ArticlePDF AvailableLiterature Review

Abstract

Introduction Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has emerged in China. There are no available vaccines or antiviral drugs for COVID-19 patients. Thence, we represented possible therapeutic agents that may stand as a potential therapy against COVID-19. Areas covered We searched PubMed, Google Scholar, and clinicaltrials.gov for relevant papers. We showed some agents with potentially favorable efficacy, acceptable safety as well as good pharmacokinetic profiles. Several therapies are under assessment to evaluate their efficacy and safety for COVID-19. Yet, some drugs were withdrawn due to their side effects after demonstrating some clinical efficacy. Indeed, the most effective therapies could be organ function support, convalescent plasma, anticoagulants, and immune as well as antiviral therapies, especially anti-influenza drugs due to the similarities between respiratory viruses regarding viral entry, uncoating, and replication. We encourage giving more attention to favipiravir, remdesivir, and measles vaccine. Expert opinion A combination, at least dual or even triple therapy, of the aforementioned efficacious and safe therapies is greatly recommended for COVID-19. Further, patients should have a routine assessment for their coagulation and bleeding profiles as well as their inflammatory and cytokine concentrations.
Full Terms & Conditions of access and use can be found at
https://www.tandfonline.com/action/journalInformation?journalCode=ierz20
Expert Review of Anti-infective Therapy
ISSN: (Print) (Online) Journal homepage: https://www.tandfonline.com/loi/ierz20
Possible therapeutic agents for COVID-19: a
comprehensive review
Khaled Mosaad Elhusseiny , Fatma Abd-Elshahed Abd-Elhay & Mohamed
Gomaa Kamel
To cite this article: Khaled Mosaad Elhusseiny , Fatma Abd-Elshahed Abd-Elhay & Mohamed
Gomaa Kamel (2020): Possible therapeutic agents for COVID-19: a comprehensive review, Expert
Review of Anti-infective Therapy, DOI: 10.1080/14787210.2020.1782742
To link to this article: https://doi.org/10.1080/14787210.2020.1782742
Accepted author version posted online: 13
Jun 2020.
Submit your article to this journal
View related articles
View Crossmark data
Publisher: Taylor & Francis & Informa UK Limited, trading as Taylor & Francis Group
Journal: Expert Review of Anti-infective Therapy
DOI: 10.1080/14787210.2020.1782742
Article type: Review
Possible therapeutic agents for COVID-19: a comprehensive review
Khaled Mosaad Elhusseiny1,2 #, Fatma Abd-Elshahed Abd-Elhay3#, Mohamed Gomaa
Kamel3#*
1Faculty of Medicine, Al-Azhar University, Cairo, Egypt.
2Sayed Galal University Hospital, Cairo, Egypt.
3Faculty of Medicine, Minia University, Minia 61519, Egypt.
#Authors equally contributed the work.
*Corresponding author:
Mohamed Gomaa Kamel,
Faculty of Medicine, Minia University,
Minia 61519, Egypt
E-mail: mohamedgomaa@s-mu.edu.eg
Phone number: 201090717950
ACCEPTED MANUSCRIPT
Abstract
Introduction: Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has emerged in
China. There are no available vaccines or antiviral drugs for COVID-19 patients. Herein, we
represented possible therapeutic agents that may stand as a potential therapy against COVID-19.
Areas covered: We searched PubMed, Google Scholar, and clinicaltrials.gov for relevant
papers. We showed some agents with potentially favorable efficacy, acceptable safety as well as
good pharmacokinetic profiles. Several therapies are under assessment to evaluate their efficacy
and safety for COVID-19. However, some drugs were withdrawn due to their side effects after
demonstrating some clinical efficacy. Indeed, the most effective therapies could be organ
function support, convalescent plasma, anticoagulants, and immune as well as antiviral
therapies, especially anti-influenza drugs due to the similarities between respiratory viruses
regarding viral entry, uncoating, and replication. We encourage giving more attention to
favipiravir, remdesivir, and measles vaccine.
Expert opinion: A combination, at least dual or even triple therapy, of the aforementioned
efficacious and safe therapies is greatly recommended for COVID-19. Further, patients should
have a routine assessment for their coagulation and bleeding profiles as well as their
inflammatory and cytokine concentrations.
Keywords: Coronavirus, Infection, COVID-19, Outbreak, Bat, Pandemic, Therapy, Drug,
Vaccine, Immunization
ACCEPTED MANUSCRIPT
Article highlights
SARS-CoV-2 is now much more hazardous than expected compared to previous outbreaks.
There are no available vaccines or specific antiviral drugs for COVID-19 patients.
Consequently, the current treatment protocols are depending mainly on supportive treatment
and trying previously used drugs due to our experience on the posology, safety profile, side
effects, as well as drug interactions of these drugs.
Herein, we investigated the potential therapeutics for COVID-19 patients based on the
previously documented antiviral activity either against SARS- and MERS-CoVs or other
viral infections and based on the ongoing or published SARS-CoV-2 studies.
Several therapies are under assessment to evaluate their efficacy and safety for COVID-19.
On the other hand, some drugs have been withdrawn due to their side effects after
demonstrating some clinical efficacy while COVID-19, as of today, is essentially
untreatable, except for supportive management.
Indeed, the most effective therapies could be organ function support, convalescent plasma,
anticoagulants, and antiviral therapy.
We encourage giving more attention to favipiravir, remdesivir, and measles vaccine.
Convalescent plasma was associated with reduced mortality during the 1918 influenza,
SARS, 2009 influenza H1N1, and Ebola outbreaks.
ALPS and ECMO should be considered under strict indications and contraindications or
that leads to waste of resources and additional complications.
Tocilizumab, as an anti-IL-6 receptor, has a promising therapeutic potential.
Antibiotics for pneumonia are essential in case of bacterial infection only otherwise it may
fuel bacterial resistance.
Undoubtedly, a combination, at least dual or even triple therapy, of the aforementioned
efficacious and safe therapies is greatly recommended for COVID-19 due to the disease
nature but giving great attention that mild, moderate, and severe patients may have different
regimens and combinations based on the disease severity is important as well.
Further, patients should have a routine assessment for their coagulation and bleeding
profiles as well as their inflammatory and cytokine concentrations.
ACCEPTED MANUSCRIPT
1. Introduction
In December 2019, an outbreak of severe acute respiratory syndrome coronavirus-2 (SARS-
CoV-2) has emerged in Wuhan, China[1]. Since then, >7 million infected cases and >400,000
deaths from 2019 CoV disease (COVID-19) have been identified exceeding the total numbers of
infected and died patients from SARS and the Middle East respiratory syndrome (MERS). The
virus transmits mainly through respiratory droplets and can be asymptomatic for 2-14 days[2].
Approximately 80% of the patients present with mild symptoms and the overall mortality rate is
nearly 5.6%, but it reaches >14% in some countries such as Italy and the UK, mainly due to the
old-age population[3]. Undoubtedly, SARS-CoV-2 is now much more hazardous than expected
compared to previous outbreaks such as the H1N1 pandemic, which caused 12,429 deaths over a
year while SARS-CoV-2 caused more than 13,000 over 5 weeks in the USA[4]. Notably,
commonly reported complications are pneumonia, acute respiratory distress syndrome (ARDS),
RNAaemia, neurological complications, and multi-organ failure that may primarily begin with
fever, cough, myalgia, and fatigue[5,6]. Developing severe COVID-19 is linked to many
possible risk factors and populations, including old-aged, hypertensive, diabetic, and/or
immunocompromised patients[7].
Unfortunately, there are no available vaccines or specific antiviral drugs for COVID-19 patients.
Consequently, the current treatment protocols are depending mainly on supportive treatment and
trying previously used drugs. Undoubtedly, underscoring the use of an old drug as an antiviral
treatment is an interesting strategy due to our experience on the posology, safety profile, side
effects, as well as drug interactions of these drugs. Phylogenetic analysis reveals that SARS-
CoV-2 has 79% identity to SARS-CoV and 50% identity to MERS-CoV[8,9], thus they share
some similar genetic and clinical characteristics. Subsequently, researchers started to try
clinically accessible drugs previously suggested for SARS- and MERS-CoVs. Thence, we here
represented possible therapeutic agents that may stand as a potential therapy against COVID-19
and shed light on their adverse effects and potential toxicities (Figure 1 and Table 1). We have
ACCEPTED MANUSCRIPT
searched PubMed, Google Scholar, and Clinicaltrials.gov for relevant papers discussing possible
therapeutic agents for COVID-19 from inception to date.
2. Antimicrobial Agents
2.1. Nucleoside Analogues (Polymerase Inhibitors)
Ribavirin is a guanosine analogue that has antiviral activity against hepatitis C virus, respiratory
syncytial virus, and viral hemorrhagic fevers[10]. It acts mainly through incorporation into viral
RNA, instead of guanosine, and this is subsequently toxic to the viral genome. Ribavirin has
been given with interferon-α to MERS-CoV patients[11], however, it failed to show response
among critically-ill patients[12]. Meanwhile, it showed uncertain clinical improvement when
used at an earlier stage[13]. Interferon-α is usually given with ribavirin to enhance and modulate
host immunity and because ribavirin was found to be not able to reduce viral RNA when given
alone[14,15]. Regarding COVID-19, a high dose of ribavirin found to limit viral infection in
vitro[16]. Thus, ribavirin might have the potential to act against COVID-19, yet this requires
large-sized clinical trials, however, caution should be taken as ribavirin has an associated risk of
hemolytic anemia and significant hemoglobin reduction. Moreover, it may result in fatigue, rash,
leukopenia, and teratogenicity[13].
Favipiravir is another guanine analogue that may act through distinctive mechanisms, including
inhibiting RNA polymerase and inducing toxic mutations in the viral genome[17]. Favipiravir
showed antiviral activity against influenza A H1N1, yellow fever, and Ebola[17–19].
Interestingly, Wang and colleagues reported that a high dose of favipiravir was able to reduce
COVID-19 loads in vitro[16]. Further, in a randomized controlled trial (RCT) of COVID-19
patients, favipiravir was compared to arbidol. The outcome of recovery was determined at day
seven post-treatment administration as return of oxygen saturation, respiratory rate, and body
temperature to their normal ranges. On the seventh day, 61.2% of patients who received
favipiravir showed clinical recovery, while only 51.7% of patients of the arbidol group
recovered. However, this difference in recovery rate was not significant. Additionally,
ACCEPTED MANUSCRIPT
favipiravir significantly relieved cough among the favipiravir group. Favipiravir is generally
tolerable; however, it is noteworthy that patients who took favipiravir experienced some adverse
events, including gastrointestinal track disturbance (13.79%), elevation of liver function
enzymes (8.62%), and psychiatric symptoms (4.31%)[20].
The therapeutic antiviral potential of remdesivir was evaluated against Ebola and Nipah viruses,
filoviruses, pneumoviruses, paramyxoviruses, as well as MERS- and SARS-CoVs[21–23]. Its
active form is analogous to adenosine, acting through inhibition of RNA polymerase and
therefore inhibiting viral replication[24–26]. It was reported to be highly potent in vivo against
SARS-CoV-2[16]. A recent study demonstrates in simian Vero E6 cells infected with SARS-
CoV-2 that remdesivir is inhibitive against SARS-CoV-2 at EC90 of 1.76 µM, a concentration
achieved in vivo in nonhuman primate models[26,27]. It has been demonstrated that remdesivir
effectively inhibited SARS-CoV-2 of human liver cancer Huh-7 cells also, which are sensitive
to SARS-CoV-2[16]. The prophylactic and therapeutic efficacious effect of remdesivir has been
shown recently against MERS-CoV in rhesus monkeys[28]. Prophylactic therapy of rhesus
monkeys with remdesivir initiated 12hours or 24hours before MERS-CoV inoculation
prevented virus-induced disease entirely, or provided a significant clinical benefit, with a
reduction in clinical signs, reduced virus replication, and prevented the lung lesions or decreased
its severity[28]. Moreover, Holshue and coworkers reported that they used remdesivir in treating
a 35-old man infected with SARS-CoV-2. The patient’s health was promoted with remdesivir
and supportive therapy[29]. In addition to that, a cohort of 53 critically-ill COVID-19 patients,
they received remdesivir with supportive measures. Of these 53 patients, 30 patients were
intubated on mechanical ventilation. On follow-up, 68% of the patients improved clinically,
including 17 of the intubated patients were extubated. After then, 47% were successfully
discharged[30]. There is very limited evidence on remdesivir safety, however, in an RCT of
patients with Ebola virus, one patient who took remdesivir had hypotension and cardiac arrest
following remdesivir cessation[31]. Yet, it was not clear if this effect was related to the
ACCEPTED MANUSCRIPT
discontinuation of remdesivir or the Ebola virus. A recent trial with remdesivir on a
compassionate use to 53 COVID-19 patients receiving oxygen support or mechanical ventilation
showed that 200mg intravenous remdesivir at day 1, followed by 100mg daily for 9 days,
resulted in clinical improvement in 68% of the patients[30]. Yet, the mortality rate was 18%
among patients receiving invasive ventilation and 5% among patients not receiving invasive
ventilation[30], suggesting that remdesivir is a good agent for COVID-19 patients not receiving
invasive ventilation. On the contrary, a recent study in China evaluated the role of remdesivir
and found that it was not associated with a difference in time to clinical improvement. Yet,
patients receiving remdesivir had an insignificant faster time to clinical improvement than those
receiving placebo among patients with symptom duration of 10 days. Besides, side effects were
revealed in 66% of remdesivir versus 64% of the placebo group while the 28-day mortality was
similar between the two groups (14 versus 13%, respectively). Consequently, remdesivir was
stopped early because of adverse events in 12% of patients versus 5% of patients who stopped
placebo early[32]. In a recent randomized open-label study, 200 severe patients received
remdesivir for 5 days, meanwhile, 197 patients took a 10-day course of remdesivir. There was
no significant difference in clinical improvement between both remdesivir treatment
courses[33]. Reported side effects in COVID-19 studies include nausea, constipation,
respiratory failure, and blood biomarkers of organ impairment, including low albumin, low
potassium, low red blood cells, and platelets that help with clotting, and jaundice. Moreover,
gastrointestinal problems, elevated liver enzymes, and infusion site reactions were reported as
well[33,34].
Of note, penciclovir was one of the first agents that were evaluated against SARS-CoV-2. It is
usually used against herpes viruses as it is a guanosine analogue that inhibits herpes DNA
polymerase enzyme, consequently, it inhibits virus replication. The in vitro study done by Wang
et al. assessed the efficacy of penciclovir against isolates of SARS-CoV-2. Penciclovir was
found to have a half-maximal effective concentration (EC50)=95.96 μM; indicating a relatively
ACCEPTED MANUSCRIPT
low efficacy against SARS-CoV-2[16,26]. Another group of researchers found that penciclovir
could effectively inhibit the RNA-polymerase of SARS-CoV-2, suggesting that it may have the
potential to be further investigated in clinical trials[35]. However, penciclovir is not absorbed
well when taken orally. Moreover, there is limited evidence regarding its safety in pregnancy
and breastfeeding with headache and nausea as the most common side effects[36].
2.2. Protease Inhibitors
Camostat mesilate, or camostat mesylate, is a synthetic serine protease inhibitor developed for
oral squamous cell carcinoma, and dystrophic epidermolysis. In a trial, 95 patients received
200mg camostat mesilate three times daily for 2 weeks and showed only mild, but no severe
side effects in patients with dyspepsia associated with non-alcoholic mild pancreatic disease[37],
demonstrating that camostat mesilate is a well-tolerated intervention. In Japan, nafamostat
mesilate, as a clinically confirmed and synthetic serine protease inhibitor, was approved for the
treatment of acute pancreatitis, disseminated intravascular coagulation, and for anticoagulation
in the extracorporeal circulation[38–40].
