COVID-19 primary series and booster vaccination and immune imprinting ===================================================================== * Hiam Chemaitelly * Houssein H. Ayoub * Patrick Tang * Peter Coyle * Hadi M. Yassine * Asmaa A. Al Thani * Hebah A. Al-Khatib * Mohammad R. Hasan * 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 * Laith J. Abu-Raddad ## Abstract **Background** Epidemiological evidence for immune imprinting was investigated in immune histories related to vaccination in Qatar from onset of the omicron wave, on December 19, 2021, through September 15, 2022. **Methods** Matched, retrospective, cohort studies were conducted to investigate differences in incidence of SARS-CoV-2 reinfection in the national cohort of persons who had a primary omicron infection, but different vaccination histories. History of primary-series (two-dose) vaccination was compared to that of no vaccination, history of booster (three-dose) vaccination was compared to that of two-dose vaccination, and history of booster vaccination was compared to that of no vaccination. Associations were estimated using Cox proportional-hazards regression models. **Results** The adjusted hazard ratio comparing incidence of reinfection in the two-dose cohort to that in the unvaccinated cohort was 0.43 (95% CI: 0.38-0.48). The adjusted hazard ratio comparing incidence of reinfection in the three-dose cohort to that in the two-dose cohort was 1.38 (95% CI: 1.16-1.65). The adjusted hazard ratio comparing incidence of reinfection in the three-dose cohort to that in the unvaccinated cohort was 0.53 (95% CI: 0.44-0.63). All adjusted hazard ratios appeared stable over 6 months of follow-up. Divergence in cumulative incidence curves in all comparisons increased markedly when incidence was dominated by BA.4/BA.5 and BA.2.75*. No reinfection in any cohort progressed to severe, critical, or fatal COVID-19. **Conclusions** History of primary-series vaccination enhanced immune protection against omicron reinfection, but history of booster vaccination compromised protection against omicron reinfection. These findings do not undermine the short-term public health utility of booster vaccination. Keywords * COVID-19 * antigenic sin * immunity * natural infection * cohort study * epidemiology ## Introduction Three years into the coronavirus disease 2019 (COVID-19) pandemic, the global population carries heterogenous immune histories derived from various exposures to infection, viral variants, and vaccination.1 Laboratory science evidence suggests the possibility of immune imprinting, a negative impact for vaccination on subsequent protective immunity against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) induced by vaccination or infection, or a combination of both.1-4 Epidemiological evidence for immune imprinting in immune histories related to infection was recently investigated, but no evidence was found for imprinting compromising protection against B.1.1.529 (omicron) subvariants.5 A pre-omicron infection followed by an omicron reinfection enhanced protection against a second omicron reinfection.5 We investigated epidemiological evidence for imprinting in immune histories related to vaccination using matched, retrospective cohort studies. We compared incidence of SARS-CoV-2 reinfection in the national cohort of individuals who had a primary documented omicron infection after primary-series (two-dose) vaccination to that in the national cohort of individuals with a documented primary omicron infection, but no vaccination history. Analogously, we also compared reinfection incidence in those who had a documented primary omicron infection after booster (third dose) vaccination to each of the two-dose and unvaccinated cohorts. These immune histories were investigated because of their pervasiveness in the global population, and because of their potential relevance to the protection of bivalent booster vaccination that is being scaled up in different countries. ## Methods ### Study population and data sources This study was conducted in the population of Qatar from onset of the omicron wave on December 19, 20216 through September 15, 2022. It analyzed the national, federated databases for COVID-19 laboratory testing, vaccination, hospitalization, and death, retrieved from the integrated, nationwide, digital-health information platform (Section S1 of the Supplementary Appendix). Databases include all SARS-CoV-2-related data with no missing information since pandemic onset, such as all polymerase chain reaction (PCR) tests, and from January 5, 2022 onward, all rapid antigen tests conducted at healthcare facilities. SARS-CoV-2 testing in Qatar is done at mass scale, mostly for routine reasons.7,8 Most infections are diagnosed not because of symptoms, but because of routine testing.7,8 Qatar launched its COVID-19 vaccination program in December of 2020 using the BNT162b2 and mRNA-1273 vaccines.9 Detailed descriptions of Qatar’s population and of the national databases have been reported previously.7,8,10-12 ### Study design and cohorts Matched, retrospective, observational cohort studies were conducted to investigate epidemiological evidence for immune imprinting in individuals who had a documented primary omicron infection, but different prior vaccination histories. A documented primary omicron infection was defined as the first record of a SARS-CoV-2-positive PCR or rapid antigen test after onset of the omicron wave in Qatar on December 19, 20216 in an individual that had no record of a prior pre-omicron infection. In the first study, we compared incidence of reinfection in the national cohort of individuals who had a primary omicron infection after primary-series (two-dose) vaccination (designated as the two-dose cohort) to that in the national cohort of individuals who had a primary omicron infection, but no vaccination history (designated as the unvaccinated cohort). In the second study, we compared incidence of reinfection in the national cohort of individuals who had a primary omicron infection after booster (third dose) vaccination (designated as the three-dose cohort) to that in the two-dose cohort. In a third study, to confirm and complement results of the first two studies, we compared incidence of reinfection in the three-dose cohort to that in the unvaccinated cohort. The majority of primary omicron infections in these three studies involved the BA.2 subvariant.7,13,14 SARS-CoV-2 reinfection was defined as a documented infection ≥90 days after an earlier infection, to avoid misclassifying prolonged positivity as reinfection.6,15,16 Children vaccinated with the pediatric dose of BNT162b2 and adults who received different vaccines were excluded. Laboratory methods are in Section S2. Classification of reinfection severity followed