SARS-CoV-2 infectious virus, viral RNA in nasopharyngeal swabs, and serostatus of symptomatic COVID-19 outpatients in the United States ======================================================================================================================================= * Katie R. Mollan * Joseph J. Eron * Taylor J. Krajewski * Wendy Painter * Elizabeth R. Duke * Caryn G. Morse * Erin A. Goecker * Lakshmanane Premkumar * Cameron R. Wolfe * Laura J. Szewczyk * Paul L. Alabanza * Amy James Loftis * Emily J. Degli-Angeli * Ariane J. Brown * Joan A. Dragavon * John J. Won * Jessica Keys * Michael G. Hudgens * David A. Wohl * Myron S. Cohen * Ralph S. Baric * Robert W. Coombs * Timothy P. Sheahan * William A. Fischer II ## Abstract **Background** SARS-CoV-2 infectious virus isolation in the upper airway of COVID-19 patients is associated with higher levels of viral RNA. However, comprehensive evaluation of the relationships between host and disease factors and infectious, replication competent virus is needed. **Methods** Symptomatic COVID-19 outpatients were enrolled from the United States. Clinical symptoms were recorded via patient diary. Nasopharyngeal swabs were collected to quantitate SARS-CoV-2 RNA by reverse transcriptase polymerase chain reaction and for infectious virus isolation in Vero E6-cells. SARS-CoV-2 antibodies were measured in serum using a validated ELISA assay. **Findings** Among 204 participants within one week of reported symptom onset (median=5, IQR 4-5 days), median age was 40 (min-max: 18-82 years), median nasopharyngeal viral RNA was 6.5 (IQR 4.7-7.6 log10 copies/mL), and 26% had detectable SARS-CoV-2 antibodies at baseline. Infectious virus was recovered in 7% of participants with antibodies compared to 58% of participants without antibodies (probability ratio (PR)=0.12, 95% CI: 0.04, 0.36; p=0.00016). Infectious virus isolation was also associated with higher levels of viral RNA (mean RNA difference +2.6 log10, 95% CI: 2.2, 3.0; p<0.0001) and fewer days since symptom onset (PR=0.79, 95% CI: 0.71, 0.88 per day; p<0.0001). **Interpretation** The presence of SARS-CoV-2 antibodies is strongly associated with clearance of infectious virus isolation. Seropositivity and viral RNA are likely more reliable markers of infectious virus suppression than subjective measure of COVID-19 symptoms. Virus-targeted treatment and prevention strategies should be administered as early as possible and ideally before seroconversion. **Funding** Ridgeback Biotherapeutics, LP and NIH **[ClinicalTrials.gov](http://ClinicalTrials.gov) Identifier** NCT04405570 **Evidence before this study** A deeper understanding of the viral and host factors associated with infectious virus detection is essential to accurately identify and quarantine contagious individuals. Several studies have reported associations between SARS-CoV-2 virus isolation and viral RNA levels or time from symptom onset. However, little is known about which host factors (i.e. demographics, comorbidities, SARS-CoV-2 seropositivity, symptomatology etc.) are associated with infectious virus detection. A search of PubMed on 12 April 2021 using keywords “COVID-19” or “SARS-CoV-2” and “infectious virus isolation” yielded 14 publications that evaluated virus isolation from respiratory samples of SARS-CoV-2 infected individuals. Five of these studies were case reports or case series that included up to 5 individuals. Seven studies included cohorts of both outpatients and hospitalized individuals and found strong associations between virus isolation and time from symptom onset and viral RNA level. Of these seven, only two studies evaluated the association between SARS-CoV-2-specific antibodies and virus isolation, but were limited by the late timing of collection (>10 days after symptom onset) or the small number of participants included. The remaining two articles compared virus isolation with antigen and molecular based diagnostics and reviewed prior literature respectively. Collectively, these studies suggest that infectious virus isolation from nasopharyngeal swab samples is possible up to 10 days from symptom onset in individuals with mild disease and for longer in those with severe illness or an underlying immune deficiency. No study has systematically evaluated host, disease, and viral factors associated with infectious virus isolation in the same cohort. **Added value of this study** To better understand the host, disease, and viral factors associated with virus isolation in ambulatory individuals with COVID-19, we analyzed demographic, symptom, virologic, and SARS-CoV-2-specific antibody data at baseline entry to outpatient care from 204 individuals enrolled in a randomized placebo-controlled study of a novel oral therapeutic. Host characteristics were self-reported and viral RNA quantitation, virus isolation, and SARS-2 specific antibody testing (IgG, IgM, IgA and total Ig) were performed at central laboratories. This represents the largest evaluation of virus isolation from symptomatic outpatients with COVID-19 reported to date, and affords an important opportunity to understand which host, virus, and disease factors are associated with the presence and clearance of infectious virus in the nasopharynx. Consistent with prior studies, we found that isolation of replication competent SARS-CoV-2 *in vitro* strongly correlated with both higher nasal viral qRT-PCR RNA levels and shorter time since symptom onset. Importantly, we also found that SARS-CoV-2 antibodies are strongly associated with the clearance of infectious virus, with virus isolation in 7% of seropositive individuals compared to 58% of seronegative individuals. **Implications of all the available evidence** Our findings provide a comprehensive analysis of key virus, host, and disease factors associated with infectious virus isolation and suggest that antibody detection appears to be a more reliable marker of infectious virus clearance than patient-reported symptom duration. Keywords * COVID-19 * infectious virus * outpatient * SARS-CoV-2 * serostatus * viral RNA ## Introduction Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), the etiological agent of coronavirus disease-2019 (COVID-19), has caused more than 153 million infections and 3.2 million deaths worldwide.