Saliva molecular testing bypassing RNA extraction is suitable for monitoring and diagnosing SARS-CoV-2 infection in children ============================================================================================================================ * Marta Alenquer * Tiago Milheiro Silva * Onome Akpogheneta * Filipe Ferreira * Sílvia Vale-Costa * Mónica Medina-Lopes * Frederico Batista * Ana Margarida Garcia * Vasco M. Barreto * Cathy Paulino * João Costa * João Sobral * Maria Diniz-da-Costa * Susana Ladeiro * José Delgado Alves * Ricardo B. Leite * Jocelyne Demengeot * Maria João Rocha Brito * Maria João Amorim ## Structured abstract **IMPORTANCE** Adults are being vaccinated against SARS-CoV-2 worldwide, but the longitudinal protection of these vaccines is uncertain, given the ongoing appearance of SARS-CoV-2 variants. Children are susceptible to infection, and some studies reported that they actively transmit the virus even when asymptomatic, thus affecting the community. Methods to easily test infected children and track the virus they carry are in demand. **OBJECTIVE** To determine if saliva is an effective sample for detecting SARS-CoV-2 RNA and antibodies in children aged 10 years and under, and associate viral RNA levels to infectivity. **DESIGN, SETTING, AND PARTICIPANTS** In this cross-sectional study, saliva SARS-CoV-2 RT-qPCR tests, with and without RNA extraction, were validated in 49 hospitalized adults. The test was then applied to 85 children, aged 10 years and under, admitted to the hospital regardless of COVID-19 symptomatology. Amongst 85 children, 29 (63.0%) presented at least one COVID-19 symptom, 46 (54.1%) were positive for SARS-CoV-2 infection, 28 (32.9%) were under the age of 1 and the mean (SD) age was 3.8 (3.4) years. Saliva samples were collected up to 48 h after a positive test by nasopharyngeal (NP) swab-RT-qPCR. **EXPOSURE** Infection by SARS-COV-2 in adults up to 8 days post-symptom onset. Children admitted to hospital for any reason and therefore with unclear onset of SARS-CoV-2 infection. **MAIN OUTCOMES AND MEASURES** Saliva RT-qPCR up to CT<37 accurately identifies SARS-CoV-2 infected children, with viral infectivity in tissue culture restricted to CT<26. **RESULTS** In adults, the accuracy of the saliva SARS-CoV-2 RT-qPCR test was 98.0% (95% confidence intervals [CI]: 89.3%–100%) as compared to NP-RT-qPCR. In children, the sensitivity, specificity, and accuracy of saliva-RT-qPCR tests compared to NP swab-RT-qPCR were, respectively, 84.8% (71.8%–92.4%), 100% (91.0%–100%), and 91.8% (84.0%– 96.6%) with RNA extraction and 81.8% (68.0%–90.5%), 100% (91.0%–100%), and 90.4% (82.1%–95.0%) without RNA extraction. The threshold for rescuing infectious particles from saliva was CT<26. There were significant IgM positive responses to the spike protein and its receptor-binding domain (RBD) among children positive for SARS-CoV-2 by NP swab and negative by saliva compared to other groups, indicating late infection onset (>7–10 days). **CONCLUSIONS AND RELEVANCE** Saliva-molecular testing is suitable in children aged 10 years and under, including infants aged <1 year, even bypassing RNA extraction methods. Importantly, the detected viral RNA levels were significantly above the infectivity threshold in several samples. Further investigation is required to understand how SARS-CoV-2 RNA levels correlate with viral transmission. **Question** Is saliva reverse transcription-quantitative polymerase chain reaction (RT-qPCR) testing (with and without RNA extraction) suitable to identify SARS-CoV-2 infected young children and can the cycle threshold (CT) be associated with infectivity in a heterogeneous population admitted to hospital for COVID-19-related and unrelated reasonsã **Findings** In this cross-sectional study of 85 children aged 10 years and under, RT-qPCR in saliva samples subjected or not to RNA extraction accurately detected SARS-CoV-2 RNA and infectious viruses could be recovered from CTs below 26. **Meaning** Saliva sampling coupled to RT-qPCR and specific antibody detection efficiently identifies infants and children infected with SARS-CoV-2. This approach is suitable for surveillance in kindergarten and school settings. ## Introduction The COVID-19 pandemic caused by SARS-CoV-2 has resulted in excess morbidity and mortality worldwide, especially in elderly populations and people with associated specific comorbidities.1-3 Adults and children above 12 years of age are being vaccinated at different paces worldwide,4 but younger children will remain more susceptible to infection. How new variants will be transmitted amongst children and affect the community remains unclear. In fact, thus far, the role of children in SARS-CoV-2 transmission remains poorly understood, mostly because the majority of children with SARS-CoV-2 display mild to no symptoms;5 it has been estimated that 93% of infected children are not identified by symptom screening.6 However, it is now well established that children are susceptible to infection,7-9 and a small percentage may develop serious complications,10 including pneumonia, myocarditis, central nervous system disorders, and multisystem inflammatory syndrome.11,12 It is critical in the next phase of the pandemic, to have readily available strategies for minimally-invasive approaches to monitor school settings; these strategies could help establish whether children are prone to evolve new variants and how variants impact viral transmission by children to the community by combining diagnosis with genotyping