Next Article in Journal
Topical Administration of Lactiplantibacillus plantarum Accelerates the Healing of Chronic Diabetic Foot Ulcers through Modifications of Infection, Angiogenesis, Macrophage Phenotype and Neutrophil Response
Next Article in Special Issue
A Calculator for COVID-19 Severity Prediction Based on Patient Risk Factors and Number of Vaccines Received
Previous Article in Journal
High-Resolution Genotyping Unveils Identical Ampicillin-Resistant Enterococcus faecium Strains in Different Sources and Countries: A One Health Approach
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Communication

Ivermectin Does Not Protect against SARS-CoV-2 Infection in the Syrian Hamster Model

1
KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, B-3000 Leuven, Belgium
2
Global Virus Network, GVN, 725 West Lombard St, Room S413, Baltimore, MD 21201, USA
3
KU Leuven Department of Imaging and Pathology, Translational Cell and Tissue Research, Division of Translational Cell and Tissue Research, B-3000 Leuven, Belgium
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Microorganisms 2022, 10(3), 633; https://doi.org/10.3390/microorganisms10030633
Submission received: 23 February 2022 / Revised: 15 March 2022 / Accepted: 15 March 2022 / Published: 16 March 2022
(This article belongs to the Special Issue SARS-CoV-2/COVID-19: Infection Models, Therapeutics and Vaccines)

Abstract

:
Ivermectin, an FDA-approved antiparasitic drug, has been reported to have in vitro activity against SARS-CoV-2. Increased off-label use of ivermectin for COVID-19 has been reported. We here assessed the effect of ivermectin in Syrian hamsters infected with the SARS-CoV-2 Beta (B.1.351) variant. Infected animals received a clinically relevant dose of ivermectin (0.4 mg/kg subcutaneously dosed) once daily for four consecutive days after which the effect was quantified. Ivermectin monotherapy did not reduce lung viral load and even significantly worsened SARS-CoV-2-induced lung pathology. Additionally, it did not potentiate the activity of molnupiravir (LagevrioTM) when combined with this drug. This study contributes to the growing body of evidence that ivermectin does not result in a beneficial effect in the treatment of COVID-19. These findings are important given the increasing, dangerous off-label use of ivermectin for the treatment of COVID-19.

1. Introduction

Two years after it was declared a global pandemic by the World Health Organization, the coronavirus disease (COVID-19) continues its devastating impact, having claimed officially over 6 million lives as of 16 March 2022 [1]. The etiological agent, the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), first emerged in Wuhan, China in December 2019 and has since evolved into variants emerging from different regions of the world, namely the Alpha, Beta, Delta, and more recently Omicron variants of concern (VoC) [2,3]. These VoC have been implicated in a resurgence of infections and mortality, with increased transmissibility and potential escape from both vaccine- and natural-infection-induced immunity [3,4,5].
Ivermectin is an FDA-approved, broad-spectrum antiparasitic drug with a range of other activities, including antiviral and host immunomodulation effects [6]. This widely accessible drug has been attracting increasing off-label use for COVID-19 since its in vitro activity against SARS-CoV-2 was reported [7], which is of growing concern. In an in vivo model of mouse hepatitis virus, a type 2 family RNA coronavirus similar to SARS-CoV-2, ivermectin reduced the viral load and protected mice from disease [8]. In Syrian hamsters infected with SARS-CoV-2, treatment with ivermectin did not result in a direct antiviral activity, but rather apparently showed some immunomodulatory effects, improved lung pathology and clinical presentations [9]. Ivermectin is being assessed as a treatment for COVID-19 in a large number of clinical trials, with no clear evidence of its clinical benefit so far [10].
Molnupiravir (LagevrioTM, EIDD-2801) is the orally bioavailable counterpart of the ribonucleoside analogue N4-hydroxycytidine (NHC, EIDD-1931), which was initially developed for the treatment of influenza [11]. NHC exerts a broad-spectrum antiviral activity against multiple RNA viruses of different families by its incorporation into viral RNA, resulting in the accumulation of deleterious mutations in the nascent viral RNA, and consequently, error catastrophe [12]. In pre-clinical infection models in mice, Syrian hamsters, and ferrets, molnupiravir demonstrated efficacy against SARS-CoV-2 [13,14,15], including against several VoC [16]. The drug has been approved in many regions for treatment of COVID-19.
Here, we evaluate the antiviral efficacy of ivermectin against SARS-CoV-2 Beta (B.1.351) VoC in a hamster infection model alone or in combination with molnupiravir; this combination was inspired by the observation of a pronounced combined activity of molnupiravir and favipiravir [17].

