An affordable anti-SARS-COV-2 spike protein ELISA test for early detection of IgG seroconversion suited for large-scale surveillance studies in low-income countries ==================================================================================================================================================================== * Renata G. F. Alvim * Tulio M. Lima * Danielle A. S. Rodrigues * Federico F. Marsili * Vicente B.T. Bozza * Luiza M. Higa * Fabio L. Monteiro * Isabela C. Leitão * Renato S. Carvalho * Rafael M. Galliez * Terezinha M. P. P. Castineiras * Alberto Nobrega * Leonardo H. Travassos * Orlando C. Ferreira, Jr. * Amilcar Tanuri * André M. Vale * Leda R. Castilho ## Abstract **Background** Accurate serological tests are essential tools to allow adequate monitoring and control of COVID-19 spread. Production of a low-cost and high-quality recombinant viral antigen can enable the development of reliable and affordable serological assays, which are urgently needed to facilitate epidemiological surveillance studies in low-income economies. **Methods** Trimeric SARS-COV-2 spike (S) protein was produced in serum-free, suspension-adapted HEK293 cells. Highly purified S protein was used to develop an ELISA, named S-UFRJ test. It was standardized to work with different types of samples: (i) plasma or serum from venous blood samples; (ii) dried blood spots (DBS) from blood drops collected by finger prick. **Findings** We developed a cost-effective, scalable technology to produce S protein based on its stable expression in HEK293 cells. The S-UFRJ ELISA displayed 98.4% specificity and sensitivity above 90% already 10 days after symptoms onset, allowing early detection of anti-S IgG seroconversion. Endpoint titers were shown to correlate with virus neutralization assessed as PRNT90. There was excellent agreement between plasma and DBS samples, significantly simplifying sample collection, storing and shipping. The overall cost per test was estimated to be approximately one US dollar. **Interpretation** The S-UFRJ assay developed herein meets the quality requirements of high sensitivity and specificity. The low cost and the use of mailable DBS samples allow for serological surveillance of populations regardless of geographical and socio-economic aspects, with special relevance for public health policy actions in low-income countries. **Funding** CTG, Senai DN/CETIQT, FAPERJ, CNPq, CAPES and Instituto Serrapilheira. Keywords * SARS-COV-2 * espike protein ELISA * serological test * dried blood spots * COVID-19 * surveillance ## Introduction The COVID-19 pandemic brought an unprecedented global health crisis. The high transmissibility of SARS-COV-2 caused an avalanche of cases within a short period of time, having caused more than 17 million cases with more than 670,000 deaths in the first 7 months of 2020. In many cities, the number of infected individuals needing medical treatment exceeded hospitals capacity, and healthcare personnel became overwhelmed by the excess of working hours and by adverse mental health outcomes. In addition to this brutal scenario, the elevated human-to-human contagious rate obliged a severe restriction of social life including, in some cases, complete lockdown except for essential activities. As a result, this situation led to economic disruption in many countries experiencing several months of partial or complete lockdown. If easing of restrictive measures is unavoidable, it cannot be done without rigorous planning and tight follow-up. Thus, epidemiological surveillance studies are imperative to measure the spread of the disease and estimate seroprevalence, guiding policy decisions regarding the need to keep or ease social distance measures and to maintain or not other preventive measures. Serological assays are essential tools for epidemiological surveillance studies to monitor and control epidemics. The structural spike (S) protein of SARS-COV-2 is known to be a major target for neutralizing antibodies, thus making it a key antigen for the development of sensitive sero-diagnostic tests. S protein contains the receptor binding domain in its S1 subunit and is also responsible for fusion to the cell membrane through its S2 subunit1. Enzyme linked immunosorbent assays (ELISA) based on the S protein have been developed, showing minimal cross-reactivity with sera against circulating “common cold” coronaviruses2 and providing correlation to virus neutralizing activity3. The currently unavailability of low-cost and high-quality recombinant S protein is a bottleneck for the development of affordable and reliable serological tests urgently needed by public health agencies, especially in low-income economies, to deal with the pandemic. Here, we focused on establishing the hallmarks for a low-cost ELISA based on S protein immunoreactivity, suited to be employed in epidemiological surveillance studies also in remote regions and low-income countries with limited clinical laboratory network. Without compromising the performance of the assay, costs were cut mainly by optimizing antigen production and by simplifying sample collection and processing. Blood collection by finger prick, using dried blood spots (DBS), greatly facilitated collection, maintenance, shipping and processing of samples. The overall cost of the assay was estimated to be approximately one