World Health Organization guidelines for COVID-19 case severity (acute-care hospitalizations),17 criticality (intensive-care-unit hospitalizations),17 and fatality18 (Section S3). ### Cohort matching and follow-up Cohorts were matched exactly one-to-one by sex, 10-year age group, nationality, and number of coexisting conditions (none, one, two, three or more comorbid conditions) to balance observed confounders between exposure groups that are related to infection risk in Qatar.10,19-22 Individuals who were first diagnosed with SARS-CoV-2 in a specific week in one cohort were matched to individuals who were first diagnosed with SARS-CoV-2 in that same calendar week in the comparator cohort, to ensure that matched pairs were exposed to the same omicron subvariants and had presence in Qatar at the same time. Cohorts were also matched exactly by testing method (PCR versus rapid antigen testing) and by reason for testing for the primary omicron infection to control for potential differences in testing modalities between cohorts. Matching was performed iteratively such that individuals in the comparator cohort were alive, had not been reinfected, and had maintained the same vaccination status at the start of follow-up. Each matched pair was followed from 90 days after the primary omicron infection of the individual in the two-dose cohort for the study comparing incidence of reinfection in that cohort with the unvaccinated cohort. Follow-up was from 90 days after the primary omicron infection of the individual in the three-dose cohort for studies comparing incidence of reinfection in that cohort to that in each of the two-dose and unvaccinated cohorts. For exchangeability,12,23 both members of each matched pair were censored as soon as one of them received a new vaccine dose (change in vaccination status; that is at earliest occurrence of an unvaccinated individual in the matched pair receiving the first dose, or the individual with two-dose vaccination receiving a third dose, or the individual with three-dose vaccination receiving a fourth dose). Accordingly, individuals were followed up until the first of any of the following events: a documented SARS-CoV-2 reinfection (defined as the first PCR-positive or rapid-antigen-positive test after the start of follow-up, regardless of symptoms), a change in vaccination status (with matched-pair censoring), or death, or end of study censoring (September 15, 2022). ### Oversight The institutional review boards at Hamad Medical Corporation and Weill Cornell Medicine– Qatar approved this retrospective study with a waiver of informed consent. The study was reported according to the Strengthening the Reporting of Observational Studies in Epidemiology (STROBE) guidelines (Table S1). The authors vouch for the accuracy and completeness of the data and for the fidelity of the study to the protocol. Data used in this study are the property of the Ministry of Public Health of Qatar and were provided to the researchers through a restricted-access agreement for preservation of confidentiality of patient data. The funders had no role in the study design; the collection, analysis, or interpretation of the data; or the writing of the manuscript. ### Statistical analysis Eligible and matched cohorts were described using frequency distributions and measures of central tendency and were compared using standardized mean differences (SMDs). An SMD of ≤0.1 indicated adequate matching.24 Cumulative incidence of reinfection (defined as proportion of individuals at risk, whose primary endpoint during follow-up was a reinfection) was estimated using the Kaplan-Meier estimator method.25 Incidence rate of reinfection in each cohort, defined as number of identified reinfections divided by number of person-weeks contributed by all individuals in the cohort, was estimated, with the corresponding 95% confidence interval (CI) using a Poisson log-likelihood regression model with the Stata 17.0 *stptime* command. Hazard ratios, comparing incidence of reinfection in the cohorts and corresponding 95% CIs, were calculated using Cox regression, adjusted for the matching factors with the Stata 17.0 *stcox* command. Hazard ratios were additionally adjusted for differences in testing frequency between cohorts. Schoenfeld residuals and log-log plots for survival curves were used to test the proportional-hazards assumption. CIs were not adjusted for multiplicity; thus, they should not be used to infer definitive differences between groups. Interactions were not considered. Statistical analyses were conducted using Stata/SE version 17.0 (Stata Corporation, College Station, TX, USA). ## Results ### Two-dose cohort versus unvaccinated cohort Figure S1 shows the study population selection process. Table 1 describes baseline characteristics of the full and matched cohorts. Matched cohorts each included 56,802 individuals. The study population is broadly representative of individuals with primary omicron infection who had received two-dose vaccination or no vaccination in Qatar (Table S2). View this table: [Table 1.](http://medrxiv.org/content/early/2022/11/01/2022.10.31.22281756/T1) Table 1. Baseline characteristics of eligible and matched cohorts in studies investigating immune protection against reinfection among those who had a primary infection with an omicron subvariant, but had a history of A) two-dose vaccination compared to no vaccination, and B) three-dose vaccination compared to two-dose vaccination. Median date of the second vaccine dose for the two-dose cohort was June 9, 2021. Median duration between the second dose and start of follow-up was 312 days (interquartile range (IQR), 264-352 days). Median duration of follow-up was 157 days (IQR, 140-164 days) for the two-dose cohort and 157 days (IQR, 139-164 days) for the unvaccinated cohort (Figure 1A). There were 573 reinfections in the two-dose cohort and 1,044 reinfections in the unvaccinated cohort during follow-up (Figure S1). None progressed to severe, critical, or fatal COVID-19. ![Figure 1.