SARS-CoV-2 viral replication persists in the human lung for several weeks after symptom onset ============================================================================================= * M Tomasicchio * S Jaumdally * A Pooran * A Esmail * L Wilson * A Kotze * L Semple * S Meier * K Pillay * R Roberts * R Kriel * R Meldau * S Oelofse * C Mandviwala * J Burns * R Londt * M Davids * C van der Merwe * A Roomaney * L Kühn * T Perumal * A.J Scott * M.J Hale * V Baillie * S Mahtab * C Williamson * R Joseph * A Sigal * I Joubert * J Piercy * D Thomson * DL Fredericks * MGA Miller * M.C Nunes * S.A Madhi * K Dheda ## ABSTRACT **Rationale** In the upper respiratory tract replicating (culturable) SARS-CoV-2 is recoverable for ∼ 4 to 8 days after symptom onset, however, there is paucity of data about the frequency or duration of replicating virus in the lower respiratory tract (i.e. the human lung). **Objectives** We undertook lung tissue sampling (needle biopsy), shortly after death, in 42 mechanically ventilated decedents during the Beta and Delta waves. An independent group of 18 ambulatory patents served as a control group. **Methods** Lung biopsy cores from decedents underwent viral culture, histopathological analysis, electron microscopy, transcriptomic profiling and immunohistochemistry. **Results** 38% (16/42) of mechanically ventilated decedents had culturable virus in the lung for a median of 15 days (persisting for up to 4 weeks) after symptom onset. Lung viral culture positivity was not associated with comorbidities or steroid use. Delta but not Beta variant lung culture positivity was associated with accelerated death and secondary bacterial infection (p<0.05). Nasopharyngeal culture was negative in 23.1% (6/26) of decedents despite lung culture positivity. This, hitherto, undescribed bio-phenotype of lung-specific persisting viral replication was associated with an enhanced transcriptomic pulmonary pro-inflammatory response but with concurrent viral culture positivity. **Conclusions** Concurrent, rather than sequential active viral replication continues to drive a heightened pro-inflammatory response in the human lung beyond the second week of illness and was associated with variant-specific increased mortality and morbidity. These findings have potential implications for the design of interventional strategies and clinical management of patients with severe COVID-19 disease. **Scientific Knowledge on the Subject** Investigations to understand SARS-CoV-2 viral shedding (determined by PCR or antigen testing) have extensively focused on samples from the upper respiratory tract. The widely accepted view is that acute severe SARS-CoV-2 infection is characterised by a viral replicative phase in the first week of symptomatic illness followed by a pro-inflammatory immunopathologic phase peaking in the second and third weeks of illness. However, it remains unclear whether detection of SARS-CoV-2 beyond 2 weeks after symptom onset in published studies represent active replication competent virus because it may represent residual genomic or antigenic material in the tissue. **What This Study Adds to the Field** We have identified a, hitherto, undescribed bio-phenotype of acute severe COVID-19 characterised by persisting viral replication in the lung for up to 4 weeks after symptom onset. ∼40% of acute severe COVID-19 intensive care unit (ICU) decedents (n=42) had nasopharyngeal swab culture positivity at ∼2 weeks post-symptom onset versus only ∼5% in a group of ambulatory control patients (n=18). There was compartment-specific (nasopharynx versus lung) discordance. The phenotype of lung-specific persisting viral replication was associated with variant-specific accelerated death, an exaggerated inflammatory response, and attenuated T-cell immunity in the lung (based on histopathological and transcriptomic studies). This challenges the traditional view that viral replication occurs during the first 5 to 10 days of illness, which is followed by an effector or hyperinflammatory phase. This is the first study, to our knowledge, to systematically culture virus from the human lung and map out its related clinical determinants, and which describes the human lung transcriptomic profile of culture-positive versus culture-negative patients with severe COVID-19 disease. Keywords * COVID-19 * SARS-CoV-2 * virus replication * mechanically ventilated patients * upper respiratory tract * immunology ## Introduction Coronavirus disease-19 (COVID-19) caused by the Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) has been the foremost killer globally over the last 3 years. Case fatality risk in hospitalised patients, and particularly in mechanically ventilated patients, during the Beta and Delta waves was particularly high [∼50%-70%; (1)]. Even with the Omicron-related variants, case fatality risk remains significant in the elderly and immunocompromised persons, and in several countries including the UK, Italy, France, Brazil, and prominently in China where there is now an ongoing epidemic of severe COVID-19 disease (2–10). Better therapeutic interventions are needed. However, despite considerable research, the pathogenesis of severe COVID-19, relative to viral kinetics, remains incompletely understood. SARS-CoV-2 detection (ascertained through PCR positivity or antigen detection) can persist for several weeks from symptom onset (11). Post-mortem studies have shown persistence of SARS-CoV-2 in tissues detected by PCR and immunohistochemistry for up to several weeks after symptom onset (12, 13). However, detection of SARS-CoV-2 in these studies may not represent replication competent virus (detectable only by viral culture) but residual genomic or antigenic material in the tissues. Shedding of replicating virus confirmed through serial viral culture (i.e. *in vitro* replication in human cell lines) from the upper respiratory tract (URT) has been shown to persist for only ∼2 to 8 days after symptom onset (11, 14–23). These findings have been confirmed in human lung challenge studies with viable pathogen where virus was cultured from the URT until a median of 4 days (and a maximum of 10 days) from symptom onset (24). However, hardly anything is known about the compartment-specific duration of actively replicating virus in the lower respiratory tract (LRT), particularly in acute severely ill hospitalised patients undergoing mechanical ventilation. We hypothesised that there is compartment-specific uncoupling of viral replication in severe COVID-19 i.e. replicating virus can persist in the LRT beyond 10 days from symptoms onset, independent of its persistence in the URT, and this persistence may be associated with an altered pulmonary immunity. ## Methods ### Patients The decedents (n=42) were recruited from Chris Hani Baragwanath Academic Hospital, Johannesburg, South Africa (n=18; Beta group) and Groote Schuur Hospital, Cape Town, South Africa (n=24; Delta group). Figure 1A outlines an overview of the study plan. Ambulatory controls (n=18) were recruited at diagnosis (baseline; ∼5 days from symptom onset), 7 days and 14 days post diagnosis. Minimally invasive tissue samples (MITS) and nasopharyngeal swabs from decedents (n=42) in the Beta and Delta waves (Figure 1B) were taken immediately after death. In addition, heart, liver, kidney, and adipose tissue samples were also taken from the Delta variant decedent cohort only. Ethical approval was obtained from the Human Research Ethics Committee (HREC) of the University of Cape Town (HREC approval number 866/2020) and University of Witwatersrand (HREC approval number M200313). Biosafety approvals were obtained from the Faculty Biosafety Committee of the University of Cape Town (IBC008-2021). ![Figure 1.