Download as pdf or txt
Download as pdf or txt
You are on page 1of 7

Antiviral Research 178 (2020) 104787

Contents lists available at ScienceDirect

Antiviral Research
journal homepage: www.elsevier.com/locate/antiviral

The FDA-approved drug ivermectin inhibits the replication of SARS-CoV-2 in T


vitro
Leon Calya, Julian D. Drucea, Mike G. Cattona, David A. Jansb, Kylie M. Wagstaffb,∗
a
Victorian Infectious Diseases Reference Laboratory, Royal Melbourne Hospital, At the Peter Doherty Institute for Infection and Immunity, Victoria, 3000, Australia
b
Biomedicine Discovery Institute, Monash University, Clayton, Vic, 3800, Australia

A B S T R A C T

Although several clinical trials are now underway to test possible therapies, the worldwide response to the COVID-19 outbreak has been largely limited to mon-
itoring/containment. We report here that Ivermectin, an FDA-approved anti-parasitic previously shown to have broad-spectrum anti-viral activity in vitro, is an
inhibitor of the causative virus (SARS-CoV-2), with a single addition to Vero-hSLAM cells 2 h post infection with SARS-CoV-2 able to effect ~5000-fold reduction in
viral RNA at 48 h. Ivermectin therefore warrants further investigation for possible benefits in humans.

1. Introduction observed for ivermectin against Zika virus (ZIKV) in mice, but the au-
thors acknowledged that study limitations justified re-evaluation of
Ivermectin is an FDA-approved broad spectrum anti-parasitic agent ivermectin's anti-ZIKV activity (Ketkar et al., 2019). Finally, ivermectin
(Gonzalez Canga et al., 2008) that in recent years we, along with other was the focus of a phase III clinical trial in Thailand in 2014–2017,
groups, have shown to have anti-viral activity against a broad range of against DENV infection, in which a single daily oral dose was observed
viruses (Gotz et al., 2016; Lundberg et al., 2013; Tay et al., 2013; to be safe and resulted in a significant reduction in serum levels of viral
Wagstaff et al., 2012) in vitro. Originally identified as an inhibitor of NS1 protein, but no change in viremia or clinical benefit was observed
interaction between the human immunodeficiency virus-1 (HIV-1) in- (see below) (Yamasmith et al., 2018).
tegrase protein (IN) and the importin (IMP) α/β1 heterodimer re- The causative agent of the current COVID-19 pandemic, SARS-CoV-
sponsible for IN nuclear import (Wagstaff et al., 2011), Ivermectin has 2, is a single stranded positive sense RNA virus that is closely related to
since been confirmed to inhibit IN nuclear import and HIV-1 replication severe acute respiratory syndrome coronavirus (SARS-CoV). Studies on
(Wagstaff et al., 2012). Other actions of ivermectin have been reported SARS-CoV proteins have revealed a potential role for IMPα/β1 during
(Mastrangelo et al., 2012), but ivermectin has been shown to inhibit infection in signal-dependent nucleocytoplasmic shutting of the SARS-
nuclear import of host (eg. (Kosyna et al., 2015; van der Watt et al., CoV Nucleocapsid protein (Rowland et al., 2005; Timani et al., 2005;
2016)) and viral proteins, including simian virus SV40 large tumour Wulan et al., 2015), that may impact host cell division (Hiscox et al.,
antigen (T-ag) and dengue virus (DENV) non-structural protein 5 2001; Wurm et al., 2001). In addition, the SARS-CoV accessory protein
(Wagstaff et al., 2012, Wagstaff et al., 2011). Importantly, it has been ORF6 has been shown to antagonize the antiviral activity of the STAT1
demonstrated to limit infection by RNA viruses such as DENV 1-4 (Tay transcription factor by sequestering IMPα/β1 on the rough ER/Golgi
et al., 2013), West Nile Virus (Yang et al., 2020), Venezuelan equine membrane (Frieman et al., 2007). Taken together, these reports sug-
encephalitis virus (VEEV) (Lundberg et al., 2013) and influenza (Gotz gested that ivermectin's nuclear transport inhibitory activity may be
et al., 2016), with this broad spectrum activity believed to be due to the effective against SARS-CoV-2.
reliance by many different RNA viruses on IMPα/β1 during infection To test the antiviral activity of ivermectin towards SARS-CoV-2, we
(Caly et al., 2012; Jans et al., 2019). Ivermectin has similarly been infected Vero/hSLAM cells with SARS-CoV-2 isolate Australia/VIC01/
shown to be effective against the DNA virus pseudorabies virus (PRV) 2020 at an MOI of 0.1 for 2 h, followed by the addition of 5 μM iver-
both in vitro and in vivo, with ivermectin treatment shown to increase mectin. Supernatant and cell pellets were harvested at days 0–3 and
survival in PRV-infected mice (Lv et al., 2018). Efficacy was not analysed by RT-PCR for the replication of SARS-CoV-2 RNA (Fig. 1A/B).