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2022/11/01/2022.10.31.22281756/F1.medium.gif) [Figure 1.](http://medrxiv.org/content/early/2022/11/01/2022.10.31.22281756/F1) Figure 1. Cumulative incidence of reinfection among those who had a primary infection with an omicron subvariant after A) two-dose vaccination compared to no vaccination, and B) three-dose vaccination compared to two-dose vaccination. Cumulative incidence of reinfection was 1.4% (95% CI: 1.2-1.5%) for the two-dose cohort and 2.4% (95% CI: 2.2-2.5%) for the unvaccinated cohort, after 165 days of follow-up (Figure 1A). In the first 70 days of follow-up, incidence was dominated by BA.2.7,13,14 Subsequently, incidence was dominated by BA.4/BA.5,6,26 and then by BA.2.75* (predominantly BA.2.75.2). Divergence between the cumulative incidence curves increased markedly when incidence was no longer dominated by BA.2. The hazard ratio comparing incidence of reinfection in the two-dose cohort to that in the unvaccinated cohort, adjusted for matching factors, was 0.59 (95% CI: 0.53-0.67; Table 2). The adjusted hazard ratio appeared stable by month of follow-up (Figure 2A). The proportion of individuals who had a test during follow-up was 48.9% for the two-dose cohort and 37.0% for the unvaccinated cohort. The testing frequency was 0.93 and 0.67 tests per person, respectively. Adjusting the hazard ratio additionally by the ratio of testing frequencies between cohorts yielded an adjusted hazard ratio of 0.43 (95% CI: 0.38-0.48). View this table: [Table 2.](http://medrxiv.org/content/early/2022/11/01/2022.10.31.22281756/T2) Table 2. Hazard ratios for incidence of SARS-CoV-2 reinfection in studies investigating immune protection among those who had a primary infection with an omicron subvariant, but different vaccination histories. ![Figure 2.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2022/11/01/2022.10.31.22281756/F2.medium.gif) [Figure 2.](http://medrxiv.org/content/early/2022/11/01/2022.10.31.22281756/F2) Figure 2. Adjusted hazard ratio by month of follow-up for SARS-CoV-2 reinfection among those who had a primary infection with an omicron subvariant A) after two-dose vaccination compared to no vaccination, and B) after three-dose vaccination compared to two-dose vaccination. ### Three-dose cohort versus two-dose cohort Figure S2 shows the study population selection process. Table 1 describes baseline characteristics of the full and matched cohorts. Matched cohorts each included 30,541 individuals. The study population is broadly representative of individuals with primary omicron infection who had received three-dose or two-dose vaccination in Qatar (Table S2). Median dates of the second and third vaccine doses for the three-dose cohort were March 26, 2021 and December 6, 2021, respectively. Median date of the second vaccine dose for the two-dose cohort was May 11, 2021. Median duration between the third dose and start of follow-up was 124 days (IQR, 103-143 days), and between the second dose and start of follow-up was 334 days (IQR, 286-371 days). Median duration of follow-up was 157 days (IQR, 135-164 days) in the three-dose cohort and 157 days (IQR, 137-164 days) in the two-dose cohort (Figure 1B). There were 480 reinfections in the three-dose cohort and 248 reinfections in the two-dose cohort during follow-up (Figure S2). None progressed to severe, critical, or fatal COVID-19. Cumulative incidence of reinfection was 2.1% (95% CI: 1.9-2.3%) for the three-dose cohort and 1.1% (95% CI: 1.0-1.3%) for the two-dose cohort, after 165 days of follow-up (Figure 1B). In the first 70 days of follow-up, incidence was dominated by BA.2.7,13,14 Subsequently, incidence was dominated by BA.4/BA.5,6,26 and then by BA.2.75*. Divergence between the cumulative incidence curves increased markedly when incidence was no longer dominated by BA.2. The adjusted hazard ratio comparing incidence of reinfection in the three-dose cohort to that in the two-dose cohort was 1.96 (95% CI: 1.64-2.34; Table 2). The adjusted hazard ratio appeared stable by month of follow-up (Figure 2B). The proportion of individuals who had a test during follow-up was 63.1% for the three-dose cohort and 49.0% for the two-dose cohort. The testing frequency was 1.39 and 0.98 tests per person, respectively. Adjusting the hazard ratio additionally by the ratio of testing frequencies between cohorts yielded an adjusted hazard ratio of 1.38 (95% CI: 1.16-1.65). ### Three-dose cohort versus unvaccinated cohort Figure S3 shows the study population selection process. Table S3 describes baseline characteristics of the full and matched cohorts. Cumulative incidence of reinfection is shown in Figure S4A. The adjusted hazard ratio comparing incidence of reinfection in the three-dose cohort to that in the unvaccinated cohort was 1.10 (95% CI: 0.92-1.31; Table 2). The adjusted hazard ratio appeared stable by month of follow-up (Figure S4B). The proportion of individuals who had a test during follow-up was 66.4% for the three-dose cohort and 36.8% for the unvaccinated cohort. The testing frequency was 1.46 and 0.70 tests per person, respectively. Adjusting the hazard ratio additionally by the ratio of testing frequencies between cohorts yielded an adjusted hazard ratio of 0.53 (95% CI: 0.44-0.63). The results of this additional study confirm the relative differences in incidence of reinfection observed in the first two studies, with incidence being lowest among the two-dose cohort and highest among the unvaccinated cohort. ## Discussion Primary-series vaccination followed by a primary omicron infection was associated with enhanced immune protection against omicron reinfection compared to primary omicron infection with no prior vaccination. This result is striking because the start of follow-up in this study was ∼1 year after the two-dose primary series. Protection of the primary series against omicron infection should have fully waned by this time, considering how rapidly vaccine protection wanes against omicron subvariants.13,27 This finding suggests that the primary omicron infection may have stimulated immune memory of the earlier primary-series immune response in a manner that enhanced protection against a subsequent omicron reinfection, particularly against BA.4/BA.5 and BA.2.75*. Remarkably, similar effect and effect size were observed recently in an analogous study.5 Incidence of reinfection among unvaccinated persons who had contracted an omicron infection following an earlier pre-omicron infection was lower than incidence of reinfection among unvaccinated persons who had only an omicron infection and no prior pre-omicron infection.5 mRNA vaccines used in Qatar are based on index-virus design.28,29 The median duration between the first and second vaccine doses was <1 month.9 Given this short duration between doses, two-dose vaccination counts perhaps as a single pre-omicron immunological event. This may explain the similarity in both effect and effect size in these two studies, since in essence, both investigate immune protection elicited by a pre-omicron immunological event followed by an omicron immunological event, compared to protection of only a single omicron event. While two-dose vaccination was associated with enhanced protection against subsequent omicron reinfection, three-dose vaccination was associated with reduced protection compared to that of two-dose vaccination. This finding suggests that the immune response against the primary omicron infection was compromised by differential immune imprinting in those who received a third