Int J Pharm Pharm Sci, Vol 14, Issue 11, 1-12Review Article

COMPREHENSIVE THERAPEUTIC INTERVENTIONS AGAINST SARS-COV-2: A REVIEW AND PROSPECTIVE

NILANJANA PRASAD*, DEBANJANA PRASAD

*Department of Biotechnology and Microbiology, Noida International University, Greater Noida 203201, Uttar Pradesh, India
*Email: nilu1545@gmail.com

Received: 27 Aug 2022, Revised and Accepted: 24 Sep 2022


ABSTRACT

In December 2019, Wuhan City, Hubei Province, China, first reported pneumonia like symptoms with unknown aetiology caused by a novel coronavirus. The novel coronavirus was renamed as Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) by Coronaviridae Study Group of the International Committee on Taxonomy of Viruses and the disease was termed as Coronavirus Disease 2019 (COVID-19). As of 19 August, 2022, the infection has reached above 220 countries, areas or territories with a total of 591 683 619 confirmed cases and 6 443 306 deaths, as published by the World Health Organization (WHO). SARS-CoV-2 is strongly contagious as it has R0, 2.2-2.6, in comparison to SARS-CoV (<1) and Middle East respiratory syndrome coronavirus (MERS-CoV) (1.4-2.5), respectively. SARS-CoV-2 might become less virulent than the SARS-CoV and MERS-CoV, with the currently analyzed mortality of COVID-19 is 3.4%. The original SARS-CoV-2 has undergone “virus evolution” with the occurrence of numerous variants such as Alpha, Beta, Gamma and Delta etc. Recently, the circulating variant of concern is Omicron subvariants. Currently, real-time reverse transcription–polymerase chain reaction-based detection of the viral genome (RNA) is the gold standard for diagnosis of SARS-CoV-2 infection. At present, Remdesivir (RDV) and Baricitinib drugs as well as vaccines Pfizer-BioNTech and Moderna have been approved for the treatment of COVID-19 by Food and Drug Administration (FDA). In this review, we summarized the existing state of knowledge on approved antiviral therapy, combination therapy, blood-derived therapeutics and immunomodulators to treat COVID-19 pandemic.

Keywords: COVID-19, SARS-CoV-2, Coronavirus, FDA, Convalescent plasma, Virus evolution


INTRODUCTION

The novel coronavirus, SARS-CoV-2 belonged to beta-coronavirus class, family Coronaviridae and order Nidovirales [1, 2]. SARS-CoV-2 has been hypothesized to originate from bat CoV, RaTG13 (96% whole genome level). Pangolin-CoV was hypothesized to be the second closest relative of SARS-CoV-2 (91.02% whole genome level) [3, 4]. Poor proofreading capability of the viral RNA polymerase and homologous recombination between CoVs might be the probable reasons of fast mutational frequency that might contribute to “cross‐species” transmission [5, 6]. COVID 19 genome consisted of single-stranded RNA composed of about 30 kb nucleotides, 5’-untranslated region (UTR), a replicase complex (orf1ab) encoding non-structural proteins (nsps), a spike protein (S) gene, envelope protein (E) gene, a membrane protein (M) gene, a nucleocapsid protein (N) gene, 3’-UTR, and several unidentified non-structural open reading frames [7, 8]. The global trend of COVID-19 confirmed cases and associated deaths from January 04, 2021 to August 22, 2022 have been reported [9] as shown in fig. 1. The highest percentage of COVID-19 confirmed cases as per WHO region was reported in Europe while the least has been reported in Africa [10] given in fig. 2.

Fig. 1: Global trend of COVID-19 cases and associated deaths

Whole genome sequencing of WH-Human 1 coronavirus (WHCV) was performed [11]. It was reported that enhancement of the nuclear localization signals in the nucleocapsid protein and well-defined inserts in the spike glycoprotein that appeared to be linked with a high case fatality rate of CoVs as well as the zoonotic transmission to humans were the main genomic features that distinguished SARS-CoV-2 from less pathogenic coronaviruses [12]. Human angiotensin-converting enzyme 2 (hACE2) has been declared to be the functional receptor of SARS-CoV-2 [13].

COVID-19 incubation period was 14 d, with a median time of 4-5 d from exposure to onset of symptoms [14-16]. The symptoms of COVID-19 reported were: fever, cough, fatigue, shortness of breath or difficulty breathing, muscle or body aches, headache, loss of taste or smell, sore throat, congestion or runny nose, nausea or vomiting and diarrhoea [17, 18-22]. The clinical manifestations of COVID-19 ranged from asymptomatic to severe pneumonia with severe acute respiratory distress syndrome (ARDS), septic shock, and ultimately multiple organ dysfunction syndrome (MODS) [23].

Fig. 2: Situation by WHO region showing confirmed cases of COVID-19 till August 24, 2022

The primary mode of infection was human-to-human transmission which occurred via respiratory secretions [24], which is why it is necessary to keep a distance of more than two meters from a sick person. Other modes included infected inanimate objects and it has been shown that the virus remains alive on surfaces for possibly up to 9 d [25, 26]. Recently, blood group A was correlated with an increased risk, whereas blood group O was associated with a decreased risk, thus indicating that the ABO blood type is a biomarker for differential susceptibility of COVID-19 [27]. In COVID-19 Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway is activated by Type I inteferons (IFNs), which lead to an antiviral gene expression program [28]. The immunopathology of the lung might be the consequence of the “cytokine storms” which lead to pneumonitis, ARDS, respiratory failure, shock, multiple organ failure and potentially death. Systemic and controlled COVID-19 drug development processes are essential approaches to combat the current COVID-19 pandemic.

Source of information

For obtaining recent quantitative, factual data about SARS-CoV-2 effects on the worldwide population (cases and deaths), diverse organizations’ such as WHO, FDA etc. were explored in depth. In addition, various electronic bibliographic databases were thoroughly searched. For formatting and presentation of information, relevant review articles were referred. Key text words, such as “virus evolution”, “therapeutic” published till 2022 were utilized in MEDLINE. Furthermore, contemporary research articles of published studies with appropriate information on the SARS-CoV-2 pathogenesis and drug therapies targeted against SARS-CoV-2 were consulted. For filling in with the fresh knowledge, relevant books, conference proceedings and public health organization survey reports were collated based on the wider objective of the review. This was achieved by looking for databases, including Web of Science, SCOPUS, EMBASE, Pubmed, Publon, PMC, Swiss rot, Google searches. From this specified methodology, findings were identified and summarized in this review. Furthermore, additional relevant references were incorporated through searching the references cited by the studies performed on the present topic.

Potential therapeutics under evaluation for the treatment of COVID-19 infection

RDV

RDV is a broad-spectrum antiviral monophosphoramidate prodrug of adenosine analogue with potent in vitro antiviral activity against a heterogenous panel of RNA viruses [29-32]. Different studies were undertaken out using RDV treatment in COVID-19 patients which resulted in clinical improvement [33-37]. RDV is the primary therapeutic antiviral treatment with proven potent efficacy against COVID-19 in an animal study which reported significant clinical benefit, a reduction in pulmonary infiltrates, and a reduction in pulmonary pathology. It further suggested that RDV treatment in COVID-19 patients should be initiated as early during infection to prevent advancement to severe pneumonia and to achieve the maximum clinical benefit because another study reported that the efficacy of direct-acting antivirals (DAAs) against acute viral infections typically decreased with delay in initiation of treatment. Adaptive COVID-19 Treatment Trial (ACTT), sponsored by the National Institute of Allergy and Infectious Diseases (NIAID), part of the National Institutes of Health (NIH) in COVID-19 hospitalized adults reported that patients who received IV RDV had a shorter time to recovery than those who received placebo and survival benefit, with a 14 d mortality rate of 7.1 % for the RDV treated group versus 11.9 % for the placebo group; although, the difference in mortality was not statistically significant. RDV treatment along with concomitant use of lopinavir-ritonavir, IFNs, and corticosteroids in adult COVID-19 patients and reported that RDV was not associated with a difference in time to clinical improvement, 28 d mortality, or rate of viral clearance between the RDV and the placebo-treated patients [38].

Chloroquine (CQ)

CQ is a 9-aminoquinoline that was synthesized in 1934 as a potent substitute for natural quinine used against malaria and was widely used to treat various human diseases [39-42]. Antiviral efficacy of CQ against COVID-19 might be evident from its past antiviral inhibitor roles against SARS-CoV, HCoV-OC43, influenza A H5N1, HCoV-229E, and EBOV in various cell line [43-48]. CQ was probably the first antiviral drug utilized in China and abroad as the front-line treatment against COVID-19 infections [49]. The first human trial conducted with chloroquine against COVID-19 reported that CQ reduced symptom duration, increased pneumonia, promoted the radiological improvement and virus-negative seroconversion without any severe adverse effects [50]. Various studies demonstrated no antiviral benefits associated with CQ in COVID-19 [51, 52].

Hydroxychloroquine (HCQ)

HCQ, an analogue of CQ, was used for the treatment of autoimmune diseases, such as systemic lupus erythematous (SLE) and rheumatoid arthritis [53]. HCQ differed from CQ by the presence of a hydroxyl group at the end of the side chain: the N-ethyl substituent is β-hydroxylated-1 [54] which conferred it with lesser risk of retinal toxicity as compared to CQ [55]. HCQ offered an immense advantage of being employed in high doses for long periods with very good tolerance and had a lower potential for drug-drug interactions than CQ [56]. A study suggested that incubation time might influence the antiviral activity of the drug because it is possible that a longer incubation time might provide more time for the drug to accumulate in higher intracellular concentrations and ultimately exhibit a potent antiviral effect [57]. Another study reported that clinical outcomes with HCQ in COVID-19 patients were overall survival, survival without ARDS, weaning from oxygen, and discharge from the hospital [58]. Diverse studies reported no clinical benefit associated with HCQ treatment against SARS-CoV-2 [59-61].

Combination therapy

Hydroxychloroquine plus azithromycin (HCQ+AZM)

Azithromycin (AZM) is utilized to treat a variety of bacterial and viral infections [62-64]. Combination treatment of AZM with HCQ has been involved in mixed clinical outcomes in COVID-19 [65-67]. Few studies have reported no clinical benefits with combination treatment of AZM with HCQ in COVID-19 [68-70].

Lopinavir/Ritonavir (LPV/RTV)

LPV, a highly potent, selective [71] and peptidomimetic [72] inhibitor of HIV type 1 aspartate protease inhibitor, usually combined with RTV to elevate its plasma half-life through the suspension of CYP450 has been reported to exhibit in vitro anti-MERS-CoV and anti SARS-CoV activity [73-75]. Combination therapy of LPV/RTV with IFN-β against MERS-CoV reported clinical benefits [76]. Various studies suggested that the administration of LPV/RTV regimen during early COVID-19 infection was effective in alleviating the viral load and improving clinical benefits in patients with mild to moderate disease [77, 78]. Numerous studies reported no clinical benefit associated with LPV/RTV treatment against SARS-CoV-2 infection [79-81].