The authors would like readers to be aware of the following letter issued by the FDA titled: “Do Not Use Ivermectin Intended for Animals as Treatment for COVID-
19 in Humans” at https://www.fda.gov/animal-veterinary/product-safety-information/fda-letter-stakeholders-do-not-use-ivermectin-intended-animals-treatment-
covid-19-humans.

Corresponding author.
E-mail address: kylie.wagstaff@monash.edu (K.M. Wagstaff).

https://doi.org/10.1016/j.antiviral.2020.104787
Received 18 March 2020; Received in revised form 27 March 2020; Accepted 29 March 2020
Available online 03 April 2020
0166-3542/ © 2020 The Author(s). Published by Elsevier B.V. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/BY-NC-ND/4.0/).
L. Caly, et al. Antiviral Research 178 (2020) 104787

(caption on next page)

2
L. Caly, et al. Antiviral Research 178 (2020) 104787

Fig. 1. Ivermectin is a potent inhibitor of the SARS-CoV-2 clinical isolate Australia/VIC01/2020. Vero/hSLAM cells were in infected with SARS-CoV-2 clinical
isolate Australia/VIC01/2020 (MOI = 0.1) for 2 h prior to addition of vehicle (DMSO) or Ivermectin at the indicated concentrations. Samples were taken at 0–3 days
post infection for quantitation of viral load using real-time PCR of cell associated virus (A) or supernatant (B). IC50 values were determined in subsequent experiments
at 48 h post infection using the indicated concentrations of Ivermectin (treated at 2 h post infection as per A/B). Triplicate real-time PCR analysis was performed on
cell associated virus (C/E) or supernatant (D/F) using probes against either the SARS-CoV-2 E (C/D) or RdRp (E/F) genes. Results represent mean ± SD (n = 3). 3
parameter dose response curves were fitted using GraphPad prism to determine IC50 values (indicated). G. Schematic of ivermectin's proposed antiviral action on
coronavirus. IMPα/β1 binds to the coronavirus cargo protein in the cytoplasm (top) and translocates it through the nuclear pore complex (NPC) into the nucleus
where the complex falls apart and the viral cargo can reduce the host cell's antiviral response, leading to enhanced infection. Ivermectin binds to and destabilises the
Impα/β1 heterodimer thereby preventing Impα/β1 from binding to the viral protein (bottom) and preventing it from entering the nucleus. This likely results in
reduced inhibition of the antiviral responses, leading to a normal, more efficient antiviral response.

