Next Article in Journal
Transcriptome and Proteomic Analysis Reveals Up-Regulation of Innate Immunity-Related Genes Expression in Caprine Herpesvirus 1 Infected Madin Darby Bovine Kidney Cells
Next Article in Special Issue
An Appraisal of the Current Scenario in Vaccine Research for COVID-19
Previous Article in Journal
Fishing for the Virome of Tropical Tuna
Previous Article in Special Issue
Effect of Aprotinin and Avifavir® Combination Therapy for Moderate COVID-19 Patients
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Potential Prophylactic Treatments for COVID-19

1
Chemical, Biological, Radiological and Nuclear Defense Division, Ministry of Defense, HaKirya, Tel-Aviv 61909, Israel
2
The Israel Defense Force Medical Corps, Tel Hashomer, Military Post 02149, Israel
3
Pulmonology Department, Edith Wolfson Medical Center, 62 Halochamim Street, Holon 5822012, Israel
4
Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
5
Israel Institute for Biological Research, Ness-Ziona 76100, Israel
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Viruses 2021, 13(7), 1292; https://doi.org/10.3390/v13071292
Submission received: 22 May 2021 / Revised: 22 June 2021 / Accepted: 28 June 2021 / Published: 2 July 2021
(This article belongs to the Special Issue Vaccines and Therapeutics against Coronaviruses)

Abstract

:
The World Health Organization declared the SARS-CoV-2 outbreak a Public Health Emergency of International Concern at the end of January 2020 and a pandemic two months later. The virus primarily spreads between humans via respiratory droplets, and is the causative agent of Coronavirus Disease 2019 (COVID-19), which can vary in severity, from asymptomatic or mild disease (the vast majority of the cases) to respiratory failure, multi-organ failure, and death. Recently, several vaccines were approved for emergency use against SARS-CoV-2. However, their worldwide availability is acutely limited, and therefore, SARS-CoV-2 is still expected to cause significant morbidity and mortality in the upcoming year. Hence, additional countermeasures are needed, particularly pharmaceutical drugs that are widely accessible, safe, scalable, and affordable. In this comprehensive review, we target the prophylactic arena, focusing on small-molecule candidates. In order to consolidate a potential list of such medications, which were categorized as either antivirals, repurposed drugs, or miscellaneous, a thorough screening for relevant clinical trials was conducted. A brief molecular and/or clinical background is provided for each potential drug, rationalizing its prophylactic use as an antiviral or inflammatory modulator. Drug safety profiles are discussed, and current medical indications and research status regarding their relevance to COVID-19 are shortly reviewed. In the near future, a significant body of information regarding the effectiveness of drugs being clinically studied for COVID-19 is expected to accumulate, in addition to information regarding the efficacy of prophylactic treatments.

1. Introduction

SARS-CoV-2 belongs to the Coronaviridae family of single-stranded RNA viruses, with a crown-like structure dictated by the spatial conformation of its spike proteins [1]. Coronavirus Disease 2019 (COVID-19), the disease resulting from exposure to this pathogen, is characterized by a pulmonary pathology which can progress to acute respiratory distress syndrome (ARDS), respiratory failure, and death [2]. The virus enters cells by binding to the angiotensin converting enzyme-2 (ACE-2) receptor expressed on subpopulations of pulmonary epithelial cells (type II epithelial cells), after which it is primed (cleavage of the spike protein) by the transmembrane serine 2 protease (TMPRSS2) as part of the fusion process with the cell membrane, and then internalized. ACE2 internalization and activation of the immune system [3] trigger pulmonary inflammation, characterized by increased levels of pro-inflammatory markers, such as interleukin-6 (IL-6) and C-reactive protein (CRP). This upregulation of inflammatory mediators progresses in some patients to a “cytokine storm” [4]. In severe cases, vascular damage, manifested by endothelial cell damage and hypercoagulation, is observed [5].
Severe morbidity and disease-related mortality are generally observed among patients aged 65 and above, as well as in individuals with comorbidities, e.g., obesity, hypertension, diabetes, heart disease, and others (herein, “at-risk population”) [6]. In addition, individuals who are in close contact with COVID-19 patients (i.e., healthcare professionals, social workers, and first-responders), as well as people living in densely populated areas or with a lifestyle that includes routine congregation (e.g., boarding schools and military units), are at higher risk of contracting the virus.
Currently, the only effective means to curb the COVID-19 pandemic is via widespread anti-SARS-CoV-2 vaccination. There are several vaccines available following emergency use authorization. However, it will take a long time until these vaccines are available for mass population vaccination on a global scale. Furthermore, vaccination is contraindicated in specific subpopulations (i.e., subjects suffering from severe allergic reaction to vaccine components) and little is known about the vaccines’ long-term efficacy. Logistic issues, such as storage, transportation, handling, and administration, also pose significant challenges. Therefore, other countermeasures are urgently required.
Apart from supportive treatment, which aims to ameliorate symptoms, provide invasive and non-invasive respiratory support, and prevent secondary complications, attempts are being made to establish a medical response via passive immunizations and pharmaceutical treatments. To date, few drugs have proven useful in treating COVID-19, most of which are administered in advanced COVID-19 disease states, i.e., steroids [7].
Prophylactic treatment for infectious diseases involves drug administration as early as possible and is generally given to healthy individuals [8]. Prophylaxis can be given before (pre-exposure) or after (post-exposure) encountering the pathogen, but prior to symptoms onset. The earlier infectious diseases are treated with antimicrobials or immunomodulatory drugs (anti-inflammatories), the better the prognosis. This is due to the lower inoculum and levels of inflammatory components (e.g., cytokines and neutrophils) at early stages of infection [9,10].
This work will review the current knowledge on COVID-19 prophylactics.

2. Methods

This review focuses on drugs reported as potentially effective for COVID-19 in the media, journals, as well as in pre-clinical or in vitro studies.
We examined pharmaceuticals with known anti-viral effects, including repurposed drugs and antibacterial drugs. A list of all relevant clinical studies, registered at the NIH website (clinicaltrails.gov) was constructed. Our search methodology in the above websites used the keywords combination of the “name of the drug” + “COVID-19”, filtering out irrelevant results. We compared all candidates’ performances based on their safety, known long-term efficacy, prophylactic usage, anti-viral coverage, and their registered clinical trials’ preliminary results. This methodology led to a list of 11 relevant drug candidates; 9 of whom have registered prophylactic studies (Table 1). After assembling the information, we summarized educated suggestions.
Inclusion criteria: from the initial search, only the studies that followed the below criteria were included in our article:
  • The drug is clinically evaluated for COVID-19.
  • The drug possesses regulatory approval from the US Food and Drug Administration (FDA), European Medicines Agency (EMA), The Medicines and Healthcare Products Regulatory Agency (MHRA), or the Pharmaceuticals and Medical Devices Agency (PMDA).
  • The drug is evaluated for prophylactic use for COVID-19/the drug is used prophylactically for other medical conditions/there’s data about previous long-term usage.

3. Current Prophylaxis Options for COVID-19

The drugs described below are currently being tested in clinical trials as COVID-19 prophylactics or as therapeutics with potential prophylactic applications (Table 2). Current status and information regarding the drugs is summarized in Table 3. The presented drugs are categorized as either antivirals, repurposed drugs, or miscellaneous.

3.1. Antivirals

3.1.1. Favipiravir

Favipiravir (T-705) is a prodrug which, after being metabolized, inhibits various viral RNA-dependent RNA polymerases (RdRps) [12]. SARS-CoV-2 RdRp activity has been found to be ten-fold higher than that of other viral RdRps; therefore, favipiravir is a good candidate for COVID-19 treatment [13]. Viruses shown to have in vitro susceptibility to this drug include polio, zika, western equine encephalitis (WEE), seasonal/pandemic influenza, rabies, ebola, arenaviruses [14], and Rift Valley fever [15]. Favipiravir has also demonstrated efficacy against pandemic influenza in a range of animal and clinical studies [16,17] and has been approved for clinical use in Japan to treat recurrent or pandemic flu. Moreover, favipiravir exhibits a very favorable clinical safety profile. Its main side effects include hyperuricemia, which is reversed upon discontinuation of treatment, and elevated liver enzyme level [18]. However, the drug is teratogenic and is therefore prohibited for use during pregnancy [19]. Prophylactic clinical use of favipiravir has been reported post-exposure to ebola [20] and rabies [21]. Owing to its broad activity against RNA viruses, favipiravir has been frequently suggested as a potential treatment for COVID-19, and has demonstrated a significant antiviral effect when administered to hamsters prior to SARS-CoV-2 infection (prophylaxis animal model) [22].
When favipiravir was tested in COVID-19 patients, viral clearance was shortened from 11 to 4 days, and 91% of the treated patients had improved pulmonary inflammation markers, compared to 62% in the control group [23]. In a randomized, comparative, open-label, multicenter, phase 3 clinical trial, the drug led to a significant shortening of clinical cure time in COVID-19 patients [24].
In the context of prophylaxis, a clinical study currently being conducted in Canada (NCT04448119, Phase 2) is assessing the efficacy of favipiravir treatment over 25 days in preventing infection in nursing homes (among the elderly, assisted-living patients, and healthcare professionals). Results obtained in another clinical trial indicated that early intervention with favipiravir is superior to late intervention in terms of viral clearance and time to defervescence, further supporting the notion of a potential benefit of prophylactic administration [25].

3.1.2. Combined Antiretroviral Medications

Lopinavir/Ritonavir

Lopinavir/ritonavir (LPV/r) has been FDA-approved since 2000 as a combination treatment for human immunodeficiency virus (HIV), and has demonstrated safety in patients without comorbidities [26]. The drug combination is safe for use in pregnant women, newborns [27,28], and infants [29].
Following demonstration of favorable clinical responses when administered to SARS-infected patients [30,31], LPV/r has been recommended for treatment of SARS-CoV-2 [32,33]. A study aiming to assess the efficacy of LPV/r alone in COVID-19 patients found no difference between the treatment and control cohorts [34].
These findings have led to treatment regimen changes and to the recommendation of the World Health Organization against LPV/r treatment in COVID-19 patients [35]. A recently published work presenting a comparative analysis of the literature related to the drug combination and coronaviruses proposed that blood concentrations of the drug suitable for HIV patients are insufficient for SARS-CoV-2 viral loads [36]. If proven correct, prophylactic LPV/r treatment at the regimens currently recommended for HIV may be effective when administered pre-exposure or at the early onset of exposure, when lower drug concentrations are required for a virucidal effect due to low inoculum. Recently published results summarizing a clinical trial in COVID-19 patients (NCT04379245) suggested that LPV/r would not be an effective preventive treatment. However, as it was a small-scale observational study [37], further assessments will be necessary before reaching final conclusions.
Two ongoing Phase 3 clinical studies are assessing the efficacy of prophylactic pre- and post (ring vaccination)—exposure treatments with LPV/r—NCT04321174 (recruiting) and NCT04364022. An additional Phase 3 study is evaluating the beneficial effects of prophylactic LPV/r treatment among healthcare workers (NCT04328285).

