Next Article in Journal
A Review on the Role of Endophytes and Plant Growth Promoting Rhizobacteria in Mitigating Heat Stress in Plants
Previous Article in Journal
Extracellular DNA: A Critical Aspect of Marine Biofilms
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Antiparasitic Drugs against SARS-CoV-2: A Comprehensive Literature Survey

1
Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122 Milan, Italy
2
Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy
3
Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy
*
Author to whom correspondence should be addressed.
Microorganisms 2022, 10(7), 1284; https://doi.org/10.3390/microorganisms10071284
Submission received: 5 May 2022 / Revised: 10 June 2022 / Accepted: 13 June 2022 / Published: 24 June 2022
(This article belongs to the Section Antimicrobial Agents and Resistance)

Abstract

:
More than two years have passed since the viral outbreak that led to the novel infectious respiratory disease COVID-19, caused by the SARS-CoV-2 coronavirus. Since then, the urgency for effective treatments resulted in unprecedented efforts to develop new vaccines and to accelerate the drug discovery pipeline, mainly through the repurposing of well-known compounds with broad antiviral effects. In particular, antiparasitic drugs historically used against human infections due to protozoa or helminth parasites have entered the main stage as a miracle cure in the fight against SARS-CoV-2. Despite having demonstrated promising anti-SARS-CoV-2 activities in vitro, conflicting results have made their translation into clinical practice more difficult than expected. Since many studies involving antiparasitic drugs are currently under investigation, the window of opportunity might be not closed yet. Here, we will review the (controversial) journey of these old antiparasitic drugs to combat the human infection caused by the novel coronavirus SARS-CoV-2.

1. Introduction

Considering that the COVID-19 pandemic to date has caused more than 6 million deaths worldwide, increasing the number of specific and effective therapeutic agents targeting SARS-CoV-2 represents an urgent and unmet need.
SARS-CoV-2 is an enveloped virus with a single stranded RNA of positive polarity, 29.9 kb in length, containing non-structural (nsps) and structural/accessory proteins [1,2]. The virus entry into host cells is mediated by the binding between the Receptor Binding Domain (RBD) of the viral structural Spike (S) protein and the Angiotensin Converting Enzyme II (ACE2) receptor on the target cells [3]. Following entry, the viral genome is released into the host cytoplasm, where two open reading frames are immediately translated into two polyproteins that are subsequently cleaved, leading to the production of the nsps. Instead, the structural/accessory proteins are synthesized through the translation of subgenomic RNAs (sgRNAs) [1,4]. In particular, viral structural proteins such as membrane (M), envelope (E) and S glycoproteins that are embedded into the viral envelope, are translated and translocated into the endoplasmic reticulum. Upon the replication of the viral genome, new single stranded RNAs of positive polarity are then incorporated into new viral particles [4]. The SARS-CoV-2 life cycle ends when the mature new virions leave the host cells through the exocytic pathway [5].
Concerning the pathogenesis of COVID-19, it greatly varies from patient to patient: most infected people develop no or mild symptoms, such as fever, cough, fatigue, dyspnea, myalgia, sputum production and headache [6], while others show severe symptoms [7]. In the latter patients, viral replication in lung parenchyma may cause severe pneumonia, with organ function damage and severe interstitial inflammation [8], which may induce an abundant production of pro-inflammatory cytokines and chemokines [7], a phenomenon called “cytokine storm” [9]. Resulting symptoms may be due to the cytokine-induced tissue damage or may result from immune-cell–mediated responses [10].
While a variety of drugs targeting many of the SARS-CoV-2 proteins have been evaluated, still no effective antiviral strategy for COVID-19 exists to date [11]. Currently, the few drugs for COVID-19 treatment that have gained full approval from the US Food and Drug Administration (FDA), or an emergency authorization, are mainly represented by repurposed agents: the antiviral remdesivir (fully approved), the antivirals Paxlovid (nirmatrelvir/ritonavir combination) and Lagevrio (molnupiravir), and the monoclonal antibodies Evusheld, Tocilizumab, and Bebtelovimab (authorized in case of emergency).
However, especially during the first period of the pandemic, when medicine seemed completely unable to find tools to cure COVID-19 or at least to reduce its high level of lethality, some antimalarial drugs with reported antiviral activities were used to fight SARS-CoV-2 [12]. Indeed, the repurposing approach, that is the usage of existing drugs for new therapeutic indications, is a well-known and applied strategy against epidemic virus, to reduce development costs, time to market, and risks of failure [13]. Hence, during the last SARS-CoV-2 pandemic, drug repurposing screens have emerged as an attractive strategy to accelerate new drug discovery and development pipelines, by focusing the attention on known drugs with direct/indirect antiviral effects [14].
Few months before the pandemic occurrence, we reviewed the use of antimalarial drugs against all known viral infections [15], concluding that their repurposing might be useful, especially in cases of antiviral drug-resistance or during the emergence of new viruses for which effective drugs were not readily available.
The aim of the present study was to review the available data on well-known antiparasitic agents with a special emphasis on antimalarial and anthelmintic drugs, given their significant potential to be repurposed as anti-SARS-CoV-2 drugs (Figure 1). The literature screen has been performed using the PubMed search engine by searching “SARS-CoV-2” or “COVID-19” and the single name of specific antiparasitic agents. Current knowledge of the efficacy of these drugs by in vitro/in silico, in vivo studies and clinical trials, along with their proposed mechanisms of action (MoA) against SARS-CoV-2 (Figure 2), will be discussed throughout the different sections of the present review.

2. Antimalarial Drugs

2.1. The Controversial Journey of 4-Aminoquinolines: Chloroquine (CQ) and Hydroxychloroquine (HCQ)

In 1880, the French military doctor Alphonse Laveran discovered the etiologic agent of malaria, a parasitic protozoan infecting the blood cells of the human host [16]. Already by that time, quinine, a natural compound derived from the bark of the cinchona tree, was widely used to treat malaria-infected patients [17]. It was not until 1934, that a 4-aminoquinoline termed chloroquine (CQ) (Figure 1) became the first synthetic quinine substitute and the drug of choice against malaria [18]. Later in 1955, continued chemical modification of CQ resulted in the introduction of its hydroxy-analogue hydroxychloroquine (HCQ) (Figure 1), which rapidly proved to be 3-fold less toxic and more water soluble [19].
Beyond their antimalarial action, CQ/HCQ exhibit anti-infective, anti-inflammatory, immunomodulating and antithrombotic therapeutic properties, resulting from not fully characterized mechanisms [20]. These pleiotropic abilities rapidly prompted the repurposing of both aminoquinolines as a potential treatment of several non-malarial diseases, ranging from infectious, rheumatological and autoimmune to neurological conditions [21].
As anti-infectives, CQ/HCQ have been successfully used to treat bacterial [22,23], and fungal infections [24,25,26]. Even though their exact antiviral mechanism of action is still unclear, CQ/HCQ may abrogate viral entry and replication inside host cells by altering endosomal acidification and pH-dependent enzymatic cleavage, by inhibiting post-translational modifications of viral proteins, or by restricting cellular iron accumulation [27,28,29]. Additionally, CQ/HCQ exert indirect immunomodulatory activities by suppressing the production of pro-inflammatory cytokines and immune hyperactivation (Figure 2) [30]. Notably, the effectiveness of CQ/HCQ analogs has been investigated in emerging and re-emerging viruses such as HIV [31,32], dengue and hepatitis C [33,34], human and avian influenza [35,36], Chikungunya virus (CHIKV) [37], Ebola and Marburg viruses [38,39], and severe acute respiratory syndrome (SARS) and Middle-East respiratory syndrome (MERS) coronaviruses [40,41], all of them representing huge challenges to human and veterinary medicine nowadays.
The current fight against SARS-CoV-2 has not been an exception, and the repositioning of CQ and HCQ as promising weapons to combat COVID-19 was rapidly investigated, partially supported by the enormous experience of CQ/HCQ use in malaria chemoprophylaxis, excellent safety and tolerability profile, very low cost and their in vitro broad-antiviral properties.

2.1.1. CQ and HCQ against SARS-CoV-2: In Silico and In Vitro Studies

Shortly after the COVID-19 outbreak and in the absence of any available pharmacological treatment and/or vaccine, one of the first published reports showed that SARS-CoV-2 infection in the common African green monkey kidney-derived Vero E6 was potently impaired by CQ treatment at low micromolar concentrations, and also suggested that therapeutic doses might be clinically achievable [42]. The same group found that HCQ was also effective in inhibiting SARS-CoV-2 infection in vitro, and more potently blocked the transport of virions into late endolysosomes (membrane-bound vesicles in the endocytic pathway) (Figure 2) [43], which was rapidly corroborated by another report [44].
Contrarily, in July 2020, a study by Hoffmann et al. compared the inhibition by CQ and HCQ of SARS-CoV-2 entry into Vero cells, Vero cells expressing TMPRSS2 (a serine protease that activates SARS-CoV-2 for entry into lung cells [45] and is essential for MERS-CoV pathogenesis in infected murine models [46]), and Calu-3 cells, airway epithelial cells that naturally express TMPRSS2 [45]. The report identified that CQ/HCQ failed to block infection with SARS-CoV-2 in TMPRSS2-Vero and Calu-3, indicating both an unlikely antiviral activity in human lung tissue and protection against COVID-19 [47].
Since CQ and HCQ are weak bases that preferentially accumulate in acidic organelles (endolysosomes and Golgi apparatus) [48], both elevate the pH of the acidic lumen and prevent the release of the viral genome into the host cytoplasm (Figure 2), as previously seen for human and avian influenza A viruses [49]. Similarly, the anti-SARS-CoV-2 lysosomotropic effects of CQ were confirmed in two human cell lines including Huh-7 (human hepatocarcinoma cells) and 293T-hACE2 (human kidney cells), showing that CQ (40 μM) markedly reduced viral replication [50]. Conversely, another report found that increasing concentrations of CQ (10, 50 and 100 μM) minimally altered the endosomal pH of human gastric epithelial cells [51]. Moreover, CQ induced Golgi-deacidification and affected post-translational modifications including glycosylation of SARS-CoV virions in vitro [40]. It is possible that another not-yet-confirmed mechanism of CQ/HCQ against SARS-CoV-2 might include the impairment of post-translational glycosylation of viral proteins, such as the Spike. Besides, CQ/HCQ can also block the uptake of SARS-CoV-2 virions by affecting the glycosylation of the ACE2 receptor in the plasma membrane (Figure 2) [43].
Considering that CQ reduces the expression of phosphatidylinositol binding clathrin assembly protein (PICALM), a clathrin adaptor essential for clathrin-mediated endocytosis [52] and blocks the endocytosis of nanoparticles in a clathrin-dependent manner [53,54], it is thus tempting to speculate that CQ might also interfere with this process during SARS-CoV-2 entry into host cells (Figure 2). However, clear evidence is still missing.
To speed up the anti-SARS-CoV-2 drug discovery pipeline, in silico bioinformatic molecular docking has been applied to find undisclosed CQ/HCQ cellular targets and to assess the binding affinities of both drugs with viral and host proteins, including: the viral protease (MPro) and host cathepsin L (CTSL), as the main proteolytic systems involved in the viral Spike protein activation [55,56]; and the RBD of the Spike protein, allowing virus entry and replication in host cells [45,57]. Results showed that HCQ achieved better interactions and affinity with ACE2 and MPro, whilst CQ achieved better results with MPro and CTSL, probably due to structural differences between the two drugs (Figure 2) [58]. In addition, Amin et al. conducted an in silico study and further demonstrated that HCQ exhibited improved binding affinities than CQ to the viral NTD-N-protein [59].
Furthermore, as a β-coronavirus, SARS-CoV-2 relies on glycoproteins and sialylated gangliosides as main attack sites of the respiratory epithelium [60]. Through molecular and structural modelling, Fantini et al. identified a ganglioside-binding site (GBS) in the N-terminal domain of the Spike glycoprotein, that may improve viral attachment to lipid rafts facilitating the interaction with the ACE2 receptor [61]. Interestingly, CQ and HCQ emerged as potential blockers of the initial S–ganglioside binding to the surface of respiratory cells (Figure 2), supporting the use of both drugs as initial therapy in infected individuals [61]. Remarkably, through in silico combination of drug-gene interaction networks, molecular docking and virus-host-drug interactome mapping [62], an unknown strong binding affinity of CQ/HCQ to TLR9 and IL-6 (components of innate immune response) was seen. Moreover, TLR9 and IL-6 were identified as targets of several SARS-CoV-2 proteins (Figure 2) [62]. Given that inhibition of TLR signaling is a promising option for COVID-19 therapeutics [63], the use of CQ/HCQ as TLR-dependent inhibitors is of significant importance.
Interestingly, in silico docking and dynamics studies have recently identified ACE2 receptor as well as novel HCQ targets including the α7 nicotinic AcetylCholine Receptor (α7 nAChR), α1D-adrenergic receptor (α1D-AR), Histamine N-Methyl Transferase (HNMT) and DNA gyrase/Topoisomerase III β (Top3β) [64]. In particular, the authors found that HCQ would block virus-binding sites on both ACE2 and α7 nAChR at the entry stage, whereas at post-entry stages, HCQ would prevent viral replication by acting against Top3β, and the “cytokine storm” by inhibiting α1D-AR.
In January 2021, Doharey et al. docking analyses identified CQ/HCQ as direct inhibitors of SARS-CoV-2 RNA-dependent RNA polymerase (RdRp), a key viral protein for RNA replication and transcription, and thus an important target for the development of antiviral drugs (Figure 2) [65]. In this regard, the authors found that CQ/HCQ showed the highest affinity towards SARS-CoV-2-RdRp, binding to its active site in the same manner as its substrate ATP binds [65].
Overall, in silico computational analyses have highlighted the potential of CQ and HCQ to interfere with SARS-CoV-2 infection at different layers, and support additional studies on the use of both drugs against COVID-19.

2.1.2. Preclinical In Vivo Use of CQ and HCQ

On 28 March 2020, the urgent need for an effective treatment against COVID-19 prompted the FDA to issue an emergency authorization to prescribe CQ and HCQ for COVID-19 patients despite the contrasting in vitro data and lack of preclinical evidence of their in vivo efficacy [66].
In May 2020, interesting data were reported on the therapeutic potential of HCQ in a ferret infection model [67], which recapitulates aspects of human SARS-CoV-2 infection and transmission [68]. In particular, HCQ-treated ferrets exhibited lower clinical scores compared to non-treated animals, whereas virus titers in nasal washes, stool specimens and respiratory tissues were similar among both groups. Two months later, the antiviral efficacy of HCQ was evaluated in SARS-CoV-2-infected cynomolgus macaques [69], a relevant model for the analysis of the early stages of SARS-CoV-2 infection in humans [70]. Differently from the in vitro data, HCQ had no significant effect on viral load, despite the high concentrations in the blood and lungs of infected animals [69]. Even more, when the drug was used as a prophylactic treatment, HCQ did not confer protection against SARS-CoV-2 infection. Consistent with these findings, in October 2020, standard human malaria HCQ prophylaxis and treatment did not benefit clinical outcome nor reduce SARS-CoV-2 titers in the upper and respiratory tract in the rhesus macaque disease model and in infected Syrian hamsters [71]. Moreover, Kaptein et al. added more negative results about the inexistent antiviral activity of HCQ in SARS-CoV-2-infected hamsters [72], suggesting no scientific basis for the use of HCQ in COVID-19 patients [72]. More recently, it was reported that HCQ and azithromycin (AZ, an antibiotic with broad-spectrum antiviral activity), alone or in combination, did not block SARS-CoV-2 replication in primary human bronchial airway epithelia, displaying no significant effect on viral replication, clinical course and lung impairment when tested in the Syrian hamster model [73].
Concerning CQ, a study searching for inhibitors of endosomal acidification to suppress SARS-CoV-2 replication and infection in vivo, showed that CQ reduced viral replication in the lungs and alleviated pneumonia reducing inflammation and infiltration in hACE2 transgenic mice [50,74].
Overall, only few reports have assessed the preclinical efficacy of CQ/HCQ against COVID-19 so far. It is thus tempting to speculate that in the absence of such an urgency of therapeutic options and vaccines against COVID-19, more preclinical studies would have possibly warned against the use of CQ/HCQ during SARS-CoV-2 infection in vivo, likely discouraging its further testing in human clinical studies.

2.1.3. CQ and HCQ in Clinical Trials

As early as in March 2020, 15 clinical trials had been already registered in the Chinese Clinical Trials Registry (ChiCRT) to test the efficacy and safety of CQ and HCQ in the treatment of COVID-19 associated pneumonia [75]. First published results from those multicenter trials showed apparent efficacy and acceptable safety against COVID-19, which encouraged the authors to further recommend the use of CQ/HCQ to treat larger infected populations [75].
In contrast, a randomized CloroCOVID-19 clinical trial aiming at comparing high vs. low dosage of CQ showed more cardiac toxicity and lethality in the high dosage arm, indicating that such dosage should be avoided for the treatment of severe COVID-19 [76]. Subsequently, in the attempt to assess the safety and benefit of the administration of CQ/HCQ in combination with AZ, an international, observational registry of 96,032 COVID-19 patients, found that the use of CQ/HCQ in different regimens was associated with ventricular arrythmia and an increased hospital death rate with no evidence of benefit of their use. Amazingly, despite unproven efficacy, CQ/HCQ were rapidly praised as a potential miracle cure for COVID-19 in the early days of the pandemic by many leaders including the US ex-president Donald Trump, circumstances that further pushed its use across the globe [77]. Indeed, by May–June 2020, the negative findings reported by Mehra et al. stopped many clinical trials in their tracks. However, on 4 June (only two weeks after its initial publication), sudden concerns were raised about the reliability of the data and analyses conducted by Surgisphere Corporation, the healthcare analytics company involved in the study, forcing the authors to request the retraction of the article from The Lancet.
On 5 June 2020, a press release announced the results from one of the largest trials, the RECOVERY randomized clinical trial on HCQ, by stating that any clinical benefit was found in hospitalized patients with COVID-19, and that the HCQ arm of the study was immediately stopped [78]. In contrast, in July 2020, Gautret and collaborators published the results from a small open-label non-randomized clinical trial which showed that HCQ treatment was significantly associated with viral load reduction in COVID-19 patients, and a synergistic and positive effect of HCQ combined with AZ [79].
Moreover, HCQ failed to prevent illness compatible with COVID-19 or confirmed infection when used as high-risk or moderate-risk post exposure prophylaxis within 4 days after exposure [80]. In addition, Mitjà et al. reported the lack of efficacy of HCQ to prevent SARS-CoV-2 infection or symptomatic COVID-19 disease in healthy persons exposed to a PCR-positive case patient [81].
Conflicting and insufficient data on the effect of CQ/HCQ in terms of all-cause mortality, progression to severe disease, clinical symptoms, and upper respiratory virologic clearance with antigen testing are available as well [82,83]. From the contrasting findings published in February 2021 by the SOLIDARITY Trial Consortium, the World Health Organization concluded that HCQ has little or no effect on hospitalized COVID-19 patients according to mortality, initiation of ventilation and duration of hospital stay [84]. Besides, the lack of efficacy of HCQ and HCQ/AZ for outpatient treatment [85], the absence of benefit of early treatment with HCQ for decreasing hospitalization [86], or the increased risk of major cardiovascular events with no effects on viral clearance rates, were reported in outpatients with early and mild COVID-19 disease [87]. More recently, in March 2022, a double-blind, multicenter, randomized, controlled trial attempted to increase the statistical power by including 1372 patients that were randomly allocated to HCQ or placebo [88], and further showed that in outpatients with mild/moderate forms of COVID-19, treatment with HCQ did not reduce the risk of hospitalization [89].
Although not always properly investigated, the use CQ/HCQ as possible treatments against COVID-19 would require a careful examination of their pharmacokinetic properties, safety profile and potential drug interactions. Despite being relatively safe in the treatment of malaria and rheumatic diseases with standardized doses, CQ/HCQ can lead to dermatological changes or adverse reactions of the gastrointestinal tract (i.e., nausea, vomiting, diarrhea), whereas the most severe side effects may include cardiotoxicity, neuromyopathy of proximal muscles, and irreversible retinopathy, mainly during prolonged treatment or high doses [90]. Although rare, case reports have also described severe hypoglycemia in malaria patients treated with CQ/HCQ, as well as in individuals with lupus and other chronic diseases [91]. Both drugs are quickly and fully absorbed after oral administration, and about 50–60% of the CQ/HCQ in blood is bound to proteins [92]. After being metabolized in the liver, high drug concentrations are found in the cardiac tissue, lungs, kidneys, liver, skeletal muscle, skin, and the eye [93]. Indeed, CQ/HCQ pharmacologically active metabolites have been involved in the drug-induced cardiotoxicity [94] and pruritus, but to date no data are available on their performance in COVID-19. Concerning cardiotoxicity, several of the studies reported above reported the risk of serious cardiovascular adverse effects (i.e., QTc prolongation, ventricular arrhythmia, atrial fibrillation and cardiac arrest) in COVID-19 patients treated with HCQ/CQ monotherapy or HCQ/CQ + AZ, which highlights the real need of evaluating the potential benefit/harm balance of CQ/HCQ in the treatment of SARS-CoV-2 infection [95]. Moreover, pharmacodynamic drug–drug interactions (DDIs) of CQ/HCQ with other medications must also be taken into consideration. For example, co-administration of HCQ with several antiviral agents including lopinavir-ritonavir, darunavir-cobicistat, and acetazolamide, resulted in QT-interval prolongation, ventricular arrhythmias, and torsade de pointes [96]. In addition, interference of HCQ with darunavir-cobicistat and tocilizumab might also lead to psychiatric disorders, such as behavioral disturbances, psychosis, agitation, delirium, and aggression [96]. Moreover, in COVID-19 patients, the administration of HCQ with acetazolamide and the anticancer ibrutinib resulted in a tachyarrhythmia as DDI [97]. Given that around 12% of hospitalizations in oncology units are due to adverse effects of anticancer agents [98], potential drug interactions with CQ/HCQ should be cautiously monitored.
Currently, 96 clinical studies are registered to ClinicalTrials.gov with the terms “chloroquine” and “COVID-19”, 35 of them already active, while HCQ accounts for a total of 291 registered trials and 92 studies under investigation (Supplementary Materials Table S1) (accessed on 3 May 2022). The main clinical studies involving CQ and HCQ are summarized in Table 1.

