Next Article in Journal
Forward Left Ventricular Ejection Fraction as a Predictor of Postoperative Left Ventricular Dysfunction in Patients with Degenerative Mitral Regurgitation
Next Article in Special Issue
Consequences of the COVID-19 Outbreak Lockdown on Non-Viral Infectious Agents as Reported by a Laboratory-Based Surveillance System at the IHU Méditerranée Infection, Marseille, France
Previous Article in Journal
Sleep Disorders in Adults with Down Syndrome
Previous Article in Special Issue
Hydroxychloroquine and Azithromycin Treatment of Hospitalized Patients Infected with SARS-CoV-2 in Senegal from March to October 2020
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

In Vitro Evaluation of the Antiviral Activity of Methylene Blue Alone or in Combination against SARS-CoV-2

1
Unité Parasitologie et Entomologie, Département Microbiologie et Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, 13005 Marseille, France
2
Aix Marseille Univ, IRD, SSA, AP-HM, VITROME, 13005 Marseille, France
3
IHU Méditerranée Infection, 13005 Marseille, France
4
Aix Marseille Univ, IRD, AP-HM, MEPHI, 13005 Marseille, France
5
Centre National de Référence du Paludisme, 13005 Marseille, France
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2021, 10(14), 3007; https://doi.org/10.3390/jcm10143007
Submission received: 26 April 2021 / Revised: 1 July 2021 / Accepted: 2 July 2021 / Published: 6 July 2021

Abstract

:
A new severe acute respiratory syndrome coronavirus (SARS-CoV-2) causing coronavirus diseases 2019 (COVID-19), which emerged in Wuhan, China in December 2019, has spread worldwide. Currently, very few treatments are officially recommended against SARS-CoV-2. Identifying effective, low-cost antiviral drugs with limited side effects that are affordable immediately is urgently needed. Methylene blue, a synthesized thiazine dye, may be a potential antiviral drug. Antiviral activity of methylene blue used alone or in combination with several antimalarial drugs or remdesivir was assessed against infected Vero E6 cells infected with two clinically isolated SARS-CoV-2 strains (IHUMI-3 and IHUMI-6). Effects both on viral entry in the cell and on post-entry were also investigated. After 48 h post-infection, the viral replication was estimated by RT-PCR. The median effective concentration (EC50) and 90% effective concentration (EC90) of methylene blue against IHUMI-3 were 0.41 ± 0.34 µM and 1.85 ± 1.41 µM, respectively; 1.06 ± 0.46 µM and 5.68 ± 1.83 µM against IHUMI-6. Methylene blue interacted at both entry and post-entry stages of SARS-CoV-2 infection in Vero E6 cells as retrieved for hydroxychloroquine. The effects of methylene blue were additive with those of quinine, mefloquine and pyronaridine. The combinations of methylene blue with chloroquine, hydroxychloroquine, desethylamodiaquine, piperaquine, lumefantrine, ferroquine, dihydroartemisinin and remdesivir were antagonist. These results support the potential interest of methylene blue to treat COVID-19.

1. Introduction

In December 2019, a new coronavirus called severe acute respiratory syndrome coronavirus (SARS-CoV-2) responsible for coronavirus diseases 2019 (COVID-19) was first detected in Wuhan, China, before spreading all over the world [1]. SARS-CoV-2 belongs to the Betacoronaviruses with similarities with viruses detected in bats [2]. SARS-CoV-2 caused a wide range of symptoms from asymptomatic to fatal respiratory tract infections [2,3,4]. The common symptoms of COVID-19 are fever with dry cough, dyspnea, headaches, myalgia, intense fatigue with gastrointestinal symptoms including vomiting, abdominal pain, loss of appetite and diarrhea [2,3,4]. Less common symptoms including hyposmia, anosmia, ageusia, maculopapular rash or urticarious lesions are observed [4]. Severe cases are associated with uncontrolled increased lung inflammatory response called cytokine storm syndrome. An interleukin release of IL-6, IL-1, IL-2, IL-10, IL-12 and IL-18 associated with tumour necrosis factor alpha (TNF-α) and other inflammatory mediators (IP-10, MCP-1, MIP-1α) are associated with the severity of pulmonary inflammation and extensive lung damages, likely leading to death [3,5]. Currently, very few treatments are officially recommended against SARS-CoV-2. Evaluation of repurposing of existing approved drugs is an efficient, low-cost approach to identify therapeutic against SARS-CoV-2. Several compounds have been already evaluated at least in vitro, including antimalarial drugs (chloroquine, mefloquine, quinine, pyronaridine, piperaquine, lumefantrine, artemisinin) [6,7,8,9], antibiotics (azithromycin, doxycycline) [10,11], antiparasitic drugs (ivermectin) [12] or antiviral agents (remdesivir, ritonavir, lopinavir, favipiravir) [8,9,13,14].
Methylene blue, a synthesized thiazine dye, is able to inactivate viruses, including Zika, yellow fever, dengue, chikungunya, Ebola viruses and Middle East respiratory syndrome coronavirus in plasma when illuminated with visible light [15,16,17,18]. Methylene blue was also shown to exert in vitro and in vivo antimicrobial effects without photoactivation, and more particularly against Plasmodium spp. [19,20,21,22,23]. The repurposed methylene blue could be a potent candidate in the treatment of COVID-19 [24]. SARS-CoV-2 in plasma or in infected Vero E6 cells was inactivated by photoactivation [25,26,27]. Moreover, methylene blue was found to inhibit SARS-CoV-2 in vitro at concentrations achievable after oral or intravenous administration [25,28].
The aim of this study was to confirm the antiviral activity of methylene blue against SARS-CoV-2, to investigate its effects on viral entry in the cell and on post-entry and its activity in combination with other potential drugs.

