Next Article in Journal
Interplay between Cellular Autophagy and Hepatitis B Virus Replication: A Systematic Review
Previous Article in Journal
Increased Uptake of Silica Nanoparticles in Inflamed Macrophages but Not upon Co-Exposure to Micron-Sized Particles
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Ivermectin as a Broad-Spectrum Host-Directed Antiviral: The Real Deal?

1
Nuclear Signaling Lab., Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Monash, Victoria 3800, Australia
2
Cancer Targeting and Nuclear Therapeutics Lab., Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Monash, Victoria 3800, Australia
*
Author to whom correspondence should be addressed.
Cells 2020, 9(9), 2100; https://doi.org/10.3390/cells9092100
Submission received: 8 August 2020 / Revised: 4 September 2020 / Accepted: 9 September 2020 / Published: 15 September 2020
(This article belongs to the Section Cell Nuclei: Function, Transport and Receptors)

Abstract

:
The small molecule macrocyclic lactone ivermectin, approved by the US Food and Drug Administration for parasitic infections, has received renewed attention in the last eight years due to its apparent exciting potential as an antiviral. It was identified in a high-throughput chemical screen as inhibiting recognition of the nuclear localizing Human Immunodeficiency Virus-1 (HIV-1) integrase protein by the host heterodimeric importin (IMP) α/β1 complex, and has since been shown to bind directly to IMPα to induce conformational changes that prevent its normal function in mediating nuclear import of key viral and host proteins. Excitingly, cell culture experiments show robust antiviral action towards HIV-1, dengue virus (DENV), Zika virus, West Nile virus, Venezuelan equine encephalitis virus, Chikungunya virus, Pseudorabies virus, adenovirus, and SARS-CoV-2 (COVID-19). Phase III human clinical trials have been completed for DENV, with >50 trials currently in progress worldwide for SARS-CoV-2. This mini-review discusses the case for ivermectin as a host-directed broad-spectrum antiviral agent for a range of viruses, including SARS-CoV-2.

1. Introduction

The 2015 Nobel Prize for medicine recognizes the seminal contribution of Campbell and Ōmura in terms of the “wonder drug” ivermectin, a macrocyclic lactone 22,23-dihydroavermectin B produced by the bacterium Streptomyces avermitilis [1], as a novel therapeutic against “infections caused by roundworm parasites”; this was alongside Tu Youyou for her seminal work on artemisinin and malaria [2]. Discovered in 1975, ivermectin was marketed successfully from 1981 for parasitic infection indications in animals, and then approved for human use for activity against onchocerciasis (river blindness) in 1987. It has since been used successfully to treat a number of human parasitic worm infestations causing river blindness/filariasis, strongyloidiasis/ascariasis, ectoparasites causing scabies, pediculosis and rosacea [1,3]. More recent applications include to control insect mediators of infection, such as malaria [1,3,4,5]. Ivermectin is on the World Health Organization’s Model List of Essential Medicines [6].
From 2012 onwards, there have multiple reports that ivermectin has antiviral properties [4,5,7,8,9,10,11,12,13,14,15,16,17] towards a growing number of RNA viruses, including human immunodeficiency virus (HIV)-1, influenza, flaviruses such as dengue virus (DENV) and Zika virus (ZIKV) and, most notably, SARS-CoV-2 (COVID-19) [17]. Evidence for activity against DNA viruses is more limited, but encompasses Pseudorabies, polyoma and adenoviruses [18,19,20]. The basis of ivermectin’s broadspectrum antiviral activity appears to relate to the fact that ivermectin binds to, and inhibits, the nuclear transport role of the host importin α (IMPα) protein [11,18,20], which is known to mediate nuclear import of various viral proteins and key host factors, but other possible antiviral actions of ivermectin have been proposed (e.g., [12]), including in the case of SARS-CoV-2 (e.g., [21,22]). This mini-review will summarise the weight of evidence for ivermectin’s broad-spectrum antiviral activity and the basis of its IMPα-directed activity in light of the possibility that ivermectin could be a critically useful antiviral in the current SARS-CoV-2 crisis [6,17].

2. Ivermectin as a US Food and Drug Administration-Approved Anti-Parasitic Agent

It is difficult to overestimate the impact of ivermectin as a therapeutic agent to control various parasitic diseases [1,2,3,4,5,6]. It is administered as a single oral yearly dose (e.g., 150 or 200 μg/kg, respectively) to treat onchocerciasis and strongyloidiasis. Lymphatic filariasis is similarly treated in endemic areas with a once-yearly dose (300–400 μg/kg), or alternatively bi-yearly dosing (150–200 μg/kg) [23]. Ivermectin’s documented antiparasitic mode of action is through potentiating GABA-mediated neurotransmission, and by binding to invertebrate glutamate-gated Cl channels to effect parasite paralysis and death [24]. Selectivity comes from the fact that ivermectin does not readily penetrate the central nervous system (CNS) of mammals, where GABA functions as a neurotransmitter [24].
Doses up to 2000 µg/kg are well tolerated in patients with parasitic infections [23,25], with analysis of the first 11 years of mass global ivermectin (Mectizan) administration indicating a cumulative incidence of one serious adverse side effect case per million [4,26]. Similarly, although drug resistance can occur in animals, no resistance in humans has yet been confirmed in over 25 years. Based on this weight of evidence, ivermectin is unquestionably a safe, potent antiparasitic agent likely to be used as such long into the future [1,4].

3. Ivermectin as an IMPα Targeting Agent with Antiviral Activity

Transport into and out of the nucleus is central to eukaryotic cell and tissue function, with a key role to play in viral infection, where a common strategy used by viruses is to antagonize the cellular antiviral response [14,27]. The targeting signal-dependent mediators of this transport are the members of the IMP superfamily of proteins, of which there are multiple α and β forms [14,27]. The pathway mediated by the IMPα/β1 heterodimer is the best characterized pathway by which host proteins, including members of the signal transducers and activators of transcription (STATs) and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) transcription factor families, enter the nucleus through nuclear envelope-embedded nuclear pores. A large number of viral proteins (e.g., [27,28]) also use this pathway (see Figure 1), where IMPα within the IMPα/β1 heterodimer performs the adaptor role of specific targeting signal recognition, while IMPβ1 performs the main nuclear roles of binding to/translocation through the nuclear pores, and release of the nuclear import cargo within the nucleus (Figure 1) [27].
The importance of nuclear targeting of viral proteins to the nucleus in the infectious cycle has been demonstrated for a number of viruses. Mutagenic analyses, for example, show that specific recognition by IMPα is critical to nuclear localization of various viral proteins, such as DENV non-structural protein (NS) 5 [30]; significantly, DENV, which shows the same reduced interaction of NS5 with IMPα is severely attenuated, underlining the importance of the NS5-IMPα interaction for dengue infection. As has since been shown using a range of different small molecules, the critical importance of this interaction to dengue infection is the basis for the fact that multiple distinct small molecules that disrupt IMPα recognition of dengue NS5 are able to limit dengue infection [7,8,11,29,31]. In the case of ivermectin, this activity extends to a large number of different viruses (see below) [7,8,9,10,11,12,13,14,15,16,17], including SARS-CoV-2 [18]. Which SARS-CoV-2 proteins may access the nucleus in infected cells has not been examined in detail but, in terms of related coronaviruses, ORF6 (Open Reading Frame 6) protein from SARS-CoV-1 has been shown to bind IMPα [32], and ORF4b from MERS-CoV (Middle Eastern Respiratory Syndrome Coronavirus) is known to access the nucleus in NLS-dependent fashion [33]. Ongoing research will establish which of the SARS-CoV-2 ORFs may play comparable roles, and be potential targets of the impact of ivermectin on IMPα.
We identified ivermectin in 2011 in a proof-of-principle, high-throughput screen using recombinantly-expressed proteins and a 1200 compound library for inhibitors of HIV-1 Integrase (IN) recognition by IMPα/β1 [34]; specific inhibitors targeting IMPα/β1 directly (such as ivermectin) and not IN (such as budenoside) were identified using a nested counterscreen strategy [34,35]. Of several compounds subsequently confirmed to be active against IMPα/β1 and possess antiviral activity as a consequence [7,14,29,36], ivermectin has been the best characterized in this regard, and shown to have broad-spectrum activities, a number of which are summarized in Table 1. It was initially shown to inhibit nuclear import not only of IN, but also of simian virus SV40 large tumour antigen (T-ag) and other IMPα/β1-dependent (but not IMPβ1-dependent) cargoes, consistent with the idea that IMPα (not IN) is the direct target [34,35]. Subsequent work has confirmed this, with ivermectin’s ability to inhibit the nuclear accumulation of various different host, including NF-kB p65 [37] and viral proteins demonstrated in transfected and infected cell systems (see Table 1) [14,34]. Ivermectin’s ability to inhibit binding of IMPα to the viral proteins NS5 and T-ag has also been confirmed in a cellular context using the biomolecular fluorescence complementation technique [11].
Although targeting of IMPα by ivermectin was clearly supported by many years of research (see also below), direct binding to IMPα was only recently formally demonstrated using a set of biophysical techniques, including thermostability, analytical ultracentrifugation, and circular dichroism (CD) [11]. Importantly, the CD/thermostability studies indicate that binding of ivermectin by IMPα induces a structural change, which is likely the basis of IMPα’s inability to bind viral nuclear import cargoes. Strikingly, the structural change also appears to impair heterodimerisation of IMPα with IMPβ1 [11]; IMPα alone cannot mediate nuclear import, only within the heterodimer with IMPβ1. Thus, ivermectin inhibits nuclear import not only by preventing signal recognition by IMPα, but also by ensuring that the IMPα/β1 complex essential to mediate subsequent transport through the nuclear pore is prevented from forming. Interestingly, GW5074 (see Table 1) appears to have a close to identical mechanism to that for ivermectin in binding to IMPα to induce structural changes that prevent both cargo recognition and specific interaction with IMPβ1 [29]; there is no such data for GSP (see Table 1), which is believed to impact the NLS-binding site of IMPα and prevent cargo recognition more directly (manuscript in preparation).