and epidemiological analyses. Saliva molecular testing has emerged as a suitable alternative to nasopharyngeal (NP) swabs for sampling and identification of SARS-CoV-2 RNA in children and adults, and for genotyping SARS-CoV-2 variants.13-15 Compared to NP swabs, saliva testing is less invasive and may be implemented in self-collected or parent-assisted contexts more easily, including the sampling of children up to 1-year-old, by low-pressure aspiration. Saliva RT-qPCR testing is also more sensitive than antigen testing.16 However, there is still some resistance to using saliva molecular testing partly because it is not a fast test, and it is unclear if the detected viral loads are substantially lower than those detected in fast lateral-flow tests using NP swab samples. In this cross-sectional study, we focused on children aged 10 years and under, admitted to hospital with COVID-19 related symptoms or with unrelated medical pathologies or surgeries, and investigated the potential of saliva for being coupled to RT-PCR testing, with saliva collected up to 48 h from a positive (or control) NP swab. Interestingly, we found non-significant differences between methods using and bypassing RNA extraction. In addition, we associated RNA levels detected in saliva with infectivity and quantified specific SARS-CoV-2 spike and receptor-binding-domain (RBD) antibodies in this type of sample. Interestingly, we only found statistically significant differences in IgM levels in samples positive in swab-molecular testing that were negative by saliva-molecular testing, suggesting that discrepancies between saliva and NP molecular tests are more frequent in children infected for more than 7-10 days, which correlates to loss of infectivity and hence transmission. Our study shows that saliva molecular testing bypassing RNA extraction is an efficient method for monitoring SARS-CoV-2 infected children in age groups up to 10 years-old. ## Methods ### Study design A total of 49 adults and 85 children (aged 10 years and under) inpatients were enrolled in this study between 25 August 2020 and 20 June 2021. NP swab samples were collected and processed at the hospital. Saliva samples were collected from adults or children, within 24 or 48 h from NP swab collection, respectively. Both SARS-CoV-2 positive and negative individuals (by NP swab) were enrolled in this study. In adults, only symptomatic patients were enrolled, whereas, for children, patients were enrolled after admission to the hospital for COVID-19 symptoms or causes non-related to COVID-19 (other medical pathologies or surgeries). This study was approved by the Ethics committees of Centro Hospitalar Universitário de Lisboa Central and Hospital Professor Doutor Fernando Fonseca, in compliance with the Declaration of Helsinki, and follows international and national guidelines for health data protection. All participants, or their guardians, provided informed written consent to take part in the study. ### Saliva collection At least 1 mL of saliva was collected with the help of a health care worker, after abstinence from food or water for at least 30 min. Participants were asked to pool saliva in the mouth and gently spit it into a sterile container without coughing or clearing their throats. For children under the age of 1 year, saliva was gently aspirated from the mouth with a suction tube. Samples were stored at 4ºC, sent to Instituto Gulbenkian de Ciência, and processed within 72 h from collection. ### SARS-CoV-2 detection Saliva was treated with Proteinase K (645 μg/mL in 160 nM SDS) for 30 min at 50ºC, followed by heat inactivation for 10 min at 98ºC. Saliva specimens with high viscosity were diluted 1:2 in TBE 2x prior to Proteinase K treatment.17 Samples were then used directly in the RT-qPCR reaction or after RNA extraction with the QIAamp Viral RNA Mini kit (Qiagen, 52906), according to the manufacturer’s instructions; 200 μL saliva were extracted and eluted in 50 μL RNase-free water; 1 μL extracted RNA or unextracted saliva were used for RT-qPCR. A one-step assay (cDNA synthesis and amplification) was performed using iTaq Universal Probes One-Step Kit (BioRad, #12013250). A master mix was prepared for each set of primer-probe CDC\_N1, CDC\_N2 and Hs\_RPP30 using: 0.5 nM of each primer pair, 125 nM of probe, 1x iTaq Universal Probes reaction mix, 2.5% (V/V) iScript Reverse Transcriptase and 10% (V/V) RNA or unextracted saliva sample. Two positive controls were performed separately per experiment (SARS-CoV-2 and Human) for N1, N2, and RP primer-probe set using 2,000 synthetic copies of nucleocapsid region of the virus or 2,000 synthetic copies of the Human single copy RPP30 gene. One negative control was performed without template for the same three conditions (N1, N2 and RP). All the primers and probes (2019-nCoV RUO Kit, 500 rxn, #10006713) were purchased from Integrated DNA Technologies, as well as the positive virus detection control (2019-nCoV_N_Positive Control, #10006625) and positive human sample control (Hs_RPP30 Positive Control, #10006626). Reactions were performed in 384 wells plates (ThermoFisher, #TF-0384) in a QuantStudio 7 Flex system (Applied Biosciences), using the fast mode, consisting of a hold stage at 50°C for 10 min, and 95°C for 3 min, followed by 45 cycles of a PCR stage at 95°C for 10 sec then 60°C for 30 sec (FAM signal acquisition step). Positive cases were considered when the two probes were amplified with a CT below 37. Negative detection was established as having no amplification or