCQ in
combination with renal failure (340). A prolonged QT interval is associated with
ventricular arrhythmia (341). Furthermore, a separate study (342) investigated the
safety associated with the use of HCQ with and without macrolides between 2000 and
2020. The study involved 900,000 cases treated with HCQ and 300,000 cases treated
with HCQ plus AZ. The results indicated that short-term use of HCQ was not associated
with additional risk but that HCQ plus AZ was associated with an enhanced risk of
cardiovascular complications (such as a 15% increased risk of chest pain, calibrated
HR of 1.15 and 95% CI of 1.05 to 1.26) and a twofold increased 30-day risk of
cardiovascular mortality (calibrated HR of 2.19; 95% CI, 1.22 to 3.94). Therefore, whether
studies utilize HCQ alone or HCQ in combination with a macrolide may be an important
consideration in assessing risk. As results from initial investigations of these drug
combinations have emerged, concerns about the efcacy and risks of treating COVID-
19 with HCQ and CQ have led to the removal of CQ/HCQ from the SOC practices in
several countries (343, 344). As of 25 May 2020, WHO had suspended administration of
HCQ as part of the worldwide Solidarity Trial (345), and later, the nal results of this
large-scale trial that compared 947 patients administered HCQ to 906 controls revealed
no effect on the primary outcome, mortality during hospitalization (rate ratio, 1.19;
P= 0.23).
Additional research has emerged largely identifying HCQ/CQ to be ineffective
against COVID-19 while simultaneously revealing a number of signicant side effects. A
randomized, open-label, nonplacebo trial of 150 COVID-19 patients was conducted in
parallel at 16 government-designated COVID-19 centers in China to assess the safety
and efcacy of HCQ (346). The trial compared treatment with HCQ in conjunction with
the SOC to the SOC alone in 150 infected patients who were assigned randomly to the
two groups (75 per group). The primary endpoint of the study was the negative
conversion rate of SARS-CoV-2 in 28 days, and the investigators found no difference in
this parameter between the groups (estimated difference between SOC plus HCQ and
SOC, 4.1%; 95% CI, 10.3% to 18.5%). The secondary endpoints were an amelioration of
the symptoms of the disease such as axillary temperature #36.6°C, pulse oximeter
blood oxygen saturation reading (SpO
2
)of.94% on room air, and disappearance of
symptoms like shortness of breath, cough, and sore throat. The median time to
symptom alleviation was similar across different conditions (19 days on HCQ plus SOC
versus 21 days on SOC; P= 0.97). Additionally, 30% of the patients receiving the SOC
plus HCQ reported adverse outcomes compared to 8.8% of patients receiving only the
SOC, with the most common adverse outcome in the SOC-plus-HCQ group being
diarrhea (10% versus 0% in the SOC group; P= 0.004). However, there are several
factors that limit the interpretability of this study. Most of the enrolled patients had
mild to moderate symptoms (98%), and the average age was 46 years. The SOC in this
study included the use of antivirals (lopinavir-ritonavir, arbidol, oseltamivir, virazole,
entecavir, ganciclovir, and interferon alfa), which the authors note could inuence the
results. Thus, they note that an ideal SOC would need to exclude the use of antivirals,
but that ceasing antiviral treatment raised ethical concerns at the time that the study
was conducted. In this trial, the samples used to test for the presence of the SARS-CoV-2
virus were collected from the upper respiratory tract, and the authors indicated that the
use of upper respiratory samples may have introduced false-negative results (e.g.,
reference 347). Another limitation of the study that the authors acknowledge was that
the HCQ treatment began, on average, at a 16-day delay from the symptom onset. The
fact that this study was open label and lacked a placebo limits interpretation, and
additional analysis is required to determine whether HCQ reduces inammatory
response. Therefore, despite some potential areas of investigation identied in post hoc
analysis, this study cannot be interpreted as providing support for HCQ as a therapeutic
Rando et al.
November/December 2021 Volume 6 Issue 6 e00233-21 msystems.asm.org 30
Downloaded from https://journals.asm.org/journal/msystems on 03 November 2021 by 2607:f470:34:2303:5040:4a48:5de1:cdb5.
against COVID-19. This study provided no support for HCQ against COVID-19, as there
was no difference between the two groups in either negative seroconversion at 28 days
or symptom alleviation, and in fact, more severe adverse outcomes were reported in
the group receiving HCQ.
Additional evidence comes from a retrospective analysis (348) that examined data
from 368 COVID-19 patients across all U.S. Veteran Health Administration medical
centers. The study retrospectively investigated the effect of the administration of HCQ
(n= 97), HCQ plus AZ (n= 113), and no HCQ (n= 158) on 368 patients. The primary
outcomes assessed were death and the need for mechanical ventilation. Standard
supportive care was rendered to all patients. Due to the low representation of women
(n= 17) in the available data, the analysis included only men, and the median age was
65 years. The rate of death was 27.8% in the HCQ-only treatment group, 22.1% in the
HCQ-plus-AZ treatment group, and 14.1% in the no-HCQ group. These data indicated a
statistically signicant elevation in the risk of death for the HCQ-only group compared
to the no-HCQ group (adjusted HR, 2.61; P= 0.03), but not for the HCQ-plus-AZ group
compared to the no-HCQ group (adjusted HR, 1.14; P= 0.72). Further, the risk of
ventilation was similar across all three groups (adjusted HR of 1.43 [P= 0.48] [HCQ] and
0.43 [P= 0.09] [HCQ plus AZ] compared to no HCQ). The study thus showed evidence of
an association between increased mortality and HCQ in this cohort of COVID-19
patients but no change in rates of mechanical ventilation among the treatment
conditions. The study had a few limitations: it was not randomized, and the baseline
vital signs, laboratory tests, and prescription drug use were signicantly different
among the three groups. All of these factors could potentially inuence treatment
outcome. Furthermore, the authors acknowledge that the effect of the drugs might be
different in females and pediatric subjects, since these subjects were not part of the
study. The reported result that HCQ plus AZ is safer than HCQ contradicts the ndings
of the previous large-scale analysis of 20 years of records that found HCQ plus AZ to be
more frequently associated with cardiac arrhythmia than HCQ alone (342); whether this
discrepancy is caused by the pathology of COVID-19, is inuenced by age or sex, or is a
statistical artifact is not presently known.
Finally, ndings from the RECOVERY trial were released on 8 October 2020. This
study used a randomized, open-label design to study the effects of HCQ compared to
the SOC in 11,197 patients at 176 hospitals in the United Kingdom (100). Patients were
randomized into either the control group or one of the treatment arms, with twice as
many patients enrolled in the control group as any treatment group. Of the patients
eligible to receive HCQ, 1,561 were randomized into the HCQ arm, and 3,155 were
randomized into the control arm. The demographics of the HCQ and control groups
were similar in terms of average age (65 years), proportion female (approximately 38%),
ethnic make-up (73% versus 76% white), and prevalence of preexisting conditions (56%
versus 57% overall). In the HCQ arm of the study, patients received 800 mg at baseline
and again after 6 h and then 400 mg at 12 h and every subsequent 12 h. The primary
outcome analyzed was all-cause mortality, and patient vital statistics were reported by
physicians upon discharge or death, or else at 28 days following HCQ administration if
they remained hospitalized. The secondary outcome assessed was the combined risk of
progression to invasive mechanical ventilation or death within 28 days. By the advice of
an external data monitoring committee, the HCQ arm of the study was reviewed early,
leading to it being closed due a lack of support for HCQ as a treatment for COVID-19.
COVID-19-related mortality was not affected by HCQ in the RECOVERY trial (rate ratio,
1.09; 95% CI, 0.97 to 1.23; P= 0.15), but cardiac events were increased in the HCQ arm
(0.4 percentage points), as was the duration of hospitalization (rate ratio for discharge
alive within 28 days, 0.90; 95% CI, 0.83 to 0.98) and likelihood of progression to
mechanical ventilation or death (risk ratio, 1.14; 95% CI, 1.03 to 1.27). This large-scale
study thus builds upon studies in the United States and China to suggest that HCQ is
not an effective treatment, and in fact may negatively impact COVID-19 patients due to
its side effects. Therefore, though none of the studies have been conducted in a blind
ID and Development of Therapeutics for COVID-19
November/December 2021 Volume 6 Issue 6 e00233-21 msystems.asm.org 31
Downloaded from https://journals.asm.org/journal/msystems on 03 November 2021 by 2607:f470:34:2303:5040:4a48:5de1:cdb5.
manner, examining them together makes it clear that the available evidence points to
signicant dangers associated with the administration of HCQ to hospitalized COVID-19
patients, without providing any support for its efcacy.
(ii) HCQ for the treatment of mild cases. One additional possible therapeutic
application of HCQ considered was the treatment of mild COVID-19 cases in otherwise
healthy individuals. This possibility was assessed in a randomized, open-label, multicenter
analysis conducted in Catalonia (Spain) (349). This analysis enrolled adults 18 and older
who had been experiencing mild symptoms of COVID-19 for fewer than 5 days.
Participants were randomized into an HCQ arm (n=136)andacontrolarm(n= 157), and
those in the treatment arm were administered 800 mg of HCQ on the rst day of
treatment followed by 400 mg on each of the subsequent 6 days. The primary outcome
assessed was viral clearance at days 3 and 7 following the onset of treatment, and
secondary outcomes were clinical progression and time to complete resolution of
symptoms. No signicant differences between the two groups were found: the difference
in viral load between the HCQ and control groups was 0.01 (95% CI, 20.28 to 0.29) at day
3and20.07 (95% CI, 20.44 to 0.29) at day 7, the relative risk of hospitalization was 0.75
(95% CI, 0.32 to 1.77), and the difference in time to complete resolution of symptoms was
22days(P= 0.38). This study thus suggests that HCQ does not improve recovery from
COVID-19, even in otherwise healthy adult patients with mild symptoms.
(iii) Prophylactic administration of HCQ. An initial study of the possible
prophylactic application of HCQ utilized a randomized, double-blind, placebo-controlled
design to analyze the administration of HCQ prophylactically (350). Asymptomatic adults
in the United States and Canada who had been exposed to SARS-CoV-2 within the past 4
days were enrolled in an online study to evaluate whether administration of HCQ over 5
days inuenced the probability of developing COVID-19 symptoms over a 14-day period.
Of the participants, 414 received HCQ and 407 received a placebo. No signicant
difference in the rate of symptomatic illness was observed between the two groups
(11.8% for HCQ, 14.3% for placebo; P= 0.35). The HCQ condition was associated withside
effects, with 40.1% of patients reporting side effects compared to 16.8% in the control
group (P,0.001). However, likely due to the high enrollment of health care workers
(66% of participants) and the well-known side effects associated with HCQ, a large
number of participants were able to correctly identify whether they were receiving HCQ
or a placebo (46.5% and 35.7%, respectively). Furthermore, due to a lack of availability of
diagnostic testing, only 20 of the 107 cases were conrmed with a PCR-based test to be
positive for SARS-CoV-2. The rest were categorized as probableor possiblecases by a
panel of four physicians who were blind to the treatment status. One possible
confounding factor is that a patient presenting one or more symptoms, which included
diarrhea, was dened as a possiblecase, but diarrhea is also a common side effect of
HCQ. Additionally, 4 of the 20 PCR-conrmed cases did not develop symptoms until after
the observation period had completed, suggesting that the 14-day trial period may not
have been long enough or that some participants also encountered secondary exposure
events. Finally, in addition to the young age of the participants in this study, which
ranged from 32 to 51 years, there were possible impediments to generalization
introduced by the selection process, as 2,237 patients who were eligible but had already
developed symptoms by day 4 were enrolled in a separate study. It is therefore likely that
asymptomatic cases were overrepresented in this sample, which would not have been
detected based on the diagnostic criteria used. Therefore, while this study does represent
the rst effort to conduct a randomized, double-blind, placebo-controlled investigation
of HCQs effect on COVID-19 prevention after SARS-CoV-2 exposure in a large sample,
the lack of PCR tests and several other design aws signicantly impede interpretation of
the results. However, in line with the results from therapeutic studies, once again no
evidence was found suggesting an effect of HCQ against COVID-19.
A second study (351) examined the effect of administering HCQ to health care
workers as a preexposure prophylactic. The primary outcome assessed was the
conversion from SARS-CoV-2-negative to SARS-CoV-2-positive status over the 8-week
study period. This study was also a randomized, double-blind, and placebo-controlled
Rando et al.
November/December 2021 Volume 6 Issue 6 e00233-21 msystems.asm.org 32
Downloaded from https://journals.asm.org/journal/msystems on 03 November 2021 by 2607:f470:34:2303:5040:4a48:5de1:cdb5.
study, and it sought to address some of the limitations of the rst prophylactic study.
The goal was to enroll 200 health care workers, preferentially those working with
COVID-19 patients, at two hospitals within the University of Pennsylvania hospital
system in Philadelphia, PA. Participants were randomized 1:1 to receive either 600 mg
of HCQ daily or a placebo, and their SARS-CoV-2 infection status and antibody status
were assessed using RT-PCR and serological testing, respectively, at baseline, 4 weeks,
and 8 weeks following the beginning of the treatment period. The statistical design of
the study accounted for interim analyses at 50 and 100 participants in case efcacy or
futility of HCQ for prophylaxis became clear earlier than completion of enrollment. The
139 individuals enrolled comprised a study population that was fairly young (average
age, 33 years) and made up largely of people who were white, women, and without
preexisting conditions. At the second interim analysis, more individuals in the
treatment group than the control group had contracted COVID-19 (four versus three),
causing the estimated z-score to fall below the preestablished threshold for futility. As a
result, the trial was terminated early, offering additional evidence against the use of
HCQ for prophylaxis.
(iv) Summary of HCQ/CQ research ndings. Early in vitro evidence indicated that
HCQ could be an effective therapeutic against SARS-CoV-2 and COVID-19, leading to
signicant media attention and public interest in its potential as both a therapeutic and
prophylactic. Initially, it was hypothesized that CQ/HCQ might be effective against
SARS-CoV-2 in part because CQ and HCQ have both been found to inhibit the
expression of CD154 in T cells and to reduce TLR signaling that leads to the production
of proinammatory cytokines (352). Clinical trials for COVID-19 have more often used
HCQ rather than CQ because it offers the advantages of being cheaper and having
fewer side effects than CQ. However, research has not found support for a positive
effect of HCQ on COVID-19 patients. Multiple clinical studies have already been carried
out to assess HCQ as a therapeutic agent for COVID-19, and many more are in progress.
To date, none of these studies have used randomized, double-blind, placebo-controlled
designs with a large sample size, which would be the gold standard. Despite the design
limitations (which would be more likely to produce false-positive results than false-
negative results), initial optimism about HCQ has largely dissipated. The most
methodologically rigorous analysis of HCQ as a prophylactic (350) found no signicant
differences between the treatment and control groups, and the WHOs global Solidarity
trial similarly reported no effect of HCQ on mortality (93). Thus, HCQ/CQ are not likely to
be effective therapeutic or prophylactic agents against COVID-19. One case study
identied drug-induced phospholipidosis as the cause of death for a COVID-19 patient
treated with HCQ (272), suggesting that in some cases, the proposed mechanism of
action may ultimately be harmful. Additionally, one study identied an increased risk of
mortality in older men receiving HCQ, and administration of HCQ and HCQ plus AZ did
not decrease the use of mechanical ventilation in these patients (348). HCQ use for
COVID-19 could also lead to shortages for antimalarial or antirheumatic use, where it
has documented efcacy. Despite signicant early attention, these drugs appear to be
ineffective against COVID-19. Several countries have now removed CQ/HCQ from their
SOC for COVID-19 due to the lack of evidence of efcacy and the frequency of adverse
effects.
ACE inhibitors and angiotensin II receptor blockers. Several clinical trials testing
the effects of ACEIs or ARBs on COVID-19 outcomes are ongoing (353359). Clinical
trials are needed because the ndings of the various observational studies bearing on
this topic cannot be interpreted as indicating a protective effect of the drug (360, 361).
Two analyses (353, 359) have reported no effect of continuing or discontinuing ARBs
and ACEIs on patients admitted to the hospital for COVID-19. The rst, known as
REPLACE COVID (156), was a randomized, open-label study that enrolled patients who
were admitted to the hospital for COVID-19 and were taking an ACEI at the time of
admission. They enrolled 152 patients at 20 hospitals in seven countries and
randomized them into two arms, continuation (n= 75) and discontinuation (n= 77).
The primary outcome evaluated was a global rank score that integrated several
ID and Development of Therapeutics for COVID-19
November/December 2021 Volume 6 Issue 6 e00233-21 msystems.asm.org 33
Downloaded from https://journals.asm.org/journal/msystems on 03 November 2021 by 2607:f470:34:2303:5040:4a48:5de1:cdb5.
dimensions of illness. The components of this global rank score, such as time to death
and length of mechanical ventilation, were evaluated as secondary endpoints. This
study reported no differences between the two groups in the primary outcome or any
of the secondary outcomes.
Similarly, a second study (157) used a randomized, open-label design to examine the
effects of continuing versus discontinuing ARBs and ACEIs on patients hospitalized for
mild to moderate COVID-19 at 29 hospitals in Brazil. This study enrolled 740 patients
but had to exclude one trial site from all analyses due to the discovery of violations of
Good Clinical Trial practice and data falsication. After this exclusion, 659 patients
remained, with 334 randomized to discontinuation and 325 to continuation. In this
study, the primary endpoint analyzed was the number of days that patients were alive
and not hospitalized within 30 days of enrollment. The secondary outcomes included
death (including in-hospital death separately), number of days hospitalized, and
specic clinical outcomes such as heart failure or stroke. Once again, no signicant
differences were found between the two groups. Initial studies of randomized
interventions therefore suggest that ACEIs and ARBs are unlikely to affect COVID-19
outcomes. These results are also consistent with ndings from observational studies
(summarized in reference 156). Additional information about ACE2, observational studies
of ACEIs and ARBs in COVID-19, and clinical trials on this topic have been summarized
(362). Therefore, despite the promising potential mechanism, initial results have not
provided support for ACEIs and ARBs as therapies for COVID-19.
Tocilizumab. Human IL-6 is a 26-kDa glycoprotein that consists of 184 amino acids
and contains two potential N-glycosylation sites and four cysteine residues. It binds to a
type I cytokine receptor (IL-6 receptor alpha [IL-6R
a
] or glycoprotein 80 [gp80]) that
exists in both membrane-bound (IL-6R
a
) and soluble (sIL-6R
a
) forms (363). It is not the
binding of IL-6 to the receptor that initiates pro- and/or anti-inammatory signaling,
but rather the binding of the complex to another subunit, known as IL-6R
b
or gp130
(363, 364). Unlike membrane-bound IL-6R
a
, which is found only on hepatocytes and
some types of leukocytes, gp130 is found on most cells (365). When IL-6 binds to sIL-
6R
a
, the complex can then bind to a gp130 protein on any cell (365). The binding of IL-
6 to IL-6R
a
is termed classical signaling, while its binding to sIL-6R
a
is termed trans-
signaling (365367). These two signaling processes are thought to play different roles in
health and illness. For example, trans-signaling may play a role in the proliferation of
mucosal T-helper TH2 cells associated with asthma, while an earlier step in this
proliferation process may be regulated by classical signaling (365). Similarly, IL-6 is
known to play a role in Crohns disease via trans-signaling, but not classical signaling
(365). Both classical and trans-signaling can occur through three independent
pathways: the Janus-activated kinase-STAT3 pathway, the Ras/mitogen-activated
protein kinase pathway and the phosphoinositol-3 kinase/Akt pathway (363). These
signaling pathways are involved in a variety of different functions, including cell type
differentiation, immunoglobulin synthesis, and cellular survival signaling pathways,
respectively (363). The ultimate result of the IL-6 cascade is to direct transcriptional
activity of various promoters of proinammatory cytokines, such as IL-1, TFN, and even
IL-6 itself, through the activity of NF-
k
B (363). IL-6 synthesis is tightly regulated both
transcriptionally and posttranscriptionally, and it has been shown that viral proteins can
enhance transcription of the IL-6 gene by strengthening the DNA-binding activity
between several transcription factors and IL-6 gene cis-regulatory elements (368).
Therefore, drugs inhibiting the binding of IL-6 to IL-6R
a
or sIL-6R
a
are of interest for
combating the hyperactive inammatory response characteristic of cytokine release
syndrome (CRS) and cytokine storm syndrome (CSS). TCZ is a humanized monoclonal
antibody that binds both to the insoluble and soluble receptor of IL-6, providing de
facto inhibition of the IL-6 immune cascade. Interest in TCZ as a possible treatment for
COVID-19 was piqued by early evidence indicating that COVID-19 deaths may be
induced by the hyperactive immune response, often referred to as CRS or CSS (170), as
IL-6 plays a key role in this response (369). The observation of elevated IL-6 in patients
Rando et al.
November/December 2021 Volume 6 Issue 6 e00233-21 msystems.asm.org 34
Downloaded from https://journals.asm.org/journal/msystems on 03 November 2021 by 2607:f470:34:2303:5040:4a48:5de1:cdb5.
who died relative to those who recovered (170) could reect an overproduction of
proinammatory interleukins, suggesting that TCZ could potentially palliate some of
the most severe symptoms of COVID-19 associated with increased cytokine production.
This early interest in TCZ as a possible treatment for COVID-19 was bolstered by a
very small retrospective study in China that examined 20 patients with severe
symptoms in early February 2020 and reported rapid improvement in symptoms
following treatment with TCZ (176). Subsequently, a number of retrospective studies
have been conducted in several countries. Many studies use a retrospective,
observational design, where they compare outcomes for COVID-19 patients who
received TCZ to those who did not over a set period of time. For example, one of the
largest retrospective, observational analyses released thus far (171), consisting of 1,351
patients admitted to several care centers in Italy, compared the rates at which patients
who received TCZ died or progressed to invasive medical ventilation over a 14-day
period compared to patients receiving only the SOC. Under this denition, the SOC
could include other drugs such as HCQ, azithromycin, lopinavir-ritonavir, darunavir-
cobicistat, or heparin. While this study was not randomized, a subset of patients who
were eligible to receive TCZ were unable to obtain it due to shortages; however, these
groups were not directly compared in the analysis. After adjusting for variables such as
age, sex, and SOFA (sequential organ failure assessment) score, they found that patients
treated with TCZ were less likely to progress to invasive medical ventilation and/or
death (adjusted HR = 0.61, 95% CI 0.40 to 0.92, P= 0.020); the results of analysis of
death and ventilation separately suggest that this effect may have been driven by
differences in the death rate (20% of control versus 7% of TCZ-treated patients). The
study reported particular benets for patients whose PaO
2
/FiO
2
ratio, also known as the
Horowitz index for lung function, fell below a 150-mm Hg threshold. They found no
differences between groups administered subcutaneous versus intravenous TCZ.
Another retrospective observational analysis of interest examined the charts of
patients at a hospital in Connecticut, United States, where 64% of all 239 COVID-19
patients in the study period were administered TCZ based on assignment by a
standardized algorithm (172). They found that TCZ administration was associated with
more similar rates of survivorship in patients with severe versus nonsevere COVID-19 at
intake, dened based on the amount of supplemental oxygen needed. They therefore
proposed that their algorithm was able to identify patients presenting with or likely to
develop CRS as good candidates for TCZ. This study also reported higher survivorship in
Black and Hispanic patients compared to white patients when adjusted for age. The
major limitation with interpretation for these studies is that there may be clinical
characteristics that inuenced medical practitionersdecisions to administer TCZ to
some patients and not others. One interesting example therefore comes from an
analysis of patients at a single hospital in Brescia, Italy, where TCZ was not available for
a period of time (173). This study compared COVID-19 patients admitted to the hospital
before and after 13 March 2020, when the hospital received TCZ. Therefore, patients
who would have been eligible for TCZ prior to this arbitrary date did not receive it as
treatment, making this retrospective analysis something of a natural experiment.
Despite this design, demographic factors did not appear to be consistent between the
two groups, and the average age of the control group was older than the average age
of the TCZ group. The control group also had a higher percentage of males and a
higher incidence of comorbidities such as diabetes and heart disease. All the same, the
multivariate HR, which adjusted for these clinical and demographic factors, found a
signicant difference between survival in the two groups (HR = 0.035, 95% CI = 0.004
to 0.347, P= 0.004). The study reported improvement of survival outcomes after the
addition of TCZ to the SOC regime, with 11 of 23 patients (47.8%) admitted prior to
March 13th dying compared to 2 of 62 (3.2%) admitted afterwards (HR = 0.035, 95% CI,
0.004 to 0.347, P= 0.004). They also reported a reduced progression to mechanical
ventilation in the TCZ group. However, this study also holds a signicant limitation: the
time delay between the two groups means that knowledge about how to treat the
ID and Development of Therapeutics for COVID-19
November/December 2021 Volume 6 Issue 6 e00233-21 msystems.asm.org 35
Downloaded from https://journals.asm.org/journal/msystems on 03 November 2021 by 2607:f470:34:2303:5040:4a48:5de1:cdb5.
disease likely improved over this time frame as well. All the same, the results of these
observational retrospective studies provide support for TCZ as a pharmaceutical of
interest for follow-up in clinical trials.
Other retrospective analyses have utilized a case-control design to match pairs of
patients with similar baseline characteristics, only one of whom received TCZ for
COVID-19. In one such study, TCZ was signicantly associated with a reduced risk of
progression to intensive care unit (ICU) admission or death (174). This study examined
only 20 patients treated with TCZ (all but one of the patients treated with TCZ in the
hospital during the study period) and compared them to 25 patients receiving the SOC.
For the combined primary endpoint of death and/or ICU admission, only 25% of
patients receiving TCZ progressed to an endpoint compared to 72% in the SOC group
(P= 0.002, presumably based on a chi-square test based on the information provided in
the text). When the two endpoints were examined separately, progression to invasive
medical ventilation remained signicant (32% SOC compared to 0% TCZ; P= 0.006) but
not for mortality (48% SOC compared to 25% TCZ; P= 0.066). In contrast, a study that
compared 96 patients treated with TCZ to 97 patients treated with the SOC only in New
York City found that differences in mortality did not differ between the two groups but
that this difference did become signicant when intubated patients were excluded
from the analysis (175). Taken together, these ndings suggest that future clinical trials
of TCZ may want to include intubation as an endpoint. However, these studies should
be approached with caution, not only because of the small number of patients enrolled
and the retrospective design but also because they performed a large number of
statistical tests and did not account for multiple hypothesis testing. In general, caution
must be exercised when interpreting subgroup analyses after a primary combined
endpoint analysis. These last ndings highlight the need to search for a balance
between impairing a harmful immune response, such as the one generated during CRS/
CSS, and preventing the worsening of the clinical picture of the patients by potential
new viral infections. Early meta-analyses and systematic reviews have investigated the
available data about TCZ for COVID-19. One meta-analysis (370) evaluated 19 studies
published or released as preprints prior to 1 July 2020 and found that the overall trends
were supportive of the frequent conclusion that TCZ does improve survivorship, with a
signicant HR of 0.41 (P,0.001). This trend improved when they excluded studies that
administered a steroid alongside TCZ, with a signicant HR of 0.04 (P,0.001). They
also found some evidence for reduced invasive ventilation or ICU admission, but only
when excluding all studies except a small number whose estimates were adjusted for
the possible bias introduced by the challenges of stringency during the enrollment
process. A systematic analysis of 16 case-control studies of TCZ estimated an odds ratio
of mortality of 0.453 (95% CI, 0.376 to 0.547;, P,0.001), suggesting possible benets
associated with TCZ treatment (371). Although these estimates are similar, it is
important to note that they are drawing from the same literature and are therefore
likely to be affected by the same potential biases in publication. A different systematic
review of studies investigating TCZ treatment for COVID-19 analyzed 31 studies that
had been published or released as preprints and reported that none carried a low risk of
bias (372). Therefore, the present evidence is not likely to be sufcient for conclusions
about the efcacy of TCZ.