2. Materials and Methods

Ivermectin was purchased from Alfa Aesar (Taiwan, China) and was first formulated as 10 mg/mL solution in propylene glycol/glycerol formal (60:40 v/v), both from Sigma (Shanghai, China) then diluted to 0.1 mg/mL stock in phosphate buffered saline (PBS). Molnupiravir (EIDD-2801) was purchased from Excenen Pharmatech Co., Ltd. (Guangzhou, China) and was formulated as 50 mg/mL stock in a vehicle containing 10% PEG400 (Sigma) and 2.5% Kolliphor-EL (Sigma) in water. The formulations of both compounds were selected based on literature [13,18].
All virus-related work was conducted in the high-containment BSL3 facilities of the KU Leuven Rega Institute (3CAPS) under licenses AMV 30112018 SBB 219 2018 0892 and AMV 23102017 SBB 219 2017 0589 according to institutional guidelines. The hamster infection model of SARS-CoV-2 has been described before [19]. Briefly, 6–8 weeks female SG hamsters were treated with vehicle, ivermectin (0.4 mg/kg, once daily (QD) subcutaneously), molnupiravir (EIDD-2801, 150 mg/kg, BID orally) or combination of both drugs (ivermectin 0.4 mg/kg QD+EIDD-2801 150 mg/kg BID) for four consecutive days starting one hour before intranasal infection with 50 µL containing 1 × 104 TCID50 of SARS-CoV-2 Beta B.1.351 variant (derived from hCoV-19/Belgium/rega-1920/2021; EPI_ISL_896474, 2021-01-11) [19]. At day four post-infection (pi), the animals were euthanized for sampling of the lungs and further analysis by i.p. injection of 500 μL Dolethal (200 mg/mL sodium pentobarbital). Lungs were collected for quantification of subgenomic viral RNA using N2 primers and probes targeting the viral nucleocapsid [20], infectious virus titers, and lung histopathology as described previously [20] (Figure 1a).