amplification of one probe above 37. Inconclusive results were considered, with only one probe being amplified with a CT less than 37. The limit of detection (LOD) of the saliva assay was performed by serial dilution of IDT synthetic copies (20,000 to 20) of SARS-CoV-2 in fresh saliva samples (non-positives) (eFigure 1). ### Infectious SARS-CoV-2 growth from saliva Saliva samples were diluted 1:4 in virus growth medium (Dulbecco’s Modified Eagle Medium [DMEM, Gibco, #21969035] supplemented with 2% fetal bovine serum [FBS, Gibco, #10500064], 2 mM L-glutamine [ThermoFisher, #25030024], 1% penicillin/streptomycin solution [Biowest, #L0022] and 2.5 μg/mL Amphotericin B [Gibco, #15290018]), vortexed, briefly centrifuged to collect debris and passed through a 40 μm filter. Two hundred μL of each sample were added in triplicate to Vero E6 cells (a kind gift from Rupert Beale, The Francis Crick Institute, UK) pre-seeded in 24 well plates with coverslips and centrifuged for 15 min at 3,500 g, 37ºC. After centrifugation, the inoculum was removed and 250 μL of fresh virus growth media were added to the cells (protocol adapted from 18). As a positive control, cells were infected with SARS-CoV-2 viral stock (strain human/DEU/HH-1/2020 from the European Virus Archive Global) at a multiplicity of infection (MOI) of 0.01. Cells were inspected daily for cytopathic effect (CPE) and considered negative if no CPE was visible for 7 days. At 1- and 4-days post-infection, a replicate was fixed with 4% formaldehyde and processed for immunofluorescence, as described in 19. Cells were stained with a rabbit polyclonal SARS-CoV-2 nucleocapsid antibody (1:1,000; ThermoFisher, #MA536270) and an anti-rabbit IgG Alexa-Fluor 488 secondary antibody (1:1,000; Invitrogen, #A-21206). Cell nuclei were stained with Hoechst 33342 (1 μg/mL, Invitrogen, #H3570). Single optical sections were imaged with a Leica SP5 confocal microscope. ### ELISA The ELISA assay used to quantify saliva IgG, IgA, and IgM anti-full-length SARS-CoV-2 spike and its receptor-binding domain (RBD) was adapted from 20 as described in 21,22, with few modifications described here. Briefly, high binding 96□well plates (Maxisorb) were coated with either RBD or spike as capture antigen at 0.5□µg/mL stored overnight at 4ºC. Plates were blocked with PBS supplemented with 2% BSA (PBS-BSA) for 1–4 hours at room temperature. Saliva samples were tested at 1 in 25 dilutions in PBS with 2% BSA and 0.1% Tween (PBS-BSA-T), in duplicate. For each isotype test plate, an IgG, IgM (GenScript [clone 2001]) or IgA (Absolute Antibody [clone 3022]) SARS-CoV-2 reactive monoclonal antibody was used to generate a concentration curve upon serial dilution and validate each plate assayed. Duplicate measurements of reference positive and negative sera samples were used at 1 in 50 dilutions in PBS-BSA-T to validate each plate assayed; negative sera were used to determine cutoff values. Samples and monoclonal antibodies were incubated for 2 hours at 37ºC, then plates were washed 3x in PBS-T. Secondary antibody goat anti□Human Fc□HRP IgG, IgA or IgM (Abcam) diluted 1 in 25,000 in PBS-BSA-T was added to respective isotype plates and incubated for 1 hour at 37ºC to reveal bound IgG, IgA or IgM. Plates were then washed 3x in PBS-T and incubated with 3,3′,5,5′□tetramethylbenzidine (BD OptEIA™, BD Biosciences) for 20 to 30 minutes at 37ºC. The reaction was stopped with sulphuric acid, and the colorimetric assay was read to provide optical density (OD) at 450□nm. Reference negative sera were collected at least 3 years before the COVID□19 pandemic and used as pooled serum from 50 samples. Isotype-specific positive and negative thresholds were determined for each assay plate from the mean negative serum value plus two standard deviations (SD). Negative sera were obtained upon informed consent in the frame of the projects “Genetic susceptibility factors and immunologic protection in COVID□19” and “Genetic variance in Portuguese population: candidate genes in COVID□19”, both approved by the IGC Ethics Committee (reference H004.2020 and H002.2020, respectively). ### Statistical analysis The values of sensitivity, specificity, and accuracy were estimated using the results of the NP swab as the reference standard. 95% confidence intervals (CI) were calculated using the Wilson method, recommended for small sample sizes.23 The analytical sensitivity of SARS-CoV-2 RNA detection between saliva and nasopharyngeal swabs from SARS-CoV-2 positive adults and children was compared using a Wilcoxon matched-pairs signed-rank test, in GraphPad Prism 9.1.2. ELISA data were cleaned, and categorical variables were created where needed. Thresholds for antibody-positive responses were defined by the negative pool (mean + 2 SD). The correlation between antibody OD values and age was assessed using Pearson’s Pairwise coefficients. Descriptive statistics were assessed as frequencies and percentages (n [%]) for categorical variables; crude associations were tested with Pearson’s Chi2 tests. Significance level was set at <0.05. STATA (StataCorp LLC, USA, V16) was used for all analyses. ## Results ### Method validation in adults We first validated our method with adult patients admitted to Hospital Fernando Fonseca with COVID-19, or without SARS-CoV-2 infection, as negative controls. Once SARS-CoV-2 infection was assessed by RT-qPCR on RNA extracted from a nasopharyngeal (NP) swab, a saliva sample was collected within the following 48 hours. Saliva specimens were pre-treated with proteinase K, RNA