AN AFFORDABLE ANTI-SARS-COV-2 SPIKE 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 We describe a cost-effective, scalable technology to produce SARS-COV-2 spike (S) protein based on stable expression in HEK293 cells, and its use to develop a highly specific and sensitive ELISA test. The assay allows early detection of anti-S IgG seroconversion and endpoint titers correlate with virus neutralization. The low-cost S-antigen production, together with sample collection by finger prick and dried blood spots, allowed the development of a half-dollar test that fits the urgent need for large-scale serological surveillance in low-income countries. Keywords * COVID-19 * epidemiology * surveillance * ELISA ## MAIN TEXT Tools to monitor and control COVID-19 are urgently needed in the context of the international public health emergency posed by SARS-COV-2 coronavirus pandemics. High-quality recombinant viral antigens are an essential tool to enable the development of serological assays needed for seroprevalence and epidemiological surveillance studies, contact mapping, identification of asymptomatic infections, evaluation of potential vaccine candidates and fundamental research on coronavirus immunity. The structural spike (S) protein of SARS-COV-2 contains the receptor binding domain in its S1 subunit and is also responsible for fusion to the cell membrane through its S2 subunit. S protein is known to be a major target for neutralizing antibodies, thus making it a key antigen for the development of sensitive diagnostic tools. Enzyme linked immunosorbent assays (ELISA) based on the S protein have been developed, showing minimal cross-reactivity with sera against commonly circulating coronaviruses [1] and providing a good correlation to neutralizing activity [2]. Here, we focused on establishing the hallmarks for a low-cost ELISA based on S protein immunoreactivity, which can be broadly employed in epidemiological surveillance studies in low-income countries. Without compromising the performance of the assay, costs were cut mainly by optimizing antigen production and adapting sample collection and processing, resulting in a total cost estimated in less than half a dollar per sample. For recombinant production and straightforward purification of the heavily glycosylated S protein, mammalian cell culture in serum-free media is the option of choice. We expressed the soluble ectodomain of the spike (S) protein of SARS-COV-2 in a stabilized prefusion conformation [3] in serum-free, suspension-adapted HEK293-3F6 cells. Differently from previous works that use transient protein expression techniques [2,3], we focused on stable, constitutive gene expression, since transgene integration in the cell genome enhances scalability and significantly decreases costs of recombinant protein production in mammalian cells [4]. Due to the challenges posed by the pandemics, 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 and an intellectual property-free plasmid containing a selection marker. A stable recombinant HEK293 cell line showing higher expression than transiently transfected HEK293 or CHO-K1 cells was generated and banked within 24 days post-transfection, and stability of expression by this cell line has so far been proven for up to 100 days post-transfection (Fig. 1a), thus making it feasible to develop less costly, long-lasting batch-refeed or perfusion technologies for cell culture. In order to decrease cell cultivation costs and logistics burden, different cell culture media available as dried powdered media were evaluated in shake flasks and stirred-tank bioreactors at 300-mL and 1.5-L scale, respectively (data not shown). The less expensive HEK-GM medium was able to provide robust cell growth and efficient recombinant protein production (Figure 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 (Figure 1b). 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 and the use of a 100-kDa cut-off membrane device, UF/DF was not able to remove smaller contaminating proteins, and an AC resin bearing an streptavidin mutein ligand was used to obtain the protein in high purity (Figure 1c). The affinity resin has shown a relatively high dynamic binding capacity for the S protein (Figure 1d), and has so far been used for 30+ adsorption/elution/regeneration cycles with no detectable decrease in performance, reducing its impact on the final costs of the purified protein. ![Figure 1:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2020/07/15/2020.07.13.20152884.1/F1.medium.gif) [Figure 1:](http://medrxiv.org/content/early/2020/07/15/2020.07.13.20152884.1/F1) Figure 1: S protein production by stable recombinant HEK293 cells, and its use to establish an ELISA. **(a)** Left panel: CHO-K1 and HEK293 were transiently transfected with plasmid DNA containing the S protein gene. Presence of S protein in the spot blots was measured using