Open access peer-reviewed chapter

Pharmacotherapy for COVID-19: A Ray of Hope

Written By

Mayank Kapoor, Prasan Kumar Panda and Vivek Mohanty

Submitted: 23 February 2021 Reviewed: 04 March 2021 Published: 17 May 2021

DOI: 10.5772/intechopen.97012

From the Edited Volume

Fighting the COVID-19 Pandemic

Edited by Manal Mohammad Baddour

Chapter metrics overview

427 Chapter Downloads

View Full Metrics

Abstract

Most viral infections have limited treatment options available and the same holds for COVID-19, its causative agent being the SARS-CoV-2 virus. Drugs used in the past against Severe Acute Respiratory Syndrome (SARS) or Middle East Respiratory Syndrome (MERS) viruses, which belong to the same family of viruses as the novel Coronavirus included ribavirin, interferon (alfa and beta), lopinavir-ritonavir combination, and corticosteroids. There remains controversy regarding their efficacy to date, except for the last one. Hence, large-scale multicentric trials are being conducted involving multiple drugs. Chloroquine and hydroxy-chloroquine were initially taking the race ahead but have now been rejected. Remdesivir was a promising candidate, for which the FDA had issued an emergency use authorization, but now is not recommended by the WHO. Convalescent plasma therapy had promising results in the early severe viremia phase, but the PLACID trial made an obscure end. Only corticosteroids have shown demonstrable benefits in improving mortality rates among severe COVID-19 cases. Many new modalities like monoclonal antibodies and tyrosine kinase inhibitors are discussed. In this chapter, we review the therapeutic drugs under investigation for the COVID-19 treatment, their mode of action, degree of effectiveness, and recommendations by different centers regarding their use in current settings.

Keywords

  • antiviral
  • monoclonal antibody
  • coronavirus disease 2019
  • dexamethasone
  • immunomodulator
  • ivermectin
  • remdesivir

1. Introduction

Because of the high rate of infectivity of the COVID-19 virus, the global burden associated with the disease, and its impact on the economies of different countries, efforts are being made to find a possible cure for the disease as soon as possible [1]. As with most viral infections, limited options are available for the treatment of COVID-19. Since there is no efficient therapy available for the same, given the public emergency, efforts are ongoing to find drugs helpful in COVID-19 infection. Drugs used in the past against Severe Acute Respiratory Syndrome (SARS) or Middle East Respiratory Syndrome (MERS), which also belong to the group of Beta coronaviruses, included ribavirin, interferon, lopinavir-ritonavir, and corticosteroids [2]. Most randomized controlled trials (RCTs) performed to test the effectiveness of these drugs have not shown any satisfying results, apart from corticosteroids. Many RCTs are still undergoing, the results of which are awaited. Studies about the virus-induced host immune response and viral processing within target cells have led to several potential therapeutic targets. We hereby discuss the potential therapeutic drugs under investigation for the COVID-19 treatment, their modes of action (Figure 1), degree of effectiveness, and recommendations (Table 1) by different centers regarding their usage in the current settings.

Figure 1.

Site of action of different possible pharmacotherapeutics used in COVID-19 treatment.

DrugMode of actionEffectivenessRecommendation
CorticosteroidsImmunosuppressantDecreased death rate in ARDS, no effect in non-ARDSWHO, CDC, and IDSA recommendations
RemdesivirRdRp inhibitorDecreases recovery timeFDA approval in October, WHO issued a conditional recommendation against use in November, IDSA suggests the use
Convalescent plasmaAnti-COVID 19 antibodiesNo benefitFDA EUA issued
Monoclonal antibodiesDirected against COVID spike proteinsBenefit in Mild cases, no benefit in hospitalized casesFDA EUA issued for OPD patients
AzithromycinImmunomodulationNo benefitNo recommendation, but widely used
IvermectinViral IMPα/β1(Importin) mediated nuclear import inhibitionBenefit in prophylaxisNIH: Insufficient data for or against the use
MelatoninPineal gland hormone, anti-inflammatoryBenefit in critical patientsNo recommendation
TocilizumabIL-6 R inhibitorReduces inflammatory markersSingle-dose in addition to dexamethasone in critical patients with rapid progression of respiratory failure may be given: NIH
FavipiravirInhibits RNA polymeraseFaster viral clearance, improved imaging findingsNo recommendation yet
RibavirinInhibits RNA polymeraseNo concrete evidenceNo recommendations yet
Chloroquine/ Hydroxy-chloroquineIncreases endosomal pH, interfere with glycosylation of receptor, immunomodulatorBenefit in clinical parameter & virological clearanceRemoved from Solidarity trial, no other recommendation
Lopinavir/ RitonavirProtease inhibitor: SARS- Cov-2 3CL proNot significantRemoved from Solidarity trial, no other recommendation
InterferonImmunomodulationNo concrete evidenceRemoved from Solidarity trial, no other recommendation
Tyrosine Kinase inhibitorsInhibit STAT phosphorylation, decrease hyperimmune stateNo concrete evidenceUse with remdesivir if corticosteroids are contraindicated: NIH/IDSA

Table 1.

Summary of various pharmacotherapeutics being considered for COVID-19 treatment.

RdRp: RNA dependent RNA polymerase; ARDS: Acute respiratory distress syndrome; WHO: World Health Organization; CDC: Centers for Disease Control and Prevention; FDA: Food and Drug Administration; IDSA: Infectious Disease Society of America; EUA: Emergency Use Authorization; NIH: National Institute of Health; Il-6 R: Interleukin-6 receptor; STAT: Signal transducer and activator of transcription.

Advertisement

2. Review of pharmacotherapy

2.1 Chloroquine/hydroxychloroquine

The first studied drugs for COVID-19 were chloroquine and hydroxychloroquine (HCQ). Chloroquine was found to be effective against Avian influenza A H5N1 virus in animal models [3, 4] and also had demonstrable activity resulting in in-vitro inhibition of SARS-CoV [5]. COVID-19 infection showed high pandemicity in countries where malaria is the least prevalent and least pandemicity where malaria is highly prevalent. This observation led to the concept that chloroquine may be beneficial in COVID-19 since it is used as an anti-malarial. The mechanism of chloroquine action depends on the pathogen involved. Chloroquine increases the endosomal pH and interferes with the glycosylation of cellular receptor [Angiotensin Converting Enzyme (ACE) II] of SARS-CoV [6]. It also inhibits quinone reductase-2, which is involved in sialic acid biosynthesis. There is inhibition of MAO-kinase, virion assembly, and processing of M protein [7]. Besides its antiviral activity, it also has immunomodulatory effects that may be synergistic. HCQ was found to be equally effective as chloroquine, although a study concluded that HCQ was more effective and less toxic than chloroquine [8]. Chloroquine inhibitory actions against SARS-CoV were equal whether the primate cells were treated before or after exposure. This suggested that chloroquine could have both prophylactic and therapeutic applications [9]. One of the first studies performed to study the effect of chloroquine was done in the Chinese population. In this trial, patients in the study group who received chloroquine had reduced symptom duration, radiological improvement, and earlier seroconversion to the virus-negative state compared to controls [10]. Following this study, the National Health Commission of the People’s Republic of China included chloroquine in its guideline for the management of pneumonia due to Covid-19. In a study conducted by Gautret et al. in France, chloroquine treatment group had significant clearing of the nasopharyngeal swab viral load compared to the control [11]. The virological clearance day-6 post inclusion (primary outcome) with HCQ vs. controls was 70% vs. 12.5% (p < 0.001). The virological clearance at day 6 in HCQ plus azithromycin, HCQ and control arms were 100%, 57.1%, and 12.5% respectively (p = 0.001) thus suggesting synergistic action of azithromycin to HCQ . Gradually the side effect profile of HCQ , that is QTc prolongation with concomitant use of Azithromycin, lead the American Heart Association (AHA) to recommend withdrawal/withholding these drugs in patients with QTc ≥ 500 millisecond (either baseline or developing during treatment). On 28 March 2020, Food and Drug Administration (FDA) had issued Emergency Use Authorization (EUA) for Chloroquine/HCQ . However, the Centers for Disease Control and Prevention (CDC) on April 7 issued a statement stating no drugs or other therapeutic measures were approved by the US FDA to prevent or treat COVID-19. In April, the FDA issued a Drug Safety Communication cautioning against the use of HCQ or chloroquine for COVID-19 outside the hospital setting or a clinical trial due to the risk of heart rhythm problems. In June 2020, it was announced by World Health Organization (WHO) that the HCQ arm of the Solidarity Trial (Multi-national trial including remdesivir, HCQ , lopinavir/ritonavir, and lopinavir/ritonavir with interferon beta-1a) would be stopped [12]. This was keeping in view the lack of any mortality benefit of HCQ . Hence in June itself, FDA revoked the EUA of HCQ and chloroquine [13]. The pre-exposure prophylaxis benefit of HCQ needs further research.