2. Materials and Methods

2.1. Drugs, Virus and Cells

Methylene blue (methylthioninium chloride; Proveblue®) was provided by Provepharm SAS (Marseille, France). Hydroxychloroquine sulfate (Sigma Aldrich, St Quentin Fallavier, France) and remdesivir (Apollo Scientific, Manchester, UK) were used as comparators. Stock solutions of methylene blue and hydroxychloroquine were prepared in water and remdesivir in DMSO/water 10%. All the stock solutions were then diluted in Minimum Essential Media (MEM, Gibco, ThermoFischer, Waltham, MA, USA) in order to have 7 final concentrations ranging from 0.1 µM to 100 µM. Two clinically-isolated SARS-CoV-2 strains (IHUMI-3 and IHUMI-6), collected in hospitalized patients during the first COVID-19 outbreak in March 2020 in Marseille [29], were maintained in production in Vero E6 cells (American type culture collection ATCC® CRL-1586™) in MEM with 4% of fetal bovine serum and 1% of glutamine (complete medium). Vero E6 cells are one of the most used cells for the culture of SARS-CoV-2 due to the presence of high expression of angiotensin converting enzyme 2 (ACE2) receptors, essential for SARS-CoV-2 cellular entry [30,31]. Vero E6 cells were found to be relevant for antiviral drug screening models [31,32].

2.2. Antiviral Activity Assay

Briefly, 96-well plates were prepared with 5 × 105 cells/mL of Vero E6 (200 µL per well), as previously described [10]. The different concentrations of methylene blue without photoactivation, hydroxychloroquine or remdesivir were added 4 h before infection. The replication of IHUMI-3 or IHUMI-6 strains in Vero E6 cells at an MOI of 0.01 was estimated 48 h after infection by RT-PCR using the Superscript III platinum one step with Rox kit (Invitrogen) after extraction with the BioExtract SuperBall kit (Biosellal, Dardilly, France). The primers used were previously described [33]. EC50 (median effective concentration) and EC90 (90% effective concentration) were estimated through nonlinear regression by using the R software (ICEstimator version 1.2). EC50 and EC90 values resulted in the mean of 6 to 12 independent experimentations.

2.3. Determination of the Inhibition Stage

Effects of methylene blue, hydroxychloroquine or remdesivir on entry and post-entry of SARS-CoV-2 were evaluated at a concentration of 10 µM. For “full-time” treatment, Vero E6 cells were infected with the IHUMI-3 strain for 48 h after pre-incubation of the cells with one of the three drugs for 4 h. For “entry” treatment, the cells were infected for 2 h after pre-incubation for 4 h and then the virus–drug mixture was replaced with fresh medium maintained for 46 h. For “post-entry” treatment, the cells were infected for 2 h and then incubated with drug for 46 h. The percentage of inhibition of SARS-CoV-2 replication by 10 µM of drug was estimated for each drug concentration as following: (mean CTdrug concentration − mean CTcontrol 0%)/(mean CTcontrol 100% − mean CTcontrol 0%) × 100. The result was the mean of 6 to 9 independent experiments.

2.4. Antiviral Activity of Drug Combinations

The antiviral activity of two concentrations of methylene blue (0.1 and 0.5 µM) was evaluated alone or in combination with four fixed concentrations of chloroquine (0.5, 1, 5 and 10 µM), hydroxychloroquine (0.5, 1, 5 and 10 µM), quinine (1, 5, 10 and 25 µM), mefloquine (0.5, 1, 5 and 10 µM), pyronaridine (0.1, 0.5, 1 and 5 µM), ferroquine (0.5, 1, 5 and 10 µM), desethylamodiaquine (0.1, 0.5, 1 and 10 µM), lumefantrine (5, 10, 25 and 50 µM), piperaquine (5, 10, 25 and 50 µM), dihydroartemisinin (5, 10, 25 and 50 µM) and remdesivir (0.05, 0.1, 0.5 and 1 µM) against the SARS-CoV-2 IHUMI-3 strain for 48 h. The percentage of inhibition of SARS-CoV-2 replication by methylene blue alone or in combination was estimated for each drug association as following: (mean CTdrug association − mean CTcontrol 0%)/(mean CTcontrol 100% − mean CTcontrol 0%) × 100. The result was the mean of 9 to 13 independent experiments.

3. Results

The antiviral activity of methylene blue against the clinically-isolated SARS-CoV-2 strains IHUMI-3 and IHUMI-6 was concentration-dependent (Figure 1).
The median effective concentration (EC50) and 90% effective concentration (EC90) of methylene blue against IHUMI-3 were 0.41 ± 0.34 µM and 1.85 ± 1.41 µM (n = 12), respectively; 1.06 ± 0.46 µM and 5.68 ± 1.83 µM against IHUMI-6 (n = 6). The difference between EC50 against the two was significant (p = 0.015, Welch two sample t-test).
In comparison, EC50 and EC90 of remdesivir against IHUMI-6 were 1.00 ± 0.41 µM and 3.2 ± 2.9 µM, respectively (n = 6). There was no significant difference between methylene blue and remdesivir EC50 or EC90 (p = 0.786 and p = 0.113, Welch two sample t-test).
EC50 and EC90 of hydroxychloroquine against IHUMI-6 were 6.25 ± 2.20 µM and 12.32 ± 2.82 µM, respectively (n = 6). Methylene blue was significantly more effective than hydroxychloroquine against IHUMI-6 (p = 0.005 for EC50 and p = 0.003 for EC90; Welch two sample t-test).
Methylene blue interacted at both entry and post-entry stages of SARS-CoV-2 infection in Vero E6 cells, as hydroxychloroquine did (Figure 2). Contrariwise, remdesivir, which is an antiviral drug, interacted only at post-entry stage.
The effects of methylene blue were additive with those of quinine (Figure 3), mefloquine (Figure 4) and pyronaridine (Figure 5).
The combinations of methylene blue with chloroquine (Figure 6), hydroxychloroquine, desethylamodiaquine, piperaquine, lumefantrine, ferroquine, dihydroartemisinin and remdesivir (data not shown) were antagonist.

4. Discussion

Our data confirmed the in vitro activity of methylene blue at very low-micromolar range with EC50 between 0.41 and 1.06 µM and EC90 between 1.85 ± 1.41 µM and 5.68 ± 1.83 µM against two strains of SARS-CoV-2 IHUMI-3 and IHUMI-6 [28,34,35]. The reduction in the viral replication is not due to methylene blue toxicity against Vero E6 cells. The 50% cytotoxic concentration (CC50) was previously evaluated (CC50 > 100 µM) [28]. According to this previous CC50, the selectivity index (SI) for methylene blue was above 100. Methylene blue was effective as antiviral remdesivir against IHUMI-6 strain and more effective than hydroxychloroquine in vitro. These effective concentrations are compatible with blood concentrations after usual oral intake or intravenous injection of methylene blue [36,37,38]. An oral uptake of 325 mg of methylene blue led to a Cmax (maximum blood concentration) value of 0.97 µg/mL (around 3 µM) [36] and a dose of 2 mg/kg intravenous showed a Cmax of 2.917 µg/mL (around 10 µM) [37]. In another study, blood concentrations of 6–7 µM were obtained after three oral daily doses of 69 mg (207 mg/day) [38]. Methylene blue EC50 and EC90 are coherent with human blood concentrations after usual uptake of methylene blue. Moreover, methylene blue is accumulated in lungs tissue. The absorption of methylene blue in lungs was around 3 to 5% of injected drug per g of tissue after a single intravenous injection of methylene blue in mice [39].
Methylene blue could be associated with antimalarial drugs such as quinine, mefloquine or pyronaridine to improve its antiviral activity. Mefloquine concentrations are 10 times higher in the lung than in the blood (a concentration which can go up to 180 mg/kg in the lung) [40]. A single oral dose of 2 mg (10 mg/kg) of pyronaridine in rats led to a blood Cmax of 223 ng/mL and a lung Cmax of 36.4 µg/g of tissue (165 more concentrated) [41]. In rat, after intravenous dose of 10 mg/kg of quinine, the observed concentration lung/blood ratio was at 246 [42]. These three drugs accumulate in lungs and could be potent partners for methylene blue for COVD-19 treatment.
Methylene blue interacted at both entry and post-entry stages of SARS-CoV-2 infection in Vero E6 cells. The inhibition of the viral entry is consistent with the results interaction between the spike protein (S) and the angiotensin converting enzyme 2 (ACE2) via its receptor binding domain (RBD), binding required for SARS-CoV-2 cell entry. Methylene blue inhibits the binding of SARS-CoV-2 spike S protein to ACE2 at micromolar range [35]. Moreover, the inhibition of both entry and viral replication after SARS-CoV-2 entry is coherent with 3D modelling approaches. Docking analysis showed that methylene blue could bind both the spike protein S of SARS-CoV-2, but lesser than hydroxychloroquine, and the main protease (M or Mpro), but lesser than remdesivir [43]. The main protein, also called 3C-like protease, is crucial in SARS-CoV-2 replication by leading to the formation of non-structural proteins (NSPs) [44]. SARS-CoV-2 needs the transmembrane protease serine 2 (TMPRSS2) for activating the spike S protein [5]. The spike S protein, TMPRSS2 and Mpro are promising anti-SARS-CoV-2 targets for enzymatic inhibitors [5,45]. Moreover, SARS-CoV-2 3D comparative modelling analyses lead to predict interactions with spike protein S and human ACE2 and to design neutralizing antibodies for blocking this binding as a new therapeutic strategy [46,47].
Besides its antiviral activity, methylene blue is reduced into leukomethylene blue which reduces the methemoglobin to hemoglobin. Methylene blue could reduce hypoxia, one of the main complications in COVID-19 patients, by decreasing methemoglobin. Moreover, methylene blue decreases inflammation and oxidative stress [48,49]. Pro-inflammatory cytokines and nitric oxide were considerably increased in the cytokine storm due to COVID-19 [50].
These results support additional in vivo studies in animal experimental models to confirm methylene blue anti-SARS-CoV-2 activity. The probable use of methylene blue to treat COVID-19 needs to be established by prospective comparative clinical studies. Methylene blue has been assessed in combination with vitamin C and N-acetyl cysteine in severe COVID-19 [51,52]. The addition of methylene blue to standard of care treatment significantly improved respiratory distress, hospital stay and mortality rate in severe patients with confirmed COVID-19 [53].

5. Conclusions

Methylene blue, an FDA-approved drug for methemoglobinemia treatment, showed potent in vitro anti-SARS-CoV-2 at micromolar range and potentiation in combination with antimalarial drugs, including quinine, mefloquine or pyronaridine. Methylene blue acted at both entry and post-entry (replication) of SARS-CoV-2 in Vero E6 cells. Methylene blue needs additional in vivo evaluation in animal models and then in human to confirm its antiviral effects.

Author Contributions

Conceptualization, M.G., B.L.S. and B.P.; validation, M.G., S.H. and B.P.; formal analysis, B.P.; investigation, M.G., O.D., M.B., J.M. and I.F.; resources, P.J., J.A., I.D., M.L.B. and S.H.; writing—original draft preparation, M.G. and B.P.; writing—review and editing, J.A., S.H. and B.L.S.; supervision, B.P.; project administration, B.P.; funding acquisition, B.L.S. and B.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the National Research Agency, program “Investissement d’avenir”grant number ANR-10-IAHU-03” and Institut Hospitalo-Universitaire (IHU) Méditerranée Infection grant number COVID-19. Manon Boxberger received a PhD grant supported by L’Occitane Society.

Data Availability Statement

The data presented in this study are available on request from the corresponding author.

Acknowledgments

The authors thank Provepharm for providing the methylene blue (Proveblue®).

Conflicts of Interest

The authors declare no conflict of interest. The funders or Provepharm 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.

Disclaimer

The findings and conclusion in this report are those of the authors and do not represent the views of the Ministère des Armées and Ministère de l’Enseignement supérieur, de la Recherche et de l’Innovation.

References

  1. Wu, F.; Zhao, S.; Yu, B.; Chen, Y.M.; Wang, W.; Song, Z.G.; Hu, Y.; Tao, Z.W.; Tian, J.H.; Pei, Y.Y.; et al. A new coronavirus associated with human respiratory disease in China. Nature 2020, 579, 365–369. [Google Scholar] [CrossRef] [Green Version]
  2. Santacroce, L.; Charitos, I.A.; Carreta, D.M.; de Nitto, E.; Lovero, R. The human coronaviruses (HCoVs) and the molecular mechanisms. J. Mol. Med. 2021, 99, 93–106. [Google Scholar] [CrossRef]
  3. Bakhiet, M.; Taurin, S. SARS-CoV-2: Targeted managements and vaccine development. Cytokine Growth Factor Rev. 2021, 58, 16–29. [Google Scholar] [CrossRef] [PubMed]
  4. Charitos, I.A.; Ballini, A.; Bottalico, L.; Cantore, S.; Passarelli, P.C.; Inchingolo, F.; D’Addona, A.; Santacroce, L. Special features of SARS-CoV-2 in daily practice. World J. Clin. Cases 2020, 8, 3920–3933. [Google Scholar] [CrossRef] [PubMed]
  5. Inchigolo, A.D.; Inchigolo, A.M.; Bordea, I.R.; Malcangi, G.; Xhajanka, E.; Scarano, A.; Lorusso, F.; Farronato, M.; Tartaglia, G.M.; Isacco, C.G.; et al. SARS-CoV-2 disease adjuvant therapies and supplements breakthrough for the infection prevention. Microorganisms 2021, 9, 525. [Google Scholar] [CrossRef] [PubMed]
  6. Gendrot, M.; Andreani, J.; Boxberger, M.; Jardot, P.; Fonta, I.; Le Bideau, M.; Duflot, I.; Mosnier, J.; Rolland, C.; Bogreau, H.; et al. Antimalarial drugs inhibit the replication of SARS-CoV-2: An in vitro evaluation. Trav. Med. Infect. Dis. 2020, 37, 101873. [Google Scholar] [CrossRef] [PubMed]
  7. Gendrot, M.; Duflot, I.; Boxberger, M.; Delandre, O.; Jardot, P.; Le Bideau, M.; Andreani, J.; Fonta, I.; Mosnier, J.; Rolland, C.; et al. Antimalarial artemisinin-based combination therapies (ACT) and COVID-19 in Africa: In vitro inhibition of SARS-CoV-2 replication by mefloquine-artesunate. Int. J. Infect. Dis. 2020, 99, 437–440. [Google Scholar] [CrossRef] [PubMed]
  8. Wang, M.; Cao, R.; Zhang, L.; Yang, X.; Liu, J.; Xu, M.; Shi, Z.; Hu, Z.; Zhong, W.; Xiao, G. Remdesivir and chloroquine effectively inhibit the recently emerged novel coronavirus (2019-nCov) in vitro. Cell Res. 2020, 30, 269–271. [Google Scholar] [CrossRef]
  9. Holwerda, M.; V’kovski, P.; Wider, M.; Thiel, V.; Djikman, R. Identification of an antiviral compound from the pandemic response box that efficiently inhibits SARS-CoV-2 infection in vitro. Microorganisms 2020, 8, 1872. [Google Scholar] [CrossRef]
  10. Andreani, J.; Le Bideau, M.; Duflot, I.; Jardot, P.; Rolland, C.; Boxberger, M.; Wurtz, N.; Rolain, J.M.; Colson, P.; La Scola, B.; et al. In vitro testing of hydroxychloroquine and azithromycin on SARS-CoV-2 shows synergistic effect. Microb. Pathog. 2020, 145, 104228. [Google Scholar] [CrossRef]
  11. Gendrot, M.; Andreani, J.; Jardot, P.; Hutter, S.; Delandre, O.; Boxberger, M.; Mosnier, J.; Le Bideau, M.; Duflot, I.; Fonta, I.; et al. In vitro antiviral activity of doxycycline against SARS-CoV-2. Molecules 2020, 25, 5064. [Google Scholar] [CrossRef] [PubMed]
  12. 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]
  13. Choy, K.T.; Wong, A.Y.L.; Kaewpreedee, P.; Sia, S.F.; Chen, D.; Hui, K.P.Y.; Chu, D.K.W.; Chan, M.C.W.; Cheung, P.P.H.; Huang, X.; et al. Remdesivir, lopinavir, emetine, and homoharringtonine inhibit SARS-CoV-2 replication in vitro. Antivir. Res. 2020, 178, 104786. [Google Scholar] [CrossRef]
  14. Zhang, L.; Liu, J.; Cao, R.; Xu, M.; Wu, Y.; Shang, W.; Wang, X.; Zhang, H.; Jiang, X.; Sun, Y.; et al. Comparative antiviral efficacy of viral protease inhibitors against the novel SARS-CoV-2 in vitro. Virol. Sin. 2020, 35, 776–784. [Google Scholar] [CrossRef]
  15. Wang, Y.; Ren, K.; Liao, X.; Luo, G.; Kumthip, K.; Leetrakool, N.; Li, S.; Chen, L.; Yang, C.; Chen, Y. Inactivation of Zika virus in plasma and derivatives by four different methods. J. Med. Virol. 2019, 91, 2059–2065. [Google Scholar] [CrossRef] [PubMed]
  16. Faddy, H.M.; Fryk, J.J.; Hal, R.A.; Young, P.R.; Reichenberg, S.; Tolksdorf, F.; Sumian, C.; Gravemann, U.; Seltsam, A.; Marks, D.C. Inactivation of yellow fever virus in plasma after treatment with methylene blue and visible light and in platelet concentrates following treatment with ultraviolet C light. Transfusion 2019, 59, 2223–2227. [Google Scholar] [CrossRef] [PubMed]
  17. Fryk, J.J.; Marks, D.C.; Hobson-Peters, J.; Prow, N.A.; Watterson, D.; Hall, R.A.; Young, P.R.; Reichenberg, S.; Sumian, C.; Faddy, H.M. Dengue and chikungunya viruses in plasma are effectively inactivated after treatment with methylene blue and visible light. Transfusion 2016, 56, 2278–2285. [Google Scholar] [CrossRef]
  18. Eickmann, M.; Gravemann, U.; Handke, W.; Tolksdorf, F.; Reichenberg, S.; Müller, T.H.; Seltsam, A. Inactivation of Ebola virus and Middle East respiratory syndrome coronavirus in platelet concentrates and plasma by ultraviolet C light and methylene blue plus visible light, respectively. Transfusion 2018, 58, 2202–2207. [Google Scholar] [CrossRef] [Green Version]
  19. Pascual, A.; Henry, M.; Briolant, S.; Charras, S.; Baret, E.; Amalvict, R.; Huyghues des Etages, E.; Feraud, M.; Rogier, C.; Pradines, B. In vitro activity of Proveblue (methylene blue) on Plasmodium falciparum strains resistant to standard antimalarial drugs. Antimicrob. Agents Chemother. 2011, 55, 2472–2474. [Google Scholar] [CrossRef] [Green Version]
  20. Fall, B.; Madamet, M.; Diawara, S.; Briolant, S.; Wade, K.A.; Lo, G.; Nakoulima, A.; Fall, M.; Bercion, R.; Kounta, M.B.; et al. Ex vivo activity of Proveblue, a methylene blue, against filed isolates of Plasmodium falciparum in Dakar, Senegal from 2013 to 2015. Int. J. Antimicrob. Agents 2017, 50, 155–158. [Google Scholar] [CrossRef]
  21. Gendrot, M.; Madamet, M.; Mosnier, J.; Fonta, I.; Amalvict, R.; Benoit, N.; Briolant, S.; Pradines, B. Baseline and multinormal distribution of Ex vivo susceptibilities of Plasmodium falciparum to methylene blue in Africa, 2013–2018. J. Antimicrob. Agents 2020, 75, 2141–2148. [Google Scholar]
  22. Dormoi, J.; Pradines, B. Dose responses of Proveblue methylene blue in an experimental murine cerebral malaria model. Antimicrob. Agents Chemother. 2013, 57, 4080–4081. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Dormoi, J.; Briolant, S.; Desgrouas, C.; Pradines, B. Efficacy of Proveblue (methylene blue) in an experimental cerebral malaria murine model. Antimicrob. Agents Chemother. 2013, 57, 3412–3414. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Ghahestani, S.M.; Shahab, E.; Karimi, S.; Madani, M.H. Methylene blue may have a role in the treatment of COVID-19. Med. Hypothesis 2020, 144, 110163. [Google Scholar] [CrossRef] [PubMed]
  25. Svyatchenko, V.A.; Nikonov, S.D.; Mayorov, A.P.; Gelfond, M.L.; Loktev, V.B. Antiviral photodynamic therapy: Inactivation and inhibition of SARS-CoV-2 in vitro using methylene blue and radachlorin. Photodagnosis Photodyn. Ther. 2021, 33, 102112. [Google Scholar] [CrossRef]
  26. Almeida, A.; Faustino, M.A.F.; Neves, M.G.P.M.S. Antimicrobial photodynamic therapy in the control of COVID-19. Antibiotics 2020, 9, 320. [Google Scholar] [CrossRef] [PubMed]
  27. Jin, C.; Yu, B.; Zhang, J.; Wu, H.; Zhou, X.; Yao, H.; Liu, F.; Lu, X.; Cheng, L.; Jiang, M.; et al. Methylene blue photochemical treatment as a reliable SARS-CoV-2 plasma virus inactivation method for blood safety and convalescent plasma therapy for COVID-19. BMC Infect. Dis. 2021, 21, 357. [Google Scholar] [CrossRef]
  28. Gendrot, M.; Andreani, J.; Duflot, I.; Boxberger, M.; Le Bideau, M.; Mosnier, J.; Jardot, P.; Fonta, I.; Rolland, C.; Bogreau, H.; et al. Methylene blue inhibits replication of SARS-CoV-2 in vitro. Int. J. Antimicrob. Agents 2020, 56, 106202. [Google Scholar] [CrossRef]
  29. Gautret, P.; Lagier, J.C.; Parola, P.; Hoang, V.T.; Meddeb, L.; Mailhe, M.; Doudier, B.; Courjon, J.; Giordanengo, V.; Vieira, V.E.; et al. Hydroxychloroquine and azithromycin as a treatment of COVID-19: Results of an open-label non-randomized clinical trial. Int. J. Antimicrob. Agents 2020, 56, 105949. [Google Scholar] [CrossRef]
  30. Wurtz, N.; Penant, G.; Jardot, P.; Duclos, N.; La Scola, B. Culture of SARS-CoV-2 in a panel of laboratory cell lines, permissivity, and differences in growth profile. Eur. J. Clin. Microbiol. Infect. Dis. 2021, 40, 477–484. [Google Scholar] [CrossRef]
  31. Kumar, S.; Sarma, P.; Kaur, H.; Prajapat, M.; Bhattacharyya, A.; Avti, P.; Sehkhar, N.; Kaur, H.; Bansal, S.; Mahendiratta, S.; et al. Clinically relevant cell culture models and their significance in isolation, pathogenesis, vaccine development, repurposing and screening of nex drugs for SARS-CoV-2: A systematic review. Tissue Cell 2021, 70, 101497. [Google Scholar] [CrossRef]
  32. Dittmar, M.; Lee, J.S.; Whig, K.; Segrist, E.; Li, M.; Kamalia, B.; Castellana, L.; Ayyanathan, K.; Cardenas-Diaz, F.L.; Morrisey, E.E.; et al. Drug repurposing screens reveal cell-type-specific entry patways and FDA-apporved drugs active against SARS-Cov-2. Cell Rep. 2021, 35, 108959. [Google Scholar] [CrossRef]
  33. Amrane, S.; Tissot-Dupont, H.; Doudier, B.; Eldin, C.; Hocquart, M.; Mailhe, M.; Dudouet, P.; Ormières, E.; Ailhaud, L.; Parila, P.; et al. Rapid viral diagnosis and ambulatory management of suspected COVID-19 cases presenting at the infectious disease referral hospital in Marseille, France, January 31st to March 1st, 2020: A respiratory virus snapshot. Travel Med. Infect. Dis. 2020, 36, 101632. [Google Scholar] [CrossRef] [PubMed]
  34. Cagno, V.; Medaglia, C.; Cerny, A.; Cerny, T.; Tapparel, C.; Cerny, E. Methylene blue has a potent antiviral activity against SARS-CoV-2 in the absence of UV-activation in vitro. BioRxiv 2020. [Google Scholar] [CrossRef]
  35. Bojadzic, D.; Alcazar, O.; Buchwald, P. Methylene blue inhibits the SARS-CoV-2 Spike-ACE2 protein-protein interaction—A mechanism that can contribute to its antiviral activity against COVID-19. Front. Pharmacol. 2021, 11, 600372. [Google Scholar] [CrossRef] [PubMed]
  36. Anh, C.X.; Chavchich, M.; Birrell, G.W.; van Breda, K.; Travers, T.; Rowcliffe, K.; Lord, A.R.; Shanks, G.D.; Edstein, M.D. Pharmacokinetics and Ex vivo antimalarial activity of artesunate-amodiaquine plus methylene blue in healthy volunteers. Antimicrob. Agents Chemother. 2020, 64, e01441-19. [Google Scholar] [CrossRef] [PubMed]
  37. Center for Drug Evaluation and Research. Clinical Pharmacology and Biopharmaceutics Review(s): Application Number 204630Orig1s000. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/nda/2016/204630Orig1s000ClinPharmR.pdf (accessed on 5 July 2021).
  38. Baddeley, T.C.; McCaffrey, J.; Storey, J.M.; Cheung, J.K.S.; Melis, V.; Horsley, D.; Harrington, C.R.; Wischik, C.M. Complex disposition of methylthioninium redox forms determines efficacy in tau aggregation inhibitor therapy for Alzheimer’s disease. J. Pharmacol. Exp. Ther. 2015, 352, 110–118. [Google Scholar] [CrossRef] [PubMed]
  39. Link, E.M.; Costa, D.C.; Lui, D.; Ell, P.J.; Blower, P.J.; Spittle, M.F. Targeting disseminated melanoma with radiolabelled methylene blue. Acta Oncolog. 1996, 35, 331–341. [Google Scholar] [CrossRef]
  40. Jones, R.; Kunsman, G.; Levine, B.; Smith, M.; Stahl, C. Mefloquine distribution in postmortem cases. Forensic Sci. Int. 1994, 68, 29–32. [Google Scholar] [CrossRef]
  41. Park, S.H.; Pradeep, K. Absorption, distribution, excretion, and pharmacokinetics of 14C-pyronaridine tetraphosphate in male and female Sprague-Dawley rats. J. Biomed. Biotech. 2010, 2010, 590707. [Google Scholar] [CrossRef] [Green Version]
  42. Minchin, R.F.; Ilett, K.F. Comparative uptake of quinine and quinidine in rat lung. J. Pharm. Pharmacol. 1981, 33, 464–466. [Google Scholar] [CrossRef]
  43. Schadeva, C.; Wadhwa, A.; Kumari, A.; Hussain, F.; Jha, P.; Kaushik, N.K. In silico potential of approved antimalarial drugs for repurposing against COVID-19. OMICS 2020, 24, 568–580. [Google Scholar] [CrossRef] [PubMed]
  44. Sencanski, M.; Perovic, V.; Pajovic, S.B.; Adzic, M.; Paessler, S.; Glisic, S. Drug repurposing for candidate SARS-CoV-2 main protease inhibitors by a novel in silico methods. Molecules 2020, 25, 3830. [Google Scholar] [CrossRef]
  45. Melo de Oliveira Campos, D.; Fulco, U.L.; Silva de Oliveira, C.B.; Oliveira, J.I.N. SARS-CoV-2 virus infection: Targets and antiviral pharmacological strategies. J. Evid. Based Med. 2020, 13, 255–260. [Google Scholar] [CrossRef] [PubMed]
  46. Pierri, C.L. SARS-CoV-2 spike protein: Flexibility as a new target for fighting infection. Signal Transduc. Target Ther. 2020, 5, 254–256. [Google Scholar] [CrossRef] [PubMed]
  47. Mercurio, I.; Tragni, V.; Busto, F.; de Grassi, A.; Pierri, C.L. Protein structure analysis of the intrecactions between SARS-CoV-2 spike protein and the human ACE2 receptor: From conformational changes to novel neutralizing antibodies. Cell. Mol. Life Sci. 2021, 78, 1501–1522. [Google Scholar] [CrossRef]
  48. Kast, R.E. Inhibiting the NLRP3 inflammasome with methylene blue as treatment adjunct in myelodysplasia. Front. Oncol. 2018, 8, 280. [Google Scholar] [CrossRef] [Green Version]
  49. Wang, L.; Chen, B.; Lin, B.; Ye, Y.; Bao, C.; Zhao, X.; Jin, L.; Xiong, X. Methylene blue attenuates lung injury induced by hindlimb ischemia reperfusion in rats. Mediators Inflamm. 2018, 2018, 2508620. [Google Scholar] [CrossRef] [PubMed]
  50. Qin, C.; Zhou, L.; Hu, Z.; Zhang, S.; Yang, S.; Tao, Y.; Xie, C.; Ma, K.; Shang, K.; Wang, W.; et al. Dysregulation of immune response in patients with COVID-19 in Wuhan, China. Clin. Infect. Dis. 2020, 71, 762–768. [Google Scholar] [CrossRef] [PubMed]
  51. Alamdari, D.H.; Moghaddam, A.B.; Amini, S.; Alamdari, A.H.; Damsaz, M.; Yarahmadi, A. The application of a reduced dye used in orthopaedics as a novel treatment against coronavirus (COVID-19): A suggested therapeutic protocol. Arch. Bone Jt. Surg. 2020, 8, 291–294. [Google Scholar]
  52. Alamdari, D.H.; Moghaddam, A.B.; Amini, S.; Keramati, M.R.; Zarmehri, A.M.; Alamdari, A.H.; Damsaz, M.; Banpour, H.; Yarahmadi, A.; Koliakos, G. Application of methylene blue-vitamin C—N-acetyl cysteine for treatment of critically ill COVID-19 patients, report of a phase-I clinical trial. Eur. J. Pharmacol. 2020, 885, 173494. [Google Scholar] [CrossRef] [PubMed]
  53. Hamidi-Alamdari, D.; Hafizi-Lotfabadi, S.; Bagheri-Moghaddam, A.; Safari, H.; Mozdourian, M.; Javidarabshahi, Z.; Peivandi-Yazdi, A.; Ali-Zeraati, A.; Sedaghat, A.; Poursadegh, F.; et al. Methylene blue for treatment of hospitalized COVID-19 patients: A randomized, controlled, open-label clinical trial, phase 2. Rev. Investig. Clin. 2021, 73, 190–198. [Google Scholar]