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2023/08/23/2023.03.06.23286834/F1.medium.gif) [Figure 1.](http://medrxiv.org/content/early/2023/08/23/2023.03.06.23286834/F1) Figure 1. Study overview including SARS-CoV-2 PCR-positive ambulatory controls (A) and mechanically ventilated decedents (B) recruited during the Beta and Delta waves. Nasopharyngeal (NP) swabs from ambulatory COVID-19 controls were obtained approximately 5 days after symptom onset (diagnosis), and then at 12- and 19-days post symptom onset. Minimally invasive tissue samples (MITS) and NP swabs were retrieved from decedents shortly after death. ### Viral culture To establish the *in vitro* viral culture model, a SARS-CoV-2 viral stock was used to infect the human lung carcinoma cell line, H1299 ACE2, in a BSL3 laboratory and infection was confirmed by light microscopy (as assessed by cytopathic effects of the virus on the cell line) and confocal microscopy (Figure S1A and B). Serial dilutions of the viral stock were used to establish the limit of detection of the PCR assay at 1×101 copies/ml (Figure S1C). Viral culture was performed on the nasopharyngeal swab and lung biopsy samples as indicated in the study overview (Figure 1) and detailed in the online supplement. Viral culture result reproducibility was good (see online supplement). ### Multiplex PCR to detect secondary bacterial infections The lung biopsy cores, stored in universal transport medium, were briefly homogenised and 200µl of the supernatant was applied to the BioFireÒ FilmArrayÒ Pneumonia panel (Bioméieux, South Africa). The panel was run using protocol BAL v3.3 according to the manufacturer’s instructions, thus generating RT-PCR readouts for 33 bacterial and viral pathogens. Bronchopneumonia was defined as histological evidence of a neutrophilic alveolar infiltration together with the detection of bacterial genomic material in the biopsy cores. ### Immunohistochemistry Immunohistochemical staining was performed using the Roche Ventana Automated platform (Ventana XT autostainer) as indicated by the manufacturer. Tissue sections were prepared, stained, and viewed using standard techniques (25). Antibodies included anti-CD3 (2GV6), anti-CD4 (SP35), anti-CD8 (SP57) and anti-CD68 (KP-1) (Roche USA). ### Haematoxylin & eosin (H&E) staining and transmission electron microscopy (TEM) H&E staining and TEM were performed according to standard procedures (25). H&E-stained slides were viewed using an Olympus BX43 microscope. TEM tissue sections were viewed using a Carl Zeiss EM109 microscope. ### SARS-CoV-2 whole genome sequencing Total SARS-CoV-2 RNA was extracted from lung biopsy samples and whole genome sequencing was performed. The generated reads were analysed with the Exatype ([https://exatype.com](https://exatype.com)) software to identify minor and major variants. The assembled consensus sequences were analysed using Nextclade Web ([https://clades.nextstrain.org](https://clades.nextstrain.org)) for further quality control and clade assignment. ### RNAseq Total RNA was extracted from lung biopsy samples from the Delta group, sequenced and mapped consecutively to the human and COVID reference genomes using the Spliced Transcripts Alignment to a Reference (STAR) software [version 2.7.7a, (26)]. A differential expression (DE) analysis was performed on the generated raw read count file with the edgeR (Version 3.38.4) R package (27). The DE results were ranked by fold change and the gseGO function, from the clusterProfiler R clusterProfiler [Version 4.0, (28)] R package was used to perform a gene set enrichment analysis (GSEA) for the Gene Ontology Biological Process pathways. Pathways with an FDR <0.05 were considered significant. ### Confocal microscopy The H1299 ACE2 cells were plated, infected with SARS-CoV-2 and allowed to adhere to coverslips slides overnight at 37°C. The next day the cells were stained with or without anti-SARS-CoV-2 S1 spike protein (ThermoFisher, USA) and the slides were mounted in Mowiol (Calbiochem, USA) containing n-propyl gallate (Sigma-Aldrich, Germany) as an anti-fading agent. Confocal microscopy was performed with a Zeiss Axiovert 200M LSM510 Meta NLO Confocal Microscope. ### Sample size calculation and statistical analysis We hypothesised that we would detect lung culture positivity at 14 days post-symptom onset in ∼33% of decedents. A sample size of ∼40 participants would allow us to ascertain that level of positivity with a 15% margin of error using 95% confidence and 80% power (OpenEpi, Version 3, opensource calculator). The Fisher Exact test was employed for categorical variables and for continuous variables, Mann-Whitney test was used for non-parametrically distributed data between the culture-negative and culture-positive groups (Stata version 17 or GraphPad, Version 9.4.1). A p-value of < 0.05 was considered significant for all statistical analyses. The multivariable analysis was performed in R by fitting a binomial Generalized Linear Model (GLM) to assess the association between steroid use and the presence of secondary bacterial infection on culture status. The tidymodels (version, 1.0.0) R package was used to perform predictive modelling using the glm binomial classification algorithm. To account for the small sample size, 1000 bootstraps were performed for each analysis. ## Results ### Demographics and clinical characteristics of the decedents The demographics of patients enrolled in the study are shown in Table S1. The median age of the patients was 53 years with 48% being males (20/42). 40.5% (17/42) had a secondary bacterial infection and 11% (4/38) had bacterial bronchopneumonia (microbiologically and histopathological confirmed). The median time from onset of symptoms to death, ICU admission to death and high flow oxygen admission to death was 17 (IQR; 9-22), 5 (2–12) and 11 (6–15) days, respectively. ### SARS-CoV-2 replicating persistence in the human lung of mechanically ventilated decedents We first ascertained the frequency and duration of replicating virus in lung tissue (which to our knowledge has not been previously undertaken). Culturable virus in the lung was present in 38.1% (16/42; Figure 2A) of mechanically ventilated ICU decedents, at a median of 15 days (and up ∼4 weeks; Figure S2) from symptom onset to sampling/death (Figure 3A). As expected, 56% (10/18) of a prospectively recruited control group of ambulatory patients had culturable virus, using nasopharyngeal swab samples, at day 5 from symptom onset (Figure 2B). In the same group of patients after 12- and 19-days after symptom onset, only 5.5% (1/18) and 0% (0/18), respectively, had culturable virus from their nasopharyngeal swab (Figure 2B). By contrast, 38% of nasopharyngeal swabs from the mechanically ventilated ICU descendants had culturable virus (Figure 2B), at a median of 13 days from symptom onset to sampling/death (Figure S3B). Additionally, SARS-CoV-2 could be detected by PCR in multiple organs in lung culture-positive decedents in the Delta cohort (biopsies other than the lung were not performed in the Beta cohort) suggesting widespread multi-organ viral dissemination (Figure 2C). SARS-CoV-2 was also detected in adipose tissue of culture-positive decedents (hitherto undescribed). Clinical characteristics, such as, age and comorbidities were similar in the lung culture-positive versus culture-negative groups (Table S1). We found no association between viral genetic variant and the phenotype of replicating viral persistence (although this might have been a factor of the limited sample size; Table S5). ![Figure 2.