27
views
0
recommends
+1 Recommend
2 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      TMPRSS2: Potential Biomarker for COVID‐19 Outcomes

      article-commentary

      Read this article at

      ScienceOpenPublisherPMC
      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

          Coronavirus disease 2019 (COVID‐19) clinical data has so far shown that the mortality rate for men is higher than for women. This disparity is observed worldwide and across different ethnic/racial groups (Table 1). Early reports from Italy and Germany show that while infection rates are similar between sexes, nearly 70% and 65%, respectively, of deaths are males. In New York City, an epicenter of the US outbreak, 54% of those infected are men, yet men account for 63% of deaths. Epidemiologic data from the previous coronavirus infections, severe acute respiratory syndrome (SARS) and the Middle East respiratory syndrome (MERS), also indicated sex‐based differences in disease susceptibility and outcomes. This discrepancy was attributed to many factors, including smoking, immune differences, and other comorbidities. An initial report released by the Centers for Disease Control and Prevention (CDC) on population‐based surveillance sampled across 14 states, representing 10% of the US population, has indicated that age and comorbidities are associated with increased hospitalization rates of patients with COVID‐19. 1 The data on sex also suggest sexual dimorphism consistent with reports from other countries (Table 1). The preliminary data on race suggest that minority populations may be disproportionately impacted by the coronavirus, where blacks contributed to 33% of the hospitalizations despite representing only 18% of the sampled population. As more data become available, correlations between race and disease severity can be interrogated more thoroughly, including the role of socioeconomic factors on influencing this disparity. Investigations into the genetic and molecular differences between women and men are warranted to identify relevant biomarkers for disease susceptibility and outcomes. Based on data from literature, we propose a novel mechanism of the observed sex differences in clinical outcomes in patients and identify a role for the transmembrane protease serine 2 (TMPRSS2) as a contributing factor to the more severe outcomes noted for COVID‐19. Table 1 Incidence of Mortality in COVID‐19 Patientsa Location Sex Incidence, % Death, % North America United Statesb Female  46g 41 Male  54 g 59 Chicago c Female 49 41 Male 50 59 New Yorkd Female 47 38 Male 53 62 Washington Statee Female 52 43 Male 45 57 Michiganf Female 54 43 Male 45 57 Canada Female 54 48 Male 46 58 Mexico Female 42 30 Male 58 70 Europe Italy Female 48 33 Male 52 67 France Female 53 39 Male 47 61 Germany Female 51 41 Male 49 59 Denmark Female 56 39 Male 44 61 Ireland Female 55 41 Male 45 59 Spain Female 51 37 Male 49 63 Switzerland Female 53 39 Male 47 61 The Netherlands Female 58 39 Male 42 61 Asia South Korea Female 60 48 Male 40 52 Philippines Female 45 31 Male 55 69 China Female 49 36 Male 51 64 South America Ecuador Female 42 30 Male 58 70 Columbia Female 49 38 Male 51 62 Peru Female 42 28 Male 58 72 Australia Australia Female 49 40 Male 51 60 Data from https://globalhealth5050.org/covid19/ and the websites in notes b‐g (accessed on April 16, 2020). https://www.cdc.gov/nchs/nvss/vsrr/COVID19/ https://www.chicago.gov/city/en/sites/covid-19/home/latest-data.html https://www1.nyc.gov/assets/doh/downloads/pdf/imm/covid-19-daily-data-summary-deaths-04072020-1.pdf and https://www1.nyc.gov/assets/doh/downloads/pdf/imm/covid-19-daily-data-summary-04072020-1.pdf https://www.doh.wa.gov/emergencies/coronavirus https://www.michigan.gov/coronavirus/0,9753,7-406-98163_98173—,00.html https://www.cdc.gov/mmwr/volumes/69/wr/mm6915e3.htm (data from hospitalized patients in 14 states). John Wiley & Sons, Ltd. This article is being made freely available through PubMed Central as part of the COVID-19 public health emergency response. It can be used for unrestricted research re-use and analysis in any form or by any means with acknowledgement of the original source, for the duration of the public health emergency. TMPRSS2 Expression as a Biomarker of Clinical Outcomes The angiotensin I–converting enzyme 2 (ACE2) and TMPRSS2 have been implicated in influenza and SARS‐corona virus (CoV) infection as well as for SARS‐CoV‐2 in mediating viral entry into the host cell. 2 Both genes mediate sex‐specific effects with ACE2, located on the X chromosome and TMPRSS2 regulated by androgen (located on 21q22.3). While ACE2 is the main receptor for the spike protein for both viruses, its expression and gene polymorphisms did not influence sex‐specific effects or outcomes for SARS 3 or COVID‐19 (based on 2 recent preprint studies published on medRxiv that have yet to be peer‐reviewed at the time of writing). 