Darunavir/Cobicistat (DRV/c)

Prophylactic DRV/c approved for the treatment of HIV-1 is a protease inhibitor. DRV acted as an inhibitor of the dimerization and of the catalytic activity of the HIV-1 protease, whereas c inhibited CYP450 that increased DRV plasma concentrations [82] Of note, DRV should not be administered without a boosting agent (RTV or c) as previous studies reported that DRV alone resulted in subtherapeutic drug levels and was linked with a higher rate of adverse events [83]. Clinical data on the use of treatment of (DRV/c) on COVID-19 is limited.

Blood derived therapeutics

Convalescent plasma (CP)

CP therapy, immunotherapy to achieve immediate short-term immunization against infectious agents where the humoral antibody (Ab) retrieved from the recovered patient was administered to a susceptible patient used to neutralise the pathogen and eventually lead to its elimination from the blood circulation [84-86]. Treatment with CP dates back to the end of the 19th century, in treatment of diphtheria and tetanus patients [87] and over the past decades, CP transfusion has proved its specificity and effectiveness as a potential treatment in patients with MERS-CoV [88], H1N15 [89], SARS-CoV [90-92] and cancer therapy which could help ameliorate survival rates in patients whose condition deteriorated even with conventional treatment. Previous studies of SARS and severe influenza recommended transfusion of CP as early as possible because the production of endogenous IgM and IgG antibodies summitted at 2 w and 4 w after infection, respectively [93]. To employ convalescent serum administration for COVID-19 the following six conditions must be met: (i) availability of donors (ii) blood banking facilities (iii) availability of assays to detect virus in serum and to estimate viral neutralization; (iv) virology lab facilities (v) randomized clinical trials (vi) regulatory compliance, comprising institutional review board approval, which might vary depending on location [94].

Different studies reported positive clinical outcomes using CP treatment in COVID-19 patients with few or no adverse events [95-99]. Neutralizing antibodies responses to SARS-CoV-2 in COVID-19 patients and convalescent patients was analyzed and reported that COVID-19 patients were seropositivity to SARS-CoV-2 during early stages, an evident neutralizing antibody response was observed in convalescent patients, and reduced antibody levels in asymptomatic or mild patients than moderate or severe patients which were consistent with results from previous studies. Another recent Chinese descriptive study in Wuhan, conducted in 6 laboratories, confirmed COVID-19 patients who received the transfusion of ABO-compatible CP and the results showed alleviation of symptoms, ameliorating radiologic abnormalities and laboratory tests and no obvious adverse effects were observed. Of note, this study speculated that anti-SARS-CoV-2 IgM and IgG directly neutralized the virus, and the anti-inflammatory contents might prevent cytokine stores recent study reported no significant clinical benefit associated with CP Therapy in severe COVID-19 patients [100].

Non-SARS-CoV-2-specific intravenous immune globulin

Liquid preparations of human immunoglobulin (pH 4) contained IgG antibody against a broad spectrum of antiviral, bacterial or other pathogens which could rapidly elevate the IgG level in the blood, directly neutralized exogenous antigens, and regulated various immune functions [101]. IVIG has been extensively utilized in the treatment of serious bacterial and viral infections and sepsis with positive and negative clinical outcomes [102-105]. A study of IVIG treatment in COVID-19 patients reported decreased 28 d mortality, minimized inflammatory response, ameliorated organ functions (all p<0.05) and 60 d mortality decreased significantly by using IVIG at an early stage (admission≤7 d) and with high dose (>15 g/d). Another study reported IVIG was associated with significantly reduced hypoxia, duration of hospital stays and need of mechanical ventilation requirement in COVID-19 patients [106].

Mesenchymal stem cell (MSC) therapy

MSCs are multipotent adult stem cells with self-renewable property i.e.; they are divided and differentiated into multiple types of tissues. MSC therapy was superior to other treatments because of: (i) easily accessed and isolated from different tissues (ii) it simply expanded to clinical volume in a suitable period of time (iii) could be stored for repetitive clinical usage; (iv) clinical trials of MSCs till date hasn’t shown adverse effects to allogeneic MSC; (v) safety and effectiveness of MSCs in several clinical trials [107]. Leng and colleagues demonstrated the safety and efficacy of IV MSC transplantation in COVID-19 patients and reported improving pulmonary status, reduced C-reactive protein (CRP) levels, and lowered inflammatory cytokines whilst IL-10 escalated and hyperactivity cytokine secreting immune cells dissipated after a few days [108]. Treatment with human umbilical cord mesenchymal stem cells (hUCMSC) in a critically ill COVID-19 patient resulted in the return of all measured parameters to normal levels and suggested that hUCMSC could be an ideal therapeutic option alone or in parallel with other immune modulators for acute COVID-19 patients [109].

Immunomodulators

Corticosteroids

Corticosteroids are systemic anti-inflammatory and classical immunosuppressive drugs which terminated or delayed the progression of pneumonia and have been shown to be effective against the treatment of ARDS [110-112]. Corticosteroids were widely used during past CoV outbreaks [113-115], in addition to other therapeutics, which reported no significant effect on clinical outcomes but increased mortality and requirement of mechanical ventilation. It was reported that it was difficult to make a clear recommendation to corticosteroid treatment of SARS patients, particularly because the drug is immunosuppressive and might delay viral clearance if administered before viral replication is controlled. The RECOVERY (Randomised Evaluation of COVid-19 therapy) trial reported that low-cost dexamethasone at a dosage of 6 mg once daily for up to 10 d alleviated 28 d mortality in COVID-19 hospitalized patients with severe respiratory complications [116]. Different studies reported that early, low-dose and short-term application of intravenous methylprednisolone in severe COVID-19 patients was associated with a rapid improvement of clinical symptoms and absorption of lung focus with little or no serious complications [117-119]. A retrospective observational study of corticosteroids on outcomes of COVID-19 patients revealed that treatment within 72 h after the first signs could reasonably reduce the risk of disease exacerbation in non-severe patients which were consistent for other virus-induced diseases [120]. Several studies reported no effective clinical outcomes associated with corticosteroid treatment in COVID-19 patients but an increased risk of mortality and secondary infections [121-123].

IFNs

IFNs are cellular surfaces’ receptor binding proteins which initiated JAK-STAT signalling cascades, with transcriptional regulation of genes and effective against certain viruses like hepatitis B virus and HCV [124]. Several MERS-CoV studies were carried out using different combinations of IFNα and IFNβ with ribavirin some of which reported an association or no association with reduction in mortality [125-127]. Varied types of IFNs belonging to the three classes (α, β and γ) reported effective anti-SARS-CoV activities [128, 129]. A study was conducted in severe COVID-19 patients with subcutaneous administration of IFNβ-1a which reported escalating discharge rate on day 14 and alleviated 28 d mortality with improved survival during the early course of administration [130]. Treatment with nebulized IFN-α2b in adults hospitalized confirmed COVID-19 patients reported shortened duration of viral shedding and synchronously decreased the markers of acute inflammation such as CRP and IL-6 [131]. Triple combination treatment with INF β-1b, LPV/RTV, and ribavirin in COVID-19 patients reported that therapy was safe and superior to LPV/RTV alone in decreasing symptoms and reducing the duration of viral shedding and hospital stay [132]. An experimental open-label study suggested that recombinant human interferon alpha (rhIFN-α) nasal drops to be a promising agent for protecting COVID-19 susceptible healthy people [133]. Combination treatment of IFN-α2b and IFN-γ with the standard of care in confirmed COVID-19 patients was associated with rapid viral elimination, reduced CRP levels and improved clinical parameters [134].

Interlukin-1 (IL-1)-Anakinra

Anakinra, a 17-kD recombinant, non-glycosylated antagonist (IL-1Ra) that only differed from the known IL-1Ra due to the presence of an extra methionine residue at the N-terminal end [135, 136]. Continuous intravenous anakinra infusion in severely ill adult patients with macrophage activation syndrome (MAS) resulted in rapid serologic and subsequent clinical improvement [137]. Various studies reported positive clinical outcomes associated with anakinra treatment in COVID-19 patients with the occurrence of few adverse events [138-140].

IL-6 Inhibitors (Tocilizumab, Sarilumab, Siltuximab)

Tocilizumab (TCZ)

TCZ is a recombinant humanized anti-interleukin-6 receptor (IL-6R). TCZ treatment in moderate to severe COVID-19 patients was found to be effective and resulted in improved survival and reduced the risk of mortality [141-143].

Siltuximab

Siltuximab is a human-mouse chimeric IL-6R monoclonal antibody [144]. IV siltuximab treatment in COVID-19 patients with pneumonia requiring ventilatory support showed reduced serum CRP levels, and improved clinical condition with increased deterioration in few patients [145]. However, clinical data on the use of siltuximab in COVID-19 treatment is limited.

Sarilumab

Sarilumab is the recombinant human monoclonal antibody IL-6 receptor antagonist approved in various countries to treat RA in adults. Phase 2/3 adaptive-designed trial of Sarilumab in severe or critical COVID-19 patients reported rapidly reduced CRP levels as the primary endpoint and “negative trends” for most outcomes in severe patients [146]. Clinical data on the use of Sarilumab in COVID-19 treatment is limited.

Bruton’s tyrosine kinase inhibitors (BTK) inhibitors

Acalabrutinib

Acalabrutinib, a highly specific covalent BTK inhibitor authorized in the U. S for the treatment of lymphoid malignancies [147]. Roschewski and colleagues conducted an off-label study using acalabrutinib in severe COVID-19 patients and reported improving oxygenation and reduced levels of inflammatory markers (CRP and IL-6) with no discerning toxicity [148]. However, clinical data on the use of acalabrutinib in COVID-19 infection is limited.

Ibrutinib

Ibrutinib is a highly potent and covalent BTK inhibitor. A study conducted by American Society of Hematology (ASH) suggested that ibrutinib might provide protection against pulmonary injury in COVID-19 patients [149]. However, clinical data on the use of ibrutinib in COVID-19 infection is limited.

JAK inhibitors

Baricitinib

Baricitinib is an oral, reversible, selective and potent JAK inhibitor approved by the FDA to treat rheumatoid arthritis and could improve the chronic inflammation seen in interferonopathies [150-153]. It was reported that therapeutic dosage of baricitinib with either 2 mg or 4 mg once daily was adequate to reach the plasma level of inhibition. In addition, he suggested that this drug could be a potential treatment for COVID-19 acute respiratory disease [154]. Baricitinib therapy in COVID-19 patients reported significantly improved clinical characteristics and none of the patients required ICU support, and the majority of the patients were discharged [155].