4. Ivermectin as an Antiviral

Consistent with the fact that many viruses are known to rely on IMPα/β1-dependent nuclear import of specific viral proteins for robust infection [14,27,28], ivermectin has been confirmed in a body of in vitro studies to be active in limiting infection by a range of different RNA viruses [10,14], including HIV-1 [7], DENV (all four serotypes) and related flaviviruses [8,11,12], influenza, and alphaviruses such as Venezuelan equine encephalitis virus (VEEV) and chikungunya [9,15,16] (see Table 1); it is also active against DNA viruses [18,19,20]. Recent studies indicate it is a potent inhibitor of SARS-CoV-2 [17].
A striking aspect of this antiviral activity is that, where determined, the EC50 for viral inhibition as assessed by a range of different techniques is in the low μM range (see right column, Table 1), interestingly aligning perfectly with its activity in inhibiting recognition of viral nuclear import cargoes by IMPα (see top of left column, Table 1). The clear implication is that the mechanism of inhibition of infectious virus production in the case of all of the viruses listed in Table 1 is largely through targeting IMPα to prevent its role in nuclear import, and of viral proteins in particular (see Figure 1). Significantly, two other small molecules (GW5074 and gossypol) that appear to target IMPα in a very similar way to prevent its nuclear import function [29] have comparable antiviral properties [13,29,36], consistent with the idea that the host protein IMPα is a key contributor to infection by a number of medically important viruses.

5. Ivermectin as an Antiviral in the Clinic

One of the greatest challenges in antiviral research, as in many other disciplines, is to transition from laboratory experiments to preclinical/clinical studies, with the question of dosing challenging [6]. However, it is important to stress the obvious in this context: that the antiviral activities of ivermectin documented in Table 1 have been derived from laboratory experiments that largely involve high, generally non-physiological, multiplicities of infection, and cell monolayer cultures, often of cell lines such as Vero cells (African green monkey kidney, impaired in interferon α/β production) that are not clinically relevant. Clearly, the results in Table 1 for low μM EC50 values should not be interpreted beyond the fact that they reveal robust, dose-dependent antiviral activity in the cell model system used, and it would be naïve to strive for μM concentrations of ivermectin in the clinic based on them.
A key consideration in any clinical intervention using ivermectin is its host-directed (IMPα-directed) mechanism of action. Host-directed agents that impact cellular activities that are essential to healthy function must be tested with caution; although ivermectin has an established safety profile in humans [23,25], and is US Food and Drug Administration-approved for a number of parasitic infections [1,3,5], it targets a host function that is unquestionably important in the antiviral response, and titration of a large proportion of the IMPα repertoire of a cell/tissue/organ likely to lead to toxicity. With this in mind, where a host-directed agent can be a “game-changer” in treating viral infection may well be in the initial stages of infection or even prophylactically (see Section 6) to keep the viral load low so that the body’s immune system has an opportunity to mount a full antiviral response [11,17].
Ivermectin’s real potential as an antiviral to treat infection can, of course, only be demonstrated in preclinical/clinical studies. Preclinical studies include a lethal Pseudorabies (PRV) mouse challenge model which showed that dosing (0.2 mg/kg) 12 h post-infection protected 50% of mice, which could be increased to 60% by administering ivermectin at the time of infection [18]. Apart from the many clinical trials currently running for SARS-CoV-2 (see below), the only other study thus far reported relates to a phase III trial for DENV infection [40]. Almost 70% of the world’s population in over 120 countries is currently threatened by mosquito-borne flaviviral infections, with an estimated 100 million symptomatic DENV infections and up to 25,000 deaths each year from dengue haemorrhagic fever [41,42], despite sophisticated large-scale vector control programs. As for the closely related ZIKV (cause of large outbreaks in the Americas in 2015/2016), the dearth of antiviral treatments and challenges in developing efficacious vaccines hamper disease control. Clinical data published in preliminary form for the phase III trial in Thailand [40] indicate antiviral activity; daily dosing (0.4 mg/kg) was concluded to be safe, and have virological efficacy, but clear clinical benefit was not reported, potentially due to the timing of the intervention. The authors concluded that dosing regimen modification was required to ensure clinical benefit [40]. This study both underlines ivermectin’s potential to reduce viral load in a clinical context, and highlights the complexities of timely intervention and effective dosing regimens to achieve real clinical benefit in the field.

6. A Viable Treatment for SARS-CoV-2?

Despite efforts in multiple domains, the current SARS-CoV-2 pandemic has now eclipsed the porcine flu epidemic in terms of numbers of infections (rapidly nearing 30 million) and deaths (>930,000) worldwide. The search for antivirals for SARS-CoV-2 through repurposing existing drugs has proved challenging (e.g., see [43,44,45,46,47]), one important aspect of repurposing being the perceived need to achieve therapeutic levels in the lung. Published pharmacokinetic modelling based on both the levels of ivermectin achievable in human serum from standard parasitic treatment dosing and robust large animal experiments where lung levels of ivermectin can be measured, indicates that concentrations of ivermectin 10 times higher than the c. 2.5 μM EC50 indicated by in vitro experiments (Table 1) are likely achievable in the lung in the case of SARS-CoV-2 [48]; modelling based on different assumptions predicts lower values, but stresses the long-term stability of ivermectin in the lung (for over 30 days) based on data from animals [49]. It should also be noted that liquid formulations for intravenous administration of long-acting ivermectin have been described, with aerosol administration also in development, to enable ivermectin administration to achieve even higher concentrations to tackle SARS-CoV-2, whilst the use of ivermectin in combination with other agents may enhance efficacy at lower doses.
There are currently more than 50 trials worldwide testing the clinical benefit of ivermectin to treat or prevent SARS-CoV-2 (see Table 2).
These include variations on combination therapies (see [51,52]), dosing regimens, and prophylactic protocols. With respect to the latter, preliminary results from recently completed study NCT04422561 (Table 2, no. 22), that examines asymptomatic family close contacts of confirmed COVID patients, show that two doses of ivermectin 72 h apart result in only 7.4% of 203 subjects reporting symptoms of SARS-CoV-2 infection, in stark contrast to control untreated subjects, of whom 58.4% reported symptoms, underlining ivermectin’s potential as a prophylactic. It is to be hoped that the results from the rigorous randomised clinical trials listed in Table 2 will emerge in the next few months to document ivermectin’s credentials as “the real deal” for COVID-19 infection or otherwise. In this context, it is noteworthy that ivermectin has already been approved for the treatment of SARS-CoV-2 in humans by the Republic of Peru [53] and in the Northeastern Beni region of Bolivia [54].