(1,2) Based on limited information about the length of infectiousness of COVID-19 patients following onset of symptoms, the US Centers for Disease Control and Prevention (CDC) recommends a physical isolation period of 10 days for individuals with mild-to-moderate COVID-19, and 20 days for individuals with severe disease.(3) However, infectious SARS-CoV-2 virus isolation has been reported beyond 20 days from patients with a compromised immune system.(4–9) While prior studies have demonstrated an association between infectious virus isolation in the upper airway and high levels of nasal viral RNA measured by quantitative reverse transcription polymerase chain reaction (qRT-PCR),(10–14) limited data are available regarding the host and disease factors associated with the presence of infectious virus in persons with mild-to-moderate COVID-19. An improved understanding of viral and host factors associated with shedding of infectious virus is essential to prevent transmission of SARS-CoV-2 and reliably evaluate the antiviral efficacy of novel therapies. Here, we provide a comprehensive analysis of demographic, immunologic, virologic, and clinical disease factors associated with infectious virus isolation and levels of viral RNA in nasopharyngeal swab samples in the largest study of symptomatic outpatient adults with COVID-19 to date. ## Methods ### Study design This cross-sectional analysis was conducted at study entry among participants who enrolled in a Phase IIa clinical trial of the oral, direct acting antiviral agent molnupiravir versus placebo. Adult outpatients with symptomatic mild-to-moderate COVID-19 in the United States were enrolled within 1 week of symptom onset.(15,16) The study protocol was approved by Western IRB (WIRB), and for each site by WIRB or a local IRB per site requirements. Ten sites enrolled participants in the following states: North Carolina, California, Washington, Texas, Florida, and Louisiana. All study participants provided written informed consent. Results from the pre-treatment, baseline visits (day 1 or screening) are reported. ### Eligibility criteria Participants were eligible for enrollment if they were 18 years of age or older, had SARS-CoV-2 infection with symptom onset within the past 7 days (168 hours), and were experiencing a fever (or feeling feverish or having chills) or signs/symptoms of respiratory illness (e.g., upper respiratory congestion, loss of sense of smell or taste, sore throat, cough, or shortness of breath). Active SARS-CoV-2 infection was confirmed by molecular testing from a sample collected within 96hr prior to enrollment. Participants who received a SARS-CoV-2 vaccine, treatment with possible anti-SARS-CoV-2 therapies within 30 days prior to enrollment, and women who were pregnant or breastfeeding were excluded. Additional eligibility criteria included: no (known) history of or current hospitalization for COVID-19, no history of significant kidney or liver disease, and no history of known blood dyscrasia, hemorrhagic cerebrovascular accident, or major bleed. A complete list of eligibility criteria is available on [clinicaltrials.gov](http://clinicaltrials.gov) ([NCT04405570](http://medrxiv.org/lookup/external-ref?link_type=CLINTRIALGOV&access_num=NCT04405570&atom=%2Fmedrxiv%2Fearly%2F2021%2F06%2F01%2F2021.05.28.21258011.atom)). ### Measurement of SARS-CoV-2 RNA, Infectious Virus, and Antibodies Nasopharyngeal (NP) swabs were collected from each participant at enrollment. Two NP swabs were collected—one from each nostril from all participants except for n=31 where a single NP swab was collected and aliquoted for testing. Nasopharyngeal swabs were placed into 3 mL of virus transport medium (VTM; supplied by COPAN), stored at 4°C prior to shipping in refrigerated containers to central laboratories, aliquoted, and stored at −80°C until tested. To measure viral RNA, qRT-PCR was performed on NP swabs using CDC primer/probe solution against the N1 region of the nucleocapsid gene (2019-nCoV_N1) at Covance CLS with Promega Maxwell for RNA extraction, manual plate build, and Quantstudio 12kflex for the 96 well qRT-PCR format (lower limit of quantification: 1,018 copies/mL).(17) To measure the presence of infectious virus in each NP swab, samples were cultured in Vero E6 cells similar to prior studies.(4) All work was performed in a UNC Department of Environmental Health (EHS)/CDC-approved BSL3 facility following approved standard operating procedures and laboratory safety plans. Briefly, VTM samples were thawed, diluted 1:1 in infection medium (MEM, 4% fetal bovine serum, 1X penicillin and streptomycin), and added in duplicate to Vero cell cultures plated 24hr prior. After 1hr at 37°C, input sample was removed (to limit contamination of the signal), monolayers were single-washed with MEM and fresh medium was added. Quadruplicate negative (infection medium only) high (500 particle forming units, PFU) and low (50 PFU) positive controls were included.(11) On 2 and 5 days post inoculation (dpi), culture positivity was determined by measuring viral RNA in heat inactivated culture medium by qRT-PCR using the Abbott m2000sp/rt quantitative SARS-CoV-2 RNA assay (limit of quantification: 100 copies/mL). The limit of culture detection was 50 PFU (MOI 0.0008)) using two criteria for positivity: (i) SARS-CoV-2 RNA copies > 1,000/*mL* in supernatant at 2dpi, or (ii) a relative change of greater than 5 in RNA copy number from 2 to 5 dpi (i.e., [*copies at* 5*dpi* / *copies at* 2*dpi*] > 5). SARS-CoV-2-specific antibodies in serum were analyzed using an antigen-capture enzyme-linked immunosorbent assay as described previously.(18) Biotinylated recombinant Spike RBD was captured on a 96-well ELISA plate coated with streptavidin. Heat inactivated 40x diluted specimen was incubated in the RBD-captured wells, and bound antigen detected using HRP conjugated anti-goat total (IgG, IgM and IgA) or individual antibody isotype on a microplate reader. Based on reference panel performance, the following optical density thresholds for seropositivity were applied: total Ig>0.376, IgG>0.376, IgA>0.30, IgM>0.31. SARS-CoV-2 antibody positive status was determined by a positive result on at least one of the following: total Ig, IgG, IgM, or IgA. ### Covariates Participant demographics and disease characteristics were collected at baseline, including medical history, hematology, and chemistry laboratory tests. Obesity was defined as body mass index (BMI) ≥30 kg/m2. Days since symptom onset was measured by participant report; and COVID-19 symptom severity was assessed using a four-point scale (absent, mild, moderate, or severe) on the Day 1 participant-reported diary. Sixteen COVID-related symptoms were collected, as well as overall severity: “*Overall, how bad are your symptoms today?”