serum of SARS-CoV-2 convalescent patients (1:1000) as primary antibody, followed by incubation with anti-human-HRP. Center panel: due to the higher levels of protein secreted by HEK293 cells, transient transfections at high (A, 2 μg/mL of pαH plasmid) and low (B, 0.75 μg/mL of pαH plasmid) DNA concentration were carried out with this cell line, along with a stable transfection enabled by co-transfecting 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 so far been confirmed for up to 100 days post-transfection. **(b)** Left panel: cultivation of the stable cell line in shake flasks and 1.5-L stirred-tank bioreactors, in batch and fed-batch mode, has shown that high titers of secreted S protein can be achieved in the cell culture supernatant; cell viability and viable cell density (VCD) are shown. Right panel: spot blot for detection of S protein in the supernatants on different days of culture. **(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: 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 cell line cultured in chemically-defined, animal component-free medium. **(d)** A typical chromatogram of the affinity chromatography purification. Fractions of the flow-through 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. **(e)** ELISA performance using low-purity (UF/DF) or high-purity (AC) S protein antigen to coat plates, 16 convalescent sera samples from COVID-19 patients (SARS-CoV-2 samples) were tested in four dilutions (1:40, 1:120, 1:360 and 1:1080). Two sera samples pre-COVID-19, and known positive and negative samples by PCR and rapid diagnostic test (RDT) were included as controls. **(f)** Titration of positive and negative samples by the ELISA. An ELISA was then established for the detection of anti-S antibodies in serum, plasma and whole blood eluates (S-UFRJ ELISA). A comparison of the UF/DF concentrated S protein to the high-purity AC sample showed that assay performance can be greatly improved by the use of the highly purified antigen (Fig. 1e). Using serum samples from negative controls and COVID-19 PCR-confirmed patients (see methods), we established that 150 ng of high-purity S protein per well enables clear discrimination of samples (Fig. 1f). Next we applied S-UFRJ ELISA to evaluate the presence of anti-S IgG in 210 serum samples, including negative controls and positive samples obtained from symptomatic individuals diagnosed by PCR as positive for the presence of SARS-COV-2 genetic material. The PCR-positive cohort consisted of 38 symptomatic individuals whose blood samples were obtained at different time points after symptoms onset, comprising a total of 66 samples. Control groups were plasma samples collected until 2018 from healthy individuals (pre-COVID-19, n=124) or collected in 2020 from individuals who tested negative for SARS-COV-2 by PCR (post-COVID-19, n=20). The assay exhibited high specificity as 122 out of 124 of pre-COVID-19 controls scored as negative, below the cut-off (98.39 % specificity) (Fig 2a). Considering all samples from PCR+ individuals, regardless of the time of collection, 53 out of 66 samples (80.30 % sensitivity) were IgG positive. For comparison, all samples were also tested for anti-SARS-COV-2 IgG using an imported rapid diagnostic test (RDT) that has been approved by the Brazilian regulatory agency (ANVISA) for commercialization in the country. Using this commercial RDT, only 30 out of 66 samples were positive for IgG (45.45% sensitivity) (Fig 2b). ![Figure 2:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2020/07/15/2020.07.13.20152884.1/F2.medium.gif) [Figure 2:](http://medrxiv.org/content/early/2020/07/15/2020.07.13.20152884.1/F2) Figure 2: Validation of S-COVID19-UFRJ ELISA for early detection and quantification of anti-S IgG serum antibodies, and optimization of sample collection amenable for field studies. **(a)** Anti-S IgG antibody detection in plasma samples from healthy individuals, obtained pre-COVID-19 (n=124) or post-COVID-19 who tested negative for the virus by qPCR (n=20), and COVID-19 patients who were qPCR positive (n=66). 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. of the negative controls in the same ELISA plate. For routine use of the S-UFRJ ELISA, sera samples were diluted at 1:40 and the cut-off (C.O.) was defined as in commercial ELISA tests: an O.D. ratio below 1 indicates a negative result (N), an O.D. ratio above 2 indicates a positive result (P), and an O.D. ratio between 1 and 2 is considered undetermined. (U). **(b)** Plasma samples from PCR positive individuals were tested for IgG anti-SARS-COV-2 by a commercial rapid immunochromatographic test (MedTeste, Medlevensohn, Brazil). Samples were then grouped by IgG reactivity according to rapid diagnostic tests (RDT) and their levels of IgG anti-S on S-UFRJ ELISA are plotted. **(c)** Levels of IgG anti-S on S-UFRJ ELISA in individuals that scored negative