Articles Genomic evidence for reinfection with SARS-CoV-2: a case study Richard L Tillett, Joel R Sevinsky, Paul D Hartley, Heather Kerwin, Natalie Crawford, Andrew Gorzalski, Chris Laverdure, Subhash C Verma, Cyprian C Rossetto, David Jackson, Megan J Farrell, Stephanie Van Hooser, Mark Pandori Summary Background The degree of protective immunity conferred by infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is currently unknown. As such, the possibility of reinfection with SARS-CoV-2 is not well understood. We describe an investigation of two instances of SARS-CoV-2 infection in the same individual. Methods A 25-year-old man who was a resident of Washoe County in the US state of Nevada presented to health authorities on two occasions with symptoms of viral infection, once at a community testing event in April, 2020, and a second time to primary care then hospital at the end of May and beginning of June, 2020. Nasopharyngeal swabs were obtained from the patient at each presentation and twice during follow-up. Nucleic acid amplification testing was done to confirm SARS-CoV-2 infection. We did next-generation sequencing of SARS-CoV-2 extracted from nasopharyngeal swabs. Sequence data were assessed by two different bioinformatic methodologies. A short tandem repeat marker was used for fragment analysis to confirm that samples from both infections came from the same individual. Findings The patient had two positive tests for SARS-CoV-2, the first on April 18, 2020, and the second on June 5, 2020, separated by two negative tests done during follow-up in May, 2020. Genomic analysis of SARS-CoV-2 showed genetically significant differences between each variant associated with each instance of infection. The second infection was symptomatically more severe than the first. Interpretation Genetic discordance of the two SARS-CoV-2 specimens was greater than could be accounted for by short-term in vivo evolution. These findings suggest that the patient was infected by SARS-CoV-2 on two separate occasions by a genetically distinct virus. Thus, previous exposure to SARS-CoV-2 might not guarantee total immunity in all cases. All individuals, whether previously diagnosed with COVID-19 or not, should take identical precautions to avoid infection with SARS-CoV-2. The implications of reinfections could be relevant for vaccine development and application. Funding Nevada IDEA Network of Biomedical Research, and the National Institute of General Medical Sciences (National Institutes of Health). Copyright © 2020 Elsevier Ltd. All rights reserved. Introduction Methods Infection with severe acute respiratory syndrome corona­ virus 2 (SARS-CoV-2) leads to a detectable immune response, but the susceptibility of previously infected individuals to reinfection with SARS-CoV-2 is not well understood. SARS-CoV-2 infection results in generation of neutralising antibodies in patients.1 However, the degree to which this immune response indicates a protective immunity to subsequent infection with SARS-CoV-2 has not yet been elucidated. In studies of immunity to other coronaviruses,2–9 loss of immunity can occur within 1–3 years. Cases of primary illness due to infection followed by a discrete secondary infection or illness with the same biological agent can best be ascertained as distinct infection events by genetic analysis of the agents associated with each illness event. Reports of secondary infection events with SARS-CoV-2 have been published from Hong Kong,10 the Netherlands and Belgium,11 and Ecuador.12 We present a case report of an individual who had two distinct COVID-19 illnesses from genetically distinct SARS-CoV-2 agents. We present a case report of a 25-year-old male patient who was a resident of Washoe County in the US state of Nevada. The patient presented to a community testing event held by the Washoe County Health District on April 18, 2020. He had symptoms consistent with viral infection (sore throat, cough, headache, nausea, and diarrhoea), which had started on March 25, 2020 (figure 1). The patient had no history of clinically significant underlying conditions, and no indications of compromised immunity were identified. During isolation, the patient’s symptoms resolved (reported on April 27, 2020) and he continued to feel well until May 28, 2020. On May 31, 2020, the patient sought care at an urgent care centre with self-reported fever, headache, dizziness, cough, nausea, and diarrhoea, at which time chest radiography was done and he was discharged home. 5 days later (on June 5, 2020), the patient presented to a primary care doctor and was found to be hypoxic with shortness of breath. He was Case history www.thelancet.com/infection