Emtricitabine/Tenofovir

Emtricitabine/tenofovir is a combination therapy used to treat and prevent HIV. The drug combination is composed of nucleoside analogues that impair viral genome replication. The combination is registered for use in adults and children under 12 years of age, and has been found effective in reducing risk of infection in virus-negative adults and as a pre-exposure prophylactic.
HIV exploits RNA-dependent DNA polymerase (RdDp), while coronaviruses use, as mentioned, RdRp. However, in vitro studies have unexpectedly demonstrated that the drug effectively inhibits the SARS-CoV-2 RdRp as well [38]. In particular, emtricitabine/tenofovir combination (the combination in DESCOVY and TRUVADA) was shown to be a terminator for the SARS-CoV-2 RdRp catalyzed reaction [39]. In light of the safety profile and potential anti-SARS-CoV-2 effects, the emtricitabine/tenofovir drug combination is currently being tested as a COVID-19 prophylactic. An ongoing Phase 3 clinical study is assessing the efficacy of a 12 week prophylaxis emtricitabine/tenofovir regimen (NCT04334928) in healthcare workers in contact with COVID-19-confirmed patients in private hospitals in Spain. Several additional studies aiming to use this drug combination in a preventive manner (NCT04519125, NCT04405271) are registered for Phase 2/3 and Phase 3, respectively, in the NIH website (clinicaltrials.gov), but are not yet recruiting participants.

3.2. Repurposed Drugs

3.2.1. Ivermectin

Ivermectin is an antiparasitic, FDA-approved (for adolescents as well as adults), inexpensive drug used for the long-term treatment of scabies, lice, river blindness, and other parasitic diseases. The drug is derived from Streptomyces avermitilis and is administered mainly via the oral route. Its main side effects at human dosages are eye redness and dry/burning sensations of the skin. More serious side effects are rare and include central nervous system (CNS) suppression due to potentiation of the gamma aminobutyric acid (GABA) neurotransmitter synapses.
Regarding its antiparasitic effect, the drug binds glutamate-gated chloride channels on nerve and muscle cells of invertebrates, leading to increased permeability of parasite membranes to chloride ions, subsequently resulting in cell hyperpolarization, paralysis, and death. Its antiviral activity may be achieved by its inhibition of importin α/β, which is responsible for viral entry into the nucleus [40,41]. Studies have shown that the drug has antiviral potential against chikungunya, yellow fever, West Nile, Venezuelan equine encephalitis virus (VEEV), dengue, and influenza. The drug is also being tested as a prophylactic treatment for malaria [42].
The drug potentially possesses a dual anti-SARS-CoV-2 effect, namely antiviral and anti-inflammatory. Regarding its antiviral effect, the drug has shown a significant anti-SARS-CoV-2 effect in vitro with a 5000-fold reduction in viral RNA in cells within 48 h of exposure. However, the dosage required to inhibit SARS-CoV-2 replication in cells is equivalent to 10,000-fold the dosage currently approved for human use [40]. Furthermore, plasma concentrations of the drug after oral administration are estimated to be too low to be effective in inhibiting viral replication [43,44]. Regarding its anti-inflammatory properties, ivermectin was shown to inhibit lipopolysaccharide (LPS)-induced prostaglandin E2 and nitric oxide production in cell culture [45]. In vivo, ivermectin inhibits the production of inflammatory cytokines (IL-6, IL-1β, TNFα) and improved survival in mice exposed to LPS [46]; it was also found to be an allosteric modulator of the α7 nicotinic acetylcholine receptor (an essential regulator of inflammation [47]) in hamsters [48]. There are currently many clinical studies testing ivermectin alone or in combination with other anti-COVID-19 treatments. A retrospective review of the medical files of COVID-19 patients in four Florida hospitals found a significant reduction in mortality rates among patients who received the drug [49]. A randomized, controlled, and double-blinded trial demonstrated a significantly shorter period of time to SARS-CoV-2 negativity in parallel to increased oxygen saturation, following intravenous administration of ivermectin. The efficacy of ivermectin was dose-dependent and no serious adverse events were reported [50].
Furthermore, several studies evaluating the potential of its prophylactic use demonstrated highly encouraging results. Prophylactic ivermectin treatment of healthcare workers in India was associated with a 73% reduction in COVID-19 infection rates [51].
The drug has also been tested for its post-exposure prophylactic effects and was found to prevent symptoms among COVID-19 household contacts (NCT04422561), where 7.4% of the subjects in the treated group developed symptoms, compared to 58.4% of those treated with placebo.
Nine additional clinical trials are registered in the NIH website, three of which were completed and one large-scale study (4257 subjects) is in course of its third phase (Table 3). In addition to the clinical evidence, it was suggested that countries with routine mass administration of prophylactic chemotherapy, including ivermectin, have a significantly lower incidence of COVID-19 [52].

3.2.2. Interferons

Interferons (IFNs) are endogenous proteins secreted following the penetration of pathogens into the body, which activate the immune system [53] and serve as a standard treatment for a range of diseases, including viral diseases, i.e., hepatitis C virus (HCV) and hepatitis B virus (HBV) [54].
IFN-α is a key IFN used in antiviral treatment [55]. The drug affects cellular regulation and prevents viral replication, while activating the adenylate cyclase enzyme on membrane receptors, which activates intracellular antiviral processes in DNA and RNA virus-mediated infections. IFN-α treatment of mice exposed to bacterial lipopolysaccharide (LPS) was associated with increased survival of mice suffering from pulmonary damage (ARDS). The increased survival was associated with the delayed onset of damage caused by neutrophil infiltration into the lungs [56]. Various IFNs have demonstrated long-term prevention of various virus-related infections, such as rhinovirus, influenza, Middle East respiratory syndrome (MERS), and SARS [57,58]. An in vitro study demonstrated their efficacy against SARS at interferon concentrations similar to those found in human serum [59]. A non-human primate study showed that treatment with a IFN-α nasal spray might prevent or reduce the pathology caused by SARS [60]. Furthermore, a clinical trial evaluating the efficacy of a recombinant human IFN-α (rhIFN-α 2b)-containing spray given twice daily to over 14,000 participants found the drug effective in preventing respiratory diseases induced by influenza, RSV, and adenoviruses, with a highly favorable safety profile [61].
Adverse effects due to IFN- α have been described in many organ systems [62]. Many side-effects are dose-dependent. The most common are flu-like symptoms, hematological toxicity, elevated hepatic transaminases, nausea, fatigue, and psychiatric sequelae. A special consideration regarding IFN treatment for SARS-CoV-2 are the IFN autoantibodies formation, which were found in severe COVID-19 patients [63,64]. It should be mentioned, though, that, although this could potentially hamper IFN bioactivity and treatment outcome, as in the case of severe COVID-19 patients, the relevance of autoantibody formation during prophylactic treatment, namely in healthy or in pre-symptomatic subjects, is yet to be determined.
A significant volume of evidence has recently accumulated regarding the potential of IFN treatment of COVID-19. SARS-CoV-2 was shown to inhibit IFN production both in vitro (membrane protein (M)-dependent inhibition) and in vivo [65]. Thus, exogenous administration of IFNs may compensate for this insufficiency [66]. In addition, PEGylated human interferon lambda-1 (PEG-IFN-λ-1a) potently delayed SARS-CoV-2 replication in epithelial cells, and prophylactic (pre-exposure) or therapeutic administration significantly lowered pulmonary viral load in a mouse model [67]. Furthermore, it was suggested that administration of recombinant or PEGylated forms of IFN-λ suppresses viral replication while preventing the onset of a “cytokine storm” [68].
There are many clinical trials currently assessing the efficacy of multiple interferons (IFN-λ-1a, IFN-β-1b, IFN-β-1a, novaferon, IFN-α, and PEG-IFN-λ-1a) in COVID-19 patients. A non-controlled study conducted in Wuhan, China weighing the efficacy of IFN-α-2b in 77 patients found that the treatment led to a significant decline in viral load in the upper respiratory tract and in IL-6 and CRP levels [69], emphasizing the potential dual effect (anti-viral and anti-inflammatory) of this drug in the course of COVID-19 treatment.
Prophylactic IFN treatments are being assessed in ongoing clinical trials (all of which are recruiting subjects to the studies, see Table 3). A Phase 3 study (NCT04320238) is evaluating the prophylactic efficacy and safety of rhIFN-α-1b nasal drops in healthy medical workers [70]. Another Phase 3 clinical trial in Chile is evaluating the post-exposure prophylactic use of three subcutaneous injections of PEG-IFNβ-1a in household contacts (NCT04552379), and a Phase 2b study conducted in the USA is evaluating the efficacy of PEG-IFN-λ-1a as a post-exposure prophylactic in high-risk, non-hospitalized individuals following household exposure (NCT04344600). A Phase 3 clinical trial is evaluating pre- and post-exposure prophylactic uses of IFNα nasal drops in oncology patients in Australia (NCT04534725).

3.2.3. Nitazoxanide

Nitazoxanide is an anti-protozoal drug belonging to the thiazolides family, which disrupts critical energy-generating pathways required for pathogen survival and replication. Nitazoxanide is a prodrug, which, after hydrolysis and conjugation, becomes an active metabolite [71].
The drug is orally administered, and has been cumulatively used by over 75 million patients across the globe [72]. Accumulated data has shown the drug to be very safe for use in humans. Furthermore, due to its low price, the drug can be affordable worldwide [73].
Nitazoxanide has demonstrated antimicrobial activity and is currently used as a broad-spectrum antiviral medication. It has been proven to be effective in chronic hepatitis patients and is also indicated for the treatment of influenza. The drug leads to reduced HIV replication and, in parallel, stimulates immune memory responses [74].
The antiviral mechanism of nitazoxanide is likely to work via activation of eukaryotic translation initiation factor 2α, which serves as an intracellular antiviral factor [75]. In coronaviruses, the drug also inhibits nucleocapsid (N) protein expression and viral replication. Moreover, studies performed in patients infected with MERS and other coronaviruses showed a reduction in proinflammatory cytokine expression [72]. Nitazoxanide inhibited IL-6 production in a mouse inflammation model [76]. Therefore, in addition to its antiviral effects, nitazoxanide may serve as an immunomodulatory drug and suppress the intensity of the “cytokine storm” in COVID-19 patients.
Preclinical and clinical trials are being performed to evaluate the efficacy of nitazoxanide against SARS-CoV-2 [72,77]. Encouraging results have been obtained in in vitro models, which showed a high correlation between the attainable maximum plasma concentrations (Cmax) and the dosage required to inhibit SARS-CoV-2 replication [78]. To date, several studies are being conducted in COVID-19 patients. In most of the studies, the drug is being combined with other drugs, but some are evaluating the efficacy of nitazoxanide as monotherapy in patients with mild-to-moderate diseases. Early administration of nitazoxanide to mild COVID-19 patients has recently been reported to reduce viral loads; no serious adverse events were reported [79](NCT04552483). Two Phase 3 studies are currently being conducted; one is aiming to assess prevention of outbreaks among healthcare workers in frequent contact with confirmed COVID-19 patients at early stages of the disease with mild symptoms (NCT04359680), and the other is evaluating post-exposure prophylaxis in nursing home residents (NCT04343248). A Phase 2 study (NCT04561063) is exploring the efficacy of nitazoxanide in preventing COVID-19 in healthcare workers at high risk of exposure. Another Phase 2 study (NCT04435314, not yet recruiting) plans to determine the effect of post-exposure nitazoxanide administration to volunteers who are at high-risk of infection. A Phase 4 study recruits volunteers in Argentina (NCT04788407).