* D-dimer and C-reactive protein (CRP) were measured at local site laboratories with varying limits of detection. D-dimer was dichotomized as <0.5 (within or near normal range) versus ≥0.5 mg/L, and CRP was dichotomized as <10 versus ≥10 mg/L. ### Data analyses This analysis includes baseline results from enrolled participants. A type I error rate of 0.05 was applied throughout (95% confidence interval (CI)) with no adjustment for multiplicity. Missing data were excluded and typically were missing due to specimen cold-chain shipping abnormalities. Analyses were conducted in Windows SAS version 9.4 (Cary, NC) and Windows R version 4.0.2 or 4.0.4. Associations between infectious virus isolation and each characteristic were estimated with a probability ratio (PR) and corresponding 95% CI from a modified Poisson model for bivariate and multivariable analyses.(19) The following host characteristics were evaluated for associations with infectious virus isolation: presence of antibodies, symptom duration, age, sex, ethnicity (Hispanic versus non-Hispanic), BMI and obesity, frequent medical conditions in our study population that are known to be associated with worse COVID-19 illness (diabetes, hypertension, and asthma), symptom severity, and five COVID-19 respiratory-related symptoms (cough, shortness of breath, sore throat, nasal obstruction, nasal discharge). Race was not evaluated because few participants self-identified as African-American or Asian. Enrollment of Latinx participants varied considerably by site, and thus *post hoc* sensitivity analyses accounting for site were conducted using a Mantel-Haenszel pooled PR. The same statistical approach was used to estimate PRs for associations of each characteristic with seropositive antibody status. Mean difference in SARS-CoV-2 RNA levels (log10 copies/mL) was estimated for each host characteristic listed above using a general linear model with heteroscedasticity-consistent standard errors. A receiver operator curve (ROC) analysis was applied to identify a SARS-CoV-2 RNA cut-point for infectious virus culture positive that minimized the Euclidean distance between the ROC curve and the (0,1) point in the ROC plane, to maximize sensitivity and specificity. Wilcoxon rank-sum tests were used to compare hematology measures by infectious virus status, and by antibody status. A chi-squared test was used to compare inflammatory markers (D-dimer, CRP) by infectious virus status, and by antibody status. ## Results ### Patient Population Between 19 June 2020 and 22 January 2021, 240 ambulatory participants were screened, and 204 participants were enrolled at ten outpatient clinical sites across the United States. Enrollment visits occurred a median of 5 (IQR 4-5) days since symptom onset. Median age was 40 (IQR 27-52) years and 51% were women. Forty-seven percent of participants were White non-Hispanic, 42% Hispanic/Latinx, 5% Black non-Hispanic, 3% Asian non-Hispanic, and 2% multiple or other races (**Table 1**). View this table: [Table 1.](http://medrxiv.org/content/early/2021/06/01/2021.05.28.21258011/T1) Table 1. Baseline characteristics of outpatients with symptomatic mild-to-moderate COVID-19 (*n* = 204) Baseline median SARS-CoV-2 viral RNA from NP swabs was 6.5 (IQR 4.7-7.6) log10 copies/mL and 8% of participants (15/195) had RNA below the limit of quantification (i.e., <1,018 copies/mL) (**Table 1**). Median viral RNA was highest ≤3 days after symptom onset (**Supplemental Figure 1**). Overall symptom severity was rated as mild for 48%, moderate for 45%, and severe for 7% of participants. The most frequent symptoms, regardless of severity, were fatigue (80%), cough (77%), stuffy nose (74%), headache (71%), and muscle aches (64%). Loss of smell and loss of taste were each experienced by 54% of participants (**Table 2**). Baseline laboratory testing was notable for a median baseline lymphocyte count of 1.4 ×109/L (IQR 1.2-1.9). Most participants had D-dimer and CRP levels within or near the normal range with 80% below 0.5 mg/L and 77% below 10 mg/L, respectively (**Table 1**). View this table: [Table 2.](http://medrxiv.org/content/early/2021/06/01/2021.05.28.21258011/T2) Table 2. Symptom severity among outpatients with mild-to-moderate COVID-19† ### SARS-CoV-2 antibodies Of 177 patients with evaluable measurements, 26% were antibody positive at baseline for at least one SARS-CoV-2 spike protein-specific immunoglobulin isotype, with 20% IgG+, 23% IgM+, and 8% IgA+ (**Table 1**). Three participants were IgG(+) with IgM(-) and IgA(-) and only one was IgA+ only (**Supplemental Table 1**). In bivariate analyses, host and disease characteristics associated with having SARS-CoV-2 specific antibodies included longer time since symptom onset (probability ratio [PR]=1.27 per 1 day, 95%CI: 1.06, 1.52, p=0.0091) and higher white blood cell counts including total white blood cells and absolute neutrophil count (**Supplemental Figure 2**). Additionally, self-described Latinx ethnicity was associated with antibody positivity in the unadjusted analysis, but in a sensitivity analysis adjusting for site, this association was no longer evident (Mantel-Haenszel PR=0.82 Latinx versus not Latinx, 95%CI: 0.41, 1.62, p=0.57). Participants with SARS-CoV-2 antibodies were more likely to have elevated D-dimer and CRP levels as compared to seronegative participants (D-dimer ≥0.5 mg/L: 39% versus 13%, p=0.00037; CRP ≥10 mg/L: 44% versus 15%, p=0.00011; **Supplemental Table 2**). ### Correlates of virus isolation Infectious virus was isolated from 45% (78/175) of participants at baseline. Those with positive virus isolation had a median symptom duration of 4 (IQR 3-5) days compared to 5 (IQR 4-6) days in participants with negative virus isolation. Mean viral RNA levels were higher among participants with infectious virus isolation compared to those who were culture negative (mean: 7.6 versus 5.0 log10 copies/mL; mean difference: 2.6, 95%CI: 2.2, 3.0, p<0.0001 **Figure 1**). An ROC curve analysis revealed that a viral RNA threshold of ≥6.4 log10 copies/mL was predictive of infectious virus isolation (**Supplemental Figure 3**), with estimated positive predictive value of 80% and a negative predictive value of 91% (**Supplemental Table 3**). ![Figure 1.