A Simple RT-PCR Melting temperature Assay to Rapidly Screen for Widely Circulating SARS-CoV-2 Variants ====================================================================================================== * Padmapriya Banada * Raquel Green * Sukalyani Banik * Abby Chopoorian * Deanna Streck * Robert Jones * Soumitesh Chakravorty * David Alland ## Abstract **Background** The emergence of more transmissible SARS-CoV-2 variants in the United Kingdom (B.1.1.7), South Africa (B1.351) and Brazil (P.1) requires a vigorous public health response, including real time strain surveillance on a global scale. Although new SARS-CoV-2 variants can be most accurately identified by genomic sequencing, this approach is time consuming and expensive. A simple and more rapid screen for the key SARS-CoV-2 mutations that define variant strains is needed. We developed a simple, rapid and high-throughput reverse-transcriptase PCR (RT-PCR) melting temperature assay that identifies the SARS-CoV-2 N501Y mutation, a key mutation which is present in all three known variant strains of concern. **Methods** RT-PCR primers and two sloppy molecular beacon (SMB) probes were designed to amplify and detect the SARS-CoV-2 N501Y (A23063T) mutation. One SMB was designed with a probe region that was complementary to the wild type sequence (WT) and a second SMB was designed with a probe region that was complementary to the mutant (MT) sequence. Each SMB was labeled with a different fluorophore, and the assay was performed in a single test well. After RT-PCR, WT versus MT SARS-CoV-2 was identified by a characteristic shift in the melting temperature (Tm) of each SMB. Assay optimization and testing was performed with RNA from SARS-CoV-2 USA WA1/2020 (WT) and SARS-CoV-2 hCoV-19/England/204820464/2020 (a B.1.17 variant). The assay was then validated using clinical samples. **Results** The limit of detection (LOD) of the assay for both the WT and the B1.1.7 variant was 4 genomic copies/reaction. The two SMBs produced Tm shifts that were 100% sensitive and 100% specific for identifying the N501Y mutation in cultured virus and in clinical samples as confirmed by Sanger sequencing. **Conclusion** We have developed a rapid screening test for the SARS-CoV-2 N501Y mutation, which is a characteristic of all three SARS-CoV-2 stains of global concern. This assay can be used to rapidly screen large numbers of patient samples for these variants, providing an early warning for the emergence and spread of these strains of concern. Key words * SARS-CoV-2 * Variants * N501Y * melting temperature (Tm) * screening test ## Introduction In December 2020, public health officials in the United Kingdom observed a surge of COVID-19 cases in Kent, England that appeared to be largely due to a specific variant of SARS-CoV-2 (1, 2). The new variant was named Variant of Concern (VOC 202012/1) or B.1.1.7 based on its phylogenetic lineage. Early reports suggested that B.1.1.7 was more transmissible and possibly more virulent than previous SARS-CoV-2 strains (3, 4), although the evidence did not suggest that B.1.1.7 caused a decrease in vaccine efficacy (5–8). The B.1.1.7 variant has a number of mutations in the spike protein including single nucleotide polymorphisms (SNPs) resulting in N501Y, A570D, D614G and P681H mutations, and deletions at amino acids 69-70 and 144Y (6, 9). The N501Y (A23063T) mutation has been identified an important contributor to the worrisome phenotype (2, 4, 10, 11). Other SARS-CoV-2 strain variants have been implicated in large outbreaks in South Africa (known as 20H/501Y.V2 or B.1.351 lineage) and Brazil (20J/501Y.V3 or P.1 lineage) (12). These variants have appeared to decrease the efficiency of some COVID-19 vaccines (2, 13). All three, B.1.1.7, B.1.352 and P.1 variants contain the N501Y mutation, while the South African and Brazilian variants additionally contain mutations in E484K and K417N (1, 12). Thus, the N501Y mutation appears to be an excellent marker for all three strains while the E484K mutation can be used to differentiate the other two strains from B.1.1.7. New SARS-CoV-2 variants have also been recently reported in the United States (1, 14) with evidence for convergent evolution at a Q667P mutation (14); however, the epidemiological and clinical importance of strains containing the Q667P mutation is not currently known. All three of the major N501Y SARS-CoV-2 variants are circulating around the