Systematic review and patient-level meta-analysis of SARS-CoV-2 viral dynamics to model response to antiviral therapies ======================================================================================================================= * Silke Gastine * Juanita Pang * Florencia A.T. Boshier * Simon J. Carter * Dagan O. Lonsdale * Mario Cortina-Borja * Ivan F.N. Hung * Judy Breuer * Frank Kloprogge * Joseph F. Standing ## SUMMARY **Background** SARS-CoV-2 viral loads change rapidly following symptom onset so to assess antivirals it is important to understand the natural history and patient factors influencing this. We undertook an individual patient-level meta-analysis of SARS-CoV-2 viral dynamics in humans to describe viral dynamics and estimate the effects of antivirals used to-date. **Methods** This systematic review identified case reports, case series and clinical trial data from publications between 1/1/2020 and 31/5/2020 following PRISMA guidelines. A multivariable Cox proportional hazards regression model (Cox-PH) of time to viral clearance was fitted to respiratory and stool samples. A simplified four parameter nonlinear mixed-effects (NLME) model was fitted to viral load trajectories in all sampling sites and covariate modelling of respiratory viral dynamics was performed to quantify time dependent drug effects. **Findings** Patient-level data from 645 individuals (age 1 month-100 years) with 6316 viral loads were extracted. Model-based simulations of viral load trajectories in samples from the upper and lower respiratory tract, stool, blood, urine, ocular secretions and breast milk were generated. Cox-PH modelling showed longer time to viral clearance in older patients, males and those with more severe disease. Remdesivir was associated with faster viral clearance (adjusted hazard ratio (AHR) = 9·19, p< 0·001), as well as interferon, particularly when combined with ribavirin (AHR = 2·2, *p* = 0·015; AHR = 6·04, *p* = 0·006). **Interpretation** Combination therapy including interferons should be further investigated. A NLME model for designing and analysing viral pharmacodynamics in trials has been established. **Evidence before this study** We performed a systematic literature search for studies reporting serial viral load measurements in humans for SARS-CoV-2 in MEDLINE and Embase, MedRxiv and BioRxiv along with the PROSPERO systematic review register. As of 27/5/20, no previous or ongoing review has systematically searched for and pooled individual patient-level serial viral load data in this disease or depicted SARS-CoV-2 viral load trajectories in a meta-analysis. In addition, modelling of SARS-CoV-2 viral dynamics and model-based quantification and simulation of antiviral drug activity is to date only available from single studies. A comprehensive dataset for distinguishing between natural history of the disease and drug effect related viral load decline was therefore lacking. **Added value of this study** This individual patient meta-analysis includes a large dataset of individual patient serial viral load measurements for SARS-CoV-2 with and without drug therapy. The full dataset contains 645 individuals with 6316 viral load samples. The majority were sampled from the upper respiratory tract. There were also samples available from the lower respiratory tract, blood, urine and stool, breast milk and ocular secretions. This enabled us to estimate parameters of a nonlinear mixed effects (NLME) viral dynamics model to characterise the course of viral shedding across each sample site. A multivariable Cox proportional hazards regression analysis revealed increasing age, disease severity and male sex to be associated with longer time to viral clearance, indicating that patients with worse clinical outcomes do have higher viral loads. A NLME model incorporating drug effects showed early initiation of antivirals shortened time to viral clearance. By including data on multiple drug combinations Cox proportional hazards suggests type I interferons plus ribavirin to be potentially synergistic *in vivo*. Our NLME model can be used to determine the sample size of future Phase II trials. For example, to detect a significant difference in proportion of patients clearing the virus after 7 days of treatment in patients starting therapy at day 3 post symptom onset with 90% power, one would require 606, 284, 90 or 31 patients per group for lopinavir/ritonavir, ribavirin, interferon or interferon plus ribavirin drug effects respectively. The later that antivirals are initiated, the smaller the drug effect and hence the larger sample size required to determine antiviral activity. **Implications of all the available evidence** This individual patient level meta-analysis provides insight into the natural history of the SARS-CoV-2 viral dynamics through the biggest serial viral load data set available to date. A NLME viral dynamics model was established enabling estimation of the impact of drug effects on viral clearance. The full data set and model code is available to as a tool to design and analyse Phase II antiviral trials. Key words * SARS-CoV-2 * COVID-19 * viral dynamics * pharmacodynamics ## INTRODUCTION Finding antivirals that target severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) will be crucial in managing the ongoing pandemic. In addition to the development of novel agents, substantial efforts are underway to establish whether currently available agents may be re-purposed1. A key biomarker for clinical antiviral activity is viral load in bodily fluids and assessing a drug’s or drug combination’s ability to reduce viral load is an important first step in identifying therapies that influence clinical outcome. To correctly assess antiviral activity, it is first necessary to understand viral load natural history. As a rapidly progressing, primarily respiratory viral infection, SARS-CoV-2 elimination from the body seems to be mainly driven by a combination of innate immune response and exhaustion of target cells available for infection2. Observational cohort studies published to date have shown that the rate of viral load decline seems slower in older patients, those with more severe disease and those with comorbidities such as diabetes mellitus and immunosuppression3–6. Interpreting these observational studies requires caution because patients have often received antiviral therapies. Due to the time point of initial infection being unknown, assessing viral load in response to treatment must account for time since symptom onset7. Since February 2020 case reports and case series of patient-level viral dynamics have been published, some of which report dosing of antiviral drugs8. Clinical trials of antivirals and their association with viral load are also beginning to read out9. Meanwhile large pragmatic trials of repurposed monotherapy antivirals have yet to find a clearly effective agent10. At this crucial juncture, it is vital to develop a pharmacodynamic modelling framework that can be used to describe the natural history of SARS-CoV-2 viral dynamics, make initial estimates on antiviral efficacy of agents used to-date, and to design and evaluate Phase II trials using viral load as a biomarker. This systematic review therefore aimed to search for case reports, case series and clinical trials reporting serial individual patient-level SARS-CoV-2 viral load measurements in humans from any sampling site upon which an individual patient-level meta-analysis was then performed. A nonlinear mixed effects (NLME) viral