In a screening of 1,100 drugs approved by the FDA, nafamostat mesilate was shown to inhibit
spike protein-mediated viral membrane fusion with transmembrane protease-serine 2
(TMPRSS2)-expressing lung Calu-3 host cells by inhibiting TMPRSS2 protease activity in
MERS-CoV[41]. Since the spike proteins of MERS-CoV and SARS-CoV-2 have similar
characteristics, nafamostat mesilate showed inhibiting action against SARS-CoV-2 in simian
Vero E6 cells[16], proposing that nafamostat mesilate can protect against the SARS-CoV-2
infection. Moreover, nafamostat mesilate was administered intravenously at 240mg per day for
5 days without severe side effects in a phase 2 RCT in severe acute pancreatitis patients[38].
Darunavir, an FDA approved antiviral agent for HIV, is an inhibitor of HIV protease
enzyme[42]. Darunavir is usually combined with other drugs such as cobicistat or ritonavir[43].
This addition leads to increased half-life and serum level of darunavir because cobicistat
suppresses CYP3A4; an enzyme that normally metabolizes darunavir. A computational-based
ACCEPTED MANUSCRIPT
study identified darunavir as a potential inhibitor of SARS-CoV-2 replication. It also suggested
that darunavir might, somehow, target the spike protein of SARS-CoV-2[44]. Of note, darunavir
is mostly tolerable with rash, nausea, and diarrhea as the commonest adverse events.
Notwithstanding, darunavir should be carefully monitored in patients with hepatitis since its
hepatotoxic effect was documented[45–47]. To date, there is limited evidence about the efficacy
of darunavir/cobicistat combination for COVID-19 patients. Therefore, we encourage
conducting further preclinical and clinical studies.
Furthermore, lopinavir/ritonavir combination has been thought to have promising antiviral
activity against SARS-CoV-2. The combination was initially developed for HIV[48]. Besides,
its efficacy was tested for SARS-, MERS-CoVs, and SARS-CoV-2[27,49,50]. Lopinavir acts
through inhibiting the HIV-protease enzyme; a vital enzyme for the virus life cycle[51] while,
ritonavir; a CYP3A4 inhibitor, was added to increase efficacy and serum levels of lopinavir[52].
Chu et al. showed in vitro antiviral activity of lopinavir/ritonavir combination against SARS-
CoV[49]. Besides, Chan et al. concluded that animals infected with MERS-CoV had lower viral
loads when treated with lopinavir/ritonavir than control animals[53]. However, another group of
researchers demonstrated that the combination has no in vitro activity against SARS-CoV[54].
Furthermore, another researchers’ group showed that lopinavir/ritonavir combination, alone, has
no role in reducing viral loads of MERS-CoV. They also showed that INF-β may be superior to
this combination in terms of reducing viral loads. Moreover, they concluded that a combination
of remdesivir and INF-β is superior, as well, to lopinavir/ritonavir combination[55]. As regard to
COVID-19, lopinavir/ritonavir revealed significant antiviral activity in three Chinese
patients[56]. Another case report showed that lopinavir/ritonavir significantly reduced viral
loads in a 54-year old Korean patient[57]. On the other side, a group of researchers conducted an
RCT to investigate lopinavir/ritonavir efficacy against COVID-19. They administered
lopinavir/ritonavir to 99 patients while standard treatment was given to 100 patients. They
reported that they found no significant difference between the two groups in terms of clinical
ACCEPTED MANUSCRIPT
improvement, mortality rate, and viral load reduction[58]. The safety profile of
lopinavir/ritonavir should not be ignored since many side effects were frequently reported
including nausea, diarrhea, and asthenia[58,59].
2.3. Other Antimicrobial Agents
Glycyrrhizin is used clinically for a long time due to having anti-inflammatory,
immunosuppressive[60], anti-tumor, and antiviral properties against several viruses such as
flaviviruses[61], and hepatitis C[62], herpes[63], as well as human immunodeficiency
viruses[64]. Of note, it is relatively nontoxic and was found to exhibit activity against SARS-
CoV in vitro[65] as well as clinically with a selectivity index (ratio of antiviral activity to
toxicity) of 67[60]. Cinatl et al. evaluated the antiviral potential of glycyrrhizin, ribavirin,
pyrazofurin, 6-azauridine, and mycophenolic acid against clinical isolates from SARS patients
and it was the most potent in the suppression of the SARS-CoV replication[60]. It suppressed
viral adsorption and penetration and was most effective when administered both during and after
the viral adsorption period[60]. It was proposed that its mode of action is mediated by inducing
the nitrous oxide pathway and also has an effect on cellular signaling pathways such as protein
kinase C, casein kinase II, as well as transcription factors[60,66]. Glycyrrhizin may be a
potential therapeutic agent for SARS-CoV-2 infection with proper monitoring since its known
adverse events (hypokalemia, hypertension, and irregular heart rhythm)[67] can be controlled
despite its administration in high dosage in clinical trials, unlike the ribavirin which had many
adverse events such as hemolysis after being administrated to SARS patients[60].
At the beginning of the SARS-CoV-2 outbreak, light has been shed over chloroquine, an
antimalarial drug, especially after an article demonstrated an in vivo high selectivity index of
chloroquine against SARS-CoV-2[16,26]. In a small RCT of 30 COVID-19 patients,
hydroxychloroquine was compared to standard supportive care. The authors did not find any
significant difference between both groups in terms of viral clearance and clinical response[68].
Meanwhile, a letter by Gao et al. reported, without patients’ data, that chloroquine phosphate
ACCEPTED MANUSCRIPT
was found to be superior to the control group, in terms of improving pneumonia and assisting
patients’ recovery[69]. Moreover, an expert consensus recommended a ten-day course of 500
mg of chloroquine phosphate twice daily[70]. Another in vitro study compared
hydroxychloroquine with chloroquine and suggested the superiority of hydroxychloroquine over
chloroquine[71], especially because it is safer, cheaper, and suitable for pregnant women.
Additionally, in a small non-randomized study, researchers reported that 14 of 20 patients,
treated with hydroxychloroquine, had negative PCR on the sixth day following the drug
administration. Meanwhile, only 2 of 16 patients had negative results in the control group on the
sixth day. They also reported that azithromycin augmented the therapeutic effect of
hydroxychloroquine on six patients compared to hydroxychloroquine alone[72]. Indeed, the use
of chloroquine had been initially supported by in vitro and weak human studies and some
countries have put it in their guidelines although both could lead to QT prolongation separately
and, thus if combined with azithromycin, they may lead to serious adverse events[73,74].
Previous reports suggested that inhibiting T-helper cell proliferation and interleukin-2 (IL-2)
production or responsiveness may boost the inflammatory response accidentally, subsequently,
it could affect patients’ outcomes unfavorably[75,76] as it was in Chikungunya viral infection.
Moreover, such combination in overdoses is extremely toxic, especially in patients with
preexisting cardiovascular problems, and could lead to hypoglycemia, neuropsychiatric effects,
drug-drug interactions, and idiosyncratic hypersensitivity events. Later on, the findings of
another uncontrolled observational study of mildly affected 80 COVID-19 patients receiving
hydroxychloroquine with azithromycin were reported[72]. Of these patients, 81.3% discharged
from the hospital, three patients were admitted to the intensive care unit (ICU), one patient died
while 14 patients were still hospitalized at the study endpoint[77]. Another observational
retrospective one-armed study of 1,061 patients, who were given hydroxychloroquine plus
azithromycin, reported that 91.7% of the patients had a good clinical and virological outcome in
10 days with the observation of mild adverse effects, including gastrointestinal, transient blurred
vision, headache, and insomnia in 2.3% of patients[78]. On the other hand, 92 COVID-19
ACCEPTED MANUSCRIPT
patients requiring oxygen therapy was administered hydroxychloroquine and compared to 89
controlled patients who did not receive hydroxychloroquine. There was no difference between
the overall survival at day 21 between both treatment and control groups (89 and 91%,
respectively)[79]. Similarly, in patients with persistent mild to moderate COVID-19,
hydroxychloroquine plus standard of care did not lead to a significantly higher probability of
negative conversion than the standard of care alone. Moreover, the hydroxychloroquine group
had higher adverse events than the control group[80]. In addition, an analysis of 96,032 COVID-
19 patients’ data revealed an increased risk of in-hospital mortality with either
hydroxychloroquine or chloroquine with or without a macrolide; compared to control patients
who were not given any of these therapeutics[81]. In a double-blinded RCT of hospitalized
COVID-19 patients, two different dosages of chloroquine, (600 mg twice daily for 10 days
compared to 450 mg twice on day 0 then 450 mg once for four days), were given as adjuvant
therapy to the standard care for both treatment arms, the researchers had to discontinue the trial
because of the observed high fatality rate among higher dosage arm (10-day chloroquine course)
and recommended against giving a high dose of chloroquine to COVID-19 patients[82].
Nitazoxanide is another potential therapeutic for COVID-19. It has shown a broad spectrum of
anti-parasitic and antiviral activity acting against rotavirus, norovirus, chronic hepatitis B,
influenza, respiratory syncytial virus, canine CoVs through distinctive mechanisms[83–87]. It
was found to overexpress interferon regulatory factor-1 that inhibited the replication of human
norovirus[87]. Moreover, it was found to be active against MERS-CoV, murine, and bovine
CoVs through inhibition of the viral N protein expression[86]. Additionally, it suppresses the
production of inflammatory cytokines, including IL-2, IL6, and IL-10[88]. Besides, the in vitro
efficacy of nitazoxanide against SARS-CoV-2 was reported[16,26]. The most commonly
reported side effects of nitazoxanide are mild, including headache, skin rash, fever, vomiting,
and abdominal pain[89,90]. To date, 11 registered trials are trying to investigate its safety and
ACCEPTED MANUSCRIPT
efficacy in COVID-19 patients either alone or in combination with ivermectin or
hydroxychloroquine with their findings yet to be revealed[91].
Umifenovir (known as arbidol) has a wide range of antiviral activity against influenza A and
B[92]. It acts mainly through inhibition of membrane fusion between the viral envelope and host
cell membrane, therefore, preventing viral entry and infectivity[93]. Arbidol, also, may enhance
the host immune response by activation of macrophages and induction of interferons[92]. In
vitro studies showed that arbidol has antiviral activity against hepatitis B, C, and Ebola viruses,
poliovirus, as well as human herpes virus-8[94,95]. This broad-spectrum antiviral activity has
made arbidol standing as a potential therapeutic against SARS-CoV-2. It is worth mentioning
that arbidol may have a synergistic effect when combined with an immunomodulatory drug
(IMOD); a drug used in HIV patients to induce the production of CD4 lymphocytes and
interferon gamma[96]. Moreover, Deng et al. conducted a retrospective cohort study, in which
16 patients received a combination of lopinavir/ritonavir plus oral arbidol compared to a group
of 17 patients who received only lopinavir/ritonavir. They found that viral loads disappeared
after 14 days in 15 of 16 and 7 of 17 patients in lopinavir/ritonavir and arbidol versus in
lopinavir/ritonavir only group, respectively[97]. Notably, arbidol is generally safe; with
gastrointestinal disturbance symptoms as the most common adverse events[98]. Currently,
registered clinical trials are investigating its safety and efficacy and comparing it to standard
care or lopinavir/ritonavir combination. We also encourage conducting more trials, however, we
also suggest evaluating its efficacy when combined with IMOD in COVID-19 patients.
Furthermore, ivermectin, an approved antiparasitic medication[99], has been proposed as a
potent inhibitor against COVID-19. Ivermectin had an in vitro antiviral activity against HIV-1
and dengue virus[100,101]. It mitigates West Nile and influenza viruses as well[102,103]. It acts
mainly by inhibiting the viral nuclear import and therefore inhibit its replication. Interestingly,
Caly et al. reported that ivermectin was able to eliminate all RNA from infected cells with
SARS-CoV-2 in 48 hours. They hypothesized that ivermectin has a high probability of being a
ACCEPTED MANUSCRIPT
strong inhibitor of COVID-19 replication in human patients, especially when given early. They,
also, found no adverse effects on tested cells[104]. However, it should be tried with caution
because it may need very high doses to work and its safety in pregnancy has not been yet
concluded[105]. Moreover, severe side effects about potentiation of the GABAergic synaptic
transmission, and causing ataxia, psychosis, depression, confusion, and convulsions were found
in a few patients[106].
Baloxavir marboxil, a potent anti-influenza drug, inhibits viral replication by inhibiting the
protein synthesis, which is a different mode of action from neuraminidase inhibitors like
oseltamivir. Thus, it was demonstrated that the baloxavir marboxil/oseltamivir combination has
a synergistic antiviral effect in experimental and clinical studies[107,108]. They also
demonstrated no side effects nor effect on their shared pharmacokinetics with no need for dose
adjustment when combined[108].
3. Targets for Angiotensin-Converting Enzyme (ACE)
ACE-II receptor has been established as a primary receptor for SARS-CoV-2. Kuba et al.
noticed a reduction in ACE-II expression, in the lung tissue, indicating that this reduction may
play a role in lung injury mediated by SARS-CoV[109]. This finding was consistent in an in
vivo study conducted on a mouse infected with influenza A as well. Notably, the administration
of recombinant ACE-II enhanced the survival of the mice, lessened lung edema, and improved
lung histopathology and function, and lessened lung edema[110]. Indeed, it does not only
compensate for the low lung ACE-II expression, but also might act, theoretically, as a decoy to
SARS-CoV-2, and subsequently naturalizing the virus through binding to the virus instead of
host receptors. Therefore, we encourage conducting RCTs investigating its safety and efficacy in
patients with COVID-19. Nevertheless, attention should be directed, primarily, to address its
safety. In hemodynamically-stable patients with ARDS, recombinant ACE-II was administered
in a pilot clinical trial. Although there were no negative hemodynamic effects, dysphagia, rash,
and acute renal failure were documented[111].
ACCEPTED MANUSCRIPT
ACE-II inhibitors were suggested as possible medications for COVID-19[112], especially in
patients with hypertension, it was noted that they reduced the pulmonary inflammatory response
and mortality. Notwithstanding, they could result in a compensatory increase in ACE-I by
negative feedback due to downregulating ACE-II, consequently that will lead to more ACE-II
resulting in excessive binding to ACE-II receptor in the lungs which increases pulmonary
vascular permeability, ultimately that results in pulmonary edema.
4. Immunotherapy and Immunomodulators
Convalescent plasma is another potential therapy against COVID-19 as well. Indeed,
convalescent plasma was suggested that it may have a good impact on SARS-CoV
patients[113,114]. It is noteworthy that the WHO recommended the use of convalescent plasma
during Ebola outbreaks[115]. It is worth mentioning as well that improvement in clinical status
in five critically ill patients with COVID-19 and ARDS was noted following the administration
of convalescent plasma containing neutralizing antibodies[116]. In contrast, in an RCT by Li et
al. who added convalescent plasma to standard treatment, compared with standard treatment
alone, did not see a statistically significant improvement in time to clinical improvement within
28 days. Of note, it was associated with some clinical improvement in severely ill patients, but
not in critically ill patients which could be because antibody therapies generally work best when
administered earlier in the disease course[117,118]. Further, its application still has many
setbacks including the accessibility to sufficient donors, risk of disease transmission, as well as,
its adverse events early after transfusion such as nausea, fever, and skin rash[119].
Monoclonal antibodies (MABs) have been suggested as a potential therapeutic for COVID-19
based on the previous evidence of their promising effect on SARS- and MERS-CoV in vivo and
in vitro[120–124]. MABs, indeed, are preferred over convalescent plasma in terms of being
specific with no risk of transmitting blood-borne infections. The mechanism of MABs is mainly
targeting a specific protein, especially the receptor-binding domain (RBD) of the S1 subunit,
thereby, blocking its binding to the host receptor, subsequently preventing viral entry[125,126].
ACCEPTED MANUSCRIPT
Another possible target is the S2 subunit which is responsible for fusion between the virus and
host cell membrane. Indeed, several MABs were previously produced to target the RBD of S1
and S2 subunits of SARS-CoV. Their preclinical studies showed promising potential treating
and preventive effects[120–124]. Therefore, owing to the similarity between the spike protein of
SARS- and SARS-CoV-2, it is worth investigating their effect on COVID-19 patients. However,
their main challenge is their production for being not cost-effective and laborious.