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2021/06/01/2021.05.28.21258011/F1.medium.gif) [Figure 1.](http://medrxiv.org/content/early/2021/06/01/2021.05.28.21258011/F1) Figure 1. SARS-CoV-2 viral RNA levels in nasopharyngeal swab by infectious virus status Nasopharyngeal viral RNA levels measured via qRT-PCR are displayed by infectious virus status, with culture negative in grey (*n* = 95) and culture positive (*n* = 78) in red; each dot represents a participant. Solid lines on the boxplots display the median and 25th-75th percentile (mean = blue dashed line) and the whiskers extend to the extrema (no more than 1.5 times the IQR from the box). LLoQ = lower limit of quantification; SD = standard deviation. Next, the association between SARS-CoV-2 specific serostatus and infectious virus isolation was evaluated. Infectious virus was isolated from 7% (3/43) of seropositive participants compared to 58% (73/125) of seronegative participants, consistent with an 88% lower prevalence of infectious virus for those with SARS-CoV-2 antibodies (probability ratio [PR]=0.12, 95%CI: 0.04, 0.36, p=0.00016). Infectious virus was isolated in only three seropositive individuals at 3, 4, and 6 days from symptom onset (all 3 had SARS-CoV-2 specific IgM; 2 had IgG; and 0 had IgA). There was also a strong association between antibody status and NP viral RNA levels (mean: 4.4 log10 copies/ml among seropositives versus 6.8 log10 copies/ml among seronegatives; mean difference: −2.4, 95%CI: −2.8, −1.9, p<0.0001). Those with viral RNA levels in the top quartile (>7.6 log10) were seronegative (100%; 43/43) with the majority (91%; 39/43) having infectious virus detected, whereas the viral RNA bottom quartile (<4.7 log10) was almost entirely infectious virus negative individuals (98%; 40/41) and many were seropositive (68%; 28/41) (**Figure 2**). ![Figure 2.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2021/06/01/2021.05.28.21258011/F2.medium.gif) [Figure 2.](http://medrxiv.org/content/early/2021/06/01/2021.05.28.21258011/F2) Figure 2. SARS-CoV-2 viral RNA, time since symptom onset, and infectious virus by SARS-CoV-2 specific antibody serostatus Panel A shows seropositive participants. Panel B shows seronegative participants. Seropositive is defined as having SARS-CoV-2 specific total Ig, IgG, IgM, or IgA antibodies. Nasopharyngeal viral RNA levels (log10 copies/mL) are shown on the y-axis and days since symptom onset on the x-axis, with infectious virus culture positive participants in red circles, and culture negative participants in grey squares. The overall viral RNA median (Q1, Q3) and LLoQ are indicated by dashed horizontal lines. Ig = immunoglobulin; LLoQ = lower limit of quantification; Q1 = 25th percentile; Q3 = 75th percentile. In a multivariable model (that included viral RNA, antibody status, and symptom duration), infectious virus isolation was more prevalent for those with higher viral RNA (PR=1.52 per 1.0 log10 copies/mL, 95%CI: 1.37, 1.70, p<0.0001), less prevalent for those with antibodies (PR=0.35 for seropositive versus seronegative, 95%CI: 0.11, 1.07, p=0.066), and no longer associated with days since symptom onset (PR=1.01 per 1 day, 95%CI: 0.91, 1.11, p=0.90). To better understand the association between infectious virus isolation and host characteristics both bivariate and multivariable analyses were performed. In bivariate analyses, infectious virus isolation was more prevalent among participants with hypertension (PR=1.62, 95%CI: 1.15, 2.28, p=0.0054) or nasal obstruction (PR=1.53, 95%CI: 0.97, 2.41, p=0.066). However, infectious virus isolation was less prevalent among participants who were further out from symptom onset (PR=0.79 per 1 day, 95%CI: 0.71, 0.88, p<0.0001), those with moderate-to-severe versus mild symptoms (PR=0.68, 95%CI: 0.48, 0.96, p=0.029), those with shortness of breath (PR=0.52, 95%CI: 0.35, 0.77, p=0.0013), and those who self-identified as Latinx compared with non-Latinx (PR=0.54, 95%CI: 0.36, 0.79, p=0.0016). In a sensitivity analysis adjusting for site, the association between Latinx ethnicity and lower prevalence of virus isolation remained (Mantel-Haenszel PR=0.52, 95%CI: 0.28, 0.96, p=0.032). In this study, infectious virus isolation was not clearly associated with age (PR=1.09 per 10 years, 95%CI: 0.98, 1.22, p=0.10) or biological sex (PR=0.85 women vs. men, 95%CI: 0.61, 1.19, p=0.34) (**Figure 3**). In a multivariable model including host characteristics (**Supplemental Table 4**), seronegativity, fewer days since symptom onset, non-Latinx ethnicity, and hypertension remained associated with higher prevalence of infectious virus isolation. ![Figure 3.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2021/06/01/2021.05.28.21258011/F3.medium.gif) [Figure 3.](http://medrxiv.org/content/early/2021/06/01/2021.05.28.21258011/F3) Figure 3. Host factor associations with infectious SARS-CoV-2 virus isolation Bivariate unadjusted analyses are shown. *Prevalence ratios for the probability of infectious virus isolation were estimated for each dichotomous characteristic and a prevalence ratio per unit change was estimated for each continuous characteristic with corresponding 95% confidence intervals. Each continuous characteristic was fit as linear in the log-prevalence of infectious virus isolation. PR = prevalence ratio. Bivariate associations between infectious virus isolation and inflammatory and hematology markers were also evaluated. Elevated inflammatory markers were numerically less frequent among participants with infectious virus culture positive compared to culture negative, however the differences were not statistically significant (D-dimer ≥0.5 mg/L: 16% versus 26%, p=0.21; CRP ≥10 mg/L: 17% versus 27%, p=0.12) (**Supplemental Table 