3.2.4. Bromhexine Hydrochloride

The mechanism of SARS-CoV-2 entry into lung epithelial cells involves binding the viral spike protein to the human ACE2 receptor, after which it is cleaved at two sites (which enables a conformational change and fusion) by the TMPRSS2 enzyme, as detailed above. Therefore, inhibition of this enzyme is an attractive target for COVID-19 treatment [3].
The role of TMPRSS2 in viral fusion with lung cells was previously demonstrated for SARS; it has been established that, in addition to its role in disease onset, it is involved in viral antibody evasion [80,81]. Furthermore, it has been proposed that TMPRSS2 inhibition by nafamostat is effective against MERS [82].
The expectorant drug bromhexine hydrochloride (HCl) has shown to inhibit TMPRSS2 (repurposed effect) [83] and has a favorable safety profile [83]. Additionally, it is low-priced and requires no prescription in many countries. Although preliminary data suggest that bromhexine HCl mucolytic doses are sub-optimal for the treatment of SARS-CoV-2 [84], there is some encouraging, though inconclusive, evidence for its repurposing potential for COVID-19 treatment [85,86].
Two clinical trials are evaluating the drug’s efficacy as COVID-19 prophylaxis for healthcare workers; one is an ongoing Phase 1 clinical trial assessing oral administration (8 mg three times daily for two months, NCT04340349), and another is a small-scale trial in 50 subjects, which was completed (NCT04405999). Results are anticipated.

3.3. Miscellaneous

3.3.1. Doxycycline

Doxycycline is a tetracycline antibacterial isolated from Streptomyces aureofaciens used to treat a broad range of infections. The drug is chiefly bacteriostatic and employs its antimicrobial effect via protein synthesis inhibition [87]. Its side effects include developing a rash, fever, lymph node swelling, flu-like symptoms, and yellow shading of the skin and eyes. In addition, doxycycline intake is associated with photosensitivity and may cause diarrhea. More severe side effects are rare. The drug is contraindicated during pregnancy (after the 18th week) and in children under eight years of age (except for emergencies) [88].
Doxycycline has been found effective against viral infections such as HIV and West Nile [89]. In a study that tested the effect of antibiotics on dengue-virus progression, doxycycline significantly inhibited the viral serine protease enzyme and significantly reduced viral replication and invasion into cells [90]. Doxycycline was also proven to reduce neurological deficits in a Zika virus mouse model [91]. Similarly, studies have suggested that tetracyclines may inhibit replication of single-stranded RNA viruses [92], including SARS-CoV-2, both in terms of cell entry and viral replication [93].
Doxycycline inhibits matrix metalloproteinases (MMPs), and, as coronaviruses exploit MMPs for a range of their essential activities (replication, cell infection, and survival), the drug may have high efficacy against SARS-CoV-2 [94]. According to other studies, doxycycline may delay COVID-19 progression via anti-inflammatory activities, including regulating the NFκB pathway and inhibition of proinflammatory cytokine levels (IL-6, IL-1β, TNFα), measured during ARDS in severely ill COVID-19 patients [95]. Earlier studies demonstrated the effectiveness of chemically modified tetracyclines against SARS, where septic shock and ARDS development were prevented [96].
Therefore, doxycycline treatment of COVID-19 patients may be effective. To date, there are several clinical trials registered on the NIH website testing its effectiveness against COVID-19. These studies are in Phases 2, 3, and 4. Although only one study evaluates doxycycline as a prophylactic agent for COVID-19 (NCT04584567, recruiting for phase 3), the drug has a prophylaxis indication, for example, in malaria. Moreover, doxycycline is administered for prolonged durationa as a post-exposure therapeutic against Q fever caused by Coxiella burnetti [97], indicating broad safety margins. Therefore, doxycycline may be a suitable prophylactic (primarily post-exposure) agent against COVID-19. Supporting the potential benefit of doxycycline prophylaxis for COVID-19, early treatment in high-risk patients with moderate-to-severe COVID-19 infections in non-hospital settings was associated with early clinical recovery, decreased hospitalization, and decreased mortality [98]. Several case reports may also support the notion of a beneficial effect of doxycycline preventive treatment. In a series of four patients with COVID-19 infection and known high-risk pulmonary disease who were placed on standard doses of doxycycline as monotherapy for a course between 5 and 14 days, a rapid clinical improvement was recorded with no safety issues noted [99]. Nevertheless, after reviewing the interim analysis of the doxycycline arm of the PRINCIPLE trial held in the UK, it was concluded that there is no beneficial effect in patients aged over 50 who are treated at home with doxycycline during the early stage of COVID-19 [100].

3.3.2. Famotidine

Famotidine is a histamine-2 receptor antagonist used to treat heartburn, gastroesophageal reflux, and peptic (stomach and duodenum) ulcers. Side effects generally include fatigue, headaches, abdominal pain, and diarrhea. Severe side effects are relatively rare [101]. Laboratory studies have found the drug to inhibit HIV replication [102]. A bioinformatic study identified the drug as a candidate inhibitor of 3-chymotrypsin-like protease (3CLpro) involved in SARS-CoV-2 replication [103]. Apart from 3CLpro inhibition, famotidine has been suggested to impart an immunomodulatory effect, since lower ferritin levels were measured in patients receiving the drug [104].
A retrospective cohort study found that COVID-19 subjects who had received famotidine (at any dosage or administration route) within 24 h of hospitalization had approximately 2-fold lower mortality and intubation rates. The treatment was seemingly given before the onset of symptoms as a treatment for heartburn, and, in most cases, it was given prior to SARS-CoV-2 infection. Famotidine self-administered by 10 non-hospitalized COVID-19 patients was found safe at high dosages, and was associated with significant self-reported symptomatic improvement [105]. To date, several clinical trials registered on the NIH website are enrolling participants to evaluate the efficacy of famotidine in COVID-19 patients, including outpatients.
In light of the high safety profile, potential antiviral effects, clinical findings mentioned above, and the possibility of long-term treatment, the drug may be suitable as a COVID-19 prophylactic. It should be mentioned, though, that several studies demonstrated conflicting data regarding famotidine efficacy in COVID-19 patients [105,106]; therefore, its clinical impact should be further validated in randomized controlled trials, particularly for prophylactic use.

3.3.3. Nitric Oxide

Nitric oxide (NO) is a neurotransmitter that interacts with many molecules, including DNA, proteins, and thiol-containing molecules. NO plays a critical role in immune system activation [107], where its derivatives (reactive nitrogen species) are generated by macrophages and other cells in response to cytokines and microbial substances. Moreover, all isoforms of the NO synthase (NOS) enzyme play a central role in the innate and adaptive immune responses.
As a smooth muscle relaxant, leading to pulmonary vasodilation, inhaled NO (iNO) is used for long-term treatment of pulmonary hypertension-associated hypoxia in infants. In a recent clinical trial conducted in several hospitals in Israel which assessed the efficacy of NO treatment in infants suffering from acute viral bronchitis [108], there were no reports of severe side effects, even after high dosage treatment (160 ppm) as an adjunct to oxygen (O2). O2 saturation in infants treated with iNO was higher and their recovery time was shorter. Safety was also demonstrated following long-term treatment (7–12 months) of adults with pulmonary hypertension [109].
Studies have shown that iNO may be an effective antiviral treatment. The literature shows that NO and its derivatives inhibit inflammatory processes via IFNγ-mediated mechanisms [110]. Furthermore, inverse correlations between NO levels and activity of viral enzymes (reductase and protease) that play essential roles in viral replication and activity have been reported [111,112]. Its antiviral effects against influenza viruses (influenza A & B), coronaviruses, vaccinia virus, herpes simplex type 1, and ectromelia have been shown in laboratory studies [113,114,115]. A small-scale study assessing the efficacy of NO treatment in patients with SARS found it to reduce viral spread in the lungs and to increase O2 saturation [116]. The recently developed nitric oxide release solution (NORS, developed by SaNOtize), which enables a delayed release of NO through a nasal spray/wash/gargling solution, was tested in the University of Utah and was found to inhibit influenza A and SARS-CoV-2 in vitro.
To date, several clinical trials are assessing the efficacy of NO treatment in COVID-19 patients. A Phase 2/3 study (NCT04842331) is recruiting 600 subjects to evaluate the efficacy of RESP301 (NO generating solution) as a post-exposure prophylaxis in household residents in the UK. Another study (NCT04312243, Phase 2) is assessing the prophylactic efficacy of NO among medical workers in close contact with confirmed SARS-CoV-2-positive patients. Additional study is assessing the efficacy of NO nasal spray (GLS-1200) given 3 times daily for 4 weeks to prevent SARS-CoV-2 infection among healthcare workers in close contact with confirmed COVID-19 patients (NCT04408183, Phase 2). An ongoing Phase 2 study (NCT04337918) is assessing NORS use as a prophylactic for individuals who came into close contact with confirmed SARS-CoV-2-positive patients. A small Phase 2 study (NCT04858451, not yet recruiting) will be conducted in order to evaluate the efficacy of RESP301 in patients at risk of viral infection (COPD and bronchiectasis patients).

3.3.4. Colchicine

Colchicine is a drug orally or intravenously administered as a long-term prophylaxis against gout, familial Mediterranean fever (FMF), and recurrent pericarditis. The drug inhibits microtubule polymerization, disrupting cell division [117]. Moreover, the drug inhibits neutrophil migration to sites of inflammation, thereby serving as an anti-inflammatory [118]. Colchicine use in pregnancy and breastfeeding is controversial with an FDA class C recommendation, although recent data suggests a more lenient approach regarding its usage during pregnancy and breastfeeding [119,120,121]. Side effects include a variety of symptoms ranging from mild and frequent (e.g., gastrointestinal symptoms such as vomiting, nausea, and abdominal pain) to rare and serious (e.g., hematological dyscrasia, myalgia, neuropathy, confusion, and convulsions) [117,122,123,124,125].
The drug is absorbed by immune cells, granulocytes, and monocytes within 24–72 h of oral administration, inducing an anti-inflammatory effect. It has been recently shown that the drug also inhibits NLRP3 inflammasome [126]. This finding is of importance, since SARS-CoV-2 has been demonstrated to activate inflammatory processes by activation of NLRP3. Specifically, NLRP3 activation takes place early in the SARS-CoV-2 infection, initiating the “cytokine storm” [127].
Colchicine is currently being evaluated in clinical trials among COVID-19 patients. In particular, recent results obtained in 4159 non-hospitalized patients have associated colchicine use with reduced risk of death and hospitalization compared to placebo. Hospitalization rates were reduced by 25%, the need for mechanical ventilation by 50%, and deaths by 44%. Treatment was initiated within 24 h of symptom onset, and inclusion criteria included at least one significant risk factor [128]. Another Phase 3 study is assessing colchicine treatment of 954 confirmed SARS-CoV-2-positive non-hospitalized patients aged 60 years and above who were at risk of developing respiratory complications (NCT04416334). An additional Phase 2/3 study (NCT04492358) is underway in Spain to evaluate the efficacy of combining colchicine and prednisone for moderate/severe COVID-19 in a vulnerable population (geriatric hospital unit/transitional care center residents).
If those studies yield positive results, and the drug proves effective in adult patients, it will lay the foundation for more extensive studies to assess its prophylactic efficacy in COVID-19 patients, particularly in elderly populations at risk of complications and mortality, with emphasis on post-exposure prophylactic administration.