dynamic model was fitted to describe the viral trajectories in each sampling site and to give a quantitative measure of viral dynamics. In data of sufficient quality, the parameters of multivariable Cox proportional hazards regression models of time to viral clearance, and NLME models of antiviral efficacy were estimated. ## METHODS ### Protocol and registration The protocol for this systematic review and individual patient meta-analysis, which follows the PRISMA Individual Patient Data systematic reviews protocol11, was first published on 27/5/2020 at: [https://github.com/ucl-pharmacometrics/SARS-CoV-2-viral-dynamic-meta-analysis](https://github.com/ucl-pharmacometrics/SARS-CoV-2-viral-dynamic-meta-analysis). The final dataset and statistical analysis code are also published here. The review was registered with PROSPERO (CRD42020189000). ### Eligibility criteria This study aimed to identify serial viral loads with time in human subjects infected with SARS-CoV-2 in order to describe and model viral load trajectory. The inclusion criteria were therefore papers containing individual subject-level reports of viral load with time, either since symptom onset or time since start of monitoring for asymptomatic subjects, and sampling site. Authors of manuscripts describing summary statistics of viral load with time were contacted requesting participant level data. Viral load was defined as either a value in copies/mL or a cycle threshold (Ct) value of an uncalibrated polymerase chain reaction (PCR) assay. ### Overall search strategy Since SARS-CoV-2 was notified to the WHO on 31/12/2019, we did not expect to find relevant papers published prior to this date. Hence, PubMed, EMBASE, medRxiv, and bioRxiv were searched with a date range of 1/1/2020 to 31/5/2020. The following search terms were used for PubMed and EMBASE: (SARS-CoV-2 OR COVID OR coronavirus OR 2019-nCoV) AND (viral load OR cycle threshold OR rtPCR OR real-time PCR OR viral kinetics OR viral dynamics OR shedding OR detection OR clinical trial). Due to character limits in the search engine, the following search terms were used for medRxiv and bioRxiv: (SARS-CoV-2 OR COVID-19 OR coronavirus) AND (viral load OR cycle threshold OR PCR OR viral dynamics OR clinical trial). After removing duplicates, two reviewers independently identified papers for full text screening, with any discrepancies resolved by a third reviewer. ### Data extraction Viral loads were reported as either numerical values in tables, figures, or in viral load versus time plots. Where possible, numerical values were copy-pasted directly into a comma separated value (csv) format from the source, whereas tabulated numerical values contained in pdf images were extracted using [https://extracttable.com/](https://extracttable.com/). Viral loads reported in plots were extracted using Web Plot Digitizer12. Each viral load was paired with a time since symptom onset or in asymptomatic subjects, the time since viral monitoring started. Furthermore, sampling site and, if viral load not reported in copies/mL, the PCR assay including the primers used, were extracted along with limit of quantification and limit of detection, if available. The following patient-level covariates were extracted if available: * Presence of fever > 37·5 °C at any time (non-time varying covariate) * age, where possible individual age but otherwise the study’s reported central measure (e.g. mean, median) * sex or the male/female ratio was extracted if patient-level data not reported * need for and days of intensive care treatment * need for and days of mechanical ventilation * whether patient died and time to death from symptom onset. In addition, a standardised disease score was constructed for each patient as follows: * 0-asymptomatic * 1-mild disease (fever, cough or other mild symptoms reported) * 2-moderate disease (in addition to mild criterion: need for supplemental oxygen /non-invasive ventilation) * 3-severe disease (requirement for mechanical ventilation) All data were stored on a shared github repository, and standardised R-scripts took data from each paper to merge into a single master dataset. A quality control (QC) check on viral load values and all covariates was performed for each paper by an independent reviewer. ### Data quality assessment #### Viral load quality score Two quality assessments were applied to each dataset. Firstly, the quality of viral load reporting was rated on a 1–3 scale. The highest quality 1 was assigned to studies reporting viral load in copies/mL or reporting a calibration curve allowing for direct conversion of Ct values to viral load. Quality 2 was assigned if viral load was reported in PCR Ct and primers used in the assay were reported, but calibration data was missing. In this case a published calibration curve for that primer from another source was used to convert to viral load in copies/mL13,14. Where more than one calibration curve was available for the same primer the mean slope and intercept was used. The lowest (quality score 3) was assigned when viral load was reported in PCR Ct but no further information was available on the PCR assay. In this instance a conversion to copies/mL was made using the mean slope and intercepts from all calibration curves. #### Drug quality score The second quality assessment on a 3-point scale related to reporting of the antiviral drug therapy administered: which drug(s) and upon which days did patients receive the drug(s). The highest quality 1 was assigned when it was reported which days each patient received each drug, or these data were provided by corresponding authors. If it was reported that no antiviral was administered this was also assigned quality 1. Quality 2 was assigned when antiviral drug treatment was reported, but ascertaining which days the patient had received the drugs was not possible. The lowest category, quality 3, was assigned when it was not possible to determine whether or not antivirals had been administered. ### Statistical analysis #### Primary analysis of time to viral clearance using Cox proportional hazards modelling The primary analysis was conducted on observed time to viral clearance, which was analysed fitting Cox proportional hazards regression models with adjusted hazard ratios estimated for each covariate. We verified the assumptions of proportional hazards using the Therneau-Grambsch test.15 The data used for this analysis were limited to respiratory and stool sampling sites only, as virus was found to be mostly undetectable at other sites. Furthermore, only data from patients with known antiviral history (drug quality 1 and 2) were used. To assess the possible risk of bias in different drug and viral load qualities, the analysis was repeated on two further subsets: Firstly, with only drug quality 1 and respiratory samples, and secondly on assay quality 1 data only. Time to viral load dropping below the limit of detection was modelled with Cox proportional hazards regression in R (version 3·6·3)16. Where a single patient contributed samples from multiple sampling sites (e.g. upper respiratory and stool), the time to the last site testing negative was used. Multivariable models for covariate effects on time to viral clearance were fitted, with additional interaction terms for drug therapies included, where multiple antiviral agents were given simultaneously. In studies reporting sex as a proportion of males, 10 000 datasets were simulated using the reported fraction of males to randomly assign individuals to being male from the