was extracted and subjected to RT-qPCR using the primer and probe sequences from the Center for Disease Control and Prevention.24 Alternatively, saliva samples treated with proteinase K were used directly for the RT-qPCR reaction, without RNA extraction. The overall concordances of saliva and NP swab were 98.0% (48/49) and 97.8% (46/47), with and without RNA extraction from saliva, respectively (Table 1). Of the patients with a positive NP swab, 100% were also positive in saliva, either with (36/36) or without (34/34) RNA extraction. The differences in CT values between saliva (with or without RNA extraction) and NP swab were not statistically significant (Figure 1A), but CT values in saliva with vs. without RNA extraction are statistically different, with extraction of RNA consistently decreasing the CT values obtained. ![Figure 1.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2021/08/12/2021.08.11.21261899/F1.medium.gif) [Figure 1.](http://medrxiv.org/content/early/2021/08/12/2021.08.11.21261899/F1) Figure 1. SARS-CoV-2 RNA detection in NP swab and saliva samples from adult patients infected with SARS-CoV-2. (A) Comparison of CT values from paired saliva (with and without RNA extraction) and NP swab specimens. Each line corresponds to a paired specimen. n.d., not detected. ns, not significant; \***|p<0.001, by Wilcoxon matched-pairs signed-rank test. (B) Saliva stability at 4ºC and -20ºC: comparison of CT values from paired saliva samples (with and without RNA extraction) processed immediately, after 3 days at 4ºC or 7 days at -20ºC. View this table: [Table 1.](http://medrxiv.org/content/early/2021/08/12/2021.08.11.21261899/T1) Table 1. Performance of saliva in adults and children. Summary of results obtained from parallel testing of swab and saliva with and without extraction of RNA. Next, we analyzed the stability of saliva samples for stored 3 days at 4ºC or 7 days at -20ºC prior to processing (Figure 1B). We detected viral RNA in all saliva samples kept at 4ºC and -20ºC, regardless of RNA extraction, demonstrating they are stable under the conditions studied. ### Performance of the method in pediatric patients Eighty-five patients aged 10 years and under admitted to Hospital Dona Estefânia between August 2020 and June 2021 were included in our analysis. The clinical characteristics of the participants are shown in Table 2. Forty-six (54.1%) were positive for SARS-CoV-2 infection and, within these, 29 (63.0%) presented at least one COVID-19 symptom, the most common being fever (50.0%), cough (28.0%), and coryza (28.0%). The remaining 17 children (37.0%) were diagnosed on routine tests prior to hospital admission for causes non-related to COVID-19. Twenty-eight (32.9%) participants were under the age of 1 and the mean (SD) age was 3.8 (3.4) years. View this table: [Table 2.](http://medrxiv.org/content/early/2021/08/12/2021.08.11.21261899/T2) Table 2. Characteristics and reported signs and symptoms of SARS-CoV-2-positive and negative children included in this study. Upon hospital admission, SARS-CoV-2 infection was assessed by RT-qPCR quantification of RNA load in NP swab samples. Saliva samples were collected within the following 48 hours and processed as described above. The overall concordances of saliva and NP swab were 91.8% (78/85) with RNA extraction and 90.4% (75/83) without RNA extraction from saliva (Table 1). Of the children with a positive NP swab, 84.8% were positive in saliva after RNA extraction (39/46) and 81.1% (36/44) were positive in saliva without RNA extraction. The specificity of the method was 100%, with the 39 patients negative by NP swab, also negative in saliva, with and without RNA extraction. When restricting the analysis to children younger than 1-year-old, the sensitivity of the method increases to 87.0% (25/28) with RNA extraction and 86.4% (19/27) without RNA extraction from saliva (Table 1). The CT values in the saliva of children were significantly different from those in NP swab (Figure 2A), with a mean CT (SD) of 22.9 (6.2) in NP swab versus 26.1 (5.1) in saliva with RNA extraction and 27.9 (4.7) without extraction. It is important to notice that there was a time interval between NP swab and saliva collection that could go up to 48 h and, therefore, these differences may not reflect a lower sensitivity of our method but rather a decrease in the patient viral load. Importantly, there was no correlation between detection of SARS-CoV-2 in saliva and the CT value of the NP swab, or the age of the patient (Figure 2B). ![Figure 2.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2021/08/12/2021.08.11.21261899/F2.medium.gif) [Figure 2.](http://medrxiv.org/content/early/2021/08/12/2021.08.11.21261899/F2) Figure 2. Detection of SARS-CoV-2 infection in NP swab and saliva samples from children. (A) Comparison of CT values from paired saliva (with and without RNA extraction) and NP swab specimens from children aged 10 years and under, positive for SARS-CoV-2 infection. Each line corresponds to a paired specimen. n.d., not detected. Each line corresponds to a paired specimen. n.d., not detected; **p<0.01, \***|p<0.001, by Wilcoxon matched-pairs signed-rank test. (B) Graphical representation of CT values in NP swabs from infected children vs. age. Positive in saliva, green dots; negative in saliva, red dots. (C) Saliva stability at 4ºC and -20ºC: comparison of CT values from paired saliva samples (with and without RNA extraction) processed immediately, after 3 days at 4ºC or 7 days at -20ºC. (D, E) Infectious SARS-CoV-2 