At 24 h, there was a 93% reduction in viral RNA present in the su- evidence to make conclusions about the safety profile in pregnancy
pernatant (indicative of released virions) of samples treated with iver- (Navarro et al., 2020; Nicolas et al., 2020). The critical next step in
mectin compared to the vehicle DMSO. Similarly a 99.8% reduction in further evaluation for possible benefit in COVID-19 patients will be to
cell-associated viral RNA (indicative of unreleased and unpackaged examine a multiple addition dosing regimen that mimics the current
virions) was observed with ivermectin treatment. By 48 h this effect approved usage of ivermectin in humans. As noted, ivermectin was the
increased to an ~5000-fold reduction of viral RNA in ivermectin- focus of a recent phase III clinical trial in dengue patients in Thailand,
treated compared to control samples, indicating that ivermectin treat- in which a single daily dose was found to be safe but did not produce
ment resulted in the effective loss of essentially all viral material by any clinical benefit. However, the investigators noted that an improved
48 h. Consistent with this idea, no further reduction in viral RNA was dosing regimen might be developed, based on pharmacokinetic data
observed at 72 h. As we have observed previously (Lundberg et al., (Yamasmith et al., 2018). Although DENV is clearly very different to
2013; Tay et al., 2013; Wagstaff et al., 2012), no toxicity of ivermectin SARS-CoV-2, this trial design should inform future work going forward.
was observed at any of the timepoints tested, in either the sample wells Altogether the current report, combined with a known-safety profile,
or in parallel tested drug alone samples. demonstrates that ivermectin is worthy of further consideration as a
To further determine the effectiveness of ivemectin, cells infected possible SARS-CoV-2 antiviral.
with SARS-CoV-2 were treated with serial dilutions of ivermectin 2 h
post infection and supernatant and cell pellets collected for real-time 2. Methods
RT-PCR at 48 h (Fig. 1C/D). As above, a > 5000 reduction in viral RNA
was observed in both supernatant and cell pellets from samples treated 2.1. Cell culture, viral infection and drug treatment
with 5 μM ivermectin at 48 h, equating to a 99.98% reduction in viral
RNA in these samples. Again, no toxicity was observed with ivermectin Vero/hSLAM cells (Ono et al., 2001) were maintained in Earle's
at any of the concentrations tested. The IC50 of ivermectin treatment Minimum Essential Medium (EMEM) containing 7% Fetal Bovine
was determined to be ~2 μM under these conditions. Underlining the Serum (FBS) (Bovogen Biologicals, Keilor East, AUS) 2 mM L-Gluta-
fact that the assay indeed specifically detected SARS-CoV-2, RT-PCR mine, 1 mM Sodium pyruvate, 1500 mg/L sodium bicarbonate, 15 mM
experiments were repeated using primers specific for the viral RdRp HEPES and 0.4 mg/ml geneticin at 37 °C, 5% CO2. Cells were seeded
gene (Fig. 1E/F) rather than the E gene (above), with nearly identical into 12-well tissue culture plates 24 h prior to infection with SARS-CoV-
results observed for both released (supernatant) and cell-associated 2 (Australia/VIC01/2020 isolate) at an MOI of 0.1 in infection media
virus. (as per maintenance media but containing only 2% FBS) for 2 h. Media
Taken together these results demonstrate that ivermectin has anti- containing inoculum was removed and replaced with 1 mL fresh media
viral action against the SARS-CoV-2 clinical isolate in vitro, with a single (2% FBS) containing Ivermectin at the indicated concentrations or
dose able to control viral replication within 24–48 h in our system. We DMSO alone and incubated as indicated for 0–3 days. At the appro-
hypothesise that this is likely through inhibiting IMPα/β1-mediated priate timepoint, cell supernatant was collected and spun for 10 min at
nuclear import of viral proteins (Fig. 1G), as shown for other RNA 6,000 g to remove debris and the supernatant transferred to fresh col-
viruses (Tay et al., 2013; Wagstaff et al., 2012; Yang et al., 2020); lection tubes. The cell monolayers were collected by scraping and re-
confirmation of this mechanism in the case of SARS-CoV-2, and iden- suspension into 1 mL fresh media (2% FBS). Toxicity controls were set
tification of the specific SARS-CoV-2 and/or host component(s) im- up in parallel in every experiment on uninfected cells.
pacted (see (Yang et al., 2020)) is an important focus future work in this
laboratory. Ultimately, development of an effective anti-viral for SARS- 2.2. Generation of SARS-CoV-2 cDNA
CoV-2, if given to patients early in infection, could help to limit the
viral load, prevent severe disease progression and limit person-person RNA was extracted from 200 μL aliquots of sample supernatant or
transmission. Benchmarking testing of ivermectin against other poten- cell suspension using the QIAamp 96 Virus QIAcube HT Kit (Qiagen,
tial antivirals for SARS-CoV-2 with alternative mechanisms of action Hilden, Germany) and eluted in 60 μl. Reverse transcription was per-
(Dong et al., 2020; Elfiky, 2020; Gordon et al., 2020; Li and De Clercq, formed using the BioLine SensiFAST cDNA kit (Bioline, London, United
2020; Wang et al., 2020) would thus be important as soon as practic- Kingdom), total reaction mixture (20 μl), containing 10 μL of RNA
able. This Brief Report raises the possibility that ivermectin could be a extract, 4 μl of 5x TransAmp buffer, 1 μl of Reverse Transcriptase and
useful antiviral to limit SARS-CoV-2, in similar fashion to those already 5 μl of Nuclease free water. The reactions were incubated at 25 °C for
reported (Dong et al., 2020; Elfiky, 2020; Gordon et al., 2020; Li and De 10 min, 42 °C for 15 min and 85 °C for 5 min.
Clercq, 2020; Wang et al., 2020); until one of these is proven to be
beneficial in a clinical setting, all should be pursued as rapidly as 2.3. Detection of SARS-CoV-2 using a TaqMan Real-time RT-PCR assay
possible.
Ivermectin has an established safety profile for human use TaqMan RT-PCR assay were performed using 2.5 μl cDNA, 10 μl
(Gonzalez Canga et al., 2008; Jans et al., 2019; Buonfrate et al., 2019), Primer Design PrecisonPLUS qPCR Master Mix 1 μM Forward (5′- AAA
and is FDA-approved for a number of parasitic infections (Gonzalez TTC TAT GGT GGT TGG CAC AAC ATG TT-3′), 1 μM Reverse (5′- TAG
Canga et al., 2008; Buonfrate et al., 2019). Importantly, recent reviews GCA TAG CTC TRT CAC AYT T-3′) primers and 0.2 μM probe (5′-FAM-
and meta-analysis indicate that high dose ivermectin has comparable TGG GTT GGG ATT ATC-MGBNFQ-3′) targeting the BetaCoV RdRp
safety as the standard low-dose treatment, although there is not enough (RNA-dependent RNA polymerase) gene or Forward (5′-ACA GGT ACG