On 11 February 2021, a preprint describing the rst randomized control trial of TCZ
was released as part of the RECOVERY trial (177). Of the 21,550 patients enrolled in the
RECOVERY trial at the time, 4,116 adults hospitalized with COVID-19 across the 131 sites
in the United Kingdom were assigned to the arm of the trial evaluating the effect of
TCZ. Of these patients, 2,022 were randomized to receive TCZ and 2,094 were
randomized to receive the SOC, with 79% of patients in each group available for
analysis at the time that the initial report was released. The primary outcome measured
was 28-day mortality, and TCZ was found to reduce 28-day mortality from 33% of
patients receiving the SOC alone to 29% of those receiving TCZ, corresponding to a rate
ratio of 0.86 (95% CI, 0.77 to 0.96; P= 0.007). TCZ was also signicantly associated with
Rando et al.
November/December 2021 Volume 6 Issue 6 e00233-21 msystems.asm.org 36
Downloaded from https://journals.asm.org/journal/msystems on 03 November 2021 by 2607:f470:34:2303:5040:4a48:5de1:cdb5.
the probability of hospital discharge within 28 days for living patients, which was 47%
in the SOC group and 54% in the TCZ group (rate ratio, 1.22; 95% CI, 1.12 to 1.34; P,
0.0001). A potential statistical interaction between TCZ and corticosteroids was
observed, with the combination providing greater mortality benets than TCZ alone,
but the authors note that caution is advisable in light of the number of statistical tests
conducted. Combining the RECOVERY trial data with data from seven smaller
randomized control trials indicates that TCZ is associated with a 13% reduction in 28-
day mortality (rate ratio, 0.87; 95% CI, 0.79 to 0.96; P= 0.005) (177).
There are possible risks associated with the administration of TCZ for COVID-19. TCZ
has been used for over a decade to treat rheumatoid arthritis (RA) (373), and a recent
study found the drug to be safe for pregnant and breastfeeding women (374).
However, TCZ may increase the risk of developing infections (373), and RA patients with
chronic hepatitis B infections had a high risk of hepatitis B virus reactivation when TCZ
was administered in combination with other RA drugs (375). As a result, TCZ is
contraindicated in patients with active infections such as tuberculosis (376). Previous
studies have investigated, with various results, a possible increased risk of infection in
RA patients administered TCZ (377, 378), although another study reported that the
incidence rate of infections was higher in clinical practice RA patients treated with TCZ
than in the rates reported by clinical trials (379). In the investigation of 544 Italian
COVID-19 patients, the group treated with TCZ was found to be more likely to develop
secondary infections, with 24% compared to 4% in the control group (P,0.0001) (171).
Reactivation of hepatitis B virus and herpes simplex virus 1 was also reported in a small
number of patients in this study, all of whom were receiving TCZ. A July 2020 case
report described negative outcomes of two COVID-19 patients after receiving TCZ,
including one death; however, both patients were intubated and had entered septic
shock prior to receiving TCZ (380), likely indicating a severe level of cytokine
production. Additionally, D-dimer and sIL2R levels were reported by one study to
increase in patients treated with TCZ, which raised concerns because of the potential
association between elevated D-dimer levels and thrombosis and between sIL2R and
diseases where T-cell regulation is compromised (172). An increased risk of bacterial
infection was also identied in a systematic review of the literature, based on the
unadjusted estimates reported (370). In the RECOVERY trial, however, only 3 out of
2,022 participants in the group receiving TCZ developed adverse reactions determined
to be associated with the intervention, and no excess deaths were reported (177). TCZ
administration to COVID-19 patients is not without risks and may introduce additional
risk of developing secondary infections; however, while caution may be prudent when
treating patients who have latent viral infections, the results of the RECOVERY trial
indicate that adverse reactions to TCZ are very rare among COVID-19 patients broadly.
In summary, approximately 33% of hospitalized COVID-19 patients develop ARDS
(381), which is caused by an excessive early response of the immune system which
can be a component of CRS/CSS (172, 376). This overwhelming inammation is
triggered by IL-6. TCZ is an inhibitor of IL-6 and therefore may neutralize the
inammatory pathway that leads to the cytokine storm. The mechanism suggests
TCZ could be benecial for the treatment of COVID-19 patients experiencing
excessive immune activity, and the RECOVERY trial reported a reduction in 28-day
mortality. Interest in TCZ as a treatment for COVID-19 was also supported by two
meta-analyses (370, 382), but a third meta-analysis found that all of the available
literature at that time carried a risk of bias (372). Additionally, different studies used
different dosages, number of doses, and methods of administration. Ongoing
research may be needed to optimize administration of TCZ (383), although similar
results were reported by one study for intravenous and subcutaneous administration
(171). Clinical trials that are in progress are likely to provide additional insight into
the effectiveness of this drug for the treatment of COVID-19 along with how it should
be administered.
ID and Development of Therapeutics for COVID-19
November/December 2021 Volume 6 Issue 6 e00233-21 msystems.asm.org 37
Downloaded from https://journals.asm.org/journal/msystems on 03 November 2021 by 2607:f470:34:2303:5040:4a48:5de1:cdb5.
Interferons. IFNs are a family of cytokines critical to activating the innate immune
response against viral infections. Interferons are classied into three categories based
on their receptor specicity: types I, II, and III (369). Specically, IFNs I (alpha interferon
[IFN-
a
] and beta interferon [IFN-
b
]) and II (gamma interferon [IFN-
g
]) induce the
expression of antiviral proteins (384). Among these IFNs, IFN-
b
has already been found
to strongly inhibit the replication of other coronaviruses, such as SARS-CoV-1, in cell
culture, while IFN-
a
and -
g
were shown to be less effective in this context (384). There is
evidence that patients with higher susceptibility to ARDS indeed show deciency in
IFN-
b
. For instance, infection with other coronaviruses impairs IFN-
b
expression and
synthesis, allowing the virus to escape the innate immune response (385). On 18 March
2020, Synairgen plc received approval to start a phase 2 trial for SNG001, an IFN-
b
-1a
formulation to be delivered to the lungs via inhalation (184). SNG001, which contains
recombinant interferon
b
-1a, was previously shown to be effective in reducing viral
load in an in vivo model of swine u and in vitro models of other coronavirus infections
(386). In July 2020, a press release from Synairgen stated that SNG001 reduced
progression to ventilation in a double-blind, placebo-controlled, multicenter study of
101 patients with an average age in the late 50s (185). These results were subsequently
published in November 2020 (186). The study reports that the participants were
assigned at a ratio of 1:1 to receive either SNG001 or a placebo that lacked the active
compound, by inhalation for up to 14 days. The primary outcome they assessed was the
change in patientsscore on the WHO Ordinal Scale for Clinical Improvement (OSCI) on
trial day 15 or 16. SNG001 was associated with an odds ratio of improvement on the
OSCI scale of 2.32 (95% CI, 1.07 to 5.04; P= 0.033) in the intention-to-treat analysis and
2.80 (95% CI, 1.21 to 6.52; P= 0.017) in the per-protocol analysis, corresponding to
signicant improvement in the SNG001 group on the OSCI on day 15 or 16. Some of the
secondary endpoints analyzed also showed differences: on day 28, the odds ratio (OR)
for clinical improvement on the OSCI was 3.15 (95% CI, 1.39 to 7.14; P= 0.006), and the
odds of recovery on day 15/16 and on day 28 were also signicant between the two
groups. Thus, this study suggested that IFN-
b
1 administered via SNG001 may improve
clinical outcomes.
In contrast, the WHO Solidarity trial reported no signicant effect of IFN-
b
-1a on
patient survival during hospitalization (93). Here, the primary outcome analyzed was in-
hospital mortality, and the rate ratio for the two groups was 1.16 (95% CI, 0.96 to 1.39; P=
0.11) administering IFN-
b
-1a to 2,050 patients and comparing their response to 2,050
controls. However, there are a few reasons that the different ndings of the two trials
might not speak to the underlying efcacy of this treatment strategy. One important
consideration is the stage of COVID-19 infection analyzed in each study. The Synairgen
trial enrolled only patients who were not receiving invasive ventilation, corresponding to
a less severe stage of disease than many patients enrolled in the Solidarity trial, as well as
a lower overall rate of mortality (387). Additionally, the method of administration differed
between the two trials, with the Solidarity trial administering IFN-
b
-1a subcutaneously
(387). The differences in ndings between the studies suggest that the method of
administration might be relevant to outcomes, with nebulized IFN-
b
-1a more directly
targeting receptors in the lungs. A trial that analyzed the effect of subcutaneously
administered IFN-
b
-1a on patients with ARDS between 2015 and 2017 had also reported
no effect on 28-day mortality (388), while a smaller study analyzing the effect of
subcutaneous IFN administration did nd a signicant improvement in 28-day mortality for
COVID-19 (389). At present, several ongoing clinical trials are investigating the potential
effects of IFN-
b
-1a, including in combination with therapeutics such as remdesivir (390)
and administered via inhalation (184). Thus, as additional information becomes available, a
more detailed understanding of whether and under which circumstances IFN-
b
-1a is
benecial to COVID-19 patients should develop.
Potential avenues of interest for therapeutic development. Given what is currently
known about these therapeutics for COVID-19, a number of related therapies beyond
those explored above may also prove to be of interest. For example, the demonstrated
Rando et al.
November/December 2021 Volume 6 Issue 6 e00233-21 msystems.asm.org 38
Downloaded from https://journals.asm.org/journal/msystems on 03 November 2021 by 2607:f470:34:2303:5040:4a48:5de1:cdb5.
benet of dexamethasone and the ongoing potential of tocilizumab for treatment of
COVID-19 suggest that other anti-inammatory agents might also hold value for the
treatment of COVID-19. Current evidence supporting the treatment of severe COVID-19
with dexamethasone suggests that the need to curtail the cytokine storm inammatory
response transcends the risks of immunosuppression, and other anti-inammatory
agents may therefore benet patients in this phase of the disease. While dexamethasone
is considered widely available and generally affordable, the high costs of biologics such
as tocilizumab therapy may present obstacles to wide-scale distribution of this drug if it
proves of value. At the doses used for RA patients, the cost for tocilizumab ranges from
$179.20 to $896 per dose for the intravenous (IV) form and $355 for the prelled syring
(391). Several other anti-inammatory agents used for the treatment of autoimmune
diseases may also be able to counter the effects of the cytokine storm induced by the
virus, and some of these, such as cyclosporine, are likely to be more cost-effective and
readily available than biologics (392). While tocilizumab targets IL-6, several other
inammatory markers could be potential targets, including TNF-
a
. Inhibition of TNF-
a
by
a compound such as etanercept was previously suggested for treatment of SARS-CoV-1
(393) and may be relevant for SARS-CoV-2 as well. Another anti-IL-6 antibody, sarilumab,
is also being investigated (394, 395). Baricitinib and other small-molecule inhibitors of the
Janus-activated kinase pathway also curtail the inammatory response and have been
suggested as potential options for SARS-CoV-2 infections (396). Baricitinib, in particular,
may be able to reduce the ability of SARS-CoV-2 to infect lung cells (397). Clinical trials
studying baricitinib in COVID-19 have already begun in the United States and in Italy
(398, 399). Identication and targeting of further inammatory markers that are relevant
in SARS-CoV-2 infection may be of value for curtailing the inammatory response and
lung damage.
In addition to immunosuppressive treatments, which are most benecial late in
disease progression, much research is focused on identifying therapeutics for early
stage patients. For example, although studies of HCQ have not supported the early
theory-driven interest in this antiviral treatment, alternative compounds with related
mechanisms may still have potential. Hydroxyferroquine derivatives of HCQ have been
described as a class of bioorganometallic compounds that exert antiviral effects with
some selectivity for SARS-CoV-1 in vitro (400). Future work could explore whether such
compounds exert antiviral effects against SARS-CoV-2 and whether they would be safer
for use in COVID-19.
Another potential approach is the development of antivirals, which could be broad
spectrum, specic to coronaviruses, or targeted to SARS-CoV-2. Development of new
antivirals is complicated by the fact that none have yet been approved for human
coronaviruses. Intriguing new options are emerging, however. Beta-D-N4-hydroxycytidine
is an orally bioavailable ribonucleotide analog showing broad-spectrum activity against
RNA viruses, which may inhibit SARS-CoV-2 replication in vitro and in vivo in mouse models
of HCoVs (401). A range of other antivirals are also in development. Development of
antivirals will be further facilitated as research reveals more information about the
interaction of SARS-CoV-2 with the host cell and host cell genome, mechanisms of viral
replication, mechanisms of viral assembly, and mechanisms of viral release to other cells;
this can allow researchers to target specic stages and structures of the viral life cycle.
Finally, antibodies against viruses, also known as antiviral monoclonal antibodies, could be
an alternative as well and are described in detail in an above section. The goal of antiviral
antibodies is to neutralize viruses through either cell-killing activity or blocking of viral
replication (402). They may also engage the host immune response, encouraging the
immune system to hone in on the virus. Given the cytokine storm that results from immune
system activation in response to the virus, which has been implicated in worsening of the
disease, a neutralizing antibody (nAb) may be preferable. Upcoming work may explore the
specicity of nAbs for their target, mechanisms by which the nAbs impede the virus, and
improvements to antibody structure that may enhance the ability of the antibody to block
viral activity.
ID and Development of Therapeutics for COVID-19
November/December 2021 Volume 6 Issue 6 e00233-21 msystems.asm.org 39
Downloaded from https://journals.asm.org/journal/msystems on 03 November 2021 by 2607:f470:34:2303:5040:4a48:5de1:cdb5.
Some research is also investigating potential therapeutics and prophylactics that
would interact with components of the innate immune response. For example, TLRs are
pattern recognition receptors that recognize pathogen- and damage-associated molecular
patterns and contribute to innate immune recognition and, more generally, promotion of both
the innate and adaptive immune responses (403). In mouse models, poly(IC) and CpG, which
are agonists of Toll-like receptors TLR3 and TLR9, respectively, showed protective effects when
administered prior to SARS-CoV-1 infection (404). Therefore, TLR agonists hold some
potential for broad-spectrum prophylaxis.
ACKNOWLEDGMENTS
James Brian Byrd was funded by FastGrants to conduct a COVID-19-related clinical
trial. Christian Brueffer is an employee and shareholder of SAGA Diagnostics AB. YoSon
Park is afliated with Pzer Worldwide Research. The author has no nancial interests to
declare and contributed as an author prior to joining Pzer, and the work was not part
of a Pzer collaboration nor was it funded by Pzer. Simina M. Boca is currently an
employee at AstraZeneca, Gaithersburg, MD, USA, and may own stock or stock options;
work was initially conducted at Georgetown University Medical Center, with writing,
reviewing, and editing continued while working at AstraZeneca. Anthony Gitter led a
patent application with the Wisconsin Alumni Research Foundation related to classifying
activated T cells. The other authors declare no conicts of interest.
We thank Nick DeVito for assistance with the Evidence-Based Medicine Data Lab
COVID-19 TrialsTracker data. We thank Yael Evelyn Marshall who contributed writing
(original draft) as well as reviewing and editing of pieces of the text but who did not
formally approve the manuscript, as well as Ronnie Russell, who contributed text to and
helped develop the structure of the manuscript early in the writing process. We are also
very grateful to James Fraser for suggestions about successes and limitations in the
area of computational screening for drug repurposing. We are grateful to the following
contributors for reviewing pieces of the text: Nadia Danilova, James Eberwine and Ipsita
Krishnan.
The members of the COVID-19 Review Consortium include the following: Vikas
Bansal, John P. Barton, Simina M. Boca, Joel D Boerckel, Christian Brueffer, James Brian
Byrd, Stephen Capone, Shikta Das, Anna Ada Dattoli, John J. Dziak, Jeffrey M. Field,
Soumita Ghosh, Anthony Gitter, Rishi Raj Goel, Casey S. Greene, Marouen Ben Guebila,
Daniel S. Himmelstein, Fengling Hu, Nasa M. Jadavji, Jeremy P. Kamil, Sergey Knyazev,
Likhitha Kolla, Alexandra J. Lee, Ronan Lordan, Tiago Lubiana, Temitayo Lukan, Adam L.
MacLean, David Mai, Serghei Mangul, David Manheim, Lucy DAgostino McGowan,
Amruta Naik, YoSon Park, Dimitri Perrin, Yanjun Qi, Diane N. Razadeh, Bharath
Ramsundar, Halie M. Rando, Sandipan Ray, Michael P. Robson, Vincent Rubinetti,
Elizabeth Sell, Lamonica Shinholster, Ashwin N. Skelly, Yuchen Sun, Yusha Sun, Gregory
L Szeto, Ryan Velazquez, Jinhui Wang, and Nils Wellhausen.
Authors with similar contributions are ordered alphabetically.
Halie M. Rando contributed to Project Administration, Software, Visualization, Writing -
Original Draft, and Writing - Review & Editing. Nils Wellhausen contributed to Project
Administration, Visualization, Writing - Original Draft, and Writing - Review & Editing.
Soumita Ghosh, Anna Ada Dattoli, and Marouen Ben Guebila contributed to Writing - Original
Draft. Alexandra J. Lee, Fengling Hu, James Brian Byrd, and Jeffrey M. Field contributed to
Writing - Original Draft and Writing - Review & Editing. Diane N. Razadeh and Ronan Lordan
contributed to Project Administration, Writing - Original Draft, and Writing - Review & Editing.
Yanjun Qi and Yuchen Sun contributed to Visualization. Christian Brueffer contributed to
Project Administration and Writing - Review & Editing. Nasa M. Jadavji contributed to
Supervision, Writing - Original Draft, and Writing - Review & Editing. Ashwin N. Skelly, Bharath
Ramsundar, Rishi Raj Goel, and YoSon Park contributed to Writing - Review & Editing. Jinhui
Wang contributed to Writing - Original Draft. The COVID-19 Review Consortium contributed to
Project Administration. Simina M. Boca and Casey S. Greene contributed to Project
Administration and Writing - Review & Editing. Anthony Gitter contributed to Project
Administration, Software, Visualization, and Writing - Review & Editing.
Rando et al.
November/December 2021 Volume 6 Issue 6 e00233-21 msystems.asm.org 40
Downloaded from https://journals.asm.org/journal/msystems on 03 November 2021 by 2607:f470:34:2303:5040:4a48:5de1:cdb5.
REFERENCES
1. Rando HM, MacLean AL, Lee AJ, Ray S, Bansal V, Skelly AN, Sell E,
Dziak JJ, Shinholster L, McGowan LD, ...Greene CS. 2021. Pathoge-
nesis, symptomatology, and transmission of SARS-CoV-2 through
analysis of viral genomics and structure. arXiv https://arxiv.org/abs/
2102.01521.
2. COVID-19 Review Consortium. 19 February 2021. SARS-CoV-2 and COVID-
19: an evolving review of diagnostics and therapeutics. https://greenelab
.github.io/covid19-review/v/d9d90fd7e88ef547fb4cbed0ef73baef5fee7fb5/
%23vaccine-development-strategies-for-sars-cov-2.
3. Hua J, Wang G, Huang M, Hua S, Yang S. 2020. A visual approach for the
SARS (severe acute respiratory syndrome) outbreak data analysis. Int J En-
viron Res PublicHealth 17:3973. https://doi.org/10.3390/ijerph17113973.
4. Center for Systems Science and Engineering at Johns Hopkins University.
COVID-19 Data Repository. GitHub https://github.com/CSSEGISandData/
COVID-19/tree/master/csse_covid_19_data/csse_covid_19_time_series.
5. Dong E, Du H, Gardner L. 2020. An interactive web-based dashboard to
track COVID-19 in real time. Lancet Infect Dis 20:5332534. https://doi
.org/10.1016/S1473-3099(20)30120-1.
6. World Health Organization. 2021. Severe acute respiratory syndrome (SARS).
World Health Organization, Geneva, Switzerland. https://www.who.int/
westernpacic/health-topics/severe-acute-respiratory-syndrome.
7. GitHub. imdevskp/sars-2003-outbreak-data-webscraping-code: repository
contains complete WHO data of 2003 outbreak with code used to web
scrap, data mung and cleaning. GitHub https://github.com/imdevskp/sars
-2003-outbreak-data-webscraping-code.
8. Guarner J. 2020. Three emerging coronaviruses in two decades. Am J
Clin Pathol 153:4202421. https://doi.org/10.1093/ajcp/aqaa029.
9. de Wit E, van Doremalen N, Falzarano D, Munster VJ. 2016. SARS and
MERS: recent insights into emerging coronaviruses. Nat Rev Microbiol
14:5232534. https://doi.org/10.1038/nrmicro.2016.81.
10. Tan W, Zhao X, Ma X, Wang W, Niu P, Xu W, Gao GF, Wu G. 21 January
2020. A novel coronavirus genome identied in a cluster of pneumonia
cases Wuhan, China 201922020. China CDC Wkly. https://doi.org/10
.46234/ccdcw2020.017.
11. Klompas M, Baker MA, Rhee C. 2020. Airborne transmission of SARS-CoV-
2: theoretical considerations and available evidence. JAMA https://doi
.org/10.1001/jama.2020.12458.
12. Goldman E. 2020. Exaggerated risk of transmission of COVID-19 by
fomites. Lancet Infect Dis 20:8922893. https://doi.org/10.1016/S1473
-3099(20)30561-2.
13. Greenhalgh T, Jimenez JL, Prather KA, Tufekci Z, Fisman D, Schooley R. 2021.
Ten scientic reasons in support of airborne transmission of SARS-CoV-2.
Lancet 397:160321605. https://doi.org/10.1016/S0140-6736(21)00869-2.
14. Tang JW, Marr LC, Li Y, Dancer SJ. 2021. Covid-19 has redened airborne
transmission. BMJ 373:n913. https://doi.org/10.1136/bmj.n913.
15. Lu R, Zhao X, Li J, Niu P, Yang B, Wu H, Wang W, Song H, Huang B, Zhu N,
Bi Y, Ma X, Zhan F, Wang L, Hu T, Zhou H, Hu Z, Zhou W, Zhao L, Chen J,
Meng Y, Wang J, Lin Y, Yuan J, Xie Z, Ma J, Liu WJ, Wang D, Xu W, Holmes
EC, Gao GF, Wu G, Chen W, Shi W, Tan W. 2020. Genomic characterisation
and epidemiology of 2019 novel coronavirus: implications for virus ori-
gins and receptor binding. Lancet 395:5652574. https://doi.org/10
.1016/s0140-6736(20)30251-8.
16. Zaki AM, van Boheemen S, Bestebroer TM, Osterhaus ADME, Fouchier
RAM. 2012. Isolation of a novel coronavirus from a man with pneumonia
in Saudi Arabia. N Engl J Med 367:181421820. https://doi.org/10.1056/
NEJMoa1211721.
17. Pushpakom S, Iorio F, Eyers PA, Escott KJ, Hopper S, Wells A, Doig A,
Guilliams T, Latimer J, McNamee C, Norris A, Sanseau P, Cavalla D,
Pirmohamed M. 2018. Drug repurposing: progress, challenges and rec-
ommendations. Nat Rev Drug Discovery 18:41258. https://doi.org/10
.1038/nrd.2018.168.
18. Mohs RC, Greig NH. 2017. Drug discovery and development: role of basic
biological research. Alzheimers Dement (N Y) 3:6512657. https://doi
.org/10.1016/j.trci.2017.10.005.
19. DeVito N, Inglesby P. 29 March 2020. Evidence-based medicine data lab
COVID-19 TrialsTracker. GitHub https://github.com/ebmdatalab/covid
_trials_tracker-covid.https://doi.org/10.5281/zenodo.3732709.
20. Rochwerg B, Agarwal A, Siemieniuk RA, Agoritsas T, Lamontagne F, Askie
L, Lytvyn L, Leo YS, Macdonald H, Zeng L, Amin W, Burhan E, Bausch FJ,
Calfee CS, Cecconi M, Chanda D, Du B, Geduld H, Gee P, Harley N,
Hashimi M, Hunt B, Kabra SK, Kanda S, Kawano-Dourado L, Kim YJ,
Kissoon N, Kwizera A, Mahaka I, Manai H, Mino G, Nsutebu E, Preller J,
Pshenichnaya N, Qadir N, Sabzwari S, Sarin R, Shankar-Hari M, Sharland
M, Shen Y, Ranganathan SS, Souza JP, Stegemann M, De Sutter A, Ugarte
S, Venkatapuram S, Dat VQ, Vuyiseka D, Wijewickrama A, Maguire B,
Zeraatkar D, et al. 2020. A living WHO guideline on drugs for covid-19.
BMJ 370:m3379. https://doi.org/10.1136/bmj.m3379.
21. Siemieniuk RA, Bartoszko JJ, Ge L, Zeraatkar D, Izcovich A, Kum E, Pardo-
Hernandez H, Qasim A, Martinez JPD, Rochwerg B, Lamontagne F, Han MA,
LiuQ,AgarwalA,AgoritsasT,ChuDK,CoubanR,CusanoE,DarziA,DevjiT,
Fang B, Fang C, Flottorp SA, Foroutan F, Ghadimi M, Heels-Ansdell D,
HonarmandK,HouL,HouX,IbrahimQ,KhamisA,LamB,LoebM,Marcucci
M, McLeod SL, Motaghi S, Murthy S, Mustafa RA, Neary JD, Rada G, Riaz IB,
Sadeghirad B, Sekercioglu N, Sheng L, Sreekanta A, Switzer C, Tendal B,
Thabane L, Tomlinson G, Turner T, Vandvik PO, et al. 2020. Drug treatments
for covid-19: living systematic review and network meta-analysis. BMJ 370:
m2980. https://doi.org/10.1136/bmj.m2980.