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2023/08/23/2023.03.06.23286834/F2.medium.gif) [Figure 2.](http://medrxiv.org/content/early/2023/08/23/2023.03.06.23286834/F2) Figure 2. Active replicating virus was recovered from the lungs of over one third of decedents (16/42). **(A)** Proportion of lung biopsy samples that were culture-positive from the decedents. **(B)** Proportion of ambulatory patient and decedent NP swab samples that were culture-positive. **(C)** PCR positivity of organs of lung culture-positive decedents from the Delta cohort. NP= nasopharyngeal. ![Figure 3.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2023/08/23/2023.03.06.23286834/F3.medium.gif) [Figure 3.](http://medrxiv.org/content/early/2023/08/23/2023.03.06.23286834/F3) Figure 3. The phenotype of replicating viral persistence, compared to the culture-negative participants, was associated with accelerated death and a higher frequency of bacterial bronchopneumonia in the Delta but not the Beta group. (**A**) The days from symptom onset to death for the culture-negative (-ve; green) and culture-positive (+ve; red) groups for both groups combined, and for the Beta (**B**) and Delta (**C**) groups alone. Proportion of samples/participants with a secondary bacterial infection in culture-negative and culture-positive decedents overall i.e. the combined groups (Beta and Delta) (**D**), Beta group only (**E**), and Delta group only (**F**). **(G)** PCR cycle threshold (Ct) value at the time of death or at admission could not discriminate or predict lung culture status. The nasopharyngeal swab PCR Ct values at admission or death were missing for some participants because they were either diagnostically confirmed by antigen testing or the Ct value was not recorded. Due to the nature of the pandemic and the burden of the disease on the healthcare infrastructure at the time, Ct values at peak periods were not recorded. We have conducted sensitivity and imputation analyses indicating that these missing data points are redundant. The nasopharyngeal swab PCR Ct values at admission or death were missing for some participants because they were either diagnostically confirmed by antigen testing or the Ct value was not recorded. Due to the nature of the pandemic and the burden of the disease on the healthcare infrastructure at the time, Ct values at peak periods were not recorded. We have conducted sensitivity and imputation analyses indicating that these missing data points are redundant. ### Time to death in the Delta and Beta groups and predictors of lung culture positivity Next, we evaluated variant-specific relationships to clinical outcomes. Mechanically ventilated patients who were SARS-CoV-2 lung culture-positive in the Delta, but not the Beta group, had accelerated death (i.e. shorter duration from symptom onset to death; Figure 3C versus 3B; p=0.004), and a higher proportion of lung-specific secondary bacterial infection (Figure 3F versus 3E; p=0.032) compared to culture-negative decedents. Similarly, to the lung culture data, the nasopharyngeal swab culture-positive Delta, but not the Beta group, had accelerated death (Figure S3D versus S3C; p=0.026). The bacterial species identified from the lung biopsies of both the Beta and Delta groups included *Streptococcus, Staphylococcus, Haemophilus, Acinetobacter*, Proteus spp, *Escherichia, Klebsiella, Enterobacter* and *Serratia* (Table S4). Overall, both groups were infected with one or more bacteria that were sensitive or resistant to β-lactams and/or carbapenems (Table S4). Key clinical and demographic characteristics such as differences in co-morbidities (age, obesity, diabetes, HIV positivity etc; Table S1) associated as drivers of severe COVID-19 disease and poor prognosis, could not explain these observations, despite the lower population-level vaccination and pre-existing COVID-19 exposure rates in the Beta cohort. Steroid usage was similar in the culture-positive and culture-negative groups (Table S1) and there was no significant (p>0.05) association between steroid use and the presence of secondary bacterial infection in a multivariable analysis. Next, we interrogated whether nasopharyngeal PCR characteristics (Ct value), either at admission or close to death, could identify the phenotype of lung replicating viral persistence. However, nasopharyngeal Ct neither at admission, nor at the time of death was associated with lung culture positivity (Figure 3G). This suggests that the kinetics of viral replication was different in the upper and the lower respiratory tract. ### Lung immunity and histology of the culture-negative versus culture-positive groups We then ascertained whether the phenotype of replicating viral persistence was associated with attenuated or modulated lung immunity in the Delta decedents (transcriptomic and flow cytometric studies were only carried out in Cape Town, i.e the Delta decedents, due to location-specific availability of assays and limited Beta group biopsy cores that had been used for unrelated studies). Immunohistochemical staining indicated that there was significantly less infiltration of CD3+ T-cells, specifically CD8+ T-cells in the alveoli and interstitium of the SARS-CoV-2 culture-positive compared with the culture-negative individuals in the Delta decedents (Figure 4A and B). ![Figure 4.