The so-called cytokine storm in severely affected patients has highlighted the rationale to
investigate the efficacy of therapeutic agents directed against cytokines. IL-6 antibody was
detected to be one of the highest cytokines among ICU COVID-19 patients with cytokine storm.
Tocilizumab, a monoclonal antibody, antagonizes IL-6 by binding to both serum and
transmembrane IL-6 receptors; preventing IL-6 from binding to its receptors and thereby
preventing its inflammatory-mediated response[127]. Tocilizumab is the first approved anti-IL-6
monoclonal antibody. It is used mainly in rheumatoid disease[128], systemic juvenile idiopathic
arthritis[129], steroid-refractory chronic graft-versus-host disease[130], and cytokine release
syndrome[127]. Xu et al. reported the use of tocilizumab in 21 Chinese patients with severe
COVID-19. Nineteen patients had lung opacities observed in their CT chest. The fever was
resolved in all patients in the first day post-tocilizumab administration. All patients had rapid
clinical improvement and discharged from the hospital with no observed tocilizumab-related
adverse events[131]. Another prospective study of 100 critical COVID-19 patients found
consistent results of rapid clinical response[132]. It has been recommended that tocilizumab
dose better to be repeated, even in COVID-19 patients who failed to show a response after the
first dose, to get a higher response[133]. Although tocilizumab has a tolerable profile in previous
indications, caution should be taken as septic shock developed in two COVID-19 patients
besides gastrointestinal perforation that occurred in one COVID-19 patient[132]. Moreover,
tocilizumab could be associated with an increased risk of opportunistic infections, leukopenia,
and/or lymphopenia. Furthermore, like other MABs, the high cost of tocilizumab might be
ACCEPTED MANUSCRIPT
challenging as well. Many registered trials are still trying to further investigate the safety and
efficacy of tocilizumab either alone or in combination with other agents such as favipiravir[134].
Other anti-IL-6 agents, including olokizumab and siltuximab, are being investigated with their
final results yet to be revealed.
In addition to that, there is rising limited evidence about the use of mesenchymal stem cells
(MSCs) in treating COVID-19-associated pneumonia. This suggestion is based on the
immunomodulatory function of MSCs. It is believed that MSCs, when injected intravenously,
may secrete anti-inflammatory paracrine factors, and therefore, they may counteract the
damaging cytokine storm of COVID-19. Additionally, they may reach the pulmonary circulation
and protect and/or regenerate the damaged pulmonary epithelial cells[135]. The effectiveness of
MSCs was previously documented in patients with acute graft-versus-host disease unresponsive
to steroid therapy, as well as, patients with systemic lupus erythematosus (SLE)[136,137].
Meanwhile, in patients with moderate-to-severe ARDS, MSCs infusion showed general
tolerability, yet, they had low efficacy that still to be investigated further[138,139]. Liang et al.
reported the use of MSCs in a critically-ill female patient affected with COVID-19. They found
that MSCs improved the clinical outcome of the patient with no adverse events[140]. Another
study enrolled seven patients, with COVID-19, and administered MSCs to them. They noticed a
modulatory effect on them, in terms of the disappearance of cytokine-producing cells. Also, they
noticed the correction of COVID-19 associated lymphocytopenia. Subsequently, the health of
the seven patients was improved[141].
Baricitinib is proposed as an immunomodulatory via inhibiting the cytokine storm caused by
COVID-19 and consequently the extensive inflammation of the lung tissue. It mostly inhibits
JAK 1 and 2 enzymes and subsequently inhibiting cytokines’ gene expression. It also might
inhibit AP2-associated protein kinase 1, an enzyme that promotes virus entry in the host cells
and the intracellular assembly of virus particles[142–144]. A caution, however, is needed when
using immunomodulators with immunocompromised patients whereas the commonest side
ACCEPTED MANUSCRIPT
effects are upper respiratory tract infections, nausea, cold sores from herpes simplex, and
shingles from herpes zoster[145].
Another proposed treatment is interferons. Interferon-α has been usually used in the treatment of
viral hepatitis[146] and some various types of cancers, including leukemia and
melanoma[147,148]. Interestingly, interferon-α was found to be effective in vitro against SARS-
CoV but with a low selectivity index than interferon-β[149]. In addition to that, a retrospective
cohort study reported that a combination of interferon-α and ribavirin improved the clinical
status of patients infected with MERS-CoV[150]. Fatigue, anorexia, and weight loss are among
the most common side effects of interferon-α but, thankfully, dose-limited[146]. Currently, its
safety and efficacy are being evaluated in clinical trials for both treating COVID-19[151,152]
and, possibly, a way of enhancing immunity and preventing COVID-19 among medical
staff[153].
It is worth mentioning that it is advised against the use of corticosteroids in COVID-19 affected
patients[154]. Corticosteroids may suppress lung inflammation in theory; however, clinical
outcomes were found to be worse in patients affected with either SARS-, or MERS-
CoVs[155,156].
5. Organ Function Support
5.1. Artificial Liver Blood Purification System (ALPS)
ALPS showed the ability to clear the serum cytokine storm which has the main role of
developing respiratory failure through the severe inflammatory response. Previously, ALPS
revealed its efficacy when used in severe H7N9 influenza critically-ill patients with cytokine
storm[157]. For COVID-19, ALPS blocked the cytokine storm successfully in severely affected
Chinese patients and supported, in part, their clinical status[158]. ALPS is indicated if serum
cytokines level is five times the upper-normal level given the daily deterioration of the lung on
imaging[159]. Yet, the high cost and less availability of ALPS make it to be used in strict
situations and upon indications.
ACCEPTED MANUSCRIPT
5.2. Extracorporeal Membrane Oxygenation (ECMO)
ECMO was previously used in the influenza A pandemic[160] and MERS-CoV[161] with
promising results. ECMO simply removes CO2 from the patient’s blood and adds oxygen,
therefore allowing a better chance for the lungs to improve under treatment. In a retrospective
cohort study on severely affected MERS patients, ECMO was placed to 17 patients in contrast to
18 control patients indicated for ECMO but it was not available for them. The ECMO group had
a significantly better PaO2/FiO2 ratio than the control group on day 14 (138 and 36,
respectively). Moreover, the ECMO group had lower mortality than the control group[161].
ECMO is reserved for COVID-19 patients with severely refractory hypoxemia. This effective
cardiopulmonary support method is, however, very limited, needs high expertise level, and
unfortunately can be a source of SARS-CoV-2 transmission between patients and treating
physicians. Hence, physicians are advised to take strict infection-control measures when dealing
with ECMO-treated patients.
6. Other Therapeutic Agents
We also highlight the importance of adding prophylactic anticoagulants in critically-ill patients
admitted to the ICU. There are two reasons behind this suggestion; firstly, owing to the
previously documented increased risk of deep venous thrombosis and pulmonary embolism in
long-time hospitalized patients[162]. Secondly, antiphospholipid antibodies can temporarily
appear in the serum of critically-ill patients with viral infections[163]. The observed adversely-
affected outcome in patients with COVID-19-related hypercoagulable state, emphasizes the
importance of prophylactic anticoagulants. Moreover, in COVID-19 mechanically-ventilated
patients, the administration of systemic anticoagulants was associated with lower in-hospital
mortality rate compared to patients who did not take systemic anticoagulants (29.1 and 62.7%,
respectively). However, for all hospitalized patients including patients who did not require
mechanical ventilation, there was no difference in in-hospital mortality[164]. Another
retrospective study of 499 severe COVID-19 patients reported a significant difference in
ACCEPTED MANUSCRIPT
mortality rate between patients (with >6 fold of upper normal D-dimer) who received low-
molecular-weight heparin (LMWH) compared to patients who did not take anticoagulant
therapy (32.8 and 52.4%, respectively)[165]. Indeed, LMWH, such as enoxaparin, acts by
inhibiting factor Xa, is preferred to use over unfractionated heparin (UFH) that acts through
binding to antithrombin III that consequently inhibits thrombin and factor Xa leading to
immediate anticoagulant action[166]. The preference of LMWH to UFH is mainly because there
is no laboratory monitoring needed. Further, resistance to UFH was observed in 80% of ICU-
admitted COVID-19 patients[167]. LMWH prophylaxis dose is indicated for all hospitalized
COVID-19 patients. It is worth mentioning if the creatinine clearance is 15-30 ml/minute; the
dose should be lowered to 30 mg once per day[168]. Yet, for patients with creatinine clearance
<15/minutes, UFH should be used instead of LMWH. Two COVID-19 patients were taking
prophylactic LMWH dose, however, they developed acute limb ischemia[169]. This may
indicate that prophylactic dose might not be effective in patients with a high D-dimer level,
thereby therapeutic doses should be given instead in those patients. Both UFH and LMWH are
associated with the risk of bleeding and heparin-induced thrombocytopenia (HIT), especially
UFH[170,171]. Thence, fondaparinux, an inhibitor of activated factor Xa, can be used instead of
heparin in patients who developed and/or have a history of HIT[172].
Dayrit and colleagues have interestingly proposed using coconut oil and its derivatives in
treating patients infected with COVID-19[173]. Indeed, coconut oil was observed to has
antiviral activity against HIV with, notably increased counts of CD4 and CD8 six weeks after its
administration[174,175]. Its antiviral activity is thought to be due to inhibition of viral
replication, disrupting viral binding to host cells, as well as, damaging the viral envelope. Based
on its reported viral activity, availability, and being safe, clinical trials of coconut oils against
COVID-19 are recommended.
Hesperidin is a natural derivative of citrus fruits. It has shown a protective effect against
influenza A virus at the early stage of infection through inhibition of viral neuraminidase
ACCEPTED MANUSCRIPT
(sialidase) enzyme, important for the virions release from infected cells[176]. A group of
researchers investigated its efficacy against SARS-CoV-2. Interestingly, hesperidin bound to
RBD of SARS-CoV-2 spike protein, and therefore inhibited the interaction between ACE-II
receptor and spike protein. This step, subsequently, inhibited viral entry inside host cells. This
finding encourages conducting more trials on hesperidin, because it is cheap, available, with no
adverse effects, except for the low incidence of nausea and vomiting[177].
It is noteworthy that we encourage conducting trials on the efficacy of live recombinant measles
vaccine (rMV) for SARS-CoV-2. Notably, a rMV, that expresses the RBD of S1 subunit of the
spike protein of SARS, was reported to be able to induce a highly robust humoral response
enabling cross-reactivity and protection against SARS, with good safety profile[178–181].
Nevertheless, this vaccine is contraindicated in pregnancy[182]. It also may result in
thrombocytopenia and febrile seizures, yet with, thankfully, low incidence[183]. The
aforementioned in vitro findings of rMV encourage giving more attention to target the spike
protein of SARS-CoV-2.
We also suggest giving COVID-19 patients influenza and pneumococcal vaccinations. In
particular, high-risk patients, including patients with severe comorbidities and elderly patients.
This will reduce the risk of developing concurrent respiratory infections. The same suggestion
was reported before among patients with SLE[184,185]. Chang et al. analyzed the risk of
mortality and morbidity in SLE patients who took and who did not take influenza vaccinations.
Vaccinated patients had fewer times of hospitalization, and ICU admission, as well as a lower
mortality rate[186]. Owing to the safety profile of influenza and pneumococcal vaccinations,
except for the reported adverse events of headache, injection site reactions, and systemic
reaction[187], it is advised that all COVID-19 patients may benefit from their vaccination
immune response even patients who are taking immunomodulatory agents since the
immunogenicity of these vaccinations were previously found to be still safe and immunogenic
with immunosuppressive therapeutics except for rituximab[184,188–192].
ACCEPTED MANUSCRIPT
7. Expert opinion
In this review, we assessed the potential therapeutics for COVID-19 patients based on the
previously documented antiviral activity either against SARS- and MERS-CoVs or other viral
infections and based on the ongoing or published SARS-CoV-2 studies. We showed some
agents with potentially favorable efficacy, acceptable safety as well as good pharmacokinetic
profiles in vitro, in vivo and/or in humans. Several therapies are under assessment to evaluate
their efficacy and safety for COVID-19. On the other hand, some drugs have been withdrawn
due to their side effects after demonstrating some clinical promise[193] while COVID-19, as of
today, is essentially untreatable, except for supportive management[194,195]. Indeed, the most
effective therapies could be organ function support, convalescent plasma, anticoagulants, and
antiviral therapy, especially anti-influenza drugs due to the similarities between respiratory
viruses regarding viral entry, uncoating, and replication[26]. We encourage giving more
attention to favipiravir, remdesivir, and measles vaccine. Of note, convalescent plasma was
associated with reduced mortality during the 1918 influenza, SARS, 2009 influenza H1N1, and
Ebola outbreaks[114,196,197]. Ribavirin is a broad-spectrum antiviral drug; yet, the clinical
effects are unclear, and side effects should be considered. Besides, ALPS and ECMO should be
considered under strict indications and contraindications or that lead to a waste of resources and
additional complications. Moreover, tocilizumab likely possesses a better effect than other anti-
IL-6 receptors MABs. Antibiotics for pneumonia are essential in case of bacterial infection only
otherwise it may fuel bacterial resistance.
Undoubtedly, a combination, at least dual or even triple therapy, of the aforementioned
efficacious and safe therapies is greatly recommended for COVID-19 due to the disease nature
but giving great attention that mild, moderate, and severe patients may have different regimens
and combinations based on the disease severity is important as well. Further, patients should
have a routine assessment for their coagulation and bleeding profiles as well as their
inflammatory and cytokine concentrations.
ACCEPTED MANUSCRIPT
Funding
This paper was not funded.
Declaration of interest
The authors have no relevant affiliations or financial involvement with any organization or
entity with a financial interest in or financial conflict with the subject matter or materials
discussed in the manuscript. This includes employment, consultancies, honoraria, stock
ownership or options, expert testimony, grants or patents received or pending, or royalties.
Reviewer disclosures
Peer reviewers on this manuscript have no relevant financial or other relationships to disclose.
ACCEPTED MANUSCRIPT
References
[1] WHO Director-General’s remarks at the media briefing on 2019-nCoV on 11 February
2020.
[2] Singhal T. A Review of Coronavirus Disease-2019 (COVID-19). Indian J. Pediatr.
Springer; 2020. p. 281–286.
[3] Organization WH. Coronavirus disease 2019 (COVID-19): situation report, 77. 2020;
[4] 2009 H1N1 Pandemic (H1N1pdm09 virus) | Pandemic Influenza (Flu) | CDC.
[5] Wang D, Hu B, Hu C, et al. Clinical Characteristics of 138 Hospitalized Patients with
2019 Novel Coronavirus-Infected Pneumonia in Wuhan, China. JAMA - J Am Med
Assoc. 2020;323:1061–1069.
[6] Elgamasy S, Kamel MG, Ghozy S, et al. First Case of Focal Epilepsy Associated with
SARSCoronavirus2. J Med Virol [Internet]. 2020 [cited 2020 Jun 4];jmv.26113.
Available from: https://onlinelibrary.wiley.com/doi/abs/10.1002/jmv.26113.
[7] Zhou F, Yu T, Du R, et al. Clinical course and risk factors for mortality of adult
inpatients with COVID-19 in Wuhan, China: a retrospective cohort study. Lancet.
2020;
[8] Lu R, Zhao X, Li J, et al. Genomic characterisation and epidemiology of 2019 novel
coronavirus: implications for virus origins and receptor binding. Lancet.
2020;395:565–574.
[9] Chan JFW, Yuan S, Kok KH, et al. A familial cluster of pneumonia associated with
the 2019 novel coronavirus indicating person-to-person transmission: a study of a
family cluster. Lancet. 2020;395:514–523.
[10] Snell NJC. Ribavirin - Current status of a broad spectrum antiviral agent. Expert Opin
Pharmacother. 2001;2:1317–1324.