dollar per test. ## Methods ### Stable cell line generation HEK293-3F6 cells (NRC Canada) growing in suspension in the chemically-defined, animal component-free HEK-TF (Xell AG) culture medium were stably transfected by lipofection using a broad-spectrum reagent (Lipofectamine 3000, Thermo Fisher) as described previously4. A total DNA concentration of 0.9 µg/mL was used, combining two vectors: the pαH vector (at 0.75 µg/mL) containing the sequence encoding the ectodomain of the spike protein in the prefusion conformation1 (kindly provided by B. Graham, Vaccine Research Center of NIAID/NIH), and an empty vector containing the neomycin phosphotransferase gene (at 0.15 µg/mL). Cells were maintained under selection pressure with 100 μg/mL G-418 sulfate (Thermo Fisher) from 2 days post-transfection on. 24 days post-transfection cell viability had recovered to over 90% viability, and a cell bank was cryopreserved. All steps and materials used during recombinant cell line generation, expansion and cell banking were documented in detail. ### Cell cultivation Stably transfected cells were maintained at 37°C and 5% CO2 under orbital agitation (180 rpm, shaker with 5-cm stroke) in vented Erlenmeyer flasks containing up to 60% of their nominal volume of HEK-GM culture medium (Xell AG). In the experiments carried out in fed-batch mode, the culture medium was supplemented with a concentrated solution of nutrients (HEK-FS, Xell AG) according to the manufacturer’s instructions. Bioreactor runs were carried out in a 1.5-L stirred-tank bioreactor (EZControl, Applikon) at setpoints of pH, temperature and dissolved oxygen of 7.1, 37°C and 40% of air saturation, respectively. Cell concentration and viability were determined by trypan blue exclusion using an automated cell counter (Vi-Cell, Beckman Coulter), whereas glucose and lactate concentrations were monitored using a metabolite analyzer (YSI 2700, Yellow Springs Instruments). Presence of S protein in the supernatants was determined by spot blots: 3 µL of each sample was applied to nitrocellulose membranes, serum of SARS-COV-2 convalescent patients (1:1000) was used as primary antibody, followed by incubation with anti-human IgG(Fc)-HRP conjugate (Sigma, #SAB3701282) and finally chemiluminescent ECL reagent (BioRad). Images were captured using a FluorChem E system (ProteinSimple), setting exposure time to be automatically determined based on the samples with strongest signal. ### S protein concentration/purification Cell suspension harvested from cell cultures was clarified by filtration using 0.45-μm PVDF membranes and used to perform either ultrafiltration/diafiltration (UF/DF) or affinity chromatography (AC) experiments. Concentration/diafiltration (90-fold concentration by volume) was achieved by UF/DF using centrifugal devices based on cellulose membranes with 100-kDa cut-off (Merck Millipore). Affinity chromatography was carried out using a 5-mL StrepTrap XT affinity chromatography column (Cytiva) following manufacturer’s instructions. Protein concentration, purity and identity in the eluted fractions were confirmed by NanoDrop (Thermo Fisher), silver-stained SDS-PAGE and Western blot analyses, respectively. For Western blots, serum of SARS-COV-2 convalescent patients (1:1000) was used as primary antibody, followed by incubation with anti-human IgG (Fc)-HRP conjugate (Sigma, #SAB3701282) and then chemiluminescent ECL reagent (BioRad). ### S-UFRJ ELISA for anti-S IgG detection High binding ELISA plates (Corning) were coated with 50 μL of SARS-COV-2 S protein in PBS (Gibco) at 4 μg/mL (unless otherwise stated in the Figures) and incubated overnight at room temperature (RT). The coating solution was removed, and 150 μL of PBS 1% BSA (blocking solution) was added to the plate and incubated at RT for 1-2 hours. The blocking solution was removed, and 50 μL of serum or plasma diluted 1:40 in PBS 1% BSA, and serially three-fold diluted, was added to the plate and incubated at RT for 2 hours. For ELISA using dried blood spots, at first circles cut from filter paper using commercially available punching devices, or single filter paper pads cut from plastic strips were used to prepare eluates by incubating for 1 hour at RT in 100 µL (filter paper circles) or 200 µL (pads) of PBS 1% BSA. In the same way as diluted serum or diluted plasma, 50 µL of eluate samples were added to the plate and incubated at RT for 2 hours. The plate was then washed five times with 150 μL of PBS. Next, 50 μL of 1:8000 goat anti-human IgG (Fc)-horseradish peroxidase antibody (Sigma, #SAB3701282) was added to the plate and incubated for 1.5 hours at RT. The plate was then washed five times with 150 μL of PBS. At the end, the plate was developed with TMB (3,3’,5,5;-tetramethylbenzidine) (Thermo Fisher). The reaction was stopped with 50 µL of 1 N HCl, and the optical density (O.D.) was read at 450 nm with 655 nm background compensation in a microplate reader (BioRad). Results are expressed either as O.D. or O.D. summation5. ### Rapid diagnostic test (RDT) A commercial rapid immunochromatographic test (MedTeste, Medlevensohn, Brazil) with separate bands for IgM and IgG detection, approved by the Brazilian regulatory agency ANVISA (Agência Nacional de Vigilância Sanitária), was used following manufacter’s instructions. ### Sample collection from human subjects Samples collected