4. Discussion

To date, nearly 180 million people worldwide have been infected with SARS-CoV-2, with approximately four million deaths suffered. While several vaccines have been approved for emergency use, there is currently a severe shortage in their global supply. Additionally, vaccine efficacy is yet to be determined, in terms of longevity and coverage of emerging SARS-CoV-2 variants, current and future [129]. Furthermore, clinical vaccine studies in children and pregnant women are only just being conducted, currently leaving these populations unvaccinated. Likewise, severely immunocompromised patients cannot be effectively vaccinated, nor can the severely allergic. Hence, there is an urgent need for applicable treatment modalities, particularly pharmaceutical drugs.
The goal of this review is to highlight the potential for prophylactic efficacy of several approved and safe drugs. Favipiravir, IFNs, ivermectin, nitazoxanide, bromhexine HCl, and NO seem to be promising candidates. In particular, Ivermectin, which dramatically reduced COVID-19 infection rates or symptoms when administered as pre-exposure or post-exposure prophylaxis, respectively. In addition to ivermectin, early treatment of at-risk hospitalized COVID-19 patients with colchicine was associated with a significant reduction in the risk of both death and the need for mechanical ventilation, and its efficacy is currently being evaluated in non-hospitalized patients aged 60 years and above. Thus, colchicine also shows high potential for prophylactic treatment (post-exposure). Lastly, early intervention with doxycycline was shown to be beneficial in a clinical trial conducted in the USA. Nevertheless, as mentioned, current reports from the PRINCIPLE trial in the UK have shown conflicting outcomes. A possible explanation for this discrepancy is the earlier drug administration in the USA trial (up to 12 h of symptom onset, in comparison to 14 days within symptoms onset in the UK trial), further supporting its prophylactic rather than therapeutic potential. Additionally, the majority of the patients in the USA trial received zinc and calcium supplements in addition to doxycycline [98]. In this regard, it would be interesting to follow the consequences of the trial (NCT04584567) currently evaluating the prophylactic potential of doxycycline co-administered with zinc. Regarding IFNs, although seemingly efficacious and safe, their high cost (Table 2) limits their potential usage for widespread prophylaxis.
It should be mentioned that combination treatments, especially with safe drugs possessing different mechanisms of action, could be of great advantage. For example, early triple antiviral therapy (LPV/r, IFN-β-1b and ribavirin) was safe and superior to LPV/r alone in alleviating symptoms and shortening viral shedding duration and hospitalization in patients with mild-to-moderate COVID-19 symptoms [130]. Furthermore, co-incubation of remdesivir and ivermectin in cell culture conferred synergistic anti-SARS-CoV-2 effects [131]. Additionally, several clinical trials are currently evaluating concomitant administration of two drugs from the list depicted in this manuscript, i.e., nitazoxanide and ivermectin (NCT04360356), nitazoxanide, and favipiravir (NCT04918927, early intervention), as well as ivermectin and doxycycline (NCT04523831 and NCT04729140—completed and phase 4, respectively). If found beneficial, it is worth evaluating these combinations as prophylactic treatments, depending on the combinations’ safety profiles.
Major drawbacks of some drugs tested for COVID-19 are their extensive side effects, e.g., chloroquine/hydroxychloroquine [132], and absorption issues which require intravenous administration, e.g., remdesivir [133,134]. Inhalation or intranasal administration routes of these drugs may overcome, at least partially, these limitations, improving their suitability for prophylactic usage. Indeed, some clinical trials are evaluating the therapeutic impact of inhaled hydroxychloroquine (NCT04477083, NCT04461353) and remdesivir (NCT04480333). Further developments regarding administration routes should be monitored.
Many of the drugs presented in this review can be characterized as broad-spectrum antivirals/antimicrobials (favipiravir, interferons, ivermectin, nitazoxanide, bromexine HCl, and NO). Extensive antiviral and antimicrobial coverage, including against influenza, could be a dual advantage, particularly during the winter season when healthcare system burdens are particularly high.
In addition to small molecule drugs for COVID-19 prophylaxis, several additional approaches not reviewed here may be relevant for prophylaxis, as detailed below.
Vitamins, dietary supplements, and antioxidants are being assessed as both preventive and adjunct-therapeutic agents. Due to their high safety profiles and indirect beneficial effects (primarily in the elderly population, who often suffer from dietary deficits or vitamin deficiencies), recommendation of usage of these products, which are being tested in clinical trials, should be considered. Moreover, due to frequent lockdowns, rates of malnutrition and vitamin D insufficiency may have increased. These supplements were extensively reviewed elsewhere [133,134] and therefore were not included in this review.
Antibody-based treatment may be a promising strategy for both pre- and post-exposure prophylaxis [132,135]. Yet, this treatment is costly compared to small molecule-based treatment, rendering it unsuitable for mass administration. Furthermore, the approach is time-consuming, requiring a parenteral route of administration and medical surveillance by highly trained medical personnel in dedicated medical facilities. This approach may also be ineffective against highly mutated variants, particularly when monoclonal antibodies are used [136].
Long-term boosting of innate immune response using vaccines which are not directed against SARS-CoV-2 (immunostimulants) are under extensive clinical evaluation for COVID-19 prevention. Decreased immune functioning may lead to a loss of control of viral replication and, as a result, increased disease severity. Long-term boosting of innate immunity by various types of immunization strategies may cause a nonspecific stimulation of the immune system. In many cases, the immediate and effective activation of the innate immune system is sufficient to cope with the invading pathogen, even without activating adaptive immunity. Long-term changes manifested by enhanced innate immune activity have been demonstrated in individuals who received live vaccines [137]. In light of these findings, various live attenuated vaccines designed originally to prevent infection with a pathogen of interest (other than SARS-CoV-2) are being clinically tested for the treatment of COVID-19. This includes the Bacillus Clamette-Guérin (BCG) vaccine which effectively protects against various infectious diseases, as well as the VPM1002 (a genetically engineered BCG strain with higher immunogenicity) [138], the oral polio vaccine, the tuberculosis vaccine [139], and the varicella-zoster vaccine (NCT04523246), which are all being clinically tested for the prevention or alleviation of COVID-19.
In summary, clinically approved drugs with well-established and favorable safety profiles repurposed as a COVID-19 prophylaxis should be considered for at-risk individuals, as well as first responders and medical teams. It is imperative to keep abreast on this topic with clinical research developments across the globe.

Funding

This research received no external funding.