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2023/08/23/2023.03.06.23286834/F4.medium.gif) [Figure 4.](http://medrxiv.org/content/early/2023/08/23/2023.03.06.23286834/F4) Figure 4. A higher proportion of T-cells, macrophages and pneumonocytes infiltrate into the lung of the culture-negative versus culture-positive decedents in the Delta group. **(A)** More CD3+ and CD8+ T-cells infiltrate into the alveoli and interstitial space of the lung culture-negative versus culture-positive group in the Delta decedents as assessed by immunohistochemistry. **(B)** Representative images (immunohistochemistry) showing increased T-cell infiltration into interstitial space (blue arrow) in the lung culture-negative versus the culture-positives in the Delta cohort. Histopathology findings **(C)** and representative images **(D)** associated with diffuse alveolar damage and microvascular thrombosis in the Delta decedents. The black arrows indicate key histopathological features. The typical histological features of severe COVID-19 (e.g. diffuse alveolar damage and microvascular thrombosis) were similar in the SARS-CoV-2 culture-positive and the culture-negative phenotype suggesting that these events occurred in the early rather than the persistent viral replication phase (Figure 4C, D and S4; Table S2, S3). Interestingly, we observed that some features of leucocyte hyperactivation (i.e., hemophagocytic syndrome) were more common in the SARS-CoV-2 culture-negative versus the culture-positive group, potentially in keeping with an aberrant immune response characterised by a lack of immune regulation, as outlined above (Figure 4C and 4D; p=0.013). The transcriptional analysis of post-mortem lung tissue identified 630 up- and 885 downregulated genes at an uncorrected p-value ≤ 0.05 when comparing the culture-positive versus negative-groups (Figure S5). After adjustment for multiple testing, the same comparison identified a total of 11 up- and 4 down-regulated genes (FDR<0.05; specific genes discussed further in the online supplement; Figure S6). The GSEA performed using the full list of differentially expressed genes ranked by fold-change, identified activated pathways that were associated with a proinflammatory response related to cytokine signalling, neutrophil and monocyte chemotaxis/recruitment, and viral entry/defence, all of which are implicated in COVID-19-related hypercytokinaemia (29) (Figure 5, S5, S6 and S7, Table S6 and S7A). Significantly repressed pathways were generally associated with body homeostasis (Figure 5A and B). There was also in tandem upregulation of Th1 and Th17 signalling pathways (Table S7B) but to a substantially lesser extent than that of innate cellular and signalling pathways (IL-1, IL-6 and neutrophil-related; Table S7A). These features may be consistent with an aberrant immune response including a lack of activation of regulatory and immune-suppressive pathways. T-cell exhaustion consistent with upregulation of PD-1, CTLA-4 and LAG (Table S7C) known to be associated with severe COVID-19, was not observed. ![Figure 5.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2023/08/23/2023.03.06.23286834/F5.medium.gif) [Figure 5.](http://medrxiv.org/content/early/2023/08/23/2023.03.06.23286834/F5) Figure 5. The transcriptomic analysis revealed that the culture-positive group, in comparison to the culture-negative group, had enrichment of activated pathways associated with inflammation, innate immunity, responses to cytokines, and responses to virus/ bacterial stimuli in the Delta descendants. Dot plot illustrating the significantly activated and suppressed pathways along with the gene count and ratio for each pathway (**A**), enrichment map illustrating the significantly activated and suppressed pathways along with the gene count and ratio for each pathway (**B**) and cnetplot similarly illustrating the overlap of genes and their fold changes for selected activated pathways (**C**). The differential expression (DE) results also revealed that a number of SARS-CoV-2 genes were significantly upregulated (FDR<0.01) in the culture-positive versus the culture-negative group including *nucleocapsid phosphoprotein* (log2 FC=8.4) and *ORF3a* (log2 FC=5.5) while the *surface/spike glycoprotein* encoding gene had a log2 FC of 5.3 and an FDR of 0.067 (Table S6). A visual inspection of the mapped SARS-CoV-2 reads revealed that those that mapped to the 5’ end of the genes were spliced with a portion mapping to the 5’ leader sequence of the genome. This suggests the reads originated from sub-genomic mRNA (sgRNA) rather than genomic RNA which is consistent with the active viral replication observed in the culture-positive group. Finally, we evaluated whether any of the DE genes could act as biomarkers discriminating between lung culture-positive and negative-individuals. Logistic regression predictive modelling revealed that *GREM1* and *FGFBP1* were associated with a sensitivity and specificity above 90% (further details in the online supplement). Future studies are warranted to determine if these lung-based biomarkers can predict patient culture status in blood samples. ## Discussion The widely accepted view in severe acute COVID-19 is that resolution of the initial viral replication phase in the first week after symptom onset is followed by an effector or hyperinflammatory phase in the second and third week of illness which is characterised by diffuse alveolar damage, thrombo-inflammation, and endotheliopathy (30). Indeed, the Infectious Disease Society of America (IDSA) recommends the use of remdesivir for only 5 days in patients with severe illness and not in mechanically ventilated patients (31). However, our results, based on post-mortem lung biopsies obtained using minimally invasive tissue sampling methods (MITS) shortly after death indicated that, in contradistinction to the URT where replication often ceases within ∼8 days from symptom onset, in the human lung virus is culturable in ∼40% of mechanically ventilated patients until death (median of 15 days and up to 4 weeks after symptom onset; see Figure 1 for the study overview). The culture-positive group in the Delta cohort had accelerated death and a higher proportion of secondary bacterial infection in the lung compared to the culture-negative group. Nasopharyngeal SARS-CoV-2 viral load (based on Ct value) neither at admission nor at death, was predictive of lung culture-positivity. SARS-CoV-2 culture-positivity in the lung of decedents was associated with attenuated pulmonary T-cell immunity and an exaggerated pro-inflammatory phenotype. Importantly, this was concurrent with, rather than sequential to the viral replication or viral culture-positive phase. These findings challenge the traditional paradigm of an initial viral replicative phase in the first week of severe illness sequentially followed by an effector or inflammatory phase (30). Our data suggest that in ∼40% of ventilated patients, viral replication persisted until death (i.e. 3rd and 4th week of illness and a median of 15 days after symptom onset) compared to ∼2 to ∼8 days in the URT as outlined in several studies including a live virus human challenge study (11, 14–24). One outlier study reported culturing virus from the URT for up to 3 weeks after symptom onset (32). However, a large proportion of patients were immunocompromised, samples at diagnosis and follow-up were combined (skewing the results), a high proportion of participants were healthcare workers (re-infection may have been a confounder), and as the authors suggested a limitation was that the Vero cell line used was overtly permissive to infection compared to the human lung carcinoma cell line, H1299 ACE2, which is a biologically representative cell line (and one that we used). Another recent study showed that infectious virus production peaked in the human lung within 2 days, but this model used *ex vivo* agarose infused devascularised and explanted human lung slices, which are not representative of what is occurring in freshly harvested human lung (33). The culture-based findings in the afore-mentioned studies must be explicitly distinguished from studies that detected residual free viral genomic RNA (but not replicating virus) embedded in the