Published online October 12, 2020 https://doi.org/10.1016/S1473-3099(20)30764-7 Lancet Infect Dis 2020 Published Online October 12, 2020 https://doi.org/10.1016/ S1473-3099(20)30764-7 See Online/Comment https://doi.org/10.1016/ S1473-3099(20)30783-0 Nevada Institute of Personalized Medicine, University of Nevada, Las Vegas, NV, USA (R L Tillett PhD); University of Nevada, Reno Center for Bioinformatics, Reno, NV, USA (R L Tillett); Theiagen Consulting LLC, Highlands Ranch, CO, USA (J R Sevinsky PhD); Nevada Genomics Center, University of Nevada, Reno, NV, USA (P D Hartley PhD); Division of Epidemiology & Public Health Preparedness, Washoe County Health District, Reno, NV, USA (H Kerwin MPH); Renown Health, Reno, NV, USA (N Crawford MD); Nevada State Public Health Laboratory, Reno, NV, USA (A Gorzalski PhD, C Laverdure BS, S Van Hooser MBA MLS(ASCP), M Pandori PhD); Department of Microbiology and Immunology (S C Verma PhD, C C Rossetto PhD) and Department of Pathology and Laboratory Medicine (M Pandori), University of Nevada, Reno School of Medicine, Reno, NV, USA; and Forensic Science Division, Washoe County Sheriff’s Office, Reno, NV, USA (D Jackson MSc, M J Farrell BS) Correspondence to: Dr Mark Pandori, Department of Pathology and Laboratory Medicine, University of Nevada, Reno School of Medicine, Reno, NV 89557, USA mpandori@med.unr.edu 1 Articles Research in context Evidence before this study We searched PubMed, preprint servers (MedRxiv, BioRxiv, and SSRN), and general news channels (via Google search) from June 30 to Sept 9, 2020, for reports of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) reinfection, using keywords including “reinfection”, “SARS-CoV-2”, and “secondary infection”. We restricted our search to publications in English. Three reports of reinfection, with variable symptom severity on reinfection, have been published worldwide to date, supporting the possibly for SARS-CoV-2 reinfection. Added value of this study We present, to our knowledge, the first North American case of reinfection with SARS-CoV-2. A 25-year-old man, who was a resident of Washoe County in the US state of Nevada, had laboratory-confirmed SARS-CoV-2 infection in April, 2020, followed by secondary infection within a period of Symptom onset March 25 * Positive real-time RT-PCR Symptom resolution Negative TMA Negative real-time RT-PCR Symptom onset April 18 April 27 May 9 May 26 May 28 First infection * Positive Positive real-time IgM and IgG RT-PCR June 5 June 6 Reinfection Figure 1: Timeline of symptom onset, molecular diagnosis, and sequencing of specimens TMA=transcription-mediated amplification. *Sequenced specimens. instructed to go to the emergency department after provision of oxygen. This work was done under an emergency order by the Chief Medical Officer of the Division of Public and Behavioral Health for the State of Nevada. Ethics approval was waived by the University of Nevada, Reno Institutional Review Board. The patient provided written consent to publish this report. Procedures Specimens were obtained from the patient by naso­ pharyngeal swab at the community testing event, during the period of isolation and recovery, and on presentation to hospital. Swabs were transported to the Nevada State Public Health Laboratory (Reno, NV, USA) in either viral transport medium or Aptima Multiswab Transport Media (Hologic, San Diego, CA, USA). Specimens were trans­ ported on cold packs and stored by refrigeration (4–8°C) for no longer than 72 h before nucleic acid extraction and subsequent real-time RT-PCR. Nucleic acid extraction was done using Omega Biotek MagBind Viral DNA/RNA 96 Kit (Omega Bio-tek, Norcross, GA, USA), per manufacturer’s instructions and with an elution volume of 100 µL. Aliquots of eluted RNA underwent real-time RT-PCR with either the Taqpath COVID-19 2 around 6 weeks, in June, 2020. The second infection was symptomatically more severe than the first. Genomic analysis showed the two viral agents were genetically distinct. The patient’s immune reaction in vitro was not assessed and, thus, conclusions cannot be made about the duration or degree of immunity. Implications of all the available evidence Reinfection with SARS-CoV-2 has been reported in at least four individuals worldwide. Thus, previous exposure to SARS-CoV-2 does not necessarily translate to guaranteed total immunity. The implications of reinfections could be relevant for vaccine development and application. From a public health perspective, all individuals—whether previously diagnosed or not—must take identical precautions to prevent infection with SARS-CoV-2. Further work is needed to assess immune reactions in vitro after