4 , 5 TMPRSS2 is an androgen‐responsive serine protease that cleaves SARS‐CoV‐2 spike protein, facilitating viral entry and activation. 2 It is expressed in many tissues outside of the lungs 6 and is well known for its predominant expression in the prostate epithelium and its role in prostate carcinogenesis. 7 , 8 The androgen‐regulated promoter of TMPRSS2 forms a fusion gene with coding regions of the proto‐oncogenic ETS transcription factor family members, which is strongly associated with prostate cancer and regulates many biological processes. 9 TMPRSS2 is also expressed on the cardiac endothelium, kidney, and digestive tract, suggesting that these may be important target organs for SARS‐CoV‐2 infection. 6 Indeed, the clinical manifestations of COVID‐19 include not only complications from acute myocardial injury, elevated liver enzymes, and acute kidney injury in patients presenting to hospitals, but also gastrointestinal symptoms in community patients experiencing milder forms of the disease. 10 , 11 Because TMPRSS2 is also expressed on microvascular endothelial cells that line blood vessels, 6 viral infection by SARS‐CoV‐2 may cause endothelial dysfunction, which can lead to thrombosis and associated complications. These data present a rationale for the role of TMPRSS2 in clinically observed complications in patients with severe disease. TMPRSS2 Single‐Nucleotide Polymorphisms as Biomarkers of Disease Outcomes Studies into TMPRSS2 single‐nucleotide polymorphisms (SNPs) have been conducted in various diseases. In breast cancer, the minor allele for rs2276205 (A>G) was associated with increased survival, potentially due to sensitization to tamoxifen. 12 In prostate cancer, the most studied disease in relation to TMPRSS2, rs12329760 (C>T) was associated with cancer in men with a family history of prostate cancer, multiple copies of the ERG gene fusion, and in whites with a doubled time to cancer diagnosis. 13 , 14 Rs2070788 (G>A) and rs383510 (T>C) were associated with severe H1N1, H7N9, and increased expression of TMPRSS2 in the lungs. 15 Most recently, a preprint study comparing genetic variants in TMPRSS2 among Italians, Europeans, and East Asians, showed allele frequency differences in rs12329760 and 2 distinct eQTL haplotypes between Italians and East Asians. 5 The study proposed that TMPRSS2 could possibly be a candidate gene that contributes to the COVID‐19 epidemiologic data seen in the Italian population, with higher death rates and differences in severity among sexes compared to East Asians. Interestingly, Asian men have a decreased incidence and mortality of prostate cancer and a lower frequency of the TMPRSS2:ERG fusion gene. 16 , 17 We may never know the true incidence rates for COVID‐19; however, looking at current data, Europe and the United States have overtaken China and eastern Asia as the epicenters of the pandemic. Accurate infection rates may demonstrate that East Asians are less susceptible to severe disease outcomes. TMPRSS2 Inhibitors, Rationale for Drug Combinations, and Clinical Biomarkers of Response TMPRSS2:ERG fusions can upregulate genes related to the nuclear factor–kB pathway including Toll‐like receptor‐4 and the Notch pathways in prostate cancer. 18 , 19 The role of TMPRSS2 and TMPRSS2:ERG fusions in nonprostatic tissue remain to be elucidated, including whether this gene can subsequently activate the Notch signaling cascade in macrophages to augment Toll‐like receptor–associated inflammatory responses and release of proinflammatory cytokines, such as tumor necrosis factor‐α, interleukin (IL)‐1β, and IL‐6. 20 Clinically, increases in IL‐6 have been linked to severe COVID‐19 cases 21 and have been discussed as a potential biomarker. 22 Cytokine release syndrome (CRS) has been noted in some severe patients of COVID‐19. 23 Coincidentally, TMPRSS2‐deficient mice were protective against SARS‐CoV infection and showed lower expression levels of cytokines and chemokines, suggesting that TMPRSS2 may be involved in regulating the production of these inflammatory markers. 