2.2 Lopinavir/ritonavir

The combination of lopinavir/ritonavir was considered as an option for the treatment of Covid-19 during initial pandemic days. Lopinavir is an HIV-1 protease inhibitor, which is combined with ritonavir to increase its half-life through cytochrome p-450 inhibition. Both anti-HIV drugs interact with residues at the active site of SARS-CoV 3C-like protease, suggesting the mechanism of action in COVID-19 [14]. Its role was first evaluated in the treatment of SARS where patients treated with lopinavir/ritonavir for 14 days combined with ribavirin for 21 days. They had a milder disease in form of less diarrhea, fever, lymphadenopathy, the incidence of nosocomial infections, viral loads, demonstration of virus in the fecal sample by reverse transcription-polymerase chain reaction (RT-PCR), and 21 days adverse outcomes [15]. The combination was tested for MERS-CoV. It was postulated that the lopinavir/ritonavir combination may inhibit the 3C-like protease of MERS-CoV and may affect apoptosis in human cells. Results revealed that treatment with lopinavir/ritonavir led to clinical, radiological, and pathological improvement. Those animals treated with this combination had the lowest mean viral load detected by RT-PCR in lung and other extrapulmonary tissue [16]. There was only a single case report of a man being treated and recovered with a combination of lopinavir/ritonavir, ribavirin, and interferon-α for the MERS [17]. Based on this data, an urgent RCT was done to study the efficacy of lopinavir/ritonavir in the Wuhan province of China [18]. The analysis revealed no significant difference in terms of time for clinical improvement and mortality at 28 days. The median time for clinical improvement was just one day shorter in the lopinavir-ritonavir group compared to the standard care group. In July 2020, WHO discontinued the lopinavir/ritonavir arm of the solidarity trial due to a lack of any mortality benefit [19]. It causes QTc prolongation, just like HCQ  [20].

2.3 Azithromycin

Azithromycin is a broad-spectrum antibiotic belonging to the macrolide group, having anti-inflammatory properties also. It is commonly used for treating atypical respiratory pathogens. Azithromycin’s anti-viral efficacy against some RNA viruses has also been described. Its efficacy has been demonstrated in-vitro against Zika virus and rhinovirus, as well as SARS-CoV-2 [21, 22]. As described, azithromycin also has immunomodulatory effects and can decrease acute exacerbations of chronic airway disease. Owing to its wide availability, excellent safety profile, and easy availability, azithromycin is one of the commonest drugs being used in the COVID-19 pandemic also. The Lancet reported the result of the COALITION II trial, [23] which was an open-label randomized trial evaluating azithromycin in addition to the standard of care (including HCQ), against the standard of care alone in severe COVID-19 patients. Azithromycin demonstrated no benefit in clinical outcome including clinical status or mortality, as compared to the standard of care alone (OR 1·36 [95% CI 0·94–1·97], p = 0·11). There was no increase in adverse events with azithromycin. In a study published in NEJM, HCQ alone or in combination with azithromycin had no demonstrable improvement in clinical status at 15 days compared with standard care in mild to moderate COVID-19 admissions [24].

2.4 Ivermectin

Ivermectin is a commonly used drug for various parasitic infestations including head lice, scabies, strongyloidiasis, ascariasis, and lymphatic filariasis. It is a macrocyclic lactone, which is derived from streptomyces avermitilis [25]. Its mechanism of activity against SARS-CoV-2 is believed to be via viral IMPα/β1 (Importin) mediated nuclear import inhibition. This leads to a decrease in the multiplication of the virus and hence the viral load [26, 27]. Ivermectin and doxycycline combination also inhibit viral entry and increases viral load clearance by the targeting of multiple viral proteins [28]. A recent study from India demonstrated that 2-dose ivermectin prophylaxis (300 micrograms/kg) within a gap of 3 days led to a 73% reduction in the number of COVID-19 infections among healthcare workers [29]. In studies conducted in Bangladesh also, the ivermectin-doxycycline combination was demonstrated to be highly effective in virological clearance in mild to moderate COVID-19 patients [30, 31]. National Institute of Health (NIH) stated in January 2021 that there was insufficient data to recommend either for or against the use of ivermectin for the treatment of COVID-19 [32].

2.5 Melatonin

Melatonin is a hormone, which is synthesized from tryptophan in the pineal gland of the body and also by mostly all the organs of the body, as its production is associated with mitochondria. Higher levels of melatonin have positive roles in health and aging. Melatonin promises to be a great adjunctive drug for viral infections owing to its anti-inflammatory, anti-apoptotic, immunomodulatory, and antioxidant activities [33]. Sirtuin-1 (SIRT1) is the proposed mediator of melatonin’s anti-inflammatory action. This is via inhibition of high mobility group boxechromosomal protein-1 (HMGB-1), leading to down-regulation of the polarization of macrophages towards pro-inflammatory type [34]. It inhibits the over-activity of the innate immune system. Hence, theoretically, the cytokine storm induced by COVID-19 can be suppressed by melatonin. But the use of melatonin in COVID-19 is still very sparse, with only a few studies evaluating the same, hence further research is warranted [35]. Owing to melatonin’s anti-inflammatory, anti-oxidant, and anti-viral actions, its role in critical illness caused due to COVID has been studied. Melatonin has easy availability, is not expensive, and has an excellent safety profile [36]. A trial (EudraCT: 2020–001808-42) is ongoing for the identification of the doses of melatonin that may prove effective in this disease. It is a phase II, single-center, double-blind, RCT to address the efficacy and safety of intravenous melatonin in COVID-19 ICU patients [37].

2.6 Remdesivir

Remdesivir is a 1′-cyano-substituted adenosine nucleotide analog prodrug, which was found to be effective against several RNA viruses. It was initially developed in 2017 by Gilead science for the treatment of the Ebola virus [38]. It has demonstrated extensive antiviral activity & effective treatment of lethal Ebola and Nipah virus infections in nonhuman primates [39]. Subsequently, it was investigated for SARS-CoV and MERS-CoV. Studies have shown that Remdesivir inhibits viral replication in human airway epithelial cell culture by affecting the early stages of viral replication through viral RNA synthesis inhibition, as an RNA-dependent RNA polymerase (RdRp) inhibitor [39]. This may be due to the absence of Exon-mediated proofreading in viruses sensitive to Remdesivir [40]. One of the first trials of Remdesivir was performed by the Gilead sciences center in hospitalized patients with confirmed SARS-CoV-2 having oxygen saturation < 94% or a need for oxygen support. Till 28 days of follow-up, the cumulative incidence of clinical improvement was 84% (95% CI 70–99) by Kaplan–Meier analysis and it was less among patients receiving invasive ventilation compared to non-invasive ventilation [41]. In another randomized, double-blind, placebo-controlled, multicentre trial at 10 hospitals in Hubei, China, Remdesivir use was not associated with any difference in time to clinical improvement [42]. In February 2020, WHO cast a vote of confidence for remdesivir, indicating that it has great potential. In April 2020, the US National Institute of Allergy and Infectious Diseases (NIAID), announced that a clinical trial in >1,000 people showed that those taking remdesivir recovered in 11 days on average, compared with 15 days for those on a placebo, even adding that remdesivir may become a standard for COVID treatment [43]. US FDA had issued a EUA for remdesivir for severe COVID-19 disease. On 22nd October 2020, the FDA approved remdesivir for use in adult and pediatric patients (≥12 years, ≥40 kg) requiring hospitalization [44]. In October 2020 itself, an interim analysis of the WHO-led, open-label, randomized SOLIDARITY trial demonstrated that 301 (11·0%) of 2743 patients who received remdesivir and 303 (11·2%) of 2708 patients who received standard care died by day 28 (Kaplan–Meier rate ratio 0·95, 95% CI 0·81–1·11; p = 0·50) [45]. The ACTT-1 study had also reported a 29-day mortality of 11·4% in patients receiving remdesivir as compared to 15·2% in placebo (hazard ratio 0·73, 95% CI 0·52–1·03) [43]. Hence in November 2020, WHO issued a conditional recommendation against remdesivir utilization in hospitalized patients, regardless of their disease severity. This was because they could not find evidence of remdesivir improving survival and other outcomes in the patients [46]. Infectious Disease Society of America (IDSA) still suggests the use of remdesivir in severe and critical patients, as does NIH [47, 48].