3. Results and Conclusions

We here tested the efficacy of single treatment with ivermectin (0.4 mg/kg, subcutaneous, once daily (QD)) or molnupiravir (150 mg/kg, oral BID) or the combination of both. Briefly, 6–8 weeks female SG hamsters were treated with the intended dose of each compound or the vehicle (i.e., the control group) for four consecutive days starting one hour before intranasal infection with SARS-CoV-2 Beta (B.1.351) variant (Figure 1a). At day 4 post-infection (pi), the animals were euthanized, and organs were collected for quantification of viral RNA, infectious virus titers, and lung histopathology (Figure 1a). Monotherapy with ivermectin (0.4 mg/kg, once daily) did not result in any significant reduction of viral RNA (Figure 1b) or infectious virus titers (Figure 1c) in the lung of treated hamsters as compared to the vehicle control. On the other hand, single treatment with molnupiravir (150 mg/kg BID) significantly reduced viral RNA and infectious virus titers in the lungs by 1.1 (p = 0.0048) and 1.8 (p = 0.0005) log10/mg lung tissue, respectively (Figure 1b,c). Treatment of infected hamsters with a combination of molnupiravir (150 mg/kg, BID) and ivermectin (0.4 mg/kg, once daily) resulted in a reduction of the lung viral RNA loads by 1.1 log10 (p = 0.022, Figure 1b) and lung infectious virus titers by 1.9 (P = 0.0002, Figure 1c) log10/mg lung tissue, which is similar in potency to the single treatment with molnupiravir (150 mg/kg, BID). No significant weight loss or toxicity signs were observed in any of the treated groups (Figure 1d). However, the average %body weight change on the day of the sacrifice (compared to day zero) in the ivermectin single treatment group (average of 1.7%) was markedly lower than the vehicle-treated group (average of 2.6%), Figure 1d.
The median histopathology score in the single ivermectin treatment group (5.8) was significantly higher than the vehicle treated control (median score of 4.5, p = 0.04) (Figure 2a). A significant improvement in lung pathology was observed in the single molnupiravir-treated group (median score of 3.5, p = 0.04), while the combined molnupiravir/ivermectin treatment resulted in a median histopathology score of 3.8 (Figure 2a). Hematoxylin/eosin (H&E)-stained images of lungs of the ivermectin-treated hamsters revealed severe perivascular inflammation with vasculitis and significant multifocal bronchopneumonia (Figure 2b). On the other hand, the lungs of animals treated with molnupiravir as a single treatment showed very limited peribronchial and perivascular inflammation (Figure 2b).
In our study, ivermectin did not result in any antiviral activity against SARS-CoV-2 in Syrian hamsters as monotherapy. This is consistent with a previous study published also in hamsters, wherein ivermectin was dosed once subcutaneously at 0.4 mg/kg (an antiparasitic dose used in the clinical setting) and monitored for four days [9]. However, unlike the previous study, we did not notice an improvement in SARS-CoV-2-induced lung pathology with ivermectin. Transcriptomic profiling of the lungs was performed in the previous study to compare expression levels of inflammatory genes with vehicle and ivermectin treatment, as well as a descriptive histopathological analysis which indicated that SARS-CoV-2-infected, ivermectin-treated hamsters exhibited reduced degrees of edema and congestion, with greater numbers of mononuclear cells in the alveolar spaces [9]. In our study, a cumulative lung score was performed based on ten different parameters, including edema and congestion, which had similar scorings in both vehicle-treated and ivermectin-treated groups. Additionally, no mononuclear cells in the alveolar spaces were observed. These differences in findings may be due to slight hamster age differences or virus strain differences used for infection, namely that the SARS-CoV-2 Beta variant was used in our study and the ancestral SARS-CoV-2 strain (BetaCoV/France/IDF00372/2020) was used in the previous study [9].
Additionally, ivermectin did not potentiate the activity of molnupiravir in our study, indicating that it is unlikely to have a significant role in directly or indirectly modulating SARS-CoV-2 replication. A Cmax of 80.2 ng/mL was measured in Syrian hamsters when treated once subcutaneously with 0.4 mg/kg of ivermectin [21], comparable to that measured in humans when dosed at similar levels orally [9,22]. Taken together with other available evidence, it suggests that in order to achieve therapeutic activity against SARS-CoV-2 (IC50 in Vero cells of 1750 ng/mL [7]), the dosing levels of ivermectin in humans would need to be substantially increased from the approved clinical dose to potentially toxic doses [23,24,25]. Recently, it has also been reported that ivermectin can inhibit SARS-CoV-2 in Vero cells, but not in human-airway-derived cells [26]. This is in line with the fact that several studies claiming the clinical benefits of ivermectin in COVID-19 have been found to be flawed [27], and several better-designed trials such as IVERCOR-COVID19 (NCT04529525) [28], I-TECH (NCT04920942) [29] and NCT04405843 [30] have demonstrated that ivermectin does not have clinical efficacy against COVID-19 and does not significantly prevent progression to severe disease or hospitalization.
In conclusion, we report that ivermectin, at a clinically relevant dose, does not reduce lung viral load nor improve lung pathology in Syrian hamsters infected with SARS-CoV-2 Beta variant. In fact, virus-induced lung pathology increases in severity in ivermectin-treated animals. Moreover, the drug does not potentiate the antiviral activity of molnupiravir (LagevrioTM). This study contributes to the growing body of evidence that ivermectin does not result in a clinical beneficial effect in the treatment of COVID-19 at the current approved dose. Hence, we strongly advise against the increasing, dangerous off-label use of ivermectin for the treatment of COVID-19.