7. Conclusions

An instinctive response in developing antiviral agents is to strive for high specificity, making the idea of virus-targeted agents specific to a particular viral component or function attractive, since, ideally, they circumvent the possibility of impacting host function. However, the high propensity of viral genomes, and particularly those of RNA viruses, to mutate and evolve means that selection for resistance can be all too prevalent (e.g., for HIV). Importantly, the high specificity of an agent to a particular virus also inevitably means that its utility against a distinct virus may be limited or non-existent. Thus, it is not surprising that repurposed antivirals active against influenza or HIV, for example, may prove in efficacious against distantly related flaviviruses or coronaviruses.
In contrast, antivirals that are host-directed can be repurposed more easily, as long as the viruses in question rely on the same host pathway/functions for robust infection, simply because the host pathway/function targeted is the same [14,28]. Although potential complication here is the viral tissue tropism (e.g., blood or lung in the case of systemic or respiratory viral infections) and accompanying pharmacokinetic considerations, selection for viral resistance is largely circumvented in this scenario. As long as toxicity is not an issue, host-directed agents thus have the potential to be genuinely broad-spectrum agents against various different viruses that rely on a common host pathway. The fact that so many viruses rely on IMPα/β1-dependent nuclear import for robust infection ([14,27,28] and see above) means that agents targeting this pathway have true potential to be broad-spectrum antivirals. After decades of use in the field, ivermectin clearly “fits the bill” here in terms of human safety, but whether it turns out to be the molecule that proves this principle will only begin to be established unequivocally, one way or another, in the ensuing months with respect to SARS-CoV-2.

Author Contributions

Conceptualization: D.A.J.; writing—original draft preparation: D.A.J., K.M.W.; writing—review and editing: D.A.J., K.M.W. All authors have read and agreed to the published version of the manuscript.

Funding

The authors acknowledge the financial support of the National Health and Medical Research Council Australia (Senior Principal Research Fellowship APP1002486/APP1103050) to D.A.J.

Acknowledgments

Andrew Owen (University of Liverpool, United Kingdom), Anudeep Talagavadi Channaiah (Topiwala National Medical College and B.Y.L. Nair Charitable Hospital, India), and Joe Mymryk (University of Western Ontario, Ontario) are thanked for sharing recent publications.

Conflicts of Interest

We confirm that there are no conflict of interest associated with this publication, with no significant financial support for this work that could have influenced its outcome.

Abbreviations

HIV-1, human immunodeficiency virus; VLP, virus like particle; PSV, Pseudorabies virus; T-ag, SV40 large T-antigen; VSV-G, vesicular stomatitis virus glycoprotein; YFV (yellow fever virus); WNV, West Nile virus; DENV, dengue virus; ZIKV, Zika virus; VEEV, Venezuelan equine encephalitis virus; PCR, polymerase chain reaction (viral genomes); CPE, cytopathogenic effect (host cell); pfu, plaque forming units (infectious virus); CFI, cell fluorescence assay (immunostaining for virus); TCID, tissue culture infectious dose (estimation of viral load based on CPE).