respiratory tract tissue of patients that had severe disease for an extended period of time (13, 34, 35). Indeed, SARS-CoV-2 RNAs have been detected in patient tissue many months after recovery from acute infection (36–38). It was initially suggested that sgRNA (sub-genomic RNA; small strands of reversely transcribed RNA) could be used as a proxy to infer viral replication. However, several recent studies have indicated that it has poor predictive value as a proxy for viral replication (39, 40). Indeed, Stein et al (13) detected sgRNA in multiple post-mortem organ biopsies, including the brain, several months after symptom onset. Thus, the data presented in this manuscript is the first to do so conclusively and comprehensively using viral culture from lung tissue (the gold standard to detect viral replication) beyond two weeks after symptom onset. We demonstrated active viral replication in the lungs of acutely ill ventilated patients for up to ∼4 weeks after symptom onset. This challenges the current practice of using antivirals like remdesivir for only 5 days and suggests that a longer duration of treatment may be required in critically ill patients. Furthermore, antivirals like remdesivir are not recommended for use by IDSA in mechanically ventilated patients (conditional recommendation) as they felt that such patients (often in the third week of their illness) are no longer in the viral replicative phase, and published controlled trial data showed no mortality benefit of remdesivir in such patients (31, 41). However, these studies demonstrated a group effect, and the analyses did not adjust for disease severity or the time from symptom onset to death in mechanically ventilated patients (42). Our data suggest that a significant number of patients may likely benefit from antivirals during mechanical ventilation. Indeed, several observational studies have found a survival benefit using remdesivir in mechanically ventilated patients, but this requires further clarification in appropriate trials (42–44). In a multivariate analysis we found no association between steroid usage and lung viral culture positivity - in fact, steroid usage was lower in the viral culture-positive group and often culture positivity persisted beyond the 10 days of steroid usage. The transcriptomic data suggested that, in a significant number of patients, the hyperinflammatory and viral replication phase occur concurrently in the 3rd and 4th week of illness, in contradistinction to the widely held view that these are sequential phases. Antiviral and selective proinflammatory responses were over-represented in the SARS-CoV-2 culture-positive compared with the culture-negative decedents, and we did not detect attenuated type 1 interferon responses at the site of disease compared with other reports (45–49). Three prior studies (one that enrolled 5 COVID-19 patients) evaluated transcriptomic lung responses in patients with severe COVID-19 versus healthy controls (45, 46, 50). These first level studies logically attempted to address the significance of transcriptomic changes specific to COVID-19 by using healthy controls or non-diseased parts of the lung from lung cancer patients. However, we specifically sought to compare culture-positive versus culture-negative groups (hitherto not undertaken) to dissect out pathways that facilitate permissiveness to ongoing viral replication. We identified two lung-based biomarkers (*GREM1* and *FGFBP1*) that could predict culture-positivity. Although these are lung-specific biomarkers, this preliminary analysis in a limited number of samples suggests that in the future, RT-PCR of tracheal aspirates or blood (if they are concordant with lung findings), could potentially serve as biomarkers to identify and direct appropriate treatment protocols to culture-positive persons but further investigation is needed. There are several limitations to our findings. Firstly, our findings are relevant to acute severe COVID-19 ARDS/pneumonia requiring mechanical ventilation and may not be applicable to milder forms of disease seen in hospitalised patients or chronic infection seen in immunocompromised patients. Secondly, we only studied patients with the Beta and Delta variants as these were the predominant variant at the time of the study. However, Omicron has also been associated with severe disease in several settings including the surge of severe COVID-19 unfolding in China. Thirdly, we did not study a control group comprising severe ARDS due to other causes because our express aim was to investigate the presence and duration of viral replication in the LRT in severe COVID-19 disease. Fourth, the sample size limited our ability to make conclusions about several aspects. However, the highly resource intensive and demanding nature of the study limited our ability to recruit higher numbers of participants. Fifthly, it could be suggested that there may have been sampling error and variability of the viral culture assay. However, the reproducibility of the viral culture technique using 6 samples across 2 separate runs had a low standard error, which was indicative of high reproducibility. Finally, the transcriptional signature and flow cytometric findings may have been affected by post-death sampling, but several detailed studies have shown (51) that most protein and RNA species are preserved and stable for several hours after death. Given that biopsies for the transcriptional studies were taken ∼2 hours after death, we feel they are broadly representative of the picture at the time of death. In summary, our data suggest that in COVID-19 disease there is considerable heterogeneity in the frequency and duration of viral replication in the upper versus the lower respiratory tract (i.e. lungs) beyond the 2nd week of illness, and that in a significant proportion of seriously ill patients, persisting viral replication occurs concurrently and may drive an exaggerated proinflammatory response (higher than in culture-negative persons), rather than sequentially as it is widely believed. These findings have potential implications for the use of antiviral therapy in seriously ill patients with COVID-19 and suggest that better biomarkers are needed to identify patient phenotypes and subsets that might benefit from concurrent anti-inflammatory and antiviral therapy. ## Supporting information Supplementary material [[supplements/286834_file03.docx]](pending:yes) ## Data Availability Individual participant data will be made available to researchers who provide a protocol that is approved by their respective human research ethics committee. All protocols will be reviewed and approved by the MITS consortium trial steering committee up to five years following publication. A data sharing agreement (DTA) will need to be concluded between the representatives of the requesting institution and the University of Cape Town Lung Institute. Data sharing requests should be directed to keertan.dheda@uct.ac.za ## Contributions K.D, MT, S.J, A.P, A.E, M.D, M.N and S.M conceived and designed experiments. K.D, A.E, S.O, L.K, T.P, A.S, I.J, J.P, D.T, D.F, M.M, M.N and S.M arranged medical ethical approval, recruitment of study participants and collection of study