growth from saliva samples. Vero E6 cells were inoculated with saliva samples from children and inspected daily for the presence of a cytopathic effect. As a positive control, cells were infected with SARS-CoV-2 viral stock at an MOI of 0.01. (D) Graphical representation of CT values in saliva vs. NP swab result, with dots representing symptomatic children, triangles asymptomatic patients, green color indicating samples where viral replication was detected, and red color samples without viral growth. n.d., non-detected. (E) At 24h post-infection, cells were fixed with 4%formaldehyde, permeabilized with 0.2% Triton X100, and stained with SARS-CoV-2 Nucleocapsid antibody (green). Cell nuclei were stained with Hoechst (blue). White bar=10µm. Images were acquired with a Leica SP5 confocal microscope. Representative images from Vero cells infected with SARS-CoV-2 viral stock or inoculated with saliva specimens with CT values of 17, 22, 25, and 30, or saliva with non-detected SARS-CoV-2 (n.d.). (F) Levels of IgG, IgA, and IgM against full-length spike and spike’s RBD measured in the saliva of children aged 10 years and under, measured by ELISA. We also analyzed the stability of saliva samples from children when stored for 3 days at 4ºC or 7 days at -20ºC prior to processing (Figure 2C). As for adults, saliva samples from children were stable under the conditions tested, since viral RNA was detectable in all samples, irrespective of whether RNA extraction was performed. Finally, we tested saliva samples on a commercial lateral-flow test (COVID19 antigen rapid test, ALL TEST, #ICOV-502). Saliva samples that tested positive in our molecular assay with CT<26 also tested positive in the chosen lateral-flow assay (eFigure 2). Of note, two other commercial lateral-flow tests performed poorly in our hands. These data are consistent with what was observed for NP swabs16 and may have direct implications for monitoring schools. ### Viral growth from saliva samples To address if we could detect infectious SARS-CoV-2 viruses in children saliva and correlate viral replication with their CT value, saliva samples were cultured in Vero cells, inspected daily for CPE, and analyzed by immunofluorescence using an antibody against SARS-CoV-2 nucleoprotein (Figure 2D). Importantly, we recovered infectious viruses from all saliva samples with CT values ≤25.6, whereas in saliva samples with higher CT values or negative, no viral replication was detected. These results suggest that children with CT values in saliva equal or higher than 26 may not be able to shed infectious viruses. ### Antibodies against SARS-CoV-2 in saliva To address the levels of anti-SARS-CoV-2 IgG, IgA and IgM antibodies in the saliva of adults and children, we performed ELISA assays against full-length spike and spike’s RBD (Figure 2F and Table 3). In adults, all saliva samples (n=21) tested negative for IgG reactivities. In contrast, reactive IgA was detected in 16 (76.2%) adults, regardless of NP swab and saliva results, suggesting that the detection of IgA antibodies does not correlate with infection. This discrepancy could, however, be explained by the presence of secreted IgAs (sIgAs) that recognize glycosylation patterns existent in spike.25,26 Only one (4.8%) participant was positive for reactive IgM. View this table: [Table 3.](http://medrxiv.org/content/early/2021/08/12/2021.08.11.21261899/T3) Table 3. Patients with antibodies in saliva against full-length spike or RBD, divided by age category and by RT-qPCR result on swab and saliva. Of the 73 saliva samples from children tested by ELISA, 3 (3.1%) were positive for specific IgG. Reactive IgA and IgM were detected in 44 (60.3%) and 5 (11.1%) participants, respectively. Four participants with discrepant NP vs. saliva RT-qPCR data (NP positive, saliva negative, n=12) presented IgM, suggesting that the corresponding samples were collected after the first immune response, estimated to be 7-10 days post-infection, and therefore after the peak of viral loads. For children up to 10 years, IgG OD responses increased significantly (p<0.05) with age for spike, while IgA OD responses to spike and RBD presented a marginally significant (p<0.1) increase with age (Figure 2F). ## Discussion The infection of SARS-CoV-2 in children remains under-diagnosed and poorly understood. In particular, the role of children in viral transmission remains unclear, especially with the emerging variants that increase viral transmission, such as the delta variant. In our work, we corroborate findings that saliva molecular testing by methods similar to those described previously15 efficiently detects infected children, even in children unable to donate saliva (Figure 2A), for whom saliva was gently aspirated. Importantly, in children, the sensitivity, specificity, and accuracy of saliva-RT-qPCR tests compared to NP swab-RT-qPCR were respectively 84.8% (71.8%–92.4%), 100% (91.0%–100%), and 91.8% (84.0%–96.6%) with RNA extraction and 81.8% (68.0%–90.5%), 100% (91.0%–100%), and 90.4% (82.1%–95.0%) without RNA extraction. Hence, we show that methods bypassing RNA extraction have a sensitivity of 97.3% (36/37) compared with assays extracting RNA, and 100% (36/36) sensitivity considering CTs up to 36 with RNA extraction. We observed that discrepancies between saliva and NP swab-based molecular tests were found in children with specific IgM responses, which could suggest lower viral loads and prolonged infection. Interestingly, our study detected a high proportion of uninfected children with IgA