22. Elezkurtaj S, Greuel S, Ihlow J, Michaelis EG, Bischoff P, Kunze CA, Sinn
BV, Gerhold M, Hauptmann K, Ingold-Heppner B, Miller F, Herbst H,
Corman VM, Martin H, Radbruch H, Heppner FL, Horst D. 2021. Causes of
death and comorbidities in patients with COVID-19. Sci Rep 11:4263.
https://doi.org/10.1038/s41598-021-82862-5.
23. Zhang B, Zhou X, Qiu Y, Song Y, Feng F, Feng J, Song Q, Jia Q, Wang J.
2020. Clinical characteristics of 82 cases of death from COVID-19. PLoS
One 15:e0235458. https://doi.org/10.1371/journal.pone.0235458.
24. Shi Y, Wang Y, Shao C, Huang J, Gan J, Huang X, Bucci E, Piacentini M,
Ippolito G, Melino G. 2020. COVID-19 infection: the perspectives on
immune responses. Cell Death Differ 27:145121454. https://doi.org/10
.1038/s41418-020-0530-3.
25. Fajgenbaum DC, June CH. 2020. Cytokine storm. N Engl J Med 383:
225522273. https://doi.org/10.1056/NEJMra2026131.
26. Nicholls JM, Poon LL, Lee KC, Ng WF, Lai ST, Leung CY, Chu CM, Hui PK,
Mak KL, Lim W, Yan KW, Chan KH, Tsang NC, Guan Y, Yuen KY, Peiris JS.
2003. Lung pathology of fatal severe acute respiratory syndrome. Lancet
361:177321778. https://doi.org/10.1016/S0140-6736(03)13413-7.
27. Gomersall CD, Kargel MJ, Lapinsky SE. 2004. Pro/con clinical debate: ste-
roids are a key component in the treatment of SARS. Critical Care 8:
1052107. https://doi.org/10.1186/cc2452.
28. Prager R, Pratte MT, Unni RR, Bala S, Ng Fat Hing N, Wu K, McGrath TA,
Thomas A, Thoma B, Kyeremanteng K. 2021. Content analysis and char-
acterization of medical tweets during the early Covid-19 pandemic. Cur-
eus 13:e13594. https://doi.org/10.7759/cureus.13594.
29. Nuventra Pharma Sciences. 2020. Points to consider in drug development of
biologics and small molecules. Nuventra Pharma Sciences, Durham, NC.
https://www.nuventra.com/resources/blog/small-molecules-versus-biologics/.
30. Drews J. 2000. Drug discovery: a historical perspective. Science 287:
196021964. https://doi.org/10.1126/science.287.5460.1960.
31. Thompson AE, Ranard BL, Wei Y, Jelic S. 2020. Prone positioning in awake,
nonintubated patients with COVID-19 hypoxemic respiratory failure. JAMA
Intern Med 180:153721539. https://doi.org/10.1001/jamainternmed.2020
.3030.
32. Stockman LJ, Bellamy R, Garner P. 2006. SARS: systematic review of treat-
ment effects. PLoS Med 3:e343. https://doi.org/10.1371/journal.pmed
.0030343.
33. Fujii T, Iwamoto A, Nakamura T, Iwamoto A. 2004. Current concepts in
SARS treatment. J Infect Chemother 10:127. https://doi.org/10.1007/
s10156-003-0296-9.
34. Stern A, Skalsky K, Avni T, Carrara E, Leibovici L, Paul M. 2017. Corticoste-
roids for pneumonia. Cochrane Database of Systematic Reviews https://
doi.org/10.1002/14651858.CD007720.pub3.
35. Chen Y, Li K, Pu H, Wu T. 2011. Corticosteroids for pneumonia. Cochrane
Database of Systematic Reviews https://doi.org/10.1002/14651858.CD007720
.pub2.
36. Sibila O, Agusti C, Torres A. 2008. Corticosteroids in severe pneumonia.
Eur Respir J 32:2592264. https://doi.org/10.1183/09031936.00154107.
37. Mikami K, Suzuki M, Kitagawa H, Kawakami M, Hirota N, Yamaguchi H,
Narumoto O, Kichikawa Y, Kawai M, Tashimo H, Arai H, Horiuchi T,
Sakamoto Y. 2007. Efcacy of corticosteroids in the treatment of commu-
nity-acquired pneumonia requiring hospitalization. Lung 185:2492255.
https://doi.org/10.1007/s00408-007-9020-3.
38. Nie W, Zhang Y, Cheng J, Xiu Q. 2012. Corticosteroids in the treatment of
community-acquired pneumonia in adults: a meta-analysis. PLoS One 7:
e47926. https://doi.org/10.1371/journal.pone.0047926.
39. Fernández-Serrano S, Dorca J, Garcia-Vidal C, Fernández-Sabé N,
Carratalà J, Fernández-Agüera A, Corominas M, Padrones S, Gudiol F,
Manresa F. 2011. Effect of corticosteroids on the clinical course of
ID and Development of Therapeutics for COVID-19
November/December 2021 Volume 6 Issue 6 e00233-21 msystems.asm.org 41
Downloaded from https://journals.asm.org/journal/msystems on 03 November 2021 by 2607:f470:34:2303:5040:4a48:5de1:cdb5.
community-acquired pneumonia: a randomized controlled trial. Crit
Care 15:R96. https://doi.org/10.1186/cc10103.
40. Villar J, Ferrando C, Martínez D, Ambrós A, Muñoz T, Soler JA, Aguilar G,
Alba F, González-Higueras E, Conesa LA, Martín-Rodríguez C, Díaz-
Domínguez FJ, Serna-Grande P, Rivas R, Ferreres J, Belda J, Capilla L, Tallet
A, Añón JM, Fernández RL, González-Martín JM, dexamethasone in ARDS
network. 2020. Dexamethasone treatment for the acute respiratory dis-
tress syndrome: a multicentre, randomised controlled trial. Lancet Respir
Med 8:2672276. https://doi.org/10.1016/S2213-2600(19)30417-5.
41. Reddy K, OKane C, McAuley D. 2020. Corticosteroids in acute respiratory
distress syndrome: a step forward, but more evidence is needed. Lancet
Respir Med 8:2202222. https://doi.org/10.1016/S2213-2600(20)30048-5.
42. Calfee CS, Matthay MA. 2007. Nonventilatory treatments for acute lung
injury and ARDS. Chest 131:9132920. https://doi.org/10.1378/chest.06
-1743.
43. Umberto Meduri G, Marik PE, Pastores SM, Annane D. 2007. Corticoste-
roids in ARDS. Chest 132:109321094. https://doi.org/10.1378/chest.07
-0714.
44. Steinberg KP, Hudson LD, Goodman RB, Hough CL, Lanken PN, Hyzy R,
Thompson BT, Ancukiewicz M, National Heart, Lung, and Blood Institute
Acute Respiratory Distress Syndrome (ARDS) Clinical Trials Network.
2006. Efcacy and safety of corticosteroids for persistent acute respira-
tory distress syndrome. N Engl J Med 354:167121684. https://doi.org/10
.1056/nejmoa051693.
45. Peter JV, John P, Graham PL, Moran JL, George IA, Bersten A. 2008. Corti-
costeroids in the prevention and treatment of acute respiratory distress
syndrome (ARDS) in adults: meta-analysis. BMJ 336:100621009. https://
doi.org/10.1136/bmj.39537.939039.BE.
46. Yu WC, Hui DSC, Chan-Yeung M. 2004. Antiviral agents and corticoste-
roids in the treatment of severe acute respiratory syndrome (SARS).
Thorax 59:6432645. https://doi.org/10.1136/thx.2003.017665.
47. Yin-Chun Yam L, Lau AC-W, Lai FY-L, Shung E, Chan J, Wong V. 2007. Cor-
ticosteroid treatment of severe acute respiratory syndrome in Hong
Kong. J Infect 54:28239. https://doi.org/10.1016/j.jinf.2006.01.005.
48. Li H, Chen C, Hu F, Wang J, Zhao Q, Gale RP, Liang Y. 2020. Impact of cor-
ticosteroid therapy on outcomes of persons with SARS-CoV-2, SARS-CoV,
or MERS-CoV infection: a systematic review and meta-analysis. Leukemia
34:150321511. https://doi.org/10.1038/s41375-020-0848-3.
49. Wenzel RP, Edmond MB. 2003. Managing SARS amidst uncertainty. N
Engl J Med 348:194721948. https://doi.org/10.1056/NEJMp030072.
50. Khan MF, Lee HJ. 2008. Synthesis and pharmacology of anti-inamma-
tory steroidal antedrugs. Chem Rev 108:513125145. https://doi.org/10
.1021/cr068203e.
51. Ferner RE, DeVito N, Aronson JK, Oxford COVID-19 Evidence Service
Team. 2020. Drug vignettes: dexamethasone. The Centre for Evidence-
Based Medicine, Nufeld Department of Primary Care Health Sciences,
University of Oxford, Oxford, United Kingdom. https://www.cebm.net/
covid-19/dexamethasone/.
52. Zabirowicz ES, Gan TJ. 2019. Pharmacology of postoperative nausea and
vomiting, p 6712692. In Hemmings HC, Jr, Egan TD (ed), Pharmacology
and physiology for anesthesia: foundations and clinical application, 2nd
ed. Elsevier, Philadelphia, PA. https://doi.org/10.1016/b978-0-323-48110
-6.00034-x.
53. Zhou W, Liu Y, Tian D, Wang C, Wang S, Cheng J, Hu M, Fang M, Gao Y.
2020. Potential benets of precise corticosteroids therapy for severe
2019-nCoV pneumonia. Signal Transduct Target Ther 5:18. https://doi
.org/10.1038/s41392-020-0127-9.
54. Arth GE, Johnston DB, Fried J, Spooncer WW, Hoff DR, Sarett LH. 2002.
16-Methylated steroids. I. 16
a
-methylated analogs of cortisone, a new
group of anti-inammatory steroids. J Am Chem Soc 80:316023161.
https://doi.org/10.1021/ja01545a061.
55. Cohen A, Goldman J, Kanenson WL, Turner R, Rose I. 1960. Treatment of
rheumatoid arthritis with dexamethasone. JAMA 174:8312834. https://
doi.org/10.1001/jama.1960.03030070009002.
56. DailyMed. 2007. Dexamethasone. DailyMed, National Institutes of Health,
Bethesda, MD. https://dailymed.nlm.nih.gov/dailymed/drugInfo.cfm?setid=
537b424a-3e07-4c81-978c-1ad99014032a.
57. Orlicka K, Barnes E, Culver EL. 2013. Prevention of infection caused by
immunosuppressive drugs in gastroenterology. Ther Adv Chronic Dis 4:
1672185. https://doi.org/10.1177/2040622313485275.
58. Mehta P, McAuley DF, BrownM, Sanchez E, Tattersall RS, Manson JJ. 2020.
COVID-19: consider cytokine storm syndromes and immunosuppression.
Lancet 395:103321034. https://doi.org/10.1016/S0140-6736(20)30628-0.
59. Russell CD, Millar JE, Baillie J. 2020. Clinical evidencedoes not support cor-
ticosteroid treatment for 2019-nCoV lung injury. Lancet 395:4732475.
https://doi.org/10.1016/S0140-6736(20)30317-2.
60. Shang L, Zhao J, Hu Y, Du R, Cao B. 2020. On the use of corticosteroids for
2019-nCoV pneumonia. Lancet 395:6832684. https://doi.org/10.1016/
S0140-6736(20)30361-5.
61. Ritchie AI, Singanayagam A. 2020. Immunosuppression for hyperinam-
mation in COVID-19: a double-edged sword? Lancet 395:1111. https://
doi.org/10.1016/S0140-6736(20)30691-7.
62. Horby P, Lim WS, Emberson J, Mafham M, Bell J, Linsell L, Staplin N,
Brightling C, Ustianowski A, Elmahi E, ... RECOVERY Collaborative Group.
2020. Effect of dexamethasone in hospitalized patients with COVID-19
preliminary report. medRxiv https://doi.org/10.1101/2020.06.22.20137273.
63. RECOVERY Collaborative Group. 17 July 2020. Dexamethasone in hospi-
talized patients with Covid-19 preliminary report. N Engl J Med
https://doi.org/10.1056/nejmoa2021436.
64. Pasin L, Navalesi P, Zangrillo A, Kuzovlev A, Likhvantsev V, Hajjar LA,
Fresilli S, Lacerda MVG, Landoni G. 2021. Corticosteroids for patients
with coronavirus disease 2019 (COVID) with different disease severity: a
meta-analysis of randomized clinical trials. J Cardiothorac Vasc Anesth
35:5782584. https://doi.org/10.1053/j.jvca.2020.11.057.
65. Lee DW, Gardner R, Porter DL, Louis CU, Ahmed N, Jensen M, Grupp SA,
Mackall CL. 2014. Current concepts in the diagnosis and management of
cytokine release syndrome. Blood 124:1882195. https://doi.org/10.1182/
blood-2014-05-552729.
66. Prescott HC, Rice TW. 2020. Corticosteroids in COVID-19 ARDS: evidence
and hope during the pandemic. JAMA 24:129221295. https://doi.org/10
.1001/jama.2020.16747.
67. Noreen S, Maqbool I, Madni A. 2021. Dexamethasone: therapeutic
potential, risks, and future projection during COVID-19 pandemic. Euro J
Pharmacol 894:173854. https://doi.org/10.1016/j.ejphar.2021.173854.
68. Mahase E. 2020. Covid-19: demand for dexamethasone surges as RECOV-
ERY trial publishes preprint. BMJ 369:m2512. https://doi.org/10.1136/
bmj.m2512.
69. Dimmock NJ, Easton AJ, Leppard KN. 2007. Introduction to modern virol-
ogy, 6th ed. Wiley-Blackwell, Hoboken, NJ.
70. Castleman Disease Collaborative Network. CORONA Data Viewer. https://
cdcn.org/corona-data-viewer/.
71. Maier HJ, Bickerton E, Britton P (ed). 2015. Methods in molecular biology,
vol 1282. Coronaviruses: methods and protocols. Springer, New York, NY.
72. Lung J, Lin Y-S, Yang Y-H, Chou Y-L, Shu L-H, Cheng Y-C, Liu HT, Wu C-Y.
2020. The potential chemical structure of anti-SARS-CoV-2 RNA-depend-
ent RNA polymerase. J Med Virol 92:6932697. https://doi.org/10.1002/
jmv.25761.
73. Totura AL, Bavari S. 2019. Broad-spectrum coronavirus antiviral drug dis-
covery. Expert Opin Drug Discov 14:3972412. https://doi.org/10.1080/
17460441.2019.1581171.
74. Tong S, Su Y, Yu Y, Wu C, Chen J, Wang S, Jiang J. 2020. Ribavirin therapy
for severe COVID-19: a retrospective cohort study. Int J Antimicrob
Agents 56:106114. https://doi.org/10.1016/j.ijantimicag.2020.106114.
75. Seley-Radtke KL, Yates MK. 2018. The evolution of nucleoside analogue
antivirals: a review for chemists and non-chemists. Part 1. Early structural
modications to the nucleoside scaffold. Antiviral Res 154:66286.
https://doi.org/10.1016/j.antiviral.2018.04.004.
76. Jin Z, Smith LK, Rajwanshi VK, Kim B, Deval J. 2013. The ambiguous base-
pairing and high substrate efciency of T-705 (favipiravir) ribofuranosyl
59-triphosphate towards inuenza A virus polymerase. PLoS One 8:
e68347. https://doi.org/10.1371/journal.pone.0068347.
77. DrugBank. 2020. Favipiravir. DrugBank. https://www.drugbank.ca/drugs/
DB12466.
78. Furuta Y, Takahashi K, Fukuda Y, Kuno M, Kamiyama T, Kozaki K, Nomura
N, Egawa H, Minami S, Watanabe Y, Narita H, Shiraki K. 2002. In vitro and
in vivo activities of anti-inuenza virus compound T-705. Antimicrob
Agents Chemother 46:9772981. https://doi.org/10.1128/AAC.46.4.977
-981.2002.
79. Sidwell RW, Barnard DL, Day CW, Smee DF, Bailey KW, Wong M-H,
Morrey JD, Furuta Y. 2007. Efcacy of orally administered T-705 on lethal
avian inuenza A (H5N1) virus infections in mice. Antimicrob Agents
Chemother 51:8452851. https://doi.org/10.1128/AAC.01051-06.
80. Furuta Y, Takahashi K, Kuno-Maekawa M, Sangawa H, Uehara S, Kozaki K,
Nomura N, Egawa H, Shiraki K. 2005. Mechanism of action of T-705
against inuenza virus. Antimicrob Agents Chemother 49:9812986.
https://doi.org/10.1128/AAC.49.3.981-986.2005.
Rando et al.
November/December 2021 Volume 6 Issue 6 e00233-21 msystems.asm.org 42
Downloaded from https://journals.asm.org/journal/msystems on 03 November 2021 by 2607:f470:34:2303:5040:4a48:5de1:cdb5.
81. Julander JG, Shafer K, Smee DF, Morrey JD, Furuta Y. 2009. Activity of T-
705 in a hamster model of yellow fever virus infection in comparison
with that of a chemically related compound, T-1106. Antimicrob Agents
Chemother 53:2022209. https://doi.org/10.1128/AAC.01074-08.
82. Gowen BB, Wong M-H, Jung K-H, Sanders AB, Mendenhall M, Bailey KW,
Furuta Y, Sidwell RW. 2007. In vitro and in vivo activities of T-705 against
arenavirus and bunyavirus infections. Antimicrob Agents Chemother 51:
316823176. https://doi.org/10.1128/AAC.00356-07.
83. Rocha-Pereira J, Jochmans D, Dallmeier K, Leyssen P, Nascimento MSJ,
Neyts J. 2012. Favipiravir (T-705) inhibits in vitro norovirus replication.
Biochem Biophys Res Commun 424:7772780. https://doi.org/10.1016/j
.bbrc.2012.07.034.
84. Mendenhall M, Russell A, Juelich T, Messina EL, Smee DF, Freiberg AN,
Holbrook MR, Furuta Y, de la Torre J-C, Nunberg JH, Gowen BB. 2011. T-
705 (favipiravir) inhibition of arenavirus replication in cell culture. Anti-
microb Agents Chemother 55:7822787. https://doi.org/10.1128/AAC
.01219-10.
85. Furuta Y, Komeno T, Nakamura T. 2017. Favipiravir (T-705), a broad spec-
trum inhibitor of viral RNA polymerase. Proc Jpn Acad Ser B Phys Biol Sci
93:4492463. https://doi.org/10.2183/pjab.93.027.
86. Gordon CJ, Tchesnokov EP, Feng JY, Porter DP, Götte M. 2020. The antivi-
ral compound remdesivir potently inhibits RNA-dependent RNA poly-
merase from Middle East respiratory syndrome coronavirus. J Biol Chem
295:477324779. https://doi.org/10.1074/jbc.AC120.013056.
87. Agostini ML, Andres EL, Sims AC, Graham RL, Sheahan TP, Lu X, Smith EC,
Case JB, Feng JY, Jordan R, Ray AS, Cihlar T, Siegel D, Mackman RL, Clarke
MO, Baric RS, Denison MR. 2018. Coronavirus susceptibility to the antivi-
ral remdesivir (GS-5734) is mediated by the viral polymerase and the
proofreading exoribonuclease. mBio 9:e00221-18. https://doi.org/10
.1128/mBio.00221-18.
88. Wang M, Cao R, Zhang L, Yang X, Liu J, Xu M, Shi Z, Hu Z, Zhong W, Xiao
G. 2020. Remdesivir and chloroquine effectively inhibit the recently
emerged novel coronavirus (2019-nCoV) in vitro. Cell Res 30:2692271.
https://doi.org/10.1038/s41422-020-0282-0.
89. Beigel JH, Tomashek KM, Dodd LE, Mehta AK, Zingman BS, Kalil AC,
Hohmann E, Chu HY, Luetkemeyer A, Kline S, Lopez de Castilla D,
Finberg RW, Dierberg K, Tapson V, Hsieh L, Patterson TF, Paredes R,
Sweeney DA, Short WR, Touloumi G, Lye DC, Ohmagari N, Oh MD, Ruiz-
Palacios GM, Beneld T, Fätkenheuer G, Kortepeter MG, Atmar RL,
Creech CB, Lundgren J, Babiker AG, Pett S, Neaton JD, Burgess TH,
Bonnett T, Green M, Makowski M, Osinusi A, Nayak S, Lane HC. 2020.
Remdesivir for the treatment of Covid-19 nal report. N Engl J Med
383:181321826. https://doi.org/10.1056/NEJMoa2007764.
90. National Institute of Allergy and Infectious Diseases (NIAID). 2020. A mul-
ticenter, adaptive, randomized blinded controlled trial of the safety and
efcacy of investigational therapeutics for the treatment of COVID-19 in
hospitalized adults. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/
NCT04280705.
91. Gilead Sciences. 2020. A phase 3 randomized study to evaluate the
safety and antiviral activity of remdesivir (GS-5734
TM
) in participants
with severe COVID-19. ClinicalTrials.gov https://clinicaltrials.gov/ct2/
show/NCT04292899.
92. Grein J, Ohmagari N, Shin D, Diaz G, Asperges E, Castagna A, Feldt T,
Green G, Green ML, Lescure FX, Nicastri E, Oda R, Yo K, Quiros-Roldan E,
Studemeister A, Redinski J, Ahmed S, Bernett J, Chelliah D, Chen D,
Chihara S, Cohen SH, Cunningham J, D'Arminio Monforte A, Ismail S,
Kato H, Lapadula G, L'Her E, Maeno T, Majumder S, Massari M, Mora-Rillo
M, Mutoh Y, Nguyen D, Verweij E, Zoufaly A, Osinusi AO, DeZure A, Zhao
Y, Zhong L, Chokkalingam A, Elboudwarej E, Telep L, Timbs L, Henne I,
Sellers S, Cao H, Tan SK, Winterbourne L, Desai P, Mera R, et al. 2020.
Compassionate use of remdesivir for patients with severe Covid-19. N
Engl J Med 382:232722336. https://doi.org/10.1056/NEJMoa2007016.
93. WHO Solidarity Trial Consortium. 2020. Repurposed antiviral drugs for
Covid-19 interim WHO Solidarity Trial results. N Engl J Med 384:
4972511. https://doi.org/10.1056/nejmoa2023184.
94. DailyMed. 2020. PLAQUENIL - hydroxychloroquine sulfate tablet. Dai-
lyMed, National Institutes of Health, Bethesda, MD. https://dailymed.nlm
.nih.gov/dailymed/drugInfo.cfm?setid=34496b43-05a2-45fb-a769-52b12e
099341.
95. Kalia S, Dutz JP. 2007. New concepts in antimalarial use and mode of
action in dermatology. Dermatol Ther 20:1602174. https://doi.org/10
.1111/j.1529-8019.2007.00131.x.
96. Vincent MJ, Bergeron E, Benjannet S, Erickson BR, Rollin PE, Ksiazek TG,
Seidah NG, Nichol ST. 2005. Chloroquine is a potent inhibitor of SARS
coronavirus infection and spread. Virol J 2:69. https://doi.org/10.1186/
1743-422X-2-69.
97. Yao X, Ye F, Zhang M, Cui C, Huang B, Niu P, Liu X, Zhao L, Dong E, Song
C, Zhan S, Lu R, Li H, Tan W, Liu D. 2020. In vitro antiviral activity and pro-
jection of optimized dosing design of hydroxychloroquine for the treat-
ment of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2).
Clin Infect Dis 71:7322739. https://doi.org/10.1093/cid/ciaa237.
98. Gautret P, Lagier JC, Parola P, Hoang VT, Meddeb L, Mailhe M, Doudier B,
Courjon J, Giordanengo V, Vieira VE, Tissot Dupont H, Honoré S, Colson
P, Chabrière E, La Scola B, Rolain JM, Brouqui P, Raoult D. 2020. Hydroxy-
chloroquine and azithromycin as a treatment of COVID-19: results of an
open-label non-randomized clinical trial. Int J Antimicrob Agents 56:
105949. https://doi.org/10.1016/j.ijantimicag.2020.105949.
99. International Society of Antimicrobial Chemotherapy. 2020. Statement
on IJAA paper. Ofcial Statement from International Society of Antimi-
crobial Chemotherapy (ISAC). Hydroxychloroquine and azithromycin as
a treatment of COVID-19: results of an open-label non-randomized clini-
cal trial (Gautret P et al. PMID 32205204). https://www.isac.world/news
-and-publications/ofcial-isac-statement.
100. RECOVERY Collaborative Group, Horby P, Mafham M, Linsell L, Bell JL,
Staplin N, Emberson JR, Wiselka M, Ustianowski A, Elmahi E, Prudon B,
Whitehouse T, Felton T, Williams J, Faccenda J, Underwood J, Baillie JK,
Chappell LC, Faust SN, Jaki T, Jeffery K, Lim WS, Montgomery A, Rowan K,
Tarning J, Watson JA, White NJ, Juszczak E, Haynes R, Landray M. 2020.
Effect of hydroxychloroquine in hospitalized patients with Covid-19. N
Engl J Med 383:203022040. https://doi.org/10.1056/nejmoa2022926.
101. RECOVERY (Randomised Evaluation of COVID-19 Therapy). 5 June 2020. No
clinical benet from use of hydroxychloroquine in hospitalised patients
with COVID-19 RECOVERY Trial. RECOVERY (Randomised Evaluation of
COVID-19 Therapy), Nufeld Department of Population Health, University
of Oxford, Oxford, United Kingdom. https://www.recoverytrial.net/news/
statement-from-the-chief-investigators-of-the-randomised-evaluation-of
-covid-19-therapy-recovery-trial-on-hydroxychloroquine-5-june-2020-no-
clinical-benet-from-use-of-hydroxychloroquine-in-hospitalised-patients
-with-covid-19.
102. Õmura S, Crump A. 2004. The life and times of ivermectin asuccess
story. Nat Rev Microbiol 2:9842989. https://doi.org/10.1038/nrmicro1048.
103. Burg RW, Miller BM, Baker EE, Birnbaum J, Currie SA, Hartman R, Kong
YL, Monaghan RL, Olson G, Putter I, Tunac JB, Wallick H, Stapley EO,
Oiwa R, Omura S. 1979. Avermectins, new family of potent anthelmintic
agents: producing organism and fermentation. Antimicrob Agents Che-
mother 15:3612367. https://doi.org/10.1128/AAC.15.3.361.
104. Crump A, Omura S. 2011. Ivermectin, 'wonder drug' from Japan: the
human use perspective. Proc Jpn Acad Ser B Phys Biol Sci 87:13228.
https://doi.org/10.2183/pjab.87.13.
105. Crump A. 2017. Ivermectin: enigmatic multifaceted wonderdrug con-
tinues to surprise and exceed expectations. J Antibiot 70:4952505.
https://doi.org/10.1038/ja.2017.11.