at the State Hematology Institute Hemorio followed a protocol approved by the local ethics committee (CEP Hemorio; approval #4008095). Samples collected at UFRJ COVID Screening Center followed a protocol approved by the national ethics committee (CONEP, Brazil; protocol #30161620000005257; approval #3953368): subjects were initially interviewed and, if they accepted to participate, they signed the informed consent, answered a questionnaire (addressing demographic data, onset and type of symptoms, history of travel abroad, among other information) and had blood (venous blood and/or finger prick) and nasopharyngeal swab collected. Only symptomatic subjects who presented at least two of the following symptoms were included: loss of taste or smell, fever, shortness of breath, diarrhea, headache, extreme tiredness, dry cough, sore throat, runny or stuffy nose, or muscle aches. Dried blood spots (DBS) were obtained by finger pricking with commercially available sterile lancets and lancing devices. Either plastic strips containing pads of filter paper, or 2.5 cm (W) x 7.5 cm (L) filter paper with three blood spots from the same volunteer, were used to collect whole blood from finger pricks. ### Plaque reduction neutralization test (PRNT) In order to determine the titers of neutralizing antibodies in serum samples, sera were first heat-inactivated at 56°C for 30 min, and two-fold serial dilutions were incubated with 100 PFU of SARS-COV-2 (GenBank #[MT126808.1](http://medrxiv.org/lookup/external-ref?link_type=GEN&access_num=MT126808.1&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F02%2F2020.07.13.20152884.atom)) for 1 hour at 37°C to enable neutralization to occur. Virus-serum mixture was inoculated into confluent monolayers of Vero cells seeded in 12-well tissue culture plates. After 1 hour, inoculum was removed and a semisolid medium (1.25% carboxymethylcellulose in alpha-MEM supplemented with 1% fetal bovine serum) was added. Cells were further incubated for 72 hours and then fixed with 4% formaldehyde solution. Viral plaques were visualized after staining with crystal violet dye solution. PRNT end-point titers were expressed as the reciprocal of the highest serum dilution for which the virus infectivity is reduced by ≥90% (PRNT90) when compared with the average plaque count of the virus control. All work involving infectious SARS-COV-2 was performed in a biosafety level 3 (BSL-3) containment laboratory. ### Role of the funding sources The funders had no role in study design, data collection, data analysis, data interpretation, or writing of the report. The corresponding authors had full access to all data in the study and had final responsibility for the decision to submit for publication. ## Results For recombinant production and straightforward purification of a heavily glycosylated protein such as the SARS-COV-2 spike protein, mammalian cell culture in serum-free media is the option of choice. We expressed the soluble ectodomain of the spike (S) protein in a stabilized prefusion conformation1 in serum-free, suspension-adapted cell lines. Initial transient transfections indicated that expression levels evaluated on days 2 and 4 post-transfection were higher in HEK293 cells than in CHO-K1 cells (Fig. 1A, left panel). However, differently from previous studies that adopt transient protein expression techniques3, we focused on stable and constitutive gene expression because transgene integration in the cell genome enhances scalability and significantly decreases costs of recombinant protein production in mammalian cells4. Due to the challenges posed by the pandemic, such as the urgency and the disruption of international supply chains for reagents and synthetic gene constructs, we decreased time and costs by using an old-fashioned technique of co-transfecting the plasmid containing the S gene with an intellectual property-free plasmid containing an eukaryotic selection marker. A stable recombinant HEK293 cell line showing higher expression than transiently transfected HEK293 cells was generated (Fig. 1A, center panel) and banked 24 days post-transfection. Stability of S protein expression by this cell line has been proven so far for up to 100 days post-transfection (Fig. 1A, right panel). Hence, confirmation of expression stability is required for developing cost-saving, long-lasting batch-refeed or perfusion technologies for cell culture. ![Figure 1:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2020/08/02/2020.07.13.20152884/F1.medium.gif) [Figure 1:](http://medrxiv.org/content/early/2020/08/02/2020.07.13.20152884/F1) Figure 1: S protein production by stable recombinant HEK293 cells and its purification from cell culture supernatant. **(A)** Left panel: CHO-K1 and HEK293-3F6 were transiently transfected with plasmid DNA containing the S protein gene. Center panel: due to the higher levels of protein secreted by HEK293 cells, this cell line was used in transient transfections at high (H, 2 µg/mL of pαH plasmid) and low (L, 0.75 µg/mL of pαH plasmid) DNA concentration, as well as in a stable transfection that comprised co-transfection of a second vector containing the neo selection marker. A well documented cell bank was cryopreserved on day 24 post-transfection. Right panel: stability of expression of the secreted protein has been confirmed so far for up to 100 days post-transfection. **(B)** Left panel: high cell viabilities and viable cell densities (VCD) were achieved in the cultivation of the stable cell line in shake flasks and 1.5-L stirred-tank bioreactors, in batch and fed-batch mode, using chemically-defined, animal component-free media. Right panel: spot blot for detection of S protein in the cell culture supernatant on different days of culture, showing high titers of secreted S protein. **(C)** S protein identity was confirmed by Western blot analysis, confirming the molecular mass of approximately 180 kDa. SDS-PAGE showed a rather low purity for the concentrated/diafiltered (UF/DF) sample, but a very high purity for the sample purified by affinity chromatography (AC). 