antibodies in saliva. Antibodies in saliva may be acquired from the blood via the gingival crevicular fluid.27 However, there is a portion of IgAs that are produced locally in mucosal tissues, such as salivary glands, including secretory IgA (sIgA).25 While some sIgA antibodies undergo affinity maturation, others recognize specific glycosylation patterns26 and could justify the high levels detected in negative patients. This cross-reactive response has been reported by others in the mucosal tissue 28,29 and is considered an important property of sIgAs for protecting the mucosal tissue against infections. In addition, our study shows that SARS-CoV-2 infectious viruses may be rescued from saliva samples with CTs lower than 26 (Figure 2D, E). Data suggest that high viral loads are prone to result in viral transmission.18 In our study, SARS-CoV-2 viral growth was not associated with symptoms in children, but the sample size used was too low to draw conclusions regarding how symptoms relate to viral loads. In fact, it has been shown that asymptomatic children have significantly lower viral loads than symptomatic children.7 Overall, it is well-established that infected children are more likely to remain asymptomatic or have milder disease than adults; children are rarely hospitalized and rarely have fatal outcomes.1,6,9,10,30 However, data accumulated from 22 centers throughout South Korea has shown that 58% of symptomatic infected children experienced symptoms 3 days (median, in a range of 1-28 days) before SARS-CoV-2-positive diagnostic and that, despite having a detectable virus load, pre-symptomatic children remained symptom-free for 2.5 days (median, in a range of 1-25 days).6 For these reasons, the vast majority of SARS-CoV-2 infected children will continue to be missed by a symptom-based testing approach.6 With the unprecedented effort of vaccinating the world population above 12-years-old, it is critical to understand how children below 12-years-old contribute to viral circulation within their community. To this need, it is essential to implement efficient, easy, non-invasive, cheap methods for accurately identifying and tracking infected children, for which our method offers a solution. It is also critical to establish if children are a potential source for emerging novel SARS-CoV-2 variants, and saliva is suitable for collecting SARS-CoV-2 for genotyping13-15 and detecting antibodies specific to spike and RBD as we show here (Figure 2F and Table 3). How infection in children will impact breakthrough infections upon vaccination, which has been demonstrated as possible,31 remains to be elucidated and is of the utmost importance. Overall, our study provides a method highly suitable to identify children positive for SARS-CoV-2. It could be used for the surveillance of kindergartens and schools, and also as the first step in genotyping efforts to monitor known variants and spot novel ones when coupled to CRISPR-based methodologies.32 ## Limitations This cross-sectional study has one key limitation. There was an interval of up to 48 hours between SARS-CoV-2 infection diagnostic in children by NP swab and the collection of a saliva sample. Given this time interval, it remains unclear whether the saliva molecular testing has a lower sensitivity than the NP test, or whether there was a real decrease in the patient viral load between the two samples collection. In support of the former scenario, other studies conducted in adults reported a small detection decrease in saliva relative to NP swabs.15,33-37 Despite the temporal limitation, our saliva testing in children up to 10-years-old indicates a high sensitivity, specificity, and accuracy with and without RNA extraction, showing that the method is suitable for detecting infected children. An important note is that children were admitted to hospital for many reasons other than COVID-19 related symptoms and hence this study is suitable to draw conclusions for children regardless of their symptoms. A second limitation of this study is that the number of samples from which we rescued infective viral particles in cultured cells does not allow a suitable statistical analysis to relate viral loads with symptoms and is not a formal demonstration of transmission from children. A third caveat is that the ELISA assay, despite being specific for sera and collected from infected people,21 was not established for saliva. In fact, we did not calibrate the ELISA with pre-pandemic saliva, and as children were not re-analyzed posteriorly for antibody development, there is a lack of saliva positive and negative controls, which also limits the conclusions we may draw from the data. ## Data Availability The authors declare that the data supporting the findings of this study are available within the paper and its Supplementary Information files. ## Supplementary Materials for ![eFigure 1.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2021/08/12/2021.08.11.21261899/F3.medium.gif) [eFigure 1.](http://medrxiv.org/content/early/2021/08/12/2021.08.11.21261899/F3) eFigure 1. Standard curves obtained by RT-qPCR amplification of dilution series of synthetic copies of SARS-CoV-2 (20000 to 20) in fresh negative saliva samples, using the N1 and the N2 primer-probe sets. The obtained cycle threshold (CT) values were plotted vs. the copy number on a logarithmic scale. Each sample was run in quadruplicate. Error bars indicate the standard deviation. ![eFigure 2.