9
views
0
recommends
+1 Recommend
2 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      The anti-influenza virus drug, arbidol is an efficient inhibitor of SARS-CoV-2 in vitro

      letter

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Dear editor, Since December 2019, a novel disease COVID-19 caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) rapidly spread to over 200 countries and infected over 1.50 million people including 92,798 deaths (data as of April 10, 2020). On March 11, the World Health Organization (WHO) characterized COVID-19 as a pandemic, and called for accelerating diagnostics, vaccines, and drugs developments to combat this novel disease. Apart of the new coronavirus, influenza virus infections have been a consistent threat to the global public health over the years. In the United States alone, the Centers for Disease Control and Prevention (CDC) estimates that, so far during the 2019–2020 winter season, there have been at least 39 million illnesses, 400,000 hospitalizations and 24,000 deaths from influenza (https://www.cdc.gov/flu/weekly/index.htm). Considering the current concomitant circulation of SARS-CoV-2 and influenza virus infections, the exploration of available and viable anti-influenza drugs to treat both diseases is of great interest. Actually, in the early stages of the outbreak of COVID-19, some anti-flu drugs (for example, oseltamivir) have been applied for the treatment of COVID-19 patients 1,2 . Previously, we reported that favipiravir (T705), an anti-influenza drug approved in Japan and China, showed a certain efficacy against SARS-CoV-2 in vitro 3 . In addition, arbidol, an anti-influenza drug targeting the viral hemagglutinin (HA) is being used in a clinical trial against COVID-19 (ChiCTR2000029573) and has been recently added to the Guidelines for the Diagnosis and Treatment of COVID-19 (sixth and seventh editions) in China. A recent retrospective study suggested that arbidol treatment showed tendency to improve the discharging rate and decrease the mortality rate of COVID-19 patients 4 . However, to our knowledge, there has been no systematical analysis about the efficacy of anti-influenza drugs against SARS-CoV-2. In this study, we evaluated six currently available and licensed anti-influenza drugs against SARS-CoV-2. The drugs include arbidol, baloxavir, laninamivir, oseltamivir, peramivir, and zanamivir 5,6 . The M2 inhibitors (amantadine and rimantadine) were not considered in this study since they were not recommended for treating influenza by WHO due to drug resistance. First, the cytotoxicity of the compounds in African green monkey kidney cells, Vero E6 (ATCC-1586) was measured by a standard cell counting kit-8 (CCK8) assay. Then, the cells were infected with SARS-CoV-2 at a multiplicity of infection (MOI) of 0.05 in the presence of either compound or dimethyl sulfoxide (DMSO) control. The dose–response curves were determined by quantification of viral RNA copy numbers in the supernatant of infected cell at 48 h post infection (p.i.). As demonstrated in Fig. 1a, arbidol efficiently inhibited virus infection in vitro. The 50% maximal effective concentration (EC50) and the 50% cytotoxic concentration (CC50) of arbidol was 4.11 (3.55–4.73) and 31.79 (29.89–33.81) μM, respectively, and the selectivity index (SI = CC50/EC50) was 7.73. Baloxavir partially inhibited SARS-CoV-2 infection (~29%) at a high concentration of 50 μM (Fig. 1a). In contrast, laninamivir, oseltamivir, peramivir, and zanamivir did not exhibit anti-SARS-CoV-2 activity even at the highest drug concentrations (Fig. 1a). The antiviral effect of the compounds was also evaluated by observing cytopathic effects (CPE) and immunofluorescence staining of infected cells. As shown in Supplementary Fig. S1, at 48 h p.i. only in cells treated with arbidol, but not with the other five drugs, viral NP expression and CPE due to SARS-CoV-2 was substantially reduced. To be noted, we also tried some human lung cell lines, for example human embryo lung fibroblasts MRC-5 and lung cancer cell line Calu-3, however, they were not very efficient for SARS-CoV-2 replication, and therefore were not used for this study. Fig. 1 Comparative antiviral efficacy of anti-influenza drugs and the mode of actions of arbidol against SARS-CoV-2 infection