Protection afforded by the BNT162b2 and mRNA-1273 COVID-19 vaccines in fully vaccinated cohorts with and without prior infection ================================================================================================================================ * Laith J. Abu-Raddad * Hiam Chemaitelly * Houssein H. Ayoub * Hadi M. Yassine * Fatiha M. Benslimane * Hebah A. Al Khatib * Patrick Tang * Mohammad R. Hasan * Peter Coyle * Zaina Al Kanaani * Einas Al Kuwari * Andrew Jeremijenko * Anvar Hassan Kaleeckal * Ali Nizar Latif * Riyazuddin Mohammad Shaik * Hanan F. Abdul Rahim * Gheyath K. Nasrallah * Mohamed Ghaith Al Kuwari * Adeel A. Butt * Hamad Eid Al Romaihi * Mohamed H. Al-Thani * Abdullatif Al Khal * Roberto Bertollini ## Abstract Effect of prior SARS-CoV-2 infection on vaccine protection remains poorly understood. Here, we investigated whether persons vaccinated after a prior infection have better protection against future infection than those vaccinated without prior infection. Effect of prior infection was assessed in Qatar’s population, where the Alpha (B.1.1.7) and Beta (B.1.351) variants dominate incidence, using two national retrospective, matched-cohort studies, one for the BNT162b2 (Pfizer-BioNTech) vaccine, and one for the mRNA-1273 (Moderna) vaccine. Incidence rates of infection among BNT162b2-vaccinated persons, with and without prior infection, were estimated, respectively, at 1.66 (95% CI: 1.26-2.18) and 11.02 (95% CI: 9.90-12.26) per 10,000 person-weeks. The incidence rate ratio was 0.15 (95% CI: 0.11-0.20). Analogous incidence rates among mRNA-1273-vaccinated persons were estimated at 1.55 (95% CI: 0.86-2.80) and 1.83 (95% CI: 1.07-3.16) per 10,000 person-weeks. The incidence rate ratio was 0.85 (95% CI: 0.34-2.05). Prior infection enhanced protection of those BNT162b2-vaccinated, but not those mRNA-1273-vaccinated. These findings may have implications for dosing, interval between doses, and potential need for booster vaccination. Keywords * SARS-CoV-2 * COVID-19 * prior infection * vaccine * cohort study * immunity * epidemiology ## Main text Effect of prior acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection on vaccine protection against acquisition of infection remains poorly understood1-3. Qatar launched Coronavirus Disease 2019 (COVID-19) immunization in December 21, 2020, first using the BNT162b24 (Pfizer-BioNTech) vaccine and subsequently adding the mRNA-12735 (Moderna) vaccine6,7. As vaccination was scaled up following the FDA-approved protocol, the country experienced two back-to-back SARS-CoV-2 waves from January-June, 2021, which were dominated by the Alpha8 (B.1.1.7) and Beta8 (B.1.351) variants6,7,9-11 (Methods). This provided an opportunity to assess whether persons vaccinated after a prior SARS-CoV-2 infection have better protection against future infection than those vaccinated without prior infection. Leveraging the national, federated databases that have captured all SARS-CoV-2 vaccinations and PCR testing since the epidemic onset (Methods), we investigated this question using two retrospective, matched-cohort studies. We compared incidence of documented SARS-CoV-2 infection in the national cohort of individuals who completed ≥14 days after the second BNT162b2 vaccine dose, but who had experienced a prior PCR-confirmed infection, with incidence among individuals who completed ≥14 days after the second BNT162b2 dose, but who had not experienced a prior infection, between December 21, 2020-June 6, 2021 (Figure 1). The same comparison was made for the mRNA-1273 vaccine (Figure 2). Cohorts were matched in a 1:1 ratio by sex, 5-year age group, nationality, and calendar week of the first vaccine dose, to control for differences in exposure risk12,13 and variant exposure6,7,9-11. Reporting of the study followed the STROBE guidelines (Supplementary Table 1). ![Figure 1.