reinfection. Emergency Use Authorized (EUA) Multiplex Assay (ThermoScientific, Waltham, MA, USA; 10 µL aliquots) or the US Centers for Disease Control and Prevention (CDC) 2019-nCoV Real-Time RT-qPCR Diagnostic Panel (CDC, Atlanta, GA, USA; 5 µL aliquots). Specimens transported on Aptima Multiswab Transport Media were tested by transcription-mediated amplifi­ ca­ tion using the Aptima SARS-CoV-2 (Panther System) assay (Hologic, Marlborough, MA, USA). Assays were done according to their respective EUA procedures, unless otherwise indicated. For the Taqpath real-time RT-PCR test, the threshold for calling a specimen positive was reactivity of two of three target sequences, each with reactivity at a cycle threshold of less than 40·00. A positive or negative result on the Hologic Aptima assay was based on proprietary processes. Antibody testing was done with the Roche Elecsys Anti SARS-CoV-2 test (Roche Diagnostics, Indianapolis, IN, USA). For viral genomic sequencing, total RNA was extracted from nasopharyngeal swabs as described. 70 µL of extracted RNA was treated for 30 min at room temper­ ature with Qiagen DNase I (Qiagen, Germantown, MD, USA) and then cleaned and concentrated with silica spin columns (Qiagen RNeasy MinElute; Qiagen) with a 12 µL water elution. A portion (7 µL) of this RNA was annealed to an rRNA inhibitor (Qiagen FastSelect rRNA HMR; Qiagen) and then reverse-transcribed (cDNA) using random hexamers. The syn­thesised DNA was strandligated and isothermally amplified into micro­grams of DNA (Qiagen FX Single Cell RNA Library Kit; Qiagen). A portion (1 µg) of this amplified DNA was sheared and ligated to Illumina-compatible sequencing adapters, followed by six cycles of PCR amplifi­cation (KAPA HiFi HotStart Library Amplification Kit; Roche Sequencing and Life Science, Kapa Biosystems, Wilmington, MA, USA) to enrich for library molecules with adapters at both ends. Next, these sequencing libraries were enriched for a sequence specific to SARS-CoV-2 using biotinylated www.thelancet.com/infection Published online October 12, 2020 https://doi.org/10.1016/S1473-3099(20)30764-7 Articles oligonucleotide baits (myBaits Expert Virus, Arbor Biosciences, Ann Arbor, MI, USA). A further eight to 16 cycles of PCR were done after enrichment (98°C for 45 s, 98°C for 15 s, 60°C for 30 s, repeat for eight to 16 cycles, then 72°C for 60 s and 4°C to complete), and these SARS-CoV-2-enriched sequen­cing libraries were pooled and sequenced with an Illumina NextSeq 500 (Illumina, San Diego, CA USA) as paired-end 2 × 75 base pair reads using the NextSeq version 2.5 mid-output 150 cycle kit (Illumina). For bioinformatics analysis of the two SARS-CoV-2 agents (referred to herein as specimen A and specimen B), after sequencing of each library, FASTQ files were imported into CLC Genomics Workbench version 20.0.4 (Qiagen A/S, Vedbæk, Denmark) with the CLC Microbial Genomics Module, CLC Genome Finishing Module, and Biomedical Genomics Analysis. Briefly, reads were imported, trimmed, and mapped to National Center for Biot­ echnology Information SARS-CoV-2 ref­ er­ ence sequence MN908947.3. The alignment was refined using the InDels and Structural Variants module, fol­lowed by the Local Realignment module. Variants were identified by a minimum coverage of five reads, mini­mum count of five, and minimum frequency of 70·0%. To ascertain repeatability of results, a second bioinfor­ matics analysis was done using an independent process and open source tools. Potential rein­ fection sequence libraries were trimmed using Trim­momatic version 0.39, with the ILLUMINACLIP adapter-clipping setting 2:30:10:2:keep­BothReads. Sequence pairs were aligned to the SARS-CoV-2 reference genome (MN908947.3) using Bowtie 2 version 2.3.13 PCR optical duplicates were flagged using Picard MarkDuplicates in picard-slim version 2.22.5. Variants were called for both samples in concert using Freebayes version 1.0.2, with ploidy settings of 1, a mini­mum allele frequency of 0·70, and a minimum depth of five reads for any variant call. The genome sequence of each sample was constructed using coverage statistics from BBtools pileup.sh and applyvariants.sh version 38.86, whereby only variants supported by coverage of five or more reads were written to bcftools consensus version 1.10.2, and all positions supported by fewer than five reads, whether reference or alternative, were replaced with Ns.14 For phylogenetic analysis, the whole genome sequen­ces of the isolates (specimen A and specimen B) were compared with those of 171 contemporaneous sequences from Nevada,15 the SARS-CoV-2 reference strain (MN908947.3), and one sequence derived from iso­late USA-WA1/2020 (Bei