3
L. Caly, et al. Antiviral Research 178 (2020) 104787

TTA ATA GTT AAT AGC GT -3′), 1 μM Reverse (5′-ATA TTG CAG CAG Cell Biol. 58, 50–60.
TAC GCA CAC A-3′) primers and 0.2 μM probe (5′-FAM-ACA CTA GCC Ketkar, H., et al., 2019. Lack of efficacy of ivermectin for prevention of a lethal Zika virus
infection in a murine system. Diagn. Microbiol. Infect. Dis. 95 (1), 38–40.
ATC CTT ACT GCG CTT CG-286 NFQ-3′) targeting the BetaCoV E-gene Kosyna, F.K., et al., 2015. The importin alpha/beta-specific inhibitor Ivermectin affects
(Corman et al., 2020). Real-time RT-PCR assays were performed on an HIF-dependent hypoxia response pathways. Biol. Chem. 396 (12), 1357–1367.
Applied Biosystems ABI 7500 Fast real-time PCR machine (Applied Li, G., De Clercq, E., 2020. Therapeutic options for the 2019 novel coronavirus (2019-
nCoV). Nat. Rev. Drug Discov. 19 (3), 149–150.
Biosystems, Foster City, CA, USA) using cycling conditions of 95 °C for Lundberg, L., et al., 2013. Nuclear import and export inhibitors alter capsid protein
2 min, 95 °C for 5 s, 60 °C for 24 s. SARS-CoV-2 cDNA (Ct~28) was used distribution in mammalian cells and reduce Venezuelan Equine Encephalitis Virus
as a positive control. Calculated Ct values were converted to fold-re- replication. Antivir. Res. 100 (3), 662–672.
Lv, C., et al., 2018. Ivermectin inhibits DNA polymerase UL42 of pseudorabies virus
duction of treated samples compared to control using the ΔCt method entrance into the nucleus and proliferation of the virus in vitro and vivo. Antivir. Res.
(fold changed in viral RNA = 2^ΔCt) and expressed as % of DMSO alone 159, 55–62.
sample. IC50 values were fitted using 3 parameter dose response curves Mastrangelo, E., et al., 2012 Aug. Ivermectin is a potent inhibitor of flavivirus replication
specifically targeting NS3 helicase activity: new prospects for an old drug. J.
in GraphPad prism.
Antimicrob. Chemother. 67 (8), 1884–1894.
Navarro, M., et al., 2020. Safety of high-dose ivermectin: a systematic review and meta-
Funding analysis. J. Antimicrob. Chemother. 75 (4), 827–834.
Nicolas, P., et al., 2020. Safety of oral ivermectin during pregnancy: a systematic review
and meta-analysis. Lancet Global Health 8 (1), e92–e100.
This work was supported by a National Breast Cancer Foundation Ono, N., et al., 2001. Measles viruses on throat swabs from measles patients use signaling
Fellowship, Australia (ECF-17-007) for KMW and an National Health lymphocytic activation molecule (CDw150) but not CD46 as a cellular receptor. J.
and Medical Research Council (NHMRC), Australia Senior Prinicple Virol. 75 (9), 4399–4401.
Rowland, R.R., et al., 2005. Intracellular localization of the severe acute respiratory
Research Fellow (SPRF) (APP1103050) for DAJ. syndrome coronavirus nucleocapsid protein: absence of nucleolar accumulation
during infection and after expression as a recombinant protein in vero cells. J. Virol.
References 79 (17), 11507–11512.
Tay, M.Y., et al., 2013. Nuclear localization of dengue virus (DENV) 1-4 non-structural
protein 5; protection against all 4 DENV serotypes by the inhibitor Ivermectin.
Buonfrate, D., et al., 2019. Multiple-dose versus single-dose ivermectin for Strongyloides Antivir. Res. 99 (3), 301–306.