2**). Participants with positive infectious virus culture had lower levels of white blood cells including total white blood cell count, absolute lymphocyte count, and absolute neutrophil count **(Supplemental Figure 4)**. ### Correlates of viral RNA Bivariate analyses of host factor associations with viral RNA level are provided in **Supplemental Figure 5**. Higher viral RNA was associated with fewer days since symptom onset (mean difference (MD) −0.5 log10 per 1 day, 95%CI: −0.6, −0.3, p<0.0001). There was a trend toward higher viral RNA levels in those with mild symptom severity (MD: −0.5 log10 for moderate-to-severe versus mild symptoms, 95%CI: −1.0, 0.07, p=0.086), with median viral RNA levels of 6.8 (IQR 4.8-8.1), 6.4 (IQR 4.6-7.3), and 5.7 (IQR 4.6-7.1) log10 copies/mL for participants with mild, moderate, and severe overall symptom severity, respectively. Viral RNA levels were higher on average for those with hypertension (MD: 1.0 log10, 95%CI: 0.3, 1.7, p=0.0036) or nasal discharge (MD: 0.7 log10, 95%CI: 0.2, 1.2, p=0.0052). Lower viral RNA levels were observed for those who self-identified as Latinx; however, in a sensitivity model adjusting for site as a covariate, the association between Latinx ethnicity and viral RNA was no longer present (MD: −0.01 log10, 95%CI: −0.7, 0.7, p=0.98). Viral RNA was not evidently associated with age in years (MD: 0.1 log10 per 10 years of age, 95%CI: −0.06, 0.3, p=0.20) or body mass index (MD: 0.02 log10 per 5 kg/m2, 95%CI: −0.2, 0.3, p=0.83). There was a trend for lower levels of viral RNA in women (MD: −0.5 log10, 95%CI: −1.0, 0.02, p=0.057). ## Discussion SARS-CoV-2 infectious virus detection within 1 week of symptom onset was prevalent and strongly associated with quantitative viral RNA levels and the absence of SARS-CoV-2 specific antibodies, suggesting that humoral immunity plays a key role in limiting infectious virus shedding. This observation is further supported by prior studies identifying prolonged viral replication in individuals with immune deficiencies, almost all of whom had specific deficiencies in B cell function.(4–8,20) Our study and others report a strong association between absence of SARS-CoV-2 antibodies and higher levels of viral RNA (21), which further supports the role of a humoral immunity in controlling virus replication. Viral RNA levels were highest soon after symptom onset (half of participants had viral RNA >3 million copies/mL at entry), with a peak in median viral RNA at or before 3 days after symptom onset, and lower levels for those presenting 5 to 7 days after symptom onset. Using a receiver operator curve analysis, we identified that viral RNA in NP swabs of 6.4 log10 copies/mL or higher is predictive of infectious virus detection, with a positive predictive value of 80% and negative predictive value of 91%, supporting prior studies suggesting a threshold of 6 log10 copies/mL.(10,11,22) However, infectious virus was isolated in three individuals with viral RNA below 5.0 log10 copies/mL (minimum: 4.3 log10 copies/mL), all three of whom were antibody negative men 6 days out from symptom onset, suggesting a delayed immune response. At entry to outpatient care for mild-to-moderate COVID-19, most individuals had a cough, nasal obstruction, fatigue, or headache; about half reported sore throat or loss of taste/smell; and 41% reported feeling feverish, having chills, or experiencing shortness of breath. Our study revealed associations between infectious virus isolation and milder symptoms, an absence of shortness of breath, and lower white blood cell populations, consistent with an early stage in the clinical course of infection and virus-associated lymphopenia. The presence of SARS-CoV-2 antibodies was associated with higher white blood cell and neutrophil counts, which is consistent with this interpretation. Prior studies report decreased virus isolation beyond 10 days from symptom onset.(11,14,23,24) In our study, when accounting for both NP RNA level and serologic response, duration of symptoms was no longer associated with virus isolation, and about half (12 of 25) of the seronegative patients who presented at day 6 or 7 from symptom onset were culture positive. In this study of outpatients with mild-to-moderate COVID-19, we did not identify associations between several pre-existing risk factors for severe COVID-19 –including age, obesity, diabetes or asthma – and infectious virus isolation or higher levels of viral RNA. However, individuals with hypertension tended to have higher levels of viral RNA and higher prevalence of infectious virus isolation, the mechanism of which is unclear. Importantly, most outpatients with mild-to-moderate symptoms, even those in higher risk groups, have a relatively low probability of developing severe COVID-19 disease.(25–27) While this study describes novel links between virologic and immunologic factors associated with isolation of infectious virus, there are important limitations to consider, including representation. There were a large number of participants who identified as Latinx, yet there was an under-representation of African-Americans and Asian-Americans compared with the US burden of COVID-19.(28) Further, this study enrolled predominantly low risk outpatients with at least one symptom of COVID-19, which limits the study population to outpatients with mild-to-moderate COVID-19. Additional research is needed to understand the factors associated with infectious virus among asymptomatic individuals. While prior studies have demonstrated prolonged virus isolation in hospitalized individuals,(11,23,29) it remains important to identify the factors associated with progression to severe disease and infectious virus shedding in outpatients. Another limitation is that seropositivity is not synonymous with virus neutralization. We observed three antibody positive participants (each had IgM; two had IgG) with positive infectious virus culture, two of whom also had RNA levels above 6.0 log10 but neutralization titers were not available. We also explicitly acknowledge that a negative culture does not exclude the presence of infectious virus, and having a negative culture is not synonymous with being non-infectious. The sensitivity of the virus isolation assay is 50pfu/ml, and thus infectious virus below this limit may not be detected. Additionally, variability in swabbing technique can result in false negative cultures. Nonetheless, the strong associations we observed between NP RNA shedding and antibody status with isolation of infectious virus provide important insight into the infectious virus burden of individuals and populations. Importantly, our study suggests that determining a patient’s serostatus and NP RNA level may help clinicians assess likelihood of infectious virus detection, which could help guide isolation, therapy, and follow-up. Longitudinal research is needed to determine if the early detection of antibodies could serve as a correlate of infectious virus clearance and whether early viral clearance decreases the likelihood of long-term COVID-19 symptoms.