Resources, Manassas, VA, USA). After trimming six 5′ uncalled bases (Ns) from specimen A and 98 Ns from specimen B, genomic sequences were aligned and related using NGPhylogeny.fr PhyML+SMS.16 Sequences were then first-aligned using MAFFT with automatic flavour selection.17 Informative regions were selected using Block Mapping and Gathering with Entropy, a sliding window size of 3, and maximum entropy of 0·5.18 Unrooted trees were constructed by PhyML with Smart Model Selection, the Akaike information criterion, and Subtree Pruning and Regrafting.19 Newick trees were visualised using Interactive Tree Of Live version 4 and rooted at the Wuhan reference strain.20 Major SARS-CoV-2 clade memberships were predicted using Nextclade. To confirm specimens A and B were from the same individual, the original swab specimens, transport media, and residual samples of extracted RNA supplied to the sequencing core facility underwent short tandem repeat (STR) analysis for identity comparison, by the Washoe County Sheriff’s Forensic Science Division (Reno, NV, USA). 2 µL of extracted DNA was quantified using the Quantifiler Trio DNA Quantification Kit (Applied Biosystems, Foster City, CA, USA) on the 7500 Real-Time PCR System and analysed with 7500 HID software version 1.3 (Applied Biosystems). Amplification of 24 GlobalFiler STR markers (Thermo Fisher Scientific, Waltham, MA, USA) was accomplished on the ProFlex PCR Instrument (Thermo Fisher Scientific) for 29 cycles. The 3500xL Genetic Analyzer (Applied Biosystems) was used for fragment analysis of the amplified STR marker regions in con­junction with HID Data Collection Software version 4.0.1 (Applied Biosystems) and Genemapper ID-X software version 1.6 (Thermo Fisher Scientific). Statistical interpretation of STR data was achieved using allele frequencies maintained in the National Institute of Standards and Technology population database.21 For more on Nextclade see https://clades.nextstrain.org Role of the funding source The funder had no role in study design, data collection, data analysis, data interpretation, or writing of the report. The corresponding author had full access to all data in the study and had final responsibility for the decision to submit for publication. Results The first nasopharygeal swab, obtained at the community screening event on April 18, 2020, was positive for SARS-CoV-2 on real-time RT-PCR testing. Two subsequent nucleic acid amplification tests obtained after resolution of symptoms were negative for SARS-CoV-2 RNA (table 1). The patient’s symptoms returned on May 28, 2020, and he was admitted to hospital on June 5, 2020, at which time a second nasopharyngeal swab was obtained and was positive for SARS-CoV-2 infection by real-time RT-PCR testing. The patient required ongoing oxygen support in hospital and reported symptoms that included myalgia, cough, and shortness of breath. Chest radiography showed development of patchy, bilateral, interstitial opacities suggestive of viral or atypical pneumonia. On June 6, 2020, the patient was tested for IgG and IgM against SARS-CoV-2 and positive results were obtained (figure 1). With two episodes of symptoms consistent with COVID-19, and two specimens positive for SARS-CoV-2 separated by a period of 48 days, in addition to resolution of symptoms and two non-reactive (negative) SARS-CoV-2 www.thelancet.com/infection Published online October 12, 2020 https://doi.org/10.1016/S1473-3099(20)30764-7 3 Articles Specimen A Specimen B April 18, 2020 May 9, 2020 May 26, 2020 June 5, 2020 June 6, 2020 Test methodology Real-time RT-PCR TMA Real-time RT-PCR Real-time RT-PCR Immunoassay (IgG and IgM antibody detection) Test result Positive Negative Negative Positive Positive Quantitative result Ct 35·24 RLU 299 ·· Ct 35·31 ·· TMA=transcription-mediated amplification. Ct=cycle threshold. RLU=relative light units. Table 1: Summary of laboratory results Coverage (reads) Allele Forward/ frequency (%) reverse balance* Average quality† Shared variants of specimens A and B versus reference genome 241C→T Specimen A 67 100% 0·37 35·6 Specimen B 6 100% 0·38 36·0 Specimen A 144 100% 0·48 35·6 Specimen B 55 0·26 35·4 0·42 35·6 0·19 35·5 0·40 35·7 0·43 35·6 1059C→T 92·7% 3037C→T See Online for appendix Specimen A 89 Specimen B 425 100% 99·8% 14408C→T‡ Specimen A 73 Specimen B 1145 100% 99·6% 23403A→G Specimen A 6859 99·9% 0·19 35·7 Specimen B 10 484 99·9% 0·46 35·6 0·45 35·2 0·48 35·4 25563G→T Specimen A 421 Specimen B 757 100% 99·1% Specimen A-specific variants versus reference genome 539C→T 141 99·3% 0·45 35·6 4113C→T 159 70·4% 0·38 35·6 7921A→G 182 98·9% 0·49 35·7 16741G→T 173 99·4% 0·47 35·6 Specimen B-specific variants versus reference genome 