for IgG rapid test grouped as IgM- and IgM+, also according to their rapid-test. **(d)** Levels of IgG anti-S in serum samples grouped according to collection on different times after symptoms onset (DASO). **(e)** Positive rates of IgG anti-S on S-UFRJ ELISA versus DASO, the identification of the IgM and IgG rapid tests, are indicated in the figure legend as S-UFRJ IgG, RT IgM and RT IgG. **(f)** Titration of serum samples of COVID-19 convalescent patients (SARS-CoV-2 samples), the highest and lowest endpoint titer samples are indicated in red and white, 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 indicated as 1. Statistical analysis was performed using Pearson’s test. **(h)** Dried blood samples (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 commercially available paper hole punching device were used to make DBS (arrowhead) from which blood was eluted for ELISA testing. **(i)** S-UFRJ ELISA comparing the O.D. values in increasing dilutions of sera and eluates. **(j)** O.D. summation of the data from the different dilutions (same experiment as in **i**). **(k)** dried blood samples collected in plastic strips containing 1, 2 or 3 pads of filter paper. **(l-n)** correlations of O.D. ratios between samples collected in the strip pad 1 and respective serum sample (**l**), respective DBS in filter paper (**m**), or pad 2 of the same strip (**n**). Serum dilution was 1:40. DBS in filter paper circles were eluted in 100 μL PBS 1% BSA. DBS in single pads were eluted in 200 μL PBS 1% BSA. 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 IgM rapid test. We found that most samples that scored negative for anti-S IgG in our ELISA, also scored as negative for IgM in the rapid test (Fig 2c). On the other hand, the majority of samples that were IgG positive in the S-UFRJ ELISA were also positive for IgM in the rapid test (Fig 2c). This result suggests that samples from PCR-positive 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, and for that reason scored negative both for IgM in the rapid test and for IgG in our S-based ELISA. Indeed, when the results of S-UFRJ ELISA were charted against DASO, this was found to be the case: samples were increasingly scored as positive for anti-S IgG according to the time point they had been collected after symptoms onset (Fig 2d). Accordingly, PCR-positive individuals that have been sampled on different DASO and scored negative in their first sampling, converted to seropositive for anti-S IgG afterwards (data not shown). Anti-S IgG seroconversion rate, detected by S-UFRJ ELISA, increases progressively from 41.66 % to 100 % as a function of DASO, being above 90 % already from day 10 after symptoms onset on (fig 2e). Of note, S-UFRJ ELISA detects anti-S IgG seroconversion much earlier than the rapid test used here for comparison, which reached a maximum positive rate as low as 71.43% even for DASO of 20 days and above. We further tested serum samples of COVID-19 convalescent patients for their SARS-COV-2 in vitro neutralization ability as measured by plaque reduction neutralization test (PRNT). Importantly, analogous to what has been shown by Amanat et al. [2], we found that sera displaying high anti-S IgG ELISA titers (Fig 2f) also presented high neutralization titers (Fig 2g), resulting in a very high correlation between those titers (Pearson’s R=0.9143, p<0.00001). Low-cost serological assays for COVID-19 are urgently needed in order to enable public health actions, especially in low-income countries. The ELISA developed herein meets this requirement: the cost of the assay in terms of consumables was calculated to be in the range of two dimes per sample. However, considering the use in epidemiological surveillance studies, especially in low-income countries, traditional blood sample collection and serum separation would imply the need for clinical laboratory services and refrigerated sample storage and transport that could jeopardize the advantages of the ELISA test. To overcome this critical limitation, we devised a simple storage and transport of blood samples collected by finger prick in filter paper strips to allow sampling in remote regions, or regions lacking a laboratory network. For that, the assay was further standardized to work using eluates from whole blood collected in filter paper (Fig 2h), a classic protocol that permits titration of antigen immunoreactivity [5]. Plastic strips with filter pads could be even more convenient for sampling and storing for retesting (Fig 2k). A first comparison of titration curves using serum samples or dried blood spots (DBS) in filter paper displayed similar results (Figs 2i-2j). O.D. ratios obtained for serum samples, and DBS either in filter paper or in pads showed very high correlation (Figs 2l-2m), as well as high reproducibility among pads from a given strip (Fig 2n). By using dried blood spots, the low cost of the