world (1). However, the distribution, extent, and spread of these variants is poorly understood in countries such as the United States where only a small fraction (0.04 to 3.5%) of COVID-19 cases are analyzed by viral genomic sequencing as per the CDC’s national genomic surveillance dashboard (15). Genome sequencing is currently the only method available to reliably detect rapidly emerging SARS-CoV-2 variants. Viral genomic sequencing has the advantage of providing a detailed map of new mutations, which supports new variant discovery as well as variant monitoring. However, genomic sequencing is expensive and difficult to perform in real time. In contrast, RT-PCR testing for SARS-CoV-2 has become widespread and is easy to perform in a high throughput manner when necessary and relatively rapid. However, routine RT-PCR tests do not differentiate among SARS-CoV-2 variants, or do so only by producing negative assay results (16), which still require sequence confirmation (17). Also, the potential for additional mutations to appear near key variant-defining alleles may complicate the development of RT-PCR assays for SARS-CoV-2 variants (18). We have previously demonstrated that sloppy molecular beacons (SMBs) combined with melting temperature (Tm) code analysis may be used to specifically detect distinct mutations in short genomic regions where a variety of mutations can exist (19). Here, we apply this same Tm-based approach to detect and differentiate variant strains of SARS-CoV-2 with high sensitivity and specificity. This approach is flexible and can be used in high throughput manner, easily allowing the addition of new mutation detecting assays as needed to identify and track SARS-CoV-2 variants as they emerge. Furthermore, this approach can be performed on a wide range of real-time PCR instrumentation as long as they have the capacity to run melt curve analysis. The wide availability of such instruments can allow quick adoption of this assay around the world, increasing access to real-time monitoring of SARS-CoV-2 variant spread. ## Methods ### Ethical considerations The use of de-identified clinical samples from confirmed COVID-19 positive and negative patients for PCR testing and sequencing was approved by the Rutgers Institutional Review Board under protocol numbers 20170001218 and 2020001541. ### Viral cultures and RNA Genomic RNA from SARS-CoV-2 USA WA1/2020 (wild type, WT) and viral culture stocks of SARS-CoV-2 hCoV-19/England/204820464/2020 (B.1.1.7 variant, MT) were obtained from BEI Resources, NIAID (Manassas, VA). RNA was isolated from the B.1.17 variant in a BSL3 laboratory, using RNAdvance viral RNA extraction kit (Beckman Coulter, Indianapolis, IN). ### Genome sequence analysis for assay design For initial analysis of the mutations in SARS-Cov-2, a total of 330,132 high quality viral genome sequences deposited in GISAID (20) as of Jan 12, 2021, were analyzed. Publicly available datasets were analyzed in this study. This data can be found here: [https://www.gisaid.org/](https://www.gisaid.org/). A 250-nucleotide region around the N501Y (A23063T) position in the reference strain (GenBank accession number [MN908947](http://medrxiv.org/lookup/external-ref?link_type=GEN&access_num=MN908947&atom=%2Fmedrxiv%2Fearly%2F2021%2F03%2F08%2F2021.03.05.21252709.atom)) was selected and used to identify the corresponding regions in the GISAID dataset using BLAST (21). These matching sequences were condensed into a set of unique sequences and aligned using a multiple sequence alignment program, MAFFT (22). Candidate amplification primers and probes were identified on the basis of sequence conservation and predicted Tm using the algorithm of SantaLucia (23) and final set of primers were designed with the help of the Primer3 program (24). SMB probe design was performed using the web servers DNA mfold ([http://www.unafold.org/mfold/applications/dna-folding-form.php](http://www.unafold.org/mfold/applications/dna-folding-form.php)) and DINAmelt ([http://www.unafold.org/hybrid2.php](http://www.unafold.org/hybrid2.php)) to predict the probe folding structures and probe-target hybrid Tm values respectively. ### Primers and Probes An 89 bp region surrounding the 23063 position was amplified using the primer set N501Y-F: 5’ggttttaattgttactttcctttacaa3’ and N501Y-R: 5’gaaagtactactactctgtatggttgg3’ at 1:10 ratio in an asymmetric PCR. Sloppy molecular beacons (SMB) were designed