106. Egerton JR, Ostlind DA, Blair LS, Eary CH, Suhayda D, Cifelli S, Riek RF,
Campbell WC. 1979. Avermectins, new family of potent anthelmintic
agents: efcacy of the B1a component. Antimicrob Agents Chemother
15:3722378. https://doi.org/10.1128/AAC.15.3.372.
107. Heidary F, Gharebaghi R. 2020. Ivermectin: a systematic review from
antiviral effects to COVID-19 complementary regimen. J Antibiot 73:
5932602. https://doi.org/10.1038/s41429-020-0336-z.
108. Sharun K, Dhama K, Patel SK, Pathak M, Tiwari R, Singh BR, Sah R, Bonilla-
Aldana D, Rodriguez-Morales AJ, Leblebicioglu H. 2020. Ivermectin, a new
candidate therapeutic against SARS-CoV-2/COVID-19. Ann Clin Microbiol
Antimicrob 19:23. https://doi.org/10.1186/s12941-020-00368-w.
109. Caly L, Druce JD, Catton MG, Jans DA, Wagstaff KM. 2020. The FDA-
approved drug ivermectin inhibits the replication of SARS-CoV-2 in vitro.
Antiviral Res 178:104787. https://doi.org/10.1016/j.antiviral.2020.104787.
110. Chaccour C, Hammann F, Ramón-García S, Rabinovich N. 2020. Ivermec-
tin and COVID-19: keeping rigor in times of urgency. Am J Trop Med Hyg
102:115621157. https://doi.org/10.4269/ajtmh.20-0271.
111. Schmith VD, Zhou JJ, Lohmer LRL. 2020. The approved dose of ivermec-
tin alone is not the ideal dose for the treatment of COVID-19. Clin Phar-
macol Ther 108:7622765. https://doi.org/10.1002/cpt.1889.
112. Momekov G, Momekova D. 2020. Ivermectin as a potential COVID-19
treatment from the pharmacokinetic point of view: antiviral levels are not
likely attainable with known dosing regimens. Biotechnol Biotechnol
Equip 34:4692474. https://doi.org/10.1080/13102818.2020.1775118.
113. Ménez C, Sutra J-F, Prichard R, Lespine A. 2012. Relative neurotoxicity of
ivermectin and moxidectin in Mdr1ab (2/2) mice and effects on
ID and Development of Therapeutics for COVID-19
November/December 2021 Volume 6 Issue 6 e00233-21 msystems.asm.org 43
Downloaded from https://journals.asm.org/journal/msystems on 03 November 2021 by 2607:f470:34:2303:5040:4a48:5de1:cdb5.
mammalian GABA(A) channel activity. PLoS Negl Trop Dis 6:e1883.
https://doi.org/10.1371/journal.pntd.0001883.
114. Rajter JC, Sherman MS, Fatteh N, Vogel F, Sacks J, Rajter J-J. 2021. Use of
ivermectin is associated with lower mortality in hospitalized patients
with coronavirus disease 2019: the ivermectin in COVID nineteen study.
Chest 159:85292. https://doi.org/10.1016/j.chest.2020.10.009.
115. Camprubí D, Almuedo-Riera A, Martí-Soler H, Soriano A, Hurtado JC,
Subirà C, Grau-Pujol B, Krolewiecki A, Muñoz J. 2020. Lack of efcacy of
standard doses of ivermectin in severe COVID-19 patients. PLoS One 15:
e0242184. https://doi.org/10.1371/journal.pone.0242184.
116. Niaee MS, Namdar P, Allami A, Zolghadr L, Javadi A, Karampour A,
Varnaseri M, Bijani B, Cheraghi F, Naderi Y, ... Gheibi N. 2021. Ivermectin
as an adjunct treatment for hospitalized adult COVID-19 patients: a
randomized multi-center clinical trial. Asian Pac J Trop Med 14:2662273.
https://doi.org/10.4103/1995-7645.318304.
117. Babalola OE, Bode CO, Ajayi AA, Alakaloko FM, Akase IE, Otrofanowei E,
Salu OB, Adeyemo WL, Ademuyiwa AO, Omilabu S. 2021. Ivermectin
shows clinical benets in mild to moderate COVID19: a randomized con-
trolled double-blind, dose-response study in Lagos. QJM https://doi.org/
10.1093/qjmed/hcab035.
118. Pott-Junior H, Paoliello MMB, Miguel ADQC, da Cunha AF, de Melo Freire
CC, Neves FF, da Silva de Avó LR, Roscani MG, Dos Santos SDS, Chachá
SGF. 2021. Use of ivermectin in the treatment of Covid-19: a pilot trial.
Toxicol Rep 8:5052510. https://doi.org/10.1016/j.toxrep.2021.03.003.
119. Ahmed S, Karim MM, Ross AG, Hossain MS, Clemens JD, Sumiya MK,
Phru CS, Rahman M, Zaman K, Somani J, Yasmin R, Hasnat MA, Kabir A,
Aziz AB, Khan WA. 2021. A ve-day course of ivermectin for the treat-
ment of COVID-19 may reduce the duration of illness. Int J Infect Dis
103:2142216. https://doi.org/10.1016/j.ijid.2020.11.191.
120. Shahbaznejad L, Davoudi A, Eslami G, Markowitz JS, Navaeifar MR,
Hosseinzadeh F, Movahedi FS, Rezai MS. 2021. Effects of ivermectin in
patients with COVID-19: a multicenter, double-blind, randomized, con-
trolled clinical trial. Clin Ther 43:100721019. https://doi.org/10.1016/j
.clinthera.2021.04.007.
121. Chaccour C, Casellas A, Blanco-Di Matteo A, Pineda I, Fernandez-
Montero A, Ruiz-Castillo P, Richardson MA, Rodríguez-Mateos M, Jordán-
Iborra C, Brew J, Carmona-Torre F, Giráldez M, Laso E, Gabaldón-Figueira
JC, Dobaño C, Moncunill G, Yuste JR, Del Pozo JL, Rabinovich NR,
Schöning V, Hammann F, Reina G, Sadaba B, Fernández-Alonso M. 2021.
The effect of early treatment with ivermectin on viral load, symptoms
and humoral response in patients with non-severe COVID-19: a pilot,
double-blind, placebo-controlled, randomized clinical trial. EClinicalMe-
dicine 32:100720. https://doi.org/10.1016/j.eclinm.2020.100720.
122. Mohan A, Tiwari P, Suri T, Mittal S, Patel A, Jain A, Velpandian T, Das UK,
Bopanna TK, Pandey RM, ... Guleria R. 2021. Ivermectin in mild and
moderate COVID-19 (RIVET-COV): a randomized, placebo-controlled
trial. Research Square https://doi.org/10.21203/rs.3.rs-191648/v1.
123. Ravikirti, Roy R, Pattadar C, Raj R, Agarwal N, Biswas B, Manjhi PK, Rai DK,
Shyama, Kumar A, Sarfaraz A. 2021. Evaluation of ivermectin as a poten-
tial treatment for mild to moderate COVID-19: a double-blind random-
ized placebo controlled trial in Eastern India. J Pharm Pharm Sci 24:
3432350. https://doi.org/10.18433/jpps32105.
124. Elgazzar A, Eltaweel A, Youssef SA, Hany B, Hafez M, Moussa H. 28 Decem-
ber 2020. Efcacy and safety of ivermectin for treatment and prophylaxis
of COVID-19 pandemic. Research Square https://doi.org/10.21203/rs.3.rs
-100956/v3.
125. Grftr News. 15 July 2021. Why was a major study on ivermectin for COVID-
19 just retracted? Grftr News. Grifter analysis and review. https://grftr
.news/why-was-a-major-study-on-ivermectin-for-covid-19-just-retracted/.
126. Brown N. 15 July 2021. Some problems in the dataset of a large study of
ivermectin for the treatment of Covid-19. Nick Browns blog. https://
steamtraen.blogspot.com/2021/07/Some-problems-with-the-data-from-a-
Covid-study.html.
127. Research Square. 14 July 2021. Research Square withdrew the preprint
Efcacy and safety of ivermectin for treatment and prophylaxis of
COVID-19 pandemic.Research Square https://doi.org/10.21203/rs.3.rs
-100956/v4.
128. López-Medina E, López P, Hurtado IC, Dávalos DM, Ramirez O, Martínez
E, Díazgranados JA, Oñate JM, Chavarriaga H, Herrera S, Parra B, Libreros
G, Jaramillo R, Avendaño AC, Toro DF, Torres M, Lesmes MC, Rios CA,
Caicedo I. 2021. Effect of ivermectin on time to resolution of symptoms
among adults with mild COVID-19. JAMA 325:142621435. https://doi
.org/10.1001/jama.2021.3071.
129. Roman YM, Burela PA, Pasupuleti V, Piscoya A, Vidal JE, Hernandez AV.
2021. Ivermectin for the treatment of COVID-19: a systematic review and
meta-analysis of randomized controlled trials. medRxiv https://doi.org/
10.1101/2021.05.21.21257595.
130. Castañeda-Sabogal A, Chambergo-Michilot D, Toro-Huamanchumo CJ,
Silva-Rengifo C, Gonzales-Zamora J, Barboza JJ. 2021. Outcomes of iver-
mectin in the treatment of COVID-19: a systematic review and meta-
analysis. medRxiv https://doi.org/10.1101/2021.01.26.21250420.
131. Hill A, Garratt A, Levi J, Falconer J, Ellis L, McCann K, Pilkington V, Qavi A,
Wang J, Wentzel H. 2021. Expression of concern: Meta-analysis of
randomized trials of ivermectin to treat SARS-CoV-2 infection.Open Fo-
rum Infect Dis 8:ofab394. https://doi.org/10.1093/od/ofab394.
132. Zein AFMZ, Sulistiyana CS, Raffaelo WM, Pranata R. 2021. Ivermectin and
mortality in patients with COVID-19: a systematic review, meta-analysis,
and meta-regression of randomized controlled trials. Diabetes Metab
Syndr 15:102186. https://doi.org/10.1016/j.dsx.2021.102186.
133. Bryant A, Lawrie TA, Dowswell T, Fordham EJ, Mitchell S, Hill SR, Tham
TC. 2021. Ivermectin for prevention and treatment of COVID-19 infec-
tion: a systematic review, meta-analysis, and trial sequential analysis to
inform clinical guidelines. Am J Ther 28:e4342e460. https://doi.org/10
.1097/MJT.0000000000001402.
134. Hariyanto TI, Halim DA, Rosalind J, Gunawan C, Kurniawan A. 2021. Iver-
mectin and outcomes from Covid-19 pneumonia: a systematic review
and meta-analysis of randomized clinical trial studies. Rev Med Virol
https://doi.org/10.1002/rmv.2265.
135. Bryant A, Lawrie TA, Dowswell T, Fordham E, Scott M, Hill SR, Tham TC.
2021. Ivermectin for prevention and treatment of COVID-19 infection: a
systematic review and meta-analysis. OSF Preprints https://doi.org/10
.31219/osf.io/k37ft.
136. Kory P, Meduri GU, Varon J, Iglesias J, Marik PE. 2021. Review of the
emerging evidence demonstrating the efcacy of ivermectin in the pro-
phylaxis and treatment of COVID-19. Am J Ther 28:e2992e318. https://
doi.org/10.1097/MJT.0000000000001377.
137. Kow CS, Merchant HA, Mustafa ZU, Hasan SS. 2021. The association
between the use of ivermectin and mortality in patients with COVID-19:
a meta-analysis. Pharmacol Rep 73:147321479. https://doi.org/10.1007/
s43440-021-00245-z.
138. Karale S, Bansal V, Makadia J, Tayyeb M, Khan H, Ghanta SS, Singh R,
Tekin A, Bhurwal A, Mutneja H, ... Kashyap R. 2021. A meta-analysis of
mortality, need for ICU admission, use of mechanical ventilation and
adverse effects with ivermectin use in COVID-19 patients. medRxiv
https://doi.org/10.1101/2021.04.30.21256415.
139. Gideon M-K. 22 June 2021. Does ivermectin work for Covid-19? Health
Nerd Medium https://gidmk.medium.com/does-ivermectin-work-for-covid
-19-1166126c364a.
140. Hill A, Garratt A, Levi J, Falconer J, Ellis L, McCann K, Pilkington V, Qavi A,
Wang J, Wentzel H. 2021. Meta-analysis of randomized trials of ivermec-
tin to treat SARS-CoV-2 infection. Open Forum Infect Dis 8:ofab358.
https://doi.org/10.1093/od/ofab358.
141. US Food and Drug Administration. 2021. FAQ: COVID-19 and ivermectin
intended for animals. Center for Veterinary Medicine, US Food and Drug
Administration, Silver Spring, MD. https://www.fda.gov/animal-veterinary/
product-safety-information/faq-covid-19-and-ivermectin-intended-animals.
142. Cardresearch. 2021. A multicenter, prospective, adaptive, double-blind,
randomized, placebo-controlled study to evaluate the effect of uvox-
amine, ivermectin, doxasozin and interferon lambda 1A in mild COVID-
19 and high risk of complications. ClinicalTrials.gov https://clinicaltrials
.gov/ct2/show/NCT04727424.
143. PRINCIPLE. 2021. Ivermectin to be investigated in adults aged 181as a
possible treatment for COVID-19 in the PRINCIPLE trial. Platform Rando-
mised Trial of Treatments in the Community for Epidemic and Pandemic
Illnesses (PRINCIPLE). https://www.principletrial.org/news/ivermectin-to
-be-investigated-as-a-possible-treatment-for-covid-19-in-oxford2019s
-principle-trial.
144. Reis G, Moreira Silva EADS, Medeiros Silva DC, Thabane L, Singh G, Park
JJH, Forrest JI, Harari O, Quirino Dos Santos CV, Guimarães de Almeida
APF, Figueiredo Neto AD, Savassi LCM, Milagres AC, Teixeira MM,
Simplicio MIC, Ribeiro LB, Oliveira R, Mills EJ, TOGETHER Investigators.
2021. Effect of early treatment with hydroxychloroquine or lopinavir
and ritonavir on risk of hospitalization among patients with COVID-19.
JAMA Netw Open 4:e216468. https://doi.org/10.1001/jamanetworkopen
.2021.6468.
145. NIH Collaboratory. 2021. August 6, 2021: early treatment of COVID-19
with repurposed therapies: the TOGETHER Adaptive Platform Trial
Rando et al.
November/December 2021 Volume 6 Issue 6 e00233-21 msystems.asm.org 44
Downloaded from https://journals.asm.org/journal/msystems on 03 November 2021 by 2607:f470:34:2303:5040:4a48:5de1:cdb5.
(Edward Mills, PhD, FRCP). Rethinking clinical trials: a living textbook of
pragmatic clinical trials. NIH Collaboratory. https://rethinkingclinicaltrials
.org/news/august-6-2021-early-treatment-of-covid-19-with-repurposed
-therapies-the-together-adaptive-platform-trial-edward-mills-phd-frcp/.
146. ClinCalc DrugStats Database. 2021. Lisinopril: drug usage statistics,
United States, 2013 2019. ClinCalc DrugStats Database. https://clincalc
.com/DrugStats/Drugs/Lisinopril.
147. Byrd J, Chertow GM, Bhalla V. 2019. Hypertension hot potato 2anatomy
of the angiotensin-receptor blocker recalls. N Engl J Med 380:158921591.
https://doi.org/10.1056/NEJMp1901657.
148. Bian B, Kelton CM, Guo JJ, Wigle PR. 2010. ACE inhibitor and ARB utiliza-
tion and expenditures in the Medicaid fee-for-service program from 1991
to 2008. J Manag Care Pharm 16:671679. https://doi.org/10.18553/jmcp
.2010.16.9.671.
149. Turner AJ, Hiscox JA, Hooper NM. 2004. ACE2: from vasopeptidase to
SARS virus receptor. Trends Pharmacol Sci 25:2912294. https://doi.org/
10.1016/j.tips.2004.04.001.
150. Huentelman MJ, Zubcevic J, Hernández Prada JA, Xiao X, Dimitrov DS,
Raizada MK, Ostrov DA. 2004. Structure-based discovery of a novel an-
giotensin-converting enzyme 2 inhibitor. Hypertension 44:9032906.
https://doi.org/10.1161/01.HYP.0000146120.29648.36.
151. Dimitrov DS. 2003. The secret life of ACE2 as a receptor for the SARS vi-
rus. Cell 115:6522653. https://doi.org/10.1016/s0092-8674(03)00976-0.
152. Kreutz R, Algharably EAE-H, Azizi M, Dobrowolski P, Guzik T, Januszewicz
A, Persu A, Prejbisz A, Riemer TG, Wang J-G, Burnier M. 2020. Hyperten-
sion, the reninangiotensin system, and the risk of lower respiratory
tract infections and lung injury: implications for COVID-19. Cardiovasc
Res 116:168821699. https://doi.org/10.1093/cvr/cvaa097.
153. Walters TE, Kalman JM, Patel SK, Mearns M, Velkoska E, Burrell LM. 2016.
Angiotensin converting enzyme 2 activity and human atrial brillation:
increased plasma angiotensin converting enzyme 2 activity is associated
with atrial brillation and more advanced left atrial structural remodelling.
Europace 19:128021287. https://doi.org/10.1093/europace/euw246.
154. Mehra MR, Desai SS, Kuy S, Henry TD, Patel AN. 2020. Cardiovascular dis-
ease, drug therapy, and mortality in Covid-19. N Engl J Med 382:e102.
https://doi.org/10.1056/NEJMc2021225.
155. Mehra MR, Desai SS, Kuy S, Henry TD, Patel AN. 2020. Retraction: cardio-
vascular disease, drug therapy, and mortality in Covid-19. N. Engl J Med
DOI: 10.1056/NEJMoa2007621. N Engl J Med 382:2582. https://doi.org/
10.1056/NEJMc2021225.
156. Cohen JB, Hanff TC, William P, Sweitzer N, Rosado-Santander NR, Medina
C, Rodriguez-Mori JE, Renna N, Chang TI, Corrales-Medina V, Andrade-
Villanueva JF, Barbagelata A, Cristodulo-Cortez R, Díaz-Cucho OA, Spaak
J, Alfonso CE, Valdivia-Vega R, Villavicencio-Carranza M, Ayala-García RJ,
Castro-Callirgos CA, González-Hernández LA, Bernales-Salas EF, Coacalla-
Guerra JC, Salinas-Herrera CD, Nicolosi L, Basconcel M, Byrd JB, Sharkoski
T, Bendezú-Huasasquiche LE, Chittams J, Edmonston DL, Vasquez CR,
Chirinos JA. 2021. Continuation versus discontinuation of reninangiotensin
system inhibitors in patients admitted to hospital with COVID-19: a prospec-
tive, randomised, open-label trial. Lancet Respir Med 9:275284. https://doi
.org/10.1016/S2213-2600(20)30558-0.
157. Lopes RD, Macedo AVS, de Barros E, Silva PGM, Moll-Bernardes RJ, Dos
Santos TM, Mazza L, Feldman A, D'Andréa Saba Arruda G, de
Albuquerque DC, Camiletti AS, de Sousa AS, de Paula TC, Giusti KGD,
Domiciano RAM, Noya-Rabelo MM, Hamilton AM, Loures VA, Dionísio
RM, Furquim TAB, De Luca FA, Dos Santos Sousa ÍB, Bandeira BS,
Zukowski CN, de Oliveira RGG, Ribeiro NB, de Moraes JL, Petriz JLF,
Pimentel AM, Miranda JS, de Jesus Abufaiad BE, Gibson CM, Granger CB,
Alexander JH, de Souza OF, BRACE CORONA Investigators. 2021. Effect
of discontinuing vs continuing angiotensin-converting enzyme inhibi-
tors and angiotensin II receptor blockers on days alive and out of the
hospital in patients admitted with COVID-19. JAMA 325:2542264.
https://doi.org/10.1001/jama.2020.25864.
158. US Food and Drug Administration. 2020. Frequently asked questions on
the revocation of the emergency use authorization for hydroxychloroquine
sulfate and chloroquine phosphate. US Food and Drug Administration, Sil-
ver Spring, MD. https://www.fda.gov/media/138946/download.
159. European Medicines Agency. 4 January 2020. COVID-19: chloroquine and
hydroxychloroquine only to be used in clinical trials or emergency use pro-
grammes. European Medicines Agency, Amsterdam, Netherlands. https://
www.ema.europa.eu/en/news/covid-19-chloroquine-hydroxychloroquine
-only-be-used-clinical-trials-emergency-use-programmes.
160. Shire SJ. 2009. Formulation and manufacturability of biologics. Curr
Opin Biotechnol 20:7082714. https://doi.org/10.1016/j.copbio.2009.10
.006.
161. Baumann A. 2006. Early development of therapeutic biologics 22
pharmacokinetics. Curr Drug Metab 7:15221. https://doi.org/10.2174/
138920006774832604.
162. Lu R-M, Hwang Y-C, Liu I-J, Lee C-C, Tsai H-Z, Li H-J, Wu H-C. 2020. Devel-
opment of therapeutic antibodies for the treatment of diseases. J
Biomed Sci 27:1. https://doi.org/10.1186/s12929-019-0592-z.
163. Corti D, Lanzavecchia A. 2013. Broadly neutralizing antiviral antibodies.
Annu Rev Immunol 31:7052742. https://doi.org/10.1146/annurev-immunol
-032712-095916.
164. Iacob SA, Iacob DG. 2017. Ibalizumab targeting CD4 receptors, an
emerging molecule in HIV therapy. Front Microbiol 8:2323. https://doi
.org/10.3389/fmicb.2017.02323.
165. Resch B. 2017. Product review on the monoclonal antibody palivizumab
for prevention of respiratory syncytial virus infection. Hum Vacc Immun-
other 13:213822149. https://doi.org/10.1080/21645515.2017.1337614.
166. Houser KV, Gretebeck L, Ying T, Wang Y, Vogel L, Lamirande EW, Bock
KW, Moore IN, Dimitrov DS, Subbarao K. 2016. Prophylaxis with a Middle
East respiratory syndrome coronavirus (MERS-CoV)specic human
monoclonal antibody protects rabbits from MERS-CoV infection. J Infect
Dis 213:155721561. https://doi.org/10.1093/infdis/jiw080.
167. van Doremalen N, Falzarano D, Ying T, de Wit E, Bushmaker T, Feldmann
F, Okumura A, Wang Y, Scott DP, Hanley PW, Feldmann H, Dimitrov DS,
Munster VJ. 2017. Efcacy of antibody-based therapies against Middle
East respiratory syndrome coronavirus (MERS-CoV) in common marmo-
sets. Antiviral Res 143:30237. https://doi.org/10.1016/j.antiviral.2017.03
.025.
168. Tanaka T, Narazaki M, Kishimoto T. 2014. IL-6 in inammation, immunity,
and disease. Cold Spring Harb Perspect Biol 6:a016295. https://doi.org/
10.1101/cshperspect.a016295.
169. DailyMed. 2020. ACTEMRA - tocilizumab. DailyMed, National Institutes of
Health, Bethesda, MD. https://dailymed.nlm.nih.gov/dailymed/drugInfo
.cfm?setid=2e5365ff-cb2a-4b16-b2c7-e35c6bf2de13.
170. Zhou F, Yu T, Du R, Fan G, Liu Y, Liu Z, Xiang J, Wang Y, Song B, Gu X, Guan
L,WeiY,LiH,WuX,XuJ,TuS,ZhangY,ChenH,CaoB.2020.Clinicalcourse
and risk factors for mortality of adult inpatients with COVID-19 in Wuhan,
China: a retrospective cohort study. Lancet 395:10541062. https://doi.org/
10.1016/S0140-6736(20)30566-3.
171. Guaraldi G, Meschiari M, Cozzi-Lepri A, Milic J, Tonelli R, Menozzi M,
Franceschini E, Cuomo G, Orlando G, Borghi V, Santoro A, Di Gaetano M,
Puzzolante C, Carli F, Bedini A, Corradi L, Fantini R, Castaniere I, Tabbì L,
Girardis M, Tedeschi S, Giannella M, Bartoletti M, Pascale R, Dolci G,
Brugioni L, Pietrangelo A, Cossarizza A, Pea F, Clini E, Salvarani C, Massari
M, Viale PL, Mussini C. 2020. Tocilizumab in patients with severe COVID-
19: a retrospective cohort study. Lancet Rheumatol 2:e474e484.
https://doi.org/10.1016/S2665-9913(20)30173-9.
172. Price CC, Altice FL, Shyr Y, Koff A, Pischel L, Goshua G, Azar MM,
Mcmanus D, Chen S-C, Gleeson SE, Britto CJ, Azmy V, Kaman K, Gaston
DC, Davis M, Burrello T, Harris Z, Villanueva MS, Aoun-Barakat L, Kang I,
Seropian S, Chupp G, Bucala R, Kaminski N, Lee AI, LoRusso PM, Topal JE,
Dela Cruz C, Malinis M. 2020. Tocilizumab treatment for cytokine release
syndrome in hospitalized patients with coronavirus disease 2019. Chest
158:13971408. https://doi.org/10.1016/j.chest.2020.06.006.
173. Capra R, De Rossi N, Mattioli F, Romanelli G, Scarpazza C, Sormani MP,
Cossi S. 2020. Impact of low dose tocilizumab on mortality rate in
patients with COVID-19 related pneumonia. Eur J Intern Med 76:3135.
https://doi.org/10.1016/j.ejim.2020.05.009.
174. Klopfenstein T, Zayet S, Lohse A, Balblanc J-C, Badie J, Royer P-Y, Toko L,
Mezher C, Kadiane-Oussou NJ, Bossert M, Bozgan A-M, Charpentier A,
Roux M-F, Contreras R, Mazurier I, Dussert P, Gendrin V, Conrozier T, HNF
Hospital Tocilizumab multidisciplinary team. 2020. Tocilizumab therapy
reduced intensive care unit admissions and/or mortality in COVID-19
patients. Med Mal Infect 50:397400. https://doi.org/10.1016/j.medmal
.2020.05.001.
175. Rojas-Marte G, Khalid M, Mukhtar O, Hashmi AT, Waheed MA, Ehrlich S,
Aslam A, Siddiqui S, Agarwal C, Malyshev Y, Henriquez-Felipe C, Sharma
D, Sharma S, Chukwuka N, Rodriguez DC, Alliu S, Le J, Shani J. 2020. Out-
comes in patients with severe COVID-19 disease treated with tocilizu-
mab: a casecontrolled study. QJM 113:5462550. https://doi.org/10
.1093/qjmed/hcaa206.
176. Xu X, Han M, Li T, Sun W, Wang D, Fu B, Zhou Y, Zheng X, Yang Y, Li X,
Zhang X, Pan A, Wei H. 2020. Effective treatment of severe COVID-19
ID and Development of Therapeutics for COVID-19
November/December 2021 Volume 6 Issue 6 e00233-21 msystems.asm.org 45
Downloaded from https://journals.asm.org/journal/msystems on 03 November 2021 by 2607:f470:34:2303:5040:4a48:5de1:cdb5.
patients with tocilizumab. Proc Natl Acad Sci U S A 117:10970210975.
https://doi.org/10.1073/pnas.2005615117.