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2021/08/12/2021.08.11.21261899/F4.medium.gif) [eFigure 2.](http://medrxiv.org/content/early/2021/08/12/2021.08.11.21261899/F4) eFigure 2. COVID-19 rapid antigen test detects SARS-CoV-2 in saliva samples positive and with CT values up to 26. Graphical representation of CT values in saliva after RNA extraction vs. rapid antigen test result, with dots representing symptomatic children, triangles asymptomatic patients. n.d., not-detected. The COVID-19 antigen rapid test from ALL TEST (ref. ICOV-502) was used accordingly to the manufacturer’s instructions, with the exception that instead of NP swab, saliva samples diluted 1:2 in extraction buffer were used. ## Acknowledgements This publication was supported by the European Virus Archive Global (EVA-GLOBAL) project that has received funding from the European Union’s Horizon 2020 research and innovation program under grant agreement No 871029. It was funded by the Fundação para a Ciência and Tecnologia (FCT, Portugal) under RESEARCH4COVID 19 call with reference 283_596885654 and co-funded by ANI under INOV4COVID (Funding to V.M.B.). M.J.A. is funded by the FCT (CEECIND/02373/2020). M.A. is funded by a Junior Researcher working contract from FCT and Instituto Gulbenkian de Ciência (IGC, Portugal). This work benefited from COVID19 emergency funds 2020 from Calouste Gulbenkian Foundation and from Oeiras city council. * Received August 11, 2021. * Revision received August 11, 2021. * Accepted August 12, 2021. * © 2021, Posted by Cold Spring Harbor Laboratory The copyright holder for this pre-print is the author. All rights reserved. The material may not be redistributed, re-used or adapted without the author's permission. ## References 1. 1.Guan WJ, Ni ZY, Hu Y, et al. Clinical Characteristics of Coronavirus Disease 2019 in China. N Engl J Med. 2020;382(18):1708–1720. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa2002032&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F08%2F12%2F2021.08.11.21261899.atom) 2. 2.Yang J, Zheng Y, Gou X, et al. Prevalence of comorbidities and its effects in patients infected with SARS-CoV-2: a systematic review and meta-analysis. Int J Infect Dis. 2020;94:91–95. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.ijid.2020.03.017&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F08%2F12%2F2021.08.11.21261899.atom) 3. 3.Wang D, Hu B, Hu C, et al. Clinical Characteristics of 138 Hospitalized Patients With 2019 Novel Coronavirus-Infected Pneumonia in Wuhan, China. JAMA. 2020;323(11):1061–1069. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1001/jama.2020.1585&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F08%2F12%2F2021.08.11.21261899.atom) 4. 4.Ledford H. Should children get COVID vaccines? What the science says. Nature. 2021;595:638–639 5. 5.Jones TC, Biele G, Muhlemann B, et al. Estimating infectiousness throughout SARS-CoV-2 infection course. Science. 2021;373(6551). 6. 6.Han MS, Choi EH, Chang SH, et al. Clinical Characteristics and Viral RNA Detection in Children With Coronavirus Disease 2019 in the Republic of Korea. JAMA Pediatr. 2021;175(1):73–80. 7. 7.Kam KQ, Thoon KC, Maiwald M, et al. SARS-CoV-2 viral RNA load dynamics in the nasopharynx of infected children. Epidemiol Infect. 2021;149:e18. 8. 8.Jones TC, Mühlemann B, Hofmann J, et al. An analysis of SARS-CoV-2 viral load by patient age. 2020. 9. 9.Lu X, Zhang L, Du H, et al. SARS-CoV-2 Infection in Children. N Engl J Med. 2020;382(17):1663–1665. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMc2005073&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F08%2F12%2F2021.08.11.21261899.atom) 10. 10.Chao JY, Derespina KR, Herold BC, et al. Clinical Characteristics and Outcomes of Hospitalized and Critically Ill Children and Adolescents with Coronavirus Disease 2019 at a Tertiary Care Medical Center in New York City. J Pediatr. 2020;223:14–19 e12. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F08%2F12%2F2021.08.11.21261899.atom) 11. 11.Mardi P, Esmaeili M, Iravani P, Abdar ME, Pourrostami K, Qorbani M. Characteristics of Children With Kawasaki Disease-Like Signs in COVID-19 Pandemic: A Systematic Review. Front Pediatr. 2021;9:625377. 12. 12.Ebina-Shibuya R, Namkoong H, Shibuya Y, Horita N. Multisystem Inflammatory Syndrome in Children (MIS-C) with COVID-19: Insights from simultaneous familial Kawasaki Disease cases. Int J Infect Dis. 2020;97:371–373. 13. 13.Carmagnola D, Pellegrini G, Canciani E, et al. Saliva Molecular Testing for SARS-CoV-2 Surveillance in Two Italian Primary Schools. Children (Basel). 2021;8(7). 14. 14.Jayamohan H, Lambert CJ, Sant HJ, et al. SARS-CoV-2 pandemic: a review of molecular diagnostic tools including sample collection and commercial response with associated advantages and limitations. Anal Bioanal Chem. 2021;413(1):49–71. 15. 15.Wyllie AL, Fournier J, Casanovas-Massana A, et al. Saliva is more sensitive for SARS-CoV-2 detection in COVID-19 patients than nasopharyngeal swabs. medRxiv. 2020;[https://doi.org/10.1101/2020.04.16.20067835](https://doi.org/10.1101/2020.04.16.20067835) 16. 16.Schildgen V, Demuth S, Lusebrink J, Schildgen O. Limits and Opportunities of SARS-CoV-2 Antigen Rapid Tests: An Experienced-Based Perspective. Pathogens. 2021;10(1). 17. 17.Ranoa DRE, Holland RL, Alnaji FG, et al. Saliva-Based Molecular Testing for SARS-CoV-2 that Bypasses RNA Extraction. bioRxiv. 2020; [https://doi.org/10.1101/2020.06.18.159434](https://doi.org/10.1101/2020.06.18.159434). 18. 18.van Kampen JJA, van de Vijver D, Fraaij PLA, et al. Duration and key determinants of infectious virus shedding in hospitalized patients with coronavirus disease-2019 (COVID-19). Nat Commun. 