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2021/07/26/2021.07.25.21261093/F1.medium.gif) [Figure 1.](http://medrxiv.org/content/early/2021/07/26/2021.07.25.21261093/F1) Figure 1. **The process for identifying SARS-CoV-2 infections in the national cohort of individuals who completed ≥14 days after the second BNT162b2 vaccine dose and who had experienced a PCR-confirmed infection before the first dose, compared with the process for identifying SARS-CoV-2 infections in the national cohort of individuals who completed ≥14 days after the second BNT162b2 vaccine dose, but who had experienced no PCR-confirmed infection before the first dose. Cohorts were matched in a 1:1 ratio by sex, 5-year age group, nationality, and calendar week of the first vaccine dose. Total follow-up time among BNT162b2-vaccinated persons, with and without prior infection, was 308,086.0 and 305,891.9 person-weeks, respectively.** ![Figure 2.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2021/07/26/2021.07.25.21261093/F2.medium.gif) [Figure 2.](http://medrxiv.org/content/early/2021/07/26/2021.07.25.21261093/F2) Figure 2. **The process for identifying SARS-CoV-2 infections in the national cohort of individuals who completed ≥14 days after the second mRNA-1273 vaccine dose and who had experienced a PCR-confirmed infection before the first dose, compared with the process for identifying SARS-CoV-2 infections in the national cohort of individuals who completed ≥14 days after the second mRNA-1273 vaccine dose, but who had experienced no PCR-confirmed infection before the first dose. Cohorts were matched in a 1:1 ratio by sex, 5-year age group, nationality, and calendar week of the first vaccine dose. Total follow-up time among mRNA-1273-vaccinated persons, with and without prior infection, was 70,729.9 and 70,872 person-weeks, respectively.** ![Figure 3.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2021/07/26/2021.07.25.21261093/F3.medium.gif) [Figure 3.](http://medrxiv.org/content/early/2021/07/26/2021.07.25.21261093/F3) Figure 3. **Kaplan-Meier curves showing the cumulative incidence of documented SARS-CoV-2 infection in the national cohort of individuals who completed ≥14 days after the second vaccine dose and who had a prior PCR-confirmed infection, compared to the cumulative incidence of documented SARS-CoV-2 infection in the matched national cohort of individuals who completed ≥14 days after the second vaccine dose, but without prior PCR-confirmed infection. The curves compare vaccination with A) the BNT162b2 (Pfizer-BioNTech) vaccine and B) the mRNA-1273 vaccine. Cohorts were matched in a 1:1 ratio by sex, 5-year age group, nationality, and calendar week of the first vaccine dose. The curves for a longer time of follow up for only the BNT162b2 vaccine are in Supplementary Figure 1. Vaccination with BNT162b2 started few weeks before vaccination with mRNA-1273.** Figures 1-2 show the process for identifying infections in these cohorts, and Table 1 presents their demographic characteristics. Using the Kaplan–Meier estimator14, cumulative infection incidence among BNT162b2-vaccinated persons, with and without prior infection, was estimated at 0.14% (95% CI: 0.11-0.19%) and 0.93% (95% CI: 0.83-1.04%), respectively, after 63 days of follow-up (Figure 1). Incidence rates of infection were estimated, respectively, at 1.66 (95% CI: 1.26-2.18) and 11.02 (95% CI: 9.90-12.26) per 10,000 person-weeks. The incidence rate ratio was estimated at 0.15 (95% CI: 0.11-0.20). View this table: [Table 1.](http://medrxiv.org/content/early/2021/07/26/2021.07.25.21261093/T1) Table 1. Demographic characteristics of matched cohorts that received the BNT162b2 and mRNA-1273 vaccines. Cumulative infection incidence among mRNA-1273-vaccinated persons, with and without prior infection, was estimated at 0.06% (95% CI: 0.03-0.12%) and 0.08% (95% CI: 0.04-0.15%), respectively, after 63 days of follow-up (Figure 1). Incidence rates were estimated, respectively, at 1.55 (95% CI: 0.86-2.80) and 1.83 (95% CI: 1.07-3.16) per 10,000 person-weeks. The incidence rate ratio was estimated at 0.85 (95% CI: 0.34-2.05). Infection incidence was low in these cohorts during a time of intense incidence in Qatar6,7,15, indicating that both vaccines were highly effective against the Alpha and Beta variants6,7, which dominated incidence9 (Methods). Still, prior infection of those BNT162b2-vaccinated further enhanced protection and reduced the incidence rate by 85% (6.6-fold) compared to those without prior infection. No evidence for such an effect was found for those mRNA-1273-vaccinated. These findings are perhaps explained by the observed differences in effectiveness of these two vaccines against the Alpha and Beta variants, estimated in Qatar at 89.5% (95% CI: 85.9-92.3%) and 75.0% (95% CI: 70.5-78.9%) for BNT162b2, respectively6, and at 100% (95% CI: 91.8-100.0%) and 96.4% (95% CI: 91.9-98.7%) for mRNA-1273, respectively7. The differences in effectiveness could have risen for a variety of reasons, such as differences in dosing, interval between doses, or the biology of both vaccines and their mechanisms of action. The dose of each of these two vaccines differed—it was 30-μg per dose for BNT162b24 and 100 μg per dose for mRNA-12735. This may have resulted in a more activated immune response for the mRNA-1273 vaccine than the BNT162b2 vaccine, and made the existence of prior immunity due to natural infection of no additional benefit for the mRNA-1273 vaccine. The interval between doses also differed and was one week longer for mRNA-12735. Evidence suggests that a longer dose interval could be associated with improved protection after receiving the second dose16. Limitations include identifying prior infection based on a record of a PCR-positive result, thereby missing those who may have been infected, but were unaware of their infection, or who did not seek testing by PCR to document the infection. Misclassification of prior infection status could lead to underestimation of the effect size of prior infection on vaccine protection. Depletion of the cohorts with prior infection due to COVID-19 mortality at time of the prior infection may have biased these cohorts toward healthier individuals with stronger immune responses. However, COVID-19 mortality has been low in Qatar’s predominantly young and working-age population12,17, and no evidence for such bias was found in the mRNA-1273 vaccine results, where the incidence rate was similar for those with and without prior infection. We assessed risk of only documented infections, but other infections may have occurred and gone undocumented, perhaps because of minimal/mild or no symptoms. Our cohorts predominantly included working-age adults; therefore, results may not necessarily be generalizable to other population groups, such as children or the elderly. Matching was done for age, sex, nationality, and calendar week of the first vaccine dose, and could not be done for other factors, such as comorbidities or additional socio-demographic factors, as these were not available to study investigators. However, matching by age and sex may have served as a proxy given that co-morbidities are associated with older age and may be different between women and men. Matching by nationality may have also captured some of the occupational risk given the distribution of the labor force in Qatar18-20. Imperfect assay sensitivity and specificity of PCR or antibody testing could have affected current or prior infection ascertainment. However, all PCR and serological testing was performed with extensively used, investigated, and validated commercial platforms with essentially 100% sensitivity and specificity (Methods). Unlike blinded, randomized clinical trials, the investigated observational cohorts were neither blinded nor randomized. Our results demonstrate low infection incidence among those vaccinated with BNT162b2 or mRNA-1273, but among those vaccinated with BNT162b2, protection against infection was further enhanced and infection incidence was further reduced by prior infection. In contrast, those