Figure 1. Anti-SARS-CoV-2 activity of methylene blue in % of antiviral inhibition on IHUMI-3 (mean of 12 independent experiments) and IHUMI-6 (mean of 6 independent experiments) clinically-isolated strains (error bar represents standard deviation).
Figure 1. Anti-SARS-CoV-2 activity of methylene blue in % of antiviral inhibition on IHUMI-3 (mean of 12 independent experiments) and IHUMI-6 (mean of 6 independent experiments) clinically-isolated strains (error bar represents standard deviation).
Jcm 10 03007 g001
Figure 2. Antiviral activities of methylene blue, hydroxychloroquine and remdesivir at 10 µM against the SARS-CoV-2 IHUMI-3 strain in vitro. For “full-time” treatment, Vero E6 cells were infected with the IHUMI-3 strain for 48 h after pre-incubation of the cells with one of the three drugs for 4 h. For “entry” treatment, the cells were infected for 2 h after pre-incubation for 4 h and then the virus–drug mixture was replaced with fresh medium maintained for 46 h. For “post-entry” treatment, the cells were infected for 2 h and then incubated with drug for 46 h. Error bars represent standard deviation of 6 to 9 independent experiments.
Figure 2. Antiviral activities of methylene blue, hydroxychloroquine and remdesivir at 10 µM against the SARS-CoV-2 IHUMI-3 strain in vitro. For “full-time” treatment, Vero E6 cells were infected with the IHUMI-3 strain for 48 h after pre-incubation of the cells with one of the three drugs for 4 h. For “entry” treatment, the cells were infected for 2 h after pre-incubation for 4 h and then the virus–drug mixture was replaced with fresh medium maintained for 46 h. For “post-entry” treatment, the cells were infected for 2 h and then incubated with drug for 46 h. Error bars represent standard deviation of 6 to 9 independent experiments.
Jcm 10 03007 g002
Figure 3. Antiviral activities of methylene blue (MB) at 0.1 and 0.5 µM in combination with quinine (QN) at 1, 5, 10 and 25 µM (error bars represent standard deviation of 13 independent experiments).
Figure 3. Antiviral activities of methylene blue (MB) at 0.1 and 0.5 µM in combination with quinine (QN) at 1, 5, 10 and 25 µM (error bars represent standard deviation of 13 independent experiments).
Jcm 10 03007 g003
Figure 4. Antiviral activities of methylene blue (MB) at 0.1 and 0.5 µM in combination with mefloquine (MQ) at 0.5, 1, 5 and 10 µM (error bars represent standard deviation of 13 independent experiments).
Figure 4. Antiviral activities of methylene blue (MB) at 0.1 and 0.5 µM in combination with mefloquine (MQ) at 0.5, 1, 5 and 10 µM (error bars represent standard deviation of 13 independent experiments).
Jcm 10 03007 g004
Figure 5. Antiviral activities of methylene blue (MB) at 0.1 and 0.5 µM in combination with pyronaridine (PND) at 0.1, 0.5, 1 and 5 µM (error bars represent standard deviation of 9 independent experiments).
Figure 5. Antiviral activities of methylene blue (MB) at 0.1 and 0.5 µM in combination with pyronaridine (PND) at 0.1, 0.5, 1 and 5 µM (error bars represent standard deviation of 9 independent experiments).
Jcm 10 03007 g005
Figure 6. Antiviral activities of methylene blue (MB) at 0.1 and 0.5 µM in combination with chloroquine (CQ) at 0.5, 1, 5 and 10 µM (error bars represent standard deviation of 9 independent experiments).
Figure 6. Antiviral activities of methylene blue (MB) at 0.1 and 0.5 µM in combination with chloroquine (CQ) at 0.5, 1, 5 and 10 µM (error bars represent standard deviation of 9 independent experiments).
Jcm 10 03007 g006
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Gendrot, M.; Jardot, P.; Delandre, O.; Boxberger, M.; Andreani, J.; Duflot, I.; Le Bideau, M.; Mosnier, J.; Fonta, I.; Hutter, S.; et al. In Vitro Evaluation of the Antiviral Activity of Methylene Blue Alone or in Combination against SARS-CoV-2. J. Clin. Med. 2021, 10, 3007. https://doi.org/10.3390/jcm10143007

AMA Style

Gendrot M, Jardot P, Delandre O, Boxberger M, Andreani J, Duflot I, Le Bideau M, Mosnier J, Fonta I, Hutter S, et al. In Vitro Evaluation of the Antiviral Activity of Methylene Blue Alone or in Combination against SARS-CoV-2. Journal of Clinical Medicine. 2021; 10(14):3007. https://doi.org/10.3390/jcm10143007

Chicago/Turabian Style

Gendrot, Mathieu, Priscilla Jardot, Océane Delandre, Manon Boxberger, Julien Andreani, Isabelle Duflot, Marion Le Bideau, Joel Mosnier, Isabelle Fonta, Sébastien Hutter, and et al. 2021. "In Vitro Evaluation of the Antiviral Activity of Methylene Blue Alone or in Combination against SARS-CoV-2" Journal of Clinical Medicine 10, no. 14: 3007. https://doi.org/10.3390/jcm10143007

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