8140C→T 1046 85·0% 0·43 35·6 11102C→T 1713 99·9% 0·44 35·5 14407C→T‡ 1145 99·7% 0·43 35·6 15190G→C 139 90·6% 0·33 35·7 15981C→T 224 26013C→T 1415 29466C→T 86 100% 0·38 35·5 99·2% 0·38 35·5 98·8% 0·07 35·8 Reference genome was Wuhan Hu 1 (GenBank MN908947.3). *Ratio of forward to reverse reads covering the locus. †Phred score. Phred is a measure of base calling accuracy, a higher score indicates higher quality. A Phred score of 30 indicates a base-calling accuracy of 99·9%. ‡CLC Genomics classified this variant as a dinucleotide multinucleotide variant. The two variants have been split in this table for clarity. Table 2: Variants noted in specimens A and B compared with the reference genome 4 test results in between positive test results, nucleic acid sequencing was done of the viruses associated with the two positive tests. Illumina sequencing yielded 738 617 read pairs for the specimen obtained in April, 2020 (specimen A), and 1 410 885 read pairs for the specimen obtained in June, 2020 (specimen B). Sequence data indicated that specimen A was a member of clade 20C, because genomic sequence analysis identified five mutations (single nucleotide variants [SNVs]) that were hallmarks of the 20C clade (3037C→T, 14408C→T, 23403A→G, 1059C→T, and 25563G→T). Specimen B was also a member of clade 20C and presented the same five hallmark SNVs. Specimen A had five further SNVs compared with the reference genome. Specimen B showed six additional SNVs and a mutation at posi­­tion 14 407, adjacent to the SNV 14408C→T and recorded as a dinucleotide multinucleotide variant (MNV) at positions 14 407 and 14 408 of the genome. Six SNVs were shared between specimen A and specimen B (table 2). Specimen A had four additional SNVs not seen in specimen B, whereas specimen B had seven SNVs that were absent in specimen A. A visualisation of the relation of sequence data sets between specimens A and B is shown in figure 2. An additional three deletions and one insertion were noted in the sequence of specimen B relative to the reference genome (appendix p 2). These findings were confirmed by additional analyses of FASTQ files generated from specimens A and B (only the SNV at locus 4113 in specimen A was not verified). Predictions of insertions and deletions were less stable, with only the deletion at loci 2084 and the insertion at 6018 confirmed. The Freebayes analysis detected a deletion at 22 832 in specimen B that was not identified by the first sequence analysis (appendix p 3), but insertion and deletion predictions from short-read alignments are less reliable than are SNV predictions22 and are merely presented for completeness. Specimens A and B were among 171 samples obtained in the US state of Nevada between March 5 and June 5, 2020, and sequenced. Phylogenetic analysis showed the relatedness of specimens A and B to each other and their comparative distance among additional positive samples (figure 3).
To rule out the possibility of specimen mishandling, or mislabelling errors during RNA extractions, forensic identity testing was done to investigate the source and intermediate materials of specimens A and B. Analysis of each of the specimens, residual extractions, and aliquot residuals showed that that specimens A and B were derived from the same individual, with a one in 53·48 × 10²⁴ chance of the specimens being from different people. Discussion Our case report presents details of the first individual in North America to have symptomatic reinfection with SARS-CoV-2. Similar to observations with the reinfection case in Ecuador,12 our patient showed increased symptom www.thelancet.com/infection Published online October 12, 2020 https://doi.org/10.1016/S1473-3099(20)30764-7 S ORF1b EM N ORF10 ORF1a ORF6 ORF7a ORF8 MN908947.3 (reference genome) ORF3a Articles Specimen A read mapping 961 359 reads Specimen B read mapping 951 644 reads Specimen A variants Specimen B variants * 5000 10 000 15 000 20 000 25 000 Figure 2: Variant mapping of specimens A and B against the reference genome ORF1a and ORF1b encode replicase proteins. The other ORFs encode assembly proteins. ORF=open reading frame. S=spike. E=envelope. M=membrane. N=nucleocapsid. *Identifies variant 14 407 in specimen A and variants 14 407 and 14 408 in specimen B. severity in their second infection, whereas the cases from Belgium and the Netherlands11 and Hong Kong10 did not show a difference in severity of symptoms. The mechanisms that could account for a more severe secondary infection can only be speculated. First, a very high dose of virus might have led to the second instance of infection and induced more severe disease.23 Second, it is possible that reinfection was caused by