2.2. Quinine and other Aryl-Aminoalcohols: In Vitro and In Vivo Studies

Aryl-aminoalcohol is a chemotype present in potent antimalarial drugs, including quinine (Q), mefloquine (MQ), lumefantrine, and halofantrine (Figure 1). Beyond their antimalarial activity, the antiviral effects of these molecules have been previously studied (reviewed in D’Alessandro et al. [15]).
Indeed, numerous manuscripts have been recently published on the possible anti-SARS-CoV-2 activity of these molecules, with a particular focus on Q and MQ. Although there is in vitro evidence that Q restricts SARS-CoV-2 infection, neither clinical nor preclinical data have been reported so far [102]. Latarissa et al. recently published a review on this topic [103]. Briefly, in silico studies indicate several possible MoA of Q, including the binding to the Lys353 residue in the peptide domain of the ACE2 [104], to the viral non-structural protein nsp12 [105], and to the SARS-CoV-2 main protease and the Spike glycoprotein (Figure 2) [106]. These computational binding affinities have been demonstrated in vitro, showing that Q exerts its antiviral activity against SARS-CoV-2 in several cell lines with IC50 values ranging from 10 μM to 60 μM [107,108].
Both in silico and in vitro tests [109,110] have been carried out to evaluate MQ efficacy against SARS-CoV-2, but studies reporting on in vivo infection models or clinical trials are still missing. Interestingly, all reports confirmed a good antiviral activity on different cell lines, although few studies attempted to understand its MoA. The most used cell line has been the Vero E6 with an IC50 ranging from 1.8 μM to 8.06 μM [111,112,113,114,115], and similar results were obtained on Caco-2 [116] and Calu cells [112] as well. MQ MoA seems to interfere with the viral load, resulting in a reduction of the cytopathic effects in vitro. An in silico study demonstrated that MQ and halofantrine bind effectively to the MPro [110]. Besides, a time-of-addition analysis performed in Vero E6/TMPRSS2 cells indicated that MQ inhibited viral entry early after viral attachment to the target cell, whereas a lower antiviral effect was observed post-entry [112]. Finally, the antiviral activity of MQ appears to be enhanced in combination with other antimalarials or with viral replication inhibitors [112,117]. MQ is usually well tolerated, but in people with active liver or thyroid disease it can cause adverse effects in the nervous and gastrointestinal systems [118,119].
Overall, despite numerous studies investigating the anti-SARS-CoV-2 effect of aryl-aminoalcohols, much work is still needed to confirm their beneficial effects against COVID-19 infection. Furthermore, the lack of in vivo and clinical trials likely suggests a difficult translation from in vitro to in vivo studies.

2.3. Artemisinin and Its Derivatives

Artemisinin (ART), also known as quinghao, is a sesquiterpene lactone with a unique peroxide structure deriving from the Chinese medicinal plant Artemisia annua L. (Figure 1) [120]. Several semi-synthetic derivative drugs have been obtained from ART: artemether, artemisone, artesunate (AS), artefenomel (OZ439) and artenimol or dihydroartemisinin (i.e., the active metabolite of ART). These drugs have been widely repurposed to treat several diseases including viral infections [121,122,123].

2.3.1. Artemisinin: In Vitro Studies

Several in silico studies have been performed to define the targets of ART both on the pathogen SARS-CoV-2 and on the host sides. Excellent affinities of ART have been demonstrated with both a close and an open conformation of the Spike glycoprotein and with one South African variant [124], with the RBD of the Spike (Figure 2) [125]. However, the energy of bond of ART to the Spike was weaker compared to other antimalarial drugs such as CQ or MQ [126,127]. Concerning MPro and thus a potential arrest of the viral replication (Figure 2), the variable binding energies obtained indicate that MPro is not an ART target [127,128,129,130,131,132], although the hybridization of ART with thymoquinone resulted in an amelioration of the affinity with the viral protease [133]. Additionally, relatively good affinities of ART to the papain-like protease [127] and the non-structural protein 1 (nsp1) [134] have also been reported (Figure 2).
Among the host’s targets, ART was found to discretely interact with ACE2, TMPRSS2 and neuropilin-1 receptors, and the Glucose Regulated Protein 78 (GRP78, an endoplasmic reticulum protein that translocates to the plasma membrane and facilitates viral entry through binding to the substrate binding domain of SARS-CoV-2) [124,132]. The ART-ACE2 interaction which appears to be strongly influenced by the polymorphisms of the host receptors, has also lowered its priority use [128].
Successive in vitro studies further showed that increasing doses of ART exhibited a reduced ability to prevent SARS-CoV-2 pseudovirus entry compared to that of HCQ on the HEK293T/ACE2 cell line [130], and its anti-SARS-CoV-2 activity was proved to be lower compared to Artemether and AS in a plaque-reduction assay in Vero E6 cells [135].
Notably, to mitigate the “cytokine storm” in COVID-19 patients, the immunosuppressive effects of ART proved to reduce the production of pro-inflammatory TNFα both from SARS-CoV-2 pseudovirus and LPS-treated THP-1 macrophages at 25 and 50 mM, together with a decrease in the production of CXCL8 from THP-1 pre-stimulated with the pseudovirus (Figure 2) [130].

2.3.2. Artemisinin: Clinical Trials

ART has also been tested in few clinical trials. An open label, prospective, multi-center, comparative and interventional study was conducted on 120 patients treated with cycles of 500 mg of ART/day (ARTIVeda™) as a dietary supplement to the different SoC, demonstrating a faster recovery when the dietary supplementation was performed in mild or moderate COVID-19 patients [136]. Details of the above-mentioned clinical trial are shown in Table 2.

2.3.3. Artemisinin Derivatives: Artefenomel (OZ439), Artemether, Artemisone, Artenimol (Dihydroartemisinin, DHA), AS

Beyond ART, the antiviral activity of its derivatives, AS, DHA and artemisone, has also been investigated in in silico studies concluding that only AS and DHA formed stable complexes with the Spike glycoprotein or the RBD (Figure 2) [137]. Instead, DHA, but not artemether, was docked with the RBD, confirming a quite good interaction [110,125]. On the other hand, focusing on inhibiting SARS-CoV-2 replication, several viral nsps were tested as hypothetical targets of the ART derivatives. Unlike AS and artefenomel, which showed a tight and stable interaction with the binding pocket of the MPro of SARS-CoV-2 (Figure 2) [138], artemether has been demonstrated once again to be the worse ART derivative [110]. Similarly, the computational binding of AS to the non-structural proteins Helicase (or nsp13), nsp10, nsp14 and nsp15 resulted to be much more favorable and stable compared to artemether [109]. Furthermore, a suitable interaction of AS with nsp1 was also found (Figure 2), including a structural topology change upon molecule binding [134]. In the same study AS and artemether were in silico docked to two viral structural proteins, the E and the N proteins, both important for the packaging of the viral genome (Figure 2) [109,139]. In contrast to ART, none of the literature examined for this review presented in silico studies on the interactions with host’s structures.
Side by side to computational in silico approaches, several in vitro investigations have been reported. AS, the best interacting molecule with SARS-CoV-2 proteins from computational studies, showed an EC50 ranging from 12.98 mM to 16.24 mM and a selectivity index (SI) from 5.10 to 7.84 calculated by RT-PCR in the supernatants of infected Vero E6 cells after 24 h [140,141]. Very potent activity of AS was also detected using Huh7.5 and A549-hACE2 cells [135]. Moreover, it reduced the expression of the viral N protein in the micromolar range (Figure 2) [140,141], and a time-of-addition assay established a post-entry stage activity by AS [135,141]. However, 30 mM AS failed to inhibit SARS-CoV-2 cytopathic effects on Vero E6 cells at 72 h post-infection and did not reduce virus replication in Calu-3 cells after 48 h of treatment [142]. Even with poor in silico evidence, DHA emerged as a promising antiviral molecule. Like AS, the EC50 of DHA by RT-PCR on infected Vero E6 cells after 24 h was 13.31 mM, but with a lower SI of 2.38 [140]. Instead, after 48 h the EC50 raised to 20.1 mM [102]. Again, DHA also reduced the viral N expression in the micromolar range (Figure 2) [140]. Likewise, artemether showed a complete inhibition of N expression at 6.25 mM, functioning both at entry and post-entry stages [141]. Moreover, the EC50 varied between 53 and 98 mg/mL on different cell types, including Vero E6, Huh7.5, A549-hACE2 lines [135]. Only one study investigated the in vitro efficacy of 30 μM OZ439 demonstrating a 1.5 log decrease of SARS-CoV-2 RNA load in Vero E6 cells, and a 1 log decrease in Calu-3 cells. Nevertheless, the discrete toxicity revealed by biomass analysis in Calu-3 discouraged its use [142].

2.3.4. AS: Clinical Trials

Among all the artemisinin derivatives, only two clinical trials evaluating the in vivo efficacy of AS are ongoing. In a Phase II randomized and double-blind clinical trial, 100 mg of AS administered once a day for 5 days will be tested and compared to placebo for its effects in reducing the length of hospital stay, and in shortening the time needed for a COVID-19 test to become negative (Supplementary Materials Table S2). Differently, the effects of AS injections 2.4 mg/kg/dose in addiction to local SoC on the hospital length of stay and mortality will be assessed in a phase IV, randomized and open label clinical trial (Supplementary Materials Table S2).

2.3.5. Artemisinin and Its Derivatives in Combination with Other Molecules: In Vitro Studies

Two in vitro studies and nine clinical trials are currently focused on the use of ART or its derivatives in combination with other drugs, including antimalarials and chemically different compounds.
Gendrot and colleagues assessed the anti-SARS-CoV-2 activity in Vero E6 of different drug combinations (AS—amodiaquine, artemether—lumefantrine, AS—pyronaridine and DHA—piperaquine), and reported the use of AS 250 mg with MQ 550 mg as the best tandem among all drugs tested [117]. Moreover, the same group also evaluated the possible synergistic effect of DHA when combined with methylene blue, which resulted, however, in an antagonistic effect [111].

2.3.6. Artemisinin and Its Derivatives in Combination with Other Molecules: Clinical Trials

Concerning the use of ART alone or in combination with other molecules (Supplementary Materials Table S2), only one clinical study has been completed to date, although the final results have not been published, yet. By evaluating the effect of an oral spray called ArtemiC (artemisinin, curcumin, frankicense and vitamin C) administered twice/day compared to placebo, excellent safety and efficacy profiles were found. In fact, such orally-administered ART used with the SoC or in combination with both SoC and OT-101 (Trabedersen, an orphan drug), is under investigation in a Phase II randomized and double-blind clinical study.
In addition to ART, the combination therapy of AS with other compounds has been also investigated (Supplementary Materials Table S2), including the use of AS and Pyramax (pyronaridine-AS) to ameliorate COVID-19 symptoms and reduce the viral load, and the use of pyronaridine-AS and the combination of AS and amodiaquine, also in the commercial form of Artecom®. Besides, a Phase III observational cohort study on malaria patients is comparing the effects of the administration of AS with pyronaridine versus another antimalarial combination treatment, artemether-lumefantrine. Finally, AS formulation with amodiaquine called Cospherunate is currently under evaluation when administered with AZ in the presence or absence of phytomedicines to treat symptomatic COVID-19 patients (Supplementary Materials Table S2).

2.4. Other Antimalarial Drugs: Atovaquone

Atovaquone is a hydroxynaphthoquinone synthetized during World War II due to the shortage of quinine. Nowadays it is used in a fixed-dose combination with Proguanil (Malarone) for the treatment of uncomplicated malaria or for prophylaxis in travelers [143]. It is also indicated for the treatment of severe pneumonia due to Pneumocystis jirovecii in immunocompromised patients [144]. In silico molecular docking studies have identified different proteins of SARS-CoV-2: the main protease [145], the B chain of the RBM [146], the NTD of the N protein [147] and the papain-like proteinase [148] as the most promising targets showing the lowest binding energies with atovaquone. Moreover, subsequent in vitro tests have showed a strong inhibition of the replication of different SARS-CoV-2 variants (alpha, beta and delta) in Vero E6 and Calu-3 cells [149]. Additionally, atovaquone was able to significantly reduced virus replication and infection in human lung cells, Vero cells and human hepatocytes [150,151]. However, concerning atovaquone’s ability to reduce virus production in airway epithelium cultures, inconsistent results have been obtained [149,150].
There are three registered clinical trials involving the use of atovaquone for COVID-19, but only one has been completed (ID NCT04456153). The results show that the addition of 1500 mg of atovaquone to the SoC twice a day, for 10 days, led to a reduction of SARS-CoV-2 load and to a better outcome in 60 patients, although not statistically significant compared to the placebo group.

3. Other Antiprotozoan Drugs: K777

In 2021, a cysteine protease inhibitor termed K777 (also known as SLV213) was reported to block SARS-CoV-2 infection in different human and monkey cell lines in vitro without cytotoxic effects [152]. K777 had previously been shown to be safe and efficacious in mice and dogs infected by Trypanosoma cruzi, the protozoan parasite which causes Chagas disease, a leading cause of death in Latin America [153,154]. As an antiparasitic agent, K777 inhibits cruzipain, the major cysteine protease of T. cruzi involved in parasite replication, host cell invasion, and subversion of the host immune response [155,156,157]. K777 also selectively inhibits the human cysteine protease cathepsin L (CTSL) (Figure 2), but not its SARS-CoV-2 counterparts papain-like and 3CL-like proteases [152]. CTSL functions by cleaving the SARS-CoV-2 Spike protein which in turn enhances virus entry. Indeed, elevated circulating levels of CTSL have been positively correlated with disease course and severity in COVID-19 patients [56]. Thus, the inhibition of CTSL by K777 may result in the loss of cathepsin L-mediated Spike processing, preventing viral infection. Indeed, K777 had already demonstrated its antiviral effects back in 2015, when Zhou et al. reported that K777 blocked the entry of SARS-CoV-1 and MERS pseudoviruses into Vero E6 and HEK293 cells, likely due to the inactivation of CTSL on cell surfaces and/or within endosomes [158].
A single, double blind, placebo-controlled clinical trial by Selva Therapeutics about the efficacy of K777 on clinical symptoms of COVID-19 is currently ongoing (ClinicalTrials.gov Identifier: NCT04843787).

4. Anthelmintic Drugs against SARS-CoV-2

4.1. Niclosamide

Niclosamide (NIC; 5-Chloro-N-(2-chloro-4-nitrophenyl)-2-hydroxybenzamide) (Figure 1) was originally developed in 1953 as a molluscicide to kill snails. Following its approval by the FDA in 1982, it was found to be an effective treatment for gastrointestinal parasitic infections caused by tapeworms in humans [159,160]. Its main MoA seems to be the blocking of glucose uptake, thus acting as an uncoupling agent for energy-generating oxidative phosphorylation, starving the worms of ATP and ultimately impacting the parasite’s pH homeostasis [159]. It also acts on other cellular signaling pathways such as Wnt/β-catenin, mTOR and JAK/STAT3. Remarkably, niclosamide has demonstrated broad antiviral effects against a wide number of different viruses such as SARS-CoV, MERS-CoV, Dengue and Zika virus, Japanese encephalitis virus, hepatitis C, Ebola, human rhinoviruses, Chikungunya virus, human adenovirus and Epstein–Barr virus [161,162]. As antiviral, niclosamide exploits the neutralization of the endo-lysosomal pH, which interferes with pH-dependent membrane fusion events, a critical step that restricts virus entry [161].

4.1.1. Niclosamide: In Vitro and In Vivo Studies

The anti-SARS-CoV-2 activity of niclosamide was reported early in July 2020, when a screening of a panel of 48 FDA-approved drugs previously preselected by an assay on SARS-CoV-2 identified niclosamide as one of the most relevant compounds in preventing the infection in Vero cells, exhibiting an IC50 value in the low micromolar range (0.28 µM) [113]. Against SARS-CoV-2, two main MoA have been considered: (i) the pH-dependent blockage of virus endocytosis; and (ii) the prevention of viral-specific autophagy through inhibition of the S-Phase kinase associated protein 2 (SKP2) (Figure 2). A third mechanism, that is, the direct inhibitory activity of niclosamide against ACE2 receptor has been proposed, but it requires further confirmation [163].
In subsequent works, using Vero E6 cells, it was however reported that niclosamide at 10.4 µM (IC50 values of 0.16) exhibited moderate virucidal activities, with a negligible reduction of viral inhibition [164]. Through molecular modeling and a virtual screening, the same authors demonstrated a strong binding affinity of the drug to MPro, which was further combined with a low binding affinity to the viral Spike glycoprotein, adding a new potential MoA of niclosamide against the virus (Figure 2) [164]. More recently, niclosamide (1 µM) was reported to inhibit the Spike-induced syncytia by suppressing the activation of TMEM16 proteins, a calcium-activated ion channel and a scramblase responsible for the exposure of phosphatidylserine on the cell surface (Figure 2) [165]. These results suggested that niclosamide could be used for the prevention of pneumocyte syncytia formation in the severe form of COVID-19 [165]. These observations were further confirmed in a study in which niclosamide was able to neutralize the pH of endosomes, block viral entry and rescue the cytopathic effects upon infection in a set of different cell lines [51].
These encouraging in vitro data prompted new investigations to overcome the problem of low absorption associated with niclosamide, by developing novel formulations that enable an effective delivery of the drug to the target tissue. Interestingly, the development of a Tween 60-coated niclosamide–montmorillonite (NIC-MMT) hybrid system based on the drug carrier MMT that possesses mucoadhesive properties, resulted in an increase of the oral bioavailability of niclosamide by more than a 1.6-fold [166]. Furthermore, using Vero E6 and ACE2-expressing lung epithelial cells, a cost-effective lipid nanoparticle formulation of niclosamide (nano NCM) that employs only the FDA-approved excipient demonstrated to be active at 0.595 nM (IC50) with a IC90 of 3.38 μM, while achieving a selectivity index (CC50/IC50 ratio) of 52, whereas the non-formulated niclosamide presented selectivity index of 464. The formulation resulted quite promising, due to its stability and the possibility to be prepared in various isotonic vehicles at neutral pH [167]. Later in 2021, universal prophylactic nasal throat sprays as early treatments against SARS-CoV-2 and its more contagious variants were investigated. In particular, a low dose, prophylactic solution of niclosamide (20 μM) at a nasally safe and acceptable pH of 7.96, and a throat spray of up to 300 μM at pH 9.19 were proposed as the simplest and potentially the most effective formulations from both an efficacy as well as manufacturing and distribution standpoints, since no cold chain would be then required [168].
Recently, the activity of niclosamide against several SARS-CoV-2 variants of concern has been investigated. The results demonstrated that niclosamide inhibited the replication of the SARS-CoV-2 original strain (Wuhan D614) in VeroE6 TMPRSS2 cells with an IC50 of 0.13 μM and IC90 of 0.16 μM, in accordance with previous studies. Importantly, niclosamide also blocked the replication of the SARS-CoV-2 D614G, B.1.1.7, B.1.351 and B.1.617.2 variants with an IC50 of 0.06 μM, 0.08 μM, 0.07 μM, and 0.08 μM, respectively, showing similar potency across the different strains compared to the original Wuhan D614 strain [169].

4.1.2. Niclosamide: Clinical Trials

A discrete number of clinical trials focused on the use of niclosamide as an effective COVID-19 treatment have been conducted in patients with different disease severities. No study results are available yet on the official website except for the study NCT04399356 (Table 3), where no significant differences in oropharyngeal clearance of SARS-CoV-2 at day 3 between placebo and niclosamide groups were reported [170]. The remaining 13 studies are summarized in Supplementary Table S3.

4.2. Nitazoxanide

Nitazoxanide (NTZ) or 2-(acetyloxy)-N-(5-nitro-2-thiazolyl) benzamide (Figure 1), was synthesized in the early 1970s from niclosamide [171] and approved by the FDA as a safe and effective oral antiprotozoal drug to treat intestinal parasitic infections caused by Cryptosporidium parvum and Giardia intestinalis both in adults and children [172,173]. In addition, NTZ and its active circulating metabolite tizoxanide (TIZ) also exhibit antimicrobial effects against bacteria including Mycobacterium tuberculosis [174,175]. In these cases, the main mechanism of action of NTZ appears to be the inhibition of pyruvate:ferredoxin oxidoreductase (crucial for anaerobic energy metabolism), along with the disruption of the microorganism’s pH homeostasis [176]. Remarkably, NTZ has shown promising activity as a broad-spectrum antiviral agent against RNA or DNA viruses in vitro, including different influenza strains, respiratory syncytial virus, parainfluenza, rotavirus, norovirus, hepatitis B and C, Dengue, yellow fever, Japanese encephalitis virus, HIV and MERS coronavirus [171,177]. Of note, both NTZ and TIZ have generally shown similar inhibitory activity against viruses in vitro [177].
In February 2020, Wang and colleagues confirmed that NTZ was able to inhibit the novel coronavirus at low micromolar concentrations (IC50  =  2.12 μM; CC50  >  35.53 μM), although the antiviral potency of the active metabolite TIZ was absent from the study [42]. In fact, only one preprint posted in December 2021 claimed that both NTZ and TIZ show similar antiviral results in Vero E6 and Caco-2 cells [178].
By April 2020, thanks to its good safety profile at approved doses along with its affordable price as a generic drug, NTZ was considered a potential treatment against COVID-19 [179]. However, concern over the unknown hepatorenal and cardiovascular effects as well as teratogenicity retarded its employment. In July 2020, based on a pathophysiological and pharmacological approach, an article complained about the “neglected” consideration of NTZ against SARS-CoV-2 infection, and recommended to initiate a clinical trial combining NTZ with AZ in early stages of COVID-19 [180]. Indeed, NTZ+AZ in mild and moderate COVID-19 patients with diarrhea (N = 20), showed a symptomatic improvement on the fifth day after treatment [181]. The same report also proposed through molecular docking that NTZ would bind to the viral ADP-ribose phosphatase domain (ADPRP or MacroD) within the non-structural nsp13 multidomain (Figure 2) [181]. Since MacroD is a key enzyme that enables the virus to bypass the host immunity [182], NTZ might provide effective antiviral therapeutics.
Considering pregnancy, in September 2020, a small prospective follow-up and report of COVID-19 cases including pregnant women (N = 20), acutely ill hospitalized (N = 5) and ambulatory patients (N = 16), found NTZ treatment to be useful against SARS-CoV-2 in an early intervention in critical conditions and during pregnancy [183]. Similarly, a cross-sectional study reported on the survival safety for COVID-19 in pregnant women treated with NTZ along with a good safety profile of the drug [184].
More recently, NTZ treatment for five days of adult patients with mild COVID-19 symptoms failed to resolve symptoms compared to placebo, although the same study also showed that early NTZ therapy was safe and significantly reduced viral load [185]. Next, the results of a randomized, double-blind pilot clinical trial comparing NTZ vs placebo for seven days, linked NTZ with improved clinical (reduced hospitalization), virologic (faster negative tests by qPCR) and lower inflammatory outcomes (ID NCT04348409) [186]. Moreover, the combination of NTZ, ribavirin and IVM plus Zinc compared to routine supportive treatment against COVID-19, led to a faster viral clearance from the nasopharynx than symptomatic therapy [187] (ID NCT04392427). Finally, evidence that oral NTZ may reduce the risk of severe illness for patients at high risk, and the time to sustained recovery for mild COVID-19 individuals has been reported (ID NCT04486313) [188].
From the mechanistic point of view, NTZ seems to impair SARS-CoV-2 Spike protein variants maturation and to induce the generation of viral protein modifications, hindering progeny virion infectivity and Spike-mediated pulmonary cell fusion (Figure 2) [189], which highlights the potential of NTZ to fight against SARS-CoV-2 infection.
As an immunomodulator, NTZ was able to suppress the production of IL-6 in vitro and in an animal model injected with lipopolysaccharide and thioglycolate [190], and to inhibit the production of other cytokines in peripheral blood mononuclear cells [191]. These results have been recently reproduced in COVID-19 patients (Figure 2) [186]. In addition, NTZ amplifies the pathways of type 1 interferons (IFN-Is) (Figure 2) [171,177], which are found quite low during severe COVID-19 immunopathology [192]. Importantly, in 2019, NTZ was proved to be a potent bronchodilator even under harsh conditions using maximally contracted airways or airways pretreated with a cytokine cocktail [193]. These findings might be of critical importance for COVID-19 patients, since NTZ might alleviate respiratory symptoms including in mechanically ventilated patients.