binomial distribution. The Cox proportional hazards regression model was then fitted to each dataset and parameter estimates compared with the model, where individual sex was assigned by rounding the fraction of males. Model parameter estimates were visualised using forest plots. #### Secondary analysis antiviral pharmacology model The secondary analysis was to use a NLME model to quantify the increase in viral elimination rate with antiviral therapy. This analysis used data only from respiratory samples and rated drug quality 1. ##### Nonlinear mixed-effects (NLME) viral dynamic model Firstly, a descriptive analysis of all data was undertaken. A NLME viral dynamics model was fitted to the individual patient-level viral load versus time data. The structural model was based on the general target cell limited model, which has previously been used to describe respiratory viral infections7,17. This model consists of three ordinary differential equations relating to changes in uninfected target cells (T), infected target cells (*I*) and free virus (*V*) over time (*t*), as follows: ![Formula][1] ![Formula][2] ![Formula][3] where *β* is the rate at which target cells become infected in the presence of virus, *δ* is the death rate of infected cells, *ρ* is the rate of viral production from infected cells and *c* is the rate of clearance of free virus. This model is structurally unidentifiable, as tested through the *IdentifiabiltyAnalysis* package in Wolfram Mathematica 12·1 (Wolfram Research, Illinois, USA)18, unless the initial condition for *T, β*, or *ρ* are known. Furthermore, the elimination rate of free virus (*c*) is likely to be much faster than the death rate of infected cells (*δ*). Hence, by assuming a quasi-steady-state between *I* and *V*, and normalising the total cell number by the number of infected cells when observations begin (*t* = 0), it is then possible to reduce the model to a structurally identifiable, two state ordinary differential equation model relating to the fraction (*f*) of infected cells with time and infected cells as a proxy for viral load as follows19: ![Formula][4] ![Formula][5] with *γ*, a new parameter equal to *ρβT/c* and interpreted to be the maximum rate of viral replication. *δ* can now be interpreted as overall viral elimination rate. This population model was then fitted to viral load data with time using the following form: ![Formula][6] where *yij* was the viral load from subject *i* at time *tij, f* is the nonlinear model defined above with parameters *φi*, and *εij* the residual between the model prediction and the observed data. Four parameters were estimated: the initial viral load at symptom onset (*V*), *β, δ* and *γ*. Interindividual variability was estimated for *V*, *β* and *δ* with each assumed to follow a log-normal distribution. Viral loads were log transformed and the residual error was assumed to follow a normal distribution. Parameter estimation by maximum likelihood was undertaken using the stochastic approximation expectation maximization (SAEM) in NONMEM version 7·420. Model evaluation was undertaken by analysis of normalised prediction distribution errors (NPDE) and visual predictive checks (VPC)21. Viral loads below the limit of detection (LOD) were included by integrating the density function from minus infinity to the limit of detection to yield a probability of the data being below the LOD (“M3 Method”)22. In some participants, multiple samples were taken at the same time point (either different sampling site or the same sample assayed by more than one method). In this case a common residual error term was used to allow for modelling one-level nested random effects. ##### Descriptive analysis of viral shedding by sample site The above model was fitted to data from each sampling site. The resulting parameters were then used to simulate the overall population viral load trajectories. For the respiratory sample sites viral area under the curve (AUC), peak viral load and half-life were derived from the model and plotted versus patient covariates. ##### Covariate analysis and antiviral drug effects modelling The initial model used only data obtained in untreated patients. A covariate analysis was undertaken testing the influence of sampling site (nasal versus oral versus lower respiratory tract), sex, age and disease status on either *V, β* or *δ*. Covariates were retained in the model based on the likelihood ratio test with a threshold level of significance of *p*< 0·01, and if the same covariate addition to *V, β* or *δ* all gave significant improvement to model fit then the model with the largest decrease in –2 log likelihood was chosen. For the final model viral area under the curve (AUC), peak viral load and half-life were derived and plotted versus patient covariates. Using the final demographic model, data from patients undergoing antiviral treatment (antiviral drug quality 1) were added. A univariable analysis was performed, testing each drug’s ability to increase *δ*. Drugs showing significant improvement in model fit (p< 0·01), according to the likelihood ratio test, were then included in the final multivariable model. ### Simulations based on the antiviral pharmacology model Simulations were performed to explore the change in viral trajectories for different time points of therapy initiation: Day 1 after symptom onset, Day 3, Day 7 and Day 10. Interferon, lopinavir/ritonavir and ribavirin monotherapy along with the following combination therapies: interferon plus ribavirin, lopinavir/ritonavir plus ribavirin and the interferon plus lopinavir/ritonavir and plus ribavirin were explored this way. A dummy population of 5100 subjects with ages uniformly distributed across 50 to 100 years, consisting of an equal ratio of males and females was created. Each regimen was simulated using the entire population, assuming sampling from the upper respiratory tract or nose for a time window of 14 days. Comparisons of the sample size required to detect a significant difference in the proportion of undetectable virus between antiviral and no treatment were made after 7 days of treatment with a 90% power and alpha level of *p*< 0·05 for antivirals starting at Days 1, 3 and 7 post symptom onset. ## RESULTS Results of the systematic search are given in Figure 1, and details of included papers in Table 1. Individual patient-level data were extracted from 45 articles reporting viral loads and/or PCR Ct values with time since symptom onset. Of these 32 papers either reported antiviral participant-level drug histories, or these were provided by the corresponding author. The full dataset contained 645 individuals contributing 6316 viral load samples. The majority of samples (*n*) were taken from the respiratory tract: nasopharyngeal (315 individuals, *n* = 2208), oropharyngeal or saliva (381 individuals, *n* = 2144) and lower respiratory tract (81 individuals, *n* = 799). The other reported samples sites were stool/rectal swabs (99 individuals, *n* = 655), blood/plasma(42 individuals, *n* = 258), urine (31 individuals, *n* = 112), ocular (16 individuals, *n* = 50), breastmilk (4 individuals, *n* = 90). Metrics of the full data set are given in Supplementary Table S1. ![Figure 1:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2020/08/23/2020.08.20.20178699/F1.medium.gif) [Figure 1:](http://medrxiv.org/content/early/2020/08/23/2020.08.20.20178699/F1) Figure 1: PRISMA diagram detailing the systematic search results View this table: [Table 1](http://medrxiv.org/content/early/2020/08/23/2020.08.20.20178699/T1) Table 1 Individual papers included in the Meta-Analysis Full details of the extracted patient-level covariates are given in Table 2. Recording of fever, days on ICU and days ventilated was largely unavailable. Therefore, no further analysis was performed on these variables. However, it was possible to categorise disease status in all drug quality 1 and 2 papers, either through reports in the manuscript or by contacting corresponding authors. Overall, most patients had mild disease (376, 66·8%), whereas 79 (14·0%) patients had moderate and 84 (14·9%) severe disease. In total 24 (4·3%) asymptomatic patients were reported. View this table: [Table 2](http://medrxiv.org/content/early/2020/08/23/2020.08.20.20178699/T2) Table 2 Overview extracted variables across different analyses, median [range] (%missing data records). n, number of individuals included. The distribution of recorded drug therapies, available for drug quality 1 data and respiratory site samples, is summarised in Supplementary Table S2. 