Author Contributions

Conceptualization, R.A., C.S.F. and J.N.; methodology, R.A. and C.S.F.; formal analysis, C.S.F., B.W. and R.A.; data curation, R.A.; validation, R.A., C.S.F. and J.N.; writing—original draft preparation, R.A. and C.S.F.; writing—review and editing, R.A. and J.N.; visualization, R.A. and B.W.; supervision, C.S.F., R.A. and J.N.; project administration, R.A. and C.S.F.; resources, S.D.J. and J.N.; funding acquisition, L.V., D.J. and J.N. All authors have read and agreed to the published version of the manuscript.

Funding

This project has received funding from the COVID-19-Fund KU Leuven/UZ Leuven and the COVID-19 call of FWO (G0G4820N), the European Union’s Horizon 2020 research and innovation program under grant agreements No 101003627 (SCORE project) and Bill & Melinda Gates Foundation (BGMF) under grant agreement INV-006366. This work also has been done under the CARE project, which has received funding from the Innovative Medicines Initiative 2 Joint Undertaking (JU) under grant agreement No 101005077. The JU receives support from the European Union’s Horizon 2020 research and innovation programme and EFPIA and Bill & Melinda Gates Foundation, Global Health Drug Discovery Institute, University of Dundee. The content of this publication only reflects the author’s view, and the JU is not responsible for any use that may be made of the information it contains.

Institutional Review Board Statement

In this published article housing conditions and experimental procedures for animal studies were done with the approval and under the guidelines of the ethics committee of animal experimentation of KU Leuven (license P065-2020, approved on 13 May 2020).

Informed Consent Statement

Not applicable.

Data Availability Statement

All of the data generated or analyzed during this study are included.

Acknowledgments

We thank Carolien De Keyzer, Lindsey Bervoets, Thibault Francken, Stijn Hendrickx, Niels Cremers for excellent technical assistance. We are grateful to Piet Maes for kindly providing the SARS-CoV-2 strain used in this study. We thank Jef Arnout and Annelies Sterckx (KU Leuven Faculty of Medicine, Biomedical Sciences Group Management) and Animalia and Biosafety Departments of KU Leuven for facilitating the animal studies. We thank the Histology department of KU Leuven for technical support for histopathological analyses. We also thank Fran Berlioz-Seux, Rob Jordan and Betsy Russell for helpful discussion.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