4.3. Ivermectin

Ivermectin (IVM) is a macrocyclic lactone made as an 80:20 mixture of two homologues, 22,23-dihydroavermectin B1a and 22,23-dihydroavermectin B1b, respectively [194]. It belongs to the avermectin family of natural compounds produced by the bacterium Streptomyces avermitilis, and was discovered as an anthelminthic agent in 1979 to treat various parasitic infections [195,196].
Although different MoA are reported for IVM, one of its main cellular activities involve the targeting of the importin alpha (IMPα) and the inhibition of normal nuclear transport [197], required for normal transcription and DNA replication [198]. IVM binds to IMPα, which prevents IMPα interaction with IMPβ and the subsequent recognition of a specific nuclear localization signal (NLS) on the cargo, thus blocking the nuclear transport.
In the last 10 years, IVM has been identified as an antiviral agent, both against RNA viruses such as HIV-1, influenza or flaviviruses, and against DNA viruses such as polyomaviruses and adenoviruses [197,199], by suppressing viral replication/maturation through the inhibition of viral proteins to be transported into and out of the nucleus [197,200]. Interestingly, the anti-SARS-CoV-2 effects of IVM seem to be linked to the inhibition of nuclear transport of viral proteins as well [201,202,203,204].

4.3.1. Ivermectin: In Silico and In Vitro Studies

During the first wave of the pandemic in Europe and America, the FDA approved the repurposing of IVM for the treatment of COVID-19. Strikingly, it was early reported that treating SARS-CoV-2-infected Vero-hSLAM cells with 5 µM of IVM 2 h post infection, induced a ~5000-fold reduction of viral RNA both inside the cells and in the cell supernatants, with an antiviral activity lasting for 48 h with no toxic effects [204].
To confirm the efficacy of IVM on SARS-CoV-2 replication, an in silico study conducted in June 2020 demonstrated that IVM may interfere with the virus attachment to the host’s ACE2 receptor (Figure 2) [205]. It was later confirmed that the 22,23-dihydroavermectin B1b homologue was able to interact with the ACE2′s RBD domain [206]. Through stable isotope labeling amino acids in cell culture (SILAC) quantitative proteomics, a wide-spectrum of antiviral effects of IVM was reported, since as much as 52 SARS-CoV-2 proteins were found to be altered after treatment with IVM [207]. In particular, it was found that IVM impairs viral replication through direct inhibition of the enzymatic activity of MPro [208,209,210,211] (Figure 2).
In vitro cell-based models cannot represent the in vivo dynamics of infection in the lung microenvironment mainly due to the lack of the corresponding immune response. To this regard, a modelling study that considered the potential immune responses, the dose and the time of administration of different drugs, identified IVM as the better drug with a better efficacy when administered immediately after disease positivity [212]. In contrast, a study conducted on Calu-3 cells that better represent the lung compartment, failed to report a reduction of SARS-CoV-2 viral load in the presence of a high IVM dose [213].
At the end of March 2021, other two in silico reports showed high binding affinities of IVM for the viral replicase, protease and RBD of SARS-CoV-2, and the human TMPRSS2 receptor (Figure 2), pushing the use of IVM as a candidate therapy against COVID-19 [214,215]. More recently, Gonzalez-Paz and colleagues evaluated for the first time the effectiveness of each homolog comprising IVM. With an elastic networks model and computational and biophysical approaches, the authors observed that 22,23-dihydroavermectin B1a exhibits high affinity for the IMPα and IMPβ subunits (Figure 2), while 22,23-dihydroavermectin B1b presents higher affinity for viral structures [216,217], especially with the SARS-CoV-2 RBD domain, further speculating that it might block the ACE2-RBD binding [206].
To increase efficacy and reduce the risk of resistance emergence, IVM was also tested in combination with other drugs. Indeed, an in vitro study on Vero E6 cells successfully demonstrated that IVM in combination with remdesivir enhanced their single antiviral activities, although it was warned that different routes of administration may modify the ability of the drugs to simultaneously reach therapeutic concentrations [218]. Similar conclusions were reached by Eweas et al. through molecular docking assays [219]. Lastly, an in vitro model of infection combined with confocal microscopy investigated the anti-SARS-CoV-2 effects of IVM in combination with atorvastatin (ATV), observing that both drugs blocked the activation of the NF-κB signaling pathway, modified gene expression of IMPα and Rho GTPase (targets of IVM and ATV, respectively), and finally suppressed IMPα accumulation in the nucleus, presenting both IVM and ATV as valid drugs against the virus [220].

4.3.2. Ivermectin: Preclinical Studies

An indirect proof of the in vivo efficacy of IVM against the novel coronavirus was first reported in November 2020, since IVM treatment succeeded in reducing the liver viral load of mouse hepatitis virus (MHV) in the murine model, a single-stranded RNA virus like SARS-CoV-2 [221]. Other important in vivo study in SARS-CoV-2 infected hamsters showed that despite no effects in the reduction of viral load in the respiratory tracts, IVM reduced the IL-6 and IL-10 ratio and induced the polarization of macrophages towards a M2 phenotype that favored a beneficial anti-inflammatory response, inferring a potential positive implication of IVM in the clinical condition of COVID-19 patients (Figure 2) [222].

4.3.3. Ivermectin: Retrospective, Observational Studies, and Clinical Trials

With such promising in silico, in vitro, and in vivo data, many clinical trials, observational trials, or retrospective studies, mainly conducted in Southern Hemisphere countries, have been performed to evaluate the potential role of IVM against COVID-19.

IVM Used as Monotherapy

Concerning the safety of IVM administration, old studies also reported a good tolerability of the drug at doses higher than those approved by the FDA for human use, and with repeated drug administrations, exhibiting no toxicity for the central nervous system [223], and during pregnancy [224]. However, in vivo studies showed that IVM may induce malformation in the fetus and, moreover, the combination with other drugs would increase the risk of such malformations [225]. It remains to be determined if IVM is safe in COVID-19 pregnant woman, mainly due to exclusion of pregnant woman in almost all trials performed so far [226].
Compared to in vitro studies, human pharmacokinetic analyses demonstrated that IVM would achieve lung concentrations over 10 times higher than the reported IC50 [227]. Interestingly, a significant early seronegativity was observed in patients receiving IVM for five days compared to placebo and the IVM-doxycycline groups in Bangladesh [228], a result that was further confirmed by African and Brazilian trials which reported a reduction in the viral load in a dose-dependent manner [229,230]. In addition, an IVM-mucoadhesive nanosuspension nasal spray showed effectiveness in mild COVID-19 patients with a faster viral clearance and a faster resolution of anosmia [231]. Despite these encouraging findings, by conducting a survey of the clinical data collected so far, there is a certain agreement that a single dose of IVM does not significantly reduce viral load. In contrast, if given early after the onset of symptoms, it helps recovering faster from anosmia and hyposmia, it prevents the progression of the disease and it improves the clinical outcome compared to untreated patients [232,233,234,235,236,237].
In different studies, however, no beneficial effects of IVM on mild or severe disease were observed, as the recovery time and the resolution of symptoms were not significantly different from untreated patients [238,239,240,241]. Contrasting results have been reported as well. An Indian clinical trial conducted by Shahbaznejad and colleagues showed that important clinical COVID-19 symptoms such as dyspnea, cough, and lymphopenia, were improved in patients treated with a single dose of IVM [242]. Even more, the administration of IVM within the first 3 days after recovery in an intensive care unit improved gastrointestinal symptoms and the number of ventilator-free days in COVID-19 patients affected by severe syndrome and subjected to ventilation [243]. Contrarily to these observations, no improvement in clinical and virological outcomes were observed among the IVM-treated compared to patients treated only with the SoC [244]. Regarding the hospitalization parameters, a reduced hospitalization trend in IVM-treated patients has been reported [245], whereas other studies did not observe such decrease in mild-moderate COVID-19 subjects [246,247]. Finally, the role of IVM in the recovery from long-COVID-19 was investigated in a retrospective study conducted in Argentina, showing that IVM induces a faster reduction of the symptoms [248].
With regard to mortality, IVM treatment seems to be associated with lower mortality rates in patients with severe pulmonary damage [249]. In a multi-center clinical trial, the mortality rate decreased by 15% only in hospitalized individuals receiving different doses of IVM and different administration routes, while those receiving HCQ did not [250]. Besides, an Egyptian study observed a decreased in IL-6 and IL-1 cytokines in COVID-19 patients treated with IVM, with a reduction in the mortality rate and the hospitalization time [251]. Conversely, studies conducted in Pakistan and China did not detect differences in mortality rate, inflammatory markers, time of hospitalization, time of resolution of symptoms and syndrome course in mild and severe COVID-19 subjects [252,253]. Likewise, an American prospective study on 286 patients treated with two doses of IVM found no benefits in hospitalization time, intensive care unit admission, intubation rate or mortality [254].

IVM Treatment in Combination with Other Drugs

To improve the effectiveness of IVM, the repurposing of this drug in combination with other drugs has been extensively tested. The first three studies were conducted in Bangladesh in the summer of 2020, demonstrating that mild-moderate COVID-19 patients receiving IVM in combination with doxycycline showed a faster negative conversion in PCR and significantly faster symptom resolution compared to the HCQ-AZ-treated group [255]. Moreover, IVM-doxycycline also resulted in a faster viral clearance [256] in severe COVID-19 patients [257], together with improvements in early recovery and prevention to progression to more serious disease [258]. In addition, in a case series of SARS-CoV-2 positive patients hospitalized in Pakistan, IVM combined with doxycycline, remdesivir, tocilizumab, enoxaparin sodium and steroids, emerged as potential new treatment options by reducing the disease-associated severity and recovery rate [259].
Besides, a retrospective case series performed in India in June 2020, evaluating the effect of IVM combined with atorvastatin or N-acetylcysteine, showed that 98.6% of patients successfully recovered, with a mortality rate lower than the national rate [260]. Furthermore, IVM in combination with AZ compared with a single IVM dose was more effective in reducing the duration of the symptoms [261]. Additionally, a small retrospective study upon a 10-day treatment schedule composed of IVM-AZ-Cholecalciferol enabled a significant decrease in the recovery time at early stages of COVID-19 [262]. Additionally, positive effects in preventing hospitalization and death were observed in Mexico, after a therapy with IVM, AZ, Montelukast, and acetylsalicylic acid [263]. Further, the combination of IVM, AZ, acetaminophen, and aspirin, given to mild COVID-19 patients with no respiratory failure, reduced the risk of hospitalization and mortality [264]. On the contrary, the combination of IVM and HCQ did not reduce the viral load nor the mortality rate [265,266].
Beyond dual combination therapies, a triple combination of IVM-HCQ-AZ resulted in a faster negative conversion compared to AZ-HCQ-treated patients [267], a positive effect on clinical recovery, reduction of positivity time, and community spread prevention [268], and an overall improvement of clinical parameters [269]. Moreover, different mixed therapies of IVM with nitazoxanide, ribavirin, and zinc supplement, led to earlier negative conversions in affected patients compared to the standard care therapy [187]. Finally, to prevent COVID-19-associated complications such as hypercoagulation, the combination of a higher approved dose of IVM with dexamethasone, enoxaparin injection and aspirin (I.D.E.A protocol) resulted in lower mortality rates and no hospitalization among mild COVID-19 patients [270].

Prophylactic Use of IVM

To avoid the spreading of the virus and to block its transmission, the prophylactic use of IVM has been investigated especially in healthcare workers, who are highly exposed to the risk of infection. During summer 2020, a protective role of IVM with very low and mild adverse effects was firstly reported: only 7.4% of asymptomatic contacts of SARS-CoV-2 positive patients that received IVM developed COVID-19 vs 58.4% of untreated subjects [271]. Preventive IVM administration was also tested on healthcare workers in India and in the Dominican Republic, with a reduction of the viral infection by 73% and 70%, respectively [271,272,273]. Consistent with these results, other observational studies found a positive involvement of IVM in preventing SARS-CoV-2 infection, since a single oral monthly dose for 4 months reduced the viral infection by ten-fold [274]. Moreover, two prophylactic doses of IVM in the following months after COVID-19 infection showed better results compared to a single IVM dose [275]. Even more, prophylactic IVM in combination with Iota-Carrageenan (a sulphated polysaccharide extracted from red seaweeds), suggested a preventive role of this combination therapy [276,277]. On the other hand, no prevention of viral infection was reported by a clinical trial conducted in Singapore, performed on 3037 COVID-19 negative migrant workers subjected to quarantine who were treated with prophylactic drug regimens, including IVM [278]. In Africa, remarkably, where IVM is massively administrated to combat non-related parasitic diseases, the reduced COVID-19 positive rates and deaths might be explained by prophylactic IVM use against SARS-CoV-2 infection [279,280].

Ivermectin: Summary

To date, 148 studies, 98 peer-reviewed, and 78 with results have been performed on the use of IVM against SARS-CoV-2 [281]. These studies present many limitations, including the small cohorts of enrolled patients, the different posology and the different disease stages at which IVM efficacy has been investigated. IVM has been officially adopted as an early COVID-19 treatment for 28% of the world population, especially in Indonesia, Bangladesh, Mexico, Egypt, Ukraine, and Venezuela [281]. Since other drugs have been concomitantly administrated with IVM, it is difficult to draw a clear conclusion about the real anti-SARS-CoV-2 activity of this antiparasitic drug. Moreover, no optimal dose has been established so far [282]. Even if some clinical trials are still ongoing (Supplementary Materials Table S4), the use of IVM as a legitimate therapeutic option to combat the novel SARS-CoV-2 infection has not yet been authorized or approved by the FDA, predominantly due to the absence of sufficient supportive data [283]. Table 4 contains available results from observational and clinical trials on the use of IVM as anti-COVID-19 therapeutics.

5. Conclusions

Over the past two years, great efforts have been made to find an effective treatment for COVID-19, and the use of antiparasitic drugs rapidly appeared as one of the most immediate solutions, based on their previously reported antiviral activities. Unfortunately, despite the promising in vitro results, the anti-SARS-CoV-2 activity of CQ and HCQ failed to be translated into clinical efficacy, as documented by several reviews and the randomized clinical trials [287]. In the case of CQ, disparity between laboratory and clinical experiments could be mainly attributed to complex pharmacokinetics, specifically its distribution time into different tissues and its prolonged elimination half-life, which has made it difficult to extrapolate drug concentrations in culture media to human doses. Prophylactic activity of HCQ was explored as well, although dosing errors and the lack of consideration to patients with preexisting conditions were shown to be present in many reported trials. It is important to note that the use of HCQ as a prophylactic drug for COVID-19 must unambiguously include an extensive investigation of its adverse events, even the mild ones, such as the observed gastrointestinal disorders, along with the costs and hassle of undergoing the administration of a preventive medication. Currently, the potential benefits associated with this prophylactic use do not overcome the arising problematic issues. Based on these considerations, we feel quite confident affirming that HCQ would not be considered as a possible alternative to the ongoing vaccination campaign to prevent COVID-19.
A fundamental support to improve and accelerate the drug repurposing strategy might derive from the use of Artificial Intelligence (AI), a powerful approach that has been widely exploited in the sequencing of SARS-CoV-2 genome from the beginning of the pandemic (reviewed in [288]). In brief, AI consists of creating synergy between the structure-based, the ligand-based screening methods and the AI algorithms, to generate accurate prediction models. A clear example has been the identification of the anti-SARS-CoV-2 properties of remdevisir, atazanavir and efavirenz, which emerged in a deep learning-based drug-target interaction prediction model, known as “Molecule Transformer-Drug Target Interaction (MT-DTI) [289]. Another subdiscipline of AI is Machine Learning (ML), which able to reveal the connections between drugs, viral and host proteins, and was successfully applied to identify Baricitinib and Rifampicin as COVID-19 treatments [290]. However, despite the many advantages of AI-based methods in drug repurposing, there is a weakness that needs to be considered: AI requires large datasets, which are not always easily accessible, structured and standardized. In addition, all the candidate drugs identified by AI need to be equally validated through classic in vitro methods.
The gap between in vitro anti-SARS-CoV-2 evidence and the transferability to clinical practice was even wider when considering ivermectin. Although the failure to repurpose antiparasitic drugs against COVID-19 can be a cause for disappointment and discouragement, on the contrary, it is necessary to draw lessons to be able to carefully define potential new treatments. As stated by Ho and colleagues in a recent review [291], future studies aiming to repurpose well-known drugs for fighting emerging infections should be based on a more accurate design of the in vitro antiviral studies, as well as on improving the validation of viral targets and on in-depth studies of the translatability from the laboratory to the clinic.
Thus, as often occurs in the biological and pharmaceutical fields, innovation is widely encouraged and must be strictly integrated with the more classic methodologies, to balance benefits and costs and to obtain solid and reliable results in the fastest way possible.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/microorganisms10071284/s1, Table S1: CQ and/or HCQ clinical trials without available results. Table S2: Clinical trials on the use of Artemisinin and derivatives without available results. Table S3: Clinical trials involving Niclosamide as antiviral agent for COVID-19 patients’ treatment without published results. Table S4: IVM clinical studies without results posted and/or published.

Author Contributions

Conceptualization, E.C.-A., S.D. (Serena Delbue), N.B., P.F., D.T. and S.P.; Methodology, E.C.-A., M.D., L.S., F.P., L.D. and S.D. (Sarah D’Alessandro); Writing—Original Draft Preparation, E.C.-A., M.D., L.S., F.P. and L.D.; Writing—Review & Editing, E.C.-A., S.D. (Serena Delbue), S.P., N.B. and S.D. (Sarah D’Alessandro); Supervision, S.D. (Serena Delbue) and E.C.-A.; Funding Acquisition, P.F., S.D. (Serena Delbue) and N.B. All authors have read and agreed to the published version of the manuscript.

Funding

The research was partially supported by Grant “PSR2020” from University of Milan to S.D. and N.B., and by the Italian Ministry of Education, Universities and Research PRIN2017 Prot. 2017KM79NN to S.D. and P.F.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Acknowledgments