ACCEPTED MANUSCRIPT
[11] Morra ME, Van Thanh L, Kamel MG, et al. Clinical outcomes of current medical
approaches for Middle East respiratory syndrome: A systematic review and meta-
analysis. Rev Med Virol. 2018;
[12] Al-Tawfiq JA, Momattin H, Dib J, et al. Ribavirin and interferon therapy in patients
infected with the Middle East respiratory syndrome coronavirus: An observational
study. Int J Infect Dis. 2014;20:42–46.
[13] Omrani AS, Saad MM, Baig K, et al. Ribavirin and interferon alfa-2a for severe
Middle East respiratory syndrome coronavirus infection: A retrospective cohort study.
Lancet Infect Dis. 2014;14:1090–1095.
[14] Dusheiko G, Main J, Thomas H, et al. Ribavirin treatment for patients with chronic
hepatitis C: Results of a placebo-controlled study. J Hepatol. 1996;25:591–598.
[15] Weiss RC, Oostrom-Ram T. Inhibitory effects of ribavirin alone or combined with
human alpha interferon on feline infectious peritonitis virus replication in vitro. Vet
Microbiol. 1989;20:255–265.
[16] Wang M, Cao R, Zhang L, et al. Remdesivir and chloroquine effectively inhibit the
recently emerged novel coronavirus (2019-nCoV) in vitro. Cell Res. 2020;30:269–271.
[17] Baranovich T, Wong S-S, Armstrong J, et al. T-705 (Favipiravir) Induces Lethal
Mutagenesis in Influenza A H1N1 Viruses In Vitro. J Virol. 2013;87:3741–3751.
[18] Sissoko D, Laouenan C, Folkesson E, et al. Experimental Treatment with Favipiravir
for Ebola Virus Disease (the JIKI Trial): A Historically Controlled, Single-Arm Proof-
of-Concept Trial in Guinea. PLoS Med. 2016;13.
[19] De Clercq E. New Nucleoside Analogues for the Treatment of Hemorrhagic Fever
Virus Infections. Chem. - An Asian J. John Wiley and Sons Ltd; 2019. p. 3962–3968.
[20] Chen C, Huang J, Yin P, et al. Favipiravir versus Arbidol for COVID-19: A
ACCEPTED MANUSCRIPT
Randomized Clinical Trial.
[21] Warren TK, Jordan R, Lo MK, et al. Therapeutic efficacy of the small molecule GS-
5734 against Ebola virus in rhesus monkeys. Nature. 2016;531:381–385.
[22] Sheahan TP, Sims AC, Graham RL, et al. Broad-spectrum antiviral GS-5734 inhibits
both epidemic and zoonotic coronaviruses. Sci Transl Med. 2017;9.
[23] Lo MK, Jordan R, Arvey A, et al. GS-5734 and its parent nucleoside analog inhibit
Filo-, Pneumo-, and Paramyxoviruses. Sci Rep. 2017;7:43395.
[24] Gordon CJ, Tchesnokov EP, Feng JY, et al. The antiviral compound remdesivir
potently inhibits RNA-dependent RNA polymerase from Middle East respiratory
syndrome coronavirus. J Biol Chem. 2020;
[25] Agostini ML, Andres EL, Sims AC, et al. Coronavirus susceptibility to the antiviral
remdesivir (GS-5734) is mediated by the viral polymerase and the proofreading
exoribonuclease. MBio. 2018;9.
[26] Carradori S. Are there any Therapeutic Options Currently Available for Wuhan
Coronavirus? Antiinflamm Antiallergy Agents Med Chem. 2020;19.
[27] Mori M, Capasso C, Carta F, et al. A deadly spillover: SARS-CoV-2 outbreak. Expert
Opin Ther Pat. 2020;1–5.
[28] De Wit E, Feldmann F, Cronin J, et al. Prophylactic and therapeutic remdesivir (GS-
5734) treatment in the rhesus macaque model of MERS-CoV infection. Natl Acad Sci
[Internet]. [cited 2020 May 5]; Available from:
www.pnas.org/cgi/doi/10.1073/pnas.1922083117.
[29] Holshue ML, DeBolt C, Lindquist S, et al. First Case of 2019 Novel Coronavirus in
the United States. N Engl J Med. 2020;382:929–936.
[30] Grein J, Ohmagari N, Shin D, et al. Compassionate Use of Remdesivir for Patients
ACCEPTED MANUSCRIPT
with Severe Covid-19. N Engl J Med. 2020;NEJMoa2007016.
[31] Mulangu S, Dodd LE, Davey RT, et al. A Randomized, Controlled Trial of Ebola
Virus Disease Therapeutics. N Engl J Med. 2019;381:2293–2303.
[32] Wang Y, Zhang D, Du G, et al. Remdesivir in adults with severe COVID-19: a
randomised, double-blind, placebo-controlled, multicentre trial. Lancet. 2020;
[33] Goldman JD, Lye DCB, Hui DS, et al. Remdesivir for 5 or 10 Days in Patients with
Severe Covid-19. N Engl J Med. 2020;NEJMoa2015301.
[34] Mehta N, Mazer-Amirshahi M, Alkindi N, et al. Pharmacotherapy in COVID-19; A
narrative review for emergency providers. Am J Emerg Med. 2020;
[35] Suramin, Penciclovir and Anidulafungin bind nsp12, which governs the RNA-
dependent-RNA polymerase activity of SARS-CoV-2, with higher interaction energy
than Remdesivir, indicating potential in the treatment of Covid-19 infection.
[36] S M, V L, FY A, et al. Famciclovir: A Focus on Efficacy and Safety. Expert Opin
Drug Saf. 2010;9.
[37] Sai JK, Suyama M, Kubokawa Y, et al. Efficacy of camostat mesilate against
dyspepsia associated with non-alcoholic mild pancreatic disease. J Gastroenterol.
2010;45:335–341.
[38] Hirota M, Shimosegawa T, Kitamura K, et al. Continuous regional arterial infusion
versus intravenous administration of the protease inhibitor nafamostat mesilate for
predicted severe acute pancreatitis: a multicenter, randomized, open-label, phase 2
trial. J Gastroenterol. 2020;55:342–352.
[39] IWAKI M, INO Y, MOTOYOSHI A, et al. Pharmacological studies of FUT-175,
nafamostat mesilate V. Effects on the pancreatic enzymes and experimental acute
pancreatitis in rats. Jpn J Pharmacol. 1986;41:155–162.
ACCEPTED MANUSCRIPT
[40] Ohkoshi M, Oka T. Clinical experience with a protease inhibitor [N, N-
dimethylcarbamoylmethyl 4-(4-guanidinobenzoyloxy)-phenylacetate] methanesulfate
for prevention of recurrence of carcinoma of the mouth and in treatment of terminal
carcinoma. J Maxillofac Surg. 1984;12:148–152.
[41] Yamamoto M, Matsuyama S, Li X, et al. Identification of nafamostat as a potent
inhibitor of Middle East respiratory syndrome coronavirus S protein-mediated
membrane fusion using the split-protein-based cell-cell fusion assay. Antimicrob
Agents Chemother. 2016;60:6532–6539.
[42] Leonis G, Czyz̀nikowska Z, Megariotis G, et al. Computational studies of darunavir
into HIV-1 protease and DMPC bilayer: Necessary conditions for effective binding
and the role of the flaps. J Chem Inf Model. 2012;52:1542–1558.
[43] Mathias AA, German P, Murray BP, et al. Pharmacokinetics and Pharmacodynamics
of GS-9350: A Novel Pharmacokinetic Enhancer Without Anti-HIV Activity. Clin
Pharmacol Ther. 2010;87:322–329.
[44] Wu C, Liu Y, Yang Y, et al. Analysis of therapeutic targets for SARS-CoV-2 and
discovery of potential drugs by computational methods. Acta Pharm Sin B. 2020;
[45] Wolfe C, Wolfe C. Profile of darunavir in the management of treatment-experienced
HIV patients. HIV/AIDS - Res Palliat Care. 2009;1:13.
[46] Rachlis A, Clotet B, Baxter J, et al. Safety, tolerability, and efficacy of darunavir
(TMC114) with low-dose ritonavir in treatment-experienced, hepatitis B or C co-
infected patients in POWER 1 and 3. HIV Clin Trials. 2007;8:213–220.
[47] Katlama C, Esposito R, Gatell JM, et al. Efficacy and safety of TMC114/ritonavir in
treatment-experienced HIV patients: 24-Week results of POWER 1. AIDS.
2007;21:395–402.
ACCEPTED MANUSCRIPT
[48] Chandwani A, Shuter J. Lopinavir/ritonavir in the treatment of HIV-1 infection: A
review. Ther. Clin. Risk Manag. Dove Press; 2008. p. 1023–1033.
[49] Chu CM, Cheng VCC, Hung IFN, et al. Role of lopinavir/ritonavir in the treatment of
SARS: Initial virological and clinical findings. Thorax. 2004;59:252–256.
[50] Kim UJ, Won EJ, Kee SJ, et al. Combination therapy with lopinavir/ritonavir, ribavirin
and interferon-a for Middle East respiratory syndrome. Antivir Ther. 2016;21:455–
459.
[51] Flexner C. HIV-protease inhibitors. N. Engl. J. Med. 1998. p. 1281–1292.
[52] Pharmacological and Therapeutic Properties of Ritonavir-Boosted Protease Inhibitor
Therapy in HIV-infected Patients - PubMed.
[53] Chan JF-W, Yao Y, Yeung M-L, et al. Treatment With Lopinavir/Ritonavir or
Interferon-β1b Improves Outcome of MERS-CoV Infection in a Nonhuman Primate
Model of Common Marmoset. J Infect Dis. 2015;212:1904–1913.
[54] Yamamoto N, Yang R, Yoshinaka Y, et al. HIV protease inhibitor nelfinavir inhibits
replication of SARS-associated coronavirus. Biochem Biophys Res Commun.
2004;318:719–725.
[55] Sheahan TP, Sims AC, Leist SR, et al. Comparative therapeutic efficacy of remdesivir
and combination lopinavir, ritonavir, and interferon beta against MERS-CoV. Nat
Commun. 2020;11.
[56] Wang Z, Chen X, Lu Y, et al. Clinical characteristics and therapeutic procedure for
four cases with 2019 novel coronavirus pneumonia receiving combined Chinese and
Western medicine treatment. Biosci Trends. 2020;14:64–68.
[57] Lim J, Jeon S, Shin HY, et al. Case of the index patient who caused tertiary
transmission of coronavirus disease 2019 in Korea: The application of
ACCEPTED MANUSCRIPT
lopinavir/ritonavir for the treatment of COVID-19 pneumonia monitored by
quantitative RT-PCR. J Korean Med Sci. 2020;35.
[58] Cao B, Wang Y, Wen D, et al. A Trial of Lopinavir-Ritonavir in Adults Hospitalized
with Severe Covid-19. N Engl J Med. 2020;
[59] Hurst M, Drugs DF-, 2000 undefined. Lopinavir. Springer.
[60] Cinatl J, Morgenstern B, Bauer G, et al. Glycyrrhizin, an active component of liquorice
roots, and replication of SARS-associated coronavirus. Lancet. 2003;361:2045–2046.
[61] Crance JM, Scaramozzino N, Jouan A, et al. Interferon, ribavirin, 6-azauridine and
glycyrrhizin: antiviral compounds active against pathogenic flaviviruses. Antiviral
Res. 2003;58:73–79.
[62] Arase Y, Ikeda K, Murashima N, et al. The long term efficacy of glycyrrhizin in
chronic hepatitis C patients. Cancer. 1997;79:1494–1500.
[63] Lampis G, Deidda D, Pinza M, et al. Enhancement of anti-herpetic activity of
glycyrrhizic acid by physiological proteins. Antivir Chem Chemother. 2001;12:125–
131.
[64] Sasaki H, Takei M, Kobayashi M, et al. Effect of glycyrrhizin, an active component of
licorice roots, on HIV replication in cultures of peripheral blood mononuclear cells
from HIV-seropositive patients. Pathobiology. 2002;70:229–236.
[65] Wu C-Y, Jan J-T, Ma S-H, et al. Small molecules targeting severe acute respiratory
syndrome human coronavirus. Proc Natl Acad Sci. 2004;101:10012–10017.
[66] Fujii T, Nakamura T, Iwamoto A. Current concepts in SARS treatment. J Infect
Chemother. 2004;10:1–7.
[67] Penninkilampi R, Eslick EM, Eslick GD. The association between consistent licorice
ingestion, hypertension and hypokalaemia: A systematic review and meta-analysis. J
ACCEPTED MANUSCRIPT
Hum Hypertens. 2017;31:699–707.
[68] CHEN J, LIU D, LIU L, et al. A pilot study of hydroxychloroquine in treatment of
patients with common coronavirus disease-19 (COVID-19). J Zhejiang Univ (Med
Sci). 2020;49:1–10.
[69] Gao J, Tian Z, Yang X. Breakthrough: Chloroquine phosphate has shown apparent
efficacy in treatment of COVID-19 associated pneumonia in clinical studies. Biosci
Trends. 2020;14:72–73.
[70] multicenter collaboration group of Department of Science and Technology of
Guangdong Province and Health Commission of Guangdong Province for chloroquine
in the treatment of novel coronavirus pneumonia. [Expert consensus on chloroquine
phosphate for the treatment of novel coronavirus pneumonia]. Zhonghua Jie He He Hu
Xi Za Zhi. 2020;43:185–188.
[71] Yao X, Ye F, Zhang M, et al. In Vitro Antiviral Activity and Projection of Optimized
Dosing Design of Hydroxychloroquine for the Treatment of Severe Acute Respiratory
Syndrome Coronavirus 2 (SARS-CoV-2). Clin Infect Dis. 2020;
[72] Gautret P, Lagier J-C, Parola P, et al. Hydroxychloroquine and azithromycin as a
treatment of COVID-19: results of an open-label non-randomized clinical trial. Int J
Antimicrob Agents. 2020;105949.
[73] Lenehan PJ, Schramm CM, Collins MS. An evaluation strategy for potential QTc
prolongation with chronic azithromycin therapy in cystic fibrosis. J Cyst Fibros.
2016;15:192–195.
[74] Chen CY, Wang FL, Lin CC. Chronic hydroxychloroquine use associated with QT
prolongation and refractory ventricular arrhythmia. Clin Toxicol. 2006;44:173–175.
[75] Guastalegname M, Vallone A. Could chloroquine/hydroxychloroquine be harmful in
ACCEPTED MANUSCRIPT
Coronavirus Disease 2019 (COVID-19) treatment? Clin Infect Dis. 2020;
[76] Juurlink DN. Safety considerations with chloroquine, hydroxychloroquine and
azithromycin in the management of SARS-CoV-2 infection. C Can Med Assoc J.
2020;192:E450–E453.
[77] Gautret P, Lagier JC, Parola P, et al. Clinical and microbiological effect of a
combination of hydroxychloroquine and azithromycin in 80 COVID-19 patients with
at least a six-day follow up: A pilot observational study. Travel Med Infect Dis.
2020;34:101663.
[78] Million M, Lagier JC, Gautret P, et al. Early treatment of COVID-19 patients with
hydroxychloroquine and azithromycin: A retrospective analysis of 1061 cases in
Marseille, France. Travel Med Infect Dis. 2020;101738.
[79] Mahévas M, Tran V-T, Roumier M, et al. Clinical efficacy of hydroxychloroquine in
patients with covid-19 pneumonia who require oxygen: observational comparative
study using routine care data. BMJ. 2020;369:m1844.
[80] Tang W, Cao Z, Han M, et al. Hydroxychloroquine in patients with mainly mild to
moderate coronavirus disease 2019: open label, randomised controlled trial. BMJ.
2020;369:m1849.
[81] Mehra MR, Desai SS, Ruschitzka F, et al. Hydroxychloroquine or chloroquine with or
without a macrolide for treatment of COVID-19: a multinational registry analysis.