1: Cell culture supernatant from non-transfected parental HEK293-3F6 cells. 2: Molecular-mass standard containing proteins at 250, 150, 100 and 75 kDa. 3: UF/DF sample after 90-fold volumetric concentration and buffer exchange into PBS. 4. Eluate from affinity chromatography. 5. Supernatant from stable recombinant cell line. **(D)** A typical chromatogram of the affinity chromatography purification. Fractions of the flow-through (fr1 to fr8) were collected during sample injection and analyzed by spot blot to evaluate progressive saturation of affinity ligands, showing a high binding capacity and a low degree of S protein leakage. For comparison, spot blots of the injected cell culture supernatant, or a 1:10 dilution, are shown in the box above the chromatogram. In all immunoblots, presence of S protein was detected using a pooled serum of SARS-COV-2 convalescent patients (1:1000) as primary antibody, followed by incubation with anti-human IgG-HRP and chemiluminescent detection. In order to decrease costs and logistics burden, cell culture media available as dried powdered media were selected to be tested both in shake flasks and stirred-tank bioreactors at 300-mL and 1.5-L scale. The chosen medium (HEK-GM) was able to provide robust cell growth and efficient recombinant protein production (Fig. 1B). Carrying out cell culture in fed-batch mode by adding pulses of a concentrated nutrient solution over cell cultivation time avoided nutrient depletion and significantly increased viable cell density and secreted S protein levels, allowing enhanced production of the recombinant protein (Fig. 1B, right panel). Protein isolation from cell culture supernatant was investigated by ultrafiltration/diafiltration (UF/DF) and affinity chromatography (AC) techniques. In spite of the large S protein size (∼180 kDa) and the use of a 100-kDa cut-off membrane device, UF/DF was not able to efficiently remove all smaller contaminating proteins, and an AC resin bearing a streptavidin mutein ligand was used to obtain the protein in high purity (Fig. 1C). The affinity resin has shown a relatively high dynamic binding capacity for the S protein (Fig. 1D), and has so far been used for 45+ adsorption/elution/regeneration cycles with no detectable decrease in performance, reducing its impact on the final costs of the purified protein. S protein preparations of lower (UF/DF) or higher purity (AC) were then used in ELISAs for the detection of anti-S antibodies in plasma samples. Nineteen positive or negative samples, collected from individuals scored as positive by PCR for SARS-COV-2 (n=16), from a non-exposed individual (“post-COVID”, n=1) or obtained before SARS-COV-2 emergence (“pre-COVID”, n=2), were tested for immunoreactivity against S protein, processed either by UF/DF or AC. Comparison of the results of the different ELISAs showed that the assay performance could be greatly improved by the use of the highly purified AC antigen, showing much better discrimination between positive samples and negative controls (Fig. 2A). For comparison, the samples were evaluated also by means of an imported rapid diagnostic test (RDT), which has been approved by the Brazilian regulatory agency (ANVISA) for commercialization in the country. Interestingly, sample #1, which was positive by PCR, but negative for IgG by RDT, clearly scored as positive in the AC S-protein ELISA. Better discrimination was also evident in titration curves (Fig. 2B), which were used to calculate the O.D. summation for each sample, evidencing again the superiority of the highly pure AC protein as ELISA antigen (Fig. 2C). Using selected plasma samples from negative controls and PCR-confirmed patients, we established that a minimum of 150 ng of high-purity S protein per well enables clear discrimination of positive and negative samples (Fig. 2D). The ELISA test with AC-purified S protein was named S-UFRJ test. ![Figure 2:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2020/08/02/2020.07.13.20152884/F2.medium.gif) [Figure 2:](http://medrxiv.org/content/early/2020/08/02/2020.07.13.20152884/F2) Figure 2: Comparison of ELISAs with UF/DF- or AC-purified S protein. **(A)** ELISA performance using low-purity (UF/DF) or high-purity (AC) S protein antigen to coat plates. A total of 22 samples were used: 15 sera from COVID-19 convalescent patients (SARS-COV-2 samples, open circle), two control samples collected until 2018 (pre-COVID-19, yellow diamond), one post-COVID-19 control sample from a healthy individual, two samples from SARS-COV-2 infected individuals characterized as PCR+/RDT-(#1 light gray filled circle, #2 dark gray