targeting both the wild type 501N (23063A) sequence (SMB-501-WT): Quasar570-5’ccgcGTT[pdU]CCAtCCCACTAATGcTG[pdU]TGGTTACCAACgcgg3’-BHQ-2 and the mutant 501Y (23063T) sequence (SMB-501-MT) : Quasar 670-5’cgcgGTT[pdU]CCAtCCCACTTATGcTG[pdU]TGGTTACCAACcgcg3’-BHQ-2, where the upper case letter indicate the SMB probe region and the lower case letter indicate the SMB stem region and pdU indicates C5 Propynyl-deoxyuridine and BHQ indicates “black hole quencher”. Primers were obtained from Millipore Sigma (The Woodlands, TX) and SMBs were synthesized by LGC Biosearch technologies (Petaluma, CA). An internal control (IC) assay developed by CDC (25, 26), targeting the human RNaseP gene was simultaneously performed for each extracted RNA specimen as a separate reaction in a separate well, using the *Taq*Man real-time PCR assay probe tagged with FAM at the 5’ end and Dabcyl quencher at the 3’ end. ### SMB-501 assay formulation and procedure TaqPath™ 1-Step RT-qPCR Master Mix, CG (ThermoFisher Scientific, Waltham, MA) was used for the RT-PCR. Each one step reaction mix was supplemented with 0.2µM of the forward primer N501Y-F and 2 µM of reverse primer N501Y-R, 0.4 µM of each of the SMB probes (SMB-501-WT and SMB-501-MT) and 1 µl of the template RNA. The internal control contained primers and probe specific for human RNAseP as described previously (25, 26). Each reaction was run in replicates of 4 in 384-well plates in Roche LightCycler 480 (Roche, Indianapolis, IN). The one-step RT-qPCR amplification was performed with the following thermocycling conditions: Uracil DNA glucosylase incubation for 2 min at 37°C and reverse transcription (RT) for 15 min at 50°C, followed by asymmetric PCR for 45 cycles (denaturation at 95 °C for 1 s, annealing/extension at 55°C for 30 s. The post-PCR melt was performed with the following conditions: denaturation at 95°C for 30s followed by cooling down to 45°C and gradual heating to 85°C, with continuous monitoring of fluorescence at the rate of 2 acquisitions per °C. The total assay time was 1h 17min. Automated Tm calls were performed by the LC480 Tm detection software at the end of the PCR. The resulting Tm for each probe was identified and matched with the Tm-signature code defined for the wildtype or the mutant variants. ### Analytical sensitivity The pre-quantitated genomic RNA from the SARS-CoV-2 USA WA1/2020 (WT) obtained from BEI resources with the stock concentration of 1.8 ×104 genomic equivalents (GE)/µl was diluted in Tris-EDTA (TE) buffer. A 1 µl of each concentration (400, 200, 40, 20 and 4 GE/µl) was added to the one-step RT-PCR mix containing the primers and probes and was evaluated in the SMB-501 assay. The RNA extracted from the variant strain was quantified against a standard curve generated with the N1 gene-specific real time RT-PCR assay (25, 26) and the concentration was determined to be 4×105 copies/µl. The mutant RNA was also serially diluted and a 1 µl of each concentration as mentioned before was evaluated in SMB-501 assay. ### Validation with patient samples Deidentified nasopharyngeal (NP) swabs obtained from patients undergoing routine clinical testing using the Xpert Xpress SARS-CoV-2 or Xpert Xpress SARS-CoV-2/Flu/RSV (4-plex) test (Cepheid, Sunnyvale CA) assay in the CLIA and CAP certified laboratory at the Public Health Research Institute (PHR), Newark, NJ, for COVID-19, were selected for this study. NP swabs were collected in 3ml viral transport media (VTM) from Hardy diagnostics (Santa Maria, CA) or Labscoop (Little Rock, AR) and were banked at either refrigerated conditions (for specimens first tested within the previous 2 weeks) or frozen (for specimens stored longer than 2 weeks). We randomly selected 20 COVID-19 positive samples of unknown genotype and a RT-PCR cycle threshold (Ct) <40 with either Xpert Xpress SARS-CoV-2 or Xpert Xpert CoV-2/Flu/RSV test, with 6 having been banked between October 2020 through December 31, 2020 and 14 having been banked from January 1, 2021 – February 2021. Thirty COVID-19 negative specimens were randomly selected from samples banked between Oct 2020 through Feb 2021. RNA was extracted from each specimen using a QiaAmp viral RNA isolation kit, following the manufacturer’s instructions (Qiagen, Valencia, CA) under BSL2 conditions. A 5µl volume of RNA was added to the one-step RT-PCR mix containing the primers and probes. All samples were tested by the SMB-501 and IC assays in two