Lancet (London, England). 2020;0.
[82] Gabriela Silva Borba M, Fonseca Almeida Val F, Sousa Sampaio V, et al. Chloroquine
diphosphate in two different dosages as adjunctive therapy of hospitalized patients
with severe respiratory syndrome in the context of coronavirus (SARS-CoV-2)
infection: Preliminary safety results of a randomized, double-blinded, phase IIb cl.
ACCEPTED MANUSCRIPT
[83] Rossignol J, … MS-ORVIA in C, 2014 undefined. Activity of thiazolides against
other respiratory viruses than influenza.
[84] Rossignol JF, La Frazia S, Chiappa L, et al. Thiazolides, a new class of anti-influenza
molecules targeting viral hemagglutinin at the post-translational level. J Biol Chem.
2009;284:29798–29808.
[85] Keeffe EB, Rossignol JF. Treatment of chronic viral hepatitis with nitazoxanide and
second generation thiazolides. World J Gastroenterol. 2009;15:1805–1808.
[86] Rossignol JF. Nitazoxanide, a new drug candidate for the treatment of Middle East
respiratory syndrome coronavirus. J Infect Public Health. 2016;9:227–230.
[87] Dang W, Xu L, Ma B, et al. Nitazoxanide inhibits human norovirus replication and
synergizes with ribavirin by activation of cellular antiviral response. Antimicrob
Agents Chemother. 2018;62.
[88] Hong S, Kim H, Song C, et al. Nitazoxanide suppresses IL-6 production in LPS-
stimulated mouse macrophages and TG-injected mice. Elsevier.
[89] Speich B, Ame SM, Ali SM, et al. Efficacy and safety of nitazoxanide, albendazole,
and nitazoxanide-albendazole against trichuris trichiura infection: A randomized
controlled trial. PLoS Negl Trop Dis. 2012;6.
[90] Stockis A, Allemon AM, De Bruyn S, et al. Nitazoxanide pharmacokinetics and
tolerability in man using single ascending oral doses. Int J Clin Pharmacol Ther.
2002;40:213–220.
[91] Search of: nitazoxanide | covid-19 - List Results - ClinicalTrials.gov.
[92] Liu Q, Xiong HR, Lu L, et al. Antiviral and anti-inflammatory activity of arbidol
hydrochloride in influenza A (H1N1) virus infection. Acta Pharmacol Sin.
2013;34:1075–1083.
ACCEPTED MANUSCRIPT
[93] Boriskin Y, Leneva I, Pecheur E-I, et al. Arbidol: A Broad-Spectrum Antiviral
Compound that Blocks Viral Fusion. Curr Med Chem. 2008;15:997–1005.
[94] Pécheur E-I, Borisevich V, Halfmann P, et al. The Synthetic Antiviral Drug Arbidol
Inhibits Globally Prevalent Pathogenic Viruses. J Virol. 2016;90:3086–3092.
[95] Boriskin YS, Pécheur EI, Polyak SJ. Arbidol: A broad-spectrum antiviral that inhibits
acute and chronic HCV infection. Virol J. 2006;3:56.
[96] Arastoo M, Khorram Khorshid HR, Radmanesh R, et al. Combination of IMODTM and
Arbidol to increase their immunomodulatory effects as a novel medicine to prevent
and cure influenza and some other infectious diseases. J Med Hypotheses Ideas.
2014;8:53–56.
[97] Deng L, Li C, Zeng Q, et al. Arbidol combined with LPV/r versus LPV/r alone against
Corona Virus Disease 2019:a retrospective cohort study. J Infect. 2020;
[98] Meng-zhao W, Long-yun C, … LN-A-A, et al. Efficacy and safety of arbidol in
treatment of naturally acquired influenza. arbidol.net [Internet]. [cited 2020 Jun 4];
Available from: http://arbidol.net/chinese003.pdf.
[99] Canga AG, Prieto AMS, Diez Liébana MJ, et al. The pharmacokinetics and
interactions of ivermectin in humans - A mini-review. AAPS J. 2008. p. 42–46.
[100] Tay M, Fraser J, Chan W, et al. Nuclear localization of dengue virus (DENV) 1–4 non-
structural protein 5; protection against all 4 DENV serotypes by the inhibitor
Ivermectin. Elsevier.
[101] Wagstaff KM, Sivakumaran H, Heaton SM, et al. Ivermectin is a specific inhibitor of
importin α/β-mediated nuclear import able to inhibit replication of HIV-1 and dengue
virus. Biochem J. 2012;443:851–856.
[102] Yang S, Atkinson S, Wang C, et al. The broad spectrum antiviral ivermectin targets the
ACCEPTED MANUSCRIPT
host nuclear transport importin α/β1 heterodimer. Elsevier [Internet]. [cited 2020 Jun
4]; Available from:
https://www.sciencedirect.com/science/article/pii/S0166354219307211.
[103] Götz V, Magar L, Dornfeld D, et al. Influenza A viruses escape from MxA restriction
at the expense of efficient nuclear vRNP import. Sci Rep. 2016;6:1–15.
[104] Caly L, Druce JD, Catton MG, et al. The FDA-approved Drug Ivermectin inhibits the
replication of SARS-CoV-2 in vitro. Antiviral Res. 2020;104787.
[105] Nicolas P, Maia M, Bassat Q, et al. Safety of oral ivermectin during pregnancy: a
systematic review and meta-analysis. Elsevier [Internet]. [cited 2020 Jun 4]; Available
from: https://www.sciencedirect.com/science/article/pii/S2214109X1930453X.
[106] Chandler RE. Serious neurological adverse events after ivermectin-do they occur
beyond the indication of onchocerciasis? Am J Trop Med Hyg. 2018;98:382–388.
[107] Mitsutaka Kitano, Atsuko Yamamoto, Takeshi Noshi, et al. Synergistic Antiviral
Activity of S-033188/S-033447, a Novel Inhibitor of Influenza Virus Cap-Dependent
Endonuclease, in Combination with Neuraminidase Inhibitors In Vitro | Open Forum
Infectious Diseases | Oxford Academic. Open Forum Infect Dis. 2017;4:S371.
[108] Kawaguchi N, Koshimichi H, Ishibashi T, et al. Evaluation of Drug–Drug Interaction
Potential between Baloxavir Marboxil and Oseltamivir in Healthy Subjects. Clin Drug
Investig. 2018;38:1053–1060.
[109] Kuba K, Imai Y, Rao S, et al. A crucial role of angiotensin converting enzyme 2
(ACE2) in SARS coronavirus-induced lung injury. Nat Med. 2005;11:875–879.
[110] Zou Z, Yan Y, Shu Y, et al. Angiotensin-converting enzyme 2 protects from lethal
avian influenza A H5N1 infections. Nat Commun. 2014;5:1–7.
[111] Khan A, Benthin C, Zeno B, et al. A pilot clinical trial of recombinant human
ACCEPTED MANUSCRIPT
angiotensin-converting enzyme 2 in acute respiratory distress syndrome. Crit Care.
2017;21:234.
[112] Meili S, Jianmin Y, Yuping S, et al. Inhibitors of RAS Might Be a Good Choice for
the Therapy of COVID-19 Pneumonia. Chinese J Tuberc Respir Dis. 2020;43:E014–
E014.
[113] Soo YOY, Cheng Y, Wong R, et al. Retrospective comparison of convalescent plasma
with continuing high-dose methylprednisolone treatment in SARS patients. Clin
Microbiol Infect. 2004;10:676–678.
[114] Cheng Y, Wong R, Soo YOY, et al. Use of convalescent plasma therapy in SARS
patients in Hong Kong. Eur J Clin Microbiol Infect Dis. 2005;24:44–46.
[115] World Health Organization. Use of Convalescent Whole Blood or Plasma Collected
from Patients Recovered from Ebola Virus Disease for Transfusion, as an Empirical
Treatment during Outbreaks Interim Guidance for National Health Authorities and
Blood Transfus. 2014.
[116] Shen C, Wang Z, Zhao F, et al. Treatment of 5 Critically Ill Patients with COVID-19
with Convalescent Plasma. JAMA - J Am Med Assoc. 2020;323:1582–1589.
[117] Li L, Zhang W, Hu Y, et al. Effect of Convalescent Plasma Therapy on Time to
Clinical Improvement in Patients With Severe and Life-threatening COVID-19: A
Randomized Clinical Trial. JAMA [Internet]. 2020 [cited 2020 Jun 4]; Available from:
http://www.ncbi.nlm.nih.gov/pubmed/32492084.
[118] Casadevall A, Joyner MJ, Pirofski L-A. A Randomized Trial of Convalescent Plasma
for COVID-19-Potentially Hopeful Signals. JAMA [Internet]. 2020 [cited 2020 Jun 4];
Available from: http://www.ncbi.nlm.nih.gov/pubmed/32492105.
[119] Van Griensven J, Edwards T, De Lamballerie X, et al. Evaluation of convalescent
ACCEPTED MANUSCRIPT
plasma for Ebola virus disease in Guinea. N Engl J Med. 2016;374:33–42.
[120] ter Meulen J, van den Brink EN, Poon LLM, et al. Human Monoclonal Antibody
Combination against SARS Coronavirus: Synergy and Coverage of Escape Mutants.
Burton DR, editor. PLoS Med. 2006;3:e237.
[121] Ng O-W, Keng C-T, Leung CS-W, et al. Substitution at Aspartic Acid 1128 in the
SARS Coronavirus Spike Glycoprotein Mediates Escape from a S2 Domain-Targeting
Neutralizing Monoclonal Antibody. Pöhlmann S, editor. PLoS One. 2014;9:e102415.
[122] Jiang L, Wang N, Zuo T, et al. Potent neutralization of MERS-CoV by human
neutralizing monoclonal antibodies to the viral spike glycoprotein. Sci Transl Med.
2014;6:234ra59.
[123] Sui J, Li W, Murakami A, et al. Potent neutralization of severe acute respiratory
syndrome (SARS) coronavirus by a human mAb to S1 protein that blocks receptor
association. Proc Natl Acad Sci U S A. 2004;101:2536–2541.
[124] Coughlin MM, Prabhakar BS. Neutralizing human monoclonal antibodies to severe
acute respiratory syndrome coronavirus: Target, mechanism of action, and therapeutic
potential. Rev Med Virol. 2012;22:2–17.
[125] Jia HP, Look DC, Shi L, et al. ACE2 Receptor Expression and Severe Acute
Respiratory Syndrome Coronavirus Infection Depend on Differentiation of Human
Airway Epithelia. J Virol. 2005;79:14614–14621.
[126] Zhou P, Yang X-L, Wang X-G, et al. A pneumonia outbreak associated with a new
coronavirus of probable bat origin. Nature. 2020;579:270–273.
[127] Tanaka T, Narazaki M, Kishimoto T. Immunotherapeutic implications of IL-6
blockade for cytokine storm. Immunotherapy. Future Medicine Ltd.; 2016. p. 959–970.
[128] Tocilizumab in Rheumatoid Arthritis: A Meta-Analysis of Efficacy and Selected
ACCEPTED MANUSCRIPT
Clinical Conundrums - PubMed.
[129] Yokota S, Miyamae T, Imagawa T, et al. Therapeutic efficacy of humanized
recombinant anti-interleukin-6 receptor antibody in children with systemic-onset
juvenile idiopathic arthritis. Arthritis Rheum. 2005;52:818–825.
[130] Drobyski WR, Pasquini M, Kovatovic K, et al. Tocilizumab for the treatment of
steroid refractory graft-versus-host disease. Biol Blood Marrow Transplant.
2011;17:1862–1868.
[131] Xu X, Han M, Li T, et al. Effective treatment of severe COVID-19 patients with
tocilizumab. Proc Natl Acad Sci. 2020;117:202005615.
[132] Toniati P, Piva S, Cattalini M, et al. Tocilizumab for the treatment of severe COVID-
19 pneumonia with hyperinflammatory syndrome and acute respiratory failure: A
single center study of 100 patients in Brescia, Italy. Autoimmun. Rev. NLM
(Medline); 2020. p. 102568.
[133] Luo P, Liu Y, Qiu L, et al. Tocilizumab treatment in COVID-19: A single center
experience. J Med Virol. 2020;
[134] Search of: Tocilizumab | COVID-19 - List Results - ClinicalTrials.gov.
[135] Atluri S, Manchikanti L, Hirsch JA. Expanded Umbilical Cord Mesenchymal Stem
Cells (UC-MSCs) as a Therapeutic Strategy in Managing Critically Ill COVID-19
Patients: The Case for Compassionate Use. Pain Physician. 2020;23:E71–E83.
[136] Hashmi S, Ahmed M, Murad MH, et al. Survival after mesenchymal stromal cell
therapy in steroid-refractory acute graft-versus-host disease: systematic review and
meta-analysis. Lancet Haematol. 2016;3:e45–e52.
[137] Wang D, Zhang H, Liang J, et al. Allogeneic mesenchymal stem cell transplantation in
severe and refractory systemic lupus erythematosus: 4 years of experience. Cell
ACCEPTED MANUSCRIPT
Transplant. 2013;22:2267–2277.
[138] Wilson JG, Liu KD, Zhuo H, et al. Mesenchymal stem (stromal) cells for treatment of
ARDS: A phase 1 clinical trial. Lancet Respir Med. 2015;3:24–32.
[139] Zheng G, Huang L, Tong H, et al. Treatment of acute respiratory distress syndrome
with allogeneic adipose-derived mesenchymal stem cells: A randomized, placebo-
controlled pilot study. Respir Res. 2014;15.
[140] Clinical remission of a critically ill COVID-19 patient treated by human umbilical cord
mesenchymal stem cells.
[141] Leng Z, Zhu R, Hou W, et al. Transplantation of ACE2- Mesenchymal Stem Cells
Improves the Outcome of Patients with COVID-19 Pneumonia. Aging Dis.
2020;11:216.
[142] Richardson P, Griffin I, Tucker C, et al. Baricitinib as potential treatment for 2019-
nCoV acute respiratory disease. Lancet. Lancet Publishing Group; 2020. p. e30–e31.
[143] Richardson PJ, Corbellino M, Stebbing J. Baricitinib for COVID-19: a suitable
treatment? – Authors’ reply. Lancet Infect Dis. 2020;
[144] Rademaker M, Baker C, Foley P, et al. Advice regarding COVID-19 and use of
immunomodulators, in patients with severe dermatological diseases. Australas J
Dermatol. 2020;
[145] Lee YH, Song GG. Relative efficacy and safety of tofacitinib, baricitinib, upadacitinib,
and filgotinib in comparison to adalimumab in patients with active rheumatoid
arthritis. Z Rheumatol. 2020;
[146] Haria M, Benfield P. Interferon-α-2a: A Review of its Pharmacological Properties and
Therapeutic Use in the Management of Viral Hepatitis. Drugs. 1995;50:873–896.
[147] Tarhini AA, Gogas H, Kirkwood JM. IFN-α in the Treatment of Melanoma. J
ACCEPTED MANUSCRIPT
Immunol. 2012;189:3789–3793.
[148] Kujawski LA, Talpaz M. The role of interferon-alpha in the treatment of chronic
myeloid leukemia. Cytokine Growth Factor Rev. 2007;18:459–471.
[149] Cinatl J, Morgenstern B, Bauer G, et al. Treatment of SARS with human interferons.
Lancet (London, England). 2003;362:293–294.
[150] Omrani AS, Saad MM, Baig K, et al. Ribavirin and interferon alfa-2a for severe
Middle East respiratory syndrome coronavirus infection: a retrospective cohort study.
www.thelancet.com/infection. 2014;14.
[151] A Prospective/Retrospective,Randomized Controlled Clinical Study of Interferon
Atomization in the 2019-nCoV Pneumonia - Full Text View - ClinicalTrials.gov.
[152] Efficacy and Safety of IFN-α2β in the Treatment of Novel Coronavirus Patients - Full
Text View - ClinicalTrials.gov.