a version of the virus that was more virulent, or more virulent in this patient’s context. Third, a mechanism of antibodydependent enhancement might be the cause, a means by which specific Fc-bearing immune cells become infected with virus by binding to specific antibodies. This mechanism has been seen previously with the beta­ coronavirus causing severe acute respiratory syndrome.24 In that case, the patient recovered and was discharged from hospital. The individual associated with these two SARS-CoV-2 infections had no immunological disorders that would imply facilitation of reinfection. They were not taking any immunosuppressive drugs. The individual was negative for HIV by antibody and RNA testing (data not shown) and had no obvious cell count abnormalities. The secondary positive case (reinfection) occurred simulta­neously to a positive case in a cohabitant (parent), who also provided a specimen on June 5, 2020, that was positive by nucleic acid amplification testing (transcription-mediated amplifi­ cation). Sequencing is underway on the co-habitant specimen to ascertain its potential role in reinfection. However, the positive specimen from the co-habitant was obtained and tested in the Hologic Aptima format, which did not align with the procedures established at our sequencing laboratory. Nevertheless, the co-habitant positive case provides a possible source for secondary exposure and reinfection of our patient. It is possible that we have reported a case of contin­ uous infection entailing deactivation and reactivation. However, for such a hypothesis to be true, a mutational rate of SARS-CoV-2 would be required that has not yet been recorded.25–28 Specimens A and B showed an extrapolated rate of SNV and MNV accumulation of 83·64 substitutions per year, a rate that greatly exceeds the currently observed rate of 23·12.28 However, even more importantly, the four substitutions noted in specimen A would have to revert to the ancestral genotype, and the odds of this reversion occurring are remote. Of course, if such an amount of base change did occur in that timeframe, the remarkable nature of specimens A and B would shift from a case of possible reinfection to one of high-rate evolution within an infected individual. Another alternative explanation for the observed differences in specimens A and B would be that of co-infection. In a co-infection hypothesis, the patient would have been infected with viruses of both genotypes at the time of sample collection. Such a hypothesis would then further require that the specimen B type virus be present, yet undetected in April, 2020, and then conversely, specimen A type virions become depleted before the June, 2020, sample collection date. Specimens A and B were both in clade 20C, which was the predominant major clade seen in northern Nevada at the time samples were obtained. Our survey of viruses in Nevada identified samples resembling each of the case genotypes.15 Although evidence exists that SARS-CoV-2 quasispecies exist at low and fluctuating frequencies in infected samples,29 whereby low-frequency (eg, 1%) SNVs could be seen in various samples from the same patient, this possible situation would not itself account for the genotype switch observed between the first infection and reinfection. Our findings have implications for the role of vaccination in response to COVID-19. If we have truly reported a case of reinfection, initial exposure to SARS-CoV-2 might not result in a level of immunity that www.thelancet.com/infection Published online October 12, 2020 https://doi.org/10.1016/S1473-3099(20)30764-7 5 Clade 19A Clade 19B Clade 20A Clade 20B Clade 20C A0033 A0058 A0059 A0105 A0112 A0079 4 A002 8 A002 32 A0 0 2 6 A00 8 7 A00 2 8 A00 16 A01 7 11 A0 7 00 A0 053 A0 066 A0 073 A0 122 A0 123 7 A0 16 1 A0 17 A0 Tree scale: 0·0001 Wuhan Hu 1 Patient's specimens A0 12 1 1 A0 24 A0 118 A0 075 1 A0 20 A0 068 0 A0 93 0 A0 76 0 A01 92 3 A01 9 8 A01 2 3 A00 8 69 A00 4 A011 3 9 A009 1 A0098 A0048 A0144 A0054 A0056 A0070 A0113 A0 A0 A0 099 A0 140 A0 06 5 A0 057 A0 097 0 A0 52 A0 141 0 A0 49 0 A01 71 3 A00 4 5 A00 5 3 A02 6 06 A018 3 A019 7 A019 8 A0199 A0194 A0203 A0018 A0022 A0129 0 08 0 A0 09 0 A 131 A0 162 A0 153 A0 133 A0 186 A0 76 1 A0 87 1 A0 81 A01 8 7 A01 8 0 A02 3 7 A01 2 A017 8 A014 A0175 A0174 Wuhan Hu 1 A0004 A0026 A0014 A0015 A0063 A0008 A001 3 A003 0 USA W A00 A1 6 A00 1 21 A00 A00 05 A0 16 0 A0 09 0 A0 06 0 A0 31 A0 010 A0 051 A0 017 A0 03 A0 04 9 11 4 5 0 04 A0 180 A0 027 A0 151 A0 025 A0 205 A0 23 0 A0 41 0 A0 42 0 A0 64 A00 52 A01 1 6 A01 0 6 A01 7 A014 6 A019 4 A020 A0201 A0200 A0143 A0046 A0047 A0045 A0192 A0210 A0108 A0114 A0145 A0146 A0095 4 A009 9 A001 3 A019 95 