booster dose, consistent with emerging laboratory science data.1-4 The booster dose, a pre-omicron immunological event, that occurred several months after the primary-series vaccination, another pre-omicron immunological event, may have trained the immune response to expect a specific narrow pre-omicron challenge; thus, the response was inferior when the actual challenge was an immune-evasive omicron subvariant. Repeat immunological events of the same kind (here pre-omicron challenge) appear associated with compromised protection against a new kind of immunological event (here omicron challenge). We investigated two immune histories with different effects for immune imprinting on each. Primary-series vaccination followed by a primary omicron infection enhanced immune protection against omicron reinfection. Booster vaccination followed by a primary omicron infection compromised protection against omicron reinfection. This highlights the complexity of the immunity landscape at this stage of the pandemic, in which people have different immune histories. These findings, however, do not undermine the utility of booster vaccination, at least in the short-term. Compromised protection was observed after booster effectiveness waned, as follow-up commenced >4 months after the booster, at a time when booster effectiveness is expected to be marginal.13,27 There is no question that the booster dose reduced infection incidence right after its administration, based on evidence from this same population.7,12,13 Nonetheless, findings indicate that short-term effects of boosters may differ from their long-term effects. Although we planned to investigate effectiveness against severe COVID-19, no reinfection in any cohort of the three studies progressed to severe, critical, or fatal COVID-19. This outcome is not unexpected given the lower severity of omicron infections30,31 and the strong protection of natural infection against severe COVID-19 at reinfection, estimated at 97% in this same population,32 as well as the long-term effectiveness of primary-series and boosters against severe COVID-19.7,8,13,27,33 While we were unable to quantify effects of immune imprinting on COVID-19 severity, the results do not suggest imprinting compromising protection against severe COVID-19. This study has limitations. We investigated incidence of documented reinfections, but undocumented reinfections may have occurred. Unvaccinated individuals are a minority in Qatar, and may not be truly immune-naïve due to undocumented prior infections or undocumented vaccinations, perhaps outside the country, especially now that we are three years into this pandemic. Bias due to unequal depletion of the unvaccinated versus vaccinated susceptible population may underestimate vaccine protection.34 With Qatar’s young population, our findings may not be generalizable to older individuals or to other countries where elderly citizens constitute a large proportion of the total population. Testing frequency differed between cohorts, reflecting different travel testing guidelines for vaccinated versus unvaccinated individuals. While adjustment for these differences affected estimated hazard ratios quantitatively, they did not materially change the findings. Home-based rapid antigen testing is not documented in Qatar (Section S1), and is not factored in these analyses. However, there is no reason to believe that home-based testing could have differentially affected the followed cohorts to alter study estimates. Matching was done while factoring key socio-demographic characteristics of the population,10,19-22 and this may also have controlled or reduced differences in home-based testing between cohorts. As an observational study, investigated cohorts were neither blinded nor randomized, so unmeasured or uncontrolled confounding cannot be excluded. Although matching covered key factors affecting infection exposure,10,19-22 it was not possible for other factors such as geography or occupation, for which data were unavailable. However, Qatar is essentially a city state and infection incidence was broadly distributed across neighborhoods. Nearly 90% of Qatar’s population are expatriates from over 150 countries, who come here for employment.10 Nationality, age, and sex provide a powerful proxy for socio-economic status in this country.10,19-22 Nationality is strongly associated with occupation.10,20-22 The matching prescription used in this study was investigated in previous studies of different epidemiologic designs, and using control groups to test for null effects.8,9,33,35,36 These control groups included unvaccinated cohorts versus vaccinated cohorts within two weeks of the first dose,8,33,35,36 when vaccine protection is negligible,28,29 and mRNA-1273-versus BNT162b2-vaccinated cohorts, also in the first two weeks after the first dose.9 These studies showed repeatedly and at different times during the pandemic that this prescription provides adequate control of differences in infection exposure,8,9,33,35,36 suggesting that the employed matching may also have controlled for differences in infection exposure in these studies. All analyses were implemented on Qatar’s total population, perhaps minimizing the likelihood of bias. In conclusion, primary-series vaccination followed by a primary omicron infection enhanced immune protection against omicron reinfection. However, booster vaccination followed by a primary omicron infection compromised protection against omicron reinfection, perhaps because it involved repeat pre-omicron immunological events before the omicron infection. These findings do not undermine the utility of booster vaccination in the short-term, but may point to potentially significant public health complexities requiring fine-tuning of booster vaccination to those who can best benefit from it, such as those most clinically vulnerable to severe COVID-19. ### Sources of support and acknowledgements We acknowledge the many dedicated individuals at Hamad Medical Corporation, the Ministry of Public Health, the Primary Health Care Corporation, Qatar Biobank, Sidra Medicine, and Weill Cornell Medicine-Qatar for their diligent efforts and contributions to make this study possible. The authors are grateful for institutional salary support from the Biomedical Research Program and the Biostatistics, Epidemiology, and Biomathematics Research Core, both at Weill Cornell Medicine-Qatar, as well as for institutional salary support provided by the Ministry of Public Health, Hamad Medical Corporation, and Sidra Medicine. The authors are also grateful for the Qatar Genome Programme and Qatar University Biomedical Research Center for institutional support for the reagents needed for 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. ## 