References

  1. Crump, A.; Omura, S. Ivermectin, wonder drug from Japan: The human use perspective. Proc. Jpn. Acad. 2011, 87, 13–28. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Nobel Foundation. The Nobel Prize in Medicine or Physiology. 2015. Available online: https://www.nobelprize.org/prizes/medicine/2015/press-release (accessed on 11 September 2020).
  3. González Canga, A.; Sahagún Prieto, A.M.; Diez Liébana, M.J.; Fernández Martínez, N.; Sierra Vega, M.; García Vieitez, J.J. The pharmacokinetics and interactions of ivermectin in humans—A mini-review. AAPSJ 2008, 10, 42–46. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Crump, A.; Omura, S. Ivermectin: Panacea for resource-poor communities? Trends Parasitol. 2014, 30, 445–455. [Google Scholar]
  5. World Health Organization. World Health Organization’s List of Essential Medicines 21st List 2019. Available online: https://apps.who.int/iris/bitstream/handle/10665/325771/WHO-MVP-EMP-IAU-2019.06-eng.pdf (accessed on 11 September 2020).
  6. Kumar, B.S.; Jeyaraman, M.; Jain, R.; Anudeep, T.C. A Wonder Drug in the Arsenal against COVID—19: Medication Evidence from Ivermectin. J. Adv. Med. Med. Res. 2020, 32, 30–37. [Google Scholar] [CrossRef]
  7. Wagstaff, K.M.; Sivakumaran, H.; Heaton, S.; 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, 851–856. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Tay, M.Y.; Fraser, J.E.; Chan, W.K.K.; Moreland, N.J.; Rathore, A.P.; Wang, C.; Vasudevan, S.G.; Jans, D.A. Nuclear localization of dengue virus (DENV) 1-4 non-structural protein 5; protection against all 4 DENV serotypes by the inhibitor Ivermectin. Antivir. Res. 2013, 99, 301–306. [Google Scholar] [CrossRef] [PubMed]
  9. Lundberg, L.; Pinkhan, C.; Baer, A.; Amaya, M.; Narayan, A.; Wagstaff, K.M.; Jans, D.A.; Kehn-Hall, K. Nuclear import and export inhibitors alter capsid protein distribution in mammalian cells and reduce Venezuelan Equine Encephalitis Virus replication. Antivir. Res. 2013, 100, 662–672. [Google Scholar] [CrossRef]
  10. Gotz, V.; Magar, L.; Dornfeld, D.; Giese, S.; Pohlmann, A.; Höper, D.; Kong, B.-W.; Jans, D.A.; Beer, M.; Haller, O.; et al. Influenza A viruses escape from MxA restriction at the expense of efficient nuclear vRNP import. Sci. Rep. 2016, 6, 23138. [Google Scholar] [CrossRef] [Green Version]
  11. Yang, S.N.Y.; Atkinson, S.C.; Wang, C.; Lee, A.; Bogoyevitch, M.A.; Borg, N.A.; Jans, D.A. The broad spectrum antiviral ivermectin targets the host nuclear transport importin α/β1 heterodimer. Antivir. Res. 2020, 177, 104760. [Google Scholar] [CrossRef]
  12. Mastrangelo, E.; Pezzullo, M.; De Burghgraeve, T.; Kaptein, S.; Pastorino, B.; Dallmeier, K.; de Lamballerie, X.; Neyts, J.; Hanson, A.M.; Frick, D.N.; et al. Lvermectin is a potent inhibitor of flavivirus replication specifically targeting NS3 helicase activity: New prospects for an old drug. J. Antimicrob. Chemother. 2012, 67, 1884–1894. [Google Scholar] [CrossRef] [Green Version]
  13. Atkinson, S.; Audsley, M.; Lieu, K.; Marsh, G.; Thomas, D.R.; Heaton, S.; Paxman, J.; Wagstaff, K.M.; Buckle, A.G.; Moseley, G.; et al. Recognition by host nuclear transport proteins drives disorder-to-order transition in Hendra virus V. Sci. Rep. 2018, 8, 23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Jans, D.A.; Martin, A.J.; Wagstaff, K.M. Inhibitors of nuclear transport. Curr. Opin. Cell Biol. 2019, 58, 50–60. [Google Scholar] [CrossRef] [PubMed]
  15. Varghese, F.S.; Kaukinen, P.; Glaesker, 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. Antivir. Res. 2016, 126, 117–124. [Google Scholar] [CrossRef] [PubMed]
  16. Thomas, D.R.; Lundberg, L.; Pinkhan, C.; Shechter, S.; Debono, A.; Baell, J.; Wagstaff, K.M.; Hick, C.A.; Kehn-Hall, K.; Jans, D.A. Identification of novel antivirals inhibiting recognition of Venezuelan equine encephalitis virus capsid protein by the Importin α/β1 heterodimer through high-throughput screening. Antivir. Res. 2018, 151, 8–19. [Google Scholar] [CrossRef] [PubMed]
  17. 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, 104787. [Google Scholar] [CrossRef] [PubMed]
  18. Lv, C.; Liu, W.; Wang, B.; Dang, R.; Qiu, L.; Ren, J.; Yan, C.; Yang, Z.; Wang, X. Ivermectin inhibits DNA polymerase UL42 of Pseudorabies virus entrance into the nucleus and proliferation of the virus in vitro and vivo. Antivir. Res. 2018, 159, 55–62. [Google Scholar] [CrossRef]
  19. Bennett, S.M.; Zhao, L.; Bosard, C.; Imperiale, M.J. Role of a nuclear localization signal on the minor capsid proteins VP2 and VP3 in BKPyV nuclear entry. Virology 2015, 474, 110–116. [Google Scholar] [CrossRef] [Green Version]
  20. King, C.R.; Tessier, T.M.; Dodge, M.J.; Weinberg, J.B.; Mymryk, J.S. Lnhibition of human adenovirus replication by the importin α/β1 nuclear import inhibitor ivermectin. J. Virol. 2020, in press. [Google Scholar] [CrossRef]
  21. Changeux, J.-P.; Amoura, Z.; Rey, F.A.; Miyara, M. A nicotinic hypothesis for Covid-19 with preventive and therapeutic implications. Comptes Rendus Biol. 2020, 343, 33–39. [Google Scholar] [CrossRef]
  22. Krause, R.M.; Buisson, B.; Bertrand, S.; Corringer, P.J.; Galzi, J.L.; Changeux., J.P.; Bertrand, D. Lvermectin: A positive allosteric effector of the alpha7 neuronal nicotinic acetylcholine receptor. Mol. Pharmacol. 1998, 53, 283–294. [Google Scholar] [CrossRef] [Green Version]
  23. Guzzo, C.A.; Furtek, C.I.; Porras, A.G.; Chen, C.; Tipping, R.; Clineschmidt, C.M.; Sciberras, D.G.; Hsieh, J.Y.K.; Lasseter, K.C. Safety, tolerability, and pharmacokinetics of escalating high doses of ivermectin in healthy adult subjects. J. Clin. Pharmacol. 2002, 42, 1122–1133. [Google Scholar] [CrossRef] [PubMed]
  24. Loukas, A.; Hotez, P.J. Chemotherapy of helminth infections. In Goodman & Gilman’s The Pharmacological Basis of Therapeutics, 11th ed.; Brunton, L.L., Lazo, J.S., Parker, K.L., Eds.; McGraw Hill: New York, NY, USA, 2006; pp. 1073–1093. [Google Scholar]
  25. Navarro, M.; Camprubí, D.; Requena-Méndez, A.; Buonfrate, D.; Giorli, G.; Kamgno, J.; Gardon, J.; Muñoz, J.; Krolewiecki, A. Safety of high-dose ivermectin: A systematic review and meta-analysis. J. Antimicrob. Chemother. 2020, 75, 827–834. [Google Scholar] [CrossRef] [PubMed]
  26. Twum-Danso, N.A.Y.; Meredith, S.E.O. Variation in incidence of serious adverse events after onchocerciasis treatment with ivermectin in areas of Cameroon co-endemic for loiasis. Trop. Med. Int. Health 2003, 8, 820–831. [Google Scholar] [CrossRef]
  27. Fulcher, A.; Jans, D.A. Regulation of nucleocytoplasmic trafficking of viral proteins; an integral role in pathogenesis ? Biochem. Biophys. Acta Mol. Cell Res. 2011, 1813, 2176–2190. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Caly, L.; Wagstaff, K.M.; Jans, D.A. Nuclear trafficking of proteins from RNA viruses: Potential target for anti-virals? Antivir. Res. 2012, 95, 202–206. [Google Scholar] [CrossRef]
  29. Yang, S.N.Y.; Atkinson, S.C.; Fraser, J.E.; Wang, C.; Maher, B.; Roman, N.; Forwood, J.K.; Wagstaff, K.M.; Borg, N.A.; Jans, D.A. Novel Flavivirus Antiviral That Targets The Host Nuclear Transport Importin α/β1 Heterodimer. Cells 2019, 8, 281. [Google Scholar] [CrossRef] [Green Version]
  30. Pryor, M.J.; Rawlinson, S.M.; Butcher, R.E.; Barton, C.L.; Waterhouse, T.A.; Vasudevan, S.G.; Bardin, P.G.; Wright, P.J.; Jans, D.A.; Davidson, A.D. Nuclear localization of dengue virus nonstructural protein 5 through its importin alpha/beta-recognized nuclear localization sequences is integral to viral infection. Traffic 2007, 8, 795–807. [Google Scholar] [CrossRef] [PubMed]