vaccinated with mRNA-1273 were as well protected as those who received the vaccine after a prior infection. These findings may have implications for the potential need of a booster vaccination. ## Data Availability The dataset of this study is a property of the Qatar Ministry of Public Health that was provided to the researchers through a restricted-access agreement that prevents sharing the dataset with a third party or publicly. Future access to this dataset can be considered through a direct application for data access to Her Excellency the Minister of Public Health ([https://www.moph.gov.qa/english/Pages/default.aspx](https://www.moph.gov.qa/english/Pages/default.aspx)). Aggregate data are available within the manuscript and its Supplementary information. ## Author contributions LJA conceived and co-designed the study, led the statistical analyses, and co-wrote the first draft of the article. HC co-designed the study, performed the statistical analyses, and co-wrote the first draft of the article. All authors contributed to data collection and acquisition, database development, discussion and interpretation of the results, and to the writing of the manuscript. All authors have read and approved the final manuscript. ## Competing interests Dr. Butt has received institutional grant funding from Gilead Sciences unrelated to the work presented in this paper. Otherwise, we declare no competing interests. ## Methods ### Data sources and study design Analyses were conducted using the centralized, integrated, and standardized national severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) databases compiled at Hamad Medical Corporation (HMC), the main public healthcare provider and the nationally designated provider for all Coronavirus Disease 2019 (COVID-19) healthcare needs. Through a nation-wide digital health information platform, these databases have captured all SARS-CoV-2-related data along with related-demographic details with no missing information since the start of the epidemic, including all records of polymerase chain reaction (PCR) testing, antibody testing, COVID-19 hospitalizations, vaccinations, infection severity classification per World Health Organization (WHO) guidelines21 (performed by trained medical personnel through individual chart reviews), and COVID-19 deaths, also assessed per WHO guidelines22. Every PCR test conducted in Qatar, regardless of location (outpatient clinic, drive-thru, or hospital, etc.), is classified on the basis of symptoms and the reason for testing (clinical symptoms, contact tracing, random testing campaigns (surveys), individual requests, routine healthcare testing, pre-travel, and port of entry). Qatar has unique demographics by sex and nationality, since expatriates from over 150 countries comprise 89% of the population12,23. The nature of circulating SARS-CoV-2 virus was informed by weekly rounds of viral genome sequencing and multiplex, quantitative, reverse-transcription PCR (RT-qPCR) variant screening24 of randomly collected clinical samples6,7,9-11, as well as by the results of deep sequencing of wastewater samples9. The weekly rounds of viral genome sequencing from January 1-May 19, 2021 identified Beta (n=623; 50.9%), Alpha (n=193; 15.8%), Delta (n=43; 3.5%), and wild-type/undetermined variants (n=366; 29.9%) in 1,225 randomly collected, PCR-positive specimens9,10. Meanwhile, the weekly rounds of multiplex RT-qPCR variant screening from March 23-May 10, 2021 identified Beta-like (n=2,605; 66.4%), Alpha-like (n=970; 24.7%), and “other” variants (n=349; 8.9%) in 3,924 randomly collected PCR-positive specimens9,11. Sanger sequencing of the receptor binding domain of SARS-CoV-2 spike protein on 109 “other” specimens confirmed that 103 were Delta-like, 3 were B.1-like, and 3 were undetermined9,11. All records of PCR testing in Qatar were examined in this study. Every individual that met the inclusion criteria in the national database, that is being vaccinated with BNT162b2 or