2.7 Tocilizumab

Tocilizumab is an Interleukin-6 (IL-6) Receptor inhibiting monoclonal antibody. Studies have shown that infection with the SARS virus leads to a cytokine storm with the release of inflammatory cytokines like IL-6, Tumor Necrosis Factor- α (TNF –α), and IL-12 [49]. Further research done on MERS-CoV showed IL-6, IL-1β, and IL-8 were elevated in these patients. In patients with confirmed COVID-19 infection who were admitted to ICU, levels of IL-2, IL-6, IL-7, IL-10, granulocyte-colony stimulating factor (G-CSF), interferon-γ-inducible protein (IP10), monocyte chemoattractant protein (MCP1), macrophage inflammatory protein 1 alpha (MIP1A), and TNF-α levels were found to be high, suggesting possible cytokine storm [50]. The first trial involving tocilizumab was performed in China in February 2020. The National Institute for Infectious disease had recommended tocilizumab in moderate to severe infections and IL-6 levels >40 pg/mL (or D-dimer levels >1000 ng/mL). However, it is not recommended by the CDC. In an RCT published in JAMA, in moderate-to-severe pneumonia, tocilizumab did not reduce the WHO Clinical Progression Scale scores. The proportion of patients with non-invasive or invasive ventilation or death at day 14 was 36% with usual care and 24% with tocilizumab. There were no differences in 28 days mortality. This meant tocilizumab could decrease the requirement for mechanical and non-invasive ventilation or death by day 14 but not mortality by day 28 [51, 52]. An RCT published by NEJM in October 2020, which included patients fulfilling at least two of the following: fever, pulmonary infiltrates, or the need for oxygen therapy to maintain oxygen saturation more than 92%, concluded that tocilizumab was not effective in preventing intubation or death in moderately ill hospitalized patients with Covid-19 [53]. Sarilumab, another IL-6 receptor antagonist was being tested in a multicentre trial for hospitalized patients with severe COVID-19 [54]. It was concluded that at 28 days, clinical improvement and mortality in severe COVID-19 were not significantly different between sarilumab and standard of care [55]. Preliminary results from the Randomized, Embedded, Multifactorial Adaptive Platform Trial for Community-Acquired Pneumonia (REMAP-CAP) were released in a non-peer-reviewed report. REMAP-CAP is the largest trial to date investigating the use of IL-6 inhibitors in COVID-19. In February 2021, after reviewing the evidence from REMAP-CAP and other trials, the NIH Panel revised the recommendations on the use of tocilizumab and sarilumab, stating there was insufficient data to recommend either for or against the use of these drugs. But given the REMAP-CAP trial, some members suggested administering a single dose of tocilizumab (8 mg/kg of actual body weight, max 800 mg) in addition to dexamethasone in the ICU patients having high oxygen requirements/invasive and non-invasive mechanical ventilation and exhibiting rapid progression of respiratory failure [56]. The number of patients receiving sarilumab in the REMAP-CAP trial was too low to assess the efficacy.

2.8 Convalescent plasma

There was a hypothesis that plasma collected from the persons who have recovered from Covid-19 may contain antibodies against SARS-CoV-2, which may be used as a treatment tool. A case series was done in China where 5 critically ill patients with confirmed Covid-19 and Acute respiratory distress syndrome (ARDS) were selected [57]. They received two consecutive transfusions of 200 mL to 250 mL of convalescent plasma (total dose: 400 mL) with a SARS-CoV-2-specific antibody (IgG) titer more than 1:1,000. After receiving the plasma, the SOFA score of the patients decreased and ventilator parameters of the patients (pO2/FiO2 ratio) of the patient improved, and viral load decreased by day 12. ARDS resolved in four patients by Day 12 and 3 were weaned off the ventilators by 2 weeks. Further trials are needed the study the effectiveness of convalescent plasma. FDA is encouraging people who have fully recovered from COVID-19 for at least two weeks to donate plasma. FDA had issued guidance providing recommendations to health care providers & investigators on administration and study of COVID-19 convalescent plasma during the public health emergency. FDA issued a EUA for convalescent plasma on August 23, 2020, although convalescent plasma did not show any stoppage of progression to severe COVID-19 or all-cause mortality in the PLACID trial [58, 59]. In a trial published in NEJM in November 2020, in 228 patients receiving convalescent plasma and 105 receiving placebo at 30 days, there was not any significant difference among the clinical outcome distribution (odds ratio [OR], 0.83 (95% CI, 0.52–1.35; P = 0.46). Mortality in the plasma group was 10.96% as compared to 11.43% in the placebo group [risk difference 0.46% points (95% CI, −7.8 to 6.8) [60].

2.9 Favipiravir

Favipiravir (FPV) is a purine nucleotide that inhibits viral RNA polymerase. It was initially used against Ebola but later found to have in-vitro activity against other RNA viruses. The EC50 (concentration of a drug that gives half-maximal response) of FPV against SARS-CoV-2 in vitro in Vero E6 cells was found to be 61.88 μM/L [6, 61]. A study investigated the effect of FPV vs. lopinavir/ritonavir on the treatment of COVID-19. FPV was independently associated with faster viral clearance and a higher improvement rate in chest imaging. These findings suggest that FPV has significantly better treatment effects on COVID-19 in terms of disease progression and viral clearance as compared with lopinavir/ritonavir [62]. In an RCT on moderate to severe COVID patients, FPV was compared with umifenovir (Arbidol) by measuring the clinical recovery at 7 days [63]. Results showed no significant differences between the 2 groups. At present, there are no recommendations for the use of FPV in Covid-19 patients. Just like HCQ & lopinavir/ritonavir combination, it also causes QT prolongation [20].

2.10 Ribavirin

Ribavirin, a guanine analog, inhibits viral RNA dependent RNA polymerase (RdRp). It has demonstrable activity against many coronaviruses, but when used against SARS-CoV it was found to have less effectiveness in vitro requiring higher doses with combination therapy. When used with interferon in the treatment of MERS-CoV, no benefits were observed in terms of clinical outcomes or the rate of virus clearance [64]. Ribavirin also causes dose-dependent hematological toxicity & transaminase elevation when used in SARS patients and being a teratogen, is contraindicated in pregnancy [65, 66]. A recent trial showed ribavirin not being associated with better negative conversion times for the SARS-CoV-2 test and not being associated with improved mortality rates [67]. Due to its lack of demonstrable efficacy against other coronaviruses and high toxicity profile, it has got a limited role in the treatment of Covid-19. However, its combination with other antivirals is being tried in the SEV trial, the result of which is yet to be published [68].

2.11 Interferons

Studies with interferon-β had shown its activity against MERS. Most studies involved combination therapy with lopinavir/ritonavir or ribavirin. However, there was no concrete evidence showing its effect on SARS- CoV-2 in-vitro and the lack of clinical trials precluded the justification for its use in Covid-19 patients and hence there are no recommendations regarding its use [69]. In a study, it was shown that early triple antiviral therapy with lopinavir/ritonavir, ribavirin, and interferon beta-1b was safe and superior to lopinavir-ritonavir combinations alone in alleviating symptoms and shortening the duration of viral shedding and hospital stay in patients with mild to moderate COVID-19 [70]. In a trial utilizing interferon β-1a, clinical response time was not significantly different between interferon and the control groups (9.7 ± 5.8 versus 8.3 ± 4.9 days, respectively, P = 0.95). On day 14, 66.7% versus 43.6% of patients in the interferon group and the control group respectively was discharged (OR, 2.5; 95% CI, 1.05–6.37). The 28-day overall mortality was significantly less in the interferon than the control group (19% versus 43.6%, respectively, P = 0.015). Early administration significantly decreased mortality (OR, 13.5; 95% CI, 1.5–1.18) [71]. Another trial testing interferon β-1b showed its effectiveness in reducing the clinical improvement time without any serious adverse events in severe COVID-19 patients. ICU admission and invasive ventilation need also decreased following administration of interferon β-1b [72]. The Lancet Respiratory Medicine showed the results of an RCT of nebulized interferon beta-1a in 101 adults admitted to the hospital with COVID-19. It demonstrated better odds of clinical improvement than placebo (OR 2·32 [95% CI 1·07–5·04]; p = 0·03). No significant difference was there in the hospital discharge odds by day 28 [73]. Recently, the SOLIDARITY trial also showed no benefit of interferon therapy [74].