177. RECOVERY Collaborative Group, Horby PW, Pessoa-Amorim G, Peto L,
Brightling CE, Sarkar R, Thomas K, Jeebun V, Ashish A, Tully R, Chadwick
D, ... Landry MJ. 2021. Tocilizumab in patients admitted to hospital
with COVID-19 (RECOVERY): preliminary results of a randomised, con-
trolled, open-label, platform trial. medRxiv https://doi.org/10.1101/2021
.02.11.21249258.
178. Rosas IO, Bräu N, Waters M, Go RC, Hunter BD, Bhagani S, Skiest D, Aziz
MS, Cooper N, Douglas IS, Savic S, Youngstein T, Del Sorbo L, Cubillo
Gracian A, De La Zerda DJ, Ustianowski A, Bao M, Dimonaco S, Graham
E, Matharu B, Spotswood H, Tsai L, Malhotra A. 2021. Tocilizumab in hos-
pitalized patients with severe Covid-19 pneumonia. N Engl J Med 384:
15031516. https://doi.org/10.1056/NEJMoa2028700.
179. Salama C, Han J, Yau L, Reiss WG, Kramer B, Neidhart JD, Criner GJ, Kaplan-
Lewis E, Baden R, Pandit L, Cameron ML, Garcia-Diaz J, Chávez V, Mekebeb-
Reuter M, Lima de Menezes F, Shah R, González-Lara MF, Assman B,
Freedman J, Mohan SV. 2021. Tocilizumab in patients hospitalized with
Covid-19 pneumonia. N Engl J Med 384:20230. https://doi.org/10.1056/
NEJMoa2030340.
180. Hoffman-La Roche. 2021. A phase III, randomized, double-blind, multi-
center study to evaluate the efcacy and safety of remdesivir plus tocili-
zumab compared with remdesivir plus placebo in hospitalized patients
with severe COVID-19 pneumonia. ClinicalTrials.gov https://clinicaltrials
.gov/ct2/show/NCT04409262.
181. Hoffman-La Roche. 2021. A randomized, double-blind, placebo-con-
trolled, multicenter study to evaluate the safety and efcacy of tocilizu-
mab in patients with severe COVID-19 pneumonia. ClinicalTrials.gov
https://clinicaltrials.gov/ct2/show/NCT04320615.
182. Genentech, Inc. 2021. A randomized, double-blind, placebo-controlled,
multicenter study to evaluate the efcacy and safety of tocilizumab in
hospitalized patients with COVID-19 pneumonia. ClinicalTrials.gov
https://clinicaltrials.gov/ct2/show/NCT04372186.
183. US Food and Drug Administration. 24 June 2021. Genentech tocilizumab
emergency use authorization 099 letter of authority. US Food and Drug
Administration, Silver Spring, MD. https://www.fda.gov/media/150319/
download.
184. Synairgen Research Ltd. 2021. A randomised double-blind placebo-con-
trolled trial to determine the safety and efcacy of inhaled SNG001 (IFN-
b
1a for nebulisation) for the treatment of patients with conrmed
SARS-CoV-2 infection. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/
NCT04385095.
185. Synairgen plc. 2020. Synairgen announces positive results from trial of
SNG001 in hospitalised COVID-19 patients. Synairgen plc, Southampton,
United Kingdom. http://synairgen.web01.hosting.bdci.co.uk/umbraco/Surface/
Download/GetFile?cid=1130026e-0983-4338-b648-4ac7928b9a37.
186. Monk PD, Marsden RJ, Tear VJ, Brookes J, Batten TN, Mankowski M,
Gabbay FJ, Davies DE, Holgate ST, Ho L-P, Clark T, Djukanovic R,
Wilkinson TMA, Crooks MG, Dosanjh DP, Siddiqui S, Rahman NM, Smith
JA, Horsley A, Harrison TW, Saralaya D, McGarvey L, Watson A, Foster E,
Fleet A, Singh D, Hemmings S, Aitken S, Dudley S, Beegan R, Thompson
A, Rodrigues PM. 2021. Safety and efcacy of inhaled nebulised inter-
feron beta-1a (SNG001) for treatment of SARS-CoV-2 infection: a rando-
mised, double-blind, placebo-controlled, phase 2 trial. Lancet Respir
Med 9:196206. https://doi.org/10.1016/S2213-2600(20)30511-7.
187. COVID-19 Review Consortium. 2021. Social factors inuencing COVID-19
exposure, and outcomes. https://greenelab.github.io/covid19-review/v/
32afa309f69f0466a91acec5d0df3151fe4d61b5/%23social-factors-inuencing-
covid-19-exposure-and-outcomes.
188. Chen L, Xiong J, Bao L, Shi Y. 2020. Convalescent plasma as a potential
therapy for COVID-19. Lancet Infect Dis 20:3982400. https://doi.org/10
.1016/S1473-3099(20)30141-9.
189. Roback JD, Guarner J. 2020. Convalescent plasma to treat COVID-19.
JAMA 323:156121562. https://doi.org/10.1001/jama.2020.4940.
190. Rajendran K, Krishnasamy N, Rangarajan J, Rathinam J, Natarajan M,
Ramachandran A. 2020. Convalescent plasma transfusion for the treatment
of COVID-19: systematic review. J Med Virol 92:147521483. https://doi.org/
10.1002/jmv.25961.
191. Janiaud P, Axfors C, Schmitt AM, Gloy V, Ebrahimi F, Hepprich M, Smith
ER, Haber NA, Khanna N, Moher D, Goodman SN, Ioannidis JPA,
Hemkens LG. 2021. Association of convalescent plasma treatment with
clinical outcomes in patients with COVID-19. JAMA 325:118521195.
https://doi.org/10.1001/jama.2021.2747.
192. Joyner MJ, Carter RE, Senefeld JW, Klassen SA, Mills JR, Johnson PW,
Theel ES, Wiggins CC, Bruno KA, Klompas AM, Lesser ER, Kunze KL,
Sexton MA, Diaz Soto JC, Baker SE, Shepherd JRA, van Helmond N,
Verdun NC, Marks P, van Buskirk CM, Winters JL, Stubbs JR, Rea RF,
Hodge DO, Herasevich V, Whelan ER, Clayburn AJ, Larson KF, Ripoll JG,
Andersen KJ, Buras MR, Vogt MNP, Dennis JJ, Regimbal RJ, Bauer PR,
Blair JE, Paneth NS, Fairweather D, Wright RS, Casadevall A. 2021. Conva-
lescent plasma antibody levels and the risk of death from Covid-19. N
Engl J Med 384:10151027. https://doi.org/10.1056/NEJMoa2031893.
193. RECOVERY Collaborative Group, Horby PW, Estcourt L, Peto L, Emberson
JR, Staplin N, Spata E, Pessoa-Amorim G, Campbell M, Roddick A,
Brunskill NE, ... Landray MJ. 2021. Convalescent plasma in patients
admitted to hospital with COVID-19 (RECOVERY): a randomised, con-
trolled, open-label, platform trial. medRxiv https://doi.org/10.1101/2021
.03.09.21252736.
194. Hsueh P-R, Huang L-M, Chen P-J, Kao C-L, Yang P-C. 2004. Chronological
evolution of IgM, IgA, IgG, and neutralisation antibodies after infection
with SARS-associated coronavirus. Clin Microbiol Infect 10:106221066.
https://doi.org/10.1111/j.1469-0691.2004.01009.x.
195. Nie Y, Wang G, Shi X, Zhang H, Qiu Y, He Z, Wang W, Lian G, Yin X, Du L,
Ren L, Wang J, He X, Li T, Deng H, Ding M. 2004. Neutralizing antibodies
in patients with severe acute respiratory syndrome-associated coronavi-
rus infection. J Infect Dis 190:111921126. https://doi.org/10.1086/
423286.
196. Prabakaran P, Zhu Z, Xiao X, Biragyn A, Dimitrov AS, Broder CC, Dimitrov
DS. 2009. Potent human monoclonal antibodies against SARS CoV,
Nipah and Hendra viruses. Expert Opin Biol Ther 9:3552368. https://doi
.org/10.1517/14712590902763755.
197. Hoffmann M, Kleine-Weber H, Schroeder S, Krüger N, Herrler T, Erichsen
S, Schiergens TS, Herrler G, Wu NH, Nitsche A, Müller MA, Drosten C,
Pöhlmann S. 2020. SARS-CoV-2 cell entry depends on ACE2 and
TMPRSS2 and is blocked by a clinically proven protease inhibitor. Cell
181:2712280.e8. https://doi.org/10.1016/j.cell.2020.02.052.
198. Walls AC, Park Y-J, Tortorici M, Wall A, McGuire AT, Veesler D. 2020.
Structure, function, and antigenicity of the SARS-CoV-2 spike glycopro-
tein. Cell 181:2812292.e6. https://doi.org/10.1016/j.cell.2020.02.058.
199. Sun C, Chen L, Yang J, Luo C, Zhang Y, Li J, Yang J, Zhang J, Xie L. 2020.
SARS-CoV-2 and SARS-CoV spike-RBD structure and receptor binding
comparison and potential implications on neutralizing antibody and vac-
cine development. bioRxiv https://doi.org/10.1101/2020.02.16.951723.
200. Renn A, Fu Y, Hu X, Hall MD, Simeonov A. 2020. Fruitful neutralizing anti-
body pipeline brings hope to defeat SARS-Cov-2. Trends Pharmacol Sci
41:8152829. https://doi.org/10.1016/j.tips.2020.07.004.
201. Pinto D, Park YJ, Beltramello M, Walls AC, Tortorici MA, Bianchi S, Jaconi
S, Culap K, Zatta F, De Marco A, Peter A, Guarino B, Spreaco R,
Cameroni E, Case JB, Chen RE, Havenar-Daughton C, Snell G, Telenti A,
Virgin HW, Lanzavecchia A, Diamond MS, Fink K, Veesler D, Corti D.
2020. Cross-neutralization of SARS-CoV-2 by a human monoclonal SARS-
CoV antibody. Nature 583:2902295. https://doi.org/10.1038/s41586-020
-2349-y.
202. Wrapp D, Wang N, Corbett KS, Goldsmith JA, Hsieh C-L, Abiona O,
Graham BS, McLellan JS. 2020. Cryo-EM structure of the 2019-nCoV spike
in the prefusion conformation. Science 367:126021263. https://doi.org/
10.1126/science.abb2507.
203. Wang C, Li W, Drabek D, Okba NM, van Haperen R, Osterhaus AD, van
Kuppeveld FJ, Haagmans BL, Grosveld F, Bosch B-J. 2020. A human
monoclonal antibody blocking SARS-CoV-2 infection. bioRxiv https://doi
.org/10.1101/2020.03.11.987958.
204. Deb P, Molla MA, Saif-Ur-Rahman KM. 2021. An update to monoclonal
antibody as therapeutic option against COVID-19. Biosaf Health 3:
87291. https://doi.org/10.1016/j.bsheal.2021.02.001.
205. Jones BE, Brown-Augsburger PL, Corbett KS, Westendorf K, Davies J,
Cujec TP, Wiethoff CM, Blackbourne JL, Heinz BA, Foster D, ... Falconer
E. 2020. LY-CoV555, a rapidly isolated potent neutralizing antibody, pro-
vides protection in a non-human primate model of SARS-CoV-2 infec-
tion. bioRxiv https://doi.org/10.1101/2020.09.30.318972.
206. Eli Lilly and Company. 2020. A randomized, placebo-controlled, double-
blind, sponsor unblinded, single ascending dose, phase 1 rst in human
study to evaluate the safety, tolerability, pharmacokinetics and pharmaco-
dynamics of intravenous LY3819253 in participants hospitalized for COVID-
19. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT04411628.
207. Eli Lilly and Company. 2020. A phase 1, randomized, placebo-controlled
study to evaluate the tolerability, safety, pharmacokinetics, and
Rando et al.
November/December 2021 Volume 6 Issue 6 e00233-21 msystems.asm.org 46
Downloaded from https://journals.asm.org/journal/msystems on 03 November 2021 by 2607:f470:34:2303:5040:4a48:5de1:cdb5.
immunogenicity of LY3832479 given as a single intravenous dose in
healthy participants. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/
NCT04441931.
208. Gottlieb RL, Nirula A, Chen P, Boscia J, Heller B, Morris J, Huhn G,
Cardona J, Mocherla B, Stosor V, Shawa I, Kumar P, Adams AC, Van
Naarden J, Custer KL, Durante M, Oakley G, Schade AE, Holzer TR, Ebert
PJ, Higgs RE, Kallewaard NL, Sabo J, Patel DR, Klekotka P, Shen L,
Skovronsky DM. 2021. Effect of bamlanivimab as monotherapy or in
combination with etesevimab on viral load in patients with mild to mod-
erate COVID-19: a randomized clinical trial. JAMA 325:6322644. https://
doi.org/10.1001/jama.2021.0202.
209. Eli Lilly and Company. 2021. A randomized, double-blind, placebo-con-
trolled, phase 2/3 study to evaluate the efcacy and safety of LY3819253
and LY3832479 in participants with mild to moderate COVID-19 illness
(BLAZE-1). ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT04427501.
210. Chen P, Nirula A, Heller B, Gottlieb RL, Boscia J, Morris J, Huhn G,
Cardona J, Mocherla B, Stosor V, Shawa I, Adams AC, Van Naarden J,
Custer KL, Shen L, Durante M, Oakley G, Schade AE, Sabo J, Patel DR,
Klekotka P, Skovronsky DM, BLAZE-1 Investigators. 2021. SARS-CoV-2
neutralizing antibody LY-CoV555 in outpatients with Covid-19. N Engl J
Med 384:2292237. https://doi.org/10.1056/NEJMoa2029849.
211. US Food and Drug Administration. 16 September 2021. Bamlanivimab
and etesevimab emergency use authorization 094 letter of authoriza-
tion. US Food and Drug Administration, Silver Spring, MD. https://www
.fda.gov/media/145801/download.
212. Hansen J, Baum A, Pascal KE, Russo V, Giordano S, Wloga E, Fulton BO,
Yan Y, Koon K, Patel K, Chung KM, Hermann A, Ullman E, Cruz J, Raque
A, Huang T, Fairhurst J, Libertiny C, Malbec M, Lee WY, Welsh R, Farr G,
Pennington S, Deshpande D, Cheng J, Watty A, Bouffard P, Babb R,
Levenkova N, Chen C, Zhang B, Romero Hernandez A, Saotome K, Zhou
Y, Franklin M, Sivapalasingam S, Lye DC, Weston S, Logue J, Haupt R,
Frieman M, Chen G, Olson W, Murphy AJ, Stahl N, Yancopoulos GD,
Kyratsous CA. 2020. Studies in humanized mice and convalescent
humans yield a SARS-CoV-2 antibody cocktail. Science 369:101021014.
https://doi.org/10.1126/science.abd0827.
213. Regeneron Pharmaceuticals. 2021. A master protocol assessing the
safety, tolerability, and efcacy of anti-spike (S) SARS-CoV-2 monoclonal
antibodies for the treatment of hospitalized patients with COVID-19.
ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT04426695.
214. Regeneron Pharmaceuticals. 2021. A master protocol assessing the
safety, tolerability, and efcacy of anti-spike (S) SARS-CoV-2 monoclonal
antibodies for the treatment of ambulatory patients with COVID-19.
ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT04425629.
215. Weinreich DM, Sivapalasingam S, Norton T, Ali S, Gao H, Bhore R, Musser
BJ, Soo Y, Rofail D, Im J, Perry C, Pan C, Hosain R, Mahmood A, Davis JD,
Turner KC, Hooper AT, Hamilton JD, Baum A, Kyratsous CA, Kim Y, Cook
A, Kampman W, Kohli A, Sachdeva Y, Graber X, Kowal B, DiCioccio T,
Stahl N, Lipsich L, Braunstein N, Herman G, Yancopoulos GD, Trial Inves-
tigators. 2021. REGN-COV2, a neutralizing antibody cocktail, in outpa-
tients with Covid-19. N Engl J Med 384:2382251. https://doi.org/10
.1056/NEJMoa2035002.
216. US Food and Drug Administration. 21 November 2020. Coronavirus
(COVID) update: fDA authorizes monoclonal antibodies for treatment of
COVID-19. US Food and Drug Administration, Silver Spring, MD. https://
www.fda.gov/news-events/press-announcements/coronavirus-covid-19
-update-fda-authorizes-monoclonal-antibodies-treatment-covid-19.
217. Traggiai E, Becker S, Subbarao K, Kolesnikova L, Uematsu Y, Gismondo
MR, Murphy BR, Rappuoli R, Lanzavecchia A. 2004. An efcient method
to make human monoclonal antibodies from memory B cells: potent
neutralization of SARS coronavirus. Nat Med 10:8712875. https://doi
.org/10.1038/nm1080.
218. Dolgin E. 2021. Super-antibodiescould curb COVID-19 and help avert
future pandemics. Nat Biotechnol 39:7832785. https://doi.org/10.1038/
s41587-021-00980-x.
219. Walker LM, Burton DR. 2018. Passive immunotherapy of viral infections:
'super-antibodies' enter the fray. Nat Rev Immunol 18:2972308. https://
doi.org/10.1038/nri.2017.148.
220. Vir Biotechnology, Inc. 2021. A randomized, multi-center, double-blind,
placebo-controlled study to assess the safety and efcacy of monoclonal
antibody VIR-7831 for the early treatment of coronavirus disease 2019
(COVID) in non-hospitalized patients. ClinicalTrials.gov https://clinicaltrials
.gov/ct2/show/NCT04545060.
221. Gupta A, Gonzalez-Rojas Y, Juarez E, Crespo Casal M, Moya J, Falci DR,
Sarkis E, Solis J, Zheng H, Scott N, ... COMET-ICE Investigators. 2021.
Early Covid-19 treatment with SARS-CoV-2 neutralizing antibody sotro-
vimab. medRxiv https://doi.org/10.1101/2021.05.27.21257096.
222. Liu Z, VanBlargan LA, Bloyet LM, Rothlauf PW, Chen RE, Stumpf S, Zhao
H, Errico JM, Theel ES, Liebeskind MJ, Alford B, Buchser WJ, Ellebedy AH,
Fremont DH, Diamond MS, Whelan SPJ. 2021. Identication of SARS-
CoV-2 spike mutations that attenuate monoclonal and serum antibody
neutralization. Cell Host Microbe 29:4772488.e4. https://doi.org/10
.1016/j.chom.2021.01.014.
223. Diamond M, Chen R, Xie X, Case J, Zhang X, VanBlargan L, Liu Y, Liu J, Errico
J, Winkler E, ... Gilchuk P. 2021. SARS-CoV-2 variants show resistance to
neutralization by many monoclonal and serum-derived polyclonal antibod-
ies. Research Square https://doi.org/10.21203/rs.3.rs-228079/v1.
224. Graham C, Seow J, Huettner I, Khan H, Kouphou N, Acors S, Winstone H,
Pickering S, Galao RP, Lista MJ, ... Doores KJ. 2021. Impact of the B.1.1.7
variant on neutralizing monoclonal antibodies recognizing diverse epi-
topes on SAR-CoV-2 spike. bioRxiv https://doi.org/10.1101/2021.02.03
.429355.
225. Wang P, Nair MS, Liu L, Iketani S, Luo Y, Guo Y, Wang M, Yu J, Zhang B,
Kwong PD, ...Ho DD. 2021. Antibody resistance of SARS-CoV-2 variants
B.1.351 and B.1.1.7. bioRxiv https://doi.org/10.1101/2021.01.25.428137.
226. US Department of Health and Human Services. 2021. Pause in the distri-
bution of bamlanivimab/etesevimab. US Department of Health and
Human Services, Washington, DC. https://www.phe.gov/emergency/
events/COVID19/investigation-MCM/Bamlanivimab-etesevimab/Pages/
bamlanivimab-etesevimab-distribution-pause.aspx.
227. Sun Z-YJ, Oh KJ, Kim M, Yu J, Brusic V, Song L, Qiao Z, Wang J-h, Wagner
G, Reinherz EL. 2008. HIV-1 broadly neutralizing antibody extracts its epi-
tope from a kinked gp41 ectodomain region on the viral membrane. Im-
munity 28:52263. https://doi.org/10.1016/j.immuni.2007.11.018.
228. Ekiert DC, Bhabha G, Elsliger M-A, Friesen RHE, Jongeneelen M, Throsby
M, Goudsmit J, Wilson IA. 2009. Antibody recognition of a highly con-
served inuenza virus epitope. Science 324:2462251. https://doi.org/10
.1126/science.1171491.
229. He Y, Li J, Du L, Yan X, Hu G, Zhou Y, Jiang S. 2006. Identication and
characterization of novel neutralizing epitopes in the receptor-binding
domain of SARS-CoV spike protein: revealing the critical antigenic deter-
minants in inactivated SARS-CoV vaccine. Vaccine 24:549825508.
https://doi.org/10.1016/j.vaccine.2006.04.054.
230. Rockx B, Donaldson E, Frieman M, Sheahan T, Corti D, Lanzavecchia A,
Baric RS. 2010. Escape from human monoclonal antibody neutralization
affects in vitro and in vivo tness of severe acute respiratory syndrome
coronavirus. J Infect Dis 201:9462955. https://doi.org/10.1086/651022.
231. Stanford University. 2021. Stanford Coronavirus Antiviral & Resistance
Database (CoVDB). Stanford University, Stanford, CA. https://covdb
.stanford.edu/page/susceptibility-data.
232. Rappazzo CG, Tse LV, Kaku CI, Wrapp D, Sakharkar M, Huang D, Deveau
LM, Yockachonis TJ, Herbert AS, Battles MB, O'Brien CM, Brown ME,
Geoghegan JC, Belk J, Peng L, Yang L, Hou Y, Scobey TD, Burton DR,
Nemazee D, Dye JM, Voss JE, Gunn BM, McLellan JS, Baric RS, Gralinski
LE, Walker LM. 2021. Broad and potent activity against SARS-like viruses
by an engineered human monoclonal antibody. Science 371:8232829.
https://doi.org/10.1126/science.abf4830.
233. Parvathaneni V, Kulkarni NS, Muth A, Gupta V. 2019. Drug repurposing: a
promising tool to accelerate the drug discovery process. Drug Discov
Today 24:207622085. https://doi.org/10.1016/j.drudis.2019.06.014.
234. Zheng W, Sun W, Simeonov A. 2018. Drug repurposing screens and syn-
ergistic drug-combinations for infectious diseases. Br J Pharmacol 175:
1812191. https://doi.org/10.1111/bph.13895.
235. Muratov EN, Amaro R, Andrade CH, Brown N, Ekins S, Fourches D, Isayev
O, Kozakov D, Medina-Franco JL, Merz KM, Oprea TI, Poroikov V,
Schneider G, Todd MH, Varnek A, Winkler DA, Zakharov AV, Cherkasov A,
Tropsha A. 2021. A critical overview of computational approaches
employed for COVID-19 drug discovery. Chem Soc Rev 50:912129151.
https://doi.org/10.1039/d0cs01065k.
236. Law G, Tisoncik-Go J, Korth MJ, Katze MG. 2013. Drug repurposing: a bet-
ter approach for infectious disease drug discovery? Curr Opin Immunol
25:5882592. https://doi.org/10.1016/j.coi.2013.08.004.
237. Pereira DA, Williams JA. 2007. Origin and evolution of high throughput
screening. Br J Pharmacol 152:53261. https://doi.org/10.1038/sj.bjp
.0707373.
238. Vincent F, Loria P, Pregel M, Stanton R, Kitching L, Nocka K, Doyonnas R,
Steppan C, Gilbert A, Schroeter T, Peakman M-C. 2015. Developing pre-
dictive assays: the phenotypic screening rule of 3.Sci Transl Med 7:
293ps15. https://doi.org/10.1126/scitranslmed.aab1201.
ID and Development of Therapeutics for COVID-19
November/December 2021 Volume 6 Issue 6 e00233-21 msystems.asm.org 47
Downloaded from https://journals.asm.org/journal/msystems on 03 November 2021 by 2607:f470:34:2303:5040:4a48:5de1:cdb5.
239. Swinney DC. 2013. Phenotypic vs. target-based drug discovery for rst-
in-class medicines. Clin Pharmacol Ther 93:2992301. https://doi.org/10
.1038/clpt.2012.236.
240. Moffat JG, Rudolph J, Bailey D. 2014. Phenotypic screening in cancer
drug discovery past, present and future. Nat Rev Drug Discov 13:
5882602. https://doi.org/10.1038/nrd4366.
241. ChemBridge Corp. 2021. Screening Libraries, Diversity Libraries, DIVER-
Set Diverse Screening Libraries. ChemBridge Corp, San Diego, CA.
https://www.chembridge.com/screening_libraries/diversity_libraries/.
242. Wagner BK, Schreiber SL. 2016. The power of sophisticated phenotypic
screening and modern mechanism-of-action methods. Cell Chem Biol
23:329. https://doi.org/10.1016/j.chembiol.2015.11.008.
243. Mercorelli B, Palù G, Loregian A. 2018. Drug repurposing for viral infec-
tious diseases: how far are we? Trends Microbiol 26:8652876. https://
doi.org/10.1016/j.tim.2018.04.004.
244. Challa AP, Lavieri RR, Lewis JT, Zaleski NM, Shirey-Rice JK, Harris PA,
Aronoff DM, Pulley JM. 2019. Systematically prioritizing candidates in
genome-based drug repurposing. Assay Drug Dev Technol 17:3522363.
https://doi.org/10.1089/adt.2019.950.
245. Edwards A. 2020. What are the odds of nding a COVID-19 drug from a
lab repurposing screen? J Chem Inf Model 60:572725729. https://doi
.org/10.1021/acs.jcim.0c00861.
246. Kido H, Okumura Y, Yamada H, Le TQ, Yano M. 2007. Proteases essential
for human inuenza virus entry into cells and their inhibitors as poten-
tial therapeutic agents. Curr Pharm Des 13:4052414. https://doi.org/10
.2174/138161207780162971.
247. Zhou Y, Vedantham P, Lu K, Agudelo J, Carrion R, Nunneley JW, Barnard
D, Pöhlmann S, McKerrow JH, Renslo AR, Simmons G. 2015. Protease
inhibitors targeting coronavirus and lovirus entry. Antiviral Res 116:
76284. https://doi.org/10.1016/j.antiviral.2015.01.011.
248. Jin Z, Du X, Xu Y, Deng Y, Liu M, Zhao Y, Zhang B, Li X, Zhang L, Peng C,
Duan Y, Yu J, Wang L, Yang K, Liu F, Jiang R, Yang X, You T, Liu X, Yang X,
Bai F, Liu H, Liu X, Guddat LW, Xu W, Xiao G, Qin C, Shi Z, Jiang H, Rao Z,
Yang H. 2020. Structure of Mpro from SARS-CoV-2 and discovery of its
inhibitors. Nature 582:2892293. https://doi.org/10.1038/s41586-020
-2223-y.