Acknowledgments

We would like to thank Avigdor Scherz for thorough discussions and generous assistance in the manuscript preparation. We would also like to acknowledge Yehudit Posen and Itai Glinert for their professional translation and linguistic editing.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Park, W.B.; Kwon, N.J.; Choi, S.J.; Kang, C.K.; Choe, P.G.; Kim, J.Y.; Yun, J.; Lee, G.W.; Seong, M.W.; Kim, N.J.; et al. Virus Isolation from the First Patient with SARS-CoV-2 in Korea. J. Korean Med. Sci. 2020, 35, e84. [Google Scholar] [CrossRef] [PubMed]
  2. Yang, X.; Yu, Y.; Xu, J.; Shu, H.; Xia, J.; Liu, H.; Wu, Y.; Zhang, L.; Yu, Z.; Fang, M.; et al. Clinical Course and Outcomes of Critically Ill Patients with SARS-CoV-2 Pneumonia in Wuhan, China: A Single-Centered, Retrospective, Observational Study. Lancet Respir. Med. 2020, 8, 475–481. [Google Scholar] [CrossRef] [Green Version]
  3. 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] [PubMed]
  4. Pedersen, S.F.; Ho, Y.-C. SARS-CoV-2: A Storm Is Raging. J. Clin. Invest. 2020, 130, 2202–2205. [Google Scholar] [CrossRef]
  5. Ackermann, M.; Verleden, S.E.; Kuehnel, M.; Haverich, A.; Welte, T.; Laenger, F.; Vanstapel, A.; Werlein, C.; Stark, H.; Tzankov, A.; et al. Pulmonary Vascular Endothelialitis, Thrombosis, and Angiogenesis in Covid-19. N. Engl. J. Med. 2020, 383, 120–128. [Google Scholar] [CrossRef] [PubMed]
  6. Richardson, S.; Hirsch, J.S.; Narasimhan, M.; Crawford, J.M.; McGinn, T.; Davidson, K.W.; The Northwell COVID-19 Research Consortium; Barnaby, D.P.; Becker, L.B.; Chelico, J.D.; et al. Presenting Characteristics, Comorbidities, and Outcomes Among 5700 Patients Hospitalized with COVID-19 in the New York City Area. JAMA 2020, 323, 2052–2059. [Google Scholar] [CrossRef]
  7. Tomazini, B.M.; Maia, I.S.; Cavalcanti, A.B.; Berwanger, O.; Rosa, R.G.; Veiga, V.C.; Avezum, A.; Lopes, R.D.; Bueno, F.R.; Silva, M.V.A.O.; et al. Effect of Dexamethasone on Days Alive and Ventilator-Free in Patients with Moderate or Severe Acute Respiratory Distress Syndrome and COVID-19: The CoDEX Randomized Clinical Trial. JAMA 2020, 324, 1307–1316. [Google Scholar] [CrossRef] [PubMed]
  8. Pre-Exposure Prophylaxis (PrEP)|HIV Risk and Prevention|HIV/AIDS|CDC. Available online: https://www.cdc.gov/hiv/risk/prep/index.html (accessed on 22 June 2021).
  9. Eagle, H. The Effect of the Size of the Inoculum and the Age of the Infection on the Curative Dose of Penicillin in Experimental Infections with Streptococci, Pneumococci, and Treponema Pallidum. J. Exp. Med. 1949, 90, 595–607. [Google Scholar] [CrossRef] [PubMed]
  10. Stevens, D.L.; Gibbons, A.E.; Bergstrom, R.; Winn, V. The Eagle Effect Revisited: Efficacy of Clindamycin, Erythromycin, and Penicillin in the Treatment of Streptococcal Myositis. J. Infect. Dis. 1988, 158, 23–28. [Google Scholar] [CrossRef] [PubMed]
  11. Drugs.Com|Prescription Drug Information, Interactions & Side Effects. Available online: https://www.drugs.com/ (accessed on 28 March 2021).
  12. Delang, L.; Abdelnabi, R.; Neyts, J. Favipiravir as a Potential Countermeasure against Neglected and Emerging RNA Viruses. Antiviral Res. 2018, 153, 85–94. [Google Scholar] [CrossRef] [PubMed]
  13. Shannon, A.; Selisko, B.; Le, N.-T.-T.; Huchting, J.; Touret, F.; Piorkowski, G.; Fattorini, V.; Ferron, F.; Decroly, E.; Meier, C.; et al. Rapid Incorporation of Favipiravir by the Fast and Permissive Viral RNA Polymerase Complex Results in SARS-CoV-2 Lethal Mutagenesis. Nat. Commun. 2020, 11, 4682. [Google Scholar] [CrossRef]
  14. Furuta, Y.; Komeno, T.; Nakamura, T. Favipiravir (T-705), a Broad Spectrum Inhibitor of Viral RNA Polymerase. Proc. Jpn. Acad. Ser. B Phys. Biol. Sci. 2017, 93, 449–463. [Google Scholar] [CrossRef] [Green Version]
  15. Borrego, B.; de Ávila, A.I.; Domingo, E.; Brun, A. Lethal Mutagenesis of Rift Valley Fever Virus Induced by Favipiravir. Antimicrob. Agents Chemother. 2019, 63, e00669-19. [Google Scholar] [CrossRef] [Green Version]
  16. Furuta, Y.; Gowen, B.B.; Takahashi, K.; Shiraki, K.; Smee, D.F.; Barnard, D.L. Favipiravir (T-705), a Novel Viral RNA Polymerase Inhibitor. Antiviral Res. 2013, 100, 446–454. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Hayden, F.G.; Shindo, N. Influenza Virus Polymerase Inhibitors in Clinical Development. Curr. Opin. Infect. Dis. 2019, 32, 176–186. [Google Scholar] [CrossRef]
  18. Mishima, E.; Anzai, N.; Miyazaki, M.; Abe, T. Uric Acid Elevation by Favipiravir, an Antiviral Drug. Tohoku J. Exp. Med. 2020, 251, 87–90. [Google Scholar] [CrossRef]
  19. Louchet, M.; Sibiude, J.; Peytavin, G.; Picone, O.; Tréluyer, J.-M.; Mandelbrot, L. Placental Transfer and Safety in Pregnancy of Medications under Investigation to Treat Coronavirus Disease 2019. Am. J. Obstet. Gynecol. MFM 2020, 2, 100159. [Google Scholar] [CrossRef] [PubMed]
  20. Jacobs, M.; Aarons, E.; Bhagani, S.; Buchanan, R.; Cropley, I.; Hopkins, S.; Lester, R.; Martin, D.; Marshall, N.; Mepham, S.; et al. Post-Exposure Prophylaxis against Ebola Virus Disease with Experimental Antiviral Agents: A Case-Series of Health-Care Workers. Lancet Infect. Dis. 2015, 15, 1300–1304. [Google Scholar] [CrossRef]
  21. Yamada, K.; Noguchi, K.; Komeno, T.; Furuta, Y.; Nishizono, A. Efficacy of Favipiravir (T-705) in Rabies Postexposure Prophylaxis. J. Infect. Dis. 2016, 213, 1253–1261. [Google Scholar] [CrossRef] [Green Version]
  22. 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, 26955–26965. [Google Scholar] [CrossRef]
  23. Cai, Q.; Yang, M.; Liu, D.; Chen, J.; Shu, D.; Xia, J.; Liao, X.; Gu, Y.; Cai, Q.; Yang, Y.; et al. Experimental Treatment with Favipiravir for COVID-19: An Open-Label Control Study. Eng. Beijing China 2020, 6, 1192–1198. [Google Scholar] [CrossRef] [PubMed]
  24. Udwadia, Z.F.; Singh, P.; Barkate, H.; Patil, S.; Rangwala, S.; Pendse, A.; Kadam, J.; Wu, W.; Caracta, C.F.; Tandon, M. Efficacy and Safety of Favipiravir, an Oral RNA-Dependent RNA Polymerase Inhibitor, in Mild-to-Moderate COVID-19: A Randomized, Comparative, Open-Label, Multicenter, Phase 3 Clinical Trial. Int. J. Infect. Dis. IJID Off. Publ. Int. Soc. Infect. Dis. 2021, 103, 62–71. [Google Scholar] [CrossRef] [PubMed]
  25. Doi, Y.; Hibino, M.; Hase, R.; Yamamoto, M.; Kasamatsu, Y.; Hirose, M.; Mutoh, Y.; Homma, Y.; Terada, M.; Ogawa, T.; et al. A Prospective, Randomized, Open-Label Trial of Early versus Late Favipiravir Therapy in Hospitalized Patients with COVID-19. Antimicrob. Agents Chemother. 2020, 64. [Google Scholar] [CrossRef] [PubMed]
  26. Vogel, M.; Rockstroh, J.K. Safety of Lopinavir/Ritonavir for the Treatment of HIV-Infection. Expert Opin. Drug Saf. 2005, 4, 403–420. [Google Scholar] [CrossRef]
  27. Tuccori, M.; Convertino, I.; Ferraro, S.; Cappello, E.; Valdiserra, G.; Focosi, D.; Blandizzi, C. The Impact of the COVID-19 “Infodemic” on Drug-Utilization Behaviors: Implications for Pharmacovigilance. Drug Saf. 2020, 43, 699–709. [Google Scholar] [CrossRef]
  28. Pasley, M.V.; Martinez, M.; Hermes, A.; d’Amico, R.; Nilius, A. Safety and Efficacy of Lopinavir/Ritonavir during Pregnancy: A Systematic Review. AIDS Rev. 2013, 15, 38–48. [Google Scholar]
  29. Lopinavir/Ritonavir (Kaletra) Use During Pregnancy. Available online: https://www.drugs.com/pregnancy/lopinavir-ritonavir.html (accessed on 28 March 2021).
  30. Chu, C.M.; Cheng, V.C.C.; Hung, I.F.N.; Wong, M.M.L.; Chan, K.H.; Chan, K.S.; Kao, R.Y.T.; Poon, L.L.M.; Wong, C.L.P.; Guan, Y.; et al. Role of Lopinavir/Ritonavir in the Treatment of SARS: Initial Virological and Clinical Findings. Thorax 2004, 59, 252–256. [Google Scholar] [CrossRef] [Green Version]
  31. Chan, K.S.; Lai, S.T.; Chu, C.M.; Tsui, E.; Tam, C.Y.; Wong, M.M.L.; Tse, M.W.; Que, T.L.; Peiris, J.S.M.; Sung, J.; et al. Treatment of Severe Acute Respiratory Syndrome with Lopinavir/Ritonavir: A Multicentre Retrospective Matched Cohort Study. Hong Kong Med. J. Xianggang Yi Xue Za Zhi 2003, 9, 399–406. [Google Scholar]
  32. Meini, S.; Pagotto, A.; Longo, B.; Vendramin, I.; Pecori, D.; Tascini, C. Role of Lopinavir/Ritonavir in the Treatment of Covid-19: A Review of Current Evidence, Guideline Recommendations, and Perspectives. J. Clin. Med. 2020, 9, 2050. [Google Scholar] [CrossRef]
  33. Magro, P.; Zanella, I.; Pescarolo, M.; Castelli, F.; Quiros-Roldan, E. Lopinavir/Ritonavir: Repurposing an Old Drug for HIV Infection in COVID-19 Treatment. Biomed. J. 2021, 44, 43–53. [Google Scholar] [CrossRef] [PubMed]
  34. Cao, B.; Wang, Y.; Wen, D.; Liu, W.; Wang, J.; Fan, G.; Ruan, L.; Song, B.; Cai, Y.; Wei, M.; et al. A Trial of Lopinavir–Ritonavir in Adults Hospitalized with Severe Covid-19. N. Engl. J. Med. 2020, 382, 1787–1799. [Google Scholar] [CrossRef] [PubMed]
  35. WHO Discontinues Hydroxychloroquine and Lopinavir/Ritonavir Treatment Arms for COVID-19. Available online: https://www.who.int/news/item/04-07-2020-who-discontinues-hydroxychloroquine-and-lopinavir-ritonavir-treatment-arms-for-covid-19 (accessed on 22 June 2021).
  36. Smolders, E.J.; Te Brake, L.H.; Burger, D.M. SARS-CoV-2 and HIV Protease Inhibitors: Why Lopinavir/Ritonavir Will Not Work for COVID-19 Infection. Antivir. Ther. 2020. [Google Scholar] [CrossRef]
  37. Charre, C.; Icard, V.; Pradat, P.; Brochier, C.; Lina, B.; Chidiac, C.; Cotte, L. Coronavirus Disease 2019 Attack Rate in HIV-Infected Patients and in Preexposure Prophylaxis Users. AIDS 2020, 34, 1765–1770. [Google Scholar] [CrossRef] [PubMed]
  38. Elfiky, A.A. Ribavirin, Remdesivir, Sofosbuvir, Galidesivir, and Tenofovir against SARS-CoV-2 RNA Dependent RNA Polymerase (RdRp): A Molecular Docking Study. Life Sci. 2020, 253, 117592. [Google Scholar] [CrossRef] [PubMed]
  39. Jockusch, S.; Tao, C.; Li, X.; Anderson, T.K.; Chien, M.; Kumar, S.; Russo, J.J.; Kirchdoerfer, R.N.; Ju, J. Triphosphates of the Two Components in DESCOVY and TRUVADA Are Inhibitors of the SARS-CoV-2 Polymerase. bioRxiv 2020. [Google Scholar] [CrossRef]