We would like to thank Rosalia Ticozzi for the daily effort in supporting the laboratory activities.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Hartenian, E.; Nandakumar, D.; Lari, A.; Ly, M.; Tucker, J.M.; Glaunsinger, B.A. The Molecular Virology of Coronaviruses. J. Biol. Chem. 2020, 295, 12910. [Google Scholar] [CrossRef] [PubMed]
  2. Naqvi, A.A.T.; Fatima, K.; Mohammad, T.; Fatima, U.; Singh, I.K.; Singh, A.; Atif, S.M.; Hariprasad, G.; Hasan, G.M.; Hassan, M.I. Insights into SARS-CoV-2 Genome, Structure, Evolution, Pathogenesis and Therapies: Structural Genomics Approach. Biochim. Biophys. Acta-Mol. Basis Disease 2020, 1866. [Google Scholar] [CrossRef] [PubMed]
  3. Li, W.; Moore, M.J.; Vasilieva, N.; Sui, J.; Wong, S.K.; Berne, M.A.; Somasundaran, M.; Sullivan, J.L.; Luzuriaga, K.; Greenough, T.C.; et al. Angiotensin-Converting Enzyme 2 Is a Functional Receptor For the SARS Coronavirus. Nature 2003, 426, 450–454. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. V’kovski, P.; Kratzel, A.; Steiner, S.; Stalder, H.; Thiel, V. Coronavirus Biology and Replication: Implications for SARS-CoV-2. Nat. Rev. Microbiol. 2021, 19, 155–170. [Google Scholar] [CrossRef] [PubMed]
  5. Kuo, L.; Masters, P.S. Functional Analysis of the Murine Coronavirus Genomic RNA Packaging Signal. J. Virol. 2013, 87, 5182–5192. [Google Scholar] [CrossRef] [Green Version]
  6. Zhang, J.J.; Dong, X.; Cao, Y.Y.; Yuan, Y.D.; Yang, Y.B.; Yan, Y.Q.; Akdis, C.A.; Gao, Y.D. Clinical Characteristics of 140 Patients Infected with SARS-CoV-2 in Wuhan, China. Allergy Eur. J. Allergy Clin. Immunol. 2020, 75, 1730–1741. [Google Scholar] [CrossRef]
  7. Jin, Y.; Yang, H.; Ji, W.; Wu, W.; Chen, S.; Zhang, W.; Duan, G. Virology, Epidemiology, Pathogenesis, and Control of COVID-19. Viruses 2020, 12, 372. [Google Scholar] [CrossRef] [Green Version]
  8. Ge, H.; Wang, X.; Yuan, X.; Xiao, G.; Wang, C.; Deng, T.; Yuan, Q.; Xiao, X. The Epidemiology and Clinical Information about COVID-19. Eur. J. Clin. Microbiol. Infect. Dis. 2020, 39, 1011–1019. [Google Scholar] [CrossRef]
  9. Coperchini, F.; Chiovato, L.; Croce, L.; Magri, F.; Rotondi, M. The Cytokine Storm in COVID-19: An Overview of the Involvement of the Chemokine/Chemokine-Receptor System. Cytokine Growth Factor Rev. 2020, 53, 25–32. [Google Scholar] [CrossRef]
  10. Fajgenbaum, D.C.; June, C.H. Cytokine Storm. N. Engl. J. Med. 2020, 7898, 2255–2273. [Google Scholar] [CrossRef]
  11. Wang, X.; Sacramento, C.Q.; Jockusch, S.; Chaves, O.A.; Tao, C.; Fintelman-Rodrigues, N.; Chien, M.; Temerozo, J.R.; Li, X.; Kumar, S.; et al. Combination of Antiviral Drugs Inhibits SARS-CoV-2 Polymerase and Exonuclease and Demonstrates COVID-19 Therapeutic Potential in Viral Cell Culture. Commun. Biol. 2022, 5, 154. [Google Scholar] [CrossRef] [PubMed]
  12. Endo, T.; Takemae, H.; Sharma, I.; Furuya, T. Multipurpose Drugs Active Against Both Plasmodium Spp. and Microorganisms: Potential Application for New Drug Development. Front. Cell. Infect. Microbiol. 2021, 11, 797509. [Google Scholar] [CrossRef] [PubMed]
  13. Pushpakom, S.; Iorio, F.; Eyers, P.A.; Jane Escott, K.; Hopper, S.; Wells, A.; Doig, A.; Guilliams, T.; Latimer, J.; McNamee, C.; et al. Drug Repurposing: Progress, Challenges and Recommendations. Nat. Rev. Drug Discov. 2019, 18, 41–58. [Google Scholar] [CrossRef] [PubMed]
  14. Zeng, X.; Song, X.; Ma, T.; Pan, X.; Zhou, Y.; Hou, Y.; Zhang, Z.; Li, K.; Karypis, G.; Cheng, F. Repurpose Open Data to Discover Therapeutics for COVID-19 Using Deep Learning. J. Proteome Res. 2020, 19, 4624–4636. [Google Scholar] [CrossRef]
  15. D’Alessandro, S.; Scaccabarozzi, D.; Signorini, L.; Perego, F.; Ilboudo, D.P.; Ferrante, P.; Delbue, S. The Use of Antimalarial Drugs against Viral Infection. Microorganisms 2020, 8, 85. [Google Scholar] [CrossRef] [Green Version]
  16. Dagen, M. History of Malaria and Its Treatment. Antimalarial Agents 2020, 1–48. [Google Scholar] [CrossRef]
  17. Christensen, S.B. Natural Products That Changed Society. Biomedicines 2021, 9, 472. [Google Scholar] [CrossRef]
  18. White, N.J. The Treatment of Malaria. N. Engl. J. Med. 1996, 335, 800–806. [Google Scholar] [CrossRef]
  19. McChesney, E.W. Animal Toxicity and Pharmacokinetics of Hydroxychloroquine Sulfate. Am. J. Med. 1983, 75, 11–18. [Google Scholar] [CrossRef]
  20. Al-Bari, M.A. Chloroquine Analogues in Drug Discovery: New Directions of Uses, Mechanisms of Actions and Toxic Manifestations from Malaria to Multifarious Diseases. J. Antimicrob. Chemother. 2015, 70, 1608–1621. [Google Scholar] [CrossRef] [Green Version]
  21. Plantone, D.; Koudriavtseva, T. Current and Future Use of Chloroquine and Hydroxychloroquine in Infectious, Immune, Neoplastic, and Neurological Diseases: A Mini-Review. Clin. Drug Investig. 2018, 38. [Google Scholar] [CrossRef] [PubMed]
  22. Landais, C.; Fenollar, F.; Thuny, F.; Raoult, D. From Acute Q Fever to Endocarditis: Serological Follow-Up Strategy. Clin. Infect. Dis. 2007, 44, 1337–1340. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Emonet, S.; Wuillemin, T.; Harbarth, S.; Wassilew, N.; Cikirikcioglu, M.; Schrenzel, J.; Lagier, J.C.; Raoult, D.; van Delden, C. Relapse of Tropheryma Whipplei Endocarditis Treated by Trimethoprim/Sulfamethoxazole, Cured by Hydroxychloroquine plus Doxycycline. Int. J. Infect. Dis. 2015, 30, 17–19. [Google Scholar] [CrossRef] [Green Version]
  24. Taramelli, D.; Tognazioli, C.; Ravagnani, F.; Leopardi, O.; Giannulis, G.; Boelaert, J.R. Inhibition of Intramacrophage Growth of Penicillium Marneffei by 4-Aminoquinolines. Antimicrob. Agents Chemother. 2001, 45, 1450–1455. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Henriet, S.S.; Jans, J.; Simonetti, E.; Kwon-Chung, K.J.; Rijs, A.J.; Hermans, P.W.; Holland, S.M.; de Jonge, M.I.; Warris, A. Chloroquine Modulates the Fungal Immune Response in Phagocytic Cells From Patients With Chronic Granulomatous Disease. J. Infect. Dis. 2013, 207, 1932–1939. [Google Scholar] [CrossRef] [PubMed]
  26. Dias-Melicio, L.A.; Moreira, A.P.; Calvi, S.A.; de Campos Soares, A.M.V. Chloroquine Inhibits Paracoccidioides Brasiliensis Survival within Human Monocytes by Limiting the Availability of Intracellular Iron. Microbiol. Immunol. 2006, 50, 307–314. [Google Scholar] [CrossRef] [PubMed]
  27. Al-Bari, M.A.A. Targeting Endosomal Acidification by Chloroquine Analogs as a Promising Strategy for the Treatment of Emerging Viral Diseases. Pharma Res. Per. 2017, 5, 293. [Google Scholar] [CrossRef]
  28. Daecke, J.; Fackler, O.T.; Dittmar, M.T.; Kräusslich, H.-G. Involvement of Clathrin-Mediated Endocytosis in Human Immunodeficiency Virus Type 1 Entry. J. Virol. 2005, 79, 1581–1594. [Google Scholar] [CrossRef] [Green Version]
  29. Boelaert, J.R.; Piette, J.; Sperber, K. The Potential Place of Chloroquine in the Treatment of HIV-1-Infected Patients. J. Clin. Virol. 2001, 20, 137–140. [Google Scholar] [CrossRef]
  30. Baize, S.; Leroy, E.M.; Georges, A.J.; Georges-Courbot, M.-C.; Capron, M.; Bedjabaga, I.; Lansoud-Soukate, J.; Mavoungou, E. Inflammatory Responses in Ebola Virus-Infected Patients. Clin. Exp. Immunol. 2002, 128, 163–168. [Google Scholar] [CrossRef]
  31. Murray, S.M.; Down, C.M.; Boulware, D.R.; Stauffer, W.M.; Cavert, W.P.; Schacker, T.W.; Brenchley, J.M.; Douek, D.C. Reduction of Immune Activation with Chloroquine Therapy during Chronic HIV Infection. J. Virol. 2010, 84, 12082–12086. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Romanelli, F.; Smith, K.M.; Hoven, A.D. Chloroquine and Hydroxychloroquine as Inhibitors of Human Immunodeficiency Virus (HIV-1) Activity. Curr. Pharm. Des. 2004, 10, 2643–2648. [Google Scholar] [CrossRef] [PubMed]
  33. Juvenal, K.; Farias, S.; Renata, P.; Machado, L.; Pereira, J.A.; Muniz, C.; Amaral Imbeloni, A.; Antô, B.; Lopes Da Fonseca, N. Antiviral Activity of Chloroquine Against Dengue Virus Type 2 Replication in Aotus Monkeys. Viral Immunol. 2015, 28, 161–169. [Google Scholar] [CrossRef] [Green Version]
  34. Mizui, T.; Shunhei, A.E.; Ae, Y.; Tanida, I.; Yoshiyuki, A.E.; Ae, T.; Ae, T.U.; Sakamoto, N.; Kenichi, A.E.; Ae, I.; et al. Inhibition of Hepatitis C Virus Replication by Chloroquine Targeting Virus-Associated Autophagy. J. Gastroenterol. 2010, 45, 195–203. [Google Scholar] [CrossRef]
  35. Loo, Y.; Xu, Y.; Cheung, Y.B.; Ooi, E.E.; Paton, N.I.; Lee, L.; Xu, Y.; Eong Ooi, E.; Bun Cheung, Y.; Archuleta, S.; et al. Chloroquine for Influenza Prevention: A Randomised, Double-Blind, Placebo Controlled Trial. Lancet Infect. Dis. 2011, 11, 677–683. [Google Scholar] [CrossRef]
  36. Jiang, K.; Li, Y.; Zhu, Q.; Xu, J.; Wang, Y.; Deng, W.; Liu, Q.; Zhang, G.; Meng, S. Pharmacological Modulation of Autophagy Enhances Newcastle Disease Virus-Mediated Oncolysis in Drug-Resistant Lung Cancer Cells. BMC Cancer 2014, 14, 551. [Google Scholar] [CrossRef] [Green Version]
  37. Roques, P.; Thiberville, S.-D.; Dupuis-Maguiraga, L.; Lum, F.-M.; Labadie, K.; Martinon, F.; Gras, G.; Lebon, P.; P Ng, L.F.; de Lamballerie, X.; et al. Paradoxical Effect of Chloroquine Treatment in Enhancing Chikungunya Virus Infection. Viruses 2018, 10, 268. [Google Scholar] [CrossRef] [Green Version]
  38. Long, J.; Wright, E.; Molesti, E.; Temperton, N.; Barclay, W. Antiviral Therapies against Ebola and Other Emerging Viral Diseases Using Existing Medicines That Block Virus Entry. F1000Res 2015. [Google Scholar] [CrossRef] [Green Version]
  39. Falzarano, D.; Safronetz, D.; Prescott, J.; Marzi, A.; Feldmann, F.; Feldmann, H. Lack of Protection Against Ebola Virus from Chloroquine in Mice and Hamsters. Emerg. Infect. Dis. 2015, 21, 1065. [Google Scholar] [CrossRef] [Green Version]
  40. Vincent, M.J.; Bergeron, E.; Benjannet, S.; Erickson, B.R.; Rollin, P.E.; Ksiazek, T.G.; Seidah, N.G.; Nichol, S.T. Chloroquine Is a Potent Inhibitor of SARS Coronavirus Infection and Spread. Virol. J. 2005, 2, 69. [Google Scholar] [CrossRef] [Green Version]
  41. De Wilde, A.H.; Jochmans, D.; Posthuma, C.C.; Zevenhoven-Dobbe, J.C.; van Nieuwkoop, S.; Bestebroer, T.M.; van den Hoogen, B.G.; Neyts, J.; Snijder, E.J. 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. 2014, 58, 4875. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Wang, M.; Cao, R.; Zhang, L.; Yang, X.; Liu, J.; Xu, M.; Shi, Z.; Hu, Z.; Zhong, W.; Xiao, G. Remdesivir and Chloroquine Effectively Inhibit the Recently Emerged Novel Coronavirus (2019-NCoV) in Vitro. Cell Res. 2020, 30, 269. [Google Scholar] [CrossRef] [PubMed]
  43. Liu, J.; Cao, R.; Xu, M.; Wang, X.; Zhang, H.; Hu, H.; Li, Y.; Hu, Z.; Zhong, W.; Wang, M. Hydroxychloroquine, a Less Toxic Derivative of Chloroquine, Is Effective in Inhibiting SARS-CoV-2 Infection in Vitro. Cell Discov. 2020, 6, 16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Yao, X.; Ye, F.; Zhang, M.; Cui, C.; Huang, B.; Niu, P.; Liu, X.; Zhao, L.; Dong, E.; Song, C.; et al. In Vitro Antiviral Activity and Projection of Optimized Dosing Design of Hydroxychloroquine for the Treatment of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). Clin. Infect. Dis. 2020, 71, 732. [Google Scholar] [CrossRef] [Green Version]
  45. Hoffmann, M.; Kleine-Weber, H.; Schroeder, S.; Krüger, N.; Herrler, T.; Erichsen, S.; Schiergens, T.S.; Herrler, G.; Wu, N.-H.; Nitsche, A.; et al. SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor. Cell 2020, 181, 271–280.e8. [Google Scholar] [CrossRef]
  46. Iwata-Yoshikawa, N.; Okamura, T.; Shimizu, Y.; Hasegawa, H.; Takeda, M.; Nagata, N. TMPRSS2 Contributes to Virus Spread and Immunopathology in the Airways of Murine Models after Coronavirus Infection. J. Virol. 2019, 93. [Google Scholar] [CrossRef] [Green Version]
  47. Hoffmann, M.; Mösbauer, K.; Hofmann-Winkler, H.; Kaul, A.; Kleine-Weber, H.; Krüger, N.; Gassen, N.C.; Müller, M.A.; Drosten, C.; Pöhlmann, S. Chloroquine Does Not Inhibit Infection of Human Lung Cells with SARS-CoV-2. Nature 2020, 585, 588. [Google Scholar] [CrossRef]
  48. Schlesinger, P.H.; Krogstad, D.J.; Herwaldt, B.L. Antimalarial Agents: Mechanisms of Action. Antimicrob. Agents Chemother. 1988, 32, 793. [Google Scholar] [CrossRef] [Green Version]
  49. Di Trani, L.; Savarino, A.; Campitelli, L.; Norelli, S.; Puzelli, S.; Vignolo, E.; Donatelli, I.; Cassone, A. Different PH Requirements Are Associated with Divergent Inhibitory Effects of Chloroquine on Human and Avian Influenza A Viruses. Virol. J. 2007, 4, 39. [Google Scholar] [CrossRef] [Green Version]
  50. Shang, C.; Zhuang, X.; Zhang, H.; Li, Y.; Zhu, Y.; Lu, J.; Ge, C.; Cong, J.; Li, T.; Tian, M.; et al. Inhibitors of Endosomal Acidification Suppress SARS-CoV-2 Replication and Relieve Viral Pneumonia in HACE2 Transgenic Mice. Virol. J. 2021, 18, 46. [Google Scholar] [CrossRef]
  51. Prabhakara, C.; Godbole, R.; Sil, P.; Jahnavi, S.; Gulzar, S.J.; van Zanten, T.S.; Sheth, D.; Subhash, N.; Chandra, A.; Shivaraj, A.; et al. Strategies to Target SARS-CoV-2 Entry and Infection Using Dual Mechanisms of Inhibition by Acidification Inhibitors. PLoS Pathog. 2021, 17, e1009706. [Google Scholar] [CrossRef] [PubMed]
  52. Bayati, A.; Kumar, R.; Francis, V.; McPherson, P.S. SARS-CoV-2 Infects Cells after Viral Entry via Clathrin-Mediated Endocytosis. J. Biol. Chem. 2021, 296, 100306. [Google Scholar] [CrossRef] [PubMed]
  53. Miller, S.E.; Mathiasen, S.; Bright, N.A.; Pierre, F.; Kelly, B.T.; Kladt, N.; Schauss, A.; Merrifield, C.J.; Stamou, D.; Höning, S.; et al. CALM Regulates Clathrin-Coated Vesicle Size and Maturation by Directly Sensing and Driving Membrane Curvature. Dev. Cell 2015, 33, 163. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Hu, T.Y.; Frieman, M.; Wolfram, J. Insights from Nanomedicine into Chloroquine Efficacy against COVID-19. Nat. Nanotechnol. 2020, 15, 247. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Simmons, G.; Bertram, S.; Glowacka, I.; Steffen, I.; Chaipan, C.; Agudelo, J.; Lu, K.; Rennekamp, A.J.; Hofmann, H.; Bates, P.; et al. Different Host Cell Proteases Activate the SARS-Coronavirus Spike-Protein for Cell-Cell and Virus-Cell Fusion. Virology 2011, 413, 265. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Zhao, M.-M.; Yang, W.-L.; Yang, F.-Y.; Zhang, L.; Huang, W.-J.; Hou, W.; Fan, C.-F.; Jin, R.-H.; Feng, Y.-M.; Wang, Y.-C.; et al. Cathepsin L Plays a Key Role in SARS-CoV-2 Infection in Humans and Humanized Mice and Is a Promising Target for New Drug Development. Signal Transduct. Target. 2021, 6, 134. [Google Scholar] [CrossRef]
  57. Yan, R.; Zhang, Y.; Li, Y.; Xia, L.; Guo, Y.; Zhou, Q. Structural Basis for the Recognition of SARS-CoV-2 by Full-Length Human ACE. Science 2020, 367, 1444. [Google Scholar] [CrossRef] [Green Version]
  58. Braz, H.L.B.; de Moraes Silveira, J.A.; Marinho, A.D.; de Moraes, M.E.A.; de Moraes Filho, M.O.; Monteiro, H.S.A.; Jorge, R.J.B. In Silico Study of Azithromycin, Chloroquine and Hydroxychloroquine and Their Potential Mechanisms of Action against SARS-CoV-2 Infection. Int. J. Antimicrob. Agents 2020, 56, 106119. [Google Scholar] [CrossRef]
  59. Amin, M.; Abbas, G. Docking Study of Chloroquine and Hydroxychloroquine Interaction with RNA Binding Domain of Nucleocapsid Phospho-Protein-an in Silico Insight into the Comparative Efficacy of Repurposing Antiviral Drugs. J. Biomol. Struct. Dyn. 2021, 39, 4243. [Google Scholar] [CrossRef]
  60. Matrosovich, M.; Herrler, G.; Klenk, H.D. Sialic Acid Receptors of Viruses. Top. Curr. Chem. 2015, 367, 1–28. [Google Scholar] [CrossRef]
  61. Fantini, J.; di Scala, C.; Chahinian, H.; Yahi, N. Structural and Molecular Modelling Studies Reveal a New Mechanism of Action of Chloroquine and Hydroxychloroquine against SARS-CoV-2 Infection. Int. J. Antimicrob. Agents 2020, 55, 105960. [Google Scholar] [CrossRef] [PubMed]
  62. Yu, W.; Bai, Y.; Raha, A.; Su, Z.; Geng, F. Integrative In Silico Investigation Reveals the Host-Virus Interactions in Repurposed Drugs Against SARS-CoV-2. Front. Bioinform. 2022, 1. [Google Scholar] [CrossRef]
  63. Sariol, A.; Perlman, S. SARS-CoV-2 Takes Its Toll. Nat. Immunol. 2021, 22, 801. [Google Scholar] [CrossRef] [PubMed]
  64. Navya, V.B.; Hosur, M.V. A Computational Study on Hydroxychloroquine Binding to Target Proteins Related to SARS-COV-2 Infection. Inform. Med. Unlocked 2021, 26, 100714. [Google Scholar] [CrossRef]
  65. Doharey, P.K.; Singh, V.; Gedda, M.R.; Sahoo, A.K.; Varadwaj, P.K.; Sharma, B. In Silico Study Indicates Antimalarials as Direct Inhibitors of SARS-CoV-2-RNA Dependent RNA Polymerase. J. Biomol. Struct. Dyn. 2021, 1–18. [Google Scholar] [CrossRef]
  66. Lenzer, J. COVID-19: US Gives Emergency Approval to Hydroxychloroquine despite Lack of Evidence. BMJ 2020, m1335. [Google Scholar] [CrossRef] [Green Version]
  67. Park, S.-J.; Yu, K.-M.; Kim, Y.-I.; Kim, S.-M.; Kim, E.-H.; Kim, S.-G.; Kim, E.J.; Casel, M.A.B.; Rollon, R.; Jang, S.-G.; et al. Antiviral Efficacies of FDA-Approved Drugs against SARS-CoV-2 Infection in Ferrets. mBio 2020, 11, 3. [Google Scholar] [CrossRef]
  68. Kim, Y.-I.; Kim, S.-G.; Kim, S.-M.; Kim, E.-H.; Park, S.-J.; Yu, K.-M.; Chang, J.-H.; Kim, E.J.; Lee, S.; Casel, M.A.B.; et al. Infection and Rapid Transmission of SARS-CoV-2 in Ferrets. Cell Host Microbe 2020, 27, 704–709.e2. [Google Scholar] [CrossRef]
  69. Maisonnasse, P.; Guedj, J.; Contreras, V.; Behillil, S.; Solas, C.; Marlin, R.; Naninck, T.; Pizzorno, A.; Lemaitre, J.; Gonçalves, A.; et al. Hydroxychloroquine Use against SARS-CoV-2 Infection in Non-Human Primates. Nature 2020, 585, 584. [Google Scholar] [CrossRef]
  70. Rockx, B.; Kuiken, T.; Herfst, S.; Bestebroer, T.; Lamers, M.M.; Oude Munnink, B.B.; de Meulder, D.; van Amerongen, G.; van den Brand, J.; Okba, N.M.A.; et al. Comparative Pathogenesis of COVID-19, MERS, and SARS in a Nonhuman Primate Model. Science 2020, 368, 1012. [Google Scholar] [CrossRef] [Green Version]