Ruxolitinib

Ruxolitinib is (JAK)1/2 inhibitor that has shown JAK-STAT inhibition in COVID-19. Ruxolitinib treatment in COVID-19 patients reported escalating clinical improvement, improved chest CT images and rapid recovery from lymphopenia and demonstrated the efficacy and safety [156]. Another study of ruxolitinib treatment in COVID-19 patients with severe systemic hyperinflammation reported sustaining clinical improvement [157].

Comprehensive, detailed information about COVID-19 treatment options is given in table 1. Recently, RDV has been approved by FDA in hospitalized adults and pediatric patients (aged ≥12 y and weighing ≥40 kg [158]. BTK and JAK inhibitors demonstrated potential anti-SARS-CoV-2 activity but data is limited. Some of them are showing encouraging results with less or no side effects but still need to investigate in details and in spite of therapeutic interventions. Other strategies like blood-derived products, immuno-modulators etc. are also in different stages of clinical trials [159-163].

Table 1: List of therapeutic interventions for COVID-19 treatment

S. No. Drug Route of administration Mechanism of action in COVID-19 Adverse effects Reference
Antiviral therapy
1. RDV IVa
  • RNAbpolymerase inhibitor with in vitro and in vivo anti SARS-CoVc-2 activity at low µm concentration (EC50d = 0.77 μM; CC50>100 μM; SI>129.87).

  • Pyrexia

  • GIe symptoms

  • Multiple organ-dysfunction syndrome

  • Acute kidney injury

  • Hypotension

  • Mortality

[35, 37, 38]
2 CQ Oral­­
  • Increases endosomal pH levels and suspends virus endosome fusion.

  • In vitro (EC50=5.47 μM) and in vivo inhibitory activity against SARS-CoV-2 at low µm concentration (EC50 = 1.13 μM).

  • Cough

  • Shortness of breath

  • GI symptoms

  • Death

[35, 52, 57, 164]
3. HCQ Oral
  • Similar to the mechanism of action of CQf.

  • Potent anti-SARS-CoV-2 activity in vitro (EC50=0.72 μM).

  • GI symptoms

  • Cardiac arrhythmia

  • Chronic kidney failure

  • Diabetes

  • Death

[57, 58, 60, 165]
Combination therapy
4. HCQ+AZM Oral
  • Induction of IFNg-stimulated genes, attenuating viral replication.

  • Immunomodulatory effects

  • Anti-Inflammatory effects

  • GI symptoms

  • Cardiac arrest

  • Diabetes mellitus

  • Chronic kidney disease

  • Death

[66, 166, 167, 168]
5 HIV Protease Inhibitors (LPV/RTV and DRV/c) Oral
  • Possible inhibitory activity on SARS-CoV-2 proteases (3CLproh and (PLproi) required for replication.

  • In vitro SARS-CoV-2 effect of Lopinavir (EC50 at 26.1 μM) in Vero E6 cells.

  • Pyrexia

  • Cough

  • GI symptoms

  • Respiratory failure/ARDSj

  • Biochemical hepatitis

  • Mortality

[79, 81, 132, 169, 170]
Blood derived therapeutics
6. CP IV
  • Passive Antibody therapy.

  • TACOk

  • TRALIl

  • Shortness of breath

  • Respiratory failure

  • Cardiac events

  • Death

[95, 171]
7. Non-SARSCoV-2 Specific IVIGm IV
  • Direct neutralization of exogenous antigens and regulation of multiple immune functions by human immunoglobulin (pH4) containing IgG antibody.

  • Bacterial super infections

  • Thrombocytopenia

  • Lymphocytopenia

  • Mortality

[101-103, 106]
8. MSCs IV
  • Robust immunomodulatory and anti-inflammatory activity.

  • Promotion of endogenous repair and improvement of the pulmonary microenvironment and function.

  • No adverse effect observed in COVID-19n.

[108]
Immunomodulators
Corticosteroids
9. Dexamethasone

Oral

IV

  • Potent anti-inflammatory and immunosuppressive effects.

  • Detrimental effect on angiotensin II which played a key role in the pathophysiology of SARS-CoV-2.

  • Cardiac arrhythmia

  • Renal replacement

  • Hyperglycaemia

  • Barotrauma

  • Mortality

[21, 123, 172-177]
10. Methylprednisolone IV
  • Hyperglycaemia

  • ARDS

  • Mortality

11. Adjuvant corticosteroid therapy (Hydrocortisone) IV
  • Secondary infections

  • Multiple organ dysfunction

  • Mortality

IFNs (alpha, beta and gamma)
12 IFN-alpha

Inhalation

Intramuscular (IFN-α2b)

  • The EC50 of IFN-α in SARS-CoV-2 treatment in Vero cells is 1.35 IU/ml.

  • Reduction of the infection rate of SARS-CoV-2 by IFNα2b spray.

  • Down regulation of the inflammatory biomarker IL-6 °by IFN-α2b.

  • Pyrexia

  • Dry Cough

  • GI symptoms

  • Worsening of the respiratory system

[131]

[178] [179]

13. IFN-β

Subcutaneous injection (IFN-β1b)

IV injection (IFN-β1a)

  • The EC50 of IFN-β in SAR-CoV-2 treatment in Vero cells is 0.76 IU/ml.

  • Upregulation of CD73p, prevention of vascular leakage and inhibition of leukocyte recruitment by IFN-β-1a.

  • Pyrexia

  • GI symptoms

  • Injection site reactions

  • Renal impairment

  • Hepatic impairment

  • Mortality

[130]

[178]

[180]

[181]

[182]

14. IFN‐γ Subcutaneous
  • Immunomodulatory effects

  • Anti-inflammatory effects

  • Restricts ACE2qexpression.

  • GI symptoms

  • Worsening of respiratory symptoms

  • Headache

[134]

[183]

IL-1
15. Anakinra

IV

Subcutaneous

  • Blocks the binding of IL-1αr and IL-1βs to IL-1Rt by mirroring the mode of action of its endogenous counterpart.

  • Neutralization of SARS-CoV-2 hyperinflammatory state.

  • Septic shock

  • Bacteremia

  • Acute respiratory failure

  • Multiorgan failure

[136]

[137]

[184]

IL-6
16. TCZ

IV

Subcutaneous

  • Humanized anti IL-6Ru antibody which competitively inhibits both the soluble and membrane-bound forms of the IL-6R.

  • Suppression of cytokine storm in critically ill COVID-19 patients.

  • Infections

  • Liver failure

  • Injection site reactions.

  • Renal replacement

  • Death

[141]

[142]

[143]

[185]

17. Sarilumab Intravenous
  • IL-6R receptor antibody which inhibits IL-6-mediated signalling. 

  • Reduction of the hyperactive inflammatory immune response associated with COVID-19.

  • Hypotension

  • Multiple organ dysfunction

  • Mortality

[146]

[186]

18. Siltuximab Intravenous
  • Chimeric antibody against IL-6R that binds to and blocks the effect of IL-6.

  • Death

  • Cerebrovascular event

[145]
Kinase inhibitors: BTK inhibitors and JAK inhibitors
19. Acalabrutinib Oral
  • Macrophage signaling and activation in COVID-19.

  • Inhibition of the BTKv induced pathological hyperinflammatory response in severe COVID-19.

  • Pyrexia

  • Headache

  • Cardiac arrhythmias

  • Opportunistic infections

  • Upper respiratory tract infections

[148]
20 Ibrutinib Oral
  • Protects against pulmonary injury in COVID-19.

  • Serious bleeding

  • Proarrhythmic

  • Effect.

  • Dyspnea

  • Hypoxia

[149]
21 Baricitinib Oral
  • JAK-STATwsignalling inhibitor.

  • Interferes with receptor-mediated endocytosis and clathrin-mediated endocytosis.

  • Upper respiratory tract infections.

[18, 151, 187]
22 Ruxolitinib Oral
  • Targeted inhibition of cytokine signaling in COVID-19.

  • Reduction of systemic inflammation in COVID-19.

  • GI symptoms

  • Hematological events

  • Headache

  • Liver dysfunction

[156]

[157]

aIntravenous; bRibonucleic acid; cSevere acute respiratory syndrome; dHalf-maximal effective concentration; eGastrointestinal; fChloroquine; gInterferon; h3-chymotrypsin-like protease; iPapain-like protease; jAcute respiratory distress syndrome; lTransfusion-related acute lung injury; mIntravenous immunoglobulin; nCoronavirus disease 2019; °Interlukin-6; pCluster of differentiation 73; qAngiotensin-converting enzyme 2; rInterleukin-1 alpha; sInterlukin-1 beta; tInterleukin-1 receptor; uInterlukin-6 receptor; vBruton’s tyrosine kinase inhibitor; wJanus kinase inhibitor-signal transducer and activator of transcription protein.

CONCLUSION

RDV has shown to be the most promising agent having potential anti-SARS-CoV-2 activity. Blood-derived therapeutics such as CP and IVIG demonstrated mixed clinical outcomes against SARS-CoV-2 with possible side effects. MSCs emerged as a safe and effective treatment of COVID-19 because of its increased proliferation rate, lowered invasive procedure, free from ethical issue and potential clinical benefits with no mortality in COVID-19 disease. Gene expression profiling study of transplanted MSCs showed that MSCs are ACE2-or TMPRSS2-indicating that MSCs are free from SARS-CoV-2 infection. Potent candidates repurposed could save time and cost, especially when vaccine is not available commercially. There is a requirement to study future COVID-19 variants, including their geographical spread, genetic changes and phenotypic characteristics to design effective vaccines against them. There is a need to increase the availability of diagnostic tests, medical devices, personal protective equipment kit and therapeutics for an effective COVID-19 response. Social distancing and caution are the magical formulas nowadays.

ACKNOWLEDGEMENT

We thank the anonymous referees for their useful suggestions.

FUNDING

Nil

AUTHORS CONTRIBUTIONS

Nilanjana Prasad and Debanjana Prasad were responsible for the conception, experiments, and writing and revising the manuscript.

CONFLICT OF INTERESTS

Declared none

REFERENCES

  1. Qadrie ZL, Wani SUD, Gautam SP, Khan MKA. Outbreak, epidemiology, therapeutics and prevention of coronavirus disease-2019: a review. Int J Curr Pharm Sci. 2020;12(5):1-4. doi: 10.22159/ijcpr.2020v12i5.39755.

  2. Grifoni A, Sidney J, Zhang Y, Scheuermann RH, Peters B, Sette A. A sequence homology and bioinformatic approach can predict candidate targets for immune responses to SARS-CoV-2. Cell Host Microbe. 2020;27(4):671-680.e2. doi: 10.1016/j.chom.2020.03.002. PMID 32183941.

  3. MA SM, Hassan S. Assess the awareness and attitude regarding prevention of coronavirus disease 2019. Asian J Pharm Clin Res. 2020;2020:95-8. doi: 10.22159/ajpcr.2020.v13i7.37804.