(30) In this study of outpatient adults with COVID-19, infectious virus isolation was associated with high viral RNA levels and an absence of SARS-CoV-2 antibodies. Ongoing studies are evaluating the neutralization capacity of the antibodies detected to understand the functional aspects of these antibodies. As SARS-CoV-2 continues to spread globally, improved understanding of host, disease, and viral factors associated with infectious virus isolation will be important to implement non-pharmacologic, vaccine, and therapeutic strategies to interrupt transmission and progression of disease. ## Data Availability Upon approval from the corresponding authors and Ridgeback Biotherapeutics, de-identified participant level data may be shared given the investigator who requests the data has approval from an Institutional Review Board, Independent Ethics Committee, or Research Ethics Board, as applicable, and executes a data use/sharing agreement. ## Funding Ridgeback Biotherapeutics LP funded this study and has subsequently entered into a collaboration with Merck to jointly develop molnupiravir. This research was also supported by the UNC-CH Center for AIDS Research P30 AI050410 (NIH); T32 ES007018 (NIEHS for TJK); a generous gift from the Chan Zuckerberg Initiative to RSB; FHCRC/UW CFAR AI-027757 and AI-106701 to RWC. ## Declarations of interest Grants or contracts were awarded to the participating sites from Ridgeback Biotherapeutics LP. KRM, CGM, and RSB have collaborations, unrelated to this study, with Gilead Sciences. JJE receives consulting honoraria outside the current study from Merck, VIR/GSK, and is a DSMB member for Adagio. WP and LJS are employed by Ridgeback Biotherapeutics LP. CGM has collaborations, unrelated to this study, with Roche, Moderna, Johnson & Johnson, Covis Pharma, and Chimerix. DAW has collaborations (advisory boards, consultancy, research funding), unrelated to this work, with Gilead Sciences. RSB has collaborations, unrelated to this study, with Moderna, Pfizer, Takeda, Eli Lily, and VaxArt. CRW has, unrelated to this study, financial disclosures for DSMB work with Biogen, Janssen, Merck and Atea, consultancy paid by Enzychem Lifesciences, and honoraria paid by Abbott Laboratories. TPS has contracts, unrelated to this work, from ViiV Healthcare and GSK. WAF has research funding from Ridgeback Biotherapeutics LP, participates on adjudication committees for Janssen, Syneos, and provides consultancy to Roche and Merck. All remaining authors have no conflicts of interest. ## Data sharing statement Upon approval from the corresponding author and Ridgeback Biotherapeutics, de-identified participant level data may be shared given the investigator who requests the data has approval from an Institutional Review Board, Independent Ethics Committee, or Research Ethics Board, as applicable, and executes a data use/sharing agreement. View this table: [Supplemental Table 1.](http://medrxiv.org/content/early/2021/06/01/2021.05.28.21258011/T3) Supplemental Table 1. SARS-CoV-2 specific antibody combinations View this table: [Supplemental Table 2.](http://medrxiv.org/content/early/2021/06/01/2021.05.28.21258011/T4) Supplemental Table 2. Associations between SARS-CoV-2 measures and inflammatory markers View this table: [Supplemental Table 3.](http://medrxiv.org/content/early/2021/06/01/2021.05.28.21258011/T5) Supplemental Table 3. Nasopharyngeal viral RNA threshold for infectious virus isolation View this table: [Supplemental Table 4.](http://medrxiv.org/content/early/2021/06/01/2021.05.28.21258011/T6) Supplemental Table 4. Multivariable model of host factor associations with infectious SARS-CoV-2 virus isolation (*n* = 162) † ## Figures ![Supplemental Figure 1.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2021/06/01/2021.05.28.21258011/F4.medium.gif) [Supplemental Figure 1.](http://medrxiv.org/content/early/2021/06/01/2021.05.28.21258011/F4) Supplemental Figure 1. SARS-CoV-2 virus isolation, viral RNA, and serostatus according to days since symptom onset Estimated prevalence of antibody positive (teal), prevalence of infectious virus positive (red), and the median viral RNA level (orange) are shown according to day since symptom onset. The left y-axis corresponds to viral RNA level and the right y-axis shows both the prevalence of infectious virus and prevalence of antibody positivity. Trend lines were estimated using a quadratic fit. ![Supplemental Figure 2.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2021/06/01/2021.05.28.21258011/F5.medium.gif) [Supplemental Figure 2.](http://medrxiv.org/content/early/2021/06/01/2021.05.28.21258011/F5) Supplemental Figure 2. Associations between hematology measures and SARS-CoV-2 specific antibody status Panels A-E: Five hematology measures are displayed by SARS-CoV-2-specific antibody status (total Ig, IgG, IgM, or IgA positivity), with antibody negative shown in grey and antibody positive shown in teal; each dot represents a participant. Solid lines on boxplots display the median and 25th-75th percentile (mean = blue dashed line). ![Supplemental Figure 3.