249. Yang H, Xie W, Xue X, Yang K, Ma J, Liang W, Zhao Q, Zhou Z, Pei D,
Ziebuhr J, Hilgenfeld R, Yuen KY, Wong L, Gao G, Chen S, Chen Z, Ma D,
Bartlam M, Rao Z. 2005. Design of wide-spectrum inhibitors targeting
coronavirus main proteases. PLoS Biol 3:e324. https://doi.org/10.1371/
journal.pbio.0030324.
250. Ren Z, Yan L, Zhang N, Guo Y, Yang C, Lou Z, Rao Z. 2013. The newly
emerged SARS-like coronavirus HCoV-EMC also has an Achillesheel:
current effective inhibitor targeting a 3C-like protease. Protein Cell 4:
2482250. https://doi.org/10.1007/s13238-013-2841-3.
251. Wang F, Chen C, Tan W, Yang K, Yang H. 2016. Structure of main protease
from human coronavirus NL63: insights for wide spectrum anti-coronavi-
rus drug design. Sci Rep 6:22677. https://doi.org/10.1038/srep22677.
252. Xue X, Yu H, Yang H, Xue F, Wu Z, Shen W, Li J, Zhou Z, Ding Y, Zhao Q,
Zhang XC, Liao M, Bartlam M, Rao Z. 2008. Structures of two coronavirus
main proteases: implications for substrate binding and antiviral drug
design. J Virol 82:251522527. https://doi.org/10.1128/JVI.02114-07.
253. Azad GK, Tomar RS. 2014. Ebselen, a promising antioxidant drug: mecha-
nisms of action and targets of biological pathways. Mol Biol Rep 41:
486524879. https://doi.org/10.1007/s11033-014-3417-x.
254. Menéndez CA, Byléhn F, Perez-Lemus GR, Alvarado W, de Pablo JJ. 2020.
Molecular characterization of ebselen binding activity to SARS-CoV-2
main protease. Sci Adv 6:eabd0345. https://doi.org/10.1126/sciadv
.abd0345.
255. Chen Z, Jiang Z, Chen N, Shi Q, Tong L, Kong F, Cheng X, Chen H, Wang
C, Tang B. 2018. Target discovery of ebselen with a biotinylated probe.
Chem Commun 54:950629509. https://doi.org/10.1039/c8cc04258f.
256. Contract Pharma. 31 August 2020. FDA clears SPIs ebselen for phase II
COVID-19 trials. Contract Pharma Publishing, Montvale, NJ. https://www
.contractpharma.com/contents/view_breaking-news/2020-08-31/fda-clears
-spis-ebselen-for-phase-ii-covid-19-trials/.
257. Sound Pharmaceuticals, Inc. 2021. A phase 2, randomized, double-blind,
placebo-controlled, dose escalation study to evaluate the safety and ef-
cacy of SPI-1005 in moderate COVID-19 patients. ClinicalTrials.gov
https://clinicaltrials.gov/ct2/show/NCT04484025.
258. Sound Pharmaceuticals, Inc. 2021. A phase 2, randomized, double-blind,
placebo-controlled, dose escalation study to evaluate the safety and ef-
cacy of SPI-1005 in severe COVID-19 patients. ClinicalTrials.gov https://
clinicaltrials.gov/ct2/show/NCT04483973.
259. Pzer. 2021. A phase 1b, 2-part, double-blind, placebo-controlled, sponsor-
open study, to evaluate the safety, tolerability and pharmacokinetics of
single ascending (24-hour, part 1) and multiple ascending (120-hour, part 2)
intravenous infusions of PF-07304814 in hospitalized participants with
covid-19. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT04535167.
260. Pzer. 2021. An interventional efcacy and safety, phase 2/3, double-blind,
2-arm study to investigate orally administered PF-07321332/ritonavir com-
pared with placebo in nonhospitalized symptomatic adult participants
with COVID-19 who are at increased risk of progressing to severe illness.
ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT04960202.
261. Qian T, Zhu S, Hoshida Y. 2019. Use of big data in drug development for
precision medicine: an update. Expert Rev Precis Med Drug Dev 4:
1892200. https://doi.org/10.1080/23808993.2019.1617632.
262. Kuleshov MV, Stein DJ, Clarke DJB, Kropiwnicki E, Jagodnik KM, Bartal A,
Evangelista JE, Hom J, Cheng M, Bailey A, Zhou A, Ferguson LB, Lachmann
A, Maayan A. 2020. The COVID-19 drug and gene set library. Patterns (N
Y) 1:100090. https://doi.org/10.1016/j.patter.2020.100090.
263. Sadegh S, Matschinske J, Blumenthal DB, Galindez G, Kacprowski T, List
M, Nasirigerdeh R, Oubounyt M, Pichlmair A, Rose TD, Salgado-Albarrán
M, Späth J, Stukalov A, Wenke NK, Yuan K, Pauling JK, Baumbach J. 2020.
Exploring the SARS-CoV-2 virus-host-drug interactome for drug repur-
posing. Nat Commun 11:3518. https://doi.org/10.1038/s41467-020
-17189-2.
264. Zhou Y, Wang F, Tang J, Nussinov R, Cheng F. 2020. Articial intelligence
in COVID-19 drug repurposing. Lancet Digit Health 2:e6672e676.
https://doi.org/10.1016/S2589-7500(20)30192-8.
265. Gordon DE, Jang GM, Bouhaddou M, Xu J, Obernier K, White KM,
O'Meara MJ, Rezelj VV, Guo JZ, Swaney DL, Tummino TA, Hüttenhain R,
Kaake RM, Richards AL, Tutuncuoglu B, Foussard H, Batra J, Haas K,
Modak M, Kim M, Haas P, Polacco BJ, Braberg H, Fabius JM, Eckhardt M,
Soucheray M, Bennett MJ, Cakir M, McGregor MJ, Li Q, Meyer B, Roesch
F, Vallet T, Mac Kain A, Miorin L, Moreno E, Naing ZZC, Zhou Y, Peng S,
Shi Y, Zhang Z, Shen W, Kirby IT, Melnyk JE, Chorba JS, Lou K, Dai SA,
Barrio-Hernandez I, Memon D, Hernandez-Armenta C, Lyu J, et al. 2020.
A SARS-CoV-2 protein interaction map reveals targets for drug repurpos-
ing. Nature 583:4592468. https://doi.org/10.1038/s41586-020-2286-9.
266. Maurice T, Su T-P. 2009. The pharmacology of sigma-1 receptors. Phar-
macol Ther 124:1952206. https://doi.org/10.1016/j.pharmthera.2009.07
.001.
267. Gordon DE, Hiatt J, Bouhaddou M, Rezelj VV, Ulferts S, Braberg H, Jureka
AS, Obernier K, Guo JZ, Batra J, Kaake RM, Weckstein AR, Owens TW,
Gupta M, Pourmal S, Titus EW, Cakir M, Soucheray M, McGregor M, Cakir
Z, Jang G, O'Meara MJ, Tummino TA, Zhang Z, Foussard H, Rojc A, Zhou
Y, Kuchenov D, Hüttenhain R, Xu J, Eckhardt M, Swaney DL, Fabius JM,
Ummadi M, Tutuncuoglu B, Rathore U, Modak M, Haas P, Haas KM,
Naing ZZC, Pulido EH, Shi Y, Barrio-Hernandez I, Memon D, Petsalaki E,
Dunham A, Marrero MC, Burke D, Koh C, Vallet T, Silvas JA, et al. 2020.
Comparative host-coronavirus protein interaction networks reveal pan-
viral disease mechanisms. Science 370:eabe9403. https://doi.org/10
.1126/science.abe9403.
268. Vela JM. 2020. Repurposing sigma-1 receptor ligands for COVID-19 ther-
apy? Front Pharmacol 11:582310. https://doi.org/10.3389/fphar.2020
.582310.
269. Hayashi T, Su T. 2005. The sigma receptor: evolution of the concept in
neuropsychopharmacology. Curr Neuropharmacol 3:2672280. https://
doi.org/10.2174/157015905774322516.
270. Tummino TA, Rezelj VV, Fischer B, Fischer A, O'Meara MJ, Monel B, Vallet
T, White KM, Zhang Z, Alon A, Schadt H, O'Donnell HR, Lyu J, Rosales R,
McGovern BL, Rathnasinghe R, Jangra S, Schotsaert M, Galarneau JR,
Krogan NJ, Urban L, Shokat KM, Kruse AC, García-Sastre A, Schwartz O,
Moretti F, Vignuzzi M, Pognan F, Shoichet BK. 2021. Drug-induced phos-
pholipidosis confounds drug repurposing for SARS-CoV-2. Science 373:
5412547. https://doi.org/10.1126/science.abi4708.
271. Breiden B, Sandhoff K. 2019. Emerging mechanisms of drug-induced
phospholipidosis. Biol Chem 401:31246. https://doi.org/10.1515/hsz
-2019-0270.
272. Obeidat M, Isaacson AL, Chen SJ, Ivanovic M, Holanda D. 2020. Zebra-
like bodies in COVID-19: is phospholipidosis evidence of hydroxychloro-
quine induced acute kidney injury? Ultrastruct Pathol 44:5192523.
https://doi.org/10.1080/01913123.2020.1850966.
273. Dittmar M, Lee JS, Whig K, Segrist E, Li M, Kamalia B, Castellana L,
Ayyanathan K, Cardenas-Diaz FL, Morrisey EE, Truitt R, Yang W, Jurado K,
Samby K, Ramage H, Schultz DC, Cherry S. 2021. Drug repurposing
screens reveal cell-type-specic entry pathways and FDA-approved
Rando et al.
November/December 2021 Volume 6 Issue 6 e00233-21 msystems.asm.org 48
Downloaded from https://journals.asm.org/journal/msystems on 03 November 2021 by 2607:f470:34:2303:5040:4a48:5de1:cdb5.
drugs active against SARS-Cov-2. Cell Rep 35:108959. https://doi.org/10
.1016/j.celrep.2021.108959.
274. Edwards A, Hartung IV. 2021. No shortcuts to SARS-CoV-2 antivirals. Sci-
ence 373:4882489. https://doi.org/10.1126/science.abj9488.
275. Hashimoto K. 2021. Repurposing of CNS drugs to treat COVID-19 infec-
tion: targeting the sigma-1 receptor. Eur Arch Psychiatry Clin Neurosci
271:2492258. https://doi.org/10.1007/s00406-020-01231-x.
276. Lenze EJ, Mattar C, Zorumski CF, Stevens A, Schweiger J, Nicol GE, Miller
J, Yang L, Yingling M, Avidan MS, Reiersen AM. 2020. Fluvoxamine vs
placebo and clinical deterioration in outpatients with symptomatic
COVID-19. JAMA 324:229222300. https://doi.org/10.1001/jama.2020
.22760.
277. Seftel D, Boulware DR. 2021. Prospective cohort of uvoxamine for early
treatment of coronavirus disease 19. Open Forum Infect Dis 8:ofab050.
https://doi.org/10.1093/od/ofab050.
278. Rosen DA, Seki SM, Fernández-Castañeda A, Beiter RM, Eccles JD,
Woodfolk JA, Gaultier A. 2019. Modulation of the sigma-1 receptorIRE1
pathway is benecial in preclinical models of inammation and sepsis. Sci
Transl Med 11:eaau5266. https://doi.org/10.1126/scitranslmed.aau5266.
279. Sukhatme VP, Reiersen AM, Vayttaden SJ, Sukhatme VV. 2021. Fluvox-
amine: a review of its mechanism of action and its role in COVID-19.
Front Pharmacol 12:652688. https://doi.org/10.3389/fphar.2021.652688.
280. Lowe D. 19 July 2021. Too many papers. In the Pipeline. https://blogs
.sciencemag.org/pipeline/archives/2021/07/19/too-many-papers.
281. Prinz F, Schlange T, Asadullah K. 2011. Believe it or not: how much can
we rely on published data on potential drug targets? Nat Rev Drug Dis-
cov 10:712. https://doi.org/10.1038/nrd3439-c1.
282. Swinney DC, Anthony J. 2011. How were new medicines discovered?
Nat Rev Drug Discov 10:5072519. https://doi.org/10.1038/nrd3480.
283. Kupferschmidt K. 2020. Big studies dim hopes for hydroxychloroquine.
Science 368:11661167. https://doi.org/10.1126/science.368.6496.1166.
284. Bugin K, Woodcock J. 2021. Trends in COVID-19 therapeutic clinical tri-
als. Nat Rev Drug Discov 20:2542255. https://doi.org/10.1038/d41573
-021-00037-3.
285. Dron L, Dillman A, Zoratti MJ, Haggstrom J, Mills EJ, Park JJ. 2021. Clinical
trial data sharing for COVID-19related research. J Med Internet Res 23:
e26718. https://doi.org/10.2196/26718.
286. Lee Z, Rayner CR, Forrest JI, Nachega JB, Senchaudhuri E, Mills EJ. 2021.
The rise and fall of hydroxychloroquine for the treatment and preven-
tion of COVID-19. Am J Trop Med Hyg 104:35238. https://doi.org/10
.4269/ajtmh.20-1320.
287. Park JJ, Dron L, Mills EJ. 2021. Moving forward in clinical research with
master protocols. Contemp Clin Trials 106:106438. https://doi.org/10
.1016/j.cct.2021.106438.
288. Diamond. 5 May 2020. Main protease structure and XChem fragment
screen. https://www.diamond.ac.uk/covid-19/for-scientists/Main-protease
-structure-and-XChem.html.
289. Barnes MA, Carson MJ, Nair MG. 2015. Non-traditional cytokines: how
catecholamines and adipokines inuence macrophages in immunity,
metabolism and the central nervous system. Cytokine 72:210219.
https://doi.org/10.1016/j.cyto.2015.01.008.
290. Elenkov IJ, Chrousos GP. 2002. Stress hormones, proinammatory and
antiinammatory cytokines, and autoimmunity. Ann N Y Acad Sci 966:
290303. https://doi.org/10.1111/j.1749-6632.2002.tb04229.x.
291. García A, Martí O, Vallès A, Dal-Zotto S, Armario A. 2000. Recovery of the
hypothalamic-pituitary-adrenal response to stress. Effect of stress inten-
sity, stress duration and previous stress exposure. Neuroendocrinology
72:114125. https://doi.org/10.1159/000054578.
292. Elenkov IJ, Papanicolaou DA, Wilder RL, Chrousos GP. 1996. Modulatory
effects of glucocorticoids and catecholamines on human interleukin-12
and interleukin-10 production: clinical implications. Proc Assoc Am Physi-
cians 108:374381. https://www.ncbi.nlm.nih.gov/pubmed/8902882.
293. Theoharides TC. 2020. Dexamethasone for COVID-19? Not so fast. J Biol
Regul Homeost Agents 34:12411243. https://doi.org/10.23812/20
-EDITORIAL_1-5.
294. Matthay MA, Thompson B. 2020. Dexamethasone in hospitalised
patients with COVID-19: addressing uncertainties. Lancet Respir Med 8:
11701172. https://doi.org/10.1016/S2213-2600(20)30503-8.
295. Brotherton H, Usuf E, Nadjm B, Forrest K, Bojang K, Samateh AL, Bittaye M,
Roberts CA, dAlessandro U, Roca A. 2020. Dexamethasone for COVID-19:
data needed from randomised clinical trials in Africa. Lancet Glob Health
8:e1125e1126. https://doi.org/10.1016/S2214-109X(20)30318-1.
296. Cai Q, Yang M, Liu D, Chen J, Shu D, Xia J, Liao X, Gu Y, Cai Q, Yang Y,
Shen C, Li X, Peng L, Huang D, Zhang J, Zhang S, Wang F, Liu J, Chen L,
Chen S, Wang Z, Zhang Z, Cao R, Zhong W, Liu Y, Liu L. 2020. Experimen-
tal treatment with favipiravir for COVID-19: an open-label control study.
Engineering 6:11921198. https://doi.org/10.1016/j.eng.2020.03.007.
297. Horby PW, Mafham M, Bell JL, Linsell L, Staplin N, Emberson J,
Palfreeman A, Raw J, Elmahi E, Prudon B, Green C, Carley S, Chadwick D,
Davies M, Wise MP, Baillie JK, Chappell LC, Faust SN, Jaki T, Jefferey K,
Lim WS, Montgomery A, Rowan K, Juszczak E, Haynes R, Landray MJ.
2020. Lopinavirritonavir in patients admitted to hospital with COVID-
19 (RECOVERY): a randomised, controlled, open-label, platform trial. Lan-
cet 396:13451352. https://doi.org/10.1016/S0140-6736(20)32013-4.
298. Harrington DP, Baden LR, Hogan JW. 2020. A large, simple trial leading
to complex questions. N Engl J Med 384:576577. https://doi.org/10
.1056/nejme2034294.
299. Retraction Watch. 29 April 2020. Retracted coronavirus (COVID) papers.
https://retractionwatch.com/retracted-coronavirus-covid-19-papers/.
300. Lou Y, Liu L, Yao H, Hu X, Su J, Xu K, Luo R, Yang X, He L, Lu X, Zhao Q,
Liang T, Qiu Y. 2021. Clinical outcomes and plasma concentrations of
baloxavir marboxil and favipiravir in COVID-19 patients: an exploratory
randomized, controlled trial. Eur J Pharm Sci 157:105631. https://doi
.org/10.1016/j.ejps.2020.105631.
301. Dabbous HM, Abd-Elsalam S, El-Sayed MH, Sherief AF, Ebeid FFS, El
Ghafar MSA, Soliman S, Elbahnasawy M, Badawi R, Tageldin MA. 2021.
Efcacy of favipiravir in COVID-19 treatment: a multi-center randomized
study. Arch Virol 166:949954. https://doi.org/10.1007/s00705-021
-04956-9.
302. Ivashchenko AA, Dmitriev KA, Vostokova NV, Azarova VN, Blinow AA,
Egorova AN, Gordeev IG, Ilin AP, Karapetian RN, Kravchenko DV,
Lomakin NV, Merkulova EA, Papazova NA, Pavlikova EP, Savchuk NP,
Simakina EN, Sitdekov TA, Smolyarchuk EA, Tikhomolova EG, Yakubova
EV, Ivachtchenko AV. 2020. AVIFAVIR for treatment of patients with
moderate coronavirus disease 2019 (COVID): interim results of a phase
II/III multicenter randomized clinical trial. Clin Infect Dis 73:531534.
https://doi.org/10.1093/cid/ciaa1176.
303. Pilkington V, Pepperrell T, Hill A. 2020. A review of the safety of favipira-
vir a potential treatment in the COVID-19 pandemic? J Virus Erad 6:
4551. https://doi.org/10.1016/S2055-6640(20)30016-9.
304. Mulangu S, Dodd LE, Davey RT Jr, Tshiani Mbaya O, Proschan M, Mukadi
D, Lusakibanza Manzo M, Nzolo D, Tshomba Oloma A, Ibanda A, Ali R,
Coulibaly S, Levine AC, Grais R, Diaz J, Lane HC, Muyembe-Tamfum
JJ; PALM, Writing Group, Sivahera B, Camara M, Kojan R, Walker R,
Dighero-Kemp B, Cao H, Mukumbayi P, Mbala-Kingebeni P, Ahuka S,
Albert S, Bonnett T, Crozier I, Duvenhage M, Proftt C, Teitelbaum M,
Moench T, Aboulhab J, Barrett K, Cahill K, Cone K, Eckes R, Hensley L,
Herpin B, Higgs E, Ledgerwood J, Pierson J, Smolskis M, Sow Y, Tierney J,
Sivapalasingam S, Holman W, Gettinger N, Vallée D, Nordwall J, PALM
Consortium Study Team. 2019. A randomized, controlled trial of Ebola
virus disease therapeutics. N Engl J Med 381:22932303. https://doi.org/
10.1056/NEJMoa1910993.
305. Sheahan TP, Sims AC, Graham RL, Menachery VD, Gralinski LE, Case JB,
Leist SR, Pyrc K, Feng JY, Trantcheva I, Bannister R, Park Y, Babusis D,
Clarke MO, Mackman RL, Spahn JE, Palmiotti CA, Siegel D, Ray AS, Cihlar
T, Jordan R, Denison MR, Baric RS. 2017. Broad-spectrum antiviral GS-
5734 inhibits both epidemic and zoonotic coronaviruses. Sci Transl Med
9:eaal3653. https://doi.org/10.1126/scitranslmed.aal3653.
306. Cohen J. 13 March 2020. Did an experimental drug help a U.S. coronavi-
rus patient? Science https://doi.org/10.1126/science.abb7243.
307. COVID-19 Investigation Team, Kujawski SA, Wong KK, Collins JP, Epstein
L, Killerby ME, Midgley CM, Abedi GR, Ahmed N, Almendares O, Alvarez
FN, ... Zhang J. 2020. First 12 patients with coronavirus disease 2019
(COVID) in the United States. medRxiv https://doi.org/10.1101/2020.03
.09.20032896.
308. Holshue ML, DeBolt C, Lindquist S, Lofy KH, Wiesman J, Bruce H, Spitters
C, Ericson K, Wilkerson S, Tural A, Diaz G, Cohn A, Fox L, Patel A, Gerber
SI, Kim L, Tong S, Lu X, Lindstrom S, Pallansch MA, Weldon WC, Biggs
HM, Uyeki TM, Pillai SK, Washington State 2019-nCoV Case Investigation
Team. 2020. First case of 2019 novel coronavirus in the United States. N
Engl J Med 382:929936. https://doi.org/10.1056/NEJMoa2001191.
309. Goldman JD, Lye DCB, Hui DS, Marks KM, Bruno R, Montejano R, Spinner
CD, Galli M, Ahn MY, Nahass RG, Chen YS, SenGupta D, Hyland RH,
Osinusi AO, Cao H, Blair C, Wei X, Gaggar A, Brainard DM, Towner WJ,
Muñoz J, Mullane KM, Marty FM, Tashima KT, Diaz G, Subramanian A,
GS-US-540-5773 Investigators. 2020. Remdesivir for 5 or 10 days in
patients with severe covid-19. N Engl J Med 383:18271837. https://doi
.org/10.1056/NEJMoa2015301.
ID and Development of Therapeutics for COVID-19
November/December 2021 Volume 6 Issue 6 e00233-21 msystems.asm.org 49
Downloaded from https://journals.asm.org/journal/msystems on 03 November 2021 by 2607:f470:34:2303:5040:4a48:5de1:cdb5.
310. US Food and Drug Administration. 22 October 2020. Remdesivir EUA
Letter of Authorization. US Food and Drug Administration, Silver Spring,
MD. https://www.fda.gov/media/137564/download.
311. Gilead Sciences. 2021. A phase 3 randomized study to evaluate the
safety and antiviral activity of remdesivir (GS-5734
TM
) in participants
with moderate COVID-19 compared to standard of care treatment. Clini-
calTrials.gov https://clinicaltrials.gov/ct2/show/NCT04292730.
312. EU Clinical Trials Register. 2020. Multi-centre, adaptive, randomized trial
of the safety and efcacy of treatments of COVID-19 in hospitalized
adults. https://www.clinicaltrialsregister.eu/ctr-search/trial/2020-000936
-23/FR.
313. Capital Medical University. A trial of remdesivir in adults with mild and
moderate COVID-19. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/
NCT04252664.
314. Capital Medical University. 2020. A phase 3 randomized, double-blind,
placebo-controlled, multicenter study to evaluate the efcacy and safety
of remdesivir in hospitalized adult patients with severe COVID-19. Clini-
calTrials.gov https://clinicaltrials.gov/ct2/show/NCT04257656.
315. US Food and Drug Administration. 22 October 2020. FDA approves
rst treatment for COVID-19. US Food and Drug Administration, Silver
Spring, MD. https://www.fda.gov/news-events/press-announcements/
fda-approves-rst-treatment-covid-19.
316. Gilead Sciences. 15 October 2020. Gilead Sciences Statement on the Sol-
idarity Trial. Gilead Sciences, Foster City, CA. https://www.gilead.com/news
-and-press/company-statements/gilead-sciences-statement-on-the-solidarity
-trial.
317. Galiuto L, Patrono C. 2020. Conicting results on the efcacy of remdesi-
vir in hospitalized Covid-19 patients: comment on the Adaptive Covid-
19 Treatment Trial. Eur Heart J 41:43874388. https://doi.org/10.1093/
Earhart/ehaa934.
318. Cohen J, Kupferschmidt K. 28 October 2020. The very, very bad lookof
remdesivir, the rst FDA-approved COVID-19 drug. Science https://www
.sciencemag.org/news/2020/10/very-very-bad-look-remdesivir-rst-fda-
approved-covid-19-drug.
319. Spinner CD, Gottlieb RL, Criner GJ, Arribas López JR, Cattelan AM,
Soriano Viladomiu A, Ogbuagu O, Malhotra P, Mullane KM, Castagna A,
Chai LYA, Roestenberg M, Tsang OTY, Bernasconi E, Le Turnier P, Chang
SC, SenGupta D, Hyland RH, Osinusi AO, Cao H, Blair C, Wang H, Gaggar
A, Brainard DM, McPhail MJ, Bhagani S, Ahn MY, Sanyal AJ, Huhn G,
Marty FM, GS-US-540-5774 Investigators. 2020. Effect of remdesivir vs
standard care on clinical status at 11 days in patients with moderate
COVID-19. JAMA 324:10481057. https://doi.org/10.1001/jama.2020
.16349.
320. Kalil AC, Patterson TF, Mehta AK, Tomashek KM, Wolfe CR, Ghazaryan V,
Marconi VC, Ruiz-Palacios GM, Hsieh L, Kline S, Tapson V, Iovine NM, Jain
MK, Sweeney DA, El Sahly HM, Branche AR, Regalado Pineda J, Lye DC,
Sandkovsky U, Luetkemeyer AF, Cohen SH, Finberg RW, Jackson PEH,
Taiwo B, Paules CI, Arguinchona H, Erdmann N, Ahuja N, Frank M, Oh
MD, Kim ES, Tan SY, Mularski RA, Nielsen H, Ponce PO, Taylor BS, Larson
L, Rouphael NG, Saklawi Y, Cantos VD, Ko ER, Engemann JJ, Amin AN,
Watanabe M, Billings J, Elie MC, Davey RT, Burgess TH, Ferreira J, Green
M, Makowski M, et al. 2020. Baricitinib plus remdesivir for hospitalized
adults with Covid-19. N Engl J Med 384:795807. https://doi.org/10
.1056/nejmoa2031994.
321. US Food and Drug Administration. 28 July 2021. Letter of Authorization:
EUA for baricitinib (Olumiant), in combination with remdesivir (Veklury),
for the treatment of suspected or laboratory conrmed coronavirus dis-
ease 2019 (COVID). US Food and Drug Administration, Silver Spring, MD.
https://www.fda.gov/media/143822/download.