67 patients did not receive antivirals. The NLME model fits to the overall data, stratified by sampling site, are provided in Supplementary Table S3 and Supplementary Figure S1. Simulations from the models for each sampling site showing the expected viral load trajectory along with the predicted proportion of samples, that would be below the limit of detection are given in Figure 2. For respiratory sites model-derived AUC, peak viral load and half-life is given in Supplementary Figure S2 ![Figure 2:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2020/08/23/2020.08.20.20178699/F2.medium.gif) [Figure 2:](http://medrxiv.org/content/early/2020/08/23/2020.08.20.20178699/F2) Figure 2: Model-predicted viral load trajectories at each sample site studied. Black lines are the median predictions, with shaded areas representing the 95% prediction interval. The percentage of samples that are predicted to be below a typical limit of detection (10 copies/mL) are given in 2-daily time bins on each plot Data on a total of 354 patients with respiratory and/or stool/rectal sampling and drug quality 1 or 2 were available. A forest plot of the parameter estimates from the Cox proportional hazards regression model is provided in Figure 3. Viral clearance was fastest from upper respiratory tract samples and slowest from stool. More sensitive assays (with lower detection limits) were associated with longer time to viral clearance and viral clearance was faster in females, younger patients and those who were asymptomatic. ![Figure 3:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2020/08/23/2020.08.20.20178699/F3.medium.gif) [Figure 3:](http://medrxiv.org/content/early/2020/08/23/2020.08.20.20178699/F3) Figure 3: Multivariable Cox proportional hazard results on all drug quality 1 and drug quality 2 data from respiratory and stool/rectal sampling sites. Adjusted hazard ratios exceeding 1 indicate virus being more likely to become undetectable Regarding antiviral therapies, only remdesivir (adjusted hazard ratio (AHR) = 9·19, p< 0·001) and interferons (AHR = 2·20, p = 0·015) were independently associated with faster viral clearance. The effect of interferon alpha and beta (Supplementary Figure S3) was similar and hence these were combined. Lopinavir/ritonavir, ribavirin and interferons were most used and also most used in combination. Adding interaction terms for interferon plus lopinavir/ritonavir, interferon plus ribavirin and lopinavir/ritonavir plus ribavirin showed a trend towards synergy between interferons and ribavirin in the full dataset (AHR = 6·04, p = 0·006 Figure 3), as well as in the additional analysis taking in quality assessments to account for potential bias: respiratory data limited to drug quality 1 (Figure 4) and in data limited to only viral load quality 1 data (Supplementary Figure S4). ![Figure 4:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2020/08/23/2020.08.20.20178699/F4.medium.gif) [Figure 4:](http://medrxiv.org/content/early/2020/08/23/2020.08.20.20178699/F4) Figure 4: Multivariable Cox proportional hazard results on drug quality 1 data from respiratory sampling sites only. Adjusted hazard ratios exceeding 1 indicate virus being more likely to become undetectable Covariate relationships and drug effects were explored through NLME modelling with parameter estimates of the model given in Supplementary Table S4 along with visual predictive checks and NPDEs in Supplementary Figures S5 and FigureS6 and visualization of viral area under the curve, peak viral load and half-life derived from the final model in Supplementary Figure S7. Drug effects were estimated to increase δ. Drug regimens containing interferon (p< 0·001), lopinavir/ritonavir (p = 0·0016) and ribavirin (p< 0·001) each improved model fit and so were taken forward to the final multivariable drug model. The final model was then used to simulate expected viral trajectories from upper respiratory sampling sites for interferon, lopinavir/ritonavir and ribavirin monotherapy, interferon plus ribavirin as well as lopinavir/ritonavir plus ribavirin and the triple combination of interferon, lopinavir/rintonavir and ribavirin started at 1, 3, 7 and 10 days post symptom onset (Figure 5). The sample sizes for hypothetical Phase II trials to detect significant differences in viral load versus no treatment after 7 days of therapy are given in Supplementary Table S5. ![Figure 5:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2020/08/23/2020.08.20.20178699/F5.medium.gif) [Figure 5:](http://medrxiv.org/content/early/2020/08/23/2020.08.20.20178699/F5) Figure 5: Simulated viral load trajectories. Simulations with a dummy population equally distributed between 50 and 100 years, and equal male/female ratio were performed for each scenario. Drugs were started at day 1 (blue), day 3 (orange), day 7 (green) or day 10 (red) post symptom onset. Mean black line and error bars represent simulations of the dummy population without drug treatment. Coloured mean lines and error bars represent the respective drug regimen. Percentage values represent expected proportion of samples below the limit of detection for no drug (black) versus drug therapy (coloured) at each time point. ## DISCUSSION This systematic review and individual level meta-analysis has identified viral load trajectories from 645 individuals aged from the first month of life to 100 years. Data from all major sampling sites showed, that: following symptom onset in most patients, upper respiratory tract viral load has peaked and is declining, whereas in the lower respiratory tract viral load peaks 2–3 days after symptom onset; virus is detectable in stool for at least 2 weeks in 75% of individuals, and virus is detected in low levels in blood, urine, ocular secretions and breast milk (Figure 2). In addition to simulating the expected trajectory of viral load at each site, we were able to simulate the percentage of samples expected to be below a typical detection limit of 10 copies/mL (Figure 2). From this it can be seen, that from day 10 post symptom onset over a quarter of upper respiratory samples have undetectable viral load. This emphasises the importance of early antiviral therapy, and for Phase II trials using viral load as an endpoint to commence therapy in the first few days of symptom onset in order to reliably differentiate antiviral effects from natural viral decline (Figure 5, Supplementary Table S5). A heterogeneous range of antivirals, administered in different combinations, was