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2021/06/01/2021.05.28.21258011/F6.medium.gif) [Supplemental Figure 3.](http://medrxiv.org/content/early/2021/06/01/2021.05.28.21258011/F6) Supplemental Figure 3. SARS-CoV-2 viral RNA by infectious virus culture – ROC Curve Panel A shows boxplots of viral RNA measured via qRT-PCR displayed by infectious virus status, with culture negative in grey and culture positive in red; each dot represents a participant. Panel B shows a receiver operator characteristic (ROC) curve. A threshold of 6.4 viral RNA log10 copies/mL maximized sensitivity and specificity for SARS-CoV-2 infectious virus isolation and is represented by the green line on the boxplots and the corresponding green dot on the ROC curve. ![Supplemental Figure 4.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2021/06/01/2021.05.28.21258011/F7.medium.gif) [Supplemental Figure 4.](http://medrxiv.org/content/early/2021/06/01/2021.05.28.21258011/F7) Supplemental Figure 4. Associations between hematology measures and infectious SARS-CoV-2 virus Panels A-E: Five hematology measures are displayed by infectious virus status, with infectious virus culture negative shown in grey and culture positive shown in red; each dot represents a participant. Solid lines on boxplots display the median and 25th-75th percentile (mean = blue dashed line). ![Supplemental Figure 5.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2021/06/01/2021.05.28.21258011/F8.medium.gif) [Supplemental Figure 5.](http://medrxiv.org/content/early/2021/06/01/2021.05.28.21258011/F8) Supplemental Figure 5. Host factor associations with SARS-CoV-2 viral RNA level Bivariate analyses are shown. Mean differences in SARS-CoV-2 viral RNA (log10 copies/mL) measured via qRT-PCR were estimated with a corresponding 95% confidence interval. MD = mean difference. ## Acknowledgements We sincerely thank all the participants and sites for their dedication and contributions to the study: Valley Clinical Trials (Northridge, CA), University of North Carolina (Chapel Hill, NC), Fred Hutchinson (Seattle, WA), Care United Research (Forney, TX), Benchmark Research (Colton, CA), FOMAT Medical Research (Oxnard, CA), Indago Research & Health Center (Hiahleah, FL), Wake Forest Baptist Health (Winston-Salem, NC), Duke University (Durham, NC), and NOLA Research Works (New Orleans, LA). This research was supported in part, by a generous gift from the Chan Zuckerberg Initiative to Dr. Ralph Baric. We thank Dr. Sarah Reifeis for data carpentry support, Ms. Ann Marie Weideman for statistical graphics advice, and Covance by Labcorp for data management support and quantification of viral RNA. ## Footnotes * kmollan{at}unc.edu * joseph_eron{at}med.unc.edu * tjk{at}live.unc.edu * painter{at}ridgebackbio.com * eduke{at}fredhutch.org * cmorse{at}wakehealth.edu * egoecker{at}uw.edu * prem{at}med.unc.edu * cameron.wolfe{at}duke.edu * laura{at}ridgebackbio.com * paul_alabanza{at}med.unc.edu * amy_james{at}med.unc.edu * edeglia1{at}uw.edu * ajbrown7{at}email.unc.edu * dragavon{at}uw.edu * jwon98{at}live.unc.edu * jkeys{at}email.unc.edu * mhudgens{at}email.unc.edu * david_wohl{at}med.unc.edu * myron_cohen{at}med.unc.edu * rbaric{at}email.unc.edu * bcoombs{at}uw.edu * sheahan{at}email.unc.edu * william_fischer{at}med.unc.edu * * co-senior authors * Received May 28, 2021. * Revision received May 28, 2021. * Accepted June 1, 2021. * © 2021, Posted by Cold Spring Harbor Laboratory This pre-print is available under a Creative Commons License (Attribution-NonCommercial-NoDerivs 4.0 International), CC BY-NC-ND 4.0, as described at [http://creativecommons.org/licenses/by-nc-nd/4.0/](http://creativecommons.org/licenses/by-nc-nd/4.0/) ## References 1. 1.Hu B, Guo H, Zhou P, Shi ZL. Characteristics of SARS-CoV-2 and COVID-19. Nat Rev Microbiol [Internet]. 2020;(December). Available from: [http://dx.doi.org/10.1038/s41579-020-00459-7](http://dx.doi.org/10.1038/s41579-020-00459-7) 2. 2.World Health Organization. WHO Coronavirus (COVID-19) Dashboard [Internet]. [cited 2021 May 5]. Available from: [https://covid19.who.int/](https://covid19.who.int/) 3. 3.CDC. Interim Guidance on Duration of Isolation and Precautions for Adults with COVID-19 [Internet]. [cited 2021 Apr 12]. Available from: [https://www.cdc.gov/coronavirus/2019-ncov/hcp/duration-isolation.html](https://www.cdc.gov/coronavirus/2019-ncov/hcp/duration-isolation.html) 4. 4.Aydillo T, Gonzalez-Reiche AS, Aslam S, van de Guchte A, Khan Z, Obla A, et al. Shedding of Viable SARS-CoV-2 after Immunosuppressive Therapy for Cancer. N Engl J Med [Internet]. 2020;383(26):2586–8. Available from: [http://www.ncbi.nlm.nih.gov/pubmed/32293753](http://www.ncbi.nlm.nih.gov/pubmed/32293753) 5. 5.Avanzato VA, Matson MJ, Seifert SN, Pryce R, Williamson BN, Anzick SL, et al. Case Study: Prolonged Infectious SARS-CoV-2 Shedding from an Asymptomatic Immunocompromised Individual with Cancer. Cell [Internet]. 2020;183(7):1901–1912.e9. Available from: [http://www.ncbi.nlm.nih.gov/pubmed/33248470](http://www.ncbi.nlm.nih.gov/pubmed/33248470) 6. 6.Baang JH, Smith C, Mirabelli C, Valesano AL, Manthei DM, Bachman MA, et al. Prolonged Severe Acute Respiratory Syndrome Coronavirus 2 Replication in an Immunocompromised Patient. J Infect Dis [Internet]. 2021;223(1):23–7. Available from: [http://www.ncbi.nlm.nih.gov/pubmed/33089317](http://www.ncbi.nlm.nih.gov/pubmed/33089317) 7. 7.Choi B, Choudhary MC, Regan J, Sparks JA, Padera RF, Qiu X, et al. Persistence and Evolution of SARS-CoV-2 in an Immunocompromised Host. N Engl J Med [Internet]. 2020;383(23):2291–3. Available from: [http://www.ncbi.nlm.nih.gov/pubmed/33176080](http://www.ncbi.nlm.nih.gov/pubmed/33176080) 8. 8.Tarhini H, Recoing A, Bridier-Nahmias A, Rahi M, Lambert C, Martres P, et al. Long term SARS-CoV-2 infectiousness among three immunocompromised patients: from prolonged viral shedding to SARS-CoV-2 superinfection. J Infect Dis [Internet]. 2021 Feb 8; Available from: [http://www.ncbi.nlm.nih.gov/pubmed/33556961](http://www.ncbi.nlm.nih.gov/pubmed/33556961) 9. 9.Walsh KA, Spillane S, Comber L, Cardwell K, Harrington P, Connell J, et al. The duration of infectiousness of individuals infected with SARS-CoV-2. J Infect [Internet]. 2020;81(6):847–56. Available from: [http://www.ncbi.nlm.nih.gov/pubmed/33049331](http://www.ncbi.nlm.nih.gov/pubmed/33049331) 10. 10.Wölfel R, Corman VM, Guggemos W, Seilmaier M, Zange S, Müller MA, 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%2F2021%2F06%2F01%2F2021.05.28.21258011.atom) 11. 11.van Kampen JJA, van de Vijver DAMC, Fraaij PLA, Haagmans BL, Lamers MM, Okba N, et al. Duration and key determinants of infectious virus shedding in hospitalized patients with coronavirus disease-2019 (COVID-19). Nat Commun [Internet]. 