References

  1. WHO. Coronavirus (COVID-19) Dashboard. Available online: https://covid19.who.int/ (accessed on 22 February 2022).
  2. Karim, S.S.A.; Karim, Q.A. Omicron SARS-CoV-2 variant: A new chapter in the COVID-19 pandemic. Lancet 2021, 398, 2126–2128. [Google Scholar] [CrossRef]
  3. Plante, J.A.; Mitchell, B.M.; Plante, K.S.; Debbink, K.; Weaver, S.C.; Menachery, V.D. The variant gambit: COVID-19′s next move. Cell Host Microbe 2021, 29, 508–515. [Google Scholar] [CrossRef] [PubMed]
  4. Davies, N.G.; Abbott, S.; Barnard, R.C.; Jarvis, C.I.; Kucharski, A.J.; Munday, J.D.; Pearson, C.A.B.; Russell, T.W.; Tully, D.C.; Washburne, A.D.; et al. Estimated transmissibility and impact of SARS-CoV-2 lineage B.1.1.7 in England. Science 2021, 372, eabg3055. [Google Scholar] [CrossRef] [PubMed]
  5. Wang, P.; Nair, M.S.; Liu, L.; Iketani, S.; Luo, Y.; Guo, Y.; Wang, M.; Yu, J.; Zhang, B.; Kwong, P.D.; et al. Antibody resistance of SARS-CoV-2 variants B.1.351 and B.1.1.7. Nature 2021, 593, 130–135. [Google Scholar] [CrossRef]
  6. Heidary, F.; Gharebaghi, R. Ivermectin: A systematic review from antiviral effects to COVID-19 complementary regimen. J. Antibiot. 2020, 73, 593–602. [Google Scholar] [CrossRef] [PubMed]
  7. Caly, L.; Druce, J.D.; Catton, M.G.; Jans, D.A.; Wagstaff, K.M. The FDA-approved drug ivermectin inhibits the replication of SARS-CoV-2 in vitro. Antiviral Res. 2020, 178, 104787. [Google Scholar] [CrossRef] [PubMed]
  8. Arévalo, A.P.; Pagotto, R.; Pórfido, J.L.; Daghero, H.; Segovia, M.; Yamasaki, K.; Varela, B.; Hill, M.; Verdes, J.M.; Vega, M.D.; et al. Ivermectin reduces in vivo coronavirus infection in a mouse experimental model. Sci. Rep. 2021, 11, 7132. [Google Scholar] [CrossRef]
  9. De Melo, G.D.; Lazarini, F.; Larrous, F.; Feige, L.; Kornobis, E.; Levallois, S.; Marchio, A.; Kergoat, L.; Hardy, D.; Cokelaer, T.; et al. Attenuation of clinical and immunological outcomes during SARS-CoV-2 infection by ivermectin. EMBO Mol. Med. 2021, 13, e14122. [Google Scholar] [CrossRef]
  10. National Institute of Health Table 2d: Ivermectin Clinical Data. Available online: https://www.covid19treatmentguidelines.nih.gov/tables/ivermectin-data/ (accessed on 22 February 2022).
  11. Toots, M.; Yoon, J.-J.; Cox, R.M.; Hart, M.; Sticher, Z.M.; Makhsous, N.; Plesker, R.; Barrena, A.H.; Reddy, P.G.; Mitchell, D.G.; et al. Characterization of orally efficacious influenza drug with high resistance barrier in ferrets and human airway epithelia. Sci. Transl. Med. 2019, 11, eaax5866. [Google Scholar] [CrossRef]
  12. Urakova, N.; Kuznetsova, V.; Crossman, D.K.; Sokratian, A.; Guthrie, D.B.; Kolykhalov, A.A.; Lockwood, M.A.; Natchus, M.G.; Crowley, M.R.; Painter, G.R.; et al. β-D-N4-Hydroxycytidine Is a Potent Anti-alphavirus Compound That Induces a High Level of Mutations in the Viral Genome. J. Virol. 2017, 92, e01965-17. [Google Scholar] [CrossRef] [Green Version]
  13. Rosenke, K.; Hansen, F.; Schwarz, B.; Feldmann, F.; Haddock, E.; Rosenke, R.; Barbian, K.; Meade-White, K.; Okumura, A.; Leventhal, S.; et al. Orally delivered MK-4482 inhibits SARS-CoV-2 replication in the Syrian hamster model. Nat. Commun. 2021, 12, 2295. [Google Scholar] [CrossRef] [PubMed]
  14. Wahl, A.; Gralinski, L.E.; Johnson, C.E.; Yao, W.; Kovarova, M.; Dinnon, K.H.; Liu, H.; Madden, V.J.; Krzystek, H.M.; De, C.; et al. SARS-CoV-2 infection is effectively treated and prevented by EIDD-2801. Nature 2021, 591, 451–457. [Google Scholar] [CrossRef] [PubMed]
  15. Cox, R.M.; Wolf, J.D.; Plemper, R.K. Therapeutically administered ribonucleoside analogue MK-4482/EIDD-2801 blocks SARS-CoV-2 transmission in ferrets. Nat. Microbiol. 2020, 6, 11–18. [Google Scholar] [CrossRef] [PubMed]