observed in our data (Supplementary Table S2) meaning multivariable modelling of time to viral clearance was used to tease out individual drug effects. No antiviral activity was seen for chloroquine/hydroxychloroquine, azithromycin, lopinavir/ritonavir, umifenovir and thymalfasin. However, remdesivir and interferons were both independently associated with shorter time to viral clearance and combination of interferons with ribavirin also appeared to reduce viral load compared with untreated patients (Figures 3, 4 and S4). Remdesivir did not however significantly decrease δ in the NLME model, but this is likely due to the low number of included patients. Our most interesting finding is the promising antiviral activity of interferons, possibly due to low endogenous interferon levels induced by SARS-CoV-227,28. Interferons (alpha and beta) have shown extensive *in vitro* activity against Severe acute respiratory syndrome-1 (SARS-CoV-1) and Middle East respiratory syndrome coronavirus (MERS-CoV)23,24.However, this has not translated into clinical effectiveness in MERS-CoV23, although results from one trial are still pending25. Although recent data suggests interferon beta may be more potent than alpha against SARS-CoV-2 *in vitro*26, possibly due to higher selective indices for interferon-beta 1b, upon finding similar effects of interferon alpha and beta in our primary analysis (Figure S3), we decided to combine the interferon effect to better explore drug combinations. Consistent across data qualities and sampling site combinations, we found either a significant or trend towards significant synergistic activity of interferon plus ribavirin (Figures 3, 4, S4). An extensive body of literature exists to show both interferon alpha and beta are synergistic with ribavirin in vitro against both SARS-CoV-1 and MERS-CoV23, and we have now shown a signal towards this *in vivo* with SARS-CoV-2. Although ribavirin monotherapy was rarely used (two patients), the finding of ribavirin plus interferon synergy is likely to be robust, since we studied large numbers of lopinavir/ritonavir plus ribavirin, lopinavir/ritonavir alone, lopinavir/ritonavir plus interferon and triple therapy with ribavirin plus lopinavir/ritonavir plus interferon in addition to patients receiving no antivirals. Our result suggests, that combining interferons with a nucleoside analogue, possibly remdesivir or favipiravir as less toxic alternatives to ribavirin, is a potentially promising combination. In our secondary analysis, we included interferon plus ribavirin in the NLME model and simulations show that virus should be suppressed 2–3 days faster compared to no treatment (Figure 5). Another main finding of our work was the limited antiviral effect of lopinavir/ritonavir, in addition to its lack of significant synergistic effect with either ribavirin or interferons. The protease inhibitor lopinavir had a modest but consistent in vitro activity against the major coronaviruses, including SARS-CoV-2, although activity is confined to concentrations at the upper end of the clinically achievable range1, and our simulations suggest monotherapy studies would require well over 500 participants per arm just to show antiviral activity (Supplementary Table S5). In SARS-CoV-1 however, lopinavir/ritonavir plus ribavirin was found to be synergistic in vitro and when initiated immediately upon diagnosis led to a significant decrease in mortality compared to historical controls30,31. Early post-exposure prophylaxis against Middle East Respiratory Syndrome (MERS-CoV) in healthcare workers showed that lopinavir/ritonavir plus ribavirin reduced the incidence of infection from 28% to 0%32. The lopinavir/ritonavir plus ribavirin combination has therefore been the basis for many clinical trials and treatment protocols, but our findings suggest that it may not be as useful in SARS-CoV-2 (Figure 3). The antiviral effects of remdesivir in vitro are well established and despite only being able to extract individual patient-level data on six patients, it produced a significantly faster viral clearance in the primary analysis (Figure 3). Despite in some cases showing promising in vitro activity, we did not find significant antiviral effects of azithromycin, chloroquine/hydroxychloroquine, thymalfasin or umifenovir. In the case of hydroxychloroquine and azithromycin the raw viral load data from the heavily criticised study by Gautret et al33 was included, but contrary to the original analysis we found no clinical antiviral activity of either drug and, in the case of hydroxychloroquine, a trend towards slower viral clearance. The reason for this difference in interpretation appears to stem from using time since symptom onset as opposed to time since starting drug and with untreated patients being monitored from an earlier day-post symptom onset. This example highlights the necessity of accounting for the time course of the infection when analysing viral loads. In our secondary NLME analysis the simplified target cell limited model provided a good fit to data from each sampling site. In many cases this approximated a mono-exponential decay, but in others, particularly in lower respiratory tract, there was a pronounced peak in the first days following symptom onset. The model was stable with high inter-individual variability on V and β, reflecting the fact that relative changes in these parameters lead to the initial part of the curve either rising then falling (in situations when V ≈ –β) or approximately monoexponentially declining (when V(0) ≫ –β). In addition, we found the model to be less sensitive to changes in γ, meaning it can take a wide range of values with little influence on model fit, hence we did not estimate an inter-individual variability term on it. In contrast to authors who have estimated parameters for more mechanistic models34, we estimated all drug effects to increase δ, which implies a mode of action relating to inhibition of viral replication or stimulation of viral clearance mechanisms. Whilst for most of the drugs studied this may be reasonable, entry inhibitors may be more appropriately described by inhibition of γ, which may not be statistically identifiable with the data possible to collect in the clinical setting. Despite this potential limitation, we found similar agents (combinations including interferons, ribavirin and lopinavir/ritonavir) to those identified in the primary analysis of time-to viral clearance. The major limitation of our work is the lack of clinical trial data and lack of data on potentially important re-purposing agents such as favipiravir and nitazoxanide and that only one of the authors of a major clinical trial agreed to share their data9. Through applying quality assessment criteria on drug history and assay reporting, before undertaking Cox proportional hazards and NLME modelling we aimed to reduce possible bias in the heterogenous data available. In conclusion, this individual patient level meta-analysis has yielded useful insights into SARS-CoV2 viral dynamics. A model-based description of viral trajectories in different sampling sites has been elucidated, and we have found covariates such as increasing age, disease severity and male sex to be associated with slower viral clearance. Our review firmly establishes a role for early viral suppression in the management of SARS-CoV-2 and an important signal as to the possible benefits of interferons as a component of antiviral therapy has been found. It has been shown that viral dynamic models such as ours