322. Ashfaq UA, Javed T, Rehman S, Nawaz Z, Riazuddin S. 2011. Lysosomo-
tropic agents as HCV entry inhibitors. Virol J 8:163. https://doi.org/10
.1186/1743-422X-8-163.
323. Zhang Y, Xiao M, Zhang S, Xia P, Cao W, Jiang W, Chen H, Ding X, Zhao
H, Zhang H, Wang C, Zhao J, Sun X, Tian R, Wu W, Wu D, Ma J, Chen Y,
Zhang D, Xie J, Yan X, Zhou X, Liu Z, Wang J, Du B, Qin Y, Gao P, Qin X,
Xu Y, Zhang W, Li T, Zhang F, Zhao Y, Li Y, Zhang S. 2020. Coagulopathy
and antiphospholipid antibodies in patients with Covid-19. N Engl J
Med 382:e38. https://doi.org/10.1056/NEJMc2007575.
324. Müller-Calleja N, Manukyan D, Ruf W, Lackner K. 2016. Mechanism of
action of hydroxychloroquine in the antiphospholipid syndrome. Blood
128:5023. https://doi.org/10.1182/blood.V128.22.5023.5023.
325. Erkan D, Aguiar CL, Andrade D, Cohen H, Cuadrado MJ, Danowski A,
Levy RA, Ortel TL, Rahman A, Salmon JE, Tektonidou MG, Willis R,
Lockshin MD. 2014. 14th International Congress on Antiphospholipid
Antibodies Task Force report on antiphospholipid syndrome treatment
trends. Autoimmun Rev 13:685696. https://doi.org/10.1016/j.autrev
.2014.01.053.
326. Wang T-F, Lim W. 2016. What is the role of hydroxychloroquine in reduc-
ing thrombotic risk in patients with antiphospholipid antibodies? Hema-
tology Am Soc Hematol Educ Program 2016:714716. https://doi.org/10
.1182/asheducation-2016.1.714.
327. Zhou D, Dai S-M, Tong Q. 2020. COVID-19: a recommendation to exam-
ine the effect of hydroxychloroquine in preventing infection and pro-
gression. J Antimicrob Chemother 75:16671670. https://doi.org/10
.1093/jac/dkaa114.
328. Sperber K, Louie M, Kraus T, Proner J, Sapira E, Lin S, Stecher V, Mayer L.
1995. Hydroxychloroquine treatment of patients with human immuno-
deciency virus type 1. Clin Ther 17:622636. https://doi.org/10.1016/
0149-2918(95)80039-5.
329. Helal GK, Gad MA, Abd-Ellah MF, Eid MS. 2016. Hydroxychloroquine aug-
ments early virological response to pegylated interferon plus ribavirin in
genotype-4 chronic hepatitis C patients. J Med Virol 88:21702178.
https://doi.org/10.1002/jmv.24575.
330. Coster WJ. 2013. Making the best match: selecting outcome measures
for clinical trials and outcome studies. Am J Occup Ther 67:162170.
https://doi.org/10.5014/ajot.2013.006015.
331. Molina JM, Delaugerre C, Le Goff J, Mela-Lima B, Ponscarme D, Goldwirt
L, de Castro N. 2020. No evidence of rapid antiviral clearance or clinical
benet with the combination of hydroxychloroquine and azithromycin
in patients with severe COVID-19 infection. Med Mal Infect 50:384.
https://doi.org/10.1016/j.medmal.2020.03.006.
332. Chen Z, Hu J, Zhang Z, Jiang S, Han S, Yan D, Zhuang R, Hu B, Zhang Z.
2020. Efcacy of hydroxychloroquine in patients with COVID-19: results
of a randomized clinical trial. medRxiv https://doi.org/10.1101/2020.03
.22.20040758.
333. Chinese Clinical Trial Registry. 2020. Therapeutic effect of hydroxychlor-
oquine on novel coronavirus pneumonia (COVID). http://www.chictr.org
.cn/showprojen.aspx?proj=48880.
334. Chen J, Liu D, Liu L, Liu P, Xu Q, Xia L, Ling Y, Huang D, Song S, Dandan
Z, ... Lu H. 2020. A pilot study of hydroxychloroquine in treatment of
patients with common coronavirus disease-19 (COVID-19). J Zhejiang
Univ (Med Sci) https://doi.org/10.3785/j.issn.1008-9292.2020.03.03.
335. Gao J, Tian Z, Yang X. 2020. Breakthrough: chloroquine phosphate has
shown apparent efcacy in treatment of COVID-19 associated pneumo-
nia in clinical studies. Biosci Trends 14:7273. https://doi.org/10.5582/
bst.2020.01047.
336. Yang N, Shen H-M. 2020. Targeting the endocytic pathway and autoph-
agy process as a novel therapeutic strategy in COVID-19. Int J Biol Sci 16:
17241731. https://doi.org/10.7150/ijbs.45498.
337. Zheng J. 2020. SARS-CoV-2: an emerging coronavirus that causes a
global threat. Int J Biol Sci 16:16781685. https://doi.org/10.7150/ijbs
.45053.
338. Mehra MR, Desai SS, Ruschitzka F, Patel AN. 2020. RETRACTED: Hydroxy-
chloroquine or chloroquine with or without a macrolide for treatment of
COVID-19: a multinational registry analysis. Lancet https://doi.org/10
.1016/S0140-6736(20)31180-6.
339. Mehra MR, Desai SS, Ruschitzka F, Patel AN. 2020. RetractionHydroxy-
chloroquine or chloroquine with or without a macrolide for treatment of
COVID-19: a multinational registry analysis. Lancet https://doi.org/10
.1016/s0140-6736(20)31324-6.
340. OLaughlin JP, Mehta PH, Wong BC. 2016. Life threatening severe QTc
prolongation in patient with systemic lupus erythematosus due to
hydroxychloroquine. Case Rep Cardiol 2016:4626279. https://doi.org/10
.1155/2016/4626279.
341. Roden DM. 2008. Keep the QT interval: it is a reliable predictor of ventric-
ular arrhythmias. Heart Rhythm 5:12131215. https://doi.org/10.1016/j
.hrthm.2008.05.008.
342. Lane JCE, Weaver J, Kostka K, Duarte-Salles T, Abrahao MTF, Alghoul H,
Alser O, Alshammari TM, Biedermann P, Burn E, ... Prieto-Alhambra D.
2020. Safety of hydroxychloroquine, alone and in combination with azi-
thromycin, in light of rapid wide-spread use for COVID-19: a multina-
tional, network cohort and self-controlled case series study. medRxiv
https://doi.org/10.1101/2020.04.08.20054551.
343. Borba MGS, Val FFA, Souza Sampaio V, Almeida Araújo Alexandre M,
Cardoso Melo G, Brito M, Gomes Mourão MP, Brito-Sousa JD, Baía-da-
Silva D, Guerra MVF, ...CloroCovid-19 Team. 2020. Chloroquine diphos-
phate in two different dosages as adjunctive therapy of hospitalized
patients with severe respiratory syndrome in the context of coronavirus
Rando et al.
November/December 2021 Volume 6 Issue 6 e00233-21 msystems.asm.org 50
Downloaded from https://journals.asm.org/journal/msystems on 03 November 2021 by 2607:f470:34:2303:5040:4a48:5de1:cdb5.
(SARS-CoV-2) infection: preliminary safety results of a randomized, dou-
ble-blinded, phase IIb clinical trial (CloroCovid-19 Study). medRxiv
https://doi.org/10.1101/2020.04.07.20056424.
344. Howard J, Cohen E, Kounang N, Nyberg P. 14 April 2020. Heart risk con-
cerns mount around use of chloroquine and hydroxychloroquine for
Covid-19 treatment. CNN https://www.cnn.com/2020/04/13/health/
chloroquine-risks-coronavirus-treatment-trials-study/index.html.
345. World Health Organization. 2020. WHO Director-General's opening
remarks at the media brieng on COVID-19 25 May 2020. World Health
Organization, Geneva, Switzerland. https://www.who.int/dg/speeches/
detail/who-director-general-s-opening-remarks-at-the-media-brieng-on-
covid-1925-may-2020.
346. Tang W, Cao Z, Han M, Wang Z, Chen J, Sun W, Wu Y, Xiao W, Liu S, Chen
E, ... Xie Q. 2020. Hydroxychloroquine in patients mainly with mild to
moderate COVID19: an openlabel, randomized, controlled trial. medR-
xiv https://doi.org/10.1101/2020.04.10.20060558.
347. Wang W, Xu Y, Gao R, Lu R, Han K, Wu G, Tan W. 2020. Detection of SARS-
CoV-2 in different types of clinical specimens. JAMA 323:184321844.
https://doi.org/10.1001/jama.2020.3786.
348. Magagnoli J, Narendran S, Pereira F, Cummings T, Hardin JW, Sutton S,
Ambati J. 2020. Outcomes of hydroxychloroquine usage in United States
veterans hospitalized with Covid-19. medRxiv https://doi.org/10.1101/
2020.04.16.20065920.
349. Mitjà O, Corbacho-Monné M, Ubals M, Tebe C, Peñael J, Tobias A,
Ballana E, Alemany A, Riera-Martí N, Pérez CA, Suñer C, Laporte P,
Admella P, Mitjà J, Clua M, Bertran L, Sarquella M, Gavilán S, Ara J,
Argimon JM, Casabona J, Cuatrecasas G, Cañadas P, Elizalde-Torrent A,
Fabregat R, Farré M, Forcada A, Flores-Mateo G, Muntada E, Nadal N,
Narejos S, Gil-Ortega AN, Prat N, Puig J, Quiñones C, Reyes-Ureña J,
Ramírez-Viaplana F, Ruiz L, Riveira-Muñoz E, Sierra A, Velasco C, Vivanco-
Hidalgo RM, Sentís A, G-Beiras C, Clotet B, Vall-Mayans M, BCN PEP-CoV-
2 RESEARCH GROUP. 2020. Hydroxychloroquine for early treatment of
adults with mild coronavirus disease 2019: a randomized, controlled
trial. Clin Infect Dis https://doi.org/10.1093/cid/ciaa1009.
350. Boulware DR, Pullen MF, Bangdiwala AS, Pastick KA, Lofgren SM, Okafor
EC, Skipper CP, Nascene AA, Nicol MR, Abassi M, Engen NW, Cheng MP,
LaBar D, Lother SA, MacKenzie LJ, Drobot G, Marten N, Zarychanski R,
Kelly LE, Schwartz IS, McDonald EG, Rajasingham R, Lee TC, Hullsiek KH.
2020. A randomized trial of hydroxychloroquine as postexposure pro-
phylaxis for Covid-19. N Engl J Med 383:5172525. https://doi.org/10
.1056/NEJMoa2016638.
351. Abella BS, Jolkovsky EL, Biney BT, Uspal JE, Hyman MC, Frank I, Hensley
SE, Gill S, Vogl DT, Maillard I, Babushok DV, Huang AC, Nasta SD, Walsh
JC, Wiletyo EP, Gimotty PA, Milone MC, Amaravadi RK, Prevention and
Treatment of COVID-19 With Hydroxychloroquine (PATCH) Investigators.
2021. Efcacy and safety of hydroxychloroquine vs placebo for pre-ex-
posure SARS-CoV-2 prophylaxis among health care workers. JAMA In-
tern Med 181:1952202. https://doi.org/10.1001/jamainternmed.2020
.6319.
352. Schrezenmeier E, Dörner T. 2020. Mechanisms of action of hydroxychlor-
oquine and chloroquine: implications for rheumatology. Nat Rev Rheu-
matol 16:1552166. https://doi.org/10.1038/s41584-020-0372-x.
353. University of Pennsylvania. Elimination or Prolongation of ACE Inhibitors
and ARB in Coronavirus Disease 2019. ClinicalTrials.gov https://clinicaltrials
.gov/ct2/show/NCT04338009.
354. Medical University Innsbruck. Stopping ACE-inhibitors in COVID-19 (ACEI-
COVID). ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT04353596.
355. University of Minnesota. Losartan for patients with COVID-19 not requir-
ing hospitalization. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/
NCT04311177.
356. Universityof Minnesota. Losartan for patients with COVID-19 requiring hospi-
talization. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT04312009
357. National University of Ireland. 2020. Coronavirus (COVID-19) ACEi/ARB
Investigation (CORONACION) ClinicalTrials.gov https://clinicaltrials.gov/
ct2/show/NCT04330300.
358. University of California, San Diego. Ramipril for the treatment of COVID-19
(RAMIC). ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT04366050.
359. DOr Institute for Research and Education. 2020. Angiotensin receptor
blockers and angiotensin-converting enzyme inhibitors and adverse
outcomes in patients with COVID19 (BRACE-CORONA). ClinicalTrials.gov
https://clinicaltrials.gov/ct2/show/NCT04364893.
360. Cohen JB, Hanff TC, South AM, Sparks MA, Hiremath S, Bress AP, Byrd JB,
Chirinos JA. 2020. Response by Cohen et al to letter regarding article,
Association of inpatient use of angiotensin-converting enzyme inhibitors
and angiotensin II receptor blockers with mortality among patients with
hypertension hospitalized with COVID-19.Circ Res 126:e1402e141.
https://doi.org/10.1161/CIRCRESAHA.120.317205.
361. Sparks MA, South A, Welling P, Luther JM, Cohen J, Byrd JB, Burrell LM,
Batlle D, Tomlinson L, Bhalla V, Rheault MN, Soler MJ, Swaminathan S,
Hiremath S. 2020. Sound science before quick judgement regarding RAS
blockade in COVID-19. Clin J Am Soc Nephrol 15:7142716. https://doi
.org/10.2215/CJN.03530320.
362. NephJC. 14 March 2020. ACE2 and hypertension. http://www.nephjc
.com/news/covidace2.
363. Luo Y, Zheng SG. 2016. Hall of fame among pro-inammatory cytokines:
interleukin-6 gene and its transcriptional regulation mechanisms. Front
Immunol 7:604. https://doi.org/10.3389/mmu.2016.00604.
364. Rose-John S. 2012. IL-6 trans-signaling via the soluble IL-6 receptor: im-
portance for the pro-inammatory activities of IL-6. Int J Biol Sci 8:
123721247. https://doi.org/10.7150/ijbs.4989.
365. Scheller J, Rose-John S. 2006. Interleukin-6 and its receptor: from bench
to bedside. Med Microbiol Immunol 195:1732183. https://doi.org/10
.1007/s00430-006-0019-9.
366. Garbers C, Hermanns HM, Schaper F, Müller-Newen G, Grötzinger J,
Rose-John S, Scheller J. 2012. Plasticity and cross-talk of interleukin 6-
type cytokines. Cytokine Growth Factor Rev 23:85297. https://doi.org/
10.1016/j.cytogfr.2012.04.001.
367. Rose-John S, Heinrich PC. 1994. Soluble receptors for cytokines and
growth factors: generation and biological function. Biochem J 300(Pt 2):
2812290. https://doi.org/10.1042/bj3000281.
368. Tanaka T, Narazaki M, Masuda K, Kishimoto T. 2013. Interleukin-6; patho-
genesis and treatment of autoimmune inammatory diseases. Inamm
Regen 33:054065. https://doi.org/10.2492/inammregen.33.054.
369. Tisoncik JR, Korth MJ, Simmons CP, Farrar J, Martin TR, Katze MG. 2012.
Into the eye of the cytokine storm. Microbiol Mol Biol Rev 76:1632.
https://doi.org/10.1128/MMBR.05015-11.
370. Watad A, Bragazzi NL, Bridgewood C, Mansour M, Mahroum N, Riccò M,
Nasr A, Hussein A, Gendelman O, Shoenfeld Y, Lindar M, Amital H, Wu J,
McGonagle D. 2020. Systematic review and meta-analysis of case-con-
trol studies from 7,000 COVID-19 pneumonia patients suggests a bene-
cial impact of tocilizumab with benet most evident in non-corticoste-
roid exposed subjects. SSRN J https://doi.org/10.2139/ssrn.3642653.
371. Kaye AG, Siegel R. 2020. The efcacy of IL-6 inhibitor tocilizumab in
reducing severe COVID-19 mortality: a systematic review. PeerJ 8:
e10322. https://doi.org/10.7717/peerj.10322.
372. Cortegiani A, Ippolito M, Greco M, Granone V, Protti A, Gregoretti C,
Giarratano A, Einav S, Cecconi M. 2021. Rationale and evidence on the
use of tocilizumab in COVID-19: a systematic review. Pulmonology 27:
52266. https://doi.org/10.1016/j.pulmoe.2020.07.003.
373. Jones G, Panova E. 2018. New insights and long-term safety of tocilizu-
mab in rheumatoid arthritis. Ther Adv Musculoskelet Dis 10:1952199.
https://doi.org/10.1177/1759720X18798462.
374. Saito J, Yakuwa N, Kaneko K, Takai C, Goto M, Nakajima K, Yamatani A,
Murashima A. 2019. Tocilizumab during pregnancy and lactation: drug
levels in maternal serum, cord blood, breast milk and infant serum.
Rheumatology 58:150521507. https://doi.org/10.1093/rheumatology/
kez100.
375. Chen L-F, Mo Y-Q, Jing J, Ma J-D, Zheng D-H, Dai L. 2017. Short-course
tocilizumab increases risk of hepatitis B virus reactivation in patients
with rheumatoid arthritis: a prospective clinical observation. Int J Rheum
Dis 20:859869. https://doi.org/10.1111/1756-185X.13010.
376. Fu B, Xu X, Wei H. 2020. Why tocilizumab could be an effective treat-
ment for severe COVID-19? J Transl Med 18:164. https://doi.org/10.1186/
s12967-020-02339-3.
377. Campbell L, Chen C, Bhagat SS, Parker RA, Ostor AJK. 2010. Risk of adverse
events including serious infections in rheumatoid arthritis patients
treated with tocilizumab: a systematic literature review and meta-analysis
of randomized controlled trials. Rheumatology 50:552562. https://doi
.org/10.1093/rheumatology/keq343.
378. Pawar A, Desai RJ, Solomon DH, Santiago Ortiz AJ, Gale S, Bao M, Sarsour
K, Schneeweiss S, Kim SC. 2019. Risk of serious infections in tocilizumab
versus other biologic drugs in patients with rheumatoid arthritis: a mul-
tidatabase cohort study. Ann Rheum Dis 78:456464. https://doi.org/10
.1136/annrheumdis-2018-214367.
379. Lang VR, Englbrecht M, Rech J, Nüsslein H, Manger K, Schuch F, Tony H-
P, Fleck M, Manger B, Schett G, Zwerina J. 2012. Risk of infections in
rheumatoid arthritis patients treated with tocilizumab. Rheumatology
51:852857. https://doi.org/10.1093/rheumatology/ker223.
ID and Development of Therapeutics for COVID-19
November/December 2021 Volume 6 Issue 6 e00233-21 msystems.asm.org 51
Downloaded from https://journals.asm.org/journal/msystems on 03 November 2021 by 2607:f470:34:2303:5040:4a48:5de1:cdb5.
380. Radbel J, Narayanan N, Bhatt PJ. 2020. Use of tocilizumab for COVID-19-
induced cytokine release syndrome: a cautionary case report. Chest 158:
e15e19. https://doi.org/10.1016/j.chest.2020.04.024.
381. Tzotzos SJ, Fischer B, Fischer H, Zeitlinger M. 2020. Incidence of ARDS
and outcomes in hospitalized patients with COVID-19: a global literature
survey. Crit Care 24:516. https://doi.org/10.1186/s13054-020-03240-7.
382. Kaye A, Siegel R. 2020. The efcacy of IL-6 inhibitor tocilizumab in reduc-
ing severe COVID-19 mortality: a systematic review. medRxiv https://doi
.org/10.1101/2020.07.10.20150938.
383. Hassoun A, Thottacherry ED, Muklewicz J, Aziz Q-U-A, Edwards J. 2020.
Utilizing tocilizumab for the treatment of cytokine release syndrome in
COVID-19. J Clin Virol 128:104443. https://doi.org/10.1016/j.jcv.2020
.104443.
384. Spiegel M, Pichlmair A, Mühlberger E, Haller O, Weber F. 2004. The anti-
viral effect of interferon-beta against SARS-coronavirus is not mediated
by MxA protein. J Clin Virol 30:211213. https://doi.org/10.1016/j.jcv
.2003.11.013.
385. DeDiego ML, Nieto-Torres JL, Jimenez-Guardeño JM, Regla-Nava JA,
Castaño-Rodriguez C, Fernandez-Delgado R, Usera F, Enjuanes L. 2014. Co-
ronavirus virulence genes with main focus on SARS-CoV envelope gene. Vi-
rus Res 194:1242137. https://doi.org/10.1016/j.virusres.2014.07.024.
386. Synairgen plc. 2020. Synairgen to start trial of SNG001 in COVID-19
imminently. Synairgen plc, Southampton, United Kingdom. http://
synairgen.web01.hosting.bdci.co.uk/umbraco/Surface/Download/GetFile?
cid=23c9b12c-508b-48c3-9081-36605c5a9ccd.
387. Peiffer-Smadja N, Yazdanpanah Y. 2021. Nebulised interferon beta-1a
for patients with COVID-19. Lancet Respir Med 9:1222123. https://doi
.org/10.1016/S2213-2600(20)30523-3.
388. Ranieri VM, Pettilä V, Karvonen MK, Jalkanen J, Nightingale P, Brealey D,
Mancebo J, Ferrer R, Mercat A, Patroniti N, Quintel M, Vincent JL,
Okkonen M, Meziani F, Bellani G, MacCallum N, Creteur J, Kluge S,
Artigas-Raventos A, Maksimow M, Piippo I, Elima K, Jalkanen S, Jalkanen
M, Bellingan G, INTEREST Study Group. 2020. Effect of intravenous inter-
feron
b
-1a on death and days free from mechanical ventilation among
patients with moderate to severe acute respiratory distress syndrome.
JAMA 323:7252733. https://doi.org/10.1001/jama.2019.22525.
389. Davoudi-Monfared E, Rahmani H, Khalili H, Hajiabdolbaghi M, Salehi M,
Abbasian L, Kazemzadeh H, Yekaninejad MS. 2020. A randomized clinical
trial of the efcacy and safety of interferon
b
-1a in treatment of severe
COVID-19. Antimicrob Agents Chemother 64:e01061-20. https://doi.org/
10.1128/AAC.01061-20.
390. National Institute of Allergy and Infectious Diseases (NIAID). 2021. A
multicenter, adaptive, randomized blinded controlled trial of the safety
and efcacy of investigational therapeutics for the treatment of COVID-
19 in hospitalized adults (ACTT-3). ClinicalTrials.gov https://clinicaltrials
.gov/ct2/show/NCT04492475.
391. Canadian Agency for Drugs and Technologies in Health. 2015. Tocilizu-
mab (Actemra): adult patients with moderately to severely active rheu-
matoid arthritis. CADTH Common Drug Reviews. Canadian Agency for
Druga and Technologies in Health, Ottawa, Ontario, Canada. https://
www.ncbi.nlm.nih.gov/books/NBK349513/table/T43/.
392. Hankin C, Feldman S, Szczotka A, Stinger R, Fish L, Hankin D. 2005. A
cost comparison of treatments of moderate to severe psoriasis. Drug
Benet Trends https://escholarship.umassmed.edu/meyers_pp/385.
393. Tobinick E. 2004. TNF-
a
inhibition for potential therapeutic modulation
of SARS coronavirus infection. Curr Med Res Opin 20:3940. https://doi
.org/10.1185/030079903125002757.
394. Sano. 16 March 2020. Sanoand Regeneron begin global Kevzara
(sarilumab) clinical trial program in patients with severe COVID-19.
Sano, Bridgewater, NJ. http://www.news.sano.us/2020-03-16-Sano
-and-Regeneron-begin-global-Kevzara-R-sarilumab-clinical-trial-program-in-
patients-with-severe-COVID-19.
395. Sano. Sarilumab COVID-19. ClinicalTrials.gov https://clinicaltrials.gov/
ct2/show/NCT04327388.
396. Stebbing J, Phelan A, GrifnI,TuckerC,OechsleO,SmithD,RichardsonP.
2020. COVID-19: combining antiviral and anti-inammatory treatments. Lan-
cet Infect Dis 20:4002402. https://doi.org/10.1016/S1473-3099(20)30132-8.
397. Richardson P, GrifnI,TuckerC,SmithD,OechsleO,PhelanA,RawlingM,
Savory E, Stebbing J. 2020. Baricitinib as potential treatment for 2019-nCoV
acute respiratory disease. Lancet 395:e30e31. https://doi.org/10.1016/
S0140-6736(20)30304-4.
398. Eli Lilly and Company. 10 April 2020. Lilly begins clinical testing of thera-
pies for COVID-19. Eli Lilly and Company, Indianapolis, IN. https://
investor.lilly.com/news-releases/news-release-details/lilly-begins-clinical
-testing-therapies-covid-19.
399. Hospital of Prado. 2020. Baricitinib combined with antiviral therapy in
symptomatic patients infected by COVID-19: an open-label, pilot study.
ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT04320277.
400. Biot C, Daher W, Chavain N, Fandeur T, Khalife J, Dive D, De Clercq E.
2006. Design and synthesis of hydroxyferroquine derivatives with anti-
malarial and antiviral activities. J Med Chem 49:284522849. https://doi
.org/10.1021/jm0601856.
401. Sheahan TP, Sims AC, Zhou S, Graham RL, Pruijssers AJ, Agostini ML,
Leist SR, Schäfer A, Dinnon KH, III, Stevens LJ, Chappell JD, Lu X, Hughes
TM, George AS, Hill CS, Montgomery SA, Brown AJ, Bluemling GR,
Natchus MG, Saindane M, Kolykhalov AA, Painter G, Harcourt J, Tamin A,
Thornburg NJ, Swanstrom R, Denison MR, Baric RS. 2020. An orally bioa-
vailable broad-spectrum antiviral inhibits SARS-CoV-2 in human airway
epithelial cell cultures and multiple coronaviruses in mice. Sci Transl
Med 12:eabb5883. https://doi.org/10.1126/scitranslmed.abb5883.
402. Pelegrin M, Naranjo-Gomez M, Piechaczyk M. 2015. Antiviral monoclonal
antibodies: can they be more than simple neutralizing agents? Trends
Microbiol 23:6532665. https://doi.org/10.1016/j.tim.2015.07.005.
403. Alberts B, Johnson A, Lewis J, Raff M, Roberts K, Walter P. 2002. Molecu-
lar biology of the cell, 4th ed. Garland Science, New York, NY.
404. Zhao J, Wohlford-Lenane C, Zhao J, Fleming E, Lane TE, McCray PB,
Perlman S. 2012. Intranasal treatment with poly(IC) protects aged mice
from lethal respiratory virus infections. J Virol 86:11416211424. https://
doi.org/10.1128/JVI.01410-12.