2.12 Corticosteroids

ARDS is a leading cause of mortality in Covid-19 pneumonia. Cytokine storm plays a key role in the pathogenesis of ARDS in Covid-19 patients and thus immunosuppression may have a role in the treatment of such patients [75]. Glucocorticoids modify the inflammation-mediated lung injury and hence can alter progression to respiratory failure and death. Studies on SARS and MERS showed that corticosteroids did not show any improvement in overall survival but showed delayed viral clearance from the respiratory tract and other steroid-related complications like Hyperglycaemia & Psychosis [76]. A retrospective study was carried out in Covid-19 patients in China who had developed ARDS. Those who received steroids had decreased death rates compared to those who did not [77]. In another study in non-ARDS patients, corticosteroid treatment did not influence virus clearance time, hospital length of stay, or duration of symptoms in mild COVID-19. Another study reported that early application of low-dose corticosteroid improves the treatment effect, presenting as improvement of hypoxia and fever, shortening disease course, and accelerating focus absorption [78]. Steroids are now the only therapy showing mortality benefit in COVID-19 severe disease. RECOVERY trial has concluded that dexamethasone 6 mg given once daily for up to 10 days decreased 28-day mortality in patients with COVID-19 on respiratory support. But a careful decision has to be made regarding severity as patients not requiring oxygen showed no benefit but had a possibility of harm with corticosteroid therapy. In the dexamethasone group, the incidence of death was less than the usual care group among patients receiving invasive mechanical ventilation (29.3% vs. 41.4%; rate ratio, 0.64; 95% CI, 0.51–0.81) and those receiving oxygen without invasive mechanical ventilation (23.3% vs. 26.2%; rate ratio, 0.82; 95% CI, 0.72–0.94). No benefit was demonstrated among those who were receiving no respiratory support at randomization (17.8% vs. 14.0%, rate ratio, 1.19; 95% CI, 0.91–1.55) [79]. Subsequent RCTs also confirmed the same. Hence, all guidelines advocated steroids as first-line therapy in severe COVID-19. In due course, specific dose, route, and duration of therapy will be answered.

2.13 Monoclonal antibodies

Various novel monoclonal antibodies are under investigation for COVID-19. In a study published in NEJM, it has been described that LY-CoV555 (bamlanivimab) (also known as LY3819253), is a potent anti-spike neutralizing monoclonal antibody [80]. It binds to the receptor-binding domain of SARS-CoV-2. It was extracted from the convalescent plasma obtained from a COVID-19 patient. The protection of bamlanivimab against SARS-CoV-2 in primates has been reported [81]. In the interim analysis of data, patients receiving LY-CoV555 reported fewer hospitalizations and a lesser symptom burden than placebo receivers. In November 2020, it got the FDA EUA [82]. According to FDA, bamlanivimab reduced COVID-19 related hospital admissions in patients who are at high risk for disease progression [83]. This authorization came even after the company making the drug, Lilly, had announced in October 2020 that it was holding the trial in the hospital admitted patients as it not showing any benefits in them (ACTIV-3 trial). Remaining studies of bamlanivimab remain ongoing, including ACTIV-2 trial which includes the newly diagnosed mild to moderate COVID-19 patients; BLAZE-1, including recently diagnosed COVID-19 patients in the ambulatory (non-hospitalized) setting, studying bamlanivimab as monotherapy and in combination with etesevimab; and BLAZE-2, a phase 3 study for COVID-19 prophylaxis. Based on BLAZE-1 data, Lilly had submitted a request for EUA for bamlanivimab for the treatment of recently diagnosed mild to moderate COVID-19 patients to the FDA [84]. FDA reported 3% hospitalizations and emergency room visits in bamlanivimab treated patients compared to 10% in placebo. The FDA has approved bamlanivimab for patients age ≥ 12, and at high risk for progressing to severe covid-19 or hospital admission. However, it is emphasized that bamlanivimab should not be given to in-hospital COVID-19 patients or those requiring oxygen therapy; as such monoclonal antibodies may worsen outcomes in these patients. Another potential antibody treatment for COVID-19, REGN-COV2, a combination of two monoclonal antibodies casirivimab and imdevimab (REGN10933 and REGN10987), also faced some issues among inpatients with high oxygen requirements. In November 2020, the FDA issued EUA to monoclonal antibodies casirivimab and imdevimab (REGN10933 and REGN10987- against spike proteins of SARS-CoV-2) to be administered together for the treatment of mild to moderate COVID-19 in adults and pediatric patients (≥12 years of age) [85]. Although, in this case also, Regeneron Pharma had to halt its antibody cocktail trial in the admitted patients due to safety concerns, hence it was approved for non-admitted patients only [86]. Interestingly, US President Donald Trump had also received this regime when he tested positive for COVID-19 [87]. Astra Zeneca’s COVID-19 Long-Acting AntiBody (LAAB) combination AZD7442 trial has also advanced into Phase III [88]. On February 9, 2021, the FDA has issued a EUA for bamlanivimab plus etesevimab for the management of mild to moderate COVID-19 in outpatients at high risk for disease progression. The data come from a randomized, double-blind, placebo-controlled clinical trial in 1,035 non-hospitalized adults with mild to moderate COVID-19, at high risk for progression to severe disease. Hospitalization or death occurred in 36 (7%) of placebo recipients compared to 11 (2%) patients treated with bamlanivimab 2,800 milligrams and etesevimab 2,800 milligrams administered together, demonstrating a 70% reduction [89].

2.14 Janus kinase (JAK) inhibitors

The kinase inhibitors are being proposed as a novel modality of COVID-19 treatment. The rationale behind this being the prevention of phosphorylation of key proteins that are involved in the signal transduction that in turn leads to immunological activation and inflammation. This includes the cellular responses to the pro-inflammatory cytokines like IL-6 [90]. JAK inhibitors interfere with the phosphorylation of signal transducer and activator of transcription (STAT) proteins [91, 92]. These proteins are in turn involved in cell signaling, growth, and survival. The immunosuppression may reduce the hyperactive immune state induced by COVID-19. Moreover, JAK inhibitors like baricitinib have a theoretical direct antiviral activity via interference with viral endocytosis. This can prevent viral entry in the cells [93]. NIH has recommended that in the rare circumstances where corticosteroids cannot be used, baricitinib in combination with remdesivir may be used for the treatment of hospitalized, non-intubated patients requiring oxygen supplementation. IDSA guidelines also suggest the use of this combination in hospitalized severe COVID-19 patients [47]. Use of baricitinib without remdesivir is not recommended, except in a clinical trial [94]. As for the use of baricitinib in combination with corticosteroids, there is still insufficient data. Both baricitinib and corticosteroids cause immuno-suppression; hence, there is an additive risk of infection.

2.15 Other miscellaneous drugs with a possible therapeutic effect

In the pathogenesis of Covid-19, ACE 2 receptors play an important role by facilitating the entry of the virus into the cell [1, 95]. Thus it could be a possible therapeutic target with the use of ACE-inhibitors and ARB [1, 96]. However, there is a concern that the use of these drugs to stop virus replication may increase the expression of ACE-2 receptors and paradoxically worsen the infection. However, no in-vitro studies are available which show either definite detrimental or protective effect of these agents. As a result, the current guidelines state to continue these drugs in patients who are already taking them [97].

Umifenovir (also known as Arbidol) is an antiviral agent with a unique mechanism of action targeting the S protein/ACE2 interaction and inhibiting membrane fusion of the viral envelope [98]. It is approved in Russia for prophylaxis and treatment of influenza. Of particular interest is its demonstrable in-vitro activity against Covid-19 [99]. In an observational study in China, patients treated with umifenovir for a median duration of 9 days had a higher discharge rate and lesser mortality [100]. But as with other agents, the lack of RCT limits the justification for its use in Covid-19. However, ACE targeting therapy is a promising one [1].

Camostat mesylate is an agent used in the treatment of pancreatitis. It inhibits host serine protease, TMPRSS2.3, and has been shown to prevent viral cell entry in-vitro and thus could be a target for future studies [101].

Nitazoxanide, an anti-helminthic with a relatively favorable safety profile has shown in-vitro activity against SARS-CoV and MERS [102]. Besides it also has additional immunomodulatory action & thus can be used in trials in Covid-19 patients as a therapeutic option.

Many non-allopathic pharmaceuticals are also in pipeline as promising COVID-19 therapy. In June 2020, yoga guru Baba Ramdev announced that his company Patanjali Ayurved had launched a drug called ‘Coronil’ that could cure COVID-19 [103]. However, no scientific basis for this claim is produced until now.

Advertisement

3. Conclusion

The Global pandemic with COVID-19 is on. Drug therapy holds the key to the treatment and containment of the disease. Hence, large-scale multicentric trials are ongoing involving multiple drugs. Until now, no therapy is absolutely effective in the treatment of the patient as infection and death rates continue to mount all over the world. Corticosteroids have shown a significant effect on reducing the mortality in severe COVID-19 patients. It is hoped that the results of the ongoing trials will open further opportunities towards understanding the disease process and designing safe and effective treatments.