  31. Fraser, J.E.; Watanabe, S.; Wang, C.; Chan, W.K.; Maher, B.; Lopez-Denman, A.; Hick, C.; Wagstaff, K.M.; Mackenzie, J.M.; Sexton, P.M.; et al. A nuclear transport inhibitor that modulates the unfolded protein response and provides in vivo protection against lethal dengue virus infection. J. Infect. Dis. 2014, 210, 1780–1791. [Google Scholar] [CrossRef] [Green Version]
  32. Frieman, M.; Yount, B.; Heise, M.; Kopecky-Bromberg, S.A.; Palese, P.; Baric, R.S. Severe Acute Respiratory Syndrome Coronavirus ORF6 Antagonizes STAT1 Function by Sequestering Nuclear Import Factors on the Rough Endoplasmic Reticulum/Golgi Membrane. J. Virol. 2007, 81, 9812–9824. [Google Scholar] [CrossRef] [Green Version]
  33. Yang, Y.; Ye1, F.; Zhu, N.; Wang, W.; Deng, Y.; Zhao, Z.; Tan, W. Middle East respiratory syndrome coronavirus ORF4b protein inhibits type I interferon production through both cytoplasmic and nuclear targets. Sci. Rep. 2015, 5, 17554. [Google Scholar] [CrossRef] [Green Version]
  34. Wagstaff, K.M.; Rawlinson, S.M.; Hearps, A.C.; Jans, D.A. An AlphaScreen(R)-based assay for high-throughput screening for specific inhibitors of nuclear import. J. Biomol. Screen. 2011, 16, 192–200. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Wagstaff, K.M.; Headey, S.; Telwatte, S.; Tyssen, D.; Hearps, A.C.; Thomas, D.R.; Tachedjian, G.; Jans, D.A. Molecular dissection of an inhibitor targeting the HIV integrase dependent preintegration complex nuclear import. Cell. Microbiol. 2018, e12953. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Lopez-Denman, A.J.; Russo, A.; Wagstaff, K.M.; White, P.A.; Jans, D.A.; Mackenzie, J.M. Nucleocytoplasmic shuttling of the West Nile virus RNA-dependent RNA polymerase NS5 is critical to infection. Cell. Microbiol. 2018, 20, e12848. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Ci, X.; Li, H.; Yu, Q.; Zhang, X.; Yu, L.; Chen, N.; Song, Y.; Deng, X. Avermectin exerts anti-inflammatory effect by downregulating the nuclear transcription factor kappa-B and mitogen-activated protein kinase activation pathway. Fundam. Clin. Pharmacol. 2009, 23, 449–455. [Google Scholar] [CrossRef]
  38. Yu, Z.-H.; Chan, H.C. Gossypol as a male antifertility agent—Why studies should have been continued. Int. J. Androl. 1998, 21, 2–7. [Google Scholar] [CrossRef] [PubMed]
  39. Porat, O. Effects of Gossypol on the Motility of Mammalian Sperm. Mol. Reprod. Dev. 1990, 21, 400–408. [Google Scholar] [CrossRef]
  40. Yamasmith, E.; Saleh-arong, F.A.; Avirutnan, P.; Angkasekwinai, N.; Mairiang, D.; Wongsawat, E.; Tanrumluk, S.; Fongsri, U.; Suputtamongkol, Y. Efficacy and Safety of Ivermectin against Dengue Infection: A Phase III, Randomized, Double-blind, Placebo-controlled Trial. In Proceedings of the 34th Annual Meeting The Royal College of Physicians of Thailand—‘Internal Medicine and One Health’, Pattaya, Thailand, 26–28 April 2018. [Google Scholar]
  41. Bhatt, S.; Gething, P.W.; Brady, O.J.; Messina, J.P.; Farlow, A.W.; Moyes, C.L.; Drake, J.M.; Brownstein, J.S.; Hoen, A.G.; Sankoh, O.; et al. The global distribution and burden of dengue. Nature 2013, 496, 504–507. [Google Scholar] [CrossRef]
  42. Brady, O.J.; Gething, P.W.; Bhatt, S.; Messina, J.P.; Brownstein, J.S.; Hoen, A.G.; Moyes, C.L.; Farlow, A.W.; Scott, T.W.; Hay, S.I. Refining the Global Spatial Limits of Dengue Virus Transmission by Evidence-Based Consensus. PLoS Negl. Trop. Dis. 2012, 6, e1760. [Google Scholar] [CrossRef]
  43. 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, 517–525. [Google Scholar] [CrossRef]
  44. US Food and Drug Administration. Coronavirus (COVID-19) Update: FDA Revokes Emergency Use Authorization for Chloroquine and Hydroxychloroquine. 15 June 2020. Available online: https://www.fda.gov/media/138945 (accessed on 11 September 2020).
  45. US Food and Drug Administration. FDA Cautions against Use of Hydroxychloroquine or Chloroquine for COVID-19 Outside of the Hospital Setting or a Clinical Trial due to Risk of Heart Rhythm Problems. 1 July 2020. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/nda/2020/OSE%20Review_Hydroxychloroquine-Cholorquine%20-%2019May2020_Redacted.pdf (accessed on 11 September 2020).
  46. Cao, B.; Wang, Y.; Wen, D.; Liu, W.; Wang, J.; Fan, G.; Ruan, R.; Song, B.; Cai, C.; 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]
  47. Wang, Y.; Zhang, D.; Du, G.; Du, R.; Zhao, J.; Jin, Y.; Fu, S.; Gao, L.; Cheng, Z.; Lu, Q.; et al. Remdesivir in adults with severe COVID-19: A randomised, double-blind, placebo-controlled, multicentre trial. Lancet 2020, 395, 1569–1578. [Google Scholar] [CrossRef]
  48. 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. Prioritisation of Anti-SARS-Cov-2 Drug Repurposing Opportunities Based on Plasma and Target Site Concentrations Derived From Their Established Human Pharmacokinetics. Clin. Pharm. Ther. 2020. [Google Scholar] [CrossRef] [PubMed]
  49. Schmith, V.D.; Zhou, J.J.; Lohmer, L.R. The Approved Dose of Ivermectin Alone is not the Ideal Dose for the Treatment of COVID-19. Clin. Pharm. Ther. 2020. [Google Scholar] [CrossRef] [PubMed]
  50. Trial Site News. Eurnekian Public Hospital Argentina’s IVERCAR Ivermectin & Carrageenan Study Shows Positive Results Targeting COVID-19. 3 September 2020. Available online: https://www.trialsitenews.com/argentinas-ivercar-ivermectin-carrageenan-study-shows-positive-results-targeting-covid-19/ (accessed on 11 September 2020).
  51. Alam, M.; 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. Bangladesh Coll. Phys. Surg. 2020, 38, 10–15. [Google Scholar] [CrossRef]
  52. 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 Hydroxychloroquine with Azithromycin in COVID-19 Patients. Bangladesh Coll. Phys. Surg. 2020, 38, 5–9. [Google Scholar] [CrossRef]
  53. Republica Del Peru Ministerio De Salud. RM_270-2020-MINSA. 8 May 2020. Available online: https://cdn.www.gob.pe/uploads/document/file/694719/RM_270-2020-MINSA.PDF (accessed on 11 September 2020).
  54. The Gobierno del Estate Plurinacional de Bolivia Misterio de Salud. Ministerial Resolution No 259 from the Gobierno del Estate Plurinacional de Bolivia Misterio de Salud. 20 May 2020. Available online: https://www.minsalud.gob.bo/component/jdownloads/send/27-comunicado-oficial/425-resolucion-ministerial-n-0259 (accessed on 11 September 2020).
Figure 1. Schematic showing IMPα’s role in nuclear transport of host and viral proteins, and mechanism of inhibition by ivermectin. (a) Host proteins, such as members of the STAT or NF-κB transcription factor families, localize in the nucleus through the action of the IMPα/β1 heterodimer, where the “IBB” (IMPβ-binding) region of IMPα (green curved line) is bound by IMPβ1 to enable cargo recognition by IMPα within the heterodimer; IMPβ1 subsequently mediates transport of the trimeric complex through the nuclear pore (NPC, nuclear pore complex) embedded within the nuclear envelope (NE) into the nucleus. This is followed by release within the nucleus to enable the transcription factors to carry out normal function in transcriptional regulation, including in the antiviral response. IMPα can only mediate nuclear import within the heterodimer with IMPβ1. (b) In viral infection, specific viral proteins (e.g., NS5 in the case of DENV, ZIKV, WNV) able to interact with IMPα utilize the IMPα/β1 heterodimer to access the nucleus and antagonize the antiviral response [14,27,28]. This is critical to enable optimal virus production as shown by mutagenic and inhibitor studies. Which SARS-CoV-2 proteins may access the nucleus in infected cells has not been examined (see Section 3). (c) The IMPα targeting compound ivermectin binds to IMPα (binding site shown as red lozenge) both within the IMPα/β heterodimer to dissociate it, and to free IMPα to prevent it binding to IMPβ1, thereby blocking NS5 nuclear import [11]. GW5074 (see Table 1) has been shown to exhibit a similar mechanism [29].