  40. 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. Antiviral Res. 2020, 178, 104787. [Google Scholar] [CrossRef] [PubMed]
  41. Jans, D.A.; Martin, A.J.; Wagstaff, K.M. Inhibitors of Nuclear Transport. Curr. Opin. Cell Biol. 2019, 58, 50–60. [Google Scholar] [CrossRef]
  42. Varghese, F.S.; Kaukinen, P.; Gläsker, S.; Bespalov, M.; Hanski, L.; Wennerberg, K.; Kümmerer, B.M.; Ahola, T. Discovery of Berberine, Abamectin and Ivermectin as Antivirals against Chikungunya and Other Alphaviruses. Antiviral Res. 2016, 126, 117–124. [Google Scholar] [CrossRef] [PubMed]
  43. Bray, M.; Rayner, C.; Noël, F.; Jans, D.; Wagstaff, K. Ivermectin and COVID-19: A Report in Antiviral Research, Widespread Interest, an FDA Warning, Two Letters to the Editor and the Authors’ Responses. Antiviral Res. 2020, 178, 104805. [Google Scholar] [CrossRef]
  44. Schmith, V.D.; Zhou, J.J.; Lohmer, L.R.L. The Approved Dose of Ivermectin Alone Is Not the Ideal Dose for the Treatment of COVID-19. Clin. Pharmacol. Ther. 2020, 108, 762–765. [Google Scholar] [CrossRef]
  45. Zhang, X.; Song, Y.; Xiong, H.; Ci, X.; Li, H.; Yu, L.; Zhang, L.; Deng, X. Inhibitory Effects of Ivermectin on Nitric Oxide and Prostaglandin E2 Production in LPS-Stimulated RAW 264.7 Macrophages. Int. Immunopharmacol. 2009, 9, 354–359. [Google Scholar] [CrossRef] [PubMed]
  46. Zhang, X.; Song, Y.; Ci, X.; An, N.; Ju, Y.; Li, H.; Wang, X.; Han, C.; Cui, J.; Deng, X. Ivermectin Inhibits LPS-Induced Production of Inflammatory Cytokines and Improves LPS-Induced Survival in Mice. Inflamm. Res. 2008, 57, 524–529. [Google Scholar] [CrossRef] [PubMed]
  47. Wang, H.; Yu, M.; Ochani, M.; Amella, C.A.; Tanovic, M.; Susarla, S.; Li, J.H.; Wang, H.; Yang, H.; Ulloa, L.; et al. Nicotinic Acetylcholine Receptor Alpha7 Subunit Is an Essential Regulator of Inflammation. Nature 2003, 421, 384–388. [Google Scholar] [CrossRef]
  48. De Melo, G.D.; Lazarini, F.; Larrous, F.; Feige, L.; Kergoat, L.; Marchio, A.; Pineau, P.; Lecuit, M.; Lledo, P.-M.; Changeux, J.-P.; et al. Anti-COVID-19 Efficacy of Ivermectin in the Golden Hamster. bioRxiv 2020. [Google Scholar] [CrossRef]
  49. 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, 85–92. [Google Scholar] [CrossRef] [PubMed]
  50. 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 Randomised Controlled Double Blind Dose Response Study in Lagos. medRxiv 2021. [Google Scholar] [CrossRef]
  51. Behera, P.; Patro, B.K.; Singh, A.K.; Chandanshive, P.D.; Pradhan, S.K.; Pentapati, S.S.K.; Batmanabane, G.; Mohapatra, P.R.; Padhy, B.M.; Bal, S.K.; 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]
  52. 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]
  53. Kumaki, Y.; Ennis, J.; Rahbar, R.; Turner, J.D.; Wandersee, M.K.; Smith, A.J.; Bailey, K.W.; Vest, Z.G.; Madsen, J.R.; Li, J.K.-K.; et al. Single-Dose Intranasal Administration with MDEF201 (Adenovirus Vectored Mouse Interferon-Alpha) Confers Protection from Mortality in a Lethal SARS-CoV BALB/c Mouse Model. Antiviral Res. 2011, 89, 75. [Google Scholar] [CrossRef] [PubMed]
  54. Robek, M.D.; Boyd, B.S.; Chisari, F.V. Lambda Interferon Inhibits Hepatitis B and C Virus Replication. J. Virol. 2005, 79, 3851–3854. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Antoniou, K.M.; Ferdoutsis, E.; Bouros, D. Interferons and Their Application in the Diseases of the Lung. Chest 2003, 123, 209–216. [Google Scholar] [CrossRef] [Green Version]
  56. Kudo, D.; Uno, K.; Aoyagi, T.; Akahori, Y.; Ishii, K.; Kanno, E.; Maruyama, R.; Kushimoto, S.; Kaku, M.; Kawakami, K. Low-Dose Interferon-α Treatment Improves Survival and Inflammatory Responses in a Mouse Model of Fulminant Acute Respiratory Distress Syndrome. Inflammation 2013, 36, 812–820. [Google Scholar] [CrossRef]
  57. Gao, L.; Yu, S.; Chen, Q.; Duan, Z.; Zhou, J.; Mao, C.; Yu, D.; Zhu, W.; Nie, J.; Hou, Y. A Randomized Controlled Trial of Low-Dose Recombinant Human Interferons Alpha-2b Nasal Spray to Prevent Acute Viral Respiratory Infections in Military Recruits. Vaccine 2010, 28, 4445–4451. [Google Scholar] [CrossRef] [PubMed]
  58. Scagnolari, C.; Vicenzi, E.; Bellomi, F.; Stillitano, M.G.; Pinna, D.; Poli, G.; Clementi, M.; Dianzani, F.; Antonelli, G. Increased Sensitivity of SARS-Coronavirus to a Combination of Human Type I and Type II Interferons. Antivir. Ther. 2004, 9, 1003–1011. [Google Scholar]
  59. Bellingan, G.; Maksimow, M.; Howell, D.C.; Stotz, M.; Beale, R.; Beatty, M.; Walsh, T.; Binning, A.; Davidson, A.; Kuper, M.; et al. The Effect of Intravenous Interferon-Beta-1a (FP-1201) on Lung CD73 Expression and on Acute Respiratory Distress Syndrome Mortality: An Open-Label Study. Lancet Respir. Med. 2014, 2, 98–107. [Google Scholar] [CrossRef]
  60. Gao, H.; Zhang, L.; Wei, Q.; Duan, Z.; Tu, X.; Yu, Z.; Deng, W.; Zhang, L.; Bao, L.; Zhang, B.; et al. Preventive and therapeutic effects of recombinant IFN-alpha2b nasal spray on SARS-CoV infection in Macaca mulata. Zhonghua Shi Yan He Lin Chuang Bing Xue Za Zhi Zhonghua Shiyan He Linchuang Bingduxue Zazhi Chin. J. Exp. Clin. Virol. 2005, 19, 207–210. [Google Scholar]
  61. Chen, Q.; Zhang, L.; Yu, D.; Yu, Z.; Liu, Y.; Zhang, L.; Li, Z.; Duan, Z.; Wang, B.; Wei, X.; et al. A field trial for evaluating the safety of recombinant human interferon alpha-2b for nasal spray. Zhonghua Shi Yan He Lin Chuang Bing Xue Za Zhi Zhonghua Shiyan He Linchuang Bingduxue Zazhi Chin. J. Exp. Clin. Virol. 2005, 19, 211–215. [Google Scholar]
  62. Sleijfer, S.; Bannink, M.; Van Gool, A.R.; Kruit, W.H.J.; Stoter, G. Side Effects of Interferon-Alpha Therapy. Pharm. World Sci. PWS 2005, 27, 423–431. [Google Scholar] [CrossRef]
  63. Bastard, P.; Rosen, L.B.; Zhang, Q.; Michailidis, E.; Hoffmann, H.-H.; Zhang, Y.; Dorgham, K.; Philippot, Q.; Rosain, J.; Béziat, V.; et al. Autoantibodies against Type I IFNs in Patients with Life-Threatening COVID-19. Science 2020, 370, eabd4585. [Google Scholar] [CrossRef] [PubMed]
  64. De Prost, N.; Bastard, P.; Arrestier, R.; Fourati, S.; Mahévas, M.; Burrel, S.; Dorgham, K.; Gorochov, G.; Tandjaoui-Lambiotte, Y.; Azzaoui, I.; et al. Plasma Exchange to Rescue Patients with Autoantibodies Against Type I Interferons and Life-Threatening COVID-19 Pneumonia. J. Clin. Immunol. 2021, 41, 536–544. [Google Scholar] [CrossRef]
  65. Acharya, D.; Liu, G.; Gack, M.U. Dysregulation of Type I Interferon Responses in COVID-19. Nat. Rev. Immunol. 2020, 20, 397–398. [Google Scholar] [CrossRef]
  66. Zheng, Y.; Zhuang, M.-W.; Han, L.; Zhang, J.; Nan, M.-L.; Zhan, P.; Kang, D.; Liu, X.; Gao, C.; Wang, P.-H. Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Membrane (M) Protein Inhibits Type I and III Interferon Production by Targeting RIG-I/MDA-5 Signaling. Signal Transduct. Target. Ther. 2020, 5, 1–13. [Google Scholar] [CrossRef]
  67. Dinnon, K.H.; Leist, S.R.; Schäfer, A.; Edwards, C.E.; Martinez, D.R.; Montgomery, S.A.; West, A.; Yount, B.L.; Hou, Y.J.; Adams, L.E.; et al. A Mouse-Adapted Model of SARS-CoV-2 to Test COVID-19 Countermeasures. Nature 2020, 586, 560–566. [Google Scholar] [CrossRef] [PubMed]
  68. Andreakos, E.; Tsiodras, S. COVID-19: Lambda Interferon against Viral Load and Hyperinflammation. EMBO Mol. Med. 2020, 12, e12465. [Google Scholar] [CrossRef] [PubMed]
  69. Zhou, Q.; Chen, V.; Shannon, C.P.; Wei, X.-S.; Xiang, X.; Wang, X.; Wang, Z.-H.; Tebbutt, S.J.; Kollmann, T.R.; Fish, E.N. Interferon-A2b Treatment for COVID-19. Front. Immunol. 2020, 11. [Google Scholar] [CrossRef] [PubMed]
  70. Meng, Z.; Wang, T.; Chen, L.; Chen, X.; Li, L.; Qin, X.; Li, H.; Luo, J. An Experimental Trial of Recombinant Human Interferon Alpha Nasal Drops to Prevent COVID-19 in Medical Staff in an Epidemic Area. medRxiv 2020. [Google Scholar] [CrossRef] [Green Version]
  71. Trabattoni, D.; Gnudi, F.; Ibba, S.V.; Saulle, I.; Agostini, S.; Masetti, M.; Biasin, M.; Rossignol, J.-F.; Clerici, M. Thiazolides Elicit Anti-Viral Innate Immunity and Reduce HIV Replication. Sci. Rep. 2016, 6, 27148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Rossignol, J.-F. Nitazoxanide, a New Drug Candidate for the Treatment of Middle East Respiratory Syndrome Coronavirus. J. Infect. Public Health 2016, 9, 227–230. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. 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, 52–60. [Google Scholar] [CrossRef]
  74. Rossignol, J.-F. Nitazoxanide: A First-in-Class Broad-Spectrum Antiviral Agent. Antiviral Res. 2014, 110, 94–103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Elazar, M.; Liu, M.; McKenna, S.A.; Liu, P.; Gehrig, E.A.; Puglisi, J.D.; Rossignol, J.-F.; Glenn, J.S. The Anti-Hepatitis C Agent Nitazoxanide Induces Phosphorylation of Eukaryotic Initiation Factor 2alpha via Protein Kinase Activated by Double-Stranded RNA Activation. Gastroenterology 2009, 137, 1827–1835. [Google Scholar] [CrossRef]
  76. 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, 23–27. [Google Scholar] [CrossRef] [PubMed]
  77. Kelleni, M.T. Nitazoxanide/Azithromycin Combination for COVID-19: A Suggested New Protocol for Early Management. Pharmacol. Res. 2020, 157, 104874. [Google Scholar] [CrossRef] [PubMed]
  78. Rajoli, R.K.; Pertinez, H.; Arshad, U.; Box, H.; Tatham, L.; Curley, P.; Neary, M.; Sharp, J.; Liptrott, N.J.; Valentijn, A.; et al. Dose Prediction for Repurposing Nitazoxanide in SARS-CoV-2 Treatment or Chemoprophylaxis. medRxiv 2020. [Google Scholar] [CrossRef] [PubMed]
  79. Rocco, P.R.M.; Silva, P.L.; Cruz, F.F.; Junior, M.A.C.M.; Tierno, P.F.G.M.M.; Moura, M.A.; De Oliveira, L.F.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. Eur. Respir. J. 2021. [Google Scholar] [CrossRef]