Advertisement

Conflict of interest

The authors declare no conflict of interest.

References

  1. 1. Kapoor M, Dhar M. A look into Possible New Treatment Modality for COVID-19: ACE 2 [Internet]. 2020. Available from: https://www.omicsonline.org/open-access/a-look-into-possible-new-treatment-modality-for-covid-19-ace-2.pdf
  2. 2. Zumla A, Chan JFW, Azhar EI, Hui DSC, Yuen KY. Coronaviruses-drug discovery and therapeutic options. Nat Rev Drug Discov. 2016;15(5):327-47.
  3. 3. Yan Y, Zou Z, Sun Y, Li X, Xu KF, Wei Y, et al. Anti-malaria drug chloroquine is highly effective in treating avian influenza A H5N1 virus infection in an animal model. Cell Res. 2013;23(2):300-2.
  4. 4. Vincent MJ, Bergeron E, Benjannet S, Erickson BR, Rollin PE, Ksiazek TG, et al. Chloroquine is a potent inhibitor of SARS coronavirus infection and spread. Virol J. 2005;2:1-10.
  5. 5. Keyaerts E, Vijgen L, Maes P, Neyts J, Ranst M Van. In vitro inhibition of severe acute respiratory syndrome coronavirus by chloroquine. Biochem Biophys Res Commun. 2004 Oct 8;323(1):264-8.
  6. 6. Wang M, Cao R, Zhang L, Yang X, Liu J, Xu M, et al. Remdesivir and chloroquine effectively inhibit the recently emerged novel coronavirus (2019-nCoV) in vitro. Cell Res. 2020;30(3):269-71.
  7. 7. Colson P, Rolain JM, Raoult D. Chloroquine for the 2019 novel coronavirus SARS-CoV-2 [Internet]. Vol. 55, International Journal of Antimicrobial Agents. Elsevier B.V.; 2020 [cited 2020 Nov 20]. p. 105923. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7134866/
  8. 8. Liu J, Cao R, Xu M, Wang X, Zhang H, Hu H, et al. Hydroxychloroquine, a less toxic derivative of chloroquine, is effective in inhibiting SARS-CoV-2 infection in vitro. Cell Discov [Internet]. 2020;6(1):6-9. Available from: http://dx.doi.org/10.1038/s41421-020-0156-0
  9. 9. Shukla AM, Archibald LK, Shukla AW, Mehta HJ, Cherabuddi K. Chloroquine and hydroxychloroquine in the context of COVID-19 [Internet]. Vol. 9, Drugs in Context. Bioexcel Publishing LTD; 2020 [cited 2020 Nov 20]. Available from: /pmc/articles/PMC7192209/?report=abstract
  10. 10. Gao J, Tian Z, Yang X. Breakthrough: Chloroquine phosphate has shown apparent efficacy in treatment of COVID-19 associated pneumonia in clinical studies [Internet]. Vol. 14, BioScience Trends. International Advancement Center for Medicine and Health Research Co., Ltd.; 2020 [cited 2020 Nov 20]. p. 72-3. Available from: www.biosciencetrends.com
  11. 11. Gautret P, Lagier JC, Parola P, Hoang VT, Meddeb L, Mailhe M, et al. Hydroxychloroquine and azithromycin as a treatment of COVID-19: results of an open-label non-randomized clinical trial. Int J Antimicrob Agents [Internet]. 2020 Jul 1 [cited 2021 Mar 4];56(1). Available from: https://pubmed.ncbi.nlm.nih.gov/32205204/
  12. 12. Coronavirus disease (COVID-19): Hydroxychloroquine [Internet]. [cited 2020 Nov 26]. Available from: https://www.who.int/news-room/q-a-detail/coronavirus-disease-covid-19-hydroxychloroquine#
  13. 13. FDA cautions against use of hydroxychloroquine or chloroquine for COVID-19 outside of the hospital setting or a clinical trial due to risk of heart rhythm problems | FDA [Internet]. [cited 2020 Nov 26]. Available from: https://www.fda.gov/drugs/drug-safety-and-availability/fda-cautions-against-use-hydroxychloroquine-or-chloroquine-covid-19-outside-hospital-setting-or
  14. 14. Nutho B, Mahalapbutr P, Hengphasatporn K, Pattaranggoon NC, Simanon N, Shigeta Y, et al. Why are lopinavir and ritonavir effective against the newly emerged coronavirus 2019? Atomistic insights into the inhibitory mechanisms. Biochemistry [Internet]. 2020 [cited 2020 Nov 20];59(18):1769-79. Available from: https://dx.doi.org/10.1021/acs.biochem.0c00160
  15. 15. Chu CM, Cheng VCC, Hung IFN, Wong MML, Chan KH, Chan KS, et al. Role of lopinavir/ritonavir in the treatment of SARS: Initial virological and clinical findings. Thorax [Internet]. 2004 Mar 1 [cited 2020 Nov 20];59(3):252-6. Available from: www.thoraxjnl.com
  16. 16. De Wilde AH, Jochmans D, Posthuma CC, Zevenhoven-Dobbe JC, Van Nieuwkoop S, Bestebroer TM, et al. Screening of an FDA-approved compound library identifies four small-molecule inhibitors of Middle East respiratory syndrome coronavirus replication in cell culture. Antimicrob Agents Chemother [Internet]. 2014 Aug 1 [cited 2020 Nov 20];58(8):4875-84. Available from: http://dx.doi.org/10.1128
  17. 17. Kim UJ, Won EJ, Kee SJ, Jung SI, Jang HC. Combination therapy with lopinavir/ritonavir, ribavirin and interferon-a for Middle East respiratory syndrome. Antivir Ther. 2016;21(5):455-9.
  18. 18. Cao B, Wang Y, Wen D, Liu W, Wang J, Fan G, et al. A Trial of Lopinavir–Ritonavir in Adults Hospitalized with Severe Covid-19. N Engl J Med [Internet]. 2020 May 7 [cited 2020 Nov 20];382(19):1787-99. Available from: https://www.nejm.org/doi/full/10.1056/NEJMoa2001282
  19. 19. WHO discontinues hydroxychloroquine and lopinavir/ritonavir treatment arms for COVID-19 [Internet]. [cited 2020 Nov 26]. Available from: https://www.who.int/news/item/04-07-2020-who-discontinues-hydroxychloroquine-and-lopinavir-ritonavir-treatment-arms-for-covid-19
  20. 20. Naksuk N, Lazar S, Peeraphatdit TB. Cardiac safety of off-label COVID-19 drug therapy: a review and proposed monitoring protocol. Eur Hear journal Acute Cardiovasc care [Internet]. 2020 May 6 [cited 2020 Nov 20];9(3):215-21. Available from: http://journals.sagepub.com/doi/10.1177/2048872620922784
  21. 21. Retallack H, Di Lullo E, Arias C, Knopp KA, Laurie MT, Sandoval-Espinosa C, et al. Zika virus cell tropism in the developing human brain and inhibition by azithromycin. Proc Natl Acad Sci U S A [Internet]. 2016 Dec 13 [cited 2020 Nov 26];113(50):14408-13. Available from: www.pnas.org/cgi/doi/10.1073/pnas.1618029113
  22. 22. Schögler A, Kopf BS, Edwards MR, Johnston SL, Casaulta C, Kieninger E, et al. Novel antiviral properties of azithromycin in cystic fibrosis airway epithelial cells. Eur Respir J [Internet]. 2015 Feb 1 [cited 2020 Nov 26];45(2):428-39. Available from: http://ow.ly/BVw2U
  23. 23. Oldenburg CE, Doan T. Azithromycin for severe COVID-19 [Internet]. Vol. 396, The Lancet. Lancet Publishing Group; 2020 [cited 2020 Nov 26]. p. 936-7. Available from: https://doi.org/10.1056/NEJMoa2019014.
  24. 24. Cavalcanti AB, Zampieri FG, Rosa RG, Azevedo LCP, Veiga VC, Avezum A, et al. Hydroxychloroquine with or without Azithromycin in Mild-to-Moderate Covid-19. N Engl J Med [Internet]. 2020 Jul 23 [cited 2020 Nov 29]; Available from: https://www.nejm.org/doi/full/10.1056/nejmoa2019014
  25. 25. Laing R, Gillan V, Devaney E. Ivermectin - Old Drug, New Tricks?. Trends Parasitol. 2017;33(6):463-472. doi:10.1016/j.pt.2017.02.004
  26. 26. Pandey S, Pathak SK, Pandey A, Salunke AA, Chawla J, Sharma A, et al. Ivermectin in COVID-19: What do we know? [Internet]. Vol. 14, Diabetes and Metabolic Syndrome: Clinical Research and Reviews. Elsevier Ltd; 2020 [cited 2020 Nov 29]. p. 1921-2. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7521351/