material. MT, SJ. A.P, L.W, A.K, S.M, K.P, R.R, R.K, R.M, C.M, J.B, R.L, M.D, C vdM, A.R, M.H, V.B, S. M, C.W, and R.J performed the experiments. M.T, S.J, A.P and M.D set up experimental assays. A.S provided the cell line. K.D, M.T, S.J, A.P, L.W, A.K, L.S, S.M, K.P, R.R, R.L, M.H, V.B, S.M, R.J, and N.M analysed and interpreted data. K.D, M.T, L.S, and S.M wrote the manuscript with input from all listed authors. ## Declaration of interests The authors have no competing interests. ## Acknowledgments The authors would like to thank Arnold-Day C, Crowther M, Fernandes N, and Mitchell L from the Division of Critical Care, Department of Anaesthesia and Perioperative Medicine, University of Cape Town, South Africa for identifying and referring potential patients into the study. The study was funded by the Bill & Melinda Gates Foundation (grant number INV-017282) and the South African Medical Research Council (grant number SHIP NCD 96756) with partial support from the Department of Science and Technology and National Research Foundation: South African Research Chair Initiative in Vaccine Preventable Diseases. The KD lab acknowledges funding from the SA MRC (RFA-EMU-02-2017), EDCTP (TMA-2015SF-1043, TMA-1051-TESAIII, TMA-CDF2015), UK Medical Research Council (MR/S03563X/1), NIH (CRDF-OISE-16-62105) and the Wellcome Trust (MR/S027777/1). This work was co-funded by The Wellcome Centre for Infectious Diseases Research in Africa is supported by core funding from the Wellcome Trust (230135/Z/16/Z) and the European Union’s Horizon Europe Research and Innovation Actions (101046041) for genomic surveillance. The authors would like to thank the families of the deceased who gave us permission to conduct the study, which may advance our understanding of SARS-CoV-2 pathogenesis to inform future clinical management of respiratory pathogen pandemics. ## Footnotes * The manuscript was amended according to reviewers comments * Received March 6, 2023. * Revision received August 22, 2023. * Accepted August 23, 2023. * © 2023, 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.Davies MA, Morden E, Rosseau P, Arendse J, Bam JL, Boloko L, Cloete K, Cohen C, Chetty N, Dane P, Heekes A, Hsiao NY, Hunter M, Hussey H, Jacobs T, Jassat W, Kariem S, Kassanjee R, Laenen I, Roux SL, Lessells R, Mahomed H, Maughan D, Meintjes G, Mendelson M, Mnguni A, Moodley M, Murie K, Naude J, Ntusi NAB, Paleker M, Parker A, Pienaar D, Preiser W, Prozesky H, Raubenheimer P, Rossouw L, Schrueder N, Smith B, Smith M, Solomon W, Symons G, Taljaard J, Wasserman S, Wilkinson RJ, Wolmarans M, Wolter N, Boulle A. Outcomes of laboratory-confirmed SARS-CoV-2 infection during resurgence driven by Omicron lineages BA.4 and BA.5 compared with previous waves in the Western Cape Province, South Africa. Int J Infect Dis 2022; 127: 63–68. 2. 2.Johns Hopkins University Center for Systems Science and Engineering. 2022. Available at: [https://coronavirus.jhu.edu/map.html](https://coronavirus.jhu.edu/map.html). 3. 3.Cai J, Deng X, Yang J, Sun K, Liu H, Chen Z, Peng C, Chen X, Wu Q, Zou J, Sun R, Zheng W, Zhao Z, Lu W, Liang Y, Zhou X, Ajelli M, Yu H. Modeling transmission of SARS-CoV-2 Omicron in China. Nat Med 2022; 28: 1468–1475. 4. 4.Chen X, Yan X, Sun K, Zheng N, Sun R, Zhou J, Deng X, Zhuang T, Cai J, Zhang J, Ajelli M, Yu H. Estimation of disease burden and clinical severity of COVID-19 caused by Omicron BA.2 in Shanghai, February-June 2022. Emerg Microbes Infect 2022; 11: 2800–2807. 5. 5.Colnago M, Benvenuto GA, Casaca W, Negri RG, Fernandes EG, Cuminato JA. Risk factors associated with mortality in hospitalized patients with COVID-19 during the Omicron wave in Brazil. Bioeng 2022; 9. 6. 6.Gautret P, Hoang VT, Jimeno MT, Lagier JC, Rossi P, Fournier PE, Colson P, Raoult D. The severity of the first 207 infections with the SARS-CoV-2 Omicron BA.2 variant, in Marseille, France, December 2021-February 2022. J Med Virol 2022; 94: 3494–3497. 7. 7.Jassat W, Mudara C, Ozougwu L, Tempia S, Blumberg L, Davies MA, Pillay Y, Carter T, Morewane R, Wolmarans M, von Gottberg A, Bhiman JN, Walaza S, Cohen C, group Da. Difference in mortality among individuals admitted to hospital with COVID-19 during the first and second waves in South Africa: a cohort study. Lancet Glob Health 2021; 9: e1216–e1225. 8. 8.Maruotti A, Ciccozzi M, Jona-Lasinio G. COVID-19-induced excess mortality in Italy during the Omicron wave. IJID Reg 2022; 4: 85–87. 9. 9.Maslo C MA, Laubscher A, Toubkin M, Sitharam L, Feldman C, Richards GA. COVID-19: A comparative study of severity of patients hospitalized during the first and the second wave in South Africa. MedRXiv 2021. 10. 10.Ward IL, Bermingham C, Ayoubkhani D, Gethings OJ, Pouwels KB, Yates T, Khunti K, Hippisley-Cox J, Banerjee A, Walker AS, Nafilyan V. Risk of covid-19 related deaths for SARS-CoV-2 Omicron (B.1.1.529) compared with Delta (B.1.617.2): retrospective cohort study. BMJ 2022; 378: e070695. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiYm1qIjtzOjU6InJlc2lkIjtzOjE5OiIzNzgvYXVnMDJfNy9lMDcwNjk1IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjMvMDgvMjMvMjAyMy4wMy4wNi4yMzI4NjgzNC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 11. 11.Fontana LM, Villamagna AH, Sikka MK, McGregor JC. Understanding viral shedding of severe acute respiratory coronavirus virus 2 (SARS-CoV-2): Review of current literature. Infect Control Hosp Epidemiol 2021; 42: 659–668. 12. 12.Ramos-Rincon JM, Herrera-Garcia C, Silva-Ortega S, Portilla-Tamarit J, Alenda C, Jaime-Sanchez FA, Arenas-Jimenez J, Fornes-Riera FE, Scholz A, Escribano I, Pedrero-Castillo V, Munoz-Miguelsanz C, Orts-Llinares P, Marti-Pastor A, Amo-Lozano A, Garcia-Sevila R, Ribes-Mengual I, Moreno-Perez O, Concepcion-Aramendia L, Merino E, Sanchez-Martinez R, Aranda I. Pathological findings associated with SARS-CoV-2 on postmortem core biopsies: correlation with clinical presentation and disease course. Front Med 2022; 9: 874307. 13. 13.Stein SR, Ramelli SC, Grazioli A, Chung JY, Singh M, Yinda CK, Winkler CW, Sun J, Dickey JM, Ylaya K, Ko SH, Platt AP, Burbelo PD, Quezado M, Pittaluga S, Purcell M, Munster VJ, Belinky F, Ramos-Benitez MJ, Boritz EA, Lach IA, Herr DL, Rabin J, Saharia KK, Madathil RJ, Tabatabai A, Soherwardi S, McCurdy MT, Consortium NC-A, Peterson KE, Cohen JI, de Wit E, Vannella KM, Hewitt SM, Kleiner DE, Chertow DS. SARS-CoV-2 infection and persistence in the human body and brain at autopsy. Nature 2022; 612: 758–763. 14. 14.Basile K, McPhie K, Carter I, Alderson S, Rahman H, Donovan L, Kumar S, Tran T, Ko D, Sivaruban T, Ngo C, Toi C, O’Sullivan MV, Sintchenko V, Chen SC, Maddocks S, Dwyer DE, Kok J. Cell-based culture informs infectivity and safe de-isolation assessments in patients with coronavirus disease 2019. Clin Infect Dis 2021; 73: e2952–e2959. 15. 15.Berengua C, Lopez M, Esteban M, Marin P, Ramos P, Cuerpo MD, Gich I, Navarro F, Miro E, Rabella N. Viral culture and immunofluorescence for the detection of SARS-CoV-2 infectivity in RT-PCR positive respiratory samples. J Clin Virol 2022; 152: 105167. 16. 16.Bullard J, Dust K, Funk D, Strong JE, Alexander D, Garnett L, Boodman C, Bello A, Hedley A, Schiffman Z, Doan K, Bastien N, Li Y, Van Caeseele PG, Poliquin G. Predicting infectious severe acute respiratory syndrome coronavirus 2 from diagnostic samples. Clin Infect Dis 2020; 71: 2663–2666. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/cid/ciaa638&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F23%2F2023.03.06.23286834.atom) 17. 17.Lan L, Xu D, Ye G, Xia C, Wang S, Li Y, Xu H. Positive RT-PCR test results in patients recovered from COVID-19. JAMA 2020; 323: 1502–1503. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F23%2F2023.03.06.23286834.atom) 18. 18.Ling Y, Xu SB, Lin YX, Tian D, Zhu ZQ, Dai FH, Wu F, Song ZG, Huang W, Chen J, Hu BJ, Wang S, Mao EQ, Zhu L, Zhang WH, Lu HZ. Persistence and clearance of viral RNA in 2019 novel coronavirus disease rehabilitation patients. Chin Med J 2020; 133: 1039–1043. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F23%2F2023.03.06.23286834.atom) 19. 19. Santos Bravo M, Berengua C, Marin P, Esteban M, Rodriguez C, Del Cuerpo M, Miro E, Cuesta G, Mosquera M, Sanchez-Palomino S, Vila J, Rabella N, Marcos MA. Viral culture confirmed SARS-CoV-2 subgenomic RNA value as a good surrogate marker of infectivity. J Clin Microbiol 2022; 60: e0160921. 20. 20.Singanayagam A, Patel M, Charlett A, Lopez Bernal J, Saliba V, Ellis J, Ladhani S, Zambon M, Gopal R. Duration of infectiousness and correlation with RT-PCR cycle threshold values in cases of COVID-19, England, January to May 2020. Euro Surveill 2020; 25. 21. 21.van Kampen JJA, van de Vijver D, Fraaij PLA, Haagmans BL, Lamers MM, Okba N, van den Akker JPC, Endeman H, Gommers D, Cornelissen JJ, Hoek RAS, van der Eerden MM, Hesselink DA, Metselaar HJ, Verbon A, de Steenwinkel JEM, Aron GI, van Gorp ECM, van Boheemen S, Voermans JC, Boucher CAB, Molenkamp R, Koopmans MPG, Geurtsvankessel C, van der Eijk AA. Duration and key determinants of infectious virus shedding in hospitalized patients with coronavirus disease-2019 (COVID-19). Nat Commun 2021; 12: 267. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41467-020-20568-4&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=33431879&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F23%2F2023.03.06.23286834.atom) 22. 22.Wolfel R, Corman VM, Guggemos W, Seilmaier M, Zange S, Muller MA, Niemeyer D, Jones TC, Vollmar P, Rothe C, Hoelscher M, Bleicker T, Brunink S, Schneider J, Ehmann R, Zwirglmaier K, Drosten C, Wendtner C. Virological assessment of hospitalized patients with COVID-2019. Nature 2020; 581: 465–469. [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%2F2023%2F08%2F23%2F2023.03.06.23286834.atom) 23. 23.Young BE, Ong SWX, Ng LFP, Anderson DE, Chia WN, Chia PY, Ang LW, Mak TM, Kalimuddin S, Chai LYA, Pada S, Tan SY, Sun L, Parthasarathy P, Fong SW, Chan YH, Tan CW, Lee B, Rotzschke O, Ding Y, Tambyah P, Low JGH, Cui L, Barkham T, Lin RTP, Leo YS, Renia L, Wang LF, Lye DC, Singapore Novel Coronavirus Outbreak Research T. Viral dynamics and immune correlates of coronavirus disease 2019 (COVID-19) severity. Clin Infect Dis 2021; 73: e2932–e2942. 24. 24.Killingley B, Mann AJ, Kalinova M, Boyers A, Goonawardane N, Zhou J, Lindsell K, Hare SS, Brown J, Frise R, Smith E, Hopkins C, Noulin N, Londt B, Wilkinson T, Harden S, McShane H, Baillet M, Gilbert A, Jacobs M, Charman C, Mande P, Nguyen-Van-Tam JS, Semple MG, Read RC, Ferguson NM, Openshaw PJ, Rapeport G, Barclay WS, Catchpole AP, Chiu C. Safety, tolerability and viral kinetics during SARS-CoV-2 human challenge in young adults. Nat Med 2022; 28: 1031–1041. 25. 25.Suvarna SK, Layton C, Bancroft JD. Banfords theory and practice of histology techniques (8th edition. Elsevier 2019. 26. 26.Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, Batut P, Chaisson M, Gingeras TR. STAR): ultrafas;t universal RNA-seq aligner. Bioinformatics 2013; 29: 15–21. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/bioinformatics/bts635&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23104886&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F23%2F2023.03.06.23286834.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000312654600003&link_type=ISI) 27. 27.Howe KL, Achuthan P, Allen J, Allen J, Alvarez-Jarreta J, Amode MR, Armean IM, Azov AG, Bennett R, Bhai J, Billis K, Boddu S, Charkhchi M, Cummins C, Da Rin Fioretto L, Davidson C, Dodiya K, El Houdaigui B, Fatima R, Gall A, Garcia Giron C, Grego T, Guijarro-Clarke C, Haggerty L, Hemrom A, Hourlier T, Izuogu OG, Juettemann T, Kaikala V, Kay M, Lavidas I, Le T, Lemos D, Gonzalez Martinez J, Marugan JC, Maurel T, McMahon AC, Mohanan S, Moore B, Muffato M, Oheh DN, Paraschas D, Parker A, Parton A, Prosovetskaia I, Sakthivel MP, Salam AIA, Schmitt BM, Schuilenburg H, Sheppard D, Steed E, Szpak M, Szuba M, Taylor K, Thormann A, Threadgold G, Walts B, Winterbottom A, Chakiachvili M, Chaubal A, De Silva N, Flint B, Frankish A, Hunt SE, GR II, Langridge N, Loveland JE, Martin FJ, Mudge JM, Morales J, Perry E, Ruffier M, Tate J, Thybert D, Trevanion SJ, Cunningham F, Yates AD, Zerbino DR, Flicek P. Ensembl 2021. Nucleic Acids Res 2021; 49: D884–D891. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/NAR/GKAA942&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F23%2F2023.03.06.23286834.atom) 28. 28.Wu T, Hu E, Xu S, Chen M, Guo P, Dai Z, Feng T, Zhou L, Tang W, Zhan L, Fu X, Liu S, Bo X, Yu G. clusterProfiler 4.0: A universal enrichment tool for interpreting omics data. Innov 2021; 2: 100141. 29. 29.Ragab D, Salah Eldin H, Taeimah M, Khattab R, Salem R. The COVID-19 cytokine storm; what we know so far. Front Immunol 2020; 11: 1446. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3389/fimmu.2020.01446&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F23%2F2023.03.06.23286834.atom) 30. 30.Merad M, Blish CA, Sallusto F, Iwasaki A. The immunology and immunopathology of COVID-19. Science 2022; 375: 1122–1127. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1126/science.abm8108&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F23%2F2023.03.06.23286834.atom) 31. 31.Bhimraj A MR, Shumaker AH, Baden L, Cheng VC, Edwards KM, Gallagher JC, Gandhi RT, Muller WJ, Nakamura MM, O’Horo JC, Shafer RW, Shoham S, Murad MH, Mustafa RA, Sultan S, Falck-Ytte Y. IDSA Guidelines on the Treatment and Management of Patients with COVID-19. 2022. Available at: [https://www.idsociety.org/practice-guideline/covid-19-guideline-treatment-and-management/#Recommendations15-17:Remdesivir](https://www.idsociety.org/practice-guideline/covid-19-guideline-treatment-and-management/#Recommendations15-17:Remdesivir). 32. 32.Folgueira MD, Luczkowiak J, Lasala F, Perez-Rivilla A, Delgado R. Prolonged SARS-CoV-2 cell culture replication in respiratory samples from patients with severe COVID-19. Clin Microbiol Infect 2021; 27: 886–891. 33. 33.Pechous RD, Malaviarachchi PA, Banerjee SK, Byrum SD, Alkam DH, Ghaffarieh A, Kurten RC, Kennedy JL, Xuming Z. An ex vivo human precision-cut lung slice platform provides insight into SARS2 CoV-2 pathogenesis and antiviral drug efficacy. bioRxiv 2023. 34. 