[153] Experimental Trial of rhIFNα Nasal Drops to Prevent 2019-nCOV in Medical Staff -
Full Text View - ClinicalTrials.gov.
[154] Russell CD, Millar JE, Baillie JK. Clinical evidence does not support corticosteroid
treatment for 2019-nCoV lung injury. Lancet. Lancet Publishing Group; 2020. p. 473–
475.
[155] Stockman LJ, Bellamy R, Garner P. SARS: Systematic review of treatment effects.
PLoS Med. 2006. p. 1525–1531.
[156] Arabi YM, Mandourah Y, Al-Hameed F, et al. Corticosteroid therapy for critically ill
patients with middle east respiratory syndrome. Am J Respir Crit Care Med.
2018;197:757–767.
[157] Liu X, Zhang Y, Xu X, et al. Evaluation of plasma exchange and continuous veno-
venous hemofiltration for the treatment of severe avian influenza A (H7N9): A cohort
ACCEPTED MANUSCRIPT
study. Ther Apher Dial. 2015;19:178–184.
[158] Xu K, Cai H, Shen Y, et al. Management of corona virus disease-19 (COVID-19): the
Zhejiang experience. Zhejiang Da Xue Xue Bao Yi Xue Ban. 2020;49:0.
[159] Zhang Y, Yu L, Tang L, et al. A Promising Anti-Cytokine-Storm Targeted Therapy for
COVID-19: The Artificial-Liver Blood-Purification System. Engineering. 2020;
[160] Davies AR, Jones D, Bailey M, et al. Extracorporeal membrane oxygenation for 2009
influenza A(H1N1) acute respiratory distress syndrome. JAMA - J Am Med Assoc.
2009;302:1888–1895.
[161] Alshahrani MS, Sindi A, Alshamsi F, et al. Extracorporeal membrane oxygenation for
severe Middle East respiratory syndrome coronavirus. Ann Intensive Care. 2018;8:3.
[162] Malato A, Dentali F, Siragusa S, et al. The impact of deep vein thrombosis in critically
ill patients: A meta-analysis of major clinical outcomes. Blood Transfus. SIMTI
Servizi Sri; 2015. p. 559–568.
[163] Uthman IW, Gharavi AE. Viral infections and antiphospholipid antibodies. Semin
Arthritis Rheum. 2002;31:256–263.
[164] Paranjpe I, Fuster V, Lala A, et al. Association of Treatment Dose Anticoagulation
with In-Hospital Survival Among Hospitalized Patients with COVID-19. J Am Coll
Cardiol. 2020;
[165] Tang N, Bai H, Chen X, et al. Anticoagulant treatment is associated with decreased
mortality in severe coronavirus disease 2019 patients with coagulopathy. J Thromb
Haemost. 2020;18.
[166] Chuang YJ, Swanson R, Raja SM, et al. Heparin enhances the specificity of
antithrombin for thrombin and factor Xa independent of the reactive center loop
sequence. Evidence for an exosite determinant of factor Xa specificity in heparin-
ACCEPTED MANUSCRIPT
activated antithrombin. J Biol Chem. 2001;276:14961–14971.
[167] White D, MacDonald S, Bull T, et al. Heparin resistance in COVID-19 patients in the
intensive care unit. J Thromb Thrombolysis. 2020;
[168] Minze MG, Kwee YY, Hall RG. Low-molecular-weight heparin prophylaxis dosing: Is
weight an issue? J. Pharm. Technol. SAGE Publications Inc.; 2016. p. 75–80.
[169] Perini P, Nabulsi B, Massoni CB, et al. Acute limb ischaemia in two young, non-
atherosclerotic patients with COVID-19. Lancet. Lancet Publishing Group; 2020. p.
1546.
[170] Lim W, Dentali F, Eikelboom JW, et al. Meta-analysis: Low-molecular-weight heparin
and bleeding in patients with severe renal insufficiency. Ann. Intern. Med. American
College of Physicians; 2006. p. 673–684.
[171] Immunoglobulin G From Patients With Heparin-Induced Thrombocytopenia Binds to
a Complex of Heparin and Platelet Factor 4 - PubMed.
[172] Warkentin TE. Fondaparinux: Does it cause HIT? can it treat HIT? Expert Rev.
Hematol. Expert Rev Hematol; 2010. p. 567–581.
[173] Dayrit FM, Newport MT. The Potential of Coconut Oil and its Derivatives as Effective
and Safe Antiviral Agents Against the Novel Coronavirus (nCoV-2019). 2020.
[174] Dharma Widhiarta K. Virgin Coconut Oil for HIV-Positive People. 2016.
[175] Conrado Dayrit BS. COCONUT OIL IN HEALTH AND DISEASE: ITS AND
MONOLAURIN’S POTENTIAL AS CURE FOR HIV/AIDS*. 2000.
[176] Saha RK, Takahashi T, Suzuki T. Glucosyl hesperidin prevents influenza A virus
replication in vitro by inhibition of viral sialidase. Biol Pharm Bull. 2009;32:1188–
1192.
ACCEPTED MANUSCRIPT
[177] Nardarajah D. Hesperidin-A short Review. Res J Pharm Tech. 2014;7.
[178] Escriou N, Callendret B, Lorin V, et al. Protection from SARS coronavirus conferred
by live measles vaccine expressing the spike glycoprotein. Virology. 2014;452–
453:32–41.
[179] Du L, Zhao G, Chan CCS, et al. Recombinant receptor-binding domain of SARS-CoV
spike protein expressed in mammalian, insect and E. coli cells elicits potent
neutralizing antibody and protective immunity. Virology. 2009;393:144–150.
[180] Bennett J V., de Castro JF, Valdespino-Gomez JL, et al. Aerosolized measles and
measles-rubella vaccines induce better measles antibody booster responses than
injected vaccines: randomized trials in Mexican schoolchildren. Bull World Health
Organ. 2002;
[181] Liniger M, Zuniga A, Tamin A, et al. Induction of neutralising antibodies and cellular
immune responses against SARS coronavirus by recombinant measles viruses.
Vaccine. 2008;26:2164–2174.
[182] Pregnancy Guidelines and Recommendations by Vaccine | CDC.
[183] Demicheli V, Rivetti A, Debalini MG, et al. Vaccines for measles, mumps and rubella
in children. Cochrane Database Syst Rev. 2012;
[184] de Roux A, Schmidt N, Rose M, et al. Immunogenity of the pneumococcal
polysaccharide vaccine in COPD patients. The effect of systemic steroids. Respir Med.
2004;98:1187–1194.
[185] Murdaca G, Orsi A, Spanò F, et al. Influenza and pneumococcal vaccinations of
patients with systemic lupus erythematosus: Current views upon safety and
immunogenicity. Autoimmun. Rev. 2014. p. 75–84.
[186] Chang CC, Chang YS, Chen WS, et al. Effects of annual influenza vaccination on
ACCEPTED MANUSCRIPT
morbidity and mortality in patients with Systemic Lupus Erythematosus: A
Nationwide Cohort Study. Sci Rep. 2016;6:1–7.
[187] Ponvert C, Ardelean-Jaby D, Colin-Gorski AM, et al. Anaphylaxis to the 23-valent
pneumococcal vaccine in child: A case-control study based on immediate responses in
skin tests and specific IgE determination. Vaccine. 2001;19:4588–4591.
[188] Elkayam O, Bashkin A, Mandelboim M, et al. The effect of infliximab and timing of
vaccination on the humoral response to influenza vaccination in patients with
rheumatoid arthritis and ankylosing spondylitis. Semin Arthritis Rheum. 2010;39:442–
447.
[189] Gelinck LBS, Van Der Bijl AE, Beyer WEP, et al. The effect of anti-tumour necrosis
factor α treatment on the antibody response to influenza vaccination. Ann Rheum Dis.
2008;67:713–716.
[190] Bingham CO, Looney RJ, Deodhar A, et al. Immunization responses in rheumatoid
arthritis patients treated with rituximab: Results from a controlled clinical trial.
Arthritis Rheum. 2010;62:64–74.
[191] Van Assen S, Holvast A, Benne CA, et al. Humoral responses after influenza
vaccination are severely reduced in patients with rheumatoid arthritis treated with
rituximab. Arthritis Rheum. 2010;62:75–81.
[192] Salemi S, Picchianti-Diamanti A, Germano V, et al. Influenza vaccine administration
in rheumatoid arthritis patients under treatment with TNFα blockers: Safety and
immunogenicity. Clin Immunol. 2010;134:113–120.
[193] Ferner R, Aronson J. Chloroquine and hydroxychloroquine in covid-19. 2020 [cited
2020 Jun 4]; Available from: https://www.bmj.com/content/369/bmj.m1432/.
[194] Jean S-S, Hsueh P-R. Old and re-purposed drugs for the treatment of COVID-19.
ACCEPTED MANUSCRIPT
Expert Rev Anti Infect Ther [Internet]. 2020 [cited 2020 Jun 4]; Available from:
http://www.ncbi.nlm.nih.gov/pubmed/32419524.
[195] Jean S, Lee P, Microbiology PH-J of, et al. Treatment options for COVID-19: The
reality and challenges. Elsevier [Internet]. [cited 2020 Jun 4]; Available from:
https://www.sciencedirect.com/science/article/pii/S1684118220300943.
[196] Luke TC, Kilbane EM, Jackson JL, et al. Meta-analysis: Convalescent blood products
for Spanish influenza pneumonia: A future H5N1 treatment? Ann. Intern. Med.
American College of Physicians; 2006. p. 599–609.
[197] Hung I, To K, Lee C, et al. Convalescent plasma treatment reduced mortality in
patients with severe pandemic influenza A (H1N1) 2009 virus infection.
academic.oup.com [Internet]. [cited 2020 Jun 4]; Available from:
https://academic.oup.com/cid/article-abstract/52/4/447/378553.
ACCEPTED MANUSCRIPT
Figure Legends
Figure 1. SARS-CoV-2 replication cycle with target therapeutic agents. Abbreviations;
CM=camostat mesilate, NM=nafamostat mesilate, IL-6=interleukin-6, ACE=angiotensin-
converting enzyme.
ACCEPTED MANUSCRIPT
Table 1. Summary of possible therapeutic agents for SARS-CoV-2. 1
Dru
g
Mechanism Common Uses Common Side Effects/Limitations
Nucleoside Analo
g
ues (Pol
y
merase Inhibitors)
Ribavirin A guanosine analogue incorporates into viral RNA, instead of
guanosine, and is toxic to the viral genome
HCV, RCV, and viral
hemorrhagic fevers
Hemolytic anemia, fatigue, rash,
leukopenia, and teratogenicity
Favipiravir A guanine analogue, inhibiting RNA polymerase and inducing
toxic mutations
influenza A H1N1, yellow
fever, and Ebola
Gastrointestinal tract disturbance, the
elevation of liver function enzymes,
and psychiatric symptoms
Remdesivir Analogous to adenosine inhibits RNA polymerase and viral
replication
Ebola Blood biomarkers of organ
impairment, gastrointestinal problems,
elevated liver enzymes, and infusion
site reactions
Penciclovir Inhibition of RNA polymerase Herpesvirus Headache and nausea
Protease Inhibitors
Camostat mesilate Serine protease enzyme inhibitor Oral squamous cell
carcinoma and dystrophic
epidermolysis
Dyspepsia
Nafamostat mesilate Serine protease enzyme inhibitor Acute pancreatitis, DIC Acute systemic reaction
Darunavir A protease enzyme inhibitor, and target for spike protein of
SARS-COV-2
HIV Rash, nausea, and diarrhea,
hepatotoxic
Cobicistat CYP3A4 inhibitor HIV Jaundice, skin rash, depression, and
headache
Lopinavir Protease enzyme inhibitor SARS-, and MERS-CoV,
and HIV
Nausea, diarrhea, and asthenia
Ritonavir CYP3A4 inhibitor SARS-, and MERS-CoV,
and HIV
Nausea, diarrhea, and asthenia
Other Antimicrobial A
ents
Glycyrrhizin Inhibits viral adsorption and penetration, induces the nitrous oxide
pathway and affects cellular signaling pathways such as protein
kinase C, transcription factors, as well as casein kinase II
HCV, upper respiratory
tract infections, and as
anti-inflammatory,
immunosuppressive, anti-
tumor, and antiviral
Hypokalemia, hypertension, and
irregular heart rhythm
ACCEPTED MANUSCRIPT
properties against
flaviviruses, herpes
viruses, and human
immunodeficiency virus
Chloroquine, and
Hydroxychloroquine
Increasing pH in host-cell lysosomes which in turn inhibits the
hydrolytic activity of protease enzymes that are required for
processing of viral glycoproteins. They also may interfere with the
terminal glycosylation of the ACE-II receptor. They could
negatively influence the virus-receptor binding and abrogate the
infection.
Malaria QT prolongation
Nitazoxanide Downregulation of expression of N protein Rotavirus and giardiasis Vomiting, fever, skin rash, and
abdominal pain
Arbidol (Umifenovir) Inhibits fusion between viral and cellular membrane, activation of
macrophages and induction of interferons
HBV, HCV, Ebola virus,
poliovirus, as well as
human herpes virus-8
Gastrointestinal disturbance
Ivermectin Inhibition of viral nuclear import and therefore inhibit its
replication
Various parasitic
infections
Neurotoxicity
Baloxavir marboxil/Oseltamivir Viral endonuclease inhibitor and neuraminidase inhibitor,
respectively
Influenza A and B viruses -
Tar
g
ets for ACE
ACE-II inhibitors Downregulating ACE-II Hypertension Increases pulmonary vascular
permeability and pulmonary edema.
Recombinant Agents Dilatation of SARS-CoV-2 levels via reduction of ACE-II
expression
SARS-CoV Dysphagia, rash, and acute renal
failure
Immunotherap
y
and Immunomodulators
Convalescent plasma Binds to SARS-CoV-2, blocks infection, binds to infected cells
and changes the immune system
Ebola, SARS-CoV Nausea, fever, and skin rash, risk of
diseases transmission
Monoclonal antibodies Targeting a specific protein, especially the RBD of S1 subunit,
thereby, blocking its binding to host receptor (viral entry) and
targeting S2 subunit (fusion between the virus and host cell
membrane)
SARS- and MERS-CoVs Not cost-effective and laborious
Tocilizumab Anti-IL-6 antibody Rheumatoid arthritis and
systemic juvenile
Increase the risk of opportunistic
infections, lymphopenia and
ACCEPTED MANUSCRIPT
idiopathic arthritis leukopenia
MSCs Immunomodulatory (secrete anti-inflammatory paracrine factors),
counteract the cytokine storm, protect and/or regenerate the
damaged pulmonary epithelial cells
Acute graft-versus-host
disease, and SLE
-
Baricitinib Inhibition of JAK 1 and 2 enzymes (cytokines’ gene expression),
AP2-associated protein kinase 1 (viral entry) and the intracellular
assembly of virus particles
SLE Immunosuppression, upper respiratory
tract infections, nausea, cold sores
from herpes simplex, and shingles
from herpes zoster
INF-α and β Stimulate innate antiviral immunity SARS-, and MERS-CoVs,
hepatitis, leukemia, and
melanoma
Fatigue, anorexia, and weight loss
Corticosteroids Cytokine storm-targeted therapy and immunosuppression SLE Immunosuppression
Or
g
an Function Support
ALPS Cytokine storm-targeted therapy and organ function support --
ECMO Cytokine storm-targeted therapy and organ function support -Very limited, high expertise level,
and a source of SARS-CoV-2
transmission
Other Therapeutic A
g
ents
Anticoagulants - DVT, PE, stroke and heart
attacks
Bleeding
Coconuts Disrupting viral binding to host cells and damaging the viral
envelope
HIV -
Hesperidin Inhibition of viral neuraminidase (sialidase) enzyme and the
interaction between the ACE-II receptor and the spike protein
(inhibition of viral entry)
Influenza A virus Nausea and vomiting
Measles vaccine
Expresses the RBD of S1 subunit of the spike protein of SARS
and induces a highly robust humoral response enabling cross-
reactivity and protection against SARS
Measles Contraindicated in pregnancy,
thrombocytopenia and febrile seizures
Abbreviations; ALPS=artificial liver blood purification system, ECMO=extracorporeal membrane oxygenation, HCV= Hepatitis C virus, 2
ACE=angiotensin-converting enzyme, MSCs= mesenchymal stem cells, INF=interferon, HIV=human immunodeficiency virus, SARS=severe acute 3
respiratory syndrome, MERS= middle east respiratory syndrome, RCV=respiratory syncytial virus, SLE=systemic lupus erythematosus, 4
RBD=receptor binding domain. 5
6
ACCEPTED MANUSCRIPT
8
9
F
i
ig
1
ACCEPTED MANUSCRIPT
... Some researchers have theorized that the measlesmumps-rubella (MMR) vaccine may play a decisive role in these disparities [1,[5][6][7][8][9][10]. There are reports that appear to suggest that several currently available vaccines (including polio, Haemophilus influenzae type-B, MMR, and pneumococcal) may offer significant protection against COVID-19 via a nonspecific immunity [8,11,12]. ...