separate wells in replicates of two. The samples that failed the IC assay were repeated starting with the RNA extraction. A subset of samples that tested positive either for 501N wildtype or 501Y mutant was confirmed by Sanger sequencing using the primer pair: F-5’ctatcaggccggtagcacac3’ and R-5’ctttcttttgaacttctacatg3’ which amplifies a 143bp segment of the S-gene inclusive of the amino acid positions at 484 and 501. The sequencing chromatograms and the sequences were analyzed using Ugene (ver 37) comparing against the known WT and MT sequences using MegAlign Pro software (DNAStar, ver16). ### Statistical analysis Standard statistical analyses (average, standard deviation) and graphing were performed using Microsoft excel (ver 2102) and GraphPad Prism 8.4.3 for Windows. ## Results ### Limit of detection The one-step RT mix containing asymmetric PCR primers and both wildtype and MT specific probes was added with either WT RNA or MT RNA at different concentrations ranging from 400 to 4 GE/reaction (N=4). After Tm analysis was performed the Tm peak heights were highest in the assays with the largest added number of GE and the peak heights progressively decreased as the number of GEs present in the reactions decreased. However, Tm peak heights produced by both SMB 501-WT and SMB 501-MT) could still be reproducibly detected at the lowest concentration tested (4 GE/reaction) when tested against both WT and N501Y mutant strains, defining the assay limit of detection as <4 GE per reaction (Fig. 1). ![Fig. 1.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2021/03/08/2021.03.05.21252709/F1.medium.gif) [Fig. 1.](http://medrxiv.org/content/early/2021/03/08/2021.03.05.21252709/F1) Fig. 1. Analytical limit of detection and Tm values generated by the SMB-501-WT (WT) and SMB501-MT (MT) probes tested against SARS-CoV-2 RNA. **A** The SMB-501-WT probe was tested in the presence of SARS-CoV-2 wildtype (WT) and B.1.1.7 mutant (MT) RNA at the indicated number of genomic equivalents (GEs). **B** The SMB-501-MT probe was tested in the presence of SARS-CoV-2 wildtype (WT) and B.1.1.7 mutant (MT) RNA at the indicated number of GEs. The effect of target concentration on the melt peak height is shown for WT (C) and MT (D). ![Fig. 2.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2021/03/08/2021.03.05.21252709/F2.medium.gif) [Fig. 2.](http://medrxiv.org/content/early/2021/03/08/2021.03.05.21252709/F2) Fig. 2. Sloppy molecular beacon (SMB) Tm profile samples tested using the SMB-501-Tm assay. Ref WT indicates the Tm profile of the reference WT SARS-CoV-2 strain and Ref MT indicates the TM profile of the reference MT SARS-CoV-2 B.1.1.7 strain. P1 – P20 indicate that Tm profiles of the 20 clinical samples tested in this study. Error bars show +/- one standard deviation. ### Tm code definition The Tm values produced by both SMBs against the reference WT and the MT SARS-CoV2 strains are listed in Table 1. The mean and standard deviations shown are derived from at least 4 replicates. Using these reference RNA samples, when RNA was tested with the SMB 501-WT probe, WT-RNA produced a Tm of 59.8°C±0.4 and MT N501Y RNA produced a Tm of 55.2°C±0.4. Similarly, when RNA was tested with the SMB 501-MT probe, WT-RNA produced a Tm of 58.2°C±1, and MT N501Y RNA produced a Tm of 62.25°C±0.6. Given that Tm values can vary slightly between clinical samples, we specified two-temperature Tm codes that identified either 501N or 501Y alleles with a Tm range approximately 1.5-4 times the experimentally verified ±SD values for each probe Tm. Thus, the 501N (WT) Tm code was defined as a SMB-501-WT Tm of 59.8 ±1.5 °C and a SMB-501-MT Tm of 58.2 ±1.5°C; and a 501Y (MT) Tm code was defined as a SMB-501-WT Tm of 55.2 ±1.5 °C and a SMB-501-MT Tm of 62.25 ±1.5°C. Any samples that failed to produce a Tm value for either of the SMBs or produced Tm values outside of the range defined for the Tm codes would have been defined as indeterminant and the assay repeated. Using this code definition, we retested our two reference RNA samples 20 times, and the correct 501 allele was corrected in each case, achieving an analytic sensitivity and specificity of 100%. These results clearly indicate that the combination of both SMB 501-WT and SMB 501-MT probes can specifically detect and differentiate the N501Y variants from the wild type strains with high confidence. View this table: [Table 