24 Furthermore, autopsy findings from COVID‐19 patients revealed that tissues that had been destroyed by SARS‐CoV‐2 exhibited characteristics of the so‐called primary cytokine storm (induced by viral infection and mainly produced by alveolar macrophages, epithelial cells, and endothelial cells), rather than those observed in secondary cytokine storm (induced by different subsets of activated T lymphocytes in late‐stage viral infections or a complication of T‐cell–engaging therapies). 25 Together with data from the TMPRSS2 knockout mice, which reveal that TMPRSS2 contributes to the spread and immunopathology of the virus at primary sites of infection, the evidence further points to an underlying mechanism that may involve endothelial dysfunction, which can lead to thrombi formation that is observed in many cases. 21 Moreover, because the physiological role of TMPRSS2 is unknown, what remains to be determined is the role of androgen/androgen signaling and other sex steroids in modulating TMPRSS2 expression during active infection and/or in driving potential novel fusions with possibly another ETS family member that regulate genes involved in the immune response. Our laboratory and others have demonstrated that antiandrogens (eg, enzalutamide, apalutamide) can decrease TMPRSS2 expression in prostate cancer cells. 26 , 27 Whether the use of these androgen pathway inhibitors to decrease TMPRSS2 expression has activity against SARS‐CoV infection also remains to be addressed. There are currently no US Food and Drug Administration–approved treatments for any human CoV infection, and new interventions are likely to require months to years to develop. In the face of a pandemic, drug repurposing, using existing medicines that having already been tested safely in humans, is an emerging strategy that offers a faster approach to identifying an effective COVID‐19 treatment. Rigorous testing in double‐blinded, randomized, controlled trials with a larger sample size are needed to determine the safety and efficacy of these new drug combinations and to guide clinical decisions. While each repurposed drug individually may not yield a significant overall clinical benefit, carefully combined cocktails could be very effective (eg, HIV), focusing on a multimodality approach. The key is to find the right combination in the case of COVID‐19. In standard drug development, many proposed combinations would usually undergo in vivo preclinical testing to provide rationale for the regimen. However, there are currently no robust mouse models that recapitulate the SARS‐CoV‐2 pathogenesis observed in humans. Due to structural differences in mouse ACE2 compared to human ACE2 (hACE2) proteins, the SARS coronaviruses exhibit poor tropism characteristics for mouse tissues and are inefficient at infecting mice. McCray et al 28 successfully developed a hACE2 transgenic mouse strain (K18‐hACE2) as a mouse model for SARS. Since SARS‐CoV‐2 shares similar mechanisms of viral entry with SARS‐CoV, this transgenic model could potentially be used to study novel therapeutics for COVID‐19. The Jackson Laboratory is currently in the process of reestablishing a new K18‐hACE2 transgenic mouse colony. Because SARS‐CoV‐2 disease severity increases with increasing age, we suggest future drug efficacy testing in CoV aged mouse models to fully recapitulate the age‐related increase in pathogenesis observed in humans. In summary, no in vivo preclinical data for SARS‐CoV‐2 are available to justify any of the treatment regimens for combination trials currently under investigation for COVID‐19, as an in vivo mouse model for SARS‐CoV‐2 is not yet available, and it would require too much time to test each proposed drug combination in primate models of human disease or use existing models for MERS‐CoV and SARS‐CoV pathogenesis. Given the urgency of the COVID‐19 pandemic, repurposing existing drugs or antiviral agents approved or in development for treating infections caused by HIV, hepatitis B/C, and influenza are mostly based on therapeutic experience with SARS/MERS. In fact, many of the proposed combination trials registered on the World Health Organization (WHO) website and ClnicalTrials.gov, including the current WHO Solidarity trial, are testing drug combinations based on existing case studies and anecdotal evidence from early treatment approaches used in China, in vitro/in vivo data