  4. Zhou P, Yang XL, Wang XG, Hu B, Zhang L, Zhang W. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature. 2020;579(7798):270-3. doi: 10.1038/s41586-020-2012-7, PMID 32015507.

  5. Sukumaran S, Sathianarayanan S. A review on covid-19 pandemic a global threat-current status and challenges and preventive strategies. Int J App Pharm. 2021;13(5):10-4. doi: 10.22159/ijap.2021v13i5.42070.

  6. Ji W, Wang W, Zhao X, Zai J, Li X. Cross-species transmission of the newly identified coronavirus 2019-nCoV. J Med Virol. 2020;92(4):433-40. doi: 10.1002/jmv.25682, PMID 31967321.

  7. Zheng M, Song L. Novel antibody epitopes dominate the antigen city of spike glycoprotein in SARS-CoV-2 compared to SARS-CoV. Cell Mol Immunol. 2020;17(5):536-8. doi: 10.1038/s41423-020-0385-z, PMID 32132669.

  8. Nandagopal M, Balakrishnan A, Padhiar C. Trinity of severe acute respiratory syndrome Coronavirus-2: origin, genotype, phenotype, and immune response in coronavirus disease-201. Asian J Pharm Clin Res. 2021. v14i9.42363;2021;14(9):28-36. doi: 10.22159/ajpcr.

  9. World Health Organization. Coronavirus disease (COVID-19) dashboard; 2020.

  10. World Health Organization. Situation by WHO region; 2020.

  11. Wu F, Zhao S, Yu B, Chen YM, Wang W, Song ZG. A new coronavirus associated with human respiratory disease in China. Nature. 2020;579(7798):265-9. doi: 10.1038/s41586-020-2008-3, PMID 32015508.

  12. Gussow AB, Auslander N, Faure G, Wolf YI, Zhang F, Koonin EV. Genomic determinants of pathogenicity in SARS-CoV-2 and other human coronaviruses. Proc Natl Acad Sci USA. 2020;117(26):15193-9. doi: 10.1073/pnas.2008176117, PMID 32522874.

  13. Hoffmann M, Kleine Weber H, Schroeder S, Kruger N, Herrler T, Erichsen S. SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor. Cell. 2020;181(2):271-280.e8. doi: 10.1016/j.cell.2020.02.052, PMID 32142651.

  14. Guan WJ, Ni ZY, Hu Y, Liang WH, Ou CQ, He JX. Clinical characteristics of coronavirus disease 2019 in China. N Engl J Med. 2020;382(18):1708-20. doi: 10.1056/NEJMoa2002032, PMID 32109013.

  15. Li Q, Guan X, Wu P, Wang X, Zhou L, Tong Y. Early transmission dynamics in Wuhan, China, of novel coronavirus-infected pneumonia. N Engl J Med. 2020;382(13):1199-207. doi: 10.1056/NEJMoa2001316, PMID 31995857.

  16. Lauer SA, Grantz KH, Bi Q, Jones FK, Zheng Q, Meredith HR. The incubation period of coronavirus disease 2019 (COVID-19) from publicly reported confirmed cases: estimation and application. Ann Intern Med. 2020;172(9):577-82. doi: 10.7326/M20-0504, PMID 32150748.

  17. Wang D, Hu B, Hu C, Zhu F, Liu X, Zhang J. Clinical characteristics of 138 hospitalized patients with 2019 novel coronavirus–infected pneumonia in Wuhan, China. JAMA. 2020;323(11):1061-9. doi: 10.1001/jama.2020.1585, PMID 32031570.

  18. Huang C, Wang Y, Li X, Ren L, Zhao J, Hu Y. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet. 2020;395(10223):497-506. doi: 10.1016/S0140-6736(20)30183-5.

  19. Chen N, Zhou M, Dong X, Qu J, Gong F, Han Y. Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: a descriptive study. Lancet. 2020;395(10223):507-13. doi: 10.1016/S0140-6736(20)30211-7, PMID 32007143.

  20. Clinical findings in a group of patients infected with the 2019 novel coronavirus (SARS-Cov-2) outside of Wuhan, China: retrospective case series. BMJ. 2020;368:m792. doi: 10.1136/bmj.m606.

  21. Wu C, Chen X, Cai Y, Xia J, Zhou X, Xu S. Risk factors associated with acute respiratory distress syndrome and death in patients with coronavirus disease 2019 pneumonia in Wuhan, China. JAMA Intern Med. 2020;180(7):934-43. doi: 10.1001/jamainternmed.2020.0994, PMID 32167524.

  22. Pan L, Mu M, Yang P, Sun Y, Wang R, Yan J. Clinical characteristics of COVID-19 patients with digestive symptoms in Hubei, China: A descriptive, cross-sectional, multicenter study. Am J Gastroenterol. 2020;115(5):766-73. doi: 10.14309/ajg.0000000000000620, PMID 32287140.

  23. Yang X, Yu Y, Xu J, Shu H, Xia J, Liu H. Clinical course and outcomes of critically ill patients with SARS-CoV-2 pneumonia in Wuhan, China: a single-centered, retrospective, observational study. Lancet Respir Med. 2020;8(5):475-81. doi: 10.1016/S2213-2600(20)30079-5, PMID 32105632.

  24. Prompetchara E, Ketloy C, Palaga T. Immune responses in COVID-19 and potential vaccines: lessons learned from SARS and MERS epidemic. Asian Pac J Allergy Immunol. 2020;38(1):1-9. doi: 10.12932/AP-200220-0772, PMID 32105090.

  25. Phan LT, Nguyen TV, Luong QC, Nguyen TV, Nguyen HT, Le HQ. Importation and human-to-human transmission of a novel coronavirus in Vietnam. N Engl J Med. 2020;382(9):872-4. doi: 10.1056/NEJMc2001272, PMID 31991079.

  26. Riou J, Althaus CL. Pattern of early human-to-human transmission of Wuhan 2019 novel coronavirus (2019-nCoV), December 2019 to January 2020. Euro Surveill. 2020;25(4):2000058. doi: 10.2807/1560-7917.ES.2020.25.4.2000058, PMID 32019669.

  27. Zhao J, Yang Y, Huang H, Li D, Gu D, Lu X. Relationship between the ABO blood group and the coronavirus disease 2019 (COVID-19) susceptibility. Clin Infect Dis. 2021;73(2):328-31. doi: 10.1093/cid/ciaa1150, PMID 32750119.

  28. de Wit E, van Doremalen N, Falzarano D, Munster VJ. SARS and MERS: recent insights into emerging coronaviruses. Nat Rev Microbiol. 2016;14(8):523-34. doi: 10.1038/nrmicro.2016.81, PMID 27344959.

  29. Lo MK, Jordan R, Arvey A, Sudhamsu J, Shrivastava Ranjan P, Hotard AL. GS-5734 and its parent nucleoside analog inhibit Filo, Pneumo, and Paramyxoviruses. Sci Rep. 2017;7:43395. doi: 10.1038/srep43395, PMID 28262699.

  30. Sheahan TP, Sims AC, Graham RL, Menachery VD, Gralinski LE, Case JB. Broad-spectrum antiviral GS-5734 inhibits both epidemic and zoonotic coronaviruses. Sci Transl Med. 2017;9(396):eaal3653. doi: 10.1126/scitranslmed.aal3653. PMID 28659436.

  31. Warren TK, Jordan R, Lo MK, Ray AS, Mackman RL, Soloveva V. Therapeutic efficacy of the small molecule GS-5734 against Ebola virus in rhesus monkeys. Nature. 2016;531(7594):381-5. doi: 10.1038/nature17180, PMID 26934220.

  32. Sheahan TP, Sims AC, Leist SR, Schafer A, Won J, Brown AJ. Comparative therapeutic efficacy of remdesivir and combination lopinavir, ritonavir, and interferon beta against MERS-CoV. Nat Commun. 2020;11(1):222. doi: 10.1038/s41467-019-13940-6, PMID 31924756.

  33. Williamson BN, Feldmann F, Schwarz B, Meade White K, Porter DP, Schulz J. Clinical benefit of remdesivir in rhesus macaques infected with SARS-CoV-2. Nature. 2020;585(7824):273-6. doi: 10.1038/s41586-020-2423-5, PMID 32516797.

  34. Sheahan TP, Sims AC, Zhou S, Graham RL, Pruijssers AJ, Agostini ML. An orally bioavailable broad-spectrum antiviral inhibits SARS-CoV-2 in human airway epithelial cell cultures and multiple coronaviruses in mice. Sci Transl Med. 2020;12(541):eabb5883. doi: 10.1126/scitranslmed.abb5883. PMID 32253226.

  35. Piscoya A, Ng-Sueng LF, Parra del Riego A, Cerna Viacava R, Pasupuleti V, Roman YM. Efficacy and harms of remdesivir for the treatment of COVID-19: A systematic review and meta-analysis. PLOS ONE. 2020;15(12):e0243705. doi: 10.1371/journal.pone.0243705. PMID 33301514.

  36. Beigel JH, Tomashek KM, Dodd LE, Mehta AK, Zingman BS, Kalil AC. Remdesivir for the treatment of covid-19–final report. N Engl J Med. 2020;383(19):1813-26. doi: 10.1056/NEJMoa2007764. PMID 32445440.

  37. Grein J, Ohmagari N, Shin D, Diaz G, Asperges E, Castagna A. Compassionate use of remdesivir for patients with severe covid-19. N Engl J Med. 2020;382(24):2327-36. doi: 10.1056/NEJMoa2007016, PMID 32275812.

  38. Wang Y, Zhang D, Du G, Du R, Zhao J, Jin Y. Remdesivir in adults with severe COVID-19: a randomised, double-blind, placebo-controlled, multicentre trial. Lancet. 2020;395(10236):1569-78. doi: 10.1016/S0140-6736(20)31022-9, PMID 32423584.

  39. Black RH, Canfield CJ, Clyde DF, Peters W. In: Bruce-Chwatt LJ, editor. Chemotherapy of malaria. 2nd ed. England: Bath Press; 1986. p. 227-44.

  40. Winzeler EA. Malaria research in the post-genomic era. Nature. 2008;455(7214):751-6. doi: 10.1038/nature07361, PMID 18843360.

  41. Parhizgar AR, Tahghighi A. Introducing new antimalarial analogues of chloroquine and amodiaquine: A narrative review. Iran J Med Sci. 2017;42(2):115-28. PMID 28360437.

  42. Savarino A, Boelaert JR, Cassone A, Majori G, Cauda R. Effects of chloroquine on viral infections: an old drug against today’s diseases? Lancet Infect Dis. 2003;3(11):722-7. doi: 10.1016/s1473-3099(03)00806-5, PMID 14592603.

  43. Keyaerts E, Vijgen L, Maes P, Neyts J, Van Ranst M. In vitro inhibition of severe acute respiratory syndrome coronavirus by chloroquine. Biochem Biophys Res Commun. 2004;323(1):264-68. doi: 10.1016/j.bbrc.2004.08.085. PMID 15351731.

  44. Keyaerts E, Li S, Vijgen L, Rysman E, Verbeeck J, Van Ranst M. Antiviral activity of chloroquine against human coronavirus OC43 infection in newborn mice. Antimicrob Agents Chemother. 2009;53(8):3416-21. doi: 10.1128/AAC.01509-08, PMID 19506054.

  45. Yan Y, Zou Z, Sun Y, Li X, Xu KF, Wei Y. 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. doi: 10.1038/cr.2012.165, PMID 23208422.

  46. Blau DM, Holmes KV. Human coronavirus HCoV-229E enters susceptible cells via the endocytic pathway. Adv Exp Med Biol. 2001;494:193-8. doi: 10.1007/978-1-4615-1325-4_31, PMID 11774468.

  47. Johansen LM, Brannan JM, Delos SE, Shoemaker CJ, Stossel A, Lear C. FDA-approved selective estrogen receptor modulators inhibit Ebola virus infection. Sci Transl Med. 2013;5(190):190ra79. doi: 10.1126/scitranslmed.3005471, PMID 23785035.

  48. Madrid PB, Chopra S, Manger ID, Gilfillan L, Keepers TR, Shurtleff AC. A systematic screen of FDA-approved drugs for inhibitors of biological threat agents. PLoS One 2013;8(4):e60579. doi: 10.1371/journal.pone.0060579.

  49. Devaux CA, Rolain JM, Colson P, Raoult D. New insights on the antiviral effects of chloroquine against coronavirus: what to expect for COVID-19? Int J Antimicrob Agents. 2020;55(5):105938. doi: 10.1016/j.ijantimicag.2020.105938. ijantimicag.2020.105938. PMID 32171740.

  50. Gao J, Tian Z, Yang X. Breakthrough: chloroquine phosphate has shown apparent efficacy in treatment of COVID-19 associated pneumonia in clinical studies. BiosSci Trends. 2020;14(1):72-3. doi: 10.5582/bst.2020.01047, PMID 32074550. 

  51. Borba MGS, Val FFA, Sampaio VS, Alexandre MAA, Melo GC, Brito M. Effect of High vs Low doses of chloroquine diphosphate as adjunctive therapy for patients hospitalized with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection: A randomized clinical trial. JAMA Netw Open. 2020;3(4):e208857. doi: 10.1001/jamanetworkopen.2020.8857, PMID 32330277.

  52. Huang M, Tang T, Pang P, Li M, Ma R, Lu J. Treating COVID-19 with Cchloroquine. J Mol Cell Biol. 2020;12(4):322-5. doi: 10.1093/jmcb/mjaa014, PMID 32236562.

  53. Net. MedicineNet. Hydroxychloroquine (Plaquenil); 2020.

  54. Zhang W, Zhao Y, Zhang F, Wang Q, Li T, Liu Z. The use of anti-inflammatory drugs in the treatment of people with severe coronavirus disease 2019 (COVID-19): the perspectives of clinical immunologists from China. Clin Immunol. 2020;214:108393. doi: 10.1016/j.clim.2020.108393. PMID 32222466.

  55. Marmor MF, Kellner U, Lai TY, Melles RB, Mieler WF, American Academy of Ophthalmology. Recommendations on screening for chloroquine and hydroxychloroquine retinopathy (2016 Revision). Ophthalmology. 2016;123(6):1386-94. doi: 10.1016/j.ophtha.2016.01.058, PMID 26992838 Revision). Ophthalmology 2016;123(6):1386-94. doi: 10.1016/j.ophtha.2016.01.058.

  56. Jean SS, Lee PI, Hsueh PR. Treatment options for COVID-19: the reality and challenges. J Microbiol Immunol Infect. 2020;53(3):436-43. doi: 10.1016/j.jmii.2020.03.034, PMID 32307245.

  57. Yao X, Ye F, Zhang M, Cui C, Huang B, Niu P. 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;1771(15):732-9. doi: 10.1093/cid/ciaa237, PMID 32150618. 

  58. Mahevas M, Tran VT, Roumier M, Chabrol A, Paule R, Guillaud C. Clinical efficacy of hydroxychloroquine in patients with covid-19 pneumonia who require oxygen: observational comparative study using routine care data. BMJ. 2020;369:m1844. doi: 10.1136/bmj.m1844. m1844. PMID 32409486.

  59. Geleris J, Sun Y, Platt J, Zucker J, Baldwin M, Hripcsak G. Observational study of hydroxychloroquine in hospitalized patients with covid-19. N Engl J Med. 2020;382(25):2411-8. doi: 10.1056/NEJMoa2012410, PMID 32379955.

  60. Magagnoli J, Narendran S, Pereira F, Cummings TH, Hardin JW, Sutton SS. Outcomes of hydroxychloroquine usage in United States veterans hospitalized with COVID-19. medRxiv [preprint]. medRxiv. 2020:2020.04.16.20065920. doi: 10.1101/2020.04.16.20065920, PMID 32511622.

  61. Tang W, Cao Z, Han M, Wang Z, Chen J, Sun W. Hydroxychloroquine in patients with mainly mild to moderate coronavirus disease 2019: open label, randomised controlled trial. BMJ. 2020;369:m1849. doi: 10.1136/bmj.m1849. PMID 32409561.

  62. Choudhary R, Sharma AK. Potential use of hydroxychloroquine, ivermectin and azithromycin drugs in fighting COVID-19: trends, scope and relevance. New Microbes New Infect. 2020;35:100684. doi: 10.1016/j.nmni.2020.100684. PMID 32322397. 

  63. Retallack H, Di Lullo E, Arias C, Knopp KA, Laurie MT, Sandoval-Espinosa C. Zika virus cell tropism in the developing human brain and inhibition by azithromycin. Proc Natl Acad Sci USA. 2016;113(50):14408-13. doi: 10.1073/pnas.1618029113, PMID 27911847. 

  64. Bosseboeuf E, Aubry M, Nhan T, de Pina JJ, Rolain JM, Raoult D. Azithromycin inhibits the replication of Zika virus. J Antivirals Antiretrovirals. 2018;10(1):6-11. doi: 10.4172/1948-5964.1000173.

  65. Gautret P, Lagier JC, Parola P, Hoang VT, Meddeb L, Mailhe M. Hydroxychloroquine and azithromycin as a treatment of COVID-19: results of an open-label non-randomized clinical trial. Int J Antimicrob Agents. 2020;56(1):105949. doi: 10.1016/j.ijantimicag.2020.105949, PMID 32205204.

  66. Gautret P, Lagier JC, Parola P, Hoang VT, Meddeb L, Sevestre J. 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. doi: 10.1016/j.tmaid.2020.101663. PMID 32289548.

  67. Mercuro NJ, Yen CF, Shim DJ, Maher TR, McCoy CM, Zimetbaum PJ. Risk of QT interval prolongation associated with use of hydroxychloroquine with or without concomitant azithromycin among hospitalized patients testing positive for coronavirus disease 2019 (COVID-19). JAMA Cardiol. 2020;5(9):1036-41. doi: 10.1001/jamacardio.2020.1834, PMID 32936252.

  68. Molina JM, Delaugerre C, Le Goff J, Mela-Lima B, Ponscarme D, Goldwirt L. No evidence of rapid antiviral clearance or clinical benefit with the combination of hydroxychloroquine and azithromycin in patients with severe COVID-19 infection. Med Mal Infect. 2020;50(4):384. doi: 10.1016/j.medmal.2020.03.006. PMID 32240719.

  69. Chorin E, Dai M, Shulman E, Wadhwani L, Bar-Cohen R, Barbhaiya C. The QT interval in patients with COVID-19 treated with hydroxychloroquine and azithromycin. Nat Med. 2020;26(6):808-9. doi: 10.1038/s41591-020-0888-2, PMID 32488217. 

  70. Cavalcanti AB, Zampieri FG, Rosa RG, Azevedo LCP, Veiga VC, Avezum A. Hydroxychloroquine with or without azithromycin in mild-to-moderate covid-19. N Engl J Med. 2020;383(21):2041-52. doi: 10.1056/NEJMoa2019014, PMID 32706953.

  71. Sham HL, Kempf DJ, Molla A, Marsh KC, Kumar GN, Chen CM. ABT-378, a highly potent inhibitor of the human immunodeficiency virus protease. Antimicrob Agents Chemother. 1998;42(12):3218-24. doi: 10.1128/AAC.42.12.3218, PMID 9835517.

  72. Debouck C. The HIV-1 protease as a therapeutic target for AIDS. AIDS Res Hum Retroviruses. 1992;8(2):153-64. doi: 10.1089/aid.1992.8.153, PMID 1540403.

  73. Chu CM, Cheng VC, Hung IF, Wong MM, Chan KH, Chan KS. Role of lopinavir/ritonavir in the treatment of SARS: initial virological and clinical findings. Thorax. 2004;59(3):252-6. doi: 10.1136/thorax.2003.012658, PMID 14985565.

  74. Chen F, Chan KH, Jiang Y, Kao RY, Lu HT, Fan KW. In vitro susceptibility of 10 clinical isolates of SARS coronavirus to selected antiviral compounds. J Clin Virol. 2004;31(1):69-75. doi: 10.1016/j.jcv.2004.03.003. PMID 15288617. 

  75. Wu CY, Jan JT, Ma SH, Kuo CJ, Juan HF, Cheng YS. Small molecules targeting severe acute respiratory syndrome human coronavirus. Proc Natl Acad Sci USA. 2004;101(27):10012-7. doi: 10.1073/pnas.0403596101, PMID 15226499.

  76. Chan JF, Yao Y, Yeung ML, Deng W, Bao L, Jia L. 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(12):1904-13. doi: 10.1093/infdis/jiv392, PMID 26198719.

  77. Lim J, Jeon S, Shin HY, Kim MJ, Seong YM, Lee WJ. Case of the index patient who caused tertiary transmission of coronavirus disease 2019 in Korea: the application of lopinavir/ritonavir for the treatment of COVID-19 pneumonia monitored by quantitative RT-PCR. J Korean Med Sci. 2020;35(6):e79. doi: 10.3346/jkms.2020.35.e79.

  78. Wang Z, Chen X, Lu Y, Chen F, Zhang W. Clinical characteristics and therapeutic procedure for four cases with 2019 novel coronavirus pneumonia receiving combined Chinese and Western medicine treatment. BioSci Trends. 2020;14(1):64-8. doi: 10.5582/bst.2020.01030, PMID 32037389.

  79. Cao B, Wang Y, Wen D, Liu W, Wang J, Fan G. A trial of lopinavir-ritonavir in adults hospitalized with severe covid-19. N Engl J Med. 2020;382(19):1787-99. doi: 10.1056/NEJMoa2001282, PMID 32187464.