  27. 27. Caly L, Druce JD, Catton MG, Jans DA, Wagstaff KM. The FDA-approved drug ivermectin inhibits the replication of SARS-CoV-2 in vitro. Antiviral Res [Internet]. 2020 Jun 1 [cited 2020 Nov 29];178:104787. Available from: /pmc/articles/PMC7129059/?report=abstract
  28. 28. Kumar Maurya D. A Combination of Ivermectin and Doxycycline Possibly Blocks the Viral Entry and Modulate the Innate Immune Response in COVID-19 Patients.
  29. 29. Behera P, Kumar Patro B, Kumar Singh A, Kumar RS, Kumar Pradhan S, Kumar Pentapati S, et al. Title: Role of ivermectin in the prevention of COVID-19 infection among healthcare workers in India: A matched case-control study. medRxiv [Internet]. 2020 Nov 3 [cited 2020 Nov 29];2020.10.29.20222661. Available from: https://doi.org/10.1101/2020.10.29.20222661
  30. 30. Alam MT, Murshed R, Bhiuyan E, Saber S, Alam RF, Robin RC. A Case Series of 100 COVID-19 Positive Patients Treated with Combination of Ivermectin and Doxycycline. J Bangladesh Coll Physicians Surg [Internet]. 2020 Jun 12 [cited 2020 Nov 29];10-5. Available from: https://doi.org/10.3329/jbcps.v38i0.47512
  31. 31. Rahman MA, Iqbal SA, Islam MA, Niaz MK, Hussain T, Siddiquee TH. Comparison of Viral Clearance between Ivermectin with Doxycycline and Hydroxychloroquine with Azithromycin in COVID-19 Patients. J Bangladesh Coll Physicians Surg [Internet]. 2020 Jun 12 [cited 2020 Nov 29];5-9. Available from: https://doi.org/10.3329/jbcps.v38i0.
  32. 32. Ivermectin | COVID-19 Treatment Guidelines [Internet]. [cited 2021 Feb 16]. Available from: https://www.covid19treatmentguidelines.nih.gov/antiviral-therapy/ivermectin/
  33. 33. Diao B, Wang C, Tan Y, Chen X, Liu Y, Ning L, et al. Reduction and Functional Exhaustion of T Cells in Patients With Coronavirus Disease 2019 (COVID-19). Front Immunol [Internet]. 2020 May 1 [cited 2020 Nov 29];11:827. Available from: https://www.frontiersin.org/article/10.3389/fimmu.2020.00827/full
  34. 34. Hardeland R. Melatonin and inflammation—Story of a double-edged blade [Internet]. Vol. 65, Journal of Pineal Research. Blackwell Publishing Ltd; 2018 [cited 2020 Nov 29]. Available from: https://onlinelibrary.wiley.com/doi/full/10.1111/jpi.12525
  35. 35. Zhang R, Wang X, Ni L, Di X, Ma B, Niu S, et al. COVID-19: Melatonin as a potential adjuvant treatment. Vol. 250, Life Sciences. Elsevier Inc.; 2020. p. 117583.
  36. 36. Reiter RJ, Abreu-Gonzalez P, Marik PE, Dominguez-Rodriguez A. Therapeutic Algorithm for Use of Melatonin in Patients With COVID-19. Front Med [Internet]. 2020 May 15 [cited 2020 Nov 29];7:226. Available from: https://www.frontiersin.org/article/10.3389/fmed.2020.00226/full
  37. 37. Acuña-Castroviejo D, Escames G, Figueira JC, de la Oliva P, Borobia AM, Acuña-Fernández C. Clinical trial to test the efficacy of melatonin in COVID-19. J Pineal Res. 2020;69(3):2-5.
  38. 38. Tchesnokov E, Feng J, Porter D, Götte M. Mechanism of Inhibition of Ebola Virus RNA-Dependent RNA Polymerase by Remdesivir. Viruses [Internet]. 2019 Apr 4 [cited 2020 Nov 20];11(4):326. Available from: https://www.mdpi.com/1999-4915/11/4/326
  39. 39. Cao Y chen, Deng Q xin, Dai S xue. Remdesivir for severe acute respiratory syndrome coronavirus 2 causing COVID-19: An evaluation of the evidence. Vol. 35, Travel Medicine and Infectious Disease. Elsevier USA; 2020. p. 101647.
  40. 40. Yethindra V. Role of GS-5734 (Remdesivir) in inhibiting SARS-CoV and MERS-CoV: The expected role of GS-5734 (remdesivir) in COVID-19 (2019-nCoV)-VYTR hypothesis. Int J Res Pharm Sci [Internet]. 2020 Mar 6 [cited 2020 Nov 20];11(Special Issue 1):1-6. Available from: https://doi.org/10.26452/ijrps.v11iSPL1.1973
  41. 41. Protocol: Specific Antiviral Therapy in the Clinical Management of Acute Respiratory Infection with SARS-CoV-2 (COVID-19) [Internet]. 2020 [cited 2020 Nov 20]. Available from: https://www.who.int/publications-detail/clinical-management-of-severe-acute-respiratory-
  42. 42. Wang Y, Zhang D, Du G, Du R, Zhao J, Jin Y, et al. Remdesivir in adults with severe COVID-19: a randomised, double-blind, placebo-controlled, multicentre trial. Lancet. 2020 May 16;395(10236):1569-78.
  43. 43. Beigel JH, Tomashek KM, Dodd LE, Mehta AK, Zingman BS, Kalil AC, et al. Remdesivir for the Treatment of Covid-19 — Final Report. N Engl J Med [Internet]. 2020 Nov 5 [cited 2020 Nov 29];383(19):1813-26. Available from: https://www.nejm.org/doi/full/10.1056/NEJMoa2007764
  44. 44. FDA. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/nda/2020/214787Orig1s000Sumr.pdf.
  45. 45. WHO Solidarity Trial Consortium, Pan H, Peto R, Henao-Restrepo A-M, Preziosi M-P, Sathiyamoorthy V, et al. Repurposed Antiviral Drugs for Covid-19 — Interim WHO Solidarity Trial Results. N Engl J Med [Internet]. 2020 Dec 2 [cited 2020 Dec 4];NEJMoa2023184. Available from: http://www.nejm.org/doi/10.1056/NEJMoa2023184
  46. 46. WHO recommends against the use of remdesivir in COVID-19 patients [Internet]. [cited 2020 Nov 29]. Available from: https://www.who.int/news-room/feature-stories/detail/who-recommends-against-the-use-of-remdesivir-in-covid-19-patients
  47. 47. Overview of IDSA COVID-19 Treatment Guidelines [Internet]. [cited 2021 Feb 16]. Available from: https://www.fda.gov/media/143603/download
  48. 48. Therapeutic Management | COVID-19 Treatment Guidelines [Internet]. [cited 2021 Feb 16]. Available from: https://www.covid19treatmentguidelines.nih.gov/therapeutic-management/
  49. 49. Li Y, Chen M, Cao H, Zhu Y, Zheng J, Zhou H. Extraordinary GU-rich single-strand RNA identified from SARS coronavirus contributes an excessive innate immune response. Microbes Infect. 2013 Feb 1;15(2):88-95.
  50. 50. Huang C, Wang Y, Li X, Ren L, Zhao J, Hu Y, et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet. 2020 Feb 15;395(10223):497-506.
  51. 51. Hermine O, Mariette X, Tharaux PL, Resche-Rigon M, Porcher R, Ravaud P. Effect of Tocilizumab vs Usual Care in Adults Hospitalized with COVID-19 and Moderate or Severe Pneumonia: A Randomized Clinical Trial. JAMA Intern Med [Internet]. 2020 [cited 2020 Nov 29]; Available from: https://jamanetwork.com/
  52. 52. Guaraldi G, Meschiari M, Cozzi-Lepri A, Milic J, Tonelli R, Menozzi M, et al. Tocilizumab in patients with severe COVID-19: a retrospective cohort study. Lancet Rheumatol. 2020 Aug 1;2(8):e474-84.
  53. 53. Stone JH, Frigault MJ, Serling-Boyd NJ, Fernandes AD, Harvey L, Foulkes AS, et al. Efficacy of Tocilizumab in Patients Hospitalized with Covid-19. N Engl J Med [Internet]. 2020 Oct 21 [cited 2020 Nov 29]; Available from: https://www.nejm.org/doi/full/10.1056/NEJMoa2028836