  71. Rosenke, K.; Jarvis, M.A.; Feldmann, F.; Schwarz, B.; Okumura, A.; Lovaglio, J.; Saturday, G.; Hanley, P.W.; Meade-White, K.; Williamson, B.N.; et al. Hydroxychloroquine Prophylaxis and Treatment Is Ineffective in Macaque and Hamster SARS-CoV-2 Disease Models. JCI Insight 2020, 5. [Google Scholar] [CrossRef] [PubMed]
  72. Kaptein, S.J.F.; Jacobs, S.; Langendries, L.; Seldeslachts, L.; ter Horst, S.; Liesenborghs, L.; Hens, B.; Vergote, V.; Heylen, E.; Barthelemy, K.; et al. Favipiravir at High Doses Has Potent Antiviral Activity in SARS-CoV-2-Infected Hamsters, Whereas Hydroxychloroquine Lacks Activity. Proc. Natl. Acad. Sci. USA 2020, 117, 26955. [Google Scholar] [CrossRef] [PubMed]
  73. Cochin, M.; Touret, F.; Driouich, J.-S.; Moureau, G.; Petit, P.-R.; Laprie, C.; Solas, C.; de Lamballerie, X.; Nougairède, A. Hydroxychloroquine and Azithromycin Used Alone or Combined Are Not Effective against SARS-CoV-2 Ex Vivo and in a Hamster Model. Antivir. Res. 2022, 197, 105212. [Google Scholar] [CrossRef] [PubMed]
  74. Bao, L.; Deng, W.; Huang, B.; Gao, H.; Liu, J.; Ren, L.; Wei, Q.; Yu, P.; Xu, Y.; Qi, F.; et al. The Pathogenicity of SARS-CoV-2 in HACE2 Transgenic Mice. Nature 2020, 583. [Google Scholar] [CrossRef]
  75. Gao, J.; Tian, Z.; Yang, X. Breakthrough: Chloroquine Phosphate Has Shown Apparent Efficacy in Treatment of COVID-19 Associated Pneumonia in Clinical Studies. BioSci. Trends 2020, 14, 72. [Google Scholar] [CrossRef] [Green Version]
  76. Borba, M.G.S.; Val, F.F.A.; Sampaio, V.S.; Alexandre, M.A.A.; Melo, G.C.; Brito, M.; Mourao, M.P.G.; Brito-Sousa, J.D.; Baia-da-Silva, D.; Guerra, M.V.F.; et al. 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, e208857. [Google Scholar] [CrossRef] [Green Version]
  77. Medical Association, A. Letters Internet Searches for Unproven COVID-19 Therapies in the United States. JAMA Intern. Med. 2020, 180, 1116. [Google Scholar] [CrossRef]
  78. Horby, P.; Mafham, M.; Linsell, L.; Phil, D.; Bell, J.L.; Staplin, N.; Emberson, J.-T.R.; Wiselka, M.; Ustianowski, A.; Elmahi, E.; et al. Effect of Hydroxychloroquine in Hospitalized Patients with COVID-19. N. Engl. J. Med. 2020, 21, 2030. [Google Scholar] [CrossRef]
  79. Gautret, P.; Lagier, J.C.; Parola, P.; Hoang, V.T.; Meddeb, L.; Mailhe, M.; Doudier, B.; Courjon, J.; Giordanengo, V.; Vieira, V.E.; et al. Hydroxychloroquine and Azithromycin as a Treatment of COVID-19: Results of an Open-Label Non-Randomized Clinical Trial. Int. J. Antimicrob. Agents 2020, 56, 105949. [Google Scholar] [CrossRef]
  80. Boulware, D.R.; Pullen, M.F.; Bangdiwala, A.S.; Pastick, K.A.; Lofgren, S.M.; Okafor, E.C.; Skipper, C.P.; Nascene, A.A.; Nicol, M.R.; Abassi, M.; et al. A Randomized Trial of Hydroxychloroquine as Postexposure Prophylaxis for COVID-19. N. Engl. J. Med. 2020, 383, 517. [Google Scholar] [CrossRef]
  81. Mitjà, O.; Corbacho-Monné, M.; Ubals, M.; Alemany, A.; Suñer, C.; Tebé, C.; Tobias, A.; Peñafiel, J.; Ballana, E.; Pérez, C.A.; et al. A Cluster-Randomized Trial of Hydroxychloroquine for Prevention of COVID-19. N. Engl. J. Med. 2021, 384, 417. [Google Scholar] [CrossRef] [PubMed]
  82. Hernandez, A.V.; Roman, Y.M.; Pasupuleti, V.; Barboza, J.J.; White, C.M. Hydroxychloroquine or Chloroquine for Treatment or Prophylaxis of COVID-19. Ann. Intern. Med. 2020, 173, 287. [Google Scholar] [CrossRef]
  83. Ip, A.; Ahn, J.; Zhou, Y.; Goy, A.H.; Hansen, E.; Pecora, A.L.; Sinclaire, B.A.; Bednarz, U.; Marafelias, M.; Sawczuk, I.S.; et al. Hydroxychloroquine in the Treatment of Outpatients with Mildly Symptomatic COVID-19: A Multi-Center Observational Study. BMC Infect. Dis 2021, 21, 72. [Google Scholar] [CrossRef] [PubMed]
  84. Pan, H.; Peto, R.; Henao-Restrepo, A.; Preziosi, M.; Sathi-yamoorthy, V.; Abdool Karim, Q.; Ale-jandria, M.; Hernández García, C.; Kie-ny, M.; Malekzadeh, R.; et al. Repurposed Antiviral Drugs for COVID-19—Interim WHO Solidarity Trial Results. N. Engl. J. Med. 2021, 384, 497. [Google Scholar] [CrossRef] [PubMed]
  85. Johnston, C.; Brown, E.R.; Stewart, J.; Karita, H.C.S.; Kissinger, P.J.; Dwyer, J.; Hosek, S.; Oyedele, T.; Paasche-Orlow, M.K.; Paolino, K.; et al. Hydroxychloroquine with or without Azithromycin for Treatment of Early SARS-CoV-2 Infection among High-Risk Outpatient Adults: A Randomized Clinical Trial. EClinicalMedicine 2021, 33, 100773. [Google Scholar] [CrossRef]
  86. Reis, G.; dos Santos Moreira Silva, E.A.; Medeiros Silva, D.C.; Thabane, L.; Singh, G.; Park, J.J.H.; Forrest, J.I.; Harari, O.; Quirino Dos Santos, C.V.; Guimarães de Almeida, A.P.F.; et al. Effect of Early Treatment With Hydroxychloroquine or Lopinavir and Ritonavir on Risk of Hospitalization Among Patients With COVID-19. The TOGETHER Randomized Clinical Trial. JAMA Netw. Open 2021. [Google Scholar] [CrossRef]
  87. Rodrigues, C.; Freitas-Santos, R.S.; Levi, J.E.; Senerchia, A.A.; Lopes, A.T.A.; Santos, S.R.; Siciliano, R.F.; Pierrotti, L.C. Hydroxychloroquine plus Azithromycin Early Treatment of Mild COVID-19 in an Outpatient Setting: A Randomized, Double-Blinded, Placebo-Controlled Clinical Trial Evaluating Viral Clearance. Int. J. Antimicrob. Agent 2021, 58, 106428. [Google Scholar] [CrossRef]
  88. Dubée, V.; Roy, P.-M.; Vielle, B.; Parot-Schinkel, E.; Blanchet, O.; Darsonval, A.; Lefeuvre, C.; Abbara, C.; Boucher, S.; Devaud, E.; et al. Hydroxychloroquine in Mild-to-Moderate Coronavirus Disease 2019: A Placebo-Controlled Double Blind Trial. Clin. Microbiol. Infect. 2021, 27, 1124. [Google Scholar] [CrossRef]
  89. Avezum, Á.; Oliveira, G.B.F.; Oliveira, H.; Lucchetta, R.C.; Pereira, V.F.A.; Dabarian, A.L.; D O Vieira, R.; Silva, D.V.; Kormann, A.P.M.; Tognon, A.P.; et al. Hydroxychloroquine versus Placebo in the Treatment of Non-Hospitalised Patients with COVID-19 (COPE-Coalition V): A Double-Blind, Multicentre, Randomised, Controlled Trial. Lancet Reg. Health Am. 2022, 11, 100243. [Google Scholar] [CrossRef]
  90. Braga, C.B.; Martins, A.C.; Cayotopa, A.D.E.; Klein, W.W.; Schlosser, A.R.; da Silva, A.F.; de Souza, M.N.; Andrade, B.W.B.; Filgueira-Júnior, J.A.; de Pinto, W.J.; et al. Side Effects of Chloroquine and Primaquine and Symptom Reduction in Malaria Endemic Area (Mâncio Lima, Acre, Brazil). Interdiscip. Perspect. Infect. Dis. 2015, 2015, 346853. [Google Scholar] [CrossRef]
  91. Juurlink, D.N. Safety Considerations with Chloroquine, Hydroxychloroquine and Azithromycin in the Management of SARS-CoV-2 Infection. Can. Med. Assoc. J. 2020, 192, E450–E453. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Augustijns, P.; Geusens, P.; Verbeke, N. Chloroquine Levels in Blood during Chronic Treatment of Patients with Rheumatoid Arthritis. Eur. J. Clin. Pharmacol. 1992, 42, 429. [Google Scholar] [CrossRef] [PubMed]
  93. Ducharme, J.; Farinotti, R. Clinical Pharmacokinetics and Metabolism of Chloroquine. Clin. Pharmacokinet. 1996, 31, 257. [Google Scholar] [CrossRef] [PubMed]
  94. Essien, E.E.; Ette, E.I. Effects of Chloroquine and Didesethylchloroquine on Rabbit Myocardium and Mitochondria. J. Pharm. Pharmacol. 1986, 38, 543. [Google Scholar] [CrossRef]
  95. Zhao, Y.; Zhang, J.; Zheng, K.; Thai, S.; Simpson, R.J.; Kinlaw, A.C.; Xu, Y.; Wei, J.; Cui, X.; Buse, J.B.; et al. Serious Cardiovascular Adverse Events Associated with Hydroxychloroquine/Chloroquine Alone or with Azithromycin in Patients with COVID-19: A Pharmacovigilance Analysis of the FDA Adverse Event Reporting System (FAERS). Drugs Real World Outcomes 2022, 9, 231. [Google Scholar] [CrossRef]
  96. Conti, V.; Sellitto, C.; Torsiello, M.; Manzo, V.; de Bellis, E.; Stefanelli, B.; Bertini, N.; Costantino, M.; Maci, C.; Raschi, E.; et al. Identification of Drug Interaction Adverse Events in Patients With COVID-19. JAMA Netw. Open 2022, 5, e227970. [Google Scholar] [CrossRef]
  97. Treon, S.P.; Castillo, J.J.; Skarbnik, A.P.; Soumerai, J.D.; Ghobrial, I.M.; Guerrera, M.L.; Meid, K.; Yang, G. The BTK Inhibitor Ibrutinib May Protect against Pulmonary Injury in COVID-19–Infected Patients. Blood 2020, 135, 1912. [Google Scholar] [CrossRef] [Green Version]
  98. Miranda, V.; Fede, A.; Nobuo, M.; Ayres, V.; Giglio, A.; Miranda, M.; Riechelmann, R.P. Adverse Drug Reactions and Drug Interactions as Causes of Hospital Admission in Oncology. J. Pain Symptom Manag. 2011, 42, 342. [Google Scholar] [CrossRef]
  99. Réa-Neto, Á.; Stradiotto Bernardelli, R.; Martins, B.; Câmara, D.; Reese, F.B.; Vinicius, M.; Queiroga, O.; Oliveira, M.C. An Open-Label Randomized Controlled Trial Evaluating the Efficacy of Chloroquine/Hydroxychloroquine in Severe COVID-19 Patients. Sci. Rep. 2021, 11, 9023. [Google Scholar] [CrossRef]
  100. Self, W.H.; Semler, M.W.; Leither, L.M.; Casey, J.D.; Angus, D.C.; Brower, R.G.; Chang, S.Y.; Collins, S.P.; Eppensteiner, J.C.; Filbin, M.R.; et al. Effect of Hydroxychloroquine on Clinical Status at 14 Days in Hospitalized Patients With COVID-19. JAMA 2020, 324, 2165. [Google Scholar] [CrossRef]
  101. Furtado, R.H.M.; Berwanger, O.; Fonseca, H.A.; Corrêa, T.D.; Ferraz, L.R.; Lapa, M.G.; Zampieri, F.G.; Veiga, V.C.; Azevedo, L.C.P.; Rosa, R.G.; et al. Azithromycin in Addition to Standard of Care versus Standard of Care Alone in the Treatment of Patients Admitted to the Hospital with Severe COVID-19 in Brazil (COALITION II): A Randomised Clinical Trial. Lancet 2020, 396, 959. [Google Scholar] [CrossRef]
  102. Gendrot, M.; Andreani, J.; Boxberger, M.; Jardot, P.; Fonta, I.; le Bideau, M.; Duflot, I.; Mosnier, J.; Rolland, C.; Bogreau, H.; et al. Antimalarial Drugs Inhibit the Replication of SARS-CoV-2: An in Vitro Evaluation. Travel. Med. Infect. Dis. 2020, 37, 101873. [Google Scholar] [CrossRef] [PubMed]
  103. Latarissa, I.R.; Barliana, M.I.; Meiliana, A.; Lestari, K. Potential of Quinine Sulfate for COVID-19 Treatment and Its Safety Profile: Review. Clin. Pharmacol. Adv. Appl. 2021, 13, 225. [Google Scholar] [CrossRef] [PubMed]
  104. Lestari, K.; Sitorus, T.; Megantara, S.; Levita, J. Molecular Docking of Quinine, Chloroquine and Hydroxychloroquine to Angiotensin Converting Enzyme 2 (ACE2) Receptor for Discovering New Potential COVID-19 Antidote. J. Adv. Pharm. Educ. Res. 2020, 10, 1–4. [Google Scholar]
  105. Sumitha, A.; Devi, P.B.; Hari, S.; Dhanasekaran, R. COVID-19—In Silico Structure Prediction and Molecular Docking Studies with Doxycycline and Quinine. Biomed. Pharmacol. J. 2020, 13, 1185. [Google Scholar] [CrossRef]
  106. Srimathi, R.; Mohan Maruga Raja, M.K.; Kathiravan, M.K. In Silico Screening of Traditional Herbal Medicine Derived Chemical Constituents for Possible Potential Inhibition against Sars-Cov-2. J. Nat. Remedies 2020, 20, 79. [Google Scholar] [CrossRef]
  107. Große, M.; Ruetalo, N.; Layer, M.; Hu, D.; Businger, R.; Rheber, S.; Setz, C.; Rauch, P.; Auth, J.; Fröba, M.; et al. Quinine Inhibits Infection of Human Cell Lines with Sars-Cov-2. Viruses 2021, 13, 647. [Google Scholar] [CrossRef]
  108. Persoons, L.; Vanderlinden, E.; Vangeel, L.; Wang, X.; Do, N.D.T.; Foo, S.Y.C.; Leyssen, P.; Neyts, J.; Jochmans, D.; Schols, D.; et al. Broad Spectrum Anti-Coronavirus Activity of a Series of Anti-Malaria Quinoline Analogues. Antivir. Res. 2021, 193. [Google Scholar] [CrossRef]
  109. Roy Chattopadhyay, N.; Chatterjee, K.; Banerjee, A.; Choudhuri, T. Combinatorial Therapeutic Trial Plans for COVID-19 Treatment Armed up with Antiviral, Antiparasitic, Cell-Entry Inhibitor, and Immune-Boosters. VirusDisease 2020, 31, 479. [Google Scholar] [CrossRef]
  110. Sachdeva, C.; Wadhwa, A.; Kumari, A.; Hussain, F.; Jha, P.; Kaushik, N.K. In Silico Potential of Approved Antimalarial Drugs for Repurposing against COVID-19. OMICS A J. Integr. Biol. 2020, 24, 568. [Google Scholar] [CrossRef]
  111. Gendrot, M.; Jardot, P.; Delandre, O.; Boxberger, M.; Andreani, J.; Duflot, I.; le Bideau, M.; Mosnier, J.; Fonta, I.; Hutter, S.; et al. In Vitro Evaluation of the Antiviral Activity of Methylene Blue Alone or in Combination against SARS-CoV-2. J. Clin. Med. 2021, 10, 3007. [Google Scholar] [CrossRef]
  112. Shionoya, K.; Yamasaki, M.; Iwanami, S.; Ito, Y.; Fukushi, S.; Ohashi, H.; Saso, W.; Tanaka, T.; Aoki, S.; Kuramochi, K.; et al. Mefloquine, a Potent Anti-Severe Acute Respiratory Syndrome-Related Coronavirus 2 (SARS-CoV-2) Drug as an Entry Inhibitor In Vitro. Front. Microbiol. 2021, 12. [Google Scholar] [CrossRef] [PubMed]
  113. Jeon, S.; Ko, M.; Lee, J.; Choi, I.; Byun, S.Y.; Park, S.; Shum, D.; Kim, S. Identification of Antiviral Drug Candidates against SARS-CoV-2 from FDA-Approved Drugs. Antimicrob. Agents Chemother. 2020, 64, e00819-20. [Google Scholar] [CrossRef] [PubMed]
  114. Weston, S.; Coleman, C.M.; Haupt, R.; Logue, J.; Matthews, K.; Li, Y.; Reyes, H.M.; Weiss, S.R.; Frieman, M.B. Broad Anti-Coronavirus Activity of Food and Drug Administration-Approved Drugs against SARS-CoV-2 In Vitro and SARS-CoV In Vivo. J. Virol. 2020, 94, e01218-20. [Google Scholar] [CrossRef] [PubMed]
  115. Jan, J.-T.; Cheng, T.-J.R.; Juang, Y.-P.; Ma, H.-H.; Wu, Y.-T.; Yang, W.-B.; Cheng, C.-W.; Chen, X.; Chou, T.-H.; Shie, J.-J.; et al. Identification of Existing Pharmaceuticals and Herbal Medicines as Inhibitors of SARS-CoV-2 Infection. Proc. Natl. Acad. Sci. USA 2021, 118, e2021579118. [Google Scholar] [CrossRef]
  116. Ellinger, B.; Bojkova, D.; Zaliani, A.; Cinatl, J.; Claussen, C.; Westhaus, S.; Keminer, O.; Reinshagen, J.; Kuzikov, M.; Wolf, M.; et al. A SARS-CoV-2 Cytopathicity Dataset Generated by High-Content Screening of a Large Drug Repurposing Collection. Sci. Data 2021, 8. [Google Scholar] [CrossRef]
  117. Gendrot, M.; Duflot, I.; Boxberger, M.; Delandre, O.; Jardot, P.; le Bideau, M.; Andreani, J.; Fonta, I.; Mosnier, J.; Rolland, C.; et al. Antimalarial Artemisinin-Based Combination Therapies (ACT) and COVID-19 in Africa: In Vitro Inhibition of SARS-CoV-2 Replication by Mefloquine-Artesunate. Int. J. Infect. Dis 2020, 99, 437. [Google Scholar] [CrossRef]
  118. Postasman, I.; Beny, A.; Seligmann, H. Neuropsychiatric Problems in 2500 Long-Term Young Travelers to the Tropics. J. Travel Med. 2000, 7, 5. [Google Scholar] [CrossRef] [Green Version]
  119. Croft, A.M.; Herxheimer, A. Adverse Effects of the Antimalaria Drug, Mefloquine: Due to Primary Liver Damage with Secondary Thyroid Involvement? BMC Public Health 2002, 2, 6. [Google Scholar] [CrossRef] [Green Version]
  120. Ma, N.; Zhang, Z.; Liao, F.; Jiang, T.; Tu, Y. The Birth of Artemisinin. Pharmacol. Ther. 2020, 216, 107658. [Google Scholar] [CrossRef]
  121. Qiu, F.; Liu, J.; Mo, X.; Liu, H.; Chen, Y.; Dai, Z. Immunoregulation by Artemisinin and Its Derivatives: A New Role for Old Antimalarial Drugs. Front. Immunol. 2021, 12. [Google Scholar] [CrossRef] [PubMed]
  122. Kiani, B.H.; Kayani, W.K.; Khayam, A.U.; Dilshad, E.; Ismail, H.; Mirza, B. Artemisinin and Its Derivatives: A Promising Cancer Therapy. Mol. Biol. Rep. 2020, 47, 6321. [Google Scholar] [CrossRef] [PubMed]
  123. Cheong, D.H.J.; Tan, D.W.S.; Wong, F.W.S.; Tran, T. Anti-Malarial Drug, Artemisinin and Its Derivatives for the Treatment of Respiratory Diseases. Pharmacol. Res. 2020, 158, 104901. [Google Scholar] [CrossRef] [PubMed]
  124. Rolta, R.; Salaria, D.; Sharma, P.P.; Sharma, B.; Kumar, V.; Rathi, B.; Verma, M.; Sourirajan, A.; Baumler, D.J.; Dev, K. Phytocompounds of Rheum Emodi, Thymus Serpyllum, and Artemisia Annua Inhibit Spike Protein of SARS-CoV-2 Binding to ACE2 Receptor: In Silico Approach. Curr. Pharmacol. Rep. 2021, 7, 135. [Google Scholar] [CrossRef]
  125. Ribaudo, G.; Coghi, P.; Yang, L.J.; Ng, J.P.L.; Mastinu, A.; Memo, M.; Wong, V.K.W.; Gianoncelli, A. Computational and Experimental Insights on the Interaction of Artemisinin, Dihydroartemisinin and Chloroquine with SARS-CoV-2 Spike Protein Receptor-Binding Domain (RBD). Nat. Prod. Res. 2021, 1–6. [Google Scholar] [CrossRef]
  126. Prashantha, C.N.; Gouthami, K.; Lavanya, L.; Bhavanam, S.; Jakhar, A.; Shakthiraju, R.G.; Suraj, V.; Sahana, K.V.; Sujana, H.S.; Guruprasad, N.M.; et al. Molecular Screening of Antimalarial, Antiviral, Anti-Inflammatory and HIV Protease Inhibitors against Spike Glycoprotein of Coronavirus. J. Mol. Graph. Model. 2021, 102. [Google Scholar] [CrossRef]
  127. Pojskic, L. Screening of Preferential Binding Affinity of Selected Natural Compounds to SARS-CoV-2 Proteins Using in Silico Methods. EJMO 2020, 4, 319. [Google Scholar] [CrossRef]
  128. Badraoui, R.; Saoudi, M.; Hamadou, W.S.; Elkahoui, S.; Siddiqui, A.J.; Alam, J.M.; Jamal, A.; Adnan, M.; Suliemen, A.M.E.; Alreshidi, M.M.; et al. Antiviral Effects of Artemisinin and Its Derivatives against SARS-CoV-2 Main Protease: Computational Evidences and Interactions with ACE2 Allelic Variants. Pharmaceuticals 2022, 15, 129. [Google Scholar] [CrossRef]
  129. Belhassan, A.; Zaki, H.; Chtita, S.; Alaqarbeh, M.; Alsakhen, N.; Benlyas, M.; Lakhlifi, T.; Bouachrine, M. Camphor, Artemisinin and Sumac Phytochemicals as Inhibitors against COVID-19: Computational Approach. Comput. Biol. Med. 2021, 136. [Google Scholar] [CrossRef]
  130. Dogan, K.; Erol, E.; Didem Orhan, M.; Degirmenci, Z.; Kan, T.; Gungor, A.; Yasa, B.; Avsar, T.; Cetin, Y.; Durdagi, S.; et al. Instant Determination of the Artemisinin from Various Artemisia Annua L. Extracts by LC-ESI-MS/MS and Their In-Silico Modelling and In Vitro Antiviral Activity Studies against SARS-CoV-2. Phytochem. Anal. 2021. [Google Scholar] [CrossRef]
  131. Hasan, A.; Jannat, K.; Bondhon, T.A.; Jahan, R.; Hossan, M.S.; de Lourdes Pereira, M.; Nissapatorn, V.; Wiart, C.; Rahmatullah, M. Can Antimalarial Phytochemicals Be a Possible Cure for COVID-19? Molecular Docking Studies of Some Phytochemicals to SARS-CoV-2 3C-like Protease. Infect. Disord. Drug Targets 2021, 21. [Google Scholar] [CrossRef]
  132. Sudeep, H. Molecular Docking Analysis of Withaferin A from Withania Somnifera with the Glucose Regulated Protein 78 (GRP78) in Comparison with the COVID-19 Main Protease. Bioinformation 2020, 16, 411. [Google Scholar] [CrossRef] [PubMed]