  80. Chen J, Ling Y, Xi X, Liu P, Li F, Li T. Efficacies of lopinavir/ritonavir and abidol in the treatment of novel coronavirus pneumonia. Chin J Infect Dis. 2020;38:86-9. doi: 10.1016/j.medj.2020.04.001.

  81. Li Y, Xie Z, Lin W, Cai W, Wen C, Guan Y. Efficacy and safety of lopinavir/ritonavir or arbidol in adult patients with mild/moderate COVID-19: an exploratory randomized controlled trial. Med (NY). 2020;1(1):105-13105-113.e4. doi: 10.1016/j.medj.2020.04.001. PMID 32838353. 

  82. Deeks ED. Darunavir/cobicistat/emtricitabine/tenofovir alafenamide: a review in HIV-1 infection. Drugs. 2018;78(10):1013-24. doi: 10.1007/s40265-018-0934-2, PMID 29915897.

  83. JohnsonandJohnson. Lack of evidence to support use of darunavir based treatment for SARS-CoV-2 [internet]. US Johnson and Johnson; 2020.

  84. Iyer M, Jayaramayya K, Subramaniam MD, Lee SB, Dayem AA, Cho SG. COVID-19: an update on diagnostic and therapeutic approaches. BMB Rep. 2020;53(4):191-205. doi: 10.5483/BMBRep.2020.53.4.080, PMID 32336317.

  85. Rajam G, Sampson J, Carlone GM, Ades EW. An augmented passive immune therapy to treat fulminant bacterial infections. Recent Pat Antiinfect Drug Discov. 2010;5(2):157-67. doi: 10.2174/157489110791233496, PMID 20370679.

  86. Virdi V, Depicker A. Role of plant expression systems in antibody production for passive immunization. Int J Dev Biol. 2013;57(6-8):587-93. doi: 10.1387/ijdb.130266ad, PMID 24166441.

  87. Roback JD, Guarner J. Convalescent plasma to treat COVID-19: possibilities and challenges. JAMA. 2020;323(16):1561-2. doi: 10.1001/jama.2020.4940, PMID 32219429.

  88. International Severe Acute Respiratory and Emerging Infection Consortium (U.K.). Treatment of MERS-CoV: information for Clinicians Clinical decision-making support for treatment of MERS-CoV patients. London: Public Health England; 2014.

  89. Hung IF, To KK, Lee CK, Lee KL, Chan K, Yan WW. Convalescent plasma treatment reduced mortality in patients with severe pandemic influenza a (H1N1) 2009 virus infection. Clin Infect Dis. 2011;155252(4):447-56. doi: 10.1093/cid/ciq106, PMID 21248066.

  90. Soo YO, Cheng Y, Wong R, Hui DS, Lee CK, Tsang KK. Retrospective comparison of convalescent plasma with continuing high-dose methylprednisolone treatment in SARS patients. Clin Microbiol Infect. 2004;10(7):676-8. doi: 10.1111/j.1469-0691.2004.00956.x.x. PMID 15214887. 

  91. Liu W, Fontanet A, Zhang PH, Zhan L, Xin ZT, Baril L. Two-year prospective study of the humoral immune response of patients with severe acute respiratory syndrome. J Infect Dis. 2006;193(6):792-5. doi: 10.1086/500469, PMID 16479513.

  92. Yeh KM, Chiueh TS, Siu LK, Lin JC, Chan PK, Peng MY. Experience of using convalescent plasma for severe acute respiratory syndrome among healthcare workers in a Taiwan hospital. J Antimicrob Chemother. 2005;56(5):919-22. doi: 10.1093/jac/dki346, PMID 16183666.

  93. Luke TC, Kilbane EM, Jackson JL, Hoffman SL. Meta-analysis: convalescent blood products for Spanish influenza pneumonia: a future H5N1 treatment? Ann Intern Med. 2006;145(8):599-609. doi: 10.7326/0003-4819-145-8-200610170-00139, PMID 16940336.

  94. Casadevall A, Pirofski LA. The convalescent sera option for containing COVID-19. J Clin Invest. 2020;130(4):1545-8. doi: 10.1172/JCI138003, PMID 32167489. 

  95. Zeng QL, Yu ZJ, Gou JJ, Li GM, Ma SH, Zhang GF. Effect of convalescent plasma therapy on viral shedding and survival in patients with coronavirus disease 2019. J Infect Dis. 2020;222(1):38-43. doi: 10.1093/infdis/jiaa228, PMID 32348485. 

  96. Duan K, Liu B, Li C, Zhang H, Yu T, Qu J. Effectiveness of convalescent plasma therapy in severe COVID-19 patients. Proc Natl Acad Sci USA. 2020;117(17):9490-6. doi: 10.1073/pnas.2004168117, PMID 32253318. 

  97. Wang X, Guo X, Xin Q, Pan Y, Hu Y, Li J. Neutralizing antibody responses to severe acute respiratory syndrome coronavirus 2 in coronavirus disease 2019 inpatients and convalescent patients. Clin Infect Dis. 2020;71(10):2688-94. doi: 10.1093/cid/ciaa721, PMID 32497196.

  98. Ye M, Fu D, Ren Y, Wang F, Wang D, Zhang F. Treatment with convalescent plasma for COVID-19 patients in Wuhan, China. J Med Virol. 2020;92(10):1890-1901. doi: 10.1002/jmv.25882, PMID 32293713. 

  99. Ahn JY, Sohn Y, Lee SH, Cho Y, Hyun JH, Baek YJ. Use of convalescent plasma therapy in two COVID-19 patients with acute respiratory distress syndrome in Korea. J Korean Med Sci. 2020;35(14):e149. doi: 10.3346/jkms.2020.35.e149. PMID 32281317.

  100. Li L, Zhang W, Hu Y, Tong X, Zheng S, Yang J. Effect of convalescent plasma therapy on time to clinical improvement in patients with severe and life-threatening COVID-19: A randomized clinical trial. JAMA. 2020;324(5):460-70. doi: 10.1001/jama.2020.10044, PMID 32492084. 

  101. Shao Z, Feng Y, Zhong L, Xie Q, Lei M, Liu Z. Clinical efficacy of intravenous immunoglobulin therapy in critical ill patients with COVID-19: a multicenter retrospective cohort study. Clin Transl Immunology. 2020;9(10):e1192. doi: 10.1002/cti2.1192, PMID 33082954.

  102. Galeotti C, Kaveri SV, Bayry J. IVIG-mediated effector functions in autoimmune and inflammatory diseases. Int Immunol. 2017;29(11):491-8. doi: 10.1093/intimm/dxx039, PMID 28666326.

  103. Hartung HP. Advances in the understanding of the mechanism of action of IVIg. J Neurol. 2008;3255Suppl 3:3-6. doi: 10.1007/s00415-008-3002-0, PMID 18685919. 

  104. Busani S, Damiani E, Cavazzuti I, Donati A, Girardis M. Intravenous immunoglobulin in septic shock: review of the mechanisms of action and meta-analysis of the clinical effectiveness. Minerva Anestesiol. 2016;82(5):559-72. PMID 26474267.

  105. Lai ST. Treatment of severe acute respiratory syndrome. Eur J Clin Microbiol Infect Dis. 2005;24(9):583-91. doi: 10.1007/s10096-005-0004-z, PMID 16172857.

  106. Sakoulas G, Geriak M, Kullar R, Greenwood Kristina HM, Vyas A. Intravenous immunoglobulin (IVIG) significantly reduces respiratory morbidity in COVID-19 pneumonia: a prospective randomized trial. medRxiv; 2020. doi: 10.1101/2020.07.20.20157891.

  107. Golchin A, Farahany TZ, Khojasteh A, Soleimanifar F, Ardeshirylajimi A. The clinical trials of mesenchymal stem cell therapy in skin diseases: an update and concise review. Curr Stem Cell Res Ther. 2019;14(1):22-33. doi: 10.2174/1574888X13666180913123424, PMID 30210006.

  108. Leng Z, Zhu R, Hou W, Feng Y, Yang Y, Han Q. Transplantation of ACE2-mesenchymal stem cells improves the outcome of patients with COVID-19 pneumonia. Aging Dis. 2020;11(2):216-28. doi: 10.14336/AD.2020.0228, PMID 32257537.

  109. Liang B, Chen J, Li T, Wu H, Yang W, Li Y. Clinical remission of a critically ill COVID-19 patient treated by human umbilical cord mesenchymal stem cells: A case report. Medicine (Baltimore). 2020;99(31):e21429. doi: 10.1097/MD.0000000000021429, PMID 32756149.

  110. Thompson BT. Glucocorticoids and acute lung injury. Crit Care Med. 2003;31(4)Suppl:S253-S257. doi: 10.1097/01.CCM.0000057900.19201.55. PMID 12682449.

  111. Monton C, Ewig S, Torres A, El-Ebiary M, Filella X, Rano A. Role of glucocorticoids on inflammatory response in non immunosuppressed patients with pneumonia: a pilot study. Eur Respir J. 1999;14(1):218-20. doi: 10.1034/j.1399-3003.1999.14a37.x.x. PMID 10489855. 

  112. Cain DW, Cidlowski JA. Immune regulation by glucocorticoids. Nat Rev Immunol. 2017;17(4):233-47. doi: 10.1038/nri.2017.1, PMID 28192415.

  113. Stockman LJ, Bellamy R, Garner P. SARS: systematic review of treatment effects. PLoOS Med. 2006;3(9):e343. doi: 10.1371/journal.pmed.0030343. PMID 16968120. 

  114. Chen RC, Tang XP, Tan SY, Liang BL, Wan ZY, Fang JQ. Treatment of severe acute respiratory syndrome with glucosteroids: the Guangzhou experience. Chest. 2006;129(6):1441-52. doi: 10.1378/chest.129.6.1441, PMID 16778260.

  115. Sung JJ, Wu A, Joynt GM, Yuen KY, Lee N, Chan PK. Severe acute respiratory syndrome: report of treatment and outcome after a major outbreak. Thorax. 2004;59(5):414-20. doi: 10.1136/thx.2003.014076, PMID 15115870. 

  116. University of Oxford. Low-cost dexamethasone reduces death by up to one-third in hospitalised patients with severe respiratory complications of COVID-19 [internet]. UK: University of Oxford; 2020. Available from: https://www.ox.ac.uk/news/2020-06-16-low-cost-dexamethasone-reduces-death-one-third-hospitalised-patients-severe. [Last accessed on 23 Jun 2020]

  117. Wang Y, Jiang W, He Q, Wang C, Wang B, Zhou P. Early, low-dose and short-term application of corticosteroid treatment in patients with severe COVID-19 pneumonia: single-center experience from Wuhan, China. medRxiv; 2020. doi: 10.1101/2020.03.06.20032342.

  118. Fadel R, Morrison AR, Vahia A, Smith ZR, Chaudhry Z, Bhargava P. Early short-course corticosteroids in hospitalized patients with COVID-19. Clin Infect Dis. 2020;71(16):2114-20. doi: 10.1093/cid/ciaa601, PMID 32427279.

  119. Corral Gudino L, Bahamonde A, Arnaiz Revillas F, Gomez Barquero J, Abadia Otero J, Garcia Ibarbia C. Methylprednisolone in adults hospitalized with COVID-19 pneumonia: an open-label randomized trial (GLUCOCOVID). Wien Klin Wochenschr. 2021;133(7-8):303-11. doi: 10.1007/s00508-020-01805-8. 

  120. Sun F, Kou H, Wang S, Lu Y, Zhao H, Li W. Medication patterns and disease progression among 165 patients with coronavirus disease 2019 (COVID-19) in Wuhan, China: A single-centered, retrospective, observational study; 2019. doi: 10.2139/ssrn.3551323.

  121. Russell CD, Millar JE, Baillie JK. Clinical evidence does not support corticosteroid treatment for 2019-nCoV lung injury. Lancet. 2020;395(10223):473-5. doi: 10.1016/S0140-6736(20)30317-2.

  122. Keller MJ, Kitsis EA, Arora S, Chen JT, Agarwal S, Ross MJ. Effect of systemic glucocorticoids on mortality or mechanical ventilation in patients with COVID-19. J Hosp Med. 2020;15(8):489-93. doi: 10.12788/jhm.3497, PMID 32804611.

  123. Yang Z, Liu J, Zhou Y, Zhao X, Zhao Q, Liu J. The effect of corticosteroid treatment on patients with coronavirus infection: a systematic review and meta-analysis. J Infect. 2020;81(1):e13-20. doi: 10.1016/j.jinf.2020.03.062, PMID 32283144.

  124. Schneider WM, Chevillotte MD, Rice CM. Interferon-stimulated genes: a complex web of host defenses. Annu Rev Immunol. 2014;32:513-45. doi: 10.1146/annurev-immunol-032713-120231, PMID 24555472.

  125. Arabi YM, Shalhoub S, Mandourah Y, Al-Hameed F, Al-Omari A, Al- Qasim E. Ribavirin and interferon therapy for critically ill patients with Middle East respiratory syndrome: A multicenter observational study. Clin Infect Dis. 2020;70(9):1837-44. doi: 10.1093/cid/ciz544, PMID 31925415.

  126. Omrani AS, Saad MM, Baig K, Bahloul A, Abdul-Matin M, Alaidaroos AY. Ribavirin and interferon alfa-2a for severe Middle East respiratory syndrome coronavirus infection: a retrospective cohort study. Lancet Infect Dis. 2014;14(11):1090-5. doi: 10.1016/S1473-3099(14)70920-X, PMID 25278221. 

  127. Shalhoub S, Farahat F, Al-Jiffri A, Simhairi R, Shamma O, Siddiqi N. IFN-α2a or IFN-β1a in combination with ribavirin to treat Middle East respiratory syndrome coronavirus pneumonia: a retrospective study. J Antimicrob Chemother. 2015;70(7):2129-32. doi: 10.1093/jac/dkv085, PMID 25900158.

  128. Wong SS, Yuen KY. The management of coronavirus infections with particular reference to SARS. J Antimicrob Chemother. 2008;62(3):437-41. doi: 10.1093/jac/dkn243, PMID 18565970.

  129. Loutfy MR, Blatt LM, Siminovitch KA, Ward S, Wolff B, Lho H. Interferon alfacon-1 plus corticosteroids in severe acute respiratory syndrome: a preliminary study. JAMA. 2003;290(24):3222-8. doi: 10.1001/jama.290.24.3222, PMID 14693875.

  130. Davoudi Monfared E, Rahmani H, Khalili H, Hajiabdolbaghi M, Salehi M, Abbasian L. A randomized clinical trial of the efficacy and safety of interferon β-1a in treatment of severe COVID-19. Antimicrob Agents Chemother. 2020;64(9):e01061-20. doi: 10.1128/AAC.01061-20, PMID 32661006.

  131. Zhou Q, Chen V, Shannon CP, Wei XS, Xiang X, Wang X. Interferon-α2b treatment for COVID-19. Front Immunol. 2020;11:1061. doi: 10.3389/fimmu.2020.01061, PMID 32574262.

  132. Hung IF, Lung KC, Tso EY, Liu R, Chung TW, Chu MY. 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;395(10238):1695-704. doi: 10.1016/S0140-6736(20)31042-4, PMID 32401715.

  133. Meng Z, Wang T, Chen L, Chen X, Li L, Qin X. The effect of recombinant human interferon alpha nasal drops to prevent COVID-19 pneumonia for medical staff in an epidemic area. Curr Top Med Chem. 2021;21(10):920-7. doi: 10.2174/1568026621666210429083050. PMID 33970846.

  134. Idelsis EM, Jesus PE, Yaquelin DR, Dania VB, Monica BR, Lisandra BR. Effect and safety of combination of interferon alpha-2b and gamma or interferon alpha-2b for negativization of SARS-CoV-2 viral RNA. Preliminary results of a randomized controlled clinical trial. medRxiv; 2020. doi: 10.1101/2020.07.29.20164251.

  135. Winthrop KL, Mariette X, Silva JT, Benamu E, Calabrese LH, Dumusc A. ESCMID study group for infections in compromised hosts (ESGICH) consensus document on the safety of targeted and biological therapies: an infectious diseases perspective (Soluble immune effector molecules [II]: agents targeting interleukins, immunoglobulins and complement factors). Clin Microbiol Infect. 2018;24Suppl 2:S21-S40. doi: 10.1016/j.cmi.2018.02.002, PMID 29447987.

  136. Shakoory B, Carcillo JA, Chatham WW, Amdur RL, Zhao H, Dinarello CA. Interleukin-1 receptor blockade is associated with reduced mortality in sepsis patients with features of macrophage activation syndrome: reanalysis of a prior Phase III trial. Crit Care Med. 2016;44(2):275-81. doi: 10.1097/CCM.0000000000001402, PMID 26584195.

  137. Monteagudo LA, Boothby A, Gertner E. Continuous intravenous anakinra infusion to calm the cytokine storm in macrophage activation syndrome. ACR Open Rheumatol. 2020;2(5):276-82. doi: 10.1002/acr2.11135, PMID 32267081.

  138. Mehta P, McAuley DF, Brown M, Sanchez E, Tattersall RS, Manson JJ. COVID-19: consider cytokine storm syndromes and immunosuppression. Lancet. 2020;395(10229):1033-4. doi: 10.1016/S0140-6736(20)30628-0, PMID 32192578.

  139. Aouba A, Baldolli A, Geffray L, Verdon R, Bergot E, Martin-Silva N. Targeting the inflammatory cascade with anakinra in moderate to severe COVID-19 pneumonia: case series. Ann Rheum Dis. 2020;79(10):1381-2. doi: 10.1136/annrheumdis-2020-217706, PMID 32376597.

  140. Cavalli G, De Luca G, Campochiaro C, Della Torre E, Ripa M, Canetti D. Interleukin-1 blockade with high-dose anakinra in patients with COVID-19, acute respiratory distress syndrome, and hyperinflammation: a retrospective cohort study. Lancet Rheumatol. 2020;2(6):e325-e331. doi: 10.1016/S2665-9913(20)30127-2, PMID 32501454.

  141. Xu X, Han M, Li T, Sun W, Wang D, Fu B. Effective treatment of severe COVID-19 patients with tocilizumab. Proc Natl Acad Sci USA. 2020;117(20):10970-5. doi: 10.1073/pnas.2005615117, PMID 32350134.

  142. Sciascia S, Apra F, Baffa A, Baldovino S, Boaro D, Boero R. Pilot prospective open, single-arm multicentre study on off-label use of tocilizumab in patients with severe COVID-19. Clin Exp Rheumatol. 2020;38(3):529-32. PMID 32359035.

  143. Guaraldi G, Meschiari M, Cozzi Lepri A, Milic J, Tonelli R, Menozzi M. Tocilizumab in patients with severe COVID-19: a retrospective cohort study. Lancet Rheumatol. 2020;2(8):e474-e484. doi: 10.1016/S2665-9913(20)30173-9, PMID 32835257.

  144. Rhee VFvan Rhee F, Wong RS, Munshi N, Rossi JF, Ke XY, Fossa A. Siltuximab for multicentric Castleman'’s disease: a randomised, double-blind, placebo-controlled trial. Lancet Oncol. 2014;15(9):966-74. doi: 10.1016/S1470-2045(14)70319-5, PMID 25042199. 

  145. Gritti G, Raimondi F, Ripamonti D, Riva I, Landi F, Alborghetti L. Use of siltuximab in patients with COVID-19 pneumonia requiring ventilatory support. medRxiv; 2020. doi: 10.1101/2020.04.01.20048561.

  146. Regeneron. Regeneron and Sanofi provide update on U.S. Phase 2/3 adaptive-designed trial of KEVZARA® (sarilumab) in hospitalized COVID-19 patients; 2020.

  147. Wang M, Rule S, Zinzani PL, Goy A, Casasnovas O, Smith SD. Acalabrutinib in relapsed or refractory mantle cell lymphoma (ACE-LY-004): a single-arm, multicentre, phase 2 trial. Lancet. 2018;391(10121):659-67. doi: 10.1016/S0140-6736(17)33108-2, PMID 29241979.

  148. Roschewski M, Lionakis MS, Sharman JP, Roswarski J, Goy A, Monticelli MA. Inhibition of bruton tyrosine kinase in patients with severe COVID-19. Sci Immunol. 2020;5(48):eabd0110. doi: 10.1126/sciimmunol.abd0110. PMID 32503877. 

  149. Treon SP, Castillo JJ, Skarbnik AP, Soumerai JD, Ghobrial IM, Guerrera ML. The BTK inhibitor ibrutinib may protect against pulmonary injury in COVID-19-infected patients. Blood. 2020;135(21):1912-5. doi: 10.1182/blood.2020006288, PMID 32302379.

  150. Dougados M, van der Heijde DVD, Chen YC, Greenwald M, Drescher E, Liu J. Baricitinib in patients with inadequate response or intolerance to conventional synthetic DMARDs: results from the RA-BUILD study. Ann Rheum Dis. 2017;76(1):88-95. doi: 10.1136/annrheumdis-2016-210094, PMID 27689735.

  151. Stebbing J, Phelan A, Griffin I, Tucker C, Oechsle O, Smith D. COVID-19: combining antiviral and anti-inflammatory treatments. Lancet Infect Dis. 2020;20(4):400-2. doi: 10.1016/S1473-3099(20)30132-8, PMID 32113509.