stercoralis infection (Strong Treat 1 to 4): a multicentre, open-label, phase 3, ran- Timani, K.A., et al., 2005. Nuclear/nucleolar localization properties of C-terminal nu-
domised controlled superiority trial. Lancet Infect. Dis. 19 (11), 1181–1190. cleocapsid protein of SARS coronavirus. Virus Res. 114 (1–2), 23–34.
Caly, L., Wagstaff, K.M., Jans, D.A., 2012. Nuclear trafficking of proteins from RNA van der Watt, P.J., et al., 2016. Targeting the nuclear import receptor Kpnbeta1 as an
viruses: potential target for anti-virals? Antivir. Res. 95, 202–206. anticancer therapeutic. Mol. Canc. Therapeut. 15 (4), 560–573.
Corman, V.M., et al., 2020. Detection of 2019 novel coronavirus (2019-nCoV) by real- Wagstaff, K.M., et al., 2011. An AlphaScreen(R)-based assay for high-throughput
time RT-PCR. Euro Surveill. 25 (3). screening for specific inhibitors of nuclear import. J. Biomol. Screen 16 (2), 192–200.
Dong, L., Hu, S., Gao, J., 2020. Discovering drugs to treat coronavirus disease 2019 Wagstaff, K.M., et al., 2012. Ivermectin is a specific inhibitor of importin alpha/beta-
(COVID-19). Drug Discov. Ther. 14 (1), 58–60. mediated nuclear import able to inhibit replication of HIV-1 and dengue virus.
Elfiky, A.A., 2020. Anti-HCV, nucleotide inhibitors, repurposing against COVID-19. Life Biochem. J. 443 (3), 851–856.
Sci. 248, 117477. Wang, M., et al., 2020. Remdesivir and chloroquine effectively inhibit the recently
Frieman, M., et al., 2007. Severe acute respiratory syndrome coronavirus ORF6 antag- emerged novel coronavirus (2019-nCoV) in vitro. Cell Res. 30 (3), 269–271.
onizes STAT1 function by sequestering nuclear import factors on the rough en- Wulan, W.N., et al., 2015. Nucleocytoplasmic transport of nucleocapsid proteins of en-
doplasmic reticulum/Golgi membrane. J. Virol. 81 (18), 9812–9824. veloped RNA viruses. Front. Microbiol. 6, 553.
Gonzalez Canga, A., et al., 2008. The pharmacokinetics and interactions of ivermectin in Wurm, T., et al., 2001. Localization to the nucleolus is a common feature of coronavirus
humans–a mini-review. AAPS J. 10 (1), 42–46. nucleoproteins, and the protein may disrupt host cell division. J. Virol. 75 (19),
Gordon, C.J., et al., 2020 Apr 10. The antiviral compound remdesivir potently inhibits 9345–9356.
RNA-dependent RNA polymerase from Middle East respiratory syndrome cor- Yamasmith, E., et al., 2018. Efficacy and safety of ivermectin against dengue infection: a
onavirus. J. Biol. Chem. 295 (15), 4773–4779. phase III, randomized, double-blind, placebo-controlled trial. In: He 34th Annual
Gotz, V., et al., 2016. Influenza A viruses escape from MxA restriction at the expense of Meeting the Royal College of Physicians of Thailand. Internal Medicine and One
efficient nuclear vRNP import. Sci. Rep. 6, 23138. Health, Chonburi, Thailand.
Hiscox, J.A., et al., 2001. The coronavirus infectious bronchitis virus nucleoprotein lo- Yang, S.N.Y., et al., 2020. The broad spectrum antiviral ivermectin targets the host nu-
calizes to the nucleolus. J. Virol. 75 (1), 506–512. clear transport importin alpha/beta1 heterodimer. Antivir. Res. 104760.
Jans, D.A., Martin, A.J., Wagstaff, K.M., 2019. Inhibitors of nuclear transport. Curr. Opin.