filled circle), and one sample from a SARS-COV-2 convalescent patient who had the severe form of the disease (#3, red square). The cut-off for antigen evaluation was defined as [mean + 3 standard deviations (X+3SD)] of the O.D. of negative controls. **(B)** Samples used in (A) were titrated in four serial dilutions (1:40, 1:120, 1:360 and 1:1080), both for UF/DF or AC ELISA. **(C)** O.D. summation of the titration curves shown in (B); symbols as in (A). Next we applied the S-UFRJ ELISA to evaluate the presence of anti-S IgG in 210 samples, including pre-COVID negative controls and samples from symptomatic individuals diagnosed by PCR as positive for SARS-COV-2. The PCR-positive cohort consisted of 38 symptomatic individuals whose blood samples were obtained at different timepoints after symptoms onset, comprising a total of 66 samples. Negative control samples were collected either until 2018 from healthy individuals (pre-COVID, n=124) or in 2020 from healthy individuals who tested negative for SARS-COV-2 by PCR (post-COVID controls, n=20), comprising a total of 144 negative control samples. The assay exhibited high specificity (98.61%), as 142 out of 144 controls scored as negative, below the cut-off (Fig. 3A). Considering all samples from PCR-positive individuals, regardless of the time of collection after symptoms onset, 53 out of 66 samples (80.30 % overall sensitivity) were IgG positive by the S-UFRJ ELISA (Fig. 3A). For comparison, all samples were also tested for IgG using the RDT, and only 30 out of 66 samples were positive for IgG (45.45% overall sensitivity for the commercial rapid test) (Fig. 3B). ![Figure 3:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2020/08/02/2020.07.13.20152884/F3.medium.gif) [Figure 3:](http://medrxiv.org/content/early/2020/08/02/2020.07.13.20152884/F3) Figure 3: Validation of S-UFRJ ELISA, which comprises the AC-purified antigen, for early detection and quantification of anti-S IgG serum antibodies. **(A)** Anti-S IgG antibody detection in samples from healthy individuals, obtained pre-COVID-19 (n=124, yellow diamond) or post-COVID-19 who tested negative for the virus by PCR (n=20, open square), and from COVID-19 patients who were PCR-positive (n=66, open circle). Relative levels of antibodies are shown as O.D. ratio of values of individual samples to the [mean + 1 standard deviation (X+1SD)] of the O.D. average of the negative controls in the same ELISA plate. For routine use of the S-UFRJ ELISA, sera samples were diluted at 1:40. An O.D. ratio below 1 indicates a negative result (N), an O.D. ratio above the cut-off (C.O.) of 2 indicates a positive result (P), and an O.D. ratio between 1 and 2 is considered undetermined (U). **(B)** The PCR-positive samples shown in (A) were tested for anti-SARS-COV-2 IgG by a commercial rapid diagnostic test (RDT) (MedTeste, Medlevensohn, Brazil). Samples were then grouped by IgG reactivity according to the RDT result (RDT IgG-, open circle; RDT IgG+, red filled circle), and their anti-S IgG levels measured by the S-UFRJ ELISA are plotted. **(C)** Anti-S IgG levels measured by the S-UFRJ ELISA for the RDT IgG-negative samples shown in (B), grouped according to the RDT IgM result as RDT IgM-(open circle) or RDT IgM+ (open red circle). **(D)** Levels of anti-S IgG in samples grouped according to the timepoint of sample collection given in days after symptoms onset (DASO); symbol legends are indicated in the figure. **(E)** Positive rates versus DASO for anti-S IgG measured by S-UFRJ ELISA (open circle), for IgM measured by RDT (open red circle) and for IgG measured by RDT (red filled circle). **(F)** S-UFRJ IgG titration of 16 samples from COVID-19 convalescent patients (SARS-COV-2 samples); samples with the highest and lowest endpoint titer of the group are indicated in solid red and solid black colors, respectively. **(G)** Correlation between S-UFRJ ELISA IgG endpoint titers and neutralization (PRNT90) for samples titrated in (F) (n=16). Three samples with PRNT90 values <10 are plotted as 1. Statistical analysis was performed using Pearson’s test. In order to gain insight into the samples from symptomatic PCR-positive individuals that scored negative for anti-S IgG in the S-UFRJ ELISA, samples were grouped as IgM- and IgM+ according to the RDT (Fig. 3C). We found that 14 out of 15 samples that scored negative for anti-S IgG in our ELISA, also scored as negative for IgM in the rapid test (Fig. 3C). This result suggests that samples from PCR+ individuals that scored negative for anti-S IgG in our ELISA, may have been collected in the very beginning of the disease, only a few days after symptoms onset (DASO), and for that reason also scored negative for IgM in the RDT. Indeed, when the results of S-UFRJ ELISA were charted against DASO, this was confirmed: samples were increasingly scored as positive for anti-S IgG according to the time point they had been collected after symptoms onset (Fig. 3D). Accordingly, PCR+ individuals that have been sequentially sampled on different DASO and scored negative in their first sampling, later converted to seropositivity for anti-S IgG (data not shown). Anti-S IgG seroconversion rate, as scored by S-UFRJ ELISA, increased progressively from 41.66% (days 0-4) to 100% (20+ days) as a function of DASO, being above 90% for all samples collected 10 or more days after symptoms onset (Fig. 3E). Of note, S-UFRJ ELISA detected anti-S IgG seroconversion of over 41% of PCR-positive individuals already at 0-4 DASO, a time window where the rapid test detected no IgG seroconversion. Even for DASO of 20+ days, the RDT reached a positive rate of 71.43%, whereas the S-UFRJ ELISA scored a rate of 100%. The results show the superiority of S-UFRJ ELISA when compared to the commercial RDT used for comparison, with higher sensitivity and earlier detection of seroconversion in PCR+ symptomatic individuals. A previous study using S protein-based ELISA has observed a correlation between anti-S or anti-RBD (the receptor binding domain within the S protein) IgG titers and virus neutralization3. To address whether the S-UFRJ ELISA could also bring information about the neutralizing capacity of positive samples, we compared anti-S IgG titers and their virus neutralizing titers. We tested plasma samples of COVID-19 convalescent donors for their SARS-COV-2 in vitro neutralization capacity as measured by classic plaque reduction neutralization test (PRNT), using a Brazilian SARS-COV-2 isolate. Importantly, analogously to what has been shown by Amanat and colleagues3, we found that the higher the anti-S IgG ELISA endpoint titers (Fig. 3F), also the higher the neutralization titers were (Fig. 3G), resulting in a high correlation between those titers (Pearson’s R=0.9143, p<0.00001). Hence, S-UFRJ ELISA also provides an important functional correlation with SARS-COV-2 neutralization capacity. The cost per sample of serological assays for COVID-19 is an important aspect concerning large-scale public health actions, especially in low-income countries. ELISA samples are routinely based on venous blood collection followed by serum or plasma preparation. This requires clinical laboratory services, as well as refrigerated sample storage and transport, which would significantly increase the cost of the S-UFRJ ELISA. More relevant, it would severely limit the usage of the assay for epidemiological surveillance studies in world regions lacking an appropriate network of clinical laboratories. To overcome this critical limitation, we evaluated a simple means for storage and transport of blood samples, by collecting blood drops obtained by finger prick in filter paper, resulting in dried blood spots (DBS) that enable sampling in remote regions, or regions lacking a laboratory network. For that, the assay was further standardized to use eluates from DBS (Fig. 4A), a simple, low-cost and low-complexity method6. Importantly, titration curves of plasma samples, or eluates from DBS collected in filter paper, displayed comparable results (Figs. 4B and 4C). Additionally, plastic strips with one or more filter paper pads allowed further precision in sampling and storing for eventual retesting (Fig. 4D). Consistently, O.D. ratios obtained for plasma samples and DBS, either in filter paper or in pads, showed very high correlation (Figs. 4E and 4F), as well as high reproducibility between pads from a given strip (Fig. 4G). Thus, by using dried blood spots, the low cost of the test was further warranted. We have confirmed that blood samples collected in filter paper and kept in plastic bags can be preserved for at least 2 months without altering their serological result (data not shown). Importantly, the elution volume of 200µL used for DBS pads allows at least four experiment replicates per pad. Additionally, as each plastic strip can contain multiple pads, it also permits additional testing for immunoreactivity against other SARS-COV-2 antigens (e.g. RBD or nucleocapsid protein), or for cross-reactivity evaluation against related viruses2. ![Figure 4:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2020/08/02/2020.07.13.20152884/F4.medium.gif) [Figure 4:](http://medrxiv.org/content/early/2020/08/02/2020.07.13.20152884/F4) Figure 4: S-UFRJ test optimization for sample collection amenable for field studies. **(A)** Dried blood spots (DBS) obtained by finger pricking with commercially available lancing devices: a 2.5 cm (W) x 7.5 cm (L) filter paper with three blood spots from the same volunteer and a commercially available paper hole punching device were used to prepare a DBS disk (arrowhead) from which blood was eluted for ELISA testing. **(B)** S-UFRJ ELISA comparing the O.D. values for plasma samples in increasing serial dilutions and for the corresponding eluates prepared by incubating the DBS disks in increasing volumes of buffer. **(C)** O.D. summation of the data shown in (B). **(D)** Dried blood spots collected in plastic strips containing 1, 2 or 3 pads of filter paper. **(E-G)** correlations of O.D. ratios between samples collected in the strip pad 1 and either the respective plasma sample (E), the respective DBS in filter paper (F), or the second pad of the same strip (G). In E-G, plasma dilution was 1:40, DBS in filter paper disks were eluted in 100 µL of PBS 1% BSA, and DBS in single pads were eluted in 200 µL PBS 1% BSA. Statistical analysis was performed using Pearson’s test. Taking into account the simplicity of the DBS method, the overall cost of the S-UFRJ test is significantly decreased. In terms of raw materials (consumables) needed for collecting samples and performing the test, a detailed calculation revealed that consumables costs vary between two and four USD