1 0 A en A cim Spe A0202 90 A01 89 1 A0 49 1 A0 65 1 A0 156 A0 154 A0 163 A0 155 A0 157 A0 164 A0 A0034 A0188 A0081 A0103 A0107 A0084 A0106 A0 1 1 1 A008 3 A0 0 86 A01 0 A00 4 8 A01 5 7 A01 7 8 A0 5 0 A0 20 0 Sp 60 A0 ecim A0 087 en B A0 137 A0 088 A0 136 A0 132 08 9 Articles Figure 3: Phylogenetic placement of specimens A and B within Nevada isolates, reference genomes, and global clades 171 sequences were from Nevada. Wuhan Hu 1 was the reference genome (GenBank MN908947.3). USA WA1 was the isolate USA-WA1/2020 (Bei Resources, Manassas, VA, USA). is 100% protective for all individuals. With respect to vaccination, this understanding is established, with influenza regularly showing the challenges of effective vaccine design.30 A major limitation of our case study is that we were unable to undertake any assessment of the immune response to the first episode of SARS-CoV-2 6 infection. We also could not assess fully the effectiveness of the immune responses (eg, neutralising antibody titres) during the second episode, when the individual was antibody-positive for total antibody assay to the SARS-CoV-2 nucleocapsid protein. If our patient is a case of natural viral evolution in vivo (although highly unlikely www.thelancet.com/infection Published online October 12, 2020 https://doi.org/10.1016/S1473-3099(20)30764-7 Articles in view of the requirement of four reversions to reference genotypes) then the implications of these data are that SARS-CoV-2 can adapt with enough genetic dexterity to avoid a natural immune response in a manner to re-establish detectable levels of infection in an individual. If our patient is a case of reinfection, it is crucial to note that the frequency of such an occurrence is not defined by one case study: this event could be rare. The absence of comprehensive genomic sequencing of positive cases in the USA and worldwide limits the advances in public health surveillance needed to find these cases. Certainly, limitations in screening and testing availability for SARS-CoV-2 exacerbate the poor surveillance efforts being undertaken not only to diagnose COVID-19 but also to obtain actionable genetic tracking of this agent. 8 Contributors RLT contributed to writing of the report and data analysis. JRS contributed to review and editing of the report and data analysis. PDH contributed to sequencing and analysis. HK contributed to public health intelligence and case identification. NC contributed to clinical data and clinical care. AG and CL contributed to diagnostics analysis. SCV and CCR contributed to writing and editing of the report and figure generation. DJ and MJF contributed to identity testing and confirmation. SVH contributed to diagnostics and laboratory management. MP had the idea for the study and contributed to diagnostics, formal analysis, and writing and editing of the report. 13 Declaration of interests JRS reports personal fees from Qiagen Digital Insights, outside of the submitted work. All other authors declare no competing interests. 17 Data sharing The CLC workflow, combined mapping report, parameters for CLC modules, FASTA-format sequences, the open-source workflow, BAM alignments, and VCF-format files are available online. 18 Acknowledgments We thank Nevada IDEA Network of Biomedical Research for funding this work. We acknowledge grants from the National Institute of General Medical Sciences (National Institutes of Health), which enabled publication of these findings (GM103440 and GM104944). We thank the Washoe County Health District and Washoe County Sheriff’s Department for helping to identify and confirm these findings. References 1 Ju B, Zhang Q, Ge J, et al. Human neutralizing antibodies elicited by SARS-CoV-2 infection. Nature 2020; 584: 115–19. 2 Callow KA, Parry HF, Sergeant M, Tyrrell DA. The time course of the immune response to experimental coronavirus infection of man. Epidemiol Infect 1990; 105: 435–46. 3 Chang S-C, Wang J-T, Huang L-M, et al. Longitudinal analysis of severe acute respiratory syndrome (SARS) coronavirus-specific antibody in SARS patients. Clin Diagn Lab Immunol 2005; 12: 1455–57. 4 Huang AT, Garcia-Carreras B, Hitchings MDT, et al. A systematic review of antibody mediated immunity to coronaviruses: antibody kinetics, correlates of protection, and association of antibody responses with severity of disease. medRxiv 2020; published online April 17. https://doi.org/10.1101/2020.04.14.20065771 (preprint). 5 Liu W, Fontanet A, Zhang P-H, et al. Two-year prospective study of the humoral immune response of patients with severe acute respiratory syndrome. J Infect Dis 2006; 193: 792–95. 