2021;12(1):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%2F2021%2F08%2F12%2F2021.08.11.21261899.atom) 19. 19.Vale-Costa S, Alenquer M, Sousa AL, et al. Influenza A virus ribonucleoproteins modulate host recycling by competing with Rab11 effectors. J Cell Sci. 2016;129(8):1697–1710. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NToiam9jZXMiO3M6NToicmVzaWQiO3M6MTA6IjEyOS84LzE2OTciO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMS8wOC8xMi8yMDIxLjA4LjExLjIxMjYxODk5LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 20. 20.Stadlbauer D, Amanat F, Chromikova V, et al. SARS-CoV-2 Seroconversion in Humans: A Detailed Protocol for a Serological Assay, Antigen Production, and Test Setup. Curr Protoc Microbiol. 2020;57(1):e100–e100. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/cpmc.100&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=32302069&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F08%2F12%2F2021.08.11.21261899.atom) 21. 21.Alenquer M, Ferreira F, Lousa D, et al. Amino acids 484 and 494 of SARS-CoV-2 spike are hotspots of immune evasion affecting antibody but not ACE2 binding. bioRxiv. 2021:2021.2004.2022.441007. 22. 22.Viana JF, Bergman M-L, Gonçalves LA, et al. Population homogeneity for the antibody response to COVID-19 BNT162b2 / Comirnaty vaccine is only reached after the second dose, across all adult age ranges. MedRxiv. 2021;2021.03.19.21253680. 23. 23.Newcombe RG. Improved confidence intervals for the difference between binomial proportions based on paired data. Stat Med. 1998;17(22):2635–2650. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/(SICI)1097-0258(19981130)17:22<2635::AID-SIM954>3.0.CO;2-C&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=9839354&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F08%2F12%2F2021.08.11.21261899.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000076941900009&link_type=ISI) 24. 24.Centers for Disease Control and Prevention USDoHaHS. Research Use Only 2019-Novel Coronavirus (2019-nCoV) Real-time RT-PCR Primers and Probes. 2020. 25. 25.Stadtmueller BM, Huey-Tubman KE, Lopez CJ, Yang Z, Hubbell WL, Bjorkman PJ. The structure and dynamics of secretory component and its interactions with polymeric immunoglobulins. Elife. 2016;5. 26. 26.Gonçalves J, Juliano AM, Charepe N, et al. Non-neutralizing secretory IgA and T cells targeting SARS-CoV-2 spike protein are transferred to the breastmilk upon BNT162b2 vaccination. medRxiv. 2021:2021.2005.2003.21256416. 27. 27.Ebersole JL. Humoral immune responses in gingival crevice fluid: local and systemic implications. Periodontol 2000. 2003;31:135–166. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1034/j.1600-0757.2003.03109.x&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=12657000&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F08%2F12%2F2021.08.11.21261899.atom) 28. 28.Woof JM, Mestecky J. Mucosal immunoglobulins. Immunological Reviews. 2005;206(1):64–82. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1111/j.0105-2896.2005.00290.x&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=16048542&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F08%2F12%2F2021.08.11.21261899.atom) 29. 29.Brandtzaeg P. Induction of secretory immunity and memory at mucosal surfaces. Vaccine. 2007;25(30):5467–5484. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.vaccine.2006.12.001&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=17227687&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F08%2F12%2F2021.08.11.21261899.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000248588200004&link_type=ISI) 30. 30.Hoang A, Chorath K, Moreira A, et al. COVID-19 in 7780 pediatric patients: A systematic review. EClinicalMedicine. 2020. 31. 31.Hacisuleyman E, Hale C, Saito Y, et al. Vaccine Breakthrough Infections with SARS-CoV-2 Variants. N Engl J Med. 2021;384(23):2212–2218. 32. 32.de Puig H, Lee RA, Najjar D, et al. Minimally instrumented SHERLOCK (miSHERLOCK) for CRISPR-based point-of-care diagnosis of SARS-CoV-2 and emerging variants. Science Advances. 2021;7(32):eabh2944. [FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6MzoiUERGIjtzOjExOiJqb3VybmFsQ29kZSI7czo4OiJhZHZhbmNlcyI7czo1OiJyZXNpZCI7czoxMzoiNy8zMi9lYWJoMjk0NCI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIxLzA4LzEyLzIwMjEuMDguMTEuMjEyNjE4OTkuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 33. 33.Chua GT, Wong JSC, To KKW, et al. Saliva viral load better correlates with clinical and immunological profiles in children with coronavirus disease 2019. Emerg Microbes Infect. 2021;10(1):235–241. 34. 34.Khurshid Z, Zohaib S, Joshi C, Moin SF, Zafar MS, Speicher DJ. Saliva as a non-invasive sample for the detection of SARS-CoV-2: a systematic review. 2020. 35. 35.Moreno-Contreras J, Espinoza MA, Sandoval-Jaime C, et al. Saliva sampling is an excellent option to increase the number of SARS CoV2 diagnostic tests in settings with supply shortages. bioRxiv. 2020;[https://doi.org/10.1101/2020.06.24.170324](https://doi.org/10.1101/2020.06.24.170324) 36. 36.Xu R, Cui B, Duan X, Zhang P, Zhou X, Yuan Q. Saliva: potential diagnostic value and transmission of 2019-nCoV. Int J Oral Sci. 2020;12(1):11. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41368-020-0080-z&link_type=DOI) 37. 37.Yee R, Truong TT, Pannaraj PS, et al. Saliva Is a Promising Alternative Specimen for the Detection of SARS-CoV-2 in Children and Adults. Journal of Clinical Microbiology. 2021;59(2):e02686–02620.