  80. Matsuyama, S.; Nagata, N.; Shirato, K.; Kawase, M.; Takeda, M.; Taguchi, F. Efficient Activation of the Severe Acute Respiratory Syndrome Coronavirus Spike Protein by the Transmembrane Protease TMPRSS2. J. Virol. 2010, 84, 12658–12664. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Glowacka, I.; Bertram, S.; Müller, M.A.; Allen, P.; Soilleux, E.; Pfefferle, S.; Steffen, I.; Tsegaye, T.S.; He, Y.; Gnirss, K.; et al. Evidence That TMPRSS2 Activates the Severe Acute Respiratory Syndrome Coronavirus Spike Protein for Membrane Fusion and Reduces Viral Control by the Humoral Immune Response. J. Virol. 2011, 85, 4122–4134. [Google Scholar] [CrossRef] [Green Version]
  82. Yamamoto, M.; Matsuyama, S.; Li, X.; Takeda, M.; Kawaguchi, Y.; Inoue, J.-I.; Matsuda, Z. Identification of Nafamostat as a Potent Inhibitor of Middle East Respiratory Syndrome Coronavirus S Protein-Mediated Membrane Fusion Using the Split-Protein-Based Cell-Cell Fusion Assay. Antimicrob. Agents Chemother. 2016, 60, 6532–6539. [Google Scholar] [CrossRef] [Green Version]
  83. Maggio, R.; Corsini, G.U. Repurposing the Mucolytic Cough Suppressant and TMPRSS2 Protease Inhibitor Bromhexine for the Prevention and Management of SARS-CoV-2 Infection. Pharmacol. Res. 2020, 157, 104837. [Google Scholar] [CrossRef]
  84. Fu, Q.; Zheng, X.; Zhou, Y.; Tang, L.; Chen, Z.; Ni, S. Re-Recognizing Bromhexine Hydrochloride: Pharmaceutical Properties and Its Possible Role in Treating Pediatric COVID-19. Eur. J. Clin. Pharmacol. 2021, 77, 261–263. [Google Scholar] [CrossRef] [PubMed]
  85. Ansarin, K.; Tolouian, R.; Ardalan, M.; Taghizadieh, A.; Varshochi, M.; Teimouri, S.; Vaezi, T.; Valizadeh, H.; Saleh, P.; Safiri, S.; et al. Effect of Bromhexine on Clinical Outcomes and Mortality in COVID-19 Patients: A Randomized Clinical Trial. BioImpacts BI 2020, 10, 209–215. [Google Scholar] [CrossRef]
  86. Li, T.; Sun, L.; Zhang, W.; Zheng, C.; Jiang, C.; Chen, M.; Chen, D.; Dai, Z.; Bao, S.; Shen, X. Bromhexine Hydrochloride Tablets for the Treatment of Moderate COVID-19: An Open-Label Randomized Controlled Pilot Study. Clin. Transl. Sci. 2020, 13, 1096–1102. [Google Scholar] [CrossRef] [PubMed]
  87. MacDougall, C. Goodman & Gilman’s: The Pharmacological Basis of Therapeutic, 13th ed.; McGraw-Hill: New York, NY, USA, 2013. [Google Scholar]
  88. Gaillard, T.; Briolant, S.; Madamet, M.; Pradines, B. The End of a Dogma: The Safety of Doxycycline Use in Young Children for Malaria Treatment. Malar. J. 2017, 16, 148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Zakeri, B.; Wright, G.D. Chemical Biology of Tetracycline Antibiotics. Biochem. Cell Biol. 2008, 86, 124–136. [Google Scholar] [CrossRef] [PubMed]
  90. Rothan, H.A.; Mohamed, Z.; Paydar, M.; Rahman, N.A.; Yusof, R. Inhibitory Effect of Doxycycline against Dengue Virus Replication in Vitro. Arch. Virol. 2014, 159, 711–718. [Google Scholar] [CrossRef] [PubMed]
  91. Abrams, R.P.M.; Yasgar, A.; Teramoto, T.; Lee, M.-H.; Dorjsuren, D.; Eastman, R.T.; Malik, N.; Zakharov, A.V.; Li, W.; Bachani, M.; et al. Therapeutic Candidates for the Zika Virus Identified by a High-Throughput Screen for Zika Protease Inhibitors. Proc. Natl. Acad. Sci. USA 2020, 117, 31365–31375. [Google Scholar] [CrossRef]
  92. Phillips, J.M.; Gallagher, T.; Weiss, S.R. Neurovirulent Murine Coronavirus JHM.SD Uses Cellular Zinc Metalloproteases for Virus Entry and Cell-Cell Fusion. J. Virol. 2017, 91. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Gendrot, M.; Andreani, J.; Jardot, P.; Hutter, S.; Delandre, O.; Boxberger, M.; Mosnier, J.; Le Bideau, M.; Duflot, I.; Fonta, I.; et al. In Vitro Antiviral Activity of Doxycycline against SARS-CoV-2. Molecules 2020, 25, 5064. [Google Scholar] [CrossRef]
  94. Dutta, K.; Basu, A. Use of Minocycline in Viral Infections. Indian J. Med. Res. 2011, 133, 467–470. [Google Scholar] [PubMed]
  95. Henehan, M.; Montuno, M.; De Benedetto, A. Doxycycline as an Anti-Inflammatory Agent: Updates in Dermatology. J. Eur. Acad. Dermatol. Venereol. JEADV 2017, 31, 1800–1808. [Google Scholar] [CrossRef]
  96. Griffin, M.O.; Fricovsky, E.; Ceballos, G.; Villarreal, F. Tetracyclines: A Pleitropic Family of Compounds with Promising Therapeutic Properties. Review of the Literature. Am. J. Physiol. Cell Physiol. 2010, 299, C539–C548. [Google Scholar] [CrossRef] [Green Version]
  97. Anderson, A.; Bijlmer, H.; Fournier, P.-E.; Graves, S.; Hartzell, J.; Kersh, G.J.; Limonard, G.; Marrie, T.J.; Massung, R.F.; McQuiston, J.H.; et al. Diagnosis and Management of Q Fever--United States, 2013: Recommendations from CDC and the Q Fever Working Group. MMWR Recomm. Rep. Morb. Mortal. Wkly. Rep. Recomm. Rep. 2013, 62, 1–30. [Google Scholar]
  98. Alam, M.M.; Mahmud, S.; Rahman, M.M.; Simpson, J.; Aggarwal, S.; Ahmed, Z. Clinical Outcomes of Early Treatment with Doxycycline for 89 High-Risk COVID-19 Patients in Long-Term Care Facilities in New York. Cureus 2020, 12. [Google Scholar] [CrossRef]
  99. Yates, P.A.; Newman, S.A.; Oshry, L.J.; Glassman, R.H.; Leone, A.M.; Reichel, E. Doxycycline Treatment of High-Risk COVID-19-Positive Patients with Comorbid Pulmonary Disease. Ther. Adv. Respir. Dis. 2020, 14, 1753466620951053. [Google Scholar] [CrossRef] [PubMed]
  100. Butler, C.C.; Dorward, J.; Yu, L.-M.; Gbinigie, O.; Hayward, G.; Saville, B.R.; Hecke, O.V.; Berry, N.; Detry, M.; Saunders, C.; et al. Azithromycin for Community Treatment of Suspected COVID-19 in People at Increased Risk of an Adverse Clinical Course in the UK (PRINCIPLE): A Randomised, Controlled, Open-Label, Adaptive Platform Trial. Lancet 2021, 397, 1063–1074. [Google Scholar] [CrossRef]
  101. Humphries, T.J.; Merritt, G.J. Review Article: Drug Interactions with Agents Used to Treat Acid-Related Diseases. Aliment. Pharmacol. Ther. 1999, 13 (Suppl. S3), 18–26. [Google Scholar] [CrossRef]
  102. Bourinbaiar, A.S.; Fruhstorfer, E.C. The Effect of Histamine Type 2 Receptor Antagonists on Human Immunodeficiency Virus (HIV) Replication: Identification of a New Class of Antiviral Agents. Life Sci. 1996, 59, 365–370. [Google Scholar] [CrossRef]
  103. Wu, C.; Liu, Y.; Yang, Y.; Zhang, P.; Zhong, W.; Wang, Y.; Wang, Q.; Xu, Y.; Li, M.; Li, X.; et al. Analysis of Therapeutic Targets for SARS-CoV-2 and Discovery of Potential Drugs by Computational Methods. Acta Pharm. Sin. B 2020, 10, 766–788. [Google Scholar] [CrossRef]
  104. Freedberg, D.E.; Conigliaro, J.; Wang, T.C.; Tracey, K.J.; Callahan, M.V.; Abrams, J.A.; Famotidine Research Group Famotidine. Use Is Associated with Improved Clinical Outcomes in Hospitalized COVID-19 Patients: A Propensity Score Matched Retrospective Cohort Study. Gastroenterology 2020, 159, 1129–1131.e3. [Google Scholar] [CrossRef]
  105. Cheung, K.S.; Hung, I.F.N.; Leung, W.K. Association Between Famotidine Use and COVID-19 Severity in Hong Kong: A Territory-Wide Study. Gastroenterology 2021, 160, 1898–1899. [Google Scholar] [CrossRef]
  106. Zhou, J.; Wang, X.; Lee, S.; Wu, W.K.K.; Cheung, B.M.Y.; Zhang, Q.; Tse, G. Proton Pump Inhibitor or Famotidine Use and Severe COVID-19 Disease: A Propensity Score-Matched Territory-Wide Study. Gut 2020. [Google Scholar] [CrossRef] [PubMed]
  107. Bogdan, C. Nitric Oxide and the Immune Response. Nat. Immunol. 2001, 2, 907–916. [Google Scholar] [CrossRef]
  108. Goldbart, A.; Golan-Tripto, I.; Pillar, G.; Livnat-Levanon, G.; Efrati, O.; Spiegel, R.; Lubetzky, R.; Lavie, M.; Carmon, L.; Ghaffari, A.; et al. Inhaled Nitric Oxide Therapy in Acute Bronchiolitis: A Multicenter Randomized Clinical Trial. Sci. Rep. 2020, 10, 9605. [Google Scholar] [CrossRef] [PubMed]
  109. Barst, R.J.; Channick, R.; Ivy, D.; Goldstein, B. Clinical Perspectives with Long-Term Pulsed Inhaled Nitric Oxide for the Treatment of Pulmonary Arterial Hypertension. Pulm. Circ. 2012, 2, 139–147. [Google Scholar] [CrossRef] [Green Version]
  110. Reiss, C.S.; Komatsu, T. Does Nitric Oxide Play a Critical Role in Viral Infections? J. Virol. 1998, 72, 4547–4551. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Benz, D.; Cadet, P.; Mantione, K.; Zhu, W.; Stefano, G. Tonal Nitric Oxide and Health—A Free Radical and a Scavenger of Free Radicals. Med. Sci. Monit. Int. Med. J. Exp. Clin. Res. 2002, 8, RA1–RA4. [Google Scholar]
  112. Saura, M.; Zaragoza, C.; McMillan, A.; Quick, R.A.; Hohenadl, C.; Lowenstein, J.M.; Lowenstein, C.J. An Antiviral Mechanism of Nitric Oxide: Inhibition of a Viral Protease. Immunity 1999, 10, 21–28. [Google Scholar] [CrossRef] [Green Version]
  113. Akerström, S.; Mousavi-Jazi, M.; Klingström, J.; Leijon, M.; Lundkvist, A.; Mirazimi, A. Nitric Oxide Inhibits the Replication Cycle of Severe Acute Respiratory Syndrome Coronavirus. J. Virol. 2005, 79, 1966–1969. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Karupiah, G.; Xie, Q.W.; Buller, R.M.; Nathan, C.; Duarte, C.; MacMicking, J.D. Inhibition of Viral Replication by Interferon-Gamma-Induced Nitric Oxide Synthase. Science 1993, 261, 1445–1448. [Google Scholar] [CrossRef] [PubMed]
  115. Rimmelzwaan, G.F.; Baars, M.M.; de Lijster, P.; Fouchier, R.A.; Osterhaus, A.D. Inhibition of Influenza Virus Replication by Nitric Oxide. J. Virol. 1999, 73, 8880–8883. [Google Scholar] [CrossRef] [Green Version]
  116. Chen, L.; Liu, P.; Gao, H.; Sun, B.; Chao, D.; Wang, F.; Zhu, Y.; Hedenstierna, G.; Wang, C.G. Inhalation of Nitric Oxide in the Treatment of Severe Acute Respiratory Syndrome: A Rescue Trial in Beijing. Clin. Infect. Dis. Off. Publ. Infect. Dis. Soc. Am. 2004, 39, 1531–1535. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Angelidis, C.; Kotsialou, Z.; Kossyvakis, C.; Vrettou, A.-R.; Zacharoulis, A.; Kolokathis, F.; Kekeris, V.; Giannopoulos, G. Colchicine Pharmacokinetics and Mechanism of Action. Curr. Pharm. Des. 2018, 24, 659–663. [Google Scholar] [CrossRef] [PubMed]
  118. Ben-Chetrit, E.; Bergmann, S.; Sood, R. Mechanism of the Anti-Inflammatory Effect of Colchicine in Rheumatic Diseases: A Possible New Outlook through Microarray Analysis. Rheumatology 2006, 45, 274–282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Herscovici, T.; Merlob, P.; Stahl, B.; Laron-Kenet, T.; Klinger, G. Colchicine Use during Breastfeeding. Breastfeed. Med. Off. J. Acad. Breastfeed. Med. 2015, 10, 92–95. [Google Scholar] [CrossRef] [PubMed]