Figure 1. Schematic showing IMPα’s role in nuclear transport of host and viral proteins, and mechanism of inhibition by ivermectin. (a) Host proteins, such as members of the STAT or NF-κB transcription factor families, localize in the nucleus through the action of the IMPα/β1 heterodimer, where the “IBB” (IMPβ-binding) region of IMPα (green curved line) is bound by IMPβ1 to enable cargo recognition by IMPα within the heterodimer; IMPβ1 subsequently mediates transport of the trimeric complex through the nuclear pore (NPC, nuclear pore complex) embedded within the nuclear envelope (NE) into the nucleus. This is followed by release within the nucleus to enable the transcription factors to carry out normal function in transcriptional regulation, including in the antiviral response. IMPα can only mediate nuclear import within the heterodimer with IMPβ1. (b) In viral infection, specific viral proteins (e.g., NS5 in the case of DENV, ZIKV, WNV) able to interact with IMPα utilize the IMPα/β1 heterodimer to access the nucleus and antagonize the antiviral response [14,27,28]. This is critical to enable optimal virus production as shown by mutagenic and inhibitor studies. Which SARS-CoV-2 proteins may access the nucleus in infected cells has not been examined (see Section 3). (c) The IMPα targeting compound ivermectin binds to IMPα (binding site shown as red lozenge) both within the IMPα/β heterodimer to dissociate it, and to free IMPα to prevent it binding to IMPβ1, thereby blocking NS5 nuclear import [11]. GW5074 (see Table 1) has been shown to exhibit a similar mechanism [29].
Cells 09 02100 g001
Table 1. In vitro properties of IMPα inhibitors with antiviral effects.
Table 1. In vitro properties of IMPα inhibitors with antiviral effects.
CompoundDocumented Action on IMPα 3Antiviral Against 4Inhibitory Concentration (Assay)/Fold reduction 5
Ivermectin 1
  • Inhibits interaction in vitro of IMPα with HIV-IN [34], DENV2 NS5 (1 μM) [7,11], T-ag [31], Hendra V (15 μM) [13], IMPβ1 (7 μM) [11]
  • Inhibits interaction of IMPα with T-ag and NS5 in a cell context as visualised by quantitative BiFc [11]
  • Inhibits CoIP from cell lysates of IMPα with T-ag, Adenovirus EIA [20]
  • Inhibits nuclear accumulation in a cellular context of IMPα/β1- but not β1-recognised viral proteins such as T-ag [7,16], DENV2 NS5 [7], VEEV Capsid [16], adenovirus E1A [20], PSV UL42 [18] as well as host cargoes (see [15,30])
  • Reduces nuclear localisation in infected cells of VEEV Capsid [9] and adenovirus E1A [20]
Coronavirus
SARS-CoV-2
HIV-1 (VSV-G-pseudotyped NL4-3.Luc.R-E-HIV)
Influenza VLPs (avian influenza A/MxA escape mutants)
Flavivirus:
YFV (17D)
DENV1 (EDEN-1)
DENV2 (NGC)
DENV2 (EDEN-2)
DENV3 (EDEN-3)
DENV4 (EDEN-4)
WNV (NY99)
WNV (MRM61C)
ZIKV (Asian/Cook Islands/2014)
Alphavirus:
Chikungunya virus (CHIKV-Rluc)
Sindbis (HR)
Semliki forest virus
VEEV (TC83)
Hendra (Hendra virus/Australia/Horse/1994)
DNA viruses
Adenovirus (HAdV-C5)
Adenovirus (HAdV-B3)
BK polyomavirus (BKPyV)
Pseudorabies
EC50 = 2.2/2.8 μM (qPCR/released/cell-associated virus) [17] 5000-fold
50 μM > 2-fold (luciferase) [7]
10 μM total inhibition (luciferase) [10]
EC50 = 5/0.5 nM (CPE/qPCR) [12]
3 μM > 50,000-fold (pfu) [15]
EC50 = 2.3/3 μM (CFI, 2 hosts) [8]
EC50 = 0.7 μM (qPCR) [12]
EC50 = 0.4/0.6 μM (pfu/qPCR) [11]
50 μM total inhibition (pfu) [7]
EC50 = 2.1/1.7 μM (CFI, 2 hosts) [8]
EC50 = 1.7 μM (CFI) [8]
EC50 = 1.9 μM (CFI) [8]
EC50 = 4 μM (qPCR) [12]
EC50 = 1/0.5 μM (pfu/qPCR) [11]
EC50 = 1.3/1.6 μM (pfu/qPCR) [11]
EC50 = 1.9/0.6 μM (luciferase, 2 hosts)
3 μM > 5000-fold (pfu) [15]
3 μM > 1000-fold (pfu) [15]
3 μM > 200-fold (pfu) [15]
1 μM c. 20-fold (pfu) [9]
est. EC50 = 2 μM (TCID/luciferase) [13]
EC50 = c. 2.5 μM; 10 μM 20-fold (qPCR) [20]
10 μM c. 8-fold (qPCR) [20]
Est. EC50 1.5 μM (PFU/CPE/qPCR) [19]
Est. EC50 c. 0.8 μM 1000-fold [18]
Gossypol 2
  • Inhibits interaction in vitro of IMPα with Hendra Virus V (10 μM) [14]
  • Inhibits nuclear accumulation in WNV infected cells of NS5 [36]
WNV (MRM61C)
Hendra (Hendra virus/Australia/Horse/1994)
10 μM 100-fold (pfu) [36]
10 μM (TCID/luciferase) [14]
GW5074
  • Inhibits interaction in vitro of IMPα with DENV2 NS5 (5 μM) [29], Hendra (V 15 uM) [13], IMPβ1 (10 μM) [29]
  • Inhibits nuclear accumulation in DENV2 infected cells of NS5 [29]
DENV2 NGC
ZIKV (Asian/Cook Islands/2014)
WNV (MRM61C)
EC50 = 0.5/1 (pfu/PCR) [29]
EC50 = 0.3/0.6 (pfu/PCR) [29]
EC50 = 5/7 (pfu/PCR) [29]
1 US Food and Drug Administration (FDA)-approved broad-spectrum antiparasitic agent, including against parasitic worm infestations and ectoparasites causing scabies, pediulosis and rosacea [1,3]. 2 Abandoned as human male contraceptive due to side effects [38,39]. 3 Inhibits helicase activity (FRET based assay) of DENV2, YFV and WNV NS3 (IC50 0.2–0.5 μM) [12]. 4 Entries in brackets indicate virus strains/constructs used. 5 Est. estimated.
Table 2. Summary of current clinical trials using ivermectin for SARS-CoV-2.
Table 2. Summary of current clinical trials using ivermectin for SARS-CoV-2.
Title, URLStatus 1N 2Interventions 3StartLocations
1Ivermectin Effect on SARS-CoV-2 Replication in Patients With COVID-19;
https://ClinicalTrials.gov/show/NCT04381884
R45Ivermectin 0.6 mg/kg QD plus SC vs. SC18.5.20 CEMIC, Buenos Aires, Ciudad De Buenos Aires, Argentina
2Ivermectin and Nitazoxanide Combination Therapy for COVID-19;
https://ClinicalTrials.gov/show/NCT04360356
NY100Ivermectin 0.2 mg/kg once plus NZX 500 mg BID for 6 days vs. SC20.5.20Tanta University, Egypt
3Ivermectin vs. Placebo for the Treatment of Patients With Mild to Moderate COVID-19;
https://ClinicalTrials.gov/show/NCT04429711
R100Ivermectin 12–15 mg/day for 3 days vs. Placebo12.5.20Sheba Medical Center, Ramat-Gan, Israel
4Hydroxychloroquine and Ivermectin for the Treatment of COVID-19 Infection;
https://ClinicalTrials.gov/show/NCT04391127
R200Ivermectin 12 mg (<80 kg) or 18 mg (>80 kg) once vs. HCQ 400 mg BID for 1 day then 200 mg BID for 4 days vs. Placebo4.5.20Jose Manuel Arreola Guerra, Aguascalientes, Mexico
5Efficacy of Ivermectin in Adult Patients With Early Stages of COVID-19.
https://ClinicalTrials.gov/show/NCT04405843
NY400Ivermectin 0.3 mg/kg daily for 5 days vs. Placebo20.6.20Colombia
6Ivermectin In Treatment of COVID 19 Patients.
https://ClinicalTrials.gov/show/NCT04425707
R100Ivermectin (dose unlisted) vs. SC vs. Ivermectin (dose unlisted) plus SC9.6.20Isolation and referral hospitals for COVID 19 patients, Cairo, Egypt
7Efficacy and Safety of Ivermectin and Doxycycline in Combination or IVE Alone in Patients With COVID-19 Infection;
https://ClinicalTrials.gov/show/NCT04407130
E72Ivermectin 0.2 mg/kg once plus 200 mg DOC day 1 followed by 100 mg DOC BID for 4 days vs. Ivermectin 0.2 mg/kg QD for 5 days vs. Placebo16.6.20Icddr, B, Dhaka, Bangladesh
8Efficacy of Ivermectin as Add on Therapy in COVID19 Patients.
https://ClinicalTrials.gov/show/NCT04343092
C100Ivermectin 0.2 mg/kg once weekly plus HCQ 400 mg QD plus ATM 500 mg QD vs. HCQ 400 mg QD plus ATM 500 mg QD18.4.20General Directorate of Medical City, Bagdad, Baghdad, Iraq
9COVidIVERmectin: Ivermectin for Treatment of Covid-19 (COVER).
https://ClinicalTrials.gov/show/NCT04438850
NY102Ivermectin 0.6 mg/kg QD for 5 days vs. Ivermectin 1.2 mg/kg QD for 5 days vs. Placebo20.6.20Negrar, Verona, Italy; Bologna, Italy; Milan, Italy; Rovereto, Italy; Turin, Italy; Barcelona, Spain; Madrid, Spain
10Efficacy, Safety and Tolerability of Ivermectin in Subjects Infected With SARS-CoV-2 With or Without Symptoms (SILVERBULLET).
https://ClinicalTrials.gov/show/NCT04407507