6 Mo H, Zeng G, Ren X, et al. Longitudinal profile of antibodies against SARS-coronavirus in SARS patients and their clinical significance. Respirology 2006; 11: 49–53. 7 Reed SE. The behaviour of recent isolates of human respiratory coronavirus in vitro and in volunteers: evidence of heterogeneity among 229E-related strains. J Med Virol 1984; 13: 179–92. 9 10 11 12 14 15 16 19 20 21 22 23 24 25 26 27 28 29 30 Woo PCY, Lau SKP, Wong BHL, et al. Longitudinal profile of immunoglobulin G (IgG), IgM, and IgA antibodies against the severe acute respiratory syndrome (SARS) coronavirus nucleocapsid protein in patients with pneumonia due to the SARS coronavirus. Clin Diagn Lab Immunol 2004; 11: 665–68. Wu L-P, Wang N-C, Chang Y-H, et al. Duration of antibody responses after severe acute respiratory syndrome. Emerg Infect Dis 2007; 13: 1562–64. To KK-W, Hung IF-N, Ip JD, et al. COVID-19 re-infection by a phylogenetically distinct SARS-coronavirus-2 strain confirmed by whole genome sequencing. Clin Infect Dis 2020; published online Aug 25. https://doi.org/10.1093/cid/ciaa1275. Van Elslande J, Vermeersch P, Vandervoort K, et al. Symptomatic SARS-CoV-2 reinfection by a phylogenetically distinct strain. Clin Infect Dis 2020; published online Sept 5. https://doi. org/10.1093/cid/ciaa1330. Prado-Vivar B, Becerra-Wong M, Guadalupe JJ, et al. COVID-19 re-infection by a phylogenetically distinct SARS-CoV-2 variant, first confirmed event in South America. SSRN 2020; published online Sept 8. https://doi.org/10.2139/ssrn.3686174 (preprint). Langmead B, Salzberg SL. Fast gapped-read alignment with Bowtie 2. Nat Methods 2012; 9: 357–59. Li H. A statistical framework for SNP calling, mutation discovery, association mapping and population genetical parameter estimation from sequencing data. Bioinformatics 2011; 27: 2987–93. Hartley P, Tillett RL, Xu Y, et al. Genomic surveillance revealed prevalence of unique SARS-CoV-2 variants bearing mutation in the RdRp gene among Nevada patients. medRxiv 2020; published online Sept 11. https://doi.org/10.1101/2020.08.21.20178863 (preprint). Lemoine F, Correia D, Lefort V, et al. NGPhylogeny.fr: new generation phylogenetic services for non-specialists. Nucleic Acids Res 2019; 47: W260–65. Katoh K, Standley DM. MAFFT multiple sequence alignment software version 7: improvements in performance and usability. Mol Biol Evol 2013; 30: 772–80. Criscuolo A, Gribaldo S. BMGE (Block Mapping and Gathering with Entropy): a new software for selection of phylogenetic informative regions from multiple sequence alignments. BMC Evol Biol 2010; 10: 210. Lefort V, Longueville J-E, Gascuel O. SMS: Smart Model Selection in PhyML. Mol Biol Evol 2017; 34: 2422–24. Letunic I, Bork P. Interactive Tree Of Life (iTOL) v4: recent updates and new developments. Nucleic Acids Res 2019; 47: W256–59. National Institute of Standards and Technology. Population studies conducted by the NIST Forensics/Human Identity Project Team. Sept 23, 2014. https://strbase.nist.gov/NISTpop.htm (accessed Sept 22, 2020). Abnizova I, te Boekhorst R, Orlov YL. Computational errors and biases in short read next generation sequencing. J Proteomics Bioinform 2017; published online Jan 26. https://doi.org/10.4172/jpb.1000420. Guallar MP, Meiriño R, Donat-Vargas C, Corral O, Jouvé N, Soriano V. Inoculum at the time of SARS-CoV-2 exposure and risk of disease severity. Int J Infect Dis 2020; 97: 290–92. Yip MS, Leung NH, Cheung CY, et al. Antibody-dependent infection of human macrophages by severe acute respiratory syndrome coronavirus. Virol J 2014; 11: 82. Hill V, Rambaut A. Phylogenetic analysis of SARS-CoV-2 genomes. March 6, 2020. https://virological.org/t/phylodynamic-analysis-ofsars-cov-2-update-2020-03-06/420 (accessed Sept 23, 2020). Mercatelli D, Giorgi FM. Geographic and genomic distribution of SARS-CoV-2 mutations. Front Microbiol 2020; 11: 1800. Pachetti M, Marini B, Benedetti F, et al. Emerging SARS-CoV-2 mutation hot spots include a novel RNA-dependent-RNA polymerase variant. J Transl Med 2020; 18: 179. Hadfield J, Megill C, Bell SM, et al. Genomic epidemiology of novel coronavirus: global subsampling. Sept 16, 2020. https://nextstrain. org/ncov/global?c=region&l=clock (accessed Sept 23, 2020). Jary A, Leducq V, Malet I, et al. Evolution of viral quasispecies during SARS-CoV-2 infection. Clin Microbiol Infect 2020; published online July 24. https://doi.org/10.1016/j.cmi.2020.07.032. Osterholm MT, Kelley NS, Sommer A, Belongia EA. Efficacy and effectiveness of influenza vaccines: a systematic review and meta-analysis. Lancet Infect Dis 2012; 12: 36–44. www.thelancet.com/infection Published online October 12, 2020 https://doi.org/10.1016/S1473-3099(20)30764-7 For shared resources see https://doi.org/10.5281/ zenodo.3988782 7