from testing for SARS/MERS, and histopathological evidence from COVID‐19 cases. Many of the proposed treatment combinations are based on demonstrated antiviral activity of the drug(s) in tissue culture coupled with scientific rationale based on proposed mechanisms of action in conjunction with the need for effective therapy against SARS‐CoV‐2 to justify the need. There is no doubt that antivirals and supportive treatments are key areas to focus on in treating patients with COVID‐19. Data from SARS‐ and MERS‐CoV infections show that the disease process is driven by both virus and host immune response factors. Our proposed treatment combination is based on existing data from in vitro/in vivo studies that have been conducted for SARS‐CoV and MERS‐CoV for each individual agent as well as clinical features of COVID‐19 since current in vivo animal models are not available for SARS‐CoV‐2. We propose a drug combination therapy that can effectively limit viral entry and replication as well as target systemic clinical manifestations of viral infection (Figure 1). For example, we propose the rationale for a treatment regimen that includes a TMPRSS2 inhibitor in combination with antivirals and/or inhibitors of CRS. Camostat and nafamostat are TMPRSS2 inhibitors currently approved to treat pancreatitis in Japan. These drugs have been shown to inhibit MERS infection by blocking viral entry, with nafamostat being the more potent of the two. 29 Both are being investigated in Japan and other countries for their ability to block SARS‐CoV‐2 entry, with Japan initiating a clinical trial for nafamostat (which is also a short‐acting anticoagulant) for COVID‐19–positive patients. 30 Direct inhibition of TMPRSS2 using protease inhibitors (nafamostat) or indirectly by downregulating TMPRSS2 expression with androgen pathway inhibitors, as with all potential novel treatments for COVID‐19, are promising hypotheses, and it remains to be determined whether this approach can mediate the sex‐specific outcomes and severity of outcomes in patients. Figure 1 Proposed COVID‐19 treatments. Rationale for combination therapy that effectively limits viral entry and replication as well as targets systemic clinical manifestations of viral infection such as cytokine storm. Direct TMPRSS2 inhibition with camostat and nafamostat. Remdesivir, an RNA‐dependent RNA polymerase inhibitor with anti‐viral activity, targets viral replication post entry in SARS‐CoV‐2. Hydroxychloroquine (HCQ) and chloroquine (CQ) exhibit broad spectrum effects that include viral inhibition, suppression of multiple cytokines, and vascular protective effects. Some agents target cytokine release such the anti‐IL‐6 antibody tocilizumab, the anti‐human GM‐CSF monoclonal antibody lenzilumab, or the JAK1/2 inhibitor ruxolitinib. ACE2, angiotensin I–converting enzyme 2; COVID‐19, coronavirus disease 2019; GM‐CSF, granulocyte macrophage colony‐stimulating factor; IL, interleukin; JAK, Janus kinase; SARS‐CoV‐2, severe acute respiratory syndrome coronavirus 2. Created using BioRender.com. Another drug under investigation for COVID‐19 is remdesivir, an RNA‐dependent RNA polymerase inhibitor with antiviral activity, which has been shown to act on viral replication after entry in SARS‐CoV‐2. 31 While remdesivir is not US Food and Drug Administration approved for any indications, the WHO considered it as a promising drug candidate to treat COVID‐19 based on its broad‐spectrum activity, in vitro/in vivo data for coronaviruses (SARS‐CoV‐2, SARS‐CoV, and MERS‐CoV), and clinical safety data from Ebola virus disease trials. Initial results of the compassionate use of remdesivir in patients with COVID‐19 have shown some encouraging results. 32 Additionally, there are clinical trials using the antimalarial drugs hydroxychloroquine (HCQ) and chloroquine to treat COVID‐19 patients. Many studies have published in vitro data discussing their effects on viral inhibition. 31 , 33 Interestingly, antimalarial drugs have been shown to inhibit multiple cytokines including IL‐6 34 and are speculated to have vascular protective effects to prevent thrombotic complications. 35 These data suggests that HCQ and chloroquine may exhibit broad‐spectrum effects as antivirals, while simultaneously decreasing a proposed marker of severity and other clinically observed complications. The clinical data for HCQ from early trials for COVID‐19 have been mixed, with some studies showing encouraging results and others demonstrating lack of efficacy. 