... Some studies have reported that the MMR vaccine may protect against or reduce the severity, hospitalization, or mortality of coronavirus disease 2019 infection [1,[5][6][7][8][9][10]. This theory was introduced by Gold et al. (2020), after observing that recent countries with large-scale MMR vaccination are associated with the fewest COVID-19 deaths [14]. ...
Article
Full-text available
Objective & aim The coronavirus disease, so far (COVID-19) has brought about millions of infections and fatalities throughout the world. Our aim was to determine the correlation between rubella IGG titers with the severity COVID-19. Materials & methods This study was conducted among COVID-19 confirmed patients over 18 years of age. The disease severity levels were categorized by WHO interim guidance. The rubella-specific IgG antibody-titer spectrum was measured (within first 48 h of hospitalization) by enzyme-linked immunosorbent assay (ELISA). Result In a study of 46 inpatients with varying COVID-19 disease severity (mild, moderate, severe, and critical), we observed a negative correlation between rubella IgG antibody titers and COVID‐19 severity (P-Value = 0.017), There was an interaction between COVID-19 vaccination history and rubella IGG on severity COVID-19 (P-Value = 0.0015). There was an interaction between age group under 44 years (including national measles- rubella (MR) vaccination in Iran) and rubella IGG titers on severity COVID-19 too (p-value = 0.014). Conclusion In conclusion, MR vaccination seems to have a positive effect in reducing the severity of the disease, emphasizing that, the important and separate effect of the IGG rubella (due to natural or extrinsic immunity) titers is determining.
... Although SARS-CoV-2 pandemic crisis has a little dissipation in some countries, it has been discovered that the countries with lower death rates and lower severity of symptoms from COVID-19 infection are those with large-scale MMR vaccination campaigns (Gold et al. 2020). Some other researchers have been more specific to propose thatthe MMR vaccine, could induce self-protection against COVID-19 infection or even decrease its severity, prevalence and mortality rate (Anbarasu et al. 2020;Elhusseiny et al. 2020;Meenakshisundaram et al. 2020;Sidiq et al. 2020). ...
Article
Full-text available
Many factors have been implicated in the pathogenesis and severity of COVID-19 pandemic. A wide variation in the susceptibility for SARS-CoV-2 infection among different population, gender and age has been observed. Multiple studies investigated the relationship between the antibody's titre of previously vaccinated individuals and the susceptibility of coronavirus infection, to find a rapid effective therapy for this pandemic. This study focused on the association between measles-mumps-rubella (MMR) antibodies titre and the severity of COVID-19 infection. We aimed to investigate the correlation between the antibody's titre of MMR and the SARS-CoV-2 infection susceptibility and disease severity, in a cohort of COVID-19 Egyptian patients, compared to a control group. MMR antibody titre was measured using enzyme Linked Immune Sorbent Assay; (ELISA) for 136 COVID-19 patients and 44 healthy individuals, as control group. There were high levels of measles and mumps antibodies titer in the deteriorating cases, which could not protect from SARS-CoV-2 infection. However, the rubella antibodies might protect from SARS-CoV-2 infection, but once the infection occurs, it may aggravate the risk of case deterioration. MMR antibodies could be used as a guideline for COVID-19 symptom-severity and, in turn, may be considered as an economic prognostic marker used for early protection from multiple autoimmune organ failure.
... Expected risk Associated with Potential Therapeutic Approaches RNA-dependent RNA polymerase inhibitors (Nucleoside Analogues): Remdesivir (RDV) Remdesivir so far considered to be the most clinically successful drug against SARS-CoV-2. The broadspectrum activity of Remdesivir was evaluated against filoviruses, paramyxoviruses, pneumoviruses, Nipah viruses, Ebola viruses, SARS-CoVs and MERS [27][28][29]. Remdesivir is an adenosine analogue works by inhibiting the viral replication through targeting the RNAdependent RNA polymerase (RdRp) cause premature termination of viral RNA transcription [30,31]. The nsp12 residue is the potential binding site of remdesivir on RNA-dependent RNA polymerase (RdRp). ...
Article
Full-text available
he emerging and re-emergence of viral outbreaks in the history of mankind has always pose severe global intimidation to public health and economy. The debilitating effects of 2019-nCoV (2019 novel coronavirus) outbreak has swiftly spread worldwide due to its highly contagious nature with severe risk of respiratory tract infections and higher mortality rate, necessitating the urgent need for the production of effective vaccine and potential therapeutic agents. The active evolution of SARS-CoV-2 strain in different population and environment strive immense challenge against anti-viral therapeutic development based on viral pathogenicity. The potential FDA drugs are evaluated based on their known safety and efficacy with exceptional pharmacokinetic profiles for the treatment of nCoV-2019. Existing knowledge related to MERS-CoV and SARS-CoV epidemic has provided a better understanding to explore purposeful therapeutics strategies against novel coronavirus disease (COVID-19). To limited extend, the ongoing promising and hopeful treatments includes convalescent plasma therapy, remdesivir, lopinavir/ritonavir, ACE inhibitors, TMPRSS2 inhibitors, hydroxychloroquine, interferon, ribavirin, tocilizumab, and corticosteroids however clinical efficacy of some of them need to be validated in randomized clinical trials (RCTs). The global struggle to make a protected and successful Coronavirus immunization is finally proving to be fruitful. Although challenges such as strain variation resistant, possible side effects, adequate supply of vaccines to all countries and limited availability of second dose still diverting the option of possible efficacious therapeutics strategies to work alongside with vaccine development with improved efficacy and safety profile. This review is focused on the potential advancement in therapeutic approaches with possible repurposing of the available drugs and explores the current status of available vaccines with hope that these strategies found to be cogent in controlling SARS-CoV-2 outbreak.
... Moreover, Saunders et al. analysed 22 individual studies from 10 countries, revealing a relationship between sedentary behaviour and depression [14]. Furthermore, the imposition of restrictions on social meetings appears to have had an impact on the deterioration of the psychological condition of many populations [15][16][17][18]. ...
Article
Full-text available
Published reports indicate the need for psychological interventions and the integration of psychiatric care into crisis management plans in people with mental health issues caused by the COVID-19 pandemic. It seems crucial to identify the root causes of the health-social-economic crisis and identify potential opportunities for widely implemented psychological assistance. This narrative literature review aims to identify the types of interventions deployed as telemedicine-based mental health support and their effectiveness. The PubMed and Web of Science electronic databases were searched. From a total of 48 articles, 46 were analysed after removing duplicates. From these, thirty-seven records were excluded according to the inclusion criteria and nine (eight RCT and one cross-over) were assessed as full texts. The included publications were randomised clinical trials or cross-over studies focused on remote mental support interventions. In all studies, participants represented both sexes and had an average age range of 6–64. Studies included participants from seven countries and the overall number of participants in the included studies was 687. The content of these intervention programmes includes both established psychotherapeutic programmes, as well as new interventions. Remote support was implemented through three approaches: phone/video calls, mobile applications, and internet-based programs. The results of the included studies indicate a higher or equal efficacy of telemedicine interventions compared to traditional forms. The review also revealed a relatively wide range of targeted research groups: from children with social anxiety through to their caregivers; adolescents with neurological disorders; and from college students to adults with psychiatric or orthopaedic disorders. Analysis of the included papers found that telemedicine interventions show promising results as an attempt to improve population mental health during the COVID-19 pandemic.
... Importantly, high throughput drug screening program, as well as machine-based learning, proved to be a boon to the medicinal chemist in expediting some of the aforementioned drug discovery endeavours. RNA-dependent RNA polymerase (RdRp) and helicase as targets for COVID19 drugs have also been the subject of explorations [23,235,236]. It has been well established that the viral spike glycoprotein S protein binding to host cell angiotensin-converting enzyme 2 (ACE2) [24,25] is responsible for SARS-CoV-2 entry into host cells. ...
Article
Full-text available
Unprecedented efforts of the researchers have been witnessed in the recent past towards the development of vaccine platforms for the control of the COVID-19 pandemic. Albeit, vaccination stands as a practical strategy to prevent SARS-CoV-2 infection, supplementing the anti-COVID19 arsenal with therapeutic options such as small molecules/peptides and antibodies is being conceived as a prudent strategy to tackle the emerging SARS-CoV-2 variants. Noteworthy to mention that collective efforts from numerous teams have led to the generation of a voluminous library composed of chemically and mechanistically diverse small molecules as anti-COVID19 scaffolds. This review article presents an overview of medicinal chemistry campaigns and drug repurposing programs that culminated in the identification of a plethora of small molecule-based anti-COVID19 drugs mediating their antiviral effects through inhibition of proteases, S protein, RdRp, ACE2, TMPRSS2, cathepsin and other targets. In light of the evidence ascertaining the potential of small molecule drugs to approach conserved proteins required for the viral replication of all coronaviruses, accelerated FDA approvals are anticipated for small molecules for the treatment of COVID19 shortly. Though the recent attempts invested in this direction in pursuit of enrichment of the anti-COVID-19 armoury (chemical tools) are praiseworthy, some strategies need to be implemented to extract conclusive benefits of the recently reported small molecule viz. (i) detailed preclinical investigation of the generated anti-COVID19 scaffolds (ii) in-vitro profiling of the inhibitors against the emerging SARS-CoV-2 variants (iii) development of assays enabling rapid screening of the libraries of anti-COVID19 scaffold (iv) leveraging the applications of machine learning based predictive models to expedite the anti-COVID19 drug discovery campaign (v) design of antibody–drug conjugates.
Article
Innovation usually requires time‐consuming exploratory approaches. However, external shocks and related crises, such as the COVID‐19 pandemic, lead to severe time pressures, which require short‐term R&D results. We investigate how organizations' prior collaboration and existing knowledge not only helped them cope with the crisis but also affected the vaccine's development performance. Specifically, we investigate the R&D outcomes of 386 organizations involved in the COVID‐19 vaccine's development within the first 18 months after the pandemic's outbreak. The results reveal that under urgency, organizations with prior scientific collaborations and technological knowledge exhibit a higher R&D performance. Furthermore, a broad network of diverse collaborators strengthened this relationship, thereby calling for more interdisciplinary R&D activities. We therefore extend the literature on innovation speed and strengthen long‐term R&D outcomes' role in organizations with a broad existing knowledge base and collaboration networks. We do so by specifically supporting such organizations' ability to integrate their previous R&D's and collaborations' knowledge to achieve rapid innovative outcomes under urgency.
Article
Full-text available
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has quickly spread all over the world. In this respect, traditional medicinal chemistry, repurposing, and computational approaches have been exploited to develop novel medicines for treating this condition. The effectiveness of chemicals and testing methods in the identification of new promising therapies, and the extent of preparedness for future pandemics, have been further highly advantaged by recent breakthroughs in introducing noble small compounds for clinical testing purposes. Currently, numerous studies are developing small-molecule (SM) therapeutic products for inhibiting SARS-CoV-2 infection and replication, as well as managing the disease-related outcomes. Transmembrane serine protease (TMPRSS2)-inhibiting medicinal products can thus prevent the entry of the SARS-CoV-2 into the cells, and constrain its spreading along with the morbidity and mortality due to the coronavirus disease 2019 (COVID-19), particularly when co-administered with inhibitors such as chloroquine (CQ) and dihydroorotate dehydrogenase (DHODH). The present review demonstrates that the clinical-stage therapeutic agents, targeting additional viral proteins, might improve the effectiveness of COVID-19 treatment if applied as an adjuvant therapy side-by-side with RNA-dependent RNA polymerase (RdRp) inhibitors.
Chapter
The emergence of mutagenic strains of severe acute respiratory syndrome-Coronavirus-2 (SARS-CoV-2) worst hit the world which already suffered from the Coronavirus disease-2019 (COVID-19) pandemic for 2 years. Due to recent advances in vaccinomics, many vaccine candidates are available but their efficacy against a mutant version of SARS-CoV-2 has remained uncertain. The immune-informatics-based reverse vaccinomic approaches have shown promising investigations recently for the development of cost-effective vaccinomics candidates in a very short period of time. The strategic vaccine development of selected epitopes using artificial intelligence for both B- and T-cells is a very crucial step in this process. This approach provides a highly effective and immunogenic vaccine that offers immunological safety against autoimmunity and other adverse effects over ethnicities, pregnant women, and vulnerable age groups. Several researchers have developed effective vaccine candidates using computational vaccinology and the immune-informatics approach. In this process, a unique peptide sequence of viral proteins such as Nucleocapsid, spike, envelope protein was identified by various in silico tools which are acting as immunological epitopes against TLRs, T-cells, and B-cells. While the conventional immunological vaccine studies take years for vaccine candidature, the immunoinformatics approach is a time-efficient way for the next generation research to study host-pathogen interactions and vaccine development. It is also cost-effective and leads to a better understanding of disease pathogenesis, diagnosis, and immunological response. Owing to the advantage of immunoinformatics-based vaccine approaches the present chapter aimed to discuss vaccine development using immunoinformatics approaches. Besides, the current challenges and future aspects have also been discussed herewith.
Chapter
The novel coronavirus infection (COVID-19) first reported in December 2019, has become serious global life-threatening disease that has created huge health care crises. Despite huge number of clinical trials for drug and vaccine, only few successful candidates are in market, which led to problem of demand and supply. The low- and middle-income countries faces major health crisis due to financial issues. Studies to search plant originated natural bioactive molecules for prophylactic and therapeutic of COVID-19 infection has gained considerable attention, due to their low-cost, easy availability and lesser side effects. Natural products and herbal medicine have long been known for their role in treating respiratory infections and many of them have been approved/under trial as drugs or over-the-counter food additives to lessen the symptoms. It is undisputable fact that herbal/natural molecules’ medicine is still a promising resource and used as precursor for drug discovery to search prospective prophylactic candidate against COVID-19. These bioactive compounds bind with potential therapeutic target of SARS-COV-2 such as ACE II, Spike protein, TMPRESS, RdRp, Main proteases and endoribonuclease and may prevent or at least slow down the SARS-CoV-2 infection. Therefore, several numbers of clinical trials have registered to investigate the potentials of natural product to halt disease progression. The main aim of present chapter is to discuss the potential role of natural molecules which can be used as therapeutic drugs for treatment of COVID-19 and thus helpful to curb down the mortality rate.