can increase the power to detect drug effects due to their utilisation of serial measures35 and our model should be useful to others in both the design and analysis of future Phase II trials, hence the model code and raw data from this analysis is made available. ## Data Availability The final dataset is available at github [https://github.com/ucl-pharmacometrics/SARS-CoV-2-viral-dynamic-meta-analysis](https://github.com/ucl-pharmacometrics/SARS-CoV-2-viral-dynamic-meta-analysis) ## CONTRIBUTOR’S STATEMENT Silke Gastine performed literature search, data interpretation, data analysis and manuscript writing. Juanita Pang performed literature search and data extraction Florencia A.T. Boshier performed literature search and data extraction Simon J. Carter performed data interpretation and data analysis Dagan O. Lonsdale supported data interpretation Mario Cortina-Borja supported data analysis Ivan F.N. Hung: performed data collection Judy Breuer: supported data interpretation Frank Kloprogge: supported data interpretation, data analysis Joseph F. Standing: performed study design, literature search, data extraction, data analysis, manuscript writing ## DATA AVAILABILITY The final dataset is available at: [https://github.com/ucl-pharmacometrics/SARS-CoV-2-viral-dynamic-meta-analysis](https://github.com/ucl-pharmacometrics/SARS-CoV-2-viral-dynamic-meta-analysis) ## CODE AVAILABILITY Model code is available at: [https://github.com/ucl-pharmacometrics/SARS-CoV-2-viral-dynamic-meta-analysis](https://github.com/ucl-pharmacometrics/SARS-CoV-2-viral-dynamic-meta-analysis) ## SOURCE OF FUNDING No specific funding was available for this work. Support at institution level came from the National Institute for Health Research Biomedical Research Centre at Great Ormond Street Hospital for Children NHS Foundation Trust and University College London, and J.F.S. and F.K. were supported by United Kingdom Medical Research Council (MRC) Fellowships (grants M008665 and P014534). JB receives funding from the NIHR UCL/UCLH Biomedical Research Centre. JP is supported by a Rosetrees Trust PhD fellowship (M876). FTB is supported by a Wellcome Trust Collaborative Award (203268) to JB. ## DISCLOSURE OF CONFLICT OF INTEREST None * Received August 20, 2020. * Revision received August 20, 2020. * Accepted August 23, 2020. * © 2020, Posted by Cold Spring Harbor Laboratory This pre-print is available under a Creative Commons License (Attribution-NoDerivs 4.0 International), CC BY-ND 4.0, as described at [http://creativecommons.org/licenses/by-nd/4.0/](http://creativecommons.org/licenses/by-nd/4.0/) ## REFERENCES 1. 1.Arshad U, Pertinez H, Box H, et al. Prioritization of Anti-SARS-Cov-2 Drug Repurposing Opportunities Based on Plasma and Target Site Concentrations Derived from their Established Human Pharmacokinetics. Clin Pharmacol Ther 2020. 2. 2.To KK, Tsang OT, Leung WS, et al. Temporal profiles of viral load in posterior oropharyngeal saliva samples and serum antibody responses during infection by SARS-CoV-2: an observational cohort study. Lancet Infect Dis 2020; 20(5): 565–74. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S1473-3099(20)30196-1&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F23%2F2020.08.20.20178699.atom) 3. 3.He X, Lau EHY, Wu P, et al. Temporal dynamics in viral shedding and transmissibility of COVID-19. Nat Med 2020; 26(5): 672–5. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.7326/M20-3012&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F23%2F2020.08.20.20178699.atom) 4. 4.Rajpal A, Rahimi L, Ismail-Beigi F. Factors Leading to High Morbidity and Mortality of COVID-19 in Patients with Type 2 Diabetes. J Diabetes 2020. 5. 5.Fishman JA, Grossi PA. Novel Coronavirus-19 (COVID-19) in the immunocompromised transplant recipient: #Flatteningthecurve. Am J Transplant 2020; 20(7): 1765–7. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=32233057&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F23%2F2020.08.20.20178699.atom) 6. 6.Zheng S, Fan J, Yu F, et al. Viral load dynamics and disease severity in patients infected with SARS-CoV-2 in Zhejiang province, China, January-March 2020: retrospective cohort study. BMJ 2020; 369: m1443. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiYm1qIjtzOjU6InJlc2lkIjtzOjE4OiIzNjkvYXByMjFfMjAvbTE0NDMiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMC8wOC8yMy8yMDIwLjA4LjIwLjIwMTc4Njk5LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 7. 7.Kamal MA, Gieschke R, Lemenuel-Diot A, Beauchemin CA, Smith PF, Rayner CR. A drug-disease model describing the effect of oseltamivir neuraminidase inhibition on influenza virus progression. Antimicrob Agents Chemother 2015; 59(9): 5388–95. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiYWFjIjtzOjU6InJlc2lkIjtzOjk6IjU5LzkvNTM4OCI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIwLzA4LzIzLzIwMjAuMDguMjAuMjAxNzg2OTkuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 8. 8.Kim JY, Ko JH, Kim Y, et al. Viral Load Kinetics of SARS-CoV-2 Infection in First Two Patients in Korea. J Korean Med Sci 2020; 35(7): e86. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3346/jkms.2020.35.e86&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F23%2F2020.08.20.20178699.atom) 9. 9.Hung IF, Lung KC, Tso EY, et al. Triple combination of interferon beta-1b, lopinavir-ritonavir, and ribavirin in the treatment of patients admitted to hospital with COVID-19: an open-label, randomised, phase 2 trial. Lancet 2020; 395(10238): 1695–704. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S0140-6736(20)31042-4&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=32401715&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F23%2F2020.08.20.20178699.atom) 10. 10.Recovery trial. Statement from the Chief Investigators of the Randomised Evaluation of COVid-19 thERapY (RECOVERY) Trial on lopinavir-ritonavir. 2020. [https://www.recoverytrial.net/files/lopinavir-ritonavir-recovery-statement-29062020_final.pdf](https://www.recoverytrial.net/files/lopinavir-ritonavir-recovery-statement-29062020_final.pdf) (accessed 29.06.2020. 11. 11.Stewart LA, Clarke M, Rovers M, et al. Preferred Reporting Items for Systematic Review and Meta-Analyses of individual participant data: the PRISMA-IPD Statement. JAMA 2015; 313(16): 1657–65. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1001/jama.2015.3656&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25919529&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F23%2F2020.08.20.20178699.atom) 12. 12.Rohatgi A. Webplotdigitizer: Version 4.2. 2019. [https://automeris.io/WebPlotDigitizer](https://automeris.io/WebPlotDigitizer). 13. 13.Vogels CBF, Brito AF, Wyllie AL, et al. Analytical sensitivity and efficiency comparisons of SARS-COV-2 qRT-PCR primer-probe sets. medRxiv 2020: 2020.03.30.20048108. 14. 14.Chu DKW, Pan Y, Cheng SMS, et al. Molecular Diagnosis of a Novel Coronavirus (2019-nCoV) Causing an Outbreak of Pneumonia. Clin Chem 2020; 66(4): 549–55. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/clinchem/hvaa029&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F23%2F2020.08.20.20178699.atom) 15. 15.Grambsch PM, Therneau TM. Proportional hazards tests and diagnostics based on weighted residuals. Biometrika 1994; 81(3): 515–26. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/biomet/81.3.515&link_type=DOI) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1994PP36700006&link_type=ISI) 16. 16.R Core Team. A language and environment for statistical computing (version 3.6.3). R Foundation for Statistical Computing, Vienna, Austria. 