  54. 54. Sanofi and Regeneron begin global Kevzara® (sarilumab) clinical trial program in patients with severe COVID-19 - Mar 16, 2020 [Internet]. [cited 2020 Nov 20]. Available from: http://www.news.sanofi.us/2020-03-16-Sanofi-and-Regeneron-begin-global-Kevzara-R-sarilumab-clinical-trial-program-in-patients-with-severe-COVID-19
  55. 55. Della-Torre E, Campochiaro C, Cavalli G, De Luca G, Napolitano A, La Marca S, et al. Interleukin-6 blockade with sarilumab in severe COVID-19 pneumonia with systemic hyperinflammation: an open-label cohort study. Ann Rheum Dis [Internet]. 2020 [cited 2020 Nov 29];79:1277-85. Available from: http://dx.doi.org/10.1136/annrheumdis-2020-218122
  56. 56. Statement on Tocilizumab | COVID-19 Treatment Guidelines [Internet]. [cited 2021 Feb 16]. Available from: https://www.covid19treatmentguidelines.nih.gov/statement-on-tocilizumab/
  57. 57. Shen C, Wang Z, Zhao F, Yang Y, Li J, Yuan J, et al. Treatment of 5 Critically Ill Patients with COVID-19 with Convalescent Plasma. JAMA - J Am Med Assoc [Internet]. 2020 Apr 28 [cited 2020 Nov 20];323(16):1582-9. Available from: https://jamanetwork.com/
  58. 58. Kadlec RP. Convalescent Plasma COVID-19 Letter of Authorization [Internet]. 2020 [cited 2020 Nov 29]. Available from: https://www.govinfo.gov/content/pkg/FR-2020-04-01/pdf/2020-06905.pdf.
  59. 59. Agarwal A, Mukherjee A, Kumar G, Chatterjee P, Bhatnagar T, Malhotra P. Convalescent plasma in the management of moderate covid-19 in adults in India: Open label phase II multicentre randomised controlled trial (PLACID Trial). BMJ [Internet]. 2020 Oct 22 [cited 2020 Nov 29];371. Available from: https://www.bmj.com/content/371/bmj.m3939
  60. 60. Simonovich VA, Burgos Pratx LD, Scibona P, Beruto M V., Vallone MG, Vázquez C, et al. A Randomized Trial of Convalescent Plasma in Covid-19 Severe Pneumonia. N Engl J Med [Internet]. 2020 Nov 24 [cited 2020 Dec 4];NEJMoa2031304. Available from: http://www.nejm.org/doi/10.1056/NEJMoa2031304
  61. 61. Furuta Y, Komeno T, Nakamura T. Favipiravir (T-705), a broad spectrum inhibitor of viral RNA polymerase. Proc Japan Acad Ser B [Internet]. 2017 Aug 2 [cited 2020 Nov 20];93(7):449-63. Available from: https://www.jstage.jst.go.jp/article/pjab/93/7/93_PJA9307B-02/_article
  62. 62. Cai Q , Yang M, Liu D, Chen J, Shu D, Xia J, et al. Experimental Treatment with Favipiravir for COVID-19: An Open-Label Control Study. Engineering. 2020 Mar 18;
  63. 63. Chen C, Zhang Y, Huang J, Yin P, Cheng Z, Wu J, et al. Favipiravir versus Arbidol for COVID-19: A Randomized Clinical Trial. [cited 2020 Nov 20]; Available from: https://doi.org/10.1101/2020.03.17.20037432
  64. 64. Arabi YM, Shalhoub S, Mandourah Y, Al-Hameed F, Al-Omari A, Qasim E Al, et al. Ribavirin and Interferon Therapy for Critically Ill Patients with Middle East Respiratory Syndrome: A Multicenter Observational Study. Clin Infect Dis [Internet]. 2020 May 1 [cited 2020 Nov 20];70(9):1837-44. Available from: https://academic.oup.com/cid/article/70/9/1837/5523209
  65. 65. Stockman LJ, Bellamy R, Garner P. SARS: Systematic Review of Treatment Effects. Low D, editor. PLoS Med [Internet]. 2006 Sep 12 [cited 2020 Nov 20];3(9):e343. Available from: https://dx.plos.org/10.1371/journal.pmed.0030343
  66. 66. Altınbas S, Holmes JA, Altınbas A. Hepatitis C Virus Infection in Pregnancy. Gastroenterol Nurs [Internet]. 2020 Jan 1 [cited 2020 Nov 20];43(1):12-21. Available from: http://journals.lww.com/10.1097/SGA.0000000000000404
  67. 67. Tong S, Su Y, Yu Y, Wu C, Chen J, Wang S, et al. Ribavirin therapy for severe COVID-19: a retrospective cohort study. Int J Antimicrob Agents. 2020 Sep 1;56(3):106114.
  68. 68. Panda PK, Bandyopadhyay A, Singh BC, Moirangthem B, Chikara G, Saha S, et al. Safety and efficacy of antiviral combination therapy in symptomatic patients of Covid-19 infection - a randomised controlled trial (SEV-COVID Trial): A structured summary of a study protocol for a randomized controlled trial [Internet]. Vol. 21, Trials. BioMed Central Ltd; 2020 [cited 2021 Mar 4]. Available from: https://pubmed.ncbi.nlm.nih.gov/33081849/
  69. 69. Totura AL, Bavari S. Broad-spectrum coronavirus antiviral drug discovery. Expert Opin Drug Discov [Internet]. 2019 Apr 3 [cited 2020 Nov 20];14(4):397-412. Available from: https://www.tandfonline.com/doi/full/10.1080/17460441.2019.1581171
  70. 70. Hung IFN, Lung KC, Tso EYK, Liu R, Chung TWH, Chu MY, et al. Triple combination of interferon beta-1b, lopinavir–ritonavir, and ribavirin in the treatment of patients admitted to hospital with COVID-19: an open-label, randomised, phase 2 trial. Lancet. 2020 May 30;395(10238):1695-704.
  71. 71. Davoudi-Monfared E, Rahmani H, Khalili H, Hajiabdolbaghi M, Salehi M, Abbasian L, et al. A randomized clinical trial of the efficacy and safety of interferon β-1a in treatment of severe COVID-19. Antimicrob Agents Chemother [Internet]. 2020 Sep 1 [cited 2020 Nov 29];64(9). Available from: https://doi.org/10.1128/AAC
  72. 72. Rahmani H, Davoudi-Monfared E, Nourian A, Khalili H, Hajizadeh N, Jalalabadi NZ, et al. Interferon β-1b in treatment of severe COVID-19: A randomized clinical trial. Int Immunopharmacol. 2020 Nov 1;88:106903.
  73. 73. Peiffer-Smadja N, Yazdanpanah Y. Nebulised interferon beta-1a for patients with COVID-19. Lancet Respir Med [Internet]. 2020 Nov 12 [cited 2020 Nov 30];0(0). Available from: http://www.ncbi.nlm.nih.gov/pubmed/33189160
  74. 74. Solidarity clinical trial for COVID-19 treatments [Internet]. [cited 2021 Feb 16]. Available from: https://www.who.int/emergencies/diseases/novel-coronavirus-2019/global-research-on-novel-coronavirus-2019-ncov/solidarity-clinical-trial-for-covid-19-treatments
  75. 75. Mehta P, McAuley DF, Brown M, Sanchez E, Tattersall RS, Manson JJ. COVID-19: consider cytokine storm syndromes and immunosuppression [Internet]. Vol. 395, The Lancet. Lancet Publishing Group; 2020 [cited 2020 Nov 20]. p. 1033-4. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7270045/
  76. 76. Arabi YM, Mandourah Y, Al-Hameed F, Sindi AA, Almekhlafi GA, Hussein MA, et al. Corticosteroid therapy for critically ill patients with middle east respiratory syndrome. Am J Respir Crit Care Med [Internet]. 2018 Mar 15 [cited 2020 Nov 20];197(6):757-67. Available from: http://www.atsjournals.org/doi/10.1164/rccm.201706-1172OC
  77. 77. Wu C, Chen X, Cai Y, Xia J, Zhou X, Xu S, et al. Risk Factors Associated With Acute Respiratory Distress Syndrome and Death in Patients With Coronavirus Disease 2019 Pneumonia in Wuhan, China. JAMA Intern Med [Internet]. 2020 Jul 1 [cited 2020 Nov 20];180(7):934. Available from: https://jamanetwork.com/journals/jamainternalmedicine/fullarticle/2763184
  78. 78. Wang Y, Jiang W, He Q , Wang C, Wang B, Zhou P, et al. Early, low-dose and short-term application of corticosteroid treatment in patients with severe COVID-19 pneumonia: single-center experience from Wuhan, China. medRxiv [Internet]. 2020 Mar 12 [cited 2020 Nov 20];2020.03.06.20032342. Available from: https://doi.org/10.1101/2020.03.06.20032342