  133. De Oliveira, V.M.; da Rocha, M.N.; Magalhães, E.P.; da Silva Mendes, F.R.; Marinho, M.M.; de Menezes, R.R.P.P.B.; Sampaio, T.L.; dos Santos, H.S.; Martins, A.M.C.; Marinho, E.S. Computational Approach towards the Design of Artemisinin–Thymoquinone Hybrids against Main Protease of SARS-COV-2. Future J. Pharm. Sci. 2021, 7. [Google Scholar] [CrossRef] [PubMed]
  134. Gurung, A.B.; Ali, M.A.; Lee, J.; Farah, M.A.; Al-Anazi, K.M.; Al-Hemaid, F. Artesunate Induces Substantial Topological Alterations in the SARS-CoV-2 Nsp1 Protein Structure. J. King Saud Univ. Sci. 2022, 34. [Google Scholar] [CrossRef] [PubMed]
  135. Zhou, Y.; Gilmore, K.; Ramirez, S.; Settels, E.; Gammeltoft, K.A.; Pham, L.V.; Fahnøe, U.; Feng, S.; Offersgaard, A.; Trimpert, J.; et al. In Vitro Efficacy of Artemisinin-Based Treatments against SARS-CoV-2. Sci. Rep. 2021, 11. [Google Scholar] [CrossRef] [PubMed]
  136. Trieu, V. Targeting TGF-β Pathway with COVID-19 Drug Candidate ARTIVeda/PulmoHeal Accelerates Recovery from Mild-Moderate COVID-19. 2021. medRxiv 2021. [Google Scholar] [CrossRef]
  137. Sehailia, M.; Chemat, S. Antimalarial-Agent Artemisinin and Derivatives Portray More Potent Binding to Lys353 and Lys31-Binding Hotspots of SARS-CoV-2 Spike Protein than Hydroxychloroquine: Potential Repurposing of Artenimol for COVID-19. J. Biomol. Struct. Dyn. 2020, 1–11. [Google Scholar] [CrossRef]
  138. Sharma, S.; Deep, S. In-Silico Drug Repurposing for Targeting SARS-CoV-2 Main Protease (Mpro). J. Biomol. Struct. Dyn. 2022, 40, 3003. [Google Scholar] [CrossRef]
  139. Dey, D.; Borkotoky, S.; Banerjee, M. In Silico Identification of Tretinoin as a SARS-CoV-2 Envelope (E) Protein Ion Channel Inhibitor. Comput. Biol. Med. 2020, 127. [Google Scholar] [CrossRef]
  140. Cao, R.; Hu, H.; Li, Y.; Wang, X.; Xu, M.; Liu, J.; Zhang, H.; Yan, Y.; Zhao, L.; Li, W.; et al. Anti-SARS-CoV-2 Potential of Artemisinins in Vitro. ACS Infect. Dis. 2020, 6, 2524. [Google Scholar] [CrossRef]
  141. Hu, Y.; Liu, M.; Qin, H.; Lin, H.; An, X.; Shi, Z.; Song, L.; Yang, X.; Fan, H.; Tong, Y. Artemether, Artesunate, Arteannuin B, Echinatin, Licochalcone B and Andrographolide Effectively Inhibit SARS-CoV-2 and Related Viruses In Vitro. Front. Cell. Infect. Microbiol. 2021, 11. [Google Scholar] [CrossRef]
  142. Ghosh, A.K.; Miller, H.; Knox, K.; Kundu, M.; Henrickson, K.J.; Arav-Boger, R. Inhibition of Human Coronaviruses by Antimalarial Peroxides. ACS Infect. Dis. 2021, 7, 1985. [Google Scholar] [CrossRef] [PubMed]
  143. Nixon, G.L.; Moss, D.M.; Shone, A.E.; Lalloo, D.G.; Fisher, N.; O’neill, P.M.; Ward, S.A.; Biagini, G.A. Antimalarial Pharmacology and Therapeutics of Atovaquone. J. Antimicrob. Chemother. 2013, 68, 977. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Weyant, R.B.; Kabbani, D.; Doucette, K.; Lau, C.; Cervera, C. Pneumocystis Jirovecii: A Review with a Focus on Prevention and Treatment. Expert Opin. Pharmacother. 2021, 22, 1579. [Google Scholar] [CrossRef] [PubMed]
  145. Florez, H.; Singh, S. Coronavirus Disease 2019 Drug Discovery through Molecular Docking. F1000Res 2020, 9. [Google Scholar] [CrossRef]
  146. Lizbeth Ramírez-Salinas, G.; Martínez-Archundia, M.; Correa-Basurto, J.; García-Machorro, J. Repositioning of Ligands That Target the Spike Glycoprotein as Potential Drugs for SARS-CoV-2 in an In Silico Study. Molecules 2020, 25, 5615. [Google Scholar] [CrossRef]
  147. Kim, J.; Hwang, S.Y.; Kim, D.; Kim, M.; Baek, K.; Kang, M.; An, S.; Gong, J.; Park, S.; Kandeel, M.; et al. Abiraterone Acetate Attenuates SARS-CoV-2 Replication by Interfering with the Structural Nucleocapsid Protein. Biomol. Ther. 2022. [Google Scholar] [CrossRef]
  148. Marak, B.N.; Dowarah, J.; Khiangte, L.; Singh, V.P. Step toward Repurposing Drug Discovery for COVID-19 Therapeutics through in Silico Approach. Drug Dev. Res. 2020, 82, 374. [Google Scholar] [CrossRef]
  149. Carter-Timofte, M.E.; Arulanandam, R.; Kurmasheva, N.; Fu, K.; Laroche, G.; Taha, Z.; van der Horst, D.; Cassin, L.; van der Sluis, R.M.; Palermo, E.; et al. Antiviral Potential of the Antimicrobial Drug Atovaquone against SARS-CoV-2 and Emerging Variants of Concern. ACS Infect. Dis. 2021, 7, 3034. [Google Scholar] [CrossRef]
  150. Rodriguez-Rodriguez, B.A.; Noval, M.G.; Kaczmarek, M.E.; Jang, K.K.; Thannickal, S.A.; Kottkamp, A.C.; Brown, R.S.; Kielian, M.; Cadwell, K.; Stapleford, K.A.; et al. Atovaquone and Berberine Chloride Reduce SARS-CoV-2 Replication In Vitro. Viruses 2021, 13, 2437. [Google Scholar] [CrossRef]
  151. Pickardid, A.; Calverleyid, B.C.; Changid, J.; Garvaid, R.; Gago, S.; Lu, Y.; Kadlerid, K.E. Discovery of Re-Purposed Drugs That Slow SARS-CoV-2 Replication in Human Cells. bioRxiv 2021. [Google Scholar] [CrossRef]
  152. Mellott, D.M.; Tseng, C.-T.; Drelich, A.; Fajtová, P.; Chenna, B.C.; Kostomiris, D.H.; Hsu, J.; Zhu, J.; Taylor, Z.W.; Kocurek, K.I.; et al. A Clinical-Stage Cysteine Protease Inhibitor Blocks SARS-CoV-2 Infection of Human and Monkey Cells. ACS Chem. Biol. 2021, 16, 642–650. [Google Scholar] [CrossRef] [PubMed]
  153. Engel, J.C.; Doyle, P.S.; Hsieh, I.; McKerrow, J.H. Cysteine Protease Inhibitors Cure an Experimental Trypanosoma Cruzi Infection. J. Exp. Med. 1998, 188, 725. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Barr, S.C.; Warner, K.L.; Kornreic, B.G.; Piscitelli, J.; Wolfe, A.; Benet, L.; McKerrow, J.H. A Cysteine Protease Inhibitor Protects Dogs from Cardiac Damage during Infection by Trypanosoma Cruzi. Antimicrob. Agents Chemother. 2005, 49, 5160. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Meirelles, M.N.; Juliano, L.; Carmona, E.; Silva, S.G.; Costa, E.M.; Murta, A.C.; Scharfstein, J. Inhibitors of the Major Cysteinyl Proteinase (GP57/51) Impair Host Cell Invasion and Arrest the Intracellular Development of Trypanosoma Cruzi in Vitro. Mol. Biochem. Parasitol. 1992, 52, 175. [Google Scholar] [CrossRef]
  156. Franke de Cazzulo, B.M.; Martínez, J.; North, M.J.; Coombs, G.H.; Cazzulo, J.J. Effects of Proteinase Inhibitors on the Growth and Differentiation of Trypanosoma Cruzi. FEMS Microbiol. Lett. 1994, 124, 81. [Google Scholar] [CrossRef]
  157. Otta, D.A.; de Araújo, F.F.; de Rezende, V.B.; Souza-Fagundes, E.M.; Elói-Santos, S.M.; Costa-Silva, M.F.; Santos, R.A.; Costa, H.A.; Siqueira-Neto, J.L.; Martins-Filho, O.A.; et al. Identification of Anti-Trypanosoma Cruzi Lead Compounds with Putative Immunomodulatory Activity. Antimicrob. Agents Chemother. 2018, 62. [Google Scholar] [CrossRef] [Green Version]
  158. Zhou, Y.; Vedantham, P.; Lu, K.; Agudelo, J.; Carrion, R.; Nunneley, J.W.; Barnard, D.; Pöhlmann, S.; McKerrow, J.H.; Renslo, A.R.; et al. Protease Inhibitors Targeting Coronavirus and Filovirus Entry. Antivir. Res. 2015, 116, 76. [Google Scholar] [CrossRef]
  159. Chen, W.; Mook, R.A.; Premont, R.T.; Wang, J. Niclosamide: Beyond an Antihelminthic Drug. Cell. Signal. 2018, 41, 89. [Google Scholar] [CrossRef]
  160. Kadri, H.; Lambourne, L.A.; Mehellou, Y. Niclosamide, ADrug with Many (Re)Purposes. Chem. Med. Chem. 2018, 13, 1088. [Google Scholar] [CrossRef]
  161. Jung, E.; Nam, S.; Oh, H.; Jun, S.; Ro, H.J.; Kim, B.; Kim, M.; Go, Y.Y. Neutralization of Acidic Intracellular Vesicles by Niclosamide Inhibits Multiple Steps of the Dengue Virus Life Cycle In Vitro. Sci Rep. 2019, 9, 8682. [Google Scholar] [CrossRef]
  162. Xu, J.; Shi, P.Y.; Li, H.; Zhou, J. Broad Spectrum Antiviral Agent Niclosamide and Its Therapeutic Potential. ACS Infect. Dis. 2020, 6, 909. [Google Scholar] [CrossRef]
  163. Pindiprolu, S.K.S.S.; Pindiprolu, S.H. Plausible Mechanisms of Niclosamide as an Antiviral Agent against COVID-19. Med. Hypotheses 2020, 140. [Google Scholar] [CrossRef] [PubMed]
  164. Mostafa, A.; Kandeil, A.; Elshaier, Y.A.M.M.; Kutkat, O.; Moatasim, Y.; Rashad, A.A.; Shehata, M.; Gomaa, M.R.; Mahrous, N.; Mahmoud, S.H.; et al. Fda-Approved Drugs with Potent in Vitro Antiviral Activity against Severe Acute Respiratory Syndrome Coronavirus 2. Pharmaceuticals 2020, 13, 443. [Google Scholar] [CrossRef] [PubMed]
  165. Braga, L.; Ali, H.; Secco, I.; Chiavacci, E.; Neves, G.; Goldhill, D.; Penn, R.; Jimenez-Guardeño, J.M.; Ortega-Prieto, A.M.; Bussani, R.; et al. Drugs That Inhibit TMEM16 Proteins Block SARS-CoV-2 Spike-Induced Syncytia. Nature 2021, 594, 88. [Google Scholar] [CrossRef]
  166. Yu, S.; Piao, H.; Sanoj Rejinold, N.; Jin, G.; Choi, G.; Choy, J.H. Niclosamide–Clay Intercalate Coated with Nonionic Polymer for Enhanced Bioavailability toward COVID-19 Treatment. Polymers 2021, 13, 1044. [Google Scholar] [CrossRef] [PubMed]
  167. Wang, G.; Gaikwad, H.; McCarthy, M.; Gonzalez-Juarrero, M.; Li, Y.; Armstrong, M.; Reisdorph, N.; Morrison, T.; Simberg, D. Lipid Nanoparticle Formulation of Niclosamide (Nano NCM) Effectively Inhibits SARS-CoV-2 Replication in Vitro. Precis. Nanomed. 2021, 4, 724. [Google Scholar] [CrossRef]
  168. Needham, D. The PH Dependence of Niclosamide Solubility, Dissolution, and Morphology: Motivation for Potentially Universal Mucin-Penetrating Nasal and Throat Sprays for COVID19, Its Variants and Other Viral Infections. Pharm. Res. 2021. [Google Scholar] [CrossRef]
  169. Weiss, A.; Touret, F.; Baronti, C.; Gilles, M.; Hoen, B.; Nougairède, A.; de Lamballerie, X.; Sommer, M.O.A. Niclosamide Shows Strong Antiviral Activity in a Human Airway Model of SARS-CoV-2 Infection and a Conserved Potency against the Alpha (B.1.1.7), Beta (B.1.351) and Delta Variant (B.1.617.2). PLoS ONE 2021, 16. [Google Scholar] [CrossRef]
  170. Cairns, D.M.; Dulko, D.; Griffiths, J.K.; Golan, Y.; Cohen, T.; Trinquart, L.; Price, L.L.; Beaulac, K.R.; Selker, H.P. Efficacy of Niclosamide vs Placebo in SARS-CoV-2 Respiratory Viral Clearance, Viral Shedding, and Duration of Symptoms Among Patients With Mild to Moderate COVID-19. JAMA Netw. Open 2022, 5, e2144942. [Google Scholar] [CrossRef]
  171. Rossignol, J.-F. Nitazoxanide: A First-in-Class Broad-Spectrum Antiviral Agent. Antivir. Res. 2014, 110, 94. [Google Scholar] [CrossRef] [Green Version]
  172. Rossignol, J.-F.A.; Ayoub, A.; Ayers, M.S. Treatment of Diarrhea Caused by Cryptosporidium Parvum: A Prospective Randomized, Double-Blind, Placebo-Controlled Study of Nitazoxanide. J. Infect. Dis. 2001, 184, 103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Ortiz, J.J.; Ayoub, A.; Gargalaà, G.; Chegne, N.L.; Favennecà, L. Randomized Clinical Study of Nitazoxanide Compared to Metronidazole in the Treatment of Symptomatic Giardiasis in Children from Northern Peru. Aliment. Pharm. 2001, 15, 1409. [Google Scholar] [CrossRef] [PubMed]
  174. Dubreuil, L.; Houcke, I.; Mouton, Y.; Rossignol, J.F. In Vitro Evaluation of Activities of Nitazoxanide and Tizoxanide against Anaerobes and Aerobic Organisms. Antimicrob. Agents Chemother. 1996, 40, 2266. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. De Carvalho, L.P.S.; Lin, G.; Jiang, X.; Nathan, C. Nitazoxanide Kills Replicating and Nonreplicating Mycobacterium Tuberculosis and Evades Resistance. J. Med. Chem. 2009, 52, 5789. [Google Scholar] [CrossRef] [PubMed]
  176. De Carvalho, L.P.S.; Darby, C.M.; Rhee, K.Y.; Nathan, C. Nitazoxanide Disrupts Membrane Potential and Intrabacterial PH Homeostasis of Mycobacterium Tuberculosis. ACS Med. Chem. Lett. 2011, 2, 849. [Google Scholar] [CrossRef]
  177. Rossignol, J.-F. Nitazoxanide, a New Drug Candidate for the Treatment of Middle East Respiratory Syndrome Coronavirus. J. Inf Pub. Health 2016, 9, 227. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  178. Driouich, J.-S.; Cochin, M.; Touret, F.; Petit, P.-R.; Gilles, M.; Moureau, G.; Barthélémy, K.; Laprie, C.; Wattanakul, T.; Chotsiri, P.; et al. Pre-Clinical Evaluation of Antiviral Activity of Nitazoxanide against Sars-CoV-2. bioRxiv 2021. [Google Scholar] [CrossRef]
  179. Pepperrell, T.; Pilkington, V.; Owen, A.; Wang, J.; Hill, A.M. Review of Safety and Minimum Pricing of Nitazoxanide for Potential Treatment of COVID-19. J. Virus Erad. 2020, 6, 52. [Google Scholar] [CrossRef]
  180. Kelleni, M.T. Nitazoxanide/Azithromycin Combination for COVID-19: A Suggested New Protocol for Early Management. Pharm. Res. 2020, 157, 104874. [Google Scholar] [CrossRef]
  181. Sayed, A.M.; Khalaf, A.M.; Mohamed, I.; Abdelrahim, E.A.; Marwa, I.; Elgendy, O. Repurposing of Some Anti-Infective Drugs for COVID-19 Treatment: A Surveillance Study Supported by an in Silico Investigation. Int. J. Clin. Pract. 2021, 75, 13877. [Google Scholar] [CrossRef]
  182. Brosey, C.A.; Houl, J.H.; Katsonis, P.; Balapiti-Modarage, L.P.F.; Bommagani, S.; Arvai, A.; Moiani, D.; Bacolla, A.; Link, T.; Warden, L.S.; et al. Targeting SARS-CoV-2 Nsp3 Macrodomain Structure with Insights from Human Poly(ADP-Ribose) Glycohydrolase (PARG) Structures with Inhibitors. Prog. Biophys. Mol. Biol. 2021, 163, 171. [Google Scholar] [CrossRef] [PubMed]
  183. Meneses Calderón, J.; Figueroa Flores, M.D.R.; Paniagua Coria, L.; Briones Garduño, J.C.; Meneses Figueroa, J.; Vargas Contretas, M.J.; de la Cruz Ávila, L.; Díaz Meza, S.; Ramírez Chacón, R.; Padmanabhan, S.; et al. Nitazoxanide against COVID-19 in Three Explorative Scenarios. J. Infect. Dev. Ctries 2020, 14, 982. [Google Scholar] [CrossRef] [PubMed]
  184. Enríquez López, K.Y.; Meneses Calderón, J.; de la Cruz Ávila, L.; López Esquivel, M.Á.; Meneses Figueroa, J.; Vargas Contreras, M.J.; Anaya Herrera, J.; Sotelo Martínez, O.R.; Mendoza López, J.A.; Mendieta Zerón, H. Evolution of COVID-19 Pregnancies Treated With Nitazoxanide in a Third-Level Hospital. Cureus 2021, 13, e15002. [Google Scholar] [CrossRef] [PubMed]
  185. Rocco, P.R.; Silva, P.L.; Cruz, F.F.; Antonio Melo-Junior, M.C.; FGMM Tierno, P.; Moura, M.A.; Frederico De Oliveira, L.G.; Lima, C.C.; dos Santos, E.A.; Junior, W.F.; et al. Early Use of Nitazoxanide in Mild COVID-19 Disease: Randomised, Placebo-Controlled Trial on Behalf of the SARITA-2 Investigators. Eur. Respir J. 2021, 58. [Google Scholar] [CrossRef] [PubMed]
  186. Blum, V.F.; Cimerman, S.; Hunter, J.R.; Tierno, P.; Lacerda, A.; Soeiro, A.; Cardoso, F.; Bellei, N.C.; Maricato, J.; Mantovani, N.; et al. Nitazoxanide Superiority to Placebo to Treat Moderate COVID-19—A Pilot Prove of Concept Randomized Double-Blind Clinical Trial. EClinicalMedicine 2021, 37, 100981. [Google Scholar] [CrossRef]
  187. Elalfy, H.; Besheer, T.; El-Mesery, A.; El-Gilany, A.H.; Soliman, M.A.A.; Alhawarey, A.; Alegezy, M.; Elhadidy, T.; Hewidy, A.A.; Zaghloul, H.; et al. Effect of a Combination of Nitazoxanide, Ribavirin, and Ivermectin plus Zinc Supplement (MANS.NRIZ Study) on the Clearance of Mild COVID-19. J. Med. Virol. 2021, 93, 3176. [Google Scholar] [CrossRef]
  188. Rossignol, J.-F.; Bardin, M.C.; Fulgencio, J.; Mogelnicki, D.; Br Echot, C. A Randomized Double-Blind Placebo-Controlled Clinical Trial of Nitazoxanide for Treatment of Mild or Moderate COVID-19. eClinicalMedicine 2022, 45, 101310. [Google Scholar] [CrossRef]
  189. Riccio, A.; Santopolo, S.; Rossi, A.; Piacentini, S.; Rossignol, J.-F.; Santoro, M. Gabriella Impairment of SARS-CoV-2 Spike Glycoprotein Maturation and Fusion Activity by Nitazoxanide: An Effect Independent of Spike Variants Emergence. Cell. Mol. Life Sci. 2022, 79, 227. [Google Scholar] [CrossRef]
  190. Hong, S.K.; Kim, H.J.; Song, C.S.; Choi, I.S.; Lee, J.B.; Park, S.Y. Nitazoxanide Suppresses IL-6 Production in LPS-Stimulated Mouse Macrophages and TG-Injected Mice. Int. Immunopharmacol. 2012, 13, 23. [Google Scholar] [CrossRef]
  191. Clerici, M.; Trabattoni, D.; Pacei, M.; Biasin, M.; Rossignol, J. The Anti-Infective Nitazoxanide Shows Strong Immumodulating Effects (155.21). J. Immunol. 2011, 186, 155. [Google Scholar]
  192. Riberoid, M.S.; Jouvenet, N.; Dreuxid, M.; Bastien Nisoleid, S. Interplay between SARS-CoV-2 and the Type I Interferon Response. PLoS Pathog. 2020, 16, e1008737. [Google Scholar] [CrossRef]
  193. Miner, K.; Labitzke, K.; Liu, B.; Wang, P.; Henckels, K.; Gaida, K.; Elliott, R.; Chen, J.J.; Liu, L.; Leith, A.; et al. Drug Repurposing: The Anthelmintics Niclosamide and Nitazoxanide Are Potent TMEM16A Antagonists That Fully Bronchodilate Airways. Front. Pharm. 2019, 10, 51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Laing, R.; Gillan, V.; Devaney, E. Ivermectin–Old Drug, New Tricks? Trends Parasitol. 2017, 33, 463. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Canga, A.G.; Prieto, A.M.S.; Diez Liébana, M.J.; Martínez, N.F.; Sierra Vega, M.; García Vieitez, J.J. The Pharmacokinetics and Interactions of Ivermectin in Humans—A Mini-Review. AAPS J. 2008, 10, 42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  196. Buonfrate, D.; Salas-Coronas, J.; Muñoz, J.; Maruri, B.T.; Rodari, P.; Castelli, F.; Zammarchi, L.; Bianchi, L.; Gobbi, F.; Cabezas-Fernández, T.; et al. Multiple-Dose versus Single-Dose Ivermectin for Strongyloides Stercoralis Infection (Strong Treat 1 to 4): A Multicentre, Open-Label, Phase 3, Randomised Controlled Superiority Trial. Lancet Infect. Dis. 2019, 19, 1181. [Google Scholar] [CrossRef]
  197. Wagstaff, K.M.; Sivakumaran, H.; Heaton, S.M.; Harrich, D.; Jans, D.A. Ivermectin Is a Specific Inhibitor of Importin α/β-Mediated Nuclear Import Able to Inhibit Replication of HIV-1 and Dengue Virus. Biochem. J. 2012, 443, 851. [Google Scholar] [CrossRef] [Green Version]
  198. Oka, M.; Yoneda, Y. Importin α: Functions as a Nuclear Transport Factor and Beyond. Proc. Jpn. Acad. Ser. B Phys. Biol. Sci. 2018, 94, 259. [Google Scholar] [CrossRef] [Green Version]
  199. Jans, D.A.; Wagstaff, K.M. The Broad Spectrum Host-Directed Agent Ivermectin as an Antiviral for SARS-CoV-2? Biochem. Biophys. Res. Commun. 2021, 538, 163. [Google Scholar] [CrossRef]
  200. Martin, A.J.; Jans, D.A. Antivirals That Target the Host IMPα/Β1-Virus Interface. Biochem. Soc. Trans. 2021, 49, 281. [Google Scholar] [CrossRef]