  152. Genovese MC, Kremer J, Zamani O, Ludivico C, Krogulec M, Xie L. Baricitinib in patients with refractory rheumatoid arthritis. N Engl J Med. 2016;374(13):1243-52. doi: 10.1056/NEJMoa1507247, PMID 27028914.

  153. Smolen JS, Genovese MC, Takeuchi T, Hyslop DL, Macias WL, Rooney T. Safety profile of baricitinib in patients with active rheumatoid arthritis with over 2 years median time in treatment. J Rheumatol. 2019;46(1):7-18. doi: 10.3899/jrheum.171361, PMID 30219772. 

  154. Richardson P, Griffin I, Tucker C, Smith D, Oechsle O, Phelan A. Baricitinib as potential treatment for 2019-nCoV acute respiratory disease. Lancet. 2020;395(10223):e30-e31. doi: 10.1016/S0140-6736(20)30304-4, PMID 32032529. 

  155. Cantini F, Niccoli L, Matarrese D, Nicastri E, Stobbione P, Goletti D. Baricitinib therapy in COVID-19: A pilot study on safety and clinical impact. J Infect. 2020;81(2):318-56. doi: 10.1016/j.jinf.2020.04.017, PMID 32333918.

  156. Cao Y, Wei J, Zou L, Jiang T, Wang G, Chen L. Ruxolitinib in treatment of severe coronavirus disease 2019 (COVID-19): A multicenter, single-blind, randomized controlled trial. J Allergy Clin Immunol. 2020;146(1):137-146.e3. doi: 10.1016/j.jaci.2020.05.019, PMID 32470486.

  157. Rosee LFLa Rosee F, Bremer HC, Gehrke I, Kehr A, Hocchhaus A, Birndt S. The Janus kinase 1/2 inhibitor ruxolitinib in COVID-19 with severe systemic hyperinflammation. Leukemia. 2020;34(7):1805-15. doi: 10.1038/s41375-020-0891-0, PMID 32518419.

  158. US Food and Drug Administration. FDA approves first treatment for COVID-19. US FDA; 2020.

  159. National Institute of Health. NIH clinical trial testing remdesivir plus interferon beta-1a for COVID-19 treatment begins; 2020.

  160. Clinical Trials. gov. Ibrutinib for the treatment of COVID-19; 2020. Patients requiring hospitalization. US Clinical Trials. gov. Available from: https://clinicaltrials.gov/ct2/show/NCT04439006. [Last accessed on Sep 26 2020].

  161. Clinical Trials. gov. Study of oral ibrutinib capsules to assess respiratory failure in adult participants with severe acute respiratory syndrome Coronavirus-2 (SARS-CoV-2) and pulmonary Injury (iNSPIRE); 2020. Available from: https://clinicaltrials.gov/ct2/show/NCT04375397. [Last accessed on 26 Mar 2021].

  162. Clinical Trials. gov. Ruxolitinib in Covid-19 Patients with defined hyperinflammation (RuxCoFlam); 2020. Available from: https://clinicaltrials.gov/ct2/show/NCT04338958. [Last accessed on 30 Mar 2021]

  163. National Institutes of Health. NIH clinical trial testing antiviral remdesivir plus anti-inflammatory drug baricitinib for COVID-19 begins; 2020. Available from: https://www.nih.gov/news-events/news-releases/nih-clinical-trial-testing-antiviral-remdesivir-plus-anti-inflammatory-drug-baricitinib-covid-19-begins. Us NIH. [Last accessed on 02 Sep 2020]

  164. Vincent MJ, Bergeron E, Benjannet S, Erickson BR, Rollin PE, Ksiazek TG. Chloroquine is a potent inhibitor of SARS coronavirus infection and spread. Virol J. 2005;2:69. doi: 10.1186/1743-422X-2-69, PMID 16115318.

  165. Ministry of Health and Family Welfare. Revised Advisory on the Use of hydroxychloroquine (HCQ) as prophylaxis for COVID-19 Infection (in supersession of previous advisory dated 23rd March (2020). New Delhi (Asia) Ministry of Health and Family Welfare; 2020 Available from: https://www.mohfw.gov.in/pdf/RevisedadvisoryontheuseofhydroxychloroquineasprophylaxisforSARSCOVID19infection.pdf. [ Last accessed on 20 Apr 2020]

  166. Gielen V, Johnston SL, Edwards MR. Azithromycin induces anti-viral responses in bronchial epithelial cells. Eur Respir J. 2010;36(3):646-54. doi: 10.1183/09031936.00095809, PMID 20150207.

  167. Culic O, Erakovic V, Cepelak I, Barisic K, Brajsa K, Ferencic Z. Azithromycin modulates neutrophil function and circulating inflammatory mediators in healthy human subjects. Eur J Pharmacol. 2002;450(3):277-89. doi: 10.1016/s0014-2999(02)02042-3, PMID 12208321.

  168. Rosenberg ES, Dufort EM, Udo T, Wilberschied LA, Kumar J, Tesoriero J. Association of treatment with hydroxychloroquine or azithromycin with in-hospital mortality in patients with COVID-19 in New York State. JAMA. 2020;323(24):2493-502. doi: 10.1001/jama.2020.8630, PMID 32392282.

  169. Choy KT, Wong AY, Kaewpreedee P, Sia SF, Chen D, Hui KPY. Remdesivir, lopinavir, emetine, and homoharringtonine inhibit SARS-CoV-2 replication in vitro. Antiviral Res. 2020;178:104786. doi: 10.1016/j.antiviral.2020.104786. PMID 32251767.

  170. Nukoolkarn V, Lee VS, Malaisree M, Aruksakulwong O, Hannongbua S. Molecular dynamic simulations analysis of ritonavir and lopinavir as SARS-CoV 3CL (pro) inhibitors. J Theor Biol. 2008;254(4):861-7. doi: 10.1172016/JCI140j.jtbi.2008.07.030, PMID 18706430.

  171. Joyner MJ, Wright RS, Fairweather D, Senefeld JW, Bruno KA, Klassen SA. Early safety indicators of COVID-19 convalescent plasma in 5000 patients. J Clin Invest 2020;130(9):4791-7. doi: 10.1172/JCI140200.

  172. Lu X, Chen T, Wang Y, Wang J, Yan F. Adjuvant corticosteroid therapy for critically ill patients with COVID‐19. Crit Care. 2020;24(1):241. doi: 10.1186/s13054-020-02964-w, PMID 32430057.

  173. Mammen MJ, Aryal K, Alhazzani W, Alexander PE. Corticosteroids for patients with acute respiratory distress syndrome: a systematic review and meta-analysis of randomized trials. Pol Arch Intern Med. 2020;130(4):276-86. doi: 10.20452/pamw.15239, PMID 32186831. 

  174. Villar J, Ferrando C, Martinez D, Ambros A, Munoz T, Soler JA. Dexamethasone treatment for the acute respiratory distress syndrome: a multicentre, randomised controlled trial. Lancet Respir Med. 2020;8(3):267-76. doi: 10.1016/S2213-2600(19)30417-5, PMID 32043986.

  175. Horby P, Lim WS, Emberson JR, Mafham M, Bell JL. Dexamethasone in hospitalized patients with covid-19-preliminary. N Engl J Med. 2020;384:693-704.

  176. Confalonieri M, Urbino R, Potena A, Piattella M, Parigi P, Puccio G. Hydrocortisone infusion for severe community-acquired pneumonia: a preliminary randomized study. Am J Respir Crit Care Med. 2005;171(3):242-8. doi: 10.1164/rccm.200406-808OC, PMID 15557131.

  177. Sichitiu J, Fakhouri F, Desseauve D. Antenatal corticosteroid therapy and COVID-19: pathophysiological considerations. Acta Obstet Gynecol Scand. 2020;99(7):952. doi: 10.1111/aogs.13887, PMID 32356302.

  178. Mantlo E, Bukreyeva N, Maruyama J, Paessler S, Huang C. Antiviral activities of type I interferons to SARS-CoV-2 infection. Antiviral Res. 2020;179:104811. doi: 10.1016/j.antiviral.2020.104811. PMID 32360182.

  179. Shen KL, Yang YH. Diagnosis and treatment of 2019 novel coronavirus infection in children: a pressing issue. World J Pediatr. 2020;16(3):219-21. doi: 10.1007/s12519-020-00344-6, PMID 32026147.

  180. Ranieri VM, Pettila V, Karvonen MK, Jalkanen J, Nightingale P, Brealey D. Effect of intravenous interferon β-1a on death and days free from mechanical ventilation among patients with moderate to severe acute respiratory distress syndrome: A randomized clinical trial. JAMA. 2020;323(8):725-33. doi: 10.1001/jama.2019.22525, PMID 32065831.

  181. Kiss J, Yegutkin GG, Koskinen K, Savunen T, Jalkanen S, Salmi M. IFN-beta protects from vascular leakage via up-regulation of CD73. Eur J Immunol. 2007;37(12):3334-8. doi: 10.1002/eji.200737793, PMID 18034430.

  182. Aeffner F, Woods PS, Davis IC. Activation of A1-adenosine receptors promotes leukocyte recruitment to the lung and attenuates acute lung injury in mice infected with influenza A/WSN/33 (H1N1) virus. J Virol. 2014;88(17):10214-27. doi: 10.1128/JVI.01068-14, PMID 24965449.

  183. Lang DA, Osterhaus AD, Haagmans BL. Interferon-gamma and interleukin-4 downregulate expression of the SARS coronavirus receptor ACE2. In: Vero E6 cells. Virol 2006;353(2):474-81. doi: 10.1016/j.virol.2006.06.011.

  184. Huet T, Beaussier H, Voisin O, Jouveshomme S, Dauriat G, Lazareth I. Anakinra for severe forms of COVID-19: a cohort study. Lancet Rheumatol. 2020;2(7):e393-e400. doi: 10.1016/S2665-9913(20)30164-8, PMID 32835245.

  185. Kewan T, Covut F, Al-Jaghbeer MJ, Rose L, Gopalakrishna KV, Akbik VB. Tocilizumab for treatment of patients with severe COVID-19: A retrospective cohort study.  Clinical Medicine. 2020;24:100418. doi: 10.1016/j.eclinm.2020.100418. PMID 32766537. 

  186. Worldpharmanews. Sanofi and Regeneron provide update on Kevzara® (sarilumab) Phase 3 U.S. trial in COVID-19 patients; 2020. Available from: https://www.worldpharmanews.UKworldpharmanews.com. [Last accessed on 20 Aug 2020]

  187. Favalli EG, Biggioggero M, Maioli G, Caporali R. Baricitinib for COVID-19: a suitable treatment? Lancet Infect Dis. 2020 2020;20(9):1012-3. doi: 10.1016/S1473-3099(20)30262-0, PMID 32251638.