dimes, for two scenarios that were considered (Supplementary Table S1): one scenario considering items sold by international vendors in the country, and the second one considering items produced locally and adopting other saving measures that we confirmed not to compromise the performance of the test (data not shown). Based on techniques routinely employed in economic feasibility studies7, as shown in Supplementary Table S2 the final price of the test was estimated by including in the computation other manufacturing costs (that include labor, utilities, laboratory charges, insurance, among others) and general expenses (which account for administrative, research, development, distribution and marketing costs). Costs can vary significantly from country to country, but we expect that the two scenarios in Tables S1 and S2 can represent most realities reasonably well. As shown in Table S2, the final cost estimated for the S-UFRJ test from sample collection to test result is approximately USD 1 per sample. ## Discussion Serological tests for the detection of anti-SARS-COV-2 antibodies are instrumental for epidemiological surveillance studies in the present COVID-19 global health crisis. Assessing the spread of SARS-COV-2 with the help of serological assays provides the data input for policy decisions about social distancing and other preventive measures. As recently discussed for the case of Spain8, population-based epidemiological studies are a key element to inform authorities about the need for maintaining public health measures to avoid new epidemic waves, thus allowing scientifically driven decision making by governments. However, reliable epidemiological data require reliable diagnostic tests. A recent systematic review and meta-analysis on serological tests for COVID-19 showed that the evidence on the accuracy of existing point-of-care lateral flow tests (rapid diagnostic tests) is particularly weak and does not support their continued use9. Traditional ELISA tests and chemiluminescent assays (CLIAs) showed better performance9, but depend on collecting venous blood to obtain serum or plasma, thus being demanding in regard to sample collection, storage and transport. Thus, S-UFRJ ELISA emerges as a promising tool for epidemiological surveillance. It combines low cost and sample collection by finger prick/DBS, with the important advantage of a high accuracy, given by a specificity of 98.61% and a sensitivity greater than 90% for 10-19 days after symptoms onset and 100% thereafter. The high quality of the assay is warranted by the use of the spike protein, which is less conserved in coronaviruses and thus less prone to induce cross-reactivity10-11. Moreover, the recombinant spike protein used herein consists of the full-length ectodomain and is produced in the prefusion trimeric conformation, thus maintaining the most neutralization-sensitive epitopes1 and reliably mimicking the antigenic properties of the virus upon infection and spread in the organism. Both the poor accuracy and high costs of rapid diagnostic tests that are currently widespread around the globe pose major obstacles for large-scale epidemiological surveillance. In some countries these assays can reach values up to USD 100 per sample. The S-UFRJ test, based on DBS eluates from finger pricks and showing a much superior performance when compared to an imported RDT broadly commercialized in Brazil, has an estimated cost of just one US dollar. Its use can not only enable broad and reliable serological surveillance in populations regardless of their geographical and socio-economic aspects, but can also allow governments worldwide to save very significant amounts of financial resources that could be invested in other types of countermeasures to fight the health and socio-economic consequences of COVID-19 pandemic. ## Data Availability The raw data of all experiments are available upon request. ## Contributors AMV and LRC had the idea for the study and contributed to study design. RGFA, TML, DASR and AMV performed most experiments and analyzed data. FFM, VBTB, RSC and LHT gave practical laboratory support and performed analyses. LMH and FLM performed the virus neutralization assays. ICL, RMG, TMPPC, OCFJ and AT contributed to sample collection and characterization. LHT, AN, OCFJ, AMV and LRC contributed to data analysis and interpretation. AN, AMV and LRC wrote the manuscript. LMH, LHT, RSC, RGFA, TML and AT contributed to a detailed revision of the manuscript. ## Declaration of interests We declare no competing interests. ## Supplementary material View this table: [Supplementary Table S1:](http://medrxiv.org/content/early/2020/08/02/2020.07.13.20152884/T1) Supplementary Table S1: Cost estimate of consumables needed for S-UFRJ test, considering two scenarios: (1) purchase of materials sold by international suppliers in the country and with no saving efforts; (2) purchase of materials from domestic suppliers and making saving efforts that were validated as not interfering in test performance (data not shown): use of skim milk (2% m/v) instead of BSA (1% m/v); in-house preparation of PBS; finger prick using just the sterile lancet, with no lancing device; and reuse of buffer reservoirs for an equivalent of 30 ELISA plates. As a conservative estimate to account for controls included in each plate and for any eventual