  201. Rowland, R.R.R.; Chauhan, V.; Fang, Y.; Pekosz, A.; Kerrigan, M.; Burton, M.D. Intracellular Localization of the Severe Acute Respiratory Syndrome Coronavirus Nucleocapsid Protein: Absence of Nucleolar Accumulation during Infection and after Expression as a Recombinant Protein in Vero Cells. J. Virol. 2005, 79, 11507. [Google Scholar] [CrossRef] [Green Version]
  202. Timani, K.A.; Liao, Q.; Ye, L.; Zeng, Y.; Liu, J.; Zheng, Y.; Ye, L.; Yang, X.; Lingbao, K.; Gao, J.; et al. Nuclear/Nucleolar Localization Properties of C-Terminal Nucleocapsid Protein of SARS Coronavirus. Virus Res. 2005, 114, 23. [Google Scholar] [CrossRef] [PubMed]
  203. Wulan, W.N.; Heydet, D.; Walker, E.J.; Gahan, M.E.; Ghildyal, R. Nucleocytoplasmic Transport of Nucleocapsid Proteins of Enveloped RNA Viruses. Front. Microbiol. 2015, 6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Caly, L.; Druce, J.D.; Catton, M.G.; Jans, D.A.; Wagstaff, K.M. The FDA-Approved Drug Ivermectin Inhibits the Replication of SARS-CoV-2 in Vitro. Antivir. Res. 2020, 178, 3. [Google Scholar] [CrossRef]
  205. Lehrer, S.; Rheinstein, P.H. Ivermectin Docks to the SARS-CoV-2 Spike Receptor-Binding Domain Attached to ACE2. In Vivo 2020, 34, 3023. [Google Scholar] [CrossRef] [PubMed]
  206. Francés-Monerris, A.; García-Iriepa, C.; Iriepa, I.; Hognon, C.; Miclot, T.; Barone, G.; Monari, A.; Marazzi, M. Microscopic Interactions between Ivermectin and Key Human and Viral Proteins Involved in SARS-CoV-2 Infection. Phys. Chem. Chem. Phys. 2021, 23, 22957. [Google Scholar] [CrossRef] [PubMed]
  207. Li, N.; Zhao, L.; Zhan, X. Quantitative Proteomics Reveals a Broad-Spectrum Antiviral Property of Ivermectin, Benefiting for COVID-19 Treatment. J. Cell Phys. 2021, 236, 2959. [Google Scholar] [CrossRef]
  208. Mody, V.; Ho, J.; Wills, S.; Mawri, A.; Lawson, L.; Ebert, M.C.C.J.C.; Fortin, G.M.; Rayalam, S.; Taval, S. Identification of 3-Chymotrypsin like Protease (3CLPro) Inhibitors as Potential Anti-SARS-CoV-2 Agents. Commun. Biol. 2021, 4. [Google Scholar] [CrossRef] [PubMed]
  209. Udofia, I.A.; Gbayo, K.O.; Oloba-Whenu, O.A.; Ogunbayo, T.B.; Isanbor, C. In Silico Studies of Selected Multi-Drug Targeting against 3CLpro and Nsp12 RNA-Dependent RNA-Polymerase Proteins of SARS-CoV-2 and SARS-CoV. Netw. Model. Anal. Health Inform. Bioinform. 2021, 10. [Google Scholar] [CrossRef]
  210. Bello, M. Elucidation of the Inhibitory Activity of Ivermectin with Host Nuclear Importin α and Several SARS-CoV-2 Targets. J. Biomol. Struct. Dyn. 2021, 1–9. [Google Scholar] [CrossRef]
  211. Qureshi, U.; Mir, S.; Naz, S.; Nur-e-Alam, M.; Ahmed, S.; Ul-Haq, Z. Mechanistic Insights into the Inhibitory Activity of FDA Approved Ivermectin against SARS-CoV-2: Old Drug with New Implications. J. Biomol. Struct. Dyn. 2021, 1–12. [Google Scholar] [CrossRef]
  212. Kern, C.; Schöning, V.; Chaccour, C.; Hammann, F. Modeling of SARS-CoV-2 Treatment Effects for Informed Drug Repurposing. Front. Pharmacol. 2021, 12, 1. [Google Scholar] [CrossRef]
  213. Dinesh Kumar, N.; ter Ellen, B.M.; Bouma, E.M.; Troost, B.; van de Pol, D.P.I.; van der Ende-Metselaar, H.H.; van Gosliga, D.; Apperloo, L.; Carpaij, O.A.; van den Berge, M.; et al. Moxidectin and Ivermectin Inhibit Sars-Cov-2 Replication in Vero E6 Cells But Not in Human Primary Airway Epithelium Cells. Antimicrob. Agent Chemother. 2021, 66, e0154321. [Google Scholar] [CrossRef] [PubMed]
  214. Choudhury, A.; Das, N.C.; Patra, R.; Bhattacharya, M.; Ghosh, P.; Patra, B.C.; Mukherjee, S. Exploring the Binding Efficacy of Ivermectin against the Key Proteins of SARS-CoV-2 Pathogenesis: An Approach. Future Virol. 2021, 16, 277. [Google Scholar] [CrossRef]
  215. Muthusamy, S.; Gopal, H.; Manivarma, T. Virtual Screening Reveals Potential Anti-Parasitic Drugs Inhibiting the Receptor Binding Domain of SARS-CoV-2 Spike Protein. J. Virol. Antivir. Res. 2021, 110. [Google Scholar] [CrossRef]
  216. González-paz, L.; Hurtado-león, M.L.; Lossada, C.; Fernández-materán, F.V. Structural Deformability Induced in Proteins of Potential Interest Associated with COVID-19 by Binding of Homologues Present in Ivermectin: Comparative Study Based in Elastic Networks Models. J. Mol. Liq. 2021, 340, 117284. [Google Scholar] [CrossRef]
  217. González-Paz, L.; Hurtado-León, M.L.; Lossada, C.; Fernández-Materán, F.V.; Vera-Villalobos, J.; Loroño, M.; Paz, J.L.; Jeffreys, L.; Alvarado, Y.J. Comparative Study of the Interaction of Ivermectin with Proteins of Interest Associated with SARS-CoV-2: A Computational and Biophysical Approach. Biophys. Chem. 2021, 278, 106677. [Google Scholar] [CrossRef]
  218. Jeffreys, L.N.; Pennington, S.H.; Duggan, J.; Caygill, C.H.; Lopeman, R.C.; Breen, A.F.; Jinks, J.B.; Ardrey, A.; Donnellan, S.; Patterson, E.I.; et al. Remdesivir-Ivermectin Combination Displays Synergistic Interaction with Improved in Vitro Activity against SARS-CoV-2. Int. J. Antimicrob. Agents 2022, 59, 106542. [Google Scholar] [CrossRef]
  219. Eweas, A.F.; Alhossary, A.A.; Abdel-Moneim, A.S. Molecular Docking Reveals Ivermectin and Remdesivir as Potential Repurposed Drugs Against SARS-CoV-2. Front. Microbiol. 2021, 11, 592908. [Google Scholar] [CrossRef]
  220. Segatori, V.I.; Garona, J.; Caligiuri, L.G.; Bizzotto, J.; Lavignolle, R.; Toro, A.; Sanchis, P.; Spitzer, E.; Krolewiecki, A.; Gueron, G.; et al. Effect of Ivermectin and Atorvastatin on Nuclear Localization of Importin Alpha and Drug Target Expression Profiling in Host Cells from Nasopharyngeal Swabs of Sars-cov-2- Positive Patients. Viruses 2021, 13, 2084. [Google Scholar] [CrossRef]
  221. Arévalo, A.P.; Pagotto, R.; Pórfido, J.L.; Daghero, H.; Segovia, M.; Yamasaki, K.; Varela, B.; Hill, M.; Verdes, J.M.; Duhalde Vega, M.; et al. Ivermectin Reduces in Vivo Coronavirus Infection in a Mouse Experimental Model. Sci. Rep. 2021, 11, 7132. [Google Scholar] [CrossRef]
  222. Melo, G.D.; Lazarini, F.; Larrous, F.; Feige, L.; Kornobis, E.; Levallois, S.; Marchio, A.; Kergoat, L.; Hardy, D.; Cokelaer, T.; et al. Attenuation of Clinical and Immunological Outcomes during SARS-CoV-2 Infection by Ivermectin. EMBO Mol. Med. 2021, 13, 1. [Google Scholar] [CrossRef] [PubMed]
  223. Guzzo, C.A.; Furtek, C.I.; Porras, A.G. Safety, Tolerability, and Pharmacokinetics of Escalating High Doses of Ivermectin in Healthy Adult Subjects. J. Clin. Pharmacol. 2002, 42, 1122. [Google Scholar] [CrossRef] [PubMed]
  224. Ndyomugyenyi, R.; Kabatereine, N.; Olsen, A.; Magnussen, P. Efficacy of Ivermectin and Albendazole Alone and in Combination for Treatment of Soil-Transmitted Helminths in Pregnancy and Adverse Events: A Randomized Open Label Controlled Intervention Trial in Masindi District, Western Uganda. Am. J. Trop. Med. Hyg. 2009, 79, 856. [Google Scholar] [CrossRef] [Green Version]
  225. Wise, L.D.; Scialli, A.R. Ivermectin for COVID-19: Concerns during Pregnancy. Reprod. Toxicol. 2022, 107, 43. [Google Scholar] [CrossRef]
  226. Taylor, M.M.; Kobeissi, L.; Kim, C.; Amin, A.; Thorson, A.E.; Bellare, N.B.; Brizuela, V.; Bonet, M.; Kara, E.; Thwin, S.S.; et al. Inclusion of Pregnant Women in COVID-19 Treatment Trials: A Review and Global Call to Action. Lancet Glob. Health 2021, 9, e366. [Google Scholar] [CrossRef]
  227. Arshad, U.; Pertinez, H.; Box, H.; Tatham, L.; Rajoli, R.K.R.; Curley, P.; Neary, M.; Sharp, J.; Liptrott, N.J.; Valentijn, A.; et al. Prioritization of Anti-SARS-Cov-2 Drug Repurposing Opportunities Based on Plasma and Target Site Concentrations Derived from Their Established Human Pharmacokinetics. Clin. Pharmacol. Ther. 2020, 108, 775. [Google Scholar] [CrossRef]
  228. Ahmed, S.; Mahbubul, M.; Ross, A.G.; Sharif, M.; Clemens, J.D.; Kibtiya, M.; Swe, C.; Zaman, K.; Somani, J.; Yasmin, R.; et al. A Five-Day Course of Ivermectin for the Treatment of COVID-19 May Reduce the Duration of Illness. Biomed. Biotechnol. Res. J. 2020, 21, 214–216. [Google Scholar] [CrossRef]
  229. Babalola, O.E.; Bode, C.O.; Ajayi, A.A.; Alakaloko, F.M.; Akase, I.E.; Otrofanowei, E.; Salu, O.B.; Adeyemo, W.L.; Ademuyiwa, A.O.; Omilabu, S. Ivermectin Shows Clinical Benefits in Mild to Moderate COVID19: A Randomized Controlled Double-Blind, Dose-Response Study in Lagos. QJM Int. J. Med. 2022, 114, 780. [Google Scholar] [CrossRef]
  230. Pott-Junior, H.; Bastos Paoliello, M.M.; de Miguel, A.Q.C.; da Cunha, A.F.; de Melo Freire, C.C.; Neves, F.F.; da Silva de Avó, L.R.; Roscani, M.G.; dos Santos, S.D.S.; Chachá, S.G.F. Use of Ivermectin in the Treatment of COVID-19: A Pilot Trial. Toxicol. Rep. 2021, 8, 505. [Google Scholar] [CrossRef]
  231. Aref, Z.F.; Bazeed, S.E.E.S.; Hassan, M.H.; Hassan, A.S.; Rashad, A.; Hassan, R.G.; Abdelmaksoud, A.A. Clinical, Biochemical and Molecular Evaluations of Ivermectin Mucoadhesive Nanosuspension Nasal Spray in Reducing Upper Respiratory Symptoms of Mild COVID-19. Int. J. Nanomed. 2021, 16, 4063. [Google Scholar] [CrossRef]
  232. Cadegiani, F.A.; Goren, A.; Wambier, C.G.; McCoy, J. Early COVID-19 Therapy with Azithromycin plus Nitazoxanide, Ivermectin or Hydroxychloroquine in Outpatient Settings Significantly Improved COVID-19 Outcomes Compared to Known Outcomes in Untreated Patients. New Microbes New Infect. 2021, 43, 100915. [Google Scholar] [CrossRef]
  233. Krolewiecki, A.; Lifschitz, A.; Moragas, M.; Travacio, M.; Valentini, R.; Alonso, D.F.; Solari, R.; Tinelli, M.A.; Cimino, R.O.; Álvarez, L.; et al. Antiviral Effect of High-Dose Ivermectin in Adults with COVID-19: A Proof-of-Concept Randomized Trial. EClinicalMedicine 2021, 37. [Google Scholar] [CrossRef] [PubMed]
  234. Chaccour, C.; Casellas, A.; Blanco-Di Matteo, A.; Pineda, I.; Fernandez-Montero, A.; Ruiz-Castillo, P.; Richardson, M.A.; Rodríguez-Mateos, M.; Jordán-Iborra, C.; Brew, J.; et al. The Effect of Early Treatment with Ivermectin on Viral Load, Symptoms and Humoral Response in Patients with Non-Severe COVID-19: A Pilot, Double-Blind, Placebo-Controlled, Randomized Clinical Trial. EClinicalMedicine 2021, 32. [Google Scholar] [CrossRef] [PubMed]
  235. Ravikirti; Roy, R.; Pattadar, C.; Raj, R.; Agarwal, N.; Biswas, B.; Manjhi, P.K.; Rai, D.K.; Shyama; Kumar, A.; et al. Evaluation of Ivermectin as a Potential Treatment for Mild to Moderate Covid-19: A Double-Blind Randomized Placebo Controlled Trial in Eastern India. J. Pharm. Pharm. Sci. 2021, 24, 343. [Google Scholar] [CrossRef]
  236. Mohan, A.; Tiwari, P.; Menon Suri, T.; Mittal, S.; Patel, A.; Jain, A.; Velpandian, T.; Das, S.; Boppana, K.; Pandey, R.M.; et al. Single-Dose Oral Ivermectin in Mild and Moderate COVID-19 (RIVET-COV): A Single-Centre Randomized, Placebo-Controlled Trial. J. Infect. Chemother. 2021, 27, 1341. [Google Scholar] [CrossRef] [PubMed]
  237. Buonfrate, D.; Chesini, F.; Martini, D.; Roncaglioni, M.C.; Ojeda Fernandez, M.L.; Alvisi, M.F.; de Simone, I.; Rulli, E.; Nobili, A.; Casalini, G.; et al. High-Dose Ivermectin for Early Treatment of COVID-19 (COVER Study): A Randomised, Double-Blind, Multicentre, Phase II, Dose-Finding, Proof-of-Concept Clinical Trial. Int. Antimicrob. Agents 2022, 19, 106516. [Google Scholar] [CrossRef]
  238. Podder, C.S.; Chowdhury, N.; Sina, M.I.; Haque, W.M.M.U. Outcome of Ivermectin Treated Mild to Moderate COVID-19 Cases: A Single-Centre, Open-Label, Randomised Controlled Study. IMC J. Med. Sci. 2021, 14, 11. [Google Scholar] [CrossRef]
  239. Zeeshan Khan Chachar, A.; Ahmad Khan, K.; Asif, M.; Tanveer, K.; Khaqan, A.; Basri, R. Effectiveness of Ivermectin in SARS-CoV-2/COVID-19 Patients. Int. J. Sci. 2020, 9, 31. [Google Scholar] [CrossRef]
  240. Lim, S.C.L.; Hor, C.P.; Tay, K.H.; Mat Jelani, A.; Tan, W.H.; Ker, H.B.; Chow, T.S.; Zaid, M.; Cheah, W.K.; Lim, H.H.; et al. Efficacy of Ivermectin Treatment on Disease Progression Among Adults With Mild to Moderate COVID-19 and Comorbidities: The I-TECH Randomized Clinical Trial. JAMA Intern. Med. 2022, 182, 426. [Google Scholar] [CrossRef]
  241. Galan, L.E.B.; Santos, N.M.D.; Asato, M.S.; Araújo, J.V.; de Lima Moreira, A.; Araújo, A.M.M.; Paiva, A.D.P.; Portella, D.G.S.; Marques, F.S.S.; Silva, G.M.A.; et al. Phase 2 Randomized Study on Chloroquine, Hydroxychloroquine or Ivermectin in Hospitalized Patients with Severe Manifestations of SARS-CoV-2 Infection. Pathog. Glob. Health 2021, 115, 235. [Google Scholar] [CrossRef]
  242. Shahbaznejad, L.; Davoudi, A.; Eslami, G.; Markowitz, J.S.; Navaeifar, M.R.; Hosseinzadeh, F.; Movahedi, F.S.; Rezai, M.S. Effects of Ivermectin in Patients With COVID-19: A Multicenter, Double-Blind, Randomized, Controlled Clinical Trial. Clin. Ther. 2021, 43, 1007. [Google Scholar] [CrossRef] [PubMed]
  243. Shimizu, K.; Hirata, H.; Kabata, D.; Tokuhira, N.; Koide, M.; Ueda, A.; Tachino, J.; Shintani, A.; Uchiyama, A.; Fujino, Y.; et al. Ivermectin Administration Is Associated with Lower Gastrointestinal Complications and Greater Ventilator-Free Days in Ventilated Patients with COVID-19: A Propensity Score Analysis. J. Infect. Chemother. 2022, 28, 548. [Google Scholar] [CrossRef] [PubMed]
  244. Camprubí, D.; Almuedo-Riera, A.; Martí-Soler, H.I.; Soriano, A.; Hurtado, J.C.; Subirà, C.; Grau-Pujol, B.; Krolewiecki, A.; Muñoz, J. Lack of Efficacy of Standard Doses of Ivermectin in Severe COVID-19 Patients. PLoS ONE 2020, 15, e0242184. [Google Scholar] [CrossRef]
  245. Abd-Elsalam, S.; Noor, R.A.; Badawi, R.; Khalaf, M.; Esmail, E.S.; Soliman, S.; Abd El Ghafar, M.S.; Elbahnasawy, M.; Moustafa, E.F.; Hassany, S.M.; et al. Clinical Study Evaluating the Efficacy of Ivermectin in COVID-19 Treatment: A Randomized Controlled Study. J. Med. Virol. 2021, 93, 5833. [Google Scholar] [CrossRef] [PubMed]
  246. Szente Fonseca, S.N.; de Queiroz Sousa, A.; Wolkoff, A.G.; Moreira, M.S.; Pinto, B.C.; Valente Takeda, C.F.; Rebouças, E.; Vasconcellos Abdon, A.P.; Nascimento, A.L.A.; Risch, H.A. Risk of Hospitalization for COVID-19 Outpatients Treated with Various Drug Regimens in Brazil: Comparative Analysis. Travel Med. Infect. Dis. 2020, 38, 101906. [Google Scholar] [CrossRef]
  247. Vallejos, J.; Zoni, R.; Bangher, M.; Villamandos, S.; Bobadilla, A.; Plano, F.; Campias, C.; Chaparro Campias, E.; Achinelli, F.; Guglielmone, H.A.; et al. Ivermectin to Prevent Hospitalizations in Patients with COVID-19 (IVERCOR-COVID19): A Structured Summary of a Study Protocol for a Randomized Controlled Trial. Trials 2020, 21, 10. [Google Scholar] [CrossRef]
  248. Franco Haroldo, D.; Hector, C.; Roberto, H. Ivermectin in Long-Covid Patients: A Retrospective Study. J. Biomed. Res. Clin. Investig. 2021, 2, 1008. [Google Scholar] [CrossRef]
  249. Rajter, J.C.; Sherman, M.S.; Fatteh, N.; Vogel, F.; Sacks, J.; Rajter, J.J. Use of Ivermectin Is Associated With Lower Mortality in Hospitalized Patients With Coronavirus Disease 2019: The Ivermectin in COVID Nineteen Study. Chest 2021, 159, 85. [Google Scholar] [CrossRef]
  250. Shakhsi Niaee, M.; Namdar, P.; Allami, A.; Zolghadr, L.; Javadi, A.; Karampour, A.; Varnaseri, M.; Bijani, B.; Cheraghi, F.; Naderi, Y.; et al. Ivermectin as an Adjunct Treatment for Hospitalized Adult COVID-19 Patients: A Randomized Multi-Center Clinical Trial. Asian Pac. J. Trop. Med. 2021, 14, 266. [Google Scholar] [CrossRef]
  251. Rezk, N.; Elsayed Sileem, A.; Gad, D.; Khalil, A.O. MiRNA-223-3p, MiRNA-2909 and Cytokines Expression in COVID-19 Patients Treated with Ivermectin. Zagazig Univ. Med. J. 2021. [Google Scholar] [CrossRef]
  252. Zubair, S.M.; Chaudhry, M.W.; Zubairi, A.B.S.; Shahzad, T.; Zahid, A.; Khan, I.A.; Khan, J.A.; Irfan, M. The Effect of Ivermectin on Non-Severe and Severe COVID-19 Disease and Gender-Based Difference of Its Effectiveness. Monaldi Arch. Chest Dis. 2022. [Google Scholar] [CrossRef]
  253. Abbas, K.U.; Muhammad, S.; Ding, S.F. The Effect of Ivermectin on Reducing Viral Symptoms in Patients with Mild COVID-19. Indian J. Pharm. Sci. 2020, 82. [Google Scholar] [CrossRef]
  254. Ozer, M.; Goksu, S.Y.; Conception, R.; Ulker, E.; Balderas, R.M.; Mahdi, M.; Manning, Z.; To, K.; Effendi, M.; Anandakrishnan, R.; et al. Effectiveness and Safety of Ivermectin in COVID-19 Patients: A Prospective Study at a Safety-Net Hospital. J. Med. Virol. 2021, 2, 1473. [Google Scholar] [CrossRef]
  255. Chowdhury, A.T.M.M.; Shahbaz, M.; Karim, M.R.; Islam, J.; Dan, G.; He, S. A Comparative Study on Ivermectin-Doxycycline and Hydroxychloro-quine-Azithromycin Therapy on COVID-19 Patients. Eurasian J. Med. Oncol. 2021, 5, 63. [Google Scholar] [CrossRef]
  256. Rahman, M.A.; Iqbal, S.A.; Islam, M.A.; Niaz, M.K.; Hussain, T.; Siddiquee, T.H. Comparison of Viral Clearance between Ivermectin with Doxycycline and Hy-droxychloroquine with Azithromycin in COVID-19 Patients. J. Bangladesh Coll. Phys. Surg. 2020, 5. [Google Scholar] [CrossRef]
  257. Alam, M.T.; Murshed, R.; Bhiuyan, E.; Saber, S.; Alam, R.F.; Robin, R.C. A Case Series of 100 COVID-19 Positive Patients Treated with Combination of Ivermectin and Doxycycline. J. Bangladesh Coll. Phys. Surg. 2020, 38, 10. [Google Scholar] [CrossRef]
  258. Mahmud, R.; Rahman, M.M.; Alam, I.; Ahmed, K.G.U.; Kabir, A.K.M.H.; Sayeed, S.K.J.B.; Rassel, M.A.; Monayem, F.B.; Islam, M.S.; Islam, M.M.; et al. Iver-mectin in Combination with Doxycycline for Treating COVID-19 Symptoms: A Randomized Trial. J. Int. Med. Res. 2021, 49. [Google Scholar] [CrossRef] [PubMed]
  259. Ahsan, T.; Rani, B.; Siddiqui, R.; D‘Souza, G.; Memon, R.; Lutfi, I.; Hasan, O.I.; Javed, R.; Khan, F.; Hassan, M. Clinical Variants, Characteristics, and Outcomes Among COVID-19 Patients: A Case Series Analysis at a Tertiary Care Hospital in Karachi, Pakistan. Cureus 2021, 13. [Google Scholar] [CrossRef]
  260. Bhattacharya, R.; Ray, I.; Mukherjee, R.; Chowdhury, S.; Kulasreshtha, M.; Ghosh, R. Observational Study on Clinical Features, Treatment and Outcome of Covid 19 in a Tertiary Care Centre in India—A Retrospective Case Series. Int. J. Sci. Res. 2020, 1. [Google Scholar] [CrossRef]
  261. Faisal, R.; Shah, S.F.A.; Hussain, M. Potential Use of Azithromycin Alone and in Combination with Ivermectin in Fighting against the Symptoms of COVID-19. Prof. Med. J. 2021, 28, 737. [Google Scholar] [CrossRef]