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 HC co-designed the study, performed the statistical analyses, and co-wrote the first draft of the article. LJA conceived and co-designed the study, led the statistical analyses, and co-wrote the first draft of the article. PT and MRH conducted multiplex, RT-qPCR variant screening and viral genome sequencing. PVC designed mass PCR testing to allow routine capture of SGTF variants and conducted viral genome sequencing. HY, HAK, and MS conducted viral genome sequencing. All authors contributed to data collection and acquisition, database development, discussion and interpretation of the results, and to the writing of the article. All authors have read and approved the final manuscript. Decision to publish the paper was by consensus among all authors. ## 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. ## Appendix ### Supplementary Appendix #### Section S1. Further details on methods ##### Data sources and testing Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) testing in the healthcare system in Qatar is done at a mass scale, and mostly for routine reasons, where about 5% of the population are tested every week.1,2 About 75% of those diagnosed are diagnosed not because of appearance of symptoms, but because of routine testing.1,2 Every polymerase chain reaction (PCR) test and an increasing proportion of the facility-based rapid antigen tests conducted in Qatar, regardless of location or setting, are classified on the basis of symptoms and the reason for testing (clinical symptoms, contact tracing, surveys or random testing campaigns, individual requests, routine healthcare testing, pre-travel, at port of entry, or other). All facility-based testing done during follow-up in the present study was factored in the analyses of this study. Rapid antigen test kits are available for purchase in pharmacies in Qatar, but outcome of home-based testing is not reported nor documented in the national databases. Since SARS-CoV-2-test outcomes are linked to specific public health measures, restrictions, and privileges, testing policy and guidelines stress facility-based testing as the core testing mechanism in the population. While facility-based testing is provided free of charge or at low subsidized costs, depending on the reason for testing, home-based rapid antigen testing is de-emphasized and not supported as part of national policy. There is no reason to believe that home-based testing could have differentially affected the followed matched cohorts to affect our results. The infection detection rate is defined as the cumulative number of documented infections, that is diagnosed and laboratory-confirmed infections, over the cumulative number of documented and undocumented infections. Serological surveys and other analyses suggest that a substantial proportion of infections in Qatar and elsewhere are undocumented.3-9 With absence of recent serological surveys in Qatar, it is difficult to estimate the current or recent infection detection rate, but mathematical modeling analyses and their recent updates suggest that at present no less than 50% of infections are never documented.7,10 However, there is no reason to believe that undocumented infections could have differentially affected the followed matched cohorts to affect our results. Qatar has unusually young, diverse demographics, in that only 9% of its residents are ≥50 years of age, and 89% are expatriates from over 150 countries.11,12 Further descriptions of the study population and these national databases were reported previously.1,2,12-14 ##### Comorbidity classification Comorbidities were ascertained and classified based on the ICD-10 codes for chronic conditions as recorded in the electronic health record encounters of each individual in the Cerner-system national database that includes all citizens and residents registered in the national and universal public healthcare system. All encounters for each individual were analyzed to determine the comorbidity classification for that individual, as part of a recent national analysis to assess healthcare needs and resource allocation. The Cerner-system national database includes encounters starting from 2013, after this system was launched in Qatar. As long as each individual had at least one encounter with a specific comorbidity diagnosis since 2013, this person was classified with this comorbidity. Individuals who have comorbidities but never sought care in the public healthcare system, or seek care exclusively in private healthcare facilities, are classified as individuals with no comorbidity due to absence of recorded encounters for them. #### Section S2. Laboratory methods and variant ascertainment ##### Real-time reverse-transcription polymerase chain reaction testing Nasopharyngeal and/or oropharyngeal swabs were collected for polymerase chain reaction (PCR) testing and placed in Universal Transport Medium (UTM). Aliquots of UTM were: 1) extracted on KingFisher Flex (Thermo Fisher Scientific, USA), MGISP-960 (MGI, China), or ExiPrep 96 Lite (Bioneer, South Korea) followed by testing with real-time reverse-transcription PCR (RT-qPCR) using TaqPath COVID-19 Combo Kits (Thermo Fisher Scientific, USA) on an ABI 7500 FAST (Thermo Fisher Scientific, USA); 2) tested directly on the Cepheid GeneXpert system using the Xpert Xpress SARS-CoV-2 (Cepheid, USA); or 3) loaded directly into a Roche cobas 6800 system and assayed with the cobas SARS-CoV-2 Test (Roche, Switzerland). The first assay targets the viral S, N, and ORF1ab gene regions. The second targets the viral N and E-gene regions, and the third targets the ORF1ab and E-gene regions. All PCR testing was conducted at the Hamad Medical Corporation Central Laboratory or Sidra Medicine Laboratory, following standardized protocols. ##### Rapid antigen testing Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) antigen tests were performed on nasopharyngeal swabs using one of the following lateral flow antigen tests: Panbio COVID-19 Ag Rapid Test Device (Abbott, USA); SARS-CoV-2 Rapid Antigen Test (Roche, Switzerland); Standard Q COVID-19 Antigen Test (SD Biosensor, Korea); or CareStart COVID-19 Antigen Test (Access Bio, USA). All antigen tests were performed point-of-care according to each manufacturer’s instructions at public or private hospitals and clinics throughout Qatar with prior authorization and training by the Ministry of Public Health (MOPH). Antigen test results were electronically reported to the MOPH in real time using the Antigen Test Management System which is integrated with the national Coronavirus Disease 2019 (COVID-19) database. ##### Classification of infections by variant type Surveillance for the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants in Qatar is based on viral genome sequencing and