2019. 17. 17.Baccam P, Beauchemin C, Macken CA, Hayden FG, Perelson AS. Kinetics of influenza A virus infection in humans. J Virol 2006; 80(15): 7590–9. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoianZpIjtzOjU6InJlc2lkIjtzOjEwOiI4MC8xNS83NTkwIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjAvMDgvMjMvMjAyMC4wOC4yMC4yMDE3ODY5OS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 18. 18.Anguelova M, Karlsson J, Jirstrand M. Minimal output sets for identifiability. Math Biosci 2012; 239(1): 139–53. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.mbs.2012.04.005&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22609467&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F23%2F2020.08.20.20178699.atom) 19. 19.Kim KS, Ejima K, Ito Y, et al. Modelling SARS-CoV-2 Dynamics: Implications for Therapy. medRxiv 2020: 2020.03.23.20040493. 20. 20.Beal SLS, L.B.; Boeckmann, A.J.; Bauer, R.J. NONMEM 7.4 users guides. (1989 –2018). doi:10.1128/AAC.00069-15. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiYWFjIjtzOjU6InJlc2lkIjtzOjk6IjU5LzkvNTM4OCI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIwLzA4LzIzLzIwMjAuMDguMjAuMjAxNzg2OTkuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 21. 21.Nguyen TH, Mouksassi MS, Holford N, et al. Model Evaluation of Continuous Data Pharmacometric Models: Metrics and Graphics. CPT Pharmacometrics Syst Pharmacol 2017; 6(2): 87–109. 22. 22.Beal SL. Ways to Fit a PK Model with Some Data Below the Quantification Limit. Journal of Pharmacokinetics and Pharmacodynamics 2001; 28(5): 481–504. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1023/A:1012299115260&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=11768292&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F23%2F2020.08.20.20178699.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000172744200004&link_type=ISI) 23. 23.Zeitlinger M, Koch BCP, Bruggemann R, et al. Pharmacokinetics/Pharmacodynamics of Antiviral Agents Used to Treat SARS-CoV-2 and Their Potential Interaction with Drugs and Other Supportive Measures: A Comprehensive Review by the PK/PD of Anti-Infectives Study Group of the European Society of Antimicrobial Agents. Clin Pharmacokinet 2020. 24. 24.Sheahan TP, Sims AC, Leist SR, et al. Comparative therapeutic efficacy of remdesivir and combination lopinavir, ritonavir, and interferon beta against MERS-CoV. Nat Commun 2020; 11(1): 222. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41467-019-13940-6&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=31924756&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F23%2F2020.08.20.20178699.atom) 25. 25.Arabi YM, Alothman A, Balkhy HH, et al. Treatment of Middle East Respiratory Syndrome with a combination of lopinavir-ritonavir and interferon-beta1b (MIRACLE trial): study protocol for a randomized controlled trial. Trials 2018; 19(1): 81. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/s13063-017-2427-0&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F23%2F2020.08.20.20178699.atom) 26. 26.Yuan S, Chan CC, Chik KK, et al. Broad-Spectrum Host-Based Antivirals Targeting the Interferon and Lipogenesis Pathways as Potential Treatment Options for the Pandemic Coronavirus Disease 2019 (COVID-19). Viruses 2020; 12(6). 27. 27.Blanco-Melo D, Nilsson-Payant BE, Liu WC, et al. Imbalanced Host Response to SARS-CoV-2 Drives Development of COVID-19. Cell 2020; 181(5): 1036–45 e9. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.cell.2020.04.026&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F23%2F2020.08.20.20178699.atom) 28. 28.Chu H, Chan JF, Wang Y, et al. Comparative replication and immune activation profiles of SARS-CoV-2 and SARS-CoV in human lungs: an ex vivo study with implications for the pathogenesis of COVID-19. Clin Infect Dis 2020. 29. 29.Cao B, Wang Y, Wen D, et al. A Trial of Lopinavir-Ritonavir in Adults Hospitalized with Severe Covid-19. N Engl J Med 2020; 382(19): 1787–99. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F23%2F2020.08.20.20178699.atom) 30. 30.Chu CM, Cheng VC, Hung IF, et al. Role of lopinavir/ritonavir in the treatment of SARS: initial virological and clinical findings. Thorax 2004; 59(3): 252–6. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6OToidGhvcmF4am5sIjtzOjU6InJlc2lkIjtzOjg6IjU5LzMvMjUyIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjAvMDgvMjMvMjAyMC4wOC4yMC4yMDE3ODY5OS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 31. 31.Chan KS, Lai ST, Chu CM, et al. Treatment of severe acute respiratory syndrome with lopinavir/ritonavir: a multicentre retrospective matched cohort study. Hong Kong Med J 2003; 9(6): 399–406. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=14660806&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F23%2F2020.08.20.20178699.atom) 32. 32.Park SY, Lee JS, Son JS, et al. Post-exposure prophylaxis for Middle East respiratory syndrome in healthcare workers. J Hosp Infect 2019; 101(1): 42–6. 33. 33.Gautret P, Lagier JC, Parola P, 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(1): 105949. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F23%2F2020.08.20.20178699.atom) 34. 34.Goncalves A, Bertrand J, Ke R, et al. Timing of antiviral treatment initiation is critical to reduce SARSCoV-2 viral load. CPT Pharmacometrics Syst Pharmacol 2020. 35. 35.Laouenan C, Guedj J, Mentre F. Clinical trial simulation to evaluate power to compare the antiviral effectiveness of two hepatitis C protease inhibitors using nonlinear mixed effect models: a viral kinetic approach. BMC Med Res Methodol 2013; 13: 60. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/1471-2288-13-60&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23617810&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F23%2F2020.08.20.20178699.atom) 36. 36.Kam KQ, Yung CF, Cui L, et al. A Well Infant With Coronavirus Disease 2019 With High Viral Load. Clin Infect Dis 2020; 71(15): 847–9. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F23%2F2020.08.20.20178699.atom) 37. 37.Liu Y, Yang Y, Zhang C, et al. Clinical and biochemical indexes from 2019-nCoV infected patients linked to viral loads and lung injury. Sci China Life Sci 2020; 63(3): 364–74. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s11427-020-1643-8&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=32048163&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F23%2F2020.08.20.20178699.atom) 38. 38.Lim J, Jeon S, Shin HY, et al. Case of the Index Patient Who Caused Tertiary Transmission of COVID-19 Infection in Korea: the Application of Lopinavir/Ritonavir for the Treatment of COVID-19 Infected Pneumonia Monitored by Quantitative RT-PCR. J Korean Med Sci 2020; 35(6): e79. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3346/jkms.2020.35.e79&link_type=DOI) 39. 39.Pan Y, Zhang D, Yang P, Poon LLM, Wang Q. Viral load of SARS-CoV-2 in clinical samples. Lancet Infect Dis 2020; 20(4): 411–2. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S1473-3099(20)30113-4&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=32105638&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F23%2F2020.08.20.20178699.atom) 40. 40.Zhang W, Du RH, Li B, et al. Molecular and serological investigation of 2019-nCoV infected patients: implication of multiple shedding routes. Emerg Microbes Infect 2020; 9(1): 386–9. 41. 41.Xu T, Chen C, Zhu Z, et al. Clinical features and dynamics of viral load in imported and non-imported patients with COVID-19. Int J Infect Dis 2020; 94:68–71. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.ijid.2020.03.022&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=32179140&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F23%2F2020.08.20.20178699.atom) 42. 42.Chen W, Lan Y, Yuan X, et al. Detectable 2019-nCoV viral RNA in blood is a strong indicator for the further clinical severity. Emerg Microbes Infect 2020; 9(1): 469–73. 43. 43.Young BE, Ong SWX, Kalimuddin S, et al. Epidemiologic Features and Clinical Course of Patients Infected With SARS-CoV-2 in Singapore. JAMA 2020. 44. 44.Shen C, Wang Z, Zhao F, et al. Treatment of 5 Critically Ill Patients With COVID-19 With Convalescent Plasma. JAMA 2020. 45. 45.Wan R, Mao ZQ, He LY, Hu YC, Wei C. Evidence from two cases of asymptomatic infection with SARSCoV-2: Are 14 days of isolation sufficient? Int J Infect Dis 2020; 95:174–5. 46. 46.Lescure FX, Bouadma L, Nguyen D, et al. Clinical and virological data of the first cases of COVID-19 in Europe: a case series. Lancet Infect Dis 2020; 20(6): 697–706. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S1473-3099(20)30200-0&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F23%2F2020.08.20.20178699.atom) 47. 47.Wolfel R, Corman VM, Guggemos W, et al. Virological assessment of hospitalized patients with COVID-2019. Nature 2020; 581(7809): 465–9. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41586-020-2196-x&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F23%2F2020.08.20.20178699.atom) 48. 48.Xu Y, Li X, Zhu B, et al. Characteristics of pediatric SARS-CoV-2 infection and potential evidence for persistent fecal viral shedding. Nat Med 2020; 26(4): 502–5. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41591-020-0817-4&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F23%2F2020.08.20.20178699.atom) 49. 49.Han MS, Seong MW, Heo EY, et al. Sequential analysis of viral load in a neonate and her mother infected with SARS-CoV-2. Clin Infect Dis 2020. 50. 50.Wyllie AL, Fournier J, Casanovas-Massana A, et al. Saliva is more sensitive for SARS-CoV-2 detection in COVID-19 patients than nasopharyngeal swabs. medRxiv 2020: 2020.04.16.20067835. 51. 51.Cheng CY, Lee YL, Chen CP, et al. Lopinavir/ritonavir did not shorten the duration of SARS CoV-2 shedding in patients with mild pneumonia in Taiwan. J Microbiol Immunol Infect 2020; 53(3): 488–92. 52. 52.Yang Y, Yang M, Shen C, et al. Evaluating the accuracy of different respiratory specimens in the laboratory diagnosis and monitoring the viral shedding of 2019-nCoV infections. medRxiv 2020: 2020.02.11.20021493. 53. 53.Yang JR, Deng DT, Wu N, Yang B, Li HJ, Pan XB. Persistent viral RNA positivity during the recovery period of a patient with SARS-CoV-2 infection. J Med Virol 2020. 54. 54.Covid-Investigation Team. Clinical and virologic characteristics of the first 12 patients with coronavirus disease 2019 (COVID-19) in the United States. Nat Med 2020; 26(6): 861–8. 55. 55.Lui G, Ling L, Lai CK, et al. Viral dynamics of SARS-CoV-2 across a spectrum of disease severity in COVID-19. J Infect 2020; 81(2): 318–56. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.jinf.2020.04.025&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F23%2F2020.08.20.20178699.atom) 56. 56.Hu Y, Shen L, Yao Y, Xu Z, Zhou J, Zhou H. A report of three COVID-19 cases with prolonged viral RNA detection in anal swabs. Clin Microbiol Infect 2020; 26(6): 786–7. 57. 57.Seah IYJ, Anderson DE, Kang AEZ, et al. Assessing Viral Shedding and Infectivity of Tears in Coronavirus Disease 2019 (COVID-19) Patients. Ophthalmology 2020; 127(7): 977–9. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F23%2F2020.08.20.20178699.atom) 58. 58.Liu WD, Chang SY, Wang JT, et al. Prolonged virus shedding even after seroconversion in a patient with COVID-19. J Infect 2020; 81(2): 318–56. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.jinf.2020.04.025&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F23%2F2020.08.20.20178699.atom) 59. 59.Xing YH, Ni W, Wu Q, et al. Prolonged viral shedding in feces of pediatric patients with coronavirus disease 2019. J Microbiol Immunol Infect 2020; 53(3): 473–80. 60. 60.Qian GQ, Chen XQ, Lv DF, et al. Duration of SARS-CoV-2 viral shedding during COVID-19 infection. Infect Dis (Lond) 2020; 52(7): 511–2. 61. 61.Kim ES, Chin BS, Kang CK, et al. Clinical Course and Outcomes of Patients with Severe Acute Respiratory Syndrome Coronavirus 2 Infection: a Preliminary Report of the First 28 Patients from the Korean Cohort Study on COVID-19. J Korean Med Sci 2020; 35(13): e142. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3346/jkms.2020.35.e142&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F23%2F2020.08.20.20178699.atom) 62. 62.Colavita F, Lapa D, Carletti F, et al. SARS-CoV-2 Isolation From Ocular Secretions of a Patient With COVID-19 in Italy With Prolonged Viral RNA Detection. Ann Intern Med 2020; 173(3): 242–3. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.7326/M20-1176&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F23%2F2020.08.20.20178699.atom) 63. 63.Hill KJ, Russell CD, Clifford S, et al. The index case of SARS-CoV-2 in Scotland. J Infect 2020; 81(1): 147–78. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F23%2F2020.08.20.20178699.atom) 64. 64.Huang Y, Chen S, Yang Z, et al. SARS-CoV-2 Viral Load in Clinical Samples from Critically III Patients. Am J Respir Crit Care Med 2020; 201(11): 1435–8. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F23%2F2020.08.20.20178699.atom) 65. 65.Zhou R, Li F, Chen F, et al. Viral dynamics in asymptomatic patients with COVID-19. Int J Infect Dis 2020; 96:288–90. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.ijid.2020.05.030&link_type=DOI) 66. 66.Qi L, Yang Y, Jiang D, et al. Factors associated with the duration of viral shedding in adults with COVID-19 outside of Wuhan, China: a retrospective cohort study. Int J Infect Dis 2020; 96:531–7. 67. 67.Yuan C, Zhu H, Yang Y, et al. Viral loads in throat and anal swabs in children infected with SARS-CoV-2. Emerg Microbes Infect 2020; 9(1): 1233–7. 68. 68.Huang JT, Ran RX, Lv ZH, et al. Chronological Changes of Viral Shedding in Adult Inpatients with COVID-19 in Wuhan, China. Clin Infect Dis 2020. 69. 69.Kong Y, Cai C, Ling L, et al. Successful treatment of a centenarian with coronavirus disease 2019 (COVID-19) using convalescent plasma. Transfus Apher Sci 2020: 102820. 70. 70.Tam PCK, Ly KM, Kernich ML, et al. Detectable severe acute respiratory syndrome coronavirus 2 (SARSCoV-2) in human breast milk of a mildly symptomatic patient with coronavirus disease 2019 (COVID-19). Clin Infect Dis 2020. 71. 71.Yoon JG, Yoon J, Song JY, et al. Clinical Significance of a High SARS-CoV-2 Viral Load in the Saliva. J Korean Med Sci 2020; 35(20): e195. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3346/jkms.2020.35.e195&link_type=DOI) 72. 72.Phan LT, Nguyen TV, Huynh LKT, et al. Clinical features, isolation, and complete genome sequence of severe acute respiratory syndrome coronavirus 2 from the first two patients in Vietnam. J Med Virol 2020. 73. 73.Klement-Frutos E, Burrel S, Peytavin G, et al. Early administration of ritonavir-boosted lopinavir could prevent severe COVID-19. J Infect 2020. 74. 74.Gross R, Conzelmann C, Muller JA, et al. Detection of SARS-CoV-2 in human breastmilk. Lancet 2020; 395(10239): 1757–8. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S0140-6736(20)31181-8&link_type=DOI) [1]: /embed/graphic-1.gif [2]: /embed/graphic-2.gif [3]: /embed/graphic-3.gif [4]: /embed/graphic-4.gif [5]: /embed/graphic-5.gif [6]: /embed/graphic-6.gif