4
Antiviral Research 178 (2020) 104805

Contents lists available at ScienceDirect

Antiviral Research
journal homepage: www.elsevier.com/locate/antiviral

Ivermectin and COVID-19: A report in Antiviral Research, widespread interest, an FDA warning, T

two letters to the editor and the authors' responses

Caly et al. at Monash University in Australia recently published a concentration of 5μM from 2 hours post-infection SARS-CoV-2
paper in Antiviral Research, reporting that ivermectin, a medication isolate Australia/VIC01/2020 until the conclusion of the experi-
widely used for the treatment of certain parasitic diseases in humans ment. SARS-CoV-2 RNA was determined by RT-PCR at Days 0–3
and livestock animals, inhibits the replication of SARS-CoV-2 in cell in both supernatant and cell pellet experiments. The authors
noted 93–99.8% reduction in viral RNA for ivermectin versus
culture (Caly et al., 2020). Despite the authors' cautious conclusion that
DMSO control at 24h in supernatant (released virions) and cell
ivermectin "warrants further investigation for possible benefits in hu-
associated viral RNA (total virus) respectively. They also describe
mans," the paper has excited widespread interest on medical and ve- by 48 hours a ∼5000-fold reduction of viral RNA and main-
terinary websites, which often incorrectly describe the drug as a tenance of effect at 72 hours. Additional experiments were con-
treatment or cure for COVID-19. These inappropriate statements led to ducted with serial dilutions of ivermectin to establish the con-
a warning by the US FDA, that ivermectin in veterinary products should centration-response profile, and the authors describe ivermectin
not be used for human therapy, as a potent inhibitor of SARS-CoV-2, with an IC50 determined to
https://www.fda.gov/animal-veterinary/product-safety- be approximately 2 μM under these conditions.
information/fda-letter-stakeholders-do-not-use-ivermectin-intended- We sought to examine the clinical relevance of the con-
animals-treatment-covid-19-humans. centrations evaluated in these in vitro experiments to those that
may be achieved with ivermectin dosing in practice, in order to
The FDA message also explains that in vitro studies such as the re-
assist in prioritizing ongoing efforts with finding therapeutics that
port in AVR are "commonly used in the early stages of drug develop-
may be effective in COVID-19.
ment." Ivermectin is one of humanity's most important medicines
The paper by Caly et al. has also elicited two letters to the editor, (Crump and Omura, 2011) and is extensively used for 5 neglected
which are printed below, followed by the authors' response to both tropical diseases at single oral doses of 150–200 μg/kg, resulting
letters. Readers should be aware that neither the letters nor the re- in the mean peak plasma concentrations of approximately 30–47
sponse has been peer-reviewed, so appropriate caution should be used ng/mL (Merck, 2009). In Phase I studies, doses up to 2000 μg/kg
in quoting or citing them. (Guzzo et al., 2002) have been administered in a fasted state or up
Mike Bray, MD to 600 μg/kg following a standard high-fat meal. Smit et al.
Editor-in-chief (2019) report that ivermectin 600 μg/kg administered orally re-
sulted in a maximum median concentrations (Cmax) in plasma of
Antiviral Research
118.9 ng/mL (p5-p95: 45.2–455.1ng/mL), with relatively rapid
Caly L, Druce JD, Catton MG, Jans DA, Wagstaff KM, 2020. The clearance and a half-life of approximately 3–5 hours.
FDA-approved Drug Ivermectin inhibits the replication of SARS-CoV-2 Similar to Yao et al. who proposed the potential for hydro-
in vitro. Antiviral Res. Apr 3:104787. xychloroquine for treating COVID-19, (Yao et al., 2020) we ap-
plied a physiologic-based pharmacokinetic (PBPK) model of
ivermectin using the Simcyp platform to explore the plasma and
Craig R. Rayner, Karen Yeo, David Wesche, Lisa Almond, lung concentrations relative to the IC50 values against SARS-CoV-
Michael Dodds, Patrick F. Smith, Mark Sullivan 2 determined in vitro. The ivermectin PBPK model was initially
Certara, Inc, 100 Overlook Center Princeton, NJ, 08540, USA developed to facilitate drug development for parasitic diseases
including onchocerciasis and is a full model that allows predic-
tion of tissue drug concentrations. The model has been in-
To the Editor dependently verified. The predicted versus observed plasma pro-
Recently Caly et al. reported in vitro activity of ivermectin files for ivermectin across clinical studies in the Mectizan NDA
against SARS-CoV-2 following a single addition to Vero-hSLAM were well aligned, Merck (1996) indicating the base model is well
cells, and suggest that these data “demonstrate that ivermectin is defined. Furthermore, the PBPK model was able to predict iver-
worthy of further consideration as a possible SARS-CoV-2 anti- mectin exposures in plasma, adipose and skin to within 1.3-fold of
viral” (Caly et al., 2020). In isolation, these in vitro data are robust observed data in patients infected with Onchocerca volvulus
and encouraging but the report does not include a correlation of (Baraka et al., 1996)
the in vitro findings with clinically achievable plasma and, more Simulations were performed using the Simcyp Simulator
relevantly, lung concentrations that would permit the determi- Version 19 Release 1. Ten virtual trials of 10 subjects aged 18–75
nation of whether the macrocyclic lactones (and specifically in years (50% female) were simulated using the Sim-NEurCaucasion
this case ivermectin) are genuine therapeutic options. population. In the simulation, high dose ivermectin (600 μg/kg)
Caly et al. bathed Vero-hSLAM cells with ivermectin at a was administered orally, daily for 3 days and the virtual study

https://doi.org/10.1016/j.antiviral.2020.104805

Available online 21 April 2020


0166-3542/ © 2020 Published by Elsevier B.V.
Antiviral Research 178 (2020) 104805

carried on to 9 days. Dosing was in the Fed state and fraction approaches integrate in vitro findings with the in vivo situation
unbound was 0.07 (plasma) and 0.13 (lung). Simulations for and may serve to prioritize existing drugs that are candidates for
mean systemic plasma and lung tissue concentrations are shown repurposing.
in Fig. 1.
Declaration of competing interest

KY, DW, LA, MD, PS and CR work for Certara, a consulting


firm in integrated drug development and have directly consulted
with a variety of not-for-profit global health organizations,
biotechnology and pharmaceutical companies and governments
with an interest in medical countermeasures against respiratory
virus infections. MS works for Medicines Development for Global
Health and the Kirby Institute and has no conflicts of interest to
declare.

Funding information

No funding was provided to write this short communication.