  16. Abdelnabi, R.; Foo, C.S.; De Jonghe, S.; Maes, P.; Weynand, B.; Neyts, J. Molnupiravir Inhibits Replication of the Emerging SARS-CoV-2 Variants of Concern in a Hamster Infection Model. J. Infect. Dis. 2021, 224, 749–753. [Google Scholar] [CrossRef]
  17. Abdelnabi, R.; Foo, C.S.; Kaptein, S.J.F.; Zhang, X.; Do, T.N.D.; Langendries, L.; Vangeel, L.; Breuer, J.; Pang, J.; Williams, R.; et al. The combined treatment of Molnupiravir and Favipiravir results in a potentiation of antiviral efficacy in a SARS-CoV-2 hamster infection model. EBioMedicine 2021, 72, 103595. [Google Scholar] [CrossRef]
  18. Lo, P.-K.A.; Fink, D.W.; Williams, J.B.; Blodinger, J. Pharmacokinetic studies of ivermectin: Effects of formulation. Vet. Res. Commun. 1985, 9, 251–268. [Google Scholar] [CrossRef]
  19. Abdelnabi, R.; Boudewijns, R.; Foo, C.S.; Seldeslachts, L.; Sanchez-Felipe, L.; Zhang, X.; Delang, L.; Maes, P.; Kaptein, S.J.F.; Weynand, B.; et al. Comparing infectivity and virulence of emerging SARS-CoV-2 variants in Syrian hamsters. EBioMedicine 2021, 68, 103403. [Google Scholar] [CrossRef]
  20. Kaptein, S.J.F.; Jacobs, S.; Langendries, L.; Seldeslachts, L.; ter Horst, S.; Liesenborghs, L.; Hens, B.; Vergote, V.; Heylen, E.; Barthelemy, K.; et al. Favipiravir at high doses has potent antiviral activity in SARS-CoV-2-infected hamsters, whereas hydroxychloroquine lacks activity. Proc. Natl. Acad. Sci. USA 2020, 117, 26955–26965. [Google Scholar] [CrossRef]
  21. Hanafi, H.A.; Szumlas, D.E.; Fryauff, D.J.; El-Hossary, S.S.; Singer, G.A.; Osman, S.G.; Watany, N.; Furman, B.D.; Hoel, D.F. Effects of Ivermectin on Blood-Feeding Phlebotomus papatasi, and the Promastigote Stage of Leishmania major. Vector-Borne Zoonotic Dis. 2011, 11, 43–52. [Google Scholar] [CrossRef]
  22. A Guzzo, C.; I Furtek, C.; 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]
  23. Chaccour, C.; Hammann, F.; Ramón-García, S.; Rabinovich, N.R. Ivermectin and COVID-19: Keeping Rigor in Times of Urgency. Am. J. Trop. Med. Hyg. 2020, 102, 1156–1157. [Google Scholar] [CrossRef]
  24. Jermain, B.; Hanafin, P.O.; Cao, Y.; Lifschitz, A.; Lanusse, C.; Rao, G.G. Development of a Minimal Physiologically-Based Pharmacokinetic Model to Simulate Lung Exposure in Humans Following Oral Administration of Ivermectin for COVID-19 Drug Repurposing. J. Pharm. Sci. 2020, 109, 3574–3578. [Google Scholar] [CrossRef] [PubMed]
  25. Schmith, V.D.; Zhou, J.; Lohmer, L.R. The Approved Dose of Ivermectin Alone is not the Ideal Dose for the Treatment of COVID-19. Clin. Pharmacol. Ther. 2020, 108, 762–765. [Google Scholar] [CrossRef] [PubMed]
  26. Dinesh Kumar, N.; Ter Ellen, B.M.; Bouma, E.M.; Troost, B.; van de Pol, D.P.I.; van der Ende-Metselaar, H.H.; van Gosliga, D.; Apperloo, L.; Carpaij, O.A.; van den Berge, M.; et al. Moxidectin and Ivermectin Inhibit SARS-CoV-2 Replication in Vero E6 Cells but Not in Human Primary Bronchial Epithelial Cells. Antimicrob. Agents Chemother. 2022, 66, e01543-21. [Google Scholar] [CrossRef] [PubMed]
  27. Lawrence, J.M.; Meyerowitz-Katz, G.; Heathers, J.A.J.; Brown, N.J.L.; Sheldrick, K.A. The lesson of ivermectin: Meta-analyses based on summary data alone are inherently unreliable. Nat. Med. 2021, 27, 1853–1854. [Google Scholar] [CrossRef] [PubMed]
  28. Vallejos, J.; Zoni, R.; Bangher, M.; Villamandos, S.; Bobadilla, A.; Plano, F.; Campias, C.; Campias, E.C.; Medina, M.F.; Achinelli, F.; et al. Ivermectin to prevent hospitalizations in patients with COVID-19 (IVERCOR-COVID19) a randomized, double-blind, placebo-controlled trial. BMC Infect. Dis. 2021, 21, 635. [Google Scholar] [CrossRef] [PubMed]