in vitro. a Antiviral activities of the drugs. The antiviral efficacy was evaluated in Vero E6 cells by qRT-PCR analysis of virus yield at 48 h p.i. Data represent the mean ± standard deviation (SD) from two independent repeats. b, c Time-of-addition experiment of arbidol. Three experimental groups (Full-time, Entry, and Post-entry) were set up as described in the Supplementary Methods. At 16 h p.i., virus yield in the cell supernatant was quantified by qRT-PCR (b), and the expression of NP in infected cells was analyzed by western blots (c). The values below the blot represent the relative band intensity (NP/GAPDH) normalized to that of the DMSO group. d Impact of arbidol on SARS-CoV-2 binding. Vero E6 cells were treated with arbidol (10 μM) or DMSO for 1 h prior to infection with SARS-CoV-2 at 4 °C for 1 h. The supernatant (unbound virions) and the cells containing bound virions (bound virions) were collected for quantification of viral RNA copies by qRT-PCR. e, f Effect of arbidol on intracellular trafficking of SARS-CoV-2. The co-localization of virions with EEs or LEs was analyzed by immunofluorescence assays as described in the Supplementary Methods. e The portion of virions that co-localized with EEs or ELs in each group (n > 150 cells) was quantified by Image J. f Representative confocal microscopic images of virions (red) and LAMP1+ ELs (green) in each group. The nuclei (blue) were stained with Hoechst 33258 dye. White arrows: virions co-localized with ELs; bars: 10 μm. For (b) and (e), statistical analysis was performed using a one-way analysis of variance (ANOVA) with GraphPad Prism. For (d), statistical analysis was performed and calculated by unpaired two-tailed t test. *P < 0.05; ***P < 0.001; ns, not significant. Apart from influenza virus, arbidol was reported to inhibit a wide array of viruses by interfering with multiple steps of the virus replication cycle 7 . The stage of SARS-CoV-2 replication targeted by arbidol was explored by conducting a preliminary time-of-addition experiment using virus at an MOI of 0.05. Arbidol was incubated with cells during the virus entry process (Entry), the post-entry stages (Post-entry), or the entire process of infection (Full-time) and progeny virus yield was quantified by qRT-PCR. The data revealed that arbidol efficiently blocked both viral entry and post-entry stages. It had a profound impact on virus Entry (~75% inhibition) with a lesser effect on Post-entry events (~55% inhibition rate) (Fig. 1b). In addition, western blot analysis (Fig. 1c) and immunofluorescence microscopy (Supplementary Fig. S2) confirmed that the expression level of viral NP was reduced drastically at Full-time (13% of the DMSO group, Fig. 1c), and showed more inhibitory effect at the Entry stage (41%) than at the Post-entry stage (61%). The details of how arbidol blocks the entry of SARS-CoV-2 into cells were further investigated. Virus (MOI = 0.05) was allowed to bind to Vero E6 cells at 4 °C for 1 h in the presence of arbidol (10 μM) or DMSO control. Virus particles bound to the cell (bound virions) and those in the supernatant (unbound virions) were analyzed by qRT-PCR. The results showed that arbidol treatment led to a significantly decreased binding efficiency (67%) compared with the control group (P < 0.05) (Fig. 1d). Correspondingly, the portion of unbound virions increased significantly to 156% of the control group after arbidol treatment (P < 0.001) (Fig. 1d). Next, we analyzed viral intracellular trafficking. As we reported recently, within infected cells, SARS-CoV-2 underwent vesicle transportation, which was first carried out by early endosomes (EEs) then further transported to endolysosomes (ELs) 8 . Co-localization of virions with EEs or ELs was visualized by immunofluorescence microscopy and statistically analyzed (n > 150 cells). As shown in Fig. 1e and Supplementary Fig. S3, in each tracked time points, there was no significant difference in the amounts of virions co-localized with EEs when comparing the DMSO- and arbidol-treated groups, although as time of infection went on (30, 60, and 90 min p.i.), the levels of co-localization considerably decreased in both DMSO- (24.0%, 5.1%, and 3.2%) and arbidol- (21.4%, 4.1%, and 2.8%) treated groups, suggesting that some virions were already transported from EEs to the next stage of vesicle transportation. By contrast, at 60 min p.i., a slightly higher percentage of virions were transported to ELs in the arbidol-treated group (22.4%) than in the DMSO group (18.3%) (P < 0.05) (Fig. 1e, f). At 90 min p.i., significantly fewer virions (~13.5%) were detected in ELs in the DMSO group; whereas significantly higher proportions of virions (~23.6%) remained within ELs in the arbidol-treated group, suggesting the drug trapped the virus in the ELs (P < 0.001) (Fig. 1e, f). Taken together, these results suggested that arbidol impeded not only viral attachment, but also release of SARS-CoV-2 from intracellular vesicles (ELs). Among the drugs tested, laninamivir, oseltamivir, peramivir, and zanamivir are neuraminidase (NA) inhibitors, which are most widely prescribed for prophylaxis and treatment of influenza. Although no NA analog exists in SARS-CoV-2, NA inhibitors such as oseltamivir nevertheless are being used clinically in treating COVID-19 patients 1,2 . Our data showed these NA inhibitors were not active against SARS-CoV-2 (Fig. 1a), which is consistent with the finding that oseltamivir and zanamivir were ineffective in inhibiting SARS-CoV 9 . Baloxavir marboxil is a new anti-influenza drug, which selectively inhibits the endonuclease activity of the viral polymerase responsible for snatching capped primers from host mRNAs to initiate viral mRNA transcription. However, this “cap-snatching” mechanism of the endonuclease is not shared by coronaviruses that encode their own enzymes to form 5ʹ-mRNA cap structures 10 . This may explain why baloxavir failed to block SARS-CoV-2 infection (Fig. 1a). During the review process of this study, Choy et al. also showed that oseltamivir and baloxavir failed to inhibit SARS-CoV-2 in vitro 11 . Arbidol, an indole-derivative, has been licensed for decades in Russia and China against influenza. It is a broad-spectrum drug against a wide range of enveloped and non-enveloped viruses. Arbidol interacts preferentially with aromatic amino acids, and it affects multiple stages of the virus life cycle, either by direct targeting viral proteins or virus-associated host factors 7 . For example, in influenza virus, crystal structures showed that arbidol inserted into a hydrophobic pocket of the fusion subunit of HA, thus hindering low-pH conformational change of HA and blocking the fusion process 12 . In hepatitis C virus, arbidol impaired both virus attachment and intracellular vesicle trafficking 13 . Likewise, we found arbidol plays a role in interfering SAS-CoV-2 binding (Fig. 1d) and intracellular vesicle trafficking (Fig. 1e, f). Arbidol can also bind to lipid membranes and may alter membrane configuration of the cytoplasm or the endosome, which are crucial for viral attachment and fusion 7 . It could be further investigated whether arbidol targets virus or/and cells by using published method 14 . In summary, among the six anti-influenza drugs, only arbidol efficiently inhibited SARS-CoV-2 infection. Functionally, it appears to block virus entry by impeding viral attachment and release from the ELs. Although the SI of arbidol is relatively low (SI = 7.73), as a repurposed drug, its pharmacokinetics profile such as maximal concentration (Cmax) is more important for predicting efficacy. It is generally believed that if the Cmax achieves EC90, the drug is very likely to be effective; while if the Cmax achieves EC50, the drug is possibly effective in vivo. In humans, a single oral administration of 800 mg of arbidol results in Cmax of ~4.1 μM 15 , and this dosage is efficacious and safe against different influenza viruses with EC50 values ranging from 2.5–20 μM 7,16 . Arbidol also showed anti-inflammatory activity, which may enhance its efficacy in vivo 16 . Considering the EC50 (4.11 μM) of arbidol against SARS-CoV-2 is comparable to, or even lower than those of influenza viruses, we, therefore, suggest that arbidol is potentially effective to treat COVID-19 patients. However, the current dose of arbidol (200 mg, 3 times/day) recommended by the Chinese Guidelines may not be able to achieve an ideal therapeutic efficacy to inhibit SARS-CoV-2 infection, and should be elevated. This needs to be verified by clinical trials. Supplementary information Supplementary Figs. S1-S3

          Related collections

          Most cited references11

          • Record: found
          • Abstract: found
          • Article: not found

          Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China

          Summary Background A recent cluster of pneumonia cases in Wuhan, China, was caused by a novel betacoronavirus, the 2019 novel coronavirus (2019-nCoV). We report the epidemiological, clinical, laboratory, and radiological characteristics and treatment and clinical outcomes of these patients. Methods All patients with suspected 2019-nCoV were admitted to a designated hospital in Wuhan. We prospectively collected and analysed data on patients with laboratory-confirmed 2019-nCoV infection by real-time RT-PCR and next-generation sequencing. Data were obtained with standardised data collection forms shared by WHO and the International Severe Acute Respiratory and Emerging Infection Consortium from electronic medical records. Researchers also directly communicated with patients or their families to ascertain epidemiological and symptom data. Outcomes were also compared between patients who had been admitted to the intensive care unit (ICU) and those who had not. Findings By Jan 2, 2020, 41 admitted hospital patients had been identified as having laboratory-confirmed 2019-nCoV infection. Most of the infected patients were men (30 [73%] of 41); less than half had underlying diseases (13 [32%]), including diabetes (eight [20%]), hypertension (six [15%]), and cardiovascular disease (six [15%]). Median age was 49·0 years (IQR 41·0–58·0). 27 (66%) of 41 patients had been exposed to Huanan seafood market. One family cluster was found. Common symptoms at onset of illness were fever (40 [98%] of 41 patients), cough (31 [76%]), and myalgia or fatigue (18 [44%]); less common symptoms were sputum production (11 [28%] of 39), headache (three [8%] of 38), haemoptysis (two [5%] of 39), and diarrhoea (one [3%] of 38). Dyspnoea developed in 22 (55%) of 40 patients (median time from illness onset to dyspnoea 8·0 days [IQR 5·0–13·0]). 26 (63%) of 41 patients had lymphopenia. All 41 patients had pneumonia with abnormal findings on chest CT. Complications included acute respiratory distress syndrome (12 [29%]), RNAaemia (six [15%]), acute cardiac injury (five [12%]) and secondary infection (four [10%]). 13 (32%) patients were admitted to an ICU and six (15%) died. Compared with non-ICU patients, ICU patients had higher plasma levels of IL2, IL7, IL10, GSCF, IP10, MCP1, MIP1A, and TNFα. Interpretation The 2019-nCoV infection caused clusters of severe respiratory illness similar to severe acute respiratory syndrome coronavirus and was associated with ICU admission and high mortality. Major gaps in our knowledge of the origin, epidemiology, duration of human transmission, and clinical spectrum of disease need fulfilment by future studies. Funding Ministry of Science and Technology, Chinese Academy of Medical Sciences, National Natural Science Foundation of China, and Beijing Municipal Science and Technology Commission.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: a descriptive study

            Summary Background In December, 2019, a pneumonia associated with the 2019 novel coronavirus (2019-nCoV) emerged in Wuhan, China. We aimed to further clarify the epidemiological and clinical characteristics of 2019-nCoV pneumonia. Methods In this retrospective, single-centre study, we included all confirmed cases of 2019-nCoV in Wuhan Jinyintan Hospital from Jan 1 to Jan 20, 2020. Cases were confirmed by real-time RT-PCR and were analysed for epidemiological, demographic, clinical, and radiological features and laboratory data. Outcomes were followed up until Jan 25, 2020. Findings Of the 99 patients with 2019-nCoV pneumonia, 49 (49%) had a history of exposure to the Huanan seafood market. The average age of the patients was 55·5 years (SD 13·1), including 67 men and 32 women. 2019-nCoV was detected in all patients by real-time RT-PCR. 50 (51%) patients had chronic diseases. Patients had clinical manifestations of fever (82 [83%] patients), cough (81 [82%] patients), shortness of breath (31 [31%] patients), muscle ache (11 [11%] patients), confusion (nine [9%] patients), headache (eight [8%] patients), sore throat (five [5%] patients), rhinorrhoea (four [4%] patients), chest pain (two [2%] patients), diarrhoea (two [2%] patients), and nausea and vomiting (one [1%] patient). According to imaging examination, 74 (75%) patients showed bilateral pneumonia, 14 (14%) patients showed multiple mottling and ground-glass opacity, and one (1%) patient had pneumothorax. 17 (17%) patients developed acute respiratory distress syndrome and, among them, 11 (11%) patients worsened in a short period of time and died of multiple organ failure. Interpretation The 2019-nCoV infection was of clustering onset, is more likely to affect older males with comorbidities, and can result in severe and even fatal respiratory diseases such as acute respiratory distress syndrome. In general, characteristics of patients who died were in line with the MuLBSTA score, an early warning model for predicting mortality in viral pneumonia. Further investigation is needed to explore the applicability of the MuLBSTA score in predicting the risk of mortality in 2019-nCoV infection. Funding National Key R&D Program of China.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: found
              Is Open Access

              Remdesivir and chloroquine effectively inhibit the recently emerged novel coronavirus (2019-nCoV) in vitro

              Dear Editor, In December 2019, a novel pneumonia caused by a previously unknown pathogen emerged in Wuhan, a city of 11 million people in central China. The initial cases were linked to exposures in a seafood market in Wuhan. 1 As of January 27, 2020, the Chinese authorities reported 2835 confirmed cases in mainland China, including 81 deaths. Additionally, 19 confirmed cases were identified in Hong Kong, Macao and Taiwan, and 39 imported cases were identified in Thailand, Japan, South Korea, United States, Vietnam, Singapore, Nepal, France, Australia and Canada. The pathogen was soon identified as a novel coronavirus (2019-nCoV), which is closely related to sever acute respiratory syndrome CoV (SARS-CoV). 2 Currently, there is no specific treatment against the new virus. Therefore, identifying effective antiviral agents to combat the disease is urgently needed. An efficient approach to drug discovery is to test whether the existing antiviral drugs are effective in treating related viral infections. The 2019-nCoV belongs to Betacoronavirus which also contains SARS-CoV and Middle East respiratory syndrome CoV (MERS-CoV). Several drugs, such as ribavirin, interferon, lopinavir-ritonavir, corticosteroids, have been used in patients with SARS or MERS, although the efficacy of some drugs remains controversial. 3 In this study, we evaluated the antiviral efficiency of five FAD-approved drugs including ribavirin, penciclovir, nitazoxanide, nafamostat, chloroquine and two well-known broad-spectrum antiviral drugs remdesivir (GS-5734) and favipiravir (T-705) against a clinical isolate of 2019-nCoV in vitro. Standard assays were carried out to measure the effects of these compounds on the cytotoxicity, virus yield and infection rates of 2019-nCoVs. Firstly, the cytotoxicity of the candidate compounds in Vero E6 cells (ATCC-1586) was determined by the CCK8 assay. Then, Vero E6 cells were infected with nCoV-2019BetaCoV/Wuhan/WIV04/2019 2 at a multiplicity of infection (MOI) of 0.05 in the presence of varying concentrations of the test drugs. DMSO was used in the controls. Efficacies were evaluated by quantification of viral copy numbers in the cell supernatant via quantitative real-time RT-PCR (qRT-PCR) and confirmed with visualization of virus nucleoprotein (NP) expression through immunofluorescence microscopy at 48 h post infection (p.i.) (cytopathic effect was not obvious at this time point of infection). Among the seven tested drugs, high concentrations of three nucleoside analogs including ribavirin (half-maximal effective concentration (EC50) = 109.50 μM, half-cytotoxic concentration (CC50) > 400 μM, selectivity index (SI) > 3.65), penciclovir (EC50 = 95.96 μM, CC50 > 400 μM, SI > 4.17) and favipiravir (EC50 = 61.88 μM, CC50 > 400 μM, SI > 6.46) were required to reduce the viral infection (Fig. 1a and Supplementary information, Fig. S1). However, favipiravir has been shown to be 100% effective in protecting mice against Ebola virus challenge, although its EC50 value in Vero E6 cells was as high as 67 μM, 4 suggesting further in vivo studies are recommended to evaluate this antiviral nucleoside. Nafamostat, a potent inhibitor of MERS-CoV, which prevents membrane fusion, was inhibitive against the 2019-nCoV infection (EC50 = 22.50 μM, CC50 > 100 μM, SI > 4.44). Nitazoxanide, a commercial antiprotozoal agent with an antiviral potential against a broad range of viruses including human and animal coronaviruses, inhibited the 2019-nCoV at a low-micromolar concentration (EC50 = 2.12 μM; CC50 > 35.53 μM; SI > 16.76). Further in vivo evaluation of this drug against 2019-nCoV infection is recommended. Notably, two compounds remdesivir (EC50 = 0.77 μM; CC50 > 100 μM; SI > 129.87) and chloroquine (EC50 = 1.13 μM; CC50 > 100 μM, SI > 88.50) potently blocked virus infection at low-micromolar concentration and showed high SI (Fig. 1a, b). Fig. 1 The antiviral activities of the test drugs against 2019-nCoV in vitro. a Vero E6 cells were infected with 2019-nCoV at an MOI of 0.05 in the treatment of different doses of the indicated antivirals for 48 h. The viral yield in the cell supernatant was then quantified by qRT-PCR. Cytotoxicity of these drugs to Vero E6 cells was measured by CCK-8 assays. The left and right Y-axis of the graphs represent mean % inhibition of virus yield and cytotoxicity of the drugs, respectively. The experiments were done in triplicates. b Immunofluorescence microscopy of virus infection upon treatment of remdesivir and chloroquine. Virus infection and drug treatment were performed as mentioned above. At 48 h p.i., the infected cells were fixed, and then probed with rabbit sera against the NP of a bat SARS-related CoV 2 as the primary antibody and Alexa 488-labeled goat anti-rabbit IgG (1:500; Abcam) as the secondary antibody, respectively. The nuclei were stained with Hoechst dye. Bars, 100 μm. c and d Time-of-addition experiment of remdesivir and chloroquine. For “Full-time” treatment, Vero E6 cells were pre-treated with the drugs for 1 h, and virus was then added to allow attachment for 2 h. Afterwards, the virus–drug mixture was removed, and the cells were cultured with drug-containing medium until the end of the experiment. For “Entry” treatment, the drugs were added to the cells for 1 h before viral attachment, and at 2 h p.i., the virus–drug mixture was replaced with fresh culture medium and maintained till the end of the experiment. For “Post-entry” experiment, drugs were added at 2 h p.i., and maintained until the end of the experiment. For all the experimental groups, cells were infected with 2019-nCoV at an MOI of 0.05, and virus yield in the infected cell supernatants was quantified by qRT-PCR c and NP expression in infected cells was analyzed by Western blot d at 14 h p.i. Remdesivir has been recently recognized as a promising antiviral drug against a wide array of RNA viruses (including SARS/MERS-CoV 5 ) infection in cultured cells, mice and nonhuman primate (NHP) models. It is currently under clinical development for the treatment of Ebola virus infection. 6 Remdesivir is an adenosine analogue, which incorporates into nascent viral RNA chains and results in pre-mature termination. 7 Our time-of-addition assay showed remdesivir functioned at a stage post virus entry (Fig. 1c, d), which is in agreement with its putative anti-viral mechanism as a nucleotide analogue. Warren et al. showed that in NHP model, intravenous administration of 10 mg/kg dose of remdesivir resulted in concomitant persistent levels of its active form in the blood (10 μM) and conferred 100% protection against Ebola virus infection. 7 Our data showed that EC90 value of remdesivir against 2019-nCoV in Vero E6 cells was 1.76 μM, suggesting its working concentration is likely to be achieved in NHP. Our preliminary data (Supplementary information, Fig. S2) showed that remdesivir also inhibited virus infection efficiently in a human cell line (human liver cancer Huh-7 cells), which is sensitive to 2019-nCoV. 2 Chloroquine, a widely-used anti-malarial and autoimmune disease drug, has recently been reported as a potential broad-spectrum antiviral drug. 8,9 Chloroquine is known to block virus infection by increasing endosomal pH required for virus/cell fusion, as well as interfering with the glycosylation of cellular receptors of SARS-CoV. 10 Our time-of-addition assay demonstrated that chloroquine functioned at both entry, and at post-entry stages of the 2019-nCoV infection in Vero E6 cells (Fig. 1c, d). Besides its antiviral activity, chloroquine has an immune-modulating activity, which may synergistically enhance its antiviral effect in vivo. Chloroquine is widely distributed in the whole body, including lung, after oral administration. The EC90 value of chloroquine against the 2019-nCoV in Vero E6 cells was 6.90 μM, which can be clinically achievable as demonstrated in the plasma of rheumatoid arthritis patients who received 500 mg administration. 11 Chloroquine is a cheap and a safe drug that has been used for more than 70 years and, therefore, it is potentially clinically applicable against the 2019-nCoV. Our findings reveal that remdesivir and chloroquine are highly effective in the control of 2019-nCoV infection in vitro. Since these compounds have been used in human patients with a safety track record and shown to be effective against various ailments, we suggest that they should be assessed in human patients suffering from the novel coronavirus disease. Supplementary information Supplementary information, Materials and Figures
                Bookmark