test is warranted. On top of the two dimes related to consumables costs per test, labor, equipment and logistics costs must be taken into consideration. These vary significantly from country to country, but we estimate that the final cost including all steps from sample collection to test result does not exceed USD 0.50 per test. Compared to USD 100.00 tests available to the customer in the US, the S-UFRJ ELISA is approximately 200-fold less expensive. Importantly, blood samples collected in filter paper and kept in sealed plastic bags can be preserved for at least 2 months without altering their serological result (data not shown). As such, the S-UFRJ ELISA, comprising the use of eluates from whole blood finger pricks as samples, allows broad serological surveillance in populations regardless of their geographical and socio-economic aspects. As recently shown for the case of Spain [6], population-based sero-epidemiological studies are essential tools to inform authorities if specific public health measures need to be maintained to avoid new epidemic waves, thus enabling scientifically driven decision making by governments. ## Data Availability The raw data of all experiments are available upon request. ## ONLINE: MATERIALS AND 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 transfected by lipofection using a broad-spectrum reagent (Lipofectamine 3000, Thermo Fisher) as described previously (Alvim et al., 2019). 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 conformation (Wrapp et al., 2020; 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 after 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 the 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 setpoins of pH, temperature and dissolved oxygen of 7.0, 37°C and 40% of air saturation, respectively. Cell concentration and viability was 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 automatically 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 UF/DF or affinity chromatography 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 finally chemiluminescent ECL reagent (BioRad). ### S-UFRJ ELISA of IgG anti-SARS-CoV-2 S protein High binding ELISA plates (Corning) were coated with 50 µL of SARS-CoV-2 S protein at 4 µg/mL in PBS (Gibco) 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, circles cut from filter paper using commercially available punching devices or single pads cut from plastic strips were used to prepare eluates by incubating for 1 hour at RT in 100 µL or 200 µL of PBS 1% BSA, respectively. 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). ### Sample collection from human subjects According to a protocol approved by the national ethics committee (CONEP, Brazil; protocol #30161620000005257; review #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 sera samples, serum samples 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%2F07%2F15%2F2020.07.13.20152884.1.atom)) for 1 hour at 37°C to enable neutralization to occur. Virus-serum mixture was inoculated into confluent monolayers of VERO cells seeded at 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. ## 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, and the team of volunteers working in the UFRJ COVID-19 screening center. This work was supported by Senai CETIQT, Senai DN and CTG, and by the Brazilian research funding agencies Fundação de Amparo a 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 * Received July 13, 2020. * Revision received July 15, 2020. * Accepted July 15, 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.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. Preprint at [https://doi.org/10.1101/2020.04.24.057323](https://doi.org/10.1101/2020.04.24.057323) (2020). 2. 2.Amanat, F., Stadlbauer, D., Strohmeier, S. et al. A serological assay to detect SARS-CoV-2 seroconversion in humans. Nat Med., [https://doi.org/10.1038/s41591-020-0913-5](https://doi.org/10.1038/s41591-020-0913-5) (2020). 3. 3.Wrapp, D., Wang, N., Corbett, K.S., et al. Cryo-EM structure of the 2019-nCoV spike in the prefusion conformation. Science 367, 1260–1263 (2020). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Mzoic2NpIjtzOjU6InJlc2lkIjtzOjEzOiIzNjcvNjQ4My8xMjYwIjtzOjQ6ImF0b20iO3M6NTI6Ii9tZWRyeGl2L2Vhcmx5LzIwMjAvMDcvMTUvMjAyMC4wNy4xMy4yMDE1Mjg4NC4xLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 4. 4.Alvim, R.G.F., Itabaiana Jr., I., Castilho, L.R. Zika virus-like particles (VLPs): stable cell lines and continuous perfusion processes as a new potential vaccine manufacturing platform. Vaccine 37, 6970–6977 (2019). 5. 5.Lim, M.D. Dried blood spots for global health diagnostics and surveillance: opportunities and challenges. Am J Trop Med Hyg., 99:256-265 (2018). 6. 6.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. The Lancet, [https://doi.org/10.1016/S0140-6736(20)31483-5](https://doi.org/10.1016/S0140-6736(20)31483-5) (2020).