References

Baraka, O.Z., et al., 1996. Ivermectin distribution in the plasma and tissues of
patients infected with Onchocerca volvulus. Eur. J. Clin. Pharmacol. 50 (5),
407–410.
Caly, L., et al., 2020. The FDA-approved drug ivermectin inhibits the replication of
SARS-CoV-2 in vitro. Antivir. Res. https://doi.org/10.1016/j.antiviral.2020.
Fig. 1. Simulated mean concentration-time profile of ivermectin in plasma 104787.
(black line) and lung tissue (blue line) following 600 μg/kg dose daily for 3 Crump, A., Omura, S., 2011. Ivermectin, 'wonder drug' from Japan: the human use
days. The 5th and 95th percentiles are also shown. The red-line is the IC50 perspective. Proc. Jpn. Acad. Ser. B Phys. Biol. Sci. 87 (2), 13–28.
(2μM) against SARS-CoV-2 determined in vitro by Caly et al. (2020). Guzzo, C.A., et al., 2002. Safety, tolerability, and pharmacokinetics of escalating
high doses of ivermectin in healthy adult subjects. J. Clin. Pharmacol. 42 (10),
1122–1133.
Pharmacodynamic response is generally achieved by ensuring Merck, 1996. Mectizan NDA 050742.
an appropriate duration of exposure above the minimum ther- Merck, 2009. Stromectol USPI. https://www.accessdata.fda.gov/drugsatfda_docs/
apeutic concentration at the site of action. Even with most gen- label/2009/050742s026lbl.pdf.
erous assumptions for clinical translation, the in vitro IC50 is > 9- Smit, M.R., et al., 2019. Pharmacokinetics-pharmacodynamics of high-dose
ivermectin with dihydroartemisinin-piperaquine on mosquitocidal activity
fold and >21-fold higher than the day 3 plasma and lung tissue
and QT-prolongation (IVERMAL). Clin. Pharmacol. Ther. 105 (2), 388–401.
simulated Cmax respectively, following a high dose ivermectin Yao, X., et al., 2020. In vitro antiviral activity and projection of optimized dosing
regimen of 600 μg/kg dose daily for 3 days. (Smit et al., 2019) design of hydroxychloroquine for the treatment of severe acute respiratory
This dose scenario, which ignores consistent exposure, exceeds syndrome coronavirus 2 (SARS-CoV-2). Clin. Infect. Dis.
the highest regulatory approved dose of ivermectin, being a 200
μg/kg single dose for the treatment of Strongyloidiasis (Merck,
2009).
Caly et al. cite the importance of regulatory approval of
ivermectin as a key part of the rationale for further evaluation François Noël
against SARS-CoV-2. However, the rigorous data review and re- Laboratory of Biochemical and Molecular Pharmacology, Institute of
assurance of a stringent regulatory authority review only applies Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro,
to currently approved doses – clinical pharmacology and tox- Brazil
icology margins (including pre-and post-natal and carcinogeni-
city studies) would, therefore, need to be recalculated. In reality,
the resultant unravelling of the supporting package of data could To the Editor,
result in lengthy delays while supporting data are revised and re- In the context of repositioning/repurposing strategy for ur-
run. gent unmet medical needs, various drugs are being proposed for
It is understandable that, faced with a devastating pandemic the treatment of COVID-19, the pandemic disease caused by
and a medical and societal imperative, there is great enthusiasm SARS-CoV-2 (Noël and Lima, 2020). This is the case for the broad-
for promising news of treatments. Picking and supporting the best spectrum macrocyclic lactone ivermectin, as reported by Caly et
therapies and preventions to tackle the COVID-19 pandemic head al. (2020) based on their data showing that ivermectin inhibits
on is one of the scientific community's most urgent priorities. To the replication of SARS-CoV-2 in vitro. However, this in vitro ac-
assist this process, the clinical pharmacological relevance of in tivity occurred at much higher concentrations (IC50 ≈ 2–3 μM)
vitro or in vivo findings should be included. In vitro promise leads than the very low (nanomolar) concentrations effective against
to clinical failure in the vast majority of cases, and in the volatile many nematode species (Geary, 2005), obtained after a usual
environment of the current pandemic, it is critical that we are dose of 200 μg/kg. This micromolar concentration is also higher
sensitive to the implications of our communication and apply our than the therapeutic peak plasma concentration (about 40 nM)
measured in humans treated for onchocerciasis control with a
resources to compounds most likely to succeed. A small window
standard dose of 150 μg/kg (Apud Shu et al., 2000) and even after
exists for the current data to have relevance for humans: we need
a high daily dose (600 μg/kg) where Cmax of 105–119 ng/ml
to confirm the effective concentrations, assess if the class of
(0.12–0.14 μM) has been obtained by PK/PD modeling (Smit et
macrocyclic lactones has similar target interactions, and under-
al., 2019).
stand the relevance of the concentrations used in vitro against
SARS-CoV-2 to those likely to be achieved at the site of action, As we previously showed (Pimenta et al., 2010) that iver-
within a dose range considered to be well tolerated. Alternative mectin is a nonselective inhibitor of three important mammalian
routes could also be considered, although these present new P-type ATPases (SERCA, Na+/K+-ATPase and H+/K+-ATPase) at
formulation and safety challenges. Modeling and simulation similar micromolar concentrations (IC50 ≈ 6–17 μM), we have to
be concerned with putative important adverse effects that this