mRNA-1273 and completing ≥14 days after the second vaccine dose, for each of these cohort studies, was classified based on infection status (with or without PCR-positive swab before the start of the study). Individuals were matched based on infection status on a 1:1 ratio by sex, 5-year age group, nationality (>75 nationality groups), and calendar week of first vaccine dose to control for differences in exposure risk12,13 and variant exposure6,7,9-11. Only matched samples were included in the analysis. Further background on Qatar’s epidemic, such as on reinfections25,26, national seroprevalence surveys12,18-20, PCR surveys12, and other epidemiological studies can be found in previous publications on this epidemic6,7,12,13,27-34. ### Laboratory methods Nasopharyngeal and/or oropharyngeal swabs (Huachenyang Technology, China) were collected for PCR testing and placed in Universal Transport Medium (UTM). Aliquots of UTM were: extracted on a QIAsymphony platform (QIAGEN, USA) and tested with real-time reverse-transcription PCR (RT-qPCR) using TaqPath™ COVID-19 Combo Kits (100% sensitivity and specificity35; Thermo Fisher Scientific, USA) on an ABI 7500 FAST (ThermoFisher, USA); extracted using a custom protocol36 on a Hamilton Microlab STAR (Hamilton, USA) and tested using AccuPower SARS-CoV-2 Real-Time RT-PCR Kits (100% sensitivity and specificity37; Bioneer, Korea) on an ABI 7500 FAST; or loaded directly into a Roche cobas® 6800 system and assayed with a cobas® SARS-CoV-2 Test (95% sensitivity, 100% specificity38; Roche, Switzerland). The first assay targets the viral S, N, and ORF1ab regions. The second targets the viral RdRp and E-gene regions, and the third targets the ORF1ab and E-gene regions. Antibodies against SARS-CoV-2 in serological samples were detected using a Roche Elecsys® Anti-SARS-CoV-2 assay (99.5% sensitivity39, 99.8% specificity39,40; Roche, Switzerland), an electrochemiluminescence immunoassay that uses a recombinant protein representing the nucleocapsid (N) antigen for antibody binding. Results were interpreted according to the manufacturer’s instructions (reactive: optical density (proxy for antibody titer41) cutoff index ≥1.0 vs. non-reactive: optical density cutoff index <1.0). All PCR tests were conducted at the Hamad Medical Corporation Central Laboratory or Sidra Medicine Laboratory, following standardized protocols. ### Statistical analysis Descriptive statistics (frequency distributions and measures of central tendency) were used to characterize study samples. Significant associations were determined using two-sided p-values. The Kaplan–Meier estimator method14 was used to estimate the cumulative risk of documented infection. Cumulative risk was defined as the proportion of individuals identified with an infection during the study period among all eligible individuals in each cohort. Incidence rates of documented infection in each cohort were calculated by dividing the number of infection cases identified during the study by the number of person-weeks contributed by all eligible individuals in the cohort. Incidence rates and corresponding 95% CIs were estimated using a Poisson log-likelihood regression model with the STATA 17.042 *stptime* command. Follow-up person-time was calculated from the day each person completed 14 days after the second vaccine dose up to the infection swab, all-cause death, or end-of-study censoring (June 6, 2021). The incidence rate ratio and corresponding 95% CI were calculated using the exact method. Statistical analyses were conducted in STATA/SE version 17.042. ## Ethical approvals The study was approved by the Hamad Medical Corporation and Weill Cornell Medicine-Qatar Institutional Review Boards with waiver of informed consent. ## Data availability The dataset of this study is a property of the Qatar Ministry of Public Health that was provided to the researchers through a restricted-access agreement that prevents sharing the dataset with a third party or publicly. Future access to this dataset can be considered through a direct application for data access to Her Excellency the Minister of Public Health ([https://www.moph.gov.qa/english/Pages/default.aspx](https://www.moph.gov.qa/english/Pages/default.aspx)). Aggregate data are available within the manuscript and its Supplementary information. ## Supplementary Material View this table: [Supplementary Table 1.](http://medrxiv.org/content/early/2021/07/26/2021.07.25.21261093/T2) Supplementary Table 1. STROBE checklist for cohort studies. ![Supplementary Figure 1.