  79. 79. Dexamethasone in Hospitalized Patients with Covid-19 — Preliminary Report. N Engl J Med [Internet]. 2020 Jul 17 [cited 2020 Nov 30]; Available from: https://www.nejm.org/doi/full/10.1056/NEJMoa2021436
  80. 80. Chen P, Nirula A, Heller B, Gottlieb RL, Boscia J, Morris J, et al. SARS-CoV-2 Neutralizing Antibody LY-CoV555 in Outpatients with Covid-19. N Engl J Med [Internet]. 2020 Oct 28 [cited 2020 Nov 29]; Available from: https://www.nejm.org/doi/full/10.1056/NEJMoa2029849
  81. 81. Jones BE, Brown-Augsburger PL, Corbett KS, Westendorf K, Davies J, Cujec TP, et al. Title: LY-CoV555, a rapidly isolated potent neutralizing antibody, provides protection in a non-human primate model of SARS-CoV-2 infection. bioRxiv [Internet]. 2020 Oct 9 [cited 2020 Nov 29];2020.09.30.318972. Available from: https://doi.org/10.1101/2020.09.30.318972
  82. 82. Coronavirus (COVID-19) Update: FDA Authorizes Monoclonal Antibody for Treatment of COVID-19 | FDA [Internet]. [cited 2020 Nov 29]. Available from: https://www.fda.gov/news-events/press-announcements/coronavirus-covid-19-update-fda-authorizes-monoclonal-antibody-treatment-covid-19
  83. 83. Mahase E. Covid-19: FDA authorises neutralising antibody bamlanivimab for non-admitted patients. BMJ [Internet]. 2020 Nov 11 [cited 2020 Nov 29];371:m4362. Available from: https://www.fda.gov/media/143602/download.
  84. 84. Lilly Statement Regarding NIH’s ACTIV-3 Clinical Trial | Eli Lilly and Company [Internet]. [cited 2020 Nov 29]. Available from: https://www.lilly.com/news/stories/statement-activ3-clinical-trial-nih-covid19
  85. 85. Coronavirus (COVID-19) Update: FDA Authorizes Monoclonal Antibodies for Treatment of COVID-19 | FDA [Internet]. [cited 2020 Nov 29]. Available from: https://www.fda.gov/news-events/press-announcements/coronavirus-covid-19-update-fda-authorizes-monoclonal-antibodies-treatment-covid-19
  86. 86. Regeneron halts trial of antibody treatment in seriously ill Covid patients | Financial Times [Internet]. [cited 2020 Nov 29]. Available from: https://www.ft.com/content/42256a8d-0073-4f57-9ac4-d3cc65a8e5c0
  87. 87. President Trump Received Regeneron Experimental Antibody Treatment - The New York Times [Internet]. [cited 2020 Nov 29]. Available from: https://www.nytimes.com/2020/10/02/health/trump-antibody-treatment.html
  88. 88. COVID-19 Long-Acting AntiBody (LAAB) combination AZD7442 rapidly advances into Phase III clinical trials | PharmaShots [Internet]. [cited 2020 Nov 29]. Available from: https://pharmashots.com/press-releases/covid-19-long-acting-antibody-laab-combination-azd7442-rapidly-advances-into-phase-iii-clinical-trials/
  89. 89. Coronavirus (COVID-19) Update: FDA Authorizes Monoclonal Antibodies for Treatment of COVID-19 | FDA [Internet]. [cited 2021 Feb 16]. Available from: https://www.fda.gov/news-events/press-announcements/coronavirus-covid-19-update-fda-authorizes-monoclonal-antibodies-treatment-covid-19-0
  90. 90. Zhang W, Zhao Y, Zhang F, Wang Q , Li T, Liu Z, et al. The use of anti-inflammatory drugs in the treatment of people with severe coronavirus disease 2019 (COVID-19): The experience of clinical immunologists from China [Internet]. Vol. 214, Clinical Immunology. Academic Press Inc.; 2020 [cited 2021 Feb 16]. Available from: https://pubmed.ncbi.nlm.nih.gov/32222466/
  91. 91. Babon JJ, Lucet IS, Murphy JM, Nicola NA, Varghese LN. The molecular regulation of Janus kinase (JAK) activation [Internet]. Vol. 462, Biochemical Journal. Portland Press Ltd; 2014 [cited 2021 Feb 16]. p. 1-13. Available from: https://pubmed.ncbi.nlm.nih.gov/25057888/
  92. 92. Bousoik E, Montazeri Aliabadi H. “Do We Know Jack” About JAK? A Closer Look at JAK/STAT Signaling Pathway [Internet]. Vol. 8, Frontiers in Oncology. Frontiers Media S.A.; 2018 [cited 2021 Feb 16]. Available from: https://pubmed.ncbi.nlm.nih.gov/30109213/
  93. 93. Stebbing J, Phelan A, Griffin I, Tucker C, Oechsle O, Smith D, et al. COVID-19: combining antiviral and anti-inflammatory treatments [Internet]. Vol. 20, The Lancet Infectious Diseases. Lancet Publishing Group; 2020 [cited 2021 Feb 16]. p. 400-2. Available from: https://pubmed.ncbi.nlm.nih.gov/32113509/
  94. 94. Kinase Inhibitors | COVID-19 Treatment Guidelines [Internet]. [cited 2021 Feb 16]. Available from: https://www.covid19treatmentguidelines.nih.gov/immunomodulators/kinase-inhibitors/
  95. 95. Chen Y, Liu Q , Guo D. Emerging coronaviruses: Genome structure, replication, and pathogenesis [Internet]. Vol. 92, Journal of Medical Virology. John Wiley and Sons Inc.; 2020 [cited 2020 Nov 20]. p. 418-23. Available from: https://onlinelibrary.wiley.com/doi/full/10.1002/jmv.25681
  96. 96. Gurwitz D. Angiotensin receptor blockers as tentative SARS-CoV-2 therapeutics [Internet]. Vol. 81, Drug Development Research. Wiley-Liss Inc.; 2020 [cited 2020 Nov 20]. p. 537-40. Available from: https://onlinelibrary.wiley.com/doi/full/10.1002/ddr.21656
  97. 97. Wang W, Zhao X, Wei W. Angiotensin-converting enzyme inhibitors (ACEI) or Angiotensin receptor blockers (ARBs) may be safe for COVID-19 patients. [cited 2020 Nov 20]; Available from: https://doi.org/10.21203/rs.3.rs-51043/v2
  98. 98. Kadam RU, Wilson IA. Structural basis of influenza virus fusion inhibition by the antiviral drug Arbidol. Proc Natl Acad Sci U S A [Internet]. 2017 Jan 10 [cited 2020 Nov 20];114(2):206-14. Available from: www.pnas.org/cgi/doi/10.1073/pnas.1617020114
  99. 99. Khamitov RA, Loginova SY, Shchukina VN, Borisevich S V., Maksimov VA, Shuster AM. Antiviral activity of arbidol and its derivatives against the pathogen of severe acute respiratory syndrome in the cell cultures. Vopr Virusol [Internet]. 2008 Jul 1 [cited 2020 Nov 20];53(4):9-13. Available from: https://europepmc.org/article/med/18756809
  100. 100. Wang Z, Yang B, Li Q , Wen L, Zhang R. Clinical features of 69 cases with coronavirus disease 2019 in Wuhan, China. Clin Infect Dis [Internet]. 2020 Aug 1 [cited 2020 Nov 20];71(15):769-77. Available from: https://academic.oup.com/cid/article/71/15/769/5807944
  101. 101. Hoffmann M, Kleine-Weber H, Schroeder S, Krüger N, Herrler T, Erichsen S, et al. SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor. Cell. 2020 Apr 16;181(2):271-280.e8.
  102. 102. Rossignol JF. Nitazoxanide, a new drug candidate for the treatment of Middle East respiratory syndrome coronavirus. J Infect Public Health. 2016 May 1;9(3):227-30.
  103. 103. Patanjali’s Coronil, 1st “proof-based” drug to fight Covid, gets govt nod | Business Standard News [Internet]. [cited 2021 Mar 4]. Available from: https://www.business-standard.com/article/current-affairs/ramdev-releases-paper-on-patanjali-s-1st-proof-based-covid-drug-coronil-coronavirus-treatment-121021900404_1.html

Written By

Mayank Kapoor, Prasan Kumar Panda and Vivek Mohanty

Submitted: 23 February 2021 Reviewed: 04 March 2021 Published: 17 May 2021