repetition needs or failures, a total of 80 tests were considered per each ELISA plate. All quotes for consumables in this table were obtained in June-July 2020 for purchase in the local market in Brazilian reais (BRL). The conversion to USD was done considering an exchange rate of 1 USD = 5.34 BRL (as of July 16, 2020). View this table: [Supplementary Table S2:](http://medrxiv.org/content/early/2020/08/02/2020.07.13.20152884/T2) Supplementary Table S2: Estimate of the final S-UFRJ test price taking 1 million tests as a calculation basis, including in the cost computation the raw materials shown in Table S1, other manufacturing costs (that include labor, utilities, laboratory charges, insurance, among others) and general expenses (which account for administrative, research, development, distribution and marketing costs)7. Profit margin and taxes were also accounted for to estimate the final S-UFRJ test price, since large-scale use of the test for epidemiological surveillance would probably require its industrial production. ## Acknowledgements Authors gratefully acknowledge Dr. B. Graham and Dr. K. Corbett (VRC/NIAID/NIH, USA) for sharing the plasmid encoding the S protein gene construct; Prof. E. Durigon (USP, Brazil) for providing the SARS-COV-2 isolate used in neutralization assays; Dr. S. Garcia and Dr. L. Amorim from Hemorio for plasma samples; all study participants for donation of blood samples; and the team of volunteers working in the UFRJ COVID-19 screening center. We thank D. Mucida, C. Lucas and M. Bellio for critical reading and editing of the manuscript. This work was supported by Senai CETIQT, Senai DN and CTG, and by the Brazilian research funding agencies Fundação de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ), Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq), Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (CAPES) and Instituto Serrapilheira. ## Footnotes * * joint senior authors * i) The main text was fully updated. ii)Figures 1 and 2 were split into four figures for clarity. iii)Supplementary Tables 1 and 2 were added to include a cost estimate of consumables needed for S-UFRJ test. * Received July 13, 2020. * Revision received August 1, 2020. * Accepted August 2, 2020. * © 2020, Posted by Cold Spring Harbor Laboratory This pre-print is available under a Creative Commons License (Attribution-NonCommercial-NoDerivs 4.0 International), CC BY-NC-ND 4.0, as described at [http://creativecommons.org/licenses/by-nc-nd/4.0/](http://creativecommons.org/licenses/by-nc-nd/4.0/) ## References 1. 1.Wrapp D, Wang N, Corbett KS, et al. Cryo-EM structure of the 2019-nCoV spike in the prefusion conformation. Science 2020; 367:1260–3. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Mzoic2NpIjtzOjU6InJlc2lkIjtzOjEzOiIzNjcvNjQ4My8xMjYwIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjAvMDgvMDIvMjAyMC4wNy4xMy4yMDE1Mjg4NC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 2. 2.Freeman B, Lester S, Mills L, et al. Validation of a SARS-CoV-2 spike protein ELISA for use in contact investigations and sero-surveillance. Biorxiv 2020; [https://doi.org/10.1101/2020.04.24.057323](https://doi.org/10.1101/2020.04.24.057323). 3. 3.Amanat F, Stadlbauer D, Strohmeier S, et al. A serological assay to detect SARS-CoV-2 seroconversion in humans. Nat Med 2020; 26:1033–6. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F08%2F02%2F2020.07.13.20152884.atom) 4. 4.Alvim RGF, Itabaiana Jr I, Castilho LR. Zika virus-like particles (VLPs): stable cell lines and continuous perfusion processes as a new potential vaccine manufacturing platform. Vaccine 2019; 37:6970–7. 5. 5.Hartman H, Wang Y, Schroeder Jr HW, Cui X. Absorbance summation: A novel approach for analyzing high-throughput ELISA data in the absence of a standard. PLoS One 2018; 13:e0198528. 6. 6.Lim MD. Dried blood spots for global health diagnostics and surveillance: opportunities and challenges. Am J Trop Med Hyg 2018; 99:256–65. 7. 7.Peters MS, Timmerhaus KD, West RE. Plant Design and Economics for Chemical Engineers, McGraw-Hill Education, New York, 5th edition, 2003. 8. 8.Pollán M, Pérez-Gómez B, Pastor-Barriuso R, et al. Prevalence of SARS-CoV-2 in Spain (ENE-COVID): a nationwide, population-based seroepidemiological study. Lancet 2020; [https://doi.org/10.1016/S0140-6736(20)31483-5](https://doi.org/10.1016/S0140-6736(20)31483-5). 9. 9.Bastos ML, Tavaziva G, Abidi SK, et al. Diagnostic accuracy of serological tests for covid-19: systematic review and meta-analysis. BMJ 2020; 370:m2516. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiYm1qIjtzOjU6InJlc2lkIjtzOjE4OiIzNzAvanVsMDFfMTEvbTI1MTYiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMC8wOC8wMi8yMDIwLjA3LjEzLjIwMTUyODg0LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 10. 10.Forcelloni S, Benedetti A, Dilucca M, Giansanti A. Identification of conserved epitopes in SARS-CoV-2 spike and nucleocapsid protein. Biorxiv 2020; [https://doi.org/10.1101/2020.05.14.095133](https://doi.org/10.1101/2020.05.14.095133). 11. 11.Krammer F, Simon V. Serology assays to manage COVID-19. Science 2020; 368:1060. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Mzoic2NpIjtzOjU6InJlc2lkIjtzOjEzOiIzNjgvNjQ5NS8xMDYwIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjAvMDgvMDIvMjAyMC4wNy4xMy4yMDE1Mjg4NC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=)