multiplex real-time reverse-transcription polymerase chain reaction (RT-qPCR) variant screening15 of random positive clinical samples,2,16-20 complemented by deep sequencing of wastewater samples.18,21,22 Further details on the viral genome sequencing and multiplex RT-qPCR variant screening throughout the SARS-CoV-2 waves in Qatar can be found in previous publications.1,2,14,16-20,23-27 #### Section S3. COVID-19 severity, criticality, and fatality classification Classification of Coronavirus Disease 2019 (COVID-19) case severity (acute-care hospitalizations),28 criticality (intensive-care-unit hospitalizations),28 and fatality29 followed World Health Organization (WHO) guidelines. Assessments were made by trained medical personnel independent of study investigators and using individual chart reviews, as part of a national protocol applied to every hospitalized COVID-19 patient. Each hospitalized COVID-19 patient underwent an infection severity assessment every three days until discharge or death. We classified individuals who progressed to severe, critical, or fatal COVID-19 between the time of the documented infection and the end of the study based on their worst outcome, starting with death,29 followed by critical disease,28 and then severe disease.28 Severe COVID-19 disease was defined per WHO classification as a SARS-CoV-2 infected person with “oxygen saturation of <90% on room air, and/or respiratory rate of >30 breaths/minute in adults and children >5 years old (or ≥60 breaths/minute in children <2 months old or ≥50 breaths/minute in children 2-11 months old or ≥40 breaths/minute in children 1–5 years old), and/or signs of severe respiratory distress (accessory muscle use and inability to complete full sentences, and, in children, very severe chest wall indrawing, grunting, central cyanosis, or presence of any other general danger signs)”.28 Detailed WHO criteria for classifying Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection severity can be found in the WHO technical report.28 Critical COVID-19 disease was defined per WHO classification as a SARS-CoV-2 infected person with “acute respiratory distress syndrome, sepsis, septic shock, or other conditions that would normally require the provision of life sustaining therapies such as mechanical ventilation (invasive or non-invasive) or vasopressor therapy”.28 Detailed WHO criteria for classifying SARS-CoV-2 infection criticality can be found in the WHO technical report.28 COVID-19 death was defined per WHO classification as “a death resulting from a clinically compatible illness, in a probable or confirmed COVID-19 case, unless there is a clear alternative cause of death that cannot be related to COVID-19 disease (e.g. trauma). There should be no period of complete recovery from COVID-19 between illness and death. A death due to COVID-19 may not be attributed to another disease (e.g. cancer) and should be counted independently of preexisting conditions that are suspected of triggering a severe course of COVID-19”. Detailed WHO criteria for classifying COVID-19 death can be found in the WHO technical report.29 View this table: [Table S1.](http://medrxiv.org/content/early/2022/11/01/2022.10.31.22281756/T3) Table S1. STROBE checklist for cohort studies. ![Figure S1.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2022/11/01/2022.10.31.22281756/F3.medium.gif) [Figure S1.](http://medrxiv.org/content/early/2022/11/01/2022.10.31.22281756/F3) Figure S1. Flowchart describing the population selection process for investigating immune protection against reinfection among those who had a primary infection with an omicron subvariant after two-dose vaccination compared to protection among those who had a primary infection with an omicron subvariant but were unvaccinated. View this table: [Table S2.](http://medrxiv.org/content/early/2022/11/01/2022.10.31.22281756/T4) Table S2. Representativeness of study participants. ![Figure S2.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2022/11/01/2022.10.31.22281756/F4.medium.gif) [Figure S2.](http://medrxiv.org/content/early/2022/11/01/2022.10.31.22281756/F4) Figure S2. Flowchart describing the population selection process for investigating immune protection against reinfection among those who had a primary infection with an omicron subvariant after three-dose vaccination compared to protection among those who had a primary infection with an omicron subvariant after two-dose vaccination. ![Figure S3.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2022/11/01/2022.10.31.22281756/F5.medium.gif) [Figure S3.](http://medrxiv.org/content/early/2022/11/01/2022.10.31.22281756/F5) Figure S3. Flowchart describing the population selection process for investigating immune protection against reinfection among those who had a primary infection with an omicron subvariant after three-dose vaccination compared to protection among those who had a primary infection with an omicron subvariant but were unvaccinated. View this table: [Table S3.](http://medrxiv.org/content/early/2022/11/01/2022.10.31.22281756/T5) Table S3. Baseline characteristics of eligible and matched cohorts in the study investigating immune protection against reinfection among those who had a primary infection with an omicron subvariant after three-dose vaccination compared to those who had a primary infection with an omicron subvariant but were unvaccinated. ![Figure S4.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2022/11/01/2022.10.31.22281756/F6.medium.gif) [Figure S4.](http://medrxiv.org/content/early/2022/11/01/2022.10.31.22281756/F6) Figure S4. A) Cumulative incidence of and B) adjusted hazard ratio by month of follow-up for SARS-CoV-2 reinfection among those who had a primary infection with an omicron subvariant after three-dose vaccination compared to those who had a primary infection with an omicron subvariant but were unvaccinated. * Received October 31, 2022. * Revision received October 31, 2022. * Accepted November 1, 2022. * © 2022, 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 CJ, Pade C, Gibbons JM, et al. Immune boosting by B.1.1.529 (Omicron) depends on previous SARS-CoV-2 exposure. Science 2022:eabq1841. 2. 2.Röltgen K, Nielsen SCA, Silva O, et al. Immune imprinting, breadth of variant recognition, and germinal center response in human SARS-CoV-2 infection and vaccination. Cell 2022;185:1025-40.e14. 3. 3.Collier A-r, Miller J, Hachmann N, et al. Immunogenicity of the BA.5 Bivalent mRNA Vaccine Boosters. bioRxiv 2022:2022.10.24.513619. 4. 4.Wang Q, Bowen A, Valdez R, et al. Antibody responses to Omicron BA.4/BA.5 bivalent mRNA vaccine booster shot. bioRxiv 2022:2022.10.22.513349. 5. 5.Chemaitelly H, Ayoub HH, Tang P, et al. Immune Imprinting and Protection against Repeat Reinfection with SARS-CoV-2. N Engl J Med 2022. 