NY66Ivermectin 12 mg/day for 3 days plus paracetamol 500 mg QID for 14 days vs. Placebo plus paracetamol 500 mg QID for 14 days20.6.20Investigacion Biomedica para el Desarrollo de Farmacos S.A. de C.V., Mexico
11Sars-CoV-2/COVID-19 Ivermectin Navarra-ISGlobal Trial (SAINT).
https://ClinicalTrials.gov/show/NCT04390022
R24Ivermectin 0.4 mg/kg once vs. Placebo14.5.20Clinica Universidad de Navarra, Pamplona, Navarra, Spain
12A Comparative Study on Ivermectin and Hydroxychloroquine on the COVID19 Patients in Bangladesh.
https://ClinicalTrials.gov/show/NCT04434144
C116Ivermectin 0.2 mg/kg once plus DOC 100 mg BID for 10 days vs. HCQ 400 mg day 1 then 200 mg BID for 9 days plus ATM 500 mg/day for 5 days2.5.20Chakoria Upazilla Health Complex, Cox’s Bazar, Bangladesh
13Ivermectin vs Combined Hydroxychloroquine and Antiretroviral Drugs (ART) Among Asymptomatic COVID-19 Infection (IDRA-COVID19).
https://ClinicalTrials.gov/show/NCT04435587
NY80Ivermectin 0,6 mg/kg daily for 3 days vs. HCQ 400 mg BID Day 1 then 200 mg BID for 4 days plus Darunavir/ritonavir (400 mg/100 mg) BID for 5 days20.7.20Siriraj Hospital, Bangkok Noi, Bangkok, Thailand
14IVERMECTIN Aspirin Dexametasone and Enoxaparin as Treatment of Covid 19.
https://ClinicalTrials.gov/show/NCT04425863
A100Ivermectin 5 mg/mL oral to be repeated 1 week later (dose unlisted)1.5.20Hospital Eurnekian, Buenos Aires, Argentina
15A Preventive Treatment for Migrant Workers at High-risk of Covid-19.
https://ClinicalTrials.gov/show/NCT04446104
R5000Ivermectin 12 mg once vs. HCQ 400 mg day 1 then 200 mg/day for 42 days vs. Zinc 80 mg/day plus vitamin C 500 mg/day for 42 days vs. Povidone-iodine throat spray TID for 42 days vs. Vitamin C 500 mg/day for 42 days13.5.20Tuas South Dormitory, Singapore, Singapore
16New Antiviral Drugs for Treatment of COVID-19.
https://ClinicalTrials.gov/show/NCT04392427
NY100Ivermectin (dose unlisted) plus NZX (dose unlisted) plus ribavirin 200 mg or 400 mg vs. Control (untreated)20.5.20Mansoura University, Mansoura, Select A State Or Province, Egypt
17Early Treatment With Ivermectin and LosarTAN for Cancer Patients With COVID-19 Infection (TITAN).
https://ClinicalTrials.gov/show/NCT04447235
NY176Ivermectin 12 mg once plus losartan 50 mg/day for 15 days vs. Placebo20.7.20Instituto do Cancer do Estado de São Paulo, Brazil
18Ivermectin in Treatment of COVID-19.
https://ClinicalTrials.gov/show/NCT04445311
R100Ivermectin daily (dose unlisted) for 3 days plus SC vs. SC31.5.20Waheed Shouman, Zagazig, Sharkia, Egypt
19Efficacy of Ivermectin in COVID-19.
https://ClinicalTrials.gov/show/NCT04392713
R100Ivermectin 12 mg once plus SC vs. SC15.4.20Combined Military Hospital Lahore, Lahore, Punjab, Pakistan
20Ivermectin and Doxycycine in COVID-19 Treatment.
https://ClinicalTrials.gov/show/NCT04403555
NY40Ivermectin (dose unlisted) plus DOC (dose unlisted) vs. CQ (dose unlisted)1.6.20Sherief Abd-Elsalam, Tanta, Egypt
21The Efficacy of Ivermectin and Nitazoxanide in COVID-19 Treatment.
https://ClinicalTrials.gov/show/NCT04351347
R300Ivermectin (dose unlisted) vs. Ivermectin (dose unlisted) plus NZX (dose unlisted) vs. Ivermectin (dose unlisted) plus CQ (dose unlisted)16.6.20Tanta University, Tanta, Egypt
22Prophylactic Ivermectin in COVID-19 Contacts.
https://ClinicalTrials.gov/show/NCT04422561
C 4304Ivermectin 15 mg (40–60 kg), 18 mg (60–80 kg) or 24 mg (> 80 kg) per day, 2 doses 72 h apart vs. Control (untreated)31.5.20Zagazig University, Zagazig, Sharkia, Egypt
23Max Ivermectin- COVID 19 Study Versus Standard of Care Treatment for COVID 19 Cases. A Pilot Study.
https://ClinicalTrials.gov/show/NCT04373824
R50Ivermectin 0.2 mg/kg daily for 2 days plus SC vs. SC25.4.20Max Super Speciality hospital, Saket (A unit of Devki Devi Foundation), New Delhi, Delhi, India
24A Study to Compare the Efficacy and Safety of Different Doses of Ivermectin for COVID-19 (IFORS)
https://ClinicalTrials.gov/show/NCT04431466
NY64Ivermectin 0.1 mg/kg once vs. Ivermectin 0.1 mg/kg day 1 and repeated after 72 h vs. Ivermectin 0.2 m/kg once vs. Ivermectin 0.2 mg/kg day 1 and repeated after 72 h vs. SC1.7.20Hospital Univeristário da Universidade Federal de São Carlos (HU-UFSCar), São Carlos, São Paulo, Brazil
25Novel Agents for Treatment of High-risk COVID-19 Positive Patients
https://ClinicalTrials.gov/show/NCT04374019
R240Ivermectin 12 mg (<75 kg) or 15 mg (>75 kg) daily for 2 days vs. HCQ 600 mg/day for 14 days plus ATM 500 mg day 1 then 250 mg/day for 4 days vs. Camostat Mesilate 200 mg TID for 14 days vs. Artemesia annua 50 mg TID for 14 days1.5.20University of Kentucky Markey Cancer Center, Lexington, Kentucky, United States
26Ivermectin-Azithromycin-Cholecalciferol (IvAzCol) Combination Therapy for COVID-19 (IvAzCol)
https://ClinicalTrials.gov/show/NCT04399746
R30Ivermectin 6 mg/day on days 0, 1, 7 and 8 plus ATM 500 mg/day 4 days plus Cholecalciferol 400 IU BID for 30 days vs. Control (untreated)15.3.20Outpatient treatment, Mexico City, Mexico
27USEFULNESS of Topic Ivermectin and Carrageenan to Prevent Contagion of Covid 19 (IVERCAR)
https://ClinicalTrials.gov/show/NCT04425850
A 51195Ivermectin (topical for oral mucosae) plus iota carrageenan (topical for oral mucosae) 5 times per day plus PPE vs. PPE only1.6.20Hospital Eurnekian, Buenos Aires, Argentina
28Novel Regimens in COVID-19 Treatment
https://ClinicalTrials.gov/show/NCT04382846
NY80Ivermectin plus CQ (dose unlisted) vs. Ivermectin plus NZX (dose unlisted) vs. Ivermectin plus NZX plus ATM (dose unlisted) vs. NZX and ATM (dose unlisted)8.5.20Tanta University, Egypt
29Anti-Androgen Treatment for COVID-19
https://ClinicalTrials.gov/show/NCT04446429
NY254Ivermectin 0.2 mg/kg QD plus ATM 500 mg QD vs. Ivermectin 0.2 mg/kg QD plus ATM 500 mg QD plus Dutasteride 0.5 mg QD26.6.20Corpometria Institute, Brasilia, Brazil
30A Real-life Experience on Treatment of Patients With COVID 19
https://ClinicalTrials.gov/show/NCT04345419
R120Ivermectin (dose unlisted) vs. CQ (dose unlisted) vs. Favipiravir (dose unlisted) vs. NZX (dose unlisted) vs. Niclosamide (dose unlisted) vs. other drugs (oseltamivir or combination of above, dose unlisted)16.6.20Tanta university hospital, Tanta, Egypt
31Worldwide Trends on COVID-19 Research After the Declaration of COVID-19 Pandemic (observational)
https://ClinicalTrials.gov/show/NCT04460547
NY200Completed interventional vs. completed observational studies on Ivermectin, Convalescent Plasma, HCQ, DAS181, or Interferon 1A 25.7.20Qassim University, Saudi Arabia
32Trial of Combination Therapy to Treat COVID-19 Infection
https://ClinicalTrials.gov/show/NCT04482686
NY300Ivermectin (dose unlisted) day 1 and 4 plus DOC (dose unlisted) for 10 days plus Zinc for 10 days plus Vitamin D3 for 10 days plus Vitamin C for 10 days vs. Placebo22.7.20ProgenaBiome, California, USA
33Randomised clinical trial of ivermectin for treatment and prophylaxis of COVID-19
https://www.clinicaltrialsregister.eu/ctr-search/trial/2020-001994-66/ES
O266Ivermectin (dose unlisted) vs. Placebo8.5.20Fundació Assistencial Mútua Terrassa, Spain
34Multicenter, randomized, double-blind, placebo-controlled study investigating efficacy, safety and tolerability of ivermectin HUVE-19 in patients with proven SARS-CoV-2 infection (COVID-19) and manifested clinical symptoms.
https://www.clinicaltrialsregister.eu/ctr-search/trial/2020-002091-12/BG
O120Ivermectin 0.4 mg/kg plus SC vs. Placebo plus SC5.5.20Bulgaria (9 sites)
35Efficacy of hydroxychloroquine, ciclesonide and ivermectin in treatment of moderate covid-19 illness: an open-label randomised controlled study (EHYCIVER-COVID)
http://ctri.nic.in/ClinicaltrialsCTRI/2020/04/024948