1.](http://medrxiv.org/content/early/2021/03/08/2021.03.05.21252709/T1) Table 1. Validation of SMB-501-assay against COVID positive clinical samples. ### Validation with patient samples A total of 50 patient samples, 20 confirmed COVID-19 positives and 30 confirmed COVID-19 negatives were tested using the SMB N501Y assay. None of the COVID-19 negative samples produced any measurable Tm values yielding a specificity of 100%. Positive samples were selected based on a wide range of N2-Ct values from 18.1 through 39 from the Xpert Xpress SARS-CoV-2 assay. As shown in Table 1, all COVID-19 positive samples produced measurable Tm values for both SMB probes in at least one of the two replicates. Although, samples with Xpert Ct values as late as 39 could be detected by our assay, we found that the four samples with a Ct >36 (Pt10-Pt13) using the Xpert assay or ≥40 Ct by the SMB-501 assay, yielded relatively stunted melt peak heights of ≤0.3 with the WT-SMB and ≤0.1 with MT-SMB similar to that observed when testing 4 GE in our limit of detection studies, which is indicative of very low viral loads in these samples. However, the Tm could still be identified for both SMB probes in all samples. Overall, 12/20 of the COVID-19 positive samples had SMB 501-WT and SMB 501-MT Tm values consistent with the WT N501 allele and 8/20 samples had Tm values consistent with mutant Y501 allele. Ten representative WT and mutant samples as predicted by the SMB assay, underwent Sanger sequencing of the PCR products for confirmation of the PCR results. In all cases, the WT or mutant sequences identified by the SMB N501Y assay were confirmed by the sequencing result. Using the confirmed sequencing results as a gold standard, 4/4 of the wild type clinical samples were detected as WT by the SMB N501Y assay, and 6/6 N501Y mutant results were detected as mutant by the assay, demonstrating a clinical sensitivity and specificity of 100%. It should also be noted that of the 6 mutants samples tested by Sanger sequencing, the sequencing study showed that one of the samples (Pt12) also had mutation at 484th amino acid position along with N501Y. Thus, the combination of our SMB 501 assay and Sanger sequencing of the mutant samples suggests that −5/6 of the mutant samples were of the B.1.1.7 lineage and the remaining mutant sample was either of the B.1.352 or P.1 lineages. ## Discussion The emergence of SARS-CoV-2 variants with the potential for increased transmission, disease severity, and resistance to vaccine induced immunity is of grave concern (27). A simple screening assay to monitor the emergence and spread of these strains may be helpful for implementing public health strategies to counter these and future strains. Our study demonstrates that our assay is simple, rapid, and sensitive and specific for detecting a key variant-identifying mutation using a high-throughput PCR assay platform. Thus, this assay has the potential for relatively inexpensive high throughput testing for rapid identification of N501Y variants. In designing this assay, we took advantage of the fact that the N501Y mutation is common to the three major SARS-CoV-2 variants of concern. Since this mutation also appears to be responsible for the increased infectivity and possibly the other adverse manifestations of these strains (4), assays which detect this mutation may also prove useful to detect any future strain that evolves to have increased transmission potential. Our assay is the first to our knowledge that uses post PCR Tm based analysis to detect and differentiate SARS-CoV-2 variants using SMB probes demonstrated in clinical samples. This assay format has the benefit of producing a measurable Tm result irrespective of whether the SMB probe is fully complementary to its target nucleic acid sequence. Instead of detecting a mutation by either producing or not producing a signal, SMBs detect mutations by producing a Tm shift. Failure to produce a Tm signal indicates an invalid assay rather than the absence of a mutation. The robustness of our assay is further increased by our use of two different SMBs, one complementary to the WT sequence and one complementary to the mutant sequence. The pattern of Tm values or “Tm signature” produced by the combined Tm values of each SMB probe provides an unequivocal identification of a WT or MT sequence. We have also shown that Tm signatures can be used to detect mixtures of mutant and WT sequences and to identify numerous mutations present in an assay’s target region (29). Thus, we expect that our assay should continue to be able to identify N501Y variants even if additional mutations develop near this primary mutation once the specific Tm signatures of each new genotype are characterized. Our assay is meant to be a screening assay which will identify samples likely to contain SARS-CoV-2 variants of concern. We suggest that the most useful public health strategy would be to also perform subsequent genomic sequencing on a subset of screen-positive samples. This sequencing would confirm the presence of the expected variants and in some cases lead to the discovery of new variants. Ongoing sequencing of a subset of all SARS-CoV-2 samples will also be required to identify completely novel variants or to investigate the epidemiology and clinical characteristics of variants such as those recently reported in the United States. Fortunately, our assay is easily extensible and additional tests for new key mutations can be added in a modular format to a screening panel when new mutations associated with critical new variants are discovered. In fact, it is our intention to continuously update this assay until our work is superseded by a better approach or much more widespread genome sequencing becomes commonplace. Key updates will also be posted on a preprint journal. In the meantime, we hope that our current test will help increase surveillance and potentially help control the spread of the new emerging variants of concern. ## Supporting information Supplemental Table 1S [[supplements/252709_file03.xlsx]](pending:yes) ## Data Availability The assay published here along with the primers and probes are made available freely for the readers ## Acknowledgements This study was funded by the National Institute of Allergy and Infectious Diseases of the National Institutes of Health under award number R01 AI131617. The following reagent was deposited by the Centers for Disease Control and Prevention and obtained through BEI Resources, NIAID, NIH: Genomic RNA from SARS-Related Coronavirus 2, Isolate USA-WA1/2020, NR-52285.The following reagent was obtained through BEI Resources, NIAID, NIH: Severe Acute Respiratory Syndrome-Related Coronavirus 2, Isolate hCoV-19/England/204820464/20200, NR-54000, contributed by Bassam Hallis. The authors wish to thank all the laboratories that contribute sequence data to the GISAID EpiCoV database. A GISAID acknowledgment table reporting the geographic origin and contributions of genomes analyzed in this study is attached as a supplementary Table 1S. D.A. receives research support and royalty payments from Cepheid, which sells the Xpert Xpress SARS-CoV-2 and Xpert Xpress SARS-CoV-2/Flu/RSV tests., S.C, is an employee of Cepheid and R.J. is a paid consultant for Cepheid. * Received March 5, 2021. * Revision received March 5, 2021. * Accepted March 8, 2021. * © 2021, Posted by Cold Spring Harbor Laboratory This pre-print is available under a Creative Commons License (Attribution-NoDerivs 4.0 International), CC BY-ND 4.0, as described at [http://creativecommons.org/licenses/by-nd/4.0/](http://creativecommons.org/licenses/by-nd/4.0/) ## References 1. 1.Galloway SE, Paul P, MacCannell DR, Johansson MA, Brooks JT, MacNeil A, Slayton RB, Tong S, Silk BJ, Armstrong GL, Biggerstaff M, Dugan VG. 2021. Emergence of SARS-CoV-2 B.1.1.7 Lineage - United States, December 29, 2020-January 12, 2021. MMWR Morb Mortal Wkly Rep 70:95–99. 2. 2.Lauring AS, Hodcroft EB. 2021. Genetic Variants of SARS-CoV-2—What Do They Mean? JAMA 325:529–531. 3. 3.Lacobucci G. 2021. Covid-19: New UK variant may be linked to increased death rate, early data indicate. BMJ 372:230. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1136/bmj.n230&link_type=DOI) 4. 4.Santos JC, Passos GA. 2021. The high infectivity of SARS-CoV-2 B.1.1.7 is associated with increased interaction force between Spike-ACE2 caused by the viral N501Y mutation. bioRxiv doi:10.1101/2020.12.29.424708:2020.12.29.424708. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoiYmlvcnhpdiI7czo1OiJyZXNpZCI7czoxOToiMjAyMC4xMi4yOS40MjQ3MDh2MSI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIxLzAzLzA4LzIwMjEuMDMuMDUuMjEyNTI3MDkuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 5. 5.Callaway E, Mallapaty S. 2021. Novavax offers first evidence that COVID vaccines protect people against variants. Nature 590:17. 6. 6.CDC. 2021. Emerging SARS-CoV-2 Variants. [https://www.cdc.gov/coronavirus/2019-ncov/more/science-and-research/scientific-brief-emerging-variants.html](https://www.cdc.gov/coronavirus/2019-ncov/more/science-and-research/scientific-brief-emerging-variants.html). Accessed 7. 7.Greaney AJ, Loes AN, Crawford KHD, Starr TN, Malone KD, Chu HY, Bloom JD. 2021. Comprehensive mapping of mutations to the SARS-CoV-2 receptor-binding domain that affect recognition by polyclonal human serum antibodies. bioRxiv doi:10.1101/2020.12.31.425021:2020.12.31.425021. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoiYmlvcnhpdiI7czo1OiJyZXNpZCI7czoxOToiMjAyMC4xMi4zMS40MjUwMjF2MSI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIxLzAzLzA4LzIwMjEuMDMuMDUuMjEyNTI3MDkuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 8. 8.Wu K, Werner AP, Moliva JI, Koch M, Choi A, Stewart-Jones Gbe, Bennett H, Boyoglu-Barnum S, Shi W, Graham BS, Carfi A, Corbett KS, Seder RA, Edwards DK. 2021. mRNA-1273 vaccine induces neutralizing antibodies against spike mutants from global SARS-CoV-2 variants. bioRxiv doi:10.1101/2021.01.25.427948. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoiYmlvcnhpdiI7czo1OiJyZXNpZCI7czoxOToiMjAyMS4wMS4yNS40Mjc5NDh2MSI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIxLzAzLzA4LzIwMjEuMDMuMDUuMjEyNTI3MDkuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 9. 9.Grubaugh ND, Hodcroft EB, Fauver JR, Phelan AL, Cevik M. 2021. Public health actions to control new SARS-CoV-2 variants. Cell doi:10.1016/j.cell.2021.01.044. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.cell.2021.01.044&link_type=DOI) 10. 10.Golubchik T, Lythgoe KA, Hall M, Ferretti L, Fryer HR, MacIntyre-Cockett G, de Cesare M, Trebes A, Piazza P, Buck D, Todd JA, Fraser C, Bonsall D. 2021. Early analysis of a potential link between viral load and the N501Y mutation in the SARS-COV-2 spike protein. medRxiv doi:10.1101/2021.01.12.20249080:2021.01.12.20249080. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoibWVkcnhpdiI7czo1OiJyZXNpZCI7czoyMToiMjAyMS4wMS4xMi4yMDI0OTA4MHYxIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjEvMDMvMDgvMjAyMS4wMy4wNS4yMTI1MjcwOS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 11. 11.Leung K, Shum MH, Leung GM, Lam TT, Wu JT. 2021. Early transmissibility assessment of the N501Y mutant strains of SARS-CoV-2 in the United Kingdom, October to November 2020. Eurosurveillance 26:2002106. 12. 12.Carolina Kymie Vasques Nonaka MMF, Tiago Gräf, Ana Verena Almeida Mendes, Renato Santana de Aguiar, Marta Giovanetti * ORCID logo, Bruno Solano de Freitas Souza. 2021. Genomic Evidence of a Sars-Cov-2 Reinfection Case With E484K Spike Mutation in Brazil. Preprints 2021010132 (doi: 10.20944/preprints202101.0132.v1). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.20944/preprints202101.0132.v1&link_type=DOI) 13. 13.Weisblum Y, Schmidt F, Zhang F, DaSilva J, Poston D, Lorenzi JC, Muecksch F, Rutkowska M, Hoffmann HH, Michailidis E, Gaebler C, Agudelo M, Cho A, Wang Z, Gazumyan A, Cipolla M, Luchsinger L, Hillyer CD, Caskey M, Robbiani DF, Rice CM, Nussenzweig MC, Hatziioannou T, Bieniasz PD. 2020. Escape from neutralizing antibodies by SARS-CoV-2 spike protein variants. Elife 9. 14. 14.Zhang W, Davis BD, Chen SS, Martinez JMS, Plummer JT, Vail E. 2021. Emergence of a novel SARS-CoV-2 strain in Southern California, USA. medRxiv doi:10.1101/2021.01.18.21249786:2021.01.18.21249786. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoibWVkcnhpdiI7czo1OiJyZXNpZCI7czoyMToiMjAyMS4wMS4xOC4yMTI0OTc4NnYxIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjEvMDMvMDgvMjAyMS4wMy4wNS4yMTI1MjcwOS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 15. 15.CDC. 2021. National Genomic Surveillance Dashboard. 16. 16. Chantal B.F. Vogels Ta, Mallery Breban, Joseph R. Fauver, Nathan D. Grubaugh. 2021. Multiplexed RT-qPCR to screen for SARS-COV-2 B.1.1.7 variants: Preliminary results. [https://virological.org/t/multiplexed-rt-qpcr-to-screen-for-sars-cov-2-b-1-1-7-variants-preliminary-results/588](https://virological.org/t/multiplexed-rt-qpcr-to-screen-for-sars-cov-2-b-1-1-7-variants-preliminary-results/588). Accessed 17. 17.Ozsolak F, Milos PM. 2011. RNA sequencing: advances, challenges and opportunities. Nat Rev Genet 12:87–98. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nrg2934&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21191423&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F03%2F08%2F2021.03.05.21252709.