Article
Full-text available
Background Being “positive” has been one of the most frustrating words anyone could hear since the end of 2019. This word had been overused globally due to the high infectious nature of SARS-CoV-2. All citizens are at risk of being infected with SARS-CoV-2, but a red warning sign has been directed towards cancer and immune-compromised patients in particular. These groups of patients are not only more prone to catch the virus but also more predisposed to its deadly consequences, something that urged the research community to seek other effective and safe solutions that could be used as a protective measurement for cancer and autoimmune patients during the pandemic. Aim The authors aimed to turn the spotlight on specific herbal remedies that showed potential anticancer activity, immuno-modulatory roles, and promising anti-SARS-CoV-2 actions. Methodology To attain the purpose of the review, the research was conducted at the States National Library of Medicine (PubMed). To search databases, the descriptors used were as follows: “COVID-19”/”SARS-CoV-2”, “Herbal Drugs”, “Autoimmune diseases”, “Rheumatoid Arthritis”, “Asthma”, “Multiple Sclerosis”, “Systemic Lupus Erythematosus” “Nutraceuticals”, “Matcha”, “EGCG”, “Quercetin”, “Cancer”, and key molecular pathways. Results This manuscript reviewed most of the herbal drugs that showed a triple action concerning anticancer, immunomodulation, and anti-SARS-CoV-2 activities. Special attention was directed towards “matcha” as a novel potential protective and therapeutic agent for cancer and immunocompromised patients during the SARS-CoV-2 pandemic. Conclusion This review sheds light on the pivotal role of “matcha” as a tri-acting herbal tea having a potent antitumorigenic effect, immunomodulatory role, and proven anti-SARS-CoV-2 activity, thus providing a powerful shield for high-risk patients such as cancer and autoimmune patients during the pandemic.
Article
Full-text available
Importance Convalescent plasma is a potential therapeutic option for patients with coronavirus disease 2019 (COVID-19), but further data from randomized clinical trials are needed. Objective To evaluate the efficacy and adverse effects of convalescent plasma therapy for patients with COVID-19. Design, Setting, and Participants Open-label, multicenter, randomized clinical trial performed in 7 medical centers in Wuhan, China, from February 14, 2020, to April 1, 2020, with final follow-up April 28, 2020. The trial included 103 participants with laboratory-confirmed COVID-19 that was severe (respiratory distress and/or hypoxemia) or life-threatening (shock, organ failure, or requiring mechanical ventilation). The trial was terminated early after 103 of a planned 200 patients were enrolled. Intervention Convalescent plasma in addition to standard treatment (n = 52) vs standard treatment alone (control) (n = 51), stratified by disease severity. Main Outcomes and Measures Primary outcome was time to clinical improvement within 28 days, defined as patient discharged alive or reduction of 2 points on a 6-point disease severity scale (ranging from 1 [discharge] to 6 [death]). Secondary outcomes included 28-day mortality, time to discharge, and the rate of viral polymerase chain reaction (PCR) results turned from positive at baseline to negative at up to 72 hours. Results Of 103 patients who were randomized (median age, 70 years; 60 [58.3%] male), 101 (98.1%) completed the trial. Clinical improvement occurred within 28 days in 51.9% (27/52) of the convalescent plasma group vs 43.1% (22/51) in the control group (difference, 8.8% [95% CI, −10.4% to 28.0%]; hazard ratio [HR], 1.40 [95% CI, 0.79-2.49]; P = .26). Among those with severe disease, the primary outcome occurred in 91.3% (21/23) of the convalescent plasma group vs 68.2% (15/22) of the control group (HR, 2.15 [95% CI, 1.07-4.32]; P = .03); among those with life-threatening disease the primary outcome occurred in 20.7% (6/29) of the convalescent plasma group vs 24.1% (7/29) of the control group (HR, 0.88 [95% CI, 0.30-2.63]; P = .83) (P for interaction = .17). There was no significant difference in 28-day mortality (15.7% vs 24.0%; OR, 0.65 [95% CI, 0.29-1.46]; P = .30) or time from randomization to discharge (51.0% vs 36.0% discharged by day 28; HR, 1.61 [95% CI, 0.88-2.93]; P = .12). Convalescent plasma treatment was associated with a negative conversion rate of viral PCR at 72 hours in 87.2% of the convalescent plasma group vs 37.5% of the control group (OR, 11.39 [95% CI, 3.91-33.18]; P < .001). Two patients in the convalescent plasma group experienced adverse events within hours after transfusion that improved with supportive care. Conclusion and Relevance Among patients with severe or life-threatening COVID-19, convalescent plasma therapy added to standard treatment, compared with standard treatment alone, did not result in a statistically significant improvement in time to clinical improvement within 28 days. Interpretation is limited by early termination of the trial, which may have been underpowered to detect a clinically important difference. Trial Registration Chinese Clinical Trial Registry: ChiCTR2000029757
Article
Full-text available
A healthy patient presented to Klinikum Altmühlfranken Weißenburg Hospital, Germany, with twomorning attacks of painful muscle spasm in left upper and lower limbs, without altered consciousness. Full examinations, radiological imaging, electroencephalography, lumbar puncture, and autoimmune profile were either normal or not consistent. Subsequent epileptic episodes were observed on admission day and following days; thus, the patient was diagnosed with focal epilepsy. The patient started to develop a fever and severe cough at day 4, and SARS-Coronavirus-2 was confirmed through a nasopharyngeal swap. She received anticonvulsants and symptomatic treatments, and completely recovered. This report emphasizes on potential nervous system involvement in SARS-Coronavirus-2 pathogenesis.
Article
Full-text available
Background Remdesivir is an RNA polymerase inhibitor with potent antiviral activity in vitro and efficacy in animal models of coronavirus disease 2019 (Covid-19). Methods We conducted a randomized, open-label, phase 3 trial involving hospitalized patients with confirmed SARS-CoV-2 infection, oxygen saturation of 94% or less while they were breathing ambient air, and radiologic evidence of pneumonia. Patients were randomly assigned in a 1:1 ratio to receive intravenous remdesivir for either 5 days or 10 days. All patients received 200 mg of remdesivir on day 1 and 100 mg once daily on subsequent days. The primary end point was clinical status on day 14, assessed on a 7-point ordinal scale. Results In total, 397 patients underwent randomization and began treatment (200 patients for 5 days and 197 for 10 days). The median duration of treatment was 5 days (interquartile range, 5 to 5) in the 5-day group and 9 days (interquartile range, 5 to 10) in the 10-day group. At baseline, patients randomly assigned to the 10-day group had significantly worse clinical status than those assigned to the 5-day group (P=0.02). By day 14, a clinical improvement of 2 points or more on the ordinal scale occurred in 64% of patients in the 5-day group and in 54% in the 10-day group. After adjustment for baseline clinical status, patients in the 10-day group had a distribution in clinical status at day 14 that was similar to that among patients in the 5-day group (P=0.14). The most common adverse events were nausea (9% of patients), worsening respiratory failure (8%), elevated alanine aminotransferase level (7%), and constipation (7%). Conclusions In patients with severe Covid-19 not requiring mechanical ventilation, our trial did not show a significant difference between a 5-day course and a 10-day course of remdesivir. With no placebo control, however, the magnitude of benefit cannot be determined. (Funded by Gilead Sciences; GS-US-540-5773 ClinicalTrials.gov number, NCT04292899.)
Article
Full-text available
Objective: To evaluate the efficacy and safety of hydroxychloroquine (HCQ) in the treatment of patients with moderate coronavirus disease 2019 (COVID-19). Methods: We prospectively enrolled 30 treatment-naïve patients with confirmed COVID-19 after informed consent at Shanghai Public Health Clinical Center. The patients were randomized 1:1 to HCQ group and the control group. Patients in HCQ group were given HCQ 400 mg per day for 5 days plus conventional treatments, while those in the control group were given conventional treatment only. The primary endpoint was negative conversion rate of SARS-CoV-2 nucleic acid in respiratory pharyngeal swab on days 7 after randomization. This study has been approved by the Ethics Committee of Shanghai Public Health Clinical Center and registered online (NCT04261517). Results: One patient in HCQ group developed to severe during the treatment. On day 7, nucleic acid of throat swabs was negative in 13 (86.7%) cases in the HCQ group and 14 (93.3%) cases in the control group (P>0.05). The median duration from hospitalization to virus nucleic acid negative conservation was 4 (1,9) days in HCQ group, which is comparable to that in the control group [2 (1,4) days, Z=1.27, P>0.05]. The median time for body temperature normalization in HCQ group was 1 (0,2) day after hospitalization, which was also comparable to that in the control group [1 (0,3) day]. Radiological progression was shown on CT images in 5 cases (33.3%) of the HCQ group and 7 cases (46.7%) of the control group, and all patients showed improvement in follow-up examinations. Four cases (26.7%) of the HCQ group and 3 cases (20%) of the control group had transient diarrhea and abnormal liver function (P>0.05). Conclusions: The prognosis of COVID-19 moderate patients is good. Larger sample size study are needed to investigate the effects of HCQ in the treatment of COVID-19. Subsequent research should determine better endpoint and fully consider the feasibility of experiments such as sample size.
Article
Full-text available
Patients with COVID-19 have a coagulopathy and high thrombotic risk. In a cohort of 69 intensive care unit (ICU) patients we investigated for evidence of heparin resistance in those that have received therapeutic anticoagulation. 15 of the patients have received therapeutic anticoagulation with either unfractionated heparin (UFH) or low molecular weight heparin (LMWH), of which full information was available on 14 patients. Heparin resistance to UFH was documented in 8/10 (80%) patients and sub-optimal peak anti-Xa following therapeutic LMWH in 5/5 (100%) patients where this was measured (some patients received both anticoagulants sequentially). Spiking plasma from 12 COVID-19 ICU patient samples demonstrated decreased in-vitro recovery of anti-Xa compared to normal pooled plasma. In conclusion, we have found evidence of heparin resistance in critically unwell COVID-19 patients. Further studies investigating this are required to determine the optimal thromboprophylaxis in COVID-19 and management of thrombotic episodes.
Article
Full-text available
Introduction: The coronavirus disease 2019 (COVID-19) caused by the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has developed since December 2019. It has caused a global pandemic with more than three hundred thousand case fatalities. However, apart from supportive care by respirators, no standard medical therapy is validated. Areas covered: This paper presents old drugs with potential in vitro efficacy against SARS-CoV-2. The in vitro database, adverse effects, and potential toxicities of these drugs are reviewed regarding their feasibility of clinical prescription for the treatment of patients with COVID-19. To obtain convincing recommendations, we referred to opinions from the US National Institute of Health regarding drugs repurposed for COVID-19 therapy. Expert opinion: Although strong evidence of well-designed randomized controlled studies regarding COVID-19 therapy is presently lacking, remdesivir, teicoplanin, hydroxychloroquine (not in combination with azithromycin), and ivermectin might be effective antiviral drugs and are deemed promising candidates for controlling SARS-CoV-2. In addition, tocilizumab might be considered as the supplementary treatment for COVID-19 patients with cytokine release syndrome. In future, clinical trials regarding a combination of potentially effective drugs against SARS-CoV-2 need to be conducted to establish the optimal regimen for the treatment of patients with moderate-to-severe COVID-19.
Article
Full-text available
Objective To assess the efficacy and safety of hydroxychloroquine plus standard of care compared with standard of care alone in adults with coronavirus disease 2019 (covid-19). Design Multicentre, open label, randomised controlled trial. Setting 16 government designated covid-19 treatment centres in China, 11 to 29 February 2020. Participants 150 patients admitted to hospital with laboratory confirmed covid-19 were included in the intention to treat analysis (75 patients assigned to hydroxychloroquine plus standard of care, 75 to standard of care alone). Interventions Hydroxychloroquine administrated at a loading dose of 1200 mg daily for three days followed by a maintenance dose of 800 mg daily (total treatment duration: two or three weeks for patients with mild to moderate or severe disease, respectively). Main outcome measure Negative conversion of severe acute respiratory syndrome coronavirus 2 by 28 days, analysed according to the intention to treat principle. Adverse events were analysed in the safety population in which hydroxychloroquine recipients were participants who received at least one dose of hydroxychloroquine and hydroxychloroquine non-recipients were those managed with standard of care alone. Results Of 150 patients, 148 had mild to moderate disease and two had severe disease. The mean duration from symptom onset to randomisation was 16.6 (SD 10.5; range 3-41) days. A total of 109 (73%) patients (56 standard of care; 53 standard of care plus hydroxychloroquine) had negative conversion well before 28 days, and the remaining 41 (27%) patients (19 standard of care; 22 standard of care plus hydroxychloroquine) were censored as they did not reach negative conversion of virus. The probability of negative conversion by 28 days in the standard of care plus hydroxychloroquine group was 85.4% (95% confidence interval 73.8% to 93.8%), similar to that in the standard of care group (81.3%, 71.2% to 89.6%). The difference between groups was 4.1% (95% confidence interval –10.3% to 18.5%). In the safety population, adverse events were recorded in 7/80 (9%) hydroxychloroquine non-recipients and in 21/70 (30%) hydroxychloroquine recipients. The most common adverse event in the hydroxychloroquine recipients was diarrhoea, reported in 7/70 (10%) patients. Two hydroxychloroquine recipients reported serious adverse events. Conclusions Administration of hydroxychloroquine did not result in a significantly higher probability of negative conversion than standard of care alone in patients admitted to hospital with mainly persistent mild to moderate covid-19. Adverse events were higher in hydroxychloroquine recipients than in non-recipients. Trial registration ChiCTR2000029868.
Article
Background Hydroxychloroquine or chloroquine, often in combination with a second-generation macrolide, are being widely used for treatment of COVID-19, despite no conclusive evidence of their benefit. Although generally safe when used for approved indications such as autoimmune disease or malaria, the safety and benefit of these treatment regimens are poorly evaluated in COVID-19. Methods We did a multinational registry analysis of the use of hydroxychloroquine or chloroquine with or without a macrolide for treatment of COVID-19. The registry comprised data from 671 hospitals in six continents. We included patients hospitalised between Dec 20, 2019, and April 14, 2020, with a positive laboratory finding for SARS-CoV-2. Patients who received one of the treatments of interest within 48 h of diagnosis were included in one of four treatment groups (chloroquine alone, chloroquine with a macrolide, hydroxychloroquine alone, or hydroxychloroquine with a macrolide), and patients who received none of these treatments formed the control group. Patients for whom one of the treatments of interest was initiated more than 48 h after diagnosis or while they were on mechanical ventilation, as well as patients who received remdesivir, were excluded. The main outcomes of interest were in-hospital mortality and the occurrence of de-novo ventricular arrhythmias (non-sustained or sustained ventricular tachycardia or ventricular fibrillation). Findings 96 032 patients (mean age 53·8 years, 46·3% women) with COVID-19 were hospitalised during the study period and met the inclusion criteria. Of these, 14 888 patients were in the treatment groups (1868 received chloroquine, 3783 received chloroquine with a macrolide, 3016 received hydroxychloroquine, and 6221 received hydroxychloroquine with a macrolide) and 81 144 patients were in the control group. 10 698 (11·1%) patients died in hospital. After controlling for multiple confounding factors (age, sex, race or ethnicity, body-mass index, underlying cardiovascular disease and its risk factors, diabetes, underlying lung disease, smoking, immunosuppressed condition, and baseline disease severity), when compared with mortality in the control group (9·3%), hydroxychloroquine (18·0%; hazard ratio 1·335, 95% CI 1·223–1·457), hydroxychloroquine with a macrolide (23·8%; 1·447, 1·368–1·531), chloroquine (16·4%; 1·365, 1·218–1·531), and chloroquine with a macrolide (22·2%; 1·368, 1·273–1·469) were each independently associated with an increased risk of in-hospital mortality. Compared with the control group (0·3%), hydroxychloroquine (6·1%; 2·369, 1·935–2·900), hydroxychloroquine with a macrolide (8·1%; 5·106, 4·106–5·983), chloroquine (4·3%; 3·561, 2·760–4·596), and chloroquine with a macrolide (6·5%; 4·011, 3·344–4·812) were independently associated with an increased risk of de-novo ventricular arrhythmia during hospitalisation. Interpretation We were unable to confirm a benefit of hydroxychloroquine or chloroquine, when used alone or with a macrolide, on in-hospital outcomes for COVID-19. Each of these drug regimens was associated with decreased in-hospital survival and an increased frequency of ventricular arrhythmias when used for treatment of COVID-19. Funding William Harvey Distinguished Chair in Advanced Cardiovascular Medicine at Brigham and Women's Hospital.