36 , 37 , 38 This is primarily due to limitations in study methodologies and small patient sample size. We eagerly await the results from larger‐scale confirmatory trials for HCQ and remdesivir. Potential inhibitors of cytokine storm have shown some clinical benefit in COVID‐19 patients and include agents that target cytokine release such the anti–IL‐6 antibody tocilizumab, the anti‐human granulocyte macrophage colony‐stimulating factor monoclonal antibody lenzilumab, or the Janus kinase 1/2 inhibitor ruxolitinib (with phase III trials initiated or being planned for COVID‐19). Sanders et al 39 provide a comprehensive review of the recent clinical experience for remdesivir, HCQ, and other therapeutics, including inhibitors of CRS that are currently under investigation as potential COVID‐19 treatments. The CDC clinical guidance currently has listed laboratory markers that are associated with increased illness severity. 40 These include lymphopenia, neutropenia, elevated serum alanine aminotransferase and aspartate aminotransferase, elevated lactate dehydrogenase, high C‐reactive protein, and high ferritin. Additionally, elevated d‐dimer and lymphopenia are also associated with mortality. Since high levels of inflammatory markers such as IL‐6 have been reported in patients experiencing COVID‐19 complications, we suggest using this potential clinical biomarker or any of the above laboratory markers as a guide to monitor patients on drug therapy. COVID‐19 treatment will involve a polypharmacy approach; we therefore expect a multitude of adverse effects from these drug combinations. Future studies will need to address the pharmacogenetics of these therapeutics and potential drug‐drug interactions and determine clinical biomarkers of response or in the management of adverse reactions. Future Studies on TMPRSS2 as a Predictive Biomarker of COVID‐19 Outcomes Many factors can be used to explain why some are at risk of more serious disease and many are currently being studied. These include socioeconomic status, access to health care, past medical history, and age. There are also many factors that can explain the difference in outcomes in men and women afflicted with COVID‐19, and TMPRSS2 emerges as an interesting candidate. We propose studies examining the differential expression of TMPRSS2 between male and female patients. Evidence of higher expression in the tissue of males can help inform further studies to correlate this expression to clinical outcomes. Moreover, genotyping studies performed on clinical specimens collected from patients on COVID‐19 trials are warranted to further evaluate known SNPs and to find additional functional polymorphisms in the TMPRSS2 gene. This will aid in understanding the impact of TMPRSS2 SNPs on disease susceptibility and its correlation with clinical outcomes, which may include assessing the severity of outcomes by organ systems to identify for sexual dimorphism. On a broader scope, multiple institutions have initiated genomic investigations in a project called the COVID Host Genetics Initiative. 41 Underlying genetic polymorphisms present a potential avenue as an actionable biomarker, enabling medical professionals to identify and treat those patients who will need an increased level of care vs those who do not need rigorous medical treatment when resources are scarce. Validated TMPRSS2 SNPs that are confirmed to be predictive biomarkers can be incorporated in the CDC's current list of clinical biomarkers for disease severity as discussed above. Furthermore, understanding the molecular differences that lead to changes in clinical outcomes, especially clear sexual dimorphisms, can help inform targeted drug therapy research and development. With the uncertainty currently surrounding the development of a vaccine for SARS‐CoV‐2, the discovery of pharmaceutical interventions is imperative, and understanding the genetics of the disease is equally as important. Funding This work was supported by the Intramural Research Program of the Center for Cancer Research, National Cancer Institute, National Institutes of Health. The content of this publication does not necessarily reflect the views or policies of the Department of Health and Human Services, nor does mention of trade names, commercial products, or organization imply endorsement by the US government. Conflicts of Interest The authors declare no conflicts of interest.