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2021/07/26/2021.07.25.21261093/F4.medium.gif) [Supplementary Figure 1.](http://medrxiv.org/content/early/2021/07/26/2021.07.25.21261093/F4) Supplementary Figure 1. **Kaplan-Meier curves showing the cumulative incidence of documented SARS-CoV-2 infection in the national cohort of individuals who completed ≥14 days after the second vaccine dose and who had a prior PCR-confirmed infection, compared to the cumulative incidence of documented SARS-CoV-2 infection in the matched national cohort of individuals who completed ≥14 days after the second vaccine dose, but without prior PCR-confirmed infection. The curves compare vaccination with A) the BNT162b2 (Pfizer-BioNTech) vaccine and B) the mRNA-1273 vaccine. Cohorts were matched in a 1:1 ratio by sex, 5-year age group, nationality, and calendar week of the first vaccine dose. The cumulative infection incidence among the BNT162b2-vaccinated persons, with and without prior infection, was estimated at 0.16% (95% CI: 0.11-0.23%) and 1.45% (95% CI: 1.20-1.76%), respectively, after 132 days of follow-up.** ## Acknowledgements We acknowledge the many dedicated individuals at Hamad Medical Corporation, the Ministry of Public Health, the Primary Health Care Corporation, and the Qatar Biobank for their diligent efforts and contributions to make this study possible. The authors are grateful for support from the Biomedical Research Program, the Biostatistics, Epidemiology, and Biomathematics Research Core, and the Genomics Core, all at Weill Cornell Medicine-Qatar, as well as for support provided by the Ministry of Public Health and Hamad Medical Corporation. The authors are also grateful for the Qatar Genome Programme for supporting the viral genome sequencing. The funders of the study had no role in study design, data collection, data analysis, data interpretation, or writing of the article. Statements made herein are solely the responsibility of the authors. * Received July 25, 2021. * Revision received July 25, 2021. * Accepted July 26, 2021. * © 2021, Posted by Cold Spring Harbor Laboratory The copyright holder for this pre-print is the author. All rights reserved. The material may not be redistributed, re-used or adapted without the author's permission. ## References 1. 1.Reynolds, C.J., et al. Prior SARS-CoV-2 infection rescues B and T cell responses to variants after first vaccine dose. Science (2021). 2. 2.Muena, N.A., et al. Long-lasting neutralizing antibody responses in SARS-CoV-2 seropositive individuals are robustly boosted by immunization with the CoronaVac and BNT162b2 vaccines. medRxiv (2021). 3. 3.Letizia, A.G., et al. SARS-CoV-2 seropositivity and subsequent infection risk in healthy young adults: a prospective cohort study. Lancet Respir Med 9, 712–720 (2021). 4. 4.Polack, F.P., et al. Safety and Efficacy of the BNT162b2 mRNA Covid-19 Vaccine. N Engl J Med (2020). 5. 5.Baden, L.R., et al. Efficacy and Safety of the mRNA-1273 SARS-CoV-2 Vaccine. N Engl J Med 384, 403–416 (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa2035389&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F07%2F26%2F2021.07.25.21261093.atom) 6. 6.Abu-Raddad, L.J., Chemaitelly, H., Butt, A.A. & National Study Group for Covid-19 Vaccination. Effectiveness of the BNT162b2 Covid-19 Vaccine against the B.1.1.7 and B.1.351 Variants. N Engl J Med (2021). 7. 