  120. Götestam Skorpen, C.; Hoeltzenbein, M.; Tincani, A.; Fischer-Betz, R.; Elefant, E.; Chambers, C.; da Silva, J.; Nelson-Piercy, C.; Cetin, I.; Costedoat-Chalumeau, N.; et al. The EULAR Points to Consider for Use of Antirheumatic Drugs before Pregnancy, and during Pregnancy and Lactation. Ann. Rheum. Dis. 2016, 75, 795–810. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  121. Indraratna, P.L.; Virk, S.; Gurram, D.; Day, R.O. Use of Colchicine in Pregnancy: A Systematic Review and Meta-Analysis. Rheumatol. Oxf. Engl. 2018, 57, 382–387. [Google Scholar] [CrossRef] [Green Version]
  122. Misra, D.P.; Gasparyan, A.Y.; Zimba, O. Benefits and Adverse Effects of Hydroxychloroquine, Methotrexate and Colchicine: Searching for Repurposable Drug Candidates. Rheumatol. Int. 2020, 40, 1741–1751. [Google Scholar] [CrossRef]
  123. Gloperba (Colchicine Oral Solution): Uses, Dosage, Side Effects, Interactions, Warning. Available online: https://www.rxlist.com/gloperba-drug.htm (accessed on 17 April 2021).
  124. Colchicine—FDA Prescribing Information, Side Effects and Uses. Available online: https://www.drugs.com/pro/colchicine.html (accessed on 17 April 2021).
  125. Mitigare Uses, Side Effects & Warnings. Available online: https://www.drugs.com/mtm/mitigare.html (accessed on 17 April 2021).
  126. Misawa, T.; Takahama, M.; Kozaki, T.; Lee, H.; Zou, J.; Saitoh, T.; Akira, S. Microtubule-Driven Spatial Arrangement of Mitochondria Promotes Activation of the NLRP3 Inflammasome. Nat. Immunol. 2013, 14, 454–460. [Google Scholar] [CrossRef]
  127. Marchetti, C.; Mould, K.; Tengesdal, I.W.; Janssen, W.J.; Dinarello, C.A. Targeting of the NLRP3 Inflammasome for Early COVID-19. bioRxiv 2021. [Google Scholar] [CrossRef]
  128. Tardif, J.-C.; Bouabdallaoui, N.; L’Allier, P.L.; Gaudet, D.; Shah, B.; Pillinger, M.H.; Lopez-Sendon, J.; da Luz, P.; Verret, L.; Audet, S.; et al. Efficacy of Colchicine in Non-Hospitalized Patients with COVID-19. medRxiv 2021. [Google Scholar] [CrossRef]
  129. Follmann, D.; Fintzi, J.; Fay, M.P.; Janes, H.E.; Baden, L.; Sahly, H.E.; Fleming, T.R.; Mehrotra, D.V.; Carpp, L.N.; Juraska, M.; et al. Assessing Durability of Vaccine Effect Following Blinded Crossover in COVID-19 Vaccine Efficacy Trials. medRxiv 2020. [Google Scholar] [CrossRef]
  130. Hung, I.; Lung, K.-C.; Tso, E.; Chung, T.; Chu, M.-Y.; Ng, Y.-Y.; Lo, J.; Chan, J.; Tam, A.; Shum, H.; et al. Triple Combination of Interferon Beta-1b, Lopinavir–Ritonavir, and Ribavirin in the Treatment of Patients Admitted to Hospital with COVID-19: An Open-Label, Randomised, Phase 2 Trial. Lancet 2020, 395. [Google Scholar] [CrossRef]
  131. Jeffreys, L.; Pennington, S.H.; Duggan, J.; Breen, A.; Jinks, J.; Ardrey, A.; Donnellan, S.; Patterson, E.I.; Hughes, G.L.; Hong, D.; et al. Remdesivir-Ivermectin Combination Displays Synergistic Interaction with Improved in Vitro Antiviral Activity against SARS-CoV-2. bioRxiv 2020. [Google Scholar] [CrossRef]
  132. Sahraei, Z.; Shabani, M.; Shokouhi, S.; Saffaei, A. Aminoquinolines against Coronavirus Disease 2019 (COVID-19): Chloroquine or Hydroxychloroquine. Int. J. Antimicrob. Agents 2020, 55, 105945. [Google Scholar] [CrossRef] [PubMed]
  133. Meo, S.A.; Zaidi, S.Z.A.; Shang, T.; Zhang, J.Y.; Al-Khlaiwi, T.; Bukhari, I.A.; Akram, J.; Klonoff, D.C. Biological, Molecular and Pharmacological Characteristics of Chloroquine, Hydroxychloroquine, Convalescent Plasma, and Remdesivir for COVID-19 Pandemic: A Comparative Analysis. J. King Saud Univ. Sci. 2020, 32, 3159–3166. [Google Scholar] [CrossRef] [PubMed]
  134. Sahebnasagh, A.; Saghafi, F.; Avan, R.; Khoshi, A.; Khataminia, M.; Safdari, M.; Habtemariam, S.; Ghaleno, H.R.; Nabavi, S.M. The Prophylaxis and Treatment Potential of Supplements for COVID-19. Eur. J. Pharmacol. 2020, 887, 173530. [Google Scholar] [CrossRef] [PubMed]
  135. Bloch, E.M.; Shoham, S.; Casadevall, A.; Sachais, B.S.; Shaz, B.; Winters, J.L.; van Buskirk, C.; Grossman, B.J.; Joyner, M.; Henderson, J.P.; et al. Deployment of Convalescent Plasma for the Prevention and Treatment of COVID-19. J. Clin. Invest. 2020, 130, 2757–2765. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Shanmugaraj, B.; Siriwattananon, K.; Wangkanont, K.; Phoolcharoen, W. Perspectives on Monoclonal Antibody Therapy as Potential Therapeutic Intervention for Coronavirus Disease-19 (COVID-19). Asian Pac. J. Allergy Immunol. 2020, 38, 10–18. [Google Scholar] [CrossRef]
  137. Netea, M.G.; Giamarellos-Bourboulis, E.J.; Domínguez-Andrés, J.; Curtis, N.; van Crevel, R.; van de Veerdonk, F.L.; Bonten, M. Trained Immunity: A Tool for Reducing Susceptibility to and the Severity of SARS-CoV-2 Infection. Cell 2020, 181, 969–977. [Google Scholar] [CrossRef]
  138. Kaufmann, S.H.; Cotton, M.F.; Eisele, B.; Gengenbacher, M.; Grode, L.; Hesseling, A.C.; Walzl, G. The BCG Replacement Vaccine VPM1002: From Drawing Board to Clinical Trial. Expert Rev. Vaccines 2014, 13, 619–630. [Google Scholar] [CrossRef] [PubMed]
  139. Rosero, S.; Torres, I.; Goodridge, A. Mast Cell C57 Activation by Mycobacterium Tuberculosis Lipids and Mycobacterium Bovis Bacille Calmette-Guerin (BCG). In C58. Tuberculosis Infection and Disease; American Thoracic Society International Conference Abstracts; American Thoracic Society: New York, NY, USA, 2016; p. A5484. [Google Scholar]
Table 1. Search outcomes summary.
Table 1. Search outcomes summary.
DrugAll Clinical TrialsProphylactic Clinical Trials
Favipiravir381
LPV/r384
Emtricitabine/Tenofovir73
Ivermectin6810
Interferons405
Nitazoxanide285
Bromhexine hydrochloride (HCl)62
Doxycycline141
Famotidine80
Nitric Oxide (NO)225
Colchicine260
Table 2. Candidate SARS-CoV-2 prophylactic drugs currently in clinical trials.
Table 2. Candidate SARS-CoV-2 prophylactic drugs currently in clinical trials.
DrugSafetyCost 2Long-Term TreatmentCOVID-19 Clinical TrialAdministration 3
(Price per Dose in USD,
Single Dose)
Age +65Prophylaxis
AntiviralFavipiravir++++N.A. 425 days (COVID-19)++per os (P.O.)
LPV/r+++3.9–4.6
(400–100 mg/5 mL)
Unlimited +P.O.
Emtricitabine/
tenofovir
+++36.02–49.32 (300 mg)Unlimited (HIV)
12 weeks (COVID-19)
+P.O.
Repurposed DrugsIvermectin+++3.95 (3 mg)15–17 year
(up to ×2 a year)
+P.O./intravenous (I.V.)
Interferons++++9274 (4 mL) 5
6985 (0.3 mg) 6
28 days (COVID-19) +Mucosal, Parenteral
Nitazoxanide++++140.8 (500 mg) 73–24 months++P.O.
Bromhexine HCl++++0.1–0.6 (8 mg)2 months (COVID-19) +P.O.
MiscellaneousDoxycycline+++0.6–2 (50/100 mg)Months P.O./I.V.
Famotidine++++0.08–2.04 (20 mg)Unlimited +/−P.O./I.V.
Nitric Oxide (NO)++++ 1N.A.5–17 months
4 weeks (COVID-19)
+Inhalation, Topical 3
Colchicine+++2.24–2.55 (0.6 mg)Unlimited
21 days (COVID-19)
+ P.O.
1 NO release solutions and nasal drops are associated with high safety margins; 2 According to online price evaluation at drugs.com [11]; 3 Nasal spray/wash/gargling; 4 Not available; 5 IFN-β-1a; 6 IFN-β-1b; 7 Cheaper generic forms are available. “++++” is considered to have a highly favorable safety profile; “+++” is considered to have a favorable safety profile.
Table 3. Clinical trials status of candidate for SARS-CoV-2 prophylactic drugs *.
Table 3. Clinical trials status of candidate for SARS-CoV-2 prophylactic drugs *.
DrugNCTPhaseParticipantsCountryRemarks
IvermectinNCT04832945Completed713Dominican Republic
NCT04668469Completed600Egypt
NCT04425850Completed229ArgentinaPositive results
NCT0444610434257Singapore
NCT048912504800ZambiaNot yet recruiting
NCT045272113550ColombiaNot yet recruiting
NCT048947212/3750ArgentinaRecruiting
NCT044225612/3340Egypt
NCT047017101/2300Argentina+Iota-carrageenan
NCT04384458N.A.400BrazilRecruiting
NitazoxanideNCT047884074456ArgentinaRecruiting
NCT0435968031407USA
NCT04343248 13800USA
NCT0456106321950South AfricaRecruiting
NCT044353142200BrazilNot yet recruiting
Emtricitabine/TenofovirNCT0433492834000SpainRecruiting
NCT0440527131378ArgentinaNot yet recruiting
NCT045191252/3950ColombiaNot yet recruiting
LPV/rNCT0432828531200France
NCT043640223326Switzerland
NCT0432117431220CanadaRecruiting
NCT04251871N.A.150ChinaRecruiting 6
InterferonsNCT0453472532282Australia 5Recruiting
NCT0432023832944ChinaRecruiting
NCT0455237931240ChileRecruiting
NCT043446002164USARecruiting
NCT04251871N.A.150ChinaRecruiting 6
DoxycyclineNCT04584567 331100TunisiaRecruiting
Nitric Oxide (NO)NCT048423312/3600UKRecruiting
NCT044081832225USARecruiting
NCT043379182143Canada
NCT04858451 22150UKNot yet recruiting
NCT04312243224USA
FavipiravirNCT044481192760Canada
Bromhexine HClNCT04405999Completed50Russia
NCT04340349 41214Mexico
* Drugs were ordered according to available data volume and clinical trial status; 1 Post-exposure prophylaxis of COVID-19 and other viral respiratory illnesses in elderly residents of Long-Term Care Facilities (LTCF); 2 In COPD and bronchiectasis patients; 3 Combined with zinc; 4 Combined with hydroxychloroquine; 5 In cancer patients; 6 Not the main subject of the study. It is a part of the standard of care treatment.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ben-Zuk, N.; Dechtman, I.-D.; Henn, I.; Weiss, L.; Afriat, A.; Krasner, E.; Gal, Y. Potential Prophylactic Treatments for COVID-19. Viruses 2021, 13, 1292. https://doi.org/10.3390/v13071292

AMA Style

Ben-Zuk N, Dechtman I-D, Henn I, Weiss L, Afriat A, Krasner E, Gal Y. Potential Prophylactic Treatments for COVID-19. Viruses. 2021; 13(7):1292. https://doi.org/10.3390/v13071292

Chicago/Turabian Style

Ben-Zuk, Noam, Ido-David Dechtman, Itai Henn, Libby Weiss, Amichay Afriat, Esther Krasner, and Yoav Gal. 2021. "Potential Prophylactic Treatments for COVID-19" Viruses 13, no. 7: 1292. https://doi.org/10.3390/v13071292

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