  262. Espitia-Hernandez, G.; Munguia, L.; Diaz-Chiguer, D.; Lopez-Elizalde, R.; Jimenez-Ponce, F. Effects of Ivermectin-Azithromycin-Cholecalciferol Combined Therapy on COVID-19 Infected Patients: A Proof of Concept Study. Biomed. Res. 2020, 31, 129. [Google Scholar]
  263. Lima-morales, R.; Méndez-hernández, P.; Flores, Y.N.; Cuecuecha-rugerio, E.; Nava-zamora, A.; Hernández-galdamez, D.R.; Romo-dueñas, D.K.; Salmerón, J. Effectiveness of a Multidrug Therapy Consisting of Ivermectin, Azithromycin, Mon-telukast, and Acetylsalicylic Acid to Prevent Hospitalization and Death among Ambulatory COVID-19 Cases in Tlaxcala, Mexico. Int. J. Infect. Dis 2021, 105, 598. [Google Scholar] [CrossRef] [PubMed]
  264. De Jesús Ascencio-Montiel, I.; Tomás-López, J.C.; Álvarez-Medina, V.; Gil-Velázquez, L.E.; Vega-Vega, H.; Vargas-Sánchez, H.R.; Cervantes-Ocampo, M.; Villasís-Keever, M.Á.; González-Bonilla, C.R.; Duque-Molina, C. A Multimodal Strategy to Reduce the Risk of Hospitalization/Death in Ambulatory Patients with COVID-19. Arch. Med. Res. 2022, 1. [Google Scholar] [CrossRef]
  265. Kishoria, N.; Mathur, S.L.; Parmar, V.; Kaur, R.J.; Agarwal, H.; Parihar, B.S.; Verma, S. Ivermectin as adjuvant to hydroxycholoroquine in patients resistant to standard treatment for SARS-CoV-2: Results of an open-label randomized clinical study. Paripex-Indian J. Res. 2020, 50. [Google Scholar] [CrossRef]
  266. Ferreira, R.M.; Beranger, R.W.; Sampaio, P.P.N.; Filho, J.M.; Lima, R.A.C. Out-comes Associated with Hydroxychloroquine and Ivermectin in Hospitalized Pa-tients with COVID-19: A Single-Center Experience. Rev. Assoc. Med. Bras. 2021, 67, 1466. [Google Scholar] [CrossRef] [PubMed]
  267. Sudhir, M.; Ajay Singh, T.; Deepti Singh, H.; Vibhu Sagar, K.; Yash, S. Comparative Analytical Study of Two Different Drug Regimens in Treatment of Covid-19 Positive Patients in Index Medical College Hospital and Research Center, Indore, India. Available online: https://ijhcr.com/index.php/ijhcr/article/view/1263/1064 (accessed on 19 February 2022).
  268. Sathi, S.; Garg, A.K.; Mittal, A.; Verma, S.; Saini, V.S.; Singh, M.K.; Vohra, D.; Kumar, S. Clinical Effect of the Combination Therapy of Hydroxychloroquine, Azithromycin and Ivermectin in Patients with COVID-19. J. Cardiovasc. Dis. Res. 2021, 12, 61. [Google Scholar] [CrossRef]
  269. Okumuş, N.; Demirtürk, N.; Çetinkaya, R.A.; Güner, R.; Avcı, İ.Y.; Orhan, S.; Konya, P.; Şaylan, B.; Karalezli, A.; Yamanel, L.; et al. Evaluation of the Effectiveness and Safety of Adding Ivermectin to Treatment in Severe COVID-19 Patients. BMC Infect. Dis. 2021, 21, 411. [Google Scholar] [CrossRef] [PubMed]
  270. Hector, C.; Roberto, H.; Protocol, C.-I.D.E.A. Safety and Efficacy of the Combined Use of Ivermectin, Dexamethasone, Enoxaparin and Aspirina against COVID-19 the I.D.E.A. Protocol. J. Clin. Trials 2021, 11, 459. [Google Scholar]
  271. Shoumann, W.M.; Hegazy, A.A.; Nafae, R.M.; Ragab, M.I.; Samra, S.R.; Ibrahim, D.A.; Al-Mahrouky, T.H.; Sileem, A.E. Use of Ivermectin as a Potential Chemopro-phylaxis for COVID-19 in Egypt: A Randomised Clinical Trial. J. Clin. Diagn. Res. 2021, 27. [Google Scholar] [CrossRef]
  272. Behera, P.; Patro, B.K.; Singh, A.K.; Chandanshive, P.D.; Ravikumar, S.R.; Pra-dhan, S.K.; Pentapati, S.S.K.; Batmanabane, G.; Mohapatra, P.R.; Padhy, B.M.; et al. Role of Ivermectin in the Prevention of SARS-CoV-2 Infection among Healthcare Workers in India: A Matched Case-Control Study. PLoS ONE 2021, 16, e0247163. [Google Scholar] [CrossRef]
  273. Morgenstern, J.; Redondo, J.N.; Olavarria, A.; Rondon, I.; Roca, S.; De Leon, A.; Canela, J.; Tavares, J.; Minaya, M.; Lopez, O.; et al. Ivermectin as a SARS-CoV-2 Pre-Exposure Prophylaxis Method in Healthcare Workers: A Propensity Score-Matched Retrospective Cohort Study. Cureus 2021, 13, e17455. [Google Scholar] [CrossRef]
  274. Alam, M.T.; Murshed, R.; Gomes, P.F.; Masud, Z.M.; Saber, S.; Chaklader, M.A.; Khanam, F.; Hossain, M.; Momen, A.B.I.M.; Yasmin, N.; et al. Ivermectin as Pre-Exposure Prophylaxis for COVID-19 among Healthcare Providers in a Selected Tertiary Hospital in Dhaka—An Observational Study. Eur. J. Med. Health Sci. 2020, 2. [Google Scholar] [CrossRef]
  275. Behera, P.; Patro, B.K.; Padhy, B.M.; Mohapatra, P.R.; Bal, S.K.; Chandanshive, P.D.; Mohanty, R.R.; Ravikumar, S.R.; Singh, A.; Singh, S.R.; et al. Prophylactic Role of Ivermectin in Severe Acute Respiratory Syndrome Coronavirus 2 Infection Among Healthcare Workers. Cureus 2021, 5, e16897. [Google Scholar] [CrossRef] [PubMed]
  276. Carvallo, H.; Hirsch, R.; Psaltis, A.; Contreras, V. Study of the Efficacy and Safety of Topical Ivermectin + Iota-Carrageenan in the Prophylaxis against COVID-19 in Health Personnel. J. Biomed. Res. Clin. Investig. 2020, 2, 1007. [Google Scholar] [CrossRef]
  277. Chahla, R.E.; Ruiz, L.M.; Ortega, E.S.; Morales, M.F.; Barreiro, F.; George, A.; Mancilla, C.; D’Amato, S.; Barrenechea, G.; Goroso, D.G.; et al. Intensive Treatment with Ivermectin and Iota-Carrageenan as Pre-Exposure Prophylaxis for COVID-19 in Health Care Workers from Tucuman, Argentina. Am. J. Ther. 2021, 28, E601. [Google Scholar] [CrossRef] [PubMed]
  278. Seet, R.C.S.; Quek, A.M.L.; Ooi, D.S.Q.; Sengupta, S.; Lakshminarasappa, S.R.; Koo, C.Y.; So, J.B.Y.; Goh, B.C.; Loh, K.S.; Fisher, D.; et al. Positive Impact of Oral Hydroxychloroquine and Povidone-Iodine Throat Spray for COVID-19 Prophylaxis: An Open-Label Randomized Trial. International. J. Infect. Dis. 2021, 106, 314. [Google Scholar] [CrossRef]
  279. Guerrero, R.; Bravo, L.E.; Muñoz, E.; Ardila, E.K.G.; Guerrero, E. COVID-19: The Ivermectin African Enigma. Colomb. Med. 2020, 51, 1–8. [Google Scholar] [CrossRef]
  280. Hellwig, M.D.; Maia, A. A COVID-19 Prophylaxis? Lower Incidence Associated with Prophylactic Administration of Ivermectin. Int. J. Antimicrob. Agents 2021, 57, 106248. [Google Scholar] [CrossRef]
  281. Available online: https://C19adoption.Com/ (accessed on 3 May 2022).
  282. Available online: https://www.Covid19treatmentguidelines.Nih.Gov/Therapies/Antiviral-Therapy/Ivermectin/ (accessed on 3 May 2022).
  283. Available online: https://www.Fda.Gov (accessed on 3 May 2022).
  284. López-Medina, E.; López, P.; Hurtado, I.C.; Dávalos, D.M.; Ramirez, O.; Mar-tínez, E.; Díazgranados, J.A.; Oñate, J.M.; Chavarriaga, H.; Herrera, S.; et al. Effect of Ivermectin on Time to Resolution of Symptoms among Adults with Mild COVID-19: A Randomized Clinical Trial. JAMA J. Am. Med. Assoc. 2021, 325, 1426–1435. [Google Scholar] [CrossRef]
  285. Sarhan, R.M.; Harb, H.S.; Abou, A.E.; Sa-lem-bekhit, M.M. Efficacy of the Early Treatment with Tocili-zumab-Hydroxychloroquine and Tocilizumab-Remdesivir in Severe COVID-19 Patients. J. Infect. Public Health. 2022, 15, 116. [Google Scholar] [CrossRef]
  286. Hazan, S.; Dolai, S.; Clancy, R.L.; Borody, T.J. Effectiveness of Ivermec-tin-Based Multidrug Therapy in Severely Hypoxic, Ambulatory COVID-19 Patients. Future Med. 2022, 17, 339. [Google Scholar] [CrossRef]
  287. Manivannan, E.; Karthikeyan, C.; Moorthy, N.S.H.N.; Chaturvedi, S.C. The Rise and Fall of Chloroquine/Hydroxychloroquine as Compassionate Therapy of COVID-19. Front. Pharmacol. 2021, 12, 584940. [Google Scholar] [CrossRef] [PubMed]
  288. Abubaker Bagabir, S.; Ibrahim, N.K.; Abubaker Bagabir, H.; Hashem Ateeq, R. COVID-19 and Artificial Intelligence: Genome Sequencing, Drug Development and Vaccine Discovery. J. Infect. Public Health 2022, 15, 289. [Google Scholar] [CrossRef] [PubMed]
  289. Beck, B.R.; Shin, B.; Choi, Y.; Park, S.; Kang, K. Predicting Commercially Avail-able Antiviral Drugs That May Act on the Novel Coronavirus (SARS-CoV-2) through a Drug-Target Interaction Deep Learning Model. Comput. Struct. Biotechnol. J. 2020, 18, 784. [Google Scholar] [CrossRef]
  290. Aghdam, R.; Habibi, M.; Taheri, G. Using Informative Features in Machine Learning Based Method for COVID-19 Drug Repurposing. J. Cheminform. 2021, 13, 70. [Google Scholar] [CrossRef]
  291. Ho, W.S.; Zhang, R.; Tan, Y.L.; Chai, C.L.L. COVID-19 and the Promise of Small Molecule Therapeutics: Are There Lessons to Be Learnt? Pharmacol. Res. 2022, 179, 106201. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Chemical structures of antiparasitic drugs reported to have anti-SARS-CoV-2 activity and included in this manuscript. Source: PubChem.
Figure 1. Chemical structures of antiparasitic drugs reported to have anti-SARS-CoV-2 activity and included in this manuscript. Source: PubChem.
Microorganisms 10 01284 g001
Figure 2. Mechanisms of action of antiparasitic drugs to interfere with SARS-CoV-2 viral infection at different levels: (a) during the infection cycle of the virus; (b) during the viral attachment to cellular receptors; and (c) as indirect immunomodulatory agents. Created with BioRender.com.
Figure 2. Mechanisms of action of antiparasitic drugs to interfere with SARS-CoV-2 viral infection at different levels: (a) during the infection cycle of the virus; (b) during the viral attachment to cellular receptors; and (c) as indirect immunomodulatory agents. Created with BioRender.com.
Microorganisms 10 01284 g002
Table 1. CQ and HCQ clinical trials against COVID-19 with published results.
Table 1. CQ and HCQ clinical trials against COVID-19 with published results.
Chloroquine (CQ)
Trial No.PhaseDrugsNo. ParticipantsStatus & Results
NCT04323527Phase 2CQ diphosphate: high dosage (600 mg twice daily for 10 days) vs. low-dosage (450 mg twice on day 1 and once daily for 4 days)278Completed. Higher CQ dosage should not be recommended for critically ill patients with COVID-19 because of its potential safety hazards, especially when taken concurrently with azithromycin and oseltamivir [76].
NCT04420247Phase 3CQ/HCQ added to standard of care (SoC)142Completed. The trial was stopped before reaching the planned sample size due to harmful effects. In patients with severe COVID-19, the use of CQ/HCQ added to SoC resulted in a significant worsening of clinical status, an increased risk of renal dysfunction and an increased need for invasive mechanical ventilation [99].
Hydroxychloroquine (HCQ)
Trial No.PhaseDrugsNo. ParticipantsStatus & Results
NCT04381936
“RECOVERY Trial”
Phase 2/Phase 3HCQ vs. SoC4716Completed (HCQ arm). COVID-19 patients receiving HCQ did not have a lower incidence of death at 28 days than those who received usual care [78].
NCT04315948
“SOLIDARITY Trial”
Phase 3HCQ vs. Remdesivir vs. Lopinavir/ritonavir vs. Interferon Beta vs. SoC vs. AZD7442 vs. Placebo2416Recruiting. HCQ has little or no effect on hospitalized COVID-19 patients according to mortality, initiation of ventilation and duration of hospital stay [84].
NCT04308668Phase 3HCQ vs. Placebo1312Completed. HCQ failed to prevent illness compatible with COVID-19 or confirmed infection when used as high-risk or moderate-risk postexposure prophylaxis within 4 days after exposure [80].
NCT04304053Phase 3HCQ as prophylactic treatment2300Completed. Postexposure therapy with HCQ did not prevent SARS-CoV-2 infection or symptomatic COVID-19 in healthy persons exposed to a PCR-positive case patient [81].
NCT04332991Phase 3HCQ vs. Placebo479Completed. Among adults hospitalized with respiratory illness from COVID-19, treatment with HCQ did not significantly improve clinical status at day 14 [100].
NCT04466540Phase 4HCQ vs. Placebo1372Completed. HCQ did not reduce the risk of hospitalization in outpatients with mild or moderate forms of COVID-19 [89].
NCT04321278Phase 3HCQ vs. HCQ + azithromycin447Completed. In patients with severe COVID-19, the use of HCQ + azithromycin did not improve clinical outcomes [101].
NCT04325893Phase 3HCQ vs. Placebo259Terminated (decrease in number of eligible patients). Trial involving mainly older patients with mild to moderate COVID-19. HCQ treatment did not result in better clinical or virological outcomes [88].
NCT04403100Phase 3HCQ Sulfate Tablets vs. Lopinavir/Ritonavir Oral Tablet vs. HCQ Sulfate Tablets +Lopinavir/Ritonavir Oral vs. Placebo1968Recruiting. Neither HCQ nor lopinavir-ritonavir showed any significant benefit for decreasing COVID-19–associated hospitalization or other secondary clinical outcomes [86].
NCT04354428Phase 2/Phase 3HCQ sulfate vs. HCQ+AZ300Active, not recruiting. HCQ and HCQ + AZ do not affect the clinical course of COVID-19 among outpatients and should not be used to treat SARS-CoV-2 infection [85].
Table 2. Artemisinin clinical trials with known results.
Table 2. Artemisinin clinical trials with known results.
Trial No.PhaseDrugsNo. ParticipantsStatus & Results
NCT05004753Phase 4ARTIVeda™
(Artemisinin) +/− SoC
120Completed. ARTIVeda™ provides a faster recovery of patients with mild-moderate COVID-19 (preliminary data on 60 patients) [136]
Table 3. Clinical trials involving niclosamide as antiviral agent for COVID-19 patients’ treatment with published results.
Table 3. Clinical trials involving niclosamide as antiviral agent for COVID-19 patients’ treatment with published results.
Trial No.PhaseDrugsNo. ParticipantsStatus & Results
NCT04399356Phase 2Niclosamide vs. Placebo73Completed. No significant difference in oropharyngeal clearance of SARS-CoV-2 at day 3 between placebo and niclosamide-treated groups [170].
Table 4. IVM: observational and clinical studies against COVID-19.
Table 4. IVM: observational and clinical studies against COVID-19.
Observational Studies
Trial No.PhaseDrugsNo. ParticipantsStatus & Results
NCT04434144NAIVM + Doxycycline/HCQ + AZ116Completed. Faster negative conversion of PCR and significantly faster symptom resolution in IVM-Doxycycline [255].
NCT04425863NAIVM + Aspirin/
IVM + Dexamethasone injection + Aspirin/
IVM + Dexamethasone injection + Enoxaparin injection
167Completed. A positive role of IVM + aspirin + dexamethasone + enoxaparin therapy. No hospitalization of mild cases and lower mortality rate compared to national rate [270].
Clinical Trials
Trial No.PhaseDrugsNo. ParticipantsStatus & Results
NCT043818842IVM and SoC vs. SoC45Completed. No differences between the two groups in viral load and in clinical evolution of the patients. Significant difference between patients that showed high IVM levels in plasma samples vs. untreated patients: correlation of IVM level with decrease of the viral load. High dose of IVM did not show toxicity [233].
NCT045916001/2IVM + Doxycycline/SoC140Completed. Therapy improvement by IVM + Doxycycline.
NCT043430921IVM vs. IVM + HCQ + AZT16Completed. Better efficacy of combination of IVM, HCQ and AZT; shorter hospitalization, and safety.
NCT044058432/3IVM vs. Placebo476Completed. No significant resolution of symptoms [284].
NCT045295252/3IVM vs. Placebo501Completed. No effect on preventing hospitalization of COVID-19 patients [247].
NCT043911273IVM vs. Placebo vs. HCQ108Completed. No efficacy of IVM or HCQ in decreasing hospitalization days, respiratory problems or deaths.
NCT045238313IVM + Doxycycline vs. SoC400Completed. Improvements in earlier recovery, prevention to progress to more serious disease (mortality) and increased likeliness to be COVID-19 negative by RT-PCR [258].
NCT047844811/2IVM254Completed. Reduction of the number of symptoms and improvement of clinical state.
NCT047017101/2IVM + Iota-carrageenan300Completed. Reduction of number of infected health workers with preventive treatment with IVM and Iotacarrigean. Prevention of severe disease.
NCT043900221/2IVM vs. Placebo24Completed. No difference between treated and untreated patients in the decrease of viral load but early recovery of anosmia and hyposmia in IVM-treated patients [234].
NCT044225612/3IVM chemoprophylaxis vs. no treatment304Completed. Significant differences among the two groups: 7.4% SARS-CoV-2 positive in IVM-treated vs. 58.4% of untreated subjects. Protective role of IVM [271].
NCT044461043IVM vs. HCQ vs. Zinc vs. Povidone-Iodine vs. Vitamin C4257Completed. No efficacy of IVM [278].
NCT044388502IVM vs. Placebo93Completed. Incidence dramatically dropped and is lack of eligible patients.
NCT046025072IVM vs. Placebo75Completed. Lack of severe COVID-19 cases in the place of study.
NCT043740192IVM vs. Camostat Mesilate vs. Artemesia annua vs. AS13Completed. Slow accrual.
NCT044314662IVM vs. SoC32Completed. IVM for SARS-CoV-2 treatment is safe. IVM antiviral effect is dose-dependent [230].
NCT047165692/3Intranasal IVM spray150Completed. Reduction of anosmia and rapid viral clearance in treated patients [231].
NCT047790474IVM vs. HCQ vs. Remdesivir vs. Tocilizumab vs. Lopinavir/Ritonavir 150150Completed. No positive effect of IVM treatment [285].
NCT044826861IVM + Doxycycline + Zinc + Vitamin D3 + Vitamin C31Completed. The combination of drugs is safe and effective [286].
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Calvo-Alvarez, E.; Dolci, M.; Perego, F.; Signorini, L.; Parapini, S.; D’Alessandro, S.; Denti, L.; Basilico, N.; Taramelli, D.; Ferrante, P.; et al. Antiparasitic Drugs against SARS-CoV-2: A Comprehensive Literature Survey. Microorganisms 2022, 10, 1284. https://doi.org/10.3390/microorganisms10071284

AMA Style

Calvo-Alvarez E, Dolci M, Perego F, Signorini L, Parapini S, D’Alessandro S, Denti L, Basilico N, Taramelli D, Ferrante P, et al. Antiparasitic Drugs against SARS-CoV-2: A Comprehensive Literature Survey. Microorganisms. 2022; 10(7):1284. https://doi.org/10.3390/microorganisms10071284

Chicago/Turabian Style

Calvo-Alvarez, Estefanía, Maria Dolci, Federica Perego, Lucia Signorini, Silvia Parapini, Sarah D’Alessandro, Luca Denti, Nicoletta Basilico, Donatella Taramelli, Pasquale Ferrante, and et al. 2022. "Antiparasitic Drugs against SARS-CoV-2: A Comprehensive Literature Survey" Microorganisms 10, no. 7: 1284. https://doi.org/10.3390/microorganisms10071284

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop