key: cord-0866126-zmq8l82a authors: Annavajhala, Medini K.; Mohri, Hiroshi; Wang, Pengfei; Nair, Manoj; Zucker, Jason E.; Sheng, Zizhang; Gomez-Simmonds, Angela; Kelley, Anne L.; Tagliavia, Maya; Huang, Yaoxing; Bedford, Trevor; Ho, David D.; Uhlemann, Anne-Catrin title: Emergence and expansion of SARS-CoV-2 B.1.526 after identification in New York date: 2021-08-24 journal: Nature DOI: 10.1038/s41586-021-03908-2 sha: 760cd448db92a5208213b871d5cf338417fae7c2 doc_id: 866126 cord_uid: zmq8l82a SARS-CoV-2 infections have surged across the globe in recent months, concomitant with considerable viral evolution(1–3). Extensive mutations in the spike protein may threaten the efficacy of vaccines and therapeutic monoclonal antibodies(4). Two signature spike mutations of concern are E484K, which has a crucial role in the loss of neutralizing activity of antibodies, and N501Y, a driver of rapid worldwide transmission of the B.1.1.7 lineage. Here we report the emergence of the variant lineage B.1.526 (also known as the Iota variant(5)), which contains E484K, and its rise to dominance in New York City in early 2021. This variant is partially or completely resistant to two therapeutic monoclonal antibodies that are in clinical use and is less susceptible to neutralization by plasma from individuals who had recovered from SARS-CoV-2 infection or serum from vaccinated individuals, posing a modest antigenic challenge. The presence of the B.1.526 lineage has now been reported in all 50 states in the United States and in many other countries. B.1.526 rapidly replaced earlier lineages in New York, with an estimated transmission advantage of 35%. These transmission dynamics, together with the relative antibody resistance of its E484K sub-lineage, are likely to have contributed to the sharp rise and rapid spread of B.1.526. Although SARS-CoV-2 B.1.526 initially outpaced B.1.1.7 in the region, its growth subsequently slowed concurrently with the rise of B.1.1.7 and ensuing variants. Evolution of SARS-CoV-2 was slow at the beginning of the global pandemic 6 ; however, multiple major variants of concern have emerged over the past year 1-3, 7 . These lineages are characterized by mutations in the spike protein, raising concerns that they may escape from therapeutic monoclonal and vaccine-induced antibodies. The hallmark mutation of B.1.1.7-a SARS-CoV-2 variant of concern first identified in the UK-is N501Y, located in the receptor-binding domain (RBD) of spike 1 . This mutation appears to render the virus more transmissible and virulent [8] [9] [10] , perhaps owing to a higher binding affinity of N501Y for the ACE2 receptor 11 or a greater propensity to evade host innate immune responses 12 . Two other variants of concern, B.1.351 2 and P.1 3 , also harbour the N501Y mutation, in addition to an E484K substitution in the RBD 2,3 . P.1 was identified as part of a second surge in Manaus, Brazil, despite a high pre-existing SARS-CoV-2 seroprevalence in the population 13, 14 . Reinfections with P.1 and another related Brazilian variant P.2 harbouring E484K have been documented 15, 16 . A previous study on B.1.351 demonstrated that this variant is refractory to neutralization by a number of monoclonal antibodies directed to the top of the RBD, including several that have received emergency use authorization 4 . B.1.351 was markedly more resistant to neutralization by plasma from individuals who had recovered from SARS-CoV-2 infection and sera from vaccinated individuals. Of note, these effects were mediated in part by the E484K mutation. These finding are concerning in light of recent reports that three vaccine trials in South Africa showed a substantial drop in efficacy 17, 18 . Similarly, P.1 was also relatively resistant to antibody neutralization, although to a lesser degree 19 . In this study, we have implemented rapid molecular screening for signature mutations implicated in the success of these early variants of concern. We next performed untargeted whole-genome nanopore sequencing of nasopharyngeal swab samples collected throughout the study period with a cycle threshold (C t ) ≤ 35. We obtained 1,507 SARS-CoV-2 whole genomes (59% of samples with C t ≤ 35; Extended Data Fig. 2 ). Sequencing results verified the E484K and N501Y substitutions in all samples identified by PCR screening. Of the sequenced N501Y isolates, 31 out of 41 (76%) were consistent with the B.1.1.7 lineage. Samples that harboured both N501Y and E484K were genotyped as P.1 (n = 6), B.1.351 (n = 1) and B.1.623 (n = 1). However, unexpectedly, the large majority of PCR-screened cases with E484K (98 out of 128 (77%)) were from the B.1.526 lineage 20 . Analysis of this genomic collection (Fig. 1b) showed that by May 2021, SARS-CoV-2 variants (including B.1.526, B.1.1.7 and, more recently, P.1) comprised two-thirds of all sequenced isolates, replacing the vast majority of earlier lineages (Fig. 1b) . The proportion of infections caused by B.1.526 rose rapidly from late 2020 to February 2021, and remained at approximately 40-50% of all sequenced cases from March to May 2021, despite a concurrent increase in B.1.1.7. Indeed, during December and January, when the prevalence of B.1.1.7 was still negligible (Fig. 1b) , the frequency of all viruses in the B.1.526 lineage increased from less than 5% to 50%, while the frequency of other lineages declined from more than 95% to 50% (Fig. 1b, where white blank space represents other lineages). Calculations using these numbers in a head-to-head comparison and an established mathematical method 21 indicate that B.1.526 has a growth advantage of approximately 5% per day. Similarly, fitting a logistic regression model to 478 individual observations from the extended timeframe of November 2020 to January 2021 shows that B.1.526 had a similar growth advantage of 4.6% per day (95% confidence interval 2.8-6.5% per day). Given that the serial interval for SARS-CoV-2 transmission is about 7 days in the absence of any intervention 22 To further investigate the evolutionary history of B.1.526, we performed phylogenetic analyses on genomes in this collection and in the GISAID collection harbouring the ORF1a-nsp6 deletion Δ106-108, along with the mutation A20262G that uniquely defines the parent clade containing B.1.526 and related viruses (Fig. 2a) . We observed a stepwise emergence of the key lineage-defining mutations, with T95I, D253G and L5F appearing in the earliest phylogenetic nodes. Isolates subsequently branched into four sub-lineages, with two major groups B.1.526-E484K and B.1.526-S477N containing A701V, and a smaller sub-lineage B.1.526-S477N containing Q957R. The B.1.526-L452R lineage-which emerged in parallel with these-is related to B.1.526-E484K and B.1.526-S477N, but forms a distinct phylogenetic branch (Extended Data Fig. 3 ). Fig. 2b shows the localization of signature B. the antigenic supersite in the N-terminal domain 24 , which is a target for neutralizing antibodies 25 , whereas E484K is situated at the RBD interface with the cellular receptor ACE2. The A701V mutation near the furin cleavage site is also shared with variant B.1.351. The effects of the signature spike protein mutations in B.1.526 on antibody neutralization were first assessed using vesicular stomatitis virus (VSV)-based pseudoviruses, as previously described 4,25 . Pseudoviruses were constructed containing S477N or E484K alone, or containing all five signature mutations (L5F, T95I, D253G, A701V and E484K or S477N) (NYΔ5(E484K) or NYΔ5(S477N)), and analysed in a neutralization assay with 12 monoclonal antibodies (including 5 with emergency use authorization), 20 plasma samples from patients who had recovered from SARS-CoV-2 infection and 22 sera from vaccinated individuals 4 . The neutralizing activities of 12 monoclonal antibodies covering a range of epitopes on RBD were essentially unaltered against the S477N and NYΔ5(S477N) pseudoviruses (Extended Data Fig. 4a) , showing that this mutation has no discernible antigenic impact, as was validated using convalescent plasma and vaccinee sera (Extended Data Fig. 4b) . However, the activities of several antibodies-including REGN10933 and LY-CoV555, which are already in clinical use-were either impaired or lost when tested against E484K and NYΔ5(E484K) pseudoviruses (Fig. 3a) . Similarly, neutralizing activities of convalescent plasma or vaccinee sera were reduced by 4.1-fold or 3.3-to 3.6-fold, respectively, against NYΔ5(E484K) (Fig. 3b) . Neutralization studies on the authentic B.1.526-E484K virus yielded similar results, although the magnitudes of resistance to convalescent plasma and vaccinee sera were slightly lower at 2.6-fold and 1.8-to 2.0-fold, respectively (Fig. 3b) . A comparative analysis with other variants of concern ( Fig. 3c) that are already in clinical use. Furthermore, neutralizing activities of plasma from individuals who have recovered from SARS-CoV-2 infection or sera from vaccinated individuals were lower against B.1.526-E484K. By contrast, the S477N mutation, a key signature of another B.1.526 sub-lineage, did not have an impact on antibody neutralization. This study has several limitations. This was a single-centre genomic survey of patients attending hospital and may not have fully captured patients with milder disease. However, our results are comparable to genomic data released by public health laboratories in the region and incorporate all publicly available data for phylogeographic context and growth rate calculations. As in all genomic surveillance studies, we predominantly sequenced samples with a C t less than 30, but this included a high proportion of samples throughout the study period. In addition, our PCR screen enabled us to obtain unbiased estimates of E484K and N501Y prevalence early on in the study. PCR approaches may be increasingly warranted for continued surveillance during non-surge periods, during which C t values trend higher. Finally, transmissibility estimates based on observed prevalence are imperfect as they reflect observed growth rates rather than intrinsic transmissibility of the virus. Together, our findings underscore the importance of the E484K mutation, which has emerged in at least 246 different lineages 28 of SARS-CoV-2, a powerful illustration of convergent evolution. This highlights that E484K can rapidly emerge in multiple clonal backgrounds and may warrant targeted screening for this key mutation in addition to robust genomic surveillance programs. However, B.1.526 is one of the few lineages with E484K that has risen to prominence. The greatest threat of B.1.526 appears to be the ease with which it spreads, with an estimated transmissibility approximately 35% higher than non-variant viruses when competing head to head. Despite the notable transmissibility of B. 1.1.7, B.1.526 was able to spread rapidly in the United States, replacing other lineages and continuing to increase in frequency in several states where both B.1.526 and B.1.1.7 were predominant. Similarly, although B.1.351 may pose the greatest antigenic challenge to antibodies and vaccines, the B.1.526-E484K sub-lineage also exhibits resistance to antibody neutralization. Our findings present a clear-cut example of SARS-CoV-2 evolution in real time. B.1.526, with its higher transmissibility, appeared suddenly and rose to dominance, only to wane as variants (B.1.1.7 and, more recently, B.1.617.2) with even higher fitness emerged. These observations are a stark reminder that increasingly concerning variants are expected to emerge if SARS-CoV-2 is allowed to continue its spread. Any methods, additional references, Nature Research reporting summaries, source data, extended data, supplementary information, acknowledgements, peer review information; details of author contributions and competing interests; and statements of data and code availability are available at https://doi.org/10.1038/s41586-021-03908-2. This observational study took place at an academic quaternary care centre in New York City. Nasopharyngeal swabs obtained as part of routine clinical care were tested by the Clinical Microbiology laboratory, and positive specimens were transferred to the Columbia University Biobank for inactivation and storage. Electronic health records data extracted for this analysis included demographics, laboratory results, admission, discharge and transfer dates, current and historical international classification of disease codes (ICD 9 and ICD 10) extracted from the clinical data warehouse. The study was reviewed and approved by the Columbia University Institutional Review Board (IRB) (AAAT0123). The IRB waived consent for the entirety of this observational study, including for the collection and sequencing of the viral samples as well as the abstraction of the clinical metadata, as this observational study met the requirements for this exception. These include minimal risks to subjects, not adversely affecting the rights and welfare of the subjects, and that the research could not be carried out without the waiver. Extended Data Fig. 1 describes the overall protocol for variant screening. To enable rapid PCR-based screening, we prepared RNA using the heat inactivation method in place of RNA-isolation methods 29 . First, 50 µl of nasal swab sample in viral transport medium was transferred to 96-well PCR plates, covered with an adhesive aluminium foil (VWR 60941-076) and incubated at 95 °C for 5 min using the PCR instrument. After the centrifugation of the plate at >2,100g for 5 min, 5 µl of the supernatant from each sample, which contains viral RNA, was used for the SNP assay. The SNP assay consists of four steps as follows: reverse transcription of viral RNA, pre-read of the SNP assay, real-time PCR and post-read of the SNP assay. 5 µl of RNA from the supernatant was added to 15 µl of the single step quantitative PCR with reverse transcription (RT-qPCR) reaction mix, which consists of 5 µl of TaqPath 1-step RT-qPCR Master Mix, CG (4×) (ThermoFisher Scientific), 500 nM of forward and reverse primers, 120 nM of VIC-MGB probe, 50 nM of FAM-MGB probe, 1/2000 volume of ROX Reference Dye (Invitrogen) as the final concentration, and nuclease-free water to adjust the total reaction volume of 20 µl. Each reaction plate included 8 control wells, 5 × 10 6 and 5 × 10 3 copies of WA-1 (wild type), B.1.1.7 and B.1.351, which were generated by PCR to match the variant sequences, and 2 wells with water as no template controls (NTC). The The reaction plate was subjected to: (1) reverse transcription reaction at 25 °C for 2 min, 50 °C for 15 min and a hold at 4 °C; (2) SNP assay (pre-read) at 60 °C for 30 s; (3) real-time PCR at 95 °C for 20 s followed by 50 cycles of two-step PCR, at 95 °C for 3 s and at 60 °C for 30 s with the fast 7500 mode; followed by (4) SNP assay (post-read) at 60 °C for 30 s using ABI 7500 Fast Dx real-time PCR instrument with SDS Software (ThermoFisher Scientific). The genotype at each key position for each sample was determined by reading the component signal of the amplification and the allelic discrimination analysis software in the program. Extended Data Fig. 2 displays a flowchart outlining samples available for this study. Isolates with C t values below 35 were selected for sequencing using the ARTIC v3 low-cost protocol targeting 400-bp amplicons 30 or Rapid Barcoding kit protocol targeting 1,200-bp amplicons 31 . In brief, RNA was extracted using the Qiagen RNeasy Mini kit or Zymo DNA/RNA Mini kit. Reverse transcription was performed using LunaScript RT SuperMix (NEB). Tiling PCR was performed on the cDNA, and amplicons were barcoded using the Oxford Nanopore Native Barcoding Expansion 96 kit. Pooled barcoded libraries were then sequenced on an Oxford Nanopore MinION sequencer using R9.4.1 flow cells. Base calling was performed in MinKNOW software v21.02.1. Sequencing runs were monitored in real time using RAMPART (https:// artic-network.github.io/rampart/) to ensure sufficient genomic coverage with minimal runtime. Consensus sequence generation was performed using the ARTIC bioinformatics pipeline (https://github. com/artic-network/artic-ncov2019). Genomes were manually curated by visually inspecting sequencing alignment files for verification of key residues in Geneious v10.2.6. Phylogenetic reconstruction of amino acid changes (Fig. 2a) was conducted using the Nextstrain 32 workflow at https://github.com/nextstrain/ncov, which aligns sequences against the Wuhan Hu-1 reference using nextalign (https://github.com/nextstrain/nextclade), constructs a maximum-likelihood phylogenetic tree via IQ-TREE 33 , estimates molecular clock branch lengths via TreeTime 34 and reconstructs nucleotide and amino acid changes (also via TreeTime). This workflow was applied to 2,309 SARS-CoV-2 genomes with the 9-bp deletion Δ106-108 in ORF1a-nsp6 along with mutation A20262G, which demarcates the parent clade to lineage B.1.526 alongside 688 global reference viruses. This analysis was conducted on data downloaded 35 from GISAID (https:// gisaid.org/) on 5 April 2021. Phylogeographic reconstruction of spread from New York state (Fig. 4e, f) was similarly conducted using the same Nextstrain workflow with the addition of performing ancestral trait reconstruction of the geographic 'division' attribute of 933 SARS-CoV-2 genomes downloaded from GISAID on 6 Jun 2021. We assayed the neutralizing activity of monoclonal antibodies, convalescent plasma and vaccinee sera against E484K, S477N and wild-type (D614G) pseudoviruses, as well as pseudovirus NYΔ5 containing all five signature mutations of B.1.526-E484K (L5F, T95I, D253G, E484K, D614G and A701V), as previously described 25 . We examined four monoclonal antibodies with emergency use authorization (CB6, REGN10987, REGN10933 and LY-CoV555), and eight additional RBD monoclonal antibodies, including from our own collection (2-15, 2-7, 1-57 and 2-36) 25 as well as S309 36 , COV2-2196 and COV2-2130 37 , and C121 38 . We also examined convalescent plasma collected in March and April 2020 (n = 20 patients), and sera from individuals who had received Moderna or Pfizer vaccine 4 (n = 22). In brief, Vero E6 cells (ATCC) were seeded in 96-well plates (2 × 10 4 cells per well). Cell lines were negative for Mycoplasma, as assessed using the Mycoplasma PCR ELISA (Sigma). Pseudoviruses were incubated with serial dilutions of the test samples in triplicate for 30 min at 37 °C. The mixture was added to cultured cells and incubated for an additional 24 h. Luminescence was measured using a Britelite plus Reporter Gene Assay System (PerkinElmer), and IC 50 was defined as the dilution at which the relative light units were reduced by 50% compared with the virus control wells (virus + cells) after subtraction of the background in the control groups with cells only. The IC 50 values were calculated using nonlinear regression in GraphPad Prism 8.0. Statistical analysis was performed using a Wilcoxon matched-pairs signed rank test. Two-tailed P-values are reported. Infectious SARS-CoV-2 isolate hCoV-19/USA/NY-NP-DOH1/2021 was isolated at the Aaron Diamond AIDS Center (Columbia University Medical Center) from a nasopharyngeal swab and propagated for one passage in Vero E6 cells (ATCC). Infectious titre of the resulting virus was determined by an end-point dilution and cytopathic effect (CPE) assay on Vero-E6 cells as described previously 25 . The virus has since been deposited at BEI Resources (catalogue (cat.) no. NR-55359). SARS-CoV-2 virus USA-WA1/2020 (WA1), obtained from BEI Resources (cat. no. NR-52281) served as the control in experiments. An end-point dilution microplate neutralization assay was performed to measure the neutralization activity of twenty convalescent patient plasma samples and twelve purified monoclonal antibodies. In brief, plasma samples were subjected to successive fivefold dilutions starting from 1:100. Similarly, antibodies were serially diluted (fivefold dilutions) starting at 50 µg ml −1 . Triplicates of each dilution were incubated with SARS-CoV-2 at an multiplicity of infection of 0.1 in Eagle's minimum essential medium (EMEM; ATCC) with 7.5% inactivated fetal calf serum (FCS) for 1 h at 37 °C. After incubation, the virus-antibody mixture was transferred to a monolayer of Vero-E6 cells grown overnight. The cells were incubated with the mixture for about 70 h. CPE of viral infection was visually scored for each well in a blinded fashion by two independent observers. The results were then converted into percentage neutralization at a given sample dilution or antibody concentration, and the mean ± s.e.m. was plotted using a five-parameter dose-response curve in GraphPad Prism v8.4. Growth dynamics of B.1.1.7 and B.1.526 were obtained through by downloading 'metadata' from GISAID on 6 June 2021 for all 422,760 viruses sampled from the United States collected after 1 January 2021. These metadata have PANGO lineages 39 already assigned to each genome sequence. Daily state-level frequencies (and frequencies for CUIMC) were extracted for plotting using seven-day sliding window averages of the prevalence of B.1.1.7 and B.1.526, calculated as the number of sequence-verified samples from each strain divided by the total number of positive samples with C t values below 35, as this threshold value was used for sequencing. Separately, a multinomial logistic regression model was fit directly to the observation data consisting of individual genomes, their dates of sampling (independent variable X in days since 1 January 2021) and their category labels (dependent variable Y, "B.1.1.7", "B.1.526" and "other"). This results in a 4-parameter model where both B.1.1.7 and B.1.526 have parameters specified for frequency at day 0 (1 January 2021) and logistic growth rate. This model was fit to the data using the Classify package of Mathematica v12.2. Further information on research design is available in the Nature Research Reporting Summary linked to this paper. All genomes and associated metadata generated as a part of this study have been uploaded to GISAID (https://gisaid.org) and NCBI GenBank (BioProject Accession PRJNA751551). Biological materials (that is, variant pseudoviruses) generated as a part of this study will be made available but may require execution of a materials transfer agreement. Data processing and visualization were performed using publicly available software and packages, primarily RStudio v1.2.5033, Graph-Pad Prism v8.4, and iTOL (https://itol.embl.de/). The exact workflows used for phylogenetic (Fig. 2a) and phylogeographic analysis of public GISAID data (Fig. 4e, f) are available at https://github.com/blab/ ncov-ny. Frequency dynamics were modelled using Mathematica in notebooks also available at https://github.com/blab/ncov-ny. Phylogenetic tree based on whole-genome alignment of genomes sequenced from our hospital centre with at least one mutation of interest or concern (E484K, N501Y, S477N, or L452R) and unique spike protein mutation constellations (n = 64). Branches are labelled according to Pangolin-assigned lineage identifications. Residues at which at least one sample harboured a mutation are displayed above the S-protein schematic. Residues highlighted in colour represent mutations when compared to the Wuhan-Hu-1 strain. Fig. 4 | Neutralization studies of B.1.526-S477N Fig. 5 | State-level growth dynamics of B.1.526 and B.1.1.7 . Owing to non-normal distribution, a Kruskal-Wallis non-parametric test was used. 4 Cycle threshold value as determined through our rapid qPCR-based screening assay on heat-inactivated nasopharyngeal swab samples. Last updated by author(s): Jul 29, 2021 Reporting Summary Nature Research wishes to improve the reproducibility of the work that we publish. This form provides structure for consistency and transparency in reporting. For further information on Nature Research policies, see our Editorial Policies and the Editorial Policy Checklist. For all statistical analyses, confirm that the following items are present in the figure legend, table legend, main text, or Methods section. The exact sample size (n) for each experimental group/condition, given as a discrete number and unit of measurement A statement on whether measurements were taken from distinct samples or whether the same sample was measured repeatedly The statistical test(s) used AND whether they are one-or two-sided Only common tests should be described solely by name; describe more complex techniques in the Methods section. A description of all covariates tested A description of any assumptions or corrections, such as tests of normality and adjustment for multiple comparisons A full description of the statistical parameters including central tendency (e.g. means) or other basic estimates (e.g. regression coefficient) AND variation (e.g. standard deviation) or associated estimates of uncertainty (e.g. confidence intervals) For null hypothesis testing, the test statistic (e.g. F, t, r) with confidence intervals, effect sizes, degrees of freedom and P value noted Policy information about availability of computer code Data collection PCR-based genotyping of nasopharyngeal swabs was performed on an ABI 7500 Fast Dx Real-Time PCR Instrument with SDS Software (ThermoFisher Scientific). Genomic sequencing was performed on an Oxford MinION and Oxford GridION and basecalling was performed using MinKNOW v21.02.1. For pseudovirus neutralization assays, SoftMax Pro v7.0.2 (Molecular Devices) was used to measure luminescence. Basecalling was performed in the MinKNOW software v21.02.1. Sequencing runs were monitored in real-time using RAMPART (https://articnetwork.github.io/rampart/) v1.2.0 or v1.2.1 to ensure sufficient genomic coverage with minimal runtime. Consensus sequence generation was performed using the ARTIC bioinformatics pipeline (https://github.com/artic-network/artic-ncov2019). Genomes were manually curated by visually inspecting sequencing alignment files for verification of key residues in Geneious v10.2.6. RStudio v1.2.5033, GraphPad Prism v8.4, and Mathematica v.12.2 were used for data representation and statistical analysis. Phylogenetic analyses utilized MAFFT v1.4.0 and IQ-TREE v2.0.3. The exact workflow used for phylogenetic analysis of public GISAID data (Fig. 2a, Fig. 4e-f ) and frequency dynamic modeling is available at https://github.com/blab/ncov-ny. For manuscripts utilizing custom algorithms or software that are central to the research but not yet described in published literature, software must be made available to editors and reviewers. We strongly encourage code deposition in a community repository (e.g. GitHub). See the Nature Research guidelines for submitting code & software for further information. Temporal signal and the phylodynamic threshold of SARS-CoV-2 Convergent evolution of SARS-CoV-2 spike mutations, L452R, E484Q and P681R, in the second wave of COVID-19 in Maharashtra Covid-19: new UK variant may be linked to increased death rate, early data indicate Transmission of SARS-CoV-2 lineage B.1.1.7 in England: insights from linking epidemiological and genetic data Emergence and rapid transmission of SARS-CoV-2 B.1.1.7 in the United States Comprehensive mapping of mutations in the SARS-CoV-2 receptor-binding domain that affect recognition by polyclonal human plasma antibodies Evolution of enhanced innate immune evasion by the SARS-CoV-2 B.1.1.7 UK variant SARS-CoV-2 reinfection by the new variant of concern (VOC) P.1 in Amazonas, Brazil Resurgence of COVID-19 in Manaus, Brazil, despite high seroprevalence Severe reinfection with South African SARS-CoV-2 variant 501Y.V2: a case report Genomic evidence of SARS-CoV-2 reinfection involving E484K spike mutation Novavax offers first evidence that COVID vaccines protect people against variants Efficacy of the ChAdOx1 nCoV-19 Covid-19 vaccine against the B.1.351 variant Increased resistance of SARS-CoV-2 variant P.1 to antibody neutralization Detection and characterization of the SARS-CoV-2 lineage B.1.526 in New York Human immunodeficiency virus fitness in vivo: calculations based on a single zidovudine resistance mutation at codon 215 of reverse transcriptase Serial interval of SARS-CoV-2 was shortened over time by nonpharmaceutical interventions SARS-CoV-2 variant classifications and definitions Potent SARS-CoV-2 neutralizing antibodies directed against spike N-terminal domain target a single supersite Potent neutralizing antibodies against multiple epitopes on SARS-CoV-2 spike COVID-19: Data The localized rise of a B.1.526 SARS-CoV-2 variant containing an E484K mutation in New York State Massive and rapid COVID-19 testing is feasible by extraction-free SARS-CoV-2 RT-PCR Artic protocol. Protocols Rapid and inexpensive whole-genome sequencing of SARS-CoV2 using 1200 bp tiled amplicons and Oxford Nanopore rapid barcoding Nextstrain: real-time tracking of pathogen evolution IQ-TREE 2: new models and efficient methods for phylogenetic inference in the genomic era TreeTime: maximum-likelihood phylodynamic analysis GISAID: global initiative on sharing all influenza data-from vision to reality Cross-neutralization of SARS-CoV-2 by a human monoclonal SARS-CoV antibody Rapid isolation and profiling of a diverse panel of human monoclonal antibodies targeting the SARS-CoV-2 spike protein Convergent antibody responses to SARS-CoV-2 in convalescent individuals A dynamic nomenclature proposal for SARS-CoV-2 lineages to assist genomic epidemiology Data Policy information about availability of data All manuscripts must include a data availability statement. This statement should provide the following information, where applicable: -Accession codes, unique identifiers, or web links for publicly available datasets -A list of figures that have associated raw data -A description of any restrictions on data availability All genomes and associated metadata generated as a part of this study have been uploaded to GISAID (gisaid.org) and NCBI GenBank (BioProject Accession PRJNA751551) For pseudovirus and B.1.526-E484K live virus assays, we obtained convalescent plasma from 20 patients, vaccinee sera from 12 Moderna SARS-Co-2 mRNA-1273 vaccine participants, and sera from 10 Pfizer BNT162b2 Covid-19 vaccine participants. This sample size is appropriate within technical capability to compare the difference between groups, as we have previously published. Phylogenetic analysis of the emergence of B.1.526 and its sublineages included 2,997 publicly available genomes from GISAID; phylogeographic analysis included 933 B.1.526 genomes from the GISAID database. Growth rate calculations per state included in total data from 422,760 genomes collected between Data exclusions Genomes with greater than 8% ambiguous bases (3000 Ns) were excluded from phylogenetic analyses, based on standard criteria used in NextStrain bioinformatics pipelines and based on GISAID genome upload quality criteria Pseudovirus experiments in this manuscript were performed in three successful replicates prior to reporting. For the B.1.526-E484K live virus experiments, a minimum of three successful replicates were used. Neutralization assays for variants not performed in this manuscript but reported here, were performed successfully in at least triplicates and the detailed summary of those experiments have been reported in Wang et al., 2021. Randomization This observational study was not randomized Reporting for specific materials, systems and methods We require information from authors about some types of materials, experimental systems and methods used in many studies. Here, indicate whether each material, system or method listed is relevant to your study )) were also provided. The remaining monoclonal antibodies used in this study (1-57, 2-7, 2-15, 2-36) were produced at Columbia University as described in Liu et al. 2020 (Ref#27) Validation All of the SARS-CoV2 spike antigen-specific monoclonal antibodies obtained from commercial sources and produced in house have been thoroughly validated by epitope mapping and binding analysis to SARS-CoV-2 spike protein and neutralization experiments to SARS-CoV-2 pseudoviruses and described in our previous publications Vero E6 (ATCC, Cat# CRL-1586) cell lines were used Authentication Cell lines were obtained from authenticated vendors; additional authentication was not performed. Cells were recovered as healthy logarithmically growing cells within 4 to 7 days after thawing. Viability was measured and found to be >90% Mycoplasma contamination All cell lines were negative for Mycoplasma, as assessed using the Mycoplasma PCR ELISA (Sigma Catalog No. 11663925910) Demographic and clinical characteristics for patients whose nasopharyngeal swab samples were included in this study are shown in Extended Data Table 1. Plasma samples were obtained from patients (median age 51.5; IQR 48-60; 13 males (65%) and 7 females (35%)) convalescing from documented SARS-CoV-2 infection approximately one month after recovery or later Acknowledgements Biospecimens used for this research were obtained from the Columbia University Biobank (CUB) with technical support from V. J. Mahadeva, S. Fernando and S. T. Parker-Jones. CUB is supported by the Irving Institute for Clinical and Translational Research (NCATS UL1TR001873). In particular, we thank M. Reilly, E. Hod and the CUB COVID-19 Genomics Consortium for facilitating this effort. We also thank L. Liu and S. Iketani for technical support, and A. Perelson for mathematical input. We gratefully acknowledge all the authors, the originating laboratories responsible for obtaining the specimens and the submitting laboratories for generating the genetic sequence and metadata and sharing via the GISAID Initiative, on which part of the presented research is based. This work was in part funded by NIH/ NIDA grant U01 DA053949 (A.-C.U. and M.K.A.) and by support from Andrew and Peggy Cherng, Samuel Yin, Barbara Picower and the JBP Foundation, Brii Biosciences, Roger and David Wu, and the Bill and Melinda Gates Foundation. T.B. is a Pew Biomedical Scholar and is supported by NIH grant no. R35 GM119774-01. Funders and funding agencies had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript. Fig. 1 | Rapid PCR-based screening assay protocol to identify samples harboring key substitutions. a, Viral RNA is prepared by heat inactivation and centrifugation. The supernatant is then used for the SNP assay, which entails four steps: the reverse transcription (RT) reaction, pre-PCR reading of the plate to assess background fluorescence (SNP pre-read), real-time PCR, and post-PCR reading of the plate to measure fluorescence (SNP post-read). The runtime for this entire protocol is approximately two hours. b, Genotype at targeted sites in COVID-19 viral RNA can be determined with two MGB probes, one for wild type (conjugated with VIC) and the other for variant type (conjugated with FAM). c, Example signals for the variant type (K484; blue), the wild type (E484; red) and samples with no signal (black) are shown. Nasopharyngeal swabs obtained as part of routine clinical care were tested by the Clinical Microbiology laboratory, and positive specimens were transferred to the Columbia University Biobank for inactivation and storage; participants were not recruited for this study. Convalescing patients volunteered for the cohort study. These cases were enrolled into an observational cohort study of convalescent patients followed at the Columbia University Irving Medical Center starting in the Spring of 2020. From their documented clinical profiles, plasma samples from ten with severe Covid-19 were selected, along with plasma from 10 with non-severe infection, for this study, as described in Wang This study was reviewed and approved by the Columbia University Institutional Review Board (protocol number AAAT0123) and deidentified viral samples were obtained under a waiver of consent. Sera and plasma was obtained from patients after informed consent.Note that full information on the approval of the study protocol must also be provided in the manuscript.