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000286319600010&link_type=ISI) 18. 18.Lopez-Rincon A, Tonda A, Mendoza-Maldonado L, Mulders D, Molenkamp R, Perez-Romero CA, Claassen E, Garssen J, Kraneveld AD. 2021. Classification and specific primer design for accurate detection of SARS-CoV-2 using deep learning. Sci Rep 11:947. 19. 19.Roh SS, Smith LE, Lee JS, Via LE, Barry CE, 3rd, Alland D, Chakravorty S. 2015. Comparative Evaluation of Sloppy Molecular Beacon and Dual-Labeled Probe Melting Temperature Assays to Identify Mutations in Mycobacterium tuberculosis Resulting in Rifampin, Fluoroquinolone and Aminoglycoside Resistance. PLoS One 10:e0126257. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1371/journal.pone.0126257&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25938476&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F03%2F08%2F2021.03.05.21252709.atom) 20. 20.Elbe S, Buckland-Merrett G. 2017. Data, disease and diplomacy: GISAID’s innovative contribution to global health. Global challenges (Hoboken, NJ) 1:33–46. 21. 21.Altschul SF, Madden TL, Schäffer AA, Zhang J, Zhang Z, Miller W, Lipman DJ. 1997. Gapped BLAST and PSI-BLAST: a new generation of protein database search programs. Nucleic Acids Res 25:3389–402. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/nar/25.17.3389&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=9254694&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F03%2F08%2F2021.03.05.21252709.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1997XU79300002&link_type=ISI) 22. 22.Katoh K, Misawa K, Kuma K, Miyata T. 2002. MAFFT: a novel method for rapid multiple sequence alignment based on fast Fourier transform. Nucleic Acids Res 30:3059–66. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/nar/gkf436&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=12136088&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F03%2F08%2F2021.03.05.21252709.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000177154300016&link_type=ISI) 23. 23.SantaLucia J, Jr.. 1998. A unified view of polymer, dumbbell, and oligonucleotide DNA nearest-neighbor thermodynamics. Proc Natl Acad Sci U S A 95:1460–5. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoicG5hcyI7czo1OiJyZXNpZCI7czo5OiI5NS80LzE0NjAiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMS8wMy8wOC8yMDIxLjAzLjA1LjIxMjUyNzA5LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 24. 24.Untergasser A, Cutcutache I, Koressaar T, Ye J, Faircloth BC, Remm M, Rozen SG. 2012. Primer3--new capabilities and interfaces. Nucleic acids research 40:e115–e115. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/nar/gks596&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22730293&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F03%2F08%2F2021.03.05.21252709.atom) 25. 25.Emery SL, Erdman DD, Bowen MD, Newton BR, Winchell JM, Meyer RF, Tong S, Cook BT, Holloway BP, McCaustland KA, Rota PA, Bankamp B, Lowe LE, Ksiazek TG, Bellini WJ, Anderson LJ. 2004. Real-time reverse transcription-polymerase chain reaction assay for SARS-associated coronavirus. Emerging infectious diseases 10:311–316. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3201/eid1002.030759&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=15030703&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F03%2F08%2F2021.03.05.21252709.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000188867700028&link_type=ISI) 26. 26.Lu X, Wang L, Sakthivel SK, Whitaker B, Murray J, Kamili S, Lynch B, Malapati L, Burke SA, Harcourt J, Tamin A, Thornburg NJ, Villanueva JM, Lindstrom S. 2020. US CDC Real-Time Reverse Transcription PCR Panel for Detection of Severe Acute Respiratory Syndrome Coronavirus 2. Emerg Infect Dis 26:1654–65. 27. 27.Nelson G, Buzko O, Spilman P, Niazi K, Rabizadeh S, Soon-Shiong P. 2021. Molecular dynamic simulation reveals E484K mutation enhances spike RBD-ACE2 affinity and the combination of E484K, K417N and N501Y mutations (501Y.V2 variant) induces conformational change greater than N501Y mutant alone, potentially resulting in an escape mutant. bioRxiv doi:10.1101/2021.01.13.426558:2021.01.13.426558. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoiYmlvcnhpdiI7czo1OiJyZXNpZCI7czoxOToiMjAyMS4wMS4xMy40MjY1NTh2MSI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIxLzAzLzA4LzIwMjEuMDMuMDUuMjEyNTI3MDkuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9)