  29. Lim, S.C.L.; Hor, C.P.; Tay, K.H.; Mat Jelani, A.; Tan, W.H.; Ker, H.B.; Chow, T.S.; Zaid, M.; Cheah, W.K.; Lim, H.H.; et al. Efficacy of Ivermectin Treatment on Disease Progression Among Adults with Mild to Moderate COVID-19 and Comorbidities: The I-TECH Randomized Clinical Trial. JAMA Intern. Med. 2022. Available online: file:///C:/Users/MDPI/Downloads/jamainternal_lim_2022_oi_220006_1644957301.61931.pdf (accessed on 22 February 2022).
  30. López-Medina, E.; López, P.; Hurtado, I.C.; Dávalos, D.M.; Ramirez, O.; Martínez, E.; Díazgranados, J.A.; Oñate, J.M.; Chavarriaga, H.; Herrera, S.; et al. Effect of Ivermectin on Time to Resolution of Symptoms among Adults with Mild COVID-19: A Randomized Clinical Trial. JAMA J. Am. Med. Assoc. 2021, 325, 1426–1435. [Google Scholar] [CrossRef]
Figure 1. Antiviral efficacy of ivermectin as a single or combination therapy against SARS-CoV-2 beta (B.1.351) variant in a hamster infection model. (a) Setup of the study. (b) Viral RNA levels in the lungs of control (vehicle-treated), ivermectin-treated (0.4 mg/kg, QD), EIDD-2801-treated (150 mg/kg, BID), and combination-treated (EIDD-2801+ivermectin at 150 mg/kg BID+0.4 mg/kg QD) SARS-CoV-2 (B.1.351)-infected hamsters at day 4 post-infection (pi) are expressed as log10 SARS-CoV-2 RNA copies per milligram lung tissue. Individual data and median values are presented. (c) Infectious viral loads in the lungs of control (vehicle-treated), ivermectin-treated, EIDD-2801-treated and combination-treated (EIDD-2801+ivermectin) SARS-CoV-2-infected hamsters at day 4 pi are expressed as log10 TCID50 per milligram lung tissue. Individual data and median values are presented. (d) Weight change at day 4 pi in percentage, normalized to the body weight at the time of infection. Bars represent means ± SD. Data were analyzed with the Mann−Whitney U test. * p < 0.05, ** p < 0.01, *** p < 0.001, ns = non-significant. EIDD=EIDD-2801. All data (panels b, c, d) are from two independent experiments with n = 14 for vehicle group and n = 10 for the other groups.
Figure 1. Antiviral efficacy of ivermectin as a single or combination therapy against SARS-CoV-2 beta (B.1.351) variant in a hamster infection model. (a) Setup of the study. (b) Viral RNA levels in the lungs of control (vehicle-treated), ivermectin-treated (0.4 mg/kg, QD), EIDD-2801-treated (150 mg/kg, BID), and combination-treated (EIDD-2801+ivermectin at 150 mg/kg BID+0.4 mg/kg QD) SARS-CoV-2 (B.1.351)-infected hamsters at day 4 post-infection (pi) are expressed as log10 SARS-CoV-2 RNA copies per milligram lung tissue. Individual data and median values are presented. (c) Infectious viral loads in the lungs of control (vehicle-treated), ivermectin-treated, EIDD-2801-treated and combination-treated (EIDD-2801+ivermectin) SARS-CoV-2-infected hamsters at day 4 pi are expressed as log10 TCID50 per milligram lung tissue. Individual data and median values are presented. (d) Weight change at day 4 pi in percentage, normalized to the body weight at the time of infection. Bars represent means ± SD. Data were analyzed with the Mann−Whitney U test. * p < 0.05, ** p < 0.01, *** p < 0.001, ns = non-significant. EIDD=EIDD-2801. All data (panels b, c, d) are from two independent experiments with n = 14 for vehicle group and n = 10 for the other groups.
Microorganisms 10 00633 g001