34.Xu Q, Milanez-Almeida P, Martins AJ, Radtke AJ, Hoehn KB, Oguz C, Chen J, Liu C, Tang J, Grubbs G, Stein S, Ramelli S, Kabat J, Behzadpour H, Karkanitsa M, Spathies J, Kalish H, Kardava L, Kirby M, Cheung F, Preite S, Duncker PC, Kitakule MM, Romero N, Preciado D, Gitman L, Koroleva G, Smith G, Shaffer A, McBain IT, McGuire PJ, Pittaluga S, Germain RN, Apps R, Schwartz DM, Sadtler K, Moir S, Chertow DS, Kleinstein SH, Khurana S, Tsang JS, Mudd P, Schwartzberg PL, Manthiram K. Adaptive immune responses to SARS-CoV-2 persist in the pharyngeal lymphoid tissue of children. Nat Immunol 2023; 24: 186–199. 35. 35.Zollner A, Koch R, Jukic A, Pfister A, Meyer M, Rossler A, Kimpel J, Adolph TE, Tilg H. Postacute COVID-19 is characterized by gut viral antigen persistence in inflammatory bowel diseases. Gastroenterology 2022; 163: 495–506 e498. 36. 36.Choi B, Choudhary MC, Regan J, Sparks JA, Padera RF, Qiu X, Solomon IH, Kuo HH, Boucau J, Bowman K, Adhikari UD, Winkler ML, Mueller AA, Hsu TY, Desjardins M, Baden LR, Chan BT, Walker BD, Lichterfeld M, Brigl M, Kwon DS, Kanjilal S, Richardson ET, Jonsson AH, Alter G, Barczak AK, Hanage WP, Yu XG, Gaiha GD, Seaman MS, Cernadas M, Li JZ. Persistence and evolution of SARS-CoV-2 in an immunocompromised host. NEJM 2020; 383: 2291–2293. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMc2031364&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F23%2F2023.03.06.23286834.atom) 37. 37.Griffin I, Woodworth KR, Galang RR, Burkel VK, Neelam V, Siebman S, Barton J, Manning SE, Aveni K, Longcore ND, Harvey EM, Ngo V, Mbotha D, Chicchelly S, Lush M, Eckert V, Dzimira P, Sokale A, Valencia-Prado M, Azziz-Baumgartner E, MacNeil A, Gilboa SM, Tong VT. Recurrent SARS-CoV-2 RNA detection after COVID-19 illness onset during pregnancy. Emerg Infect Dis 2022; 28: 873–876. 38. 38.Rodriguez-Grande C, Alcala L, Estevez A, Sola-Campoy PJ, Buenestado-Serrano S, Martinez-Laperche C, Manuel de la Cueva V, Alonso R, Andres-Zayas C, Adan-Jimenez J, Losada C, Rico-Luna C, Comas I, Gonzalez-Candelas F, Catalan P, Munoz P, Perez-Lago L, Garcia de Viedma D, Gregorio Maranon Microbiology IDCSG. Systematic genomic and clinical analysis of severe acute respiratory syndrome coronavirus 2 reinfections and recurrences involving the same strain. Emerg Infect Dis 2022; 28: 85-94. 39. 39.Alexandersen S, Chamings A, Bhatta TR. SARS-CoV-2 genomic and subgenomic RNAs in diagnostic samples are not an indicator of active replication. Nat Commun 2020; 11: 6059. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41467-020-19883-7&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F23%2F2023.03.06.23286834.atom) 40. 40.Hwang HS, Lo CM, Murphy M, Grudda T, Gallagher N, Luo CH, Robinson ML, Mirza A, Conte M, Conte A, Zhou R, Vergara C, Brooke CB, Pekosz A, Mostafa HH, Manabe YC, Thio CL, Balagopal A. Characterizing SARS-CoV-2 Transcription of Subgenomic and Genomic RNAs During Early Human Infection Using Multiplexed Droplet Digital Polymerase Chain Reaction. J Infect Dis 2023; 227: 981–992. 41. 41.Consortium WHOST. Remdesivir and three other drugs for hospitalised patients with COVID-19: final results of the WHO Solidarity randomised trial and updated meta-analyses. Lancet 2022; 399: 1941–1953. 42. 42.Beigel JH, Tomashek KM, Dodd LE, Mehta AK, Zingman BS, Kalil AC, Hohmann E, Chu HY, Luetkemeyer A, Kline S, Lopez de Castilla D, Finberg RW, Dierberg K, Tapson V, Hsieh L, Patterson TF, Paredes R, Sweeney DA, Short WR, Touloumi G, Lye DC, Ohmagari N, Oh MD, Ruiz-Palacios GM, Benfield T, Fatkenheuer G, Kortepeter MG, Atmar RL, Creech CB, Lundgren J, Babiker AG, Pett S, Neaton JD, Burgess TH, Bonnett T, Green M, Makowski M, Osinusi A, Nayak S, Lane HC, Members A-SG. Remdesivir for the treatment of Covid-19 - final report. NEJM 2020; 383: 1813–1826. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa2007764&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F23%2F2023.03.06.23286834.atom) 43. 43.Lapadula G, Bernasconi DP, Bellani G, Soria A, Rona R, Bombino M, Avalli L, Rondelli E, Cortinovis B, Colombo E, Valsecchi MG, Migliorino GM, Bonfanti P, Foti G, Remdesivir-Ria Study G. Remdesivir use in patients requiring mechanical ventilation due to COVID-19. Open Forum Infect Dis 2020; 7: ofaa481. 44. 44.Pasquini Z, Montalti R, Temperoni C, Canovari B, Mancini M, Tempesta M, Pimpini D, Zallocco N, Barchiesi F. Effectiveness of remdesivir in patients with COVID-19 under mechanical ventilation in an Italian ICU. J Antimicrob Chemother 2020; 75: 3359–3365. 45. 45.Budhraja A, Basu A, Gheware A, Abhilash D, Rajagopala S, Pakala S, Sumit M, Ray A, Subramaniam A, Mathur P, Nambirajan A, Kumar S, Gupta R, Wig N, Trikha A, Guleria R, Sarkar C, Gupta I, Jain D. Molecular signature of postmortem lung tissue from COVID-19 patients suggests distinct trajectories driving mortality. Dis Model Mech 2022; 15. 46. 46.Melms JC, Biermann J, Huang H, Wang Y, Nair A, Tagore S, Katsyv I, Rendeiro AF, Amin AD, Schapiro D, Frangieh CJ, Luoma AM, Filliol A, Fang Y, Ravichandran H, Clausi MG, Alba GA, Rogava M, Chen SW, Ho P, Montoro DT, Kornberg AE, Han AS, Bakhoum MF, Anandasabapathy N, Suarez-Farinas M, Bakhoum SF, Bram Y, Borczuk A, Guo XV, Lefkowitch JH, Marboe C, Lagana SM, Del Portillo A, Tsai EJ, Zorn E, Markowitz GS, Schwabe RF, Schwartz RE, Elemento O, Saqi A, Hibshoosh H, Que J, Izar B. A molecular single-cell lung atlas of lethal COVID-19. Nature 2021; 595: 114–119. 47. 47.Tay MZ, Poh CM, Renia L, MacAry PA, Ng LFP. The trinity of COVID-19: immunity, inflammation and intervention. Nat Rev Immunol2020; 20: 363–374. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41577-020-0311-8&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F23%2F2023.03.06.23286834.atom) 48. 48.van de Veerdonk FL, Giamarellos-Bourboulis E, Pickkers P, Derde L, Leavis H, van Crevel R, Engel JJ, Wiersinga WJ, Vlaar APJ, Shankar-Hari M, van der Poll T, Bonten M, Angus DC, van der Meer JWM, Netea MG. A guide to immunotherapy for COVID-19. Nat Med 2022; 28: 39–50. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41591-021-01643-9&link_type=DOI) 49. 49.Wang EY, Mao T, Klein J, Dai Y, Huck JD, Jaycox JR, Liu F, Zhou T, Israelow B, Wong P, Coppi A, Lucas C, Silva J, Oh JE, Song E, Perotti ES, Zheng NS, Fischer S, Campbell M, Fournier JB, Wyllie AL, Vogels CBF, Ott IM, Kalinich CC, Petrone ME, Watkins AE, Yale IT, Dela Cruz C, Farhadian SF, Schulz WL, Ma S, Grubaugh ND, Ko AI, Iwasaki A, Ring AM. Diverse functional autoantibodies in patients with COVID-19. Nature 2021; 595: 283–288. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41586-021-03631-y&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F23%2F2023.03.06.23286834.atom) 50. 50.Wang S, Yao X, Ma S, Ping Y, Fan Y, Sun S, He Z, Shi Y, Sun L, Xiao S, Song M, Cai J, Li J, Tang R, Zhao L, Wang C, Wang Q, Zhao L, Hu H, Liu X, Sun G, Chen L, Pan G, Chen H, Li Q, Zhang P, Xu Y, Feng H, Zhao GG, Wen T, Yang Y, Huang X, Li W, Liu Z, Wang H, Wu H, Hu B, Ren Y, Zhou Q, Qu J, Zhang W, Liu GH, Bian XW. A single-cell transcriptomic landscape of the lungs of patients with COVID-19. Nat Cell Biol 2021; 23: 1314–1328. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41556-021-00796-6&link_type=DOI) 51. 51.Deinhardt-Emmer S, Wittschieber D, Sanft J, Kleemann S, Elschner S, Haupt KF, Vau V, Haring C, Rodel J, Henke A, Ehrhardt C, Bauer M, Philipp M, Gassler N, Nietzsche S, Loffler B, Mall G. Early postmortem mapping of SARS-CoV-2 RNA in patients with COVID-19 and the correlation with tissue damage. Elife 2021; 10.