6. 6.Altarawneh HN, Chemaitelly H, Hasan MR, et al. Protection against the Omicron Variant from Previous SARS-CoV-2 Infection. N Engl J Med 2022;386:1288–90. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F01%2F2022.10.31.22281756.atom) 7. 7.Altarawneh HN, Chemaitelly H, Ayoub HH, et al. Effects of Previous Infection and Vaccination on Symptomatic Omicron Infections. N Engl J Med 2022;387:21–34. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F01%2F2022.10.31.22281756.atom) 8. 8.Chemaitelly H, Tang P, Hasan MR, et al. Waning of BNT162b2 Vaccine Protection against SARS-CoV-2 Infection in Qatar. N Engl J Med 2021;385:e83. [CrossRef](http://medrxiv.org/lookup/external-ref?access\_num=10.1056/NEJMOA2114114/SUPPL_FILE/NEJMOA2114114_DISCLOSURES.PDF&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F01%2F2022.10.31.22281756.atom) 9. 9.Abu-Raddad LJ, Chemaitelly H, Bertollini R, National Study Group for Covid Vaccination. Effectiveness of mRNA-1273 and BNT162b2 Vaccines in Qatar. N Engl J Med 2022;386:799–800. 10. 10.Abu-Raddad LJ, Chemaitelly H, Ayoub HH, et al. Characterizing the Qatar advanced-phase SARS-CoV-2 epidemic. Sci Rep 2021;11:6233. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41598-021-85428-7&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F01%2F2022.10.31.22281756.atom) 11. 11.Chemaitelly H, Bertollini R, Abu-Raddad LJ, National Study Group for Covid Epidemiology. Efficacy of Natural Immunity against SARS-CoV-2 Reinfection with the Beta Variant. N Engl J Med 2021;385:2585–6. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMc2110300&link_type=DOI) 12. 12.Abu-Raddad LJ, Chemaitelly H, Ayoub HH, et al. Effect of mRNA Vaccine Boosters against SARS-CoV-2 Omicron Infection in Qatar. N Engl J Med 2022;386:1804–16. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F01%2F2022.10.31.22281756.atom) 13. 13.Chemaitelly H, Ayoub HH, AlMukdad S, et al. Duration of mRNA vaccine protection against SARS-CoV-2 Omicron BA.1 and BA.2 subvariants in Qatar. Nat Commun 2022;13:3082. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41467-022-30895-3&link_type=DOI) 14. 14.Chemaitelly H, Ayoub HH, Coyle P, et al. Protection of Omicron sub-lineage infection against reinfection with another Omicron sub-lineage. Nat Commun 2022;13:4675. 15. 15.Kojima N, Shrestha NK, Klausner JD. A Systematic Review of the Protective Effect of Prior SARS-CoV-2 Infection on Repeat Infection. Eval Health Prof 2021;44:327–32. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1177/01632787211047932&link_type=DOI) 16. 16.Pilz S, Theiler-Schwetz V, Trummer C, Krause R, Ioannidis JPA. SARS-CoV-2 reinfections: Overview of efficacy and duration of natural and hybrid immunity. Environ Res 2022:112911. 17. 17.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. 18. 18.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. 19. 19.Ayoub HH, Chemaitelly H, Seedat S, 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 2021;11:05005. 20. 20.Coyle PV, Chemaitelly H, Ben Hadj Kacem MA, 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 2021;24:102646. 21. 21.Al-Thani MH, Farag E, Bertollini R, et al. SARS-CoV-2 Infection Is at Herd Immunity in the Majority Segment of the Population of Qatar. Open Forum Infect Dis 2021;8:ofab221. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/ofid/ofab221&link_type=DOI) 22. 22.Jeremijenko A, Chemaitelly H, Ayoub HH, et al. Herd Immunity against Severe Acute Respiratory Syndrome Coronavirus 2 Infection in 10 Communities, Qatar. Emerg Infect Dis 2021;27:1343–52. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3201/eid2705.204365&link_type=DOI) 23. 23.Barda N, Dagan N, Cohen C, et al. Effectiveness of a third dose of the BNT162b2 mRNA COVID-19 vaccine for preventing severe outcomes in Israel: an observational study. Lancet 2021;398:2093–100. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S0140-6736(21)02249-2/ATTACHMENT/588607F0-89F7-4E80-8E48-FB7CE8C7F7EC/MMC1.PDF&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F01%2F2022.10.31.22281756.atom) 24. 24.Austin PC. Using the Standardized Difference to Compare the Prevalence of a Binary Variable Between Two Groups in Observational Research. Communications in Statistics - Simulation and Computation 2009;38:1228–34. 25. 25.Kaplan EL, Meier P. Nonparametric Estimation from Incomplete Observations. J Am Stat Assoc 1958;53:457–81. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.2307/2281868&link_type=DOI) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1958WX09300012&link_type=ISI) 26. 26.Altarawneh HN, Chemaitelly H, Ayoub HH, et al. Protective Effect of Previous SARS-CoV-2 Infection against Omicron BA.4 and BA.5 Subvariants. N Engl J Med 2022. 27. 27.Andrews N, Stowe J, Kirsebom F, et al. Covid-19 Vaccine Effectiveness against the Omicron (B.1.1.529) Variant. N Engl J Med 2022;386:1532–46. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/nejmoa2119451&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F01%2F2022.10.31.22281756.atom) 28. 28.Polack FP, Thomas SJ, Kitchin N, et al. Safety and Efficacy of the BNT162b2 mRNA Covid-19 Vaccine. N Engl J Med 2020;383:2603–15. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa2034577&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F01%2F2022.10.31.22281756.atom) 29. 29.Baden LR, El Sahly HM, Essink B, et al. Efficacy and Safety of the mRNA-1273 SARS-CoV-2 Vaccine. N Engl J Med 2021;384:403–16. [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%2F2022%2F11%2F01%2F2022.10.31.22281756.atom) 30. 30.Butt AA, Dargham SR, Coyle P, et al. COVID-19 Disease Severity in Persons Infected With Omicron BA.1 and BA.2 Sublineages and Association With Vaccination Status. JAMA Intern Med 2022. 31. 31.Butt AA, Dargham SR, Tang P, et al. COVID-19 disease severity in persons infected with the Omicron variant compared with the Delta variant in Qatar. J Glob Health 2022;12:05032. 32. 32.Chemaitelly H, Nagelkerke N, Ayoub HH, et al. Duration of immune protection of SARS-CoV-2 natural infection against reinfection. J Travel Med 2022. 33. 33.Abu-Raddad LJ, Chemaitelly H, Bertollini R, National Study Group for Covid Vaccination. Waning mRNA-1273 Vaccine Effectiveness against SARS-CoV-2 Infection in Qatar. N Engl J Med 2022;386:1091–3. 34. 34.Ray GT, Lewis N, Klein NP, Daley MF, Lipsitch M, Fireman B. Depletion-of-susceptibles Bias in Analyses of Intra-season Waning of Influenza Vaccine Effectiveness. Clin Infect Dis 2020;70:1484–6. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/cid/ciz706&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=31351439&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F01%2F2022.10.31.22281756.atom) 35. 35.Abu-Raddad LJ, Chemaitelly H, Yassine HM, et al. Pfizer-BioNTech mRNA BNT162b2 Covid-19 vaccine protection against variants of concern after one versus two doses. J Travel Med 2021;28. 