2
Antiviral Research 178 (2020) 104805

drug could produce at the higher than usual doses that would be activity against a number of different RNA viruses in vitro (Tay et
necessary for treating COVID-19 patients. As a result, a phase 1 al., 2013; Yang et al., 2020). The way a HDA can reduce viral load
study is absolutely needed before using ivermectin since a recent is by inhibiting a key cellular process that the virus hijacks to
meta-analysis concluded that there are not enough data to sup- enhance infection by suppressing the host antiviral response.
port a recommendation for its use in higher-than-approved doses Reducing viral load by even a modest amount by using a HDA at
(Navarro et al., 2020). low dose early in infection can be the key to enabling the body's
immune system to begin to mount the full antiviral response
Declaration of competing interest before the infection takes control.
Pharmaceutical research efforts are currently underway to
No conflict. refine liquid formulations for intravenous administration of long-
acting ivermectin, develop aerosol administration, and consider
using ivermectin in combination with other agents to enhance
References efficacy at low doses. However, it is important to urge great
caution in approaching the use of ivermectin in this simplistic
Caly, L., Druce, J.D., Catton, M.G., Jans, D.A., Wagstaff, K.M., 2020c. The way, precisely because ivermectin is a HDA. Because it targets a
FDA-approved Drug Ivermectin inhibits the replication of SARS-CoV-2 in host component, it cannot be assumed that even doses lower than
vitro. Antivir. Res. https://doi.org/10.1016/j.antiviral.2020.104787.
Navarro, M., Camprubí, D., Requena-Méndez, A., Buonfrate, D.,
those discussed by Yeo et al. (2020) and Noël and Lima (2020)
Giorli, G., Kamgno, J., et al., 2020. Safety of high-dose ivermectin: a are safe in the context of a burgeoning viral infection, where a
systematic review and meta-analysis. J. Antimicrob. Chemother. 75 (4), measured immune response is key to recovery. Clinical testing of
827–834. ivermectin at any dose in the fight against viral infection must include
Noël, F., Lima, J., 2020. Pharmacological aspects and clues for the rationale use of intensive monitoring of patient well-being, to pre-empt any im-
Chloroquine/Hydroxychloroquine facing the therapeutic challenges of
munosuppressive or other adverse reactions as early as possible.
COVID-19 pandemic. Lat. Am. J. Clin. Sci. Med. Technol. 2, 28–34.
Pimenta, P.H.C., Silva, C.L.M., Noël, F., 2010. Ivermectin is a nonselective Finally, it is critically important to remember that ivermectin
inhibitor of mammalian P-type ATPases. Naunyn-Schmied Arch. Pharmacol. as an antiviral is in a very early phase – under no circumstances
381, 147–152. should self-medication be considered without the guidance of a qua-
Smit, M.R., Ochomo, E.O., Waterhouse, D., Kwambai, T.K., Abong'o, B.O., lified physician, and especially not using therapeutics designed for
Bousema, T., et al., 2019. Pharmacokinetics-pharmacodynamics of high dose
veterinary purposes!
ivermectin with Dihydroartemisinin-piperaquine on mosquitocidal activity
and QT-prolongation (IVERMAL). Clin. Pharmacol. Ther. 105 (2), 388–401.
Declaration of competing interest

Authors have no conflict of interest, with no link to any


pharma company.
David A. Jans, Kylie M. Wagstaff
Nuclear Signalling Laboratory, Monash Biomedicine Discovery Institute, Funding information
Department of Biochemistry and Molecular Biology, Monash University,
Clayton, Victoria, 3800, Australia No funding supported this letter to the editor.

Response of the authors References


To the Editor
Yeo et al. and Noël aptly point out that published studies show
Caly, L., Druce, J.D., Catton, M.G., Jans, D.A., Wagstaff, K.M., 2020b. The
that blood levels of ivermectin achieved during standard therapy FDA-approved Drug Ivermectin inhibits the replication of SARS-CoV-2 in
are much lower than the concentrations we reported as inhibitory vitro. Antiviral Res. Apr 3, 104787.
for SARS-CoV-2 in cell culture (Caly et al., 2020). Yeo et al. Tay, M.Y., Fraser, J.E., Chan, W.K., Moreland, N.J., Rathore, A.P., Wang, C.,
(2020) further explore the question via pharmacokinetic mod- Vasudevan, S.G., Jans, D.A., 2013. Nuclear localization of dengue virus
(DENV) 1-4 non-structural protein 5; protection against all 4 DENV serotypes
eling (from Certara Inc.), while Noël and Lima (2020) voices
by the inhibitor Ivermectin. Antivir. Res. 99 (3), 301–306. https://doi.org/10.
concern that if high concentrations of ivermectin could be 1016/j. 2013 Sep.
achieved, this would likely be toxic. These points are well made, Yang, S.N.Y., Atkinson, S.C., Wang, C., Lee, A., Bogoyevitch, M.A., Borg, N.A.,
and we are in agreement, but they do not address the reported Jans, D.A., 2020. The broad spectrum antiviral ivermectin targets the host
mechanism of action of ivermectin (Yang et al., 2020), and nuclear transport importin α/β1 heterodimer. Antiviral Res. 2, 104760 2020
thereby fail to highlight a further vitally important reason to be Mar.
very cautious in considering ivermectin as a therapeutic for viral
infection in human patients.
Ivermectin's key direct target in mammalian cells is a not a
Mike Bray∗, Craig Rayner, François Noël, David Jans, Kylie Wagstaff,
viral component, but a host protein important in intracellular
transport (Yang et al., 2020); the fact that it is a host-directed E-mail address: mikebrayavr@gmail.com (M. Bray).
agent (HDA) is almost certainly the basis of its broad-spectrum


Corresponding author.

You might also like