NY120Ivermectin 12 mg/day for 7 days vs. Ciclesonide 0.2 mg/kg BID for 7 days vs. HCQ 400 mg BID Day 1 then 200 mg BID for 6 days vs. SC15.5.20New Delhi, India
36A Phase IIB open label randomized controlled trial to evaluate the efficacy and safety of Ivermectin in reducing viral loads in patients with hematological disorders who are admitted with COVID 19 infection
http://ctri.nic.in/ClinicaltrialsCTRI/2020/04/025068
NY50Ivermectin 3 mg (15–24 kg) or 6 mg (25–35 kg) or 9 mg (36–50 kg) or 12 mg (51–65 kg) or 15 mg (66–79 kg) or 0.2 mg/kg (80 kg) once vs. SC27.5.20Christian Medical College Vellore, TAMIL NADU, India
37Interventional study to assess the efficacy of Ivermectin with standard of care treatment versus standard of care in patients of COVID-19 at R D Gardi Medical College, Ujjain, India
http://ctri.nic.in/ClinicaltrialsCTRI/2020/04/025224
NY50Ivermectin 12 mg/day for 2 days plus SC vs. SC24.5.20R D Gardi Medical College, Ujjain, Madhya Pradesh, India
38Study to assess the efficacy of Ivermectin as prophylaxis of COVID 19 among health care workers and COVID 19 contacts in Ujjain, India;
http://ctri.nic.in/ClinicaltrialsCTRI/2020/04/025333
NY2000Ivermectin 12 mg/day (adult) or 6 mg/day (children) for 2 days vs. Control27.5.20R D Gardi Medical College, Ujjain, Madhya Pradesh, India
39Randomised Controlled Trial of Ivermectin in hospitalised patients with COVID19 (RIVET-COV).
http://ctri.nic.in/ClinicaltrialsCTRI/2020/04/026001
NY60Ivermectin single dosing of 0.2 mg/kg vs. Ivermectin 0.4 mg/kg vs. Ivermectin 0.8 mg/kg vs. Ivermectin 1.6 mg/kg vs. Ivermectin 2 mg/kg vs. SC25.6.20New Delhi, India
40A Prospective, randomized, single centred, open labelled, two arm, placebo-controlled trial to evaluate efficacy and safety of Ivermectin drug in patients infected with SARS-CoV-2 virus;
http://ctri.nic.in/ClinicaltrialsCTRI/2020/04/025960
NY100Ivermectin 12 mg/day for 3 days vs. SC18.6.20Symbiosis University Hospital and Research Centre, Maharashtra, India
41A Clinical Trial to Study the Efficacy of “Ivermectin” in the prevention of Covid-19. A Single Arm Study.
http://ctri.nic.in/ClinicaltrialsCTRI/2020/04/026232
NY50Ivermectin 0.2 mg/kg once10.7.20DVFM, Andhra Pradesh, India
42Ivermectin Nasal Spray for COVID19 Patients.
https://ClinicalTrials.gov/show/NCT04510233
NY60Ivermectin nasal spray (1 mL) in each nostril BID vs. Ivermectin oral (6 mg) TID vs. SC10.8.20Tanta University, Tanta, Egypt
43Outpatient use of ivermectin in COVID-19.
https://ClinicalTrials.gov/show/NCT04530474
NY200Ivermectin 0.15–0.2 mg/kg (max 12 mg) once vs. Placebo26.8.20Temple University Hospital, Philadelphia, USA
44Ivermectin to prevent hospitilizations in COIVD-19.
https://ClinicalTrials.gov/show/NCT04529525
R500Ivermectin 12 mg (48–80 kg) or 18 mg (80–110 kg) or 24 mg (>100 kg) at inclusion and again at 24h vs. Placebo21.8.20Ministry of Public Health, Province of Corrientes, Argentina
45Clinical trial of ivermectin plus doxycycline for the treatment of confirmed Covid-19 infection.
https://ClinicalTrials.gov/show/NCT04523831
R400Ivermectin 6 mg and doxycycline 100 mg BID for 5 days vs. Placebo19.8.20Dhaka Medical College, Dhaka Bangladesh
46Pilot study to evaluate the potential of ivermectin to reduce COVID-19 transmission.
https://www.clinicaltrialsregister.eu/ctr-search/trial/2020-001474-29/ES
O24Ivermectin (dose unlisted) vs. Placebo8.5.20Clinica Universidad de Navarra, Pamplona, Spain
47Dose-Finding study of Ivermectin treatment on patients infected with Covid-19: A clinical trial.
https://en.irct.ir/trial/47012
A125Ivermectin 0.2 mg/kg single dose plus SC vs. Ivermectin 0.2 mg/kg day 1, 2, 5 plus SC vs. Placebo plus SC vs. Ivermectin 0.4 mg/kg day 1 and 0.2 mg/kg day 2, 5 vs. SC4.5.20Qazvin University of Medical Sciences, Qazvin, Iran
48In vivo use of ivermectin (IVR) for treatment for corona virus infected patients: a randomized controlled trial.
http://www.chictr.org.cn/showprojen.aspx?proj=54707
NY60Ivermectin single dose 0.2 mg/kg vs. Placebo10.6.20Rayak Hospital, Riyaq, Lebanon
49A randomized clinical trial study, comparison of the therapeutic effects of Ivermectin, Kaletra and Chloroquine with Kaletra and Chloroquine in the treatment of patients with coronavirus 2019 (COVID-19).
http://en.irct.ir/trial/48444
A60Ivermectin 0.15–0.2 mg/kg single dose day 1 plus HCQ 200 mg day 1 plus Lopinavir/Ritonavir 400/100 mg days 2–6 vs. HCQ 200 mg day 1 plus Lopinavir/Ritonavir 400/100 mg days 2-630.5.20Ahvaz Razi Hospital, Ahvaz, Iran
50A double-blind clinical trial to repurpose and assess the efficacy and safety of ivermectin in COVID-19.
http://isrctn.com/ISRCTN40302986
R45Ivermectin 6 mg every 3.5 days for 2 weeks vs. Ivermectin 12 mg every 3.5 days for 2 weeks vs. Placebo23.4.20Lagos University Teaching Hospital, Lagos, Nigeria
51Effectiveness of Ivermectin in the Treatment of Coronavirus Infection in Patients admitted to Educational Hospitals of Mazandaran in 2020.
https://en.irct.ir/trial/49174
R60Ivermectin 0.2 mg/kg once plus SC vs. SC21.5.20Bouali Hospital, Sari, Iran
52Subcutaneous Ivermectin in Combination With and Without Oral Zinc and Nigella Sativa: a Placebo Randomized Control Trial on Mild to Moderate COVID-19 Patients.
https://clinicaltrials.gov/ct2/show/study/NCT04472585
R40Ivermectin 0.2 mg/kg subcutaneous injection every 2 days plus SC vs. Ivermectin 0.2 mg/kg subcutaneous injection every 2 days plus 80 mg/kg Nigella Sativa oral QD plus SC vs. Ivermectin 0.2 mg/kg subcutaneous injection every 2 days plus 20 mg Zinc Sulfate oral TID plus SC vs. Placebo plus SC14.7.20Shaikh Zayed Hospital, Lahore, Pakistan
53Pragmatic study “CORIVER”: Ivermectin as antiviral treatment for patients infected by SARS-COV2 (COVID-19).
https://www.clinicaltrialsregister.eu/ctr-search/trial/2020-001971-33/ES
O45Ivermectin 0.2–0.4 mg/kg (regime unlisted) vs. HCQ 400 mg vs. ATM 500 mg vs. Placebo22.7.20Hospital Universitario Virgen de las Nieves, Granada, Spain
54Effectiveness and Safety of Ivermectin for the Prevention of Covid-19 Infection in Colombian Health Personnel at All Levels of Care, During the 2020 Pandemic: A Randomized Clinical Controled Trial.
https://clinicaltrials.gov/ct2/show/record/NCT04527211
NY550Ivermectin 0.2 mg/kg weekly for 7 weeks vs. Placebo7.9.20Pontificia Universidad Javeriana, Valle Del Cauca, Colombia
1 R, Recruiting, NY, Not yet recruiting, A, Active not recruiting, C, Completed, E, Enrolling by invitation, O, ongoing 2 Number of patients 3 SC, standard care, QD, once per day, BID, twice daily, QID, 4 times daily, TID, 3 times daily, PPE, personal protective equipment, vs. versus, HCQ, hydroxychloroquine (US Food and Drug Administration approval was rescinded for COVID-19) [44,45]; DOC, doxycycline; CQ, chloroquine, ATM, Azithromycin, NZX, Nanozoxide 4 Raw data for asymptomatic family close contacts of confirmed COVID patients show that 2 doses of ivermectin 72 h apart resulted in only 7.4% of 203 subjects reporting symptoms of SARS-CoV-2 infection, in contrast to control untreated subjects, of whom 58.4% reported symptoms; evidence of prophylaxis. 5 Preliminary results for 1195 subjects are consistent with prophylaxis [50].

Share and Cite

MDPI and ACS Style

Jans, D.A.; Wagstaff, K.M. Ivermectin as a Broad-Spectrum Host-Directed Antiviral: The Real Deal? Cells 2020, 9, 2100. https://doi.org/10.3390/cells9092100

AMA Style

Jans DA, Wagstaff KM. Ivermectin as a Broad-Spectrum Host-Directed Antiviral: The Real Deal? Cells. 2020; 9(9):2100. https://doi.org/10.3390/cells9092100

Chicago/Turabian Style

Jans, David A., and Kylie M. Wagstaff. 2020. "Ivermectin as a Broad-Spectrum Host-Directed Antiviral: The Real Deal?" Cells 9, no. 9: 2100. https://doi.org/10.3390/cells9092100

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