7.Chemaitelly, H., et al. mRNA-1273 COVID-19 vaccine effectiveness against the B.1.1.7 and B.1.351 variants and severe COVID-19 disease in Qatar. Nat Med (2021). 8. 8.World Health Organization. Tracking SARS-CoV-2 variants. Available from: [https://www.who.int/en/activities/tracking-SARS-CoV-2-variants/](https://www.who.int/en/activities/tracking-SARS-CoV-2-variants/). Accessed on: June 5, 2021. (2021). 9. 9.National Project of Surveillance for Variants of Concern and Viral Genome Sequencing. Qatar viral genome sequencing data. Data on randomly collected samples. [https://www.gisaid.org/phylodynamics/global/nextstrain/](https://www.gisaid.org/phylodynamics/global/nextstrain/). (2021). 10. 10.Benslimane, F.M., et al. One year of SARS-CoV-2: Genomic characterization of COVID-19 outbreak in Qatar. medRxiv, 2021.2005.2019.21257433 (2021). 11. 11.Hasan, M.R., et al. Real-Time SARS-CoV-2 Genotyping by High-Throughput Multiplex PCR Reveals the Epidemiology of the Variants of Concern in Qatar. medRxiv, 2021.2007.2018.21260718 (2021). 12. 12.Abu-Raddad, L.J., et al. Characterizing the Qatar advanced-phase SARS-CoV-2 epidemic. Scientific Reports 11, 6233 (2021). 13. 13.Ayoub, H.H., et al. Mathematical modeling of the SARS-CoV-2 epidemic in Qatar and its impact on the national response to COVID-19. J Glob Health 11, 05005 (2021). 14. 14.Kaplan, E.L. & Meier, P. Nonparametric estimation from incomplete observations. The Journal of the American Statistical Association 53, 457–481 (1958). 15. 15.Chemaitelly, H., et al. Reinfections with the SARS-CoV-2 B.1.351 variant and efficacy of natural immunity against reinfection under review at New England Journal of Medicine (2021). 16. 16.Voysey, M., et al. Single-dose administration and the influence of the timing of the booster dose on immunogenicity and efficacy of ChAdOx1 nCoV-19 (AZD1222) vaccine: a pooled analysis of four randomised trials. Lancet 397, 881–891 (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/s0140-6736(21)00432-3&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=33617777&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F07%2F26%2F2021.07.25.21261093.atom) 17. 17.Seedat, S., et al. SARS-CoV-2 infection hospitalization, severity, criticality, and fatality rates. medRxiv 2020.11.29.20240416 (non-peer-reviewed preprint) (2020). 18. 18.Jeremijenko, A., et al. Herd Immunity against Severe Acute Respiratory Syndrome Coronavirus 2 Infection in 10 Communities, Qatar. Emerg Infect Dis 27, 1343–1352 (2021). 19. 19.Al-Thani, M.H., et al. SARS-CoV-2 infection is at herd immunity in the majority segment of the population of Qatar. Open Forum Infectious Diseases (2021). 20. 20.Coyle, P.V., et al. SARS-CoV-2 seroprevalence in the urban population of Qatar: An analysis of antibody testing on a sample of 112,941 individuals. iScience, 102646 (2021). 21. 21.World Health Organization. COVID-19 clinical management: living guidance. Available from: [https://www.who.int/publications/i/item/WHO-2019-nCoV-clinical-2021-1](https://www.who.int/publications/i/item/WHO-2019-nCoV-clinical-2021-1). Accessed on: May 15, 2021. (2021). 22. 22.World Health Organization. International guidelines for certification and classification (coding) of COVID-19 as cause of death. Available from: [https://www.who.int/classifications/icd/Guidelines\_Cause\_of\_Death\_COVID-19-20200420-EN.pdf?ua=1](https://www.who.int/classifications/icd/Guidelines\_Cause_of_Death_COVID-19-20200420-EN.pdf?ua=1). Document Number: WHO/HQ/DDI/DNA/CAT. Accessed on May 15, 2021. (2020). 23. 23.Planning and Statistics Authority-State of Qatar. Qatar Monthly Statistics. Available from: [https://www.psa.gov.qa/en/pages/default.aspx](https://www.psa.gov.qa/en/pages/default.aspx). Accessed on: May 26, 2020. (2020). 24. 24.Vogels, C., Fauver, J. & Grubaugh, N. Multiplexed RT-qPCR to screen for SARS-COV-2 B.1.1.7, B.1.351, and P.1 variants of concern V.3. dx.doi.org/10.17504/protocols.io.br9vm966. (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.17504/protocols.io.br9vm966&link_type=DOI) 25. 