2021;12(1):8–13. Available from: [http://dx.doi.org/10.1038/s41467-020-20568-4](http://dx.doi.org/10.1038/s41467-020-20568-4) 12. 12.Liu Y, Yan LM, Wan L, Xiang TX, L. A, Liu JM, et al. Viral dynamics in mild and severe cases of COVID-19. Lancet Infect Dis. 2020;20(6):656–7. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F06%2F01%2F2021.05.28.21258011.atom) 13. 13.Chen J, Qi T, Liu L, Ling Y, Qian Z, Li T, et al. Clinical progression of patients with COVID-19 in Shanghai, China. J Infect [Internet]. 2020;80(5):e1–6. Available from: [http://www.ncbi.nlm.nih.gov/pubmed/32171869](http://www.ncbi.nlm.nih.gov/pubmed/32171869) 14. 14.Bullard J, Dust K, Funk D, Strong JE, Alexander D, Garnett L, et al. Predicting infectious severe acute respiratory syndrome coronavirus 2 from diagnostic samples. Clin Infect Dis. 2020;71(10):2663–6. 15. 15.Sheahan TP, Sims AC, Zhou S, Graham RL, Pruijssers AJ, Agostini ML, et al. An orally bioavailable broad-spectrum antiviral inhibits SARS-CoV-2 in human airway epithelial cell cultures and multiple coronaviruses in mice. Sci Transl Med [Internet]. 2020;12(541). Available from: [http://www.ncbi.nlm.nih.gov/pubmed/32253226](http://www.ncbi.nlm.nih.gov/pubmed/32253226) 16. 16.Painter WP, Holman W, Bush JA, Almazedi F, Malik H, Eraut Ncje, et al. Human Safety, Tolerability, and Pharmacokinetics of Molnupiravir, a Novel Broad-Spectrum Oral Antiviral Agent with Activity Against SARS-CoV-2. Antimicrob Agents Chemother [Internet]. 2021;(March). Available from: [http://www.ncbi.nlm.nih.gov/pubmed/33649113](http://www.ncbi.nlm.nih.gov/pubmed/33649113) 17. 17.CDC Diagnostic Test for COVID-19 [Internet]. 2020 [cited 2021 Apr 12]. Available from: [https://www.cdc.gov/coronavirus/2019-ncov/lab/virus-requests.html](https://www.cdc.gov/coronavirus/2019-ncov/lab/virus-requests.html) 18. 18.Premkumar L, Segovia-Chumbez B, Jadi R, Martinez DR, Raut R, Markmann A, et al. The receptor binding domain of the viral spike protein is an immunodominant and highly specific target of antibodies in SARS-CoV-2 patients. Sci Immunol [Internet]. 2020 Jun 11;5(48):eabc8413. Available from: [http://www.ncbi.nlm.nih.gov/pubmed/32527802](http://www.ncbi.nlm.nih.gov/pubmed/32527802) 19. 19.Zou G. A Modified Poisson Regression Approach to Prospective Studies with Binary Data. Am J Epidemiol. 2004;159(7):702–6. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/aje/kwh090&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=15033648&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F06%2F01%2F2021.05.28.21258011.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000220484900009&link_type=ISI) 20. 20.Buckland MS, Galloway JB, Fhogartaigh CN, Meredith L, Provine NM, Bloor S, et al. Treatment of COVID-19 with remdesivir in the absence of humoral immunity: a case report. Nat Commun [Internet]. 2020;11(1):6385. Available from: [http://www.ncbi.nlm.nih.gov/pubmed/33318491](http://www.ncbi.nlm.nih.gov/pubmed/33318491) 21. 21.Borremans B, Gamble A, Prager KC, Helman SK, McClain AM, Cox C, et al. Quantifying antibody kinetics and RNA detection during early-phase SARS-CoV-2 infection by time since symptom onset. Elife [Internet]. 2020;9:1–27. Available from: [http://www.ncbi.nlm.nih.gov/pubmed/32894217](http://www.ncbi.nlm.nih.gov/pubmed/32894217) 22. 22.Huang CG, Lee KM, Hsiao MJ, Yang SL, Huang PN, Gong YN, et al. Culture-based virus isolation to evaluate potential infectivity of clinical specimens tested for COVID-19. J Clin Microbiol. 2020;58(8):1–8. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1128/JCM.02015-19&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F06%2F01%2F2021.05.28.21258011.atom) 23. 23.Folgueira MD, Luczkowiak J, Lasala F, Pérez-Rivilla A, Delgado R. Prolonged SARS-CoV-2 cell culture replication in respiratory samples from patients with severe COVID-19. Clin Microbiol Infect [Internet]. 2021 Feb 22; Available from: [http://www.ncbi.nlm.nih.gov/pubmed/33631334](http://www.ncbi.nlm.nih.gov/pubmed/33631334) 24. 24.Singanayagam A, Patel M, Charlett A, Lopez Bernal J, Saliba V, Ellis J, et al. Duration of infectiousness and correlation with RT-PCR cycle threshold values in cases of COVID-19, England, January to May 2020. Euro Surveill [Internet]. 2020;25(32). Available from: [http://www.ncbi.nlm.nih.gov/pubmed/32794447](http://www.ncbi.nlm.nih.gov/pubmed/32794447) 25. 25.Gottlieb RL, Nirula A, Chen P, Boscia J, Heller B, Morris J, et al. Effect of bamlanivimab as monotherapy or in combination with etesevimab on viral load in patients with mild to moderate COVID-19: A randomized clinical trial. JAMA - J Am Med Assoc. 2021;325(7):632–44. 26. 26.Chen P, Nirula A, Heller B, Gottlieb RL, Boscia J, Morris J, et al. SARS-CoV-2 Neutralizing Antibody LY-CoV555 in Outpatients with Covid-19. N Engl J Med. 2021;384(3):229–37. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F06%2F01%2F2021.05.28.21258011.atom) 27. 27.Weinreich DM, Sivapalasingam S, Norton T, Ali S, Gao H, Bhore R, et al. REGN-COV2, a Neutralizing Antibody Cocktail, in Outpatients with Covid-19. N Engl J Med. 2021;384(3):238–51. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F06%2F01%2F2021.05.28.21258011.atom) 28. 28.Rossen LM, Branum AM, Ahmad FB, Sutton P, Anderson RN. Excess Deaths Associated with COVID-19, by Age and Race and Ethnicity — United States, January 26–October 3, 2020. MMWR Morb Mortal Wkly Rep. 2020;69(42):1522–7. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.15585/mmwr.mm6942e2&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=33090978&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F06%2F01%2F2021.05.28.21258011.atom) 29. 29.Jeong HW, Kim S-M, Kim H-S, Kim Y-I, Kim JH, Cho JY, et al. Viable SARS-CoV-2 in various specimens from COVID-19 patients. Clin Microbiol Infect [Internet]. 2020 Nov;26(11):1520–4. Available from: [http://www.ncbi.nlm.nih.gov/pubmed/32711057](http://www.ncbi.nlm.nih.gov/pubmed/32711057) 30. 30.Nalbandian A, Sehgal K, Gupta A, Madhavan M V, McGroder C, Stevens JS, et al. Post-acute COVID-19 syndrome. Nat Med [Internet]. 2021 Mar 22; Available from: [http://www.ncbi.nlm.nih.gov/pubmed/33753937](http://www.ncbi.nlm.nih.gov/pubmed/33753937)