36. 36.Chemaitelly H, Yassine HM, Benslimane FM, 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;27:1614–21. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F01%2F2022.10.31.22281756.atom) ## References 1. 1.Altarawneh HN, Chemaitelly H, Ayoub HH, et al. Effects of Previous Infection and Vaccination on Symptomatic Omicron Infections. N Engl J Med 2022;387:21–34. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F01%2F2022.10.31.22281756.atom) 2. 2.Chemaitelly H, Tang P, Hasan MR, et al. Waning of BNT162b2 Vaccine Protection against SARS-CoV-2 Infection in Qatar. N Engl J Med 2021;385:e83. [CrossRef](http://medrxiv.org/lookup/external-ref?access\_num=10.1056/NEJMOA2114114/SUPPL_FILE/NEJMOA2114114_DISCLOSURES.PDF&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F01%2F2022.10.31.22281756.atom) 3. 3.Al-Thani MH, Farag E, Bertollini R, et al. SARS-CoV-2 infection is at herd immunity in the majority segment of the population of Qatar. Open Forum Infectious Diseases 2021. 4. 4.Coyle PV, Chemaitelly H, Ben Hadj Kacem MA, 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 2021;24:102646. 5. 5.Jeremijenko A, Chemaitelly H, Ayoub HH, et al. Herd Immunity against Severe Acute Respiratory Syndrome Coronavirus 2 Infection in 10 Communities, Qatar. Emerg Infect Dis 2021;27:1343–52. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3201/eid2705.204365&link_type=DOI) 6. 6.Seedat S, Chemaitelly H, Ayoub HH, et al. SARS-CoV-2 infection hospitalization, severity, criticality, and fatality rates in Qatar. Sci Rep 2021;11:18182. 7. 7.Ayoub HH, Chemaitelly H, Seedat S, 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 2021;11:05005. 8. 8.Ayoub HH, Mumtaz GR, Seedat S, Makhoul M, Chemaitelly H, Abu-Raddad LJ. Estimates of global SARS-CoV-2 infection exposure, infection morbidity, and infection mortality rates in 2020. Glob Epidemiol 2021;3:100068. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F01%2F2022.10.31.22281756.atom) 9. 9.Angulo FJ, Finelli L, Swerdlow DL. Estimation of US SARS-CoV-2 Infections, Symptomatic Infections, Hospitalizations, and Deaths Using Seroprevalence Surveys. JAMA Netw Open 2021;4:e2033706. 10. 10.Ayoub HH, Chemaitelly H, Makhoul M, et al. Epidemiological impact of prioritising SARS-CoV-2 vaccination by antibody status: mathematical modelling analyses. BMJ Innov 2021;7:327–36. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6ODoiYm1qaW5ub3YiO3M6NToicmVzaWQiO3M6NzoiNy8yLzMyNyI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIyLzExLzAxLzIwMjIuMTAuMzEuMjIyODE3NTYuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 11. 11.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. 12. 12.Abu-Raddad LJ, Chemaitelly H, Ayoub HH, et al. Characterizing the Qatar advanced-phase SARS-CoV-2 epidemic. Sci Rep 2021;11:6233. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41598-021-85428-7&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F01%2F2022.10.31.22281756.atom) 13. 13.Chemaitelly H, Bertollini R, Abu-Raddad LJ, National Study Group for Covid Epidemiology. Efficacy of Natural Immunity against SARS-CoV-2 Reinfection with the Beta Variant. N Engl J Med 2021;385:2585–6. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMc2110300&link_type=DOI) 14. 14.Abu-Raddad LJ, Chemaitelly H, Ayoub HH, et al. Effect of mRNA Vaccine Boosters against SARS-CoV-2 Omicron Infection in Qatar. N Engl J Med 2022;386:1804–16. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F01%2F2022.10.31.22281756.atom) 15. 15.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. (Accessed June 6, 2021, at [https://www.protocols.io/view/multiplexed-rt-qpcr-to-screen-for-sars-cov-2-b-1-1-br9vm966](https://www.protocols.io/view/multiplexed-rt-qpcr-to-screen-for-sars-cov-2-b-1-1-br9vm966).) 16. 16.Abu-Raddad LJ, Chemaitelly H, Butt AA, National Study Group for Covid Vaccination. Effectiveness of the BNT162b2 Covid-19 Vaccine against the B.1.1.7 and B.1.351 Variants. N Engl J Med 2021;385:187–9. [CrossRef](http://medrxiv.org/lookup/external-ref?access\_num=10.1056/NEJMC2104974/SUPPL_FILE/NEJMC2104974_DISCLOSURES.PDF&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F01%2F2022.10.31.22281756.atom) 17. 17.Chemaitelly H, Yassine HM, Benslimane FM, 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;27:1614–21. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F01%2F2022.10.31.22281756.atom) 18. 18.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. at [https://www.gisaid.org/phylodynamics/global/nextstrain/](https://www.gisaid.org/phylodynamics/global/nextstrain/).) 19. 19.Benslimane FM, Al Khatib HA, Al-Jamal O, et al. One Year of SARS-CoV-2: Genomic Characterization of COVID-19 Outbreak in Qatar. Front Cell Infect Microbiol 2021;11:768883. 20. 20.Hasan MR, Kalikiri MKR, Mirza F, et al. Real-Time SARS-CoV-2 Genotyping by High-Throughput Multiplex PCR Reveals the Epidemiology of the Variants of Concern in Qatar. Int J Infect Dis 2021;112:52–4. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.ijid.2021.09.006&link_type=DOI) 21. 21.Saththasivam J, El-Malah SS, Gomez TA, et al. COVID-19 (SARS-CoV-2) outbreak monitoring using wastewater-based epidemiology in Qatar. Sci Total Environ 2021;774:145608. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.scitotenv.2021.145608&link_type=DOI) 22. 22.El-Malah SS, Saththasivam J, Jabbar KA, et al. Application of human RNase P normalization for the realistic estimation of SARS-CoV-2 viral load in wastewater: A perspective from Qatar wastewater surveillance. Environ Technol Innov 2022;27:102775. 23. 23.Tang P, Hasan MR, Chemaitelly H, et al. BNT162b2 and mRNA-1273 COVID-19 vaccine effectiveness against the SARS-CoV-2 Delta variant in Qatar. Nat Med 2021;27:2136–43. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F01%2F2022.10.31.22281756.atom) 24. 24.Altarawneh HN, Chemaitelly H, Hasan MR, et al. Protection against the Omicron Variant from Previous SARS-CoV-2 Infection. N Engl J Med 2022;386:1288–90. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F11%2F01%2F2022.10.31.22281756.atom) 25. 25.Chemaitelly H, Ayoub HH, AlMukdad S, et al. Duration of mRNA vaccine protection against SARS-CoV-2 Omicron BA.1 and BA.2 subvariants in Qatar. Nat Commun 2022;13:3082. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41467-022-30895-3&link_type=DOI) 26. 26.Qassim SH, Chemaitelly H, Ayoub HH, et al. Effects of BA.1/BA.2 subvariant, vaccination, and prior infection on infectiousness of SARS-CoV-2 omicron infections. J Travel Med 2022. 27. 27.Altarawneh HN, Chemaitelly H, Ayoub HH, et al. Protective Effect of Previous SARS-CoV-2 Infection against Omicron BA.4 and BA.5 Subvariants. N Engl J Med 2022. 28. 28.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. 29. 29.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.