Rando et al.
November/December 2021 Volume 6 Issue 6 e00233-21 msystems.asm.org 52
Downloaded from https://journals.asm.org/journal/msystems on 03 November 2021 by 2607:f470:34:2303:5040:4a48:5de1:cdb5.
... 4 Efforts to develop interventions for those with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection have resulted in evidence-based therapies. [5][6][7][8][9][10][11] However, these advances are limited and may not have shown unequivocal efficacy. 7 Vaccination is protective against severe infection; however, incomplete acceptance of vaccination, incomplete protection for immunocompromised patients, and variant emergence highlight the ongoing need for the development of additional COVID-19 therapies. ...
... [5][6][7][8][9][10][11] However, these advances are limited and may not have shown unequivocal efficacy. 7 Vaccination is protective against severe infection; however, incomplete acceptance of vaccination, incomplete protection for immunocompromised patients, and variant emergence highlight the ongoing need for the development of additional COVID-19 therapies. 8,[11][12][13] Ideally, new treatment options for COVID-19, and other respiratory viral illnesses, will be virus and variant independent, well tolerated, and easily administered. ...
... 1,11 Most disease-specific treatments developed to date (antivirals and monoclonal antibodies) are virus-and/or variant-specific. 7,8,11 Non-virus-specific therapies (e.g., systemic corticosteroids) are principally utilized in hospitalized patients and often require intravenous administration. [5][6][7] Recently, non-virus-specific antivirals have been developed. ...
Article
Full-text available
Introduction Granulocyte-macrophage colony-stimulating factor (GM-CSF), a protein produced in the lung, is essential for pulmonary host defense and alveolar integrity. Prior studies suggest potential benefits in several pulmonary conditions, including acute respiratory distress syndrome and viral infections. This trial evaluated the effect of the addition of inhaled sargramostim (yeast-derived, glycosylated recombinant human GM-CSF) to standard of care (SOC) on oxygenation and clinical outcomes in patients with COVID-19-associated acute hypoxemia. Materials and Methods A randomized, controlled, open-label trial of hospitalized adults with COVID-19-associated hypoxemia (oxygen saturation <93% on ≥2 L/min oxygen supplementation and/or PaO2/FiO2 <350) randomized 2:1 to inhaled sargramostim (125 mcg twice daily for 5 days) plus SOC versus SOC alone. Institutional SOC before and during the study was not limited. Primary outcomes were change in the alveolar–arterial oxygen gradient (P(A–a)O2) by day 6 and the percentage of patients intubated within 14 days. Safety evaluations included treatment-emergent adverse events. Efficacy analyses were based on the modified intent-to-treat population, the subset of the intent-to-treat population that received ≥1 dose of any study treatment (sargramostim and/or SOC). An analysis of covariance approach was used to analyze changes in oxygenation measures. The intubation rate was analyzed using the chi-squared test. All analyses are considered descriptive. The study was institutional review board approved. Results In total, 122 patients were treated (sargramostim, n = 78; SOC, n = 44). The sargramostim arm experienced greater improvement in P(A–a)O2 by day 6 compared to SOC alone (least squares [LS] mean change from baseline [SE]: −102.3 [19.4] versus −30.5 [26.9] mmHg; LS mean difference: −71.7 [SE 33.2, 95% CI −137.7 to −5.8]; P = .033; n = 96). By day 14, 11.5% (9/78) of sargramostim and 15.9% (7/44) of SOC arms required intubation (P = .49). The 28-day mortality was 11.5% (9/78) and 13.6% (6/44) in the sargramostim and SOC arms, respectively (hazard ratio 0.85; P = .76). Treatment-emergent adverse events occurred in 67.9% (53/78) and 70.5% (31/44) on the sargramostim and SOC arms, respectively. Conclusions The addition of inhaled sargramostim to SOC improved P(A–a)O2, a measure of oxygenation, by day 6 in hospitalized patients with COVID-19-associated acute hypoxemia and was well tolerated. Inhaled sargramostim is delivered directly to the lung, minimizing systemic effects, and is simple to administer making it a feasible treatment option in patients in settings where other therapy routes may be difficult. Although proportionally lower rates of intubation and mortality were observed in sargramostim-treated patients, this study was insufficiently powered to demonstrate significant changes in these outcomes. However, the significant improvement in gas exchange with sargramostim shows this inhalational treatment enhances pulmonary efficiency in this severe respiratory illness. These data provide strong support for further evaluation of sargramostim in high-risk patients with COVID-19.
... The pressing need for results, as well as inherent public and governmental pressure during a pandemic, stressed the integrity of the scientific process and required unconventional methods to work so many lines of inquiry in parallel. Finances and timelines factor heavily into therapeutic development, and repurposing existing medications for immediate use became an attractive option, as it is both cost-effective and time-saving [3]. Many clinicians will remember the state of COVID-19 research early on in the pandemic, which evolved from individual clinicians posting their experiences on Twitter, advancing to small, preliminary, and sometimes rushed clinical trials, and finally progressing to larger, more rigorous studies. ...
Article
Full-text available
“In the midst of chaos, there is also opportunity”—Sun Tzu, The Art of War [...]
... COVID-19 could be mild to severe, and 2% of the diagnosed die from acute respiratory distress syndrome (ARDS), while acute lung injury or ARDS is the main symptom of severe malaria. This can occur even after antimalarial treatment [12,13]. Malaria prevention and treatment in hypoendemic areas is even more crucial during the era of the COVID-19 pandemic to sustain the gains made [14]. ...
Article
Full-text available
Coronavirus disease 2019 (COVID-19) contact tracing and malaria reactive case detection (RACD) are effective strategies for disease control. The emergence of the COVID-19 pandemic and the global attention COVID-19 has received in the recent past and present has hampered malaria control efforts. Among these are difficulties in finding and treating malaria-infected individuals in hypoendemic settings in the community, due to lockdown restrictions by countries. It is common knowledge that malaria cases that cannot be identified remain untreated. To sustain the gains made in malaria control, we proposed a two-pronged hybrid approach for COVID-19 contact tracing and malaria RACD in communities with COVID-19 and malaria coinfections. Such an approach would equally factor the burden of malaria cases and COVID-19 to support an effective strategy for responding to current and future pandemics.
Article
Full-text available
Over the past 150 years, vaccines have revolutionized the relationship between people and disease. During the COVID-19 pandemic, technologies such as mRNA vaccines have received attention due to their novelty and successes. However, more traditional vaccine development platforms have also yielded important tools in the worldwide fight against the SARS-CoV-2 virus. A variety of approaches have been used to develop COVID-19 vaccines that are now authorized for use in countries around the world. In this review, we highlight strategies that focus on the viral capsid and outwards, rather than on the nucleic acids inside. These approaches fall into two broad categories: whole-virus vaccines and subunit vaccines. Whole-virus vaccines use the virus itself, either in an inactivated or attenuated state. Subunit vaccines contain instead an isolated, immunogenic component of the virus. Here, we highlight vaccine candidates that apply these approaches against SARS-CoV-2 in different ways. In a companion manuscript, we review the more recent and novel development of nucleic-acid based vaccine technologies. We further consider the role that these COVID-19 vaccine development programs have played in prophylaxis at the global scale. Well-established vaccine technologies have proved especially important to making vaccines accessible in low- and middle-income countries. Vaccine development programs that use established platforms have been undertaken in a much wider range of countries than those using nucleic-acid-based technologies, which have been led by wealthy Western countries. Therefore, these vaccine platforms, though less novel from a biotechnological standpoint, have proven to be extremely important to the management of SARS-CoV-2.
Article
Full-text available
In the 21st century, several emergent viruses have posed a global threat. Each pathogen has emphasized the value of rapid and scalable vaccine development programs. The ongoing severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has made the importance of such efforts especially clear. New biotechnological advances in vaccinology allow for recent advances that provide only the nucleic acid building blocks of an antigen, eliminating many safety concerns. During the COVID-19 pandemic, these DNA and RNA vaccines have facilitated the development and deployment of vaccines at an unprecedented pace. This success was attributable at least in part to broader shifts in scientific research relative to prior epidemics: the genome of SARS-CoV-2 was available as early as January 2020, facilitating global efforts in the development of DNA and RNA vaccines within 2 weeks of the international community becoming aware of the new viral threat. Additionally, these technologies that were previously only theoretical are not only safe but also highly efficacious. Although historically a slow process, the rapid development of vaccines during the COVID-19 crisis reveals a major shift in vaccine technologies. Here, we provide historical context for the emergence of these paradigm-shifting vaccines. We describe several DNA and RNA vaccines in terms of their efficacy, safety, and approval status. We also discuss patterns in worldwide distribution. The advances made since early 2020 provide an exceptional illustration of how rapidly vaccine development technology has advanced in the last 2 decades in particular and suggest a new era in vaccines against emerging pathogens. IMPORTANCE The SARS-CoV-2 pandemic has caused untold damage globally, presenting unusual demands on but also unique opportunities for vaccine development. The development, production, and distribution of vaccines are imperative to saving lives, preventing severe illness, and reducing the economic and social burdens caused by the COVID-19 pandemic. Although vaccine technologies that provide the DNA or RNA sequence of an antigen had never previously been approved for use in humans, they have played a major role in the management of SARS-CoV-2. In this review, we discuss the history of these vaccines and how they have been applied to SARS-CoV-2. Additionally, given that the evolution of new SARS-CoV-2 variants continues to present a significant challenge in 2022, these vaccines remain an important and evolving tool in the biomedical response to the pandemic.
Article
Full-text available
Over the past 150 years, vaccines have revolutionized the relationship between people and disease. During the COVID-19 pandemic, technologies such as mRNA vaccines have received attention due to their novelty and successes. However, more traditional vaccine development platforms have also yielded important tools in the worldwide fight against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). A variety of approaches have been used to develop COVID-19 vaccines that are now authorized for use in countries around the world. In this review, we highlight strategies that focus on the viral capsid and outwards, rather than on the nucleic acids inside. These approaches fall into two broad categories: whole-virus vaccines and subunit vaccines. Whole-virus vaccines use the virus itself, in either an inactivated or an attenuated state. Subunit vaccines contain instead an isolated, immunogenic component of the virus. Here, we highlight vaccine candidates that apply these approaches against SARS-CoV-2 in different ways. In a companion article (H. M. Rando, R. Lordan, L. Kolla, E. Sell, et al., mSystems 8:e00928-22, 2023, https://doi.org/10.1128/mSystems.00928-22), we review the more recent and novel development of nucleic acid-based vaccine technologies. We further consider the role that these COVID-19 vaccine development programs have played in prophylaxis at the global scale. Well-established vaccine technologies have proved especially important to making vaccines accessible in low- and middle-income countries. Vaccine development programs that use established platforms have been undertaken in a much wider range of countries than those using nucleic acid-based technologies, which have been led by wealthy Western countries. Therefore, these vaccine platforms, though less novel from a biotechnological standpoint, have proven to be extremely important to the management of SARS-CoV-2.
Article
Full-text available
Severe acute respiratory syndrome coronavirus 2 (SARS‑CoV‑2) induced cytokine storm is the major cause of COVID‑19 related deaths. Patients have been treated with drugs that work by inhibiting a specific protein partly responsible for the cytokines production. This approach provided very limited success, since there are multiple proteins involved in the complex cell signaling disease mechanisms. We targeted five proteins: Angiotensin II receptor type 1 (AT1R), A disintegrin and metalloprotease 17 (ADAM17), Nuclear Factor‑Kappa B (NF‑κB), Janus kinase 1 (JAK1) and Signal Transducer and Activator of Transcription 3 (STAT3), which are involved in the SARS‑CoV‑2 induced cytokine storm pathway. We developed machine learning (ML) models for these five proteins, using known active inhibitors. After developing the model for each of these proteins, FDA-approved drugs were screened to find novel therapeutics for COVID‑19. We identified twenty drugs that are active for four proteins with predicted scores greater than 0.8 and eight drugs active for all five proteins with predicted scores over 0.85. Mitomycin C is the most active drug across all five proteins with an average prediction score of 0.886. For further validation of these results, we used the PyRx software to conduct protein–ligand docking experiments and calculated the binding affinity. The docking results support findings by the ML model. This research study predicted that several drugs can target multiple proteins simultaneously in cytokine storm-related pathway. These may be useful drugs to treat patients because these therapies can fight cytokine storm caused by the virus at multiple points of inhibition, leading to synergistically effective treatments.
Article
Full-text available
In the 21st century, several emergent viruses have posed a global threat. Each pathogen has emphasized the value of rapid and scalable vaccine development programs. The ongoing SARS-CoV-2 pandemic has made the importance of such efforts especially clear. New biotechnological advances in vaccinology allow for recent advances that provide only the nucleic acid building blocks of an antigen, eliminating many safety concerns. During the COVID-19 pandemic, these DNA and RNA vaccines have facilitated the development and deployment of vaccines at an unprecedented pace. This success was attributable at least in part to broader shifts in scientific research relative to prior epidemics; the genome of SARS-CoV-2 was available as early as January 2020, facilitating global efforts in the development of DNA and RNA vaccines within two weeks of the international community becoming aware of the new viral threat. Additionally, these technologies that were previously only theoretical are not only safe but also highly efficacious. Although historically a slow process, the rapid development of vaccines during the COVID-19 crisis reveals a major shift in vaccine technologies. Here, we provide historical context for the emergence of these paradigm-shifting vaccines. We describe several DNA and RNA vaccines and in terms of their efficacy, safety, and approval status. We also discuss patterns in worldwide distribution. The advances made since early 2020 provide an exceptional illustration of how rapidly vaccine development technology has advanced in the last two decades in particular and suggest a new era in vaccines against emerging pathogens.
Preprint
Full-text available
In the 21st century, several emergent viruses have posed a global threat. Each of these pathogens has emphasized the value of rapid and scalable vaccine development programs. The ongoing SARS-CoV-2 pandemic has made the importance of such efforts especially clear. In particular, new biotechnological advances in vaccine design allowed for new advances in vaccines that provide only the nucleic acid building blocks of an antigen, eliminating many safety concerns. During the COVID-19 pandemic, these DNA and RNA vaccines have facilitated the development and deployment of vaccines at an unprecedented pace. This success was attributable at least in part to broader shifts in scientific research relative to prior epidemics. For example, the genome of SARS-CoV-2 was available as early as January 2020, allowing for global efforts in the development of DNA and RNA vaccines to begin within two weeks of the international community becoming aware of the new viral threat. Additionally, these technologies that were previously only theoretical were found to be not only safe but also to have high vaccine efficacy. Although historically a slow process, the rapid development of vaccines during the COVID-19 public health crisis reveals a major shift in vaccine technologies. In this review, we provide historical context for the emergence of these paradigm-shifting vaccines. Further context is provided in a companion review exploring more established vaccines platforms. We describe several DNA and RNA vaccines and in terms of their efficacy, safety, and approval status. We also discuss patterns in worldwide distribution. The advances made since early 2020 provide an exceptional illustration of how rapidly vaccine development technology has advanced in the last two decades in particular, and suggest a new era in vaccines against emerging pathogens.
Article
Full-text available
The continuing emergence of variants of the SARS-CoV-2 virus requires the development of modular molecular therapies. Here, we engineered a recombinant amphiphilic protein, oleosin, to spontaneously self-assemble into multivalent micellar nanostructures which can block the Spike S1 protein of SARS-CoV-2 pseudoviruses (PVs). Short recombinant proteins like oleosin can be formulated more easily than antibodies and can be functionalized with precision through genetic engineering. We cloned S1-binding mini-protein genes called LCBx, previously designed by David Baker's laboratory (UW Seattle), to the N-terminus of oleosin, expressing Oleo-LCBx proteins in E. coli. These proteins largely formed 10-100 nm micelles as verified by dynamic light scattering. Two proteins, Oleo-LCB1 and Oleo-LCB3, were seen to completely and irreversibly block transduction by both wild-type and delta variant PVs into 293T-hsACE2 cells at 10 μM. Presented in multivalent micelles, these proteins reduced transduction by PVs down to a functional protein concentration of 5 nM. Additionally, Oleo-LCB1 micelles outperformed corresponding synthetic LCB1 mini-proteins in reducing transduction by PVs. Tunable aqueous solubility of recombinant oleosin allowed incorporation of peptides/mini-proteins at high concentrations within micelles, thus enhancing drug loading. To validate the potential multifunctionality of the micelles, we showed that certain combinations of Oleo-LCB1 and Oleo-LCB3 performed much better than the individual proteins at the same concentration. These micelles, which we showed to be non-toxic to human cells, are thus a promising step toward the design of modular, multifunctional therapeutics that could bind to and inactivate multiple receptors and proteins necessary for the infection of the SARS-CoV-2 virus.
Article
Full-text available
Background: Coronavirus disease 2019 (Covid-19) disproportionately results in hospitalization or death in older patients and those with underlying conditions. Sotrovimab is a pan-sarbecovirus monoclonal antibody that was designed to prevent progression of Covid-19 in high-risk patients early in the course of disease. Methods: In this ongoing, multicenter, double-blind, phase 3 trial, we randomly assigned, in a 1:1 ratio, nonhospitalized patients with symptomatic Covid-19 (≤5 days after the onset of symptoms) and at least one risk factor for disease progression to receive a single infusion of sotrovimab at a dose of 500 mg or placebo. The primary efficacy outcome was hospitalization (for >24 hours) for any cause or death within 29 days after randomization. Results: In this prespecified interim analysis, which included an intention-to-treat population of 583 patients (291 in the sotrovimab group and 292 in the placebo group), 3 patients (1%) in the sotrovimab group, as compared with 21 patients (7%) in the placebo group, had disease progression leading to hospitalization or death (relative risk reduction, 85%; 97.24% confidence interval, 44 to 96; P = 0.002). In the placebo group, 5 patients were admitted to the intensive care unit, including 1 who died by day 29. Safety was assessed in 868 patients (430 in the sotrovimab group and 438 in the placebo group). Adverse events were reported by 17% of the patients in the sotrovimab group and 19% of those in the placebo group; serious adverse events were less common with sotrovimab than with placebo (in 2% and 6% of the patients, respectively). Conclusions: Among high-risk patients with mild-to-moderate Covid-19, sotrovimab reduced the risk of disease progression. No safety signals were identified. (Funded by Vir Biotechnology and GlaxoSmithKline; COMET-ICE ClinicalTrials.gov number, NCT04545060.).
Article
Full-text available
The novel coronavirus SARS-CoV-2, which emerged in late 2019, has since spread around the world and infected hundreds of millions of people with coronavirus disease 2019 (COVID-19). While this viral species was unknown prior to January 2020, its similarity to other coronaviruses that infect humans has allowed for rapid insight into the mechanisms that it uses to infect human hosts, as well as the ways in which the human
Article
Full-text available
Ivermectin is an antiparasitic drug being investigated for repurposing against SARS-CoV-2. Ivermectin showed in-vitro activity against SARS-COV-2 at high concentrations. This meta-analysis investigated ivermectin in 24 randomized clinical trials (3328 patients) identified through systematic searches of PUBMED, EMBASE, MedRxiv and trial registries. Ivermectin was associated with reduced inflammatory markers (C-Reactive Protein, d-dimer and ferritin) and faster viral clearance by PCR. Viral clearance was treatment dose- and duration-dependent. In 11 randomized trials of moderate/severe infection, there was a 56% reduction in mortality (Relative Risk 0.44 [95%CI 0.25-0.77]; p=0.004; 35/1064 (3%) deaths on ivermectin; 93/1063 (9%) deaths in controls) with favorable clinical recovery and reduced hospitalization. Many studies included were not peer reviewed and a wide range of doses were evaluated. Currently, WHO recommends the use of ivermectin only inside clinical trials. A network of large clinical trials is in progress to validate the results seen to date.
Article
Full-text available
COVID-19 has resulted in huge numbers of infections and deaths worldwide and brought the most severe disruptions to societies and economies since the Great Depression. Massive experimental and computational research effort to understand and characterize the disease and rapidly develop diagnostics, vaccines, and drugs has emerged in response to this devastating pandemic and more than 130 000 COVID-19-related research papers have been published in peer-reviewed journals or deposited in preprint servers. Much of the research effort has focused on the discovery of novel drug candidates or repurposing of existing drugs against COVID-19, and many such projects have been either exclusively computational or computer-aided experimental studies. Herein, we provide an expert overview of the key computational methods and their applications for the discovery of COVID-19 small-molecule therapeutics that have been reported in the research literature. We further outline that, after the first year the COVID-19 pandemic, it appears that drug repurposing has not produced rapid and global solutions. However, several known drugs have been used in the clinic to cure COVID-19 patients, and a few repurposed drugs continue to be considered in clinical trials, along with several novel clinical candidates. We posit that truly impactful computational tools must deliver actionable, experimentally testable hypotheses enabling the discovery of novel drugs and drug combinations, and that open science and rapid sharing of research results are critical to accelerate the development of novel, much needed therapeutics for COVID-19.
Article
Full-text available
Background We systematically assessed benefits and harms of the use of ivermectin (IVM) in COVID-19 patients. Methods Published and preprint randomized controlled trials (RCTs) assessing IVM effects on COVID-19 adult patients were searched until March 22, 2021 in five engines. Primary outcomes were all-cause mortality, length of stay (LOS), and adverse events (AE). Secondary outcomes included viral clearance and severe AEs. Risk of bias (RoB) was evaluated using Cochrane RoB 2·0 tool. Inverse variance random effect meta-analyses were performed. with quality of evidence (QoE) evaluated using GRADE methodology. Results Ten RCTs (n=1173) were included. Controls were standard of care [SOC] in five RCTs and placebo in five RCTs. COVID-19 disease severity was mild in 8 RCTs, moderate in one RCT, and mild and moderate in one RCT. IVM did not reduce all-cause mortality vs. controls (RR 0.37, 95%CI 0.12 to 1.13, very low QoE) or LOS vs. controls (MD 0.72 days, 95%CI −0.86 to 2.29, very low QoE). AEs, severe AE and viral clearance were similar between IVM and controls (all outcomes: low QoE). Subgroups by severity of COVID-19 or RoB were mostly consistent with main analyses; all-cause mortality in three RCTs at high RoB was reduced with IVM. Conclusions In comparison to SOC or placebo, IVM did not reduce all-cause mortality, length of stay or viral clearance in RCTs in COVID-19 patients with mostly mild disease. IVM did not have an effect on AEs or severe AEs. IVM is not a viable option to treat COVID-19 patients.
Article
Full-text available
Aims This systematic review and meta-analysis aims to investigate the effect of ivermectin on mortality in patients with COVID-19. Methods A comprehensive systematic literature search was performed using PubMed, Scopus, Embase, and Clinicaltrials.gov from the inception of databases up until April 9, 2021. The intervention group was ivermectin and the control group was standard of care or placebo. The primary outcome was mortality reported as risk ratio (RR). Results There were 9 RCTs comprising of 1788 patients included in this meta-analysis. Ivermectin was associated with decreased mortality (RR 0.39 [95% 0.20–0.74], p = 0.004; I²: 58.2%, p = 0.051). Subgroup analysis in patients with severe COVID-19 showed borderline statistical significance towards mortality reduction (RR 0.42 [95% 0.18–1.00], p = 0.052; I²: 68.3, p = 0.013). The benefit of ivermectin and mortality was reduced by hypertension (RR 1.08 [95% CI 1.03–1.13], p = 0.001); but was not influenced by age (p = 0.657), sex (p = 0.466), diabetes (p = 0.429). Sensitivity analysis using fixed-effect model showed that ivermectin decreased mortality in general (RR 0.43 [95% CI 0.29–0.62], p < 0.001) and severe COVID-19 subgroup (RR 0.48 [95% CI 0.32–0.72], p < 0.001). Conclusions Ivermectin was associated with decreased mortality in COVID-19 with a low certainty of evidence. Further adequately powered double-blinded placebo-controlled RCTs are required for definite conclusion.
Article
Full-text available
Objective: To evaluate different doses of ivermectin in adult patients with mild COVID-19 and to evaluate the effect of ivermectin on mortality and clinical consequences. Methods: A randomized, double-blind, placebo-controlled, multicenter clinical trial was performed at five hospitals. A total of 180 mild hospitalized patients with COVID-19 confirmed by PCR or chest image tests were enrolled and allocated to six arms including hydroxychloroquine 200 mg twice per day, placebo plus hydroxychloroquine 200 mg twice per day, single dose ivermectin (200 μg/kg), three low interval doses of ivermectin (200, 200, 200 μg/kg), single dose ivermectin (400 μg/kg), and three high interval doses of ivermectin (400, 200, 200 μg/kg). The primary endpoint of this trial was all-cause of mortality or clinical recovery. The radiographic findings, hospitalization and low O2 saturation duration, and hematological variables of blood samples were analyzed. Results: A total of 16.7% (5/30) and 20.0% (6/30) patients died in arms treated with hydroxychloroquine 200 mg twice per day and placebo plus hydroxychloroquine 200 mg twice per day, respectively, and a reduction in mortality rate in patients receiving ivermectin treatment to 0%, 10%, 0% and 3.3% for arms 1-4 were observed. Risk of mortality was also decreased about 15% in the ivermectin treated arms. Conclusions: Ivermectin as an adjunct reduces the rate of mortality, time of low O2 saturation, and duration of hospitalization in adult COVID-19 patients. The improvement of other clinical parameters shows that ivermectin, with a wide margin of safety, had a high therapeutic effect on COVID-19.
Article
Introduction Ivermectin is an antiparasitic drug which has in-vitro efficacy in reducing severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) viral load. Hence, Ivermectin is under investigation as a repurposed agent for treating COVID-19. Methods In this pilot, double blind, randomized controlled trial, hospitalized patients with mild-to-moderate COVID-19 were assigned to a single oral administration of an elixir formulation of Ivermectin at either 24 mg or 12 mg dose, or placebo in a 1:1:1 ratio. The co-primary outcomes were conversion of RT-PCR to negative result and the decline of viral load at day 5 of enrolment. Safety outcomes included total and serious adverse events. The primary outcomes were assessed in patients who had positive RT-PCR at enrolment (modified intention-to-treat population). Safety outcomes were assessed in all patients who received the intervention (intention-to-treat population). Results Among the 157 patients randomized, 125 were included in modified intention-to-treat analysis. 40 patients each were assigned to Ivermectin 24 mg and 12 mg, and 45 patients to placebo. The RT-PCR negativity at day 5 was higher in the two Ivermectin arms but failed to attain statistical significance (Ivermectin 24 mg, 47.5%; 12 mg arm, 35.0%; and placebo arm, 31.1%; p-value = 0.30). The decline of viral load at day 5 was similar in each arm. No serious adverse events occurred. Conclusions In patients with mild and moderate COVID-19, a single oral administration of Ivermectin did not significantly increase either the negativity of RT-PCR or decline in viral load at day 5 of enrolment compared with placebo.
Article
Are many drug repurposers pursuing artifacts?