25.Abu-Raddad, L.J., et al. Assessment of the risk of SARS-CoV-2 reinfection in an intense re-exposure setting. Clin Infect Dis (2020). 26. 26.Abu-Raddad, L.J., et al. SARS-CoV-2 antibody-positivity protects against reinfection for at least seven months with 95% efficacy. EClinicalMedicine 35, 100861 (2021). 27. 27.Al Kuwari, H.M., et al. Epidemiological investigation of the first 5685 cases of SARS-CoV-2 infection in Qatar, 28 February-18 April 2020. BMJ Open 10, e040428 (2020). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1136/bmjopen-2020-040428&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.ncbi.nlm.nih.gov/pubmed/33033033&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F07%2F26%2F2021.07.25.21261093.atom) 28. 28.Ayoub, H.H., et al. Epidemiological impact of prioritising SARS-CoV-2 vaccination by antibody status: mathematical modelling analyses. BMJ Innovations, bmjinnov-2021-000677 (2021). 29. 29.Butt, A.A., et al. Hospital admission rates, length of stay, and in-hospital mortality for common acute care conditions in COVID-19 vs. pre-COVID-19 era. Public Health 189, 6–11 (2020). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.puhe.2020.09.010&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=33126120&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F07%2F26%2F2021.07.25.21261093.atom) 30. 30.Saththasivam, J., et al. COVID-19 (SARS-CoV-2) outbreak monitoring using wastewater-based epidemiology in Qatar. Sci Total Environ 774, 145608 (2021). 31. 31.Seedat, S., et al. SARS-CoV-2 infection hospitalization, severity, criticality, and fatality rates. medRxiv 2020.11.29.20240416 (2020). 32. 32.Abu-Raddad, L.J., et al. Two prolonged viremic SARS-CoV-2 infections with conserved viral genome for two months. Infect Genet Evol 88, 104684 (2020). 33. 33.Abu-Raddad, L.J., et al. Pfizer-BioNTech mRNA BNT162b2 Covid-19 vaccine protection against variants of concern after one versus two doses. J Travel Med (2021). 34. 34.Nasrallah, G.K., et al. Analytic comparison between three high-throughput commercial SARS-CoV-2 antibody assays reveals minor discrepancies in a high-incidence population. Sci Rep 11, 11837 (2021). 35. 35.Thermo Fisher Scientific. TaqPath™ COVID-19 CE-IVD RT-PCR Kit instructions for use. Available from: [https://assets.thermofisher.com/TFS-Assets/LSG/manuals/MAN0019215\_TaqPathCOVID-19\_CE-IVD\_RT-PCR%20Kit\_IFU.pdf](https://assets.thermofisher.com/TFS-Assets/LSG/manuals/MAN0019215\_TaqPathCOVID-19_CE-IVD_RT-PCR%20Kit_IFU.pdf). Accessed on December 02, 2020. (2020). 36. 36.Kalikiri, M.K.R., et al. High-throughput extraction of SARS-CoV-2 RNA from nasopharyngeal swabs using solid-phase reverse immobilization beads. medRxiv, 2020.2004.2008.20055731 (2020). 37. 37.Kubina, R. & Dziedzic, A. Molecular and Serological Tests for COVID-19 a Comparative Review of SARS-CoV-2 Coronavirus Laboratory and Point-of-Care Diagnostics. Diagnostics (Basel) 10(2020). 38. 38.US Food and Drug Administration. Cobas® SARS-CoV-2: Qualitative assay for use on the cobas® 6800/8800 Systems. Avilable from: [https://www.fda.gov/media/136049/download](https://www.fda.gov/media/136049/download). Accessed on: December 02, 2020. (2020). 39. 39.Muench, P., et al. Development and Validation of the Elecsys Anti-SARS-CoV-2 Immunoassay as a Highly Specific Tool for Determining Past Exposure to SARS-CoV-2. J Clin Microbiol 58(2020). 40. 40.The Roche Group.Roche’s COVID-19 antibody test receives FDA Emergency Use Authorization and is Available in markets accepting the CE mark. Available from: [https://www.roche.com/media/releases/med-cor-2020-05-03.htm](https://www.roche.com/media/releases/med-cor-2020-05-03.htm). Accessed on: June 5, 2020. (2020). 41. 41.Oved, K., et al. Multi-center nationwide comparison of seven serology assays reveals a SARS-CoV-2 non-responding seronegative subpopulation. EClinicalMedicine 29, 100651 (2020). 42. 42.StataCorp. Stata Statistical Software: Release 17. College Station, TX: StataCorp LLC. (2021).