          Related collections

          Most cited references29

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

          Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: a retrospective cohort study

          Summary Background Since December, 2019, Wuhan, China, has experienced an outbreak of coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Epidemiological and clinical characteristics of patients with COVID-19 have been reported but risk factors for mortality and a detailed clinical course of illness, including viral shedding, have not been well described. Methods In this retrospective, multicentre cohort study, we included all adult inpatients (≥18 years old) with laboratory-confirmed COVID-19 from Jinyintan Hospital and Wuhan Pulmonary Hospital (Wuhan, China) who had been discharged or had died by Jan 31, 2020. Demographic, clinical, treatment, and laboratory data, including serial samples for viral RNA detection, were extracted from electronic medical records and compared between survivors and non-survivors. We used univariable and multivariable logistic regression methods to explore the risk factors associated with in-hospital death. Findings 191 patients (135 from Jinyintan Hospital and 56 from Wuhan Pulmonary Hospital) were included in this study, of whom 137 were discharged and 54 died in hospital. 91 (48%) patients had a comorbidity, with hypertension being the most common (58 [30%] patients), followed by diabetes (36 [19%] patients) and coronary heart disease (15 [8%] patients). Multivariable regression showed increasing odds of in-hospital death associated with older age (odds ratio 1·10, 95% CI 1·03–1·17, per year increase; p=0·0043), higher Sequential Organ Failure Assessment (SOFA) score (5·65, 2·61–12·23; p<0·0001), and d-dimer greater than 1 μg/mL (18·42, 2·64–128·55; p=0·0033) on admission. Median duration of viral shedding was 20·0 days (IQR 17·0–24·0) in survivors, but SARS-CoV-2 was detectable until death in non-survivors. The longest observed duration of viral shedding in survivors was 37 days. Interpretation The potential risk factors of older age, high SOFA score, and d-dimer greater than 1 μg/mL could help clinicians to identify patients with poor prognosis at an early stage. Prolonged viral shedding provides the rationale for a strategy of isolation of infected patients and optimal antiviral interventions in the future. Funding Chinese Academy of Medical Sciences Innovation Fund for Medical Sciences; National Science Grant for Distinguished Young Scholars; National Key Research and Development Program of China; The Beijing Science and Technology Project; and Major Projects of National Science and Technology on New Drug Creation and Development.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor

            Summary The recent emergence of the novel, pathogenic SARS-coronavirus 2 (SARS-CoV-2) in China and its rapid national and international spread pose a global health emergency. Cell entry of coronaviruses depends on binding of the viral spike (S) proteins to cellular receptors and on S protein priming by host cell proteases. Unravelling which cellular factors are used by SARS-CoV-2 for entry might provide insights into viral transmission and reveal therapeutic targets. Here, we demonstrate that SARS-CoV-2 uses the SARS-CoV receptor ACE2 for entry and the serine protease TMPRSS2 for S protein priming. A TMPRSS2 inhibitor approved for clinical use blocked entry and might constitute a treatment option. Finally, we show that the sera from convalescent SARS patients cross-neutralized SARS-2-S-driven entry. Our results reveal important commonalities between SARS-CoV-2 and SARS-CoV infection and identify a potential target for antiviral intervention.
              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
                figgw@mail.nih.gov
                Journal
                J Clin Pharmacol
                J Clin Pharmacol
                10.1002/(ISSN)1552-4604
                JCPH
                Journal of Clinical Pharmacology
                John Wiley and Sons Inc. (Hoboken )
                0091-2700
                1552-4604
                21 May 2020
                : 10.1002/jcph.1641
                Affiliations
                [ 1 ] Genitourinary Malignancies Branch Center for Cancer Research National Cancer Institute National Institutes of Health Bethesda Maryland USA
                Author notes
                [*] [* ] Corresponding Author:

                William D. Figg, PharmD, Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Building 10/Room 5A03, Bethesda, MD 20892

                Email: figgw@ 123456mail.nih.gov

                [†]

                Both authors contributed equally to this work.

                Article
                JCPH1641
                10.1002/jcph.1641
                7280622
                32437018
                5542b3b6-c3ce-4eaf-aaa7-b31ab746d93d
                Published 2020. This article is a U.S. Government work and is in the public domain in the USA. The Journal of Clinical Pharmacology published by Wiley Periodicals LLC on behalf of American College of Clinical Pharmacology

                This is an open access article under the terms of the http://creativecommons.org/licenses/by-nc-nd/4.0/ License, which permits use and distribution in any medium, provided the original work is properly cited, the use is non‐commercial and no modifications or adaptations are made.

                History
                : 13 April 2020
                : 17 April 2020
                Page count
                Figures: 1, Tables: 1, Pages: 7, Words: 4402
                Funding
                Funded by: National Cancer Institute , open-funder-registry 10.13039/100000054;
                Categories
                Commentary
                Commentary
                Custom metadata
                2.0
                corrected-proof
                Converter:WILEY_ML3GV2_TO_JATSPMC version:5.8.4 mode:remove_FC converted:09.06.2020

                biomarker,coronavirus,covid‐19,sars‐cov‐2,tmprss2
                biomarker, coronavirus, covid‐19, sars‐cov‐2, tmprss2

                Comments

                Comment on this article