                Author and article information

                Contributors
                huzh@wh.iov.cn
                zhongwu@bmi.ac.cn
                wangml@wh.iov.cn
                Journal
                Cell Discov
                Cell Discov
                Cell Discovery
                Springer Singapore (Singapore )
                2056-5968
                2 May 2020
                2 May 2020
                2020
                : 6
                : 28
                Affiliations
                [1 ]ISNI 0000000119573309, GRID grid.9227.e, State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, , Chinese Academy of Sciences, ; Wuhan, 430071 China
                [2 ]ISNI 0000 0004 1803 4911, GRID grid.410740.6, National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, ; Beijing, 100850 China
                [3 ]ISNI 0000 0004 1797 8419, GRID grid.410726.6, University of the Chinese Academy of Sciences, ; Beijing, 100049 China
                [4 ]ISNI 0000000119573309, GRID grid.9227.e, National Virus Resource Center, Wuhan Institute of Virology, , Chinese Academy of Sciences, ; Wuhan, 430071 China
                Author information
                http://orcid.org/0000-0002-1560-0928
                http://orcid.org/0000-0001-8701-3530
                Article
                169
                10.1038/s41421-020-0169-8
                7195821
                31934347
                de5c05f3-745b-4da3-8d38-0f5c0f11f2fe
                © The Author(s) 2020

                Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.

                History
                : 29 March 2020
                : 11 April 2020
                Funding
                Funded by: the National Key R&D program of China (grant no. 2020YFC0841700)
                Funded by: the National Natural Science Foundation of China (grant no. 31621061 to Z.H.).
                Funded by: the National Science and Technology Major Projects for “Major New Drugs Innovation and Development” (grant no. 2018ZX09711003), the National Key R&D program of China (grant no. 2020YFC0841700)
                Categories
                Correspondence
                Custom metadata
                © The Author(s) 2020

                cell biology,molecular biology
                cell biology, molecular biology

                Comments

                Comment on this article