Figure 2. Lung histopathology of SARS-CoV-2-infected Syrian hamsters following treatment with ivermectin as a single or combination therapy. (a) Cumulative severity score from H&E stained slides of lungs from control (vehicle-treated ivermectin-treated (0.4 mg/kg, QD), EIDD-2801-treated (150 mg/kg, BID) and combination-treated (EIDD-2801+ivermectin at 150 mg/kg BID+0.4 mg/kg QD) SARS-CoV-2 (B.1.351)-infected hamsters at day 4 post-infection (pi). Individual data and median values are presented, and the dotted line represents the median score of untreated non-infected hamsters. Data were analyzed with the Mann−Whitney U test. * p < 0.05, ns = non-significant. Data are from two independent experiments with n = 14 for vehicle group and n = 10 for the other groups. EIDD = EIDD-2801. (b) Representative H&E images of lungs of control (vehicle-treated), ivermectin-treated and EIDD-2801-treated SARS-CoV-2 (B.1.351)-infected hamsters at day 4 post-infection (pi). Lung from vehicle control group showed peribronchial inflammation with intrabronchial cell debris (blue arrows), perivascular inflammation with endothelialitis (red arrows), and bronchopneumonia (green arrows) surrounded by intra-alveolar hemorrhage. Image of the lung from the ivermectin-treated animal showed severe perivascular inflammation with vasculitis (red arrows), limited and focal peri-bronchial inflammation (blue arrows), and significant and multifocal bronchopneumonia (green arrows). On the other hand, the lung from the molnupiravir-treated animal (EIDD-2801) revealed very limited peribronchial (blue arrows) and peri-vascular (red arrows) inflammation. Scale bar 100 µM.
Figure 2. Lung histopathology of SARS-CoV-2-infected Syrian hamsters following treatment with ivermectin as a single or combination therapy. (a) Cumulative severity score from H&E stained slides of lungs from control (vehicle-treated ivermectin-treated (0.4 mg/kg, QD), EIDD-2801-treated (150 mg/kg, BID) and combination-treated (EIDD-2801+ivermectin at 150 mg/kg BID+0.4 mg/kg QD) SARS-CoV-2 (B.1.351)-infected hamsters at day 4 post-infection (pi). Individual data and median values are presented, and the dotted line represents the median score of untreated non-infected hamsters. Data were analyzed with the Mann−Whitney U test. * p < 0.05, ns = non-significant. Data are from two independent experiments with n = 14 for vehicle group and n = 10 for the other groups. EIDD = EIDD-2801. (b) Representative H&E images of lungs of control (vehicle-treated), ivermectin-treated and EIDD-2801-treated SARS-CoV-2 (B.1.351)-infected hamsters at day 4 post-infection (pi). Lung from vehicle control group showed peribronchial inflammation with intrabronchial cell debris (blue arrows), perivascular inflammation with endothelialitis (red arrows), and bronchopneumonia (green arrows) surrounded by intra-alveolar hemorrhage. Image of the lung from the ivermectin-treated animal showed severe perivascular inflammation with vasculitis (red arrows), limited and focal peri-bronchial inflammation (blue arrows), and significant and multifocal bronchopneumonia (green arrows). On the other hand, the lung from the molnupiravir-treated animal (EIDD-2801) revealed very limited peribronchial (blue arrows) and peri-vascular (red arrows) inflammation. Scale bar 100 µM.
Microorganisms 10 00633 g002
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Foo, C.S.; Abdelnabi, R.; Vangeel, L.; De Jonghe, S.; Jochmans, D.; Weynand, B.; Neyts, J. Ivermectin Does Not Protect against SARS-CoV-2 Infection in the Syrian Hamster Model. Microorganisms 2022, 10, 633. https://doi.org/10.3390/microorganisms10030633

AMA Style

Foo CS, Abdelnabi R, Vangeel L, De Jonghe S, Jochmans D, Weynand B, Neyts J. Ivermectin Does Not Protect against SARS-CoV-2 Infection in the Syrian Hamster Model. Microorganisms. 2022; 10(3):633. https://doi.org/10.3390/microorganisms10030633

Chicago/Turabian Style

Foo, Caroline S., Rana Abdelnabi, Laura Vangeel, Steven De Jonghe, Dirk Jochmans, Birgit Weynand, and Johan Neyts. 2022. "Ivermectin Does Not Protect against SARS-CoV-2 Infection in the Syrian Hamster Model" Microorganisms 10, no. 3: 633. https://doi.org/10.3390/microorganisms10030633

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